library(tidyverse)
library(knitr)

WEEK 1

L1.1 INTRODUCTION TO GENETICS

Objective 1. Explain the molecular differences between DNA and RNA and their roles in the cell

Core differences

Feature DNA RNA
Sugar Deoxyribose, no 2′-OH Ribose, 2′-OH present
Bases A, T, G, C A, U, G, C
Strandedness Usually double-stranded helix Usually single-stranded*
Stability More chemically stable Less stable
Location Stays in nucleus Moves throughout cell
Primary role Long-term information storage Multiple roles in expression

Slides note exceptions such as dsRNA viruses and structured functional RNAs.

Roles tied to those differences

  • DNA stores stable information in the nucleus.
  • RNA performs multiple expression roles:
    • mRNA carries coding sequence.
    • rRNA forms ribosomal structure.
    • tRNA brings amino acids.
  • Only a small fraction of the genome encodes proteins; the rest still needs packaging and regulation.

Mermaid figure

flowchart LR
  DNA[(DNA, nucleus)] -->|transcription| mRNA[mRNA]
  mRNA -->|translation at ribosome| Protein[Protein]
  rRNA[rRNA] --- Ribosome
  tRNA[tRNA + amino acids] --> Ribosome

TLDR (Obj 1) DNA is stable, double-stranded, nuclear information storage. RNA is less stable, mobile, and supports expression as mRNA, rRNA, and tRNA.


Objective 2. Explain the steps through which DNA is expressed as protein

Stepwise overview

  1. Transcription DNA → pre-mRNA.
  2. Processing Remove introns, join exons, add cap and poly-A tail.
  3. Export mRNA leaves nucleus.
  4. Translation Ribosome reads codons; tRNAs deliver amino acids → protein.

Alignment table

Stage Input Key event Output
Transcription DNA RNA polymerase copies gene region pre-mRNA
Processing pre-mRNA Splice introns, add cap/tail mature mRNA
Export mRNA Nuclear pore transit cytosolic mRNA
Translation mRNA, tRNA, rRNA Peptide synthesis at ribosome polypeptide → protein

Codon logic

Three nucleotides = one codon = one amino acid.

TLDR (Obj 2) DNA → pre-mRNA → processed mRNA → translated codons → polypeptide → protein.


Objective 3. Explain how these steps are regulated

1) Access to DNA

  • Chromatin packaging regulates transcription factor and polymerase access.

2) Transcriptional output

  • Cells vary transcript number to vary protein amount.

3) RNA processing and stability

  • Capping, splicing, and tailing determine how much mRNA survives and is translated.

Flow summary

flowchart TD
  A[Chromatin state] --> B(Transcription)
  B --> C[mRNA processing]
  C --> D[Nuclear export]
  D --> E[Translation rate]
  E --> F[Protein level]

TLDR (Obj 3) Regulation occurs at chromatin access, transcription level, and RNA processing. The goal is correct protein amounts at correct times.


Objective 4. Apply key terms (slide 4) to describe the amount and organisation of DNA

Key terms with definitions

Term Definition Usage
Nucleotide Sugar + phosphate + base Three nucleotides form a codon.
Gene DNA sequence encoding RNA/protein HBB gene encodes beta-globin.
Allele Alternate gene version ΔF508 is an allele of CFTR.
Locus Physical chromosomal position HBB locus is on 11p15.5.
Genome Full DNA content Human haploid genome ≈ 3.2 Gb.
Chromosome One DNA molecule with proteins Humans have 23 pairs.
Chromatin DNA + proteins (packaging) Open chromatin increases access.
Nucleosome DNA wrapped around histone octamer Controls access.
Ploidy Sets of chromosomes Somatic cells are diploid.
Haploid One set Gametes are haploid.
Diploid Two sets Somatic cells.
Karyotype Chromosome number/appearance 46, XX or 46, XY.
mtDNA Circular mitochondrial genome ~16.5 kb, maternal.
Operon Cluster under one promoter Common in bacteria.
Plasmid Small circular DNA in bacteria Often carries resistance genes.

Amount of DNA

  • Haploid = ~3.2 Gb
  • Diploid = ~6.4 Gb
  • mtDNA = 16.5 kb, many copies/cell
  • 46 chromosomes in somatic cells, 23 in gametes

Organisation hierarchy

nucleotide → double helix → nucleosome → chromatin fibre → loop/domains → chromosome → genome

flowchart TD
  A[Nucleotide] --> B[Double helix]
  B --> C[Nucleosome]
  C --> D[Chromatin fibre]
  D --> E[Loops]
  E --> F[Chromosome]
  F --> G[Genome]

Example applications

Human somatic cell
  • Diploid, 46 chromosomes, 6.4 Gb nuclear DNA + mtDNA.
  • Packaged as: helix → nucleosome → chromatin → chromosome.
Human gamete
  • Haploid, 23 chromosomes, 3.2 Gb DNA.
Bacterium
  • One circular chromosome, plasmids common.
  • No nucleus; operons common.

Prokaryote vs eukaryote summary

Feature Prokaryote Eukaryote
Genome Circular chromosome, plasmids Multiple linear chromosomes
Packaging Nucleoid proteins Histones and nucleosomes
Compartment Nucleoid Nucleus
Gene organisation Operons Mostly monocistronic
mtDNA No Present

TLDR (Obj 4) Human somatic cells are diploid with 46 chromosomes and ~6.4 Gb DNA. DNA is organised from nucleotides → helix → nucleosomes → chromatin → chromosomes → genome. Bacteria have circular DNA, plasmids, operons, and nucleoid structure.


L1.2 GENETICS & HEREDITARY

Objective: Explain how different alleles give rise to different traits

Key terms

  • Allele, genotype, phenotype, dominant, recessive, homozygous, heterozygous.

Logic

Genotype Molecular state Phenotype
AA Dominant protein present Dominant trait
Aa Dominant protein present Dominant trait
aa No dominant product Recessive trait

Punnett logic

Aa × Aa →

  • 25% AA
  • 50% Aa
  • 25% aa
  • Phenotype ratio 3:1
flowchart LR
  P1[A or a] --> Offspring
  P2[A or a] --> Offspring

TLDR Dominant alleles show with 1 or 2 copies. Recessive traits require 2 copies. Punnett squares convert parent genotypes into offspring probabilities.


Objective: Describe autosomal vs sex-linked inheritance patterns

Core differences

Feature Autosomal Sex-linked
Chromosomes 1–22 X or Y
Copies in XY Two One X, one Y
Examples Widow’s peak Haemophilia, red-green colour blindness
Pitfall Misjudging heterozygotes Forgetting XY is hemizygous

Rules

  • Autosomal recessive: aa affected; carriers common; often skips generations.
  • Autosomal dominant: AA/Aa affected; appears every generation.
  • X-linked recessive: mostly males; no male→male; daughters can be carriers.
  • X-linked dominant: affected father gives to all daughters, no sons.
  • Y-linked: only males, father → all sons.

Worked example: X-linked recessive

XAXa × XAY →

  • Sons: 50% affected
  • Daughters: 50% carriers
flowchart TB
  A[XA] --> D1[XAXA]
  A2[Xa] --> D2[XAXa]
  B[XA] --> S1[XAY]
  C[Y] --> S2[XaY]

TLDR Autosomal traits act the same in XX and XY. X-linked traits differ because XY has only one X. Male→male transmission excludes X-linked patterns. Affected fathers passing only to daughters suggests X-linked dominant.


Objective: Read and interpret pedigrees

What to check

  • Pattern by sex and generation
  • Male→male transmission
  • All daughters affected?
  • Skips or clusters?
  • Maternal-only transmission
  • Consanguinity

Patterns

Mode Hallmarks
Autosomal dominant Present every generation
Autosomal recessive Unaffected parents → affected children
X-linked recessive Mostly males; no father→son
X-linked dominant All daughters of affected father
Y-linked Father → all sons
Mitochondrial Affected mother → all children

Carrier probability (AR example)

Unaffected sibling of affected: 2/3 carrier.

TLDR Scan gender pattern, male→male lines, father→daughter patterns, skipped generations, and maternal transmission. Match to inheritance mode and compute genotype risks.


L1.3 PROPERTIES OF NUCLEIC ACIDS

Objective 1. Compare and contrast the structure, properties and roles of DNA and RNA

Structure and chemistry

Feature DNA RNA Why it matters
Sugar Deoxyribose Ribose RNA less stable
Bases A,T,G,C A,U,G,C U replaces T
Pairing A=T, G≡C A=U, G≡C Stability differs
Strandedness Double-stranded Single-stranded, folds Functional shapes
Direction 5′→3′ 5′→3′ Same chemistry
Charge Negative backbone Negative backbone Binds basic proteins
Spectrophotometry A260 = 50 ng/µL dsDNA A260 = 40 ng/µL RNA Prac relevance

Higher-order organisation

  • DNA stored as chromatin; euchromatin accessible, heterochromatin condensed.

Roles

  • DNA stores genetic information.
  • RNA runs expression: mRNA, rRNA, tRNA, regulatory RNAs.

TLDR DNA is stable and double-stranded in chromatin. RNA is less stable, single-stranded, and functional in expression.


Objective 2. Explain transcription, translation, and RNA processing

2a. Transcription

  • Template strand guides RNA; coding strand matches RNA except U for T.
  • RNA polymerase binds promoter and synthesises RNA 5′→3′.

2b. Translation

  • Ribosome reads codons; tRNAs match amino acids; ends at stop codon.

2c. Processing

  • 5′ cap, splicing, 3′ poly(A) tail → mature mRNA.

TLDR DNA → pre-mRNA → capped/spliced/polyadenylated → mRNA → ribosome → protein.


Objective 3. Describe features of the genetic code

Essentials

  • Triplet, non-overlapping, comma-less, degenerate, unambiguous, near-universal.
  • Start = AUG; Stops = UAA, UAG, UGA.

Wobble

  • Flexibility at third position allows fewer tRNAs.

Mutation types

  • Synonymous, missense, nonsense, frameshift.

TLDR 64 triplet codons specify 20 amino acids + stops. Wobble explains degeneracy.


Objective 4. Appreciate how different molecules bind DNA

Binding mechanisms

  • Electrostatic (histones).
  • Major/minor groove recognition (TFs).
  • Intercalation (aromatic drugs).
  • Covalent crosslinking (cisplatin).
  • Topoisomerase interactions.

Medical examples

  • Cisplatin, doxorubicin, actinomycin D.

TLDR DNA binding uses charge interactions, groove readout, intercalation, or covalent adducts. These changes alter transcription, replication, or structure.

WEEK 2

L2.1 MUTATIONS

Objective: Understand the different types of mutations

What a mutation is

  • A mutation is a permanent change in DNA sequence. It is copied during replication and can be germline or somatic. Not all mutations are harmful; some are neutral or beneficial.

Where mutations occur

  • Scale ranges from single-base changes to large chromosomal segments.

DNA-level classes

Class Definition Typical consequence
Base substitution One base replaced by another Silent, missense, or nonsense
Insertion One or more bases added Frameshift if not multiple of 3
Deletion One or more bases removed Frameshift if not multiple of 3
Inversion Segment flipped Local disruption or altered regulation
Duplication Segment copied Dosage increase or frame disruption
Translocation Segment moved Fusion genes, mis-regulation
Mobile element insertion Transposon/retroelement insertion Disrupts coding or regulatory DNA

Protein-level classes

Class Definition Notes
Silent Codon change, same amino acid Can affect mRNA kinetics
Missense Codon change to different amino acid Severity depends on chemistry and position
Nonsense Codon to premature stop Truncation or no protein
Frameshift Indel not in multiples of 3 Alters downstream codons, usually severe

Functional descriptors

  • Null, leaky, silent.

Reading-frame logic

  • Three reading frames per strand.
  • Single-base indels shift the frame.
  • Multiples of 3 preserve it.
  • Base change to stop codon yields nonsense mutation.
flowchart TD
  A[Normal codons] --> B[+1 insertion]
  B --> C[Frameshift → early stop]
  A --> D[+3 insertion]
  D --> E[Frame preserved]
  A --> F[Substitution]
  F --> G[Nonsense/Missense]

Germline vs somatic

  • Germline mutations are heritable.
  • Somatic mutations are not inherited and drive cancer evolution.

Context that modulates impact

  • Site (active site, structural region).
  • Environment (temperature sensitivity).
  • Chemistry of replaced residue.

TLDR – Types of mutations DNA-level: substitution, indels, inversion, duplication, translocation, mobile element insertion. Protein-level: silent, missense, nonsense, frameshift. Frameshifts arise from non-triplet indels and usually abolish function. Impact depends on site, context, and residue chemistry.


Objective: Describe the effect of mutations on RNA and protein sequence and function

DNA → RNA → protein logic

DNA change RNA effect Protein effect Functional outcome
Synonymous substitution Same amino acid, may alter stability or translation speed None Usually neutral, subtle kinetic effects
Missense substitution Different codon One amino acid changed Neutral to severe; includes leaky, LOF, GOF, dominant-negative
Nonsense substitution Premature stop Truncated protein or none Often NMD if stop is >50–55 nt upstream of last junction
In-frame indel Frame preserved Adds/removes residues Motif disruption
Frameshift indel New codons downstream Radical sequence change, early stop Usually severe loss-of-function
Splice-site mutation Exon skipping, intron retention, cryptic splicing In-frame or frameshift outcomes Often NMD or dysfunctional isoforms
Promoter/enhancer mutation Changes transcription No coding change Altered expression
UTR mutations mRNA stability/translation changes No coding change Up/downregulation
CNV (deletion/duplication) Dose change Less or more protein Haploinsufficiency, overexpression
Dominant-negative missense Normal RNA Mutant subunit poisons complex Worse than simple LOF

Chemistry and protein fate

  • Charge swaps, hydrophobic↔︎polar, proline disruptions.
  • Misfolding → chaperones → proteasome.
  • Truncation → mislocalisation.

NMD linkage

flowchart TD
  DNA --> pre[pre-mRNA]
  pre --> spliced[mRNA]
  spliced -->|PTC upstream| NMD
  spliced -->|no PTC| Translation

TLDR – Effects of mutations DNA changes propagate through RNA processing to protein. Nonsense and frameshift often trigger NMD. Splice-site and UTR changes alter expression. Outcomes: neutral, partial loss, complete loss, gain of function, or dominant-negative.


Objective: Understand how errors in replication result in substitutions

Origin during replication

  • Tautomeric shifts → misincorporation.
  • Proofreading failures → mismatch persists.
  • Mismatch repair failure → mismatch becomes fixed mutation.

Chemical lesions

  • Deamination C→U, 5mC→T → G·C→A·T transitions.
  • 8-oxo-G pairs with A → G·C→T·A transversions.
  • Alkylation (O6-methyl-G→T).
  • Base analogs (5-BU).

Transition vs transversion

  • Transition: purine↔︎purine, pyrimidine↔︎pyrimidine.
  • Transversion: purine↔︎pyrimidine.

Replication slippage

  • At repeats → indels (MMR repairs loops).

Flow

flowchart LR
  Fork --> Mis[Mispair]
  Mis -->|proofread| Correct
  Mis -->|escape| MMR
  MMR -->|missed| NextRound
  NextRound --> Sub[Fixed substitution]

TLDR – Replication errors Mispaired bases from rare tautomers or damaged DNA cause mismatches. Proofreading and MMR correct most. Unrepaired mismatches → permanent transition or transversion.


L2.2 DETECTION OF MUTATIONS

Objective 1. Appreciate the different methods for detecting large versus small mutations

Large-scale variants

Method Detects Size Readout Example
Karyotyping Aneuploidy, large rearrangements >5 Mb G-banded metaphase spreads Trisomy 21
FISH Targeted deletions/translocations >100–200kb Fluorescent probe signals BCR-ABL1 in CML

Small-scale variants

Method Detects Principle Output
RFLP Changes in restriction sites Enzyme cutting + gel Fragment pattern differences
PCR Presence/absence, small indels Locus amplification Amplicon detection
Sanger sequencing Base-level variants ddNTP chain termination Exact sequence
flowchart LR
  Q[Clinical question] --> S{Variant size?}
  S -->|Large| Karyotype
  S -->|Sub-Mb| FISH
  S -->|1–1000 bp| PCR
  S -->|Base-level| Sanger

TLDR – Detection Karyotype for Mb-scale events. FISH for targeted sub-Mb. PCR/RFLP for small variants. Sanger for exact base calls.


Objective 2. Understand the molecular principles of PCR and Sanger sequencing

PCR

  • Components: template, primers, polymerase, dNTPs.
  • Denaturation → annealing → extension.
  • Exponential amplification.
flowchart TD
  D[dsDNA] --> S[ssDNA]
  S --> P[Primers]
  P --> E[Extension]
  E --> Copies

Sanger sequencing

  • Template + primer + dNTPs + fluorescent ddNTPs.
  • ddNTPs terminate extension.
  • Capillary electrophoresis resolves fragments.
flowchart LR
  Template --> Mix[dNTP + ddNTP]
  Mix --> Term[Terminated fragments]
  Term --> CE[Capillary]
  CE --> Seq[Sequence trace]

PCR vs Sanger

Feature PCR Sanger
Goal Detect/amplify Read sequence
Output Amplicon Base-by-base
Sensitivity Very high High
Use Presence/absence Variant identification

TLDR – PCR vs Sanger PCR amplifies loci from minimal input. Sanger reads exact sequences using ddNTP termination.


Objective: Appreciate that DNA sequence changes can be exploited in clinical diagnosis

Applications

  • Genetic disease: PCR for known loci; Sanger for CFTR, BRCA1/2.
  • Oncology: FISH for BCR-ABL1; PCR/Sanger for oncogenes.
  • Infectious disease: PCR for viral genomes, variants, load.
  • Forensics/identity: RFLP patterns.
  • Newborn screening: Mass spectrometry for metabolic disorders.
  • Large events: Karyotyping for aneuploidy.
flowchart LR
  Q[Clinical question] --> Size{Event size}
  Size --> Cytogenetics
  Size --> PCR
  Size --> Sanger
  Size --> Protein

TLDR – Clinical use Chromosomal tests detect large events. PCR handles small, known sequence differences. Sanger confirms exact variants. PCR also supports pathogen detection and viral load tracking.


L2.3 MECHANISMS OF GENE EXPRESSION

Objective: Understand that each cell contains the same DNA but expresses different genes

Core idea

  • All somatic cells share the genome; differential gene expression generates different cell types.

Why it matters

  • Drives differentiation, function, and adaptation.
  • Controlled at DNA, RNA, and protein levels.
flowchart LR
  SameDNA --> Programs
  Programs --> Expression
  Expression --> CellTypes

TLDR Same DNA, different expression profiles → different tissues.


Objective: Understand that DNA accessibility affects gene expression

Chromatin states

State Definition Effect
Heterochromatin Condensed Repressive
Euchromatin Open Permissive

Dynamic control

  • Histone modifications, nucleosome repositioning.
  • X-inactivation as chromosome-scale example.
flowchart TD
  Packed --> LowExpr
  Open --> HighExpr
  Signals --> Open
  Signals --> Packed

TLDR Open chromatin enables transcription. Closed chromatin represses it.


Objective: Understand the role of transcription factors and regulatory elements

Regulatory DNA

  • Core promoter, promoter-proximal elements, enhancers, silencers, insulators.

TFs

  • General TFs recruit Pol II.
  • Sequence-specific TFs bind motifs, recruit co-activators or co-repressors.
  • Combinatorial control sets precision.

Objective: Understand gene expression regulation at RNA and protein levels

RNA-level control

  • Splicing, editing, poly(A) tailing.
  • mRNA stability via UTRs and miRNAs.
  • Translation initiation control.
  • NMD removes faulty mRNAs.

Protein-level control

  • PTMs (phosphorylation, acetylation, glycosylation).
  • Proteasome/lysosome degradation.
  • Localization and complex formation.
flowchart LR
  premRNA --> splice
  splice --> mRNA
  mRNA --> decay
  mRNA --> translation
  translation --> protein
  protein --> PTM
  PTM --> fate

TLDR RNA-level steps shape isoforms, stability, and translation. PTMs and degradation fine-tune protein abundance and activity.

WEEK 3

L3.1 Genomic Variability in Populations

Objective: Explain the definition of population genetics and common terminologies in this field

What population genetics studies

  • Population genetics: branch of genetics that examines genetic variation to explain differences within an interbreeding population in a defined region. A population is all individuals of a species living in the same area who interbreed.

Core terms used throughout the lecture

Term Exam-grade definition One-line usage
Allele Alternative DNA sequence at a gene (locus) “W and w are alleles for flower colour.”
Genotype Allele pair at a locus in a diploid individual “WW, Ww, ww are genotypes.”
Phenotype Observable trait influenced by genotype and context “WW and Ww give purple; ww gives white.”
Homozygous Two identical alleles (AA or aa) “WW and ww are homozygous.”
Heterozygous Two different alleles (Aa) “Ww is heterozygous.”
Allele frequency Proportion of all gene copies that are a specific allele “W = 13/18 = 0.72; w = 5/18 = 0.28; p + q = 1.”
Gene pool All copies of a gene in the population “18 alleles across 9 plants is the gene pool.”
Genotype frequency Proportion of individuals with a genotype “WW 6/9 = 0.67; Ww 1/9 = 0.11; ww 2/9 = 0.22.”

Why these frequencies matter

  • Track evolutionary change.
  • Support public-health predictions using genotype distributions.

TLDR
Population genetics quantifies allele and genotype frequencies in interbreeding populations. Know allele, genotype, phenotype, homozygous, heterozygous, allele frequency, genotype frequency, and p + q = 1. These measures track population structure and evolutionary change.


Objective: Understand the principle of Hardy–Weinberg equilibrium (HWE), its assumptions, and importance

The HWE baseline

  • Large, non-evolving population under specific conditions maintains stable allele and genotype frequencies.

Required equations

  • Alleles: p + q = 1
  • Genotypes: p² + 2pq + q² = 1

Five assumptions

  1. Very large population (no drift)
  2. Random mating
  3. No mutation
  4. No migration
  5. No natural selection

Worked example (from lecture)

  • 1000 mice; ww = 400 → q² = 0.4 → q = 0.63246 → p = 0.36754.
  • Expected: p² = 0.135, 2pq = 0.4649, q² = 0.4 → matched actual counts.

Why HWE matters

  • Baseline for detecting evolutionary forces.
  • Useful for estimating carrier frequencies.

TLDR
HWE gives expected genotype frequencies (p², 2pq, q²) when no evolutionary forces act. Deviations signal drift, selection, migration, mutation, or non-random mating. The 1000-mouse example shows perfect HWE alignment.


Objective: Understand and calculate genotype and phenotype frequencies

Definitions

  • Allele frequency: proportion of allele copies.
  • Genotype frequency: proportion of individuals with each genotype.

Direct counting example

  • WW = 6, Ww = 1, ww = 2 (n=9).
  • Alleles: W = 13/18 = 0.72; w = 5/18 = 0.28.

Using HWE to infer genotypes

  • p = 0.72; q = 0.28 → p² = 0.5184, 2pq = 0.4032, q² = 0.0784.

Worked mouse example (dominant trait)

  • Black = p² + 2pq; white = q².

Minimal workflow

  • Use recessive phenotype frequency as q² when only phenotypes are known.

TLDR
Use genotype counts when available. Otherwise infer q from recessive phenotype (q²), then compute p = 1 − q, then genotype and phenotype frequencies.


Objective: Understand that populations evolve over time due to external factors

Evolutionary forces

  • Mutation
  • Gene flow
  • Genetic drift
  • Natural selection
  • Non-random mating

Why HWE rarely holds

  • Real populations violate assumptions (size, selection, migration, etc.).

Interpretation logic

  • Low heterozygosity → possible inbreeding or selection.
  • Allele frequency shifts → drift/selection/migration.
  • Large random swings in small populations → drift.

Human variation context (lecture)

  • Human–human: ~1/1000 bp
  • Human–chimp: ~1/100
  • Human–mouse: ~1/6–1/3

TLDR
Populations evolve because of mutation, migration, drift, selection, and non-random mating. HWE is a null model; deviations identify which forces act.


L3.2 Capturing Diversity

Objective: Explain the definition of genetic diversity and variation

Definitions

  • Genetic diversity: range of traits in a population.
  • Genetic variation: DNA differences enabling diversity.

Key facts

  • Unrelated humans differ at ~0.1% of bases.
  • SNPs ≈ 1 per 1000 bases; ≥1% frequency to be called SNP.
  • Highest human diversity in African populations.

Diversity vs variation

  • Variation is DNA-level difference; diversity is trait-level outcome.

TLDR
Genetic variation (SNPs, indels, structural variants) underpins genetic diversity. Humans differ at ~0.1% of bases; SNP frequency ~1/1000. African populations show highest diversity.


Objective: Understand the importance of genetic diversity and variation

Why diversity matters (population/ecosystem)

  • Boosts adaptability, resilience, and long-term survival.

Why diversity matters (human health)

  • Modulates disease susceptibility (e.g., blood group and norovirus).
  • Arises from drift, gene flow, selection, mutation.

Advantages/disadvantages

Aspect Advantage Disadvantage
Population survival Higher adaptability Harder to find causal variants
Human health Protective variants exist More opportunities for pathogenic variants
Public health Supports risk assessment Complexity for association studies

TLDR
Diversity improves resilience and shapes disease risk. Forces include drift, gene flow, selection, and mutations (~10⁻⁴–10⁻⁶ per gene per generation).


Objective: Understand importance and application of SNPs in medicine and health

Why SNPs matter

  • Explain differences in disease risk, drug response, toxicity.

Applications

Application What you do Value
GWAS risk scores Combine risk SNPs Stratify prevention/screening
Pharmacogenomics Genotype drug-response SNPs Guide therapy
Carrier/cascade testing Genotype in families Identify carriers
Molecular diagnosis Use tag SNPs/founder markers Speed diagnosis
Ancestry/panel use Genome-wide SNP sets Population context
Target discovery Map loci to effector genes Prioritise biological pathways

Pharmacogenomics examples

  • CYP2C19 & clopidogrel activation
  • HLA-B*57:01 & abacavir hypersensitivity
  • DPYD & fluoropyrimidine toxicity
  • SLCO1B1 & statin myopathy

Clinical workflow

  1. Define question.
  2. Select validated SNPs/panel.
  3. Genotype.
  4. Interpret with population/LD context.
  5. Act using guideline recommendations.

TLDR SNPs guide risk prediction, diagnosis, carrier testing, and drug selection. Pharmacogenomics relies heavily on SNPs with strong clinical evidence.


L3.3 Genome Analysis

Objective: Explain how a GWAS is conducted and what it requires

Purpose

  • Compare allele frequencies between cases and controls genome-wide.

Requirements

  • Ancestry-matched cohorts.
  • Large sample size (>1000; ideally >100k).
  • Clear phenotype.
  • High-quality genotyping (arrays).

How samples are genotyped

  • DNA → genotyping array with allele-specific probes → fluorescence scan → genotype call.

Pipeline

flowchart LR
  A[Define + recruit] --> B[Genotype + QC + impute]
  B --> C[Association tests]
  C --> D[Replication/meta]
  D --> E[Interpret loci]
  E --> F[Functional follow-up]

Arrays vs WGS

  • Arrays cheap and scalable; WGS expensive and data-heavy.

TLDR GWAS requires large, ancestry-matched cohorts and high-quality array genotypes. Workflow: recruit → genotype → QC/impute → test → replicate → interpret.


Objective: Understand how to read a Manhattan plot and interpret basic GWAS results

Outputs

  • Association table (CHR, SNP ID, BP, A1, OR, SE, P).
  • Manhattan plot (−log10 P vs genome).
  • QQ plot.

Reading the Manhattan plot

  • Highest peaks = strongest associations.
  • Look at chromosome and region.

Reading the table

  • A1 = effect allele.
  • OR > 1 = increased odds; OR < 1 = protective.
  • P-value indicates strength.

Follow-up questions

  • Which gene?
  • Coding or regulatory?
  • Functional impact?
  • Frequency in populations?

TLDR Use Manhattan plots to locate strong signals, then read table entries for effect allele, OR, and P. Follow-up requires mapping regions to genes and functions.


Objective: Understand the concept of LD and tag SNPs

Define LD

  • Non-random association of alleles at nearby loci.

Why LD exists

  • Nearby variants inherited together on same chromosome segment.

Practical use

  • Use tag SNPs to represent an LD block.
flowchart TD
  Block --> Tag
  Tag --> Array
  Array --> Infer

TLDR LD lets one SNP proxy a block of linked variants. GWAS arrays genotype tag SNPs to minimise redundancy.


Objective: Understand the limitations of GWAS

Limitations

Limitation Explanation
Sample size/power Needs huge N; small studies produce false positives.
Common-variant focus Poor at detecting rare variants.
Interpretation difficulty Most hits are non-coding.
Missing heritability Many traits not fully explained by common variants.
Ancestry confounding Must match ancestry to avoid spurious associations.

Implications

  • Build large matched cohorts, replicate findings, and use sequencing and functional studies to fill gaps.

TLDR GWAS is limited by power, its focus on common variants, and difficulty interpreting non-coding hits. It captures only part of heritability and must avoid ancestry confounding.

WEEK 4

L4.1 Non-Eukaryotic Genomes

Appreciate the difference in the size and complexity of genomes encoded by cellular and non-cellular life

What to compare

  • Genome size and coding capacity
  • Molecular form of the genome (DNA vs RNA, single- vs double-stranded, segmented vs non-segmented)
  • Packaging and storage (chromosomes, plasmids, nucleosomes vs none)
  • Replication context (cellular division vs obligate intracellular replication)

Size and complexity at a glance

Replicator Typical genome form Size range taught Coding content and packaging Notes for exam answers
Bacteria Usually circular DNA chromosome; plus plasmids Chromosome ~1×105–1×107 bp; plasmids ~5×103–2×106 bp Chromosome carries essential genes, vertically inherited; plasmids carry non-essential/fitness genes, often horizontally transferred; genomes are mosaic with mobile elements Know “chromosome vs plasmid” and the mobilome concept (transposons, insertion sequences).
Viruses DNA or RNA; ss or ds; segmented, non-segmented, sometimes multipartite ~2 kb to >2 Mb No nucleus; no histone packaging; wide architectural diversity; giant viruses encode thousands Some giant viruses exceed small bacteria in size.
Viroids Small circular RNA, no proteins ~300 nt RNA acts as information and enzyme; no protein coding Counterexample to the central dogma.

Storage and organisation contrasts

  • Cells vs non-cells: eukaryotes store DNA in nuclei with nucleosomes; bacteria have nucleoids and plasmids; viruses/viroids have no chromosomes.
  • Bacterial mosaic genomes: mobile elements move between chromosome and plasmids, driving rapid adaptation such as antibiotic resistance.
  • Viral architectures: ss/ds DNA or RNA, segmented or multipartite; some RNA viruses present translation-ready genomes; retroviruses integrate.

Why complexity differs

  • Gene number increases roughly with genome size but not linearly.
  • Giant viruses have large gene sets with many unknown functions.
  • Inheritance context: bacteria inherit chromosomes vertically but gain accessory genes horizontally; viruses replicate clonally inside hosts.

TLDR Cellular genomes (bacteria) have large, structured DNA chromosomes plus plasmids with vertical and horizontal inheritance. Non-cellular genomes span extremes, from ~2 kb RNA viruses to >2 Mb giant DNA viruses, with viroids at ~300 nt. Differences in packaging, replication, and inheritance explain the range in size and complexity.


Compare and contrast how non-eukaryotic genetic material is inherited/acquired

Overview

  • Two axes: vertical inheritance and horizontal acquisition.
  • Bacteria/archaea use both; viruses depend on host cycles; viroids use rolling-circle RNA replication.

Cellular prokaryotes (bacteria/archaea)

Vertical inheritance
  • Chromosome replicates and segregates during binary fission.
  • Plasmids partition by copy-number control and partition systems.
Horizontal gene transfer (HGT)
Mechanism Core steps Vehicles/DNA forms Notes
Transformation Uptake of free DNA; integrates by homologous recombination Linear fragments or plasmids Natural competence is species-specific.
Conjugation Donor plasmid forms a pilus; DNA transfer begins at oriT F plasmids; Hfr strains mobilise chromosomal segments Major route for antibiotic resistance spread.
Transduction Phage packages host DNA accidentally (generalised) or adjacent DNA on excision (specialised) Phage particles Moves gene cassettes between strains and species.
  • Mobile elements (transposons, integrons) jump between chromosome and plasmids.

Viruses

Vertical vs horizontal
  • Horizontal: virion-mediated transmission between hosts.
  • Vertical-like: integrated prophage or provirus replicates with host genome.
Genetic exchange in viruses
Route Applies to What occurs Why it matters
Reassortment Segmented RNA viruses Swap genome segments during co-infection Sudden antigenic shifts; altered host range/virulence.
Recombination Many DNA viruses, some RNA viruses Polymerase template-switching Creates hybrid genomes.
Integration Temperate phages, retroviruses Insert/remove viral DNA in host genome Stable persistence, occasional host-gene capture.

Viroids

Characteristics
  • Small circular RNA replicating by rolling-circle mechanisms using host polymerases.
  • Spread cell-to-cell and between plants via mechanical transfer or vectors.
  • No DNA or proteins; transmission is purely RNA-based.

Compare and contrast summary

Feature Bacteria/Archaea Viruses Viroids
Vertical inheritance Chromosome + plasmid replication Only when integrated RNA copied in host cells
Horizontal acquisition Transformation, conjugation, transduction Reassortment, recombination, co-infection Cell-to-cell RNA movement
Vehicles Free DNA, plasmids, phages Virions, genome segments Naked RNA
Dependency on host Moderate Complete Complete

TLDR

Prokaryotes inherit chromosomes vertically but acquire new genes by transformation, conjugation, and transduction. Viruses exchange genetic material via infection, reassortment, and recombination, and persist vertically only when integrated. Viroids replicate as RNA circles and transmit through cell-to-cell spread.


L4.2 Genetics as a Diagnostic Tool

Explain the similarities and differences between PCR and qPCR

Shared features

  • Primer-directed DNA synthesis by thermostable polymerase.
  • Amplification cycles: denaturation, annealing, extension.
  • Require known target sequence.

Key differences

Feature PCR qPCR
Readout End-point gel band Fluorescence measured every cycle
Quantification Qualitative only Ct value gives relative quantity
Reagents Primers, polymerase Primers + fluorescent probe/dye
Interpretation Amplicon present/absent Lower Ct = more template
Vulnerabilities Mispriming, contamination All PCR issues + probe mismatch from mutations

Minimal workflow

PCR: extract → (RT) → amplification → gel
qPCR: extract → (RT) → amplification → real-time fluorescence → Ct

TLDR

PCR is qualitative and uses gels. qPCR adds fluorescence and reports a Ct, a relative measure of template quantity. Multi-target panels guard against target mutations.


Understand how qPCR is applied to real-world scenarios

Clinical use

  • Respiratory pathogen panels (SARS-CoV-2, Influenza A/B, RSV, etc.).

Ct interpretation

  • Ct is relative and lab-specific.
  • Low Ct → high template.
  • High Ct (>44) → negative in course example.

Multi-target justification

  • Single-gene dropouts occur when mutations disrupt primer/probe binding.
  • Example: SARS-CoV-2 S-gene target failure due to 69–70 deletion.

Workflow

  1. Panel run.
  2. Review Ct.
  3. If one target drops out, consider mutation.
  4. If all negative, proceed to untargeted sequencing.

TLDR

Ct reflects relative template quantity. Multi-target panels detect pathogens even when mutations disrupt one target. Negative panels with ongoing suspicion lead to sequencing.


Identify when untargeted sequencing is useful

What “untargeted” means

  • Sequencing all RNA in the sample.
  • No pathogen-specific primers.

Useful when

  • All targeted tests negative.
  • Novel, diverse, or unexpected pathogens suspected.
  • Need full genome for phylogenetics.

Not useful when

  • A qPCR panel already covers likely pathogens.
  • Rapid quantitative thresholds needed.
  • Limited resources or bioinformatics capability.

Process summary

Extract total RNA → build library → sequence → analyse → phylogenetics

TLDR Untargeted sequencing identifies unknown or novel pathogens and recovers full genomes. It is not first-line when targeted qPCR is available.


L4.3 Intro to Phylogenetics

Compare and contrast pedigrees and phylogenies

Table

Feature Pedigree Phylogeny
Shows Parent–offspring relationships Evolutionary relationships among sequences
Nodes Actual individuals Inferred ancestors
Tips People Sampled genomes
Branch lengths Generations Time or mutations

TLDR

Pedigrees track parents and offspring. Phylogenies infer evolutionary relationships; internal nodes are ancestral populations and branch lengths encode time or mutation counts.


Describe the relationship between a phylogeny and an ancestral population

Key points

  • Internal nodes represent inferred ancestral populations.
  • Branch length reflects time or mutation accumulation.
  • Clades = sets of tips sharing a most recent inferred ancestor.
tips → inferred ancestor → deeper ancestor

TLDR Internal nodes correspond to ancestral populations and branch lengths show how far tips diverged from them.


Explain how mutations trace pathogen spread

Core logic

  • Mutations accumulate at characteristic rates.
  • Differences in genomes indicate relatedness.
  • Trees built from sequences reveal introductions, clusters, and spread direction.

Workflow

  1. Collect time- and place-stamped samples.
  2. Sequence genomes.
  3. Build time-scaled phylogeny.
  4. Map clusters, introductions, host jumps.
  5. Integrate with epidemiological metadata.

TLDR Mutations act as markers for transmission. Time-scaled phylogenies reconstruct spread and inform public health action.

WEEK 5

L5.1 Variations in Human Health — Exam notes

Objective 1. Explain how monozygotic (MZ) and dizygotic (DZ) twin studies help disentangle genetic and environmental contributions

Core logic

  • MZ twins arise from one zygote that splits, so they share ~100% of segregating DNA; DZ twins arise from two zygotes, so they share ~50% on average, like ordinary siblings. When MZ pairs are more similar than DZ pairs, genetics explains part of the variance. When MZ ≈ DZ, shared environment or chance dominates.

Binary traits: concordance comparison

  • Concordance = percent of twin pairs where both twins have the trait. Higher MZ than DZ concordance supports a genetic contribution, but not genetic determinism. Example values taught: schizophrenia 46% MZ vs 15% DZ; bipolar 62% vs 8%; type 1 diabetes 40% vs 5%.

Quantitative traits: correlation comparison

  • Replace concordance with correlation (r). Typical pattern in the slides: height r_MZ ≈ 0.90, r_DZ ≈ 0.50, implying strong genetic influence with room for environment/chance because r_MZ < 1.0.

TLDR Use MZ vs DZ similarity to partition influences: larger MZ–DZ gaps imply genetic effects; small gaps imply environment or chance. Binary traits use concordance, quantitative traits use correlation. Even high MZ similarity leaves space for non-genetic factors.


Objective 2. Interpret concordance rates and heritability estimates, and describe what they do (and do not) tell us

Reading concordance

  • Definition: percent of twin pairs with the same status.
  • Interpretation: MZ ≫ DZ suggests a genetic component; MZ < 100% shows environment, stochastic events, or epigenetic/accessibility effects also matter. Use disorder tables as context, not proof of determinism.

Estimating heritability (twin method)

  • Heritability (broad-sense here) is the proportion of trait variance in a population attributable to genetic differences.
  • Falconer’s estimator from twin correlations: ( h^2 = 2 × (r_MZ − r_DZ) ). Example from the slides: height with (r_MZ = 0.90), (r_DZ = 0.50) → (h^2 = 2 × (0.90 − 0.50) = 0.80) (80%).

What heritability does tell you

  • A population-level summary under current environments.
  • A signal that genetics explains some fraction of observed variance.
  • A guide for expecting familial resemblance and for prioritising genetic studies.

What heritability does not tell you

  • Not individual causation (you cannot say “your trait is X% genetic”).
  • Not immutability: high heritability ≠ inevitability (e.g., nutrition alters height; diet prevents PKU disease).
  • Not mechanism: it does not identify specific genes, pathways, or modifiable exposures.
  • Not unbiased if assumptions fail: equal-environments assumption, shared prenatal/postnatal settings, and twin generalisability can inflate estimates.

Quick checklist for exam items

  • Quote concordance with both MZ and DZ values when provided.
  • For quantitative traits, state r_MZ and r_DZ, then compute (h^2) with Falconer’s formula.
  • Add one limitation line (equal environments, representativeness) and one “high heritability ≠ destiny” example.

TLDR Concordance compares twin pair similarity for binary traits; MZ ≫ DZ implies genetic influence but never proves determinism. Heritability (for example Falconer’s h^2) quantifies the genetic share of variance in a population, not for individuals. It does not fix outcomes or identify mechanisms and can be biased if twin assumptions are violated. Use it alongside context and limitations.


Objective 3. Recognise how gene–environment interactions and modifier genes contribute to variable disease expression

What to state first

  • Same genotype, different outcomes when environment differs or when modifier genes alter pathways upstream, downstream, or parallel to the primary gene. Penetrance and expressivity reflect this variability.

Gene–environment interactions (G×E)

  • Exposure changes outcome for a given genotype.

    • Diet-controlled PKU: pathogenic PAH alleles produce disease only when dietary phenylalanine is unmanaged; dietary restriction reduces phenylalanine and prevents neurotoxicity.
    • Smoking and lung function: genotypes affecting protease–antiprotease balance or ciliary function show worse trajectories with smoke exposure.
    • Infections, hormones, temperature, and microbiome shift expression, timing, and severity in genetically predisposed people.
How to write it in exams
  • Name the genotype. Name the exposure. State the direction of effect (exposure raises or lowers risk or severity). Tie to penetrance or expressivity.

Modifier genes

  • Second loci change severity or age of onset without being the primary cause.

    • Modifiers alter dosage (transcription), processing (splicing), protein folding or clearance, or parallel pathways that compensate or exacerbate.
    • Expect intrafamilial variability even with the same causal allele because family members carry different modifiers.
Workflow to reason about variability
  1. Confirm primary genotype.
  2. List relevant environments (diet, toxins, infections, hormones).
  3. List candidate modifiers (pathway members, transporters, chaperones, inflammatory mediators).
  4. Explain penetrance or expressivity through these levers.

TLDR Variable expression arises when environmental exposures interact with genotype and when modifier genes tune pathway flux. Report outcome in terms of penetrance and expressivity. Use PKU–diet and exposure-dependent lung disease as standard illustrations, and explain intrafamilial variability through modifiers acting on the same pathway.


Objective 4. Appreciate that random change (genetic drift, stochastic events) also shapes phenotypic outcomes

Stochastic sources within individuals

  • Developmental noise: small random differences during development lead to measurable trait differences even in MZ twins raised together.
  • Epigenetic drift: random changes in chromatin marks alter expression over time.
  • Somatic mosaicism: post-zygotic mutations create clone-specific differences between tissues or twins.
  • Random X-inactivation in XX individuals produces tissue mosaics; the proportion of cells inactivating each X varies, shifting phenotype severity for X-linked traits.

Population-level randomness

  • Genetic drift: in finite populations, allele frequencies fluctuate by chance across generations; effects are strongest in small populations and during founder events or bottlenecks. Drift changes disease-allele frequencies independent of selection.

How to incorporate randomness in answers

  • After discussing genes and environment, add a stochastic term: random X-inactivation, somatic mutation, or developmental noise explains residual discordance.
  • For populations, state whether observed frequency change could reflect drift given population size and recent bottlenecks.

TLDR Not all variation is deterministic. Within individuals, random X-inactivation, epigenetic drift, and somatic mutations change expression and severity. Across populations, genetic drift shifts allele frequencies by chance, especially after bottlenecks. Use these to explain residual MZ twin discordance and population frequency changes that lack a selection signal.


L5.2 Introduction to Monogenic Diseases

Objective: Distinguish between coding, splicing, regulatory, expression, and localisation mutations in monogenic disease

The framework

Mutations in a single gene can disrupt different steps between DNA and protein. Name the class, define the mechanism, and give a disease example.

Class What changes Molecular effect Typical outcome Lecture examples
Coding (missense, nonsense, frameshift, silent) Base(s) within the coding sequence Missense swaps one amino acid. Nonsense creates a premature stop and often triggers NMD. Frameshift (indel not ×3) rewrites downstream codons. Silent keeps amino acid but can still affect expression. Usually loss of function; occasionally toxic gain of function Sickle cell disease, single-aa missense in β-globin; Duchenne muscular dystrophy, frameshift in dystrophin; silent defined in slides.
Splicing (non-coding) Splice donor or acceptor, branch point, enhancers Exon skipping, intron retention, cryptic splice site use; frameshift and or premature stop common Reduced or absent functional protein; often NMD Spinal muscular atrophy (SMN1 splicing defects); β-thalassemia splice-site variants.
Regulatory (promoter or enhancer or 5′UTR) Promoter and other cis-elements Transcription initiation or efficiency altered; epigenetic silencing in some repeats Normal protein sequence but too little (or no) mRNA β-thalassemia promoter variants; Fragile X, 5′UTR CGG expansion silencing FMR1.
Expression level (dosage) Gene copy number or one functional allele Haploinsufficiency reduces dosage; duplication or amplification increases dosage Too little or too much protein; dosage-sensitive phenotypes Williams syndrome (ELN deletion → low elastin); CMT1A (PMP22 duplication → overexpression).
Localisation or folding Protein folding or trafficking signals Misfolded protein degraded (ERAD) or trapped; mislocalised protein fails to reach compartment Loss of function at site of action; possible toxic gain from aggregates CFTR ΔF508 misfolding and ER retention; α1-antitrypsin Z variant accumulates in liver (deficiency in lung and hepatotoxicity); primary ciliary dyskinesia (dynein mislocalisation).

Quick decision map (use in short answers)

flowchart TD
  DNA[DNA] -->|coding| ProteinSeq[Protein sequence changed]
  DNA -->|splicing| RNAProc[RNA processing altered]
  DNA -->|regulatory| Transcription[Transcription altered]
  GeneDose[Gene dosage] --> Expression[Protein amount altered]
  ProteinSeq --> FoldLoc[Fold/traffic to correct compartment?]
  FoldLoc --> Function[Function at site]

Phrases that score

  • “Nonsense → premature stop → NMD likely when early.”
  • “Frameshift (not ×3) rewrites downstream codons → null.”
  • “Promoter or 5′UTR variant → reduced transcription, normal protein sequence.”
  • “Haploinsufficiency: one working copy is not enough.”
  • “ΔF508 CFTR misfolds and is retained in ER, so channel never reaches membrane.”

TLDR Distinguish where the defect acts. Coding alters the protein sequence (missense, nonsense, frameshift). Splicing alters exon–intron processing. Regulatory variants alter transcription without changing sequence. Expression level problems change dose (haploinsufficiency or duplication). Localisation or folding defects produce, but misplace or degrade, the protein. Anchor each class with the lecture’s disease examples and state the molecular consequence succinctly.


Objective 2. Recognise that the same disease phenotype can result from different molecular mechanisms

Three layers of many paths to one phenotype

  • Locus heterogeneity: different genes, same clinical picture. Distinct genes in the same pathway can each cause the disease phenotype.
  • Allelic heterogeneity: different variants in the same gene produce the same diagnosis but with variable severity or age at onset (missense vs nonsense vs splice).
  • Mechanistic heterogeneity within one gene: the phenotype arises via different molecular routes in the same locus (for example sequence change, mis-splicing, or mis-localisation), yielding overlapping clinical presentations.

Lecture-aligned exemplars (anchor these in short answers)

Phenotype Molecular routes in lecture Consequence pattern
β-thalassemia Promoter variants (reduced transcription), splice-site variants (exon skipping or cryptic splice), coding nonsense or frameshift (PTC → NMD) Reduced β-globin dose; severity tracks residual expression
Cystic fibrosis (CFTR) Missense gating defects (reduced channel opening), folding or trafficking defects (ΔF508 ER retention), splice defects (aberrant mRNA), rarer promoter or 5′UTR effects Common phenotype with variable sweat chloride, pulmonary and pancreatic involvement
Dystrophinopathies Frameshift (DMD, null), in-frame deletions (BMD, partial function), splice variants creating either outcome Shared muscle phenotype with severity gradient
A1AT deficiency Missense misfolding (Z allele, polymer accumulation, gain-of-toxicity in liver plus loss in lung), rarer null alleles (severe loss without hepatic polymer) Same diagnosis via distinct mechanisms (toxic gain plus loss, or pure loss)

TLDR The same diagnosis can arise from different genes (locus heterogeneity), different variants in one gene (allelic heterogeneity), and different mechanisms within one locus (regulatory, splicing, coding, localisation). Use β-thal, CFTR, dystrophin, and A1AT as worked anchors from the lecture.


Objective 3. Appreciate that monogenic diseases are often considered potentially druggable

Why monogenic targets are attractive

  • Single dominant node: one gene or pathway node explains most of the phenotype, simplifying target selection and trial endpoints.
  • High effect size: large, reproducible biology improves signal-to-noise in small studies.
  • Mechanism clarity: knowing where the defect acts (transcription, splicing, folding, trafficking, function) points directly to a modality.

Mechanism → modality mapping used in the lecture

Defect class Therapeutic idea Lecture exemplars to cite
Gating or function defect Small-molecule potentiator to increase activity CFTR gating variants treated with potentiators
Folding or trafficking defects Pharmacological chaperone or corrector to improve folding and delivery CFTR ΔF508 with correctors; A1AT strategies framed as folding or aggregation problem
Splicing error Antisense oligonucleotide to modulate splicing; exon-skipping strategies SMA splicing rescue; exon-skipping in dystrophin
Nonsense or early stop Readthrough agents or gene addition or editing Nonsense-focused approaches summarised; gene addition listed for null states
Low transcription or regulatory Transcriptional up-regulation, gene addition, or vector-mediated expression β-thalassemia or hemoglobinopathies presented as dose restoration
Pure loss of enzyme Enzyme or protein replacement or gene addition Enzyme deficiency archetypes; augmentation for deficiency states
Clinical logic in short answers
  1. Identify mechanism class (coding, splicing, regulatory, localisation, dose).
  2. Pick modality that directly counters that class.
  3. Note that combinations are common (for example CFTR corrector plus potentiator).
  4. Add a monitoring readout aligned with mechanism (for example sweat chloride for CFTR).

Practical cautions

  • Genotype specificity: many drugs help only certain variant classes.
  • Delivery and durability: gene and antisense therapies depend on tissue delivery and persistence.
  • Toxicity trade-offs: chaperoning misfolded proteins can reduce degradation but risk aggregation.

TLDR Monogenic diseases are druggable because mechanism is clear and concentrated in a single node. Match defect class to modality: potentiators for gating, correctors for folding or trafficking, antisense oligos for splicing, readthrough or gene therapy for nonsense or null, replacement for enzyme loss, and transcriptional up-regulation or gene addition for regulatory or dose problems. Many therapies are genotype-specific and delivery or durability constraints matter.


L5.3 Introduction to Polygenic Diseases

Objective 1. Explain what a PRS is and how it is calculated

Definition

  • Polygenic risk score (PRS) is a single number that summarises inherited predisposition for a trait or disease by combining the effects of many SNPs identified in GWAS. It is the weighted sum of risk alleles across selected SNPs, where weights are GWAS effect sizes.

Inputs you must name

  • A curated list of SNPs associated with the trait from GWAS, each with:

    • Effect allele (the allele associated with higher or lower risk)
    • Effect size (β_i) from the GWAS result
  • The individual’s genotype for each SNP, coded as the count of effect alleles (G_i ∈ {0,1,2}).

Calculation formula

[ = _{i=1}^{m} _i G_i]

  • Positive β increases risk score. Negative β is protective.
  • Scores are often standardised in a reference cohort to allow percentile interpretation.

Minimal workflow

  1. Start with GWAS summary statistics for the trait.
  2. Select SNPs and their β values.
  3. Genotype the individual and code G_i.
  4. Compute the weighted sum.
  5. Place the score on the population distribution to obtain a percentile.

Notes that score well

  • Most single SNP effects are small; the cumulative sum matters.
  • Protective and risk alleles both contribute; the net sum determines direction.
  • PRS is relative to the population used for weights and normalisation.

TLDR PRS is a weighted sum of effect alleles across GWAS SNPs. Count the effect alleles per SNP (G_i), multiply by the GWAS effect (β_i), then sum. Interpret the score against a reference population to get a percentile.


Objective 2. Describe how PRS informs risk, not determines it

What PRS tells you

  • Relative genetic risk, suitable for risk stratification across a population.
  • Scores distribute approximately bell-shaped. People in the high tail can have several-fold higher odds than the population average. People in the low tail are relatively protected.

What PRS does not tell you

  • Not a diagnosis and not deterministic for an individual.
  • Predictive value depends on ancestry match between the person and the GWAS that supplied the weights.
  • Does not incorporate environment or lifestyle by itself.

Interpreting a person’s score

  • Report percentile in a matched population reference.
  • Translate to relative risk categories, not absolute probability.
  • Combine with environment and lifestyle to refine overall risk. The lecture’s matrix shows high PRS plus poor environment yields the highest risk, while good environment buffers even high PRS.

Communication phrases to use

  • “Higher than average genetic liability, not a guarantee.”
  • “Environment can amplify or buffer genetic predisposition.”
  • “Accuracy is best when ancestry matches the discovery GWAS.”

TLDR PRS ranks genetic predisposition. It improves stratification, not diagnosis. Outcomes still depend on environment, lifestyle, and chance, and transferability depends on ancestry alignment with the GWAS that produced the weights. Use percentiles and relative-risk language.


Objective 3. Understand gene–environment interaction in polygenic disease

Define the concept

  • Gene–environment interaction (G×E) occurs when the effect of a person’s genetic background on a trait depends on their exposures, or vice versa. Environment can magnify, reduce, or reverse genetic risk.

What the lecture emphasised

  • PRS is probabilistic. It shifts likelihood, not certainty. Outcomes still depend on lifestyle, environment, and chance.
  • The slides present a matrix: high PRS with poor environment gives highest risk; high PRS with good environment reduces risk; low PRS with poor environment can still lead to disease; low PRS with good environment gives lowest risk.

How to reason about G×E in answers

  1. State genetic liability (for example high vs low PRS).
  2. Specify the exposure set (diet, smoking, activity, stress).
  3. Combine them to place the person in a risk cell (highest, moderate, lowest) using the matrix logic.

Worked patterns from the slides and transcript

  • Coronary artery disease: high PRS plus smoking, inactivity, and poor diet yields the highest risk; lifestyle improvement buffers risk even with high PRS.

  • Type 2 diabetes: many variants raise susceptibility, but diet and exercise move risk up or down. High heritability does not imply inevitability.

  • Alcohol use disorder case study:

    • Genetics contributes about half the variance in liability in population studies; MZ concordance exceeds DZ, showing genetic influence.
    • Individual SNP effects are modest; the score aggregates many loci. Social context, stress, trauma, access, and norms strongly modify risk.

Communication phrases to reuse

  • “High PRS raises relative risk; environment determines where the person lands on the matrix.”
  • “Low PRS does not guarantee protection in a poor environment.”
  • “Report risk by percentile and describe exposures that amplify or buffer genetic liability.”

TLDR G×E means genetic effects depend on exposure. The course matrix shows four cells: high PRS with poor environment is highest risk; high PRS with good environment is buffered; low PRS with poor environment can still lead to disease; low PRS with good environment is lowest. Use this framework to interpret CAD, T2D, and AUD examples. Always phrase PRS as probability, shaped by lifestyle and chance.

WEEK 6

L6.1 Sickle Cell Disease

Objective 1. Describe the difference between sickle cell trait and sickle cell anaemia, and how sickle cell trait protects against severe malaria

Genotype, phenotype, and clinical contrast

Feature Sickle cell trait Sickle cell anaemia
Genotype Heterozygous HbAS (one HbS allele, one normal HbA) Homozygous HbSS (two HbS alleles)
Hemoglobin mixture ~40–50% HbS, remainder HbA Nearly all HbS
Sickling Limited sickling under severe hypoxia; usually asymptomatic Marked sickling in deoxygenated blood; repeated cycles cause irreversible sickling
Pathophysiology Minimal polymer formation because HbA disrupts HbS polymer growth HbS polymerises into rigid fibres in low O₂, distorting RBCs, causing vaso-occlusion, hemolysis, pain crises, organ damage
Population pattern Common in malaria-endemic regions 300,000–400,000 new cases per year worldwide; distribution tracks malaria maps

How sickle cell trait protects against severe malaria

  • In HbAS erythrocytes, Plasmodium replication is impaired. Infected cells are more prone to membrane damage and rupture, interrupting the parasite life cycle. This lowers risk of severe malaria. The global distribution of HbS mirrors malaria endemicity.

TLDR Trait (HbAS) mixes HbS with HbA, which limits polymer growth and sickling, so people are usually asymptomatic. Disease (HbSS) is near-pure HbS, which polymerises in low oxygen, causing hemolysis and vaso-occlusion. Trait confers protection against severe malaria because infected RBCs rupture more readily, disrupting parasite replication.


Objective 2. Explain how the Glu6Val mutation disrupts hemoglobin structure and function, and how this leads to sickle cell trait or anaemia

Molecular substitution and its consequence

  • Mutation: β-globin Glu6Val (HBB; HbS). Glutamate is polar and surface-exposed. Valine is hydrophobic and creates an exposed hydrophobic patch on HbS.
  • State dependence: In the deoxygenated conformation, Val6 fits a hydrophobic pocket on a neighbouring hemoglobin (lined by Phe85 and Leu88), driving HbS–HbS contacts. This nucleates long polymers that stiffen the cell. Oxygenated Hb does not expose the same interface to the same extent.

From polymer to disease

  • HbSS: High HbS concentration promotes polymer nucleation and fibre growth with each deoxygenation. Repeated cycles damage membranes, create permanently sickled RBCs, and cause vaso-occlusion and hemolysis.
  • HbAS: HbA disrupts HbS–HbS packing. Polymer formation is limited, so sickling is minimal except under extreme hypoxia. This explains the trait’s usual lack of symptoms.

TLDR Glu→Val at β6 creates a hydrophobic patch on HbS. In deoxygenated blood, Val6 docks into a hydrophobic pocket on another hemoglobin (Phe85, Leu88), triggering polymerisation. In HbSS, high HbS allows robust fibre formation, producing rigid, hemolysis-prone cells and vaso-occlusion. In HbAS, HbA dilutes and disrupts HbS packing, so polymerisation and sickling remain limited.


Objective 3. Describe the different forms of haemoglobin throughout development, and the advantage of increased fetal haemoglobin levels in sickle cell patients

Developmental haemoglobin series you must name

Stage Tetramer (globin chains) Where the chains come from Notes for exams
Embryonic (very early) Gower I ζ₂ε₂, Gower II α₂ε₂, Portland ζ₂γ₂ α-like cluster (ζ→α) on chr16; β-like cluster (ε→γ→δ/β) on chr11 Short-lived; appear before placental HbF dominance
Fetal (mid–late gestation → early infancy) HbF α₂γ₂ α from chr16, γ from chr11 High O₂ affinity supports placental transfer; persists for months after birth
Adult (post-infancy) HbA α₂β₂ (major), HbA₂ α₂δ₂ (minor) β and δ from chr11 HbA replaces HbF through developmental switching

The slides group these as a developmental switch from ε/ζ to γ to β/δ across the two loci. You should state that the β-like cluster is ordered 5′→3′ roughly in the order of expression (ε→γ→δ/β).

Why HbF helps in sickle cell disease

  • HbF does not participate in HbS polymer contacts. The γ chain lacks the β-chain surface features that accept Val6 from HbS in deoxygenated blood, so mixed tetramers and HbF-rich cells are less prone to polymerisation and sickling.

  • Clinical correlations from the lecture:

    • Higher HbF levels reduce hemolysis and vaso-occlusive crises.
    • Hydroxyurea increases HbF and improves outcomes; induction of HbF is a core disease-modifying strategy.
    • Curative approaches (e.g., gene addition/editing) also aim to reactivate HbF by altering γ-globin regulation.

Phrases that score

  • “HbF (α₂γ₂) persists after birth, then declines as HbA (α₂β₂) rises.”
  • “γ-globin reduces HbS polymerisation; more HbF means fewer crises.”
  • “Therapies that raise HbF are disease-modifying.”

TLDR Development moves from embryonic ζ or ε or γ tetramers to HbF (α₂γ₂) in the fetus, then to adult HbA (α₂β₂) with minor HbA₂ (α₂δ₂). HbF protects because γ chains do not accept the HbS Val6 interaction that drives polymerisation, so cells with more HbF sickle less. Treatments that raise HbF (for example hydroxyurea or HbF-reactivation strategies) reduce crises and hemolysis.


L6.2 Cystic Fibrosis

Objective 1. Explain the genetic and physiological basis of Cystic Fibrosis

Genetics

  • Cause: autosomal recessive disease from CFTR gene mutations.
  • Carrier frequency: ~1 in 25 Australians; prevalence <5 in 10,000.
  • Diagnosis: newborn heel-prick screen with immunoreactive trypsinogen, then confirm with CFTR genotyping and sweat chloride test (gold standard).

CFTR protein and physiology

  • Location: apical membrane of epithelial cells in airways, pancreas, sweat glands, and male reproductive tract.
  • Function: ATP-gated chloride channel in the ABC transporter family. ATP binding dimerises the NBDs and opens the pore; ATP hydrolysis returns it to closed. An R-domain with phosphorylation sites regulates opening.
  • Airway mechanism: CFTR-mediated Cl⁻ secretion draws water into airway surface liquid (ASL). Hydrated ASL lets cilia beat and mucus clear. Loss of CFTR → reduced Cl⁻ secretion → ASL dehydrationthick mucus, obstruction, infection, inflammation.
  • Sweat mechanism: CFTR dysfunction impairs salt reabsorption → elevated sweat chloride (up to ~4× normal), basis of sweat test.
  • Pancreas mechanism: reduced Cl⁻ or water secretion from duct cells → viscous secretions → duct blockage → failure of digestive enzyme delivery → malabsorption and malnutrition.

TLDR CF is autosomal recessive and caused by CFTR mutations. CFTR is an ATP-gated Cl⁻ channel; loss of function dehydrates airway liquid, raises sweat chloride, and thickens pancreatic secretions. Screen with IRT, confirm by genetics and sweat test.


Objective 2. Describe why Cystic Fibrosis is a multi-organ disease, and identify the systems most commonly affected

Why multi-organ

  • CFTR is expressed in many epithelia, so defective chloride and water transport affects multiple exocrine systems.

Systems and consequences

System Primary epithelial defect Dominant consequences
Respiratory (upper and lower airways) ↓ CFTR Cl⁻ secretion → ASL dehydration → impaired mucociliary clearance Chronic mucus obstruction, recurrent infection, persistent inflammation, airway damage
Pancreas / GI Thick ductal secretions block enzyme flow Pancreatic insufficiency, maldigestion, malnutrition, risk of insulin insufficiency
Sweat glands Impaired salt reabsorption in ducts High sweat chloride (diagnostic)
Male reproductive tract CFTR in exocrine epithelia of vas or epididymis Obstructive phenomena contributing to infertility patterns (not detailed mechanistically in this lecturette beyond organ listing)

Clinical framing used in the lecture

  • Respiratory and GI systems are the major contributors to morbidity in routine care pathways for CF.

TLDR CF is multi-organ because CFTR functions in many epithelia. Airways: dehydrated ASL → thick mucus, infection, inflammation. Pancreas: blocked ducts → enzyme delivery failure and malnutrition. Sweat glands: high sweat chloride. Male tract: epithelial involvement consistent with obstructive phenotypes.


Objective 3. Define the most common mutations in the CFTR gene associated with Cystic Fibrosis, and explain how they affect CFTR protein structure and function

Variant classes highlighted in the lecture

Mutation (example) Molecular defect Structural or functional consequence Clinical note from slides or transcript
F508del (ΔF508) Misfolding of CFTR, chiefly in NBD1 ER retention and degradation, so little channel reaches the apical membrane; reduced Cl⁻ transport Most common worldwide; archetype of folding or trafficking defect.
Gating defects (e.g., G551D) Channel reaches membrane but fails to open efficiently Low open probability despite membrane localisation; ATP-gating cycle is impaired Distinct from folding; important because it is potentiator-responsive.
Nonsense / early stop Premature termination codon → truncated protein mRNA often degraded by NMD; absent protein Results in severe loss of function; grouped as “minimal function” in clinic.
Splicing variants Altered exon inclusion Reduced or abnormal CFTR; may introduce frameshift or PTC Severity tracks residual correctly spliced mRNA.

How these produce disease physiology (link back to Obj. 1): Any mechanism that lowers functional CFTR at the apical membrane reduces Cl⁻ secretion, dehydrates airway surface liquid, and thickens secretions in affected organs.

TLDR Common CFTR mutations fall into folding or trafficking (for example F508del, ER retention), gating (for example G551D, low open probability at membrane), nonsense (absent protein via NMD), and splicing (aberrant transcripts). All reduce functional CFTR, lowering Cl⁻ transport and driving CF pathology.


Objective 4. Demonstrate an understanding of the key treatments used to manage Cystic Fibrosis symptoms

Disease-modifying CFTR-targeted therapy (variant-guided)

Defect category Modulator strategy Lecture framing
Gating (e.g., G551D) Potentiator to increase channel open probability at the membrane Increases Cl⁻ flux in variants where the channel is present but opens poorly.
Folding or trafficking (e.g., F508del) Corrector(s) to improve folding or processing and deliver CFTR to the surface; often combined with a potentiator Addresses misfolding plus low Po; combination therapy highlighted.
Minimal-function (nonsense or severe splice) Limited modulator benefit; non-CFTR approaches remain central Slides flag limited responsiveness; manage complications aggressively.

Organ-system management emphasised in the lecture

System Main interventions and why
Respiratory Airway clearance techniques; nebulised therapies to hydrate or loosen mucus; antibiotics for acute infections and chronic suppression; vaccination; regular physiotherapy. Goal: improve mucociliary clearance, reduce bacterial load, blunt inflammation.
Pancreas / GI Pancreatic enzyme replacement therapy (PERT); high-calorie, high-fat diet; fat-soluble vitamin supplementation; monitor for CF-related diabetes. Goal: restore digestion and nutrition.
Sweat/fluids & general Maintain hydration, electrolyte awareness (sweat salt loss). Multidisciplinary care with regular monitoring.

Diagnostic–treatment linkage referenced in class: Newborn IRT screen → sweat chloride or genotype confirms CF and guides variant-matched modulator consideration, alongside initiation of airway and nutritional care pathways.

TLDR Management combines variant-guided CFTR modulators (potentiators for gating; corrector±potentiator for F508del) with organ-system care: airway clearance and antibiotics for lungs, PERT and nutrition for pancreas or GI, and comprehensive monitoring. Minimal-function variants rely more on supportive care. Screening and genotyping steer therapy selection.


L6.3 Type 2 Diabetes

Objective 1. Recognise that DNA sequence is not the only determinant of phenotype

Core claim

  • T2D phenotype reflects genes + transcriptional regulation + epigenetics + environment + behaviour. DNA sequence sets liability, but regulation and exposure shape expression.

What your slides emphasise

  • Polygenic architecture: hundreds of loci with small effects; identified SNPs explain only a fraction of genetic predisposition, leaving “missing heritability.”
  • Transcriptional regulation: risk variants often act by altering gene expression in β-cells, adipose, liver, and muscle.
  • Epigenetics: DNA methylation and histone acetylation change DNA accessibility. Modifications can be environment-induced, heritable, persistent, and sometimes reversible.
  • Heterogeneity: same diagnosis, different pathways engaged (β-cell dysfunction versus insulin resistance).
  • Family risk: strong familial loading (for example higher lifetime risk with affected parent[s]) indicates genetics and shared environment.

Practical framing for answers

  • When asked “why sequence isn’t enough,” state:

    • Risk variants are mostly non-coding and tune transcription.
    • Epigenetic state integrates exposures across life (including in utero), shifting gene expression without changing sequence.
    • Population rise in T2D over decades outpaces genetic change, so environmental shifts must drive incidence.

Compact diagram

flowchart LR
  DNA[DNA sequence] -->|sets liability| GR[Gene regulation]
  Env[Environment & behaviour] -->|alters| Epig[Epigenetic state]
  Epig --> GR
  GR --> Phys[β-cell function & insulin sensitivity]
  Phys --> Phen[T2D phenotype]

TLDR Sequence sets baseline risk, but transcriptional regulation and epigenetics translate exposures into expression changes that determine β-cell output and insulin action. Rapid increases in T2D reflect environment and behaviour, not DNA sequence alone.


Objective 2. Understand that environmental factors can interact with genetic predisposition to trigger disease

Gene–environment interaction (G×E) in T2D

  • Adiposity is the strongest modifiable driver. Diet, inactivity, medications (for example long-term corticosteroids), and comorbidities shift insulin resistance and β-cell demand.
  • Environment shapes exposure: food availability, built environment, socioeconomic factors, stress, and norms influence diet and activity.
  • Genetics shapes response to environment: differences in taste preference, satiety signalling, resting metabolic rate, activity patterns, and microbiome mean the same environment yields different outcomes across genotypes.

How to reason through a case in exams

  1. State genetic liability (family history, PRS if given).
  2. Specify exposures (diet, activity, meds, sleep, stress, socioeconomic context).
  3. Link to pathways: exposure raises insulin resistance and lipotoxicity → greater β-cell workload → decompensation in genetically susceptible individuals.
  4. Add epigenetic dimension when appropriate (maternal hyperglycaemia, early-life nutrition).

Evidence threads to cite verbally

  • Family risks and twin or heritability ranges show a genetic component, but the 1990s–present incidence surge signals dominant environmental influence.
  • Population differences in risk reflect both allele frequencies and environmental exposures.

Minimal decision flow

High genetic liability + adverse environment → earliest onset / greatest risk
High genetic liability + supportive environment → buffered risk
Low genetic liability + adverse environment → still possible T2D (later onset)
Low genetic liability + supportive environment → lowest risk

TLDR Environment and behaviour modulate inherited risk. Obesity, diet, and inactivity push insulin resistance; susceptible genotypes fail β-cell compensation sooner. The same environment does not affect everyone equally because genetic and epigenetic context govern response.


Objective 3. Recognise that successful disease interventions do not have to target a specific gene but can act on the regulatory network

Key idea

  • In T2D, changing network state works even when you do not change DNA. You can target hormonal signalling, nutrient flux, chromatin state, organ cross-talk, and behavioural inputs to shift glycaemia, insulin sensitivity, and β-cell workload.

Network-level levers highlighted in lecture

Intervention class Primary network nodes System effects that matter for glycaemia
Lifestyle therapy (dietary pattern, caloric balance, physical activity, sleep) Energy balance, insulin signalling in muscle and liver, adipose inflammation, gut hormones ↓ insulin resistance, ↓ hepatic glucose output, improved glucose disposal; durable risk reduction when maintained
Metformin AMPK activation in liver and gut signalling ↓ hepatic gluconeogenesis, beneficial gut–liver crosstalk; lowers fasting glucose without changing DNA
GLP-1 receptor agonists Incretin signalling (islet–brain–gut axis) ↑ glucose-dependent insulin, ↓ glucagon, slower gastric emptying, reduced appetite and body mass; β-cell “rest”
SGLT2 inhibitors Renal glucose transport Glycosuria independent of insulin; lowers glucotoxicity and relieves β-cell stress; cardiorenal benefits
Thiazolidinediones (where used) PPARγ transcriptional control in adipose Re-partitioning of lipid, ↓ ectopic fat, improved insulin sensitivity
Bariatric/metabolic surgery Gut hormones, bile acids, nutrient flow Rapid incretin shifts, reduced insulin resistance, remission in subsets without gene editing
Epigenetic/behavioural milieu (weight loss, activity, sleep regularity) Chromatin accessibility, transcriptional programs Partly reversible methylation or acetylation states; sustained improvements in tissue insulin response

These act upstream, in parallel, or downstream of risk variants to change pathway flux and phenotypes, illustrating that pathway control is sufficient for clinical benefit.

flowchart LR
  Env[Lifestyle / Surgery / Drugs] --> Signals[Hormones & Nutrient Signals]
  Signals --> Reg[Transcriptional & Epigenetic Programs]
  Reg --> Tissues[Liver • Muscle • Adipose • Islet • Kidney • Gut]
  Tissues --> Outcomes[↓ Insulin resistance • ↓ Gluconeogenesis • ↓ Glucotoxicity]
  Outcomes --> Glyc[Improved glycaemia & risk]

How to write this in exams

  • Name the node (for example AMPK, GLP-1R, SGLT2, PPARγ, renal glucose transport).
  • State the pathway shift (for example ↓ hepatic glucose output, ↑ insulin in a glucose-dependent way, glycosuria).
  • Link to phenotype (lower HbA1c, less β-cell stress, weight loss, cardiorenal protection).
  • Close with “effective without altering DNA sequence,” aligning with the unit theme.

TLDR You do not need to edit genes to treat T2D. Interventions that act on regulatory networks, such as energy balance, AMPK, incretins, renal glucose handling, PPARγ programs, and gut–brain–liver axes, shift pathway flux to reduce insulin resistance, gluconeogenesis, and glucotoxicity. These network shifts deliver clinical benefit even though DNA sequence is unchanged.

WEEK 7

L7.1 Introduction to heritable vs non-heritable diseases

Objective 1. Distinguish between heritable and non-heritable diseases

Plain definitions

  • Heritable disease: caused by germline mutations present in the egg or sperm; mutation is in every cell of the offspring; can pass to the next generation. Examples: cystic fibrosis, sickle cell disease, some inherited cancers.
  • Non-heritable disease: caused by somatic mutations acquired after conception (environment, lifestyle, or random mitotic errors); affects only the individual; not passed to children. Examples: UV-induced skin cancer, many sporadic cancers.

What to compare in answers

Feature Heritable Non-heritable
Origin Germline mutation in egg or sperm Somatic mutation in body cells
Cell ploidy Gametes are haploid (23) Somatic cells diploid (46)
Transmission Passed via meiosis to offspring Not transmitted (no gamete involvement)
Drivers Single-gene, chromosomal, mitochondrial, de novo germline Mutagens (UV, chemicals, viruses), lifestyle, random mitosis errors
Examples CF, SCD, Huntington disease, familial cancers UV skin cancer, smoking-related lung cancer
Family pattern Often multi-generational Usually sporadic

Mutagen classes to name for non-heritable disease: physical (UV, X-rays), chemical (tobacco smoke, pollutants), biological (HPV). Their damage accumulates in somatic cells and can drive cancer.

TLDR Heritable = germline mutation, present in all cells, passed to children. Non-heritable = somatic mutation from environment, lifestyle, or random division, confined to the individual. Name origin, transmission route, and a matching example for full marks.


Objective 2. Explain how meiosis and mitosis contribute to germline and somatic mutations

Mitosis → somatic mutations (non-heritable)

  • What mitosis does: one division makes two genetically identical somatic cells for growth and repair.
  • Where mutations arise: replication errors, mis-segregation, DNA damage during the cell cycle.
  • Outcomes: clonal somatic variants; can drive cancer when they hit growth-control genes; not passed to offspring.

Meiosis → germline mutations (heritable)

  • What meiosis does: two divisions make four genetically unique haploid gametes (23 chromosomes). Diversity arises via independent assortment, crossing-over (recombination), and random fertilisation.

  • Where mutations and chromosomal errors arise:

    • Recombination errorstranslocations (e.g., Robertsonian; familial Down syndrome).
    • Copy-number errorsdeletions/duplications (e.g., 22q11.2 deletion, PMP22 duplication in CMT1A).
    • Base-level de novo germline mutations during gametogenesis.
  • Outcomes: variants are present in every cell of the child; can follow autosomal dominant/recessive, X-linked, or mitochondrial patterns.

Mini-schema to memorise

MITOSIS (somatic) → growth/repair → replication errors ↦ somatic mutations → cancer risk → not inherited
MEIOSIS (germline) → gametes/diversity → assortment + crossing-over ↦ germline mutations/CNVs → inherited

TLDR Mitosis errors create somatic mutations that can cause cancer but do not transmit. Meiosis generates variation for the next generation and, when errors occur, produces germline mutations or chromosomal rearrangements that are heritable. Name a diversity mechanism and a meiotic error with an example.

Objective: Describe key mechanisms of genetic inheritance and how meiosis promotes genetic diversity

Mechanisms of genetic inheritance (as taught)

  • Mendelian segregation: each parent contributes one allele per gene to offspring; alleles segregate during meiosis I. Use for trait-tracking and Punnett squares.
  • Autosomal dominant: one pathogenic allele causes disease; vertical transmission; M = F affected. Reduced penetrance and variable expressivity can blur patterns.
  • Autosomal recessive: disease when both alleles are pathogenic; often skips generations; increased risk with consanguinity.
  • X-linked: recessive usually affects males; carrier females may be variably affected (skewed X-inactivation). Dominant shows female bias but male lethality in some disorders.
  • Mitochondrial: maternal inheritance; all children of an affected mother inherit mtDNA, severity varies with heteroplasmy.
  • Non-Mendelian modifiers: penetrance, expressivity, de novo variants, and chromosomal changes (CNVs, translocations) alter observed pedigrees.

How meiosis promotes genetic diversity

  • Independent assortment (meiosis I): homologous chromosome pairs align independently. Gamete combinations ≈ (2^{n}) for n chromosome pairs. With 23 pairs, assortment alone yields a vast number of haploid combinations.
  • Crossing-over (homologous recombination): exchange between non-sister chromatids at chiasmata creates new haplotypes within chromosomes, breaking up linkage blocks. Frequency relates to genetic distance (map units/cM).
  • Random fertilisation: any sperm can fuse with any egg, multiplying assortment and recombination diversity.
  • Outcomes: unique gametes every meiosis; new allele combinations each generation; foundation for population-level variation and for mapping traits by recombination.

Quick exam workflow

  1. Identify the inheritance mode from pedigree features (AD/AR/X-linked/mitochondrial) and note any modifiers (penetrance, de novo, CNV).
  2. Link to meiosis: segregation explains allele transmission; assortment and recombination explain diversity and occasional unexpected haplotypes.
  3. If mapping is mentioned, relate recombination rate to genetic distance and LD blocks.

TLDR Inheritance follows allele segregation with modes AD, AR, X-linked, and mitochondrial, modulated by penetrance, expressivity, and structural variation. Meiosis generates diversity through independent assortment, crossing-over, and random fertilisation, yielding novel haplotypes and enabling recombination-based trait mapping.

Objective: Identify genetic, chromosomal, and mitochondrial causes of heritable diseases

1) Genetic (single-gene and small-scale sequence changes)

  • Autosomal recessive: two pathogenic alleles. Examples: cystic fibrosis (CFTR), phenylketonuria (PAH), sickle cell disease (HBB).
  • Autosomal dominant: one pathogenic allele. Examples: Huntington disease (HTT CAG repeat expansion), Marfan syndrome (FBN1).
  • X-linked: pathogenic variant on X. Examples: Duchenne muscular dystrophy (DMD), haemophilia A (F8).
  • Splicing/promoter/UTR variants: reduce correct transcript or expression. Examples: β-thalassaemia splice or promoter variants.
  • Nonsense or frameshift: premature stop, loss of function.
  • Missense: altered function or stability.
  • Imprinting defects: parent-of-origin expression errors. Examples: Prader–Willi, Angelman.
  • Repeat expansions: dynamic increases in short repeats. Examples: Fragile X (FMR1), Huntington (HTT).

2) Chromosomal (copy-number and structural)

  • Aneuploidy: whole-chromosome number change from meiotic nondisjunction.

    • Trisomy 21 (Down syndrome), trisomy 18, trisomy 13.
    • Sex chromosome aneuploidy: Turner (45,X), Klinefelter (47,XXY).
  • Microdeletions and microduplications: sub-microscopic CNVs.

    • 22q11.2 deletion syndrome.
    • Williams syndrome (7q11.23 deletion).
    • CMT1A (1.4–1.5 Mb PMP22 duplication).
  • Balanced rearrangements: reciprocal or Robertsonian translocations in a parent. May be phenotypically silent but produce unbalanced gametes and affected offspring.

  • Unbalanced structural changes: terminal or interstitial deletions/duplications that alter dosage of multiple genes.

3) Mitochondrial (mtDNA)

  • Maternal inheritance. Both sons and daughters can be affected. Only affected mothers transmit.

  • Heteroplasmy and threshold. Mix of normal and mutant mtDNA per cell. Disease manifests when mutant load exceeds a tissue-specific threshold.

  • Common examples:

    • LHON (Leber hereditary optic neuropathy; complex I subunits).
    • MELAS (m.3243A>G in MT-TL1).
    • MERRF (MT-TK).
  • Bottleneck in oogenesis drives variable heteroplasmy among siblings and tissues.

TLDR Heritable disease can arise from single-gene variants (AD, AR, X-linked, imprinting, repeat expansions), chromosomal errors (aneuploidy, CNVs, translocations), or mitochondrial mutations with maternal inheritance and heteroplasmy. Name class, mechanism, and one example.


Objective: Understand how mutations and environmental influences affect disease risk

A simple liability model

[ = + + + ]

Use this to structure any answer.

Genetic effects

  • Germline variants set baseline risk: monogenic, polygenic, or chromosomal.
  • Penetrance and expressivity determine presence and severity.
  • Modifier genes in the same pathway shift outcome in families with the same primary variant.

Environment and exposures

  • Diet and nutrients: folate status and neural-tube risk; low-phenylalanine diet prevents PKU disease; obesity and T2D onset.
  • Toxins and lifestyle: smoking and COPD or lung cancer; alcohol and liver disease.
  • Infections and microbiome: trigger or modulate immune and metabolic pathways.
  • Medications and hormones: corticosteroids increase insulin resistance; teratogens raise malformation risk.

Gene–environment interaction (G×E)

  • The same genotype yields different outcomes under different exposures.

  • The same exposure yields different outcomes across genotypes.

  • Examples:

    • α1-antitrypsin deficiency with smoking accelerates emphysema.
    • HBB Glu6Val plus malaria pressure maintains the HbS allele at high frequency due to heterozygote advantage.
    • High PRS for T2D plus adverse environment triggers earlier disease; supportive environment buffers risk.

Stochastic influences to mention

  • Random X-inactivation alters severity in X-linked disease.
  • Somatic mosaicism creates tissue-specific differences.
  • Developmental noise and epigenetic drift add person-specific variation.

Exam checklist

  • Identify variant class and inheritance.
  • Add environmental levers that raise or lower risk.
  • State one G×E interaction.
  • Mention penetrance/expressivity or stochastic factors if discordance is in the stem.

TLDR Mutations define baseline liability, but environment and G×E move people across the disease threshold. Add penetrance, modifier genes, and stochastic processes to explain variable expression, twin discordance, and shifting population risk.

L7.2 Chromosomal Abnormalities

Objective 1. Define chromosomal abnormalities and explain their role in human health

Definition

  • Any change in number or structure of chromosomes that alters normal development or physiology.
  • Human cells carry 46 chromosomes in 23 pairs. Errors in formation or segregation lead to disease.
  • Consequences include developmental delay, congenital anomalies, pregnancy loss, cancer risk, and variable organ involvement.

TLDR Chromosomal abnormalities are numerical or structural changes to chromosomes. They disrupt accurate genome distribution and lead to developmental and health problems across the lifespan.


Objective 2. Differentiate numerical and structural abnormalities with relevant examples

Numerical abnormalities

  • Trisomy: extra whole chromosome. Examples: Trisomy 21 (Down syndrome), Trisomy 18, Trisomy 13.
  • Monosomy: loss of a chromosome. Example: Monosomy X (Turner syndrome).
  • Mechanism: meiotic nondisjunction.

Structural abnormalities

  • Deletion: loss of a segment. Example: cri-du-chat (5p deletion).
  • Duplication: extra copy of a segment. Example: CMT1A from PMP22 duplication.
  • Translocation: segment moves to another chromosome. Examples: Philadelphia chromosome t(9;22) in CML; unbalanced translocations in some Down syndrome cases.
  • Inversion: segment flips within the same chromosome. Example on slides: hemophilia A case linked to inversion.
  • Mechanism: breakage and faulty repair or unequal recombination.

One-look comparison

Class What changes Typical mechanism Example(s)
Numerical Whole chromosome count Nondisjunction in meiosis Trisomy 21, 18, 13; Monosomy X
Structural A chromosome segment Breakage, misrepair, unequal recombination 5p deletion, PMP22 duplication, t(9;22), factor VIII inversion

TLDR Numerical errors add or remove entire chromosomes (trisomy, monosomy). Structural errors rearrange segments (deletions, duplications, translocations, inversions). Know one clear example for each.


Objective 3. Distinguish between heritable vs non-heritable chromosomal changes

Non-heritable (de novo)

  • Arise spontaneously in a gamete or early embryo.
  • Not present in either parent.
  • Recurrence risk is low for future pregnancies.
  • Includes most trisomies, many segmental deletions/duplications, and de novo unbalanced translocations.

Heritable

  • A parent carries a balanced rearrangement (e.g., balanced or Robertsonian translocation, inversion).
  • Parent is usually healthy but can produce unbalanced gametes.
  • Offspring risk for miscarriage or congenital anomalies is increased.

Exam sentences

  • “De novo CNV detected by microarray with normal parental studies → low recurrence.”
  • “Parental balanced Robertsonian translocation → increased risk of unbalanced conceptions; offer counselling.”

TLDR De novo changes originate in a gamete or early embryo and usually do not recur. Heritable changes come from a parent with a balanced rearrangement and carry recurrence risk due to unbalanced gametes. Cite one example of each in answers.

Objective 4. Describe key diagnostic methods for detecting chromosomal abnormalities

What to order and why

Test What it detects well When to use Caveats
Karyotype (G-banding) Whole-chromosome changes and large structural rearrangements; visualises balanced translocations First-line when you suspect aneuploidy, translocation, or infertility with possible Robertsonian/inversion carrier Lower resolution; misses small CNVs
FISH (targeted probes) Rapid confirmation of a suspected locus or aneuploidy; detects mosaicism in targeted cells Follow-up of a known or strongly suspected region; rapid prenatal or neonatal answers Only the probed region; not genome-wide
Chromosomal microarray (array CGH/SNP array) Genome-wide copy-number gains/losses and many microdeletions/duplications Developmental delay, congenital anomalies, autism spectrum, multiple malformations Does not detect balanced rearrangements
QF-PCR/targeted PCR Fast aneuploidy check for common trisomies Prenatal rapid screen or confirm with specific markers Limited to the loci assayed
NIPT (cell-free DNA) Screening for common fetal trisomies from maternal blood Population screening before invasive testing Screening only; positives need diagnostic confirmation
Invasive prenatal sampling Chorionic villus sampling (CVS) or amniocentesis to obtain fetal cells When NIPT/ultrasound suggests an abnormality or there is high a-priori risk Procedure risks; then run karyotype/CMA/FISH on the sample

These methods appear in your diagnostic flow: screen when appropriate, then confirm with a diagnostic test matched to the suspected class of abnormality.

TLDR Use karyotype for aneuploidy and balanced rearrangements, CMA for genome-wide CNVs, FISH/QF-PCR for rapid targeted questions, and NIPT as screening before CVS/amnio. Match the test to the suspected abnormality and confirm positives with a diagnostic assay.


Objective 5. Recognise the importance of early and accurate detection in improving health

Why timing and accuracy matter

  • Guides pregnancy care: early detection informs options, delivery planning, and neonatal support.
  • Directs therapy and surveillance: identify syndromes with cardiac, renal, endocrine, or neurodevelopmental risks and start targeted care plans.
  • Reduces diagnostic odyssey: early, correct diagnosis prevents unnecessary tests and delays.
  • Enables family counselling: clarifies recurrence risk; detects balanced carriers in parents when relevant.
  • Improves resource allocation: connects families to allied health, education support, and registries.

Safe reporting language (use in OSCE-style answers)

  • “This screening test suggests risk; we will confirm with a diagnostic test.”
  • “Results guide obstetric planning and postnatal specialty review.”
  • “If a parent is a balanced carrier, recurrence risk is higher; offer genetic counselling.”

TLDR Early, accurate detection changes outcomes: it shapes pregnancy decisions, starts targeted surveillance and interventions, shortens the diagnostic journey, and gives families clear recurrence information. Screen first when appropriate, then confirm with the right diagnostic test.

L7.3 Example of Heritable vs Non-heritable Polygenic Diseases

Objective 1. Differentiate between heritable and non-heritable genetic risk factors for breast cancer

One-look comparison

Dimension Heritable risk Non-heritable (polygenic + somatic) risk
Genetic architecture High-penetrance single genes, classically BRCA1/BRCA2 Many low-penetrance variants across the genome, plus somatic mutations acquired over life
Pattern Runs in families; mutation present in every cell from birth Often no clear family history; risk emerges from the sum of small effects and exposures
Share of total cases ~5–10% of all breast cancers Majority of cases
Typical quantification Pathogenic-variant status (e.g., BRCA1/2) Polygenic Risk Score (PRS) plus environment and age
Prevention focus Genetic counselling, enhanced screening, risk-reducing surgery when appropriate Risk stratification for screening, lifestyle modification, manage exposures
Treatment implications Sensitivity to DNA-damaging agents and PARP inhibitors Standard of care guided by tumour features; PRS informs population screening rather than therapy choice

TLDR Heritable breast cancer reflects high-penetrance variants such as BRCA1/2 and accounts for ~5–10% of cases. Non-heritable risk dominates and comes from many small-effect variants plus environmental exposures; PRS summarises the inherited polygenic part and supports screening and prevention planning.


Objective 2. Explain how BRCA1 and BRCA2 mutations increase cancer risk and guide prevention strategies

Biology you must state

  • BRCA1 and BRCA2 are tumour suppressors that repair DNA double-strand breaks by homologous recombination. They maintain genomic stability and prevent mutation accumulation. Mutations impair repair, driving genomic instability and cancer risk.

Risk figures to remember

  • Heritable breast cancer share: ~5–10% of all cases.
  • Lifetime risk: BRCA1 ~55–72%; BRCA2 ~45%. These values guided the lecture examples and prevention framing.

How BRCA status guides prevention and care

  • Genetic testing and counselling for individuals with suggestive personal or family history.
  • Enhanced screening pathways when positive (earlier start, higher frequency, MRI where indicated).
  • Risk-reducing surgery discussion for high-risk carriers (e.g., bilateral mastectomy; salpingo-oophorectomy based on age and risk).
  • Therapeutic implications in cancer: tumours with BRCA loss often show heightened response to platinum chemotherapy and to PARP inhibitors that exploit homologous recombination deficiency.

Exam phrasing

  • BRCA1/2 normally mediate HR repair. Pathogenic variants cause HR deficiency, raising lifetime risk. A positive result prompts counselling, intensified screening, consideration of risk-reducing surgery, and, if cancer occurs, supports PARP or platinum-based regimens.”

TLDR BRCA1/2 safeguard the genome by HR repair. Pathogenic variants disable this function, causing genomic instability and large lifetime risk increases. Management: test and counsel, intensify screening, consider risk-reducing surgery, and use HR-defect-targeted therapies when cancer is present.

Objective 3. Describe how Polygenic Risk Scores assess non-heritable risk across populations

What PRS is doing here

  • PRS summarises common, small-effect germline variants into a single score to rank relative breast-cancer risk across a population. It complements, but does not replace, high-penetrance genes.

Calculation (recap)

[ =_{i=1}^{m}_i G_i]

  • ( _i ) = GWAS effect size for SNP (i); (G_i) = count of risk alleles (0,1,2). Scores are standardised to a reference cohort to give percentiles.

How it is used across populations

  • Risk stratification: higher PRS percentiles → higher relative risk; used to tailor screening intensity and age of initiation.
  • Portability caveat: accuracy depends on ancestry match between the person and the GWAS used for weights; transfer across ancestries degrades calibration. Use ancestry-matched references.

Strengths and limits

Strength Limit
Captures polygenic liability invisible to single-gene tests Not diagnostic; gives relative risk only
Helps target scarce screening resources Sensitive to ancestry mismatch
Can combine with age, family history, lifestyle Does not include environment unless explicitly modelled

TLDR PRS is a weighted sum of many common variants that ranks relative risk and helps stratify screening. It requires an ancestry-matched reference and does not diagnose disease.


Objective 4. Analyse the impact of Angelina Jolie’s case on public awareness, genetic testing and preventative medicine

What happened and why it mattered

  • A high-profile BRCA1 carrier publicly described risk-reducing surgery, which increased public awareness, referrals for genetic counselling, and uptake of testing among eligible families; it also normalised discussion of risk-reducing mastectomy and oophorectomy in appropriate carriers.

Teaching points from the lecture

  • Positive effects: earlier identification of high-risk carriers; timely prevention planning; broader understanding of tumour-suppressor genetics in the public.
  • Operational cautions: ensure referrals are criteria-based; avoid inappropriate testing in low-risk individuals; provide pre-/post-test counselling to set expectations.

TLDR The “Angelina effect” boosted awareness, counselling referrals, and evidence-based prevention for true high-risk carriers, with the caveat to maintain criteria-based testing and robust counselling.


WEEK 8

L8.1 Coding vs Non-coding DNA

Objective 1. Define non-coding DNA and understand its characteristics

Definition

  • Coding DNA produces protein-coding genes. Non-coding DNA comprises DNA that does not encode proteins. In humans, about 98% of the genome is non-coding.

Core characteristics from the lecture

  • Widespread across the genome, including introns, telomeres, satellite DNA, non-coding RNA genes, and gene-regulatory sequences such as promoters, enhancers, and silencers.
  • Controls gene expression and RNA fate. Non-coding regions influence transcription, splicing, mRNA stability, export, and localisation. Introns retained in mRNA can trigger nonsense-mediated decay (NMD).
  • Shapes chromosome integrity and lifespan. Telomeres cap chromosome ends, prevent fusion, and relate to cellular ageing.
  • Contributes to chromosome structure. Satellite DNA supports centromere and kinetochore function and heterochromatin formation.
  • Mutations in non-coding DNA can alter TF binding, enhancer–promoter communication, or non-coding RNA production, leading to disease.

TLDR Non-coding DNA is the majority of the human genome. It regulates genes and RNAs, preserves chromosome integrity, and organises chromatin. Changes in these regions can shift expression programs and cause disease.


Objective 2. Describe the different types of non-coding DNAs

The landscape

Class Where it sits What it does Disease links from lecture
Introns Within genes between exons Control expression, RNA stability, export, localisation. Provide sequences for non-coding RNAs. Intron retention can trigger NMD and repress genes. Mutations at splice dinucleotides cause intron retention and gene silencing, including in cancers; muscular dystrophy examples noted.
Telomeres Chromosome ends Protect ends, prevent end-to-end fusion, maintain stability, relate to replicative lifespan. Short telomeres raise risk of CVD, dementia, cancer, osteoporosis; excessively long telomeres associate with some cancers.
Satellite DNA Intergenic, pericentromeric, subtelomeric, intronic arrays Builds centromere/kinetochore, supports spindle assembly, drives heterochromatin and genome organisation. Repeat changes can disrupt centromeres → missegregation and genomic instability, contribute to cancer; developmental disorders discussed, with Fragile X example on slides.
Non-coding RNA genes Discrete loci Encode tRNA, rRNA, miRNA and other RNAs that act structurally or regulatory. Dysregulation contributes to disease mechanisms; set up for later modules in your course.
Gene-regulatory sequences Promoters upstream of genes; enhancers and silencers can be distal Promoters bind TFs to initiate transcription. Enhancers facilitate promoter interactions. Silencers inhibit them. DNA loops enable distal contacts. Mutations reduce or increase TF binding or enhancer–promoter communication, linked to cancer, diabetes, neurological and developmental disorders. Parkinson’s enhancer SNP example shown.

Concept diagram you can sketch quickly

Genome
├─ Coding DNA → proteins
└─ Non-coding DNA
   ├─ Introns → splicing, RNA fate, NMD
   ├─ Telomeres → end protection, ageing
   ├─ Satellite DNA → centromere, heterochromatin
   ├─ ncRNA genes → tRNA, rRNA, miRNA
   └─ Regulatory DNA → promoters, enhancers, silencers (TF binding, looping)

TLDR Non-coding DNA comprises introns, telomeres, satellite repeats, non-coding RNA genes, and regulatory elements. Each class has a distinct role in expression control or chromosome biology. Mutations in any class can perturb regulation or chromosome stability and lead to disease.


Objective 4. Understand the importance of studying non-coding DNA mutations/alterations in human health and medicine

Why this matters clinically and scientifically

  • Explains missing heritability: many GWAS hits lie in regulatory DNA. Knowing which enhancer or promoter is causal clarifies biology and risk.

  • Improves diagnostics: add non-coding variant interpretation when coding tests are negative; evaluate splice-site and promoter variants; report intron-retention mechanisms.

  • Guides therapy:

    • ASOs to correct splicing or reduce toxic transcripts.
    • CRISPRi/CRISPRa or small molecules to tune enhancers or promoters for dose correction.
    • Telomere maintenance and chromatin drugs in selected cancers.
  • Stratifies prevention: regulatory variants in metabolic or neurodegenerative pathways refine risk models.

  • Safety in gene therapy: avoid creating harmful enhancer contacts or disrupting insulators.

Practical workflow to mention

  1. Prioritise non-coding variants that overlap TF motifs, chromatin marks, or splice signals.
  2. Use RNA evidence: aberrant splicing, intron retention, or expression changes.
  3. Link to phenotype and, where relevant, propose modality (ASO, expression tuning).

TLDR Studying non-coding changes sharpens diagnosis, reveals mechanisms behind GWAS signals, and opens therapeutic levers that tune expression or splicing rather than protein sequence. Include non-coding analysis when coding tests are unrevealing.


L8.2 Non-coding mutations in thalassaemia

Objective 1. Describe the genetic cause of α- and β-thalassaemia

Loci and allele counts

  • α-globin: two α-genes (HBA1, HBA2) on chr16, so four α-alleles per diploid genome.
  • β-globin: one β-gene (HBB) on chr11, so two β-alleles.

α-thalassaemia, usually deletions

Genotype pattern Typical label Molecular basis Clinical pattern
aa/aa Unaffected All four α present Normal
−a/aa Silent carrier Deletion of one α-gene Asymptomatic
−−/aa or −a/−a α-thalassaemia trait Two-gene deletion, either cis (−−/aa) or trans (−a/−a) Mild microcytic anaemia
−−/−a HbH disease Three-gene deletion Moderate–severe anaemia; HbH (β₄) present
−−/−− Hydrops fetalis Four-gene deletion Usually lethal in utero

β-thalassaemia, usually point mutations (often non-coding)

  • Promoter variants reduce transcription initiation.
  • Intronic/non-coding variants reduce RNA stability or processing.
  • Splice-site variants (5′ GT / 3′ AG and nearby motifs) cause exon skipping, intron retention, or cryptic splice use.
  • β⁰: no β-globin made. β⁺: reduced β-globin.
  • Large deletions are uncommon.

β-thalassaemia genotype → label

Genotype examples Label Phenotype
β/β Unaffected Normal
β⁺/β or β⁰/β Minor (trait) Asymptomatic to mild anaemia
β⁺/β⁺ or β⁰/β⁺ Intermedia Mild–moderate; usually non-transfusion dependent
β⁰/β⁰ Major Severe anaemia; transfusion dependent

TLDR α-thal is mainly gene deletions on chr16 (1→4 genes lost; cis/trans matters). β-thal is mainly non-coding point mutations in promoter/introns/splice sites of HBB on chr11, classified β⁰ (none) or β⁺ (reduced). Genotype sets clinical class from silent carrier to trait, HbH, hydrops (α) and from trait to intermedia to major (β).


Objective 2. Explain how globin-chain imbalances drive pathophysiology, and why combined α+β thalassaemia is milder

Chain-imbalance mechanism

  1. Reduced synthesis of one chain (α or β) creates excess unpaired partner chains.
  2. Unpaired chains precipitate in erythroid precursors, damaging membranes.
  3. Precursors undergo apoptosis (ineffective erythropoiesis); circulating RBCs are fragile and undergo hemolysis.
  4. Result: microcytic, hypochromic anaemia, marrow expansion, extramedullary haematopoiesis, splenomegaly, jaundice from increased bilirubin.

Direction of excess chains

  • β-thal: too few β chains → excess α chains precipitate; severe membrane damage.
  • α-thal: too few α chains → excess β chains form unstable β₄ (HbH) tetramers; poor oxygen carriage and membrane injury.

Why co-inheritance (α + β thal) can reduce severity

  • When both production streams are down, the pool of unpaired chains shrinks.
  • Fewer precipitates mean less ineffective erythropoiesis and less hemolysis, so the phenotype shifts toward milder anaemia.
Pathway sketch you can reproduce fast
↓One chain  →  Excess partner chains  →  Precipitation in precursors
                                        ↓
                              Membrane injury, apoptosis (ineffective erythropoiesis)
                                        ↓
                          Few, fragile RBCs → hemolysis → anaemia, hypoxia

TLDR Imbalance drives disease: the surplus chain aggregates, injures precursors (ineffective erythropoiesis), and shortens RBC survival (hemolysis). In β-thal, α-excess is toxic; in α-thal, β-excess forms unstable HbH. Co-inheritance of α + β thal balances chain supply, so fewer unpaired chains and milder disease.


Objective 3. Compare and contrast haemoglobin composition in unaffected vs thalassaemia patients

Key patterns to memorise

  • β-thalassaemia: HbA2 and HbF rise as β-chain output falls. Hallmark for β-thal is ↑HbA2 with ±↑HbF.
  • α-thalassaemia: HbA2 and HbF are usually normal; HbH (β4) appears in HbH disease. DNA testing is needed when electrophoresis looks “normal”.

Haemoglobin subtype table (from your slide)

Phenotype HbA (~97%) HbF (~1%) HbA2 (~2%) HbH
Unaffected ~97 ~1 ~2
β-thal major (β⁰/β⁰) 0 up to 95 >5
β-thal intermedia (β⁰/β⁺) 10–30 70–90 >4
β-thal minor/trait (β⁺/β) >88 <5 >4
HbH disease (−−/−α) 60–90 ~1 ~2 1–40
α-thal trait (−α/−α) >95 ~1 ~2

TLDR β-thal: think ↑HbA2 ± ↑HbF. α-thal: electrophoresis often normal, unless HbH is present. Use DNA tests to confirm α-thal trait.


Objective 4. Use CBC, ferritin, and electrophoresis to identify thalassaemia and distinguish it from other microcytic anaemias

Stepwise diagnostic approach (what the lecture taught)

  1. CBC + smear

    • Microcytic, hypochromic anaemia. Smear may show target cells, anisopoikilocytosis, ± nucleated RBCs in more severe disease.
  2. Iron studies

    • Normal or elevated ferritin supports thalassaemia rather than iron deficiency when microcytosis is present.
  3. Haemoglobin electrophoresis

    • β-thal: ↑HbA2 (>3.5%) and ↑HbF.
    • α-thal: usually normal HbF/HbA2; HbH may appear in HbH disease.
  4. If α-thal suspected with normal electrophoresis

    • Proceed to DNA testing for α-gene deletions (cis/trans).

Differentiating microcytic anaemias quickly

  • Thalassaemia vs iron deficiency

    • Both microcytic/hypochromic on CBC/smear. In thalassaemia, ferritin is normal/high; in iron deficiency, ferritin is low. Then use electrophoresis patterns above.
  • α vs β thalassaemia

    • β-thal trait/intermedia/major shows ↑HbA2 ± ↑HbF with severity gradient. α-thal trait is electrophoresis-normal, HbH disease shows HbH band.

Pathophysiology tie-in for viva answers

  • Imbalanced chains cause ineffective erythropoiesis and haemolysis, explaining microcytosis and anaemia severity.

TLDR Start with CBC/smear → microcytic, hypochromic. Check ferritin: normal/high points to thalassaemia. Use electrophoresis: β-thal = ↑HbA2 ± ↑HbF, α-thal = usually normal, HbH if HbH disease. Confirm α-deletions by DNA when electrophoresis is non-diagnostic.


L8.3 Non-coding RNA diseases

Objective 1. Define non-coding RNA (ncRNA) and understand its characteristics

Definition

  • ncRNAs are RNA molecules that do not encode proteins yet are functional in the cell. Two size classes: small (<200 nt) and long (lncRNA, >200 nt). They can be regulatory or housekeeping/structural.
  • Regulatory ncRNAs act at chromatin, transcriptional, and post-transcriptional levels; they localise to nucleus and/or cytoplasm.

General characteristics highlighted in the lecture

  • Small ncRNAs: include microRNAs (miRNAs) that regulate gene expression post-transcriptionally.
  • Long ncRNAs (lncRNAs): >200 nt; functions include chromatin remodelling, transcriptional activation/repression, RNA splicing, protein scaffolding, subcellular architecture, protein localisation/transport, translational control, miRNA sponging, and serving as precursors for other small RNAs.
  • miRNA biogenesis and action: pri-miRNA → pre-miRNA (nucleus) → export → Dicer processing → loaded into RISC → binds 3′UTR of targets (partial complementarity) to induce mRNA cleavage, translation repression, or deadenylation.
  • Origins: miRNAs can derive from introns, exons, or other ncRNA loci (e.g., snoRNA-derived).
  • Breadth: miRNAs occur across plants and animals; the slide cites miRBase v22.1 count (~38k entries).

TLDR ncRNAs are functional RNAs that do not code for protein. They are grouped into small and long classes and control gene expression at multiple levels. miRNAs act via RISC at 3′UTRs to reduce protein output; lncRNAs regulate from chromatin to translation.


Objective 2. Provide examples of non-coding RNAs and their functions

A. microRNAs (miRNAs)

  • Core function: 18–25 nt single-stranded RNAs that down-regulate gene expression post-transcriptionally via RISC at 3′UTRs.
  • Biogenesis: pri-miRNA → pre-miRNA → cytoplasmic processing → RISC loading; act with partial complementarity to many targets.
Disease-linked examples from the lecture
  • miR-125b and Fragile X syndrome: interacts with FMRP and binds the PSD-95 mRNA 3′UTR, suppressing translation and impairing synaptic function.
  • miR-125b and Type 2 diabetes: negative regulator of insulin secretion in β-cells; increased miR-125b lowers glucose-stimulated insulin release/content.
  • Broader panels show many cardiometabolic and oncologic miRNAs (oncomiRs and tumour-suppressor miRs) dysregulated across diseases.

B. Long non-coding RNAs (lncRNAs)

  • Definition: >200 nt; nuclear or cytoplasmic; multifunctional regulators (chromatin, transcription, splicing, scaffolding, localisation, translation, miRNA sponge, small-RNA precursor).
Disease-linked examples from the lecture
  • Atherosclerosis

    • H19: overexpression → lipid accumulation, inflammation, cell proliferation; promotes plaque instability and rupture.
    • ANRIL: overexpression → vascular cell proliferation and arterial wall thickening.
  • Diabetes and complications

    • MALAT1: regulates β-cell differentiation/survival/function; upregulated in T1DM, inhibiting insulin secretion.
    • Reg1cp: SNP/mutation increases T2DM risk via insulin-resistance mechanisms.
    • RPL13p5: upregulated in GDM, promotes insulin resistance.
    • Diabetic kidney disease: aberrant H19 expression associated with pathology.
    • Diabetic retinopathy: MIAT upregulated (retinal inflammation and cell death); NEAT1 promotes pathologic retinal angiogenesis with fragile, leaky vessels.

TLDR miRNAs repress target mRNAs post-transcriptionally (e.g., miR-125b in Fragile X and β-cell insulin secretion). lncRNAs regulate from chromatin to translation; examples include H19 and ANRIL in atherosclerosis, MALAT1/Reg1cp/RPL13p5 in diabetes, and MIAT/NEAT1 in diabetic retinopathy.


Objective 3. Understand that abnormal changes to non-coding RNAs are implicated in human disease

Mechanisms of pathogenicity

  • Dosage change: over- or under-expression alters pathway flux (e.g., cell cycle, inflammation).
  • Sequence/structure change: seed or binding motif changes rewire targets.
  • Processing defects: pri→pre→mature steps fail, so functional species drop.
  • Localization change: nuclear vs cytoplasmic shift alters which processes are regulated.
  • Competing interactions: lncRNAs and circular RNAs can sponge miRNAs, changing effective miRNA dose.

Disease examples mapped to mechanism

ncRNA Change Primary effect Disease context (from lecture)
miR-125b Overexpression Represses synaptic PSD-95; impairs β-cell insulin release Fragile X synaptic dysfunction; reduced glucose-stimulated insulin secretion in T2D models
H19 (lncRNA) Overexpression Pro-proliferative, pro-inflammatory programs Atherosclerotic plaque growth and instability
ANRIL (lncRNA) Overexpression Vascular cell cycle activation Arterial thickening, atherosclerosis
MALAT1 (lncRNA) Up in diabetes β-cell dysfunction; insulin secretion block T1D/T2D pathology contributions
MIAT / NEAT1 (lncRNAs) Up in retina Inflammation, abnormal angiogenesis Diabetic retinopathy (fragile, leaky vessels)

Why small changes matter

  • Each miRNA targets many mRNAs. Modest fold-changes propagate through networks and produce measurable phenotypes. lncRNAs act as scaffolds, guiding chromatin and protein complexes, so mis-expression shifts entire programs.

TLDR ncRNA alterations cause disease by changing dose, sequence, processing, localization, or sponging. Small shifts at the ncRNA layer reprogram networks and drive pathology in neuro, cardiometabolic, and ocular settings (e.g., miR-125b, H19, ANRIL, MALAT1, MIAT, NEAT1).


Objective 4. Understand the importance of studying non-coding RNA mutations/alterations in human health and medicine

Clinical value

  • Diagnostics: add ncRNA analysis when coding tests are negative. Use expression panels or single-ncRNA assays as biomarkers for risk, diagnosis, and activity.

  • Prognosis and stratification: ncRNA signatures separate patient subgroups and predict progression (e.g., vascular risk, retinopathy progression).

  • Therapeutic targets:

    • miRNA inhibitors (antagomiRs) to block pathogenic miRNAs.
    • miRNA mimics to restore lost repression.
    • ASOs or siRNAs against pathogenic lncRNAs.
    • CRISPRi/CRISPRa at ncRNA promoters or enhancers to tune dose.
  • Monitoring: circulating ncRNAs enable minimally invasive follow-up.

Practical study workflow from the lecture

  1. Prioritise candidates by overlap with disease tissues and pathways.
  2. Measure expression and processing (pri/pre/mature miRNA; lncRNA isoforms).
  3. Validate direct targets (reporters, pulldowns, CLIP-style evidence when available).
  4. Perturb with gain/loss tools and read out pathway and phenotype.
  5. Translate to clinic: assay performance, stability, and delivery.

Caveats to state in exams

  • Specificity: miRNAs have many targets; off-target risk exists.
  • Delivery: tissue targeting and stability limit in-vivo therapies.
  • Context: ncRNA function is cell-type and state dependent; results may not transfer across tissues or ancestries without recalibration.

TLDR Studying ncRNAs improves diagnosis, risk stratification, and therapy design. Use antagomiRs, mimics, ASOs, or CRISPR-based tuning to correct pathogenic ncRNA programs, with careful attention to specificity, delivery, and context.

WEEK 9

L9.1 Precision Medicine

Objective 1. Describe “precision medicine” and the role of genetics

Definition

  • Precision medicine tailors therapy to the individual using clinical, environmental, and genomic data to pick the right drug, right dose, right patient, right time.

Why it is needed

  • Drug response shows a population distribution from minimal to excessive effect. Many patients sit outside the average dose window and need adjustment. Precision approaches reduce non-response and toxicity.

Role of genetics

  • PK layer: variants in metabolic enzymes and transporters shift exposure and half-life.
  • PD layer: variants in drug targets alter binding and downstream signalling.
  • Genomic information separates poor, intermediate, normal, and ultra-rapid phenotypes for dosing and selection.

TLDR Precision medicine individualises therapy using genomics with clinical context. Genetics affects PK through enzymes and transporters and PD through targets, which guides drug and dose to maximise benefit and minimise harm.


Objective 2. Outline key pharmacology core concepts relevant to precision medicine

The three pillars to name in exams

  1. Pharmacokinetics (PK) — movement of drug in the body

    • ADME: Absorption, Distribution, Metabolism, Elimination.
    • PK parameters: bioavailability, clearance, half-life, volume of distribution, steady state, first vs zero-order kinetics.
  2. Pharmacodynamics (PD) — what drug does to the body

    • Drug–target interaction, mechanism of action, selectivity, affinity, efficacy, potency.
    • Dose–response curves, therapeutic window, agonist vs antagonist.
    • Structure–activity relationships and binding pockets determine response and genomic sensitivity.
  3. Integrative/system concepts

    • Inter-individual variability, drug interactions, adverse drug reactions, tolerance. These modulate PK and PD at the bedside.

Where genetics fits inside the pillars

Pillar Genetic levers Consequence for practice
PK: Metabolism Phase I enzymes, mainly CYPs: CYP2C9, CYP2C19, CYP2D6, CYP3A4/5; plus FMOs, ADH, etc. Alters activation/inactivation rate, exposure, and half-life. Dose selection and contraindications.
PK: Transport ABCB1 (P-gp), ABCG2 (BCRP), OATP1B1 (SLCO1B1) Changes absorption, tissue entry, biliary/renal efflux. Predicts toxicity for substrates like statins or digoxin.
PD: Targets Receptor or enzyme variants, especially GPCRs and other targets 5-fold potency differences observed with common missense variants; informs selection of class or dose.

Linking PK and PD to clinical optimisation

  • Build the exposure–response picture: administer → concentration–time → target binding → effect, then balance benefit and ADRs within the therapeutic window. Use genomics where it shifts this curve.

TLDR Precision work uses PK, PD, and systems concepts. In PK, ADME and parameters drive exposure and are gene-sensitive through CYPs and transporters. In PD, target variation shifts binding, potency, and efficacy. Integrate these with variability, interactions, and ADRs to personalise therapy.


Objective 3. Explain core pharmacodynamic and pharmacokinetic concepts of drugs

Pharmacokinetics (PK): movement of drug

ADME and key parameters

  • Absorption: extent and rate into blood. Bioavailability (F).
  • Distribution: into tissues. Volume of distribution (V_d).
  • Metabolism: biotransformation (Phase I, II).
  • Elimination: renal + biliary. Clearance (CL). Half-life (t_{1/2}).

Equations you should quote

  • (t_{1/2}=0.693V_d/CL)
  • Loading dose (=)
  • Maintenance dosing rate (=)
  • Steady state in ~4–5 half-lives (first-order).

First- vs capacity-limited elimination

  • First-order: constant fraction cleared per time; linear PK.
  • Zero-/mixed-order: pathways saturated near therapeutic range; disproportionate exposure rise with dose.

Organ determinants

  • Hepatic: intrinsic clearance, enzyme content, extraction ratio.
  • Renal: filtration, secretion, reabsorption, fraction unbound (f_u).

Where genetics acts in PK

  • Enzymes (CYP2C9, 2C19, 2D6, 3A, UGT1A1) alter CL and (t_{1/2}).
  • Transporters (OATP1B1, P-gp/ABCB1, BCRP/ABCG2) alter absorption, hepatic uptake, biliary/renal efflux.

Pharmacodynamics (PD): what drug does

Dose–response

  • Potency (EC({50})) vs efficacy (E()).
  • Emax/Hill model: (E=).
  • Therapeutic window: balance effect and toxicity.

Ligand classes

  • Full/partial/inverse agonists; competitive antagonists shift EC({50}) right without lowering E(); non-competitive reduce E(_). Tolerance/desensitisation, spare receptors.

Where genetics acts in PD

  • Target variants (e.g., GPCR missense) change affinity/efficacy, shifting curves at a given concentration.

TLDR PK sets exposure (ADME, (CL), (V_d), (t_{1/2}), (F)); PD links concentration to effect (potency, efficacy, therapeutic window). Genetics shifts PK via enzymes/transporters and PD via targets, so equations and curves move for different genotypes.


Objective 4. Detail exemplar genetic variants that contribute to altered pharmacology of drugs

Gene / variant (phenotype) Drug(s) Mechanism → clinical effect Action from lecture
CYP2C19 2/3 (loss), *17 (gain) Clopidogrel Prodrug activation depends on CYP2C19. LoF → ↓ active metabolite → ↓ antiplatelet effect; *17 → ↑ activation → bleeding risk Avoid clopidogrel in PM/IM; consider alternatives. Dose/bleeding caution in *17.
CYP2D6 gene duplications (UM) or LoF (PM) Codeine, tramadol Codeine/tramadol need 2D6 for active opioid formation. UM → toxicity; PM → no analgesia Prefer non-2D6 opioids in UM/PM.
CYP2C9 2/3 and VKORC1 −1639G>A Warfarin 2C9 LoF ↓ S-warfarin clearance; VKORC1 A lowers target expression → higher sensitivity Use genotype-guided lower initial doses; careful INR titration.
SLCO1B1 c.521T>C (OATP1B1↓) Simvastatin (statins) ↓ Hepatic uptake → ↑ plasma exposure → myopathy risk Avoid high-dose simvastatin; choose alternative or lower dose.
DPYD decreased-function alleles 5-FU/capecitabine DPD deficiency → ↓ catabolism → severe toxicity Reduce/avoid; titrate to tolerance when variants present.
UGT1A1 28/6 Irinotecan ↓ Glucuronidation of SN-38 → neutropenia/diarrhoea Start lower dose in high-risk genotypes.
TPMT or NUDT15 LoF Azathioprine/6-MP ↓ Inactivation or altered nucleotide sanitation → myelosuppression Use reduced doses or alternative; monitor counts closely.
**HLA-B*57:01** Abacavir Immune presentation of drug–peptide → hypersensitivity Screen and avoid if positive.
**HLA-B*15:02** (ancestry-specific risk) Carbamazepine T-cell–mediated SJS/TEN risk Screen in high-risk ancestries; avoid if positive.

Transporter and target notes

  • ABCB1 (P-gp) and ABCG2 (BCRP) variants shift CNS entry and biliary/renal clearance of substrates in smaller, drug-specific ways.
  • Target variants in GPCRs and enzymes can shift **EC(_{50})** or **E(_)** by several-fold, altering class choice or dose.

How to write genotype-guided dosing in exams

  1. State gene–drug pair and phenotype.
  2. Name the mechanism (activation, clearance, transport, immune risk).
  3. Give the action (avoid, lower dose, use alternative, monitor).
  4. Tie back to PK/PD: exposure or potency shift moves the dose–response curve.

TLDR Know the anchor pairs: CYP2C19–clopidogrel, CYP2D6–codeine, CYP2C9/VKORC1–warfarin, SLCO1B1–simvastatin, DPYD–5-FU, UGT1A1–irinotecan, TPMT/NUDT15–thiopurines, HLA-B57:01–abacavir, HLA-B15:02–carbamazepine. For each, name mechanism and adjust drug or dose accordingly.


L9.2 Intro to Genomic Drug Design & Development

Objective. Outline the role of genomics across the drug-development pipeline

One-page view (pipeline × genomics)

Stage What genomics adds Concrete tools/examples
Disease biology → Target ID Finds causal genes, pathways, variants; shows which tissues and cell types matter GWAS to nominate loci; NGS and single-cell sequencing to map expression and cell states; variant–function links to nominate targets
Target validation Tests necessity/sufficiency of genes and variants CRISPR knock-out/knock-in; rescue assays; pathway rewiring to confirm on-target effects
Lead discovery / optimisation Aligns chemistry with variant-defined binding pockets and allosteric sites; anticipates resistance In-silico modelling of mutant proteins; SAR guided by variant maps; avoid metabolism-prone motifs based on enzyme knowledge
Preclinical (PK/PD, tox) Predicts metabolism and transport; flags toxicity risks across genotypes Map substrates to CYPs (2D6, 2C19, 3A4/5) and transporters (OATP1B1, P-gp, BCRP); toxicogenomics panels
Clinical trials Designs genotype-aware trials; improves power and safety Enrichment/stratification by pharmacogene or target genotype; genotype-guided dosing arms (e.g., CYP2C19–clopidogrel)
Post-approval Explains variable effectiveness and rare AEs; updates labels Pharmacovigilance signals traced to variants (e.g., **HLA-B*57:01–abacavir**); real-world PGx implementation and label changes

Quick pipeline diagram

Genomic discovery → Target validation → Lead/SAR → Preclinical PK/PD → Genotype-aware trials → Post-market PGx + safety
      (GWAS, scRNA-seq)         (CRISPR)        (mutant structures)     (CYPs/transporters)     (enrich/stratify)     (labels, RWE)

Case anchors from the lecture

  • Breast cancer subtyping: ER/PR, HER2 status directs tamoxifen/aromatase inhibitors or trastuzumab.
  • Warfarin: VKORC1 and CYP2C9 genotypes guide starting dose in a narrow window.
  • Abacavir: **HLA-B*57:01** screening prevents hypersensitivity.

Implementation challenges to name

  • Data integration/standardisation across platforms.
  • Regulatory clarity for validation and clinical use.
  • Collaboration and infrastructure to share and analyse multi-omics at scale.
  • Ethics: privacy, consent, and equity of access.

TLDR Genomics informs every stage: it finds and validates targets, shapes SAR against variant-defined proteins, predicts PK/PD via enzymes and transporters, stratifies trials, and guides post-market safety and dosing. Examples: ER/PR/HER2 therapy selection, VKORC1/CYP2C9–warfarin, **HLA-B*57:01–abacavir**.


Objective. Discuss the genomic basis that informs design of molecular-targeted therapies

1) Start from genotype → structure → function

  • Mutation class changes the 3D pocket or allosteric network of a target.
  • Structural consequences guide whether to design orthosteric, allosteric, or covalent ligands and what to avoid in SAR.
  • Use variant-resolved models (crystal, cryo-EM, AlphaFold-refined) to see pocket volume, electrostatics, waters, and gatekeeper residues.

2) Recurrent design patterns you should name

Genomic change in target Structural/biophysical effect Design tactic What to watch
Activating missense in the active site or switch loop Pocket reshapes, altered H-bond network, constitutive signaling Fit the new pocket; exploit mutant-specific cavities for selectivity Keep WT off-target low; avoid steric clash in WT
Gatekeeper mutation in kinases Blocks first-gen inhibitors Add narrow hinge vectors or covalent warheads to bypass steric block Balance reactivity with selectivity
Allosteric mutations Shift inactive/active equilibrium Target allosteric pockets to re-weight conformational ensemble Maintain activity across resistance variants
Loss of repair pathway (e.g., HRD) New collateral sensitivity Synthetic lethality drugs (e.g., PARP) Patient selection by genotype and biomarkers
Fusion proteins Aberrant dimerisation, novel interfaces Design interface or ATP-site inhibitors with fusion-aware SAR Monitor resistance clones

3) Case anchors from the lecture

  • EGFR activating mutations → pocket re-shape. First-line ATP-site inhibitors designed for the mutant pocket. Resistance gatekeeper variants prompted next-gen designs that bypass steric blocks and adjust H-bond vectors.
  • BCR-ABL kinase: early ATP-site inhibitor success followed by resistance mutations at the gatekeeper. Next-gen inhibitors restored binding by threading past the mutated residue or engaging distinct subpockets.
  • KRAS hotspot design: covalent ligands lock a mutant-created pocket, guided by the unique cysteine and switch-II groove geometry.
  • HER2 amplification: target the amplified receptor with antibodies or kinase inhibitors; genomic amplification selects the pathway and predicts benefit.
  • BRCA1/2 loss: leverage homologous recombination deficiency with PARP inhibitors as a synthetic-lethal strategy.

4) SAR workflow the lecture emphasised

  1. Variant triage: which mutations drive disease and occur at meaningful frequency.
  2. Structure hypotheses: mutant vs WT pockets, waters, protonation, dynamics.
  3. Chemotype choice: orthosteric, allosteric, covalent if a nucleophilic residue is uniquely positioned in the mutant.
  4. Iterative SAR: potency on mutant, selectivity vs WT and kinome panels, ADME flags, transporter liabilities.
  5. Resistance planning: map likely secondary mutations and pre-empt with breadth across common variants.
  6. Translation: genotype-enriched preclinical and clinical testing, with PK/PD and exposure–response anchored to the mutant target.

5) When to go covalent, allosteric, or degraders

  • Covalent: mutant-specific nucleophile near the pocket, clean WT offset, benefit from prolonged target occupancy.
  • Allosteric: resistance at ATP site or need for higher selectivity by exploiting distal pockets.
  • Targeted degraders: when occupancy does not silence function or multiple functions need removal.

6) PK/PGx considerations baked into design

  • Anticipate metabolism and transport using CYP and transporter knowledge to avoid suboptimal exposure or DDIs.
  • Plan genotype-aware trials and companion diagnostics so the molecule is tested where the genomic rationale holds.

TLDR Start with genotype and translate it into mutant structure and mechanism. Choose orthosteric, allosteric, covalent, or synthetic-lethal strategies that fit the altered pocket or pathway. Plan around resistance and PK early, then run genotype-enriched trials.


L9.3 Intro to Pharmacogenomics

Objective 1. Define pharmacogenomics and describe its role in influencing individual variability in medication safety and response

Definition

  • Pharmacogenomics studies how inherited genetic variation changes medication response, including efficacy and adverse effects.

Why response varies

  • Only about 35–45% of patients achieve remission with the first antidepressant started. Variability is common across drug classes.
  • Adverse drug reactions cause roughly 5–10% of hospital admissions, with higher burden in older adults.
  • Genetics is one factor among many that explains variability; it acts through PK and PD mechanisms.

Where genetics acts

  • PK: variants in metabolic enzymes and transporters shift absorption, clearance, exposure, and half-life. Example: CYP2C19 poor metabolisers have higher voriconazole concentrations and toxicity risk at standard doses.
  • PD: variants alter drug–target binding and downstream signaling. Example: HLA risk alleles predispose to severe immune-mediated hypersensitivity.

Clinical role

  • PGx shifts practice from one-dose-fits-all to genotype-aware selection and dosing, reducing toxicity and non-response.
  • Large multicentre evidence shows PGx-guided care lowers adverse drug reactions versus usual care.

TLDR Pharmacogenomics explains part of inter-patient variability in benefit and harm by altering PK and PD. It enables genotype-based drug choice and dosing that improves outcomes and reduces adverse events.


Objective 2. Identify common genomic mutations tested in PGx and explain their impact on metabolism or response

Core pharmacogenes and typical clinical consequences (as taught)

Gene / locus Variant pattern (functional effect) Primary role Drug examples Expected impact on patient
CYP2C19 No-function 2, 3; increased-function *17 Phase I metabolism Voriconazole, clopidogrel Poor metabolisers accumulate voriconazole and risk toxicity; increased-function can over-activate prodrugs or reduce exposure to active drugs.
CYP2D6 No-function 4, decreased 10, *41; gene duplications → UM Phase I metabolism Codeine, tramadol, many psychotropics PMs fail to form morphine from codeine; UMs over-produce morphine and risk respiratory depression.
CYP2C9 Reduced-function 2, 3 Phase I metabolism Warfarin Reduced clearance of S-warfarin, higher bleeding risk at standard doses. Dose reductions needed, combined with VKORC1.
VKORC1 −1639G>A regulatory variant Drug target (vitamin K cycle) Warfarin Increased sensitivity to warfarin. Lower starting doses.
DPYD Decreased-function alleles Catabolism of 5-FU 5-FU, capecitabine Severe toxicity with standard doses; reduce or avoid.
TPMT Low- or no-function alleles Thiopurine inactivation Azathioprine, 6-MP Myelosuppression at standard doses; use reduced dose or alternative.
UGT1A1 28, 6 Phase II glucuronidation Irinotecan Higher SN-38 exposure, neutropenia and diarrhoea risk; start lower.
SLCO1B1 (OATP1B1) c.521T>C reduced function Hepatic uptake transporter Statins (e.g., simvastatin) Higher plasma statin, increased myopathy risk; choose alternative or lower dose.
**HLA-B*57:01** Risk allele Immune presentation Abacavir High risk of hypersensitivity; screen and avoid if positive.
HLA-B15:02, HLA-B58:01 Risk alleles Immune presentation Carbamazepine; allopurinol Elevated SJS/TEN risk; allopurinol hypersensitivity risk ~hundreds-fold with HLA-B*58:01. Screen in susceptible groups.

From genotype to phenotype to dosing

  • Star-allele nomenclature defines function per allele. Genotype combinations map to metaboliser phenotype: poor, intermediate, normal, rapid, ultra-rapid. Dose and drug selection should follow phenotype to avoid toxicity or non-response.

Population notes used in triage

  • Many Australians carry at least one actionable genotype. Phenotype distributions differ by ancestry, which guides pre-test probability and interpretation.

TLDR Testing focuses on high-yield pharmacogenes: CYP2C19, CYP2D6, CYP2C9, VKORC1, DPYD, TPMT, UGT1A1, SLCO1B1, and HLA. These variants change exposure or trigger immune reactions, so match drug and dose to phenotype to prevent toxicity and treatment failure.


Objective 3. Illustrate how genetic variation alters structure and function of proteins involved in drug metabolism or action

Genotype → protein → PK/PD → clinical effect

DNA variant
   ↓
Protein change (amount, structure, localisation, kinetics)
   ↓
PK (CL, t1/2, exposure) or PD (EC50, Emax, immune risk)
   ↓
Efficacy and toxicity at a given dose

What variants do at the protein level (with exam-ready examples)

Protein class Variant type Molecular consequence PK/PD effect Lecture examples you can cite
Drug-metabolising enzymes (e.g., CYP2C19, CYP2D6, CYP2C9, UGT1A1, TPMT, DPYD) Missense in active site or heme pocket Changes pocket geometry or stability, alters Vmax and Km Slower or faster clearance, changed t1/2 and exposure CYP2D6 missense or deletion → codeine not activated; CYP2C9 2/3 → slow S-warfarin clearance
Splice/frameshift/nonsense Truncated or absent enzyme Functional knockout **CYP2C19 *2** splice defect → minimal voriconazole metabolism, high toxicity risk at standard dose
Promoter/UGT repeats Lower expression Lower enzyme abundance, decreased Vmax **UGT1A1*28** lower SN-38 glucuronidation → irinotecan neutropenia risk
Transporters (OATP1B1/SLCO1B1, ABCB1/P-gp, ABCG2/BCRP) Missense in transmembrane domain or NBD Reduced substrate translocation, misfolding or mis-trafficking Lower hepatic uptake or efflux, higher plasma Cmax/AUC SLCO1B1 c.521T>C lowers hepatic statin uptake → simvastatin myopathy risk
Drug targets (receptors, enzymes) Missense in binding pocket Affinity or efficacy shift (EC50, Emax) Different potency at same concentration GPCR variation on slides with several-fold potency shifts across common missense variants
Immune presentation (HLA) Allelic variant in peptide-binding groove Presents drug-peptide adducts, triggers T-cell response Severe idiosyncratic toxicity independent of dose HLA-B57:01 with abacavir; HLA-B15:02 with carbamazepine SJS/TEN

Make the PK/PD linkage explicit in answers

  • PK (exposure shift)

    • Enzyme LoF: (CL ), (t_{1/2} ), **AUC * → toxicity at usual dose.
    • Enzyme GoF: (CL ), (t_{1/2} ), **AUC * → non-response unless dose increased.
    • Transporter LoF: reduced uptake or efflux → **AUC * in plasma or in tissues depending on transporter site.
  • PD (sensitivity shift)

    • Target missense: **EC(_{50})** shifts right (lower potency) or left (higher potency).
    • Immune risk alleles: dose-independent hypersensitivity, so selection, not dose, is the lever.

Worked mini-cases from the lecture logic

  • CYP2C19 poor metaboliser + voriconazole Structure/function: splice LoF → minimal enzyme. PK: AUC↑, crosses toxic threshold on graph. Action: lower dose or alternative antifungal.

  • CYP2D6 poor or ultra-rapid + codeine Structure/function: absent enzyme (PM) or gene duplication (UM). PD surrogate: active metabolite (morphine) formation ↓ in PM (no effect), ↑ in UM (toxicity). Action: avoid codeine in PM and UM, pick non-2D6 opioid.

  • VKORC1 −1639G>A + CYP2C92/3 + warfarin Structure/function: VKORC1 promoter variant lowers enzyme abundance (higher drug sensitivity). CYP2C9 missense slows clearance. Combined effect: higher sensitivity and slower CL. Action: lower initial dose and tighter INR titration.

  • SLCO1B1 c.521T>C + simvastatin Structure/function: transporter activity ↓. PK: hepatic uptake ↓, plasma AUC↑, muscle exposure higher. Action: avoid high-dose simvastatin; choose alternative statin or lower dose.

Sentence templates for OSCE or SAQ

  • “This variant changes the protein’s [active site / expression / localisation], which shifts [CL, EC50, AUC], so at a standard dose the patient has [toxicity / non-response]. The fix is [alternative drug / lower dose / genotype-guided monitoring].”

TLDR Variants change protein amount, structure, localisation, or kinetics in enzymes, transporters, targets, or HLA. That shifts PK (exposure) or PD (sensitivity) and explains toxicity and non-response. Use the allele→protein→PK/PD→action chain with the lecture’s anchors: CYP2C19–voriconazole, CYP2D6–codeine, VKORC1/CYP2C9–warfarin, SLCO1B1–statins, and HLA–hypersensitivity.

WEEK 10

L10.1 Precision Medicine: Development of CF Modulators

Objective 1. Drug discovery process that led to CFTR modulators

Milestones you should name

  • 1989: CFTR gene identified. The CF Foundation funds basic mapping of functional domains and common mutations.
  • 2005–2006: Vertex and the CF Foundation run high-throughput screening (HTS) to find small-molecule potentiators and correctors.
  • Assay: NIH/3T3 cells overexpressing WT and ΔF508 CFTR. Readout is a fluorescent chloride-efflux assay. Library size ~164,000 compounds. 185 positives carried forward for medicinal chemistry.
  • Lead optimisation → Ivacaftor (VX-770).
  • Phase 3 trials show clinical benefit in selected genotypes.
  • 2012: FDA approves ivacaftor for patients ≥6 years with at least one G551D allele.
  • Post-ivacaftor: discovery and optimisation of correctors that improve folding and trafficking.
  • Trikafta (elexacaftor + tezacaftor + ivacaftor) emerges from the insight that combining two complementary correctors with a potentiator maximises surface CFTR and channel opening.
  • Real-world: PBS listings in Australia expand access; registry tracks survival gains. Cost, access, and equity remain major issues.

Why the screens hit and how SAR evolved

  • HTS found molecules that increase CFTR-mediated Cl⁻ flux. Early chemistry pruned liabilities and improved potency and lipophilicity to match a membrane protein target.
  • Later structural work with ΔF508 CFTR bound to Trikafta components showed binding in transmembrane domains (TMDs), explaining synergy between correctors and the potentiator at the surface.

Sketch the pipeline in exams

Gene + domain mapping → HTS (chloride-efflux) → hits (n=185) → med-chem → ivacaftor
→ genotype-specific Phase 3 → approval (G551D) → corrector discovery → triple combo
→ label expansion + PBS access; ongoing cost and equity challenges

TLDR Discovery moved from gene mapping to HTS in CFTR-overexpressing cells, to ivacaftor as the first potentiator, then to correctors that rescue folding. Structural data located modulator binding in TMDs, justifying the triple therapy strategy and broader genotype coverage.


Objective 2. Types of CFTR modulators and their modes of action

CFTR biology anchor

  • CFTR is an ABC transporter with TMDs and two NBDs. Channel activity requires PKA-dependent phosphorylation. Many CF mutations reduce folding, trafficking, or gating.

What each class does

Class Representative drugs Primary action Mechanistic details you can state
Potentiators Ivacaftor; deutivacaftor (next-gen, longer half-life) Increase open probability of CFTR at the cell surface Allosteric. Keeps CFTR open in a largely ATP-independent way, but requires phosphorylated CFTR to work. Improves WT and gating-mutant activity at the membrane.
Correctors, Class I Lumacaftor, tezacaftor Stabilise TMD1, aid co-translational folding and ER exit Extend ER residence to allow proper folding of WT and mutant CFTR so more reaches the surface.
Correctors, Class II Elexacaftor, vanzacaftor Enhance Class I effect by binding distinct TMD sites Complementary binding sites produce additive rescue. Some Class II show secondary potentiator-like effects in cells.

Why triple therapy works

  • Tezacaftor improves folding and trafficking.
  • Elexacaftor binds elsewhere in TMDs to add further rescue.
  • Ivacaftor then increases gating once CFTR is at the surface.
  • Cryo-EM with ΔF508 CFTR supports TMD binding for all three.

Genotype logic you will use later

  • Gating mutations (for example G551D) respond to potentiator.
  • Folding/trafficking mutations (for example ΔF508) need corrector(s) to raise surface expression plus a potentiator to maximise function.

TLDR Potentiators open CFTR channels already at the surface. Correctors raise the amount of CFTR that reaches the surface by stabilising folding in the ER. Combining two complementary correctors with a potentiator yields maximal function, especially for ΔF508 backgrounds. Structural studies place all three ligands in TMDs, consistent with their effects.

Objective 3. Know which CFTR modulators are effective for specific mutations, and why

Match the defect class to the modulator

CFTR defect (example) Molecular problem Effective modulator(s) Why it works
Class III, “gating” (e.g., G551D) Channel reaches membrane but won’t open efficiently Ivacaftor monotherapy Potentiator raises open probability of surface CFTR → restored Cl⁻ flux.
Class II, folding/trafficking (e.g., F508del) Misfolded CFTR, ER retention, little reaches surface Tezacaftor/ivacaftor or lumacaftor/ivacaftor; best response with elexacaftor/tezacaftor/ivacaftor Correctors stabilise folding and trafficking; adding a potentiator maximises activity of rescued channels. Two complementary correctors (elexacaftor + tezacaftor) target distinct TMD sites → additive rescue.
Residual function (Class IV/V) (selected splice or conductance variants) Reduced conductance or reduced transcript, but some CFTR at surface Ivacaftor ± a corrector depending on baseline function Potentiation improves opening; some variants gain further benefit when more protein is trafficked.
Minimal/no CFTR production (Class I, nonsense) No full-length protein Current modulators ineffective (consider non-modulator approaches) No substrate for potentiation or correction; requires read-through or gene/additive therapies (not the focus of modulators).

Why triple therapy dominates F508del care Cryo-EM and structure-function data in the slides show elexacaftor and tezacaftor engage distinct TMD sites, stabilising different folding bottlenecks; ivacaftor then improves gating once rescued protein reaches the surface. This three-point rescue (folding A + folding B + gating) yields the largest functional gain in F508del backgrounds.

TLDR Gating mutations respond to ivacaftor alone. Folding/trafficking mutations (F508del) need corrector(s) and do best with elexacaftor/tezacaftor/ivacaftor. Residual-function variants often benefit from ivacaftor ± a corrector. No-protein nonsense variants are not helped by modulators.


Objective 4. Benefits and common side effects of CFTR modulators (with monitoring and DDI points)

Clinical benefits highlighted in the lecture

  • Pulmonary: improved FEV₁ percent predicted; fewer pulmonary exacerbations.
  • Sweat chloride: significant reduction consistent with restored CFTR function.
  • Nutrition/weight: increased BMI/weight gain where malabsorption had been limiting.
  • Quality of life: improved patient-reported respiratory and global scores.

Common adverse effects and cautions (by regimen)

Regimen Common AEs in slides/transcript Monitoring / DDI notes
Ivacaftor Headache, GI upset, transaminase elevations; paediatric cataract signal noted historically LFTs at baseline and periodically; check for CYP3A interactions (ivacaftor is a substrate).
Lumacaftor/ivacaftor Dyspnoea/chest tightness, cough; LFT elevations Lumacaftor is a strong CYP3A inducer → reduced exposure to many drugs including some hormonal contraceptives; review meds carefully; monitor LFTs.
Tezacaftor/ivacaftor Generally better tolerated; LFT elevations possible LFTs; fewer DDIs than lumacaftor combo but maintain CYP3A awareness.
Elexacaftor/tezacaftor/ivacaftor Rash, LFT elevations, ↑CK in some; GI symptoms; headache LFTs at baseline and during therapy; manage rash (more frequent in some populations); check for CYP3A substrate/inhibitor co-meds.

Practice notes the lecturer emphasised

  • Start with baseline LFTs, repeat per label or clinic protocol, and after dose changes.
  • For lumacaftor/ivacaftor, perform a structured DDI review due to CYP3A induction, including hormonal contraceptives and azoles where relevant.
  • Counsel on expected time course of benefit (weeks) and the possibility of early respiratory symptoms with lumacaftor combinations that usually settle.

TLDR Modulators improve lung function, sweat chloride, exacerbations, weight, and QoL. Main risks: LFT elevations across regimens; dyspnoea/chest tightness with lumacaftor/ivacaftor; rash with elexacaftor/tezacaftor/ivacaftor; and CYP3A DDIs (notably with lumacaftor). Do baseline and periodic LFTs, screen for DDIs, and counsel on early symptom trajectories.

L10.2 Precision Medicine: Lung Cancer & EGFR Kinase Inhibitors

Objective 1. Describe the importance of EGFR as a critical drug target in lung cancer

What EGFR is

  • EGFR, also called HER1 or ERBB1, is a membrane receptor with an extracellular ligand-binding region, a single transmembrane helix, and an intracellular tyrosine kinase (TK) domain. The ATP site in the TK domain centers on D855. Activation requires ligand-driven dimerisation and conformational changes that open the A-loop and reposition the C-helix.

Why it matters in NSCLC

  • EGFR is an oncogenic driver in non-small-cell lung cancer and its signaling pathway is altered in a large fraction of tumours. EGFR activates multiple survival and proliferation cascades, including RAF–MAPK, PI3K, STAT3, and NF-κB, which promote growth, angiogenesis, invasion, and apoptosis evasion.

Mutations that make EGFR druggable

  • Tumour sequencing linked exon 19 deletions (Ex19del) and L858R in exon 21 to marked sensitivity to ATP-site TKIs. These are the two most common activating mutations. Early clinical series and meta-analyses showed superior progression-free survival in mutant tumours versus wild type when treated with EGFR TKIs.

Clinical take-home

  • EGFR is essential biology and a validated target. Mutation testing stratifies patients for TKIs and explains dramatic early responses seen in responders.

TLDR EGFR drives NSCLC through a kinase domain that controls key growth pathways. Activating mutations (Ex19del, L858R) reshape the ATP pocket and predict strong benefit from EGFR TKIs. Testing EGFR is central to first-line treatment selection.


Objective 2. Explore the genetic basis for chemotherapy drug resistance

Two levels of resistance

  • On-target (EGFR) resistance: new mutations in the kinase domain reduce inhibitor binding but preserve ATP binding.
  • Off-target or bypass: activation of parallel pathways, gene amplification, or histologic change that restores downstream signaling.

Canonical on-target sequence

  • Gatekeeper T790M

    • Location: gatekeeper residue controlling access to the ATP cleft.
    • Effect: bulkier methionine introduces steric hindrance, decreases binding of first- and second-generation TKIs, while ATP still fits. Clinical resistance often emerges at 1–2 years.
  • Third-generation solution: osimertinib

    • Binds covalently to C797 in the ATP site. High selectivity for mutant EGFR that carries T790M, effective first-line as well.
  • Next resistance: C797S

    • Mutation removes the covalent anchor, reducing osimertinib binding and driving the need for fourth-generation inhibitors.

Other genetic routes

  • Gene amplifications and pathway switches can bypass EGFR dependence. Histologic transformation can also occur under drug pressure. These events restore signalling despite EGFR blockade.

Resistance timeline and design logic

  • First/second-generation reversible ATP-site inhibitors succeed in Ex19del/L858R.
  • T790M emerges, so covalent third-generation drugs target C797.
  • C797S then appears, prompting new chemotypes and binding strategies.

Quick reference table

Setting Mutation pressure Binding consequence Clinical move
1st/2nd gen TKI on Ex19del/L858R Selects T790M Steric block of reversible TKIs, ATP still binds Switch to osimertinib
Osimertinib on T790M Selects C797S Loss of covalent bond Consider 4th-gen TKI trials or alternate strategies
Drug pressure on pathway Amplifications / bypass Downstream reactivation Combine or switch based on driver detected

TLDR Resistance is genetic. T790M blocks first/second-generation TKIs. Osimertinib overcomes T790M by covalently binding C797. C797S then removes the covalent handle and drives next-gen design. Bypass activation and histologic shifts are additional routes. Track mutations and adapt therapy accordingly.

Objective 3. Explain how specific EGFR kinase-domain mutations guide drug design

Where binding happens and why mutations matter

  • EGFR TKIs bind in the ATP cleft of the kinase domain. Key activation and binding features include the C-helix/K721 salt bridge, DFG-D831, HRD-D813, and the ATP site centred on D855. Amino acids that frame this pocket determine drug fit; mutations here change inhibitor binding and clinical response.

Activating mutations that create sensitivity

  • Exon 19 deletions and L858R reshape loops that flank the ATP pocket, stabilise the active conformation, lower the activation threshold, and increase TKI sensitivity. Early sequencing of complete responders located these lesions within the TK domain near the inhibitor site.

Resistance mutations that reshape design

  • T790M (gatekeeper) adds bulk at the gate to the ATP site. It sterically hinders first- and second-generation reversible TKIs while permitting ATP, driving relapse. This required new chemotypes.
  • Osimertinib (3rd gen) solved T790M by forming a covalent bond to C797 in the ATP site, regaining potency and selectivity for mutant EGFR.
  • C797S then removes the covalent anchor, leading to post-osimertinib resistance and motivating fourth-generation designs.

Design logic across generations

Generation Core design feature Pocket consequence addressed Representative drugs
1st Reversible ATP-mimetics that H-bond in the hinge (near D855 region) Leverage sensitising pocket of Ex19del/L858R Gefitinib, erlotinib
2nd Irreversible pan-ERBB; broader reactivity Partial workarounds but limited by T790M at clinical exposures Afatinib; irreversible scaffolds context
3rd Mutant-selective covalent binders targeting C797 Bypass T790M steric block, prefer mutant over WT Osimertinib; WZ4002 (tool)
4th (in development) Non-covalent or alternate-covalent strategies Overcome C797S loss of covalent anchor “4th gen” TKI concept on slides

TLDR Map the mutation to the pocket change. Ex19del/L858R open and stabilise the active cleft so reversible ATP-mimetics work. T790M blocks access, so C797-covalent design restores binding (osimertinib). C797S then removes that handle, prompting new scaffolds.


Objective 4. Match common actionable EGFR mutations with appropriate TKIs

High-frequency actionable mutations

EGFR alteration Typical effect on pocket Preferred TKI(s) per lecture Rationale
Exon 19 deletion (Ex19del) Activates and stabilises kinase; increases TKI sensitivity Gefitinib, erlotinib, afatinib, osimertinib Sensitising mutation; all generations active, 3rd gen widely used first-line.
L858R (exon 21) Activating point mutation near activation loop Gefitinib, erlotinib, afatinib, osimertinib Same sensitising logic as Ex19del; strong clinical benefit.
T790M (gatekeeper) Steric hindrance to reversible binders; ATP still fits Osimertinib Covalent C797 engagement overcomes T790M at clinical exposure.
C797S (post-osimertinib) Loss of covalent anchor Investigational 4th-gen TKIs/strategies C797S prevents covalent adduct; next-gen designs required.
Atypical mutations (ex18/ex20 subsets) Heterogeneous structural changes Variable; outcomes generally poorer vs classical Structure-based classes predict variable response; emerging focus.

Evidence anchors to cite in answers

  • Early responders had TK-domain mutations at Ex19 and L858R near the inhibitor site in the ATP cleft.
  • TKI benefit favours mutant over wild type on PFS meta-analysis.
  • Resistance plateaus at 1–2 years and arises through on-target mutations (T790M, then C797S) or bypass routes.

TLDR Ex19del/L858R → first-/second-generation TKIs work, osimertinib now common first-line. T790Mosimertinib. C797S → no approved TKI on the slides; look to 4th-gen strategies. Atypical mutations are heterogeneous and need structure-informed choices.


L10.3 Precision Medicine: mRNA Vaccines

Objective 1. How genomics enables rapid mRNA-vaccine design and why this platform has advantages over traditional vaccines

From sequence to vial, fast

Genomics collapses the front end of vaccine R&D into a digital design step.

  1. Pathogen genome posted → antigen chosen in silico (e.g., SARS-CoV-2 spike).
  2. mRNA designed in hours (codon-optimised, stabilised, capped, modified nucleosides).
  3. In-vitro transcription produces the mRNA.
  4. Formulate in lipid nanoparticles (LNPs) to protect RNA and deliver it to the cytoplasm.
  5. Fill–finish and QC → first-in-human. This is a platform: the manufacturing process stays the same while you swap the sequence for new targets or variants.

COVID-19 timeline as proof

  • 10 Jan 2020: SARS-CoV-2 genome posted online.
  • Within 2 days: spike antigen agreed.
  • ~1 hour: vaccine mRNA sequence designed.
  • ~45 days: clinical-grade RNA manufactured, QC’d, filled, and shipped for trials.
  • Outcome: the same sequence reached approval (Spikevax).

Why genomics keeps it fast after launch

  • Global genomic surveillance (e.g., GISAID) tracks spike mutations; boosters are updated by editing the mRNA code while keeping the same platform.

Mechanism recap

  • mRNA is a transient instruction read by ribosomes in the cytoplasm; it does not enter the nucleus and degrades after use. The encoded antigen is expressed and presented to the immune system.

Platform advantages vs traditional vaccines

Feature mRNA vaccines Traditional platforms (inactivated, live-attenuated, protein subunit)
Design speed Digital antigen selection from genome; hours–days to design, weeks to clinical lot Requires pathogen growth, protein purification, or attenuation; months–years
Manufacturing Single IVT + LNP process reused across targets; scale by sequence swap Process differs per pathogen; bespoke cell culture and purification
Update agility Rapid code edits follow genomic surveillance (variant boosters) Reformulation and new production cycles
No pathogen No handling of live virus during manufacture Live/attenuated lines or viral propagation often required
Antigen fidelity Precise sequence control (codons, stabilising mutations) Biologic variability in antigen prep
Cytoplasmic expression No nuclear entry; transient expression Variable depending on platform
Platform extensibility Same chassis for infectious disease and oncology neoantigens Usually bespoke per disease area

Schematic you can sketch quickly

Genome posted → Antigen picked (in silico) → mRNA designed (1–2 h)
→ IVT + cap/modified nucleosides → LNP formulation → QC/fill-finish → Trials
                 ↑ same chassis, swap the sequence for variants

TLDR Genomics turns vaccine design into a software-like step: sequence in, sequence out. mRNA platforms keep process constant while swapping code, enabling hour-scale design, week-scale manufacture, and fast variant updates, without growing virus.


Objective 2. How mRNA technologies enable precision medicine (variant-specific vaccines and personalised cancer immunotherapies)

A) Variant-specific infectious-disease vaccines

Precision workflow (what changes and what stays constant)

  • Genomics in → sequence out: global surveillance (e.g., GISAID) flags spike mutations; teams edit the mRNA coding sequence accordingly while keeping the same IVT + LNP platform.
  • Manufacturing/CMC: cap chemistry, modified nucleosides, and LNP composition are conserved, so design-to-batch timelines compress; only the antigen sequence changes.

Why this is precision medicine

  • Target matching: boosters can match circulating variants (monovalent/bivalent designs) to maintain neutralisation breadth.
  • Population tailoring: same platform supports age- or risk-specific dose levels and schedules without re-engineering biology.

Schematic you can sketch

Surveillance (global genomes) → Pick variant antigen in silico
→ Edit mRNA code (codon/stability) → IVT + LNP (unchanged process)
→ QC/fill → Variant-matched booster rollout

B) Personalised cancer mRNA vaccines (neoantigen vaccines)

Patient-specific pipeline the lecture outlined

  1. Tumour & normal sequencing (WES/RNA-seq).
  2. Neoantigen prediction: call somatic mutations, infer HLA type, predict high-affinity binders; prioritise clonal, expressed mutations.
  3. mRNA construct: encode a poly-epitope string (CD8 and CD4 targets), add UTR/cap/modified nucleosides.
  4. IVT + LNP manufacture (platform unchanged), then dosing with or without a checkpoint inhibitor.
  5. Readouts: expansion of neoantigen-specific T cells, tumour regression signals.

Why this is precision medicine

  • Individual tumour genetics → bespoke immunogen (mutation-unique antigens reduce off-tumour risk).
  • Rapid, repeatable chassis enables patient-by-patient manufacturing with identical chemistry/CMC, only the sequence differs.

C) Side-by-side: variant boosters vs personalised neoantigen vaccines

Aspect Variant-specific infectious vaccine Personalised cancer vaccine
Input data Population viral genomes (surveillance) Individual tumour + normal genomes (WES/RNA-seq)
Antigen choice Variant spike (or other pathogen protein) Patient’s neoantigens (somatic mutations, HLA-matched)
Construct Single or bivalent mRNA Poly-epitope mRNA (CD8/CD4)
Platform Same IVT + LNP Same IVT + LNP
Goal Restore neutralisation against circulating variants Elicit tumour-specific T cells, often with checkpoint inhibitors
Precision lever Match to current variant Match to patient’s tumour mutations

TLDR mRNA is a platform for precision medicine. For infectious disease, genomic surveillance drives variant-matched boosters by swapping code, not process. For oncology, a patient’s tumour genome drives neoantigen-encoded, poly-epitope mRNA to prime personalised T-cell responses, frequently paired with checkpoint blockade. Both reuse IVT + LNP with only the sequence changing.

WEEK 11

L11.1 Clinical Implementation & Application of Pharmacogenomics

Explain how genomics-based diagnostics are used to guide therapeutic decision-making in clinical settings.

The clinical workflow you must name

  1. Indication: pick a gene–drug pair where results change therapy. Examples: CYP2C19 for clopidogrel or voriconazole, HLA-B*57:01 for abacavir, DPYD for 5-FU/capecitabine, TPMT/NUDT15 for thiopurines.
  2. Order test from a NATA-accredited lab; obtain informed consent.
  3. Collect sample: blood (higher DNA yield) or saliva/buccal swab (convenient); label and transport per pre-analytical requirements.
  4. Lab analysis: DNA extraction → genotyping/sequencing → variant calling → report with genotype, mapped phenotype and clinical advice.
  5. Therapeutic action using guideline mapping from genotype→phenotype→dose/drug choice.
Indication → Consent/Order (NATA) → Sample (blood/saliva/buccal) 
→ Genotype → Phenotype → Action (avoid/swap/adjust/monitor)

When to test and how it changes care

  • Reactive testing: patient is on, or about to start, a specific medicine; use a single-gene or panel as appropriate.
  • Pre-emptive testing: store panel results in the record for future prescribing to avoid delays and repeated tests.

Use Australian indications to triage

  • RCPA indications classify gene–drug pairs as Recommended, Consider, or Available (no consensus). Several tests attract Medicare rebates (e.g., HLA-B57:01, HLA-B15:02, HLA-A*31:01, DPYD, TPMT).

How results change decisions (anchor examples)

  • Avoid drug: HLA-B57:01 → avoid abacavir. HLA-B15:02 → avoid carbamazepine in at-risk groups.
  • Swap drug: CYP2C19 poor/intermediate → avoid clopidogrel; use ticagrelor/prasugrel.
  • Adjust dose: TPMT/NUDT15 reduced activity → lower thiopurine dose; DPYD decreased function → reduce/avoid fluoropyrimidines.
  • Optimise exposure: CYP2C19 phenotype predicts voriconazole levels; adjust dose or choose alternative.

Context always matters

  • Integrate age, renal/hepatic function, comorbidities, and DDIs. Strong inhibitors/inducers can phenoconvert the observed response and override genotype.

TLDR Follow a fixed pathway: indication → accredited test → sample → genotype→phenotype → action. Use RCPA indications to decide who to test, then apply results to avoid, swap, or adjust therapy, while accounting for DDIs and patient factors.


Demonstrate how to use pharmacogenomics resources (e.g., CPIC, ClinPGx) to support clinical decision-making for treatment selection and dose optimisation.

Five-step playbook (apply this in exams and OSCEs)

  1. Confirm gene–drug pair and phenotype on the report (e.g., CYP2C19 IM).

  2. Open guideline: CPIC or DPWG for dosing and alternatives; ClinPGx for evidence strength and summaries.

  3. Select action:

    • Avoid/substitute when guidelines say “Do not use” for the phenotype.
    • Dose change when a starting dose reduction or ceiling is recommended.
    • Monitoring if therapeutic drug monitoring or safety labs are advised.
  4. Check context: DDIs, renal/hepatic function, comorbidities. Guard against phenoconversion.

  5. Document: genotype, phenotype, source guideline, chosen action, and follow-up plan. Store results accessibly for future use.

Worked mini-workflows from the lecture

  • Clopidogrel after PCI

    • Report: CYP2C19 PM/IM.
    • CPIC/DPWG: Avoid clopidogrel; choose ticagrelor/prasugrel.
    • Action: Substitute, document rationale; no need to “dose-up” a prodrug that will not activate.
  • Thiopurines in IBD

    • Report: TPMT or NUDT15 decreased function.
    • CPIC/DPWG: Start at reduced dose or choose alternative based on phenotype severity.
    • Action: Lower dose, schedule FBC monitoring for myelosuppression.
  • 5-FU/Capecitabine

    • Report: DPYD decreased function.
    • CPIC/DPWG: Reduce or avoid depending on activity score.
    • Action: Start reduced, consider alternative regimen; monitor for toxicity.
  • Abacavir

    • Report: **HLA-B*57:01 positive**.
    • CPIC/DPWG: Contraindicated.
    • Action: Select alternative NRTI; record allergy/intolerance.

Quick reference: where each resource fits

Task Use this Why
Dosing and alternatives CPIC, DPWG Actionable, phenotype-based recommendations
Evidence strength, summaries, visuals ClinPGx Curated evidence levels; mechanism diagrams
Local “should I test?” triage RCPA indications Australian categories: Recommended / Consider / Available

Practical notes for implementation

  • Turnaround: single-gene fast; panel supports pre-emptive storage; WES/WGS is rare in routine care. Plan accordingly.
  • Cost/rebate: some tests are Medicare-rebated; panel pricing often competitive with serial single-gene orders.
  • Documentation: include guideline name/version in your note to support prescribing and future reuse.

TLDR Start from the reported phenotype, open CPIC/DPWG for the action, check ClinPGx for evidence strength, adjust for DDIs and organ function, then document the plan. Use RCPA indications to decide who to test and panel testing to future-proof prescribing.


Compare and contrast different pharmacogenomic testing approaches and their implications for patient care.

What you are choosing between

Approach Typical methods Scope Turnaround Strengths Limitations Best use
Single-gene targeted test Targeted genotyping or small amplicon NGS One drug–gene (e.g., CYP2C19, **HLA-B*57:01**) Fast Quick, inexpensive, simple consent/report Narrow scope; can miss rare alleles or CYP2D6 CNVs; repeats testing later Reactive prescribing when one decision is urgent
Focused PGx panel Array or amplicon NGS with star-allele translation; CNV calling for CYP2D6 10–25+ pharmacogenes (CYPs, UGT1A1, SLCO1B1, DPYD, TPMT/NUDT15, HLA) Moderate Pre-emptive value across many drugs; fewer repeat tests; better allele coverage Interpretation complexity; incidental non-actionable variants; still misses some rare/hybrid events Pre-emptive storage in EHR; multi-drug clinics (cardiology, oncology, psych)
WES/WGS Exome or whole-genome sequencing Broadest Slowest Future-proof, finds rare/novel variants and non-PGx findings if consented Cost; variable coverage of PGx loci, CYP2D6 structural complexity; star-allele translation non-trivial; secondary findings and data-governance issues Research, complex cases, systems building; seldom first-line in routine PGx

Key lecture points to state

  • Pre-emptive panels reduce downstream delays and duplicate tests; store phenotypes in the record for reuse.
  • Reactive single-gene tests remain appropriate when one imminent prescribing decision hinges on one gene.
  • CYP2D6 requires CNV-aware methods for accurate phenotype.
  • Consent, storage, and local reimbursement differ by test type; some single-gene tests are Medicare-rebated.
  • Whatever the assay, you must still account for phenoconversion from strong inhibitors/inducers, organ dysfunction, and clinical context.

TLDR Match the clinical question to the assay. Use single-gene for urgent, one-off decisions, PGx panels for pre-emptive multi-drug value with CYP2D6 CNV support, and WES/WGS rarely, when broad discovery is required. Regardless of assay, adjust for DDIs, organ function, and phenoconversion.


Assess the clinical utility of pharmacogenomic information in a case-based scenario involving drug selection or dose adjustment.

Case A. PCI on dual antiplatelet therapy

Indication Post-stent patient planned for clopidogrel. Order CYP2C19.

Result Genotype: 2/2 → Poor metaboliser (PM). Report maps to phenotype and provides action statements.

Guideline lookup Open CPIC/DPWG; avoid clopidogrel in PM/IM; choose ticagrelor or prasugrel. Optionally confirm in ClinPGx for evidence strength.

Context checks

  • Contraindications to alternatives, bleeding risk.
  • DDIs that might phenoconvert.
  • Renal/hepatic function.

Plan note (exam-ready wording) “CYP2C19 2/2, phenotype poor metaboliser. Per CPIC/DPWG, clopidogrel is unlikely to be effective. Substitute ticagrelor. Document genotype, phenotype, source guideline, and counsel on bleeding and dyspnoea risk.”

Utility Prevents treatment failure and stent thrombosis risk; demonstrates actionable change in drug selection.


Case B. Initiating thiopurine for IBD

Indication Starting azathioprine. Order TPMT/NUDT15 (panel preferred if future PGx use anticipated).

Result TPMT 1/3A (IM) or NUDT15 decreased function.

Guideline lookup CPIC/DPWG: start at reduced dose or consider alternative depending on activity score; schedule serial FBC to monitor for myelosuppression.

Context checks Concomitant myelosuppressants, infection risk, liver tests.

Utility Mitigates severe myelosuppression and hospitalisation; shows how genotype changes dose and monitoring.


Case C. Starting 5-FU/capecitabine

Indication Adjuvant chemotherapy. Order DPYD before first cycle.

Result Activity score indicates decreased function.

Guideline lookup Reduce or avoid fluoropyrimidines based on activity score; consider alternative regimen; institute enhanced toxicity monitoring.

Utility Prevents life-threatening toxicity; high yield and often reimbursed.


What to document every time

  • Genotype → phenotype, guideline version, decision (avoid/swap/adjust), monitoring plan, and where the result is stored for future prescribing. Build in an alert for phenoconversion risks.

TLDR Clinical utility is proven when genotype changes management and improves outcomes. Use a fixed playbook: order wisely, map to phenotype, apply CPIC/DPWG, check context and phenoconversion, and document so results are reusable. Panels maximise future value; single-gene tests solve urgent questions now.


L11.2 Sample to Prescription

Understand the process of pharmacogenomic testing from sample to prescription.

End-to-end clinical pathway

  1. Indication and consent Identify a drug–gene question. Obtain consent. Choose an accredited lab.

  2. Collection and transport Buccal swab at home or clinic. Use barcoded kit and biohazard bag. Mail to lab. Swab design preserves DNA integrity in transit.

  3. Accessioning and pre-analytics Receive, label, and check for damage or low yield.

  4. DNA extraction and QC Lyse cells, bind DNA to magnetic beads, wash, elute. Quantify and assess purity by spectrophotometer to confirm suitability for downstream assays.

  5. Target amplification (PCR) Amplify PGx loci to create ample template for genotyping. Primer pairs flank variant sites.

  6. Genotyping Use TaqMan OpenArray or Agena MassARRAY to call alleles at predefined variants. Labs may also deploy NGS panels.

  7. Bioinformatics → diplotype → phenotype Call variants and CNVs. Map to star-alleles using PharmVar rules. Translate diplotype to phenotype with guideline logic (e.g., CYP2D6 1/4 → IM; CYP2C19 2/2 → PM).

  8. Report and delivery Secure report to clinician or pharmacist with action categories (e.g., traffic-light). Store results for reuse.

  9. Therapeutic action and counselling Apply guideline-based action: avoid, switch, or adjust dose. Integrate age, organ function, and DDIs. Watch for phenoconversion from inhibitors or inducers.

Indication → Swab/Ship → Accession → Extract + QC → PCR → Genotype
→ Bioinformatics (PharmVar) → Diplotype → Phenotype → Report → Action

TLDR Follow a fixed chain: collect → extract/QC → PCR → genotype → star-allele → phenotype → report → prescribe. Results guide avoid/switch/dose actions, but you still check comorbidities and DDIs before writing the script.


Learn about DNA extraction, PCR, and genotyping methods.

DNA extraction and QC

  • Lysis with detergent and protease releases nucleic acids.
  • Magnetic-bead purification isolates DNA at scale.
  • QC by spectrophotometer confirms concentration and purity for reliable amplification.

PCR fundamentals

  • Purpose: enrich specific loci carrying pharmacogenetic variants.
  • Cycle: denaturation → annealing (primers bind) → extension (polymerase synthesises).
  • qPCR adds real-time detection; labs miniaturise into OpenArray plates for throughput.

Genotyping methods used in PGx

1) TaqMan OpenArray (real-time PCR with allele-specific probes)
  • Primers amplify the target.
  • Allele probes each carry a reporter dye and a quencher.
  • When polymerase cleaves a bound probe, the reporter separates from the quencher and fluoresces.
  • Color readout identifies genotype: one dye = homozygote, both dyes = heterozygote.
  • Strengths: fast, scalable, well-validated assays. Limits: focused to known variants.
2) Agena MassARRAY (MALDI-TOF with single-base extension)
  • PCR first, then a single-base extension primer adds one terminating nucleotide at the SNP.
  • MALDI-TOF measures mass-to-charge; each allele gives a distinct mass peak.
  • Peaks call AA, AB, or BB genotypes at high precision.
  • Strengths: accurate multiplexing. Limits: targets predefined loci.
3) NGS (emerging clinical standard)
  • Broader coverage across PGx genes, captures rare or novel variants and supports future re-analysis.
  • Trade-offs include turnaround and star-allele translation complexity in some loci.

Method comparison (what to say in exams)

Step TaqMan OpenArray Agena MassARRAY NGS PGx panels
Chemistry Allele-specific probes with fluorescent reporters Single-base extension + MALDI-TOF mass readout Targeted capture or amplicon sequencing
Output Fluorescence cluster → genotype Mass peaks → genotype VCFs → star-alleles → phenotype
Scope Known variants on array Known variants with multiplexing Broad, includes rare/novel variants
Strength Speed, throughput, mature assays Precision, multiplex, low per-variant cost Comprehensive, future-proof data
Caveat Limited to panel content Limited to panel content Longer TAT, complex interpretation in some loci

TLDR Extract with beads and QC first. PCR enriches targets. Labs then call alleles by fluorescent probes (TaqMan) or by mass (MassARRAY). NGS expands coverage and future-proofs data. All paths flow into star-alleles → phenotype → action.


Comprehend the translation of genotype into clinical phenotype.

The mapping chain you must name

Variants (incl. CNVs) → Star-alleles (PharmVar rules) → Diplotype → Activity score (if applicable) → Phenotype → Action

Core principles

  • Star-alleles encode function per gene. Combine two star-alleles to get a diplotype.
  • Some genes use an activity score (AS) to convert diplotype into phenotype.
  • Copy-number matters (e.g., CYP2D6 gene duplications or deletions).
  • Always account for phenoconversion (strong inhibitors/inducers can override genotype).

Exam table: genotype → phenotype → action

Gene Diplotype examples → AS Phenotype Typical action from report
CYP2D6 1/1 (AS 2), 1/4 (AS 1), 4/4 (AS 0), 1xN/1 (AS >2) NM, IM, PM, UM Codeine/tramadol: avoid in PM/UM; pick non-2D6 opioid. Psychotropics: adjust per guideline.
CYP2C19 1/1, 1/2, 2/2, 1/17, 17/17 NM, IM, PM, RM/UM Clopidogrel: avoid in IM/PM; voriconazole: dose or switch by phenotype.
DPYD Activity score by variant set Normal, Intermediate, Poor 5-FU/capecitabine: reduce or avoid by AS.
TPMT / NUDT15 Decreased-function diplotypes IM, PM Thiopurines: start low or use alternative; monitor FBC.
UGT1A1 1/28, 28/28, *6 etc. Reduced function Irinotecan: reduce dose in high-risk genotypes.
SLCO1B1 c.521T>C diplotypes Reduced function Statins (simvastatin): avoid high dose or switch.
HLA-B 57:01, 15:02 Positive Abacavir: contraindicated; Carbamazepine: avoid in at-risk groups.

Read and act like this (report language)

  • CYP2C19 2/2 → PM. Use ticagrelor/prasugrel rather than clopidogrel.”
  • TPMT IM. Start reduced thiopurine dose. Monitor FBC.”
  • DPYD decreased function. Reduce 5-FU or choose alternative. Intensify toxicity monitoring.”

TLDR Convert star-alleles to a diplotype, then to activity/phenotype, then to a clear action. Do not forget CNVs (CYP2D6) or phenoconversion from DDIs. Quote the phenotype and the concrete change to the script.


Recognise the real-world application of PGx in personalised medicine.

High-yield clinical use-cases from the lecture

Scenario What you order Typical result Action you write Why it matters
Post-PCI dual antiplatelet therapy CYP2C19 IM/PM Avoid clopidogrel → ticagrelor/prasugrel Prevents stent thrombosis from non-response.
New start codeine for pain CYP2D6 (CNV-aware) PM or UM Avoid codeine; pick non-2D6 opioid Avoids no analgesia in PM and toxicity in UM.
Initiating thiopurine in IBD TPMT/NUDT15 IM or PM Reduce dose or alternative; monitor FBC Prevents severe myelosuppression.
Planning fluoropyrimidine chemotherapy DPYD Decreased function Reduce/avoid per activity score; monitor toxicity Reduces life-threatening toxicity.
Statin myopathy risk SLCO1B1 Reduced function Avoid high-dose simvastatin; choose alternative Lowers myopathy risk and improves adherence.
Abacavir initiation **HLA-B*57:01** Positive Do not use abacavir Prevents hypersensitivity.

Pre-emptive vs reactive in practice

  • Reactive: order a single-gene test when one drug decision is urgent.
  • Pre-emptive panel: store phenotypes in the record to automate future alerts and avoid repeat testing.
  • Build the note template once; reuse across cases.

One-minute prescribing note (template)

  • Result: “CYP2C19 2/2 → PM.”
  • Guideline: “Per CPIC/DPWG, avoid clopidogrel.”
  • Plan: “Use ticagrelor. Review DDIs and bleeding risk.”
  • Follow-up: “Document phenotype in EHR, enable alerts.”

TLDR PGx changes drug choice and dose in real clinics. Use reactive single-gene tests for urgent calls and pre-emptive panels for long-term value. Write a short note that states phenotype, cites a guideline, records the action, and sets follow-up.


L11.3 HLA Testing & Hypersensitivity Reactions

Explain the role of HLA genes in immune function and their association with drug-induced hypersensitivity reactions.

HLA in immunity

  • HLA genes sit in the MHC on chromosome 6. Class I loci (HLA-A, -B, -C) present intracellular peptides to CD8⁺ T cells. Class II present extracellular peptides to CD4⁺ T cells.
  • Extreme polymorphism at HLA changes the peptide-binding groove, shaping which self or pathogen peptides are displayed to T cells.

Why HLA matters for adverse drug reactions

  • Some drugs or metabolites form drug–HLA–peptide complexes that are seen as “non-self,” triggering clonal T-cell activation and type IV hypersensitivity. Clinical outcomes include SCARs: DRESS, SJS, and TEN.

High-yield drug–allele associations from the lecture

  • **HLA-B*57:01 → abacavir hypersensitivity. Testing made routine worldwide and cut incidence from about 7–12% to near zero in some regions. Abacavir is contraindicated if positive.**
  • **HLA-B*58:01 → allopurinol**-induced SCARs. High carriage in East Asians; many jurisdictions mandate testing before first-line urate-lowering therapy.
  • HLA-B15:02 and HLA-A31:01 → carbamazepine-induced SJS/TEN/DRESS. B15:02 is enriched in Southeast Asian populations; A31:01 higher in Europeans and Japanese.

Population impact

  • Allele frequencies vary by ancestry, so pre-test probability and the cost–benefit of screening are population-specific. The lecture highlighted higher B58:01 in East Asia and meaningful B57:01 carriage in parts of South Asia and Europe.

Core table to memorise

Drug HLA allele Risked syndrome(s) Action if positive
Abacavir HLA-B*57:01 Multisystem hypersensitivity Contraindicated; use non-abacavir regimen
Allopurinol HLA-B*58:01 SJS/TEN, DRESS Avoid; choose alternative urate-lowering therapy
Carbamazepine / Oxcarbazepine HLA-B15:02; HLA-A31:01 SJS/TEN; SJS/TEN/DRESS Avoid; select non-aromatic anticonvulsant where possible

TLDR HLA molecules present peptides to T cells. Certain alleles bind drug-derived antigens and trigger T-cell–mediated SCARs. The highest-yield pairs in practice are B57:01–abacavir, B58:01–allopurinol, and B15:02/A31:01–carbamazepine. Pre-treatment HLA genotyping prevents life-threatening reactions and is prioritised based on allele frequency in the local population.


Describe the mechanisms by which drug–HLA interactions lead to severe cutaneous adverse reactions (SCARs) such as SJS, TEN, and DRESS.

Mechanistic models highlighted

  • Hapten/pro-hapten: a drug or reactive metabolite covalently modifies a self protein, creating a neo-antigen that loads into the HLA groove.
  • p-i (pharmacologic interaction): a drug binds non-covalently to HLA or directly to the T-cell receptor, altering recognition thresholds without antigen processing.

Immunopathology sequence

  1. Drug–HLA–peptide complex is displayed on APCs.
  2. CD8⁺ and CD4⁺ T cells recognise the complex and clonally expand.
  3. Cytokines escalate inflammation.
  4. Cytotoxic T cells and mediators induce keratinocyte apoptosis and tissue injury.
  5. Clinical syndromes emerge on characteristic timelines.

Syndrome profiles to quote

Syndrome Onset after exposure Hallmark features Mortality Common triggers in slides
DRESS 2–6 weeks Fever, facial oedema, diffuse rash >50% BSA, lymphadenopathy, hepatitis, nephritis, pneumonitis ~1–10% Carbamazepine, lamotrigine, phenytoin, allopurinol, sulfonamides, some antivirals/antibiotics
SJS/TEN 4–28 days Prodrome then blistering, mucosal erosions, skin detachment; organ involvement ~10–40% (TEN higher) Antiepileptics, allopurinol, sulfonamides, NSAIDs, abacavir

Why pre-emptive HLA testing works

  • Removing exposure in genetically susceptible patients stops the initiating step. Programs that introduced HLA-B57:01 testing virtually eliminated abacavir hypersensitivity in several cohorts. Similar reductions were noted for B58:01–allopurinol and B15:02/A31:01–carbamazepine.

One-line flow you can sketch in exams

Drug (or metabolite)
   ↓ binds covalently (hapten) or non-covalently (p-i)
HLA groove / TCR
   ↓
T-cell activation (CD8/CD4) → cytokines → cytotoxicity
   ↓
Keratinocyte apoptosis → DRESS / SJS / TEN

TLDR SCARs are T-cell–mediated. Drugs trigger them by covalently creating neo-antigens or by non-covalently altering HLA/TCR recognition. Activated T cells and cytokines drive keratinocyte apoptosis, producing DRESS at 2–6 weeks and SJS/TEN at 4–28 days with high mortality. Pre-treatment HLA testing interrupts this cascade.


Interpret HLA genotyping results (e.g., HLA-B57:01, HLA-B58:01, HLA-B15:02, HLA-A31:01) to guide safe prescribing decisions.

How to read the report

  • Locus and allele: e.g., **HLA-B*57:01** present.
  • Zygosity: most calls are heterozygous; action does not change with zygosity for these drug–allele pairs.
  • Assay resolution: high-resolution typing preferred; if imputed/low-res, confirm when action has major consequences.
  • Action statement: usually categorical (contraindicated/avoid/use alternative).

Actionable pairs and what to write

Result Drug(s) affected Risked syndrome(s) Action you take Note to document
**HLA-B*57:01 positive** Abacavir Multisystem hypersensitivity Contraindicated “Avoid abacavir. Record as genomic contraindication.”
**HLA-B*58:01 positive** Allopurinol SJS/TEN, DRESS Avoid “Use febuxostat or non-allopurinol strategy. Counsel on SCAR symptoms.”
**HLA-B*15:02 positive** Carbamazepine, oxcarbazepine SJS/TEN Avoid “Select non-aromatic anticonvulsant (e.g., levetiracetam, valproate if appropriate).”
**HLA-A*31:01 positive** Carbamazepine SJS/TEN/DRESS Avoid / consider alternative “Risk extends beyond SJS/TEN, include DRESS. Choose alternative if possible.”

Practical cautions

  • Negative does not remove all risk, but it lowers it to background for that allele-mediated mechanism.
  • Reactions can occur with related drugs (e.g., oxcarbazepine with B*15:02).
  • Record HLA results in the allergy/adverse reaction section and problem list so they fire decision-support alerts.

TLDR Read the allele, ignore zygosity for these pairs, act categorically: B57:01→no abacavir, B58:01→no allopurinol, B15:02/A31:01→avoid carbamazepine. Store results where prescribing systems can see them.


Evaluate clinical guidelines (CPIC, RCPA, ClinPGx) to determine appropriate therapeutic alternatives for patients with high-risk HLA alleles.

Who does what

Task Best source How you use it
Phenotype-to-action dosing/avoidance CPIC Clear, graded recommendations (e.g., “Strong: Do not use abacavir if B*57:01 positive”).
Local testing indications and funding RCPA Decides who you test pre-treatment in Australia; lists “Recommended/Consider/Available.”
Evidence snapshots and visuals ClinPGx Summaries of mechanism, effect sizes, allele frequencies, and quick action tables.

Worked substitutions (lecture anchors)

  • **B*57:01+ patient needing NRTI: Avoid abacavir; use tenofovir-based** backbones as appropriate to regimen.
  • **B*58:01+ gout: Avoid allopurinol; consider febuxostat** or non-urate-lowering approaches if contraindicated.
  • **B*15:02+ epilepsy (East/Southeast Asian ancestry): Avoid carbamazepine/oxcarbazepine; choose levetiracetam, valproate** (if clinically suitable), or other non-aromatic options.
  • **A*31:01+**: prefer non-CBZ regimens; if unavoidable, document risk discussion and plan close monitoring.

Documentation template

  • “**HLA-B*58:01 positive. Per CPIC and RCPA, avoid allopurinol due to high SCAR risk. Plan febuxostat**. Education given on SCAR symptoms. Result filed to fire future alerts.”

TLDR Open CPIC for the action, RCPA to justify testing and align with local practice, and ClinPGx for quick evidence context. Default to substitution, not “test dosing,” for high-risk alleles.


Assess the impact of population-specific HLA allele frequencies on the implementation of pharmacogenomic testing strategies.

Why frequency matters

  • Pre-test probability and number-needed-to-genotype depend on allele prevalence. Screening yield rises with higher carriage.

Key patterns highlighted

  • **HLA-B*58:01: higher in East Asians**; routine pre-allopurinol testing yields large absolute risk reduction.
  • **HLA-B*15:02: concentrated in Southeast/East Asian** populations; targeted testing before carbamazepine is high value.
  • **HLA-A*31:01: notable in Europeans and Japanese**; raises broad CBZ hypersensitivity risk.
  • **HLA-B*57:01**: present in many populations (Europe, South Asia); universal pre-abacavir testing is justified.

Implementation models

  • Universal testing when allele is common and the reaction is severe and preventable (e.g., **B*57:01 → abacavir**).
  • Targeted testing by ancestry when carriage is clustered (e.g., **B*15:02 → carbamazepine** in Southeast/East Asians).
  • Always confirm self-reported ancestry is incomplete; when uncertain, test rather than assume low risk.

Clinic checklist

  • Map local demography to screening policy.
  • Build EHR order sets that suggest HLA tests based on medication and recorded ancestry.
  • Track outcomes to refine policy.

TLDR Match screening breadth to allele frequency and reaction severity. Use universal pre-abacavir testing and targeted pre-CBZ/allopurinol testing in high-frequency groups. When ancestry is unclear, test.

WEEK 12

L12.1 CYP2C19 & Clopidogrel

Objective 1. Define the role of clopidogrel and its clinical uses

What it is

  • Oral antiplatelet prodrug. Irreversibly inhibits platelet P2Y12 (ADP) receptor once activated. Prevents ADP-mediated activation and aggregation.

Where you use it

  • Acute coronary syndrome, post-PCI stent protection (dual antiplatelet therapy with aspirin), secondary prevention after MI or ischemic stroke, peripheral artery disease.

Real-world

  • Most PCI patients receive a P2Y12 inhibitor. Clopidogrel remains common due to cost and tolerability, despite greater efficacy of prasugrel or ticagrelor in selected settings.

DAPT snapshot (post-PCI)

  • Loading: clopidogrel 600 mg then 75 mg daily; alternatives: ticagrelor 180 mg then 90 mg twice daily, prasugrel 60 mg then 10 mg daily. Choice depends on bleeding risk, cost, comorbidity, and PGx.

TLDR Clopidogrel is a P2Y12 inhibitor used for ACS, post-stent care, and secondary prevention. It is a prodrug, so clinical effect depends on metabolic activation. It remains widely prescribed in PCI pathways alongside aspirin.


Objective 2. Explain clopidogrel metabolism and the role of CYP2C19

Pathway overview

Clopidogrel (prodrug) → hepatic CYPs → (active thiol metabolite)
                           ↑
                        CYP2C19 is the key rate-limiting step
Active metabolite → covalent block of platelet P2Y12 → ↓ aggregation
  • CYP2C19 converts clopidogrel to its active thiol. Other CYPs contribute, but CYP2C19 is the major determinant of exposure.

CYP2C19 alleles you must name

  • 1 normal function; 2 and 3 loss-of-function; 17 gain-of-function. Population frequencies vary by ancestry.

PK/PD evidence

  • Healthy-volunteer study: IM/PM phenotypes produce about 30% less active metabolite across doses. Associated reduction in antiplatelet effect at 4 hours.

TLDR Clopidogrel needs CYP2C19 to form its active metabolite. 2/3 reduce activation, *17 increases it. Loss-of-function lowers active-metabolite exposure and blunts platelet inhibition.


Objective 4. Compare poor, intermediate, normal, rapid, and ultrarapid phenotypes for clopidogrel

Phenotype Common diplotypes Active metabolite exposure Platelet inhibition Clinical outcomes on clopidogrel Recommended action (ACS/PCI) Notes
Poor metaboliser (PM) 2/2, 2/3, 3/3 Markedly ↓ Blunted Highest on-treatment platelet reactivity, higher MACE and stent thrombosis Avoid clopidogrel. Use ticagrelor or prasugrel if not contraindicated FDA boxed warning aligns; “dose-up” clopidogrel is not recommended
Intermediate metaboliser (IM) 1/2, 1/3, 2/17 Reduced Elevated MACE risk vs NM Prefer ticagrelor/prasugrel over clopidogrel If clopidogrel must be used, document risk and monitor closely
Normal metaboliser (NM) 1/1 Baseline Expected Reference risk Clopidogrel acceptable at standard dosing Choose alternative P2Y12 if other clinical factors favour it
Rapid (RM) 1/17 Slightly ↑ Slightly ↑ Similar or slightly better efficacy vs NM without clear bleeding excess Clopidogrel acceptable Consider clinical context rather than genotype alone
Ultrarapid (UM) 17/17 No consistent harm signal in lecture material Clopidogrel acceptable Monitor for bleeding with concomitant anticoagulants or high bleeding risk

Key comparisons to state in answers

  • PM vs IM: both have reduced activation; PM carries the largest risk and requires switching. IM also prefer switch, especially after PCI.
  • UM/RM vs NM: similar prescribing as NM; no guideline-driven dose increase or avoidance solely for UM/RM.
  • Dose escalation of clopidogrel does not reliably overcome LOF genotypes after PCI and is not the recommended workaround.
  • Always integrate DDIs and clinical factors: strong CYP2C19 inhibitors, prior stroke/TIA (prasugrel contraindicated), bleeding risk, cost, adherence, dyspnoea risk with ticagrelor.

TLDR IM and PM get less benefit from clopidogrel; switch to ticagrelor or prasugrel for ACS/PCI. NM/RM/UM can use standard clopidogrel. Do not rely on higher clopidogrel dosing to rescue IM/PM.


Objective 5. Evaluate the role of pharmacogenetic testing for clopidogrel in personalised care

Why it matters

  • Clopidogrel is a prodrug. CYP2C19 genotype explains a significant share of inter-patient variability in active-metabolite exposure, platelet inhibition, and cardiovascular outcomes after PCI.
  • Evidence syntheses show PGx-guided P2Y12 selection reduces major adverse cardiovascular events vs standard care, without a bleeding penalty when switching appropriately.

When to test

  • Reactive testing: before or immediately after PCI when clopidogrel is being considered. Highest yield because results change the first-line P2Y12 choice.
  • Pre-emptive panels: store CYP2C19 (and other PGx) results in the record for future cardiovascular decisions.

How to use the result (exam algorithm)

CYP2C19 phenotype → 
  PM or IM → avoid clopidogrel → choose ticagrelor or prasugrel*
  NM/RM/UM → clopidogrel acceptable → weigh bleeding risk, comorbidities, cost
*Avoid prasugrel if prior stroke/TIA; consider age/weight/bleeding risk

Practical considerations that strengthen or limit utility

  • Phenoconversion: strong CYP2C19 inhibitors (e.g., omeprazole at high doses, fluvoxamine) can make a genotypic NM behave like an IM or PM. Check the med list.
  • DDIs for alternatives: ticagrelor has dyspnoea and bradyarrhythmia risks; prasugrel has higher bleeding risk and specific contraindications.
  • Health-system fit: panels lower repeat-testing; results should be stored prominently and trigger EHR prescribing alerts.
  • Equity and cost: clopidogrel is inexpensive; genotype helps target costlier agents to those unlikely to benefit from clopidogrel.

Documentation you can reuse

  • “CYP2C19 2/2 → Poor metaboliser. Per guideline, avoid clopidogrel; start ticagrelor. Reviewed DDIs and bleeding risk. Phenotype stored in EHR for future prescribing.”

TLDR Testing before PCI guides the P2Y12 choice at the point of maximum impact. IM/PM should not receive clopidogrel; NM/RM/UM can. Always check DDIs, bleeding risk, and contraindications, then document genotype, phenotype, action, and follow-up.


L12.2 PGx in Mental Health

Objective 1. Define pharmacogenomics and identify key genes (CYP2D6, CYP2C19, SLC6A4)

What pharmacogenomics is

  • Use genetic variation to predict drug exposure, efficacy, and toxicity, then tailor drug and dose.

Key genes in antidepressant therapy

  • CYP2D6: major clearance pathway for many SSRIs, SNRIs, and TCAs; copy-number variation drives UM phenotypes.
  • CYP2C19: major for citalopram/escitalopram and contributes to TCA metabolism; PMs show higher concentrations and side-effect risk (QT for citalopram).
  • SLC6A4 (5-HTT): SSRI target; 5-HTTLPR S allele lowers transporter expression. Evidence for guiding selection is inconsistent; not used for dose changes in guidelines.
  • ABCB1 (P-gp): limits CNS entry of some antidepressants; associations reported, not yet actionable.

High-yield summary table (from lecture)

Drug class / examples Primary gene(s) PGx consideration (lecture framing)
SSRIs — citalopram, escitalopram, sertraline CYP2C19 PM → ↑ levels and side-effects; consider alternative SSRI or lower dose (QT caution for citalopram).
SSRIs — fluoxetine, paroxetine CYP2D6 PM → ↑ levels/toxicity; UM → ↓ efficacy at standard dose.
SNRIs — venlafaxine, duloxetine CYP2D6 PM → ↑ side-effects; NM → standard dosing.
TCAs — amitriptyline, nortriptyline CYP2D6, CYP2C19 PM → ↑ concentrations/toxicity; adjust dose or choose alternative; TDM helpful.
All SSRIs (target) SLC6A4 5-HTTLPR S allele linked to reduced response in some studies; guideline action not established.

TLDR (Objective 1) Focus on CYP2D6 and CYP2C19: they drive exposure for most SSRIs/SNRIs/TCAs. SLC6A4 and ABCB1 findings exist but are not dosing levers. Use PGx to choose safer agents or adjust dose when CYP function is low or high.


Objective 2. Explain how CYP variation changes antidepressant levels and outcomes

CYP2D6 phenotype effects

  • PM (no function): higher plasma levels, more adverse effects (e.g., paroxetine, fluoxetine, venlafaxine, TCAs).
  • UM (gene duplication): low levels and non-response risk (clear with nortriptyline example).
  • IM/NM: intermediate to expected exposure. Lecture data: nortriptyline PK spans from very high (PM) to very low (UM with multiple copies); PGx- or phenotype-guided dosing improves target attainment vs standard dosing.

CYP2C19 phenotype effects

  • PM: higher SSRI exposure (notably citalopram/escitalopram), more side-effects; QT risk highlighted for citalopram.
  • RM/UM: lower exposure, possible non-response at standard dose for CYP2C19-substrate SSRIs.
  • IM/NM: intermediate to expected exposure.

Clinical consequences and actions (lecture framing)

  • Escitalopram/citalopram in CYP2C19 PM: choose a non-CYP2C19 SSRI/SNRI, or start ~50% lower if must use; monitor for side-effects and QT.
  • Paroxetine/fluoxetine in CYP2D6 PM: expect intolerance; consider alternative or dose reduction.
  • Venlafaxine in CYP2D6 PM: higher side-effect risk; consider alternative or careful titration.
  • TCAs: use genotype to adjust starting dose; use TDM to stay within the therapeutic window.

Case anchor (from lecture)

  • Escitalopram non-response/intolerance with CYP2C192/2 (PM) and CYP2D6 IM → switched to desvenlafaxine (not a CYP2D6/2C19 substrate) → symptoms improved in 4 weeks.

Evidence of benefit

  • Meta-analysis of RCTs: PGx-guided antidepressant therapy increased remission ~1.4× vs standard care.

Practical cautions

  • Phenoconversion: strong CYP inhibitors/inducers can override genotype; review co-medications (e.g., potent CYP2D6 or CYP2C19 inhibitors).
  • Consider comorbidities (e.g., QT risk, hepatic function) and patient preferences.

TLDR (Objective 2) Lower CYP activity → higher levels/toxicity; higher activity → lower levels/non-response. Use PGx to avoid SSRIs cleared by the impaired pathway (e.g., CYP2C19 PM on escitalopram) or to select agents outside CYP2D6/2C19 (e.g., desvenlafaxine). PGx-guided care improves remission rates versus trial-and-error.


Objective 3. Interpret a PGx report to recommend an antidepressant

How to read the report (use this order)

  1. Genes and phenotypes: CYP2D6, CYP2C19 first. Note UM/RM/NM/IM/PM.
  2. Assay notes: CNV status for CYP2D6. List any strong inhibitors that would phenoconvert.
  3. Drug table: map each candidate to the clearing enzyme.
  4. Action: avoid, switch, or dose-adjust. Add monitoring.

Decision grid to apply

Phenotype Avoid / prefer Dosing notes
CYP2C19 PM Avoid citalopram/escitalopram; prefer sertraline, fluvoxamine, or non-CYP2C19 options (e.g., desvenlafaxine) If use is unavoidable, start ~50% lower and watch QT for citalopram
CYP2C19 UM/RM Consider agents not reliant on 2C19 if non-response history on escitalopram/citalopram If using 2C19 SSRIs, titrate to effect; consider TDM if available
CYP2D6 PM Avoid paroxetine, fluoxetine high doses, venlafaxine, TCAs; prefer sertraline, escitalopram (if 2C19 OK), desvenlafaxine, mirtazapine If using a 2D6 substrate, start lower; consider TDM for TCAs
CYP2D6 UM Avoid nortriptyline, paroxetine, venlafaxine monotherapy; prefer drugs outside 2D6 (e.g., desvenlafaxine) If unavoidable, higher doses may still underperform; TDM helps

Worked case 1 (from slides): “Emily”

  • Report: CYP2C19 2/2 → PM, CYP2D6 IM.
  • Problem: escitalopram non-response and side-effects.
  • Action: switch to desvenlafaxine (minimal CYP2D6/2C19 reliance).
  • Outcome: symptom improvement at 4 weeks.

Worked case 2 (TCA)

  • Report: CYP2D6 PM, CYP2C19 IM.
  • Goal: nortriptyline for neuropathic pain.
  • Action: choose alternative first. If used, reduce starting dose and use TDM to target range.

One-minute note template

  • “PGx: CYP2C19 PM, CYP2D6 IM. Avoid escitalopram/citalopram. Start desvenlafaxine. Review co-meds for CYP inhibitors. Plan follow-up in 2–4 weeks; monitor BP and adherence.”

TLDR Read CYP2C19 and CYP2D6 first. Avoid substrates of the impaired pathway. Prefer agents outside that pathway or adjust dose with monitoring. Record phenotype, action, and follow-up.


Objective 4. Assess benefits and limitations of PGx-guided prescribing in mental health

Benefits (lecture data)

  • Higher remission with PGx-guided care vs usual care (meta-analysis ~1.4×).
  • Fewer adverse effects when avoiding high-exposure scenarios (e.g., 2C19 PM on citalopram).
  • Faster effective selection after prior non-response.
  • TDM synergy for TCAs when genotype predicts extremes.

Limitations and cautions

  • Phenoconversion: potent inhibitors/inducers can override genotype; always check the med list.
  • Evidence heterogeneity: not all endpoints or classes show equal benefit; SLC6A4/ABCB1 remain non-actionable for dosing in guidelines.
  • Complex phenotypes: depression outcomes depend on adherence, comorbidity, environment, and diagnosis accuracy.
  • Assay scope: inaccurate CYP2D6 phenotype if CNVs not measured.
  • Workflow: turnaround, cost, and access vary; store results in the record to reuse.

When PGx adds the most value

  • Multiple medication failures or intolerance.
  • Planned use of TCAs or high-risk SSRIs (e.g., citalopram in 2C19 PM).
  • Polypharmacy where phenoconversion risk is high and you need a CYP-sparing option.

Clinic checklist

  • Confirm phenotypes for CYP2D6 and CYP2C19.
  • Map candidate drugs to clearing enzymes.
  • Screen for DDIs and QT risk.
  • Prefer non-CYP or CYP-sparing agents when phenotypes are extreme.
  • Document plan and schedule follow-up within 2–4 weeks.

TLDR PGx improves remission and tolerability when you align drugs with CYP2D6/2C19 phenotypes. Limits arise from phenoconversion, variable evidence for non-CYP genes, and access. Use PGx where the decision stakes are high and store results for reuse.


L12.3 Drug Safety for 5-FU in Cancer

Objective 1. How chemotherapy drugs work and why they have side effects

  • Goal: reduce tumour growth and spread by killing rapidly dividing cells. Targets include DNA/RNA synthesis, microtubules, signalling pathways, and nucleotide biosynthesis. Anti-metabolites such as 5-fluorouracil (5-FU) block nucleotide synthesis.
  • Low therapeutic index explains toxicity. Doses that suppress tumours also injure normal fast-turnover tissues: bone marrow, GI mucosa, hair follicles. Side effects include myelosuppression, mucositis, diarrhoea, and alopecia.
  • Fluoropyrimidines in context: used in metastatic or hard-to-treat cancers (e.g., colorectal, breast, upper GI, head and neck). Capecitabine is an oral prodrug of 5-FU.

TLDR Cytotoxics kill fast-dividing cells. That includes tumours and normal renewal tissues, so toxicity is common. 5-FU is an anti-metabolite used widely in solid tumours. Capecitabine is its prodrug.


Objective 2. Metabolic pathways relevant to fluoropyrimidine activation and elimination

Activation

  • 5-FU is converted in cells to FdUMP, which inhibits thymidylate synthase, blocking de novo thymidylate synthesis and DNA replication.
  • Capecitabine → 5-FU: sequential activation culminates in conversion by thymidine phosphorylase (TP). TP activity is higher in some tumours, which can concentrate 5-FU in tumour tissue relative to normal tissue.
Capecitabine (oral) --(TP, higher in tumours)--> 5-FU --(cell enzymes)--> FdUMP
FdUMP ┤ Thymidylate synthase → ↓ dTMP → DNA synthesis blocked

Elimination

  • Rate-limiting catabolism: dihydropyrimidine dehydrogenase (DPD), encoded by DPYD, inactivates 5-FU. Reduced DPD activity elevates circulating active species and toxicity risk.
  • Tumour DPD matters: higher tumour DPD correlates with poor response to 5-FU infusion, consistent with intratumoural inactivation.
  • Clinical impact of DPYD variants: four clinically relevant variants highlighted (e.g., c.1905+1G>A [*2A], c.1679C>G [*13], c.2846A>T, HAPB3). Variant carriers have markedly higher risk of severe toxicity and treatment-related death; a pooled analysis showed a ~35-fold increase in treatment-related mortality among variant carriers vs noncarriers.
  • Genotype vs phenotype testing: genotype by PCR for key DPYD variants; phenotype by measuring DPD activity in PBMCs. Both approaches were described, with wide inter-individual variability in activity.

TLDR Activation: 5-FU → FdUMP inhibits thymidylate synthase; capecitabine uses tumour-enriched TP to generate 5-FU locally. Elimination: DPD (DPYD) inactivates 5-FU. Low DPD activity from DPYD variants drives severe toxicity and higher treatment-related mortality; tumour DPD can blunt efficacy.


Objective 3. Side effects specific to 5-FU

Toxicity profile you should name

  • Myelosuppression: neutropenia, thrombocytopenia; earlier and more severe with bolus dosing.
  • Gastrointestinal: mucositis, stomatitis, diarrhoea; more common with continuous infusion and capecitabine.
  • Hand–foot syndrome: palmar-plantar erythrodysesthesia; prominent with capecitabine and infusional 5-FU.
  • Cardiotoxicity: chest pain from coronary vasospasm; monitor during infusion, stop drug and manage urgently.
  • Neurotoxicity: acute encephalopathy with hyperammonaemia; rare but severe.
  • Ocular/dermatologic: conjunctivitis, photosensitivity, nail changes.

Red flags for early severe toxicity

  • Onset after first cycle with disproportionate mucositis, diarrhoea, neutropenia, or neurotoxicity suggests DPD deficiency. Urgently stop therapy and evaluate.

Bolus vs infusion quick table

Schedule More likely toxicities
Bolus 5-FU Myelosuppression
Infusional 5-FU / Capecitabine Mucositis, diarrhoea, hand–foot syndrome, cardiotoxicity events during infusion

TLDR 5-FU toxicities cluster by schedule. Bolus drives marrow suppression. Infusion/capecitabine drive mucositis, diarrhoea, and hand–foot syndrome, with infusion-time vasospasm risk. Severe early toxicity signals DPD deficiency.


Objective 4. Dose adjustment guidelines by genotype and current Australian practice

Clinically relevant DPYD variants covered in the lecture

  • c.1905+1G>A [*2A], c.1679C>G [*13], c.2846A>T, c.1129-5923C>G (HAPB3). These reduce DPD activity and increase risk of severe toxicity and treatment-related death.

Activity score (AS) and CPIC-style actions shown on slides

DPYD activity score Phenotype label Starting recommendation for 5-FU or capecitabine Follow-up
2.0 Normal Full dose Usual monitoring
1.5–1.0 Intermediate Reduce starting dose by 50% (range 25–50% per regimen), then titrate using toxicity and, if available, TDM Escalate cautiously if well tolerated
0.5–0 Poor Avoid fluoropyrimidines Choose non-fluoropyrimidine regimen

Genotype vs phenotype testing

  • Genotype detects the main risk alleles above.
  • Phenotype measures DPD activity in PBMCs and captures non-genotyped causes of low activity. The lecture described both and showed wide activity variability.

Australian implementation highlights

  • Pre-treatment DPYD testing is available, with the four-variant panel common in clinical labs.
  • Slides note Australian rebate pathways and the RCPA context for indications. Use local lab guidance for ordering and reporting.

How to write the prescribing note

  • DPYD AS 1.0 (intermediate) due to c.2846A>T. Start capecitabine at 50%, review toxicity day 7–14, adjust by tolerance. Consider TDM if available.”

Clinical cautions from the lecture

  • Do not “catch up” quickly after a reduced start.
  • Watch for phenoconversion from interacting drugs or organ dysfunction.
  • If severe toxicity occurs despite dose reduction, stop and reassess genotype/phenotype.

TLDR Use DPYD activity score to set the first dose. AS 2 gets full dose. AS 1.5–1 starts at 50% with careful titration. AS ≤0.5 avoids 5-FU/capecitabine. Australian labs offer four-variant panels with rebate notes on the slides. Document AS, starting dose, and the titration plan.