Immune 4 - Tumour immunology and immunotherapy of cancer Flashcards

1
Q

What causes paraneoplastic cerebellar degeneration (PCD)

A

Anti-CDR2 antibody

CDR2 = cerebellum degeneration-related antigen 2

Elimination of purkinje cells by tumour induced auto-immune response

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

What is meant by tumour “immunosurveillance”

A

Malignant cells - generally controlled by the action of the immune system

Immunotherapy tries to enhance immune responses to cancer

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

Highlight quick differences between T cells and B cells

A

T cells = “MHC restricted”. Alpha and beta T cell receptor

B cells = vast range of molecules recognised by antibody

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

Explain the cancer-immunity cycle

A
  1. Death of cancer cell - loads of antigen released
  2. Cancer antigen presented on APC
  3. T cell recognition at lymph node - T cell activation
  4. T cell moves to tumours
  5. T cell infiltration into tumours (TIL)
  6. Recognition of cancer cells by T cells
  7. Killing of cancer cells (consequently more antigens are released)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Certain inhibitory signals are involved in the cancer immunity cycle - these are targeted in cancer immunotherapy. Eg?

“immune checkpoint blockade”

A
  1. CTL-A4 / PD-L1 / PD-1
  2. PD-L1

Were trying to remove the negative signals inhibiting the immune response

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

What are the 2 requirements for activation of an adaptive anti-tumour immune response?

A
  1. Local inflammation in the tumour (“danger signal”)
  2. Expression and recognition of tumour antigens

Tumour inflammation takes a while to trigger adaptive immune response

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

What are the problems with immune surveillance

A
  1. It takes the tumour a while to cause local inflammation

2. Antigenic differences between normal and tumour cells may be very subtle - e.g. a small number of point mutations

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

What is the basis of cancer immunotherapy

A

“teaching” the adaptive immune system to selectively detect and destroy tumour cells - potential alternative/supplement to conventional therapies

usually minimal side effects

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

How might immune responses to tumours be similar to virus infected cells?

A

T cells can see inside cells - and can recognise tumour specific antigens (presented on MHC class 1/2).

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

Name some tumour specific antigens to target

A

Viral proteins:

  1. Epstein Barr virus (EBV)
  2. Human Papillomavirus (HPV)

Mutated cellular proteins:
1. TGF-beta receptor

(The above all may cause cancer)

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

Which opportunistic malignancies can cause cancer (viral origin) in immunosuppressed individuals

A
  1. EBV positive lymphoma - post transplant immunosuppression

2. HHV8-positive Kaposi sarcoma: HIV

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

In immunocompetent individuals, what cancers arise from viral origin

A
  1. HTLV1-associated leukaemia/lymphoma
  2. HepB and HepC virus-associated hepatocellular carcinoma
  3. HPV - positive genital tumours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Which are the oncoproteins of HPV that induce and maintain cervical cancer

A

E6 and E7

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

What are the target antigens for preventive HPV vaccination

A

“Late genes”
L1 and L2

These are surface proteins, incorporated into Virus-Like particles (VLPs)

(we don’t use E6 & E7 oncoproteins - as potentially dangerous)

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

To whom is the HPV vaccine given therapeutically?

NB can also be given preventively

A

To those who are infected by HPV16 and have not been successful in clearing it (immune failure)

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

What are tumour-associated antigens (TAAs)

A

They are normal cellular proteins which are aberrantly expressed (timing / location / quantity)

They are normal self proteins, so tolerance may need to be overcome to generate an immune response

17
Q

Tumour associated antigens are ectopically expressed auto-antigens. They are so called developmental antigens, meaning?

A

They are silent in normal adult tissues except male germ cells (some expressed in placenta).

18
Q

Give an example of a tumour associated antigen

A
  1. e.g. MAGE family - melanoma associated antigens.
  2. Human epidermal growth factor receptor 2 (HER2) - overexpressed in some breast cancers
  3. Mucin 1 (MUC-1) - membrane associated glycoprotein - over expressed in many cancers
  4. Carcinoembryonic antigeen (CEA) - normally only expressed in foetus/embryo - but overexpressed in variety of cancers

Prostate:

Prostate specific antigen (PSA), Prostate-specific membrane antigen (PSMA), Prostatic acid phosphatase (PAP)

19
Q

Some tumours escape central tolerance - these are often the targets of cancer therapy. The may express auto-immune reactivity against normal cells. Give an example

A

Melanocyte/melanoma - tyrosinase (melanin production) - has poor self tolerance

20
Q

What are 2 major problems with targeting tumour associated auto-antigens for T cell mediated immunotherapy of cancer?

A
  1. Auto-immune responses against normal tissues
  2. Immunological tolerance:
    - normal tolerance to auto-antigens
    - tumour induced tolerance
21
Q

Describe monoclonal antibody based therapy for cancer

A

Can be:

  1. “naked” - e.g. Perception (anti-HER2)
  2. “Conjugated” - radioactive particle, e.g. anti-CD20 linked to yttrium-90, or anti-HER2 linked to cytotoxic drug (trastuzumab emtansine)
  3. “Bispecific antibodies” - genetically engineered - combines 2 specificities, e.g. anti-CD3 and anti-CD19 (e.g. Blinatumomab, used in some patients with B cell tumours)
22
Q

Give an example of therapeutic cancer vaccination

A

Provenge (Sipuleucel-T) - not NICE approved but FDA approved

Patients own WBC treated with fusion protein between prostatic acid phosphatase (PAP) and GM-CSF cytokine. Stimulates DC maturation and enhances PAP-specific T cell responses

23
Q

Describe “personalised” tumour specific cancer vaccines

A

Normal cell DNA and tumour cell RNA taken, and sequenced. HLA typing done. Neoantigens formed, which are combined with an adjuvant and given as a personalised vaccine

24
Q

Describe the basis for immune checkpoint blockade approach to cancer treatment

A

This approach reduces/removes negative regulatory controls of existing T cell responses

Targets CTLA-4 and PD-1 pathways.

CTLA-4 = expressed on activated and regulatory T cells, binds to CD80/86

PD-1 = expressed on activated T cells, binds to PD-L1/L2

e.g. Ipilimumab given (anti-CTLA-4), Nivolumab (anti-PD-1), antagonistic antibodies

problem is, it may incite an autoimmune response

25
Q

Describe what adoptive transfer of cells (ACT) is

A
  1. Take WBCs from patient with tumour
  2. Expand T cells in vitro (can be antigen specific expansion or non specific expansion)
  3. We can also genetically engineer the T cells, e.g. by introducing new antigens into them
  4. Once T cell culture has expanded, we can put them back into the human
26
Q

Give an example of a genetic-engineering type approach to cancer therapy

It has been used for B cell tumours using CD19

A

CARs (Chimaeric Antigen Receptors)

Can introduce a CAR into a T cell where the binding bit of receptor has been derived from an antibody - variable region of antibody can be scFv (Single chain fragment variable)

scFV stuck on the outside of the cell - an extension of the T cell receptor chain

If CAR binds to (tumour) antigen –> it will deliver an activating signal to the T cell –> then destroys T cell