You are on page 1of 22

Congenital Coagulation Disorders

7
Danielle Sterrenberg and Sucha Nand

The congenital coagulation disorders encompass


a wide variety of inherited diseases that can affect Inherited Disorders of Secondary
all aspects of coagulation including the disrup- Hemostasis
tion of both primary and secondary hemostasis.
Most of these disorders are quite rare but some, Von Willebrand Disease
such as von Willebrand disease, can be seen
rather frequently. Given the disruption of the Von Willebrand disease (VWD) is the most com-
coagulation pathways, they all have in common mon inherited disorder of coagulation affecting
an enhanced predilection for hemorrhagic epi- nearly 1 % of the general population [1]. Some
sodes both spontaneous and as a result of epidemiologic studies suggest that 1:100 people
hemostatic challenges like trauma or surgical have some form of VWD but only about 1:1000
procedures. As such, it is important to identify are symptomatic [2, 3]. There are several forms
affected patients in order to prevent excessive of VWD, the most common of which are inher-
bleeding in the perioperative period. Figure 7.1 is ited in an autosomal dominant fashion and affect
a summary of coagulation defects and their male and females nearly equally (Table 7.1).
impact on either primary hemostasis, or forma-
tion of the platelet plug, and secondary hemo- Von Willebrand Factor
stasis, i.e., generation of thrombin and clot The von Willebrand factor (VWF) gene is located
stabilization. Inherited collagen vascular disor- on the short arm of chromosome 12 at 12p13.3
ders are not discussed in this review. We review and it encodes for the pro-coagulant adhesive
the inherited disorders of secondary hemostasis protein, VWF. VWF is produced and stored as an
first as these disorders are encountered much ultralarge multimeric protein in endothelial cells
more frequently in clinical practice and then and is also produced in megakaryocytes and
move on to discuss disorders of primary stored in alpha granules of platelets. Endothelial
hemostasis. cells release VWF constitutively into plasma
whereas platelets release VWF into circulation
only after their activation [4]. In the event of
sheer stress, this ultralarge VWF binds collagen
in exposed subendothelium and platelets forming
D. Sterrenberg, M.D. • S. Nand, M.D. (*) a bridge. This exposes the A1 domain, a binding
Loyola University Medical Center,
Maywood, IL, USA site for the cleavage protein, ADAMTS13.
e-mail: dsterrenberg@lumc.edu; snand@lumc.edu ADAMTS13 cleaves the ultralarge VWF into

© Springer International Publishing Switzerland 2016 93


C.M. Loftus (ed.), Anticoagulation and Hemostasis in Neurosurgery,
DOI 10.1007/978-3-319-27327-3_7
94 D. Sterrenberg and S. Nand

Primary Secondary
Hemostasis Hemostasis

Hemophilia A and B
Factor XI deficiency
Platelet Function Disorders von Willebrand disease
Rare Inherited Coagulation
Type I (fibrinogen Defects
deficiency)

Glanzmann Thrombasthenia
Bernard-Souiller Syndrome Collagen Vascular Disorders
Ehlers Danlos
Storage Pool Disorders
Wiskott Aldridge Syndrome Osler Weber Rendu

Fig. 7.1 Causes of congenital coagulation disorders and their effect on primary hemostasis, secondary hemostasis, or both

Table 7.1 Laboratory diagnosis of VWD [71]


*Ratio of
*VWF:RCo *VWF:Ag VWF:RCo **VWF
Diagnosis (IU/dL) (IU/dL) *Factor VIII to VWF:Ag **RIPA multimers
Type 1 ↓ <30 ↓ <30 ↓ or normal >0.6 Normal Normal
Type 2A ↓ <30 ↓ <30–200 ↓ or normal <0.6 ↓ ↓HMW
multimers
Type 2B ↓ <30 ↓ <30–200 ↓ or normal Usually ↑ ↓HMW
<0.6 multimers
Type 2M ↓ <30 ↓ <30–200 ↓ or normal <0.6 ↓ Normal
Type 2N ↓ or normal; ↓ or normal; ↓↓ >0.6 Normal Normal
30–200 30–200
Type 3 ↓↓ <3 ↓↓<3 ↓↓↓ (<10 IU/dL) NA Absent NA
Normal 50–200 50–200 100 IU/dL >0.6 Normal Normal

smaller multimers of various molecular weights, IIIa that allow for it to function at multiple steps
preventing the formation of platelet thrombi and in the hemostatic process [6]. VWF has 2 main
enhancing the function of VWF [5]. roles in primary hemostasis; it mediates platelet
Von Willebrand factor (VWF) is essential to adhesion to the sub endothelial surface through
both primary and secondary hemostasis. It is a the GP1b/IX/V receptor and, in coordination
bridging molecule with binding sites for factor with fibrinogen, allows for platelet aggregation
VIII, platelet glycoprotein Ib, exposed collagen via the GPIIb/IIIa receptor. Its role in secondary
on the vessel wall, and platelet glycoprotein IIb/ hemostasis is as a plasma stabilizer for the fragile
7 Congenital Coagulation Disorders 95

factor VIII, carrying factor VIII and thereby Type I VWD


preventing its proteolytic degradation in plasma Type I VWD is the most common subtype rep-
by activated protein C and delivering it to sites of resenting around 75–80 % of all cases. It is a
vascular injury [7]. quantitative defect in VWF leading to reduced,
but not absent, levels in plasma. It is inherited
Diagnosis and Classification in an autosomal dominant (AD) fashion, but has
There are 3 main types of VWD characterized by varying degrees of expression and penetrance
a quantitative defect in the VWF, either partial making characterization of the responsible muta-
loss of VWF as in Type I or full loss of the protein tions and mechanisms of disease as well as diag-
as is seen in Type 3, or by qualitative defects in nosis difficult. Furthermore, varying VWF levels
the function of VWF as is seen in Type 2 VWD. throughout the aging process and throughout
Initial laboratory evaluation for patients sus- blood groups have made this even more chal-
pected of having VWD includes a complete lenging. For instance, VWF levels are known to
blood count (CBC), which is usually normal but increase with age and during times of stress or
in certain variants of VWD can reveal a con- inflammation or pregnancy and to be higher in
current thrombocytopenia. Coagulation studies those individuals with AB blood types than in
show an abnormal bleeding time, a normal pro- those with type O blood [8, 9]. However, recent
thrombin time (PT) and, depending on the variant studies have shown that the decreased VWF level
of VWD, a normal or prolonged activated partial seen in Type I VWD is likely due, in some
thromboplastin time (aPTT). Laboratory studies instances, to enhanced clearance of VWF out of
that are specific to VWD include the von circulation or to intracellular retention of VWF
Willebrand factor antigen (VWF:Ag) which [10]. Because the VWF function remains intact
measures circulating von Willebrand factor, von in Type I VWD, the use of desmopressin
Willebrand Ristocetin cofactor assay (VWF:RCo) (DDAVP) may be useful.
which measures the functional ability of VWF by
mimicking the physiologic interaction between Type 2 VWD
VWF and Gp1b on the platelet surface, and the This subtype comprises a group of abnormalities
factor VIII concentration (FVIII:C). The ratio of in VWF that is further subdivided into four vari-
VWF:Ag to VWF:RCo can help to identify the ants (2A, 2B, 2M, and 2N) all of which are char-
subtype of VWD. For instance, in patients with acterized by qualitative deficits in VWF and
quantitative defects in VWF, the ratio will be collectively account for ~10–20 % of VWD.
near 1 with the dysfunction of VWF related to the Essentially, the production and release of VWF is
degree of deficiency of the protein. However, in normal, but the protein itself is dysfunctional due
the case of qualitative defects, the ratio will be to a variety of inherited mutations.
<0.6 as the circulating antigen levels are near Type 2A: Most frequent form of Type 2 VWD,
normal but their function is impaired. Once the comprising 20–25 % of VWD. It is caused by
patient is confirmed to have VWD with tests genetic mutations that impair the multimerization
listed above, it is important to classify which type of VWF or enhance its sensitivity to ADAMTS13
of VWD they have as the treatment, bleeding cleavage causing loss of high molecular weight
risk, and prognosis may vary. The two tests (HMW) and intermediate molecular weight
utilized most often in classifying VWD are (IMW) multimers and decreased activity of the
ristocetin-induced platelet aggregation (RIPA), binding domains for GPIB on the surface of
used to identify Type 2B in particular, and the platelets. The key laboratory findings are both a
VWF multimer analysis, used to distinguish Type reduced VWF:Ag and VWF:RCo level but with
2A from Type 2M. a disproportionate decrease in the functional
96 D. Sterrenberg and S. Nand

component such that their ratio is <0.6, in similar to that seen in severe hemophilia A. Given
addition to a VWF multimer profile exhibiting a these patients have virtually no VWF, they do not
loss of IMW and HMW multimers. respond to DDAVP.
Type 2B: Gain of function mutations in the
Gp1Ba binding domain, inherited in an Auto- Clinical Presentation
somal Dominant fashion, enable the VWF to Patients with von Willebrand disease experience
bind platelets more avidly leading to enhanced mucocutaneous bleeding, epistaxis, menorrhagia
clearance of HMW multimers and platelets. The as well as prolonged and at times severe bleeding
hemostatic findings are identical to that seen in after surgery or trauma. One of the most common
Type 2A, however, ristocetin induced platelet sources of hemorrhage in VWD patients is men-
aggregation (RIPA) would show enhanced aggre- orrhagia, with 20 % of women with menorrhagia
gation to low dose ristocetin, distinguishing Type having VWD [12]. Life threatening central ner-
2A from 2B. In addition, Type 2B can be asso- vous system (CNS) and gastrointestinal (GI)
ciated with thrombocytopenia. DDAVP is con- bleeding is rare but can occur in patients with
traindicated in this subtype due to the risk of Type 3 VWD or less frequently in type 2 VWD.
exacerbating thrombocytopenia.
Type 2M: Caused by loss of function muta- Treatment
tions in the platelet or collagen binding domains There are 3 main therapeutic strategies for man-
that impair VWF dependent platelet adhesion. As aging VWD. The first strategy involves enhanc-
with all Type 2 mutations, the ratio of VWF:Ag ing the release of VWF from endogenous
to VWF:RCo is <0.6 but VWF multimers remain compartments into plasma through the use of
normally distributed. DDAVP. The second strategy is to replace endog-
Type 2N (Normandy): This variant involves enous VWF with exogenous, virally inactivated
mutations inherited in an autosomal recessive plasma concentrates of VWF and factor VIII, and
(AR) fashion that interfere with the functional the third strategy is to enhance hemostatic con-
ability of VWF to bind and carry FVIII leading to trol utilizing agents that are extraneous to the
enhanced proteolytic degradation of FVIII and a VWF/FVIII complex.
clinical syndrome that is similar to mild hemo- DDAVP is the treatment of choice for VWD.
philia A. Distinguishing hemophilia A from It is a synthetic derivative of vasopressin that
VWD Type 2N can be challenging as the facilitates the release of VWF from endothelial
VWF:Ag and VWF:RCo can be reduced or nor- compartments via V2 receptors thereby increas-
mal and factor VIII will be reduced. VWF multi- ing VWF levels in circulation [13]. It also raises
mers are usually normal as well. Distinction may FVIII levels, although this mechanism is not as
require assays of factor VIII–VWF binding [11]. well understood. All patients with VWD, except
those with Type 2B, should undergo a DDAVP
Type 3 infusion test near the time of diagnosis to see if
This type is the most rare form of VWD, affect- they respond to DDAVP as this treatment can be
ing only 1 in 1 million people and is the most given to correct acute bleeding episodes or given
severe [10]. It is caused by mutations, inherited prophylactically prior to necessary surgeries.
as an autosomal recessive trait, that result in near Even though patients with Type 3 VWD usually
total absence of VWF. This results in extremely do not respond to DDAVP, an infusion study
low VWF:Ag and VWF:RCo as well as very low should still be performed, as there are small sub-
factor VIII levels that are in the range of those sets of patients who will respond to therapy [14].
with severe hemophilia A. The clinical presenta- Combination VWF and factor VIII concen-
tion of patients with Type 3 is different from all trates are indicated in Type 2B VWD and in
other forms of VWD as patients usually experi- patients who are actively bleeding, for the pre-
ence sever bleeding episodes with spontaneous vention of excessive or delayed bleeding from
hemorrhage into synovial space and musculature trauma or surgical procedures with high risk of
7 Congenital Coagulation Disorders 97

hemorrhage, and those patients who fail to The ability to use DDAVP perioperatively to
respond to DDAVP. Humate-P is a VWF/factor prevent bleeding depends on a variety of factors.
VIII concentrate that is most commonly used in Patient responsiveness is a main factor, but the
this setting as it has been the most studied. Its extent of hemostatic challenge provided during
ratio of VWF/RCo to factor VIII is very similar the operation and the time needed to heal must be
to that seen in normal plasma and has been shown considered. In general, major surgery requires
to have efficacy in restoring normal hemostasis. hemostasis for 7–14 days while minor surgeries
A new agent, BAX111, is a recombinant human only require hemostasis for 1–5 days. Common
VWF that has been shown to be efficacious major and minor surgical procedures are listed in
in controlling bleeding in patients with VWD in Table 7.1. If more than 3 days of hemostasis is
phase I and Phase III trials [15]. It is currently necessary, then DDAVP is not the only agent
awaiting FDA approval. needed to provide coverage given its propensity
Other agents can be used to halt or prevent for tachyphylaxis after 3–4 doses. As such, factor
bleeding events in patients with VWD. For replacement is necessary in these patients.
instance, oral contraceptives or intrauterine Patients who are at very high risk of intraopera-
devices can be given to prevent menstrual blood tive bleeding due to the nature of the surgery
loss while fibrin clot stabilizers such as epsilon should be covered by factor replacement as well.
aminocaproic acid (EACA or Amicar) or trane- If the procedure is of mild hemorrhagic risk, such
xamic acid (Cyklokapron) are often used to arrest as a dental procedure, and the patient has been
minor mucosal bleeding or menorrhagia both in shown to have a response to DDAVP infusion,
combination with DDAVP or factor replacement then the patient can be treated with DDAVP prior
or as sole agents [16]. Both are available in oral, to surgery and then repeated every 8–24 h as
intravenous, and topical formulations and exert needed for ~3 doses. However, factor VIII level
their effect by binding plasminogen and prevent- and VWF:RCo should be checked after every
ing its activation to plasmin, thereby preventing administration because of the occurrence of
fibrinolysis [17]. They are contraindicated in tachyphylaxis. Usually for mild procedures,
patients with hematuria given the risk of clot for- DDAVP is administered in conjunction with anti-
mation in the renal collecting system leading to fibrinolytics such as Amicar or tranexamic acid.
obstructive uropathy. The antifibrinolytics are usually continued for
5–7 days after mild procedures.
Perioperative Management of VWD Humate P and Alphanate are two commer-
Preoperative evaluation of a patient with VWD cially available VWF factor replacements that
should include a detailed history of previous bleed- are approved for the prophylactic prevention of
ing episodes and surgical outcomes to fully assess bleeding in the surgical patient. However, Humate
the bleeding risk of the planned surgery or proce- P is the most widely used and most studied in both
dure. If the patient has not previously had a desmo- the setting of reversing acute bleeding and in the
pressin infusion test performed, it should be done prophylactic management of surgical patients.
prior to surgery. Response patterns to DDAVP Several studies have shown that it achieves excel-
remain uniform throughout life, so if a patient has lent to good hemostasis in 91–100 % of surgical
had a prior documented response to DDAVP, then procedures [18, 19]. Humate P is usually given
this does not need to be repeated prior to surgery. once daily with the exact dose based on the risk
Laboratory evaluations should include a CBC, PT, of intraoperative bleeding. For instance, during
PTT, and factor VIII level as well as a VWF:Ag major surgical procedures Humate P is given once
level and VWF:RCo assay. Clinical data suggest daily and dosed to raise the factor VIII and
that the FVIII level is the best predictor of soft VWF:RCo levels to 80–100 IU/dL the day of sur-
tissue, surgical, and delayed bleeding in VWD gery and then maintained >50 IU/dL for 7–14
patients while the VWF:RCo is the component days after surgery. For minor procedures, FVIII
responsible for mucosal bleeding [16]. and VWF:RCo target levels are >50 IU/dL on the
98 D. Sterrenberg and S. Nand

day of surgery and then continued for a target with hemophilia B have causative missense
level of >30 IU/dl for 1–5 days after surgery. For mutations and inversions are in general not
dental extractions or minimally invasive proce- seen [24].
dures, patients should receive a one-time dose of Hemophilia A is more prevalent, affecting
Humate P to raise their FVIII or VWF:RCo to around 1:5000 males with approximately 13,000
>50 IU/dL [20]. new cases identified per year. Hemophilia B
Patients with VWD, despite their coagulopa- affects approximately 1:30,000 males with around
thy, can still develop thromboembolic disease. In 3600 new cases of hemophilia B in the USA per
fact, several studies have shown cases of deep year [25, 26].
venous thrombosis (DVT) or pulmonary embo-
lism (PE) after factor VIII and VWF replacement Hemophilia A
in VWD patients undergoing surgery [21]. Hemophilia A is an X-linked recessive (XLR)
A review of the patients who developed these genetic deficiency of the functional plasma clot-
venous thromboembolic events (VTE) shows ting factor, factor VIII (FVIII) resulting from
that several of them had very high FVIII levels mutations in the FVIII gene located on the long
and additional risks for VTE such as advanced arm of the X chromosome. This results in low to
age, estrogen use, and recent surgical procedures. absent factor VIII levels and a disruption in the
Thus, care should be taken to not overcorrect the intrinsic pathway of the coagulation cascade
factor VIII level by checking both a FVIII in causing a lack of thrombin or fibrin generation
addition to VWF:RCo during factor replacement and thus inability to stabilize the platelet plug
and to consider anticoagulant prophylaxis in sur- formed during primary hemostasis resulting in
gical patients especially in those with other risk excessive, often delayed, hemorrhage both in
factors for VTE development [22]. response to trauma or arising spontaneously.
Over 70 % of cases are inherited in an XLR
pattern, but around 30 % of cases arise as sponta-
Hemophilia neous mutations in the FVIII gene. Given its X
linked pattern of inheritance it is a disease seen
Hemophilia is a group of disorders resulting from almost exclusively in males. However, mild
genetic mutations in either factor VIII (hemo- hemophilia A and, very rarely, even a severe
philia A) or factor IX (hemophilia B). Both genes hemophilia phenotype are known to occur in het-
are located on the long arm of the X chromosome erozygous females due to various degrees of
at Xq28 and Xq27 respectively and thus are chromosomal X inactivation [27]. Given this and
inherited in an X-linked fashion [23]. Most the fact that female carriers, while asymptomatic,
affected men display symptoms while carrier can also have reduced factor VIII levels, moni-
females are usually asymptomatic. With the use toring of factor VIII levels should be considered
of molecular genetics, a wide spectrum of muta- in female patients with a family history of hemo-
tions in the factor VIII and factor IX genes have philia or evidence of exaggerated hemorrhage in
been identified and known to contribute to the response to trauma or spontaneous hemorrhage.
respective clinical syndrome. In fact, according The clinical severity, or risk of bleeding, in
to an online database, www.factorviii-db.org, patients with hemophilia A can be predicted by
there are currently (as of February 2015) 2,015 the coagulation factor activity level present in
unique mutations identified as causing hemo- plasma. Table 7.2 shows the clinical classifica-
philia A. By far the most common mutations tion of patients with hemophilia A. In general,
identified are intron 22 inversions that affects factor levels (<1 IU/dL) are considered severe
more than 45 % of severe hemophilia A patients. with patients presenting, oftentimes in infancy,
Hemophilia B has a similar online database at with episodes of spontaneous hemorrhage most
www.factorix.org that has identified 1095 unique commonly into their joints or muscles. Less
variants of F9 mutations. Almost 70 % of patients severe phenotypes are associated with some
7 Congenital Coagulation Disorders 99

Table 7.2 Suggested duration of VWF replacement for different surgical procedures [72]
Other procedures, if uncomplicated,
Major surgery (7–14 days) Minor surgery (1–5 days) single VWF treatment
Cardiothoracic Biopsy: breast, cervical Cardiac catheterization
Cesarean Section Complicated dental extractions Cataract surgery
Craniotomy Gingival surgery Endoscopy (without biopsy)
Hysterectomy Central line placement Liver biopsy
Open cholecystectomy Laparoscopic procedures Lacerations
Prostatectomy Simple dental extractions

residual factor VIII levels and are characterized FVIII Protein and the Pathophysiology
by excessive bleeding at sites of trauma. In gen- of Hemophilia A
eral, factor VIII levels >30 % to that of normal are FVIII circulates in plasma as an inactive precursor
enough to prevent spontaneous hemorrhage and to that is in a complex with von Willebrand factor.
maintain a normal hemostatic system. An epide- It has no intrinsic enzymatic activity but rather
miologic survey conducted in the 1990s identified functions as a cofactor for FIX in the activation of
about 43 % of hemophiliacs as severe [28]. FX to its active form, FXa [31]. It is a highly
Both hemophilia A and B have identical inher- unstable protein and requires the covalent binding
itance patterns and clinical presentation but have of von Willebrand factor to prevent its proteolytic
different factor deficiencies. However, there are degradation in plasma. This instability of FVIII in
some studies suggesting a higher bleeding risk in plasma is why, in some forms of von Willebrand
patients with hemophilia A for the same level of disease, FVIII levels can be low [32]. Factor VIII
factor activity [29]. Also, with hemophilia A the and factor IX deficiency results in the same coagu-
factor levels generally remain constant through- lation derangement: namely, the inability to gener-
out a patients’ life but with hemophilia B there ate thrombin and fibrin to secure the primary
can be some increase in FIX levels after puberty platelet plug formed during primary hemostasis.
such that a patient with a moderate form of hemo-
philia may become more mild due to a small but Hemophilia B
clinically significant rise in FIX levels in response The clinical presentation of hemophilia B is
to androgen production during adolescence. Prior nearly identical to that of hemophilia A. In addi-
to modern factor replacement, the median overall tion, Both the FVIII and the FIX genes are located
survival for patients with severe hemophilia, A or on the X chromosome in close proximity to one
B, was 11.3 years. However, more recent esti- another. Also as seen in hemophilia A, 70 % of
mates in patients treated with factor replacement mutations are inherited in an XLR pattern but
and after appropriate viral eradication of blood 30 % of mutations arise de novo.
products was possible in them show an average FIX circulates in plasma as a serine protease
survival of 63 years for males with severe zymogen that requires FVIII as a cofactor for its
phenotype and 73 years for males with mild to functional activity. Together with FVIII, a com-
moderate hemophilia [30]. These numbers plex is formed that cleaves FX into its active
approach that seen with the normal population. form, FXa. However, unlike FVIII, FIX is depen-
As a consequence, more and more patients are dent on posttranslational vitamin K-dependent
developing additional medical comorbidities and processing by gamma carboxylase for full func-
are requiring surgical procedures for a number of tional activity. Clinical severity and management
common aging maladies such as heart disease of hemophilia B is also dependent on the amount
and cancer. of factor IX functional activity retained [33].
100 D. Sterrenberg and S. Nand

Clinical Manifestations of Hemophilia Diagnosis


A and B Approximately 50–70 % of newborns with
Bleeding is the hallmark of hemophilia. However, hemophilia have a positive family history and
clinical presentation and prognosis in hemophilia in these patients the diagnosis is usually made
varies significantly based on the residual factor around the time of birth by checking factor VIII
levels. Patients with severe disease are often or factor IX levels on cord blood. In the remain-
afflicted by recurrent non-traumatic bleeds. Birth ing patients the disease is diagnosed when symp-
represents the first hemostatic challenge in the toms begin to manifest, which is usually very
life of a hemophiliac and is when some patients early in infancy for severe patients and later in
present with symptoms. Common sites of bleed- childhood or in adulthood for mild cases. Workup
ing during the first month of life for patients with consists of a detailed family history, a detailed
hemophilia include post-circumcision bleeds bleeding history and surgical history, as well as
(~48 %), followed by intracranial hemorrhage appropriate laboratory testing. A CBC is neces-
(ICH) seen in 19 % of cases [34]. Multiple stud- sary in the evaluation of a potential patient and is
ies have shown that the rate of ICH in hemo- usually normal but can have an associated micro-
philia, regardless of disease severity, is around cytic anemia depending on the amount of prior
1–4 % with a 20 % mortality rate [34, 35]. After blood loss. The PT is normal but aPTT is pro-
the neonatal period, patients tend to develop longed. If performed, the bleeding time is usually
hemorrhage at alternative sites. normal. A mixing study will correct the aPTT
Hemarthrosis is the most common site of to within normal range, but in patients with an
spontaneous hemorrhage. While any joint can be inhibitor, the correction may be partial. Factor
affected, this is seen most commonly in the knee, VIII or factor IX levels are reduced or absent in
elbow and ankles usually starting after affected hemophilia A or B respectively. Because VWD,
individuals begin to crawl or walk. Hemarthrosis which is the most common congenital coagula-
results in chronic synovitis which can lead a tion disorder, can also present with reduced factor
patient to develop hemophilic arthropathy, which VIII levels, it is important to rule this out via
severely affects joint integrity and in cases of appropriate laboratory assessment prior to a for-
recurrent bleeds, can lead to a deformed and mal diagnosis of hemophilia A.
essentially dysfunctional joint [36].
Intramuscular hematomas are the second most Treatment
common site of hemorrhage in hemophiliacs. Replacement of the deficient factor with either
The hematoma can be large and in severe cases, plasma derived or recombinant factor VIII or fac-
depending on the location, can be associated with tor IX concentrate is the hallmark of hemophilia
compartment syndrome, major blood loss in the therapy. Decisions regarding when to treat, what
setting of iliopsoas hematomas, and local muscle dose to use, and how often to administer depend
or tendon damage [37]. on the severity of disease and whether or not
Other common forms of hemorrhage include there is active hemorrhage occurring or the
mucocutaneous, gastrointestinal and genitouri- patient is undergoing a surgical procedure.
nary. Intracranial hemorrhage is one of the most The treatment of hemophilia is based on the
worrisome sequelae of hemophilia and remains a clinical severity of the syndrome. Individuals
risk throughout the patients’ life. In a case– with severe deficiency, require regular prophy-
control trial of hemophilia patients, high inhibi- lactic administration of either plasma derived
tor titers, severe hemophilia, and a prior history or recombinant factor VIII or factor IX to pre-
of ICH were all associated with higher rates of vent spontaneous hemorrhage and consequent
ICH [38]. joint damage that occurs with synovial bleeds.
7 Congenital Coagulation Disorders 101

Multiple studies have been performed showing dental extractions, thereby decreasing or
that in severe cases, prophylactic administration eliminating the use of homologous factor replace-
of factor replacement reduces the annual bleeding ment in select patients [44]. Despite marked vari-
rates and subsequent joint damage seen in hemo- ability between patients, DDAVP can be seen to
philia patients versus on-demand replacement increase factor VIII levels three to fivefold that of
strategies [39]. Patients with mild to moderate baseline in patients with mild hemophilia A or
forms can utilize on demand scheduling of factor von Willebrand disease and this increase is often
replacement or alternative agents to reduce the enough to terminate mild bleeding episodes and
bleeding risk. In cases of prophylactic adminis- provide adequate coverage for low risk surgical
tration or minor mucosal bleeding, raising factor procedures. Patients with severe factor VIII defi-
VIII level to 30 % (~0.3 IU/dL) is usually suffi- ciency or hemophilia B do not respond to DDAVP
cient for normal hemostasis. and this agent should not be considered.
The fibrin clot stabilizers Amicar and tran-
Active Hemorrhage examic acid are seldom utilized in hemophilia.
During acute life threatening hemorrhage or However, they can be considered for mild muco-
major surgical procedures a normal factor VIII or sal or uterine bleeding or after dental extractions
factor IX level should be maintained at all times again in an effort to enhance local control of
[40]. The half-life of both plasma derived and minor bleeding without the use of blood prod-
recombinant factor VIII is 8–12 h necessitating ucts. As mentioned earlier, they are contraindi-
twice daily administration in the setting of active cated in patients with hematuria.
hemorrhage or major surgery, whereas the half-
life of factor IX in serum is around 20 h allowing Inhibitors
for daily dosing [41, 42]. The dose of concentrate Now that blood products used for replacement in
to be administered is calculated based on the sub- hemophilia utilize adequate viral eradication
type of hemophilia, the baseline factor levels, strategies, the transmission of HIV and hepatitis
and the target factor level. For mild/moderate that once plagued the hemophilia world and led
bleeding, the target range for factor levels is to significant morbidity and mortality is essen-
around 0.3 IU/dL (30 % normal), however for tially a concern of the past. However, the main
acute severe hemorrhage, the goal is 1 IU/dL or concern now with the use of factor replacement is
near 100 % normal range [43]. Factor concen- the development of inhibitors. These inhibitors
trates can be administered in bolus or continuous are allo-antibodies to factor VIII and factor IX
dosing, with some studies recommending a con- that decrease levels of endogenous factors and
tinuous infusion in cases of life threatening hem- also neutralize the administration of exogenous
orrhage in order to prevent the peaks and troughs factors during replacement strategies thus incre-
seen with bolus dosing. asing a patient’s risk of bleeding both spontane-
ously and in response to trauma and making the
Desmopressin (DDAVP) correction of their coagulopathy more difficult.
Desmopressin, or DDAVP, can be used to treat In hemophilia A, about 30 % of patients with
patients with mild or moderate factor VIII defi- severe disease will develop inhibitors that can
ciency in which a prior rise of their factor VIII bind to factor VIII [45]. They are typically IgG4
levels in response to therapy has been demon- and can render factor replacement completely
strated. DDAVP is a derivative of antidiuretic ineffective. For reasons not completely known,
hormone and has been shown to increase plasma they occur more frequently in patients with
levels of factor VIII and von Willebrand factor severe disease and are more common in African
through increasing the release of this complex Americans, young patients undergoing replace-
from endothelial cells. It can therefore be used in ment for the first time, patients getting factor
cases of mild mucosal bleeding or periope- replacement in preparation for surgery, and in
ratively surrounding mild procedures, such as those with certain genetic mutations responsible
102 D. Sterrenberg and S. Nand

for hemophilia [46–48]. They occur less efficacious therapy, eradicating inhibitors in
frequently in hemophilia B, usually in about 3 % about 70 % of patients. However, it takes 9–12
of affected patients [45]. Not only do inhibitors months of treatment to obtain this result. There-
increase a patient’s bleeding risk but they also fore, bypassing agents are often required in
make the prophylaxis and treatment of hemo- patients prior to or during ITI or in the event of
philia much more challenging. relapse. Immune tolerance is achieved using fre-
The severity of inhibitors is based on the activ- quent and prolonged high doses of FVIII either
ity of the antibodies, which is measured in anti- with or without concurrent immunosuppressive
body titers. Patients with low-titer inhibitors, i.e., therapy [51].
<5 BU/ml (Bethesda units), often have transient
antibodies that can subside without therapy and Perioperative Treatment of Hemophilia
can be treated with higher doses of factor VIII or The risk of bleeding is high for patients with
factor IX alone. However, high-titer inhibitors hemophilia who require surgery and all appropri-
(>5 BU/ml) often will not respond to increasing ate preoperative measures should be taken. A
doses of factor replacement and require alterna- detailed surgical and family history of all surgical
tive forms of therapy. As in the patient without patients should be recorded prior to their proce-
inhibitors, one can consider DDAVP, Amicar, and dure and if a potential female hemophilia carrier
tranexamic acid to control or decrease the risk of is identified, then she would require a more in
bleeding. However, in patients with severe acute depth hematologic assessment, as her FVIII lev-
bleeding or at high risk of spontaneous hemor- els can be variably reduced, predisposing her
rhage, bypass agents are used. to excessive perioperative hemorrhage and pot-
entially requiring treatment prior to surgery.
Factor VIII Inhibitor Bypassing Agents In anticipation of any elective surgical procedure,
(FEIBA) a multidisciplinary perioperative plan should be
FEIBA is an activated prothrombin complex arranged. Care should to be taken to ensure that
concentrate (aPCC) containing all the vitamin K adequate factor replacement is available to
dependent coagulation factors (FII, FVII, FIX, administer to patients before, during, and after
and FX) and factor VIII in both zymogen and the procedure. A detailed presurgical workup
active forms. The mechanism of action is com- should be performed a few weeks prior to surgery
plex, but is thought to involve thrombin activa- (in the case of elective procedures) and should
tion via prothrombin and factor X [49]. It has a include a repeat CBC, PT, aPTT, and factor lev-
half-life of 6–12 h and thus doses are usually els. Laboratory assessment for the development
repeated every 6–12 h depending of bleeding of inhibitors should also be performed. Adequate
severity and clinical response. preoperative testing can take more than a week
to obtain results and should be planned
Recombinant Factor VIIa (rVIIa) accordingly.
rVIIa, also known as Novoseven, is a recombi- Dosing of clotting factors varies on the type of
nant factor VIIa that facilitates hemostasis by surgery performed. For major surgery, FVIII and
activating FXa directly on platelet surfaces, thus FIX levels should be around 100 % of normal.
bypassing the factor VIII complex entirely. It has This can be accomplished with perioperative and
a short half-life of 2.3 h and thus must be admin- postoperative administration of factor concen-
istered frequently in the setting of active major trate provided the patient has not developed any
hemorrhage [50]. inhibitors. Treatment should begin prior to sur-
gery and be continued for at least 10–14 days
Immune Tolerance Induction postoperatively and in some cases several weeks
Therapy (ITI) after major surgery depending on the ongoing
The long-term treatment of inhibitor develop- bleeding risk. For moderate risk procedures, such
ment in hemophilia is aimed at eliminating the as dental extractions, factor levels can be
causative antibodies. ITI is the most studied and maintained around 50 % normal value and do not
7 Congenital Coagulation Disorders 103

need to be continued weeks after the procedure. Unlike hemophilia A and B, patients with FXI
Factor replacement can be can be delivered in a deficiencies usually do not develop spontaneous
bolus fashion (usually every 12 h for FVIII hemorrhage but rather have excessive bleeding
replacement and daily for FIX replacement) or as due to surgery or trauma. Also unlike hemo-
a continuous infusion. As in the setting of severe philia, the plasma level of FXI does not correlate
acute hemorrhage, some studies recommend con- with bleeding risk of the patient. For example,
tinuous infusion perioperatively surrounding major homozygous patients with FXI levels <20 IU/dL
surgical procedures with high bleeding risk. can either have adequate hemostasis after trauma
In the event the hemophilic patient has devel- or surgery or experience significant bleeding
oped either factor VIII or factor IX inhibitors, the risks. Heterozygotes with a partial deficiency
perioperative management is more complicated. and relatively high levels of FXI, around
However, several treatment options are available, 60–70 IU/dL can also experience significant
and the choice of treatment depends upon the bleeding complications [53, 54]. As such, a
severity of expected bleeding. It should be noted detailed family history and prior bleeding history
that patients with inhibitors have a higher risk of is of utmost importance because the phenotype
bleeding. of the patient and their family is the best way to
Novoseven (recombinant factor VII or rFVIIa) truly assess bleeding risk as one cannot currently
has been utilized in the perioperative manage- predict risk based on factor levels or other labo-
ment of hemophiliacs who have acquired inhibi- ratory tests.
tors. Prothrombin factor concentrates (FEIBA,
Autoplex) are also routinely used in this setting. Diagnosis
However, the monitoring of their efficacy is more As most FXI deficient patients are asymptom-
difficult, as one cannot rely upon the usual atic, the diagnosis of FXI deficiency is usually
increase in factor levels. As such, the monitoring not made until late childhood or adulthood, gen-
of their efficacy occurs largely by their ability to erally after excessive bleeding is noted after
lower the aPTT level and clinically by the amount surgery or during a preoperative workup of a
of hemostasis achieved in the patient. prolonged aPTT. Similar to hemophilia A and B,
Other agents such Amicar or tranexamic acid initial workup of suspected individuals should
can be given after minor procedures such as den- include a detailed history and laboratory exami-
tal extractions to decrease the risk of bleeding but nation. CBC will usually be normal as will a pro-
their use and benefit in major neurologic proce- thrombin time but the aPTT will likely be
dures is minimal, if any. However, they should prolonged. A workup for more common causes
not be used in conjunction with bypassing agents of excessive hemorrhage such as von Willebrand
as this could increase the patients’ risk of throm- disease or hemophilia should be undertaken and
boembolic phenomenon. ruled out. In certain patient populations such as
Ashkenazi Jews, checking for factor XI defi-
Factor XI Deficiency ciency should be considered prior to a workup
Factor XI (FXI) deficiency, sometimes referred for hemophilia or VWD given its higher fre-
to as hemophilia C, is a rare autosomal recessive quency in this patient population. The diagnosis
inherited coagulation disorder caused by a muta- is confirmed after demonstration of low plasma
tion in the F11 gene on the short arm of factor XI levels. Of note, infants normally have
Chromosome 4. It affects about 1 in 100,000 low factor XI levels that then rise to normal lev-
people. However, it is much more prevalent in els after 6 months of age with no additional age
Ashkenazi and Iraqi Jewish populations, with related variations thereafter and patients with
one in eight Ashkenazi Jews being heterozygous synthetic liver dysfunction will have lower fac-
for the F11 gene mutation and one in 190 having tor XI levels as the liver is the primary site of
a homozygous deficiency [52]. FXI production [55].
104 D. Sterrenberg and S. Nand

Treatment either FFP or FXI depending on availability,


There is some anecdotal evidence that DDAVP assessing each patient for IgA levels, inhibitor
can be helpful at inducing hemostasis in mild formation, and comorbidities that may enhance
cases of bleeding with FXI deficiency due to an their risk of volume overload with FFP prior to
increase in FVIII levels and vWF levels follow- surgery in order to optimize factor replacement.
ing its administration and may be considered in Furthermore, given the risk of thrombotic events
mild cases of FXI deficiency [56]. However, its with both FFP and especially with FIXC, factor
use is controversial. In general, the treatment repletion aim for target levels around 60–70 IU/
includes either factor replacement or the use of dL and, if given in a prophylactic setting, should
antifibrinolytic agents. have prophylactic anticoagulation administered
Fresh Frozen Plasma (FFP), which contains simultaneously if safe.
all factors of the soluble coagulation system, was As is the case with most inherited defects
the initial treatment of choice for FXI deficiency of coagulation, antifibrinolytic agents, given in
and remains so in areas, such as the USA, where either an oral or topical or intravenous route, can
FXI concentrates (FXIC) are unavailable. Since be used to stop or prevent minor cases of bleed-
the early 1990s, FXIC has overtaken FFP in cen- ing. However, their use in conjunction with FXI
ters where it is available, like the UK, as it has a replacement should be avoided due to the risk of
longer half-life of around 45 h, infuses only fac- thrombotic events [58].
tor XI and not additional pro-coagulant agents,
and can be given in a much lower volume thereby Factor XI Inhibitors
reducing the risk of volume overload and other The development of FXI inhibitors after plasma
complications seen with FFP. For prophylactic infusion is rare, but can occur as frequently as
treatment of major surgical procedures with high 33 % in specific gene mutations within FXI defi-
bleeding risk or in the treatment of severe acute ciency [59]. In general, these inhibitors can be
bleeding, FXI concentrate, if available is the bypassed with rFVIIa [60].
treatment of choice and is usually given every
other day and continued for 10–14 days after Rare Inherited Coagulation Disorders
major surgery and for 1 week postoperatively The Rare Inherited Coagulation disorders (RICD)
after minor procedures. In the USA, FFP given comprise only 2–5 % of all inherited disorders of
perioperatively for the same duration is the stan- coagulation and include deficiencies of factor I
dard. There are a few risks that are unique to fac- (afibrinogenemia, hypofigrinogemia, dysfibrino-
tor replacement for FXI deficiency. That is, with genemia), factor II (prothrombin), factor V, com-
both FFP and FXI concentrates there is a risk of bined deficiency of factor V and factor VIII,
severe anaphylaxis in patients with IgA defi- factor VII deficiency, factor X deficiency, FXIII
ciency, a risk of inhibitor formation, and a risk of deficiency and combined deficiency of vitamin K
severe volume overload with the high volume of dependent factors. Inheritance patterns, epidemi-
FFP needed to correct the deficiency. With FXIC, ology, and treatment of these various deficiencies
there is a well-documented risk of thrombosis is listed in Table 7.3. These deficiencies are a
that must be considered. Given the risks associ- clinically heterogeneous group of disorders that
ated with factor repletion, studies are underway arise from autosomal recessive inheritance of
to develop laboratory measures that would ade- DNA mutations in genes encoding the corre-
quately quantify bleeding risk in FXI deficiency sponding coagulation factors leading to decreased
in order to identify patients in whom factor levels of the protein or, more rarely, dysfunction
replacement can be avoided. A thrombin genera- of the involved protein. The exceptions to this are
tion test (TGT) has shown promise in some stud- in the combined FV and FVIII deficiency in
ies, but further validation is necessary [57]. At which there is a mutation in a protein regulating
present, the standard approach is to give prophy- intracellular transport of the two factors [61] and
lactic therapy for all major surgical patients with in the deficiency of vitamin K dependent Factors
7 Congenital Coagulation Disorders 105

(VKDFD) whereby a defect in genes encoding Table 7.3 The severity of bleeding manifestations in
enzymes involved in the vitamin K dependent hemophilia A and B is correlated to residual clotting fac-
tor level
posttranslational modification of proteins lead to
a deficiency of FII, VII, IX, and X (as well as pro- Clotting
factor level %
coagulant factors protein C and S) and systemic activity
issues related to deranged vitamin K metabolism Severity (IU/mL) Bleeding episodes
[62]. Bleeding is more common in homozygotes Severe 1 % (<0.01) Spontaneous bleeding,
with RICD but heterozygotes can also have an usually into joints or
increased propensity for bleeding after episodes muscles. Prophylactic
infusions of factor
of hemostatic stress. replacement is required.
The hallmark of RICD is excessive hemor- Moderate 1–5 % Occasional spontaneous
rhage, the exception to this being FXII deficiency (0.01–0.05) bleeding and severe
that is not associated with an increased bleeding bleeding with trauma or
surgery. Prophylactic
risk and thus will not be discussed in this chapter.
infusion of factor
The clinical presentation of the RICD is variable replacement is usually
with patients being diagnosed anywhere from required.
infancy to late adulthood. In general, their bleed- Mild 4–40 % Severe bleeding with
ing risk is less than that seen with hemophilia A (0.05–0.40) major trauma or surgery.
Prophylaxis is not
and B, but some of these syndromes are associ- generally required.
ated with serious bleeding complications and in
the case of FXIII deficiency with relatively high
rates of spontaneous intracranial hemorrhage Fresh Frozen Plasma
(34 % in severe patients) [63]. FFP is beneficial for several disorders as it
In all forms of RICD, patients can develop contains all the factors of the coagulations
menorrhagia, mucocutaneous bleeding and epi- system.
staxis and excessive bleeding following surgical Prothrombin Complex Concentrates.
procedures. In severe forms of the deficiencies, Prothrombin complexes are a combination of FII,
spontaneous hemorrhage into synovial spaces or VII, IX, and X and protein C and S prepared
muscles can occur as well. With some syndromes from fresh frozen human plasma.
there is a clear correlation with factor level and Cryoprecipitated antihemophilic factor (cryopre-
bleeding risk but in others this linear correlation cipitate).
is less clear. Heterozygotes of these disorders can Cryo is a frozen plasma product rich in fibrino-
exhibit excessive bleeding but usually only after gen and also containing factors VIII, VWF,
significant hemostatic challenge. The diagnosis and FXIII and is used most frequently in the
usually is made by characteristic abnormalities in treatment of factor I or fibrinogen deficiency.
laboratory analysis of coagulation in addition to
corresponding factor deficiencies. As with other coagulation disorders, the antifi-
brinolytic agents can be useful in the setting of an
Treatment acute bleeding episode, as well as to prevent peri-
Treatment should be individualized to the patient operative hemorrhage alone in the setting of
and their specific deficiency but common themes minor deficiencies and minor procedures or in
exist. Acute bleeding is managed by replacement combination with factor replacement. However,
of the deficient factor via either recombinant care should be given when using it with full dose
forms or plasma concentrates, if such factors are rVII as this can potentiate thrombotic events.
available, or pooled plasma combination of fac- Treatment strategies for all RICD are listed in
tors listed below. Table 7.3.
106 D. Sterrenberg and S. Nand

Inhibitors well as an exaggerated hemorrhagic response


Inhibitors can develop in RICD but at a lower following surgical procedures. Given the risk
frequency than that seen in hemophilia and is of enhanced perioperative bleeding, a detailed
on the order of <5 %. This is usually managed workup for IDP should be conducted in the
with rVIIa similar to inhibitor development with patient with normal coagulation screening tests
hemophilia A, B, or C. and a history of unexplained or exaggerated
bleeding [66]. Please see Figs. 7.2 and 7.3 for a
list of platelet disorders and where they affect
Inherited Disorders of Primary platelet function. Our discussion will be limited
Hemostasis to the most commonly occurring and most severe
inherited disorder.
Role of Platelets in Clot Formation

Platelets are the main actors in primary hemo- Glanzmann’s Thrombasthenia (GT)
stasis. They are small enucleate cells that are
released from the megakaryocyte and circulate GT is the most severe of the IDP, and according
inactive in plasma with an average life span of 10 to some reports, affects around 33 % of IDP
days. However, after vessel wall damage, suben- patients. GT is a severe bleeding disorder with
dothelial components such as collagen and VWF affected individuals presenting early in life with
are exposed and bind to and activate platelets. excessive bruising and bleeding in response to
Exposed collagen binds the GPVI receptor and trauma but also with spontaneous hemorrhage
VWF binds to platelets via the GPIb/IX/V recep- including intracranial hemorrhage [67]. It is an
tor anchoring them into place and recruiting other autosomal recessive disorder that is caused by
platelets to the site. Once there, the platelets are qualitative or quantitative defects in the GpIIb/
then activated by a variety of platelet agonists IIIa (fibrinogen) receptors on the platelet surface
such as ADP and thromboxane that, through G [68]. This prevents platelets from aggregating
protein receptor activation, cause a change in the and thus from forming a hemostatic plug.
platelet cytoskeleton that leads to alteration in the
shape of platelets allowing them to spread and
release the contents stored in their alpha and delta Bernard Soulier Syndrome (BSS)
intracellular granules. This results in release of
thromboxane A2 (TXA2) and production of a BSS can at times be a severe bleeding disorder.
procoagulant surface whereby secondary hemo- It is an autosomal recessive congenital bleeding
stasis can be conducted, and activation of GPIIb/ disorder caused by quantitative or qualitative
IIIa receptors resulting in platelet aggregation defects in the GPIb-IX-V VWF (VWF) receptor
and the subsequent formation of a platelet impairing normal adhesion of platelets to sites of
plug limiting blood flow to the site of vascular vascular damage . It is characterized by thrombo-
damage [64]. cytopenia and large platelets and a prolonged
bleeding time. Like GT, the bleeding diathesis
can be severe at times.
Inherited Disorders of Platelets (IDP)

Genetic mutations causing alterations in any one Storage Pool Disorders (SBP)
of the steps involved in primary hemostasis can
result in IDP. IDP are rare and very heteroge- The storage pool disorders, listed in Table 7.4
neous group of disorders, the true incidence of and shown in Fig. 7.3, are a heterogeneous group
which is unknown [65]. They mostly result in of diseases characterized by deregulated platelet
mild to moderate mucocutaneous bleeding as aggregation from various genetic mutations
7 Congenital Coagulation Disorders 107

Reduced response to collagen

Glanzmann thrombasthenia: IV Scott syndrome:


no aggregation with VI CD9
decreased
natural agonists prothrombin conversion

αIIbβ3
α6β1

α5β1

Reduced response to
collagen
α2β1
IX
Bernard-Soulier
V Ibβ syndrome:
Altered response to stimuli: ADP
lack of adhesion to
(P2Y12), TXA2 (TPa), 5-HT Ibα
VWF and abnormal
PAF, adrenaline... Platelet-type VWD:
response to thrombin
spontaneous binding
of VWF to GP Iba

Fig. 7.2 Disorders that principally affect surface components of platelets [75]. Reprinted from Nurden and Nurden
et al. Congenital Disorders with Associated Platelet Dysfunction

ultimately causing disordered release of platelet mucocutaneous, gastrointestinal, genitourinary,


granular contents. The mutations involved in these or intracranial hemorrhage. Most of the defects
disorders can extend beyond the megakaryocytic are inherited in an autosomal recessive fashion,
lineage causing a constellation of systemic find- but there are rare X-linked and autosomal
ings that can include severe immunodeficiency, dominant.
albinism and neurologic deficits [69].

Diagnosis
Clinical Presentation
The diagnosis of IPD can be challenging. In gen-
Patients can present in infancy or into early adult- eral patients in whom there is concern for an
hood with a bleeding diathesis caused by IDP. IDP should undergo simultaneously workup for
Usually the bleeding manifestations of affected acquired disorders that could lead to platelet dys-
individuals include unexplained bruising, epi- function or medications that could interfere with
staxis, menorrhagia, mucocutaneous bleeding, the function of platelets. A detailed drug history
and excessive bleeding following surgical is important as many common over-the-counter
procedures such as dental extractions or after medications can interfere with platelet function,
traumatic events. Certain IPD can cause a severe most notable being NSAIDS. However, occa-
bleeding diathesis that manifest as spontaneous sional herbal supplements can interfere with
108 D. Sterrenberg and S. Nand

a-GRANULES:
Gray platelet syndrome
Quebec platelet syndrome, a,d-SPD

METABOLISM:
IV VI CD9 Glycogen synthetase
Production of ATP

αIIbβ3
α6β1

ENZYMES:
Cyclooxygenase
TXA2 synthetase α5β1
Lipoxygenase

SIGNALING PATHWAYS:
α2β1 G-proteins (Ga9Ga11)
IX Phospholipases
V Phosphorylations
Ibβ
DENSE GRANULES: Ibα
Hermansky-Pudlak, Chediak-Higashi CYTOSKELETON:
and Griscelli syndromes, d-SPD MYH9 disorders and
giant platelet syndromes
Wiskott-Aldrich syndrome

Fig. 7.3 Disorders affecting intracellular organelles or cytosolic portions of proteins of platelets [75]. Reprinted from
Nurden and Nurden et al. Congenital Disorders with Associated Platelet Dysfunction

platelet function too. Patients should also undergo abnormal in most IPD, especially in Glanzmann
screening for more common causes of coagulop- Thrombasthenia or Bernard-Soulier where the
athy such as VWD or hemophilia. In practicality closure time can be especially prolonged. How-
this often occurs alongside the workup for a ever, there have been false negatives recorded
platelet function disorder [70]. with PFA-100 especially with the various SPD
Laboratory examination should include a and as such a normal PFA-100 should not stop a
CBC with peripheral blood smear examination workup for IDP. Platelet aggregations studies are
under the microscope looking primarily at the generally the next step in the diagnosis of a pos-
size and morphology of the platelets as well as sible IDP. They measure the ability of platelets
any concurrent cellular changes in the leukocytes to aggregate in response to typical agonists to
or red blood cells. All patients should have coag- aggregation, ADP, adrenaline, collagen, arachi-
ulation studies performed including a PT and donic acid, ristocetin, and thromboxane. Res-
aPTT, which are usually normal and a workup of ponse curves are then generated for each agonist
VWD including a VWF:Ag and VWF:RCo. If a and compared to a reference range. However, this
bleeding time is sent it can be normal, minimally test can be inaccurate in the setting of NSAID use
prolonged, or severely prolonged depending on or thrombocytopenia. Once again a normal plate-
the disorder and its severity. The Platelet Function let aggregation study does not necessarily rule
Analyzer-100 is a screening test measuring out a diagnosis of IDP, and if this remains a clini-
platelet adhesion to a variety of surfaces and is cal concern patients should undergo specialized
7

Table 7.4 Prevalence, inheritance patterns, bleeding risk, and treatment of RICD [5, 8, 63, 73, 74]
Correlation of
factor levels to Spontaneous ICH, % of
Factor deficiency Prevalence Inheritance Lab findings bleeding risk bleeding patients Other associations Treatment
Factor I 1:1,000,000 AR ↑PT Strong Yes Yes, 5 % Thrombosis, FIC
Afibrinogenemia AR ↑PTT pregnancy loss cryo
Congenital Coagulation Disorders

Hypofibrinogenemia AD ↑TT
dysfibrinogenemia ↑reptilase test
Factor II prothrombin 1:2,000,000 AR ↑PT Unknown Yes Yes, 7 % PCC, FFP
↑ aPTT
Factor V 1:1,000,000 AR ↑PT Weak Yes Yes, 8 % thrombosis FFP
↑ aPTT
Factor V + factor VIII 1:2,000,000 AR ↑PT Strong No No FFP
↑ aPTT
Factor VII 1:500,000 AR ↑PT Weak Yes Yes, 20 % Thrombosis rVIIa
nl aPTT
Factor X 1:1,000,000 AR ↑PT Strong Yes Yes, 13 % Pregnancy loss PCC
↑ aPTT
Factor XIII 1:2,000,000 AR nl PT Strong Yes Yes, 34 % Pregnancy loss, rXIII
nl aPTT impaired wound FXIIIC
healing FFP
VKDFD AR ↑PT Unknown Yes Yes Skeletal abnormalities Vitamin K
↑ aPTT FFP
PCC
rVIIa
109
110 D. Sterrenberg and S. Nand

additional testing such as measurement of ATP/ Table 7.5 Classification of inherited disorders of platelet
ADP content release via a platelet nucleotide/ function [69]
ATP release, platelet flow cytometry to measure 1. Disorders of Adhesion: Defects in platelet–vessel
platelet activation or surface glycoproteins, or wall interaction
dense granule release or phospholipid expression (a) von Willebrand disease
(b) Bernard–Soulier syndrome (deficiency or
(GPIB/IX/V and GPIIb/IIIa), or molecular analy-
defect in GPIb)
sis can be considered [70]. These tests are 2. Disorders of aggregation: Defects in platelet–
only performed at a small number of specialized platelet interaction
centers. (a) Congenital afibrinogenemia (deficiency or
dysfunctional plasma fibrinogen)
(b) Glanzmann’s thrombasthenia (deficiency or
defect in GPIIb-IIIa)
Treatment
3. Disorders of platelet secretion and abnormalities of
granules
Despite the clinical heterogeneity seen with IDPs (a) Storage pool deficiency
their treatment is largely the same and involves • α-granule disorders
the administration of donor platelets in order to – Gray platelet syndrome
achieve an adequate number of active platelets – Quebec platelet disorder
for normal hemostasis. Severe IDP such as • Dense granule disorders
Glanzmann Thrombasthenia or Bernard Soulier – Hemansky–Pudlak syndrome
Syndrome may need prophylactic administration – Chediak–Higashi syndrome
of platelets to prevent spontaneous hemorrhage. – Idiopathic dense granule disease
The main concern with the use of platelets 4. Disorders of platelet secretion and signal
with these disorders is that patients may, and transduction
often will, become alloimmunized and thus (a) Receptor defects: Defects in platelet–agonist
interaction with thromboxane a2, collagen,
refractory to platelet transfusions. Alloimmunity ADP, epinephrine
is the development of antibodies against either (b) Defects in G-protein activation
HLA Antigen mismatch on the surface of donor • Gαq deficiency
platelets or the development of antibodies against • Gαs abnormalities
the GpIIb/IIIa or GPIb/V/IX receptors on donor • Gαi1 deficiency
platelets that are lacking on platelets of patients (c) Defects in phosphatidylinositol metabolism:
with GT or BSS, respectively. Once the patient Phospholipase C-2 deficiency
becomes alloimmunized they are refractory to (d) Defects in calcium mobilization
subsequent platelet transfusions. For patients (e) Defects in protein phosphorylation (pleckstrin)
PKD-y deficiency
who are alloimmunized due to an HLA mis-
(f) Abnormalities in arachidonic acid pathways
match, they can be given HLA matched single and thromboxane A2 synthesis impaired
donor platelets if available. In all patients with a liberation of arachidonic acid
platelet function disorder, they should have • Cyclooxygenase deficiency
leukocyte reduced platelet transfusions in order • Thromboxane synthase deficiency
to try and prevent this from occurring. In addi- 5. Defects in cytoskeletal regulation
tion, platelet transfusions should be performed (a) Wiskott–Aldrich
only if necessary. Unfortunately for GT and BSS, 6. Disorders of platelet–coagulant protein interaction
(membrane phospholipid defects)
these patients often require prophylactic adminis-
(a) Scott Syndrome
tration of platelets in order to prevent spontane-
7. Miscellaneous
ous hemorrhage.
In milder phenotypes of IDP, patients can take
antifibrinolytics or DDAVP perioperatively sur- in the setting of major surgical procedures and
rounding mild procedures or dental extractions or acute severe hemorrhage, platelet transfusions
acutely in the setting of mild bleeding episodes in are required but can be given in addition to
order to prevent platelet transfusions. However, Amicar, tranexamic acid, or DDAVP. For patients
7
Table 7.6 Inherited thrombocytopenia: genetic mutations and associated phenotype [75]. Reprinted from Nurden and Nurden et al. Congenital Disorders with Associated
Platelet Dysfunction
Affected gene
Syndrome chromosomal location Inheritance Associated phenotype
MYH9-related diseases: May–Hegglin MYH9 AD Various combinations of leukocyte inclusions,
anomaly, Fechtner, Epstein, and 22q12–13 deafness, nephritis, cataracts. Large platelets
Sebastian syndromes
Mediterranean GPIBA, possibly others AD Large platelets
macrothrombocytopenia 17p 13
Bernard–Soulier syndrome GPIBA, GPIBB, GF9 AR Giant platelets, platelet adhesion defect, abnormal
17p 13, 22q11, and 3q21 aggregation of thrombin
Platelet-type VWD GPIBA AD Enlarged platelets. Defective adhesion due DO
Congenital Coagulation Disorders

17p 13 spontaneous binding of VWF to GPIbα and cleavage


of VWF multimers
Familial platelet disorder/acute RUNXI (CBFA2, AMLI) AD Myelodysplasia, propensity for leukemia. Platelet
myelogenous leukemia 21q22 dysfunction
Chromosome 10/THC2 FLJ14813 AD None
10p 11–12
Paris-Trousseau/Jacobsen syndromes Hemizygous deletion AD Cardiac and facial defects, mental retardation
including FL11 Enlarged platelets and large granules
11q23
Gray platelet Syndrome Unknown Mostly recessive Myelofibrosis, Enlarged platelets with no α-granules,
platelet dysfunction
Congenital amegakaryocytic c-MPL AR Severe thrombocytopenia at birth. Progressive aplasia
thrombocytopenia (CAMT) 1p34
Thrombocytopenia and absent radii Large deletion AR Shortened/absent radii bilaterally
(TAR) 1q21.1
Amegakaryocytic thrombocytopenia HOXA11 AD Fused radius, incomplete range of motion
with radio-ulnar synostosis 7pl5–14
Wiskott–Aldrich syndrome WAS X-linked Immunodeficiency, eczema, lymphoma, small
Xp 11.23-p11.22 platelets. Defective platelet and lymphocyte function
X-linked thrombocytopenia (XLT) WAS X-linked Small platelets, no immune problems
GATA-1-related thrombocytopenia GATAI X-linked Dyserythropoiesis ± anemia, β-thalassemia in some
with dyserythropoiesis Xp11.23 patients (XLTT). Platelet dysfunction, large platelets
AD autosomal dominant, AR autosomal recessive
AP autosomal recessive inheritance, AD autosomal dominant inheritance. The DiGeorge velocardiofacial syndrome is occasionally familial but largely de novo in origin and is
111

not included in the Table Type 2BVWD is accompanied by a familial thrombocytopenia is some families
112 D. Sterrenberg and S. Nand

that have developed allo-immunity, there have 13. Kaufmann JE, Vischer UM. Cellular mechanisms of
been studies reporting adequate hemostasis after the hemostatic effects of desmopressin (DDAVP).
J Thromb Haemost. 2003;1:682–9. doi:10.1046/
rVIIa (Novoseven), particularly in patients with j.1538-7836.2003.00190.x.
GT (Tables 7.5 and 7.6). 14. Castaman G, Lattuada A, Mannucci PM, Rodeghiero
F. Factor VIII:C Increases after Desmopressin in a
Subgroup of Patients with Autosomal Recessive
Severe von Willebrand Disease. Br J Haematol.
References 1995;89(1):147–51.
15. Mannucci PM, Kempton C, Millar C, et al. Pharma-
1. Rodeghiero F, Castaman G, Dini E. Epidemiological cokinetics and safety of a novel recombinant human
investigation of the prevalence of von Willebrand’s von Willebrand factor manufactured with a plasma-
disease. Blood. 1987;69:454–9. free method: A prospective clinical trial. Blood. 2013;
2. Werner EJ, Broxson EH, Tucker EL, Giroux DS, 122:648–57. doi:10.1182/blood-2013-01-479527.
Shults J, Abshire TC. Prevalence of von Willebrand 16. Rodeghiero F, Castaman G, Tosetto A. How I treat
disease in children: a multiethnic study. J Pediatr. von Willebrand disease. Blood. 2009;114:1158–65.
1993;123:893–8. doi:10.1182/blood-2009-01-153296.
3. Bowman M, Hopman W, Rapson D, Lilicrap D, Silva 17. Verstraete M. Clinical application of inhibitors of
M, James P. Brief Report: A Prospective Evaluation of fibrinolysis. Drugs. 1985;29:236–61. doi:10.2165/
the Prevalence of Symptomatic von Willebrand 00003495-198529030-00003.
Disease (VWD) in a Pediatric Primary Care Popu- 18. Thompson AR, Gill JC, Ewenstein BM, Mueller-
lation. Pediatr Blood Cancer. 2010;55:171–3. Velten G, Schwartz BA. Successful Treatment for
doi:10.1002/pbc. Patients with von Willebrand Disease Undergoing
4. Goodeve AC. The genetic basis of von Willebrand Urgent Surgery Using Factor VIII/VWF Concentrate
disease. Blood Rev. 2010;24:123–34. doi:10.1016/j. (Humate-P). Haemophilia. 2004;10(1):42–51.
blre.2010.03.003. 19. Federici AB, Castaman G, Franchini M, et al. Clinical
5. Zhang X, Halvorsen K, Zhang C-Z, Wong WP, use of Haemate P in inherited von Willebrand’s
Springer TA. Mechanoenzymatic cleavage of the ultra- disease: a cohort study on 100 Italian patients.
large vascular protein von Willebrand factor. Science. Haematologica. 2007;92:944–51. http://www.ncbi.
2009;324:1330–4. doi:10.1126/science.1170905. nlm.nih.gov/pubmed/17606445.
6. James PD, Lillicrap D. The molecular characteri- 20. Franchini M. Surgical prophylaxis in von Willebrand’s
zation of von Willebrand disease: Good in parts. disease: A difficult balance to manage. Blood Transfus.
Br J Haematol. 2013;161:166–76. doi:10.1111/bjh. 2008;6 Suppl 2:s33–8. doi:10.2450/2008.0035-08.
12249. 21. Makris M, Colvin B, Gupta V, Shields ML, Smith
7. Koppelman SJ, van Hoeij M, Vink T, et al. MP. Venous thrombosis following the use of interme-
Requirements of von Willebrand factor to protect diate purity FVIII concentrate to treat patients with
factor VIII from inactivation by activated protein von Willebrand’s disease. Thromb Haemost.
C. Blood. 1996;87:2292–300. 2002;88:387–8. doi:10.1267/THRO88020987.
8. Conlan MG, Folsom AR, Finch A, et al. Associations 22. Mannucci PM. Treatment of von Willebrand’s dis-
of factor VIII and von Willebrand factor with age, ease. N Engl J Med. 2004;351:683–94. doi:10.1056/
race, sex, and risk factors for atherosclerosis: The NEJMra040403.
Atherosclerosis Risk in Communities (ARIC) Study. 23. Mannucci PM. Back to the future: A recent history of
Thromb Haemost. 1993;70:380–5. haemophilia treatment. Haemophilia. 2008;14:10–8.
9. Jenkins PV. O'Donnell JS. ABO blood group deter- doi:10.1111/j.1365-2516.2008.01708.x.
mines plasma von Willebrand factor levels: A biologic 24. Giangrande PLF. The molecular basis of hemophilia.
function after all? Transfusion. 2006;46:1836–44. In: Molecular Hematology: Third Edition; 2010:219–
doi:10.1111/j.1537-2995.2006.00975.x. 232. doi:10.1002/9781444318531.ch18.
10. Sadler JE, Budde U, Eikenboom JCJ, et al. Update on 25. Stonebraker JS, Bolton-Maggs PHB, Michael Soucie
the pathophysiology and classification of von Willebrand J, Walker I, Brooker M. A study of variations in the
disease: a report of the Subcommittee on von Willebrand reported haemophilia A prevalence around the world.
Factor. J Thromb Haemost. 2006;4:2103–14. Haemophilia. 2010;16:20–32. doi:10.1111/j.1365-
doi:10.1111/j.1538-7836.2006.02146.x. 2516.2009.02127.x.
11. von Lillicrap D. Willebrand disease: advances in 26. Kasper CK, Lin JC. Prevalence of sporadic and
pathogenetic understanding, diagnosis, and therapy. familial haemophilia. Haemophilia. 2007;13:90–2.
Blood. 2013;122:3735–40. doi:10.1182/blood-2013- doi:10.1111/j.1365-2516.2006.01397.x.
06-498303. 27. Di Michele DM, Gibb C, Lefkowitz JM, Ni Q, Gerber
12. Ragni MV, Bontempo FA, Cortese HA. Von Willebrand LM, Ganguly A. Severe and moderate haemophilia A
disease and bleeding in women. Haemophilia. 1999; and B in US females. Haemophilia. 2014;20(2):e136–
5:313–7. doi:10.1046/j.1365-2516.1999.00342.x. 43. doi:10.1111/hae.12364.
7 Congenital Coagulation Disorders 113

28. Soucie JM, Evatt B, Jackson D. Occurrence of treatment regimens. J Thromb Haemost. 2010;8:269–
hemophilia in the United States. The Hemophilia 75. doi:10.1111/j.1538-7836.2009.03703.x.
Surveillance System Project Investigators. Am 42. Barnes C. Importance of pharmacokinetics in the
J Hematol. 1998;59:288–94. doi:10.1002/(SICI)1096- management of hemophilia. Pediatr Blood Cancer.
8652(199812)59:4<288::AID-AJH4>3.0.CO; 2013;60 Suppl 1:S27–9. doi:10.1002/pbc.24339.
2-I [pii]. 43. Srivastava A, Brewer AK, Mauser-Bunschoten EP,
29. Mannucci PM, Franchini M. Is haemophilia B less et al. Guidelines for the management of hemophilia.
severe than haemophilia A? Haemophilia. 2013;19: Haemophilia. 2013;19(1):e1–47. doi:10.1111/j.1365-
499–502. doi:10.1111/hae.12133. 2516.2012.02909.x.
30. Plug I, Van Der Bom JG, Peters M, et al. Mortality 44. Mannucci PM. Desmopressin (DDAVP) in the treat-
and causes of death in patients with hemophilia, ment of bleeding disorders: the first 20 years. Blood.
1992-2001: a prospective cohort study. J Thromb 1997;90:2515–21.
Haemost. 2006;4:510–6. doi:10.1111/j.1538-7836. 45. Brettler DB. Inhibitors in congenital haemophilia.
2006.01808.x. Baillieres Clin Haematol. 1996;9:319–29. doi:10.1016/
31. Bowen DJ. Haemophilia A, and haemophilia B: S0950-3536(96)80066-7.
molecular insights. Mol Pathol. 2002;55:127–44. 46. Gouw SC, Van Den Berg HM, Oldenburg J, et al. F8
doi:10.1136/mp.55.2.127. gene mutation type and inhibitor development in
32. Federici AB. The factor VIII/von Willebrand factor patients with severe hemophilia A: Systematic review
complex: basic and clinical issues. Haematologica. and meta-analysis. Blood. 2012;119:2922–34.
2003;88:EREP02. doi:10.1182/blood-2011-09-379453.
33. Lillicrap D. The molecular basis of haemophilia 47. Astermark J, Oldenburg J, Escobar M, White GC,
B. Haemophilia. 1998;4:350–7. Berntorp E. The Malmö International Brother Study
34. Kulkarni R, Presley R, Lusher J, et al. Complications (MIBS). Genetic defects and inhibitor development in
of hemophilia in children in the first two years of life: siblings with severe hemophilia A. Haematologica.
A report from the centers for disease control (CDC) 2005;90:924–30.
universal data collection (UDC) system. Haemophilia. 48. Astermark J, Berntorp E, White GC, Kroner BL. The
2012;18. Malmo International Brother Study (MIBS): further
35. Richards M, Lavigne Lissalde G, Combescure C, support for genetic predisposition to inhibitor devel-
et al. Neonatal bleeding in haemophilia: A European opment in hemophilia patients. Haemophilia.
cohort study. Br J Haematol. 2012;156:374–82. 2001;7:267–72.
doi:10.1111/j.1365-2141.2011.08967.x. 49. Turecek PL, Váradi K, Gritsch H, Schwarz HP.
36. Valentino LA. Blood-induced joint disease: The FEIBA: mode of action. Haemophilia. 2004;10 Suppl
pathophysiology of hemophilic arthropathy. J Thromb 2:3–9. doi:10.1111/j.1365-2516.2004.00934.x.
Haemost. 2010;8:1895–902. doi:10.1111/j.1538-7836. 50. Lindley CM, Sawyer WT, Macik BG, et al. Pharma-
2010.03962.x. cokinetics and Pharmacodynamics of Recombinant
37. Sørensen B, Benson GM, Bladen M, et al. Mana- Factor VIIa. Clin Pharmacol Ther. 1994;55:638–48.
gement of muscle haematomas in patients with severe 51. Kempton CL, White GC. How we treat a hemophilia
haemophilia in an evidence-poor world. Haemophilia. A patient with a factor VIII inhibitor. Blood.
2012;18:598–606. doi:10.1111/j.1365-2516.2011. 2009;113:11–7. doi:10.1182/blood-2008-06-160432.
02720.x. 52. Asakai R, Chung DW, Davie EW, Seligsohn U.
38. Witmer C, Presley R, Kulkarni R, Michael Soucie J, Factor XI deficiency in Ashkenazi Jews in Israel.
Manno CS, Raffini L. Associations between intracra- N Engl J Med. 1991;325:153–8. doi:10.1056/
nial haemorrhage and prescribed prophylaxis in a NEJM199107183250303.
large cohort of haemophilia patients in the United 53. Bolton-Maggs PHB, Patterson DA, Wensley RT,
States. Br J Haematol. 2011;152:211–6. doi:10.1111/ Tuddenham EGD. Definition of the bleeding tendency
j.1365-2141.2010.08469.x. in factor XI-deficient kindreds: A clinical and labora-
39. Iorio A, Marchesini E, Marcucci M, Stobart K, Chan tory study. Thromb Haemost. 1995;73:194–202.
AK. Clotting factor concentrates given to prevent 54. Bolton-Maggs PH, Young Wan-Yin B, McCraw AH,
bleeding and bleeding-related complications in peo- Slack J, Kernoff PB. Inheritance and bleeding in fac-
ple with hemophilia A or B. Cochrane Database tor XI deficiency. Br J Haematol. 1988;69:521–8.
Syst Rev. 2011;9, CD003429. doi:10.1002/14651858. 55. Kadir RA, Lee CA, Sabin CA, Pollard D, Economides
CD003429.pub3. DL. Pregnancy in women with von Willebrand’s dis-
40. Josephson N. The hemophilias and their clinical man- ease or factor XI deficiency. Br J Obstet Gynaecol.
agement. ASH Educ Progr B. 2013;2013:261–7. 1998;105:314–21.
doi:10.1182/asheducation-2013.1.261. 56. Castaman G, Tosetto A, Rodeghiero F. Reduced von
41. Collins PW, Björkman S, Fischer K, et al. Factor VIII Willebrand factor survival in von Willebrand disease:
requirement to maintain a target plasma level in the pathophysiologic and clinical relevance. J Thromb
prophylactic treatment of severe hemophilia A: Haemost. 2009;7 Suppl 1:71–4. doi:10.1111/
Influences of variance in pharmacokinetics and j.1538-7836.2009.03381.x.
114 D. Sterrenberg and S. Nand

57. Livnat T, Martinowitz U, Mansharov R, Ariella Z. Disorders in Children. Transfus Med Hemother.
Thrombin Generation Test Can Predict Bleeding 2010;37:231–5. doi:10.1159/000320762.
Tendency In Patients With Severe Factor XI Deficiency. 66. Hayward CPM, Rao AK, Cattaneo M. Congenital
Blood. 2013;122:3600. http://bloodjournal.hematolo- platelet disorders: Overview of their mechanisms,
gylibrary.org/content/122/21/3600.abstract. diagnostic evaluation and treatment. Haemophilia.
58. Bolton-Maggs PHB, Perry DJ, Chalmers EA, et al. 2006;12:128–36. doi:10.1111/j.1365-2516.2006.
The rare coagulation disorders - Review with guide- 01270.x.
lines for management from the United Kingdom 67. Knöfler R, Olivieri M, Weickardt S, Eberl W, Streif
Haemophilia Centre Doctors’ Organisation. W. First results of the THROMKID study: a quality
Haemophilia. 2004;10:593–628. doi:10.1111/j.1365- project for the registration of children und adolescents
2516.2004.00944.x. with hereditary platelet function defects in Germany,
59. Salomon O, Zivelin A, Livnat T, et al. Prevalence, Austria, and Switzerland. Hamostaseologie. 2007;
causes, and characterization of factor XI inhibitors in 27:48–53. doi:07010048 [pii].
patients with inherited factor XI deficiency. Blood. 68. Salles II, Feys HB, Iserbyt BF, De Meyer SF,
2003;101:4783–8. doi:10.1182/blood-2002-09-2794. Vanhoorelbeke K, Deckmyn H. Inherited traits affect-
60. Livnat T, Tamarin I, Mor Y, et al. Recombinant acti- ing platelet function. Blood Rev. 2008;22:155–72.
vated factor VII and tranexamic acid are haemosta- doi:10.1016/j.blre.2007.11.002.
tically effective during major surgery in factor 69. Handin RI. Inherited platelet disorders. Hematology
XI-deficient patients with inhibitor antibodies. Am Soc Hematol Educ Program. 2005:396–402.
Thromb Haemost. 2009;102:487–92. doi:10.1160/ doi:10.1182/asheducation-2005.1.396.
TH09-03-0172. 70. Kirchmaier CM, Pillitteri D. Diagnosis and mana-
61. Nichols WC, Seligsohn U, Zivelin A, et al. Mutations gement of inherited platelet disorders. Transfus Med
in the ER-Golgi intermediate compartment protein Hemother. 2010;37:237–46. doi:10.1159/000320257.
ERGIC-53 cause combined deficiency of coagulation 71. Yawn B, Nichols WL, Rick ME. Diagnosis and
factors V and VIII. Cell. 1998;93:61–70. doi:10.1016/ management of von Willebrand disease: guidelines
S0092-8674(00)81146-0. for primary care. Am Fam Physician. 2009;80:
62. Napolitano M, Mariani G, Lapecorella M. Hereditary 1261–8.
combined deficiency of the vitamin K-dependent clot- 72. Nichols WL, Hultin MB, James AH, et al. von
ting factors. Orphanet J Rare Dis. 2010;5:21. Willebrand disease (VWD): evidence-based diagno-
doi:10.1186/1750-1172-5-21. sis and management guidelines, the National Heart,
63. Acharya SS, Coughlin A, Dimichele DM, et al. Rare Lung, and Blood Institute (NHLBI) Expert Panel
bleeding disorder registry: Deficiencies of factors II, report (USA). Haemophilia. 2008;14:171–232.
V, VII, X, XIII, fibrinogen and dysfibrinogenemias. doi:10.1111/j.1365-2516.2007.01643.x.
J Thromb Haemost. 2004;2:248–56. doi:10.1111/ 73. Escobar MA. Advances in the treatment of inherited
j.1538-7836.2003.t01-1-00553.x. coagulation disorders. Haemophilia. 2013;19:648–59.
64. Bolton-Maggs PHB, Chalmers EA, Collins PW, et al. doi:10.1111/hae.12137.
A review of inherited platelet disorders with guide- 74. Acharya SS, Dimichele DM. Rare inherited disorders
lines for their management on behalf of the UKHCDO. of fibrinogen. Haemophilia. 2008;14:1151–8.
Br J Haematol. 2006;135:603–33. doi:10.1111/ doi:10.1111/j.1365-2516.2008.01831.x.
j.1365-2141.2006.06343.x. 75. Nurden P, Nurden AT. Congenital disorders associ-
65. Knöfler R, Streif W. Strategies in Clinical and ated with platelet dysfunctions. Thromb Haemost.
Laboratory Diagnosis of Inherited Platelet Function 2008;99:253–63. doi:10.1160/TH07-09-0568.

You might also like