Sunteți pe pagina 1din 24

4

Hemostasis, Surgical Bleeding,


and Transfusion
chapter Bryan Cotton, John B. Holcomb, Matthew Pommerening,
Kenneth Jastrow, and Rosemary A. Kozar

Biology of Hemostasis 85 Acquired Hypofibrinogenemia / 92 Indications for Replacement of Blood and


Vascular Constriction / 85 Myeloproliferative Diseases / 92 Its Elements / 97
Platelet Function / 85 Coagulopathy of Liver Disease / 92 Volume Replacement / 98
Coagulation / 86 Coagulopathy of Trauma / 93 New Concepts in Resuscitation / 98
Fibrinolysis / 88 Acquired Coagulation Inhibitors / 93 Complications of Transfusion / 100
Congenital Factor Deficiencies 88 Anticoagulation and Bleeding / 94 Tests of Hemostasis and
Coagulation Factor Deficiencies / 88 Transfusion 96 Blood Coagulation 102
Platelet Functional Defects / 89 Background / 96 Evaluation of Excessive
Acquired Hemostatic Defects 90 Replacement Therapy / 96 Intraoperative or
Postoperative Bleeding 104
Platelet Abnormalities / 90

BIOLOGY OF HEMOSTASIS platelets are normally removed by the spleen and have an aver-
age life span of 7 to 10 days.
Hemostasis is a complex process whose function is to limit Platelets play an integral role in hemostasis by forming
blood loss from an injured vessel. Four major physiologic a hemostatic plug and by contributing to thrombin formation
events participate in the hemostatic process: vascular constric- (Fig. 4-2). Platelets do not normally adhere to each other or
tion, platelet plug formation, fibrin formation, and fibrinolysis. to the vessel wall but can form a plug that aids in cessation of
Although each tends to be activated in order, the four pro- bleeding when vascular disruption occurs. Injury to the intimal
cesses are interrelated so that there is a continuum and mul- layer in the vascular wall exposes subendothelial collagen to
tiple reinforcements. The process is shown schematically in which platelets adhere. This process requires von Willebrand
Fig. 4-1. factor (vWF), a protein in the subendothelium that is lacking in
Vascular Constriction patients with von Willebrand’s disease. vWF binds to glycopro-
Vascular constriction is the initial response to vessel injury. It is tein (GP) I/IX/V on the platelet membrane. Following adhesion,
more pronounced in vessels with medial smooth muscles and is platelets initiate a release reaction that recruits other platelets
dependent on local contraction of smooth muscle. Vasoconstric- from the circulating blood to seal the disrupted vessel. Up to
tion is subsequently linked to platelet plug formation. Throm- this point, this process is known as primary hemostasis. Platelet
boxane A2 (TXA2) is produced locally at the site if injury via aggregation is reversible and is not associated with secretion.
the release of arachidonic acid from platelet membranes and Additionally, heparin does not interfere with this reaction, and
is a potent constrictor of smooth muscle. Similarly, endothelin thus, hemostasis can occur in the heparinized patient. Adenosine
synthesized by injured endothelium and serotonin (5-hydroxy- diphosphate (ADP) and serotonin are the principal mediators in
tryptamine [5-HT]) released during platelet aggregation are platelet aggregation.
potent vasoconstrictors. Lastly, bradykinin and fibrinopeptides, Arachidonic acid released from the platelet membranes is
which are involved in the coagulation schema, are also capable converted by cyclooxygenase to prostaglandin G2 (PGG2) and
of contracting vascular smooth muscle. then to prostaglandin H2 (PGH2), which, in turn, is converted to
The extent of vasoconstriction varies with the degree of TXA2. TXA2 has potent vasoconstriction and platelet aggrega-
vessel injury. A small artery with a lateral incision may remain tion effects. Arachidonic acid may also be shuttled to adjacent
open due to physical forces, whereas a similarly sized vessel endothelial cells and converted to prostacyclin (PGI2), which
that is completely transected may contract to the extent that is a vasodilator and acts to inhibit platelet aggregation. Platelet
bleeding ceases spontaneously. cyclooxygenase is irreversibly inhibited by aspirin and revers-
ibly blocked by nonsteroidal anti-inflammatory agents, but is
Platelet Function not affected by cyclooxygenase-2 (COX-2) inhibitors.
Platelets are anucleate fragments of megakaryocytes. The nor- In the second wave of platelet aggregation, a release reac-
mal circulating number of platelets ranges between 150,000 and tion occurs in which several substances including ADP, Ca2+,
400,000/μL. Up to 30% of circulating platelets may be seques- serotonin, TXA2, and α-granule proteins are discharged. Fibrin-
tered in the spleen. If not consumed in a clotting reaction, ogen is a required cofactor for this process, acting as a bridge for

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
Key Points
1 The life span of platelets ranges from 7 to 10 days. Drugs that 4 Therapeutic anticoagulation preoperatively and postop-
interfere with platelet function include aspirin, clopidogrel, pra- eratively is becoming increasingly more common. The
sugrel, dipyridamole, and the glycoprotein IIb/IIIa (GP IIb/IIIa) patient’s risk of intraoperative and postoperative bleeding
inhibitors. Approximately 5 to 7 days should pass from the time should guide the need for reversal of anticoagulation
the drug is stopped until an elective procedure is performed. therapy preoperatively and the timing of its reinstatement
2 The acute coagulopathy of trauma results from a combination postoperatively.
of activation of protein C and hyperfibrinolysis. It is distinct 5 Damage control resuscitation has three basic components:
from disseminated intravascular coagulation, is present on permissive hypotension, minimizing crystalloid-based
arrival to the emergency department, and is associated with an resuscitation, and the administration of predefined blood
increase in mortality. products.
3 Newer anticoagulants like dabigatran and rivaroxaban have no 6 The need for massive transfusion should be anticipated,
readily available method of detection of the degree of antico- and guidelines should be in place to provide early and
agulation and may not be readily reversible. increased amounts of red blood cells, plasma, and platelets.

the GP IIb/IIIa receptor on the activated platelets. The release thrombin (factor IIa) by activated factor X (Xa) in the pres-
reaction results in compaction of the platelets into a plug, a pro- ence of factor V and calcium, and it is involved in the reaction
cess that is no longer reversible. Thrombospondin, another pro- by which activated factor IX (IXa), factor VIII, and calcium
tein secreted by the α-granule, stabilizes fibrinogen binding to activate factor X. Platelets may also play a role in the initial
the activated platelet surface and strengthens the platelet-platelet activation of factors XI and XII.
interactions. Platelet factor 4 (PF4) and α-thromboglobulin are
also secreted during the release reaction. PF4 is a potent heparin Coagulation
antagonist. The second wave of platelet aggregation is inhibited Hemostasis involves a complex interplay and combination of
by aspirin and nonsteroidal anti-inflammatory drugs, by cyclic interactions between platelets, the endothelium, and multiple
adenosine monophosphate (cAMP), and by nitric oxide. As a circulating or membrane-bound coagulation factors. While
consequence of the release reaction, alterations occur in the a bit simplistic and not reflective of the depth or complexity
phospholipids of the platelet membrane that allow calcium and of these interactions, the coagulation cascade has traditionally
clotting factors to bind to the platelet surface, forming enzymati- been depicted as two possible pathways converging into a single
cally active complexes. The altered lipoprotein surface (some- common pathway (Fig. 4-3). While this pathway reflects the
times referred to as platelet factor 3) catalyzes reactions that basic process and sequences that lead to the formation of a clot,
are involved in the conversion of prothrombin (factor II) to the numerous feedback loops, endothelial interplay, and platelet

1. Vascular phase 2. Platelet phase


(Vasoconstriction) (Platelets aggregate)
Common pathway
Intrinsic pathway Prothrombin Extrinsic pathway

Thrombin CA2+
CA2+v
Clotting factors Clotting factors
VIII, IX, X, XI, XII VII
Fibrin

3. Coagulation phase (Clot formation)


Figure 4-1.  Biology of hemostasis. The four phys-
iologic processes that interrelate to limit blood loss
from an injured vessel are illustrated and include
vascular constriction, platelet plug formation,
86 (Clot retraction) 4. Fibrinolysis (Clot destruction) fibrin clot formation, and fibrinolysis.

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
tests segregate abnormalities of clotting to one of the two arms. 87
Vascular endothelial
injury An elevated activated partial thromboplastin time (aPTT) is
associated with abnormal function of the intrinsic arm of the
Platelet hemostatic Vasoconstriction cascade (II, IX, X, XI, XII), while the prothrombin time (PT) is

CHAPTER 4
function associated with the extrinsic arm (II, VII, X). Vitamin K defi-
Subendothelial collagen ciency or warfarin use affects factors II, VII, IX, and X
Expanding from the basic concept of Fig. 4-3, the primary
pathway for coagulation is initiated by TF exposure following
Platelet adhesion secretion Coagulation activation
via tissue factor- subendothelial injury. Clot propagation ensues with what is a
ADP, serotonin,
(Reversible) factor VIIa sequence of four similar enzymatic reactions, each involving a
Ca2+, fibrinogen

Hemostasis, Surgical Bleeding, and Transfusion


proteolytic enzyme generating the next enzyme by cleaving its
Platelet aggregation secretion
IXa, Xa proenzyme, a phospholipid surface (e.g., platelet membrane) in
ADP, serotonin,
(Irreversible) Complexes on the presence of ionized calcium, and a helper protein. TF binds to
Ca2+, fibrinogen activated platelets
VIIa, and this complex catalyzes the activation of factor X to Xa.
Platelet aggregation
This complex is four orders of magnitude more active at convert-
Thrombin ing factor X than is factor VIIa alone and also activates factor IX
Platelet-fibrin + to IXa. Factor Xa, together with Va, calcium, and phospholipid,
thrombus Fibrinogen
composes the prothrombinase complex that converts prothrom-
Figure 4-2.  Schematic of platelet activation and thrombus function. bin to thrombin. The prothrombinase complex is significantly
more effective at catalyzing its substrate than is factor Xa alone.
Thrombin is then involved with the conversion of fibrinogen to
functions are not included. The intrinsic pathway begins with fibrin and activation of factors V, VII, VIII, XI, and XIII.
the activation of factor XII that subsequently activates factors In building on the redundancy inherent in the coagu-
XI, IX, and VIII. In this pathway, each of the primary factors lation system, factor VIIIa combines with IXa to form the
is “intrinsic” to the circulating plasma, whereby no surface is intrinsic factor complex. Factor IXa is responsible for the bulk
required to initiate the process. In the extrinsic pathway, tissue of the conversion of factor X to Xa. This complex (VIIIa-IXa)
factor (TF) is released or exposed on the surface of the endo- is 50 times more effective at catalyzing factor X activation
thelium, binding to circulating factor VII, facilitating its activa- than is the extrinsic (TF-VIIa) complex and five to six orders
tion to VIIa. Each of these pathways continues on to a common of magnitude more effective than factor IXa alone.
sequence that begins with the activation of factor X to Xa Once formed, thrombin leaves the membrane surface
(in the presence of VIIIa). Subsequently, Xa (with the help of and converts fibrinogen by two cleavage steps into fibrin and
factor Va) converts factor II (prothrombin) to thrombin and then two small peptides termed fibrinopeptides A and B. Removal
factor I (fibrinogen) to fibrin. Clot formation occurs after fibrin of fibrinopeptide A permits end-to-end polymerization of the
monomers are cross-linked to polymers with the assistance of fibrin molecules, whereas cleavage of fibrinopeptide B allows
factor XIII. side-to-side polymerization of the fibrin clot. This latter step is
One convenient feature of depicting the coagulation cas- facilitated by thrombin-activatable fibrinolysis inhibitor (TAFI),
cade with two merging arms is that commonly used laboratory which acts to stabilize the resultant clot.

Extrinsic Intrinsic

Surface Inflammation
Vascular injury
Factor XII Factor XIIa Complement activation
Fibrinolysis
Kallikrein Prekallikrein
Tissue factor +
HMW kininogen ? Physiologic
factor VII
Surface

Factor XIa Factor XI


Tissue factor-Factor VIIa 2+
Ca

Factor IX Factor IXa Factor VIIIa Factor VIII


Ca
2+ Ca2+
Factor X Factor Xa Phospholipid
Factor Va
Ca2+ Factor V
Phospholipid
Prothrombin Thrombin
Factor XIII
(factor II) (factor IIa)
Ca2+ Fibrin
Fibrinogen Fibrin Figure 4-3. Schematic of the
Factor XIIIa coagulation system. HMW = high
X-Linked fibrin molecular weight.

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
88 In seeking to balance profound bleeding with overwhelm-
ing clot burden, several related processes exist to prevent prop-
Platelet Thrombin Fibrin FDP
agation of the clot beyond the site of injury.1 First, feedback
inhibition on the coagulation cascade deactivates the enzyme
complexes leading to thrombin formation. Thrombomodulin Plasminogen Plasmin
PART I

(TM) presented by the endothelium serves as a “thrombin sink”


by forming a complex with thrombin, rendering it no longer
available to cleave fibrinogen. This then activates protein C
(APC) and reduces further thrombin generation by inhibiting
factors V and VIII. Second, tissue plasminogen activator (tPA) tPA
BASIC CONSIDERATIONS

is released from the endothelium following injury, cleaving


plasminogen to initiate fibrinolysis. APC then consumes plas-
minogen activator inhibitor-1 (PAI-1), leading to increased tPA
Endothelium
activity and fibrinolysis. Building on the anticoagulant response
to inhibit thrombin formation, tissue factor pathway inhibitor Figure 4-4.  Formation of fibrin degradation products (FDPs). tPA =
(TFPI) is released, blocking the TF-VIIa complex and reducing tissue plasminogen activator.
the production of factors Xa and IXa. Antithrombin III (AT-
III) then neutralizes all of the procoagulant serine proteases and
also inhibits the TF-VIIa complex. The most potent mechanism As with clot formation, fibrinolysis is also kept in check
of thrombin inhibition involves the APC system. APC forms a through several robust mechanisms. tPA activates plasmino-
complex with its cofactor, protein S, on a phospholipid surface. gen more efficiently when it is bound to fibrin, so that plasmin
This complex then cleaves factors Va and VIIIa so they are no is formed selectively on the clot. Plasmin is inhibited by α2-
longer able to participate in the formation of TF-VIIa or pro- antiplasmin, a protein that is cross-linked to fibrin by factor XIII,
thrombinase complexes. This is of interest clinically in the form which helps to ensure that clot lysis does not occur too quickly.
of a genetic mutation, called factor V Leiden. In this setting, fac- Any circulating plasmin is also inhibited by α2-antiplasmin and
tor V is resistant to cleavage by APC, thereby remaining active circulating tPA or urokinase. Clot lysis yields FDPs including
as a procoagulant. Patients with factor V Leiden are predisposed E-nodules and D-dimers. These smaller fragments interfere with
to venous thromboembolic events. normal platelet aggregation, and the larger fragments may be
Degradation of fibrin clot is accomplished by plasmin, a incorporated into the clot in lieu of normal fibrin monomers.
serine protease derived from the proenzyme plasminogen. Plas- This may result in an unstable clot as seen in cases of severe
min formation occurs as a result of one of several plasminogen coagulopathy such as hyperfibrinolysis associated with trauma-
activators. tPA is made by the endothelium and other cells of induced coagulopathy or disseminated intravascular coagulopa-
the vascular wall and is the main circulating form of this family thy. The presence of D-dimers in the circulation may serve as a
of enzymes. tPA is selective for fibrin-bound plasminogen so marker of thrombosis or other conditions in which a significant
that endogenous fibrinolytic activity occurs predominately at activation of the fibrinolytic system is present. Another inhibi-
the site of clot formation. The other major plasminogen activa- tor of the fibrinolytic system is TAFI, which removes lysine
tor, urokinase plasminogen activator (uPA), also produced by residues from fibrin that are essential for binding plasminogen.
endothelial cells as well as by urothelium, is not selective for
fibrin-bound plasminogen. Of note, the thrombin-TM complex CONGENITAL FACTOR DEFICIENCIES
activates TAFI, leading to a mixed effect on clot stability. In
addition to inhibiting fibrinolysis directly, removal of the termi- Coagulation Factor Deficiencies
nal lysine on the fibrin molecule by TAFI renders the clot more Inherited deficiencies of all of the coagulation factors are seen.
susceptible to lysis by plasmin. However, the three most frequent are factor VIII deficiency
(hemophilia A and von Willebrand’s disease), factor IX defi-
Fibrinolysis ciency (hemophilia B or Christmas disease), and factor
Fibrin clot breakdown (lysis) allows restoration of blood flow XI deficiency. Hemophilia A and hemophilia B are inherited as
during the healing process following injury and begins at the sex-linked recessive disorders with males being affected almost
same time clot formation is initiated. Fibrin polymers are exclusively. The clinical severity of hemophilia A and hemo-
degraded by plasmin, a serine protease derived from the pro- philia B depends on the measurable level of factor VIII or factor
enzyme plasminogen. Plasminogen is converted to plasmin by IX in the patient’s plasma. Plasma factor levels less than 1%
one of several plasminogen activators, including tPA. Plasmin of normal are considered severe disease, factor levels between
then degrades the fibrin mesh at various places, leading to the 1% and 5% moderately severe disease, and levels between 5%
production of circulating fragments, termed fibrin degradation and 30% mild disease. Patients with severe hemophilia have
products (FDPs), cleared by other proteases or by the kidney and spontaneous bleeds, frequently into joints, leading to crippling
liver (Fig. 4-4). Fibrinolysis is directed by circulating kinases, arthropathies. Intracranial bleeding, intramuscular hematomas,
tissue activators, and kallikrein present in vascular endothelium. retroperitoneal hematomas, and gastrointestinal, genitourinary,
tPA is synthesized by endothelial cells and released by the cells and retropharyngeal bleeding are added clinical sequelae seen
on thrombin stimulation. Bradykinin, a potent endothelial- with severe disease. Patients with moderately severe hemophilia
dependent vasodilator, is cleaved from high molecular weight have less spontaneous bleeding but are likely to bleed severely
kininogen by kallikrein and enhances the release of tPA. Both after trauma or surgery. Mild hemophiliacs do not bleed sponta-
tPA and plasminogen bind to fibrin as it forms, and this trimo- neously and have only minor bleeding after major trauma or sur-
lecular complex cleaves fibrin very efficiently. After plasmin is gery. Since platelet function is normal in hemophiliacs, patients
generated, however, it cleaves fibrin somewhat less efficiently. may not bleed immediately after an injury or minor surgery as
VRG RELEASE: tahir99
https://kickass.to/user/tahir99/uploads/
they have a normal response with platelet activation and forma- of each per milliliter. However, factor V activity is decreased 89
tion of a platelet plug. At times, the diagnosis of hemophilia is because of its inherent instability. The half-life of prothrom-
not made in these patients until after their first minor procedure bin (factor II) is long (approximately 72 hours), and only about
(e.g., tooth extraction or tonsillectomy). 25% of a normal level is needed for hemostasis. Prothrombin

CHAPTER 4
Patients with hemophilia A or B are treated with factor complex concentrates can be used to treat deficiencies of pro-
VIII or factor IX concentrate, respectively. Recombinant factor thrombin or factor X. Daily infusions of FFP are used to treat
VIII is strongly recommended for patients not treated previously bleeding in factor V deficiency, with a goal of 20% to 25%
and is generally recommended for patients who are both human activity. Factor V deficiency may be coinherited with factor
immunodeficiency virus (HIV) and hepatitis C virus (HCV) VIII deficiency. Treatment of bleeding in individuals with the
seronegative. For factor IX replacement, the preferred products combined deficiency requires factor VIII concentrate and FFP.

Hemostasis, Surgical Bleeding, and Transfusion


are recombinant or high-purity factor IX. In general, activity Some patients with factor V deficiency are also lacking the fac-
levels should be restored to 30% to 40% for mild hemorrhage, tor V normally present in platelets and may need platelet trans-
50% for severe bleeding, and 80% to 100% for life-threatening fusions as well as FFP.
bleeding. Up to 20% of hemophiliacs with factor VIII defi-
Factor VII Deficiency.  Inherited factor VII deficiency is a
ciency develop inhibitors that can neutralize FVIII. For patients
rare autosomal recessive disorder. Clinical bleeding can vary
with low titers, inhibitors can be overcome with higher doses of
widely and does not always correlate with the level of FVII
factor VIII. For patients with high titer inhibitors, alternate treat-
coagulant activity in plasma. Bleeding is uncommon unless the
ments should be used and may include porcine factor VIII, pro-
level is less than 3%. The most common bleeding manifesta-
thrombin complex concentrates, activated prothrombin complex
tions involve easy bruising and mucosal bleeding, particularly
concentrates, or recombinant factor VIIa. For patients undergo-
epistaxis or oral mucosal bleeding. Postoperative bleeding is
ing elective surgical procedures, a multidisciplinary approach
also common, reported in 30% of surgical procedures.6 Treat-
with preoperative planning and replacement is recommended.2
ment is with FFP or recombinant factor VIIa. The half-life of
von Willebrand’s Disease.  von Willebrand’s disease (vWD), recombinant factor VIIa is only approximately 2 hours, but
the most common congenital bleeding disorder, is characterized excellent hemostasis can be achieved with frequent infusions.
by a quantitative or qualitative defect in vWF, a large glycopro- The half-life of factor VII in FFP is up to 4 hours.
tein responsible for carrying factor VIII and platelet adhesion. Factor XIII Deficiency.  Congenital factor XIII (FXIII) defi-
The latter is important for normal platelet adhesion to exposed ciency, originally recognized by Duckert in 1960, is a rare
subendothelium and for aggregation under high shear condi- autosomal recessive disease usually associated with a severe
tions. Patients with vWD have bleeding that is characteristic bleeding diathesis.7 The male-to-female ratio is 1:1. Although
of platelet disorders such as easy bruising and mucosal bleed- acquired FXIII deficiency has been described in association
ing. Menorrhagia is common in women. vWD is classified into with hepatic failure, inflammatory bowel disease, and myeloid
three types. Type I is a partial quantitative deficiency, type leukemia, the only significant association with bleeding in chil-
II is a qualitative defect, and type III is total deficiency. For dren is the inherited deficiency.8 Bleeding is typically delayed
bleeding, type I patients usually respond well to desmopressin because clots form normally but are susceptible to fibrinolysis.
(DDAVP). Type II patients may respond, depending on the par- Umbilical stump bleeding is characteristic, and there is a high
ticular defect. Type III patients are usually unresponsive. These risk of intracranial bleeding. Spontaneous abortion is usual in
patients may require vWF concentrates.3 women with factor XIII deficiency unless they receive replace-
Factor XI Deficiency.  Factor XI deficiency, an autosomal ment therapy. Replacement can be accomplished with FFP,
recessive inherited condition sometimes referred to as hemo- cryoprecipitate, or a factor XIII concentrate. Levels of 1% to
philia C, is more prevalent in the Ashkenazi Jewish population 2% are usually adequate for hemostasis.
but found in all races. Spontaneous bleeding is rare, but bleeding
may occur after surgery, trauma, or invasive procedures. Treat-
Platelet Functional Defects
Inherited platelet functional defects include abnormalities of
ment of patients with factor XI deficiency who present with
platelet surface proteins, abnormalities of platelet granules, and
bleeding or in whom surgery is planned and who are known
enzyme defects. The major surface protein abnormalities are
to have bled previously is with fresh frozen plasma (FFP).
thrombasthenia and Bernard-Soulier syndrome. Thrombasthe-
Each milliliter of plasma contains 1 unit of factor XI activity,
nia, or Glanzmann thrombasthenia, is a rare genetic platelet
so the volume needed depends on the patient’s baseline level,
disorder, inherited in an autosomal recessive pattern, in which
the desired level, and the plasma volume. Antifibrinolytics may
the platelet glycoprotein IIb/IIIa (GP IIb/IIIa) complex is either
be useful in patients with menorrhagia. Factor VIIa is recom-
lacking or present but dysfunctional. This defect leads to faulty
mended for patients with anti-factor XI antibodies, although
platelet aggregation and subsequent bleeding. The disorder was
thrombosis has been reported.4 There has been renewed interest
first described by Dr. Eduard Glanzmann in 1918.9 Bleeding
in factor XI inhibitors as antithrombotic agents, because patients
in thrombasthenic patients must be treated with platelet trans-
with factor XI deficiency generally have only minimal bleeding
fusions. The Bernard-Soulier syndrome is caused by a defect
risk unless a severe deficiency is present and seem to be pro-
in the GP Ib/IX/V receptor for vWF, which is necessary for
tected from thrombosis.5
platelet adhesion to the subendothelium. Transfusion of normal
Deficiency of Factors II (Prothrombin), V, and X.  Inher- platelets is required for bleeding in these patients.
ited deficiencies of factors II, V, and X are rare. These deficien- The most common intrinsic platelet defect is storage pool
cies are inherited as autosomal recessive. Significant bleeding disease. It involves loss of dense granules (storage sites for
in homozygotes with less than 1% of normal activity is encoun- ADP, adenosine triphosphate [ATP], Ca2+, and inorganic phos-
tered. Bleeding with any of these deficiencies is treated with phate) and α-granules. Dense granule deficiency is the most
FFP. Similar to factor XI, FFP contains one unit of activity prevalent of these. It may be an isolated defect or occur with
VRG RELEASE: tahir99
https://kickass.to/user/tahir99/uploads/
90 partial albinism in the Hermansky-Pudlak syndrome. Bleed- Quantitative Defects.  Shortened platelet survival is seen in
ing is variable, depending on the severity of the granule defect. immune thrombocytopenia, disseminated intravascular coagu-
Bleeding is caused by the decreased release of ADP from these lation, or disorders characterized by platelet thrombi such as
platelets. A few patients have been reported who have decreased thrombotic thrombocytopenic purpura and hemolytic uremic
numbers of both dense and α-granules. They have a more severe syndrome. Immune thrombocytopenia may be idiopathic or
PART I

bleeding disorder. Patients with mild bleeding as a consequence associated with other autoimmune disorders or low-grade B-cell
of a form of storage pool disease can be treated with DDAVP. It malignancies, and it may also be secondary to viral infections
is likely that the high levels of vWF in the plasma after DDAVP (including HIV) or drugs. Secondary immune thrombocytopenia
somehow compensate for the intrinsic platelet defect. With often presents with a very low platelet count, petechiae and pur-
more severe bleeding, platelet transfusion is required. pura, and epistaxis. Large platelets are seen on peripheral smear.
BASIC CONSIDERATIONS

Initial treatment consists of corticosteroids, intravenous gamma


Acquired Hemostatic Defects globulin, or anti-D immunoglobulin in patients who are Rh posi-
tive. Both gamma globulin and anti-D immunoglobulin are rapid
Platelet Abnormalities in onset. Platelet transfusions are not usually needed unless cen-
Acquired abnormalities of platelets are much more common tral nervous system bleeding or active bleeding from other sites
than acquired defects and may be quantitative or qualitative, occurs. Survival of the transfused platelets is usually short.
although some patients have both types of defects. Quantita- Primary immune thrombocytopenia is also known as
tive defects may be a result of failure of production, shortened idiopathic thrombocytopenic purpura (ITP). In children, it is
survival, or sequestration. Failure of production is generally a usually acute in onset, short lived, and typically follows a viral
result of bone marrow disorders such as leukemia, myelodys- illness. In contrast, ITP in adults is gradual in onset, chronic
plastic syndrome, severe vitamin B12 or folate deficiency, che- in nature, and has no identifiable cause. Because the circulat-
motherapeutic drugs, radiation, acute ethanol intoxication, or ing platelets in ITP are young and functional, bleeding is less
viral infection. If a quantitative abnormality exists and treatment for a given platelet count than when there is failure of platelet
is indicated either due to symptoms or the need for an invasive production. The pathophysiology of ITP is believed to involve
procedure, platelet transfusion is utilized. The etiologies of both both impaired platelet production and T cell–mediated plate-
qualitative and quantitative defects are reviewed in Table 4-1. let destruction.10 Management options are summarized in
Table 4-2.11 Treatment of drug-induced immune thrombocyto-
penia may simply entail withdrawal of the offending drug, but
Table 4-1 corticosteroids, gamma globulin, and anti-D immunoglobulin
Etiology of platelet disorders may hasten recovery of the count. Heparin-induced thrombo-
cytopenia (HIT) is a form of drug-induced immune thrombo-
A.  Quantitative Disorders
cytopenia. It is an immunologic event during which antibodies
1. Failure of production: related to impairment in bone
against platelet factor 4 (PF4) formed during exposure to hep-
marrow function
arin affect platelet activation and endothelial function with
a. Leukemia
resultant thrombocytopenia and intravascular thrombosis.12
b.  Myeloproliferative disorders
The platelet count typically begins to fall 5 to 7 days after
c. B12 or folate deficiencies
d.  Chemotherapy or radiation therapy
e.  Acute alcohol intoxication
f.  Viral infections
2.  Decreased survival Table 4-2
a. Immune-mediated Management of idiopathic thrombocytopenic purpura
1)  Idiopathic thrombocytopenia (ITP) (ITP) in adults
2)  Heparin-induced thrombocytopenia
First Line
3)  Autoimmune disorders or B-cell malignancies
  a. Corticosteroids: The majority of patients respond but
4)  Secondary thrombocytopenia
only a few long term
b.  Disseminated intravascular coagulation (DIC)
  b. Intravenous immunoglobulin (IVIG) or anti-D
c.  Related to platelet thrombi
immunoglobulin: indicated for clinical bleeding
1)  Thrombocytopenic purpura (TTP)
2)  Hemolytic uremic syndrome (HS) Second Line. Required in most patients
3. Sequestration   a. Splenectomy: open or laparoscopic. Criteria include
a.  Portal hypertension severe thrombocytopenia, high risk of bleeding, and
b. Sarcoid continued need for steroids. Failure may be due to
c. Lymphoma retained accessory splenic tissue.
d.  Gaucher’s Disease   b. Rituximab, an anti-CD 20 monoclonal antibody
B.  Qualitative Disorders   c. Thrombopoietin (TPO) receptor agonists such as
1. Massive transfusion romiplostim and eltrombopag
2. Therapeutic platelet inhibitors Third Line. To be used after failure of splenectomy and
3. Disease states rituximab
a.  Myeloproliferative disorders   a. TPO receptor agonists
b.  Monoclonal gammopathies   b. Immunosuppressive agents. For failure of TPO receptor
c.  Liver disease agonists

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
heparin has been started, but if it is a re-exposure, the decrease is frequently used, but it is not clear what etiologic factor is 91
in count may occur within 1 to 2 days. HIT should be suspected being removed by the pheresis.
if the platelet count falls to less than 100,000 or if it drops by Sequestration is another important cause of thrombocyto-
50% from baseline in a patient receiving heparin. While HIT penia and usually involves trapping of platelets in an enlarged

CHAPTER 4
is more common with full-dose unfractionated heparin (1%– spleen typically related to portal hypertension, sarcoid, lym-
3%), it can also occur with prophylactic doses or with low phoma, or Gaucher’s disease. The total body platelet mass is
molecular weight heparins. Interestingly, approximately 17% essentially normal in patients with hypersplenism, but a much
of patients receiving unfractionated heparin and 8% receiving larger fraction of the platelets are in the enlarged spleen. Platelet
low molecular weight heparin develop antibodies against PF4, survival is mildly decreased. Bleeding is less than anticipated
yet a much smaller percentage develop thrombocytopenia and from the count because sequestered platelets can be mobilized to

Hemostasis, Surgical Bleeding, and Transfusion


even fewer develop clinical HIT.13 In addition to the mild to some extent and enter the circulation. Platelet transfusion does
moderate thrombocytopenia, this disorder is characterized by not increase the platelet count as much as it would in a normal
a high incidence of thrombosis that may be arterial or venous. person because the transfused platelets are similarly sequestered
Importantly, the absence of thrombocytopenia in these patients in the spleen. Splenectomy is not indicated to correct the throm-
does not preclude the diagnosis of HIT. bocytopenia of hypersplenism caused by portal hypertension.
The diagnosis of HIT may be made by using either a sero- Thrombocytopenia is the most common abnormality of
tonin release assay (SRA) or an enzyme-linked immunosorbent hemostasis that results in bleeding in the surgical patient. The
assay (ELISA). The SRA is highly specific but not sensitive, patient may have a reduced platelet count as a result of a vari-
so a positive test supports the diagnosis but a negative test does ety of disease processes, as discussed earlier. In these circum-
not exclude HIT.12 On the other hand, the ELISA has a low stances, the marrow usually demonstrates a normal or increased
specificity, so although a positive ELISA confirms the presence number of megakaryocytes. By contrast, when thrombocytope-
of anti-heparin-PF4, it does not help in the diagnosis of clinical nia occurs in patients with leukemia or uremia and in patients
HIT. A negative ELISA, however, essentially rules out HIT. on cytotoxic therapy, there are generally a reduced number of
The initial treatment of suspected HIT is to stop heparin megakaryocytes in the marrow. Thrombocytopenia also occurs
and begin an alternative anticoagulant. Stopping heparin with- in surgical patients as a result of massive blood loss with prod-
out addition of another anticoagulant is not adequate to prevent uct replacement deficient in platelets. Thrombocytopenia may
thrombosis in this setting. Alternative anticoagulants are pri- also be induced by heparin administration during cardiac and
marily thrombin inhibitors. The most recent guideline by the vascular cases, as in the case of HIT, or may be associated with
American College of Chest Physicians recommends lepiru- thrombotic and hemorrhagic complications. When thrombocy-
din, argatroban, or danaparoid for patients with normal renal topenia is present in a patient for whom an elective operation
function and argatroban for patients with renal insufficiency.14 is being considered, management is contingent upon the extent
Because of warfarin’s early induction of a hypercoagulable and cause of platelet reduction. A count of greater than 50,000/μL
state, warfarin should be instituted only once full anticoagula- generally requires no specific therapy.
tion with an alternative agent has been accomplished and the Early platelet administration has now become part of mas-
platelet count has begun to recover. sive transfusion protocols.18,19 Platelets are also administered
These are also disorders in which thrombocytopenia is a preoperatively to rapidly increase the platelet count in surgical
result of platelet activation and formation of platelet thrombi. In patients with underlying thrombocytopenia. One unit of plate-
thrombotic thrombocytopenic purpura (TTP), large vWF mol- let concentrate contains approximately 5.5 × 1010 platelets and
ecules interact with platelets, leading to activation. These large would be expected to increase the circulating platelet count by
molecules result from inhibition of a metalloproteinase enzyme, about 10,000/μL in the average 70-kg person. Fever, infection,
ADAMtS13, which cleaves the large vWF molecules.15 TTP is hepatosplenomegaly, and the presence of antiplatelet alloan-
classically characterized by thrombocytopenia, microangio- tibodies decrease the effectiveness of platelet transfusions. In
pathic hemolytic anemia, fever, and renal and neurologic signs patients refractory to standard platelet transfusion, the use of
or symptoms. The finding of schistocytes on a peripheral blood human leukocyte antigen (HLA)-compatible platelets coupled
smear aids in the diagnosis. Plasma exchange with replacement with special processors has proved effective.
of FFP is the treatment for acute TTP.16 Additionally, rituximab,
a monoclonal antibody against the CD20 protein on B lympho- Qualitative Platelet Defects.  Impaired platelet function often
cytes, has shown promise as an immunomodulatory therapy accompanies thrombocytopenia but may also occur in the pres-
directed against patients with acquired TTP, of which the major- ence of a normal platelet count. The importance of this is obvi-
ity are autoimmune mediated.17 ous when one considers that 80% of overall strength is related
Hemolytic uremic syndrome (HUS) often occurs second- to platelet function. The life span of platelets ranges from 7 to
ary to infection by Escherichia coli 0157:H7 or other Shiga 10 days, placing them at increased risk for impairment by medi-
toxin-producing bacteria. The metalloproteinase is normal in cal disorders and prescription and over-the-counter medications.
these cases. HUS is usually associated with some degree of renal Impairment of ADP-stimulated aggregation occurs with
failure, with many patients requiring renal replacement therapy. 1 massive transfusion of blood products. Uremia may be
Neurologic symptoms are less frequent. A number of patients associated with increased bleeding time and impaired aggrega-
develop features of both TTP and HUS. This may occur with tion. Defective aggregation and platelet dysfunction are also
autoimmune diseases, especially systemic lupus erythematosus seen in patients with thrombocythemia, polycythemia vera, and
and HIV infection, or in association with certain drugs (such as myelofibrosis.
ticlopidine, mitomycin C, gemcitabine) or immunosuppressive Drugs that interfere with platelet function include aspirin,
agents (such as cyclosporine and tacrolimus). Discontinuation clopidogrel, prasugrel, dipyridamole, and GP IIb/IIIa inhibi-
of the involved drug is the mainstay of therapy. Plasmapheresis tors. Aspirin, clopidogrel, and prasugrel all irreversibly inhibit

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
92 platelet function. Clopidogrel and prasugrel do so through selec- correlation between an increasing DIC score and mortality, espe-
tive irreversible inhibition of ADP-induced platelet aggrega- cially in patients with infections.26
tion.20 Aspirin works through irreversible acetylation of platelet The most important facets of treatment are relieving the
prostaglandin synthase. patient’s causative primary medical or surgical problem and
There are no prospective randomized trials in general sur- maintaining adequate perfusion. If there is active bleeding,
PART I

gical patients to guide the timing of surgery in patients on aspi- hemostatic factors should be replaced with FFP, which is usu-
rin, clopidogrel, or prasugrel.21 The general recommendation is ally sufficient to correct the hypofibrinogenemia, although
that approximately 5 to 7 days should pass from the time the cryoprecipitate, fibrinogen concentrates, or platelet concentrates
drug is stopped until an elective procedure is performed.22 Tim- may also be needed. Given the formation of microthrombi
ing of urgent and emergent surgeries is even more unclear. Pre- in DIC, heparin therapy has also been proposed. Most stud-
BASIC CONSIDERATIONS

operative platelet transfusions may be beneficial, but there are ies, however, have shown that heparin is not helpful in acute
no good data to guide their administration. However, new func- forms of DIC, but may be indicated in cases where thrombosis
tional tests are becoming available that may better demonstrate predominates, such as arterial or venous thromboembolism and
defects in platelet function and may serve to guide the timing severe purpura fulminans.
of operation or when platelet transfusions might be indicated. Primary Fibrinolysis.  An acquired hypofibrinogenic state in
Other disorders associated with abnormal platelet func- the surgical patient can be a result of pathologic fibrinolysis.
tion include uremia, myeloproliferative disorders, monoclonal This may occur in patients following prostate resection when
gammopathies, and liver disease. In the surgical patient, plate- urokinase is released during surgical manipulation of the pros-
let dysfunction of uremia can often be corrected by dialysis or tate or in patients undergoing extracorporeal bypass. The sever-
the administration of DDAVP. Platelet transfusion may not be ity of fibrinolytic bleeding is dependent on the concentration of
helpful if the patient is uremic when the platelets are given and breakdown products in the circulation. Antifibrinolytic agents,
only serve to increase antibodies. Platelet dysfunction in myelo- such as ε-aminocaproic acid and tranexamic acid, interfere with
proliferative disorders is intrinsic to the platelets and usually fibrinolysis by inhibiting plasminogen activation.
improves if the platelet count can be reduced to normal with
chemotherapy. If possible, surgery should be delayed until the Myeloproliferative Diseases
count has been decreased. These patients are at risk for both Polycythemia, or an excess of red blood cells, places surgical
bleeding and thrombosis. Platelet dysfunction in patients with patients at risk. Spontaneous thrombosis is a complication of
monoclonal gammopathies is a result of interaction of the mono- polycythemia vera, a myeloproliferative neoplasm, and can be
clonal protein with platelets. Treatment with chemotherapy or, explained in part by increased blood viscosity, increased plate-
occasionally, plasmapheresis to lower the amount of monoclo- let count, and an increased tendency toward stasis. Paradoxi-
nal protein improves hemostasis. cally, a significant tendency toward spontaneous hemorrhage
Acquired Hypofibrinogenemia also is noted in these patients. Thrombocytosis can be reduced
by the administration of low-dose aspirin, phlebotomy, and
Disseminated Intravascular Coagulation (DIC).  DIC is hydroxyurea.27
an acquired syndrome characterized by systemic activation of
coagulation pathways that result in excessive thrombin genera- Coagulopathy of Liver Disease
tion and the diffuse formation of microthrombi. This disturbance The liver plays a key role in hemostasis because it is responsible
ultimately leads to consumption and depletion of platelets and for the synthesis of many of the coagulation factors (Table 4-3).
coagulation factors with the resultant classic picture of diffuse Patients with liver disease, therefore, have decreased production
bleeding. Fibrin thrombi developing in the microcirculation may of several key non-endothelial cell-derived coagulation factors
cause microvascular ischemia and subsequent end-organ failure as well as natural anticoagulant proteins, causing a disturbance
if severe. There are many different conditions that predispose in the balance between procoagulant and anticoagulant path-
a patient to DIC, and the presence of an underlying condition ways. This disturbance in coagulation mechanisms causes a
is required for the diagnosis. For example, injuries resulting in complex paradigm of both increased bleeding risk and increased
embolization of materials such as brain matter, bone marrow, or thrombotic risk. The most common coagulation abnormalities
amniotic fluid can act as potent thromboplastins that activate the
DIC cascade.23 Additional etiologies include malignancy, organ
injury (such as severe pancreatitis), liver failure, certain vascu-
lar abnormalities (such as large aneurysms), snake bites, illicit Table 4-3
drugs, transfusion reactions, transplant rejection, and sepsis.24 In
Coagulation factors synthesized by the liver
fact, DIC frequently accompanies sepsis and may be associated
with multiple organ failure. As of yet, scoring systems for organ Vitamin K–dependent factors: II (prothrombin factor), VII,
failure do not routinely incorporate DIC. The important interplay IX, X
between sepsis and coagulation abnormalities was demonstrated Fibrinogen
by Dhainaut et al who showed that activated protein C was effec- Factor V
tive in septic patients with DIC.25 The diagnosis of DIC is made
Factor VIII
based on an inciting etiology with associated thrombocytopenia,
prolongation of the prothrombin time, a low fibrinogen level, and Factors XI, XII, XIII
elevated fibrin markers (FDPs, D-dimer, soluble fibrin mono- Antithrombin III
mers). A scoring system developed by the International Society Plasminogen
for Thrombosis and Hemostasis has been shown to have high
Protein C and protein S
sensitivity and specificity for diagnosing DIC as well as a strong

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
associated with liver dysfunction are thrombocytopenia and mixed following insertion of a transjugular intrahepatic porto- 93
impaired humoral coagulation function manifested as prolonga- systemic shunt (TIPS). Therefore, treatment of thrombocytope-
tion of the prothrombin time and international normalized ratio nia should not be the primary indication for a TIPS procedure.
(INR). The etiology of thrombocytopenia in patients with liver Decreased production or increased destruction of coagula-

CHAPTER 4
disease is typically related to hypersplenism, reduced produc- tion factors as well as vitamin K deficiency can all contribute
tion of thrombopoietin, and immune-mediated destruction of to a prolonged PT and INR in patients with liver disease. As
platelets. The total body platelet mass is often normal in patients liver dysfunction worsens, so does the liver’s synthetic func-
with hypersplenism, but a much larger fraction of the platelets tion, which results in decreased production of coagulation fac-
is sequestered in the enlarged spleen. Bleeding may be less than tors. Additionally, laboratory abnormalities may mimic those of
anticipated because sequestered platelets can be mobilized to DIC. Elevated D-dimers have been reported to increase the risk

Hemostasis, Surgical Bleeding, and Transfusion


some extent and enter the circulation. Thrombopoietin, the pri- of variceal bleeding. The absorption of vitamin K is dependent
mary stimulus for thrombopoiesis, may be responsible for some on bile production. Therefore, liver patients with impaired bile
cases of thrombocytopenia in cirrhotic patients, although its role production and cholestatic disease may be at risk for vitamin K
is not well delineated. Finally, immune-mediated thrombocyto- deficiency.
penia may also occur in cirrhotics, especially those with hepatitis Similar to thrombocytopenia, correction of coagulopathy
C and primary biliary cirrhosis.28 In addition to thrombocytope- should be reserved for treatment of active bleeding and prophy-
nia, these patients also exhibit platelet dysfunction via defective laxis for invasive procedures and surgery. Treatment of coagu-
interactions between platelets and the endothelium, and possibly lopathy caused by liver disease is usually done with FFP, but
due to uremia and changes in endothelial function in the setting because the coagulopathy is usually not a result of decreased
of concomitant renal insufficiency. Hypocoagulopathy is fur- levels of factor V, complete correction is not usually possible.
ther exacerbated with low platelet counts because platelets help If the fibrinogen is less than 200 mg/dL, administration of cryo-
facilitate thrombin generation by assembling coagulation factors precipitate may be helpful. Cryoprecipitate is also a source of
on their surfaces. In conditions mimicking intravascular flow, factor VIII for the rare patient with a low factor VIII level.
low hematocrit and low platelet counts contributed to decreased
adhesion of platelets to endothelial cells, although increased
Coagulopathy of Trauma
Traditional teaching regarding trauma-related coagulopathy
vWF, a common finding in cirrhotic patients, may offset this
attributed its development to acidosis, hypothermia, and dilution
change in patients with cirrhosis.29 Hypercoagulability of liver
of coagulation factors. Recent data, however, have shown that
disease has recently gained increased attention, with more evi-
over one third of injured patients have evidence of coagulopa-
dence demonstrating the increased incidence of thromboem-
thy at the time of admission.36 More importantly, patients
bolism despite thrombocytopenia and a hypocoagulable state 2 arriving with coagulopathy are at a significantly higher
on conventional blood tests.30,31 This is attributed to decreased
risk of mortality, especially in the first 24 hours after injury. In
production of liver-synthesized proteins C and S, antithrombin,
light of these findings, a dramatic increase in research focused
and plasminogen levels, as well as elevated levels of endothe-
on the optimal management of the acute coagulopathy of trauma
lial-derived vWF and factor VIII, a potent driver of thrombin
(ACoT) has been observed over the past several years. ACoT
generation.32,33 Given the concomitant hypo- and hypercoagu-
is not a simple dilutional coagulopathy but a complex problem
lable features seen in patients with liver disease, conventional
with multiple mechanisms.37 Whereas multiple contributing fac-
coagulation tests may be difficult to interpret, and alternative
tors exist, the key initiators to the process of ACoT are shock
tests such as thromboelastography (TEG) may be more informa-
and tissue injury. ACoT is a separate and distinct process from
tive of the functional status of clot formation and stability in cir-
DIC, with its own specific components of hemostatic failure.
rhotic patients. Several studies imply that TEG provides a better
Brohi et al have demonstrated that only patients in shock arrive
assessment of bleeding risk than standard tests of hemostasis in
coagulopathic and that it is the shock that induces coagulopathy
patients with liver disease; however, no studies have directly
through systemic activation of anticoagulant and fibrinolytic
tested this, and future prospective trials are needed.34
pathways.38 As shown in Fig. 4-5, hypoperfusion causes activa-
Before instituting any therapy to ameliorate thrombocyto-
tion of TM on the surface of endothelial cells. Thrombin-TM
penia, the actual need for correction should be strongly consid-
complexes induce an anticoagulant state through activation of
ered. In general, correction based solely on a low platelet count
protein C and enhancement of fibrinolysis. This same complex
should be discouraged. Most often, treatment should be with-
also limits the availability of thrombin to cleave fibrinogen to
held for invasive procedures and surgery. Platelet transfusions
fibrin, which may explain why injured patients rarely have low
are the mainstay of therapy; however, the effect typically lasts
levels of fibrinogen.
only several hours. Risks associated with transfusions in gen-
eral and the development of antiplatelet antibodies in a patient Acquired Coagulation Inhibitors
population likely to need recurrent correction should be consid- Among the most common acquired coagulation inhibitors is the
ered. A potential alternative strategy involves administration of antiphospholipid syndrome (APLS), which includes the lupus
interleukin-11 (IL-11), a cytokine that stimulates proliferation anticoagulant and anticardiolipin antibodies. These antibodies
of hematopoietic stem cells and megakaryocyte progenitors.26 may be associated with either venous or arterial thrombosis,
Most studies using IL-11 have been in cancer patients, although or both. In fact, patients presenting with recurrent thrombosis
some evidence exists that it may be beneficial in cirrhotics as should be evaluated for APLS. Antiphospholipid antibodies are
well. Significant side effects limit its usefulness.35 A less very common in patients with systemic lupus but may also be
well-accepted option is splenectomy or splenic embolization to seen in association with rheumatoid arthritis and Sjögren’s syn-
reduce hypersplenism. In addition to the risks associated with drome. There are also individuals who will have no autoimmune
these techniques, reduced splenic blood flow can reduce portal disorders but develop transient antibodies in response to infections
vein flow with subsequent portal vein thrombosis. Results are or those who develop drug-induced APLS. The hallmark of

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
94 Central role of thrombomodulin in Table 4-4
acute traumatic coagulopathy (ATC)
Medications that can alter warfarin dosing
Shock ↓ warfarin effect Barbiturates, oral contraceptives,
↑ warfarin requirements estrogen-containing compounds,
PART I

corticosteroids, adrenocorticotropic
Hypoperfusion
hormone
Thrombin Maintains ↑ warfarin effect Phenylbutazone, clofibrate,
↑ Thrombomodulin fibrinogen ↓ warfarin requirements anabolic steroids, L-thyroxine,
level
BASIC CONSIDERATIONS

glucagons, amiodarone, quinidine,


Thrombomodulin/ cephalosporins
Thrombin complex

Activation PAI-1
can be dramatically reduced. There are several reversal options
ActivationTAFI that include vitamin K administration, plasma, cryoprecipitate,
of Consumption
protein C recombinant factor VIIa, and factor concentrates. Urgent rever-
sal for life-threatening bleeding should include vitamin K and
a rapid reversal agent such as plasma or prothrombin complex
Fibrinolysis concentrate. In the elderly or those with intracranial hemor-
ATC
rhage, concentrates are preferred, whereas in situations with
hypovolemia from hemorrhage, plasma should be used.
Figure 4-5. Illustration of the pathophysiologic mechanism
Newer anticoagulants like dabigatran and rivaroxaban
responsible for the acute coagulopathy of trauma. PAI-1 = plas- have no readily available method of detection of the degree of
minogen activator inhibitor 1; TAFI = thrombin-activatable fibri- anticoagulation. More concerning is the absence of any
nolysis inhibitor. 3 available reversal agent. Unlike warfarin, the nonrevers-
ible coagulopathy associated with dabigatran and rivaroxaban
is of great concern to those providing emergent care to these
APLS is a prolonged aPTT in vitro but an increased risk of patients.39
thrombosis in vivo. The only possible strategy to reverse the coagulopathy
associated with dabigatran may be emergent dialysis. Unfor-
Anticoagulation and Bleeding tunately, the ability to rapidly dialyze the hemodynamically
Spontaneous bleeding can be a complication of any antico- unstable bleeding patient or rapidly dialyze the anticoagulated
agulant therapy whether it is heparin, low molecular weight patient with an intracranial bleed is challenging even at large
heparins, warfarin, factor Xa inhibitors, or new direct thrombin medical centers. Recent data suggest that rivaroxaban, however,
inhibitors. The risk of spontaneous bleeding related to heparin may be reversed with the use of prothrombin complex concen-
is reduced with a continuous infusion technique. Therapeutic trates (four-factor concentrates only: II, VII, IX, and X).40 In
anticoagulation is more reliably achieved with a low molecu- less urgent states, these drugs can be held for 36 to 48 hours
lar weight heparin. However, laboratory testing is more chal- prior to surgery without increased risk of bleeding in those with
lenging with these medications, as they are not detected with normal renal function. Alternatively, activated clotting time
conventional coagulation testing. However, their more reliable (stand alone or with rapid TEG) or ecarin clotting time can be
therapeutic levels (compared to heparin) make them an attrac- obtained in those on dabigatran, and anti-factor Xa assays can
tive option for outpatient anticoagulation and more cost-effec- be obtained in those taking rivaroxaban.
tive for the inpatient setting. If monitoring is required (e.g., in Bleeding complications in patients on anticoagulants
the presence of renal insufficiency or severe obesity), the drug include hematuria, soft tissue bleeding, intracerebral bleeding,
effect should be determined with an assay for anti-Xa activity. skin necrosis, and abdominal bleeding. Bleeding secondary to
Warfarin is used for long-term anticoagulation in various anticoagulation therapy is also not an uncommon cause of a
clinical conditions including deep vein thrombosis, pulmonary rectus sheath hematomas. In most of these cases, reversal of
embolism, valvular heart disease, atrial fibrillation, recurrent anticoagulation is the only treatment that is necessary. Lastly, it
systemic emboli, recurrent myocardial infarction, prosthetic is important to remember that symptoms of an underlying tumor
heart valves, and prosthetic implants. Due to the interaction may first present with bleeding while on anticoagulation.
of the P450 system, the anticoagulant effect of the warfarin Surgical intervention may prove necessary in patients
is reduced (e.g., increases dose required) in patients receiv- receiving anticoagulation therapy. Increasing experience suggests
ing barbiturates as well as in patients with diets low in vita- that surgical treatment can be undertaken without full reversal of
min K. Increased warfarin requirements may also be needed the anticoagulant, depending on the procedure being performed.41
in patients taking contraceptives or estrogen-containing com- When the aPTT is less than 1.3 times control in a heparinized
pounds, corticosteroids, and adrenocorticotropic hormone patient or when the INR is less than 1.5 in a patient on warfarin,
(ACTH). Medications that can alter warfarin requirements are reversal of anticoagulation therapy may not be necessary. How-
shown in Table 4-4. ever, meticulous surgical technique is mandatory, and the patient
Although warfarin use is often associated with a significant must be observed closely throughout the postoperative period.
increase in morbidity and mortality in acutely injured and emer- Certain surgical procedures should not be performed in
gency surgery patients, with rapid reversal, these complications concert with anticoagulation. In particular, cases where even

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
minor bleeding can cause great morbidity, such as the central accepted transfusion thresholds. Platelet concentrates are given 95
nervous system and the eye, surgery should be avoided. Emergency for bleeding patients in the immediate postoperative period;
operations are occasionally necessary in patients who have been however, studies have shown that indiscriminate platelet ther-
heparinized. The first step in these patients is to discontinue apy conferred no therapeutic advantage.46 It is in these patients

CHAPTER 4
heparin. For more rapid reversal, protamine sulfate is effective. where rapid coagulation testing is required to assist with directed
However, significant adverse reactions, especially in patients transfusion therapy.47 Many institutions now use antifibrinolyt-
with severe fish allergies, may be encountered when administer- ics, such as ε-aminocaproic acid and tranexamic acid, at the time
ing protamine.42 Symptoms include hypotension, flushing, bra- of anesthesia induction after several studies have shown that
dycardia, nausea, and vomiting. Prolongation of the aPTT after such treatment reduced postoperative bleeding and reoperation.
heparin neutralization with protamine may also be a result of the Aprotinin, a protease inhibitor that acts as an antifibrinolytic

Hemostasis, Surgical Bleeding, and Transfusion


anticoagulant effect of protamine. In the elective surgical patient agent, has been shown to reduce transfusion requirements asso-
who is receiving coumarin-derivative therapy sufficient to effect ciated with cardiac surgery.48 Desmopressin acetate stimulates
anticoagulation, the drug can be discontinued several days before release of factor VIII from endothelial cells and may also be
operation and the prothrombin concentration then checked (a effective in reducing blood loss during cardiac surgery. The use
level >50% is considered safe).43 Rapid reversal of anticoagula- of recombinant factor VIIa has also been studied but with con-
tion can be accomplished with plasma or prothrombin complex flicting results between improved hemostasis and thrombotic
concentrates in the emergent situation. Parenteral administration events and mortality, and thus its use is often employed only as
of vitamin K also is indicated in elective surgical treatment of a measure of last resort.45,49
patients with biliary obstruction or malabsorption who may be Local Hemostasis.  Significant surgical bleeding is usually
vitamin K deficient. However, if low levels of factors II, VII, IX, caused by ineffective local hemostasis. The goal is therefore
and X (vitamin K–dependent factors) exist as a result of hepa- to prevent further blood loss from a disrupted vessel that has
tocellular dysfunction, vitamin K administration is ineffective. been incised or transected. Hemostasis may be accomplished by
The perioperative management of patients receiving long- interrupting the flow of blood to the involved area or by direct
term oral anticoagulation therapy is an increasingly common closure of the blood vessel wall defect.
problem. Definitive evidence-based guidelines regarding
4 which patients require perioperative “bridging” anticoagu- Mechanical Procedures.  The oldest mechanical method of
lation and the most effective way to bridge are lacking. However, bleeding cessation is application of direct digital pressure, either
the American College of Chest Physicians Evidence-Based Clin- at the site of bleeding or proximally to permit more definitive
ical Practice Guidelines do serve as best practice for these situa- action. An extremity tourniquet that occludes a major vessel
tions.44 A few clinical scenarios exist where the patient should be proximal to the bleeding site or the Pringle maneuver for liver
transitioned to intravenous heparin from oral anticoagulants. A bleeding are good examples. Direct digital pressure is very
heparin infusion should be held for 4 to 6 hours before the proce- effective and has the advantage of being less traumatic than
dure and restarted within 12 to 24 hours of the end of its comple- hemostatic or even “atraumatic” clamps.
tion. The primary indication for this level of aggressiveness is When a small vessel is transected, a simple ligature is usu-
patients with mechanical heart valves. Other indications include ally sufficient. However, for larger pulsating arteries, a transfix-
a recent (within 30 days) myocardial infarction, stroke, or pul- ion suture to prevent slipping is indicated. All sutures represent
monary embolism. Situations such as thromboembolic events foreign material, and selection should be based on their intrinsic
greater than 30 days prior, hypercoagulable history, and atrial characteristics and the state of the wound. Direct pressure applied
fibrillation do not require such stringent restarting strategies. by “packing” a wound with gauze or laparotomy pads affords
the best method of controlling diffuse bleeding from large areas,
Cardiopulmonary Bypass.  Under normal conditions, homeo-
such as in the trauma situation. Packing bone wax on the raw
stasis of the coagulation system is maintained by complex inter-
surface to effect pressure can control bleeding from cut bone.
actions between the endothelium, platelets, and coagulation
factors. In patients undergoing cardiopulmonary bypass (CPB), Thermal Agents.  Heat achieves hemostasis by denaturation of
contact with circuit tubing and membranes results in abnormal protein that results in coagulation of large areas of tissue. Elec-
platelet and clotting factor activation, as well as activation of trocautery generates heat by induction from an alternating cur-
inflammatory cascades, that ultimately result in excessive fibri- rent source, which is then transmitted via conduction from the
nolysis and a combination of both quantitative and qualitative instrument directly to the tissue. The amplitude setting should
platelet defects. Platelets undergo reversible alterations in mor- be high enough to produce prompt coagulation, but not so high
phology and their ability to aggregate, which causes sequestra- as to set up an arc between the tissue and the cautery tip. This
tion in the filter, partially degranulated platelets, and platelet avoids thermal injury outside of the operative field and also pre-
fragments. This multifactorial coagulopathy is compounded by vents exit of current through electrocardiographic leads, other
the effects of shear stress in the system, induced hypothermia, monitoring devices, or permanent pacemakers or defibrillators.
hemodilution, and anticoagulation.45 A negative grounding plate should be placed beneath the patient
While on pump, activated clotting time measurements are to avoid severe skin burns, and caution should be used with cer-
obtained along with blood gas measurements; however, con- tain anesthetic agents (diethyl ether, divinyl ether, ethyl chloride,
ventional coagulation assays and platelet counts are not nor- ethylene, and cyclopropane) because of the hazard of explosion.
mally performed until rewarming and after a standard dose of A direct current also can result in hemostasis. Because the
protamine has been given. TEG may give a better estimate of protein moieties and cellular elements of blood have a nega-
the extent of coagulopathy and may also be used to anticipate tive surface charge, they are attracted to a positive pole where a
transfusion requirements if bleeding is present.45 thrombus is formed. Direct currents in the 20- to 100-mA range
Empiric treatment with FFP and cryoprecipitate is often have successfully controlled diffuse bleeding from raw surfaces,
used for bleeding patients; however, there are no universally as has argon gas.

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
96 Topical Hemostatic Agents.  Topical hemostatic agents can be transfused only with Rh-negative blood. However, this group
play an important role in helping to facilitate surgical hemo- represents only 15% of the population. Therefore, the adminis-
stasis. These agents are classified based on their mechanism tration of Rh-positive blood is acceptable if Rh-negative blood
of action, and many act at specific stages in the coagulation is not available. However, Rh-positive blood should not be
cascade and take advantage of natural physiologic responses to transfused to Rh-negative females who are of child-bearing age.
PART I

bleeding.50 The ideal topical hemostatic agent has significant In emergency situations, type O-negative blood may be
hemostatic action, minimal tissue reactivity, nonantigenicity, in transfused to all recipients. O-negative and type-specific red
vivo biodegradability, ease of sterilization, low cost, and can be blood cells are equally safe for emergency transfusion. Prob-
tailored to specific needs.51 lems are associated with the administration of four or more
In 2010, Achneck et al published a comprehensive over- units of O-negative blood because there is a significant increase
BASIC CONSIDERATIONS

view of absorbable, biologic, and synthetic agents.52 Absorb- in the risk of hemolysis. In patients with clinically significant
able agents include gelatin foams (Gelfoam), oxidized cellulose cold agglutinins, blood should be administered through a blood
(Surgicel), and microfibrillar collagens (Avitene). Both gelatin warmer. If these antibodies are present in high titer, hypother-
foam and oxidized cellulose provide a physical matrix for clot- mia is contraindicated.
ting initiation, while microfibrillar collagens facilitate plate- In patients who have been multiply transfused and who
let adherence and activation. Biologic agents include topical have developed alloantibodies or who have autoimmune hemo-
thrombin, fibrin sealants (FloSeal), and platelet sealants (Vita- lytic anemia with pan-red blood cell antibodies, typing and
gel). Human or recombinant thrombin derivatives, which facili- cross-matching is often difficult, and sufficient time should
tate the formation of fibrin clots and subsequent activation of be allotted preoperatively to accumulate blood that might be
several clotting factors, take advantage of natural physiologic required during the operation. Cross-matching should always
processes, thereby avoiding foreign body or inflammatory be performed before the administration of dextran because it
reactions.51 Caution must be taken in judging vessel caliber in interferes with the typing procedure.
the wound because thrombin entry into larger caliber vessels The use of autologous transfusion is growing. Up to 5 units
can result in systemic exposure to thrombin with a risk of dis- can be collected for subsequent use during elective procedures.
seminated intravascular clotting or death. They are particularly Patients can donate and store their own blood if their hemoglo-
effective in controlling capillary bed bleeding when pressure or bin concentration exceeds 11 g/dL or if the hematocrit is greater
ligation is insufficient; however, the bovine derivatives should than 34%. The first procurement is performed 40 days before
be used with caution due to the potential immunologic response the planned operation, and the last one is performed 3 days
and worsened coagulopathy. Fibrin sealants are prepared from before the operation. Donations can be scheduled at intervals
cryoprecipitate (homologous or synthetic) and have the advan- of 3 to 4 days. Recombinant human erythropoietin (rHuEPO)
tage of not promoting inflammation or tissue necrosis.53 Platelet accelerates generation of red blood cells and allows for more
sealants are a mixture of collagen and thrombin combined with frequent harvesting of blood.
plasma-derived fibrinogen and platelets from the patient, which Banked Whole Blood. Once the gold standard, whole
requires the additional need for centrifugation and processing. blood is rarely available in Western countries. With sequen-
Topical agents are not a substitute for meticulous surgical tial changes in storage solutions, the shelf life of red blood
technique and only function as adjuncts to help facilitate surgi- cells is now 42 days. Recent evidence has demonstrated that
cal hemostasis. The advantages and disadvantages of each agent the age of red cells may play a significant role in the inflam-
must be considered, and use should be limited to the minimum matory response and incidence of multiple organ failure.54 The
amount necessary to minimize toxicity, adverse reactions, inter- changes in the red blood cells that occur during storage include
ference with wound healing, and procedural costs. reduction of intracellular ADP and 2,3-diphosphoglycerate (2,3-
DPG), which alters the oxygen dissociation curve of hemoglo-
TRANSFUSION bin, resulting in a decrease in oxygen transport. Stored RBCs
progressively becomes acidodic with elevated levels of lactate,
Background potassium, and ammonia.
Human blood replacement therapy was accepted in the late nine-
Red Blood Cells and Frozen Red Blood Cells.  Red blood
teenth century. This was followed by the introduction of blood
cells are the product of choice for most clinical situations requir-
grouping by Landsteiner who identified the major A, B, and
ing resuscitation. Concentrated suspensions of red blood cells
O groups in 1900, resulting in widespread use of blood prod-
can be prepared by removing most of the supernatant plasma
ucts in World War I. Levine and Stetson in 1939 followed with
after centrifugation. The preparation reduces but does not elimi-
the concept of Rh grouping. These breakthroughs established
nate reactions caused by plasma components. Frozen red blood
the foundation from which the field of transfusion medicine has
cells are not currently available for use in emergencies, as the
grown. Whole blood was considered the standard in transfu-
thawing and preparation time is measured in hours. They are
sion until the late 1970s when component therapy began to take
used for patients who are known to have been previously sensi-
prominence. This change in practice was made possible by the
tized. The red blood cell viability is improved, and the ATP and
development of improved collection strategies, infectious dis-
2,3-DPG concentrations are maintained.
ease testing, and advances in preservative solutions and storage.
Leukocyte-Reduced and Leukocyte-Reduced/Washed Red
Replacement Therapy Blood Cells.  These products are prepared by filtration that
Typing and Cross-Matching.  Serologic compatibility for A, removes about 99.9% of the white blood cells and most of the
B, O, and Rh groups is established routinely. Cross-matching platelets (leukocyte-reduced red blood cells) and, if necessary,
between the donors’ red blood cells and the recipients’ sera (the by additional saline washing (leukocyte-reduced/washed red
major cross-match) is performed. Rh-negative recipients should blood cells). Leukocyte reduction prevents almost all febrile,

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
nonhemolytic transfusion reactions (fever and/or rigors), allo- hemorrhage, and the majority died on the day of randomiza- 97
immunization to HLA class I antigens, and platelet transfusion tion. The authors reported that TXA use resulted in a statisti-
refractoriness and cytomegalovirus transmission. In most cally significant reduction in the relative risk (RR) of all-cause
Western nations, it is the standard red blood cell transfusion mortality of 9% (14.5 vs. 16.0%, RR 0.91, confidence interval

CHAPTER 4
product. Supporters of universal leukocyte reduction argue that [CI] 0.85–0.97; P = .0035). A recent post hoc analysis of the
allogenic transfusion of white cells predisposes to postoperative CRASH-2 data showed that the greatest benefit of TXA admin-
bacterial infection and multiorgan failure. Reviews of random- istration occurred when patients received the medication soon
ized trials and meta-analyses have not provided convincing evi- after injury.61 In this analysis, TXA given between 1 and 3 hours
dence either way,55,56 although a large Canadian retrospective after trauma reduced the risk of death due to bleeding by 21%
study suggests a decrease in mortality and infections.57 (147/3037 [4.8%] vs. 184/2996 [6.1%], RR 0.79, CI 0.64–0.97;

Hemostasis, Surgical Bleeding, and Transfusion


Platelet Concentrates.  The indications for platelet transfu- P = .03). Treatment given after 3 hours increased the risk of
sion include thrombocytopenia caused by massive blood loss death due to bleeding (144/3272 [4.4%] vs. 103/3362 [3.1%],
and replacement with platelet-poor products, thrombocytopenia RR 1.44, CI 1.12–1.84; P = .004). Finally, a recent meta-analysis
caused by inadequate production, and qualitative platelet disor- reported that TXA is effective for preventing blood loss in
ders. The shelf life of platelets is 120 hours from time of donation. surgery and reducing transfusion and was not associated with
One unit of platelet concentrate has a volume of approximately increased vascular occlusive events.62
50 mL. Platelet preparations are capable of transmitting infectious Adverse events associated with TXA use have been
diseases and can account for allergic reactions similar to those reported. These include acute gastrointestinal disturbances
caused by red blood cell transfusion. A therapeutic level of plate- (nausea, vomiting, and diarrhea, generally dose-related), visual
lets is in the range of 50,000 to 100,000/μL but is very dependent disturbances (blurry vision and changes in color perception,
on the clinical situation. Recent evidence suggests that earlier use especially with prolonged use), and occasional thromboem-
of platelets may improve outcomes in bleeding patients.58 bolic events (e.g., deep venous thrombosis and pulmonary
In rare cases, in patients who become alloimmunized embolism, generally observed in the setting of active intravas-
through previous transfusion or patients who are refractory from cular clotting). Its use is thus contraindicated in the settings of
sensitization through prior pregnancies, HLA-matched platelets acquired defective color vision and active intravascular clot-
can be used. ting. TXA should be used with caution in the setting of urinary
tract bleeding because ureteral obstruction due to clotting has
Fresh Frozen Plasma.  Fresh frozen plasma (FFP) prepared been reported. TXA is contraindicated in patients with aneu-
from freshly donated blood is the usual source of the vitamin rysmal subarachnoid hemorrhage; however, there have been
K-dependent factors and is the only source of factor V. FFP car- no reported complications associated with intra- or extracranial
ries similar infectious risks as other component therapies. Use of hemorrhage associated with trauma. TXA should not be given
plasma as a primary resuscitation modality in patients who are with activated prothrombin complex concentrate or factor IX
rapidly bleeding has received attention over the last few years, complex concentrates because these may increase the risk of
and ongoing studies are under way to evaluate this concept. FFP thrombosis.
can be thawed and stored for up to 5 days, greatly increasing TXA is an antifibrinolytic that inhibits both plasminogen
its immediate availability. In an effort to increase the shelf life activation and plasmin activity, thus preventing clot breakdown
and avoid the need for refrigeration, lyophilized plasma is being rather than promoting new clot formation. TXA is an inhibitor
tested. Preliminary animal studies suggest that it preserves the of plasminogen activation and an inhibitor of plasmin activity.
beneficial effects of FFP.59 It occupies the lysine binding sites on plasminogen, thus pre-
Concentrates and Recombinant DNA Technology.  Techno- venting its binding to lysine residues on fibrin. This reduces
logic advancements have made the majority of clotting factors plasminogen activation to plasmin. Similarly, blockade of
and albumin readily available as concentrates. These products lysine-binding sites on circulating plasmin prevents binding
are readily available and carry none of the inherent infectious to fibrin and thus prevents clot breakdown. TXA is 10 times
risks as other component therapies. more potent in vitro than aminocaproic acid. At therapeutically
Tranexamic Acid.  Tranexamic acid (TXA; trade name: Cyk- relevant concentrations, TXA does not affect platelet count or
lokapron), an antifibrinolytic agent, has been used to decrease aggregation or coagulation parameters. It is excreted largely
bleeding and the need for blood transfusions in coronary artery unchanged in urine and has a half-life of about 2 hours in cir-
bypass grafting (CABG), orthotopic liver transplantation, hip culation. While prolonged use requires that dosing be adjusted
and knee arthroplasty, and other surgical settings. The safety for renal impairment, use in the acute trauma situation does
and efficacy of using TXA to treat trauma patients was recently not appear to require adjustment. No adjustment is needed
evaluated in a large randomized, placebo-controlled clinical for hepatic impairment. Based on the CRASH-2 trial, TXA is
trial.60 In this trial, 20,211 adult trauma patients in 274 hospitals becoming more widely used in the United States for patients
in 40 countries with significant hemorrhage (heart rate >110 with ongoing bleeding, especially those with documented evi-
beats per minute and systolic blood pressure <90 mmHg or dence of fibrinolysis. Careful analysis of recently ongoing trials
both) or judged to be at risk for significant hemorrhage were will further elucidate the safety profile of this powerful drug.63
randomized to either TXA or placebo administered as a load-
Indications for Replacement of
ing dose of 1 g over 10 minutes followed by an infusion of 1 g
over 8 hours. It is important to understand that the responsible Blood and Its Elements
physician did not randomize patients with either a clear indica- Improvement in Oxygen-Carrying Capacity.  Oxygen-
tion or a clear contraindication to TXA. The overall mortality carrying capacity is primarily a function of the red blood cells.
rate in the cohort studied was 15.3%, of whom 35.3% died on Thus, transfusion of red blood cells should augment oxygen-
the day of randomization. A total of 1063 patients died due to carrying capacity. Additionally, hemoglobin is fundamental to

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
98 arterial oxygen content and thus oxygen delivery. Despite this are nonpreventable.70 Patients who survive to an emergency
obvious association, there is little evidence that actually sup- center have a high incidence of truncal hemorrhage, and deaths
ports the premise that transfusion of red blood cells equates with in this group of patients may be potentially preventable. Trun-
enhanced cellular delivery and utilization. The reasons for this cal hemorrhage patients in shock often present with the early
apparent discrepancy are related to changes that occur with stor- coagulopathy of trauma in the emergency department and are at
PART I

age of blood. The decrease in 2,3-DPG and P50 impair oxygen significant risk of dying.71-73
offloading, and deformation of the red cells impairs microcir- Many of these patients have suffered substantial bleeding
culatory perfusion.64 and may receive a significant transfusion, generally defined as
Treatment of Anemia: Transfusion Triggers. A 1988 the administration of ≥4 to 6 units of red blood cells within 4 to
National Institutes of Health Consensus Report challenged the 6 hours of admission. This definition is admittedly arbitrary.
BASIC CONSIDERATIONS

dictum that a hemoglobin value of less than 10 g/dL or a hema- Although 25% of all trauma admissions receive a unit of blood
tocrit level less than 30% indicates a need for preoperative red early after admission, only a small percentage of patients receive
blood cell transfusion. This was verified in a prospective ran- a massive transfusion. In the military setting, the percentage of
domized controlled trial in critically ill patients that compared massive transfusion patients almost doubles.74
a restrictive transfusion threshold to a more liberal strategy and Damage Control Resuscitation.  Standard advanced trauma
demonstrated that maintaining hemoglobin levels between 7 and life support guidelines start resuscitation with crystalloid, fol-
9 g/dL had no adverse effect on mortality. In fact, patients with lowed by packed red blood cells.75 Only after several liters of
APACHE II scores of ≤20 or patients age <55 years actually had crystalloid have been transfused does transfusion of units of
a lower mortality.65 plasma or platelets begin. This conventional massive trans-
Despite these results, change in daily clinical practice has fusion practice was based on a several small uncontrolled
been slow. Critically ill patients still frequently receive transfu- retrospective studies that used blood products containing
sions, with the pretransfusion hemoglobin approaching 9 g/dL increased amounts of plasma, which are no longer available.76
in a recent large observational study.66 This outdated approach Because of the known early coagulopathy of trauma, the cur-
unnecessarily exposes patients to increased risk and little benefit. rent approach to managing the exsanguinating patient involves
One unresolved issue related to transfusion triggers is early implementation of damage control resuscitation
the safety of maintaining a hemoglobin of 7 g/dL in a patient 5 (DCR). Although most of the attention to hemorrhagic
with ischemic heart disease. Data on this subject are mixed, and shock resuscitation has centered on higher ratios of plasma and
many studies have significant design flaws, including their ret- platelets, DCR is actually composed of three basic components:
rospective nature. However, the majority of the published data permissive hypotension, minimizing crystalloid-based resusci-
favors a restrictive transfusion trigger for patients with non-ST tation, and the immediate release and administration of pre-
elevation acute coronary syndrome, with many reporting worse defined blood products (red blood cells, plasma, and platelets)
outcomes in those patients receiving transfusions.67,68 in ratios similar to those of whole blood.
In Iraq and Afghanistan, DCR practices are demonstrating
Volume Replacement unprecedented success with improved overall survival.77 Civil-
The most common indication for blood transfusion in surgical ian data also suggest that a balanced resuscitation approach
patients is the replenishment of the blood volume; however, a yields improved outcome in severely injured and bleeding
deficit is difficult to evaluate. Measurements of hemoglobin trauma patients.69 To verify military and single-institution civil-
or hematocrit levels are frequently used to assess blood loss. ian data on DCR, a multicenter retrospective study of modern
These measurements can be occasionally misleading in the face transfusion practice at 17 leading civilian trauma centers was
of acute loss. Both the amount and the rate of bleeding are fac- performed.78 It was found that plasma:platelet:red blood cell
tors in the development of signs and symptoms of blood loss. ratios varied from 1:1:1 to 0.3:0.1:1, with corresponding sur-
Loss of blood in the operating room can be roughly evalu- vival rates ranging from 71% to 41%. A significant center effect
ated by estimating the amount of blood in the wound and on the was seen, documenting wide variation in both transfusion prac-
drapes, weighing the sponges, and quantifying blood suctioned tice and outcomes between Level 1 trauma centers. This varia-
from the operative field. In patients with normal preoperative tion correlated with blood product ratios. Increased plasma- and
values, blood loss up to 20% of total blood volume can be platelet-to-RBC ratios significantly decreased truncal hem-
replaced with crystalloid or colloid solutions. Blood loss above orrhagic death and 30-day mortality without a concomitant
this value may require the addition of a balanced resuscitation increase in multiple organ failure as a cause of death. A prospec-
including red blood cells, FFP, and platelets (detailed later in tive observational study evaluating current transfusion practice
this chapter) (Table 4-5). at 10 Level 1 centers was recently published, again documenting
the wide variability in practice and improved outcomes with
New Concepts in Resuscitation earlier use of increased ratios of plasma and platelets.79 Patients
Traditional resuscitation algorithms are sequentially based on receiving ratios less than 1:2 were four times more likely to die
crystalloid followed by red blood cells and then plasma and than patients with ratios of 1:1 or higher.
platelet transfusions and have been in widespread use since the Regardless of the optimal ratio, it is essential that the
1970s. No quality clinical data supported this concept. Recently trauma center has an established mechanism to deliver these
the damage control resuscitation (DCR) strategy, aimed at halt- products quickly and in the correct amounts to these critically
ing and/or preventing rather than treating the lethal triad of injured patients. In fact, several authors have shown that a
coagulopathy, acidosis, and hypothermia, has challenged tradi- well-developed massive transfusion protocol is associated with
tional thinking on early resuscitation strategies.69 improved outcomes independent of the ratios chosen.80 This
Rationale.  In civilian trauma systems, nearly half of all aggressive delivery of predefined blood products should begin
deaths happen before a patient reaches the hospital, and many prior to any laboratory-defined anemia or coagulopathy.

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
Table 4-5
Replacement of clotting factors

Life Span In Vivo Fate during Level Required for Ideal Agent ACD Bank Ideal Agent for
Factor Normal Level (Half-Life) Coagulation Safe Hemostasis Blood (4°C [39.2°F]) Replacing Deficit
I (fibrinogen) 200–400 mg/100 mL 72 h Consumed 60–100 mg/100 mL Very stable Bank blood; concentrated
fibrinogen
II (prothrombin) 20 mg/100 mL (100% of 72 h Consumed 15%–20% Stable Bank blood; concentrated
normal level) preparation
V (proaccelerin, accelerator 100% of normal level 36 h Consumed 5%–20% Labile (40% of normal Fresh frozen plasma; blood
globulin, labile factor) level at 1 wk) under 7 d
VII (proconvertin, serum 100% of normal level 5h Survives 5%–30% Stable Bank blood; concentrated
prothrombin conversion preparation
accelerator, stable factor)
VIII (antihemophilic factor, 100% of normal level 6–12 h Consumed 30% Labile (20%–40% of Fresh frozen plasma;
antihemophilic globulin) (50%–150% of normal normal level at 1 wk) concentrated antihemophilic
level) factor; cryoprecipitate
IX (Christmas factor, plasma 100% of normal level 24 h Survives 20%–30% Stable Fresh-frozen plasma; bank
thromboplastin component) blood; concentrated preparation
X (Stuart-Prower factor) 100% of normal level 40 h Survives 15%–20% Stable Bank blood; concentrated
preparation
XI (plasma thromboplastin 100% of normal level Probably 40–80 h Survives 10% Probably stable Bank blood
antecedent)
XII (Hageman factor) 100% of normal level Unknown Survives Deficit produces no Stable Replacement not required
bleeding tendency
XIII (fibrinase, fibrin- 100% of normal level 4–7 d Survives Probably <1% Stable Bank blood
stabilizing factor)
Platelets 150,000–400,000/μL 8–11 d Consumed 60,000–100,000/μL Very labile (40% of Fresh blood or plasma; fresh
normal level at 20 h; platelet concentrate (not frozen
0 at 48 h) plasma)
ACD = acid-citrate-dextrose.
Source: Reproduced with permission from Salzman EW: Hemorrhagic disorders. In: Kinney JM, Egdahl RH, Zuidema GD, eds. Manual of Preoperative and Postoperative Care. 2nd ed. Philadelphia: WB Saunders;
1971:157. Copyright Elsevier.

CHAPTER 4

99
Hemostasis, Surgical Bleeding, and Transfusion
VRG RELEASE: tahir99
https://kickass.to/user/tahir99/uploads/
100 Table 4-6
Adult Transfusion Clinical Practice Guideline
A.  Initial Transfusion of Red Blood Cells (RBCs):
1. Notify blood bank immediately of urgent need for RBCs.
PART I

O negative uncross-matched (available immediately).


As soon as possible, switch to O negative for females and O positive for males.
Type-specific uncross-matched (available in approximately 5–10 min).
Completely cross-matched (available in approximately 40 min).
2.  A blood sample must be sent to blood bank for a type and cross.
BASIC CONSIDERATIONS

3. The Emergency Release of Blood form must be completed. If the blood type is not known and blood is needed immediately,
O-negative RBCs should be issued.
4.  RBCs will be transfused in the standard fashion. All patients must be identified (name and number) prior to transfusion.
5. Patients who are unstable or receive 1–2 RBCs and do not rapidly respond should be considered candidates for the massive
transfusion (MT) guideline.
B.  Adult Massive Transfusion Guideline:
1. The Massive Transfusion Guideline (MTG) should be initiated as soon as it is anticipated that a patient will require massive
transfusion (≥10 U RBCs in 24 h). The Blood Bank should strive to deliver plasma, platelets, and RBCs in a 1:1:1 ratio. To
be effective and minimize further dilutional coagulopathy, the 1:1:1 ratio must be initiated early, ideally with the first
2 units of transfused RBCs. Crystalloid infusion should be minimized.
2. Once the MTG is activated, the Blood Bank will have 6 RBCs, 6 FFP, and a 6 pack of platelets packed in a cooler available
for rapid transport. If 6 units of thawed FFP are not immediately available, the Blood Bank will issue units that are ready
and notify appropriate personnel when the remainder is thawed. Every attempt should be made to obtain a 1:1:1 ratio of
plasma:platelets:RBCs.
3. Once initiated, the MT will continue until stopped by the attending physician. MT should be terminated once the patient is
no longer actively bleeding.
4.  No blood components will be issued without a pickup slip with the recipient’s medical record number and name.
5. Basic laboratory tests should be drawn immediately on ED arrival and optimally performed on point-of-care devices,
facilitating timely delivery of relevant information to the attending clinicians. These tests should be repeated as clinically
indicated (e.g., after each cooler of products has been transfused). Suggested laboratory values are:
•  CBC
•  INR, fibrinogen
•  pH and/or base deficit
•  TEG, where available
CBC = complete blood count; ED = emergency department; FFP = fresh frozen plasma; INR = international normalized ratio; TEG = thromboelastography.

An example of an adult massive transfusion clinical guide- transfusions). Preformed cytokines in donated blood and recipi-
line specifying the early use of component therapy is shown in ent antibodies reacting with donated antibodies are postulated eti-
Table 4-6. Specific recommendations for the administra- ologies. The incidence of febrile reactions can be greatly reduced
6 tion of component therapy during a massive transfusion are by the use of leukocyte-reduced blood products. Pretreatment
shown in Table 4-7. Because only a small percentage of trauma with acetaminophen reduces the severity of the reaction.
patients require a massive transfusion and because blood prod- Bacterial contamination of infused blood is rare. Gram-
ucts in general are in short supply, the need for early prediction negative organisms, which are capable of growth at 4°C, are the
models has been studied and a comparison of results from both most common cause. Most cases, however, are associated with
civilian and military studies is shown in Table 4-8.81-85 While the administration of platelets that are stored at 20°C or, even
compelling, none of these algorithms have been prospectively more commonly, with apheresis platelets stored at room tem-
validated. perature. Cases from FFP thawed in contaminated water baths
have also been reported.87 Bacterial contamination can result in
Complications of Transfusion (Table 4-9) sepsis and death in up 25% of patients.88 Clinical manifestations
Transfusion-related complications are primarily related to includes systemic signs such as fever and chills, tachycardia and
blood-induced proinflammatory responses. Transfusion-related hypotension, and gastrointestinal symptoms (abdominal cramps,
events are estimated to occur in approximately 10% of all trans- vomiting, and diarrhea). If the diagnosis is suspected, the trans-
fusions, but less than 0.5% are serious in nature. Transfusion- fusion should be discontinued and the blood cultured. Emer-
related deaths, although rare, do occur and are related primarily gency treatment includes oxygen, adrenergic blocking agents,
to transfusion-related acute lung injury (TRALI) (16%–22%), and antibiotics.
ABO hemolytic transfusion reactions (12%–15%), and bacterial
contamination of platelets (11%–18%).86 Allergic Reactions.  Allergic reactions are relatively frequent,
Nonhemolytic Reactions.  Febrile, nonhemolytic reactions occurring in about 1% of all transfusions. Reactions are usu-
are defined as an increase in temperature (>1°C) associated with ally mild and consist of rash, urticaria, and flushing. In rare
a transfusion and are fairly common (approximately 1% of all instances, anaphylactic shock develops. Allergic reactions are

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
The syndrome of TRALI is defined as noncardiogenic 101
Table 4-7
pulmonary edema related to transfusion.89 It can occur with
Component therapy administration during massive the administration of any plasma-containing blood product.
transfusion Symptoms are similar to circulatory overload with dyspnea

CHAPTER 4
Fresh frozen As soon as the need for massive transfusion and associated hypoxemia. However, TRALI is characterized
plasma (FFP) is recognized. as noncardiogenic and is often accompanied by fever, rigors,
For every 6 red blood cells (RBCs), give and bilateral pulmonary infiltrates on chest x-ray. It most com-
6 FFP (1:1 ratio). monly occurs within 1 to 2 hours after the onset of transfusion
but virtually always before 6 hours. Toy et al recently reported
Platelets For every 6 RBCs and plasma, give one
a decrease in the incidence of TRALI with the reduction trans-

Hemostasis, Surgical Bleeding, and Transfusion


6 pack of platelets. 6 random-donor platelet
fusion of plasma from female donors, due to a combination of
packs = 1 apheresis platelet unit.
reduced transfusion of strong cognate HLA class II antibod-
Platelets are in every cooler.
ies and HNA antibodies in patients with risk factors for acute
Keep platelet counts >100,000.
lung injury.90 Treatment of TRALI entails discontinuation of
Cryoprecipitate After first 6 RBCs, check fibrinogen any transfusion, notification of the transfusion service, and pul-
level. If ≤200 mg/dL, give 20 units monary support, which may vary from supplemental oxygen to
cryoprecipitate (2 g fibrinogen). Repeat mechanical ventilation.
as needed, depending on fibrinogen Hemolytic Reactions.  Hemolytic reactions can be classified
level, and request appropriate amount of as either acute of delayed. Acute hemolytic reactions occur
cryoprecipitate. with the administration of ABO-incompatible blood and can be
fatal in up to 6% of cases. Contributing factors include errors in
the laboratory of a technical or clerical nature or the administra-
caused by the transfusion of antibodies from hypersensitive tion of the wrong blood type. Immediate hemolytic reactions
donors or the transfusion of antigens to which the recipient is are characterized by intravascular destruction of red blood
hypersensitive. Allergic reactions can occur after the adminis- cells and consequent hemoglobinemia and hemoglobinuria.
tration of any blood product but are commonly associated with DIC can be initiated by antibody-antigen complexes activat-
FFP and platelets. Treatment and prophylaxis consist of the ing factor XII and complement, leading to activation of the
administration of antihistamines. In more serious cases, epi- coagulation cascade. Finally, acute renal insufficiency results
nephrine or steroids may be indicated. from the toxicity associated with free hemoglobin in the plasma,
Respiratory Complications.  Respiratory compromise may resulting in tubular necrosis and precipitation of hemoglobin
be associated with transfusion-associated circulatory overload within the tubules.
(TACO), which is an avoidable complication. It can occur with Delayed hemolytic transfusion reactions occur 2 to 10 days
rapid infusion of blood, plasma expanders, and crystalloids, par- after transfusion and are characterized by extravascular hemo-
ticularly in older patients with underlying heart disease. Central lysis, mild anemia, and indirect (unconjugated) hyperbilirubine-
venous pressure monitoring should be considered whenever mia. They occur when an individual has a low antibody titer at
large amounts of fluid are administered. Overload is manifest the time of transfusion, but the titer increases after transfusion
by a rise in venous pressure, dyspnea, and cough. Rales can gen- as a result of an anamnestic response. Reactions to non-ABO
erally be heard at the lung bases. Treatment consists of diuresis, antigens involve immunoglobulin G-mediated clearance by the
slowing the rate of blood administration, and minimizing fluids reticuloendothelial system.
while blood products are being transfused. If the patient is awake, the most common symptoms of
acute transfusion reactions are pain at the site of transfusion,
facial flushing, and back and chest pain. Associated symptoms
Table 4-8 include fever, respiratory distress, hypotension, and tachycardia.
In anesthetized patients, diffuse bleeding and hypotension are the
Comparison of massive transfusion prediction studies
hallmarks. A high index of suspicion is needed to make the diag-
Author Variables ROC AUC Value nosis. The laboratory criteria for a transfusion reaction are hemo-
globinuria and serologic criteria that show incompatibility of the
McLaughlin et al 81
SBP, HR, pH, Hct 0.839 donor and recipient blood. A positive Coombs’ test indicates
Yücel et al 82
SBP, HR, BD, Hgb, 0.892 transfused cells coated with patient antibody and is diagnostic.
Male, + FAST, long Delayed hemolytic transfusions may also be manifest by fever
bone/pelvic fracture and recurrent anemia. Jaundice and decreased haptoglobin usu-
Moore et al83 SBP, pH, ISS >25 0.804 ally occur, and low-grade hemoglobinemia and hemoglobinuria
may be seen. The Coombs’ test is usually positive, and the blood
Schreiber et al 84
Hgb ≤11, INR >1.5, 0.80
bank must identify the antigen to prevent subsequent reactions.
penetrating injury
If an immediate hemolytic transfusion reaction is sus-
Cotton et al85 HR, SBP, FAST, 0.83-0.90 pected, the transfusion should be stopped immediately, and
penetrating injury a sample of the recipient’s blood drawn and sent along with
AUC = area under the curve; BD = base deficit; FAST = Focused assess- the suspected unit to the blood bank for comparison with the
ment with sonography for trauma; Hct = hematocrit; Hgb = hemoglobin; pretransfusion samples. Urine output should be monitored and
HR = heart rate; INR = international normalized ratio; ISS = injury adequate hydration maintained to prevent precipitation of hemo-
severity score; ROC = receiver operating characteristic; SBP = systolic globin within the tubules. Delayed hemolytic transfusion reac-
blood pressure.
tions do not usually require specific intervention.

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
102 Table 4-9
Transfusion-related complications

Abbreviation Complication Signs and Symptoms Frequency Mechanism Prevention


PART I

NHTR Febrile, Fever 0.5%–1.5% of Preformed cytokines Use leukocyte-


 nonhemolytic  transfusions Host Ab to donor   reduced blood
 transfusion  lymphocytes Store platelets <5 d
 reaction
BASIC CONSIDERATIONS

Bacterial High fever, chills <<0.05% of blood Infusion of


 contamination Hemodynamic changes 0.05% of platelets   contaminated blood
DIC
Emesis, diarrhea
Hemoglobinuria
Allergic reactions Rash, hives 0.1%–0.3% of Soluble transfusion Provide
Itching  units  constituents  antihistamine
 prophylaxis
TACO Transfusion- Pulmonary edema ? 1:200–1:10,00 of Large volume of Increase transfusion
 associated  transfused   blood transfused  time
 circulatory  patients   into an older patient Administer diuretics
 overload   with CHF Minimize
  associated fluids
TRALI Transfusion-related Acute (<6 h) hypoxemia Anti-HLA or Limit female donors
  acute lung injury Bilateral infiltrates ±   anti-HNA Ab in
Tachycardia,   transfused blood
hypotension   attacks circulatory
  and pulmonary
 leukocytes
Hemolytic reaction, Fever 1:33,000– Transfusion of ABO- Transfuse
 acute Hypotension   1:1,500,000 units   incompatible blood  appropriately
DIC Preformed IgM Ab to   matched blood
Hemoglobinuria   ABO Ag
Hemoglobinemia
Renal insufficiency
Hemolytic reaction, Anemia IgG mediated Identify patient’s
  delayed (2–10 d) Indirect   Ag to prevent
 hyperbilirubinemia  recurrence
Decreased haptoglobin
 level
Positive result on direct
  Coombs’ test
Ab = antibody; Ag = antigen; CHF = congestive heart failure; DIC = disseminated intravascular coagulation; HLA = human leukocyte antigen; HNA =
anti-human neutrophil antigen; IgG = immunoglobulin G; IgM = immunoglobulin M.

Transmission of Disease.  Malaria, Chagas’ disease, brucel- the decreased rates of transmission. Recent concerns about the
losis, and, very rarely, syphilis are among the diseases that have rare transmission of these and other pathogens, such as West
been transmitted by transfusion. Malaria can be transmitted by Nile virus, are being addressed by current trials of “pathogen
all blood components. The species most commonly implicated inactivation systems” that reduce infectious levels of all viruses
is Plasmodium malariae. The incubation period ranges from and bacteria known to be transmittable by transfusion. Prion dis-
8 to 100 days; the initial manifestations are shaking chills and orders (e.g., Creutzfeldt-Jakob disease) also are transmissible by
spiking fever. Cytomegalovirus (CMV) infection resembling transfusion, but there is currently no information on inactivation
infectious mononucleosis also has occurred. of prions in blood products for transfusion.
Transmission of hepatitis C and HIV-1 has been dra-
matically minimized by the introduction of better antibody and
nucleic acid screening for these pathogens. The residual risk TESTS OF HEMOSTASIS AND BLOOD COAGULATION
among allogeneic donations is now estimated to be less than The initial approach to assessing hemostatic function is a care-
1 per 1,000,000 donations. The residual risk of hepatitis B is ful review of the patient’s clinical history (including previous
approximately 1 per 300,000 donations.91 Hepatitis A is very abnormal bleeding or bruising), drug use, and basic laboratory
rarely transmitted because there is no asymptomatic carrier testing. Common screening laboratory testing includes platelet
state. Improved donor selection and testing are responsible for count, PT or INR, and aPTT. Platelet dysfunction can occur

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
at either extreme of platelet count. The normal platelet count Additional medications may significantly impair hemo- 103
ranges from 150,000 to 400,000/μL. Whereas a platelet count static function, such as antiplatelet agents (clopidogrel and GP
greater than 1,000,000/μL may be associated with bleeding or IIb/IIIa inhibitors), anticoagulant agents (hirudin, chondroitin
thrombotic complications, increased bleeding complications sulfate, dermatan sulfate), and thrombolytic agents (streptokinase,

CHAPTER 4
may be observed with major surgical procedures when the plate- tPA). If abnormalities in any of the coagulation studies cannot
lets are below 50,000/μL and with minor surgical procedures be explained by known medications, congenital abnormalities of
when counts are below 30,000/μL, and spontaneous hemorrhage coagulation or comorbid disease should be considered.
can occur when the counts fall below 20,000/μL. Despite a lack Unfortunately, while these conventional tests (PT, aPTT)
of evidence supporting their use, platelet transfusions are still capture the classic intrinsic and extrinsic coagulation cascade,
recommended in ophthalmologic and neurosurgical procedures they do not reflect the complexity of in vivo coagulation.92

Hemostasis, Surgical Bleeding, and Transfusion


when the platelet count is less than 100,000/μL. Although they are useful to follow warfarin and heparin thera-
The PT and aPTT are variations of plasma recalcification pies, they poorly reflect the status of actively bleeding patients.
times initiated by the addition of a thromboplastic agent. The This is not surprising given that these tests use only plasma and
PT reagent contains thromboplastin and calcium that, when not whole blood to provide their assessment of the patient’s clot-
added to plasma, leads to the formation of a fibrin clot. The ting status. To better assess the complex and rapidly changing
PT test measures the function of factors I, II, V, VII, and X. interactions of an actively bleeding patient, many centers have
Factor VII is part of the extrinsic pathway, and the remaining moved to whole blood-viscoelastic testing such as TEG or rota-
factors are part of the common pathway. Factor VII has the tional thromboelastometry (ROTEM). In addition, some centers
shortest half-life of the coagulation factors, and its synthesis have demonstrated that the graphical display options allow for
is vitamin K dependent. The PT test is best suited to detect more rapid return of results and that these tests are actually less
abnormal coagulation caused by vitamin K deficiencies and expensive than standard coagulation panels.
warfarin therapy. TEG was originally described by Hartert in 1948.93 Con-
Due to variations in thromboplastin activity, it can be dif- tinuous improvements in this technique have made this test a
ficult to accurately assess the degree of anticoagulation on the valuable tool for the medical personnel interested in coagula-
basis of PT alone. To account for these variations, the INR is tion. The TEG monitors hemostasis as a dynamic process rather
now the method of choice for reporting PT values. The Interna- than revealing information of isolated conventional coagula-
tional Sensitivity Index (ISI) is unique to each batch of thrombo- tion screens.94 The TEG measures the viscoelastic properties of
plastin and is furnished by the manufacturer to the hematology blood as it is induced to clot under a low-shear environment
laboratory. Human brain thromboplastin has an ISI of 1, and the (resembling sluggish venous flow). The patterns of change in
optimal reagent has an ISI between 1.3 and 1.5. shear-elasticity enable the determination of the kinetics of clot
The INR is a calculated number derived from the follow- formation and growth as well as the strength and stability of
ing equation: the formed clot. The strength and stability provide information
about the ability of the clot to perform the work of hemostasis,
INR = (measured PT/normal PT)ISI
while the kinetics determines the adequacy of quantitative fac-
The aPTT reagent contains a phospholipid substitute, tors available for clot formation. A sample of celite-activated
activator, and calcium, which in the presence of plasma leads whole blood is placed into a prewarmed cuvette, and the clotting
to fibrin clot formation. The aPTT measures function of fac- process is activated with kaolin with standard TEG and kaolin
tors I, II, and V of the common pathway and factors VIII, IX, plus tissue factor with rapid TEG. A suspended piston is then
X, and XII of the intrinsic pathway. Heparin therapy is often lowered into the cuvette that moves in rotation of a 4.5-degree
monitored by following aPTT values with a therapeutic target arc backward and forward. The normal clot goes through accel-
range of 1.5 to 2.5 times the control value (approximately 50 to eration and strengthening phase. The fiber strands that interact
80 seconds). Low molecular weight heparins are selective Xa with activated platelets attach to the surface of the cuvette and
inhibitors that may mildly elevate the aPTT, but therapeutic the suspended piston. The clot forming in the cuvette trans-
monitoring is not routinely recommended. mits its movement onto the suspended piston. A “weak” clot
The bleeding time is used to evaluate platelet and vascular stretches and therefore delays the arc movement of the piston,
dysfunction, although not as frequently as in the past. Several which is graphically expressed as a narrow TEG. A strong clot,
standard methods have been described; however, the Ivy bleed- in contrast, will move the piston simultaneously and propor-
ing time is most commonly used. It is conducted by placing a tionally to the cuvette’s movements, creating a thick TEG. The
sphygmomanometer on the upper arm and inflating it to 40 mmHg, strength of a clot is graphically represented over time as a char-
and then a 5-mm stab incision is made on the flexor surface of acteristic cigar-shape figure (Fig. 4-6).
the forearm. The time is measured to cessation of bleeding, and
the upper limit or normal bleeding time with the Ivy test is 7
minutes. A template aids in administering a uniform test and
Coagulation Fibrinolysis
adds to the reproducibility of the results. An abnormal bleeding
time suggests platelet dysfunction (intrinsic or drug-induced), LY
vWD, or certain vascular defects. Many laboratories are replac- Angle
ing the template bleeding time with an in vitro test in which
blood is sucked through a capillary and the platelets adhere to
R K MA
the walls of the capillary and aggregate. The closure time in this
system appears to be more reproducible than the bleeding time
and also correlates with bleeding in vWD, primary platelet func- Figure 4-6.  Illustration of a thromboelastogram (TEG) tracing. K =
tion disorders, and patients who are taking aspirin. clot kinetics; LY = lysis; MA = maximal amplitude; R = reaction time.

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
104 Several parameters are generated from the TEG tracing. The platelets sensitize the recipient, who makes antibody to the
The r-value (reaction time) represents the time between the foreign platelet antigen. The foreign platelet antigen does not
start of the assay and initial clot formation. This reflects clot- completely disappear from the recipient circulation but attaches
ting factor activity and initial fibrin formation and is increased to the recipient’s own platelets. The antibody then destroys the
with factor deficiency or severe hemodilution. The k-time (clot recipient’s own platelets. The resultant thrombocytopenia and
PART I

kinetics) is the time needed to reach specified clot strength and bleeding may continue for several weeks. This uncommon cause
represents the interactions of clotting factors and platelets. As of thrombocytopenia should be considered if bleeding follows
such, the k-time is prolonged with hypofibrinogenemia and sig- transfusion by 5 or 6 days. Platelet transfusions are of little help
nificant factor deficiency. Prolonged r-value and k-time are com- in the management of this syndrome because the new donor
monly addressed with plasma transfusions. The alpha or angle platelets usually are subject to the binding of antigen and dam-
BASIC CONSIDERATIONS

(∝) is the slope of the tracing and reflects clot acceleration. The age from the antibody. Corticosteroids may be of some help
angle reflects the interactions of clotting factors and platelets. in reducing the bleeding tendency. Posttransfusion purpura is
The slope is decreased with hypofibrinogenemia and platelet self-limited, and the passage of several weeks inevitably leads
dysfunction. Decreased angles are treated with cryoprecipitate to subsidence of the problem.
transfusion or fibrinogen administration. The maximal ampli- DIC is characterized by systemic activation of the coagu-
tude (mA) is the greatest height of the tracing and represents clot lation system, which results in the deposition of fibrin clots and
strength. Its height is reduced with dysfunction or deficiencies in microvascular ischemia and may contribute to the development
platelets or fibrinogen. Decreased mA is addressed with platelet of multiorgan failure. Consumption and subsequent exhaustion
transfusion and, in cases where the angle is also decreased, with of coagulation proteins and platelets due to the ongoing acti-
cryoprecipitate (or fibrinogen) as well. The G-value is a paramet- vation of the coagulation system may induce severe bleeding
ric measure derived from the mA value and reflects overall clot complications.
strength or firmness. An increased G-value is associated with Lastly, severe hemorrhagic disorders due to thrombo-
hypercoagulability, whereas a decrease is seen with hypocoagu- cytopenia have occurred as a result of gram-negative sepsis.
lable states. Finally, the LY30 is the amount of lysis occurring in Defibrination and hemostatic failure also may occur with menin-
the clot, and the value is the percentage of amplitude reduction gococcemia, Clostridium perfringens sepsis, and staphylococcal
at 30 minutes after mA is achieved. The LY30 represents clot sepsis. Hemolysis appears to be one mechanism in sepsis leading
stability and when increased fibrinolysis is present. to defibrination.
TEG is the only test measuring all dynamic steps of clot for-
mation until eventual clot lysis or retraction. TEG has also been
REFERENCES
shown to identify on admission those patients likely to develop
thromboembolic complications after injury and postoperatively.95-97 Entries highlighted in blue are key references.
Recent trauma data have shown TEG to be useful in pre-    1. Brohi K, Cohen MJ, Davenport RA. Acute coagulopathy of
dicting early transfusion of red blood cells, plasma, platelets, trauma: mechanism, identification and effect. Curr Opin Crit
and cryoprecipitate.98 TEG can also predict the need for life- Care. 2007;13:680-685.
saving interventions shortly after arrival and to predict 24-hour    2. Kulkarani R. Comprehensive care of the patient with haemo-
and 30-day mortality.99 Lastly, TEG can be useful to guide philia and inhibitors undergoing surgery: practical aspects.
Hemophilia. 2013;19(1):2-10.
administration of TXA to injured patients with hyperfibrinoly-
   3. Federicini AB, Mannucci PM. Management of inherited von
sis.100 Our center now uses TEG rather than PT and a PTT to Willebrand disease in 2007. Ann Med. 2007;39:(5):346-358.
evaluate injured patients in the emergency room.101    4. Girolami A, de Marinis GB, Bonamigo E, Lombardi AM.
Recombinant FVIIa concentrate-associated thrombotic events
in congenital bleeding disorders other than hemophilias.
EVALUATION OF EXCESSIVE INTRAOPERATIVE Hematology. 2012;17(6):346-349.
OR POSTOPERATIVE BLEEDING    5. Peyvandi F, Bolton-Maggs PHB, Batorova A, De Moerloose
Excessive bleeding during or after a surgical procedure may be P. Rare bleeding disorders. Haemophilia. 2012;18(Suppl 4):
the result of ineffective hemostasis, blood transfusion, unde- 148-153.
tected hemostatic defect, consumptive coagulopathy, and/or   6. Peyvandi F, Mannucci PM. Rare coagulation disorders.
Thromb Haemost. 1999;82(4):1207-1214.
fibrinolysis. Excessive bleeding from the operative field unas-
   7. Anwar R, Miloszewski KJ. Factor XIII deficiency. Br J
sociated with bleeding from other sites usually suggests inad- Haematol. 1999;107(3):468-484.
equate mechanical hemostasis.    8. Anwar R, Minford A, Gallivan L, Trinh CH, Markham AF.
Massive blood transfusion is a well-known cause of throm- Delayed umbilical bleeding—a presenting feature for factor
bocytopenia. Bleeding following massive transfusion can occur XIII deficiency: clinical features, genetics, and management.
due to hypothermia, dilutional coagulopathy, platelet dysfunction, Pediatrics. 2002;109(2):E32.
fibrinolysis, or hypofibrinogenemia. Another cause of hemostatic    9. George JN, Caen JP, Nurden AT. Glanzmann’s thrombas-
failure related to the administration of blood is a hemolytic trans- thenia: the spectrum of clinical disease. Blood. 1990;75(7):
fusion reaction. The first sign of a transfusion reaction may be 1383-1395.
diffuse bleeding. The pathogenesis of this bleeding is thought to   10. Stasi R, Evangelista ML, Stipa E, et al. Idiopathic thrombo-
cytopenic purpura: current concepts in pathophysiology and
be related to the release of ADP from hemolyzed red blood cells,
management. Thromb Haemost. 2008;99:4-13.
resulting in diffuse platelet aggregation, after which the platelet   11. George JN. Sequence of treatments for adults with primary
clumps are removed out of the circulation. immune thrombocytopenia. Am J Hematol. 2012;87:S12-S15.
Transfusion purpura occurs when the donor platelets are   12. Baldwin ZK, Spitzer AL, Ng VL, Harkin AH. Contemporary
of the uncommon PlA1 group. This is an uncommon cause of standards for the diagnosis and treatment of heparin-induced
thrombocytopenia and associated bleeding after transfusion. thrombocytopenia (HIT). Surgery. 2008;143:305-312.

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
  13. Amiral J, Peynaud-Debayle E, Wolf M, et al. Generation of   32. Tripodi A, Mannucci PM. The coagulopathy of chronic liver 105
antibodies to heparin-PF4 complexes without thrombocyto- disease. N Engl J Med. 2011;365:147.
penia in patients treated with unfractionated or low-molecular   33. Tripodi A, Primignani M, Lemma L, et al. Detection of the
weight heparin. Am J Hematol. 1996;52:90-95. imbalance of procoagulant versus anticoagulant factors

CHAPTER 4
  14. Linkins LA, Dans AL, Moores LK, Bona R, Davidson BL, in cirrhosis by a simple laboratory method. Hepatology.
Schulman S, Crowther M; American College of Chest 2010;52:249-255.
Physicians. Treatment and prevention of heparin-induced   34. Stavitz RT. Potential applications of thromboelastography in
thrombocytopenia: Antithrombotic Therapy and Preven- patients with acute and chronic liver disease. Gastroenterol
tion of Thrombosis, 9th ed: American College of Chest Hepatol. 2012;8:513-520.
Physicians Evidence-Based Clinical Practice Guidelines.   35. Ghalib R, Levine C, Hassan M, et al. Recombinant human
Chest. 2012;141(2 Suppl):e495S-530S. interleukin-11 improves thrombocytopenia in patients with

Hemostasis, Surgical Bleeding, and Transfusion


 15. Zimrin AB, Hess JR. Thrombocytopenic purpura: going cirrhosis. Hepatology. 2003;37:1165-1171.
against the evidence. Crit Care Med. 2006;34:2247-2248.   36. Nunez TC, Cotton BA. Transfusion therapy in hemorrhagic
 16. Kremer Hovinga JA, Meyer SC. Current management of shock. Curr Opin Crit Care. 2009;15:536-541.
thrombotic thrombocytopenic purpura. Curr Opin Hematol.   37. Hess JR, Brohi K, Dutton RP, et al. The coagulopathy of
2008;15(5):445-450. trauma: a review of mechanisms. J Trauma. 2008;65:748-754.
  17. Fakhouri F, Vernant JP, Veyradier A, et al. Efficiency of cura-   38. Brohi K, Cohen MJ, Ganter MT, et al. Acute coagulopathy of
tive and prophylactic treatment with rituximab in ADAMT13- trauma: hypoperfusion induces systemic anticoagulation and
deficient thrombotic thrombocytopenic purpura: a study of hyperfibrinolysis. J Trauma. 2008;64:1211-1217.
11 cases. Blood. 2005;106:1932-1937.   39. Cotton BA, McCarthy J, Holcomb JB. The (irreversible) harm
  18. Brown LM, Call MS, Knudson M, et al. A normal platelet of dabigatran etexilate in acutely injured patients. N Eng J
count may not be enough: the impact of admission platelet Med. 2011;365(21):2039-2040.
count on mortality and transfusion in severely injured trauma   40. Eerenberg ES, Kamphuisen PW, Sijpkens MK, et al. Reversal
patients. J Trauma. 2011;71(2 Suppl 3):S337-S342. of rivaroxaban and dabigatran by prothrombin complex con-
  19. Holcomb JB, Zarzabal LA, Michalek JE, et al. Increased platelet: centrate. A randomized, placebo-controlled, crossover study
RBC ratios are associated with improved survival after massive in healthy subjects. Circulation. 2011;124:1573-1579.
transfusion. J Trauma. 2011;71(2 Suppl 3): S318-S328.   41. Kearon C, Hirsh J. Management of anticoagulation before and
  20. Eikelboom JW, Hirsh J, Spencer FA, Baglin TP, Weitz JI. after elective surgery. N Engl J Med. 1997;336:1506.
Antiplatelet Drugs: Antithrombotic Therapy and Prevention   42. Lindblad B. Protamine sulfate: a review of its effects—
of Thrombosis, 9th ed: American College of Chest Physi- hypersensitivity and toxicity. Eur J Vasc Surg. 1989;3:195.
cians Evidence-Based Clinical Practice Guidelines. Chest   43. Dentali, F, Ageno, W, Crowther, M. Treatment of coumarin-
2012;141(2 Suppl):e89S-e119S. associated coagulopathy: a systematic review and proposed
  21. Sarode R. How do I transfuse platelets (PLTs) to reverse anti- treatment algorithms. J Thromb Haemost. 2006;4:1853.
PLT drug effect? Transfusion. 2012;52:695-701.   44. Douketis JD Spyropoulos AC, Spencer FA, et al. Periopera-
  22. Lavelle WF, Lavell EA, Uhl R. Operative delay for orthopedic tive Management of Antithrombotic Therapy Antithrombotic
patients on clopidogrel (Plavix): a complete lack of consensus. Therapy and Prevention of Thrombosis, 9th ed: American
J Trauma. 2008;64:996. College of Chest Physicians Evidence-Based Clinical Prac-
  23. Hess JR, Lawson JH. The coagulopathy of trauma versus dissem- tice Guidelines. Chest. 2012;141(2 Suppl):e326S-e350S.
inated intravascular coagulation. J Trauma. 2006;60:S12-S19.   45. Besser MW, Klein AA. The coagulopathy of cardiopulmonary
  24. Taylor FB, Toh CH, Hoots WK, Wada H, Levi M. Towards bypass. Crit Rev Clin Lab Sci. 2011;47(5–6):197-212.
definition, clinical and laboratory criteria, and a scoring sys-   46. Simon TL, Akl BF, Murphy W. Controlled trial of routine
tem for disseminated intravascular coagulation. Thromb administration of platelet concentrates in cardiopulmonary
Haemost. 2001;86:1327-1330. bypass surgery. Ann Thorac Surg. 1984;37:359-364.
  25. Dhainaut JF, Yan SB, Joyce DE, et al. Treatment effects of   47. Taneja R, Fernandes P, Marwaha G, Cheng D, Bainbridge D.
drotrecogin alfa (activated) in patients with or without overt Perioperative coagulation management and blood conserva-
disseminated intravascular coagulation. J Thromb Haemost. tion in cardiac surgery: a Canadian survey. J Cardiothorac
2004;2:1924-1933. Vasc Anesth. 2008;22:662-669.
  26. Angstwurm MW, Dempfle CE, Spannagl M. New dissemi-   48. Murkin JM, Lux J, Shannon NA, et al: Aprotinin significantly
nated intravascular coagulation score: a useful tool to predict decreases bleeding and transfusion requirements in patients
mortality in comparison with Acute Physiology and Chronic receiving aspirin and undergoing cardiac operations. J Thorac
Health Evaluation II and Logistic Organ Dysfunction scores. Cardiovasc Surg. 1994;107:554.
Crit Care Med. 2006;34:314-320.   49. Andersen ND, Syamal D, Williams JB, et al. Intraoperative use
  27. Tefferi A. Polycythemia vera and essential thrombocythemia: of low-dose recombinant activated Factor VII during thoracic
2012 update on diagnosis, risk stratification, and management. aortic operations. Ann Thoracic Surg. 2012;93(6):1921-1929.
Am J Hematol. 2012;87(3):285-293.   50. Palm M, Altman J. Topical hemostatic agents: a review.
  28. Feistauer SM, Penner E, Mayr WR, et al. Target platelet anti- Dermatol Surg. 2008;34:431-445.
gen of autoantibodies in patients with primary biliary cirrho-   51. Larson PO. Topical hemostatic agents for dermatologic sur-
sis. Hepatology. 1997;25:1343. gery. J Dermatol Surg Oncol. 1988;14:623-632.
  29. Lisman T, Bongers TN, Adelmeijer J, et al. Elevated levels of von   52. Achneck HE, Sileshi, B, Jamiolkowski, RM, et al. A Com-
Willebrand factor in cirrhosis support platelet adhesion despite prehensive review of topical hemostatic agents: Efficacy
reduced functional capacity. Hepatology. 2006;44:53-61. and recommendations for use. Ann Surg 2010; 25:217-228.
  30. Northup PG, McMahon MM, Ruhl AP, et al. Coagulopathy  53. Martinowitz U, Schulman S. Fibrin sealant in surgery of
does not fully protect hospitalized cirrhosis patients from patients with a hemorrhagic diathesis. Thromb Haemost.
peripheral venous thromboembolism. Am J Gastroenterol. 1995;74:486-492.
2006;101:1524.  54. Kiraly LN, Underwood S, Differding JA, Schreiber MA.
  31. Gatt A, Riddell A, Calvaruso V, et al. Enhanced thrombin Transfusion of aged packed red blood cells results in decreased
generation in patients with cirrhosis-induced coagulopathy. tissue oxygenation in critically injured trauma patients.
J Thromb Haemost. 2010;8:1994. J Trauma. 2009;67(1):29-32.

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
106   55. McAlister FA, Clark HD, Wells PS, Laupacis A. Perioperative and outcomes in massively transfused combat casualties. J
allogeneic blood transfusion does not cause adverse sequelae Trauma Acute Care Surg. 2012;73(2 Suppl 1):S89-S94.
in patients with cander: a meta-analysis of unconfounded stud-   75. Carrico CJ, Canizaro PC, Shires GT. Fluid resuscitation fol-
ies. Br J Surg. 1998;85:171-178. lowing injury: rationale for the use of balanced salt solutions.
  56. Vamvakas EC, Blajchman MA. Universal WBC reduction: the Crit Care Med. 1976;4:46.
PART I

case for and against. Transfusion. 2001;41:691-712.   76. Harrigan C, Lucas CE, Ledgerwood AM, et al. Serial changes
  57. Hebert PC, Fergusson D, Blajchman MA, Leukoreduction in primary hemostasis after massive transfusion. Surgery.
Study Investigators. Clinical outcomes following institution of 1985;98:836.
the Canadian universal leukoreduction program for red blood   77. Borgman MA, Spinella PC, Perkins JG, et al. The ratio of
cell transfusions. JAMA. 2003;289(15):1941-1949. blood products transfused affects mortality in patients receiv-
  58. Inaba K, Lustenberger T, Rhee P, et al. The impact of platelet ing massive transfusions at a combat support hospital. J Trauma.
BASIC CONSIDERATIONS

transfusion in massively transfused trauma patients. J Am Coll 2007;63:805.


Surg. 2010;211(5):573-579.  78. Holcomb JB, Wade CE, Michalek JE, et al. Increased
  59. Sondeen JL, Prince MD, Medina L, et al. Comparison of plasma and platelet to red blood cell ratios improves out-
lyophilized swine plasma to fresh frozen plasma plus two come in 466 massively transfused civilian trauma patients.
ratios of packed red blood cells in a cold, coagulopathic, Ann Surg. 2008;248:447-458.
poly trauma severe hemorrhage shock swine model. Shock.   79. Holcomb JB, del Junco DJ, Fox EE, et al, for the PROM-
2008;29(suppl 1):13-14. MTT Study Group. The prospective, observational, multi-
  60. CRASH-2 Collaborators, Shakur H, Roberts I, et al. Effects center, massive transfusion study, PROMMTT: comparative
of tranexamic acid on death, vascular occlusive events, effectiveness of a time-varying treatment and competing risks.
and blood transfusion in trauma patients with significant Arch Surg. 2012;15:1-10.
haemorrhage (CRASH-2): a randomised, placebo-controlled   80. Cotton BA, Au BK, Nunez TC, et al. Predefined massive
trial. Lancet. 2010 Jul 3;376(9734):23-32. transfusion protocols are associated with a reduction in organ
  61. CRASH-2 Collaborators, Roberts I, Shakur H, Afolabi A, failure and postinjury complications. J Trauma. 2009;66:
et al. The importance of early treatment with tranexamic 41-48; discussion 48-49.
acid in bleeding trauma patients: an exploratory analy-   81. McLaughlin DF, Niles SE, Salinas J, et al. A predictive model
sis of the CRASH-2 randomized controlled trial. Lancet. for massive transfusion in combat casualty patients. J Trauma.
2011;377:1096. 2008;64(2 Suppl):S57.
  62. Henry DA, Carless PA, Moxey AJ, et al. Antifibrinolytic use   82. Yücel N, Lefering R, Maegele M, et al. Trauma-Associated
for minimizing perioperative allogeneic blood transfusion. Severe Hemorrhage (TASH) score: probability of mass trans-
Cochrane Database Syst Rev. 2011;(3):CD001886. fusion as surrogate for life threatening hemorrhage after mul-
 63. Kirkpatrick AW, Holcomb JB, Stephens MH, Gruen R; tiple trauma. J Trauma. 2006;60:1228.
Members of the Evidence-Based Reviews in Surgery Group.   83. Moore FA, Nelson T, McKinley BA, et al. Massive transfu-
Tranexamic acid effects in trauma patients with significant sion in trauma patients: tissue hemoglobin oxygen saturation
hemorrhage. J Am Coll Surg. 2012;215(3):438-440. predicts poor outcome. J Trauma. 2008;64:1010.
  64. Gerber DR. Transfusion of packed red blood cells in patients   84. Schreiber MA, Perkins J, Kiraly L, et al. Early predictors of
with ischemic heart disease. Crit Care Med. 2008;36: massive transfusion in combat casualties. J Am Coll Surg.
1068-1074. 2007;205:541.
  65. Herbert PC, Wells GW, Blajchman MA, et al. A multicenter,   85. Cotton BA, Dossett LA, Haut ER, et al. Multicenter validation
randomized, controlled clinical trial of transfusion require- of a simplified score to predict massive transfusion in trauma.
ment in critical care. N Engl J Med. 1999;340:409-417. J Trauma. 2010;69(Suppl 1):S33-S39.
  66. Corwin HL, Gettinger A, Pearl RG, et al. The CRIT Study: ane-   86. Despotis GJ, Zhang L, Lublin DM. Transfusion risks and
mia and blood transfusion in the critically ill—current clinical transfusion-related pro-inflammatory responses. Hematol
practice in the United States. Crit Care Med. 2004;32:399-352. Oncol Clin N Am. 2007;21:147.
  67. Carson JL, Terrin ML, Noveck H, et al. Liberal or restrictive   87. Pandey S, Vyas GN. Adverse-effects of plasma transfusion.
transfusion in high-risk patients after hip surgery. N Engl J Transfusion. 2012;52:65S-79S.
Med. 2011;365(26):2453-2462.   88. Goodnough LT, Brecher ME, Kanter MH: Transfusion medi-
  68. Carson JL, Carless PA, Hebert PC. Transfusion thresholds and cine: blood transfusion. N Engl J Med. 1999;340:438.
other strategies for guiding allogeneic red blood cell transfu-   89. Looney MR, Gropper MA, Matthay MA. Transfusion-related
sion. Cochrane Database Syst Rev. 2012;4:CD002042. acute lung injury. Chest. 2004;126:249.
  69. Cotton BA, Reddy N, Hatch QM, et al. Damage control  90. Toy P, Gajic O, Bacchetti P, et al. Transfusion-related
resuscitation is associated with a reduction in resuscitation acute lung injury: incidence and risk factors. Blood.
volumes and improvement in survival in 390 damage con- 2012;119(7):1757-1767.
trol laparotomy patients. Ann Surg. 2011;254(4):598-605.   91. Zou S, Stramer SL, Dodd RY. Donor testing and risk: cur-
  70. Kauvar DS, Lefering R, Wade CE. Impact of hemorrhage rent prevalence, incidence, and residual risk of transfusion-
on trauma outcome: an overview of epidemiology, clini- transmissible agents in US allogeneic donations. Transfusion
cal presentations, and therapeutic considerations. J Trauma. Med Rev. 2012;26(2):119-128.
2006;60(6 Suppl):S3.   92. Hoffman M, Monroe DM. Coagulation 2006: a modern view
  71. Niles SE, McLaughlin DF, Perkins J, et al. Increased mortal- of hemostasis. Hematol Oncol Clin North Am. 2007;21:1-11.
ity associated with early coagulopathy after trauma in combat   93. Hartert H. Blutgerinnungsstudien mit der thrombelastogra-
casualties. J Trauma. 2008;64:1459-1463. phie, einem neuen untersuchungsverfahren. Klin Wochenschr.
  72. Macleod J, Lynn M, McKenney MG, Jeroukhimov I, Cohn 1948;26:577.
SM. Predictors of mortality in trauma patients. Am Surg.   94. Mallet SV, Cox DJA. Thromboelastography: a review article.
2004;70:805-810. Br J Anaesth. 1992;69:307.
  73. Brohi K, Singh J, Heron M, Coats T. Acute traumatic coagu-  95. Cotton BA, Radwan ZA, Matijevic N, et al. Admission
lopathy. J Trauma. 2003;54:1127-1130. rapid thromboelastography (rTEG) predicts development of
  74. Cap AP, Spinella PC, Borgman MA, Blackbourne LH, Per- pulmonary embolism in trauma patients. J Trauma. 2012;72(6):
kins JG. Timing and location of blood product transfusion 1470-1477.

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
  96. Caprini JA, Arcelus JI, Laubach M, et al. Postoperative hyper- 100. Cotton BA, Harvin JA, Kostousouv V, et al. Hyperfibrinolysis 107
coagulability and deep-vein thrombosis after laparoscopic on admission is an uncommon but highly lethal event asso-
cholecystectomy. Surg Endosc. 1995;9:304-309. ciated with shock and pre-hospital fluid administration.
  97. Dai Y, Lee A, Critchley LA, et al. Does thromboelastography J Trauma. 2012;72(2):365-370.

CHAPTER 4
predict postoperative thromboembolic events? A systematic 101. Holcomb JB, Minei KM, Scerbo ML, et al. Admission rapid
review of the literature. Anesth Analg. 2009;108:734-742. thromboelastography (r-TEG) can replace conventional
  98. Cotton BA, Faz G, Hatch Q, et al. Rapid thromboelastogra- coagulation tests in the emergency department: experi-
phy (r-TEG) delivers real-time results that predict transfusion ence with 1974 consecutive trauma patients. Ann Surg.
within one hour of admission. J Trauma. 2011;71(2):407-417. 2012;256(3):476-486.
  99. Schöchl H, Cotton BA, Inaba K, et al. FIBTEM provides
early prediction of massive transfusion in trauma. Crit Care.

Hemostasis, Surgical Bleeding, and Transfusion


2011;15:R265-R271.

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/
This page intentionally left blank

VRG RELEASE: tahir99


https://kickass.to/user/tahir99/uploads/

S-ar putea să vă placă și