Hemostasis Flashcards

1
Q

How does the vascular endothelium reduce unprovoked thrombosis?

A

Healthy endothelial cells possess antiplatelet and anticoagulant activity that functions to inhibit clot formation. The negatively charged vascular endothelium repels platelets, and endothelial cells produce potent platelet inhibitors such as prostacyclin (prostaglandin I2) and nitric oxide that prevent adhe- sion of quiescent platelets. An adenosine diphosphatase (CD39) expressed on the surface of vascular endothelial cells also serves to block platelet activation through degradation of adenosine diphosphate (ADP), a potent platelet activator. The vascular endothelium also plays a pivotal anticoagulant role by expressing several inhibi- tors of plasma-mediated hemostasis. Endothelial cells can increase activation of protein C, an anticoagulant, via surface expression of thrombomodulin (TM), which acts as a cofactor for thrombin-mediated activation of protein C, making its activation 1000 times faster. Endothelial cells also produce tissue factor pathway inhibitor (TFPI), which inhibits the procoagulant activity of factor Xa and the TF–VIIa complex. Finally, the vascular endothelium synthesizes tissue plasminogen activator (tPA), which is responsible for activating fibrinolysis, a primary counter-regulatory mechanism limiting clot propagation.

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

Describe what occures with platelets after an endothelium injury

A

Damage to vascular endothelial cells exposes the underlying
extracellular matrix (ECM), which contains collagen, von
Willebrand factor (vWF), and other platelet-adhesive
glycoproteins. Platelet receptors for vWF (glycoprotein
Ib-IX-V complex) and collagen (integrin α2β1) facilitate
platelet adhesion to the site of vessel injury. Absence of either vWF (von Willebrand disease) or glycoprotein Ib-IX-V complex receptors (Bernard–Soulier syndrome) results in a clinically significant bleeding disorder.

In addition to promoting their adhesion to the vessel wall, the platelet interaction with collagen serves as a potent stimulus for the subsequent phase of platelet activation. During the activation phase, platelets secrete agonists such as thromboxane A2
(TxA2 ) and release granular
contents, resulting in recruitment and activation of
additional platelets and propagation of plasma-mediated
coagulation. Platelets contain two specific types of storage granules: α-granules and dense bodies. α-Granules contain numerous proteins essential to hemostasis and wound repair, including fibrinogen, coagulation factors V and VIII, vWF, platelet-derived growth factor, and
others. Dense bodies contain the adenine nucleotides
ADP and adenosine triphosphate (ATP), in addition to calcium, serotonin, histamine, and epinephrine. Redistribution of platelet membrane phospholipids during
activation exposes newly activated platelet surface receptors and binding sites for calcium and coagulation factor.

activation complexes, which is critical to propagation of
plasma-mediated hemostasis. During activation, platelets
also undergo structural changes to develop pseudopodlike membrane extensions and to release physiologically active microparticles, which serve to dramatically increase the platelet membrane surface area.
During the final phase, platelet aggregation, activators
released during the activation phase recruit additional
platelets to the site of injury. Newly active glycoprotein
IIb/IIIa receptors on the platelet surface gain higher affinity for fibrinogen, thereby promoting crosslinking and aggregation with adjacent platelets. The importance of these receptors is reflected by the bleeding disorder associated with their hereditary deficiency, Glanzmann thrombasthenia

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

Describe the extrinsic pahtway of coagulation

A

The extrinsic pathway of coagulation is now understood to represent the initiation phase of plasma-mediated
hemostasis and begins with exposure of blood plasma to
tissue factor (TF).

After vascular injury, small concentrations of factor VIIa circulating in plasma form phospholipid-bound activation complexes with TF, factor X, and calcium to promote conversion
of factor X to Xa. Additionally, the TF/factor VIIa complex activates factor IX of the intrinsic pathway, further demonstrating the key role of TF in initiating hemostasis

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

Describe the intrinsic pathway of coagulation

A

The intrinsic pathway begins when factor XII or the Hageman factor is exposed to collagen, kallikrein, and high molecular weight kininogen (HMWK) and is subsequently activated. Factor XIIa activates factor XI into XIa. With a calcium ion, factor XIa activates factor IX. Then, factor IXa, factor VIIIa, and calcium form a complex to activate factor X. Factor VIII is found in the blood and is often activated by thrombin (factor IIa)

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

Describe the common pathway of Coagulation

A

The final pathway, common to both extrinsic and intrinsic coagulation cascades, depicts thrombin generation and subsequent fibrin formation.

Signal amplification results from activation of factor X by both intrinsic (FIXa, FVIIIa, Ca2+) and extrinsic (TF, FVIIa, Ca2+) tenase
complexes. The tenase complexes in turn facilitate
formation of the prothrombinase complex (FXa, FII [prothrombin], FVa [cofactor], and Ca2+), which mediates a surge in thrombin generation from prothrombin. Thrombin proteolytically cleaves fibrinogen molecules to generate fibrin monomers, which polymerize into fibrin strands to form a clot. Finally, factor XIII is activated by
thrombin and acts to covalently crosslink fibrin strands producing an insoluble fibrin clot resistant to fibrinolytic
degradation

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

Which are the major contrarregulatory pathways of coagulation?

A

Four major counterregulatory pathways have been identified
that appear particularly important for downregulating
hemostasis: fibrinolysis, tissue factor pathway inhibitor (TFPI) , the protein C system, and
serine protease inhibitors (SERPINs)

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

In vivo, plasmin generation
is most often accomplished by release of … from the vascular endothelium

A

tissue plasminogen activator (tPA) or urokinase

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

How the normal fibrinolysis is limited to the area of clot formation?

A

Activity of tPA and urokinase is accelerated in the presence of fibrin, thereby limiting fibrinolysis to areas of clot formation

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

How does plasmin acts?

A

Promotes enzymatic degradation of fibrin and fibrinogen and inhibits coagulation by degrading essential cofactors V and VIII and reducing platelet glycoprotein surface receptors essential to adhesion and aggregation.

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

What is the role of the serine protease inhibitors (SERPINs) in the coagulation process?

A

excessive fibrinolysis is prevented by the function
of two key SERPINs, namely plasmin-activator inhibitor-1 (PAI-1) and α2‐antiplasmin. PAI-1 serves as the primary inhibitor of tPA and urokinase, thereby decreasing plasmin generation, whereas α2‐antiplasmin directly
inactivates circulating plasmin

In addition, one of the most significant SERPINs regulating hemostasis is antithrombin (AT, formerly antithrombin III).
AT can inhibit all procoagulant proteases of the blood clotting cascade, but its primary targets appear to be thrombin and factors Xa (FXa) - less efficiently IXa (FIXa) XI, XII and the others.
Heparin binds AT, causing a conformational change that
accelerates AT-mediated inhibition of targeted enzymes
by over1000-fold

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

What is the tissue factor pathway inhibitor role in the coagulation?

A

TFPI binds and inhibits factor Xa through the formation of membrane-bound complexes. These factor Xa–TFPI complexes also act to inhibit TF/factor VIIa complexes, thereby downregulating the extrinsic coagulation pathway

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

How the protein C system acts?

A

The protein C system proves particularly important in regulating coagulation through inhibition of thrombin and the essential cofactors Va and VIIIa.

After binding to thrombomodulin on the surface of the endothelial cell, thrombin’s procoagulant function decreases and instead its ability to activate protein C is
augmented. Activated protein C (APC), complexed with the cofactor protein S, degrades both factors Va and VIIIa. Loss of these critical cofactors limits formation of tenase and prothrombinase complexes essential to formation of factor Xa and thrombin, respectively

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

Von Willebrand factor function

A

Under normal conditions vWF plays a critical role in platelet
adhesion to the extracelular matrix and prevents degradation of factor
VIII by serving as a carrier molecule.

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

Von Willebrand Disease presentation and labs

A

Classically, patients with vWD describe a history of easy
bruising, recurrent epistaxis, and menorrhagia, which are characteristic of defects in platelet-mediated hemostasis.
In more severe cases (i.e., type 3 vWD), concomitant
reductions in factor VIII may lead to serious spontaneous
hemorrhage, including hemarthroses.

Routine coagulation studies are generally not helpful in the diagnosis of vWD, as the platelet count and prothrombin time (PT) will be normal in most patients
and the activated partial thromboplastin time (aPTT) may
demonstrate mild-to-moderate prolongation depending
on the level of factor VIII reduction. Initial screening tests
for vWD involve measurement of vWF levels (vWF antigen) and vWF platelet binding activity in the presence of the ristocetin cofactor, which leads to platelet agglutination.

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

Presentation and labs of hemophilia A and B

A

Hemophilia A (factor VIII deficiency) and hemophilia B
(factor IX deficiency) are both X-linked inherited bleeding disorders most frequently presenting in childhood as
spontaneous hemorrhage involving joints and/or deep
muscles.The severity of the disease is dependent
on an individual’s baseline factor activity level. Severe disease, defined by less than 1% of coagulation factor activity, occurs in approximately two-thirds of patients with hemophilia A and one half of patients with hemophilia B.

Classically, laboratory testing in patients with hemophilia reveals prolongation of the aPTT, whereas the PT, bleeding time, and platelet count remain within normal limits. However, a normal aPTT may also be seen in
mild forms of hemophilia, and it is important to exclude
vWD as a cause of factor VIII deficiency

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

What is the hemophilia C?

A

Factor XI deficiency, known as hemophilia C or Rosenthal syndrome (prevalence: 1 in 1,000,000), is characterized by isolated prolongation in aPTT and variable bleeding severity. Factor XI activity levels, however, do not correlate well with bleeding risk. Most individuals
do not experience spontaneous bleeding, hemarthrosis, or
muscle hematomas, though bleeding episodes can occur
under situations of hemostatic challenge such as trauma,
surgery, or childbirth

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

Presentation and coagulogram of Factor XIII deficiency

A

Factor XIII is involved in stabilizing the fibrin clot. Factor
XIII deficiency (prevalence: 1 in 2,000,000) presents with delayed bleeding after hemostasis, impaired wound healing, and, occasionally, pregnancy loss.

Laboratory evaluation in these patients will demonstrate normal aPTT and PT, but the diagnosis can be confirmed by measurement of factor XIII activity levels

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

Explain the vitamine F deficiency ant it’s aassociation with bleeding

A

Vitamin K is an essential fat-soluble vitamin that is required for the carboxylation of factors II, VII, IX, and X and proteins C and S. Without carboxylation, these factors cannot bind to the phospholipid membrane of
platelets and participate in hemostasis.

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

How does liver diseas affects coagulation?

A

The liver is the primary site for production of procoagulant factors, including fibrinogen; prothrombin (factor
II); factors V, VII, IX, X, XI, and XII; the anticoagulants
protein C and S; and AT. Severe liver disease impairs synthesis of coagulation factors, produces quantitative and
qualitative platelet dysfunction, and impedes clearance
of activated clotting and fibrinolytic proteins. Laboratory
findings commonly associated with liver disease include
a prolonged PT and possible prolongation of the aPTT,
suggesting that these individuals are at increased risk of
bleeding. However, these abnormal values only reflect
decreases in procoagulant factors and do not account
for parallel decreases in anticoagulant factors (protein
C, protein S, and AT). As a result, patients with chronic
liver disease are thought to have a rebalanced hemostasis
and actually generate amounts of thrombin equivalent to
healthy individuals.6
Similarly, thrombocytopenia from platelet sequestration in the spleen is often observed in patients with liver disease and portal hypertension. However, levels of the plasma metalloprotease ADAMTS13, responsible for cleaving vWF multimers, are also decreased in chronic liver disease and result in high circulating levels of large
vWF multimers, which promote platelet aggregation.
Consequently, this increase in vWF may partially correct
for thrombocytopenia and platelet dysfunction but can
also result in a prothrombotic state and increase clotting
risk.
Fibrinolysis of a formed clot is also aberrant in patients with liver disease. Excessive fibrinolysis is prevented by thrombin-activatable fibrinolysis inhibitor (TAFI), which blocks activation of plasmin from plasminogen. TAFI is synthesized by the liver, and because levels are decreased in patients with chronic liver disease,
it was believed that such individuals are at increased
bleeding risk because of hyperfibrinolysis. However,
levels of PAI-1, an inhibitor of tPA and urokinase, are
also increased in liver disease and may serve to normalize fibrinolytic activity. Thus, in patients with chronic
liver disease, hemostatic mechanisms are rebalanced,
but decreases in procoagulant and anticoagulant factors
create a tenuous equilibrium that is easily disrupted. As
a result, these patients are at risk for both bleeding and
inappropriate clotting.

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

How does renal disease affects the coagulation?

A

Platelet dysfunction commonly occurs in association
with chronic renal failure and uremia and has primarily been attributed to decreased platelet aggregation and
adhesion to injured vessel walls. Impaired adhesion is
likely the result of defects of glycoprotein IIb/IIIa, which
facilitates platelet binding of fibrinogen and vWF.
Additionally, accumulation of guanidinosuccinic acid
and the resulting increase in endothelial nitric oxide
synthesis further decrease platelet responsiveness. Red
blood cell (RBC) concentration has also been suggested
to contribute to impaired platelet activity, as correction
of anemia shortens bleeding times. This is thought to be
the result of the increased RBC mass displacing platelets
from the center of the vessel and bringing them into
close proximity of the endothelium, thereby promoting
adhesion

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

Presentation and labs of disseminated intravascular coagulation

A

Disseminated intravascular coagulation (DIC) is a pathologic hemostatic response to TF/factor VIIa complex that
leads to excessive activation of the extrinsic pathway, which overwhelms natural
nticoagulant mechanisms
and generates intravascular thrombin

Most often, DIC presents clinically as a diffuse bleeding disorder associated with consumption of coagulation factors and platelets during widespread microvascular thrombotic activity resulting in multiorgan dysfunction.

Laboratory findings typical of DIC include reductions in platelet count; prolongation of the PT, aPTT, and thrombin time (TT); and elevated concentrations of soluble fibrin and fibrin degradation products (D-dimers). However, DIC is both
a clinical and laboratory diagnosis, so laboratory data
alone do not provide sufficient sensitivity or specificity to confirm a diagnosis

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

Conditions associated with DIC

A

1) Infections:
- Bacterial (gram-negative bacilli, grampositive cocci)
- Viral (CMV, EBV, HIV, VZV, hepatitis)
- Fungal (histoplasma)
- Parasites (malaria)

2) Malignancy
- Hematologic (AML)
- Solid tumors (prostate cancer, pancreatic cancer)
- Malignant tumors (mucin secreting adenocarcinoma)

3) Obstetric causes
- Amniotic fluid embolism
- Preeclampsia/eclampsia
- Placental abruption
- Acute fatty liver of pregnancy
- Intrauterine fetal demise

4) Massive inflammation
- Severe trauma
- Burns
- Traumatic brain injury
- Crush injury
- Severe pancreatitis

5) Toxic/ immunologic
- Snake envenomation
- Massive transfusion
- ABO blood type incompatibility
- Graft versus host disease

6) Other
- Liver disease/fulminant hepatic failure
- Vascular disease (aortic aneurysms, giant hemangiomas)
- Ventricular assist devices

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Explain the cardiopulmonary bypass-associated coagulopathy

A

Initial priming of the bypass circuit results in hemodilution and thrombocytopenia. Adhesion of platelets
to the synthetic surfaces of the bypass circuit further
decreases platelet counts and contributes to platelet
dysfunction. During CPB, expression of platelet surface
receptors important for adhesion and aggregation (GPIb,
GPIIb/IIIa) are downregulated and the number of vWFcontaining α-granules are decreased, thereby impairing
platelet function. Furthermore, induced hypothermia
during CPB results in reduced platelet aggregation and
plasma-mediated coagulation by decreasing clotting
factor production and enzymatic activity. Increased
plasmin generation may also occur during CPB, a process that accelerates clot lysis

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

Explain the trauma induced coagulopathy

A

Coagulopathy in this setting may be the result of acidosis, hypothermia, and hemodilution from resuscitation; however, an independent trauma-induced
coagulopathy (TIC) is also experienced by these individuals.
The anticoagulant effect of APC (activated protein C) is thought to play a primary role in TIC by decreasing thrombin generation via inhibition of factor Va and VIIIa and promoting fibrinolysis through inhibition of PAI-1. Hypoperfusion subsequent to traumatic injury is thought to be the stimulus for APC activation. Additionally, tissue damage results in shedding of the endothelial glycocalyx
(EG), a gel-like matrix with anticoagulant properties that
lines the vascular endothelium. The EG contains proteoglycans such as syndecan-1, hyaluronic acid, heparan sulfate, and chondroitin sulfate, that when shed during endothelial injury, result in an “autoheparinization”
phenomenon that contributes to TIC. Impaired platelet
responsiveness also contributes to increased bleeding
in TIC

25
Q

Treatment of von Willebrand disease

A

Mild cases of vWD often respond to DDAVP, which causes the release of vWF from endothelial cells. One dose of DDAVP (0.3 μg/kg IV) will produce a complete or near-complete response in the majority of patients with type 1 vWD. DDAVP is contraindicated in type 2B vWD, as it may precipitate thrombocytopenia, which
could worsen bleeding.

In the setting of more significant
surgical bleeding, use of plasma-derived vWF:factor VIII concentrate (Humate-P) or recombinant vWF (Vonvendi)
is indicated.

If vWF concentrates are not available, cryoprecipitate, which contains high levels of vWF, can be used, but its use is considered second line, as most cryoprecipitate has not undergone the pathogen inactivation processes used in preparing vWF concentrates

26
Q

Treatment os hemophilia A and B

A

In most cases, perioperative management of patients wit hemophilia A or B necessitates consultation with a hematologist and administration of recombinant or purified factor VIII or factor IX concentrates, respectively.

Mild cases of hemophilia A may be treated with desmopressin.

An increasingly common complication of hemophilia, particularly in the case of hemophilia A, has been the development of alloantibodies directed against the factor VIII protein. Administration of factor VIII concentrates will fail to control bleeding in patients with high-titer antibodies. Several approaches to reduce bleeding in these patients include substitution of porcine factor VIII, administration of activated factor VIII bypass activity (FEIBA) or nonactivated prothrombin complex concentrates (PCCs), or treatment with recombinant factor VIIa (rFVIIa).

27
Q

The prothrombin G20210A
gene mutation causes increased plasma concentrations
of … resulting in a …

A

prothrombin

hypercoagulable state

28
Q

How does Factor V Leiden mutation increase the risk of thrombosis?

A

The mutation results in
APC resistance whereby factor Va is no longer susceptible to APC-mediated degradation. This inability of APC to counterregulate the coagulation system results in a prothrombotic state that is found in approximately 5%
of the Caucasian population

29
Q

Protein C deficiency is an autosomal dominant trait
affecting approximately 1 in 500 individuals in the general population. Deficiency can be the result of reduced
concentrations or function of protein C and may result
in …

A
  • VTE
  • warfarin-induced skin necrosis - neonatal purpura (in homozygous neonates)
  • fetal loss
30
Q

Acquired protein C deficiency can be seen in …

A

DIC, liver disease, severe infection (especially meningococcemia), uremia, and those on vitamin K antagonist (VKA) anticoagulation.

31
Q

Acquired protein S deficiency has been associated with …

A

pregnancy, oral contraceptive use, DIC, human immunodeficiency virus (HIV) infection, nephrotic syndrome, and liver disease

32
Q

Physiopathology oh heparin-induced thrombocytopenia

A

Heparin-induced thrombocytopenia (HIT) describes an autoimmune-mediated drug reaction occurring in as many as 5% of patients receiving heparin therapy.
Patients with HIT experience a mild to moderate thrombocytopenia. However, unlike other drug-induced thrombocytopenias, HIT results in platelet activation and increased risk of venous and arterial thromboses. Evidence suggests that HIT is mediated by immunoglobulin G (IgG) antibodies, which bind to platelet factor 4 (PF4).
Complexes composed of antibody, PF4, and heparin bind to platelet Fcγ receptors, thereby activating them

33
Q

Risck factor for heparin-induced thrombocytopenia

A
  • Prolonged exposure to heparin therapy (>5 days)
  • UFH > LMWH > fondaparinux (UFH conveys a risk 10 times greater thanthat of LMWH, whereas the penta-saccharide fondaparinux is rarely associatedwith HIT)
  • Surgical (particularly cardiac and orthopedic) or trauma patients (1% to 5%)
  • female patients have approximately twice the risk for developing HIT
34
Q

Diagnosis of heparin-induced thrombocytopenia

A

A diagnosis of HIT should be considered for any patient experiencing thrombosis or thrombocytopenia (absolute or relative ≥50% reduction in platelet count) during or after heparin administration.

Although HIT remains a clinical diagnosis (4Ts Score), HIT antibody testing should be undertaken to confirm the diagnosis.

The enzyme-linked immunosorbent assay (ELISA) is sensitive, but not as specific as the serotonin release assay (SRA), because the SRA indicates heparin-induced platelet activation. For many intensive care unit (ICU) patients, a positive ELISA test does not lead to a positive SRA, which
means these patients are unlikely to have HIT

35
Q

What is the single most effective predictor of perioperative bleeding

A

a carefully performed bleeding history

36
Q

Brief description of prothrombine time test

A

PT assesses the integrity of the extrinsic and common pathways of plasma-mediated hemostasis. It measures time required in seconds for fibrin clot formation to occur after mixing a sample of patient plasma with TF
(thromboplastin) and calcium. It is sensitive to deficiencies in fibrinogen and factors II, V, VII, or X. As three of these factors have vitamin K–dependent synthesis (factors II, VII, and X), the PT assay has been used to monitor anticoagulation with VKAs such as warfarin.
Heparin, LMWH, and fondaparinux inhibit thrombin and therefore should prolong the PT. However, most PT reagents contain heparin-binding chemicals that block this effect; thus, the PT remains normal in the setting of these medications

37
Q

Why the international normalized ratio was created?

A

The thromboplastin reagent, derived from animal or recombinant sources, can vary in its ability to bind factor VII and initiate coagulation, which limits interlaboratory comparisons. Given the importance of monitoring PT results for patients on long-term warfarin therapy, the
INR was introduced by the World Health Organization as a means of normalizing PT results among different laboratories.

38
Q

Brief description of Activated Partial Thromboplastin Time test

A

aPTT assesses integrity of the intrinsic and common pathways of plasma-mediated hemostasis. It measures the time required in seconds for fibrin clot formation to occur after mixing a sample of patient plasma with phospholipid, calcium, and an activator of the intrinsic pathway of coagulation (e.g., celite, kaolin, silica, or ellagic acid). It can detect low levels of prekallikrein; high-molecular-weight kininogen; factors XII, XI, IX, and VIII (intrinsic pathway); and low levels of factors II, V, and X and fibrinogen (common pathway). The aPTT is
more sensitive to deficiencies in factors VIII and IX than other factors in the intrinsic and common pathways. In most cases, coagulation factor levels below 30% to 40% of normal are detectable

39
Q

Crief description of Anti–Factor Xa Activity test

A

The anti–factor Xa activity assay or factor Xa inhibition test is being used with increasing frequency to monitor heparin anticoagulation instead of, or in addition to, the aPTT assay. The assay involves combining patient plasma with reagent factor Xa and an artificial substrate that releases a colorimetric signal after factor Xa
cleavage, thereby providing a functional assessment of heparin anticoagulant effect. Although aPTT values can be affected by several patient factors such as coagulation factor deficiencies, factor inhibitors, or the presence of
lupus anticoagulant, measurement of the heparin-bound AT inhibition of factor Xa activity is not influenced by these variables. Anti–factor Xa testing can also be used to measure the effect of other anticoagulants such as LMWH, fondaparinux, and factor Xa inhibitors. Data
supporting the use of anti–factor Xa over aPTT is sparse; however, it may be helpful to use anti–factor Xa testing in combination with the aPTT to monitor heparin effect

40
Q

Brief description of thrombine time test

A

The TT measures the final step of the clotting pathway where fibrinogen is converted to fibrin. It measures the time required in seconds for fibrin clot formation to occur after mixing a sample of patient plasma with calcium and thrombin. Conditions that prolong the thrombin time include therapy with anticoagulants (including heparin and DTIs), hypofibrinogenemia (<100 mg/dL), dysfibrinogenemia (presence of abnormal fibrinogen), DIC, liver disease, high concentrations of serum proteins (multiple myeloma, amyloidosis), and circulating bovine
thrombin antibodies (formed after exposure to topical bovine thrombin used for hemostasis during surgery).

41
Q

Brief description of bleeding time test

A

The bleeding time test was the first in vivo assay of platelet function. To perform the test, a blood pressure cuff is inflated on the upper part of the arm to 40 mm Hg and a standardized 9-mm long and 1-mm deep incision is made on the volar surface of the forearm.

Blood is blotted away every 30 seconds with filter paper until bleeding stops. A prolonged bleeding time (>11 minutes) can signify either platelet dysfunction or thrombocytopenia (<100,000/μL).

The bleeding time has benefits of assessing natural hemostasis, does not require specialized equipment, and is not susceptible to artifacts from anticoagulant medications

A normal bleeding time does not predict adequate hemostasis during surgery, nor
does an abnormal bleeding time predict abnormal surgical bleeding. As a result, the bleeding time may be used to help with diagnosis of inherited platelet disorders, but is not recommended as a preoperative screening test

42
Q

Crier description of activated clotting time

A

ACT measures the time in seconds for formation of a clot after a contact activation initiator (e.g., celite, kaolin) is added to a sample of freshly drawn whole blood. Because the ACT measures fibrin clot formation by way of intrin- sic and common pathways, heparin and other anticoagu- lants prolong time to clot formation. Although the aPTT assay has replaced it in most clinical situations, the ACT’s simplicity, low cost, and linear response at high heparin concentrations make it a popular perioperative coagula- tion monitor during surgical cases requiring high doses of heparin (e.g., cardiac and vascular surgery)

43
Q

How does aspirin reduces platelets aggregation?

A

There are two primary COX isozymes: COX-1 and COX-2. COX-1 maintains the integrity of the gastric lining and renal blood flow and initiates the formation of TxA2, which is important for platelet aggregation and secretion. COX-2 is responsible for synthesizing the prostaglandin mediators in pain and inflammation.

Small doses of aspirin irreversibly inhibit COX-1; however, COX-2 is 170 times less sensitive than COX-1 to aspirin, so only at high doses can aspirin irreversibly inhibit both COX-1 and COX-2. Because platelets have no deoxyribonucleic acid (DNA), they are unable to synthesize new COX-1 once aspirin has irreversibly inhibited the enzyme. As a result, despite its short half- life (15 to 20 minutes), aspirin’s platelet-inhibitory effect persists for 7 to 10 days, which is the expected lifetime of anucleated platelets. The recovery of platelet function after aspirin requires generation of new platelets. Generally, megakaryocytes produce 10% to 12% of circulating platelets daily, so near-normal hemostasis is expected in 2 to 3 days after the last dose of aspirin, assuming normal platelet turnover. Otherwise, immediate reversal can only be achieved with platelet transfusions

44
Q

Why selective cox-2 inhibitors are associated with an increase in the thrombotic risck?

A

Although COX-2 is not expressed by platelets, the increased thrombotic risk is thought to be caused, in part, by inhibition of prostacyclin (PGI2) without inhibition of TxA2, thus tipping the balance toward thrombosis

45
Q

How do Thienopyridines (e.g., clopidogrel, ticlopidine, and prasugrel) and nucleoside analogs (e.g., ticagrelor and cangrelor) interfere with platelet function?

A

They interfere with platelet function as antagonists of the P2Y12 receptor. Both classes prevent binding of ADP by the P2Y12 receptor, which impairs platelet adhesion and aggregation by preventing the expression of GP IIb/IIIa on the surface of activated platelets.

Thienopyridines are prodrugs requiring hepatic metabolism to generate the active metabolite that then irreversibly inactivates the ADP-binding site of the P2Y12 receptor. Platelet function normalizes 7 days after discontinuing clopidogrel and 14 to 21 days after discontinuing ticlopidine

Ticagrelor and cangrelor function as reversible inhibitors that change the conformation of the P2Y12 receptor.

46
Q

Why some patients are resistant to clopidogrel?

A

Clopidogrel requires metabolism by CYP2C19 for activation and as a result has wide interindividual variability in inhibiting ADP-induced platelet function. Although many factors may be involved in this variability, genetic polymorphism of CYP2C19 along with ABCB1, which affects the intestinal permeability and oral bioavailability of clopidogrel, are thought to play a significant role. Patients treated with clopidogrel who have decreased CYP2C19 activity were shown to have significantly increased risk of major cardiovascular events. The Food and Drug Administration (FDA) issued a black box warning on clopidogrel to make patients and health care providers aware that patients who are CYP2C19-poor metabolizers, who represent up to 14% of patients, are at high risk of treatment failure and that genotype testing may be helpful.

47
Q

Particularities of ticagrelor and cangrelor

A

Ticagrelor has much lower interindividual variability because it binds to a separate site on the P2Y12 receptor, and both the parent drug and the active metabolite have antiplatelet effects. Because it is much shorter acting than clopidogrel, ticagrelor must be dosed twice daily, which may be of benefit before surgery.

Cangrelor is the only P2Y12 inhibitor available for intravenous administration and exhibits rapid onset (seconds) and offset, with platelet function normalizing within 60 minutes of discontinuation. This rapid offset may allow for bridging therapy in patients with drug-eluting stents who require surger

48
Q

Mechanisms of action of GPIIb/IIIa Inhibitors and particularities of the agents in this class

A

Glycoprotein IIb/IIIa inhibitors (GPIs) (e.g., abciximab, eptifibatide, and tirofiban) prevent platelet aggregation by decreasing the binding of fibrinogen and vWF to glycoprotein IIb/IIIa receptors on the surface of activated platelets. They are administered intravenously in order to limit ongoing arterial thrombosis or to prevent formation of occlusive thrombi and restenosis in diseased vessels.

Abciximab is a noncompetitive, irreversible inhibitor of GPIIb/IIIa, whereas eptifibatide and tirofiban are competitive, reversible GPIIb/IIIa antagonists.

The inhibition provided by abciximab continues at various levels for several days after the infusion has been discontinued; however, platelet aggregation normalizes within 24 to 48 hours.

Platelet aggregation returns to normal 8 hours after discontinuing eptifibatide and tirofiban.

All of these medications can cause thrombocytopenia, but the effect is strongest with abciximab (incidence of about 2.5%

49
Q

Brief general description of vitamine K antagonists

A

Warfarin, the most frequently used oral VKA, inhibits the synthesis of factors II, VII, IX, and X and proteins C and S. Without vitamin K, these proteins do not undergo γ-carboxylation and therefore are unable to bind to phospholipid membranes during hemostasis. Warfarin has a long half-life (∼36 hours), and the complete anti- coagulant effect can take 3 to 4 days to emerge because of the long half-lives of the preexisting coagulation factors. Prothrombin (factor II) has the longest half-life (∼60 hours), whereas factor VII and protein C have the shortest half-lives (3 to 6 hours). During the initiation of warfarin therapy, early reductions in the anticoagu- lant protein C relative to other coagulation factors can produce a hypercoagulable state, resulting in thrombosis or warfarin-induced skin necrosis. As a result, patients at high risk for thromboembolism must be bridged with another anticoagulant (usually heparin) until warfarin’s full anticoagulant effect is achieved.
Warfarin is monitored using the INR, and the therapeutic range for warfarin anticoagulation is generally an INR of 2.0 to 3.0, except for patients with mechanical heart valves, where higher values are necessary (INR 2.5 to 3.5). The INR is not calibrated to evaluate nonwarfarin factor deficiencies such as from liver disease and should not be used to evaluate therapeutic effects of other anti- coagulants. Warfarin has a very narrow therapeutic win- dow, which can be easily affected by other medications, foods, and alcohol. Warfarin’s pharmacokinetics are also affected by genetic variations in cytochrome P450 (CYP2C9 gene), which can result in altered metabolism.

50
Q

Brief description of unfractionated heparin

A

UFH is a mixture of different-length polysaccharides with a high molecular weight. UFH indirectly inhibits thrombin (factor IIa) and factor Xa by binding to AT. The benefits of heparin are its short half-life and ability to be fully reversed with protamine, a positively charged protein isolated from salmon. Patients may be resistant to UFH if they have hereditary insufficiency of AT or an acquired deficiency of AT from prolonged heparin administration. Treatment should be with recombinant or plasma-derived AT concentrates. If not available, plasma transfusions may be used, but this increases the risk of transfusion-associated circulatory overload (TACO) or other transfusion reactions.
Heparin therapy is monitored with the aPTT or ACT. Full-dose heparin for cardiac surgery is administered as an intravenous bolus of 300 to 400 U/kg. An ACT greater than 400 seconds is usually considered safe for initiation of CPB. One mg protamine to 100 units of heparin is the reversal dose used at the conclusion of CPB

51
Q

Brief description of Low-Molecular-Weight Heparin and foundaparinux

A

LMWH is produced by cleaving UFH into shorter fragments, which results in greater indirect (AT-mediated) inhibition of factor Xa compared with that of thrombin (factor IIa).

Similarly, fondaparinux, a synthetic pentasaccharide of the AT binding region of heparin, selectively inhibits factor Xa via AT.

LMWH and fondaparinux III cannot be monitored using the aPTT assay, but routine laboratory monitoring is usually not needed. However, in patients with renal failure, which affects drug excretion, or in pregnant women, obese patients, and neonates for whom drug levels are less certain after subcutaneous injection, drug levels can be assessed using anti–factor Xa activity assays.

LMWH and fondaparinux have longer half-lives than heparin and can be administered subcutaneously either once or twice daily.

Protamine is only partially effective in reversing LMWH and not effective for fondaparinux.

LMWH is contraindicated in patients with HIT. Although fondaparinux does not interact with PF4 to form the antigen responsible for HIT, data supporting its use in HIT are limited.

52
Q

Brief description of direct thrombin inhibitors

A

DTIs bind directly to thrombin and do not require a cofactor such as AT to exert their effect. As their name implies, DTIs (e.g., lepirudin, argatroban, bivalirudin) bind directly to thrombin and do not require a cofactor such as AT to inhibit their activity. All DTIs inhibit thrombin in its free and fibrin-bound states, unlike heparin, which only has an effect on free thrombin.

Clinical effects can be monitored with aPTT or ACT assays.

Hirudin is a naturally occurring DTI found in leeches. Lepirudin is a recombinant hirudin analog, whereas argatroban and bivalirudin are synthetic agents.

Argatroban, which has a half-life of 45 minutes, is the pre- ferred DTI in patients with renal insufficiency because it is hepatically eliminated. Bivalirudin is a reversible DTI and is metabolized by plasma proteases and renally excreted. Bivalirudin is often chosen in patients with both renal and hepatic dysfunction because of its short half-life of 25 minutes, although dose adjustments are still necessary.

There are no antidotes for any of the DTIs, so reversal depends on their clearance. All DTIs will interfere with the INR, but argatroban has the greatest effect, which can complicate the transition to long-term warfarin anticoagulation

53
Q

Brief description of thrombolytics

A

Most thrombolytic agents are serine proteases that work by converting plasminogen to plasmin (plasminogen activators). Plasmin then degrades fibrinogen, fibrin, and crosslinked fibrin (found in the clot), thereby generating fibrin-degradation products.

Thrombolytic drugs are often divided into two categories: (1) non–fibrin-specific agents or (2) fibrin-specific agents.

Streptokinase, produced by β-hemolytic streptococci, is a non–fibrin-specific thrombolytic and was the first clinically used thrombolytic agent. Because streptokinase is a bacterial protein, it can elicit an immune response, including allergic or anaphylactic reactions.

Fibrin-specific thrombolytic drugs include recombinant tPAs such as alteplase, reteplase, and tenecteplase. The ability of a thrombolytic agent to selectively recognize plasminogen bound to fibrin surfaces rather than plas- minogen in the circulation dictates its fibrin specificity. Fibrin-specific thrombolytic agents may confer a lower risk of hemorrhagic complications by limiting lysis to the site of thrombosis; however, data regarding such a benefit are conflicting.

In addition to lysing clots, tPAs function as anticoagulants through the liberation of fibrin degradation products during fibrinolysis. These degradation products include fragment X (from fibrinogenolysis) and D-dimer (from crosslinked fibrin), which inhibit platelet aggregation. Surgery or puncture of noncompressible vessels is contraindicated within a 10-day period after the use of thrombolytic drugs.

54
Q

Brief description of antifibrinolytics

A

There are two types of antifibrinolytics: the lysine analogs (epsilonaminocaproic acid [EACA] and tranexamic acid [TXA]) and a serine protease inhibitor, aprotinin. Aprotinin was removed from the U.S. market because of concerns of renal and cardiovascular toxicity and is now only available in Europe and Canada.

The lysine analogs act to impair fibrinolysis by competitively inhibiting the binding site on plasminogen, leading to inhibition of plasminogen activation in addition to preventing plasminogen binding of fibrin. TXA and EACA likely have equivalent efficacy and decrease perioperative blood loss in cardiac surgery, liver transplantation, and orthopedic surgery.

Overall, TXA and EACA appear to be inexpensive and low-risk adjunctive agents that should be considered for use in major surgery or critical bleeding. Administration of lysine analogs perioperatively does not appear to increase the risk of thrombosis, but further studies are necessary before this can be definitively concluded.
However, there are reports of high-dose TXA causing seizures in patients undergoing cardiac surgery.This is thought to be the result of TXA binding to GABAA receptors, which in turn reduces GABAA-mediated inhibition in the central nervous system (CNS). Because of this effect, it is recommended that doses for cardiac surgery be limited to a loading dose of 10 mg/kg followed by an infusion of 1 mg/kg/hr.

55
Q

Brief description of prothrombin complex concentrate

A

factors are preserved in the inactive state, with the aim of decreasing thrombogenic risk; however, FEIBA (Factor eight inhibitor bypassing activity or Anti-Inhibitor Coagulant Complex) is a fourfactor PCC that contains activated factor VII. Although PCCs are derived from human plasma, they are treated with at least one viral reduction process, which reduces the risk for infectious and noninfectious transfusion reactions. Because of their improved safety profile and small volume of administration compared with plasma, PCCs are the first-line treatment for emergent reversal of VKAs.

56
Q

Describe the perioperative thromboembolism risk stratification

A

High
Mechanical heart valve
Rheumatic valvular heart disease
CHADS score ≥5
VTE within 3 months or history of VTE when VKAs are discontinued

Moderate
CHADS score of 3 or 4
VTE between 3 and 12 months or history of recurrence
Active cancer

Low

CHADS score 0–2
VTE >12 months prior and no other risk factors

57
Q

Perioperative management of patients using vitamine K antagonists

A

For patients taking VKAs, the current recommendation is to stop VKAs 5 days before surgery for those who are at low risk for perioperative thromboembolism. VKAs should be restarted 12 to 24 hours postoperatively if there is adequate hemostasis.

Patients at high risk of thromboembolism should be placed on bridging anticoagulation with UFH or LMWH after discontinuation of VKAs. For patients at intermediate thromboembolic risk, there is no definitive guidance regarding bridging therapy, so the approach chosen should be based on individual patient and surgical risk factors

58
Q

Perioperative management of patients using heparins

A

For those patients receiving bridging therapy with UFH, the infusion should be stopped 4 to 6 hours before
surgery and resumed without a bolus dose no sooner than 12 hours postoperatively. In surgeries with high postoperative bleeding risk, resumption of UFH should be delayed 48 to 72 hours until adequate hemostasis has been achieved.

In patients receiving bridging therapy with LMWH, the last dose of LMWH should be administered 24 hours before surgery and dosing should be resumed 24 hours postoperatively in low-bleeding-risk surgery and delayed until 48 to 72 hours postoperatively for surgeries with high bleeding risk