Expired activity
Please go to the PowerPak homepage and select a course.

Immune Globulin Therapies—How the Appropriate Selection and Use of Immune Globulin Products Can Impact Patient Care

Introduction/Background

Intravenous immunoglobulins (IVIGs) are blood products approved to treat antibody deficiency and certain autoimmune diseases. They are also often used off-label to treat many neurological, hematological, rheumatological, and dermatological diseases.1 When discussing formulation and indications, this activity will primarily focus on intravenous formulations of immunoglobulins and their U.S. Food and Drug Administration (FDA) approved indications. IVIGs are produced by pooling plasma from a large number of healthy donors, typically ranging from 1000 to 40,000 donors per batch.1,2 The manufacturing process for IVIGs also includes purification, stabilization, and viral reduction methods. IVIGs have been associated with a number of adverse effects that range from mild to severe. These adverse effects can be potentiated by the intervariable characteristics among different IVIG products and patient characteristics or comorbidities that predispose them to developing certain side effects. Thus, it is important for pharmacists to enhance their knowledge about these products in order to select the appropriate IVIG products for the right patients.

Manufacturing Process for IVIG Products

IVIG products are produced through a number of sequential steps. The initial step in manufacturing IVIG products requires the collection of donated human plasma through either whole blood donation or a process called plasmapheresis. Plasma derived from whole blood donation is called recovered plasma. The whole blood is centrifuged, usually within 12 hours of collection, to separate the cellular components from the plasma.2 Within the United States, approximately 250 milliliters of plasma are obtained from a single pint of donated whole blood.3 Alternatively, plasma derived from plasmapheresis is referred to as source plasma. Plasmapheresis is an automated process using specialized equipment that removes whole blood from a donor, immediately separates it into plasma and cellular components, and then returns the cellular components to the donor in sterile saline solution to help the body replace the removed plasma.2,4 About 600 to 800 milliliters of source plasma can be obtained from one donation.3 In addition, source plasma can be donated up to 2 times within a 7 day period, whereas recovered plasma from whole blood can only be donated once every 8 weeks.5,6 The major manufacturers of IVIG products, which include Baxter, CSL Behring, and Grifols, primarily use source plasma donated at certified plasma collection centers.

Individuals who choose to donate source plasma must be determined as suitable donors based on qualifications provided in the Code of Federal Regulations (CFR).6 These qualifications serve to protect the health and safety of the donor, as well as the safety of therapeutic products produced from the donated plasma. Prospective donors must first provide their informed consent before undergoing the plasma collection procedure. Next, a qualified, licensed physician or an individual under their supervision completes an initial medical examination of each donor on the day of first donation or no more than 1 week prior to the first donation to certify that the donor is in good health. Subsequent medical examinations should be conducted, as well, at intervals of no longer than 1 year.

A donor is considered to be in good health on the day of their donation if the following qualifications are met: normal temperature, systolic and diastolic blood pressures within normal limits, blood hemoglobin level of no less than 12.5 g/dL or a hematocrit level of no less than 38%, normal pulse rate, total serum or total plasma protein of no less than 6 g/dL, and a body weight of at least 110 pounds. Additionally, the donor must not have any of the following: acute respiratory diseases, skin infections at the site of phlebotomy, any disease transmissible by blood transfusion, skin punctures or scars on arms and forearms indicative of addiction to self-injected narcotics, history of viral hepatitis after the eleventh birthday, history of close contact within 12 months of donation with an individual having viral hepatitis, and history of having received, within 12 months, human blood or any derivative that is a possible source of viral hepatitis. Along with meeting the qualifications of good health described above, each donor needs to provide a blood sample on the day of their initial medical examination or first plasma donation, whichever comes first, and at least every 4 months thereafter. The blood sample is used to test for syphilis and to quantify the immunoglobulin composition of the donor’s plasma.6

According to the Plasma Protein Therapeutics Association (PPTA), an organization that represents the collectors of source plasma used to derive treatments, the following 2 types of source plasma donors exist: applicant donors and qualified donors. An applicant donor is an individual who initially presents to donate source plasma and fulfills the CFR and International Quality Plasma Program (IQPP) screening requirements. Developed by PPTA, the voluntary IQPP standards serve to enhance the quality and safety of collected source plasma. The IQPP screening requirements for applicant donors include completing a physical examination, checking the donor’s name against the National Donor Deferral Registry (NDDR), and educating the donor abotut high-risk activities. An applicant donor can be reclassified as a qualified donor if he or she passes the requirements mentioned above at a follow-up screening and provides a subsequent donation of either a complete unit of source plasma or a blood/plasma sample only. The qualified donor must test nonreactive for human immunodeficiency virus (HIV), hepatitis B virus (HBV), and hepatitis C virus (HCV). In addition, the follow-up screening to become a qualified donor must occur no later than 6 months after the previous screening. Most importantly, the IQPP standards state that no units of source plasma from applicant donors will be acceptable for manufacturing therapeutic plasma products until the individual achieves the criteria of a qualified donor.7 When a qualified donor no longer fulfills the screening qualifications, the individual is permanently deferred from donating plasma.8

Along with IQPP, the PPTA has another set of voluntary standards that go beyond regulatory requirements to improve the safety of source plasma. Known as the Quality Standards of Excellence, Assurance, and Leadership (QSEAL), this set of standards pertains to the manufacturing of plasma protein therapies. Inventory hold, one of the QSEAL standards, requires all source plasma to be held in inventory for at least 60 days from the time of collection. Used as an added source of protection prior to preparing pools of plasma, inventory hold allows manufacturers to retrieve units of plasma from disqualified donors as determined by postdonation information (i.e., delayed admission of high-risk behavior or international travel, becoming reactive for HIV, HBV, or HCV). Under the inventory hold standard, manufacturers are required to document and verify that each unit of source plasma was not released for further manufacture until after the 60-day hold period expired.9

Once the source plasma is collected, it is analyzed for known viruses using nucleic acid testing (NAT) and released from inventory where it will be pooled with thousands of other plasma donations. After pooling the plasma, the next step is to isolate the immunoglobulin G (IgG). Isolating IgG is accomplished through fractionation techniques. Following IgG fractionation, the product undergoes purification, stabilization, and various types of viral reduction treatments.10

Cold ethanol fractionation is the process that is used to separate IgG from other proteins and contaminants in the pooled plasma. The following 2 types of cold ethanol fractionation exist: the Cohn-Oncley process and the Kistler-Nitschmann method, which is a variation of the former. For each of these fractionation methods, plasma proteins are selectively precipitated into several fractions based on their solubility in solutions of different alcohol concentrations, pH values, ionic strengths, and temperatures.11 An important difference between these 2 methods is that the Cohn-Oncley process removes immunoglobulin A (IgA) while the Kistler-Nitschmann method does not.12 The IgA content of IVIG products may affect patient tolerability and is discussed in the Product Characteristics section. In terms of viral reduction, cold ethanol fractionation inactivates and removes HIV, but does not eliminate the risk for infection with HCV.13

Following cold ethanol fractionation, the isolated IgG is purified through a process called ion exchange chromatography. This process involves separating IgG from contaminants based on the total charge. The impure IgG sample is loaded into an ion exchange chromatography column at a specific pH that causes contaminants to bind to the column while IgG remain in the solution.14 To prevent the aggregation of IgGs during manufacturing and storage, various types of stabilizers are added to IVIG preparations. Examples of stabilizers include sucrose, mannitol, and glycine and are described in more detail in a later section. Preservatives are not added to IVIG preparations because of concerns about patient safety and immunoglobulin integrity.13

Several viral inactivation and elimination treatments are applied to IVIG products during the manufacturing process. Infectious agents that are potentially transmissible by infusing IVIG preparations include the following: hepatitis A, B, C, D, and G, HIV, parvovirus, West Nile virus, severe acute respiratory syndrome (SARS), Monkeypox, Creutzfeldt-Jakob disease, and Mad deer disease.12 The most commonly used treatments to reduce the transmission of these infectious agents are solvent/detergent (S/D), pasteurization, acidic pH, nanofiltration, and caprylate precipitation. The viral reduction steps used to manufacture each IVIG product are listed in the prescribing information for each respective product.15-24

S/D treatments consist of incubating pooled plasma with a nonvolatile organic solvent and 1 of 2 detergents, Tween-80 or triton X-100.25,26 S/D treatments inactivate viruses by disrupting the structural integrity of lipid-enveloped viruses without denaturing plasma proteins. However, S/D treatment is not effective against nonenveloped viruses, such as hepatitis A virus and parvovirus, and requires additional manufacturing steps to eliminate the detergent.27 Pasteurization is a different viral inactivation process that involves a heat treatment of the product at 60°C for 10 hours. Through the denaturation of essential viral components, pasteurization inactivates both enveloped, and to a lesser extent, some nonenveloped viruses.28,29 Another method of eliminating viruses is nanofiltration, which uses filters with pore sizes of 15 to 45 nanometers to retain and remove both enveloped and nonenveloped viruses from IgG preparations based on their size. In addition, nanofiltration may potentially remove prions from IgG products.2,27 Caprylate is a plant-derived fatty acid that quickly inactivates many viral species and does not significantly impact IgG concentration or activity. Because caprylate treatment is gentler than S/D methods, caprylate is considered an alternative viral reduction process to S/D.12 Finally, low pH treatments inactivate enveloped viruses, but have limited efficacy against nonenveloped viruses. Incubating IgG preparations at low pH can also maintain IgG monomer content and limits the reaggregation of IgGs.12,27

Mechanisms of Action

The mechanisms of action through which IVIGs work are complex and depend on the dose of IVIG and the pathogenesis of the patient’s disease.1 In general, IVIG products have inter-related anti-infective and immunomodulatory mechanisms. The anti-infective mechanisms hypothesized consist of precipitation, agglutination, and neutralization of antigens, activation of phagocytosis, complement-mediated cytolysis, and natural killer cell-mediated cytolysis. Hypothesized immunomodulatory mechanisms of IVIG products include neutralization of autoantibodies, downregulation of B-cell and T-cell function, cytokine regulation, and Fc receptor blockade. There are also anti-infective and immunomodulatory mechanisms that overlap. These mechanisms include neutralization of superantigens and elimination of complement-activating circulating immune complexes.30 The mechanism of IVIG efficacy in a particular disease state can be matched to the underlying disease etiologies. IVIG deficiencies and immunosuppressed states utilize all types of mechanisms to benefit patients, whereas diseases with autoimmune etiologies typically benefit from the immunomodulatory effects of IVIG therapy.

FDA Approved Indications

IVIGs are approved for a number of immunodeficiency and autoimmune disorders.31 When treating immunodeficiency disorders, such as primary humoral immunodeficiency (PID) and B-cell chronic lymphocytic leukemia (CLL), a secondary immunodeficiency, a replacement dose ranging from 200 to 800 mg/kg is given every 3 to 4 weeks.1,32 When treating many autoimmune and inflammatory disorders, high dose IVIG therapy, typically 2 g/kg/cycle, is used. These autoimmune diseases include the following: idiopathic thrombocytopenic purpura (ITP), Kawasaki disease, multifocal motor neuropathy (MMN), and chronic inflammatory demyelinating polyneuropathy (CIPD).

All of the IVIG products currently on the market in the United States are approved for the treatment of antibody deficiency. However, not all products have the various autoimmune disease indications listed above on their label and dosing varies as well. Refer to the package insert for a specific product to see which indications it is approved for, as well as for dosing directions. Table 1 presents the immune globulin products that are approved in the United States and their FDA indications.

Table 1. FDA Approved Immunoglobulin Products and Indications15-24,40-42
Product Name Manufacturer FDA Approved Indications
Bivigam Biotest Pharmaceuticals Corporation
  • Primary Humoral Immunodeficiency
Carimune NF CSL Behring
  • Primary Humoral Immunodeficiency
  • Immune Thrombocytopenic Purpura
Flebogamma DIF 5% & 10% Instituto Grifols, SA
  • Primary Humoral Immunodeficiency
Gammagard Liquid Baxter Healthcare Corporation
  • Primary Humoral Immunodeficiency
  • Multifocal Motor Neuropathy
Gammagard S/D Baxter Healthcare Corporation
  • Primary Humoral Immunodeficiency
  • B-cell Chronic Lymphocytic Leukemia
  • Immune Thrombocytopenic Purpura
  • Kawasaki syndrome
Gammaked Grifols Therapeutics Inc
  • Primary Humoral Immunodeficiency
  • Immune Thrombocytopenic Purpura
  • Chronic Inflammatory Demyelinating Polyneuropathy
Gammaplex Bio Products Laboratory Limited
  • Primary Humoral Immunodeficiency
  • Immune Thrombocytopenic Purpura
Gamunex-C Grifols Therapeutics, Inc.
  • Primary Humoral Immunodeficiency
  • Immune Thrombocytopenic Purpura
  • Chronic Inflammatory Demyelinating Polyneuropathy
Octagam Octapharma
  • Primary Humoral Immunodeficiency
Privigen CSL Behring
  • Primary Humoral Immunodeficiency
  • Immune Thrombocytopenic Purpura
Hizentra (SC) CSL Behring
  • Primary Humoral Immunodeficiency
HyQvia (SC) Baxter Healthcare Corporation
  • Primary Humoral Immunodeficiency
GamaSTAN S/D (IM) Grifols Therapeutics, Inc.
  • Hepatitis A prophylaxis
  • Prevention or modification of measles (rubeola)
  • Passive immunization against varicella in immunosuppressed patients
  • Prophylaxis of rubella in early pregnancy
FDA = U.S. Food and Drug Administration; NF = nanofiltered; S/D = solvent/detergent; SC = subcutaneous; IM = intramuscular

Although the efficacy of different IVIG products is often believed to be consistent, they are not pharmaceutically equivalent.31,33 Depending on the processes and techniques used to manufacture each product, the various immunoglobulin levels will vary. Thus, if a patient requires long-term treatment with IVIG therapy, it would be a good practice to maintain him or her on the same product.1

IVIGs are also used off-label to treat many diseases that affect the neurological, hematological, dermatological, and rheumatological systems. Some examples of these off-label treatments include the following: Guillain Barré syndrome, myasthenia gravis, immune hemolytic anemia, dermatomyositis, and systemic lupus erythematosus.

Differences in Product Characteristics

There are currently 10 manufactured IVIG products in the United States34 with varying properties that are dependent upon the fractionation, purification (i.e., ion exchange chromatography), and viral inactivation (e.g., nanofiltration) processes by which they are produced.31 These properties include the following: addition of stabilizers (e.g., sugars, amino acids), pH value, sodium content, osmolarity, osmolality, volume load, and IgA concentration.31

Sugars and amino acids are added to IVIGs to stabilize the product by preventing IgG aggregate formation.12,13 The sugars that are used to stabilize IVIGs include the following: sucrose, glucose, maltose, or D-sorbitol. As an alternative, the amino-acids that can be used for IVIG stabilization are glycine, L-proline, or L-isoleucine. The presence of these stabilizers can increase the likelihood of adverse reactions for certain patient populations (e.g., those with renal impairment and those with diabetes). Thus, the stabilizer used to manufacture the IVIG product should be considered when selecting an appropriate product to treat a patient.

The stability of IVIG products are also dependent on their pH, which varies among products based on the methods used to prepare them. The typical range in pH for these products is between 4.0 and 7.2. However, to avoid infusion-related reactions, most products will ultimately be at a near neutral or physiological pH after preparation.12 Whether sugar or amino acid stabilizers will be needed is also determined by the pH of the IVIG product. A low pH (4.0 to 4.5) will be able to stabilize the product by preventing IgG reaggregation, whereas a near physiological pH will require the addition of stabilizers.12

Sodium content also varies, ranging from trace amounts up to 1.8%.12,13 The concentration of sodium will determine the osmolality of the prepared solution that will be infused.12 Products with a high sodium content will need to be avoided for certain patients to avoid serious adverse reactions. This is described in more detail in a later section.

Infusion volumes will be determined by the concentration of IVIG being administered. Higher concentration doses will require smaller volumes for preparation and infusion, while lower concentration doses will have higher volume load. Certain patient populations (e.g., older adults and neonates) and those with comorbidities may not be able to tolerate preparations that are high in volume.31

In addition to IgG, all IVIG products will have some level of IgA antibodies that may vary significantly among different products. This can range from trace amounts up to 720 μg/mL. The main concern with the IgA content is anaphylactic reactions that may arise in patients who have IgA deficiency and may have developed antibodies against IgA. Proper selection of IVIG products with only trace amounts of IgA will be important when treating these patients.31,35

Adverse Events Associated With Immune Globulins

IVIGs have been associated with many adverse effects that range in terms of severity and seriousness. These adverse events can be local or systemic. Although rare, local adverse reactions at the site of infusion may be observed when administering IVIGs. These local reactions may include pain, bleeding, or bruising at the infusion site.36

Mild adverse events commonly observed include headaches, nausea, malaise, myalgia, arthralgia, chills, anxiety, flushing, abdominal cramps, rash, low-grade fever, and leukopenia.35 Some of these mild side effects can be controlled or treated by premedicating patients prior to infusion. An example of this includes patients who experience urticaria from IVIG products. Giving diphenhydramine or small doses of intravenous corticosteroids to these patients prior to infusion can minimize this adverse event. Additionally, other common side effects, such as fever and headaches or postinfusion nausea, can be treated after they arise with acetaminophen or NSAIDs and prochlorperazine or ondansetron, respectively.35

Serious adverse effects are rare (ranging from < 1% to 6%)32,35-37 and can include the following: acute renal failure, stroke, myocardial infarction, venous thromboembolism, anaphylaxis, aseptic meningitis, and hemolysis.31,35 Patients’ risk factors and comorbid conditions can predispose them to developing these adverse effects.31 Thus, selecting the appropriate IVIG product, based on patients’ characteristics, will be critical in preventing adverse events, especially those that are severe and potentially fatal. Table 2 provides examples of adverse events that are associated with IVIG products.

Table 2. Examples of Adverse Events Associated with IVIG Therapya,10,31,35,38
Mild adverse effects
  • Headache
  • Nausea
  • Malaise
  • Myalgia
  • Arthralgias
  • Chills
  • Fever
  • Shortness of breath
  • Cough
  • Abdominal pain
  • Direct Coombs seroconversion
  • Hypertension
  • Flushing
  • Tachycardia
  • Dyspnea
  • Anxiety
  • Leukopenia
  • Urticaria
Severe adverse effects
  • Anaphylactic reaction
  • Acute renal failure
  • Thromboembolism
  • Aseptic meningitis
  • Myocardial infarction
  • Hemolysis
aFor a complete list of adverse events, refer to the package insert for each product

Patient Risk Factors for Adverse Reactions

Identifying patient characteristics that may put an individual at higher risk for specific adverse reactions to various IVIG products will be important for proper product selection. Key characteristics/comorbidities may include patients at risk for renal failure (e.g., preexisting renal impairment, ingesting nephrotoxic agents concurrently, diabetes, older than 65 years of age), those with cardiovascular disease or those who are at risk for thromboembolic events, congestive heart failure, and IgA deficiency with anti-IgA antibody production.31

Acute renal failure (ARF) is one of the serious adverse effects associated with IVIG treatments. Data collected about patients who have developed ARF indicate that IVIG products stabilized with sucrose increase the possibility of this serious adverse effect. When administered orally, sucrose is enzymatically broken down into glucose and fructose. However, this cleaving of sucrose does not occur when the route of administration is intravenous because there is a lack of the necessary enzyme in the blood.35 This results in the presence of sucrose in the kidneys, which increases the osmotic gradient and results in swelling of proximal tubular cells and decreased renal function.35 Doses of IVIG on the high end (2 g/kg/cycle) of the approved doses are more strongly associated with ARF as compared with lower dose regimens. Thus, the FDA has recommended a maximum infusion rate of 3 mg/kg/min for products containing sucrose.35 In addition to IVIG product properties, patient characteristics also play an important role in increasing the risk of ARF development. These characteristics may include baseline renal insufficiency, hypertension, diabetes mellitus, dehydration, older than 65 years of age, ingesting nephrotoxic agents concomitantly, and paraproteinemia.31,35

Patients with known cardiovascular disease or elevated risk of thromboembolic events should avoid high-dose IVIG products containing high sodium levels, high osmolality/osmolarity, and high volume load.31 A mechanism by which IVIG may induce a thromboembolic event is through the increase of serum viscosity and or the presence of prothrombotic Factor XI.35-36 In healthy patients without significant risk factors for stroke and vascular disease, this elevation in viscosity will not have a negative effect. This same mechanism is thought to be the cause of myocardial infarctions that develop in patients receiving IVIG therapy. Most cases of myocardial infarction attributed to IVIG treatment affect patients who have substantial cardiovascular risk factors.35

Anaphylactic reactions, although rare, are also a concern with IVIGs. The primary risk factor to evaluate is whether the patient has IgA deficiency leading to development of anti-IgA antibodies. If an IVIG product that has a high concentration of IgA (e.g., Carimune NF) is administered, this could potentially lead to anaphylaxis. Therefore, it is important to assess whether the patient has developed anti-IgA antibodies as the result of an IgA deficiency before selecting an appropriate product with low or trace amounts of IgA.31,35

Although reported with very low incidence, aseptic meningitis has been associated with IVIG in several cases. The proposed underlying risk for developing this adverse reaction stems from both IVIG therapy and patient risk factors. It has been reported that slowing the rate of infusion may decrease the chance of a patient developing aseptic meningitis, although there are cases where it has still occurred. Higher doses (2 g/kg/cycle) have also been associated with aseptic meningitis. Additionally, it has been reported that a history of migraine headaches may predispose patients to aseptic meningitis when receiving IVIG therapy. Thus, antimigraine prophylaxis might be considered for these patients prior to receiving IVIGs. No deaths have been attributed to IVIG-related aseptic meningitis and symptoms are self-limiting in nature. Treatment should be supportive and systemic therapy may not be necessary.35

Age may also subject patients to certain adverse events and will require careful product selection. Older adult patients (aged 65 years and older) may have less tolerance for products that have high concentrations of sugars and/or salts, as well as hyperosmolar preparations. Increased monitoring should be conducted for this patient population. In addition, older adult patients are more likely to have comorbidities that may make them more sensitive to adverse events.32 Neonatal and pediatric patients have smaller peripheral veins and blood volume, putting them at risk for infusion-related reactions and intolerance toward products that have a high infusion volume, acidic pH, and that are hyperosmolar.32

Patients with diabetes are at risk of increased serum glucose levels when administered IVIG products stabilized with sugar. However, only those products in which glucose is the stabilizing agent will increase glucose levels in the blood. Sucrose and maltose will not have this elevating effect; but maltose can potentially cause false-positive spikes in certain glucose monitor readings.32 Table 3 presents information regarding what IVIG product characteristics to avoid in patients with various risk factors. Table 4 represents the key IVIG product characteristics of selected products that may impact the choice of products in selected patient populations.

Table 3. IVIG Product Characteristics to Avoid in Patients with Specific Risk Factors10,31
Patient Risk Factors High Sugar Content High Sodium Content Volume Load High Osmolality/Osmolarity IgA Low pH
Renal Impairment X X X X    
Cardiovascular Disease   X X X    
Thromboembolic Risk   X X X    
Older Adult Patients X X X X    
Diabetes Mellitus Xa          
Pediatric/Neonatal Patients   X X X   X
Anti-IgA Antibodies         X  
IVIG = intravenous immunoglobulin; IgA = immunoglobulin A
aGlucose-containing products
Table 4. IVIG Key Product Characteristics
Product Stabilizer Sodium Content Osmolality/Osmolarity IgA Content
Bivigam 10% Glycine 100 - 140 mEq/L ≤ 510 mOsm/kg ≤ 200 mcg/mL
Carimune NF 5% sucrose
(3 g vial only)
< 20 mg/g
protein
In sterile water:
3%, 192 mOsm/kg;
6%, 384 mOsm/kg;
12%, 768 mOsm/kg
1000-2000
mcg/mL (6%)
Flebogamma 5% DIF
Flebogamma 10% DIF
5% sorbitol (polyol) Trace
(< 3.2 mEq/L)
326 ± 5.1 mOsm/kg (5%)
343 ± 6.1 mOsm/kg (10% )
< 3.2 mcg/mL
Gammagard Liquid 10% Glycine No sodium added 240 – 300 mOsm/kg 37 mcg/mL
Gammagard S/D 5% 2% glucose, glycine 0.85% 636 mOsm/L (5%);
1250 mOsm/L (10%)
< 1 mcg/mL
Gamunex-C 10% Glycine Trace
(< 7 mEq/L)
258 ± 3 mOsm/kg 46 mcg/mL
Octagam 5% 10% maltose ≤ 30 mEq/L 310 – 380 mOsm/kg < 200 mcg/mL
Privigen 10% Proline Trace 240 – 440 mOsm/kg ≤ 25 mcg/mL
IgA = immunoglobulin A; NF = nanofiltered; S/D = solvent/detergent;

Pharmacoeconomic Considerations

In addition to specific product characteristics and patient risk factors, the price of IVIG products and other related costs may also factor into the selection of a product. Pharmacy and Therapeutics (P&T) committees often considered patient demographics and patient risk factors, tolerability and cost when deciding to place a specific product on formulary. The average wholesale prices differ among products and can be found in the Red Book; however, other factors such as contract pricing can affect the final price a health system of institution pays for IVIG products. Other related costs that affect formulary and product seletion decisions that impact pharmacy, nursing, and patient care include preparation time, infusion time, storage requirements, and management of adverse events.12

Patient Counseling and IVIG Administration Strategies

When counseling a patient about IVIG therapy, the commonly observed adverse effects should be communicated. The patient should understand that they may experience infusion-related reactions that occur more frequently during their early doses and that these reactions typically alleviate after a few hours.15-24 During the infusion, they should also be monitoring for common systemic side effects. When counseling a patient about the proper storage of IVIG medications, always refer to the specific package insert storage instructions.

When considering dosing and infusion rates, high-concentration and high-infusion rate IVIG administration should be reserved for patients found to have no risk factors for severe adverse events that have been associated with IVIGs.31 When treating a patient with comorbidities, there are many things to consider while selecting the most appropriate agent. Subcutaneously administered products typically have much faster infusion rates than those recommended for IVIG administration; refer to specific product information for recommendations.

Patients who have, or are at risk of developing, renal impairment should address concerns and conduct assessments prior to selecting and administering IVIG therapy. The patient should be adequately hydrated and serum creatinine, blood urea nitrogen, and urinary output should be monitored. If available, select an agent that has been stabilized with something other than sugar, avoiding sucrose in particular. An isotonic preparation is also preferred for these patients.32

Select products that are low in sodium content, have small volume load (10% solutions are preferred), and have isotonic osmolality when treating a patient with cardiovascular disease or one who is at risk for thromboembolic event.32

After careful selection, when starting a patient on IVIG therapy, begin with a slow infusion rate and titrate based on tolerability. If adverse events are observed or experienced by the patient, a decision to either slow the rate of infusion or stopping it will have to be made. Whenever reasonable, slowing the infusion should be the first course of action.33 However, if slowing the rate of infusion does not immediately improve tolerability, the infusion should be stopped and the necessary treatment for the adverse event should be provided. Initial, maintenance, and maximum infusion rates for each product can be found in their respective package inserts.33

There are times when switching among products will be necessary because of the following: clinical efficacy, hospital formulary, patient tolerability, and availability.33 When changing to another product, it is important to increase the monitoring of the patient and infuse at a conservative rate, as if dosing a patient who is receiving his or her first dose or a dose early in the treatment. The variability in product characteristics may affect how the patient tolerates the new IVIG therapy.

Other Formulations and Routes of Administration

Immune globulin therapy was originally administered as either intramuscular (IM) or subcutaneous (SC) injections. After the IV formulation was introduced in the 1970s, these products were primarily administered via IV or SC routes.39

Several immunoglobulin products are administered by SC infusions. In addition to IV infusion, Gammagard Liquid and Gamunex-C can also be administered subcutaneously, but only for their primary humoral immunodeficiency indications.18,22 Two immunoglobulin products on the market are only administered by SC infusions. The first is Hizentra, which is indicated for the treatment of primary humoral immunodeficiency in adults and pediatric patients 2 years of age and older.40 HyQvia is another SC immunoglobulin product indicated for primary humoral immunodeficiency treatment in adults. Along with immune globulin, HyQvia also consists of recombinant human hyaluronidase. The hyaluronidase is administered first, followed by the immune globulin portion of HyQvia within 10 minutes.41

Currently in the United States, GamaSTAN S/D is the only IM formulation of immunoglobulins on the market. The approved indications and uses for this product are unique compared with the IV products. GamaSTAN S/D is indicated for hepatitis A prophylaxis, prevention or modification of measles (rubella/rubeola), passive immunization against varicella in immunosuppressed patients, and prophylaxis of rubella in early pregnancy.42 This product must be administered intramuscularly.

Conclusion

Pharmacists have an essential role in optimizing treatment for patients who require immunoglobulin therapy. The majority of products are manufactured as an IV formulation and are FDA approved for the treatment of several immunodeficiency and autoimmune disorders. There are substantial differences in terms of product characteristics (e.g., stabilizing agent, salt content, pH) among the available IVIG products. The manufacturing processes and preparation methods for each product will determine the characteristics most likely to impact their safety profiles. Patient risk factors (e.g., renal insufficiency, predisposition for thromboembolism) may elevate the patient’s risk for experiencing various adverse events, some of which can be avoided by selecting appropriate products with the right characteristics. Pharmacists can make a substantial impact by understanding IVIG products and their suitability for the treatment of different patient populations in consideration of risk factors. This will allow them to make recommendations and help select the ideal product.

References

  1. Jolles S, Sewell WA, Misbah SA. Clinical uses of intravenous immunoglobulin. Clin Exp Immunol. 2005;142(1):1-11.
  2. Radosevich M, Burnouf T. Intravenous immunoglobulin G: trends in production methods, quality control and quality assurance. Vox Sang. 2010;98(1):12-28.
  3. Baxter. How Plasma-Derived Treatments are Produced. Baxter Web site. http://www.baxter.com/press_room/factsheets/plasma_therapies_overview.html. Accessed April 12, 2015.
  4. DonatingPlasma. Donor Frequently Asked Questions. DonatingPlasma Web site. http://www.donatingplasma.org/donation/donor-faq. Accessed April 12, 2015.
  5. Plasma Protein Therapeutics Association (PPTA). What is Plasma? PPTA Web site. http://www.pptaglobal.org/plasma/what-is-plasma. Accessed April 12, 2015.
  6. U.S. Food and Drug Administration (FDA). Additional Standards for Human Blood and Blood Products, 21 C.F.R § 640.65 (2014). FDA Web site. http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=640.65. Accessed June 3, 2015.
  7. Plasma Protein Therapeutics Association (PPTA). International Quality Plasma Program (IQPP): Qualified Donor Standard. PPTA Web site. http://www.pptaglobal.org/images/IQPP/QualifiedDonorStdFinal1.pdf. Accessed April 12, 2015.
  8. Donor Selection. CSL Behring Web site. http://www.cslbehring.com/quality-safety/integrated-safety-system/plasma-donor-selection.htm. Accessed April 12, 2015.
  9. Plasma Protein Therapeutics Association (PPTA). QSEAL Inventory Hold Standard. http://www.pptaglobal.org/safety-quality/standards/qseal. Accessed April 12, 2015.
  10. Gelfand EW. Differences between IGIV products: impact on clinical outcome. Int Immunopharmacol. 2006;6(4):592-599.
  11. Matejtschuk P, Dash CH, Gascoigne EW. Production of human albumin solution: a continually developing colloid. Br J Anaesth. 2000;85(6):887-895.
  12. Shah S. Pharmacy considerations for the use of IGIV therapy. Am J Health Syst Pharm. 2005;62(16 suppl 3):S5-S11.
  13. Siegel J. Safety considerations in IGIV utilization. Int Immunopharmacol. 2006;6(4):523-527.
  14. Page M, Thorpe R. “Purification of IgG using DEAE-Sepharose Chromatography .“ In: The Protein Protocols Handbook, Second Edition. Walker JM, ed. Totowa, NJ: Humana Press; 2002:987-988.
  15. Bivigam (immune globulin intravenous [human]) [package insert]. Boca Raton, FL: Biotest Pharmaceuticals; 2014.
  16. Carimune Nanofiltered (immune globulin intravenous [human]) [package insert]. Kankakee, IL: CSL Behring; 2013.
  17. Flebogamma DIF (immune globulin intravenous [human]) [package insert]. Barcelona, Spain: Grifols USA, LLC; 2014.
  18. Gammagard Liquid (immune globulin intravenous [human]) [package insert]. Westlake Village, CA: Baxter Healthcare Corporation; 2013.
  19. Gammagard S/D (immune globulin intravenous [human]) [package insert]. Westlake Village, CA: Baxter Healthcare Corporation; 2010.
  20. Gammaked (immune globulin injection [human]) [package insert]. Research Triangle Park, NC: Grifols Therapeutics Inc.; 2013.
  21. Gammaplex (immune globulin intravenous [human]) [package insert]. Raleigh, NC: Bio Products Laboratory; 2014.
  22. Gamunex-C (immune globulin injection [human]) [package insert]. Research Triangle Park, NC: Grifols Therapeutics Inc.; 2014.
  23. Octagam (immune globulin intravenous [human]) [package insert]. Hoboken, NJ: Octapharma USA Inc.; 2014.
  24. Privigen (immune globulin intravenous [human]) [package insert]. Bern, Switzerland: CSL Behring; 2013.
  25. Horowitz B, Wiebe ME, Lippin A, Stryker MH. Inactivation of viruses in labile blood derivatives. I. Disruption of lipid-enveloped viruses by tri(n-butyl)phosphate detergent combinations. Transfusion. 1985;25(6):516-522.
  26. Horowitz B, Lazo A, Grossberg H, et al. Virus inactivation by solvent/detergent treatment and the manufacture of SD-plasma. Vox Sang. 1998;74(suppl 1):203-206.
  27. Burnouf T, Radosevich M. Reducing the risk of infection from plasma products: specific preventative strategies. Blood Reviews. 2000;14(2):94-110.
  28. Uemura Y, Yang YH, Heldebrant CM, et al. Inactivation and elimination of viruses during preparation of human intravenous immunoglobulin. Vox Sang. 1994;67(3):246-254.
  29. Chang CE, Eo HG, Lee YS, et al. Human intravenous immunoglobulin preparation and virus inactivation by pasteurization and solvent detergent treatment. Prep Biochem Biotechnol. 2000;30(3):177-197.
  30. Ballow M. Clinical and investigational considerations for the use of IGIV therapy. Am J Health Syst Pharm. 2005;62(16 suppl 3):S12-S18.
  31. Chérin P, Cabane J. Relevant criteria for selecting an intravenous immunoglobulin preparation for clinical use. BioDrugs. 2010;24(4):211-223.
  32. Siegel J. IVIG Medication Safety: A Stepwise Guide to Product Selection and Use. Pharmacy Practice News. 2010:1-8. Pharmacy Practice News Web site (McMahon Publishing). http://www.pharmacypracticenews.com/download/IVIG_safety_ppn0910_ WM.pdf. Accessed June 8, 2015.
  33. Siegel J. Immune Globulins: Therapeutic, Pharmaceutical, Cost, and Administration Considerations. Pharmacy Practice News Web site (McMahon Publishing). [Login necessary] http://www.pharmacypracticenews.com/index.asp?show=pocketguide&section_id=159 &article_id=15544. Accessed April 22, 2015.
  34. U.S. Food and Drug Administration (FDA.) Immune Globulin Intravenous (IGIV) Indications. Consumer Affairs Branch, 10 June 2013. Web. 18 Apr. 2015. FDA Web site. http://www.fda.gov/BiologicsBloodVaccines/BloodBloodProducts/ApprovedProducts/LicensedProductsBLAs/FractionatedPlasmaProducts/ucm133691.htm. Accessed June 8, 2015.
  35. Hamrock DJ. Adverse events associated with intravenous immunoglobulin therapy. Int Immunopharmacol. 2006;6(4):535-542.
  36. Wolberg AS, Kon RH, Monroe DM, Hoffman M. Coagulation factor XI is a contaminant in intravenous immunoglobulin preparations. Am J Hematol. 2000;65(1):30-34.
  37. Stiehm ER. Adverse effects of human immunoglobulin therapy. Transfus Med Rev. 2013;27(3):171-178.
  38. Pierce LR, Jain N. Risks associated with the use of intravenous immunoglobulin. Transfus Med Rev. 2003;17(4):241-251.
  39. Caspi A, Sherritze S, Robins C, et al. Privigen immune globulin intravenous (human), 10% liquid. P T (Product Profiler) 2011;36(8):2-31.
  40. Hizentra (immune globulin subcutaneous [human]) [package insert]. Bern, Switzerland: CSL Behring; 2015.
  41. HyQvia (immune globulin infusion 10% [human]) [package insert]. Westlake Village, CA: Baxter Healthcare Corporation; 2014.
  42. GamaSTAN S/D (immune globulin [human]) [package insert]. Research Triangle Park, NC: Grifols Therapeutics Inc.; 2013.

Back to Top