Week 6: Cancer Chemotherapy Flashcards

1
Q

The two main categories of genes acted upon by mutation are recognised as:

A

 Proto-oncogenes (inactive) transformed by mutation to oncogenes (active). These genes control cell division apoptosis and differentiation.
 Tumour suppressor genes. The loss of function these suppressor genes can be critical in triggering carcinogenesis.

Transformation of a normal cell into a cancer cell requires a number of these control elements to be lost. Once this loss has taken place, the following characteristics of cancer cells define the magnitude of malignant threat.

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

Describe Loss of cell growth control as one of the hallmarks of cancer

A

The key feature of cancer cells is that the normal mechanism controlling proliferation and ordered cell death (apoptosis) is lost. This does not mean that the cancer cells are necessarily proliferating faster than normal cells, but that the ordered control of cell division balanced by apoptosis is lost in cancer cells.

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

Describe De-differentiation and loss of specific function as one of the hallmarks of cancer

A

Normal cells have a specific function to fulfil, which is lost when they turn cancerous. The degree of de-differentiation in cancers varies considerably, but the greater this loss of original identity, the worse the malignant challenge to the body generally is.

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

Describe blood supply as one of the hallmarks of cancer

A

As the cancer cells proliferate to reach the dimensions of a clinically discernible tumour, there is a natural limit to growth due to limited blood supply. This would restrict tumour growth to about 1-2 mm in diameter. To overcome this limitation, the tumour cells produce their own local angiogenesis factors to promote vessel growth into the tumour, so it can continue to develop.

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

Describe Metastasis and invasiveness as one of the hallmarks of cancer

A

Even with the additional vascular supply, if these de-differentiated cells in the tumour stayed in one location, then killing them would be easier. The threat they pose could be better contained through direct surgery and irradiation. However, malignant cells may also lose the very specific positional sense and identity within their parent tissue. This arises due to changes in cell surface proteins. These changes allow them to migrate or metastasise throughout the body.
This takes place via the lymphatic or vascular system or more locally by invasion of an adjacent body cavity. They can then proliferate further afield and produce new tumours. In addition as the cancer progresses over time, further mutations in cancer cells result in increased heterogeneity, with distinct cell lines arising that increase the malignant challenge to be met by chemotherapy

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

Given the physiological limitations on tumour growth, not all cells in a tumour are involved in active proliferation and normally belong to one of three compartments. These compartments tend also to relate to their spatial position in the body of the tumour. Outline the compartments.

A

 Compartment A – Dividing cells receiving adequate nutrient/vascular supply
 Compartment B – Resting cells remain in ‘G0’ phase but able to re- join Compartment
A if there are changes in cell signalling or the /local environment. – e.g. following surgery. More likely to situated in the middle of the body of the tumour.
 Compartment C – Cells no longer able to divide, but act to contribute to the overall bulk of a tumour. These present no challenge.
Note: cancer cells can still die off on their own if minimal nutrient supply is not maintained.
Compartment A may represent between 5-20% of the actual tumour cell population and is the one most susceptible to chemotherapy targeting cells at one or more stages in the cell cycle.
Therapeutically, cells in Compartment B present a real problem. They may be affected to some degree by chemotherapy as some resting RNA and protein synthesis will be taking place. But the proportion of chromosomal DNA in ‘G0’ that is open to attack is much more limited. This makes the effective kill ratio attainable in this compartment much lower. They will thus be 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
7
Q

when is a tumor clinically detectable?

A

Typically, a tumour needs to consist of a cluster of about 109cells before it is clinically detectable or to reach the size of a small grape.

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

Describe The log kill ratio

A

Typically, a tumour needs to consist of a cluster of about 109cells before it is clinically detectable or to reach the size of a small grape. The underlying model utilized by clinicians is based on log kill ratios. This means that if a given treatment kills 104 or 99.99% of cancer cells then for a population of 109 cells, this would mean a reduction to 105 or to 0.01% of the original population of 109 cells. The 105 cancer cells remaining would resemble a sphere the size of a full stop.
This would be a four log kill ratio and further treatment would be warranted so another dose of the same four log kill therapy would then result in only 101 or just 10 cancer cells remaining. This proportionate killing is comparable with the ideas around first order pharmacokinetics covered in the first part of the Workbook.
In this theoretical model this sounds really good, but the factors outlined earlier mean that of the 109 cells perhaps only 107- 108 may be available for direct attack (Compartment A) with the more resistant resting Compartment B cells able to revert hydra like, to aggressive ‘A’ types post therapy.
In reality, more modest kill ratios than given in example above may be attained that require repeated chemotherapy. The kill rate for cancer cells has to be weighed up against the kill rate for healthy cells in susceptible tissues.

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

Cancer chemotherapy: Agents directly modifying DNA structure

DNA Intercalation and Topoisomerase II Inhibition – Anthracycline Antibiotics. Describe the mechanism of action.

A

The anthracycline antibiotics are used in synergistic combination with other drugs with different mechanisms to reduce the risk of additive ADRs. Members of this group include doxorubicin and daunorubicin. Their discrete molecular ring structure enables them to intercalate between the spaces between DNA base pairs. This intercalation would interfere with normal transcription and replication. The anthracyclines particularly affect the activity of Topoisomerase II.
Topoisomerase IIenables the breaking, rotation and re-ligation of DNA strands during DNA replication and repair. The intercalated antibiotic molecule physically interferes with this. This action is further augmented by the anthracycline binding to the Topoisomerase II thus forming a tripartite DNA – anthracycline – Topoisomerase II complex. This hinders its availability for deployment elsewhere. The resulting non-ligated free strands of DNA act as a trigger to apoptosis.
As a secondary action, the anthracyclines are able to generate free radicals by binding free Fe2+. The locally produced free radicals then go onto further damage the DNA. Overall, the damage caused by anthracyclines is detected by DNA damage sensing mechanisms in the cell which then trigger apoptosis. In contrast to bleomycin, (see below) their action is non-cell cycle specific

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

Cancer chemotherapy: Agents directly modifying DNA structure
DNA Scission –Bleomycin.
Describe the mechanism of action

A

Bleomycin is a glycopeptide antibiotic derived from Streptomyces fungi. Bleomycin can both bind with DNA and chelate with free Fe2+ ions. Its structure allows it to closely align itself within DNA by both intercalation but also by binding via its terminal NH2 group to DNA. When it chelates with free cytoplasmic Fe2+ ions, this reaction site then catalyses production of both superoxide and hydroxide free radical species These highly reactive free radical species then attack phosphodiester bonds in the DNA strand resulting in a cutting or scission of DNA strands. This scission is considered to be the primary mechanism underlying its cytotoxicity and is comparable with the anthracyclines in this respect. The production of free radicals is also considered to underlie its pulmonary toxicity. Bleomycin is most effective during G2, but also has some effect in non-replicating G0 cells.

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

Cancer chemotherapy: Agents directly modifying DNA structure.
Alkylating Agents Related Compounds and The Platins
Describe the mechanism of action

A

Clinically, the name is often used to refer to compounds used in chemotherapy that covalently bind to DNA. The primary underlying mechanism for these related agents is that they typically possess two highly labile or ‘leaving’ groups such as – Cl, that are necessary for instigating the covalent reaction with the DNA.
Nucleophilic target sites for these agents are spread along the whole length of DNA
Along the length of DNA strands the specific molecular target sites are nucleophilic, or positive charge attracting, groups. These are very common and their locations are very well defined. These include:
N7 and O6 atoms in guanine; N1 and N3 in adenine; N3 in cytosine. These all have spare electron pairs to donate to a vacant electron orbital. They can therefore make a covalent bond with those agents the alkylating group. The reactive group on alkylating agents are therefore electrophilic or ‘electron attracting’

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

Antimetabolites – interference with precursors to purine and pyrimidine synthesis
Describe the mechanism of action

A

Antimetabolites used in cancer chemotherapy are structurally related to precursors involved in DNA synthesis. They act to interfere with the production of the purine or pyrimidine nucleotides. This is by acting as a direct competitive analogue in DNA or RNA synthesis such as 6- Mercaptopurine (a purine analogue), 5-Fluorouracil or 5-FU (a pyrimidine analogue).
In addition, inhibition of key enzymes involved in precursor synthesis is a common therapeutic option, with methotrexate used as a highly potent inhibitor of Dihydrofolate Reductase or DHFR. Brief consideration is given here to the mechanism of methotrexate and 5-fluorouracil from this large group of therapeutic agents.

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

Methotrexate acts by inhibiting DHFR to interfere with folate metabolism. Describe this process

A

Folates are vital for production of purine nucleotides and thymidilate. These feed into the pathway that enables DNA and RNA synthesis. After being actively taken up into the cell, folate is converted in to a polyglutamate. Then, the polyglutamated folate is reduced in two steps by the enzyme DHFR to produce a tetradyhdrofolate, or FH4.
Polyglutamated FH4acts as a co-factor acting as a methyl group carrier. This methyl group is then used in the transformation of 2 - deoxyuridylate or DUMP into 2- deoxythymidylate or DTMP. This transformation is carried out by the Thymidylate Synthase enzyme.
Along with other related pathways, the production of DTMP contributes to the production of the purines and pyrimidines that act as molecular building blocks for both DNA and RNA. Methotrexate is structurally very similar to folic acid and is actively transported into the cell and also polyglutamated. Methotrexate on its own has a much higher (x1000) affinity for DHFR
As a result, both Methotrexate alone and its polyglutamated form act to vastly reduce the metabolism of folate to reduce nucleotide production. The decrease in thymidine levels being the most marked. The controlled dosages used in cancer chemotherapy are much higher than used in RA and the aim is to reduce cancer cell growth as rapidly as possible by effectively removing nucleotide precursors from the cell metabolite pool.

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

why is chemotherapy most commonly prescribed IV?

A

With many of these drugs, their toxicity rules out oral delivery as they would severely damage the GI tract. In addition bioavailability is often variable and only a few agents are given orally. In addition, the patient is highly likely to experience nausea and vomiting throughout treatment further limiting practicability of the oral route.
The most practical route for systemic administration of these agents is intravenous. This allows fine control of delivery by injected bolus an infusion bag or pump infusion. If an emergency arises due to appearance of adverse effects, the infusion can be stopped immediately.
If a tumour is localised, then direct intraregional delivery may be possible. If it is within a defined space for example the bladder or lung effusion may be performed. Intrathecal and intraventricular delivery is used for treating tumours in the CNS.

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

Drug resistance in cancer chemotherapy - Primary and acquired resistance

A

Traditional agents used in cancer chemotherapy are, by definition, cytotoxic. Neither healthy or cancer ‘know’ these drugs are being used to defeat a systemic threat to the whole body. Consequently they will treat many of these agents in the way they would deal with any other potentially toxic xenobiotic molecule.
The terms primary and acquired resistance are respectively defined as resistance being present prior to drug exposure and subsequent to drug exposure. These resistances in tumour cells can be due to their adapting and upregulating responses to repeated ‘xenobiotic’ chemotherapeutic challenge, or they can arise as a result of mutations in the cancer cell genome driving increased MDRP expression/activity.

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

Main factors contributing to chemotherapy drug resistance

A

Multidrug Resistance Protein – MDRP. This P-Glycoprotein (P is for ‘permeability’) is generally expressed throughout most healthy cells at low levels. High levels are notably expressed in the kidney, liver and GI tract. MDRP functions to generically remove hydrophobic (ie charged) large xenobiotics
If cancer cells repeatedly encounters one or more of the chemotherapeutic drugs described here then expression of MDRP may increase as a result. Importantly, as MDRP activity is non-specific, it can then act can remove structurally dissimilar molecules used in combination chemotherapy. For example prior exposure to doxorubicin can then also increase efflux and resistance to vinblastine or cis-platin to limit efficacy of future dosing.
Other inducible mechanisms – Drug exposure can also act to decrease the rate of active uptake of drugs such as methotrexate or cis-platin by downregulation of the active carrier.
Conversely, drug target enzymes expression can be upregulated to offset the decrease in production of metabolites e.g. DHFR ↑ with methotrexate. Further examples include increased rate of repair of drug induced lesions in DNA or membrane following increased expression of repair enzymes in cancer cells. For example, this is seen with use of the alkylating agents or bleomycin which act to damage DNA.

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

Cancer chemotherapeutic ADRs

A

As they are primarily targeted at rapidly dividing cells, they also affect normal cells with a higher rate of proliferation. This includes most of the GI tract, especially the buccal mucosa, and hair follicles. There are however other ADRs, which are manifest with specific groups or drugs.
Common ADRs
These include severe nausea, vomiting and diarrhoea. Mucositis, alopecia and myelosuppresion, impaired wound healing skin toxicity are also common. More specific within group ADRs, include neurotoxicity, cardio, bladder, lung, and renal toxicity. The latter three are irreversible.
Of the ADRs, the most frequent dose limiting is haematological toxicity and is the most frequent cause of death during chemotherapy. Different agents have varying effects in both degree and lineage e.g. neutrophils and platelets.
In addition, acute renal failure is also an expected ADR. Hyperuricaemia caused by rapid tumour lysis during treatment can result in a large increases in purines being released into the circulation. The subsequent increase in purine metabolism generates urates. This can lead to precipitation of urate crystals in renal tubules and in extreme cases subsequent kidney failure and death.

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

Some side effects associated with main cytotoxic groups

A

Antimetabolites – The example drugs given here largely exhibit all the general ones listed. With high dose methotrexate treatment there is higher risk of renal failure.
Cytotoxic antibiotics 1 – Cardiotoxicity: The anthracyclines doxorubicin and daunorubicin are especially cardiotoxic due to free radical generation.
Cytotoxic antibiotics 2 – Pulmonary Toxicity: Risk of pulmonary fibrosis with bleomycin is high at about 10% with a 1% fatality rate. These effects of both are cumulative and irreversible.
Alkylating agents – Cis-platin at high dose, peripheral, sensory and motor neuropathy. High frequency ototoxicity is common.
Mitotic spindle inhibitors – High dose neurotoxicity as ‘stocking and glove’ peripheral neuropathy is often reported.

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

describe the basic components of DNA

A
  • Nucleotide = sugarphosphate- base
  • DNA = double helix of nucleotides
  • Purines = Adenine & Guanine
  • Pyridimines = Cytosine & Thymine (Uracil in RNA)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Classification of Tumours According to Chemo-sensitivity

A

Highly Sensitive
Lymphomas
Germ cell tumours Small cell lung Neuroblastoma Wilm’s tumour
Modest Sensitivity
Breast Colorectal Bladder Ovary Cervix
Low Sensitivity
Prostate Renal cell Brain tumours Endometrial

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

site of Action of antimetabolites

A

DNA synthesis

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

site of Action of alkylating agents

A

DNA

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

site of Action of intercalating agents

A

dna transcription and duplication

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

site of Action of spindle poisons

A

mitosis
• Once chromosomes are aligned at metaphase plate, spindle microtubules depolymerize, moving sister chromatids toward opposite poles
• Nuclear membrane re-forms and cytoplasm divides

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

Microtubule-binding agents affect microtubule dynamics in 2 ways

A

o Inhibit polymerisation

o Stimulate polymerisation and prevent depolymerisation

26
Q

Alkylating Agents: Mechanism of Resistance

A

deactivation entry or increased exit of agent
inactivation of agent in cell
enhanced repair of DNA lesions produced by alkylation

27
Q

Chemotherapy: clinical indications

A

• May seem obvious… Cancer
• but aim is very different in different malignancies
• predicted response is also different within the same cancer based on:
o performance score
o clinical stage
o prognostic factors or score (often involving biological factors) o molecular or cytogenetic markers
• Side effects vs anticipated or best outcome

28
Q

Chemotherapy: regimens

A
  • A single drug
  • Or
  • Combination chemotherapy
  • consisting of a number of different drugs • usually given an acronym
29
Q

Chemotherapy: route of administration

A

o IV is the most common – bolus, infusional bag, continuous
pump infusion
o PO convenient, dependent on oral bioavailability
o SC convenient in community setting
o Into a body cavity – bladder, pleural effusion
o Intralesional - directly into a cancerous area – consider pH
o Intrathecal - into the CSF – by lumbar puncture or omaya reservoir (directly into ventricles)
o Topical -medication will be applied onto the skin o IM rarely

30
Q

name two types of IV pumps

A

PICC Line

Hickman Line

31
Q

Chemotherapy adverse effects:

due to effect of treatment on the tumour

A
  • Acute renal failure - often multifactorial – hyperuricaemia caused by rapid tumour lysis leads to precipitation of urate crystals in renal tubules
  • GI perforation at site of tumour – reported in lymphoma
  • Disseminated intravascular coagulopathy eg onset within a few hours of starting treatment for acute myeloid leukaemia
32
Q

Chemotherapy: side effects Vomiting

A

• Multifactorial but includes direct action of chemotherapy drugs on the central chemoreceptor trigger zone
• Patterns of emesis
o acute phase 4 - 12 hours
o delayed onset, 2 - 5 days later
o chronic phase, may persist up to 14 days

33
Q

Chemotherapy: side effects Alopecia

A
  • Hair thins at 2 - 3 weeks
  • May be total
  • May re-grow during therapy
  • Marked with doxorubicin, vinca alkaloids, cyclophosphamide
  • Minimal with platinums
  • Help sometimes with scalp cooling
34
Q

Chemotherapy: side effects Skin Toxicity

A
Local
o Irritation and thrombophlebitis of veins o Extravasation
• General
o Bleomycin
• Hyperkeratosis
• Hyperpigmentation
• Ulcerated pressure sores
o Busulphan, doxorubicin, cyclophosphamide, actinomycin D
• Hyperpigmentation
35
Q

Chemotherapy: side effects Mucositis

A

• Gastrointestinal tract epithelial damage
• May be profound and involve whole tract • Most commonly worst in oropharynx
• Presents as
o Sore mouth/throat o Diarrhoea
o G.I. bleed

36
Q

Chemotherapy: side effects Cardio-Toxicity

A

• Cardio-myopathy
o doxorubicin ++ (> 550 mg/m2) o high dose cyclophosphamide o mortality approx. 50%
• Arrhythmias
o cyclophosphamide o etoposide

37
Q

Chemotherapy: side effects Lung Toxicity

A
• Bleomycin
o pulmonary fibrosis
o beware concurrent radiotherapy
• Mitomycin C, cyclophosphamide, melphalan, chlorambucil
o pulmonary fibrosis
38
Q

Haematological Toxicity of Cancer Therapy

A

• Most frequent dose limiting toxicity
• Most frequent cause of death from toxicity
• Different agents cause variable effects on degree and lineages
o Neutrophils o Platelets

39
Q

Chemotherapy: cytotoxic drugs

A

• Considered ‘dangerous’ drugs, need specialists to prescribe because:
• Narrow therapeutic indices
• Significant side effect profile
• Dose needs to be altered for the individual patient
• Treatment phasing requires a balance
See next slide for some criteria
• Hence…weigh up the role and dose of chemotherapy for every cancer patient individually and always remember the aim of the treatment

40
Q

Chemotherapy: individual factors

A
  • Dose needs to be altered for the individual patient based on
  • their surface area and/or body mass index
  • drug handling ability (eg liver function, renal function… dependent on the metabolism and excretion routes)
  • general wellbeing (performance status and comorbidity)
  • Treatment phasing needs to take into account balance between:
  • growth fraction
  • the ‘cell kill’ of each cycle of the chemotherapy regimen
  • marrow and GI tract recovery before next cycle
  • how tolerable is the regimen – both short term organ toxicity & physical side effects & long term damage causing late effects
41
Q

Pharmacokinetics and chemotherapy

What causes variability?

A
• Abnormalities in absorption
N+V, compliance, gut problems
• Abnormalities in distribution
Weight loss, reduced body fat, ascites etc
• Abnormalities in elimination
Liver and renal dysfunction, other meds
• Abnormalities in protein binding
Low alb, other drugs
42
Q

Chemotherapy: important drug interactions

A

• Other drugs may increase plasma levels of the chemotherapy drug (and therefore side effects)
o Vincristine and itraconazole (a commonly used antifungal) leads to more neuropathy
o Capecitabine (oral 5FU) and warfarin
o Methotrexate – caution with prescribing penicillin,
NSAIDs
o Capecitabine and St Johns Wort, grapefruit juice

43
Q

Chemotherapy: monitoring during treatment

A

• Response of the cancer
o Radiological imaging
o Tumour marker blood tests o Bone marrow/cytogenetics
• Drug levels
o Eg Methotrexate drug assays taken on serial days to ensure
clearance from the blood after folinic acid rescue • Checks for organ damage
o Creatinine clearance o Echocardiogram

44
Q

Define Neoadjuvant chemotherapy

A

given before surgery or radiotherapy for the primary cancer

45
Q

Define Adjuvant chemotherapy

A

given after surgery to excise the primary cancer, aiming to reduce relapse risk eg breast cancer

46
Q

Define Palliative chemotherapy

A

to treat current or anticipated symptoms without curative intent

47
Q

Define Primary chemotherapy

A

1st line treatment of cancer.. In many haematological cancers this will be with curative intent, initially aiming for remissiom

48
Q

Define Salvage chemotherapy

A

– chemotherapy for relapsed disease

49
Q

A 60 year man (PMH hypertension, osteoarthritis; heavy smoker; good exercise tolerance; prescribed medication is bendrofluazide and aspirin with prn ibuprofen) was admitted to hospital dehydrated with a 6 week history of progressive dysphagia and is diagnosed at endoscopy with oesophageal cancer.
A CT scan of thorax and abdomen suggests this has not spread. His case is discussed at the upper GI malignancy Multidisciplinary meeting. He attends the outpatient clinic one week after discharge.
He is offered surgical resection of the tumour and neoadjuvant chemotherapy.
What is the meaning of neoadjuvant in this context? What is the role of the chemotherapy?

A

Neoadjuvant chemotherapy: chemotherapy given before surgery to shrink the size of the tumour
Reducing the size of the tumour gives more surgical options

50
Q

A 60 year man (PMH hypertension, osteoarthritis; heavy smoker; good exercise tolerance; prescribed medication is bendrofluazide and aspirin with prn ibuprofen) was admitted to hospital dehydrated with a 6 week history of progressive dysphagia and is diagnosed at endoscopy with oesophageal cancer
What tests do you want to do prior to dosing his chemotherapy, particularly in view of his PMH and his clinical presentation?

A

Definite requirements:-

Renal , liver function tests and electrolytes

51
Q

The dose of his chemotherapy drugs is going to be worked out based on his body surface area (BSA).
What measurements need taking in clinic? Find out the formula for calculating BSA.

A

square root [(height in cm x weight kg) /3600]

52
Q

A 60 year man (PMH hypertension, osteoarthritis; heavy smoker; good exercise tolerance; prescribed medication is bendrofluazide and aspirin with prn ibuprofen) was admitted to hospital dehydrated with a 6 week history of progressive dysphagia and is diagnosed at endoscopy with oesophageal cancer.
A CT scan of thorax and abdomen suggests this has not spread. His case is discussed at the upper GI malignancy Multidisciplinary meeting. He attends the outpatient clinic one week after discharge.
He is offered surgical resection of the tumour and neoadjuvant chemotherapy.

You read the side effect profile of this regimen and discover there is a moderately high chance of him becoming thrombocytopenic to the extent of causing easy bruising and bleeding.
4. What advice should you give him about his existing medication?

A

Aspirin and ibuprofen both cause thrombocytopenia.
While aspirin is thought to complement cisplatin it is recommended to stop with erubicin which lowers platelet count.
General recommendation that aspirin is stopped before starting chemotherapy

53
Q

List 5 common general side effects of chemotherapy

A
General side effects-
 Severe nausea and vomiting
 Diarrhoea
 Mucositis/Stomatitis
 Bone marrow suppression/ myelosuppression
 Alopecia
Some specific toxicities:–
Bleomycin- pulmonary fibrosis Doxorubicin- cardiotoxic Cyclophosphomide- bladder toxicity Methotrexate- megaloblastic anaemia
54
Q

What is Tamoxifen?

A

Tamoxifen is a selective oestrogen receptor modulator (SERM). SERMs are oestrogen related compounds that display selective agonist or antagonist activity.
Tamoxifen competes with oestrogen for binding with oestrogen receptor in breast tissue. Hence causing regression of some breast tumours.

55
Q

What are the risks and side effects of taking tamoxifen?

A

Hot flushes and nausea are the most frequent side effects Vaginal dryness
Endometrial hyperplasia and malignancy
Increase risk of deep vein thrombosis and pulmonary embolism

56
Q

A 45 yr old woman has had a mastectomy for breast cancer after she found a lump. She attends the clinic after the operation to discuss future treatment. Her breast cancer is Grade 3, tumour size 2.5cm, Lymph nodes were involved. Oestrogen receptor (ER) status negative. Her treatment options are discussed. An on-line prognostic factor based treatment model is used.
Should this patient be offered Tamoxifen?

A

No

Her oestrogen receptor status is negative hence not be offered

57
Q

A 45 yr old woman has had a mastectomy for breast cancer after she found a lump. She attends the clinic after the operation to discuss future treatment. Her breast cancer is Grade 3, tumour size 2.5cm, Lymph nodes were involved. Oestrogen receptor (ER) status negative. Her treatment options are discussed. She is told that with no further treatment she has 32.9% chance of being alive in 10 yrs time, 65.4% chance of having died of her cancer and 1.7% chance of having died of another cause. With combination chemotherapy her chance of being alive in 10 yrs time is increased by 27.3%.
What is her chance of being alive in 10 yrs with chemotherapy?

A

Chance of being alive is increased by 27.3%, means 32.9% is increased by 27.3%.
27.3% of 32.9% = 8.98% (9.0%)
So chance of being alive after chemotherapy = 32.9 + 9.0 = 41.9%

58
Q

List 3 classes of antiemetics which may be helpful and give an example of a drug for each class.

A

Phenothazines- Prochloperazine

5-HT3 receptor antagonist (setrons)- Ondansetron Dopamine receptor antagonist- Metoclopramide

59
Q

A 70 yr old woman has had a mastectomy for breast cancer picked up during mammography. She attends the clinic after the operation to discuss future treatment. Her breast cancer is Grade 2, tumour size 2.5cm, Lymph nodes were not involved. Oestrogen receptor (ER) status positive. Her treatment options are discussed. An on-line prognostic factor based treatment model is used.
She is told that with no further treatment she has 61.8% chance of being alive in 10yrs time, 14.9% chance of having died of her cancer and 23% chance of having died of another cause. With combination chemotherapy her chance of being alive in 10yrs time is increased by 3.1%. With hormone therapy her chance of being alive in 10yrs time is increased by 3.9%.
What features of this woman’s breast cancer lead her to have a higher survival rate without treatment?

A

Lymph nodes not involved
Mastectomy reduced the tumour bulk
Grade of cancer is lower
(Oestrogen receptor status positive hence will respond to tamoxifen not stated if tamoxifen was administered prior to mastectomy)

60
Q

The 70yr old patient declines chemo but starts tamoxifen. 6 months later she falls and sustains a non-pathological fracture of neck of femur. Post-op she then develops a lower limb deep venous thrombosis.
List her 3 risk factors in this case for VTE.

A

Tamoxifen

Fracture neck of femur Breast cancer

61
Q

What are the 2 current outpatient anticoagulant treatments available? For each list the delivery route and the mode of therapeutic monitoring required (in the absence of renal failure).

A

Warfarin:
 coumarin anticoagulants-antagonize the cofactor functions of vitamin K
 Given orally as tablets
 Need to monitor INR and adjust the dose , due to risk of bleeding
Heparin:
 Inhibits action of coagulation factors
 Administered subcutaneously or intravenously
 Monitor for bleeding