Solid Tumours Flashcards

1
Q

What is the most common cancer wordlwide?

A

Female breast cancer

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

What is the incidence of breast cancer?

A

15% of all new cancer cases
Over 2.3 million new cases a year and 685,000 deaths.

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

What is the prognosis of breast cancer?

A

80.4% survive for 10 years or more

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

What is the average age of diagnosis of breast cancer?

A

63 years

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

What is the lifetime probability of developing invasive breast cancer for women?

A

12.3% or 1 in 7

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

What are the risk factors for breast cancer?

A

Age: >50
Personal history of breast or ovarian cancer
Family history of breast or ovarian cancer (HBOC)
Menstruation: Periods before age 12 and after 55
Pregnancy: Not being pregnant before age of 35
Hormone replacement therapy after menopause
Oral contraceptive
Radiation exposure at a young age
Lifestyle e.g. obesity, alcohol, food, exercise

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

What is atypical hyperplasia of the breast?

A

This diagnosis increases the risk of developing breast cancer in the future.

Typical ductal hyperplasia (ADH) differential diagnosis with ductal carcinoma in situ (DCIS)- same but smaller

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

What is Lobular carcinoma in situ (LCIS)?

A

LCIS refers to abnormal cells found in the lobules or glands of the breast. It is not considered cancer. However, LCIS in 1 breast increases the risk of developing invasive breast cancer in either breast in the future.

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

How does breast cancer present?

A

Most of the patients discover their disease in the context of routine screening.
The presence of breast lump, changes of breast shape or size, or nipple discharge should alert a woman. Mastalgia may also occur.

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

What are the two main types of breast cancer?

A

Breast cancer is characterized as noninvasive and invasive cancer mainly based on the location of the breast cancer cells i.e. if they are confined to the mammary ductal-lobular system or they have spread outside of the milk ducts or lobules into the surrounding breast tissue.

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

What is the most common non invasive breast cancer?

A

The most common noninvasive breast cancer type is DCIS, ductal carcinoma in situ, considered as Stage 0. It is graded as low, intermediate ad high based on the degree of differentiation of tumour cells.

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

What are the chances of DCIS progressing to invasive cancer?

A

There is at least 30% to 50% risk of untreated DCIS progressing to invasive carcinoma in the ipsilateral breast 10 to 20 years after initial diagnosis if no treatment applied. Therefore, surgical intervention to remove DCIS often followed by radiotherapy and/or medical treatment is highly recommended.

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

What are the types of invasive breast cancer?

A

Invasive or infiltrating ductal carcinoma- most common

Invasive lobular carcinoma (10% to 15% of all invasive breast cancers).

Less common invasive types are medullary, mucinous, tubular, metaplastic, papillary, micropapillary, apocrine and inflammatory breast cancer, an aggressive type (~1% to 5% of all invasive breast cancers).

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

What molecular markers are used to classify breast cancer?

A

Oestrogen recepter (ER)
Progesterone receptor (PR)
HER2

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

What is the significance or ER testing in breast cancer?

A

80% of breast cancer patients overexpress ER which drives the cancer
These patients are likely to respsonds to hormone therapy such as aromatase inhibitors and/or tamoxifen (SERM) which blocks oestrogen binding to ER

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

How are ER/PR/HER2 expression investigated?

A

IHC
Use the alfred score to determine if positive

HER2 is reflexed to FISH to confirm amplification if staining is greater than 2

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

What is the significance of PR testing in breast cancer?

A

65% of breast cancers are both ER and PR positive. PR is a target gene of ER and its expression is dependent upon oestrogen
ER/PR positive tumours respond better to ER modulator therapy (SERM)
PR positive breast cancers have a better prognosis

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

What is HER2?

A

ERBB2 part of the EGFR pathway
proto-oncogene

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

What is the significance of HER2 testing in breast cancer?

A

Overexpression of HER2 occurs in 15-20% of cancers and is associated with aggressive subtype and poor prognosis
Anti-HER2 herceptin is available as a treatment for these patients

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

What are HER2 low breast cancers?

A

Cancers assigned with low levels of HER2 protein and/or few copies of HER2 gene are sometimes now called as HER2-low. When categorizing breast cancer by HER2 expression, “HER2-low” cancers make up most cases. There has been promising results of targeted therapy with Tdx-D(Enhertu - trastuzumab deruxtecan

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

What is triple negative breast cancer (TNBC)?

A

TNBC accounts for 10-20% and lacks expression of all three receptors (ER, PR, and Her2). Thus, it cannot be treated using anti-estrogen hormonal therapies or trastuzumab (monoclonal antibody targeting HER2 protein). Associated with poorer prognosis

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

What mutations are found in TNBC?

A
  • BRCA1 and 2/HRD: 10% germline BRCA and 35% other causes of HRD
  • PIK3/AKT pathway
    -Cell cycle checkpoints
  • Notch signalling
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What potential targeted therapies are there for TNBC?

A

PARP inhibitors
PIK3CA inhibitors
Immunotherapy (PD-L1)

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

What is on the test directory for ER positive HER2 negative early breast cancer?

A

Gene expression assays: M3.2 OnocoType Dx, M3.3 EndoPredict, M3.3 Prosignia

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

What is the oncotype DX and why is it used in early stage ER+/HER- breast cancer?

A

21 genes analysed to predict chemotherapy benefit and prevent overtreatment of patients who do not need it

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

What is the significance of PD-L1 in breast cancer?

A

Atezolizumab is a PD-LA inhibitor that blocks the interaction between PD-L1 and PD-1, thereby promoting T cell activity.

PD-L1-positive patients with metastatic TNBC showed improved progression free survival (PFS) when pembrolizumab was given in combination with chemotherapy

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

What is the response rate for anti-PD-L1 therapy in breast cancer?

A

10-30%
Can have bad side effects including autoimmune disease
IHC of PD-L1 is used to determine who is likely to respond

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

What tests are on the test directory for breast cancer?

A

M3.5 NTRK testing
M3.6 PIK3CA
M3.7 DPYD
Oncotype

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

What is the clinical significance of PIK3CA on breast cancer?

A

Most recurrently mutated gene in breast cancer
Activating variants allows may respond to PIK3CA inhibitors- according to the SOLAR trial (11 specific variants but others may also respond)

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

What is the significance of ESR1 in breast cancer?

A

More common in metastatic cancers and associated with shorter progression free survival
They have been shown to be resistant to treatment with hormone therapies (AI, tamoxifen) but may respond to treatment with CDK4/6 inhibitors- clinical trials available
Elacestrant (a selective estrogen receptor degrader) has been FDA approved for ESR1 mutated breast cancers- NICE under development

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

What is the role of ctDNA in breast cancer?

A

Currently not on the test directory but likely to be added soon for ESR1
- ESR1 is a known acquired resistance mechanism to hormone therapies
- ctDNA would allow early detection of this resistance and variant is often not present in the primary tumour
- Potential access to other therapies such as Elacestrant

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

What is the significance of HRD in breast cancer?

A

It is derived by combining 3 main characteristics: numbers of telomeric allelic imbalances, large scale transitions, and loss of heterozygosity events (LOH), all of which are indicative of deficiency of the DNA repair system.
Predicts response to PARP inhibitors- currently in clinical trials BrightNess trial

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

How can breast cancer be monitored molecularly?

A

ctDNA Liquid biopsy blood test, for detecting the PIK3CA mutation, is approved by the FDA for breast cancer that can be used in clinical practice.

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

How is TNBC treated?

A
  • Surgery/readiotherapy
  • Chemotherapy
  • Targeted: PIK3CA, NTRK, PARP, PD-L1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What are the ESMO guidelines recommendations for breast cancer treatment?

A
  • Luminal A-like: endocrine therapy (ET) alone or with chemotherapy, in case of G3, T3 or ≥ 4 positive lymph nodes (IA)
  • Luminal B-like (HER2-negative): ET and chemotherapy, but not concomitantly (IID)
  • Luminal B-like (HER2-positive): chemotherapy and anti-HER2 (trastuzumab) and ET (IA). Only for selected cases, in which chemotherapy is contraindicated or refused by the patients, ET and trastuzumab may be considered as acceptable (VA)
  • HER2-positive (non-luminal): chemotherapy and anti-HER2 (trastuzumab) (IA)

-Triple-negative (ductal): chemotherapy (IA)

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

What is the ESMO guidelines on breast cancer screening?

A

ESMO recommends periodically mammography screening for women between 50 and 69 years old (IA). More specific are the NICE and German guidelines which recommend mammography for women between 50 and 70 years old every 3 and 2 years.

ESMO also recommends yearly screening with MRI and mammography together or alternating every 6 months for women with a family history of breast cancer, starting 10 years prior to the diagnosis age of the earliest case in the family (IIIB).

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

What is the incidence of high grade serous ovarian cancer (HGSOC)?

A

Over 300,000 cases diagnosed worldwide per year, distribution varies internationally. Average 1.1% lifetime risk
6th most common cancer in women

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

What are the risk factors for ovarian cancer?

A

Family history
- First degree relative with breast/ovarian cancer, prostate cancer
- Ashkenazi Jewish heritage
Reproductive history
- Early menarche, late menopause, fewer pregnancies
- Pregnancy (before 25y), breastfeeding, late menarche, early menopause, contraceptive pill for 3+ years (↓ risk by 30-50%)
Obesity
Increasing age
Genetic predisposition
- Previous cancer such as breast
- Mutations in DNA damage repair (HRD, BRCA1, BRCA2)
Risk reduction
- Tubal ligation, prophylactic hysterectomy/salpingo/oophrectomy

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

What is the clinical presentation of ovarian cancer?

A

Patients may be asymptomatic or have non-specific symptoms that are related to involvement of abdominopelvic organs.

Currently no accurate medical screening available for early-stage detection

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

What is the prognosis of ovarian cancer?

A
  • Only 20% diagnosed in early stages, when 5y survival is 94%
  • 80% patients present with FIGO stage III or IV, leading to low median survival (5y <30%)
  • Most reocur
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What is HRD?

A

The inability to repair double stranded breaks by homologous recombination
This results in error prone DNA repair and genomic instability

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

How common is HRD in High grade serous ovarian cancer (HGSOC)?

A

Approximately 50% HGSOC harbour defects in HRR pathway genes
- most are either germline or somatic BRCA variants
- variants in other HRR genes e,g, RAD51D/D, BRIP1, RAD50, BARD1, CHEK2, MRE11A, NBN, PALB2- leads to a BRCA like phenotype

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

Outline the HRR pathway?

A

Requires recruitment of BRCA1 to double-stranded break (DSB), which then stimulates exonuclease-mediated 5’-3’ end resection to create single strand overhang.
RAD51-DNA filament is created by exchange of RPA for RAD51 by complex containing RAD51, PALB2 and BRCA2. Sister chromatid is used as a template for high-fidelity DNA repair.

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

What percentage of HGSOC have BRCA variants?

A

Pathogenic variants are early events in ~20% cases (account for most dHRR), consisting of germline in ~15% HGSOC patients and somatic in ~6-7% HGSOC cases

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

What are reversion mutations in BRCA?

A

Reversion mutations in the BRCA genes that can reinstate homologous recombination proficiency (HRP) (i.e. cells/tumour cells are able to effectively repair DNA damage by HRR) have also been identified. This is often associated with primary or acquired Fce to PARP inhibitors, topoisomerase inhibitors or platinum salt

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

What other genes have clinical relevance in ovarian cancer?

A

TP53
- >95% HGSOC present TP53 variants early in oncogenesis and contribute to genomic instability

SMARCA4
- present in small cell carcinoma of the ovary

DICER1
- Seen somatically in ~50% of SLCT and at younger patient age and with moderately/poorly differentiated tumours

FOXL2
- c.402C>G, p.(Cys134Trp)-mutant
- Nearly all adult granulosa cell tumours

CTNNB1 and APC
- Variants in most microcystic stromal tumour (MCST)

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

What testing is on the test directory for ovarian cancer?

A

M2.1: BRCA1/BRCA2, SMARCA4
M2.3: NTRK
M2.5: HRD
M233.1: WGS
M245.1: ovarian sex cord stromal tumours- FOXL2, CTNNB1, APC, DICER1
R207: germline SNV and CNV- BRCA1; BRCA2; BRIP1; MLH1; MSH2; MSH6; PALB2; RAD51C; RAD51D

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

How is ovarian cancer diagnosed?

A

Physical exam
Imagining: Transvaginal ultrasound, CT and chest X ray
Biochemistry: elevated serum CA125 (>35 units/mL but non specific)
Histology: pleomorphic nuclei, high mitotic activity, necrosis, multi-nucleated cells common, prominent lymphocytic infiltrate.

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

What IHC is used to diagnose HGS ovarian cancer?

A

~90% demonstrate nuclear WT1,
~95% abnormal p53 (strong diffuse staining in >80% cells, or no staining), CK7/CA125/PAX8 typically positive
ER often expressed, p16 (CDKN2A) strong diffuse staining in ≥50% tumours,
PTEN and ARID1A retained.
These are useful in differentiating HGSOC from emdometroid

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

What is mainstreaming?

A

All ovarian and breast cancer patients are eligible for germline BRCA testing directly from oncologists to allow for treatment with PARP inhibitors

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

When is somatic BRCA testing required in ovarian cancer?

A

Tumour molecular testing recommended by NCCN after primary surgery or at recurrence/persistence if not performed at this timepoint, to inform utility of PARP inhibitors

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

Is ovarian cancer monitored?

A

Monitoring not currently performed through molecular testing equally across the U.K., CA125 routinely reviewed, however ctTNA is a focus of development for NHSE across solid tumours.

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

What is the NICE recommendation for treatment of stage 1 ovarian cancer?

A
  • Optimal surgical staging (TAH, BSO, infracolic omentectomy, bx of peritoneal deposits)
  • Platinum based chemotherapy- adjuvant chemo (6 x carboplatin) for high-risk Stage I (G3, or Ic) not low-risk (G1/2, Ia/b)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What is the treatment of stage II-IV ovarian cancer?

A
  • Surgical objective to complete resection of all macroscopic disease
  • Intraperitoneal chemotherapy only as part of a clinical trial
  • Chemotherapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

How is recurrent ovarian cancer treated? (or maintenance therapy)

A

PARP inhibitors (dependent on BRCA and HRD status)
- Niraparib/rucaparib maintenance +/- after 1st line PBCT (2+ courses)
- Olaparib maintenance through CDF for FIGO 3/4 after complete/partial response to 1st line PBCT+bev and is HRD+
+/- bevacizumab

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

What are PARP inhibitors?

A
  • PARP is required in the Base Excision Repair pathway, inhibition of which causes an accumulation of DSB
  • Patients with deficient HRR are unable to HiFi repair DSBs, which accumulate and catastrophic DNA damage and cell death
  • This leads to Synthetic lethality
  • Most benefit derived from 1st line PBCT leading to SSB/DNA damage, then PARPi to inhibit mechanism of repair
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What guidelines are used in ovarian cancer?

A

NICE
ESMO-ESGO consensus recommendations
- recommends BRCA germline and somatic testing and HRD testing to identify patients who would respond to PARP inhibitors for maintenance therapy after platinum based chemo

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

What is the prostate?

A

The prostate is a male sex glandular structure
Its main functions are to provide force to ejaculate semen and to add nutrient-rich alkaline fluid to the semen to maintain spermatic health post-ejaculation and enhance fertility

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

What is the process of malignant transformation of the prostate?

A

Malignant transformation of the prostate follows a multistep process, initiating as Prostatic Intraepithelial Neoplasia (PIN) followed by localized prostate cancer and then advanced prostate adenocarcinoma with local invasion, culminating in metastatic prostate cancer.
PIN can be divided into two grades, low (LGPIN) and high (HGPIN). HGPIN has a high predictive value for predicting progression to adenocarcinoma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What is the incidence of prostate cancer?

A

1 in 6 men in the UK will be diagnosed with prostate cancer in their lifetime.
In the UK, it is the most common malignancy in men with around 52,000 diagnosed with prostate cancer each year.

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

What are the risk factors for prostate cancer?

A
  • Age
  • Black ethnicity
  • Family history
  • Obesity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

How does prostate cancer present?

A

Often asymptomatic

  • Lower Urinary Tract (LUT) symptoms (e.g. nocturia, frequency, hesitancy, urgency or retention)
  • Visible Haematuria
  • Abnormal Digital Rectal Examination (DRE)
  • Symptoms of advanced disease (lower back pain/bone pain secondary to bony metastasis, weight loss
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What fusion is present in up to 50% of prostate cancers?

A

The TMPRSS2-ERG fusion plays a significant role in the occurrence and progression of approximately 50% of prostate cancers. This fusion involves the E-26 transformation-specific (ETS)-related gene (ERG) and the transmembrane serine protease 2 (TMPRSS2).

TMPRSS2-ERG regulates androgen receptor (AR) signalling

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

Is there targeted treatments available for prostate cancers with TMPRSS2-ERG fusions?

A

Enzalutamide, inhibits androgen, is more effective in cells or tumors with TMPRSS2-ERG translocations. These tumors exhibit increased AR signaling, making them more responsive to enzalutamide treatment

Investigation into targeted therapies is ongoing

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

What targets are covered on the test directory for prostate cancer?

A

BRCA1, BRCA2
TMPRSS2-ERG
NTRK1/2/3

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

Why is BRCA tested in prostate cancer?

A

Both somatic and germline BRCA variants have been reported in prostate cancer

These patients are eligible for treatment with Olaparib. Testing on somatic tissue should be done first and then blood if tissue not available (R444)

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

What is the diagnostic pathway for prostate cancer?

A

According to ESWMP guidelines
GP
- a digital rectal examination (DRE)
- a Prostate Specific Antigen (PSA) blood test

Imaging
- If elevated PSA, MRI

If positive
- transrectal ultrasound-guided (TRUS) biopsy
- histology, grade, stage and risk stratification

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

What is PSA?

A

Prostate Specific Antigen (PSA) is a protein produced by prostate epithelial cells. PSA is produced by normal prostate tissue, however levels in the blood tend to increase in malignancy. It may be used both in the diagnosis and surveillance of prostate cancer but is not perfect- threshold still in discussion

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

Is there a screening programme for prostate cancer?

A

No
There have been a number of studies that have tried to establish if a screening programme would be an overall benefit to the population.

PSA testing is used sometimes but has a high false positive rate. PSA testing can be discussed with men over 50, and should be offered to those men over 50 who request it.

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

How is prostate cancer staged?

A

TNM
using the 2018 classification for adenocarcinoma of the prostate based on primary tumour (T), lymph node involvement (N) and metastases (M)

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

How is prostate cancer graded?

A

The Gleason score is a histological grade assigned to prostate cancers. From the biopsy, the most common and second most common tumour pattern is assigned a score of 1 to 5 (5 being the highest grade) to give a combined score of 2 to 10.

Grade 1 (score of 6): Cells look simialr to normal prostate cells

Grade 5 (score 10): Cells look very abnormal

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

How is prostate cancer stratified based on risk?

A

Using the Cambridge Prognostic Group (CPG) as low, intermediate and high on the basis the PSA, Gleason score and clinical stage.

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

How is low risk prostate cancer treated?

A
  • Active surveillance: an option in low-risk localised prostate cancer. It involves regular PSA measurements, digital rectal examinations and multiparametric MRIs. It is used as many with low-risk localised disease will have years without disease progression.
  • Radical prostatectomy: is a definitive treatment option for localised prostate cancer. It involves the removal of the entire prostate gland and surrounding tissues.
  • Radical radiotherapy: is a definitive treatment option for localised prostate cancer. May be combined with brachytherapy (implanting radioactive seeds directly in the prostate)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

How is intermediate risk prostate cancer treated?

A
  • Androgen-depravation therapy: this treatment aims to lower androgen levels (most common)
  • Gonadotropin-releasing hormone (GnRH) agonist: cause a ‘chemical castration’. Goserelin is a commonly used GnRH agonist
  • Bicalutamide (an anti-androgen)
  • Bilateral orchidectomy (castration)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

How is high risk prostate cancer treated?

A
  • Neoadjuvent chemotherapy (Docetaxel)
  • Radiotherapy
  • Radical prosteoectomy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

How are relapsed prostate cancer patients treated?

A

Olaparib is NICE recommneded for relapsed BRCA positive prostate cancer patients

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

What is endometrial carcinoma?

A

Endometrial cancer arises from the epithelial lining of the uterus. Historically classified into two classes
1.Hormonally dependent (Indolent, low grade, endometrioid carcinomas)
2.Hormonally independent – (associated with poorer prognosis, non-endometrioid – serous and clear cell morphology, clinically aggressive, that are unrelated to oestrogen stimulation).

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

What are the four classes of endometrial cancer according to The Cancer Genome Atlas (TCGA), ?

A
  • Group 1, with POLE mutations, is associated with a good prognosis.
  • Group 2, with MMR deficiency, is associated with an intermediate prognosis.
  • Group 3, showing low–copy-number alterations, is also associated with an intermediate prognosis.
  • Group 4 tumours, with high–copy-number alterations and TP53 mutations are associated with a poor prognosis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What is the incidence of endometrial cancer?

A

3.4% of all new cancers

Most common invasive cancer of female genital tract

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

What is the prognosis of endometrial cancer?

A

81% 5 year survival rate

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

How does endometrial cancer present?

A
  • The first sign is most often vaginal bleeding not associated with a menstrual period.
  • Pain with urination,
  • Pain during sexual intercourse,
  • Pelvic pain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What are the risk factors for endometrial cancers?

A
  • Obesity (In postmenopausal women, there is greater synthesis of oestrogens in body fat)
  • Diabetes (abnormal glucose tolerance is found in more than in 60%).
  • Hypertension
  • Infertility
  • Women with oestrogen secreting tumours have a higher risk of endometrial cancer.
  • Endometrial cancer is extremely rare in women with no ovaries
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What are the types of endometrial cancer?

A

Endometrioid carcinoma
Serous carcinoma
Clear cell carcinoma
Undifferentiated carcinoma
Mixed carcinoma
Other
Carcinosarcoma

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

How is endometrial cancer graded?

A

FIGO criteria
- Non-gladular, non squamous growth and cytological atypia increases grade

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

What IHC markers are used to identify endometrial carcinoma?

A

Low grade
- ER/PR, p16
High grade
- ARID1A, PTEN, MMR, p53

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

What is serous endometrial carcinoma?

A

Serous carcinomas represent approximately 10% of all endometrial carcinomas but account for as many as 40% of endometrial cancer–related deaths.
The presence of TP53 is supportive of serous carcinoma.

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

What is the function of POLE?

A

Polymerase δ (pol δ) and Polymerase ε (pol ε) are the principal DNA replicases in eukaryotic cells.
-They are coded for by the genes POLD1and POLE
- Pol ε ensures accurate synthesis of the leading strand during DNA replication and is involved in proof reading and error correction

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

What is the affect of POLE LOF variants?

A

Loss of proofreading, catalytic and error-correction during DNA replication that results in an ultramutational state, leading to the accumulation of mutations in the genome

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

What is the frequency of POLE variants in endometrial carcinoma?

A

7-12% have variants in exonuclease domain

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

What variants are known in POLE?

A

11 distinct pathogenic variants have been classified
- accounts for 2/3s of POLE mutant EC

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

What is the MMR system?

A
  • Identifies and corrects base-base mismatches and insertions/deletions
  • Reduces replication-associated errors and contributes to cell cycle arrest and programmed cell death
  • Defects in MMR increase spontaneous mutation rate
  • Leads to mutagenesis in the short term and tumorigenesis in the long term
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

How is MMR endometrial cancer group identified?

A

IHC for MMR genes
- complete loss of one or one heterodimer of MMR
e.g. MLH1 and PMS2 or MSH2 and MSH6

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

What testing does WHO 2022 recommend for classification of endometrial cancer?

A

Reflex testing if negative in the following order:
1. POLE status (NGS)
2. MMR (IHC
3. P53 status (IHC, if inconclusive than NGS)

NGS for TP53 is much more accurate than IHC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

Why is it important to determine the molecular subtype of endometrial cancer?

A
  1. Groups are prognostically distinct
  2. Groups exhibit characteristic responses to therapy that are not captured in existing treatment algorithms- POLE mutated patients are being overtreated
  3. Certain groups are associated with hereditary cancer syndromes (MMR-d and Lynch Syndrome; p53-mut and BRCA1 and BRCA2) – role in screening and prevention
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What does NICE recommend about MMR testing in endometrial cancer?

A

All endometrial cancers are MMR tested using IHC
- if loss of expression of MLH1/PMS2 then reflex for MLH1 promoter hypermethylation as most common cause of somatic loss
- If negative, refer to clinical genetics and send for germline MMR testing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What is the incidence of colorectal cancer?

A

42,896 new cases each year
16,808 deaths
6-7% lifetime risk

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What is the prognosis of colorectal cancer?

A

53% >10 year survival

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What are the risk factors for colorectal cancer?

A
  • Older (age-specific incidence rates increase steeply after age 50, with the highest rates above age 85 years)
  • Male (RR~1.83)
  • Family history of CRC
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

What is the clinical presentation of colorectal cancer?

A
  • Change to bowel habits
  • Loose poo or diarrhoea outside normal for person pattern
  • Constipation outside normal for person pattern
  • Blood in poo
  • Increased frequency of passing stools/ increased urge to pass stool.
  • Abdominal pain
  • Abdominal bloating
  • Losing weight without trying
  • Feeling unusually tired
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What are the WHO classification of colorectal cancers?

A

Adenocarcinoma
Mucinous
Signet ring cell carcinoma
Squamous cell carcinoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What is the diagnostic pathway for colorectal cancer?

A

Colonoscopy
Biopsy
Imaging

ESMO, NICe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What IHC markers are used to identify colorectal cancer?

A

Cytokeritin 20+
Cytokeritin 7-
CDX2+

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What somatic testing is included in the test directory for colorectal cancer?

A

M1.1- KRAS, NRAS, BRAF
M1.4- MSI
M1.5- MLH1 promoter hypermethylation
M1.6- NTRK1/2/3
M1.9- MLH1, MSH2, MSH6, PMS2, POLE, POLD1 (somatic lynch)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

Why are KRAS and NRAS targets for colorectal cancer?

A
  • Activating variants in KRAS and NRAS result in constitutive activation of the RAS/RAF/MEK/ERK pathway.
  • CRC would be eligible for treatment with anti-EGFR therapy e.g. cetuximab or panitumumab.
  • KRAS/NRAS are downstream of this point to inhibition via anti-EGFR therapy would be ineffective.
  • intermediate prognosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

Why is BRAF testing carried out in colorectal cancer?

A

BRAF variants are present in approx 5-10% of CRC cases
Associated with:
- Right side tumours
- Older age
- Female
- MSI-high
Activating variants in BRAF result in constitutive activation of the RAS/RAF/MEK/ERK pathway and will not respond to anti-EGFR therapy
- BRAF V600E carrying CRC can access treatment with encorafenib plus cetuximab per licensing agreement.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What is the prognosis significance of BRAF in colorectal cancer?

A

Prognostic marker:
- poor prognosis if present in microsatellite stable tumour
- good prognosis if present in microsatellite unstable tumour

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

Why is MMR IHC carried out on all colorectal cancers?

A

NICE improving outcomes recommends
- Loss of MMR is indicative of lynch syndrome and starts the lynch reflex pathway

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

What is the prognostic significance of MMRd colorectal tumours?

A

Improved prognosis and good response to immunotherapy
Poor response to 5 FU chemotherapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

What pathway is used if MMRd is identified?

A

Wither IHC or MSI used to identtify potential lynch cases
- MLH1 promoter hypermethylation
- BRAF V600E testing- somatic marker
- Germline testing
- Somatic lynch testing if still no cause identified

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

Why is microsatellite testing used in colorectal cancer?

A
  • When repair pathways are dysfunctional then there can increased instability at repetitive regions of the genome.
  • Microsatellite can be assessed for integrity – please review other notes for details.
  • Microsatellites that show variations in size and slippage indicate that the repair pathways in a tumour have been disrupted and the patient may have Lynch syndrome or somatic Lynch-like syndrome.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

Why is MLH1 promoter hypermethylation used in colorectal cancer?

A

Colorectal and endometrial cancers are all screened by IHC to identify cases showing loss of expression of mismatch repair proteins (MLH1, MSH2, MSH6 and PMS2)

Loss of MLH1 and PMS2 is more commonly due to sporadic events so further testing is required to investigate these IHC results.

MLH1 promoter hypermethylation testing will identify cases where hypermethylation the promoter region prevents expression of MLH1.

This is usually due to hypermethylation in the tumour genome but on occasion germline constitutional hypermethylation of MLH1 promoter has been observed.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

Why are MMR, POLE and POLD1 (M1.9) tested in the tumour of colorectal cancer?

A

Look for the somatic second hit in a Lynch syndrome patient’s tumour
Two somatic pathogenic variants to explain IHC expression loss in a tumour when germline showed no evidence of Lynch syndrome

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

How is localised colorectal cancer treated according to NICE?

A

Surgery + neoadjuvent therapy
If MMR-P- 5-FU
If MMR-d- Capectiabine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

How is metastatic CRC treated according to NICE?

A

MMR-P
- Adjuvent chemotherapy and anti-EGFR therapy

MMR-D
- Adjuvent chemotherapy and anti-EGFR therapy
- immunotherapy)
KRAS/NRAS mut and MMR-p
- adjuvent chemo

KRAS/NRAS mut and MMR-d
- Adjuvent chemo and immunotherapy

BRAF
- BRAF inhibitor plus anti-EGFR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What are the main classifications of lung cancer?

A

Small cell lung cancer (SCLC) - 15%

Non-small cell lung cancer (NSCLC) – 85%
- Adenocarcinoma
- Squamous cell carcinoma (SCC)
- Large cell neuroendocrine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What is the incidence of lung cancer?

A

5-10 per 100K in never-smokers, 20-30x as high in smokers
According to the World Health Organization (WHO), lung cancer accounted for 11.4% of all new cancer cases in 2020

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What are the risk factors for lung cancer?

A
  • Smoking
  • Radon exposure
  • Occupational agents (asbestos, aluminium production, arsenic, coal, dioxin, radiation etc)
  • Air pollution
  • Family history
  • Personal history of lung disease- e.g. COPD
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

How does lung cancer present?

A

> 80% present with locally advanced or metastatic disease​

Primary tumour​
- Persistent cough,
- Haemoptysis (coughing up blood),
- Dyspnea (shortness of breath), ​
- Chest pain​

Metastasis​
- Headache/seizures​
- Bone pone​
- Weight loss​
- Fatigue

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

How is lung cancer diagnosed?

A

Imaging
- Chest X ray followed by PET, CT
Biopsy
-Histological subtyping and TNM staging
- IHC used

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

What IHC markers are used to histologically subtype SCLC?

A

Cytokeratin AE1/3 +
TTF1 +
Synaptophysin +
CD56 +
Napsin A and p40 -

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

What IHC markers are used to histologically subtype lung adenocarcinoma?

A

Cytokeratin AE1/3 +
TTF1 +
Napsin A +
Synaptophysin -
CD56 -
p40 -

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What IHC markers are used to histologically subtype lung squamous cell carcinoma?

A

Cytokeratin AE1/3 +
p40 +
TTF1 -
Napsin A -
Synaptophysin -
CD56 -

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

How is SCLC treated?

A

Stage 1: Surgery and adjuvent chemo/radiation
Stage 2-4: incurable, no targeted therapies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What is the characteristic tumour profile of SCLC?

A

TP53 and RB1 LOF
High TMB
Associated with heavy smokers so G>T and C>A transversions
High number of CNVs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What are the SCLC test directory targets?

A

RB1 CNV: used in equivocal cases to aid diagnosis and to identify possible resistance mechanism to chemotherapy
NTRK fusions: treatment eligibility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

How is stage 4 NSCLC treated?

A

Incurable so treated with systemic therapies
- Based on the presence of targeted therapy and PD-L1 expression
- If <50% PD-L1: platinum based chemo +/- pembrolizumab
- if >50% PD-L1: pembrolizumab +/+ pemetrexed and carboplatin
- if targeted event

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What test directory targets are there for NSCLC?

A

SNV: EGFR, KRAS, BRAF, MET, ALK
CNV: MET
Structural: ALK, ROS1, RET, NTRK1, NTRK2, NTRK3
ctDNA: EGFR, ALK

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

What is the frequency, common mutations and clinical relevance of EGFR variants in NSCLC?

A
  • 10-15%
  • Activating missense variants e.g. L858R, deletion in exon 19 and insertions in exon 20
  • Predicts response to EGFR TKI’s
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What acquired resistance mechanisms are seen in EGFR treated NSCLC?

A

EGFR variants e.g. T790M, C797S
MET amplification

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What EGFR TKIs are used to treat NSCLC?

A

Erlotinib and Gefitinib
- First-generation EGFR inhibition
- Most common resistance mechanism is Thr790Met in exon 20

Afatinib, Dacomitinib
- Second-generation ERBB family blockade

Osimertinib
- Third-generation, targets T790M
- Most common EGFR-based resistance mechanism is Cys797Ser in exon 20

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What is the clinical significance of EGFR exon 20 insertion in NSCLC?

A

Will not respond to standard EGFR TKIs

Mobercertinib
- TKI specific for EGFR exon 20
-Now removed from NICE commissioning due to adverse affects

Amivantamab
- Monoclonal antibody for EGFR exon 20
- Licensed but not yet NICE recommended, may access through special circumstances

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What is the frequency, common mutations and clinical relevance of KRAS variants in NSCLC?

A

25%
SNVs and indels affecting G12 and Q61
Poor prognosis and G12C predicts repsonse to KRAS G12C specific TKIs
- Co-occurring TP53 associated with increased immunotherapy response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What targeted treatments are available for KRAS mutated NSCLC?

A

KRAS notoriously difficult to target therapeutically
Sotorasib
- G12C specific
- Available through CDF
- Second line treatment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What treatments are available for BRAF mutated NSCLC?

A

Only for Class 1 V600 variants
Dabrafenib plus trametinib (BRAF/MEK combination therapy)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What is the frequency, common mutations and clinical relevance of BRAF variants in NSCLC?

A

1-2%
Three classes of BRAF variants
- Class 1: V600 variants
- Class 2: Non-v600 variants, activating
- Class 3: Kinase dead
Class 1 BRAF variants are associated with poor prognosis and lower response to immune checkpoint inhibitors but response to BRAF.MEK inhibition

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What is the frequency, common mutations in MET variants in NSCLC?

A

3-4% Splicing (exon 14 skipping)
2-4% Amplification

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What is the clinical significance of MET exon 14 skipping variants in NSCLC?

A

Predicts response to MET TKI- Tepotonib

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What is the clinical significance of MET amplification in NSCLC?

A

Secondary MET amplification is associated with acquired resistance to EGFR TKI
Can also be primary oncogenic event

No NICE approved therapy but some evidence of response to criztonib

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

How are MET variants detected?

A

Exon skipping
- DNA splicing variants
- RNA exon skipping
- between 60-90% concordance in DNA but RNA is gold standard

Amplification
- NGS CNV calling
- FISH (confirmation)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What is the frequency, common mutations and clinical relevance of ALK variants in NSCLC?

A

ALK Fusions e,g, ELM4-ALK (3-7%) inv(2)(p21;p23)
Predicts response to ALK inhibitor therapy- rapid response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What treatments are available for ALK rearranged NSCLC?

A

Crizotinib
- First generation
- Approved for first and second line

Certinib and Alectinib
- Second generation

Lorlatinib
- Third generation
- Sensitive to ALK resistance mutations including G1202R
- no longer approved first-line (July 2023), second line only

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What resistance mechanisms are there to ALK TKIs in NSCLC?

A

ALK SNVs e.g. G1202R, L1196M
ALK amplification
MET amplification

Different mechanisms of resistance may be sensitive to different lines of treatment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

What is the frequency, common mutations and clinical relevance of RET variants in NSCLC?

A

RET fusions 1%
KIF5B-RET​, CCDC6-RET
Response to Selpercatinib

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What is the frequency, common mutations and clinical relevance of ROS1 variants in NSCLC?

A

ROS1 Fusions 2%
CD74-ROS1​
EZR-ROS1
Response to Crizotinib​
- ROS1 and ALK share 49% amino acid sequence homology so crizotinib can bind to both

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What is the frequency, common mutations and clinical relevance of NTRK variants in NSCLC?

A

NTRK fusions 1%
LMNA-NTRK1​
ETV6-NTRK3 t (12; 15) (p13; q25)
Response to Larotrectinib and Entrectinib​
- Available once all treatment options have been exhausted

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

What other non-TD targets are used in NSCLC?

A

ERBB2
- Response to ERBB2 TKI’s
- Amplification is a resistance mechanism to EGFR TKI
PIK3CA
- response to targeted therapy
TP53
- increased response to immune checkpoint

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

How are structural variants detected in NSCLC?

A

RNA sequencing panels
FISH
IHC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

What is the role of ctDNA in NSCLC?

A

EGFR and ALK ctDNA currently on the test directory​
- Currently delivered by a national 2-hub model for NSCLC​
- Can be used when a tumour is inaccessible to biopsy ​
- Potential disease monitoring mechanism and potentially identify resistance variants prior to disease progression​
- More representative picture of the tumour heterogeneity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

What is the prognosis of lung cancer?

A

NSCLC: 5-year survival 5%
SCLC: 5 year survival at 2%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

What guidelines are used in NSCLC?

A

NICE
ESMO

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

What is melanoma?

A

Melanoma arises from an accumulation of mutations that transforms melanocytes, most commonly in the skin.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

How is melanoma classified according to the WHO?

A

Melanoma arising due to UV radiation
- Low-CSD (cumulative sun damage) melanoma- superficial spreading melanoma
- high-CSD melanoma- Lentigo maligna melanoma , Desmoplastic melanoma 

Melanoma not associated with CSD 
- Spitz
- Acral
- Mucosal
-Uveal
- Melanoma arising within blue naevi 
- Melanoma arising in congenital naevi 

Nodular Melanoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

How does melanoma evolve from a benign naevus?

A

Benign naevus
- Harbours only a single mutation and no other pathogenic alterations
- e.g. BRAF V600E

Melanocytoma
- Evolved from benign naevi with additional pathogenic mutations but have not yet reached malignant state

Malignant melanoma
- acquired additional pathogenic mutations and becomes malignant

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What is the incidence of melanoma?

A

5th most common cancer- 4% of all new cancer cases
Adult incidence: 20 million cases per year

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What are the most common melanoma in different populations?

A

Caucasain: superficial spreading
Asian, hispanic and African- Acral melanoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What are the risk factors of melanoma?

A

86% of cases are preventable
- Fitzpatrick skin type 1: never tans (always burns), fair skin, blue eyes, red/blond hair, freckles
- history of blistering sunburn
- genetic predisposition
- immunosupression
- Congenital nevi
- Environmental factors (tanning beds, sun exposure)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

How does cutaneous melanoma present?

A

New/changing pigmented skin or mucosa lesions. Lesions may be crusting, itching and may bleed.

ABCDE model
A - Asymmetry: Asymmetric in shape, size, or color.
B - Border Irregularity: The edges of the mole or lesion are irregular, ragged, notched, or blurred, rather than smooth and well-defined.
C - Colour Variation: The mole or lesion exhibits uneven colouring or multiple colours
D - Diameter: >5mm or is increasing in size over time.
E - Evolution: The mole or lesion has undergone changes in size, shape, colour, or texture over time, or new symptoms such as itching, tenderness, or bleeding have developed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

What test directory targets are there for melanoma?

A

SNVs: BRAF, NRAS, KIT
Structural: NTRK1/2/3
CNVs: MYB, RREB1, CCND1, MYC, CDKN2A- for equivocal cases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What is the frequency, common mutations and clinical relevance of BRAF variants in melanoma?

A

50% - most commonly superficial spreading melanoma
BRAF V600 are the most common
Associated with aggressive disease and makes patient eligible for NICE approved targeted therapy BRAF/MEK combination therapy (only V600 variants): Dabrafenib+Trametinib/ Encorafenib+binimetinib

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

What is the frequency, common mutations and clinical relevance of NRAS variants in melanoma?

A

15-20% of melanoma
NRAS Q61 are the most common, typically mutually exclusive with BRAF V600
Associated with aggressive disease
There is no targeted treatment but there is some evidence that patients may respond to MEK inhibitors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

What is the relevance of concurrent NRAS and BRAF variants in melanoma?

A

NRAS variants can be found with Class 3 BRAF variants. This results in elevated oncogenic potential through activation of the MEK/ERK pathway and these tumours may be more likely to respond to MEK inhibitors.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

What is the frequency, common mutations and clinical relevance of KIT variants in melanoma?

A

Present in 10.8% of melanomas, most commonly high CSD (Lentigo maligna melanoma)
Targeted treatment: Fair responses to TKIs such as imatinib and sorafenib but this is not licensed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

What is the frequency, common mutations and clinical relevance of NTRK variants in melanoma?

A

NTRK fusions are present in <1% cutaneous/mucosal melanoma but 20-30% spitzoid melanomas

ETV6:NTRK3 t(12 15)(p13 q25)

Can aid in diagnosis of spitzoid neoplasm

NICE approved targeted treatment available for these patients- NTRK inhibitors Larotrectinib and entrectinib- only once all treatment options have been exhausted

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

What TD targets are there for uveal melanoma?

A

BRAF, NRAS, NF1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

What is the frequency, common mutations and clinical relevance of NF1 variants in melanoma?

A

10-15% of melanomas

LOF variants in NF1 are particularly common in lentigo maligna melanoma.
Associated with very high TMB
Not a test directory target for melanoma but there is some preclinical evidence that these patients may respond to MEK inhibition.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

What are the common mutations and clinical relevance of GNAQ and GNA11 variants in melanoma?

A

Codon Q209 hotspot variant dominates Uveal (and Blue Neavi) melanoma, whilst BRAF V600 is unusual in this subtype

Some response to MEK inhibitor, but no improvement in overall survival

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What is the frequency, common mutations and clinical relevance of BAP1 variants in melanoma?

A

3-4% ocular melanoma
PARP inhibitors being explored for these patients
Germline variants associated with BAP1 predisposition syndrome
Associated with greater metastatic disease potential

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

What is the frequency, common mutations and clinical relevance of TERT promoter variants in melanoma?

A

43% melanomas, especially non-acral skin​
Associated with older age, sun exposure, thicker tumours, BRAF variants​
Most common in superficial spreading​
Associated with poorer prognosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

What molecular analysis can be used to help diagnose equivocal melanocytic lesions?

A

Copy number panel either using array or FISH
- RREB1
- MYB
- CCND1
- C-MYC
- CDKN2A
- Centromere 6

This is particularly useful when diagnosing spitz melanoma which can be diagnostically challenging to distinguish Spitzoid melanoma from benign Spitzoid melanocytic lesions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

What is TMB?

A

Tumour Mutation Burden (TMB)

The total number of somatic/acquired mutations per coding area of a tumour genome (Mut/Mb).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

Why is TMB important in melanoma?

A

Melanoma is known to have some of the highest TMB scores compared with other solid tumour types.
TMB however varies within melanoma subtypes, with NF1 mutated and High CSD cutaneous melanoma having the highest TMB
High TMB is a positive predictor of response to immunotherapy with checkpoint inhibitors such as anti-PD1 (nivolumab, pembrolizumab) and anti-CTLA4 (ipilimumab),

Not all high TMB tumours will respond to immunotherapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

How is melanoma diagnosed?

A

Imaging
Sentinel lymph node biopsy
Scar re-excision
Histology to subtype and stage
IHC for protein overexpression or loss of expression, e.g p16 (CDKN2A)
Equivacol cases: FISH, SNP arrays or NGS to detect a CNV ‘melanoma pattern’ or variants that may inform on subtypes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

What IHC is used in the diagnosis of melanoma?

A

S100
MelanA
HMB45
SOX10

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

How is melanoma staged?

A

TNM: tumour nodes metastases​

T​: Breslow thickness​ (depth of tumour in mm from skin), Ulceration​

N​: Largely based on number of involved nodes​

M: Location of distant metastases​

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

How is melanoma prognosis determined?

A

Stage
Serum LDH
BAP1 loss/monosomy 3 is poor prognosis in uveal melanoma
BRAF/NRAS poor prognosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

How is localised melanoma treated?

A

Surgical removal/resection, with or without radiotherapy (depending on histological types and resection margins)

Sentinel lymph node biopsy is recommended for Breslow thickness of >1mm, ulcerated thinner melanoma may also be indicated for SLNB

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

How is metastatic melanoma treated?

A

Stage III
Surgical resection followed by PD-1 inhibitor OR
BRAF plus MEK inhibitor in V600 mutant melanoma
Lymphadectomy of involved LN if benefit is likely to outweigh the significant rate of morbidity associated with this procedure

Unresectable III and untreated stage IV
Checkpoint inhibitor combination therapy Pembrolizumab, Nivolumab (anti-PD-1); Ipilimumab (anti-CTLA-4))
BRAF plus MEK inhibitor if immunotherapy unsuitable or rapid progression/high disease burden) - Dabrafenib+Trametinib/Encorafenib+binimetinib;
Chemotherapy or best supportive care

Previously treated stage IV
Check point inhibitor Combination or monotherapy
BRAF-MEK inhibitor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

What is the prognosis of melanoma?

A

Patients with localised disease, and tumours <1mm deep have a >95% survival rate at 5 years.
Relapse occurs in a significant proportion of people with stage IIA to IIC melanoma (up to 50% at 5 years in people with stage IIC melanoma)
<30% diagnosed with distant metastasized melanoma survive over 5 years

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

What guidelines are used in melanoma?

A

ESMO
NICE
NCCN

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

What are the considerations when deciding whther a melanoma patient should start on immunotherapy or targeted therapy?

A

Start with immunotherapy
Long-lasting effect, and still have targeted therapy in the bag if/when needed​
But relatively high rate of side effects, and significant rate of non-response​
But fairly slow onset​

Start with targeted therapy
Rapid effect (days/weeks)​
Generally higher response rate​
Lower rate of side effects​
But relatively short duration of response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

What is the incidence of CNS tumours?

A

Glioblastoma: annual incidence is approximately 3 per 100,000 per year and increases with age and male sex.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

What are the risk factors associated with CNS tumours?

A

Age
Obesity
Exposure to vinyl chloride – glioma
EBV infection – implicated in primary CNS lymphoma
Transplant recipients and patients with AIDS – increased risk of primary CNS lymphoma
Ionising radiation
Radiofrequency radiation
Occupational exposures e.g. Meningiomas may be associated with lead exposure.
Family syndromes: NF1, NF2, VHL, LFS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

How do patients with CNS tumours present?

A

Headaches
Seizures
Visual changes
GI symptoms e.g. loss of appetite, nausea, vomiting
Personality, mood, mental capacity, concentration changes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

How are CNS tumours diagnosed?

A

Imaging
MRI: used as standard as has good tissue resolution. CT scan used when MRI is contradictory
PET: used to determine tumour from necrotic regions (metabolically active)

Histology
Biopsy- morphology, staining, IHC to determine grade/stage

Molecular
IDH1, MGMT, ATRX, 1p/19q codeletion, histone mutations
All involved in determining the subtype

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

What poor prognostic factors are associated with CNS tumours?

A

Age >40 years
Progressive disease
Tumour size >5cm
Tumour crossing midline
Contrast enhancement on MRI
WHO performance status (>1)
Neurological symptoms
Less than gross total resection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

How have the WHO 2021 CNS classification guidelines changed from the 2016 guidelines?

A

The 2021 WHO classification emphasizes molecular diagnostics alongside traditional histopathology- integrated diagnosis.
Gliomas have also been distinguished into adult and paediatric
Introduced grading within tumour types

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

How are CNS tumours classified?

A

Adult-type diffuse gliomas
Pediatric-type diffuse low-grade gliomas
Pediatric-type diffuse high-grade gliomas
Circumscribed astrocytic gliomas
Glioneuronal and neuronal tumors
Ependymal tumors
Embryonal tumors
Meningioma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

What are the major classifications of adult gliomas according to WHO 2021?

A
  1. Diffuse Astrocytic and Oligodendroglial Tumors
    IDH-mutant gliomas
    - Astrocytoma, IDH-mutant (graded 2, 3, or 4)
    - Oligodendroglioma, IDH-mutant and 1p/19q-codeleted (graded 2 or 3)
    IDH-wildtype gliomas
    - Glioblastoma, IDH-wildtype (automatically grade 4)
    - Diffuse midline glioma, H3 K27-altered (grade 4)
    - Diffuse hemispheric glioma, H3 G34-mutant (grade 4)
  2. Circumscribed Astrocytic Gliomas (Non-diffuse)
    - Pilocytic astrocytoma (grade 1)
    - Pleomorphic xanthoastrocytoma (grade 2)
    - Subependymal giant cell astrocytoma (grade 1)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

What molecular markers are used to classify adult gliomas according to WHO 2021?

A

Learn diagram

IDH (Isocitrate Dehydrogenase) Status
IDH-mutant: Associated with a better prognosis.
IDH-wildtype: Often associated with a poorer prognosis

1p/19q Codeletion
Presence of this codeletion is characteristic of oligodendrogliomas.

Histone Mutations
Mutations in histone genes such as H3 K27M (common in diffuse midline gliomas) and H3 G34R/V.

ATRX: rules out oligodendroglioma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

How are CNS tumours graded according to WHO 2021?

A

Grade 1: Typically benign, slow-growing, and well-circumscribed tumors (e.g., Pilocytic astrocytoma).

Grade 2: Low-grade, but infiltrative and have potential for malignant transformation.

Grade 3: Anaplastic, more aggressive than grade 2.

Grade 4: Highly malignant and aggressive, such as glioblastomas and certain diffuse midline gliomas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

What TD targets are there for gliomas?

A

SNVs: IDH1, IDH2, ATRX, H3-3A,H3C2, BRAF, TERT promoter
Structural: NTRK, EGFR vIII, BRAF
Methylation: MGMT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

What are the common mutations and the clinical significance of IDH1/2 in gliomas?

A

IDH1 R132 and IDH2 R172
First stage of WHO classification (astrocytomas and oligodendrogliomas)
Better prognosis compared to IDH-wildtype gliomas.
Associated with a more favorable response to treatment, including radiotherapy and chemotherapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

What is the role of IDH enzymes and how do mutant IDH drive CNS tumours?

A

Isocitrate dehydrogenase (IDH) enzymes, of which there are three isoforms, are essential enzymes that participate in several major metabolic processes, such as the Krebs cycle, glutamine metabolism, lipogenesis and redox regulation.

Mutations usually in the isocitrate binding site, reducing isocitrate binding and altering cellular metabolism

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

How is IDH mutant detected in CNS?

A

IHC
- R132H-specific Ab
- 85-90% R132H but can’t detect other variants

NGS
- Panel genes, full gene sequencing

Targeted hosptot

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
195
Q

What is the clinical significance of 1p/19q codeletion in gliomas?

A

Characteristic of oligodendrogliomas.

Associated with a better prognosis and response to therapy.

Key criterion for differentiating between oligodendrogliomas and astrocytomas.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
196
Q

How is 1p/19q codeletion detected in gliomas?

A

ATRX IHC: Surrogate marker based upon mutual-exclusivity with 1p19q co-deletion. Rapid, relatively cheap, and labour unintensive.

FISH: Reliable and cost effective. FISH cannot differentiate between whole chr. Arm deletion from smaller focal deletions.

SNP arrays: can identify whole-arm co-deletion with higher reliability – better choice but labour intensity

PCR based microsatellite analysis: can allow detection of LOH at selected loci and NGS-based methods also useful

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

What is the clinical significance of ATRX in gliomas?

A

Commonly found in IDH-mutant astrocytomas, rules out oligodendroglioma.

ATRX mutation is mutually exclusive with 1p19q co-del

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

What is the clinical significance of TP53 in gliomas?

A

Frequently found in IDH-mutant astrocytomas

Can be detected by IHC or NGS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

What is the clinical significance of MGMT in gliomas?

A

Promoter methylation

Predicts response to alkylating agent chemotherapy (e.g., temozolomide) in glioblastomas- associated with longer survival

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

What is the role of MGMT?

A

Removes alkyl groups and repairs mutagenic DNA lesions, preventing DNA damage and subsequent apoptosis. Therefore key in maintaining DNA integrity, but can provide a survival / cell death resistance mechanism by preventing cancer cells from entering the apoptotic pathway when subject to chemo-radio therapy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
201
Q

How is MGMT promoter hypermethylation detected in gliomas?

A

Bisulfite conversion: Convert unmethylated cytosine residues to uracil, using bisulphitation of DNA (methylated cytosine is resistant to this chemical alteration)

Pyrosequencing
MLPA
Methylation specific PCR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

What is the clinical significance of TERT in gliomas?

A

Almost all IDH-mutant, 1p/19q-codeleted oligodendroglial tumours have activating mutations in TERT gene promoter region – valuable diagnostic marker.
Associated with a poor prognosis.
TERT promoter mutations, along with EGFR amplification and +7/-10 copy number changes, are indicative of glioblastoma, IDH-wildtype.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
203
Q

What are the common TERT mutations seen in glioma and what do they do?

A

c.-124C>T, c.-146C>T

Produces a binding site for ETS transcription factors- gains the ability to recruit transcription factors that might upregulate TERT protein expression.

Leads to telomere maintenance in tumour cells

Detected by NGS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

What is the clinical significance of histone H3 mutations in gliomas?

A

H3 K27M: Found in diffuse midline gliomas, particularly in the brainstem, thalamus, and spinal cord; associated with a very poor prognosis (2 year surival <10%) and classified as WHO grade 4, paediatric

H3 G34R/V: Found in diffuse hemispheric gliomas, typically in children and young adults; also associated with a poor prognosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
205
Q

What is the clinical significance of BRAF mutations in gliomas?

A

Common in pediatric low-grade gliomas

V600E in ganglioglioma

KIAA1594-BRAF fusion present in 70% pilocytic astrocytoma- aids diagnosis

Can be targeted by specific inhibitors (e.g., vemurafenib) in cases of refractory or recurrent tumors.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
206
Q

How are BRAF mutated gliomas treated?

A

V600E
- BRAF/MEKi FDA approved but not yet NICE approved
- Could access compassionally

KIAA1549:BRAF
- MEK inhibitor trial

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

What is the clinical significance of CDK2NA/B mutations in gliomas?

A

Indicative of a more aggressive tumor and associated with poor prognosis in IDH-mutant astrocytomas.

Important for grading and determining the aggressiveness of the tumor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

What is the clinical significance of EGFR mutations in gliomas?

A

Most frequently amplified in glioblastoma (30-40%), often associated with structural abnormalities e.g. mutant protein EGFRvIII – exon 2-7 skipping, characterised by deletion of extracellular domain (seen in ½ of all with amplification).

Most useful in diffuse gliomas lacking IDH mutations which fail to show high grade features (necrosis, MVP), a confounding presentation; EGFR amp confirms GBM, IDH-WT, WHO IV.

Poor prognosis

Potential therapeutic target

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
209
Q

How are paediatric gliomas classified according to the WHO?

A

Pediatric-type diffuse low-grade gliomas
- Diffuse astrocytoma, MYB- or MYBL1-altered
- Angiocentric glioma
- Polymorphous low-grade neuroepithelial tumor of the young
- Diffuse low-grade glioma, MAPK pathway-altered

Pediatric-type diffuse high-grade gliomas
- Diffuse midline glioma, H3 K27-altered
- Diffuse hemispheric glioma, H3 G34-mutant
- Diffuse pediatric-type high-grade glioma, H3-wildtype and IDH-wildtype
- Infant-type hemispheric glioma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
210
Q

What are embryonal tumours?

A

Replaces Peripheral neuroectodermal tumour (PNET)
Predominantly occur in children
Histologically characterised by high cellularity with densely packed, poorly differentiated small cells and marked mitotic activity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
211
Q

What are the types of CNS embryonal tumours?

A

Includes:
Medulloblastomas
Embryonal tumours with multi-layered rosettes (ETMRs)
Atypical teratoid/rhabdoid tumours (AT/RTs)
+ heterogenous group of other embryonal CNS tumours

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
212
Q

How are medulloblastomas classified?

A

WNT activated

SHH activated/TP53 WT

SHH activated/TP53 mutant

Non-WNT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
213
Q

Outline WNT activated meduloblastoma

A

10%
>90% carry CTNNB1 mutation
Children have good prognosis but adults less favourable

214
Q

Outline SHH activated/TP53 WT meduloblastoma

A

30%
Infants and adulthood
PTCH1 or SUFU germline mutations

215
Q

Outline SHH activated/TP53 mutant meduloblastoma

A

Small percentage of SHH-activated medulloblastomas
Predominantly children
Show large cell/anaplastic phenotype
Poor prognosis
Up to 50% have germline TP53 mutation

216
Q

Outline non WNT/SHH mutant meduloblastoma

A

Group 3 and group 4” molecular categories

Group 3 = 20%
Group 4=40%

Children and infants

Lack of easily accessible diagnostic tools - grouped together

Histologically almost always show classic or large cell/anaplastic phenotype

217
Q

What are Atypical teratoid/rhabdoid tumours?

A

Underlying defect in SMARCB1 or rarely SMARCA4

Products of these are essential components of SWI/SNF chromatic remodelling complex.

218
Q

What are Embryonal tumours with multi-layered rosettes

A

Primarily affects children
Highly aggressive

C19MC Amplification or TTYHI-C19MC. This amplification is a key diagnostic marker.

LIN28A overexpression, a protein involved in the regulation of microRNAs and embryonic development.

219
Q

What mutation are associated with Embryonal tumours of the pineal and pituitary region?

A

RB1 (RB transcriptional corepressor 1) mutations (+/- germline defect) and DICER1 (Dicer 1, ribonuclease III) mutations linked to pineoblastoma

Pituitary – DICER1 mutation key predisposing event

220
Q

How is glioblastoma treated?

A

The standard of care in newly diagnosed GBMM includes maximal safe surgical resection, followed by concurrent radiotherapy and temozolomide (TMZ) and six monthly cycles of adjuvant TMZ.

Treating symptoms- steroids, Anti-convulsant (seizures)

221
Q

What is the recurrence rate of glioblastoma?

A

90%

Median PFS of 1.5–6 months and median OS of 2–9 months

222
Q

What guidelines are used in CNS tumours?

A

WHO
NICE
ESMO

223
Q

What is the thyroid gland?

A

The thyroid gland is an endocrine gland in your neck. It makes two hormones that are secreted into the blood: thyroxine (T4) and triiodothyronine (T3). Regulates vital functions including breathing, heart rate, nervous system and body temperature

224
Q

What is the incidence of thyroid cancer?

A

GLOBOCAN 2018 database, thyroid cancer (TC) accounts for 3.1% of all new cancer diagnoses worldwide and, therefore, ranks in 9th position with regard to incidence
CRUK states TC represents 1% of all UK cancers (2016-18 data)- 20th most common in UK
Estimated annual 41,000 deaths,

225
Q

What are the risk factors associated with thyroid cancer?

A

History of radiation therapy administered in infancy or childhood for benign H&N conditions
Radiation exposure as a consequence of radioactive fallout
Family history of thyroid disease or multiple endocrine neoplasia (MEN) / other inherited syndromes
Frequent sporadic variants
A history of goitre (swelling of thyroid, deficient function, >90% due to iodine deficiency)
Female sex (2-3 times more likely)
Age (in 30’s and over 60s)

226
Q

How does thyroid cancer present?

A

Majority are asymptomatic
May present witha painless lump or swelling in the front of the neck
Sore throat
Difficulty swallowing (dysphagia)
Unexplained hoarseness

227
Q

What are thyroid nodules?

A

Thyroid lesion that is palpable and/or distinct from the surrounding area
Over 90% are small, non-palpable, benign lesions;
however, identification of clinically significant, malignant thyroid nodules that will cause morbidity if not diagnosed early is important.

228
Q

How is thyroid cancer diagnosed?

A

Physical examination and history
Ultrasound
Laryngoscopy
Fine-needle aspiration biopsy of the thyroid
Tissue Biopsy
Blood test: Blood hormone studies (TSH…), blood chemistry studies (calcitonin, CEA)
CT scan

229
Q

How are thyroid cancers subtyped?

A

Usually by ultrasound and histopathology
Sometimes definitive classification can be challenging and molecular profiling can aid classification as well as stratify targeted therapy

230
Q

How are mailgnant thyroid neoplasms classified?

A

Thyroid cancers are derived from either follicular thyroid cells or neuroendocrine cells
According to WHO 2022
Differentiated thyroid cancer
- Papillary
- Follicular
- Poorly differentiated
Undifferentated thyroid cancer
- Anaplastic
Neuroendocrine
- Medullary

231
Q

What are the proportion and prognosis of papillary thyroid cancer?

A

60-80% of all cases
5 year survival of 90%
Good prognosis
Mean age <40

232
Q

What are the proportion and prognosis of follicular thyroid cancer?

A

30-50%
5 year survival of 90%
Good prognosis
Mean age 40-60

233
Q

What is poorly differentiated thyroid cancer?

A

5-7%
More aggressive, less differentiated form of follicular-derived thyroid cancer, intermediate between differentiated thyroid cancer and anaplastic thyroid cancer.

234
Q

What are the proportion and prognosis of anaplastic thyroid cancer?

A

2%
5 year survival of 10%
Very poor prognosis
Mean age >60
Almost always presents with metastasis, mortality is 100%

235
Q

What are the proportion and prognosis of medullary thyroid cancer?

A

5-7%
5 year survival of 80%
Poor prognosis
Mean age 10-60

236
Q

What molecular events define thyroid cancer development?

A

Progression from Follicular thyroid cells to papillary thyroid cancer (PTC) or follicular thyroid cancer (FTC) is marked by mutations activating the MAPK- or PI3K-AKT signalling pathways
Further progression to poorly differentiated thyroid cancer (PDTC) is characterised by additional mutations and rearrangements, affecting both signalling pathways simultaneously.
Anaplastic thyroid cancer (ATC) is characterised by further genetic events, especially in oncogenes like TP53, as well as epigenetic alterations and infiltration of immune cells, creating an intracellular microclimate that favours genetic instability and oncogenesis.

237
Q

What is familial medullary thyroid cancer?

A

Caused by germline change in the RET gene

About 20% cases are caused by this

If multiple tumours seen then known as multiple endocrine neoplasia type 2 (MEN2)

MEN2A is associated with pheochromocytomas (tumours that make adrenaline) and with parathyroid gland tumours.

MEN2B is associated with pheochromocytomas and with benign growths of nerve tissue on the tongue and elsewhere called neuromas

238
Q

Outline the MAPK pathway

A

The MAPK-signaling pathway is crucial for growth and proliferation of the cell.
The signaling cascade is initiated on the cell surface by a “mitogen” stimuli, a growth factor, that binds to the tyrosine kinase receptor (RTK) and ultimately results in the phosphorylation and activation of Mitogen-Activated Protein Kinase (MAP2K), which is also known as MEK, and the downstream extra cellular signal-regulated-kinase (ERK).

239
Q

Outline the PI3K/AKT signaling pathway

A

The PI3K/AKT signalling pathway is involved in the regulation of apoptosis, proliferation, cell cycle progression, angiogenesis, cytoskeleton integrity, and energy metabolism, and ultimately leads to AKT and mTOR activation.

240
Q

What is the role of RAS in the MAPK and PIK3 signaling pathway in thyroid?

A

RAS mutations can activate both MAPK- and PI3K/AKT pathways. However, it seems like RAS mutations preferable activate the PI3K-AKT pathway in thyroid oncogenesis.

There are three isoforms of RAS-NRAS, HRAS and KRAS, where NRAS is the most commonly mutated in human TC.

RAS mutation is thought to be a premalignant mutation, and that additional mutations are needed to set off carcinogenesis

241
Q

What TD targets are there for thyroid cancer?

A

SNVs: BRAF, KRAS, NRAS, HRAS, RET, TERT promoter, TP53

Structural: RET, ALK, NTRK1, NTRK2 and NTRK3

242
Q

What is the clinical significance of BRAF variants in thyroid cancer?

A

BRAF V600E

Exclusive in papillary thyroid cancer and papillary thyroid cancer derived anaplastic thyroid cancer

Poor prognosis

May respond to BRAF/MEK inhibition- not NICE approved, FDA approved

243
Q

What is the clinical significance of KRAS, NRAS and HRAS variants in thyroid cancer?

A

Codons 61, 12 and 13

Common in follicular adenomas

244
Q

What is the clinical significance of TERT promoter variants in thyroid cancer?

A

Poor prognostic marker, associated with aggressive disease

Even worse prognosis if found in combination with BRAF V600E

245
Q

What is the clinical significance of RET SNVs variants in thyroid cancer?

A

M918T most common

Found in medullary thyroid carcinoma

Associated with poor prognosis

RET inhibitors available for these patients- cabozantinib is approved and may be given regardless of RET status. Selpercatinib available through CDF as second line

246
Q

What is the clinical significance of RET fusions variants in thyroid cancer?

A

CCD6::RET
NCOA4::RET

Found in 7% of papillary thyroid cancers- highly specific. Also ATC (derived from papillary)

RET inhibitors available for these patients- cabozantinib is approved and may be given regardless of RET status. Selpercatinib available through CDF as second line

247
Q

What is the clinical significance of TP53 variants in thyroid cancer?

A

TP53 mutations are rare in differentiated thyroid cancers but are more common in anaplastic thyroid carcinoma (ATC) and poorly differentiated thyroid carcinoma (PDTC).

They are associated with:
Highly aggressive disease.
Poor prognosis.
Resistance to conventional therapies.

248
Q

What is the clinical significance of ALK fusions in thyroid cancer?

A

Seen in a small number of anaplastic thyroid cases

Associated with younger patient age

Potential response to ALK inhibitors e.g. Crizotinib but not NICE approved

249
Q

What is the clinical significance of NTRK fusions in thyroid cancer?

A

LMNA-NTRK1
ETV6-NTRK3 l t(12;15) (p13; q25)

Most commonly seen in papillary thyroid cancer but still rare

Eligibility for treatment with NTRK inhibitors entrectinib and lorentrectinib- exhausted all treatment options

250
Q

How is differentiated thyroid cancer treated?

A

ESMO guidelines indicate that asymptomatic patients with stable disease should undergo active surveillance only (CT; serum TG and TgAb)

Progressive disease:
- thyroidectomy (low risk can have lobectomy)
- If high risk ( tumour >2cmm, nodal and metastasis), radioidine therapy is used

If refractory, sorafenib / lenvatinib > cabozantinib TKI’s which are VEGFR inhibitors

NTRK/RET inhibitors if suitable

251
Q

How is medullary thyroid carcinoma treated?

A

Surgery resection is primary treatment- thyroidectomy

Do no respond to radioactive iodine or conventional chemotherapy

Two kinase inhibitors, vandetanib (multi-kinase inhibitor of VEGFR, EGFR, RET) and cabozantinib, are approved for advanced medullary thyroid cancer without predictive testing but reserved for patients with structural disease progression

Pralsetinib and selpercatinib should be considered for RET mutant tumours.

252
Q

How is anaplastic thyroid cancer treated?

A

Debulking surgery to remove tumour blocking the airway

External beam irradiation therapy with radio-sensitizing chemotherapy

Adjuvent radiotherapy in all cases

Consider targeted therapies e.g. BRAF/MEKi

253
Q

What is the role of miRNA in thyroid cancer?

A

Diagnostic power of miRNAs in thyroid cancer has been evaluated by several groups. miRNAs not only can differentiate malignant tissues from non-malignant tissues, but also have differential expression in different stages of thyroid cancer. Assessment of serum levels of miRNAs is a practical non-invasive method for follow-up of patients after thyroidectomy.

254
Q

What is the incidence of GIST?

A

Gastrointestinal stromal tumours (GISTs) are the most common mesenchymal neoplasms of the gastrointestinal tract with an annual incidence of approximately 10–15 cases per million.

They usually present sporadically in older adults (median age 60–65 years) with slight male predominance. Strong female predilection is seen in paediatric SDH-deficient GIST.

255
Q

What are the risk factors of GIST?

A

Hereditary GIST
- Germline variants in KIT or PDGFRA (5%)

256
Q

How do GISTs present?

A

GISTs present asymptomatically in 18% of cases, especially in cases of smaller tumours of the intestinal tract. These tumours are usually found incidentally on abdominal CT scans, during endoscopy, or during surgical procedures for other manifestations.

Symptomatic patients may present with nonspecific symptoms of nausea, vomiting, abdominal distension, early satiety, abdominal pain, and rarely as a palpable abdominal mass.

Larger tumours may cause obstruction of the gastrointestinal lumen leading to dysphagia, obstructive jaundice, or constipation, depending on the location of the mass.

257
Q

How are GISTs diagnosed?

A

Imaging
- CT
- MRI
- PET

Histology
- Biopsy
- IHC, normally positive for CD117 and/or DOG1- helps distinguish GISTs from other mesenchymal tumours

Molecular
- KIT and PDGFRA mutations can help diagnose
- In KIT/PDGFRA wt GIST, SDHB to idenify SDH- deficient GIST
- BRAF and NTRK variants can also be found
- In quadruple-negative GIST (for KIT/PDGFRs/BRAF/SDH), an unrecognised underlying NF1 syndrome should be excluded

258
Q

How is GIST prognosis determined?

A

Tumour
- Size: larger tumours increased risk
- Mitotic rate: >5 mitoses per 50HPF
- Location: Gastric GISTs better prognosis than intestine
- Metastasis: worse prognosis

Molecular
- KIT mutated more aggressive than PDGFRA
- KIT exon 9 have the worst outcome
- SDH deficient tumours are less predictable

259
Q

What is GIST prognosis?

A

Localised: 95%.

Metastasis - 84%.

Distant metastasis - 52%.

260
Q

What TD targets are there for GIST?

A

SNVs: KIT, PDGFRA
RNA: NTRK1/2/3

261
Q

What molecular abnormalities are associated with sporadic paediatric GIST?

A

SDH Mutations:
Pediatric GISTs often exhibit a deficiency in the succinate dehydrogenase complex, particularly SDH subunits (SDHA, SDHB, SDHC, SDHD).

Loss of SDHB Expression: This is a hallmark of SDH-deficient GISTs, identified by immunohistochemistry.

NF1 related deficiency

KIT and PDGFRA variants not normally seen

262
Q

What syndromes are associated with paediatric GIST?

A

Carney-Stratakis Syndrome: A hereditary condition characterized by the presence of GIST and paragangliomas due to germline mutations in SDH subunits.

Carney Triad: A non-hereditary syndrome involving GIST, paragangliomas, and pulmonary chondromas, often associated with SDH deficiency.

NF1- associated GISTs

263
Q

What is the incidence and significance of KIT variants in GIST?

A

80% of GISTs

Lead to constitutive activation which activates downstream Ras/Raf/MAPK and the PI3K/AKT pathway

264
Q

What is the clinical significance of KIT exon 11 variants in GIST?

A

Most frequent, affecting the juxta-membrane domain.

They are mostly caused by in-frame deletions within codon Gln550 and Glu560, missense mutations and duplications

Deletions affecting codons 557–558 of exon 11 of the KIT gene have been reported in up to 28% of all GISTs and have been associated with high-risk tumors

Repsond to imatinib

265
Q

What is the clinical significance of KIT exon 9 variants in GIST?

A

10% of cases

Affecting extracellular domains

Duplications of Ala502- Tyr503

These tumors are commonly located in the small bowel and are often associated with a more aggressive phenotype.

Double dose Imatinib is recommended for these variants.

266
Q

What is the percentage, common mutations and clinical significance of PDGFRA in GISTs?

A

8-10% of GISTs

Exon 12, 14 and 18 (most common) mutations

Favourable prognosis

Respond to treatment with Imatinib

Often located in the stomach

267
Q

What is the significance of the D842V PDGFRA mutation?

A

Results in a distortion of the kinase activation loop, tilting the protein conformation in favour of the activated structure. Imatinib can only bind inactive form

Resistant to treatment with imatinib

Avapritinib is specifically designed to target D842V

268
Q

What primary resistance mechanisms are associated with GIST treatment?

A

Primary resistance- tumour progression within the first 6 months of treatment

PDGFRA D842V Mutation- prevents imatinib from binding

Wild type GIST: lacking both KIT and PDGFRA variants

KIT exon 9 mutations: require double dosage, standard dose is insufficient

269
Q

What secondary resistance mechanisms are associated with GIST treatment?

A

Secondary KIT mutations- usually occur in 12-36 months

  • These mutations typically occur in exons encoding the ATP-binding pocket (exon 13 and 14) or the activation loop (exon 17 and 18)- alter the binding site of the TKI, reducing its effectiveness e.g. V654A in exon 13 of the KIT gene.

Alternative pathway activation: activation of the PI3K/AKT/mTOR pathway or the upregulation of other receptor tyrosine kinases.

270
Q

How is GIST treated (first line)?

A

Surgical excision of lesion

Adjuvant therapy with imatinib 400 mg/day for 3 years for patients with a significant risk of relapse.

KIT exon 9 mutation: adjuvant imatinib at a higher dose of 800 mg

PDGFRA exon 18 D842V: avapritinib (specifically designed)

271
Q

In the case of imatinib resistance, what other treatments can be used in GIST?

A

Standard second-line treatment is sunitinib.

Regorafenib is the standard third-line therapy.

Ripretinib is the standard fourth-line treatment.

272
Q

What is wild type GIST?

A

Approximately 10–12% of all GISTs lack mutations in KIT and PDGFRA and are called wt-GISTs.

Can be sub-classified in an SDH-competent and an SDH-deficient group

SDH IHC used to help classify

273
Q

What are the types of SDH competent wt GIST?

A

NF1 mutant GIST
- NF1 is an inherited, autosomal dominant disease
- Approximately 7% of patients with NF1 develop GIST
- somatic inactivating NF1 mutations have also been seen

BRAF, KRAS and PIK3CA-Mutant GISTs
- BRAF mutations (V600E) have been found in approximately 8–13% of wt-GISTs
- mutually exclusive with KIT/ PDGFRA
- Rare
- BRAF/MEKi available

ETV6-NTRK3
- Rare
- NTRK inhibitors

274
Q

What is SDH deficient wt GIST?

A

Defect in the SDH complex (mitochondrial complex or succinate reductase) which is composed of 4 subunits SDHA, SDHB, SDHC and SDHD.

The SDH enzyme is a key enzyme in the Krebs cycle and electron transport chain

SDH-deficient GISTs usually occur in patients younger than 40 years of age, have a female predilection, occur in the stomach

275
Q

What targeted treatments are available in GISTs?

A

KIT, PDGFRA: Imatinib

PDGFRA D842V: Avapritinib

NTRK Fusion: Entrectinib, loretrectinib

BRAF: BRAF/MEKi (not NICE approved)

276
Q

What are sarcomas?

A

Tumours that develop in the muscle, bone, nerves, cartilage, tendons, blood vessels and the fatty and fibrous tissues. Named after tissue in which they originate.

Include:
- Soft tissue
- Primary bone
- GIST

Commonly harbour gene fusions

277
Q

What are soft tissue tumours?

A

Soft tissue tumours are a type of sarcoma and are very rare (<1% of all cancers).

Largely benign with a very high cure rate after surgical excision.

Soft tissue tumours are a therapeutic challenge as there over 100 histological subtypes.

~30% of soft tissue tumours have disease specific rearrangements; the rest can often have random complex karyotypes.

278
Q

How are recurrent translocations detected in soft tissue tumours?

A

G banding
- Rarely used but is possible
- Difficult on FFPE as poor quality

FISH
- Used as standard
- Quick, can be done on small amounts of tissue with low number of abnormal cells
- but itnerpretation is difficult, partner not identified and can miss some rearrangements

RT-PCR
- Cheap and quick
- Need to know partner to have correct primers

NGS
- Often used as well as FISH
- High throughput, can detect partners
- Poor quality RNA leads to high failure rate and longer TaT

279
Q

What are small round blue cell tumours?

A

Group of tumours aising from different tissues (including sarcomas) characterised by small, round, relatively undifferentiated cells, which range from benign to incredibly aggressive

Appear ‘blue’ when H&E stained due to the high nuclei to cytoplasm ratio

Most commonly seen in children so often fall under the umbrella of ‘paediatric tumours’ (but can be seen in adults)

10% have germline predisposition

280
Q

How are small round cell tumours classified?

A

Typically very difficult to classify due to the similarities in histology (lack of differentiation), demographic prevalence, clinical history and disease site

Includes:
Ewing sarcoma
Rhabdomyosarcoma
Synovial sarcoma
Neuroblastoma
Medulloblastoma
Desmoplastic small round cell tumours
Rhabdoid tumours
Liposarcoma
Inflammatory myofibroblastic tumour

281
Q

What are the types of undifferentiated small round cell sarcomas according to WHO 2022?

A

Ewing sarcoma

Round cell sarcoma with EWSR1::non-ETS fusions

CIC-rearranged sarcoma

Sarcoma with BCOR genetic alterations

282
Q

What is Ewing’s Sarcoma?

A

Ewing’s sarcoma is a malignant tumor that grows in the bones or the soft tissue, such as cartilage.

6-8% of primary bone tumours

It is most common in children and young adults. In rare cases, Ewing sarcoma occurs in adults.

283
Q

How does Ewing’s sarcoma present?

A

Palpable mass: Most often in the long bones of the arms and legs, the pelvis, or the chest. It can also develop in the skull or the flat bones of the trunk. Lesion shows ‘onion skin’

Fever, weight loss, swelling (metastatic)

The tumour often metastasises to the lungs and other bones.
At the time of diagnosis, spread is seen in about one third of children with Ewing sarcoma.

284
Q

What is the prognosis of Ewings sarcoma?

A

Five year survival is ~75%
25% if metastatic at diagnosis

Favourable prognosis if complete pathological response

Prognostic role of STAG2, CDKN2A, TP53 variants being validated- rare

285
Q

How is Ewings sarcoma monitored?

A

Future:

Potential for use of ctDNA to detect EWSR1::FLI1

Potential for circulating tumour cells to detect CD99+ cells

Likely to emerge in the coming years

286
Q

How is Ewings sarcoma diagnosed?

A

Core needle biopsy

Essential diagnostic criteria:
- diffuse infiltrate of small round cell morphology
- CD99+ by IHC- strong diffuse membranous staining

Desirable criteria:
- identification of FET:ETS fusion usually EWSR1
- EWSR1 rearrangement by FISH and NGS

Imaging
- MRI to detect solid tumour mass

287
Q

What molecular rearrangements are seen in Ewing’s sarcoma?

A

EWSR1-FLI1 (85%)- t(11;22)(q24;q12)

EWSR1::ERG (10%),

Other EWSR1/FUS fusions (<10%)

Fusion partner may have prognostic value so usually detected by EWSR1 break apart FISH, followed by NGS

Additional mutations at diagnosis are rare

288
Q

How does EWSR1-FL1 drive Ewing sarcoma?

A

Transcriptional activating domain of EWSR1 and DNA binding domain of FL1
Producing an aberrant transcription factor that drives oncogenesis by dysregulating gene expression and promoting uncontrolled cell proliferation and survival.

289
Q

What is the on the test directory for Ewing sarcoma?

A

FISH: EWSR1 rearrangement

WGS

Structural: EWSR1, NTRK

290
Q

How is Ewings sarcoma treated?

A

Local surgery/resection

And/or Radiotherapy

And Intensive multi-agent chemotherapy (good repsonse)

Currently no targeted therapies available- difficult to target as fusion protein lacks enzymatic activity and lack obvious pockets for small molecule binding

291
Q

What is the rate of relapse for Ewing sarcoma?

A

Relapse is mostly systemic (71-73%), followed by combined (12-18%) and local (11-15%) relapse

5 year post relapse survival rate of 15-25%, with local recurrence faring better than systemic

292
Q

What is rhabdomyosarcoma?

A

Small round cell tumour

Mesenchymal tumours typically associated with the skeletal muscle, , and approximately 50% of cases are diagnosed in the first decade of life.

293
Q

What is the incidence of rhabdomyosarcoma?

A

The most common soft-tissue sarcoma of childhood, annual incidence of 4.5/1 million children.

Accounts for 5% of malignancies in children

294
Q

What are the subtypes of rhabdomyosarcoma

A

Embryonal rhabdomyosarcoma (ERMS) – 80% , best prognosis

Alveolar rhabdomyosarcoma (ARMS) – 20%, unfavourable prognosis.

Two rare subgroups recognized by WHO: spindle cell and pleomoprhic

295
Q

What molecular abnormalities are associated with Rhabdomyosarcoma?

A

Alveolar type: PAX3::FOXO1 t(2;13)(q35;q14),
PAX7::FOXO1 (more favourable outcome compared to PAX3).

Spindle cell: MYOD1 L112R (very poor prognosis)

Embryonal type: RAS variants common but no recurrent chromosomal abnormalities

296
Q

How is rhabdomyosarcoma diagnosed?

A

Histology
- Small round cell morphology
- Some skeletal muscle differentiation
- Desmin+ by IHC

Molecular
- FOXO1 rearrangement by FISH and NGS

Imaging

297
Q

What is the incidence of rhabdomyosarcoma and how does it present?

A

4.5 per million per year
Lump/swelling, painless mass

298
Q

What is the prognosis of rhabdomyosarcoma?

A

Low-risk RMS five-year survival ~80%;
high-risk RMS five-year survival ~20-30%

299
Q

What is synovial sarcoma?

A

Small round cell tumour
Rare, highly aggressive

5-10% of all soft tissue tumours and10-20% in young adults

Age range 5-85, median age 35 years.

Small proportion caused by LFS or NF1

300
Q

What is the prognosis of synovial sarcoma?

A

5-year survival rate is as low as 36%.

301
Q

Who gets synovial sarcoma?

A

1/3 occur in <20 years

77% before 50y

Females more commonly affected

302
Q

How does synovial sarcoma present?

A

Painful mass usually near joints of arm, leg or neck. Initial slow growth often delays Dx. Aggressive growth invade adjacent bone

50% patients exhibit metastatic disease, of which 74-81% are to the lungs.

303
Q

What molecular abnormalities are associated with synovial sarcoma?

A

SS18::SSX1 (approx. 60% case) - t(X;18)(p11.2;q11)
SS18::SSX2 (30%), SS18::SSX4 (<10%)

SYT-SSX1 fusions have poorer prognosis- controversial

304
Q

How is synvovial sarcoma treated?

A

Surgery

Chemotherapy

B-catenin inhibitors, MTOR inhibitors, RTKs inhibitors (inhibit PDGFR), BCL2 and EZH2 inhibitors

305
Q

What is neuroblastoma?

A

Affects immature nerve cells (neuroblasts), typically arising in the adrenal glands or along the sympathetic nervous system.

Most often affects children <5 years and rarely adults, more common in boys

306
Q

What molecular abnormalities are associated with neuroblastoma?

A

Familial neuroblastoma – germline mutations in the ALK and PHOX2B

The PHOX2B gene is important for the formation and differentiation of nerve cells.

Deletion of regions of chromosome 1 and chromosome 11 are associated with neuroblastoma.

~ 25 percent have MYCN amplification- unclear clinical relevance

307
Q

What is Medulloblastoma?

A

Small round cell tumour

Medulloblastoma (MB) is one of the most common childhood malignant brain tumours.

Originates within the brainstem or the cerebellum

Although many patients now survive, can develop chronic health complications due to treatments

308
Q

What is the incidence of medulloblastoma and how does it present?

A

5 cases per million per year

Headaches, poor academic performance, hydrocephaly

Associated with Turcot syndrome, Gorlin syndrome

309
Q

What are the subgroups of medulloblastoma?

A

WNT (10%)
- Muations affecting WNT pathway e.g. CTNNB1, DDX3X and SMARCA4
- low risk

SHH (30%)
- Mutations in SHH pathway e.g. PTCH1, SMO, SUFU
- further classified into TP53 mut (very high risk) and TP53 wt (intermediate risk)

Group 3 (20%)
- MYC amplification
- Intermediate to high risk

Group 4 (40%)
- PRDM6 overexpression
- Low to intermediate risk

310
Q

What is desmoplastic small round cell tumour?

A

Small round cell tumour

Tumors that grow in the abdomen and pelvic area of the body. DSRCT is very rare. DSRCT occurs most often in young white males between the ages of 10 and 30.

311
Q

How does DSRCT present?

A

Often asymptomatic
When the tumors get larger, symptoms can include: pain, nausea and vomiting diarrhea, constipation, swelling in the abdomen.

312
Q

What is the prognosis of DSRCT?

A

5 year survival- 15%

313
Q

What is the molecular abnormality associated with DSRCT?

A

Characterised by the EWSR1::WT1
t(11;22)(p13;q12)

314
Q

How is DSRCT treated?

A

Surgery

Chemotherapy

Hyperthermic intraperitoneal chemotherapy (HIPEC): This treatment washes the inside of the abdomen with warm chemotherapy drugs- kills the tumor cells in the abdomen without exposing the rest of the body to the drugs, which can cause side effects.

Radiation Therapy

315
Q

What are rhabdoid tumours?

A

Small round cell tumour

Rare, aggressive type of childhood cancer that can start in the kidneys, soft tissues or CNS.

Primarily affects infants and young children

316
Q

What is the prognosis of rhabdoid tumours?

A

Poor prognosis with median survival 18 months,
5 year survival 30%

317
Q

What molecular abnormality is associated with rhabdoid tumours?

A

Majority of RTs arise as a consequence of biallelic inactivation of SMARCB1

Can be detected by IHC, PCR, NGS - not specific to diagnosis

25-30% with rhabdoid tumour on brain have a germline variant in SMARCB1

318
Q

How are rhabdoid tumours treated?

A

Surgery, radiotherapy and chemotherapy

Current preclinical trials looking at targeting of epigenetic proteins e.g. HDAC inhibitors, DNMT inhibitors (azacitadine)

319
Q

What are lipomas?

A

Lipomas are benign tumour’s composed of fat cells.

Most common adipocytic neoplasm (and also the most common soft tissue neoplasm).

Commonly have rearrangements of HMGA2 and rarely HMGA1

320
Q

What is liopsarcoma?

A

Small round cell tumour

Rare type of cancer that develops in the adipose tissue, which can be divided into four main types:

Well-differentiated liposarcoma (most common, 50%)

Dedifferentiated liposarcoma (progresses from well-differentiated liposarcoma)

Myxoid liposarcoma (more frequently found in limbs) 15-20%

Pleomorphic liposarcoma (rare, fast-growing)

321
Q

What is well differentiated liposarcoma?

A

Most common liposarcoma

Also known as atypical lipomatous tumor as these tumours show no potential for metastasis

Characterized by MDM2 amplification

Low grade

322
Q

What molecular abnormalities are seen in liposarcoma?

A

Well differentiated and de-differentiated:
MDM2 amplification

Myxoid:
FUS-DDIT3 t(12;16)(q13;p11) or EWS-DDIT3

323
Q

What is clear cell sarcoma?

A

CCS is a malignant neoplasm typically involving deep soft tissue of the extremities in close proximity to tendons and aponeurotic structures.

Very rare tumor of young adults with melanocytic differentiation.

324
Q

What is the incidence of clear cell sarcoma?

A

The exact incidence is largely unknown, although occasional case series mention CCS comprising less than 1% of all soft tissue sarcoma

325
Q

What molecular abnormalities are seen in clear cell sarcoma?

A

Hallmark is t(12;22)(q13;q12): EWSR1::ATF1- 90% of cases.

A related variant translocation t(2;22)(q32.2;q12) involves EWSR1-CREB1 (6%).

326
Q

What is extraskeletal myxoid chondrosarcoma?

A

Extraskeletal myxoid chondrosarcoma (EMC) is a rare soft-tissue malignancy

EMC is a malignant mesenchymal neoplasm of uncertain differentiation characterized by abundant myxoid matrix.

Chondrosarcoma usually refers to a cancer derived from cells that produce cartilage, however despite its name there is no convincing evidence that EMC has cartilaginous differentiation.

Aggressive but good prognosis

327
Q

What molecular abnormalities are seen in extraskeletal myxoid chondrosarcoma?

A

Characterised by t(9;22)(q22;q12.2)- EWSR1::NR4A3

Small proportion have BP56-NR4A3

NR4A3 fusions present in >90% of EMCs and are not present in any other sarcoma- diagnostic

328
Q

What are the subtypes of renal cell carcinoma?

A

Clear cell RCC (ccRCC) – most common 75%-80% of the cases

Papillary RCC (pRCC) – 15% of the cases – previously has been subdivided to type 1 and 2, which has now been removed in the new WHO 5th edition (2022).

Chromophobe RCC (chRCC) – 5% of the cases

TFE3 rearranged RCC

TFEB rearranged RCC

SDH deficienct RCC

hereditary leiomyomatosis

329
Q

What is the prognosis of renal cell carcinoma?

A

Different RCC histological subtypes were associated with specific survival outcomes.

Patients with ccRCC had significantly poorer survival than those with pRCC or chRCC (P = 0.0001 and P = 0.0008, respectively)

TP53 poor prognosis in ccRCC, pRCC

CDKN2A loss or PTEN mutation poor prognosis in chRCC

330
Q

How has the WHO 5th edition changed for RCC?

A

Uses molecular classifications more than orevious additions

Changed clear cell papillary renal cell carcinoma to clear cell papillary renal cell tumour as there has been no evidence of metastasis in this indolent tumour

331
Q

What is the incidence of RCC?

A

Renal cell carcinoma (RCC) accounts for 2% of all cancers globally and is the cause of 2% of cancer deaths.

Slight male prevalence – M:F = 1.58:1 (2017).

332
Q

What are the risk factors for RCC?

A

Obesity
Cigarette smoking
Hypertension
Acute kidney- papillary RCC.
Patients on long-term haemodialysis due to end-stage renal disease develop renal cysts and have an increased risk of renal cancer (3–7%)

Most are sporadic but there are herediatary syndromes associated with RCC

333
Q

How does RCC present?

A

Haematuria (blood in the urine),
loin pain,
malaise,
weight loss,
anaemia (from depression of EPO)
hypertension (due to the excess secretion of renin by the tumour). Approximately 5% of patients will have polycythaemia.

334
Q

How is RCC diagnosed?

A

Most cases of RCC are strongly suspected by imaging.
Suspicion of RCC should prompt laboratory examinations of serum creatinine, haemoglobin, leukocyte and platelet counts, lymphocyte to neutrophil ratio, lactate dehydrogenase, C-reactive protein (CRP) and serum-corrected calcium.

335
Q

What TD targets are there in RCC?

A

Used to classify subtypes rather than targeted treatments

SNV: FH, BRAF, MET, VHL, SDHA, SDHB, SDHC, SDHD, TCEB-1 (ELOC), TSC1, TSC2

Structural: TFEB, TFE3 and NTRK

336
Q

What is clear cell renal cell carcinoma?

A

Derived from renal tubular epithelial cells
80% of all RCC.
The cells have clear and occasionally eosinophilic cytoplasm

Associated with biallelic VHL inactivation.

337
Q

What abnormalities are associated with clear cell RCC?

A

Inactivation of the VHL complex is the defining signature event in ccRCC.

Normally del3p affecting one allele and inactivating intragenic mutations or promoter region methylation of the second VHL allele

Also frequent gain of 5q, loss of 14q

338
Q

When might clear cell RCC suggest hereditary cancer?

A

Multifocal and/or bilaterar
Early age of onset
Suggests a hereditary cancer syndrome such as von Hippel–Lindau syndrome.

5% of cases

339
Q

What is papillary RCC?

A

Papillary adenomas usually occur in the renal cortex, particularly in the capsular or subcapsular area.

Virtually all papillary adenomas are clinically silent and discovered as incidental findings.

340
Q

What abnormalities are associated with papillary RCC?

A

MET, EGFR, KMT2C, trisomy 7 and 17 and loss of Y

No relevant diagnostic abnormalities according to WHO

341
Q

What is chromophobe RCC?

A

The main goal in diagnosis of chRCC, is the differential diagnosis with oncocytoma.

Chromophobe RCCs have diffuse positivity for cytokeratin 7 (CK7), whereas oncocytomas are negative or present focal positivity for CK7.

342
Q

What molecular abnormalities are associated with chromophobe RCC?

A

Known pattern of single-copy chromosome losses of chromosomes 1, 2, 6, 10, 13 and 17

PTEN, TSC1, TSC2 and MTOR, TERT promoter

343
Q

Why are the DNA targeted for RCC tested for?

A

VHL: associated with clear cell renal cell carcinoma, aid diagnosis and response to VEGF treatment

MET: associated with papillary RCC. Potential repsonse to MET inhibitors

FH: Classify as FH deficient RCC, Germline mutations in the FH gene are associated with hereditary leiomyomatosis and renal cell cancer (HLRCC).

SDHA-D: Variants allow classification of Succinate Dehydrogenase (SDH) Deficient Renal Cell Carcinoma

ELOC: ELOC rearranged RCC

344
Q

What is the clinical significance of TFE3 and TFEB rearrangments in RCC?

A

TFE3 rearrangements are characteristic of TFE3 rearranged RCC, which is a distinct subtype of RCC, formally MITF Xp11 translocation RCC). These tumors typically occur in children and young adults but can also be found in older patients.

TFEB rearrangements are associated with a rare subtype of RCC known as TFEB rearranged RCC. These tumors are also known as TFEB-amplified RCC

345
Q

What are cholangiocarcinomas?

A

Tumour located in the periphery of the second-order bile ducts, ranging from segmental bile ducts to smaller branches of the intrahepatic biliary tree.

346
Q

How are cholangiocarcinomas classified?

A

Intrahepatic CCA (iCCA): located in the periphery of the second-order bile ducts,
Perihilar CCA: arises at the junction where the right and left hepatic ducts meet,
Distal CAA: common bile duct

347
Q

How does cholangiocarcnioma present?

A

CA19-0 elevated

Clinical symptoms are dependent on where the lesions originate in the biliary tree and are typically absent in early stage tumours.

Nonspecific symptoms (such as weight loss, abdominal discomfort, and malaise).

Jaundice, pruritus, and pale stool when the tumour is large enough to obstruct the biliary flow.

348
Q

What is the incidence of cholangiocarcinoma?

A

Rare adult tumour with a global incidence of <6 in 100,000, although incidence in rising

Intrahepatic CCA is second most common primary hepatic malignancy behind hepatocellular carcinoma and accounts for 10-15% of cases (2).

Higher incidence in south-east Asia (Thailand >80 in 100,000).

349
Q

What are the risk factors for cholangiocarcinoma?

A

Obesity
Smoking
Hypertension
IBS
Primary sclerosing cholangitis
Chronic biliary stones
Chronic liver disease
Family history

350
Q

What is the prognosis of cholangiocarcinoma?

A

Prognosis is poor (5-year OS of <20%) due to late diagnosis and high rate of relapse

351
Q

How is cholangiocarcinoma diagnosed?

A

Blood test
- CA19-9 raised
- Elevated liver function tests

Imaging
- Ultrasound, CT, MRI

Histology

352
Q

What is on the TD for cholangiocarcinoma?

A

SNV: IDH1

Structural: FGFR2, NTRK

MSI- lynch syndrome suspected

353
Q

What molecular abnormalities are seen in cholangiocarcinoma?

A

iCCA
- IDH1/2 mutations (20%)
- BAP1 mutations (10–20%)
- FGFR2 fusions (15%).

dCCA
KRAS (20%)
SMAD4 (10–20%).

Mutations in TP53 are observed in either type (30%).

Loss of MMR (could be indicative of lynch)

354
Q

What targeted treatments are available for cholangiocarcinoma?

A

IDH1 R132 variant: Ivosidenib (second line)

FGFR2 fusion: Pemigatinib (relapsed)

MMR loss/ MSI-high: Pembrolizumab

355
Q

How is cholangiocarcinoma treated?

A

Surgery is currently the only curative treatment option
Due to tumour heterogeneity, chemotherapy has poor effectiveness Radiotherapy is rarely used against this tumour type.

Stage I-II
- Surgery of possible
- Adjuvent chemotherapy if non curative resection

Stage III-IV
- Unresectable
- Chemotherapy: Cisplatin
- Immunotherapy /targeted treatments

356
Q

What is the recurrence rate of cholangiocarcinoma?

A

Tumour recurrence occurs in ~70% of patients

Long-term survival can rarely be expected once tumour recurrence occurs because the recurrence means ‘systematic disease’

357
Q

What is heptocellular carcinoma?

A

Hepatocellular carcinoma (HCC) is the most common form of liver cancer and accounts for ~90% of cases.

358
Q

How does heptocellular carcinoma develop?

A

Pre-cancerous cirrhotic nodules with low-grade dysplasia, called low-grade dysplastic nodules (LGDNs).

LGDNs subsequently develop into high-grade dysplastic nodules (HGDNs) that can transform into early-stage HCC (stages 0 and A) and progress into more advanced HCC (stages B and C).

359
Q

What is the incidence of heptocellular carcinoma?

A

906 000 people were diagnosed with liver cancer globally.

5-year survival rate of approximately 18%.

age between 60 and 70

predominantly affects men.

360
Q

What are the risk factors for heptocellular carcinoma?

A

Over 90% of HCC cases occur in the setting of chronic liver disease as a consequence of chronic infections with the hepatitis B virus (HBV) or hepatitis C virus (HCV), alcohol abuse or alcoholic steatohepatitis (ASH), and nonalcoholic fatty liver disease (NAFLD) or non-alcoholic steatohepatitis (NASH).

Obesity, diabetes, and smoking

Haemochromatosis or hereditary tyrosinaemia type 1.

HIV

361
Q

How is heptocellular carcinoma diagnosed?

A

Solid hepatic nodules ≥1 cm, especially in patients with liver cirrhosis trigger investigation

Imaging

Biopsy
- IHC for glypican 3, HSP70 and Glutamine synthetase
- All 3 markers allows diagnosis

362
Q

What is on the test directory for heptocellular carcinoma?

A

NTRK- for NTRK inhibitors

363
Q

What molecular abnormalities are associated with heptocellular carcinoma?

A

Currently no mutations are used to predict therapeutic response (other than NTRK)

TERT promoter (60% of cases)
TP53 (30%)
CTNNB1 (30%)
ARID1A (10%)

364
Q

What causes germline predisposition to heptocellular carcinoma?

A

A1ATD is characterized by low levels of serum A1AT and affects 1:1800 to 1:2000 live births.

It is the most common genetic cause of liver disease in children and increases risk for cirrhosis and HCC.

365
Q

How is hepocellular carcinoma treated

A

Liver resection or transplantation is the main curative option (5 year survival 75-80%)

Thermal ablation- radiofrequency ablation to injure the tumour through EM energy

Systemic disease
- Sorafenib and Lenvatinib- first line TKI
- Regorafenib- second line TKI
- Pembrolizumab

366
Q

What is oesophageal cancer?

A

Histopathological subtypes are: Oesophageal squamous cell carcinoma (ESCC) and oesophageal adenocarcinoma (EAC).​

ESCC is the predominant subtype that usually arises from squamous epithelial cells of the oesophagus, whereas EAC originates from glandular cells present near the stomach and is believed to be largely related to acid exposure of the lower oesophagus.​

367
Q

What is the prognosis of oesophageal cancer?

A

EC is a highly aggressive, malignancy with over 400,000 deaths annually, worldwide. Both ESCC and EAC carry poor outcomes with a five-year overall survival rates of 15%​

368
Q

What are the risk factors of oesophagael cancer?

A

Smoking
Alcohol
Hot beverage drinking
Poor nutrition

369
Q

What molecular abnormalities are seen in oeasophageal carcinoma

A

HER2- only approved biomarker

TP53 (72%), ELMO1 (25%), DOCK2 (12%), CDKN2A (12%), ARID1A (9%), SMAD4 (8%) and PIK3CA (6%). ​

Gain of KRAS (21%), HER2 (19%), EGFR (16%), CND1 (10%) and MET (6%)

Loss of SMAD4 (34%), CDKN2A (32%) and ARID1A (10%) are found.​

370
Q

What targeted therapies are there in oesophageal cancer?

A

HER2 testing – trastuzumab

PD-L1 testing - found in 35% to 45% of oesophageal cancers. Pembrolizumab

MMR and MSI testing -Pembrolizumab.

371
Q

What is the prognosis of gastric cancer?

A

Annual 723,100 deaths
Recurrence occurs in up to 30–40% of patients within 5 years

372
Q

What are the risk facts of gastric cancer?

A

Diet
Smoking
Alcohol
EBV infection
Family history

373
Q

What hereditary cancer syndromes are associated with gastric cancer?

A

The strongest association is found in hereditary diffuse gastric cancer (CDH1) syndrome, in which approximately 80% of patients will develop gastric cancer.​

Others with a much lower risk include:​
Lynch, ​
Hereditary breast and ovarian cancer (BRCA), ​
Li-Fraumeni, ​
Familial adenomatous polyposis, and ​
Peutz-Jegher syndromes.​

374
Q

What are the molecular subtypes of gastric cancer?

A

Subtype 1 (ARID1A+ type) is characterized by high ARID1A and PIK3CA mutations and is associated with favorable prognosis

Subtype 2 (TP53+ type) harbors a highly recurrent TP53 mutation, is associated with poor prognosis

Subtype 3 (CDH1+ type) is accompanied by a high CHD1 mutation and is associated with a poor prognosis

375
Q

How does gastric cancer present?

A

Often present with an upper endoscopy report performed for symptoms, including dyspepsia and reflux,​

Also with symptoms or signs that may indicate advanced disease, such as dysphagia, weight loss, gastrointestinal bleeding, anemia, and emesis.

376
Q

How is gastric cancer treated?

A

Stage 1-3
Resectable: gastrectomy
- Triplet chemotherapy

Stage IV
- Palliative resections
- Endoscopic stents
- Palliative chemotherapy
- HER2+ - trastuzumab
- PD-L1/MSI - pembrolizumab

377
Q

What is a small bowel adenocarcinoma?

A

Rare malignancy with worse prognosis compared to other cancers of the gastrointestinal tract. ​

378
Q

What is the incidence of small bowel adenocarcinoma?

A

11,390 new cases of small bowel cancer were estimated, with 2100 cancer-related death. ​

379
Q

What targetable mutations are seen in small bowel adenocarcinoma?

A

HER2

BRAF V600E

dMMR/TMB

380
Q

What is the incidence of adenoid cystic carcinoma?

A

3-4.5/million annual incidence; 1% of all head and neck cancer

Most common of minor salivary glands

381
Q

How does adenoid cystic carcinoma present?

A

Mean age range 50-60.

Indolent and slow growing, with few symptoms

1/3 patients present at with T4 tumour

Symptoms, when eventually occurring, may include facial nerve paralysis, oroantral fistulas, and nasal obstruction.

382
Q

How is adenoid cystic carcinoma diagnosed?

A

Biopsy
- histological subtypes tubular, cribriform, solid
- MYB IHC used
- often followed up by sequencing
- MYB::NFIB most common (80%)

Test directory
- MYB, NTRK

383
Q

What molecular abnormalities are seen in adenoid cystic carcinoma?

A

MYB rearrangements
- MYB::NFIB (80% of cases) and MYBL1::NFIB (5%)
- Rearrangements lead to deletion of 3’ UTR regions, disabling the regulatory activity of miRNAs and resulting in overexpression of MYB genes.

Copy number
- KIT gain: poor prognosis

384
Q

How is adenoid cystic carcinoma treated?

A

Curative surgery

5-FU + CAP (cisplatin, doxorubicin, and cyclophosphamide)

Radiotherapy

385
Q

What is the incidence of adrenal cortical carcinoma?

A

In the UK, around 220 people are diagnosed with adrenal gland cancer every year (ACC and phaeochromocytoma). ACC found in 1-2/million people (1).

Commonly detected in fifth/sixth decades of life, but also a secondary peak in children <10 years

386
Q

What are the risk factors of adrenal cortical carcinoma?

A

Smoking
Oral contraceptives
Germline predisposition (10%): LFS, lynch, FAP, MEN1

387
Q

How does adrenal cortical carcinoma present?

A

Patients can present with Cushing’s syndrome- symptoms can include central obesity, muscle weakness, easy bruising, fatigue, and depression.

Generic symptoms relating to presence of a mass- abdo pain, tiredness, weight loss, nausea, and vomiting.

Female patients can be affected by increased androgens: excess facial and body hair (hirsutism), baldness, acne, deepening of the voice, increased muscularity, and an increased sex drive

Metastatic disease 40-60%

388
Q

What molecular pathways are affected in adrenal cortical carcinoma?

A

p53/RB
- TP53 (most common)
- RB1
- MDM2

Wnt pathway
- APC
- CTNNB1
- MEN1

CNVs
- CDK4 gain
- RB1 loss

389
Q

What TD targets are there for adrenal cortical carcinoma?

A

TP53 SNVs and CNVs

NTRK

390
Q

How is adrenal cortical carcinoma diagnosed after biopsy?

A

IHC
- Ki67- prognostic biomarker.
- SF1- diagnostic
- BUB1B and PINK1 strong poor prognosis predictors

391
Q

How are adrenal cortical carcinoma classified into risk groups?

A

Low risk: no mutation in TP53 and Wnt pathways, do not display whole-genome doubling.

High risk: frequent TP53 and Wnt pathway gene mutations, numerous non-recurrent alterations with potentially whole-genome doubling.

392
Q

How is adrenal cortical carcinoma monitored?

A

Currently CT/MRI

Evidence supporting the use of ctDNA
- high level of detectable mutations in ctDNA

393
Q

How is adrenal cortical carcinoma treated?

A

Stage I-III
Complete surgical resection

Stage IV
- Systemic treatments
- Mitotane- acts specifically in the adrenal cortex, disrupts mitochondrial membranes and cytochrome enzymes leading to cell death
- Anti-VEGF agents disrupt tumour angiogenesis.
- IGF-I inhibitors
- CDK4/6 inhibitors- targets CDK4 gains
- Very limited evidence to suggest targeted therapies are affective

394
Q

What is the prognosis of adrenal cortical carcinoma?

A

The 5-year overall survival rate is 60–80% for tumours confined to the adrenal space

35–50% for locally advanced disease37-47%

Potentially curable disease if tumour is completely excised with no angioinvasion.

Stage IV ACC unlikely to survive>5 years.

395
Q

What is the incidence of head and neck squamous cell carcinoma (HNSCC)?

A

7th most common cancer globally, accounting for an estimated 890,000 new cases

Roughly 4.5% of all cancer diagnoses around the world)

450,000 deaths per year (roughly 4.6% of global cancer deaths).

396
Q

What are the risk factors of HNSCC?

A

Smoking (leading risk factor)
Alcohol
HPV- cause of 72% cases
EBV
Areca nut

Genetic: fanconi aneamia

397
Q

How does HNSCC present?

A

Most present with advanced HNSCC

small proportion present with pre mlaignant lesions

Some symptoms: non-healing ulcer, palpable mass, issues speaking/eating et

398
Q

What TD targets are there for HNSCC?

A

SNVs: CDKN2A, EGFR, TP53

CNVs: TP53, CDKN2A

Structural variants: RET, NTRK

399
Q

How is HNSCC diagnosed?

A

Biopsy/ FNA
- Histology to determine squamous differentiation

IHC
- HPV status (use p16 as a marker)
- Squamous markers: CK5, CK6, p63

Genetics
- TP53, CDKN2A

400
Q

How is HNSCC treated?

A

Local resection

Systemic therapy:
- Chemotherapy (cisplatin) +cetuximab
- Pembrolizumab/nivolumab- liscensed for use with >1% anti-PD-L1 IHC companion diagnostic

Other trials:
- HRAS inhibitor tipifarnib

401
Q

What is the prognosis of HNSCC?

A

Poor, median OS 10-15 months

402
Q

What are the types of bladder cancer?

A

Urothelial carcinoma (80-90%)
Squamous cell carcinoma (1-2%)
Adenocarcinoma (1%)

403
Q

What is urothelial carcinoma?

A

Formally known as transitional cell carcinoma

Most common form of bladder cancer and starts in the urothelial cells in the bladder walls inner layer

404
Q

What is the incidence of bladder cancer?

A

10,300 new bladder cancer cases in the UK every year, and is the 11th most common cancer in the UK.

More common in males and most common >85 years

405
Q

What are the risk factors for bladder cancer?

A

Smoking
Chronic irritations
Chronic inflamation

406
Q

How does bladder cancer present?

A

Often asumptomatic

Haematuria (blood in urine)
Dysuria
Increased frequency, urgency

407
Q

How is bladder cancer classified?

A

NMIBC and MIBC. MIBC has a high mutational burden

Luminal
- Better prognosis
- FGFR2/3 mutations

Basal
- Poor prognosis
- TP53 mutations and DNA repair pathways
- Respond to better platinum based chemotherapy

408
Q

How is bladder cancer diagnosed?

A

Cystoscopy
CT scan/MRI

409
Q

What is the prognosis of bladder cancer?

A

70% NMIBC
- 60-70% reoccur

25% MIBC
- 5 year survival is 78%
- median survival 18 months

410
Q

How is bladder cancer treated?

A

Low stage
- Transurethral resection of the bladder (TUR-BT)
- Intravesival BCG
- Chemotherapy

High stage
- Radical cystectomy
- Radiation therapy

Erdafitinib- FGFR inhibitors in NICE development currently

411
Q

What is mesothelioma?

A

Malignant neoplasm of mesothelial differentiation that arises from mesothelial lining cells of the pleura

412
Q

What is the incidence of mesothelioma?

A

Up to 30 cases/million people/year

An estimated 28,000 - 43,000 people die from malignant pleural mesothelioma worldwide every year.

413
Q

What are the risk factors of mesothelioma?

A

Strong association with asbestos exposure- latency period of 20 to > 40 years

Ionizing radiation for treatment of malignancy (Hodgkin lymphoma, non-Hodgkin lymphoma, testicular cancer)

414
Q

How does mesothelioma present?

A

Parietal, visceral, mediastinal or diaphragmatic pleura

Shortness of breath, Chest wall pain, pleurisy, Cough, Weight loss,

Occasionally asymptomatic when discovered at early stage.

415
Q

What molecular markers are used to distinguish reactive and malignant mesothelioma?

A

Loss of BAP1

Loss of MTAP

Homozygous deletion of CDKN2A

416
Q

What TD targets are there for mesothelioma?

A

CDKN2A CN loss (NGS and FISH)

NTRK

417
Q

How is mesothelioma diagnosed?

A

Pleural thickening/ pleural effusion on X ray

IHC
- Loss of MTAP and BAP1
- Homozygous deletion of CDKN2A
- When used in combination, 84% sensitive, 100% specific for mesothelioma diagnosis

418
Q

How is mesothelioma treated?

A

Most common treatment is chemotherapy alone

Also surgery and radiation therapy

419
Q

What subtype of mesothelioma has the best prognosis?

A

Epithelioid subtype (best prognosis) > biphasic > sarcomatoid / desmoplastic subtype (worst prognosis)

420
Q

What is mucoepidermoid carcinoma?

A

MEC is the most common salivary gland malignancy in children and adults, and the most common malignancy of the major and minor salivary glands.

421
Q

What is the incidence of mucoepidermoid carcinoma?

A

2.5 to 3.0 cases per 100,000 individuals a year, wide age range from childhood to elderly

422
Q

What are the risk factors for mucoepidermoid carcinoma?

A

Possible association with HPV and ionizing radiation.

423
Q

What is the clinical presentation of mucoepidermoid carcinoma?

A

33% of patients are asymptomatic
Firm or fixed tumour, occasionally largely cystic.

424
Q

What molecular abnormalities are there in mucoepidermoid carcinoma?

A

CRTC1::MAML2 t(11;19)(q21;p13)-80% of MECs

Also CRTC3:MAML2

425
Q

What TD targets are there for mucoepidermoid carcinoma?

A

MAML2 rearrangements by FISH or NGS
NTRK

426
Q

How is mucoepidermoid carcinoma diagnosed?

A

CT, MRI

IHC
- Positive: Pancytokeratin, CK5/6, p63, p40,
- Negative: S100, HER2, DOG1

427
Q

What is the prognosis of mucoepidermoid carcinoma?

A

Excellent prognosis, with approximately 98.8% 5 year survival rate in low grade and 97.4% in intermediate grade tumors. About 67% 5 year survival rate for high grade tumors

428
Q

How are mucoepidermoid carcinoma treated?

A

Conservative excision with preservation of the facial nerve- low and intermediate-grade

Radical neck dissection is performed in patients with cervical node metastasis and T3 lesion.

Adjuvant radiotherapy and chemotherapy might be considered for higher grade tumors

429
Q

What is the incidence of pancreatic cancer?

A

Approximately 10,500 new cases of pancreatic cancer each year in the UK (statistics from (1))

Tenth most common cancer in the UK.

430
Q

What is the prognosis of pancreatic cancer?

A

21% one-year survival, 3% five-year survival, 1% ten-year survival

Fifth commonest cause of cancer death in the UK and accounts for 6% of all cancer deaths, but pancreatic cancer incidence in on the rise.

431
Q

What are the types of pancreatic cancer?

A

95% exocrine tumours

Most commonly adenocarcinoma

5% are pancreatic neuroendocrine neoplasms

432
Q

How does pancreatic cancer present?

A

Non-specific symptoms leading to late stage disease

Symptoms:
- Abdominal and back pain
- Unexplained weight loss
- Jaundice and itchy skin
- Indigestion
- Loss of appetite
- Change in bowel habits and fatty stools
- Bloating and feeling full quickly
- Tiredness

433
Q

How is pancreatic cancer diagnosed?

A

Blood test
- CA19-9 established biomarker for pancreatic ductal adenocarcinoma

Imaging
- CT
- Endoscopic ultrasound
- MRI

Biliary brushing for cytology

Resection of pancreatic cysts

434
Q

When do patients undergo surveillance for pancreatic cancer?

A

hereditary pancreatitis and a PRSS1 mutation

BRCA1, BRCA2, PALB2 or CDKN2A (p16) mutations, and one or more first-degree relatives with pancreatic cancer

Peutz–Jeghers syndrome.

Lynch syndrome

435
Q

What genes are commonly mutated in somatic pancreatic cancer?

A

KRAS, TP53, CDKN2A, GNAS, SMARCB1, SMAD4 and RB1

436
Q

What syndromes are associated with familial pancreatic cancer?

A

10% of pancreatic cancer is hereditary

HBOC (breast and ovarian)- 7%
- BRCA1/2

FAMMM (Familial atypical multiple mole melanoma)
- CDKN2A

PJS (Peutz-Jeghers syndrome)
- STK11

LFS (Li-Fraumeni syndrome)
- TP53

FAP (Familial adenomatous polyposis)
- APC

Hereditary pancreatitis
- PRSS1 and SPINK1

Lynch syndrome
- MLH1, MSH2, MSH6, PMS2

437
Q

How is inherited pancreatic cancer tested for according to the test directory?

A

NGS +/MLPA

BRCA1
BRCA2
CDKN2A
PALB2
CDK4
MLH1
MSH2
MSH6
PMS2
PRSS1
RABL3
STK11

438
Q

What TD targets are there for somatic pancreatic cancer?

A

BRCA1/2 - PARP eligibility

NTRK- NTRK inhibitors

MSI- lynch pathway

439
Q

is HRD testing available for pancreatic cancer?

A

No

But evidence shows that pancreatic has high HRD and that these may respond to PARP inhibitors

These tumours also respond well to platinum based chemotherapy

440
Q

How is pancreatic cancer treated?

A

First line
- FOLFIRINOX

Second line
- oxaliplatin-based chemotherapy

Resection only curative option but more than 50% are not eligible

PARP inhibitors if BRCA variant

Immunotherapy if MSI/MMR deficient

441
Q

Is pancreatic cancer screened for?

A

There is no NHS national screening program due to the rarity of pancreatic cancer, the costs of screening and the fact that screening isn’t currently reliable and accurate enough.

High risk (family history) received feedback. Screening begins around age 40 and involves CT scans or endoluminal ultrasound test, and sampling of pancreatic juices every three years. This pancreatic juice is subjected to mutational analysis for KRAS, TP53 and CDKN2A variants. If variants are discovered, screening frequency is increased to annual screening.

442
Q

What are Phaeochromocytomas and paragangliomas (PPGL’s)?

A

Rare neuroendocrine neoplasms

Phaeochromocytomas originate from chromaffin cells of the adrenal medulla.

Paragangliomas occur in the paraganglia outside the adrenal medulla and can migrate to the head, neck and pelvis

443
Q

What are the genetic types of Phaeochromocytomas according to the cancer gnome atlas?

A

1: Mutations of genes that affect hypoxic signalling, usually germline affecting VHL (15-20% of PPGL’s) and TCA-cycle (SDHx genes, 10-15% PPGL’s).

  1. Tyrosine kinase cluster’- mutations of RET, NF1, HRAS, TMEM127. These mutations are found in 50-60% of PCC/PPGL’s and can be germline or somatic mutations. Lower risk of mets
  2. Somatic mutations in the WNT-altered pathway (MAML3 oncogene fusions and CSDE1 mutations). Likely aggressive. Found in 5-10% of PCC/PPGL’s.
444
Q

What genetic predispositions are there to PPGL?

A

Von Hippel-Lindau (VHL), syndrome, MEN2, NF1, FH - 1/3 casees

Familial PPGL, SDHA-related autosomal-dominant syndromes

445
Q

What is the incidence of PPGL?

A

Ranges from 0.4-9.5 per million per year

50-60 years age

10-20% occur in children

446
Q

What genes are associated with hereditary PPGL?

A

RET, NF1, VHL, TMEM127, SDHA, SDHB, SDHC, SDHD, SDHAF2, FH,

447
Q

How does PGAL present?

A

Many present with few symptoms or can be asymptomatic (25% cases) but mostly present with catecholamine excess

Hypertension

Other symptoms may include abdominal pain, constipation, weight loss, tremor, facial pallor, panic attacks, fatigue and seizures

448
Q

What TD targets are there for somatic PPGL?

A

SNVs RET

NTRK

449
Q

How is PPGAL diagnosed according to the WHO/

A

Essential: Neuroendocrine neoplasm arising from the adrenal medulla with consistent morphology

Desirable:

CT/MRI

IHC:
+ tyrsoine hydroxylase, GATA3, synaptophysin
-ve for S200, SOX10
SDHx to guide subsequent genetic testing

Routine assessment of cell proliferation (mitotic count, Ki67/MIB-1 labelling index); Mitoses are often very rare or absent

Measurement of all 3 metabolites of parent catecholamines (including 3-methoxytyramine along with metanephrine and normetanephrine levels.

450
Q

How are patients with PPGL montiored?

A

ESE guidelines recommend monitoring for 10 years. High-risk patients (young patients and those with a genetic disease, a large tumour and/or a paraganglioma) should be monitored for life

Biochemical tests yearly including chromogranin-A

Imaging every 1-2 years

451
Q

How are are PPGL treated?

A

Metanephrine blockade (alpha/beta blockers), phenoxybenzamine

Resection

cyclophosphamide, vincristine and dacarbazine (CVD) chemotherapy, the most widely used regimen

Clinical trials using olaparib with temozolomide

452
Q

What is the prognosis of PPGL?

A

Risk of metastasis ranges from ~5 to 15% . 5yr survival rate for adults was 40-95%, and 98% in children (

Germline SDHB mutations confer the highest risk of metastasis, 5yr survival rate drop s to 36-92% for adults and 76% for children

453
Q

What is thymic cancer?

A

Thymic cancer is a rare epithelial type of cancer arises from the thymus gland.

Thymic epithelial tumours represent a heterogeneous group of rare malignancies that include:

  • thymomas
    *thymic carcinoma (account for ~20% of all thymus tumours)
    *thymic neuroendocrine tumours
454
Q

What is the difference between thymomas and thymic carcinomas?

A

Thymic carcinomas are much less common than thymomas, accounting for 14–22% of all thymic epithelial tumours.​

They are more aggressive in terms of recurrence and metastasising than thymomas, and are more resistant to chemotherapy, they are not usually detected until it has become invasive.​

455
Q

What is the incidence of thymic carcinoma?

A

Annual incidence ranging from 1.3 to 3.2 per million.

Mean age at diagnosis is 50–60 years of age

456
Q

What are the risk factors for thymic carcinoma?

A

No environmental or infectious factors have been demonstrated to play a role in the pathogenesis of thymic epithelial tumours.

Genetic risk factors, such as multiple endocrine neoplasia 1 (MEN1), may influence the development of thymomas (but this is less common in thymic carcinoma)

457
Q

How does thymic carcinoma present?

A

1/3 are asymptomatic

*Coughing
* Chest pain
* Shortness of breath
* Difficulty swallowing
* Fatigue
* Weight loss

458
Q

How is thymic carcinoma diagnosed?

A

CT/MRI

IHC
- CD117/KIT and CD5+ determine thymic origin
- EGFR/HER2 overexpression

459
Q

What molecular abnormalities are associated with thymic carcinoma?

A

Poorly understood

Thymic squamous cell carcinoma
- Loss of chromosome 16p

Translocations:
Thymic mucoepidermoid carcinoma (with MAML2 gene fusion)​

NUT carcinoma (with NUTM1 gene fusion)​

A subtype of clear cell carcinoma (hyalinizing clear cell carcinoma, with EWSR1 gene fusion)​

SNVs: TP53, CDKN2A, KIT and PTEN.​

460
Q

What TD targets are there for thymic carcinoma and why?

A

KIT
- Common mutations are L576P, D820E
- Predicts repsonse to imatinib

461
Q

How is thymic carcinoma treated?

A

Chemotherapy: ​
- better response for thymomas than carcinomas
- Cisplatin based regimens

Immunotherapy:
- ICIs; normally high PD-L1 expression
- clinical trial of pembrolizumab.​

Other options:
- Imatinib- KIT mutated
- Sunitinib is an option as 2nd line treatment (independent from KIT status)​
- Everolimus (mTOR inhibitor) may represent an option for refractory tumours​

462
Q

How often do thymic carcinomas reoccur?

A

Recurrences of thymic epithelial tumours are not uncommon (∼10%–15% of all-stage resected tumours

463
Q

What is CUP?

A

Cancer of unknown primary

CUP is defined as metastatic cancer in the absence of a clinically detectable anatomically defined primary tumour.

Often challenging because it tends to be aggressive and has often spread to many parts of the body before it is found (often lymph nodes, liver, lung, peritoneum (lining of the bowel), or bone)

464
Q

What is the incidence of CUP?

A

3-5% of all newly diagnosed cancers

1 year survival 10-20%

465
Q

What are the risk factors of CUP?

A

Smokers are at risk of developing CUP and this risk correlates with the level of tobacco exposure

Type 2 diabetes

autoimmune disorders (Human papillomavirus (HPV)

High body mass index, waist circumference, low socioeconomic status and black ethnic background may be additional risk factors

466
Q

How does CUP present?

A

Depends on which organs spread to. Can include:

-Swollen, firm, non-tender lymph nodes
- A mass in the abdomen
-Shortness of breath
-Bone pain

467
Q

What TD targets are there for CUP?

A

NTRK

WGS- when standard of care is exhausted

468
Q

What diagnostic strategies are used to aid diagnosis of CUP?

A

Morphological pattern- differentiate between epithelial, round, spindle-shaped and anaplastic cancers to identify the pattern of tissue organisation regarding entity and tissue of origin.

Immunohistochemistry (IHC)- markers to help determine tissue lineage

MSI testing

CT/MRI- locate primary tumour

Genetics
- Structural can aid diagnosis
- gene expression profiles

469
Q

What are common diagnostic difficulties in CUP?

A

Two common diagnostic difficulties in CUP work-up are poorly differentiated or undifferentiated cancer and better differentiated carcinoma, especially adenocarcinoma, without an obvious primary site.

Can’t offer a molecular panel if do not know the primary

470
Q

What targeted treatments can be used for CUP?

A

NTRK inhibitiors

MSI-H/TMB- Pembrolizumab

If no druggable target, treated with empiric therapy

471
Q

What is immunotherapy?

A

Immunotherapy is the utilisation of cellular immune responses to target cancer cells and ultimately trigger apoptosis of these cells

472
Q

What is the tumor microenvironment?

A

Tumour microenvironment consists of the environment surrounding cancer cells and the crosstalk and interplay between the tumour and surrounding cells. These surrounding cells can be cells of the innate and adaptive immune system as well as local cells of the affected organ.

This is important for the use of immunotherapies as the presence of immune cells is required

473
Q

What factors in the tumor microenvironment result in immune suppression?

A

1.Stroma
2. Carcinoma-Associated Fibroblasts (CAFs)
3. Tumour-Associated Macrophages (TAM)
4. Cytokines

474
Q

How does the stroma of the TME promote immune suppression?

A

The stroma comprises various non-cancerous cells, extracellular matrix (ECM), and signalling molecules surrounding the tumour cells. It provides structural support to the tumour. Increased collagen can create physical barriers that impede immune cell infiltration and drug delivery

475
Q

How does the Carcinoma-Associated Fibroblasts (CAFs) of the TME promote immune suppression?

A

They secrete growth factors, cytokines, and ECM components that support cancer cell survival, proliferation, and invasion

476
Q

What is the role of Tumour-Associated Macrophages (TAMs) in the TME?

A

Can exhibit pro-tumour or anti-tumour functions depending on their activation state.

In the TME, TAMs often adopt an immunosuppressive phenotype, promoting tumour growth and metastasis. They secrete cytokines, such as TGF-β and IL-10, which suppress the activity of cytotoxic T cells and natural killer (NK) cells, thereby facilitating immune evasion by cancer cells.

477
Q

What is the role of cytokines in the TME?

A

Certain cytokines, such as TGF-β, IL-10, and IL-6, have immunosuppressive properties and can inhibit the activity of immune cells within the TME.
Cytokines like vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) promote angiogenesis, facilitating the formation of new blood vessels to supply nutrients and oxygen to the growing tumour.

478
Q

What is meant by a hot tumour microenvironment?

A

There is a significant infiltration of immune cells, particularly cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells.
These immune cells are actively engaged in recognizing and attacking cancer cells, contributing to an anti-tumour immune response.
These tumours are more repsonsive to immunotherapies.

However, even in hot TMEs, there may be immunosuppressive elements like regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs)

479
Q

What is meant by a cold tumour microenvironment?

A

There is a limited infiltration of immune cells, particularly cytotoxic T lymphocytes and NK cells.
Low levels of inflammatory cytokines and chemokines.
Tumours often resistant to immunotherapies

480
Q

As well as hot/cold TME, what other barriers are there to immunotherapies?

A

Tumour Heterogeneity:
varying molecular characteristics which respond differently to immunotherapy

Resistance Mechanisms: upregulation of alternative immune checkpoints, alterations in antigen presentation, and recruitment of immunosuppressive cells.

Physical Barriers: The dense extracellular matrix (ECM) and abnormal tumour vasculature within the TME can physically impede the infiltration of immune cells and the delivery of immunotherapeutic agents to the tumour site.

481
Q

What are immune checkpoints?

A

Programmed cell death 1 (PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) are co-inhibitory receptors expressed on the surface of T cells to negatively regulate T cell-mediated immune responses. These receptors bind ligands expressed by host cells

This is normal function of the immune system to prevent an inappropriately excessive immune response which may be harmful

cancer cells can use checkpoint inhibitor proteins to inhibit immune surveillance

482
Q

What are immune checkpoint inhibitiors?

A

Drugs to attach to the co-inhibitory receptors on T cells/tumour cells, blocking the immune inhibition, thereby reactivating the immune response against tumour cells.

Examples are anti-CTLA-4, anti-PD-1, and anti-PD-L1

483
Q

What approved anti-CTLA-4 checkpoint inhibitors are there?

A

Ipilimumab
- Melanoma, RCC, colorectal, NSCLC etc

CTLA-4 is constitutively expressed in regulatory T cells

484
Q

What approved anti-PD-1 checkpoint inhibitors are there?

A

Pembrolizumab
- approved in 2014
- Melanoma, NSCLC, RCC, hodgkins lymphoma, endometrial

Nivolumab
- approved in 2014
- Melanoma, NSCLC, RCC, hodgkins lymphoma, colorectal etc

485
Q

What anti PD-L1 checkpoint inhibitors are there?

A

Atezolizumab
- Bladder, NSCLC, breast

486
Q

What are neo-antigens?

A

Cancer antigens produced as a result of mutations and presented by major histocompatibility complex (MHC) proteins on the surface of cancer cells

This targets those cells for destruction by the immune system

487
Q

What biomarkers are there for repsonse to immunotherapy?

A

PD-L1 IHC

TMB

MMR/MSI

488
Q

How does TMB help predict reponse to immunotherapy?

A

Once immune checkpoints are blocked and T cells are able to target cancer cells, they still need to different the tumour cells from normal cells. The more neo-antigens present the easier this is

The higher number of mutations present (both driver and passenger), the more likely mutant proteins will be expressed at the cell surface and are therefore more likely to repsond to immunotherapy

Tumour mutational burden is a measure of the number of mutations present in a cell and correlates to response

489
Q

What is TMB?

A

TMB is expressed as the number of acquired mutations per megabase (mut/Mb) of sequenced DNA commonly assessed by next generation sequencing (NGS) in the tumour genome.

490
Q

How is TMB measured?

A

Gold standard TMB measurement is derived from WGS/WES- tissue availablity and long turnaround times means this is using routinely

Smaller targeted panels that are already in widespread use have generally been used to measure TMB including, TSO500 from Illumina (1.33Mb, 523 genes) and Foundation One CDx from Foundation Medicine (0.8Mb, 324 genes),

the latter which is FDA approved as a companion diagnostic for the ICI drug pembrolizumab (if (≥10 mut/Mb)

491
Q

What factor impact TMB measurement?

A

Sample issues
- Low TC will underestimate TMB
- Low quality of sample, particularly from FFPE samples may cause an overestimation of TMB

Analytical
- Using small panels reduces confidence in score
- High variability in BI processes- what types of variants are included

492
Q

What are some limitations of TMB as a biomarker for ICI response?

A

TMB values vary widely between tumour types so it is difficult to set a cut off that suits all- likely each tumour type needs different

TMB is not a sole predictor neo-antigenicity : The recognition of an antigen as a foreign entity will differ depending on the mutation profile present- indels and structural variants more likely than SNVs

Doesn’t consider TME- are there a high number of activated T cells (Hot TME)

493
Q

How does MMRd predict immunotherapy response?

A

Inactivation of this complex is associated with reduced DNA replication fidelity, leading to microsatellite instability (MSI) and the mutator phenotype often seen in colorectal and other cancers.

Again likley to result in neo-antigens (correlates but is not an independent predictor as dependent upon mutation profile)

494
Q

How can MMRd be measured for immunotherapy response?

A

IHC
- testing for loss of expression of the key mismatch repair proteins MLH1, MSH2, MSH6 and PMS2

MSI
- fragment analysis of microsatellite markers

495
Q

What is the primary objective of cancer vaccination?

A

To activate the immune system to recognize and combat cancer cells, prevent tumour growth, recurrence, or metastasis, and enhance the immune system’s capacity to eliminate cancer cells

It involves eliciting an immune response by targeting specific tumour-associated antigens (TAAs), which are proteins expressed by cancer cells. A further aim of cancer vaccines is to induce a memory response, allowing for a more effective immune response upon subsequent encounters with tumour cells.

496
Q

What is the difference between prophylactic and therapeutic cancer vaccines?

A

Prophylactic vaccines prevent cancer in high-risk populations, while therapeutic vaccines treat individuals already diagnosed with cancer

497
Q

How do cell-based cancer vaccines work?

A

They use immune cells, such as dendritic cells, to stimulate an immune response against cancer cells by collecting from a patients blood or tumour, activating, and expanding these cells in a lab and reintroducing them into the patient’s body

There are two approaches: Autologous vaccines utilize the patient’s own tumour cells, ensuring antigen compatibility but at the expense of increased costs and time requirements. Allogeneic vaccines, while lacking personalization, present a time-saving advantage.

498
Q

How do dendritic cell vaccines function?

A

Dendritic Cell Vaccines are collected from the patient’s blood or generated in the laboratory. They are then matured and activated using immune-stimulating molecules or tumour specific antigens (TSAS). The loaded DCs are administered back to the patient. These DCs migrate to lymphoid organs, where they interact with immune cells, such as T cells, B cells, and NK) cells.

The DCs present the tumour antigens to CD4+ helper T cells and CD8+ cytotoxic T lymphocytes (CTLs), leading to their activation. The activated T cells provide help signals to other immune cells, enhancing the immune response against tumour cell, engaging both the innate and adaptive immunities and leading to the development of immunological memory in the case of tumour relapse.

499
Q

How do whole cell vaccines function?

A

Cancer cells are collected from the patient’s tumour or established cancer cell lines. These cells are inactivated or genetically modified to reduce their ability to grow and cause disease. When administered back to the patient, the whole cells are recognized by various immune cells, including DCs, macrophages, and NK cells, triggering an immediate non-specific inflammatory response.

The activated immune cells, in turn, process the tumour antigens, present them to T cells, and initiate an immune response. CD4+ helper T cells provide help signals to other immune cells, while CD8+ CTLs recognize and eliminate tumour cells expressing the presented antigens. Whole-cell cancer vaccines also aim to induce a memory response for enhanced immune protection against tumour recurrence.

500
Q

What is the historical significance of Bacillus of Calmette-Guérin (BCG) in cancer vaccines?

A

BCG became the first approved whole-cell vaccine for cancer therapy in 1990, specifically for bladder cancer, and was approved by the FDA in 1998

501
Q

Describe peptide cancer vaccines

A

They involve the administration of peptides derived from TAAs, which are taken up by antigen-presenting cells (APCs) and presented to T cells, leading to an immune response

502
Q

How do DNA and RNA cancer vaccines work?

A

These vaccines deliver DNA or RNA encoding TAAs, which are taken up by cells to produce the antigens, then processed and presented by APCs to activate T cells.

503
Q

What are induced pluripotent stem cell (iPSC)-based cancer vaccines?

A

iPSCs are differentiated into tumour microenvironment-specific cells expressing antigens, which activate immune cells and initiate a robust immune response upon administration.

504
Q

How do viral-based cancer vaccines activate the immune response?

A

The modified virus interacts with immune cells, including DCs, macrophages, and NK cells, triggering an inflammatory response and the release of pro-inflammatory cytokines and chemokines. The virus particles are phagocytosed by immune cells, and the TAAs expressed by the virus or delivered to infected cells are processed and presented to T cells. CD8+ CTLs recognize the presented TAAs, leading to their activation and expansion.

505
Q

What is the role of memory response in cancer vaccines?

A

To enable a more effective immune response upon subsequent encounters with tumour cells

506
Q

What are oncolytic viruses?

A

Viruses used as a vector to target and kill cancer cells, including natural and genetically modified viruses that reproduce in cancer cells without harming healthy cells

Their ability to specifically bind and infect cells, replicate using cellular machinery, and lyse host cells, releasing new viruses to infect more cells

507
Q

How can oncolytic viruses alter the tumour microenvironment (TME)?

A

By provoking an inflammatory response and triggering an immune response against cancer cells, potentially turning the TME from a ‘cold zone’ to a ‘hot zone’.

When the cancer cells are lysed, they release tumour-associated antigens (TAAs), cytokines, and other molecules that promote an adaptive immune response and activate various immune cells.

508
Q

What is an advantage of virus vaccines over monoclonal antibodies?

A

Can pass the blood brain barrier

509
Q

How can viruses be used to deliver therapeutics directly to cancer cells?

A
  • ‘suicide gene’: Suicide gene therapy involves delivery of a gene which codes for a cytotoxic product or a gene which has the ability to correct mutated proapoptotic genes, inducing apoptosis
  • Cytokines: e.g. TGF-β are known to play a key role in the TME of tumour cells, and targeted delivery may alter the balance against tumour cells. Can be toxic
  • Genes that inhibit angiogenesis resulting in oxygen starvation in the tumour.
  • Sodium-iodide symporter (NIS) gene to the viral genome. When combined with radioiodine therapy it allows local radiotherapy of the tumour, as used to treat thyroid cancer.
510
Q

What is a major obstacle for oncolytic virus therapy?

A

The patient’s immune system, which tries to deactivate the virus, especially after intravenous injection.

It has been shown that immunosuppression by chemotherapy and inhibition of the complement system can enhance oncolytic virus therapy.

511
Q

What are some strategies to avoid immune system detection of oncolytic viruses?

A

Using non-common human pathogens, coating the virus with polymers, or hiding the virus inside macrophages.

512
Q

What was the first oncolytic virus approved by the FDA and for what condition?

A

T-VEC, a genetically engineered oncolytic herpes simplex virus type 1, approved in 2015 for treating metastatic melanoma.

513
Q

What are monoclonal antibodies?

A

Monoclonal antibodies are identical clones of an antibody used to target a specific antigen epitope. Their use as a cancer therapeutic is in selectively identifying a specific cell as a target rather than a broad response which may affect all cells

514
Q

How do ligand/receptor blockade monoclonal antibodies work?

A

Some cancer types are characterised by overexpression/inappropriate receptor signalling pathway activation which may cause uncontrolled growth. Use of monoclonal antibodies against these receptors or their ligands can bind and block these signalling pathways fuelling tumour growth.

An example of receptor blocking is HER2 positive breast cancer which can be treated with Herceptin (trastuzumab).

515
Q

How do ADCC (antibody-dependent cell cytotoxicity) monoclocal antibodies work?

A

The specific antigen binding fragment (Fab) of the monoclonal antibody identifies the region of interest, while the Fc domain binds NK cells which perforate and kill the target cell.

An example of this is Rituximab, an anti CD-20 monoclonal antibody which is used in the treatment of CD-20 positive leukaemias.

CD-20 is abundantly expressed on cell surface of mature B-NHL cancer cells while immature naïve B cells do not, theoretically allowing the maturation of normal functional B cells while targeting malignant B cells.

516
Q

How do Antibody–drug conjugates (ADCs) monoclocal antibodies work?

A

Mabs bearing cytotoxic drugs covalently bound via a chemical linker and can be defined as prodrugs

Serves as targeted delivery system to the tumour expressing the antigen/target recognized by the antibody

517
Q

What are CAR-T cells?

A

CAR-T cells are engineered synthetic T cell receptors that redirect lymphocytes to recognize and eliminate cells expressing a specific target antigen, independent from the MHC receptor.

518
Q

What are the four main components of a Chimeric antigen receptors (CAR)?

A

The four main components of a CAR are:

Extracellular target antigen-binding domain.
Hinge region.
Transmembrane domain.
Intracellular signalling domains.

519
Q

What is the function of the antigen-binding domain in CAR-T cells?

A

The antigen-binding domain confers target antigen specificity and is derived from the variable heavy (VH) and light (VL) chains of monoclonal antibodies, forming a single-chain variable fragment (scFv) that targets extracellular surface cancer antigens.

520
Q

Describe the role of the hinge region in CAR-T cells.

A

The hinge region extends the binding units from the transmembrane domain, providing flexibility to overcome steric hindrance and allowing the antigen-binding domain to access the targeted epitope.

521
Q

What is the transmembrane domain in CAR-T cells responsible for?

A

The transmembrane domain anchors the CAR to the T cell membrane and is usually derived from natural proteins like CD3ζ, CD4, CD8α, or CD28.

522
Q

Explain the function of the intracellular signalling domains in CAR-T cells.

A

Intracellular signalling domains transmit activation signals. They determine the generation of the CAR, classified into first (CD3z only), second (one costimulatory domain + CD3z), or third generation (multiple costimulatory domains + CD3z).

523
Q

What are the main challenges in CAR-T cell therapy?

A

The main challenges include:

Antigen escape.
On-target off-tumour effects.
CAR-T cell trafficking and tumour infiltration.
CAR-T cell-associated toxicities.
CAR immunogenicity.

524
Q

What is antigen escape and how can it be countered in CAR-T cell therapy?

A

Antigen escape is the development of tumour resistance by the tumour downregulating the target antigen, preventing recognition by the T cells. Cancer cells may also show hetergoenity where not all cells express the same antigen, preventing all cells being targeted

It can be countered by targeting multiple antigens using either dual CAR constructs or tandem CARs.

525
Q

What are on-target off-tumour effects?

A

On-target off-tumour effects occur when CAR-T cells target antigens expressed on both tumour and normal tissues, leading to toxicity. This can be mitigated by targeting tumour-restricted post-translational modifications.

526
Q

How can CAR-T cell tumour infiltration be improved?

A

In solid tumours, it can be challenging for the CAR-T cells to access the tumour cells due to physical barriers e.g. stroma or the TME. Strategies to overcome this include:

Local delivery of CAR-T cells.
Expressing chemokine receptors on CAR-T cells.
Removing physical barriers like tumour stroma.
Combating the immunosuppressive tumour microenvironment.

527
Q

What is cytokine-release syndrome (CRS) and how is it managed?

A

CRS is a result of excessive cytokine release by activated CAR-T cells, managed primarily with IL-6 receptor blockade using tocilizumab and corticosteroids.

528
Q

What are some strategies to overcome the immunosuppressive tumour microenvironment?

A

Strategies include combination immunotherapy, armored CARs, and engineering CAR-T cells to secrete pro-inflammatory cytokines or express cytokine receptors.

529
Q

What is immune effector cell-associated neurotoxicity syndrome (ICANS)?

A

ICANS is characterized by elevated cerebrospinal fluid cytokine levels and blood-brain barrier disruption, managed with corticosteroids

530
Q

How can CAR-T cell-associated toxicities be ameliorated?

A

Toxicities can be ameliorated by altering CAR structure, reducing antigen binding domain specificity, modifying hinge and transmembrane regions, and using different costimulatory domains

531
Q

What are engineered T cell receptor (TCR)-T cells?

A

TCR-T cells are engineered T cells with exogenous T cell receptors directed towards specific tumour antigens, capable of targeting peptides from both intracellular and extracellular proteins

532
Q

What are the advantages and disadvantages of engineered TCR-T cells over CAR-T cells?

A

Advantages include less cytokine-induced toxicity and the ability to target intracellular and extracellular proteins presented by HLA molecules

Disadvantages include the requirement for intact antigen processing and presentation machinery and potential suppression by immunomodulatory factors within the tumour microenvironment.