BN - Precision medicine and personalised cancer immunotherapies Flashcards

(16 cards)

1
Q

Q1: What is precision medicine and how does it differ from personalised medicine? (4)

A
  • Precision medicine: Tailors treatment based on genetic, environmental, and lifestyle differences across patient subgroups.
  • Focuses on population-level characteristics and molecular phenotypes (e.g. genome, proteome, metabolome).
  • Personalised medicine: Tailored to an individual patient’s unique profile.
  • While often used interchangeably, personalised is more individualised than precision medicine.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Q2: What technological advancements are enabling precision medicine? (3)

A
  • Omics technologies: Genomics, transcriptomics, proteomics, metabolomics, lipidomics.
  • Especially pharmacogenomics: Studies how genetic makeup influences drug response.
  • Use of next-generation sequencing enables rapid identification of disease-causing mutations.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Q3: What are some clinical applications of precision medicine? (5)

A
  • Gene therapy for spinal muscular atrophy, cystic fibrosis, and sickle cell disease.
  • Targeted drugs for HER2+ breast cancer (e.g., trastuzumab).
  • BCR-ABL inhibitors (e.g., imatinib) for CML.
  • Anti-CD20/CD19/CD79b mAbs in lymphomas.
  • Over 200 FDA-approved drugs now include pharmacogenomic label info.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Q4: How does precision medicine improve cancer care? (3)

A
  • Helps stratify patients based on molecular phenotype to optimise therapy.
  • Targets known molecular pathways or tumour locations.
  • Improves treatment precision via proteogenomic stratification (integrates genomic + proteomic data).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Q5: How does the immune system naturally respond to tumours? (6)

A
  • Innate immunity: Rapid, non-specific response; presents antigens to adaptive system.
  • Adaptive immunity: Slow, specific, memory-forming response.
  • Immune system normally recognises and removes abnormal cells (immune surveillance).
  • Tumours evade detection via weak immune responses, loss of MHC, or being immunocompromised.
  • Tumour-associated antigens (TAAs) are often weak and hard to target.
  • T cell responses are often weak or exhausted, allowing tumour survival.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Q6: What makes tumour cells recognizable or invisible to the immune system? (5)

A
  • Some tumours contain microbes or arise from viruses (foreign signal).
  • Many lose MHC (HLA) display, making them “invisible” to T cells.
  • Tumours express TAAs (e.g., NY-ESO-1, MAGE) – abnormal in time, location, or amount.
  • Tumour-specific antigens (TSAs) arise from mutations and are truly foreign.
  • These differences determine how “self” or “non-self” a tumour appears immunologically.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Q7: How do lymph nodes contribute to anti-tumour immunity? (5)

A
  • Serve as immune hubs that filter lymph fluid for antigens.
  • B cells in the cortex respond to matching antigens via BCRs.
  • B cells interact with helper T cells → undergo class switching and differentiation.
  • T cells and dendritic cells are drawn to nodes via CCR7 signaling.
  • Lymph nodes facilitate T and B cell activation and coordinate immune attack.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Q8: How are tumour antigens presented to T cells via MHC pathways? (3)

A
  1. Endogenous (MHC I):
    • Intracellular proteins degraded by proteasome, peptides loaded onto MHC I via TAP, presented to CD8+ T cells.
  2. Exogenous (MHC II):
    • Engulfed proteins are processed in lysosomes, loaded onto MHC II via HLA-DM, presented to CD4+ helper T cells.
  3. Cross-Presentation:
    • APCs can present exogenous proteins on MHC I, crucial for initiating cytotoxic T cell responses to tumours.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Q9: What are the major immunotherapy strategies for cancer? (5)

A
  • Immune checkpoint inhibitors – e.g., anti-PD-1/PD-L1 to boost T cell activity.
  • Adoptive T cell therapy – e.g., CAR-T cell therapy.
  • Monoclonal antibodies – e.g., anti-TAA antibodies.
  • Therapeutic vaccines – deliver TAAs to stimulate immunity.
  • Immunomodulators – e.g., cytokines, BCG, angiogenesis inhibitors.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Q10: What is T cell exhaustion and how is it reversed by checkpoint inhibitors? (4)

A
  • Acute stimulation triggers response, followed by downregulation via PD-1/PD-L1 to avoid autoimmunity.
  • Chronic stimulation in tumours leads to T cell exhaustion (upregulation of PD-1, Tim-3, LAG-3).
  • Exhausted T cells are ineffective against tumours.
  • Checkpoint inhibitors block PD-1/PD-L1, reinvigorating T cells to attack cancer.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Q11: What is the relationship between mutational burden and checkpoint inhibitor efficacy? (4)

A
  • High TMB increases the likelihood of neoantigen formation.
  • More mutations = more T cell-recognisable targets.
  • Cancer types with high TMB (e.g., melanoma, lung cancer) respond better to PD-1 blockade.
  • TMB is now used as a biomarker for immunotherapy suitability.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Q12: What types of gene mutations contribute to tumour formation? (4)

A
  • Driver mutations: Promote tumour growth.
  • Passenger mutations: Accidental, neutral.
  • Proto-oncogenes: Normal growth genes; when mutated → oncogenes (overactive).
  • Tumour suppressor genes & DNA repair genes: When inactivated → loss of growth control and mutation accumulation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Q13: What causes mutations leading to cancer and how do they affect proteins? (5)

A
  • Causes: UV, smoke, viruses, alcohol, cell division errors, inheritance.
  • Missense mutations → amino acid changes = neoantigens.
  • Mutated p53 = common loss-of-function mutation, promotes tumour formation.
  • Mutant proteins can disrupt cell growth regulation.
  • If recognised as “non-self”, these proteins can be T cell targets.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Q14: How do tumour-associated antigens (TAAs) differ from tumour-specific antigens (TSAs)? (4)

A

TAAs:

  • Overexpressed in tumours but also found in healthy tissues.
  • Risk of off-target effects in therapy.

TSAs (Neoantigens):

  • Result from unique mutations in tumours.
  • Not found elsewhere in the body → high specificity, low side effects.

P53 can act as either, depending on context.

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

Q15: How do T cells recognise and respond to tumour mutations? (4)

A
  • T cells detect peptides presented by MHC molecules.
  • Tumours can produce abnormal or developmental proteins (CTAs, splice variants).
  • Challenge: TCRs do not reveal the identity of target peptides.
  • Identifying true neoantigens remains difficult; only 5–20 clonotypes usually active per tumour.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Q16: What are the main takeaways from precision cancer immunotherapy? (6)

A
  • Omics enables stratification and targeted treatment.
  • Mutations drive tumour formation and define therapy responses.
  • Checkpoint blockade works best in high-TMB tumours.
  • Neoantigens from non-synonymous mutations are ideal immune targets.
  • Effective therapy requires recognition of MHC-bound peptides.
  • Precision targeting of mutated proteins is key to advancing safe, potent immunotherapies.