Final Flashcards

1
Q

When does the FDA regulate a therapy as a drug?

A

The only two approved cell-based therapies are hematopoietic stem cell transplantation and CAR-T (for blood and immune system).

FDA regulates when there was more than minimal manipulation of the cells (can’t do anything to the cell that you would do in a lab).

In order to not be regulated, all of these must be true: Minimally manipulated, homologous use, no excipients or stabilizers or anything, any systemic effect?

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

What are the different types of cell-based therapies and what are the differences between them?

A

Stem cells & stem cell-derived products: Don’t involve immune cells. These cells can be differentiated into any type of cell.
- Utilize the property of stem cells (differentiation).

Immunotherapies: Involves immune cells and activation of the immune system.

  • Utilizes already mature cells in the body. We use these cells and infuse them back into the patient.
  • Adoptive cell transfers, cancer vaccines, etc.
  • Not just T cells, there’s also dendritic cells and NK cells.

Cell-based therapy is just cell therapy when it does not involve the immune system (such as stem cell tissue regeneration). Cell-based therapy is immunotherapy when it utilizes immune cells (such as CAR-T).

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

What are the basic components of a CAR-T therapy?

A
  • Force immune cells to find the genes and stick to them. There are FDA approved drugs. They are very efficacious.

Chimeric Antigen receptor + T cell (or NK, macrophages, or dendritic cells)

Cells are removed, genetically engineered (inject cells with gene), expanded, and delivered back to the patient.

CARs are engineered molecules that are expressed on the surface of immune cells that can recognize specific proteins on the target cells (TSAs and TSAs) and then deliver an activation signal to the cells.

Includes antigen receptor (cancer cell) and co-stimulatory domains that send activation signal to cause cell death.

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

What is the difference between autologous and allogeneic cell-based therapy?

A

Autologous - The patient is their own donor. They can only donate to themselves. This is good for rejection. It’s harder to source because it’s hard to rely on your own amount of cells/immune system.

Allogenic - Anyone can donate to anyone (future of stem cell therapy?). Easier to source. Can induce an immune response/rejection because our bloods don’t always match.

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

What are some problems that may arise when developing drugs that target a single tumor antigen?

A

The cancer cell is smart. It may down-regulate that antigen that it knows the drug is targeting, leaving that drug pretty useless. For this reason, we want to make drugs that have multiple antigens as the target.

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

What are the main safety issues that are associated with cell-based gene therapies?

A

CAR failure - It just doesn’t work. Ex. can’t harvest enough T cells, CAR T cells don’t expand, limited CAR T cell persistence in vivo

Antigen modulation - Antigen loss or down-modulation. Every cancer pt with brain/breast cancer, the cancer expresses different protein (it escapes the CAR T cell target). This is why we may use two different antigens as targets.

CAR toxicities - Ex. Neurotoxicity

Unmet needs - Doesn’t work that well

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

What are two reasons why cell-based therapies have been more successful (and approved by the FDA) for blood cancers, but not solid tumors?

A

Solid tumors - They are heterogeneous, they have immunosuppressive factors, the immune cell infiltration is low, and the cells in solid tumors are dysfunctional.
- We try to target cytokine networks, immunosuppresive populations, immune checkpoints, and tumor stroma to get around this. (Anything that supports the tumor).

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

What is one way to clinically mitigate or manage cytokine release syndrome?

A

Antibodies against toxins and cytokines - block IL-6 (ex. tocilizumab). ***

Avoid steroids (they may impact CAR-T cells)

Use cells that induce no or limited CRS (NK cells)

Hemofiltrition to filter out cytokines

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

What is the drug substance and the drug product in cell-based therapy?

A

Drug substance: engineered molecular component (ex. chimeric antigen receptor or gene)

Drug product: Immune cell + gene (ex. CAR-T/NK cell)

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

What is ADCC? What are multi-specific engagers?

A

ADCC - antibody-dependent cell-mediated cytotoxicity. When a pt gets Ab therapy, the Ab will not kill the cancer cell, it’ll just block it. The killing is done by the immune system. The Ab interacts with CD16 on NK cells to stimulate the killing.

Multi-specific engagers - Bridge the gap between the cancer cell and the T cell for killing.

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

What is an on-target, off-tumor effect?

A

On-target, off-tumor: Cell recognizes the right protein on the wrong place. Ex. CD19 on B cells. This can result in severe toxicities, because you’re killing off healthy cells.

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

What are some common problems with targeting tumor antigens with cell-based immunotherapies?

A

Tumor evasion - The shutting down of proteins on the cancer cell that are trying to be targeted. This results in resistance to treatment.

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

What are the potential risks of cell-based therapies?

A

Neurotoxicity - Due to immune cell activity, can be fatal

Cytokine release syndrome (CRS) and GvHD - CRS is a systemic inflammatory response (aka cytokine storm) that results in pro-inflammatory cytokines attacking the pts immune system. GvHD is when the donor tissue recognizes the exogenous cells as foreign, and then starts attacking them.

Toxicity associated with viral genomic DNA - Need to make sure the virus is completely dead.

Cells that persist for an extended period - Could increase or prolong adverse reactions. We try to mitigate this with suicide switches.

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

Is the viral engineering method of cell based therapy permanent or transient?

A

Permanent. This is the most common type of cell engineering.

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

What is lymphodepletion and what drugs are commonly used for it?

A

Lymphodepletion - Donor matching and pre-treatment of patients. This is needed in all cases. It eliminates immune system so that it has no ability to reject the drug.

Cyclophosphamide & Fludarabine - Take these once a day for 7 days.

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

What are the targets of the first two FDA-approved CAR-T cell therapies?

A

Large B-Cell Lymphoma??

Blood cancers

17
Q

What is the role of the extracellular region of CAR?

A

It binds to the antigen on the tumor cell.

18
Q

What agent is most commonly used for cryopreservation of immune cell therapy products?

A

Cryopreservation is the process of freezing the cell therapy drug product. These minimize damage as the cells are frozen.

We use DMSO most commonly. This stops the water loss from the cell.

If you freeze cell too slowly, you’ll over dehydrate it. Too fast, and it turns into ice. You have to find the middle ground.

DMSO is often washed off before infusion into patient.

19
Q

What’s the difference between native cell therapies and cell-based gene therapies?

A

Native - Unmodified immune cells (ex. TILs, Hematopoietic stem cell transplants)

Cell-based - Gene-modified cells (CAR-T/NK, TCR-T cell therapies)

20
Q

What is the gut microbiota and microbiome?

A

Gut microbiota - Collective term for microorganisms in the human gut (bacteria, fungi, virus).

Gut microbiome - The collection of genetic information that is encoded within all genomes present in the microbiota.

21
Q

What are some characteristics of human gut microbiota?

A

There are 10^13-14 microbial cells in the intestinal tract. This is more than the amount of human cells there are in the body.

The largest amount of microbes can be found in the lower GI tract. These are anaerobic microbes. As food goes through the GI tract, the environment gets more and more anaerobic. The food near the lower GI tract cannot be metabolized by human enzymes and they are now nutrients for the microbes.

22
Q

What are the roles of the gut microbiota in physiology and pathology?

A

Physiology - The microbiota has immune, GI function, metabolic and digestive function. They also effect most of the major organs and tissues.

Pathology -

  • Obesity: The obesity phenotype is transferred by the gut microbiota. Essentially the gut microbiota plays a role in developing obesity.
  • CV disease: High levels of TMAO in the blood is associated with an increase in major adverse CV and liver disease. It is found that the microbiota can produce this.
  • C. diff: Gut microbiota provides colonization resistance to C. diff. If you have a healthy gut microbiota, you won’t get the C. diff infection, but if you get rid of your normal gut microbiota, you’ll probably get the infection.
23
Q

How can the gut microbiota be manipulated to improve health? Ex. Fecal microbiota transplant (FMT), Probiotics, small molecules

A

Fecal microbiota transplant - Give whole collection of microbes to the gut (restore gut microbiota). This is the gold standard for C. diff infection.

Probiotics - These are specific live microorganisms that help the host when taken. The underlying mechanisms are unclear, but there are some health benefits (ex. maintain good digestion, improve neurotransmitter balance, etc). However, these benefits are stain specific, so we don’t really know what benefits we are getting.

Chemical inhibitors - They can inhibit the more harmful metabolites that are made from the gut microbiota.

24
Q

What is the impact of the gut microbiota on drug metabolism and toxicity?

A

The gut microbiota can metabolize certain drugs. Ex. Etoposide is a low solubility and low permeability drug that has relatively low systemic exposure. However, with antibiotic pre-treatment, the concentration of Etoposide in the blood goes up.

Antibiotics can be used for pre-treatment to get rid of this microbiota, so the drug doesn’t get metabolized as much anymore, resulting is better systemic drug exposure.

25
Q

Why are nanoparticles considered for drug delivery?

A

Nanoparticles are small enough that they can be modified to enter cells easily (packaging the small cells makes them selective for the type of cell we want them to enter, ex. cancer cells). They can be injected IV, they can be used with cellular uptake (pinocytosis, phagocytosis, etc.), extravasation, adsorption of ligands, etc. They also have a unique biodistribution that is different than free drug distribution.

They have the potential to modify biodistribution of a drug and increase drug delivery to target tissues through IV injection.

26
Q

What are the different nanoparticulate drug delivery systems, what are they made of, how are they prepared?

A

Liposomes - The phospholipids will automatically arrange into this liposome, and the drug and go inside of it. Cholesterol may be used to stabilize the liposome.

Protein nanoparticles - Albumin is a natural carrier of hydrophobic drugs. Albumin interacts with cell receptors, so it can actually help deliver the drug to its target (ex. SPARC, gp60 receptor).

27
Q

What is Doxil, Taxol, and Abraxane?

A

Doxil - Doxorubicin in Liposome. This is an anticancer drug with dose liminiting cardiotoxicity, so with the liposome, it changed the distribution of the drug so that it doesn’t get into the heart anymore, thus reducing its toxic effect. The liposomes are PEGylated. It’s made through remote loading, so the lysosome is formed, neutral Doxorubicin enters the lysosome, the acidic conditions charges the doxorubicin, so now it’s trapped in there.
- Used for ovarian cancer, AIDS-related Kaposi’s sarcoma, and multiple myeloma.

Taxol - Paclitaxel + Cremophor EL. Cremophor EL is a non-ionic surfactant that forms a micelle around Paclitaxel.
- Used for advanced carcinoma of the ovary, breast cancer, lung cancer, and Kaposi’s sarcoma.

Albraxane - Paclitaxel + human albumin for injectable suspension. Using albumin avoids the complications related to surfactants, so you can give more of the drug, leading to a better effect.
- Used for metastatic breast cancer

28
Q

What are the two main problems with Cremophor EL?

A
  1. It has potential to leach out plasticizers from the standard intravenous tubing, so we need to use special infusion sets.
  2. It may cause hypersensitivity reactions. Due to these reactions, we usually want to pre-medicate the patient with corticosteroids and antihistamines.
29
Q

What is the critical micelle concentration (CMC) and how is it related to micelle stability?

A

Micelles - surfactants will aggregate in aqueous solution to form micelles at a certain concentration and temp.

CMC - When the surfactants or block polymers (PEG, PLA) are present above a certain concentration, micelles will form. That concentration is the CMC. At a concentration lower than the CMC, micelles will not form.

A high CMC means that there will be fast disintegration of micelles upon dilution, which is not desirable for systemic drug delivery (we want to drug to stay inside the micelle). We want a low CMC.

  • Polymeric micelles help solubilize hydrophobic drugs in water and they are less toxic than surfactants (like Cremophor EL and Polysorbate 80).
30
Q

How does polyethylene glycol (PEG) surface provide nanoparticles with the “stealth” function?

A

PEG prevents opsonization by shielding the surface charge of the phospholipid, increasing surface hydrophilicity (making a hydrated layer that prevents opsonins from forming), and enhancing repulsive interaction between liposomes and blood components.

This increases the circulation time of the liposome and decreases the reticuloendothelial system accumulation.

31
Q

What components of mRNA vaccines are critical to complexation with mRNA?

A

Gene vaccines introduce a new gene into the body to help fight a disease. mRNA vaccines are an example of this.

Having an ionizable lipid is critical for the lipid nanoparticle to bind to mRNA. This can be seen in a tertiary amine.

32
Q

What are the differences between Moderna, Pfizer, and Janssen covid vaccines?

A

Moderna and Pfizer are RNA vaccines that use lipid nanoparticles (non-viral) as a carrier. the mRNA used encodes a spike protein.

Janssen is DNA that encodes a spike protein and is incorporated into Adenovirus vector and then injected.