Anaesthetic considerations in patients with inherited disorders of coagulation

Similar documents
Alternative Haemostatic Agents in the Management of Obstetric Haemorrhage

Disclosure. Hemophilia: The Royal Treatment. Objectives. Background. History of Hemophilia. Epidemiology 1/4/2018

The Coagulation Workup In The Office Setting

Common Inherited Bleeding Disorders

Prior Authorization Criteria Hemophilia/Blood Factor Products

Manejo de la transfusión de plaquetas. Ileana López-Plaza, MD

Clinical Policy: Humate-P (Antihemophiliac Factor/von Willebrand Factor Complex Human) Reference Number: CP.MP.404

Common Inherited Coagulation Disorders. Inherited Coagulation Disorders. All coagulation factors and platelets in circulation (unactivated)

LABORATORY APPROACH TO BLEEDING DISORDERS DR NISHANTH PG 1 ST YEAR DEPARTMENT OF PATHOLOGY

Factor Concentrates. More is better? Alexander Duncan MD Emory Medical Labs

Factor VIII Concentrate Factor IX Complex (Coagulation Factors, II, VII, IX, X) Concentrate

Local vasoconstriction. is due to local spasm of the smooth muscle (symp. reflex) can be maintained by platelet vasoconstrictors

Platelet type bleeding: (vwd) - Petechiae, purpura, ecchymoses, bruising - Menorrhagia, hematuria, occult GI bleed - Gingival bleeding, epistaxis

Perioperative management of patients on warfarin requiring elective surgery Dr K Boyd, Mrs S Doyle

Congenital Fibrinogen Deficiency. Claude Negrier, MD, PhD Hôpital Louis Pradel Université Claude Bernard Lyon 1

CLOTTING FACTORS AND COAGULANT BLOOD PRODUCTS

Session 1 Topics. Vascular Phase of Hemostasis. Coagulation Pathway. Action of Unfractionated Heparin. Laboratory Monitoring of Anticoagulant Therapy

LABORATORY DIAGNOSIS OF BLEEDING DISORDERS

CLOTTING FACTORS, COAGULANT BLOOD PRODUCTS & OTHER HEMOSTATICS

CLOTTING FACTOR REPLACEMENT THERAPY

Primary hemostasis. Vascular endothelium Vasoconstriction : local tissue factor, nervous system

Category Storage Shelf Life Additional Criteria

Platelet Factor IV- Heparin Antibodies. Presenter: Michael J. Warhol, M.D.

Laboratory investigation in the Bleeding Patient. Dr Craig Taylor Consultant Haematologist May 2016

Current and Emerging Treatment Paradigms in the Management of Hemophilia

How to Approach a Patient with Bleeding. Nigel Key MB ChB FRCP. November 1, ISTH Advanced Training Course. ISTH Advanced Training Course

Coagulation Cascade. TF-VIIa. Tissue Factor + VII. XIIa. Prothrombin. XIa. IXa. VIIIa. Thrombin. XL- Fibrin. XIII Monomer. Fibrinogen.

Consulted With; Post/Committee/Group Date Haematology Dr Shereen Elshazly (Lead for Anticoagulation) January 2016 Department of Anaesthesia

Adv Pathophysiology Unit 4 Page 1 of 10. Learning Objectives for this file:

Plasma Testing in the Clinical Coagulation Laboratory: New drugs, new problems.

BLOOD COAGULATION. 1. The initial phase of the process is vascular constriction. This limits the flow of blood to the area of injury.

AUSTRALIAN BLEEDING DISORDERS REGISTRY

ANTICOAGULANT THERAPY ANTICOAGULANT THERAPY REVISITED Thrombosis is a complication of underaggressive anticoagulant therapy

CLOTTING FACTORS AND COAGULANT BLOOD PRODUCTS

Hypercoagulation. CP Conference 11/14/2006

HaematologY. Dr Tom Bate Again!

Haemostasis. The function of haemostasis is: to prevent blood loss from injured vessels. to stop bleeding. to prevent thrombosis

New Anticoagulants Linda Liu, M.D.

LVHN Scholarly Works. Lehigh Valley Health Network. Joseph G. Ottinger RPh, MS, MBA, BCPS Lehigh Valley Health Network,

iccnet CHSA Clinical Protocol - HEPARIN

Coagulation in perspective: Blood management. Objectives

Index. Autoimmune thrombocytopenic purpura (AITP), 100, 102, 105, 108, 187 Automated platelet counters, , 108

Clotting Disorder Therapy

Introduction Hemostasis: Tourniquet Test & Bleeding Time. Hematology-Immunology System Faculty of Medicine Universitas Padjadjaran LOGO

Lina Al-Lawama. Rama Al-Ashqar. Malik Al-Zohlof

These handouts are only meant as a guide while you follow the presentation on the screen. Sometimes the speaker will change some of the slides.

4/5/2017. Molecular Testing Applications in Coagulation. Disclosures. Objectives. Human Genome Chromosome Analysis.

Patients who Decline Blood Products:- Haematological Aspects of Care. Dr Catherine Flynn Consultant Haematologist CWIUH

UW Medicine Alternative Monitoring for Antithrombotic Agents

Clinical Procedure. ROTEM Trauma, Resuscitation. Princess Alexandra Hospital Emergency Department. 1 Introduction

Coagulation Mechanisms Dr. Nervana Bayoumy

Case 1. Case 1. Case 2. Case 2. Review of Bleeding Disorders Case 1

Thriving Amid The Turbulent Ride

Waterfall/ Cascade Model:1960 s. Coagulation Challenges During the Perioperative Period

Guideline on core SmPC for human fibrinogen products

LABORATORY APPROCH TO THE BLEEDING PATIENT

Blood transfusion in special situations and poly-trauma. Dr Marina Karakantza Consultant Haematologist in Transfusion Medicine, LTHT

Special Coagulation - APC Resistance. DiaPharma Group, Inc. Customer Service: Technical Support:

Antihemophilic Products

Princess Alexandra Hospital Emergency Department. Clinical Procedure. 1 Introduction

Omar A. Elkashef, MD

Emergency and Perioperative Hemostasis Testing: Which Assays Provide Helpful Information. Wayne Chandler, MD Laboratory Medicine Seattle Children s

Evaluation of Complex Coagulation Cases: Case-Based Illustrations of Important Issues

ANEMIA. Oral iron. IV iron gluconate (order set #233)

Anticlotting mechanisms 1: physiology and pathology

Clinical Use of Plasma for Transfusion. Irma Szymanski, MD Professor of Pathology, Emerita University of Massachusetts Medical School

Personal history of excessive mucocutaneous bleeding. Family history of excessive bleeding. Laboratory tests of hemostasis consistent with VWD

Using TEG in the ED, OR, and ICU. Don H. Van Boerum, MD, FACS

General Principles of. Hemostasis. Kristine Krafts, M.D.

GUIDELINES FOR THE TREATMENT OF CONGENITAL DISORDERS OF FIBRINOGEN. Australian Haemophilia Centre Directors Organisation

A Review of Coagulation Factor Disorders

Anticoagulation in VTE The Haematologist s Perspective. Dr. M.D. Maina FRCP Edin.

Drugs used in Thromboembolic Disease. Munir Gharaibeh, MD, PhD, MHPE Department of Pharmacology Faculty of Medicine October 2014

Human coagulation factor VIII and human von Willebrand factor complex, powder for injection.

Managing Massive Transfusions in diverse Patient Populations in a Non-Metropolitan Area

Sign up to receive ATOTW weekly -

ANTICOAGULANT THERAPY ANTICOAGULANT THERAPY REVISITED 2005

ANTICOAGULANT THERAPY REVISITED Thrombosis is a complication of underaggressive anticoagulant therapy ANTICOAGULANT THERAPY.

DOACs: When and how to measure their anticoagulant effect

Type of intervention Treatment; secondary prevention. Economic study type Cost-effectiveness analysis.

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP) CORE SPC FOR HUMAN PLASMA DERIVED AND RECOMBINANT COAGULATION FACTOR IX PRODUCTS

Heparin Induced Thrombocytopenia. Heparin Induced Thrombocytopenia. Heparin Induced Thrombocytopenia. Temporal Aspects.

Managing the Risks Associated with Anticoagulant Therapy. Steve McGlynn Specialist Principal Pharmacist (Cardiology) NHS Greater Glasgow and Clyde

Anti- THrombosis with Enoxaparin in intubated Adolescents

Riastap (fibrinogen concentrate, human) Public Summary of Risk Management Plan (Extract from the EU Risk Management Plan Version 3.

Transfusion & Non-transfusion Approaches to Bleeding

TABLE I. PRODUCTS LICENSED IN THE U.S. TO TREAT HEMOPHILIA A. A. Recombinant FACTOR VIII Concentrates

The Combined Use of Bypassing Agents with Antithrombin Reduction in Plasma of Hemophilia A and B Patients with Inhibitors

Subject: Clotting Factors and Coagulant Blood Products

P. W. HOWIE M.D., M.R.C.O.G. first method of activation is the intrinsic system. Hageman factor, which in turn activates a series of

Novel Anti-coagulant Agents. David G Hovord BA MB BChir FRCA Clinical Assistant Professor University of Michigan

Disclosures. Thromboelastography. TEG Methodology. TEG Output. Thromboelastography (TEG): Basics & Clinical Applications

Disclosure (s) Relevant financial relationship(s) None Off-label usage None Change in slide set You betcha! (YES!) 2015 MFMER slide-1

Scottish Medicines Consortium

Guideline on core SmPC for human plasma derived and recombinant coagulation factor VIII products

The legally binding text is the original French version TRANSPARENCY COMMITTEE OPINION. 24 June 2009

If not heparin for bypass then what? Dr Tony Moriarty Consultant Cardiac Anaesthetist Birmingham United Kingdom

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP) CORE SPC FOR HUMAN PLASMA DERIVED ANTITHROMBIN

FACTOR XIII DEFICIENCY IN PAKISTAN

Transcription:

in patients with inherited disorders of coagulation Ushma Jitendra Shah MB BS DNB EDA EDRA Madan Narayanan MB BS MD FRCA FCARCSI EDIC J Graham Smith BSc MB BCh MD FRCP FRCPath Matrix reference 1A02, 2A03, 2A05 Key points Congenital disorders of coagulation are uncommon but present significant challenges in the perioperative period. Multidisciplinary input and an individualized management plan depending on the type of disorder and surgery is essential to optimize outcome. Fresh-frozen plasma should not be used to replace clotting factors. Specific recombinant factor replacement for the inherited bleeding disorders and therapeutic anticoagulation for clotting disorders are the cornerstones of management. Recombinant activated factor 7 has revolutionized the management of haemophilia patients with inhibitors. Non-steroidal anti-inflammatory drugs and other drugs altering the coagulation are in general avoided. No specific guidelines exist for neuraxial techniques or peripheral nerve blocks in these patients. Risk benefit assessment on an individual basisiscrucial. Ushma Jitendra Shah MB BS DNB EDA EDRA Clinical Fellow Anaesthesia Department of Anaesthetics Frimley Park NHS Trust Camberley GU16 7UJ, UK Madan Narayanan MB BS MD FRCA FCARCSI EDIC Consultant in Anaesthesia and Intensive Care Department of Anaesthetics Frimley Park NHS Trust Portsmouth Road Camberley GU16 7UJ, UK, Tel: þ44 1276604161 E-mail: madan.narayanan@gmail.com (for correspondence) J Graham Smith BSc MB BCh MD FRCP FRCPath Consultant Haematologist Frimley Park NHS Trust and University of Surrey, Guildford Camberley GU16 7UJ, UK 26 Inherited bleeding (haemostatic) and clotting (thrombotic) disorders are rare but sometimes encountered by anaesthetists during emergency or elective surgeries. The perioperative management of these uncommon conditions can be challenging. In this article, we discuss the most common inherited coagulation disorders, their pathophysiology, and perioperative anaesthetic considerations. Inherited bleeding disorders The most frequent inherited bleeding disorders and their inheritance are summarized in Table 1. Of all the congenital bleeding disorders, haemophilia and von Willebrand s disease (VWD) are the most common. Pathophysiology and diagnosis Haemophilia Haemophilia can be classified as haemophilia A, B, or C depending on the deficiency of the coagulation factors VIII, IX, or XI respectively. Haemophilia A and B are inherited as X-linked recessive (XLR) disorders due to mutation in the long arm of chromosome X at F8 and F9 genes, respectively. 1 As with any XLR disorder, males are affected and females are carriers. One-third of the patients presenting with haemophilia have no family history. Factors VIII and IX mainly play an important role in the intrinsic pathway of the clotting cascade. These factors are required for thrombin generation and fibrin formation. The plasma concentration of factors VIII and IX can be expressed in IU ml 21 or as percentages of normal pooled plasma. 1 IU is the concentration of coagulation factor in 1 ml of normal pooled plasma. The normal value is 0.5 1.5 IU ml 21 or 50 150%. Diagnosis of haemophilia is usually suspected when bleeding symptoms occur spontaneously or after trauma. The risk of bleeding increases as the factor levels decrease (Table 2). Patients usually present with bleeding into the weight-bearing joints (knees, ankles and elbows), muscles and rarely the genitourinary system. The most common cause of death is intracranial bleed. The routine clotting screen may be normal and the only abnormality may be a prolonged activated partial thromboplastin time. The definitive diagnosis is made only by a factor level assay. Von Willebrand s disease VWD is the most common of inherited bleeding disorders. The prevalence of VWD is one in 100 but is asymptomatic in the majority of patients and is clinically significant in only one in 10 000 patients. 2,3 VWD is caused by either a quantitative or qualitative defect in von Willebrand s factor (VWF). VWF is a plasma glycoprotein which plays a vital role in platelet adhesion, aggregation, and also acts as a carrier for factor VIII and thereby decreasing its clearance from plasma. VWF is synthesized in bone megakaryocytes and vascular endothelium and stored in Weibel Palade bodies in the endothelial cells. Deficiency of VWF leads to easy bruising from trivial trauma; in particular, bleeding from mucosal surfaces, that is, epistaxis, gums, and bowel. Depending upon the type of VWD, patients can have prolonged bleeding after surgery. Of all blood groups, people with blood group O tend to have low VWF levels. VWD is classified by the International Society on Thrombosis and Haemostasis (ISTH) based on quantitative or qualitative defect of VWF into three types. 2 In type 1 VWD, there is a quantitative defect, whereas in type 2 VWD, there is a qualitative defect. Type 2 is further subclassified into four types (type 2A, 2B, 2M, 2N) depending upon the associated platelet binding and function, factor VIII binding doi:10.1093/bjaceaccp/mku007 Advance Access publication 24 April, 2014 Continuing Education in Anaesthesia, Critical Care & Pain Volume 15 Number 1 2015 & The Author [2014]. Published by Oxford University Press on behalf of the British Journal of Anaesthesia. All rights reserved. For Permissions, please email: journals.permissions@oup.com

Table 1 Inherited bleeding disorders. XLR, X-linked recessive; AD, autosomal-dominant; AR, autosomal-recessive; VWF, von Willebrand s factor Disorder Inheritance Primary defect Incidence Haemophilia A XLR Factor VIII deficiency 1/5000 male births Haemophilia B XLR Factor IX deficiency 1/25 000 male births Haemophilia C AD/AR Factor XI deficiency Von Willebrand s disease AD/AR VWF qualitative or quantitative defect 1/10 000 Rare bleeding disorders Factor II, V, VII, X, XIII deficiency AR,1/million Dysfibrinogenaemia AD,1/million Table 2 Grading of severity of haemophilia Haemophilia (A, B, or C) Mild Moderate Severe % activity of factors 5 40 1 5,1 Factor levels (IU ml 21 ) 0.05 0.40 0.01 0.05,0.01 capacity, and number of high molecular weight VWF multimers. In type 3 VWD, there is a complete absence of VWF. Type 1 VWD is a mild moderate bleeding disorder, whereas type 3 VWD is a severe bleeding disorder. VWD shows autosomal inheritance (dominant or recessive) depending on the subtype. Often the only abnormality is a prolonged bleeding time as normal screening tests do not rule out VWD (Table 3). In general, lab diagnosis of VWD is made with quantitative factor VIII, VWF, collagen binding assays, ristocetin cofactor activity, and multimeric analysis. The qualitative assays include tests such as glycoprotein binding assay, ristocetin cofactor activity (VWF:RCo), and ristocetin-induced platelet agglutination. 3 Anaesthetic considerations Preoperative assessment A detailed history about the type of haemophilia and VWD and its severity must be obtained. Prior information about response to DDAVP, use of recombinant factors VIII and IX, and previous transfusion of blood will be useful. A complete blood count, coagulation profile and fibrinogen level, and specific factor assays must be done if indicated. All patients must be evaluated for the presence of transfusion-related infections such as HIV and hepatitis B and C. Examination for the presence of joint deformities, contractures, and a thorough airway assessment. Preoperative assessment ideally must be done by a team of haematologist, surgeon, and anaesthetist, so that a tailored individual plan is formulated and discussed with the patient. General principles Multidisciplinary management comprising haematologist, surgeon, anaesthetist, physiotherapist, and occupational therapist. Liaison with laboratory services to ensure that appropriate factor concentrates are available and in sufficient quantity. Elective surgery scheduled early during the week and preferably in the morning. Table 3 Screening blood tests in haemophilia and von Willebrand s disease (VWD) Test Haemophilia VWD Platelet count Normal Normal or reduced Bleeding time Normal Normal or prolonged Prothrombin time Normal Normal Activated partial thromboplastin time Prolonged Normal or prolonged Preoperative clotting screen and specific factor assays depending on the type of bleeding disorder and preoperative transfusion of recombinant factors 30 60 min before the surgical procedure. Perioperative avoidance of mucosal trauma, i.m. injections, maintenance of normothermia, and pressure point care. Care with vascular access and invasive monitoring. Consider early use of ultrasound. Avoidance of tachycardia and hypertension. Risk benefits for neuraxial block and regional blocks need to be assessed individually and in general avoided. Early mobilization, consider mechanical deep venous thrombosis prophylaxis. Risk benefits of pharmacological methods must be considered and discussed with the surgeon and haematologist. Multimodal pain management, avoid non-steroidal antiinflammatory drugs. Specific management Haemophilia Before introduction of viral inactivation methods, the most common cause of death in haemophilliacs was due to transfusion of transmitted viral infections such as Hepatitis C and HIV. The general move over the years has been away from pooled plasma products to recombinant factors. Patients with haemophilia need 80 100% correction of their factor VIII before any major surgical procedure and this must be confirmed before surgery. Postoperatively, levels should be maintained for up to 6 weeks after orthopaedic procedures and 1 2 weeks for other procedures. 1,4,5 Factors and blood products (i) Recombinant factors VIII and IX: this is mainly helpful in haemophilia A and B. Commercial preparations of factors VIII Continuing Education in Anaesthesia, Critical Care & Pain j Volume 15 Number 1 2015 27

and IX as lyophilized powder are available. It is free from the risk of disease transmission as seen with blood products, but the limiting factor being its cost. The dosage of factors is as follows: Each FVIII unit per kilogram of body weight infused i.v. will raise the plasma FVIII level by 2% (t 1/2 factor VIII¼8 12 h). No of units of FVIII required¼weight of patient% factor level desired0.5. Each FIX unit per kilogram of body weight infused i.v. will raise the plasma FIX level 1% (t 1/2 factor IX¼18 24 h). No of units of FIX required¼weight of patient% factor level desired. Patients must be screened for the presence of inhibitors to factor VIII or IX. Depending upon the amount of inhibitors present, patients are classified as low risk (inhibitor level,5 Bethesda units ml 21 ) or high risk (.5 Bethesda units ml 21 ). The low-risk group requires higher dose of the deficient factor, but the high-risk group requires approved alternative regimens like recombinant activated factor VII (rfviia) or Factor Eight Inhibitor Bypassing Activity (FEIBA). (ii) Intermediate and high purity factor VIII concentrates: plasma derived, can have other non-factor VIII proteins, alternative when recombinant factors are not available. (iii) Cryoprecipitate: it is a source of factor VIII, VWF, factor XIII, and fibrinogen. It carries a risk of transmission of blood-borne infections and must be used only when recombinant factors are not available. Methylene blue-treated non-uk sourced cryoprecipitate is used for children,16 yr old. (iv) Prothrombin complex concentrate (PCC): it is a combination of blood clotting factors II, VII, IX, and X, protein C and S. Although the current main indication of PCC is for emergency reversal of warfarin therapy, PCC were initially developed for treatment of haemophilia B; they have been sparingly used after the development of highly purified and recombinant factors. Its thrombogenic calculated units are based on factor IX levels. The dosage is 15 50 units kg 21 (maximum 5000 units stat). Most PCC contain heparin to prevent activation of the factors. The plasma-derived activated PCCs (FEIBA) are licensed for use in patients with inhibitors to factor VIII and IX. Commercially available products include Beriplex w and Octaplex w and differ in the concentrations of the factors, manufacturing process, methods for viral inactivation, and dosage. (v) Recombinant factor VIIa: it can bind to the surface of activated platelets, thereby directly activating factor X and leading to an improved generation of thrombin. rfviia has been shown to be effective in achieving haemostasis in haemophilia patients with inhibitors in about 80% of cases. The thrombogenic activity of rfviia is optimized when fibrinogen levels and ph are within the normal range. It is also licensed for use in platelet dysfunctional disorders (Glanzman s thromboasthenia) and FVII deficiency. Fresh-frozen plasma is no longer used in perioperative management of haemophilia as it carries the risk of blood-borne infections and volume overload. Pharmacological options (i) Desmopressin (1-desamino-8-D-arginine vasopressin) or DDAVP: is an analogue of vasopressin hormone; it acts by releasing VWF which in turn forms a complex with factor VIII, thereby preventing its breakdown. It is mainly used in treatment of mild form of haemophilia A and VWD. DDAVP produces a two- to five-fold increase in factor VIII levels which may be sufficient in mild haemophilia for minor surgery. It is ineffective in severe haemophilia A or in haemophilia B. It is available as intranasal spray, oral or sublingual tablet, or i.v. preparation. The peak effect of i.v. dose is at 60 min and s.c./intranasal is 90 120 min. The i.v./s.c dose is 0.3 mgkg 21. Adult nasal spray dosage of 300 mg is equivalent to the standard i.v. dose of 0.2 mg kg 21. A dose of 300 mg for those over 50 kg and 150 mg for those up to 50 kg is suggested. DDAVP is not approved for use in children,2 yr or in pregnant women. (ii) Tranexamic acid: it is a synthetic derivative of amino acid lysine. Its antifibrinolytic action is due to competitive inhibition of conversion of plasminogen to plasmin which degrades fibrin. The plasma half-life is 2 h. The optimal dosage is unknown and has been successfully used in a wide dose range. It can be used orally (15 25 mg kg 21 8th hourly) or i.v. (10 mg kg 21 8th hourly). Conventional oral dosage is 3 g in three divided doses. It should be avoided in patients with haematuria due to obstructive uropathy. It is helpful in both haemophilia and VWD. Von Willebrand s disease The majority of cases of VWD do not need blood components to control haemorrhage, pharmacological management would suffice. In the high-risk subtypes, it is recommended that the VWF:RCo level is maintained at about 100 IU dl 21 perioperatively and.50 IU dl 21 in the immediate postoperative period. The FVIII plasma concentration should be above 100 IU dl 21 to cover major surgery and sustained above 50 IU dl 21 in the postoperative period. Major surgery requires treatment for 7 14 days and minor surgery for 1 5 days. 3 (i) DDAVP: it is approved for use in VWD type 1 and is of no use in type 3 VWD; its use in type 2 VWD must be discussed with a haematologist because of variable effect according to subtypes and its capacity to cause thrombocytopenia. DDAVP must be given at least 90 min before operation in the abovementioned doses. Further doses may not be beneficial due to rapid tachyphylaxis. DDAVP infusion can cause hypotension, facial flushing, and hyponatraemia. Consider fluid restriction. (ii) Antifibrinolytics: Tranexamic acid in the above mentioned doses may be useful. 28 Continuing Education in Anaesthesia, Critical Care & Pain j Volume 15 Number 1 2015

(iii) VWF/factor VIII concentrates: indicated in severe cases, type 3 VWD, and qualitative defects of VWF. These concentrates are derived from pooled plasma and are subjected to viral inactivation methods. Various preparations with different ratios of VWF/factor VIII:C and VWF:RCo levels are available (Humate P w and Alphanate SD/HT w ). Products like recombinant factor VIII which contain no VWF are not used in VWD. Venous thromboembolism (VTE) can occur with replacement therapy. 3 Allo-antibody formation occurs in 10 15% of type III VWD. In patients with inhibitors, there is very limited experience reported in the literature but recombinant VIIa (rviia) and continuous infusion of high doses of rfviii have been successfully used. (iv) Platelet infusions: should be considered if bleeding persists, despite DDAVP and replacement VWF concentrates. (v) Cryoprecipitate: unpredictable effect. Risk of transmissible infections. Should not be used for management of VWD unless other treatment modalities have failed. Special population and situations Regional anaesthesia Although there are guidelines about the use of regional anaesthesia in patients on anticoagulants, there are no clear-cut guidelines in patients with coagulation disorders. There are case series and reports of central neuraxial blocks with catheters in haemophiliac patients undergoing lower limb orthopaedic surgeries after correction of factor levels, but there are no randomized controlled trials. Therefore, the risk benefit ratio must be assessed individually on a case-to-case basis. The minimum safe factor levels and platelet count for neuraxial block in both the obstetric and general populations and evidence-based recommendations for neuraxial techniques in the setting of haemophilia and VWD cannot be offered. 6 Obstetrics patients with haemophilia Females are usually carriers as haemophilia is an XLR disorder. All female relatives of a patient with haemophilia must be screened to document FVIII level. Once the carriers are identified, they must be educated and counselled about the risk of having a child with haemophilia and other reproductive options. Such patients should be managed with a multidisciplinary team right from the antenatal period. Sex of the child and therefore the haemophilia status may be identified by polymerase chain reaction around 10 weeks of gestation or by ultrasound around 18 20 weeks. Factor VIII and IX levels must be measured immediately before delivery and postpartum with an aim to maintain above 0.5 IU ml 21. Controversies exist about the mode of delivery. Instrumental delivery poses a high risk of intracranial haemorrhage in a fetus with confirmed haemophilia status. Elective Caesarean section must be debated individually according to the risk to mother and child taking into consideration the haemophilia status of the fetus. There is no contraindication to normal vaginal delivery; however, prolonged labour must be avoided. There is no evidence that routine ultrasound screening or prophylactic administration of clotting factors reduces the risk of intracranial haemorrhage in the newborn. Labour analgesia The risk of neuraxial analgesia must be assessed individually after checking platelet and factor VIII levels (.0.5 IU ml 21 ). Other options need to be considered like remifentanil patient controlled analgesia (PCA) with continuous monitoring of heart rate and oxygen saturation. Von Willebrand s disease In pregnancy, VWF begins to increase from 6 weeks of gestation and increases by three- to four-fold by the third trimester. Levels of VWF must be assessed at 34 36 weeks in type 1 and 2 VWD. VWF activity (VWF:RCo).40 IU dl 21 is required for safe vaginal delivery and.50 IU dl 21 for Caesarean section. 3 Post-Caesarean section patients usually need treatment with DDAVP or VWF concentrates for a week to maintain haemostasis. Similarly, patients with post-partum haemorrhage must receive active management with suitable replacement therapy. Monitoring of VWF and factor VIII must be done within 24 h post-partum as levels of VWF decline rapidly back to baseline. Labour analgesia Epidural analgesia may be considered in type1 VWD after thorough evaluation of coagulation profile and VWF levels. The risk benefit ratio must be assessed and must be performed by a senior anaesthetist. Epidural anaesthesia is not recommended in type 2 or 3 VWD. Other recessively inherited coagulation disorders Deficiency of fibrinogen, prothrombin, clotting factors V, VII, X, XI, and XIII are recessively inherited and are very rare with prevalence ranging from one in two million for factor II (prothrombin) and factor XIII (FXIII) deficiency to one in 500 000 for factor VII (FVII) deficiency. Replacement factors such as fibrinogen concentrates (RiaSTAP w ), rfviia (NovoSeven w ), and F XIII (Corifact w ) are available and must be considered along with the use of PCC and cryoprecipitate. Inherited clotting disorders (thrombophilias) Thrombotic and embolic events cause significant mortality and morbidity. Thrombotic tendency can be caused by either inherited or acquired defects in the clotting cascade. Venous thrombosis has an annual incidence of one in 1000. Deficiencies of antithrombin, protein C, and protein S account usually for,5% of venous thrombosis in all age groups. The inherited thrombophilias are listed in Table 4. 7 Continuing Education in Anaesthesia, Critical Care & Pain j Volume 15 Number 1 2015 29

Table 4 Inherited thrombophilias Abnormality Factor Inheritance Life time probability of thrombosis Excess procoagulants Factors VII, VIII, IX, XI Abnormal cofactors (mutations) Factor V Leiden 2.2 Prothrombin Dysfibrinogenaemia Deficiency of anticoagulants Antithrombin (AT) AD 8.1 Protein C AD 7.3 Protein S AD 8.5 Abnormalities of fibrinolysis tpa (tissue plasminogen activator) Plasminogen PAI (plasminogen activator inhibitor) Other allied inherited conditions Hyperhomocysteinaemia Antithrombin deficiency Antithrombin functions primarily by inactivating thrombin and activated factor X and secondarily by inactivating factors VII, IX, and XII. The normal levels of antithrombin are 70 132% and,60% results in thrombosis. Protein C and protein S deficiency Protein C mainly functions by inactivating activated factors V and VIII and stimulating plasminogen activator. Protein S is a cofactor of protein C, which enhances its action. The normal levels of protein C and protein S are 70 164% and 63 160%, respectively, and any level below the normal is thrombotic. Factor V Leiden mutation Factor V mainly acts as a cofactor for activated factor X. Activity of factor V is limited by activated protein C which degrades it. The factor V Leiden mutation results in resistance to activated protein C thereby causing thrombosis. Abnormalities of fibrinolytic system In normal subjects, plasminogen is converted to plasmin which lyses clots. It is mainly activated by tissue plasminogen activator (tpa) and inhibited by plasminogen activator inhibitor (PAI). Deficiency of plasminogen, tpa, or excess of PAI may lead to a hypercoagulable state (Table 5). Qualitative defects in fibrinogen (dysfibrinogenaemia) may also lead to thrombosis. Treatment Anticoagulants such as unfractionated heparin, low molecular weight heparin (LMWH) and warfarin are the main stay of treatment. For specific conditions, replacement factors are available like cryoprecipitates, protein C, and antithrombin. Fresh-frozen plasma is rarely used as a source of anticoagulants and may be used in the case of their deficiency. Consideration should be given to preoperative placement of inferior vena cava (IVC) filters in high-risk patients undergoing high-risk surgery with anticipated prolonged immobility. Table 5 Diagnosis Defect Preoperative assessment A detailed coagulation screen must be done to evaluate the diagnosis and to monitor treatment as patients are on anticoagulation. The decision to administer fresh-frozen plasma before surgery must be done in consultation with the haematologist. (Fresh-frozen plasma is only used for factor V deficiency and plasma exchange in patients with thrombotic thrombocytopenic purpura and haemolytic uraemic syndrome.) General principles Tests Dysfibrinogenaemia Prolonged thrombin time (normal: 18 22 s) Fibrinolysis defect Prolonged euglobulin clot lysis time (normal: 90 240 min) Anticoagulant deficiency Normal thrombin time and euglobulin (protein C, protein S, antithrombin) clot lysis time As for bleeding disorders, multidisciplinary team management, elective scheduled surgery, ultrasound for vascular access, and multimodal analgesia. Preoperative clotting screen/anti-xa levels depending on the medication used to bridge perioperative anticoagulation. Bridging therapy with unfractionated/lmwhs for high-risk patients. Avoid dehydration and promote early mobilization, use of mechanical compression devices. Anaesthetic considerations Choice of general anaesthesia vs regional anaesthesia should be individualized as per patient and surgery. Guidelines exist for regional anaesthesia in a patient on anticoagulation by American and European societies of regional anaesthesia. In high-risk patients, warfarin must be converted to heparin before surgery with monitoring of coagulation. The use of mechanical measures like graduated 30 Continuing Education in Anaesthesia, Critical Care & Pain j Volume 15 Number 1 2015

elastic stockings and intermittent pneumatic compression devices must be used. A plan for restarting anticoagulants after surgery must be discussed with the surgeons and haematologist. Conclusion Patients with inherited coagulation disorders can have the same perioperative outcome as a normal patient if care is taken to maintain the deficient factor levels in bleeding disorders or time anticoagulation in clotting disorders in the perioperative period. Also the availability of recombinant factor VIIa has revolutionized treatment in complicated haemophilia patients with inhibitors. Declaration of interest None declared. References 1. Fijnvandraat K, Cnossen MH, Leebeek FWG, Peters M. Diagnosis and management of haemophilia. Br Med J 2012; 344: e2707 2. Sadler JE, Mannucci PM, Berntorp E et al. Impact, diagnosis and treatment of von Willebrand disease. Thromb Haemost 2000; 84: 160 17 3. Pasi KJ, Collins PW, Keeling DM et al. Management of von Willebrand disease: a guideline from the UK Haemophilia Centre Doctors Organization. Haemophilia 2004; 10: 218 31 4. Martlew VJ. Peri-operative management of patients with coagulation disorders. Br J Anaesth 2000; 85: 446 55 5. DiMichele D, Neufeld EJ. Hemophilia. A new approach to an old disease. Hematol Oncol Clin North Am 1998; 12: 1315 44 6. Choi S, Brull R. Neuraxial techniques in obstetric and non-obstetric patients with common bleeding diathesis. Anesth Analg 2009; 109: 648 60 7. Khan S, Dickerman JD. Hereditary thrombophilia. Thromb J 2006; 4: 15 Please see multiple choice questions 17 20. Continuing Education in Anaesthesia, Critical Care & Pain j Volume 15 Number 1 2015 31