CCC Flashcards

1
Q

Which cancers are associated with the following chemicals?
1. aromatic amines
2. benzene
3. wood dust
4. vinyl chloride

A
  1. bladder cancer
  2. leukaemia
  3. nasal adenocarcinoma
  4. angiosarcomas
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How does radiation increase the risk of cancer?

A

by increasing DNA damage leading to the accumulation of mutations in tumour-supressor genes and oncogenes

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

which carcinoma are low fibre diets in the West associated with? why does the diet lead to carcinomas?

A

associated with colorectal carcinoma
low fibre diets lead to an increased transit time through the bowel, thereby increasing exposure to carcinogenic substances

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

which carcinoma is associated with smoked food in Japan?

A

gastric carcinoma

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

characteristic translocations can be induced by which cytotoxic drugs, and what cancer can they lead to?

A

may be induced by topoisomerase inhibitors and lead to an acute leukaemia

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

How does HPV cause cancer? Which cancers does it cause?

A

the E6 protein produced by HPV16 binds to and inactivates the p53 protein
this leads to dysregulation of the cell cycle and apoptotic pathways and subsequent malignant transformation of epithelial cells infected
it causes cervical and anal cancers - associated with sexual transmission of HPV

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

infection of epstein barr virus is associate with which cancers?

A

associated with non-Hodgkin’s lymphoma and other lymphomas

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

what is the most common genetic abnormality of epstein barr virus?

A

caused by epstein barr nuclear antigens (EBNA), an 8:14 translocation in which the proto-oncogene of c-myc on chromosome 8 becomes transcriptionally controlled by the conrol elements of immunoglobulin genes on chromosome 14

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

which cancers are associated with hepatitis B virus?

A

hepatocellular cancer, and leads to a greater than 100-fold increased risk

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

how do retrovirus infections cause cancers?

A

by integration into the cellular retroviruses can cause abnormal overexpression of oncogenes. Retroviral infection has been implicated in numerous animal tumours

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

in humans, the HTLV1 retrovirus infection is associated with which cancer?

A

T-cell lymphomas

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

which cancer is helicobacter pylori associated with?

A

causative factor in malignancy, particularly mucosal associated lymphoid tissue (MALT) tumours

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

what symptoms do cancer patients usually present with?

A

lumps - breast lumps, change in moles, nodes, nodules and MSK lumps
bleeding - haemoptysis, rectal bleeding, haematuria, post-menopausal or irregular menstrual bleeding
pain - chest or abdo pain, headache
change in function - change in bowel habit, new cough, dyspnoea, weight loss, fever, acute confusional state

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

what do these stages of T (primary tumour) of the TNM mean?
Tx
T0
Tis
T1, 2, 3, 4

A

Tx = primary tumour cannot be assessed
T0 = no evidence of primary tumour
Tis = carcinoma in situ
T1, 2, 3, 4 = increasing size and/or local extent of the primary tumour

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

what do these stages of N (regional lymph nodes) of TNM mean?
Nx
N0
N1, 2, 3

A

Nx = regional lymph nodes cannot be assessed
N0 = no regional lymph node metastasis
N1, 2, 3 = increasing involvement of regional lymph nodes

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

what do these stages of M (distant/organ metastasis) of TNM mean?
Mx
M0
M1

A

presence of distant metastasis cannot be assessed
no distant metastasis
distant metastasis

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

what does the grade of a tumour mean?

A

refers to the extent the tumour resembles normal tissue or has a bizarre appearance

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

what do the following grades mean?
GX
G1
G2
G3

A

GX = grade of differentiation cannot be assessed
G1 = well-differentiated: similarities remain to normal tissue of the organ of origin
G2 = moderately differentiated
G3 = poorly differentiated: bizarre cells

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

for which parts of the body is CT and PET-CT used?

A

evaluation of chest and abdominal malignancies

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

for which parts of the body is MRI used?

A

bone and soft tissue lesions, and regions where bone causes artefact in the CT appearances such as the pelvis or the posterior fossa of the brain

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

in the RECIST system for comparing clinical trials, what do the following terms mean?
complete response (CR)
partial response (PR)
stable disease (SD)
progressive disease (PD)

A

CR = no disease detectable radiologically
PR = all lesions have shrunk by at least 30% but disease still present
SD = less than 20% increase in size or less than 30% decrease in size
PD = new lesions or lesions that have increased in size by more than 20%

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

what is the rate of additional cancers for each CT scan?

A

one additional cancer per 1000-2000 scans

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

which tumours are MRIs the gold standard for imaging?

A

neurospinal, rectal, prostate and MSK tumours, and staging for subtypes of head and neck cancer

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

which devices are vulnerable to the effects of MRIs?

A

most pacemakers and implantable cardiac defibrillators
metallic foreign bodies eg vascular clips, surgical staples

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

what is the principal investigation for detection of skeletal metastases?

A

bone scintography (bone scan)

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

how does PET scanning work?

A

positron emission tomography detects high-energy emitted by short-lived radioisotopes, which can be chemically tethered to molecules such as glucose or somatostatin to form a trace eg fluorine 18 deoxyglucose (SDG-18), a radioactive form of glucose

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

what does the sensitivity and specificity of a tumour marker mean?

A

sensitivity of a marker describes its ability to detect those with a certain disease. If 100 people have the disease and the marker is elevated in only 95, sensitivity is 0.95
specificity describes its ability to accurately define those who are disease free. If in 100 disease-free people the marker is negative in only 90 (ie there are 10 false positives) the specificity of the test is 0.9

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

which tumour markers should a young male with disseminated bone metastases be tested for?

A

serum LDH, aFP, and BHCG (pregnancy test) to diagnose chemo sensitive and potentially curable germ cell tumours

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

which tumour markers are used in testicular teratoma?

A

HCG and aFP

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

when is CEA high?

A

in the setting of colorectal carcinoma
also more common in people who smoke, or have IBD, hepatitis, pancreatitis or gastritis

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

when are CA125 levels elevated?

A

mainly used as a tumour marker in ovarian carcinoma
also elevated in pancreatic, lung, colorectal and breast cancer; usually where these are disseminated to the abdominal cavity

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

what is alpha fetoprotein (aFP) and when is it elevated? when is it undetectable?

A

it is a glycoprotein produced by the normal foetal yolk sac, liver and intestines
levels are moderately elevated in hepatitis, but high levels are also produced by hepatocellular carcinoma and cancers containing yolk sac elements eg teratoma
it is undetectable in normal individuals after the first year of life

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

what is human chorionic gonadotrophin (HCG) and when is it elevated?

A

it is a glycoprotein consisting of two subunits
HCG is elevated in patients with gestational trophoblastic disease (hydatiform mole, choriosarcoma)
there is also a specific elevation of the B-subunit with non-seminomatous testicular cancers and some with seminoma
It is also raised in pregnancy!

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

what is prostate specific antigen (PSA) and when is it elevated?

A

it is a protein produced by prostatic cells
levels are raised in prostate cancers but also with benign hypertrophy of the prostate - a near ubiquitous phenomenon as men age
may also be elevated by rectal exam, in prostatitis and UTI

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

when are immunoglobulins used as tumour markers? how are they measured?

A

can be a measure of the paraproteinaemias eg myeloma and Waldenstrom’s macroglobulinaemia, and occasionally non-Hodgkin’s lymphoma
can be measured in the blood or their excretion can be measured as light chains in the urine (Bence-Jones protein), which occurs in 40-50% of all cases of myeloma

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

what are the different types of biopsies?

A

fine needle aspiration cytology
tru-cut needle biopsy = a piece of the tumour is sampled under local anaesthetic
incisional biopsy = a piece of the tumour is sampled at surgery
excisional biopsy = the whole of a mass is removed

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

what is neoadjuvant chemotherapy? what are the aims? for which cancers is it used?

A

pre-operative treatment of an operable tumour before definitive surgical intervention
aims are to make the tumour smaller, to allow less radical surgery, while at the same time treating occult micro metastases
established for osteosarcoma, and is being tested in clinical trials for other malignancies such as breast cancer

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

what is primary chemotherapy and when is it used?

A

initial chemotherapy for a tumour that is inoperable or of uncertain operability, where a reduction in the tumour bulk in a pre-defined manner may make surgery with curative intent feasible

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

what is adjuvant chemotherapy?

A

it is the use of chemotherapy following a complete macroscopic clearance at surgery
chemotherapy in this setting treats the occult microscopic metastases which we know usually lead to relapse after surgery for lymph node positive disease eg breast cancer and colorectal cancer.

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

when is tamoxifen used as a hormonal treatment for cancer?

A

used for in-situ breast cancer before invasive carcinoma is recognised (ie before overt malignancy appears)

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

which cells are usually affected by chemotherapy?

A

haematopoietic stem cells and the lining of the GI tract, producing low blood counts (myelosuppression) and mucositis

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

what is the usual schedule of cycles and for how long?

A

treatment every 3-4 weeks, and maximally effective after a 6 month course

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

which chemotherapy drugs are available orally?

A

cyclophosphamide, etoposide, capecitabine and tamoxifen

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

what is the route of most chemotherapy?

A

given IV as bolus injection or short infusion

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

when is chemotherapy given intravesical? what is the advantage of this?

A

chemotherapy is routinely given this way in the management of superficial bladder cancer
has the advantage of producing high doses at the site of the tumour, with negligible systemic absorption and hence minimal systemic toxicity

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

when is chemotherapy given intraperitoneal?

A

can be administered directly into the peritoneal cavity in the context of tumours that spread trans-coelomically (eg ovarian cancer)

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

when is chemotherapy given intra-arterial? what is the advantage?

A

any tumour that has a well-defined blood supply is potentially suitable eg hepatic artery infusion for liver metastases
this allows higher doses to be delivered to the involved site and reduces systemic toxicity

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

how are routine cytotoxic chemotherapy doses calculated?

A

according to the patient’s body surface area - most commonly used formula is that of DuBois and DuBois

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

which is the only chemotherapy drug to have its own dose calculation?

A

carboplatin - has its own dose calculated according to renal function

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

how does nausea arise as a side effect of chemotherapy? what can be used to control this?

A

nausea arises from a combination of direct simulation of the vomiting centre, peripheral stimulation and anticipatory causes
the use of 5-HT antagonist drugs like ondansetron can help with this

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

how is myelosuppression caused by chemotherapy? what is the lowest point of the drop known as? what is the neutrophil count? how long does it take to recover?

A

chemotherapy causes bone marrow suppression by killing haematopoietic progenitor cells
this leads to a leucopenia and thrombocytopenia generally after 10-14 days from the beginning of each cycle
the lowest point of this drop is known as the NADIR!
a neutrophil cunt greater than 1x10^9/1 is rarely associated with a clinical infection. However, a risk of infection with a count less than 0.5 x 10^9/1 is significant
haematopoietic recovery usually occurs after 3-4 weeks, enabling further cycles of chemotherapy to be given

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

what is oral mucositis indicating?

A

may reflect more general damage to the whole GI epithelium, a rapidly dividing cell population susceptible to cytotoxic chemotherapy

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

why does diarrhoea occur as a side effect of chemotherapy?

A

due to colitis or small bowel mucosal inflammation

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

why is constipation a side effect of chemotherapy?

A

constipation is usually due to dehydration with reduced oral intake due to nausea, and to adverse effects of other meds being taken eg opiate analgesics or 5-HT antagonists

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

what can be used to control the side effect alopecia?

A

can be controlled by the use of a cold cap which reduces the blood flow to the scalp

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

when does peripheral neuropathy occur as a side effect? how long does it take to recover from it?

A

these occur with the platinum drugs (particularly cisplatin), taxanes, and vinca alkaloids
neuropathy, principally affecting sensory nerves, may recover partially over a period of months, but patients are usually left with a residual deficit

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

which drugs are related to central neurological toxicity?

A

eg ifosfamide-induced encephalopathy and 5-FU induced cerebellar toxicity

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

how does cisplatin cause ototoxicity?

A

cochlear damage rather than auditory nerve damage is believed to be responsible for the high tone hearing loss associated with cisplatin
the effect is permanent
pre-existing high-tone hearing damage precludes the use of cisplatin

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

when does nephrotoxicity occur as a side effect of chemotherapy?

A

this occurs with platinum agents, principally cisplatin, and with the alkylating agent ifosfamide
renal excretion of many cytotoxics means that adequate renal function is required to reduce overall toxicity

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

which drugs cause bladder toxicity? give an example of an antidote

A

cyclophosphamide and ifosfamide cause haemorrhagic cystitis in a dose-dependent manner
antidotes exist such as Mesna

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

which chemotherapy drugs are associated with cardiac side effects?

A

doxorubicin and paclitaxel are both associated with acute arrhythmias
5-FU (and related drugs such as capecitabine) may cause coronary artery spasm and therefore induce cardiac ischaemia

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

when does extravasation occur as a side effect of chemotherapy?

A

some cytotoxic drugs are highly vesicant and cause tissue damage on extravasation. they are administered through fast-running drips under direct observation, to dilute any vesicant action and to make nurses administering treatment aware immediately if extravasation occurs

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

what is hand-foot syndrome? how is it resolved?

A

ie palmar plantar erythema
erythema of the palms of the hands and soles of the feet is frequently seen with 5-FU, capecitabine and some of the targeted agents eg sunitinib, erlotinib
side effect usually responds to drug withdrawal and emollients but patients will need to be reviewed for other drug side effects eg bowel toxicity in the case of 5-FU

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

which drugs cause photosensitivity?

A

some drugs such as 5-FU cause photosensitivity
patients should be advised regarding the use of high-factor sun blocks

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

which drugs cause pigmentation as a side effect?

A

bleomycin leads to skin and nail pigmentation
it occurs in combination with pulmonary fibrosis and a common pathogenic process is thought to be responsible

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

which drug causes myalgia and arthralgia as a side effect?

A

with use of paclitaxel and are usually well controlled with non-steroidal analgesia

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

which drugs are associated with frequent hypersensitivity reactions on administration?

A

both paclitaxel and docetaxel cause allergic reactions

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

how do chemotherapy drugs cause second malignancies? what is the most carcinogenic of the anti-cancer drugs?

A

some cause sub-lethal DNA damage that may eventually lead to the genetic changes require to induce a second malignancy
the most carcinogenic anti-cancer drugs are alkylating agents and procarbazine

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

which chemotherapy drugs cause infertility?

A

most drugs are associated with a reduction in fertility
some drugs, notably alkylating agents, render patients infertile at standard doses
most patients who have high-dose treatments become infertile

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

which chemotherapy drugs cause long term pulmonary damage?

A

may result from fibrosis induced by drugs such as bleomycin and busulphan
high-dose or prolonged administration of most alkylating agents is associated with pulmonary fibrosis or pneumonitis

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

which cancers can cause bone marrow replacement by malignant infiltration and produce pancytopenia?

A

more common in haematological malignancies and certain solid cancers eg breast, lung and prostate cancer

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

what is the type of anaemia usually cause by repeated chemotherapy?

A

often macrocytic but not megaloblastic

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

what are the investigations for myelosuppression?

A

a transient nadir in blood counts following chemotherapy can be observed
full evaluation (to check for bone marrow infiltration) includes a blood film, measurement of haematinics, bone marrow aspirate and trephine

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

what is the treatment for anaemia in myelosuppression?

A

heamoglobin less than 10 g/dl - may benefit from blood transfusion
use of recombinant erythropoetin in preventing symptomatic anaemia can be benficial and reduce risks of transfusion reactions and viral transmission

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

what are the clinical signs of significant thrombocytopenia?

A

petechial haemorrhage
spontaneous nose bleeds
corneal haemorrhage
haematuria

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

at what level would you give a patient a platelet transfusion due to thrombocytopenia?

A

platelet count less than 20 x 10^9/L

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

what are the potential consequences of neutropenia in chemo patients? when should patients seek help?

A

may quickly lead to multi-organ failure associated with septic shock - should seek immediate attention from the unit delivering their radiotherapy if they develop a fever

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

when does a patient require Abx in neutropenic sepsis?

A

total white counts less than 1 x 10^9/L with an associated fever require immediate in-patient management with broad spectrum antibiotics

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

why should rectal and vaginal examinations not be performed in suspected neutropenic sepsis?

A

due to the risk of causing bacteraemia if the mucosa is breached

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

which investigations are carried out for neutropenic sepsis?

A

extensive cultures of blood, urine, sputum, throat etc and a chest X-ray

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

when would a change in IV Abx occur with neutropenic sepsis? what would you give if the patient has a persistent fever despite appropriate antibiotic treatment?

A

failure to respond to initial antibiotics within approximately 48 hours requires a change to second-line broad-spectrum antibiotics
give patient additional antifungal or antiviral agents

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

when would prophylactic antibiotics be administered for neutropenic sepsis?

A

should be considered in the presence of chronic obstructive airways disease and the use of co-trimoxazole in patients with lymphoma at risk of pneumocystis pneumonia

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

what are the current targets of immunotherapy in cancer?

A

Cytotoxic T Lymphocyte Antigen 4 (CTLA4) expressed on T lymphocytes
Programmed cell death protein 1 (PD1) more broadly expressed across a range of immune cells including B cells and antigen-presenting cells, and its ligand
Programmed death Ligand 1 (PDL1) expressed on a broad range of both immune andcancer cells

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

how do long-acting monoclonal antibodies work?

A

they act via immune checkpoint blockage to release anti-tumour immunity
some may also act against cancer cells through complement pathways and antibody-dependent cytotoxicity

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

how are immunotherapy drugs administered and how are the doses calculated? give some examples

A

all are delivered intravenously and dosing is either calculated by weight or flat dosing based on idealised body weight
examples: ipilimumab, nivolumab, pembrolizumab and atezolizumab

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

is immunotherapy curative or palliative?

A

most treatment is palliative, for patients with advanced disease

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

what are the unusual features of cancer immune checkpoint inhibitor side effects?

A
  1. first side effects can occur any time from the start of first infusion until many months after the drug has been withdrawn
  2. many side effects do not wax and wane with the cycle of treatment (in contrast to chemo)
  3. peak incidence of new immune mediated side effects is 6-8 weeks after first treatment but the range is very wide
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

what are the types of rashes that can occur as a side effect of immunotherapy?

A

typically patients present with a maculopapular eczema type rash on the trunk. other patterns have been described including psoriatic plaques, angioedema and vitiligo > these will respond to topical emollients and steroid cream
rare but serious include steven johnson syndrome > these patients require high dose IV steroids and specialist referral

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

what are the symptoms of pneumonitis from immunotherapy?

A

can present with often dry cough, SOB, reduced exercise tolerance and fatigue
symptoms may develop rapidly over a few days or more slowly over several weeks
median time to onset after single agent nivolumab is 3-4 months and symptoms usually take 4-6 weeks to improve

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

what are the investigations and treatment of penumonitis?

A

initial: obs, FBC, CRP and CXR. full biochem screen can pick up other side effects and sputum, and screening for viral, opportunistic and bacterial chest infections is of benefit
most patients will benefit form oral steroids though some eg with pyrexia, high neutrophil and CRP, will require treatment for infection first

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

What does pneumonitis look like on Chest X-ray?

A

can look like infection
CXR shows consolidation and haziness

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

at what stage will diarrhoea be a side effect of immunotherapy?

A

within the first 2 months

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

what are. the features of concern with diarrhoea/colitis?

A

frequency >6 times a day
associated abdo pain
bloody diarrhoea
nausea
nocturnal diarrhoea

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

What are the initial assessments for diarrhoea/colitis as a side effect in immunotherapy? what investigation is useful in treatment later?

A

include obs, fluid balance, FBC, CRP, stool sample (for MCS & C.difficile toxin) and AXR. Flexible sigmoidoscopy is useful in guiding later treatment

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

what are the initial assessments for hepatitis as a side effect on immunotherapy?

A

should include a med review (consider recent statins and Abx), alcohol history, hepatitis blood screen (inc. viral hepatitis and iron studies) and imaging (usually USS) to investigate thrombosis and possible metastases

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

how are mild and serious cases of hepatitis treated as a side effect of immunotherapy managed?

A

mild: withhold cancer immunotherapy treatment and frequent biochemical re-assessment
serious: require immunosuppression with high dose steroids and specialist referral

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

how common is nephritis in cancer immunotherapy? what are the symptoms?

A

less common occurring in 1-4% of patients
consider if patients present with AKI
nephritis can be asymptomatic at first or present with symptoms of uraemia: weakness, fatigue, anorexia, malaise, thirst and reduced urine output

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

what are the initial assessments for nephritis caused by cancer immunotherapy?

A

include a careful history, focus on meds and infection and/or lower urinary tract symptoms
next evaluate fluid balance and perform biochemistry, urine dipstick (send for MCS to microbiology if appropriate) and urine protein/creatinine ratio

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

what are the differentials of nephritis caused by cancer immunotherapy?

A

dehydration , recent IV contrast, infection, meds, hypotension, obstructive uropathy

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

what is the management of serious cases of nephritis caused by cancer immunotherapy?

A

require immunosuppression with high dose steroids and specialist referral

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

what is the commonest pattern of nephritis in cancer immunotherapy-caused nephritis?

A

acute tubulo-interstitial nephritis with lymphocytic infiltration

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

what is the difference between myalgia and arthralgia? which is more common?

A

myalgia is muscle pain
arthralgia is joint pain without swelling
myalgia is more common than arthralgia

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

what are some more serious rheumatological complications of cancer immunotherapy?

A

acute myositis, polymyalgia rheumatica and arthritis

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

what are the initial assessments for myalgia/arthralgia as a side effect of cancer immunotherapy?

A

rheumatological history, exam of skin and joints and use of plain x-rays to exclude bone metastases

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

what is the management of myalgia/arthralgia as a side effect of cancer immunotherapy?

A

mild cases: manage symptomatically eg paracetamol and or ibuprofen
moderate or severe cases (ie patients with significant pain or swelling affecting function esp activities of daily living) benefit from specialist referral and oral or intra-articular steroids

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

what are the symptoms of endocrinopathies caused by cancer immunotherapy?

A

often present with non-specific symptoms such as fatigue, mild headache, mood change, malaise, thirst, weight change or feeling generally unwell

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

what are the standard safety bloods for patients on cancer immunotherapy? what are the assessments for toxicity?

A

should include regular assessment of thyroid function (T4, TSH) and cortisol (unless on systemic steroids)
acute assessment of toxicity may require a wider panel of hormone bloods along with simple obs (BP critical) and biochemistry (look for low sodium in adrenal insufficiency)

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

what is a common endocrine disorder caused by cancer immunotherapy?

A

thyroid disorders - in many cases acute thyroiditis cause hyperthyroidism (high T4 low TSH) followed after a delay of some weeks by hypothyroidism (low T4 high TSH)

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

what should you always check for and treat before starting thyroid replacement in cancer immunotherapy?

A

always check and treat any abnormality of adrenal function

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

acute pituitary inflammation (hypophysitis), caused by cancer immunotherapy, presents in which way and causes what? what are the tests for it? what is the management?

A

can cause headache, visual disturbance and a wide range of hormone abnormalities inc. adrenal insufficiency and secondary hypothyroidism (low T4 low TSH)
blood cortisol levels vary across the day (9am sample is most useful) and formal assessment of adrenal function with a short Synacthen test is ideal
patients with proven adrenal insufficiency or high clinical index of suspicion should start hydrocortisone replacement with adive about sick day rules (double dose), back up IM injection if vomiting and specialist referral

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

what is a short Synacthen test?

A

it is based on measurement of serum cortisol before and after an injection of synthetic ACTH - it assesses adrenal function

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

what are the side effects of steroids? what are the long term issues?

A

sleep disturbance, mood change, indigestion (+/- gastrointestinal bleeding), weight gain, high BP and increased risk of infection
long term: diabetes, osteopenia/osteoporosis, and proximal myopathy

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

what does it mean when cancers are hormone-dependent?

A

their rate of growth is influenced by levekls of hormones, and interfering with those hormones in one way or another may lead to growth arrest or tumour regression.

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

which cancers are most commonly hormone sensitive?

A

those arising within tissues under hormonal control of normal cellular proliferation or survival
including: prostate, breast and endometrium (sex hormones), lymphocytic malignancies such as lymphoma, leukaemia, and myeloma (corticosteroids)

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

at which stages are hormonal treatments used in cancer treatment?

A

neo-adjuvant or instead of surgery (primary medical therapy)
adjuvant therapy
to shrink established metastases and improve quality and duration of life (palliative therapy)

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

what receptors can be used in tumour cells to predict hormone sensitivity? give an example

A

the presence of cytoplasmic steroid receptors
eg oestrogen receptor (ER) in breast cancer

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

what is a medicinal way of inducing reversible “medical castration” in both men and women? how do they work? for which people is this unsuitable and why?

A

long-acting LHRH analogues eg goserelin, leuprorelin
by receptor down-regulation in the pituitary, block LH and FSH production and, in turn, gonadal hormone output
unsuitable for postmenopausal women for whom sex hormone production is mainly extra-gonadal, in fat and adrenal glands

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

The rate-limiting step in oestrogen synthesis is the conversion of androstenedione to oestrone by the enzyme aromatase. In postmenopausal women, andostenedione is secreted by what? why is this an important step?

A

it is secreted by the adrenal and aromatized in other tissues including fat and liver. This step is the target for aromatase inhibitors

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

what is aminoglutethimide and what does it need alongside it?

A

it was the first aromatase inhibitor and is non-specifc
as it also blocks an earlier stage in steroid synthesis, corticosteroid supplementation is required

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

what kind of drug is tamoxifen?

A

hormone inhibitor and acts as an anti-oestrogen

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

what are the 2 types of anti-androgen? how do they work?

A
  1. steroidal anti-androgens eg cyproterone acetate - have a dual action. in tumour cells they inhibit the androgen receptor, but in the hypothalamus they substitute for testosterone, so stimulate negative feedback inhibition with subsequent decrease in LHRH release
  2. non-steroidal anti-androgens eg bicalutamide - inhibit testosterone in both tumour cells and hypothalamus, so feedback inhibition is lost and serum testosterone levels rise
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

what is the “maximum androgen blockade”?

A

describes the combination of a non-steroidal anti androgen with an LHRH analogue to prevent the effect of non-steroidals, where they increase the serum testosterone levels, and is used as a therapeutic strategy in prostate cancer

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

how are glucocorticoids useful in cancer hormonal treatment?

A

glucocorticoids in high concentration induce apoptosis in some malignant lymphoid cells and form an important component of treatments for lymphoid leukaemias, lymphomas, myeloma and Hodgkin’s disease

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

what is the aim of using hormone supplementation in certain sex-hormone sensitive cancers?

A

to induce negative feedback loops eg oestrogen to down-regulate hypothalamic LHRH in prostate cancer, or tachyphylaxis (down-regulation) of receptors eg high-dose oestrogens in breast cancer

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

how do progestogens work? why are they widely used in palliative medicine?

A

may give direct inhibition of tumour growth via acting as an agonist of the progesterone receptor, but also produce negative feedback on the pituitary/gonadal axis
these drugs may also stimulate the appetite and are widely used in palliative medicine for that reason

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

what are the ways radiotherapy is delivered and what is the most common form in the UK?

A

using photons/x-rays electrons, radio-isotopes or protons
external beam radiotherapy using photons/x-rays is the most common form in the UK

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

how does radiotherapy work?

A

X-rays penetrate deep into body tissue whilst sparing the over-lying skin, where they produce secondary electrons and free radicals which cause DNA damage to both cancer cells and normal cells - this is the ‘skin-sparing’ effect

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

how is it that normal cells can repair after radiotherapy but cancer cells can not?

A

Normal cells can often repair the DNA damage and therefore survive. Conversely, cancer cells commonly have defective DNA repair pathways and are unable to repair radiotherapy induced DNA damage, with cancer cells subsequently undergoing cell death at the time of cell division (mitotic cell death) or apoptotic cell death

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

what does Gy mean?

A

the dose of radiation delivered to body tissue is expressed in the unit Gray (Gy)

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

what is a fraction?

A

radiotherapy is commonly delivered as a series of small doses called fractions rather than as a single large dose

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

what is a typical schedule for a head and neck cancer patient?

A

70Gy in 35 fractions over 7 weeks

132
Q

what are commonly used palliative schedules for radiotherapy?

A

8Gy in 1 fraction
20Gy in 5 fractions
30Gy in 10 fractions

133
Q

which cancers are highly radiosensitive and which do not respond well to radiotherapy?

A

seminoma and Hodgkin’s disease are highly radiosensitive and respond well to low doses of radiation whereas cancers such as a glioblastoma multiforme are relatively radio-resistant to even high doses of radiotherapy

134
Q

what is the therapeutic index with regards to radiotherapy?

A

the balance between tumour control and the side effects of radiotherapy

135
Q

what is concurrent chemotherapy?

A

chemotherapy given to patients during their radiotherapy treatment - it is thought to act as a radiosensitiser, increasing the sensitivity of the cells to radiation

136
Q

what is the most common external beam radiotherapy used in the UK?

A

3D-conformal radiotherapy

137
Q

what is the gross tumour volume, clinical target volume and planning target volume?

A

GTV: tumour is delineated on each CT slice it appears on
CTV: margin is then added for microscopic disease spread, which may include adjacent nodal groups
PTV: finally a further margin is added to allow for minor daily variations in patient and tumour position

138
Q

what is the multi-leaf collimator?

A

MLC: part of the head of the radiotherapy machine and helps to shape the beam of radiation

139
Q

does radiotherapy hurt?

A

radiotherapy treatment itself is painless and the patient neither sees nor fells anything as it is being delivered

140
Q

what are the 2 main types of side effects for radiotherapy and give examples for each?

A
  1. acute: usually after first 5-10 fractions. side effects tend to increase during treatment and often hit a peak in the first few weeks following the end of treatment eg localised skin reaction, oral mucositis and diarrhoea
  2. late: develop at least 3 months after radiotherapy and sometimes manifest years later. long-term toxicity occurs as some of the damage to normal cells cannot be repaired and partly due to the development of fibrosis and blood vessel damage within the irradiated tissue eg lung fibrosis, skin atrophy and infertility
141
Q

does having radiotherapy increase your chances of getting cancer?

A

yes - radiotherapy itself is a carcinogenic and there is a risk of a second malignancy developing as a result of treatment

142
Q

should you give radiotherapy to pregnant women?

A

radiotherapy is a teratogenic and therefore must be avoided in any woman who is pregnant

143
Q

what is brachytherapy?

A

a form of radiation treatment where radiation sources are placed within or close to the tumour
it allows the delivery of a localised high radiation dose to a small tumour volume, increasing the chance of tumour control whilst minimising the dose to surrounding normal tissue

144
Q

which cancers commonly have brachytherapy?

A

prostate, gynaecological, oesophageal and head and neck cancers

145
Q

what are the two main types of brachytherapy?

A

intracavity: the radioactive material is placed inside a body cavity such as the uterus and cervix
interstitial: where the material is put into the target such as the prostate

146
Q

what is important to notify a patient after brachytherapy?

A

Radiation protection is of particular importance in brachytherapy. Whilst a radioactive source is in situ the patient will be radioactive. Before discharge a patient will be advised about their on-going radiation risk to others.

147
Q

what is a radio-isotope? what is the most commonly used form and when is it used?

A

an unstable form of a chemical element which emits radiation when it decays and is another method of radiation delivery
The most commonly used form is radioactive iodine, I-131, which is used in the management of the most common forms of thyroid cancer

148
Q

how does iodine work as a radioisotope?

A

the iodine is preferentially taken up by and concentrated in any remaining thyroid tissue, normal or malignant, where it emits radiation as it undergoes radioactive decay, ablating the thyroid cells
few other tissues in the body take up iodine, and so it is selectively delivered radiation

149
Q

how long does it take for a patient to have radiation low enough to be safe to others after receiving iodine radioisotope therapy?

A

The patient will need to remain in the lead lined room until the level of radiation they are emitting is low enough to be safe to others, which is usually around four days

150
Q

what is stereotactic radiotherapy? why is it useful and when is it used?

A

when a tumour deposit is well defined and small eg brain metastases or lung primary, can deliver large dose of radiotherapy to target with a very small margin. Th biological dose (a concept that quantifies the chance of cell death within the target) can be increased by giving a small number of large fractions
used to treat oligometastatic disease since they generally have minimal side effects and yet produce high levels of local control
also possible in patients unfit for surgery

151
Q

what is arc therapy? what is the advantage?

A

allow the treatment unit to deliver treatment continuously as it moves through an arc around the patient
the MLC leaves can also be moved throughout this process
advantages: treatment delivery can be very fast (so more patients treated per day on individual machine), dose can effectively be ‘painted’ onto the target volume with much greater accuracy and with sparing of surrounding tissue

152
Q

what is intensity modulation?

A

refers to the moving of the MLC leaves during treatment delivery to build up dose in different parts of the treated volume - in Leeds volumetric modulated arc therapy is a type of intensity modulated radiotherapy used

153
Q

what is phase 1 of clinical trials?

A

aim is to determine toxicity (previously tested in vitro and in animals) and establish maximum tolerated dose (MTD)
drugs with no clinical activity seen are NOT rejected at this phase
dose escalation is performed, commencing at 10% of the dose /kg that is lethal in 10% of mice (‘LD10’). in general, 3 patients are treated at each dose level until side effects are seen and then 6 patients per group until maximum tolerated dose is reached
it is performed on patients with any tumour, in whom no conventional therapy is appropriate
patients must remain generally fit and have near-normal renal and hepatic function
trials are not randomized or comparative

154
Q

what is phase 2 of clinical trials?

A

once drug doses and scheduling have been defined from phase 1 trials, aim of phase 2 is to assess the particular anti-tumour activity of a new treatment in a range of different cancers
the radiological tumour shrinkage (‘response rate’) is the primary outcome measure for most phase 2 trials
it is not necessary to have a control arm and random allocation of treatment in all phase 2 trials, but can be useful

155
Q

what is phase 3 of clinical trials?

A

randomised trials comparing new with established treatments
primary endpoints assessed are usually length of life whatever the cause of death (overall survival), or length of life until the cancer grows (progression-free survival)
radiological shrinkage (response rate) and QOL are often secondary
if benefit of a new treatment is thought to be small, a large sample size will be required to achieve statistical significance
for this reason most of these trials are run in multiple centres and countries

156
Q

which criteria is used to define tumour responses radiologically? which definitions are used after treatment for target lesions?

A

RECIST (response evaluation criteria in solid tumours)
definitions:
complete response (disappearance of all target lesions)
partial response (at least a 30% decrease in the sum of longest diameters of target lesions taking as reference the baseline LD sum)
progression disease (at least a 20% increase in sum of LD of target lesions taking as reference the smallest sum LD recorded since treatment started or the appearance of one or more new lesions)
stable disease (neither sufficient shrinkage to qualify for PR, nor sufficient increase to qualify for PD, taking as references the smallest sum LD since the treatment started

157
Q

what do the following terms mean?
overall survival
disease free survival
progression free survival

A

overall survival: the length of time between entry to the trial and death from whatever causes
disease free survival: the time between entry into the trial and recurrence of the tumour or death from other causes
progression free survival: time between entry into the trial and disease progression or recurrence

158
Q

which toxicity in clinical trials, on the scale 1 to 4, which is most toxic?

A

grade 1 = least toxic
grade 4 = most toxic

159
Q

what do the following trials involve?
basket trials
umbrella trials
platform trials

A

“Basket trials” - where tumours harbouring a particular genetic mutation (of whatever primary site) are tested with a new drug that targets that mutation.
“Umbrella trials” - where patients with a particular type of cancer (e.g. non-small cell lung cancer) are all tested to establish the genetics of their tumours and then offered different experimental treatments depending on the results.
“Platform trials” - where multiple experimental arms are compared with a single control arm to reduce costs and increase the number of patients able to receive novel therapies.

160
Q

What would you prescribe for a 73 y.o. man with advanced renal cancer and multiple lung metastases who is complaining of progressive breathlessness?

A

oramorph PO

161
Q

what do the different scores for performance status mean?

A

0 = normal
1 = symptomatic and ambulatory cares for self
2 = ambulatory >50% time
3 = ambulatory <50% time nursing care required
4 = bedridden

162
Q

A 66 y.o. man presented with a change in bowel habit. Subsequent investigations show a right-sided colonic mass but no metastatic disease. He undergoes a colectomy and histology confirms a Duke’s C adenocarcinoma (indicates LN involvement). What treatment should he be offered next?
Adjuvant chemotherapy
Radical radiotherapy
Surgical excision
Surveillance
Systemic hormone therapy

A

adjuvant chemotherapy

163
Q

A 77 y.o. lady presents to her GP with hip pain. X-ray shows mixed sclerotic/lytic lesion in the iliac blade and subsequent imaging and biopsy confirm small primary HER2- ER+ breast cancer. Which is the most appropriate initial treatment approach?
Palliative chemotherapy
Palliative radiotherapy
Palliative systemic hormone treatment
Surgical excision
Symptom management alone

A

Palliative systemic hormone treatment
Hormone therapy appropriate - chemo may be considered if liver involvement. Bisphosphonates may be added for prevention of skeletal related events

164
Q

A 64 y.o. woman with hyperthyroidism and alcohol dependence complains of episodes of palpitations associated with breathlessness. She is a non-smoker and is otherwise well. On examination her pulse is 88bpm and regular, heart sounds normal, chest clear and no ankle oedema.
Which is the most appropriate diagnosis?
Anaemia
Anxiety
COPD
Heart failure
Paroxysmal AF

A

Paroxysmal AF
Thyrotoxicosis and alcohol are RFs for AF. The episodic nature of her symptoms suggests paroxysmal AF depsite a normal HR on exam

165
Q

What is the first line treatment for animal bites?

A

Co-amoxiclav

166
Q

What is the 2nd line treatment of skin infections given to patients who are allergic to penicillin?

A

erythromycin

167
Q

A 69 y.o. man with myeloma is admitted to the oncology ward with constant nausea. He has vomited small amounts of bile stained fluid for the last 3 days and feels thirsty.
Which anti-emetic do you prescribe?

A

Haloperidol
The patient most likely has a chemical or ‘ toxic’ cause for his vomiting (stimulation of the chemoreceptor trigger zone) probably hypercalcaemia and therefore the most suitable FIRST line antiemetic would be haloperidol
Haloperidol is usually drug of choice in renal impairment (likely with this story)

168
Q

A 71 y.o. woman with advanced lung cancer complains of passing hard stool. She is opening her bowels once a day as previously but this is now painful. What is the most suitable laxative to prescribe?
Bisacodyl suppositories
Co-danthramer suspension
Docusate capsules
Fybogel sachets
Senna tablets

A

Docusate capsules
The patient is having difficulty passing hard stool but has no change in the frequency of opening her bowels. She therefore needs a laxative that will soften the stool. Docusate is the only laxative in the list with only softening action

169
Q

A 63 y.o. man undergoing treatment for prostate cancer complains his left leg ‘gave way’ yesterday and feels odd. He denies any back pain. Which is the most appropriate investigation?
CT scan brain
Isotope bone scan
MRI spine
PSA
X-ray thoracic spine

A

MRI spine
symptoms are highly suggestive of a neurological problem. Spinal cord compression is an oncological emergency and is best excluded or characterised if present by MRI, which can also determine whether surgery, radiotherapy or supportive care are best management. It is possible to have cord compression without back pain, Brain mets are very uncommon in prostate cancer

170
Q

An 81 y.o. woman presents to her GP with gradual onset of breathlessness on exertion. She has a history of angina and hypertension. On exam, she has a systolic murmur, fine crackles at both lung bases and pitting oedema of the ankles. Her ECG shows left ventricular hypertrophy. What are the most appropriate investigations to perform?
CXR and coronary angiogram
CXR, Echo and bloods for BNP
Exercise tolerance test
Spirometry and bloods for FBC, U&E
24 hour ECG and echocardiogram

A

CXR, Echo and bloods for BNP
The patients appears to have heart failure as a result of ischaemic heart disease and hypertension. Her ECG shows LVH and she may also have some valvular disease. NICE guidelines for Heart Failure 2010 suggest BNP and Echo in all patients suspected of having heart failure. A CXR would also be helpful.

171
Q

A 41 y.o. woman presents to her GP with 3 weeks history of dysphagia. She has no weight loss or abdominal pain but has vomited on several occasions. She is HIV positive and is on antiretroviral treatment but has no other recent medical history. Which is the most appropriate investigation to arrange?
Urgent bloods for FBC, HIV viral and CD4 count
Urgent CXR
Urgent direct access CT scan
Urgent direct access US
Urgent direct access upper GI endoscopy

A

Urgent direct access upper GI endoscopy
NICE guidance offer urgent direct access upper GI endoscopy (to be performed within 2 weeks) to access for oesophageal cancer or stomach cancer in people:
with dysphagia
55 and over with weight loss and upper abdominal pain
or reflux or dyspepsia
The HIV is a red herring - it should not change your investigation

172
Q

A 69 y.o. man with metastatic mesothelioma was dying at home but had a fall and the paramedic ambulance brought him to the Emergency Department. He did not sustain any injuries but died on the ward the following day. What is the most appropriate cause of death to write on the medical cause of death certificate?
1a fall
1a metastatic mesothelioma
1a fall, 2 metastatic mesothelioma
1a fall 1b frailty 1c metastatic mesothelioma
Do not issue a certificate and refer to HM coroner

A

Patient had an occupational-related illness therefore must be discussed with HM coroner who may give permission for death certificate to be issued (as per D)

173
Q

A 60 y.o. man visits the practice nurse to discuss lifestyle modification after a friend died of cancer. The patient’s BMI is 35. Which cancer is a man who is obese at more risk of?
Bladder
Colon
Lung
Melanoma
Prostate

A

Colon cancer
Cancer Research Uk estimates obesity is one of the most important causes of colon cancer and accounts for 11-14% of cases

174
Q

You see a 65 y.o. woman in the surgery, on examining her pulse you notice she is in AF at a rate of 110bpm she is asymptomatic, you arrange an ECG which confirms AF. She has an active lifestyle but has asthma for which she takes salbutamol and an inhaled corticosteroid/long acting beta agonist. She takes no other medications and has no other PMH. You calculate her CHAD2DS2VASc score as 2. Which is the most appropriate management?
Beta blocker and aspirin
Calcium channel blocker
Calcium channel blocker and oral anticoagulant
Digoxin
Refer for cardioversion

A

Calcium channel blocker and anticoagulant
Beta blockers are contraindicated in patients with asthma
NICE no longer recommend aspirin monotherapy for stroke prevention in AF
Rhythm control not appropriate as you don’t know how long she has had AF and there is no known underlying cause

175
Q

A 70 y.o. woman complains that soon after waking she had a fleeting loss of vision in her left eye followed by a change in sensation affecting the right side of her body. Neurological exam later that day shows no abnormalities. Which is the most likely diagnosis?
Acute anxiety
Bell’s palsy
Migraine
Multiple sclerosis
Transient ischaemic attack

A

Transient ischaemic attack
sudden onset of symptoms. Amaurosis fugax and sensory change which lasts less than 24 hours

176
Q

A 73 y.o. man with prostate cancer and multiple bone and lymph node metastases is admitted to MAU with increasing fatigue and swollen ankles. He is currently taking paracetamol 1mg QDS, oxycodone 20mg BD and dexamethasone 0.5mg daily.
His blood tests on admission show: sodium 141 (137-144), potassium 5.9 (3.5-4.9), urea 25 (2.5-7) and creatinine 487 (60-110)
What is the most likely cause of his renal failure?
dehydration
hypercalcaemia
medication
obstructive uropathy
polycystic kidney disease

A

Obstructive uropathy
In dehydration urea and sodium likely to be higher and clinical story to match
hypercalcaemia unusual in prostate cancer (sclerotic not lytic mets) and creatinine usually rises to est 200 only
medication only likely if on ACE/NSAIDs/diuretics
Obstructive uropathy very common in prostate cancer: prostate and retroperitoneal LN obstruct ureters

177
Q

A 10 y.o. boy with asthma comes to surgery for his routine review. He is using his salbutamol inhaler every day about 2 or 3 times but more when he is playing football. He wakes up coughing most nights and occasionally has to sit out of PE due to wheezing. Which is the most appropriate additional treatment?
Antimuscarinic eg ipratropium bromide
Leukotriene receptor antagonist eg montelukast
Oral theophylline
Regular long-acting beta2 agonist eg salmeterol
Regular standard dose inhaled corticosteroid eg beclometasone

A

Regular standard dose inhaled corticosteroid
the patient is not controlled on step 1 of the BTS (british thoracic society) guidelines so must move up to step 2 which involves adding a regular steroid inhaler
Long acting beta 2 agonists are step 3 and should only be started in patients already on regular inhaled steroid preventer therapy. What would you add next? NICE = leukotriene receptor antagonist LTRA eg montelukast (cheaper but less effective) SIGN/BTS = long acting B2 agonist inhaler LABA eg salmeterol

178
Q

An 81 y.o. lady has her annual review for hypertension. Her BP is 100/60 and U&Es show the following: sodium 128 (NR 135-145), potassium 4.5 (NR 3.5-5), urea 7.0 (NR 3.0-8.3), creatinine (NR 44-133). Which of her medications would you stop?
Amlodipine
Atenolol
Bendroflumethiazide
Paracetamol
Ramipril

A

Bendroflumethiazide
thiazide diuretics are a common cause of hyponatraemia in the elderly

179
Q

What is a normal spirometry?

A

FEV1 and FVC greater than 80% of predicted and FEV1/FVC greater than 70% indicate normal spirometry
restrictive = FVC reduced, FEV1 reduced <80%, ratio normal
Obstructive = FVC normal, FEV1 reduced <80%, ratio reduced

180
Q

what are the side effects and signs of opioid toxicity?

A

side effects: constipation, nausea and vomiting, drowsiness (usually subsides after 48-72 hours)
toxicity: confusion, hallucinations, myoclonus, pupillary constriction, respiratory depression in overdose

181
Q

what is the conversion of oral morphine to the following?
subcutaneous morphine
oral oxycodone
oral codeine
subcutaneous diamorphine

A

subcutaneous morphine: divide by 2
oral oxycodone: divide by 2
oral codeine: x10
subcutaneous diamorphine: divide by 3

182
Q

what characteristics of tumours is needed for screening?

A

tumours should ideally:
be curable when detected early in the majority of the patients
be relatively common
have a ling pre-invasive or non-metastatic stage
be able to be detected by relatively simple tests
be distinct from benign lesions

183
Q

what are the ideal characteristics of a screening test?

A

tests should ideally:
be able to detect cancer at an early enough stage to implement effective treatment
be sensitive (ie be able to distinguish tumours clearly and give a low false negative value) - not miss actual cases
be specific (ie would not give false positive results)
be well tolerated hence improving compliance
be easy to administer or perform
be inexpensive
be well publicised in order to ensure high uptake amongst the target population

184
Q

what are the potential advantages and disadvantages of screeening?

A

advantages:
reduction of mortality by detecting early disease that is curable
less radical treatment hence reducing morbidity
saving on health service resources by increased cure rates
reassurance given by a negative tes

disadvantages:
increased length of anxiety and morbidity if no effective intervention is possible
the over-investigation of false positive cases with associated morbidity
over-treatment of borderline cases that do not require treatment
false reassurance from a false negative result
possible harmful effects of the screening test
cost of screening a large population

185
Q

what are the screening programmes in the UK?

A

cervical cancer
breast cancer
colorectal cancer

186
Q

what are the ages at which the following are screened?
cervical cancer
breast cancer
colorectal cancer

A

cervical: all women aged between 25 and 64; women aged 25-49 invited for cervical smears every 3 years, after that invited every 5 years
breast: regular mammography offered to all women between ages of 50 and 70 every 3 years
colorectal: men and women offered bowel screening using faecal occult blood every 2 years from 60 to 69 - this is being extended to 74

187
Q

What is advanced care planning?

A

a voluntary process of discussion and review to help an individual who has capacity to anticipate how their condition may affect them in the future and, if they wish, set on record choices about their care and treatment
outcomes include:
advance statement of wishes to inform subsequent best interest judgements
advance decisions to refuse treatment which are legally binding if valid and applicable
appointment of lasting powers of attorney for health and welfare and/or property and affairs

188
Q

what are the 3 causes of pain in cancer patients?

A

the disease itself eg bone invasion
the treatment eg radiotherapy induced oesophagitis
a concurrent disease eg osteoarthritis

189
Q

what are the features of bone pain in cancer patients and what is the treatment?

A

features: either a dull ache over a large area or well localised tenderness over the bone. Often worse on weight bearing or with movement
treatment: NSAIDs eg diclofenac 50mg tds, radiotherapy and bisphosphonates eg pamidronate infusion

190
Q

what are the features and treatment of visceral pain in cancer patients?

A

features: dull, deep-seated, poorly localised pain. There may be tenderness over a particular organ eg liver. Some visceral pain is spasmodic such as bladder spasm or bowel colic.
treatment: for constant dull visceral pains follow the analgesic ladder. Pain caused by visceral stretch such as liver capsule pain can be treated by NSAIDs or corticosteroids to reduce inflammation. Colic pain responds to anticholinergic drugs such as subcutaneous hyoscine butylbromide for bowel colic and oral oxybutynin for bladder spasm

191
Q

what are the features and treatment of headache due to raised in cancer patients?

A

features: dull, oppressive pain usually worse on waking, coughing, sneezing and may be associated with nausea and vomiting
treatment: corticosteroids to reduce oedema eg 16mg po dexamethasone daily, reduce to lowest effective dose, NSAIDs and paracetamol

192
Q

what are the features and treatment of neuropathic pain in cancer patients?

A

features: pain in an area of abnormal sensation. It may be localised to specific dermatomes or over a wider, less defined area. There may be altered sensation in the area such as numbness or hyperaesthesia and autonomic changes such as pallor or sweating. the patient may describe the pain as ‘pins and needles’ or burning
treatment: antidepressants (amitriptyline 10-75mg nocte) and anticonvulsants (gabapentin 100-1200mg tds, pregabalin 25-300mg bd). Compression of a nerve may be helped by corticosteroids

193
Q

what are the 3 stages of the analgesic ladder?

A

step 1: paracetamol
step 2: weak opioid eg codeine plus paracetamol eg co-codamol
step 3: strong opioids eg morphine, diamorphine
NSAIDs may be used at any step; other adjuvant drugs include antiepileptics, antidepressants, corticosteroids

194
Q

what are the 3 strengths of co-codamol? what should the dose be for progressing from step 1 of the analgesic ladder?

A

co-codamol come as either 8mg codeine and 500mg paracetamol (weak), 15mg codeine and 500mg paracetamol, or 30mg codeine and 500mg paracetamol (strong)
generally when progressing from step 1 the preparation with codeine 30mg should be prescribed

195
Q

what are the side effects of strong opioids?

A

constipation - always prescribe a laxative eg co-danthramer
N&V (usually settles within a few days) - prescribe a prn antiemetic eg haloperidol, and consider a regular antiemetic if patient is already nauseates or has had sickness in the past
drowsiness when starting or changing dose (usually improves within 48 hours)
confusion and visual hallucinations are rare if correct dose given - check dose and renal function and consider an alternative option if confusion persists
respiratory depression is rare
psychological dependence and addiction are common fears among patients but are rarely a problem in clinic practice

196
Q

what is the usual starting dose of MST after co-codamol?

A

if patient has been on max strength of co-codamol then MST 20mg bd is usually appropriate

197
Q

how long do fentanyl transdermal patches last?

A

duration of action of 72 hours

198
Q

which patients are suitable to have fentanyl transdermal patches?

A

mainly suitable for patients with severe chronic pain already stabilised on other opioids

199
Q

what is xerostomia?

A

dry mouth

200
Q

what is the treatment of oral thrush (candidiasis)?

A

treat with systemic antifungals - fluconazole 50mg od 7 days - or topical agents - nystatin 1ml qds 7 days

201
Q

what are the features and treatment of gastric stasis/irritation caused N&V?

A

features: early satiety, epigastric fullness, hiccups, heartburn, often minimal nausea between vomits. May be caused by tumour, hepatomegaly, ascites (‘squashed stomach’) and dysmotility (drugs, autonomic failure)
treatment: metoclopramide 10-20mg po/sc 30 mins before meals or 30-60mg sc over 24 hours. Stop any causative drugs if possible, consider PPI if gastric irritation

202
Q

what are the features and treatment of toxic causes of N&V?

A

features: persistent or intermittent nausea, small vomits, ‘possets’, and retching. May be caused by drugs (opioids, digoxin, antiepileptics), hypercalcaemi, uraemia and infections (UTI, pneumonia)
treatment: haloperidol 1.5-5mg po/sc nocte

203
Q

what are the features and treatment of cerebral causes of N&V?

A

features: early morning headache, vomiting, may be little nausea. Associated neurological symptoms/signs
treatment: dexamethasone 8-16mg po od (reduced if possible) plus cyclizine 50mg tds po/sc or 150mg sc/24 hours

204
Q

what are the features and treatment of anxiety/anticipatory N&V?

A

features: may have specific precipitants such as certain situations, overly anxious or depressed
treatment: benzodiazepines, CBT, complementary therapies

205
Q

what is the treatment of indeterminate N&V?

A

levopromazine 6.25-12.5mg nocte po/sc

206
Q

what are the features and treatment of vestibular causes of N&V?

A

features: may be associated with movement, hearing loss, vertigo or tinnitus
treatment: consider cyclizine, hyoscine or cinnarizine

207
Q

what are the 3 classes of laxatives? give examples

A

bulk forming - fybogel (rarely appropriate in palliative care)
stool softeners - lactulose and sodium docusate, movicol (predominantly a softener but may also stimulate bowel motions, eac sachet requires 125mls of liquid to be swallowed)
stimulants - senna and dantron - avoid if patient has colic

208
Q

what is a down-side to lactulose?

A

may cause significant bloating and flatulence

209
Q

what are the laxatives of choice for opioid induced constipation?

A

co-danthrusate
co-danthramer
movicol

210
Q

what should you do if you have given laxatives but the patient still hasn’t passed bowels in 3 days?

A

consider rectal exam and the use of suppositories and enemas

211
Q

which cancers usually get intestinal obstruction?

A

ovarian and bowel cancer

212
Q

what are the symptoms of intestinal obstruction in cancer patients?

A

N&V
colicky pain
abdo distension
dull aching pain
diarrhoea and/or constipation

213
Q

which medication are given to cancer patients who have intestinal obstruction?

A

combination of antiemetics, analgesics, antisposmodics
if colic is a feature, stimulant laxatives and prokinetic drugs (metoclopramide) should be stopped and antispasmodics prescribes (hyoscine butylbromide)
dexamethasone and octreotide may also be used

214
Q

what are 3 possible causes of dyspnoae in cancer patients? give treatment for each

A
  1. asthma - bronchodilators
  2. pulmonary oedema - diuretics, diamorphine
  3. pulmonary embolism - anticoagulants eg tinzaparin
215
Q

what are 4 possible causes of dyspnoea arising over several days in cancer patients? give treatments for each

A
  1. exacerbation of COPD - antibiotics, bronchodilators
  2. pneumonia - antibiotics, physiotherapy
  3. bronchial obstruction by tumour - dexamethasone, stents or laser
  4. superior vena cava obstruction - dexamethasone, urgent stenting
216
Q

what are 5 possible causes of more gradual onset of dyspnoea? give treatments for each

A
  1. congestive cardiac failure - diuretics, digoxin, ACE inhibitors
  2. anaemia - tranfusion?
  3. pleural effusion - pleural aspiration/pleurodesis
  4. ascites - paracentesis if appropriate
  5. lymphangitis carcinomatosis - trial dexamethasone
217
Q

what is the non-pharmacological treatment of dyspnoea (if cause cannot be reversed or is inadequately reversed)?

A

discussion of fears and explanation with patient, family and staff
modification of lifestyle, breathing retraining and relaxation
oxygen may help acute dyspnoea, but palliative patients will not benefit from ocygen therapy. A fan directed onto the face often helps

218
Q

what are the pharmacological treatments of dyspnoea (if cause cannot be reversed or is inadequately reversed)?

A

opioids: decrease resipratory effort and therefore breathlessness eg oramorph 2.5mg four hourly if opioid naive - gradually titrate up the dose
benzodiazepines: lorazepam (0.5mg-1mg sub-lingual) may help anxiety associated breathlessness or give relief during panic attacks
midazolam (2.5mg-5mg subcut) may benefit patients who cannot tolerate oral/sub-lingual route

219
Q

what is the management of cough in cancer patients?

A

if patient having difficulty expectorating, a trial of saline nebulisers may be helpful
if cough is dry and irritating then simple linctus may help
alternatively opioids are cough suppressants eg codeine linctus, normal release oral morphine

220
Q

which cancers commonly cause metastatic spinal cord compression?

A

breast, bronchus and prostate

221
Q

what is the pathophysiology of metastatic spinal cord compression?

A

it is caused by tumour or metastass in the vertebral body or paraspinal region pressing on the spinal cord. 2/3 occur in the thoracic region and the remainder in the cervical or lumbar sppine

222
Q

what are the complications of metastatic spinal cord compression?

A

can even lead to paraparesis, paraplegia and incontinence

223
Q

what are the symptoms of metastatic spinal cord compression?

A

back pain or nerve root pain - either unilateral or bilateral, which may be aggravated by movement, coughing or lying flat. Pain may precede other symptoms or may be absent in some patients
motor weakness - may be rapid or slow in onset, and can be subtle in the early stages; descriptions of perceived changes in strength are important
subjective sensory disturbance - often precedes objective physical signs eg ‘it feels like I am walking on cotton wool’
bladder/bowel dysfunction - generally occurs late, urinary retention often develops insidiously

224
Q

what are the signs of metastatic spinal cord compression?

A

weakness/paraparesis/paraplegia
changes in sensation occur below the level of compression; may be asymmetrical and may not necessarily be complete
reflexes are usually increased below the level of the lesion
clonus and painless bladder distension may be present

225
Q

what are the investigations of metastatic spinal cord compression?

A

whole spine MRI!

226
Q

what is the management of metastatic spinal cord compression?

A

corticosteroids (dexamethasone 16mg) should be commenced if there is a clinical suspicion of cord compression, pending definitive investigations. Subsequent treatment may involve surgery, radiotherapy or chemotherapy, either alone or in combination.
Surgery is usually favoured in situations of mechanical collapse of a vertebral body (where radiotherapy will rarely reverse the situation) but is less likely to be used if there is extensive disease elsewhere.
Management is usually coordinated by the on call oncology service, who will liaise with the neurosurgeons as required.
Management is time critical. Outcomes correlate with the level of function at the time of treatment. Complete paralysis, that has been present for several days, is almost never reversible.

227
Q

what is superior vena cava obstruction?

A

an extrinsic compression, thrombosis, or invasion of the wall of the superior vena cava

228
Q

what commonly causes superior vena cava obstruction?

A

most commonly caused by extensive lymphadenopathy in the upper mediastinum (often in patients with lung cancer or lymphoma), but can occur with any solid tumour
rarer causes include germ cell, breast or oesophageal cancer

229
Q

how is superior vena cava obstruction diagnosed and is it serious?

A

essentially a clinical diagnosis
should be regarded as an emergency since the patient’s condition may deteriorate rapidly

230
Q

what are the symptoms of superior vena cava obstruction?

A
  • Headache or a “feeling of fullness” in the head
  • Facial swelling
  • Dyspnoea (often worse on lying flat)
  • Cough
  • Hoarse voice
231
Q

what are the signs of superior vena cava obstruction?

A
  • Facial / upper limb oedema
  • Prominent blood vessels on the neck, trunk and arms
  • Cyanosis
232
Q

what is the management of superior vena cava obstructin?

A

commence on corticosteroids (dexamethasone 16mg daily) which can sometimes produce short-term relief of symptoms
urgent vascular stenting is the usual treatment of choice - often followed by radiotherapy or chemotherapy depending on the primary tumour type

233
Q

what investigations are important if a patient presented with superior vena cava obstruction as a first sign of caancer?

A

biopsy is important
if a germ cell tumour is possible then tumour markers may be useful eg AFP, BhCG, LDH

234
Q

when is hypercalcaemia commonly seen in cancer patients?

A

commonly seen in breast cancer, lung cancer, squamous cell carcinomas and myeloma

235
Q

what are the symptoms of hypercalcaemia?

A

symptoms may develop insidiously and hypercalcaemia may be missed as a cause of some commonly occurring symptoms
early: lethargy, malaise, anorexia, polyuria, thirst, nausea, vomiting, constipation
late: confusion, drowsiness, fits, coma

236
Q

what is the investigation of hypercalcaemia in cancer patients?

A

serum calcium corrected for serum albumin

237
Q

what is the management of hypercalcaemia?

A

rehydration using normal saline followed by IV bisphosphonates such as pamidronate or zoledronic acid
about 70% of patients respond
maximum response is seen after 6-11 days with an average duration of response of 3-4 weeks

238
Q

which cancers usually cause major haemorrhage?

A

eg a head and neck cancer eroding into a major vessel

239
Q

what is the management of major haemorrhage in cancer patients?

A

keep green towels close by to absorb and reduce the visual impact of blood loss
if time allows, IM into the deltoid muscle or SC midazolam as a sedative and amnesic can be given

240
Q

which symptoms and signs may indicate that a palliative patient has a short prognosis?

A

profound weakness
confined to bed for most of the day
drowsy for extended periods
disorientated
severely limited attention span
losing interest in food and drink
too weak to swallow medication

241
Q

what are the nursing needs of palliative patients?

A

treat dry mouth with good regular mouth care
assess immobility and pressure areas - special beds or mattresses may be needed
consider catheter, convene, or pads for incontinence
consider bowel care only if constipation is causing discomfort or agitation
fast track/continuing care funding will enable patients. to die at home or in a care home

242
Q

what are the possible benefits of withdrawing artificial hydration/nutrition in a palliative person at the end of their life?

A

less vomiting and incontinence
reduction in barriers between patient and family/carers
prevention of painful venepuncture

243
Q

which medications are commonly stopped when a palliative patient is no longer able to swallow?

A

vitamins/iron
hormones
anticoagulants
corticosteroids
antibiotics
antidepressants
cardiovascular drugs
anticonvulsants used for pain

244
Q

what are the 4 medications that patients should be prescribed when in the end of their life?

A

analgesic, antiemetic, anti-secretory and anxiolytic

245
Q

what is the ‘death rattle’ and how is it treated?

A

this is rattling noise produced by the movement of secretions in the upper airways, generally in patients who are too weak to expectorate effectively
patients are generally not distressed by the noise but relatives and carers may find this distressing
treatment: reposition the patient, antisecretory drugs eg hyoscine butylbromide (buscopan) or hyoscine hydrobromide (which may cause paradoxical agitation) may be needed

246
Q

what is the most commonly used syringe driver?

A

the McKinley T34

247
Q

what are the indications for a syringe driver?

A
  • Inability to swallow drugs due to reduced conscious level, often in the last few days of life
  • Persistent nausea and vomiting
  • Intestinal obstruction
  • Malabsorption of drugs
  • Dysphagia
248
Q

is inadequate pain control an indication for syringe driver use?

A

it is not an indication unless there is reason to believe oral analgesics are not being absorbed

249
Q

what is a side effect of cyclizine in a syringe driver?

A

may cause skin irritation

250
Q

why is hyoscine butylbromide preferred over hyoscine hydrobromide as an antisecretory and antispasmodic?

A

hyoscine butylbromide (buscopan) does NOT cross the blood brain barrier
hyoscine hydrobromide may cause sedation or agitation

251
Q

which drugs are unsuitable for SC administration?

A

as they are too irritant:
diazepam, chlorpromazine, prochlorperazine

252
Q

which are the big 4 cancers?

A

breast, lung, prostate and colorectal

253
Q

what is the incidence of breast cancer in the UK?

A

the most common cancer in women, accounting for 15% of all new cases of female cancer
in UK: 55000 new cases diagnosed each year; approx 11000 women die from the disease
in UK: 1 in 8 women and 1 in 870 men will develop breast cancer

254
Q

what are the risk factors of breast cancer?

A

increasing age
increasing periods of oestrogen exposure: late childbearing, nulliparity, early menarche, late menopause, obesity
COCP and some types of HRT
obesity, alcohol
ionising radiation
FHx (first degree relative, particularly premenopausal)
genetics: BRCA1 gene (breast and ovarian cancer), BRCA2 gene (linked to early onset breast cancer and male breast cancer, but not with ovarian cancer)

255
Q

what is the histology of breast cancer?

A

Infiltrating or invasive ductal carcinoma is the most common cell type, comprising 70-80% of all cases. Lobular carcinoma, comprising approximately 10% of cases, is characterized by a higher incidence of multicentric tumours within the same or opposite breast. Other less common histological types include medullary, colloid, comedo and papillary.
Tumours are graded according to differentiation with grade 1 lesions being well differentiated and grade 3 lesions being poorly differentiated.

256
Q

what is the presentation of breast cancer?

A

most patients present with a breast mass
less common presentations include nipple discharge, regional lymphadenopathy (axillary or supraclavicular nodes) or symptoms of metastatic disease

257
Q

how are patients diagnosed with breast cancer?

A

referred to breast clinic either by GP (2ww) or through NHS breast screening programme
triple assessment:
1. clinical assessment (exam and history)
2. bilateral mammography - to detect multicentric tumours or synchronous primaries in the opposite breast
3. targeted US and biopsy of symptomatic breast area or area of mammographic abnormality. patients will also have. anUS of axillae with/without biopsy of suspicious nodes
cytological diagnosis should be confirmed by fine needle aspiration cytology (FNAC), needle biopsy, incisional or excisional biopsy

258
Q

when is MRI performed in breast cancer?

A

if there is discrepancy between clinical exam, mammogram and US findings, if breast density preludes accurate mammogram assessment and if the histology is lobular (as there is a higher frequency of multicentric disease)

259
Q

what do the following staging of breast cancer mean?
T0, Tis, T1, T2, T3, T4

A

T0 = no primary tumour
Tis = in situ disease, non-invasive
T1 = invasive tumour less than 2cm
T2 = primary tumour between 2 and 5cm
T3 = primary tumour greater than 5cm
T4 = skin involvement

260
Q

what do the following staging of breast cancer mean?
N0, N1, N2, N3
M0, M1

A

N0 = no lymph nodes
N1 = mobile axillary nodes
N2 = fixed axillary nodes
N3 = internal mammary nodes
M0 = no metastases
M1 = distant metastases

261
Q

what is the stage of the following breast cancers?
1. T or N > stage II, M0
2. Tis, N0, M0
3. T1, N0, M0
4. Any T, Any N, M1
5. T2/3, N0, M0 or T0/1/2, N1, M0

A
  1. stage 4
  2. stage 0
  3. stage 1
  4. stage 5
  5. stage 2
262
Q

what is the management of localised breast cancer?

A

Each patient’s management plan should be discussed at a multidisciplinary meeting (MDT).
The standard treatment would be surgery first. However for a certain subset of patients neoadjuvant chemotherapy is considered (chemotherapy before surgery) such as when
* Initial surgery is not possible due to the size of the tumour
* To allow for breast conservation
* Her2 positive or triple negative breast cancer (ER, PR and Her 2 negative) as high response rates are possible

263
Q

what are the surgical options in breast cancer?

A

mastectomy or conservative surgery eg wide local excision, with postoperative radiotherapy

264
Q

how does the assessment of axillary lymph nodes. inbreast cancer determine the management?

A

If the initial assessment shows evidence of metastatic involvement of the lymph nodes then patients will have an axillary clearance.
If there is no evidence of metastatic involvement of the lymph nodes, patients will have a sentinel node biopsy (SNB). The sentinel nodes are the first few lymph nodes into which a tumour drains. Sentinel node biopsy involves injecting a tracer material that helps the surgeon locate the sentinel nodes during surgery. The sentinel nodes are removed and analyzed in a laboratory. If they are positive then patients will go on to have an axillary clearance or radiotherapy to the axillae.

265
Q

which factors determine whether breast cancer patients receive adjuvant systemic therapy with endocrine treatment or with cytotoxic chemotherapy?

A
  • Hormone receptor status [oestrogen receptor (ER) status]
  • HER-2 receptor status
  • Menopausal status
  • Tumour size and grade
  • Nodal involvement
  • Performance status
266
Q

what is trastuzamab and when is it used?

A

Therapy with Trastuzamab (Herceptin®) is effective in metastatic and localised disease where the cancer over-expresses the target epithelial growth factor receptor (HER-2) eg breast cancer. In the adjuvant setting it is given for 12 months. It can affect cardiac function and therefore patients will need to have regular cardiac monitoring (normally with a MUGA scan).

267
Q

which endocrine therapy is used in breast cancer?

A

tamoxifen - normally prescribed for premenopausal women who have tumours that are ER/PR positive (if no contraindications); 20mg/day reduces risk of recurrence and death in the adjuvant setting; increased thrombotic complications are reported and increased risk of endometrial cancer > normally given. for5 years
Aromatase Inhibitors. e.g. anastrazole, letrozole are demonstrated to have superior efficacy to tamoxifen in post-menopausal women with breast cancer. They have a different side effect profile including fewer vascular and malignant events, but more problems with osteoporosis.

268
Q

what do all breast cancer patients require following conservative surgery?

A

all patients require radiotherapy to the residual breast tissue to reduce chances of locoregional cancer recurrence
standard treatment is daily, monday to friday, for 3 weeks

269
Q

when can endocrine treatment be given as primary treatment in breast cancer?

A

can be used in ER/PR positive disease in slowly progressive disease, especially where there is no visceral involvement and the patient has minimal symptoms
median duration of response to a single endocrine therapy is 1-2 years

270
Q

higher response rate to endocrine therapy can be related to which factors in breast cancer?

A
  • The dominant site of disease (highest in women with disease in soft tissue, less in those with bone metastases and less again in those with visceral metastases). This may simply reflect ER status
  • An objective response to prior endocrine treatment.
  • Greater duration of previous disease free interval.
271
Q

when is ovarian ablation used in breast cancer?

A

in premenopausal women, endogenous ovarian oestrogen production may be stopped by ovarian ablation (surgically or radiotherapy induced) or by the use of luteinizing hormone releasing hormone (LHRH) agonists

272
Q

what is the 5 year survival of the following stages of breast cancer?
stage 1
stage 2
stage 3
stage 4

A

stage 1 = 95%
stage 2 = 80%
stage 3 = 60%
stage 4 = 25%

273
Q

what is the epidemiology of lung cancer in the UK?

A
  • Lung cancer is the 3rd most common cancer in the UK.
  • 1 in every 13 men and 1 in every 15 women are diagnosed with lung cancer in their lifetime
  • Lung cancer accounts for 22% of cancer related deaths in the UK
  • Only 10% of patients who are diagnosed with lung cancer live for 5 years or more.
274
Q

what are the risk factors of lung cancer?

A

genetics
smoking
increasing age
history of COPD
industrial exposure to asbestos, chromium, arsenic and iron oxide
exposure to radiation

275
Q

where do lung tumours usually arise from histologically?

A

Tumours usually arise from the epithelium of the large and medium sized bronchi (only rarely from the lung parenchyma itself).

276
Q

how much do small cell lung cancers make up lung cancers? what are the features of them? what are they associates with?

A

15%
These are highly aggressive tumours which grow rapidly. They have usually metastasised and become inoperable prior to diagnosis. They can be very responsive to chemotherapy initially, but often relapse quickly. Overall, prognosis is generally poor.
They can also be associated with paraneoplastic syndromes - most commonly causing SIADH, Cushing’s syndrome or Lambert Eaton Myesthaenic syndrome (LEMS).

277
Q

what percentage of lung cancer is non-small cell?

A

85%

278
Q

what are the types of non-small cell lunger cancer? give features for each

A

squamous cell carcinoma (42%): often found centrally close to the bronchi; can present with bronchial obstruction; closely linked to cigarette smoking; squamous cell cancers can also secrete PTH relate peptide (PTHrp) which can lead to malignancy-related hypercalcaemia
adenocarcinoma (39%): often peripheral, more frequent in women, non-smokers and patients with previous asbestos exposure; more commonly associated with activating mutations. in EGFR and ALK
large cell carcinomas (8%): less differentiated than other NSCLCs and tend to metastasise early
other types = carcinoid, mesothelioma, sarcoma and lymphoma

279
Q

what are the clinical features of lung cancer?

A

majority present with non-specific symptoms of the primary tumour eg cough, dyspnoea, haemoptysis, chest pain, or recurrent chest infections
lat presentation is frequent as most of these symptoms are common in smokers
Occasionally, tumours are identified incidentally on x-rays or scans performed for some other reason.

280
Q

how do the following lung cancers present?
apical tumours
medistinal disease

A

apical tumours may invade the brachial plexus or sympathetic chain producing Horner’s syndrome or pain in the distribution of the nerve routes involved (Pancoast’s syndrome). Recurrent laryngeal nerve palsy and superior vena cava obstruction may be associated with mediastinal disease

281
Q

which lung cancer more commonly presents with clubbing, and which is more commonly presents with sputum production?

A

clubbing is more frequent with squamous cell carcinoma. Sputum production may be excessive in bronchio-alveolar carcinoma

282
Q

what are the investigations of lung cancer?

A

CXR
CT chest and upper abdomen
PET scan - used in patients who are thought to have operable disease to check for distant metastases which may not be picked up on CT.
bronchoscopy
Trans-thoracic biopsy under radiological guidance is sometimes required for the biopsy of peripheral lung tumours, or distant metastatic disease e.g. liver metastases.
tumour markers - neuron specific enolase and lactate dehydrogenase (not routinely used)
pulmonary function tests
cardiopulmonary exercise testing - important for patients who are being considered for surgical resection to ensure they are fit enough to undergo an operation

283
Q

what is the management of small cell lung cancer?

A

SCLC is generally considered a systemic disease at presentation. As such, the vast majority of patients will receive palliative chemotherapy.
If the disease appears “limited stage” at diagnosis (i.e. encompassable within a high dose radiotherapy field) then radical radiotherapy may be given (either alongside or following) the chemotherapy.
If the disease is more extensive at presentation, then chemotherapy is the mainstay of treatment although consolidation thoracic radiotherapy may also be used in patients who have a good response to chemotherapy.
Prophylactic cranial irradiation (see below) is also used in patients with limited disease and in those with extensive disease who respond to chemotherapy.

284
Q

what is the difference in management between complications of small cell and non-small cell lung cancer?

A

SCLC is one of the most chemo-sensitive solid tumours. Chemotherapy is therefore the mainstay of SCLC management with responses occurring within days. Complications such as superior vena cava obstruction or spinal cord compression can be treated with chemotherapy rather than radiotherapy. This is in contrast to NSCLC.

285
Q

what are the 2 indications for radiotherapy in the management of small cell lung cancer?

A
  1. treatment of primary tumour
  2. prophylactic cranial irradiation - brain mets frequent in SCLC; can have significant toxicities eg memory impairment, functional deficit and dementia
  3. palliative
286
Q

is surgery appropriate for small cell lung cancer?

A

The early dissemination of SCLC means it should be considered a systemic disease at presentation. Surgical intervention is therefore inappropriate for the majority of cases.

287
Q

what is the prognosis of small cell lung cancer? what are the prognostic factors?

A

without treatment: poor, median survival of 2-4 months
treatment with systemic chemo: median survival 6-12 months
prognostic factors: extent of disease at presentation, number of metastatic sites, performance status, degree of weight loss and biochemical abnormalities (elevated LDH or low sodium or albumin)

288
Q

when is surgery used in non-small cell lung cancer? what is a definite contraindication?

A

stage 1 and 2 - surgical resection has a good prognosis and possible cure
mediastinal involvement is considered a contraindication to surgery

289
Q

when is radiotherapy used in non-small cell lung cancer?

A

In patients with early stage disease, who are not suitable for surgery, radical radiotherapy has often been used and produces 20% 5-year survival for patients with stage 1 or 2 disease. Continuous, hyperfractionated accelerated radiotherapy (CHART) is radiotherapy which is given three times a day for 12 consecutive days, and compared with conventional radiotherapy (often 20 to 30 once daily treatments), results in improved survival.
Giving radiotherapy at the same time as chemotherapy (concurrent chemo-radiotherapy) may also improve outcomes compared to giving chemotherapy first followed by radiotherapy (sequential chemo-radiotherapy). This is usually used in patients with Stage II-III dise
early NSCLC located more peripherally in the lungs is treated by stereotactic ablative body radiotherapy (SABR). This delivers a small number (≤5) of very large doses of radiotherapy to a small, highly conformal volume around the lung tumour.
Palliative radiotherapy is used frequently for symptomatic disease such as spinal cord compression, painful bone metastases or significant chest symptoms due to central disease (e.g. haemoptysis, cough).

290
Q

what are some examples of chemotherapy used in non-small cell lung cancer?

A

combination regimens are used, either Carboplatin and Gemcitabine or Carboplatin/Cisplatin and Pemetrexed. Docetaxel can be used as a 2nd line option

291
Q

when is targeted chemotherapy used in non-small cell lung cancer?

A

beneficial in some patients with advanced non-small cell lung cancer. Patients with adenocarcinoma are tested for mutations in EGFR and ALK at the time of diagnosis. If these are positive, tyrosine kinase inhibitors (TKIs) can be used (e.g Afatinib/Erlotinib/Gefitinib for EGFR mutations and Crizotinib for ALK mutations). The proportion of patients suitable for these treatments is < 10% overall but is higher in some ethnic groups and non-smokers. Patients on these targeted therapies can be treated with second line targeted therapies (Osimertinib/Ceritinib) rather than the standard chemotherapy described above.

292
Q

when is immunotherapy (pembrolizumab) used in non-small cell lung cancer?

A

recently been approved for patients with advanced NSCLC in patients with high PDL1 expression (a molecule involved in controlling the normal immune response). This can be used either before or after chemotherapy and a small percentage of patients can have a prolonged benefit with this.

293
Q

what is the prognosis and prognostic factors of non-ssmall cell lung cancer?

A

Without treatment, prognosis is likely to be short, and is usually measured between 3-6 months. Provided patients are fit enough to tolerate chemotherapy, this can extend average life expectancy by a number of months, possibly to just over a year. If patients are suitable for targeted treatment or immunotherapy, this can be substantially longer and prognosis may be around 2 years in some cases.

294
Q

what is the most common cancer in males?

A

prostate cancer

295
Q

what is the epidemiology of prostate cancer?

A

Prostate cancer is the commonest cancer in males, with more than 47000 new cases diagnosed in the UK in 2015. It is predominantly a disease of older men, with more than 50% occurring over the age of 75.

296
Q

what are risk factors for prostate cancer?

A

increasing age
higher in men from African and Afro-Caribbean backgrounds
Fhx, and men whose mothers were diagnosed with breast cancer
BRCA2 and pTEN gene mutations

297
Q

what is the histology of prostate cancer?

A

over 95% are adenocarcinomas, developing in glandular tissue in posterior or peripheral parts of the prostate
(benign prostatic hyperplasia more commonly arises in centre of gland)
histology defined by Gleason system

298
Q

what is the presentation of prostate cancer?

A

may be asymptomatic and diagnosed with rectal exam or PSA blood test
may present with lower urinary tract symptoms: poor stream, nocturia, dribbling and increased frequency, impotence is common
1 in 5 patients present with metastatic prostate cancer and may have bone complications such as anaemia, pain, pathological fracture, or spinal cord compression.
Typical rectal examination findings would include an enlarged, hard, craggy gland (or nodule). Ultimately there is obliteration of the median sulcus.

299
Q

what are the investigations of prostate cancer?

A

initial: DRE and serum PSA
if radical treatment appropriate then an MRI scan can visualise the prostate well and delineate any extra-capsular spread
if metastatic disease concerns then isotope radionucleotide bone scan
most patients will have US guided transrectal biopsy to confirm the diagnosis but where clinical suspicion is very high (e.g. PSA >100 with positive bone scan) this is not required

300
Q

What do the following stages of prostate cancer mean?
TX
T0
T1
T2
T3
T3a
T3b
T4

A

TX = primary tumour cannot be assessed
T0 = no evidence of primary tumour
T1 = clinically unapparent tumour not palpable nor visible by imaging
T2 = tumour confined within prostate
T3 = tumour extends through the prostate capsule
T3a = extracapsular extension (unilateral or bilateral)
T3b = tumour invades seminal vesicles
T4 = Tumour is fixed or invades adjacent structures other than seminal vesicles: bladder neck, external sphincter, rectum, levator muscles, and/or pelvic wall

301
Q

what do the following stage of prostate cancer mean?
N0
N1
M0
M1
M1a
M1b
M1c

A

N0 = no regional lymph node involvement
N1 = regional lymph node involvement
M0 = no distant metastases
M1 = distant metastases
M1a = non-regional lymph nodes(s)
M1b = bone(s)
M1c = other or multiple site(s) with or without bone disease

302
Q

what is the grading system of prostate cancer?

A

Gleason system scores tumours from 2 to 10 on the basis of histological patterns in the 2 most dominant areas of the tumour eg Gleason 4 + 3 means main area is 4 with the second area 3

303
Q

what is the general management of prostate cancer?

A

PSA, histological grade and stage can be used to stratify patients with local prostate cancer into risk groups and to counsel patients on management options. Treatment of locally advanced and metastatic patients is broadly palliative but can lead to durable symptomatic benefit and many patients live with their disease for years. . The most appropriate options are influenced by the patient’s age, fitness, existing medical issues and preferences

304
Q

when may observation be appropriate as management of prostate cancer?

A

In patients with asymptomatic disease, confined to the prostate, observation rather than active treatment may be appropriate, particularly where other conditions limit the length of survival that is expected. Radical treatment is unlikely to benefit men with life expectancy of less than ten years, some of whom will not require on-going surveillance. The optimum management of patients with an elevated PSA but no clinical evidence of prostate cancer is unknown

305
Q

when is surgery an appropriate management of prostate cancer?

A

Patients with localised disease (T2 disease or less) can be treated by radical prostatectomy with curative intent and this can be performed by perineal or retroperineal routes. Robotic laparoscopic surgery leads to reduced operating times, less blood loss and shorter hospital stays but, though mortality is low, prostatectomy can lead to temporary or lasting impotence and incontinence.
Palliative surgical techniques such as trans-urethral resections may be used to relieve prostatic symptoms or urinary obstruction in some men.

306
Q

when is radiotherapy used in the management of prostate cancer?

A

Radical radiotherapy may be used as an alternative to surgery in T1 and T2 tumours, where PSA is low, suggesting no occult metastases. It is also more appropriate for the control of more advanced local disease. Adjuvant radiotherapy may also be given following radical surgery if there is concern for residual disease.
Definitive radiotherapy should be delayed until at least six weeks following trans-urethral resection to prevent stricture formation.
Radiotherapy may be performed by external beam irradiation, by the interstitial implantation of radioisotopes (brachytherapy) or by a combination of these.
Side effects can include dysuria, rectal bleeding, diarrhoea, impotence and incontinence.
Palliative radiotherapy is used to palliate the primary tumour and/or to treat metastatic complications e.g. recurrent haematuria due to bleeding from prostate bed, bone pain from metastatic disease.

307
Q

how do LHRH agonists act as hormonal therapy for prostate cancer? give examples and what are the side effects?

A

eg leuprorelin, goserelin
Luteinizing-hormone releasing hormone (LHRH) interferes with the normal release of gonadotropins from the pituitary. This reduces the level of circulating testosterone to those following castration. LHRH agonists are given by monthly or 3 monthly subcutaneous or intra-muscular depot injections. Side effects include impotence, loss of libido and tumour flare. Tumour flare occurs on initiation of treatment (prior to the down regulation of gonadotropin) and is avoided by short-term concomitant anti-androgen therapy. Long-term consequences of medical castration include increased cardiac risk and osteoporosis.

308
Q

how do gonadortophin-releasing hormone antagonist act as hormone therapy for prostate cancer? give examples

A

leads to castrate levels of testosterone in 96% of patients within 3 days (without risk of tumour flare). Given by monthly subcutaneous injection it is indicated when tumour flare may lead to significant symptoms e.g. a patient presenting with metastatic spinal cord compression.

309
Q

how does oestrogen therapy treat prostate cancer? what are the side effects?

A

Oestrogens inhibit LHRH production from the hypothalamus but are rarely the best option due to side effects: impotence, loss of libido, gynaecomastia, myocardial infarction, stroke and pulmonary emboli.

310
Q

how do anti-androgens treat prostate cancer? give examples

A

e.g. bicalutamide, enzalutamide) compete with androgens for sites on the androgen receptor. Enzalutamide shows progression and survival benefit when combined with LHRH therapy in advanced prostate cancer.

311
Q

how is chemotherapy used to treat prostate cancer? give examples

A

Cytotoxic treatment with docetaxel (in combination with prednisolone) and cabazitaxel has been shown to improve quality of life and overall survival in patients with castrate-refractory metastatic disease. Recent data supports use of early docetaxel chemotherapy (+/- prednisolone) around the time of diagnosis of metastatic prostate cancer: six cycles of chemotherapy in fit men prolongs survival (median 60 months, ie an extra 15 months, HR 0.76, p=0.005) and has led to a change in practice.

312
Q

what is the prognosis of prostate cancer?

A

Survival is related to the extent of the disease.
Prostate cancer survival of men with low risk localised prostate cancer is excellent (99%) at 10 years whether they choose active surveillance, radiotherapy or surgery.
Prostate cancer 10 year survival (all UK patients) is around 84%.
Patients with metastatic disease have a median survival of 3.5 years. Pattern of disease (bone metastases), aggressive pathology and younger age are associated with a poorer prognosis.

313
Q

what is the epidemiology of colorectal cancer?

A

41000 new cases diagnosed every year in UK
4th most common malignancy after lung cancer - 10-15% of all malignancies, causes 16000 deaths each year
cancer. ofcolon is 1.5 times more common than rectal carcinoma

314
Q

what are the risk factors for colorectal cancer?

A

diet - rich in animal fats and meat and poor in fibre; common in Western countries
IBD - association with ulcerative colitis which is related to the extent of the bowel involvement and duration of inflammation
familial association - hereditary non-polyposis colon cancer (HNPCC), familial adenomatous polyposis (FAP) and Gardner’s syndrome; mutation sin APC gene (5q21-22 for FAP coli) and mutations in DNA mismatch repair genes (HNPCC)

315
Q

what is the stepwise process of progression from benign to invasive carcinoma in colorectal cancer? what is the pathophysiology?

A

APC mutations are associated with the development of benign adenomas whereas the progression to invasive carcinoma requires further mutations eg p53, DCC and RAS
these mutations may be caused by carcinogens present in faeces
in principle diets low in fibre prolong transit time and may prolong exposure to carcinogens
bile salts may act as carcinogens both directly and when degraded by bacteria present in the faeces

316
Q

what is the histology of colorectal cancer? where are they most commonly found?

A

40% of large bowel cancers occur in the rectum, 20% in the sigmoid colon, 6% in the caecum and the rest in the remaining colon
the histological types of cancer that occur are:
- epithelial: 90-95% adenocarcinoma (mucinous or signet ring). Rare others include squamous cell carcinoma and adenosquamous carcinoma.
- carcinoid
- gastrointestinal stromal tumour
- primary malignant lymphoma
colorectal cancer normally spreads by local invasion, lymphatic, venous and trans-coelomic spread within the peritoneal cavity

317
Q

what is the usual presentation of colorectal cancer?

A

presentation can be with classical features of altered bowel habit, weight loss, rectal bleeding and vague abdominal pain, although clinical features vary according to the site of the primary tumour and degree of spread
more occult tumours, typically of the right side of the colon and the caecum, can present with iron deficiency anaemia

318
Q

what are the investigations of colorectal cancer?

A

RECTAL EXAM IS ESSENTIAL
direct visualisation of the bowel with rigid sigmoidoscopy, flexible sigmooidoscopy and colonoscopy allows biopsy of suspicious lesions
double contrast barium enema is less useful as no histology is provided
CT provides staging and a useful evaluation of the bowel in some cases
CT colonoscopy combines CT scanning with insufflation of the whole colon with gas - this can help identify synchronous polyps within the bowel where colonoscopy is not possible, however it does not facilitate biopsy
other: tumour marker CEA (carcino-embryonic antigen) - although elevation of this protein is not diagnostic it can be useful to monitor

319
Q

what do the following stages of colorectal cancer mean?
TX
T0
T1
T2
T3
T4

A

TX - primary tumour cannot be assessed
T0 - no evidence of primary tumour
T1 - tumour invades submucosa
T2 - tumour invades muscularis propria
T3 - tumour extends through muscularis propria into peri-colic tissues
T4 - tumour invades visceral peritoneum or invades / adheres to adjacent organ or structure

320
Q

what do the following stages of colorectal cancer mean?
N0
N1
N2
MX
M0
M1
M1a
M1b
M1c

A

N0 - no regional lymph node involvement
N1 - involvement of 1-3 lymph nodes
N2 - involvement of 4 or more lymph nodes
MX - distant metastasis cannot be assessed (not evaluated by any modality)
M0 - no distant metastasis
M1 - distant metastasis
M1a - confined to one organ or site eg liver or lung, but not peritoneum
M1b - 2 or more sites (but not peritoneum)
M1c - peritoneal spread

321
Q

what is Duke’s staging used for and what do the stages A-D represent each?

A

A invasion into but not through the bowel wall
B invasion through the bowel wall but not into nodes
C lymph node involvement
D distant metastases

322
Q

when is surgery used as management for colorectal cancer?

A

Radical resection is the standard treatment for primary colorectal carcinoma because of the risk of unsuspected nodal metastases. Patients with early stage colorectal carcinoma are usually cured by surgical resection alone. The long-term results with rectal carcinoma are related to the initial surgical resection.
Surgery may also be indicated in patients with advanced disease. Resection of liver metastases in addition to the primary may be beneficial. Further resection of a local recurrence is associated with improved long-term survival.
Surgery or colonic stenting may be used in the palliative setting to manage or prevent an obstructing lesion.

323
Q

when is radiotherapy used as management for colorectal cancer?

A

predominantly used in treatment of rectal carcinomas
not commonly used in management of colon cancers due to the potential toxicity to adjacent organs and mobility of tumours
pre-operative (or rarely adjuvant) radiotherapy is indicated in high risk rectal carcinomas before (or rarely following) total resection
cases are selected on the basis of pelvic MRI staging scans
local recurrences can be palliated with radiotherapy
metastatic disease may also respond to palliative radiotherapy

324
Q

when is chemotherapy used as management for colorectal cancer?

A

adjuvant chemo for high-risk colorectal cancer eg Duke’s C carcinoma
5-FU is the most active agent in colorectal carcinoma with a response rate of approximately 25% - newer drugs such as Oxaliplatin and Irinotecan are now also in standard use

325
Q

what is the prognosis for each stage of Duke’s for colorectal cancer? also for age

A

5 year survival:
A - 80%
B - 50%
C - 15-40%
D - 5%
age below 40 is an adverse prognostic factor reflecting a biologically more aggressive tumour

326
Q

what is the screening for colorectal cancer?

A

faecal occult blood testing of average-risk populations (with follow up colonoscopy for positive cases)
given to people aged 60-74 every 2 years

327
Q

What is the difference between using the CHADVASC score and ORBIT score?

A

CHADVASC is used for the stroke risk of AF patients
ORBIT is used as a screening tool for the bleeding risk of AF patients and the need for anticoagulation