Unit 6 - Targeted Therapy Flashcards

1
Q

What is personalised medicine?

A

An approach to give a specific form of treatment (either drug choice or dose regime) to a patient sub-group in order to maximise treatment efficacy and minimize any side-effects

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

What factors could be responsible for the difference in patient response (HER2+ breast cancer) to neratinib?

A
  • Extrinsic factors (diet, stress – as cortisol can activate cAMP pathway modifying drug response)
  • Host genetics – pharmacogenetics – perhaps SNPs in CYP enzymes for instance
  • Other tumour characteristics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What ways can a molecular driver of tumour growth be identified?

A

Techniques like NGS or gene microarray – from biopsies or CTC/CFDNA.
Importantly this must be causative not correlative.

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

What are the two types of biomarkers?

A

Prognostic – indication of patient outcome (e.g., ER+ breast cancer has favourable outcome)
Predictive – suggests a patient might respond to a certain type of treatment/therapy – HER2+ breast cancer for Tratuzumab or olaparib with BRCA mutated ovarian cancer

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

Describe the ways we can analyse biomarkers

A

IHC for ER/PR
NGS for specific mutations like L858R in NSCLC
Oncotype DX looks at 21 genes associated with different processes in breast cancer – (compared to reference) and then determines with there is a high risk of recurrence and subsequently the severity of treatment (chemotherapy vs anti-hormones)

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

What are the differences between nonsynonymous and synonymous mutation and how could this be important in drug response?

A

A non-synonymous mutation changes the residue in the protein, whilst a synonymous mutation does not!
This could either effect protein structure and activity or potentially affect promoter activity or mRNA conformation/stability

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

Give examples of where SNPs in non-target genes can result in changes in drug activity

A
  • DPYD and 5-FU – Dihydropyrimidine dehydrogenase which detoxifies 5-FU – common T>G reduces its activity resulting in increased activity and toxicity
  • CYP2D6 and tamoxifen – enzyme helps metabolise tamoxifen to endoxifen (active form) – hence mutations may reduce activity
  • XRCC1 and platinum agents – increased activity will mean cells are more able to repair DNA damage – hence will change sensitivity
  • ABC1/MDR1 – alters ability for cell to efflux drug – therefore will impact on cellular concentration and activity
    Note the increased activity of the drug may increase efficacy and/or toxicity and decrease will obviously decrease its efficacy.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Give examples of how somatic mutations in tumours may effect drug response

A

GISTs often have KIT mutations (MAPK/PI3K…) which can be inhibited by imatinib – those with exon 11 mutations respond better to those with exon 9 or no mutation!

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

Discuss the problems that exist with personalised medicine approaches

A
  • Tumours are very heterogenous so only certain populations may respond resulting in tumour evolution and resistance
  • The biopsy quality of tumours is questionable as around 20% cannot be used for biomarker assessment!
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What types of NSCLC are most common and which of these are used by smoking?

A

Non-squamous are 70% - non-smoking whist Squamous are 30% - caused by smoking

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

Discuss how the stage of NSCLC effects what treatment is chosen

A

Stage 0 – removal by surgery
Stage I-IIIA – surgery and adjuvant chemotherapy or chemotherapy and radiotherapy
Stage IIIB – chemotherapy and radiotherapy
Stage IV – SACT – usually involving platinum doublet (cisplatin + pemetrexed)

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

Outline the 3 ways of targeted therapy in NSCLC.

A
  • A target associated with a particular phenotype – EGFR inhibitors in east Asian female, non-smokers, or adenocarcinoma patients (much better response rate)
  • Target a process central for cancer progression – bevacizumab in angiogenesis – alongside immunotherapy and platinum doublet or nintedanib
  • A particular molecular sub-group based on mutation profiling – certain mutations within EGFR
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What were the 1st line EGFR inhibitors?

A

Gefitinib and Erlotinib (competitive reversible inhibitors) 😉 form an acneiform rash - THESE ARE ANILOQUINAZOLINES

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

Why is bevacizumab given alongside immunotherapy?

A

VEGF is an immunomodulator which decreases T-cell activity, hence immunotherapy is used to recover immune activity

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

List a mutation of EGFR that are associated with a)drug sensitivity, b) primary drug resistance and c)mutations associated with acquired resistance

A

a) Exon 19 deletion, L858R – 100x more sensitive to 1st generation than wild-type tumours (70% response rate in those VS very low in WT)
b) Exon 20 insertions
c) T790M

30% of NSCLCs have activating EGFRs

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

How common are EGFR mutations in NSCLC and where do they most commonly occur?

A

Around 30% of patients – with mutations most commonly in exons 19-21 – TK domain (mostly exon 19 deletion of L858R)

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

What is the impact of the L858R mutation?

A

This residue is within the activation loop – which regulates the binding of ATP to the active site – this charge change results in a conformational change which promotes the open/active form – increasing the enzymes activity – but also making it far more sensitive to these first generation inhibitors compared to the wild-type molecule!!
X100 times for sensitive to the 1st generation TKI’s

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

What are the 2nd generation EGFR inhibitors?

A

Afatinib (blocks all EGFR family) and dacomitinib – have improved PFS compared to 1st generation – also competitive inhibitors - but irreversible because they form covalent bonds

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

How often does resistance to the 1st generation EGFR inhibitors occur and describe the impact of one of these secondary mutations

A

Most patients see resistance within 12 months and about 50% have the T790M mutations. This is the gate keeper residue – increased size sterically blocks the entry of the inhibitors but not ATP into the active site.
This also increases its affinity for ATP – increasing oncogenic signalling!!

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

Describe some of the ways EGFR inhibitor resistance occurs (1st generation)

A
  • Parallel RTK hyperactivation via mutations/amplification of ligand or receptor – HER2, MET (can also interact with HER3) -crizotinib
  • Downstream pathway activation
    a. Kras (12/13/61) – in younger, western or smokers with adenocarcinoma
    b. BRAF – in 2% - dabrafenib
    c. PI3KCA/PTEN mutations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Why are 2nd generation EGFR inhibitors less affected by T790M compared to 1st generation?

A

This is because they can still form covalent interactions with C797 which increases its affinity – but still unsuccessful at tolerable doses

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

Name a 3rd generation EGFR inhibitor

A

Osimertinib – binds irreversibly and inhibits the T790M mutants and is less effective against the WT (hence less cytotoxic) – forms a covalent bond with C797 -Can cross the BBB

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

Describe a way we can monitor patients for EGFR secondary mutations?

A

Using liquid biopsies with cfDNA

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

What mutations lead to resistance against Osimertinib?

A

C797S – prevents this covalent bond that is required for its activity!!

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

What other mutations are seen in lung cancer and how can they be targeted (apart from EGFR)?

A

Met – crizotinib
KRAS – smokers – AMG510
BRAF – smokers- dabrafenib
PI3K
ALK – younger, men, non-smokers — gene fusions with EML4 – active receptor tyrosine kinase via dimerization enhancement->MAPK and PI3K – crizotinib (L1196M – resistance), 2nd generation alectinib (resistance via G1202R), 3rd generation loralatinib (can across BBB)
ROS-1 – RTK - fusion gene ->PI3K/MAPK -can use crizotinib (G202R4/gatekeeper resistance) or Entrectinib.

26
Q

What treatments are available for NSCLC patients who have no mutations which are associated with targeted therapy?

A

75% of patients Immune checkpoint inhibitors like pembrolizumab/nivolumab (anti-PD-1 – prevents interaction with PD-L1) – prevent T-cell exhaustion/inactivation. Atezolizumab is anti-PDL1 – and can potentially work in those regardless of PD-L1
Can be used in mutation positive with TKIs/bevacizumab/platinum chemotherapy

27
Q

Why is treatment of NSCLC so difficult?

A
  • Most patients present with late stages of the disease – already very advanced
  • There are lots of pathways which can be switched between for resistance
  • Brain metastases is very common – and many molecules don’t cross the BBB
28
Q

What are the risk factors for HCC?

A
  • Underlying chronic liver disease/cirrhosis – like haemochromatosis – cirrhosis – regeneration and inflammation which can promote tumorigenesis
  • Obesity, drug use, anabolic steroids, drinking and smoking
  • Viruses – Hepatitis B/C
29
Q

How do hepatitis B and C cause cancer?

A

Hepatitis B
Responsible for around 80% of cases – increases risk-100-fold. DSDNA virus encoding a weakly oncogenic protein X
Hepatitis C
Transfected via blood – increases risk 20-fold – Positive strand- RNA virus can cause cancer in many ways:
- X protein can induce DNA transcription alterations – BCL-2 and miRNAs (that regulate HIF-1-alpha/MMPs?) promoting proliferation
- Can induce DNA damage like p53 (G->T mutation) as work in synergy with aflatoxin B1
- Viruses induce chronic inflammation – GFs like PDGF or IL’s and can also promote angiogenesis (infecting endothelial cells)
-

30
Q

How can HCC be prevented?

A
  • Hepatitis B vaccination
  • Infection control (screening transfusions)
  • Anti-viral treatments
  • Screening at risk cohorts
  • Screening patients with chronic HCV/HBC
  • Lifestyle changes – drinking/smoking/drug use
31
Q

How is liver cancer staged?

A

Using the Barcelona clinic liver classification system
condition.
- Stage 0 – single nodule and ≤ 2cm
- Early stage (A)- single node or ≤ 3 nodules ≤ 3cm and ECOG0
- Intermediate stage (B)– Multinodular but ECOG 0
- Advanced Stage – Portal Invasion N1, M1, ECOG 1-2
- Terminal Stage (D)
Can also look at increase alpha-fetoprotein in the blood

32
Q

How is stage 1 HCC treated?

A

This stage is operable which means they have good survival rates. There are 3 types of surgery:
Operable disease – completely removed the liver with a transplant if possible (confined disease)
Partial resection – removes entire cancerous lesion (liver can re-generate)
Ex-vivo liver resection – removal of disease area and reimplantation of remaining tumour-free liver

33
Q

How is stage B HCC treated?

A

Transarterial chemoembolization – TACE – where chemotherapy (doxorubicin/cisplatin) is injected via a catheter into the hepatic artery to cut off blood flow causing ischemia and a high level of drug in the liver (minimises toxicity)
Bland/Transarterial embolization – TAE – particles injected via catheter to block hepatic blood flow
Transarterial chemoembolization with drug-eluting beads – DEB-TACE – beads block blood flow and elute chemotherapy
Necrosis is never complete, can rely on portal blood supply and angiogenesis can promote tumour survival.

34
Q

Describe the many ways in which stage C HCC is treated

A

HCC associated with many RTK hyperactivations as well as angiogenesis – because of this many pan-RTK inhibitors are used.
- Sorafenib – RAF1, BRAF, VEGFR2, PDGFRβ – MAIN THERAPY
- Lenvatinib – VEGFR1-3, PDGFRα, FGFR1-4 and RET
- Regorafenib – VEGFR1-3, BRAF V600, PDGFR, FGFR (after progression on sorafenib – attacks the pathways that may lead to resistance
- Cabozanitinib – MET, VEGFRs and KIT – targets MET+
Resistance is common in these therapies as they can switch signalling to other RTKs and they are associated with severe toxicities like hypertension, diarrhoea or hand-foot syndrome
Ramucirumab – VEGFR2 antibody
Atezolizumab – PDL1 antibody with bevacizumab
Nivolumab – PD1 inhibitor

35
Q

What are the reasons why brain tumours are difficult to treat?

A
  • Tumours are localised in the brain so difficult to remove surgically
  • Limited capacity of the brain to repair itself
  • Disrupted blood supply can reduce drug delivery and may lead to accumulation of fluid around the tumour – intracranial hypertension
  • Therapeutic insensitivity
  • Treatment neurotoxicity
  • They are able to activate many different pathways leading to resistance as well as significant tumour heterogeneity!
36
Q

What cells do gliomas originate from?

A

Astrocytes, oligodendrocytes, Schwann cells]]] glial cells - non-neuronal cells that maintain homeostasis (neurotransmitter/ions), maintain the blood-brain barrier and produce myelin

37
Q

Discuss the mechanism of temozolomide and what is a predictive factor for its efficacy?

A

Temozolomide is an DNA damaging agent which can help add a methyl-group to guanosine bases at position O6- this can lead to a substitution as it forms a base-pair with T.
The methylation status of methylguanine DNA methyltransferase is key to this as this enzyme carries out the removal of this methyl group – hence promoter methylation and gene silencing will lead to a better response to treatment.
Used in brain cancer

38
Q

What mutations are common in glioblastomas?

A
  • RTK (EGFR/PDGFR), Ras, PI3K in 88% (as well as PTEN loss)
  • P53 altered in 87% (or CDKN2A inactivation- inhibitor of CDK INHIBITOR 2A that regulates P16 and P14 which are TS, inhibiting CDK4/6 and activating p53 respectively)
  • Rb inactivation in 77%
39
Q

What targeted therapies exist for glioblastoma treatment?

A

Targeting EGFR – common EGFRVIII mutation/loss of exons 2-7 (ligand-binding domain) or activate in autocrine manner
1st generation gefitinib and erlotinib not associated with good outcome due to cross talk with IGFR and poor intratumoural levels/pharmacology
Antibodies like cetuximab are too large to cross the BBB
2nd generation TKIs under investigation like lapatinib
PDGFR – itself and ligand overexpressed
Imatinib show poor efficacy as well as antibody ramucirumab
RAS
Farnesyltransferase inhibitors like lonafarnib as well as MEK inhibitors like sorafenib
PI3K/Akt
Perfosine – poor toxicity

40
Q

Describe the approaches of targeted therapy in melanoma

A

BRAF inhibition – mutations in around 60%
BRAF is a main component of MAPK signalling – the majority of cases are associated with the V600E mutations ->MAPK pathway but also NF-κB and A/C-RAFs
Inhibitors like vemurafenib
Resistance occurs via RTK hyperactivation to regulate other pathways are mutations in MEK/RAS to hyperactivate the MAPK pathway
Immunotherapy
The anti-CTLA4 (0n the T-cells) ipilimumab to prevent interaction with CD80/86 (B7 receptor) involved in T-cell inactivation
Same for Pembrolizumab (anti-PD1) and nivolumab (anti-PD1) – T-cells

41
Q

Describe the activation of the ERK pathway

A
  • RTK-ligand binding leads to receptor dimerization, phosphorylation and activation
  • GRB2 binding helps bind the GEF SOS which can activate Ras (via transformation from inactive GDP-from to GTP-form)
  • This can then phosphorylate and activate RAF->MEK->ERK -> expression of cyclin D1
42
Q

Explain why the combination of immunotherapy and targeted therapy in melanoma might be a good approach

A

Because immunotherapies see improvements later on in treatment regimes, whilst targeted therapies are preferential short-term

43
Q

Describe the two types of ovarian cancer and which type of cell is usually responsible.

A

Type I – chemoresistant but low grade – KRAS/PI3K/ARD1A (part of SWI/SNF of chromatin remodelling complex)
Type II – chemo sensitive and usually associated with BRCA/P53 mutations lots of different mutations in small amounts - meaning its difficult to target all (IGFR/RAS/Notch)
Both associated with epithelial cells

44
Q

Describe the type of approaches of targeted therapy in ovarian cancer.

A

Angiogenesis – inhibitors like cediranib and bevacizumab
PARP inhibitors in those with BRCA mutations
MEK inhibitors like selumetinib due to mutations in BRAF/KRAS and PTEN or BRAF/VEGFR inhibitor sorafenib

45
Q

Describe the concept of synthetic lethality

A

Were the loss of function of two genes (usually involved in a similar process) together, but not individually will lead to cell death

46
Q

Describe how PARP inhibitors work in those with BRCA mutations

A

BRCA1//2 play a role in double-strand repair by helping recruit molecules like RAD51 to help repair the DNA using the second copy of the chromosome in a process known as homologous recombination. Tumour cells that have mutations in these genes are less able to repair DNA as they instead use an error prone process – NHEJ which can often lead to cell death
PARP is an enzyme that helps repair single-strand breaks via BER – recruiting molecules like XRCC1 to the damage site. The use of competitive PARP inhibitors like olaparib will prevent single strand breaks from being repaired in ALL cells. Normal cells have functional HR pathway so will be able to repair the subsequent double-strand breaks so will be largely unaffected – however tumour cells will have to use NHEJ – which will lead to tumour cell death
Resistance is quite common though through restoration of HR or Increased PARP activity
Is a possibility can be used in patients with mutations in other HR genes like PALB2 or RAD51 which cumulatively constitute a significant proportion of patient

47
Q

Explain the ways in which ovarian cancer cells can become resistant to chemotherapies

A

BRCA1/2 can affect sensitivity to platinum based agents as they help repair any DNA damage the agent produces
Signalling pathways like PI3K up-regulated (Either via RTK or intracellular kinase)
Efflux proteins like p-glycoprotein

48
Q

Describe the possible treatments for TNBC?

A

Immunotherapies like pembrolizumab or atezolizumab
PARP inhibitors like olaparib in those with BRCA mutations
ADCs – like Sacituzumab govitecan – Trop2 (expressed by lots of TNBC) and a topoisomerase inhibitor
PI3K inhibition like ipatasertib or Capivasertib

49
Q
  1. Describe the two functions of the pancreas and what two cell types are involved
A

Exocrine – regulate digestion
Acinar cells secrete chymotyprsinogen, lipases and amylases
Ductal cells secrete bicarbonate to regulate the pH
Endocrine – regulates glucose metabolism
α/β cell secrete glucagon and insulin
PP and Δ cells regulator secretory activity of other cells through pancreatic polypeptide which inhibits enzyme secretion and somatostatin effects neurotransmitters

50
Q

What are the risk factors associated with pancreatic cancer

A
  • Age
  • Smoking/alcohol/poor diet
  • Chronic pancreatitis
  • H.pylori infection
  • Genes like APC/BRCA/P53
51
Q

In pancreatic cancer what is PANIN?

A

Pancreatic intraepithelial neoplasia – ERBB/KRAS….INK4….P53 mutations….Telomerase

52
Q

What types of non-targeted therapy are used in pancreatic cancer

A

Whipple procedure – not many can have this can procedure is complex
Chemotherapy like carboplatin, 5-FU and docetaxel
Radiotherapy – brachytherapy

53
Q

What kinds of targeted therapy exist in pancreatic cancer?

A

MEK inhibitor pimasertib to inhibit those with RAS mutations which are an early event and found in many pancreatic cancer patients
Anti-IGFR antibody dalotuzumab – hyperactivation is also common and autocrine/paracrine activity
SMO inhibitors like saridegib

54
Q

Explain how the hedgehog pathway can promote pancreatic cancer progression

A

Patch receptor inhibits SMO which will lead to the cos-2 complex binding to Gli, cleaving it so it cannot enter the nucleus
SHH binding to patch relinquishes this inhibition meaning the Cos2 complex is bound to Smo. Gli can then enter the nucleus and transcribe genes like mTOR, EGF and TGFβ.
SHH can be overexpressed in cancer due to Ras hyperactivity and TAFs

55
Q

Describe the risks associated with CRC?

A
80% non-inherited 
-	Age
-	Type II diabetes 
-	Obesity 
-	Smoking/alcohol 
-	High fat/red meat
20% hereditary 
-	FAP – APC mutations 
-	Lynch syndrome – Mut genes MMR
56
Q

What is the most common early event in CRC development?#

A

Loss of APC – promotes beta-catenin signalling.

Can be followed both other changes like loss of p53 and activation of KRAS/RAF/PI3K as well as epigenetic changes

57
Q

What are the four stages of Duke staging in CRC?

A
  • A – confined to mucosa
  • B -Perforates outer serosa
  • C – lymph nodes close to bowel
  • D – distant metastasis
58
Q

Describe the non-targeted chemotherapeutic approaches in CRC

A
  • The first agent will likely be capecitabine (anti-metabolite ->5-FU) +/- oxaliplatin (XELOX) – DNA strand breaks, or 5-FU (thymidyl synthase inhibitor) + folonic acid (enhance 5-FU toxicity as stabilises enzyme complex) +/- oxaliplatin (FOLFOX)
  • 2nd line therapy is 5-FU + folonic acid + irinotecan (FOLFIRI) topoisomerase inhibitor or irinotecan monotherapy if FOLFOX 1ST line
59
Q

What are the targeted therapy agents used in CRC?

A

EGFR is overexpressed in 80% of patients
Cetuximab and panitumumab – prevent ligand binding and dimerization (G1 arrest) – have shown potential with FOLFOX and FOLFIRI
Anti-angiogenic agents
Bevacizumab – binds to VEGF-A- again can be used alongside FOLFIRI – resistance occurs via KRAS/BRAF mutations or via chronic treatment resulting in an angiogenic switch
Aflibercept is an anti-VEGF recombinant protein attached to an FC-region
Immune checkpoint inhibitors
MMR proteins (MSH2/6 mutation or MLH1 promoter methylation) can lead to mutations resulting in abnormal protein production that trigger T-cell infiltration
Pembrolizumab and nivolumab (PD-1 antibodies) can activate T-CELLs as many of these tumour cells will also produce PDL1 to cause T-cell exhaustion

60
Q

Describe the mechanisms CRC patients have to become resistance to EGFR therapies.

A

Non-genetic:
- Increased EGFR//EGF expression
- Altered binding affinity of EGFR ligands or extracellular domain mutations
- Dysregulation of EGFR internalisation
- Overexpression of other receptors like HER2/MET
- PI3K/AKT up-regulation
Genomic
- BRAF mutations – V600E – co-treat with Encorafenib – really important to determine appropriate therapy as not-responsive to EGFR treatment
- RAS mutations like G12C – is REALLY IMPORTANT to determine if chemo/EGFR therapy used!!!! Mutants do not respond to EGFR therapy - hence sequencing is commonly used to detect

61
Q

What is Lonsurf?

A

The use of the nucleoside analogue trifluridine and the thymidine phosphorylase inhibitor tipiracil -given after failure of EGFR inhibitors in CRC