Haematology EMQs Flashcards

1
Q
A Iron deficiency anaemia
B β-Thalassaemia
C Anaemia of chronic disease
D Blood loss
E Alcohol
F Vitamin B12 deficiency
G Renal failure
H Aplastic anaemia
I Lead poisoning

A 35-year-old man presents to his GP with a 1-month history of increased
tiredness. The patient also admits to diarrhoea and minor abdominal pain during
this period. His blood tests reveal the following:
Hb 9.5 (13–18 g/dL)
MCV 64 (76–96 fL)
Fe 12.2 (14–31 μmol/L)
TIBC 74 (45–66 μmol/L)
Ferritin 9.2 (12–200 μg/L)

A

A Iron deficiency anaemia

Iron deficiency anaemia (IDA; A) causes a hypochromic (pallor of the
red blood cells on blood film due to reduced Hb synthesis), microcytic
(small size) anaemia (low haemoglobin). A reduction in serum iron can
be caused by a number of factors, including inadequate intake, malabsorption
(coeliac disease; most likely cause in this case given diarrhoea
and abdominal pain), increased demand (pregnancy) and increased
losses (bleeding and parasitic infections). Further studies are required to
distinguish IDA from other causes of microcytic anaemia: serum ferritin
will be low, while total iron binding capacity (TIBC) and transferrin will
be high.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q
A Iron deficiency anaemia
B β-Thalassaemia
C Anaemia of chronic disease
D Blood loss
E Alcohol
F Vitamin B12 deficiency
G Renal failure
H Aplastic anaemia
I Lead poisoning

A 56-year-old vagrant man presents to the accident and emergency department
with weakness in his legs. The patient has a history of poorly controlled
Crohn’s disease. His blood tests demonstrate Hb 9.4 (13–18 g/dL) and MCV 121
(
76–96 fL). A blood film reveals the presence of hypersegmented neutrophils.

A

F Vitamin B12 deficiency

The majority of cases of vitamin B12 deficiency (F) occur secondary to
malabsorption: reduced intrinsic factor production due to pernicious
anaemia or post-gastrectomy, as well as disease of the terminal ileum.
Clinical features will be similar to those of anaemia in mild cases,
progressing to neuropsychiatric symptoms and subacute degeneration
of the spinal cord (SDSC) in severe cases. Vitamin B12 deficiency
results in a macrocytic megaloblastic anaemia as a result of inhibited
DNA synthesis (B12 is responsible for the production of thymidine).
Hypersegmented neutrophils are pathognomonic of megaloblastic
anaemia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q
A Iron deficiency anaemia
B β-Thalassaemia
C Anaemia of chronic disease
D Blood loss
E Alcohol
F Vitamin B12 deficiency
G Renal failure
H Aplastic anaemia
I Lead poisoning

A 65-year-old man is referred to the haematology department by his GP after
initially presenting with tiredness, palpitations, petechiae and recent pneumonia.
His blood tests reveal Hb 9.8 (13–18 g/dL), MCV 128 (76–96 fL), reticulocyte
count 18 (25–100 × 109/L), 1.2 (2–7.5 × 109/L) and platelet count 125
(150–400 × 109/L).

A

H Aplastic anaemia

Aplastic anaemia (H) is caused by failure of the bone marrow resulting
in a pancytopenia and hypocellular bone marrow. Eighty per cent
of cases are idiopathic, although 10 per cent are primary (dyskeratosis
congenita and Fanconi anaemia) and 10 per cent are secondary (viruses,
SLE, drugs and radiation). The pathological process involves CD8+/
HLA-DR+ T cell destruction of bone marrow resulting in fatty changes.
Investigations will reveal reduced Hb, reticulocytes, neutrophils, platelets
and bone marrow cellularity as well as a raised MCV. Macrocytosis
results from the release of fetal haemoglobin in an attempt to compensate
for reduced red cell production.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q
A Iron deficiency anaemia
B β-Thalassaemia
C Anaemia of chronic disease
D Blood loss
E Alcohol
F Vitamin B12 deficiency
G Renal failure
H Aplastic anaemia
I Lead poisoning
A 56-year-old woman presents to her GP with increased tiredness in the past
few weeks. A past medical history of rheumatoid arthritis is noted. Her blood
tests demonstrate the following:
Hb 8.6 (11.5–16 g/dL)
MCV 62 (76–96 fL)
Fe 10.2 (11–30 μmol/L)
TIBC 38 (45–66 μmol/L)
Ferritin 220 (12–200 μg/L)
A

C Anaemia of chronic disease

Anaemia of chronic disease (ACD; C) occurs in states of chronic infection
and inflammation, for example in tuberculosis (TB), rheumatoid
arthritis, inflammatory bowel disease and malignant disease. ACD is
mediated by IL-6 produced by macrophages which induces hepcidin
production by the liver. Hepcidin has the effect of retaining iron in
macrophages (reduced delivery to red blood cells for erythropoiesis) and
reduces export from enterocytes (reduced plasma iron levels). Laboratory features of ACD include a microcytic hypochromic anaemia, rouleaux
formation (increased plasma proteins), raised ferritin (acute phase protein)
as well as reduced serum iron and TIBC.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q
A Iron deficiency anaemia
B β-Thalassaemia
C Anaemia of chronic disease
D Blood loss
E Alcohol
F Vitamin B12 deficiency
G Renal failure
H Aplastic anaemia
I Lead poisoning

A 12-year-old Mediterranean boy presents to his GP with increased tiredness
over the past few weeks which is affecting his ability to concentrate at school.
Examination is normal. Blood tests demonstrate the following:
Hb 9.5 (13–18 g/dL)
MCV 69 (76–96 fL)
Fe 18.2 (14–31 μmol/L)
TIBC 54 (45–66 μmol/L)
Ferritin 124 (12–200 μg/L)

A

B β-Thalassaemia

β-Thalassaemia (B) is a genetic disorder characterized by the reduced or
absent production of β-chains of haemoglobin. Mutations affecting the
β-globin genes on chromosome 11 lead to a spectrum of clinical features
depending on the combinations of chains affected. β-Thalassaemia
minor affects one β-globin chain and is usually asymptomatic, but may
present with mild features of anaemia. Haematological tests reveal a
microcytic anaemia but iron studies will be normal, differentiating from
iron deficiency anaemia. β-Thalassaemia major occurs due to defects
of both β-globin chains and results in severe anaemia requiring regular
blood transfusions, as well as skull bossing and hepatosplenomegaly.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q
A Hereditary sherocytosis
B Sickle cell anaemia
C β-Thalassaemia
D Glucose-6-phosphate
dehydrogenase deficiency
E Pyruvate kinase deficiency
F Autoimmune haemolytic anaemia
G Haemolytic disease of the
newborn
H Paroxysmal nocturnal
haemoglobinuria
I Microangiopathic haemolytic
anaemia

A 48-year-old woman diagnosed with chronic lymphocytic leukaemia develops
jaundice and on examination is found to have conjunctival pallor. Direct
antiglobulin
test is found to be positive at 37°C.

A

F Autoimmune haemolytic anaemia

Autoimmune haemolytic anaemia (AIHA; F) is caused by autoantibodies
that bind to red blood cells (RBCs) leading to splenic destruction. AIHA
can be classified as either ‘warm’ or ‘cold’ depending on the temperature
at which antibodies bind to RBCs. Warm AIHA is IgG mediated,
which binds to RBCs at 37°C; causes include lymphoproliferative disorders,
drugs (penicillin) and autoimmune diseases (SLE). Cold AIHA is
IgM mediated which binds to RBCs at temperatures less than 4°C; this
phenomenon usually occurs after an infection by mycoplasma or EBV.
Direct antiglobulin test (DAT) is positive in AIHA and spherocytes are
seen on blood film.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q
A Hereditary sherocytosis
B Sickle cell anaemia
C β-Thalassaemia
D Glucose-6-phosphate
dehydrogenase deficiency
E Pyruvate kinase deficiency
F Autoimmune haemolytic anaemia
G Haemolytic disease of the
newborn
H Paroxysmal nocturnal
haemoglobinuria
I Microangiopathic haemolytic
anaemia

An 18-year-old man presents to accident and emergency after eating a
meal containing Fava beans. He is evidently jaundiced and has signs suggestive
of anaemia. The patient’s blood film reveals the presence of Heinz
bodies.

A

D Glucose-6-phosphate

Glucose-6-phosphate dehydrogenase deficiency (G6PD deficiency; D) is
caused by an X-linked recessive enzyme defect. G6PD is an essential
enzyme in the red blood cell pentose phosphate pathway; the pathway maintains NADPH levels which in turn supply glutathione to neutralize
free radicals that may otherwise cause oxidative damage. Therefore,
G6PD deficient patients are at risk of oxidative crises which may be
precipitated by certain drugs (primaquine, sulphonamides and aspirin),
fava beans and henna. Attacks result in rapid anaemia, jaundice and a
blood film will demonstrate the presence of bite cells and Heinz bodies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q
A Hereditary sherocytosis
B Sickle cell anaemia
C β-Thalassaemia
D Glucose-6-phosphate
dehydrogenase deficiency
E Pyruvate kinase deficiency
F Autoimmune haemolytic anaemia
G Haemolytic disease of the
newborn
H Paroxysmal nocturnal
haemoglobinuria
I Microangiopathic haemolytic
anaemia

A 10-year-old girl presents to accident and emergency with jaundice. Blood
tests reveal uraemia and thrombocytopenia. A peripheral blood film demonstrates
the presence of schistocytes.

A

I Microangiopathic haemolytic
anaemia

Microangiopathic haemolytic anaemia (I) is caused by the mechanical
destruction of RBCs in circulation. Causes include thrombotic thrombocytopenic
pupura (TTP), haemolytic uraemic syndrome (HUS; E. coli
O157:57), disseminated intravascular coagulation (DIC) and systemic
lupus erythematosus (SLE). In all underlying causes, the potentiation
of coagulation pathways creates a mesh which leads to the intravascular
destruction of RBCs and produces schistocytes (helmet cells).
Schistocytes are broken down in the spleen, raising bilirubin levels and
initiating jaundice.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q
A Hereditary sherocytosis
B Sickle cell anaemia
C β-Thalassaemia
D Glucose-6-phosphate
dehydrogenase deficiency
E Pyruvate kinase deficiency
F Autoimmune haemolytic anaemia
G Haemolytic disease of the
newborn
H Paroxysmal nocturnal
haemoglobinuria
I Microangiopathic haemolytic
anaemia

A 9-year-old boy from sub-Saharan Africa presents to accident and emergency
with abdominal pain. On examination the child is found to have dactylitis.
Blood haemoglobin is found to be 6.2 g/dL and electrophoresis reveals the
diagnosis.

A

B Sickle cell anaemia

Sickle cell anaemia (B) is an autosomal recessive genetic haematological
condition due to a point mutation in the β-globin chain of haemoglobin
(chromosome 11); this mutation causes glumatic acid at position six to
be substituted by valine. Homozygotes for the mutation (HbSS) have
sickle cell anaemia while heterozygotes (HbAS) have sickle cell trait.
The mutation results in reduced RBC elasticity; RBCs therefore assume a
sickle shape which leads to the numerous complications associated with
a crisis. Blood tests will reveal an anaemia, reticulocytosis and raised
bilirubin. Haemoglobin electrophoresis will distinguish between HbSS
and HbAS.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q
A Hereditary sherocytosis
B Sickle cell anaemia
C β-Thalassaemia
D Glucose-6-phosphate
dehydrogenase deficiency
E Pyruvate kinase deficiency
F Autoimmune haemolytic anaemia
G Haemolytic disease of the
newborn
H Paroxysmal nocturnal
haemoglobinuria
I Microangiopathic haemolytic
anaemia

A 1-day old baby has developed severe jaundice on the neonatal ward.
The mother is rhesus negative and has had one previous pregnancy. Due
to having her first baby abroad, she was not administered prophylactic
anti-
D.

A

G Haemolytic disease of the
newborn

Haemolytic disease of the newborn (G) occurs when the mother’s blood is
rhesus negative and the fetus’ blood is rhesus positive. A first pregnancy
or a sensitizing event such as an abortion, miscarriage or antepartum
haemorrhage leads to fetal red blood cells entering the maternal circulation
resulting in the formation of anti-D IgG. In a second pregnancy,
maternal anti-D IgG will cross the placenta and coat fetal red blood cells
which are subsequently haemolyzed in the spleen and liver. Therefore,
anti-D prophylaxis is given to at-risk mothers; anti-D will coat any fetal
red blood cells in the maternal circulation causing them to be removed
by the spleen prior to potentially harmful IgG production.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q
A Anisocytosis
B Howell–Jolly bodies
C Heinz bodies
D Rouleaux formation
E Spherocytes
F Target cells
G Cabot rings
H Pappenheimer bodies
I Tear-drop cells

A 34-year-old man, who has a past medical history of splenectomy following
splenic trauma, presents to his GP with malaise 2 weeks after returning from
abroad. Routine blood results are found to be normal but a blood film demonstrates
inclusions within erythrocytes.

A

B Howell–Jolly bodies

Howell–Jolly bodies (B) are nuclear DNA remnants found in circulating
erythrocytes. On haematoxylin and eosin stained blood film they appear
as purple spheres within erythrocytes. In healthy individuals erythrocytes
expel nuclear DNA during the maturation process within the
bone marrow; the few erythrocytes containing Howell–Jolly bodies are
removed by the spleen. Common causes of Howell–Jolly bodies include
splenectomy secondary to trauma and autosplenectomy resulting from
sickle cell disease.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q
A Anisocytosis
B Howell–Jolly bodies
C Heinz bodies
D Rouleaux formation
E Spherocytes
F Target cells
G Cabot rings
H Pappenheimer bodies
I Tear-drop cells

A 66-year-old man has a gastroscopy and colonoscopy following a blood test
which demonstrated a microcytic anaemia. The patient had complained of tiredness
and significant weight loss over a 1-month period.

A

A Anisocytosis

Anisocytosis (A) is defined as the variation in the size of circulating
erythrocytes. The most common cause is iron deficiency anaemia (IDA),
but thalassaemia, megaloblastic anaemia and sideroblastic anaemia are
all causative. As well as blood film analysis, anisocytosis may be detected
as a raised red cell distribution width (RDW), a measure of variation
in size of red blood cells. In the case of IDA, anisocytosis results due to
deficient iron supply to produce haemoglobin.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q
A Anisocytosis
B Howell–Jolly bodies
C Heinz bodies
D Rouleaux formation
E Spherocytes
F Target cells
G Cabot rings
H Pappenheimer bodies
I Tear-drop cells

A 36-year-old woman presents to her GP after a 1-month history of tiredness
and recurrent chest infections. Blood tests reveal a pancytopenia and a subsequent
bone marrow aspirate reveals a dry tap.

A

I Tear-drop cells

Tear-drop cells (I), also known as dacrocytes, are caused by myelofibrosis.
The pathogenesis of myelofibrosis is defined by the bone marrow
undergoing fibrosis, usually following a myeloproliferative disorder
such as polycythaemia rubra vera or essential thrombocytosis. Bone
marrow production of blood cells decreases resulting in a pancytopenia.
The body compensates with extra-medullary haemopoiesis causing
hepatosplenomegaly. Blood film will demonstrate leuko-erythroblasts,
tear-drop cells and circulating megakaryocytes. Bone marrow aspirate is
described as a ‘dry and bloody’ tap.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q
A Anisocytosis
B Howell–Jolly bodies
C Heinz bodies
D Rouleaux formation
E Spherocytes
F Target cells
G Cabot rings
H Pappenheimer bodies
I Tear-drop cells

A 3-week-old neonate is found to have prolonged jaundice with serious risk of
kernicterus. Blood film demonstrates the presence of ‘bite cells’ as well as inclusions
within erythrocytes.

A

C Heinz bodies

Heinz bodies (C) are inclusion bodies found within erythrocytes that
represent denatured haemoglobin as a result of reactive oxygen species.
Heinz bodies are most commonly caused by erythrocyte enzyme deficiencies
such as glucose-6-phosphate dehydrogenase (G6PD) deficiency,
which may present in neonates with prolonged jaundice and NADPH
deficiency (leading to accumulation of hydrogen peroxide), as well
as chronic liver disease and α-thalassaemia. Damaged erythrocytes are
removed in the spleen by macrophages leading to the formation of ‘bite
cells’.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q
A Anisocytosis
B Howell–Jolly bodies
C Heinz bodies
D Rouleaux formation
E Spherocytes
F Target cells
G Cabot rings
H Pappenheimer bodies
I Tear-drop cells

A 45-year-old woman with known Graves’ diseases presents to her GP with
increased tiredness. She is found to have a megaloblastic anaemia.

A

G Cabot rings

Cabot rings (G) are looped structures found within erythrocytes which
may be caused by megaloblastic anaemia, i.e. inhibition of erythrocyte
production occurring as a result of reduced DNA synthesis secondary to vitamin B12 deficiency. Vitamin B12 deficiency is most commonly caused
by intrinsic factor (a protein required for vitamin B12 absorption) deficiency
as a result of pernicious anaemia. Pernicious anaemia is caused
by antibody destruction of gastric parietal cells which produce intrinsic
factor and may be associated with other autoimmune diseases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q
A Immune thrombocytopenic
purpura
B Idiopathic thrombotic
thrombocytopenic purpura
C Disseminated intravascular
coagulation
D Glanzmann’s thrombasthenia
E Von Willebrand disease
F Haemophilia A
G Haemophilia B
H Hereditary haemorrhagic
telangiectasia
I Bernard–Soulier syndrome

A 4-year-old girl is seen by her GP due to recent onset petechiae on her feet
and bleeding of her gums when she brushes her teeth. The child’s platelet count
is found to be 12 500 per μL. The GP prescribes prednisolone and reassures the
child’s mother that the bleeding will resolve.

A

A Immune thrombocytopenic
purpura

Immune thrombocytopenic purpura (ITP; A) may follow either an acute or
chronic disease process. Acute ITP most commonly occurs in children, usually
occurring 2 weeks after a viral illness. It is a type 2 hypersensitivity
reaction, with IgG binding to virus-coated platelets. The fall in platelets is
very low (less than 20 × 109/L) but is a self-limiting condition (few weeks).
Chronic ITP is gradual in onset with no history of previous viral infection.
It is also a type 2 hypersensitivity reaction with IgG targeting GLP-2b/3a.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q
A Immune thrombocytopenic
purpura
B Idiopathic thrombotic
thrombocytopenic purpura
C Disseminated intravascular
coagulation
D Glanzmann’s thrombasthenia
E Von Willebrand disease
F Haemophilia A
G Haemophilia B
H Hereditary haemorrhagic
telangiectasia
I Bernard–Soulier syndrome

A 28-year-old man attends the haematology outpatient clinic regarding a longstanding
condition he has suffered from. His disorder is related to a deficiency
in factor 8 and therefore requires regular transfusions to replace this clotting
factor.

A

F Haemophilia A

Haemophilia A (F) is an X-linked genetic disorder and hence only
affects men. Haemophilia A is characterized by a deficiency in factor 8.
Haemophilia A is diagnosed by a reduced APTT as well as reduced
factor
8. Symptoms depend on severity of disease: mild disease features
bleeding after surgery/trauma; moderate disease results in bleeding
after minor trauma; severe disease causes frequent spontaneous bleeds.
Clinical features include haemarthrosis (causing fixed joints) and muscle
haematoma (causing atrophy and short tendons).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q
A Immune thrombocytopenic
purpura
B Idiopathic thrombotic
thrombocytopenic purpura
C Disseminated intravascular
coagulation
D Glanzmann’s thrombasthenia
E Von Willebrand disease
F Haemophilia A
G Haemophilia B
H Hereditary haemorrhagic
telangiectasia
I Bernard–Soulier syndrome

A 34-year-old man is taken to the local accident and emergency after suffering
an episode of jaundice, fever and worsening headache. Blood tests reveal a low
platelet count and blood film is suggestive of a microangiopathic haemolytic
anaemia picture.

A

B Idiopathic thrombotic
thrombocytopenic purpura

Idiopathic thrombotic thrombocytopenic purpura (B) occurs due to
platelet microthrombi. Presenting features include microangiopathic
haemolytic anaemia (red blood cells coming into contact with microscopic
clots are damaged by shear stress), renal failure, thrombocytopenia,
fever and neurological signs (hallucinations/stroke/headache). A
mutation in the ADAM-ST13 gene, coding for a protease that cleaves
von Willebrand factor (vWF) allows for the formation of vWF multimers
enabling platelet thrombi to form causing organ damage.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q
A Immune thrombocytopenic
purpura
B Idiopathic thrombotic
thrombocytopenic purpura
C Disseminated intravascular
coagulation
D Glanzmann’s thrombasthenia
E Von Willebrand disease
F Haemophilia A
G Haemophilia B
H Hereditary haemorrhagic
telangiectasia
I Bernard–Soulier syndrome

A 68-year-old man on the Care of the Elderly ward is confirmed to have Gramnegative
sepsis. The patient is bleeding from his mouth and is in shock. Initial
blood tests reveal a reduced platelet count, anaemia and renal failure.

A

C Disseminated intravascular
coagulation

Disseminated intravascular coagulation (DIC; C) may be caused by
Gram-negative sepsis, malignancy, trauma, placental abruption or
amniotic fluid embolus. Tissue factor is released which triggers the
activation of the clotting cascade, leading to platelet activation
(thrombosis in microcirculation) and fibrin deposition (haemolysis).
The consumption of platelets and clotting factors predisposes to
bleeding. Plasmin is also generated in DIC which causes fibrinolysis,
perpetuating the bleeding risk. The clinical manifestations of
DIC are therefore linked to microthombus production (renal failure
and neurological signs) and reduced platelets, clotting factors
and increased fibrinolysis (bruising, gastrointestinal bleeding and
shock).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q
A Immune thrombocytopenic
purpura
B Idiopathic thrombotic
thrombocytopenic purpura
C Disseminated intravascular
coagulation
D Glanzmann’s thrombasthenia
E Von Willebrand disease
F Haemophilia A
G Haemophilia B
H Hereditary haemorrhagic
telangiectasia
I Bernard–Soulier syndrome

A 2-year-old boy is taken to see the GP due to his mother noticing bruising
on his arms and legs after playing in the park. The parent mentions that she
has also noticed several recent nose bleeds in her son but thought he would
‘grow out of it’. Investigations reveal a low APTT, low factor 8 levels and low
Ristocetein cofactor activity.

A

E Von Willebrand disease

von Willebrand disease (vWD; E) is an autosomal dominant condition
caused by a mutation on chromosome 12. Physiologically, von
Willebrand factor (vWF) has two roles: platelet adhesion and factor 8
production. Therefore, in vWD, where there is a deficiency in vWF,
there is a defect in platelet plug formation as well as low levels of factor
8. Clinically, patients will present with gum bleeding, epistaxis or
prolonged bleeding after surgery. Investigations will reveal a high/
normal APTT, low factor 8 levels, low ristocetin cofactor activity, poor
ristocetin aggregation and normal PTT,

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q
A Factor V Leiden
B Antiphospholipid syndrome
C Malignancy
D Protein S deficiency
E Antithrombin deficiency
F Prothrombin G20210A mutation
G Oral contraceptive pill
H Buerger’s disease
I Chronic liver disease

A 35-year-old Caucasian man presents to accident and emergency with deep
pain and swelling in his left calf. His past medical history reveals history of
recurrent DVTs. The patient’s notes reveal a letter from his haematologist who
had diagnosed a condition caused by a substitution mutation.

A

F Prothrombin G20210A mutation

Prothrombin G20210A (F) is an inherited thrombophilia caused by the
substitution of guanine with adenine at the 20210 position of the prothrombin
gene. Physiologically, prothrombin promotes clotting after
a blood vessel has been damaged. The G20210A causes the amplification
of prothrombin production thereby increasing the risk of clotting,
and causing a predisposition to deep vein thrombosis and pulmonary embolism.
The prevalence of the mutation is approximately 5 per cent
in the Caucasian population, the race with the greatest preponderance.

22
Q
A Factor V Leiden
B Antiphospholipid syndrome
C Malignancy
D Protein S deficiency
E Antithrombin deficiency
F Prothrombin G20210A mutation
G Oral contraceptive pill
H Buerger’s disease
I Chronic liver disease

A 38-year-old woman presents to accident and emergency with abdominal
pain as well as passing blood and tissue per vagina. Ectopic pregnancy
is diagnosed after ultrasound. The patient’s past medical history includes a
haematological
condition in which a clotting factor is unable to be degraded
by activated protein C.

A

A Factor V Leiden

Factor V Leiden (A) is an autosomal dominant inherited thrombophilia.
Under normal circumstances protein C inhibits factor 5. In Factor V
Leiden a mutation of the F5 gene that codes for factor 5, whereby an
arginine codon is substituted for a glutamine codon, results in impaired
degradation of factor 5 by protein C. As a result, patients are at risk of
deep vein thrombosis and miscarriage. Diagnostic tests determine the
functionality of activated protein C.

23
Q
A Factor V Leiden
B Antiphospholipid syndrome
C Malignancy
D Protein S deficiency
E Antithrombin deficiency
F Prothrombin G20210A mutation
G Oral contraceptive pill
H Buerger’s disease
I Chronic liver disease

A 32-year-old woman is seen by her rheumatologist to follow up her longstanding
systemic lupus erythematosus (SLE). The patient has a history of
recurrent
miscarriages. The woman is positive for anti-cardiolipin antibodies and
lupus anticoagulant.

A

B Antiphospholipid syndrome

Antiphospholipid syndrome (APLS; B) is an autoimmune disorder that
may present with stroke (arterial thrombosis), deep vein thrombosis
(venous thrombosis) and/or recurrent miscarriages. APLS may be primary
(not associated with autoimmune disease) or secondary to autoimmune
disease such as SLE. Anti-cardiolipin antibodies and lupus
anticoagulant bind to phospholipids on cell surface membranes of cells
causing the activation of the coagulation cascade and thereby promoting
clot formation. Diagnosis involves demonstrating the presence of
circulating anti-cardiolipin antibodies and lupus anticoagulant.

24
Q
A Factor V Leiden
B Antiphospholipid syndrome
C Malignancy
D Protein S deficiency
E Antithrombin deficiency
F Prothrombin G20210A mutation
G Oral contraceptive pill
H Buerger’s disease
I Chronic liver disease

A 45-year-old man, who has a 50 pack/year history of smoking, is referred to
the vascular outpatient clinic by his GP after suffering intermittent claudication.
A diagnostic angiogram reveals a corkscrew appearance of his lower
limb arteries.

A

H Buerger’s disease

Buerger’s disease (thromboangitis obliterans; H) is a vasculitis of small/
medium arteries and veins of the hands and feet; it is strongly related
to smoking. Claudication may be the initial presentation but as the
disease progresses there is an association with recurrent arterial and
venous thrombosis leading to gangrene and amputation in severe cases.
Angiograms of the upper and lower limbs are helpful in the diagnosis of
Buerger’s disease; a corkscrew appearance of the arteries may arise due
to persistent vascular damage.

25
Q
A Factor V Leiden
B Antiphospholipid syndrome
C Malignancy
D Protein S deficiency
E Antithrombin deficiency
F Prothrombin G20210A mutation
G Oral contraceptive pill
H Buerger’s disease
I Chronic liver disease

A 37-year-old man presents to accident and emergency with shortness of
breath and severe pleuritic chest pain. A CTPA reveals the diagnosis of pulmonary
embolism. The patient’s haematological records state the patient has a
condition that leads to the persistence of factors 5a and 8a causing increased
risk of venous thrombosis.

A

D Protein S deficiency

Protein S deficiency (D) is associated with the impaired degradation of
factors Va and VIIIa. Protein S and protein C are physiological anticoagulants.
Deficiency of protein S leads to persistence of factors 5a and
8a in the circulation and hence patients have a susceptibility to venous
thrombosis. Three types of protein S deficiency exist: type I (quantitative
defect) and types II and III (qualitative defect). Since protein S is a
vitamin K dependent anticoagulant, warfarin treatment and liver disease
may also lead to venous thrombosis in rare cases (the majority of cases
show increased bleeding).

26
Q
A Immediate haemolytic transfusion
reaction
B Febrile non-haemolytic reaction
C Iron overload
D IgA deficiency
E Transfusion related lung injury
F Bacterial infection
G Delayed haemolytic transfusion
reaction
H Fluid overload
I Graft versus host disease

An 82-year-old man has just received a blood transfusion following a low haemoglobin
level on the Care of the Elderly ward. He is now short of breath and is
coughing up pink frothy sputum.

A

H Fluid overload

Fluid overload (H) is an immediate complication of blood transfusion.
Clinical features suggestive of fluid overload will include dyspnoea,
distended neck veins and pink frothy sputum. Usually fluid overload
occurs in situations where the blood transfusion rate is too fast; a
transfusion would generally have to run at more than 2 mL/kg/hour to
induce fluid overload. Patients with pre-existing cardiac or renal failure
are prone to fluid overload as a result of blood transfusion.

27
Q
A Immediate haemolytic transfusion
reaction
B Febrile non-haemolytic reaction
C Iron overload
D IgA deficiency
E Transfusion related lung injury
F Bacterial infection
G Delayed haemolytic transfusion
reaction
H Fluid overload
I Graft versus host disease

A 34-year-old HIV-positive man receives a regular blood transfusion as part of
his beta-thalassaemia major treatment regimen. He soon develops diarrhoea
and a maculopapular rash on his limbs.

A

I Graft versus host disease

Graft versus host disease (GVHD; I) occurs due to the transfer of donor
lymphocytes to the recipient in a blood transfusion in patients who are
immunosuppressed. Normally, the immune system is strong enough to
detect and destroy donor lymphocytes. However, in immunosuppression
(stem cell transplant patients/chemotherapy/malignancy/HIV) the donor
lymphocytes cannot be destroyed; these foreign lymphocytes persist
and target host tissue, especially the gastrointestinal tract and skin.
Symptoms of GVHD include diarrhoea, maculopapular rash and skin
necrosis. To minimize GVHD, donor blood is irradiated to remove
lymphocytes.

28
Q
A Immediate haemolytic transfusion
reaction
B Febrile non-haemolytic reaction
C Iron overload
D IgA deficiency
E Transfusion related lung injury
F Bacterial infection
G Delayed haemolytic transfusion
reaction
H Fluid overload
I Graft versus host disease

A 34-year-old man requires a blood transfusion following a road traffic accident.
However, soon after the transfusion, the patient is dyspnoeic and hypotensive.
Investigation into the patient’s past medical history reveals a history of
recurrent chest and gastrointestinal infections.

A

D IgA deficiency

IgA deficiency (D) leads to recurrent mild infections of the mucous
membranes lining the airways and digestive tract. In affected patients
serum IgA levels are undetectable but IgG and IgM levels are normal.
IgA is found in mucous secretions from the respiratory and gastrointestinal
tracts and plays a key role in mucosal immunity. IgA deficient
patients are also predisposed to severe anaphylactic reactions to blood
transfusions due to the presence of IgA in donor blood.

29
Q
A Immediate haemolytic transfusion
reaction
B Febrile non-haemolytic reaction
C Iron overload
D IgA deficiency
E Transfusion related lung injury
F Bacterial infection
G Delayed haemolytic transfusion
reaction
H Fluid overload
I Graft versus host disease

A 56-year-old man is given a blood transfusion following severe blood loss
after a hip replacement operation. Three hours after the transfusion, the patient
develops shortness of breath, a dry cough and a fever of 39°C.

A

E Transfusion related lung injury

Transfusion-related lung injury (TRALI; E) is characterized by acute
non-cardiogenic pulmonary oedema that occurs within 6 hours following
blood transfusion. The pathogenesis of TRALI involves the presence
of anti-white blood cell antibodies in the donor blood that attack host
leukocytes; sensitizing events in donors include previous blood transfusion
or transplantation. Clinical features of TRALI are dry cough, dyspnoea
and fever.

30
Q
A Immediate haemolytic transfusion
reaction
B Febrile non-haemolytic reaction
C Iron overload
D IgA deficiency
E Transfusion related lung injury
F Bacterial infection
G Delayed haemolytic transfusion
reaction
H Fluid overload
I Graft versus host disease

A 29-year-old woman requires an immediate blood transfusion after suffering
a post-partum haemorrhage. However, 30 minutes after her transfusion she
develops abdominal pain, facial flushing and vomiting. Analysis of the woman’s
urine reveals the presence of haemoglobin.

A

A Immediate haemolytic transfusion
reaction

Immediate haemolytic transfusion reaction (IHTR; A) is characterized
by ABO incompatibility and occurs 1–2 hours post-transfusion. Clinical
features include abdominal pain, loin pain, facial flushing, vomiting and
haemoglobinuria. Host IgG and IgM target donor red blood cells which are
subsequently removed by the reticuloendothelial system. The most severe
reaction occurs if a group O patient is transfused with group A blood.

31
Q
A Acute lymphoblastic leukaemia
B Acute promyelocytic leukaemia
C Chronic myeloid leukaemia
D Chronic lymphocytic leukaemia
E Hairy cell leukaemia
F T-cell prolymphocytic leukaemia
G Large granular lymphocytic
leukaemia
H Adult T-cell leukaemia
I Acute myeloid leukaemia

A 62-year-old woman is seen by a GP due to a recent chest infection that has
been troubling her. Initial blood tests show an elevated white cell count with
specifically raised granulocytes. Following referral to a haematologist, a bone
marrow biopsy reveals a hypercellular bone marrow and cytogenetic screening
suggests a translocation between chromosomes 9 and 22.

A

C Chronic myeloid leukaemia

Chronic myeloid leukaemia (CML; C) is most prevalent in the elderly
population and is commonly suspected secondary to routine blood tests.
Blood results will show an elevated level of granulocytes (neutrophils,
basophils and eosinophils). Blood film will demonstrate myeloid cells
at different stages of maturation. Bone marrow biopsy in CML patients
suggests hypercellularity. Ninety-five per cent of cases are caused by
the Philadelphia chromosome, a chromosomal translocation between
chromosomes 9 and 22; this results in the BCR-Abl fusion oncogene
that has tyrosine kinase activity. Recent novel therapies for CML include
imatinib, a BCR-Abl inhibitor.

32
Q
A Acute lymphoblastic leukaemia
B Acute promyelocytic leukaemia
C Chronic myeloid leukaemia
D Chronic lymphocytic leukaemia
E Hairy cell leukaemia
F T-cell prolymphocytic leukaemia
G Large granular lymphocytic
leukaemia
H Adult T-cell leukaemia
I Acute myeloid leukaemia

A 41-year-old man is referred to a haematologist by his general practitioner
after several recent chest infections and tiredness. On examination, bruises
are seen on his lower limbs as well as splenomegaly. Initial blood tests reveal a
pancytopenia. Further testing demonstrates the presence of tumour cells that
express tartrate-resistant acid phosphatase.

A

E Hairy cell leukaemia

Hairy cell leukaemia (HCL; E) is a haematological malignancy of
B lymphocytes and a subtype of chronic lymphocytic leukaemia. It
most commonly occurs in middle-aged men. The cancer derives its
name from the fine hair-like projections that are seen on tumour cells
on microscopy. Cell surface markers include CD25 (IL-2 receptor) and
CD11c (adhesion molecule). Diagnosis can be confirmed by the presence
of tartrate-resistant acid phosphatase (TRAP) on cytochemical analysis.
Clinical features relate to invasion of the spleen (splenomegaly), liver
(hepatomegaly) and bone marrow (pancytopenia).

33
Q
A Acute lymphoblastic leukaemia
B Acute promyelocytic leukaemia
C Chronic myeloid leukaemia
D Chronic lymphocytic leukaemia
E Hairy cell leukaemia
F T-cell prolymphocytic leukaemia
G Large granular lymphocytic
leukaemia
H Adult T-cell leukaemia
I Acute myeloid leukaemia

A 60-year-old man presents to his GP with fever, malaise and cough. On examination,
the man is found to have petechiae on his legs as well as gum hypertrophy.
Blood tests reveal anaemia, leukocytopenia and thrombocytopenia. A blood
film demonstrates the presence of Auer rods within blast cells.

A

I Acute myeloid leukaemia

Acute myeloid leukaemia (AML; I) is characterized by more than 20 per
cent myleoblasts in the bone marrow. AML also causes proliferation
of megakaryocytes and erythrocytes. Mutations that can cause AML
include internal tandem duplications of the FLT3 gene (coding for a
tyrosine kinase) and t(8;21) (a translocation causing a compressor complex
to inhibit haematopoietic differentiation. Primary causes include
Down’s syndrome; secondary causes include myeloproliferative disease. Blood tests will reveal a variable white cell count, anaemia,
thrombocytopenia and reduced neutrophil count. Auer rods on blood
film are pathognomonic, which will also be leuko-erythroblastic.
Immunophenotyping of CD13, CD33 or CD34 can also aid diagnosis.

34
Q
A Acute lymphoblastic leukaemia
B Acute promyelocytic leukaemia
C Chronic myeloid leukaemia
D Chronic lymphocytic leukaemia
E Hairy cell leukaemia
F T-cell prolymphocytic leukaemia
G Large granular lymphocytic
leukaemia
H Adult T-cell leukaemia
I Acute myeloid leukaemia

A 42-year-old Japanese migrant presents to his GP with generalized lymphadenopathy
and nodules on his arms. On examination the patient has hepatosplenomegaly.
Blood tests reveal lymphocytosis and a raised calcium level.

A

H Adult T-cell leukaemia

Adult T-cell leukaemia (adult T-cell lymphoma; ATL; H) is a rare haematological
malignancy with poor prognosis. It is caused by human T-cell
leukaemia virus type 1 (HTLV-1), endemic in Japan and the Caribbean.
Tumour cells express the cell surface protein CD4 and will contain the
HTLV-1 virus within; the nuclei of ATL cells have a characteristic cloverleaf
appearance. Clinical features include lymphadenopathy, hepatosplenomegaly,
skin lesions and hypercalcaemia.

35
Q
A Acute lymphoblastic leukaemia
B Acute promyelocytic leukaemia
C Chronic myeloid leukaemia
D Chronic lymphocytic leukaemia
E Hairy cell leukaemia
F T-cell prolymphocytic leukaemia
G Large granular lymphocytic
leukaemia
H Adult T-cell leukaemia
I Acute myeloid leukaemia

A 70-year-old man is reviewed by his GP after having felt tired and experienced
weight loss over a 2-month period. The patient has lymphadenopathy on
examination. Blood tests demonstrates a lymphocytosis of 4500 cells per microlitre
and smudge cells can be visualized on a peripheral blood film.

A

D Chronic lymphocytic leukaemia

Chronic lymphocytic leukaemia (CLL; D) is a B-cell neoplasm characterized
by a lymphocyte count of over 4000 cells per microlitre. CLL
most commonly occurs in elderly men. The cancer presents primarily
in the lymph nodes with small lymphocytes containing irregular nuclei
mixed with larger prolymphocytes. Prolymphocytes may aggregate to
form pathognomonic proliferation centres. Blood film may reveal the
presence of smudge cells. Clinical features are non-specific and include
tiredness and weight loss. Hypogammaglobulinaemia is an associated
immune phenomenon. CLL may convert into more aggressive forms
including prolymphocytic transformation and diffuse-large B-cell lymphoma
(Richter’s syndrome).

36
Q
A Diffuse large B-cell lymphoma
B Burkitt lymphoma
C Follicular lymphoma
D Small lymphocytic leukaemia
E Mantle cell lymphoma
F Peripheral T-cell lymphoma
G Mycosis fungoides
H Angiocentric lymphoma
I Hodgkin’s lymphoma

A 5-year-old boy is seen by a volunteer doctor at an Ethiopian refugee camp.
On examination the child has a prominent swelling on the left side of his jaw.
A tissue sample of the mass demonstrates a ‘starry sky’ appearance on light
microscopy.

A

B Burkitt lymphoma

Burkitt lymphoma (BL; B) is a haematological cancer of B lymphocytes
caused by latent Epstein–Barr viral (EBV) infection and is most prevalent
in Africa, affecting children and teenagers. Subtypes of BL include
endemic, sporadic and immunodeficiency-associated disease. Endemic
BL presents with a mandibular mass whereas non-endemic types present with an abdominal mass. All forms are highly associated with translocations
of the c-myc gene on chromosome 8 (the most common with the
Ig heavy chain on chromosome 14). A ‘starry sky’ appearance is characteristic
when viewing BL cells under microscopy.

37
Q
A Diffuse large B-cell lymphoma
B Burkitt lymphoma
C Follicular lymphoma
D Small lymphocytic leukaemia
E Mantle cell lymphoma
F Peripheral T-cell lymphoma
G Mycosis fungoides
H Angiocentric lymphoma
I Hodgkin’s lymphoma

A 52-year-old man presents to his GP with painless lymphadenopathy which he
describes as having fluctuated in size over the past month, as well as experiencing
night sweats and weight loss. He also mentions the lumps become painful
when he drinks alcohol. Further biopsy of the lumps reveals the presence of
Reed–Sternberg cells.

A

I Hodgkin’s lymphoma

Hodgkin’s lymphoma (I) results from the proliferation of B cells from
the germinal centre. The pathogenesis is linked to EBV infection which
activates NF-κB, preventing apoptosis of infected cells. Release of IL-5
from B-cells activates eosinophils, prolonging the life of B cells further.
Histologically, Hodgkin’s lymphoma is characterized by the presence
of Reed–Sternberg cells (binucleate/multinucleate cells with abundant
cytoplasm, inclusion-like nucleoli and surrounded by eosinophils).
Lymphadenopathy associated with Hodgkin’s lymphoma is usually painless,
asymmetrical, fluctuates in size and is painful with alcohol intake.
Other clinical features include fever, night sweats, weight loss and Pel–
Ebstein fever (intermittent fever every 2 weeks). Unlike non-Hodgkin’s
lymphoma, extra-nodal involvement is rare.

38
Q
A Diffuse large B-cell lymphoma
B Burkitt lymphoma
C Follicular lymphoma
D Small lymphocytic leukaemia
E Mantle cell lymphoma
F Peripheral T-cell lymphoma
G Mycosis fungoides
H Angiocentric lymphoma
I Hodgkin’s lymphoma

A 60-year-old man presents to his GP with malaise, night sweats and weight
loss. On examination the patient is found to have generalized lymphadenopathy
and hepatomegaly. Cytogenetic investigation a few weeks later by a haematologist
reveals a translocation between chromosomes 11 and 14, which has caused
overexpression of the BCL-2 protein.

A

E Mantle cell lymphoma

Mantle cell lymphoma (MCL; E) is an aggressive B-cell lymphoma primarily
affecting elderly men. The most common cause is a translocation
between chromosomes 11 and 14, involving the BCL-1 locus and
Ig heavy chain locus, therefore leading to over-expression of cyclin D1.
Over-expression of cyclin D1 leads to dysregulation of the cell cycle.
Clinically, generalized lymphadenopathy, as well as bone marrow and
liver infiltration, are common. Hodgkin’s lymphoma can be split into
classical and lymphocyte predominant nodular (LPN) subtypes.

39
Q
A Diffuse large B-cell lymphoma
B Burkitt lymphoma
C Follicular lymphoma
D Small lymphocytic leukaemia
E Mantle cell lymphoma
F Peripheral T-cell lymphoma
G Mycosis fungoides
H Angiocentric lymphoma
I Hodgkin’s lymphoma

A 40-year-old woman is referred to a haematologist after she is found to have
generalized, painless lymphadenopathy. A report on tumour cell morphology
states the presence of both centrocytes and centroblasts.

A

C Follicular lymphoma

Follicular lymphoma (C) is caused most commonly by a translocation
between chromosomes 14 and 18, leading to over-expression of the
BCL-2 protein. Over-expression of BCL-2 causes inhibition of apoptosis,
promoting the survival of tumour cells. Tumour cells in follicular
lymphoma are characterized by centrocytes (small B cells with irregular
nuclei and reduced cytoplasm) and centroblasts (larger B cells with
multiple nuclei). Clinical features include painless, generalized lymphadenopathy.
Follicular lymphoma usually presents in middle-aged patients
and has a non-aggressive course but is difficult to cure.

40
Q
A Diffuse large B-cell lymphoma
B Burkitt lymphoma
C Follicular lymphoma
D Small lymphocytic leukaemia
E Mantle cell lymphoma
F Peripheral T-cell lymphoma
G Mycosis fungoides
H Angiocentric lymphoma
I Hodgkin’s lymphoma

A 62-year-old HIV-positive man presents to a haematologist with a 3-month
history of weight loss and tiredness. On examination, the patient has a mass on
his neck which the patient states has been rapidly growing. Staining of biopsy
tissue demonstrates the present of large B cells which are positive for EBV.

A

A Diffuse large B-cell lymphoma

Diffuse large B-cell lymphoma (DLBL; A) is a haematological malignancy
most commonly affecting the elderly, characterized by large lymphocytes
which have a diffuse pattern of growth. Common chromosomal abnormalities
which contribute to the development of DLBL include the t(14;18)
translocation which is characteristic of follicular lymphoma; this suggests
that follicular lymphoma may undergo a degree of transformation to cause
DLBL in such circumstances. Tumour cells that have follicular lymphoma
morphology may be present at other sites. Two subtypes of DLBL have been
described, both of which are associated with immunodeficiency: immunodeficiency-
associated large B-cell lymphoma (linked to latent EBV infection)
and body cavity-based large cell lymphoma (linked to HHV8 infection).

41
Q
A Essential thrombocythaemia
B Myelofibrosis
C Chronic myelo-monocytic
leukaemia
D Refractory anaemia with excess
blasts
E Polycythaemia rubravera
F Refractory anaemia with ringed
sideroblasts
G Refractory anaemia
H 5q-Syndrome
I Multiple myeloma

A 40-year-old man is referred to a haematologist after suffering an episode
of petechiae on his legs followed by a burning sensation in his fingers and
deep vein thrombosis a few weeks later. Blood tests reveal a platelet count of
850 × 109/L.

A

A Essential thrombocythaemia

Essential thrombocythaemia (A) results in a high platelet count, which
quickly become dysfunctional; it is characterized by periods of bleeding
or thrombosis. Clinical features of bleeding events include gastrointestinal
bleeding, bruising, petechiae and/or menorrhagia. Thrombotic events
manifest as erythromelalgia (erythema, swelling, pain and/or burning
sensation in the extremities), digital ischaemia, cerebrovascular accident,
deep vein thrombosis and Budd–Chiari syndrome. Blood tests will demonstrate
a platelet count of over 600 × 109/L and the bone marrow will
be hypercellular with giant platelets, as well as megakaryocyte clustering
and hyperplasia. Treatment options include hydroxyurea or anagrelide.

42
Q
A Essential thrombocythaemia
B Myelofibrosis
C Chronic myelo-monocytic
leukaemia
D Refractory anaemia with excess
blasts
E Polycythaemia rubravera
F Refractory anaemia with ringed
sideroblasts
G Refractory anaemia
H 5q-Syndrome
I Multiple myeloma

A 52-year-old woman presents to her GP due to increased tiredness. The patient
also reports easy bruising and numerous bouts of pneumonia which have
occurred over the past 6 months. On examination, the patient has splenomegaly.
Blood tests reveal a low white cell and platelet count. Blood film reveals the presence
of tear drop cells and on bone marrow aspiration there is a ‘dry’ tap.

A

B Myelofibrosis

In myelofibrosis (B) the bone marrow undergoes fibrosis, the cause of which
is unknown. The body compensates with extra-medullary haemopoiesis
causing enlargement of the spleen and liver. The underlying pathogenesis
is related to abnormal megakaryocytes releasing PDGF and TGF-β which
stimulate fibroblast proliferation. Blood tests will show an initial rise in
white cell and platelet counts during the compensatory phase; as fibrosis
progresses the bone marrow reduces white cell and platelet production.
Blood film will be leukoerythroblastic, with tear-drop cells and circulating
megakaryocytes (fibrosis causes ejection of megakaryocytes from the bone
marrow). Bone marrow aspirate will demonstrate a ‘dry’ or bloody tap.

43
Q
A Essential thrombocythaemia
B Myelofibrosis
C Chronic myelo-monocytic
leukaemia
D Refractory anaemia with excess
blasts
E Polycythaemia rubravera
F Refractory anaemia with ringed
sideroblasts
G Refractory anaemia
H 5q-Syndrome
I Multiple myeloma

A 60-year-old man is referred to a haematologist after complaining of back
pain and tiredness as well as recent onset low mood. Urine tests reveal the presence
of Bence–Jones proteins. An X-ray of the patient’s spine shows the presence
of lytic lesions.

A

I Multiple myeloma

Multiple myeloma (I) is defined as the proliferation of plasma cells in
the bone marrow (>10 per cent plasma cells). Myeloma cells release
monoclonal antibodies (most commonly IgG or IgA) and/or light chains
(paraproteins); IgA production significantly increases the viscosity of
the blood. Diagnosis is based on paraprotein bands of greater than
30 g/L on electrophoresis. Blood tests will demonstrate an increased ESR and calcium levels as well as rouleaux formation on blood film.
Bence–Jones proteins (immunoglobulin light chains) may be present in
the urine. Plasma cells visualized from bone marrow biopsy are atypical,
with multiple nuclei, prominent nucleoli and cytoplasmic granules (containing
immunoglobulin). X-rays may reveal punched-out lytic lesions.

44
Q
A Essential thrombocythaemia
B Myelofibrosis
C Chronic myelo-monocytic
leukaemia
D Refractory anaemia with excess
blasts
E Polycythaemia rubravera
F Refractory anaemia with ringed
sideroblasts
G Refractory anaemia
H 5q-Syndrome
I Multiple myeloma

A 72-year-old man presents with a 1-month history of fever, night sweats and
weight loss. Blood tests reveal a monocyte count of 1400/mm3 in the peripheral
blood and a bone marrow biopsy demonstrates that myeloblasts constitute 16
per cent of his bone marrow

A

C Chronic myelo-monocytic

Chronic myelo-monocytic leukaemia (CMML; C) is a myelodysplastic/
myeloproliferative disease which most commonly affects the elderly
population, defined by a monocytosis of >1000/mm3 and increased
number of monocytes in the bone marrow. Myeloblasts make up

45
Q
A Essential thrombocythaemia
B Myelofibrosis
C Chronic myelo-monocytic
leukaemia
D Refractory anaemia with excess
blasts
E Polycythaemia rubravera
F Refractory anaemia with ringed
sideroblasts
G Refractory anaemia
H 5q-Syndrome
I Multiple myeloma

A 43-year-old woman presents to her general practitioner with headaches, episodes
of dizziness and a strange itching sensation after she comes out of the
bath. On examination a plethoric appearance is noted. Blood tests reveal a haemoglobin
of 19 g/dL and erythropoietin levels are suppressed.

A

E Polycythaemia rubravera

Polycythaemia rubra vera (PRV; E) is characterized by proliferation of
erythroid, granulocytic and megakaryocyte lines. Many PRV cases are due
to a V167F mutation on exon 2 of the JAK2 gene, leading to uncontrolled
stem cell proliferation. Clinical features include hyperviscosity (headaches,
dizziness and stroke), hyper-mast-cell degranulation (pruritis after hot
baths, plethoric skin and peptic ulceration) and increased cell turnover
(gout). Blood tests will reveal a haemoglobin concentration above 18 g/dL,
leukocytosis and thrombocytosis. Erythropoietin levels are low due to a
negative-feedback response from increased erythrocyte production.

46
Q
A Temporal arteritis
B Renal cell carcinoma
C Colorectal cancer
D Rheumatoid arthritis
E Miliary tuberculosis
F Acute pancreatitis
G Schistosomiasis
H Sarcoidosis
I Epstein–Barr infection

A 56-year-old woman visits her GP for a regular check-up for a chronic condition
she suffers from. On examination, she has signs of long-term steroid
therapy. There is ulnar deviation at her metacarpophalangeal joints. Blood
tests reveal a microcytic hypochromic anaemia, low iron and total iron binding
capacity, but a raised ferritin level.

A

D Rheumatoid arthritis

Rheumatoid arthritis (RA; D) is an inflammatory disease that mainly
affects the small joints of the hands but systemic involvement can be
a feature, manifesting in the lungs (fibrosis), heart (pericarditis) and eyes (scleritis). RA is a cause of anaemia of chronic disease (ACD),
which is mediated by IL-6 produced by macrophages. IL-6 induces
hepcidin production by the liver which has the effect of retaining iron
in macrophages (reduced delivery to red blood cells for erythropoiesis)
and decreases export from enterocytes (reduced plasma iron levels).
Laboratory features of ACD include a microcytic hypochromic anaemia,
rouleaux formation and raised ferritin (acute phase protein).

47
Q
A Temporal arteritis
B Renal cell carcinoma
C Colorectal cancer
D Rheumatoid arthritis
E Miliary tuberculosis
F Acute pancreatitis
G Schistosomiasis
H Sarcoidosis
I Epstein–Barr infection

A 45-year-old man presents to accident and emergency with an excruciating
headache. Blood tests show an erythrocyte sedimentation rate of 110 mm/hour.

A

A Temporal arteritis

Temporal arteritis (A) is a vasculitis most commonly affecting the medium
and large arteries of the head. It is also known as giant cell arteritis
due to the inflammatory cells that are visualized on biopsy. Prominent
temporal arteries with regional tenderness, coupled with an erythrocyte
sedimentation rate (ESR) of more than 60 mm/hour is highly suggestive
of temporal arteritis. ESR may be raised due to increase plasma proteins
(fibrinogen, acute phase proteins or immunoglobulin) or due to reduced
packing of red blood cells (anaemia). Other causes of a raised ESR
include myeloma, polymyalgia rheumatica and autoimmune disease.

48
Q
A Temporal arteritis
B Renal cell carcinoma
C Colorectal cancer
D Rheumatoid arthritis
E Miliary tuberculosis
F Acute pancreatitis
G Schistosomiasis
H Sarcoidosis
I Epstein–Barr infection

A 38-year-old man from Nigeria presents to his GP with progressive shortness
of breath, cough and painful rashes on his lower legs. Blood tests reveal a
monocytosis. Chest X-ray demonstrates bihilar lymphadenopathy.

A

H Sarcoidosis

Sarcoidosis (H) is a granulomatous disease characterized by the presence
of non-caseating granulomas in multiple organs, most commonly affecting
the lungs. Diagnosis of sarcoidosis is usually a matter of excluding
other diseases but chest X-ray (bihilar lymphadenopathy), CT scanning
and lung biopsy can all help. Blood tests commonly reveal a monocytosis;
monocytes are contributory to the pathogenesis of granulomatous
disease. Other causes of monocytosis include brucellosis, typhoid, varicella
zoster infection and chronic myelo-monocytic leukaemia (CMML).

49
Q
A Temporal arteritis
B Renal cell carcinoma
C Colorectal cancer
D Rheumatoid arthritis
E Miliary tuberculosis
F Acute pancreatitis
G Schistosomiasis
H Sarcoidosis
I Epstein–Barr infection

A 66-year-old presents to his GP with severe weight loss over 1 month as well
as tiredness. Blood tests reveal an increased erythrocyte, haemoglobin and
erythropoietin count.

A

B Renal cell carcinoma

Renal cell carcinoma (RCC; B) is the most common type of renal cancer.
Secondary polycythaemia may be associated with RCC as a result
of increased erythropoietin (EPO) production. Secondary polycythaemia
can be distinguished from primary polycythaemia as in the former there
is an increase in blood EPO levels, whereas in the latter EPO levels
decrease. Other causes of secondary polycythaemia include chronic
hypoxia (high altitude, smoking, lung disease, cyanotic heart disease),
renal disease (cysts, renal artery stenosis, hydronephrosis) and solid
tumours (renal cell carcinoma and hepatocellular carcinoma).

50
Q
A Temporal arteritis
B Renal cell carcinoma
C Colorectal cancer
D Rheumatoid arthritis
E Miliary tuberculosis
F Acute pancreatitis
G Schistosomiasis
H Sarcoidosis
I Epstein–Barr infection

A 24-year-old man has recently returned from a trip to Kenya. He presents to
his GP with abdominal pain, fever and on examination has hepatosplenomegaly.
Blood tests reveal a marked eosinophilia.

A

G Schistosomiasis

Schistosomiasis (G) is a parasitic disease caused by Schistosoma spp.
It is particularly common in Asia, Africa and South America. The risk
of bladder cancer is increased in urinary forms of schistosomiasis. The
immune response to parasitic infection involves eosinophils and hence
a marked eosinophilia is characteristic. Other causes of eosinophilia
besides parasitic infection include allergic disease (asthma, rheumatoid
arthritis, polyarteritis), neoplasms (Hodgkin’s lymphoma, non-Hodgkin’s
lymphoma) as well as certain drugs (NSAIDs).