Hemostatic disorders

Physiology of hemostasis

As a result of injury to the blood vessel endothelium, three events take place concurrently:

  • 1.

    vasoconstriction (vascular phase),

  • 2.

    platelet plug formation (primary hemostatic mechanism—platelet phase), and

  • 3.

    fibrin thrombus formation (initiation, amplification, and propagation phases).

There are three integral components to hemostasis:

  • 1.

    endothelial cells

  • 2.

    platelets

  • 3.

    plasma coagulation factors

    Endothelial cells secrete substances that:

    • repel platelets [prostaglandin I2, adenosine diphosphate (ADP), and nitric oxide],

    • initiate coagulation (collagen, fibronectin),

    • promote platelet adhesion [von Willebrand factor (vWF)] and fibrin dissolution (tissue plasminogen activator, t-PA),

    • catalyze the inhibition of thrombin (heparin and thrombomodulin), and

    • inhibit the initiation of fibrin dissolution (t-PA inhibitor).

Participation of platelets in hemostasis is a fundamental component of the physiologic process of coagulation. Platelet interactions in coagulation are initiated by adhesion to areas of vascular injury. Subsequent activation of platelets results in release of ADP, serotonin, and calcium from “dense bodies” and fibrinogen, vWF, factor V (FV), high-molecular-weight (HMW) kininogen, fibronectin, α-1-antitrypsin, α-thromboglobulin, platelet factor 4 (PF4), and platelet-derived growth factor from α granules. Platelets provide surfaces for the assembly of coagulation factors (e.g., VIIIa/Ca21/IXa and Va/Ca21/Xa complexes). The platelets aggregate and increase the mass of the hemostatic plug. They also mediate blood vessel constriction (by releasing serotonin) and neutralize heparin.

All of the plasma coagulation factors are produced in the liver; factor VIII (FVIII) is also produced by endothelial cells. Table 13.1 lists the half-life and plasma levels of the coagulation factors. Factor II (FII), factor VII (FVII), factor IX (FIX), and factor X (FX) are vitamin K–dependent and require vitamin K in order to undergo posttranslational gamma-carboxylation. These vitamin K–dependent factors circulate in zymogen form, are activated on platelet phospholipid surfaces and, upon activation, have serine protease activity. The plasma coagulation factors work in an interdependent manner to generate thrombin (FIIa) from prothrombin (FII); thrombin then converts fibrinogen to form fibrin monomers. Fibrin monomers polymerize and establish a network. Thrombin activates factor XIII (FXIII), which in turn cross-links the fibrin network. By incorporating into the hemostatic plug, thrombin becomes inactivated. Thrombin plays a central bioregulatory role, promoting platelet aggregation and release reactions and generating a positive biofeedback loop to form more thrombin at a faster rate. Thrombin and thrombin complexed to thrombomodulin also activate thrombin activatable fibrinolysis inhibitor (TAFI), a procarboxypeptidase found in plasma, which attenuates fibrinolysis of the clot. The regulation of hemostasis is well orchestrated with its three major components (blood vessels, platelets, and plasma coagulation factors), which are integrated and interdependent.

Table 13.1
Half-life and plasma levels of coagulation factors.
Factors Common name Biologic half-life (h) Plasma concentration (nM) Plasma levels (units/dL)
I Fibrinogen 56–82 8800 200–400 a
II Prothrombin b 45–60 1400 50–150
III Tissue thromboplastin N/A 0
V Proaccelerin, labile factor 36 20 50–150
VII Proconvertin, b stable factor 5 10 50–150
VIII Antihemophilic factor 8–12 0.7 50–150
IX Christmas factor b 12–24 90 50–150
X Stuart factor b 24–60 170 50–150
XI Plasma thromboplastin antecedent 48 30 50–150
XII Hageman factor 48–52 375 50–150
XIII Fibrin-stabilizing factor 168–240 70 50–150
High-molecular-weight kininogen Fitzgerald factor 136 6000
Prekallikrein Fletcher factor N/A 450

a In mg/dL.

b Vitamin K dependent.

Primary hemostatic mechanism (platelet phase)

Primary hemostasis leads to the formation of a reversible aggregate of platelets: a temporary platelet hemostatic plug with endothelial injury exposing vWF and collagen from the subendothelial matrix to flowing blood and shear forces. Plasma vWF binds to the exposed collagen, uncoils its structure and, in synergy with collagen, supports the adhesion of platelets. Initially, the vWF interacts with the GPIb platelet receptor, tethering the platelets. As the platelet collagen receptors GPVI and α2β1 bind to collagen, the platelets adhere and become activated with a resulting release of platelet alpha and dense granule contents. Platelet activation results in a conformational change in the αIIbβ3 receptor, activating it and enhancing its avidity for vWF, for vessel wall ligands and fibrinogen. The enhanced avidity for vWF and fibrinogen mediates platelet-to-platelet interactions that eventually lead to platelet plug formation.

The fibrin thrombus formation component of hemostasis occurs in three overlapping phases ( Fig. 13.1 ):

  • 1.

    initiation

  • 2.

    amplification

  • 3.

    propagation

Figure 13.1, A cell-based model of coagulation. The three phases of coagulation occur on different cell surfaces: Initiation on the tissue-factor bearing cell: Amplification on the platelet as it becomes activated; and Propogation on the activated platelet surface.

The initiation phase begins with cell-based expression of tissue factor (TF) at the site of endothelial injury. FVII binds to the exposed TF and is rapidly activated. The FVIIa/TF complex in turn generates factor Xa (FXa) and factor IXa (FIXa). FXa can activate FV that complexes with FXa and generates small amounts of thrombin. During the amplification phase the procoagulant stimulus is transferred to the surface of platelets at the site of injury. The small amounts of thrombin enhance platelet adhesion, fully activate the platelets, and activate FV, FVIII, and FXI. In the propagation phase the “tenase” complex of FIXa FVIIIa is assembled on the platelet surface and efficiently generates FXa. Similarly, the “prothrombinase” complex of FXa FVa is assembled on the platelet surface and efficiently generates thrombin. Unlike FXa generated from TF FVIIa interactions, FXa complexed to FV is protected from inactivation by TF pathway inhibitor (TFPI), assuring adequate thrombin generation. The resulting procoagulant, thrombin, activates FXIII and cleaves fibrinopeptides A and B from fibrinogen. The residual peptide chains aggregate by means of loose hydrogen bonds to form fibrin monomers. Under the influence of FXIIIa, fibrin monomers are converted into fibrin polymers, forming a stable fibrin clot. In the presence of thrombin the mass of loosely aggregated intact platelets is transformed into a densely packed mass that is bound together by strands of fibrin to form a definitive hemostatic barrier against the loss of blood.

Platelet vessel interaction

The vasculature forms a circuit that maintains blood in a fluid state and free of leaks. With vascular injury, platelets and the coagulation system temporarily close the rent and repair the leak. Blood vessel wall characteristics exhibit properties that contribute to hemostasis or stop hemorrhage as well as prevent thrombosis. The media and adventitia of the vessel wall enable vessels to dilate or constrict. The subendothelial basement membranes contain adhesive proteins that provide binding sites for platelet and leukocytes. Remodeling that occurs after injury is enhanced by extracellular matrix metalloproteinases.

Fibrinolysis

The fibrinolytic system provides a mechanism for the removal of physiologically deposited fibrin. Clot lysis is brought about by the action of plasmin on fibrin. Fibrinolytic events are shown in Fig. 13.2 . Plasminogen from circulating plasma is laid down with fibrin during the formation of thrombin. Plasminogen is primarily synthesized in the liver and circulates in two forms, one with an NH 2 -terminal glutamic acid residue (glu-plasminogen) and a second form with an NH 2 -terminal lysine, valine, or methionine residue (lys-plasminogen). Glu-plasminogen can be converted to lys-plasminogen by limited proteolytic degradation.

Figure 13.2, The fibrinolytic pathway. Plasminogen is converted enzymatically to plasmin by t-PA or by u-PA. Plasmin cleaves fibrin and fibrinogen into fibrin degradation products. Major inhibitors of the fibrinolytic pathway are depicted. PAI-1 and PAI-2 inhibit t-PA. Plasmin activity is inhibited by a2-AP. Abbreviations: t-PA , tissue plasminogen activator; u-PA , urokinase; PAI-1 , plasminogen activator inhibitor 1; PAI-2 , plasminogen activator inhibitor 2; a2-AP , alpha 2 anti-plasmin. (T=inhibition).

Lys-plasminogen has a higher affinity for fibrin and cellular receptors; it is also more readily activated to plasmin than glu-plasminogen. Both forms of plasminogen bind to fibrin through specific lysine-binding sites. These lysine-binding sites also mediate the interaction of plasminogen with its inhibitor, α2-antiplasmin (α2AP). TAFI-mediated removal of C -terminal lysine and arginine residues will prevent high-affinity plasminogen binding and will attenuate fibrinolysis. Plasminogen is converted to its enzymatically active form, plasmin, by several activators. These activators are widely distributed in body tissues and fluids. t-PA is the principal intravascular activator of plasminogen. t-PA is a serine protease that binds to fibrin through lysine-binding sites. When t-PA is bound to fibrin, its plasmin generation efficiency increases markedly. Urokinase-type plasminogen activator, a second physiological activator of plasminogen, is present in urine and activates plasminogen to plasmin independent of the presence of fibrin. Plasmin splits fibrin and fibrinogen into fibrin-degradation products (FDPs): fragments X, Y, D, and E, some of which are measured clinically as d -dimers.

Properties attributed to the various fibrin split products (FSPs) include heparin-like effects, inhibition of platelet adhesion and aggregation, potentiation of the hypotensive effect of bradykinin and chemotactic properties (monocytes and neutrophils). Increased fibrinolysis is usually a reaction to intravascular coagulation (secondary fibrinolysis) rather than the initial event (primary fibrinolysis). The action of plasmin is negatively regulated by several inhibitors (shown in Fig. 13.2 ) that include α2AP and α2-macroglobulin. PA is in turn regulated by two inhibitors, plasminogen activator inhibitor-1 (PAI-1) and PAI-2. PAI-1 is the physiologically important of these inhibitors.

Natural inhibitors of coagulation

In addition to the physiologic role of fibrinolysis, other inhibitors play critical roles in the control of hemostasis. Table 13.2 lists the plasma fibrinolytic components and hemostatic inhibitors and their principal substrates. All members of this group have overlapping roles in the control of coagulation and fibrinolysis. Major antiproteases of this group of inhibitors include antithrombin (AT), α2AP, α2-macroglobulin, the inhibitor of the activated first component of complement (C1 inhibitor), and α1-antitrypsin.

Table 13.2
Plasma fibrinolytic components and hemostatic inhibitors.
Biologic half-life Proteases inhibited Concentration in plasma (mg/dL)
Fibrinolytic components
Plasminogen a 48 h 10–15
Plasminogen activators
Tissue 3–4 min
Urokinase 9–16 min
Plasminogen activator inhibitor a Plasminogen activator, XII a 60–200 b
Inhibitors
Antithrombin a (heparin cofactor) 17–76 h XII a , XI a , IX a , X a , thrombin, kallikrein, plasmin 10–14104–121 b
α 2 -Plasmin inhibitor a (antiplasmin) 30 h XII a , XI a , kallikrein, Plasmin, thrombin 6–880–120 b
α 2 -Macroglobulin a XII a , XI a , thrombin, kallikrein, plasmin 190–310
C1 inhibitor a XII a , kallikrein 20–25
α 1 -Antitrypsin a Thrombin, XI a , kallikrein 245–325
Thrombin activatable inhibitor of fibrinolysis 20–400 c
Tissue factor pathway inhibitor Factor VIIa/tissue factor complex Endothelial bound
Protein C a 6 h V a ,VIII a , plaminogen activator inhibitor 0.4–0.671–109 b
Protein S 60 h V a , VIII a 95–125 b
Protein C inhibitor a Protein C a 0.5

a Enzymatic activity: serine protease.

b Activity in plasma (%).

c Activity expressed as nM/L.

AT neutralizes the procoagulant thrombin, FIXa, FXa, and FXIa ( Fig. 13.3 ). When bound to circulating heparin or heparin sulfate on endothelial cells, AT undergoes a conformational change with a dramatic increase in this activity. TFPI is responsible for inactivation of the FXa/FVIIa/TF complex.

Figure 13.3, Major inhibitory proteins of coagulation. TFPI, AT, protein C and protein S are depicted with their target coagulation factor substrates. Abbreviations: TFPI , tissue factor pathway inhibitor; AT , antithrombin. (T=inhibition).

The vitamin K–dependent zymogen, protein C (PC), and its cofactor protein S (PS), which is also a vitamin K protein, play an important role in the control of hemostasis by inhibiting activated FV and VIII ( Fig. 13.3 ). Binding of thrombin to thrombomodulin on endothelial cells of small blood vessels neutralizes the procoagulant activities of thrombin and activates PC. PC binds to a specific receptor and the binding augments the activation of PC by thrombin. Activated PC (APC) inactivates FVa and FVIIIa in a reaction that is greatly accelerated by the presence of free PS and phospholipids, thereby inhibiting the generation of thrombin. Free PS itself has anticoagulant effects: it inhibits the prothrombinase complex (FXa, FVa, and phospholipid), which converts prothrombin to thrombin and inhibits the complex of FIXa, FVIIIa, and phospholipid, which converts FX to FXa.

Hemostasis in the newborn

In comparison with hemostatic mechanisms in older children and adults, those of newborn infants are not uniformly developed. Table 13.3 lists the hemostatic values in healthy preterm and term infants.

Table 13.3
Hemostatic values in healthy preterm and term infants.
Normal adults/children Preterm infant (28–32 weeks) Preterm infant (33–36 weeks) Term infant
PT (s) 10.8–13.9 14.6–16.9 10.6–16.2 10.1–15.9
aPTT (s) 26.6–40.3 80–168 27.5–79.4 31.3–54.3
Fibrinogen (mg/dL) 95–425 160–346 150–310 150–280
II (%) 100 a 16–46 20–47 30–60
V (%) 100 a 45–118 50–120 56–138
VII (%) 100 a 24–50 26–55 40–73
VIII (%) 100 a 75–105 130–150 154–180
vWF Ag (%) 100 a 82–224 147–224 67–178
vWF (%) 100 a 83–223 78–210 50–200
IX (%) 100 a 17–27 10–30 20–38
X (%) 100 a 20–56 24–60 30–54
XI (%) 100 a 12–28 20–36 20–64
XII (%) 100 a 9–35 10–36 16–72
XIII (%) 100 a 35–127 30–122
PK (%) 100 a 14–38 20–46 16–56
HMW-K (%) 100 a 20–36 40–62 50–78
Abbreviations: HMW-K , High-molecular-weight kininogen; PK , prekallikrein.

a Expressed as a percentage of activity in pooled control plasma.

Plasma factors

In newborns, FVIII and vWF are normal or higher than adult levels. Plasminogen levels are only 50% of adult values and α2AP levels are 80% of adult values, whereas PAI-1 and t-PA levels are significantly increased over adult values. The increased plasma levels of t-PA and PAI-1 in newborns on day 1 of life are in marked contrast to values from cord blood, in which concentrations of these two proteins are significantly lower than in adults. Newborns also have decreased levels of vitamin K–dependent procoagulants (FII, FVII, FIX, FX) and activity of anticoagulant factors, especially AT, PC, and PS.

Blood vessels

  • Capillary fragility is increased.

  • Prostacyclin production is increased.

Platelets

  • Platelet adhesion is increased due to increased vWF and increased HMW vWF multimers.

  • Epinephrine-induced aggregation is decreased due to decreased platelet receptors for epinephrine.

  • Ristocetin-induced aggregation is increased due to increased vWF and increased HMW vWF multimers.

  • Platelet activation is increased: as evidenced by elevated levels of thromboxane A2, β-thromboglobulin, and PF4.

Approach to a bleeding child

Evaluation of a patient for a hemostatic defect generally entails the following:

  • 1.

    Detailed history (see Table 13.4 for initial features suggestive of pathological bleeding in children):

    • a.

      Symptoms: epistaxis, gingival bleeding, easy bruising, menorrhagia, hematuria, neonatal bleeding (heel stick, umbilicus), gastrointestinal (GI) bleeding, hemarthrosis, prolonged bleeding after lacerations, heavy menses.

    • b.

      Response to hemostatic challenge: circumcision, surgery, phlebotomy, immunization/intramuscular injection, suture placement/removal, dental procedures.

    • c.

      Underlying medical conditions: known associations with hemostatic defects (liver disease, renal failure, vitamin K deficiency).

    • d.

      Medications: antiplatelet drugs (nonsteroidal antiinflammatory drugs), anticoagulants [warfarin, heparin, low-molecular-weight heparin (LMWH)], antimetabolites ( l -asparaginase), prolonged use of antibiotics causing vitamin K deficiency, long-term use of iron suggestive of ongoing blood loss causing iron-deficiency anemia.

    • e.

      Family history: symptoms, response to hemostatic challenge (siblings, parents, aunts, uncles, grandparents), red cell transfusions after surgeries, iron deficiency after surgery or menorrhagia, hemoperitoneum/hemorrhagic ovarian cysts in a menstruating female.

    Table 13.4
    Initial features suggestive of pathologic bleeding in children.
    Reproduced with permission from Acharya, S.S., 2013. Rare bleeding disorders in children: identification and primary care management. Pediatrics 132, Page 883, Copyright© 2013 by the AAP.
    • Age related

    • Bleeding (e.g., umbilical stump bleeding, intracranial hemorrhage, excessive and prolonged bleeding postcircumcision or after heel stick or intramuscular injection) during neonatal period.

    • Palpable and multiple bruises in infants and older children who are not independently mobile.

    • Persistent palpable bruising in an older mobile child.

    • Spontaneous bleeding in the absence of anatomic causes.

    • Personal history of

    • Recurrent (especially excessive and spontaneous) mucocutaneous bleeding.

    • Atypical bleeds (e.g., hemarthroses, retroperitoneal bleeding), whether spontaneous or provoked.

    • Excessive or prolonged bleeding after hemostatic challenges (i.e., trauma, dental procedures, or surgery).

    • Menorrhagia in adolescent girls: menstrual bleeding for >7 days or 80-mL blood loss per menstrual cycle [as evidenced by soaking through a pad or tampon within 1 h or change of pads or tampons every hour or passage of large (>1.1-in. diameter) clots].

    • Traumatic bleeding that is out of proportion to or inconsistent with reported injury (consider nonaccidental trauma).

    • Family history of

    • Recurrent bleeding symptoms.

    • Excessive or prolonged bleeding after trauma or invasive procedures.

    • Known or suspected bleeding diatheses.

    • Physical findings

    • Multiple bleeding stigmata.

    • Physical findings suggestive of specific underlying causes (e.g., petechiae in platelet disorders, jaundice in liver disease, hypermobility, vascular malformations, musculoskeletal abnormalities).

    • Pallor/anemia.

  • 2.

    Complete physical examination.

    Signs consistent with past coagulopathy: petechiae, ecchymosis, hematomas, synovitis/joint effusion, arthropathy, muscle atrophy, evidence of joint laxity or hyperextensibility which can exacerbate the bleeding phenotype. The Beighton score ( Fig. 13.4 ) is useful in evaluating hyperextensibility in children older than 7 years of age. In very young patients, parental joint mobility should be assessed.

    Figure 13.4, Beighton Score for Joint Hypermobility assessment. Degree of mobility assessed by passive maneuvers in 5 joints. Total score: 0–9. Hypermobility score: ≥5.

    Several congenital syndromes such as Down syndrome, Turner syndrome, Noonan syndrome, and Jacobsen syndrome are associated with underlying bleeding diathesis and close attention must be paid to the bleeding history while evaluating these patients.

  • 3.

    Laboratory evaluation (see Table 13.5 ).

    Table 13.5
    Coagulation tests and normal values.
    Test Normal value Clinical application
    Platelet function
    Template bleeding time (min) <9 Crude, lack of reproducibility
    Platelet aggregation Described in Chapter 12 on platelets
    Platelet factor 3 availability Screens for platelet procoagulant activity
    Clot retraction Starts at hour 1; completes at hour 24 Measures platelet interaction with fibrin
    Intrinsic system
    Activated partial thromboplastin time (s) 25–35
    Extrinsic system
    Prothrombin time (s) 10–12
    Factor assays See Tables 13.1 and 13.3
    Thrombin time (s) <24 Prolonged in hypofibrinogenemia, dysfibrinogenemia, hypoalbuminemia, liver disease, neonates
    Reptilase time (s) <25 Modification of thrombin time, unaffected by presence of heparin
    Antiphospholipid assays
    Dilute Russell’s viper venom time (s) 29–42
    Kaolin clotting time (s) Sensitive test even in presence of heparin
    Fibrinolytic system
    Euglobulin clot lysis time (mins) 90–240 Prolonged with hypofibrinolysis

Initial screening tests

  • 1.

    Complete blood count (CBC): quantitative assessment of platelets and review of blood smear to assess platelet morphology.

  • 2.

    Assessments of platelet function:

    • a.

      Platelet function analyzer (PFA-100): assesses flow through a membrane with an aperture coated with platelet agonists; membrane closure time is measured in response to ADP and to epinephrine. The closure time is often prolonged with impaired platelet function and von Willebrand disease (vWD).

    • b.

      Bleeding time should not be used because of difficulties in validation.

  • 3.

    Coagulation factor screening tests:

    • a.

      From a laboratory perspective the coagulation system is divided into the intrinsic pathway, the extrinsic pathway, and the common pathway. Such an artificial division is not based on coagulation physiology but is useful for conceptualizing in vitro laboratory testing ( Fig. 13.5 ).

      Figure 13.5, A conceptualization of commonly used screening tests of coagulation and the coagulation parameters they measure. Abbreviations: PTT , partial thromboplastin time; PT , prothrombin time.

  • 4.

    Prothrombin time (PT) assay (assesses the extrinsic system): this test utilizes tissue thromboplastin and calcium chloride, to initiate the formation of thrombin via the extrinsic pathway. The international normalized ratio (INR) is used to correct for differences between thromboplastin sources across laboratories.

  • 5.

    Activated partial thromboplastin time (aPTT) assay (assesses the intrinsic system): this test utilizes a phospholipid reagent, a particulate activator (e.g., ellagic acid, kaolin, silica, soy extract) and calcium chloride to start the enzyme reaction that leads to the formation of thrombin via the intrinsic pathway.

Common confirmatory coagulation assays

  • 1.

    Fibrinogen: quantitative measurement of fibrinogen, deficiency suggested when both the PT and the aPTT are prolonged.

  • 2.

    Thrombin time (TT): prolonged when fibrinogen is reduced or abnormal, in the presence of inhibitors (FDPs, d -dimers) and in the presence of thrombin-inhibiting drugs and hypoalbuminemia. This is a useful test to diagnose dysfibrinogenemia (qualitatively abnormal fibrinogen). Also useful when both the PT and aPTT are prolonged. The reptilase time is a modification of the TT, using the purified enzyme reptilase instead of thrombin and is unaffected by heparin and heparin-like anticoagulants.

  • 3.

    Mixing studies (performed to evaluate a prolonged PT or aPTT): the respective assay is performed following the addition of normal pooled plasma to patient plasma. Normalization indicates a clotting factor deficiency that was corrected by addition of normal pooled plasma. Continued prolongation indicates the presence of a coagulation inhibitor. Such inhibitors may be physiologically relevant or only detected in vitro.

  • 4.

    Clotting factor activity assays: performed to identify clotting factor deficiencies if mixing studies normalize. FXII, FXI, FIX, and FVIII assays are useful if the aPTT normalizes in mixing studies. The FVII assay is useful if the PT normalizes in mixing studies. FX, FV, FII, and fibrinogen assays are useful if both PT and aPTT normalize in mixing studies. (Note: FXIII deficiency does not result in prolongation of the PT or aPTT.); confirmation needs assays for FXIII antigen and FXIII activity; the 5-M urea lysis assay should not be used to confirm FXIII deficiency given its poor sensitivity.

  • 5.

    von Willebrand panel: von Willebrand antigen (vWF:Ag) is the quantitative assay for vWF, von Willebrand Ristocetin cofactor activity (vWF:RCo) is the functional or qualitative assay for VWF. They are both useful when PFA-100 closure time is prolonged or when vWD is suspected. vWF multimers are useful to diagnose qualitative VWF abnormalities (type 2 vWD) when discrepancies between vWF:Ag (normal to low) and vWF:RCo (extremely low); ratio of vWF:RCo to vWF:Ag<0.6).

  • 6.

    Platelet aggregation studies: a qualitative assessment of platelet function, useful when platelet function disorders are suspected or PFA-100 closure time is prolonged.

Global hemostatic tests

Hemostasis is a complex interplay of simultaneously occurring events, with current assays only reflecting snapshots of this dynamic process. Global hemostatic tests can provide detailed information on thrombin generation and processes downstream, including fibrin polymerization and fibrin dissolution.

  • 1.

    Thrombin generation assay: the calibrated automated thrombogram system uses a fluorogenic substrate to continuously measure the generated thrombin. The endogenous thrombin potential, which can be measured by calculating the area under the curve from the thrombogram, has shown correlation with the bleeding phenotypes in hemophilia, in hemophilia patients with inhibitors, and factor XI deficiency.

  • 2.

    Viscoelastic tests.

Thromboelastography (TEG) is performed on whole blood, assessing the viscoelastic property of clot formation under low shear condition after the addition of specific coagulation activators. Improvement in the methodology is widely expanding the use of TEG from preclinical research settings to point-of-care testing in intensive care units and operating rooms. The device has a metal pin suspended by a torsion wire immersed into a cup that holds the whole blood. Once clotting starts, fibrin strands formed increase the torque between the pin and the cup that is measured electronically. TEG provides various data relating to clot formation and fibrinolysis (the lag time before the clot starts to form, the rate at which clotting occurs, the maximal amplitude of the trace or clot strength, and the extent and rate of amplitude).

Rotational thromboelastography is almost identical to TEG; however, instead of the cup rotating, here the sensor rod rotates and is operator independent. The output data are named differently as also different assays are available for analysis of different aspects of the coagulation system such as EXTEM (extrinsic pathway), INTEM (intrinsic pathway), FIBTEM (fibrinogen contribution), HEPTEM (heparin effect), and APTEM (thrombolysis reversal).

Preoperative evaluation of hemostasis

  • 1.

    History: the history is perhaps the most important element of the evaluation. In an effort to standardize bleeding histories, a number of quantitative bleeding assessment tools (BATs) have been developed and validated in individuals with known vWD (Vicenza-based BAT, the condensed Molecular and Clinical Markers for the Diagnosis and Management of Type 1 vWD Questionnaire (MCMDM-1 vWD) International Society on Thrombosis and Haemostasis (ISTH), and the more pediatric-specific Pediatric Bleeding Questionnaire (PBQ)), although whether these tools can directly predict future bleeding episodes needs further study.

    • a.

      If negative: no coagulation tests are indicated.

    • b.

      If positive or unreliable: the following tests should be performed: CBC, PFAs, PT, aPTT, and fibrinogen, TT, von Willebrand panel.

  • 2.

    Abnormal tests require further investigation (see Fig. 13.6 ).

    Figure 13.6, Coagulation tests and interpretation. Abbreviations: PTT , activated partial thromboplastin time; PT , prothrombin time; TT , thrombin time.

  • 3.

    In a patient with a significant bleeding history, if all screening tests and von Willebrand panel are normal, consider FXIII, PAI-1 activity, α2AP, and platelet aggregation studies, vitamin C levels and rule out hyperextensibility on clinical examination (see Figure 13.4 ).

Acquired coagulation factor disorders

Vitamin K deficiency

The normal full-term infant is born with levels of FII, FVII, FIX, and FX that are low by adult standards ( Table 13.3 ). The coagulation factors drop even lower over the first few days of life, reaching their nadir on the third day. This is due to the low body stores of vitamin K at birth. As little as 25-μg vitamin K can prevent this fall in activity of the vitamin K–dependent clotting factors. The vitamin K content of cow’s milk is only about 6 μg/dL and that of breast milk 1.5 μg/dL. Moreover, breastfed infants are colonized by lactobacilli that do not synthesize gut vitamin K. It is a combination of low initial stores and subsequent poor intake of vitamin K that occasionally produces an aggravation of the coagulation defect causing primary hemorrhagic disease of the newborn. Vitamin K deficiency results in hemorrhagic disease between the second and fourth days of life and is manifested by GI hemorrhage, hemorrhage from the umbilicus, or internal hemorrhage (classic hemorrhagic disease of the newborn). Bleeding attributable to this cause is responsive to parenteral vitamin K therapy; for this reason, parenteral vitamin K is routinely administered to newborns. Serious occurrences of vitamin K deficiency bleeding continue to occur due to parental refusal of vitamin K prophylaxis after birth and increased prevalence of home deliveries with the aid of midwives who may not be licensed to administer intramuscular vitamin K after birth.

In premature infants with low birth weight, both the vitamin K stores and the level of coagulation factors are lower than in term infants. The response to vitamin K is slow and inconsistent, suggesting that the immature liver has reduced synthetic capability. Maternal ingestion of certain drugs may result in neonatal hypoprothrombinemia and reduction in FVII, FIX, and FX, resulting in early hemorrhagic disease of the newborn. These drugs include oral anticoagulants and anticonvulsants (phenytoin, primidone, and phenobarbital). This can be prevented by administration of vitamin K to the mother 2–4 weeks prior to delivery.

Late hemorrhagic disease of the newborn occurs between day 7 and 6 months of life in the absence of adequate vitamin K prophylaxis at birth. It may be idiopathic or exacerbated by malabsorption or liver disease. Consider investigating for biliary atresia, alpha 1 antitrypsin deficiency, and celiac disease, cystic fibrosis associated with malabsorption if hemorrhagic disease of the newborn occurs after adequate vitamin K prophylaxis at birth. Table 13.6 lists the laboratory findings in vitamin K deficiency in relationship to the findings in liver disease and disseminated intravascular coagulation (DIC). Any transient inability of the newborn liver to synthesize necessary coagulation factors, even in the presence of vitamin K, can result in hemorrhagic disease that is nonresponsive to vitamin K therapy.

Table 13.6
Laboratory findings in vitamin K deficiency, liver disease, and disseminated intravascular coagulation.
Component Vitamin K deficiency Liver disease DIC
Red cell morphology Normal Target cells Fragmented cells, burr cellshelmet cells, schistocytes
aPTT Prolonged Prolonged Prolonged
PT Prolonged Prolonged Prolonged
Fibrin split products Normal Normal or slightly increased Markedly increased
Platelets Normal Normal Reduced
Factors decreased II, VII, IX, X I, II, V, VII, IX, X Assays are of limited utility

Hepatic dysfunction

Hepatic dysfunction as a result of immaturity, infection, hypoxia, or under perfusion of the liver can all result in transient inability of the liver to synthesize coagulation factors. This is more prominent in small premature infants. The sites of bleeding in these cases are usually pulmonary and intracerebral with a high mortality. Other causes of hepatocellular dysfunction affecting patients of all ages include hepatitis, cirrhosis, Wilson disease, and Reye syndrome. In liver disease, vitamin K–dependent factors, FV, and fibrinogen are usually decreased, and FSPs may be elevated due to impaired clearance ( Table 13.6 ). In contrast, FVIII levels are usually normal. There is no response to vitamin K. There is usually a clinical response to clotting factor replacement therapy, using fresh frozen plasma (FFP) and cryoprecipitate (replacement guidelines are the same as those outlined in Table 13.7 ).

Table 13.7
Treatment of disseminated intravascular coagulation and purpura fulminans.
Treatment of the underlying disorder

  • Treatment of infections with appropriate antiinfectives

  • (antibiotics, antiviral drugs, antifungal drugs)

  • Correction of electrolyte imbalances, acidosis, and shock

  • Appropriate antineoplastic therapy

  • Removal of triggering stimulus

Replacement therapy as indicated

  • Platelet concentrates (1 unit/10 kg)

  • Cryoprecipitate (50–100 mg/kg fibrinogen) a

  • Fresh frozen plasma (10–15 mL/kg, initially; may need 5 mL/kg q6h)

Intravenous heparinization b
Intravenous direct thrombin inhibitors
Antiplatelet drugs
Antithrombin concentrate
Activated protein C concentrate

a One bag of cryoprecipitate contains about 200 mg fibrinogen.

b See “Heparin therapy” section.

Disseminated intravascular coagulation

DIC is characterized by the intravascular consumption of platelets and plasma clotting factors. Widespread coagulation within the vasculature results in the deposition of fibrin thrombi and the production of a hemorrhagic state when the rapid consumption of platelets, pro-, and anticoagulant factors results in levels inadequate to maintain hemostasis. The accumulation of fibrin in the microcirculation leads to mechanical injury to the red cells, resulting in erythrocyte fragmentation and microangiopathic hemolytic anemia. Widespread activation of the coagulation cascade rapidly results in the depletion of many clotting factors as fibrinogen is converted to fibrin throughout the body as follows:

  • 1.

    The generation of thrombin results in intravascular coagulation, rapidly falling platelet count, fibrinogen, and FV, FVIII, and FXIII levels. Paradoxically, in vitro bioassays for these factors may be elevated owing to generalized activation of the coagulation system.

  • 2.

    Concurrently, plasminogen is converted to its enzymatic form (plasmin) by t-PA. Plasmin breaks down fibrinogen and fibrin (secondary fibrinolysis) into FSPs, resulting in clot lysis.

Diagnosis of DIC relies on the presence of a well-defined clinical situation associated with a thrombo-hemorrhagic disorder. Table 13.6 lists the typical diagnostic findings in DIC. The ISTH DIC scoring system used in adults has been validated in pediatrics and can be used to establish diagnosis. Disease states associated with DIC and low-grade DIC are listed in Table 13.8 . Generally available treatment options for the treatment of DIC are shown in Table 13.7 .

Table 13.8
Disease states associated with disseminated intravascular coagulation.
Causative factors Clinical situation
Tissue injury
  • Trauma/crush injuries

  • Head injury

  • Major surgery

  • Heat stroke

  • Burns

  • Venoms

  • Malignancy

  • Obstetrical accidents

  • Amniotic fluid embolism

  • Placental abruption

  • Stillborn fetus

  • Abortion

  • Fat embolism

Endothelial cell injury Infection (bacterial, viral, protozoal)
AND/OR Immune complexes
Abnormal vascular surfaces
  • Eclampsia

  • Postpartum renal failure

  • Oral contraceptives

  • Cardiopulmonary bypass

  • Giant hemangioma

  • Vascular aneurysm

  • Cirrhosis

  • Malignancy

  • Respiratory distress syndrome

Platelet, leukocyte, or red cell injury
  • Incompatible blood transfusion

  • Infection

  • Allograft rejection

  • Hemolytic syndromes

  • Drug hypersensitivity

  • Malignancy

Localized intravascular coagulopathy
  • Kasabach–Merritt syndrome

  • Chronic inflammatory disorders

  • Arteriovenous fistulae

  • Vascular prosthesis

  • Glomerulonephritis

Low-grade DIC has the potential to accelerate into fulminant DIC. Careful monitoring in terms of detecting the presence of fragmented red blood cells, mild decrease in platelets, and low fibrinogen levels usually indicates high fibrinolysis and increased risk of bleeding. Treatment of low-grade DIC involves treating the underlying disease triggering it.

Inherited coagulation factor disorders

Table 13.9 lists the genetics, prevalence, coagulation studies, and symptoms of inherited coagulation factor disorders.

Table 13.9
Genetics, prevalence, coagulation studies and symptoms of inherited coagulation factor deficiencies.
Recent data from EN-RBD study—Peyvandi, F., Mannucci, P.M., Garagiola, I., et al., 2016. A randomized trial of factor VIII and neutralizing antibodies in hemophilia A. N. Engl. J. Med. 374 (21), 2054–2064.
Factor deficiency Genetics Estimated prevalence Prevalence of ICH (upper limits) APTT PT Associated with bleeding episodes
Afibrinogenemia AR 1:500,000 10% P P ++
Dysfibrinogenemia AR 1:1million Single case N/P P +/− thrombosis
II AR 1:2 million 11% P P ++
V (parahemophilia) AR 1:1 million 8% of homozygotes P P ++
VII AR 1:500,000 4–6.5% N P + b
VIII (hemophilia A) XLR 1:5000 males 5–12% P N +++
von Willebrand’s disease 1:1000 Extremely rare
Type 1 AD N/P N +
Type 2 AD N/P N ++
Type 3 AR P N ++
IX (hemophilia B) XLR 1;30,000 males 5–12% P N +++
X AR 1:1 million 21% P N ++
XI (hemophilia C) AV 1:1 million Extremely rare P N + b
XII AD P N
XIII AR 1:2 million 33% N N + c
Prekallikrein (Fletcher trait) AD P a N
HMW kininogen (Fitzgerald trait) AR P N
Passovoy (?) AR P N +/–
Prolonged when associated with platelet dysfunction.
Notes : Bleeding episodes occur in most individuals homozygous for the disorder. However, with FXI and FVII deficiency, there is no correlation between factor levels and bleeding phenotype. Most studies consider factor levels <10% to be associated with bleeding symptoms. Abbreviations: AD , Autosomal dominant; AR , autosomal recessive; AV , autosomal variable; BT , bleeding time; HMW , high-molecular-weight; N , normal; P , prolonged; XLR , X-linked recessive.

a Shortened with prolonged exposure to kaolin.

b Bleeding episodes occur in most individuals homozygous for the disorder. However, with FXI and FVII deficiency there is no correlation between factor levels and bleeding phenotype. Most studies consider factor levels < 10% to be associated with bleeding symptoms, –recent data from EN-RBD study- Peyvandi F. et al., 2012.

c Umbilical stump bleeding; need FXIII activity and gene sequencing for FXIII—A and B—subunit for accurate diagnosis. The 5-M urea lysis solubility test not a sensitive test for diagnosis.

Hemophilia A and B

Hemophilia A is an X-linked recessive bleeding disorder resulting in decreased blood levels of functional procoagulant FVIII (VIII: C, antihemophilic factor). Hemophilia B is also an X-linked recessive disorder and is indistinguishable from hemophilia A with respect to its clinical manifestations. In hemophilia B the defect is a decreased level of functional procoagulant FIX (IX: C, plasma thromboplastin component or Christmas factor). The incidence of hemophilia A is approximately 1 per 5000 males and hemophilia B around 1 in 30,000. Thus FVIII deficiency accounts for 80–85% of cases of hemophilia, with FIX deficiency accounting for the remainder.

Genetics

Given X-linked recessive inheritance females are carriers for hemophilia or may have mild hemophilia depending on factor levels and accompanying bleeding symptoms. Most usually have variable factor levels but typically will have enough levels to be in the hemostatic range. Excessive lionization, however, may lead to symptomatic females with undetectable factor levels and bleeding symptoms requiring management as their male counterparts. The FVIII common intron 22 inversion, resulting from an intrachromosomal precombination, is identifiable in 45% of severe hemophilia A patients. For the remaining 55% of patients with severe hemophilia A, as well as all those with mild and moderate hemophilia A, the molecular defects can usually be detected by efficient screening of all 26 FVIII exons and splice junctions. Targeted mutation analysis is the most accurate test for carrier detection and prenatal diagnosis for severe hemophilia A. For rare patients in whom a precise mutation cannot be identified or gene sequencing is not an option, intragenetic and extragenetic linkage analysis of DNA polymorphisms can be useful with up to 99.9% precision (when an affected male patient and his related family members are available). Preimplantation genetic diagnosis is a reproductive option available to carrier females. When definitive diagnosis of the carrier state cannot be made, determination of the FVIII/vWF:Ag ratio (<1.0) can be used to detect 80% of hemophilia A carriers with 95% accuracy when done in laboratories with careful standardization procedures.

Hemophilia B carriers have a wide range of FIX levels but, in a subset of cases, can be detected by the measurement of reduced plasma FIX activity (60–70% of cases). The FIX gene is located centromeric to the FVIII gene in the terminus of the long arm of the X chromosome. The 34-kb FIX coding sequence comprises eight exons and encodes a 461-amino-acid precursor protein that is approximately one-third the size of the FVIII complementary Deoxyribonucleic Acid (cDNA). Because of the smaller gene size, FIX mutations can be identified in nearly all patients. Direct FIX mutation testing is available through DNA diagnostic laboratories, with linkage analysis used in those cases where the responsible mutation cannot be identified.

Prenatal diagnosis of hemophilia can be performed by either chorionic villus sampling at 10–12 weeks gestation or by amniocentesis after 15 weeks gestation. If DNA analysis is not available or if a woman’s carrier status cannot be determined, fetal blood sampling can be performed at 18–20 weeks gestation for direct fetal FVIII plasma activity level. The normal fetus at 18–20 weeks gestation has a very low FIX level, which an expert laboratory can distinguish from the virtual absence of FIX in a fetus with severe hemophilia B.

Clinical course of hemophilia

Hemophilia should be suspected when unusual bleeding is encountered in a male patient. Clinical presentations of hemophilia A and hemophilia B are indistinguishable. The frequency and severity of bleeding in hemophilia are usually related to the plasma levels of FVIII or FIX ( Table 13.10 ), although some genetic modifiers of hemophilia severity have been identified. The median age for first joint bleed is 10 months, corresponding to the age at which the infant becomes mobile. Table 13.11 shows the common sites of hemorrhage in hemophilia. The incidence of severity and clinical manifestations of hemophilia are listed in Table 13.12 . For hemophilia B the Leyden phenotype (severe hemophilia as a child that becomes mild after puberty) has been described in families with defects in the androgen-sensitive promoter region of the gene.

Table 13.10
Relationship of factor levels to severity of clinical manifestations of hemophilia A and B.
Type Percentage factor VIII/IX Type of hemorrhage
Severe < 1 Spontaneous; hemarthroses and deep soft tissue hemorrhages
Moderate 1–5 Gross bleeding following mild-to-moderate trauma; some hemarthrosis; seldom spontaneous hemorrhage
Mild 5–40 Severe hemorrhage only following moderate-to-severe trauma or surgery
High-risk carrier females variable Gynecologic and obstetric hemorrhage common, other symptoms depend on plasma factor level

Table 13.11
Common sites of hemorrhage in hemophilia.
Hemarthrosis
Intramuscular hematoma
Hematuria
Mucous membrane hemorrhage

  • Mouth

  • Dental

  • Epistaxis

  • Gastrointestinal

High-risk hemorrhage

  • Central nervous system

  • Intracranial

  • Intraspinal

  • Retropharyngeal

  • Retroperitoneal

  • Hemorrhage causing compartment syndrome/nerve compression

  • Femoral (iliopsoas muscle)

  • Sciatic (buttock)

  • Tibial (calf muscle)

  • Perineal (anterior compartment of leg)

  • Median and ulnar nerve (flexor muscles of forearm)

Table 13.12
Incidence of severity and clinical manifestations of hemophilia.
Severity Severe Moderate Mild
Incidence
Hemophilia A 70% 15% 15%
Hemophilia B 50% 30% 20%
Bleeding manifestations
Age of onset ≤1 year 1–2 years 2 years (adult)
Neonatal hemorrhages
Following circumcision Common Common None
Intracranial Occasionally Rare Rare
Post Neonatal period
Muscle/joint hemorrhage Spontaneous Following minor trauma Following major trauma
CNS hemorrhage High risk Moderate risk Rare a
Postsurgical hemorrhage Common Common Rare a
Oral hemorrhage b Common Common Rare a

a FVIII, > 25; FIX, > 15.

b Following trauma or tooth extraction.

Managing newborns with known or suspected hemophilia

Known female carriers or females with positive family history should plan deliveries at an obstetric center affiliated with hemophilia centers whenever possible, although 30% of newborns with hemophilia due to de novo mutations will not have a family history. There is no contraindication to vaginal delivery and the option of elective C-section should be considered based on obstetrical factors and individualized. Forceps application, vacuum delivery, fetal scalp electrodes, and fetal blood sampling should be avoided. Uncontaminated cord blood should be collected and sent to a laboratory that can expediently run FVIII/FIX levels. Mild FVIII deficiency may be missed at birth and any low normal levels should be repeated at approximately 6 months of life. FIX levels could be physiologically low at birth and should be repeated beyond age 3 months to confirm a diagnosis of mild/ moderate hemophilia B. Intramuscular injections should be avoided in patients with severe hemophilia to avoid muscle bleeds. If heelsticks are conducted, adequate pressure should be applied to achieve hemostasis. Acute bleeding should be managed by recombinant factor support. 1-Deamino-8- d -arginine vasopressin (DDAVP) is strictly contraindicated in newborns due to the risk for hyponatremic seizures. Screening cranial ultrasound should be performed to rule out intracranial hemorrhage (ICH) in all newborns diagnosed with hemophilia A/B. In a symptomatic newborn with suspected ICH but a normal ultrasound, magnetic resonance imaging (MRI)/computed tomography (CT) scan should be pursued. The decision for circumcision should involve consultation with a hematologist. For newborns without forewarning who have bleeding symptoms and an elevated aPTT, FFP treatment (15–25 mL/kg) can be initiated prior to receiving confirmatory factor levels.

Treatment (factor replacement therapy)

Factor replacement therapy is the mainstay of hemophilia treatment. The degree of factor correction required to achieve hemostasis is largely determined by the site and nature of the particular bleeding episode. Commercially available products for replacement therapy are listed in Table 13.13 . Source plasma for all plasma-derived factor concentrates undergoes donor screening and nucleic acid testing for a variety of viral pathogens. In addition, all plasma-derived and many recombinant factor concentrates undergo a viral inactivation treatment, typically with solvent detergent, wet or dry heat treatment, pasteurization, or nanofiltration. Recombinant factor concentrates are widely accepted as the treatment of choice for previously untreated patients, minimally treated patients, and patients who have not had transfusion-associated infections. These products, however, need frequent administration via intravenous (IV) infusions due to their short half-life.

Table 13.13
Commercially available coagulation factor concentrates.
Class Product Dosage (prophylaxis) Dosage <12 years Half life (hours) Primary use
Plasma derived Koate-DVI 16.12 Hemophilia A
Humate P 12.2 Hemophilia A/von Willebrand
Alphanate SD 17.9 Hemophilia A/von Willebrand
Hemofil M 14.8 Hemophilia A
Monoclate P Hemophilia A
Alphanine 21.3 Hemophilia B
Mononine 22.6 Hemophilia B
Recombinant Recombinate 11.16 Hemophilia A
Helixate Hemophilia A
ReFacto Hemophilia A
Novoeight 20–50 IU/kg 3/wk 25–60 IU/kg 3/wk 7.7–10 Hemophilia A
Nuwiq 30–40 IU/kg 2 days 30–50 IU/kg 2 days–3/wk 11.9–17.4 Hemophilia A
Kogenate FS 25 IU/kg 2 days 14.6 Hemophilia A
Kovaltry 20–40 IU/kg 2–3/wk 25–50 IU/kg 2–3/wk 11.7–14.3 Hemophilia A
Advate 20–40 IU/kg 2–3 days 12.03 Hemophilia A
Xyntha 11.2 Hemophilia A
Benefix 20.2 Hemophilia B
Ixinity 24 Hemophilia B
Rixubis 40–60 IU/kg 2/wk 60–80 IU/kg 2/wk 23.2–27.7 Hemophilia B
EHL products
Fc fusion Eloctate 50 IU/kg 4 days 12.7–19.7 Hemophilia A
Fc fusion Alprolix 50 IU/kg /wk 60 IU/kg/wk 86–97 Hemophilia B
Albumin fusion Idelvion 25–40 IU/kg/wk 40–55 IU/kg/wk 104–118 Hemophilia B
PEGylated Adynovate 40–50 IU/kg 2/wk 55 IU/kg 11.8–14.69 Hemophilia A
PEGylated Esperoct 50 IU/kg 4 days 65 IU/kg 2/wk 13.8–21.7 Hemophilia A
PEGylated Jivi 30–40 IU/kg 2/wk 17.4–21.4 Hemophilia A
PEGylated Rebinyn Not indicated 103–114 Hemophilia B
Single chain Afstyla 20–50 IU/kg 2–3/wk 50 IU/kg 10.2–14.3 Hemophilia A
Bispecific monoclonal antibody Hemlibra 3 mg/kg sc/wk×4 weeks followed by 1.5 mg/kg sc/wk Hemophilia A
Prothrombin complex Profilnine SD 24.68 Hemophilia B
Bebulin VH Kcentra 19.4–24.6 Hemophilia BAnticoagulant reversal
aPCC FEIBA/Autoplex 85 units/kg/2 days Hemophilia A+B inhibitor
Bypassing agent NovoSeven 90 µg/kg 2 h (treatment dose) 2.6–3.1 Inhibitor bypass therapy, FVII deficiency, Glanzman
Specialty items Proplex T FVII replacement (3.5 U FVII/U FIX)
Abbreviation: SC , Subcutaneously.

Extended half-life (EHL) recombinant factor products with the FVIII or FIX protein fused to the Fc portion of immunoglobulin 1 are available, which prolong the half-life of the factor in circulation. This enables prophylaxis dosing regimens occurring every 3–5 days for FVIII-deficient patients and 7–10 days dosing for FIX-deficient patients. PEGylated products are available that have longer half-lives. By increasing the molecular mass, glomerular filtration, proteolytic degradation, and clearance are reduced leading to an increased half-life of the PEGylated protein. Several other strategies being investigated include single-chain rFVIII that more effectively binds to vWF, bioengineered antibodies, and short interfering RNAs that slow down the endogenous AT synthesis.

Emicizumab is a bispecific humanized antibody acting as an FVIIIa “mimic” binding to FIXa and FXa and thereby accelerating progression of the coagulation cascade toward thrombin generation. The HAVEN trials established that emicizumab prophylaxis can achieve remarkable reduction in bleeding rates regardless of age or FVIIIa inhibitor status. Its singular advantage is that it can be given subcutaneously at weekly/bimonthly schedules. Breakthrough bleedings during the trials did require additional hemostatic support. The main safety concerns are from increased thrombotic risks and thrombotic microangiopathy, paucity of assays to assess hemostatic drug levels, and potential for the development of antidrug antibodies. Patients with high activity levels or involved in high-impact sports may not be suitable candidates for this therapy.

Gene therapy offers the promise of curing hemophilia by inserting the deficient gene into the patient’s tissue that can then restore circulating factor levels. Notably, the gene for FIX is small and easy to insert into many vectors. In 2011 Nathwani and colleagues successfully transduced 10 hemophilia B patients using adeno-associated viral (AAV) vectors, with persistent FIX levels in all patients up to 9 years, with 90% reduction in bleeding episodes. Gene therapy in hemophilia A has been slower due to the larger size of the FVIII gene. In 2017 a truncated FVIII cDNA was used to incorporate into AAV vector, which leads to successful FVIII expression up to 2 years. This is currently being reviewed by the US Food and Drug Administration. A number of phase 1–3 gene therapy trials are currently ongoing and the results could potentially change the treatment landscape of hemophilia.

Strategies for hemophilia care include on-demand treatment of acute bleeding episodes or, for severe hemophilia patients, prophylactic administration of clotting factor concentrate to maintain trough factor levels >1% augmented with on-demand treatment of breakthrough bleeding episodes. The latter strategy pharmacologically converts the severe hemophilia phenotype to a moderate phenotype with an attendant reduction in frequency of bleeding episodes. A randomized multicenter US national study suggested a markedly reduced incidence of hemophilic arthropathy when prophylaxis was instituted prior to onset of recurrent joint bleeds. This benefit must be balanced with the need for frequent prophylactic infusions (3–4 times/wk for FVIII, 2 times/wk for FIX), venous access considerations, potential requirements for central venous access devices, increased cost of treatment, and the occasional patients who have a mild clinical course. EHL products are useful to mitigate some of these limitations. However, recovery studies maybe needed to determine adequate dosing frequency and treatment individualized for the patient’s activities. Most providers will still use short-acting products for breakthrough bleeds. Table 13.14 provides generally accepted guidelines for treatment of most types of hemophilic bleeding. When a bleeding episode is suspected, hemostatic treatment should be rendered first and diagnostic evaluation(s) should be performed later to prevent adverse consequence of the bleed.

Table 13.14
Treatment of bleeding episodes.
Type of hemorrhage Hemostatic factor level Hemophilia A Hemophilia B Comment/adjuncts
Hemarthrosis 30–50% minimum FVIII 20–40 units/kg q12–24 h as needed; if joint still painful after 24 h, treat for further 2 days FIX 30–40 units/kg q24h as needed; if joint still painful after 24 h, treat for further 2 days Rest, immobilization, cold compress, elevation
Muscle 40–50% minimum, for iliopsoas or compartment syndrome 100% then 50–100%×2–4 days
  • 20–40 units/kg q12–24 h as needed

  • For iliopsoas or compartment syndrome initial dose is 50 units/kg

  • 40–60 units/kg every 24 h as needed

  • For iliopsoas or compartment syndrome initial dose is 60–80 units/kg

Calf/forearm bleeds can be limb-threatening. Significant blood loss can occur with femoral-retroperitoneal bleed
Oral mucosa Initially 50%, then EACA at 50 mg/kg q6h×7 days usually suffices 25 units/kg 50 units/kg Antifibrinolytic therapy is critical. Do not use with PCC or aPCC
Epistaxis Initially 30–40%, use of EACA 50 mg/kg q6h until healing occurs may be helpful 15–20 units/kg 30–40 units/kg Local measures: pressure, packing
Gastrointestinal Initially 100% then 50% until healing occurs FVIII 50 units/kg, then 25 units/kg q12h FIX 100 units/kg, then 50 units/kg q day Lesion is usually found, endoscopy is recommended, antifibrinolytic may be helpful
Hematuria Painless hematuria can be treated with complete bed rest and vigorous hydration for 48 h. For pain or persistent hematuria 100% FVIII 50 units/kg, if not resolved 30–40 units/kg every day until resolved FIX 80–100 units/kg, if not resolved then 30–40 units/kg every day until resolved Evaluate for stones or urinary tract infection. Lesion may not be found. Prednisone 1–2 mg/kg/d×5–7 days may be helpful. Avoid antifibrinolytics
Central nervous system Initially 100% then 50–100% for 14 days 50 units/kg; then 25 units/kg q12h 80–100 units/kg; then 50 units/kg q24h Treat presumptively before evaluating, hospitalize, Lumbar puncture requires prophylactic factor coverage
Retroperitoneal or retropharyngeal Initially 80–100% then 50–100% until complete resolution FVIII 50 units/kg; then 25 units/kg q12 h until resolved FIX 100 units/kg; then 50 units/kg q24 h until resolved Hospitalize
Trauma or surgery Initially 100%; then 50% until would healing is complete 50 units/kg; then 25 units/kg q12 h 100 units/kg; then 50 units/kg q24 h Evaluate for inhibitor prior to elective surgery
Note: Antifibrinolytic= EACA ; syrup, 250 mg/5 mL; tablet, 500 mg, 1000 mg.

Ancillary therapy

DDAVP: in hemophilia A patients, DDAVP increases plasma FVIII levels twofold to fivefold. It is commonly used to treat selected hemorrhagic episodes in mild hemophilia A patients.

When used intravenously the dose is 0.3 μg/kg (maximum dose 25 μg) administered in 25–50 mL normal saline over 15–20 minutes. Its peak effect is observed in 30–60 minutes. Subcutaneous DDAVP is as effective as IV DDAVP, facilitating treatment of very young patients with limited venous access. Concentrated intranasal DDAVP (Stimate, 0.1%), available as a 1.5 mg/mL preparation, can be used age 6 years or older. Care should be exercised to avoid inadvertent dispensing of the dilute intranasal DDAVP (0.01%) commonly used for treatment of diabetes insipidus. The peak effect of intranasal Stimate is observed 60–90 minutes after administration.

Recommended dosage for use of intranasal Stimate:

  • Body weight <50 kg: 150 μg (one-metered dose).

  • Body weight >50 kg: 300 μg (two-metered doses).

Precautions with DDAVP use:

  • 1.

    Mild fluid restriction to two-thirds maintenance fluids and drinking to thirst, only electrolyte-containing fluids; avoidance of free water.

  • 2.

    Monitoring urinary output and daily weights may be useful to track fluid retention.

A test dose of DDAVP should be administered at the time of diagnosis or in advance of an invasive procedure to assess the magnitude of the patient’s response. DDAVP administration may be repeated at 24-h intervals according to the severity and nature of the bleeding. Administration of DDAVP at shorter intervals results in a progressive tachyphylaxis over a period of 4–5 days. Depending on the response after a trial, DDAVP could be recommended for most minor procedures. For major procedures requiring maintenance of factor activity >80%, factor concentrates may be needed even in mild hemophilia A patients.

Side effects of DDAVP:

Asymptomatic facial flushing.

  • Thrombosis (a rarely reported complication).

  • Hyponatremia, more common in very young patients, in patients receiving repeated doses of DDAVP or large volumes of oral or IV fluids; hyponatremic seizures have been reported in children under 2 years of age. DDAVP is, therefore, contraindicated in children less than 2 years of age.

Antifibrinolytic therapy

Antifibrinolytic drugs inhibit fibrinolysis by preventing activation of the proenzyme plasminogen to plasmin, mostly by activating TAFI. This intervention is useful for preventing clot degradation in areas rich in fibrinolytic activity, including the oral cavity, the nasal cavity, and the female reproductive tract. Approved antifibrinolytic drugs include:

Epsilon aminocaproic acid (EACA; Amicar): available as a pill and liquid, prescribed in a dose of 50–100 mg/kg every 6 hours (maximum, 20 g total dose/d). GI symptoms may occur at higher doses; therefore the preferred starting dose is 50 mg/kg. The drug is available as 500 mg, 1000 mg tabs, or as a flavored syrup (250 mg/mL).

Tranexamic acid (Cyklokapron, Lysteda): 20–25 mg/kg (maximum, 1.5 g) orally or 10 mg/kg (maximum, 1.0 g) intravenously every 8 hours. This is approved for use in women with bleeding disorders with menorrhagia with expanded use in bleeding disorder patients.

Antifibrinolytic therapy should not be utilized in patients with urinary tract bleeding because of the potential of intrarenal clot formation. To treat spontaneous oral hemorrhage or to prevent bleeding from dental procedures in pediatric patients with hemophilia, either drug is begun in conjunction with DDAVP or factor replacement therapy and continued for up to 7 days or until mucosal healing is complete. Antifibrinolytic drugs also have efficacy as an adjunct treatment for epistaxis and for menorrhagia. Antifibrinolytic drugs are safe to use in hemophilia B patients receiving coagulation FIX concentrates but should not be used concurrently with activated prothrombin complex concentrates (aPCCs) because of the thrombotic potential of the combination. Initiation of oral antifibrinolytic drug therapy 4 hours after the last dose of aPCCs appears to be well tolerated.

Management of inhibitors in hemophilia

Approximately 30% of patients with severe hemophilia A can develop neutralizing alloantibodies (inhibitors) directed against FVIII. Inhibitors are a major cause of morbidity and mortality in hemophilia. Risk factors for inhibitors include early age at exposure, presence of the common inversion mutation, large deletions of the FVIII gene, African-American ethnicity, and a sibling with hemophilia and an inhibitor. The SIPPETT study prospectively randomized previously untreated patients to receive a plasma-derived product or a recombinant product. There was a twofold higher incidence of inhibitors in the recombinant product group. The INSIGHT group demonstrated an incidence of 6.7% by 50 exposure days and 13.3% by 100 exposure days.

Inhibitors are quantified using the Bethesda assay. Low-responder inhibitors have titers <5 Bethesda units (BU) and do not exhibit anamnesis upon repeated exposure to FVIII. A 1 BU neutralizes 50% of FVIII/FIX activity. Approximately half of these patients with inhibitors will be low responders and, of these, approximately half will have transient inhibitors. Hemophilia A patients with low-responder inhibitors can generally be treated with FVIII concentrate, albeit at an increased dosing intensity because of reduced in vivo recovery and a shortened half-life of the FVIII. High-responder inhibitors have titers ≥5 BU, and although the titer may decay in the absence of FVIII exposure, these patients will display an anamnestic rise in titer upon rechallenge with FVIII. The clinical approach is different for high and low responders ( Table 13.15 ).

Table 13.15
Recommendations for replacement therapy for treatment of bleeding in patients with factor VIII inhibitors.
Type of patient Type of bleed Recommended treatment
Low responder a ( < 5 BU) Minor or major bleed Factor VIII infusions using adequate amounts of factor VIII to achieve a circulating hemostatic level
High responder b with low Minor/major bleed PCC, aPCC or rVIIa infusions
Inhibitor level ( < 5 BU) Life-threatening bleed Factor VIII infusions until anamnestic response occurs, then aPCC or rVIIa infusions
High responder with high Minor bleed PCC, aPCC or rVIIa infusions
inhibitor level ( > 5 BU) Major bleed PCC, aPCC or rVIIa infusions
Notes: aPCC, for example, Autoplex (Hyland) and FEIBA. Nonactivated PCC, for example, Konyne (Cutter) and Proplex (Hyland).

a Rise of inhibitor titer is slow to factor VIII challenge.

b Rise of inhibitor titer is rapid to factor VIII challenge.

Low responders

For serious limb- or life-threatening bleeding, a bolus infusion of 100 units/kg FVIII is administered, repeat doses of 100 units/kg are administered at 12-h intervals or, alternatively, the level is maintained with a continuous infusion rate based on the inhibitor titer, recovery and estimated half-life of the factor. An FVIII assay should be obtained 15 minutes after the bolus infusion and trough or steady-state FVIII levels should be followed at least daily thereafter. During prolonged treatment in vivo recovery and half-life may transiently improve as inhibitor antibody is adsorbed by the FVIII.

High responders

Treatment of an acute bleed is achieved by the use of bypassing agents [aPCCs and recombinant VIIa (rVIIa)] and inhibitor eradication is achieved by initiating immune tolerance. In high-responder inhibitor patients with limb- or life-threatening bleeding, if the inhibitor titer is <20 BU, high-dose continuous infusion of FVIII may saturate the antibody permitting a therapeutic FVIII level. The dose can be based on recovery and half-life studies. If an FVIII level is not attainable or the antibody level is greater than 20 BU, then bypassing agents to initiate hemostasis independent of FVIII should be employed.

Agents used to manage bleeding:

  • 1.

    aPCC [FVIII inhibitor bypassing activity (FEIBA)]

    These products have increased amounts of activated FVII (FVIIa), FX (FXa), and thrombin and are effective in patients even with high-titer inhibitors (>50 BU). The initial dose of 75–100 units/kg can be repeated in 8–12 hours not to exceed 200 units/kg/d. Generally, for a joint bleed, four to five doses at 12-h intervals are employed after which the risk of thrombogenicity is increased. Approximately 75% of patients with inhibitors respond to aPCC infusions. For some patients, trace amounts of FVIII in aPCC products may cause anamnesis of the inhibitor titer. If multiple doses are administered, the patient should be monitored for the development of DIC and thromboembolic complications. The simultaneous use of antifibrinolytic therapy (e.g., Amicar) should be avoided. An Oral antifibrinolytic drug therapy 4 hours after the last dose of aPCC, however, appears to be safer.

  • 2.

    Recombinant FVIIa (NovoSeven)

    Recombinant activated FVII (rFVIIa) concentrate can be administered to achieve hemostasis in patients with high-titer inhibitors. The recommended dose is 90-μg/kg rFVIIa repeated every 2 hours for two to three infusions. Higher doses up to 200 μg/kg for two to three doses have been used in pediatric patients in life-threatening situations to achieve better hemostasis. Single high dose of 270 μg/kg has been shown to be effective in the outpatient setting for the treatment of an acute bleed. The subsequent frequency of infusion and duration of therapy must then be individualized, based on the clinical response and severity of bleeding. Early initiation of hemostatic treatment (within 8 hours of a bleed) with rFVIIa produces response rates on the order of 90%. Treatment failures with conventional doses of rFVIIa may respond to higher doses. The incidence of thrombotic complications with this product has been low and anamnesis of the inhibitor does not occur.

  • 3.

    Emicizumab:

    Emicizumab should be considered at the onset of inhibitor development prior to the start of immune tolerance induction (ITI) (see next). Breakthrough bleeding should be treated with rVIIa due to the increased thrombotic risk with aPCC use. Concurrent use of ITI with emicizumab is under investigation to assess its ability to achieve and maintain tolerance.

Modalities used to eradicate inhibitors:

  • 1.

    Plasmapheresis with immunoadsorption

    When bleeding persists despite active treatment, extracorporeal plasmapheresis over staphylococcal A columns may rapidly reduce the inhibitor titer (up to 40%) by adsorbing offending inhibitory IgG antibodies. This approach is cumbersome and may produce significant fever and hypotension due to the release of staphylococcal A protein. However, it can be lifesaving in desperate situations and its efficacy can be enhanced by concomitant replacement therapy with FVIII containing concentrates.

  • 2.

    ITI

    This intervention is instituted for inhibitor eradication in high-responder inhibitors and involves frequent administration of FVIII concentrate to induce immune tolerance to exogenous FVIII. The goals of ITI are an undetectable inhibitor titer, restoring the ability to treat bleeds with FVIII concentrates and restoration of normal in vivo FVIII recovery and half-life. A variety of regimens have been used for ITI ( Table 13.16 ), including daily high-dose FVIII (100 units/kg twice daily or 200 units/kg daily regimen) with or without immunomodulatory therapy, daily intermediate-dose FVIII (50–100 units/kg/d), and alternate-day low-dose FVIII (25 IU/kg).

    Table 13.16
    Selected immune tolerance induction regimens for hemophilia associated inhibitors.
    Protocol FVIII dose (IU/kg) Other agents % Success rate (no. of patients in trial) Inhibitor elimination time (range in months) Predictors of success ( P value)
    High dose
    Bonn ( ) (Brackman et al.) 100–150 twice daily aPCC as required 100 (60) 1–2
    Malm ( ) (Berntorp/Nilsson et al.) Maintain FVIII>0.40 units/mL Cyclophosphamide; 12- to 15-mg/kg IV daily for 2 days followed by 2–3 mg/kg orally given daily for 8–10 days IV Ig; dose is 0.4 g/kg daily for 5 days immunoadsorption 63 (16) 1–2
    Intermediate-dose
    Kasper/Ewing ( ) 50–100 daily Oral prednisone PRN 79 (12) 1–10
    Low-dose
    Dutch ( ) (Mauser-Bunschoten) 25 alternate days 87 (24) 2–28
    Registry
    International, IITR ( ) >200 daily: 32% Steroids 51% 10.5 (time to success) Age at ITI start (.008)
    100–199 daily: 20% Pre-ITI inhibitor titer (.04)
    50–100 daily: 23% Historical peak titer (.04)
    <50 daily: 25% VII dose (higher, .03)
    North American NAITR ( ) >200 daily: 14% Immunomodulators 63% 16.3 (time to success) Pre-ITI inhibitor titer (.005)
    100–199 daily: 33% Historical peak titer (.04)
    50–100 daily: 28% Peak titer on ITI (.0001)
    <50 daily: 25% FVII dose (lower, .01)
    German GITR ( ) 200–300 daily 76% 15.5 (time to success) Historical peak titer (.0012)

    The international ITI study demonstrated that a higher dose (200 IU/kg/d) prevented more early bleeding events and achieved faster tolerance than the lower dose, but immune tolerance was eventually achieved in 70% of patients. Until tolerance is successfully attained episodic bleeds require treatment with bypassing agents. A low historical peak inhibitor titer, a low inhibitor titer at initiation of ITI (<10 BU), and a low maximum inhibitor titer during ITI all favor success. Success rates may also be higher in young patients and in patients treated on higher dose regimens. Data from the Italian ITI study indicate that the relationship between FVIII mutations and rate of inhibitor development is likely to correlate with ITI outcome. Large FVIII gene deletions known to be associated with the highest risk of inhibitor development also showed the highest ITI failure rates. Cost and venous access are added obstacles to successfully completing immune tolerance. In the subset of children who fail ITI, anti-CD20-antibody (rituximab) has been used with and without ITI with varying success rates. Clinically significant responses were observed with concurrent ITI in 47% of patients with only 14% patients achieving durable responses.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here