Immunoglobulin therapy plays an increasingly important role in the treatment of a variety of medical conditions, not only to prevent or ameliorate infection but also because of antiinflammatory and immunomodulating effects. Passive immunization provides exogenous, preformed antibodies that can prevent or treat certain infectious diseases. In the early 20th century, hyperimmune animal sera were produced to treat specific infections. After human plasma fractionation was developed (during World War II), immune globulin (human) (IG) became available for passive immunization. Although this development was a major breakthrough, intravenous (IV) infusion of this product evoked severe adverse reactions that made IV administration not feasible. Standard IG continues to be limited to intramuscular (IM) or subcutaneous administration. For other indications, such as for management of a primary immunodeficiency disorder, the volume and hence the amount of immunoglobulin G (IgG) that can be administered by the IM route often is suboptimal. In 1981, the US Food and Drug Administration (FDA) approved the first US-licensed (human) intravenous immune globulin (IGIV). IGIV enables IV administration of large doses of IgG with minimal discomfort, producing an immediate rise in both total plasma IgG concentration and concentrations of specific antibodies.

The products available for passive immunization can be grouped into four basic categories: (1) standard IG for IM administration (hepatitis A, measles, rubella); (2) hyperimmune polyclonal globulins that can be administered by the IM route (hepatitis B IG [HBIG], rabies IG, tetanus IG [TIG], varicella IG) or by the IV route (cytomegalovirus [CMV], vaccinia, botulism IGs); (3) standard IGIV that can be administered subcutaneously or intravenously; and (4) monoclonal antibodies (mAbs) ( Table 5.1 ). In addition, two antitoxins of animal origin are available for limited distribution. (An antitoxin is an antibody with the ability to neutralize a specific toxin.)

TABLE 5.1
Immune Globulins Prepared From Human Plasma
Nonproprietary Name Abbreviation
For Intramuscular Administration
Standard immune globulin (human) IG
Hepatitis B immune globulin (human) HBIG
Rabies immune globulin (human) a RIG
Tetanus immune globulin (human) TIG
Varicella-zoster immune globulin (human) b VariZIG
For Intravenous Administration
Immune globulin intravenous (human) IGIV
Cytomegalovirus immune globulin intravenous (human) CMV-IGIV
Botulism immune globulin intravenous (human) BIG-IGIV
Vaccinia immune globulin intravenous (human) VIG-IGIV
Anthrax immune globulin intravenous (human) AIG-IGIV
AIG, anthrax IG; CMV, cytomegalovirus; HBIG, hepatitis BIG; IG, immune globulin (human); IGIV, intravenous immune globulin; RIG, rabies immunoglobulin; TIG, tetanus IG; VIG, vaccinia IG (human).

a As much of the dose as possible should be instilled around the wound.

b See text.

Passive immunization is used for a variety of indications, including the following: (1) treatment of a primary immunodeficiency and, in certain cases, a secondary immunodeficiency; and (2) prophylaxis against infections with specific organisms. IG is less useful for treatment of established infections. However, the use of IGIV for the treatment of certain conditions that involve immune dysregulation, such as immune thrombocytopenic purpura (ITP) and Kawasaki disease, is the standard of care.

In the following sections, the various products and their uses are discussed. Pathogen-specific and disease-specific chapters should be consulted. An effort has been made to use nonproprietary product names as they appear in the labeling, but for brevity, abbreviations are used. Emphasis is placed on FDA-approved indications (i.e., those that appear in the product package inserts). In the case of IGIV, not all products have been approved or studied for all indications ( Table 5.2 ). Moreover, in contrast to IG for IM administration, IGIV products are produced by a variety of manufacturing processes that use different stabilizers and excipients, various concentrations, and physical states and they can differ in IgA content.

TABLE 5.2
Available Immune Globulin Intravenous Products in the US
Product Registered Name Concentration, Route Manufacturer FDA-Approved Indication
Bivigam 10% (IV) Biotest Pharmaceuticals PID
Gammaplex 5%, 10% (IV) Bio Products Laboratory ITP, PID
Carimune NF
Lyophilized (IV)
CSL Behring ITP, PID
Privigen, 10% (IV) CSL Behring PID, chronic ITP
Flebogamma DIF
5%, 10% (IV)
Instituto Grifols, SA PID
Gammagard Liquid 5% (IV, SC) Baxter Healthcare PID, MMN
Gammaplex 5% (IV) Baxter Healthcare CLL, ITP, KD, PID
Gamunex-C 10% (IV, SC) Grifols Therapeutics, Inc. ITP, PID, CIDP
Octagam 5%, 10% (IV) Octapharma USA PID
GamaSTAN S/D 15%–18% (IM) Grifols Therapeutics, Inc. Hepatitis A, measles, varicella prophylaxis
Hizentra 20% (SC) CSL Behring AG PID
HYQVIA 10% (SC) Baxter Healthcare PID
Privigen 10% (IV) CSL Behring AG PID, ITP
Cuvitru 20% (SC) Octapharma PID
Gammaked 10% (IV) Takeda PID, ITP
WinRho SDF Cangene Chronic or acute ITP and HIV-immune related conditions
CIDP, chronic inflammatory demyelinating polyneuropathy; CLL, chronic lymphocytic leukemia; IM, intramuscular; ITP, immune thrombocytopenia purpura; KD, Kawasaki disease; PID, primary immunodeficiency; MMN, multifocal motor neuropathy; SC, subcutaneous.

Standard Immune Globulin (Human)

IG is prepared by cold alcohol fractionation of pooled human plasma. It is formulated as a 16.5% protein solution. At least 96% of the total protein is IgG, but small quantities of IgM and IgA may be present. Each lot represents the pooled plasma of at least 1000 donors, ensuring a wide diversity of antibodies. A single lot may represent up to 60,000 donors. Similar to all human plasma-derived products, donors are screened for certain viruses (hepatitis B, hepatitis C, human immunodeficiency viruses 1 and 2 [HIV-1 and [HIV-2], human T-lymphotropic virus 1 and 2 [HTLV-1 and HTLV-2], West Nile virus, Zika virus) as well as Trypanosoma cruzi and Treponema pallidum, and, in endemic areas, Babesia microti, to minimize the potential for transmission. Other steps taken to minimize such transmission are noted at the end of this chapter.

Hepatitis A

A major use of IM standard IG is for preexposure and postexposure prophylaxis of hepatitis A virus (HAV) in a susceptible person. When administered within 14 days of exposure, IG is 80%–90% effective in preventing clinical hepatitis. IG is most effective when administered early in the incubation period. When administered later in the incubation period, IG still may attenuate HAV disease. The usual dose of IM IG varies by age and should be administered as soon as possible after exposure. For persons 12 months–40 years of age, HAV vaccine is preferred for postexposure prophylaxis. IG is recommended for people >40 years of age because of the absence of data regarding vaccine use in adults >40 years of age and the severity of HAV infection in older adults. Children <12 months of age, immunocompromised people, people with chronic liver disease, and others who may have a diminished response to vaccine should receive IG.

IG may be effective for preexposure prophylaxis in travelers to areas where HAV is prevalent. However, for travelers who are ≥1 year old and whose departure is not imminent, HAV vaccine largely has replaced IG. Due to concern that preexposure prophylaxis with IG may decrease the effectiveness of measles, mumps, and rubella (MMR) vaccine (which is recommended for infants aged 6 months–11 months who are traveling internationally), an off-label dose of HAV vaccine instead of IG is recommended. The dose of IG depends on the age of the traveler and the length of stay. For a child <1 year old, the dose is 0.01–0.02 mL/kg if the anticipated stay is ≤2 months and 0.06 mL/kg if longer. In older persons whose departure is imminent, the dose of IG is 0.01–0.02 mL/kg, concomitant with the first dose of vaccine, for a stay of up to 5 months, and 0.06 mL/kg, concomitant with the first dose of vaccine, for a longer stay.

For people ≥12 months of age for whom IG is given for preexposure or postexposure prophylaxis, hepatitis A vaccine is given simultaneously, at a separate site and using a separate syringe.

Measles

Vaccination is the major strategy for achieving protection against measles. More than 98% of persons who receive 2 doses of measles vaccine (with the first dose administered no earlier than the first birthday and the second dose administered at least 28 days after the first dose) develop serologic evidence of immunity. Nonetheless, measles postexposure prophylaxis with IG is important for children <12 months of age (for age 6–11 months, MMR vaccine can be substituted for IG if within 72 hours of exposure), seronegative persons who cannot be vaccinated within 72 hours of exposure, seronegative pregnant women, and immunocompromised children not receiving routine immunoglobulin replacement therapy. Prophylaxis is indicated for susceptible household or hospital contacts of a case. A single dose of 0.5 mL/kg administered within 6 days of exposure may prevent or modify disease and provide temporary protection. No more than 15 mL should be injected; small children should be given no more than 3 mL at any single site. Manufacturer instructions should be consulted for maximum volume injected into a single muscle. A child receiving routine IgG replacement therapy is likely to have a protective concentration of measles-neutralizing antibody. If the child is ≥12 months of age, live measles vaccine should be administered ~6 months after IG administration if no contraindication exists. For children 6–11 months of age who are traveling to areas where measles is endemic, MMR vaccine (rather than IG) should be administered; this dose is not counted in the required 2-dose series at ≥12 months of age. IGIV is recommended for seronegative pregnant woman and for severely immunocompromised persons.

Rubella

IG can be considered for prophylaxis of rubella-susceptible women who are exposed to rubella early in pregnancy. As many as 85% of infants born to mothers who are infected in the first trimester of pregnancy will have signs of congenital rubella syndrome. The IG option is recommended only for women if termination of pregnancy is declined. Limited data indicate that IM IG in a dose of 0.55 mL/kg may decrease clinically apparent infection, virus shedding, and rates of viremia in infected susceptible people. Theoretically, the reduction in viral replication may decrease the likelihood of fetal infection. The absence of clinical signs in a woman who has received IG does not guarantee that fetal infection has been prevented. Infants with congenital rubella have been born to mothers who received IG shortly after exposure.

Specific Immune Globulins for Intramuscular Administration

Specific hyperimmune IGs for IM use are IgG preparations produced from plasma that is selected by screening donations or collected from donors who have been purposefully immunized or who have received a booster. Either approach can ensure the presence of high concentrations of antibody directed against one or more specific antigens. The manufacturing process is essentially the same as that used to prepare IG. The protein concentration differs for individual products and may be found in the package insert. Specific products available for IM administration are listed in Table 5.1 . Hyperimmune globulins are used to prevent hepatitis B, rabies, tetanus, and varicella-zoster virus (VZV) infection. A hyperimmune globulin typically contains antibody against the intended virus or toxin that is 8–10 times higher than standard IG.

Hepatitis B Immune Globulin

All pregnant women should be tested for circulating hepatitis B surface antigen (HBsAg). Infants born to women who are HBsAg positive (either acutely or chronically infected with hepatitis B virus [HBV]) are at high risk of developing acute and chronic HBV infection. The HBV vaccination series accompanied by a single dose of HBIG is 85%–95% effective in preventing HBV infection in the infant. Infants born to an HBsAg-positive mother should receive HBIG (0.5 mL) within 12 hours of birth. The first of 3 doses of HBV vaccine should be given at the same time as HBIG but at a different site.

In general, HBIG is not recommended if the mother’s HBsAg status is not known, but HBV vaccination series should be begun within 12 hours of birth. Ideally, in such a case, the mother should be tested for HBsAg as soon as possible after delivery. If the test result is positive, the infant should receive 0.5 mL of HBIG IM as soon as possible (within 12–24 hours) and if delay is unavoidable, at least within 7 days of birth; vaccination series should be completed according to schedule. Preterm infants (<2 kg) constitute a special class. If the mother’s HBsAg status is unknown and cannot be determined within 12 hours, the neonate should be given 0.5 mL of HBIG as well as vaccine to provide passive antibody before onset of the active immune response to the vaccine.

After a percutaneous (needlestick, laceration, or bite) or mucosal exposure that contains or may contain HBV, blood should be obtained from the person who was the source of the exposure (if possible) to determine his or her HBsAg status. Management of the exposed person depends on the HBsAg status of the source and the vaccination and antibody to HBsAg response status of the exposed person. Recommended postexposure prophylaxis for different situations has been outlined by the Centers for Disease Control and Prevention (CDC).

Human Rabies Immune Globulin

Rabies IG (human) (HRIG) is always used in association with rabies vaccine for postexposure prophylaxis. , HRIG is administered to previously unvaccinated persons to provide rabies virus-neutralizing antibody until the patient responds to vaccination by actively producing virus-neutralizing antibodies. HRIG is administered once, at the same time postexposure prophylaxis is initiated with the human rabies vaccine.

The dose of HRIG is 20 IU/kg. If anatomically feasible, the dose should be infiltrated around the wound, and any remaining dose should be administered by the IM route at an anatomic site distant from the vaccine administration. HRIG is supplied in 2-mL (300 IU) and 10-mL (1500 IU) vials with an average potency of 150 IU/mL. Thus the contents of a 2-mL vial can be used to treat a child of up to 15 kg; for larger children, additional vials are required. A 10-mL vial is sufficient to treat a 75-kg adult and as a result is less suitable for pediatric use. However, if the smaller vials are not available, an appropriate portion can be withdrawn to treat a child. The product contains no preservative; once a vial has been entered, the contents should be administered promptly, and the remainder should be discarded.

Tetanus Immune Globulin

TIG was one of the first specific IG products. TIG is recommended for the treatment of tetanus and for prophylaxis in certain circumstances. A patient with tetanus should receive a single dose of TIG, although the optimal dose has not been determined. For postexposure prophylaxis, the need for active immunization with or without passive immunization depends on the condition of the wound and the patient’s immunization history. Specific guidance has been provided by the Advisory Committee on Immunization Practices. Persons with wounds that are neither clean nor minor and who have had 0–2 previous doses of tetanus toxoid or have an uncertain history of previous doses should receive TIG, as well as diphtheria and tetanus toxoids (DTs), tetanus and diphtheria toxoids (Td), diphtheria and tetanus toxoids and acellular pertussis (DTaP), or tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis (Tdap), depending on age and other needed vaccines. TIG can neutralize only unbound tetanus toxin; it cannot affect toxin bound to nerve endings. A single IM dose of 3000–5000 units TIG is generally recommended for treatment of children and adults, with part of the dose infiltrated around the wound (250 units is maximum used for prophylaxis). IGIV contains tetanus antitoxin and can be used if TIG is not available.

Varicella-Zoster Immune Globulin

Postexposure prophylaxis should be targeted toward patients without evidence of immunity to varicella who are at high risk for severe disease and complications and who have been exposed to and likely infected by VZV. Such persons include the following: (1) immunocompromised people; (2) nonimmune pregnant women; (3) neonates whose mothers develop signs and symptoms of varicella within 5 days before delivery to 48 hours after delivery; (4) premature infants born at 28 weeks of gestation or later who are exposed in the neonatal period and whose mothers do not have evidence of immunity; and (5) premature infants born before 28 weeks of gestation or who weigh <1000 g at birth and are exposed during the neonatal period regardless of maternal history of varicella disease or vaccination (because transfer of maternal antibody may not have occurred). Significant exposure can occur through residence in the same household, close contact with a playmate, proximity to a contagious patient, or contact with a contagious visitor or hospital staff member.

In December 2012, the FDA approved VariZIG, a varicella-zoster IG preparation (Cangene Corporation, Winnipeg, Canada) for postexposure prophylaxis for persons at high risk of severe disease who lack evidence of immunity to varicella and for whom varicella vaccine is contraindicated. VariZIG is a lyophilized product and, after reconstitution, is administered IM. VariZIG is approved for administration as soon as possible following VZV exposure, ideally within 96 hours (4 days) for greatest effectiveness, but if it is administered within 10 days, it may result in disease attenuation. The dose is 125 U (1 vial)/10 kg body weight up to a maximum of 625 IU (5 vials). The minimum dose is 125 IU, or 62.5 IU if weight is ≤2 kg.

If VariZIG cannot be obtained within 96 hours after exposure, IGIV can be substituted at a dose of 400 mg/kg. Although licensed IGIV preparations are known to contain varicella antibody, the titer of any specific lot is uncertain. An alternative to postexposure prophylaxis with VariZIG or IGIV is administration of oral acyclovir or valacyclovir, which should be started 7–10 days after exposure and continued for a total of 7 days. This approach can be considered in immunocompetent, seronegative people with significant exposure who cannot be vaccinated. The oral acyclovir dose for adults is 800 mg given 4 times a day. If a child requires acyclovir preemptively, the dose is 40–80 mg/kg/day, divided into 4 doses (maximum daily dose 3200 mg) or valacyclovir 60 mg/kg/day divided into 3 doses (maximum daily dose 3000 mg). As an alternative, exposed immunocompetent children and adults can be given varicella vaccine within 3–5 days of exposure if they have not previously received 2 doses of vaccine and have no contraindication.

Immune Globulin Intravenous (Human)

IGIV, like IG, is prepared from large pools of donor derived plasma. The early steps in IGIV manufacture are similar to those used for preparing IG. Further processing of individual products is performed by a variety of techniques, including steps such as polyethylene glycol precipitation, ion exchange chromatography, and exposure to low pH. This variation reflects, in part, the lack of consensus on the optimal procedure for preparing IGIV on a commercial scale in a form that is safe for IV administration. Final formulations are diverse, such as freeze-dried or in solution, different protein concentrations of the solution, different pH, and various stabilizers and other excipients that can be used in several different combinations. The product package label should be consulted for a synopsis of the product’s preparation and properties.

Each lot of IGIV must contain minimum levels of antibodies to certain infectious organisms or toxoids (measles, poliovirus, and diphtheria). These concentrations were established to ensure lot-to-lot consistency rather than to match the clinical indications for individual products. Even though the use of large plasma pools enhances consistency, IGIV products differ in the concentration of antibodies to specific antigens. As a result, antibody titers to other bacterial and viral pathogens can vary significantly among preparations and lots. , In addition, processing steps can alter functional capabilities of IgG or change the relative distribution of immunoglobulin subclasses. These changes may or may not be reflected in antibody titers measured by nonfunctional assays (e.g., enzyme-linked immunosorbent assay [ELISA]). In addition, individual products have undergone clinical trials for only a certain subset of indications. Even when several IGIV products carry the same indication, direct comparisons of the efficacy of multiple products in a single clinical trial are uncommon. For these reasons, IGIV cannot be considered a uniform generic product. Available products in the US are shown in Table 5.2 . Certain physiologic properties intrinsic to IGIV preparations (and variable among products) should be considered in all persons for whom administration is considered and especially in persons who have underlying conditions. Such considerations include the following: high volume load, sodium content, and osmolality, especially in neonates and young children, as well as in patients with cardiac disease, renal impairment, or thromboembolic risk; 2%–10% carbohydrate content in children with diabetes or renal impairment; low pH in neonates; and IgA content in persons who may have preexisting anti-IgA antibodies.

Approved Indications

Current FDA-approved available IGIV products in the US have the following indications: (1) replacement therapy for primary immunodeficiency disorders associated with defects in humoral immunity; (2) treatment of ITP to induce a rapid rise in platelet count when needed to prevent or control bleeding; (3) prevention of coronary artery aneurysms associated with Kawasaki disease; (4) prevention of bacterial infections in patients with hypogammaglobulinemia and recurrent bacterial infections associated with B-cell chronic lymphocytic leukemia; (5) prevention of infections, pneumonitis, and graft-versus-host disease after bone marrow transplantation; (6) maintenance therapy to improve muscle strength in adults with multifocal motor neuropathy; (7) improvement of neuromuscular disability and maintenance therapy to prevent relapse of chronic inflammatory demyelinating polyneuropathy; (8) pediatric patients with HIV infection to decrease the frequency of bacterial infections and the frequency of hospitalization (IGIV use in HIV infected patients has been reduced in the era of highly active antiretroviral therapy) ( Table 5.3 ).

TABLE 5.3
Approved Indications for Immune Globulin Intravenous Therapy
Indications Dosage Comments
Replacement Therapy
Primary immunodeficiency ∼400 mg/kg q4 wk (average dose) Adjust dose according to individual response
Chronic lymphocytic leukemia 400 mg/kg q3–4 wk Cost effectiveness questioned
Bone marrow transplantation 500 mg/kg q1 wk Other antiinfective therapies also effective
Human immunodeficiency virus infection in children 400 mg/kg q2–4 wk Useful in selected symptomatic patients
Immune Modulation
Idiopathic thrombocytopenic purpura 400 mg/kg daily for 5 days, or 1 g/kg single dose Useful in acute and chronic cases
Kawasaki disease 2 g/kg single dose 10%–20% of patients may need a second dose
Multifocal motor neuropathy 400 mg/kg daily for 5 days Repeat doses dependent on clinical response
Chronic inflammatory demyelinating polyneuropathy 400 mg/kg daily for 5 days Repeat doses dependent on clinical response

Primary Immune Deficiencies

IGIV is standard replacement therapy in primary immunodeficiency, and all IGIV products include this indication. Clinical experience with IGIV in many different types of primary immunodeficiency has been favorable. Patients with the following primary immunodeficiencies have associated hypogammaglobulinemia and may benefit from replacement IGIV therapy: X-linked agammaglobulinemia, common variable immunodeficiency, severe combined immunodeficiency, hyper-IgM syndrome, congenital agammaglobulinemia, and Wiskott-Aldrich syndrome.

The objective of replacement therapy is to determine the dose and frequency that will result in freedom from serious infections. Monitoring the trough levels of IgG in parallel with the patient’s clinical course has been helpful in meeting this goal. A range of effective doses, dosing schedules, and trough levels has been observed. The usual schedule for IGIV infusion in patients with hypogammaglobulinemia is once every 3–4 weeks; the typical dose is 400 mg/kg (range, 300–800 mg/kg). Trough concentrations of IgG should be maintained in the range of 400–500 mg/dL.

Replacement therapy is unlikely to be beneficial for isolated IgE deficiency, isolated IgG subclass 4 deficiency, selective IgA deficiency, or isolated IgM deficiency.

Secondary Immune Deficiencies

Chronic lymphocytic leukemia and the immune suppression that occurs in patients undergoing bone marrow transplantation result in quantitative and qualitative humoral immunodeficiency. Although IGIV reduces infections in these conditions, unresolved issues are selection of patients most likely to benefit, optimal timing and duration of administration, and the relative effectiveness of IGIV and other antiinfective therapies.

Infection with HIV causes dysregulation of humoral immunity with impaired functional antibody activity. Some HIV-infected children have fewer bacterial infections while they are receiving IGIV; hence IGIV may benefit selected patients. With the use of combined antiretroviral therapy, antimicrobial prophylaxis (e.g., trimethoprim-sulfamethoxazole), and vaccination, the need for IGIV has decreased, but it can be considered in patients with recurrent serious bacterial infections and IgG of ≤400 mg/dL or evidence of functional antibody deficiency.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here