Oncolytic Viruses II - Potency Flashcards

1
Q

how does changing serotypes improve virus replication in cancer cells/how is this done? explain with adenovirus example

A

Directed evolution: passage on HT-29 –> selected “ColoAd1” = Ad3/Ad11p chimeric virus –> uses CD46 R which is higher in cancers that the Ad5 CAR receptor which is low on cancers

Ad3 and Ad11p have reduced seroprevelance –> less preexisiting immunity

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

how was Ad5 adapted towards cancer through mutations?

A

PCR Ad5 on “mutator cells” which express error-prone pol. –> mutants –> select best mutant on human cancer cell line (largest plaque = better virus/highly prolific)

best mutatns changed splicing ratio –> increased expression of ADP (adenovirus death protein) which is a transmembrane protein that induces nuclear membrane instability –> cell lysis (proinflammatory) and virus release

decreased 6.7K = inhibitor of extrinisc cell death/induced apoptosis
decreased 19K = inhibitor of MHC peptide loading

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

which mutation in virus could promote replication/killing/spread in cancer cells?

A

SV5 (mutant) binds less to cells –> spreads a farther distance before re-infection –> infects more layers than WT

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

why would different binding efficiencies favour different niches?

A

in gut: bind or be eliminated – fast flow, few input viruses, lots of immune cells –> binding efficiency must be strong here

in tumor: bind too strong and never move – limited flow (dense and high pressure), lots of input viruses, less immune cells –> efficient binding restrcits distance of spread

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

what are two ways to increase immunity against tumor Ags?

A
  1. add stimulatory genes
  2. delete immunomodulatory genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

how does HSV ICP47 block MHC1-peptide loading?

A
  • bind the transporters associated with antigen processing (TAP)
  • blocks loading of peptides onto MHC-1 molecules in the ER
  • infected cells are masked for immune recognition of cytotoxic T cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

what do the viroreceptors and virokines (immunomodulatory genes ) in poxviruses do?

A
  • viroreceptors: solube or cell-surface decoys that bind host-cell cytokines or chemokines –> block: IFN signaling, cytokine activity, complement
  • virokines: secreted agonistic or antagonisitic ligands for host cellular receptors –> activates EGRF/Ras signaling
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

what are 6 general ways for resisting neutralization by Abs or complement?

A
  1. high dose – admin so much virus –> takes a while for body to catch up
  2. immunosuppressive drugs (downside: susceptible to other pathogens + need IS to clear cancer cells)
  3. specific inhibitors
  4. genetic – change epitopes
  5. biochemical virus shielding
  6. change oncolytic virus at every treatment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

what components of human serum inactivate oncolytic VV? what does heating serum do?

A

complements and Abs

heating serum denatures complement

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

does inhibiting complement promote oncolysis by VV? how can we inhibit complement, why is this not the best solution?

A

CVF = cobra venom factor – C3 complement depleting –> cobras are an endagered species

when complement is reduced, virus reduced tumor size –> once intitial virus gets to tumor, it can subdue complement because it expresses its own complement inhibitor

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

biochemical targeting was not ideal for an oncolytic virus, why is biochemical shielding a good approach?

A

helps hide the virus so it can get to its target

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

what are 3 challenges for changing OV at every treatment?

A
  1. overwhelming the IS, forgets about the tumor?
  2. politics, patent your virus
  3. can only use the one’s that are approved, takes a while to develop
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

what effect do factors in breast tumor environments have on reovirus?

A

when mouse breast tunor extracellular extract (tumor juice) is combined with reovirus in a mouse –> smaller viral titre than virus alone

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

how do breast cancer factor inactivate reovirus?

A

breast tumor metalloproteases cleave the attatchment protein of reovirus: cleaves sigma1’s C-term head off –> tail (sialic acid) –> can’t bind to cells

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

can we overcome the cleavage of sigma1 and inactivation by breast cancer proteases in reovirus?

A

point mutation to remove proteolysis-sensitive site –> no cleavage by proteases –> reovirus can bind to cells

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

does overcoming sigma1 cleavage help oncolysis of reovirus?

A

single point mutated reovirus delayes tumor growth (takes longer to reach same volume as controls)

17
Q

what is the difference between additive, antagonistics, and synergistic effects?

A

addivitive: not helping each other but doing their own thing
antagonisitic: worse off than either therapy alone (common)
synergistic: working together and having an effect

18
Q

what can we combine OVs with in a combination therapy? what can these do to a tumor?

A
  • OV + radiotherapy
  • OV + HDACi
  • OV + chemotherapy
  • OV + immunotherapy

tumor regression. inhibition of tumor associated angiogenesis, apoptosis, tumor cell lysis

19
Q

what is a pro-drug converting enzyme – OV combination?

A

viral vector which encodes an enzyme infects tumor cell –> prodrug is added and converted by the enzyme to a toxin –> toxin spreads to neighbors

localize a toxin

20
Q

give an example of a prodrug converting enzyme OV combo

A

inject gene vector containing e. coli PNP into tumor cells –> cells produce PNP protein –> inject fludarabine (IV) –> PNP converts fludarbine into cytotoxic fluoroadenine –> fluroadenine can diffuse into neighboring cells that do not express e. coli PNP

21
Q

what are major advantages on OV therapy?

A
  1. self-replilcating/self-dosing
  2. can target changes in networks.hubs rather than individual-specific changes in cancer = less toxic than chemotherapy but more broad-reaching than directed therapies
  3. genetically modifiable – increase potency, increase specificity, can combine mutliple mechanisms
  4. promote anti-tumor immunity – clearance and protection
22
Q
A
23
Q

what is a major disadvantage of OV therapy?

A

clearance by IS