Oncology Flashcards

0
Q

What is the most common cause of cancer death in 20-34 year olds?

A

Melanoma.

Australia has highest incidence in the world.
Incidence doubled over last 20y.
4th most common cancer overall. 10%.

Stage IV confers a 10-20% 5yr survival rate.

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

What is the 5 leading causes of cancer death in Australia?

A

Lung > bowel > PROSTATE > breast > pancreas

In males: lung > prostate > bowel > pancreas > CUP (unknown primary)

In women: lung > breast > bowel > pancreas > CUP (unknown primary)

Cancer is the second leading cause of mortality
Most common overall: prostate, bowel, breast, melanoma, lung

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

What are the agents available for metastatic melanoma?

A
  1. Targeting the MAP kinase pathway:
    note 45% of melanomas have BRAF mutation –> constitutive activation of the MAP kinase pathway –> promotes proliferation, prevents apoptosis
    - BRAF INHIBITORS: Vemurafenib, **Dabrafenib
    - MEK INHIBITORS: Trametenib, Cobimetenib
  2. Immunotherapy
    - Anti-CTLA4 Antibody - **Ipilimumab
    - Anti-PD1 Antibody - **Nivolumab, Pembrolizumab
    - Anti-PD-L1 Antibody
    - Anti Interleukin 2 (not in Australia)

Older agents - Dacarbazine (alkylating agent) and Fotemustine
- Do NOT work, no improvement in overall survival

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

What are the side effects of the BRAF inhibitors?

A

SKRAP the Dog says “RAF RAF RAF!” (From Playschool)

Side effects of the BRAFs - Vemurafenib and Dabrafenib:
S - SCCs (20%) treat by excision, well tolerated)
K - keratoacanthoma (10%)
R - Rash
A - Arthralgias
P - Photosensitivity
(Also causes deranged LFTs)

“Dab your forehead - you have a fever!”
Dabrafenib also causes FEVER & alopecia

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

What is Trametinib?

What are the side effects?

A

Trametinib is a MEK inhibitor. (MEK is downstream of BRAF in the map kinase pathway)
Improves overall survival in metastatic melanoma.

Side effects:
Heart failure - reduced LVEF!
Diarrhoea
Rash
Ocular side effects
Peripheral oedema
(NO SCCs!)

(Trams in MElbourne make my “Heart-DROP”…..
TRAMetenib = MEK = side effects: Heart-DROP)

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

Is there a role for combination BRAF/MEK inhibition?

A

Yes. New data from 2014 supports combination as 1st line for metastatic melanoma (but not PBS approved)

Double inhibition shows progression-free survival, and combination Dabrafenib/Trametinib showed better overall survival,

with

NO significant increase in toxicity and LESS SCCs (1%)

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

What is the best evidence for the duration of adjuvant tamoxifen therapy for breast cancer?

A

New data from 2014 shows evidence for 10 YEARS of tamoxifen. Greater benefit than 5 years.

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

What is Ipilimumab?

What are the side effects?

A

Ipilimumab is a mab against the inhibitory CTLA-4 Tcell receptor.
Ipilimumab is a CTLA-4 BLOCKER used in metastatic melanoma.

As CTLA-4 usually stops T cell activation and proliferation, Ipilimumab

  • -> “inhibits the inhibition”
  • -> acts as a T cell agonist
  • -> promotes anti-tumour immunity in metastatic melanoma

(Note this is OPPOSITE to the action of Abatacept, which is a fusion Ig for CTLA-4, is used in RA (not as good as the TNFs or infliximab) –> potentiates the inhibitory action of CTLA-4 –> PREVENTS T cell activation and proliferation)

SIDE EFFECTS of CTLA-4 INHIB = “CaPiTaLS”
These IRAEs or Immune-related Adverse Events (75% of patients) are good signs that the drug is working. Side effects are reversible and easily treated with steroids +/- TNF blockade.
C - colitis
P - Pneumonitis, pituitary dysfunction (hypopituitarism)
T - thyroid hyper/hypo
L - liver dysfunction / hepatitis
S - SKIN RASH = ***VITILIGO a good prognostic sign, Pruritis, SJS, TEN.

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

What is Nivolumab?

What are the side effects?

A

Nivolumab is an Anti-PD1 antibody, used in metastatic melanoma.
(New evidence for use in NSCLC)
(New agent probably, not in written exam)

** USED IN BRAF NEGATIVE PATIENTS **

Note that PD1 and CTLA4 are both inhibitory receptors.
The PD1 receptor (Programmed Death 1) on Tcells is an immune checkpoint receptor that binds to the PD1 Ligand on tumour cells and makes them “invisible” –> renders them invisible from destruction by CTLs and CD4 Tcells.

Hence Nivolumab BINDS to PD1 receptor on Tcells and inhibits this anti-tumour response –> allows destruction of tumour cells by CTLs and CD4 Tcells.

SIDE EFFECTS:
Immune related adverse events (“CaPiTaLS” = same as for CTLA4 blockers) but also UVEITIS

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

What cancers do we recommend screening for?

A

Breast
Colorectal
CERVICAL

Not routinely recommended for- prostate, lung, ovarian, melanoma

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

Guidelines for breast cancer screening.

A

Age 50-74 - Mammograms every 2 years

(If patients are under 50 (>40) or over 75, can still have mammograms but these women are not formally invited to the Breast Ca Screening program)

NOT evidence for breast self examination as a stand-alone screening tool. Must use mammograms.

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

What are the guidelines for Screening for Colorectal Cancer?

A

Normal people, Age 50-75:

  • Annual FOBT
  • Colonoscopy every 10 years
  • Flexisig every 5 years

MODERATE Risk FHx (1st deg relative younger than 55)
- Colonoscopy every 5 years from age 50,
or 10 years younger than age of relative

SCREENING IN FAMILIAL CANCER SYNDROMES:
1. FAP / Familial Adenomatous Polyposis
(Auto Dom, mutated APC gene, get adenomas in childhood and CRCs by age 45. Prevention of bowel cancer with surgery/Colectomy usually at age 18)
- Sigmoidoscopy/Colonoscopy yearly from ADOLESCENCE, age 12-15!!!
- Endoscopic & Duodenal screening from age 25, or from time of Colectomy (to look for Duodenal adenomas and rectal carcinomas)

  1. HNPCC (2-4%)
    (Auto Dom, mismatch repair gene defect –> CRCs and increased endometrial and GU cancers)
    - Colonoscopy ONCE A YEAR from age 25,
    or 5 years younger than age of relative
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Spinal Cord Compression = Oncological emergency.

What is the most common mechanism of Spinal Cord Compression in Oncology patients?
How do patients present?
What cancers are usually associated?
What is the Mx?
What is the BEST predictor of outcome in these patients?

A

Most common mechanism is **epidural extension from vertebral body or pedicle.
Other mechanisms are:
- Extension of paravertebral tumour
- Pathological collapse of vertebral body
- Metastasis to meninges or cord.

Most common associated tumours: lung, breast, prostate

Presentation:
Back pain (>90%) worse at night and lying down.
Motor changes, sensory changes (saddle sensory loss in cauda equina), bowel and bladder dysfunction.

MANAGEMENT:
MRI whole spine
High dose dexamethasone ASAP! 10mg stat then 4mg QID.
Surgery to stabilise spine.
Radiotherapy alone if radio sensitive tumour, or radiotherapy after surgery.
(not chemo)

BEST PREDICTOR of outcome is the neurological status at the START of treatment.

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

An oncology patient presents with:

  • facial and arm swelling (often worse with bending forward / lying)
  • headaches / pressure sensation in head
  • SOB
  • cough
  • hoarse voice
  • epistaxis

What is the condition?
What are the associations?
Management?

A

Superior Vena Cava Syndrome = Oncological Emergency!
- partial or complete obstruction of bloodflow from SVC to right atrium due to tumour compression, invasion and/or thrombosis

May develop laryngeal oedema / stridor from airway obstruction, or cerebral oedema / confusion / coma.

Associated with Lung Ca > NHL > other

MANAGEMENT:
- CT Chest to look at SVC - diagnostic.
- remove any central lines if present that may be causing thrombus
- ANTICOAGULATE if thrombus
(No evidence for steroids or diuretics for facial oedema)

  • CHEMOTHERAPY for SCLC, NHL, germ cell tumours
  • **ENDOLUMINAL STENT!
  • -> rapid and sustained symptom improvement
  • -> no need to get histology first
  • RADIOTHERAPY either alone or after stent
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the benefits of measuring CEA?

A
  • good indicator of PROGNOSIS (if elevated CEA pre-operatively, then worse prognosis)
  • used to monitor for disease RELAPSE post curative surgery
  • used to monitor RESPONSE TO TREATMENT of metastatic disease

(But CEA also raised in other cancers- most adenocarcinomas, medullary thyroid Ca, PUD/gastritis, COPD, diabetes, liver disease, SMOKING, inflammation)

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

Which tumour markers are used to monitor for RELAPSE and RESPONSE TO TREATMENT?

Which tumour markers have PROGNOSTIC value?

A

Monitors RELAPSE and RESPONSE TO TREATMENT:
CEA in CRC
Ca 19-9 in pancreatic Ca
AFP (also used in screening for HBV/HCV/cirrhosis- high risk patients)
Ca 125 in ovarian Ca
AFP/betaHCG/LDH in testicular cancer
PSA in prostate cancer
ChromograninA (CgA) in neuroendocrine tumours
Thyroglobulin in thyroid Ca
Calcitonin and CEA in medullary thyroid cancer

Monitors RESPONSE TO TREATMENT ONLY:
Ca 15-3 and CEA in breast cancer is only used to monitor response to treatment in metastatic disease
(NO ROLE for measuring for disease relapse)

PROGNOSTIC value:
CEA
Ca 19-9
AFP/betaHCG/LDH
Thyroglobulin!
Calcitonin and CEA in medullary thyroid cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Germ Cell Tumours / Testicular Cancer.

How do you differentiate Pure Seminomas from Non-Seminomas.

A

Look at the AFP.

Pure Seminomas:
BetaHCG normal/high
LDH normal/high
AFP NORMAL

Non-seminomatous Germ Cell tumours:
BetaHCG normal/high
LDH normal/high
**AFP INCREASED (in 80%)

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

What are the benefits of measuring Chromogranin A?

A

Chromogranin A is more sensitive than Urine 5HIAA for neuroendocrine tumours but it is less specific –> better for monitoring RECURRENCE of disease.

Used to monitor disease progression, relapse, response to treatment.

Benign causes - PPI use, renal and liver dis, atrophied gastritis, IBD

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

What are the gene characteristics of the BrCa1 and BrCa2 genes?

What is the usual function of the BrCa1 and 2 genes?

A

BrCa1 gene
- on chromosome 17q/22
> 80% of mutations are truncating
- Higher (female) cancer risk compared with BrCa2

BrCa2 gene

  • on chromosome 13q
  • 27 exons which include the OCCR Ovarian Cancer Cluster Region on exon 11
  • Higher risk of MALE breast cancer, prostate cancer, pancreatic cancer
  • BRCA2-associated Prostate cancer is more aggressive, with greater chance of T3/4 disease and node positivity at diagnosis. Gleason score often > 8. –> **High risk therefore need Immediate Radical Prostatectomy!!

GENE FUNCTIONS:

  • both have similar roles in dsDNA repair
  • function as TUMOUR SUPPRESSOR GENES
    • -> inactivation of these genes leaves DNA damage unrepaired –> leads to cancer.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What are the tumour features associated with BRCA 1 and 2 mutations?

A

Tumours associated with BRCA1 appear to have typical histological features:

  • high grade
  • hormone receptor NEGATIVE in 75%!!!
  • pushing borders
  • less DCIS
  • medullary or atypical medullary histology
  • basal epithelial phenotype

BRCA2 tumours do NOT have any specific phenotypic features

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

What are the characteristics of Hereditary epithelial ovarian cancer?

A

These are ovarian cancers that develop in BRCA1 and 2 mutation carriers.
Recent data shows BRCA mutation rate in invasive epithelial ovarian cancers = 14%.
Highest mutation rate is in invasive SEROUS ovarian cancers.

Classically:

  • SEROUS PAPILLARY ovarian cancers (85% vs 40% in sporadic ovarian ca’s)
  • less common to see other histological subtypes such as clear cell, endometrioid ca.
  • MORE RESPONSIVE TO PLATINUM CHEMO (better overall survival) compared with non-BrCa-associated Ovarian ca’s
  • ** Guidelines suggest to TEST ALL CASES OF INVASIVE EPITHELIAL CANCERS FOR BRCA MUTATIONS

(Note - mucinous adenocarcinomas are NOT associated)

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

In which patients is it appropriate to offer Gene Testing for Familial Breast and Ovarian cancer (BRCA gene testing)?

A

Offered to HIGH RISK families.

High risk includes:

  • 3 or more cases of breast/ovarian cancer in 1st or 2nd degree relatives
  • 2 or more cases of breast/ovarian cancer in 1st or 2nd degree relatives, PLUS one high risk feature
  • bilateral breast cancer
  • male breast cancers
  • age under 40
  • OVARIAN cancer

(There are computer programs that can calculate the patient’s risk of mutation - generally the arbitrary cut off is to offer gene testing if the probability is >10%)

If a BRCA MUTATION IS IDENTIFIED –> can offer other family members predictive gene testing. (If positive in 1st deg relative, the risk of having a mutation is 50% based on auto dominant inheritance)

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

If someone is a BRCA gene carrier, what are their options for:

  1. surveillance (watchful waiting)
  2. prophylactic surgery
  3. chemoprevention?
A
  1. SURVEILLANCE
    - YEARLY MAMMOGRAMS AND BREAST MRI from age 25-30,
    or 5-10 years before the age of the youngest affected relative.

(Breast MRI highly sensitive in detection but NO proven survival benefit)
(No proven benefit in clinical breast examination, Ca 125 monitoring or yearly transvaginal ultrasounds)

  1. PROPHYLACTIC SURGERY FOR BREAST OR OVARY
    BREAST SURGERY
    - bilateral mastectomy (with or without breast reconstruction) offers best protection against breast cancer - leaves a residual risk of 1-3%
    - age < 50 years best outcome
    - the younger the patient, the higher the relative risk of developing breast cancer
    eg. 30yo has 60% lifetime risk, 60yo has 15% lifetime risk.
    Also
    SECONDARY PREVENTION - if Br Ca before age 40, there is a 50% risk of developing a second primary Bc Ca in the next 25 years.

OVARY / SALPINGO-OOPHRECTOMY

  • at age 40-45
  • consider at age 35 if patient BRCA1 carrier (as 4% risk of ovarian cancer before age 40)
  • reduces risk of ovarian cancer by 80%. Leaves a residual risk of primary peritoneal cancer of 2-5%
  • reduces risk of breast cancer if performed before menopause (RR 0.5)
  • must follow up BMD and CV health!!
  1. CHEMOPREVENTION = “Risk Reducing Medication”
    - -> TAMOXIFEN (Anti-oestrogen) daily for 5 years, but benefits extend to 10 years. Reduces risk of hormone positive breast cancer by 30-40% but no proven survival benefit)

side effects:
Increased risk endometrial cancer (with Tamoxifen only, in postmenopausal), VTEs.

–> HRT: used after BSO to age 50. Never use if prior breast cancer.

–> OCP: protective against ovarian cancer. Issue of possible increase in breast cancer risk- need balance.

  • -> PARP INHIBITORS (OLAPARIB)
  • active in breast and ovarian metastatic disease.
  • increased response rates and PFS but not overall survival.

polyADP ribose polymerase enzymes are involved in an alternative DNA repair pathway (base excision repair) – tumour cells have defective dsDNA repair (methylation or somatic BRCA mutation) and rely on alternative DNA repair pathways such as PARP.

  • -> BLOCKING this repair pathway in BRCA-associated tumours is LETHAL to the cancer.
  • -> confers “selective tumour cytotoxicity” because normal tissues have normal BRCA function but cancer tissues have no normal BRCA function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is Hereditary breast/ovarian cancer syndrome?

What are the risk factors / when would you suspect this syndrome in your patient?

A

Hereditary breast/ovarian cancer syndrome:

  • BrCa genes 1 and 2
  • penetrance 50-80% of gene carriers develop cancer
  • explains most of high risk breast and ovarian cancer families, and approx 30% of high risk Br Ca families

Suspect in:
- early age

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

What is the lifetime risk of cancer in:
BRCA1
And
BRCA2?

A

BRCA1

  • Breast cancer –> 40-80%
  • Ovarian cancer –> 20-40%
  • Prostate cancer in males –> Increased relative risk by 1.5-2

BRCA2

  • Breast cancer –> 40-60%
  • Ovarian cancer –> 10-20%
  • Breast cancer in males –> 6%
  • Pancreatic cancer –> 3.5%
  • Pancreatic cancer high grade –> Increased RR by 4+
  • Melanoma –> Incresed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is Li Fraumeni Syndrome?

A

p53 p53 p53!!!!
(“P” Frau”MANY” Cancers!)
CANCERS IN 90% by age 60!!! - BLAGS (see below)

Auto dominant GERMLINE p53 mutation on chromosome 17!

Multiple primary tumours common.
Prevalence 1 in 20,000. Rare.

PRESENTATION: “BLAGS”
Breast > Leukaemia > Adrenal > GBM > Sarcomas

“Breast-sarcoma syndrome”

  • often HER2 positive pre-menopausal breast cancer
  • SARCOMAS (non Ewing)
  • brain tumours - GBM
  • leukaemia
  • **Paediatric cancers
  • **Adrenocortical cancers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is Cowden Syndrome?

A

Auto dominant GERMLINE PTEN mutation on chromosome 10.

Characterised by:

  • Breast cancer (25-50% risk) and fibrocystic disease
  • Thyroid cancer (10%) and benign thyroid disease
  • Cutaneous lesions (Oral papilloma and skin keratoses)
  • Macrocephaly
27
Q

What is Gardner syndrome?

What is Turcot syndrome?

A

These are “FAP - Plus” Syndromes.

Gardner = FAP + extraintestinal features
(Osteomas, desmoid tumours, epidermoid cysts)

Turcot = familial colon cancer (either FAP/HNPCC) + brain tumour
That is,
FAP + medulloblastoma
Or 
HNPCC/Lynch + GBM
28
Q

What are the genes involved in HNPCC (Lynch syndrome) and what is their tumour-promoting mechanism?

A
MLH1
MSH2
MSH6
PMS2
------>> defective mismatch repair

Auto dominant tumour suppressor genes.
Caused by mutations in DNA MISMATCH REPAIR GENES –> leads to genetic instability –> random mismatches in DNA are left uncorrected.

Explains 3-4% of CRC.

29
Q

What features are more common in HNPCC compared to other colorectal cancers?

What are the other cancers that have increased risk in HNPCC?

A
Proximal disease (right sided) (70% in HNPCC cf 30% in other CRC)
Microsatellite instability (95% cf 10%)
Poorly differentiated
MUCINOUS
Lymphoid reaction

Younger age 40-50 (cf age 65-70 in other CRC)
Affects M=F (cf M>F in other CRC)

OTHER CANCER RISK IN HNPCC-
Endometrial (30-50%), stomach, ovarian

30
Q

What are Familial Paraganglioma Syndromes?

A

Paraganglioma = tumour of the autonomic NS.
Auto dominant.

Caused by mutations in the genes encoding MITOCHONDRIAL enzymes.

** Genes mainly SDHB and SDHD **

Tumours are usually benign and can be from base of skull to sacrum.
Young age at diagnosis 20-30’s.
** Phaeo’s occur in these syndromes! **

(“The “GANG” in the SyDney HUB” is “DOMinant and MIGHTY”
= ParanGANGlioma = SDHB gene, Auto Dom, MITochondria)

31
Q

What is Von Hippel Lindau syndrome?

A

A GERMLINE mutation of the VHL a gene on chromosome 3p.
Auto dominant.

Diagnosis (need 2 or more of):
- retinal angiomas
- CNS haemangioblastoma
- renal cell ca - clear cell
- Phaeo
(Also p/w pancreatic/renal cysts)
32
Q

New syndromes.

What is a BAP1 mutation?

A

BAP1 = a BrCa1-Associated Protein
A protein that binds BRCA1
Acts as a tumour suppressor gene
GERMLINE mutations –> result in benign MELANOCYTIC skin lesions

In young: atypical skin lesions, atypical Spitz naevus

In adults: mesothelioma, melanomas - uveal and cutaneous, lung adeno’s, meningitis

DO SKIN CHECKS AND EYE EXAMS FROM AGE 18 in patients with positive mutation

33
Q

New mutations.

What are POLD1 and POLE mutations?

A

(“Polly” genes –> result in “Polyposis”)
Cause POLYPOSIS or early age COLORECTAL CA that is Microsatellite-stable (No Microsatellite instability)

Encode DNA polymerase enzymes.
Repair DNA a damage.

POLD1 also increases risk of endometrial cancer

34
Q

What is the mechanism of action of Abiraterone and what are the side effects?

A

Abiraterone is a CYP17 Antagonist –> inhibits the action of 17-alpha Hydroxylase at all sites (Adrenals, testes, prostate cancer) hence Pregnenolone not converted to 17-OH progesterone
–> inhibits Androgen biosynthesis.

The side effect is that the Pregnenolone is shunted to the mineralocorticoid pathway –> INCREASED ALDOSTERONE
–> Hypokalaemia, HTN, Fluid retention

There is also DECREASED CORTISOL production (due to lack of upstream 17-OH Progesterone) –> Addisons
HENCE ALL PATIENTS ON ABIRATERONE MUST BE STARTED ON PREDNISONE TO AVOID ADDISONIAN CRISIS.

35
Q

What are the recommendations for screening in Prostate cancer?

A

None in Australia. Controversial.

Recent data shows benefit if screening with PSA is largest in men 50-64 yo with life exp > 10 years.

36
Q

What are the treatments available for metastatic Prostate Cancer?

A

Firstly classify metastatic Prostate Cancer into 2 groups,
1. Hormone sensitive (early) and
2. Castrate-resistant
secondly, treat Bone Metastases.

  1. HORMONE-SENSITIVE PrCa:
    Treat with ANDROGEN DEPRIVATION THERAPY
    = GnRH (super)AGONIST eg. Gosrelin, Leuproline
  • initially causes a flare phenomenon for 2 weeks –> rise in LH and FSH –> rise in Testosterone,
    HENCE MUST ALSO GIVE A TESTOSTERONE ANTAGONIST (FLUTAMIDE, NILUTAMINE) for 2/52 prior and post starting Gosrelin

Eventual effect of GnRH Agonist Gosrelin is to DOWNREGULATE LH/FSH RECEPTORS –> HYPOGONADISM/decreased Testosterone.

–> note this treatment cases Osteoporosis 2ry to hypogonadism

2. CASTRATE-RESISTANT PROSTATE CANCER
Management is broken up into 3 phases
- Pre-Docetaxel
- DOCETAXEL = Microtubule inhibitor
- Post-Docetaxel 

PRE-DOCETAXEL AGENTS
= Anti-Androgens
1. Abiraterone (CYP17 inhibitor/ Adrenal Biosynthesis inhibitor) + give with PREDNISONE.
Or
2. Enzalutamide (an androgen receptor antagonist –> blocks testosterone from exerting any effect)
Or
3. Evidence for Radium 223 if not fit for chemo, or Sipuleucel-T (expensive, controversial American drug but survival benefit)

DOXETACEL
= gold standard treatment for all castrate-resistant PrCa’s
= increases overall survival and reduces PSA levels by >50%
S/E peripheral neuropathy, pulm toxicity, diahrroea, fluid ret, febrile neutropaenia

POST-DOCETAXEL CHEMOTHERAPY
Survival benefits with:
1. CABAZITAXEL = overcomes Docetaxel resistance!
2. Antiandrogens
Abiraterone or Enzalutamide
3. Radium 223

TREAT BONE METASTASES / OSTEOPOROSIS FROM TREATMENT
(Any old Male with pancytopaenia, think of bone marrow infiltration from prostate cancer or lymphoma)
1. Zoledronic acid
2. Denosumab
(These improve BMD and lengthen time to first skeletal event, but NO SURVIVAL BENEFIT)

**Denosumab is SUPERIOR to ZA in castrate-resistant Pr Ca **
(non-inferior to ZA in other cancers)
Denosumab can be used in renal impairment

37
Q

What are the genetic mutations found in metastatic melanoma?

What are the drugs that target these mutations?

A

DRUG TARGETS IN MET MELANOMA:
(Think of Australia as the melanoma capital of the world -
“We visit Brisbane, Cairns and Melbourne which are “checkpoints” for melanoma”)
= B, C, M, plus the immune checkpoint inhibitors (CTLA4 and PD1)
= BRAF, C-Kit, MEK, CTLA, PD1

Mutations:
BRAF
C-Kit
MEK
NRAS
--> all of these mutations cause constitutive activation of the MAP kinase ONCOGENIC pathway

BRAF - Dabrafenib, Vemurafenib
C-Kit - Imatinib (TKI)
MEK - Trametenib
NRAS - NONE!

(Side note) - 
Also use Immune checkpoint inhibitors for met melanoma:
CTLA4 - Ipilimumab
PD1 - Nivolumab
Side effects are IRAE's
38
Q

What chemotherapy drugs cause CARDIOTOXICITY?

A
5FU
Capecitabine (= oral 5FU)
Anthracyclines - rubicins 
Traztuzumab
Bevacizumab
Sunitinib

5FU/ Capecitabine

  • chest pain from coronary artery VASOSPASM
  • RFs: dose-dependent, concurrent radiotherapy, infusions (cardiac disease NOT predictive)

Anthracyclines - RUBICINS

  • Cardiomyopathy and Heart Failure
  • acute: arrhythmias / heart block
  • RFs: CUMULATIVE dose
  • Mx: treat Heart failure

Traztuzumab

  • decreases LVEF –> clinical heart failure (reversible)
  • RFs: age, anthracyclines, pre-existing heart dysfunction

Bevacizumab
- HTN

Sunitinib, Sorafenib
- decreases LVEF but rarely causes HF

39
Q

What chemotherapy drugs cause PULMONARY TOXICITY?

A
Bleomycin
Taxanes 
Gefitinib, Erlotinib
Everolimus 
Other agents - Gemcitabine, CTX, Temozolomide 

BLEOMYCIN

  • -> Interstitial Pulm Fibrosis!!! May be FATAL.
  • less common, COP & Hypersensitivity pneumonitis
  • do a baseline DLCO

Taxanes

  • diffuse interstitial pneumonia
  • pulmonary oedema & pleural effusions
  • RFs: combination chemotherapy, radiotherapy

Gefitinib, Erlotinib (EGFR TKIs)

  • Insterstitial lung disease –> fatal in 1/3 of patients
  • RFs: smoking, preexisting lung disease

Everolimus
- Pneumonitis

40
Q

What chemotherapy drugs cause NEUROTOXICITY?

A

Platinum agents
Taxanes
Vinca Alkaloids
Bevacizumab

Platinum agents -
Cisplatin worse > Carpoplatin:
- peripheral neuropathy
- OTOTOXICITY - Tinnitus and high-freq hearing loss

Oxaliplatin

  • ATAXIC neuropathy (large fibres involved), acute neurotoxicity
  • COLD DYSASTHESIAS

Taxanes
- Nab-paclitaxel > Paclitaxel > Docetaxel > Cabazitaxel
- glove & stocking PURE SENSORY neuropathy with LOSS OF REFLEXES, motor function preserved.
(Occasionally impaired with Paclitaxel)

Vinca Alkaloids

  • AXONAL NEUROPATHY, *** sensory&raquo_space; motor
  • AUTONOMIC NEUROPATHY

Bevacizumab

  • (rare) Reversible posterior leukoencephalopathy syndrome
  • headaches, confusion, visual changes, seizures
41
Q

What are the targets for biological agents in Clear Cell Renal cancer?

What are the drugs used and what are their main side effects?

A
  1. VEGF (ligand and receptor)
  2. PDGF Receptor
  3. mTOR
    (RENA said, “ My VEGgie Patch needs MoniTORing after I CUT OUT the weeds!”)

** ALWAYS CUT OUT / perform a NEPHRECTOMY in ALL PATIENTS even in metastatic disease **

  1. VEGF inhibitors are first line treatment
    - TKIs increase progression-free survival but NOT overall survival/mortality
    - SUNITINIB blocks both VEGF and PDGF RECEPTORS
    - PAZOPANIB only blocks VEGF1 receptor
    - Both Suni and Pazopanib have same effects but Pazop slightly better quality of life.
- S/E SUNITINIB: 
HTN =good!
THYROID dysfunction
Hand and foot rash
Cardiotoxic/ LV dysfunction (worse with Sorafenib)
  • S/E PAZOPANIB:
    HTN = good
    LIVER derangement
  1. 2nd line treatment
    - BEVACIZUMAB, VEGF “LIGAND” inhibitor, S/E HTN
  • mTOR INHIBITORS = TEMSIROLIMUS IV, EVEROLIMUS PO
  • S/E are METABOLIC derangement & pulm toxicity
    HI BSLs
    HIGH LIPIDS (TG in the 20’s!)
    Pneumonitis

TEMSIROLIMUS is the only drug with a SURVIVAL BENEFIT

42
Q

What are these tumour are these markers associated with?

  1. Beta HCG
  2. PLAP / Placental alkaline phosphatase
  3. 5HIAA
  4. Chromogranin A
  5. Calcitonin
  6. Thyroglobulin
  7. AFP / Alpha Feto Protein
A
  1. Beta HCG - germ cell tumours (testicular, choriocarcinoma etc)
  2. PLAP / Placental alkaline phosphatase - SEMINOMA testicular cancer (better prognosis)
  3. 5HIAA - “hire a car” CARCINOID tumours, mainly in small bowel
  4. Chromogranin A - PhaeoCHROMOcytoma, neuroendocrine tumours
  5. Calcitonin - MEDULLARY thyroid cancer
  6. Thyroglobulin - Differentiated (non-medullary) thyroid cancer, usually PAPILLARY thyroid Ca
  7. AFP / Alpha Feto Protein - NON-SEMINOMA Testicular cancer (worse prognosis), germ cell tumours, HCC
43
Q

What are the risk factors for Renal Cell Carcinoma?

A

Smoking
HTN
Obesity

VHL auto dominant gene (2-3%) - Von Hippel Lindau is the most common cause of inherited RCC –> accumulation of HIF-alpha –> increased growth factors VEGF and PDGF.

44
Q

What is the management for Testicular cancer?

A

Seminomas (50%) and Non-Seminomas (50%) differentiated by histology and presence of PLAP/AFP.

Mx of testicular cancer IS PLATINUM BASED CHEMO.

SEMINOMAS
- better prognosis, virtually ALL curable
- Options for management are Orchidectomy and surveillance,
Or
Orchidectomy and adjuvant chemo with CARBOPLATIN
= 99% cure rate.
NEVER Radiotherapy as increased risk of 2nd malignancy.

Basically DO EVERYTHING to cure testicular cancer!!!
- New evidence for giving high-dose CTX (Cyclophosphamide) and STEM CELL RESCUE for metastatic testicular cancer

NON-SEMINOMAS
- worse prognosis, hence need 3 AGENTS “BEP”
- Orchidectomy cures 60%
- need active surveillance with
AFP
BetaHCG
LDH
CT imaging
- BEP = BLEO, ETOPOSIDE, CISPLATIN (platinum agent the most important)
- BLEOMYCIN S/E: Pneumonitis, can be fatal, hence monitor DLCO. Hypersensitivity reaction.
- CISPLATIN S/E: peripheral neuropathy, tinnitus/Ototoxicity, renal impairment

45
Q

Young man presents with mediastinal mass. What are your main DDx?

A
Sarcoidosis
Lymphoma
TESTICULAR CANCER (as gonadal tissue arises from chest embryonically)
46
Q

Testicular cancer patient presents with residual mass post curative chemotherapy. What is the diagnosis and what is your management?

A

Most masses are fibrosis or TERATOMA which has a risk of malignant transformation into an adenocarcinoma.

Hence all post-chemo residual masses need to be surgically resected.

47
Q

What are the risk factors for Colorectal Cancer? Which is the most important?

A
Personal or Family Hx - most important
Ulcerative colitis increases risk by 15x
IBD/Crohns but less risk than UC
Obesity
T2DM
Alcohol
48
Q

What is the minimum number of lymph nodes needed to sample in colorectal cancer?

A

Need minimum 12 lymph nodes!!!

Increased risk of recurrence when 11 or less LN’s sampled

49
Q

What genetic mutations do you test in Colorectal Cancer?

What biologic agents target these?

A

“Big KEV has colorectal cancer”

Bad = KRAS mutation
EGFR
VEGF
(Also if you suspect HNPCC then do MLH1 and MSH2)

Biological targets:
KRAS mutation - none, but important to know if using Cetuximab
EGFR - Cetuximab
VEGF - Bevacizumab

** Important to note:
KRAS is DOWNSTREAM of the EGFR receptor, hence blocking EGFR with Cetuximab will have no effect because the downstream KRAS is constitutively active
–> Hence patient needs to have a NORMAL KRAS (ie KRAS WILDTYPE) to qualify for Cetuximab. **

So treatment is limited BUT can use Bevacizumab in these patients.

MANAGEMENT OF CRC:
1. Chemo (FOLFOX or FOLFIRI) plus BEVACIZUMAB
= first line treatment

  1. 2nd line treatment for disease progression -
    CETUXIMAB (EGFR inhibitor) but only if KRAS normal/wildtype.
    - S/E of Cetuximab is rash = good!
50
Q

What is the management for metastatic colorectal cancer?

What are the side effects of this adjuvant chemo?

A
  1. First line = FOLFOX / FOLFIRI “plus” BEVACIZUMAB
    (Big KEV fears foxes)

FOLFOX - Folinic acid, 5FU, Oxaliplatin
FOLFIRI - Folinic acid, 5FU, Irinocetan (a Topoisomerase inhibitor)
** Note Capecitabine = oral 5FU ***

S/E 5FU/Capecitabine:

  • Cardiotoxicity/ vasospasm
  • plantar palmar erythema (red, tender hands) - worse with Capecit.
  • diahrroea
  • mucositis

S/E OXALIPLATIN:

  • peripheral neuropathy
  • renal impairment

S/E IRINOCETAN:

  • cholinergic effects
  • diahrroea

–> Folic acid and B12 is given to reduce toxic side effects

51
Q

What is the Mx for pancreatic cancer?

A

Whipples cure rate is only 25% - but tumours rarely resectable at presentation anyway

Adjuvant chemo:

  • single agent 5FU or Gemcitabine
  • 4-agent chemo for metastatic disease: FOLFIRINOX (ie the agents that we use in CRC) –> improves survival from 7 to 11 months compared to single agent Gemcitabine
  • can also try Taxanes
  • ERLOTINIB (EGFR inib) as 40-80% express EGFR mutation
52
Q

How does Methotrexate work?
How does MTX toxicity present?
What is the treatment for toxicity?

A

MTX is a dihydrofolate reductase (DHFR) inhibitor

  • -> blocks purine/pyrimidine synthesis
  • -> blocks DNA/RNA synthesis

–> Also causes Folate deficiency (ANTIFOLATE AGENT)
Hence Folate supplementation reduces side effects

MTX toxicity:

  • GI symptoms
  • Bone Marrow toxicity
  • CNS toxicity

Treatment is FOLINIC ACID RESCUE for 3 days

53
Q

What are the poor prognostic features of breast cancer?

Which is the worst prognostic feature?

A

Worst feature is LN involvement (MCQ) - axillary

Poor prognostic features:

  • positive nodes
  • ECOG >3 at diagnosis
  • increasing in size
  • NEGATIVE E & P Hormone receptors (ER or PR neg)
  • POSITIVE HER2 mutation (15%) = bad because cells are constitutively active and increased risk of mets and relapse
  • young age
54
Q

What is the treatment for receptor-positive Breast Cancer?

What are the side effects of treatment?

A

EARLY STAGE:

  • surgery and radiotherapy (all pts who have surgery also get RTx)
  • Neo-Adjuvant chemotherapy

(oncotype microarray testing provides a 10-yr “ recurrence score”. If >30 then high risk)

ADJUVANT CHEMO:
ANTHRACYCLINE (RUBICINS) and TAXANE based chemotherapy

TAMOXIFEN = a SERM
** In cancer cells - selectively blocks the oestrogen receptor (ER ANTAGONIST)
But
** In bones and endometrium, it potentiates the oestrogen effect (ER AGONIST) –> good for bones (improves BMD!) but bad for endometrium (increases risk of UTERINE Cancer)

  • Tamoxifen reduces the recurrence rate of Br Ca by 40%
  • S/E Tamoxifen
  • DVT/PE (3% increased risk over 5 years)
  • Uterine cancer

ANASTROZOLE = Aromatase inhibitor
- works only on adrenal/non-ovarian tissue to reduce formation of oestrogens.
(Not useful in premenopausal women as their ovaries are still producing oestrogen)

*** note that the Aromatase that converts Testosterone to Oestradiol is more dominant in PREmenopausal women, whereas the Aromatase that converts DHEAS/Androstenedione to Oestrone is more dominant in POSTmenopausal women)

  • S/E of Aromatase Inhibitors
  • Osteoporosis (worsens BMD as less Oestrogen!)
  • Hypercholesterolaemia
  • IHD
  • Arthralgias
    (NO increased risk of DVT/PE or Uterine Ca)
55
Q

What is the treatment for HER2 positive Breast Cancer?

A

TRAZTUZUMAB for 12 months

S/E

  • cardiotoxicity–> REVERSIBLE cardiomyopathy; need to do a TTE every 3 months
  • allergic reaction
  • Trastuzumab has NO CSF PENETRATION hence use LAPATINIB (a small TKI) if not responsive to Trastuzumab.
  • Note you need 3+++ positive HER2 results on immunohistochemistry to make a diagnosis of HER2 positivity. (HER2 1+ is negative)
56
Q

What is the treatment for TRIPLE NEGATIVE metastatic breast cancer?

A

PARP Inhibitors!!! (Very examinable!)
Poly-adenosine diposphate ribose polynuclease
“BMN 673”

Also Radiotherapy for symptoms of:
Bone pain, SC compression, Brain mets, Ulcerating cancer

bone Mets / skeletal protection use Bisphosphonates and Denosumab plus Vit D

57
Q

What are the indications for Radiotherapy in metastatic breast cancer?

A

Metastatic disease is generally incurable, so use chemo and/or Radiotherapy only to improve symptoms.

Indications for Radiotherapy:
Bone pain
SC compression
Brain mets
Ulcerating cancer
58
Q

Compare and contrast NSCLC and SCLC.

A

NSCLC more common (85%) and SCLC (13%)

NSCLC has 3 main subtypes -
1. Adenocarcinoma (better prognosis) - common 40%
- subtype Broncheoalveolar has better survival, seen in Females and nonsmokers
2. SCC
3. Large cell (worse prognosis) - most common (70%)
Others are
- Carcinoid (slow growing neuroendo tumour)
- mesothelioma

SCLC

  • more aggressive, spreads early to brain
  • 70% of patients have mets on presentation
  • smokers
  • involves larger airways
  • assoc with SIADH, PARANEOPLASTIC processes
  • ** sensitive to Platinum Doublet chemo (Plat and etop) but response is NOT durable***
59
Q

What mutations do you test for in lung cancer?

What are the biological targets for these?

A

EGFR and ALK
*** And the T790M mutation tells you if patient will be resistant to EGFRs.

EGFR MUTATION:

  • usually in FEMALE, ASIAN, NONSMOKERS
  • present in 50-70% of NSCLC, 15% in adenocarcinomas
  • GEFITINIB / ERLOTINIB = EGFR receptor blocker TKI’s
  • or CETUXIMAB = EGFR ligand blocker mab

USE GEFITINIB or ERLOTINIB TKI’s as FIRST LINE AGENT MONOTHERAPY!!! More effective than chemo.

S/E for all of the above EGFR blockers:
Rash
Pulmonary fibrosis
Diarrhoea

  • *T790M mutation is a C–>T mutation, associated with EGFR TKI RESISTANCE
  • *New evidence that an agent called CO-1686 can cause tumour inhibition that is independent of EGFR signalling

ALK MUTATIONS:

  • 5% of adeno’s
  • CRIZOTINIB is an inhibitor of ALK phosphorylation
60
Q

Which chemo drugs are highly emetic?
Best treatment for chemo-induced nausea?
What is the mechanism of action of these drugs?

A

highly emetic - PLATINUM chemo agents
(“These PLAITS make me nauseous, my head spins!”)

TRIPLE THERAPY - “I get nausea coming back from my ADO”
A- Aprepitant
D- dexamethasone
O- Ondansetron
- superior to ANY single agent, and ANY metoclopramide combo’s

Mechanism of action:
APREPITANT: a neurokinin1 receptor antagonist –> good for treatment and PREVENTION of nausea

Dexamethasone

Ondansetron is a 5HT3 Serotonin antagonist

61
Q

How do you diagnose a Carcinoid tumour?

A

Clinical presentation - “Your CAR is the latest FAD”
Facial FLUSHING
Abdo pain
Diarrhoea

Also has cardiac, hepatic manifestations if mets are present.

Dx= 5HIAA on a 24-HOUR URINE sample

5HIAA is a metabolite of Serotonin/5HT that is responsible for the diarrhoea +/- cardiac manifestations

COMMON SITES OF TUMOUR:
Terminal ileum - MOST COMMON
appendix
Rectum

Mx: OCTREOTIDE. Inhibits hormone production by several tumours, as it is a somatostatin analogue

62
Q

Define the ECOG Performance Statuses.

A

0 - fully able
1 - Ambulatory but limited strenuous activity
2 - Less than 50% bed bound, still able to perform self-care
3 - More than 50% bed bound, needs assistance with ADLs
4 - 100% confined to bed

63
Q

What type of chemo agent is Cyclophosphamide?

A

CTX is an alkylating agent. Cross links DNA.

64
Q

TAMOXIFEN. What is the mode of action and what are the side effects?

A

TAMOXIFEN = a SERM (ER Antagonist)
** In cancer cells - selectively blocks the oestrogen receptor
But
** In bones and endometrium, it potentiates the oestrogen effect (ER AGONIST) –> good for bones (improves BMD!) but bad for endometrium (increases risk of UTERINE Cancer)

  • Tamoxifen reduces the recurrence rate of Br Ca by 40%

SIDE EFFECTS:
(“TAMara is doing ENDO training in Pow” = TAMoxifen - ENDOmetrial cancer, “P”E risk)

  • DVT/PE (3% increased risk over 5 years)
  • Endometrial/Uterine cancer
65
Q

AROMATASE INHIBITORS eg. ANASTROZOLE

what is the mode of action and what are the side effects?

A

ANASTROZOLE = Aromatase inhibitor

  • works only on adrenal (+/- non-ovarian) tissue to reduce formation of oestrogens.
  • BLOCKS conversion of
    Androstenedione –> Oestrone
    Testosterone –> Oestrogen
  • DO NOT USE in premenopausal women as their ovaries are still producing oestrogen
  • SIDE EFFECTS:
    (“dr ANA Ananda works with OSTEO and ARTHRALGIAs and her HEART is CHOLD (COLD)!!” = ANAstrasole –> Osteoporosis, Arthralgias, Ischaemic HEART disease, CHOLesterol)
  • OSTEOPOROSIS (worsens BMD as less Oestrogen!)
  • Hypercholesterolaemia
  • IHD
  • Arthralgias

(NO increased risk of DVT/PE or Uterine Ca)

*** note that the Aromatase that converts Testosterone to Oestradiol is more dominant in PREmenopausal women, whereas the Aromatase that converts DHEAS/Androstenedione to Oestrone is more dominant in POSTmenopausal women