Pharmacology 10: Cancer Chemotherapy Flashcards

1
Q

characteristics of cancer cells which define the magnitude of malignant threat?

A

loss of cell growth control- ordered control of cell division not balanced by apoptosis
de-differentiation and loss of specific function
blood supply- tumour cells release local angiogenesis factors to promote vessel growth
metastasis and invasiveness- loss of positional sense, disruption to E-cadherin?
tumour compartmentation

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

which 3 compartments to tumour cells belong to?

A
A= dividing cells receiving adequate nutrient/vascular supply
B= resting cells in G0, able to rejoin A if changes in cell signalling/local environ e.g. following surgery, more likely to be situated in middle of tumour.
C= cells no longer able to divide, contribute to overall tumour bulk, present no challenge.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

which compartment of tumour cells is most susceptible to chemotherapy?

A

A

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

how many cells does a tumour need to consist of to be clinically detectable or to reach size of a small grape?

A

10^9 cells

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

what is the difficulty of targeting compartment B tumour cells with chemotherapy?

A

proportion of chromosomal DNA in G0 open to attack is much more limited, making effective kill ratio much lower, and cells therefore available to re-enter compartment A, even following intense chemotherapy.

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

what is the general aim of all cytotoxic agents used in cancer chemotherapy?

A

drive a therapeutically higher rate of apoptotic death in cancer cells over that caused in normal cells

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

how do the anthracycline antibiotics work in cancer chemotherapy?

A

their discrete molecular ring structure enables them to intercalate between spaces between DNA base pairs, interfering with normal transcription and replication.
Antibiotic also binds to Topoisomerase II, forming a tripartitie DNA-anthracycline-Topoisomerase II complex, and topoisomerase II enables breaking, rotation and re-ligation of DNA strands in DNA replication and repair, resulting non-ligated free DNA strands act as trigger for apoptosis.
Anthracyclines can also generate free radicals by binding to Fe2+, which go on to damage DNA, which is detected by DNA damage sensing mechanisms, which trigger apoptosis.
Action non-cell cycle specific

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

in which phase of the cell cycle is bleomycin, a glycopeptide antibiotic, most effective?

A

G2

but also some effect in non-replicating G0 cells.

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

from which bacteria is the glycopeptide antibiotic bleomycin derived?

A

streptomyces fungi

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

how does bleomycin work to directly modify DNA structure?

A

binds with DNA and chelates with free Fe2+ ions
structure allows it to closely align itself within DNA by intercalation and by binding via its terminal NH2 group to DNA
reaction site when it chelates with Fe2+ then catalyses production of superoxide and OH free radical species- attack phosphodiester bonds in DNA, causing cutting of DNA strands= primary mechanism underlying cytotoxicity

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

what is the reactive group on alkylating agents?

A

electrophilic- attracts e- from nucleophilic target sites along length of DNA strands

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

when does the maximal effect of alkylating agents occur?

A

during S phase of cell cycle as DNA replicating and large sections of DNA strands exposed and unpaired
but affect cell function in all phases so cell-cycle nonspecific

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

examples of antimetabolites which interfere with precursors to purine and pyrimidine synthesis?

A

6-mercaptopurine (from azathioprine)
methotrexate
5-fluorouracil

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

which nucleoside has the most marked reduction in response to methotrexate?

A

thymidine*

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

enzyme inhibited by methotrexate?

A

dihydrofolate reductase, inhibiting production of tetrahydrofolate= methyl group carrier, co-factor

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

how does 5-fluorouracil act?

A

irreversibly inhibits thymidylate synthase

analogue of uracil= a pyrimidine base

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

where do antimetabolites act in the cell cycle?

A

specific to S phase

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

how do vinca alkaloids e.g. vincristine cause cell apoptosis?

A

inhibit microtubule formation by binding to beta-tubulin subunit, preventing microfilament formation that make up microtubule, so mitotic spindle doesn’t form and cells arrested in mitotic metaphase. chromosome cannot segregate and affected cell cannot proliferate.

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

how do taxanes e.g. paclitaxel and docetaxal, cause cell apoptosis?

A

promote and stabilise formation of tubulin polymer into microtubules, reversibly bind to beta-tubulin subunit,stopping microtubules from disassemblying so rendered non-functional, chromosome cannot be pulled apart, cell stuck in metaphase.

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

most common adminstration route for cancer chemotherapy agents?

A

IV- often their toxicity rules out oral delivery as would severely damage GI tract.
also, bioavailability often variable and ptnt likely suffer nausea and vomiting throughout tment.
IV allows fine delivery control by injected bolus, infusion bag or pump, and if emergency, infusion can immediately be stopped.

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

what route of delivery is used for tumours in CNS?

A

intrathecal and intraventricular

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

2 types of resistances of cancer cells to chemotherapy?

A
primary= resistance prior to drug exposure
acquired= after drug exposure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

how can acquired drug resistance come about?

A

multidrug resistance protein= expression may increase if cancer cells exposed to 1 or more chemotherapeutic drugs, functions to generically remove hydrophobic large xenobiotics.

may be downregulation of active carrier e.g. with methotrexate and cis-platin drugs, drug exposure decreases rate of active drug uptake

drug target enzymes may be upregulated to offset decrease in metabolite prod e.g. increase dihydrofolate reductase with methotrexate.

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

how can drug resistance be offset clinically?

A

utilise high dose, short term intermittent repeated therapy with drugs given in optimal combination.

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

common ADRs with cancer chemotherapeutics?

A

nausea
vomiting- acute phase 4 - 12 hours, delayed onset, 2 - 5 days later, chronic phase- may persist up to 14 days, action on central chemoreceptor trigger zone
diarrhoea
mucositis
alopecia
myelosuppression, impaired wound healing and skin toxicity

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

most frequent cause of death during chemotherapy?

A

haematological toxicity

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

why can acute renal failure occur with cancer chemotherapeutics?

A

rapid tumour lysis causes large increases in purines being released in to the circulation, increased purine met. generates urates which precipitate urate crystal formation in renal tubules, can then cause kidney failure and death.
=tumour lysis syndrome

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

why are anthracyclines especially cardiotoxic?

A

free radical generation

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

what high risk of about 10% is assoc with bleomycin?

A

pulmonary fibrosis

especially if patient also on O2 therapy

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

ADRs assoc with alkylating agents?

A

periperal, sensory and motor neuropathy

high frequency ototoxicity

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

ADRs assoc with mitiotic spindle inhibitors e.g. vincristine and taxanes?

A

neurotoxicity as ‘glove and stocking’ peripheral neuropathy often reported

32
Q

why are gaps between chemotherapy dosing required, and hence why is it given in ‘pulses’?

A

allow time for IS to recover, so give pulses of chemotherapy when BM cells have recovered but tumour cells haven’t significantly recovered.

33
Q

examples of tumours highly sensitive to chemotherapy?

A
lymphomas
germ cell tumours
neuroblastoma
small cell lung cancer
Wilm's tumour (nephroblastoma)- tumour of kidneys which occurs in children
34
Q

examples of tumours with modest sensitivity to chemotherapy?

A
breast
colorectal
bladder
ovary
cervix
35
Q

examples of tumours with low sensitivity to chemotherapy?

A

prostate
brain
endometrial
renal cell

36
Q

alkylating agents damage DNA, where do they act in the cell cycle?

A

all phases, and so are cell-cycle non-specific

physical disruption to DNA interferes with DNA replication and RNA transcription

37
Q

how do alkylating agents damage DNA?

A

they covalently bind to the 2 strands forming the double helix of DNA, preventing these strands from separating so DNA unable to unravel for replication.

38
Q

examples of atoms along DNA with spare e- pairs for forming covalent bonds with DNA alkylating agents?

A

N7 and O6 atoms in guanine
N1 and N in adenine
N3 in cytosine

these site are nucleophilic- they are +ve charge attracting as have spare e- pairs.

39
Q

common steps in the chemical reactions that alkylating agents undergo with DNA?

A

given IV, and in plasma Cl- groups stay attached to drug as relatively high plasma Cl- conc.
drug enters cell, where labile -vly charged Cl- groups are easier to lose as lower Cl- cytoplasmic conc.
when close to their exposed DNA target site, loss of Cl- results in net +vly charged carbonium ion, or +vly charged Pt2+ in platins.
These groups can then react with nucleophilic DNA groups on DNA bases forming stable covalent bonds*(contrast to ionic bonds drugs such as NSAIDs and warfarin form with plasma proteins which means other drugs can displace them.)
as 2 sets of individual e- pair accepting sites on each carbonium ion, cross linking occurs at 2 spatially separate sites on DNA strands.

both inter and intra strand links can then be formed producing DNA adducts. cross linking can also occur between DNA and proteins.

40
Q

primary cause of cell death mediated by DNA alkylating agents?

A

inhibition of DNA replication

41
Q

specific ADRs with alkylating drugs?

A

infertility, especially in males

acute non-lymphocytic leukaemia

42
Q

examples of cancers platinum compounds (alkylating agents) are used in?

A

colorectal

ovarian

43
Q

active modeity of 5-FU? how does this affect DNA synthesis?

A

5-FdUMP

binds to thymidylate synthase and inhibits its action, stopping the synthesis of pyrimidines required in DNA

44
Q

how does MTX act in cancer chemotherapy?

A

DHFR inhibitor

turns off folate cycle, so don’t produce purines= A and G, necessary in DNA

45
Q

cancers MTX is used in?

A

brain tumours

leukaemia

46
Q

cancer 5-FU partic. used in?

A

GI cancers

47
Q

what exactly do vinca alkaloids inhibit to stop microtubule formation?

A

polymerisation

48
Q

example vinca alkaloid drug used in lung cancer?

A

vinorelbine

49
Q

example of tumour treated with the taxane paclitaxel?

A

ovarian cancer

50
Q

which drugs stimulate polymerisation and inhibit depolymerisation of microtubules during mitosis of cells?

A

taxanes e.g. paclitaxel

51
Q

stage of cell cycle cancer cells are stuck in due to action of taxanes and vinca alkaloids (spindle poisons)?

A

metaphase

52
Q

which pump on cells is important for trying to efflux cancer chemotherapy drugs from cell?

A

P-glycoprotein

53
Q

how can glutathione be involved in development of resistance to chemotherapy drugs?

A

binds to the drug and blocks its activity

54
Q

3 mechanisms of resistance to cancer chemotherapy drugs?

A

increased efflux or decreased influx
inactivation in cell
enhanced repair of DNA lesions

55
Q

when is the performance status significant?

A

in the palliative setting, not using chemotherapy to cure the patient, not useful if poor performance status drug’s desired effect doesn’t outweigh significant ADRs- ptnt will have serious problems with toxicity e.g. neutropenic sepsis and life threatening infections.

56
Q

why are IV pumps e.g. PICC and Hickman lines, good for use in chemotherapy drug administration?

A

less risk of arm pain and phlebitis

57
Q

how can we give chemotherapy tment to try and avoid emergence of resistance?

A

combine drugs together from different classes- have differing mechanisms of resistance

58
Q

what must be considered when giving combination chemotherapy?

A

NON OVERLAPPING TOXICITY- drugs that are given have different ADRs

59
Q

how can acute renal failure due to rapid tumour lysis be prevented?

A

can give allopurinol

60
Q

how can chemotherapy targeting lymphomas have an adverse effect?

A

can cause GI perforation at site of tumour

61
Q

what adverse effect might happen if treating acute myeloid leukaemia with chemotherapy?

A

disseminated intravascular coagulopathy within a few hrs of starting tment

62
Q

how might alopecia be coped with?

A

scalp cooling

63
Q

skin toxicity caused by bleomycin?

A

hyperkeratosis
hyperpigmentation
ulcerated pressure sores

64
Q

what are Beau’s lines?

A

deep lines running side to side on the fingernail
can occur with chemotherapy as each round stops nail growth, so white line appears, number of lines= number of chemotherapy sessions had.

65
Q

symptoms of mucositis?

A

sore mouth/throat
diarrhoea
GI bleed

66
Q

cardiotoxicity caused by chemotherapy?

A

cardiomyopathy e.g. caused by doxorubicin= anthracycline, and high dose cyclophosphamide= alkylating agent- also used in SLE.
arrhythmias e.g. with cyclophosphamide and etoposide= topoisomerase inhibitor.

67
Q

most frequent dose limiting toxicity of chemotherapeutics?

A

haematological toxicity

68
Q

what factors cause abnormalities in drug distribution of chemotherapeutics?

A

weight loss
ascites
reduced body fat

69
Q

important drug interaction of vincristine?

A

with anti-fungal= itraconazole, leads to more neuropathy

70
Q

important drug interaction of oral 5-FU?

A

with warfarin- INR increased, may be serious bleeding
St John’s wart
grapefruit juice

71
Q

important drug interactions of MTX?

A

penicillin- reduced clearance of MTX- MTX toxicity e.g. pulmonary fibrosis and heaptotoxicity, NSAIDs- protein binding, increase MTX toxicity

72
Q

when might 5-FU be used as a cream (topical tment)?

A

skin cancer

73
Q

examples of monitoring required during tment with chemotherapy?

A

response of cancer= tumour marker blood tests
radiological imaging
BM/cytogenics

drug levels e.g. MTX- drug assays to check clearance from blood

checks for organ damage= creatinine clearance
ECG

74
Q

define chemotherapy

A

tment of cancer with cytotoxic drugs

75
Q

what does the log cell kill model of tumour growth and regression predict?

A

that the effects of chemotherapy can be modeled as a 1st order process, so a given dose of drug kills a constant fraction of tumour cells, and the number of cells killed depends on the total no. of cells remaining.

giving higher doses to ensure eradication is difficult due to toxic ADRs and development of resistance- multidrug resistance proteins= actively transport a range of hydrophobic molecules out of the cell.

so a log kill ratio of 3 means that if 10^9 tumour cells, 1 round of chemotherapy will result in 10^6 tumour cells left, so will be left with 1000000 cells, so 999,000,000 cells have been killed. with a 2nd round of chemotherapy, 10^3 cells will be left, so this time 999,000 cells have been killed.

76
Q

what factors confound and constrain the log kill ratio?

A

compartmentalisation

drug resistance