Year 3 Flashcards

1
Q

What is flow cytometry used for?

A

To count and analyse the size, shape and properties of individual cells within a heterogeneous population of cells

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

What type of data is in flow cytometry?

A

Quantatative

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

What is analysed in a flow cytometer?

A
  • Cells
  • Tissues
  • Organisms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is the flow order of a flow cytometer?

A
  • Sample
  • Fluidics that move the sample into the flow cytometer
  • Lasers
  • Optics which gather the light
  • Detectors to sense the light
  • Computer system to output the data into a form that can be analysed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the forward scatter proportional to in a flow cytometer?

A

The size of the cell

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

What occurs to the forward (side) scattered light in a flow cytometer?

A
  • Detector converts the scattered light into a voltage which is directly proportional to the amount of forward scattered light
  • The computer converts this data into a histogram plot with amount of forward scattered light on the x-axis and the number of cells on the y-axis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is the side scattered light proportional to in a flow cytometer?

A

Shape and internal complexity of a cell

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

What form does the data of fluorescence take?

A
  • Histogram plot

* Dot plot

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

What are the uses of antibodies?

A
•	Blocking/neutralising
o	Receptors
o	Cytokines
o	Pathogens
o	Toxins 
•	Capturing 
o	Precipitation
o	ELISA
o	Purification of cells
•	Immunotherapy
•	Scientific and clinical study
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What is immunodiagnostics?

A

Diagnostic methodology that uses antigen-antibody reaction as their primary means of detection

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

What can be used to link as a label with an antibody?

A
  • Radiation
  • Enzyme
  • Fluorescent tag
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What diagnostic methods can use labelling?

A
  • Western blotting (antigen-enzyme)
  • Immunohistochemistry
  • Flow cytometry (antigen-fluoresce)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What does CD number stand for?

A

Cluster of differentiation number

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

What are CD numbers used for?

A

Used for the identification and investigation of cell surface molecules providing targets for immunophenotyping of cells

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

What does CD4- mean?

A

The patient does not have CD4 cells

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

What does CD4+ mean?

A

The patient has CD4 cells

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

What does CD16low mean?

A

The patient has a low concentration of CD16

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

What does CD16high mean?

A

The patient has a high concentration of CD16

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

On a histogram where is the positive and negative CD numbers shown?

A
  • Negative to the left

* Positive to the right

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What diseases are due to too much immune reactivity?

A
•	Autoimmune diseases
o	Rheumatoid arthritis 
o	Multiple sclerosis
o	Lupus 
•	Atopic diseases
o	Asthma
o	Siriasis 
o	Anaphylaxis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What diseases are due to too little immune reactivity?

A
  • Infection

* Cancer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the definition of tolerance?

A
  • Teaches what is “self” allowing the adaptive immune system to recognise “non-self”
  • Essential to prevent autoimmune disease
  • Immunocompetent host fails to respond to an immunogenic challenge with a specific antigen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Who discovered that tolerance is acquired and specific?

A

Peter Medewar by doing skin grafts on mice

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is the danger hypothesis?

A

If encounter an antigen in a safe environment, the body is tolerant
• If encounter an antigen in a dangerous environment, the body is not tolerant

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is central tolerance?

A
  • Immature self-reactive lymphocytes that recognise self-antigens in generative (“central”) lymphoid organs die by apoptosis; other fates
  • This occurs during lymphocyte development
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is peripheral tolerance?

A
  • Mature self-reactive lymphocytes that recognise self-antigens in peripheral tissues are inactivated (anergy), killed (deletion) or supressed
  • Occurs after lymphocytes leave the primary lymphoid organs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What features does tolerance have the same as an immune response?

A
  • Antigen specific
  • Can exist in T cells, B cells or both
  • Has immunological memory (T cell tolerance is more profound and lasts longer then tolerance at the B cell level)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Where do B cells originate from?

A

Bone marrow

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Where do T cells originate from?

A

Thymus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What does kinetic proofreading do?

A
  • Measure of affinity

* Assesses how well the T cell receptor and its target antigen is bound

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What does a high affinity in kinetic proofreading mean?

A

Strong binding which lasts between T cell receptor and its target antigen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What does a low affinity in kinetic proofreading mean?

A

Weak binding which falls apart between T cell receptor and its target antigen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What does a resting T cell receptor in a cell membrane that is unphosphorylated mean?

A

That the T cell receptor is turned off

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What does Lck want to do to a T cell receptor?

A

Phosphorylate TCR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What does CD45 want to do to a T cell receptor?

A

Dephosphorylate TCR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What does ZAP70 do to a T cell receptor?

A

Binds to phosphorylated TCRs, signals TCR activation to the rest of the cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What occurs on the cell membrane of a T cell with regards to a resting TCR?

A

Lck and CD45 compete = no overall receptor phosphorylation and no ZAP70 binding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What occurs on the cell membrane of a T cell with regards to the TCR and an irrelevant target antigen?

A
  • No strong interaction = no overall activation

* No ZAP70 binding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What occurs on the cell membrane of a T cell with regards to the TCR and a specific target antigen?

A
  • Strong/close interaction = overall activation
  • CD45 is too big and is excluded
  • Lck is small and can still get to phosphorylate TCR
  • ZAP70 binding = T cell becomes activated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What is an immature T cell called?

A

Thymocyte

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What question must be asked before a thymocyte can be released?

A
  • Does it see self MHC at all?
  • Does it see self MHC too much?
  • Does it react with self-peptides?
  • Will it fight intracellular or extracellular infections?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Why is a mechanism for repertoire selection and self-tolerance needed?

A
  • Generation of the TCR repertoire involves many random mechanisms
  • The specificity of TCR in the immature repertoire is also random and will include cells with receptors that are harmful (self-antigen recognition), useless and useful (foreign antigen recognition)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Do processing pathways distinguish between self and non-self?

A

No

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What type of antigen is most presented by an antigen presenting cell?

A

Self-antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What is the thymus?

A
  • Lobulated structure with a stroma of epithelial cells and connective tissue
  • Stroma provides microenvironment for T cell development and selection
  • Lobules differentiate into an outer cortex and inner medulla, both filled with bone-marrow-derived thymocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What cells are in the cortex of the thymus?

A
  • Thymocyte
  • Cortical epithelial cell
  • Immature double positive or double negative thymocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What cells are in the medulla of the thymus?

A
  • Thymocyte
  • Dendritic cell
  • Macrophages
  • Medullary epithelial cells
  • Mature single positive or single negative thymocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

How are harmful T cells removed?

A

Negative selection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

How are useless T cells removed?

A

Neglect - no signal sent to keep it alive

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What selection occurs to useful T cells?

A

Positive selection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What is required for T cell maturation?

A

Thymus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What happens to T cells if the thymus is removed from an adult and a neonate?

A
  • Adult will have mature T cells within its body - we need the thymus to develop/mature T cells not to maintain them – bone marrow supplies T cells and they mature in the thymus
  • Neonates will have no T cells as the T cells have not yet left the thymus in neonates
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What percentage of T cells will die in the thymus as a result of stringent processing to find the “useful” T cells?

A

Approximately 98%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What gets rid of the apoptotic thymocytes?

A

Thymic macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What is the flow order of T cell development?

A
  • Double negative
  • Large double positive
  • Small double positive
  • Either single positive (with CD4) or single positive (with CD8)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What are the TCR associated molecules and coreceptors of double negative T cells?

A
  • CD3/TCR-

* CD4-/CD8-

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What are the TCR associated molecules and coreceptors of large double positive T cells?

A
  • CD3+
  • TCR+
  • CD4+/CD8+
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What are the TCR associated molecules and coreceptors of small double positive T cells?

A
  • CD3+
  • TCR+
  • CD4+
  • CD8+
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What are the TCR associated molecules and coreceptors of single positive T cells (with CD4+)?

A
  • CD3+
  • TCR+
  • CD4+
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What are the TCR assocaited molecules and coreceptors of single positive T cells (with CD8+)?

A
  • CD3+
  • TCR+
  • CD8+
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

How does the thymus choose which of the cells entering the thymus are useful, harmful or useless?

A
  • Goes through both positive and negative selection

* If lives through both it is useful

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What is positive selection?

A

Retention of thymocytes expressing TCR that are restricted in their recognition of antigen by self MHC i.e. selection of the useful

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What is negative selection?

A

Removal of thymocytes expressing TCR that either recognise self-antigens presented by self MHC or that have no affinity for self MHC i.e. selection of the harmful and the useless

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What is MHC restriction?

A
  • Antigen can be seen by the TCR only in the context of an MHC molecule
  • TCR will not bind to an MHC molecule unless there is an antigen in the groove
  • In the presence of antigen, the TCR must have some affinity for the MHC molecule
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What do studies in bone marrow chimeras show in the thymus?

A

MHC restriction is learnt in the thymus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What MHC haplotype will T cells recognise in bone marrow chimera experiment?

A
  • Mice of a particular MHC haplotype only make T cells restricted by that haplotype
  • Bone marrow must contain potential to make T cells restricted by A and B MHC molecules (cross)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What does irradiation prevent in bone marrow chimera?

A
  • Prevents the bone marrow from generating lymphocytes

* Irradiation destroys the immune system but has no effect on the epithelial or dendritic cells of the thymus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

How is MHC restriction in the thymus learned?

A

By positive selection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What cells are restricted by MHC type of the thymus that they mature in, in bone marrow chimera?

A

Peripheral T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

How is neglect achieved in useless thymocyte cells?

A
  • Peptide is not recognised or irrelevant

* Thymocyte receives no signal, fails to be positively selected and dies by apoptosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

How is positive selection achieved in useful thymocyte cells?

A
  • Peptide is a partial agonist
  • Thymocyte receives a partial signal and is rescued from apoptosis i.e. the cell is positively selected to survive and mature
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

How is negative selection achieved in harmful thymocyte cells?

A
  • Peptide is an agonist

* Thymocyte receives a powerful (full) signal and undergoes apoptosis i.e. the cell is negatively selected and dies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

How accurate is the positive selection model?

A
  • Relied on very complex chimera experiments
  • Relied on proof of MHC restriction as an outcome which is tested in an ‘unnatural’ response using MHC mismatched presenting cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

How accurate is the negative selection model?

A

Relied on exceptionally powerful superantigens operating outside the normal mechanisms of antigen recognition

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

In a TCR from MHC class I restricted T cell, which type of cells mature?

A

CD8

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

In a TCR from MHC class II restricted T cell, which type of cells mature?

A

CD4

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What are the models for double positive to single positive transition?

A
  • Instructive model

* Stochastic/selection model

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What is the instructive model?

A

Signal from CD4 silences the CD8 expression and vice versa

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What is the stochastic/selection model?

A

Cells randomly inactivate CD4 or CD8 gene, whilst testing a match of TcR restriction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What is the differential effect on the mature and immature repertoire in deletion of cells in the thymus?

A
  • TcR from T cell specific for hen egg lysosome (HEL)
  • Approximately 100% of T cells express anti-HEL TcR
  • Thymocytes activated by antigen in the thymic environment die
  • T cells activated by antigen in the periphery proliferate = evidence for negative selection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What occurs to thymocytes in negative selection when in the presence of ecotopic expression of host peptides?

A

Development of regulatory T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What are the consequences of AIRE mutation?

A
  • Human disease

* Mouse knockout

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What human disease is a result of AIRE mutation?

A
  • Autoimmune polyendocrinopathy with candidiasis and ectodermal dysplasia (APECED, also called autoimmune polyendocrine syndrome (APSS-1)
  • Associated gene identified by positional cloning, named AIRE (“autoimmune regulator”)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What is mouse knockout?

A
  • Autoantibodies against multiple endocrine organs, retina

* Failure to express many self-antigens in the thymus -> failure of negative selection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

How are self-peptides presented in the thymus?

A
  • AIRE (autoimmune regulator) is a regulator of gene transcription that stimulates thymic expression of many self-antigens which are largely restricted to peripheral tissues
  • Discovered as the genetic cause of a human autoimmune disease APS-1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

Summarise positive selection

A
  • Bone marrow chimeras show that MHC restriction is learnt in the thymus
  • T cells are ‘educated’ in the thymus to recognise antigens only in the context of self MHC
  • MHC restriction is learnt in the thymus by positive selection
  • The MHC haplotype of the environment in which T cells mature determines their MHC restriction element
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

Summarise negative selection

A

• Two different models can explain the choice whether to become CD4 or CD8 single positive T cells
• AIRE causes thymic epithelial cells to produce many self-proteins
o But not proteins that are in immune privileged sites
o These are captured and presented by professional APCs in the thymic medulla
• Mutations in AIRE lead to failure in negative selection and profound autoimmunity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What is the significance of tolerance?

A
  • All individuals are tolerant of their own antigens (self-tolerance); breakdown of self-tolerance results in autoimmunity
  • Therapeutic potential
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What is the therapeutic potential of tolerance?

A

Inducing tolerance may be exploited to prevent graft rejection, treat autoimmunity and allergic diseases, and prevent immune responses in gene therapy, perhaps stem cell transplantation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What is “clonal ignorance”?

A

Mature self-reactive lymphocyte clones do not encounter or respond to self-antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What does anergy in peripheral tolerance result in?

A

Functional unresponsiveness as anergy inactivates T cells by turning off signals with APCs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What does deletion in peripheral tolerance result in?

A

Apoptosis after activating cell with APCs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

What does suppression in peripheral tolerance result in?

A

Block in activation by regulatory T cells between APCs and T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What does T cell anergy in peripheral tolerance do?

A

Either blocks signal or engages inhibitory receptors (e.g. CTLA-4)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What are the features of anergy in peripheral tolerance?

A
  • Induced by self-antigens that are displayed to the immune system without inflammation or innate immune response (prolonged signal 1, i.e. antigen, inadequate signal 2)
  • Recognition of such antigens may lead to signalling block and/or engagement of inhibitory receptors
  • Role of anergy in self-tolerance in humans is unclear (therapeutic potential?)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What does deletion in peripheral tolerance do?

A

Either activation induced cell death (engagement of death receptors) or activation induced cell death (expression of pro-apoptotic proteins) both ending in apoptosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What are the features of deletion in peripheral tolerance?

A

Stimulation of T cells by self-antigen triggers apoptosis by engagement of death receptors (death receptor pathway) or imbalanced expression of pro-apoptotic proteins (mitochondrial pathway)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What evidence is there to support the importance of ACID in maintenance of self-tolerance?

A
  • Mice with mutations in Fas or Fas ligand develop a lupus-like autoimmune disease
  • Humans with mutations in Fas or enzyme involved death receptor-induced apoptosis (caspases): the autoimmune lymphoproliferative syndrome (ALPS)
  • Eliminating both death pathways in mice -> “spontaneous” systemic autoimmune disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

What does suppression in peripheral tolerance do?

A
  • Recognition of self-antigen in either thymus or peripheral tissues by regulatory T cells
  • Inhibit T cell activation (dendritic cell with naïve T cell) or inhibit T cell effector functions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What are the features of suppression in peripheral tolerance?

A
  • Regulatory T cells are CD4+ cells that express high levels of CD25 (IL-2 receptor alpha chain)
  • Generated by self-antigen recognition in the thymus or peripheral tissues
  • Generation requires a transcription factor called Foxp3 (mutations in Foxp3 are the cause of a severe autoimmune disease in humans and mice
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What are the mechanisms of action in suppression in peripheral tolerance?

A
  • May be multiple
  • Secretion of immune-suppressive cytokines (IL-10, TGFβ) – inhibits macrophages, dendritic cells and interferon gamma
  • CTLA-4 on Tregs blocks B7 on APCs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What is the significance of suppression in peripheral tolerance with regards to self-tolerance?

A
  • Some autoimmune diseases may be associated with defects in regulatory T cells or resistance of responding cells to suppression
  • Therapeutic potential of cellular therapy (autoimmune diseases, graft rejection etc)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What is the largest immunologic organ in the body?

A

The gastrointestinal tract

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

In the GIT what might take up antigens in oral tolerance?

A
  • Peyer’s patches
  • Dendritic cells
  • Epithelial cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What is the most important immune cell in oral tolerance induction?

A

Regulatory T cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What factors can influence oral tolerance induction?

A
  • Antigen related
  • Inherent to the host
  • Disturbances at different steps can be described in food hypersensitivity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

The introduction of antigens to the body has different routes with distinct outcomes. What routes lead to immunity?

A
  • Subcutaneous, intramuscular and intradermal

* Infection/injury

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

The introduction of antigens to the body has different routes with distinct outcomes. What routes lead to tolerance?

A
  • Intravenous
  • Portal vein
  • Anterior chamber of the eye
  • Mucosal
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

What can oral feeding of antigen induce?

A

An active (but inhibitory) immune response that is mediated by T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

What factors allow oral tolerance to be multifactorial?

A
  • Induction of clonal anergy
  • Induction of clonal deletion
  • Induction of T regulatory cells which produce suppressor cytokines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What does a low dose oral tolerance mean?

A

Immunoregulation – some immune cells prevent others from responding to antigen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

What does a high dose oral tolerance mean?

A

Anergy/deletion – the cells themselves lose the ability to respond to the antigen used to induce oral tolerance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

Compare use of antigen between oral tolerance and vaccines?

A
  • Tolerance has a soluble antigen

* Vaccines have an antigen plus adjuvant, particulate antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

Compare number of dosages between oral tolerance and vaccines?

A
  • Tolerance has repeated doses (prolonged and sustained)

* Vaccines have a limited number of vaccine doses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

Compare amount of dosage between oral tolerance and vaccines?

A
  • Bolus dose of 20-500mg or multiple injections of 1mg

* Vaccines have a low antigen dose, typically in ug concentrations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What are some types of viral infections?

A
  • Encephalitis/meningitis
  • Eye infection
  • Common cold
  • Parotitis
  • Pharyngitis
  • Gingivostomatitis
  • Cardiovascular
  • Pneumonia
  • Myelitis
  • Hepatitis
  • Pancreatitis
  • Gastroenteritis
  • Skin infections
  • Sexually transmitted diseases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What diseases are linked (not all fully confirmed) to infection?

A
  • Alzheimer’s disease
  • Asthma
  • Atherosclerosis
  • Autoimmune diseases
  • Cancer coronary heart disease dementia
  • Stroke etc
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What autoimmune diseases are linked (not all fully confirmed) to infections?

A
  • Crohn’s
  • Lupus
  • Multiple sclerosis
  • Psoriasis etc
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

What does Epstein Barr Virus cause?

A
  • Autoimmune diseases
  • Breast cancer
  • Oesophageal cancer
  • Hodgkin’s lymphoma
  • Chronic obstructive pulmonary disease
  • Seasonal affective disorder
  • Lupus multiple sclerosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

How many amino acids is the class I epitopes of antigens?

A

8-10

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

How many amino acids is the class II epitopes of antigens?

A

18-20

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What are some features of viruses?

A
  • Simple structure (subcellular level)
  • Obligate intracellular agents – unable to replicate outside cells, enter cells via receptors
  • Induce diseases through damage of cell in which they replicate or induction of the immune response
  • Infection can be either acute or chronic (active and latent)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What are the mechanisms of the innate immunity as a defence against viruses?

A
  • Inhibition of infection and induction of antiviral state type I interferons (IFN-α and β)
  • Killing of infected cells (NK cells)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

How is antiviral action of type I interferons done?

A

Once a cell is infected with a virus it will secrete type I interferons which leads to the upregulation of enzymes that inhibit replication (non-specific) of uninfected cells with IFN receptors protecting it from infection. The type I interferons also increase expression of class I MHC molecules which promotes cytotoxic T cells to kill the infected cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What happens in the destruction of infected cells by NK cells when the inhibitory receptor is engaged?

A

NK cell binds to a normal autologous cell by the activated receptor to the ligand for NK cell and the inhibitory receptor that binds to the self-class I MHC – self peptide complex. This leads to the NK cell not activating so the normal autologous cell is not killed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

What happens in the destruction of infected cells by NK cells when the inhibitory receptor is not engaged?

A

NK cell only binds to the ligand for NK cell of the virus infected cell (class I MHC negative – virus inhibits class I MHC expression). the NK cells are activated and kills the infected cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What are the mechanisms of the adaptive immunity as a defence against viruses?

A

• Humoral immunity – B cells and antibody lead
o Neutralization (IgG and IgA)
o Antibody-dependent cell mediated cytotoxicity (IgG)
o Opsonization (IgG)
• Cell mediated immunity – CD8+ and CD4+ T cells lead
o Killing of infected cells (CD8+ T cells)
o Activation of CD8+ T cells and B cells (CD4+ helper T cells)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

How does neutralization of viruses occur without antibodies present?

A
  • No neutralization
  • Microbe can penetrate the cells to the epithelial cells where it can bind to tissue cell receptor and infect the tissue cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

How does neutralization of viruses occur with antibodies present?

A
  • Neutralization

* Antibody blocks binding of microbe to the cells so they cannot bind to the cell and infect them

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What are the protective mechanisms of antibodies?

A
  • An antibody can bind to the virus and C1q can bin to the antibody. This activates complement with complement components binding to complement receptors on the macrophage allowing phagocytosis
  • An antibody can bind to the Fc receptor on a macrophage blocking the binding of the virus to that Fc receptor allowing phagocytosis
  • Antibodies can bind to the virus blocking the uptake through cellular receptors on target cells
  • Antibodies can signal to NK cells that they have bound to a virus on a (infected) target cell. The NK cell then kills the target cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What is the mechanism of killing by CTLs (cytotoxic leukocytes)

A
  • Antigen recognition and conjugate formation
  • CTL activation
  • CTL granule exocytosis
  • Apoptosis of target cell
  • Granzymes enter cell via receptor-mediated endocytosis and enter cytoplasm via perforin-dependent mechanisms -> activation of apoptotic pathways
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

How does an NK cell kill?

A
  • Either by perforin and granzymes resulting in apoptosis

* Or FasL and Fas receptor binding resulting in apoptosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What are some mechanisms of immune evasion of viruses?

A
  • Antigenic variation (influenza, HIV …)
  • Inhibition of antigen processing and presentation (many viruses)
  • Inhibition of immune response (many viruses)
  • Infection of immune cells (HIV…)
  • Establishment of latency (HSV, HIV…)
  • Inhibition of apoptosis (Herpes and Pox viruses …)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

How has the Influenza virus gone through antigenic variation?

A
  • Influenza infected humans and birds

* An epidemic strain that affects both birds and humans then infect a pig with a pandemic strain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

How does a virus inhibit antigen processing and presentation?

A

• They can inhibit the class I MHC pathway of antigen processing
o They inhibit the proteasomal activity (done by EBV and human CMV)
o Block the TAP transport (HSV)
o Block MHC synthesis and/or ER retention (adenovirus or human CMV)
o Removal of class I from ER (CMV)
o Interference with CTL recognition by “decoy” vital class I-like molecules (murine CMV)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

How can viruses inhibit immune responses?

A

Through production of virokines and viroreceptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

How does Epstein Barr Virus inhibit the immune response?

A

EBV infected B lymphocyte inhibits macrophage activation by secreting IL-10 and binding to the receptor on the macrophage inhibiting it

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

How does Poxx Virus inhibit the immune response of the body?

A

• Pox virus infected tissue produces IL-1 and IFN-γ receptors (viroreceptors) to block IL-1 and IFN- γ activation on the macrophage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What is a potential disadvantage of antigenic variation?

A

Requires mutation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What are the benefits of antigenic variation?

A
  • Extend the length of infection

* Infect hosts with prior exposure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

Why is an extended length of infection a benefit of antigenic variation?

A
  • The new variants allow the virus to stay one step ahead of the immune response as with each new mutant strain the host must generate a new response - Use high mutation rates to stay ahead of the adaptive immune response
  • Some viruses, like hepatitis C virus, escape by evading antibody response or a persistent infection with a high variability in the envelope protein indicating positive selection (that the infected cell in useful)
  • Use antigenic variation to escape surveillance (Trypanosoma brucei achieves this not by mutation but by differential expression)
  • Some viruses persist in vivo by ceasing to replicate until immunity wanes (latency – not transcriptionally active, and causes no disease)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

How is antigenic variation achieved?

A
  • Mutation
  • Recombination
  • Differential expression of archived variants
  • HIV evades CTL responses by changing their dominant epitopes
  • Epitopic space allows for a higher capacity for infection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

How does CTLs kill in a normal immune response?

A
  • MHC molecules present antigen from cytosol

* Cytotoxic T cell recognises complex of viral peptide with MHC class I and kills infected cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What peptides does class I present?

A

Cell made intracellular peptides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What peptides does class II present?

A

Found exogenous peptides from the environement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

How do viruses escape CTLs?

A

Amino acid substitutions of the epitopes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

How can amino acid substitutions of the epitopes allow them to escape recognition?

A
  • Can interfere with processing and transport of peptides
  • Can reduce binding to MHC molecules
  • Can reduce the affinity of TCR receptor binding
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

How can CTL escape be done by interfering with processing/transport?

A

A study of murine leukaemia virus (MuLV – oncogenic retrovirus) showed that a single amino acid substitution in a viral peptide can alter cleavage pattern and hence epitope presentation and hence CTL response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

What type of mutation is the single amino acid substitution responsible for CTL escape?

A

Single point mutations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

What is the end result of interfering with processing/transport in CTL escape?

A

The epitope is unavailable to MHC and CTL responses to FMR type is weak

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

How can CTL escape be done by reducing MHC binding?

A
  • Mutations that reduce peptide-MHC binding
  • This can either prevent MHC from dragging the peptide successfully to the cell surface, or from holding on to it once there
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

What is an example of reduced MHC binding for CTL escape?

A

• SIV/macaques used as a model for HIV
o An epitope Tat was shown to have mutation in the Tat viral epitope that destroyed binding to the MHC
o Little variation outside of the epitope
o End result - Positive selection to block MHC binding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What is an example of reduced TCR binding to escape CTLs?

A
  • LCMV system shows a single amino acid change that can lead to a decline in affinity for the TCR
  • Tissot et al (2000) showed a Y to F substitution in one immunodominant epitope caused a 100-fold reduction in affinity for the TCR
  • End result – escape mutation that destroy the immune system’s ability to see that viral epitope
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What are some examples of pathogens that sample from a pool of archival genomic variation?

A
  • Trypanosoma brucei
  • Borrelia hermsii
  • Plasmodium falciparum
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What does Trypanosoma brucei cause?

A

Sleeping sickness

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What does Borrelia hermsii cause?

A

Relapsing fever

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

What does Plasmodium falciparum cause?

A

Malaria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

Why do sensory neurons remain infected?

A
  • The virus remains quiescent, few viral proteins are produced and hence there are few virus-derived proteins to present on MHC class I
  • Neurons carry low levels of MHC class I molecules making it harder for CTLs to recognise and kill them
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

Why would neurons have low MHC I expression?

A

Low levels of MHC I expression may be beneficial to the host since it reduces the risk that neurons, which cannot regenerate, will be attacked inappropriately by CTLs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

How does Mycobacterium tuberculosis use antigenic variation to extend the length of infection?

A
  • Resists destruction b host defence mechanisms/exploits them
  • It is taken up by macrophages but prevents the fusion of the phagosome with the lysosome, effectively hiding from antibody-mediated immunity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

What vaccine is a good vaccine in that is has excellent cross-reactivity?

A

Measles virus vaccine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

When does cross-reaction occur?

A

Occurs when the host can use its specific recognition from prior exposure to fight against a later, slightly different antigenic variant

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

What causes antigenic drift?

A

Point mutations in the genes encoding surface proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

What causes antigenic shift?

A

Reassortments leading to novel surface proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

What are features of antigenic drift?

A
  • Seasonal (Influenza)
  • Predictable annual occurrence
  • Residual immunity in population
  • Neutralizing antibodies against hemagglutinin/neuraminidase block binding to cells
  • Mutations alter hemagglutinin/neuraminidase epitopes so that neutralizing antibody no longer binds
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

What are features of antigenic shift?

A
  • Pandemic (Influenza)
  • Unpredictable rare occurrence
  • “Naïve” population
  • Antigenic shift occurs when RNA segments are exchanged between viral strains in a secondary host
  • No cross-protective immunity to virus expressing novel hemagglutinin/neuraminidase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What are the features of primary immunodeficiencies?

A
  • Genetic basis
  • Majority are diagnosed in children
  • Milder forms may not be recognized until adulthood
  • About 1 in 500 people is born with a primary immunodeficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

What is the genetic basis of primary immunodeficiencies?

A
  • Inherited
  • Familial
  • Often monogenic
169
Q

What is monogenic?

A

Often caused by a single gene mutation or deficiency

170
Q

What are the features of secondary immunodeficiencies?

A

• Also known as acquired or immunosuppression
• Defect following exposure to
o Disease/infection
o Drug treatment
o Environmental exposure
• Can also be caused by aging and malnutrition

171
Q

How is primary immunodeficiency varied?

A
  • Ethnic origins

* Phenotype between ethnic origins

172
Q

How does ethnic origins effect primary immunodeficiency disease prevalence?

A

Prevalence in 100,000 inhabitants in the Middle East and North Africa region compared with countries from other world region

173
Q

What is variation in ethnic origins due to?

A
  • Different ethnicities

* Familial practices

174
Q

How is chemotaxis of leukocytes achieved?

A

Leukocyte needs to roll along the surface of the vasculature under the influence of selectins and mucins. Chemokines and chemokine receptors are presented by glycosaminoglycans which activates integrins, firm adhesion and extravasation (migration into tissues)

175
Q

What is opsonization?

A

Enhancing phagocytosis of antigens

176
Q

What is chemotaxis?

A
  • Directional movement

* Attracting macrophages and neutrophils

177
Q

What is cell lysis?

A

Rupturing membranes of foreign cells

178
Q

What is the function of phagocytes?

A
  • Engulf and destroy pathogens using a plethora of microbial agents
  • Participate in antigen presentation of engulfed proteins
179
Q

What is the function of NK cells?

A
  • Deal with intracellular pathogens (shielded from phagocytes and complement)
  • Antiviral, protect from cancer and malignancies
  • Induce apoptosis (by perforin and granzymes) and secrete cytolytic granules
180
Q

What are the functions of dendritic cells?

A
  • Sample environment by pinocytosis (“drinking”) or phagocytosis (“eating”)
  • Resident in tissues and mature upon activation and travel to local nodes
  • In nodes antigen presentation and co-stimulation
181
Q

What are the functions of CD4 T cells?

A
  • TH1 macrophage activating
  • TH2 B cell activating
  • Treg control inflammation
182
Q

What are the functions of CD8 T cells?

A
  • Cytotoxic response to infected cells

* Antiviral and anti-cancer response

183
Q

What are the functions of B cells?

A
  • Memory cells in circulation

* Plasma cells produced in bone marrow produce antibodies

184
Q

What are the functions of IgM?

A
  • Produced early
  • Low affinity
  • Activate complement
185
Q

What are the functions of IgA?

A

Secreted across mucosa to prevent re-infection

186
Q

What are the functions of IgE?

A

Bind to mast cells to clear multicellular parasites

187
Q

What are the functions of IgG?

A
  • Sterilising/neutralising
  • Cross placenta from mother to baby
  • Signal NK cells and phagocytes
188
Q

What does PID stand for?

A

Primary immunodeficiency diseases

189
Q

What is PID?

A
  • They are genetic, familial, and heritable
  • They impact culture and society – predominant in cultures with consanguineous marriage
  • Predisposed to specific infection – teaches us how the immune system works
190
Q

What defects can occur in PID?

A
  • Defects in pattern recognition
  • Defects in phagocytes
  • Defects in migration
  • Defects in complement
  • Defects in B cells
  • Defects in T cells
191
Q

What is the defect in pattern recognition in patients with PID?

A
  • Toll-like receptors (TLRs) are crucial in recognition of pathogen associated molecular patterns (PAMPs)
  • MyD88 is an adaptor required for signalling
  • MyD88 deficiency results in severe life-threatening infection with pyogenic bacteria including pneumococci
192
Q

What is pyogenic?

A

Pus forming

193
Q

What is an example of a defect in phagocytes in patients with PID

A
  • Chronic granulomatous disease and p91 mutation
  • Neutrophils fail to produce reactive oxygen species (superoxide – phorbol ester)
  • Poor ability to kill
  • Aspergillus fumigatus pneumonitis (an e.g.)
194
Q

What is an example of a defect in migration in PID?

A
  • Lack of CD 18 subunit of β2 integrins results in “Leukocyte adhesion deficiency” (LAD) and recurrent bacterial infections
  • Only neutrophils are affected
  • Other leukocytes can migrate using β1 integrins
195
Q

What is an example of defects in complement in PID?

A
  • Defective terminal membrane attack complex proteins (C7, C9 or factor D) all results in Neisseria gonorrhoeae and N. meningitidis
  • Deficiencies in several other components predisposes to development of autoimmune disease
196
Q

What is an example of defects in B cells?

A
•	X-linked agammaglobulinemia that more likely affects males results in grossly depressed Ig levels
o	Cell mediated immune responses normal
o	Pyogenic infections common
•	Selective IgA deficiency
o	Most common of all PIDs
o	Affects circulating and secretory
197
Q

What is an example of defects in T cell?

A
  • Often impact on B-cell function
  • DI-George syndrome due to mutations in TBX1 transcription factor T cell development is disturbed (malformed T cells – 6% of normal level)
  • Hyper-IgM syndrome due to deficiency in transcription factor STAT3
198
Q

What are the features of Hyper-IgM syndrome due to deficiency to STAT3 in defected T cells?

A
  • Decreasing expression of CD40
  • Absence of IgG, IgA and IgE
  • Infections with Toxoplasma
199
Q

What does SCID stand for?

A

Severe combined immunodeficiencies

200
Q

What are the features of SCID?

A
  • Absolute failure in T cell development
  • Affects 1 in 8000
  • Profound defects in cellular and humoral immunity
  • Death within first year of life due to severe and recurrent opportunistic infections
  • Vulnerable to vaccination with attenuated organisms (e.g. rota virus vaccine)
201
Q

What are some causes of SCID?

A
  • T-B-NK+ SCID – RAG-1, Rag-2
  • T-B-NK- SCID - ADA
  • T-B+NK+ SCID – IL-7Rα chain
  • T-B+NK- SCID – gamma-c chain – cytokine signalling pathways (IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21)
  • RD – AK2
  • Artemis
  • JAK-3
202
Q

What are the causes of secondary immunodeficiencies?

A
•	Infection
o	HIV
o	Measles – increased IL-12 production by virus protein cross-linking CD46
•	Side-effects
o	X-rays
o	Cytotoxic drugs
o	Malignancy
203
Q

What tests are used to identify immunodeficiencies?

A
  • ELISA – be careful of children (immunoglobulins aren’t all present at birth)
  • Test humoral response
  • Phenotype lymphocytes by flowcytometry
  • Sheep red blood cell lysis – complement
204
Q

What therapies are available for immunodeficiencies?

A
  • Early intervention with antibiotics, antifungals, and antivirals
  • Replace missing components
  • Gene therapy
  • Transplantation
205
Q

What are some examples of replacing missing components in therapies for immunodeficiencies?

A
  • Adenosine deaminase deficiency due to SCID – enzyme can be injected weekly
  • Defects in humoral response – pooled Ig from several donors will contain antibodies to many common infections
206
Q

How is gene therapy done?

A

Retroviral gene therapy vector into a packaging line inserted into the host cell

207
Q

Is gene therapy a success?

A
  • There are some successes and some failures

* In 2008 in a study for SCID half of the patients developed malignancies and one of them died

208
Q

What are some features of transplantation as a therapy for immunodeficiencies?

A
  • Replace entire immune system with stem cells from healthy donor
  • MHC polymorphism is limiting factor – risk of graft-vs-host disease
  • Radioblation or cytotoxic drugs may be required to minimise host-vs-graft disease
209
Q

What are the features of haematopoietic stem cell transplantation?

A
  • Conditioning regime of chemo/radio therapy
  • Infused with donor haematopoietic stem cells
  • Immunodeficiency improves as their immune system is replaced by donor lymphoid cells
210
Q

What are the features of graft vs host disease (GvHD)?

A
  • Characteristic targets of liver, skin, and gut
  • Graded according to severity from 0-IV
  • Incidence of stage II-IV is 35-50% (5500/year)
  • T cell mediated allo-immune response
211
Q

What is the criteria for a GvHD?

A
  • The donor graft contains immune competent cells
  • There is an immunological disparity between donor and host
  • The host immune system is incapable of mounting a response
  • Donor effector cells must be able to migrate to the host target tissue
212
Q

What is the three phases model of GvHD?

A
  • Commonly used to describe the pathophysiology of acute GvHD
  • Phase 1 occurs prior to transplantation, the conditioning regime of chemo and/or radio therapy causes damage to the target organs which leads to the production of pro-inflammatory cytokines or ‘danger signals’
  • Phase 2 occurs after transplantation, host antigen presenting cells activate donor T cells leading to activation and proliferation of donor T cells.
  • Phase 3 is the effector phase where the donor T cells migrate to the host tissues and cause damage by a variety of mechanisms
213
Q

Why in phase 2 of GvHD does activation and proliferation of donor T cells occur?

A

Because donor T cells are presented with antigens which appear foreign to them due to the genetic differences between the donor and the recipient

214
Q

What is the current clinical management of GvHD?

A
  • Immune suppression
  • Treatment with steroids
  • Regulatory T cells have been shown to prevent GvHD in mouse models and more recently clinical trials
215
Q

What is the difference between GvHD and HvGD?

A
  • In GvHD mature T cells from graft recognise host cell as foreign
  • In HvGD mature T cells in host recognise graft cells as foreign
216
Q

What is the hallmark of a successful graft for immunodeficiency?

A

No immune response by T cell depleted graft. Stem cells proliferate and reconstitute host immune system

217
Q

What is the demographic of the HIV pandemic?

A
  • Killed 25 million by 2008
  • More than 35 million infected
  • 3 million new cases in 2008 alone
  • 14 million children orphaned
  • Females over 16 account for >50% of cases
  • Young people (15-24) account for 34% of cases
  • First reported case in 1981
  • Syndrome characterised with predisposition
  • HIV-1 isolated in 1983
218
Q

What are the predispositions of HIV?

A
  • Opportunistic infections
  • Aggressive form of Kaposi’s sarcoma
  • Concurrent depletion of CD4 cells
219
Q

What are the categories of HIV?

A
•	HIV-1
o	M
o	O
o	N
o	P
•	HIV2
o	Eight groups
o	Less virulent
220
Q

Where is the prevalence for HIV?

A
  • HIV-1M is spread worldwide (subcategories A-K)

* HIV1-O/N/P and HIV-2 is predominant in West Africa

221
Q

What is the clinical course of HIV?

A
  • Infection
  • Acute viremia
  • Latency
  • Early AIDS
  • Terminal AIDS
222
Q

What can infect someone with HIV?

A
  • Exposure to bodily fluids
  • Sexual intercourse (most common)
  • Intravenous drug delivery (sharing of needles)
  • Blood/blood products
  • Breastfeeding (25% perinatal transmission)
223
Q

What bodily fluids can infect someone with HIV?

A
  • Semen
  • Vaginal fluid
  • Milk
  • Blood
224
Q

How is someone infected with HIV?

A
  • Intraepithelial dendritic cells bind HIV using DC-SIGN
  • HIV is internalized into early endosomes
  • Dendritic cells that have migrated to lymph nodes transfer HIV to CD4 T cells
225
Q

What occurs in acute viremia of HIV?

A
  • 2-8 weeks after infection, explosion of viral replication (particularly in the gut)
  • Symptoms occur
  • Spontaneously resolves after 1-4 weeks
226
Q

What are the symptoms during acute viremia of HIV?

A
  • High fever
  • Sore throat
  • Headaches
  • Swollen lymph nodes
  • Diarrhoea
227
Q

How is HIV spontaneously resolved in acute viremia?

A
  • Strong HIV specific CD8 killing of infected cells

* Production of HIV-specific antibodies

228
Q

What occurs in latency of HIV?

A
  • CD4 levels rebound but still low (800/ml compared with 1200/ml in healthy)
  • Baseline level of virus in the blood – a “set point” which is best indicator of individual prognosis
  • Gradual decline in number and function of CD4 cells
  • Asymptomatic for 2-15 years
229
Q

How does the number and function of CD4 cells decline in latency of HIV?

A
  • Direct cytopathic effect of viral proteins on host cell
  • Infected cells have increased susceptibility to apoptosis
  • Elimination of infected CD4 cells by CD8 cells
230
Q

What occurs in early AIDS within the clinical course of HIV?

A
  • Number of functional CD4 drops below 400cells/ml
  • A drop below 200cells/ml is classified as AIDS
  • Opportunistic infections appear to exploit weakened immune system (e.g. early Candida (oral thrush) and M. tuberculosis)
231
Q

What does AIDS stand for?

A

Acquired immunodeficiency syndrome

232
Q

What occurs in terminal AIDS within the clinical course of HIV?

A
  • Reactivation of latent infections
  • Fungal pneumonia – highly fatal without treatment
  • Mycobacterium avium and cytomegalovirus – treatment resistant environmental infections
  • Respiratory infections are the major cause of death
233
Q

What latent infections can be reactivated in terminal AIDS?

A
  • Shingles (chicken pox)
  • B cell lymphomas (Glandular fever, EBV)
  • Kaposi’s sarcoma (Herpes)
234
Q

What is the structure of HIV?

A
  • Retrovirus – RNA genome but replication passes through DNA with involvement of reverse transcriptase
  • Lentivirus – prolonged course of disease
235
Q

How many genes does the HIV genome have?

A

10

236
Q

What are the genes of HIV genome?

A
  • LTR (long terminal repeat)
  • gag (Pr55gag)
  • pol (polymerase)
  • vif (viral infectivity factor (p23))
  • vpr (viral protein R (p15))
  • rev (regulator of viral gene expression (p14))
  • tat (transcriptional activator (p14))
  • vpu (viral protein U)
  • env (gp160 envelope protein)
  • nef (negative effector (p24))
237
Q

How does HIV enter the cells?

A
  • Envelope spike which engages CD4 on – T lymphocytes, macrophages and dendritic cells
  • Binding event changes shape of gp120 to form a binding site for coreceptors CCR5 and CXCR4
  • Leads to gp41 (highly hydrophobic) acting like a harpoon to destabilise T cell membrane and promote fusion of virus to cell
238
Q

How is HIV integrated?

A
  • Genome consists of 2 single stranded RNA molecules
  • Reverse transcriptase has no proof reading – 1 mutation per genome per reverse transcription
  • Converts to DNA in cytoplasm then translocates to nucleus and preferentially targets areas of active transcription (but can integrate in latent areas)
  • Viral sequence now part of genome host cell
239
Q

How is HIV replicated?

A

• Production of viral transcripts by RNA polymerases and transcription factors of host cell
o Activation of T vells results in expression of NFkB transcription factor, this also binds HIV promotor
• Viral proteins expressed in sequence as needed by alternative splicing of transcripts
• New viron assembled at plasma membrane

240
Q

How is HIV tested for?

A
  • 3rd generation tests
  • 4th generation tests
  • PCR testing
241
Q

What are the features of 3rd generation tests of HIV?

A
  • Tests for anti-HIV antibodies
  • Good for long term screening
  • Can give false negatives during window period of initial infection (3 weeks – 2 months, prior to seroconversion, production of anti-HIV antibodies)
242
Q

What are the features of 4th generation tests of HIV?

A
  • Tests for anti-HIV antibodies
  • P24 antigen (rises sharply during primary infection)
  • Shorter window period
  • 20 samples are pooled and then singly tested if any are positive
243
Q

What are the features of PCR testing of HIV?

A
  • Common in USA as alternative to 4th generation (window period 5 days shorter)
  • Pools of 90 together, then batches of 10 then singly to identify patients
  • Test more expensive
244
Q

What are some potential drug targets in the virus life cycle from most potential to least?

A
  • Reverse transcriptase (current clinical use)
  • Entry (CCU)
  • Fusion (CCU)
  • Integration 9CCU)
  • Protein production (CCU)
  • Assembly
  • Maturation
245
Q

What are the features of HIV therapeutics?

A

Potential drug targets in the virus life cycle

246
Q

What potential drug targets are in HIV therapeutics?

A
  • Entry, maraviroc (oral, blocks co-receptor CCR5)
  • Fusion, enfuviritide (rescue drug, twice daily sub cut, expensive, binds gp41)
  • Reverse transcription, nucleotide analogues or allosteric inhibitors
  • Integration, raltegravir (oral twice daily)
  • Protein production, saquinavir (oral)
247
Q

What does each person vary with drug efficacy?

A
  • Error prone nature of virus
  • Large viral load
  • Rapid rate of replication
248
Q

What is the drug efficacy and resistance in HIV drugs?

A
  • Each person has many variants
  • Some protease inhibitors rendered ineffective by single mutation
  • Others (e.g. zidouvudine) require multiple mutations
  • Most effective to give a “cocktail” of drugs with different mechanisms
249
Q

What does a HIV vaccine need to be?

A
  • Effective – must evoke protective levels of immunity
  • Available
  • Stable – preferably not requiring refrigeration
  • Cheap
  • Safe – eliminate pathogenicity
250
Q

What does current research on HIV aim to generate?

A
  • “blunting” antibodies to protect against disease rather than infection
  • Strategies to increase cellular immune responses and reduce viral set point
251
Q

What are the four main cancers in the UK?

A
  • Breast
  • Lung
  • Large bowel
  • Prostate
252
Q

What rank is cancer in the cause of mortality in the UK?

A

2nd

253
Q

What is cancer caused by?

A

Progressive growth of the progeny of a single transformed cell

254
Q

What is the simplified process of tumour cell growth?

A
  • Initial modified tumour cell
  • The modified tumour cell begins to proliferate into a localised benign tumour
  • The tumour cells become invasive into the tissues
  • The tumour cells invade blood vessels allowing metastasis to occur
255
Q

What is metastasis?

A

Spread of cancer to different parts of the body travelled through the blood or lymph

256
Q

What does curing cancer require?

A

Removal or destruction of all malignant cells without harming the patient

257
Q

What is the leading cancer with the most cases in men?

A

Prostate

258
Q

What is the leading cancer with the most cases in women?

A

Breast

259
Q

What is the leading cancer with the most deaths in men and women?

A

Lung and bronchus

260
Q

Which cancer has the highest survival rate?

A

Prostate - 99%

261
Q

Which cancer has the second highest survival rate?

A

Melanoma of the skin - 91%

262
Q

Which cancer has the lowest survival rate?

A

Pancreas - 4%

263
Q

What can be the causative agents of cancer?

A
  • Chemical carcinogens
  • Spontaneous
  • UV and ionizing radiation
  • Virus-induced
  • Genetic abnormalities
  • Immunosuppression
264
Q

What are some examples of chemical carcinogens that cause cancer?

A
  • Formaldehyde
  • Ethanol
  • Methanol (in antifreeze)
265
Q

What are some examples of spontaneous events that cause cancer?

A
  • Random mutation event
  • Cell division
  • Cell proliferation
266
Q

What are some examples of UV and ionising radiation that cause cancer?

A
  • X-rays

* Sun exposure

267
Q

What are some examples of virus-induced cancer causing agents?

A
  • Hepatitis C
  • Epstein Barr Virus
  • Human papillomavirus
268
Q

What are some examples of genetic abnormalities causing cancer?

A
  • Xeroderma pigmentosum (XP)

* Families can develop errors in DNA proofreading

269
Q

What is xeroderma pigmentosum (XP)?

A

Decreased ability to repair DNA damage such as that caused by UV light

270
Q

What is the characteristic of normal cells regarding cell renewal and cell death?

A
  • Mature cells have a given life span

* Replaced by proliferation and differentiation of stem cells

271
Q

What disrupts normal healthy cells to make cancerous cells?

A
  • Chemical
  • Physical
  • Viral agents
  • All that mutate the DNA
272
Q

What are the types of oncogenes?

A
  • v-onv (virus oncogenes)

* c-onc (cellular oncogenes)

273
Q

What are oncogenes?

A

A gene that has the potential to cause cancer

274
Q

What type of gene is affected by mutations that induce tumour formation?

A
  • Induce cellular proliferation
  • Inhibit cellular proliferation
  • Regulate programmed cell death
275
Q

What gene induces cellular proliferation?

A

Proto-oncogenes

276
Q

What gene inhibits cellular proliferation?

A

Tumour suppressor genes

277
Q

What gene regulates programmed cell death?

A

Tumour suppressor genes

278
Q

Why can cells not proliferate indefinitely?

A

Limited availability of growth factors

279
Q

What is a feature of benign tumours?

A
  • Do not grow indefinitely

* Do not significantly invade surrounding tissue

280
Q

What is a feature of malignant tumours?

A
  • Become progressively invasive

* Can metastasise

281
Q

Where are carcinomas derived from?

A

Endodermal and ectodermal tissues

282
Q

Where are leukaemia and lymphomas derived from?

A

Hematopoietic cells

283
Q

Where are sarcomas derived from?

A

Mesodermal connective tissue

284
Q

What is the development of a normal cell to a metastatic cell?

A
  • A multi-step process of clonal evolution driven by several mutations
  • It is a sequential discreet evolution of cancer
285
Q

What is the flow order of the development of a normal cell to a metastatic cell?

A
  • Normal
  • Hyperplastic
  • Dysplastic
  • Neoplastic
  • Metastatic
286
Q

How are cancer cells different?

A
  • Clonal in origin
  • Deregulated growth and lifespan
  • Altered tissue affinity
  • Resistance to control via apoptotic signals
  • Change in surface phenotype and markers
  • Structural and biochemical changes
  • Presence of tumour-specific antigens
287
Q

What differences of cancer cells to normal cells are responsible for an immune attack?

A
  • Change in surface phenotype and markers
  • Structural and biochemical changes
  • Presence of tumour-specific antigens
288
Q

When was immune surveillance of cancer proposed and refined?

A
  • Proposed in 1909

* Refined in 1950s

289
Q

Who proposed and refined immune surveillance of cancer?

A
  • Proposed by Paul Ehrlich

* Refined by Burnet and Thomas

290
Q

What was the proposed immune surveillance of cancer?

A
  • “In animals … genetic changes must be common and a proportion … will represent a step towards malignancy.
  • … there should be some mechanism for eliminating such potentially dangerous mutant cells ad it is postulated that this mechanism is of immunological character”
291
Q

When was evidence of immune surveillance of cancer found?

A

2001 by Schreiber et al

292
Q

What did the evidence find about immune surveillance of cancer?

A

IFN-γ and lymphocytes prevent primary tumour development and shape tumour immunogenicity

293
Q

What does more recent studies on immune surveillance of cancer tell us?

A
  • Identify effector populations
  • Knockout models utilised
  • Nude mice studies
294
Q

What is the evidence for immune surveillance in mice?

A
  • Animal models showed that pre-treatment of mice with killed tumour material could protect against a subsequent challenge
  • This is T cell based
295
Q

How were the mice models used in immune surveillance of cancer?

A
  • Immunized mouse with irradiated tumour cells
  • In one mouse inject viable cells of same tumour
  • In another inject viable cells of a different tumour
  • Those with same tumour cells responded with tumour rejection antigens eliminating the tumour
  • Those with different tumour cells did not eliminate tumour
296
Q

What is the evidence that immune surveillance of cancer of T based?

A
  • T cell removal from mice removed protection to tumour

* Transfer of T cells from immunized mouse can protect a naïve mouse from tumour challenge

297
Q

What evidence is there for immune surveillance in humans?

A
  • Immunosuppression leads to increased development of viral-derived tumours
  • Organ transplant
  • High tumour-infiltrating lymphocyte number correlates with improved survival
  • NK cell loss corelates with increased tumour pathogenicity
298
Q

What is an example of viral-derived tumour from immunosuppression?

A

Kaposi sarcoma from herpes in AIDS

299
Q

How are organ transplants evidence for immune surveillance?

A
  • Increases risk of malignant melanoma

* 3-fold higher risk of sarcoma

300
Q

What antigens does a cancer cell have?

A
  • Viral
  • Silent
  • Mutant
  • Immunological escape changes
  • Loss of organ-specific antigen
  • Altered carbohydrate (cell division)
  • Metastatic molecules
301
Q

What part of the immune system do tumour antigens trigger?

A
  • Humoral

* Cell-mediated

302
Q

What tumour antigens are recognised by T cells?

A
  • Antigens only expressed by tumours
  • Mutated normal antigens
  • Antigens expressed at wrong stage of cell growth
  • Overexpressed antigens
303
Q

How does a mutated antigen lead to a new epitope?

A
  • Point mutation in either self-protein or self-peptide
  • Mutation in shape (self-peptide) – immune system recognises it as a new antigen
  • New epitope within cell (self-protein) due to binding of a new peptide to MHC molecule
304
Q

How does an abnormal expression lead to a new epitope?

A
  • In a normal cell either too little expressed or overexpressed
  • Too little results in presentation of antigens novel to the adult immune system
  • Overexpressed leads to more of one peptide being presented, allowing recognition by T cell
305
Q

Why is T cell recognition not likely in a solid organ cancer?

A
  • Normally naïve T cells do not move into tissues – do not see tumour antigens
  • Tumour does not express co-receptors to activate naïve T cells
  • Tumour metastasis to lymph node is too late
  • Tumour mutation could prevent CD8 T cells recognition
306
Q

Which tumour mutations could prevent CD8 T cell recognition?

A

MHC or TAP downregulation

307
Q

Why can T cells recognise cancer cells in haematopoietic cancers?

A
  • Cancerous blood cells encounter naïve T cells
  • Some haematopoietic cells express high levels of B7
  • Not 100% effective
308
Q

How does the adaptive immune response target tumours?

A

• When tumour cell is dying, antigens are released
• Antigens are engulfed by APCs taken to lymph node
• Presented to T cell which stimulate:
o Antibody/ADCC/complement is activated by helper T cells that educate other T/B cells what is occurring
o CTL recognise and destroy tumour cells

309
Q

What are some effector mechanisms against cancer that the body releases?

A
  • Monocyte/macrophage release lytic enzymes and phagocytose necrotic cells
  • Antibody against tumour antigens
  • Initiation of NK cytotoxic responses
  • Induction of tumour specific CD8 T cells
  • Promotes proinflammatory anti-cancer environment
310
Q

What responses are initiated by NK cells as an effector mechanism against cancer?

A
  • Antibody-dependant cellular cytotoxicity (ADCC)

* MHC I surveillance

311
Q

What does induction of tumour specific CD8 T cells do as an effector mechanism?

A

CD8 T cell migrates into tumour site and initiates cytotoxic responses

312
Q

How does the effector mechanism of the body promote pro-inflammatory anti-cancer environment?

A
  • Release cytokines/chemokines (TNFα, IFNs)

* Release antiangiogenic factors

313
Q

How are tumour cells killed by NK or CD8 T cells?

A

1) Class I + Ag on the tumour cell binds to TCR on the CTL
This triggers perforin and granzyme B to be released
Perforates tumour cell
2) Fas (CD95) on the tumour cell bind to FasL on the CTL
This triggers tumour cell death

314
Q

What is the immune response tumour elimination?

A
  • Innate immune response recognises tumour cell establishment
  • NK cells and other effectors recruited to site by chemokines – target tumour growth directly
  • Tumour-specific T cells home to site along with macrophages and other effectors to eliminate tumour cells
315
Q

When does immune response tumour elimination occur?

A

Early in tumour development

316
Q

What is a drawback of immune response tumour elimination?

A

Not all tumour cells will be killed

317
Q

What is one way that ‘clever’ cancer cells use to escape attack?

A

Downregulate MHC I to evade T cells

318
Q

What is the problem for cancer cells with downregulated MHC I on a cancer cell?

A
  • NK cells desire to kill but this is negated by high expression of MHC I on our healthy cells
  • So, the ‘clever’ cancer cells with low expression of MHC I will be killed by NK cells
319
Q

What is evidence of NK cell control of cancer?

A

Maintenance of remission in acute leukaemia’s dependent upon CD56+/CD8α+ NK cells – Lowdell et al (2002)

320
Q

What ways can tumour cells evade the immune response?

A
  • Downregulation of MHC I and TAP

* May provide poor co-stimulation

321
Q

What are the three E’s of immunoediting?

A
  • Elimination
  • Equilibrium
  • Escape
322
Q

What occurs in elimination of immunoediting?

A
  • NK, CTL and CD4 cells kill tumour cells

* Not all

323
Q

What occurs in equilibrium of immunoediting?

A

There is the same number of immune cells to tumour cells

324
Q

What occurs in escape of immunoediting?

A

Cells avoid immunological control and proliferate

325
Q

What mechanisms by which tumours can avoid immune recognition?

A
  • Low immunogenicity
  • Tumour treated as self-antigen
  • Antigenic modulation
  • Tumour-induced immune suppression
  • Tumour-induced privileged site
326
Q

What does low immunogenicity involve?

A
  • No peptide: MHC ligand
  • No adhesion molecules
  • No co-stimulatory molecules
  • No way of cells binding to the tumour
327
Q

What does antigenic modulation involve?

A
  • Antibody against tumour cell-surface antigens induces endocytosis and degradation of antigen. Immune selection of antigen-loss variants
  • Antibody binds to the surface of the tumour which engulfs, and lysis occurs
328
Q

What does tumour treated as self-antigen involve?

A
  • Tumour antigens taken up and presented by APCs in absence of co-stimulation tolerize T cells
  • This induces tolerance in the form of anergy (prolonged signal 1, no signal 2)
329
Q

What does tumour-induced immune suppression involve?

A

Factors secreted by tumour cells inhibit T cell and induces Treg

330
Q

What does tumour-induced privileged site involve?

A

Factors secreted by tumour cells create a physical barrier to the immune system

331
Q

What factors are secreted in tumour-induced immune suppression?

A
  • TGFβ

* IL-10

332
Q

What does TGFβ do?

A
  • Important in the function of regulatory T cells

* Actively inhibits T cell proliferation

333
Q

What does IL-10 do?

A

Actively inhibits maturation of dendritic cells

334
Q

What are the features of an immature dendritic cell?

A
  • Static – keeps position in tissues
  • Constantly phagocytosing
  • Low surface class II MHC
  • No expression of co-stimulatory molecules
335
Q

What are the features of a mature dendritic cell?

A
  • Highly mobile – migrates to lymph
  • Stops phagocytosing
  • High surface class II MHC
  • High expression of co-stimulatory molecules
336
Q

How does an immune tolerance generate with dendritic cells?

A
  • Signal 1 (antigen) is present
  • Signal 2 (co-stimulation – danger signal) – not present
  • Dendritic cell not activated
  • Tumour cells in lymph are perceived not dangerous
  • Anergy (peripheral tolerance)
337
Q

What are the types of tumour associated macrophages?

A
  • M1

* M2

338
Q

What is M1 activated in the presence of?

A

IFN-γ

339
Q

What is M2 activated in the presence of?

A

IL-10

340
Q

What are the features of M1?

A
  • Defence against bacteria
  • Tumour suppression
  • Immune stimulation
341
Q

What are the features of M2?

A
  • Tissue repair and angiogenesis
  • Tumour promotion
  • Downregulation of M1 and adaptive immunity
342
Q

What are the types of anti-cancer therapies?

A
  • Radiation
  • Alkylating
  • Anti-metabolites
  • Natural products
  • Metals
  • New drugs
343
Q

What forms can immunotherapy take?

A
  • Passive transfer

* Active immunization/vaccination/immunotherapy

344
Q

What is passive transfer in immunotherapy?

A

Anti-tumour antibody (a range of monoclonal antibodies)

345
Q

What is active immunization in immunotherapy?

A

Strategies that generate an anti-tumour immune response (normally cytotoxic T cells)

346
Q

What are tumour rejection antigens?

A
  • The antigens expressed by experimentally induced murine tumours
  • Peptides of tumour cell proteins that are presented to T ells by MCH molecules
347
Q

What are the types of antigens that are targets of T cell responses in tumour rejection antigens?

A
  • Antigens not normally expressed on somatic cells

* Antigens that are normally expressed on somatic cells

348
Q

What are some tumour-specific antigens?

A
  • Bcr-abl

* CDK-4/β-catenin

349
Q

What cancer is Bcr-abl associated with?

A

Chronic myeloid leukaemia

350
Q

What cancer is CDK-4/β-catenin associated with?

A

Melanoma

351
Q

What are some testes-specific antigens?

A
  • MAGE 1-3

* NY-ESO-1

352
Q

What cancer is MAGE 1-3 associated with?

A
  • Melanoma

* Colorectal cancers

353
Q

What cancer is NY-ESO-1 associated with?

A

Melanoma

354
Q

What are some differentiation antigens (found in the womb in development)?

A
  • Tyrosinase (TRP-1/2)
  • Melan-A
  • Monoclonal Ab
355
Q

What cancer is Tyrosine (TRP-1/2) associated with?

A

Melanoma

356
Q

What cancer is Melan-A associated with?

A

Melanoma

357
Q

What cancer is Monoclonal Ab associated with?

A

Myeloma

358
Q

What are some tumour-associated antigens?

A
  • MUC-1
  • α-fetoprotein
  • Her-2/neu
  • WT-1
  • Myeloblastin
  • Survinin
359
Q

What cancer is MUC-1 associated with?

A
  • Myeloma
  • Breast
  • etc
360
Q

What cancer is α-fetoprotein associated with?

A
  • Liver
  • Ovaries
  • Testes
  • Lymphoma
  • etc
361
Q

What cancer is HER-2/neu associated with?

A

Breast

362
Q

What cancer is WT-1 associated with?

A
  • Many
  • Leukaemia
  • Solid tumours
363
Q

What cancer is myeloblastin associated with?

A

Leukaemia

364
Q

What cancer is Survinin associated with?

A

Many

365
Q

What are some antigens that are not normally expressed on somatic cells?

A
  • Antigens only expressed in embryo
  • Gene products of mutated or recombined cellular genes
  • Sequestered antigens (cancer-testis family – apart from tumours only expressed in testis)
  • Viral antigens
366
Q

In principle what is the ideal target for immunotherapy?

A
  • Immune response not limited by self-tolerance

* Responses to the antigens are unlikely to inflict damage on normal healthy cells

367
Q

What percentage of colorectal cancers have MAGE-3 expressed?

A

25%

368
Q

What can vaccines containing MAGE-3 induce?

A
  • Both tumour regression and cytotoxic T cell response in patients with advanced melanoma
  • Tumour regression correlates with the number of anti-MAGE-3 CD8 T cells
369
Q

What cancer does human papillomavirus (HPV) cause?

A

Cervical cancer

370
Q

What percentage of American women have contracted at least one strain of HPV by the age of 50?

A

More than 80%

371
Q

What rank is cervical cancer on the deadliest cancers of women?

A

5th

372
Q

How many HPV strains are there?

A

120

373
Q

How many HPV strains cause cervical cancer

A

20

374
Q

Which strains of HPV are the more dominant types in cervical cancer?

A
  • HPV 16

* HPV 18

375
Q

Which oncogenes are responsible for cervical carcinogenesis?

A
  • E7

* E6

376
Q

What does E7 do in cervical cancer?

A

E7 causes uncontrolled cell proliferation as it complexes with Rb gene

377
Q

What does E7 do in cervical cancer?

A

E6 causes the loss of DNA repair function and prevents cell undergoing apoptosis as it binds to and degrades p53 gene protein product

378
Q

What HPV strains does Gardasil protect against?

A
  • HPV 6
  • HPV 11
  • HPV 16
  • HPV 18
379
Q

What vaccine is used for HPV cervical cancer?

A

DNA free virus like particles (VLP) synthesised by self-assembly of the fusion proteins of the major capsid antigen L1 induce strong humoral response with neutralizing antibodies

380
Q

What are the trade names for HPV vaccine for cervical cancer?

A
  • Gardasil

* Cervarix

381
Q

What trials were done for HPV vaccine in cervical cancer?

A

MERCK and GSK using HPV 16 and HPV 18 L1

382
Q

What are some potential tumour antigen targets?

A
  • Tissue specific antigens – melanoma/melanocyte antigens

* Antigens that are widely expressed at very low levels on normal tissues

383
Q

What must be considered when using tissue specific antigens and low level widely expressed antigens?

A
  • Existence of immunological tolerance

* Possibility of inducing autoimmune diseases

384
Q

What are some anti-cancer antibody immunotherapies?

A
  • Naked Mab
  • Immunoconjugates
  • Multistep targeting
385
Q

What does naked Mab trigger?

A
  • Antibody-dependent cell-mediated cytotoxicity (ADCC)

* Complement-dependent cytotoxicity (CDC)

386
Q

What are some immunoconjugates?

A
  • Radioimmunoconjugate (radionuclide binds to antibody)
  • Immunocytokine (cytokine binds to antibody)
  • Immunotoxin (toxin binds to antibody)
  • Antibody-directed enzyme prodrug therapy (ADEPT)
  • Immunoliposome
  • Cellular immunoconjugates (killer cells binding to antibody)
387
Q

What is multistep targeting?

A
  • Biotinylated radioactive ligand binds to streptavidin (amplifier) which is bound to an antigen on the surface of the tumour cell
  • Biospecific MAb (monoclonal antibody) on antibody binds to radionuclide
388
Q

What does Rituximab (Rituxan) target?

A

CD20 cells in Non-Hodgkins Lymphoma found on cancerous B cells

389
Q

When was rituximab approed?

A

1997

390
Q

What type of antibody is rituximab?

A

Chimeric IgG1

391
Q

What is a drawback of rituximab?

A

Kills all B cells - even normal healthy B cells

392
Q

What is rituximab used in?

A
  • Autoimmunity

* Transplantation

393
Q

What does rituximab do?

A

Destroys CD20 cells found on cancerous B cells either by antibody mediated cellular cytotoxicity or complement dependent cytotoxicity

394
Q

How does complement dependent cytotoxicity kill the B cell?

A
  • C3 -> C3b
  • C3b binds to MAC (membrane attack complex) or antibody bound to CD20
  • This activates cell death
395
Q

How does antibody-dependent cell-mediated cytotoxicity kill the B cell?

A
  • FcγR/CR-mediated opsonic phagocytosis – FcγR binds to antibody bound to CD20 – activates phagocytosis cell death
  • Complement receptor on macrophage binds to rituximab bound to antibody bound to CD20 on B cell – activates cell death
396
Q

How does NK cells kill the B cell?

A

Same as macrophage killing but with a NK cell ending in direct lysis with perforin and granzyme B

397
Q

What does Trastuzumab (Herceptin) target?

A

Targets HER-2 (human epidermal growth factor receptor 2) which is expressed at high levels in some breast cancers

398
Q

What antibody is used in Trastuzumab?

A

Humanized IgG1

399
Q

When was Trastuzumab approved?

A

1998

400
Q

What does Trastuzumab do?

A
  • Acts by the inhibition of HER-2 mediated tumour cell proliferation and migration
  • It is only active in cells that overexpress HER-2
401
Q

What percentage of breast cancer patients overexpress HER-2?

A

25%

402
Q

What does Gemtuzumab ozagamicin (mylotrag) target?

A

Targets CD33 which is expressed in 80% acute myeloid leukaemia cases but not in normal hematopoietic cells

403
Q

What antibody is used in Gemtuzumab ozagamicin?

A

Humanized IgG4

404
Q

What is humanized IgG4 linked/bound to in Gemtuzumab ozagamicin?

A

Calicheamicin

405
Q

When was Gemtuzumab ozagamicin approved?

A

2000

406
Q

What dpes Gemtuzumab ozagamicin do?

A
  • Antibody binds to CD33 resulting in a complex which is internalised
  • The calicheamicin derivative is released into the lysosome of the myeloid cell
  • The calicheamicin derivative binds to DNA resulting in the breakage of the strands and apoptosis
407
Q

What does Alemtuzumab (Campath) target?

A

Targets CD52 which is expressed on B cells in patients with B cell chronic lymphocytic lymphoma

408
Q

What antibody is used in Alemtuzumab?

A

Humanized IgG1

409
Q

When was Alemtuzumab approved?

A

2001

410
Q

What does Alemtuzumab do?

A

Acts by antibody-dependant cellular cytotoxicity and complement dependent cytotoxicity

411
Q

What is Alemtuzumab used in?

A
  • Transplants

* Under trail for MS (multiple sclerosis)

412
Q

What does Ibritumomab tiuxetetan (Zevalin) target?

A

CD20 on B cells in Non-Hodgkins Lymphoma

413
Q

What antibody is used in Ibritumomab tiuxetetan?

A

Murine IgG1 90Y conjugate

414
Q

When was Ibritumomab tiuxetetan approved?

A

2002

415
Q

What does Ibritumomab tiuxetetan do?

A
  • Delivery of cytotoxic radiation leading to apoptosis
  • Yttrium 90 decays by emission of Beta particles with a half-life of 64 hours
  • Also induces ADCC and CDC
416
Q

What is Ipilimumab used for?

A

Melanoma treatement

417
Q

What antibody is used in Ipilimumab (Yervoy)?

A

Human monoclonal antibody

418
Q

What treatment is Ipilimumab undergoing clinical trials for?

A
  • Non-small cell lung carcinoma (NSCLC)
  • Small cell lung cancer (SCLC)
  • Metastatic hormone-refractory prostate cancer
419
Q

What does Ipilimumab do?

A
  • Blocks CTLA-4 interaction with B7, preventing anti-inflammatory signalling
  • CTLA-4 is on the T cell and B7 on tumour cell