Other Flashcards

1
Q

What genes have been linked with hereditary breast and ovarian cancer?

A

There are two primary genes linked with the majority of HBOC families; BRCA1 and BRCA2. There are also other genes which have been linked to an increased risk of developing breast and other cancers including TP53, PTEN, CDH1, ATM, CHEK2 or PALB2. The genes currently tested as part of the national test directory are BRCA1, BRCA2, PALB2, ATM and CHEK2

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

What is the inheritance pattern of BRCA1 and BRCA2 germline mutations and associated familial risks?

A

HBOC follows an autosomal dominant pattern of inheritance. Therefore a mutation need only occur in one copy of the gene to have an increased risk of cancer. Parents may pass along a copy of the gene with a mutation with the child having a 50% chance of inheriting the change. A sibling or parent will also have 50% chance of inheriting the mutation, however if the parents test negative for the mutation, the siblings risk will significantly decrease but still may be higher than an average risk.

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

What is the incidence of hereditary breast and ovarian cancer?

A

Most breast and ovarian cancers are sporadic. Currently it is estimated that <1% of the general population have a mutation in BRCA1 or BRCA2 and up to 10% of women and 20% of men who develop breast or ovarian cancer will have a mutation in one of these genes. 10-30% of women younger than 60 diagnosed with triple negative breast cancer, cancer which are negative for HER2, progesterone and oestrogen, have a BRCA1 or BRCA2 mutation. Others may have mutations in other associated genes.

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

Which sub-population has an increased risk of hereditary breast and ovarian cancer and why?

A

Families with Ashkenazi Jewish ancestry have an increase chance through the occurance of three ‘founder mutations’; 185delAG in BRCA1, 5382insC in BRCA1 and 6174delT in BRCA2. It is estimated that 1 in 40 people of Ashkenazi Jewish ancestry will have one of these three mutations.

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

What is the estimated risk of breast and ovarian cancer for women with a BRCA1 or BRCA2 germline mutation?

A

Cancer risks for women with HBOC:
• Lifetime risk of breast cancer – 45-75%
• Lifetime risk of ovarian cancer – BRCA1 25-40%, BRCA2 10-20%
• Developing a second breast cancer – 20-40%

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

What is the estimated risk of breast and ovarian cancer for men with a BRCA1 or BRCA2 germline mutation?

A

• Lifetime risk of breast cancer – BRCA1 1-2%, BRCA2 6%
• Risk of prostate cancer – BRCA1 some increased risk, BRCA2 – 20%
• Men with a BRCA2 mutation have significantly increase risk of developing more aggressive prostate cancer before the age of 65 and therefore screening should begin at age 40

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

What are the clinical features of neurofibromatosis type 1?

A

Multiple light brown patches of skin pigments (café au lait spots), skinfold freckling, visible neurofibromas under the skin and small modules in the iris

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

What type of mutations in NF1 cause neurofibromatosis type 1?

A

Point mutations are responsible for 90% of NF1 patients with a single exon or whole NF1 gene deletion associated with the remaining 5-7%.

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

What type of mutations in NF1 cause neurofibromatosis type 1?

A

Point mutations are responsible for 90% of NF1 patients with a single exon or whole NF1 gene deletion associated with the remaining 5-7%.

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

What are the clinical features of neurofibromatosis type 2?

A

NF2 patients typically develop schwannomas on the bilateral vestibular portion of the eight cranial nerve and on other cranial nerves, spinal roots or peripheral nerves. In addition NF2 patients often develop multiple meningiomas and ependymomas at an early age. NF2 patients often experience hearing loss, balance problems, flesh coloured skin flap and muscle wasting. Visual impairment is likely due to cataracts, optic nerve meningiomas and retinal hamartomas.

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

What is the most frequent type of mutation in NF2 which causes neurofibromatosis type 2?

A

Truncating mutations are the most frequent germline event and cause the most severe disease. The presence of truncated protein is also associated with a younger age at diagnosis and a higher prevalence of meningiomas, spinal tumours and cranial nerve tumours.

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

Selumetinib is what type of inhibitor and what is it used to treat?

A

Selumetinib is a MEK inhibitor which is approved by NICE for NF1 for treating symptomatic and inoperable plexiform neurofibromas (PN) associated with type 1 neurofibromatosis (NF1) in children aged 3 and over.

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

What drug can cause toxicity in patients with TPMT mutations?

A

Mercaptopurine, Azathioprine and Thioguanine.
mercaptopurine and azathioprine are generally used for non-malignant immunogenic disorders and thioguanine for myeloid leukaemias.

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

What is the frequency of TPMT mutations causing TMPT deficiency?

A

• 1%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are the four TPMT deficiency alleles which account for 95% of TMPT deficiency cases?

A

o TPMT 2* (238G>C) – 100x decrease in activity (very low)
o TPMT *3A (contains two SNPs, *3B and *3C) – No detectable enzyme activity
o TPMT *3B (460G>A) – 4x decrease in activity (very low)
o TPMT *3C (719A>G)) – 1.4x decrease in activity

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

What starting does alterations for azathioprine are carried out according to TPMT testing?

A

o Homozygous wild-type – Normal dose
o Heterozygous – one functional and one non-functional – reduce dose by 30-70%
o Homozygous / Compound het – Two non functional alleles – Consider alternative treatments for non-malignancy. For malignancy start at a drastically reduced dose.

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

What type of treatment does DPD efficiency cause toxicity to?

A

Treatment with fluoropyrimidines - 5-fluorouracil (5FU) and capecitabine

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

What does the dihydropyrimadine dehydrogenase (DPD) enzyme do and what happens when DPD is deficient?

A

DPD is the first step and rate limiting enzyme for the catabolism of fluoropyrimidines. Reduced DPD activity results in reduced clearance and increased half life of fluoropyrimidines (5FU and capecitabine) and can cause profound dose related toxicities.

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

What are the four clinically relevant DPYD variants and their associated impact on DPD function?

A

o c.1905+1G>A, activity 0
o c.1679T>G, activity 0
o c.2846A>T, activity 0.5
o c.1129-5923C>G (c.1236G>A), activity 0.5

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

What variant is in perfect linkage disequilibrium with the DPYD c.1129-5923C>G variant?

A

The synonymous variant c.1236G>A
This can be used as a proxy when testing

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

What percentage of the European population is estimated to have a clinically relevance DPYD variant?

A

7%

22
Q

What are the proven biomarkers for response to immunotherapy?

A

High Tumour Mutational Burden
MSI High / dMMR
POLE or POLD1 mutation
PD-L1 expression on IHC

23
Q

Why is PD-L1 expression a predictor of immunotherapy response?

A

PD-L1 is a protein which helps to keep immune cells from attacking non harmful cells in the body. Some cancers have high amounts of PD-L1 which allows the cancer to ‘trick’ the immune system and avoid being attacked as a foreign, harmful substance.

24
Q

What is tumour mutational burden (TMB)?

A

TMB refers to the number of somatic gene mutations present in a tumour. It is defined as the number of mutations per megabase of DNA analysed. This varies across tumour types

25
Q

What tumour features can impact TMB calculations?

A

-Variations across different cancer types in levels of TMB
-Tumour heterogeneity
-Primary/Metastatic - Primary tumours have lower heterogeneity compared to metastatic (higher TMB)

26
Q

How can TMB influence the effect of the immune system in recognising tumour cells?

A

Cancers with low TMB have fewer mutations and therefore decreases the chance that one will activate the immune system

Cancer cells with high TMB have more mutations, increasing the chance that one will activate the immune system

27
Q

How do immune checkpoint inhibitors work?

A

One mechanism that tumours use to survive is to increase immune checkpoint molecules that can help to suppress antitumor immune responses. ICIs reinvigorate antitumor immune responses by interrupting co-inhibitory signaling pathways and promote immune-mediated elimination of tumor cells.

28
Q

Why is TMB a marker for ICI response?

A

Tumours with high TMB had a higher number of neoantigens and therefore can identify patients who would in turn benefit from ICI therapy.

29
Q

What are the high/low thresholds or TMB and what response is seen in patients treated with ICIs?

A

TMB-High - >20mutations/Mb, response 58%
TMB-Low - <20mutations/Mb, response 20%

30
Q

What methods can be used to detect TMB?

A

Calculation of TMB is generally carried out using WGS, WES or panel-based approachess. Research has favoured WGS/WES whereas clinical applications have more often utilised panels due to a lower sequencing cost, lower DNA inpur requirement and a shorter turnaround time

31
Q

What are the primary factors which influence how TMB is calculated?

A

1) Tumour cell content & sequencing coverage
Quality of TMB data is dependant on greater tumour content and sequencing coverage. Targeted panels allow for sequencing to a greater depth than WGS/WES enabling a higher sensitivity when the tumour content is low (<10%) and can detect variants down to lower frequencies.

2) Pre analytical/processing factors
Tissue is normally fixed in formaldehyde (FFPE) which can lead to significant noise in NGS and impact TMB calculations

3) Sequencing strategy and size of assay
As the size of a panel becomes smaller. the uncertainty associated with TMB becomes greater with the coefficient of variation rapidly increasing when the panel size is less than 1Mb

4) Bioinformatics pipelines
Pipeline filtering can differ between analysis methodologies. Most will filter out synonymous and germline variants as they are unlikely to be involved in creating neoantigens however this is not standardised. Lack of matched tumour-germline analysis in standard clinical practice means germline variants cannot be filtered out. Choice of variant caller (and its sensitivity) can impact how TMB calculated

5)TMB thresholds
Not yet standardised. Different studies have utilised different cut offs for high TMB

32
Q

What are the two indicators of a homologous recombination deficient tumour?

A

1) Mutation in a HRD gene (e.g. BRCA1/2, RAD51C)
2) Identification of an HRD signature (e.g. HRD Scar, Genomic Instability Score, COSMIC Sig 3)

33
Q

What types of variants are the feature of an HRD scar?

A

Loss of heterozygosity (LOH)
Telomere allelic imbalance (TAI)
Large Scale State Transitions (LST)

34
Q

What is telomeric allelic imbalance?

A

Telomeric allelic imbalance, is defined as the number of regions with allelic imbalance extending to the sub telomere but not crossing the centromere

35
Q

What is a large scale state transition?

A

Chromosomal breakage that generates 10 Mb or larger fragments. The quantification of these breaks can be used as a surrogate measure for genomic instability

36
Q

What is the main limitation of genomic scar assays?

A

A major limitation of the genomic scar assays is the potential lack of representing of the current HRD status when analysing archival tissue. Tumour cells previously shown to be HR deficient may have restored their proficiency by resistance mechanisms such as reverse mutations in HRD genes.

37
Q

What techniques can be used to detect HRD?

A
  • Single nucleotide polymorphism (SNP) arrays
  • Whole Genome Sequencing
  • Targeted Panel Sequencing
38
Q

What are the names of some bioinformatics tools which can be used to generate an HRD score form WGS?

A

HRDetect and scarHRD

39
Q

What test method is approved in the UK for detecting HRD and what does it test?

A

Myriad myChoice assay
- Tests BRCA1/2 for likely pathogenic/pathogenic variants
- Generated a genomic instability (GIS) score from loss of heterozygosity, telomere allelic imbalance and large scale state transitions. a GIS >= 42 or BRCA1/2 +ve is considered positive for HRD

40
Q

What is the main difficulty in implementing HRD scoring in clinical practice?

A

Scores which are used between different bioinformatics tools or assays are not comparable which makes validation and threshold determination difficult.

41
Q

How is deparaffinisation of FFPE samples carried out?

A

The solvent xylene is typically used to remove all paraffin from the tissue sections attached to the slides/curls. Prior to deparaffinisation, slides are heated to melt the wax, they are then washed multiple times with xylene to solubilise and remove the paraffin. Next xylene is removed by washed with xylene and ethanol. Finally the sample is rehydrated with graded concentrations of ethanol in water ending in a final rise of pure water. Xylene is hazardous and non-hazardous alternatives are available to de-wax the sample.

42
Q

What are the four types of damage caused by formalin fixation?

A

Formaldehyde induced crosslinks
DNA fragmentation
Abasic Sites
Deamination of cytosine bases

43
Q

What is the basic procedure for DNA extraction and the enzymes involved?

A

Basic Procedure:
1) Cell lysis to break the cell membranes and expose DNA along with the cytoplasm
2) Lipids from cell membranes broken down with detergents and surfactants
3) Proteins broken down with protease (optional) using proteinase K
4) RNA broken down with RNAse (optional)
5) Solution treated with a concentrated salt solution (saline) to make debris such a broken proteins, lipids and RNA clump together.
6) Centrifugation of the solution to separate clumped debris from the DNA
7) DNA purification from detergents and debris. Most commonly used is
a. Ethanol precipitation usually by ice cold ethanol or isopropanol. Since DNA is insoluble in these alcohols It will aggregate together and form a pellet on centrifugation.
b. Phenol-choloform extraction in which phenol denatures proteins in the same.
8) DNA is precipitated in a slightly alkaline buffer (usually a TE buffer)

44
Q

What are the two methods for DNA quantification and name associated commercial example?

A

Spectrophotometry - NanoDrop Spectrophotometer (Thermo Fisher)
Fluorometry - Quantus (Progema)

45
Q

How is spectrometry used to evaluate DNA concentration and quality?

A

DNA concentration can be determined by measuring the absorbance at 260nm(A260) in a spectrophotometer using a quartz cuvette. The ratio of readings at 260nm and 280nm provides an estimate of the DNA purity with respect to contaminants that absorb light.

46
Q

How is fluorometry used to evaluate DNA concentration?

A

Fluorometry allows specific and sensitive measurement (more than spectrophotometry) of DNA concentration by use of a florescent dye, a common dye is Pico Green. The fluorescently labelled DNA can then be run on an agarose gel alongside a DNA ‘ladder’ consisting of known DNA quantities (e.g. Agarose gel electrophoresis or capillary gel electrophoresis). The amount of DNA can then be estimated by comparison of the band intensity with the standards either visually or using a scanner or imaging system.

47
Q

Pathogenic mutations in which genes are causative of lynch syndrome and what is the function of this group of genes?

A

MLH1, MSH2, MSH6 and PMS2

DNA mismatch repair genes (MMR)

48
Q

What are the main barriers to introducing a pharmacogenomics service?

A
  • Clinically urgent
  • High volume of samples
  • Clinical Relevance
49
Q

What is the name for the DPD enzyme and what is its function?

A

Dehydropyrimidine hydrogenase (DPD) is an enzyme which catabolises thymine and uracil. DPD catalyses the first step In the 5-FU degradation pathway, converting around 80% of 5-FU to its inactive metabolite.

50
Q

How does 5-FU (& capecitobine) work?

A
  • 5-FU works as an antimetabolite, blocking the enzyme thymidylate synthase and therefore preventing the thymidine formation required for DNA synthesis and thereby preventing cell proliferation
51
Q

What are the clinical considerations for germline focussed tumour analysis?

A

On tumour / Off tumour
Actionability (penetrance/risk, severity, preventative measures, strength of evidence)
Consent & Patient Education - Explicit consent is not routine, requested at dif stages of the pathway, counselling & confirmation)

52
Q

What are the lab considerations for germline focussed tumour analysis?

A

Tumour heterogenity / contamination (VAFS)
Analysis (ACMG))