
Contents
-
-
-
-
-
-
-
-
-
-
-
Introduction Introduction
-
Flow cytometry Flow cytometry
-
Tissue culture Tissue culture
-
Proliferation assays Proliferation assays
-
Immunohistology Immunohistology
-
Cytokine, chemokine, soluble protein assays Cytokine, chemokine, soluble protein assays
-
Apoptosis assays Apoptosis assays
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
Adhesion markers Adhesion markers
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
Bronchoalveolar lavage (BAL) studies Bronchoalveolar lavage (BAL) studies
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
CD40 ligand expression CD40 ligand expression
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
Complement membrane regulatory factors Complement membrane regulatory factors
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
Cytokine and cytokine receptor measurement Cytokine and cytokine receptor measurement
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
Cytotoxic T cells Cytotoxic T cells
-
FOXP3 (regulatory T cells—IPEX syndrome) FOXP3 (regulatory T cells—IPEX syndrome)
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
Genetic and protein studies Genetic and protein studies
-
Leukaemia phenotyping Leukaemia phenotyping
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
Leukaemia phenotyping panel Leukaemia phenotyping panel
-
-
Lymphocyte subsets Lymphocyte subsets
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
B-cell function: in vivo assays B-cell function: in vivo assays
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
B-cell function: in vitro assays B-cell function: in vitro assays
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
T-cell function: in vivo assays T-cell function: in vivo assays
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
T-cell function: in vitro assays T-cell function: in vitro assays
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
Lymphoma diagnosis Lymphoma diagnosis
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
Neutrophil function testing Neutrophil function testing
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
NK-cell function NK-cell function
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
NK granule release NK granule release
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
Perforin expression Perforin expression
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
Toll-like receptors (TLRs) Toll-like receptors (TLRs)
-
Principles of testing Principles of testing
-
Indications for testing Indications for testing
-
Interpretation Interpretation
-
-
TRECs TRECs
-
T-cell receptor and immunoglobulin heavy-chain gene rearrangements T-cell receptor and immunoglobulin heavy-chain gene rearrangements
-
-
-
-
-
-
Cite
Introduction
Cellular investigations include:
identification of cell-surface phenotype
identification of intracellular proteins
identification of cellular function, including activation
identification of secreted products (cytokines, chemokines)
identification of abnormal cellular constituents (leukaemia/lymphoma).
Few functional assays are standardized and gold standard assays have not been defined.
EQA exists only for basic lymphocyte phenotyping and then only in the context of testing for HIV.
Flow cytometry
Flow cytometry provides the cornerstone of diagnostic cellular immunology and depends on the availability of monoclonal antibody reagents that react with human surface and intracellular antigens. Fluorescent intercalating dyes can be used to detect DNA semi-quantitatively (for cell cycle analysis).
The technique involves the flow of fluorescent-labelled cells past the exciting laser and subsequent detectors.
Method is only applicable to single-cell suspensions, e.g. blood-derived cells or cultured cells.
It is possible to use disaggregated solid tissues, such as tumours.
Modern flow cytometers use a single exciting laser (monochromatic light) and can detect multiple different wavelengths of light emitted by fluorescent dyes.
There are detectors for forward and 90° light scatter, which are related to cell size and cell granularity, respectively.
Software permits complex multiparameter gating and analysis, including real-time data collection and analysis.
Fluorescent-conjugated monoclonal antibodies are used against surface antigens.
Cell permeabilization techniques are available to enable staining of intracellular antigens.
Surface and intracellular stains can be combined.
Appropriate controls are required to detect non-specific staining.
The major advantage of flow cytometry for analysis is that it is semi-automated and can analyse very large numbers of cells very rapidly compared with fluorescence microscopy. It is much more accurate.
Accurate absolute counts are available on single-platform analysers using bead technology. This obviates the errors from using haematology counter total lymphocyte counts.
Regular calibration of the instrument is required, and it is essential that compensation between the fluorescence detectors is correctly set up. This is done with beads of known fluorescence.
Tissue culture
In vitro functional studies of cells may require purified cells (blood, other fluids).
This is done by density gradient centrifugation, using Ficoll, metrizamide, or dextran solutions.
The different buoyant densities of blood cells permit separation when blood is centrifuged through the dense medium.
Further purification of lymphocyte populations can be undertaken using:
rosetting with sheep red cells
magnetic separation using monoclonal antibodies coupled to magnetic microspheres.
The more cells are handled in vitro, the more their characteristics are altered.
This affects activation parameters in particular.
Cell culture is usually carried out in tissue-culture medium supplemented with:
antibiotics to prevent contamination with bacteria
fetal calf serum (FCS) or human AB serum (no isoagglutinins)
other ‘black-box’ factors that are required for optimal cell growth (glutamine is added as this is labile).
Where proliferation assays are being carried out, it is essential to screen the FCS first, as some batches are mitogenic in their own right.
Good sterile technique is essential.
Culture is carried out in a 37°C humidified incubator, with a controlled atmosphere (usually 5% CO2) to maintain pH.
Many different types of tissue culture media are available. Most contain pH buffers (bicarbonate) and pH indicators.
Proliferation assays
There are numerous mitogenic stimuli that can be used (see ‘T-cell function: in vitro assays’, p.570).
These are added at the initiation of the culture.
Cells are pulsed with tritiated thymidine, which is taken up into newly synthesized DNA in dividing cells: this remains the gold standard.
Cells are then harvested onto filter papers and exposed to scintillant fluid.
Counts per minute are determined using a beta-counter.
Alternative assays have been described using flow cytometers (no radio-isotopes):
CD69 expression
DNA analysis with intercalating dyes (cell cycle analysis).
These do not give comparable results to those from tritiated thymidine uptake and appear less sensitive.
Immunohistology
Immunoperoxidase and other enzymatic immunostains are used in the diagnosis of lymph-node disease.
Multiple monoclonal antibodies which recognize different stages of lymphoid development or particular subsets of cells are used.
Many of the antibodies used will also work on paraffin-embedded sections, but this depends on whether the target antigen is stable under the conditions of fixation. Frozen sections are better at present.
In situ hybridization is used to detect viral nucleic acid (EBV, other herpesviruses).
Cytokine, chemokine, soluble protein assays
Detection of specific cellular products, such as antibodies, cytokines, and shed surface molecules (soluble CD8 etc.) are usually undertaken using EIA or RIA techniques, as described in Chapter 18.
Cytokines may also be detected by bioassays using cell lines whose growth depends on a given cytokine.
Strictly, both types of assay should be used, as EIA techniques may give spurious results due to naturally occurring cytokine-binding proteins in serum (soluble receptors, binding factors).
Bioassays are notoriously difficult to standardize and reproduce, and are not suited to routine diagnostic use.
Detection of intracellular cytokines with fluorescently labelled monoclonal antibodies in permeabilized cells has been used in conjunction with surface staining.
This does not indicate that the cytokines are secreted and therefore does not equate to functional assays of cytokines.
Apoptosis assays
Principles of testing
Flow cytometric assays exist for identification of degraded DNA in apoptotic cells.
Preferred method is the TUNEL method, using fluorescent nucleotides enzyme-inserted into DNA strand breaks present in apoptotic cells. Commercial assays are available.
Fluorochrome-conjugated annexin V can be used to detect surface phosphatidylserine which is exposed on the cell surface in apoptotic cells, but not in normal cells.
Expression of fas and fas-ligand by flow cytometry.
A functional assay is available using PHA+IL-2 stimulated T cells. These express high levels of Fas and can be induced to apoptose by the addition of Fas-ligand. Co-staining with annexin V (apoptotic cells) and propidium iodide (identifies dead cells) allows the response of the patient’s cells to be compared with a normal control. To improve the reliability of the assay, multiple dilutions of fas ligand are used.
Protein and molecular follow-up tests are required to confirm defects: few PID centres have the capacity to run the necessary assays.
Indications for testing
Suspected apoptotic defect (ALPS, caspase deficiency).
Interpretation
Careful use of controls is required.
Samples must be run fresh.
Microscopic confirmation of assay results is advised to exclude artefacts.
Adhesion markers
Principles of testing
Flow cytometry.
Analysis should be carried out with CD18, CD11a (LFA-1), CD11b (Mac-1, CR3), and CD11c (CR4) for LAD-1, and CD15 for LAD-2. Neutrophils and lymphocytes should be tested.
Stimulation studies for upregulation in the presence of PMA or γ-IFN may be required where there is partial expression of CD18.
Indications for testing
Suspected leucocyte adhesion molecule deficiency (see Chapter 1).
Interpretation
LAD-1 is associated normally with deficiency of CD18, the common β-chain for the integrins, which leads to absence of CD11a, CD11b, and CD11c, as well as CD18.
Absence of β-chains has been reported but is extremely rare.
LAD-2 is exceptionally rare and is associated with deficiency of the hapten-X receptor on neutrophils (CD15).
Under certain circumstances it may be appropriate to look at the expression of the other complement receptors: CR1 (expressed on red cells, eosinophils, and B cells) and CR2 (CD21, EBV receptor expressed on B cells, NK cells, and follicular dendritic cells).
Reduction of red cell CR1 has been found in SLE.
Some patients with CVID may lack CD21 on some of their B cells.
Bronchoalveolar lavage (BAL) studies
Normal adult values for non-smokers:
total cells, 130–180×103/mL
macrophages, 80–95%
lymphocytes, <15%
neutrophils, <3%
eosinophils, <0.5%.
Normal adult values for smokers:
total cells, 300–500×103/mL
macrophages, 85–98%
lymphocytes, <10%
neutrophils, <5%
eosinophils, <3%.
Principles of testing
Cells recovered from bronchi by saline lavage during bronchoscopy can be stained and counted using neat BAL fluid. Total count and percentage differential counts are required.
Subsets of lymphocytes can be analysed by flow cytometry.
Indications for testing
Unexplained interstitial lung disease.
Sarcoidosis.
Hypersensitivity pneumonitis.
Idiopathic pulmonary fibrosis (IPF).
Eosinophilic granuloma.
Connective tissue diseases.
Interpretation
In sarcoidosis, there is a marked increase in lymphocytes (to about 30% of the total cells), predominantly CD4+ T cells, giving a CD4:CD8 ratio (which is normally 2:1) of between 4:1 and 10:1.
Values improve with treatment, but the levels and the ratio do not predict the severity of the disease.
Occasionally there is an increase in neutrophils and mast cells, which is said to indicate a poorer prognosis.
In hypersensitivity pneumonitis, the BAL lymphocytosis comprises mainly CD8+ cells, with the highest levels occurring in the acutely exposed.
In IPF, a neutrophilia in excess of 10%, particularly if there is an increase in eosinophils, is associated with a poor prognosis.
A lymphocytosis (a rare finding) is associated with a better prognosis and indicates a probable response to steroids.
In eosinophilic granuloma (histiocytosis X), there is an increase in OKT6-positive (S-100, CD1+) histiocytic cells, up to 20% of total cells, which is diagnostic.
CD40 ligand expression
Principles of testing
Flow cytometry is used to demonstrate upregulation of expression of CD40-ligand (CD154) upon stimulation of T cells in vitro with mitogens (PMA).
CD69 expression is used as an activation control.
Indications for testing
Suspected CD40-ligand deficiency.
Interpretation
Gating stimulated cells can be difficult because of clumping and size changes: this makes the activation control important.
Variants of CD40-ligand deficiency have been identified in which there is normal upregulation of non-functional ligand. Normal results do not exclude the diagnosis.
Failure of upregulation is highly suggestive of CD40-ligand deficiency.
Abnormal results should be followed up with genetic testing for mutations in the CD40-ligand gene.
Complement membrane regulatory factors
Principles of testing
Flow cytometry is now used exclusively.
Functional assays of cell lysis (Ham’s test) have been withdrawn.
Indications for testing
Suspected paroxysmal nocturnal haemoglobinuria.
Interpretation
Deficiencies of a group of surface proteins with an unusual glycosyl-phosphatidylinositol membrane binding are associated with paroxysmal nocturnal haemoglobinuria (PNH).
This is a clonal disorder leading to unusual susceptibility to homologous complement lysis, particularly of red cells.
The proteins in question are regulatory proteins which prevent destruction of cells by homologous complement and include:
decay accelerating factor (DAF, CD55)
homologous restriction factor-20 (HRF20, CD59)
C8-binding protein (HRF65)
acetylcholinesterase.
Cytokine and cytokine receptor measurement
Principles of testing
Enzyme immunoassay (serum, cell culture supernatant).
Bioassay.
Flow cytometry for intracellular cytokines; surface staining for receptors.
Immunoblotting.
In vitro stimulation assays with mycobacterial and salmonella antigens may be required to demonstrate defects.
Elispot assays can identify specific cytokine production in response to stimulation.
Indications for testing
Only absolute indication is suspected cytokine/cytokine receptor deficiency (e.g. IL-12, γ-IFN receptor deficiencies).
Intracellular cytokines have been used to identify functional Th1/Th2 balance.
Interpretation
EIA assays are unreliable because of the presence of natural cytokine-binding proteins and soluble receptors.
Bioassays are difficult to standardize, time-consuming, and unsuitable for routine diagnostic use.
IL-6 rises very early in acute-phase responses, before a rise in the CRP can be detected. However:
CRP is readily available and an is acceptable surrogate for IL-6
CRP levels are raised in myeloma, reflecting elevated IL-6
CRP levels are also raised in Castleman’s syndrome, reflecting raised IL-6.
Cytokine and cytokine receptor deficiencies are exceptionally rare (see Chapter 1).
Flow cytometric tests for intracellular cytokine detection are available.
Technique works well for IL-2 and γ-IFN but poorly for IL-4.
It has the significant advantage that specific T-cell subpopulations can be studied using multicolour flow cytometry.
Cytotoxic T cells
Cytotoxic T cells can be generated during a one-way mixed lymphocyte reaction (sMLR), stimulating the responding cells with irradiated or mitomycin-treated allogeneic target cells and then assessing the ability of the responders to kill Cr51-labelled targets, in a similar assay to the NK-cell assay (see ‘NK-cell function’, p.575).
This is a complex and fiddly assay, and has been used mainly as part of the cross-matching procedure (see Chapter 21).
FOXP3 (regulatory T cells—IPEX syndrome)
Principles of testing
Flow cytometric test to detect the presence of regulatory T cells (Treg) by intracellular detection of FOXP3.
This requires a permeabilization step on separated lymphocytes.
Treg are FOXP3+ CD4+ CD25bright and CD127weak (CD127 is IL-7 receptor-A).
Indications for testing
Investigation of suspected IPEX syndrome (see Chapter 1).
Interpretation
Assays that involve permeabilization of separated lymphocytes are intrinsically more prone to technical problems.
The assay must be run on fresh samples with a normal control.
This is a screening, not a quantitative, assay and will also pick up non-functional FOXP3—essential to do follow-up genetic analysis.
Genetic and protein studies
Protein and genetic studies are essential for the identification of gene defects in primary immunodeficiencies.
Surface proteins and some intracellular proteins, relevant to the diagnosis of primary immune deficiencies, can be identified by flow cytometry.
Protein studies, including surface and intracellular protein detection, are frequently used as a screening test prior to genetic testing, e.g. for:
XLA (BTK)
CGD (phox proteins)
CD40 ligand deficiency
IPEX (FOXP3)
ICOS deficiency (ICOS)
SAP, XIAP
CD3ζ.
Abnormal protein expression should be followed up by molecular mutation analysis.
Molecular analysis is also required where there is a high degree of clinical suspicion but apparently normal protein expression (expression of non-functional protein).
Family studies are valuable to identify asymptomatic carriers, who can then receive appropriate counselling.
Leukaemia phenotyping
Leukaemia phenotyping is undertaken to identify the origin of the malignant cell and the presence or absence of markers that are known to be of prognostic significance. This will always be undertaken in conjunction with other studies, including examinations of blood films, bone marrow smears, and trephines stained for enzymatic cytoplasmic and membrane markers.
Principles of testing
Flow cytometry of peripheral blood and bone marrow:
surface markers
intracellular markers.
Morphology on peripheral blood and bone marrow.
Enzymatic studies.
Molecular studies:
oncogene expression
Ig heavy chain and Tcr gene rearrangements
karyotype and chromosomal abnormalities.
Indications for testing
Suspected leukaemia or pre-leukaemia.
Interpretation
Diagnosis depends on the use of multiple markers and techniques.
Follow-up panels may be required (see next section).
Leukaemic cells often correspond to particular stages of cellular differentiation, which can be matched to normal cell ontogeny.
Aberrant antigens, expressed out of sequence, may occur.
This may give rise to biphenotypic leukaemias.
Bone marrow studies are complex because of the very different light-scattering properties of the cellular constituents.
Familiarity with the patterns of antigen expression at each stage of differentiation for each lineage is required.
Re-examination of bone marrow after treatment is important to detect the presence of minimal residual disease. This can be done using the following techniques.
Flow cytometry, which can detect one leukaemic cell in 10 000 cells.
PCR techniques where the leukaemic cells carry an abnormal genetic marker (oncogene) or have a specific rearrangement of either the immunoglobulin (B lineage) or T-cell receptor (T lineage) genes. These techniques are even more sensitive.
Flow cytometric karyotyping is now possible as an alternative to molecular techniques.
Leukaemia phenotyping panel
A primary panel for acute leukaemias will usually include the following.
B lineage:
CD10 (CALLA), CD19, CD24, HLA-DR, cytoplasmic Ig (μ heavy chains) and surface Ig.
T lineage:
CD2, cytoplasmic CD3, CD7.
Lymphoblast:
TdT.
AML lineage:
CD13, CD14, CD33.
Erythroid:
glycophorin A.
Megakaryocyte:
CD41.
A secondary panel may be used in difficult cases and may include the following.
B lineage:
cytoplasmic CD22.
T lineage:
CD1, CD3, CD4, CD8.
AML:
CD15, cytoplasmic myeloperoxidase.
For chronic lymphoid disorders the panel will be slightly different, as follows.
B lineage, primary:
CD10, CD20, CD5, surface Ig.
B lineage, secondary:
CD11c, CD25, CD38, and FMC7.
T lineage, primary:
CD3.
T lineage, secondary:
CD4, CD8, CD11b, CD16, CD57.
Lymphocyte subsets
Normal adult ranges:
• . | total T cells (CD3+) . | 0.69–2.54 109/L . |
---|---|---|
• | CD4+ T cells | 0.41–1.59 109/L |
• | CD8+ T cells | 0.19–1.14 109/L |
• | total B cells (CD19+) | 0.09–0.66 109/L |
• | NK cells (CD16+ CD56+) | 0.09–0.56 109/L |
• | activated T cells (CD3+CD25+) | 0.1–0.4 109/L |
• . | total T cells (CD3+) . | 0.69–2.54 109/L . |
---|---|---|
• | CD4+ T cells | 0.41–1.59 109/L |
• | CD8+ T cells | 0.19–1.14 109/L |
• | total B cells (CD19+) | 0.09–0.66 109/L |
• | NK cells (CD16+ CD56+) | 0.09–0.56 109/L |
• | activated T cells (CD3+CD25+) | 0.1–0.4 109/L |
Principles of testing
Single-platform flow cytometry is considered the gold standard.
Fluorescence microscopy should not be used.
Direct conjugation of the fluorochrome to the antibody is preferred.
Beads are used to calibrated absolute counts.
Results should be reported as absolute counts: percentages and ratios are not helpful for the basic markers but may be useful for extended panels (see next section).
Robust EQA schemes exist for common markers.
Indications for testing
Diagnosis and monitoring of immunodeficiency states.
Monitoring immunotherapeutic agents (anti-T-cell antibodies, cytotoxic drugs).
Interpretation
Wide availability of commercial reagents with different fluorochromes allows many permutations and combinations, using multichannel flow cytometers.
Some of the fluorochromes are large molecules, and multiple staining of markers on cells may give rise to steric hindrance and reduced binding.
Correct set-up of compensation for the flow cytometer is essential. This must be checked regularly and especially after servicing.
Regular quality control checks should be carried out with commercial fluorochrome coupled beads.
A basic panel for primary immunodeficiency work should include the following.
T cells: CD3, CD4, CD8.
B cells: CD19 or CD20.
NK cells: CD16 and CD56.
activated cells: CD25, MHC class II.
Additional markers may include the following.
CD45RA, CD45RO, CD27 (naive and effector T cells):
CD4+CD45RA+CD27+ = naive T cells
CD4–CD45RA+CD27+ = naive T cells
CD4–CD45RA+CD27– = effector T cells.
CD27, sIgM, sIgD (naive, memory, and class-switch memory):
CD27–sIgM+sIgD+ = naive B cells
CD27+sIgM+sIgD+ = memory B cells
CD27+sIgM–sIgD– = class-switch memory B cells.
Tcr aβ and γd.
Leucocyte adhesion and complement receptors (see above).
MHC class I (bare lymphocyte syndrome).
Lymphocyte numbers have a marked circadian rhythm. In serial monitoring, samples must be taken at the same time of day.
In a baby with suspected SCID, the presence of mainly activated CD8+ T cells raises suspicion of materno-fetal engraftment, while the presence of activated CD4+ T cells, in the presence of large numbers of eosinophils, is suggestive of Omenn’s syndrome.
T-cell receptor gene rearrangements will show an oligoclonal response.
Absence of CD8+ T cells is a feature of ZAP-70 kinase deficiency.
Abnormalities of T- and B-cell populations are also seen in CVID, with CD4+ T-cell lymphopenia, affecting particularly CD45RA+ T cells, and absence of class switch memory B cells.
After HSCT, high levels of activated T cells often indicate GvHD.
Very low CD4+ T-cell counts are not a diagnostic feature of HIV disease.
Temporary reductions in the CD4+ T-cell count are seen with a number of trivial viral infections, particularly in the acute phase.
This is often accompanied by an elevation of the CD8+ T cells and NK cells.
Lymphocyte phenotyping must not be used as a surrogate for HIV testing without consent.
CD4:CD8 ratio is of little value. Risk of opportunist infections is determined by absolute CD4 count.
Basic panel of CD3, CD4, and CD8 is all that is required (with viral load monitoring).
Recovery of cell numbers may be seen with HAART.
Persistent CD4+ T-cell lymphopenia has also been reported as a cause of opportunistic infections in the absence of any evidence for infection with either HIV-1 or HIV-2 (idiopathic CD4+ T-cell lymphopenia).
Rare deficiency of the binding site for the anti-CD4 mAb OKT3 is recognized, giving spuriously low CD4 counts. This variant CD4 appears functionally normal.
Abnormal lymphocyte profiles are also seen in:
lymphoma
malignancy
chronic fatigue syndromes
protein-losing enteropathy
overtraining syndrome.
Generalized proportionate reductions in lymphocyte counts are seen with long-term immunosuppressive therapy.
CD4/CD8 double-negative Tcr γδ T cells are increased in ALPS (see Chapter 1).
B-cell function: in vivo assays
Principles of testing
In vivo antibody production is measured by detection of serum antibody levels and rise in titre after deliberate test immunization.
Antibody levels should be measured to exposure and immunization antigens.
Testing should include protein and polysaccharide antigens (see Table 20.1).
Subclass-specific responses can be measured to some antigens.
Serotype-specific responses can be measured to pneumococcal polysaccharides (not all serotypes are equally immunogenic).
Isohaemagglutinins, in appropriate blood group patients allow detection of IgM responses.
In the USA, the bacteriophage Φ'174 is used as an immunogen. This neoantigen permits detection of primary and secondary antibody responses.
Antibodies will normally be detected by EIA on serum, or by complement-fixation assays (viral antibodies).
ELISPOT assays allow the detection of specific antibody-producing B cells.
EQA schemes exist for viral and bacterial antibodies.
Antigen . | Type of antigen . | Isotype/subclass . | Robust assays available? . |
---|---|---|---|
Pneumovax® | Polysaccharide | IgG, IgG2 | Yes? |
Salmonella Vi | Polysaccharide | IgG, IgG2 | Yes |
Tetanus toxoid | Protein | IgG, IgG1 | Yes |
Diphtheria toxoid | Protein | IgG, IgG1 | No |
Hib capsular polysaccharide | Polysaccharide | IgG, IgG1 (when conjugated to protein), IgG2 | Yes |
Meningococcal group C capsular polysaccharide | Polysaccharide | IgG, IgG1 (when conjugated to protein), IgG2 | No |
Poliovirus | Protein | IgG, IgM, IgG1, IgG3 | Yes |
MMR | Multiple proteins | IgG, IgM, IgG1, IgG2 | Yes |
Hepatitis B virus surface antigen | Protein | IgG, IgG1, IgG3 | Yes |
Isohaemagglutinins | Polysaccharides | IgM | Yes |
Antigen . | Type of antigen . | Isotype/subclass . | Robust assays available? . |
---|---|---|---|
Pneumovax® | Polysaccharide | IgG, IgG2 | Yes? |
Salmonella Vi | Polysaccharide | IgG, IgG2 | Yes |
Tetanus toxoid | Protein | IgG, IgG1 | Yes |
Diphtheria toxoid | Protein | IgG, IgG1 | No |
Hib capsular polysaccharide | Polysaccharide | IgG, IgG1 (when conjugated to protein), IgG2 | Yes |
Meningococcal group C capsular polysaccharide | Polysaccharide | IgG, IgG1 (when conjugated to protein), IgG2 | No |
Poliovirus | Protein | IgG, IgM, IgG1, IgG3 | Yes |
MMR | Multiple proteins | IgG, IgM, IgG1, IgG2 | Yes |
Hepatitis B virus surface antigen | Protein | IgG, IgG1, IgG3 | Yes |
Isohaemagglutinins | Polysaccharides | IgM | Yes |
Indications for testing
Suspected antibody deficiency.
Interpretation
Full infection and immunization history is required to evaluate responses.
Dynamic responses after immunization give a better view of B-cell function.
Target should be a rise into the normal/protective range with a minimum of a fourfold rise in titre.
Assays for bacterial antibodies are poor with CVs of 15–25%. Pre- and post-immunization samples should be run on the same assay.
Assays must be interpreted with caution.
Only killed or subunit vaccines should be given to patients with suspected immunodeficiency.
Role of subclass- and serotype-specific assays is uncertain at present. Multiplex assays may be valuable for rapid screening of responses to multiple serotypes.
Move to conjugated polysaccharide vaccines may lead to loss of pure polysaccharides for test immunization.
B-cell function: in vitro assays
Principles of testing
In vitro B-cell function is usually tested by stimulation of purified mononuclear cells by:
pokeweed mitogen (PWM)
anti-IgM + IL-2
Staphylococcus strain A Cowan (SAC)
Epstein–Barr virus (EBV).
IgG, IgA, and IgM production can be measured at 7 days by sensitive ELISA of the supernatant.
Testing is time-consuming.
Indications for testing
There are few clinical indications for this at present.
Flow cytometric detection of class-switch memory B cells is quicker and easier than using anti-IgM + IL-2 system to identify prognostically important subgroups of common variable immunodeficiency (Chapter 1).
Interpretation
Interpretation depends on the type of assay used and the establishment of appropriate ranges for age and sex.
T-cell function: in vivo assays
Principles of testing
T-cell function in vivo is tested by delayed-type hypersensitivity.
Antigens are pricked through the skin (Merieux Multitest CMI®) or injected intradermally.
Most useful antigens include PPD, Candida, mumps, tetanus, and streptokinase/streptodornase, which are available (some with difficulty) as single antigens, or are part of the battery in the Multitest.
Indications for testing
Testing is of limited value except in the circumstance of chronic mucocutaneous candidiasis, where there is often specific anergy to Candida with reasonable responses to other antigens.
Interpretation
There may be early reactions but these are due to mechanisms not involving T cells.
At 72–96 hours, in a positive reaction, there will be a cellular infiltrate that is palpable, with overlying erythema.
Reactivity to the panel is low in early childhood and increases with age.
Poor responses are seen in:
T-cell immune deficiencies (primary and secondary)
combined immune deficiency
some patients with CVID
leukaemias
lymphomas
other malignant disease
renal failure
during some chronic infections (late HIV).
T-cell function: in vitro assays
Principles of testing
In vitro T-cell function testing is carried out by inducing the T cells to proliferate by exposure to either mitogens or antigens.
Mononuclear cells are separated from neutrophils by density gradient centrifugation (Ficoll®).
Proliferation of the T cells is measured by the incorporation into DNA of tritiated thymidine in replicating cells.
Other methods used to study T-cell function in vitro include flow cytometric tests for the following.
Measurement of calcium flux.
DNA replication (a non-isotopic alternative to the standard proliferation assay).
Changes in surface antigen expression in response to activation (IL-2 receptor, CD25, transferrin receptor, CD71, CD69, and the nuclear antigen Ki-67).
Intracellular cytokines: cytokine production in culture can be measured, but this is not done routinely and the flow cytometric determination of intracellular cytokine is likely to be of more value.
There is little value in the MLR as a test of T-cell function, although it forms a part of cross-matching bone marrow.
ELISPOT assays can be used to measure cytokine production in response to antigens and will also give a precursor frequency.
Indications for testing
Suspected primary T-cell or combined immune deficiency.
Functional assays are rarely required in secondary T-cell immunodeficiency.
Monitoring of the T-cell proliferative response after HSCT provides a useful marker of returning function that will determine safe release from laminar flow.
Interpretation
Results will be reported as counts per minute (cpm) for the unstimulated and stimulated cells and as a stimulation index.
For PHA-stimulated cells the uptake should be >5000cpm and the increment over the unstimulated cells should be >4000cpm. The stimulation index should be >10.
For antigens such as Candida, the response is smaller and an increment of 2000cpm and a stimulation index of 3.0 are satisfactory.
The requesting clinician must also arrange a control sample from a healthy volunteer, where possible of the same age/sex as the patient.
This is necessary, as there are wide variations in individual responses, even in healthy individuals, and there are variations with age.
Each laboratory should establish its own age- and sex-specific normal ranges for each mitogen.
The most useful stimuli are the following.
Phytohaemagglutinin (PHA): a lectin (sugar-binding molecule) derived from kidney beans. This binds to sugar residues on a number of surface molecules, thus activating cells by several pathways simultaneously, including via the CD3–Tcr complex.
Concanavalin A (ConA): a lectin derived from jack beans. Its effect is similar to that of PHA except that it depends on normal monocyte accessory function.
Mitogenic anti-CD3 monoclonal antibodies: soluble and immobilized anti-CD3 cause specific stimulation of T cells via the CD3–Tcr complex, mimicking antigen.
Phorbol esters (phorbol myristate acetate, PMA): this molecule activates protein kinase C directly in cells, bypassing the need for membrane events. The addition of a calcium ionophore, which raises the intracellular calcium by inserting unregulated calcium channels in the membrane, increases the effect of PMA on PKC, as it is a calcium-dependent enzyme.
Interleukin-2: this has very little effect on its own but is synergistic with anti-CD3. Restoration of proliferative responses to other stimuli by the addition of IL-2 suggests a downstream defect leading to reduced/absent IL-2 production.
Antigens: many antigens can be used, but the most useful are Candida, tetanus, PPD, and viral antigens (CMV, HSV, rubella), as patients are likely to have been exposed or immunized. Responses are lower, as the frequency of T cells with the correct Tcr will be small.
Lymphoma diagnosis
Principles of testing
Diagnosis of lymphoma follows principles similar to those for leukaemia typing, except that the cells are in a solid organ.
Single-cell suspensions produced by disaggregating the tissue can be used.
Most information is gained from looking at tissue sections.
Staining is usually done with monoclonal antibodies followed by anti-mouse antibody conjugated to a reagent for developing a colour reaction (peroxidase, alkaline phosphatase–anti-alkaline phosphatase, etc.).
Most studies can now be carried out on paraffin sections.
Immunophenotyping will normally be undertaken in parallel with morphological, virological, and enzymatic studies.
The primary panel usually includes:
CD45 (leucocyte common antigen)
CD45RA (minority of T cells and B cells)
CD3 (T cells)
CD4 (T-helper cells plus macrophages and dendritic cells)
CD8
C3bR (follicular dendritic cells, B cells, macrophages)
HLA-DR (B cells, activated T cells)
surface immunoglobulins (heavy and light chains)
Ki-67 (nuclear antigen expressed in proliferating cells).
The secondary panel for T-cell antigens includes:
CD2, CD5, CD7, and CD1, although the latter is also expressed on dendritic cells and macrophages.
The secondary panel for B-lineage antigens includes CD10, CD21, CD22, CD23, CD24, CD79a, and CD5 (also expressed on T cells).
Confirmation of the presence of Reed–Sternberg cells can be obtained by using CD30 and CD15.
Histiocytes are reactive with CD68.
LMP-1 is a surface marker of EBV+ cells and is expressed on Reed–Sternberg cells.
In situ hybridization can be used to detect oncogene expression and viral genes.
Initial and supplementary antibody panels will be used.
Extracted material can be used as a source of DNA for molecular studies of T-cell receptor and Ig heavy-chain gene rearrangements as markers of clonality.
EQA systems exist.
Indications for testing
Any excised lymphoid tissue where lymphadenopathy is a feature should be examined for evidence of lymphoma.
Interpretation
In the differential diagnosis of an abnormal lymph node the question ‘Is this a malignant process or a reactive process?’ must be answered.
Lymphomas often express aberrant patterns of surface and cellular antigens:
κ:λ ratios > 10:1
sIg negative, B-lineage antigen positive
co-expression of B-lineage antigens and CD5, CD10, CD43, or CD6
loss of an expected T-lineage antigen
dual expression of CD4 and CD8 (outside thymus)
expression of terminal deoxytransferase (TdT) or CD1a (outside thymus).
Lymphoid tumours need to be distinguished from other (metastatic) malignancy:
cells of lymphoid origin usually express CD45
other markers are available to distinguish cells from other sources, including carcinoembryonic antigen, cytokeratin, chromogranin, desmin, and S-100.
Hodgkin’s disease is distinguished by the presence of characteristic Reed–Sternberg cells:
usually detectable by standard histology, although they may be sparse
can be identified by their reaction with CD30 and CD15, without reactivity with CD45 or T/B lineage antigens.
Evidence of clonality can now be obtained by studies of Ig and Tcr gene rearrangements by molecular techniques:
this can be carried out even on DNA extracted from paraffin sections
because of the use of PCR techniques to amplify the DNA of interest, very small samples can be analysed.
Abnormal expression of oncogenes and tumour suppressor genes can be detected by in situ hybridization. Oncogenes routinely screened for include:
Bcl-2
cyclin D1
p53
Bcl-6
CD99 (myc-2)
c-myc.
Viral screens include EBV, CMV, HHV6, HHV8.
The classification of lymphomas is constantly being revised in the light of new findings. Readers are advised to consult an up-to-date detailed text to understand the process.
Neutrophil function testing
Principles of testing
First test is neutrophil count (serial counts required for cyclic neutropenia—3 times weekly for 6 weeks).
Screening test for defects of oxidative metabolism (CGD) is nitroblue tetrazolium reduction test, in which a colourless intracellular dye is reduced to an insoluble blue compound, formazan, when the neutrophil’s oxidative machinery is activated.
Usually done as a simple slide test.
Can be done as a quantitative assay, with extraction of the formazan and quantitation by colorimetry.
Flow cytometry using dye reduction (dihydrorhodamine (DHR)) allows more cells to be analysed more quickly.
NBT and DHR tests should be done in parallel, as there are examples of neutrophil deficiencies where one test is abnormal but the other is not.
Other tests of the oxidative machinery include chemiluminescence (amplified by luminol) and the iodination test, which relates to hydrogen peroxide production.
Phagocytosis can be measured by simply counting the number of latex beads or yeasts ingested by neutrophils or, more accurately, by flow cytometry using labelled bacteria.
Bacterial killing assays allow the whole process to be tested, including opsonization, phagocytosis, and oxidative metabolism.
Test organism is incubated with patient’s serum or control serum and then each is incubated with either normal or patient’s neutrophils.
At a fixed time thereafter, the cells are lysed and the lysate plated out to allow residual live bacteria to grow.
Normally all bacteria will be killed within 30 minutes.
Chemotaxis assays are usually carried out by measuring migration under agarose or by the Boyden chamber method, in which cells migrate into a microporous filter which is examined under a microscope with a vernier gauge on the focusing ring, allowing the distance travelled to the leading edge to be measured.
Monocytes can be studied on flow cytometers at the same time as neutrophils.
Specific defects include absence of γ-IFN receptors, IL-12.
No EQA schemes exist. Therefore laboratories must establish their own normal ranges and set up normal controls in parallel with patient samples for all assays.
Indications for testing
Any patient with suspected neutrophil disorder:
recurrent abscesses, especially if deep-seated (liver)
extensive oral ulceration/gingivitis
atypical granulomatous disease
unusual bacterial or fungal infections—especially catalase-positive organisms (aspergillus, staphylococcus).
Interpretation
Slide NBT tests may miss some cases of chronic granulomatous disease and, if there is a high degree of suspicion, it is essential to perform a more sensitive flow cytometric assay.
With sensitive flow cytometric assays, heterozygotes for CGD mutations may have half the normal activity.
Bacterial killing assays may be abnormal in healthy children under the age of 2 years.
Chemotaxis is an important part of the process and rare defects due to the lack of anaphylotoxin receptors have been reported.
Both methods for chemotaxis give wide ranges even for normal individuals, so determining what is abnormal is often difficult.
Neutrophil function testing should always include testing for adhesion molecule deficiency (see ‘Adhesion markers’, p.559) and for neutrophil enzymes, especially myeloperoxidase (a common deficiency of doubtful significance), G6PD, and alkaline phosphatase (reduced in specific granule deficiency).
Neutrophil assays must be done with fresh samples.
Any intercurrent infection will cause abnormal function.
Follow-up protein studies and genetic investigations are required where defects of oxidative metabolism are identified in screening tests.
NK-cell function
Principles of testing
Activity of MHC non-restricted killer cells (natural killer cells) can be assessed in vitro.
Erythroleukaemia cell line K562 is known to be susceptible to lysis by NK cells.
Assay is carried out by incubating mononuclear cells with labelled K562 cells at varying effector target cell ratios and then identifying the death of the targets.
Conventional method is to surface label the targets with Cr51, and then measure the release of the isotope into the medium on cell death.
Appropriate controls are required to identify spontaneous release of the isotope and target cell death unrelated to effector cell activity (should be less than 5%).
Flow cytometric assays use a green fluorescent membrane-bound dye to label the targets.
Cell death is identified by the uptake of propidium iodide, which gives a red fluorescence.
Thus live and dead targets can be separated from the unlabelled effector cells by their staining.
The assays can be modified using different targets to look at antibody-dependent cell-mediated cytotoxicity (ADCC) and lymphokine-activated killer (LAK) activity.
No EQA exists. Laboratories must establish normal ranges and run normal controls with each assay.
The test depends on availability of high-quality K562 cells. Long-term cell culture must be meticulous.
Indications for testing
Indications are limited.
NK-cell deficiency has been reported (rarely) causing severe infections with herpesviruses.
Routine screening of patients with simple cold sores is not justified.
NK-cell function may be relevant in graft rejection and assessment of rare NK-cell leukaemias.
Interpretation
Flow cytometric assay is more sensitive to minor target-cell damage, permeabilizing the cell to the red dye. Therefore oversensitivity is a problem.
Chromium-release assay depends on the complete disintegration of the cell.
Clinical diagnostic value of the NK assay remains to be fully evaluated. Routine evaluation of ADCC and LAK activity is not undertaken.
Excessive NK-cell activity has been associated with an increased risk of graft loss in mismatched bone marrow transplants (particularly host NK-cell activity).
Low/absent NK activity has been reported in rare patients with recurrent infections with herpes family viruses.
Very high activity may be found in NK-cell leukaemias.
Number of NK cells identified by flow cytometry does not necessarily correlate with the activity.
NK granule release
Principles of testing
CD107a (lysosomal-associated membrane protein 1 (LAMP1)) is expressed on the surface of NK cells that have degranulated.
K562 cells are used as a target for NK cells to stimulate degranulation, as they do not express MHC Class I antigens.
Expression of CD107a on NK cells (CD3+ CD56+) is detected by flow cytometry after incubation of separated PBMNC with K562 cells.
PHA is also used as a non-specific activator of degranulation.
Indications for testing
Suspected familial haemophagocytic lymphohistiocytosis (FHLH).
Interpretation
Patients with FHLH due to Munc 13–4 and Syntaxin 11 deficiency will have absent degranulation.
Patients with perforin deficiency will have normal granule release.
A normal control is required with each run.
Very few NK cells should express CD107a unstimulated; >7% should express it when stimulated.
Assay depends on ready access to high-quality K562 cells.
Perforin expression
Principles of testing
Flow cytometric detection of intracellular perforin, using a permeabilization technique on separated PBMNC.
Perforin is expressed in the granules of NK cells, some CD8+ T cells, CD56+ T cells, and δ T cells.
Indications for testing
Suspected familial haemophagocytic lymphohistiocytosis (FLH) (see Chapter 1).
Interpretation
Only 30% of patients with FLH will have a perforin defect. Follow-up degranulation assays should be carried out if perforin expression is normal and there is a high degree of suspicion (see ‘NK granule release’, p.576).
XLPS should be excluded in males.
Toll-like receptors (TLRs)
Principles of testing
Flow cytometric assay based on the shedding of CD62L (L-selectin) by neutrophils when activated.
All 10 TLRs identified signal via MyD88 and IRAK-4 (clinical deficiencies of both are described (see Chapter 1)). Four TLRs signal via UNC-93B (TLR3, TLR7, TLR8, TLR9).
Activation of neutrophils via TLRs will lead to loss of CD62L.
Lipopolysaccharide (LPS) is the ligand for TLR4; CL097 (an imidazoquinoline) is a ligand for TLR7/8. PMA is used as a positive control (bypasses TLRs to activate neutrophils).
Indications for testing
Suspected deficiency of IRAK-4, MyD88, or UNC-93B.
Interpretation
IRAK-4- and MyD88-deficient patients will shed CD62L normally with PMA, but not with LPS or CL097.
UNC-93B-deficient patients will shed CD62L normally with PMA and LPS but not with CL097.
TRECs
Analysis of T-cell receptor excision circles (TRECs) is valuable in assessing thymic output, e.g. post-HSCT, in HIV patients on HAART (low levels may predict disease progression).
As TRECs do not replicate during cell division, progressive dilution occurs post-emigration.
Assays are not yet widely available for routine use.
TRECs can be detected on magnetic-bead-separated T-cell subpopulations by quantitative PCR techniques.
T-cell receptor and immunoglobulin heavy-chain gene rearrangements
Testing is carried out by multiplex PCR amplification followed by PAGE.
Panels of Tcr Vβ-specific labelled monoclonal antibodies can be used to carry out testing by flow cytometry.
This enables monoclonal expansions in the T-cell repertoire (lymphoma, response to chronic infection) and selective clonal loss (e.g. Omenn’s syndrome, DiGeorge syndrome) to be identified.
Spectra type of Tcr Vβ and IgH gene usage can be constructed to demonstrate polyclonal, oligoclonal, and monoclonal gene usage, using fluorescent PCR products.
This type of testing is crucial in lymphoma diagnosis, and in the diagnosis of oligoclonal states such as Omenn’s syndrome.
Month: | Total Views: |
---|---|
October 2022 | 1 |
April 2024 | 1 |
June 2024 | 2 |