Gene Transfer Therapy Flashcards

1
Q

How is gene therapy defined?

A

The deliberate alteration of the nucleic acid content in somatic cells for therapeutic benefit.

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

How is the wider field of gene therapy operated historically?

A

This is a concept that has been being tested since 1989, with over 2200 clinical trials since then; around 64% of which targeted cancer, 9.5 monogenic diseases (including SCID) and 7.9% for both cardiovascular and infectious diseases (esp. HIV).

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

What is the safety/success record of gene therapy so far?

A

80% of all trials have used viral vectors. While there have been some clear benefits shown, particularly for SCID, cancer and CVD, there has been relatively low progression to phase II and III trials.

While the safety of gene therapy treatments has been largely positive, there have been some deaths associated with it, largely confined to a SCID trial which induced leukaemia in some patients.

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

What is the main benefit of gene therapy?

A

The potential to cure a disease with a single treatment, rather than a lifelong drug regimen.

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

What are the different approaches one can take to gene therapy?

A

They are classified based on their effect on the target gene:

• Gene transfer
o Plasmids, adenovirus, adeno-associated virus

• Gene silencing
o ASO, RNAi, miRNA, CRISPR-interference

• Gene manipulation
o Exon splicing

•	Gene alteration
o	SFHR (small fragment homologous replacement), CRISPR/Cas9, Zinc-Finger Nucleases, TALENs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is the most common way of enabling gene therapy?

A

Most gene therapy approaches utilise viruses for their ability to transfer genetic material with high efficiency both in- and ex-vivo.

Earlier studies tended to use ssRNA retroviruses, or dsDNA adenoviruses – which do actually still predominate.

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

What is one way to deal with the size restrictions imposed by vectors?

A

The human genes used in transfer do not tend to be the full length chromosomal genes due to the huge amplification in length caused by the introns (often more than 3x).

Instead the cDNA is often used, although this can lead to loss of the long term expression control that the intron sequences can provide.

To compensate for this, mini-genes are often designed with a few introns inserted into the sequence, typically in a concatenated bunch after 2-3 exons, to augment proper expression.

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

What are some considerations that must be made when designing an expression system?

A

Which gene to target/transfer, whether long or short term expression is required, the safe and effective expression level, tissue/organ specific expression, the dividing activity of the cells, and whether the gene is required to be inducible.

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

What is short term expression useful for in therapy? Give examples of this.

A

Short term expression is useful for the trials which aim not to cure a monogenetic disease, but to temporarily treat an existing acquired condition using the therapy.

For example, in CVD this may take the form of attempting to regress or stabilise plaques by short term upregulation of ApoA1 and thus HDLs.

This has also employed ectopic expression; engineering muscle cells to secrete ApoE3 to increase its plasma concentration, and engineering macrophages to secrete ApoA1 specifically at lesion sites.

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

What is perhaps the biggest challenge to adequate gene therapy treatment?

A

Vector delivery.

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

What are the main categories of vector delivery mechanisms?

A
  • Receptor mediated delivery
  • Membrane fusion
  • Ultrasound permeabilisation
  • Ex-vivo techniques
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What is receptor mediated vector delivery?

A

Certain viruses display ligands that are recognised by membrane receptors, allowing for invasion. These can be exploited for delivery, including that of plasmids coated in lipid nanoparticles.

By tagging a virus with a particular ligand, organ specificity can be achieved.

Galactose tagging enables specific uptake into liver in mice.

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

What is membrane fusion mediated delivery?

A

This uses artificial vesicles containing plasmids, which can fuse with the plasma membrane to deliver their genetic payload.

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

What is ultrasound permeabilisation in vector delivery?

A

Used to allow the vectors to escape blood vessels in specific locations.

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

What are the ex-vivo techniques for ex-vivo vector delivery?

A

o Microinjection

o Electroporation

o Gene gun
 Fires genes in gold nanoparticles

o Useful for cellular therapy

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

What are the advantages of plasmid vectors?

A

Interest in plasmids as a vector has been renewed of late. Although they have been disfavoured due to difficulties in establishing efficient expression, they can be manufactured with both high yield and purity – a feat that is difficult to match with viral vectors.

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

How are plasmid vectors delivered?

A

In order to deliver the plasmids, electroporation can be used in animals for ex-vivo transfection, and hydrodynamic injection (rapid intravenous hepatic injection of a large dose) is effective in rodents, but may prove difficult in larger organisms.

Less invasive techniques include coating the plasmids in cationic liposomes. These can be targeted to specific organs by chemical addition of ligands – particularly using galactose. Tetrameric galactose is often used to increase uptake via the asialoglycoprotein receptor.

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

How can plasmid vectors be modified to improve performance after delivery?

A

S/MAR motifs are tetrameric 155bp additions used to increase retention of the episome after division by binding it to the nucleus. These are, however, difficult to construct and modify for use, and virus-based constructs still give far more stable expression.

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

What are the two broad classes of viral vectors?

A

Viral vectors can be classified as integrating or non-integrating, depending on whether the DNA payload is incorporated into the human genome of whether the vector instead forms an extrachromosomal element.

Integrating vectors, including lentiviruses and γ-retroviral vectors are stably incorporated upon cell division, but their recombination can disrupt genes – as occurred in the SCID treatment failure.

Non-integrating vectors, such as adenoviruses and AAVs, can transduce quiescent cells effectively, but are quickly lost in fast-dividing cells.

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

How can more body-amenable viral vectors be produced?

A

Molecular techniques now enable recombinant viruses to be produced with ‘scrambled’ capsids. Such a library allows direct in-vivo panning (screening) to select viral vectors with specific properties e.g. tissue tropism or low immunogenicity.

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

What stages must a viral vector carry themselves through to express a gene? How can this be avoided?

A
Cell association
endocytosis
DNA release
nuclear uptake
transcription
translation

ASO and RNAi treatments bypass many of the steps by acting within the cytoplasm, including the complex formation of dsDNA within the nucleus required for integration or episomal expression (a process which varies between vectors).

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

What viral function must be removed to produce a useful vector?

A

Viral vectors can either be replication-competent of replication-defective. Replication-competent retroviruses possess all the genes requires to continue propagating themselves after infection, whereas those that are defective deliver their payload but fail to continue the typical lytic pathway.

Replication defective retroviruses are far more commonly used, primarily for safety reasons but also because the lack of many parts of the viral genome creates more space for the transgene.

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

What problem does the use of replication-defective viruses cause?

A

Because the vectors produced must be unable to replicate, this makes them more difficult to produce in the first place.

24
Q

How are replication defective viruses made?

A

Bby infecting producer cells (typically human embryonic kidney (HEK-293) cell lines) with two sets of DNA; the replication-defective vector DNA (containing the therapeutic expression cassette) and helper DNA which contains the genes required for replication and capsid synthesis removed from the vector DNA.

The helper DNA replicates and produces all the capsid components, and enables replication of the vector DNA. However, it lacks the packaging domain (psi) that is recognised by the developing capsid preventing it from producing viable viruses. The vector DNA does contain the psi domain, so only it is packaged into the capsids produced from the helper DNA, producing large amounts of a virus which cannot replicate.

25
Q

What are retroviruses?

A

These are integrative (RT encoding) ssRNA vectors which generally have high efficiency in delivering to dividing cells ex-vivo, having been predominantly used in this manner to treat haematopoietic and immune cells in SCID, leukaemia and thalassaemia. One example is the murine leukaemia virus. Replication-defective retroviruses have a capacity of up to 8-10kb.

26
Q

What are enveloped retroviruses?

A

Enveloped retroviruses use membrane fusion to deliver their genomes into target cells. Following nonspecific adhesion to the cell surface, viral attachment glycoproteins bind specifically to their cognate receptors, whereupon binding becomes irreversible and the membranes fuse. The host range of the retrovirus is determined by the ability of the viral envelope protein (Env) to bind to the cellular receptor.

27
Q

What are lentiviruses?

A

Lentiviruses are a commonly used subclass of retroviruses (including HIV-1) which also infect non-dividing cells (giving a wider tissue tropism – variety of tissues that the gene can be delivered to). This includes the ability to target livers in-vivo. Lentiviruses are always replication-defective for safety reasons.

28
Q

Where do lentiviruses integrate?

A

Recent research has shown that their integration is not random, but directed by integrase to specific active genes – particularly those localised to the periphery of the nucleus (Marini et al, 2015).

They do appear to have lower tendency to integrate into tumorigenic areas than γ-retroviral vectors (Cattoglio et al, 2007)

29
Q

What are the advantages of using lentiviruses?

A

Lentiviruses have the advantages (other than the permanent, multigenerational integration) of low immunogenicity, a packaging capacity of up to ~7.5kb (al Yacoub et al 2007) and the ability to be pseudotyped with alternative envelopes to alter tropism.

30
Q

What is an alternative use of lentiviruses made possible by editing?

A

Point mutations that inactivate integrase, or long terminal repeats, have been used to abolish the genome integrating capacity of lentiviruses to produce short-term, non-integrating vectors with greater safety profiles. These have been considered for use in vaccination and tumour-cytotoxic therapies.

31
Q

What are the advantages of adenovirus vectors?

A

Ad or adenoviruses have now overtaken RV as the vector most used in clinical trials.

They are efficient, easy to prepare in good yield and high concentration, infect a broad range of cells and are non-tumourigenic as their DNA does not integrate, though this also limits their use due to their lack of replication in cells during division.

By using a ‘gutless’ vector with a helper virus (production of which complicates purification) you can produce huge capacity – up to 35kb.

32
Q

What adenovirus feature can be exploited to promote delivery?

A

Adenoviruses also have various serotypes (which variation of capsid proteins are used) which can be exploited for cell invasion, such as Ad serotype 5 which binds initially to Coxsackie and Adenovirus Receptor (CAR), with integrin co-receptors allowing for receptor-mediated endocytosis.

33
Q

What poses a significant barrier to adenovirus activity?

A

Due to human exposure to pathogenic adenoviruses, many patients have latent antibodies against them which may prevent delivery of the vector. The use of adenoviruses that exclusively infect non-human animals is being explored to compensate for this.

34
Q

What leads some to have reservations over the use of adenovirus vectors?

A

Adenoviruses were the vector used in the clinical trial the resulted in the highly publicised death (from massive immune response) of an 18-year-old – the first recorded gene therapy death – amidst accusations of misconduct in the trial due to financial conflicts of interest.

This led to significant reduction in the use of adenoviruses, with more emphasis being placed upon vectors such as Adeno-associated viruses (AAV).

35
Q

What is AAV and what advantages does it have?

A

AAV, a specific parvovirus species, has high uptake by both dividing and non-dividing cells.

They have low immunogenicity, not being known to cause any human disease, meaning that there are rarely existing antibodies. If there are, this can be dealt with using immunosupressors or by switching serotype.

Despite usually being non-integrating episomes their expression is often sustained (only wild-type integrates, purely episomal as a vector). They also have the advantage of being able to scale up pharmaceutically. However, their capacity is limited to 4.5kb.

36
Q

Which AAV serotype enables improved delivery?

A

AAV2 is taken up by HSPGs, αVβ5-integrins and FGFR-1. The HSPG acts as the primary receptor, with the other two being co-receptors that allow for clathrin-coated pit localisation and receptor-mediated endocytosis.

The pH drop in the late endosome leads to viral release and a change in conformation of the capsid proteins, allowing for nuclear pore complex interaction and release of the DNA into the nucleus, where it is replicated (in replication-competent varieties) or sometimes integrated into the host genome.

37
Q

How do the various AAV serotypes compare?

A

AAV2, although effective in vitro, has performed poorly in vivo. Others have greater tissue tropism and uptake rate.

AAV1, 5 and 7 all have high transduction rates in mouse skeletal muscle. AAV6 has a high transduction rate for airway epithelial cells, and AAV8 is highly efficient at transducing hepatocytes.

Isolation of new serotypes is difficult, but once done their capsid protein encoding genes can be engineered into helper plasmids to produce vectors with that serotype.

38
Q

What is the disadvantage with AAV2 serotype?

A

The AAV2 serotype uncoats in the nucleus slower than AAV8 which leads to the ssDNA transgenes being released more individually.

This is not conducive to the annealing of the strands to form the dsDNA required for transcription. The ssDNA is more likely to be recognised and broken down.

39
Q

How can the disadvantage with AAV2 uncoating be overcome?

A

AAV8 serotypes uncoat faster, producing a higher ssDNA concentration leading to faster annealing, increasing the lifespan and activity of the DNA.

To acquire both this and strong uptake, AAV2/8 vectors are used which cross-package the AAV2 genome into an AAV8 serotype.

A recombinant AAV vector can be produced using different serotypes to produce serotype hybrids, which can compromise or combine benefits of either.

40
Q

What is the composition of AAV capsids?

A

There are three protein subunits that comprise the capsid, VP1, VP2 and VP3.

All share the majority of their structure, with VP3 being only the commonly shared structure, VP2 having that with an additional section, and VP1 having both of those sections with another addition. VP1, 2 and 3 form a capsid by combining in a ratio of 1:1:18.

41
Q

How can AAV capsid proteins be strategically modified?

A

This can be exploited for adding peptide sequences to increase uptake or tropism. By only adding the peptide sequence to VP1 or VP2 + 1, the disruption is reduced and infectivity is maintained. 14 residue core RGD peptide insertion is possible in several sites in VP3 without disruption, allowing them to interact with integrins to greatly increase transduction efficiency.

42
Q

What are mosaic and chimeric AAV vectors?

A

Mosaic vectors can be produced by using different subunits from different AAV serotypes (i.e some of each VP from different serotypes), and chimeric vectors have the VP proteins genetically engineered with residues or domains swapped between the different serotypes to create more intimate hybrids.

Since there are 10-12 well characterised serotypes, the possibilities are huge, and may lead to great potential in increasing transduction efficiency and tissue specificity.

43
Q

What is scAAV?

A

AAV are usually ssDNA vectors, with a mixture of sense and antisense genomes being packaged individually into the vectors to anneal in the nucleus.

Self-complementary AAV (scAAV) vectors skip this step by encapsulating the fully formed intramolecular dsDNA thus enabling far more rapid transgene expression. However, in order to do this the genome must be approximately half the size of the full capacity in order to fit into the capsid.

44
Q

How are AAV vectors made?

A

To produce an AAV vector, the vector plasmid is inserted into the producer cells with inverted terminal repeats at either end of the transgene, which form hairpins allowing the host cell replication machinery to synthesise both the sense and antisense versions depending on which end it began at, hence how each gets packaged.

45
Q

How are scAAV vectors made?

A

In scAAV production, one of the ITRs is mutated. This means that replication can only initiate at the wildtype ITR and proceeds through the mutant end without terminal resolution, using the opposite strand as a template, to create the dimer. The product is a self-complementary genome with the mutant ITR in the middle.

46
Q

How have scAAV been used in therapy?

A

A UCL team designed the hepatocyte-specific scAAV2/8.LP1 vector to treat haemophilia B (Factor IX deficiency). The long term results indicate effective therapeutic benefits with no long term toxic effects.

47
Q

What are the gene therapy targets which would be overexpressed to treat CVD?

A
o	LDLR
o	ApoA1
o	ApoE3
o	ABCA1
o	LPL
48
Q

Which is the only OE CVD gene therapy target in trials?

A

LPL is the only target listed with gene therapy in clinical trial for hyperchylomicronaemia treatment.

Glybera is used for this, a variant of the human LPL gene with higher activity, AAV vector injected intramuscularly. This was the first EU approved gene therapy.

49
Q

What potential targets for gene silencing therapy are there in CVD? Describe the available treatments.

A

o ApoB
 Exon skipping strategies also used
 Kynamro ASO treatment

o MTP
 Juxtapid ASO treatment

o CETP

o PCSK9
 RNAi in human trials (lipid nanoparticle suspension vector)

o ApoCIII

50
Q

What ex vivo CVD-targeting gene therapy strategies have been attempted?

A

A 1995 clinical trial, hepatocytes were isolated from FH patients and transduced with RV expressing the human LDLR gene, in an attempt to increase expression, before they were re-implanted into the patients.

There was limited success in terms of efficacy, but did confirm the safety and feasibility of such treatments with continued expression. Two out of the five patients did see a clear benefit lasting for 2.5 years. However, the method is highly labour intensive, and no use of similar strategies has since been attempted.

51
Q

Why are ApoE (-/-) mice commonly used as models for CVD gene therapy?

A

These are a useful model due to the relative ease of gene transfer therapy. A wide range of over or under-expressed genes have been trialled in this model to ameliorate the hypercholesterolaemia, including the added expression of mouse LDLR, as would later be trialled in humans, and that of human ApoA1 to raise HDL levels.

52
Q

What trials were performed on ApoE mice?

A

Hepatic targeted gene therapy centred around expression of ApoE3 in these mice (using first generation Adenovirus vectors).

The previously mentioned scAAV2/8.LP1 vector has also now been used to transfer ApoE3 and ApoA1 to mouse hepatocytes in an update of the previous experiment.

53
Q

What was the outcome of the first ApoE trial?

A

This greatly reduced plasma cholesterol initially, however the effects were lost with the ApoE transgene expression which progressively declined to negligible levels after 56 days. However, this did during this time lead to a reduction in lesion area of 38%.

54
Q

What caused the loss of expression in the initial ApoE trial?

A

The loss of expression may have been due to immune response or promoter shutdown (by epigenetic repression). These studies used strong viral promoters such as those from cytomegaloviruses (CMV) which give strong expression but elicit faster epigenetic response.

Immune response is also likely because not only could there be a response to the capsid or the expressed ApoE3, but the first generation adenoviruses were ‘leaky’ and prone to expressing small amounts of viral protein which could be immunogenic. Second generation adenoviruses, and the ‘gutless Ad’ vectors do not have this issue.

55
Q

What was the outcome of the scAAV ApoE trial?

A

Once again the transfer of ApoE3 essentially normalised the lipoprotein profile, and significantly reduced the lesion coverage, with expression lasting far longer than the previous trial.

The addition of ApoA1 raised the HDL level but did little to lower the LDL profile. ApoA1-Milano, a natural atheroprotective mutation (R173C) was also used, but suprisingly performed more poorly than wildtype.

56
Q

What notable phenomenon was shown in both ApoE expression gene therapy mouse trials?

A

In both experiments there was a significant gender bias, with males responding far more positively than females, thought to be due to slower uncoating, though the reasons for this remain unclear. It is known to affect single-strand vectors via an androgen-dependent pathway, as castrated males show similar expression to females.