Tumour biology Flashcards

You may prefer our related Brainscape-certified flashcards:
1
Q

Name the purines and pyrimidines in DNA and who they pair with

DEFINITELY IN CURRICULUM

A

Purines : adenine, guanine
Pyrimidine: cytosine, thymine

A = T (2 hydrogen bonds)
G 三 C (3 hydrogen bonds)

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

How is DNA packaged?

DEFINITELY IN CURRICULUM

A

DNA is an acid with highly negative charge. Double-helix - 2 anti-parallel strands – polymers of nucleotides
DNA helix wraps around nucleosomes consisting of histones (highly +ve charge) -> forms chromatin fibre -> coiled and packaged into chromosomes

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

Name the bases in RNA? What is the difference between RNA and DNA?

DEFINITELY IN CURRICULUM

A
  • Single stranded, shorter, U instead of T

Purines: adenine, guanine
Pyrimdine: cytosine, uracil

A = U (2 hydrogen bonds)
G 三 C (3 hydrogen bonds)

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

Name the difference types of RNA and their assoc RNA polymerases?

A

rRNA (ribosomal)- up to 5kb- structural - made by RNA Pol 1
mRNA (messenger) - 1-10kb- carry messages to encode proteins - made by RNA Pol II
tRNA (transfer)- 76-90bp (very small) - made by RNA pol III

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

What are exons and introns?

DEFINITELY IN CURRICULUM

A

Exons: coding DNA
introns: non-coding DNA

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

Define anaplasia?

DEFINITELY IN CURRICULUM

A

Lack of differentiation and loss of morphological characteristics. Cellular and nuclear pleomorphism (different sizes). Hyperchromatic nuclei. Loss of orientation/polarity. Increased nuclear:cytoplasm ratio. Frequent mitoses +/- multipolar spindles. Giant cells

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

What is dysplasia and CIS?

DEFINITELY IN CURRICULUM

A

Dysplasia is disordered growth, usually in epithelium. Loss of cellular uniformity and architectural orientation, pleomorphism, increased mitotic figures in abnormal location (not just basal layer). Cells retain their polarity (unlike anaplasia). Usual progressive maturation is lost

Carcinoma In situ: full thickness dysplasia with basement membrane intact and no extension to subepithelium

Both can progress to cancer but can normalise

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

What is metaplasia?

DEFINITELY IN CURRICULUM

A

Reversible change of one cell type to another cell type- eg Barretts (squamous to columnar) or smoker’s bronchial epithelium (columnar to squamous). - may be pre-malignant

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

What is hyperplasia?

DEFINITELY IN CURRICULUM

A

Increase in number of cells in a tissue eg HRT and endometrium. May be pre-malignant

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

What are the basic steps of making a protein from DNA? What are post-translational modifications?

DEFINITELY IN CURRICULUM

A

Transcription (DNA transcribed 5’ to 3’ by RNA polymerase binding to promoter and unzipping DNA strand until reaches a terminator sequence. Forms pre-mRNA which gets 5’ cap and introns spliced out and polyA tail (facilitates binding to ribosome) to form mRNA and travels to ribosome from nucleus)

Translation - involves all 3 types of RNA, occurs in a ribosome (made of 2 structural rRNA/protein subunits - ribosome moves to each codon and tRNA brings the amino acids binding to codon by its complementary anticodon sequence- can be free floating in cytoplasm or attached to endoplasmic reticulum) - starts at AUG (methionine/open reading frame) - stops at stop codon (UAG/UGA/UAA) - polypeptide chain formed. Genetic code is redundant – more codons for 1 AA (64 codons = 20 AAs)

Post-translational modifications.
- Chemical modifications that generate heterogeity in proteins, modifying end product of expression and
regulating protein function
- Cleaving – Proteases cut sections leading to activation
- Ubiquitination – tagging with Ubiquitin, leading to degredation by Proteosomes
- Phosphorylation – adding Phosphate groups to change function – activate/deactivate
- Also Acetylation and Methylation – NB: Glycosylation is NOT a post-translational modification

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

Describe the process of transcription? Where and how does it start?

DEFINITELY IN CURRICULUM

A

Usually started at 5’ end of DNA - contains a nucleotide sequence that make up the promoter region.
TATA box - located near the start of transcription is one of the most important regulatory elements
TBP (tata box-binding protein) is a generic transcription factor crucial for the initiation.

Response elements: short DNA sequence in promoter recognised by specific transcription factor.

Reaches stop codon - 3 codons always indicates stop protein synthesis - UAA, UAG, UGA

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

What is a somatic vs germ line mutation?

DEFINITELY IN CURRICULUM

A

Somatic mutation- occur in somatic cells and only affect the individual in which the mutation arises
Germ-line mutation- alter gametes and passed on to offspring

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

Name some types of point mutations?

DEFINITELY IN CURRICULUM

A

Substitutions- transitions/transversions
Deletions
Insertions

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

Describe the two types of base pair substitutions?

DEFINITELY IN CURRICULUM

A

Transitions: convert a purine to another purine

  • 4 types - A↔G, T↔C
  • most result in a synonymous substitution (no change in amino acid due to degenerate code)

Transversions: convert a purine to a pyrimidine and vice versa

  • 8 types
  • more likely to result in non-synonymous mutations

Can result in:
- Nonsynonymous/misense mutation - base pair substitution results in a different amino acid eg sickle cell

  • Nonsense mutation: base pair substitution results in stop codon (short protein)
  • Neutral non-synonymous mutation: base pair substituion results in substitution of an AA with similar chemical properties (does not affect function)
  • synonymous/silent mutation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What type of mutation does an insertion/deletion of a base cause?

DEFINITELY IN CURRICULUM

A

Frameshift mutation: deletions or insertions non divisible by 3 result in translation of incorrect AAs/codons.

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

What is a misense mutation? What is a nonsense mutation? What is a silent mutation?

DEFINITELY IN CURRICULUM

A

Missense: Change from one AA to another due to a base pair substitution.

Nonsense: Base pair substitution results in a stop codon

Silent: which code for the same amino acid due to degenerate nature

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

What is an inversion? What is a duplication?

DEFINITELY IN CURRICULUM

A

Duplications – Repeat of 1 or more nucleotides to another DNA sequence

Inversions – inverted sequence of 2 or more nucleotides within a DNA sequence

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

What is chromothripsis?

DEFINITELY IN CURRICULUM

A

When a chromosome shatters and in an attempt to repair the damage many incorrect junctions occur. Can disrupt tumour suppressor genes and produce oncogenic fusion genes. Shattered DNA fragments may also form extrachromosomal DNA.
CAUSES CANCER
Produces multiple mutations at once. Detected by FISH.

Potential causes include ionising radiation, telomere dysfunction, aborted apoptosis.

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

Which type of UV radiation causes the most cancer?

DEFINITELY IN CURRICULUM

A

UVB

UVA - reaches most acellular dermis (wavelength og 320-380nm)
UVB- reaches epidermis (wavelength 290-320nm)
UVC- absorbed by ozone, rarely reaches skin

Note need a wavelength of <100nm to cause ionising event - hence UV radiation is not ionising. Does have enough energy to break chemical bonds.

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

How does UV radiation cause cancer?

DEFINITELY IN CURRICULUM

A

UVB

Conjugated double bonds in the rings of the nitrogenous bases of DNA absorb UV radiation.
- causes CYCLOBUTANE PYRIMIDINE DIMERS- cause a bend in DNA helix so DNA polymerase cannot read DNA template -> it preferentially incorporates an A reside so TC/CC dimers restored to TT dimers -> result in transitions (TC -> TT, CC-> TT). PYRIMIDINE DIMERS UNIQUE TO SKIN CANCER. Induces NER.
- Also get 6,4 photoproducts- abasic site

UVA

  • indirectly damages DNA via free-radicals, water is fragmented generating ROS -> cause DNA damage
  • G-> T transversions characteristic

DNA-protein crosslinks are also important lesions in cells exposed to UV radiation. Crosslinks are
particularly disruptive, as they occur mostly in the area of the chromosome that is undergoing replication.

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

What is characteristic of UVB radiation damage?

DEFINITELY IN CURRICULUM

A

Pyramidine dimers - 2 types

  • cyclobutane pyrimidine dimers (2/3)
  • 6,4 photoproducts (1/3)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the carcinogen in coal tar from cigarettes and how does it cause mutations?

DEFINITELY IN CURRICULUM

A

Polycyclic aromatic hydrocarbons : Benzo (a)pyrene (the most well-known carcinogen in tobacco smoke)
PAHs metabolised (by CYP1A1 enzyme) -> forms ultimate carcinogen -> forms adducts with purine bases -> results in G-> T transversions.

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

What in nitrosamines and nitrosamines causes cancer?

DEFINITELY IN CURRICULUM

A

Found in tobacco, preserved fish and meats during smoking. Principal carcinogenic product is alkylated O6 guanine derivatives.

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

What are DNA mismatches?

DEFINITELY IN CURRICULUM

A

DNA can base pair incorrectly leading to DNA structure distortion
Tautomeric shifts
Deamination
Loss of bases: depurination, depyrimidination

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

What is a tautomeric shift?

A

A tautomer is a structural isomer.

Thymine and guanine usually in keto forms (C=O) -> undergo spontanous isomerisation to enol form (C-OH) - means they can join to keto forms of T and G

Cytosine and adenine usually in amino forms (C-NH2) -> undergoes spontaneous isomerisation to imino form (C=NH) -> means they can join to amino forms of C and A

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

What is deamination

A

Type of mismatch: Loss of an amino group (from C, G or A) can happen spontaneously and result in conversion of bases.
Cytosine -> uracil
Adenine-> hypoxanthine
Guanine -> xanthine

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

Name the different types of DNA repair?

DEFINITELY IN CURRICULUM

A
Direct repair
Base exicision repair- most common
Nucleotide exicison repair
Mismatch repair
DSB repair
- HR
- NHEJ
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What is direct DNA repair and name some examples of direct DNA repair?

DEFINITELY IN CURRICULUM

A

Damage is recognised by a protein factor and directly chemically reversed

Bulky alkyl adducts

  • recognised by O6- alkylguanine DNA alkyltransferase (AGT/ MGMT), the damaged base is flipped out of the DNA helix- methyl moeity is transfered to AGT protein
  • MGMT, known as O6-methylguanine-DNA methyltransferase,
  • AGT is a suicide enzyme- only does one round of demethylation and it is not regenerated.

Pyrimidine dimer - NOT in placental mammals
- recognised by photolyase which absorbs blue light and breaks the cyclobutane ring

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

Describe the process of base excision repair.

DEFINITELY IN CURRICULUM

A

Targets chemically altered bases due to oxidation, deamination and alkylation (eg 8-oxoguanine- failure to remove this results in G->T transversion mutation)

Initiated by specific DNA glycosylases (eg OGG1, MUTYH) that recognise and remove specific damage to leave an AP- apurinic/apyrimidinic site

  • Scaffold protein XRCC1
  • AP site cleaved by endonuclease
  • repair takes place
  • DNA polymerase B replaces the nucleotide and ligase 3 and 1 fills the gap
  • Poly (ADP-ribose) polymerase- PARP- interacts with single strand breaks and synthesise poly (ADP-ribose) chain that signal to other DNA repair proteins and leads to modification of histones and relaxation of chromatin to increase accessibility

Short patch repair = one nucleotide. Long patch = 2-10 nucleotides.

Single strand breaks recognised by alkaline solution assay.

Important in repairing radiation induced damage.

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

What is the role of PARP in DNA repair?

DEFINITELY IN CURRICULUM

A

SSB
- Poly (ADP-ribose) polymerase- PARP- interacts with single strand breaks and synthesise poly (ADP-ribose) chain that signal to other DNA repair proteins and leads to modification of histones and relaxation of chromatin to increase accessibility

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

What types of damage is NER useful for repairing?

DEFINITELY IN CURRICULUM

A

Specific to helix distorting lesions e.g.

UV induced DNA damage pyrimidine dimers
Bulky DNA adducts e.g. polycyclic aromatic hydrocarbons
Cisplatin

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

Describe NER?

DEFINITELY IN CURRICULUM

A

Helix-distorting lesions
Recognition of damaged site (XPC and XPA) lead to unwinding of DNA by helicases (XPB and XPD) and excision of a short ssDNA segment by ERCC1-XP F nuclease (usually 10-20 bases) containing damage
Lots of other XP proteins involved
DNA polymerase copies the undamaged strand and DNA ligase seals off the ends.
No loss of information. PCNA (proliferating cell nuclear antigen) is associated with DNA polymerase.

Two forms- differ in how they recognise the damage
- GG-NER- global genome repair occurs continuously but takes a long time to spot the abnormality - continuously scanning for helix distortion
- TC-NER- transcription coupled repair- actively transcribed strand of DNA is repaired with greater efficacy - RNA polymerase comes across the DNA backbone kink and stalls - NER recruited and repairs - transcription can continue

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

When does mismatch repair occur?

DEFINITELY IN CURRICULUM

A

Corrects errors that arise spontaneously during DNA replication. When there are base-base mismatches or insertions/deletions introduced due to slippage during replication of repetitive sequences (e.g. microsatellites).

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

What disease is caused by NER deficiencies?

DEFINITELY IN CURRICULUM

A

Xeroderma pigmentosum
Defects in 1 of 7 proteins XP A-G
Rare AR disorder
Extremely sensitive to UV light
No radiosensitvity
1000x risk Skin cancer, 20x risk other malignancies, life expectancies 20-30s, keratitis, mental retardation, premature dementia

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

How does mismatch repair work?

DEFINITELY IN CURRICULUM

A

hMSH2/3 (indels) or 2/6 (mismatch) (also called MutS) recognises distorted structure in DNA and recruits MutL heterodimers (MLH1/PMS1) which have endonuclease activity - this is the MMR complex - allows strand discrimination by putting nicks into the incorrect strand. PCNA is also recruited. Exonuclease cuts out the nucleotides. DNA polymerase/ligase resolves. Gap is filled by polymerase and sealed by ligase.

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

Which cancer syndrome is caused by mismatch repair deficiencies?

DEFINITELY IN CURRICULUM

A

HNPCC/lynch - hereditary non-polyposis colorectal carcinoma = colorectal/endometrial/ovarian cancer. Autosomal dominant. Less severe than FAP.

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

Describe the process of NHEJ?

DEFINITELY IN CURRICULUM

A
  • DNA ends recognised by Ku 70/80 (XRCC6/5) proteins -> recruit DNA-PK catalytic subunit. The Ku heterodimer has a high affinity for DNA ends and forms a close-fitting asymmetrical ring that
    threads onto a free end of DNA
  • DNA-PKcs associates with Ku70/80 to form the DNA-PK holo-enzyme. DNA-PK holds ends together. DNA-PK phosphorylates H2Ax so opening chromatin.
  • recruits artemis protein which trims ends.
  • DNA ligase IV forms a tight complex with XRCC4. XRCC4 and ligase 4 rejoin ends
    If DNA ends compatible they can be directly ligated together but often processed (resected) then ligated back together. Error-prone, mutagenic, quick, efficient, throughout cell cycle, can lead to chromosome translocations and telomere fusion

Important in repairing radiation induced damage, particularly in late responding tissues.
important in V(D)J recombination - therefore get immunodeficient if mutated - SCID

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

What are the pros and cons and NHEJ?

DEFINITELY IN CURRICULUM

A

Quick 2-4hrs

LOST DNA, MUTAGENIC

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

When in the cell cycle does NHEJ occur?

DEFINITELY IN CURRICULUM

A

Can occur at any point.
Very important in G1 PHASE of cell cycle as HR doe not occur.
Non proliferating normal tissues with low mitotic rate eg brain, kidney cord sit in G1 phase so if you were to inhibit NHEJ then would get back late toxicity.

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

What has overriding control and orchestrates response to DSBs?

DEFINITELY IN CURRICULUM

A

ATM

  • phosphorylation of histone H2AX by ATM/DNA-PK causes recruitment of proteins to site of damage. H2AX phosphorylation represents an important step in the formation of nuclear foci of DNA repair
    proteins (also phosphorylated by ATR and DNA-PK)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Describe HR

DEFINITELY IN CURRICULUM

A

MRN complex- made up of Rad50/Mre11/Nbs1 complex binds to ends of DNA and uses endonuclease activity to create single strand 3’ end overhangs. (Mre11 also activates ATM)
Rad52 binds to DNA termini
RAD51 is a recombinase and forms a nucleoprotein filament that facilitates strand invasion
for homologous recombination. Rad51 binds to exposed ends to form nucleoprotein - BRCA 1/2 aids in nuclear transport of Rad51 and Rad52 helps with binding of Rad 51 to exposed ends. Needs the BRCA1-PALB2-BRCA2 complex.
Rad51 helps to search for homologous template
When homologue is found the 3’ end of ssDNA serves as a primer to initiate DNA synthesis.
BCM protein (deficient in Bloom’s syndrome) help migrate the junctions towards each other
Resolvases restore the junctions known as Holliday junctions.

Important in repairing radiation induced damage. No loss of information

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

What does the fanconi anaemia pathway do?

A

Repair inter- strand cross link which block replication forks - repairs stalled replication forks. Bulky adducts so NER started - causes dsDNA breaks as on both sides so HR then started. Fanconi proteins important in this NER/HR process of interstrand repair. Therefore Fanconi anaemia patients very sensitive to cisplatin.
Assembly of FA complex which recruits nucleases.
Breaks DNA and lesion is bypassed by translesional synthesis and further repaired with NER
Gap in 2nd strand is a double strand break and is repaired by HR

Fanconi anaemia patients get a lot of quadriradials

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

What is the double hit hypothesis

A

One copy of a gene is inherited mutated (germ line) in all cells of the body. Later on a second copy gets randomly mutated (somatic) which pushes the cell towards oncogenic transformation

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

What is synthetic lethality?

A

Genetic concept that describes buffering effect genes have on each other functions. If one targets a synthetic lethality partner of mutated gene one will achieve selected cell death in only cancer. PARP1 inhibitors in BRCA1/HR deficient cancers is the main example.

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

Why are BRCA and PARP synthetically lethal?

A

PARP is a key mediator in BER
BRCA1/2 key mediator in HR
In BRCA mutant cell HR is dysfunctional so cells heavily rely on BER. Because BER is blocked the single strand breaks become DSBs and HR not working to toxic to cells.

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

What causes genetic instability?

A

Replication errors- eg mismatch repair
Replication problems- impeded progress
Damage to DNA
Mitotic errors - chromosome segregation defects eg spindle assembly checkpoints.

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

What is a transcription factor?

DEFINITELY IN CURRICULUM

A

Protein that binds to gene promoters and regulates transcription
3000 transcription factors regulate 20,000 genes

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

What is the structure of a transcription factor?

DEFINITELY IN CURRICULUM

A

Contain a DNA binding domain, transcriptional activation domain (Recruits the generaly replication complex), dimerisation domain and ligand binding domain (e.g. steroid hormone for steroid receptors).

4 types of DNA binding domains:
- helix-turn-helix motif
- Leucine- zipper motif
- helix- loop-helix motif
- Zinc finger motif
These domains are characteristic protein formations that enable transcription factor to bind to DNA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

How is the activity of a transcription factor regulated?

DEFINITELY IN CURRICULUM

A

Synthesis in particular cell types only
Covalent modifications eg phosphorylation
ligand binding
Dimerisation - exchange of partner proteins

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

Give some examples of transcription factors?

A

AP-1 (jun and fos family, dimerise in different ways)
Myc family (Myc, max, mad, mxi)- dimerise in different ways
Steroid hormones
RAR - retinoic acid receptor
p53 - mutated in 50% cancers

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

What is AP-1 and how does it work?

DEFINITELY IN CURRICULUM

A

Transcription factor.
AP1- binds to TPA response element or to cyclic AMP response element in the promoter of target genes

AP-1 is made up of two components and is produced by dimers from 4 families: Jun, Fos, ATF/CREB and MAF family. 18 possible combinations. Fos and jun contain leucine zipper dimerisation domain. Promote proliferation generally.
Jun B acts as a negative regulator of growth when dimerised with Jun

AP-1 activated by specific signals e.g. growth factors (MEK/ERK pathway), ROS, radiation

AP-1 made from c-Fos and c-Jun can transform a normal cell to malignant cell

Activates cyclin D

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

How do steroid hormone receptors act as transcription factors?

A

Superfamily of steroid hormone receptors act as ligand-dependent transcription factors.
Nuclear receptor family (48 members) eg Androgen receptor, oestrogen receptor, glucocorticoid receptor, mineralocorticoid receptor, progresterone, PPAR, retinoic acid, thyroid hormone receptor, vitamin D receptor.
Contain a zinc finger DNA binding domain, ligand binding domain for specific steroid hormones and dimerisation domain.
Each domain specific for that hormone.
Steroid hormones pass through cell membrane and bind to intracellular receptor in cytoplasm or nucleus -> receptors move to nucleus and activate transcription through specific DNA response elements.

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

What is the retinoic acid receptor (RAR) and how does it work?

A

Transcription factor
RAR is located in nucleus and is a heterodimer with RXR. This heterodimer acts as a transcriptional repressor in absence of retinoic acid (derived from vit A) by recruiting co-repressors. It binds to RA response element (RARE) in target genes. In vitamin A presence, it binds to co-activators instead.
Aberrant forms of RARs are characteristic in some forms of leukaemia. PML-RAR recruits HDACs and DNMTs to silence differentiation genes - all-trans retinoic acid is used in treatment of APML to change the PML-RARA to an activating rather than silencing transcription factor = differentiation.

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

What can a somatic mutation upstream of TAL1 gene produce?

A

TAL1 (T cell Acute Leukaemia 1)- oncogene which codes for basic helix-loop-helix transcription factor -> create a super enhancer which upregulates expression -> loads of transcription factors.

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

What makes up chromatin, nucleosomes and histones

DEFINITELY IN CURRICULUM

A

Chromatin : thread of DNA 1m long when stretched out (60%), assoc RNA (5%), protein (35%). Structural scaffold tightly wound to form chromosomes.

Nucleosomes formed into fibres then radial loops.

Nucleosome: 147 base pairs of DNA wrapped 1.7 x around core histone proteins. Histone core is an octomer of histones (2 of each of H2A/H2B/H3/H4 and then H1 is associated with the linker DNA between nucleosomes)

Histone: contains domains for histone-histone, histone-DNA interactions and NH2-terminal lysine rich; COOH rich terminal tail domains which can be modified eg methylated/phosphorylated.

  • Histones play a role in protecting against ionising radiation induced damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What is epigenetics?

DEFINITELY IN CURRICULUM

A

Heritable information that is encoded by modifications of genome/chromatin components (not a change in DNA sequence so NOT mutations)

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

What are the two most common types of epigenetic modification?

DEFINITELY IN CURRICULUM

A

Histone modification
DNA methylation
Both can be acquired or inherited.

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

What types of histone modification are there?

DEFINITELY IN CURRICULUM

A

Acetylation (euchromatin)
Methylation (H3K4me3 euchromatin; H3K9me, H3K27me3 is heterochromatin)
Phosphorylation
Ubquination

Measured by ChIP-Seq

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

Describe histone acetylation?

DEFINITELY IN CURRICULUM

A
Alters chromatin structure and effects gene expression. 
Acts as a docking signal for recruitment/replusion of chromatin
Histone acetyltransferases (HATs) - add acetyl group to histone tail lysines - neutralises positive charge on lysine residues and relaxes chromatin folding  - transcriptional activators recruit HATs

Histone deacetylases (HDACs) - remove acetyl group.

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

Name some examples of histone acetylation causing cancer.

A

EP300 gene codes for p300 protein a HAT
P300 usually acts as a tumour suppressor. Mutated in epithelial cell tumours.

Chromosomal translocation produces PML-RAR (APML)- recruits HDAC to promoter region of RA target genes and represses the expression of genes -> blocks cell differentiation. Give trans-retinoic acid = no longer recruits HDAC - genes activated instead = differentiation.

Oncohistones = mutated histones that cause cancer.

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

Where does DNA methylation occur on DNA?

DEFINITELY IN CURRICULUM

A

Addition of methyl group to position 5 of cytosine in CG dinucleotide

ONLY occurs at cytosine nucleotides which are situated 5’ to guanine (CpGs)

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

Where do you find the most CpGs?

DEFINITELY IN CURRICULUM

A

CpG is unequally represented in genome- which may be due to 5-methylcytosine easily deaminating to thymine causing a C-> T transition.
CpG islands- clusters of CpG located in promoter region of genes.

Methylated cytosines are mainly found in repressed genes eg X chromosome, inactivated genes, imprinted genes -> methylation is a heritable signal and assoc with compact chromatin structure and maintains gene silencing

Not clear how it works to silence - perhaps stops binding of transcription factor; recruit chromatin modifying enzymes; induce mutation due to deamination.

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

What are DNMTs and what types are there?

DEFINITELY IN CURRICULUM

A

DNA methyltransferases - mediate the covalent addision of a methyl group from a methyl carrier SAM (S-adenosyl-methionine).

Three DNMTs:

  • DNMT1- during DNA replication this methylates DNA if original strand was methylated- allows inheritance of methylation
  • DNMT3a, DNMT 3b- involved in de novo methylation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

Overall do cancer cell have more or less methylation of DNA than normal cells?

DEFINITELY IN CURRICULUM

A

20-60% less methylation - causes genetic instability
Global hypomethylation with hypermethylation of specific gene promoters
30% breast cancers ER negative due to hypermethylation of ERalpha.
BRCA1 can be inactivated by methylation
MGMT (glioblastoma), APC (colorectal), MLH1- commonly methylated.

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

How do you detect DNA methylation?

A

Sodium bisulfite treatment- converts unmethylated cytosine to uracil by deamination.
Then do methylation specific PCR
Change grading of GBM (MGMT promoter methylation)

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

What are microRNAs (miRNAs)

A

Small non-protein coding RNAs (18-25 nucleotides) regulate expression of mRNAs
Able to repress hundreds of gene targets post transcriptionally -> powerful regulators of growth, differentiation and apoptosis.

Repress gene targets binding to 3’UTR of their target mRNA blocks translation
Can be oncogenes or tumour suppressors

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

What are telomeres?

DEFINITELY IN CURRICULUM

A

Protect ends of the chromosomes from digestion by nuclear enzymes and prevent induction of mechanisms of repair of DNA double strand breaks.
Composed of several thousand TTAGGG repeats bound by a protein complex called shelterin complex on 3’ overhang - forms T loop/G quadruplexes so no dsDNA break detected

Telomeres can be transcribed into a lncRNA that contains telomere repeat-containing RNA (TERRA) - essential in maintaining length of telomeres.

Telomere length and distance to a gene can also affect gene expression: telomere position effect.

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

What is the end replication problem? What is the Hayflick limit?

DEFINITELY IN CURRICULUM

A

DNA shortens by 100-200 DNA bases with each round of replication due to limits of DNA polymerases. DNA synthesised in 5’ to 3’ direction therefore complementary strand is synthesised in fragments (Okazaki fragments) with a RNA primer to initiate - these primers are lost so therefore the strand shrinks. Enter senescence once threshold length reached. If cells bypass senescence due to mutation, telomeres become critically short&raquo_space; chromosome instability&raquo_space;apoptosis

Hayflick limit: number of times a cell can replicate - limited by number of telomeres.

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

What is telomerase?

DEFINITELY IN CURRICULUM

A

A ribonucleoprotein containing human telomerase reverse transcriptase (hTERT) and human telomerase RNA (hTR) which maintains the telomere length eg stem cells.
hTERT uses hTR as a template to add new repeats to telomeric DNA
AGAINST CENTRAL DOGMA OF BIOLOGY as synthesising DNA from RNA

  • Physiologically observed in stem cells, germ cells and hair follicles – cells that need to be immortal
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

How does cancer affect telomeres?

DEFINITELY IN CURRICULUM

A

90% cancer upregulates telomerase, likely through reversal of epigenetic changes that occur on differentiation to
somatic cell lineages

Patients with lower TERT levels have better prognosis, promoter mutations in TERT&raquo_space; increase telomerase activity&raquo_space; cancer

  • melanoma and other cancers have found mutations in TERT promoter.
  • c-myc increases expression of hTERT gene.

Telomere shortening occurs in response to replication, ssDNA breaks and oxidative damage so may act as a tumour suppressor limiting replicative potential

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

What are the 4 types of protein involved in the transcription of growth factor signal?

A

Growth factors
Growth factor receptors (many are TKIs)
Intracellular signal transducers
Nuclear transcription factors

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

Name some types of EGFRs and what is their general function?

DEFINITELY IN CURRICULUM

A
- EGFR/ErbB1/HER1
- ErbB2/HER2 - no ligand binding activity - acts as a heterodimer for others
- ErbB3/HER3 - no tyrosine kinase activity
ErbB4/HER4

Family of receptor tyrosine kinases - important for transduction of a signal from an EXTRACELLULAR growth factor through the cell where regulates gene expression. Form homodimers or heterodimers after ligand binding.

RTKs can be indirectly activated by cross talk:
o G-protein coupled receptors activate proteases&raquo_space; cleave precursor growth factors&raquo_space; release of
ligands&raquo_space; RTK activation
o ErbB2 receptors trans-activated by other RTKs and integrins

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

What is the structure of an EGFR

A

Extracellular ligand binding domain (cetuximab binding)
Single hydrophobic transmembrane domain
Cytoplasmic protein tyrosine kinase domain (except HER2 does not bind a known ligand, just acts as a co-receptor and HER3 only has weak kinase activity) (Osimertinib etc binding)

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

How does the EGF signal get inside the cell?

DEFINITELY IN CURRICULUM

A

Binding of EGF to receptor
EGFR receptor dimerisation (one EGF bound to each receptor)
Conformational change in the receptor causes autophosphorylation (one half of dimer phosphorylates other half due to kinase activation)
The phosphorylated tyrosine residues create high affinity binding sites for Src homology 2 (SH2) domains. GRB2 binds to SH2 domains on the EGFR and binds SOS via its SH3 domain so translocating it to the cell membrane. SH3 domains interact with SOS which recruits Ras. Causes Ras to release GDP and bind GTP, activating it.
Ras activates RAF/MEK/ERK and PI3K

SH2&3 domains mediate protein-protein interaction in pathways activated by TKs. Proteins that contain SH2/3 domains = Grb2, ABL, SRC and PI3K

Signal can be terminated by further phosphorylation of EGFR, removal of tyrosine phosphorylation, receptor endocytosis and degradation, and binding of receptor inhibitors.

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

What is Ras and what is its function

DEFINITELY IN CURRICULUM

A

Ras is a GTP binding protein. It is a GTPase. Intracellular transducer. When bound to GDP inactive. SOS releases Ras from GDP and it binds to GTP. (SOS [son of sevenless] is a guanine nucleotide exchange factor)

Binary switch

K-RAS, N-RAS, H-RAS

Ras is loosely bound to inside of cell membrane (also found on subcellular membrane compartments such as endoplasmic reticulum)

Causes activation of Raf (MAPKKK) and PI3K

Mutation results in Ras protein that can’t covert GTP&raquo_space; GDP – therefore remains permanently “on”
- Most commonly mutated oncogene in humans - 30% of human tumours carry Ras mutation – (mainly in
Codons 12, 13, 61) e.g. K-Ras in colorectal cancer, N-Ras in thyroid cancer
- Anti-cancer targets against Ras oncogene include farneslyation transferase inhibitors (Ras requires posttranslational modification by farneslation to become active), up-stream targets (e.g. EGF-receptor -Cetuximab or HER2 – Trastuzumab) and down-stream receptors (e.g. Raf – Sorafenib or MEK – Trametinib)
- Ras hard to target directly (“undruggable”) – binding affinity of GTP to Ras is much higher than that of ATP to kinases and there are no binding domains for small molecule TKIs

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

What is Raf?

A

A serine/threonine kinase- MAPKKK
- Raf recruited to cell membrane and binds to RAS-GTP -> activates it -> signal transducer and carries the signal away from the cell membrane.

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

Describe the Ras-Raf Pathway?

DEFINITELY IN CURRICULUM

A

GF - extracellular
EGFR - dimerisation and autophosphylation -> SH2/3 domains
RAS - GTP (liberated by SOS from GDP)
RAF (MAPKKK)
phosphorylates MEK (MAPKK) (dual tyrosine and serine/threonine kinase)
Phosphorylates MAPK/ERK - serine-threonine kinase
MAPKs -> enter nucleus through facilitated diffusion and phosphorylates Fos to activate AP-1 transcription factors (made up of Jun and Fos family members) and Myc family transcription factors (myc, max, mad, Mxi) dimerise in different ways.

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

Describe the PI3-k pathway?

DEFINITELY IN CURRICULUM

A

PI3K - a lipid kinase interacts directly with Ras. PI3K phosphorylates PIP2 to PIP3 (in cell membrane).
PIP-3 recruits serine/threonine kinase PDK-1 to cell membrane
Then serine/threonine kinase AKT recruited and phosphorylated and activated by PDK-1.
Activated AKT takes signal from the membrane and is involved in anti-apoptotic signals by phosphorylating distant target proteins
- mTOR (serine/threonine kinase) - downstream target Akt which is involved in promoting anabolic programmes eg lipid/nucleotide synthesis.

Akt can also travel to nucleus to the nucleus where is can phosphorylate transcription factors like FOXO (forkhead box O!!!!!!!!)

INHIBITED BY PTEN via PIP3

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

What is Src?

A

Intracellular tyrosine kinase- plays role in proliferation, adhesion, invasion and motility
Src normally phosphorylated which blocks its SH2 and SH3 domain.
Src activated by tyrosine kinase receptors like EGFR - reveals its active domains.
Integrins activate focal adhesion kinase/Src complex. Activated FAK-Src functions to promote cell motility (by inhibiting E cadherin and promoting disassembly of focal adhesions), cell cycle progression and cell survival.

First oncogene discovered - from Rous Sarcoma Virus. v-SRC - lacks negative regulatory domain so constitutively active.

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

What is an oncogene? What is a proto-oncogene?

A

Proto-oncogene: normal cellular genes that encode proteins that stimulate proliferation/cell viability/inhibit cell
death (GO). c-GENE NAME

Oncogenes: Mutated genes whose protein product is produced in higher quantities or whose altered product has increased activity and therefore acts in a dominant manner and contributes to carcinogenesis. v-GENE NAME
Discovered in transforming retroviruses in animals (gene “captured” from host organism), v-onc found to be homologous to mammalian genes. Origin of names e.g. K-Ras – Kirsten rat sarcoma virus (K-Ras)
Types
- retroviruses - rous sarcoma virus
- growth factors e.g. PDGF
- growth factor receptor e.g. RET, EGFR, HER2
- Intracellular signal tranducers eg Ras (mutated in 30% tumours), BRaf
- Transcription factors

> 100 oncogenes have been identified.

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

Give an example of growth factor as an oncogene

A

Proto-oncogene = c-sis produces PDGF
Oncogene = v-sis causes unregulated growth via activation of PDGF pathway - inappropriately cytoplasmic so constitutively active.

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

Give an example of a growth factor receptor oncogene

A

Proto-oncogene- RET - TK receptor
Transduces signal to glial derived neurotrophic factor (GDNF) family ligands when in heterodimer with cofactors
- familial medullary thyroid cancer
MEN2A and MEN2B
Oncogenic activation can lead to constitutive activation by dimerisation or increased kinase activity

v-ERBB = truncated EGFR = no extracellular domain = constitutively active. Also often activated by point mutations infering with growth factor binding and gene amplification.
HER2 over-expression driving cancer cell proliferation – a genetic copy number
variation or epigenetic change&raquo_space; over-expression or ErbB2. ErbB2 homo/heterodimerization&raquo_space; amplified oncogenic signalling
Mutations&raquo_space; inducing activation/inhibiting inactivation E.g. L858R mutation of EGFR – most common EGFR mutation causing cancer, leads to permanently ‘active’ structural conformation, or E.g. EGFR viii deletion mutation – loss of extra-cellular domain, therefore ligand independent and constitutionally ‘active’ – linked to cisplatin/cetuximab resistance in oral
cancers
Geneomic rearrangement/translocation&raquo_space; improper localisation/activation E.g. Fusion of intracellular domain of ALK with dimerization/oligomerization domain of
another protein

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

Name some intracellular transducers that can become oncogenes

A

RAS

  • 30% human cancers
  • loss of GTPase activity- usually required to return active RAS-GTP to RAS-GDP causing constitutive activation

B-RAF
- melanoma (V600E)- causes constitutive kinase activity and insensitivity to feedback

Genes that code for cytoplasmic TKs can become oncogenes:

  • SRC- colon cancer Tyr530 on Src cannot form its inactive phosphorylated form so always active
  • MAPK
  • ABL (nuclear tyrosine kinase - 9:22 translocation forms BCR-ABL which form homooligomeric complexes that autophosphorylate so constitutively active. Also stays in cytoplasm instead of nucleus)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

Name some transcription factors that can become oncogenes?

A

AP-1

  • components of AP-1- jun and fos are encoded by protooncogenes c-jun, c-fos.
  • truncation at end of v-fos means it produces an mRNA with a longer half life.
  • both c-fos and c-jun increased by overexpression and amplification

C-myc
- chromosomal translocation of myc (chromosome 8) to a location that falls within regulation of strong promoter of imumunoglobulin genes (chromsome 14) increases expression of myc gene -> Burkitts lymphoma

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

What are the mechanisms of oncogenic activation

DEFINITELY IN CURRICULUM

A
  1. Point mutations and deletions in coding regions e.g. loss of GTPase function in Ras so constituvely activated
  2. Mutations in gene promoter region
  3. Chromosomal translocations e.g. BCR-ABL, IMH-myc + insertional mutagenesis
  4. Gene amplification from the normal 2 copies in diploid genome eg HER2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

How long is the cell cycle?

DEFINITELY IN CURRICULUM

A

Average length 16hrs
15hrs interphase (G1 is most variable and longest in length)
1hr mitosis (shortest phase)
Varies depending on cell type
Chromosomes can only be observed in mitosis due to condensation

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

When is a cell irreversibly committed to the cell cycle?

DEFINITELY IN CURRICULUM

A

On passing G1 restriction point

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

Describe the order of the cell cycle

DEFINITELY IN CURRICULUM

A

G0: Terminally differentiated – not dividing but expressing proteins required for cellular house-keeping. Most lymphocytes are in G0 and awaiting the appropriate antigen, which will stimulate to re-enter cell cycle at G1. mitogens/growth factors induce cells to re-enter cycle

Gap1: Longest part of cell cycle/most variable. Intense (mRNA) transcription of proteins for DNA synthesis
Check-point at G1 ensures favourable cell division conditions

G1/S checkpoint: Cyclin D + cdk4/6 = Rb hypophosphorylation + cyclin E/CDK2 = Rb hyperphosphorylation = S phase proteins + cyclin E/A/CDK2 = origin of replication complex.

S phase: DNA duplicated into 2 sister chromatids. Replicate two complete sets of all 46 chromosomes (2n to 4n DNA)

G2 phase: Synthesis of protein required for mitosis and cytokinesis. Checkpoint at G2 to confirm DNA replicated correctly and fit to proceed to Mitotic phase

G2/M checkpoint: Cyclin B/A + cdk1 = chromosome condensation/nuclear envelope breakdown/spindle formation

Mitosis: prophase, metaphase, anaphase, telophase

M checkpoint: chromatids lined up on metaphase plate = anaphase promoting complex = anaphase occurs

G2/M (due to mitotic catastrophe) >G1>early S>late S (due to presence of sister chromatid to use for HR) = radiosensitivity

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

How do cyclin-cdk complexes exert their effect?

DEFINITELY IN CURRICULUM

A

Cyclins: Regulate progression through the cell cycle. Binding of Cyclin to CDK = conformational change = reveals active site of CDK. Concentration in cell rise and fall throughout cell cycle. Cyclins are present only during short periods within the cell cycle and are controlled by their own degradation. Degraded by ubiquitin proteasome.

CDKs: concentration is constant. When activated by cyclins, they phosphorylate target proteins including transcriptional regulators, cytoskeleton proteins, nuclear pore, envelope proteins, histones. Dephosphorylation is important for resetting the cycle.

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

Where does cyclin D act and how?

DEFINITELY IN CURRICULUM

A

Drives progression through G1
Binds to cdk4/6
ONe of its final targets is EGF signalling pathway
Phosphorylates Rb = releases HDAC = transcription of cyclin E

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

Where does cyclin E act and how?

DEFINITELY IN CURRICULUM

A

Important for G1-S phase progression. Cyclin E/CDK2.
Hyperphosphorylates Rb = E2F released and drives transcription of S phase proteins.

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

Where does cyclin A act and how?

DEFINITELY IN CURRICULUM

A

Important for S phase progression. Formation of origin of replication complex with CDK2
Directs G2 and G2->M phase. Drives nuclear envelope breakdown, spindle formation and chromosome condensation.Important for G2/M checkpoint with CDK1:

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

Where does cyclin B act and how?

DEFINITELY IN CURRICULUM

A

Directs G2 and G2->M phase. Drives nuclear envelope breakdown, spindle formation and chromosome condensation.
Binds to CDK1

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

What are cyclin dependent kinases?

DEFINITELY IN CURRICULUM

A

Serine/threonine kinases that regulate progression of the cell cycle via phosphorylation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What are the mechanisms of cdk regulation

DEFINITELY IN CURRICULUM

A
  • Association with cyclins - activates cdk by conformational change revealing active site, cyclins degraded by proteasomes after being flagged by ubiquitin
  • Association with cdk inhibitors - 2 families p16ink4a (INK) family and p21 (cip/kip) family.
  • INK4 proteins (p16, p15, p18, p19) bind cdks 4/6 and interfere with binding to cycle D. p14 ARF inhibits the MDM2-mediated degradation of p53. p16INK4A is a cell cycle inhibitor that prevents phosphorylation of RB by CDK4. p14ARFis an MDM2 inhibitor thereby causing p53 levels to increase, resulting in greater cell cycle inhibition.
  • p21 family members (p21, p27, p57) interact with both cyclins and their associated cdks and block ATP binding sites
  • phosphorylation causing activation e.g. by CDK activating kinase (CAK)

-phosphorylation causing inactivation e.g. by Wee1 kinase.
- dephosphorylation causing activation (cell cycle division 25 phosphatase cdc25 dephosphorylates Wee1 site). CHK1/2 (serine/threonine kinase) phosphorylate and inhibits cdc25 - activated by DNA damage.

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

What is a key substrate of cyclin d - cdk4/6 complex?

DEFINITELY IN CURRICULUM

A

Rb protein

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

How does Rb protein regulate the G1 checkpoint?

DEFINITELY IN CURRICULUM

A

Rb protein is a key substrate of cyclin d-cdk4/6 substrate
Rb regulates activity of E2F transcription factor- crucial for expression of genes needed for S phase.

Hypophosphorylated Rb sequesters HDAC and E2F so transcription repressed.
Cyclin d-cdk4/6 causes partial phosphorylation of Rb and release of HDAC -> repression relieved for some genes like cyclin E
Additional phosphorylation by cyclin E- cdk2 causing release of E2F
-E2F (transcription factor) initiates the transcription of genes required to enter S phase including cyclin A

This occurs in response to growth signals

DNA damage is detected and repaired to ensure accurate transmission of genetic
material – halts cell cycle at G1 until DNA repaired, ensuring faulty DNA is not
replicated in S phase. DNA damaged detected by p53. ↑ p53 protein → ↑p21 level → inhibits cyclinE-cdk2 complexes →inhibits phosphorylation of Rb protein
i.e Rb proteins remain bound to transcription factor E2F→ G1/S arrest for DNA repair

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

What happens at the G2 checkpoint?

DEFINITELY IN CURRICULUM

A

Blocks entry into M phase in cells that have incurred DNA damage - allowing repair
DNA damage activates either -> ATM or ATR -> phosphorylate and activate Chk 2 and Chk1 respectively
Chk 1/2 prevents cdks from becoming active by phosphorylating and inactivating Cdc25.
Halts cell cycle progression
After successful repair- PLK1 which targets Chk1 for degradation and inhibits Chk2.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

Describe the steps in mitosis

DEFINITELY IN CURRICULUM

A

Prophase- appearance of chromosomes as result of condensation, nuclear membrane breakdown, separation of duplicated centrosomes, Mitotic spindle assembled (a set of microtubules extending from the centriole which will
later attach to chromosomes)

Metaphase- align chromosomes on metaphase/equatorial plate and assembly of microtubules to form mitotic spindle

Anaphase- spindle pulling apart and separating chromatids, move towards spindle poles

Telophase- accumulation of chromosomes at their respective poles, reforming nuclear membrane, chromosome decondensation and cytokinesis (cytoplasmic division)

Chromosome refers to the whole package of DNA: pre S phase this contains 2n (double-stranded helix), post S phase contains 4n (two sister chromatids with double-stranded helix). Chromatid is 2n.

Centromere is part of chromosome where spindle attaches

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What happens at the spindle assembly checkpoint/mitotic checkpoint?

DEFINITELY IN CURRICULUM

A

Signalling cascade that ensures the correct chromosomal segregation during mitosis and production of two genetically identical nuclei. Anaphase-promoting complex (ubiquitin-ligase) inhibits securin which usually inhibits separase - therefore APC activates separase which separates the chromatids by degrading cohesin so activating anaphase. APC is inhibited until the chromatids are lined up on the metaphase plate.

APC also inactivates cyclins so prevents re-entry into cell cycle.

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

What does anaphase complex do?

DEFINITELY IN CURRICULUM

A

An ubiquitin–protein ligase that regulates mitosis
During metaphase unattached chromatid pairs recruit proteins to form the mitotic checkpoint complex that inhibits anaphase complex. Once chromatids attached to microtubules they stop inhibiting it.
Anaphase complex targets securin - once degraded protease separase is activated
Separase cleaves cohesin link between sister chromatids allowing them to separate

APC also marks cyclins for degradation

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

What are aurora kinases?

A

Aurora kinases A, B, C
Regulate important aspects of mitosis
Serine -threonine kinases that phosphorylate target proteins

Aurora kinase A (STK15): localises to centrosomes during interphase; thought to play role in centromere maturation and spindle formation.
Aurora kinase B (STK12): involved later in mitosis, role in spindle attachment to chromosomal centromeres and the
spindle checkpoint, in chromosomal segregation and cytokinesis
Aurora kinase C (STK13): active during late mitosis. Localises to spindle poles

Aurora kinases frequently overexpressed in cancers

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

What mutations in cdks are you aware of?

A

Miscoding mutation in cdk4 stops it binding to INK4 inhibitors in subset melanoma patients
Cdk4 required for development of mammary gland tumours
Overexpression of cdk6 in some leukaemias

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

Name some tumour suppressor genes?

A

BRCA1/2
PTEN
Rb
p53

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What is a tumour suppressor gene?

DEFINITELY IN CURRICULUM

A

Tumor suppressor genes represent the opposite side of cell growth control, normally acting to inhibit cell proliferation and tumor development. In many tumors, these genes are lost or inactivated, thereby removing negative regulators of cell proliferation and contributing to the abnormal proliferation of tumor cells.

Functions in DNA damage repair, blocking cell division and controlling apoptosis. Are usually recessive – both genes need to be mutated (exceptions are P53 and P27). Exceptions are due to haploinsufficiency – ONE mutated allele can lead to cancer as only half the normal quantity of protein is produced which isn’t enough to stop tumour formation in that specific case

Most familial cancer syndromes are due to mutations in tumour suppressor genes apart from MEN2A/2B which is RET.

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

What is the role of BRCA1?

A

Recruitment of Rad51 to DSBs - HR. (Also has broader role in NHEJ and RNA polymerase - ubiquitination. Thought to inhibit oestrogen receptor mediated transcription hence less breast/ovarian.

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

What is the role of BRCA2?

A

The localization of RAD51 to the DNA double-strand break requires the formation of the BRCA1-PALB2-BRCA2 complex.

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

What is PTEN?

A

Gene encoding a phosphatase with dual specificity. It can act as a protein and lipid phosphatase.
PTEN -> dephosphorylates the membrane lipid PIP3 -> PIP2
so inhibiting the PI3K pathway

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

How does PTEN mutation cause cancer?

DEFINITELY IN CURRICULUM

A

Loss of inhibitory dephosphorylation activity of PTEN (for PIP3) can result in a consituitively active PI3K pathway.

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

What syndromes does a germline mutation of PTEN cause?

DEFINITELY IN CURRICULUM

A

Cowden disease aka hamartoma syndrome. Autosomal dominant. BET – breast, endometrial and thyroid cancers.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

Describe the differences in the sporadic and familial form of retinoblastoma

DEFINITELY IN CURRICULUM

A

Tumour suppressor gene
Familial (40% cases)

  • one germline mutation and second sporadic- often results from somatic mitotic recombination in which normal gene is replaced with the mutant copy - Knudson’s 2 hit hypothesis
  • often bilateral
  • autosomal dominant
  • also associated with small cell lung cancer

Sporadic form (60% cases)

  • both mutations occur somatically in the same retinoblast.
  • low chance of this occuring > once so usually only affects one eye
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

What is Rb?

DEFINITELY IN CURRICULUM

A

Transcriptional co-factor that can bind to transcription factors and either inhibit or induce transcription factor activity
Rb has >100 known protein binding partners
Main role is to regulate G1 -> S phase transition
Facilitates activity of E2F and chromatin remodelling enzymes
Cell cycle arrest can be induced by Rb via stabilisation of CDK inhibitor p27.

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

What are the upstream activators of p53?

A

DNA damage
Aberrant growth signals
Cell stress- radiation, drug, hypoxia, nucleotide depletion

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

What are the downstream responses possible when p53 is activated?

A

Cell cycle arrest or senescence (p21 - inhibits CDKs so inhibits G1/S and G2/M checkpoint)
DNA repair (DNA repair proteins e.g. XPC)
Apoptosis (e.g. PUMA)
Inhibition of angiogenesis (thrombospondin 2)

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

What is the structure of p53 protein

A

p53 - phosphoprotein transcription factor containing 4 distinct domains:

  • Amino-terminal transactivation domain and MDM2 binding site
  • DNA binding domain containing Zn ion
  • Tetramerisation domain
  • carboxy-terminal regulatory domain

-p53 binds as a tetramer to p53 response element

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

How is p53 regulated?

DEFINITELY IN CURRICULUM

A

Regulated at level of protein degradation not gene expression
Main regulator = MDM2

p53 present transiently as constantly being degraded.

Other regulators: MDMX and HAUSP (removes Ubiquitin form p53)

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

What is MDM2?

A

Main regulator of p53 protein
Ubiquitin ligase (flags protein for proteolysis)
Modifies the carboxy-terminal domain of p53 tagging it for degradation.
It also binds and inhibits p53 transactivation domain and transports protein into cytoplasm away from nucleus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

How is MDM2 regulated?

A

p53 stimulates production of MDM2

Low amounts of p53 will reduce transcription of MDM2

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

What is the activation pathway (upstream of p53) when a DSB occurs?

A

DSB -> stimulates ATM -> phosphorylates and activates Chk2 -> ATM + Chk2 phophorylate amino-terminal sites of p53 -> interferes with binding of MDM2

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

What is the activation pathway (upstream of p53) when cellular stress occurs?

A

Cell stress -> activates ATR -> casein kinase II -> phosphorylates p53

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

What is the activation pathway (upstream of p53) for oncogene activation?

A

Activated oncogenes eg Ras -> activity of protein p14arf -> sequesters MDM2 to nucleolus of nucleus

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

What are the downstream activators once p53 has been activated?

A

INHIBITION OF CELL CYCLE
Transcriptional induction of p21 gene -> product p21 inhibits several cyclin-cdk complexes and causes pause in G1->2 transition

APOPTOSIS

  • several mediators of apoptosis transcriptionally regulated by p53
  • induces pro-apoptotic proteins NOXA, PUMA, p53AIP1
  • tips balance regulated by Bcl-2 towards apoptosis

DNA repair and angiogenesis

  • Gene XPC is involved in nucleoside excision repair and is regulated by p53
  • Thrombospondin an inhibitor of angiogenesis is also regulated by p53
  • Induction of miRNAs is important for inhibiting stem call and preventing mets
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

How does p53 decide which of the downstream outcomes occur?

A

P53 chooses between apoptosis and cell cycle pausing via – phosphorylation of Ser46 (apoptosis); pulses (pause) vs sustained expression (apoptosis); presence of cofactors e.g. ASPP (apoptosis stimulating proteins of p53) push to apoptosis

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

What regulates p21?

A

p53 and MIZ 1 bind to promoter and induce transcription resulting in cell cycle inhibition

However if myc is present it competes with p53 and binds to p21 and inhibits transcription blocking cell cycle inhibition

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

What is the most common type of mutation in p53? How does this compare to other tumour suppressor genes?

A

> 75% p53 mutations = missense mutations - most of these are located in the DNA binding domain

Differs from classical tumour suppression genes -> tend to have nonsense or frameshift mutations that lead to inactivated truncated proteins.

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

What is Li-Fraumeni and how is it inherited?

DEFINITELY IN CURRICULUM

A

Germline mutation of p53
AD disease - haploinsufficiency can be sufficient to cause cancer. p53 important in apoptosis, DNA repair, senescence.
25 x increase risk of developing cancer <50yrs. 50% will have cancer before aged 30.
Sarcomas, HER2+ breast cancer, leukaemia, brain tumours, choroid plexus tumours (specific - refer for testing), adrenocortical tumours (specific - refer for testing. Very sensitive to carcinogens (if smoker will definitely get lung cancer, if have ionising radiation for cancer then will get radiation induced sarcoma)

RADIOSENSITIVE - sarcomas

However, their tumours are actually radioresistant so radiotherapy less effective.

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

Patients with Li-fraumeni syndrome do not usually lose other p53 gene, remain heterozygous. This does not fit with Knudsons hypothesis. Why might this be?

DEFINITELY IN CURRICULUM

A

Reduced amounts of p53 (haploinsufficiency) can cause transformation, also specific mutations may result in varied amounts of tumour suppressor.
Some p53 do not lead to loss of function but instead form altered protein that interacts with the normal p53 and inactivates it. p53 is tetramer so just one abnomral protein inactivates whole tetramer

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

What is the continuum model of tumour suppression?

A

Integrates the two hit hypothesis with broader concept
Subtle dosage effects of tumour suppressor either as changes in level of expression or protein activity
eg p53- one dominant negative mutation can cause tumour suppression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What can viruses do to p53?

A

Adenovirus E1A, papillomavirus E6 & E7 etc inactivate Rb and p53.
Some do this using uquitin-proteosome system to degrade it.

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

How does HPV virus affect p53?

A

p53 is rarely mutated
binding of HPV protein E6 to p53 causes it to be flagged for degradation

Polymorphisms in p53 gene leads to differences in risk for cervical cancer. eg pts with 2 allelles coding for Arg have 7 x increased risk as it is more susceptible to degradation by HPV E6

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

What is apoptosis?

DEFINITELY IN CURRICULUM

A

Highly regulated process of programmed cell death, active process. ATP dependent.

  • Highly regulated, not associated with an inflammatory response
  • Reduction of cellular/nuclear volume
  • Chromain condesation/DNA ladder formation - multiples of 80bp
  • Nuclear fragmentation and plasma blebbing
  • Cell contents packaged in membrane bound bodies
  • Engulfment by phagocytes - The exposure of phosphatidylserines (phospholipids) on the exterior of the plasma membrane is the signal that initially recruits phagocytes. Ordinarily, phosphatidylserine is sequestered on the inner leaflet of the phospholipid bilayer and is not displayed on the cell’s surface. Macrophages then produce antiinflammatory TGFbeta to ensure no inflammatory response is produced.
  • 2 pathways - Intrinsic (mitochondrial driven) and Extrinsic (receptor driven signalling)
  • Both mediated by Caspases (Cysteine-rich ASpartate ProteASES):
    o first synthesised as inactive Procaspases – activated by cleavage of Aspartate residues, then cause
    further cleavage&raquo_space; cascade of amplification&raquo_space; amplification of apoptosis signal
    o Initiator Caspases = 2/8/9/10, Executioner Caspases = 3/6/7 (note Caspase 3 serves as a paracrine signal from dying cells to stimulate proliferation of surviving cells)
  • Can be stimulated by extracellular signals - “death factors” or by intracellular insults – DNA damage/oxidative
    stress
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What is necrosis?

DEFINITELY IN CURRICULUM

A

Sloppy process whereby cells swell, cell membranes become leaky and cells spill out contents into surrounding tissue and cause inflammation. Uncontrolled, in response to trauma. Swelling of cytoplasmic organelles. Moderate chromatin condensation

Necroptosis: Necroptosis is a programmed form of necrotic cell death. Necrotic cell death has been considered a form of passive cell death. However, the discovery that TNF-alpha mediated necrosis can be inhibited by a specific inhibitor of RIP1 kinase, necrostatin-1, led to the concept of necroptosis. Necroptosis has now been established as a regulated necrotic cell death pathway controlled by RIP1 and RIP3 kinases. Under conditions that are insufficient to trigger apoptosis, TNF-alpha activates TNFR1 and in turn induces the recruitment of RIP1 kinase and other proteins to form complex I. Subsequently, these proteins dissociate from TNFR1 and RIP1 can be found in the cytosol in complex IIb, which includes RIP1 and RIP3. The formation of complex IIb leads to necroptosis

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

What are caspases?

A

Specific proteases that act like molecular scissors to cleave intracellular proteins at aspartate residues

he initiator caspases (caspases-2, -8, -9 and -10), which activate the downstream caspases, and the executioner caspases (caspases-3, -6 and -7), which cleave cellular substrates.

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

Describe the extrinsic pathway of apoptosis?

DEFINITELY IN CURRICULUM

A

Death factor (eg Fas ligand/TNF) -> binds to transmembrane death receptors (Fas/TNF-r). FAS is transmembrane on T cells, TNFa is free.
Receptors form homotrimers undergo conformational change and expose intracellular death domains.

Intracellular adaptor proteins (FADD/TRADD) transduce signal to caspases
Recruit pro-caspase 8 via death effector domains.
Pro-caspase 8 activate when close together by self cleavage
CASPASE 8 IS INITIATOR -> cascade of caspases -> executioner caspases (3,6,7)
Proteolysis of target proteins
(caspases also cleave Rb suggesting it may have a role in inhibiting apoptosis)

A death factor (e.g. FAS ligand) bound to plasma membrane of another cell or a soluble factor (e.g TNF or
TRAIL) is received by a transmembrane death receptor/TNF receptor
Receptor undergoes conformational change (e.g. trimerisation) which exposes the intracellular Death Domain
- This allows intracellular adaptor proteins e.g. FAS associated death domain (FADD) protein or TNF-receptor
associated death domain (TRADD) protein to bind to Death Domain and recruit Procaspase 8 (The ligandreceptor-death domain protein-Procaspase 8) complex is called a Death Inducing Signalling Complex - DISC
- Self cleavage and activation of Procaspase 8&raquo_space; Caspase 8&raquo_space; cascade of Caspase activation&raquo_space; activation of
executioner Caspases 3/6/7
- Inhibited by c-FLIP which blocks Procaspase 8 recruitment and activation

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

What is the initiator caspase in extrinsic apoptosis?

DEFINITELY IN CURRICULUM

A

Caspase 8

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

What inhibits extrinisic apoptosis?

DEFINITELY IN CURRICULUM

A

c-Flip

Binds to FADD or caspase 8 and inhibits caspase 8 recruitment and activation

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

Describe the process of intrinsic apoptosis?

DEFINITELY IN CURRICULUM

A

Stimuli inside the cell eg DNA damage
Induced via Bcl-2 family which act at outer mitochondrial membrane.
One group of Bcl proteins inhibit and one group promote apoptosis.
On activation of BH3 only proteins, Bid and bim activate Bax -> translocates to mitochondrial membrane -> oligomerises into the membrane -> causes membrane to become more permeable and release apoptotic mediators -> cytochrome c joins to Apaf-1 and recRuits pro-caspase 9 to form apoptosome 7 spoked wheel of death -> activates caspase cascade
Smac/DIABLO released from mitochondria inhibit IAPs that normally block caspases.

  • Regulated by BCL-2 family which are divided into two groups with opposing functions:
  • Pro-apoptotic: Bax, Bak and also BH3-only proteins: Bim, Bid, Bax, Bak, Noxa, Puma
    o These stimulate pore formation on Mitochondrial membrane through which Cytochrome C and
    Procaspase 9 are relseased
    o Apoptotic signal&raquo_space; BIM + BID bind to and activate BAX&raquo_space; conformal change in BAX&raquo_space; BAX monomers
    insert into Mitochondrial membrane&raquo_space; form oligomers&raquo_space; pore formation
  • Pro-survival: BCL-2, BCL-xl, BCL-W, MCL-1
    o These bind and sequester Pro-apoptotic factors to inhibit apoptosis
    o Pro-survival factors e.g. BCL-xl cause dissociation of BAX oligomers
  • Cytochrome C release triggers Caspase activation and formation of Apoptosome (Seven-spoked wheel of
    death:
    o Spokes: APAF-1 (bind to Procaspase 9 via CARD domain)
    o Tip of spokes: Cytochrome C
    o Dome (Hub): Procaspase 9
    o Binding of APAF-1 to Procaspase 9&raquo_space; activation to (initiator) Caspase 9&raquo_space; begins caspase cascade&raquo_space;
    activation of (executioner) Caspases 3/6/7
  • Inhibitors of apoptosis (IAPs) can bind and inhibit activated caspases
  • Smac/DIABLO, also released from mitochondrion inhibit IAPs
  • Intrinsic pathway triggered by DNA damage OR damage to plasma membrane – radiation activates
    sphingomyelinase on plasma membrane&raquo_space; generates ceramide&raquo_space; second messenger to activate apoptosis via mitochondria
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

How do the anti-apoptotic proteins work in intrinsic apoptosis?

DEFINITELY IN CURRICULUM

A

Cause dissociation of Bax oligomers from the mitochondrial membrane

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

What are the pro apoptotic members of the Bcl family?

A

BH3- only, pro apoptotic

  • Bad
  • Bik
  • Bid
  • Bim
  • Noxa
  • Puma

Pro apoptotic other members

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

What are the anti- apoptotic members of the Bcl family?

A
Bcl-2
Bcl-w
A1
Boo
Mcl-1
Bind to and inhibit pro-apoptotic proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What is the process of release of apoptotic mediators form mitochondrial membrane called?

DEFINITELY IN CURRICULUM

A

MOMP

Mitochondrial Outer Membrane Permeabilisation

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

How is the extrinsic and intrinsic pathway of apoptosis linked?

DEFINITELY IN CURRICULUM

A

Caspase 8 (extrinsic pathway) can cleave and activate Bid -> stimulate intrinsic pathway via Bax.
ATM kinase phosphorylates Bid and is required to cause cell cycle arrest

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

How is p53 involved in apoptosis?

DEFINITELY IN CURRICULUM

A

p53 functions both in the nucleus and cytoplasm by transcription dependent and independent means.
FUNCTIONS LINKED TO PUMA
- p53 can repress expression of anti-apoptotic factors (Bcl-2, IAPs)
- PUMA (p53 upregulated modulator of apoptosis) - member of Bcl-2 family is essential for p53 induced apoptosis
p53 activates PUMA which acts as an enabler for release of Bcl-x from p53 so p53 can activate Bax

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

Are cancer cells “closer” to apoptosis?

DEFINITELY IN CURRICULUM

A

Yes. Cancer cells are in a pro-apoptotic state that is inhibited by IAPs, whereas normal cells are in a non-apoptotic state that require caspase activation to initiate apoptosis.

TNF-related apoptosis-inducing ligand (TRAIL/APO-2L) is a member of the TNF family that promotes apoptosis by binding to the transmembrane receptors TRAIL-R1/DR4 and TRAIL-R2/DR5. Its cytotoxic activity is relatively selective to the human tumor cell lines without much effect on the normal cells. Potential target for drugs.

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

How can cancer alter the extrinsic pathway of apoptosis?

DEFINITELY IN CURRICULUM

A

Sunburn/UV causes clustering of Fas death receptors and activation of caspase cascade (mutation in Fas-r may lead to increased risk of skin cancer)

Loss of caspase 8 in SCLC and neuroblastomas

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

How can cancer alter the intrinsic pathway of apoptosis? How does this affect treatment?

DEFINITELY IN CURRICULUM

A

More common than extrinsic pathway
Mutations in p53/MDM2/ATM/Chk2
Bcl-2 translocation t(14,18) - B cell lymphoma
Loss of BH3-only proteins e.g. deletion/mutation.
Mutations in bax and bid (Bax >50% mutated in colorectal)
Apaf-1 (co-activator of caspase 9) mutated and repressed in mets melanoma
XIAP induced leukaemia, lung, prostate cancer (supress caspase 9/3/7)

MAKE CANCERS RESISTANT TO CHEMOTHERAPY
eg loss of bax makes resistant to 5FU in colorectal

PI3K:
- Activation of PI3K&raquo_space; activates AKT, AKT blocks pro-apoptotic BIM + BAD
- Therefore PI3K activation inhibits apoptosis&raquo_space; increased risk of proliferation and cancer

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

What are alternative death pathways that are caspase independent and what do they involve?

A

Use alternative proteases eg calpains, cathepsins, serine proteases
eg autophagy, mitotic catastrophe

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

What are cancer stem cells?

DEFINITELY IN CURRICULUM

A

Rare cells within a tumour that have the ability to self renew and give rise to phenotypically diverse cancer cells with limited replicative potential that make up the rest of the tumour. This self renewal provides an extended window for mutations. Cancer stem cells are heterogeneous but are generally more radioresistant and chemoresistant than the more proliferative tumor cell populations, which can be attributed to high anti-oxidant levels, slow cycling, mdr1 expression, and reprogramming. Cancer stem cells appear to have higher levels of free radical scavengers, abnormal activation of developmental pathways, hyperphosphorylation of checkpoint kinases which collectively tend to drive their superb resistance to radiation.

Cancer stem cells = clonogenic cells that survive treatment (can be selected for by chemo/radiotherapy)
They:
- Are capable of self-renewal
- Have differentiation capacity
- Can continually sustain the tumour
- Are resistant to treatment – e.g. by DNA repair, drug efflux, anti-apoptotic
- Are responsible for recurrence

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

How is it proposed that the dergulation of self renewal of stem cells occurs in cancer?

A

Normal stem cells maintain a balance between self renewal and differentiation. Loss of balance can lead to unregulated self renewal.
Alternatively differentiated cells may acquire a mutation which reactivates self renewal programme

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

What is wnt signalling pathway?

A

evolutionarily conserved cell-cell communication system that is important for stem cell renewal, cell proliferation and cell differentiation both during embryogenesis and during adult tissue homeostasis.
19 members of Wnt proteins - INTER cellular signalling molecules.
Lipid modficiations of Wnt by protein porcupine play a role in its secretion from cell.

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

Describe the Wnt pathway when no Wnt ligand present?

A

In cytoplasm (intracellular)

  • Several proteins (APC, Axin, glycogen synthase kinase & casein kinase) aggregate together to form a degradation complex
  • complex targets β- catenin (a transcriptional co-activator) for degradation by phosphoylating and ubquintinating it.
  • In absence of β-catenin transcription is repressed by transcriptional repressor Groucho
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

Describe the Wnt pathway when Wnt ligand is present?

A

Wnt (extracellular) binds to its transmembrane receptor (can be G protein coupled & TKRs) called Frizzled.
Frizzled co receptor LRP undergos change and its cytoplasmic tail is phosphorylated by GSK3 &CKI)
This allows β-catenin to escape from degradation complex and move to nucleus
Acts as a co-activator (with Bcl-9 & pygopus) of T cell factor/LEF family of transcription factors (eg c-myc, cyclin D)

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

How can Wnt1 be affected by cancer?

A

Wnt1 is a proto-oncogene

Mutations constiutively activated Wnt pathway

154
Q

Which cancer is most likely to be caused by mutations in Wnt pathway?

A

Colorectal , >90%

Most mutations inactivate APC or activate β-catenin (rarely affect Wnt)

155
Q

What is familial adenomatous polyposis?

A

Genetic Abnormality: autosomal dominant – mutation of APC tumour suppressor gene (usually truncating mutation) chromosome 5q21. Activates Wnt pathway
Mechanism of Action: APC binds and regulates B-catenin activity, unregulated B catenin&raquo_space; constitutive activation of Tcf = uncontrolled growth and proliferation
Associated cancers: Multiple adenomatous polyps from adolescence – 100% risk of colorectal cancer. Have prophylactic colectomy before age 30.
Loss of APC (hyperplastic)&raquo_space; DNA hypomethlylation (early adenoma)&raquo_space; K-Ras activation (intermediate adenoma)&raquo_space; LOH 18q ?TSG (late)&raquo_space; loss of p53
(carcinoma)

polyps + medulloblastoma = turcot syndrome; polyps + benigns omas e.g. osteomas = gardner syndrome.

156
Q

What is the role of the hedgehog signalling pathway?

A

Role in embryonic development, tissue self renewal and carcinogenesis
Cilia

157
Q

Describe hedgehog family and the transmembrane receptors with which they interact?

A

3 members Hh family: Sonic, Desert and Indian = INTERcellular signalling molecules

2 transmembrane receptors: patched and smoothened (related to Frizzled)- responsible for signal transduction by Hh- interaction regulated within cilia.

158
Q

Describe the hedgehog signalling pathway is absence of any hedgehogs?

A

Patched is localised in the cilia (at top) and inhibits smoothened (from coming to top of cilia)

Gli (zinc fingered transcription factor) - sequestered by a protein complex in cytoplasm which induces cleavage of Gli by proteosomes -> results in a repressor -> nucleus and inhibits transcription of Hh target genes

159
Q

Describe the hedgehog signalling pathway in presence of hedgehogs?

A

Sonic, desert or Indian bind to patched
Patched is translocated from cilia allowing smoothened to relocate there.
Smoothened tranduces a signal into cell - causes large protein complex to dissociate and release Gli
Gli moves to nucleus and regulates expression of Hh genes (VEGF, cyclin D, Bcl2, Snail)

160
Q

How is the hedgehog signalling pathway related to cancer?

A

Patched is a tumour suppressor gene

161
Q

What does a mutation in patched cause?

Not in curriculum

A

Gorlin syndrome- germline mutation in one copy of patched -> BCC, medulloblastoma, rhabdomyosarcomas

All sporadic BCCs
30% sporadic medulloblastomas

162
Q

What is organotropism?

DEFINITELY IN CURRICULUM

A

Specific cancers metastasise to particular sites
Many can be explained by direction of blood flow
1/3 cannot > seed and soil

Seed & Soil Analogy: Seeds are carried in all directions but can only live and grow if they fall on congenial soil:
- E.g. Spleen and Kidneys – rich blood flow but rare sites of metastases
- E.g. Breast and prostate metastasise to bone more than expected
- E.g. Unusual sites for metastaes such as Ocular Melanoma to Liver

163
Q

How do tumours spread? What are the 3 theories of metastatic potential?

DEFINITELY IN CURRICULUM

A

Can be monoclonal (seeded by one cell)
Or polyclonal (seeded by two or more cells)
Can also be seeded from other metastases

  1. Classical theory: clonal selection during cancer progression
    * Primary tumour heterogenous, cells seeded continuosly, cells may not initially have full metastatic potential but can remain dormant and evolve with additional genetic/epigenetic changes independent of but parallel to primary tumour progression
    * Primary tumour can seed >2 mets at once and mets can further seed mets
  2. Predetermined genetic predisposition theory
    * Metastatic potential is an intrinsic property of each tumour from the start – therefore should be predictable from the primary tumour and detectable at first diagnosis – thus aiding management
    decisions
  3. Host predisposition theory
    * Host genotype (and microenvironment) is predisposed to be metastases permissive
164
Q

How do cells break free from normal molecular constraints in order to metastasise?

DEFINITELY IN CURRICULUM

A

EMT- epithelial-mesenchymal transition. Transient loss of closely connected epithelial characteristics and acqusition of a more motile (fibroblast-like), mesenchymal phenotype. An important process in embryonic development

As a result, cancer cells are more easily able to cross the extra-cellular matrix and basement membrane borders
between organs

165
Q

What is EMT characterised by?

DEFINITELY IN CURRICULUM

A

Loss of polarity and cytoskeleton rearrangements

Deconstruction of epithelial cell-cell junctions

Changes in cell shape

Downregulation of epithelial markers eg e-cadherin - the predominant cell-cell adhesion molecule which acts as a TSG by securing cell-cell adhesion and suppressing metastases. EGFR binds SRC via its SH2 domain; SRC promotes motility; FAK activates SRC; The (FAK-SRC)- complex breaks E-Cadherin-Catenin bonds

Upregulation of mesenchymal proteins eg n-cadherin and vimentin

Secretion of specific proteases

Increased motility and invasive potential, increased matrix deposition and resistance to apoptosis

166
Q

How is EMT induced?

DEFINITELY IN CURRICULUM

A

Can be induced via a variety of transcription factors which can be activated via growth factors (eg HGF, EGF, PDGF, TGF) via MAPK, PI3K pathway

Ultimately specific transcription factors eg Twist, snail, slug (Twist is a
key regulator of metastasis)

Reversed once tumour cell establishes secondary sites

Key regulators:
o Envirnomental factors e.g. hypoxia and inflammation
o Wnt, Notch and Hedgehog pathways

167
Q

What anchors cells in place extracellularly and what is the their intracellular component?

A

CAMS and cadherins
Cadherins are calclium dependent transmembrane glycoproteins that have an extracellular hook and bind to other caherins on cell next to them.
They interact via their intracellular catenins.
Catenins can also bind transcription factors and induce gene expression in nucleus

168
Q

What is the predominant CAM in epithelial cells?

A

E-cadherin - secure cell-cell adhesion and suppresses metastasis.
Mutations of extracellular domain and methyltion in promoter region of E-cadherin gene have been identified in gastric/prostate cancer

169
Q

What are integrin receptors?

A

Integrins – cell interactions with the extracellular matrix e.g. collagen/laminins/fibronectins. Inhibit anoikis (apoptosis) when bound to ECM.

As well as breaking free from caherins. Cells must break free from normal molecular constraints by ECM and avoid anoikis.
Integrin receptors are a family of >24 heterodimers made up of a range of alpha and beta subunits that mediate cell-ECM interactions.
They also help with some inside -> outside and outside -> inside signalling. Induce conformational change in the extracellular domain changing the affinity of integrins for their ECM ligands

170
Q

What is involved in the ECM?

A

ECM components eg collagen, fibronectin, laminin
Recognised by integrin receptors
Upon ligand binding the integrins clusster in the membrane and affect the cytoskeleton through interaction with actin-binding proteins and kinases like focal adhesion kiase(FAK)

171
Q

How are integrin receptors changed in cancer?

A

Altered expression of receptors observed in tumour cells during EMT. Have to avoid integrin-induced anoikis in response to loss of ECM contact.

172
Q

What is the role of serine proteases and MMPs?

DEFINITELY IN CURRICULUM

A

Invasion of tumour cells into surrounding tissue requires the action of specific proteases that degrade a path through the ECM and stroma
MMPs can cleave the extracellular domain of E-cadherin and so contribute to loss of epithelial cell-cell junctions.

Tumour cells produce proteases and induce surrounding stromal cells to do so also

Plasminogen and Collagenase also involved in ECM degredation

173
Q

What regulates MMPs?

A

Carefully regulated as they are synthesised as latent enzymes requiring cleavage.
TIMPs regulate their function. Tissue inhibitors of metalloproteinases (TIMPs)

Extracellular matrix metalloproteinase inducer (EMMPRIN) is upregulated on membrane of tumour cells and induces production of MMPs in adjacent stromal cells.

174
Q

Describe the process of intravasation of a tumour cells?

DEFINITELY IN CURRICULUM

A
  1. Cell must attach to the stromal face of vessel
  2. Degrade basement membrane (using MMPs/serine proteases)
  3. Pass between endothelial cells (transendothelial migration) into the blood stream. (tumour associated neovascularisation results in leaky tortuous blood
    vessels allowing easy access)

tumour microenvironment of metastasis doorway (perivascular macrophage activated by tumour cell via colony stimulating factor-1) which activates tumour cell via EGF secretion – migrates along collagen fibres towards blood vessel following the HGF gradient from endothelial cells (chemotaxis) – once gets to the endothelial cell VEGF released by macrophages induces vascular permeabilization and tumour cell gets through

175
Q

What helps to guide the tumour cells into the vessels? How do they do this?

DEFINITELY IN CURRICULUM

A

Tumour associated macrophages
Involves colony-stimulating factor 1 (CSF 1) receptor on macrophages and EGFR on tumour cells.
Macrophage associate with blood vessels and produce EGF -> binds to EGFR on tumour cells -> tumour cells produce CSF 1 which interacts with macrophages and leads to chemotaxis-mediated co-migration along HGF gradient.

176
Q

What are CTCs?

A

Tumour cells within the bloodstream

Can travel singly or in clumps together with platelets as emboli

177
Q

Describe the process of extravasation of a tumour cell?

DEFINITELY IN CURRICULUM

A
  1. Tumour cell must attach to the endothelial side of the blood vessel. tumour cells bind to endothelial cells based on their selectins (Different endothelial cells have different selectins – seed and soil) and do transendothelial migration).
  2. Pass through the endothelial cells and basement membrane (transendothelial migration). Proteases break BM.
  3. Migrate into the surrounding stroma
  4. Either grow or remain dormant depending on metastatic niche
178
Q

How do CTCs bind to the endothelium?

A

E- selectin is important
Binding to endothelium via e-selectin receptors on endothelium mediates adhesion and also triggers a signal cascade by activating stress-activated protein kinase 2 (SAPK2) - necessary for transendothelial migration.

179
Q

What are DTCs?

A

Disseminated tumour cells

cells that have spread but have not yet colonised.

180
Q

What is the pre-metastatic niche?

DEFINITELY IN CURRICULUM

A

A site of future mets that is alerted as a result of factors released by primary tumour in preparation for the arrival of tumour cells.
Supports the seed and soil theory

Pathfinder cells (systemic mediators) e.g VEGF, TGF-B, precondition the distant soil ready for arrival of metastatic cells

181
Q

What are exosomes and how can they contribute to a pre-mestastatic niche?

A

Small vesicles that carry protein and nucleic acids which can travel in the circulation.
They can carry DNA, RNA and protein to cells to which they can fuse- called horizontal transfer -> role in promoting mets.
Can provide “education” to and change behaviour of receiving cells.

182
Q

What are metastatic suppressor genes?

A

Definited by ability to inhibit overt metastases without affecting growth of primary tumour
23 identified
- NM23
-MKK4
Both promote dormancy/apoptosis of micromets

183
Q

Name some pro angiogenic factors?

A

Non specific growth factors
- EGF, HGF, FGF, PDGF

Vascular endothelium specific growth factors

  • VEGF & VEGF-R - MAIN PLAYER
  • angiopoeitins
  • Tie receptors
184
Q

Describe the VEGF family

DEFINITELY IN CURRICULUM

A

5 members
VEGF-A-D and placental growth factor (PIGF)

3 VEGF tyrosine kinase receptors
- VEGFR -1, -2, -3

(VEFG-A/VEGFR-2) complex key to angiogenesis

185
Q

What does VEGFR-1 do?

DEFINITELY IN CURRICULUM

A

VEGFR-1 acts as a decoy by regulating the amount of VEGF-A available to VEGFR-2 as the binding affinity for VEGFR-1 is higher but its kinase activity lower so it restricts angiogenic response. Competitive inhibition.

186
Q

What does VEGF-C bind to and whats its role?

DEFINITELY IN CURRICULUM

A

Binds to VEGFR-3 and plays a role in lymphangiogenesis.

VEGFC produced by tumours so promotes expansion of lymphatic vessels in the tumour periphery, increasing risk of lymphatic spread

187
Q

Describe the signal transduction when VEGF-A is made.

DEFINITELY IN CURRICULUM

A

VEGF-A produced by tumour cells and can be induced in surrounding normal cells.

  • One VEGF-A molecule Binds to two VEGFR-2 receptors -> dimerisation and phosphorylation.
  • intracellular proteins containing SH2 domains bind to phosphorylated receptor -> trigger
    • Ras-Raf-MAPK pathway - stimulated VEGF genes
    • PI3K pathway - AKT leads to inhibition of apoptosis (by inhibition of BAD)and production of endothelial nitric oxide synthase (eNOS) that stimulates endothelial permeability

VEGF responsive genes include EGFR ligand, epiregulin, COX2, MMP1, MMP2

188
Q

Name some angiogenic inhibitors?

DEFINITELY IN CURRICULUM

A

Angiostatin (binds annexin II)
Endostatin (blocks MAPK and MMPs)
Prolactin
P53
Thrombospondin-1, -2
Notch
Top1

189
Q

How is angiostatin produced and what is its role?

A

Plasminogen can be cleaved by proteinases including several MMPs to release angiostatin.
Angiostatin binds to endothelial cell surface receptor, annexin II to exert its inhibitory effects on angiogenesis

190
Q

How is endostatin produced and what is its role?

A

Fragment of collagen XVIII
Can be proteolytically released by elastase and cathepsin
Blocks MAPK activation in endothelial cells and also MMPs

191
Q

Why might surgery removing the primary tumour cause domant mets to grow?

A

? production of anti-angiogenic factors by certain tumours inhibits growth of micro-mets

Surgery induces angiogenic growth factors

192
Q

What is HIF?

DEFINITELY IN CURRICULUM

A

HIF is a heterodimic transcription factor comprising of one HIF-1α and HIF-1β
Activity of HIF is regulated by oxygen concentration, not mRNA expression via HIF-1α

HIF is a driver of tumour progression – transcriptionally activates genes linked to invasion, metastases and resistance. HIF regulates
glycolysis (e.g GLUT-1), angiogenesis (e.g. VEGF), erythropoiesis (e.g. EPO), and pH (e.g. CA9).

193
Q

What happens to HIF under normoxic conditions?

DEFINITELY IN CURRICULUM

A

HIF-1α hydroxylated by prolyl-4 hydroxylase - acts as a direct oxygen sensor by linking molecular oxygen to specific proline residues
VHL binds to hydroxylated HIF-1α and activates proteins which ubuitinate it -> degraded -> target genes are not activated

194
Q

What happens to HIF under hypoxic conditions?

DEFINITELY IN CURRICULUM

A

Enzymes prolyl-4 hydroxylase is inactivated.
HIF-1α is rapidly stabilised and transported to nucleus
Heterodimeric HIF transcription factor can activate its target genes via hypoxia response element (HRE)
Most notable target is VEGF gene which has a HRE in its promoter region

195
Q

What happens to HIF-1α in kaposi’s sarcoma?

A

HIghly vascularised tumour caused by herpes virus
three protein products of viral genome increase HIF-1α half life, nuclear localisation and transactivation under normoxic conditions mimicking hypoxia.

196
Q

30 oncoproteins have been shown to tip balance towards angiogenesis, can you name the star players?

A

EGFR - receptor tyrosine kinase
Src - intracellular tyrosine kinase
Ras - intracellular transducer
Fos and Jun - transcription factors
p53- normally binds to and activates the promoter of thrombospondin- 1 gene (anti-angiogenic), when p53 is mutated it does not activate it

197
Q

Describe the process of angiogenic sprouting?

DEFINITELY IN CURRICULUM

A

In response to angiogenic signals, endothelial cells extend filopodia and migrate towards signal
At location of highest concentraion of VEGFA, VEGFR-2 is activated.
Signal enhanced by co-receptor neuropilin-1 (Nrp-1) and transduced via MAPK cascade
Stimulates formation of tip cell at forefront of a sprout
Behind tip cell are the proliferating stalk cells that extent the sprouting vessel
Upon VEGFR2 activation tip cells produce and release Notch ligand called Delta-like 4 (DLL4)
DLL4 binds to notch receptor on neighbouring cells -> NICD (notch intracellular domain) is released intracellularly -> travels to nucleus and represses VEGFR2 expression and increases VEGFR1 expression.
VEGFR-1 reduces concentration of VEGF available for VEGFR-2 therefore the growing sprout moves along a VEGF gradient

198
Q

Name some carcinogenic contaminants on foods?

DEFINITELY IN CURRICULUM

A

Farmed salmon - contains polychlorinated biphenols and other pesticides, eating >once a month increases cancer risk

Red meat - cooking at high temps produces heterocyclic aromatic amines -> metabolised by p450 N-acetyltransferase which turns to carcinogen ->DNA adducts -> base substitutions and mutations. N-acetyltransferase polymorphisms affect colon cancer risk.

Peanuts-> contaminated with aflatoxin B, a fungal product of aspergillus flavus -> cause GC->TA transversions -> HCC

Nitrosamines in food preservatives such as sodium nitrite -> O6 guanine alkylation.

199
Q

What is folate and why is it so important?

DEFINITELY IN CURRICULUM

A

Vitamin B9
CAn accept or donate one carbon units in metabolic reactions
Critical co-enzyme for nucleotide synthesis and DNA methylation (methionine ->SAM)- depletion can interfere with this and cause cancer (associated with colorectal cancer).

200
Q

What happens to DNA synthesis in a person with folate def?

DEFINITELY IN CURRICULUM

A

dTMP synthesis is inhibited in a low folate state and imbalance of nucelotides results in incorporation of uracil into DNA -> DNA strand breaks trying to repair this

Also get disruption in DNA methylation may cause genomic hypomethylation
- methyl groups used for methylation are supplied by folate -> decrease in synthesis of methionine and so genomic hypomethylation

MUTATION AND HYPOMETHYLATION

201
Q

Why does obesity cause cancer?

DEFINITELY IN CURRICULUM

A

Adipose tissue is an endocrine tissue that can release free fatty acids, peptide and steroid hormones. Obesity is associated with breast, endometrial, colon, kidney, liver, pancreas and oesophageal cancer.

ALTERED SEX HORMONE METABOLISM

  • synthesis of oestrogen from androgens using aromatase
  • elevated cholesterol - 27-hydroxycholesterol is a ligand for oestrogen receptor

INCREASED PRODUCTION OF FAT CELL HORMONES DUE TO ADIPOCYTE HYPERTROPHY - ADIPOKINES
- causes chronic inflammatory response -> IL6, TNF

INCREASED INSULIN SIGNALLING PATHWAYS
- insulin resistance increases insulin levels and IGFs
- promotion of proliferation and inhibition of apoptosis

DIETARY ALTERATIONS OF GUT MICROBIOTA (dysbiosis)
- promotes liver cancer -> more gram +ve => causes increase in bacterial metabolite deoxycholic acid made from cholesterol-derived bile acids -> chronic DNA damage in liver as human liver cannot metabolise it

ASSOCIATED WITH NAFLD -> HCC probably due to cirrhosis/inflammation

(Dietary effects on cancer: caloric restriction reduces cancer. High fat high carb diets = cancer. High fibre = reduced colorectal cancer)

(Physical activity also associated with reduced cancer - perhaps due to improved insulin resistance.)

202
Q

Why does alcohol cause cancer? Which cancers does it cause?

DEFINITELY IN CURRICULUM

A

Metabolised by enzyme alcohol dehydrogenase to form acetaldehyde -> binds to DNA -> forms adducts
Get more acetaldehyde in saliva (10-100x higher) as bacteria in saliva convert it (hence higher risk of mouth cancer).

Acetaldehyde is oxidised by enzymes acetaldehyde dehydrogenase (single nucleotide polymorphism of gene causes intolerance and increased risk of oesophageal cancer)

Alcohol dehydrogenase can metabolise oestrogen - excess alcohol competes with oestrogen for metabolism so more metabolism levels -> breast cancer.

Liver cirrhosis (inflammation) -> HCC

Causes oropharynx, larynx, oesophagus, liver. Increased risk of breast/colorectal cancer

203
Q

How is fruit and veg intake linked to cancer?

DEFINITELY IN CURRICULUM

A

Inverse assoc between total fruit and veg intake and risk of cancer
Microconstituents of food play a role in cancer prevention directly via free radical scavenging or indirectly regulating expression of genes that code for phase I and II reactions

Phase I P450 and II conjugation reactions are major defence mechanism against xenobiotics (foreign substances) - often phase I converts to ultimate carcinogen and phase II allows removal.

204
Q

How does vitamin c help protect against cancer?

A

Water soluble vit C can donate electrons to a free radical (e.g.superoxide, hydrogen peroxide - note hydroxyl radicals are too reactive to be affected by antioxidants)
Oxidised vit c forms ascorbyl radical that is fairly stable and unreactive
Vit C reductase can regenerate vit C from ascorbyl radical for reuse or ascorbyl radical may lose another electron and become degraded
Vit C needs to be replenished daily
Lipid soluble vit E acts as a free radical scavenger in a similar way

Note supplemental vitamin C has no effect on cancer.
Vitamin D also supposed by good for cancer but supplemental vitamin D has no effect. Steroid hormone receptor family.

205
Q

What is the role of antioxidants in cancer?

A

Antioxidant response element (ARE) in promoter region of several genes encoding detoxification and antioxidant enzymes (eg glutathione s-transferase, NADPH:quinone oxidoreductase, phase II reaction enzymes)

KEAP1 inhibits NRF2 by forming complex with ubiquitin ligase. ROS interact with cysteines of KEAP1, stopping it interacting with NRF2. Therefore NRF2 drive transcription of ARE genes. Antioxidants also inhibit KEAP1.

Nrf2 binds to Maf (member of basic leucine zipper family) and activates transcription of detoxification enzymes via ARE

Antioxidants:

  • Isopthiocyanates in broccoli
  • EGCG in green tea

Nrf2 antioxidant pathway often upregulated in cancer - perhaps protects cells/DNA from chemotherapy/ROS induced damage.

Therefore Nrf2 pathway is to inhibit carcinogenesis but gets hijacked by cancer cells to protect themselves and promote tumour survival.

206
Q

What is the warburg effect?

A

Observation that cancer cells carry out aerobic glycolysis, converting glucose to lactate in presence of oxygen.

207
Q

Normally in aerobic conditions how does a cell make ATP?

A
  1. Glucose undergoes glycolysis in cytoplasm to pyruvate x2
  2. Pyruvate converted to acetyl coA by pyruvate dehydrogenase - oxidative process, NADH and CO2 formed.
  3. Citric acid/Krebs cycle - series of reactions breaks down acetyl coA - > forms ATP, NADPH, FADH2
  4. Electron transport chain - series of mitochondrial membrane proteins that transfer electron donors to electron receptors
208
Q

What is the bodies 1st, 2nd and 3rd line defense of pathogens?

DEFINITELY IN CURRICULUM

A

1st line = skin and mucous membranes

2nd line = no memory, no prior exposure required, quick, non-specific - innate immune system

  • macrophages - phagocytose foreign substances + dead host tissue - constantly processing their microenvironment - present antigens via MHC class II
  • NK cells - targets abnormal host cells e.g. tumour, virus-infected - cytotoxicity through granzymes and inflammation through interferon gamma
  • neutrophils (granulocyte and phagocyte - target bacteria)
  • eosinophils (granulocyte - parasite)
  • basophils (granulocyte - allergy e.g. hayfever/anaphylaxis)
  • dendritic cells (APCs to activate the adaptive immune system) - links the innate and adaptive immune system
  • inflammation
  • complement
  • cytokines

3rd line = specific and adaptive; recombination required; has memory

  • b cells - antibodies (humoral immunity)
  • t lymphocytes - CD4 T helper (activates B cells and CD8 cells) and CD8 T killer cells (cell mediated immunity - infected/abnormal host cells through granzymes/perforin/granulysin) and CD4 T reg cells (immunosuppressive)
  • CD4 cells activated by antigen presenting cells - dendritic cells and macrophages
209
Q

What are pattern recognition receptors PRRs?

A

Pattern recognition e.g. toll like receptors
Mainly found on antigen presenting cells (APCs) like dendritic cells, monocytes, NK cells, also found on other non-immune cells.

Recognise PAMPs (pathogen assoc molecular patterns) and TFs
INNATE IMMUNITY
When PAMP binds to PRR -> recruitment of APCs, upregulation of MHC and secretion of cyto/chemokines

210
Q

What happens in complement cascade?

A

Innate immunity

Classical pathway -> activated by antigen/antibody complexes -> starts at C1

Alternative pathway -> activated by cell membranes -> starts at C3b

Lectin pathway -> activated by carbohydrate structures -> starts at MBL

OUTCOMES:
C3b -> causes OPSONISATION (phagocytosis)
C3a & C5a -> inflammation, mast cell activation/chemotaxin
MAC -> LYSIS

211
Q

What causes opsonisation

A

Activation of complement cascade. Opsonisation = tagging a target with an opsonin allowing it to be phagocytosed by phagocytes.

Specifically C3b

212
Q

Name some acute phase proteins and how do they affect the complement system? What causes production of acute phase proteins?

May be in curriculum

A

IL6, IL1, TNF-α -> cause acute phase proteins to be made -> CRP, mannan-binding lectin, serum amyloid protein A

Maximise activation of complement system
CRP and serum amyloid protein A bind to DNA and other nuclear material from cells helping in their clearance

213
Q

What are interferons and when are they produced?

May be in curriculum

A

Produced in reponse to viral infection. Inhibit protein synthesis
Interferon type I (alpha and beta): produced in response to viral molecular patterns e.g. by dendritic cells and macrophages. Antagonise viral replication. Recruit NK cells.
Interferon type 2 (gamma): produced by NK cells.

214
Q

What are lymphokines/cytokines that stimulate lymphocyte growth?

May be in curriculum

A

GF for lymphocytes eg IL2, IL4, influence nature of immune reponse
IL2 and IL9 are produced by T cells to stimulate T cell growth.
IL4 and IL5 are produced by T cells to stimulate B cell growth.

215
Q

What are monokines/cytokines that activate inflammation?

May be in curriculum

A

Critical to immune defence and inflammation eg IL1, TNF, IL6- activate lymphocytes, increase body temp, activate mobilise phagocytes, activate vascular endothelium.
IL1 = fever. IL6 = acute phase proteins. TNFalpha = both, general inflammation.

216
Q

What are chemokines?

May be in curriculum

A

Activate and direct effector cells expressing appropriate chemokine receptors to sites of tissue damage and regulate leukocyte migration to tissues. eg CXCL-8, CCL2

217
Q

What are MHC class I cells?

DEFINITELY IN CURRICULUM

A

Most nucleated cells. Present intracellular antigen (peptide fragments bound to MHC class I in the rough endoplasmic reticulum) to cytotoxic CD8 T cells (via MHC receptor). MHC I – 3 alpha units and a beta-2 microglobulin; MHC II – 2 alpha and 2 beta chains

218
Q

What MHC class II cells?

DEFINITELY IN CURRICULUM

A

B cells, macrophages and dendritic cells. Antigen presenting cells. Present antigen to helper CD4 T cells. Extracellular antigens are endocytosed/phagocytosed and degraded by lysosomal enzymes. Bound to MHC class Ii in the lysosome and transferred to cell membrane. MHC I – 3 alpha units and a beta-2 microglobulin; MHC II – 2 alpha and 2 beta chains

219
Q

What is CD8+? Which cells have it and what does it do?

DEFINITELY IN CURRICULUM

A

CD8 is a transmembrane glycoprotein that serves as a co-receptor for the T cell receptor.
SPECIFIC FOR MHC CLASS I
CD8+ cells become cytotoxic T cells on activation. They turn to memory T cells after activation and clearance of infection. Overtime, if they are not reacted, they lose their function = T cell exhaustion.

220
Q

What is CD4+? Which cells have it and what does it do?

DEFINITELY IN CURRICULUM

A

CD4 is a transmembrane glycoprotein that serves as a co-receptor for the T cell receptor.
SPECIFIC FOR MHC CLASS II

CD4 cells can be T helper cells, monocytes, macrophages, dendritic cells.

221
Q

What encodes the MHC proteins in humans?

DEFINITELY IN CURRICULUM

A

HLA genes on chromosome 6

222
Q

What makes up a T cell receptor?

May be in curriculum

A

2 different protein chains - heterodimer

  • 95% are α/β chains
  • 5% are γ/δ chains

Transmembrane region
Constant region
Variable region at top with antigen binding site

223
Q

What 3 signals does a naive T cell need for activation?

DEFINITELY IN CURRICULUM

A
  1. Antigen presented by APC via MHC recognised by TCR (e.g. MHC class II and CD4)
  2. Co-stimulatory activation by checkpoint molecules eg CD28 (t cell) and B7 (APC) - this is inhibited by CTLA4 on T cells competing to bind B7 instead CD28
  3. Cytokines
224
Q

What inhibitory molecules are activated following naive T cell activation?

DEFINITELY IN CURRICULUM

A

CTLA4- co-inhibitory molecule is rapidly mobilised from intracellular vesicles within T cell membrane
It competes for B7 (CD80/CD86) ligand (on APC) with CD28, it has a higher affinity for it so inhibits it - ipilimumab

PD1 (pembrolizumab/nivolumab) and its ligands PDL1 (atezolizumab/durvalumab) and PDL2 are also co-inhibitory.
Interaction of PDL1 on tumour cells and its receptor on activated effector T cells causes inhibition.

Inhibition is due to recruitment of SHP-2 phosphatases inactivating PI3K and protein kinase C activation. Blocks production and secretion of molecules required for cytotoxic response.

. Naive and memory T cells express high levels of cell surface CD28 but do not express
CTLA4 on their surface. Instead, CTLA4 is sequestered in intracellular vesicles. After the TCR is
triggered by antigen encounter, CTLA4 is transported to the cell surface. The stronger the
stimulation through the TCR (and CD28), the greater the amount of CTLA4 that is deposited on
the T cell surface. Therefore, CTLA4 functions as a signal dampener to maintain a consistent
level of T cell activation in the face of widely varying concentrations and affinities of ligand for
the TCR. By contrast, the major role of the programmed cell death protein 1 (PD-1) pathway is
not at the initial T cell activation stage but rather to regulate inflammatory responses in tissues
by effector T cells recognizing antigen in peripheral tissues. Activated T cells upregulate PD-1
and continue to express it in tissues. Inflammatory signals in the tissues induce the expression of
PD-1 ligands, which downregulate the activity of T cells and thus limit collateral tissue damage in
response to a microorganism infection in that tissue. The best characterized signal for PD-1
ligand 1 (PD-L1; also known as B7 H1) induction is IFNg, which is predominantly produced by T
helper 1 (TH1) cells, although many of the signals have not yet been defined completely.
Excessive induction of PD-1 on T cells in the setting of chronic antigen exposure can induce an
exhaustive or anergic state.

225
Q

What cells have PD1, PDL1 and PDL2 receptors?

DEFINITELY IN CURRICULUM

A

PDL1 - tumour cells and many other types of cells after exposure to interferon gamma.
PDL2 - APCs

PD1 - T cells

226
Q

If there is an intracellular cytosolic microbe - describe the process of presentation? Which MHC class is used?

DEFINITELY IN CURRICULUM

A

Antigen processed - peptides. Taken into the endoplasmic reticulum. Bind to CLASS I MHC -> golgi -> vesicle and transported to cell surface

227
Q

If there is an extracellular antigen in vesicles - describe the process of presentation? Which MHC class is used?

DEFINITELY IN CURRICULUM

A

Antigen taken up into cell via endocytosis
Degraded by phagosomes in vesicle
MHC class II released from ER
Travels to vesicle and combine with antigen
Travels to cells surface and presents it

228
Q

What do antibodies do?

DEFINITELY IN CURRICULUM

A

Bind soluble antigens or can bind antigen on cell surface.
Neutralisation e.g. stopping PD1 binding to PDL1
Opsonisation (Opsonization is an immune process which uses opsonins to tag foreign pathogens for elimination by phagocytes. Without an opsonin, such as an antibody, the negatively-charged cell walls of the pathogen and phagocyte repel each other.)
Complement activation
Antibody-dependent cellular cytotoxicity (granzymes/perforin - NK cells/CD8 T cells)

229
Q

Describe the components of an antibody?

DEFINITELY IN CURRICULUM

A

Constant region (most of the antibody): 2 identical heavy chains linked by a disulphide bond, 2 identical light chains on outer side bound by disulphide bond to heavy chain = these are the same in all types of immunoglobulins.
Variable region is the tip = antigen binding site. Produced by genetic recombination (NHEJ)
Fc = fragment crystallisable = the stick of the Y.
Fab = fragment antigen binding = the arms of the Y = includes the light chains and the antigen binding site.

230
Q

What is IgM, IgG, IgA, IgE, IgD - how are thye made up, where are they important and what do they do?

DEFINITELY IN CURRICULUM

A

IgM (6%) - pentamer- activates complement cascade = initial response
IgG (80%) - monomer - neutralises toxins, opsonisation, complement, binds to phagocytes = memory response
IgA (13%)- dimer- secreted in tears, mucus, saliva
IgE (0.002%)- monomer- allergy, binds to mast cells/basophils
IgD (1%) - monomer- B cell receptor

231
Q

What do B cells do?

DEFINITELY IN CURRICULUM

A

Antigen presented via MHC class II on APC + CD4 T cell cells = activation to plasma cells that produce antibodies. B cells/T cells interaction occurs in lymph nodes. Turns to memory B cell. Produces one type of antibody (multiple B cells target different part of the foreign protein = polyclonal).

232
Q

How do T cells build tolerance?

DEFINITELY IN CURRICULUM

A

Central - thymus - killed if they respond to host tissue too strongly

  • negative selection
  • receptor editing
  • generation of regulatory t cells

Peripheral - suppress autoreactive T cells

  • clonal anergy (unresponsive, prevent overactivation)
  • clonal deletion
  • regulatory t cells
  • T-T cell interactions

Maturation happens in response to antigen

233
Q

What are Treg cells and what is their role in cancer?

DEFINITELY IN CURRICULUM

A

Type of differentiated T helper cell -> T regulatory cell -> influence tolerance and immune suppression
Secrete immunosuppresive cytokines (IL10, TGFb)
Block T cell proliferation, inhibit NK function
High levels of Tregs = poor prognosis in cancer

234
Q

What is immunoediting? What is the immunoscore?

DEFINITELY IN CURRICULUM

A

Elimination -> equilibrium -> escape
Elimination of cancer; equilibrium where less immunogenic cancer cells are selected for; escape where tumour evades immune system. Evidence to support this is that immunodeficient hosts produce more immunogenic tumours than immunosufficient hosts; the number and type of tumour infiltrating lymphocytes gives an immunoscore which correlates with cell survival

General cancer immune avoidance facts: LET survival curve is exponential.
The process by which tumor cells change phenotype in response to an immune response (cytotoxicity or inflammation) in an attempt to avoid recognition (i.e. the induction of PD-1, PD-L1, and IDO following antigen recognition and the production of IFNγ) is characteristic of an adaptive immune response.
Epitope spreading (or antigen cascade, antigen spread, determinant spread) describes a phenomenon where the immune response evolves and expands from focusing on a single antigenic epitope, into a multi-epitopic response be it naturally or following therapeutic intervention e.g. vaccination or radiotherapy. This process is dynamic and may continue to expand over time. Antigen spreading of the anti-tumor immune response from one antigen to another antigen has been linked to superior clinical outcome with the assumption that it counteracts tumor immune evasion. The somatic mutation load of cancers is thought to be a major determinant of response to immune checkpoint blockade through the generation of neoantigens targetable by the immune system.

235
Q

What are T infiltrating lymphocytes and how do they affect prognosis?

DEFINITELY IN CURRICULUM

A

T infiltrating lymphocytes (TILs)

  • can predict favourable outcome in some tumour types
  • infiltrating NK and CD8 T cells assoc with favourable prognosis

o Immune desert (no T cells in tumour); immune excluded (T cells surround tumour but cannot infiltrate for unknown reason); immune inflamed (T cells infiltrate tumour). You want tumour to be immune inflamed to induce tumour death.

236
Q

Why doesn’t our immune system protect us from cancer?

DEFINITELY IN CURRICULUM

A

Peripheral tolerance is required to regulate self reactive T cells and NK cells- these regulatory mechanisms are often exploited by tumours to evade immune recognition

TUMOUR IMMUNE INVASION VIA:

  • MODIFYING MICROENVIRONMENT- secrete immunosuppressive agents eg TGF-B, IL-10, VEGF
  • EVADES T CELL RECOGNITION- downregulates class I MHC (NK cells should still recognise cells that have lost MHC)
  • DYSREGULATION OF ANTIGEN PROCESSING/PRESENTATION PATHWAYS
237
Q

What are good immune cells? (Activating/effector cells)

DEFINITELY IN CURRICULUM

A

CD8 T cells, NK cells, CD4 (Th1), M1 polarised macrophages (encourage inflammation)

238
Q

What are the bad immune cells? (Immunosuppressive cells)

DEFINITELY IN CURRICULUM

A

CD4 Tregs, Myeloid-derived suppressor cells (MDSCs), tumour-associated macrophages (TAM), M2 macrophages (decrease inflammation and encourage tissue repair)

239
Q

What are types of tumour associated antigens?

DEFINITELY IN CURRICULUM

A

Can be unique to cancer- neoantigens e.g. mutated p53 peptides.
Can be overexpressed normal antigens eg HER2
Differentially expressed antigens eg cancer testis antigens which are normally restricted to testis germ cells
Presented to T cells via APCs. Neoantigens will be particularly immunogenic = more T infiltrating lymphocytes.

240
Q

What are dendritic cells?

DEFINITELY IN CURRICULUM

A

Type of APC - constantly sampling environment for foreign substances and activate both innate and adaptive immune system

Can be activated by toll like receptors.
Cause inflammation by releasing cytokines.
Recruit NK cells to do cell mediated cytoxicity via interferons.
Perform phagocytosis to present antigens to T cells.

Have class I and II MHC
Immature dendritic cells - specialised for antigen acquisition, highly phagocytic, low in class II, co-stimulatory, no NK activation

Mature DC: antigen and danger signal specialised for antigen presentation of T cells, less phagocytic, high class II, co-stimulatory molecules, activate NK cells

241
Q

What danger signals activate innate and adaptive immunity?

A

Tumour necrotis cores
Hypoxia
Cell derived danger signals - heat shock proteins, ATP, RNA, DNA, uric acid, hyaluronic acid, NK ligands, IFN-a

242
Q

How does radiotherapy cause immunogenic cell death?

A

senses presence of cytosolic DNA -> activates CGAS -> STING (stimulator of interferon genes)

Inhibited by TREX1

243
Q

What is a tumour assoc macrophage and what does it mean in cancer?

DEFINITELY IN CURRICULUM

A

Poor prognosis
Secrete IL10, TGFb
Enhance tumour proliferation through NFKB/STAT signalling

244
Q

What is the danger hypothesis? What is immunosurveillance?

DEFINITELY IN CURRICULUM

A

Based on idea that the immune system does not differentiate from self/non-self but by things that might cause danger via danger signals. Explains why immunosuppressed patients get more cancers.
Immunosurveillance is a term used to describe the processes by which cells of the immune system look for and recognise foreign pathogens, such as bacteria and viruses, or pre-cancerous and cancerous cells in the body.

245
Q

What is the infectious non-self model?

DEFINITELY IN CURRICULUM

A

That APCs are activated via pattern recognition receptors (PRRs) which recognize evolutionary distant conserved patterns. These pathogen-associated molecular patterns (PAMPs) on such organisms as bacteria are recognized as infectious non-self, whereas PRRs are not activated by non-infectious self.

DOES NOT EXPLAIN ANTI-TUMOUR IMMUNITY

246
Q

What is a myeloid suppressor cell?

A

Necrosis releases endogenous DAMPs (damage-associated molecular patterns), starts the cycle of chronic inflammation and recruits MDSC
Produce NO and ROS may cause DNA damage and mutations
Support tumour progression by increasing angiogenesis, tumour cell stemness, metastasis

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid progenitor cells that fail to differentiate into granulocytes, macrophages, or dendritic cells. These immature cells have the capacity to suppress immune responses mediated by natural killer (NK) cells and NKT cells, as well as CD8+ and CD4+ T cells. MDSCs have been suggested to have a causative role in promoting tumor-associated immunosuppression. They accumulate in the blood, bone marrow, and secondary lymphoid organs of tumor-bearing mice and cancer patients, where circulating levels of these cells have been shown to correlate with clinical stage, metastatic burden, and chemoresistance.

247
Q

What is EBV and how does it cause cancer?

DEFINITELY IN CURRICULUM

A

DNA virus (herpes virus 4)
Causes lymphoma eg Burkitts, nasopharyngeal cancer
EBV encodes several viral proteins that affect host gene expression- oncoprotein LMP1 able to transform cells in culture
Activates inflammatory transcription factors NFKB and Stat and inhibits apoptosis via TRADD

248
Q

What is human papilloma virus and what types cause cancer? How do they do this?

DEFINITELY IN CURRICULUM

A

DNA virus (papovavirus).
HPV 16 & 18 account for 70%
HPV gene products E6 and E7 are major players that target tumour suppressors - both are required to immortalise cells in culture.

  • E7 binds to Rb at HDAC binding site and triggers ubiquitin-mediated degradation of Rb preventing sequestering of E2F -> constitutive action of cyclin A/E

-E6 forms a complex with ubiquitin ligase binds to p53 and targets it for degradation. Also inhibits transactivation activity of p53. Polymorphisms in p53 at amino acid 72 lead to differences in risk of cervical cancer - if both alleles code of Arg they have 7 x risk cervical cancer than those with Pro at site - Arg at 72 is more susceptible to E6 degradation.

Sexually transmitted: Can cause cervical (100% caused by HPV, lag time 10 years), oral (50% caused by HPV), penile, vulval, anal cancer

HPV vaccine gardasil

249
Q

What is HTLV-1? How do you get it and how does it cause cancer? (Human T cell lymphotropic virus type 1)

DEFINITELY IN CURRICULUM

A

RETROVIRUS (RNA virus)
adult T cell leukaemia (2-5% affected individuals get leukaemia) - 100% cases of T cell leukaemia have the virus - prevalent in Japan.
Transmission via breast milk, semen, blood
Genomic RNA copied into DNA by reverse transcriptase before viral proteins are synthesised by the host cells machinery
-TAX protein and HBZ are required to transform T cells in culture - work by protein-protein interactions. TAX activates NFKB pathway. HBZ activates Jun.

To prevent they limit breastfeeding to 6 months in Japan

250
Q

What is hepatitis B and how does it cause cancer?

DEFINITELY IN CURRICULUM

A

DNA virus. Integrates into host genome.
Chronic infection causes HCC - 10-25x risk
Causes cancer via:
- Indirect mechanism: evoking immune response, chronic inflammation - liver necrosis and regeneration
- Direct mechanism: insertional mutagenesis at specific sites-> integration into human telomerase reverse transcriptase (hTERT) gene occurs frequently, results in upregulation due to close proximity with viral enhancer
- Direct mechanism: HBV X protein activates proto-oncogenes and inhibits tumour suppressor genes (main effector) - induces liver cancer in mice.

(Hepatitis C also causes HCC - RNA virus)

hep B vaccine to prevent, check blood products

251
Q

What causes primary effusion lymphomas and Kaposi’s sarcoma and how does it cause them?

DEFINITELY IN CURRICULUM

A

Herpes virus 8 or kaposi’s sarcoma assoc herpes virus
DNA virus
Thought to infect circulating endothelial cells.
Often assoc with AIDs, immunosuppression.
KSHV produces viral cyclin, viral anti-apoptosis proteins eg vBcl2, vFLIP.
Also viral encoded mRNAs and viral protein LANA (main effector) which interferes with p53 and Rb. LANA has same mechanism as HPV.
They require autocrine and paracrine factors for cell transformation - induce neighbouring cells to produce growth factors and cytokines (by releasing paracrine factor vGCPR) which stimulate NFKB and MEK/ERK pathways in the HHV8 infected cells - tumourigenesis by “remote control”.

252
Q

What is the multistep inflammatory process for inducing gastric cancer?

DEFINITELY IN CURRICULUM

A

75% gastric cancers caused by H pylori
Chronic superficial gastritis -> atrophic gastritis -> intestinal dysplasia -> gastric carcinoma.

Gastric atrophy characterised by loss of normal glandular tissue and results in reduced acid production -> other bacteria colonise -> inflammatory response

H pylori eradication - PPI/2 antibiotics

253
Q

How does H.pylori cause cancer? (Molecular mechanism)

DEFINITELY IN CURRICULUM

A

Code for protein called cytotoxin-associated antigen (Cag A)- effector protein injected into cells via integrin receptor -> phosphorylated by tyrosine kinases e.g. Src and ABL -> activates SH2 containing proteins e.g. SHP2 -> MAPK pathway

Unphosphorylated Cag A interacts with e-cadherin causing it to release b-catenin -> nucleus and is transcriptional regulator of genes required for metaplasia

Also H-pylori induced inflammation results in hypermethylation of promoter of e-cadherin, reducing its expression. Also general inflammation will upregulate NFKB etc.

Cag-A expression induced in high salt conditions - high salt diet associated with gastric diet.

254
Q

The inflammatory pathways involved in oncogenesis activate 2 important transcription factors, what are they?
What other ways does inflammation cause cancer?

DEFINITELY IN CURRICULUM

A

JAK/STAT pathway -> induced by IL6 - induces proliferation (cyclin D), survival (Bcl-xl), inflammation (IL6), angiogenesis (VEGF)
NF-κB -> regulate pro-inflammatory factors

Tumour associated macrophages:
Macrophages produce ROS and RNS (Reactive nitrogen species) to help fight infection but can also cause DNA damage
TNF-a can affect cell motility and tumour metastasis -> nitric oxide synthase is stimulated by TNF-a.
IL-6 important in hepatocarcinogenesis - downregulation of IL6 by macrophages in response to oestrogen makes women less susceptible

Inflammation can also attract stem cells e.g. chronic inflammation in prostate cayses recruitment of mesenchymal stem cells.

Induce DNA damage/genomic instability; stimulate cell growth/angiogenesis/EMT/motility/breakdown of ECM. Effect is on tumour cell themselves and stromal cells.

255
Q

What is NF-κB? What is its role in cancer?

May be in curriculum

A

Transcription factor. Produced by macrophages and tumour cells.
Activated by specific inflammatory agents eg bacteria, viruses, cytokines eg TNF-a, stress, hypoxia, smoke, carcinogens, radiation

Causes inflammation (Cox2, IL6, TNFa), promotes metastasis (MMP9, TWIST), angiogenesis (VEGF), promotes proliferation (cyclin D, MDM2) and inhibits apoptosis (bcl-xl, cIAP, c-FLIP)
Target genes : c-Flip, Cox-2, MMP9, VEGF, IL6, TNF-a, cyclin D, MDM2

Normally bound to IκB proteins which inhibits NF-κB, if these are degraded by IKK phosphorylation then NF-κB released and travels to nucleus as a hetero or homodimer.

Can get activating mutations of the NFKB gene and also deletions of IκB gene

256
Q

What are the hallmarks of cancer?

DEFINITELY IN CURRICULUM

A
  1. Sustained proliferative signalling: Acquired mutations short-circuit growth factor pathways leading to unregulated growth (oncogenes). EGFR inhibitors target this.
  2. Insensitivity to anti-growth signals: Acquired mutations or gene silencing interfere with growth inhibition pathways (tumour suppressor genes). CDK inhibitors.
  3. Evasion of programmed cell death. Cancer cells evade apoptotic signals. Pro-apoptotic BH3 mimetics.
  4. Replicative immortality: Replication of normal cells limited by telomere
    shortening. Altered regulation of telomere maintenance results in unlimited replicative potential. Telomerase inhibitors
  5. Induced angiogenesis: Cancer cells alter the balance between angiogenic
    inducers and inhibitors to activate the angiogenic
    switch. VEGF inhibitors.
  6. Activating invasion + metastasis: Genomic alteration affects enzymes involved in
    invasion or molecules involved in cell-cell/cellular-extracellular adhesion. Inhibitors of HGF/c-Met

+ emerging

  1. Avoid immune destruction. Immunotherapy agents
  2. Disregulating cellular energetics: Cancer cells carry out glycolysis even in the presence of oxygen – glycolysis intermediates are used in biosynthetic pathways = Warburg effect. Aerobic glycolysis inhibitors

+ enabling

  1. Genome instability and mutations: Faulty DNA repair leads to genomic instability. PARP inhibitors.
  2. Tumour promoting inflammation: Inflammation facilitates the ability to acquire core hallmarks of cancer and release of mutagenic oxygen
    species. Aspirin.
257
Q

What is a single nucleotide polymorphism?

DEFINITELY IN CURRICULUM

A

Change in a single base eg A-> G
Most either synonymous or benign
Specific SNPs in particular genes may be assoc with increased or decreased risk of cancer.

Many don’t cause alteration in protein, may not even be in a disease associated gene. THey are useful as if they are associated with a disease (GWAS study) that usually means they are near a gene.

Most SNPs are bi-allelic. Very useful as abundant and can by typed using microarray (hybridisation)

258
Q

How are mutations classified?

A

5 point classification - whether a change in amino acid changes function of protein
1. clearly not pathogenic
2. Likely non-pathogenic
3. Uncertain
4. Likely pathogenic- exchange hydrophobic for hydrophilic which changes structure of protein
5, clearly pathogenic

259
Q

What is hereditary non-polyposis colorectal cancer/HNPCC/Lynch syndrome and how is it inherited?

DEFINITELY IN CURRICULUM

A

Genetic Abnormality: Autosomal Dominant – defects in MSH2 and MLH 1 account for 90% of cases, also PMS1, PMS2, MSH6
Mechanism of Action: Loss of protein products coded by these genes&raquo_space; loss of MMR&raquo_space; reduced fidelity of DNA replication by orders of magnitude = microsatellite instability.
Associated cancers: Colorectal Cancer
Amsterdam II criteria: >3 family members (one a first degree relative of other two),
two successive generations, >1 diagnosed < 50 years, FAP excluded.
Also endometrial, small bowel, ovarian serous cystadenoma. Don’t tend to get polyps unlike FAP.

260
Q

Name some gain of function mutations of CPGs that cause cancer

A

11 CPGs activated by mutations - all autosomal dominant
ALK- generally only somatic
EGFR- generally on somatic
RET- common somatic, can be germline in MEN2

261
Q

Name some loss of funtion mutations in CPGs?

A

103 CPGs contain loss of function mutations
TP53
BRCA1
MLH1
Herogenous - retinoblastoma (single gene)

262
Q

What are cancer associated SNPs?

A

single nucleotide polymorphism
Contribute to increased risk but not high risk like CPGs
Some protective, some increase risk
Multiplicative effect as each one may only increase risk a small amount but together risk might be much higher

263
Q

Which patients would you consider testing for Li-fraumeni? If test was positive how would you follow them up?

DEFINITELY IN CURRICULUM

A

Breast cancer <30yrs esp if HER2 +ve
FH sarcoma
Choroid plexus tumours
Very high rates of early onset solid tumours over multiple generations

Annual MRI breast from age 20yrs
? annual whole body MRI

264
Q

What is MEN1?
Inheritance
Cancers

DEFINITELY IN CURRICULUM

A

Mutation of MEN1 gene (tumour suppressor menin)
AD

PPP
- Parathyroid -90% onset 20s
- Pancreatic - 70% - gastrinoma>non-functioning>insulinoma
- Pituitary - 40%, (non-functioning>prolactinoma)

265
Q

How do you monitor and treat people with MEN1?

DEFINITELY IN CURRICULUM

A

Parathyroid- annual PTH, calcium, prophylactic parathyroidectomy (removal of 3.5/4 parathyroid glands)
Pancreatic- annual gastrin, annual imaging e.g. CT/endoscopic USS
Gastrinomas- PPI/resection
Pituitary - annual prolactin and IGFR

266
Q

What is MEN2A?
Inheritance
Cancers

DEFINITELY IN CURRICULUM

A

RET gene - mutations in extracellular cysteines cause intermolecular disulfide bonds -> constitutive RET dimerisation and aberrant activation. ONCOGENE
Autosomal dominant

TAP (thyroid/adrenal/parathyroid)
Medullary thyroid cancer- 90%
Phaeochromocytoma 50%
Parathyroid adenoma 20-30%

Familial medullary thyroid cancer- 100% risk but don’t get other cancers, 10 people in family with medullary thyroid cancer, same mutation.

267
Q

How do you treat people with MEN2A?

DEFINITELY IN CURRICULUM

A

Annual calcitonin
Prophylactic Thyroidectomy (timing depends on severity of mutation)
From age of 8 or 20 - annual calcium PTH, urinary metanephrines and MRI abdo if urinary metanephrine abnormal

268
Q

What is MEN2B?
Inheritance
Cancers

DEFINITELY IN CURRICULUM

A

RET mutation - alters substrate binding pocket of tyrosine kinase domain via substitution -> increase kinase activity. Autosomal dominant. usually denovo RET mutation M918T variant in 95% cases.

More severe/earlier onset than MEN2a

TAM (thyroid, adrenal, all the Ms)
Medullary thyroid cancer >90%
Phaeochromocytoma 50%
Assoc abnormalities 50%
- marfanoid
- mucosal neuromas
- megacolon

269
Q

What is von hippel lindau syndrome and what are the features?

DEFINITELY IN CURRICULUM

A

VHL gene mutation (tumour suppressor gene): ubiquitin ligase that marks HIF for degradation in presence of oxygen so no angiogenesis
Autosomal dominant

Clear cell renal cancer (bilateral, young, familial history)
Cysts in kidneys, pancreas, genital tract
Retinal angiomas - Haemangioblastomas
Cerebellar or spinal haemangiomas
Retinal haemangioblastomas (blindness)
Phaeochromocytomas
Endocrine pancreatic tumours
Cystadenomas
Endolymphatic sac tumours of inner ear

Belzutifan for RCC in VHL patients – HIF2a inhibitor

270
Q

Would you test all patients with clear cell renal cancer?

A

Yes if any other features of VHL /other genetic syndrome e.g. cerebellar haemangioblastoma
Or if :
<40
Bilateral
Multifocal
First/second degree relative with renal cancer

271
Q

What causes familial melanoma and when do you develop melanoma?

(Not in curriculum)

A

CDKN2A mutations - gene contains instructions for making p16 and p14
CDK4 mutations

Multiple primaries at young age of onset

  • at least 3 melanomas in family
  • risk pancreatic cancer
272
Q

What are the major and micro criteria for Cowdens syndrome?

DEFINITELY IN CURRICULUM

A

Major criteria (3 or more, must be macrocephaly)

  • Macrocephaly (>97th centile) ALWAYs - do they have trouble buying hats
  • Breast cancer
  • Non-medullary thyroid cancer
  • Endometrial cancer

Minor criteria (2 major + 3 minor)

  • other thyroid lesions
  • IQ <75
  • Hamartomatous intestinal polyps
  • fibrocystic disease of breast
  • GU tumours or malformations
  • uterine fibroids
273
Q

What is the mutation responsible for cowdens syndrome?

DEFINITELY IN CURRICULUM

A

PTEN. Also called hamartoma syndrome. Autosomal dominant. Activation of PI3K pathway constitutively.

274
Q

What is peutz-jegher syndrome?
Inheritance
Features

DEFINITELY IN CURRICULUM

A

Mutations in STK11/LKB1 gene- tumour suppressor gene
AD (although usually spontaneous mutation)
Increase in hamartomatous polyps throughout GI tract
Dark brown macules around mouth and oral mucosa
Cutaneous and GI symptoms
50% have cancer by age 60yrs- breast, GI, pancreas, ovary, uterus, testicle, oesophagus, lung, 15 x risk of colorectal cancer

Surveillance

275
Q

What is the STK11 gene?

DEFINITELY IN CURRICULUM

A

Mutated in peutz-jegher syndrome. Serine/threonine kinase 11 - a tumour suppressor that helps to polarise cells and promotes apoptosis

276
Q

What is gardner syndrome?

A

Subtype of FAP characterised by osteomas, dental anomalies, epidermal cysts and soft tissue tumours

276
Q

Discuss neurofibromatosis type 1

DEFINITELY IN CURRICULUM

Features

A

Mutation of NF1 gene -> makes protein neurofibromin- acts as a tumour suppressor in oligodendrocytes, schwann cells. Neurofibromin is a negative regulator of the Ras pathway by stimulating the GTPase activity of Ras so turning it off. Non functioning neurofibromin cannot regulate cell growth and division -> tumours along nerves. Autosomal dominant.

  • neurofibromas
  • Lisch nodules (eyes)
  • cafe au lait and axillary/groin freckles
  • malignant peripheral nerve sheath tumours
  • optic glioma
  • learning difficulties
  • increased risk of brain tumours and leukaemias
  • pectus excavatum, scoliosis, short stature
  • sarcomas
277
Q

How is xeroderma pigmentosa inherited?

DEFINITELY IN CURRICULUM

A

AR

278
Q

What does xeroderma pigmentosa cause ?

DEFINITELY IN CURRICULUM

A

Phenotype severity varies according to genetic defect.

Extreme sensitivity to UV sunlight - x 1000risk skin cancer.
Burn at young age, by age of 2 freckling in sun exposed areas
1/2 develop skin cancer by age 10 without sun protection (SCC, BCC or melanoma)
Blindness/cataracts/corneal ulcerations/ketatitis and 30% get progressive neurological deficits involving eyes, ears, balance
20 x risk other cancers.
Mental retardation, premature dementia
Life expectancy 20-30s.
Note – these patients do not show more radiosensitivity – therefore NER is not important in DNA repair post radiotherapy.

279
Q

What is xeroderma pigmentosa caused by?

DEFINITELY IN CURRICULUM

A

7 types of mutation. Mainly in NER (nucleotide exicision repair) - XPC, ERCC2, POLH account for most. XPA-G

280
Q

What is Cockaynes syndrome?
Features
Inheritence

(Not in curriculum)

A

Rare form of dwarfism
+ deafness and retinal atrophy, microcephaly
UV sensitivity
Age quickly, 3 types starting at birth or childhood

AR mutation of ERCC6/8 gene -> involved in NER

281
Q

What syndromes cause radiosensitvity?

A

Li-fraumeni - sarcomas
Ataxia telangiectasia - most radiosensitive
Bloom syndrome
Nijemegen breakage syndrome
Gorlin syndrome
Fanconi syndrome

282
Q

How is ataxia telangiectasia inherited ?

DEFINITELY IN CURRICULUM

A

AR

283
Q

What causes ataxia telangiectasia? What are the features?

DEFINITELY IN CURRICULUM

A

ATM mutation (Ataxia Telangiectasia Mutated kinase (ATM) mutation). Chromosome 11q22

ATM is an inactive homodimer&raquo_space; activated by DSBs (ionising radiation)&raquo_space; phosphorylated to active monomers&raquo_space; phosphorylate P53 & Chk1 (G1/S checkpoint), Chk2 (G2/M checkpoint) causing either cell cycle arrest and DNA
repair or if damage too severe, apoptosis. ATM deficient cells are therefore very radiosensitive and unable to repair DSBs.

Severity of phenotypes varies depending on mutation

  • progressive cerebellar ataxia from childhood (mask-like face, slow eye movements, ataxia)
  • telangiectasia (skin, conjunctiva)
  • radiosensitive
  • cancer risk x 100 of lymphoma + leukaemia
  • immune defects
  • radiosensitivity
  • sterility
  • normal IQ
    1/100-200 people are carriers- higher in askenazi jews - carriers have increased risk of breast cancer – it is included in the breast cancer genetic panel - 1/40000 have disease
  • life expectancy 25 years – death due to cancer (usually lymphoma/leukaemia)p
284
Q

What is nijemegen breakage syndrome?

(Not in curriculum)

A
Mutation of NBN gene - part of MRN complex (MRE11, RAD50, NBS) used in HR and activated by ATM
AR
Short stature
Mental retardation
Microcephaly
Facial dysmorphism
Immunodeficiency
Increased risk of breast cancer

RADIOSENSITIVITY

285
Q

What is Bloom syndrome?

(Not in curriculum)

A

Mutation in BLM gene - codes for RecQ helicase - responsbile for maintaining DNA structure and unwinding it -> increased sister chromatid exchanges

Short stature
Sun sensitive rash
Bird like features
high pitched voice
COPD
DM
Immuno def
Increased cancer risk - H+N, SCC
Radiosensitivity
286
Q

Cancer cells need rapid ATP production but glycolysis only produces 2 ATP, how else can they make ATP and synthesis macromolecules?

A

Upregulate glutamine -> enter TCA cycle
Glutamine can do reverse TCA cycle and once converted to a-ketoglutarate this can undergo reductive carboxylation and form citrate -> major substrate for lipid synthesis.

Glutamine important in producing carbon intermediates and reduced nitrogen for nucleotide synthesis.

287
Q

How is the PI3K pathway influencing cancer metabolism?

A

PI3K/AKT pathway may regulate glucose metabolism by:

  1. Regulating glucose transporter expression through AKT -> stimulates transcription of GLUT1
  2. Enhancing glucose capture by HK2 and inducing aerobic glycolysis by promoting HK2 bidning to voltage dependent anion channels
  3. Stimulates PFK1 activity
288
Q

After irradiation which part of cell cycle is a cell most likely to arrest in?

A

Following ionizing radiation exposure, virtually all proliferating cells (independent of p53 (TP53) status) will show a radiation dose-dependent arrest in the G2 phase. Typically, cells with functional wild-type p53 will also arrest in the G1 phase of the cell cycle, and some cells may also arrest in S phase after radiation.

289
Q

What is the structure of a nucleotide?

DEFINITELY IN CURRICULUM

A

Nucleotides are composed of three subunit molecules: a nucleobase, a five-carbon sugar (ribose or deoxyribose), and a phosphate group consisting of one to three phosphates. DNA (deoxyribonucleic acid) is an acid (from the phosphate group) - the base forms bonds with the complementary base so doesn’t have basic activity.

290
Q

What the immunosuppressive cytokines?

May be in curriculum

A

IL10 (inhibits T cells/macrophages/dendritic cells) - via STAT3 pathways, TGF beta
(Tumour cells also produce IDO (indoleamine-2,3-dioxygenase) - role in maternal tolerance to foetal tissue - causes reduction in tryptophan for T cells to use - potential target to restore immunity)

291
Q

How to NK cells contribute to immune-drive tumour suppression?

May be in curriculum

A

Part of innate immune system. Perform cell mediated immunity by release cytotoxins e.g. granzymes and perforin.
1) Recognise stress ligands on tumour cells surface e.g. MIC through their stress ligand receptor (e.g. NKG2D)
2) Abundant cytosolic nucleic acids in tumour cells (due to genomic instability) activates a sensor called STING (stimulator of interferon gene). This induces interferon production which recruits NK cells.
3) Can perform ADCC (antibody dependent cellular toxicity)

292
Q

Discuss Wilm’s tumour

DEFINITELY IN CURRICULUM

A

Genetic Abnormality: Mutation of Wt1 or Wt2 tumour suppressor gene (transcriptional regulator) - seen in some genetic syndromes such as Beckwith-Wiedemann syndrome, Frasier syndrom, WAGR syndrome

Associated cancers: Nephroblastoma (kidney cancer in children aged 4-5 years - can metastasise to the lung - form from nephrogenic rests)

293
Q

Discuss familial breast cancer

DEFINITELY IN CURRICULUM

A

Genetic Abnormality: BRCA 1 (17q) and 2 (13q)
Mechanism of Action: autosomal dominant – involved In HR pathway of DNA repair. In patients with mutated BRCA, normal tissues are heterozygous – no increased sensitivity. A spontaneous somatic mutation in other BRCA allele in later life causes cells to be deficient in HR repair&raquo_space; genomic instability&raquo_space; sensitivity to DNA crosslinking agents (e.g. Cisplatin)
Synthetic lethality:
- PARP used in BER to repair SSBs. If left unrepaired (e.g. in context of PARPis) – replication converts these to DSBs. BRCA used in HR to repair DSBs
- Normal cells treated with PARPis – functional HR – viable
- BRCA mutant tumour cells treated with PARPis - loss of BER and HR – synthetic lethality - death
Associated cancers: breast, ovarian cancer, prostate, fallopian tube,
leukaemia/lymphoma. BRCA2 also in Fanconi anaemia (recessive)

In the NHS currently the genetic breast cancer screening panel includes - BRCA1 (HR), BRCA2 (HR), PALB2 (HR – aids BRCA2), ATM (DSB) and CHEK2 (DSB)
PALB2: Tumour suppressor gene that is a partner of both BRCA1 and BRCA2 so part of HR pathway: Breast cancer (lesser extent ovarian and pancreatic cancer
Chk2: familial breast cancer due to mutation 1100delC in Chk2, 37% risk of breast cancer at age 70 in women

The prevalence of BRCA1 mutation is slightly lower than that of BRCA2 mutations in the general US population (1:500 for BRCA1 vs 1:222 for BRCA2). The breast cancer risks for carriers of BRCA1 and BRCA2
mutations are similar at about 70% by the age of 80, but with earlier age of disease onset for the BRCA1 mutation (peaks in 4th decade) compared to BRCA2 carriers (peaks in 5th decade).

294
Q

What are external triggers for inflammation-induced cancer?

DEFINITELY IN CURRICULUM

A

Cancer is also known as the “wound that never heals” - chronic inflammation. Inflammation and infection cause 20% cancers. Potential treatment/prevention e.g. aspirin, celecoxib (used in FAP) - inhibits prostaglandin production from arachidonic acid by COX2.
1) Infectious agents e.g. hepatitis B
2) Tobacco smoke (also has 82 carcinogens)
3) Asbestos
4) Obesity - enlarged adipocytes (hypertrophy) are source of pro-inflammatory molecules
5) Inflammatory bowel disease = increased risk of colorectal cancer
6) Cirrhosis and HCC e.g. obesity/alcohol/infectious driven
7) Dysbiosis (imbalance in gut microbiota) may contribute to cancer in obesity
8) E coli, Strep bovis, H pulori = colorectal cancer

295
Q

Name some parasitic causes of cancer?

DEFINITELY IN CURRICULUM

A

Schistosoma haematobium = bladder (liver fluke)
Opisthorchi viverrini = liver cancer (liver fluke)
Clonorchi sinensis = liver cancer (liver fluke)

296
Q

What cancers is HIV associated with?

DEFINITELY IN CURRICULUM

A

HIV – Kaposi’s, non-hodgkins lymphoma, hodgkins

Retrovirus.

297
Q

Discuss smoking and cancer

DEFINITELY IN CURRICULUM

A

Causes lung, oropharynx, larynx, oesophagus, pancreas, bladder cancer.
81 carcinogens: benzo [a] pyrene (polycyclic aromatic hydrocarbon) which causes G to T transversions and activated by CYP1A1; nitrosamines cause O6 guanine alkyation.
Polymorphisms in CYP1A1 can vary how much carcinogen is activated so affects cancer-rates in smokers.
Inflammation

298
Q

Name some environmental causes of cancer

DEFINITELY IN CURRICULUM

A
  • Occupational e.g scrotal and nasal cancer in chimney sweeps from soot, bladder TCC from aromatic amines and azo-dyes (textile industry), mesothelioma from asbestos
  • Toxic agents in urban air, organic compounds from agriculture, chlorination/contaminants of drinking water, UV light
  • Diet – stomach cancer from high-nitrate foods; liver cancer from mouldy grain (aflatoxins)
299
Q

Name some lifestyle factors that contribute to cancer

DEFINITELY IN CURRICULUM

A

1) Smoking (note there is synergy between smoking and alcohol in carcinogenesis)
2) Alcohol
3) Obesity
4) Physical activity
5) Reproductive choices: having children, late menarche, early menopause, breastfeeding, not using oestrogen-based contraceptives = less breast cancer. (Oestrogen is a mitogen for breast cancer and also thought the oestrogen metabolites are genotoxic)
6) Sexual activity e.g. HPV
7) IV drug use e.g. Hep B/C = HCC, HIV = kaposis/lymphoma.
8) Age is the most important risk factor

300
Q

Discuss radiation-induced damage

DEFINITELY IN CURRICULUM

A

Ionising radiation e.g. alpha/beta particles and gamma rays. Ionises by mobilising an electron. Can directly ionise DNA or with water (radiolysis) to form hydroxyl radical (OH) then hydrogen peroxide (H2O2) then superoxide radical (O2-). Causes dsDNA breaks = mitotic catastrophe. Leukaemia is most frequent ionising-radiation induced cancer. Solid tumours increase with dose in linear fasion. Thymol glycol [5,6-dihydroxy-5,6-dihydrothymine] and oxidized guanine [8-oxo-7,8-dihydroguanine (8-oxoG)] are DNA base lesions present in clustered DNA damage induced in cells by ionizing radiation. These other forms of DNA damage, however, are more readily repaired and are less likely to result in cell death.

Ultraviolet radiation - UVB - cyclobutane pyrimidine dimers and 6,4 photoproducts. BCC/SCC/MM. Also DNA/protein crosslinks - mostly occur in euchromatin

Tumour cells die by mitotic catastrophe in radiation. Lymphoid and some epithelial normal tissues die by apoptosis in ionising radiation - this occurs in interphase.

301
Q

What is paracrine/autocrine/endocrine signalling?

DEFINITELY IN CURRICULUM

A

Paracrine signaling acts on nearby cells, endocrine signaling uses the circulatory system to transport ligands, and autocrine signaling acts on the signaling cell.

302
Q

What is kataegis?

DEFINITELY IN CURRICULUM

A

Small areas of hypermutation in cancer genomes. “Thunderstorm”

303
Q

What is the significance of extrachromosomal DNA?

DEFINITELY IN CURRICULUM

A

Tumour cells contain large circular extrachromosomal DNAs - rarely found in normal cells. They can replicate but do not have centromeres so are not equally distributed into daughter cells. Causes tumour cell heterogeneity. Is a source of amplified genes, chimeric rearrangements and reintegration into the linear genome of chromosomes. They can also be excluded from cells temporarily e.g. to cause drug resistance in presence of chemotherapy then reabsorbed once chemotherapy gone - allows flexibility to tumour. Non-mendelian inheritance. Chromatin in ecDNA is more open than conventional linear DNA

304
Q

What is an enhancer?

A

Regulatory DNA sequences that are position and orientation independent relative to the promoter. Important for tissue and stage specific expression.
Clusters of enhancers = super-enhancers.

305
Q

What are driver and passenger mutations? What is a mutator phenotype?

DEFINITELY IN CURRICULUM

A

Driver mutations = confer a growth advantage.

Passenger mutations = do not confer a growth advantage.

Mutations in DNA repair = mutator phenotype.

306
Q

What are hetero and euchromatin

DEFINITELY IN CURRICULUM

A

Heterochromatin - highly compact chromatin, no transcription - “silenced”.

Euchromatin - open accessible chromatin, transcription occurs.

307
Q

Describe DNA replication

DEFINITELY IN CURRICULUM

A
  • DNA Helicase unwinds the helix, DNA Polymerases synthesise DNA, always in the 5’&raquo_space; 3’ direction
  • Copied in a semi-conservative manner, each strand encodes its own complimentary strand, therefore each
    DNA molecule contains one parental strand and one de novo synthesised strand
308
Q

What is a gene?

DEFINITELY IN CURRICULUM

A

The molecular gene is a sequence of nucleotides in DNA that is transcribed to produce a functional RNA. There are two types of molecular genes: protein-coding genes and non-coding genes.

309
Q

What are mini- and microsatellites?

DEFINITELY IN CURRICULUM

A
  • Tandemly repeated DNA (also found as centromeric DNA)
  • Mini – up to 20kb length and repeat units up to 25bp, telomeric DNA (TTAGGG repeat) is an example
  • Micro – up to 150bp and repeat units of 1-4bp (function unknown)
310
Q

What is a polymorphism?

DEFINITELY IN CURRICULUM

A

Polymorphism, as related to genomics, refers to the presence of two or more variant forms of a specific DNA sequence that can occur among different individuals or populations. Used as DNA markers

SNPs is common type.

RFLP – restriction fragment length polymorphisms: Allelic variation in site for restriction enzyme cutting, Bi-allelic, so not that useful

SSLP – simple sequence length polymorphisms: Array of repeat sequences that have variable lengths,
Multi-allelic, Minisatellites – variable number of tandem repeats (VNTRs), less useful as tend to be found in telomeric regions and less good for PCR due to length, Microsatellites – simple tandem repeats (STRs), preferred

311
Q

What is senescence?

DEFINITELY IN CURRICULUM

A
  • Halting of DNA replication and cell cycle proliferation, via activation of pRB and p53
  • Cells can only replicate a limited no. of times (Hayflick Limit) as they’re unable to copy chromosome caps
    (Telomeres)
  • loss of senescence (limitless replicative potential) is a cancer hallmark
  • p21 is temporary senescence, p16 is permanent
312
Q

What is MYC?

DEFINITELY IN CURRICULUM

A

Family of transcription factors: MYC, MAX, MAD, MXI. Dimerise in different ways and lead to distinct biological effects of growth, differentiation and death.

MYC promotes proliferation including E2F, hTERT, p21 and CDK4. Requires heterodimerisation with MAX to function via basic helix-loop-helix leucin zipper domains - bind to E-box in target genes. MAX and MAD/MXI repress transcription at E-box target genes so inhibit proliferation.

  • t(8:14) translocation places c-MYC under control of immunoglobulin heavy or light chain gene transcriptional promoter&raquo_space; increased cell proliferation
313
Q

Define neoplasia

DEFINITELY IN CURRICULUM

A

Neoplasia: abnormal proliferation of cells.
* Benign neoplasm: -oma.
* Malignant neoplasm: cancer, -carcinoma, -sarcoma etc.

–carcinoma = epithelial cell cancer – accounts for 85% of all cancers
–sarcoma = mesodermal (muscle/bone) cancer
–adenocarcinoma = secretary/glandular tissue e.g. breast

314
Q

What are the stages of metastasis?

DEFINITELY IN CURRICULUM

A

Stages of metastasis:
1. Primary tumour formation
2. Local tissue invasion
3. Intravasation
4. Transport in circulation
5. Lodgement in distant tissues
6. Extravasation
7. Micrometastasis
8. Metastasis – dormancy or outgrowth

315
Q

Describe the local effects of tumours?

DEFINITELY IN CURRICULUM

A
  • Pain (somatic, neuropathic)
  • Pressure and Obstruction
  • Ulceration and bleeding
  • Invasion – organ dysfunction e.g. seizures
316
Q

What are the distant effects of tumours?

DEFINITELY IN CURRICULUM

A

Cancer cachexia (due to cytokines - NOT nutritional demands of cancer)

Paraneoplastic syndromes:
o Cushing’s syndrome (ACTH), SIADH (hyponatraemia) – SCLC
o Hypercalcaemia – NSCLC, Breast, Prostate – PTHrp
o Hypoglycaemia – HCC, sarcomas – Insulin/Insulin like substances
o Myaesthenia, peripheral neuropathy – bronchogenic carcinoma
o Acanthosis nigricans (gastric), Dermatomyositis photosensitive rash (bronchogenic)
o Hypertrophic osteoarthropathy and clubbing
o Venous thrombosis
o Nephrotic syndrome
o polycythaemia = EPO from RCC

317
Q

What is the angiogenic switch?

DEFINITELY IN CURRICULUM

A

Angiogenic Switch – tumour creates conditions of hypoxia leading to:
- over-expression of angiogenic-inducers (e.g. VEGF, PDGF, Hepatocyte-derived Growth Factor, Fibroblast growth factor, Epidermal Growth Factor)
- down-regulation of angiogenic-inhibitors (e.g. Angiostatin, Endostatin, p53, Thrombospondin 1/2)

318
Q

What are features of the tumour vasculature?

DEFINITELY IN CURRICULUM

A
  • chaotic and inefficient resulting in poor-perfusion, limited access by cytotoxic agents, hypoxia and subsequent resistance to radiotherapy and promotion of further angiogenesis
  • immature vessels lack pericytes, therefore haemorrhage, tumour cell escape and ascites
319
Q

Name the mechanisms of tumour vascularisation

DEFINITELY IN CURRICULUM

A

In the absence of angiogenesis, tumors would only be expected to reach a diameter of about 2 mm
1. Activation of nearby vessels and induction of sprouting (angiogenic switch)
2. Co-option of existing vessels in the peri-tumoral area – tumour grows along vessel walls
3. vasculogenesis is the formation of blood vessels from circulating bone-marrow endothelial progenitor cells.
4. Vascular mimicry: channels lined by tumour cells – common in melanoma

320
Q

What are the sources of mutation?

DEFINITELY IN CURRICULUM

A

Internal: - error-prone synthesis or repair, cellular metabolism, reactive oxygen species
External: - Ionising radiation, UV light, chemicals, toxins, virus, chemotherapy

321
Q

What is autophagy? What is the difference to apoptosis?

A
  • Mechanism of survival under stress
  • Cell breaks down own organelles using lysosomes to provide for more essential processes - can be selective to the organelle (mitophagy, ribophagy etc) or non-selective to the cytoplasma (macroautophagy)
  • Lack of chromatin condensation
  • Accumulation of (double-membraned) autophagic vacuoles (autophagosomes) which fuse with lysosomes (autolysosomes)
  • Little or no uptake by phagocytic cells
  • whole process is tightly regulated through at least 30 Atg-autophagy genes

Not taken up by phagocytic cells, no chromatin condensation. Beclin is a protein important in the development of the autophagosome for autophagy.

Autophagy is a process in which cells generate energy and metabolites by digesting their own organelles and macromolecules and as such it is a survival mechanism. Autophagy allows a starving cell, or a cell that is deprived of growth factors to survive up to a point. Cells that do not receive nutrients for extended periods ultimately digest all available substrates and die through autophagic death. Supplying nutrients before this critical point would restore the cell’s health

322
Q

What are experimental ways to detect apoptosis?

A
  1. Annexin V – most commonly used method via flow cytometry
  2. Tunel assay – detects DNA fragments
  3. DNA ladder – via gel electrophoresis of pooled DNA

During the execution phase of apoptosis, nucleases are activated which cleave DNA into 180-200 base pair increments. Several assays are available to measure this phenotype. The TUNEL method identifies apoptotic cells by using terminal deoxynucleotidyl transferase (TdT) to transfer biotin-dUTP to strand breaks of cleaved DNA. The Annexin V Assay, a classical technique for detecting apoptosis, is the most commonly used method for detecting apoptosis by flow cytometry. Annexin V is a calcium-dependent phospholipid binding protein that has a high affinity for the phophatidylserine (PS), a plasma membrane phospholipid. One of the earliest features of apoptosis is the translocation of PS from the inner to the outer leaflet of the plasma membrane, thereby exposing PS to the external environment. Annexin V binds to PS exposed on the cell surface and identifies cells at an earlier stage of apoptosis than assays based on DNA fragmentation. DNA ladder formation is detected by gel electrophoresis of pooled DNA. Diamidino-2-phenylindole (DAPI) is DNA-specific dye that displays a blue fluorescence. This dye could be used to assess the nuclear morphology of normal versus apoptotic cells by fluorescence microscopy.

323
Q

What does ATM do?

A

Pauses cell cycle via p53 (and less extent CDC25) - p53 triggers apoptosis if needed.
Activates DNA repair via H2AX, Nbs1, DNAPKc, BRCA2, BLM1 = HR

ATM is activated following irradiation by auto-phosphorylation and conversion from an
inactive homodimer to an active monomer

324
Q

Which point of cell cycle is most radiosensitivity?

A

G2/M>G1>early S>late S

HR needs second chromatid so only active in late S - fixes radiation induced dsDNA breaks.
G2/M is beyond the DNA repair checkpoint so too late to fix radiation induced dsDNA breaks so mitotic catastrophe occurs

325
Q

What effect does hyperfractionation have on tumour control, late toxicities and early toxicities?

A

<1.8Gy/fraction
Tumour control (Assuming high alpha/beta ratio) = no effect
Early toxicity (high alpha/beta ratio) = no effect
Late toxicity (low alpha/beta ratio) = reduced toxicity
As alpha/beta ratio is inversely proportional to sensitivity to fractionation

326
Q

What effect does hypofractionation have on tumour control, late toxicities and early toxicities?

A

> 2Gy/fraction
Tumour control = no effect
Early toxicity = no effect
Late toxicity = increased toxicity

327
Q

What effect does acceleration have on tumour control, late toxicities and early toxicities?

A

Acceleration = >10Gy/week
Tumour control = increased control (Assuming you went beyond T delay initially) as less repopulation
Early toxicity = increased as less repopulation
Late toxicity = no effect as no repopulation

328
Q

What is the effect of having two fractions/day on tumour control/early toxicities/late toxicities? (Assuming total treatment time and dose is same)

A

Tumour control: no effect due to rapid DNA repair
Early toxicities: no effect due to rapid DNA repair
Late toxicities: increased as slow DNA repair

329
Q

What is the effect of increasing total radiation dose on tumour control/early toxicities/late toxicities?

A

Tumour control = increased
Early toxicity = increased
Late toxicity = increased

330
Q

Which DNA repair pathways are important in radiation?

A

NHEJ > HR for causing radiosensitivity if mutated. BER does not usually cause radiosensitivity if mutated (Apart from XRCC1 gene)

ssDNA breaks (BER/PARP) - most common as ssDNA breaks more common than dsDNA breaks
dsDNA break - NHEJ (throughout cell cycle, fast, inaccurate/mutagenic, most used by cells. most important in radiation repair) and HR (S/G2 phase, slow, accurate)

Loss of NHEJ = increase in radiosensitivity and lose the alpha part of the curve.
Loss of HR = sensitive to PARP inhibitors = synthetic lethality

331
Q

What does alpha and beta represent in the alpha and beta ratio?

A

Alpha = sensitivity to low doses of radiation, linear bit of the curve. Represents dsDNA breaks and/or lethal damage and/or DNA repair pathways not saturated and/or direct damage. Gray-1

Beta = sensitivity to high doses of radiation, exponential bit of curve. Represents multiple ssDNA breaks in one cells and/or multiple sublethal damage hits and/or DNA repair pathways saturated and/or indirect damage. Gray-2

Alpha/beta ratio = inversely proportional to sensitivity to changes in fractionation. High = straight, low = curved. Gray. Is where AD=BD2. Linear component of cell kill = quadratic component. Determines the bendiness of the survival curve. The A/B ratio is the dose at which the linear and quadratic contributions to cell killing are equal.
High alpha/beta ratio in tumour/acute toxicities: usually highly proliferating tissues, tissues with more lethal damage and less sublethal damage
Low alpha/beta ratio: late toxicities, lower proliferating tissues, tissues with more sublethal damage repair

332
Q

What are the alpha/beta ratio for late toxicity, early toxicity, squamous cell e.g. H+N, breast adenocarcinoma and prostate adenocarcinoma?

A

Late toxicity: 3
(Spinal cord: 2)
Prostate cancer: 1.5 (hypofractionate)
Breast: 4-4.5 (hypofractionate)
Early toxicity: 10
H+N: 10 (hyperfractionate)
Skin: fibrosis = 3, erythema = 10

333
Q

What is SF2?

A

Surviving fraction at 2Gy
A measure of radiosensitivity

334
Q

How are survival curves plotted for radiation? What are dose response curves?

A

Semi-logarithmic (dose on x axis, logarithm of survival fraction on y axis) = allows visualisation of low doses as you want to get to 0 or as low as possible

Dose response curves = total dose on x axis, response on y axis = sigmoid shape (low dose doesn’t cure much, then get a lot of cure, then not much cure). Radiosensitiser shifts the tumour control curve to the left. Hypoxia shifts to the right. Complication rate must be to the right otherwise you cannot give treatment. Is parallel to the tumour control. Radioprotection moves complication rate to the right (but also protects the tumour hence none are used in practice)

335
Q

What are the 5 Rs of radiobiology?

A
  • Repair: DNA repair of sublethal damage - SLD mostly in low alpha beta tissues I e. late toxicities. Fractionation = time for late toxicity repair = better tolerability, no effect on tumour control.
  • Radiosensitivity: depends on tumour. Can increase by radiosensitisers e.g. chemotherapy, PARP inhibitors, treating anaemia. The law of Bergonie and Tribondeau states: The radiosensitivity of a cell is directly proportional to reproductive rate and is inversely proportional to its degree of differentiation.
  • Repopulation: early toxicities and tumour repopulate. Fractionation = more repopulation of tumour = worse control.
  • Redistribution: G2/M>G1>early S>late S. Catch cells at different phases by fractionation = better tumour control.
  • Reoxygenation: oxygen = ROS = more radiosensitive. normal tissues are not hypoxic so only applies to tumour Fractionation = opens temporarily closed vessels + changes architecture so cells get oxygenated + oxygen can get to inside cells = better tumour control.
336
Q

What is the equation for BED?

A

Biologically effective dose on a specific effect e.g. tumour control or late toxicity
BED = nd (1+d/alphabeta ratio)-K x (T-T delay)

n = number of fractions
d = dose/fraction
(nd = total dose)
K = constant for each effect = Gy/day - dose lost per day of repopulation
Tdelay = days until repopulation occurs

(Tdelay usually 28 days, K = 0.9 for H+N)

337
Q

What is the total dose required to get a radiotherapy regimen for prostate cancer equivalent to 74Gy in 2Gy/fraction (new regimen in 3Gy/fraction)

A

EQD2 = nd (d1+alpha/beta) / (d2+alpha/beta)
74 = nd (3Gy + 1.5)/(2Gy + 1.5)
74 (2+1.5)/(3+1.5) = nd

Used to compare different regimens with different dose per fraction.

EQD2 gives you a practical dose in 2Gy factions whereas BED gives you a theoretical biological effect dose.

338
Q

Describe radiation damage to the eyes

A

o Early:keratoconjunctivitis
o Late: cataracts – occurs at low doses but is easily treatable with surgery. Occur at 2Gy. TD5/5 10Gy.
o Late: “dry eye” due to damage to lacrimal glands – this is dose limiting effect. Causes chronic corneal ulceration and loss of eye. TD5/5 is 30Gy of dry eye.
A radiation-induced cataract is one of the few examples of a radiation injury which does have distinct pathognomonic characteristics that identify it as having been induced by ionizing radiation; radiation-induced cataracts typically begin in the posterior portion of the lens, unlike age-related cataracts.

339
Q

Describe radiation damage to the ears

A

o Early: otitis media
o Late: permanent hearing loss if >30 Gy

340
Q

Describe radiation damage to taste

A

o Get loss of taste due to direct damage and due to xerostomia (dry mouth) from salivary glands after 30Gy – usually resolves but may not.

341
Q

Describe radiation damage to cartilage and bone

A

o Epiphysial plate: very radiosensitive (4Gy) get loss of growth.
o Adult cartilage: radioresistant but can get osteoradionecrosis as a late effect.
(Femoral head TD5/5 is 52Gy)
Bone marrow: aplasia, pancytopenia.
TD5/5 2.5Gy

342
Q

Describe radiation damage to the heart

A

o Late: reversible changes in ECG that are not predictive; reversible pericarditis with pericardial effusion – usually asymptomatic and occurs within 2 years; radiation-induced cardiomyopathy with reduced EF or conduction blocks – takes 10-20 years to develop; ischaemic heart disease. Occurs at 50Gy in 2Gy fractions.
o Exact damage thought to be due to volume effects and also sensitive substructures – in future may volume the heart in sections e.g. coronary arteries, valves.
A number of large clinical trials, particularly those performed in Hodgkin’s disease patients, have indicated that the populations most at risk for RIHD are young females and the elderly, and that the important factors governing tissue tolerance are total dose, fraction size, and volume irradiated.
(V40 = 50%) TD5/5 40Gy.

343
Q

Describe radiation damage to peripheral nerves e.g. brachial plexus

A

o Late effects: sensory and motor defects

344
Q

Describe radiation damage to spinal cord

A

o Earliest late effect: Lhermitte sign (reversible demyelinating reaction) – not predictive for other effects.
o Late effect: permanent myelopathy (paralysis) – can be due to demyelination and necrosis of white matter or due to vasculopathy.
o Tolerance of spinal cord dependent on size of dose per fraction so giving repair time doesn’t have much effect.
o Generally radioresistant compared to other organs but very important so use a TD5 (5% complication level)
TD5/5 = 45Gy

345
Q

Describe radiation damage to the brain

A

o Transient demyelination = somnolence syndrome.
o Leukoencephalopathy.
o Radionecrosis - Initially can white matter necrosis and demyelination then get vascular changes with telangiectasis and focal haemorrhages.
o Children’s brains more sensitive than adults.
o Generally radioresistant compared to other organs but very important so use a TD5 (5% complication level)
The effects of radiation on the nervous system arise primarily as a
consequence of damage to oligodendrocytes and glial cells
TD5/5 = 45Gy

346
Q

Describe radiation damage to the urinary bladder

A

o Get reduced storage so increased urinary frequency both early and late but due to different pathologies
o Early: hyperaemia and mucosal oedema. Worsened by UTI which can cause desquamation and ulceration. These early changes contribute to the severity of the late changes.
o Late: progressive mucosal breakdown resulting in ulceration and fistulae. Can get fibrosis of bladder wall. Can get bleeding episodes due to telangiectasia. Can take up to 10 years – the higher the radiation dose, the quicker the formation of complications.
Urethra TD5/5: 70Gy
Bladder TD5/5: 75Gy

347
Q

Describe radiation damage to the kidney

A

second most sensitive volume effect
o Late: radiation nephropathy – proteinuria, hypertension, anaemia and impairment in urine concentration. Glomerular injury then glomerular sclerosis then tubule-interstitial fibrosis. RAAS system plays a role - may be mitigated/prophylaxis with ACEi.
o Low alpha/beta ratio.
o Dose tolerated by kidney reduces with time as you develop the radiation nephropathy – therefore retreatment is best avoided.
o Dependent on volume effect – if only partially irradiated, the remaining kidney can hypertrophy to compensate for the damaged area.
The kidney has a relatively low tolerance dose because of the limited number of clonogens within each nephron, although the cells comprising the functional subunits of the kidney are not particularly radiosensitive. The kidney exhibits substantial sparing with fractionation and displays little or no tolerance to re-irradiation. (V20 = 20%) TD5/5 23Gy.

348
Q

Describe radiation damage to the lung

A

most sensitive volume effect
o Early: pneumonitis. SOB, dry cough, reduced gas exchange, reduced pulmonary compliance.
o Late: lung fibrosis (chronic radiation pneumopathy). Very radiosensitive, significant volume effect (lung is the dose-limiting organ in total body irradiation). Increasde age and tamoxifen treatment increase risk of fibrosis.
The best predictor for late effects has been found to be the V20/V30, that is, the percentage of normal lung volume that receives should not receive more than 20 Gy or 30 Gy, respectively. (V20 =
30%). T5/5 17.5Gy.

349
Q

Describe radiation damage to the liver

A

third most sensitive volume effect
o Radiosensitive, with tolerance dose of 30Gy in 2Gy fractions, volume dependent e.g. in total body irradiation for bone marrow transplantation (liver acini are the FSUs).
o Early: acute radiation hepatitis with venoocclusive disease (central vein thrombosis) causing liver enlargement and ascites
o Late: chronic hepatopathy – fibrosis of liver.
(V30 = 40%)* TD5/5 30Gy.

350
Q

Describe radiation damage to the intestine

A

o Early: get initial increase in motility then atonic phase followed by loss of proliferating epithelium (atrophic villi) which causes diarrhoea (Water and electrolyte loss) and resulted absorption/resorption (affects oral drug absorption). This can cause sepsis. “Fixed” intestinal loops e.g. due to adhesions and the rectum are more at risk as they will be exposed to radiation on each dose as they cannot move.
o Late: chronic ulcers; stenosis; ileus; telangiectasis causing bleeding. These changes are induced by the early damage.

in the rectum, it is the percentage of the wall that has received 40-50 Gy that determines the likelihood of rectal bleeding, although the extent of reserve, unirradiated tissue is also a factor.

The small intestine is the most sensitive component of the GI tract. TD5/5S:
Small intestine: 40Gy
Colon: 45Gy
Rectum: 60Gy

351
Q

Describe radiation damage to the stomach

A

o Early: gastritis
o Late: ulceration due to vascular damage
TD5/5 50 Gy

352
Q

Describe radiation damage to the salivary gland

A

o Early: transient hypersalivation then stop salivation after 40Gy. Volume effect very pronounced so need to irradiate entire salivary gland to get these complications.
o Late: chronic xerostomia if doses >60Gy.
Parallel organ
The best way to spare the parotid gland is to decrease the volume of the gland irradiated.
The parotid exhibits relatively little sparing with fractionation so use of either a hyperfractionated or hypofractionated protocol would have only a modest impact. Prolongation or acceleration of treatment would have little effect on the parotid. The serous acinar cells of the parotid and submaxillary glands are Iconsidered to be the targets for radiation-induced salivary gland damage.
Quantec 1 gland dose <20Gy

353
Q

Describe radiation damage to the teeth

A

o Early: radiation caries due to direct damage at dentine-enamel border and due to xerostomia and changes in the microbiome of the mouth.
o Patients should have dental treatment prior to radiation as if they have it after they can get osteoradionecrosis.
Radiation tolerance of the mandible is also affected by pre-irradiation dental disease, fraction size and gender (males more susceptible).

354
Q

Describe radiation damage to the oral mucosa and oesophagus

A

o Early: oral mucositis and ulceration. Dose limiting in radical head and neck radiation. Oesophagitis causing dysphagia.
o Late: mucosal atrophy and ulceration; telangiectasia (bleeding); strictures in the oesophagus causing chronic dysphagia. TD5/5 is 55Gy.

355
Q

Describe radiation damage to the skin

A

o Early: temporary erythema (1 day) dry desquamation (Radiodermatitis sicca) then moist desquamation (4 weeks)
o Late: subcutaneous fibrosis (induration); telangiectasia. Can occur without any early changes as it occurs in different skin layer.
o Hair loss: transient or permanent depending on dose – can grow back with discolouration.
o Loss of sebaceous and perspiratory glands resulting in dry skin.
Following irradiation of the skin, the dose and time course for epilation and loss of sebaceous gland secretion are similar. Following skin irradiation, the first visible evidence of damage is a transient erythema that is observed within 24 hours following irradiation, whereas moist desquamation would only be observed after a few weeks. Epilation is observed at doses similar to those that cause the main wave of erythema that is typically manifested about one week following irradiation. Pigment changes typically appear long after irradiation due to the low proliferation rate of melanoblasts. It is usually not possible to predict the extent of late reactions based upon the severity of early reactions because early reactions result from killing of epidermal stem cells, whereas late reactions likely occur due to vascular damage in the dermis.

356
Q

How should missed radiotherapy doses be addressed?

A

The ideal procedure is to transfer all patients to a matched linear accelerator on the day of interruption. Where this is not possible the following approaches are recommended.
– Where possible, there should be the facility that allows patients who have missed scheduled weekday treatments to be treated at the weekend. Departmental protocols must ensure that complex treatments can be safely delivered out of normal hours.
– Patients can be treated twice daily, with a minimum of six hours between therapies.
– Use of biologically equivalent dose (BED) calculations to derive an alternative schedule involving a modified number of treatment fractions with which to complete the radiotherapy course in the planned overall time, but perhaps accepting a higher BED in normal tissues.
– The addition of extra treatment fractions where compensation cannot be achieved within the original overall planned time

357
Q

What is the acceptable radiotherapy treatment delay for category 1 patients? Which cancers are included in this category?

A

Treatment duration MUST not be prolonged by more than two days over the original prescription.

Radical therapies for:
§ Squamous cell carcinoma of the head and neck region
§ Non-small cell and small cell lung carcinoma
§ Squamous cell carcinoma of the cervix
§ Squamous cell and adenocarcinoma of oesophagus
§ Squamous cell carcinoma of skin, vagina or vulva
§ Squamous cell carcinoma of the anus
§ Medulloblastoma and primitive neuroectodermal tumours (PNET)
§ Patients with tumours with a short mass-doubling time

358
Q

What is the acceptable radiotherapy treatment delay for category 2 patients? Which cancers are included in this category?

A

It is ADVISED that their treatment should not be prolonged by more than two days over the original prescription. Must not go above 5 days.

Generally adenocarcinoma
§ adenocarcinoma of the breast
§ transitional cell carcinoma of the bladder
§ carcinoma of the prostate

359
Q

What is the acceptable radiotherapy treatment delay for category 3 patients? Which cancers are included in this category?

A

These are patients being treated with palliative intent. Overall time is less critical in achieving the desired palliative outcomes. Prolonged interruptions, which may occur because of intercurrent illness, may require compensation, particularly if longer than seven days.

360
Q

What is the definition of cell kill in radiobiology?

A

Radiobiologically speaking, a cell is ‘killed’ if it is rendered unable to divide and cause further growth.

361
Q

What is the bystander effect?

A

Non-targeted, radiation-induced bystander effects are effects that appear in unirradiated cells in the
presence of irradiated cells.

Cells growing in the growth media of cells that have been irradiated demonstrate effects of radiation
Ability of transfected cells to transfer death signals to neighboring tumor cells through GAP junctions e.g. reactive oxygen/nitrogen species or extracellular signalling via TGFbeta or cytokines.

362
Q

How much damage does 1Gy radiation cause?

A

1000ssDNA breaks and 40 dsDNA breaks

363
Q

Name some methods to detect DNA breaks?

A

Neutral comic assay can only detect Double strand DNA, thus reflect the only double strand DNA breaks. Alkaline comic assay can detect both double and single stranded DNA, thus theoretically can be
used to measure single strand breaks as well as double strand break.
Pulse Filed Gel Electrophoresis (PFGE) can detect fragmented DNA to assess level of DNA double strand breaks.
Neutral comet assay can measure DNA double strand breaks.
Specific antibodies can be used to detect oxidated bases, such as 8-Oxoguanine (8-OxoG).
Phosphorylated H2AX (gH2AX) can be used as a surrogate marker for DNA double strand breaks -, most accurate marker.

364
Q

What is the abscopal effect?

A

The abscopal effect is a phenomenon in the treatment of metastatic cancer where localized irradiation of a tumor causes, not only a shrinking of the irradiated tumor, but also a shrinking of tumors far from the irradiated area. Thought to be due to immune activation.

365
Q

What is the dose rate effect?

A

The lower the dose rate, the lower the cell kill even for the same total dose. This is due to increased DNA repair of sublethal DNA damage. Therefore if you inhibit DNA repair = no dose rate effect. The traditional dose-rate effect is most pronounced between dose rates of 1Gy/min and 0.3Gy/hour. Within this range, as dose rate decreases, cell killing decreases and the slope of the survival cure becomes more shallow, not steeper. The magnitude of dose-rate effect varies significantly among different cell types due to inherent differences in capacity for sublethal damage repair.

High dose rate = less sublethal damage repair = the beta component of cell killing will increase.

High alpha/beta ratio = beta component not very important = not much sublethal damage = no dose rate effect

High LET = less sublethal DNA damage = less dose rate effect. If you go below 1cGy/min then death increases due to asynchronous cell population becomes synchronized as sensitive G2 cells: inverse dose rate effect.

366
Q

What are the different types of cellular damage (lethal/sublethal/potentially lethal)?

A

PLDR is best demonstrated with a “delayed plating” experiment, and is operationally defined as an increase in the surviving fraction resulting from prolonged incubation of cells under non-growth conditions following irradiation. If non-cycling cells are forced to re-enter the cell cycle immediately after irradiation, rather than remaining quiescent, potentially lethal damage will be “expressed” and therefore the surviving fraction will be lower. PLD is believed to be complex double strand breaks (DSBs) that are repaired slowly as compared to simple DSBs. Therefore, cells that are left in stationary phase after irradiation display enhanced survival as they have
time to repair complex DSBs before resuming progression through the cell cycle.

Sublethal Damage Repair (SLDR) denotes the phenomenon that cell survival increases when irradiated with split doses compared to a single dose of irradiation. Demonstrated by a split dose experiment. Repair occurs mostly within the first hour, starting to plateau after 2h for most proliferating mammalian cells.

Repair of DNA damage and rejoining of chromosome breaks presumably underlie both the sublethal and potentially lethal damage recovery

367
Q

What is linear energy transfer?

A

High LET = high RBE (until 100kev/um) = low OER = low energy = densely ionising = less sublethal damage repair = less dose rate effect = less fractionation sparing

High LET particles have a higher RBE in hypoxic cells than oxygenated cells (as they are equally as effective in both cells unlike the reference radiation)
High LET particles have a higher RBE in fractionated regimens than acute regimens as they don’t show sparing caused by fractionation.

368
Q

Tumour response to radiation (cellular level)

A

Tumor response following radiation into has been described to occur in 4 discrete stages where stage I involves massive endothelial apoptosis or at least stopping of endothelial proliferation, stage II involves tumor regression via tumor cell death, stage III involves early regrowth of vessels from remnant endothelial and tumor cells with some growth factor support from myeloid bone marrow-derived cells, and stage IV involves late recurrence due to the tumor bed effect mediated by defective neovascularization.

369
Q

What are the effects on radiation on the testis?

A

Dose fractionation increases the risk for sterility in the male; the TD5 and TD50 for sterility are 2 Gy and 8 Gy, respectively, for a single dose of X-rays, whereas these values decrease to 1 Gy and 2 Gy for fractionated irradiation. This effect results from spreading the dose over time permitting reassortment sensitization to occur for spermatogonia, which have a large variation in radiation sensitivity through the course of their cell cycle, and more than compensating for any repair that might occur between fractions. Thus, spermatogonia located in a relatively radioresistant portion of the cell cycle may progress into a more radiosensitive part of the cell cycle at the time of the second and subsequent irradiations.
Spermatids and spermatozoa are relatively radioresistant, whereas spermatogonia are
radiosensitive. A drop in testosterone levels would not be detectable following a scattered dose of 0.1 Gy to the testes, particularly to an adult. Following a moderate dose of radiation, which kills a large number of spermatogonia, there may be relatively little effect on the levels of spermatocytes, spermatids and spermatozoa initially, since a period of 67 days is required for maturation of a spermatogonial stem cell to a mature spermatozoan. Hence, there may be very little drop in sperm count for the first month following irradiation, although the sperm count will decrease at a later time. Full recovery of a normal sperm count following radiation-induced azoospermia caused by exposure of the testes to a dose of 6 Gy, would require a period of at least 2 years.

Based on animal data, a
minimum waiting period of 3-6 months is recommended for both men and women before attempting procreation following radiotherapy in order to reduce the risk of radiation-induced genetic effects.

Hormone failure TD/5 is 30Gy as Leydig cells can tolerate much higher doses - these secrete testosterone.

We therefore see sterility with minimal effect on libido.

370
Q

What are isoeffect curves for?

A

When plotted as the log of the total dose to produce a given isoeffect as a function of the log of the dose per fraction (plotted on a reverse scale), most late
responding normal tissues are characterized by steep isoeffect curves, whereas those for early responding normal tissues and most tumors tend to be shallow.
They allow identification of the optimal range of fraction sizes to use for treatment based on isoeffect on tumour control probability and late normal tissue complication probability.

Isoeffect curves are often plotted with the log of the total dose on the y-axis and the log of the fraction size (from high to low) on the x-axis. Tissues with a greater
repair capacity will show greater sparing with increasing Fractionation (smaller fraction sizes) and therefore will have steeper isoeffect curves.

371
Q

What is an endonuclease? What is an exonuclease?

A

An exonuclease cleaves one nucleotide at a time beginning at the end of a DNA strand.

Endonucleases produce nicks within intact
DNA strands

372
Q

What are radiation effects on the ovaries?

A

Sterilization (different to testis – fixed number from birth - EXTREMELY RADIOSENSITIVE &
IMMEDIATE) - TD5/5 2Gy

Menopause (Hormonal secretion is associated with follicular maturation, sterilisation by radiation leads to a loss of libido and menopause) - TD5/5 20Gy

373
Q

What is the lethal dose of total body irradiation?

A

LD50/60 - radiation dose that leads to death within 60 days of 50% population.
3.5Gy without medical treatment (die of haematopoietic syndrome)
7Gy with medical care (die of GI syndrome)

374
Q

What symptoms do you get for each dose of total body irradiation? What is the treatment?

A

Time course and severity of clinical features are a function of overall body volume irradiated, inhomogeneity of dose exposure, absorbed dose, dose rate and particle type.

> =1Gy = prodromal reaction.
1Gy (mild)-2.5Gy (significant) = haematopoietic syndrome - time to death is 1-2 months.
5Gy (mild)-8Gy (significant) = GI syndrome = death in 3-10 days.
20Gy = cerebrovascular syndrome = death within 24-48hrs

Treatment:
Accidental 2Gy exposure: monitor at home, prophylactic antibiotics and monitor blood counts
Accidental 3Gy exposure: haematopoietic syndrome - hospitalised in reverse air flow isolation with supportive care and observation for 2-4 weeks, prophylactic antibiotics, transfusion of blood products.
Accidental 8Gy exposure: likely death due to GI syndrome. Supportive care includes symptomatic management with antiemetics, IV fluid and electrolyte replacement. Can consider bone marrow transplant if survive the GI syndrome Don’t give steroids.
Accidental 10Gy exposure = death.

374
Q

Discuss the haematopoietic syndrome caused by total body irradiation?

A

Occurs mildly at 1Gy, significantly at 2.5Gy.
The chronological order for decline of the components of peripheral blood after irradiation is lymphocytes, granulocytes (7-10days), platelets (20 days) and lastly erythrocytes (based on transit time so time to drop - as the cell that actually dies is the bone marrow stem cells.

Death in 1-2 months with no treatment. Death due to infection and haemorrhage.

Treat with antibiotics and blood product transfusions and reverse air room and supportive care.

375
Q

Discuss the GI syndrome caused by total body irradiation?

A

Mild symptoms ats at 5Gy, significant symptoms at 8Gy, definite death at 10Gy.

Nausea, vomiting, blood diarrhoea (due to depopulation of epithelial lining due to sterilisation of crypt cells). Death in 3-7 days. Small bowel paneth cells are most sensitive.

Treatment is supportive e.g. antiemetics, IV fluids, IV electrolytes, psychological support. No steroids. (Note if they survive the GI syndrome then you have to manage the haematopoeitic syndrome e.g. with bone marrow transplant)

376
Q

Discuss the cerebrovascular syndrome caused by total body irradiation?

A

Occurs at 20Gy. Death within 24-48hrs - no treatment.

Cerebral oedema, microvasculitis, brain necrosis.

377
Q

Discuss the prodromal phase of radiation sickness?

A

Nausea, vomiting, anorexia, fatigue = sublethal dose.
Fever, diarrhoea, headache, hypotension, apathy, skin erythema = supralethal dose
Occurs quicker and more severe with higher doses - can get vomiting within a few minutes with high exposure - usually starts within 2-6hours. Duration of phase is inversely proportional to dose - at high doses go straight to the manifest symptoms e.g. cerebrovascular syndrome. Usually 1-3 days. Can get a latent phase with “apparent cure” after prodromal phase before the patient gets the manifest illness phase - this latent phase can last hours-weeks.

378
Q

How do you detect total body irradiation?

A

the minimum whole body dose that can be detected through measurement of dicentric chromosomes in peripheral blood lymphocytes is approximately 0.25 Gy.

The most reliable approach to estimate dose one month following a radiation exposure is to karyotype peripheral blood lymphocytes to detect chromosomal aberrations, particularly dicentric chromosomes, which are normally not found in unirradiated people.

379
Q

What do we know about retreatment with radiation?

A
  1. If the tolerance within a given tissue volume has already been exceeded during the first treatment, and loss of function is present or expected soon, then re-irradiation is not possible without loss of function.
  2. For early effects, restitution of the original tolerance may be complete after low to moderate doses, and after tissue-specific and dose-dependent time intervals. At high doses, residual damage may remain for longer intervals, particularly at the stem cell level, which is not necessarily reflected by the number of differentiated cells in the functional tissue compartments (e.g. blood cell counts for bone marrow).
  3. For some late-responding tissues, partial (central nervous system, lung) or complete (skin) restoration of tolerance is observed after low and moderate initial doses (60 per cent of the initial tolerance). For example, spinal cord can safely be re-irradiated to a total of 140 per cent of the initial EQD2tol.
  4. In some late responding tissues (kidney,
    urinary bladder), progression of damage at a subclinical level must be expected, thus precluding re-irradiation without exceeding tolerance.
  5. Alternative treatment options must be considered before re-irradiation.
  6. If (curative) re-irradiation is to be administered, optimum treatment planning (dose conformation) and a proper choice of fractionation protocol (hyperfractionation) are required