Hemophilia TREATMENT HK

Embed Size (px)

DESCRIPTION

PTCB

Citation preview

  • TAILORING PROPHYLAXIS AND TREATMENT OF

    HEMOPHILIA SANDEEP DEVABHAKTHUNI, PHARM.D.

  • 1

    TAILORING PROPHYLAXIS AND TREATMENT OF HEMOPHILIA

    ACTIVITY DESCRIPTION Patients with hemophilia and their health care providers

    often search for a treatment solution that is just right. The

    most effective prophylaxis protocols are tailored to the

    individual based on many factors (such as age, bleeding

    patterns, joint health and levels of physical activity). The

    science of treating hemophilia continues to improve;

    getting the art of individualizing treatment continues to be

    a challenge. As the bridge between patients and

    physicians, pharmacists are in the position to play an

    integral part of a multi-pronged solution to this challenge

    of individualizing treatment. Because pharmacotherapy

    and knowledge of a patients pharmacokinetics play a

    prominent role in individualizing treating of hemophilia, it

    is also a perfect opportunity for the pharmacist to be

    involved. This program will satisfy the education need by

    creating a program for pharmacists that will enhance their

    understanding of hemophilia, pharmacotherapy,

    counseling points, and information needed to work with

    the patient to maximize the benefits of medications, limit

    side effects and identify drug-drug or drug-disease

    interactions.

    TARGET AUDIENCE

    The target audience for this activity is pharmacists,

    pharmacy technicians and nurses in hospital,

    community, and retail pharmacy settings.

    LEARNING OBJECTIVES After completing this activity, the pharmacist and nurse

    will be able to:

    Outline the many factors such as age, bleeding

    patterns, joint health and levels of physical

    activity) that must be considered when tailoring

    prophylaxis protocols for individual treatment

    Review the current and emerging

    pharmacological approaches to the management

    of hemophilia (pharmacologic profiles, efficacy,

    side effects, & adverse events)

    Describe the role pharmacists can play in

    counseling hemophiliac patients on lifestyle

    changes, drug treatment strategies and

    medication adherence to improve quality of life

    After completing this activity, the pharmacy technician

    will be able to:

    List symptoms of hemophilia

    List treatments available for hemophilia

    ACCREDITATION

    PHARMACY

    PharmCon, Inc. is accredited by the

    Accreditation Council for Pharmacy

    Education as a provider of continuing

    pharmacy education.

    NURSING

    PharmCon, Inc. is approved by the California Board of

    Registered Nursing (Provider Number CEP 13649) and

    the Florida Board of Nursing (Provider Number 50-

    3515). Activities approved by the CA BRN and the FL

    BN are accepted by most State Boards of Nursing.

    CE hours provided by PharmCon, Inc. meet the ANCC criteria

    for formally approved continuing education hours. The ACPE

    is listed by the AANP as an acceptable, accredited continuing

    education organization for applicants seeking renewal

    through continuing education credit. For additional

    information, please visit

    http://www.nursecredentialing.org/RenewalRequirements.aspx

    Universal Activity No.: 0798-0000-14-191-H01-P&T

    Credits: 2 contact hours (0.2 CEU)

    Release Date: December 15, 2014

    Expiration Date: December 15, 2016

    ACTIVITY TYPE

    Knowledge-Based Home Study Monograph

    FINANCIAL SUPPORT BY

    Baxter

  • 2

    ABOUT THE AUTHOR

    Dr. Sandeep Devabhakthuni is an Assistant Professor in

    the Department of Pharmacy Practice and Science at

    the University of Maryland School of Pharmacy. He

    graduated with a Bachelor of Engineering in Biomedical

    Engineering degree from University of Pittsburgh

    School of Engineering and a Doctor of Pharmacy

    degree from the University of Pittsburgh School of

    Pharmacy. He then completed his pharmacy practice

    residency at the University of Maryland Medical Center.

    He also completed his specialty residency in Cardiology

    and Critical Care at the University of Pittsburgh Medical

    Center. Currently, Dr. Devabhakthuni is a board certified

    pharmacotherapy specialist at the University of

    Maryland Medical Center, and he has a clinical practice

    on the Cardiology and Medical Intensive Care services.

    Sandeep Devabhakthuni, PharmD, BCPS

    Assistant Professor, University of Maryland

    School of Pharmacy

    FACULTY DISCLOSURE

    It is the policy of PharmCon, Inc. to require the

    disclosure of the existence of any significant financial

    interest or any other relationship a faculty member or

    a sponsor has with the manufacturer of any

    commercial product(s) and/or service(s) discussed in

    an educational activity. Sandeep Devabhakthuni

    reports no actual or potential conflict of interest in

    relation to this activity.

    Peer review of the material in this CE activity was

    conducted to assess and resolve potential conflict of

    interest. Reviewers unanimously found that the

    activity is fair balanced and lacks commercial bias.

    Please Note: PharmCon, Inc. does not view the existence of

    relationships as an implication of bias or that the value of

    the material is decreased. The content of the activity was

    planned to be balanced and objective. Occasionally,

    authors may express opinions that represent their own

    viewpoint. Participants have an implied responsibility to use

    the newly acquired information to enhance patient

    outcomes and their own professional development. The

    information presented in this activity is not meant to serve

    as a guideline for patient or pharmacy management.

    Conclusions drawn by participants should be derived from

    objective analysis of scientific data presented from this

    monograph and other unrelated sources.

  • 3

    Introduction

    Hemophilia A and B are rare congenital bleeding disorders caused by a deficiency or

    absence of coagulation factor VIII (FVIII) or factor IX (FIX), respectively.1 Hemophilia is a genetic

    disorder that affects over 400,000 people worldwide with a majority of them as males.2

    Hemophilia A is the most common form of hemophilia, counting for 80-85% of cases. Because

    there is no cure for these X-linked disorders, appropriate management is necessary to avoid

    devastating consequences including crippling arthropathy. The severity of the disorder is

    typically characterized by the residual endogenous FVIII/FIX concentrations. Patients with a

    factor level of < 0.01 IU/mL are classified as severe hemophiliacs and represent about half of

    diagnosed cases. Patients with factor levels between 0.01-0.05 IU/mL and > 0.05 IU/mL have

    moderate and mild hemophilia, respectively. While the bleeding phenotype may be

    heterogeneous even in severe hemophilia, this classification by FVIII/FIX concentrations

    correlates with the severity of clinical symptoms, with spontaneous joint and muscle bleeds

    being largely confined to patients with severe hemophilia.3

    Hemophilia A and B are difficult to differentiate from clinical presentation.

    Replacement of hemostatic concentrations of the deficient factor is the mainstay of treatment

    for bleeding episodes according to type and severity of bleeds. Without proper management,

    patients can experience recurrent joint bleeds, leading to mobility problems, which was the

    classic progression of this disease prior to 1970s when coagulation factors were not yet

    available.4 Prior to development of coagulation factors, mortality was extremely high, and the

    life expectancy of people with hemophilia was lower when compared to the general

    population.5 Then, the discovery of cryoprecipitate and lyophilized formulations of factor

  • 4

    concentrates served as a catalyst for the more effective hemophilia replacement therapy. Also,

    these innovative strategies allowed for increased patient convenience including the possibility

    of home therapy and treating joint bleeds as soon as possible. This also led to significant

    reduction in potential complications and improved the quality of life for patients with

    hemophilia.

    In the late 1970s and early 1980s, there was a huge concern when widespread blood-

    borne virus transmission occurred due to use of pooled plasma in manufacturing of factor

    concentrates. This event triggered a closer inspection to ensure safety of treatment for a

    population that is at a higher risk of fatal complications. This led to the development of viral

    inactivation techniques for the production of plasma-derived factor concentrates. Then,

    recombinant gene technology and protein purification were implemented, which led to the

    creation of highly purified recombinant FVIII and FIX products, which have become the first-line

    agents for correction of factor deficiencies associated with hemophilia.6 These advances in

    management significantly improved treatment for hemophilia patients and contributed to the

    increased use of primary prophylaxis, where regular infusion of factor concentrates are

    administered to prevent bleeding and resulting joint damage. With the development of

    strategies to improve viral safety, the most serious and challenging complication of treatment is

    the risk of inhibitory alloantibodies.7

    With recent technological advances, patients with hemophilia may now receive optimal

    treatment and can achieve excellent quality of life if effective approaches are used to provide

    multidisciplinary comprehensive care. The objectives of this review are to address current

  • 5

    advances and emerging pharmacological approaches for treatment of hemophilia and to

    describe ongoing issues and importance of multidisciplinary comprehensive care.

    Clinical Assessment of Hemophilia

    Symptoms of hemophilia can range from mild to severe depending on the amount of

    clotting factors present in blood. In general, patients with hemophilia bleed for a longer period

    of time compared to healthy people because of coagulation factor deficiency. Common

    symptoms can include large bruises, spontaneous bleeding from gums or nose, pain or

    tightness in joints, and blood in stool or urine. Severe symptoms usually involve bleeding into

    the joints, brain, or internal organs or substantial bleeding after injury or surgery that could

    potentially be fatal. Furthermore, bleeding in the joints can develop into swelling that can lead

    to breakdown of cartilage, resulting in chronic pain and immobility that can be permanent.8

    An accurate diagnosis is essential to ensure that a patient receives the appropriate

    treatment since different bleeding disorders may have very similar symptoms. A correct

    diagnosis can only be made with the support of comprehensive coagulation laboratory testing.

    This testing helps clinicians to understand the clinical features of hemophilia. Using screening

    tests can identify potential causes of bleeding. For patients with hemophilia, the coagulation

    tests are characterized by a normal prothrombin time (PT), bleeding time (BT), and platelet

    count, but the activated partial thromboplastin time (aPTT) is prolonged. The reason aPTT is

    prolonged is because this test measures the intrinsic pathway for the coagulation cascade,

    whereas PT measures the extrinsic pathway. The intrinsic pathway requires adequate sources

    of both factors VIII and IX to function properly. Since either hemophilia type A or B is

  • 6

    characterized by a deficiency in one of these factors, this leads to problems with the intrinsic

    pathway, which ultimately can result in bleeding consequences.8

    In addition to evaluating coagulation tests, assessment of factor assays is needed to

    confirm diagnosis since a deficiency in a coagulation factor can guide clinicians in determining

    the type of hemophilia. The severity of the disease is correlated with the degree of the

    deficiency, and the classification of hemophilia severity by the factor concentration is shown in

    Table 1.8 The most common hindrance in hemophilia treatment is the production of inhibitors,

    or antibodies against injected coagulation factor replacement, which can occur up to 20% of

    patients.9 In hemophilia patients with suspected inhibitors, testing can be performed using a

    Bethesda titer to determine presence of antibodies to specific clotting factors.

    Preventive and Supportive Measures for Hemophilia

    When managing a patient diagnosed with hemophilia, the goals of therapy include

    promotion of adequate hemostasis with minimal side effects with deficient clotting factor,

    prevention of viral transmission, promotion of hemostasis in the presence of inhibitors, and

    optimizing patient adherence by considering cost and ease of use. Because early recognition is

    crucial to prevent mobility complications, patients need to be educated on signs/symptoms of

    bleeding, injury avoidance, prompt self treatment, and need for immunizations against

    Hepatitis A and B prior to receiving replacement coagulation factors. Acute bleeds should be

    treated as quickly as possible, preferably within two hours.8 Most patients should be counseled

    on home treatment since this strategy can improve quality of life due to less pain and disability,

    fewer hospitalizations, and decreased time away from work or school.10 In addition, patients

  • 7

    should be prepared for any surgical interventions by maintaining factor levels of at least 0.5-0.7

    units/mL (50-70%). Dental extraction in hemophiliacs is also associated with a high risk of

    bleeding and requires a multidisciplinary approach and stringent protocol.11

    Besides considering preventative strategies, patients may require supportive therapy

    depending on the severity of hemophilia. If patients have significant swelling in their joints, this

    may lead to either acute pain from bleeding or chronic pain from joint damage due to cartilage

    destruction. Adequate assessment of the cause of pain is necessary to guide proper

    management. Hemophilia patients can experience pain due to venous access, joint or muscle

    bleeding, operation, or chronic hemophilic arthropathy. For appropriate management of pain,

    clinicians can consider corticosteroids, acetaminophen, and narcotics.12 For chronic hemophilic

    arthropathy, cyclooxygenase-2 (COX-2) inhibitors have a greater role in management. If pain is

    disabling, orthopedic surgery may be indicated.13-16

    Treatment Options for Hemophilia

    The types of treatment methods available today are plasma-derived products,

    recombinant coagulation factors, and gene therapy. Dosing for coagulation factors is based on

    volume of distribution (both intravascular and extravascular compartments), half-life, and

    factor level required for hemostasis. The available plasma-derived products or recombinant

    clotting factors are listed in Table 2. Patients who develop an immune response to therapy or

    have acquired hemophilia are extremely difficult to manage.17 The treatment options for this

    complication include prothrombin complex concentrates, activated recombinant factor VII

    therapies, and immunosuppressive medications.

  • 8

    Plasma-Derived Coagulation Factors

    Plasma-derived coagulation factors in whole blood and in plasma fractions are used as

    replacements for any absent factors in hemophiliacs.18 Available plasma products include fresh

    frozen plasma, freeze-dried concentrates, and cryoprecipitate (slowly thawed plasma

    precipitate).19 Transfusions with healthy blood were the earliest successful treatment of

    hemophilia. Blood transfusions provide the patient with missing clotting factors and replenish

    lost blood volume during bleeds. However, these products need to be used repeatedly in order

    to replenish live tissue cells.

    Treatment with these products requires suitable methods to cleanse blood and plasma

    from pathogens. For plasma-derived products, the most common methods for purification

    include moderate dry heating, strong dry heating, and wet heating of blood products. The use

    of these methods decreased the incidence of Human Immunodeficiency Virus (HIV) and

    Hepatitis C transmission, which are deadly blood-borne pathogens.17 In the 1980s, 60-80% of

    patients with severe hemophilia contracted HIV from blood-derived products. Most of the

    viruses commonly transmitted through blood transfusions have a long, asymptomatic carrier

    state, which is problematic since healthy donors may actually be carriers of a pathogen.18

    Standard screenings have been placed to remove blood samples with these viruses, but

    there still is a potential risk for transmission. Viruses can evolve into different strains or

    different pathogens, which may not be detected by routine techniques. So there is a potential

    threat of emerging viruses because a pathogen can make contact with a new species or

    population and establish itself in that vulnerable population, like patients with hemophilia.

  • 9

    Thus, there have been improvements made in detection methods such as nucleic-acid

    screening and incorporation of products that reduce viral activity, making blood products safe

    from HIV and Hepatitis B and C.18 There is an increased focus on the use of reagents that are

    completely independent of human plasma.18 Another concern with plasma-derived factors is

    the possibility of an adverse immune reaction, leading to decreased efficacy with repeat

    administration.17

    Blood products are generally cost effective; yet, proper purification techniques are not

    easily accessible in some third-world countries, which increases the risk of viral transmission

    and causes complications in hemophilia patients. Approximately 80% of hemophiliacs continue

    to use plasma-derived factors due to limited resources.20

    Recombinant Coagulation Factors

    Recombinant coagulation factors treatments deliver clotting factors that hemophilia

    patients are missing and have become the first-line agents for management of acute bleeds.17,21

    The first generation recombinant factors were derived from DNA by using albumin in the

    synthetic steps. However, albumin was not included in the final product, which decreases the

    risk of hypersensitivity reactions. Newer recombinant factors have been produced that use no

    human proteins in the synthetic or final stages of production. Because of this process, these

    factors are though to be safer in terms of viral transmission compared to plasma-derived

    factors; however, the risk is not completely eliminated.17 The recovery time for patients using

    recombinant factors has been shown to be slower than those for plasma-derived factors.22

  • 10

    A single dose of recombinant factors can eliminate 80% of uncontrolled bleeds and

    subsequent use increases the success rate to 90-95%.17 Recombinant factors are considered as

    effective as plasma-derived coagulation factors. In a study of 95 children with moderate to

    severe hemophilia A, recombinant FVIII for average of 1.5 years was given in response to

    excessive bleeds and prior to surgical/dental procedures. The average number of transfusions

    needed was 34.9 infusions per individual, with effective response and minimal side effects

    reported.23 Another study suggested a different method of administration, which was

    continuous injection of clotting factor to prevent highs and lows in coagulating factor level.

    This promotes homeostasis and stops large bleeds before they occur. This method also reduces

    the overall amount of factor required for treatment.24

    Several factors are considered when determining appropriate dosing for recombinant

    coagulation factors, including site of bleeding, volume of distribution, half-life, and joint health.

    Dosing for factor replacement is dependent on site of bleed, which helps to determine percent

    correction to target factor concentration that is needed as well as duration. Guidelines for

    factor replacement based on site of bleed and need for prophylaxis for surgery are provided in

    Table 3. Specific dosing instructions for recombinant FVIII and FIX products are provided in

    Table 4. Because recombinant FVIII concentrates are larger molecules, the volume of

    distribution is 0.5 so this requires half the amount needed for appropriate recombinant FIX

    concentrate replacement. Recombinant FIX concentrates also have a longer half-life and

    require less frequent dosing compared to recombinant FVIII products. These recombinant

    concentrates should be infused slowly by intravenous injection at a rate not to exceed 3

    mL/min in adults and 100 units/min in young children.

  • 11

    Administration of recombinant factor concentrates requires close monitoring.

    Important efficacy parameters include plasma factor levels 15 minutes after infusion completed

    to verify accuracy of calculated dose and control of bleeding. If the target factor concentration

    is not achieved with appropriate dosing, then testing for inhibitor development is warranted.

    For monitoring of safety, patients should be educated on the possibility of hypotension,

    injection site reaction/pain, dyspnea, hypersensitivity, and thrombosis (more common with

    recombinant FIX products). In addition, recombinant FIX concentrates have an increased risk

    for hypersensitivity reaction and disseminated intravascular coagulopathy in patients who have

    liver disease, are undergoing surgery, or are neonates.25,26

    The main concern with recombinant factors is the development of inhibitors

    (approximately 28-33%).25,26 For hemophiliac patients, a normally functional coagulation factor

    is deficient. So the bodys immune system will see a recombinant product derived from foreign

    DNA as a pathogen. Thus, an immune response is mounted to destroy and remove the infused

    coagulating factor.27 If not recognized, hemophilia patients with inhibiting antibodies could

    have an increased bleeding risk that is potentially fatal.28 The reported incidence of inhibitor

    development has been variable due to different study designs. A study by Knobe and

    colleagues tested 116 people with hemophilia for presence of inhibitors after factor

    replacement treatment that last 14-16 days. Of these people, 19% of hemophilia A patients

    developed inhibitors compared to 37% of hemophilia B patients. The study suggested that

    inhibitor development could be genetically related because all patients who had inhibitors were

    found to have impaired protein synthesis due to a mutation.29

  • 12

    Despite recombinant factors being more expensive than plasma-based factors by 20-

    50%, these factors are used by 60-70% of severe hemophiliacs in the United States and all

    patients in Canada and Ireland use recombinant factors over plasma-derived products.

    Increased cost is likely due to amount of coagulation factor that can be extracted from a blood

    sample is only 5-10% of the quantity of factor present in the sample.17 Although dosing

    patterns will vary, a typical individual with hemophilia receiving primary prophylaxis will need

    approximately 2000 IU of clotting factor 3 times per week with the average cost per dose

    ranging from $1,000 to $2,000. This translates into an approximate monthly cost of $12,000 to

    $24,000 or $1444,000 to $288,000 per year in medication expenses alone.30 The discrepancy in

    price is the reason why it is challenging to provide recombinant factors in developing

    countries.31 Methods to decrease amount needed for replacement such as factor clearance

    receptor antagonist, continuous infusion of product, and increased half-life of recombinant

    factor may lead to substantial healthcare cost savings.17,24 Possible solutions to prevent

    inhibitor development include re-engineering the recombinant factor so that it is less likely to

    induce an immune response.32

    Adjunctive Therapies for Hemophilia

    While coagulation factor replacement therapy is the mainstay treatment for hemophilia

    patients, there are adjunctive therapies that can be used depending on the type and severity of

    hemophilia. Desmopressin is a vasopressin synthetic analog that causes release of von

    Willebrand factor and factor VIII, which makes it suitable for treatment of hemophilia A only.33

    Desmopressin is frequently used for treatment of mild or moderate bleeding episodes in

    patients with hemophilia A with a good response rate of 80-90%. It can be given either

  • 13

    intravenously by administering 0.3 mcg/kg in 50 mL of 0.9% sodium chloride over 15-30

    minutes or intranasally with 150 300 mcg per dose (1 spray = 150 mcg). Desmopressin can be

    given daily for 2-3 days, but a 30% lower response is expected with second dose due to

    tachyphylaxis.34,35 Major adverse effects of treatment include flushing (most common),

    thrombosis (rare), headaches, tachycardia, and hypotension. Because of its antidiuretic effect,

    desmopressin also promotes fluid retention, which can cause profound hyponatremia so it

    should be used cautiously in patients with heart failure experiencing fluid overload or existing

    hyponatremia.34,35

    Antifibrinolytic therapy can be used as adjunctive therapy for procedures expected to

    cause mucosal bleeding. This type of therapy prevents fibrin breakdown by inhibiting

    fibrinolytic enzymes found in saliva. Aminocaproic acid can be given as oral or intravenous dose

    of 100 mg/kg (maximum of 6 grams) every 6 hours. Aminocaproic acid is used less often due to

    short half-life, low potency and potential for toxicity. Tranexamic acid is 10 times more potent

    compared to aminocaproic acid so it can be administered as 25 mg/kg (maximum 1.5 grams)

    orally every 8 hours or 10 mg/kg (maximum 1 gram) intravenously every 8 hours.36,37 Either

    agent is usually given for 7 days following dental extractions to prevent post-operative

    bleeding. They are contraindicated in hematuria since they may cause serious obstructive

    uropathy and should be avoided in combination with prothrombin complex concentrates due to

    additive risk of thromboembolism. These agents can cause thrombosis, headache, renal failure

    (requiring dose adjustments), and hypotension. Aminocaproic acid also can cause

    rhabdomyolosis, and tranexamic acid has been associated with visual disturbances and

    increased incidence of anaphylaxis.36,37

  • 14

    Another adjunctive therapy for patients with Hemophilia B is the use of prothrombin

    complex concentrates (PCCs). These products contain non-activated factors II, VII, IX, and X.

    Activated PCCs contain greater quantities of the activated factors. However, PCCs are not used

    as first-line for management of patients with Hemophilia B because of lower purity and risk of

    thrombosis. The risk of thrombosis is higher in patients with hepatic disease, neonates, and

    patients experiencing crush injury or major surgery. Other adverse effects include dizziness,

    nausea, hives, flushing, headaches, and hypersensitivity reactions. Other adjunctive therapies

    include fresh frozen plasma and cryoprecipitate, but neither of these agents is recommended

    routinely due to concerns about safety and quality.

    Prophylaxis versus On-Demand Replacement Therapy

    Current factor treatment regimens include on-demand treatment, which is infusing

    clotting factor when a bleed occurs, or prophylaxis, where factors are infused to prevent bleeds

    by maintaining levels of FVIII or FIX at appropriate levels. The use of prophylaxis is intended to

    prevent bleeding episodes through the administration of regular infusions. The frequency of

    prophylactic infusion depends on several factors and is determined by the patient and primary

    care provider. The prophylaxis options are provided in Table 5. Several studies have

    demonstrated that prophylaxis therapy gives children the best chance to reach adulthood

    without damage to their joints.38-40 If patients have recurrent joint bleeds, this can lead to

    severe and debilitating injuries that often require physical therapy. Also prophylactic

    administration can convert severe hemophilia into milder form with much lower incidence of

    chronic arthropathy.

  • 15

    To prevent bleeding and joint destruction by preserving normal musculoskeletal

    function, the typical goal of infusion is to maintain at minimum of 0.01 units/mL (1%). Studies

    have shown that this can still prevent joint bleeds even if the goal cannot be achieved.38-40 For

    FVIII and FIX, the most common regimens studied are 25 50 units/kg three times weekly and

    40-100 units/kg twice weekly, respectively. These regimens have been proven to be cost-

    effective long-term because they eliminate the high cost associated with subsequent

    management of damaged joints and improves quality of life.30,41 Primary prophylaxis is typically

    started before 2 years of age with almost no previous history of bleeding episodes and normal

    joint evaluations. However, this type of prophylaxis is not widely accepted because of high

    cost, inconvenience to families leading to noncompliance, and risk of infection or thrombosis

    with central venous access. Secondary prophylaxis after significant joint bleeding is more

    common and is associated with significant reduction in number of recurrent episodes; however,

    radiological evidence of joint disease rarely improves and often progresses despite

    prophylaxis.41 The National Hemophilia Foundations Medical and Scientific Advisory Council

    recommend that prophylaxis be considered optimal therapy for individuals for severe

    hemophilia A or B. Prophylactic therapy should be instituted early (prior to onset of frequent

    bleeding) with the aim of keeping FVIII/FIX level above 1% between doses.42

    Treatment Options for Hemophilia Patients with Inhibitors

    One of the most serious complications of hemophilia is the development of inhibitors or

    neutralizing antibodies to the infused clotting factor. In some individuals with hemophilia A,

    the factor product used to prevent or treat bleeds is viewed as a foreign body. This results in an

    immune reaction by the body to make that infused clotting factor inactive and harmless to the

  • 16

    system, which leaves the individual unprotected from bleeds. Inhibitors develop in about 30%

    of patients with severe Hemophilia A and up to 5% of those with hemophilia B.30 Individuals

    with inhibitors are certainly at higher risk for serious bleeding episodes and significant joint

    damage.

    There are numerous risk factors for the development of inhibitors that have been

    identified. These risk factors include age, race, type of hemophilia, presence of other immune

    disorder, and frequency and dose of factor. High-intensity treatment with factor replacement

    is a major risk factor for inhibitor development. Also, choosing continuous infusion of clotting

    factor over bolus dosing can increase the risk. Other risk factors include surgical procedure

    during first 50 exposure days, severe hemophilia A, family history, certain FVIII and FIX genetic

    mutations, and other genetic factors (African American, Asian, and Hispanic ethnicity).43

    Inhibitor development is more common during the first year of treatment, but it can occur at

    anytime. Inhibitor development should be suspected with decreased clinical response to factor

    replacement, and lab testing should be considered in this situation.

    When hemophilia patients develop inhibitors, treatment goals are to treat acute

    bleeding episodes and eradicate inhibitor development if possible.44-46 The decision to treat

    hemophilia patients with inhibitors is dependent on whether they are having an active bleed. If

    the patients are bleeding, then treatment strategies are dependent on the degree of inhibitor

    development. If a low titer < 5 BU is measured, then high-dose factor replacement is the best

    option, which would require 2-3 times the usual replacement doses more frequently. However,

    if a high titer > 5 BU is measured, then other alternative strategies are warranted, which include

  • 17

    the use of the use of PCCs, recombinant FVIIa concentrate, or porcine FVIII concentrate (for

    patients with hemophilia A only).45,46

    For hemophilia patients with high titers of inhibitors, activated PCCs will be more

    effective since PCC includes only trace amounts of FVIII and larger amounts of FIX, which is

    more helpful in patients with hemophilia B. Activated PCCs also contain FVIII Inhibitor

    Bypassing Activity, which activates the synthesis of thrombin by stimulating prothrombinase,

    which bypasses the synthesis of FIX and FVIII. Using aPCCs is an option for either type of

    hemophilia with inhibitors present. The disadvantages of this treatment strategy is that the

    products have lower purity, high risk of thrombosis, no suitable monitoring strategy, and

    possibility of an anamnestic response, which means that the body may develop an immune

    response upon repeated exposure. For these reasons, recombinant FVIIa (NovoSeven) is a

    preferred strategy to manage bleeds in hemophilia patients with high titers of inhibitors.

    Recombinant FVIIa bypasses the factor deficiency and activates factor X, which can initiate

    thrombin formation, and it is only active at site of tissue injury. Recombinant FVIIa can be used

    for prevention and treatment of bleeding. The major limitation of this product is that it has a

    very short half-life and requires redosing every 2 hours. So continuous infusion is a more

    convenient and cost-effective method to administer recombinant FVIIa, and the interval can be

    extended once hemostasis is achieved. There is a less risk of viral transmission and anamnestic

    response compared to aPCCs. There are no routine laboratory tests that can accurately

    measure the efficacy of recombinant FVIIa infusion.44-46 Recombinant FVIIa has been

    demonstrated to stop bleeding episodes effectively with one dose as opposed to treatment

  • 18

    with aPCCs.47 Due to a reduced need for re-infusion of product, recombinanat FVIIa is overall

    more cost-effective than aPCCs.47

    Finally, the last option available to treat an acute bleed for hemophilia patients with

    high titers of inhibitors is the use of porcine factor VIII. This option is only available for patients

    with hemophilia A with inhibitor development. Since this is obtained from a foreign source,

    there is a high risk of a severe allergic reaction when given. Because of the risk of

    hypersensitivity and thrombocytopenia, porcine factor VIII is no longer commercially available,

    but it can be obtained for specific patients that have no response to recombinant FVIIa or PCC

    or have severe hemorrhages.44

    If the hemophilia patient with inhibitors is not actively bleeding and has a low titer < 5

    BU, then immune tolerance therapy is recommended so that maintenance factor replacement

    is a viable option. Providing high doses of factor concentrate therapy as a regular infusion

    ranging from 25 units/kg every other day to 200 units/kg daily has been shown to eradicate

    inhibitors.44,46 High-dose factor treatment has been successful in 70% of patients.48 Drugs that

    suppress the immune system have also been investigated for inhibitor eradication in

    hemophilia patients. Rituximab is an anti-CD20 antibody that destroys existing B-cells, which

    are present in immune response.49 Other immunosuppressive agents used to control inhibitors

    include corticosteroids, cyclophosphamide, immunoglobulin, and prednisone.46 These drugs

    can be used alone or in combination to reduce immune activity. However, these treatments

    are not usually pursued because of notable adverse effects. For example, corticosteroids can

    cause mood instability, weight gain, hypertension, and hyperglycemia. Because these agents

    suppress the immune system, patients are higher risk for deadly infections. The process by

  • 19

    which immune tolerance is induced creates high costs, estimating up to 4 times higher than

    patients without inhibitors ($697,000 vs. $155,000).30 In addition, all of these treatments

    would require adequate monitoring to ensure both efficacy and safety.

    Gene Therapy

    In the recent years, there has been a focus on developing gene therapy for management

    of hemophilia because the disease is usually caused by a single gene defect.50 The treatment

    looks promising when tested in dogs and mice with knock-out mutations for the coagulation

    factor gene. However, when used in human models, the same degree of coagulation factor

    production was not achieved compared to animal models.17 Though gene therapy is promising,

    it is currently not a viable option for mass use among hemophilia patients. Further

    investigation is warranted to demonstrate universal success of gene therapy.

    Hemophilia Products in Development

    For the last three decades, the hemophilia market has been dominated by recombinant

    clotting factors produced by specific manufacturers including Baxter, Bayer, and CSL Behring.

    There have been several generations of recombinant coagulation factors. The most recent

    generation (third-generation recombinant factors) lack bovine or human proteins in the

    synthesis of coagulation factors or in the final products), which lowers the risk of viral

    transmission. Aside from this advance in manufacturing, there have been no other major

    changes that have affected management of hemophilia patients. Table 6 summarizes the

    products that are currently being developed or recently approved. There are two new

    recombinant products approved in 2013 (Rixubis and Alprolix) and one in 2014

  • 20

    (NovoEight).51,52 Also, there are many longer-acting versions of clotting factors in

    development, which may be helpful since there has been an increased use of prophylaxis.

    Comprehensive Care

    Hemophilia is a rare disorder that is complex to manage and requires optimal care of

    patients, especially in those with severe forms of the disease. Comprehensive care goes

    beyond treatment of acute bleeds and is crucial to promote physical and psychosocial health

    and quality of life and can potentially decrease morbidity and mortality. A multidisciplinary

    care team is needed to address prevention and treatment, as well as vein and dental care.

    Because an acute life-threatening bleed can occur anytime at any location patients should carry

    easily accessible identification indicating diagnosis, type and severity of hemophilia, inhibitor

    status, type of factor needed for repletion, initial dosage for treatment of mild, moderate, and

    severe bleeding as well as contact information.53,54 Adequate emergency care should be

    available at all times including access to a coagulation laboratory capable of performing clotting

    factor assays, provision of appropriate clotting factor concentrates, blood products if factor

    concentrates not available, and casting and/or splinting for immobilization and

    mobility/support aids as needed.

    A comprehensive care program is needed to coordinate inpatient and outpatient care

    and services to patients and their family. Patients should be seen by all multisdisciplinary team

    members on a yearly basis for a complete hematologic, musculoskeletal, and psychosocial

    assessment.8 The management plan should be developed in collaboration with the patient and

    communicated to all practitioners involved in the patients care. Communication among

  • 21

    healthcare providers is key. In addition, the patient, family members, and other caregivers

    should be educated on potential consequences to ensure that optimal care is provided.

    Pharmacist Role

    Pharmacists play an important role in making sure that patients are receiving

    appropriate care. The most important responsibility is the evaluation of factor concentrate

    dosing regimens for treatment and prophylaxis. Pharmacists can also perform medication

    regimen reviews to ensure that the patient is not receiving non-steroidal anti-inflammatory

    drugs, aspirin, or drugs affecting platelet adhesion since these can put the patient at a higher

    risk for uncontrolled bleeding. Pharmacists can also assist with insurance authorizations to

    secure reimbursement for clotting factors and communicate any issues with the hemophilia

    comprehensive care team. There are also specialty pharmacies that manage and coordinate

    care of hemophilia patients, and this is where majority of clotting factors are dispensed.

    Treatment with clotting factor requires intravenous access, and patients and their caregivers

    require training on how to infuse clotting factors at home.

    Pharmacists can provide hemophilia disease education to patients, families, and other

    involved team members. They can assist families with making treatment decisions to provide

    effective therapy with minimal side effects. They can minimize barriers to access clotting

    factors, manage refills appropriately, and provide infusion training techniques and medication

    education on clotting factor storage, preparation, and reconstitution. Pharmacists should take

    the initiative to contact patients routinely to assess compliance, especially if patients are on

    prophylaxis or immune tolerance therapy, to ensure proper adherence. Finally, pharmacists

  • 22

    can assess each patients current regimen and determine the best treatment option using

    evidence-based medicine.

    Conclusion

    Hemophilia is a chronic illness that requires complex and comprehensive medical care

    to optimize patient outcomes including extending life expectancy and improving quality of life.

    Indeed, if patients receive appropriate comprehensive care at a hemophilia treatment center

    and follow preventive care, these patients with hemophilia A or B can have a long life

    expectancy without disability. There have been major advancements in the treatment and

    prevention of bleeds that have improved quality of life for these patients. In the future, gene

    therapy may serve as a cure for hemophilia, but further investigation is needed to clarify the

    place in therapy. Current research is focused on improving treatment administration to

    increase compliance for patients and allow them to function appropriately in society. Choosing

    the appropriate therapy is dependent on several factors such as age, bleeding patterns, joint

    health, and levels of physical activity) and should be determined on an individual case basis.

    Multidisciplinary healthcare team coordination is necessary to ensure that patients are aware

    of risks of hemophilia treatment so that they opt for safer methods of treatments. Both the

    healthcare team and patients must understand the efficacy and safety of each treatment to

    make an appropriate decision for management of hemophilia.

  • 23

    References:

    1. Mannucci PM, Tuddenham EG. The hemophilias from royal genes to gene therapy. N Engl

    J Med. 2001;344:1773-1779.

    2. Stonebraker JS, Bolton-Maggs PH, Soucie JM, Walker I, Brooker M. A study of variations in

    the reported haemophilia A prevalence around the world. Haemophilia. 2010;16:20-32.

    3. Van Dijk K, Fishcer K, van der Bom JG, Grobbee DE, van den Berg HM. Variability in clinical

    phenotype of severe haemophilia: The role of the first joint bleed. Haemophilia.

    2005;11:438-443.

    4. Rosendaal G, Lafeber FP, Pathogenesis of hemophilic arthropathy. Haemophilia.

    2006;12:117-121.

    5. Mannucci PM. Back to the future: A recent history of hemophilia treatment. Haemophilia.

    2008;14:10-18.

    6. Plug I, van der Bom JG, Peters M, et al. Thirty years of hemophilia treatment in the

    Netherlands, 1972-2001. Blood. 2004;104:3494-3500.

    7. Wight J, Paisley S. The epidemiology of inhibitors in haemophilia A: A systematic review.

    Haemophilia. 2003;9:418-435.

    8. Srivastava A, Brewer AK, Mauser-Bunschoten EP, et al. Guidelines for the management of

    hemophilia. Hemophilia. 2013;19:e1-e47.

    9. High K. The leak stops here: platelets as delivery vehicles for coagulation factors. J Clin

    Invest. 2006;116:1840-1842.

    10. Teitel JM, Barnard D, Israels S, Lillicrap D, Poon MC, Sek J. Home management of

    haemophilia. Haemophilia. 2004;10:118-133.

  • 24

    11. Frachon X, Pommereuil M, Berthier AM, et al. Management options for dental extraction in

    hemophiliacs: a study of 55 extractions (2000-2002). Oral Surg Oral Med Oral Pathol Oral

    Radiol Endod. 2005;99:270-275.

    12. Rattray B, Nugent DJ, Young G. Celecoxib in the treatment of haemophilic synovitis, target

    joints, and pain in adults and children with haemophilia. Haemophilia. 2006;12:S14-17.

    13. Hermans C, de Moerloose P, Fischer K, et al. Haemophilia Therapy Standardisation Board.

    Management of acute haemarthrosis in haemophilia A without inhibitors: literature review,

    European survey and recommendations. Haemophilia. 2011; 17:383-92.

    14. Rattray B, Nugent DJ, Youn G. Celecoxib in the treatment of haemophilic synovitis, target

    joints, and pain in adults and children with haemophilia. Haemophilia. 2006;12:514-517.

    15. Tsoukas C, Eyster ME, Shingo S, et al. Evaluation of the efficacy and safety of etoricoxib in

    the treatment of hemophilic arthropathy. Blood. 2006;107:1785-1790.

    16. Rodriguez-Merchan EC. Musculoskeletal complications of hemophilia. HSSJ. 2010;6:37-42.

    17. Mannucci P. Hemophilia: treatment options in the twenty-first century. Thromb Haemost.

    2003;1:1349-1355.

    18. Ludlam A, Powderly WG, Bozzetta S. Clinical perspectives of emerging pathogens in

    bleeding disorders. Lancet. 2006;367:252-261.

    19. Ingram C. The history of haemophilia. J Clin Pathol. 1976;29:469-479.

    20. Farrugia A. Evolving perspectives in product safety for haemophilia. Haemophilia.

    2002;8:236-243.

    21. Pipe SW. Recombinant clotting factors. Thromb Haemost. 99:840-850.

  • 25

    22. White G, Shapiro A, Ragni M, et al. Clinical evaluation of recombinant factor IX. Semin

    Hematol. 1998;35:33-38.

    23. Lusher JM, Arkin S, Abildgaard CF, Schwartz RS. Recombinant factor VIII for the treatment of

    previously untreated patients with hemophilia A. Safety, efficacy, and development of

    inhibitors. Kogenate Previously Untreated Patient Study Group. N Engl J Med.

    1993;328:453-459.

    24. Bartorova A, Martinowitz U. Continuous infusion of coagulation factors. Haemophilia.

    2002;8:170-177.

    25. Kasper CK. Products for clotting factor replacement in developing countries. Seminr Thromb

    Hemost. 2005;31:507-512.

    26. Giangrande PL. Blood products for hemophilia: past, present and future. Bio Drugs.

    2004;18:225-234.

    27. Fanchini M, Salvagno GL, Lippi G. Inhibitors in mild/moderate haemophilia A: an update.

    Thromb Haemost. 2006;96:113-118.

    28. Goudemand J, Laurian Y, Calvez T. Risk of inhibitors in haemophilia and the type of factor

    replacement. Curr Opin Hematol. 2006; 13:316-322.

    29. Knobe KE, Sjorin E, Tengborn LI, Petrini P, Ljung RC. Inhibitors in the Swedish population

    with severe haemophilia A and B: a 20-year survey. Acta Paediatr. 2002;91:910-914.

    30. Kessler C, Santilli M. Understanding hemophilia: a manafed care review. CDMI Report. Fall

    2013;32-38.

    31. Farrugia A. Evolving perspectives in product safety for haemophilia. Haemophilia.

    2002;8:236-243.

  • 26

    32. Barrow RT, Healey JF, Gailani D, Scandella D, Lollar P. Reduction of the antigenicity of factor

    VIII toward complex inhibitory antibody plasmas using multiply-substituted hybrid

    human/porcine factor VIII molecules. Blood. 2000;95:564-568.

    33. Castaman G. Desmopressin for the treatment of haemophilia. Haemophilia. 2008;14:15-20.

    34. Manucci PM. Desmopressin (DDAVP) in the treatment of bleeding disorders: the first 20

    years. Blood. 1997;90:2515-21.

    35. Lessinger C, Becton D, Cornell C Jr, Cox Gill J. High-dose DDAVP intranasal spray (Stimate)

    for the prevention and treatment of bleeding in patients with mild haemophilia A, mild or

    moderate type 1 von Willebrand disease and symptomatic carriers of haemophilia A.

    Haemophilia. 2001;7:258-266.

    36. Coetzee MJ. The use of topical crushed tranexamic acid tablets to control bleeding after

    dental surgery and from skin ulcers in haemophilia. Haemophilia. 2007;13:443-444.

    37. Hvas AM, Sorensen HT, Norengaard L, Christiansen K, Igerslev J, Sorensen B. Tranexamic

    acid combined with recombinant factor VIII increases clot resistance to accelerated

    fibrinolysis in severe hemophilia A. J Thromb Haemost. 2007;5:2408-2414.

    38. Aronstam A, Arblaster PG, Rainsford SG, et al. Prophylaxis in haemophilia: a double-blind

    controlled trial. Br J Haematol. 1976;33:81-90.

    39. Manco-Johnson MJ, Abshire TC, Shapiro AD, et al. Prophylaxis versus episodic treatment to

    prevent joint disease in boys with severe hemophilia. N Engl J Med. 2007;357:535-544.

    40. Bianchette VS. Prophylaxis in the haemophilia population. Haemophilia. 2010;16:181-188.

  • 27

    41. Gringeri A, Lundin B, von Mackensen S, et al. ESPRIT Study Group. A randomized clinical trial

    of prophylaxis in children with hemophilia A (the ESPRIT study). J Thromb Haemost.

    2011;9:700-710.

    42. National Hemophilia Foundation. MASAC Recommendation #179 MASAC recommendation

    concerning prophylaxis (regular administration of clotting factor concentrate to prevent

    bleeding).

    http://www.hemophilia.org/NHFWeb/MainPgs/MainNHF.aspx?menuid=57&contentid=100

    7. Accessed 2014 November 29.

    43. Astermark J, Satagostino E, Hoots KW. Clinical issues in inhibitors. Haemophilia. 2010;16:54-

    60.

    44. Wight J, Paisley S. The epidemiology of inhibitors in haemophilia A: a systematic

    review. Haemophilia. 2003;9:418-435.

    45. Hay CR. Factor VII inhibitors in mild and moderate-severity haemophilia A.

    Haemophilia. 1998;4:558-563.

    46. Berntorp E, Collins P, DOrion R, et al. Identifying non-responsive bleeding episodes

    in patients with haemophilia and inhibitors: a consensus definition. Haemophilia.

    2011;17:e202-210.

    47. Joshi AV, Stephens JM, Munro V, Mathew P, Botteman MF. Pharmacoeconomic

    analysis of recombinant factor VIIa versus APCC in the treatment of minor-to-

    moderate bleeds in hemophilia patients with inhibitors. Curr med Res Opin.

  • 28

    2006;22:23-31.

    48. Manno CS. Management of bleeding disorders in children. Hematology Am Soc

    Hematol Educ Program. 2005;416-22.

    49. Wiestner A, Cho HJ, Asch AS, et al. Rituximab in the treatment of acquired factor

    VIII inhibitors. Blood. 2002;100:3426-3428.

    50. Gan SU, Kon OL, Calne RY. Genetic engineering for haemophilia A. Expert Opin

    Biol Ther. 2006;6:1023-30.

    51. Gouw S, van der Bom J, Ljung et al. Factor VII products and inhibitor development

    in severe hemophilia A. N Engl J Med. 2013;368:231-239.

    52. Clement P New factor concentrates. The future is now. Parent Empowerment

    Newsletter. 2013;23:10-11.

    53. Evatt BL. The natural evolution of haemophilia care: developing and sustaining

    comprehensive care globally. Haemophilia. 2006;12:13-21.

    54. Evatt BL, Black C, Batarova A, Street A, Srivastava A. Comprehensive care for

    haemophilia around the world. Haemophilia. 2004;10:9-13.

  • 29

    Table 1. Hemophilia Severity Classification by Factor Concentration8

    Classification Factor Concentration* (Units/mL)

    Clinical Manifestations

    Mild > 0.05 0.4 Units/mL (>5 40%)

    Hemorrhage with magor trauma or surgery May go years without diagnosis

    Moderate 0.01 0.05 Units/mL (1 5%)

    Occasional spontaneous hemorrhages Hemorrhage with mild trauma/surgery that is prolonged

    Severe < 0.01 Units/mL (

  • 30

    Table 3. Guidelines for Factor Replacement

    Hemorrhage Factor VIII (% of normal)

    Factor IX (% of normal)

    Duration (days)

    Severe

    Intracranial

    Retroperitoneal

    Retropharyngeal

    60 100 50 100 10 14

    Moderate

    Hemarthroses

    Hematoma

    Hematuria

    30 60 25 50 3 7

    Minor

    Epistaxis

    Oral (mild)

    20 40 15 30 1 3

    Surgery* 50 100 50 100 Up to 14

    *For minor surgery, can consider lower goal of 30-60% of normal. For major surgery, can

    consider higher goal of 100% before surgery begins and may repeat after 6-12 hours and

    continue until healing is complete.

    Table 4. Dosing of Recombinant Coagulation Factors

    Factor VIII Factor IX

    Initial Dose (units) Desired increase (%) x Weight (kg) x 0.5

    Pediatric: Desired increase (%) x Weight (kg) x 1.4 Adult: Desired increase (%) x Weight (kg) x 1.2

    Maintenance Dose (units)

    50% of initial dose 50% of initial dose

    Expected Response (0.02 Units/mL)

    (0.01 Units/mL)

    Half-life (hours) 8 15 11 27 Dosing Interval 12 24 (minor bleed)

    8 12 (moderate severe bleed)

    12 24

  • 31

    Table 5. Prophylactic Factor Replacement

    Protocol Definition

    Episodic Treatment Treatment given at time of clinically relevant bleeding

    Continuous Prophylaxis Primary Prophylaxis Regular continuous treatment in the

    absence of documented osteochondrial joint disease and started before second clinical evident large joint bleed and age 3 years

    Secondary Prophylaxis Regular continuous treatment after 2 or more bleeds into large joints and before onset of joint disease

    Tertiary Prophylaxis Regular continuous treatment started after onset of joint disease

    Intermitent Prophylaxis Treatment given to prevent bleeding for periods not exceeding 45 weeks in a year

    Table 6. Emerging Hemophilia Products in Development51,52

    Product Name (Manufacturer) Product Type/Indication Distinguishing Feature

    Regulatory Status

    Factor VIII

    BAX 855 (Baxter) Pegylated rFVIII Long acting Phase II/III

    BAY 81-8973 (Bayer) BDD rFVIII 3rd generation; normal t1/2

    Phase III completed March 2013

    BAY 94-9027 (Bayer) Pegylated rFVIII Long acting Phase III

    BIIB 031 (Blogen Idec) BDD rVIII-Fc fusion Long acting Phase III completed March 2014

    CSL627 (CSL Behring) Single-chain rFVIII Improved stability during manufacturing

    Phase II/III

    GreenGene F (Green Cross Corporation)

    rFVIII New in the United States

    Phase III

    Turoctocog alfa (Novo Nordisk NovoEight)

    BDD rFVIII Normal t1/2 Approved 2013

    NN7088 (Novo Nordisk N8-GP) Glyco-pegylated rFVIII Long acting Phase III

  • 32

    Human-cl rhFVIII (Octapharma) Hemophilia A Normal t1/2 Phase III

    NecLip-pdFVIII (Recoly NV LongAte)

    Plasma-derived factor VIII formulated with NecLip

    Long acting Approved in Russia

    Factor IX

    BAX 326 (Baxter Rixubis) Third-generation rFIX First 3rd generation product

    Approved June 2013

    BIIB 029 (Biogen Idec/Swedish Orphan Biovitrium Alprolix)

    rFIX-Fc fusion Long acting Approved March 2014

    IB1001 (Cangene Corporation) rFIX Approval on hold pending data requested by FDA

    CSL654 (CSL Behring) rFIX albumin fusion Long acting Phase II/III

    NN7999 (Novo Nordisk N9-GP) Glyco-pegylated rFIX Long acting Phase III

    Inhibitors of Factor VIIA

    BAX 817 (Baxter) Hemophilia with inhibitors rFVIIa

    Normal t1/2 Phase III

    OBI-1 (Baxter) Hemophilia A with inhibitors or acquired hemophilia A

    Recombinant (porcine)

    Phase III

    CSL589 rVIIa-FP (CSL Behring) Hemophilia A or B with inhibitors; rFVIIa-albumin fusion

    Long acting Phase I

    LA-rFVIIa (Novo Nordisk) rFVIIa Long acting Phase I/II

    PF-05280602 (Pfizer/Catalyst Biosciences)

    Hemophilia with inhibitors; rFVIIa

    Produced by human cell line

    Phase I

    NecLip-rFVIIa (Recoly NV LongSeven)

    Hemophilia with inhibitors; rFVIIa formulated with NecLip

    Long acting Phase I/II

    LR769 (rEVO biologics/LFB Biotechnologies)

    Hemophilia A or B with inhibitors; transgenic rFVIIa

    Produced in rabbit milk

    Phase II

    rFIII, recombinant factor VIII; BDD, B-domain deletion; t1/2, half-life; rFIX, recombinant factor

    FIX; rFVIIa, recombinant factor VIIa; NecLip, non-encapsulating liposomes; FDA, Food and Drug

    Administration

  • 33

    ACTIVITY TEST

    1. Which of the following statements is TRUE regarding epidemiology of hemophilia?

    A. Hemophilia B is the most common form counting for 80-85% of cases

    B. Majority of hemophilia cases occur in females

    C. Severe hemophilia is characterized by a coagulation factor level > 5 IU/dL

    D. Severe hemophilia is associated with spontaneous joint and muscle bleeds.

    2. Hemophilia B is caused by a deficiency of which clotting factor?

    A. Factor VII

    B. Factor VIII

    C. Factor IX

    D. Factor XII

    3. Which of the following abnormal laboratory results is (are) consistent with a diagnosis of

    hemophilia type B?

    I. Prolonged activated partial thromboplastin time (aPTT)

    II. Prolonged prothrombin time (PT)

    III. Decreased factor VIII level

    IV. Decreased factor IX level

    A. I and III

    B. I and IV

    C. II and IV

    D. I, II and III

    E. I, II, IV

    4. JK is a 10 year old boy who was recently diagnosed with hemophilia A, and his hematologist

    has ordered coagulation tests to determine severity. The test results have revealed that his

    factor VIII level is 0.1 Units/mL (10%). How would you classify JKs severity based on this

    information?

    A. Mild hemophilia A

    B. Moderate hemophilia A

    C. Severe hemophilia A

    D. Cannot determine severity due to insufficient information provided

    5. Which of the following severity of hemophilia is correctly matched with the associated

    clinical manifestations?

    A. Mild hemophilia occasional spontaneous hemorrhages

    B. Moderate hemophilia hemorrhage with mild trauma/surgery is usually prolonged

    C. Moderate hemophilia life-threatening hemorrhages

    D. Severe hemophilia may go years without diagnosis

  • 34

    6. All of the following statements regarding preventive and supportive measures is true

    EXCEPT:

    A. Acute bleeds should be treated within 2 hours to prevent joint damage in hemophilia patients

    B. Patients should be educated on home treatment since it can improve quality of life and decrease

    pain and disability.

    C. Immunizations against Hepatitis C and Human Immunodeficiency Virus (HIV) are necessary prior to

    receiving replacement clotting factors.

    D. Maintaining factor levels of at least 0.5-0.7 units/mL (50-70%) should be adequate when

    performing minor surgical interventions in patients with hemophilia.

    7. Assuming that there are adequate resources and cost is not an issue, which of the following

    treatment options is the most appropriate treatment option for managing an acute bleed for a

    patient with hemophilia B?

    A. Cryoprecipitate

    B. Recombinant factor IX

    C. Activated prothrombin complex concentrate

    D. Aminocaproic acid

    8. Which of the following options is the most appropriate option to manage an acute bleed for

    a patient with a history of hemophilia A, who has the presence of inhibitors (measured as 3

    Bethesda Units)?

    A. Cryoprecipitate.

    B. Tranexamic acid

    C. Recombinant Factor VIIa

    D. High doses of recombinant factor VIII

    9. Which of the following statements is FALSE regarding prophylactic factor replacement for

    hemophilia patients?

    A. The goal is to maintain the deficient factor level at a minimum of 0.01 units/mL (1%).

    B. It is more beneficial to initiate prophylaxis in hemophilia patients when they reach adulthood.

    C. Providing prophylactic replacement has been shown to prevent bleeding and joint destruction.

    D. Providing tertiary prophylaxis (initiation after onset of joint disease) can prevent significant

    progression of joint disease.

    10. Other than factor replacement, which of the following treatments is the most appropriate

    option for management of epistaxis in a patient with mild hemophilia A?

    A. Cryoprecipitate

    B. Fresh frozen plasma

    C. Desmopressin

    D. Aminocaproic acid

  • 35

    11. Which of the following initial dosing regimens for recombinant factor VIII concentrate

    would you recommend for a patient with Hemophilia A who is experiencing an intracranial

    hemorrhage (assume the goal is to increase factor level to 100% of normal)? The patients

    weight is 165 pounds, and the factor VIII level is 0.007 Units/mL (0.7%).

    A. 37.5 units intravenously once

    B. 90 units intravenously once

    C. 3750 units intravenously once

    D. 9000 units intravenously once

    12. Which of the following parameters would you recommend to monitor when administering

    recombinant clotting factor to manage a hematoma in a patient with hemophilia B?

    A. Factor VIII level 15 minutes after completed infusion

    B. Hypertension

    C. Thrombosis

    D. Thrombocytopenia

    13. Which of the following medications is correctly matched with an associated adverse effect?

    A. Desmopressin hyponatremia

    B. Aminocaproic acid visual disturbances

    C. Recombinant Factor VIIa thrombocytopenia

    D. Recombinant Factor VIII - rhabdomyolysis

    14. Which of the following pharmacologic options is NOT appropriate for a hemophilia patient

    who is experiencing chronic pain in his right knee due to a history of hemarthrosis (joint

    bleeding)?

    A. Acetaminophen

    B. Ibuprofen

    C. Oxycodone

    D. Dexamethasone

    15. Which of the following statements is TRUE when counseling a hemophilia patient on

    medication management?

    A. Plasma-derived coagulation factors are first line agents for factor replacement.

    B. Prophylactic replacement is not recommended since it does not prevent recurrent joint bleeds.

    C. You do not need to receive factor replacement if you are undergoing a dental procedure.

    D. Home treatment can decrease pain and reduce risk of musculoskeletal dysfunction and long-term

    disability.

  • 36

    16. Which of the following is considered a major risk factor for the development of inhibitors in

    a patient with hemophilia?

    A. Mild hemophilia

    B. Hemophilia B

    C. High-intensity factor replacement therapy

    D. Bolus dosing of factor replacement therapy

    17. What would be the most appropriate recommendation for managing a patient hemophilia

    B found to have presence of inhibitors with a low titer of 4 BU that is not actively bleeding?

    A. Cyclophosphamide

    B. Immunoglobulin

    C. Prednisone

    D. Immune tolerance therapy with recombinant factor IX

    18. If a patient with hemophilia A is found to have presence of inhibitors with a high titer of 12

    BU, what would be the most appropriate recommendation for managing an acute

    retroperitoneal bleed?

    A. Activated prothrombin complex concentrates

    B. Porcine factor VIII

    C. Recombinant factor VIIa

    D. High-dose replacement of recombinant factor VIII

    19. Which of the following is TRUE regarding emerging therapies for hemophilia that are

    currently in development?

    A. Third generation recombinant clotting factors are associated with low risk since they contain

    human proteins.

    B. Gene therapy has been demonstrated to be effective in humans and will be available for use in the

    near future.

    C. Many of the new products in development are longer-acting versions of clotting factors to increase

    ease of use.

    D. The new products in development have been associated with a higher risk of viral transmission.

    20. Pharmacists can play an important role in the coordination and management of care for

    patients with hemophilia. Which of the following are responsibilities that pharmacists have

    when taking care of hemophilia patients?

    A. Evaluate factor concentrate dosing regimens for treatment and prophylaxis.

    B. Train patients and caregivers on proper infusion technique of clotting factors at home.

    C. Contact patients routinely to assess patient adherence to prescribed factor replacement regimen.

    D. All of the above

    Please submit your final responses on freeCE.com. Thank you.