Cell therapeutics Flashcards

1
Q

What is adoptive cell therapy

A

Adoptive Cell Therapies: using a patient’s immune cells to fight Off disease

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

What is a DC is and its role in immunity?

A
  • Professional APC - antigen processing
  • Bridges adaptive and innate immunity
  • Most potent APC: In cancer, they present tumor antigens
  • T cell responses are initiated by activated DCs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is DC in vivo activation, in vivo expansion, and blocking of inhibitory signals

A

in vivo activation: Provide exogenous activation signals (attempt to reverse supression)

in vivo expansion: DCs are not high in number in the TME. Hypothesis: increasing the number of intratumoral cDC could increase the cumultative function of the population

blocking of inhibitory signals: A suppression microenvironment is a key regulator of cDC function. Hypothesis: BLocking inhibitory pathways that reduce cDC functionality may enhance the activation state of tumor cDC

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

What is a γδ Τ cell is and its role in healthy individuals compared to cancer patients

A

A subset of T cells, bearing a γδ heterodimer as a T cell receptor (TCR)

Healthy individuals:
γδT cells are naturally biased towards a cytotoxic phenotype,
producing antitumor cytokines IFNγ-, TNF.
Newborns harbor a population of circulating IFNγ- and TNF-positive
γδT cells.
Diversity generated by microbial encounter, leading to functional
diversity.

Cancer patients:
Tumour encounter through γδT cells can result in anergy or the
deletion of γδT cells.
IL-17 production by γδT cells enhances tumour progression.
Increase the expression of myeloid derived suppressor cells (MDSCs)
in the TME

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

Describe different ways that γδ Τ cells can be exploited for therapies. Focus mainly on DOT cells

A

Goal: To induce de novo expression of natural cytotoxic receptors, particularly NKP30 and nkP44, while also upregulating the expression of NKG2D and DNAAM1
–> enhances an NK cell response

Method: This is achieved by a Vδ1-enriched (>60%) γδT cell product for adoptive transfer, produced in a period of over 3 weeks under stimulation with TCR agonists and cytokines.

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

What is a NK cell and its biological role

A

-Innate immune cell
-2 - 18% of human peripheral blood lymphocytes
-Widely found in the spleen and lymph nodes

-Able to distinguish between heathly, stressed and transformed cells

-Activated;
a) via receptors: Able to lyse cells without prior stimulation

b) Via antibody-dependent cellular cytotoxicity

-Primed in response to various cytokines (e.g. IL-2, IL-15)
-Release cytotoxic granules (e.g. perforin, granzyme)

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

Describe the three main treatment modalities targeting NKs (cytokine supplements, monoclonal antibodies, adoptive cell therapy)

A

Cytokine supplements:

IL-15: expands NK cells

IL-21: important in maturation

IL-12: Critical for NK memory cell formation

IL-2: enhance their cytolytic activity

Can be combined with other therapies to give synergistic effect.

Monoclonal antibodies:
Blocking KIRs would allow a more efficient NK cell cytotoxicity

Epidermal growth factor receptor inhibitor treatment: NK cells are more enriched in the TME

MICA antibodies: prevention of NK cell recognition –> prevention of inhibition in TME

Adoptive cell therapy:

adoptive transfer:
Transfer of NK cell along with CD34+ HSCs is promising

Antibody engineering:
BiKES and TRiKES

BiKE connects a single-
chain variable fragment (scFv) to the anti-CD16 recogni-
tion site with the scFv of a tumor specific antigen, such
as CD19/CD20 for non-Hodgkin lymphomas

TriKE consists of a BiKE and cyto-
kine IL-15, which boost NK cell function and survival

Genitic modification:
Culture in vitro and differentiate in CAR-NK in vivo
Or deleting surface molecules (e.g. CD38) which enhances cytotoxic ability

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

What is the difference of engineered vs non-engineered T cell therapies?

A

Non-engineered immune cell therapies typically include apheresis, turmor resection or banked donor T cells which are subjected to Treg isolation and expansion; T cells isolation and expansion; Expansion, respectively. These will in turn yield Tregs, TILs and Viral-specific CTLs

Engineered immune cell therapies are generated by
first apheresing or drawing blood from the patient, isolating T cells, and using viral or non-
viral approaches to insert a transgene encoding a synthetic receptor. These can yield engineered TCRs, CAR-T, CAAR-T, CAR-Treg or CAR-NK

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

Describe the three main non-engineered treatment modalities targeting T cells (TILs, viral-specific CTLs, Treg cells)

A

TILs: tumor-infiltrating lymphocytes
Ex vivo expansion and re-infused in patients with IL-2

Viral-specific CTLs: cytotoxic t lymphocytes
Infusion of donor leukocytes pre sensitized to EBV

ex/in vivo expansion and transfusion of autologous or allogeneic Tregs Therapeutic strategy!

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

Reflect on the advantages of cellular therapeutics over another type of biopharmaceutical

A

Cellular therapies are precise and lower the risk of drug resistance, although ADAs are a risk. Cell therapeutics are a first gen-technology and expensive and often personalized

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

Drawback of DC in vivo activation/expansion?

A

Single-agent administration in patients is not efficacious

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

DOT therapy drawback?

A

Drawback: potential downraguation of these naturally expressed receptors once the cells are infused into a patient

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

What is the NK cell therapy goal?

A

To enhance NK cell function and cytotoxicity

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

What is a drawback with TILs?

A

Not enough to remove tumor on its own

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

What is a drawback with viral-specific CTLs?

A

Patients can be immunized against donor cell products. Alternative methods use the patients’s own virus-specific immune cells or modify T cells with virus-specific properties

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