1/48
Looks like no tags are added yet.
Name | Mastery | Learn | Test | Matching | Spaced |
---|
No study sessions yet.
Diseases being targeted by gene therapy
cancer
monogenic disease
infectious diseases
cardiovascular diseases
Gene therapy strategies
aim to deliver a gene in order to overcome disease/infection
non functional gene is replaced with functioning gene
suicide gene
for cancer
use enzyme not found in cells, substrate binding = production of toxic substance that kills cells
for a nonfunctional gene, deliver a gene which produces a product that blocks the expression of the faulty gene
gene editing to correct defects
Gene therapy: In Situ Therapeutic Protein Production
in situ - on site
therapeutic proteins are hard to produce and expensive
e.g. deliver anti-her2 mAb gene for herceptin for her-2 +ve cancer and get pt own cells to produce herceptin and release into systemic circulation
Suicide genes
gene-directed enzyme pro-drug therapy (GDEPT)
often uses a combination of HSV thymidine kinase and ganciclovir
HSV TK has > 1000 fold greater efficiency in phosphorylating ganciclovir than mammalian kianses
not well phosphorylated normally
incorporated into DNA as its similar to guanosine and induces apoptosis
nucleic acid based therapies
decoy oligonucleotides - interfere with transcription of a gene by binding tot he promoter gene and stop it being expressed
antisense oligonucleotide - stop translation by being complementary to a specific sequence and stopping the translation into protein that causes disease
RNA interference
siRNA
knock down gene expression if sequence is known
binds to mRNA and induces cleavage which degrades mRNA
reduce expression of gene at protein level
miRNA
micro RNA
derived from genes that specifically code mRNAs and not proteins
important in regulating gene expression
cause destabilisation and reduction of translation into protein instead of mRNA destruction
over and underexpressed in many cancers
role of miRNA-34a
involved in pathways that drive proliferation survival of cells
regulates pathways that control cell proliferation, migration and invasion, resistance to apoptosis and immune evasion
involved in cancers
targets include androgen receptors (prostate), PDL-1, C-MYC and CD44
miRNA in clinic
chemically modified to make stable in serum
specific targeting
inhibition of in vivo tumour growth
caused complete cure in one animal
direct delivery of genes/nucleic acids into cells
put therapeutic gene/oligonucleotide/miRNA in a suitable vector (virus)
inject into patient
targeting will allow it to reach the cell and delivery
cell-based delivery of genes/nucleic acids into cells
take a patients own cells
modify in vitro
select for cells that have been modified and put them back in
could be used in sickle cell anaemia
challenges to deliver
complex formation
unstable in serum
don’t get into cells well
need to get through nuclear membrane to get into nucleus
may need to wait until dividing
transport from delivery site
Retrovirus
ssRNA genome
100nm in diameter
some infect only dividing cells
lentiviruses can do it all
can be used to target specific cells
risk of immune response and tumour development
Lentiviruses
randomly insert into genome
if they insert into a tumour suppressor gene then they can stop it acting
if they insert near a proto-oncogene they can turn it on
Adenovirus
Infects dividing and non-dividing cells
Doesn’t integrate
Can be used to target specific cell types
Possible immune response
Gendicine
recombinant andenovirus with p53
used to treat head and neck cancer
used in combo with chemotherapy
Ocorine
oncolytic virus - a virus that selectively infects and kills tumour cells
Oncolytic viruses are engineered to take advantage of key regulatory factors of the cell cycle and the virus life cycle
Oncorine can’t replicate in normal cells due to induction of p53, but can do so in p53-deficient tumour cells
modified virus will proliferate in p53 deficient cancer cells and kill them
Adeno-associated viruses
ssDNA genome
Genome ~5 kbp
~20 nm diameter
Infects dividing and non-dividing cells
Specific integration (chr. 19)
Can be used to target specific cell types
Little immune response
Glybera
Adeno-associated virus serotype used as vector
For the treatment of lipoprotein lipase deficiency (LPLD)
Rare disease, affecting 1-2 people per million
Causes dramatically increased fat levels in the blood
Glybera was administered as a one-time series of small i.m. injections into the leg
Physical non-viral nucleic acid delivery methods
Physical
direct
gene gun
ultrasound
electroporation
Chemical non-viral nucleic acid delivery methods
for in vivo
encapsulation with polymer
complexation
Advantages and disadvantages of non-viral vectors
ADV:
Easy to prepare and modify (e.g. targeted) because they’re synthetic
Reduced immunogenicity
Large capacity
DADV:
Transient
Poor efficiency
Types of non-viral vectors
liposomes
dendrimers
cationic polymers
cell penetrating peptides
Lipoplexes
DNA/RNA + liposome
interact with cell surface and get taken up into cell
Dendrimers
Wide range of chemical approaches and functional groups
Surface groups can be functionalized for targeting, stealth, etc
Can be designed to release under specific conditions
Amine-rich dendrimers for complexation with negatively charged nucleic acids (N means it’s positively charged)
Cationic Polymers
mix with nucelic acid
form a circular complex called toroid
Cell-penetrating peptides
HIV TAT protein and Drosophila Antennapedia transcription factor shown to translocate cell membranes
Many CPPs discovered to date - potential for delivery of a variety of cargoes (including liposomes, nanoparticles, etc for biological entities and traditional drugs)
Low cytotoxicity, dose-dependent, can be conjugated to carriers
Tend to be arginine rich (e.g. TAT (48-60) = GRKKRRQRRRPPQ)
Various uptake mechanisms, including direct translocation and the different types of endocytosis
Particularly promising for oligonucleotides - packaging and delivery in one
Potential for Non-viral vectors: what characteristics do we want ideally?
Small
Stable in serum
Low toxicity
Specific cell targeting
Cytosolic release of cargo
Nuclear targeting (depending on cargo)
High transfection efficiency
Anti-angiogenesis strategies
Anti-sense RNA or siRNA to block translation of GF or its receptor
Oligonucleotides to block transcription of GF or its receptor
Gene therapy to induce production of soluble receptor or mAb (against receptor of GF)
mop up existing GF and stops it binding to receptors on cells that drive angiogenesis
Glioblastoma tissue
VEGF is highly up-regulated in GBM tissue and levels correlate to disease progression
Gene for soluble VEGF receptor delivered in murine glioma model - mops up VEGF and reduces angiogenesis
RGD
tripeptide
targets intergrin receptors
integrin receptors upregulated in cancers
RGD binds to them in cancer cells and aids in targeting them to tumour cells
Haematopoietic Stem Cell Transplantation (HSCT)
For peripheral blood cells:
Pre-treatment with GCSF prior to collection
G-PBMNCs collected
Cell-based immunotherapy
Allogeneic T cells (from same donor) transfected with HSV-TK – Zalmoxis approved in 08/16
no longer authorised
Engrafts and helps reconstitute the immune system alongside HSCT from same donor, improves overall survival and non-relapse mortality
Ganciclovir administered if graft versus host disease develops
when too much t-cell produced
ganciclovir kills them off
immunotherapy in cancer
Cytotoxic T cells (Tc cells) are activated by antigen presenting cells such as dendritic cells, which prime them with specific antigens for target cells (e.g. infected cells, cancers)
presented on surface via MHC
Tc cells then recognize and kill cells which express these antigens via release of perforin and granzyme, which induce apoptosis
The efficacy of this process can be massively improved using cell engineering approaches
APCs
Antigen Presenting Cell
used to train t-cells to respond to the cancerous antigen
Artificial APCs are currently an emerging approach – decorate cells or microparticles with antigens in order to prime T cells
“Off the shelf” - does not require harvesting from the patient
CAR T-Cells
T cells genetically engineered to express a chimeric antigen receptor (CAR)
CAR T cells proliferate and kill tumour cells upon contact with the antigen they recognize
Kymriah
first ever FDA approved CAR-T cell therapy
for b-cell lymphocytic leukemia in children and young adults
recognises CD19
IMLYGIC
HSV-1 engineered to be oncolytic
kills cancer and doesn’t harm healthy cells
it attracts cells of immune system to help fight the leukemia
Mesenchymal Stem Cell Therapy
Ability to home to site of injury
Anti-inflammatory properties
Hypoimmunogenic - no immune response so dont need to match to donor
All makes them suitable as therapeutic cells
Pro-tumorgenic effects of MSC
promote tumor cell ggrowth invasion and metastasis and support tumour fbrovascular network and promote angionesis
support the tumor-induced vascular network
MSC in cancer patients
if delivered into patients with cancer it will hone and embed into cancer cell and produce trail ligand
trail ligand will react with trail receptor on cells and induce apoptosis
Anticancer effects of MSCs
anti-angiogenesis
cytokine delivery
to deliver miRNA
vehicle for anti-cancer drugs
CRISPR/Cas9
Clustered Regularly Interspaced Short Palindromic Repeats
CRISPR-associated (Cas) genes
Targeted CRISPR/Cas9 treatment
head and neck cancer due to overexpression of SOX2 gene in cancer stem cells which promototes growth and resistance to apoptosis
specifically knocking down SOX2 in tumour cells using CRISPR/Cas9 should cause cancer regression
found a 90% inhibition of growth, 90% increase in survival and tumour disappeared in 50% of mice
dCas9
a mutated cas9 which deactivates nuclease activity and enables it to be used to control gene expression
guide RNAs localise dCas9 to specific genes where transcription is activated or repressed
CRISPR-Based cell therapies
trials with engineered t-cells with `
CRISPR knocks out endogenous T-cell receptor (stops knocking out what it’s linked well) and PD-1 (stops cancer cell from inhibiting t-cells)
therapy was well-tolerated
but only 10% of t-cells in trial has all of the genetic modifications needed
Using genome editing to be able to target drug delivery
looked at cancer patients and looked at the 1000+ variations of p53
tested variations to see if specific mutations caused cancer to see if there are any biomarkers for cancer or any specific druggable targets
Advantages of gene editing
Small (potentially easier to deliver), inexpensive, specific, versatile
Developments in base and prime editing offer more control
Disadvantages of gene editing
Off-target effects - unintentional editing similar genomic regions to the target, possibly oncogenic
Autoimmune risks - possible autoimmune responses or unintended effects on healthy tissues
Efficient delivery to all target cells or a specific subsets remains challenging
Ethical Concerns
Limited simultaneous changes - CRISPR currently allows limited simultaneous changes per cell
HDR to NHEJ ratio - the balance between the two affects editing efficiency
Abuse, terrorism - people injecting themselves with CRISPR/Cas9