Pharmacology Flashcards

1
Q

Which drugs are alkylating agents?

A

Cyclophosphomide
Ifosfomide
Temozolamide
Mitomycin C (also tumour antibiotic)

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

How do alkylating agents work?

A

Transfer an alkyl group (CH3+) to guanine base on DNA in N7 position of purine ring.
Can be monofunctional (one DNA strand) - prevents effective DNA replication (NO crosslinking)
Bifunctional - majority- 2 seperate DNA strands to form a covalent crosslink (inter and intra stand crosslinks)

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

Describe the MOA of cyclophosphamide

A

Prodrug -> metabolised by p450 -> 4-hydroxycyclophosphamide which is in equilibrium with aldophosphamide
Aldophosphamide -> phosphoramide mustard (active) + acrolein (urotoxic)
Aldophosphamide deactivated by aldehyde dehydrogenase (levels high in GI and haem so less toxic)

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

where does cyclophosphamide work in the cell cycle?

A

Non specific

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

How can you give cyclophosphamide?

A

Oral or IV

GOod oral bioavailability 75%

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

WHat is the distribution of cyclosphosphamide?

A

Throughout body including brain and CSF, 60% phosphoramide musturd bound to plasma proteins

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

How is cyclophosphamide excreted?

A

Urine

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

Would you dose reduce cyclophosphamide in renal/liver impairment?

A

If CrCl <20 75% dose, if <10 50% dose

Liver: risk of decreased activation and increased risk of veno-occlusive disease

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

When might you use ifosphamide?

A

Sarcomas, germ cell tumours, VIP, paediatric tumours

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

What is the MOA of ifosfamide?

A

Activated by cytochrome p450 enzymes -> 4-hydroxyifosfamide which is in equilibrium with aldoiphosphamide -> ifosfamide musturd (active) + acrolein

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

What causes neurotoxicity with ifosfamide?

A

Chloroethyl side chain oxidation (occurs more than cyclosphosphamide) -> accumulation of chloroaldehyde which is neurotoxic

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

What is excretion of ifosfamide?

A

Urinary 50-70%

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

Would you dose reduce in liver/renal impairment with ifosfamide?

A

CrCl <60 give 70% dose. Cr CL <45 don’t give.

Not recommended if bili >ULN or ALT/ALP >2.5x

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

What type of drug is temozolamide?

A
Alkylating agent
Trazine analogue (similar to decarbazine)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Are alkylating agents cell cycle specific?

A

No

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

Describe the MOA of temozolamide?

A

Spontaneously decomposes to MTIC at physiological pH
Precise mechanism unclear- drug methylates guanine residues in DNA and inhibits DNA, RNA and protein synthesis.
does NOT cross link DNA strands.

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

What is the bioavailabiliy of TMZ and does it cross blood brain barrier?

A

Oral - 100% bioavailable

Lipophylic so crosses BBB

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

How is TMZ metabolised and excreted?

A

Metabolism: non enzymatic hydrolysis

Urine

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

Do you need to dose reduce TMZ in renal/liver impairment?

A

no, caution if severe

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

What are mechanisms of resistance to alkylating agents?

A

Glutathione inactivates all alkylating agents ( increase these levels)
Glutathione-S transferase catylases the conjugation of glutathione with alkylating agents (increased levels)

Induction of DNA repair- O6 alkylguanine-DNA alkyltransferase enzyme (MGMT) removes alkyl group from O6 guanine. MGMT levels assoc with resistance in glioma

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

What are the side effects of alkylating agents?

A

Myelosuppression ( cyclophosphamide plt sparing due to high ADH in megakaryocytes), nadir 8-16 days, recovery by D20. Prolonged with nitrosureas + mitomycin

Alopecia - as lipophilic- mostly with cyclo/ifosfamide

GI toxicity - less emetogenic than platinium
CNS toxicity - mainly ifosfamide
GONADAL- testicular failure (depletion of germ cells) dose dependent, may be reversable. Ovarian failure.
Haemorrhagic cystitis
IP + fibrosis

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

Which classes of chemotherapy drugs cause alopecia?

A

Alkylating agents, taxanes, vincristine and anthracyclines

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

Describe the CNS toxicity caused by ifosfamide, its treatment and the risk factors?

A

Accumulation of neurotoxin chloroaldehyde
From mild confusion -> somnolence, seizure, coma
Ifosfamide induced encephalopathy - onset 2-48hrs post dose, resolves on stopping drug over 1-3 days
Methylene blue - used as treatment
Risk factors: low albumin, alcohol excess, renal failure.

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

How do you treat haemorrhagic cystitis?

A
Irritation of bladder mucosa by urinary metabolites (cyclophosphamide and ifosfamide) -> acrolein, phospharamide musturd and chloroacetaldehyde irritate bladder
Always adminster:
Mesna - detoxifies metaolites
Hydration
Close observation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

How do antimetabolites work?

A

structually similar to metabolites so mistaken by the cell and processed - interfere with production of nucleic acids/DNA/RNA -> block purine/pyramidine synthesis

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

Which part of the cell cycle do antimetabolites work in?

A

G1/S phase

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

What is 5FU?

A

Antimetabolite

Pyramidine

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

What is the mechanism of action of 5FU?

A

Inert drug
5-FU -> F-dUMP which competes with natural dUMP for catalytic side of thymidylate synthase that produces dTMP

F-dUMP forms a complex with TS and acts as a suicide inhibitor -> causes decrease in dTMP & dTTP levels and dUMP and dUTP accumulate.
(principle MOA)
Also get FUTP misincorperation in RNA and FDUTP misincorporation into DNA>

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

How is 5FU given?

A

Oral absorption unreliable
IV pharmacokinetics vary depending on duration of infusion
- bolus vs continuous
Half life short- ? increased duration of exposure improves survival

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

How are the side effects different whether you give 5FU as a bolus or continuous infusion?

A

Bolus - neutropenia, mucositis and diarrhoea

Infusion- hand-food syndrome, (diarrhoea and mucositis but worse with bolus)

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

How is 5FU excreted?

A

Biliary secretion- stool

<10% renal

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

How do you dose reduce 5FU in liver or renal impairment?

A

Renal - nil

Liver- moderate impairment reduce by 1/3, severe reduce by 1/2

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

What is the MOA of capecitabine?

A

Absorbed GI tract as pro-drug
Undergoes 3 step metabolsim
Capectabine -> via hepatic carboxylesterase -> 5’DFCR (5′-deoxy-5-fluorocytidine)-> via cytidine deaminase -> 5’DFUR -> thymidine phosphorylase -> 5-FU

Thymdine phosphorylase is preferentially expressed in tumour tissue providing a degree of targeting for generation of 5-FU in tumour.

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

How is capecitabine excreted?

A

Mainly as metabolites - renal

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

How would you dose reduce capecitabine if renal/liver impairment

A

CrCL 30-50 75% dose
CrCl <30 C/I

Bili >3x, ALT/AST >2/5x omit

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

What are the main side effects of cap/5FU?

A
DIarrhoea
PPE
pancytopenia
Cardiac- ECG changes/chest pain
Increase in liver enzymes
N+V
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What is DPD deficiency?

A

Dihydropyrimidine dehydrogenase usually degrades thymine and uracil
Widely present in liver
AR, mutation DYPD gene on chromosome 1p21
If severe notice in childhood- seizures, microcephaly etc.
Different penetrances so often unknown
Severe reaction to fluropyrimidines

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

What is gemcitabine?

A

Antimetabolite

Pyramidine

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

What is the mechanism of action of gemcitabine?

A

2,2-diflurodeoxycyitidine (dFdC) - actively transported across cell membrane by nucleoside transporters as lipophilic

dFdc -> via deoxycytidine kinase -> dFdC monophosphate -> via deoxycytidylate kinase -> dFdC diphosphate (inhibits ribonucleotide reductase) -> via dFdC trisphosphate (inhibits DNA synthesis via misincorperation)

Phosphorylated
Deoxycytidine kinase is rate limiting enzyme
Incorperated into DNA/RA causing cell death

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

How is gemcitabine metabolised?

A

Extensive metabolism and deamination to dFdU in liver, plasma, tissues by cytidine deaminase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

How is gemcitabine given?

A

Not oral - as poor availability, extensive deamination in GI tract
IV infusion if <70 mins then not extensively distirbuted (half life 30-90mins), if longer than more widely distibuted (half life 4-10hrs)
FOr lung cancer given over 30 mins

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

Does gemcitabine cross BBB?

A

No

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

What drugs interact with capecitabine?

A

Warfarin

Phenytoin

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

What drugs can rescue against effects of 5FU?

A

Thymidine (only if continious infustion)- rescues TS effects

Vistonuridine

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

What are the mechanisms of resistance of 5FU?

A

Increased expression of thymidylate synthase.
• Decreased levels of reduced folate substrate 5, 10-
methylenetetrahydrofolate for TS reaction.
• Decreased incorporation of 5-FU into RNA/DNA.
Increased activity of DNA repair enzymes, uracil glycosylase, and
dUTPase.
• Increased salvage of physiologic nucleosides including thymidine.
• Increased expression of dihydropyrimidine dehydrogenase.
• Decreased expression of mismatch repair enzymes (hMLH1, hMSH2).
• Alterations in TS with decreased binding affinity of enzyme for FdUMP.

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

Which part of the cell cycle is gemcitabine specific for?

A

S phase

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

How is gemcitabine excreted?

A

Renal

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

Would you dose reduce gemcitabine in liver/renal impairment?

A

If CrCl<30 consider dose reduction

If bili >27 then dose reduce

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

What are the side effects of gemcitabine?

A
Myelosuppression (DOSE LIMITING)- neutropenia> thrombocytopenia
Mild N+V
Flu like symtpoms
Transient hepatic dysfunction (ALT, bili)
Pneumonitis
Oedema
Mild proteinutia/haematuria
Rarely HUS/TTP
Maculopapular rash
Alopecia is rare
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are the mechanisms of resistance of gemcitabine?

A

Descreased activation via decreased deoxyctidine kinase
Increased breakdown via cytidine deaminase
Decreased nucleoside transport of drug into cell
Increased expression of competing nucleotide dCTP via expression of dCTP synthetase

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

What is methotrexate used for?

A

Lymphoma
Leukaemia
Gestational trophoblastic disease
Antimetabolite- antifolate

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

What is the mechanism of action for methotrexate?

A

Enters cell via RFC (reduced folate carrier)
Undergoes intracellular polyglutamation using folylpolyglutamyl synthetase (FPGS) -> MTXPG (adds 5-7 glutamyl groups) -> competitive inhibitor of DHFR -> no thymine is made -> inhibits dna/rna synthesis
Thymineless cell

Tumour cells have higher levels of FPGS than normal cells so are more susceptible

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

What breaks down MTXPG?

A

Gamma-glutamyl hydrolase

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

How is methotrexate given?

A

Usually IV as oral bioavailability is only 40%- metabolism by intestine and deglutamation and 1st pass hydroxylation by liver

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

What is the distribution of MTX?

A

Wide
Third space accumulation which prolong drug exposure and toxicity.
If given intrathecally- can diffuse back into plasma causing toxicity

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

How is MTX excreted? What reduces the excretion

A

Renal- probably active proximal tubular secretion (distal tubular reabsorption may occur)
- reduced by weak organic acids eg NSAIDs, penicillin, PPIs

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

Would you dose reduce MTX in renal impairment?

A

Yes

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

How should you monitor a patient after giving high dose MTX?

A

Serum levels at 24hr & 48hrs
24-36hrs give leucovorin/folinic acid rescue
High volumes IVF

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

What side effects can MTX have?

A
Nephrotoxicity
Bone marrow suppression
Mucositis
Hepatotoxicity- transient transaminitis, cirrhosis with prolonged oral dosing. 
Neurotoxicity
Pneumonitis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What causes nephrotoxicity with MTX and how would you avoid and treat it?

A

Rapid drug excretion into urine at rate >solubility -> precipitation. Accumulation in renal cortex.
Amelioration
- IV fluids - aim UO >100mls/hr
- Urinary alkalinasation - Na bicarb- aim pH >7
- Avoid nephrotoxins
- avoid those that reduce renal excretion - weak acids - NSAIDs, penicillins, cipro, PPIs

Rescue Glucarpidase (costs £80,000) - not useful after 96hrs

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

How does folinic acid rescue work for methotrexate

A

5-formyl derivative of tetrahydrofolic acid
Converted to other reduced folic acid derivatives
Does NOT require action of dihydrofolate reductase

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

What is pemetrexed and what is it used for?

A

Antimetabolite- antifolate

NSCLC, mesothelioma

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

What are the resistance mechanisms of MTX?

A

Increased expression of DHFR or alterations in its binding affinity with MTX
Decreased carrier mediated transport with RFC/FRP
Decreased active FRGS or increase gamma-glutamyl hydrolase

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

What is mechanism of action of pemetrexed?

A

Transported into cell via RFC
Metabolised to higher polyglutamate form FPGS
Pentaglutamate form is 60x more potent and predominantly remains intracellular with prolonged cellular retention
INHIBITS TS -> causes accumulation of dUMP and incorperation of dUTP into DNA -> inhibition of funtion
INHIBITS DIHYDROFOLATE REDUCTASE -> depletion of reduce folates and critical one carbon carriers for cellular metabolism
INHIBITS GART & AICART which prevents de novo purine biosynthesis

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

How is pemetrexed excreted?

A

Renal 90%

NSAIDs, aspiring may inhibit renal excretion

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

What needs to be given before starting pemetrexed?

A

Vit B12

Folic acid- on regular low dose 400mcg

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

What side effects can you get with pemetrexed? When would you worry about increased toxicity?

A
Myelosuppression
Skin rash- hand-ffot syndrome
Mucositis
Diarrhoea
Transient elevation of serum transaminases
Fatigue

Wide vol of distribution so caution for 3rd space accumulation

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

What are the mechanisms of resistance to pemetrexed?

A

Increased expression of TS
Alteration in binding affinity of TS
Decreased transport of drug into cell via decreased RFC/FRP

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

What is trifluradine-tipiracil?

A

Anti-metabolite
Anti-folate
Used in mets colorectal and gastric cancer

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

How does trifluradine-tipiracil work?

A

Trifluridine taken into cell -> phosphorylated by thymidylate kinase -> TF-TMP which inhibits TS and accumulation of dUMP

Also TF-TMP can undergo further phosphorylation to TF-TTP -> misincorperated into DNA

Tipiracil inhibits the metabolism of trifluridine by inhibiting thymidine phosphorylase

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

What are the side effects of trifluradine-tipiracil?

A
Pancytopenia
Fatigue
DIarrhoea
Vomiting
Pyrexia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

How is trifluridine-tipiracil excreted?

A

Renal

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

What anti-tumour antibiotics do you know?

A

Anthracyclines- doxorubicin, epiprubicin
Mitomycin (also alkylating agent)
Bleomycin

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

What are the main biological reactions caused by anthracyclines?

A

Topo-2 inhibition
DNA intercalation
Helicase inhibition
One and two electron reduction of anthracycline molecule with production of ROS

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

What is bleomycin used for?

A

Testicular cancer
Hodgkins lymphoma (ABVD)
SCC skin

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

What is the MOA for bleomycin?

A

Small molecule that contains a DNA binding and iron binding region at opposite ends.
Iron is necessary as cofactor for free-radical generation - reduction oxygen by Fe ions chelated by bleomycin -> ROS
Binds to DNA by intercalation of bithiazole moety between base pairs of DNA
DNA-bleomycin-Fe complex -> undergo oxidation to bleomycin-Fe which releases superoxide/hydroxyl radicals -> cause SSB/DSBs DNA

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

How is bleomycin inactivated?

A

By bleomycin hydrolase (present throughout the body except skin/lungs)

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

WHat is the volume of distribution of bleomycin?

A

Rapidly absorbed - 30mins
Half life 2hrs
WIde vol. distirbution, <10% protein bound.
Accumulates in skin and lung nodes

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

How is bleomycin excreted?

A

Renal

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

What should you monitor when giving bleomycin?

A

Pulmonary function tests

CXR

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

What are the side effects of bleomycin?

A
Fever
Rash, pigmentation of skin
Alopecia
Raynauds
Hypersensitivity (premedicate with acetaminophen)
Pulmonary
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

Is bleomycin cell cycle specific?

A

Yes

G2 phase

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

What types of pulmonary toxicity do you get with bleomycin and what are the possible mechanisms that cause it?

A

Hypersensitivity pneumonintis, bronchiolitis obliterans, acute interstitial pneumonitis, pul. fibrosis
Occurs in up to 10%
May involve:
- oxidative damage
- bleomycin hydrolase deficiency - degraded bleomycin, present in all tissues except lung/skin
- stimulates alveolar macrophages to secrete inflammatory cytokines
- genetic susceptibility

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

What are the mechanisms of resistance to bleomycin?

A

Increased expression of DNA repair enzymes

Altered drug uptake

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

What are the uses of doxorubicin?

A

Breast cancer, lymphoma, sarcoma, ovarian, lung bladder, thyroid, gastric, wilms tumour

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

What is the MOA of doxorubicin?

A

Multiple mechanisms

  1. Intercalates into DNA resulting in inhibition of DNA synthesis and function
  2. Inhibits transcription through inhibiting DNA- dependent RNA polymerase
  3. inhibits topoisomerase II be forming cleavable complex with DNA and topoisomerase II to create uncompensated DNA helix torsional tension and SSB/DSBs.
  4. Formation of cytotoxic ROS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

How is doxorubicin given? Does it cross the BBB?

A

IV, not absorbed orally
Wide distribution
Does NOT cross BBB
75% plasma protein bound

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

How is doxorubicin metabolised?

A

Extensively in liver to active hydroxylated metabolite doxorubincinol then reduced by NAPH-dependnet aldo-keto reductases - one electron reduction, two electron reduction & deglycosidation

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

HOw is doxorubicin excreted?

A

Biliary - stool

<10% renal

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

What drugs does doxorubicin interact with?

A

Dexamethasone, 5FU and heparin will lead to precipitate if given together
Increased risk of haemorrhagic cysitis and cardiotoxicity if given with cyclophosphamide

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

What should you monitor with anthracyclines?

A

Cardiac function - ECHO/MUGA

Liver function

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

What are the side effects/risks with anthracyclines?

A
STRONG VESICANT
cardiotoxic
Skin toxicity- sensitive to sunlight
Hyperpigmentation
Alopecia
N+V
Mucositis
Diarhhoea
Red/orange urine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

What are the mechanisms of resistance to anthracyclines?

A

ELevated p170 levels -> drug efflux
Decreased expression of topoisomerase II
Increased expression of glutathione

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

What is the function of topoisomerase?

A

Help to unwind and untangle DNA for repair/replication
T1 create SSB
T2 create DSB

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

What is the function of topoisomerase?

A

Nuclear enzymes that help to unwind and untangle (break, rewind and rejoin) DNA for repair/replication
T1 create SSB allow uncoiling supercoiled DNA
T2 catalyses both SSB & DSBs

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

What is calyx and how is it different to normal drug?

A
Liposomal doxorubicin
Liposomal encapsulation leads to longer half life and possible preferential accumulation in tumour
Increased half life 45-90hrs
Reduced N+V and alopecia
Reduced cardiotoxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What is the MOA of mitomycin

A

Acts as an alkylating agent.
Cross link DNA
Activated by NADH cytochrome p450 reducatase to ROS, seimquinone and hydroquininone species which target DNA.
PREFERENTIAL ACTION IS HYPOXIC TUMOUR CELLS

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

How is mitomycin given and does it cross the BBB?

A

IV, not absorbed oral

WIdely distributed but does NOT cross BBB

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

How is mitomycin metabolised and excreted?

A

Extensively by p450 enzymes in liver

Bile -> faeces

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

What are the side effects of mitomycin?

A
STRONG VESICANT
myelosuppression - DOSE LIMITING
N+V
Mucositis
Anorexia
Fatigue
HUS
Pneumonitis
Heparo0venous disease
Chemical cystitis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What are the resistance mechanisms to mitomycin?

A

Elevated p170 levels -> drug efflux
Increased activity of DNA repair enzymes
Increased expression of glutathione and glutathione detoxifying enzymes

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

Which drugs are tubulin binding agents?

A

Vinca alkaloids: vincristine, vinblastine, vinorelbine, eribulin
Taxanes: paclitaxel, docetaxel
VDA: combrestatin
Antibody drug conjugates: Trastuzumab- emtandine (T-DM1)

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

What are microtubules?

A
Components of cytoskeleton
Essential in all eukaryotic cells for:
- development
- maintenence of cell shape
- transport of vesicles, mitochondria
- cell signalling
- cell division
- MITOSIS
Made of alpha and beta-tubulin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What part of the cell cycle are tubulin binding agents specific to?

A

G2/M phase

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

How do microtubules work?

A

Treadmilling- allows motion. Net growth at one end, net shortening at other end. Flow from +ve to -ve, length unchanged.

Dynamic instability

    • end switches between phases of growth and shortening
  • relatively long periods of slow lengthening
  • then rapid periods of shortening
  • rate of DI increases in mitosis x 100. Formation and attachment of mitotic spindles to chromosomes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

How are microtubules regulated?

A

Microtubule assoc proteins (MAPs) - MAP4 is a tau protein
Epigenetic alerations
Variable expression of tubulin isotu[es
Tubulin mutations

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

What are the advantages and disadvantages of targeting microtubules?

A

Advantages:

  • tubulin essential for rapidly dividing cells
  • > 1 binding site thus can overcome cross resistance
  • anti-angiogenic and anti-vascular effects

Disadvantages:

  • not tumour selective and overexpression does not drive cancer
  • needs apoptotic mechanisms eg p53 to be active
  • susceptible to mechanisms of resistance eg MDR
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

What is the MOA of the vinca alkaloids?

A

High affinity for microtubule ends (low affinity for sides)

  • increase tubulin autoaffinity-> leads to spiral aggregates
  • Destabilising agent: causes depolymerisation, suppress treadmilling an dynamic instability, inhibits mitotic progression and causes cell death by apoptosis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

How are the vica alkaloids metabolised and excreted?

A

Cytochrome p450

excreted in bile -> stools

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

Do vinca alkaloids cross BBB?

A

No

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

What are the side effects of vincristine and vinorelbine?

A
VESICANT
CONSTIPATION- start laxatives
NEUROPATHY- dose limiting
Hair loss
Fatal IT
SIADH

Vinorelbine + myelotoxicity

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

What are the mechanisms of resistance to vinca alkaloids?

A

P170 drug efflux

Mutations in alpha and beta tubulin proteins with decreased affinity for drug

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

How do taxanes work?

A

Microtubule stabilising agents, active M phase
Bind beta subunit of tubulin interiorly
- induce stabilisation
- increase polymerisation
- suppression of microtubule dynamics
- cell cycle arrest in G2/M and ultimately apoptosis
- sensitivity/resistance to taxanes is dependent on beta-tubulin isoforms

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

What is the metabolism, VOD for paclitaxel? Does it cross the BBB.

A

Wide VOD, distributes to 3rd spaces
90-95% protein bound
Metabolism p450 enzymes liver -> excretion faeces
Does NOT cross BBB

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

What are the side effects of paclitaxel and docetaxel?

A
Hypersensitivity
Myelotoxicity
Neurotoxicity
Myalgia
Oedema
Alopexia
Moderate nausea + vomting
Arrythmia
Lower back muscles can contract - occasionally muscle rupture-> high CK

Docetaxel
++ myelotoxicity
– neuro toxicity

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

What is difference between weekly and 3 weekly paclitaxel?

A

Weekly ? antiangiogenic effect- watch for MI

Weekly - more anaemia + neuropathy, less neutropenia

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

What is the VD and excretion of docetaxel?

A

VD 1-2l
Hepatic excretion
95% protein bound

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

What are the side effects of cabazitaxel?

A

Hypersensitivity
Neutropenia- dose limiting
Less neuropathy than other taxanes

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

How is cabazitaxel excreted and would you dose reduce in renal/liver impairment?

A

Metabolism by p450 and Hepatic excretion
Large VD
AST/ALT >1.5 C/I
Use with caution in renal impairment

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

What are the mechanisms of resistance to taxanes?

A

Alterations in the tubulin binding affinity

P-glycoprotein efflux pump (EXCEPT cabazitaxel which is a poor substrate for pump so may be useful in MDR disease)

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

What is the MOA of eribulin?

A

Halichondrin B analogue (non competitive inhibitor of vinca)
Suppress microtubule growth
Inhibits polymerisation inducing cell cycle arrest and apoptosis
Unique tubulin interaction as inhibits MT growth without shortening by sequestering tubulin in non-productive aggregates.
- binds to caps
- included aggregates that compete with soluble tubulin

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

How is eribulin distributed and eliminated?

A

Extensively distributed

ELimination hepatobiliary- dose reduce in liver impairment

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

What does eribulin interact with?

A

Drugs that prolong QTc so monitor ECG

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

What are the mechanisms of resistance to eribulin?

A

Appears less susceptible to p-glycoprotein pump

Maintains activity in various taxane resistant tumours

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

Name some drugs that inhibit topoisomerase I and II?

A

I: irinotecan
II: etoposide, anthracyclines

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

How does topoisomerase I work?

A

Main function is to maintain 3D conformity of DNA removing torsional stresses.

  1. Causes a transient enzyme bridge with DNA SSB (topo-1 cleavable complex, TOP1ccs) through which the strand can pass.
  2. Topo-1 reseals the cleaved strand

TOP1ccs is stabilised by the binding of camptothecins. Once stabilised the complex leaded to arrest of DNA replication fork and formation of SSBs.

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

How does irinotecan work?

A

Semisynthetic derivative of camptothecin
Converted to active metabolite SN-38 by carboxylesterases in liver.
SN-38 binds to and stabilised by topoisomerase I -DNA complex and prevents re-ligation of DNA after it has been cleaved -> causes DSB
Colorectal tumours have high levels of topoisomerase I

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

Is irinotecan cell cycle specific

A

Non specific

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

What can cause worse toxicity in a patient on irinotecan?

A

UGT1A1 def
SN-38 metabolised by glucocuronidation (UGT1A1) to SN-38 glucuronide
Reduced metabolism
significantly higher risk of diarrhoea and neutropenia x 4

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

How is irinotecan excreted? should you dose reduce in hepatic dysfunction?

A

64% faecal, 28% urine

Reduce dose in gilberts or any elevation of bilirubin

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

What are the side effects caused by irinotecan?

A

Cholinergic syndrome

  • N+V, salivation, facial flushing, diarrhoea (early onset), abdo cramps,
  • managed atropine

Late onset diarrhoea - 7-10 days

  • delayed mucosal cytotoxicity
  • cholera like diarrhoea
  • persistnet explosive watery stools
  • early loperamide

Neutropenia
Hepatotoxicity
anorexia

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

What are the mechanisms of resistance to irinotecan?

A

Decreased expression or mutations of topoisomerase I
P170 drug efflux
Decreased carboxylesterase enzyme so decreased activity of SN-38

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

How does topoisomerase II inhibitors work?

A

Topo II: Mediates ATP- dependent induction of nicks in both strands of DNA duplex allowing relaxation of superhelical DNA during DNA replication and transcription
Exists in alpha and beta isoforms.
Alpha isoforms
- interfere with enzyme function by stabilising a reaction intermediate in which DNA strands are cut and covalently linked to protein tyrosine residues forming a cleavable complex
- high levels of TOP2-DNA covalent complexes
- Stabilisation of this complex blocks transcription and replication

Non-intercalating- directly bind to enzyme: etoposide
Intercalating:doxorubicin, mitoxantrone

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

Is etoposide cell cycle specific?

A

Yes Late S and G2 phase

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

How is etoposide given and is it protein bound?

A

IV
Oral availability 50% of IV dose
90-95% albumin bound so low albumin might cause more toxicity
Enters CSF poorly

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

How is etoposide excreted?

A

44% faeces
- bili 26-51, AST 60-180 - give 50% dose
54% urine
- CrCl 15-50ml/min : 75% dose

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

What are the side effects of etoposide?

A

Myelosuppression- mainly leukopenia - 5-15days
N+V, mucositis, diarrhoea
Alopecia
Hypersensitivity
2nd cancer risk - mixed lineage leukaemia after 2-3 yrs

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

What are the mechanisms of resistance for etoposide?

A

P170 efflux- cross resistance to vinca alkaloids, anthracycline, taxanes
Decreased expression of topoisomerase II, decreased binding affinity for it
ENhanced DNA repair

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

What is the MOA of cisplatin?

A

Intracellular
Aquation- Cl- ion is displaced for H2O
H2O ligand is displaced so platinum ion can bind to bases.
PREFERENCE FOR GUANINE- N7 position
Crosslinks to another guanine by displacment of another Cl- ion.
Intra-strand links - poorly repaired
Inter-strand links- repaired more efficiently

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

What are the side effects of cisplatin?

A

High doses it is a vesicant
Highly emetogenic - use NK receptor blocker
Nephrotoxicity- 28-38%
Neurotoxicity
Ototoxicity
Myelosuppression
Electrolyte disturbance - hypo ca, K, Mg, Na
Arterial and venous thromboembolism (all platiniums, reduced with carbo). Small increased risk of MI/CVA

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

Describe the issues with nephrotoxicity with cisplatin?

A

Renal impairment can be progressive
Thrombotic microangiopathy- occurs when given with bleomycin
Hypomagnaesaemia- due to urinary magnesium wasting. Occurs in 50% cases.
Anaemia- renal tubular injury results in epo deficiency
Fanconi like syndrome- increased urinary excretion of glucose, AA. Lactate and pyruvate in urine.
Increase pro inflammatory cytokines
Vasoconstriction in renal microvasculature

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

What does cisplatin interact with?

A
Nephrotoxics
cisplatin reduces phenytoin dose
reduces renal clearance of etoposide
methotrexate
ifosfamide
bleomycin
should be given AFTER paclitaxel - prevents delayed excretion and toxicity (carbo as well)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

How is carboplatin different in its MOA from cisplatin?

A

Less potent cisplatin
Binds to guanine and adenine preferentially
aquation occurs at a slower rate 4:1 compaired to cisplatin.
Clearance proportional to GFR
2-6hrs in pts with normal GFR, up to 18hrs with abnormal GFR

144
Q

What are the difference in side effects for carbo to cisplatin?

A
Less nephrotoxic
Less emetogenic
Peripheral neuropathy uncommon
Trade off is more myelosuppressive
Reactions

Adminster AFTER PACLITAXEL

145
Q

Does haemodialysis clear carboplatin?

A

HD clears carbo at rate of 25% renal

PD does not

146
Q

What is the calculation for AUC?

A

Area under the curve on a plot of drug concentration in plasma against time
AUC measures total drug exposure over time- dependent on dose & rate of elimination

Carbo dose (mg)= target AUC (mg/ml x min) x (GFR +25)

147
Q

What are the mechanisms of resistance to carbo/cisplatin?

A

Reduced accumulation due to alterations in cellular transport
Increased inactivation by thiol containing proteins eg GLUTATHIONE (main mechanism)
Enhanced DNA repair enzyme activity
Def in mismatch repair enzymes

148
Q

What is oxaliplatin and how does it work?

A
Adds 2 bidentate groups- 1,2-diaminocyclohexane
Forms bulkier adducts
More effective at blocking replication
Forms inter- and intra strand crosslinks
Undergoes aquation like cis/carboplatin
149
Q

How is oxaliplatin excreted

A

> 50% renal - use with caution in CrCl <20

150
Q

What is the VD of oxaliplatin?

A

40% sequestered in RBCs
Wide VD
Extensively protien bound.

151
Q

What are the side effects of oxaliplatin?

A

Ototoxicity- rare compared to cisplatin
Nephrotoxicity - less than cisplatin
Acute neurosensory complex- during or after 1st inufsions.
- >85% pts. Discomfort swallowing cold items and throat discomfort, sensitivity touching cold items, paraesthesia & dyesthesias of hands, feet, perioral region.
Cumulative sensory neuropathies
- Dose dependent sensory, distal axonal neuropathy without motor involvement. Forms fewer adducts than cisplatin so less neurotoxic.

Hepatotoxicity - sinusoidal injury-portal htn, ascites
Rare cases of bronchiolitis obliterans pneumonia

152
Q

Why do you get acute neurosensory complex with oxaliplatin and how can you reduce acute neurotoxicity?

A

Chelation of Ca by metabolite leading to hyperexcitability of peripheral nerves
Magnesium and calcium infusions before and after can help

153
Q

What are the mechanisms of resistance to oxaliplatin?

A

Increased glutathione
Increase DNA repair
Non-cross resistant to cis/carboplatin in tumour cells that are deficient in MMR enzymes (MMR enzymes not required for oxaliplatin adducts due to bulkier size)

154
Q

Name the egfr inhibitors and which generation they are from?

A

1st gen: erlotinib, gefitinib
2nd gen: afatinib
3rd gen: osimertinib

155
Q

How do erlotinib and gefitinib work?

A

Reversible EGFR inhibition, bind to WT and mutant receptor

156
Q

What is the difference between the EGFR TKIs and cetuximab?

A

EGFR TKIs are small molecules that work on the the intracellular portion of EGFR receptor
Cetuximab: chimeric human-mouse antibody - blocks extracellular receptor

157
Q

How do afatinib work?

A

Irreversible eGFR inhibitors, bind to EGFR (incl WT) and HER2 & HER4

158
Q

How does osimertinib work?

A

Superior to any other eGFR inhibitors. Irreversible inhibition of mutated EGFR including T790M. Does not bind to WT EGFR so less toxicity.

159
Q

How are EGFR inhibitors metabolised and excreted?

A

Metabolism: liver CYP3A4 enzymes
Elimination: hepatic -> faeces

160
Q

What are the side effects of EGFR inhibitors and how do you manage them?

A

Rash (75%)

  • prevention: emollients, sun protection
  • G1/2: topical hydrocortisone/1% clindamycin, oral abx
  • G3/4: systemic abx, steroids, interrupt/reduce

Diarrhoea (50%)

  • G1/2 loperamide, codeine
  • G3/4 interrupt, stool sample, fluids, dose reduce

ILD

  • risk 1-8% with EGFR, 10% with mTOR
  • can be fatal
  • usually within 1 month of starting
  • stop, steroids, oxygen

Keratitis/conjunctivitis

Hepatitis
GI perforation
Hair and nail changes

161
Q

What do EGFR inhibitors interact with?

A
Warfarin
Cigarettes
PPI
Antacids
p450 inhibitors/inducers
162
Q

What are the mechanisms of resistance to EGFR inhibitors?

A

Mutations in ErbB tyrosine kinases leading to decreased binding affinity to afatinib
Presence of KRAS mutations
Presence of BRAF mutations
Activation/induction of alternative cellular signalling pathways such as PI3K/AKT, IGR-1R, c-MMET
Increased expression of mTORC1 signalling pathway

163
Q

Name an ALK inhibitor

A

Crizotinib

Alectinib - better

164
Q

What is the mechanisms of action of crizotinib?

A

Inhibits multiple TK receptors- ALK and c-MET
Metabolism in liver by CYP3A4/A5 enzymes
Excretion in faeces

165
Q

What are the side effects of crizotinib?

A

Common: visual disturbances, fatigue, GI toxicity, oedema

Rare but serious: hepatitis, pneumonitis, QT prolongation, cytopenias

166
Q

What do you monitor for patients on crizotinib?

A

Liver enzymes monthly

QT initially on ECG

167
Q

What drugs should be avoided with crizotinib?

A

PPIs

P450 enzyme inducers/inhibitors

168
Q

What drug is licenced in lung cancer patients with a ROS1 mutation?

A

Crizotinib

169
Q

What is bevacizumab?

A

zumab = humanised mouse antibody
against VEGF-A
Works extracellularly

170
Q

What are the side effects of bevacizumab?

A

HTN, MI, stroke, proteinuria/nephrotic syndrome
Poor wound healing (need 4-6wks off prior to op)
GI perforation (do not use if bowel involvement)
Posterior leukoencephalopathy syndrome

171
Q

What are the mechanisms of resistance to bevacizumab??

A

Increased expression of pro-angiogenic factor ligands, PIGF, bFGF, hepatocyte growth factor (HGF)
Recruitment of bone marrow derived cells which circumvents the requirment of VEGF signalling.
Increased pericyte coverage of tumour vasculature which supports its integrity and reduces the need for VEGF mediated survival signalling.
Activation and enhancement of invasion and mets to provide access to normal tissue vasculature without obligate neovascularisation.

172
Q

What are the VEGF TKIs used for

A

RCC
1st line: sunitinib, pazopanib
2nd line: cabozantinib (sorafenib)

GIST - sunitinib
PNET- sunitinib

173
Q

How are all the VEGF TKIs metabolised and excreted?

A

Liver and excreted faeces.
Metabolised by CYP3A4 p450 enzymes
Affected by p450 drugs

174
Q

What is the mechanism of action of sunitinib?

A

Multi targeted TKI

Inhibits phosphorylation of VEGFR1-3, PDGF beta, KIT, FLT-3

175
Q

Should you dose reduce in liver/renal impairment with sunitinib?

A

Renal- not needed, can take after dialysis

Liver- mild-mod not needed, severe not evaluated

176
Q

How should you monitor patients on sunitinib?

A

BP
Evidence of CCF
TFTs

177
Q

What are the side effects of sunitinib?

A
HTN (in nearly 30%)
Hand-foot sundrome
Yellow discoluration of skin
Bleeding/epistaxis
LVF
GI symptoms
myelosuppression
Adrenal suppression/hypothyroidism
178
Q

What is the dosing schedule for sunitinib?

A

4 weeks on, 2 weeks off

One dose/schedule does not lead to same oral bioavailability in all

179
Q

What is pazopanib?

A

Multi-targeted TKI

Inhibits phosphorylation of VEFR1-3, PDGF alpha & beta, KIT, bFGF

180
Q

Can you take sunitinib with food?

A

Yes, unaffected by food

181
Q

Can you take pazopanib with food?

A

Exposure increased if tablet crushed or if taken with food.

Take 1hr before or 2hrs after food

182
Q

What do you need to monitor in patients on pazopanib?

A
LFTS every few weeks esp when starting
QTc
HTN
Proteinuria on urine dip
TFTs
183
Q

What are the side effects of pazopanib?

A
Hepatic failure- SEVERE AND FATAL
HTN in 50%- related to plasma concentration
GI effects with elevated lipase and risk of perforation
MI/Angina/stoke
Hypothyroidism
Proteinuria
QTc prolongation
Myelosuppression
184
Q

Can you give pazopanib in liver/renal failure.

A

Hepatic impairment- can be caused by pazopanib, not usually sunitinib
- Mild: ALT>ULN, bili 1-1.5x = normal dose
Mod: bili 1.5-3x : max dose 200mg OD
Severe: bili >3x: CI
Renal impairment: dose adjustment unnecesary

185
Q

What is sorafenib?

A

Multi-targeted TKI
Inhibits phosphorylation of: b & c-RAF, KIT, FLT-3, RET, VEGFR2, PDGFbeta, KIT, VEGFR1-3
Used in RCC and HCC

186
Q

What are the side effects of sorafenib?

A
HTN
Hand-foot skin reactions
Wound healing issues
Diarrhoea
Hypophosphataemia
MONITOR BP
187
Q

Can you take sorafenib with food?

A

Absorption reduced by high fat food so take without food.

188
Q

Do you dose reduce sorafenib in liver and renal impairment?

A

Renal: no
Liver: mild-mod not needed

189
Q

What is cabozantinib?

A

Multi targeted TKI
Inhibits phosphorylation of VEGFR1-2, also MET and AXL (implicated in resistance to VEGF other TKIs)

Used 1st or 2nd line RCC

190
Q

Can you take cabozantinib with food?

A

Take one hour before or 2hrs after

191
Q

How do you monitor a patient on cabozantinib?

A

BP, urine protein regularly

Monitor for GI perforation, osteonecrosis

192
Q

What are the side effects of cabozantinib?

A
Hand-foot syndrome
diarrhoea, N+V, mucositis, 
Proteinuria, HTN
Bleeding
Hepatotoxicity
Reversible PRES syndrome
Osteonecrosis of jaw
193
Q

Can you give cabozantibib in renal/liver impairment

A

Renal impairment: mild/mod use with caution, severe not recommended
Liver impairment: mild/mod use 40mg OD< severe not recommended.
Start at 60mg OD normally

194
Q

What is cetuximab?

A

Monoclonal recombinant chimeric IgG antibody against EGFR
Binds with 10x greater affinity to EGFR than normal ligands blocking it.
Used in H+N, colorectal (ONLY WHEN KRAS WT)

195
Q

What should you monitor when a patient is on cetuximab?

A

Magnesium

196
Q

What are the side effects of cetuximab?

A
90% infusion related reaction
Skin reactions - shows drug is working
ILD
Paronychia
Hypomagnaesaemia
197
Q

What is denosumab and how does it work?

A

Fully human monoclonal antibody (igG) to RANKL
Prevents RANKL binding to RANK receptor on osteoclast surface reducing osteoclast survival and function -> less bone resorption

198
Q

What are the side effects of denosumab?

A
Hypocalcaemia
Bone pain
Abdo pain
constipation
Cataracts
osteonecrosis of jaw
Atypical femoral fractures.
199
Q

What is imatinib?

A

Phenylaminopyrimidine methanesulfonate compound that occupies the ATP binding site of the BCR/ABL protein and very limited number of other TKIs (PDGFR, c-KIT)
Results in inhibition of substrate phosphorylation
Induces apoptosis in BCR-ABL and Ph1 cells

200
Q

What is imatinib used for?

A

CML
ALL with Ph +ve
GIST expressing c-KIT

201
Q

What is the metabolism, excretion and interactions with imatinib?

A

100% oral bioavailability, extensively bound to plasma protiens
Metabolism: CYP4A enzymes
Excretion: faeces
Interactions: p450 enzyme inducers/inhibitors

202
Q

Can you take imatinib with food?

A

Yes should be taken with food

203
Q

What do you monitor if a patient is on imatinib?

A

ECHO/MUGA
mood for dpression
weight due to fluid retention

204
Q

What are the side effects of imatinib?

A
N+V (irritates stomach lining)
Fluid retention
CCF- rare but serioud
DIarrhoea
Myelosuppression
Skin toxicity
depression
205
Q

What are the mechanisms of resistance to imatinib?

A

Increased expresison of Bcr-abl tyrosine kinase
Mutations in Bcr/abl TK resulting in altered binding affinity of drug
Increased expression of P170 glycoprotein -> drug efflux
Increased expression of c-KIT

206
Q

What is nintedanib?

A

Small molecule TKI to PDGF, FGFR, VEGFR

Used in NSCLC with docetaxel initially

207
Q

How does nintedanib work?

A

Low oral bioavailability of 5% due to p-glycoprotein transporting drug back out of lumen and high 1st pass metabolism in liver
Inactivated by esterases to free carboxylic acid which is then glucuronidated and excreted in bile and faeces

208
Q

Is nintedanib effected by p450 enzymes?

A

NOT effected

209
Q

What are the side effects of nintedanib?

A

Deranged liver enzymes
GI disturbance
Strokes/MI
Poor wound healing

210
Q

What is olaparib?

A

PARP-i

Small molecule

211
Q

How does olaparib work?

A

Inhibits PARP which is key in base excision repair,

Synthetic lethality with BRCA mutations

212
Q

How is olaparib metabolised and what does it interact with?

A

Metabolism CYP3A
Interactions with those enzymes
Excretion urine and faeces

213
Q

What are the side effects of olaparib?

A
Pancytopenia
N+V
Arthralgia
Flu like symptoms
Diarrhoea
dyspepsia
214
Q

What is palbociclib

A

oral, reversible, selective, small-molecule inhibitor of CDK4 and CDK6

215
Q

Should you take palbociclib with food?

A

Yes

216
Q

What is the metabolism and elimination of palbociclib

A

Bioavailability 46%, 85% protein bound
Hepatic metabolism via CYP3A and SULTA1
ELimination faeces
Interacts with CYP3A drugs

217
Q

What the side effects of palbociclib?

A
Neutropenia -common
fatigue
nausea
Diarrhoea
rash
infections
thrombocytopenia
Reduce dose in severe hepatic impairment and monitor in renal impairment
218
Q

What is pertuzumab and how does it work?

A

Monoclonal recombinant humanised IgG1 antibody directed against extracellular dimerisation domain of HER2 receptor- subdomain II
Binding of pertuzumab leads to HER2 inhibition of heterodimerisation of HER2 with family members including EGFR, HER3, HER4.
Leads to inhibition of MAPK, PI3K

219
Q

How do you monitor patients on pertuzumab and what are the common side effects?

A

ECHO

Infusion reactions, fatigue, CCF

220
Q

What is trastuzumab and how does it work?

A

Recombinant monoclonal antibody directed against extracellular domain of HER2 receptor- subdomain IV

221
Q

What is transtuzumab-emtansine and how does it work?

A

HER2 targeted antibody drug conjugate made up of trastuzumab and a small molecule microtubule inhibitor DM1
Upon binding of HER2 receptor- kadcyla undergoes receptor mediated internalisation and lysosomal degradation leading to intracellular release of DM1 molecule.
Binding of DM1 to tubulin leads to disruption of microtubule ntwork and cell cycle arrest/apoptosis.
DM1 metabolised by CYP3A4/5

222
Q

What are the side effects of trastuzumab-emtansine?

A
Severe hepatotoxicity
CCF
Pulmonary syndromes
Neurotoxicity and peripheral neuropathy
Fatigue
223
Q

What is rituximab and how does it work?

A

Chimeric anti-CD20 antibody
Consists of a human IgG1-k constant regions and variable regions from murine monoclonal antibody
Targets CD20 antigen, a cell surface phosphoprotein expressed during early pre-B cell development until mature B stage but lost in plasma stage.
Binding results in inhibition of CD20 mediated signalling that leads to inhibition of cell activation and cell cycle progression
Mediates complement dependent lysis in presence of human complement and antibody dependent cellular cytotoxicity with human effector cells.

224
Q

What are the side effects of rituximab?

A

Infusion reactions common
C/I if hypersensitivity to murine products
Tumour lysis syndrome
Arrythmias and cardiotoxicity
N+V
Skin reaction- including Stephens johnson

225
Q

What is trametinib and how does it work?

A

Reversible inhibitor and mitogen-activated extracellular signal related kinase I (MEK1) and kinase 2 (MEK2)
Results in inhibition of downstrea, regulators of extracellular signal related kinase (ERK) pathway leading to inhibition of cellular proliferation.

226
Q

What is the availability and metabolism of trametinib?

A

Oral availability 70%
Food with high fat content reduces cmax, tmax, AUC
Extensive binding to plasma proteins
metabolism: mainly via deacetylation (by hydrolytic enzymes eg carboxylesterases) or mono-oxidation in combination with glucorinidation.

227
Q

How do you monitor patients on trametinib?

A

Cardiomyopathy - ECHO

Eye exams- retinal detachment/retinal vein occlusion

228
Q

What are the side effects of trametinib?

A
Cardiomyopathy - 10% develop median time of 63 days
Visual disturbances
Rashes 
Pul toxicity
GI side effects
HTN
Lymphoedema
229
Q

What is dabrafenib and how does it work?

A

Inhibits mutant forms of BRAF serine-threnonine kinase BRAF-V600E which results in constutive activation of microtubule-assoc protein kinase (MAPK) pathway.
Inhibits wildtype BRAF and CRAF kinases aswell

230
Q

Describe the bindong, metabolism and elimination of dabrafenib?

A
High oral bioavailability 95%
Avoid with food with high fat content
Extensive plasma protein binding
Metabolism: CYP2C8, CYP3A4  in liver to hydroxy-dabrafenib metabolite -> carboxy metabolite
Elimination: faeces
231
Q

How should you monitor somebody on dabrafenib?

A

Skin exams for SCC

Glucose

232
Q

What are the side effects of dabrafenib?

A
SCC/keratoacanthomas
Skin reactions
fever
hyperglycaemia
Arthralgia
OPthalmologic - uveitis, iritis
233
Q

What are the mechanisms of resistance to BRAF/MEK

A

Increased expression of MAPK

Activation of an alternative cellular signalling pathways eg FDF-R, PI3K

234
Q

What are bisphosphonates and how do they work?

A

Inorganic pyrophosphate analogues
Attach to hydroxypatite binding sites on boy surfaces. When osteoclasts begin to resorb bone, the bisphosphonate realeased impairs the ability of osteoclast to form a ruffled border to adher to the bony surface so cannot continue to resorb bone.
Also reduce osteoclast progenitor development and recruitment

235
Q

What is the excretion of bisphosphonates?

A

20-80% taken up by bone the rest is excreted renally.

Half life in circulation short at 2hrs but once taken up by bone, half life is years

236
Q

What is ipilimumab?

A

A fully monoclonal antibody against CTLA4 (T lymphocyte assoc antigen 4) expressed on surface of CD4 and CD8 lymphocytes.
This results in the inhibition of the interaction between CTLA4 and its target ligand CD80/CD86

237
Q

What is pembro/nivolumab?

A

Anti-PD1 fully human IgG4 antibodies
PD1 (on T cell) receptor inhibits T-cells activity by interactions with its ligands PDL-1 on tumour cells, APCs, and PDL-2 on activated monocytes and dendritic cells.
This usually results in an immunosuppressive tumour environment so helps to interrupt this by blocking it.

238
Q

What should you monitor for people on immunotherapy?

A

LFTs
TFTs
Cortisol

239
Q

Describe the hypothalamic/pit pathway that produces testosterone

A

Hypothalamus produces GnRH -> anterior pit produces LH and FSH
FSH -> sertoli cells -> spermatogenesis, oestrogens, androgen binding protein
LH -> leydig cells -> produce androgens

240
Q

How does testosterone change the transcription of particular genes/DNA?

A

Testosterone bound to SHBG extracellularly
Enters cell and coverted to DHG by 5 α-reductase
Binds to androgen receptor
AR undergoes dimerisation and phosporylation
Enters nucleus and binds to androgen response element of DNA - changes transcription of genes
Leads to increased growth, PSA and survival

241
Q

Name some LHRH analogues and describe how they work?

A

Goserelin, Leuprolin, Triptorelin
Buserelin (can give intranasally or by SC injection)

Stimulate LH receptors of pituitary
Initial testosterone flare
Then leads to LH receptor downregulation

242
Q

How quickly do you reach castrate levels of testosterone with LHRH analogues? What else might you need to give

A

May get testosterone flare in 1st 2-3 weeks - co treatment with an anti-androgen (eg bicalutamide/casodex) for 1 week before and 2-3 weeks after starting is advised

Reach castrate levels testosterone in 2-4 weeks

243
Q

Name a LHRH antagonist and describe its mechanism of action?

A

Degaralix
Binds to LHRH receptors in pituitary - pure antagonist with reversible/complete binding.
No initial agonist action (no LG release) so no flare.
Rapid and profound testosterone suppression
? better cardiaac risks than LHRH agonists.
Given SC

244
Q

How is zoladex/goserelin excreted?

A

Usually an implant- slowly released drug
Half life of 2-4hrs in blood. Poorly protein bound
Renally excreted
No dose change in liver/renal dysfunction

245
Q

How is degarelix metabolised and excreted?

A

90% plasma protien bound

Metabolism via hydrolysis and excreted in peptide fragments in faeces

246
Q

What are the side effects of androgen suppression?

A
Hot flushes
Fatigue
Impotence/loss of libido
Loss of bone strength and muscle -> DEXA scan
Mood changes
Metabolic syndrome -> diabetes, CVD, MI, sudden death
- insulin resistance
- arterial stiffness
- increased BMI
247
Q

How does prostate cancer become castrate resistant?

A

AR overexpression
AR mutation
Signalling crosstalk- alternative AR activation
Androgen indepedent AR activation

248
Q

Name the anti-androgens and describe how they work?

A

20-30% response- usually used with LHRH agonists
Bind to and block AR

1st gen

  • Bicalutamide - metabolised by p450
  • Flutamide
  • Cyproterone acetate - potent progestogen, moderately potent anti-androgen, weak glucocorticoid

2nd gen
- enzalutamide

Other drugs that inihibit adrenal androgen production:

  • corticosteroids
  • ketoconazole
  • abiraterone acetate (x10 more potent than ketoconazole)
  • oestrogens- diethylstilbestrol
249
Q

What is enzalutamide and how does it work?

A
Novel AR antagonist
Three mechanisms of actioN:
- blocks testosterone binding to AR
- impedes movement of AR to nucleus
- inhibits DNA binding
Causes cell death in cancers resistant to bicalutamide
250
Q

How is enzalutamide metabolised?

A

Extensive binding to plasma proteins
Metabolism : liver CYP2C8, CYP3A4
Interacts with those drugs - clopidogrel

Not tested in CrCl <30 or severe hepatic dysfunction

251
Q

What are the side effects of enzalutamide?

A
Fatigue
MSK- back pain
Coronary syndromes
Diarrhoea
Hot flushes
Oedema
Seizures <1%
252
Q

How does abiraterone work?

A

Derivative of steroidal progesterone
Inhibits androgen production from 3 sources- adrenal gland, testes and prostate tumour cells.

Blocks CYP17 : 17α-hydroxylase and 20-lyase

This prevents the conversion of cholesterol to cortisol (vis 17α- hydroxyprogesterone) or to testosterone (via DHEA)
Instead cortisol is converted to aldosterone (via progesterone)
Build up of mineralocorticoids.

253
Q

How is abiraterone metabolised and excreted?

A

Rapidly hydrolysed to its active metabolite via esterases.
CYP3A4 and SULT2A1 further metabolizes abiraterone into two inactive metabolites called abiraterone sulfate and N-oxide abiraterone sulfate.
90% excreted in faeces

INTERACTS with CYP3A4 drugs

254
Q

What are the side effects of abiraterone?

A
Fatigue
Oedema
Arthralgia
HTN
Hypokalaemia
Coronary syndromes
255
Q

What is the mechanisms of resistance to abiraterone?

A

Upregulation of CYP17
Induction of AR and AR splice variants tha result in a ligand-independent AR transactivation
Expression of truncated androgen receptors.

256
Q

What is the MOA of anastrazole and letrozole?

A

Non-steroidal aromatase inhibitors
INhibit conversion of adrenal androgens (androstenedione and testosterone) to oestrogens.
Serum oestradiol suppressed by 90% at 2 weeks and completely at 6 weeks.

257
Q

How is anastrazole metabolised and excreted

A

In liver by N-dealkylation, hydroxylation and glucoronidation by cytochrome p450 enzymes
90% excreted faeces
Avoid if CrCl <20

258
Q

How is letrozole metabolised and excreted?

A

In liver via CYP3A4 and CYP2A6.
Glucorinidation leads to inactive metabolites
75% renally excreted.
Manufacturer advises caution if CrCl <10

259
Q

What are the side effects of aromatase inhibitors?

A
N+V
Hot flushes
Dry skin
Arthralgia
Headache
Oedema
Osteoporosis - DEXA monitoring
260
Q

What is exemestane and how does it work?

A

Steroidal aromatase inhibitor

Binds to and permanently , irreversible inactivates aromatase.

261
Q

How is exemestane metabolised and excreted?

A

CYP3A4 enzymes

Excreted in faeces

262
Q

What is tamoxifen?

A

Non steroidal anti-oestrogen with weak oestrogen agonist effects.
Competes with oestrogen for binding to ERs (inside cells), when tamoxifen binds it leads to ER dimerisation and tamoxifen ER dimer is transported to nucleus where it binds to DNA sequence referred to as ER elements.
Inhibits transcriptional process and transduction pathways

Also inhibits TGF-B which inhibits TGF-a, IGF-1 - involved in proliferation

263
Q

Is tamoxifen cell cycle specific?

A

Yes blocks cell at mid G1 phase.

264
Q

How is tamoxifen metabolised and excreted?

A

Plasma protein bound
Metabolised by cytochrome p450 to main metabolite N-desmethyl tamoxifen- same biological activity as tamoxifen..
Excreted in faeces

265
Q

What does tamoxifen interact with?

A

Cytochrome p450 (CYP2D6, CYP3A, CYP2B6, and CYP2C1)
SSRIs/SNRIs
antipsychotics

266
Q

What are the side effects of tamoxifen?

A
Endometrial cancer
VTE
Menopausal symptoms
elevated triglycerides
Myelosuppression -rare
Pruitis
Initiation can cause a tumour flare- MSCC/bone mets worse
267
Q

What are the mechanisms of resistance to tamoxifen?

A

Decreased ER expression/mutations leading to decreased binding affinity
Over-expression of growth factor receptors- HER, EGFR< IGF-1, TGF-beta
ESRi mutation

268
Q

What is medroxyprogesterone acetate and how does it work?

A
Steroidal progestin (synthetic variant of progesterone)
Binds to progesterone receptors and blunts GnRH release
Anti-proliferative effect on endometrium
Used in endometrial/breast/renal cancer
269
Q

What are the side effects of medroxyprogesterone acetate?

A
Loss of BMD
Bleeding irregularities
Cancer risks
VTE
CCF
270
Q

What is octreotide?

A

Somatostatin analogue
Inhibits hormone secretions in carcinoid syndrome
Octreotide binds to somatostatin receptors highly expressed on surface of carcinoid tumours, where it inhibits the release of gastrin, VIP, secretin, motilin and serotonin.

271
Q

How is octreotide given, metabolised and what does it interact with?

A

SC injection, half life 100mins (longer than somatostatin)
Metabolised by p450 enzymes
Interacts: cyclosporin, insulin, oral hypoglycaemic agents, beta blockers, bromocriptine.

272
Q

What are the side effects of octreotide?

A
Biliary abnormalities
Nausea
Abdo pain
Constipation
Myalgia
Changes in glucose
273
Q

What chemotherapy drugs are never given intrathecally and which are?

A

NEVER VINCA ALKALOIDS

Methotrexate, cytarabine

274
Q

What are the safety conditions when giving intrathecal chemo?

A

All drugs labelled with FOR INTRATHECAL USE ONLY - should be transported seperately to IV drugs

Only >ST3 can prescribe if appropriately trained
Given in designated area
Given in normal working hours
Given AFTER IV chemo
Purpose designated intrathecal chemo chart
Register of delegated personel who have been traned to prescribe, authorise, dispense and administer intrathecal chemo.

275
Q

What are pharmacokinetics?

A

The times course of a drug in the body (what a body does to the drug)

276
Q

What are pharmacodynamics?

A

the time course of the effect pf a drug (what the drug does to the body)

277
Q

What is the description of the time course of a drug in the body?

A
LADME
Liberation
Absorption
Distribution
Metabolism
Excretion
278
Q

What effects rate of absorption of a drug?

A

Oral- absorption depends on the lipid solubility of a drug

- non-ionised molecules are favoured as far more lipid soluble than ionised (H+) which have a shell of water molecules

279
Q

What effects the distribution of a drug?

A

Lipid soluble drugs are generally rapidly distributed and protein bound.

280
Q

What is the volume of distribution equal to?

A

VD: apparent volume into which the drug is distributed following IV injection (IV drugs have 100% bioavailability)

VD= dose/concentration at time

281
Q

If you give 100mg of drug A IV and measure the concentration at Time T as 1420mcg/L. What is the volume of distribution at time T?

A

100mg = 100,000mcg

100,000/1429 = 70l

VD = dose/concentration at time

282
Q
If the volume of distribution is :
1. 3l
2. 5l
3. 15l
4. 42l 
Where does it suggest the drug has been distributed to?
A
3l (4%) = plasma
5l (7%) = blood
15l (21%) = extracellular fluid
42l (60%) = total body water
>42l = tissue distribution

Women have more fat so total body water is lower than men.

283
Q

How do you calculate the concentration of a drug?

A

Concentration decreases exponentially
Plasma concentration = initial concentration x e^ - λt

λ = elimination rate constant = ln2/ t1/2

284
Q

How many half lives doe sit take to reach a steady state?

A

5

285
Q

What is clearance of a drug equal to?

A

Cl = λ x VD

286
Q

If λ = 0.02 min ^-1 , what does that mean?

A

λ is elimination rate constant

2% of drug is eliminated per minute

287
Q

How do you calculate the dosing rate of a drug?

A

Dosing rate = clearance x steady state plasma concentration

If giving orally the dosing rate = bioavailability x dose /dosing interval = plasma clearance x plasma concentration

288
Q

What different concentration effect relationships do you know?

A
Complicated to model
- linear
- quadratic
- logarithmic
- Emax and sigmoid Emax
Complicated by hysteresis
289
Q

What is hysteresis?

A

Peak effect doe not happen at peak concentration but after the peak concentration is dropping
can model this with effect compartment or indirect response model

290
Q

Which drugs are vesicants?

A

Anthracyclines - tend to be the worse
Mitomycin C
Vinca alkaloids
Taxanes

291
Q

Which drugs are irritants but NOT vesicants?

A
5-FU
Platinums
Topoisomerase inhibitors
Liposomal doxorubicin
Ifosfamide
292
Q

What drug can you use in anthracycline extravasations?

A

Dexrazoxane
Water soluble ring close analog of the iron chelator ethylenediaminetretraacetic acid (EDTA)- like form which is a strong iron chelator and has ability to displace iron for th anthracycline

293
Q

What are the steps for dealing with extravasation?

A
  1. Stop and disconnect infusion, leave needle in place
  2. Identify the extravasated agent
  3. Try to aspirate as much as possible and record the volume remove. Avoid manual pressure over the area and remove cannula.
  4. Mark with a pen or outline the area
  5. notify plastics

If anthracycline, antibiotics, alkylating agents

  • apply dry cold compresses
  • use specific antidotes - anthracyclines - topical DMSO, dexrazosane, for mitomycine topical DMSO

If vinca alkaloids, taxanes, platinium

  • apply warm compresses
  • adminster hyaluronidase- increases resorption

Elevation and analgesia.

294
Q

Where are opioid receptors found and name them?

A

Peripheral nerves
Spinal cord
Mutiple regions of brain

4 opioid receptors
Mu (70%) - u
Delta (24%)
Kappa (6%)
ORL-1 (opiod receptor like-1)
295
Q

How is morphine metabolised and what receptor does it exert its effect?

A

35% PO bioavailability
Half life 1.5hrs
Metabolised (glucorinidation) by enzyme UGT2B7 to major metabolites morphine-3-glucuronide (60- 80% and is inactive), morphine-6-glucouronide (10-15% and is active)
M6G acts of u- receptors

296
Q

What is diamorphine and how does it work?

A

Pro-drug of morphine
More soluble and more lipophilic
Half life 3 mins IV
M-6-G acts on u receptors.

297
Q

How is oxycodone metabolised and which receptors does it act on?

A

CYP2D6
CYP3A4

u & k opioid receptors

298
Q

What is fentanyl and what is its MOA?

A
Synthetic opioid
80x more potent than morphine
Transdermal preparation
- lipophilic low molecular weight
Increased absorption with pyrexia

Metabolism: CYP3A4 & CYP3A5 -> norfentanyl

Acts on u- opioid receptors

299
Q

What is buprenorphine?

A
Semi-synthetic opioid
25-30 x more potent then morphine
Partial agonist
- partial Mu receptor agonist
- Kappa receptor antagonist
- weak delta receptor agonist

Highly lipid soluble
Safe to use in renal impairment

300
Q

How is buprenorphine metabolised?

A

CYP3A4

301
Q

What is methadone and how does it work?

A

Synthetic opioid
Broad receptor profile:
- Mu receptor, delta receptor, NMDA, presynaptic blocker of serotonin reuptake.

302
Q

Describe the distribution and excretion of methadone

A

HIghly variable half life- 8-75hrs
Rapid distribution and slow elimination
Accumulates in fat stores, only 1% stays in plasma
Excreted unchanged by kidney or metabolised liver (CYP2B6 & CYP3A4) to inactive metabolites
Can use in renal failure

303
Q

How does alfentanil work

A

Synthetic derivative of fentayl
Rapid onset and short duration of action
1/10 PRN dose but rarely used as lasts around 30 mints.
Metabolism by CYP3A4 to inactive metabolites- excreted in urine

304
Q

How do NSAIDs work?

A

Inhibit COX-1 and COX-2 - produce prostaglandins and thromboxanes

305
Q

How do corticosteroids work?

A

Diffuse into cell and bind to glucocorticoid receptors (usually kept inactive by hsp90)
When binds to cortisol it moves to nucleus and stimulates synthesis of proteins.

306
Q

What are the effects of steroids?

A

Reduces inflammation and immunological responses
Increases liver glycogen deposition and increases gluconeogenesis
Increases bone catabolism - osteoporosis
Lifts mood/psychosis
Increases gastric acid
Suppresses adrenals
Mineralocorticoid effects - K+ excretion, Na reabsorption
HTN

307
Q

How do tricyclic antidepressants work?

A

Blocks the presynaptic re-uptake of serotonin and noradrenaline in the CNS, enhancing the actiosn of the descending inhibitory pathways.

308
Q

Name some TCAs. What are there side effects?

A

2 classes

  • tertiary amines eg amitripyline
  • secondary amines eg notrtiptyline

Side effects: anti-cholinergic- dry mouth, blurred vision, constipation, postural hypotension, heart block, arrythmias
Caution in cardiac diseas and hepatic disease

309
Q

How does gabapentin/pregabalin work?

A

Chemical analogue of GABA
Reduced calcium influx of hyper-excited neurons.
Caution in renal impairment

310
Q

How do benzodiazepines work?

A

GABA agonists- inhibit release of excitatory glutamate decreasing spasm in skeletal muscle

311
Q

How does baclofen work?

A

Derivative of GABA
Works by activating GABAb receptors
Excreted renally

312
Q

How does ketamine work?

A

NMDA receptor antagonist

  • calcium channel involved in central sensitisation
  • normally blocked by magnesium and inactive
  • prolonged excitation unblocks channel and causes increased pain, expansion of field of pain and opioid resistance.
  • Also acts on opioid receptors, monoaminergic receptor,s muscarinic receptors, voltage sensitive calcium ions.
    NOT GABA

Extensive 1st pass metabolism to norketamine via CP3A4
Max blood concentration greater aft PO than injection

313
Q

Describe the adjuvant analgesic step ladder for neuropathic pain?

A
  1. Corticosteroids
  2. TCA or anti-epileptics
  3. TCA & anti-epileptic
  4. NMDA receptor blocker
  5. Spinal analgesia.
314
Q

Which areas of the brain cause vomiting and what receptors do they have

A

Vestibular system - H1 receptors, M1 receptors (muscarinic)
CTZ in medulla: D2, NK, 5HT2 receptors
Vomiting centre in medulla: H1, NK, M1 receptors
GI tract: D2 receptors, 5HT3 receptors.

315
Q

How does metoclopramide work and what are the side effects?

A

D2 receptor antagnost
5HT4 receptor antagonist
In addition to prokinetic effect on gut it also acts on CTZ

Side effects: Acute dystonia, oculogyric crisis, extrapyramidal side effects, galactorrhoea, gynaecomastia

316
Q

How does domperidone work?

A

D2 receptor antagonist
Acts on receptors in CTZ and stomach
DOes not cross BBB

Side effects; Gynaecomastia, galactorrhoea, amenorrhoea

317
Q

How does haloperidol work?

A

D2 receptor antagonist in CTZ

Causes sedation and extra-pyramidal side effects

318
Q

How does cyclizine work>

A

H1 and ACh antagonists - acts on inner ear and vomtiing centre.

319
Q

What are the uses and side effects of cyclizine?

A

Use: vertigo and labyrinthine disorders, mechanical bowel obstruction and raised ICP
Side effects: drowsiness, caution in heart failure as anti=muscarinic effect can cause tachycardia

320
Q

How does levomepromazine work?

A

Antagonist at D2, H1, M1, 5HT2 & alpha1-adrenergic receptors
Used for refractory nausea
Side effects: drowsiness, postural hypotension, antimuscarinic effects.

321
Q

How does ondansetron work?

A

Blocks the amplifying effect of excess 5HT on vagal nerve fibres
5HT3 antagonist
Used specficially for highly emetogenic radiotherapy and chemotherapy (combined with NK often)

322
Q

What are the side effects of ondansetron?

A

Constipation and headaches.
Reduces analgesic efficacy of tramadol
dose reduce in hepatic impairment

323
Q

What is aprepitant and how does it work?

A

NK1 receptor antagonist

  • G protein coupled receptors located in peripheral and central nervous system
  • receptor has dominant ligand - substance P. SP is a neuropeptide which is found in high concentrations in the vomiting centre and whne activated results in vomiting reflex
  • aprepitant blocks substance P

Metabolised by CYP3A4

324
Q

What are the side effects of ondansetron?

A

Headache, hiccups, constipation, reduced appetite

325
Q

How does GCSF work?

A

Binds to cell surface receptors on haemopoetic cells in bone marrow and stimulates neutrophil progenitor cell proliferation and differentiation
Also speeds up neutrophil maturations leading to increased number being released.

326
Q

What are the possible side effects of GCSF?

A
Allergic reaction
splenic rupture
ARDS
Sickle cell criss
Glomerulonephritis
Alveolar haemorrhage
Capillary leak syndrome
low plts
Bone pain
Rash 
Diarrrhoea/constipation
Alopecia
DONT USE IN MDS and CML
327
Q

What is pegylated GCSF?

A

Addition of polyethelene glycol PEG moetiy to filgrastim
Long acting form - too large for renal clearance
Neutrophil mediated clearance- half life around 42hrs although dependent on neutrophil count.
Half life of GCSF 3.5hrs

328
Q

What are pharmacodynamic drug interactions?

A

Where the effects of one drug change be the presence of another substance at its site of action eg folinic acid and 5FU

329
Q

What are pharmacokinetic drug interactions?

A

Those which affect absorption, distribution, metabolism, elimination

330
Q

Why are drugs metabolised?

A

Protein bound drug is not filtered by renal glomerulus.
Free drug readily diffuses back from tubule into blood
If relied on renal excretion along than drugs would have a very long half life so most drugs are metabolised.

331
Q

What happens during drug metabolism

A

Process in which drugs converted to a more water soluble form that can be filtered by the kidneys

332
Q

What is a phase I reaction?

A

Oxidation, reduction or hydrolysis.
Oxidation is most common.
CYP450 enzymes account for 90% drug metabolism.
CYP3A4 accounting for 50%.

Other enzymes involved in metabolism are xanthine oxidase- metabolises mercaptopurine and azathioprine

333
Q

Name some CYP3A4 inducers?

A
Carbamazepine
Phenytoin
Modafinil
St Johns wort
Rifampicin
Dexamethasone
334
Q

Name some CYP3A inhibitors

A
Aprepitant
Antifungals eg ketoconazole
Grapefruit juice
Clarithromycin/erythromycin
Ciprofloxacin
Lapatinib
Imatinib
Verapamil
335
Q

What is a phase 2 reaction?

A

Coupling/conjugation with other substances. Usually to less active polar molecules that are regularly excreted by kidneys

  • glucuronidation
  • acetylation
  • glutathione
  • glycination
  • sulphation
  • mehtylation
336
Q

What effects elimination of drugs?

A

Tends to be excreted in bile or urine
Changes in pH, tubular active transport systems
Reduced renal blood flow - methotrexate with NSAIDS

337
Q

What is the max CrCl for carboplatin dosing?

A

125ml/min is cap

338
Q

What are the risk factors for tubular necrosis while on cisplatin?

A
33% get nephrotoxicity
CrCl reduced, avoid if <40
Dose
Rate of administration
Hydration status
Low albumin
Other nephrotoxics
339
Q

What dose is considered high dose methotrexate?

A

> 1g/m2

340
Q

Name some drugs which may cause permanent loss of fertility?

A
Cyclophosphamide
Cisplatin
Doxorubicin
Irinotecan
Ifosfamide
Gemcitabine
? etoposide
341
Q

How can chemotherapy cause women to become infertile?

A

Affect on follicular growth and maturation within the ovary, caused by cell-cycle specific chemotherapies, or can result from death of the resting oocytes within the ovaries, caused particularly by alkylating agents. Destruction of the
oocytes results in a decreased number of steroid producing cells, and thus a fall in the oestrogen levels, and a direct impact on the normal cycling of gonadotrophins. The effect on ovary function may be temporary, or may, if
oocyte loss has occurred, be permanent.
Chemotherapy can cause a mitotic arrest of the endometrial epithelium, which will limit fertility. This would normally be temporary.

342
Q

How can chemotherapy cause infertility in men?

A

Cause infertility or impotence
Affect sertoli cells- sperm made
Affect Leydig cells- reduce testosterone -> reduced sperm, impotence
If stem cell compartment of testes significantly affected it will not recover

Impotence - neurotoxicity - platinum based chemo

343
Q

What chemo drugs do you dose reduce in liver impairment?

A

Anthracyclines
Taxanes
Paclitaxel 3 weekly: if bilirubin < 1.25 x ULN and ALT < 10 x ULN, proceed with full dose.

Vinca alkaloids

344
Q

Which drugs develop resistance with p170 glycoprotein efflux pump?

A

anthracyclines, vinca alkaloids, etoposide and taxanes

345
Q

How does cisplatin cause nephrotoxicity?

A

Proximal tubular necrosis and apoptosis
Vasoconstriction
Pro-inflammation

346
Q

What does non-linear pharmacokinetics mean and why does this usually happen?

A

the clearance of a drug is not proportional to the concentration for all concentrations. This is usually due to saturation of metabolic pathways, which results in a decrease in (relative) clearance as the concentration of drug increases
Zero order kinetics (rather than 1st order)

347
Q

What are the clinical implications of non-linear pharmacokinetics?

A

clinical implications of this are that above a certain dose for drugs with non-linear pharmacokinetics the concentration and hence toxicity may increase rapidly and disproportionately with dose

348
Q

What is paclitaxel mixed with and why? What side effect does this cause?

A

Highly lipophilic
polyoxyethylated castor oil (Cremophor EL) in a 1 : 1 v/v mixture with dehydrated ethanol, a powerful solubilizer combo
Reactions
probably by inclusion in micelles, is the cause of the apparent nonlinear plasma pharmacokinetics of paclitax

349
Q

Name 4 consequences of combining paclitaxel with a pGP inhibitor?

A

Pharmacokinetics – increased half-life

Pharmacokinetics – decreased clearance

Pharmacodynamics – increased efficacy

Pharmacodynamics – increased toxicity

350
Q

Why are liposomal drugs removed from the body and what can be added to them to prevent this from happening?

A

Readily opsonised and caught by mononuclear phagocyte system + removed.
Add PEG - prevents binding of opsonins (stealth liposome), also increased uptake in tumour

351
Q

Define oral bioavailability

A

Extent and rate at which active drug enters circulation
Assessed using AUC - conc/time curve
Oral bioavailability = AUC drug adminstered orally/ AUC drug administered IV

352
Q

What is a therapeutic index?

A

toxic dose/ dose for therapeutic response
Usually TD50 / ED 50

ED Effective dose

353
Q

What is the classical MDR phenotype and what drugs are susceptible?

A

p-glycoprotein pump
resistant to the so-called naturally occurring anti-cancer drugs, such as anthracyclines, Vinca alkaloids and epipodophyllotoxins (etoposide)

354
Q

What is atypical MDR?

A

Atypical MDR is assoc with changes to topoisomerase IIa

Also non-Pgp MDR phenotype is caused by overexpression of the multidrug resistance-associated protein (MRP) gene -> effects same drugs a pGP

355
Q

MMR deficiency leads to resistance towards which drugs?

A

Capecitabine/5FU
Carboplatin/Cisplatin
Alkylating agents together with MGMT