Solid Tumours Flashcards

1
Q

What are the genes and variant types which are targetable in Lung Cancer?

A
EGFR mutations
ALK rearrangements
RET rearrangements
ROS1 rearrangements
MET amplification
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Response to what treatment is associated with EGFR mutations in lung cancer?

A

EGFR Tyrosine Kinase inhibitors

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

What types of EGFR variants are activating and associated with response to EGFR TKI’s in lung cancer and what are the most common variants?

A
  • Point mutations (Missense), small insertions and small deletions

Most common (90%)

  • In frame deletions in exon 19
  • Point mutation (L858R) in exon 21

Other known sensitising variants

  • Exon 19 insertions
  • L861Q
  • G719X
  • S768I
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is the most common mechanism for resistance in EGFR activating mutation positive lung cancer patients hen treated with EGFR TKI’s?

A

The most common resistance mechanism is the T790M mutation in EGFR.

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

What type of EGFR variant is associated with a poor prognosis and lack of response to EGFR TKI’s

A

Exon 20 insertions

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

What is the clinical significance of BRAF variants (V600E and non-V600E) in lung cancer?

A

BRAF driver variants occur in 1-2% of lung cancer cases.

BRAF V600E typically occurs in current/former smokers and do not typically occur alongside EGFR, MET ex14, RET, ALK or ROS variants.

For patients with the BRAF V600E variant, combination therapy with dabrafenib and trametinib is recommended (BRAF inhibitor and MEK inhibitor)

Non-BRAF V600E variants impact on therapy is yet to be established.

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

What phenotype are KRAS variants associated with in lung cancer and what are there clinical significance?

A

KRAS variants are associated with cigarette smoking and are associated with a shorter survival than patients who are KRAS wildtype. Patients with KRAS driver variants do not respond to EGFR TKIs.

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

What types of variants can be found in MET in lung cancer and what is the function of MET

A

MET (C-MET), the hepatocyte growth factor (HGF) receptor is a tyrosine kinase receptor that is involved in cell survival and proliferation.

Driver variants in MET include exon14 skipping mutations, MET gene copy number gain/amplification and MET overexpression.

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

What is the clinical significance of MET variants in lung cancer?

A

Capmatinib (MET inhibitor) has been approved for treatment by the FDA but is still under review by NICE.

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

What is the function of RET/ALK/ROS1, what types of RET/ALK/ROS1 variants are identified in lung cancer and what is the clinical significance?

A

RET, ALK and ROS1 aretyrosine kinase receptors involved in cell proliferation.

RET, ALK and ROS1 rearrangements can be identified in lung cancer and may occur with a number of genes (e.g. KIF5B, CCDC6) which leads to overexpression of the RET, ALK or ROS1 protein.

Patients with rearrangements in these genes have been shown to respond to RET, ALK or ROS1 tyrosine kinase inhibitors.

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

Under what circumstances may a Lung patient be treated with immunotherapy.

A

Positive PD-L1 staining (>50%)

No approved targetable genetic mutations (EGFR / ALK/ ROS1 etc)

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

What are the tests which can be used to detect mismatch repair deficiency in colorectal cancer?

A

IHC for MLH1, MSH2, MSH6 and PMS2
or
Microsatellite Instability (MSI) analysis

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

What proportion of colorectal cancer patients have MMR defects and what proportion can be attributed to a germline cause?

A

15-20% are identified as having an MMR defect

3% have a germline cause

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

Describe the Lynch screening pathway with IHC as the starting test

A

IHC for MLH1, MSH2, MSH6 and PMS2
->
If MHS2, MSH6 or PMS2 IHC is abnormal proceed to Lynch germline analysis
->
If MLH1 is abnormal continue to BRAF V600E analysis
->
If BRAF V600E detected -> Likely Sporadic
->
If no BRAF V600E is detected proceed to MLH1 promoter hypermethylation analysis
->
If MLH1 promoter is methylated -> Likely Sporadic
->
If MLH1 promoter unmethylated -> Proceed to Lynch germline analysis

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

Describe the Lynch screening pathway with MSI as a starting test

A

MSI analysis
->
If MSI is normal/low -> Likely somatic
->
If MSI is high (unstable) proceed to BRAF V600E analysis
->
If BRAF V600E detected -> Likely Sporadic
->
If no BRAF V600E is detected proceed to MLH1 promoter hypermethylation analysis
->
If MLH1 promoter is methylated -> Likely Sporadic
->
If MLH1 promoter unmethylated -> Proceed to Lynch germline analysis

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

IHC uses antibodies to detect the expression of MMR proteins in Colorectal cancers. What does decreased or abnormal staining indicate?

A

Absent or reduced signaling of the proteins can indicate there may be a mutation (somatic or germline) in one of those genes.

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

What are microsatellites and why is the analysis of these regions used to test for MMR deficiency?

A

Microsatellites are repetitive sequences of DNA that are at increased risk of copying errors during replication.

In tumours with a defective MMR system, errors in copying microsatellite sequences cause them to vary in length - this is known as microsatellite instability.

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

What are the tests used following IHC or MMR to determine wether a colorectal is likely to be sporadic and why are they used?

A

MLH1 promoter hypermethylation - Sporadic CRC can show loss of MLH1 protein expression cause by mutations in the promoter.

BRAF V600E testing - BRAF V600E variants are associated with sporadic mutations in MLH1

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

What is the clinical significance for testing for KRAS and NRAS in Colorectal cancer?

A

KRAS or NRAS mutations are associated with a lack of response to EGFR inhibitors. This is because the proteins operate downstream from EGFR in the RAS/RAF/MEK/ERK pathway.

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

What are the hotspots for KRAS and NRAS?

A

Exon 2 - codon 12 & 13
Exon 3 - codon 59 & 61
Exon 4 - codon 117 & 146

In both genes

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

What is the clinical significance of BRAF V600E (therapy related) and non codon 600 BRAF variants in colorectal cancer?

A

BRAF V600E -associated with a worse prognosis and lack of response to EGFR inhibitors unless given with a BRAF inhibitor

Non-V600E - Currently uncertain significance

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

What treatment are colorectal patients who are MSI high eligible for?

A

MSI-H tumours or those with MMR deficiency have been shown to respond to the PD-1 inhibitor Pembroluzima (Immunotherapy)

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

What are the different classes of BRAF mutations?

A

Class I: Found at the V600 locus, defined as those with extremely strong kinase activity

Class II: Non-Codon 600 variants with intermediate to high activity

Class III: Lack of impaired kinase activity

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

What is the clinical significance of the occurrence of BRAF V600 and non V600 BRAF mutations in melanoma?

A
V600 - Approved use of BRAF inhibitors
Non-V600 - Not approved but class II may respond to BRAF inhibitors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is the clinical significance of KIT driver variants in melanoma?

A

KIT driver variants are more frequently identified in acural and mucosal melanoma. Patients with KIT variants have been shown to respond to the TKI imatinib but it is not currently approved.

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

What is the clinical significance of NRAS driver variants in melanoma?

A

NRAS driver variants are associated with poor survival in local and advanced melanoma. MEK inhibitors produce a response in a minority of patients.

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

Driver variants in what genes are associated with uveal melanoma and in what proportion of cases are they identified?

A

GNAQ and GNA11 - >80% of cases

28
Q

What is the response rate to TKIs of the commonly identified KIT driver variants in GIST and in what proportion of cases are they identified?

A

80% of GISTS will have a KIT driver variant

90% of KIT exon 11 mutant patients will respond to treatment with imatinib (TKI)

50% of KIT exon 9 mutant patients will respond to treatment with imatinib

29
Q

What proportion of GIST cases have a PDGFRA driver variant and what is the clinical significance?

A

5-10% of GISTS will have a PDGFRA driver variant

Most PDGFRA variants are associated with response to imatinib, apart from the D842V variant which is unlikely to respond to imatinib and other approved TKIs.

30
Q

What mechanism causes resistance to imatinib in GIST and what treatment is recommended when this occurs?

A

Drug resistance typically occurs due to secondary variants in KIT or PDGFRA.

Patients who are resistant to imatinib should proceed to treatment with Sunitnib and for patients resistant to imatinib and sunitinib patients should be treated with Regarafenib.

31
Q

In what complex are mutations often identified in GIST patients without KIT or PDGFRA mutations and what is the clinical significance?

A

10-15% of GIST patients which lack mutations in KIT/PDGFRA have functional inactivation of the succinate dehydrogenase (SDH) complex which can be detected by a lack of SDHB expression on IHC.

GISTS with SDH inactivation usually arise in the stomach of younger individuals. Patients may benefit from treatment with sunitinib or regorafinib.

Referral to clin gen is recommended for patients with SDH deficient GIST or those with NF1 or SDH mutations

32
Q

What are the prognostic implications/grading associated with an IDH1/2 variant in a glioma and what testing methods can be used?

A

IDH1/2 variants define grade 2/3 astrocytomas and oligodendrogliomas and the secondary grade 4 glioblastomas into which astrocytomas can evolve. Their presence distinguishes lower grade gliomas from primary glioblastomas which are IDH wildtype.

IDH1/2 variants are associated with a relatively favourable prognosis and are commonly associated with MGMT methylation. In grade 2/3 gliomas, wild type IDH1/2 is associated with increased risk of progressive disease.

Diffusely infiltrative astrocytomas with IDH1/2 are mostly grade 2/3, however some develop grade 4 histological features associated with worse prognosis.

The most common IDH1 variant R132 can be detected by IHC with sequencing of IDH1/2 recommended if negative

33
Q

What is 1p/19q co-deletion diagnostic of, how is it tested for and what is the associated prognosis.

A

The occurance of a 1p/19q co deletion is diagnostic of oligodendroglioma and is always seen alongside an IDH1/2 variant which is also needed for a diagnosis of oligodendroglioma.

The co-deletion is associated with a favourable prognosis.

Detection can be carried out by FISH, PCR, Array based (e.g. SNP array) or NGS

34
Q

For which types of brain tumour is MGMT promoter methylation carried out, what is the role of the MGMT protein, how is it tested for and what genetic changes is it associated with?

A

MGMT promoter methylation is essential in the workup of all high grade gliomas - grade 3/4.

MGMT is a DNA repair enzyme which reverses the DNA damage caused by alkylating agents.MGMT unmethylated is active and results in resistance to temozolomide chemotherapy. MGMT methylation silences the protein and makes it more sensitive to treatment with alkylating agents.

MGMT promoter methylation can be tested by methylation specific PCR, methylation specific high res melt analysis, pyrosequencing and ddPCR. Pyrosequencing is considered the best method and prognostic stratifier in GBMs treated with temozolomide.

MGMT methylation is strongly associated with IDH mutations and genome-wide methylation epigenetic changes (G-CIMP phenotype)

35
Q

What type of brain tumour is ATRX mutations associated with, what genetic changes are they seen with and not seen with and how can it be tested for?

A

ATRX mutations are associated with astrocytomas.

They are strongly associated with IDH mutations and are mutually exclusive with 1p/19q codeletions.

ATRX mutations can be detected by IHC and/or sequencing.

36
Q

What is the clinical significance of the K27M in H3F3A/HIST1H3B and G34V variants in H3F3A in brain tumours?

A

Histone variants tend to occur in paediatric midline gliomas. The K27M variant is an adverse prognostic marker and the G34V variant does not have a prognostic association after a diagnosis of GBM is made.

37
Q

What is the clinical significance of the BRAF V600E and BRAF Fusion variants in brain tumours?

A

BRAF V600E - present in 60-80% of supratentorial grade 2/3 pelomorphic xanthoastrocytomas (PXA), 30% of dysembryoplastic neuroepithelial tumours, 20% of grade 1 gangliomas and 5% of grade 1 astrocytomas.

The presence of a BRAF fusion is reliable evidence that the tumour is a pliocytic astrocytoma.

BRAF fusions tend to be indolent but BRAF V600E is associated which a much greater range of outcomes and may be associated with response to BRAF inhibitors.

38
Q

What are the molecular hallmarks of a glioblastoma?

A

EGFR amplification/mutation
IDH wt
TERT mut
chr10/PTEN del

39
Q

Fusions involving which gene is characteristic of a diagnosis of Ewings Sarcoma?

A

EWSR1 fusions

Note this is not diagnostic as also seen in other tumour types

40
Q

What group of genes does EWSR1 frequently fused to in Ewings Sarcoma?

A

It can be found most frequently fused to members of the E26 transformation specific / Erythroblast transformation specific (ETS) family of transcription factors including FLI1, ERG, ETV1, ETV4 and FEV.

The ETS family comprises of 29 genes all of which contain a highly conserved DNA binding domain called the ETS domain

There are a number of non-ETS gene fusions also seen in other types of soft tissue sarcomas.

41
Q

What are fusions in EWSR1 in Ewings Sarcoma often caused by and what is the prognostic significance?

A

Fusions are generated by chromoplexy in 42% of cases which disrupts multiple genes and is associated with a more aggressive disease.

42
Q

What secondary events are often seen in Ewings Sarcoma and what is the associated prognosis?

A

Gain of chr8 (30-35%) - No significance
Gain of chr12 (25-33%) - Adverse prognosis
Gains of chr2 - Positive prognosis
Unbalanced translocation t(1;16)(q12;q11.2) (10-30%) - Poor prognosis
Deletions in short arm of chr9 (9p21, CDKN2A) - Poor prognosis
Inactivating mutations in STAG2 and TP53
CDKN2A Deletions

43
Q

What testing is carried out for Ewings Sarcoma?

A
  • Cytogenetics - Detect translocations etc

- Break apart of EWRS1 (FISH, RT-PCR, NGS), where possible techniques which can identify the partner are preferable

44
Q

What could cause an EWSR1 fusion to not be detected in an Ewings Sarcoma?

A

A fusion involving the FUS gene will give a false negative result when using breakapart probe FISH as EWSR1 is not translocated in these cases

EWSR1-FUS fusion neg undifferentiated round cell sarcoma are characterised by CIC-DUX4 fusions (t(4;19), t(10,19))

45
Q

What is homologous recombination deficiency (HRD)?

A

The homologous recombination pathway allows the repair of double stranded DNA breaks. HRD is a deficiency in the HR system for repairing double stranded breaks.

46
Q

Genomic instability is a hallmark for disruption of which pathway?

A

Homologous recombination

47
Q

What are the main (known) causes of HRD?

A

Germline or somatic mutations in BRCA1/2
Mutations on the Fanconi Anaemia pathway genes
Mutations in the mismatch repair genes

48
Q

What is the currently only known mechanism for PARPi resistance (primary or acquired)

A

Reverse mutations in the BRCA genes that can reinstate the homologous recombination pathway.

49
Q

How do PARP inhibitors work?

A

PARP is an enzyme required to repair single-stranded DNA breaks. When PARP is inhibited, it leads to an accumulation of single-strand breaks. When PARPi are used in a patient deficient in double-stranded DNA breaks, this leads to tumour death because neither single strand or double strand breaks can be repaired. This is a phenomenon called synthetic lethality.

50
Q

What are the main categories of HRD test?

A

1) HRR pathway related genes - specific cause of HRD
2) Genomic ‘scars’ or mutational signatres which measure the pattern of somatic mutations that accumulate rather than the underlying defect.
3) Functional assays that can provide a real time read out of HRD.

51
Q

What gene level tests can be used to detect HRR deficiency?

A

1) Germline testing to detect inherited pathogenic variants including the large dels/dups senn in BRCA1/2
2) Tumour testing through extraction of DNA from the ovarian tumour and tested for driver/pathogenic variants. - About 2/3 of BRCA variants have been shown to be germline in origin

52
Q

What types of variants are associated with HRD?

A

Driver variants in HR pathway genes
Thousands of somatic mutations - SNVs and short deletions and tandem duplications
BRCA1 / RAD51 promoter methylation
Deletions of large stretches of DNA (<15Mb)
Telomeric Allelic Imbalance
Large scale transitions

53
Q

What is the commercial assay for HRD currently available through funding from AstraZeneca and how does it work?

A

MyChoice HRD - Myriad
Diagnostic test using FFPE - FDA approved
Assessed SNVs, insertions, deletions and large rearrangements to determine a genomic instability score (GIS). A positive test has a GIS >42

54
Q

What is the genomic signature which is a proposed biomarker for HRD?

A

SBS Signature 3 (COSMIC)
Associated with BRCA1/2 mutation and BRCA1 promoter hypermethylation

Unlikely to be a robust clinical biomarker in isolation

55
Q

How does the HRDetect algorithm work?

A

Algorithm uses a combination of all four mutation classes and measures the features with different weighting

  • Indels (2.398)
  • SBS Sig 3 (1/611) and Sig 8 (0.091)
  • SVS Sig 3 and Sig 5 (0.847)
  • CNV HRD Score (Same as myraid) (0.667)
56
Q

What are the advantages and disadvantages of using a mutational signature for the detection of HRD?

A

Advantages
-Strong clinical evidence i can outperform scar assays (myriad)

Disadvantages

  • Needs to be done on fresh tissue as there are too many artefacts from FFPE
  • Provides a historical representation of HRD and doesn’t consider HRD status could be reinstated
57
Q

What does a typical NTRK gene rearrangement look like?

A

Typically NTRK rearrangements are caused by intra or inter chromosomal rearrangements to from hybrid genes in which the 3’ sequence of NTRK1/2/3 that includes the kinase domain are juxtaposed to the 5’ sequence of a different gene. This produced a chimeric oncoprotein characterised by ligand dependant constitutive activation of the TRK kinase.

Upstream partners often contain oligodimerisation domains such as coiled-coil, zinc finger or WD repeats.

58
Q

In which cancer types are NTRK1/2/3 rearrangements most common?

A

Rare cancer types are highly enriched for NTRK1/2/3 fusions with a prevalence of up to 90% in some cancer types.

NTRK fusions are found in more common cancers at a much lower frequency (5-25%)

59
Q

What testing methods can be used to detected an NTRK1/2/3 fusion?

A
  • FISH
  • RT-PCR
  • However, most commonly used is NGS. RNA-Seq is typically used as large intronic regions in NTRK2 or NTRK3 can lead to difficulties in detection using DNA
60
Q

What are the approved TRK inhibitors (NICE) for use for patients with NTRK1/2/3 rearrangements?

A

Larotrectinib and Entrectinib.

61
Q

What causes the secondary resistance to TRK inhibition?

A

The only mechanism identified to date is the acquisition of mutations in the kinase domain of the fusion.

62
Q

What type of treatment does DPD efficiency cause toxicity to?

A

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

63
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.

64
Q

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

A

c. 1905+1G>A (DPYD2A) - null function
c. 1979T>A (DPYD
13) - null function
c. 2846A>T - Moderate function
c. 1129-5923C>G (HapB3) - Moderate function

65
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

66
Q

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

A

7%

67
Q

What are the three categories of DPD metabolisers and associated activity and dosing of these?

A

DPYD Poor metabolisers (DPYD AS:0.5 or 0)

  • Carriers of two no function DPYD variants (AS 0)
  • Carriers of one no function and one decreased function DPYD variant (AS 0.5)
  • AS 0.5 - Avoid use of 5FU. If alt not available should be used at a strongly reduced dose with careful monitoring
  • AS 0 - Avoid use of 5FU

DPYD Intermediate metabolisers (DPYD AS:1 or 1.5)

  • Carriers of one no function DPYD variant (AS 1)
  • Carriers of one decreased function DPYD variants (AS 1.5)
  • Carriers of two decreased function DPYD variants (AS 1)
  • AS 1 - Reduce dose by 50%
  • AS 1.5 - Reduce dose by 25-50%
  • Intermediate metabolisers may tolerate full dose so dosing should be increased in subsequent cycles when patients experience none or clinically tolerable toxicity in the first two cycles.

DPYD normal metabolisers (DPYD AS:2)

  • No clinically relevant DPYD variants
  • Normal dosing as per = manufacturers instructions