Unit 2 - Traditional Therapies Flashcards

1
Q

Who first had the idea of radiation therapy in cancer and who first showed that it could be used after surgery to minimise the radicalness of the surgery

A

Emil Grubbe

Fisher and Veronesi

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

What radioactive molecules were among the first two be used in cancer treatment?

A

Radon gas and radium capsules

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

Describe the history of chemotherapies

A
  • Originally with Paracelsus in the 1500’s use of arsenic, lead and mercury (see also Pare’s La method)
  • Paul Ehrilich 1st researcher looking at selectivity in drugs in many diseases using dyes and postulated the magic bullet idea
  • Goodman and Gilman -> toxic mustard gasses from WWI to be used in lymphoma/leukaemia and went onto develop cyclophosphamide
  • Sidney Faber rationalised use of anti-folates like aminopterin (block dihydrofolate reductase in nucleotide synthesis) -> many similar molecules work in a similar way like methotrexate and 5-FU
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Who were the first people to use combinations of chemotherapies – what agents did this include?

A

Frei and Freireich – VAMP regime (60’s/70’s) – Vincristine, amethopterin, 6-mercaptropurine and prednisone in childhood leukaemia

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

Who were the first people to look at adjuvant chemotherapy

A

Bonnadona and Fischer used 5-FU, methotrexate and cyclophosphamide in breast cancer to improve survival and prevent relapse -> lead to development of neoadjuvant too

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

What was the first discovered oncogene and who discovered it?

A

Ras and Robert Weinberg

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

What is brachytherapy and how does it differ to other kinds of radiotherapy?

A

Brachytherapy is where the radiation is placed inside the body!! Although patients can be radioactive after treatment, so may have to isolate after therapy!!

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

What proportion of cancer patients have radiotherapy?

A

About 50%, generally more for radical intent but some cases are associated with palliative care (pain reduction, to improve breathing etc and uses a lower dose)

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

Describe some ways in which the targeted area is minimised in radiotherapy (within about 2mm)

A
  • Using imaging techniques like MRI/CT/RNI to ensure only the specific region is targeted like CT’s – often a radiation plan is produced using 3 planes around an isocentre
  • Minimise patient movement via immobilisation devices such as those for the head and neck or thorax monitoring devices which may be used to stop treatment when there is too much movement.
  • Can use multiple planes/fields to reduce dose reaching sensitive tissues
  • Certain radioactive substance which are specific to tissue (e.g., I-131 in thyroid)
  • Intensity modulated radiotherapy and multi-leaf collimation can help shape the beam to with different intensities to prevent off-target effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Describe the two types of ways brachytherapy can be given and for each of these describe how some examples of how it is used?

A

Sealed – sits within capsule (Ir-192)
Generally quite mobile devices. Given via intercavitary (existing cavity – ovarian cancer), intraluminal (catheter through the nose ->oesophagus) and interstitial (in a body cavity via an applicator which is computer controlled– such a breast and prostate) requiring many incisions.
Unsealed – is in form of liquid or gas (I-131) and dissipates throughout the body
Radioactive isotope therapy (given via IV/orally) – isotope will generally accumulate in a certain tissue to minimise off-target effects – not recovered- therefore radioactive and may need to isolate/minimise contact.
External beam – delivered by linear accelerators (Co-60) – directed outside the body, usually combined with a CT scanner to avoid other organs (image quality not as good)

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

What type of beam energies are used in radiotherapy and where would they be used?

A

Kilovoltage – uses electronic beam energy
Superficial X-rays (90-150kv) – X-rays on or near body surface
Deep X-ray therapy – 150-300kv – higher energy and penetrate deeper ( a few cm’s) – vertebrate metastatic disease
Megavoltage (6-10MVs) via a linear accelerator (large structures) in X-rays and particles. Can be used for deeper tumours. Will also have imaging panels attached to aid accuracy as well as light to guide the rays!

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

What are the acute and chronic side-effects of radiotherapy treatment?

A

Acute – damage to skins like erythema and dry desquamation (given emollients)
Chronic – telangiectasia, lymphedema, alopecia, anaemia, organ damage
Related to the dose used, the field size. The target organ and fractionation

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

List two types of particle radiotherapy

A

Electron radiotherapy and proton radiotherapy (require large particle accelerator which is expensive x10 vs radiotherapy)

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

Discuss the relationship of energy of electron radiotherapy and its intensity at different distances.

A

Electrons produced by linear accelerator – energies between 6-20MeV – which each having higher intensities at greater distances of penetration. 75% at skin surfaces – rises to a peak (2-6cm) deep and then a sharp drop-off – minimises off-target effects!!

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

How may radiotherapy cause damage to non-tumorous cells?

A

The ionising radiation can result in DNA damage, primarily SS/DS breaks – the higher the dose the more damage. This will result in cell cycle arrest at G1/S and G2/M if this damage is not repaired ->apoptosis!!!
The patient may not see damage immediately as cells go through the cell cycle at different times/rates.

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

Rank the four stages of the cell cycle in terms of radiosensitivity

A

Most sensitive: mitosis , G1/2, S-phase

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

What is a therapeutic ratio?

A

The therapeutic index describe the difference in susceptibility of damage to target (tumour) cells compared to non-target cells at a given dose.

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

What are the three phases in a radiation event- describe each of these in detail?

A

Physical phase – very quick
Incoming photon interacts with orbital electrons – ionisation – and the photon is scattered to interact with another atom (Compton Scatter)
Chemical damage – within 1ms
Primary target is water, radiolysis results in ROS like hydroxyl free radical – SS/DB breaks and 8-Oxo-G
Biological phase – over longer periods (months)
Where the DNA damage can be (or not) repaired – this stage looks at the repair by enzymes or the subsequent cell cycle arrest/apoptosis – which can result in side-effects (anaemia/alopecia/lymphedema/erythema…)

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

What is the oxygen effect in radiotherapy?

A

The amount of oxygen present plays a significant role in the effectiveness of radiotherapy, decreasing amount decreases efficiency!!
This is because most DNA damage in radiotherapy which interact a free-radical which interacts with oxygen to form free-radical species (short half-life) – hence hypoxia will result in less of these species hence less direct damage to DNA.
A large proportion of tumours are actually hypoxic (can changes over time)– which means these regions will likely survive treatment and be able to allow tumour recurrence.

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

What ways can we enhance the oxygen enhancement ratio to aid radiotherapy?

A
  • Supplying additional oxygen to the patient
  • Improve patients own oxygen delivery (exercise/health)
  • Monitor levels of haemoglobin to suspend treatment to ensure most effective efficacy – Cherenkov excited luminescence imaging
  • Oxygen mimic drug (nitroimidazoles)
  • Selectively targeting hypoxic cells via hypoxia activated prodrugs like mitomycin C
  • Increase oxygen flow by oxygen carriers
  • Target DNA repair pathway – 5-FU or ATR/Chk1 inhibitors in early development
  • Vasodilation (nicotinamide)
  • Anti-angiogenic agents
  • Hyperbaric chambers
    But if they will actually increase oxygen at permanent hypoxic areas may effect efficiency!! Onion peel model!! – But they may help to reduce the dose required to reduce impact on non-tumorous tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Using the 5R’s describe the impact of fractionation on radiotherapy results?

A

Radiosensitivity
Each cell has its own radiosensitivity – some cells are very radioresistant like melanoma so there is no point using it here. The amount of radiation per fraction and number of fractions is based on the radiosensitivity of the cells.
Reoxygenation
Increased oxygen concentration increases the susceptibility to radiation damage and will prevent recurrence due to (temporary) hypoxia – hence fractionation reduces this impact and increases the chances of ‘catching’ the tumour cells when they are not hypoxia (only temporary)
Reassortment
Efficacy also depends on the stage of the cell cycle, those in M-phase are more sensitive – hence like above – increased fractionation means that over the entire regime it is likely you will catch these cells in their most sensitive cell cycle state – again to prevent recurrence
Repopulation
The space between doses will allow tumours to proliferate and re-populate the tumour – meaning an increasing dose would then be required – which could be a disadvantage – this could be due to the enriched nutrient supply to these tumours. But normal tissue has more time to recover. Accelerated radiotherapy protocols can be used for some tumours with fast repopulation rates
Recovery
Where cells sustain sub-lethal damage, the longer period between radiotherapy can increase the chance of recovery and a higher dose is required.

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

In radiotherapy, what is the tolerance dose?

A

This is the maximum amount of radiation a tissue can receive before becoming permanently damaged – it depends how much of the organ is present in the radiation field (although shielding can sometimes reduce this). The TD is given as X/Y – X is the % probability of complication after Y years!!

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

What side-effects are most chemotherapies associated with and why?

A

Chemotherapies are usually associated with blocking certain stages of the cell cycle – of which all cells progress through. These agents preferentially target rapidly dividing cells, which will include tumour cells, but also other fast-dividing cells like red blood cells (anaemia), white blood cells (immune deficiency), gastrointestinal issues and hair loss.

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

What factors can influence the efficacy of a chemotherapy?

A
  • Tumour cell burden
  • Sensitivity – do they up-regulate MDR1 or other pro-survival genes (DNA repair for alkylating agents)
  • Dose intensity
  • Treatment schedule
  • Proportion of tumour population that is actively dividing
  • Location of the tumour (BBB)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is the fractional kill hypothesis and why does this explain why chemotherapy is given in cycles?

A

At a given a proportion of tumour cells will be dividing – so only a few of the cells will be targeted by these agents – hence only a proportion of the tumour population will be killed (growth fraction) – also treatment schedule, dose intensity, sensitivity and tumour cell burden are important.
Therefore, sustained chemotherapy will mean that not all tumour cells will be killed and there are increased side-effects to the patient. Hence it is given in cycles.
This is a cycle of treatment and cessation for the normal cells (as well as some tumour cells) to recovery to maximise tumour cell death whilst minimising damage to normal cells!!

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

What does neoadjuvant chemotherapy mean?

A

That chemotherapy is given before surgery to shrink the tumour and perhaps make the surgery less complex.

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

Give an example of chemotherapy in palliative care

A

Gemcitabine in pancreatic cancer to prevent bone compression on nervous tissue

28
Q

Give an example of where chemotherapy can be used with another modality

A

5-FU to radiosensitize cells (radiotherapy) prevents DNA repair

29
Q

Give ways that drugs can be administered

A

IV/IM/ subcutaneously
Orally like capecitabine
Intrathecal – between peamator and arachnoid mator in spinal cord (leukaemia/lymphoma) like methotrexate OR ommaya reservoir in the scalp
Intraperitoneal – catheter for ovarian cancer (cisplatin)
Hepatic artery (catheter for liver) again cisplatin
Surgical implantation like wafers in the brain that release carmustine

30
Q

Why is Paclitaxel given before Cisplatin

A

The two agents are synergistic – if cisplatin is given first then it can damage tubular cells of the kidney impairing its function – hence preventing the excretion of paclitaxel which will subsequently have a longer half-life and be associated with greater toxicity

31
Q

What pre-medication is given before paclitaxel therapy?

A

Generally given antihistamines and corticosteroids to prevent hypersensitivity reactions has it is emulsified with a Cremophore and ethanol to improve pharmacokinetic parameters (aqueous solubility) which may increase its chances of a reaction

32
Q

Why is paclitaxel not given alongside cimetidine or clopidogrel?

A

Cimetidine is a histamine receptor antagonist (heart burn) and clopidogrel is a blood thinner (heart disease). They both inhibit members of the CYP family (CYP2C8 and CYP3A4) which are involved in metabolising paclitaxel – hence inhibiting these enzymes will reduce the clearance of paclitaxel resulting in increased levels and increased toxicities.

33
Q

How do alkylating agents work? Give examples

A

They interact with nucleotides, usually purines in the major groove (G7) – this can cause kinks in the DNA preventing it from being transcribed or replicated and potentially strand cleavage – which will result in G1/S cell cycle arrest (but can work at all phases)
Examples are carmustine and nitrosoureas (cross BBB)

34
Q

How do anti-metabolites work and give some examples?

A

They prevent the synthesis of nucleotides which is essential for DNA replication and transcription – nucleotide depletion leads to cell death!
1) Hydroxyurea inhibits ribonucleotide reductase – nucleotide depletion
2) Methotrexate (via dihydrofolate reductase inhibition) – nucleotide depletion
3) Capecitabine (->5-FU) inhibits thymidylate synthase which inverts dUMP to dTMP and can increase levels of dUTP which can be misincorporated into DNA. 5-FU can also be incorporated into the RNA – nucleotide depletion
4) Mercaptopurine – inhibits purine synthesis early in process and can be further converted into molecules that can be converted into DNA and RNA blocking the synthesis of these polymers – nucleotide depletion and DNA synthesis inhibitor.
5) Gemcitabine (after modification in cells) which can also be inserted into DNA preventing elongation (DNA replication) – inhibits ribonucleotide reductase and can be inserted into DNA blocking replication.
Work during S-phase (where DNA replication occurs) - So both inhibit nucleotide synthesis and can be incorporated into DNA

35
Q

How do anti-tumour antibiotics work, give an example?

A

They create double-strand breaks in DNA strands – bleomycin chelates iron and intercalates between DNA and results in irons oxidation. The liberated electron can result in superoxide free radicles which can result in SSB/DSB and potential oxidation of Guanosine leading to mispairing!

36
Q

Name some topoisomerase inhibitors involved in cancer treatment, how do they work?

A

Doxorubicin (TOPII) and Topotecan (TOPI) – these enzymes are essential for unwinding DNA in processes like transcription and replication.

37
Q

How does platinum agents work in chemotherapy, give examples?

A

They act in a similar fashion to alkylating agents, usually interacting/crosslinking two guanosine bases in close proximity – resulting to intra (more)/interstrand adducts which can cause DS/SS breaks.
Examples – cisplatin/oxaliplatin

38
Q
  1. How do vinca alkaloids act – give examples
A

They bind to beta-tubulin preventing microtubule polymerisation – arresting the cells in metaphase of the cell cycle
Most are extracted from the Madagascan Periwinkle – vincristine, vinblastine, vinorelbine

39
Q

What is the evidence that oestrogen is important in driving breast cancer?

A
  • The rates of breast cancer are much higher in women compared to men
  • It is related to age (exposure)
  • Earlier menstruation and later menopause increase the chances
  • Protective effect of removing ovaries
  • Oestrogen receptor is overexpressed in many breast cancer cells in the luminal classes
40
Q

Describe the four pathways involved in E-ER signalling?

A

The classical pathway
E enters the cells and binds to ER, causes conformational changes with helix 12 sealing the lid. Loss of Hsp90 allows dimerization. E-ER dimer ten binds to ERE on DNA to promote transcription of cyclin D1 and Myc (by AF2 domain now able to interact with co-activators like SRC to recruit co-activators.
Tethered
E-ER binds to TFs like Fos and Jun which can then recruit co-activators to promote transcription
Non-genomic
Interacts with GPR30 in calveoli rafts->promote cAMP and EGFR signalling
Genomic-non-ligand bound
Various molecules can phosphorylate the AF-1 domain (MAPK118) which increase its ability to recruit various co-activators

41
Q

Describe the importance of antihormone therapy in breast cancer.

A
  • Is beneficial in the majority those with early and advanced ER+ disease
  • Has increased the 5-year survival rate significantly (<80%)
  • Is useful after surgery in advanced cases
  • Can be used alongside other agents like CDK inhibitors
  • Patients can be cycled through many different anti-hormones before late-line chemotherapy
42
Q

List the three kinds of anti-oestrogens

A
  • Non-steroidal antioestrogens which block E-ER binding like tamoxifen (SERMs – selective oestrogen receptor modulator)
  • Competitive inhibitors with additional effects on ER – steroidal anti-oestrogens (faslodex) (SERDs – selective oestrogen receptor down regulator)
  • Block oestrogen production – aromatase inhibitors anastrozole and LH-RH agonists like Zoladex
43
Q

How do non-steroidal anti-oestrogens work and who is it used in?

A

Tamoxifen mimics oestrogen (and can be converted to 4-OH-Tam by CYP450’s) hence can compete with it for binding to oestrogen receptor pocket. Binding results in similar changes (loss of HSP90, dimerization and binding to ERE’s) but the bulky alkyalminoethoxy side chain and trans configuration of tamoxifen prevents helix 12 from sealing the pocket – so helix 12 then blocks the AF2 regions so it cannot recruit activators like SRC.
Used in ER+ pre-menopausal breast cancer patients or in patients who cannot take other forms of anti-ER therapies.

44
Q

What side-effects are associated with tamoxifen use?

A
  • Menopausal-like hot flashes
    Also can oestrogenic effect – as its activity does not effect the AF1 domain which can still be phosphorylated can help recruit co-activators – this is the dominant form in some tissues depending on levels of activators/repressors or kinase activity… this results in…
    Good: anti-osteoporotic and can low blood cholesterol levels
    Bad: Stimulates uterus (uterine cancer) and thromboembolic events (by decreasing liver levels of anti-thrombin
45
Q

How do the ER-inhibitors with additional effects/Steroidals (SERDs) work and how do they compared to tamoxifen?

A

Faslodex are more than 100-fold more effective at ER binding than tamoxifen as they are oestradiol derivatives – and also there is no pro-oestrogenic activity.
The very long side chain disrupts many functions of ER –prevents helix 12 closure - preventing dimerization and nuclear localisation, resulting in degradation in the cytoplasm. This means both AF-1 and AF-2 activity are blocked.
It can be used in post-menopausal women when anti-hormones fail (with agents like CDK4/6 inhibitors) and is the only agent used for those with mutated (hyperactivated) ER. Also gives fewer hot flushes as it doesn’t cross the BBB and has no agonistic activity.

But is has to be given IM injections which limits the dosage – although non-steroidal antagonists like Elacestrant are under investigation which can be taken orally!

46
Q

What is the structure of aromatase and what reaction does it carry out?

A

Made of two components:
P450 aromatase – a haemoprotein responsible for binding the androgen and carried out the reduction reaction
Flavoprotein NADPH-cytochrome P450 reductase – takes an electron from NADPH to p450 aromatase
This helps carry out a reduction reaction producing a C18 steroid (oestrogen) from a C19 steroid (androgen)

47
Q

Where is oestrogen produce in premenopausal women and post menopausal women?

A

Pre-menopausal women – lots made during menstrual cycle in ovarian granulosa cells
Post-menopausal – peripheral aromatisation in fat/muscle cells

48
Q

What group of patients are aromatase inhibitors like letrozole/anastrozole used in?

A

In postmenopausal ER+ patients (lots better than tamoxifen) – but in younger women this would result in a gonadotropin surge (feedback loop) which will result in hyper stimulation of ovaries to produce more oestrogen.
Can also prevent relapse and be used preventively in high risk groups.

49
Q

Giving examples, describe the two types of aromatase inhibitors

A

Steroidal – irreversible -compete with active site after enzyme metabolism – exemestane
Non-steroidal – reversible – compete with androgens for the active site due to azole nitrogens binding haem group – letrozole and anastrozole

50
Q

What are the side-effects of aromatase inhibitors?

A

Fracture/joint problems and sever hot flushes – but no uterus stimulation or thromboembolism events

51
Q

What treatments are available for premenopausal women in breast cancer.

A

LH-RH agonists like the peptide mimetic Zoladex.
This hormone stimulates LH-RHr in the pituitary gland which release LH and FSH which stimulate oestrogen synthesis – hence blocking this will prevent stimulation of synthesis of oestrogen!!
Importantly in young women this is reversible.
Is associated with hot flushes, bone loss and temporary menopause and can be used with tamoxifen.

52
Q

Describe the ways in which androgen is produced.

A

LHRH (GnRH) is made by the hypothalamus and binds to GnRH receptor on the anterior pituitary gland which release FSH and LG into the blood. LH binds to receptors on Leydig cells in the testes which produce testosterone from androgens and FSH binds to receptors on nurse cells which support the Leydig cells.
Residual androgens can also be made from the adrenal gland like DHEA
Testosterone and inhibin (from Sertoli cells) can feed back on this axis to regulate this

53
Q

How can testosterone be taken up by cells and promote proliferation?

A

Testosterone is taken up by cells and converted to 5-alpha dihydroxytestosterone by 5-alpha reductase (the potent metabolite) which then binds to the androgen receptor in the cytoplasm.
This binding results in a conformational changes, dissociation of HSP70 proteins and dimerization with the helix 12 sealing the androgen in the pocket revealing a hydrophobic AF-2 domain to bind LXXLL motifs.
The conformational change also exposes the NLS allowing nuclear localisation (via importin proteins) and eventual binding to the AREs in the androgen-regulated genes allowing it to recruit FOXA1A (unwinds chromatin) and SRC to promote gene expression
AF-1 domain can also be activated by phosphorylation e.g/, Akt-s213

54
Q

What is PSA?

A

Prostate-specific androgen – levels of this in the blood are used to help diagnose prostate cancer – higher levels are associated with higher disease risk/grade and can be monitor patients and help predict relapse. But high levels can be caused by other factors like BPH or infection!
Is an ARE-expressed gene!!!!

55
Q

Describe the various androgen deprivation therapy methods?

A

Blocking testosterone production by the testis by – surgical castration or chemical castration (GnRH agonists) and blocking androgen activity by AR antagonists like bicalutamide/enzalutamide
Continuous GnRH agonists are used with short-term use of AR antagonists

56
Q

Why are prostate cancer and breast cancer so reliant on anti-hormone therapy?

A

Because both cancers have a significant proportion of patients who are ER/PR+ and this signalling is usually essential in driving proliferative signalling!
Although these are often given with chemotherapy or surgery

57
Q

What are the disadvantages of androgen deprivation therapy?

A
  • Side effects: Erectile dysfunction, loss of libido, muscle loss, osteoporosis, infertility
  • Acquired resistance is common within 2-3 years – this castration resistant form is generally incurable
58
Q

Describe surgical castration in ADT in prostate cancer.

A

This would have commonly been a bilateral orchidectomy but now a suprascapular orchidectomy – only 5% of patients have this because it is reversible (physical and psychological effects) but is an option in non-sexually active men or those non-compliant to other ADTs.
But can have a rapid reduction on testosterone levels.

59
Q

Describe the mechanism of action of chemical castration?

A

GnRH is a decapeptide which is involved in the release of FSH and LH from the pituitary gland. G-6 residue is important in its degradation (short half-life).
Super-agonists are peptide mimetics which are modified at G6 so they are no longer degraded so its levels are sustained as well as G10 which can enhance binding to the receptor like Zoladex.
This continuous agonistic activity will reduce the levels of the receptors, desensitising cells to GnRH (after an initial surge of LH and FHS – takes a few weeks to be seen)

60
Q

What are the advantages and disadvantages of GnRH agonists like Zoladex.

A

Advantages:
- Avoids traumatic and irreversible surgery
- Effective in advanced disease in high-risk early disease with chemotherapy
Disadvantages
- Must be given every month as injection and may take a while to achieve stable reduction
- Side-effects of ADT
- Only blocks testicular synthesis – doesn’t effect adrenal glands
- Flare of testosterone in initial treatment may accelerate cancer and associated problems

61
Q

How are the surges/flares of testosterone caused by Zoladex treatment in prostate cancer being overcome?

A

GnRH antagonists – like Degarelix which are also decapeptides but do not cause agonistic behaviour due to modifications of first three residues.

62
Q

Describe with examples the activity of anti-androgens

A

Original molecules non-steroidal (bicalutamide) These molecules bind to the pocket of AR, out-competing the androgen – it still allows dimerization and ARE binding – but prevents activation of AF-2 by preventing Helix-12 folding and co-repressors are bound instead. But allow A-F1 activity.
2nd generation molecules like enzalutamide have no agonistic activity
These molecules are usually not given as a monotherapy as they promote a normal feedback mechanism and the testosterone levels eventually increased – but can be used with younger patients due to less side-effects

63
Q

Describe the ways in which prostate cancer patients can become anti-hormone resistant.

A
  • Residual androgens – or produced by the tumour
  • AR overexpression/amplification – in around 50%
  • Ligand-independent activation through PTMs of AR due to signalling of IGF, EGF)
  • AR LBD mutations or splice variants (ARV567) – cant bind enzalutamide or mutations that can increase affinity of other ligands like oestrogen or progesterone!
64
Q

What molecules are used to target castration-resistance prostate cancer?

A
  • Taxanes with…
  • 2nd generation anti-androgens
  • Abiraterone acetate to block CYP17A1 to inhibit androgen synthesis or via dexamethasone

Olaparib too being investigated

65
Q

Describe how Proton therapy intensity differs through tissue depth

A

Will have low intensity at low tissue depths and then have a sharp peak around 15cm into tissue, whilst will then drop suddenly
Has ability to target deep tumours with minimal toxicity

66
Q

How can the Bragg Peak be spread out in particle radiotherapy?

A

Using beams of various energies together

67
Q

What percentage of prostate cancers are dependent on AR signalling

A

80%