Haematology 3 Flashcards

1
Q

What’s VTE?

A

Venous Thromboembolism

Made up of:
Deep Vein Thrombosis (DVT)
Pulmonary Embolism (PE)

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

Virchow’s triad and thrombosis they predispose to

A

venous (stasis and hypercoaguability)

arterial (vessel wall)

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

Risk factors for VTE

A
  • age
  • previous VTE
  • cancer
  • immobility/paresis
  • surgery/trauma
  • other illness (e.g. Inflammatory bowel
    disease; Behcet’s; Paroxysmal noctural
    haemoglobinuria; myeloproliferative diseases)
  • COCP/HRT
  • pregnancy/puerperium
  • inherited thrombophilia
  • antiphospholipid Abs
  • obesity
  • smoking
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

puerperium

A

the period of about six weeks after childbirth during which the mother’s reproductive organs return to their original non-pregnant condition

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

Definition of Hereditary thrombophilia

A

A genetically determined disorder of haemostasis which increases the probability of venous thromboembolism (VTE)

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

What are the 5 inherited thrombophilias

A
1965 Antithrombin      1 in 3,000
1981 Protein C              1 in 300
1984 Protein S              1 in 300
1993/4 Factor V Leiden 1 in 20
1996 PTG20210A         1 in 50
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Function of factor Va

A

Factor Va orientates X and PT so the cleavage is better.

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

What’s AT and what does it do?

A

Anti-thrombin is a serine protease inhibitor (serpin) and inhibits Xa and IIa (thrombin)

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

What’s PC and what does it do when activated?

A

Protein C, also known as autoprothrombin IIA and blood coagulation factor XIV, is a zymogen, the activated form of which plays an important role in regulating anticoagulation, inflammation, cell death, and maintaining the permeability of blood vessel walls in humans and other animals. Activated protein C (APC) performs these operations primarily by proteolytically inactivating proteins Factor Va and Factor VIIIa. APC is classified as a serine protease as it contains a residue of serine in its active site. The zymogenic form of protein C is a vitamin K-dependent glycoprotein that circulates in blood plasma.

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

What’s protein S?

A

A cofactor for protein C

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

Thrombin negative feed back

A

Thrombin bound to thrombomodulin activates protein C, an inhibitor of the coagulation cascade. The activation of protein C is greatly enhanced following the binding of thrombin to thrombomodulin, an integral membrane protein expressed by endothelial cells.

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

Inheritance of anticoagulation defects

A

Autosomal dominant
e.g AT, PC, PS
Only one allele leads to an increased risk of VTEs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What’s factor V Leiden?

A

R506Q (arginine to glutamine)
Cleavage by PC takes place 10x more slowly

1 in 20 so common!

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

What’s PTG20210A?

A

The “G20210A” refers to the fact that the mutation is a guanine (G) to adenine (A) substitution at position 20210 of the DNA of the prothrombin gene. This mutation (or more accurately, single-nucleotide polymorphism or variant), is commonly associated with increased risk of occurrence and recurrence of the disease venous thromboembolism (VTE), including both deep vein thrombosis (DVT) and pulmonary embolism (PE).

The polymorphism is located in a noncoding region of the prothrombin gene (3’ untranslated region nucleotide 20210), replacing guanine with adenine. The position is at or near where the pre-mRNA will have the poly-A tail attached.

Genetic code causes the body to make too much of the prothrombin protein. By having too much prothrombin, it increases the chances the blood clotting.

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

Antiphospholipid Syndrome Clinical Criteria

A

Arterial or Venous Thrombosis (young people with strokes)
or
Pregnancy Morbidity (recurrent miscarriage)
• ≥ 1 fetal death(s) after 10 weeks
• ≥ 1 premature birth(s) (≤ 34 weeks) due to preeclampsia, eclampsia or placental insufficiency
• ≥ 3 consecutive spontaneous abortions < 10 weeks

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

Antiphospholipid antibodies

A

Antiphospholipid antibodies are not directed against phospholipids but against proteins that bind to negatively charged phospholipids

Antibodies to ß2glycoprotein I (ß2GPI) are probably pathogenic, antibodies to prothrombin probably not

Detected as:
Lupus Anticoagulant
Anticardiolipin Antibodies
Antiß2GPI Antibodies (binds cardiolipin)

Affect intracellular signalling.
Paradoxically slow clotting in test tube.

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

Signs and symptoms of DVTs

A
Pain and tenderness
Swelling
Pitting oedema
Increased warmth
Collateral superficial veins
Change in skin colour
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Wells score

A

Pretest probability assessment for DVT

active cancer 1
paralysis, plaster 1
bed > 3d, surgery within 4w 1
tenderness along veins 1
entire leg swollen 1
calf swollen > 3cm 1
pitting oedema 1
collateral veins 1
previous DVT 1
alternative diagnosis -2
Low <1
Moderate 1-2
High >2
Unlikely ≤1
Likely ≥2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What’s the D-dimer test?

A

A monoclonal antibody against D-dimers.
During clotting fibrinogen (D-domain, E-domain, D-domain) crosslinks to form fibrin (D-domain, E-domain, D-domain D-domain, E-domain, D-domain, etc).

D-domains are cleaved together.
Suggests there has been a clot.

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

Use of pre-test probability when

considering D-dimer result

A

Sens 0.96 spec 0.44 LR neg = 0.091
Useful for negatives.

If unlikely DVT based on wells score and D dimer negative then 0.5% chance. Don’t need to treat.

If likely DVT based on wells score and D dimer negative then 3.4% chance. Must treat

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

What type of DVTs is US good at detecting

A

Proximal DVTs (popliteal and above)

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

When to treat a DVT? Draw the flow chart

A

First do Wells score:

Unlikely -> D-dimer:

  • if negative discharge
  • if positive -> proximal US:
    • if negative discharge
    • if positive treat

Likely -> Proximal US

  • if positive treat for DVT
  • if negative -> D-dimer:
    • if negative discharge
    • if positive -> repeat US 6-8 days later: if neg discharge, if positive treat for DVT
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Signs and symptoms of PE

A
Shortness of breath
Chest pain - pleuritic
Tachycardia
Haemoptysis
Circulatory collapse
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Features of the ideal anticoagulant

A
  • oral
  • a wide therapeutic index
  • predictable pharmacokinetics and dynamics negating the need for monitoring
  • a rapid onset of action
  • an antidote
  • minimal non-anticoagulant side-effects
  • minimal interactions with other drugs and food
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Heparins

A
  • Heparin is a mixture of glycosaminoglycan chains extracted from porcine mucosa.
  • Unfractionated heparin (UFH) chains average 15,000 Da.
  • Low molecular weight heparin (LMWH) is a mixture of smaller chains – average 5,000 Da.
  • All heparins depend on a specific pentasaccharide sequence for binding to antithrombin. They bind AT and potentiates its action.
  • 5 saccharides in a row where sulphides are in the right orientation are able to bind to AT
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is fondaparinux?

A

A chemically synthesised pentasaccharide that has the sulphates in the right position to bind antithrombin III and inhibit factor Xa.
Unlike heparin, it is selective for factor Xa.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

LMWH preferentially inhibits…

A

IIa (thrombin) more than Xa

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

UFH only inhibits

A

IIa

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

Unfractionated heparin

A

Given by continuous intravenous infusion.

Requires monitoring by APTT with adjustment of infusion rate to keep in the therapeutic range (often quoted a 1.5 – 2.5 normal – with our laboratory reagent 45 – 70 seconds)

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

LMWH

A

Low molecular weight heparin

The key property of LMWHs is that they
produce a much more predictable anticoagulant response than UFH.

They also have very high bioavailability after s/c injection.

The dose can therefore calculated by
body weight and be given s/c od without
any monitoring or dose adjustment.

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

What’s protamine sulphate and when is it used?

A

It is a highly cationic peptide that binds to either heparin or low molecular weight heparin (LMWH) to form a stable ion pair, which does not have anticoagulant activity. The ionic complex is then removed and broken down by the reticuloendothelial system. In large doses, protamine sulfate may also have an independent—however weak—anticoagulant effect.

Works better with longer chains.
UFH – yes
LMWH – partial
Fondaparinux - no

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

Vitamin K

A

Vitamin K is needed to perform an essential posttranslational modification of key clotting factors (II, VII, IX, X)

Vitamin K antagonists (VKA) can therefore be used as anticoagulants

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

Warfarin mechanism

A

Warfarin inhibits the vitamin K-dependent synthesis of biologically active forms of the calcium-dependent clotting factors II, VII, IX and X, as well as the regulatory factors protein C, protein S, and protein Z. Other proteins not involved in blood clotting, such as osteocalcin, or matrix Gla protein, may also be affected. The precursors of these factors require gamma carboxylation of their glutamic acid residues to allow the coagulation factors to bind to phospholipid surfaces inside blood vessels, on the vascular endothelium. The enzyme that carries out the carboxylation of glutamic acid is gamma-glutamyl carboxylase. The carboxylation reaction will proceed only if the carboxylase enzyme is able to convert a reduced form of vitamin K (vitamin K hydroquinone) to vitamin K epoxide at the same time. The vitamin K epoxide is in turn recycled back to vitamin K and vitamin K hydroquinone by another enzyme, the vitamin K epoxide reductase (VKOR). Warfarin inhibits epoxide reductase

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

PT/INR

A

The PT expressed as an INR measures the degree of anticoagulation

INR
1.0 normal, not anticoagulated
1.0 – 2.0 under-anticoagulated
2.0 – 3.0 correct degree of anticoagulation for most
3.0 – 4.0 extra anticoagulation for some eg MHV
>4.0 over-anticoagulated

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

Reversal of warfarin

A

Major bleeding:
give PCC (30 u/kg)
and vitamin K 5 mg iv

Non-major bleeding:
give vitamin K 1-3 mg iv

INR > 8.0 without bleeding:
give vitamin K 1-5 mg orally

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

What’s PCC?

A

Prothrombin complex concentrate

Made up of factor II, IX and X (sometimes VII)

Acts within minutes

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

Warfarin is not the ideal anticoagulant because…

A
  • very narrow therapeutic range
  • unpredictable
  • slow onset so cover with heparin
  • interacts with other drugs and food
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Direct oral anti coagulants that inhibit Xa

A

Rivaroxaban
Apixaban
Edoxaban

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

Direct oral anti coagulant that inhibits IIa

A

Dabigatran

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

Dabigatran

Target
Half life
Renal clearance
Bioavailability
Peak
A
Target  IIa
Half life  12-17 h
Renal clearance  80%
Bioavailability  6%
Peak  2 h
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Rivaroxaban

Target
Half life
Renal clearance
Bioavailability
Peak
A
Target  Xa
Half life  7-13 h
Renal clearance  33%
Bioavailability  80%
Peak  2-4 h
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Apixaban

Target
Half life
Renal clearance
Bioavailability
Peak
A
Target  Xa
Half life  10-14 h
Renal clearance  25%
Bioavailability  50%
Peak 2-4 h
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Edoxaban

Target
Half life
Renal clearance
Bioavailability
Peak
A
Target Xa
Half life  8-10 h
Renal clearance  50%
Bioavailability  62%
Peak  2-4 h
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

Antidote to dabigatran

A

Idarucizumab

  • Monoclonal mouse antibody developed with high dabigatran binding affinity
  • Monoclonal antibody was then humanized and directly expressed as a Fab fragment in mammalian cells (hamster)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Antidotes to Xa inhibitors that are in clinical trials

A

Andexanet

A truncated inactive recombinant FXa, expressed in CHO cells, that avidly binds Xa inhibitors.
• Lacks the GLA domain and replaces the activation peptide with a linker connecting light and heavy chains.
• Catalytically inactive because of a mutation (S419A) in the catalytic triad.

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

Stages of haemostatic plug formation

A

Vessel constriction

Formation of an unstable platelet plug

  • platelet adhesion
  • platelet aggregation

Stabilisation of the plug with fibrin
-blood coagulation

Dissolution of clot and vessel repair
-fibrinolysis

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

Primary haemostasis

A

Formation of an unstable plug

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

Secondary haemostasis

A

Stabilisation of the plug with fibrin

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

Why do people bleed?

A
• Almost always a defect of procoagulant effect
– Acquired or congenital
– Includes:
• adhesive functions of platelet-VWF
• stabilising effect of FXIII

• Acquired disorders are most common in hospital

• Very rarely excess anticoagulant effect
– except for anticoagulant drugs (warfarin, NOACs)

• Very rarely excess fibrinolytic effect
– except therapeutic

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

Types of defects of procoagulant function and how to test for them

A
• Too little protein (quantitative)
– Usually inherited (specific assays)
– Excess consumption (global pattern)
– Liver failure (global pattern)
– Drugs, antibody, hormonal (specific assays)

• Defective function (qualitative)
– Usually inherited (specific assays)

• Inhibited function
– Antibody present (specific assays)
– Drugs

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

Tests of haemostasis

A

• Screening tests
– Clotting screen (PT/APTT)
– Fibrinogen
– Full blood count (platelets)

  • Specific factor assays (eg Factor VIII)
  • Platelet function (PFA-100, platelet aggregation)
  • Global tests: ROTEM, TEG, thrombin generation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Which clotting screen is used for the extrinsic pathway?

A

Prothrombin time (PT)

VIIa, V, X, II

Extrinsic pathway activated by exposed tissue

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

Which clotting screen is used for the intrinsic pathway?

A

Activated partial thromboplastin time (APTT)

XII, XI, VIII, IX, V, X, II

contact activated

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

Why are coagulation screens done? What’s the problem?

A
  • Detect a bleeding tendency
  • Detect a cause for bleeding
  • Detect a systemic disease
  • Detect a thrombotic disorder
  • ‘Reassurance’

In fact they are reliable for none of these
things:
• Unphysiological
• Limited sensitivity & specificity
• Test a very limited portion of haemostasis
• i.e. practical but unreliable

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

Bleeding disorders not detected by routine clotting tests

A
  • Mild factor deficiencies
  • Platelet disorders
  • von Willebrand disease
  • Factor XIII deficiency (cross linking)
  • Excessive fibrinolysis
  • Vessel wall disorders
  • Metabolic disorders (e.g. uraemia)
  • (Thrombotic disorders)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

Should you do pre-operative coagulation tests?

A

No!

Patients should have a bleeding history taken and do a coagulation test if cause for concern.

– previous surgery and trauma
– family history
– anti-thrombotic medication

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

Categories of coagulation disorders

A

Reduced production of coagulation factors
– Hereditary failure of production:
• Factor VIII/IX, haemophilia A/B
– Acquired:
• Liver disease
• Dilution
• Anticoagulant drugs – warfarin, direct oral anticoagulants

Increased consumption
– Acquired:
• Disseminated intravascular coagulation (DIC)
• Immune - autoantibodies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What proportion of each factor is sufficient for haemostasis?

A

Approx 40%

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

Features of haemophilia A and B

A

• Haemophilia
– A – deficiency factor VIII
– B – deficiency factor IX

• Congenital, X-linked recessive
– Inversion intron 22 (50% haemophilia A), point mutations, deletions

• Incidence
– A - 1 in 10,000
– B – 1 in 50,000

• Factor VIII and Factor IX deficiency are clinically indistinguishable using
– coagulation screen results
– pattern of bleeding
– severity

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

Platelet function testing

A
  • Platelet number (FBC)
  • Platelet aggregation (measure aggregation in response to standard agonists)
  • Global tests (bleeding time, PFA)
  • Platelet granule contents (ATP:ADP ratio)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What’s TEG and what’s it used for?

A
  • Thromboelastogram
  • whole blood in cuvette, contact activation next to pin, slows rotation of the blood
  • developed for use with celite activator
  • measures changes in clot strength
  • includes all phases of haemostatic mechanism including fibrinolysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Inheritance of haemophilia

A
• For a female heterozygote (carrier)
– Half of sons are affected
– Half of daughters are carriers
• For a male haemophiliac
– All daughters are heterozygotes
– All sons are unaffected
• Approx one third are new mutations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Abnormal test in haemophilia

A

APTT Increased

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

Pattern of bleeding in haemophilia

A

• Not from superficial cuts or small vessels (eg
nosebleeds)
• Delayed
• Deep: muscle and joint

Problems with primary haematostasis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

Categories of bleeding in haemophilia

A

Severe (≤1%)
Joints and muscle
Bleeding often delayed, but prolonged
Risk of intracranial haemorrhage and chronic arthropathy (disability)

Moderate (2-5%)
Prolonged or excessive bleeding after minor trauma
Rarely bleeds into joints

Mild (6-40%)
‘Spontaneous’ bleeds
Excessive bleeding only after major trauma or surgery

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

History of haemophilia treatment

A
1950’s whole blood, plasma
1960’s cryoprecipitate
1970’s factor concentrates
1984 factor VIII cloned
1990’s recombinant FVIII and FIX
2000 protein free production
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

Half lives of factor replacements

A

Factor VIII 8-12 hours

Factor IX 18-24 hours

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

What’s ‘on demand’ factor replacement therapy?

A

Treat bleeds as they occur

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

What’s ‘prophylactic’ factor replacement therapy?

A

Treat before bleeds occur

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

Which haemophiliac patients are treated prophylactically?

A

Severe or moderates that behave as severe.

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

What are the non-concentrate treatments (not factor) for haemophiliacs?

A
  • Local measures
  • Tranexamic acid:
  • Binds to plasminogen
  • Blocks binding to fibrin
  • Clot lysis is reduced

•Desmopressin (DDAVP)

  • Vasopressin derivative
  • Acts via V2 receptors
  • 2-5 fold rise in VWF-VIII (VIII>VWF); DDAVP induces vWF secretion by endothelial cells by binding to V2R and activating cAMP-mediated signaling in endothelial cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

Complications of haemophilia

A
  • Acute bleeds
  • Long term arthropathy
  • Development of inhibitors (antibodies) to FVIII or IX
  • Transmission of hep B, hep C, HIV
  • vCJD
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What’s Von Willebrand’s disease?

A
• Quantitative or qualitative deficiency of von
Willebrand factor (VWF)
• Mostly autosomal dominant
• Chromosome 22
•  The four hereditary types of vWD described are type 1, type 2, type 3, and pseudo- or platelet-type. 
– Quantitative
• Type 1
– partial deficiency
– most common (75%), prevalence 10^-3-10^-4
• Type 3
– complete deficiency
– 5%, prevalence 10^-6

– Qualitative
• Type 2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What are the VWF functions and the related features of VWD?

A

– Bridge between collagen on damaged vessel wall and
platelets under shear (1° haemostasis)
• → platelet-type mucosal bleeding

– Stabilise and protect factor VIII (2° haemostasis)
• → coagulation defect bleeding

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

Type 2 VWD

A

Type 2 vWD (20-30% of cases) is a qualitative defect and the bleeding tendency can vary between individuals. Four subtypes exist: 2A, 2B, 2M, and 2N.

Type 2A
The vWF is quantitatively normal but qualitatively defective. The ability of the defective von Willebrand factors to coalesce and form large vWF multimers is impaired, resulting in decreased quantity of large vWF multimers and low RCoF activity. Only small multimer units are detected in the circulation. Von Willebrand factor antigen (vWF:Ag) assay is low or normal.

Type 2B
This is a “gain of function” defect. The ability of the qualitatively defective vWF to bind to glycoprotein Ib (GPIb) receptor on the platelet membrane is abnormally enhanced, leading to its spontaneous binding to platelets and subsequent rapid clearance of the bound platelets and of the large vWF multimers. Thrombocytopenia may occur. Large vWF multimers are reduced or absent from the circulation.

Type 2M
Type 2M vWD is a qualitative defect of vWF characterized by its decreased ability to bind to GPIb receptor on the platelet membrane and normal capability at multimerization. The vWF antigen levels are normal. The ristocetin cofactor activity is decreased and high molecular weight large vWF multimers are present in the circulation.

Type 2N (Normandy)
This is a deficiency of the binding of vWF to coagulation factor VIII. The vWF antigen test is normal, indicating normal quantity of vWF. The ristocetin cofactor assay is normal. Assay for coagulation factor VIII revealed marked quantitative decrease equivalent to levels seen in hemophilia A. This has led to some vWD type 2N patients being misdiagnosed as having hemophilia A.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

VWD treatment

A
• Generally “on demand”
• Local – OCP (oral contraceptive pill), nasal cauterisation
• Tranexamic acid
• DDAVP – nasal, s/c, IV
• Concentrates
– plasma-derived
– recombinant VWF now in clinical trial
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What causes HDN?

A
  • First described in 1894 by Townsend
  • Identified as due to vitamin K deficiency in 1930s
  • Vitamin K dependent coagulation factors: II, VII, IX, X
  • Treatment and prophylaxis of neonates first described in 1939
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

Causes of low vitamin K in the neonate

A
  • Poor placental transfer of vitamin K
  • Low fetal vitamin K stores
  • Low vitamin K content of breast milk (1-2 mg/L) compared to formula
  • Absent bacterial vitamin K synthesis in neonatal gut
  • Functional immaturity of fetal liver
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

Time course, associated causes and features of classic HDN

A

Time: Day 1-7

Associated causes: “Physiological”

Features: Bruising and purpura, post circumcision, GI haemorrhage

80
Q

Time course and associated causes of early HDN

A

Time: < 24 hours

Associated causes: Maternal meds: antiepileptic, anti-TB

81
Q

Time course, associated causes and features of late HDN

A

Time: 2-12 weeks

Associated causes: Malabsorption of vitamin K: underlying cholestatic disease, cystic fibrosis, diarrhoea

Features: Intracranial haemorrhage (up to 50%)

82
Q

Diagnosis of HDN

A
  • prolonged PT and APTT
  • Factor assay II, VII, IX, X if in doubt
  • Correction of coag abnormality in approx 24hrs with parenteral vit K confirms diagnosis
83
Q

Management HDN

A

– Treatment
• General support
• Vitamin K 1mg IV
• FFP (fresh frozen plasma) if severe bleeding

– Prevention
• 1mg IM at birth
• Oral vitamin K (2-3mg) different regimens

84
Q

Prevalence of HDN

A
  • Without prophylaxis 1 in 400 neonates
  • With IM prophylaxis 1 in 400,000 neonates

• NB Previous controversy
– Possible increased risk (overall 1.0-1.5-fold increase) of childhood leukaemias after IM vitamin K.

85
Q

Size of a platelet

A

 2-4 μm diameter

 Discoid & anucleate

86
Q

Lifespan of a platelet

A

7-10 days

87
Q

Platelet cell of origin

A

megakaryocytes

88
Q

Platelet number controlled by

A

TPO (Thrombopoietin)

89
Q

Normal platelet count

A

150-450 x10^9/l

90
Q

Platelet functions

A

 To help achieve a stable clot after injury to minimise blood loss

 Acts with endothelium (binds to vWF) & provides a PL surface for coagulation factors

91
Q

Platelet response to injury

A

Adhesion
surface glycoproteins, vWF – slows the plt down

Activation
ADP, thrombin, thromboxane

Releasereactions
platelet granule contents

Aggregation
ADP, thromboxane A2

92
Q

Diseases of platelets

A

 Abnormality of number:

  • Thrombocytopenia
  • Thrombocytosis

 Abnormality of platelet function

 Combination of the two

93
Q

Too many platelets

A

Thrombocytosis

94
Q

Not enough platelets

A

Thrombocytopenia

95
Q

Platelet count of mild thrombocytopenia

A

100-150 x 10^9/l

96
Q

Platelet count where you’re at risk of surgical bleeding

A

<50 x 10^9/l

97
Q

Platelet count at risk of spontaneous bleeding

A

10-20 x 10^9/l

98
Q

Categories of things causing thrombocytopenia

A

 Artefactual- react to EDTA in FBC tube, clump together and are counted as RBCs. You can see it on a blood film. Use a citrated tube (no EDTA)
 Increased destruction
 Reduced production

99
Q

Causes of increased platelet destruction

A

 Autoimmune: ITP, CTD
 Drug related: heparin (HIT), quinine, penicillins
 Microangiopathic: TTP, HUS, DIC
 Hypersplenism: Portal hypertension, haematological malignancy
 Alloimmune: NAIT, PTP

100
Q

What’s ITP?

A

Idiopathic thrombocytopenic purpura

101
Q

What’s NAIT?

A

Neonatal alloimmune thrombocytopenia

102
Q

What’s PTP?

A

Post-transfusion purpura

103
Q

What’s TTP?

A

Thrombotic thrombocytopenic purpura

104
Q

Causes of reduced production of platelets

A
 B12/folate deficiency
 Liver disease/alcohol
 Infection: (HIV, hep C, CMV, severe sepsis)
 Bone marrow infiltration:
 malignancy, haematological disease
 Drugs: (cytotoxics)
 Aplasia
105
Q

Symptoms of platelet disease

A

 Mucosal surface bleeding;

  • Bruising, petechiae, ecchymoses
  • Epistaxis, menorrhagia
  • GI bleeding, gum bleeding

 Immediate and continued bleeding after injury
 Mucosal surfaces affected most commonly – Menorrhagia, gum bleeding, epistaxis
 Anaemia
 Joint and muscle bleeding uncommon

106
Q

Approach to diagnosing thrombocytopenia

A

Symptoms of platelet deficiency

 Detailed history:
 Onset of symptoms
 Family history (inherited disease)
 Bleeding history
 Bleeding assessment tools – Vincenza, PBAC, ISTH
 Past medical history
 Drug history: especially over the counter stuff
 Alcohol history
107
Q

Differential diagnosis of thrombocytopenia

A

 Von Willebrand’s disease
 Clotting pathway defects
 Vasculitis: palpable purpura (raised, tend to be on limbs)
 CTD- Ehlers-Danlos (collagen deficiency, blood vessels normally constrict when damaged but in ED they can’t)

108
Q

Initial approach to investigating thrombocytopenia

A
  • Historical full blood count (FBC)
    Trends in platelet counts provide clues to underlying diagnosis
  • FBC and reticulocyte count
    A high reticulocyte count suggests haemolysis.
    A low reticulocyte count points towards reduced red cell production.
    Cytopenias involving other cell lineages are suggestive of disorders involving the bone marrow.
  • Blood film Essential
  • Prothrombin time (PT), activated partial thromboplastin time (APTT), Clauss fibrinogen +/- d-dimers
  • Exclude disseminated intravascular coagulation or an additional coagulation abnormality
  • Renal and liver function
109
Q

What causes changes in blood film?

A

 Pseudothrombocytopenia (platelet clumps)
 B12 changes (hypersegmented neutrophils)
 Leucoerythroblastic (red and white cell precursors)
 HUS/TTP
 ITP
 Haem malign

110
Q

Specific diseases: ITP

A

 Auto-immune disease

  • Anti-platelet antibodies – IgG against platelet glycoprotein leading to destruction in the spleen
  • Recently – more complex T cell related mechanisms
  • Defined as plt count <100
  • M = F (except 30 – 60 yrs: F>M)

 Aetiology = unknown

 Occasionally 2° to:

  • SLE or CTD
  • lymphoproliferative disease
111
Q

ITP diagnosis

A
 Diagnosis: by exclusion
 No single test
 Blood film:  Giant platelets
 Bone marrow may be
helpful (++ megas)
112
Q

Specific tests for ITP

A

Reticulocyte count & Direct antiglobulin test (DAT):
Determines presence of haemolysis
Important if considering anti-D therapy

Immunoglobulin quantification Looking for: common variable immunodeficiency and IgA
deficiency

Blood group and Rhesus status
Informative if considering anti-D therapy

H. pylori serology
Urea breath test
Eradication of H. pylori has been shown to lead to
resolution of thrombocytopenia in some instances

Anti-phospholipid antibodies & lupus anticoagulant
Positive in 40% of individuals with ITP

Pregnancy test in women of
childbearing age
Management may differ in pregnancy

Anti-nuclear antibodies
Can help diagnose SLE and is also an indicator of chronicity
in childhood ITP

Viral PCR for CMV and parvovirus
Chronic infection can cause thrombocytopenia

113
Q

ITP management

A

Haemorrhagic:
 Steroids
 i.v. Ig
 Platelet transfusion should only be considered in life- threatening bleeding

Non-haemorrhagic:
 Children: observe (often recover within 6 weeks)
 Adults: assess risk for bleeding
1st line: steroids (prednisolone 1mg/kg p.o.)
2nd line: i.v. Ig especially for improving counts for surgery, anti-D, splenectomy, rituximab, TPO-analogues (romiplostim)

114
Q

First line therapies for ITP

A

Corticosteroids
IV anti-D
IVIg

115
Q

Second line therapies for ITP

A

Rituximab
TPO receptor agonists (ramiplostin, eltrombopag)
Splenectomy

116
Q

Specific diseases: microangiopathy and thrombocytopenia

A

 Disseminated Intravascular Coagulopathy (DIC)
 Haemolytic Uraemic Syndrome (HUS)
 Thrombotic Thrombocytopenia Purpura (TTP)

Lead to:
Thrombosis in microcirculation

Resulting in:

  • Organ failure
  • Fragmentation haemolysis
117
Q

What’s MAHA?

A

Microangiopathic haemolytic anaemia

 Deposition of fibrin mesh, in small vessels

 Due to:

  • Platelet rich thrombi
  • Red cells then get damaged by the fibrin mesh - schistocytes
118
Q

Features of TTP

A

 Rare disease
 Inherited or acquired (Acquired causes: pregnancy, HIV)
 Pentad of symptoms: fever, neurological signs, renal
failure, haemolysis, low platelets

 APTT/PT normal
 LDH raised (due to RBC lysis), platelets low, Hb low (~80), Coombs negative (DAT -ve), film MAHA, raised unconjugated bilirubin

119
Q

Mechanism underlying TTP

A

 In health, vWF is cleaved by ADAMTS13 protease
When none present:
 ++ activation of platelets
 Thrombi formed
 Diagnosis: low ADAMTS13 level - <5-10% (± ADAMTS Ab present)

120
Q

Investigations in TTP

A

To establish MAHA:
Full blood count, blood film, reticulocytes, LDH, bilirubin, coagulation screen (PT, APTT, fibrinogen), Coombs test

To look for organ involvement:
Renal function, troponin, ECG and consider echocardiogram (EF 15-20%) and CT/MRI

To look for underlying cause:
Pregnancy test, HIV, hepatitis A/B/C,
autoantibody screen including ANA, dsDNA,
anticardiolipin antibody, beta2glycoprotein-1, lupus
anticoagulant screen, B12, folate, thyroid function, amylase, stool culture, CT chest/abdomen/pelvis

To confirm diagnosis:
ADAMTS13 activity assay, anti-ADAMTS13 antibody assay

121
Q

TTP treatment

A

 TTP is a medical emergency
 Up to 20% mortality

 Treatment:

  • plasma exchange (usually daily – 1 – 1.5 plasma volumes)
  • Rituximab and steroids may be used
  • Aspirin once plt count > 50
  • Avoid platelet transfusions (bleeding is rare)

 Congenital TTP: born with lack of ADAMTS13
- Rx: regular infusions of intermediate purity Factor VIII or FFP

122
Q

Haemolytic uraemic syndrome

A

Same as TTP but more severe renal failure, less severe neurological symptoms.

 More common in children
 Strong association with E Coli 0157 (less common Shigella)
 Diarrhoea

 Low platelets, anaemia, microangiopathic blood film and renal failure
 APTT/PT normal
 Haemodialysis may be required – supportive measures

123
Q

DIC

A

 Acquired disorder
 Patients are often really sick (acute DIC)
 Associations: sepsis, trauma, obstetric emergencies, malignancy
 Chronic DIC is more commonly found with malignancy
 Bleeding: Venepuncture sites, etc
 Thrombosis: Digital gangrene

124
Q

Features of DIC

A
May have bleeding or thrombotic features – depends on balance of coagulation
 Prolonged APTT/PT
 Low fibrinogen
 Low platelets
 Elevated D-Dimer: fibrinolysis
 Low Hb
 Film: MAHA
125
Q

Management of DIC

A
 Treat the underlying cause
 If bleeding:
- Give RBC as required
- FFP 15ml/kg aiming for APTT/PT <1.5 normal
- Platelets to aim >50x10^9/L
- Cryoprecipitate aiming for Fg >1.0g/L

(FFP: fresh frozen plasma – contains clotting factors,
cryoprecipitate – contains fibrinogen)

126
Q

HIT

A

 Clinico-pathological syndrome

 Clinical:

  • Thrombocytopenia
  • Thrombosis

 Pathology:
Antibodies vs. PF4-heparin complex which activate platelets and lead to consumption

127
Q

Presentation of HIT

A

 Platelets fall 5-10 days following heparin use
 Occasionally fall in 1st day if recent previous exposure
 Count falls by at least 30% from normal, and rarely below 20x10^9/L
 Patient may develop a venous/arterial clot
 Patient may complain of pain at the site of – heparin infusion, or LMWH/UFH injection sites (bright red painful areas around injection site

128
Q

Diagnosing HIT

A

4 Ts

 Thrombocytopenia (> 50% decrease)
 Timing 5-10 days after starting heparin
 Thrombosis
 OTher cause not apparent
 Test for HIT antibodies positive
(Not all have to be present but the more features there are, the more likely the diagnosis is HIT)

129
Q

HIT treatment

A

 Stop heparin!
 Start alternative anti-coagulant e.g. danaparoid, fondaparinux, lepirudin, argatroban
 Start definitive anticoagulant once platelets into normal range
 Warfarin or other suitable anticoagulant:
- 3 months if a thrombosis
- 4 weeks if HIT only

130
Q

Disorders affecting number of platelets

A
Thrombocytosis:
 Bleeding
 IDA
 Infection/inflammation
 Splenectomy
 Malignancy
 MPD: ET, PRV, CML, MF
131
Q

Disorders affecting quality of platelets

A
 Drugs – aspirin, anti-platelets
 Uraemia
 Liver disease
 Haematological (Myeloma, MPD, MDS, leukaemia)
 Cardiopulmonary bypass
132
Q

Inherited platelet disorders

A

 Vessel wall abnormality (Collagen vascular dx, vWD)
 Adhesion (Bernard Soulier GP1b)
 Aggregation (Glanzmann’s GPIIb/IIa)
 Secretion (Hermansky Pudlak, Grey platelet syndrome)
 Procoagulant activity (Scott’s syndrome)
 Signal transduction (Thromboxane receptor abnormality)

133
Q

Diferential diagnosis for lymphadenopathy

A
  1. Infection
    • Viral: EBV, CMV, HIV, rubella (posterior auricular)
    • Bacterial: endocarditis, TB, lyme disease, cat scratch
    • Parasitic: toxoplasmosis
  2. Malignancy: local met, CLL, any lymphoma, AML
  3. Drugs: phenytoin (many others reported)
  4. Dermatopathic lymphadenopathy
  5. Autoimmune: SLE, rheumatoid
  6. Other: Kikuchi’s (necrotising lymphadenitis)
134
Q

When would you biopsy a lymph node?

A

In an adult, lymph node present for > 6 weeks

135
Q

How would you investigate a para-aortic lymph node?

A

CT guided biopsy

136
Q

A patient presents with fatigue and breathlessness on exertion over the last few months.

Blood film shows pancytopenia.

Bone marrow aspirate: hypocellular particles, no marked dysplasia.

Trephine: markedly hypocellular with increased fat spaces.

Diagnosis?

A

Acquired aplastic anaemia

137
Q

Definition of pancytopenia

A

Definition: A reduction in all the cellular elements of the blood

138
Q

Causes of pancytopenia

A

Majority:
Reduced production
• Infection (including HIV) / sepsis
• Megaloblastic anaemia (B12 / folate deficiency)
• Drugs: chemo, RT, antiretrovirals etc
• Primary marrow failure: AA, myelodysplasia, Fanconi
• Marrow infiltration: secondary cancer, acute leukaemia, lymphoma, myelofibrosis
• Malnutrition / eating disorder (anaorexia)
• Paroxysmal nocturnal haemoglobinuria

Minority:
Peripheral sequestration - splenomegaly

139
Q

Complication of prolongued B12 deficiency and what does it affect?

A

Subacute combined degeneration of spinal cord

Also known as Lichtheim’s disease, refers to degeneration of the posterior and lateral columns of the spinal cord as a result of vitamin B12 deficiency (most common), vitamin E deficiency, and copper deficiency. It is usually associated with pernicious anemia.

140
Q

What is paroxysmal nocturnal haemoglobinuria?

A

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired, life-threatening disease of the blood. The disease is characterized by destruction of red blood cells (hemolytic anemia), blood clots (thrombosis), and impaired bone marrow function (not making enough of the three blood components).

Characterized by destruction of red blood cells by the complement system, a part of the body’s innate immune system. This destructive process occurs due to the presence of defective surface proteins on the red blood cell, which normally function to inhibit such immune reactions. Since the complement cascade attacks the red blood cells within the blood vessels of the circulatory system, the red blood cell destruction (hemolysis) is considered an intravascular hemolytic anemia.

141
Q

What is the telltale sign of PNH and what proportion of people have it?

A

Red urine in the morning (originally gave the condition its name)

26%

142
Q

A child with pancytopenia think….

A

bone marrow failure syndrome or acute leukaemia

143
Q

Someone presents with pancytopenia. What tests should you do to help with the diagnosis?

A

Examine for splenomegaly

FBC: high MCV - rule out megaloblastic anaemia

Reticulocytes - consumption or production problem

Blood film:
• leucoerythroblastic, think sepsis, marrow infiltration or fibrosis (useful to look at CRP)
• dysplastic changes, think myelodysplasia
• circulating blasts - acute leukaemia

144
Q

Hypogranular neutrophils…think…

A

Myelodysplasia

145
Q

Causes of bone marrow failure

A

Congenital:
• Fanconi anaemia - defect in DNA repair, become cytopenic 5-10yoa, increased risk AML, most have some other anomaly
• Schwachman-Diamond - neutropenia, exocrine pancreas insufficiency, skeletal abnormalities
• Dyskeratosis congenita - premature aging, skin / nail dystrophy, marrow failure. Gene encodes protein involved in telomerase function

Acquired:
Aplastic anaemia - rare autoimmune condition. Treatment with immunosuppresives (ATG, cyclosporine) or transplant

146
Q

Normal range of neutrophils

A

2-7.5 x 10^9 /l

147
Q

Normal range of eosinophils

A

0.04-0.4 x 10^9 /l

148
Q

Normal range of basophils

A

<0.1 x 10^9/l

149
Q

Normal range of monocytes

A

0.2-0.8 x 10^9/l

150
Q

Normal range of lymphocytes

A

1.5-4 x 10^9/l

151
Q

Who has more lymphocytes? children or adults

A

Children

152
Q

Effect of exercise on white cell count

A

Increases

153
Q

Effect of smoking on white cell count

A

Mild leucocytosis (neutrophils)

154
Q

Effect of pregnancy on white cell count

A

Mild leucocytosis (neutrophils)

155
Q

Causes of neutrophilia

A
  1. ‘Normal’ variations: pregnancy, smoking
  2. Infection
  3. Inflammatory disorder e.g. post-op, MI, vasculitis
  4. Underlying malignancy
  5. Drugs: corticosteroids, adrenaline, G-CSF
  6. Metabolic: DKA, renal failure, pre-eclampsia
  7. Acute haemorrhage
  8. Haem: myeloproliferative conditions (PV, ET, CML)
156
Q

Causes of eosinophilia

A
  1. Allergy / atopy: eczema, asthma
  2. Drugs: idiosyncratic drug reactions often associated with eosinophilia
  3. Parasitic infection especially worms
  4. Skin diseases: pemphigus, dermatitis herpetiformis
  5. Autoimmune conditions e.g. Churg-Strauss
  6. Lymphoma e.g. Hodgkin’s Lymphoma
  7. Eosinophilic lung diseases e.g. Eosinophilic pneumonia
  8. Addison’s disease
  9. Primary hypereosinophilic syndromes
157
Q

Causes of monocytosis

A

Isolated monocytosis is NOT common.

Main causes are:
1. Chronic infection: endocarditis, TB, CMV, malaria
2. Haematological disorders e.g. chronic myelomonocytic leukaemia (a form of myelodysplastic syndrome / myeloproliferative disorder)
Other conditions can cause monocytosis but not commonly

158
Q

Causes of basophilia

A

Basophilia is even less common than eosinophilia. Really only 1 cause:
Myeloproliferative condition
(e.g. CML, PV, ET, MF)

159
Q

Causes of a raised MCV

A
  • Folate and B12 deficiency
  • Alcohol / liver disease
  • Pregnancy
  • Drugs: azathioprine, hydroxycarbamide, methotrexate
  • Hypothyroidism
  • Myelodysplastic syndrome
  • Haemolysis (reticulocytes)
  • Myeloma
160
Q

What is oral hairy leukoplakia?

A

EBV driven condition in the immunosuppressed.

Test HIV status

161
Q

Causes of pure red cell aplasia

A

• Congenital: Diamond-Blackfan anaemia
• Parvovirus infection
 May be chronic if immunosuppressed
 Causes aplastic crisis in patients with chronic haemolysis e.g. sickle cell
• Autoimmune conditions e.g. SLE
• Lymphoproliferative conditions e.g. CLL
• Thymoma
• Treatment with erythropoietin (extremely rare)

162
Q

What is Diamond-Blackfan anaemia?

A

A congenital erythroid aplasia that usually presents in infancy. DBA causes low red blood cell counts (anemia), without substantially affecting the other blood components (the platelets and the white blood cells), which are usually normal.

About 25-50% of the causes of DBA have been tied to abnormal ribosomal protein genes.

163
Q

Presence of spherocytes suggests….

A

extravascular haemolysis

164
Q

What test should you do if you suspect extravascular haemolysis?

A
  • A direct antiglobulin test (aka direct Coombs’ test)
  • Reticulocyte count (raised)
  • Investigate the underlying cause as 50% have other diseases (HIV, CMV, malignancy, SLE)
165
Q

Treatment for autoimmune haemolytic anaemia

A
  • oral steroids (prednisolone)
  • folic acid supplementation ( so the increased rate of red blood cell production and destruction does not result in folate deficiency)(Remember that the body’s stores of folate are small compared to stores of B12.)
  • if bad splenectomy (take out sight of breakdown)
166
Q

What causes spherocytosis?

A
  • Hereditary spherocytosis
  • allo-immune haemolysis (this typically being post transfusion)
  • severe burns
  • clostridial infections
167
Q

Low hb with microcytic, hypochromic red cells, pencil cells suggests?

A
Iron deficiency
(might see a raised platelet count)
168
Q

The left supra-clavicular lymph node is known as?

A

Virchow’s node

169
Q

How do you confirm iron deficiency?

A
  • serum ferritin (low)
  • transferrin (high)
  • serum iron (low)
    NB but remember that infection and the acute phase response can sometimes make these data difficult to interpret.
170
Q

How does sickle cell cause vaso-occulsive crisis?

A

Briefly: the mutation in the beta globin gene means that the haemoglobin in sickle cell disease (HbS) is much less soluble than normal haemoglobin (HbA) when deoxygenated. Therefore, under hypoxic
conditions, HbS comes out of solution and polymerizes, producing long tactoids that resulting in sickling of the red cells.

Sickled cells are more rigid and less deformable than normal erythrocytes, and therefore lodge in the microvasculature with subsequent hypoperfusion and infarction. Although sickled cells can revert to the normal red cell shape on reoxygenation, repeated cycles of sickling and de-sickling will eventually irreversibly damage the cells which therefore have a shorter lifespan than normal red cells.

Increasing evidence suggests that endothelial activation and increase interaction between sickle red cells and the endothelial cells is also implicated.

171
Q

Why would a sickle cell person take penicillin and folic acid?

A

The spleen in patients with sickle cell disease is subject to episodes of sickling and auto-infarction.

Consequently, by adulthood, patients with sickle cell anaemia are functionally hyposplenic. Since the spleen is centrally important in protection against encapsulated organisms they take prophylactic antibiotics.

Folic acid taken as they have an increased rate of RBC synthesis

172
Q

What crises can complicate sickle cell disease?

A
  • acute chest syndrome
  • Painful crises
  • aplastic crises (e.g. caused by parvovirus B19, which prevents normal red cell
    maturation)
  • hepatic or splenic sequestration crises
173
Q

How do you treat someone in sickle cell crisis?

A
  • supportive care (fluids, oxygen, analgesia and antibiotic treatment)
  • red cell exchange transfusion (removing sickling blood, and replacing with non-sickling blood, to lower the HbS percentage).
174
Q

At what age will someone present with beta thalassaemia major?

A

Around 1 year once the switch from fetal to adult haemoglobin (HbF to HbA) has occurred.

175
Q

What’s the type of thalassaemia?

Requires frequent blood transfusions

A

Major

176
Q

What’s the type of thalassaemia?

anaemic but require only occasional transfusions (e.g. in the context of intercurrent infection)

A

Intermedia

177
Q

What molecular defects can cause alpha and beta thalassaemia?

A

Usually:

  • point mutations or small deletions for beta thalassaemia
  • gene deletions in alpha thalassaemia.
178
Q

Why would a thalassaemia patient take deferasirox?

What problems are they likely to develop without it?

A

This is an iron chelator. Patients with thalassaemia who received regular transfusions will become iron-loaded very quickly, since there is no physiological mechanism for enhancing iron excretion from the body. The iron is deposited in the heart, liver, and endocrine organs where is can cause significant dysfunction. Without good iron chelation, patients with thalassaemia major may die of the consequences of iron overload, with heart failure being the usual cause. Significant endocrine problems include growth retardation, delayed puberty and infertility

179
Q

What’s g the threshold at which myelodysplasia is said to have progressed to AML?

A

When the blast count in the marrow is over 20%

180
Q

Treatment for neutropenic sepsis

A

Medical emergency!

Neutrophils are critical for the maintenance of mucosal integrity and are also essential for the normal response to bacterial infection. Neutropenic patients are therefore at much increased risk of serious infection, especially with Gram negative organisms. Theu can deteriorate and succumb to infection exceedingly quickly. Any neutropenic patient with a fever should be treated aggressively with fluids as required, plus broad spectrum
antibiotic cover (remember to take cultures first!). Tazocin is a good first line choice.
  • Some need transfusion with red cells.
  • Platelet tranfusions required if patient develops haemorrhagic features.
181
Q

What’s the sudan black stain used for?

A

Sudan black will stain myeloblasts but not lymphoblasts.

182
Q

Causes of high HCT (haematocrit)

A

A high haemoglobin may therefore be consequence of either a high epo level (which may be appropriate in the face of hypoxia, or inappropriate if there is no hypoxia), or autonomous proliferation of the red cells in the marrow, independent of epo.

Primary polycythaemia (AKA polycythaemia vera) is the latter – autonomous red cell production. It is one of the myeloproliferative disorders.

Secondary polycythaemia is the kind driven by too much epo. If the patient is chronically hypoxic (maybe he has COPD? Maybe he has cyanotic heart disease?) he will naturally produce more epo; this will result in an ‘appropriate’ or compensatory high Hb. In some cases, there is inappropriate epo secretion; this typically occurs from a renal tumour or polycystic kidney.

A third subtype is ‘relative polycythaemia’. In this condition, the HCT is high NOT because of increased red cell production, but because of reduced plasma volume. This might be due to diuretic treatment, for
example.

If the patient also has a high platelet count…. This elevation of another myeloid cell line hints that this might be a primary myeloproliferative disorder; though the platelet count can also rise in the context of reactive inflammatory or malignant disease

183
Q

Common mutation for PV patients and what proportion have it?

A

JAK2 V617F

95%

184
Q

Complications of PV

A

 Thrombosis
 Haemorrhage
 Fibrotic change in the marrow (ie.e. the development of secondary myelofibrosis)
 (Rarely) transformation to AML

185
Q

Treatment of PV patients

A

venesection

JAK2 inhibitors (e.g. ruxolitinib) are also now available for patients whose disease is difficult to control by venesection

anticoagulation

186
Q

Mutations associated with essential thrombocythaemia

A
  • JAK2 mutation
  • MPL (the thrombopoietin receptor)
  • CALR (calreticulin) gene
187
Q

Tests to do if someone has thrombocytopenia

A
  • look for molecular clonal marker in their blood
  • consider inflammatory/infective markers (e.g. CRP) and ferritin
  • a urine dip for blood or subclinical
    infection
  • look for cancer
  • if things are still unclear after these tests, a bone marrow biopsy may be needed.
188
Q

What features are needed to make a diagnosis of essential thrombocythaemia?

A

WHO criteria:
– the need for a SUSTAINED elevated platelet count
– the absence of other myeloid malignancy
– the absence of a reactive cause for thrombocytosis
– presence of an acquired mutation e.g. JAK2 V617F as a supportive but not essential factor.

189
Q

What complication explains the abdominal pain, nausea and constipation seen in MM patients?

A

hypercalcaemia- osteoclast action

also in other malignancies with bony metastases

190
Q

Symptoms of MM

A
CRAB: 
C = calcium (elevated)
R = renal failure
A = anemia
B = bone lesions
191
Q

If you suspect someone has MM, what tests should you do?

A
  • serum protein electrophoresis to look for a monoclonal band (paraprotein) and serum free light chains to look for clonal light chain production
  • Urine electrophoresis will also be useful to pick up a monoclonal light chain band.

Making a formal diagnosis of myeloma, however, requires a bone marrow biopsy, which will demonstrate an increased
population of plasma cells.

192
Q

Prognosis of myeloma

A

The 5 year survival is just under 50%. 10 year survival is approximately 33%

193
Q

Smear cells

A

CLL

194
Q

Examples of low grade lymphoproliferative disorders

A
  • chronic lymphocytic leukaemia (CLL)
  • follicular lymphoma
  • MALT lymphoma (aka marginal zone lymphoma)
  • lymphoplasmacytic lymphoma
195
Q

Complications of low grade lymphoproliferative disorders

A

Immune complications are not uncommon (e.g. autoimmune haemolytic anaemia, immune thrombocytopenic purpura).

Marrow failure may arise if the lymphocyte clone expands sufficiently to impact on the production of other cell lines.

Finally, all low grade lymphoproliferative disorders may transform to become high grade. In CLL, this is known as Richter’s transformation, and has a very poor prognosis.