Lecture 13 - Tumour Immunology Flashcards

You may prefer our related Brainscape-certified flashcards:
1
Q

Can the immune system protect the host from cancer?

Give 3 examples of historical experiments that tried to associate the immune system with cancer?

A
  • 1890’s: William Coley treated cancer patients with bacterial extracts to activate general systemic immunity
  • 1950’s: Using rodent models, it was relatively easy to immunise against transplantable tumours and ‘tumour immunology’ was an ‘optimistic’ field. However, this was an artefact of allogenic responses (transplant rejection)
  • Second of 20thC: experiments by Ludvig Gross, George Klein and more recently Theirry Boon demonstrated that a protective response can be generated against a ‘non-immunogenic murine tumour
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Explain the mice model experiments that showed that tumours can be immunogenic

A
  • inject carcingon
  • tumour develops, removed
  • irradiate cells and vaccination naive mouse with them
  • challenge vaccinated mouse with live tumour cells and mouse remains tumour free
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Explain the mice model experiments that showed that tumours can induce immunological memory

A
  • culture tumour cells in vitro
  • challenge naive mouse with live tumour cells –> tumour develops
  • challenge original mouse with live tumour cells –> mouse remains tumour free
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the three forms of evidence of tumour protective immunity in humans?

A
  1. immunosuppressed individuals more frequently develop cancer (especially virus-associated cancers) than immunocompetent individuals
  2. cancer patients can develop spontaneous immune response to their own tumours
  3. the presence of immune cells within some tumours correlates with improved prognosis e.g. colorectal cancers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is cancer immunosurveillance?

A
  • this concept predicts that the immune system can recognise precursors of cancer and, in most cases, destroy these precursors before they become clinically apparent
  • this theory was controversial because early studies in ‘immunocompromised’ mice did not support it. (however not good models of immunocrompromisation)
  • More recent work with fully immunocompromised mice indicates that the theory is correct
  • however, the immune system not only protects the host for tumour development but also sculpts, or edits, the immunogenicity of tumours that may eventually form
  • Therefore a new term was introduced –> cancer immunoediting
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are the 3 E’s of Cancer Immunoediting?

A
  1. Elimination - immune-mediated destruction of most cancer cells
  2. Equilibrium - a dynamic equilibrium between the immune system and any tumour cell variant that has survived the elimination phase. The immune response is enough to contain, but not fully extinguish these genetically unstable and mutating tumour cells
  3. Escape - tumour cell variants selected in the equilibrium phase now growth out in a immunologically intact environment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the 4 key areas of T cells for cancer therapy?

A
  1. How how might T cells detect and destroy cancer cells?
  2. Mechanisms by which tumours might escape the immune response
  3. Developing T cell-based therapies for cancer
  4. Potential side effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Describe the normal interactive between APC’s and T helper cells

A
  • T cells recognise antigens displayed on the cell surface in the form of short peptide fragments bounds to molecules of MHC
  • Signal 1 = TCR interacting with MHC II
  • Signal 2 = costimulatory molecole interaction e.g. CD28 on T cells interacts with CD80/CD86 on APC
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What happens once a T cell has been activated by an APC?

A

It leaves the lymph node and enters the circulation

Moves back to the tumour site to carry out immune response

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

What are the 6 categories of tumour-associated antigens? Give examples

A
  • Mutated self proteins (e.g. from DNA damage) = CDK in melanoma, beta-catenin in melanoma, Capase 8 in squamous cell carcinoma
  • Aberrantly or over-expressed self protein = oncofoetal antigens, cancer-testis antigens, telomerase in many cancers
  • Lineage specific (differentiation) antigens = mart1/MelanA, gp100 and tyrosinase in melanoma, surface immunoglobulin in melanoma
  • Abnormal post-translocational modifications of self proteins = MUC1 over expressed in an underglycosylated form in breast and pancreatic cancer
  • Viral proteins = HPV in cervial cancer, EBV in Hodgkin’s lymphoma, Nasopharyngeal carcinoma
    Alternatively, target the tumour stroma? (e.g. tumour endothelial markers)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What 4 things makes a good target antigen for tumour immunotherapy?

A
  1. tumour specific –> reduce toxicity
  2. shared amongst patients with the same and different tumour types –> widely applicable
  3. Critical for tumour growth/survival –> lack of antigen-loss variants
  4. lack of immunological tolerance –> high avidity T cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What are good and bad examples of tumour specific antigens?

A
Good
- mutated self-proteins 
- viral antigens
- cancer-testis antigens 
Bad
- lineage specific antigens 
- over expressed self proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What are good and bad examples of antigens shared amongst tumours?

A

Good
- mutated self or viral proteins involved in oncogenesis
- lineage specific antigens
Bad
- mutated self proteins incidental to oncogenesis

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

What are good and bad examples of antigens critical for tumour growth/survival?

A
Good
- mutated self or viral proteins
- overexpressed self proteins 
Bad 
- mutated self proteins incidental to oncogenesis 
- lineage specifically antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are good and bad examples of antigens that lead to lack of immunological tolerance?

A
Good
- mutated self proteins 
- viral antigens 
Bad
- lineage specific antigen 
- over-expressed self proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

List the 7 mechanisms whereby tumours might escape the immune response

A
  • loss of HLA class I expression (e.g. lung cancer, prostate, melanoma etc.)
  • reduced expression of other molecules involved in antigen processing/presnetation (TAP1, LMP2, LMP7 and tapasin) (e.g. colorectal cancer)
  • loss of costimulatory molecule expression (CD80, CD86)
  • loss of adhesion molecule expression (ICAMI)
  • loss of target antigen (e.g. melanoma)
  • inhibiting T cell infiltration
  • immunosuppression at the tumour site
17
Q

Describe inhibiting T cell infiltration

A

i) endothelia B receptor expression ont the tumour endothelium signals to prevent modulation of ICAM and thereby T cell adhesion to the vascular endothelium
ii) nitrosylation of chemokines can keep T cells from entering the tumour core

18
Q

Describe immunosuppression at the tumour site

A
  • Regulatory T cells - functionally define by their ability to inhibit an immune response by influencing the activity of another cell type
  • CD4, FOXP3, CD35, CD127, mediate function through cytokine realise, and/or direct cell-cell contact
  • Important int controlling immune responses to self-antigens
  • may play a tole in tumour escape
  • may be selectivity recruited to the tumour site by chemokines
    see lecture slides
19
Q

What are the 3 types of T cell-based therapies for cancer?

A
  1. Non-specific T cell stimulation
  2. Vaccination
  3. Adoptive T cell therapy
20
Q

Describe non-specific T cell stimulation

A
  • immunostimulatory cytokines e.g. IL2 for melanoma and renal cell carcinoma
  • blockade of immunologic checkpoints e.g. administering antagonists antibodies (Ipilimumab( specific for the coinhibitory receptor CTLA4 improved overall survival in patients with previously treated metastatic melanoma
21
Q

Give examples of some vaccines already being teseted

A
  • Tumour cells/lines (irradiated or lysed) - may be combined with strategies designed to enhance the immunogenicity of the tumour cells (e.g. induce expression of co-stim molecules or cytokines)
  • defined peptides or protein antigens (purified or expressed from recombinant viruses, DNA or RNA)
  • Dendritic cell based vaccines e.g. Sipuleucel used to treat metastatic prostate cancer
22
Q

What is TIL therapy

A
  • used successfully in melanoma
  • tumour biopsy
  • tumour-infiltrating lymphocytes removed
  • select tumour-specific TIL
  • infuse >10^10 cells and high dose IL2
  • also non-myeloablative conditioning (to make room for new lymphocytes
23
Q

What is the main side effect of T cell therapy in cancer?

A

Autoimmunity

  • studies in mouse models suggest that potent tumour immunity can be induced by vaccination with specific self canteens or unfractioned tumour derived material with little or no evidence of autoimmunity but…
  • TIL therapy results in symptoms of autoimmune melanocyte destruction inlcuding vitiligo or uveitis in some melanoma patients
  • Administering antagonistic antibodys specific for CTLA4 or PD1/PDL-1 resulted in anti-tumour responses in a number of patient but this was associated with severe autoimmune responses
24
Q

What are the immune mediated mechanisms of action for monoclonal antibodies for cancer therapy?

A
  • complement mediated lysis = antibody binding to to CD20 activates complement cascade, forming MAC
  • antibody-dependent cellular cytotoxicity (ADCC) = antibody binds to cancer cell via CD20, engage NK cell which binds to antibody via Fc gamma receptor –> kills with perforin and granzymes
25
Q

What are the direct efffects of monoclonal antibodies for cancer therapy?

A
  • Blocking receptor ligand interactions
  • Anti-angiogenesis
  • Cytotoxic activity of conjugated antibodies