Mutation Detection Flashcards

1
Q

How many rare diseases have been described, and how many people do they affect in the UK?

A

Approximately 7,000 rare diseases have been described, affecting about 1 in 17 of the UK population (approximately 3.5 million individuals

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How many rare diseases are caused by highly penetrant single nucleotide variants (SNV), small indels, or CNVs?

A

5000

Relies on the identification of a disease causing germline variant

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is the primary challenge in interpreting whole exome and whole genome sequencing for inherited diseases?

A

Whittling down a list of candidate variants to identify the disease-causing one(s).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What guidelines are used for germline variant interpretation?

A

In 2015, the American College of Medical Genetics (ACMG) published guidelines to a series of criteria in Mendelian disorders.

In 2016, adopted by the Association for Clinical Genomic Science (ACGS): ACGS Best Practice Guidelines for Variant Classification in Rare Disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What evidence is used to classify germline variants using ACGS?

A

Collate evidence from population data, computational data, functional data, segregation data, literature evidence, de novo data

Many software packages to aid interpretation (for example, Alamut, Congenica

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What classification system does ACGS use?

A

ACMG/ACGS guidelines use a five-class system:

5: Pathogenic >99% probability of a variant being disease-causing
4: Likely pathogenic >90%
3: Uncertain significance
2: Likely benign <10%
1: Benign <0.1%

Each evidence relates to a criterion and is worth a specific number of points. evidence points for pathogenicity (Very Strong= 8, Strong= 4, Moderate= 2, Supporting= 1) or benignity (Strong= -4, Moderate= -2, Supporting= -1). Evidence point thresholds for the 5 classes are: ≥10 (Pathogenic), 6-9 (Likely Pathogenic), -1 to -5 (Likely Benign), ≤-6 (Benign)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What other germline interpretation guidelines are used?

A

Cancer specific: CanVig- adapts ACGS to be suitable for cancer predisposition genes

Disease specific e.g. BRCA, Lynch

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What somatic variant interpretation guidelines are used?

A

Association for Molecular Pathology (AMP) Guidelines (Li et al 2017)

S-VIG also been developed but often not fully used in routine practice

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the AMP guidelines?

A

4 tier system based on clinical actionability

Uses:

Population data e.g gnomAD

Functional data

Predictive data

Cancer hotspots e.g. cancer databases COSMIC, mycancergenome etc

Drug approval guidelines: NICE, CDF, NCCN

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

How does AMP guidelines classify variants?

A

Tier I, variants of strong clinical significance - made up of variants with level A & B evidence (clinically actionable)

Tier II, variants of potential clinical significance- made up of variants with level C &D evidence (clinically actionable)

Tier III, variants of unknown significance- evidence may be conflicting or absent- (VUS)

Tier IV, benign or likely benign variants- there is evidence a variant does not have any actionability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is meant by a driver mutation?

A

Confer growth advantage on the cells carrying them and have been positively selected during the evolution of the cancer. Non-recurrent variants are unlikely to be drivers otherwise they would more than likely been seen previously

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What is different about SVIG and AMP guidelines?

A

SVIG aims to help standardise somatic VI and bring in line with germline ACGS guidelines

Uses evidence based points system to help determine oncogenicity of result

Has a list of known hotspot variants that are oncogenic

Can only be used on SNVs, not suitable for structural or copy number variants

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What variant nomenclature is used?

A

Human genome variation society (HGVS)
- Clinical reports should include sequence reference) to ensure unambiguous naming of the variant at the DNA level as well as provide coding and protein nomenclature
(e.g., “c.” for coding DNA sequence, “p.” for protein,

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are external bioinformatic databases?

A

External Bioinformatic Databases (DBs) can be described as databases which store biological data information. The data included in the databases/resources can be split into the main following areas:

  1. genome and sequence data (sequence alignment, variant databases, phylogenetic and splicing predictions)
  2. transcriptomics data (e.g. full length cDNAs or mRNAs),
  3. proteomics data (e.g. protein databases, protein structure, family and domain classification)
  4. other specialised databases (e.g. cancer and methylation databases).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are primary and secondary databases?

A

Primary databases consist of experimentally derived data (e.g. nucleotide and protein sequences).

Secondary databases consist of data produced from the analysis of primary data. Secondary databases often include data from a combination of other databases (both primary and secondary databases) and other (e.g. literature).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Give some examples of genomic databases?

A

Primary
- EMBL (European Bioinformatics institute) (Europe)
- GenBank (National Centre for Biotechnology Information) (USA)

Secondary
OMIM (Online Mendelian Inheritance in Man)
RefSeq
Decipher
ClinVar
The cancer Genome Atlas (TCGA)
Cosmic
ClinGen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is germline conversion rate?

A

Number of pathogenic variants of true germline origin× 100/total number of tumour-detected pathogenic variants

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are Cancer Susceptibility Genes?

A

A term used to describe a gene that may increase a person’s risk of developing some types of cancer if it has certain mutations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

When should variants identified in tumour be investigated for germline significance?

A

Increased tumour testing increase in detection of secondary/incidental findings – some of which are germline in origin but not feasible to carry out paired tumour/germline on all samples to confirm

ESMO Guidelines for Germline-focussed analysis of tumour-only sequencing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What genes should always be followed up for germline analysis after tumour testing?

A

ESMO guidelines identified 7 most actioonable genes which hava a high germline conversion factor: BRCA1/BRCA2/MLH1/MSH2/MSH6/PALB2/RET

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What do the ESMO guidelines suggest about germline testing of tumour variants?

A

Four potential strategies for clinical labs. Intermediate/conservative suggested for UK/Europe

Permissive: germline follow-up for all 40 genes in all tumour types.

Intermediate-permissive: germline follow-up for all 23 MA-CSGs/HA-CSGs in all tumour types but germline follow-up only in ‘associated’ tumour types for 17 SA-CSGs.

Intermediate-conservative: germline follow-up in all tumour types for the 7 most actionable (MA-CSGs) but germline follow-up only in ‘associated’ tumour types for the other 33 HA-CSGs/SA-CSGs (highly actionable/standard actionable)

Conservative: germline follow-up only in ‘associated’ tumour types for all 40 genes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is an incidental finding?

A

defined incidental findings as “results that are not related to the indication for ordering the sequencing but that may nonetheless be of medical value or utility

Now referred to as secondary findings as incidental gives a sense of insignificance

Off target finding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What guidelines are used to determine when secondary/incidental findings be reported?

A

The ACMG published a minimum list of 59 genes to be reported as incidental or secondary findings

The ACMG subsequently established the Secondary Findings Maintenance Working Group to develop a process for curating and updating the list over time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is required for individuals undergoing clinical genomic sequencing regarding secondary findings?

A

Informed consent and an option to opt-out of receiving secondary findings.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

On what basis were genes included in the ACMG secondary findings list?

A

Based on clinical features, likelihood of early diagnosis, molecular characteristics, clinical testing options, and medical actionability.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

When should secondary findings be reported according to ACGM guidelines?

A

For clinically significant findings that have potential health implications, these include:
- the timing of the impact upon health, when this will come about, now or in the future
- its scope, who it affects, the individual, their offspring or other family members
- its scale, whether its impact upon health is significant or trivial
- The probability of impact, whether the variant is completely penetrant or only marginally so.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Why do some patients receive genetic testing through research?

A

The boundary between clinical and research activities is becoming increasingly blurred, particularly in the subspecialty of clinical genetics and is growing with the use of genomic technology.

This arises because in a resource scarce environment, some genetic tests (e.g for rare diseases) can only be accessed through research protocols because there are no relevant genetic tests validated for clinical use within the NHS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

When should variants identified in research be reported?

A

Beneficence - it has the potential for reducing the risk of, or preventing, disease
Non-maleficence - to do no harm
Justice
respect for autonomy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

When might secondary findings from WGS be repoted?

A

Discussed at GTAB:
- The seriousness of the presenting problem and the nature of the other findings
- Whether the finding represents a known clinical entity or risk factor or a finding that requires further investigation (e.g. variants of uncertain clinical significance, VUCS)
- Whether the finding has been validated to an acceptable standard
- The availability of any treatment/prophylaxis, and its likely success
- Whether the finding is a risk factor for disease or represents a disease process
- The age of the patient and co-existing morbidities and conditions
- Prior knowledge of the patient’s wishes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What does the term “opportunistic screening” refer to in the context of secondary findings?

A

The intentional search for additional pathogenic variants during genomic sequencing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is a bioinformatics pipeline?

A

A pipeline is a term in computer science for chaining or connecting software tools/programs/scripts creating a stepwise workflow to execute the analytical steps necessary for a complete bioinformatic analysis. Each step in the pipeline is typically designed to take input from the previous step and generate output that is used as input for the next step.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What are FASTQ

A

Contains raw sequencing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What are FASTQ files?

A

Text file containing demultiplexed sequence reads

Often used as the first input for bioinformatics pipelines to generate quality and alignment metrics

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What are BAM files?

A

Generated from FASTQs.

Reads are mapped against the reference genomes to allow for aligned reads to be viewed. Also provides some quality information e.g. depth of sequencing at different locations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What are CRAM files?

A

Compressed BAM files to help with space saving

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What is VCF file?

A

File contains information about a position in the genome, usually variants. May also include annotations

This is the output of variant calling section of BI pipeline

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What is a BED file?

A

Stores genomic regoins and is usually used to define the regions fo interest for an assay e.g. variants will only be called within the regions defined by the BED file

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What components are normally included in a bioinformatics pipeline?

A

The input- FASTQ
Quality control- filter out poor quality reads and removing artefacts
Sequence alignment- map to reference genome and produce BAMs
Variant calling- Identify variations between sequence and reference
Variant filtering: filter out false positives or poor quality variants (e.g. due to poor mapping, strand bias)
Variant annotation: characterize variant with location, HGVS, VAF, databases
Variant prioritisation: some pipelines may only show variants that are thought to be clinically relevant, filter out polymorphisms- must be fully validated or could miss important variant

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What are the advantages of using an in house BI pipeline?

A

Full control
- if commercial, an organisation must put their trust into the claims made by a commercial operator
- allows modification of filtering criteria, annotations and databases

Data security
- all in house so less cocersn than with a commercial operator

Cost
- often cheaper than regularly paying for commercial BI

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What are the disadvantages of using an in house BI pipeline?

A

Cost
-Can be costly to set up at the beginning

Expertise
- Requires a high amount of expertise and validation to ensure pipelines are effective and that variants of interest are not filtered out

Time
- likely to take a considerable amount of time to work through bugs and test quality of outputs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Give some examples of cases that should be used when validating a pipeline?

A

Low VAF- close to LOD

Low TC samples

Identify areas of poor quality/drop out

Large deletions/insertions

Horizontally complex variant: 2 or more sequence alterations on same read in close proximity, so that they may resemble single variant.

Vertically complex variant: three or more alleles are represented by different sequence reads

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What are the common causes of poor sequencing data?

A

GC rich regions

Homopolymeric tracts

Strand bias

Artefacts due to poor quality input DNA (e.g. deamination from FFPE)

Insufficient template DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Why might sequencing reads not always align correctly to the reference genome?

A

Variant present which affects alignment
Read maps to multiple locations in the reference genome (e.g. pseudogene)​
Reference genome is incomplete so sequence is missing (e.g. centromeric regions)​
Errors introduced during sequencing​

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

Why might a sequencing read map to more than one location on the reference genome?

A

segmental duplications or pseudogenes can result in the same sequence being present in 2 or more locations in the genome.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Why do pseudogenes make NGS difficult?

A

NGS sequence reads that map to these duplicated regions e.g. psuedogenes will not have unique mapping and therefore may be removed from downstream analyses.

If clinically relevant genes have a pseudogene it may be difficult to get sufficient coverage of the gene for variant calling

Alternatively, called variants may be in the pseudogene and not the gene itself

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What gene is difficult to analyse due to pseudogene?

A

PMS2 and Lynch Syndrome

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What is paired end sequencing and why is this better than single end?

A

Paired-end sequencing- sequence both ends of the DNA fragment producing forward and reverse reads

Paired-end sequencing can be useful for detecting structural variants (deletions, insertions or inversions)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What is FISH, and what are its advantages?

A

FISH (Fluorescence in situ hybridization) is a technique using DNA probes with fluorophore-coupled nucleotides to detect complementary sequences in fixed cells or tissues.

Its advantages include high sensitivity, direct application to both metaphase chromosomes and interphase nuclei, and single-cell level visualization

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

Describe the basic principles of FISH and its technique

A

FISH involves denaturation of probe. Treated with formadimide which reduces the melting point of the DNA and helps in faster hybridization, where the probe binds to complementary sequences on the slide.

The procedure includes slide preparation, codenaturation, hybridization, stringent washing, counterstaining, and visualization using a fluorescent microscope

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are the three main types of FISH probes, and what are their applications?

A

The three main types of FISH probes are locus-specific probes, alphoid or centromeric repeat probes, and whole chromosome probes. They are used for detecting specific genetic regions, determining chromosome number, and examining complex chromosomal abnormalities, respectively.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What are the main clinical applications of FISH?

A

FISH is widely used in oncology for diagnosing and prognosticating various cancers such as sarcoma, CML, CLL, ALL, AML, NHL, and MCL. It helps in defining treatment options, disease monitoring, and treatment response assessment.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What are the advantages of FISH?

A

The advantages of FISH include high resolution, applicability to various cell types (including non-living, fixed, and paraffin-embedded cells), relatively fast results (usually within 24 hours), and the ability to count many cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What are the disadvantages of FISH?

A

The disadvantages of FISH include its limited scope (it will not detect abnormalities in regions not probed), lack of high throughput, and lower resolution compared to PCR techniques.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What are locus-specific probes, and what are their applications?

A

Locus-specific probes bind to particular regions of a chromosome and are used to determine chromosome location or copy number of specific genes. They can be breakapart probes for detecting translocations, dual-color dual-fusion probes for known translocations, or enumeration probes for detecting deletions, duplications, and chromosome ploidy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What techniques can be used to detect copy number changes?

A

G banding
FISH
QF-PCR
RT-qPCR
MLPA
SNP arrays
NGS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What are the disadvantages of G-banding?

A

Low resolution (>5Mb), labor-intensive, slow turnaround time, unable to detect UPD, requires dividing cells and manipulation of the cell cycle, risk of cultural artefacts, and some abnormalities not detected in cultured cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What are the key steps in the principle of MLPA?

A

Hybridisation of probes to DNA, ligation of probes, PCR amplification of ligated probes, separation by capillary electrophoresis, and analysis and quantification.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What is the purpose of the stuffer sequence in MLPA probes?

A

It allows for the generation of different sized products for electrophoretic resolution.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

List the basic procedure steps for carrying out MLPA.

A
  1. Denaturation of genomic DNA
  2. Hybridisation of probes to the sample
  3. Ligation of probes
  4. PCR amplification of ligated probes using universal primers
  5. Separation of amplified products by capillary electrophoresis
  6. Analysis and quantification
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What is MS-MLPA?

A

Allows copy number detection and methylation profiling

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

How does MS-MLPA detect methylation?

A

By using probes with a methylation-sensitive restriction site and restriction enzume Hha1; methylated DNA remains undigested and generates a signal during PCR.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

How does RT-MLPA differ from standard MLPA in its procedure?

A

It requires reverse transcriptase to create cDNA from RNA before proceeding with the MLPA reaction.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What is RT-MLPA used for?

A

mRNA expression profiling.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What are the four main categories of copy number detection methods in NGS?

A

Split Read (SR)
Read Pair (RP)
Assembly-based (AS)
Read Depth (RD)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

Why is the combined analysis (CA) method popular in NGS?

A

Because no single method is comprehensive enough to detect the full range of DNA variations on its own.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What are some limitations of the Split Read (SR) method in NGS?

A

Limited by the length of reads and less reliable in regions with duplications.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What is a primary advantage of Read Depth (RD) in NGS?

A

It is reliable for detecting deletions and duplications and can count the number of CNVs, though it has poor breakpoint resolution.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What is a SNP?

A

Single nucleotide polymorphism (SNP) - a DNA sequence variation occurring commonly within a population (e.g. >1%) in which there is a single nucleotide change e.g. C to T.

On average, SNPs occur every 1000bp meaning that there are roughly 4 to 5 million SNPs per person’s genome.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What are the types of SNPs?

A

Synonymous SNPs (sSNP) do not cause a change in amino acid.

Non-synonymous (nsSNP) when an amino acid is altered and are nonsense or missense

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

Outline the principle of SNP arrays

A

Patients DNA is fragmented and amplified
Bead cheap containing thousands of probes containing common SNPs

Patients DNA hybridized to probe and single base extension with dNTPs. These are fluorescently labelled.

Beads are scanned and the detection system interprets hybridization signal and coverts intensity into genotype

DNA binds if the patient contains the SNPs. This leads to the some heterozygous calls

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What are the two main types of SNP arrays?

A

Illumina
- 850K SNPs
- 3262 target regions
- Uses single base extension with a fluorescently labelled dNTP (green if one base and red if another)

Affymetrix array
- 1.8 million markers. 946,000 for CNV detection, 906,600 SNPs
- All targets bind all probes and the presence of SNPs reduce the affinity of a signal and weaker fluorescence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What is the B allele frequency?

A

By chance, people will be homozygous for some SNPs and heterozygous for other SNPs. The SNPs selected in the bead-chip type arrays are carefully chosen for variability within a population.

In the B-Allele Chart, BB homozygotes have a data value of 1.0, AA homozygotes have a data value of 0.0 and AB heterozygotes have a data value of 0.5. This results in three clusters on the BAF plot

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

How can the BAF plot show a copy number gain or a loss?

A

AB genotype

Normally: B/A- 0.5 BAF

Gain
- Duplication- 0.33 or 0.66 BAF

Deletion
- BAF 0 or 1

Can also be used to determine clone level- based on how separated the BAF is

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

How can BAF be used to identify copy neutral loss of heterozygosity ?

A

Both copies of the chromosome are from the same parent (either inherited or acquired in cancer), there will be two copies of each SNP, but because both regions are identical, every SNP will be homozygous. This will look the same as a deletion in the B-Allele frequency chart, but no copy number change will be visible in the LogR ratio chart.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What can cause CN-LOH

A

LOH as a cancer mechanism

Uniparental disomy (inherited or acquired)

Consanguinity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What are the advantages of SNP arrays?

A

Can detect CN-LOH

Higher resolution than G banding

More accurate clone level compared to G-banding, FISH but limited below 20%

Less subjective analysis

High throughput

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What are the disadvantages of SNP arrays?

A

Can’t detect balanced translocations

5% limit of detections

Can’t detect SNVs

High level of artefacts

Quite laborious

Clonality limited to 20%ish so can’t be used for MRD

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What diseases regularly use SNP arrays?

A

MDS
ALL
MM
CNS (methylation array)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What can SNP arrays be used to detect in cancer?

A

LOH

CNVs

Methylation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What is the difference between array CGH and SNP arrays?

A

Array CGH: It focuses on detecting variations in DNA copy number by comparing the fluorescence intensity of a test sample against a reference sample.

SNP Array: In addition to detecting copy number changes, SNP arrays assess the presence of single nucleotide polymorphisms SNPs and uses in silico tools for CNV calling.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What are Array Comparative Genomic Hybridisation (CGH)?

A

Patient and control DNA are labeled with fluorescent dyes and applied to bead chip with thousands of probes for regions fo interest

Patient and control DNA compete to hybridize which is then scanned my microarray

If gain then patient DNA will be more present than control and if loss then control DNA more present

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What are the advantages of oligo array CGH?

A

Can detect genomic imbalances as small as 100bp, depending on coverage

More cost effective

better reproducibility and less batch-to-batch variation

Multiple consecutive probes indicating the same copy-number change are required to determine a gain or loss. This enhances the accuracy of the interpretation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What are the disadvantages of oligo array CGH?

A

Low detection frequency of mosaicism (<30% of abnormal cells)

False positives

Can’t detect LOH or UPD

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What are Bacterial Artificial Chromosome (BAC) arrays?

A

Type of CGH

BAC clones are propagated in vectors in bacteria, purified, amplified and then spotted onto a glass slide using ultra fine needles. Multiple copies of each BAC are spotted onto the array and distributed across the array

Due to the large size, BACs are very stable and hybridisation is specific

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What are the advantages of BAC arrays?

A

Less CNVs of uncertain significance detected

High signal to noise ratio

accurate copy number information

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What are the disadvantages of BAC arrays?

A

Abnormalities may be missed as unable to distinguish gains/losses <85kb or those which fall in 600kb gaps,

LOH/UPD not detected

less sub arrays per slide therefore expensive

Not really used in routine practice

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What are expression arrays

A

Expression arrays allow the simultaneous investigation of the expression of thousands of genes and typically involve comparing two or more highly related cellular or tissue sources that differ in an informative way.

Used to investigate expression profiling in tumours and expression profiling of MicroRNAs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What kind of methylation arrays are there?

A

Bilsulphite modification followed by microarray hybridisation: CpG islands in promoters of specific genes are hypermethylated in cancer genomes. Sodium bisulphite converts cytosine to uracil but leaves 5-methylcytosine (methylated cytosine) unchanged. Oligonucleotide probes on the microarray hybridise specifically to either the converted or unconverted sequence.

ChIP-on-chip, which locates protein binding sites that may help identify functional elements in the genome. For example, in the case of a transcription factor as a protein of interest, one can determine its transcription factor binding sites throughout the genome.

DamID (DNA adenine methyltransferase identification): an alternative to ChIP-chip, transcription factor or chromatin-binding proteins of interest are fused to DNA adenine methyltransferase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

Give an example of expression arrays in tumour profiling

A

Microarrays are now widely applied to the study of human cancer, for delineating molecular subtypes, and for assessing disease progression and treatment response

Breast
- 70-gene prognostic signature (Mammaprint) developed on Agilent platform found to be strong predictor for metastasis-free survival
- not yet used in diagnostic setting

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

Briefly define RNA interference (RNAi) and how recent array technologies have contributed in related research studies.

A

RNA interference (RNAi) is a post-transcriptional method of gene silencing.

The technique involves printing RNAi reagents onto a standard glass microarray slide, which is then placed in tissue culture dishes and cultured cells in medium are added to the arrays.
Cells that adhere to the spots internalise the printed material and become transfected, leading to silencing of a specific gene, and the remaining cells form a non-transfected lawn between spots.

Microarrays are then fixed and prepared for immunofluorescence, staining for DNA and F-actin, in situ hybridisation, apoptosis detection or other assays.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What are copy number variants?

A

A copy number variant (CNV) is a loss (deletion) or gain (duplications and triplications) of genomic material relative to the reference genome. CNVs can be intragenic, typically deletions/duplications larger than 50bp, or intergenic, involving multiple genes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

Name some of the current best practice guidelines used to interpret CNVs

A

Intergenic CNVs: The ACMG technical standards for the interpretation and reporting of constitutional copy-number variants- these guidelines are used for both rare disease and inherited cancer but not for acquired neoplasia

Intragenic CNVs: ACGS Best Practice Guidelines for Variant Classification in Rare Disease 2023 and CanVIG-UK Consensus Specification for Cancer Susceptibility genes (CSGs) of ACGS Best Practice Guidelines for Variant Classification 2023.

Acquired CNVs: The 2020 ACMG technical standards do not apply to acquired CNVs in neoplasia and there are currently no consensus guidelines adopted in the UK.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

Outline what evidence is used in the interpretation of CNVs

A

Population frequency: presence in unaffected individuals supports benign classification. E.g. Database of Genomic Variants (DGV), gnomAD, In-house laboratory databases

Genomic Content of CNV: Includes known pathogenic or cancer associated genes.

DECIPHER database, COSMIC, In-house databases, ClinVar.

Professional guidelines including NICE, NCCN, WHO

Size of CNVs- how many coding genes are involved.

Haploinsufficiency vs triplosensitivity: Consider whether one allele sufficient for normal function or if an additional copy would change the function.

Literature evidence: has this CNV been reported previously in an affected individual.

94
Q

Define the terms haploinsufficient and triplosensivity

A

Haploinsufficiency is the most common mechanism of pathogenicity for heterozygous deletions where loss of one allele resulting in a single normal allele at a particular locus is inadequate for normal function resulting in a phenotype. Regions of copy number loss containing established HI genes are more likely to be pathogenic.

TS is where an additional copy of a gene/genomic region results in a phenotype. Fewer genes appear to exhibit TS than HI and therefore copy number gains are more likely to result in a milder phenotype or to be benign than the reciprocal deletion.

95
Q

Give 2 examples of inherited cancer syndromes that require CNV testing and the gene targets associated

A

Inherited breast and ovarian cancer: intragenic CNVs BRCA1; BRCA2; PALB2; ATM; CHEK2; RAD51C; RAD51D.

Lynch syndrome: Intragenic CNVs in MLH1, MSH2, MSH6, PMS2

Usually tested for by NGS or MLPA

96
Q

Give 2 examples of acquired cancers that require CNV testing and the gene targets associated

A

Myelodysplastic syndrome (MDS): genome wide screen of copy number changes including -7/del7q, -5/del5q, i(17q)/t(17p), -13/del13q, +8; +19. Usually tested for by G banding or SNP arrays

Non small cell lung cancer (NSCLC): MET amplification. Usually tested for by FISH or NGS

97
Q

What technologies have superseded mutation scanning technologies in routine diagnostic testing?

A

Used historically as diagnostic pre-screening tool when DNA sequencing was labour intensive. This is now largely superseded by NGS and arrays and the scanning techniques below are no longer used routinely in diagnostic genetics laboratories.

98
Q

What is pyrosequencing?

A

Pyrosequencing is a sequencing method where pyrophosphate release during nucleotide incorporation generates a light signal that indicates the sequence.

99
Q

What are the principles of pyrosequencing?

A

DNA is labelled with biotin and amplified

During sequencing, each addition of a dNTP releases a pyrophosphate which is coverted to ATP

Luciferase uses ATP to produce light

This is measured and produces a trace where the peaks are proportional to the number of nucleotides added

100
Q

What are the pros and cons of pyrosequencing compared to Sanger sequencing?

A

Pros include fewer steps and better detection limits (~5%).

Cons include shorter read lengths (100-400 bp) and issues with homopolymers, potential for mutation masking due to suboptimal dispensation

101
Q

What is pyrosequencing used for?

A

Methylation analysis with bisulphite conversion (can be used for MLH1 promoter hypermethylation)

Detecting KRAS mutations and quantifying mutant alleles, which helps in selecting patients for TKI therapy in NSCLC.

102
Q

What is electrophoresis?

A

Electrophoresis is a technique used to separate DNA fragments according to size using an electric field running through a matrix.

DNA is negatively charged, so it migrates through agarose gel to the positively charged anode.

103
Q

What is a key advantage of polyacrylamide gel electrophoresis (PAGE)?

A

PAGE is more sensitive than agarose gels and can resolve fragments less than 50 bp in length.

104
Q

What is the purpose of capillary electrophoresis in DNA fragment sizing?

A

Capillary electrophoresis uses fluorescent tags to size DNA fragments within 1 bp of each other.

105
Q

What does the Agilent Bioanalyzer do?

A

Capillary electropheresis

It performs size fractionation and quantification of DNA, RNA, or protein samples using nanofluidics, electrophoresis, and flow cytometry

106
Q

What methods can be used to detect methylation?

A

Pyrosequencing
Combined Bisulphite Restriction Analysis (COBRA)
Methylation-sensitive melt curve analysis
MS-MLPA
NGS
Arrays

107
Q

What is DNA methylation?

A

An epigenetic event that alters gene expression by the addition of a methyl group to the 5-carbon of cytosine in a CpG dinucleotide, catalyzed by DNA methyltransferase (DNMT).

108
Q

Why is DNA modification necessary in methylation studies involving PCR?

A

DNA polymerase cannot distinguish between methylated and unmethylated cytosines during PCR, so DNA must be modified to preserve methylation information.

109
Q

What is bisulfite modification used for in targeted methylation analysis and how does it work?

A

To convert unmethylated cytosines to uracil, allowing differentiation between methylated and unmethylated cytosines.

Sodium bisulphite deaminates unmethylated cytosine to uracil

The methyl group on 5-methylcytosine protects the amino group from the deamination.

110
Q

How does pyrosequencing detect methylation?

A

Uses bisulphite coversion first

Unmethylated regions will show C to T changes and methylated regions will retain C

111
Q

What does Combined Bisulfite Restriction Analysis (COBRA) rely on?

A

The differences in sequence between methylated and unmethylated DNA after bisulphite modification can lead to the creation of new methylation-dependent restriction sites (e.g. conversion of CmCGA to the Taq I recognition site TCGA) or the maintenance of restriction sites in a methylation-dependent manner.

The COBRA assay, which relies on the separation of digested PCR products followed by hybridisation of fluorescently labelled probes.

Bisulfite conversion of non-methylated cytosines to uracils,

Locus-specific PCR amplification of converted DNA,

Restriction digestion,

Analysis of restriction patterns on the gel, and the quantification of these restriction patterns using ImageJ or a similar program

112
Q

What are the disadvantages of COBRA for methylation?

A

not all sequence changes resulting from bisulphite modification result in the formation/abolition of a commercially available restriction enzyme site.

Technique with restriction digestion post-PCR is prone to error due to the formation of heteroduplexes between the converted methylated and unmethylated DNA. This heteroduplex will not be cleaved by restriction enzyme.

113
Q

What is methyLight?

A

RQ-PCR method for Methylation

ommercial TaqMan assays using MethyLight technology are available for CDKN2A, ERBB2 and others

Uses methylation insensitive TaqMan probes which target regions in between methylation-specific primer sites (2 different primer pairs are used in separate reactions, one specific for methylated and converted DNA, the other specific for unmethylated and converted DNA).

114
Q

How do methylation arrays work?

A

Bisulphite modification of DNA

Whole-genome multiple displacement amplification via random hexamer priming and Φ29 DNA polymerase, which has a proofreading activity resulting in error rates 100 times lower than the Taq polymerase. Enzyme shearing.

Hybridisation and single step extension

Fluorescence staining and chip scanning

Repeated round of staining of hapten-labelled dNTPS

Scanning and data analysis

115
Q

What methylation arrays are available?

A

Illumina currently market The HumanMethylation27 BeadChip which allows researchers to interrogate 27,578 CpG loci, covering more than 14,000 genes.

The HumanMethylation450 BeadChip offers coverage of >450,000 methylation sites, and is marketed as being high throughput and low price, therefore suitable for screening genome-wide association study (GWAS) populations, or tumour samples for tumour-wide methylation status.

116
Q

How does MS-MLPA work?

A

Use of a methylation-sensitive restriction enzyme HhaI

Two reactions: undigested for CNV and digested for methylation

Hybrids of probes and unmethylated sample DNA are digested by the HhaI enzyme. Digested probes cannot be amplified during PCR and hence will not produce a signal during capillary electrophoresis. In contrast, if the sample DNA is methylated, the DNA-probe hybrids are protected against HhaI digestion and the ligated probes will generate a peak.

117
Q

What sequencing techniques can be used to detect methylation?

A

Long read sequencing: nanopore

118
Q

How is methylation testing used in colorectal cancer?

A

Hypermethylation of the hMLH1 promoter, which results in transcriptional silencing of hMLH1 and is commonly seen in sporadic colon and endometrial cancers with microsatellite instability (MSI) is now routinely offered in diagnostic labs.

routinely offered in diagnostic labs.

Analysis of tumour DNA can aid discrimination between those individuals likely to have sporadic forms of colon and endometrial cancer and those likely to benefit from mutation screening of Lynch Syndrome genes.

119
Q

Why is MGMT methylation analysis used?

A

Hypermethylation of the MGMT promoter is associated with chemosensitivity and is seen in a wide spectrum of tumours, including gliomas. Therefore, use of this kit can enable identification of those tumours likely to respond to treatment with alkylating agents

120
Q

Why is methylation analysis used in CNS tumours?

A

WHO 2022 includes methylation status in classification of CNS tumours

DNA methylation has been shown to stratify meningiomas into methylation classes that more accurately than histopathology identify patients at high risk of recurrence. Molecular classification of meningiomas based on copy number variation, point mutations, methylation, and transcriptomic and proteomic data stands out as a future diagnostic work-up of meningiomas.

121
Q

What are the advantages of methylation analysis?

A

Important in lynch (reduces germline screening) and CNS and classification of many tumours

Techniques can involve lab standard protocols such as PCR and may not require additional equipment

Techniques can be flexible and designed for specific targets

Some techniques such as RT-PCR can be used for a wide range of samples e.g. fresh, frozen, or formalin-fixed paraffin-embedded tissues and remote samples, such as serum, plasma, and urine

122
Q

What are the cons of methylation analysis?

A

Techniques can be time-consuming, prone to contamination, and difficult to interpret in non-binary samples.

123
Q

What is RQ-PCR?

A

Real time quantitive PCR

Patient RNA converted to cDNA with reverse transcriptase

A fluorescently labelled probe for the region of interest has a quencher molecular and binds to the region on cDNA

cDNA is amplified with taq polymerase, this digests the probe releasing the quencher

This increases the fluorescence with each round of amplification

This is measured in real time and the ct value is taken (the number of cycles taken to pass background fluorescence). This is directly proportional to the amount of template present

ct value is then compared to standard curve of known concentrations for absolute quantification

Normalised to housekeeping gene (ABL1)

124
Q

How does fluorescence resonance energy transfer (FRET) hybridisation work?

A

Pair of fluorescent probes labelled at 5’ and 3’

When bound these probes are end-to-end allowing a transfer of energy from one fluorophore to the other

PCR extension cleaves the probe and it is unable to transfer energy

This allows detection during annealing or extension phase

Perform melt curve analysis

125
Q

What is RQ-PCR used for?

A

qPCR (commonly Taqman) is used for monitoring MRD

CML – qPCR of BCR-ABL1 transcript levels is useful for the early detection of relapse of CML. During treatment, European LeukaemiaNet (ELN) currently suggests BCR-ABL1 RQ-PCR every 3 months until a major molecular response is confirmed, then every 3-6 months (PB is sufficient).

ALL – to monitor gene fusions (e.g. BCR-ABL1, MLL-AF4, TCF3(E2A)-PBX1, and ETV6-RUNX1), or if absent, a rearrangement of immunoglobulin or T-cell receptor genes resulting in unique molecular signature.

AML – to monitor gene fusions (e.g. PML-RARA, RUNX1-RUNXT1 and CBFB-MYH11), mutations (e.g. NPM1) and transcripts that are commonly unregulated in AML, particularly WT1. The MRC AML15 trial showed sequential monitoring by RQ-PCR for the PML-RARA transcript is the strongest predictor of relapse in APML.

126
Q

What is the principle of ddPCR?

A

Sample divided to 20,000 tiny droplets, allows thousands of independent amplification events in a single sample.

Each droplet contains either a positive (fluorescent) or negative (non-fluorescent) template.

This digital counting directly translates to the absolute number of target molecules

A Taqman mastermix is prepared with fluorescent probes, primers, and the template nucleic acid. A control signal (ROX) indicates the presence of DNA in the droplet, while target and reference signals are typically FAM and VIC.

The quantity of the target can be calculated either absolutely or relative to a reference.

127
Q

How is the absolute quantification of ddPCR determined?

A

The exact number of fragments per partition is not known – there could be none or more than one fragment present in each partition.

The Poisson distribution is the statistical probability of each partition containing 0, 1, 2, 3, 4+ fragments of DNA – this is used to correct and account for fragment number variability.

The equation used for this correction is A=– loge(1–P) where A is average number of fragments per partition, and P is the proportion of positive partitions.

128
Q

How is the relatively quantification of ddPCR determined?

A

The number of positive partitions is compared to the number of reference partitions e.g. variant allele v wild type allele.

The quantity of the reference is normally known before e.g. a diploid cell is known to have 2 copies.

The Poisson correction equation is applied first, then the quantity is normalised to the reference quantity to give relative abundance.

129
Q

What are the advantages of ddPCR?

A

High sensitivity- the presence of a single target molecule within a droplet is sufficient for detection

Highly accurate

Reduced Bias: minimizes bias inherent in traditional PCR methods, such as competition between target and reference sequences and challenges associated with determining Ct values.

Low input volume required 

130
Q

What are the limitations of ddPCR?

A

Cost: ddPCR instruments and consumables tend to be more expensive compared to conventional PCR setups.

Throughput: Due to individual droplet analysis, ddPCR has a lower throughput

Technical Expertise: ddPCR workflows can be more complex

Ultra-sensitivity means contamination is easier.

Uniformity of partition size and correct starting dilution of DNA are essential for accuracy.

Can only detect a single variant in each assay.

131
Q

What are the applications of ddPCR?

A

JAK2 V617F in MPN

EGFR Thr790Met (T790M) testing of ctDNA in relapsing lung cancer patients

HER2 copy number in Breast Cancer Patients

KIT Asp816Val (D816V) for suspected systemic mastocytosis

BCR::ABL1 monitoring in CML - mainly RQ-PCR used as specified by guidelines

132
Q

What are the principles of IHC?

A

A solution containing the primary antibody is added to the tissue section which binds to target protein

Unbound and surplus antibodies are washed away and the secondary antibody is added.

The secondary antibody, which carries a linker molecule with horseradish peroxidase (HRP) enzymes binds to the primary antibody, followed by another washing step. After this, 3,3’ Diaminobenzidine (DAB) is added.

The HRP enzyme transforms the DAB substrate into a brownish precipitate that is deposited in the tissue at the site of the reaction, thus producing a visual representation of where the primary antibody first bound its target.

133
Q

What tissues can be used for IHC?

A

Tissues are typically fixed with formalin and embedded in paraffin wax (FFPE) to preserve the epitopes and morphology.

Frozen tissue sections can also be used

134
Q

What is antigen retrieval in IHC?

A

Formalin can mask protein targets. This step helps “unmask” them for antibody binding.

The most common method for antigen retrieval in FFPE samples is to pressure-boil the tissue slides in an acidic citrate buffer for around 15-20 minutes

135
Q

What are the two main types of antibodies used in IHC?

A

Polyclonal: Diverse antibodies targeting various regions of the protein. sensitive but potentially less specific

Monoclonal: Highly specific antibodies targeting a single protein region. can still generate results that are hard to interpret if the target epitope is present in low abundance.

136
Q

What are the advantages of IHC?

A

Enables assessment of protein expression and localization.

Well-established and widely used in pathology labs.

Provides spatial information within the tissue context.

Rapid and cheap

137
Q

What are the disadvantages of IHC?

A

Limited to known protein markers.

Does not directly detect genetic mutations.

138
Q

What is sanger sequencing?

A

Sanger sequencing gives high-quality sequence for long stretches of DNA (900 bp).

Gold standard- often used to confirm NGS

139
Q

What are the principles of sanger sequencing?

A

Template DNA denatured and primers annealed

This chain-termination method makes use of chemical analogues of the dNTPs called dideoxy nucleotides (ddNTPs). The ddNTPs lack a hydroxyl group (OH) on the 3’ carbon of the sugar ring needed to form the phosphodiester bond between one nucleotide and the next during DNA strand elongation. This inhibits further strand extension.

The chain ends with the dideoxynucleotide, which is marked with a particular colour of dye/radioactive label depending on the base (A, T, C or G) that it carries.

Exonuclease I (Exo) digests residual single-stranded DNA primers and any extraneous single-stranded DNA fragments produced during the PCR process. Shrimp Alkaline Phosphatase (SAP) dephosphorylates remaining dNTPs (free nucleotides) from the PCR product so they do not interfere with the sequencing reaction

As a result, different DNA fragments of varying length are produced.

These differently sized fragments can be size-fractionated on a polyacrylamide gel or by capillary electrophoresis.

140
Q

How is sanger sequencing analysed?

A

A commonly used program is Mutation Surveyor. The software translates the raw data into the corresponding nucleotide bases, and also assigns a quality score to each base, which predicts the probability of a base call error (i.e. how reliable the call is for any given base).
Forward sequence shows clear and distinct peaks.

141
Q

What are some advantages of sanger sequencing over NGS?

A

Cost-efficient sequencing of single samples

In some cases, analysis of longer fragments (~1,000 bp in length)

Less error prone so can confirm NGS or fill in the gaps needed for NGS

Less reliant on computational tools

Requires more DNA and higher quality

142
Q

What are the limitations of sanger?

A

SNPs under probes can prevent amplification

Poor sequencing close primer

Sensitivity limited to 15-20%

Requires high DNA input

Low throughput

Only analyse 1 target

143
Q

What is next generation sequencing?

A

High throughput massive parallel sequencing

144
Q

What are the stages of NGS sequencing?

A

DNA/RNA extraction
Library preparation and enrichment
Sequencing by synthesis

145
Q

How are sequencing libraries prepared for NGS?

A

Fragmentation: Break the DNA/RNA into smaller fragments 300-400bp.
Methods: Mechanical shearing (e.g., sonication), enzymatic digestion, or tagmentation (transposon-mediated fragmentation and tagging).

End Repair and A-tailing: Prepare DNA ends for adapter ligation. Enzymatic treatments to produce blunt ends, followed by the addition of an adenine (A) overhang.

Adapter Ligation: Attach platform-specific adapters to the DNA fragments and indexes. Ligase enzyme to attach adapters, which contain sequences necessary for binding to the sequencing platform.

PCR Amplification: Amplify the library to obtain sufficient quantities for sequencing

146
Q

What enrichment strategies are used in NGS?

A

Hybrid Capture
- Enrich specific regions of interest from the genome by uses biotin labelled probes specific to target regions and using magnetic beads to pull down the probes along with the sequence. This is then eluted

Amplicon based
- Amplify specific regions of interest using PCR and primers for specific regions

147
Q

What are the advantages and disadvantages of hybrid capture and amplicon based enrichment?

A

Hybrid
- Highly specific and uniform as no amplification
- Scalable for large panels

Amplicon
- highly specific
- quick and cost effective
- but potential amplification bias

148
Q

How do illumina sequencing work?

A

Flow cell contains probes complimentary to the library adaptors which binds the library fragments. This is extended and bends over and binds to another probe- bridge amplification. This results in clusters all across the flow cell.

Each dNTPs are fluorescently labelled and as it is added it is excited and scanned. The consensus fluorescence at each position in each cluster is used to determine the nucleotide

149
Q

How does ion torrent sequencing work?

A

Uses amplicon based enrichment

Flow cell contains millions of wells with semi conductors.

Sequencing by synthesis- as each base dNTP is added to a H+ ion is released changing the pH. The semi conductors can determine the base at each position based on the change in pH- massive parallel sequencing

150
Q

What are the key types of NGS?

A

Targeted panels
Whole Exome Sequencing
Whole genome sequencing
Whole transcriptome sequencing
Epigenomics

151
Q

What are the advantages of NGS?

A

Cost-effectiveness,
unprecedented sequencing speed,
high resolution (detect SNVs, CNVs, indels)
Highly accurate (although sometimes confirmed with sanger)
Analyse large numbers of genes at once
High throughput

152
Q

What is limitations of NGS?

A

Massive volume of data generation
- creates problems with data storage
- creates large amount of data which requires interpretation

Short reads so is unable to call large structural variants

Accuracy reduces towards end of reads- phasing

pseudogenes and repetitive sequences are difficult to sequence

153
Q

What is sequencing depth (coverage)?

A

The average number of times a base pair is sequenced

154
Q

How do NGS platforms ensure the quality of individual base calls?

A

By providing confidence scores for each base call, allowing for quality filtering during data mining

155
Q

What is bias in NGS coverage?

A

High GC content reads can be favoured in sequence leading to non-uniform coverage

Nonuniform coverage can leave portions of the genome undersequenced, making it difficult to identify SNPs, point mutations, or structural variants.

Deeper sequencing can help to compensate for this limitation

156
Q

What is the goal of personalized medicine?

A

To customize health management based on an individual’s genetic profile, improving disease prevention, early detection, and treatment.

NGS has helped improve this

157
Q

What is the 100,000 Genomes Project?

A

An NHS initiative to decode the human genome in people with rare diseases and cancer to predict disease development, diagnose conditions, and identify treatments.

158
Q

What is the National Genomic Test Directory and how was it developed?

A

Identifies the most appropriate tests for each clinical indication and the testing methodology by which it should be delivered.

Through expert panels including clinicians, scientists, health economists, policy experts, public representatives, and patient organizations.

159
Q

What is third generation sequencing?

A

The term generally given to technologies capable of sequencing single DNA molecules without amplification.  Also known as long-read sequencing (LRS).

Removes the need for PCR-based amplification or the requirement to halt between read steps

160
Q

What are the advantages of third generation sequencing?

A

Small amount of starting material

Higher throughput –hundreds to millions of sequencing reactions can be carried out asynchronously.

Lower projected cost per base

Longer read lengths (10,000-100,000bp)- allowing enhanced de novo assembly; mapping certainty; haplotype detection; chromosome phasing; CNV detection; recognition of insertions, deletions, and translocations

Sequencing of repetitive elements

Provide discrimination between genes and pseudogenes.

Produces more uniform coverage of the genome- less sensitive to GC content than SGS (dependent on tech).

Potential to detect epigenetic modifications such as methylation.

Redundant sequencing will result in higher consensus accuracy to enable rare variant detection and simplified data analysis.

No amplification so less bias

161
Q

What are the disadvantages of 3rd generation sequencing?

A

Higher error rate per read;
fewer reads per run;
more expensive per base (more relevant to PacBio than ONT)
limited standardisation raises challenges for use in clinical diagnostics.

162
Q

What are some types of third generation sequencing?

A

PacBio Single molecule real time (SMRT) sequencing

Oxford Nanopore

FRET sequencing (Life Technology)

163
Q

What is PacBio SMRT sequencing?

A

This method is based on monitoring polymerase activity while incorporating differently labelled nucleotides into the DNA strand. Each nucleotide carries a base-specific fluorescent label on its phosphate group, which is released when being incorporated by the polymerase. dNTP incorporation on each single molecule template per well is continuously visualised, with a laser and camera that records the colour and duration of emitted light
DNA sequencing is performed on SMRT cells, which contain tens of thousands of ZMWs.

SMRT technology utilises zero-mode waveguide (ZMW) – a hole measuring 30-70 nm in diameter, fabricated in 100nm thick metal film deposited on a silicon dioxide substrate. It can detect the activity of a single molecule amongst a background of thousands of labelled nucleotides

164
Q

What are the advantages of pacbio SMRT?

A

Fast – DNA polymerase continues to incorporate bases at a speed of tens per second.

Circular consensus sequence- reduce error rates to from 1-5% to 0.1-0.5% (>Q30).

Discriminate between methylated and unmethylated bases- The polymerase pauses for longer at modified vs. unmodified bases, which can be detected by measuring a metric called the interpulse duration.

165
Q

What are the disadvantages of pacbio?

A

Limited throughput: Limited number of ZMWs that can be read per SMRT cell. Also, longer molecules take longer to pass through the polymerase in the ZMW.

Expensive ~$1000/Gb.

166
Q

What is oxford nanopore?

A

3rd gen sequencing

ssDNA molecule is electrophoretically driven through a nanoscale pore (a very small hole through a thin membrane submerged in salt buffer solution).

When an electrical potential is applied to solutions on either side of the pore, it drives dissolved salt ions through the pore, establishing electrical current.

Molecules passing through the pore can measurably change the ionic current, and the amplitude and duration is determined by the molecule

Due to the length of the pore the shift in voltage due to the presence of a DNA molecule is caused by the string of bases (called a k-mer) within the pore rather than by an individual nucleotide- this detects the composition of the DNA molecule

167
Q

What are some types of nanopores?

A

Can be biological or synthetic (synthetic are more stable but only biological have been used)

Biological
- α-hemolysin- This is an exotoxin secreted from Staphylococcus aureus.
- MspA- mycobacterium smegmatis porin A channel is 1-1.2nm at its narrowest point.
- Bacteriophage phi29.

168
Q

What is a disadvantage of oxford nanopore and why?

A

There are more than 1000 possible signals instead of just 4– one for each k-mer (which is increased if modified bases are taken into account).

This results in a large error rate of up to 30%, particularly with indels, and homopolymers longer than the k-mer length can be difficult to identify.

Can also miss SNPs due to the speed that DNA passes through the pores

169
Q

What are some nanopores?

A

MinION
- Portable
- Max time 72 hours

GridION
- 5 flow cells

PromethION
- 48 flow cells

170
Q

What is FRET sequencing?

A

FRET fluorescence resonance energy transfer. DNA polymerase is tagged with a fluorophore that when brought into close proximity to a nucleotide, tagged with an acceptor fluorophore, would emit a fluorescence resonance energy transfer (FRET) signal. After incorporation, the fluorophore label on the nucleotide can be released

gDNA is chemically attached to the coverslip of a standard TIRF-based (Total Internal Reflection) microscope. Universal primers are annealed to the gDNA. Nanoparticle-labelled DNA polymerase is added to the slide. Dye attachment to the nucleotide is via the terminal phosphate, released into solution after base-incorporation.

171
Q

What are third generation mapping technologies?

A

Mapping technologies determine the large-scale sequence structure of DNA without sequencing every base

Irys system from BioNano Genomics

Dovetail Genomics cHiCago protocol

172
Q

What is the analytical process for WGS?

A

Primary analysis – involves the alignment of the short reads of the sequencing data (FASTQ) to a reference sequence, with the resulting alignments typically stored in binary alignment/map (BAM) file format.

Secondary analysis – is the calling of the somatic variants in the BAM files which creates an output as a variant calling file (VCF)

Tertiary or downstream analysis – involves the interpretation and annotation of the somatic variants called in the VCF as well as identifying mutational signatures, tumour mutation burden (TMB), HRD, MSI

173
Q

What is required for tumour WGS?

A

Tumour sample

Germline sample- to determine which variants are somatic and which are germline

Record of discussion (consent form)

Referral form (TOF)

174
Q

What QC metrics are required for WGS?

A

samples (tumour and normal) are not contaminated by DNA from another patient.

sequencing data are of sufficient quality with uniform coverage, the DNA fragment has a length of ~400 bp, there is a high mapping rate of sequenced reads to the reference genome and there is a low number of chimeric fragments.

tumour content (purity or cellularity) is above the limit of detection for somatic variants (>30%)

tumour in normal contamination (TINC) level for haematological samples is low. High TINC can result in inappropriate subtraction of true somatic variants resulting in false negatives.

175
Q

Why are virtual panels used in WGS?

A

Interpretation of somatic mutations can be particularly challenging as some tumour types can carry tens or even hundreds of thousands of somatic mutations, yet only a small number justify clinical review. Therefore, variant prioritisation is essential, but requires careful design and implementation to ensure that clinically relevant variants are not missed.

The virtual panel used is dependent on the sample type.

176
Q

How are WGS variants classified into domains?

A

Domain 1: Variants in a virtual panel of potentially actionable genes related to tumour type are reported in domain 1

Domain 2: Variants in a virtual panel of cancer-related genes (536 genes, listed in the Cancer census genes)

Domain 3: Small variants in genes not included in domains 1 & 2 are reported in domain 3.

177
Q

How is WGS data presented?

A

Circus plot key:
Track 1 (innermost track): chromosomes. Tracks 2 and 3: number of somatic single nucleotide variants (in red) and indels (in green)
Track 4: ratio of normalised depth of coverage for tumour versus normal Copy number losses are indicated in red, CNVs indicated
Track 5: absolute depth of coverage in tumour sample. Structural variants are indicated by arcs inside the plot

178
Q

How are germline WGS variants reported?

A

Tier 1: pathogenic or likely pathogenic variants conferring susceptibility to the relevant clinical indication using a tumour-type specific panel. For genes with a biallelic mode of inheritance, only homozygous or potential compound heterozygous variants are reported.

Tier 3: Variants are prioritised to tier 3 using a broad gene panels spanning cancer susceptibility genes in addition to the tumour-type specific panel.

179
Q

What are the advantages of WGS?

A

Every single variant in the genome is identified by one test – may reduce need for multiple tests of same sample in the future.

High resolution of WGS- potential to replace SNP arrays and allows identification of the precise location of fusion breakpoints (in frame)

Distinguish between somatic or germline variants.

Identifies biomarkers with emerging clinical actionability (e.g. TMB, mutational signatures for HRD and MSI etc)

Reanalysis for any targets

180
Q

What are the limitations of WGS?

A

Lower read depth coverage compared with targeted NGS panels means reduced sensitivity so low-level variants could be missed.

Tumour in normal contamination (TINC) - a risk of an increased number of false negative somatic variants as true somatic variants may be inappropriately subtracted in the analysis (most commonly observed in haematological cancers)

Long TaT

FFPE is not suitable for WGS due to poor quality, uses fresh frozen

Germline sample difficult to ensure cancer free in HO conditions- needs to be post treatment blood or skin/saliva (dependant upon condition)

Complex consent process

181
Q

What cancers are currently eligible for WGS?

A

Does not replace standard of care

all paediatric cancers, neurological and sarcomas and acute leukaemias presenting at any age,

182
Q

What is the difference between cfDNA and ctDNA?

A

cfDNA are fragmented DNA that have been released from cells and are circulating in the blood stream

This is found in both healthy and cancer populations

Cancer populations have a higher level of cfDNA in the blood due to the presence of circulating tumour DNA (ctDNA) which has been released from tumour cells

Different tumours shed different amounts of ctDNA

183
Q

Can ctDNA and cfDNA be distinguished from?

A

It is not possible to be sure that a sample has ctDNA in it- limitation of the test

Presence of cancer specific abnormalities may help determine

It is thought that ctDNA tends to be shorter fragments than cfDNA which can be used to help determine ratio e.g. with tapestation

184
Q

How does ctDNA arise?

A

Tumour cells dying though necrosis and apoptosis release ctDNA from tumour cells

DNA released from apoptosis is normally around 160-200bp and necrosis is larger

Found to be poorer integrity than cfDNA

185
Q

How is ctDNA obtained?

A

Normally extracted as cfDNA, not specifically ctDNA

Extracted from plasma portion of EDTA whole blood sample which has been separated by centrifugation

Number of extraction kits available e.g. QIAamp, cobas, genexus purification system

Bead based methodologies such as MagMax are thought to favour short fragments and help select for ctDNA

186
Q

What pre-analytical variables impact ctDNA analysis?

A

Blood storage
- If using EDTA tubes, ctDNA needs to be processed within 6 hours due to high clearance and degradation
- Better to use cell stabilization tubes such as streck tubes- can stabilize for up to 7 days
- No refrigeration- results in more cell lysis which will release gDNA and contaminate the ctDNA

Patient history
- different tumours shed different amounts
- Smoking, diabetes, exercise can impact shedding

187
Q

How can variants be detected from ctDNA?

A

Requires highly sensitive test due to very low level variants

Targeted PCR (small number of variants)
- Roche cobas EGFR- 42 EGFR variants using RQ-PCR
- Qiagen EGFR

NGS (many potential targets)
- Thermofisher OPA
- Agilent
- TSO500

188
Q

How have NGS technologies for ctDNA improved?

A

Improved sensitivity/depth of coverage by he introduction of unique molecular identifiers (UMIs)

These label each fragment with a UMI during the first PCR cycle during the library preparation stage; this means that any PCR products which have derived from this fragment can be combined into a consensus sequence.

This minimises PCR artefacts and errors

189
Q

What is ctDNA currently used for?

A

Not used widely and unlikely to completely replace tumour testing

TD
- NSCLC: EGFR (T790M) and ALK resistance variants
- companion diagnostic for osimertinib if T790M detected
- Future- ESR1 in breast for resistance hormone therapy

FDA
- colorectal for KRAS, BRAF and NRAS

190
Q

What are the advantages of ctDNA?

A

Minimally invasive
- for patients were tumour is inaccessible
- allow repeat biopsy for monitoring - MRD and resistance variants

Relatively cheap and quick
- doesn’t require tissue processing or going to pathology

Accounts for tumour heterogeneity

Could in theory allow earlier diagnosis of tumours in the future

191
Q

What are the limitations of ctDNA?

A

Requires highly sensitive tests (0.5% VAF due to dlituion in cfDNA)
- need high coverage test

False negatives
- Difficult to confirm ctDNA actually present and at what burden- may be below LOD

False positives
- 10% of actionable variants are thought to actually be CHIP
- May also detect incidental germline findings

Not gold standard
- tissue is gold standard which has standardised processing
- No standardised method for ctDNA extraction or processing
- doesn’t go to pathology so doesn’t have additional tests e.g. morphology

Requires storage in specific ctDNA tubes to prevent degradation- e.g. streck tube

Can’t confirm result is from that tumour
- concurrent primaries or unknown HM

192
Q

What are circulating tumour cells?

A

Circulating tumour cells (CTC’s) can be released into the bloodstream from the primary tumour and may result in metastasis into distant organs.

Occur at very low concetrations and require enrichment to detect

193
Q

What is the clinical utility of circulating tumour cells?

A

Liquid Biopsy: CTCs can be detected in a blood sample, offering a non-invasive method for early cancer detection, potentially before tumors are visible through imaging.

Prognostic value: The number of CTCs can correlate with prognosis. Higher CTC counts often indicate more aggressive disease

Tracking Changes: Regular monitoring can provide real-time information on disease progression or response to treatment

Personalized Therapy: Analyzing the genetic and molecular characteristics

194
Q

How are CTCs enriched for?

A

Size-Based Filtration: Utilizes filters to separate CTCs from blood cells based on their larger size.

Density Gradient Centrifugation: Isolates CTCs based on their different densities compared to other blood cells.

Immunomagnetic Separation: Uses magnetic beads coated with antibodies specific to surface markers on CTCs (e.g., EpCAM) to capture and isolate them or beads specific to the depletion of other cells e.g. CD45+

195
Q

How are CTCs detected?

A

Immunocytochemistry (ICC): Stains CTCs with fluorescent antibodies against specific markers (e.g., cytokeratins) and uses microscopy to identify them.

Flow Cytometry: Labels CTCs with fluorescent antibodies and uses a flow cytometer to count and sort the cells.

RT-PCR: Amplifies and detects specific RNA markers that are characteristic of CTCs.

Next-Generation Sequencing (NGS): Analyzes the genetic material of CTCs for mutations and alterations, providing detailed molecular information.

Microfluidic Devices: Use specialized chips with microchannels and antibodies to capture and isolate CTCs efficiently from blood samples.

196
Q

What limitations are there for the uses of PCR based assays to detect CTCs?

A

The need to perform cellular lysis, which prevents cell counting

The possibility of false positive results due to illegitimate gene transcription in non-tumour cells

The amplification of cell free nucleic acids potentially present in blood

The possibility of false positive results derived from the use of unspecific markers

The presence in the blood of non-malignant epithelial cells, released for example after an invasive procedure

The possibility of false negative results due to low PCR sensitivity connected to limited expression of the tumour marker or to the presence of PCR inhibitors

SNPs affecting PCR primer binding

197
Q

What commerical platforms are available to detect CTCs?

A

CellSearch System: The only FDA-approved method for CTC detection, combining immunomagnetic separation and immunofluorescent staining to identify and count CTCs.

CTC-chip: an array of 78.000 microposts coated with anti-EpCAM antibodies. Whole blood is pumped through the chip and EpCAM positive cells are captured and detected by cameras recognising their morphology, their viability and the expression of tumour markers

198
Q

What are the challenges with using CTCs?

A

Low Abundance: CTCs are rare in the bloodstream, making detection technically challenging.

Heterogeneity: CTCs can be highly heterogeneous, complicating the detection and analysis.

Standardization: Need for standardized methods and protocols for consistent and reliable results

199
Q

How are tissue samples processed for molecular testing?

A

Fixation with 10% neutral buffered formalin to preserve tissues

Tissue processing
Consists of three steps; 1) Dehydration with increasing conc of alcohol, 2) Clearing with xylene, 3) Infiltration (lasting between 8-12 hours)

Tissue embedding into paraffin

Sectioning with a microtome

Deparrafinisation and rehyrdration
- Xylene and ethanol to disolve parafine
- Heat to melt parafin

DNA/RNA extraction

200
Q

Why are tissues fixed?

A

Preserves tissues irreversibly in a stable and reproducible state, through chemical interactions between fixative and cellular components

This prevents autolysis (digestion of cellular components by digestive enzymes) and putrefaction (digestion by bacterial enzymes)

This retains the living state as much as possible and allow visualization through staining

The most widely practiced methods for clinical sample preservation and archiving.

Allows storage of samples for years

201
Q

What factors impact tissue quality?

A

Fixation
Extraction method
Fragmentation
Deamination artefacts
Tumour heterogeneity
Presence of normal tissue

202
Q

How does formalin fixation impact genomic testing?

A

Detrimental to testing

Fragmentation

Deamination artefacts

Formaldehyde-induced crosslinks

203
Q

What does fragmentation due to FFPE lead to?

A
  • lead to low amounts of amplifiable template for PCR amplification and increases sequence artefacts
  • Associated with poor coverage
  • low pH formalin over time increases the rate of apurinic/apyrimidinic site formation and eventually decomposition and fragmentation
204
Q

What is deamination from FFPE and what does it cause?

A
  • Hydrolytic deamination of cytosine to form uracil (or thymine if the cytosine is methylated). This results in non-reproducible C>T/G>A transition sequencing artefacts
  • Occur to some degree in every sample
  • Can result in false positives
205
Q

What other variables affect tumour DNA quality?

A

The duration of transport from patient to laboratory.

Size of tumour specimens.

Environmental factors such as exposure to heat, light and the concentration and age of formalin used for fixation.

The age of blocks examined is another consideration as long-term storage in suboptimal environments can cause significant DNA damage.

Tumour heterogeneity, and the presence of normal tissue.

Chemistry and design strategy used for NGS (hybridisation vs amplicon).

206
Q

What are abasic sites in FFPE and how does this impact downstream analysis?

A

Formaldehyde is readily oxidized to formic acid in the reaction with atmospheric oxygen. The formation of formic acid reduces the pH of formalin

The N-glycosidic bonds of the purine bases to the sugar backbone are susceptible to hydrolysis at low pH, generating abasic sites in the DNA. Thus, fixation of tissues in unbuffered formalin will significantly lower the amount of amplifiable DNA templates.

DNA polymerases have generally low bypass efficiencies and can lead to sequencing artefacts (SNVs and deletions)

207
Q

What is the relevance of tumour content in FFPE?

A

Presence of normal tissue will dilute the tumour DNA down and mean that variants are present at a lower frequency. This may push them below the limit of detection for an assay

Tumour content requirements vary between assays- often 20-30% for detection of variants at 5% VAF

208
Q

What alternatives to FFPE are there?

A

Fresh frozen tissue (requires institutional change)

Cell free tumour DNA (liquid biopsy).

209
Q

Does FFPE DNA concentration determine the amount of amplifiable DNA?

A

No- often overestimates as lots of the DNA will be fragmented and will not be amplified

210
Q

What DNA repeair pathways are there?

A

Base Excision Repair removes damaged bases that do not significantly alter the overall structure of DNA,

Nucleotide Excision Repair: can recognise and repair the bulky, structurally altering ssDNA lesions

Mis-Match Repair: functions as a method of proofreading after DNA replication to catch any mismatched base pairs

Non-Homologous End Joining: a form of repair template-independent repair and thus is an error-prone mechanism

Homologous Recombination Repair: is considered to be an error-free method of DSB repair .

211
Q

What is the clinical significance of Homologous Recombination deficiency (HRD)?

A

Present in ovarian 50%, breast 15%, pancreatic 8%, and prostate cancer 5%.

Predictive biomarker for repsonse to platinum based chemotherapy

Predictive biomarker for PARP inhibitors: exploiting and inducing cancer-specific synthetic lethality.

212
Q

What genomic alterations are associated with HRD?

A

BRCA1/2
- Germline or somatic

BRCA-like tumours
- ARID1A, ATM, PALB2, RAD51, RAD54, ATR
- Mutations in other HR genes

213
Q

How do PARP inhibitors work?

A

PARP inhibitors block PARP enzyme which is essential for base excision repair, leadinf tot he accumulation of DNA breaks

Synthetic Lethality: In HRD-positive cells, the inability to repair single-strand breaks (due to PARP inhibition) leads to the formation of double-strand breaks, which these cells cannot efficiently repair due to their deficient HR pathway. This results in cell death, selectively targeting HRD-positive cancer cells.

214
Q

How can PARP inhibitor resistance occir?

A

Restoring BRCA 1/2 activity.

Recently BRCA1/2, RAD51C/D and PALB2 reversions or secondary mutations have been described to re-store homologous recombination.

215
Q

How is HRD detected?

A

BRCA, HRD variants

Genomic scarring (panels, WGS, myriad)
- Looks for the consequence rather than the cause
- The level of chromsomal abnormlaities correlates with HRD status
- Loss of heterozygosity (LOH)- regions of intermediate size >15MB and <whole chromosome
- Telomeric allelic imbalance (TAI)- the number of regions with allelic imbalance which extend to the sub-telomere
- Large-scale state transitions (LST)-

RAD51 foci assay
- Assesses ability of cells to repair DSBs by monitoring the recuritment of RAD51 to the breaks

216
Q

How can mutational signatures be used to identify HRD?

A

Mutational signatures are patterns of somatic passenger mutations that arise during tumorigenesis and can provide insights into the underlying causes of individual cancers.

HRD predominantly manifests as small indels and genome rearrangements due to abnormal double strand break repair but also in the form of this base substitution signature.

Single-base substitution Signature 3 (SBS3): SBS3 is strongly associated with germline and somatic BRCA1 and BRCA2 mutations and BRCA1 promoter methylation- can predict HRD

217
Q

What are the limitations of HRD assays?

A

Resistance pathways would not remove the existing “scar” of prior HR.

FFPE may overestimate HRD

Most HRD assays have not been validated in a clinical trial of PARP inhibition now myriad not widely used

Lack of standaridation of assays and HRD thresholds/ guidelines

A negative HRD score does not exclude HRD

Tumour heterogeneity

218
Q

What are driver and passenger mutations?

A

‘driver’ mutations -confer growth and survival advantage to the cell, promote oncogenesis.

Most are ‘passenger’ mutations- do not confer growth advantage but accumulate as a consequence of inherent genomic instability of cancer cells.

219
Q

How do mutational signatures in tumours arise?

A

External environmental exposures e.g. smokers with lung cancer

Internal biological processes e.g. defective DNA repair

220
Q

What variant types are used to determine mutational signature?

A
  1. Single Base Substitution (SBS) Signatures- SNVs
  2. Doublet Base Substitution (DBS) Signatures- concurrent modification of two consecutive nucleotides
  3. ID Signatures-Small insertions and deletions (ID) Signatures- are defined as the incorporation or loss of small fragments of DNA (usually between 1 and 50 base pairs) in a specific genomic location.
  4. Copy Number Variations (CN) Signatures
  5. Structural Variations (SV)
  6. RNA Single Base Substitution (RNA-SBS)
221
Q

How are mutational signatures detected?

A

Sequencing
- NGS or WGS

Classification of signature with BI
- Signature Extraction Algorithms: Tools such as MutationalPatterns, SigProfiler, and DeconstructSigs analyze sequencing data to extract and identify mutational signatures.
- COSMIC provides a reference set of mutational signatures identified in different cancer types.

222
Q

What are the limitations of using mutational signatures?

A

Tissue availability/low tumour purity: impacts the ability to detect somatic sequence alterations near the limit of detection

Need germline sample to filter our variants

Artifacts: from tissue preservation, sequencing, oxidative damage and formalin artifacts

Poor validation of mutational signatures and clinical based correlations

Lack of standardised reporting.

Some mutational signatures exhibit overlapping features e.g colorectal cancers arising from MUTYH-associated polyposis exhibit an enrichment of C>A transversions, and the resulting mutational signature shares features with the mutational signature associated with tobacco smoke.

223
Q

What are the clinical applications of mutational signatures?

A

At present, largely used in research setting, only available through WGS .

Diagnosis: in CUP e.g. in melanoma

Assessment of HRD
– PARPi indications if HRD identified in ovarian

Assessment of MMR deficiency
- Immunotherapy: use in guiding treatment options, since MMR-deficient tumours demonstrate increased sensitivity to immune checkpoint inhibitors.
- Screening tool for Lynch syndrome

Public Health: Identifying mutational signatures from prior exposures from environmental toxins (such as smoking and UV light) can help quantifying the relative cancer risk of potential genotoxins.

224
Q

What is MRD?

A

Minimal residual disease (MRD) - low level cancerous cells that remain following treatment, and are only detectable by highly sensitive techniques.

225
Q

What are the uses if MRD?

A

High-resolution determination of efficacy of therapy

To allow target-driven titration of dose and duration of treatment

Relapse risk stratification after induction to allow triage to optimal consolidation therapy

To determine prognosis after completion of standard treatment

To spare toxicity and cost of SCT in those with low risk of relapse

Assignment to maintenance therapy after completion of standard treatment

226
Q

What techniques can be used to monitor MRD?

A

FISH
- Low sensitivity

RQ-PCR
- Real time measuring of fusion transcripts

Fragment analysis for tandem duplications
- Used for FLT3-ITD, can use allelic ratio

Flow cytometry
- monitoring of leukaemic associated immunophenotype (LAIP)

QF-PCR
- highly polymorphic markers is used to detect chimeris

227
Q

How is MRD monitored in ALL?

A

Flow
- LAIP
- Can relaspe with different immunophenotype

RQ-PCR
- IG/TCR clonal rearrangments
- gene fusions such as BCR-ABL1

228
Q

How is MRD monitored in AML?

A

RQ-PCR
- Fusion genes and NPM1

Fragment analysis
- FLT3- ITD status

Flow
- LAIP

WT1
- WT1 is overexpressed at the mRNA level in 80–90% of AML cases
- ELN found that reduction is an independant prognostic factor

229
Q

How is MRD monitored in CML?

A

RQ-PCR
- BCR-ABL1

FISH

230
Q

How is MRD monitored in lymphoma?

A

RC-PCR
- Ig/TCR clonal rearrangmenets
- Gene fusions e.g. IGH-BCL2

231
Q

How are bone marrow transplants monitored?

A

Sex mismatched BMT: XY FISH, Amelogenin-XY marker on PCR.

Sex matched BMT: PCR using short tandem repeat microsatellite markers.