L10, Gene therapy Flashcards

1
Q

Gene therapy definition:

A
  • The introduction, using a vector, of nucleic acids into cells with the intention of altering gene expression to prevent, halt or reverse a pathological process
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

4 Key groups of targets for gene therapy:

Include examples for each

A
  • Single gene recessive LOF -> Gene addition or replacement (e.g. CF, haemophilia)
  • Single gene haploinsufficiency -> Gene addition (e.g. DSH)
  • Single gene dominant negative -> allele silencing or replacement (e.g. HD)
  • Multi-gene or acquired -> addition of therapeutic gene (e.g. cancer, heart disease, rheumatoid arthritis)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Gene therapies of cancer:

A
  • Mostly CAR-T
  • Largest portion of gene therapies in use
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

In vivo gene therapy:

A
  • Single step
  • Vector administered (injected or inhaled) directly to patient, targeted specifically to organ or tissue
  • Targeting somatic cells only
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Ex vivo gene therapy:

A
  • Two steps
  • Cells removed from patient, vector added to cells in vitro then engineered cells returned to patient
  • May be combined with (stem) cell-based therapy
  • Targeting somatic cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Discuss the processes and relevant barriers to gene therapy:

A
  • Circulating antibodies target the vector for immune destruction -> often a basis for exclusion from trials
  • Uptake into cells requires expression of specific receptors
  • Transport into nucleus
  • Must replicate either by integrating into host DNA forming an episome for independent replication
  • Transcript must be processed to produce a protein; epigenetic effects can prevent this (e.g. insertion into heterochromatic region) -> no control over where vector inserts into genome
  • If the protein is unrecognised by the immune system, it will be attacked (e.g. in MD if patient totally lacks dystrophin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

List possible vectors for gene therapy with examples where relevant:

A
  • Adenovirus
  • Adeno-associated virus
  • Gamma-retrovirus (e.g. Moloney murine leukaemia virus-derived)
  • Lentivirus (e.g. HIV-derived)
  • Routine plasmids
  • Mini circles
  • Transposons (e.g. sleeping beauty)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Sites for in vivo gene therapy:

A
  • Accessible organs: lungs, skin, muscles
  • Inaccessible: liver, retina, brain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Vectors and key targets for treatment (in vivo):

A
  • Adenovirus, AAV, some use of retroviral vectors
  • Single gene disorders and acquired disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Advantages and disadvantages of adenoviral vectors:

A

+

  • Large capacity
  • Easily purified
  • Infect broad range of cell types
  • Efficient transduction

-

  • High incidence of neutralising antibodies (common cold)
  • Capsid protein is highly immunogenic
  • Potentially fatal inflammatory response
  • Transient expression of transgene
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Adeno-associated virus: Overview

A
  • Small
  • Non-pathogenic, minimal immune response
  • rep and cap genes can be replaced with expression cassettes (limited capacity)
  • Can be used in non dividing cells (maintained as episome)
  • Different serotypes target different tissues
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Leber’s congenital amaurosis (LCA):

Clinical features and molecular basis:

A
  • Amaurosis: vision loss without obvious physical signs
  • Early onset blindness
  • Autosomal recessive (14 genes including RPE65)
  • RPE65 codes for retinal pigment epithelium-specific 65 kDa protein -> required for photoreceptor function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Use of gene therapy for LCA:

A
  • Vision restored in mouse and dog LCA models using AAV vectors containing RPE65
  • Successful phase II clinical trials
  • Subretinal injection -> AAV2 serotype capsids -> virus taken up by retinal epithelium
  • -> RPE65 gene expressed from episomal vector -> light sensitivity restored, maintained for >3 yrs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Ex-vivo gene therapy for SCID: Basic principle

A
  • IL2RG or ADA gene
  • Use f a gamma-retroviral vector
  • Aiming for long-term reconstitution of lymphoid lineages
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Potential target organs for AAV vectors:

A
  • Liver: Gene factory, metabolic disorders
  • Muscle (gene factory; trials for haemophilia B, alpha antitrypsin deficiency, LPL deficiency
  • Repair of muscle disorders (DMD)
  • Brain (immunoprivileged site, BBB, trials for Parkinson’s disease, Canavan’s disease, Batten’s disease)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

SCID gene therapy trials: ADA vs X-linked

A
  • gamma-retrovirus vectors
  • ADA enzyme replacement therapy withdrawn ensuring transduced cells have selective advantage -> 9 out of 10 had apparently permanent curative effect
  • X-linked therapy, immune function restored in all but 5 out of 20 developed leukaemia (insertion into LMO2 proto-oncogene -> activation acts synergistically with IL-2R to promote cell proliferation)
  • Issue: gamma-retroviruses tend to insert near promoters (LTR: Enhancer activity for nearby promoters)
16
Q

Alternatives to gamma-retroviruses:

A
  • Lentiviruses (LTRs lack strong enhancer; self-inactivating vectors delete LTRs for additional safety)
  • DNA vectors (simple plasmids/minicircles; no pre-existing immunity, high capacity but delivery in vivo is very difficult)
17
Q

Targeted changes for gene therapy:

A
  • Programmeable nucleases to recognise specific site (e.g. CRISPR-Cas9)
  • DSBs induced-> NHEJ leads to gene disruption
  • Gene editing or replacement by homology directed repair is more efficient, with staggered cuts
  • Challenging to avoid apoptosis, off-target mutations, optimal vector design, need for DNA replication for HDR
18
Q

+ 3 types of programmable nucleases for genome engineering:

A
  • Zinc finger nucleases (ZFNs)
  • Transcription-activator-like effector nucleases (TALENs)
  • Cas9 RNA-guided engineered nucleases (RGENs)
19
Q

+ Outline the basic principle of genome engineering with programmable nucleases:

A
  • DSBs induced -> repaired by homology-directed repair
  • Leads to gene insertion, correction and point mutagenesis/NHEJ -> gene disruptions
20
Q

+ What is an alternative to programmable nucleases which induces single breaks:

A
  • Programmable nickases -> single-strand breaks
  • Repair of these leads to precise genome editing
  • Can be used in pairs -> much more specific than nucleases