Antiviral Agents / Virotherapy Flashcards

1
Q

While antiviral ______ are the classical approaches, antiviral _________ is an upcoming field of research and development for the treatment of infectious diseases.

A

Drugs; Immunotherapy

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

For which viral infections do we rely on antiviral drugs?

A

For those that cannot be effectively controlled via public health measures and vaccines.

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

(T/F) To date, most antiviral drugs are targeted against HIV and herpesvirus due to their clinical significance.

A

True!

Millions of lives have been saved by the use of antiviral drugs!

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

What are the three major limitations in antiviral drug development?

A

1) Requirement for a high degree of SAFETY (must ensure compound does not have deleterious effects on host cell)

2) Requirement for POTENCY (modest viral growth in the presence of inhibitor is NOT good enough; allows for mutants)

3) Some viruses difficult to PROPAGATE IN VITRO in the lab or there may be a LACK of animal models to test safety + potency

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

What does R and D mean for R&D of antiviral drugs?

A

R = Research and compound identification (only the beginning of the process of producing a drug)

D = Development, compromises all steps necessary to take an antiviral lead compound through safety testing, scale-up of synthesis, formulation, pharmacokinetic studies, and clinical trials

Takes 5-15 years!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Briefly describe the descending staircase of antiviral drug discovery.

A

First you have thousands of compounds synthesized or purified from the library.

Then you have preclinical testing where you test potency and toxicity in cells and then animals.

Finally, you have clinical testing where you test toxicity and potency in humans.

In each step, the numbers of promising candidates are decreasing, resulting in a descending staircase.

It takes several years before a final product that is safe and effective is produced!

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

Clinical trials occur in a series of phases. Match the phases to their definitions.

1) Phase I
2) Phase II
3) Phase III
4) Phase IV

A) focus on EFFECTIVENESS, slightly large groups (<100), still continue to monitor safety

B) once the drug is approved; further evaluation of EFFECTIVENESS and LONG TERM SAFETY, groups of hundreds to thousands

C) SAFETY assessment in small groups (20-80), early evidence of efficacy

D) focus on EFFECTIVENESS, SIDE EFFECTS compared to standard therapy, larger groups (100-thousands) who are randomly assigned to “trail arms”

A

Phase I: SAFETY assessment in small groups (20-80), early evidence of efficacy

Phase II: focus on EFFECTIVENESS, slightly large groups (<100), still continue to monitor safety

Phase III: focus on EFFECTIVENESS, SIDE EFFECTS compared to standard therapy, larger groups (100-thousands) who are randomly assigned to “trial arms”

Phase IV: once the drug is approved; further evaluation of EFFECTIVENESS and LONG TERM SAFETY, groups of hundreds to thousands

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

True or False

1) It takes around 10-15 years to discover a drug. We can fast track certain drugs using phase I-II (safety + efficacy in the same phase).

2) Pharmaceutical/biotech companies pay for clinical trials in collaboration with clinical investigators.

3) Phase I and Phase II take around 1 year altogether.

A

1) True

2) True

3) False; 2-4 years

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

How are antiviral compounds discovered?

A

Knowledge of viral life cycle IDENTIFIES TARGET for antiviral drug discovery:

The life cycle of many viruses are known, allowing for identification of several targets for intervention.

Viral genes essential for reproduction can be cloned and expressed in genetically tractable organisms and their products can be purified and analyzed in molecular and anatomical detail - INHIBITORS OF CRITICAL PROCESSES CAN BE IDENITFIED

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

What are some of the challenges/things to consider after a drug target is identified?

A

Must determine if compound:

1) will get to the right place at the right concentration (bioavailability)

2) will persist in the body long enough to be effective (pharmacokinetics)

3) will be tolerated or toxic

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

Differentiate mechanism-based screens with cell-based screens.

A

Mechanism-based screens: seeks to identify compounds that affect the function of a known viral target such as enzymes, transcriptional activators, cell surface receptors, ion channels. often screening is conducted with purified protein in formats that facilitate automated assays of many samples.

Cell-based screens: elements of the mechanism to be inhibited are engineered into an appropriate cell system. provides information not only about target inhibition, but also cytotoxicity & specificity. the use of multiple reporter molecules may also allow for detection for more than one event at a time.

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

Briefly describe a mechanism based screen for inhibitors of a viral protease.

A

A FLUOROGENIC MOLECULE is covalently linked to the N-terminus of the substrate (peptide) of the protease and the C-terminus is linked to a bead.

When the protease cleaves the peptide, the fluorogenic N-terminus is released into the soluble fraction which is separated from the insoluble beads containing C-terminus fragment.

Protease activity is assayed by the appearance of soluble fluorescent peptide.

Fluorescence activity if enzyme is NOT inhibited, while there will no fluorescence activity when enzyme is inhibited.

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

Briefly describe a cell based screen for inhibitors of a viral protease.

A

Tetracycline-resistant bacteria have tetracycline efflux protein on the membrane with a HIV protease site added.

When there is expression of a (HIV) protease, there is inactivation of the tetracycline efflux protein.

Inactivation causes the bacteria to be tetracycline sensitive! It leads to no bacterial growth on tetracycline-containing media.

A functional inhibitor should result in antibiotic resistant bacteria as the protease site would not be cleaved!

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

(T/F) Cell based screens can be done in eukaryotic cells. However, bacterial cells are faster and cheaper!

A

True!

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

Advances in protein separation techniques, _____ _______ and bioinformatics allow to determine the total protein repertoire aka ________ of virus samples.

We are now able to determine protein interactions of _____ proteins with _____ proteins, identifying critical nodes or _________ ____.

A

Mass spectrometry; proteome

viral; host; INTERACTION HUBS

*an example of this is the systematic survey of the interaction between cellular and viral proteins of HIV I. these interactions provide clues of potential targets for drugs!

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

(T/F) After administration, the drug must reach the correct site in the body and must remain at an effective concentration to allow for the inhibition of viral reproduction.

A

True!

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

How can we modify compounds to improve absorption? Give an example.

A

Add side chains!

ACYCLOVIR is not effectively taken up after oral ingestion. But the derivative VALACYCLOVIR has 5x oral bioavailability through the addition of a side group, which allows increased passage of drug from the intestines into the bloodstream..

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

Match the following drugs to their definitions:

1) Acyclovir (Zovirax)

2) AZT (Retrovir)

3) Amantadine (Symmetrel)

4) Tamiflu

A) Targets INFLUENZA A virus M2 protein which forms a tetramer that creates a transmembrane ion channel to transport protons. By binding to the outside of the tetramer, it blocks the entry of protons.

B) Specific non-toxic drug - effective against HERPES virus. Nucleoside analog related to guanosine. Mediates chain termination of DNA synthesis.

C) Blocks the function of Neuraminidase, an INFLUENZA enzyme that cleaves sialic acid group from glycoproteins to release virion from host cell.

D) First drug licensed for the treatment of AIDS. Analog of thymidine. Mediates chain termination of DNA synthesis.

A

Acyclovir: Specific non-toxic drug - effective against HERPES virus. Nucleoside analog related to guanosine. Mediates chain termination of DNA synthesis.

AZT: First drug licensed for the treatment of AIDS. Analog of thymidine. Mediates chain termination of DNA synthesis.

Amantadine: Targets INFLUENZA A virus M2 protein which forms a tetramer that creates a transmembrane ion channel to transport protons. By binding to the outside of the tetramer, it blocks the entry of protons.

Tamiflu: Blocks the function of Neuraminidase, an INFLUENZA enzyme that cleaves sialic acid group from glycoproteins to release virion from host cell.

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

Two major success stories of antiviral drug development: ______ and _________.

A

HIV; Hepatitis C

*Steps in the reproduction of HIV and HCV are targeted by antiviral drugs.

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

What is immunotherapy?

A

Therapeutic strategies to stimulate or restore the ability of the patient’s immune system to fight disease (mimick immune system). These therapies normally employ biological products, known as BIOLOGICAL RESPONSE MODIFIERS.

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

What are the different kinds of immunotherapies?

A

Cytokines (interferon-α) and antibodies (super-antibodies) which are PASSIVE immunotherapy.

Lymphocytes (T cells) which are ACTIVE immunotherapy.

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

(T/F) When cells get infected with pathogens, they will secrete signalling molecules called CYTOKINES to neighbouring cells to notify them of infection, and also to train and boost the immune system.

A

True!

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

The cytokine, interferon-α, was the first approved therapy for _________ virus and _______ virus.

The effects on _______ infection are potent, while on ________ are modest.

A

Hepatitis B; Hepatitis C

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Immunomodulatory agents, such as ___________, which can stimulate the T cell responses or ________ which stimulates Natural Killer (NK) cells, are being investigated for their ability to reduce _____ ____ and/or moderate _______ of persistent infections (HPV/HIV-1)

A

Interferon-α; Interleukin-12; viral load; complications

25
Q

What are the limitations to cytokine therapy?

A

1) Biological activity is generally short (not very stable)
2) High side effects (flu-like symptoms, bone marrow suppression, autoimmune disease)
3) Expensive

26
Q

1) What are monoclonal antibodies?

2) How long have they been in use? Are they common?

3) What are their limitations?

A

1) Monoclonal antibodies are laboratory produced molecules engineered to serve as “substitute” antibodies. They are generated by B-cell clones. They RECOGNIZE and NEUTRALIZE antigens.

2) They have been used for more than 1000 years for the treatment + prevention of infectious diseases but a single anti-viral antibody, the Respiratory syncytial virus (RSV) specific antibody is in widespread clinical use.

3) High cost; Difficulties in administration; Belief that antibodies are effective only in prophylactic (preventive) settings - which are already achieved by the cheaper vaccines.

27
Q

What are super-antibodies?

A

Super-antibodies are a new generation of highly potent and/or broadly cross-reactive human monoclonal antibodies.

They are typically generated infrequently and/or in a limited number of individuals during natural infections.

Isolation of these has been achieved by large-scale screening for suitable donors and new single B cell approaches to human monoclonal antibody generation.

28
Q

What are the potential advantages of super antibodies?

A

1) Higher potency (decreased cost, low treatment dosages, different administration routes, extended half-life)

2) Higher cross-reactivity (could target multiple viruses of a given family with a single antibody)

29
Q

What are the current approaches for Viral-specific T cells antiviral immunotherapy?

A

1) Banking T cells from donors who have already experienced certain viral infections

2) Training or genetically engineering virus-naive T cells taken from unexposed tissues. These approaches could make antiviral cellular immunotherapy more robust and more widely used.

30
Q

(T/F) The clinical data looks promising to use donor T cells as antiviral therapies for bone marrow transplant patients who are vulnerable to infections from common viruses.

A

True!

31
Q

What are the limitations of T-cell immunotherapy?

A

1) Very expensive
2) Not possible for a massive outbreak

32
Q

What is virotherapy? What are the two arms of virotherapy?

A

Virotherapy is a treatment using biotechnology to convert viruses into therapeutic agents by selecting or reprogramming viruses to treat diseases.

The two arms of virotherapy:
1) Viral vectors for gene therapy
2) Cancer-killing (oncolytic) viruses

33
Q

What is gene therapy?

A

Gene therapy is the process of removing, modifying, or replacing defective genes with healthy ones.

Though it has been around 45 years, it is still being developed as treatments are rarely making past the clinical trial stages.

34
Q

Recombinant, ___________ _____ vectors were the first molecular tool enabling efficient, nontoxic gene transfer into human somatic cells.

__________ and _________ virus have shown the most clinical promise and are the carriers (vectors) of several recently approved gene therapies.

A

Replication-defective viral vectors

Retroviruses; Adeno-associated virus (AAV)

*we can’t use just any virus for gene therapy! we have to select certain viruses.

35
Q

Gene therapy using an adenovirus vector can be used to treat or cure certain genetic diseases in which a patient has a defective gene.

What are the mechanisms of gene therapy?

A

1) Gene insertion: a new version of gene is introduced into the treated cell

2) Gene modification: an exciting gene is modified

3) Gene surgery/removal: a particular gene is excised and may also be replaced by its healthy counterpart

*modify viral vector to express healthy gene of interest
*virions infect cells + deliver the gene

36
Q

Adeno-associated viral vectors are engineered from a ________, _______ parvovirus that is naturally replication-defective. This means wildtype AAV requires another virus such as _______ or a ________ to replicate.

All viral coding sequences of AAVs are ______ with a gene expression cassette of interest.

A

Non-pathogenic; non-enveloped; adenovirus; herpesvirus

Replaced

37
Q

What are some limitations of AAV vectors?

A

1) They can not package more than ~5 kb of DNA.

2) They are predominantly NON-INTEGRATING. This lessens risks related to integration but also LIMITS long-term expression from AAV vectors to long-lived post-mitotic cells.

38
Q

Early AAV trials established safety but were limited by insufficient dosing, and anti-AAV immune responses, most likely because many people carry neutralizing antibodies and memory T cells directed against the AAV capsid.

How was this overcome?

A

1) Increase dosage

2) Use recombinant vectors that allow them to be not sensitive to the neutralizing vectors

39
Q

What are γ-retroviral vectors?

A

These vectors were used in 1st generation clinical trials to DELIVER A NORMAL COPY of a specific defective gene into the genome of T cells or HSCs from patients with immunodeficiency or blood cancers.

40
Q

What were the second generation of retroviral vectors? How are they different from the first generation?

A

LENTIVIRUSES and SPUMAVIRUSES were the second generations of retroviral vectors.

In contrast to γ-retroviral vectors, lentiviral vectors enabled gene transfer into NON-DIVIDING cells but still left quiescent G0 cells out of reach.

Lentiviral vectors also can carry larger and more complex gene cassettes than γ-retroviral vectors and thus development provided a critical advance for HEMOGLOBINOPATHIES.

41
Q

Briefly answer the following questions regarding retroviral vectors.

1) Which vectors are currently the tools of choice for most HSC applications?

2) What is another approach that decreases the risk of genotoxicity?

A

1) Lentiviral vectors

2) Removal of endogenous strong enhancer from lentiviral and γ-retroviral vectors is another approach that decreases the risk of genotoxicity.

42
Q

What must happen to a virus before being used to carry therapeutic genes into human cells?

A

It is modified to remove its ability to cause infectious diseases (disease-causing gene is excised).

43
Q

Briefly answer the following questions regarding AAVs:

1) What technique is used to produce recombinant adeno-associated virus (rAAV) vectors?

2) What is one of the central challenges of rAAV production?

A

1) rAAV vectors are produced using the triple transfection technique.

2) One of the central challenges of rAAV production is the need for a “HELPER VIRUS” to facilitate replication within a host mammalian cell.

The combined activity of the adenovirus-derived genes on the helper plasmid with the rAAV genes on the second plasmid proved sufficient to generate functional rAAV particles containing the transgene from the third plasmid.

44
Q

Gene therapy can be used to modify cells inside or outside the body.

Differentiate between ex vivo (cell-based delivery) and in vivo (direct delivery).

A

Cell-based delivery: treatment gene is added to a harmless lentivirus or retrovirus vector which is introduced to the isolated stem cells of patient. The stem cells containing the treatment gene are returned to the patient.

Direct delivery: Doctor injects the vector carrying the gene directly into the part of the body that has defective cells.

45
Q

(T/F) Tumours are composed only of cancer cells.

A

False!

Tumour microenvironment is very heterogeneous.

Tumours are more than just cancer cells. They include tumour stroma which is made of non-cancer cells such as endothelial cells, immune cells, stem cells, etc that allow the cancer cells to grow.

46
Q

What are the three types of anti-cancer therapies? Briefly describe them.

A

1) Chemotherapy (drugs that affect cells THAT ARE DOUBLING, not specific, and highly cytotoxic)

2) Cancer targeted therapies (agents that inhibit or attack a more specific target in cancer cells. attempt to take advantage of genetic changes in malignant cells. ex: small molecules, antibodies, anti-angiogenic drugs)

3) Oncolytic viruses (exploit tumour cell-specific genetic defects)

47
Q

What is oncolytic virus therapy based on?

A

Oncolytic virus therapy is based on selective replication of viruses in cancer cells and their subsequent spread within a tumour without causing damage to normal tissue.

They represent a unique class of cancer therapeutics with distinct mechanisms of action. The activity of oncolytic viruses is very much a reflection of the underlying biology of the viruses from which they are derived.

48
Q

(T/F) The same biological processes that control cell growth and death also control the ability of individual cells to fight virus infections!

A

True!

49
Q

Interferon production:

A

1) locally prevents further infection

2) prevents unrestricted cell growth

3) induces apoptosis

4) prevents angiogenesis

5) activates adaptive immunity

*not compatible with replication of cancer cells

50
Q

________ cells have an intact interferon response, therefore virus replication is prevented.

________ cells have impaired interferon production (no production), therefore virus replication is able to proceed.

A

Healthy; Cancer

*healthy cells fight virus infections by producing interferons (cytokines)

*interferons expressed at low levels without viral infections

51
Q

(T/F) Oncolytic viruses infect cancer cells only not healthy because they impair interferon production, which is desired in cancer cells.

A

True!

52
Q

(T/F) The biological processes underlying anti-viral defense are incompatible with efficient tumor evolution. Cancer driver mutations are oncolytic virus driver mutations.

A

True!

53
Q

What are the two classes of oncolytic viruses?

A

1) Viruses that naturally replicate preferentially in cancer cells and are non-pathogenic in humans often due to elevated sensitivity to innate antiviral signalling (can’t defend against interferons) or dependence on oncogenic signalling pathways.

2) Viruses that are genetically manipulated for use as vaccine vectors and/or those genetically engineered with mutations/deletions in genes required for replication in normal, but not cancer cells.

54
Q

What are the features of oncolytic viruses (OVs) that make them advantageous and distinct from current therapeutic modalities (resulting in a high therapeutic index)?

A

1) There is a low probability for the generation of resistance, as OVs often target multiple oncogenic pathways and employ multiple means for cytotoxicity

2) They replicate in a tumour-selective fashion and are non-pathogenic, only minimal systemic toxicity has been detected

3) Virus dose in the tumour increases with time due to in situ virus amplification, as opposed to classical drug pharmacokinetics that decreases with time

4) Safety features can be built in, such as drug and immune sensitivity

55
Q

Oncolytic viruses have been engineered or selected to initiate a MULTI-MODAL cancer attack that uses complementary mechanisms to destroy the tumour.

What are the three mechanisms?

A

1) Direct virus-mediated cell lysis (infect cancer cells only which die eventually)

2) Anti-tumour immunity (cancer + viral antigens released when cell dies; “cancer vaccines”)

3) Disruption of tumour vasculature (viruses infect endothelial cells in blood vessels that are present in the tumour; they shut down blood vessels; no nutrient + oxygen 4 tumour)

56
Q

_______ virus can be genetically modified to deliver therapeutic payloads against tumours. It has an excellent safety record in humans.

A

Vaccinia

57
Q

How does a systemically delivered oncolytic virus enter the tumour bed?

A

Tumors have leaky blood vessels!

58
Q

(T/F) Anti-tumor immunity is just as important of a mechanism as cell lysis of oncolytic viruses.

A

True!

59
Q

1) What happens if a mouse whose colon tumour was treated with an oncolytic virus was challenged with colon cancer cells?

2) What are athymic mice?

A

1) (Immune competent) mice will reject the colon tumours! T cell-based immunity leads to long-term immune surveillance and anti-tumour immune responses.

2) Athymic mice are strains that have genetic mutations that cause a deteriorated or absent thymus, resulting in a compromised immune system due to a greatly reduced number of cells.