Hemostasis Flashcards

1
Q

Identify the elements that compose the hemostatic system. Understand the basic paradigm for coagulation factor activation

A

Components:

Coagulation factors (13 known)

Platelets

Endothelial wall (site of injury)

General idea: Endothelial wall becomes damaged, exposing collagen, tissue factor, and von Willebrand factor. (vWF, if you’re interested, is produced constitutively in endothelia and binds platelets once the endothelium is damaged. Also binds Factor VIII and keeps it viable.)

Platelets bind to the site of injury and change configuration, exposing certain proteins that attract fibrinogen and vWF (thus binding more platelets). They also release granules containing thromboxane (leading to vasoconstriction of damaged vessel), vWF and ADP (to bind and activate other platelets), various clotting factors, etc. They also serve as a ground on which clotting factors can easily activate.

Note that the endothelium is normally actively preventing coagulation; prevents platelet aggregation, promotes clot breakdown. As with so many other things, it’s a balancing act. When the endothelium is breached, the balance shifts (towards pro-coagulation) until it’s resolved.

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

Describe the blood clotting pathway. List which components of the system are vitamin K dependent factors. Distinguish the extrinsic and intrinsic pathways and describe the screening tests to measure both (PT and APTT, respectively)

A

A few notes about the components of the clotting pathway:
Several clotting factors are called by name, most are called by numeral. An “a” after the numeral indicates that the factor has been activated and is enzymatic.
By name: Prothrombin (factor II), Fibrinogen (factor I). These, when activated, are called thrombin and fibrin respectively.
By numeral: Factors III-XIII.
Several clotting factors are large enzymes that serve to orient the others.
These are: Factors V and VIII. Tissue factor also serves this role.
Most of the rest of the clotting factors are proenzyme serine proteases which, when cleaved and activated, cleave and activate each other.
These are: Factors II, VII, IX, X, XI, and XII.
The others seem to have particular, difficult-to-generalize-about roles. For example, Factor XIII cross-links fibrin fibers to make a ‘hard clot.’
Some clotting factors require carboxylation in the liver by vitamin K (recall from the MCAT that it’s a fat-soluble vitamin stored in the liver). If there’s no vitamin K available or there’s a lot of liver damage, these factors may not function correctly. Note that she emphasized this a lot in lecture.
Vitamin K-dependent factors: Factors II, VII, IX, and X.
Note that several important anti-clotting factors are also vitamin K-dependent: proteins C and S.
So the end goal of clotting is to produce, cross-link, and harden a mat of fibrin fibers to plug the endothelial damage. There are two pathways, the extrinsic and the intrinsic, to get things set up to do this; both of them end by activating Factor X to feed into the same clot formation mechanism, the so-called “common pathway.”
Extrinsic pathway (Factor VII-mediated, triggered by release of tissue factor from damaged endothelium):
Tissue factor serves as a co-factor with Factor VIIa (it’s not clear what activates VII) to activate Factor X to Xa.
Note VIIa can activate elements in the intrinsic pathway as well (XI, IX).
Intrinsic pathway (Factor VIII-mediated, Factor VII and tissue factor independent):
Factor XII is activated (unclear what does this).
XIIa activates XI.
XIa activates IX.
IXa, with Factor VIII as its co-factor, activates Factor X to Xa.
Common pathway (Factor V-mediated):
Factor Xa, oriented by Factor V, cleaves prothrombin (Factor II) to thrombin (Factor IIa).
Thrombin cleaves fibrinogen (I) to fibrin (Ia), a soluble fiber that begins to polymerize.
Factor XIII cross-links fibrin polymers to form a hard, insoluble clot.
And that’s clotting. To reiterate:
Extrinsic: tissue factor + VIIa -> Xa.
Intrinsic: XIIa -> XIa -> IXa + VIII -> Xa.
Common: Xa -> IIa -> Ia + XIII -> clot.
Several notes:
Notice that many steps in these pathways require Ca2+. No calcium, no (or little) clotting.
Fibrin clots binds the platelets at the site together and attaches them more firmly to the vessel wall.

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

Explain how thrombin acts as the central regulation point for coagulation. Describe normal homeostasis in the coagulation system

A

Thrombin (IIa) is the central regulation point for coagulation. Activates platelets, cleaves fibrinogen to form fibrin. It also activates Factors VIII and V, which further accelerates the clotting cascade (recall that these are the two large orientation co-factors).

Normal hemostasis involves a certain amount of regulation of thrombin. There are anti-thrombin proteins that normally circulate to bind free thrombin to prevent its being activated. Thrombin also activates another anti-coagulation protein, protein C, which cleaves Factors Va and VIIIa to slow down clotting. There’s also a protein called tissue factor pathway inhibitor that inhibits Xa and the tissue factor-factor VIIa complex. Point is that there’s a balance going on.

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

Explain the function of platelets in hemostasis. Describe the process of platelet aggregation

A

As mentioned, adhere to site of endothelial injury, partly mediated by vWF. Activated by thrombin (IIa) to release vasoconstrictors (TXA2), calcium, and ADP, as well as changing conformation, exposing proteins (factors IIb and IIIa, if I’m reading my notes right) to bind more vWF and fibrinogen, thus binding and activating more platelets.

Platelets also have receptors for Factor V, which (recall) is the orienting co-factor for the conversion of prothrombin to thrombin. This speeds up that conversion.

Platelets also expose the phospholipid complex, which provides a greatly pro-clotting enzymatic effect on the coagulation cascade.

As mentioned just now, platelets adhere due mainly to vWF but are activated largely by thrombin (from clotting factor activity). Once activated, platelets expose receptors which bind vWF and thus bind other platelets to the site, as well as releasing vWF, ADP, etc in granules to recruit and activate them.

Note epinephrine, ADP, and collagen also activate platelets.
Drugs: see “Pharmacology of Anticoagulation Therapy.” Essentially aspirin, ADP receptor blockers, and adhesion glycoprotein inhibitors.

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

Explain the contribution and influence of endothelial cells on coagulation

A

As mentioned, when damaged, they expose collagen (mainly type II), vWF, and tissue factor.
Collagen and vWF bind platelets.
Tissue factor activates the extrinsic clotting pathway.

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

Describe the regulatory mechanisms of coagulation, i.e., protein C pathway, antithrombin, fibrinolytic pathway

A

Fibrinolytics:
Fibrin (clot material) is degraded by plasmin, which is cleaved and activated by tissue plasminogen activator (TPA). After degradation of fibrin, fibrin split products (also called fibrin degradation products) are formed, which are also anti-coagulation factors.

Protein C pathway:
Activated thrombin (IIa) activates Protein C, with thrombomodulin as a cofactor.
Protein C, with Protein S as co-factor, inactivates Factors Va and VIIIa.
Note that a mutation in Va can cause resistance to its inactivation.

Antithrombin:
Antithrombin III forms a complex with thrombin and other serine proteases to block their activity.
It’s activated by heparin (endogenously produced anti-clotting substance).
Note that, in the absence of endothelial damage, the balance is generally in favor of anti-clotting activity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Review events occuring during hemostasis. Compare primary and secondary hemostasis.

A

Adhesion, activation, and aggregation of platelets to form a platelet plug constitute the first events in formation of a clot (primary hemostasis). The platelet plug is stabilized by formation of a fibrin network generated through the coagulation cascade (secondary hemostasis). Optimal numbers and function of platelets are key to cessation of bleeding from small vascular injuries.

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

Diagram the structure of a mature platelet and show the location of: dense granules, alpha granules, glycoprotein 1a, glycoprotein Ib, glycoprotein IIb/IIIa, and phospholipids.

A

Dense Granules: located inside the platelet cell, release ADP which stimulates local platelets to aggregrate and become activated. Dense granules are less common inside the platelet cell than alpha granules.

Alpha Granules: located inside the platelet cell, release growth factors, fibrinogen, VWF

Glycoprotein 1a: Receptor on the surface of the platelet cell that binds collagen of the injured subendothelial tissue.

Glycoprotein 1b: Receptor on the surface of the platelet cell that binds to VWF

Glycoprotein IIb/IIIa: Receptor on the surface of the platelet cell that binds fibrinogen and links platelets together to form clot.

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

List three functions of platelets.

A
  1. Platelets play several important roles in hemostasis, including adhesion to the vascular subendothelium at sites of injury to begin the hemostatic process.
  2. Activation of intracellular signaling pathways leading to cytoskeletal changes and release of intracellular granules to enhance platelet plug formation, aggregation to form the platelet plug, and
  3. Support of thrombin generation by providing a phospholipid surface for the coagulation cascade to take place.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Construct a simple diagram that depicts the process of platelet adhesion. Include in the drawing subendothelial collagen, von Willebrand factor, and glycoprotein Ib. Explain why platelet adhesion to blood vessels does not occur under normal circumstances.

A

Platelet adhesion to blood vessels does not occur under normal circumstances. The endothelial cells of intact vessels prevent blood coagulation by secretion of a heparin-like molecule and through expression of thrombomodulin, which when bound to thrombin activates protein C and S (inhibit factor Va and VIIIa). Intact endothelial cells prevent platelet aggregation by the secretion of nitric oxide and prostacyclin, inhibitors of platelet activation.

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

Similarly, construct a simple diagram that shows the process of platelet aggregation; include the release reaction (ADP), thromboxane synthesis, ADP and thromboxane receptors, glycoprotein IIb/IIIa, and fibrinogen.

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

List and describe three mechanisms that could lead to thrombocytopenia.

A
  • decreased platelet production (due to aplastic anemia, myelodysplasia, and leukemia, infection, severe B12/folate deficiency)
  • increased platelet destruction or consumption (immune thrombocytopenic purpura - ITP, autoimmunity against platelets; DIC)
  • sequestration of platelets in the spleen.

A normal platelet count is between 150,000 and 400,000/uL. Spontaneous hemorrhage and increased risk of hemorrhage with trauma or surgery may be seen with platelet counts <50,000/uL, and with platelet counts less than 10-20,000/uL, life-threatening spontaneous hemorrhage, such as spontaneous intracranial hemorrhage, can be seen.

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

Identify three methods of treating ITP and the mechanism by which they increase the platelet count.

A

Acute ITP (more common in kids) may spontaneously resolve or can be treated with steroids.

Chronic ITP (more common in adults) almost always requires treatment. The most commonly used treatment options include corticosteroids, intravenous immunoglobulin (IVIG), and splenectomy. Steroids work by dampening proliferation of the B cell clone making the autoantibody. An effect is usually seen within 7 to 10 days of starting treatment. IVIG acts by blocking splenic Fc receptors to prevent their binding to antibody-coated platelets, with an effect being seen within 1 to 2 days. Splenectomy works by removing the site of autoantibody-induced platelet removal and leads to lasting responses in 60 to 70% of patients.

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

Describe the molecular defect, typical clinical course, and general approach to treatment for a patient with Von Willebrand Disease.

A

Molecular Defect: Abnormality in platelet / endothelial interaction

Clinical Course: Mucosal bleeding, nose bleeds, GI bleeds, Menorrhagia, bleeding after surgery if no correction.

Diagnosis: PFA is a screening test that would confirm a bleeding disorder/functionality of VWF protein, can directly measure the level of Factor VIII, Von Willlebrand Antigen measures amount of Von Willebrand protein, can measure Von Willebrand activity via donor platelets

Treatment:

  • Avoid aspirin, NDSAIDS, and other platelet inhibiting drugs
  • prophylaxis tx not generallly required
  • DDAVP (arginine vasopressin) enhances already synthesized VWF release from endothelial stores (only effective in type 1 VWF disease)
  • Replacement therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

List important questions to ask when obtaining a bleeding history in a patient with excessive bleeding.

A

A detailed history of the type, frequency, and amount of bleeding is essential when evaluating a patient for a suspected bleeding disorder. Some patients may consider an appropriate amount of bruising or bleeding to be excessive, making the evaluation even more challenging.

Questions to address include:

  • Does the patient display excessive, prolonged, recurrent, or delayed bleeding?
  • Has the patient ever had the opportunity to bleed excessively (physical trauma, skin lacerations, surgery)?
  • Is there a family history of significant bleeding?

Other:

  • Is the bleeding real?
  • Is it platelet type or coagulation factor type bleeding?
  • Is it acquired or congenital?
  • What tests do I order and how do I interpret them?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

List important laboratory studies to obtain when evaluating a patient with excessive bleeding.

A

Basic screening tests when evaluating excessive bleeding can include:
• Platelet count and blood smear to evaluate for thrombocytopenia or other hematologic abnormalities
• Bleeding time or platelet function analyzer (PFA-100) to evaluate primary hemostasis
• APTT as a screening test for the intrinsic coagulation pathway
• PT/INR as a screening test for the extrinsic coagulation pathway
• Thrombin clotting time (TCT) to evaluate for fibrinogen defects, the presence of fibrin split products, or heparin effects
• Fibrinogen level

Other, more advanced tests

  • Appropriate factor levels to rule out factor deficiency
  • 1:1 mixing of normal plasma with patient plasma to evaluate for a lupus anticoagulant or factor inhibitor can then be done.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

List some of the major congenital or acquired disease states causing bleeding and/or clotting.

A

Common: Hempophilia A (Factor VIII deficiency) and B (factor IX deficiency) and C (factor XI deficiency) Von Willerbrand disease Less Common: Factor VII deficiency Hypo or dysfibrinogenemia Rare: Factore XIII, V, X, and II (very rare, not likely compatible with life)

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

Explain what PT/INR is, and what they are testing. Provide a ddx of an abnormal PT/INR. Provide ddx of abnormal findings.

A

Protime/International Normalized Ratio Calcium and thromboplastin are added to citrated plasma and time to clotting is measured in seconds. Thromboplastin varies by manufacturer and time in seconds is normalized by adjusting for known potency (INR=1 is normal time)

Prolonged with II, VII, V, X and fibrinogen deficiencies. Prolonged with vitamin K deficiency and liver disease.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Explain what APTT is, and what they are testing. Provide a ddx of an abnormal APTT. Provide ddx of abnormal findings.

A

activated Partial Thromboplastin Time Surface activating agent and phospholipid is added to citrated plasma. Activated plasma is then re-calcified and time to clotting measured in seconds.

Measures the activity of entire pathway except factor VII

DDX Patients who bleed: VIII (hemophilia A), IX (hemophelia B), XI, XII deficiencies, severe von Willebrand Disease Sensitive to inhibition by heparin, and fibrin split and degradation products

DDX patients who do not bleed: Factor XII deficiency Lupus anticoagulant (clot too much)

Causes of a Prolonged PTT
Heparin in Sample
Hemophilia A and B (Factor VIII or IX Deficiency)
Factor XI Deficiency
Factor XII Deficiency*
Acquired Hemophilia
von Willebrand Disease
Lupus Anticoagulant*
(*Patients do not bleed)

20
Q

Explain what TT is, and what they are testing. Provide a ddx of an abnormal TT. Provide ddx of abnormal findings.

A

Thrombin Time Excess thrombin is added to plasma, which detects low or abnormal fibrinogen, fibrin split products or heparin. Fibrin activity should also be measured.

21
Q

Explain what bleeding time/PFA is, and what they are testing. Provide a ddx of an abnormal bleeding/PFA time. Provide ddx of abnormal findings.

A

Bleeding Time/Platelet Function Analyzer Bleeding time measures platelet function, vessel wall and skin integrity. A template device makes a cut on the forearm and time to clot (2-9 min) is measured. Platelet Function Analyzer can determine an in vitro bleeding time with agonists.

22
Q

Describe some of the other tests used to evaluate patients with thrombotic or bleeding disorders.

A

Mixing to detect inhibitors

Coagulation factor testing

23
Q

Describe the clinical features and molecular basis for hemophilia A and hemophilia B

A

X-linked defiency of Factor VIII (A) or Factor IX (B) Males primarily affected, females often symptomatic Elevated PTT is the ONLY abnormal screening test Family hx important but 30% of time due to new mutations. Hemophilia A (Factor VIII) more common, Hemophilia B (Factor IX) less common DX: screen males with unexplained hematomas, bruises or post-surgical/traumatic bleeding PTT ranges from high normal 38 sec to prolonged 100 sec Assay Factor activity to distinguish VIII from IX

24
Q

Describe the clinical features and molecular basis for factor VII deficiency

A

Rare

Abnormal PT is the only abnormal test observed.

25
Q

Describe the clinical features and molecular basis for von Willebrand disease

A

Common, mild; deficiency in vWF, with associated difficulties aggregating and binding platelets.
Shows an abnormal bleeding time; also can show an increased PTT due to vWF’s effects to prolong the half-life of Factor VIII.

26
Q

Describe the role of liver disease in coagulopathy

A

Liver disease:
Decreased synthesis of most factors aside from VIII and IX; can’t use vitamin K to carboxylate II, VII, IX, and X.
Can also stop making fibrinogen (I) and also increased consumption of platelets by spleen due to venous backup in portal hypertension.
[For diagnosis: both PT and aPTT prolonged, but PT much more so than aPTT (no carboxylation of Factor VII).]
[Treatment:]
give plasma infusion; give vitamin K; can use recombinant factor VII (VII has shortest half-life, dies out of FFP quickly); platelet transfusions.

27
Q

Describe disseminated intravascular coagulation (DIC) with its associated conditions. Understand diagnostic testing for DIC and associated conditions

A

Disseminated Intravascular Coagulation is a systemic activation of coagulation, which can result in intravascular depostion of fibrin and thrombosis of small and midsized vessels leading to organ failure (hypoxia / infarction) OR progressive depletion of platelets and coagulation factors leading to bleeding.

Peripheral smear can be used to detect microangiopathic (fragmented RBC) cells.

Common clinical conditions associated with DIC: Sepsis Trauma Malignancy (APL) Obstetrical complications (amniotic fluid embolism) Vascular disorders (aneurysm, venous malformity) Reaction to Toxin (snake venom) Immunologic disorders

Lab: PTT and protime both abnormal, but PTT much higher than protime (reverse of liver disease).
On smear: looking for schistocytes (helmet cells, have sharp points) and an abnormally low number of platelets.

Caused by:

  • Sepsis (most common)
  • Trauma with fat embolism (bone, head trauma)
  • Cancer
  • Obstetric complication (amniotic fluid embolism)
  • Dissecting aortic aneurysm or other vascular disorders
  • Some snake venoms
  • Mismatched blood transfusion (immediate hemolytic reaction)

Long PTT usually reflects nonspecific inhibition by fibrin split products
Low fibrinogen is the key indicator– low is bad, higher is better.
Note you don’t generally treat DIC directly- treat the underlying disorder. Trying to stop progression with anticoagulants is sometimes tried but can be dangerous due to bleeding.
Replace blood products and factor if bleeding out.

28
Q

Explain what a lupus anticoagulant is, how it affects coagulation, and ways to test for it

A

Lupus anticoagulant are antibodies to phospholipid in platelets which bind

Delay the PTT test reaction, so in vitro there is a prolonged PTT but in vivo there are thrombotic disorders.

Test for it by using a procedure that absorbs the antiphospholipid antibodies prior to assaying coagualation time - Russel Viper Veno Test or Platelet Neutralization test with correction are common tests.

You could also assay Factor VIII, IX, XI, XII and check for inhibitory pattern on all factors.

Can be treated with heparin. Note that it’s really important to make sure it’s this disease, since treating most of the other prolonged PTT disease states will not react kindly to heparin (anticoagulant + bleeding disorder = bad news).

29
Q

Explain how a 1:1 mixing study can distinguish a clotting factor deficiency from an inhibitor of coagulation

A

Mix patient and control plasma 1:1 and assay for Partial Thrombostin Time. If it corrects you have a factor deficiency, then incubate (2 hr @ 37C) and re-assay PTT, if stay corrected then factor deficiency, if not, then it is a specific factor VIII inhibitor (acquired hemophilia). If it does not correct initially then it is an inhibitor, likely LA.

30
Q

Describe the mechanism of action and pharmacokinetics of heparin and low molecular weight heparins, and differences in management of patients on these therapies

A

Heparins interfere with the coagulation cascade and prevent the formation of thrombin which is needed to convert fibrinogen to fibrin to form solid secondary clots. The mechanism of action is heparin binds to the Anti-Thrombin III serine protease inhibitor which enhances its ability to bind coagulation factors (100x) to prevent coagulation and also accelerate their decay. Pharmacokinetics Unfractioned heparin is administered IV or sub-cutaneously, poor pharmacokinetics and bioavailibilty (dense negative charge), dose response is unpredictable, requires hospital admission and monitoring, doesn’t cross placenta Low molecular weight heparin and fondaparinux are adminstered subcutaneously, better bioavailability, more predictable dose response, longer half life, less monitoring (outpatient)

31
Q

Describe the complications associated with heparin therapy, including excessive bleeding and heparin-induced thrombocytopenia with associated thrombosis.

A

Bleeding/hemorrhage can occur, heparin and its effects disappear after hours of its discontinuation but its effects can be immediately reversed with protamine sulfate - a positively charged molecule that neutralizes heparin. Heparin-induced thrombocytopenia syndrome (HIT), is when the platelet count decreases >50% 5-10 days after administration. This is caused by the development of antibodies to the platelet factor 4/heparin complexes which bind to and activate platelets resulting in a prothrombotic state and clot formation which decreases the peripheral platelet counts as the clots accumulate.

32
Q

Describe the alternative anticoagulant therapies used for patients with heparin-induced thrombocytopenia.

A

HIT is less common with low molecular weight heparin. Nonetheless, if it does happen Argatroban and Lepirudan can be used as direct thrombin inhibitors which act as anti-coagulants and will resolve the prothrombotic state.

33
Q

Describe the mechanism of action, pharmacokinetic and uses of oral anticoagulant warfarin.

A

Warfarin inhibits enzymes that use vitamin K as a co-factor. Specifically, this affects coagulation proteins which need vitamin K to undergo gamma carboxylation in order to bind Ca and become functional. Warfarin depletes vitamin K to cause this effect, but it does not occur for 3-5 days because the remaining functional coagulation proteins must first naturally degrade. Warfarin itself, is rapidly absorbed, has good bioavailability, and a long half life (36-48 hours). Warfarin is typically used to prevent venous thrombosis (in combination with heparin), systemic embolism in patients with prosthetic heart valves/atrial fibrillation, stroke, recurrent infarction, or death in patients with acute MI

34
Q

Describe the adverse effects and potential complications associated with use of warfarin.

A

Hemorrhage is a potential complication. In this case, the drug will be stopped and vitamin K will be administered. It takes 24-48 hrs for vitamin K reversal b/c new coagulation factors have to be synthesized, so coagulation factors will be infused as well. Warfarin crosses the placenta and is teratogenic, so cannot be used during pregnancy.

35
Q

Describe the relationship between mechanisms of action and speed of onset of action of heparin and oral anticoagulants.

A

Heparin = rapid onset Warfarin = delayed onset Oral anticoagulants = rapid onset Heparin acts rapidly by enhancing anti-thrombin III’s ability to bind coagulation factors and prevent their function, whereas oral anticoagulants are direct inhibitors of thrombin or factor Xa. Thus, oral anticoagulants have one less step in the process and are slightly more efficacious than heparin.

36
Q

Describe the mechanisms of action and uses of fibrinolytic agents.

A

Fibrinolytic agents work by converting plasminogen to plasmin, an active protease that degrades fibrin clots. Fibrinolytic agents are used for: Treatment of acute MI (<3 hours) DVT and pulmonary embolism

37
Q

Describe the mechanisms of action and uses of antiplatelet agents.

A

There are 3 types of antiplatelet agents, each with specific mechanisms. 1. Aspirin - Inhibits thromboxane A2 production (which stimulates platelet activation) by irreversibly inactivating cyclooxygenase 1. Uses: after AMI and thrombotic stroke (with thrombolytics) to prevent AMI and stroke in high-risk patients 2. ADP receptor blockers a. Thienopyridines-Ticlopidine (Ticlid), Clopidogrel (Plavix), prasugrel bind irreversibly to the ADP receptor to block granule secretion and expression of adhesion proteins (GPIIB/GPIIIa), Used to prevent AMI and thrombotic stroke (with aspirin) b. Ticagrelor binds reversibly to ADP receptor, acts more rapidly 3. Glycoprotein IIb/IIIa inhibitors - block binding of fibrinogen to the adhesion protein GPIIb/IIIa. Used IV for inpatient use before, during and after angioplasty to prevent recurrence of stenosis and AMI (with aspirin and heparin), treatment of AMI (with fibrinolytics), for unstable angina

38
Q

Heparin is typically indicated for the treatment or prevention of:

A

Venous thrombosis, pulmonary embolism (heparin acts rapidly), management of angina, acute MI, coronary angioplasty, stent placement, surgery requiring cardiac bypass, abdominal surgery/hip/knee replacement, kidney dialysis

39
Q

Identify the componenets of Virchow’s triad and their pathophysiologic contribution to thrombosis.

A

vascular obstruction due to thrombosis resulted from three interrelated factors:
1. Decreased blood flow (venous stasis)

  • immobility
  • paralysis
  • reduced flow states
  1. Inflammation of or near the blood vessels (altered vessels)
  • inflammatory damage
  • mechanical injury
  • hypoxia
  1. Intrinsic alterations in the nature of the blood itself (altered coagulability)
  • inflammatory stimuli
  • consumption of endogenous anticoagulants
40
Q

Describe at least three major clinical symptoms that occur when a patient suffers from an acute iliofemoral thrombosis of the leg, and indicate the pathophysiologic reason for each one (for example, dilated superficial veins of the calf due to obstruction of venous return in the occluded deep veins)

A

Clinical Symptoms and respective pathophysiology:

  1. Dilated superficial veins of the calf – due to obsruction of venous return in the occluded deep veins and blood flow via alternative superficial routes
  2. Swelling, redness/dusky color warmth – obstruction of deoxygenated blood and inflammatory response
  3. Pitting edema of distal extremity – transudate/increased hydrostatic pressure from back up of blood flow
  4. Size differences between left and right – obstructed on one side but not the other
  5. Pain – pressure from blood back-up
41
Q

Compare and contrast the cause and mechanism of a thrombus occurring in the arterial circulation (such as acute coronary artery thrombosis) from one that develops in a deep vein of the leg. Include the instigating factor(s) and composition of the clot.

A

Arterial Thrombus:

  • Occur under conditions of high shear stress
  • Composed primarily of aggregated platelets (appear white b/c low levels of fibrin and RBCs)
  • Can develop ischemia in downstream tissues
  • Abnormalities of blood flow that can contribute
    • hypertension
    • turbulent flow at arterial branch points
    • sites of focal atherosclerosis
  • Tx via antiplatelet therapy
  • Most inheritable hypercoaguable states do not cause arterial thrombosis

Leg Thrombus: venous in nature, not arterial.

  • Most events in high risk settings, in otherwise healthy people, are venous
  • develop under conditions of slow blood flow (low shear stress)
  • Large amounts of fibrin and RBCs (thrombi appear red)
  • stasis due to:
    • right sided heart failure
    • pre-existing venous thrombosis
    • extrinsic vascular compression by tumor
    • immobility
    • obesity
    • chronic venous insufficiency
  • Altered coagubility
    • inherited
    • oral contraceptives
    • age
42
Q

List three clinical clues suggesting an inherited hypercoagulable disorder.

A
  1. First thrombosis age <50
  2. Recurrent episodes of thrombosis
  3. Family history of thrombosis
  4. Thrombosis at unusual sites
  5. Neonatal thrombosis
  6. Thrombosis without apparent antecedent thrombogenic event (idiopathic)
43
Q

Briefly describe at the molecular level the pathophysiologic reason that patients with deficiencies of antithrombin, protein C, protein S, factor V Leiden or the prothrombin gene mutation are likely to have thrombosis. Explain what tests are used to identify these patients.

A

Deficiency:

  • Antithrombin: regulates coagulation by inactivating thrombin and Factors Xa, IXa, XIa and XIIa.
  • Protein C: a vitamin K dependent plasma protein that inactivates Factors Va and VIIIa to inhibit coagulation.
  • Protein S: a vitamin K dependent plasma protein that facilitates the anticoagulant activity of activated Protein C. Dx by measuring Protein C activity.
  • Factor V Leiden: mutation of the Factor V gene that leads to a structural change and partial resistance to inactivation through proteolytic cleavage by protein C. Dx by DNA analysis
  • Prothrombin Gene Mutation: mutation likely leads to elevated concentrations of plasma prothrombin.

Tests:

  • D-Dimer: formed when cross-linked fibrin has been degraded by plasmin through fibrinolysis.
  • CT scan: for PE suspicion
  • V/Q scan: for PE suspicion
  • Ultrasound: for DVT suspicion
  • DNA analysis: confirm type of deficiency
  • History
44
Q

List and describe at least three acquired disorders that are associated with recurrent venous or arterial thromboembolism.

A
  • Mild hyperhomocysteinemia (increased platelet activation and adhesion due to endothelial cell injury)
  • Increased age
  • Beta-2 glycoprotein 1 antibodies
  • Lupus anticoagulant
  • Elevated anticardiolipin antibodies
  • Oral contraceptives
  • Pregnancy
  • Malignancy
  • Hyperviscosity
  • Presence of antiphospholipid antibodies
  • Presence of central venous catheter
  • Surgery, especially orthopedic
  • Trauma
  • Hormone replacement therapy or tamoxifen
  • Anti-angiogenesis (inhibited growth of new blood vessels)
  • Congestive heart failure
  • Myeloproliferative disorders
  • Paroxysmal nocturnal hemoglobinuria
  • Inflammatory Bowel Disease
  • Nephrotic Syndrome
  • Marked leukocytosis in acute leukemia
  • Sickle cell anemia
  • HIV/AIDS
45
Q

Describe the clinical features and criteria for diagnosis of antiphospholipid antibody syndrome.

A

A transient syndrome that causes thrombotic (venous and arterial) or obstetric complications caused by antibodies produced in association with drug exposure, infections or accute illness.

Clinical

  • Vascular thrombosis
    • One or more clinical episodes of arterial, venous or small-vessel thrombosis of any organ or tissue
  • Complications of pregnancy
    • One or more unexplained death of nl fetus  10 wks
    • One or more premature births of nl neonates  34 wks
    • Three or more unexplained consecutive abortions < 10 wks
  • Other indicators
    • Thrombocytopenia (40-50%), Hemolytic anemia (14-23%), Transient cerebral ischemia, Transverse myelopathy/myelitis, Livedo reticularis (11-22%), Multiple sclerosis-like syndrome, Chorea, Migraine

Laboratory criteria

  • Must have detected 2 or more occasions at least 12 weeks apart
    • Anticardiolipin Antibodies
    • Lupus anticoagulant
    • Beta2-glycoprotein-I antibodies
46
Q

Explain the key factor in determining how long someone should be anticoagulated for a venous thrombosis.

A

Key in the decision-making process is determining if the DVT occurred due to transient risk factors (such as surgery, temporary immobilization, trauma, or pregnancy) or if the patient has an underlying hypercoagulable disorder or ongoing risk factors that require longer treatment.