• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/98

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

98 Cards in this Set

  • Front
  • Back
  • 3rd side (hint)

HIT


4T score


Points for different risk categories

Thrombocytopenia


2 50% drop, nadir > 20 (rarely drops lower than this


1 30-50% drop, or nadir 10-20



Timing of platelet fall


2. 5-10 days after start of exposure, or within 24 hours and has had a separate heparin exposure within 1 month


1. Possibly within 5-10 days but missing counts; onset >10 days; or within 24 hours and has a separate heparin exposure within 30-100 days




Thrombosis or other sequelae


2. Confirmed, or skin necrosis, or systemic reaction to iv injection


1. Suspected/progressive thrombosis, non-necrotising skin lesion



Other causes


2. Non apparent


1. Possible



3 or less: low risk. Dont lab test unless missing data has led to this score


4-5: intermediate risk


6 or more: high risk

Raised INR from warfarin (2x3 categories)

No bleeding


<4.5: nil


4.5-10: consider vit K if high bleeding risk


>10: Vit K. Consider prothrombinex



Bleeding


Minor: consider vit K if high bleeding risk or >4.5


Clinically significant: IV Vit K + prothrombinex


Life-threatening: IV vit K + prothrimbinex + FFP

HIT mechanism

Heparin normally binds with the cytokine, platelet-activating factor 4, forming a complex.


In HIT, IgG forms against this complex and binds it, which changes the Fc domain of these IgG, causing them to activate platelets, causing them to degranulate

(Not enough paf 4 me)

Reversal of dabigatran / apixaban / rivaroxaban

Dabigatran: idarucizumab. Rapidly reverses dabigatran. MAB. Seems to be available



Anti Xa: andexanet alpha is in phase 3. Not available

MDS subtypes

1. MDS with single lineage dysplasia with or without ringed sideroblasts (>15%)


- Blasts <5%


2. MDS with multi-lineage dysplasia with or without ringed sideroblasts (>15%)


- Blasts <5%


3. MDS with excess blasts


- Blasts 5-19%


4. Isolated del(5q) abnormality


5. Chronic myelomonocytic leukaemia (CMML)


- Blood monocytes >1, <20% blasts


6. Juvenile myelomonocytic leukaemia (JMML)


- RAS / NF-1 mutations

MDS diagnosis (4)

1. 1 or more cytopenias


- Anaemia is often macrocytic


- Cells are hypofunctional also (neutrophils are hypogranular), so may be disproportionately symptomatic to cell count


2. >10% dysplasia in a cell line (on BMAT)


3. <20% blasts (BMAT)


4. Characteristic cytogenetic findings

MDS prognostic score - name and 4 factors

IPSS score (low, intermediate, high)


Blasts (<5, 5-10, 11-20, 21-30)


Karyotype (good, intermediate, poor)


Cytopenias (0/1; 2/3)


Age

MDS treatment options


- Low risk (3)


- High risk (3)

Low risk patients - supportive cares


- Transfusions


- EPO in select patients


- Chelation in select patients (usually >20 transfusions)




High risk patients


- Young patients: stem cell transplant


- Other: azacitidine


- 5q syndrome: lenalidamide (strong response)

Most common cause of death in MDS

Infection


Then AML


Then haemorrhage

Treatment of HIT (3 aspects)

Stop heparin


Alternative anti coagulant


- Anti-Xa: lepirudin/argatroban


- Direct thrombin: danaparoid (?fondaparinux & bivalirudin also options?)


Start warfarin once platelet count improves and bridge. Continue for 1-3 months

ITP


2 types and associated ages


Most common antibody

Acute: more so children


Chronic: more so adults. Can relapse and remit with unpredictable timecourse


AntiGP2b3a

ITP acute treatment

Plt >30 and no/mild bleeding: observation


Plt >30 and symptoms: treat


Plt <30: treat




Prednisone 1-2mg/kg - good at inducing remission but relapse common


IVIG - quicker induction


Splenectomy - best difinitive treatment


Rituximab


Romiplostin (thrombopoietin receptor agonist) or eltrombopag (TPO mimetic) - mist be post-splenectomy, habing failed steroids/IVIG

ITP investigations

Film to exclude pseudothrombocytopenia (plt clumping) or platelet abnormalities (should be normal or mildly enlarged in ITP)


HIV, hepatitis C

Amyloidosis overview


3 major types


7 general possible features


Diagnosis

Types


Over 36 forms of amyloid


3 most common are


- AL (primary)


- AA (secondary):


- ATTR (cardiac only)


Other: dialysis-related




General possible features


Waxy skin


Enlarged muscles (tongue, deltoid)


Cardiac: heart failure, conduction problems


Hepatomegaly with greatly elevated ALP


Proteinuria/nephrotic syndrome (see renal slide)


Peripheral/autonomic neuropathy


Coagulopathy




Diagnosis


Biopsy


- If single organ involvement: biopsy that organ


- If multi-organ, biopsy fat pad (less bleeding risk)

AA amyloid

Most common form in resource-limited countries

Commonly complicates chronic inflammation (chronic infections or inflammatory diseases)




Fibrils of SAA protein (serum amyloid A), an acute phase reactant




Diagnosis: as per slide above


Other investigations:


- Raised CRP, ESR


- Raised serum SAA (but often not available)




Management: treat underlying condition

3 types of primary TMA


5 broad causes of secondary TMA


1 other atypical type to note

TTP (ADAMTS13)


Shiga-toxic associated HUS


Complement-mediated TMA (AKA atypical HUS)


Secondary TMA


- Malignancy


- Transplant


- Infection


- Drugs


- Pregnancy



Renal-limited TMA - when there's TMA on renal biopsy without systemic features (probably like C3 bloke from ICU)

Complement mediated TMA (atypical HUS)


Pathogenesis


Treatment (1)


- Side effect of

5-10% of HUS cases


Genetic mutations in complement regulatory proteins resulting in excessive complement activation. Can also be complement regular auto antibodies


Ecalizumab or revalizumab - block cleavage (activation) of C5 to C5a. Result is susceptible to infection (especially neisseria), ?because of lack of C5b and therefore membrane attack complex



Above is indicates in those where complemented mediated is the most likely cause for the TMA based on clinical features (see other card), while awaiting confirmation by complement testing

Shiga toxin-producing E coli HUS (STEC-HUS)


Pathophysiology


Effect on coagulation screen and D.dimer

Shiga toxin binds Gb3 found most in glomerular endothelium (and more in children than adults)


Results in platelet activation by unknown mechanism


Also inacticates ADAMTS13 (?but doesn't cause a low level)


Also causes complement activation



Coags/D.dimer normal (as opposed to DIC)



Diagnosis: clinically TMA with positive shiga toxin +/- negative ADAMTS13 deficiency



Management: supportive. Uptodate recommends eculizumab (C5 MAB) for those with severe neurological impairment. It doesnt recommend plasma exchange as no evidence

Causes of MAHA

Primary TMA (TTP, shiga toxin HUS, complement TMA)


DIC


Malignancy


Autoimmune conditions (SLE)


Antiphospholipid syndrome


Malignant hypertension


Valvular heart disease


Drugs


Pregnancy

MAHA and TMA definitions

MAHA is any non-immune haemolysis resulting in fragmentation of red blood cells intravascularly producing schistocytes. Some sites say that thrombocytopenia is also part of the definition, but others not so necessarily (especially since it can be caused by valve problems)


TMA is MAHA plus thrombocytopenia and organ damage. And it seems that pathologically it should show a characteristic endothelial injury

Similarities and differences between DIC and TMA

Both have thrombocytopenia


Both cause organ failure


Both cause MAHA with schistocytes and helmet cells (but not as bad in DIC)


Both can cause anaemia, but less common/severe in DIC


BP low in DIC, high in TMA



DIC: Everything is activated from platelets to coagulation to fibrinolysis. Clot is fibrin rich


Abnornal coags.


High D dimer.


Low fibrinogen (but can be normal in inflammatory conditions as it's an acute phase reactant)



TMA:Predominantly platelets alone are activated without so much coagulation and fibrinolysis. Clot is platelet rich/hyaline - rich (not fibrin rich)


Normal coags and ?D dimer

Paroxysmal nocturnal haemaglobinuria


Pathogenesis


Clinical features


Diagnosis


Treatment


Other condition it's associated with

Pathogenesis


Due to an acquired mutation in a glycolipid (glycosylphosphatidylinositol) needed to anchor other surface proteins (complement inhibitors), which protect red cells from the complement system.


Specifically either DAT (CD55) or protectin (CD59)


When these are lost, complement destroys cells intravascularly (CD59 prevents C9 from forming the membrane attack complex)


The gene that encodes it is the PIGA gene


The mutation has to happens in a haematopoetic stem cell for it to affect other cells




Clinical features


1. Non-immune (normal direct coombs), non-microangiopathic haemolytic anaemia


2. Venous thrombosis (especially in uncommon areas like DVST, budd-chiari, portal vein thrombosis, mesenteric thrombosis


3. Characteristic red urine in the morning (due to urine being more concentrated then) is uncommon


4. Abdo pain, erectile dysfunction, fatigue, oesophageal spasm - due to vasoconstriction from loss of NO from free Hb which scavenges it


5. Bone marrow failure if aplastic anaemia also present


Pathogenesis of thrombosis not known




Diagnosis


Suspected based on above


Confirmed by flow cytometry for CD55 (DAF) and CD59 (protectin)




Treatment


1. Mild symptoms: watchful waiting


2. Bad symptoms without aplastic anaemia/MDS: C5 inhibitor (ravalizumab or ecalizumab)


3. Bad symptoms with aplastic anaemia or MDS: consider SCT

When to suspect complement mediated TMA over other TMAs


Confirmation of the diagnosis

Renal failure-predominant TMA without recent diarrhoeal illness or culprit medications


Family Hx of the same



TTP is more neurological


STEC-HUS is renal predominant but preceded by the diarrhoeal illness


Medication induced is self explanatory



Confirmed by complement testing

HIT 2 lab tests and their relative sensitivity/specificity

ELISA to detect antibodies against the heparin/platelet-activating factor 4 complex (ELISA tubes coated with PAF4/heparin complex. Patient's serum added, then washed off. Enzume-linked animal ABs against human ABs added to the assay which bind any human ABs. Then a substrate is added which reacts to the linked enzym to create a colour change)


- Doesn't tell you whether they're actually causing platelet activation tho




Functional assays, where a platelet-rich assay is incubated with the patient's serum to see if the platelets activate or not




Both are sensitive, the functional is more specific


But functional takes longer and may not be available




Uptodate suggests doing ELISA first only doing the second if equivocal/in a certain range.

Vaccination-induced thrombotic thrombocytopenia (VITT)




Pathophys


Clinical features / scores


Lab testing


Management

Vaccine induces antibodies against PF4 (platelet-activating factor 4), the Fc domain of which causes platelet activation.


Similar to HIT, except that the antibodies are to PF4 alone rather than the heparin/PF4 complex. Also very high d-dimer


Causes a similar syndrome to HIT, where thrombosis is the primary issue rather than the thrombocytopenia




Score is a modified 4T score with


Timing post vaccine


Degree of thrombocytopenia


Presence of thrombosis


Ruling out other causes




Lab test is an ELIZA for PF4 antibodies, or a functional essay looking for platelet degranulation. Similar to HIT




Management:


- Anticoagulation in everyone, prefer to use non-heparin based


- IVIG in everyone


- Plasma exchange in refractory cases

ITP treatment


Critical bleeding


Severe bleeding


Minor bleeding (plt <50) or no bleeding and plt <20)

IV steroids + IVIG


IV steroids or IVIG


Consider IVIG or steroids, especially if plt <10



Above should be a single course, with a taper in the case of steroids. Then cease and observe

ITP secondary/further options

If recurrence or lack of response to initial steroids / IVIG:


Splenectomy


If not a candidate for splenectomy (surg risk or infection risk) or pt prefers to avoid it, or splenectomy is ineffective, next line is rituximab as a single course over 4 weeks

Waldenstrom Macroglobulinaemia


What is it


Clinical features - 3-3-3:


- Syndrome it causes and associated triad


- 3 other features (2 are in a similar group)


- 3 differences from myeloma


5 key investigation findings


Treatment

What is it


B cell neoplasm (in the bone marrow) that secretes monoclonal IgM (IgM paraproteinaemia)


Macroglobulin as in large immunoglobulin (i.e. IgM pentamer)




Clinical features


Hyperviscosity syndrome


- Neuro problems


- Visual problems


- Bleeding





Autoimmune reactions:


- AI haemolytic anaemia - cold subtype


- Autoimmune neuropathy (AB agaibst myelin-associated glycoproteins)


- Raynaud's syndrome from cyroglobulinaemia





Difference from myeloma


- Rarely get bone pain


- Enlarged lymph nodes


- Hepatosplenomegaly (from the IgM?)




Investigations


Normocytic normochromic anaemia with positive coombs test


Film: rouleaux


SPEP: M spike. On immunofluorescence : IgM


High serum viscosity




Bone marrow: >10% lymphocytic cells




Treatment


Chemo


Plasma exchange if hyperviscosity syndrome


IVIG

Hereditary haemochromatosis


3 most common gene mutations, and which 2 combos mostly cause symptomatic disease


Ethnicity


6 main organ effects


Diagnosis


Features reversible with treatment


Management


Screening of relatives

C282Y is the only one that can cause iron overload - either homozygous C282Y, less commonly C282Y/H63D heterozygote (specifically that heterozygote - C282Y when paired with a non-H63D has no risk of iron overload).


Although deltamed says that his combo doesn't result in clinical significant haemochromatosis


But not everyone homozygote has clinical iron overload


Northern European


Results in hepcidin deficiency/resistance





Cardiac: heart failure, arrythmias


Liver: cirrhosis/HCC


Pancreas: diabetes


Joints: arthropathy


Skin: pigmentation / bronze tan


Pituitary - hypogonadism (erectile dysfunction)




Diagnosis



High ferritin and transferrin saturation ,Low total iron binding capacity


Then genetic testing


Can do MRI to look for black liver, white pancreas




Liver biopsy (if done): Perl's stain





Heart failure and skin pigmentation are reversible with treatment





Management


Phlebotomy if ferritin >1000 or end organ damage.


Target ferritin in low-normal range (~50)



Screen first degree relatives of homozygotes

Gilbert syndrome


Cause


Inheritance


Diagnostic test (which isn't actually needed)

Mutation in promotor of gene that conjugates (via glucuronidation) bilirubin



Results in unconjugated hyperbilirubinaemia, especially during times of infection, exercise and fasting



Diagnosed by persistent isolated high unconjucated bilirubin



Can be confirmed by administration of nicotinic acid



Autosomal recessive

Essential thrombocytosis


Age


Cause including 3 mutations in order


3 clinical symptoms


3 complications


1 clinical sign


3 main investigations and features


Diagnostic criteria

Middle to old (mean 50)


Excessive clonal platelet production


Somatic mutation (mostly sporadic) in


- JAK2 60%


- CALR 20%


- MPL 5%


Half are incidentally found


Signs


- Neuro: visual change, headache, dizziness


- Complications: bleeding, thrombosis, 1st trimester fetal loss


- Splenomegaly without hepatomegaly or lymphadenopathy (think pure platelet consumption)


Film: playelet anisocytosis (variable size)


Bone marrow:


- Normal to moderate hypercellularity


- Prominent large megakarycytes with hyperlobulated nuclei


- Shouldnt be any highly displastic features , significant fibrosis or increased myeloblasts - suggests something else


Genotype: mutations as above (but 10% have a different one). This differentiates from reactive thrombocytosis


Test for BCR-ABL, and von willebrand disease (if plt >1000)


Jak2 mutation more likely ro transform into polycythemia RV with high RBC and WCC


Diagnosis


Plt count over 450


BMAT showing the abovr


Rule out BCR-ABL CMR and other alternatives


A mutation is present

Essential thrombocytosis


2 things it can transform into


Management

AML and myelofibrosis (but most end up having a normal life expectancy)


Use risk score


Intermed-high risk: platelet lowering agent and asprin. If hx of VTE, use anticoagulation instead of aspirin


Low risk: Aspirin (which also helos symptoms) or observation


Main plt lowering agent of choice is hydroxyurea

CLL


What is it


Characteristic cells on smear


How to diagnose when suspected off FBC/film

Leukaemia of well differentiated cells, almost all are B cells




Smear


Smudge cells




Diagnosis


1. Suspect from high lymphocyte count (with smudge cells on film)


2. Confirm with immunophenotyping by flow cytometry


- CD19, CD20 (weak), CD23 (B cell markers)


- CD5 (mostly T cell antigen)


- Very low levels of surface membrane immunoglobulin (SmIg) that is restricted (is only either light or heavy chain)


3. BMAT not necessary unless unexplained cytopenias


4. SLL is diagnosed with LN biopsy (because they don't have the lymphocytosis to point you in that direction)

CLL indications for treatment

1. B symptoms impacting on life


2. Massive/symptomatic splenomegaly, hepatomegaly or lymphadenopathy


3. Symptomatic extranodal disease


4. Progressive marrow failure: rough indications: anaemia <100 or theombocytopenia <100 (when excluded other causes)


5. Lymphocyte doubling rate less than 6 months, or >50% over 2 months



Treatment: FCR (fludarabine, cyclophosphamide, rituximab)

CLL treatment options

Age <60: FCR


17p or P53 deletion: ibrutinib


Frail elderly: Obinutuzumab (type 2 CD20 AB with enhancedantibody-dependent cytotoxicity) + chlorambucil



Relapsed disease:


- Ibrutinib


- Venetoclax: BCL2 inhibitor (aka BH3 mimetic, which is a natural BCL2 inhibitor that induces apoptosis in times of stress), which is an anti-apoptosis gene. Need graduated oral dosing to avoid tumour lysis syndrome

Monoclonal B cell lymphocytosis


What distinguishes it from CLL

Monoclonal lymphocyte count <5


No other clinical features of anything (B symptoms, lymphadenopathy, hepatosplenomegaly, cytopenias)


And no secondary causes (infection, auto-immune)




Do the usual flow cytometry as for CLL, to classify it into:


- CLL-type: CD5+, CD19+, CD23+, SmIg +ve, CD20 dim/-ve


- Atypical: as above but CD20 strongly positive. Nede to exclude mantle cell lympoma


- Non-CLL type: different. Need to exclude marginal zone lymphoma




Need to exclude atypical and non-CLL from mantel cell lympoma




Rate of progression to CLL: 1-2% per year

Ibrutinib side-effects

- Redistribution lymphocytosis (not so much aside-effect): initial worsening of lymphocytosis- Significant bruising (withhold 7 days prior tomajor surgery)


- Diarrhoea


- AF

Fludarabine mechanism

Purine analogue

Acute intermittent porphyria - 3 classifications

Inherited disorders of haem synthesis - accumulation of porphyrin precursors


Except porphyria cutanea tarda which is due to an acquired inhibitor



8 enzymes in the pathway



Categorized into 3 ways they present clinically, with the 3 most common nicely slotting each into a different one of the 3





Neuro visceral or 'hepatic', although it doesn't seem to actually affect the liver? Symptoms are due to neuropathic effect


Poster boy: acute intermittent porphyria (2nd)


- Abdo pain is central symptom (neuropathic visceral pain) with normal exam findings


- Vomiting, constipation


- Psychosis, agitation, seizure, hyponatraemia (for which these symptoms are mistaken as a result of)


- Peripheral neuropathy


- In AIP, attacks occur acutely, precipitated by certain drugs


- Diagnosis: urine porphobilinogen (remains elevated between attacks)





Blistering skin


Poster boy: porphyria cutanea tarda (1st)


- Blisters/scars/abnormal pigment on sun exposed areas


- Triggers: haemochromatosis, hep C, HIV, liver damage


- Diagnosis: urine porphyrins





Non-blistering skin


Poster boy: erythropoietic protoporphyria (3rd)


Pain/swelling immediately after sun exposure, but no lasting blisters or scars


Diagnosis: erythrocyte total protoporphyrin

Acute intermittent porphyria treatment

4 things to take at a bar - benzos, alcohol, selfie, OCP


Avoid precipitants.




Barbituates


Benzos


Alcohol


OCP


Sulphonamides




Acute attacks: Haemin

Which thrombophilias you can test for during acute thrombosis

Prothrombin gene mutation


Factor 5 leiden (both of these are mutations so can test any time)


APLS (but can be false pos, so repeat in 12 weeks)


Cant test for protein C/S



Leiden/prothrombin not affecting by anti coagulant


Cardiolipin and b2micro arent




Protein C/S

Blood film terms


Pencil cells


Howell-Jolly bodies


Spur cells/acanthocytes

Pencil cells: IDA




Oval cells: megaloblastic




Hypersegmented neutrophils: B12 deficiency




Target cells: thalassaemia, hyposplenism, liver failure




Rouleaux




Auer rods: APML. Also, AML




Helmut cells: schistocytest




Spherocytes: AIHA or hereditary spherocytosis




Note megaloblastic means large size relative to what you'd expect for nuclear material




Teardrop cells: myelofibrosis (amongst other things)




Heinz bodies: G6PD, A thalassaemia




Blister cells - white blister on the edge of the cell (G6PD)


Bite cells - the cell after the blister is popped or removed (G6PD)




Howell-Jolly: hyposplenism




Spur cells (severe acanthocytes, meaning very spiculated): haemolysis from advanced cirrhosis

Clotting cascade


Which do APTT and INR provide info on

Extrinsic (loner) is just tissue factor activating 7




Intrinsic 12 - 11 - 9




Factor 8 helps 9 -> 10


Factor 5 helps 10 -> 2




Protein C/S inhibit 8 and 5




INR - extrinsic (includes 7) - tissue is extrinsic


APTT - intrinsic




Warfarin mostly affects extrinsic, even though it also depletes factors 9 from intrinsic, because factor 7 has the shortest half life, so is the first to run out (according to google)

Thymoma associated with

Myasthenia Gravis


Red cell aplasia


Dermatomyositis


SLE


SIADH

Methaemaglobinaemia


What is it


Features


Common medication triggers

What is it


Haem has oxidised form of iron (Fe3+ instead of Fe2+) – unable to bind oxygen





Features


1. Usual signs of hypoxia (cyanosis, dyspnoea, endorgan effects)


2. Pulse oximeter sats are low - normally 85-90% regardless of actual Hb sats as it generally mucks up the interpretation


- Doesn't change with supplementation oxygen


3. ABG PaO2 and sats are normal (ABG sats are based on the PaO2?)


4. Blood: chocolate brown hue




Causes


1. Congenital


- Fault in enzyme that reduces the haemoglobin


2. Drugs


- Hydralazine, sulfur based drugs, nitrates




Management


Acquired: methylene blue. Ascorbic acid second line

Findings of methaemaglboin vs carbon monoxide poisoning vs cyanide

Methaemaglobin


- Low sats, normal pO2




CO poisoning


- Low or falsely normal sats (depending on oximeter), low pO2




Cyanide poisoning


- Normal pO2 and sats

Thymoma associated with

Myasthenia Gravis


Red cell aplasia


Dermatomyositis


SLE


SIADH

CML diagnosis

Elevated WCC/neutrophilia - low leukocyte alkaline phosphatase distinguises from reactive leukocytosis


Immature granulocyes with myelocyte bulge


Mild anaemia, raised platelets, raised basophils, raised eosinophils



Confirm by demonstration of Philadelphia chromosome by conventional cytogenetics or demonstration of mRNA for BCR-ABL

Causes of hyposplenism and hypersplenism

Hyposplenism


Splenectomy


Sickle cell syndrome


Coeliac disease




Hypersplenism


- Cirrhosis

Polycythaemia diagnosis


3 major


1 minor if all of above not met

3 major criteria


1. Persistently raised Hb (on repeated test) or Hct


2. JAK2 mutation (V617F or exon 12)


3. Bone marrow: hypercellularity with panmyelosis(trilineage growth), including pleomorphic megakaryocytes




Minor criterion: low EPO (only done if not all of abovemet)

Polcythaemia management

For all


- Phlebotomy for all, aiming haematocrit <0.45(reduces thrombosis)


- Aspirin


– at least daily (twice daily if prevvascular event or microvascular symptoms or high CVD risk)




High risk (previous thrombosis / age >60 / plt>1500/WCC >15 / uncontrolled systemic symptoms / increasing splenomegaly)


- Previous arterial thrombosis: BD aspirin


- Previous venous thombosis: anti-coagulation(warfarin preferred as insufficient evidence for DOAC, but some use)


- Cytoreductive therapy: hydroxyurea (akahydroxcarbamide) or interferon

Translocations


Burkitt


CML


Mantle cell


Follicular


APML

8 - 14 (B looks like 8) - C-myc


9 - 22


11 - 14


Follicular: 14 - 18 (fourlicular): BCL-2 (anti-apoptosis) with Ig heavy chain promotor


15 - 17

Viruses that cause lymphomas


Connective tisse disease


Endocrine causes

EBV - hodgkin lymphoma and burkitt NHL


HIV - Burkitt


Sjogrens: non-hodgkin




Hashimoto: non-hodgkin thyroid lymphoma

Causes of microcytic anaemia

Common:


IDA


Thalassaemia




Less common:


Anaemia of chronic disease


Siderblastic anaemia


Hyperthyroidism


Heavy metal poisoning

Key regulator of iron metabolism

Hepcidin - released from liver, acts on intestinal epithelium to down regulate ferroportin, preventing release of iron-bound transferrin into circulation

Also traps iron in macrophages




Correlates with ferritin levels - high in iron overload, low in iron deficiency

Drugs that cause macrocytic anaemia

Anti-folate: trimethoprim, methotrexate, pentamidine




DNA-synthesis: azathioprine, hydroxyurea,

B12 deficiency


3 findings on FBC


Finding on film


3 other organ effects apart from blood


Most common cause and 2 blood tests for this

Pancytopenia


Hypersegmented neutrophils, irregularly-shaped cells. Severe cases can have haemolysis




Subacute combined degeneration of the cord (demyelination) - this is the reason to replace first before folate


Glossitis + angular stomatitis


Neuropsych symptoms




Pernicious anaemia


Anti-intrinsic factor ABs - specific, not sensitive


Anti-parietal cell ABs - sensitive but not specific (remember because parietal cells do other things)

Causes of fragmented red cells

MAHA - TTP, HUS, complement associated TMA


HELPP


Malignant HTN


DIC


Valvular heart disease


Pre-eclampsia


Direct damage by heat/toxins

Cut off of ADAMTS13 for TTP

<5%

4 causes of intravascular haemolysis

MAHA


G6PD


Paroxysmal nocturnal haemoglobinuria


Paroxysmal cold haemoglobinuria

Investigations for someone with iron deficiency anaemia that:


1. Is persistently iron deficiency despite ORAL supplementation


2. Is persistently microcytic anaemic despite normalisation of iron stores

Rule out coeliac


Rule out thalassaemia

Sickle cell anaemia treatment (3)

Hydroxyurea (increased HbH)


Red cell exchange


Vaccinations if hyposplenism

Things causing prolongation of:




Isolated APTT (4)


Isolated INR (2)


Both (4)

Isolated APTT
Heparin/clexane


Lupus anticoagulant


Deficiencies of the intrinsic factors (12, 11, 9, 8)


Von Willebrand disease




Isolated INR


Vit K deficiency/antagonist


Factor 7 deficiency




Both prolonged


Liver failure (all factors deficient)


Fibrinogen deficiency (DIC)


Deficiencies of common factors (10, 2, 5)


Inhibitors of common factors (direct thrombin / 10a)

Which of INR, APTT, thrombin time to the NOACs prolong


% renal clearance


% protein bound


Metabolism enzyme effects


Severe bleeding

Dabigatran


- APTT, TCT prolonged


- INR dose dependent increase


- 80% renal. 35% protein bound


- P-GP substrate


- Bleeding: idaracizumab, dialysis




Rivaroxaban


- APTT: dose-dependent prolongation


- INR: sometimes. TCT no


- 1/3 renal clearance, 95% protein-bound


- Bleeding: consider FFP




Apixaban


- APTT: dose-dependent


- INR & TCT both no


- 1/4 renal, 87% protein bound


- Bleeding consider FFP

Transfusions reactions - pathophys and clinical features

FNHTR


- 1-6 hours: fever with no haemolysis


- Cytokines from donor WBCs (the 3 macrophage cytokines) - cytokines released during storage, so increased storage times = higher risk


- Prevent by leukodepletion




Acute haemolytic


- Within 15 mins. Fever, flank pain, bleeding


- Do a DAT


- Pathophys: ABO / rhesus incompatibility




TACO


- Pulmonary infiltrates with hypertension and no fever




TRALI


- Pulmonary infiltrates with fever and hypotension


- Pre-transplant stress activates lung PMNs. Donor anti-HLA ABs attack primed PMNs




Sepsis


- More likely in platelets




Urticaria / anaphylaxis


- Recipient IgE to donor proteins (e.g. in IgA deficiency)


- Prevent by washing




TA-GVHD


- 2 days to 6 weeks


- Pancytopenia, diarrhoea, rash, liver failure


- Prevent with irradiation

Blood transfusions - purpose of the following:



Radiation


Red cell washing




Main cause of anaphylaxis to red cell transfusions

Radiation: depletes white cells, reduces chance of graft vs host disease. Used in immunocompromised patients.




Red cell washing: removes immunoglobulin. Reduces the chance of anaphylaxis in patients with IgA deficiency




Loucodepletion: routinely performed. Reduces chance of FNHTRs and CMV transmission




Anaphylaxis is from a foreign substance in the donor bloods - e.g. IgA in IgA deficient people. Other example is haptoglobin-deficiency

Main organs involved in graft vs host disease


Timecourse




Main situations that it occurs in

Skin, liver, gut and bone marrow (in patients that still have a bone marrow)


2 days to 6 weeks




Blood transfusions to close relatives


Allogenic stem cell transplants


Usually, the host's WBCs kill any transfused white cells before they can engraft. In the above 2 situations, this doesnt occur (situation 1 - relatives have similar HLA and not recognised as foreign; situation 2 - host has no immune system with which to destroy donated white cells)




Transfusion associated GVHD is 90% fatal

Melphalan mechanism


Side effects

Alkylates guanine



Nausea


Myelosuppression


Pulmonary fibrosis

Lenalidomide


Mechanism


Side effects


Uses

Mechanism


1. Inhibits angiogenesis


2. Induces apoptosis


3. Immunomodulation



Use: myeloma (not exhaustive)

2 Major complications of CAR T cell therapy



3 others

Cytokine release syndrome


- Fever, hypotension, hypoxia (think TRALI)


- IL-6 key role in pathogenesis


- Treat with tocalizumab +/- steroids




ICANS - Immune-effector Cell Associated Neurotoxicity Syndrome


- Neurological symptoms: headache, confusion, ALOC, seizures


- Day 7 median onset. Commonly occurs after CRS


- IL-1beta key role in pathogenesis. Disturbed blood brain barrier


- Treat with dexamethasone/methylpred




Tumour lysis syndrome


B cell aplasia (B cells never recover)


Relapse of leukaemia

TTP treatment options in rural centre

1st line is PLEX + pred


+/- ritux


+/- caplacizumab for severe disease (anti-VWF, only new)




If PLEX not available


Pred should be


Can also use FFP to replace the ADAMTS13

APML


Diagnosis


Complication


Management


Management complication

Film


Prominent granulation with Auer rods




Diagnosis


Rapid diagnosis with PML-PARA PCR (protein produced from the below translocation)


or


FISH: t(15;17)


Later confirmed with BMAT




Complication


DIC




Management


DIC control: platelet transfusion / fibrinogen


Disease modifying - 'differentiation' therapy:


- ATRA (modified vitamin A derivative), which acts on retinol (vit A) receptor to induce differentiation of the blasts into more mature myeloid cells


- Arsenic




Complication of treatment


Differentiation syndrome


- New differentiated promyelocytes abnormaly bind endothelium (particular lung)


- Fever + TACO-like syndrome (oedema, lung infiltrates, hypoxia) + renal/liver dysfunction


- Treatment: dexamethasone +/- delay chemo

MDS most frequently mutated genes




Abormality that has treatment significance

Most commonly mutated


- SF3B1


- TET2


- SRSF2


- ASXL1




Treatment significant


- 5q deletion: treat with leflunamide

Von Willebrand disease


Inheritance


3 types


4 investigations


Management

Inheritance


Autosomal dominant (think, it's the most common inherited bleeding disorder, so dominant makes sense)




Types


1. Low (antigen) levels (75% of cases)


2. Normal levels, low function (10-20%)


- Low activity:antigen ratio


3. Absent levels (rare)




Clinical features


Platelet pattern bleeding




Investigations


Plt: normal. Mildly low in 2


APTT mildly raised due to low factor 8 (VWF carries this)


VWF antigen levels AND activity




Management


Severe bleeding: recombinant VWF


TXA as adjunct


DDAVP: triggers release of stored VWF from epithelial cells (not useful in type 2)


- Small effect, only works in some people

Hodgkin lymphoma


Age


2 random symptoms


Characteristic cells


Prognosis relative to NHL


Diagnosis (2)

Age: 20s and 60s




2 random symptoms


- Itchy


- Lymph pain after alcohol




B symptoms are less common and imply worse prognosis




Diagnosis


Flow cytometry for CD15 and CD30


And I presume biopsy

Hodkin lymphoma 4 types


Which is most common


Which has worst prognosis and best prognosis


Which 2 have particular blood film features

Nodular sclerosing (70%) - lacunar cells


Mixed cellularity (20%) - large number of reed-sternberg


Lymphocyte predominant (5) - best prognosis


Lymphocyte deplete (rare) - worst prognosis

Classical hodgkin lymphoma staging

1. Single LN group


2. >1 LN group, one side of diaphragm


3. LN groups both sides of diaphragm


4. Extranodal or non-contiguous involvement

Hodgkin lymphoma 6 poor prognostic factors

Male


Age >45


Stage 4




Hb <105


Alb <40


WCC >15, or lymphocytes <8% of WCC (i.e. lymphocyte deplete form?)

Hodgkin lymphoma management

Combination chemo + radiotherapy


- Usually ABVD (i.e. DBVD – doxorubicin, bleomycin,vinblastine, dacarbazine)


- BEACOPP is an alternative for advanced disease


- Brentuximab or pembrolizumab for relapseddisease

MGUS risk of progression to myeloma

1-2% per year (2% if restricted light chain)

Haemophilia A and B


Diagnosis

Inheritance


X-linked recessive


1/3 are de novo mutations




Diagnosis


Gene mutation, or low factor 8/9 level


- Mild: 5-40% of normal factor level


- Mod: 1-5%


- Severe: <1%




Exclude Von Willeband disease as a cause (for haemophilia A)




Management


Prophylaxis




Acute bleed


Give factor 8/9, aiming for


- Severe bleed: 80-100% of normal level


- Minor bleeds: aim lower

Haemophilia - management of acute bleed

Important point: when given the recombinant replacementfactor, the patient can develop antibodies to it, since he doesn’t normallyhave it, so it’s not ‘self’ No factor antibodies present- Give recombinant factor 8 or 9- Target activity: 80-100% of normal in severebleeding, lower in less severe bleeding or pre-emptive surgery (50% in haemarthrosis) Factor antibodies present – options:- FEIBA (factor eight bypassing products): bunchof activated factors that allow direct activation of factor 10- Factor 7a – directly activates factor 10 If unknown- Uptodate suggests giving recombinant factorfirst, but if activity is not coming up, then give bypassing products as above

Haemophilia prophylaxis

- Low risk of bleeding: individualised. Can begiven, or given at certain times or not given- Mod-high risk – options:1. Emicizumab: MAB to factor 9 that activates it –i.e. the MAB does the role of factor 82. Previously factor 8 was given, but commonlyantibodies were developed against it (since the patient never has had factor 8,so it’s not ‘self’). When antibodies were present, activated factor 7 was givento activate the other pathways Cryoprecipitate can be used if factors aren’t available

How to remember which thrombophilia screens can be done during acute VTE, and on anticoagulation


4 are good for all


2 are good for one but not the other


1 is bad for all

- 4 are good for all: 2 gene mutations (F5L,prothrombin), 2 APLS with antibody in their names


- 2 are good for 1 thing only: lupus “anticoagulant”– bad for “anticoagulants”, protein C/S – okay for S/C anticoagulants


- 1 is bad for all: AT (All Trash)

Follicular lymphoma


Translocation and association mutated protein


What is the function of this protein


OVerall outlook of follicular

14-18 (fourlicular)


BCL-2 with Ig heavy chain promotor: inhibits apoptosis


Indolent lymphoma




2nd most common NHL




Treatment


- Stage 1: radiotherapy (can be curative)


- Stage 3-4: focus on alleviating symptoms as disease has 20 year survival

Universal donor for plasma products

Universal donor is AB




Opposite to PRB transfusions

Dilute Russell Viper venom test

Detects lupus anticoagulants




The venom activates factor 10 directly, in the presence of phospholipid




Anti phospholipid antibodies (especially anti beta2 glycoprotein) interfer with the phospholipids, and so prolong the clotting time




Which kind of doesn't make sense because antiphospholipids are supposed to be pro-coagulant

AML


Pathogenesis


3 common mutations

Pathogenesis


Mutations lead to a leukaemic stem cell, which gives riseto blasts. The stem cell itself has a relatively small number of mutationscompared to other cancers, and is fairly quiescent. But the blasts that itgives rise to rapidly multiply. Chemo often can kill the blasts easily, butdoesn’t always get rid of the leukaemic stem cell.


Initially, it’s oligoclonal. Chemo may knock out one ormore clonal populations, but another may persist




3 common mutations


FLT3 (receptor tyrosine kinase)


NPM1


DNMT3a

AML


4 risk factors

1. Other haematological disease


2. Previous chemo or exposure to radiation/benzene


3. Age >65 (significantly increases after thisthreshold)


4. Genetic disorders (chromosomal)

AML 5 investigations

Film: myeloid blasts with auer rods and phi bodies


- Can be difficult to differentiate visibly fromALL blasts, so perform flow




Flow cytometry


- AML: myeloperoxidase positive (myel in both)


- ALL: TdT (terminal deoxynucelotidal transferase)positive




BMAT


- >20% blasts




Cytogenetics




FLIT3 (rapid PCR)

AML diagnosis 4 criteria

Confirmed by:


1. BMAT: >20% blasts


2. Flow cytometry: surface proteins consistent withAML


3. Genetics (genetic mutations and chromosomalabnormalities on karyotyping)


4. Clinicalsyndrome consistent

AML management

Induction chemotherapy


- For suitable candidates only – purpose is toachieve initial remission, to then assess options for further management


- “7+3”. 7 days of cytarabine then 3 days of an anthracycline(daunorubicin or idarubicin)- If FLIT3 positive, add midostaurin




If survive above, repeat BMAT. Remission = blasts <5%




Not a candidate for induction chemoCharacteristics


- Age >75


- Age >60 and ECOG 2, EF <50%, Cr Cl <45or DLCO <65%




Options


- Palliation


- Low dose chemo: cytarabine +/- venetoclax; orazacitadine

AML post-induction assessment including mutations

Favourable


NPM1 without FLT3




Intermediate






Unfavourable


Secondary AML
Remission not achieved


Wild type NPM1, with ?mutated FLT3

Cryoprecipitate contains


Use

Fibrinogen


VWF


Factor 8, 13




Use: fibrinogen replacement (DIC)




Only use in haemophilia A and von willebrand disease if nothing else available

Prothrombinex contains


Indication

Factors 2, 9, 10


Indications: warfarin reversal

FFP contains


Indication

All factors + fibrinogen




Use: anything, but higher chance of transfusion reactions

Systemic Mastocytosis


Mutation

Kit (receptor that causes mast cell proliferation) - mutated in 90% of SM


It is the receptor for 'stem cell factor'




(Mast kit for your sailing boat)

What is the CD of haematopoietic stem cells in transplantation

CD34