IC2 Flashcards

1
Q

List 2 types of immunotherapies and how they work

A
  • Activation type of immunotherapies: involve use of agents that augment and/or reestablish the immune system’s ability to prevent and fight the disease.
  • Suppression type of immunotherapies: involve use of agents that reduce or suppress an immune response.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is an immunotherapy?

A

Treatment of a disease by intervening the immune system.

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

Examples of therapeutic strategies in immunotherapy

A
  1. Cytokines (humoral component of innate & adaptive immune response)
  2. Antibodies (humoral component of adaptive immune response)
  3. T cells (include cell- based immunotherapy and checkpoint inhibitors)
  4. Cancer vaccines and many more due to advances in molecular cloning techniques and biotechnology!
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Properties/ facts of cytokine

A

• Though belonging to diverse groups, are usually small proteins of < 20 kDa.
• Are usually glycoproteins (glycosylated). Glycosylation sometimes not essential for cytokine’s biological activity, sometimes contributes to half-life of cytokine.
• Usually have short half-lives.
• Are secreted out of the cell producing it. Usually act at short range in an autocrine (i.e. on the cells that produce them) or paracrine (i.e. on cells nearby) manner.
• Binds specifically to its corresponding cytokine receptor expressed on surface of effector cell -> trigger biological effects.

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

Two types of IFN and example

A
  • Type I (IFN alpha, beta)
  • Type II (IFN gamma)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Which cell produces IFN-alpha?

A

leukocytes

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

what cell produces IFN-beta?

A

somatic cells, including fibroblasts and epithelial cells.

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

what condition is IFN-beta effective against?

A

multiple sclerosis

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

what cell produces IFN-gamma?

A

T lymphotcytes

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

production of antiserum

A
  • whole blood from immunized animal collected → left to clot or add coagulant → clotting factors removed → serum obtained as supernatant after centrifugation to separate cellular components.
  • Raw serum further purified by eliminating serum proteins and enriching the fraction of Ig that reacts with the target Ag (by protein A/G purification or immunoaffinity column chromatography)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

what is an antiserum?

A

Animal sera containing polyclonal antibodies raised by immunizing the animal with a particular antigen.

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

drawback of antiserum

A

immunogenicity issues

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

what is a monoclonal antibody?

A

Ab produced from 1 B-cell clone, recognize just 1 epitope of the antigen.

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

characteristic of monoclonal Ab and its usefulness

A

• Possess high specificity → Useful for:
– Recombination protein purification work (used in immunoaffinity chromatographic purification of proteins).
• Possess high homogeneity → Effects obtained highly reproducible → Useful for:
– Commercial development into target-specific therapeutic molecules for treatment of diseases with less side effects.
– Commercial development into diagnostic test kits.
– Many experimental research techniques.

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

characteristic of monoclonal Ab and its usefulness

A

• Possess high specificity → Useful for:
– Recombination protein purification work (used in immunoaffinity chromatographic purification of proteins).
• Possess high homogeneity → Effects obtained highly reproducible → Useful for:
– Commercial development into target-specific therapeutic molecules for treatment of diseases with less side effects.
– Commercial development into diagnostic test kits.
– Many experimental research techniques.

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

Limitations of murine Mabs

A

• human-anti-mouse antibody response (HAMA) characterized by joint swelling, rashes and kidney failure
• Fail to trigger a number of effector functions.
• Shorter half-lives (30 - 40 h)

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

% of human sequences in chimeric Mab and how is this achieved?

A

• To replace amino acid sequences on the CH and CL of murine Mab that are not essential for antigen binding with human sequences. Antigen-binding sequences in VH and VL fragments conserved.
• Chimeric Mab retains antigen selectivity and affinity similar to parent murine Mab.
• ~ 75% human

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

% of human sequences in humanised Mab and how is this achieved?

A

• Replacing all mouse amino acid sequences except the hypervariable CDR domains of Ig (reside within the VH and VL fragments) in chimeric Mabs → Humanized Mabs.
• > 90% human (reduced immunogenicity as compared to chimeric Mabs).

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

remaining immunogenicity concerns for recombinant human monoclonal antibodies

A

impurity proteins from mammalian host cells not removed

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

Nomenclature of 4 types of Monoclonal Antibody

A

– Mice (infix, -o-)
– Chimeric (infix, -xi-)
– Humanized (infix, -zu-)
– Antibodies originating in humans (infix, -u-)

20
Q

Role of pepsin & papain

A

proteases that cleave peptide bonds to yield F(ab’)2 and Fab respectively.

21
Q

what is ScFv

A

single chain variable fragment; contains amino acid sequences of antigen- binding CDRs in the VH and VL in the Fab arm of an Ig in a single polypeptide chain.

22
Q

what is BiTEs?

A

Bispecific T cell engagers (BiTEs) - have 2 distinct Fab regions and are capable to bind to two distinct epitopes. Lack a Fc domain.

23
Q

what is a triomab?

A

Trifunctional mAbs (triomabs) - bind to 2 different antigens while maintaining the capacity to mediate Fc-dependent effector functions. Possess a Fc domain.

24
Q

In naturally occurring IgG, is the Fc domain fucosylated? (Y/N)

A

Yes

25
Q

Fucosylated Fc domain of IgG causes _____

A

reduced affinity of the Fc domain to bind to an subtype of activating Fc receptor (FcyRIII) found on effector cells -> reduced ADCC induction by effector cells -> reduced efficacy of fucosylated antibodies

26
Q

what is TIL therapy

A

TIL (tumour infiltrating lymphocytes) therapy:
- Isolated TILs from patient’s tumor masses consist of T and NK cells. Isolated T cells are polyclonal, have diverse antigen specificity
- Isolated TILs, mainly T cells are expanded in vitro by a rapid expansion process (REP) - exposed to high dose IL2 + anti-CD3 antibody (to activate CD3 in TCR) + irradiated feeder cells [usually autologous PBMCs (peripheral blood mononuclear cells) obtained from patient]
- Large quantities (~10-150 billion) of lymphocytes produced and infused into patient

27
Q

what are TCR-T and CAR-T therapies (in general)

A

T cell receptor engineered T cell (TCR-T) therapy and Chimeric antigen receptor T cell (CAR-T) therapy:
1. T cell isolation from patient’s peripheral blood
2. Genetic modification with CAR/ TCR
3. Genetically engineered T cells (TCR or CAR) expanded in vitro, then infused back into the
patient -> recognize tumor antigens and attack cancer cells.

28
Q

Advantages & disadvantages of TIL therapy

A

Advantages: Generally safe compared to CAR-T/ TCR-T
Limitations:
- For some patients, excised tumor masses are devoid of or contain very low quantities of TILs.
- Expanded naturally occurring tumor-specific T cells are heterogeneous possessing varying antigen specificities -> TIL reinfusion may not be lethal enough to attack and eradicate cancer cells.
- Limited or none of the expanded naturally occurring tumor antigen- specific T cells possess high affinity.

29
Q

Principle of TCR-T therapy

A
  • Genes encoding Va and Vb within the a and b chains that make up a T cell receptor (TCR) are tumor antigen-specific.
  • Genes are cloned into retro- or lentiviral vectors, which in turn are used to transduce T cells isolated from patient’s peripheral blood
  • Genetically modified T cells less heterogeneous and high transduction efficiency of retro- or lentiviral vectors ensures that reinfused T cells possess high tumor antigen specificity
30
Q

Advantages of TCR-T therapy

A

Advantages:
- Engineered TCR-T cells possess full TCR complex -> can recognize antigens expressed at both cell surface/tumor surface and within tumor cell/tumor mass -> can penetrate tumors -> effective against solid and hematological tumors.
- Compared to CAR-T cells, TCR-T cells use full TCR complex for antigen recognition and signal transduction (CD3 adaptor proteins)
-> 1. can be fully activated at low target cell antigen densities,
2. onset of signaling slow but of longer duration and
3. execute more extended killing -> more effective than CAR-T therapy for cancer treatment.

31
Q

Disadvantages of of TCR-T therapy

A

Disadvantages:
- Engineered TCR-T cells expressing a particular tumor-specific TCR only limited for use in a patient subpopulation carrying a specific MHC/HLA allele recognized by TCR. (recall: MHC is polygenic and polymorphic)
- Less safe than TIL therapy –
1. On-target off-tumor toxicity (TCR-T cells target normal tissue expressing same antigen. E.g.
both normal melanocytes and melanoma cells express gp100 and MART-1)
2. Off-target toxicity (TCR-T cells not specific and cross-react with other antigenic fragments)
3. Cytokine-release syndrome (CRS) (infusion of TCR-T cells induce a cytokine storm).

32
Q

Where do CDR 1,2,3 in Va and Vb of TCR bind to?

A

CDR1: bind to each terminus of the peptide in peptide-MHC
CDR2: recognize and bind to MHC in peptide-MHC
CDR3: recognize and bind to peptide in peptide-MHC
Recall: Strength of interaction between TCR and peptide-MHC contributes to extent of T cell activation and development of antigen-specific T cells as memory T cells

33
Q

Principle of CAR-T therapy

A
  • Construction of chimeric antigen receptor (CAR): genetic sequence encoding for specific antigen-binding sites within VH and VL in the Fab domain of an identified antibody (known to be targeting specifically a cell surface molecule of interest) is cloned into a retro- or lentiviral vector.
  • T cells isolated from patient’s peripheral blood are transduced by vectors carrying CAR gene -> Transduced T cells express CAR on surface (now called CAR-T cells) -> In vitro expansion of CAR-T cells and cells are infused into the patient to bind to antigens on cancer cells to kill them.
34
Q

Similarities and differences btw the 4 generations of CAR-T cells

A

Similarity: All contains only ONE extracellular domain -> scFv -> R&B to antigen
Differences:
- 1st gen: 1 intracellular domain CD3z
- 2nd gen: 2 intracellular domain CD3z + CD28/ 4-IBB
- 3nd gen: 3 intracellular domain CD3z + CD28 + 4-IBB
- 4th gen: 3 intracellular domain CD3z + CD28 + 4-IBB + 1 transgene (upon target antigen binding, activated to express cytokines -> activate more T cells to kill cancer cells)

35
Q

Advantages of CAR-T therapy

A
  1. CAR-T cells recognize and bind to unprocessed tumor surface antigens without MHC processing. possess full TCR complex -> can recognize antigens without MHC proteins.
  2. scFv domain only binds to cell surface antigens -> effective against hematological tumors (patients with acute lymphoid leukemia respond very well), but not effective against solid tumors
36
Q

Disadvantages of CAR-T therapy

A
  1. scFv may guide CAR-T cells into an antigen- independent mechanism -> failed therapy
  2. Less efficient than TCR-T therapy
    - Activated only at higher target cell surface antigen densities
    - Only one subunit (scFv) binding to target cell surface antigen -> weaker CAR signaling and activation -> execute faster killing function but lack extended killing.
  3. Adverse effects:
    - On-target off-tumor toxicity mainly limited to B cell aplasia (due to CD19-specific CAR-T cells attacking and killing normal B cells also expressing CD19 like the malignant CD19+ B cells)
    - Off-target toxicity (not highly reported)
    - Cytokine-release syndrome (CRS) (induce CRS more than TCR-T therapy)
37
Q

Which cell expresses checkpoint molecules proteins on their surface?

A

T cell

38
Q

Native function of checkpoint proteins

A

to counter overstimulation of T cell activity and to prevent autoimmune responses.

39
Q

Examples of checkpoint molecules identified as targets for development of therapeutics

A
  1. Cytotoxic T-lymphocyte associated protein 4 (CTLA-4)
  2. Programmed death 1 (PD-1)
40
Q

Principle of anti-CTLA4 Mab

A

CTLA-4 and CD28 expressed on activated T cells. During antigen presentation by APC, costimulatory molecule CD80/C86 expressed on APC binds simultaneously to CD28 on
T cells to increase T cell activation -> activated T cell executes cell killing effect.

Expressed CTLA-4 competes with CD28 for binding with CD80/C86 -> reduces CD80/C86 - CD28 binding -> T cell activation inhibited

anti-CTLA4 Mab used to promote T cell activation

41
Q

Principle of PD-1 inhibitors/ PD-L1 inhibitor

A

PD-1 expressed on activated T cells.

In the intratumor microenvironment, a number of cells including DCs, tumor- associated macrophage (subset of macrophage), fibroblasts and even tumor cells themselves express PD- L1 (PD-1 ligand) and/or PD-L2

PD- L1/PD-L2 binds to PD-1 and suppress T cell activity.

PD-1 inhibitors/ PD-L1 inhibitor used to promote T cell activity.

42
Q

3 categories of cancer vaccines

A

– Cell vaccines
– Protein/peptide vaccines
– Nucleic acid (DNA, RNA) vaccines

43
Q

Designs for Cell Vaccines

A
  • Tumor cell vaccine -> antigens expressed by tumor cells to induce T cells in patient.
  • DC vaccine -> tumor antigenic proteins/peptides or tumor cells loaded on DCs, DCs administered into cancer patient for tumor antigens to induce T cells
44
Q

Principle of Protein/Peptide Vaccines

A

Vaccine formulations contain antigenic peptide fragments derived from tumor-associated antigens -> upon administration, recognized by T cells and activate T cells.

45
Q

Why are peptide/ protein vaccines not found to produce strong immune responses?

A
  1. Neoantigens (arising from specific gene mutations in a cancer patient) are not included.
  2. Antigenic peptides of short chain restricted to MHC class I binding -> MHC class II binding not engaged and hence subsequent activation of CD4+ helper T cells followed by CTL induction is absent.
  3. Antigenic peptides of short chain may bind to any cell without triggering further processing -> may induce anergy (lack of immune response to an antigen) and cause immune tolerance.
46
Q

Principle of Nucleic acid Vaccines

A

Vaccine formulations contain DNA or RNA that code for tumor associated-antigenic proteins/peptides. Can be delivered using a viral vector (efficient delivery of DNA/RNA into cells). Non vector- based delivery system also used (e.g. liposomal formulation).

47
Q

Concern of DNA vaccine

A

Nucleic acid gets incorporated into host cell genome -> more lasting expression of antigenic proteins/peptides, but risk of inducing carcinogenicity if insertion of gene causes insertional mutagenesis and switch on oncogenes.

48
Q

Concern for RNA vaccine

A

No risk of carcinogenicity caused by insertional mutagenesis since RNA not incorporated into host cell genome. Main concern is RNA stability in formulation and upon administration.