Haematology Flashcards

1
Q

what is myeloma?

A
  • a malignant (cancer) disease of differentiated bone marrow plasma cells, accounting for 1% of plasma cells
  • clonal expansion of abnormal, proliferating plasma cells producing a monoclonal paraprotein
  • accumulation of malignant plasma cells in the bone marrow leads to progressive bone marrow failure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

what is the monoclonal paraprotein that is produced in myeloma?

A

mainly IgG (55%) or IgA (20%) and rarely IgM and IgD

  • produced by clonal expansion of abnormal, proliferating plasma cells
  • malignant plasma cells just produce an excess of one type of immunoglobulin (monoclonal paraprotein)
  • may be associated with excretion of light chains in urine (Bence Jones protein), kappa or lambda
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

what are differences in production of paraprotein/light chain production in myeloma?

A
  • production of paraprotein may be associated with excretion of light chains in urine (Bence Jones protein), kappa or lambda
  • in 20% there is no paraproteinaemia, only light chains in urine
  • non secretory myeloma in <5%: no paraprotein or light chains
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

what is the epidemiology of myeloma?

A
  • disease of the elderly
  • median age at presentation is over 60yrs
  • rare under 40yrs
  • annual incidence is 4 per 100000
  • commoner in males and black Africans
  • less common in asians
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

what are clinical features of myeloma?

A
  • bone destruction often causing long bone fractures or vertebral collapse, osteolytic lesions, pathological fractures, spinal cord compression and hypercalcaemia and back pain
  • soft tissue plasmacytomas
  • bone marrow infiltration with plasma cells -> anaemia, neutropenia, thrombocytopenia, paraprotein production which may lead to hyperviscosity, infections and bleeding
  • kidney injury
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

how can myeloma cause kidney injury?

A
  • deposition of light chains in the renal tubules
  • hypercalcaemia
  • hyperuricaemia
  • use of NSAIDs
  • deposition of AL amyloid (rare)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

what types of cytogenetic abnormalities have been found in myeloma?

A

associated with poor survival

  • chromosome 13
  • hypodiploidy (<45 chromosomes)
  • t(4;14), t(14;16) and p53 (17p) deletions

associated with better prognosis

  • t(11:14)
  • hyperdiploidy (>50 chromosomes)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

what is the WHO (2008) classification of plasma cell neoplasms?

A
  • monoclonal gammopathy of undetermined significance
  • plasma cell myeloma
  • solitary plasmacytoma of bone
  • extraosseous plasmacytoma
  • monoclonal immunoglobin deposition diseases
  • heavy chain diseases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

what are types of heavy chain diseases?

A

Gamma heavy chain disease
Mu heavy chain disease
Alpha heavy chain disease

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

what are features of bone disease caused by myeloma?

A
  • dysregulation of bone remodelling -> lytic lesions in spine, skull, long bones, ribs
  • increased osteoclastic activity with no increased osteoblast formation of bone
  • adhesion of stromal cells to myeloma cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

what does adhesion of stromal cells to myeloma cells stimulate?

A

production of RANKL, IL-6 and VEGF

  • RANKL stimulates osteoclast formation and the lytic lesions
  • myeloma cells produce dickkopf-1 (DKK1) which inhibits osteoblast activity and production of new bone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

what is DKK1? what is its action in myeloma?

A

dickkopf-1

  • produced by myeloma cells
  • inhibits osteoblast activity and production of new bone
  • binds to Wnt co-receptor, lipoprotein receptor-related protein 5 (LRP5), inhibiting Wnt signalling and osteoblast differentiation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

what are life-threatening complications of myeloma? how are they treated?

A
  • renal impairment (due to hypercalcaemia) -> urgent attention and long-term dialysis
  • hypercalcaemia treated by rehydration and bisphosphonates e.g. pamidronate
  • spinal cord compression treated by dexamethosome and radiotherapy
  • hyperviscosity due to paraprotein treated by plasmapheresis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

what are general investigations of myeloma and what may be found?

A
  • FBC (Hb, WCC and platelet count are normal/low; shows anaemia, thrombocytopenia and leucopenia)
  • ESR (often high)
  • blood film (may be roleaux formation due to paraprotein and circulating plasma cells)
  • U+E (evidence of kidney infection)
  • serum biochemistry (calcium normal or raised; serum alkaline phosphatase usually normal)
  • serum beta2-microglobulin and albumin used in prognosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

what are immunoglobin investigations of myeloma?

A
  • total protein normal or raised
  • serum protein electrophoresis and immunofixation shows monoclonal band and immune paresis
  • serum free light chain assay (abnormal ratio and increased total amount of light chains)
  • 24hr urine electrophoresis and immunofixation for light chain excretion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

what are radiological investigations of myeloma?

A

skeletal survey

  • lytic lesions, esp. in skull
  • CT, MRI and PET used in plasmacytomas
  • MRI spine useful if back pain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

when can symptomatic myeloma (SMM) be diagnosed?

A

if either of the following are present:

  • significant paraproteinaemia
  • increased bone marrow plasma cells (>10%)

with evidence of end organ failure i.e. hypercalcaemia, renal impairment, anaemia, lytic bone lesions (CRAB)

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

what is AMM? how is it diagnosed?

A

asymptomatic myeloma

  • 10% of cases
  • significant paraprotein (IgG or IgA >30g/dL or urinary light chain excretion >1g/day) and/or marrow plasmacytosis but no end organ damage
  • time to progression ~2-3yrs
  • risk is highest for IgA isotype and light chains in urine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

what is MGUS? how is it diagnosed?

A

monoclonal gammopathy of unknown significance

  • isolated finding of a monoclonal paraprotein in serum that doesn’t fulfil diagnostic criteria for SMM or AMM
  • raised level of paraprotein (usually IgA) found in blood, without other features of myeloma
  • usually in elderly
  • 20-30% develop muliple myeloma over 25yrs
  • low risk: IgG subtype, paraprotein <15g/dL and normal serum free light chain ratio
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

what is plasmacytoma? how is it diagnosed?

A
  • isolated tumour of neoplastic plasma cells
  • no evidence of multiple myeloma
  • may be solitary plasmacytoma outside the marrow cavity typically in upper aerodigestive tract
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

what are features of supportive therapy for myeloma?

A
  • anaemia corrected by blood transfusion and/or erythropoietin
  • hypercalcaemia, kidney injury and hyperviscosity treated
  • infection treated with antibiotics
  • yearly flu vaccinations
  • bone pain helped by radiotherapy and systemic chemotherapy or high-dose dextamethasone, and analgesia
  • hyperviscosity treated by plasmapheresis together with systemic therapy
  • avoid NSAIDs due to risk of renal impairment
  • bisphosponates to reduce progression of bone disease; they inhibit osteoclastic activity
  • pathological fractures prevented by orthopaedic surgery with pinning of lytic bone lesions at critical sites seen on skeletal surgery
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

what are examples of specific therapy for myeloma?

A
  • thalidomide
  • chemotherapy
  • stem cell transport
  • high-dose melphalan autograft
  • allogeneic transplant
  • melphalan and prednisolone
  • bortezomib
  • bisphosphonates e.g. Zolendronate
  • lenalidomide
  • rehydrate and ensure 3L/day to prevent further renal damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

how does myeloma lead to immunoparesis?

A

monoclonal paraprotein production -> other immunoglobin levels are low, leading to increased susceptibility to infections

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

what does OLD CRAB mean, in relation to myeloma?

A

clinical presentation of myeloma
• old age
• calcium elevated
• renal failure - nephrotic syndrome, due to raised immunoglobulins which are proteins which precipitate (especially immunoglobulin light chains) and deposit in organs especially the kidneys - results in thirst due to lack of water retention due to failure
• anaemia - neutropenia or thrombocytopenia resulting in infection, bleeding and fatigue and pallor
• bone lytic lesions - back pain

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

what is seen in blood tests in myeloma?

A
  • normocytic normochromic anaemia
  • raised ESR
  • rouleaux formation on blood film
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

how is U+E used to diagnose myeloma?

A
  • high calcium
  • high alkaline phosphatase
  • Bence-jones protein in urine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

what is seen in plain X-ray in myeloma?

A
• lytic ‘punched-out’ lesions:
- pepper-pot skull
- vertebral collapse
• fractures
• osteoporosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

what types of chemotherapy are used to treat myeloma?

A

CTD: Cyclophosphamide, Thalidomide and Dexamethsasone; max 8 cycles, for less fit people

VAD: Vincristine, Adriamycine and Dexamethasone in fitter people; max 6 cycles

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

what are lymphomas?

A
  • lymphomas are disorders caused by malignant proliferations of B and T lymphocytes
  • these accumulate in the lymph nodes causing lymphadenopathy, may also be found in the peripheral blood or infiltrate organs
  • predominantly in lymph nodes but also blood, bone marrow, liver, spleen, anywhere
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

what can lymphomas be histologically divided into?

A

Hodgkins and Non-Hodgkins lymphoma

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

what characterises Hodgkins lymphoma?

A

have characteristic cells with mirror-image nuclei called Reed-Sternberg cells

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

what characterises non-Hodgkins lymphoma?

A
  • do not have characteristic cells
  • low grade e.g. Follicular Lymphoma
  • high grade e.g. Diffuse Large B Cell Lymphoma
  • very high grade e.g. Burkitt’s Lymphoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

what is the causes of lymphomas?

A

primary immunodeficiency:

  • ataxia telangiectasia
  • Wiscott-Aldrich syndrome
  • common variable immunodeficiency

secondary immunodeficiency:

  • HIV
  • transplant recipients

infection:

  • Epstein-Barr Virus (EBV)
  • Human T-lymphotropic virus
  • Helicobacter pylori

autoimmune disorders e.g. SLE

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

what is the epidemiology of Hodgkins lymphoma?

A
  • male predominance
  • majority of cases occur in teenagers e.g. 13-19 and elderly e.g. over 65
  • 2 peaks of incidence; teenagers and elderly
  • Epstein-Barr Virus (EBV) has a suggested role in pathogenesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

what can Hodgkins lymphoma be further divided into?

A
  • classical Hodgkins lymphoma (cHL)

- nodular lymphocyte predominant Hodgkins lymphoma (NLPHL)

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

what is classical Hodgkins lymphoma characterised by?

A
  • Reed-Sternberg cell with mirror image nuclei

- 90-95% of cases

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

what is nodular lymphocyte predominant Hodgkins lymphoma characterised by?

A

the Reed-Sternberg variant, the popcorn cell

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

what are risk factors for Hodgkins lymphoma?

A
  • affected sibling
  • Epstein-Barr virus (glandular fever) - ‘hide’ in white blood cells and can result in malignancy
  • SLE
  • obese
  • post-transplantation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

what is clinical presentation of Hodgkins lymphoma?

A
  • most commonly there is painless cervical lymphadenopathy, described on examination as ‘rubbery’
  • a smaller proportion of patients (often young women) present with disease localised to the mediastinum with cough due to mediastinal lymphadenopathy
  • generalised disease including hepatosplenomegaly and constitutional B symptoms (weight loss, fever, night sweats)
  • other constitutional symptoms e.g. pruritus, fatigue, anorexia and alcohol-induced pain at the site of the enlarged lymph nodes also occur
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

what are some constitutional B symptoms in Hodgkins lymphoma?

A

weight loss, fever, night sweats

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

what is the emergency presentation of Hodgkins lymphoma?

A
  • infection
  • superior vena cava obstruction with increased jugular venous pressure (JVP)
  • sensation of fullness in the head
  • dyspnoea
  • blackouts
  • facial oedema
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

how is Hodgkins lymphoma diagnosed?

A
  • CT/MRI of chest, abdomen and pelvis for staging (Ann Arbor); may show intrathoracic, abdominal and pelvic nodes
  • lymph node excision or bone marrow biopsy; shows mirror-image nuclei Reed-Sternberg cells or popcorn cells
  • liver biochemistry may be abnormal, with or without liver involvement
  • bloods film and bone marrow:
    • high ESR or Low Hb - indicate worse prognosis
    • high serum lactate dehydrogenase
  • immunophenotyping
  • cytogenetics
  • PET scan
  • molecular techniques (PCR)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

what is used to stage Hodgkins lymphoma?

A

Ann Arbor classification

  • 4 stages
  • each stage is either A, B, X or E
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

what is stage I Hodgkins lymphoma?

A

involvement of a single lymph node region or a single extralymphatic organ or site

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

what is stage II Hodgkins lymphoma?

A

involvement of two or more lymph node regions on the same side of the diaphragm, or localized involvement of an extralymphatic organ or site and of one or more lymph node regions on the same side of the diaphragm

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

what is stage III Hodgkins lymphoma?

A

involvement of lymph node regions on both sides of the diaphragm, which may also be accompanied by involvement of the spleen (IIIS) or by localized involvement of an extralymphatic organ (IIIE) or site or both (IIISE)

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

what is stage IV Hodgkins lymphoma?

A

diffuse or disseminated involvement of one or more extralymphatic organs or tissues, with or without associated lymph node involvement

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

what does A, B, X and E mean in the Ann Arbor classification of Hodgkins lymphoma?

A
  • A: no systemic symptoms other than pruritus (severe itching of skin)
  • B: presence of B symptoms such as fever, weight loss and night sweats
  • X: bulky disease
  • E: involvement of a single extranodal site that is contiguous or proximal to the known nodal site
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

what is the treatment of Hodgkins lymphoma?

A
combination chemotherapy - ABVD:
• A - Adriamycin
• B - Bleomycin
• V - Vinblastine
• D - Dacarbazine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

how is ABVD combination chemotherapy used to treat Hodgkins lymphoma?

A

stages I-A to II-A (with less than 3 areas involved and no bulk):
• short course of ABVD followed by involved field irradiation

stages II-A to IV-B (with more than 3 areas involved):
• longer course of ABVD
• cyclical combination chemotherapy (8 cycles of AVBD) with irradiation at sites of bulk disease
• PET/CT used to detect disease activity after treatment and to distinguish between active tumour (PET-positive) and necrosis or fibrosis (PET-negative) in residual masses
- irradiation, with its attendant complications, can be omitted in PET-negative masses after chemotherapy

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

what are complications of treatment of Hodgkin’s lymphoma?

A

radiotherapy:

  • may increase risk of second malignancies - solid tumours especially in the lung, breast, melanoma, stomach, sarcoma and thyroid
  • increase risk of ischaemic heart disease, hypothyroidism and lung fibrosis due to radiation field

chemotherapy:

  • myelosuppression, nausea, alopecia (hair loss) and infection
  • infertility
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

what is the epidemiology of non-Hodgkin’s lymphoma?

A
  • includes all lymphomas without Reed-Sternberg cells
  • around 80% is of B-cell origin, diffuse large B-cell lymphoma (DLBCL) is commonest
  • around 20% is of T-cell origin
  • generally more varied in terms of presentation, sub-types, treatments and outcomes
  • presentation rare before 40yrs
  • not all centre on nodes
  • there is a strong link with Epstein-Barr virus and Burkitts lymphoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

what are risk factors for non-Hodgkin’s lymphoma?

A
  • nodal disease (75%) e.g. superficial lymphadenopathy
  • extranodal disease (25%): skin (esp. T cell lymphomas and oropharynx, gut, small bowel, bone, CNS and lungs)
  • systemic B symptoms
  • pancytopenia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

what is non-Hodgkin’s lymphoma classified into?

A

low/indolent grade and high grade

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

what are features and an example of low/indolent non-Hodgkin’s lymphoma?

A

example is Follicular Lymphoma

  • slow growing
  • usually advanced at presentation
  • incurable
  • median survival 9-11 years
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

what are features and an example of high grade non-Hodgkin’s lymphoma?

A

example is Diffuse Large B-cell Lymphoma

  • usually has nodal presentation
  • 1/3 cases have extranodal involvement
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

how is non-Hodgkin’s lymphoma diagnosed?

A
  • raised lactate dehydrogenase reflects worse prognosis since its a sign of increased cell turnover and thus cell proliferation
  • blood count: may show anaemia, ESR may be raised, elevated WCC or thrombocytopenia suggests marrow involvement
  • liver biochemistry abnormal if liver involved
  • serum lactate dehydrogenase and beta2-microglobulin are prognostic indicators
  • CXR, CT, PET and gallium scans helpful in staging
  • lymph node excision or bone marrow biopsy; will not see mirror-image nuclei Reed-Sternberg cells or popcorn cells
  • marrow aspiration and trephine biopsy confirms marrow involvement
  • CT/MRI of chest, abdomen and pelvis for staging (Ann Arbor)
  • immunophenotyping
  • cytogenetics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

what is treatment of non-Hodgkin’s lymphoma?

A
R-CHOP regimen:
• R - Rituximab (monoclonal antibody - minimal side effects)
• C - Cyclophosphamide
• H - Hydroxy-daunorubicin
• O - Vincristine (Oncovin brand name)
• P - Prednisolone

with field irradiation for those with bulky disease
60-70% of those with early-stage disease are cured with this regimen

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

what is Rituximab?

A
  • monoclonal antibody
  • targets CD20 expressed on cell surface B cells
  • chimeric mouse/human protein
  • minimal side effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

how is low grade non-Hodgkin’s lymphoma treated?

A
  • none may be required

* radiotherapy may be curative in localised disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

what is treatment for high grade non-Hodgkin’s lymphoma?

A

early:
- 3 months R-CHOP regimen with radiotherapy

late:
- 6 months R-CHOP regimen with radiotherapy

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

what is Burkitt’s lymphoma? what is the treatment?

A
  • endemic, sporadic and AIDS related types
  • occurs mainly in African children
  • associated with EBV infection
  • usually B cells with jaw lymphadenopathy in children, usually with gastrointestinal involvement
  • treatment with cyclical combination chemotherapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

what are the four main subtypes of leukaemia?

A
  • acute lymphoblastic leukaemia (ALL)
  • acute myeloid leukaemia (AML)
  • chronic myeloid leukaemia (CML)
  • chronic lymphocytic leukaemia (CLL)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

what is leukaemia?

A
  • malignant neoplasms of haemopoietic stem cells (can be precursors of RBCs, platelets or white cells) in the bone marrow that are non functional
  • proliferate rapidly
  • characterised by diffuse replacement of bone marrow by neoplastic cells
  • leukaemic cells usually spill over into the blood, where they may be seen in large numbers
  • may also infiltrate the liver, spleen, lymph nodes and other tissues
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

what is the basic pathophysiology of leukaemia?

A
  • firstly, the leukaemia cells are dividing rapidly but serve no function and are wasting energy making useless cells and there is less energy available to make useful functional cells
  • secondly, due to rapid replication these cells take up a lot of space within the bone marrow meaning there is little space and food for other cells to grow
    • the bone marrow is unable to make as many normal functioning cells resulting in non-functional cells in the blood and symptoms of leukaemia
    • eventually when there is no longer space in the bone marrow, leukaemia cells will be present in the blood too
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

what is the epidemiology of acute lymphoblastic leukaemia?

A
  • most common between 2 and 4 years of age
  • the commonest cancer in childhood
  • thought to develop from a combination of genetic susceptibility and an environmental trigger
  • ionising radiation e.g. x-rays during pregnancy and Down’s syndrome (trisomy 21) are important associations
  • CNS involvement is common
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

what is acute lymphoblastic leukaemia? what happens in it?

A
  • malignancy of immature lymphoid cells (give rise to T cells and B cells)
  • affects B or T lymphocyte cell lines, arrests the maturation and promotes uncontrolled proliferation of immature blast cells (immature precursor of
    myeloid cells (myeloblasts) or lymphoid cells (lymphoblast))
  • majority of cases derive from B-cell precursors
  • there is increased proliferation of immature lymphoblast cells (B or T cell precursors) in the bone marrow:
    • if all B cells = children
    • if all T cells = adults
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

what are clinical presentations of acute lymphoblastic leukaemia?

A
  • marrow failure
  • organ infiltration
  • bone marrow infiltration resulting in bone pain and anaemia, bleeding and infection
  • liver/spleen infiltration resulting in hepatosplenomegaly
  • node infiltration resulting in lymphadenopathy
  • CNS infiltration resulting headache and cranial nerve palsies
  • mediastinum infiltration resulting in mediastinal masses with superior vena cava obstruction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

what are features of marrow failure in acute lymphoblastic leukaemia?

A

anaemia - low Hb:

  • resulting in breathlessness, fatigue, angina and claudication
  • there is pallor and cardiac flow murmur

infection - low WCC:

  • resulting in infections
  • there is fever and mouth ulcers

bleeding - low platelets:
- resulting in bleeding and bruising

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

how is acute lymphoblastic leukaemia diagnosed?

A
  • FBC and blood film:
    • WCC is usually high
    • blast cells on film and in bone marrow
  • CXR and CT scan to look for mediastinal and abdominal lymphadenopathy
  • lumbar puncture to look for CNS involvement
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

what is the treatment of acute lymphoblastic leukaemia?

A
  • blood and platelet transfusions
  • neutropenia may lead to deadly infections; treat with prophylactic antivirals, antibacterials and antifungals
  • allopurinol (prevents tumour lysis syndrome)
  • IV fluids - insert Hickman line (permanent cannula into main vessel, tunnelled under sub-cutaneous fat so harder for infection to arise) so can easily take blood for testing and administer drugs and fluids
  • remission induction with combination chemotherapy including vincristine, dexamethasone, asparaginase and daunorubicin
  • involves CNS, so prophylaxis of intrathecal drugs, methotrexate or cytosine arabinoside and occasionally cranial irradiation is used
  • marrow transplantation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

what is acute myeloid leukaemia?

A

the neoplastic proliferation of blast cells derived from marrow myeloid (gives rise to basophils, neutrophils and eosinophils) elements

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

what is the epidemiology of acute myeloid leukaemia?

A
  • progresses rapidly with death in 2 months if untreated
  • affects mostly children
  • the commonest acute leukaemia of adults
  • associated with radiation and syndromes such as Down’s (trisomy 21)
  • complete remission is usually achieved in about 80% of patients under 60 years with no comorbidity, in whom treatment is offered with curative intent
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

what is clinical presentation of acute myeloid leukaemia?

A
  • marrow failure
  • hepatomegaly and splenomegaly occur due to infiltration
  • gum hypertrophy
  • DIC occurs in a subtype of AML where there is release of thromboplastin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

how is acute myeloid leukaemia diagnosed?

A
  • WCC is often raised, but can be normal or even low
  • may be few blast cells in the peripheral blood so diagnosis depends on bone marrow biopsy
  • differentiation from acute lymphocytic leukaemia is based on microscopy, immunophenotyping and
    molecular methods
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

what are complications of acute myeloid leukaemia?

A
  • infection is a major issue; be alert to septicaemia

- causes common organisms to present oddly, with few antibodies being made

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

what is treatment of acute myeloid leukaemia?

A
  • blood and platelet transfusions
  • neutropenia may lead to deadly infections; treat with prophylactic antivirals, antibacterial and antifungals
  • allopurinol (prevents tumour lysis syndrome)
  • IV fluids - insert Hickman line (permanent cannula into main vessel, tunnelled under sub-cutaneous fat so harder for infection to arise) so can easily take blood for testing and administer drugs and fluids
  • chemotherapy
  • marrow transplantation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

what is the epidemiology of chronic myeloid leukaemia?

A
  • most exclusively a disease of adults
  • uncontrolled clonal proliferation of myeloid cells
  • occurs most often between 40-60 yrs
  • slight male predominance
  • rare in childhood
  • insidious onset
  • more than 80% have the Philadelphia chromosome which forms a fusion gene BCR/ABL on chromosome 22, which has tyrosine kinase activity: stimulates cell division
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

what is the clinical presentation of chronic myeloid leukaemia?

A
  • chronic phase lasts 3-4 years, followed by blast transformation with development of acute leukaemia and rapid death
  • may also transform into myelofibrosis, with death from bone marrow failure
  • symptomatic anaemia e.g. shortness of breath
  • abdominal discomfort due to splenomegaly
  • weight loss
  • tiredness
  • pallor
  • fever and sweats in the absence of infection
  • may be features of gout due to purine breakdown
  • may be bleeding due to platelet dysfunction
  • 40-60yrs
  • metabolic features
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

what is the action of the Philadelphia chromosome in leukaemia?

A
  • esp. CML
  • abnormality in chromosome 22
  • defective and unusually short due to reciprocal translocation of genetic material between chromosome 9 and 22
  • contains fusion gene called BCR-ABL1
  • codes for a hybrid 210-kDa fusion protein: a tyrosine kinase signalling protein that is always on, causing the cell to divide uncontrollably by interrupting stability of the genome and impairing signalling pathways
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

how is chronic myeloid leukaemia diagnosed?

A

blood count:
• very high WCC, often > 100 × 109/L; with whole spectrum of myeloid cells i.e. increased neutrophils, myelocytes, basophils and eosinophils
• low Hb (normochromic and normocytic anaemia)
• platelets are low, normal or raised

bone marrow aspirate:
- hypercellular (increased myeloid progenitors)

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

what is the treatment of chronic myeloid leukaemia?

A
  • oral Imatinib (Glivec); specific BCR/ABL1 tyrosine kinase inhibitor; produces a complete haematological response in over 95% of patients
  • Nilotinib, Dasatinib, Ponatinib (tyrosine kinase inhibitors)
  • stem cell transplant
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

what is the epidemiology of chronic lymphocytic leukaemia? what is it?

A
  • the most common leukaemia
  • often incidental finding on FBC
  • occurs predominantly in later life
  • it is the uncontrolled proliferation and accumulation of mature B cells that have escaped programmed cell death and undergone cell-cycle arrest
  • mutations, trisomies and deletions influence risk
  • pneumonia may be a triggering event
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

what is the most common leukaemia?

A

chronic lymphocytic leukaemia

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

what is the clinical presentation of chronic lymphocytic leukaemia?

A
  • often no symptoms, presenting as a surprise on a routine FBC (done pre-op)
  • may be anaemic (due to haemolysis or marrow infiltration) or infection prone
  • if severe then weight loss, sweats and anorexia
  • hepatosplenomegaly
  • enlarged, rubbery, non-tender nodes
  • indolent corse
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

how is chronic lymphocytic leukaemia diagnosed with blood count and blood film?

A

blood count
• normal or low Hb
• raised WCC with lymphocytosis (> 5 × 109/L)
• may be anaemia and thrombocytopenia

blood film
• smudge cells may be seen in vitro; these are small lymphocytes of mature appearance with are an artefactual finding due to cell rupture while the film is being made

bone marrow
• reflects peripheral blood usually heavily infiltrated with lymphocytes

immunophenotyping and cytogenetics

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

what are the complications of chronic lymphocytic leukaemia?

A
  • autoimmune haemolysis
  • increased infection risk due hypogammaglobulinaemia (low IgG); bacterial and viral especially herpes zoster
  • marrow failure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

what is the progression of chronic lymphocytic leukaemia?

A
  • many stay stable for years and may even regress
  • death is often due to complication of infection
  • may transform into aggressive lymphoma = Richter’s syndrome
  • the median survival from diagnosis is very variable and correlates closely with disease stage at diagnosis and cytogenetic findings, e.g. patients with either 11q or 17p deletions (the sites of two tumour suppressor genes) are at high risk of not responding to initial treatment and rapid progression. In other patients there is near-normal life expectancy.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

what is the treatment of chronic lymphocytic leukaemia?

A
  • blood transfusions
  • combination therapy with fludarabine, cyclophosphamide and rituximab is standard first line therapy
  • chlorambucil usually reduces lymphocytosis, lymphadenopathy and splenomegaly in older patients
  • alemtuxumab used in patients where there is disease progression after treatment with Fludarabine
  • human IV immunoglobulins
  • chemotherapy or radiotherapy
  • monoclonal antibodies (e.g. anti-CD20: rituximab)
  • try stem cell/bone marrow transplant
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

what is the rule of 3s of prognosis for chronic lymphocytic leukaemia?

A
  • 1/3 will never progress
  • 1/3 progress slowly
  • 1/3 progress actively
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

what are late effects of lymphoma?

A
  • infertility
  • anthracyclines; cardiomyopathy
  • bleomycin; lung damage
  • vinca alkaloids; peripheral neuropathy
  • second cancers
  • psychological cancers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

what is treatment of indolent non-Hodgkin’s lymphoma?

A
  • alkylating agents
  • combination chemotherapy
  • purine analogues
  • monoclonal antibodies
  • radio-immunoconjugates
  • new oral targeted agents
  • radiotherapy
  • bone marrow transplant
  • do nothing
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

what haemopoietic cells does each type of leukaemia affect?

A

acute myeloid leukaemia = myeloblast
chronic myeloid leukaemia = basophil, neutrophil, eosinophil
acute lymphoblastic leukaemia = lymphoblast
chronic lymphocytic leukaemia = B lymphocyte

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

what can a multipotential haematopoietic stem cell differentiate into?

A
  • common myeloid progenitor

- common lymphoid progenitor

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

what can a common myeloid progenitor differentiate into?

A
  • megakaryocyte
  • erythrocyte
  • mast cell
  • myeloblast
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

what can a megakaryocyte differentiate into?

A

platelets

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

what can myeloblasts differentiate into?

A
  • basophil
  • neutrophil
  • eosinophil
  • monocyte
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

what can a monocyte differentiate into?

A

macrophage (in tissues)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

what can common lymphoid progenitors differentiate into?

A
  • natural killer cell (large granular lymphocyte)

- small lymphocyte

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

what can small lymphocytes differentiate into?

A
  • T lymphocyte

- B lymphocyte

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

what can B lymphocytes differentiate into?

A

plasma cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

what increases risk of acute myeloid leukaemia?

A
  • preceeding haematological disorders
  • prior chemotherapy
  • exposure to ionising radiation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

what is supportive care for acute myeloid leukaemia?

A
  • HML
  • blood product support
  • prompt treatment of infectiosn
  • recognition of atypical/unusual infections
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

what abnormalities are present in a good risk category for acute myeloid leukaemia?

A

t(8;21), t(15;17), inv(16)

- t(15;17) = acute promyelocytic leukaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

what abnormalities are present in intermediate risk category for acute myeloid leukaemia?

A

normal, +8, +21, +22, del(7q), del(9q), abnormal 11q23, all other structural or numerical changes

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

what abnormalities are present in poor risk category for acute myeloid leukaemia?

A

-5, -7, del(5q), abnormal 3q, complex cytogenetics

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

what are features of acute promyelocytic leukaemia?

A
  • 5-8% of AML in adults
  • t(15;17) – PML/RARA gene
  • blocks differentiation of promyelocyte to mature granulocyte
  • haematological emergency – DIC
  • 90% remission rate
  • targeted treatment = ATRA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

what are the outcomes of acute lymphoblastic leukaemia?

A
  • children majority cured
  • TYA (16-25yrs) most cured with Paediatric style treatment
  • adults up to ½ cured
  • depends critically on age and cytogenetics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

what is the clinical staging of CLL?

A

Binet Stage

Stage A: lymphocytosis (<3 nodal areas): survival 10-15yrs

Stage B: nodes (3+ nodal areas): survival 5-7yrs

Stage C: anaemia and/or thrombocytopenia: survival 2-3yrs

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

what is autologous stem cell transplantation? what are advantages/disadvantages?

A

own

  • enables escalation of chemo with stem cell rescue
  • relatively straightforward
  • mortality 2%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

what is allogenic stem cell transplantation? what are advantages/disadvantages?

A
  • much more toxic
  • mortality 15-30%
  • stem cells attack residual tumour (GVL)
  • stem cells also attack recipient (GVHD)
  • used in acute and chronic leukaemias
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

what is the definition of anaemia?

A

present when there is a decrease of haemoglobin in the blood below the reference level for the age and sex of the individual

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

what is the lifespan of erythrocytes?

A

120 days

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

what can cause anaemia?

A
  • may be due either to a low red cell mass (RCM) or increased plasma volume (e.g. in pregnancy where there is small increase in red cell mass as well as high
    plasma volume so concentration is decreased thus look anaemic)
  • may be due to reduced production from bone marrow or increased loss of RBC’s i.e. by the spleen, liver, bone marrow and blood loss and has many causes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

what can cause reduced production from bone marrow or increased loss of RBCs?

A
  • the test to determine if bone marrow production is the issue is to look at the reticulocyte count which is a count of immature RBC’s in the bone marrow
  • if production is the issue then the reticulocyte count will be low
  • if removal is the issue then the reticulocyte count will be high
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

what causes a falsely high haemoglobin? where can this be seen?

A

reduction in plasma volume

- seen in dehydration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

what is MCV? how is it calculated?

A
  • a measure of the average volume of a red blood corpuscle (or red blood cell)
  • the measure is attained by multiplying a volume of blood by the proportion of blood that is cellular (the hematocrit), and dividing that product by the number of erythrocytes (red blood cells) in that volume
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

what are indices in anaemia?

A
  • low MCV <80fL
  • high MCV >96fL
  • normal MCV
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

what are causes of microcytic anaemia?

A
  • iron deficiency
  • Thalassaemia
  • anaemia of chronic disease
  • sideroblastic anaemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

what can the appearance of bone marrow be with macrocytic erythrocytes?

A
  • megaloblastic

- normoblastic

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

what can be diagnosed with macrocytic erythrocytes with megaloblastic appearance of bone marrow?

A

vitamin B12 or folate deficiency

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

what can be diagnosed with macrocytic erythrocytes, with normoblastic appearance of bone marrow?

A
  • alcohol
  • increased reticulocytes (e.g. haemolysis or haemorrhage)
  • liver disease
  • hypothyroidism
  • drug therapy e.g. azathioprine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

what are causes of normocytic anaemia?

A
  • acute blood loss
  • anaemia of chronic disease
  • CKD
  • combined deficiency, e.g. iron and folate
  • autoimmune rheumatic disease
  • marrow infiltration/fibrosis
  • endocrine disease
  • haemolytic anaemias
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

what are consequences of anaemia?

A
  • reduced O2 transport
  • tissue hypoxia
  • compensatory changes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

what are compensatory changes that occur due to anaemia?

A
  • increased tissue perfusion
  • increased O2 transfer to tissues
  • increased RBC production
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

what are pathological consequences of anaemia?

A
  • myocardial fatty change
  • fatty change in liver
  • aggravates angina and claudication
  • skin and nail atrophic changes
  • CNS cell death (cortex and basal ganglia)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

what are non-specific symptoms of anaemia?

A
  • fatigue, headaches and faintness
  • dyspnoea and breathlessness
  • angina if there is pre-existing coronary disease
  • anorexia
  • intermittent claudication
  • palpitations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

what are signs of anaemia?

A
  • may be absent even in severe anaemia - this is because a very slowly falling level of Hb allows for haemodynamic compensation and enhancement of oxygen-carrying capacity of the blood
  • pallor - skin and mucous membranes are pale
  • tachycardia
  • systolic flow murmur
  • cardiac failure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

what is the most common cause worldwide of microcytic anaemia?

A

iron deficiency anaemia

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

what is the average daily intake of iron? how much is absorbed?

A

average daily intake of iron is 15-20mg, although normally only 10% is absorbed in the duodenum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

how is iron absorbed?

A
  • actively transported into the duodenal intestinal epithelial cells by the intestinal haem transporter (HCP1) which is highly expressed in the duodenum
  • some is incorporated into ferritin (protein-iron complex) that acts as an intracellular store for iron
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

what happens to absorbed iron that does not bind to ferritin?

A

released into the blood where it is able to circulate around the body bound to the plasma protein transferrin

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

what is the role of transferrin in iron transport?

A
  • is a plasma protein which binds to absorbed iron that doesn’t bind to ferritin
  • allows iron to circulate around the body in the blood
  • transports iron in the blood plasma to the bone marrow to be incorporated into new erythrocytes
  • synthesised in the liver
  • normally about 1/3 saturated with iron
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

what happens to the majority of iron?

A

incorporated into haemoglobin in developing erythroid precursors and mature red cells

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

what happens to iron that is not incorporated into haemoglobin? where is it stored?

A

the rest is stored in reticuloendothelial cells, hepatocytes and skeletal muscle cells either as ferritin (majority; more easily mobilised than haemosiderin for Hb formation, found in small amounts in plasma and in
most cells especially liver, spleen and bone marrow) or haemosiderin in hepatocytes, skeletal muscle and reticuloendothelial macrophages

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

what is the epidemiology of iron deficiency anaemia?

A
  • most common cause of anaemia worldwide
  • seen in 14% of menstruating women
  • develops when there is inadequate iron for haemoglobin synthesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

what are the causes of iron deficiency anaemia?

A
  • blood loss
  • poor intake; rare in developed countries
  • increased demands e.g. in growth and pregnancy
  • malabsorption/decreased absorption, e.g. small bowel disease or post-gastrectomy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

what can cause blood loss that leads to iron deficiency anaemia?

A
  • menorrhagia (severe menstruation with heavy blood loss)
  • pregnancy and breast feeding
  • GI bleeding
  • hookworm - the leading cause of iron deficiency worldwide resulting in intestinal blood loss and iron deficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

what are risk factors for iron deficiency anaemia?

A
  • undeveloped countries
  • high vegetable diet
  • premature infants
  • introduction of mixed feeding delayed, since breast milk contains low iron
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

what is the pathophysiology of iron deficiency anaemia?

A

less iron is available for haemoglobin synthesis; this is crucial for haemoglobin production, thus reduction in iron will result in a decrease in haemoglobin and smaller RBC’s resulting in microcytic anaemia

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

what is the clinical presentation of iron deficiency anaemia?

A
  • fatigue, headaches and faintness
  • dyspnoea and breathlessness
  • angina if there is pre-existing coronary disease
  • anorexia
  • intermittent claudication
  • palpitations
  • may be absent even in severe anaemia
  • pallor
  • tachycardia
  • systolic flow murmur
  • cardiac failure
  • brittle nails and hair
  • spoon-shaped nails: koilonychia
  • atrophy of the papillae of the tongue: atrophic glossitis
  • angular stomatitis/cheilosis: ulceration of the corners of the mouth
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

what are differential diagnoses of iron deficiency anaemia?

A
  • thalassaemia
  • sideroblastic anaemia
  • anaemia of chronic disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

what is used to diagnose iron deficiency anaemia?

A
  • blood count and film
  • serum ferritin
  • serum iron
  • serum soluble transferrin receptors
  • further investigations into cause of blood loss e.g. GI tract examination
  • low reticulocyte count
  • bone marrow examination is generally unnecessary
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

what is seen on a blood count and film in iron deficiency anaemia?

A
  • RBCs are microcytic (MCV <80fL) and hypochromic (MCH <27pg) (pale)
  • there is poikilocytosis (variation in RBC shape) and anisocytosis (variation in RBC size)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

what is seen on serum ferritin in iron deficiency anaemia?

A
  • the level of serum ferritin reflects the amount of stored iron
  • in simple iron deficiency this is low - confirms diagnosis
  • however may be normal in malignancy or infection (since it rises in these events i.e. inflammation)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

what is seen on serum iron in iron deficiency anaemia?

A
  • serum iron is low
  • total iron-binding capacity (TIBC) rises compared to normal, iron deficiency is present with the transferrin saturation (serum iron divided by TBIC) falls below 19%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

what is treatment of iron deficiency anaemia?

A
  • find and treat underlying cause
  • oral iron e.g. ferrous sulphate or ferrous gluconate
  • parenteral iron e.g IV iron or deep intramuscular iron in extreme cases e.g. severe malabsorption or when patients are intolerant
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

what are examples of oral and parenteral iron given to treat iron deficiency anaemia?

A
  • oral iron e.g. ferrous sulphate or ferrous gluconate

- parenteral iron e.g. IV iron or deep intramuscular iron

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

what are side effects of oral iron? what can be done about this?

A
  • nausea, abdominal discomfort, diarrhoea/constipation, black stools
  • can give ferrous gluconate if side effects are bad
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

what is anaemia in chronic disease? what are the red cells like?

A
  • anaemia that is secondary to a chronic disease; if the body is sick then the bone marrow will be too, resulting in anaemia
  • occurs in patients with chronic inflammatory disease (e.g. Crohn’s and RA), chronic infections (e.g. TB), malignancy and CKD
  • RBC’s are often normocytic but they can be microcytic, especially in rheumatoid arthritis and Crohn’s disease, or can be normochromic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

what is the epidemiology of anaemia of chronic disease?

A
  • the second most common anaemia
  • the commonest anaemia in hospital patients
  • occurs in individuals with chronic infections such as:
    • tuberculosis
    • Crohn’s
    • rheumatoid arthritis
    • SLE
    • malignant disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

what are risk factors for anaemia of chronic disease?

A
have a chronic disease, e.g.
• tuberculosis
• Crohn’s
• rheumatoid arthritis
• SLE
• malignant disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

what is the pathophysiology of anaemia of chronic disease?

A
  • there is decreased release of iron from the bone marrow to developing erythroblasts (early RBC, before reticulocyte)
  • an inadequate erythropoietin response (cytokine which increases RBC production) to anaemia
  • high levels of hepcidin expression (binds to export transport protein, ferroportin, in the iron-absorbing cells in the duodenum, causing its degradation, with consequent reduction in trasnport of iron from duodenal cells into the plasma)
  • decreased RBC survival
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

what is the clinical presentation of anaemia of chronic disease?

A
  • fatigue, headaches and faintness
  • dyspnoea and breathlessness
  • angina if there is pre-existing coronary disease
  • anorexia
  • intermittent claudication
  • palpitations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

how is anaemia of chronic disease diagnosed?

A
  • serum iron and total iron-binding capacity (TIBC) are low
  • serum ferritin is normal or raised due to the inflammatory process
  • serum soluble transferrin receptor level is normal
  • blood count and film:
    • RBC’s are normocytic or microcytic and hypochromic as in rheumatoid arthritis and Crohn’s
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

what is seen on blood count and film in anaemia of chronic disease?

A

RBC’s are normocytic or microcytic and hypochromic as in rheumatoid arthritis and Crohn’s

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

what is treatment of anaemia of chronic disease?

A
  • treat underlying chronic cause
  • erythropoietin is effective in raising the haemoglobin level and is used in anaemia of renal disease and inflammatory disease e.g. rheumatoid arthritis and inflammatory bowel disease
    • side effects; flu-like symptoms, hypertension, mild rise in the platelet count and thromboembolism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

what are side effects of erythropoietin used in anaemia of chronic disease?

A

flu-like symptoms, hypertension, mild rise in the platelet count and thromboembolism

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

what is the clinical presentation of normocytic anaemia?

A
  • fatigue, headaches and faintness
  • dyspnoea and breathlessness
  • angina if there is pre-existing coronary disease
  • anorexia
  • intermittent claudication
  • palpitations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

how is normocytic anaemia diagnosed?

A
  • normal B12 and folate
  • raised reticulocytes
  • Hb down
  • blood count and film: RBCs are normocytic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

what is the treatment of normocytic anaemia?

A
  • treat underlying cause
  • improve diet with plenty of vitamins
  • erythropoietin injections
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

what are megaloblasts?

A
  • erythroblasts with delayed nuclear maturation because of delayed DNA synthesis
  • they are large (i.e. high MCV) and have no nuclei
  • may also affect the white cells (causing hypersegmented neutrophil nuclei with six lobes, and sometimes leucopenia) and platelets (causing thrombocytopenia)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

what is pernicious anaemia?

A
  • B12 deficiency
  • type of megaloblastic anaemia
  • pernicious anaemia is an autoimmune disorder where the parietal cells of the stomach are attacked resulting in atrophic gastritis and the loss of intrinsic factor production and thus vitamin B12 malabsorption
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

where is vitamin B12 found?

A

meat, fish and dairy products but not plants

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

how long do body stores of vitamin B12 last?

A

4 years

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

how is vitamin B12 absorbed?

A
  • B12 is absorbed by binding to intrinsic factor produced by the parietal cells of the stomach then being absorbed in the terminal ileum of the small intestines by carrier protein transcobalamin II
  • B12 is liberated from protein complexes in food by gastric acid and pepsin
  • binds to vitamin B12-binding protein (R binder) derived from saliva
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

what is vitamin B12 used for in the body?

A
  • B12 is essential for thymidine and thus DNA synthesis
  • thus in B12 deficiency there is an impairment of DNA synthesis resulting in delayed nuclear maturation resulting in large RBCs as well as decreased RBC production in the bone marrow
  • this DNA impairment will affect all cells, but bone marrow is most affected since its the most active in terms of cell division
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

what are some causes of vitamin B12 deficiency? what is the most common cause?

A
  • dietary (vegans)
  • malabsorption (lack of intrinsic factor or terminal ileum removed/ileal resection)
  • pernicious anaemia (most common cause)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

what is the epidemiology of pernicious anaemia?

A
  • the disease is common in the elderly (over 60)
  • seen in all races but more common in fair-haired, blue eyed individuals and those who have blood group A
  • more common in females than males
  • there is also an association with other autoimmune disease e.g. thyroid disease, Addison’s disease and vitiligo
  • many cases are undiagnosed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

what are risk factors for pernicious anaemia?

A
  • elderly
  • female
  • fair-haired, blue eyes
  • blood group A
  • thyroid and Addison’s disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

what is the pathophysiology of pernicious anaemia?

A
  • parietal cell antibodies are present in the serum in 90% of patients with pernicious anaemia, and also in 10% of normal individuals
  • intrinsic factor antibodies, although found in only 50% of patients with pernicious anaemia, are specific for diagnosis
  • atrophic gastritis (plasma and lymphoid cell infiltration in the fundus)
  • the parietal and chief cells are replaced by mucin-secreting cells
  • there is achlorhydria (reduced HCL acid production) and the absent secretion of intrinsic factor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

what is achlorhydria?

A

reduced HCL acid production

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

what is clinical presentation of pernicious anaemia?

A
  • onset is insidious with progressively increasing symptoms of anaemia e.g. fatigue, headache, pallor, dyspnoea, anorexia, tachycardia and palpitations
  • may have a lemon-yellow skin colour due to the combination of pallor and mild jaundice caused by excess breakdown of haemoglobin (due to fact that body will try to remove defective large RBCs)
  • red sore tongue (glossitis) and angular stomatitis/cheilosis (ulceration of the corners of the mouth) may be present
  • neurological features can occur with very low levels of serum B12 and include a polyneuropathy caused by symmetrical damage to the peripheral nerves and posterior and lateral columns of the spinal cord (subacute combined degeneration of the cord).
  • the latter presents with progressive weakness, ataxia and eventually paraplegia if untreated.
  • dementia and visual disturbances due to optic atrophy may also occur.
  • there is a higher incidence of gastric carcinoma with pernicious anaemia than in the general population.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

why may patients with pernicious anaemia have a lemon-yellow skin colour?

A

due to the combination of pallor and mild jaundice caused by excess breakdown of haemoglobin (due to fact that body will try to remove defective large RBCs)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

what are neurological features seen with pernicious anaemia?

A
  • only occur with very low levels of B12
  • symmetrical paresthesia (burning or prickling pain, tingling) in fingers and toes
  • early loss of vibration sense and proprioception
  • progressive weakness and ataxia
  • paraplegia may result
  • dementia, psychiatric problems, hallucinations, delusions and optic atrophy may occur from vitamin B12 deficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

what are differential diagnoses of pernicious anaemia?

A
  • must differentiate from the other cause of megaloblastic anaemia - folate deficiency
  • differentiate from other causes of B12 deficiency
  • any disease in the terminal ileum or bacterial overgrowth in the small bowel can result in B12 deficiency
  • gastrectomy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

how is pernicious anaemia diagnosed?

A
  • blood count and film
  • serum bilirubin may be raised as a result of ineffective erythropoiesis resulting in increased RBC breakdown
  • serum B12 is low, frequently <50ng/L
  • red cell folate may be reduced because vitamin B12 is needed to convert serum folate to the active intracellular form
  • Hb is low
  • reticulocyte count is low
  • intrinsic factor antibodies are diagnostic but lower sensitivity i.e. not present in all patients
  • small bowel barium follow-through and distal duodenal biopsies may be needed
  • bone marrow examination shows hypercellular bone marrow with megaloblastic changes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

what is seen on a blood count and film in pernicious anaemia?

A
  • typical of megaloblastic anaemia
  • RBCs are macrocytic (MCV often >110fL)
  • peripheral film shows oval macrocytes (large RBC’s) with hypersegmented neutrophil polymorphs with six or more lobes in the nucleus
  • in severe cases, leucopenia and thrombocytopenia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

what is the treatment of pernicious anaemia?

A
  • if not pernicious anaemia then treat cause
  • if a low B12 is due to malabsorption then injections are required
  • if cause is dietary then give oral B12 2mg per day
  • replenish B12 stores by giving IM hydroxocobalamin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

where is folate found?

A

green vegetables e.g. spinach and broccoli, nuts, yeast and offal e.g. liver and kidney

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

in which patients may folate deficiency develop rapidly?

A

both poor intake and excess utilisation of folate e.g. in ICU

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

what is folate absorbed by?

A

duodenum/proximal jejunum (upper small intestine)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

what is folate used for by the body?

A
  • folate is essential for DNA synthesis
  • thus in folate deficiency there is an impairment of DNA synthesis resulting in delayed nuclear maturation and in large RBCs as well as decreased RBC production in the bone marrow
  • this DNA impairment will affect all cells, but bone marrow is most affected since it’s the most active in terms of cell division
  • folate is also essential for fetal development; deficiency can result in neural tube defects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

what are causes of folate deficiency?`

A
  • main cause is poor intake e.g. poverty, alcoholics and elderly
  • increased demand/utilisation e.g. pregnancy or increased cell turnover i.e. haemolysis, malignancy, inflammatory disease and renal dialysis
  • malabsorption e.g. coeliac disease or Crohn’s disease
  • antifolate drugs e.g. methotrexate and trimethoprim
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

what are examples of some antifolate drugs?

A

methotrexate and trimethoprim

186
Q

what are risk factors for folate deficiency?

A
  • elderly
  • poverty
  • alcoholic
  • pregnant
  • Crohn’s or coeliac disease
187
Q

what is clinical presentation of folate deficiency?

A
  • patients may be asymptomatic
  • may present with symptoms of anaemia e.g. pallor, fatigue, dyspnoea, anorexia and headache
  • glossitis (red sore tongue) can occur
  • no neuropathy/neurological defects unlike B12 deficiency
188
Q

how can you differentiate between B12 and folate deficiency?

A

in folate deficiency there are no neuropathy/neurological problems, and in B12 deficiency there are

189
Q

how is folate deficiency diagnosed?

A
  • blood count and film
  • serum and red cell folate is low; red cell is more accurate guide to tissue folate than serum folate, which is usually low
  • GI investigation e.g. small bowel biopsy to exclude occult GI disease
  • serum bilirubin may be raised due to ineffective erythropoiesis resulting in increased RBC breakdown
  • if the history does not suggest dietary deficiency as the cause, further investigations such as endoscopic small bowel biopsy should be performed to look for small bowel disease
190
Q

what is seen on a blood count and film in folate deficiency?

A
  • typical of megaloblastic anaemia
  • RBC’s are macrocytic
  • peripheral film shows oval macrocytes (large RBCs) with hypersegmented neutrophil polymorphs with six or more lobes in the nucleus
191
Q

what is the treatment of folate deficiency?

A
  • treat underlying cause
  • give oral folic acid 5mg daily for 4 months; never without B12 as well (unless the patient is known to have normal B12) since in low B12 states it may precipitate/worsen subacute combined degeneration of the spinal cord
  • higher daily doses may be needed with malabsorption
  • prophylactic folic acid given to patients with chronic haemolysis (5mg weekly)
192
Q

what is haemolytic anaemia?

A

the premature breakdown of RBCs, before their normal lifespan of around 120 days

193
Q

where does haemolysis of RBCs occur?

A

within the circulation (intravascular):
• when RBCs are rapidly destroyed in circulation, haemoglobin is liberated
• this is initially bound to haptoglobulin but these soon become saturated
• excess free plasma haemoglobin is filtered by the renal glomerulus and enters the urine, although small amounts are reabsorbed by the renal tubules
• in renal tubular cells, haemoglobin is broken down and becomes deposited in the cells as haemosiderin

the reticuloendothelial system i.e. by macrophages of the liver, spleen (in particular) and bone marrow (extravascular)

194
Q

what does free haemoglobin bind to?

A

haptoglobulin (soon becomes saturated)

195
Q

what happens to haemoglobin in renal tubular cells?

A

broken down and becomes deposited in the cells as haemosiderin

196
Q

what are consequences of haemolysis?

A
  • shortening of red cell survival does not always result in anaemia, as there is a compensatory increase in red cell production by the bone marrow
  • if the red cell loss can be contained within the marrow’s capacity for increased output, then a haemolytic state can exist with anaemia; known as
    compensated haemolytic disease
  • the bone marrow can increase its output by 6-8 times by increasing the proportion of cells committed to erythropoiesis (RBC production) (erythroid hyperplasia) and by expanding the volume of active marrow
  • additionally, reticulocytes (immature RBCs) are released prematurely
  • reticulocytes are larger than mature cells, hence macrocytic, and stain with light blue tinge on a peripheral blood stain (polychromasia)
197
Q

what are reticulocytes like, compared to mature RBCs?

A

reticulocytes are larger than mature cells, hence macrocytic, and stain with light blue tinge on a peripheral blood stain (polychromasia)

198
Q

what are the main causes of haemolytic anaemia?

A
  • RBC membrane defects: hereditary spherocytosis
  • enzyme defects: glucose-6-phosphate dehydrogenase (G6PD) deficiency
  • haemoglobinopathies: beta thalassaemia, alpha thalassaemia, sickle cell disease
  • autoimmune haemolytic anaemia
199
Q

what are features of haemolytic anaemia?

A
  • high serum unconjugated bilirubin
  • high urinary urobilinogen
  • high faecal stercobilinogen
  • splenomegaly
  • bone marrow expansion
  • reticulocytosis
200
Q

what are requirements for red cell formation?

A

hormones, metals, iron, B12, folic acid, other vitamins

201
Q

what is bilirubin bound to in the plasma?

A

albumin

202
Q

what is the epidemiology of hereditary spherocytosis?

A
  • the most common inherited haemolytic anaemia in Northern Europeans
  • autosomal dominant
  • however in 25% it can occur spontaneously with neither parent affected
203
Q

what are risk factors for hereditary spherocytosis?

A

family history

204
Q

what is the pathophysiology of hereditary spherocytosis?

A
  • caused by defects in the red cell membrane resulting in RBCs losing part of the cell membrane as they pass through the spleen, possibly because the lipid bilayer is inadequately supported by the membrane skeleton due to deficiency in the structural protein spectrin
  • the abnormal cell membrane is associated functionally with an increased permeability to Na+ requiring an increased rate of active transport of Na+ out of the cells, which is dependent on ATP produced by glycolysis
  • the surface to volume ratio decreases and the cells become spherocytic/spherical in shape
  • spherocytes are more rigid and less deformable than normal red cells
  • they are unable to pass through the splenic microcirculation so they become trapped in the spleen and thus have a shortened lifespan and are destroyed via extravascular haemolysis
205
Q

what are spherocytes like compared to normal RBCs?

A

more rigid and less deformable

206
Q

what is the clinical presentation of hereditary spherocytosis?

A
  • jaundice at birth
  • however the onset of jaundice can be delayed for many years and some patients may go through life with no symptoms and are detected only during family studies
  • varies from symptom-free carrier to severe haemolysis with anaemia, jaundice and splenomegaly
  • splenomegaly
  • ulcers on the leg
  • chronic haemolysis leads to the formation of pigment gallstones
207
Q

what may interrupt the course of hereditary spherocytosis?

A
  • aplastic anaemia (sudden stop in RBC production) e.g. after infections, particularly with erythrovirus (parvovirus)
  • megaloblastic anaemia (result of folate depletion) caused by the hyperactivity of the bone marrow
  • haemolytic crises
208
Q

how is hereditary spherocytosis diagnosed?

A
  • blood film
  • blood count
  • haemolysis
  • direct antiglobulin (Coomb’s test)
  • diagnosis is usually straightforward and made on the basis of clinical features, family history, laboratory investigation and exclusion of other causes of haemolytic anaemia, particularly autoimmune haemolytic anaemia.
209
Q

what is seen on a blood film in hereditary spherocytosis?

A

shows spherocytes (spherical RBCs) and reticulocytes

210
Q

what is seen on a blood count in hereditary spherocytosis?

A

anaemia (Hb low) and increased reticulocytes

211
Q

what is seen in a direct antiglobulin (Coombs’) test in hereditary spherocytosis?

A

this is negative, virtually ruling out an autoimmune haemolytic anaemia in which spherocytes are also commonly present

212
Q

what is treatment of hereditary spherocytosis?

A

splenectomy:
• to relieve symptoms due to anaemia or splenomegaly, reverse growth failure and prevent recurrent gallstones
• best to postpone until after childhood due to infection risk post-op
• after operation do appropriate immunisations and life-long penicillin prophylaxis

213
Q

what is the epidemiology of glucose-6-phosphate dehydrogenase (G6PD) deficiency?

A
  • presents with haemolytic anaemia and affects millions of people worldwide, particularly in Africa, around the Mediterranean and in the Middle East and SE Asia
  • heterogenous X-linked; thus more common in males than females
  • however, female heterozygotes can also have clinical problems due to lyonisation whereby, because of random X-chromosome inactivation, female heterozygotes have two populations of red cells; a normal one and a G6PD deficient one
  • there are over 400 structural types of G6PD and mutations are mostly single amino acid substitutions (missense point mutations)
214
Q

what is the pathophysiology of G6PD deficiency?

A
  • G6PD is vital for a reaction that is necessary for RBCs by providing a NADPH which is used with glutathione to protect the RBC from oxidative damage from compounds such as hydrogen peroxide
  • this inherited enzyme deficiency thus results in reduced RBC lifespan due to oxidant damage
  • gene for G6PD is localised to chromosome Xq28 near the factor VIII gene
215
Q

where is the gene for G6PD?

A

localised to chromosome Xq28 near the factor VIII gene

216
Q

what reaction is G6PD involved in?

A

glucose-6-phosphate -> 6-phosphogluconate

  • catalyses this reaction
  • NADP+ -> NADPH
  • NADPH -> NADP+//GSSG -> GSH catalyesd by glutathione reductase
217
Q

what is the clinical presentation of G6PD deficiency?

A
  • most are asymptomatic but may get oxidative crisis due to reduction in glutathione production
  • chronic haemolytic anaemia
  • neonatal jaundice
  • acute haemolysis precipitated by ingestion of fava beans and drugs e.g. quinine, sulphonamides, quinolones and nitrofurantoin
  • clinical features are due to rapid intravascular haemolysis with symptoms of anaemia, jaundice and haemoglobinuria
218
Q

what precipitates oxidative crisis in G6PD deficiency?

A
  • occurs due to reduction in glutathione production
  • acute drug-induced haemolysis
  • made worse by ingesting fava beans
219
Q

what can cause acute drug induced haemolysis in G6PD deficiency?

A
  • aspirin
  • antimalarials such as: primaquine, quinine and chloroquine
  • antibacterials such as; most sulphonamides, nitrofurantoin and chloramphenicol
  • dapsone
  • quinidine
220
Q

how is G6PD diagnosed? what is the blood film like during attacks?

A
  • blood count is normal between attacks
  • blood film during attack:
    • irregularly contracted cells
    • bite cells (cells with an indentation in the membrane)
    • reticulocytosis - increased reticulocytes
  • G6PD enzyme levels - will be low (but note, immediately after attack the test may be normal since the oldest RBCs with the least G6PD activity are destroyed selectively, so results may show falsely high concentration of G6PD)
221
Q

what is the treatment of G6PD deficiency?

A
  • stop offending drugs and/or fava beans
  • blood transfusion may be lifesaving
  • splenectomy is not usually helpful
  • avoid precipitating factors
222
Q

what is the structure of normal haemoglobin?

A

HbA = haem + 2 alpha chains + 2 beta chains

- 97% of adult haemoglobin (Hb)

223
Q

what is the structure of foetal haemoglobin?

A

HbF = haem + 2 alpha chains + 2 gamma chains

- normal Hb in foetus from 3rd to 9th month; increased in beta-thalassaemia

224
Q

what is the structure of delta haemoglobin?

A

HbA2 = haem + 2 alpha chains + 2 delta chains

- 2% of adult Hb; elevated in beta-thalassaemia

225
Q

what are the distributions of haemoglobin in an adult?

A
HbA = 97%
HbA2 = 2%
HbF = 1%
226
Q

what are the thalassaemias?

A
  • disorders of quantity (reduced production)
    • normally, there is balanced 1:1 production of alpha and beta chains
    • these are genetic diseases of unbalanced Hb synthesis, with under production (or no production) of one globin chain. arise from one or multiple gene defects
    • the precipitation of the imbalanced globin chains within red cell precursors results in cell damage and death of precursors in bone marrow i.e ineffective erythropoiesis
    • the precipitation of these imbalanced globin chains in mature red cells leads to haemolysis
    • these are common in areas from the Mediterranean to the Far East
227
Q

what happens in beta thalassaemia?

A
  • little or no beta chain production resulting in excess alpha chains
  • these excess alpha chains combine with whatever delta and gamma chains are produced
  • resulting in increased HbA2 (Hb delta) and HbF (Hb gamma)
  • caused by over 200 genetic defects
228
Q

what is the pathophysiology of beta thalassaemia?

A
  • unlike alpha-thalassaemia, the defects are normally point mutations as opposed to gene deletions
  • the mutations result in defects in transcription, RNA splicing and modification, translation via frame shifts and nonsense codons producing highly unstable beta globulin which cannot be utilised
  • in heterozygous beta-thalassaemia there is usually asymptomatic microcytosis with or without mild haemolysis
229
Q

what can beta thalassaemia be divided into?

A
  • B thalassaemia minor (trait carrier)
  • B thalassaemia intermedia
  • B thalassaemia major
230
Q

what are clinical presentations of beta thalassaemia minor?

A
  • common carrier state: heterozygous beta thalassaemia
  • asymptomatic
  • anaemia is mild or absent
  • RBCs are hypochromic (pale) and microcytic with a low MCV and MCH
  • can be confused with iron deficiency but can be distinguished since serum ferritin and the iron stores are normal
  • Hb electrophoresis shows a raised HbA2 and often a raised HbF
231
Q

how can beta thaassaemia minor be distinguished from iron deficiency?

A

has normal serum ferritin and iron stores

232
Q

what are clinical presentations of beta thalassaemia intermedia?

A
  • includes those who are symptomatic with moderate anaemia (Hb 70-100g/L) that do not require regular transfusions
  • splenomegaly
  • bone deformities
  • recurrent leg ulcers
  • gallstones
  • infections
  • may be caused by combination of homozygous beta and alpha thalassaemias
233
Q

what are clinical presentations of beta thalassaemia major?

A
  • presents in children with homozygous beta thalassaemia in the first year of life with severe anaemia (Cooley’s anaemia), failure to thrive and recurrent infections
  • life-long transfusion dependant
  • hypertrophy of ineffective bone marrow leads to bone abnormalities: the thalassaemic facies, with an enlarged maxilla and prominent frontal and parietal bones
  • skull X-ray shows ‘hair on end’ sign due to increased marrow activity
  • blood results: MCV very low; microcytic
  • blood film: large and small irregular hypochromic RBCs
  • serum ferritin is normal
234
Q

what how is beta thalassaemia diagnosed? what are features of it on blood count and film?

A
  • in homozygous disease; blood count and film show:
    • hypochromic/microcytic anaemia
    • raised reticulocyte count
    • nucleated RBC in peripheral circulation
  • haemoglobin electrophoresis shows increased HbF (gamma) and absent or reduced HbA (normal)
235
Q

what is treatment of beta thalassaemia?

A
  • regular (every 2-4 weeks) life-long transfusions to keep Hb above 100g/L and to suppress the ineffective extramedullary haematopoiesis and to allow normal growth
  • long-term folic acid supplements
  • iron overload by repeated blood transfusions may lead to damage of endocrine glands, liver, pancreas and heart, with death in the second decade from cardiac failure
  • iron-chelating agents to prevent iron overload; oral deferiprone and SC desderrioxamine
  • 200mg daily ascorbic acid to increase urinary excretion of iron
  • splenectomy if hypersplenism persists with increasing transfusion requirements, but do after childhood to reduce infection risks
  • bone marrow transplant
236
Q

what are the side effects of iron chelating agents?

A

pain, deafness, cataracts and retinal damage

237
Q

why is ascorbic acid used in beta thalassaemia?

A

to increase urinary excretion of iron

238
Q

what is the aim of regular life-long transfusions in beta thalassaemia?

A
  • keep Hb above 90g/L
  • suppress ineffective extramedullary haematopoiesis
  • allow normal growth
239
Q

what are complications of transfusions in beta thalassaemia?

A
  • progressive increase in body iron load
  • mainly deposited in the liver and spleen resulting in liver fibrosis and cirrhosis
  • also deposited in the endocrine glands and heart resulting in diabetes, hypothyroidism, hypocalcaemia and premature death
240
Q

what is the pathophysiology of alpha thalassaemia?

A
  • in contrast to beta-thalassaemia, alpha-thalassaemia is often caused by gene deletions
  • there are four alpha-globin genes per cell
  • the gene for alpha-globin chains is duplicated on both chromosomes 16
  • the deletion of one alpha chain or both alpha-chain genes on each chromosome 16 may occur (deletion of one alpha chain is most common)
241
Q

what can alpha thalassaemia be divided into?

A

four, three, two and one gene deletion and whether a chain synthesis is partial or completely absent

242
Q

what is the clinical presentation of four gene deletion alpha thalassaemia?

A
  • deletion of both genes on both chromosomes
    • there is no alpha chain synthesis
    • only Hb Barts (4 gamma chains) is present
    • Hb Barts cannot carry O2 and is incompatible with life
    • infants are either stillborn or die very shortly after birth
    • they are pale and oedematous and have enormous livers and spleens; known as hydrops fetalis
243
Q

what is Hb Barts?

A
  • 4 gamma chains present
  • occurs in 4 gene deletion alpha thalassaemia
  • cannot carry O2 and is incompatible with life
244
Q

what is the clinical presentation of three gene deletion alpha thalassaemia?

A
  • severe reduction in alpha chain synthesis results in HbH disease, which is common in parts of Asia
  • HbH has 4 beta-chains
  • moderate anaemia and splenomegaly
  • patient is usually not transfusion dependent
245
Q

what is HbH?

A

4 beta chains

- found in alpha thalassaemia, biologically useless

246
Q

what is the clinical presentation of two gene deletion alpha thalassaemia?

A
  • microcytosis with or without mild anaemia

- carrier

247
Q

what is the clinical presentation of one gene deletion alpha thalassaemia?

A

usually a normal blood picture

248
Q

what is sickle cell anaemia?

A

a disorder of quality (abnormal molecule or variant haemoglobins) where the sickle beta-globin gene is inherited

249
Q

what is the epidemiology of sickle cell anaemia?

A
  • commonest in Africans but also in India, Middle East and southern Europe
  • autosomal recessive disorder causing the production of abnormal beta globin chains
  • 1 in 4 chance of disease
  • 50% chance of being a carrier
  • 1 in 4 chance of being disease free
250
Q

what are risk factors for sickle cell anaemia?

A
  • African

- family history

251
Q

what is the pathophysiology of sickle cell anaemia?

A
  • sickle cell haemoglobin (HbS) results from a single-base mutation of adenine to thymine which produces a substitution of valine for glutamic acid at the sixth codon of the beta-globin chain
  • in the heterozygous state (sickle cell trait), only one chromosome carries the gene; these are carriers of the disease
  • in the homozygous state (sickle cell anaemia), both genes are abnormal
  • since the synthesis of HbF (gamma or fetal) is normal, the disease does not manifest itself until the HbF decreases to adult levels at about 6 months of age
  • sickle cell haemoglobin (HbS) is insoluble and polymerises when deoxygenated
  • the flexibility of the cells is decreased and they become rigid and take up their characteristic sickle appearance
  • process is initially reversible but, with repeated sickling, the cells eventually lose their membrane flexibility and become irreversibly sickled
  • irreversibly sickled cells are dehydrated and dense, and will not return to normal when oxygenated
252
Q

what does sickling of RBCs result in?

A
  • a shortened RBC survival resulting in haemolysis
  • impaired passage of cells through the microcirculation (vasoocclusion), leading to obstruction of small vessels and tissue infarction and thus intense pain
253
Q

what precipitates sickling of RBCs?

A

infection, dehydration, cold, acidosis or hypoxia

254
Q

why do sickle cell patients often feel well despite being anaemic?

A
  • HbS releases its oxygen to the tissues more readily than normal RBCs
  • except in crises or complications
255
Q

what is the clinical presentation of heterozygous sickle cell trait?

A
  • symptom free with no disability expect in hypoxia e.g. in unpressurised aircraft or anaesthesia when vaso-occlusive events may occur
  • carriage offers protection against falciparum malaria
256
Q

what does sickle cell carriage offer protection against?

A

Falciparum malaria

257
Q

what are clinical presentations of homozygous sickle cell anaemia?

A
  • vaso occlusive crises
  • acute chest syndrome
  • pulmonary hypertension
  • anaemia
258
Q

what are features of vaso-occlusive crises in homozygous sickle cell anaemia?

A
  • acute pain in the hands and feet (dactylitis) owing to vaso-occlusion of the small vessels and avascular necrosis of the bone marrow in children
  • in adults there is pain in the long bones such as the femur, spine, ribs and pelvis due to avascular necrosis of the bone marrow
  • possible to have CNS infarction in children leading to stroke, seizures and cognitive defects
  • attacks vary in frequency and can occur as much as every day to once a year depending on patient
259
Q

what is the most common cause of death in adults with sickle cell anaemia?

A

pulmonary hypertension and chronic lung disease

260
Q

what are features of acute chest syndrome in homozygous sickle cell anaemia? what is it caused by?

A
  • a vaso-occlusive crisis of the pulmonary vasculature
  • occurs in 30%
  • pulmonary hypertension and chronic lung disease are the most common causes of death in adults with sickle cell disease
  • caused by infection (due to Chlamydia, mycoplasma and Streptococcus pneumoniae), fat embolism from necrotic bone marrow or pulmonary infarction due to sequestration of sickle cells (where sickle cells get trapped in the pulmonary vasculature)
  • results in shortness of breath, chest pain, hypoxia, fever, cough, dyspnoea and pulmonary infiltrates on CXR
261
Q

what are features of pulmonary hypertension in homozygous sickle cell anaemia? what is it caused by?

A
  • defined as a mean pulmonary artery pressure greater than 25mmHg by right heart catheterisation
  • occurs in 10% of patients
  • most probably caused by damage from repeated chest crises and repeated thromboembolism and intravascular haemolysis
  • increases the risk of hypoxaemia and worsening sickle cell crises
262
Q

what are features of anaemia in homozygous sickle cell anaemia?

A

chronic haemolysis produces a stable haemoglobin level (60-80g/L) with a high reticulocyte count (10-20%) however an acute fall in the haemoglobin level can occur

263
Q

why may an acute fall in the haemoglobin level occur in homozygous sickle cell anaemia?

A

splenic sequestration (where sickle cells get trapped in the spleen due to their shape):

  • resulting in the acute painful enlargement of spleen and an acute fall in Hb
  • can eventually lead to a fibrotic, non-functioning spleen
  • liver sequestration can also occur

bone marrow aplasia:
- most commonly follows infection with erythrovirus B19, which invades proliferating erythroid progenitors
- there is a rapid fall in haemoglobin with no
reticulocytes in the peripheral blood because of the failure of erythropoiesis in the marrow - aplastic crisis

further haemolysis due to drugs or acute infection

264
Q

what are long-term problems caused by sickle cell anaemia?

A
  • growth and development
  • avascular necrosis of bones results in shortened, deformed bones in children
  • infection in bones, lungs and kidneys (most susceptible to vaso-occlusive crisis)
  • splenic atrophy (resulting in susceptibility to infection with pneumococcus, salmonella species and haemophilus)
  • cardiac
  • neurological (fits and strokes)
  • liver: chronic hepatomegaly and liver dysfunction due to trapping of sickle cells
  • sequestration of red cells within corpora cavernosa causes priapism (prolonged painful erections) and eventual impotence
  • renal: chronic tubulointerstitial nephritis, CKD
  • eye: retinopathy, vitreous haemorrhage and retinal detachments
  • pregnancy: impaired placental blood flow can result in spontaneous abortion
265
Q

how can sickle cell anaemia cause problems in bones?

A
  • these are common sites for vaso-occlusive crises resulting in chronic infarcts
  • avascular necrosis of the hips and shoulders, compression of vertebrae and shortening of bones in the hands and feet occur
  • osteomyelitis can occur due to Staphylococcus Aureus, Staph. pneumoniae and salmonella
266
Q

how is sickle cell anaemia diagnosed? what are the results of the tests used?

A

blood count:
• level of Hb is in the range of 60-80 g/L
• raised reticulocyte count

blood films:
• sickled erythrocytes shown

sickle solubility test will be positive

Hb electrophoresis:
• confirms diagnosis
• shows 80-95% HbS and absent HbA
• aim for diagnosis at birth (cord blood) to aid prompt pneumococcal prophylaxis

sickling can be induced in vitro with sodium dithionite

267
Q

what is treatment of sickle cell anaemia?

A
  • precipitating factors such as infection, cold and dehydration should be avoided or treated quickly e.g prophylaxis vaccines
  • transfusion are not routinely given in steady-state anaemia
  • pneumococcal and influenza vaccination given routinely and daily penicillin 500mg orally
  • folic acid for all haemolysis patients
  • hospital admission when pain not controlled by non-opiate analgesia or if there are complications
268
Q

what is the treatment for acute painful attacks in sickle cell anaemia?

A
  • IV fluids
  • analgesia: morphine, codeine, paracetamol and NSAIDs
  • oxygen and antibiotics if required
269
Q

when is blood transfusion given for sickle cell anaemia?

A
  • acute chest syndrome
  • acute anaemia due to acute splenic sequestration
  • aplastic crisis
  • stroke
  • heart failure
  • note: increased risk of iron overload
270
Q

what is the treatment for anaemia in sickle cell anaemia?

A
  • oral hydroxycarbamide: increases HbF concentrations

* stem cell transplant

271
Q

what is aplastic anaemia due to bone marrow failure?

A
  • rare but serious stem cell disorder
  • defined as pancocytopenia (this is a reduction in all the major cell lines; red cells, white cells and platelets) with hypocellularity (aplasia) of the bone marrow (marrow stops making cells)
  • it is usually an acquired condition but may be inherited
272
Q

what are the main causes of aplastic anaemia due to bone marrow failure?

A
  • congenital e.g. Fanconi’s anaemia
  • idiopathic acquired (67% of cases)
  • benzene, toluene, insecticides and glue sniffing
  • cytotoxic drugs and radiation
  • chemotherapeutic drugs
  • antibiotics such as chloramphenicol, carbamazepine and azathioprine
  • infections such as Epstein-Barr virus, HIV, hepatitis and TB
  • paroxysmal nocturnal haemoglobinuria
  • miscellaneous, e.g. pregnancy
273
Q

what is the pathophysiology of aplastic anaemia due to bone marrow failure?

A
  • caused by a reduction in the number of pluripotent stem cells, together with a fault in those remaining or an immune reaction against them so that they are unable to repopulate the bone marrow
  • failure of only one cell line may also occur, resulting in isolated deficiencies, e.g. red cell aplasia
274
Q

what is the clinical presentation of aplastic anaemia due to bone marrow failure?

A

symptoms result because of RBC, WBC and platelet deficiency:
• anaemia
• increased susceptibility to infection
• bleeding
• bleeding gums, epitaxis, mouth infections, bruising with minimal trauma and blood blisters in mouth

275
Q

what are the differential diagnoses of aplastic anaemia due to bone marrow failure?

A

must be differentiated from other causes of pancocytopenia:

  • aplastic anaemia
  • drugs
  • megaloblastic anaemia
  • bone marrow infiltration or replacement: lymphoma, acute leukaemia, myeloma, secondary carcinoma, myelofibrosis
  • hypersplenism
  • SLE
  • disseminated TB
  • paroxysmal nocturnal haemoglobinuria
  • overwhelming sepsis
276
Q

how can aplastic anaemia due to bone marrow failure be diagnosed?

A

blood count:
• pancocytopenia with low or absent reticulocyte count

bone marrow examination:
• hypocellular marrow with increased fat spaces

277
Q

what is the treatment of aplastic anaemia due to bone marrow failure?

A
  • treatment of cause
  • withdrawal of offending agent, supportive care and some form of definitive therapy
  • main concern is infection thus any patient with severe neutropenia with a fever needs to be given broad-spectrum parenteral antibiotics urgently
  • give red cell transfusion and platelets
  • bone marrow transplant:
    • from HLA identical sibling or donor treatment of choice in those under 40
    • blood and platelet transfusion used cautiously in patients who are candidates for bone marrow transplant to avoid sensitisation
  • immunosuppressive therapy with antithymocyte globulin (ATG) and ciclosporin in those:
    • over 40
    • below 40 with severe disease who do not have a HLA identical sibling donor
    • those who are transfusion dependent
278
Q

what is given for immunosuppressive therapy for aplastic anaemia due to bone marrow failure?

A

antithymocyte globulin (ATG) and ciclosporin

279
Q

when is immunosuppressive therapy given in aplastic anaemia due to bone marrow failure?

A
  • over 40
  • below 40 with severe disease who do not have a HLA identical sibling donor
  • those who are transfusion dependent
280
Q

what is the definition of polycythema?

A
  • can also be referred to erythrocytosis, which is essentially is an increased red blood cell mass, whereas polycythaemia refers to any increase in RBCs
  • defined as an increase in haemoglobin, packed cell volume (PCV) aka haematocrit and RCC - these measurements are all concentrations and are directly dependent on plasma volume as well as RBC mass
  • PCV is a more reliable indicator of polycythaemia than Hb, which may be disproportionally low in iron deficiency
281
Q

what can polycythaemia be divided into?

A
  • absolute (i.e. due to increase in RBC mass; primary or secondary)
  • relative (i.e. decreased plasma volume and normal RBC mass; apparent polycythaemia or dehydration)
282
Q

what are causes of primary and secondary absolute polycythemia?

A

due to an increase in RBC mass

primary:

  • polycthaemia vera (PV) or
  • mutations in erythropoietin receptor
  • high oxygen affinity haemoglobins

secondary:

  • hypoxia e.g. high altitude, chronic lung disease, cyanotic congenital heart disease or heavy smoking (appropriate increase in EPO)
  • inappropriately high erythropoietin secretion e.g. in renal carcinoma and hepatocellular carcinoma
283
Q

what is the epidemiology/causes of Polycythaemia Vera?

A
  • arises from a single haematopoietic progenitor cell and leads to excessive proliferation of red cells and variable increase in platelets and myeloid cells
  • over 95% of patients have acquired mutations of the gene Janus kinase 2 (JAK2)
  • in the vast majority of cases there is a point mutation that causes the substitution of phenylalanine for valine at position 617
  • JAK2 is a cytoplasmic tyrosine kinase that transduces signals, especially those triggered by haemopoietic growth factors such as erythropoietin
  • commoner if over 60
284
Q

what is JAK2?

A

Janus Kinase 2

- cytoplasmic tyrosine kinase that tranduces signals, especially those triggered by haemopoietic growth factors e.g. EPO

285
Q

what is the pathophysiology of Polycythaemia Vera?

A
  • a clonal stem cell disorder resulting in a malignant proliferation of a clone derived from a single haematopoietic progenitor cell
  • the erythroid progenitor offspring are unusual in not needing erythropoietin to avoid apoptosis
  • this results in the excess proliferation of RBCs, white blood cells and platelets which causes a raised haematocrit (PCV) resulting in hyperviscosity and thrombosis
  • major complications are thrombosis and haemorrhage
286
Q

what is the definition of Polycythaemia Vera?

A

a clonal stem cell disorder resulting in a malignant proliferation of a clone derived from a single haematopoietic progenitor cell

287
Q

what is the clinical presentation of Polycythaemia Vera?

A
  • symptoms and signs are due to hypervolaemia and hyperviscosity
  • may be asymptomatic and only detected on FBC
  • usually presents in patients over 60
  • headache, vertigo, tinnitus, visual disturbance, angina pectoris, intermittent claudication, pruritus and venous thrombosis
  • severe itching after a hot bath or when the patient is warm
  • erythromelalgia; burning sensation in fingers and toes
  • increased risk of gout due to increased cell turnover and uric acid production
  • intermittent claudication
  • plethoric complexion; congested or swollen with blood in facial skin
  • hepatosplenomegaly; due to extramedullary haemopoiesis - distinguishes PV from secondary polycythaemia
  • risk of thrombosis and haemorrhage due to friable haemostatic plugs
288
Q

what are some vague symptoms due to hyperviscosity that occur in polycythaemia vera?

A
• headaches
• itching
• tiredness
• dizziness
• tinnitus
• visual disturbance
note: these symptoms are common in those over 60 hence why PV is often missed
289
Q

how is polycythaemia vera diagnosed? what is the major/minor criteria?

A

blood count showing raised WCC and platelet counts suggests PV as opposed to other causes of polycythaemia

  • demonstrating evidence of increased red cell volume (Hb >185g/L in men and >165g/L in women) and a gain of function mutation in JAK2 with at least one of:
  • erythroid hyperplasia, with increased numbers of megakaryocytes and granulocytes on bone marrow examination
  • serum erythropoietin levels below normal
  • erythroid colony formation in vitro in the absence of exogenous erythropoietin.
290
Q

what is the treatment of polycythaemia vera?

A
  • no cure
  • treatment aims to maintain a normal blood count and to prevent complications of the disease, particularly thrombosis and haemorrhage
  • venesection to maintain PCV<0.45L/L
  • chemotherapy: hydroxycarbamide and busulfan used to reduce platelet count
  • low dose aspirin alongside above treatments
  • anagrelide inhibits megakaryocyte differentiation and is useful for thrombolysis
  • radioactive phosphorus but only in those over 70 due to increased risk of acute leukaemia
  • allopurinol to block uric acid production and reduce gout
291
Q

why is venesection used in Polycythaemia Vera?

A
  • aims to lower PCV and platelet count
  • removal of 400-500mL of blood weekly to relieve symptoms
  • often used as sole treatment
292
Q

what chemotherapy is used in Polycythaemia Vera?

A

hydroxycarbamide and low-dose busulfan for those who do not tolerate venesection or have poorly controlled features of the disease e.g. thrombocytosis (too many platelets)

293
Q

why is allopurinol used in Polycythaemia Vera?

A

to block uric acid production and reduce gout

294
Q

what are the consequences of arterial thrombi formed in different places?

A

coronary circulation = MI
cerebral circulation = TIA or stroke
peripheral circulation = peripheral vascular disease

295
Q

what are the risk factors for arterial thrombosis?

A
  • smoking
  • hypertension
  • diabetes
  • hyperlipidaemia
  • obesity/sedentary lifestyle
  • stress
296
Q

what is the pathophysiology and causes of arterial thrombosis?

A
  • result of an atheroma that forms in areas of turbulent blood flow such as the bifurcation of arteries
  • platelets adhere to the damaged vascular endothelium and aggregate in response to ADP and thromboxane A2
  • plaque rupture leads to the exposure of blood containing factor VIIa to tissue factor within the plaque which may trigger blood coagulation and lead to thrombus formation
  • this can result in complete occlusion of the vessel or embolisation that produces distal obstruction
297
Q

what is done to prevent an arterial thrombosis?

A

• COX inhibitor e.g. aspirin which inhibits platelet aggregation
• P2Y12 inhibitor e.g. clopidogrel which inhibits ADP from binding to the P2Y12 receptor
• dipyramidole inhibits phosphordiesterase-mediated breakdown of cyclic AMP which prevents platelet activation
• antibodies, peptides and non-peptide antagonists block GpIIb/IIIa receptor, inhibiting the final common pathway of platelet aggregation
- epoprostenol is a prostacyclin which is used to inhibit platelet aggregation during renal dialysis (with or without heparin) and is used in primary pulmonary hypertension

298
Q

what is the epidemiology of a DVT?

A
  • deep vein thrombosis (DVT) occurs in 25-50% of surgical patients and many non-surgical patients
  • 65% of below-knee DVTs are asymptomatic and these rarely embolise to the lung
  • commonly occurs after periods of immobilisation but it can occur in normal individuals for no obvious reasons
299
Q

what are genetic causes of DVT?

A
  • Factor V Leiden (5%) - common in caucasians, increase risk of thrombus
  • PT20210A (3%) - common in caucasians, increase risk of thrombus

rare:
• antithrombin deficiency
• Protein C or Protein S deficiency

300
Q

what are acquired causes of DVT?

A
  • anti-phospholipid syndrome
  • lupus anticoagulant
  • hyperhomocysteinaemia - high levels are reduced by taking folate
301
Q

where can DVTs occur? how severe can they be?

A
  • can occur in any vein of the leg or pelvis, but is particularly found in the veins of the calf
  • unlike arterial thrombosis, venous thrombosis usually occurs in normal vessels, often in the deep veins of the leg.
  • originates around the valves as red thrombi consisting of red cells and fibrin.
  • propagation occurs, inducing a risk of embolization to the pulmonary vessels.
  • chronic venous obstruction in the leg results in a permanently swollen leg
  • below the calf there are 6 veins, from the popliteal fossa upwards they merge into one
  • a DVT in the calf below the knee is not too much of a concern
  • a DVT in above the knee is potentially fatal and usually occurs in the proximal leg vein
302
Q

what are risk factors for a DVT?

A
  • increased age
  • pregnancy
  • synthetic oestrogen
  • trauma
  • surgery (especially pelvic or orthopaedic)
  • past DVT
  • cancer
  • obesity
  • immobility
  • family history
  • varicose veins with phlebitis
  • thrombophilia
  • IBD
  • nephrotic syndrome
303
Q

what is the clinical presentation of a DVT?

A
  • may be asymptomatic
  • pain in the calf, often with swelling, redness and engorged superficial veins
  • the affected calf is often warmer and there may be ankle oedema
  • with complete occlusion, particularly in a large vein can result in cyanotic discolouration of the limb and severe oedema
  • pulmonary embolism can occur with any DVT but is more frequent from an iliofemoral thrombosis and is rare with thrombosis confined to the veins below the knee
304
Q

what is used to diagnose a DVT?

A
  • plasma D-dimer
  • compression ultrasound
  • venography
  • Doppler ultrasound
305
Q

how can a plasma D-dimer be used to diagnose a DVT?

A
  • type of fibrinogen degradation product that is released into the circulation when a clot begins to dissolve
  • D-dimers are elevated in other conditions e.g. cancer, pregnancy and post-operatively and a positive result is not diagnostic of DVT and requires further imaging
  • a normal result excludes DVT
306
Q

what is treatment of DVT?

A
  • low molecular weight heparin for a minimum of 5 days (at first)
  • oral warfarin with a target INR of 2.5 (2-3) for 6 months after
  • direct acting oral anti-Coagulants (DOAC) similar to warfarin but do not need monitoring
  • compression stockings
  • inferior vena cava filters to reduce risk of pulmonary emboli
307
Q

how can DVTs be prevented?

A
  • early mobilisation post-op
  • compression stockings
  • thrombophylaxis for both low risk and high risk
  • assess risk of patients on admission
  • recent long distance, sedentary travel
308
Q

what are platelets? what are features of them?

A
  • platelets are anucleate cells formed by fragmentation of megakaryocytic (MK) cytoplasm in bone marrow
  • disc shape allows them to flow close to the endothelium
  • play an important role in primary haemostasis (stopping of bleeding)
  • life span of 7-10 days
  • old platelets are phagocytosed by splenic macrophages in red pulp
  • have surface glycoproteins
309
Q

what is the lifespan of a platelet?

A

7-10 days

310
Q

what controls platelet production?

A

thrombopoietin (TPO)
• produced mainly by the liver thus if liver damage then there will be reduced thrombopoietin and thus decreased platelets
• stimulates the production of platelets by megakaryocytes (MKs)
• binds to platelet and MK receptors
• decrease in platelets = less bound TPO = more TPO able to bind to MK = increased platelet production

311
Q

what are some important platelet receptors?

A
  • thromboxane A2 (TXA2)
  • P2Y12
  • glycoprotein IIb/IIIa
312
Q

what is the function of thromboxane A2?

A
  • platelet receptor
    • synthesised from arachidonic acid in platelets via cyclooxygenase (COX-1)
    • induces platelet aggregation and vasoconstriction
313
Q

what is the function of P2Y12?

A
  • receptor on platelets that is activated by ADP

* amplifies activation of platelets and helps activate glycoprotein IIb/IIIa

314
Q

what is the function of glycoprotein IIb/IIIa?

A
  • acts as a receptor for fibrinogen and von Willebrand Factor (vWF)
  • vWF is a clotting factor that is essential for platelets to adhere to damaged blood vessels
  • aids platelet adherence and aggregation
315
Q

what can cause normal numbers of platelets but reduced function?

A
  • congenital abnormality in platelet function
  • medication e.g. aspirin
  • von Willebrand disease resulting in reduced vWF activity meaning platelets are unable to bind to damaged blood vessels resulting in platelet dysfunction and thus muco-cutaneous bleeding
  • uraemia (raised urea levels in blood)
316
Q

what is thrombocytopenia?

A
  • this is a deficiency of platelets in the blood

- can either be due to decreased production or increased destruction

317
Q

what causes decreased production in thrombocytopenia?

A
  • congenital thrombocytopenia: absent/reduced/malfunctioning megakaryocytes (form platelets) in bone marrow
  • infiltration of bone marrow: leukaemia, metastatic malignancy (breast and prostate), lymphoma, myeloma, myelofibrosis
  • reduced platelet production by bone marrow
  • dysfunctional production of platelets in bone marrow: myelodysplasia
318
Q

what causes reduced platelet production by bone marrow?

A
  • low B12/folate
  • reduced thrombopoietin (TPO) e.g. in liver disease
  • medication: methotrexate and chemotherapy
  • toxins e.g. alcohol
  • infections e.g. viral (HIV) or TB
  • aplastic anaemia (autoimmune)
319
Q

what causes increased destruction of platelets?

A
  • autoimmune: immune thrombocytopenia purpura (ITP); primary or secondary
  • hypersplenism: portal hypertension and splenomegaly
  • drug related immune destruction: e.g. heparin induced thrombocytopenia
  • consumption of platelets: disseminated intravascular coagulopathy (DIC) or thrombotic thrombocytopenic purpura (TTP)
320
Q

what is immune thrombocytopenia purpura (ITP)?

A
  • thrombocytopenia due to immune destruction of platelets
  • the antibody-coated platelets are removed following binding to Fc receptors on macrophages
  • IgG antibodies form to platelets and megakaryocytes
321
Q

what are features of ITP in children?

A
  • primary, acute
  • occurs most commonly in the 2-6 year age group
  • has an acute onset with muco-cutaneous bleeding and there may be a history or recent viral infection including varicella zoster (chickenpox) or measles
  • may also follow immunisation
  • although bleeding may be severe, life-threatening
    haemorrhage is rare
  • sudden self-limiting purpura
322
Q

what are features of ITP in adults?

A
  • secondary, chronic
  • usually is less acute than in children
  • characteristically seen in women and may be associated with other autoimmune disorders such as SLE, thyroid disease and autoimmune haemolytic anaemia
  • it is also seen in patients with chronic lymphocytic leukaemia (CLL) and solid tumours, and after infections with viruses like HIV or Hep C
  • platelet autoantibodies are detected in about 60-70% of patients, and are presumed to be present, although not detectable in the remaining patients; the antibodies often have specificity for platelet membrane glycoproteins IIb/IIIa and/or Ib
323
Q

what is the clinical presentation of ITP?

A
  • easy bruising
  • epistaxis (nose bleed)
  • menorrhagia (heavy menstruation)
  • purpura (red or purple spots on the skin caused by bleeding underneath skin)
  • gum bleeding
  • major haemorrhage is rare
  • splenomegaly is rare
324
Q

how is ITP diagnosed?

A
  • bone marrow examination: shows thrombocytopenia with increased or normal megakaryocytes in the marrow
  • platelet autoantibodies (present in 60-70%); not needed for diagnosis
325
Q

what is first line treatment for ITP?

A
  • oral corticosteroids 1mg/kg body weight produces a response in 2/3rds of patients but relapse is common when dose is reduced.
  • IV immunoglobulin - raises platelet count more rapidly than steroids, thus useful for surgery
326
Q

what is second line treatment for ITP?

A
  • splenectomy
  • if splenectomy fails then high dose corticosteroids and immunosuppressive agents
  • thrombopoietin receptor agonists, e.g. romiplostim and eltrobopag, drive increased platelet production and may be used in refractory ITP
327
Q

what is ITP? what is its definition and presentation?

A

Immune Thrombocytopenia Purpura

  • defined as an isolated low platelet count with a normal bone marrow in absence of other causes of low platelets
  • causes a characteristic bruise-like rash and an increased tendency to bleed
328
Q

what is TTP? what is its pathophysiology?

A

Thrombotic Thrombocytopenic Purpura
• widespread adhesion and aggregation of platelets leads to microvascular thrombosis and thus consumption of platelets and thus profound
thrombocytopenia, low RBCs and often kidney, heart and brain dysfunction
• occurs due to a reduction in ADAMTS-13 (which is attacked by the immune system) - a protease that is normally responsible for the degradation of vWF
• large multimers of vWF form resulting in platelet aggregation and fibrin deposition in small vessels leading to microthrombi

329
Q

what are causes of TTP?

A
  • idiopathic
  • autoimmune e.g. SLE
  • cancer
  • pregnancy
  • drug associated e.g. quinine
330
Q

what is the clinical presentation of TTP?

A
  • florid purpura
  • fever
  • fluctuating cerebral dysfunction
  • haemolytic anaemia with red cell fragmentation, often accompanied by acute kidney injury
331
Q

how is TTP diagnosed?

A
  • coagulation screen is normal

- lactate dehydrogenase is raised as a result of haemolysis

332
Q

what is the treatment for TTP?

A
  • plasma exchange to remove the antibody to ADAMTS-13 as well as providing a source of ADAMTS-13
  • IV methylprednisolone
  • IV rituximab
  • platelet concentrates are contraindicated
333
Q

what is DIC?

A

disseminated intravascular coagulation

  • widespread generation of fibrin within blood vessels, caused by initiation of the coagulation pathway
  • consumption of platelets and coagulation factors occurs, as well as secondary activation of fibrinolysis, leading to production of FDPs, which contribute to bleeding by inhibiting fibrin polymerisation
334
Q

what is the epidemiology/pathophysiology of DIC?

A
  • never occurs in isolation
  • recognition that the patient has a clinical disorder that may result in DIC is the key to investigation and management
  • DIC arises because of systemic activation of coagulation either by release of procoagulant material, such as tissue factor or via cytokine pathways as part of the inflammatory response
  • such systemic activation leads to widespread generation of fibrin and depositing in the blood vessels, leading to thrombosis and multiorgan failure
  • also results in the consumption of platelets and clotting factors with increased risk of bleeding
335
Q

what are causes of DIC?

A

• massive activation of the coagulation cascade
• initiating factors are:
- extensive damage to vascular endothelium thereby exposing tissue factor
- enhance expression of tissue factor by monocytes in response to cytokines
• sepsis
• major trauma and tissue destruction
• advanced cancer
• obstetric complications

336
Q

what is the pathophysiology of DIC?

A
  • cytokine release in response to SIRS (systemic inflammatory response syndrome); usually caused by sepsis, trauma, pancreatitis, obstetric emergency or malignancy
  • widespread systemic generation of fibrin within blood vessels caused by the initiation of the coagulation pathway
  • either causes microvascular thrombosis and thus organ failure OR
  • the consumption of platelets and coagulation factors, leading to bleeding by inhibiting fibrin polymerisation (thus fibrin cannot polymerise)
337
Q

what is the clinical presentation of DIC?

A
  • patient is often acutely ill and shocked
  • bleeding may occur from the mouth, nose and venepuncture sites and there may be widespread ecchymoses (discolouration of skin due to bleeding
    caused by bruising)
  • confusion
  • bruising
  • thrombotic events occur as a result of vessel occlusion by fibrin and platelets; any organ may be involved but the skin, brain and kidneys are most affected
338
Q

how is DIC diagnosed?

A
  • severe thrombocytopenia
  • diagnosis can be suggested from history e.g severe sepsis, trauma or malignancy, clinical presentation and thrombocytopenia
  • decreased fibrinogen
  • elevated FDPs (fibrin degradation products) e.g. D-dimer due to the intense fibrinolytic activity that is stimulated by the presence of fibrin in the circulation
  • blood film shows fragmented RBCs
  • prolonged prothrombin time (PT), activated partial thromboplastin time (APTT) and thrombin time (TT)
339
Q

what is the treatment of DIC?

A
  • treat underlying condition
  • replace platelets if they are very low via transfusion
  • maintain count > 50 x 10^9/L
  • fresh frozen plasma (FFP) to replace the coagulation factors
  • cryoprecipitate to replace fibrinogen and some coagulation factors
  • red cell transfusion in patients who are bleeding
340
Q

what is heparin induced thrombocytopenia?

A

development of an IgG antibody against a complex formed between platelets and heparin

341
Q

what is the pathophysiology of heparin induced thrombocytopenia?

A
  • heparin binds to a protein in the blood called platelet factor 4 (PF4), forming a complex - PF4/Heparin
  • IgG then binds to this complex forming IgG/PF4/heparin which in turn then binds and activates platelets
  • this results in platelet consumption and thus thrombocytopenia
  • also results in thrombosis (arterial or venous) as well as skin necrosis
342
Q

who is most at risk for heparin induced thrombocytopenia?

A

cardiac bypass surgery (since lots of heparin is used) and those on unfractioned heparin treatment

343
Q

what is the presentation of heparin induced thrombocytopenia?

A

typically seen as a sharp fall in platelets around 5-10 days starting heparin treatment

344
Q

what is the treatment of heparin induced thrombocytopenia?

A
  • can be life-threatening and need to stop heparin immediately and try alternative anticoagulation even if platelets are low
  • never reexpose patient to heparin
345
Q

what is the definition of febrile neutropenia?

A

temperature recorded as above 38°C in a patient with absolute neutrophil count <1.0x109/L
• emergency since if not recognised or acted on quickly then patients can rapidly deteriorate and some will die

346
Q

what are at risk patients for febrile neutropenia?

A
  • any patient that has had chemotherapy less than 6 weeks ago
  • any patient who has had a stem cell transplant or high dose chemotherapy within the last year
  • any haematological condition resulting in neutropenia:
    • aplastic anaemia - bone marrow failure syndrome
    • autoimmune disease
    • leukaemia
  • people on methotrexate, carbimazole and clozapine
347
Q

what is the clinical presentation of febrile neutropenia?

A
  • pyrexia > 38°C
  • generally unwell with malaise
  • sweats/rigors
  • cough, sore throat, abdominal pain or diarrhoea
  • pain or erythema around central venous catheter
  • tachycardia, hypotension and raised respiratory rate
  • may not always have temperature/rigors due to inability to amount immune response
348
Q

what is the management of febrile neutropenia?

A
  • start broad spectrum IV antibiotics without waiting for results ideally within 1 hour of admission
  • do not catheterise if neutropenic due to increased infection risk
349
Q

what are at risk patients for malignant spinal cord compression?

A
  • any malignancy can cause spinal cord compression:
    • bone metastasis and vertebral collapse
    • local tumour extension
    • deposition of malignant cells within cord
  • commonly seen in:
    • myeloma
    • lymphoma
350
Q

what is clinical presentation of malignant spinal cord compression?

A
  • back pain
  • weakness/numbness in legs
  • inability to control bladder/bowel
  • saddle paraesthesia
  • uni/bilateral leg weakness
  • decreased perineal sensation
  • decreased anal tone
  • in acute cord compression, tone and reflexes will be reduced
351
Q

what is management of malignant spinal cord compression?

A
  • strict bed rest
  • high dose steroid e.g. oral dexamethosone
  • analgesia
  • urgent MRI of whole spine
352
Q

what is the pathophysiology of tumour lysis syndrome?

A

tumour cell breakdown

  • > hyperkalaemia -> cardiac instability and muscle cramps
  • > nucleic acid catabolism -> high uric acid -> uric acid crystal formation -> acute renal failure -> hyperkalaemia
  • > hyperphosphataemia -> precipitation calcium phosphate -> acute renal failure
  • > hyperphosphataemia -> hypocalcaemia -> tetany, muscle cramps, irritability, seizures
353
Q

what are at risk patients for tumour lysis syndrome?

A
  • high tumour burden
  • high grade disease i.e. rapid cell turnover
  • pre-existing renal impairment
  • increasing age
  • on drugs that increase uric acid formation
354
Q

what is the definition of hyperviscosity syndrome? what can it be caused by?

A
  • increase in blood viscosity
  • usually due to high levels of immunoglobulins:
    • seen in multiple myeloma and waldenstrom macroglobulinemia
  • can be due to high cell numbers e.g. leukaemia or polycythaemia
  • hyperviscosity results in vascular stasis and hypoperfusion
355
Q

what are clinical presentations of hyperviscosity syndrome?

A
  • mucosal bleeding
  • visual change due to hypoperfusion to retina
  • neurological disturbance due to brain hypoperfusion: vertigo, hearing loss, paraesthesia, ataxia, headache, seizure or stupor
  • shortness of breath
  • fatigue/anorexia
  • bruises/gum bleeding
  • dilated retinal veins/retinal haemorrhages on fundoscopy
  • ataxia/nystagmus
  • evidence of volume overload
356
Q

how is hyperviscosity syndrome diagnosed?

A
  • plasma viscosity level
  • CT head to exclude other causes of neurological signs
  • immunoglobulin levels
  • FBC
357
Q

what is the management of hyperviscosity syndrome?

A
  • keep hydrated
  • avoid blood transfusion since this will only make blood thicker
  • plasmapheresis; removes circulating immunoglobulins (paraprotein) to decrease serum viscosity, symptoms will improve
358
Q

what is the clinical presentation of hypercalcaemia?

A
  • confusion
  • bone pain
  • constipation
  • nausea
  • polyuria
  • abdominal pain
  • anorexia
  • renal stones
  • patients can get shortening QT interval and suffer cardiac arrest
359
Q

what is the treatment of hypercalcaemia?

A
  • IV hydration, 3-4L a day

- bisphosphonates e.g. zolendronate, reduces Ca2+ production but takes a few days to come into effect

360
Q

what are features of Ondansetron?

A
  • anti-emetic drug

- is a 5HT3 antagonist

361
Q

what are features of Heparin?

A
  • often used in bypass surgery IV
  • glycoaminoglycan
  • binds to antithrombin and increases its activity
  • indirect thrombin inhibitor
  • monitor with Activated Partial Thromboplastin Time (APTT)
  • given by continuous IV infusions
  • easily reversed by proteamine
362
Q

what are features of low molecular weight Heparin?

A
  • smaller molecule
  • less variation in dose
  • renally excreted
  • given once daily, weight-adjusted dose given sub-cutaneously (SC)
  • used for treatment and prophylaxis
363
Q

what are features of Aspirin?

A
  • inhibits cyclo-oxygenase (COX) IRREVERSIBLY
  • acts for the lifetime of platelets which is around 7-10 days
  • inhibits thromboxane formation and hence platelet aggregation
  • used in arterial thrombosis given orally once daily
  • Clopidogrel is similar but inhibits ADP induced platelet aggregation
  • need to stop aspirin 5 days before surgery so platelets can recover
364
Q

what are features of Warfarin?

A
  • orally active
  • prevents the synthesis of active factors II, VII, IX, X (1972)
  • antagonist/blocker of Vitamin K (required for II, VII, IX X factor synthesis since it makes post-translational changes on them to make them active)
  • long half life (36 hours)
  • prolongs the prothrombin time
  • can reverse with Vitamin K but takes time to produce new active factors
  • difficult to use - due to the presence of Vitamin K in the diet so if vitamin K increases then your warfarin requirement will also need to increase
  • thus need to monitor using INR (international normalised ratio which standardises prothrombin time)
  • usual target range = 2-3
  • higher range = 3-4.5
365
Q

what is INR? what is its usual target range?

A

International Normalised Ratio

  • standardises prothrombin time
  • usual target range = 2-3
  • higher target range = 3-4.5
366
Q

what are features of new oral anticoagulant drugs?

A
  • orally active - like aspirin and warfarin
  • direct action on factor II or X
  • no blood tests or monitoring required
  • has a shorter half life so given twice/once daily
  • used for extended thromboprophylaxis and treatment of atrial fibrillation and DVT and pulmonary embolism
  • effect is equivalent to INR 2-3, not higher, thus cannot be used with heart valves
  • not used in pregnancy
367
Q

what is the initial treatment of myeloma?

A

an alkylator (cyclophosphamide or melphalan), steroid (prednisolone or dexamethasone) and novel agent (bortezomib or thalidomide)

368
Q

what is the diagnosis of Hodgkin’s lymphoma? what is seen histologically?

A
  • by lymph node biopsy and histological examination
  • shows Reed-Sternberg cells (binucleate or multinucleate malignant B lymphocytes) in a background rich in benign small lymphocytes and histiocytes
369
Q

what are differential diagnoses of localised lymphadenopathy (Hodgkin’s lymphoma)?

A
  • local infection
  • pyogenic infection, e.g. tonsillitis
  • tuberculosis
  • lymphoma
  • secondary carcinoma
370
Q

what are differential diagnoses of generalised lymphadenopathy (Hodgkin’s lymphoma)?

A
  • infection
  • EBV
  • cytomegalovirus
  • Toxoplasma sp.
  • tuberculosis
  • HIV infection
  • lymphoma
  • leukaemia
  • systemic disease: systemic lupus erythematosus, sarcoidosis, rheumatoid arthritis
  • drug reaction e.g. phenytoin
371
Q

what is the prognosis of Hodgkin’s lymphoma?

A

prognosis is related to the stage of disease at presentation, with a 5-year survival rate of over 90% in stage I declining to 60% in stage IV. the presence of B symptoms indicates more severe disease with a worse prognosis

372
Q

how is PET/CT used after treatment of Hodgkin’s lymphoma?

A
  • PET/CT is used to detect disease activity after treatment and to distinguish between active tumour (PET-positive) and necrosis or fibrosis (PET-negative) in residual masses
  • irradiation, with its attendant complications, can be omitted in PET-negative masses after chemotherapy
373
Q

how is non-Hodgkin’s lymphoma classified?

A
  • malignant clonal expansion of lymphocytes, occurring at different stages of lymphocyte development
  • 80% of B cell origin
  • neoplasms of non-dividing mature lymphocytes are indolent, whereas those of proliferating cells (e.g. lymphoblasts, immunoblasts) are much more aggressive
  • B and T/natural killer cell lymphomas are further divided on this basis, e.g. precursor B cell lymphoma
  • then again subdivided based on cytogenetics and immunophenotyping
374
Q

what is the clinical presentation of non-Hodgkin’s lymphoma?

A
  • rare before 40yrs
  • painless peripheral lymph node enlargement
  • systemic symptoms may occur
  • extranodal involvement more common than in Hodgkin’s lymphoma; almost any organ in body can be involved
  • bone marrow infiltration -> anaemia, recurrent infections and bleeding
  • skin involvement with T cell lymphoma presents as mycosis fungoides and Sezary syndrome
375
Q

what is seen in the blood in non-Hodgkin’s lymphoma?

A
  • may show anaemia
  • elevated WCC or thrombocytopenia suggests bone marrow involvement
  • ESR may be raised
376
Q

what is myeloablative therapy?

A

the term used for treatment that employs high-dose chemotherapy or chemotherapy plus radiation, with the aim of clearing the bone marrow completely of both benign and malignant cells. without bone marrow replacement or ‘transplantation’, the patient would die of bone marrow failure

377
Q

what is allogenic bone marrow transplantation? what is the prognosis?

A
  • bone marrow or peripheral blood stem cells from another individual, usually a human leucocyte antigen (HLA)-identical sibling, are infused intravenously following myeloablative therapy
  • immunosuppression is required to prevent host rejection and graft-versus-host disease (GVHD)
  • blood count usually recovers within 3-4 weeks
  • mortality rate is 20-40% depending on age, often due to infection of GVHD
378
Q

what are some oncological emergencies?

A
  • neutropenic sepsis
  • superior vena cava syndrome
  • acute tumour lysis syndrome
  • spinal cord compression
  • hypercalcaemia
  • PE
  • raised intracranial pressure
379
Q

what is superior vena cava syndrome? what is its presentation and treatment?

A
  • can arise from any upper mediastinal mass but is most commonly associated with lung cancer and lymphoma.
  • presentation is with difficulty breathing and/or swallowing, oedematous facies and arms, and venous congestion in the neck with dilated veins in the upper chest and arms.
  • treatment is with immediate steroids, vascular stents, radiotherapy and chemotherapy for sensitive tumours.
380
Q

what is acute tumour lysis syndrome? what is its clinical presentation?

A
  • occurs as a result of treatment producing massive and rapid breakdown of tumour cells, leading to increased serum level of urate, potassium and phosphate with secondary hypocalcaemia
  • it is most commonly seen as a complication of treatment of acute leukaemia and high-grade lymphoma unless preventive measures are taken.
  • hyperuricaemia and hyperphosphataemia result in acute kidney injury through urate and calcium phosphate deposition in the renal tubules
381
Q

what is the prevention and treatment of acute tumour lysis syndrome?

A
  • prevention and treatment is with allopurinol, rasburicase (urate oxidase)
  • high fluid loads, e.g. 4–5 L daily by intravenous (i.v.) infusion prior to, and continuing during, chemotherapy
  • allopurinol (xanthine oxidase inhibitor) or rascurbicase (recombinant urate oxidase) both work to reduce uric acid production
  • monitor electrolytes
  • refer for dialysis if required
382
Q

what are genetic features of acute promyelotic leukaemia (APML)?

A
  • have the translocation t(15;17) involving the retinoic acid receptor alpha (RARa) on chromosome 17 and the promyelocytic leukaemia gene (PML) on chromosome 15
  • resulting PML-RARa fusion protein shows reduced sensitivity to retinoic acid and prevents differentiation of myeloid cells
383
Q

what is the translocation t(15;17)? what is it associated with? what are the genes involved?

A
  • associated with APML
  • retinoic acid receptor alpha (RARa) on chromosome 17
  • promyelocytic leukaemia gene (PML) on chromosome 15
  • resulting PML-RARa fusion protein shows reduced sensitivity to retinoic acid and prevents differentiation of myeloid cells
384
Q

what is a histological feature of AML?

A

Auer rods (a rod-like conglomeration of granules in the cytoplasm) within blast cells are pathognomonic of AML

385
Q

what does bone marrow aspirate show in acute leukaemias?

A

increased cellularity, with high percentage of abnormal lymphoid or myeloid blast cells

386
Q

what are different WHO classifications of AML (acute myeloid leukaemia)?

A
  1. AML with recurrent cytogenetic abnormalities (including acute promyelocytic leukaemia with t(15;17) or variants)
  2. AML with multilineage dysplasia (often secondary to pre-existing MDS)
  3. AML and MDS, therapy related, occurring after chemotherapy or radiotherapy
  4. AML - not otherwise categorised
387
Q

what are different WHO classifications of ALL (acute lymphoblastic leukaemia)?

A
  1. precursor B cell ALL
  2. Burkitt cell leukaemia
  3. precursor T cell ALL
388
Q

what is the management of acute leukaemias?

A
  • the initial requirement of therapy is to return the peripheral blood and bone marrow to normal (complete remission) with ‘induction chemotherapy’ tailored to the particular leukaemia and the individual patient’s risk factors.
  • the risk of failure of treatment is based on the cytogenetic pattern.
  • successful remission induction is always followed by further treatment (consolidation), the details being determined by the type of leukaemia and the patient’s risk factors (and the patient’s tolerance of treatment).
  • recurrence is almost invariable if ‘consolidation’ therapy is not given.
389
Q

what is supportive care for acute leukaemias?

A
  • correction of anaemia, thrombocytopenia and coagulation abnormalities by administration of blood, platelets and blood products
  • treatment of infection with IV antibiotics
  • prevention of acute tumour lysis syndrome
390
Q

what is treatment for AML with low risk of treatment failure (based on cytogenetic pattern)?

A

a moderately intensive combination of i.v. chemotherapy, e.g. cytosine arabinoside (cytarabine) and daunorubicin, is given at intervals to allow marrow recovery in between. this is followed by consolidation therapy with a minimum of four cycles of treatment given at 3–4-week intervals.

391
Q

what is treatment for AML with intermediate risk of treatment failure?

A

consolidating chemotherapy to induce remission followed by sibling-matched allogeneic bone marrow transplantation, despite its attendant risks.

392
Q

what is treatment for AML with high risk of treatment failure?

A

this is only curable with allogeneic transplantation but unfortunately ‘high risk’ is more common with advancing age when the toxicity of this treatment increases greatly.

393
Q

what is treatment of acute promyelocytic leukaemia?

A
  • APML is a variant of AML that is specifically associated with DIC which may worsen when treatment is started
  • combine chemotherapy with all-trans-retinoid acid (ATRA) which causes differentiation of promyelocytes and rapid reversal of bleeding tendency caused by DIC
  • successful remission induction followed with maintenance ATRA
394
Q

what is ATRA? how is it used to treat APML?

A

all-trans-retinoic acid

  • combined with chemotherapy
  • causes differentiation of promyelocytes and rapid reversal of bleeding tendency caused by DIC (which is associated with APML)
395
Q

what is the prognosis of ALL?

A
  • the prognosis in children with ALL is excellent, with almost all achieving complete remission and with 80% being alive and disease free at 5 years.
  • the results in adults are not as good, the prognosis getting worse with advancing years.
  • overall, about 70–80% achieve complete remission with only about 30% being cured.
396
Q

what is the treatment of ALL?

A
  • combination chemotherapy including vincristine, dexamethasone, asparaginase and daunorubicin.
  • details of consolidation will be determined by the anticipated risk of failure but is usually with intensive chemotherapy and then maintenance therapy for 2 years to reduce the risk of disease recurrence.
  • unlike AML, ALL has a propensity to involve the CNS, so treatment also includes prophylactic intrathecal drugs, methotrexate or cytosine arabinoside (cytarabine).
  • cranial irradiation is used in those at very high risk or in those with symptoms.
397
Q

what are clinical features of CML?

A
  • CML occurs most commonly in middle age and is characterized by the presence of the Ph chromosome.
  • there is an insidious onset, with fever, weight loss, sweating and symptoms of anaemia.
  • massive splenomegaly is characteristic.
398
Q

what is the prognosis of CML?

A
  • untreated, this chronic phase lasts 3–4 years.
  • this is usually followed by blast transformation, with the development of acute leukaemia (usually acute myeloid) and, commonly, rapid death.
  • less frequently, CML transforms into myelofibrosis, death ensuing from bone marrow failure.
399
Q

what are investigations of CML? what does the blood count/bone marrow aspirate show?

A
  • blood count usually shows anaemia and a raised white cell count (often > 100 × 109/L). the platelet count may be low, normal or raised.
  • bone marrow aspirate shows a hypercellular marrow with an increase in myeloid progenitors.
  • the Ph chromosome and the BCR-ABL oncogene are shown by cytogenetics and reverse transcriptase polymerase chain reaction (RT-PCR).
400
Q

what is the management of CML?

A
  • imatinib, a tyrosine kinase inhibitor that specifically blocks the enzymatic action of the BCR-ABL fusion protein, is first-line treatment for the chronic phase.
  • imatinib produces a complete haematological response in over 95% of patients, and 70–80% of these have no detectable BCR-ABL transcripts in the blood.
  • event-free, and overall, survival appear to be better than for other treatments - imatinib can be continued indefinitely.
  • in the acute phase (blast transformation) most patients have only a short-lived response to imatinib, and other chemotherapy as for acute leukaemia is used in the hope of achieving a second chronic phase.
401
Q

what is CLL?

A

CLL, the most common form of leukaemia, is an incurable disease of older people, characterized by an uncontrolled proliferation and accumulation of mature B lymphocytes.

402
Q

what are clinical features of CLL?

A
  • CLL usually follows an indolent course.
  • early CLL is generally asymptomatic and isolated peripheral blood lymphocytosis is frequent.
  • symptoms are a consequence of bone marrow failure: anaemia, infections and bleeding.
  • an autoimmune haemolysis contributes to the anaemia.
  • some patients may be asymptomatic, the diagnosis being a chance finding on the basis of a blood count performed for a different reason.
  • there may be lymphadenopathy and, in advanced disease, hepatosplenomegaly.
403
Q

what are investigations of CLL? what do they show?

A
  • blood count shows a raised white cell count with lymphocytosis (> 5 × 109/L). there may be anaemia and thrombocytopenia.
  • blood film shows small lymphocytes of mature appearance with ‘smear or smudge cells’, an artefactual finding due to cell rupture while the film is being made.
  • bone marrow reflects peripheral blood usually heavily infiltrated with lymphocytes.
  • immunophenotyping is essential to exclude reactive lymphocytosis and other lymphoid neoplasms.
  • cytogenetics to characterize the specific mutation can be helpful in assessing prognosis.
404
Q

what is the treatment of CLL?

A
  • combination therapy with fludarabine, cyclophosphamide and rituximab has become standard first-line therapy and can induce complete remission.
  • for older patients, chlorambucil usually reduces lymphocytosis, lymphadenopathy and splenomegaly to palliate the disease.
  • alemtuzumab is used in patients in whom there is disease progression after treatment with fludarabine.
  • new generation anti-CD20 monoclonal antibodies, such as ofatumumab and obinutuzumab, are also used in fludarabine- or alemtuzumab-refractory CLL or in combination with chlorambucil.
405
Q

what are examples of new generation anti-CD20 monoclonal antibodies used to treat CLL?

A

ofatumunab and obinutuzumab

- can be used in fludarabine or alemtuzumab-refractory CLL or in combination with chlorambucil

406
Q

what is the prognosis of CLL? what does it depend on?

A
  • the median survival from diagnosis is very variable and correlates closely with disease stage at diagnosis and cytogenetic findings, e.g. patients with either 11q or 17p deletions (the sites of two tumour suppressor genes) are at high risk of not responding to initial treatment and rapid progression.
  • in other patients there is near-normal life expectancy.
407
Q

what are the constituents of blood?

A
  • blood consists of red cells, white cells, platelets and plasma.
  • plasma is the liquid component of blood which contains soluble fibrinogen and in which the other components are suspended.
  • serum is what remains after the formation of the fibrin clot.
408
Q

what are stimulatory/inhibitory growth factors?

A

stimulatory

  • erythropoietin (red cells)
  • thrombopoietin (platelets)
  • neutrophils (G-CSF0)
  • interleukin-5 (eosinophils)

inhibitory
- TNF

409
Q

what are reticulocytes? what does the reticulocyte indicate/

A
  • reticulocytes are young red cells recently released from the bone marrow and still contain RNA.
  • they are larger than mature red cells and normally represent 0.5–2.5% of total circulating red blood cells.
  • the reticulocyte count gives a guide to the erythroid activity in the bone marrow and there is normally an increase with haemorrhage, haemolysis and after treatment with specific haematinics in deficiency states.
410
Q

what can cause an increased reticulocyte count?

A
  • haemorrhage
  • haemolysis
  • after treatment with specific haematinics in deficiency states
411
Q

what is the structure of Hb?

A
  • the principal function of haemoglobin (Hb) is to deliver oxygen to the tissues from the lungs.
  • Hb is a tetramer consisting of two pairs of globin polypeptide chains.
  • a haem group, consisting of a single molecule of protoporphyrin IX bound to a single ferrous ion (Fe2+) is linked covalently at a specific site to each globin chain.
  • oxygenation and deoxygenation of Hb occur at the haem iron.
412
Q

what are the normal values for MCV (fL)?

A

80-96

413
Q

what are the normal values for MCH (mean corpuscular Hb, pg)?

A

27-32

414
Q

what are the normal values for ESR (mm/h)^9?

A

<20

415
Q

what are factors that promote intestinal absorption?

A

gastric acid, iron deficiency and increased erythropoietic activity

416
Q

how is iron eliminated?

A
  • fixed at 1mg/day
  • occurs through shedding of skin and mucosal cells and excretion in sweat, urine and faeces
  • loss during menses in women
417
Q

what are the two forms of dietary iron?

A
  • non-haem iron forms the main part of dietary iron and is derived from fortified cereals and vegetables. it is dissolved in the low pH of the stomach and reduced from the ferric to the ferrous form by a brush border ferrireductase before transportation across the mucosal cells.
  • haem iron is derived from Hb and myoglobin in red or organ meats. haem iron is better absorbed than non-haem iron.
418
Q

where is non-haem iron derived from?

A

fortified cereals and vegetables

419
Q

how is non-haem iron absorbed?

A

it is dissolved in the low pH of the stomach and reduced from the ferric to the ferrous form by a brush border ferrireductase before transportation across the mucosal cells

420
Q

where is haem iron derived from?

A

Hb and myoglobin in red or organ meats

421
Q

what is a response to iron treatment characterised by?

A

a response to iron treatment is characterized by an increase in the reticulocyte count followed by an increase in Hb at a rate of about 10 g/L every week until the Hb concentration is normal

422
Q

what is the function of hepcidin?

A

hepcidin binds to the export transport protein, ferroportin, in the iron-absorbing cells in the duodenum, thereby causing its degradation, with a consequent reduction in the transport of iron from duodenal cells into the plasma

423
Q

what are clinical features of sideroblastic anaemia? what is its treatment?

A
  • sideroblastic anaemia is a rare disorder of haem synthesis characterized by a refractory anaemia with hypochromic cells in the peripheral blood and ring sideroblasts in the bone marrow.
  • ring sideroblasts are erythroblasts with iron deposited in mitochondria and reflect impaired utilization of iron delivered to the developing erythroblast.
  • it may be inherited or acquired (secondary to myelodysplasia, alcohol excess, lead toxicity, isoniazid).
  • treatment is to withdraw the causative agents and some cases respond to pyridoxine (vitamin B6).
424
Q

what is the daily requirement for B12?

A

1mg, which is easily supplied by balanced Western diet (containing 5-30μg daily)

425
Q

what liberates B12 from protein complexes in food? what does it then bind to?

A
  • liberated by gastric acid and pepsin

- binds to a vitamin B12-binding protein (R binder) derived from saliva

426
Q

what releases free B12? what happens to it?

A
  • released by pancreatic enzymes
  • becomes bound to intrinsic factor, which, along with H+ ions, is secreted from gastric parietal cells
  • complex is delivered to terminal ileum, where vitamin B12 is absorbed and transported to tissues
427
Q

what absorbs and transports B12 in the terminal ileum?

A

carrier protein transcobalamin II

428
Q

how is an oral dose of B12 absorbed?

A

about 1% of an oral dose of B12 is absorbed passively without the need for intrinsic factor, mainly through the duodenum and ileum

429
Q

what are the causes of vitamin B12 deficiency?

A
  • low dietary intake
  • impaired absorption
  • abnormal utilisation
430
Q

what can cause impaired absorption of vitamin B12 in the stomach?

A
  • pernicious anaemia
  • gastrectomy
  • congenital deficiency of intrinsic factor
431
Q

what can cause impaired absorption of vitamin B12 in the small bowel?

A
  • ileal disease or resection, e.g. Crohn’s disease
  • coeliac disease
  • tropical sprue
  • bacterial overgrowth
  • fish tapeworm (Diphyllobothrium latum)
432
Q

what can cause abnormal utilisation of vitamin B12 which can lead to deficiency?

A
  • congenital transcobalamin II deficiency (rare)

- nitrous oxide (inactivates B12)

433
Q

how long are body stores of folate sufficient for?

A

4 months

434
Q

what are causes of poor intake which lead to folate deficiency?

A
  • old age
  • poverty
  • alcohol excess
  • anorexia
435
Q

what are causes of malabsorption which lead to folate deficiency?

A
  • Coeliac disease
  • Crohn’s disease
  • tropical sprue
436
Q

what are causes of excess utilisation of folate which lead to folate deficiency?

A

physiological: pregnancy, lactation, prematurity
pathological: chronic haemolytic anaemia, malignant and inflammatory diseases, renal dialysis

437
Q

what are drugs that can cause folate deficiency?

A
  • phenytoin
  • trimethoprim
  • sulfasalazine
  • methotrexate
438
Q

how can folic acid be used to prevent neural tube defects?

A
  • to prevent first occurrence of neural tube defects, women should be advised to take folate supplements (at least 400 μg/day) before conception and during pregnancy.
  • larger doses (5 mg daily) are recommended for mothers who already have an infant with a neural tube defect.
439
Q

what are physiological causes of macrocytosis other than megaloblastic anaemia?

A
  • pregnancy

- newborn

440
Q

what are pathological causes of macrocytosis other than megaloblastic anaemia?

A
  • alcohol excess
  • liver disease
  • reticulocytosis (e.g. due to haemolysis)
  • hypothyroidism
  • haematological disorders: myelodysplastic syndrome, sideroblastic anaemia and aplastic anaemia
  • drugs: hydroxycarbamide (hydroxyurea) and azathioprine
  • cold agglutinins
441
Q

what are causes of pancytopenia?

A
  • aplastic anaemia
  • drugs
  • megaloblastic anaemia
  • bone marrow infiltration or replacement: lymphoma, acute leukaemia, myeloma, secondary carcinoma, myelofibrosis
  • hypersplenism
  • systemic lupus erythematosus
  • disseminated tuberculosis
  • paroxysmal nocturnal haemoglobinuria
  • overwhelming sepsis
442
Q

when should neutropenic sepsis be suspected in a neutropenic patient?

A

suspect neutropenic sepsis in a neutropenic patient (neutrophil count < 1 × 10^9/L) who is pyrexial or has new-onset confusion, tachycardia, hypotension, dyspnoea or hypothermia.

443
Q

what is the assessment of suspected neutropenic sepsis?

A

history and physical examination including mucous membranes, oropharynx (?thrush, erythema), surgical sites and intravenous lines

444
Q

what investigations are done for suspected neutropenic sepsis?

A
  • bloods: full blood count and differential white cell count, C-reactive protein, urea and electrolytes, liver biochemistry, clotting, blood cultures
  • radiology: chest X-ray. consider further imaging if localizing signs, e.g. computed tomography (CT) scan of abdomen and pelvis
  • microbiology: microscopy and culture of peripheral blood and taken from central lines, sputum, urine, stool (if diarrhoea)
445
Q

what are inherited causes of haemolytic anaemia?

A
  • red cell membrane defect
  • hereditary spherocytosis
  • hereditary elliptocytosis
  • Hb abnormalities
  • thalassaemia
  • sickle cell disease
  • metabolic defects
  • G6PD deficiency
  • pyruvate kinase deficiency
  • pyrimidine kinase deficiency
446
Q

what are types of deficiency that can cause haemolytic anaemia?

A
  • G6PD deficeincy
  • pyruvate kinase deficiency
  • pyrimidine kinase deficiency
447
Q

what are acquired causes of haemolytic anaemia?

A

immune

  • autoantibodies
  • alloimmune
  • drug-induced antibodies

non-immune

  • paroxysmal nocturnal haemoglobulinuria
  • mechanical destruction: microangiopathic
  • haemolytic anaemia, damaged artificial heart valves, march haemoglobulinuria
  • secondary to systemic disease, e.g. liver failure
  • infections
  • drugs/chemicals
  • hypersplenism
448
Q

what are features of haemolysis caused by increased red cell production?

A
  • increased reticulocytosis/polychromasia

- erythroid hyperplasia of bone marrow

449
Q

what are features of haemolysis caused by increased red cell breakdown?

A
  • anaemia
  • increased serum bilirubin
  • increased LDH
  • increased urobilinogen
  • positive urinary haemosiderin
  • decreased plasma haptoglobulin
450
Q

what is seen in a blood film in haemolysis?

A
  • abnormal red cells
  • spherocytes
  • bite cells, blister cells, Heinz bodies
  • fragmentation
451
Q

how can abnormal red cells on a blood film in haemolysis be further investigated, and what can be found?

A

haemoglobin electrophoresis

  • thalassaemia
  • sickle cell diseae
  • HB C, D, E, H
452
Q

what can cause a DCT positive result in spherocytes on a blood film in haemolysis?

A
  • drugs e.g. methyldopa, quinine, NSAIDs, interferon, ribavarin
  • autoimmune haemolytic anaemia: IgG warm or cold
453
Q

what can cause a DCT negative result in spherocytes on a blood film in haemolysis?

A
  • hereditary spherocytosis

- malaria, Clostridium welchii

454
Q

what can cause bite cells, blister cells and Heinz bodies on a blood film in haemolysis?

A

enzyme defects, e.g.:

  • decreased G6PD
  • decreased pyruvate kinase
  • decreased pyrimidine 5’ nucleotidase
455
Q

what can cause fragmentation on a blood film in haemolysis?

A

microangiopathic haemolytic anaemia e.g.:

  • eclampsia
  • haemolytic uraemic syndrome
  • thrombotic thrombocytopenic purpura
  • DIC
456
Q

what is hereditary elliptocytosis?

A

hereditary elliptocytosis is similar to spherocytosis, but the red cells are elliptical in shape. it is milder clinically and usually does not require splenectomy.

457
Q

what is the structure of HbS?

A

HbS = alpha2beta2

- substitution of valine for glutamic acid in position 6 of beta chain

458
Q

what is the structure of HbC?

A

HbC = alpha2beta2

- substitution of lysine for glutamic acid in position 6 of beta chain

459
Q

what is Hb SC disease?

A

inheritance of the HbS gene from one parent and HbC from the other parent gives rise to Hb SC disease, which tends to run a milder clinical course than sickle cell disease but with more thromboses

460
Q

what are complications of sickle cell anaemia requiring inpatient management?

A
  • pain – uncontrolled by non-opiate analgesia
  • swollen painful joints
  • acute sickle chest syndrome or pneumonia
  • mesenteric sickling and bowel ischaemia
  • splenic or hepatic sequestration
  • central nervous system deficit
  • cholecystitis (pigment stones)
  • cardiac arrhythmias
  • renal papillary necrosis resulting in colic or severe haematuria
  • hyphema (layer of red cells in anterior chamber of eye) or retinal detachment
  • priapism
461
Q

when are blood transfusions given in sickle cell anaemia?

A
  • after TIAs and stroke
  • acute chest syndrome
  • splenic sequestration crisis and aplastic crises
  • before elective operation
  • during pregnancy