Immunology Flashcards

1
Q

What are the constitutive barriers to infection?

A
  • Skin
  • Mucosal surfaces
  • Commensal bacteria
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How does the skin act as a constitutive barriers to infection?

A
  • Consists of tightly packed keratinised cells - physically limits colonisation by microorganisms
  • Physiological factors - low pH and low oxygen tension
  • Sebaceous glands - produce hydrophobic oils, lysozymes and ammonia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

How does the mucosal surfaces act as a constitutive barriers to infection?

A
  • Mucus = physical/trapping barrier - contains secretory IgA, lysozymes, antimicrobial peptides and lactoferrin
  • Cilia directly trap pathogens - assisted by physical manoeuvres (sneezing/coughing)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

How does the commensal bacteria act as a constitutive barriers to infection?

A
  • 100 trillion bacteria normally reside at the surfaces
  • Compete with pathogenic bacteria for scarce resources
  • Produce fatty acids and bactericidins - inhibit the growth of many pathogens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What are the cells of the innate immune system?

A

Polymorphonuclear cells

  • Neutrophils
  • Eosinophils
  • Basophils
  • Monocytes and macrophages
  • NK cells
  • Dendritic cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are the soluble components of the innate immune system?

A
  • Complement
  • Acute phase proteins
  • Cytokines and chemokines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the key features of the innate immune response?

A

Identical responses in all individuals Cells express receptors that allow them to detect and home to sites of infection Cells express genetically encoded receptors (PRRs) Phagocytic capacity to engulf pathogens Cells secrete cytokines and chemokines that regulate the immune response

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

Describe the role of neutrophils.

A
  • Acute inflammation/Migrate rapidly to injury site
  • Express receptors for cytokines and chemokines to detect inflammation
  • Express Pattern Recognition Receptors to detect pathogens
  • Express Fc receptors for immunoglobulin to detect immune complexes
  • Perform phagocytosis and oxidative and non-oxidative killing
  • Release enzymes, histamine, lipid mediators of inflammation from granules
  • Secrete cytokines and chemokines to regulate inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Describe the role of macrophages.

A

Present within tissue - Express receptors for cytokines and chemokines, express Fc receptors for immune complexes, express PRR - Perform phagocytosis and oxidative and non-oxidative killing - Secrete chemokines and cytokines to regulate inflammation - CAN process and present antigen to T cells

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

Describe phagocyte recruitment.

A
  • Cellular damage and bacterial products trigger local production of inflammatory mediators (cytokines and chemokines)
    • Cytokines activate vascular endothelium increasing permeability
    • Chemokines attract phagocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

How does the innate immune system recognise microorganisms?

A

Pattern recognition receptors (PRRs) - Toll-like receptors and mannose receptors recognise generic motifs known as pathogen-associated molecular patterns (PAMPs) e.g. bacterial sugars, DNA and RNA Fc receptors on innate immune cells allows them to bind to Fc portion of immunoglobulins to allow phagocytes to recognise immune complexes

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

Describe endocytosis.

A
  1. Opsonins act as a bridge between the pathogen and the phagocyte receptors
  2. Antibodies bind to Fc receptors on phagocytes
  • Complement components can bind to complement receptors (e.g. CR1)
  • Acute phase proteins (e.g. CRP) will also promote phagocytosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is a phagolysosome?

A

Phagolysosome = a phagosome fused with a lysosome

  • forms a protected compartment in which killing of the organism occurs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Describe oxidative killing.

A
  1. NADPH oxidase converts oxygen into reactive oxygen species - superoxide and hydrogen peroxide
  2. Myeloperoxidase catalyses the production of hydrochlorous acid from hydrogen peroxide and chloride
  3. Hydrochlorous acid is a highly effective oxidant and anti-microbial
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Describe non-oxidative killing.

A

Killing by release of bactericidal enzymes e.g. lactoferrin and lysozyme into the phagolysosome - The range of enzymes results in a broad range of cover against bacteria and fungi

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Describe the role of natural killer cells.

A

Speacialised lymphocytes that are in the blood and may migrate to inflamed tissue

Express inhibitory receptors for self HLA class 1 which prevents inappropriate activation by normal self-cells

Express range of activatory receptors including natural cytotoxicity receptors that recognise heparan sulphate proteoglycans

Integrate signals from inhibitory and activatory receptors (usually inhibitory signals dominate)

Cytotoxic - kill ‘altered self’ cells (i.e. malignancy or virus-infected cells)

Secrete cytokines to regulate inflammation and promote dendritic cell function

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

Where are dendritic cells found?

A

Peripheral tissue

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

Describe the role of immature dendritic cells.

A

Detect inflammation - express receptors for cytokines and chemokines

Detect pathogens - express pathogen recognition receptors

Detect immune complexes - express Fc receptors for immunoglobulin

Capable of phagocytosis

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

What triggers dendritic cells to mature?

A

They do so following phagocytosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Describe the role of mature dendritic cells.

A

Upregulate expression of HLA molecules

Express co-stimulatory molecules

Migrate via lymphatics to lymph nodes

Present processed antigen to T cells in lymph nodes to prime the adaptive immune system

Express cytokines to regulate the immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What are the components of the adaptive immune system?

A

Humoral Immunity - B lymphocytes and antibody

Cellular Immunity - T lymphocytes (CD4+ and CD8+)

Soluble Components - cytokines and chemokines

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

What are the 4 key features of the adaptive immune system?

A
  1. Wide repertoire of antigen receptors - VDJ recombination with mechanisms to delete or tolerate these cells
  2. Exquisite specificity
  3. Clonal expansion - cells with appropriate specificity will proliferate during infection
  4. Immunological memory
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Define primary lymphoid organ and describe each one.

A

Organs involved in lymphocyte development

  • Bone marrow - both T and B lymphocytes are derived from haematopoietic stem cells in BM but only B cells mature here
  • Thymus - site of T cell maturation - most active in fetal and neonatal period, involutes after puberty
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Define the secondary lymphoid organs and state what they are.

A

Anatomical sites of interaction between naïve lymphocytes and microorganisms in which immune reaction occurs

  • Spleen
  • Lymph nodes
  • Mucosal associated lymphoid tissue (MALT)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Describe T lymphocyte maturation.

A
  1. Arise from haematopoietic stem cells
  2. Exported as immature cells to thymus where they undergo selection
  3. Mature T cells enter circulation and reside in secondary lymphoid organs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What do CD8 T cells recognise?

A

Peptides presented by HLA Class I

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

What do CD4 T cells recognise?

A

Peptides presented by HLA Class II

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

What selection process of T cells occurs?

A

Based on affinity to HLA molecules

  • Low affinity = useless so removed
  • High affinity = dangerous/autoimmune so removed
  • 10% develop further
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is the role of CD4 T cells?

A

Recognise peptides derived from EC proteins

Provide help for development of full B cell response

Provide help for the development of some CD8+ T cell responses

Peptides usually presented on HLA Class 2

Immunoregulatory functions via cell: cell interactions and cytokine expression

  • AKA T helper cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is the role of Th1 CD4 T cells?

A

Express CD4 and secrete IFN-gamma and IL2 to help CD8 and macrophages

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

What is the role of T follicular CD4 T cells?

A

Promote germinal centre reactions and differentiation of B cells into IgG and IgA secreting plasma cells

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

What is the role of regulatory CD4 T cells?

A

Express CD25 and transcription factor Foxp3

Secrete IL10 (immunosuppressive cytokine) to inhibit immune response

Express CTLA4 to directly inhibit T cell activation

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

What is the role of CD8 cytotoxic T cells?

A

Recognise peptides derived from intracellular proteins presented on HLA Class 1

Kills cells directly via perforin (pore-forming) and granzymes (expression of Fas ligand)

Secrete cytokines (e.g. IFN-gamma, TNF-alpha)

Important against viruses and tumours

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

Describe T cell memory.

A

Response to successive exposures of antigen

Pool of memory T cells ready to respond to antigen

More easily activated than naïve cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What is the tolerance mechaniusm of B cells?

A

If they recognise self then they are negatively selected/deleted

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What happens when B cells encounter antigens?

A

Early IgM Response - Cell differentiates into IgM secreting plasma cell

Germinal Centre Reaction - Somatic hypermutation and isotype switching from IgM to IgG/A/E

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

Describe the process of germinal centre reactions that develop an antibody response.

A

Dependent on T helper cells

  1. Dendritic cells prime CD4+ T helper cells
  2. CD4+ cells then help B cell differentiation (mediated by CD40L: CD40)
  3. B cells proliferate with the help of CD4 T cells
  4. Undergo somatic hypermutation and isotype switching (from IgM to IgG/A/E)
  5. Become plasma cells - produce antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Describe the structure of an immunoglobulin.

A

Soluble proteins made up of 2 heavy chains + 2 light chains

Heavy chain determines antibody class (e.g. M, G, A)

Effector function determined by constant region (Fc) - heavy chain

Antigen binding region (Fab) recognise antigens - heavy and light chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What are the functions of antibodies?

A

Identify pathogens and toxins (Fab-mediated)

Interact with other components of immune system to remove pathogens (Fc-mediated) - Complement, Phagocytes, NK cells

Particularly important in defence against BACTERIA

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

How do antibodies provide defence against infection?

A

Neutralisation - binds to receptors on target molecule so it cannot bind to anything else thereby blocking interaction with cells

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

Describe IgA antibodies.

A

Protection of mucosal surfaces via salivary, respiratory, gastrointestinal and lacrimal secretions = secretory function

  • Dimer form with 4 binding sites found in mucosal and epithelial surfaces e.g. gut, mouth
  • Monomer circulates in serum

IgA main antibody in breast milk, providing passive immunity in neonates

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Describe IgG antibodies.

A

Most abundant antibody type

Monomor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Describe IgM antibodies.

A

Pentamer

Primary response against pathogens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

Describe IgE antibodies.

A

Parasites and Type I hypersensitivity reactions

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

Describe B cell memory.

A

Decreased lag time between antigen exposure and antibody production (to 2-3 days)

Increased antibody titres produced

Response dominated by IgG antibodies of high affinity - compared to IgM in primary response

Response can be independent of help from CD4 cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What is complement?

A

Over 20 tightly regulated linked proteins that are produced by liver

Inactive in the circulation

When activated by enzymes they activate other proteins in the biological cascade to produce a rapid, highly amplified response

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

What are the 3 pathways of complement activation?

A

Classical - C1, C2 and C4

Mannose Binding Lectin (MBL) - C2 and C4

Alternative

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What is the final common pathway in complement activation?

A

All routes converge on C3

Final common pathway C5-C9 to membrane attack complex

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

Describe the classical pathway of complement activation.

A

Activated by immune complexes

  1. Formation of antibody-antigen immune complexes results in conformational change in antibody shape which exposes binding site for C1
  2. Binding of C1 to antibody results in cascade activation (dependent on the adaptive immune response so its not an early response)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Describe the Mannose Binding Lectin (MBL) pathway of complement activation.

A

MBL → C2, C4 → C3

  • Binding of MBL to microbial cell surface CHO → Direct stimulation of classical pathway (only C4, C2)
  • Not dependant on acquired immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

Describe the alternative pathway of complement activation.

A

Directly triggered by binding of C3 to bacterial cell wall components (lipopolysaccharide of gram-negative bacteria)

Not dependent on adaptive immune response

Involves factors: B, I, P

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

Describe the common pathway of complement activation.

A

Convergence occurs at C3

  1. Activation of C3 convertase = major amplification step
  2. Triggers formation of membrane attack complex MAC via C5-9
  3. MAC makes holes in membranes hence killing pathogen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What are the roles of complement fragments released during complement activation?

A

Increase vascular permeability and cell trafficking to inflammation site

Opsonisation of immune complexes to keep them soluble

Opsonisation of pathogens to promote phagocytosis

Phagocyte activation

Promotes mast cells/basophil degranulation

Makes holes in bacterial membranes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What are cytokines?

A

Small protein messengers with immunomodulatory function

Autocrine and paracrine dependent action

  • IL2, IL6, IL10, IL12, TNF-alpha, TGF-beta
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What are chemokines?

A

Type of cytokines

Direct recruitment of leukocytes in inflammatory response via CCL19 and CCL21 = ligands for CCR7 (receptors)

Important in directing dendritic cell trafficking to lymph nodes

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

What are the causes of secondary immune deficiences?

A
  • Malnutrition - most common globally
  • Infections - HIV, Measles, TB, SARS-CoV-2
  • Age extremities
  • Splenic surgery/trauma
  • Drugs - glucocorticoids (commonest), cytotoxic agents, chemotherapy, calcineurin inhibitors, antiepileptics, DMARDs, JAK inhibitors
  • Genetic or Metabolic disease
  • Haematological cancers
  • Biological agents/cellular therapy - anti-CD20 etc or rituximab (antibody therapies)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What is Good’s syndrome?

A
  • Thymoma and Antibody deficiency
    • Combined T and B cell absence/defect
    • CMV PJP and muco-cutaneous candida
    • Increased risks of autoimmune disease
      • Pure red cell aplasia, Myasthenia gravis, Lichen planus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What questions should you ask to evaluate secondary immune deficiency?

A
  • Clinical history of infection
  • Childhood illnesses/loss of schooling
  • Other illness/PMH
  • Family history of infection/autoimmune/cancers
  • Medication history
  • Vaccine history and any reactions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What investigation should be done for immune defects?

A
  • FISH for immunodeficiency - Picks up 85% of immune defects
    • Full blood count
    • Immunoglobulins
    • Serum complement (C3 and C4 – serum complex disease, SLE, C1 inhibitor deficiency (a primary immune deficiency))
    • HIV test
  • First Line Investigations
    • Renal and liver profile
    • Calcium and bone profile
    • Total protein and albumin
    • Urine protein/Cr ratio
    • Serum protein electrophoresis (monoclonal proteins found in MM, WMG, NHL and MGUS)
    • Serum free light chains (b-lymphoma)
  • Second Line Investigations
    • Measure conc of vaccine antibodies - tetanus (protein antigen) and pneumovax (carbohydrate antigen of 23 types)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What does an isolated reduction in IgG suggest?

A
  • Protein loosing enteropathy
  • Prednisolone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What does a reduction in IgG and IgM suggest?

A
  • B cell neoplasm
  • Rituximab
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What does a reduction in IgG and IgA suggest?

A
  • Primary antibody deficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is the management of secondary immune deficiencies?

A
  • Treat underlying cause
  • Advise measures to reduce exposure
  • Immunisation against respiratory viruses and bacteria and offer vaccines to household contacts
  • Education to treat bacterial infections promptly (use of rescue antibiotics)
  • Prophylactic antibiotics for confirmed recurrent bacterial infections
  • IgG replacement therapy – if other management have failed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

Describe the structure of HIV?

A
  • HIV has an icosahedral (20-faced) structure
  • Double-stranded RNA
  • Genome is diploid
  • Contains 9 genes that encode 15 structural, regulatory and auxiliary proteins
    • I.E. gp120, gp41 and RT
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

How does HIV replicate?

A

Inside a cell using Reverse Transcriptase to convert RNA to DNA which is integrated into the host genome

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What is the target of HIV?

A
  • CD4+ T-helper cells
  • CD4+ monocytes
  • CD4+ dendritic cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What are the possible routes of transmission of HIV?

A
  • Sexually
  • Infected blood – transfusion, needle sharing, blood products
  • Vertical (mother to child) – ante-/intra-partum, breastmilk
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

Describe the life cycle of HIV?

A
  1. Attachment and entry
  2. Reverse transcription and DNA synthesis
  3. Integration
  4. Viral transcription
  5. Viral protein synthesis
  6. Assembly of virus and release of virus
  7. Maturation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

Describe the clinical course of HIV.

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

What are the immunological features of HIV infection?

A
  • CD4 T cell depletion
  • Chronic immune activation
  • Impairment of CD4 and CD8 T cell function
  • Disruption of lymph node architecture
  • Impaired ability to generate protective T and B cell immune responses
  • Loss of antigen-specific humoral immune responses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What are the mechanisms of HIV long-term non-progressors?

A
  • Host genetic factors:
    • HLA profile - Heterozygosity for 32-bp deletion in chemokine-r CCR5
    • MBL alleles - TNF c2 microsatellite alleles
    • Gc vitamin D-binding factor alleles
  • Host immune response factors
    • Effective CTL, HTL and humoral responses
    • Secretion of:
      • CD8 antiviral factor
      • IL-16
      • Secretion of chemokines that block HIV entry co-receptors CCR5 and CXCR4
    • Maintenance of functional lymphoid tissue architecture
  • Virologic factors
    • Infection with attenuated strains of HIV
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

How is HIV diagnosed and monitored?

A
  • Detection via:
    • Anti-HIV antibodies (ELISA) – screening test
    • Anti-HIV antibodies (Western Blot) – confirmation test
    • Viral load (viral RNA detection using PCR) – very sensitive; steps:
  • ​CD4+ T Cell Counts – monitored using flow cytometry
    • The onset of AIDS correlates with a decrease in the number of CD4+ T cells
      • Antigens on T cells:
        • CD3, CD4, CD8, CD19, CD56
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What is the best predictor of HIV prognosis?

A

Initial baseline plasma viral load

  • Higher the load the sooner the progression to AIDS if not treated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

At what CD4 T cell counts should preventative measure be taking against:

  • PCP
  • Toxoplasma gondii
  • Mycobacterium avium complex (MAC)
A
  • PCP = 200 x 109/L
  • Toxoplasma gondii = 100 x 109/L
  • Mycobacterium avium complex (MAC disease) = 75 x 109/L
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

How can resistance against anti-retroviral therapy be tested for?

A
  • Phenotypic - viral replication is measured in cell cultures under selective pressure of increasing concentrations of antiretroviral drugs (compared to wildtype)
  • Genotypic - mutations detected by sequencing amplified HIV genome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What is contained within HAART?

A
  • Combination of ≥3 ART drugs = Triple Therapy:
    • 2 backbone drugs - x2 NRTIs
    • ≥1 binding agent - x1 NNRTI or INI
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

Name some examples of each type of ART backbone?

A
  • Nucleoside Reverse Transcriptase Inhibitors (NRTI) – e.g. Zidovudine/AZT, abacavir
  • Nucleotide RTI – e.g. Tenofovir
  • Non-NRTI (NNRTI) – e.g. Efavirenz
  • Protease inhibitor (PI) – e.g. Indinavir
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

Name some examples of each type of ART binding agents?

A
  • Integrase inhibitors (INI) – e.g. Raltegravir
  • Attachment inhibitors (AI) – e.g. Maraviroc
  • Fusion inhibitors (FI) – e.g. Enfuvirtide
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What interactions exist with HAART?

A
  • PIblock cytochrome P450
  • EfavirenzP450 inducer
  • INIinteracts with indigestion remedies (Gaviscon, aluminium salts, calcium salts) and is sequestered which can be very bad as some INI is absorbed but very little and so resistance is bred very quickly
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What are the limitations and complications of HAART?

A
  • Adherence – most common reason for failure
  • Does NOT eradicate latent HIV-1
  • Fails to restore HIV-specific T cell responses
  • Threat of drug resistance
  • Significant toxicities
  • High pill burden
  • Quality of life
  • Cost - >40% with no access
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What methods work to reduce the spread of HIV?

A
  • Male circumcision (APCs in foreskin at a high density)
  • Condoms
  • PrEP (Truvada)
  • TasP (Treatment as Prevention)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What cures exist for HIV?

A
  • Allogenic SCT from a CCRδ32 HLA matched donor
    • CCRδ32 are unable to contract HIV – found in 1% of north-west European patients
  • Shock and Kill
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

Describe B lymphocyte maturation.

A
  1. Initially exist in periphery as IgM B cells
  2. Undergo germinal centre reaction to differentiate into plasma cells expressing IgG, IgE and IgA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What are the most relevant proteins in terms of transplantation?

A
  • ABO blood group
  • HLA - MHC
  • Other minor histocompatibility genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What are the two major forms of rejection?

A
  • T cell-mediated rejection
  • Antibody-mediated rejection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

Where are HLA expressed?

A
  • HLA Class I (A, B and C) = ALL cells
    • Thought to be the most immunogenic
  • HLA Class II (DR, DQ, DP) = APCs
    • Upregulated on other cells under stress
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

What are the features of HLA?

A
  • They are highly polymorphic with hundreds of alleles for each locus
  • High degree of variability from the areas of protein lining the peptide-binding groove which allows us to present a wide variety of antigens in that peptide-binding groove to the cells of the immune system
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What HLA molecules are the most important to match in transplantation?

A

A, B and DR

  • DR > B > A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

Describe T-cell mediated rejection.

A
  • Phase 1 = Activation of T-cells
    • Presentation of foreign HLA antigens in MHC by APCs and co-stimulatory signals activate T-cells within lymph nodes
    • This leads to effector phase of rejection → inflammation caused leads to graft dysfunction
  • Phase 2 = Actions of activated T cells
    • Proliferation
    • Product cytokines (IL2 is important)
    • Provide help to CD8+ cells
    • Provide help for antibody production
    • Recruit phagocytic cells
  • Phase 3 = Effector Phase (Image)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What are the effects of cytotoxic T cells have on a transplanted organ?

A
  • Granzyme B (toxin)
  • Perforin (punch holes)
  • Fas-ligand (apoptosis)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What are the effects of macrophages have on a transplanted organ?

A
  • Phagocytosis
  • Proteolytic enzymes
  • Cytokine release
  • O2 and N2 radicals
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What are the histological features of T cell mediated rejection?

A
  • Lymphocytic interstitial infiltration
  • Ruptured tubular basement membrane
  • Tubulitis - inflammatory cells within the tubular epithelium
  • Macrophages
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

Describe antibody mediated rejection.

A
  • Phase 1: recognition of foreign antigens
  • Phase 2: activation of antigen-specific lymphocytes
  • Phase 3: effector phase - antibodies in graft
    • Antibodies bind to antigens (HLA) on the endothelium of the blood vessels in the transplanted organ
    • Antibodies activate complement
      • Form MAC → endothelial cell lysis
      • Recruit inflammatory cells to the microcirculation
      • Antibodies crosslink the MHC molecules
      • The antibodies can also directly recruit mononuclear cells, NK cells and neutrophils → capillaritis
  • Phase 4: graft fibrosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What are the cardinal features of antibody-mediated rejection?

A

Capillaritis = inflammatory cells in capillaries of the kidney → injury

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What is the histology of antibody-mediated rejection?

A
  • Inflammatory cell infiltrate
  • Capillaritis
  • Fixation of complement fragments on endothelial cell surfaces
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What kind of damage is caused by T-cell and antibody medtaited rejection?

A
  • T-cells = Interstitial damage
  • Antibodies = Endothelial damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

How is graft rejection prevented?

A
  • HLA typing
  • Screening for anti-HLA antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

Describe anti-HLA antibody screening.

A
  • Cytotoxicity assays
    • Inspects if recipient’s serum will kill the lymphocytes of the donor
    • Positive crossmatch suggests that there is cell lysis
  • Flow cytometry
    • Inspects if recipient’s serum binds to the donor’s lymphocytes using fluorescent anti-human immunoglobulin
  • Solid phase assays
    • Recipient’s serum is mixed with beads and fluorescently labelled immunoglobulin is used to determine which HLA epitopes the antibodies bind to
    • Patients that have antibodies to lots of different types of antibodies are regarded as highly sensitised
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

How can organ mismatch issues be overcome?

A
  • Improve transplantation across tissue barriers
  • More donors
  • Organ exchange programmes
  • Future: xenotransplantation (animals), stem cell research
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What are the 3 signals to activate T-cells?

A
  • APC MHC to T-cell TCR = Main signal
  • APC CD80/CD86 to T-cell CD28 - CD80/86 to CTLA4 suppresses immune reactions
  • Cytokine IL-2 to T-cell CD25 - after T-cell activation, autocrine IL-2 is released to further activate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What immunosuppressants are used in the management/prevention of T cell mediated rejection?

A
  • Steroids - prevent general T-cell mediated rejection
  • Inhibitors of cell signalling - Calcineurin inhibitors
  • Anti-proliferative agents
  • Inhibitors of cell surface receptors
    • Alemtuzumab = anti-CD52 monoclonal antibody that causes lysis of T cells
    • Basiliximab = anti-CD25 monoclonal antibody which targets the IL-2-R à less proliferation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

What are the main targets for immunosuppression for antibody-mediated rejection?

A
  • B-cell activation
  • Plasma cell secretion of antibodies
  • Antibody effects on endothelium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

What immunosuppressants are used in the management/prevention of antibody-mediated rejection?

A
  • Rituximab - B cell depletion
  • BAFF inhibitors
  • Proteasome inhibitors - block production of antibodies by plasma cells
  • Complement inhibitors - block complement binding to endothelial cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

What is the modern transplant immunosuppression regimen?

A
  • Induction agent = OXT3/ATG, anti-CD52, anti-CD25
    • Given at time of transplantation or just before to prepare the patient to receive the foreign organ
  • Baseline immunosuppression = calcineurin inhibitor + mycophenolate mofetil / azathioprine ± steroids
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What is the management of acute transplant rejection?

A
  • Cellular = Steroids, OKT3, ATG
  • Antibody-mediated = IVIG, Plasmapheresis, Anti-C5, Anti-CD20
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

Describe post-transplant malignancy.

A
  • Viral-associated malignancies are much more common
    • Kaposi sarcoma (HHV8)
    • Lymphoproliferative disease (EBV)
  • Skin cancer is 20x more common
  • Risk of other cancers is also increased
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

What are the features of immunodeficiency?

A
  • Infection
    • Two major or one major and recurrent minor infections in one year
    • Atypical organisms
    • Unusual sites
    • Poor response to treatment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

What are the features of primary immune deficiency?

A
  • Infection
  • Family history
  • Young age at presentation
  • Failure to thrive
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What are the common investigations for primary immune deficiency?

A
  • White cells
    • Full blood count
    • Lymphocyte subsets
    • White cell migration/function
  • Immunoglobulins
    • IgM, IgG, IgA
    • Specific Igs and response to vaccination
  • Complement
    • Complement function
    • Individual complement components
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What is the role of phagocytes?

A
  • Express cytokine/chemokine receptors to locate sites of infection
  • Express genetically encoded receptors for detection of pathogens at site of infection
    • Pattern recognition receptors which recognise generic motifs known as pathogen-associated molecular patterns (PAMPs) such as bacterial sugars, DNA, RNA
  • Express Fc receptors for detection of immune complexes
  • Phagocytic capacity to engulf the pathogens
  • Secrete cytokines and chemokines to regulate immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What are the types of primary phagocyte deficiency?

A
  • Failure to produce neutrophils
    • Failure of stem cells to differentiate along myeloid or lymphoid lineage
      • Reticular dysgenesis = Autosomal recessive severe SCID
    • Specific failure of neutrophil maturation
      • Kostmann syndrome
      • Cyclic neutropenia
  • Defect of phagocyte migration
    • Leukocyte adhesion deficiency
  • Failure of oxidative killing mechanisms
    • Chronic granulomatous disease
  • Cytokine deficiency
    • IL12, IL12R, IFNg or IFNg R deficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What is Reticular dysgenesis?

A
  • Autosomal recessive severe SCID
    • Mutation in mitochondrial energy metabolism enzyme adenylate kinase 2 (AK2)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

What is Kostmann syndrome?

A
  • Autosomal recessive severe congenital neutropenia
    • Classical form due to mutation in HCLS1-associated protein X-1 (HAX1)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

What is Cyclic neutropenia?

A
  • Autosomal dominant episodic neutropenia every 4-6 weeks
    • Mutation in neutrophil elastase (ELA-2)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

What is Leukocyte adhesion deficiency?

A
  • Deficiency of CD18 (b2 integrin subunit)
    • CD11a/CD18 (LFA-1) is expressed on neutrophils
      • Binds to ligand (ICAM-1) on endothelial cells and so regulates neutrophil adhesion/transmigration
    • In Leukocyte adhesion deficiency the neutrophils lack these adhesion molecules and fail to exit from the bloodstream
      • Very high neutrophil counts in blood
      • Absence of pus formation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What are the features of leukocyte adhesion deficiency?

A
  • Very high neutrophil counts in blood
  • Absence of pus formation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What are the features of chronic granulomatous disease?

A
  • Absent respiratory burst
  • Excessive inflammation
  • Granuloma formation
  • Lymphadenopathy
  • Hepatosplenomegaly
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What is chronic granulomatous disease?

A
  • Deficiency of one of components of NADPH oxidase
    • Inability to generate oxygen free radicals results in impaired killing
  • Persistent neutrophil/macrophage accumulation
    • Failure to degrade antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What are the appropriate investigations for suspected chronic granulomatous disease?

A
  • Nitroblue tetrazolium (NBT) test
    • NBT is a dye that changes colour from yellow to blue, following interaction with hydrogen peroxide
  • Dihydrorhodamine (DHR) flow cytometry test
    • DHR is oxidised to rhodamine which is strongly fluorescent, following interaction with hydrogen peroxide
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

What infections are common in patients with phagocyte deficiency?

A
  • Bacterial infections
    • Staphylococcus aureus
    • Enteric bacteria
  • Fungal infections
    • Candida albicans
    • Aspergillus fumigatus and flavus
  • Mycobacterial infection
    • Mycobacterium tuberculosis
    • Atypical Mycobacteria
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What is the management of phagocyte deficiency?

A
  • Aggressive management of infection
    • Infection prophylaxis
      • Antibiotics → e.g. Septrin
      • Anti-fungals → e.g. Itraconazole
    • Oral/intravenous antibiotics as needed
  • Definitive therapy
    • Haematopoietic stem cell transplantation
    • Specific treatment for CGD → Interferon gamma therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

What are the types of primary natural killer cells deficiency?

A
  • Classical NK deficiency
    • Absence of NK cells within peripheral blood
    • Abnormalities described in GATA2 or MCM4 genes in subtypes 1 and 2
  • Functional NK deficiency
    • NK cells present but function is abnormal
    • Abnormality described in FCGR3A gene in subtype 1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What infections are common in primary natural killer cells deficiency?

A
  • Virus infection
    • Herpes virus infection
      • Herpes Simplex Virus I and II
      • Varicella Zoster Virus
      • Epstein Barr Virus
      • Cytomegalovirus
      • Papillomavirus infection → associated cancers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What infections are common in patients with natural killer cells deficiency?

A
  • No good trial data
  • Prophylactic antiviral drugs such as acyclovir or gancyclovir
  • Cytokines such as IFN-alpha to stimulate NK cytotoxic function
  • Haematopoietic stem cell transplantation in severe phenotypes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What are the types of primary deficiency of complement?

A
  • Complement deficiency
  • Classical pathway deficiency (C2, C1q)
  • MBL deficiency MBL2 mutations are common but not usually associated with immunodeficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What is Complement deficiency?

A
  • Susceptibility to bacterial infections → especially encapsulated bacteria
    • Neisseria meningitides → properidin and C5-9 deficiency
    • Haemophilus influenzae
    • Streptococcus pneumoniae
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What is Classical pathway deficiency?

A
  • Deficiency in C2 and C1q
    • Susceptibility to SLE
    • Failure of phagocytosis of dead cells
      • Increased nuclear debris
    • Failure to clear immune complexes
      • Immune complex deposition in blood vessels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

How can SLE lead to to functional complement deficiency?

A
  1. Active lupus causes persistent production of immune complexes
  2. Consumption of complement leading to functional complement deficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

How can nephritic factors lead to functional complement deficiency?

A
  • Nephritic factors are auto-antibodies directed against components of the complement pathway
  • Nephritic factors stabilise C3 convertases resulting in C3 activation and consumption
  • Often associated with glomerulonephritis (classically membranoproliferative)
  • May be associated with partial lipodystrophy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What are the appropriate investigation for deficiency in complement?

A
  • Measure C3 and C4
  • Functional
    • CH50 - classical pathway
    • AP50 - alternative pathway
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What is the management of complement deficiences?

A
  • Vaccination
    • Boost protection mediated by other arms of the immune system
    • Meningovax, Pneumovax and HIB vaccines
  • Prophylactic antibiotics
  • Treat infection aggressively
  • Screening of family members
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What is the pathophysiology of SCID?

A
  • Defects of Haemopoetic stem cells
    • Reticular dysgenesis - most severe SCID
  • Defects of Lymphoid precursors
    • Less severe forms of SCID
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

Describe X-linked SCID.

A
  • 45% of all SCID
  • Mutation of common gamma chain on chromosome Xq13.1
    • Shared by cytokine receptors for IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21
    • Inability to respond to cytokines causes early arrest of T cell and NK cell development and production of immature B cells
  • Phenotype
    • Very low or absent T cell numbers
    • Very low or absent NK cell numbers
    • Normal or increased B cell numbers but low Igs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

Describe ADA deficiency.

A
  • 16.5% of all SCID
  • Adenosine Deaminase Deficiency
    • Enzyme required for cell metabolism in lymphocytes
  • Phenotype
    • Very low or absent T cell numbers
    • Very low or absent B cell numbers
    • Very low or absent NK cell numbers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What protects the SCID neonate in their first 3 months of life?

A

Source of circulating IgG in the neonate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What are the signs of SCID?

A
  • Unwell by 3 months of age
  • Infections of all types
  • Failure to thrive
  • Persistent diarrhoea
  • Unusual skin disease
    • Colonisation of infant’s empty bone marrow by maternal lymphocytes
    • Graft versus host disease
  • Family history of early infant death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

Describe T cell selection and central tolerance.

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

Describe CD8+ T celss.

A

Specialised cytotoxic cells

  • Recognise peptides derived from intracellular proteins in association with HLA class I
    • HLA-A, HLA-B, HLA-C
  • Kill cells directly
    • Perforin (pore forming) and granzymes
    • Expression of Fas ligand
  • Secrete cytokines
  • Particularly important in defence against viral infections and tumours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

Describe CD4+ cells.

A

Specialised helper cells

  • Recognise
    • Peptides derived from extracellular proteins
    • Presented on HLA Class II molecules (HLA-DR, HLA-DP HLA-DQ)
  • Immunoregulatory functions via cell:cell interactions and expression of cytokines
    • Provide help for development of full B cell response
    • Provide help for development of some CD8+ T cell responses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

What are the CD4+ T cell subsets?

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

What is DiGeorge syndrome?

A
  • Deletion at 22q11.2 → TBX1 may be responsible for some features
  • Usually sporadic rather than inherited
  • Normal numbers B cells
  • Reduced numbers T cells
  • Homeostatic proliferation with age
  • Immune function usually only mildly impaired and improves with age
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

What are the signs of DiGeorge syndrome?

A
  • High forehead
  • Developmental defects in the pharyngeal pouch
    • Low set
    • Abnormally folded ear
    • Cleft palate
    • Small mouth and jaw
  • Hypocalcaemia
  • Oesophaegeal atresia
  • Underdeveloped thymus
  • Complex congenital heart disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

What is the Bare lymphocyte syndrome - type 2?

A
  • Defect in one of the regulatory proteins involved in Class II gene expression
    • Regulatory factor X
    • Class II transactivator
  • Absent expression of MHC Class II molecules
  • Profound deficiency of CD4+ cells
    • Usually have normal number of CD8+ cells
    • Normal number of B cells
    • Low IgG or IgA antibody due to lack of CD4+ T cell help

BLS type 1 also exists due to failure of expression of HLA class I

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

What are the signs of bare lymphocyte syndrome?

A
  • Unwell by 3 months of age
  • Infections of all types
  • Failure to thrive
  • Family history of early infant death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

What are the clinical features of lymphocyte deficincies?

A
  • T cell deficiency
    • Viral infections
      • Cytomegalovirus
    • Fungal infection
      • Pneumocystis
      • Cryptosporidium
    • Some bacterial infections
      • Especially intracellular organisms
      • Mycobacteria tuberculosis
      • Salmonella
    • Early malignancy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

What are the appropriate investigations for suspected T cell deficiences?

A
  • Total white cell count and differential
  • Lymphocyte subsets
    • Quantify CD8 T cells, CD4 T cells as well as B cells and NK cells
  • Immunoglobulins
    • If CD4 T cell deficient
  • Functional tests of T cell activation and proliferation
  • HIV test
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What are the results of investigations for T cell deficiencies?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What is the management of T cell immunodeficiency?

A
  • Aggressive prophylaxis/treatment of infection
  • Haematopoieitic stem cell transplantation
    • Replace abnormal populations in SCID
    • Replace abnormal cells - class II deficient APCs in BLS
  • Enzyme replacement therapy
    • PEG-ADA for ADA SCID
  • Gene therapy
  • Thymic transplantation
    • Promote T cell differentiation in Di George syndrome
    • Cultured donor thymic tissue transplanted to quadriceps muscle
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

Describe B cell central tolerance.

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

What are the types of primary T cell immunodeficiency?

A
  • SCID
  • DiGeorge
  • BLS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

Describe immunoglobulins?

A
  • Soluble proteins made up of two heavy and two light chains
    • Heavy chain determines the antibody class
      • IgM, IgG, IgA, IgE, IgD
    • Antigen is recognised by the antigen binding regions (Fab) of both heavy and light chains
    • Effector function is determined by the constant region of the heavy chain (Fc)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

What is the function of antibodies?

A
  • Identification of pathogens and toxins (Fab mediated)
  • Interact with other components of immune response to remove pathogens (Fc mediated)
    • Complement
    • Phagocytes
    • Natural killer cells
  • Particularly important in defence against bacteria of all kinds
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What is Bruton’s X-linked a-gammaglobulinaemia?

A
  • Abnormal B cell tyrosine kinase (BTK) gene
    • Pre B cells cannot develop to mature B cells
  • Absence of mature B cells
  • No circulating Ig after 3 months
160
Q

What are the signs of Bruton’s X-linked a-gammaglobulinaemia?

A
  • Boys present in first few years of life
  • Recurrent bacterial infections
    • Otitis media
    • Sinusitis
    • Pneumonia
    • Osteomyelitis
    • Septic arthritis
    • Gastroenteritis
  • Viral, Fungal and Parasitic infections
    • Enterovirus, Pneumocystis
  • Failure to thrive
161
Q

What is Hyper IgM syndrome?

A
  • Mutation in CD40 ligand gene (CD40L, CD154)
    • Member of TNF Receptor family
    • Encoded on Xq26
    • Involved in T-B cell communication
    • Expressed by activated T cells but not B cells
  • Normal number circulating B cells
  • Normal number of T cells but activated cells do not express CD40 ligand
  • No germinal centre development within lymph nodes and spleen
  • Failure of isotype switching
  • Elevated serum IgM
  • Undetectable IgA, IgE, IgG
162
Q

What are the signs of Hyper IgM syndrome?

A
  • Boys present in first few years of life
  • Recurrent infections → particularly bacterial
  • Subtle abnormality in T cell function predisposes to:
    • Pneumocystis jiroveci infection
    • Autoimmune diseas
    • Malignancy
  • Failure to thrive
163
Q

What is Common variable immune deficiency?

A
  • Heterogenous group of disorders
    • Many different genetic defects → many unidentified
    • Failure of full differentiation/function of B lymphocytes
  • Defined by
    • Marked reduction in IgG, with low IgA or IgM
    • Poor/absent response to immunisation
    • Absence of other defined immunodeficiency
164
Q

What are the signs of common variable immune deficiency?

A
  • Recurrent bacterial infections
    • Pneumonia
    • Persistent sinusitis
    • Gastroenteritis
    • Often with severe end-organ damage
  • Pulmonary disease
    • Interstitial lung disease
    • Granulomatous interstitial lung disease (also LN, spleen)
    • Obstructive airways disease
  • Gastrointestinal disease
    • Inflammatory bowel like disease
    • Sprue like illness
    • Bacterial overgrowth
  • Autoimmune disease
    • Autoimmune haemolytic anaemia or thrombocytopenia
    • Rheumatoid arthritis
    • Pernicious anaemia
    • Thyroiditis
    • Vitiligo
  • Malignancy
    • Non-Hodgkin lymphoma
165
Q

What is Selective IgA deficiency?

A
  • Genetic condition with unknown cause
  • Prevalence = 1:600
    • 2/3rd = Asymptomatic
    • 1/3rd = Recurrent respiratory tract infections
166
Q

What are the types of primary B cell immunodeficiency?

A
  • SCID
  • Bruton’s X-linked a-gammaglobulinaemia
  • Hyper IgM syndrome
  • Common variable immune deficiency
  • Selective IgA deficienc
167
Q

What are the appropriate investigations for suspected B cell deficiences?

A
  • Total white cell count and differential
  • Lymphocyte subsets
    • Quantify B cells as well as CD4 T cells, CD8 T cells and NK cells
  • Serum immunoglobulins and protein electrophoresis
  • Functional tests of B cell function
    • Specific antibody responses to known pathogens/immunisations → measure IgG antibodies against tetanus, Haemophilus influenzae B and S. pneumoniae
    • If specific antibody levels are low, immunise with the appropriate killed vaccine and repeat antibody measurement 6–8 weeks later
168
Q

What are the results of investigations for B cell deficiencies?

A
169
Q

What is the management of B cell immunodeficiency?

A
  • Aggressive prophylaxis and Treatment of infection
  • Immunoglobulin replacement if required
    • Contains IgG antibodies to a wide variety of common organisms
    • Treatment is life-long
  • Immunisation
    • For selective IgA deficiency – Not otherwise effective because of defect in IgG antibody production
170
Q

What are the types of primary immunodeficiency?

A
171
Q

What are the appropriate investigations for suspected primary immunodeficiency?

A
  • White cells
    • Full blood count
    • Lymphocyte subsets
    • Special tests for white cell migration/function
    • Adhesion molecules – eg CD18
    • Test for oxidative killing – DHR test
  • Immunoglobulins
    • IgM, IgG, IgA
    • Specific Igs and response to vaccination
  • Complement
    • Complement function - CH50 and AP50
    • Individual complement components
172
Q

Define Allergic Disorder.

A

Immunological process that results in immediate and reproducible symptoms after exposure to an allergen.

  • Immunological process usually involves an IgE mediated type 1 hypersensitivity reaction
173
Q

Define Allergen.

A

Harmless substance that can trigger an IgE mediated immune response → clinical symptoms

174
Q

Define Sensitisation.

A

Detection of specific IgE either by skin prick testing or in vitro blood tests.

175
Q

How does the Th2 immune response occur?

A
  • Stressed or damaged epithelium will release signalling cytokines
  • Cytokines act on Th2, Th9 and ILC2 → promote the section of IL4, IL5 and IL13
    • IL-4 acts on both pathways
    • IL-4 stimulates B-cells to produce IgE and IgG4
    • These then act on eosinophils and basophils which plays a role in the expulsion of parasites and allergens but can also contribute to tissue injury
    • The TSLP and other cytokines released by the damaged epithelium can also activate follicular Th2 cells which then releases IL4
176
Q

Describe induction of Th2 immune response.

A
  • The primary defect is thought to be in the epithelial barrier (i.e. eczema and skin defect)
  • Follicular dendritic cells (Langerhans cells and dermal dendritic cells) promote secretion of Th2 cytokines more efficiently than other dendritic cell subtypes
  • IL4 secretion is only induced by peptide-MHC presentation to TCR or naïve/memory Th2 cells
177
Q

How does oral and skin/respiratory exposure alter the Th2 immune response?

A
  • Oral exposure promotes immune tolerance whereas skin and respiratory exposure induces IgE sensitisation
    • When an allergen is ingested through the oral route → T-regulatory cells inhibit IgE synthesis
      • Oral tolerance requires induction of CD4+ T-reg cells
    • T-regs inhibit multiple pro-allergic functions such as inhibiting DC APC function, secretion of IL-10, etc
178
Q

Describe the Th2 immune memory response.

A
  • The sensor is the mast cell → allergen causes cross-linking of IgE → histamine, prostaglandins and leukotrienes
  • These act on the endothelium → increased permeability, smooth muscle contraction and neuronal itch
  • Response acts to expel parasite/allergen OR will be responsible for symptoms of asthma, eczema and hay fever
179
Q

What are the theories to why allergic disease is increasing?

A
  • Hygiene Hypothesis - lack of childhood exposure to infectious agents increases susceptibility to allergic diseases by suppressing natural development of the immune system
  • Lack of vitamin D in infancy - leading to food allergy
  • Alternation of diversity in intestinal microbiome
  • High concentration of dietary advanced glycation end-products and pro-glycating sugars - immune system mistakenly recognises as causing tissue damage (e.g. fast food and soda)
180
Q

Describe the history of allergic disease.

A
  • Age of onset of the allergic disease:
    • Infants
      • Atopic dermatitis
      • Food allergy (milk, egg, nuts)
    • Children
      • Asthma (house dust mite, pets)
      • Allergic rhinitis
    • Adults
      • Drug allergy
      • Bee allergy
      • Oral allergy syndrome
      • Occupational allergy
181
Q

What are the signs and symptoms of allergic disease?

A
  • Occurs minutes to 3 hours after exposure
  • Symptoms – at least 2 organ systems usually involved
    • Skin symptoms = urticaria, angioedema
    • GI symptoms = D&V
    • Respiratory tract symptoms = SoB, cough, wheeze
    • Vasculature symptoms = hypotension, impending doom
  • Symptoms reproducible
  • Symptoms may be triggered by co-factors → i.e. NSAIDs in asthma, virus in children to VIF
182
Q

What are the appropriate investigations for suspected allergic disease?

A
  • Allergen-specific IgE tests
    • Skin prick and Intradermal testing
    • IgE RAST blood tests
  • Functional allergen tests
    • In vitro
      • Serial mast cell tryptase
      • Basophil activation test
    • In vivo
      • Open or blinded allergen challenge
183
Q

Describe the skin prick and intradermal testing.

A
  • Use skin test solutions
    • Positive control = histamine
    • Negative control = diluent
  • Positive outcome = wheal ≥3mm than negative control
  • Antihistamines discontinued for 48 hours beforehand (checked with positive control)
  • More sensitive and specific than blood tests to diagnose allergy in clinical practice
184
Q

What are the advantages and disadvantages of skin prick and intradermal testing?

A
  • Advantages
    • Rapid - 15-20 mins
    • Cheap and easy
    • Excellent negative predictive value - >95%
    • Increasing size = higher probability of allergy
    • Patient can see response
  • Disadvantages
    • Requires experience to interpret
    • Risk of anaphylaxis - 1 in 3,000
    • Poor positive predictive value/High false positive rate
    • Limited value in patients with dermatological conditions
    • False negative results with labile commercial food extracts
185
Q

Describe the method of IgE RAST blood tests.

A
  1. Allergen bound to sponge in a plastic cap and patient’s serum is added
  2. Specific IgE (if present) binds to allergen
  3. Anti-IgE antibody tagged with a fluorescent label is added
  4. Amount of IgE/Anti-IgE is measured by fluorescent light signal
186
Q

What are the indications for a IgE RAST blood test?

A
  • Patients who can’t stop antihistamines (otherwise do skin test)
  • Patients with dermatographism or extensive eczema
  • History of anaphylaxis
  • Borderline/equivocal skin prick test results
187
Q

Describe serial mast cell tryptase.

A
  • Tryptase is a pre-formed protein found in mast cell granules
  • Systemic degranulation of mast cells in anaphylaxis = increased serum tryptase
  • Peak concentration = 1-2 hours; baseline = 6-12 hours
    • Failure of baseline return after anaphylaxis = query systemic mastocytosis
  • Useful if diagnosis of anaphylaxis uncertain - e.g. hypotension/rash in anaesthesia
  • Reduced sensitivity for food-induced anaphylaxis
188
Q

Describe basophil activation testing.

A
  • Measurement of basophil response to allergen IgE cross-linking
  • Activated basophils increase expression of CD63, CD203 and CD300 protein
  • Increasingly used in the diagnosis of food and drug allergy
189
Q

What is the gold standard test for food and drug allergy diagnosis?

A

Allergen challenge - open or blinded

190
Q

Describe allergen testing.

A
  • Increasing volumes of the offending food/drug are ingested
  • Take place under close medical supervision
  • Difficult to interpret mild symptoms
  • Risk of SEVERE reaction
191
Q

What does a positive IgE test confirm?

A
  • Demonstrates Sensitisation, not clinical allergy
    • Detection of IgE is necessary but not sufficient to make a diagnosis of allergic disease
192
Q

How can serum IgE be used to predict allergic symptoms?

A
  • Concentration – higher levels = more symptoms
  • Affinity to target – higher affinity = increased risk
  • Molecular target within whole extract or even individual epitope can be linked to symptoms
  • Capacity of IgE antibody to induce mast cell degranulation
193
Q

What is Component Resolved Diagnostics?

A
  • Blood test to detect IgE to single protein components – abundance and stability of protein contributes to risk of allergic disease:
    • Useful for peanut and hazelnut allergy - may reduce need for food challenges
194
Q

What are the indications of allergen component testing?

A
  • Detect primary sensitisation
  • Confirm cross-reactivity
  • Define risk of serious reaction for stable allergens
  • Improve diagnostic sensitivity for components poorly represented in whole food extracts
  • Improve diagnostic sensitivity for unstable molecules in whole food extracts
195
Q

Define Anaphylaxis.

A

Severe potentially systemic hypersensitivity reaction.

Rapid onset, life-threatening airway, breathing and circulatory problems which is usually but not always associated with skin and mucosal changes.

196
Q

What are the signs and symptoms of anaphylaxis?

A
  • Skin is the most frequent organ involved (84%)
  • Cardiovascular
    • Collapse
    • Syncope
    • Drop in BP
  • Respiratory compromise
    • SoB
    • Wheeze
    • Stridor - more common in children
197
Q

What is the mechanism of IgE anaphylaxis?

A
  • Cells
    • Mast cells
    • Basophils
  • Mediators
    • Histamine
    • Platelet activating factor
198
Q

What is the mechanism of IgG anaphylaxis?

A
  • Cells
    • Macrophages
    • Neutrophils
  • Mediators
    • Histamine
    • Platelet activating factor
199
Q

What is the mechanism of complement anaphylaxis?

A
  • Cells
    • Mast cells
    • Macrophages
  • Mediators
    • Histamine
    • Platelet activating factor
200
Q

What is the mechanism of IgE anaphylaxis?

A
  • Cells
    • Mast cells
  • Mediators
    • Leukotrienes
    • Histamine
201
Q

What allergens causes IgE anaphylaxis?

A
  • Food
  • Insect venom
  • Ticks
  • Penicillin
202
Q

What allergens causes IgG anaphylaxis?

A
  • Biologicals
  • Blood and IgG Transfusions
203
Q

What allergens causes complement anaphylaxis?

A
  • Lipid excipients
  • Liposomes
  • Dialysis membranes and PEG
204
Q

What allergens causes pharmacological anaphylaxis?

A
  • NSAID including aspirin
  • Opiates
  • Neuromuscular drugs
  • Quinolones drug
205
Q

What reactions can mimic anaphylaxis?

A
  • SKIN: Chronic urticaria and angioedema (ACE inhibitors)
  • THROAT SWELLING: C1 inhibitor deficiency
  • CARDIOVASCULAR: Myocardial infarction and PE
  • RESPIRATORY: Very severe asthma, vocal cord dysfunction, inhaled FB
  • NEUROPSYCHIATRIC: Anxiety or panic disorder
  • ENDOCRINE: carcinoid and phaeochromocytoma
  • TOXIC: Scromboid toxicity (Histamine poisoning)
  • IMMUNE: Systemic mastocytosis
206
Q

How is a diagnosis of anaphylaxis confirmed?

A
  • Serial measurement of serum tryptase
    • Samples taken 1 hour, 3 hours and 24 hours post episode of anaphylaxis
    • The rise in tryptase concentration is directly proportional to fall in BP
  • DIAGNOSIS = persistent rise in tryptase 24 hours after allergic reaction suggestive of systemic mast cell disease
207
Q

What is management of anaphylaxis?

A
  • IM ADRENALINE
  • Supportive Treatments
    • Adjust body position
    • 100% Oxygen
    • Fluid replacement
    • Inhaled bronchodilators
    • Hydrocortisone 100 mg IV (prevent late phase response)
    • Chlorpheniramine 10 mg IV (skin rash)
208
Q

What is the mechanism of adrenaline in anaphylaxis?

A
  • Alpha 1 → causes peripheral vasoconstriction, reverses low BP and mucosal oedema
  • Beta 1 → increases heart rate, contractility and BP
  • Beta 2 → relaxes bronchial smooth muscle and reduces the release of inflammatory mediators
209
Q

Define Food Allergy and Food Intolerance.

A
  • Food ALLERGY = adverse health effect from specific immune response that occurs reproducibly on exposure to food
  • Food INTOLERANCE = non-immune reactions which include metabolic, pharmacological and unknown mechanisms
210
Q

What are the important questions in a clinical history for suspected allergy?

A
  • What does the patient mean by allergy?
  • Distinguish between IgE and non IgE mediated symptoms
  • Dose, how food is prepared, and co-factors can influence clinical symptoms
  • Does the patient have any history of atopic disease?
  • Enquire about previous investigations for food allergy i.e. SPT, IgE blood tests and complementary medical tests
  • Has elimination of food made any difference to symptoms?
  • Consider other differential diagnoses (food intolerance, eating disorders, coeliac disease)
211
Q

What are the appropriate investigations for suspected food allergy?

A
  • Clinical history
  • Skin-prick test or specific IgE blood test
    • +ve = sensitisation but not necessarily allergy
    • Higher IgE levels or larger wheals indicate a higher chance of allergy
    • Individual allergen protein component can distinguish between IgE-sensitisation and IgE-mediated allergy
  • Oral food challenge = gold standard
212
Q

What is the management of food allergy?

A
  • Avoidance – education, dietician, acknowledge anxiety, etc.
  • Anaphylaxis guidance for emergencies
  • Prevention – earlier introduction in high-risk families reduces chances of later development
213
Q

What are the IgE-mediated food allergy syndromes?

A
  • Anaphylaxis
  • Food associated exercise induced anaphylaxis
  • Delayed food-induced anaphylaxis
  • Oral allergy syndrome
214
Q

What is food associated exercise induced anaphylaxis?

A
  • Food induces anaphylaxis if individual exercises within 4-6 hours of ingestion
  • Common food triggers are wheat, shellfish, celery
215
Q

What is delayed food-induced anaphylaxis ?

A
  • Symptoms occur 3-6 hours after eating red meat and gelatin
  • IgE antibody to oligosaccharide alpha-gal (α1, 3-galactose) found in gut bacteria
  • Induced by tick bites which should be avoided
216
Q

What is oral allergy syndrome?

A
  • Limited to oral cavity, swelling and itch
  • Sensitisation to inhalant pollen protein lead to cross reactive IgE to food
  • Onset after pollen allergy established: affect adults > young children
  • Respiratory exposure to pollen (birch) results in IgE directed to homologous proteins in stone fruits (apple, pear) vegetables (carrot) and nuts (peanut, hazelnut)
  • Cooked fruits, vegetables and nut cause no symptoms → heat labile allergens detected by component allergen tests
  • 1-2% cases progresses to anaphylaxis
217
Q

Define Monogenic Auto-inflammatory Disease.

A

Mutations in a single gene encoding a protein involved in a pathway associated with innate immune cell function.

  • Abnormal signalling via cytokine pathways involving TNFα or IL1 is common
218
Q

Name some monogenic auto-inflammatory disease along with the gene mutation and inheritance pattern.

A
  • Muckle Wells Syndrome
    • NLRP3 - gain of function
    • Autosomal dominant
  • Familial Cold Auto-inflammatory Syndrome
    • NLRP3 - gain of function
    • Autosomal dominant
  • Chronic Infantile Neurological Cutaneous Articular Syndrome
    • NLRP3 - gain of function
    • Autosomal dominant
  • TNF Receptor Associated Periodic Syndrome
    • TNFRSF1
    • Autosomal dominant
  • Hyper IgD with Periodic Fever Syndrome
    • MK
    • Autosomal recessive
  • Familial Mediterranean Fever
    • MEFV
    • Autosomal recessive
219
Q

Describe the pathogenesis of familial Mediterranean fever.

A
  • Mutations in MEFV gene → Inactivated pyrin-marenostrin
    • Expressed mainly in neutrophils
  • Increased pro-caspase 1 and increased inflammation driven by lots of neutrophils
    • IL-1
    • NFKappaB → TNFa
    • Apoptosis
220
Q

What are the signs and symptoms of familial Mediterranean fever?

A
  • Periodic fevers lasting 48-96 hours associated with
    • Abdominal pain (peritonitis)
    • Chest pain (pleurisy and pericarditis)
    • Arthritis
    • Rash
221
Q

What condition do familial Mediterranean fever patients often go on to suffer with?

A

AA Amyloidosis

222
Q

What conditions are associated with AA amyloidosis?

A
  • Autoimmune diseases – rheumatoid arthritis, ankylosing spondylitis, IBD (CD & UC)
  • Autoinflammatory diseases – familial Mediterranean fever, Muckle–Wells syndrome
  • Chronic infections – TB, bronchiectasis, chronic osteomyelitis
  • Cancer – Hodgkin’s lymphoma, renal cell carcinoma
  • Chronic foreign body reaction
  • HIV/AIDS
223
Q

What are the appropriate investigations for suspected familial Mediterranean fever?

A
  • CRP - high
  • SAA - high
  • Blood to specialist genetics lab to detect mutation
224
Q

What is the management of familial Mediterranean fever?

A
  • Colchicine - 500μg BD
  • IL-1 and TNFa inhibitors
    • Anakinra - IL1 receptor antagonist
    • Etanercept - TNFα inhibitor
225
Q

Define Monogenic Auto-immune Disease.

A

Mutation in a gene encoding a protein involved in the adaptive immune response.

226
Q

Name some monogenic auto-inflammatory disease along with the gene mutation.

A
  • APS-1/APECED
    • AIRE - abnormality in tolerance
  • IPEX
    • FOXP3 - abnormality in regulatory T cells
  • ALPS
    • Abnormality of lymphocyte apoptosis
227
Q

Describe the pathogenesis of APS1/APECED.

A
  • Defect in autoimmune regulator = AIRE mutation
    • Transcription factor involved in T cell tolerance in the thymus
      • Upregulates expression of self-antigens by thymic cells
      • Promotes T cell apoptosis
  • Causes failure of central tolerance
    • Autoreactive T cells
    • Autoreactive B-cells - co-dependent on T cells
228
Q

What are the autoimmune diseases associated with APS1/APECED?

A
  • Hypoparathyroidism - 85%
  • Addison’s - 78%
  • Candidiasis - 98%
  • Hypothyroidism - 14%
  • Diabetes - 13%
  • Vitiligo - 27%
  • Enteropathy - 22%
229
Q

Describe the pathogenesis of IPEX.

A
  • Mutations in FOXP3 which is required for development of T-reg cells
  • Failure to negatively regulate T-cell responses → auto-reactive B-cells → autoantibody formation
230
Q

What are the autoimmune diseases associated with IPEX?

A
  • ‘Diarrhoea, Dermatitis and Diabetes’
    • Enteropathy - 98%
    • Diabetes mellitus - 35%
    • Dermatitis - 69%
    • Hypothyroidism - 35%
231
Q

Describe the pathogenesis of ALPS.

A
  • Mutations in FAS pathway → e.g. mutations in TNFRSF6 to encode FAS
  • Defect in apoptosis of lymphocytes
    • Failure of tolerance
    • Failure of lymphocyte homeostasis
232
Q

What are the signs and symptoms of ALPS?

A
  • Lymphocytosis → large lymph nodes
  • Splenomegaly
  • Autoimmune diseases (i.e. autoimmune cytopenias)
  • Lymphoma
233
Q

Define Polygenic Auto-inflammatory Diseases.

A

Mutations in genes encoding proteins involved in pathways associated with innate immune cell function.

234
Q

What are the features of polygenic auto-inflammatory disease?

A
  • Local factors at sites predisposed to disease lead to activation of innate immune cells such as macrophages, neutrophils with resulting tissue damage
  • HLA associations are usually less strong
  • These diseases are not characterised by presence of autoantibodies
235
Q

What gene has been identified to be linked to Crohn’s?

A
  • IBD1 gene on Chr16 identified as NOD2 → 3 different mutations
    • NOD2 gene mutations are present in 30% of patients
    • Abnormal allele of NOD2 increases risk of Crohn’s by:
      • 1 copy present = 1.5-3x
      • 2 copies present = 14-44x
  • NOD2 = a cytoplasmic microbial sensor in myeloid cells – recognises muramyl dipeptide → stimulates NFK-beta
  • Activation induces autophagy in dendritic cells
236
Q

What are the clinical features of Crohn’s?

A
  • Abdominal pain/tenderness
  • Diarrhoea – blood, pus, mucus
  • Fevers and malaise
237
Q

What is the management of Crohn’s disease?

A
  • Flare-up
    • Corticosteroid
    • Anti TNFα antibody
  • Maintenance/Remission
    • Diet-induced remission - whole protein modular diet
    • Aminosalicylates 5-ASA
238
Q

Define Mixed Pattern Auto-inflammatory/immune Disease.

A

Mutations in genes encoding proteins in pathways of innate AND adaptive immune cell function.

239
Q

What are the features of mixed pattern auto-inflammatory/immune disease?

A
  • HLA associations may be present
  • Autoantibodies not usually a feature
240
Q

What genetic polymorphisms are associated with ankylosing spondylitis?

A
241
Q

What are the signs and symptoms of ankylosing spondylitis?

A
  • Inflammation at sites of high tensile force
    • Low back pain and stiffness
    • Enthesitis
    • Large joint arthritis
242
Q

What is the management of ankylosing spondylitis?

A
  • NSAIDs
  • Immunosuppression – anti TNFα or anti IL17
243
Q

Define Polygenic Autoimmune Disease.

A

Mutations in genes encoding proteins in pathways of adaptive immune cell function.

244
Q

What are the features of polygenic autoimmune disease?

A
  • HLA associations common
  • Autoantibodies are found
245
Q

What diseases is the HLA-DR15 allele associated with?

A

Goodpasture

246
Q

What diseases is the HLA-DR3 allele associated with?

A
  • Graves
  • SLE
  • T1DM
247
Q

What diseases is the HLA-DR4 allele associated with?

A
  • T1DM
  • Rheumatoid arthritis
248
Q

What diseases is the HLA-DR1 allele associated with?

A

Rheumatoid arthritis

249
Q

What are the Gel and Coombs types?

A
  • Type 1 = Immediate hypersensitivity IgE mediated
    • Anaphylactic hypersensitivity
  • Type 2 = Antibody reacting with cellular antigen
    • Cytotoxic hypersensitivity
  • Type 3 = Antibody soluble antigen form immune complexes
    • Immune complex hypersensitivity
  • Type 4 = Delayed type hypersensitivity
    • Delayed hypersensitivity
250
Q

What is the pathogenesis of Type 1 (immediate hypersensitivity IgE mediated) autoimmune disease?

A

Rapid allergic reaction

  • Pre-existing IgE antibodies to allergen → IgE bound to Fc
    • Common allergens = pollens, drugs, food, insect, animal hair
  • Receptors on mast cells and basophils → cell degranulation
  • Release of inflammatory mediators
  • Increased vascular permeability, leukocyte chemotaxis and SM contraction
251
Q

What are the mechanisms of Type 2 (antibody reacting with cellular antigen) autoimmune disease?

A
252
Q

Name some Type 2 (antibody reacting with cellular antigen) autoimmune disease, their auto-antigen and symptoms.

A
  • Goodpasture
    • Non-collagenous domain of collagen type IV BM
    • Glomerulonephritis, pulmonary haemorrhage
  • Pemphigus vulgaris
    • Epidermal cadherin
    • Blistering of skin
  • Graves
    • TSH receptor
    • Hyperthyroidism
  • Myasthenia gravis
    • Acetylcholine receptor
    • Muscle weakness
253
Q

What is the mechanism of Type 3 (antibody reacting with soluble antigen to form immune complexes) autoimmune disease?

A
  • Antibodies bind to soluble antigen to form circulating immune complex
  • Immune complexes deposit in blood vessels
    • Complement activation, infiltration of macrophages and neutrophil
    • Cytokine and chemokine expression
    • Granule release from neutrophils
    • Increased vascular permeability
  • Inflammation and damage to vessels
    • Cutaneous vasculitis
    • Glomerulonephritis
    • Arthritis
254
Q

Name some Type 3 (antibody reacting with soluble antigen to form immune complexes) autoimmune disease, their auto-antigen and symptoms.

A
  • SLE
    • DNA, histones, RNP
    • Rash, glomerulonephritis, arthritis
  • Rheumatoid arthritis
    • Fc region of IgG
    • Arthritis
255
Q

What is the mechanism of Type 4 (delayed type hypersensitivity) autoimmune disease?

A
  • T cell mediated response → tissue destruction
    • HLA class 1 present SELF-antigens to CD8 T cells → cell lysis
    • HLA class 2 present SELF-antigen to CD4 T cells → cytokine production → inflammation and tissue damage
256
Q

Name some Type 4 (delayed type hypersensitivity) autoimmune disease, their auto-antigen and symptoms.

A
  • T1DM
    • Pancreatic beta-cell
    • Diabetes
  • Rheumatoid arthritis
    • Unknown synovial joint
    • Arthritis
  • MS
    • Myelin basic protein → brain infiltration by CD4+ T-cells
    • Neurological features, fatigue, vision blurring/loss
257
Q

A patient present with nervousness, palpitations, heat intolerant, diarrhoea, exophthalmos. What is the diagnosis?

A

Graves’

258
Q

What is the pathophysiology of Graves’?

A

T2 hypersensitivity → IgG antibodies which stimulate TSH receptor

  • Stimulating autoantibodies against TSH-receptor bind to receptor, acting as TSH agonists
  • Induce uncontrolled overproduction of thyroid hormones
  • Negative feedback cannot override antibody stimulation
    • Babies born to mothers with Graves’ may show transient hyperthyroidism
259
Q

A patient present with lethargy, dry skin and hair, constipation, cold intolerant . What is the diagnosis?

A

Hashimoto’s thyroiditis

260
Q

What is the pathophysiology of Hashimoto’s thyroiditis?

A

T2 and T4 hypersensitivity → anti-thyroid peroxidase antibodies (TPO) and anti-thyroglobulin antibodies and T + B cell thyroid infiltration

  • Goitre → enlarged thyroid infiltrated by T and B cells
    • Antibodies are not part of the diagnosis
  • Commonest cause of hypothyroidism in iodine-replete areas
261
Q

A patient present at 8yo with thirst, polyuria, malaise, urine dip confirms glycosuria. What is the diagnosis?

A

Type 1 DM

262
Q

What is the pathophysiology of T1DM?

A

T4 pathology → CD8+ T-cell (with T-cell clones) infiltration of pancreas leading to destruction of pancreatic islet cells + anti-islet cell, anti-insulin, anti-GAD and anti-IA-2 antibodies

  • Individuals with 3-4 of the above antibodies likely to develop T1DM
  • Detection of antibodies does not currently play a role in dx
263
Q

A patient present tired, pale, mild numbness of feet. Her blood results show anaemic (Hb 8.4), macrocytosis (MCV 108), urine dip negative (so not diabetes), folate normal, vitamin B12 very low . What is the diagnosis?

A

Pernicious anaemia

264
Q

What is the pathophysiology of Pernicious anaemia?

A

T2 reaction → autoantibodies against intrinsic factor → no absorption of vitamin B12 → pernicious anaemia and SCDC

  • Vit B12 deficiency → macrocytosis, fatigue, anaemia, pallor, etc.
  • Sub-acute Combined Degeneration of the Cord / SCDC à peripheral neuropathy, optic neuropathy
  • Antibodies to gastric parietal cells or intrinsic factor are useful in dx
265
Q

A patient present with drooping eyelids, weakness particularly on repetitive activity, symptoms worse at end of day. What is the diagnosis?

A

Myasthenia gravis

266
Q

What is the pathophysiology of myasthenia gravis?

A

T2 reaction → autoantibodies against ACh receptors → failure of depolarisation → absence of muscle action potential

  • Fluctuating weakness
  • Extraocular weakness, ptosis very common
  • EMG studies normal
  • Anti-ACh-R ABs present in ~75% patients à useful in diagnosis
  • Offspring of affected mothers may experience transient neonatal myasthenia
267
Q

What is the appropriate investigations for suspected myasthenia gravis?

A
  • Tensilon test positive (House episode)
    • Inject edrophonium (anti-cholinesterase) to prolong life of ACh and allow it to act on the receptors
268
Q

A patient present at 48yo with haemoptysis with widespread crackles in lungs, swelling of legs and reduced urine output.

Investigations show creatinine 472, microscopic haematuria and proteinuria, CXR – widespread shadowing, elevated TLCO suggesting pulmonary haemorrhage.

Further investigations show anti-neutrophil cytoplasmic antibody -ve, anti-basement membrane antibody +ve and crescentic nephritis on biopsy

What is the diagnosis?

A

Goodpasture’s

269
Q

What is the pathophysiology of Goodpasture’s?

A

T2 reaction → autoantibodies against basement membrane

270
Q

A patient presents with pain, stiffness and swelling of multiple small joints within hands, normochromic anaemia, high ESR, CRP. What is the diagnosis?

A

Rheumatoid arthritis

271
Q

What is the pathophysiology of Rheumatoid arthritis?

A
  • B cell involvement
    • T2 response – antibody binding to citrullinated proteins leading to
      • Complement activation
      • Macrophage activation via Fc R and complement receptors
      • NK cell activation with ADCC
    • T3 response
      • Immune complex formation – RF and anti CCP, deposition with complement activation
  • T cell involvement
    • T4 response – antigen presenting cells → CD4+ T cell → production of IFγ and IL17 → production of MMPs and IL1, TNFα
272
Q

What factors are involved in rheumatoid arthritis?

A
  • Genetic:
    • HLA DR4, HLA DR1 alleles → bind to citrullinated peptides with a higher affinity
    • PAD2 and PAD4 polymorphisms → associated with higher levels of citrullination
    • PTPN22 polymorphism (± CTLA4)
  • Environmental
    • Smoking → development of erosive disease, and increased citrullination
    • Gum infection with Porphyromonas gingivalis associated with RhA
      • Porphyromonas gingivalis expresses PAD enzyme
273
Q

What antibodies are found in Rheumatoid Arthritis?

A
  • Antibodies to cyclic citrullinated peptide (Anti CCP Antibody)
    • Bind to peptides in which arginine has been converted to citrulline by PAD
    • 95% specificity and 60% sensitivity for dx of RhA
  • Rheumatoid factor antibody (an antibody directed against Fc region of human IgG)
    • IgM anti-IgG most common RF antibody tested
    • Although IgA and IgG rheumatoid factors may present in some individuals
    • 60-70% specificity and sensitivity for dx of rheumatoid arthritis
274
Q

A patient present at 19yo with 4m history of fatigue, generalised arthralgia particularly small joints of hands, hair loss, mouth ulcers, butterfly rash. What is the diagnosis?

A

Systemic lupus erythematous - SLE

275
Q

What are the conditions associated with SLE?

A
  • CNS – seizures
  • Skin – butterfly rash, discoid lupus
  • Heart – endocarditis, myocarditis, serositis, pleuritis, pericarditis
  • Glomerulonephritis
  • Haematological – haemolytic anaemia, leukopenia, thrombocytopenia
  • Arthritis and lymphadenopathy
276
Q

What is the pathophysiology of SLE?

A

T3 hypersensitivity → anti-nuclear antibodies

  • Antibodies bind to antigens to form immune complexes
  • Immune complexes deposit in tissues → skin, joints, kidney
    • Activate complement → classical pathway
    • Stimulate cells expressing Fc and complement receptors
277
Q

What is the genetic predisposition to SLE?

A
278
Q

What immunological investigations are there for suspected SLE?

A
  • ANA → SLE = 1:640 titre
    • Anti-dsDNA
    • Anti-ENA
    • Anti-topoisomerase
    • Anti-centromere
  • Complement
    • Depletion of C4 and then C3
      • Inactive lupus = normal C3 and C4
      • Moderate active lupus = low C4
      • Very active lupus = low C3 and C4
279
Q

What are the targets of anti-nuclear antigens in SLE?

A
  • Anti-dsDNA Ab
    • Highly specific for SLE (95%)
    • High titres = severe disease - useful in disease monitoring
    • Homogenous staining on ELISA
  • Anti-ENA Ab
    • Ribonucleoproteins - Ro, La, Sm, U1RNP
      • May be found in SLE but not needed
    • Speckled pattern on ELISA
  • Anti-topoisomerase Ab - against enzymes (Anti-SCL70 AB)
    • Diffuse CREST
  • Anti-centromere Ab
    • Limited CREST
280
Q

What antibodies are found in Sjogren’s syndrome?

A

Anti-ENA → anti-Ro and anti-La

281
Q

What are the signs and symptoms of anti-phospholipid?

A
  • Recurrent venous or arterial thrombosis
  • Recurrent miscarriage
  • May occur alone/primary or in with autoimmune disease/secondary
282
Q

What are the two major antibodies in anti-phospholipid syndrome?

A
  • Anti-cardiolipin antibody
  • Lupus anti-coagulant → cannot be assessed if the patient is on anticoagulant therapy
283
Q

A patient presents with thickening around the mouth and microstomia. What is the diagnosis?

A

Systemic sclerosis

284
Q

What are the features of diffuse cutaneous systemic sclerosis?

A
  • GI, pulmonary & renal involvement
  • CREST features
    • Skin involvement of hands AND PROXIMAL PAST the forearms (unlike CREST)
285
Q

What is the pathophysiology of diffuse and limited cutaneous systemic sclerosis?

A
286
Q

What are the features of limited cutaneous systemic sclerosis?

A
  • ONLY the hands involved (does not spread proximally up the forearms)
  • Involves peri-oral skin
  • CREST-P
    • Calcinosis
    • Raynaud’s
    • Oesophageal dysmotility
    • Sclerodactyly
    • Telangiectasia
    • Primary pulmonary HTN
287
Q

A patient presents with weakness, malaise, periorbital heliotropic rash, Gottron’s papules. What is the diagnosis?

A

Idiopathic Inflammatory Myopathy → Dermatomyositis

288
Q

Describe dermatomyositis.

A
  • Periorbital rash + Gottron’s papules
    • Within muscle → perivascular CD4 T and B cells
    • Immune complex mediated vasculitis – T3 response
289
Q

Describe polymyositis.

A
  • No rash
    • Within muscle – CD8 T cells surround HLA Class 1 expressing myofibres
    • CD8 cells kill myofibres via perforin/granzymes – T4 response
290
Q

What antibodies are present in idiopathic inflammatory myopathy?

A
  • Positive ANA – extended myositis panel
    • Anti-Jo1 antibodies
    • Anti-Mi2
    • Anti-SRP (signal recognition peptide)
291
Q
A

SLE

292
Q

A patient presents at 54yo with recurrent nosebleeds and breathlessness. Cavitating pulmonary lesions on CXR. What is the diagnosis?

A

Systemic vasculitis

293
Q

What are the types of systemic vasculitis?

A
  • Large vessel
    • Takayasu’s arteritis
    • Giant cell arteritis / polymyalgia rheumatica
  • Medium vessel
    • Polyarteritis nodosa
    • Kawasaki disease
  • Small vessel ANCA-associated
    • Microscopic polyangiitis
    • Granulomatosis with polyangiitis (Wegner’s - crushed nose)
    • Eosinophilic granulomatosis with polyangiitis (Churg-Strauss)
  • Small vessel immune complex
    • Anti-GBM disease
    • IgA disease
    • Cryoglobulinemia
294
Q

What methods can be used to boost the immune system?

A
  • Vaccination
  • Replacement of missing components
  • Blocking immune checkpoints
  • Cytokine therapy
295
Q

What methods can be used to suppress the immune system?

A
  • Steroids
  • Anti-proliferative / Cytotoxic agents
  • Plasmapheresis
  • Inhibitors of cell signalling
  • Agents directed at cell surface antigens
  • Agents directed at cytokines
296
Q

Name the antigen presenting cells.

A
  • Dendritic cells
  • Macrophages
    • Langerhans cells
    • Mesangial cells
    • Kupffer cells
    • Osteoclasts
    • Microglia
  • B-lymphocytes
297
Q

Describe T-cell memory.

A
  • Longevity → persist without antigen via low level proliferation in response to cytokines
  • Different cell surface proteins → chemotaxis / adhesion to access non-lymphoid tissues
  • Rapid, robust response to subsequent antigen exposure
298
Q

Describe B-cell memory.

A
  • Longevity
  • Pre-formed, high affinity IgG antibodies
  • Rapid, robust response
299
Q

What are the aims of vaccines?

A
  • Memory → generate protective, long-lasting immune response
  • No adverse reactions
  • Practical
    • Single shot
    • Easy storage
    • Cheap
300
Q

How does the immune system fight influenza?

A
  • Although CD8 T cells control the viral load antibodies are responsible for providing a protective response
  • Anti-Haemagglutinin ABs
    • A membrane fusion glycoprotein of influenza virus
301
Q

How does protection from an influenza vaccine last?

A
  • Antibody protection begins 7 days after vaccine and last for around 6 months
302
Q

What test can be used to check for previous TB exposure?

A
  • Mantoux Test
    • Inject a small amount of liquid tuberculin intradermally
    • Area of injection is examined 48-72 hours after tuberculin injection
    • The reaction is an area of swelling around the injection site
      • Positive reaction wheal:
        • >5mm if high-risk → immunocompromised, living with someone with TB)
        • >10mm if medium-risk → healthcare professionals
        • >15mm if low-risk
303
Q

What are the different types of vaccine?

A
  • Live attenuated vaccines
  • Inactivated/component vaccines
  • DNA/RNA vaccines
  • Adenovirus
  • Dendritic cell vaccines
304
Q

Name some live attenuated vaccines.

A
  • MMR
  • BCG
  • Yellow fever
  • Typhoid
  • Polio (Sabin)
  • Vaccinia
  • Nasal influenza (aged 5-17)
305
Q

What are the advantages and disadvantages of live attenuated vaccines?

A
  • Advantages
    • Establishes infections - ideally mild symptoms
    • Raises broad immune response to multiple antigens - offer protection against different strains
    • Activates all phases of immune system
    • Often confer life-long immunity after one dose
  • Disadvantages:
    • Storage problems
    • Possible reversion to virulence - vaccine associated paralytic poliomyelitis (1 in 750,000)
    • Infection in immunosuppressed
    • Spread to contacts - spread to immunocompromised/immunosuppressed
306
Q

Name some inactivated vaccines.

A
  • Influenza
  • Cholera
  • Polio (Salk)
  • HAV
  • Pertussis (Whooping cough)
  • Rabies
307
Q

Name some component / subunit vaccines.

A
  • HbS antigen
  • HPV (capsid)
  • Influenza
308
Q

Name some toxoid vaccines.

A
  • Diphtheria
  • Tetanus
309
Q

What are the advantages and disadvantages of inactivated / component / toxoid vaccines?

A
  • Advantages:
    • No mutation or reversion Can be used in immunodeficient patients
    • Easier storage
    • Lower cost
  • Disadvantages:
    • Don’t follow normal route of infection
    • Poor immunogenicity
    • May require multiple injections, conjugates or adjuvants
310
Q

What are conjugate vaccines?

A

Polysaccharide + Protein carrier

  • Polysaccharide induces a T cell-independent B cell response (transient)
  • Protein carrier promotes T cell-dependant B cell response (long-term)
  • Examples
    • HiB
    • Meningococcus
    • Pneumococcus
311
Q

What are adjuvant vaccines?

A

Increase the immune response without altering its specificity → mimic the action of PAMPs on TLR and other PRRs

  • Useful in older people who produce a weaker immune response
  • Examples of adjuvants:
    • Aluminium salts - Hep A and B, HiB
      • Mechanism is not fully understood → could be:
        • Prolonged antigenic stimulation
        • Induce mild inflammation → development of immune response
        • Activate Gr1+ IL4+ eosinophils → prime naïve B cells à antibody response
    • Lipids (monophosphoryl lipid A; humans)
    • Oils (Freund’s adjuvant; animals)
    • ISCOMS
    • CpG DNA
312
Q

How do mRNA vaccines work?

A
  • Plasmid containing a gene of choice is harvest
  • The DNA is excised and transcribed to form mRNA
  • A complex containing the mRNA and lipids envelope is created - provides stability
  • mRNA enters cells and the protein is synthesised → immune response when presented on cell surface - spike protein is commonly used
  • Mimics the normal action of a virally infected cell and it stimulates T cell responses
313
Q

Describe adenoviral vector vaccines?

A

AstraZeneca, Sputnik COVID vaccine

  1. DNA of relevant protein inserted into viral vector
  2. Infect cells in vivo
  3. Transcription/translation to produce protein
  4. Stimulate immune response including B cells / antibodies and T cells
314
Q

What are the advantages and disadvantages of DNA/RNA vaccines?

A
  • Advantages
    • Mimics virus cell
    • May be used to develop a cancer vaccine
  • Disadvantages
    • Storage
    • In theory plasmid may integrate into host DNA → autoimmune diseases
315
Q

Describe dendritic cell vaccines.

A
  • Malignancy not infection → used against tumours where dendritic cell function may be compromised
  • Take a patient’s dendritic cells and load them with a tumour antigen and reintroduce them to the patient to try and boost the immune response against the tumour antigens
  • Requires antigens specific to the tumour and distinct from normal cells
  • Example
    • Sipuleucel-T Provenge → immunotherapy against prostate cancer
316
Q

What techniques work by replacing missing compounds?

A
  • Haematopoietic stem cell transplantation
  • Antibody replacement
  • Specific Immunoglobulin
  • Adoptive cell transfer
317
Q

What are the indications for haematopoietic stem cell transplantation?

A
  • Life-threatening immunodeficiency → e.g. SCID, leucocyte adhesion defect
  • Haematological malignancy
318
Q

What are the indications for antibody replacement therapy?

A
  • Primary antibody deficiency
    • Bruton’s X-linked hypogammaglobulinemia
    • X-linked hyper-IgM syndrome
    • Common variable immunodeficiency
  • Secondary antibody deficiency
    • Haematological malignancies
    • Post BM transplantation
319
Q

What are the indications for specific immunoglobulin?

A
  • Passive immunisation – human Ig used for post-exposure prophylaxis
    • HBV
    • Tetanus
    • Rabies
  • VZV Ig in early pregnancy (<20 weeks)
    • Acyclovir is contraindicated
320
Q

What are the different types of adoptive cell transfer?

A
  • Virus specific T cell therapy
  • Tumour infiltrating lymphocyte T cell therapy
  • T cell receptor T cells
  • CAR-T - Chimeric antigen receptor T cell therapy
321
Q

Describe virus specific T cell therapy.

A
  1. Blood is taken from the patient or from a matched individual
  2. Peripheral blood lymphocytes isolated → stimulated with pathogen peptides
  3. Expansion of pathogen-specific T cells → infused back into the patient
  • i.e. → EBV in those immunosuppressed to prevent development of B cell lymphoproliferative disease
    • Doesn’t involve any stimulation of B-cells → no B cell lymphoproliferative disease
322
Q

Describe tumour infiltrating lymphocyte T cell therapy.

A
  1. Remove tumour from patient
  2. Stimulate T cells within tumour with cytokines (e.g. IL-2) so they develop a response against tumour
  3. Select and expand the tumour infiltrating lymphocytes and reinfuse back into the patient
323
Q

Describe T cell adoptive therapies.

A
  • T cells taken from patient and viral / non-viral vectors used to insert gene fragments into T cells
  • Gene fragments encode receptors
    • TCR therapy
      • Insert a gene that encodes a specific TCR (e.g. against a tumour cell antigen)
        • Recognises MHC presented peptides
    • CAR therapy
      • Receptors are chimeric (it contains both B and T cell components)
        • Recognises cell surface CD markers
      • Standard CAR therapy = targeting CD19
        • Receptors on the CAR cell have an Ig-variable domain at the end which is joined onto the remainder of the TCR
        • Signals through the usual TCR pathway but recognises CD19 through an Ig-domain → T cells to kill the B cells
        • This is increasingly being used in ALL and NHL → not useful in solid tumours
324
Q

Name 3 immune checkpoint blockers.

A
  • CTL-A4 specific
    • Ipilimumab
  • PD-1 specific
    • Pembrolizumab
    • Nivolumab
325
Q

Describe ipilimumab.

A

Antibody specific for CTL-A4 → treat melanoma

  • CTLA4 and CD28 both expressed by T cells and both recognise the same antigens on APCs (CD80 and CD86)
    • APC CD80 and CD86 interact with CD28 → transmit a stimulatory signal
    • APC CD80 and CD86 interact with CTLA4 → transmit an inhibitory signal
  • Binds to CTLA4 → all CD80 and CD86 interactions occur through CD28 → boosted T cell response
  • Can cause autoimmune disease
326
Q

Describe Pembrolizumab and Nivolumab.

A

Specific for PD-1 → treat advanced melanoma and metastatic renal cell carcinoma

  • PD-1 is found on T-regulatory cells
  • Its ligands (PDL-1 and PDL-2) are present on APCs and some tumour cells → prevent death
  • ABs against PD-1 prevent the inhibitory effect of binding PD1 and PD1-L → activating the T cells to kill
  • Patients tend to develop autoimmune diseases - rheumatoid arthritis, thyroid disease, diabetes, SLE
327
Q

Describe some cytokine therapies.

A
  • INF-a – antiviral
    • Hepatitis B and C
  • INF-a2
    • Behçet’s disease
  • INF-g
    • Chronic granulomatous disease
  • IL-2 – increases clonal expansion
    • Renal cell cancer
328
Q

What is the mechanism of steroids in terms of immunosuppression?

A
  • Effects on Prostaglandins = inhibit phospholipase A2
    • Phospholipids blocked from becoming prostaglandins and leukotrienes which are proinflammatory
  • Effects on Phagocytes
    • Decrease chemotaxis
    • Decreased phagocytosis
    • Decreased release of proteolytic enzymes
  • Effects on Lymphocytes
    • Lymphopaenia (CD4 > CD8 > B cells)
    • Blocks cytokine gene expression
    • Decreased antibody production
    • Promotes apoptosis
329
Q

What are the side effects of corticosteroids?

A
  • Metabolic = Cushing’s Syndrome
    • Central obesity
    • Moon face
    • Diabetes
    • Lipid disorders
    • Osteoporosis
    • Hirsutism
    • Adrenal suppression
  • Cataracts
  • Glaucoma
  • Peptic ulceration
  • Pancreatitis
  • Avascular necrosis
  • Immunosuppression
330
Q

What are the indications for glucocorticoids?

A
  • Allergic disorder
  • Auto-immune
  • Auto-inflammatory
  • Transplantation
  • Malignant disease
331
Q

What is the overall mechanism of action of cytotoxic agents?

A

Inhibit DNA synthesis → cells with rapid turnover are most affected

332
Q

What forms of toxicity are cytotoxic agents?

A
  • BM suppression
    • Infection
    • Malignancy
  • Teratogenic
333
Q

Name at least 3 cytotoxic agents.

A
  • Cyclophosphamide
  • Mycophenolate
  • Azathioprine
334
Q

What is the mechanism of action of cyclophosphamide?

A

Alkylating agent

  • Alkylates guanine base of DNA
  • Damages DNA and prevent cell replication
  • Affects B cells > T cells
    • Used in antibody-mediated disorders
335
Q

What is the mechanism of action of cyclophosphamide?

A

Alkylating agent

  • Alkylates guanine base of DNA
  • Damages DNA and prevent cell replication
  • Affects B cells > T cells
    • Used in antibody-mediated disorders
336
Q

What are the indications for cyclophosphamide?

A
  • Multisystem connective tissue disease
  • Vasculitis with severe end-organ involvement - GPA, SLE
  • Cancer
337
Q

What are the side effects of cyclophosphamide?

A
  • Toxic to proliferating cells
    • Bone marrow suppression
    • Sterility (mainly in males)
    • Hair loss
  • Haemorrhagic cystitis - toxic metabolite is excreted in urine
  • Malignancy
    • Bladder cancer
    • Haematological malignancies
    • Non-melanoma skin cancer
  • Teratogenic
  • (Opportunistic) Infection
338
Q

What is the mechanism of action of azathioprine?

A

Purine analogue

  • Metabolised by the liver to 6-mercaptopurine = purine analogue
  • Blocks de novo purine synthesis (adenine and guanine) → prevents replication of DNA
  • Affects T-cells > B-cells
339
Q

What are the indications for azathioprine?

A
  • Transplantation
  • Auto-immune disease
  • Auto-inflammatory disease (e.g. Crohn’s, UC)
340
Q

What are the side effects of azathioprine?

A
  • Bone marrow suppression
    • Check TPMT activity before treatment started
  • Hepatotoxicity
  • Infection - less common than with cyclophosphamide
341
Q

What is the mechanism of action of mycophenolate mofetil?

A

Anti-metabolite

  • Blocks de novo guanosine nucleotide synthesis → prevents replication of DNA
  • Prevent T cell > B cell proliferation
342
Q

What are the indications for mycophenolate mofetil?

A
  • Transplantation
  • Auto-immune disease
  • Vasculitis
343
Q

What are the side effects of mycophenolate mofetil?

A
  • Bone marrow suppression
  • Teratogenic
  • Infection
    • HSV reactivation
    • Progressive multifocal leukoencephalopathy / JC virus
344
Q

What is Plasmapheresis?

A

Removal of pathogenic antibodies

  • Patient’s blood is passed through a separator and their own cellular constituents are reinfused
  • Plasma treated to remove Ig and is then reinfused (or replaced with albumin in plasma exchange)
345
Q

What are the issues with plasmapheresis?

A
  • Rebound antibody production
    • Although ABs gone plasma cells are still there → limits efficacy
    • Solution = co-administer an anti-proliferative agent (cyclophosphamide)
346
Q

What are the indications for plasmapheresis?

A
  • Severe antibody-mediated disease (type 2 hypersensitivity)
    • Goodpasture’s syndrome (anti-GBM)
    • Severe acute myasthenia gravis (anti-ACh-R)
    • Severe transplant rejection (antibodies against donor HLA)
347
Q

Name some inhibitors of cell signalling.

A
  • Calcineurin
    • Ciclosporin
    • Tacrolimus
  • mTOR
    • Rapamycin
  • JAK
    • Tofacitinib
    • Ruxolotinib
  • PDE4
    • Apremilast
    • Anagrelide
348
Q

What is the mechanism of ciclosporin and tacrolimus?

A

Inhibitors of calcineurin

  1. Prevent T cell signalling
  2. Blocks IL2 production
  3. Reduced activation of naive T-cells after successful APC interaction
  4. Reduced clonal expansion and proliferation
349
Q

What are the side effects of calcineurin inhibitors?

A
  • Nephrotoxicity - ciclosporin = tacrolimus
  • HTN - ciclosporin = tacrolimus
  • Neurotoxic - ciclosporin = tacrolimus
  • Diabetogenic - ciclosporin < tacrolimus
  • Dysmorphic features - ciclosporin
    • Hirsutism
    • Gingival hypertrophy
350
Q

What is the mechanism of rapamycin?

A
  • Inhibit T cell proliferation and function
    • Used in transplantation
351
Q

What is the mechanism of tofacitinib and and ruxolotinib?

A

JAK inhibitors / Jakinibs

  • Tofacitinib = JAK1 and JAK 3 inhibitor
  • Ruxolotinib = JAK 2 inhibitor
  • Interferes with JAK-STAT signalling
  • Influences gene transcription
  • Inhibits the production of inflammatory molecules
352
Q

What are the indications for tofacitinib and ruxolotinib (JAK inhibitors)?

A

Rheumatoid and Psoriatic arthritis

353
Q

What is the mechanism of apremilast and anagrelide?

A

PDE4 inhibitors

  • Increases cAMP → activate PKA → prevent activation of transcription factors
    • Decrease in cytokine production
354
Q

What are the indications for apremilast and anagrelide (PDE4 inhibitors)?

A

Rheumatoid and Psoriatic arthritis

355
Q

Match the following side effects with the most likely drug?

  • Osteoporosis
  • Infertility
  • Progressive multifocal leukoencephalopathy
  • Neutropenia (low TPMT)
  • Neuphrotoxicity
  • Cyclophosphamide
  • Prednisolone
  • Azathioprine
  • Tacrolimus
  • Mycophenolate
A
  • Osteoporosis = Prednisolone
  • Infertility = Cyclophosphamide
  • Progressive multifocal leukoencephalopathy = Mycophenolate
  • Neutropenia (low TPMT) = Azathioprine
  • Nephrotoxicity = Tacrolimus
356
Q

Name drugs that are directed at cell surface antigens?

A
  • T cells - ABA
    • Anti-thymocyte globulin (rabbit)
    • Basiliximab (anti-CD25, IL-2-R)
    • Abatacept (CTLA4-Ig)
  • T cell migration
    • Vedolizumab (anti-a4b7 integrin)
    • Natalizumab (anti-a4b7 integrin)
  • B cells
    • Rituximab (anti-CD20)
  • T cell, B cell, neutrophils, macrophages
    • Tocilizumab (anti-IL-6 receptor)
    • Sarlimumab (anti-IL-6 receptor)
357
Q

What are the indication for anti-thymocyte?

A
  • Allograft rejection - renal, heart
    • Daily IV infusion
  • Depletes T-cells
358
Q

What are the side effects of anti-thymocyte globulin?

A
  • Infusion reactions
  • Leucopaenia
  • Infection
  • Malignancy
359
Q

What is the mechanism of basiliximab?

A

Anti-CD25 / IL-2 receptor AB – inhibits T-cell proliferation

  • Targets parts of IL2 receptor (potential alpha, beta and gamma components)
    • Gamma chain shared amongst many IL-receptors, so not a good target
    • Alpha chain (aka CD25) more specific for the IL2 receptor
360
Q

What are the indications for basiliximab?

A

Prophylaxis of allograft rejection → before and after transplant

361
Q

What are the side effects of basiliximab?

A
  • Infusion reaction
  • Infection
  • Long-term malignancy risk
362
Q

What is the mechanism of abatacept?

A

CTLA4-Ig fusion protein - opposite of Ipilimumab

  • Abatacept = receptor made from a fusion of CTLA4 and IgG Fc component
  • CTLA4 and CD28 both expressed by T cells and both recognise the same antigens on APCs (CD80 and CD86)
    • APC CD80 and CD86 interact with CD28 → transmit a stimulatory signal
    • APC CD80 and CD86 interact with CTLA4 → transmit an inhibitory signal
      • Abatacept CTLA4-Ig binds APCs’ CD80 and CD86 = upregulated CTLA4-mediated reduced T cell activation
      • Ipilimumab anti-CTLA4 binds T-cell’s CTLA4 à blocks CTLA4-mediated reduced T cell activation à up activation
363
Q

What are the indications for abatacept?

A

Rheumatoid arthritis

364
Q

What are the side effects of abatcept?

A
  • Infusion reactions
  • Infection → screen for chronic infection (TB, HBV, HCV) before starting
  • Caution with malignancy
365
Q

What is the mechanism of rituximab?

A

Anti-CD20 - depletion of B cells

  • CD20 is expressed on mature B cells but not plasma cells → depletion of mature B cells
366
Q

What are the indications for rituximab?

A
  • Lymphoma
  • Rheumatoid arthritis
  • SLE
367
Q

What are the side effects of rituximab?

A
  • Infusion reactions
  • Infection - PML / JC virus
  • Exacerbation of cardiovascular disease
368
Q

What is the mechanism of vedolizumab and natalizumab?

A

Anti-alpha-4-beta-7 integrin – inhibits T-cell migration

  • Alpha 4 is expressed with either beta-1 or beta-7 integrin
  • This complex binds to MadCAM1 to mediate leukocyte epithelial rolling and arrest to enter tissues
  • Blocking this complex then inhibits leucocyte migration to tissues
369
Q

What are the indications for vedolizumab and natalizumab?

A
  • IBD
  • Remitting/relapsing MS
370
Q

What are the side effects of vedolizumab and natalizumab?

A
  • Infusion reactions
  • Infection - PML / JC virus
  • Hepatotoxic
  • Concerns regarding malignancy
371
Q

What is the mechanism of tocilizumab and sarlimumab?

A

Anti-IL-6 receptor AB - reduces activation of macrophages, T cells, B cells and neutrophils

  • Inhibits fusion of lymphoid and myeloid cells
372
Q

What are the indications for tocilizumab and sarlimumab?

A
  • Castleman’s disease - IL-6 producing tumour
  • Rheumatoid arthritis
373
Q

What are the side effects of tocilizumab and sarlimumab?

A
  • Infusion reactions
  • Infection
  • Hepatotoxic
  • Elevated lipids
  • Caution with malignancy
374
Q

Name drugs that are directed at cytokines?

A
  • Anti-TNFa
    • Infliximab
    • Adalimumab
    • Certolizumab
    • Golimumab
  • TNF decoy receptor
    • Etanercept
  • Anti-IL23
    • Ustekinumab (also anti-IL-12)
    • Guselkumab
  • Anti-IL-17
    • Secukinumab
  • Anti-RANK ligand
    • Denosumab
375
Q

What is the mechanism of infliximab, adalimumab, certolizumab and golimumab?

A

Anti-TNFa - bind to TNFa

376
Q

What are the indications for infliximab, adalimumab, certolizumab and golimumab?

A
  • Rheumatoid arthritis
  • Ankylosing spondylitis
  • Psoriasis and psoriatic arthritis
  • Inflammatory bowel disease
  • Familial Mediterranean fever
  • Other monogenic inflammatory diseases
377
Q

What are the side effects of infliximab, adalimumab, certolizumab and golimumab?

A
  • Infusion or injection site reactions
  • Infection (TB, HBV, HCV)
  • Lupus-like conditions
  • Demyelination
  • Malignancy
378
Q

What is the mechanism of etanercept?

A

TNFa antagonist / decoy receptor → Inhibits action of TNF-alpha and TNF-beta

379
Q

What are the indications for etanercept?

A
  • Rheumatoid arthritis
  • Ankylosing spondylitis
  • Psoriasis and psoriatic arthritis
380
Q

What are the side effects of etanercept?

A
  • Injection site reactions
  • Infection (TB, HBV, HCV)
  • Lupus-like conditions
  • Demyelination
  • Malignancy
381
Q

Describe IL-1 productions and state some conditions that can be managed by blocking it?

A
  • Familial Mediterranean fever → MEFV gene in mutated some abnormal pyrin-marenostrin
  • Gout → urate and the cryopyrin pathway
  • Fever
  • Adult onset Stills disease
382
Q

Describe IL-6 productions and state some conditions that can be managed by blocking it?

A

Rheumatoid arthritis

383
Q

What is the mechanism of ustekinumab?

A

Anti-p40 of IL-12 and IL-23

  • Targeting p40 will inhibit both cytokines
    • IL-12 = p40 and p35
    • IL-23 = p40 and p19
  • These cytokines mainly act on T cells and NK cells thereby modulating their activity
384
Q

What are the indications for ustekinumab?

A
  • Psoriasis and psoriatic arthritis
  • Crohn’s disease
385
Q

What are the side effects of ustekinumab?

A
  • Injection site reactions
  • Infection (TB)
  • Concern about malignancy
386
Q

What is the mechanism of guselkumab?

A

Anti-p19a in IL-23

  • IL-23 comprises p40 and p19
    • Acts on T cells and NK cells thereby modulating their activity
387
Q

What are the indications for ustekinumab?

A
  • Psoriasis
388
Q

What are the side effects of ustekinumab?

A
  • Injection site reactions
  • Infection (TB)
  • Concern about malignancy
389
Q

What is the mechanism of secukinumab?

A

Anti-IL17a

  • Dimer of IL17A or IL17A/F will bind to IL17RA/RC receptor
390
Q

What are the indications for secukinumab?

A
  • Psoriasis and psoriatic arthritis
  • Ankylosing spondylitis
391
Q

What are the side effects of secukinumab?

A
  • Infection (TB)
392
Q

Which interleukins are important in asthma and eczema?

A
  • IL-4, IL-5 and IL-13 → Th2 and eosinophil response
    • IL-4/13 blockade antibody = asthma and eczema
    • Anti-IL-13 antibody = eczema
    • Anti-IL-5 antibody = eosinophilic asthma
393
Q

What is the mechanism of denosumab?

A

Anti-RANK-ligand antibody

  • RANKL from osteoblasts acts on RANK receptor on osteoclasts → osteoclast differentiation → resorb bone
  • Denosumab is an antibody directed against RANKL = prevents RANKL binding to RANK on osteoclasts
394
Q

What are the indications for denosumab?

A

Osteoporosis

395
Q

What are the side effects of denosumab?

A
  • Injection site reactions
  • Infection - mildly immunosuppressive as RANKL has some role in other parts of the immune system
  • Avascular necrosis of the jaw