Coverage Policy Manual
Policy #: 1997113
Category: Pharmacy
Initiated: October 1993
Last Review: September 2023
  Immune Globulin, Intravenous and Subcutaneous

Description:
Human immune globulin replacement therapy provides a broad spectrum of opsonizing and neutralizing immunoglobulin G (IgG) antibodies against a wide variety of bacterial and viral antigens. Three formulations of human IgG are available for delivery by intravenous infusion (IVIg), by subcutaneous infusion (SCIg), or by intramuscular (IMIg) depot injections. IMIg has been largely abandoned in the U.S. because volume constraints and pain preclude delivery of sufficient product weekly into each buttock to yield therapeutic serum levels of IgG, leaving recipients susceptible to infections.
 
IVIg is an antibody-containing solution obtained from the pooled plasma of healthy blood donors that contains antibodies to greater than 10 million antigens. IVIg has been used to correct immune deficiencies in patients with either inherited or acquired immunodeficiencies and has also been investigated as an immunomodulator in diseases thought to have an autoimmune basis.  Several IVIg products are available for clinical use in the U.S.  This policy only addresses nonspecific pooled preparations of IVIg, including Gammagard (Baxter), Gamimune (Bayer), Sandoglobulin (Sandoz), Iveegam (Immuno), Venoglobulin (Alpha Therapeutics), Gammar (Centeon), and Polygam (American Red Cross). This policy DOES NOT address other immunoglobulin preparations that are specifically used for passive immunization to prevent or attenuate infection with specific viral diseases such as respiratory syncytial virus, cytomegalovirus, or hepatitis B.
 
One IVIG product (Gamunex) received FDA approved indication for treatment of chronic Inflammatory demyelinating polyneuropathy in September 2008 (FDA, 9/12/08).
 
Subcutaneous infusion immune globulin is used for treating patients with primary immunodeficiencies (PID). A genetic basis for more than 80 different types of PID has been discovered, the most common being primary antibody deficiency (PAD) that is associated with low levels or total lack of normal circulating immunoglobulins. The first FDA-approved SCIg product, Vivaglobin, is a pasteurized, polyvalent human normal immune globulin product that is manufactured from large pools of human plasma by cold alcohol fractionation with no chemical or enzymatic alterations. Vivaglobin administration produces relatively stable steady-state serum levels of IgG that are representative of those seen in a normal human population. Applications of this product for conditions other than primary immunodeficiencies are considered off-label in the United States and are not addressed in this policy. In recent years, other SCIg products have also received FDA-marketing approval.
 
Regulatory Status
Several IVIg products have been approved by FDA. These include Carimune® (ZLB Bioplasma), Flebogamma® (Grifols), Gammagard® (Baxter), Gamunex-C® (Grifols), Octagam® (Octapharma), Polygam® S/D (Baxter) Privigen® (CSL Behring), and BIVIGAM™ (Biotest Pharmaceuticals).
At least 1 IVIg product is FDA-approved to treat the following conditions:
  • Primary Humoral Immunodeficiency
  • Multifocal Motor Neuropathy
  •  B-cell Chronic Lymphocytic Leukemia
  • Immune (aka Idiopathic) Thrombocytopenic Purpura
  •  Kawasaki syndrome
  • Chronic Inflammatory Demyelinating Polyneuropathy
 
Several SCIg products have received FDA marketing approval for primary immunodeficiencies. These include Vivaglobin® (ZLB Behring, Kankakee, IL, discontinued by the company in 2013), Hizentra® (ZLB Behring, Kankakee, IL), Gamunex-C® (Grifols), and Gammaked® (Kedrion Biopharma, Cambridge, MA).
 
Coding
There are CPT and HCPCS codes that describe IVIg and SCIg products:
90283: Immune globulin (IgIV), human, for intravenous use
90284:  Immune globulin (SCIg), human, for use in subcutaneous infusions, 100 mg, each
J1459:  Injection, immune globulin (Privigen), intravenous, non-lyophilized (eg, liquid), 500 mg
J1557:  Injection, immune globulin (Gammaplex), intravenous, non-lyophilized (eg, liquid), 500 mg
J1556: Injection, immune globulin (bivigam), 500 mg
J1559   Injection, immune globulin (Hizentra), 100 mg
J1561:  Injection, immune globulin (Gamunex-C/Gammaked), non-lyophilized (eg, liquid), 500 mg
J1562:  Injection, immune globulin (Vivaglobin), subcutaneous, 100 mg
J1566:  Injection, immune globulin, intravenous, lyophilized (eg, powder), not otherwise specified, 500 mg
J1568:  Injection, immune globulin (Octagam) intravenous, non-lyophilized (eg, liquid), 500 mg
J1569:  Injection, immune globulin (Gammagard Liquid) intravenous, non-lyophilized (eg, liquid), 500 mg
J1572:  Injection, immune globulin (Flebogamma/Flebogamma DIF), intravenous, non-lyophilized (eg, liquid), 500 mg
J1575  Injection, immune globulin/hyaluronidase, 100 mg immunoglobulin
J1599:  Injection, immune globulin, intravenous, non-lyophilized (eg, liquid), not otherwise specified, 500 mg
J1554   Injection, immune globulin (asceniv), 500 mg
J1551   Injection, immune globulin (cutaquig), 100 mg (Effective July 1, 2022)
 
The following CPT drug administration codes would be used for the administration of these products:
96365-96366 for intravenous infusions; and
96369-96371 for subcutaneous infusions
 

Policy/
Coverage:
For contracts subject to Arkansas Act 1054/Act 876(f) USE OF DRUG TREATMENTS TO TREAT PATIENTS DIAGNOSED WITH PEDIATRIC ACUTE- ONSET NEUROPSYCHIATRIC SYNDROME AND PEDIATRIC AUTOIMMUNE NEUROPSYCHIATRIC DISORDERS ASSOCIATED WITH STREPTOCOCCAL INFECTION [individual, blanket, or any group plan, policy, or contract for healthcare services issued, renewed, or extended in this state by a healthcare insurer, health maintenance organization, hospital medical service corporation, or self-insured governmental or church plan in this state (all fully-insured and state government such as ASE/PSE, ASP)] coverage shall be provided for use of intravenous immunoglobulin, also known as "IVIG", to treat individuals diagnosed with pediatric acute-onset neuropsychiatric syndrome or pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection, or both, on or after January 1, 2024, if the pediatric patient's primary care physician, in consultation with an Arkansas licensed pediatric psychiatrist and an Arkansas licensed physician who practices in at least one (1) pediatric subspecialty, including a neurologist, rheumatologist, or infectious disease physician who has treated the pediatric patient determines and agrees that the treatment is necessary and follows a patient-specific treatment plan. Effective April 13, 2023, this benefit and associated drug treatment NO LONGER REQUIRES prior authorization.
 
For contracts not subject to Arkansas Act 1054/Act 876, the use of IVIG in the treatment of PANDAS/PANS is NOT COVERED.
 
For members of plans that utilize an oncology benefits management program, Prior Authorization is required for this service and is managed through the oncology benefits management program for oncologic related indications.
 
Effective January 1, 2022
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
SUBCUTANEOUS IMMUNOGLOBULIN
 
SCIg may meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness for the treatment of primary immunodeficiencies, including:
 
        • congenital agammaglobulinemia,
        • hypogammaglobulinemia,
        • common variable immunodeficiency (CVID),
        • severe combined immunodeficiency,
        • Wiskott-Aldrich syndrome,
        • X-linked agammaglobulinemia (XLA); OR
        • when coverage criteria have been met for intravenous immune globulin (IVIG) but patient is unable to tolerate IVIG because of side effects from the IVIG treatment. (van Schaik, 2018)
 
INTRAVENOUS IMMUNOGLOBULIN (FDA APPROVED INDICATIONS)
 
The administration of intravenous immune globulin meets member benefit certificate primary coverage criteria for effectiveness for the following FDA approved indications:
 
        • Bone marrow transplantation: to prevent risk of acute graft-versus-host disease, associated interstitial pneumonia (infectious or idiopathic), and infections (e.g., CMV, varicella-zoster viral infection, and recurrent bacterial infection) after allogeneic bone marrow transplantation in patients 20 years of age or older in the first 100 days after transplantation;
        • Chronic inflammatory demyelinating polyneuropathy (FDA approved indication) when all of the following criteria are documented:
            • Progressive or relapsing motor AND sensory dysfunction of more than one limb; AND
            • Significant disability in arm or leg function developing over at least two months; AND
            • Hypo- or areflexia; AND
            • One of the following:
                • partial conduction block or abnormal temporal dispersion of conduction must be present in at least 2 nerves and there must be significantly reduced conduction velocity, or significantly prolonged distal motor latency, or absent or significantly prolonged minimum Fwave latency in at least one other nerve; OR
                • in the absence of block or dispersion, significantly reduced conduction velocity, or significantly prolonged distal motor latency, or absent or significantly prolonged minimum F-wave latency must be present in at least 3 nerves; OR
                • in the presence of significant neurophysiologic abnormalities in only 2 nerves, unequivocal histological evidence of demyelinated nerve fibers in a nerve biopsy must also be present. CSF Studies are encouraged. An elevated protein with a CSF white count of <10/mm3 in HIV-1 negative patients would be expected in patients with CIDP.
        • Chronic immune thrombocytopenic purpura (ITP).
        • Chronic Lymphocytic Leukemia with associated hypogammaglobulinemia in patients (FDA approved indication) who have demonstrated frequent and recurrent bacterial infections or who demonstrate impaired response to bacterial polysaccharides or proteins (study results);
        • Common Variable Immunodeficiency (panhypogammaglobulinemia)
        • Congenital Agammaglobulinemia (X-linked agammaglobulinemia)
        • Dermatomyositis (DM)
        • Human immunodeficiency virus infection (HIV) in children to decrease the risk of serious bacterial infection (FDA approved indication). IVIG is covered only in children who have IgG less than 400 mg/dL or functional antibody deficiency is demonstrated by poor specific antibody titers or recurrent bacterial infections (ACIP, AAP, CDC, NIH, IDSA, PIDS recommendation).
        • Idiopathic thrombocytopenic purpura, acute, or chronic, when a rapid rise in the platelet count is required, such as prior to surgery or to prevent/control excessive bleeding (FDA approved indication) when the platelet count is less than 30,000 (study results).
        • Kawasaki’s Disease (Mucocutaneous lymph node syndrome) when used in conjunction with ASA and treatment is started within the first 10 days of illness (FDA approved indication) or as soon as possible after the disease is diagnosed or strongly suspected (AAP, AHA, ACCP recommendation);
        • Multifocal Motor Neuropathy (MMN) when used as initial and maintenance therapy to improve muscle strength and disability in adult patients. (EFFECTIVE June 2013)
        • Primary Humoral Immunodeficiency (PI)  
        • Severe Combined Immunodeficiency
        • Toxic Shock Syndrome  
        • Wiskott-Aldrich Syndrome
        • X-linked hyper-IgM syndrome and ataxia telangiectasia  (Effective January 2017)
        • X-linked immunodeficiency hyperimmunoglobulin IgM
 
OFF LABEL INDICATIONS FOR INTRAVENOUS IMMUNOGLOBULIN
 
These off label indications are considered eligible for coverage based on one or more of the following:
        • High quality systematic reviews of randomized controlled trials by independent review organizations (e.g., Cochrane Systematic Reviews, AHRQ Evidence Based Practice Centers);
        • adequately powered randomized controlled trials published in peer-reviewed medical literature;
        • single or multiple randomized controlled trials, adequately populated, with risk of bias, but recommended in publications in peer-reviewed journals;
        • prospective, non-randomized, adequately populated case-control or cohort study(s) with low risk of confounding or bias;
        • expert opinion, presented as published position papers by national medical societies supporting this opinion (unless in conflict with other expert opinion); and
        • rare life threatening conditions for which:  
            • only case series have been reported in peer reviewed medical literature;
            • more than one series has reported significant, lasting improvement;
            • no conflicting evidence is known; and
            • because of the rarity of the condition, adequate appropriate trials would be unlikely.  
 
There is no coverage for IVIG for indications other than those specifically listed as covered; requests for coverage of conditions not listed in the covered section should be submitted for review, and include references from peer reviewed journals which support coverage for the proposed condition.
 
Off-label use of IVIG Meets Member benefit certificate primary coverage criteria for the following conditions:
 
        • Acquired Factor VIII Deficiency/Acquired Hemophilia for patients unresponsive to, or cannot tolerate, standard therapy
        • Acquired hypogammaglobulinemia associated with hematological malignancy in patients with recurrent bacterial/fungal infection, and/or who have no antibody response to immunization with bacterial antigen
        • Acquired hypogammaglobulinemia following solid organ transplant in patients who have recurrent bacterial/fungal infection
        • Anti-neutrophil cytoplasmic antibody vasculitis (e.g., Wegener’s granulomatosis, Churg-Strauss syndrome) that has not responded to standard therapy)
        • Autoimmune encephalitis (AE) when the following criteria are met (Zuliani, 2019; Lancaster, 2016):
            • No eveidence of neopleasm or paraneoplstic syndrome is associated with AE; AND
            • All of the following apply:
                • Detection of a spcific autoantibody associated with AE, including but not limited to:
                  • NMDAR, LGI1, Caspr2, AMPAR, GABA-A or GABA-B receptor, IgLON5, DPPX, GlyR, mGluR1, mGluR2, mGluR5, Neurexin 3-alpha, or dopamine-2 receptor (D2R); AND
                • Clinical presentation includes neurological symptoms (for example, memory deficits, seizures, movement disorders, speech disturbances, behavioral changes, or psychiatric symptoms); AND
                • Alternative etiologies of encephalitis syndrome have been ruled out, such as infectious etiologies, other neurological disorders, or other autoimmune conditions.
            • Continued use of Ig after initial trial when the following criteria are met:
                • There are clinically significant improvements in symptoms on physical examination; AND
                • Continued need is demonstrated by clinical effect (for example, patient has a positive response and stable on current dose, or worsening of symptoms occurs from a dose decrease or increase in dose intervals, or previous discontinuation resulted in relapse);
                • There continues to be no evidence of cancer or paraneoplastic syndrome
        • Autoimmune encephalopathy (e.g., Hashimoto’s encephalopathy, voltage-gated potassium channel complex antibody-associated encephalopathy, progressive encephalomyelopathy) in patients refractory to, or cannot tolerate, glucocorticoids
        • Autoimmune hemolytic anemia, warm type, that has not responded to standard therapy
        • Autoimmune hemolytic anemia, life threatening
        • Autoimmune hemolytic anemia, neonatal
        • Autoimmune, non-paraneoplastic, encephalopathy (Hashimoto’s encephalopathy, voltage-gated potassium channel antibody encephalopathy), refractory to standard therapy (Effective, Nov 2011)
        • Autoimmune retinopathy
        • Catastrophic antiphospholipid syndrome. (Effective September 2019)
        • Dermatomyositis, refractory to glucocorticoids and at least one other immunosuppressant drug.  There must be laboratory confirmation that the patient does not have inclusion body myositis. Coverage for more than four months will require documentation of response to IVIG
        • Encephalitis, acute disseminated, refractory to corticosteroid therapy
        • End-Stage Renal Disease who have been on a transplant waiting list for 2 years and are otherwise eligible for transplantation but have been unable to find a suitable donor because of high, unacceptable levels of anti-HLA Panel Reactive Antibodies (>=50%).  The dose covered is 2g/kg of body weight per month for 3 months.
        • Fetal/maternal red blood cell alloimmunization
        • Good Syndrome (Durable Hypogammaglobulinemia Associated with Thymoma) (Effective, April 2012)
        • Graves ophthalmopathy, refractory to standard therapy
        • Guillain-Barre, within 2-weeks of symptom onset
        • Guillain-Barre, relapse occurring following initial response to IVIG
        • Hemolytic disease of newborn with hyperbilirubinemia (Neonatal Iso-immune hemolytic jaundice) when there is parental refusal for exchange transfusion or where appropriate blood components for exchange transfusion are unavailable
        • Hemolytic transfusion reaction, acute, severe, refractory to standard therapy
        • Hemolytic transfusion reaction in sickle cell disease, life threatening
        • Hemolytic uremic syndrome in adults, refractory to standard therapy
        • Hemolytic uremic syndrome, children, refractory to standard therapy
        • Hyperimmunoglobulin E syndrome
        • Hyperimmunoglobulin M syndrome, X-linked, or autosomal recessive (Effective, April 2012)
        • IgG subclass deficiency only when there is also a demonstrated deficiency in the ability to form antibodies against a variety of polysaccharide AND protein antigens
        • IgG/IgA Gammopathy of Unknown Significance, with polyneuropathy
        • IRAK4 Immunodeficiency, homozygous or compound heterozygous (Effective, March 2012)
        • Isaac’s Syndrome (Neuromyotonia), refractory to standard treatment
        • Lupus Erythematosus, refractory to standard treatment  
        • Multiple Myeloma, in patients with recurrent infections who are unable to respond to pneumococcal vaccine (as demonstrated by absence of or low titer to pneumococcal vaccine, determined by serological studies)
        • Multiple Myeloma, in patients who are post hematopoietic stem cell transplant, who have hypogammaglobulinemia (IgG < 400 mg/dL), who have had two documented (significant) bacterial infections
        • Multiple Sclerosis, Marburg disease
        • Multiple Sclerosis, acute optic neuritis
        • Multiple Sclerosis, relapsing-remitting type only, when interferon therapy has failed.  Coverage requires written documentation
        • Myasthenia Gravis, Pediatric, for the treatment of acute crisis or worsening of disease unresponsive to conventional therapy. Dosage: IVIg given at 1g/kg body weight for 1 to 2 daily treatments.
        • Myasthenia Gravis, Pediatric, for maintenance Dosage: Following the loading dose, 1 treatment every 2 to 3 weeks, followed by a gradual taper to complete withdrawal as tolerated.
        • Myasthenia Gravis, Neonatal, for the treatment of acute crisis or worsening of disease unresponsive to conventional therapy.
        • Myasthenia Gravis, Adult, for the treatment of acute crisis or worsening of disease unresponsive to conventional therapy. Dosage: IVIG total of 2 g/kg which may be given in divided doses over 2 to 14 days.
        • Myasthenia Gravis, Adult, for maintenance Dosage: Following the loading dose, maintenance doses of 0.4 g/kg every 4-12 weeks, as necessary.  
        • Myasthenia Gravis, Stabilization Prior to Surgery
        • Opsoclonus-Myoclonus Syndrome, refractory to standard therapy
        • Paraneoplastic Retinopathy (Cancer Associated Retinopathy; Melanoma Associated Retinopathy)
        • Paraneoplastic Syndrome (Lambert-Eaton myasthenia), refractory to standard therapy
        • Parvovirus B19 in members with anemia who are immunodeficient or who have sickle cell disease.  
        • Pemphigus & Other Autoimmune Blistering Diseases refractory to standard therapy
        • Polymyositis, refractory to standard therapy
        • Post-transfusion Purpura
        • Pure Red Cell Aplasia (PRCA), refractory to standard therapy
        • Stevens-Johnson Syndrome (NCCN 2A, 2021)
        • Stiff-person syndrome
        • Thrombocytopenia, Alloimmune, Neonatal
        • Thrombocytopenia, HIV associated
        • Thrombocytopenic Purpura, Idiopathic, Acute, Adult, with acute bleeding
        • Thrombocytopenic Purpura, Idiopathic, Acute, Adult, Severe (platelets less than 30,000), without bleeding, refractory to standard therapy
        • Thrombocytopenic Purpura, Idiopathic, Children, platelets less than 30,000, or with acute bleeding, refractory to standard therapy
        • Thrombocytopenic Purpura, Idiopathic, to allow patient to undergo surgery
        • Thrombotic Thrombocytopenic Purpura, refractory to standard therapy
        • Toxic Epidermal Necrolysis
        • Transient Hypogammaglobulinemia of Childhood (Effective, April 2012)
        • Transplant, Kidney, End-Stage Renal Disease, on transplant waiting list for 2 years and are otherwise eligible for transplantation but have been unable to find a suitable donor because of high, unacceptable levels of anti-HLA Panel Reactive Antibodies (>=50%)
        • Transplant, Kidney, undergoing rejection due to pathologically demonstrated vascular rejection (February 2012)
        • Transplant, following solid-organ transplant, treatment of antibody-mediated rejection. A total dose of 2g/kg is covered. (Revised December 2013)
        • Transplant, prior to solid organ transplant, treatment of patients at high risk of antibody-mediated rejection, including highly sensitized patients and those receiving an ABO incompatible organ.  A total dose of 2 g/kg is covered. (Effective December, 2013)
        • Von Willebrand Disease, Acquired, refractory to standard therapy; not covered for routine use
        • West Nile Virus Encephalomyelitis, as treatment and not prophylaxis (Effective, March 2012)
        • West Syndrome, refractory to standard therapy
 
Please refer to a separate policy on Site of Care or Site of Service Review (policy #2018030) for pharmacologic/biologic medications.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
IVIG, human-sira (Asceniv™) does not meet member benefit certificate primary coverage criteria because there is a lack of data indicating this intervention is more effective than other interventions that are less costly.  (Effective 04/01/2021)
 
For members without contracts without primary coverage criteria, IVIG, human-sira (Asceniv™) is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage. (Effective 04/01/2021)
 
IVIG does not meet member benefit certificate primary coverage criteria for effectiveness, or is considered investigational for those contracts without primary coverage criteria including but not limited to the following off label uses:
 
        • Adrenoleukodystrophy (lack of evidence of effectiveness – absence of clinical trials; conflicting case reports; expert opinion recommends against)
        • Alzheimer’s Disease (lack of evidence of effectiveness – absence of clinical trial outcomes; IVIG for Alzheimer’s disease is presently being studied in clinical trials to determine safety and effectiveness; expert opinion recommends against)
        • Amyotrophic Lateral Sclerosis (lack of evidence of effectiveness – absence of clinical trials; expert opinion recommends against)
        • Aplastic Anemia (lack of evidence of effectiveness – absence of clinical trials; conflicting case reports; expert opinion recommends against)
        • Asthma (lack of evidence of effectiveness – randomized controlled trials described no benefit; expert opinion recommends against)
        • Autism (lack of evidence of effectiveness – absence of clinical trials; case reports show conflicting results; expert opinion recommends against)
        • Autism, Oral Human Immunoglobulin for Associated Gastrointestinal Symptoms (lack of evidence of effectiveness – randomized controlled trial described no benefit)
        • Autoimmune Urticaria/Angioedema (lack of effectiveness – absence of clinical trials)
        • Birdshot Retinopathy (Effective July 2013)
        • Cardiomyopathy, Idiopathic (lack of evidence of effectiveness – randomized controlled trial described no benefit)
        • Chronic Fatigue Syndrome (lack of evidence of effectiveness – randomized controlled trial described no benefit; expert opinion recommends against)
        • Clostridium difficile enterocolitis (lack of evidence of effectiveness – no trials establishing benefit; mixed results from uncontrolled studies)
        • Congenital Heart Block, In Utero, to prevent progression of 1st or 2nd degree heart block to complete heart block (lack of evidence of effectiveness – no trials establishing benefit, and conflicting evidence from rare case reports)
        • Congenital Heart Block, In Utero, to prevent heart block in the fetus of a mother with fetal heart block in an earlier pregnancy (lack of evidence of effectiveness – randomized controlled trials have shown no benefit)
        • Diabetes Mellitus (lack of evidence of effectiveness – randomized controlled trial described no benefit; not recommended by any appropriate medical society)
        • Diabetic Amyotrophy (lack of evidence of effectiveness – lack of clinical trials; expert opinion recommends against)
        • DiGeorge Syndrome (lack of evidence of effectiveness – expert opinion recommends against)
        • Epidermolysis Bullosa Acquisita (Effective July 2013)
        • Fisher Syndrome (lack of evidence of effectiveness – lack of clinical trials; expert opinion recommends against)
        • Hemophagocytic syndrome (i.e., hemophagocytic lymphohistiocytosis)
        • Inclusion Body Myositis (lack of evidence of effectiveness – randomized controlled trials described no benefit; expert opinion recommends against)
        • In-Vitro Fertilization Failure
        • MYD88 gene immunodeficiency, homozygous or compound heterozygous mutations (lack of evidence of effectiveness – expert opinion does not recommend)
        • Neonatal sepsis (lack of evidence of effectiveness – randomized trials showed no benefit; Cochrane Review concluded no benefit)
        • Necrotizing Fasciitis (Effective July 2013)
        • Pediatric autoimmune neuropsychiatric disorder associated with group A streptococci (PANDAS); Pediatric acute-onset neuropsychiatric syndrome (PANS); childhood acute-onset neuropsychiatric syndrome (CANS) [Sigra, 2018; Williams, Swedo, 2016; UTD, Pichichero, 2021]
        • Polyradiculoeruopathy (other than CIDP) Effective July 2013)
        • Post-Cardiac Transplant Hypogammaglobulinemia (lack of evidence of effectiveness – presently under study in clinical trial)
        • Post-Polio Syndrome
        • Prevention of recurrent pregnancy loss (Alloimmune, Antiphospholipid Antibody Syndrome or any other indication)
        • Rheumatic Fever, Acute (lack of evidence of effectiveness – randomized trial described no benefit; expert opinion recommends against  
        • Rippling muscle disease, no medical literature reporting use of IVIG identified. (EFFECTIVE June, 2013)
 
There is no coverage for IVIG for indications other than those specifically listed as covered based on primary coverage criteria for effectiveness.
 
Effective February 2020 to December 31, 2021
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
SUBCUTANEOUS IMMUNOGLOBULIN
 
SCIg may meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness for the treatment of primary immunodeficiencies, including:
 
    • congenital agammaglobulinemia,
    • hypogammaglobulinemia,
    • common variable immunodeficiency (CVID),
    • severe combined immunodeficiency,
    • Wiskott-Aldrich syndrome,
    • X-linked agammaglobulinemia (XLA); OR
    • when coverage criteria have been met for intravenous immune globulin (IVIG) but patient is unable to tolerate IVIG because of side effects from the IVIG treatment.
 
INTRAVENOUS IMMUNOGLOBULIN (FDA APPROVED INDICATIONS)
 
The administration of intravenous immune globulin meets member benefit certificate primary coverage criteria for effectiveness for the following FDA approved indications:
 
    • Congenital Agammaglobulinemia (X-linked agammaglobulinemia)
    • Common Variable Immunodeficiency (panhypogammaglobulinemia)
    • X-linked immunodeficiency hyperimmunoglobulin IgM
    • X-linked hyper-IgM syndrome and ataxia telangiectasia  (Effective January 2017)
    • Severe Combined Immunodeficiency
    • Wiskott-Aldrich Syndrome
    • Bone marrow transplantation: to prevent risk of acute graft-versus-host disease, associated interstitial pneumonia (infectious or idiopathic), and infections (e.g., CMV, varicella-zoster viral infection, and recurrent bacterial infection) after allogeneic bone marrow transplantation in patients 20 years of age or older in the first 100 days after transplantation;
    • Chronic inflammatory demyelinating polyneuropathy (FDA approved indication) when all of the following criteria are documented:
        • Progressive or relapsing motor AND sensory dysfunction of more than one limb; AND
        • Significant disability in arm or leg function developing over at least two months; AND
        • Hypo- or areflexia; AND
        • One of the following:
            • partial conduction block or abnormal temporal dispersion of conduction must be present in at least 2 nerves and there must be significantly reduced conduction velocity, or significantly prolonged distal motor latency, or absent or significantly prolonged minimum Fwave latency in at least one other nerve; OR
            • in the absence of block or dispersion, significantly reduced conduction velocity, or significantly prolonged distal motor latency, or absent or significantly prolonged minimum F-wave latency must be present in at least 3 nerves; OR
            • in the presence of significant neurophysiologic abnormalities in only 2 nerves, unequivocal histological evidence of demyelinated nerve fibers in a nerve biopsy must also be present. CSF Studies are encouraged. An elevated protein with a CSF white count of <10/mm3 in HIV-1 negative patients would be expected in patients with CIDP.
    • Chronic Lymphocytic Leukemia with associated hypogammaglobulinemia in patients (FDA approved indication) who have demonstrated frequent and recurrent bacterial infections or who demonstrate impaired response to bacterial polysaccharides or proteins (study results);
    • Human immunodeficiency virus infection (HIV) in children to decrease the risk of serious bacterial infection (FDA approved indication). IVIG is covered only in children who have IgG less than 400 mg/dL or functional antibody deficiency is demonstrated by poor specific antibody titers or recurrent bacterial infections (ACIP, AAP, CDC, NIH, IDSA, PIDS recommendation).
    • Idiopathic thrombocytopenic purpura, acute, or chronic, when a rapid rise in the platelet count is required, such as prior to surgery or to prevent/control excessive bleeding (FDA approved indication) when the platelet count is less than 30,000 (study results).
    • Kawasaki’s Disease (Mucocutaneous lymph node syndrome) when used in conjunction with ASA and treatment is started within the first 10 days of illness (FDA approved indication) or as soon as possible after the disease is diagnosed or strongly suspected (AAP, AHA, ACCP recommendation);
    • Multifocal Motor Neuropathy (MMN) when used as initial and maintenance therapy to improve muscle strength and disability in adult patients. (EFFECTIVE June 2013)
    • Toxic Shock Syndrome  
 
OFF LABEL INDICATIONS FOR INTRAVENOUS IMMUNOGLOBULIN
 
These off label indications are considered eligible for coverage based on one or more of the following:
 
    • High quality systematic reviews of randomized controlled trials by independent review organizations (e.g., Cochrane Systematic Reviews, AHRQ Evidence Based Practice Centers);
    • adequately powered randomized controlled trials published in peer-reviewed medical literature;
    • single or multiple randomized controlled trials, adequately populated, with risk of bias, but recommended in publications in peer-reviewed journals;
    • prospective, non-randomized, adequately populated case-control or cohort study(s) with low risk of confounding or bias;
    • expert opinion, presented as published position papers by national medical societies supporting this opinion (unless in conflict with other expert opinion); and
    • rare life threatening conditions for which:  
        • only case series have been reported in peer reviewed medical literature;
        • more than one series has reported significant, lasting improvement;
        • no conflicting evidence is known; and
        • because of the rarity of the condition, adequate appropriate trials would be unlikely.  
 
There is no coverage for IVIG for indications other than those specifically listed as covered; requests for coverage of conditions not listed in the covered section should be submitted for review, and include references from peer reviewed journals which support coverage for the proposed condition.
 
Off-label use of IVIG Meets Member benefit certificate primary coverage criteria for the following conditions:
 
    • Acquired Factor VIII Deficiency/Acquired Hemophilia for patients unresponsive to, or cannot tolerate, standard therapy
    • Acquired hypogammaglobulinemia associated with hematological malignancy in patients with recurrent bacterial/fungal infection, and/or who have no antibody response to immunization with bacterial antigen
    • Acquired hypogammaglobulinemia following solid organ transplant in patients who have recurrent bacterial/fungal infection
    • Anti-neutrophil cytoplasmic antibody vasculitis (e.g., Wegener’s granulomatosis, Churg-Strauss syndrome) that has not responded to standard therapy)
    • Autoimmune encephalopathy (e.g., Hashimoto’s encephalopathy, voltage-gated potassium channel complex antibody-associated encephalopathy, progressive encephalomyelopathy) in patients refractory to, or cannot tolerate, glucocorticoids
    • Autoimmune hemolytic anemia, warm type, that has not responded to standard therapy
    • Autoimmune hemolytic anemia, life threatening
    • Autoimmune hemolytic anemia, neonatal
    • Autoimmune, non-paraneoplastic, encephalopathy (Hashimoto’s encephalopathy, voltage-gated potassium channel antibody encephalopathy),  refractory to standard therapy (Effective, Nov 2011)
    • Autoimmune retinopathy
    • Catastrophic antiphospholipid syndrome. (Effective September 2019)
    • Dermatomyositis, refractory to glucocorticoids and at least one other immunosuppressant drug.  There must be laboratory confirmation that the patient does not have inclusion body myositis. Coverage for more than four months will require documentation of response to IVIG
    • Encephalitis, acute disseminated, refractory to corticosteroid therapy
    • End-Stage Renal Disease who have been on a transplant waiting list for 2 years and are otherwise eligible for transplantation but have been unable to find a suitable donor because of high, unacceptable levels of anti-HLA Panel Reactive Antibodies (>=50%).  The dose covered is 2g/kg of body weight per month for 3 months.
    • Fetal/maternal red blood cell alloimmunization
    • Good Syndrome (Durable Hypogammaglobulinemia Associated with Thymoma) (Effective, April 2012)
    • Graves ophthalmopathy, refractory to standard therapy
    • Guillain-Barre, within 2-weeks of symptom onset
    • Guillain-Barre, relapse occurring following initial response to IVIG
    • Hemolytic disease of newborn with hyperbilirubinemia (Neonatal Iso-immune hemolytic jaundice) when there is parental refusal for exchange transfusion or where appropriate blood components for exchange transfusion are unavailable
    • Hemolytic transfusion reaction, acute, severe, refractory to standard therapy
    • Hemolytic transfusion reaction in sickle cell disease, life threatening
    • Hemolytic uremic syndrome in adults, refractory to standard therapy
    • Hemolytic uremic syndrome, children, refractory to standard therapy
    • Hyperimmunoglobulin E syndrome
    • Hyperimmunoglobulin M syndrome, X-linked, or autosomal recessive (Effective, April 2012)
    • IgG subclass deficiency only when there is also a demonstrated deficiency in the ability to form antibodies against a variety of polysaccharide AND protein antigens
    • IgG/IgA Gammopathy of Unknown Significance, with polyneuropathy
    • IRAK4 Immunodeficiency, homozygous or compound heterozygous (Effective, March 2012)
    • Isaac’s Syndrome (Neuromyotonia), refractory to standard treatment
    • Lupus Erythematosus, refractory to standard treatment  
    • Multiple Myeloma, in patients with recurrent infections who are unable to respond to pneumococcal vaccine (as demonstrated by absence of or low titer to pneumococcal vaccine, determined by serological studies)
    • Multiple Myeloma, in patients who are post hematopoietic stem cell transplant, who have hypogammaglobulinemia (IgG < 400 mg/dL), who have had two documented (significant) bacterial infections
    • Multiple Sclerosis, Marburg disease
    • Multiple Sclerosis, acute optic neuritis
    • Multiple Sclerosis, relapsing-remitting type only, when interferon therapy has failed.  Coverage requires written documentation
    • Myasthenia Gravis, Pediatric, for the treatment of acute crisis or worsening of disease unresponsive to conventional therapy. Dosage: IVIg given at 1g/kg body weight for 1 to 2 daily treatments.
    • Myasthenia Gravis, Pediatric, for maintenance Dosage: Following the loading dose, 1 treatment every 2 to 3 weeks, followed by a gradual taper to complete withdrawal as tolerated.
    • Myasthenia Gravis, Neonatal, for the treatment of acute crisis or worsening of disease unresponsive to conventional therapy.
    • Myasthenia Gravis, Adult, for the treatment of acute crisis or worsening of disease unresponsive to conventional therapy. Dosage: IVIG total of 2 g/kg which may be given in divided doses over 2 to 14 days.
    • Myasthenia Gravis, Adult, for maintenance Dosage: Following the loading dose, maintenance doses of 0.4 g/kg every 4-12 weeks, as necessary.  
    • Myasthenia Gravis, Stabilization Prior to Surgery
    • Opsoclonus-Myoclonus Syndrome, refractory to standard therapy
    • Paraneoplastic Retinopathy (Cancer Associated Retinopathy; Melanoma Associated Retinopathy)
    • Paraneoplastic Syndrome (Lambert-Eaton myasthenia), refractory to standard therapy
    • Parvovirus B19 in members with anemia who are immunodeficient or who have sickle cell disease.  
    • Pemphigus & Other Autoimmune Blistering Diseases refractory to standard therapy
    • Polymyositis, refractory to standard therapy
    • Post-transfusion Purpura
    • Pure Red Cell Aplasia (PRCA), refractory to standard therapy
    • Stevens-Johnson Syndrome
    • Stiff-person syndrome
    • Thrombocytopenia, Alloimmune, Neonatal
    • Thrombocytopenia, HIV associated
    • Thrombocytopenic Purpura, Idiopathic, Acute, Adult, with acute bleeding
    • Thrombocytopenic Purpura, Idiopathic, Acute, Adult, Severe (platelets less than 30,000), without bleeding, refractory to standard therapy
    • Thrombocytopenic Purpura, Idiopathic, Children, platelets less than 30,000, or with acute bleeding, refractory to standard therapy
    • Thrombocytopenic Purpura, Idiopathic, to allow patient to undergo surgery
    • Thrombotic Thrombocytopenic Purpura, refractory to standard therapy
    • Toxic Epidermal Necrolysis
    • Transient Hypogammaglobulinemia of Childhood (Effective, April 2012)
    • Transplant, Kidney, End-Stage Renal Disease, on transplant waiting list for 2 years and are otherwise eligible for transplantation but have been unable to find a suitable donor because of high, unacceptable levels of anti-HLA Panel Reactive Antibodies (>=50%)
    • Transplant, Kidney, undergoing rejection due to pathologically demonstrated vascular rejection (February 2012)
    • Transplant, following solid-organ transplant, treatment of antibody-mediated rejection. A total dose of 2g/kg is covered. (Revised December 2013)
    • Transplant, prior to solid organ transplant, treatment of patients at high risk of antibody-mediated rejection, including highly sensitized patients and those receiving an ABO incompatible organ.  A total dose of 2 g/kg is covered. (Effective December, 2013)
    • Von Willebrand Disease, Acquired, refractory to standard therapy;  not covered for routine use
    • West Nile Virus Encephalomyelitis, as treatment and not prophylaxis (Effective, March 2012)
    • West Syndrome, refractory to standard therapy
 
Please refer to a separate policy on Site of Care or Site of Service Review (policy #2018030) for pharmacologic/biologic medications.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
IVIG, human-sira (Asceniv™) does not meet member benefit certificate primary coverage criteria because there is a lack of data indicating this intervention is more effective than other interventions that are less costly.  (Effective 04/01/2021)
 
For members without contracts without primary coverage criteria, IVIG, human-sira (Asceniv™) is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage. (Effective 04/01/2021)
 
IVIG does not meet member benefit certificate primary coverage criteria for effectiveness, or is considered investigational for those contracts without primary coverage criteria including but not limited to the following off label uses:
 
    • Adrenoleukodystrophy (lack of evidence of effectiveness – absence of clinical trials; conflicting case reports; expert opinion recommends against)
    • Alzheimer’s Disease (lack of evidence of effectiveness – absence of clinical trial outcomes; IVIG for Alzheimer’s disease is presently being studied in clinical trials to determine safety and effectiveness; expert opinion recommends against)
    • Amyotrophic Lateral Sclerosis (lack of evidence of effectiveness – absence of clinical trials; expert opinion recommends against)
    • Aplastic Anemia (lack of evidence of effectiveness – absence of clinical trials; conflicting case reports; expert opinion recommends against)
    • Asthma (lack of evidence of effectiveness – randomized controlled trials described no benefit; expert opinion recommends against)
    • Autism (lack of evidence of effectiveness – absence of clinical trials; case reports show conflicting results; expert opinion recommends against)
    • Autism, Oral Human Immunoglobulin for Associated Gastrointestinal Symptoms (lack of evidence of effectiveness – randomized controlled trial described no benefit)
    • Autoimmune Urticaria/Angioedema (lack of effectiveness – absence of clinical trials)
    • Birdshot Retinopathy (Effective July 2013)
    • Cardiomyopathy, Idiopathic  (lack of evidence of effectiveness – randomized controlled trial described no benefit)
    • Chronic Fatigue Syndrome (lack of evidence of effectiveness – randomized controlled trial described no benefit; expert opinion recommends against)
    • Clostridium difficile enterocolitis (lack of evidence of effectiveness – no trials establishing benefit; mixed results from uncontrolled studies)
    • Congenital Heart Block, In Utero, to prevent progression of 1st or 2nd degree heart block to complete heart block (lack of evidence of effectiveness – no trials establishing benefit, and conflicting evidence from rare case reports)
    • Congenital Heart Block, In Utero, to prevent heart block in the fetus of a mother with fetal heart block in an earlier pregnancy (lack of evidence of effectiveness – randomized controlled trials have shown no benefit)
    • Diabetes Mellitus (lack of evidence of effectiveness – randomized controlled trial described no benefit; not recommended by any appropriate medical society)
    • Diabetic Amyotrophy (lack of evidence of effectiveness – lack of clinical trials; expert opinion recommends against)
    • DiGeorge Syndrome (lack of evidence of effectiveness – expert opinion recommends against)
    • Epidermolysis Bullosa Acquisita (Effective July 2013)
    • Fisher Syndrome (lack of evidence of effectiveness – lack of clinical trials; expert opinion recommends against)
    • Hemophagocytic syndrome (i.e., hemophagocytic lymphohistiocytosis)
    • Inclusion Body Myositis (lack of evidence of effectiveness – randomized controlled trials described no benefit; expert opinion recommends against)
    • In-Vitro Fertilization Failure
    • MYD88 gene immunodeficiency, homozygous or compound heterozygous mutations (lack of evidence of effectiveness – expert opinion does not recommend)
    • Neonatal sepsis (lack of evidence of effectiveness – randomized trials showed no benefit; Cochrane Review concluded no benefit)
    • Necrotizing Fasciitis (Effective July 2013)
    • Polyradiculoeruopathy (other than CIDP) Effective July 2013)
    • Post-Cardiac Transplant Hypogammaglobulinemia (lack of evidence of effectiveness – presently under study in clinical trial)
    • Post-Polio Syndrome
    • Prevention of recurrent pregnancy loss (Alloimmune, Antiphospholipid Antibody Syndrome or any other indication)
    • Rheumatic Fever, Acute (lack of evidence of effectiveness – randomized trial described no benefit; expert opinion recommends against  
    • Rippling muscle disease, no medical literature reporting use of IVIG identified. (EFFECTIVE June, 2013)
 
There is no coverage for IVIG for indications other than those specifically listed as covered based on primary coverage criteria for effectiveness.
 

Rationale:
Due to the detail of the rationale, the complete document is not online. If you would like a hardcopy print, please email: codespecificinquiry@arkbluecross.com
 
Given the heterogeneous nature and relapsing-remitting course of many of the diseases for which IVIg has been investigated as therapy, randomized, controlled trials (RCTs) are important for evaluating true benefit. However, in the case of rare disease, RCTs may be neither likely nor feasible. In these cases, reports of series data from at least 10 patients and consistent trends in results may support conclusions. Aside from the labeled indications, the use of IVIg has been investigated in a variety of diseases as follows:
 
ACQUIRED FACTOR VIII DEFICIENCY/ACQUIRED HEMOPHILIA
Acquired factor VIII deficiency/Acquired hemophilia is a coagulopathy caused by the development of autoantibodies directed against specific domains of the coagulation Factor VIII molecule.  Factor VIII becomes bound in the coagulation cascade which results in bleeding.  Mortality rates as high as 21% have been reported.  There are no randomized trials, and standard therapy, based on a randomized trial has been corticosteroids and cyclophosphamide (Green, 1993).  Cyclosporine has been substituted because of side of effects of long-term corticosteroid + cyclophosphamide.  Treatment with IVIg has been reported in case series (Bossi, 1998), but a meta-analysis of 245 patients with acquired Factor VIII hemophilia found that IVIG had a poor complete response rate (Delgado, 2003).  A protocol consisting of antibody depletion through immunoadsorption, IVIG, immunosuppression, and high dose factor VIII supplementation, reported in 2005, described 35 patients with an 88% long-term response rate (93% if 5 patients with malignancy were excluded) (Modified Bonn-Malmö Protocol) (Zeitler, 2005).   Neither the AHFS nor the Clinical Pharmacology Online drug compendia recommend IVIG for treatment of acquired hemophilia.  The Canadian Expert Panel on IVIG for hematological uses and the Academy of Allergy and Immunology Expert Panel on IVIG recommend IVIG for treatment of acquired hemophilia if standard therapy has failed.
 
ADRENOLEUKODYSTROPHY
X-linked adrenoleukodystrophy is an inborn error of peroxisomal metabolism characterized by progressive demyelination of the CNS and by hypoadrenalism.  There is very little information on the use of IVIG for adrenoleukodystrophy, and those results are conflicting (Miike, 1989; Cappa, 1994).  The Canadian IVIG Hematology & Neurology Expert Panel (Robinson, 2007) and the American Academy of Allergy, Asthma, and Immunology (Orange, 2006) recommend against the use of IVIG in adrenoleukodystrophy.
 
ALZHEIMER’S DISEASE
Antibodies against beta-amyloid have been found in human immunoglobulin preparations, leading to potential use of IVIG as treatment of Alzheimer’s disease.  A claims based retrospective review of IVIg for Alzheimer’s disease was published in 2009 (Fillit H) showed that previous treatment with IVIG was associated with a reduced risk of development of Alzheimer’s and related disorders in patients greater than or equal to 65 years of age.  No review articles recommend IVIG treatment of Alzheimer’s disease.  Use of IVIG for Alzheimer’s disease is being studied in clinical trials to determine safety and effectiveness (NCT00818662, NCT00299988, NCT00812565).   
 
AMYOTROPHIC LATERAL SCLEROSIS
Small prospective case series showed no benefit of IVIG for treatment of ALS (Dalakas, 1994; Meucci, 1996).   The Canadian IVIG Hematology & Neurology Expert Panel (Robinson, 2007) and the American Academy of Allergy, Asthma, and Immunology (Orange, 2006) recommend against the use of IVIG in adrenoleukodystrophy.
 
APLASTIC ANEMIA
There have been no clinical trials on IVIG for aplastic anemia.  Rare case reports (Sadowitz, 1990; Chen, 1995) and letters to the editor (Bodenstein, 1991) have described benefit, but other studies have shown no benefit (Miceli, 1990; Sumimoto, 1992; Bejaoui, 1993).  The Canadian IVIG Hematology & Neurology Expert Panel (Robinson, 2007) recommends against IVIG for aplastic anemia.  Neither the AHFS nor Clinical Pharmacology Online drug compendia recommend use of IVIG in aplastic anemia.
 
ASTHMA
There have been 3 double-blind, placebo-controlled studies of IVIG in asthma (Niggemann, 1998;).  Two of the 3 showed no significant improvement (Kishiyama, 1999; Salum, 1999), and the third reported significant steroid-sparing effect in a subgroup that required relatively high daily doses of oral steroids.  The American Academy of Allergy and Immunology expert panel recommends against the use of IVIG for asthma (Orange, 2006).  Neither the AHFS nor the Clinical Pharmacology Online compendia recommend IVIG for asthma.  
 
AUTISM
There are no clinical trials reported on the use of IVIG to treat the neurological effects of autism, nor are there any ongoing trials of IVIG for autism listed on clinicaltrial.gov. (Oct 2010).  A randomized controlled trial of IVIG given orally for treatment of gastrointestinal symptoms of associated with autism has been reported.  This trial (NCT00110708) found no benefit (Handen, 2009).  An uncontrolled, unblinded study of 10 autistic children who received IVIG showed benefit in one child (Plioplys, 1998).  A brief report of a pilot trial described no benefit (DelGiudice-Asch, 1999).  The  American Academy of Allergy, Asthma & Immunology expert panel (Orange, 2006), and the Canadian IVIG Hematology & Neurology Expert Panel (Robinson, 2007) recommend against IVIG for autism.
 
AUTOIMMUNE BLISTERING DISEASES, INCLUDING PEMPHIGUS
Corticosteroids and immunosuppressive agents are considered first line therapy. IVIG therapy is the preferred therapy for patients who are resistant, nonresponsive, have contraindications, or develop serious catastrophic adverse effects (Ahmed, 2003).  No clinical trials have been reported for IVIG for pemphigus or other autoimmune blistering diseases.  Two randomized controlled trials are listed on clinicaltrial.gov.  NCT00483119 is an ongoing FDA sponsored trial at NYU Medical Center, initiated in 2007 with estimated completion date of 2011, comparing IVIG versus IVIG plus cyclophosphamide in patients with pemphigus that are not responding to, or have developed complications from, standard therapy.  A second study, in Japan (NCT00809822) recently completed but unreported, is a randomized, placebo controlled trial of IVIG for patients with bullous pemphigoid unresponsive to corticosteroids.   Prospective, non-randomized. open label clinical studies on patients who are unresponsive to corticosteroids and immunosuppressive agents have shown response to IVIG (Toth, 1999; Engineer, 2000; Ahmed, 2001; Sami, 2002; Bystryn, 2002; Baum, 2002; Ahmed, 2002).  IVIG has not been compared to newer biologics (e.g., anti-tumor necrosis alfa drugs).
 
AUTOIMMUNE ENCEPHALOPATHY
This condition was originally designated in 1966 as “Hashimoto’s encephalopathy”, but more recently has been termed “autoimmune encephalopathy” (Caselli, 2010; Flanagan, 2010).  There are few reports of this disease, and treatment has been with glucocorticoids.  A few case reports of IVIG for steroid refractory disease have indicated both acute and longer-term benefit from IVIG (Flanagan, 2010; Berger, 2010; Drulovic, 2011).  In addition to Hashimoto’s encephalopathy, voltage-gated potassium channel complex antibody-associated encephalopathy (Tan, 2008), and Morvan syndrome (progressive encephaloencephalopathy with rigidity and myoclonus) have been included as autoimmune encephalopathies. IVIG for Hashimoto’s encephalopathy was not been reviewed or commented on by the American Academy of Allergy & Immunology expert panel (2006); the European Federation of Neurological Sciences expert panel (2006); the Canadian expert panel on IVIG for neurological diseases (2007); the Gold, Stangel, and Delakas review of IVIG for neurological diseases (2007); the American Academy of Neurology & Electrodiagnostic Medicine expert panel (2009); or the AHFS or Clinical Pharmacology Online drug compendia (2011).  Because of the rarity of this disorder, it is unlikely that prospective trials will be performed.  A case series of 72 patients (Flanagan, 2010), reported from the Mayo Clinic showed improvement with glucocorticoids, but relapsed after discontinuation.  Some patients responded only to IVIG or received IVIG after failing corticosteroids.  Because of the rarity of this disorder, it is unlikely that prospective trials will be performed.
 
AUTOIMMUNE URTICARIA (ANGIOEDEMA)
There are rare case reports and two case series (Pereira, 2007; Mitzel-Kaoukhov, 2010) which describe IVIG treatment for autoimmune urticaria.  Neither the AHFS nor the Clinical Pharmacology Online drug compendia recommend IVIG for treatment of autoimmune urticaria.  No major medical group recommends IVIG for autoimmune angioedema.  The American Academy of Allergy & Immunology expert panel states, “Because there is not clear evidence that the use of IVIG benefits patients with chronic urticaria, additional studies are needed” (Orange, 2006).  
 
CARDIOMYOPATHY, DILATED CARDIOMYOPATHY
The Intervention in Myocarditis and Acute Cardiomyopathy trial, a multi-center, placebo-controlled trial to determine whether IVIG improves left ventricular ejection fraction in adults with recent onset of idiopathic dilated cardiomyopathy or myocarditis was reported in 2001 (McNamara, 2001).  This trial was done because of reports from uncontrolled studies which purported to show benefit.  The study failed to demonstrate evidence of therapeutic efficacy.   
 
CHRONIC FATIGUE SYNDROME
A randomized, double-blind, placebo-controlled trial of 3 different doses of IVIG in 99 adult patients with chronic fatigue syndrome was reported in 1999 (Volmer-Conna, 1997).  Outcome was assessed by changes in a series of self-reported measures (quality of life, standardized diaries of daily activities, profile of mood states, and Karnofsky performance scale).  No dose of IVIG was associated with a specific therapeutic benefit.  The American Academy of Allergy and Immunology recommends against the use of IVIG for chronic fatigue syndrome (Orange, 2008).
 
CHRONIC INFLAMMATORY DEMYELINATING POLYNEUROPATHY
A double-blind placebo-controlled study comparing IVIg to placebo in patients with progressive or relapsing CIDP reported a significant effect of IVIg in 63% of patients compared to no effect in the placebo group.  A randomized single-blinded study comparing IVIg to plasma exchange reported equivalent beneficial outcomes for both therapies. Open-label treatment of 26 patients with type 2 diabetes and meeting electrophysiologic criteria for CIDP showed significant improvement in 21, suggesting benefit in diabetic CIDP as well. A possible advantage to IVIg treatment is the ability to administer the drug in the home.
 
A multicenter randomized, placebo-controlled trial with a 10% caprylate-chromatography purified intravenous immune globulin product given every 3 weeks for up to 24 weeks showed improvement in inflammatory neuropathy cause and treatment criteria (INCAT) in 54% of treated patients compared to 21% of placebo that was maintained through 24 week 24.   During an extension period of the trial, participants who continued to receive IVIG-C had a longer time to relapse than did patients treated with placebo (Hughes, 2008).
 
CLOSTRIDIUM DIFFICILE ENTEROCOLITIS
IVIG has been reported in single case reports and small case series to be beneficial for treatment of patients with enterocolitis unresponsive to vancomycin and metronidazole (Leung, 1991; Salcedo, 1997; McPherson, 2006; Murphy, 2006), and has been ineffective in other studies (Leung, 2007). The American Academy of Allergy & Immunology expert panel concluded that IVIG “might be beneficial” (Orange 2006).  The Medical Letter recommends IVIG for treatment of frequent relapse of C. difficile enterocolitis, based on the McPherson paper (Med Letter Drugs Ther, 2006a).  A 2008 review concluded, “Despite multiple case reports of successful treatment with IVIG, good sound clinical trials are lacking and thus make it difficult to recommend this for treatment of CDI at this time (Hedge, 2008).
 
Dermatomyositis
A randomized controlled trial comparing IVIg to placebo in 15 patients with refractory dermatomyositis reported significant increases in muscle strength in the IVIg group.  Repeated transfusions every 6 to 8 weeks may be required to maintain a benefit. (Delakas, 1993)  In 2 case series of 18 and 19 patients, a significant number of patients had reduction in corticosteroid use or were otherwise considered responders. In a non-randomized comparison of prednisone plus cyclosporine A, with or without IVIg, patients (12 with DM, 8 with PM) given IVIg had a higher probability of remission. Thus, the available data suggest benefit in DM, but are insufficient for firm conclusions. A recent patient series of IVIg in patients with refractory PM showed significant clinical improvement in more than two-thirds of patients.  IVIG for treatment of refractory dermatomyositis is recommended by the AHFS and Clinical Pharmacology Online drug compendia, and by multiple expert panels.
 
DIABETES MELLITUS, AUTOIMMUNE
A major cause of type I diabetes mellitus is considered to be autoimmune disease which destroys the insulin producing islet cells of the pancreas.  IVIG was studied in a randomized, placebo-controlled trial and found to have no benefit (Colagiuri, 1996).  The American Diabetes Association does not recommend IVIG to treat type I or type II diabetes mellitus.  Neither the AHFS nor the Clinical Pharmacology Online drug compendia recommend IVIG to treat autoimmune diabetes mellitus.
 
DIABETIC AMYOTROPHY
There are a few case reports of IVIG for amyotrophy, but no trials have been reported.  Monotherapy with IVIG was given to a total of 17 patients from 3 studies; all were considered to have improved. (Krendel, 1995; Pascoe, 1997; Jaradeh, 1999).  A randomized controlled trial (NCT00004407) was scheduled by the Mayo Clinic, but has not recruited participants.  The Canadian IVIG Hematology & Neurology Expert Panels recommends against the use of IVIG in diabetic polyneuropathy, mononeuropathy, or proximal lower limb neuropathy (Robinson, 2007).  A 2009 Cochrane review of immunotherapy for diabetic amyotrophy (Chan, 2009) concluded, “there is presently no evidence from randomized trials to support any recommendation on the use of any immunotherapy treatment in diabetic amyotrophy.”
 
DI GEORGE SYNDROME
Di George syndrome is primarily a congenital T-cell deficiency associated with defective development of organs dependent on cells of embryonic neural crest origin, and includes congenital cardiac defects, failure of parathyroid development, and absence of the thymus.  Facial abnormalities also may occur.  Serum immunoglobulin concentrations are frequently normal, but impaired antibody responses to polysaccharide and other antigens have been found in approximately one third of patients with Di George syndrome, but they seldom warrant IgG administration (Kobrynski, 2006).  
 
ENCEPHALITIS, ACUTE, DISSEMINATED
Acute disseminated encephalomyelitis (ADEM) is an immune-mediated inflammatory disorder of the CNS, commonly preceded by an infection, and predominately affects the white matter of the brain and spinal cord. It occurs predominately in pediatric patients, but can occur in adults.  It is classically described as monophasic, but can be recurrent and multiphasic, with wide variation of severity.  There is no standard therapy; most treatment approaches have employed some form of nonspecific immunosuppressant therapy including steroids, IVIG (Kleiman, 1995, Nishikawa, 1999; Sahlas, 2000; Marchioni, 2002; Ravaglia, 2006), or plasmapheresis.  No randomized controlled trials have been performed, and there have been no studies which have compared IVIG with steroids, plasmapheresis, or other immunomodulatory treatments.  The American Academy of Allergy & Immunology (Orange, 2006), the International Pediatric MS Study Group (Tenembaum, 2007), and the Canadian IVIG Hematology & Neurology Expert Panels (Robinson, 2007) recommend IVIG if corticosteroids are ineffective.
 
FISHER SYNDROME
Fisher syndrome is a rare, acute neurological disorder characterized by a triad of clinical manifestations that includes brain-damage associated abnormal coordination (ataxia), a condition that involves the paralysis of the eyes called ophthalmoplegia, and a generalized absence of reflexes (areflexia).  A Cochrane Neuromuscular Review in 2007 identified no randomized prospective controlled trials of immunotherapy in Fisher syndrome (Overall JR, 2007).  Several case reports and case series have been published on the treatment of Fisher syndrome with IVIG, and patients who received IVIG experienced clinical recovery in a slightly shorter time frame than would be expected based on the natural history of the disease (Arakawa, 1993; Zitko, 1994; Benito-Leon, 1996; Bushra, 2000; Kaushik, 2005; Mori, 2007).  Neither AHFS nor Clinical Pharmacology Online recommends IVIG for Fisher’s syndrome.  The AANEM states, “On the basis of the single retrospective analysis and case reports, it is difficult to clearly define the role of VIG in treating Fisher syndrome.  The literature suggests that best medical management may suffice for many patients (Donofrio, 2009).
 
GOOD SYNDROME (Durable Hypogammaglobulinemia Associated with Thymoma)
Good syndrome is characterized by hypogammaglobulinemia and acquired immunodeficiency associated with benign or malignant thymoma (Good, 1954).  Autoimmunity to to B lymphocyte lineage causes severe deficiency in B lymphocytes and hypogammaglobulinemia, resulting in acquired immunodeficiency vulnerable especially to bacterial infection (Kelleher, 2003).  Hypogammaglobulinemia and susceptibility to infection may persist for several years after resection of the thymoma (Tarr, 2001; Kitamura, 2009).  IVIG replacement is recommended by the International Union of Immunological Societies (IUIS Scientific Committee, 1999)
 
Guillain-Barre Syndrome
Two randomized studies comparing IVIg with plasma exchange reported equivalent outcomes for the two treatment approaches. In addition, one study has reported that there is no health benefit in combining plasma exchange with IVIg. Therefore, choice of therapy may be dictated by practical concerns, i.e., access to plasma exchange and need for hospitalization.
 
HEMOLYTIC DISEASE OF NEWBORN WITH JAUNDICE (ISO-IMMUNE HEMOLYTIC DISEASE OF NEWBORN)
A Cochrane review of 3 randomized controlled trials totalling 189 infants with rhesus and ABO incompatibility concluded, “Although the results show a significant reduction in the need for exchange transfusion in those treated with IVIG, the applicability of the results is limited.  The number of studies and infants included is small and none of the 3 included studies was of high quality…Further well designed studies are needed before routine use of IVIG can be recommended for the treatment of iso immune hemolytic jaundice (Alcock, 2002).  This review was updated in 2009 with a similar conclusion: “The role of IVIG remains uncertain, particularly in hospitals that do not use early rate of rise of bilirubin as an indicator for exchange transfusion.  Since it appears safe, it may have a role in special circumstances such as parental refusal for exchange transfusion, or where appropriate blood components for exchange transfusion are unavailable.  However, routine use of IVIG for the treatment of iso immune hemolytic jaundice cannot yet be recommended (Alcock, 2009).  The American Academy of Allergy and Immunology states, “Multiple anecdotal reports demonstrate benefit from the use of IVIG in autoimmune hemolytic anemia, but the use of IVIG should be considered only when other therapeutic modalities fail.  IVIG might decrease the need for exchange transfusion in neonates with iso-immune hemolytic jaundice.  However, there are methodologic flaws with these studies, and routine use in this setting is not recommended.”  Two randomized controlled trials reported in 2011 found conflicting results: A trial of 80 infants from the Netherlands, which randomly assigned neonates with rhesus hemolytic disease for IVIG or placebo, found no reduction in the need for exchange transfusion or rates of other adverse neonatal outcomes (Smits-Wintjens, 2011).  A study from Egypt of 90 infants were randomly assigned to 3 groups: low dose IVIG (0.5gm/kg), high dose IVIG (1g/Kg), and placebo.  There was a more rapid decrease in bilirubin, the duration of phototherapy, and hospital stay (Elalfy, 2011).
 
HYPERIMMUNOGLOBULIN M SYNDROME
Hyper-IgM syndromes are a group of disorders characterized by hypogammaglobulinemia with severely impaired production of specific antibody.  Children with hyper-IgM syndrome have decreased levels of IgG and IgA and increased or normal levels of IgM.  Although B cells are present, there is an inability to generate specific antibody.  As a result, these individuals have recurrent infections similar to those of patients with agammaglobulinemia.  Regular replacement therapy with IVIG is crucial for individuals with this disorder, whether it is due to the X-linked or autosomal recessive varieties (Levy, 1997; Winkelstein, 2003; Quartier, 2004).  Hyperimmunoglobulin M is also seen in patients with Waldenstrom’s hyperglobulinemia, and rarely in B-cell lymphomas, and may be associated with low immunoglobulin G.  The American Academy of Allergy and Immunology (Orange, 2006) recommends IVIG for children with X-linked hyper-IgM syndrome and other autosomal recessive varieties (Orange, 2006).
 
INCLUSION BODY MYOSITIS
Inclusion body myositis (IBM) is a late onset inflammatory muscle disease (myopathy) with a distinctive pattern of proximal and distal limb atrophy and weakness.  In published series it has accounted for between 17 to 30% of all idiopathic inflammatory myopathies but the true proportion may be much higher.  In neuromuscular disease referral centers IBM is the commonest acquired myopathy over the age of 50 years.  Muscle biopsy is essential for diagnosis and can be pathognomonic. It usually shows varying degrees of inflammation, with muscle fibres containing rimmed vacuoles but also additional features which include the presence of nuclear and cytoplasmic inclusions and the abnormal deposition of proteins such as amyloid, ubiquitin and phosphorylated tau. There are agreed criteria for the diagnosis of IBM utilizing clinical, laboratory and biopsy findings.  In the majority of cases IBM is a sporadic and isolated disorder. However there are a few cases where IBM occurs in association with an autoimmune disease.  In rare instances familial cases of IBM have occurred but as these are usually not associated with inflammation they are called hereditary inclusion body myopathies (h-IBM) They are a heterogenous group of disorders characterized clinically by progressive muscular weakness and atrophy beginning in the hands or feet. So far, five different hereditary forms (1 autosomal recessive, 4 autosomal dominant) have been characterized. Dalakas and colleagues have reported on a double-blind, placebo-controlled crossover study comparing IVIg to placebo in 19 patients with inclusion body myositis (Delakas, 1997).  There was no statistically significant improvement in overall muscle strength in the IVIg group compared to the control placebo group. (Delakas, 2001) reported a RCT of 36 patients with biopsy-proven IBM who received IVIG + prednisone versus placebo + prednisone.  IVIG combined with prednisone for a 3-month period was not effective in IBM.  A crossover study of 22 patients with sporadic IBM were treated with high-dose IVIG versus placebo (Walter, 2000).  Treatment was considered of mild benefit.  The European Federation of Neurological Sciences (Skeie, 2006); the American Academy of Allergy & Immunology (Orange, 2006); and the Canadian IVIG Hematology & Neurology Expert Panels (Robinson, 2007) recommend against the use of IVIG for treatment of inclusion body myositis.
 
IRAK4  IMMUNODEFICIENCY
Four Mendelian primary immunodeficiencies associated with impaired signaling of the Toll Like Receptor (TLR) pathway have been reported, one of which is IRAK4 (Interleukin-1 Receptor Associated Kinase 4).  Autosomal recessive IRAK-4 deficiency was first discovered in 2003 (Picard, 2003) and has been described in a less than 100 patients as of 2011.  Patients with IRAK-4 deficiency present with invasive and non-invasive pyogenic bacterial infections, but have normal resistance to common fungi, parasites, viruses, and many bacteria.  Clinical status andoutcome improve with age thought to be due to the development of adaptrive antigen-specific T- and B-lymphocyte responses.  IVIG is recommended empirically until the patient is at least 10 years old (Picard, 2011).  IVIG for IRAK4 immunodeficiency meets coverage criteria based on Expert Opinion, as the number of cases would not allow for appropriate treatment trials, and case studies to date indicate benefit .
 
Kawasaki Syndrome and Other Vasculitides
Randomized, multicenter studies have shown that high-dose IVIg plus aspirin, given within the first 10 days after the onset of fever, is safe and effective in reducing the prevalence of coronary artery abnormalities in Kawasaki Syndrome.
 
A RCT of single course IVIg (n=17) vs. placebo (n=17) in patients with persistent active Wegener’s granulomatosis or microscopic polyangiitis associated with anti-neutrophil cytoplasmic antibody found significantly more responders in the IVIg treatment group at 3 months, but no significant differences after 3 months or in the frequency of relapse or use of other medications.  Data are inadequate regarding the effectiveness of IVIg in other vasculitides including polyarteritis nodosa and rheumatoid arthritis.
 
MULTIFOCAL MOTOR NEUROPATHY
In 2010, the Joint Task Force of the European Federation of Neurological Societies and the Peripheral Nerve Society published guidelines on the management of multifocal motor neuropathy (MMN). The guideline recommends the use of IVIg in the initial and maintenance treatment in patients with MMN (Joint Task Force of the EFNS and the PNS, 2010). Four randomized, controlled, double-blind trials of IVIg for treating MMN (Azulay et al., 1994; Van den Berg et al., 1995; Federico et al., 2000; Le´ ger et al., 2001) have been summarized in a Cochrane systematic review (van Schaik et al., 2005). IVIg treatment was superior to placebo in inducing an improvement in muscle strength in patients with MMN.
 
Multiple Myeloma
A review of the current literature shows several review and meta-analysis articles but no reports of trials using IVIG for myeloma patients before or after stem cell transplant.
 
Blade and Rosifiol (2007) stated the highest risk of infection in myeloma patients is within the first two months of initiation of therapy and in patients with relapsed and refractory disease.  The increased susceptibility to infections is multifactorial with the major cause being the impaired antibody production leading to a decrease in uninvolved immunoglobulins.  There has been controversy about infection prophylaxis in patients with MM but high-dose immunoglobulins are not recommended.
 
Raanani (2009) also noted in his meta-analysis that causes for infections in patients who receive HSCT are multifactorial and consist also of causes other than hypogammaglobulinemia.  The authors concluded there was no advantage to the use of IVIG in terms of survival or infection prevention.
 
Other authors have also recommended the limited use of IVIG for myeloma patients: Nucci (2009): consider IVIG if serum IgG level < 500 mg/dL and recurrent serious infection despite prophylactic antibiotics.  Ludwig (2007): prophylactic measures such as long-term antibiotic or antiviral medication or use of intravenous immunoglobulins is recommended in patients with repeated infectious complications.
 
Multiple Sclerosis
In terms of its impact on the acute exacerbation rate in patients with relapsing-remitting MS, IVIg is comparable to treatment with beta interferon. Despite a reduction in the acute exacerbation rate, data are inconclusive regarding the impact of IVIg on patients’ disability. There are insufficient data to evaluate the effectiveness of IVIg in treating patients with chronic progressive MS.
 
A high-quality RCT compared IVIg to placebo in 67 patients, the majority of whom had chronic progressive disease; the trial was terminated early due to lack of benefit either in patients who remained clinically stable or in those with evidence of disease activity. Small series data also suggest no benefit.
 
In 2002 the American Academy of Neurology published a technology assessment on therapies for multiple sclerosis.  This assessment provided a rating of the recommendations, including A (established as effective), B (probably effective, ineffective, or harmful), C (possibly effective, ineffective or harmful) or U (data inadequate). This assessment offered the following recommendation regarding IVIg:  The studies of intravenous immunoglobulin (IVIg) to date, have generally involved small numbers of patients, have lacked complete data on clinical and MRI outcomes, or have used methods that have been questioned. It is, therefore, only possible that IVIg reduces the attack rate in relapsing-remitting multiple sclerosis. (Type C recommendation)
 
The current evidence suggests that IVIg is of little benefit with regard to slowing disease progression. (Type C recommendation). In contrast, the American Academy of Neurology recommended the use of interferon beta (Type B recommendation) and glatiramer acetate (Type A recommendation). This assessment suggests that IVIg is no longer considered a drug of choice for relapsing-remitting MS, and thus the policy statement in this policy has been revised to indicate that IVIg is not medically necessary. A literature search for the period of 2002 to December 2004 did not identify any additional randomized trials that would prompt reconsideration of the conclusions of the American Academy of Neurology assessment.
 
Myasthenia Gravis
One RCT (total n=87) and one retrospective chart review (total n=54)  compared IVIg treatment to plasma exchange in acute myasthenic crisis. Myasthenic crisis was defined as an acute episode of respiratory muscle weakness, defined by an forced vital capacity (FVC) of <=1.0 liter or negative inspiratory force of <= 20 cm H20, or requirement of mechanical ventilation. One crossover study compared these therapies in 12 patients with moderate-to-severe disease in a stable phase. Results for all 3 trials showed that IVIg and plasma exchange had similar efficacy over time, although improvement may be more rapid with plasma exchange. Series data support benefit with IVIg treatment in patients with acute exacerbations and with refractory disease or who are unable to tolerate standard treatment. One series of 10 children with refractory disease suggests short-term benefit with IVIg but limited long-term benefit.  Refractory myasthenia gravis has been defined as those patients with persistent symptoms despite immunosuppressive treatment with prednisone and/or azathioprine or those unable to tolerate steroid therapy.
 
Feasby et al.: Given the limited evidence available, the expert panel agreed IVIG should not be used as maintenance therapy for chronic myasthenia gravis.
 
Skeie et al.: Trials have not shown evidence of improved functional outcome or steroid-sparing effect with repeated use of IVIG in moderate or severe stable MG (class I evidence).
 
Multiple resources were reviewed for dosing recommendations for Myasthenia Gravis.  In a study by Barth D, et al (2011), 84 patients were randomized with moderate to severe MG defined as a Quantitative Myasthenia Gravis Score for disease severity (QMGS) of >10.5 and worsening weakness to IVIg (Gamunex®, Talecris Biotherapeutics) 1 g/kg/day for 2 consecutive days or PLEX (Caridian Spectra) 1.0 plasma volume exchanges for 5 exchanges. The patients were evaluated at day 14 after treatment for the primary efficacy parameter of change in QMGS and secondary clinical and electrophysiologic parameters and were followed for a total of 60 days.  Findings showed that both IVIg and PLEX reduced the QMGS and IVIg was comparable to PLEX in efficacy.  
 
In a Cochrane review, seven RCTs comparing IVIg to no treatment, placebo or PLEX, in people with MG were identified.  One trial with 168 people with exacerbations, showed no evidence of superiority of IVIg 2g/kg over IVIg 1 g/kg on the change of MMS after 15 days. (MD 3.84; 95% Cl -0.98 to 8.66).
 
Cosi V et al, (1991) had 37 patients affected by autoimmune generalized myasthenia gravis (MG) with highdose intravenous gammaglobulin (HDIVIg), 400 mg/kg per day on 5 consecutive days. A onedegree improvement of Oosterhuis global clinical classification of myasthenic severity (OGCCMS), the disappearance of bulbar involvement or both were recorded 12 days after the beginning of the treatment in 70.3% of the patients and persisted up to 60 days in 58.7%. A twodegree improvement of OGCCMS was recorded in 54.1% of the patients and it was maintained up to 60 days in 37.8%. The percentage of improvement did not significantly differ between patients entering the treatment in a longstanding, drugrefractory stationary phase of the illness (n = 26) and patients who received HDIVIg in an acute phase of MG (n = 11).
 
MYD88 gene deficiency
Four Mendelian primary immunodeficiencies associated with impaired signaling of the Toll Like Receptor (TLR) pathway have been reported, one of which is autosomal recessive MyD88 deficiency which was first discovered in 2008. (von Bernuth, 2008).  Deficiency in the MYD88 gene results in invasive pyogenic infections.  IVIG is not recommended for patients with MYD88 gene deficiency by any review group, and is not recommended in the review of this deficiency by Picard, et al (Picard, 2011).
 
NEONATAL SEPSIS
A 2010 Cochrane review found no benefit, but recommended waiting on the International Neonatal Immunotherapy Study results before a definitive conclusion was reached.  The INIS Collaborative Group randomized controlled trial was reported in September 2011 (Brocklehurst, 2011) and found no benefit of IVIG in infants with neonatal sepsis.
 
PARANEOPLASTIC RETINOPATHY (CANCER ASSOCIATED RETINOPATHY, MELANOMA ASSOCIATED RETINOPATHY, AUTOIMMUNE RETINOPATHY ASSOCIATED WITH NEOPLASM) Paraneoplastic vision loss is characterized by sudden, progressive loss of vision associated with photosensitivity, ring scotomata, visual field defects, and abnormal electroretinogram believed to be caused by an autoimmune effect on the retina or optic nerve from anti-retinal autoantibodies associated with a remote cancer (Jacobson, 1990; Guy, 1999; Katsuta, 2002).  Autoantibodies (anti-recoverin, anti-enolase, anti-carbonic anhydrase II, and anti-transducin) affect both rod and/or cone function, causing rapid visual loss (Murphy, 1997; Adamus, 2004; Dot, 2005; Weleber, 2005; Adamus, 2009).  No clinical trials have been published on the use of IVIG for treatment of paraneoplastic vision loss, cancer associated retinopathy, or melanoma associated retinopathy.  No published paper from any IVIG expert panel has reviewed IVIG for treatment of paraneoplastic vision loss.  A few case reports suggest improvement following IVIG for paraneoplastic vision loss (Guy,1999; Keltner, 2001; Subhadra, 2008), and reduction in anti-enolase antibody has been described following IVIG (Adamus (2004).  Neither AHFS nor Clinical Pharmacology Online drug compendia recommend IVIG for treatment of paraneoplastic retinopathy.  IVIG for paraneoplastic retinopathy is considered to meet Primary Coverage Criteria because 1) of the critical nature of this disorder; 2) of the lack of other appropriate therapeutic measures; 3) it is unlikely that prospective trials will be performed due to the rarity of this condition; and 4) there are published case series indicating benefit.  
 
Paraneoplastic Syndrome (Eaton-Lambert Disease)
Results of a double-blind, placebo-controlled, crossover, randomized study of IVIg versus placebo in 11 patients with paraproteinemic IgM demyelinating polyneuropathy showed only a mild and transitory effect in 3 patients.  There are inadequate data on the use of IVIg in paraneoplastic syndromes, such as Eaton-Lambert disease.  
 
PEMPHIGUS AND OTHER AUTOIMMUNE BLISTERING DISEASES
Corticosteroids and immunosuppressive agents are considered first line therapy. IVIG has been shown to decrease circulating autoantibodies in pemphigus, and addition of immunosuppressive drugs to IVIG has an additive effect.  Based on uncontrolled studies (Ahmed, 2003), IVIG therapy is the preferred therapy for patients who are resistant, nonresponsive, have contraindications, or develop serious catastrophic adverse effects.  No clinical trials have been reported for IVIG for pemphigus or other autoimmune blistering diseases.  Two randomized controlled trials are listed on clinicaltrial.gov.  NCT00483119 is an ongoing FDA sponsored trial at NYU Medical Center, initiated in 2007 with estimated completion date of 2011, comparing IVIG versus IVIG plus cyclophosphamide in patients with pemphigus that are not responding to, or have developed complications from, standard therapy.  A second study, in Japan (NCT00809822) recently completed but unreported, is a randomized, placebo controlled trial of IVIG for patients with bullous pemphigoid unresponsive to corticosteroids.   Prospective, non-randomized, open label clinical studies on patients who are unresponsive to corticosteroids and immunosuppressive agents have shown response to IVIG (Toth, 1999; Engineer, 2000; Ahmed, 2001; Sami, 2002; Bystryn, 2002; Ahmed, 2002; Baum, 2006).  IVIG has not been compared to newer biologics (e.g., anti-tumor necrosis alfa drugs).
 
POST TRANSPLANT HYPOGAMMAGLOBULINEMIA
Hypogammaglobulinemia has been identified following lung (Kawut, 2005; Yip, 2006; Robertson, 2009), heart (Corales, 2000), AND kidney (Boddana, 2011) transplantation.  There has been consideration that this may be due to the use of mycophenolate mofetil to prevent allograft rejection, and this drug is known to be associated with an increased risk of bacterial infection.   
 
POST TRANSPLANT LYMPHOPROLIFERATIVE DISORDER
PDLDs are a heterogenous group of abnormal lymphoid proliferations that occur after solid organ transplant or hematopoietic transplantation.  PTLDs range from hyperplasia to aggressive lymphomas with 60-70% being Epstein-Barr virus positive.
 
PREVENT RECURRENT PREGNANCY LOSS IN UNSELECTED POPULATION  
A Cochrane review on Immunotherapy for recurrent miscarriage published initially in 2006, and edited in 2010, looked at 20 trials on various forms of immunotherapy, including 7 with IVIG, for treatment of recurrent abortion.  They concluded that IVIG provided no significant beneficial effect over placebo in approving live birth rate (Porter, 2010).  Another systematic review of eight trials (Hutton, 2007) involving 442 women found that IVIG did not significantly increase the odds ratio of live birth when compared with placebo for treatment of primary recurrent miscarriage (primary - miscarriage occurring in women with no recognized cause, e.g, antiphospholipid syndrome).  
 
PREVENT RECURRENT PREGNANCY LOSS IN ANTIPHOSPHOLIPID SYNDROME
The Pregnancy Loss Study Group multicenter, placebo-controlled pilot study of IVIG treatment of antiphospholipid syndrome during pregnancy compared treatment with heparin and low-dose aspirin plus IVIG with heparin and low-dose aspirin plus placebo in a group of 16 women who met strict criteria for antiphospholipid syndrome in 16 women (Branch, 2000).  Obstetrical outcomes were excellent in both groups, with all women being delivered of live-born infants after 32 weeks gestation.  A second randomized controlled trial of 40 women with recurrent abortion (at least 3 occurrences) and repeatedly positive test results for anticardiolipin or lupus anticoagulant were randomized to receive IVIG or low molecular weight heparin plus low dose aspirin (Triolo, 2004).  The women treated with low molecular weight heparin plus low-dose aspirin had a higher rate of live births (84%) than those treated with IVIG (57%).  A third randomized controlled trial (Dendrinos, 2009) randomly allocated 85 women to receive low molecular weight heparin plus low-dose aspirin or IVIG.  Low molecular weight heparin plus low-dose aspirin (72.5%) resulted in a significantly higher live birth rate than IVIG (39.5%) in treatment of antiphospholipid antibody syndrome in women with recurrent abortion.  The American Academy of Allergy, Asthma, and Immunology recommends against use of IVIG to prevent pregnancy loss in women with antiphospholipid syndrome (Orange, 2006).
 
PURE RED CELL APLASIA (PRCA)
Pure red-cell aplasia is defined as the absence of mature erythroid precursors in a bone marrow that otherwise exhibits normal cellularity.  Acquired pure red-cell aplasia may occur in association with neoplasms (such as lymphoproliferative disorders), thymoma, autoimmune disorders, pregnancy, or as a consequence of chronic human parvovirus B19 infection in an immunologically incompetent host.  PRCA may also develop after exposure to drugs (erythropoietin or tacrolimus).  
 
There are case reports of effectiveness of IVIG in these conditions, and also in patients with HIV.  The Canadian Hematological Expert Panel recommends IVIG for pure red blood cell aplasia if the PRCA is refractory to standard therapy.
 
RHEUMATIC FEVER, ACUTE
A randomized placebo-controlled trial in 59 patients with rheumatic fever who were treated with either IVIG or placebo revealed that IVIG treatment did not reduce the valvulitis or altered the natural history of the disease (Voss, 2001). A Cochrane Review concludes that IVIG is not indicated in treatment of acute rheumatic fever (Cillers, 2003).
 
STAPHYLOCOCCAL TOXIC SHOCK SYNDROME
According to the Drug Compendia, the American College of Pediatrics suggests that IGIV may be considered in the management of severe staphylococcal toxic shock syndrome when the infection is refractory to several hours of aggressive therapy, an undrainable focus is present, or the patient has persistent oliguria with pulmonary edema.  The American Academy of Allergy, Asthma, and Immunology lists IVIG as “probably beneficial” in staphylococcal toxic shock (Orange, 2006) although they provide no references.  A review of therapeutic uses of immunoglobulins in pediatrics (Stiehm, 2010) lists IVIG as probably beneficial, and references from two textbooks (Pickering, 2009; Feigen, 2009), support this use.
 
STEVENS-JOHNSON SYNDROME
Stevens-Johnson syndrome is a potentially fatal disorder.  A prospective non-comparative study of IVIG showed no benefit on mortality or progression (Bachot, 2003).   The American Academy of Allergy, Asthma, and Immunology recommends IVIG as “probably beneficial” for treatment of Stevens-Johnson syndrome (Orange, 2006), although the reference given in support of that decision (Trent, 2003) addresses toxic epidermal necrolysis only.  The Medical Letter (2006b) states that there is no clear evidence of benefit for Stevens Johnson syndrome, “but [it is]thought to be potentially life-saving.  Gurcan and Ahmed in their review of IVIG state, “The current viewpoint is that IVIG may be effective in Stevens-Johnson syndrome and toxic epidermal necrolysis if used very early (within 48 h of onset of clinical features) and at high doses for 3-5 days consecutively (Gurcan, 2007).  
 
THROMBOCYTOPENIA, NEONATAL ALLOIMMUNE
IVIG, to prevent severe intra-uterine thrombocytopenia, given to an expectant mother who had previously delivered an infant with neonatal alloimmune thrombocytopenia, was initially described in a prospective case series of 7 women  (Bussel, 1988).  Since that time there have been a number of case series and prospective single arm or comparative drug trials of IVIG (Bussel, 1996; Berkowitz, 2006; Berkowitz, 2007), but there have been no randomized controlled trials (RCTs) comparing IVIG to no therapy (Rayment, 2005; Bassler, 2008).
 
The American Academy of Allergy and Immunology states, “there are anecdotal data supporting the use of IVIG for antenatal therapy of fetomaternal alloimmune thrombocytopenia.  Although there are no randomized trials to support this practice, use of IVIG has become routine first-line therapy in this setting.” (Orange, 2006).  A Cochrane review of IVIG for antenatal interventions for fetomaternal alloimmune thrombocytopenia concluded, “There are insufficient data from randomized controlled trials to determine the optimal antenatal management of fetomaternal alloimmune thrombocytopenia” (Rayment, 2005).  The American College of Obstetrics and Gynecology published a state of the art paper in 2006 stating this therapy is the treatment of choice (Berkowitz, 2006).  In a review of neonatal alloimmune thrombocytopenia, (Vinograd, 2010), the authors concluded, “[N]oninvasive approaches and the implementation of risk stratification (including combination therapy of IVIG with steroids and/or the use of more than 1gm/kg/week of IVIG) are appropriate in the management of fetal alloimmune thrombocytopenia.  Neither the AHFS nor the Clinical Pharmacology Online compendia have position statements on this therapy.  A RCT of antenatal IVIG given in either 2gm/kg/wk or 1gm/kg/wk plus prednisone starting at approximately 20 weeks of gestation, in women with very high risk, high risk and standard risk for neonatal alloimmune thrombocytopenia was initiated in 2001 (expected completion date Feb 2012) at Weill Medical College of Cornell University (NCT00194987), but results of the full study are not yet available.  The subset of 73 women in the standard risk group (defined as women with documented alloimmune thrombocytopenia who had not delivered an infant with an intracranial hemorrhage in a prior pregnancy) showed “excellent” outcomes inb both groups (Berkowitz, 2007).
 
TRANSPLANTATION, ANTIBODY MEDIATED REJECTION
One RCT of 30 patients published in 2001 suggested that IVIg is at least as good as anti-CD3 in combating corticosteroid-resistant rejection of kidney transplants (Casadei, 2001). Later, in 2003-4, findings from the NIH IG02, a double-blind placebo-controlled trial, were published (Jordan, 2003).  The trial randomized 101 highly sensitized renal transplant candidates to receive either 4 monthly infusions of IVIg or placebo prior to transplant. If transplanted, additional infusions were given monthly for 4 months. IVIg significantly reduced PRA levels in study subjects compared to placebo, resulting in a higher transplant rate. For example, a total of 24 patients subsequently underwent transplant, 16 in the IVIg group and 8 in the placebo group. There was acceptable graft survival in both groups. Desensitization protocols varied among transplant centers; certain protocols commonly used are referred to as the Cedars-Sinai protocol and the Johns Hopkins protocol. The Cedars-Sinai protocol consisted of high-dose IVIg (2 g/kg) and was offered to patients awaiting either a deceased or live donor (Jordan, 2003). The Johns Hopkins protocol consisted of low-dose IVIg (100 mg/kg) in combination with plasmapheresis with or without treatment with anti-CD-20 (i.e., Rituxan) (Montgomery, 2004).
 
A retrospective cohort study published in 2009 compared outcomes in pediatric liver transplant patients entered into a multicenter Registry who did (n=336) and did not (n=1,612) receive IVIg within 7 days of transplantation (Bucuvalas, 2009). The investigators assumed that IVIg given within this timeframe was used for prophylaxis of antibody mediated rejection (AMR), rather than for treatment. The Kaplan-Meier probability of patient survival was not significantly different between groups (hazard ratio [HR]: 0.97, 95% CI: 0.71-1.39). However, the risk of graft rejection was significantly lower in patients treated with immunoglobulin. In the first 3 months after transplant, 31% of patients who received immunoglobulin and 40% of those not treated had an episode of graft rejection (p=0.02). Similarly, the proportion of patients with 2 or more episodes of graft rejection was significantly lower among those who received immunoglobulin (13.1%) than those who did not (19.2%), p=0.009. Patients were not randomized to treatment group, and there may have been differences in those treated or not treated with immunoglobulin that affected outcomes.
 
A variety of protocols also have been developed for the treatment of AMR, often in combination with other therapies, such as plasmapheresis or anti-CD-20 (Casadei, 2001; Ibernon, 2005; Jordan, 2005; Lehrich, 2005). The majority of studies of IVIg in the transplant setting are retrospective case series from single institutions. Therefore, it is not possible to compare immunomodulatory regimens to determine their relative efficacy. Nevertheless, in part based on the large volume of literature published on this subject, it appears that IVIg is a component of the standard of care for the management of AMR.
  
VON WILLEBRAND’S DISEASE, ACQUIRED  
The Canadian IVIG Hematology Expert Panel does not recommend IVIG for routine use in the treatment of acquired Von Willebrand’s disease, but states “IVIG may be considered one option among adjunctive therapies in the treatment of acquired von Willebrand’s disease in urgent situations (e.g., active bleeding or preoperatively) (Robinson, 2007).  No other major medical society has published recommendations on the use of IVIG in von Willebrand’s disease.  Neither the AHFS nor Clinical Pharmacology Online recommend coverage of IVIG for von Willebrand’s disease (2011).
 
WEST NILE VIRUS FEVER
The West Nile virus has become endemic in the U.S. since 1999 (Lanciotti, 1999), has been found in mosquitoes, birds, horses, and other animals (Rappole, 2000; Campbell, 2002), and antibodies against West Nile virus have become much more common in the plasma of blood donors.  Reports have been published of transmission of the disease following solid organ transplantation (Iwamoto, 2003; Rhee 2011).  There is no specific treatment for West Nile virus infection, and in most cases the disease is self-limited and treated with supportive care (although some have predicted long-term neurological deficit (Sejvar, 2007).  But in persons who are immunosuppressed, or who are elderly, are at risk of fatal meningoencephalitis.  There are single and multi-case reports of use of IVIG with high specific antibody titers against West Nile virus with apparent clinical improvement (Shimoni, 2001; Hamdan, 2002; Haley, 2003; Makhoul, 2009; Saqueib, 2008; Rhee, 2011).  IVIG is recommended for treatment of confirmed West Nile encephalomyelitis by Clinical Pharmacology Online (2012).
 
2019 Update
A literature search conducted through October 2019 did not reveal any new information that would prompt a change in the coverage statement.
 
2020 Update
A literature search conducted through October 2020 did not reveal any new information that would prompt a change in the coverage statement.
 
2021 Update
A literature search conducted through October 2021 did not reveal any new information that would prompt a change in the coverage statement.
 
2022 Update
Annual policy review completed with a literature search using the MEDLINE database through September 2022. No new literature was identified that would prompt a change in the coverage statement.
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through September 2023. No new literature was identified that would prompt a change in the coverage statement.
 

CPT/HCPCS:
90283Immune globulin (IgIV), human, for intravenous use
90284Immune globulin (SCIg), human, for use in subcutaneous infusions, 100 mg, each
C9399Unclassified drugs or biologicals
J1459Injection, immune globulin (privigen), intravenous, non lyophilized (e.g., liquid), 500 mg
J1460Injection, gamma globulin, intramuscular, 1 cc
J1551Injection, immune globulin (cutaquig), 100 mg
J1554Injection, immune globulin (asceniv), 500 mg
J1555Injection, immune globulin (cuvitru), 100 mg
J1556Injection, immune globulin (bivigam), 500 mg
J1557Injection, immune globulin, (gammaplex), intravenous, non lyophilized (e.g., liquid), 500 mg
J1558Injection, immune globulin (xembify), 100 mg
J1559Injection, immune globulin (hizentra), 100 mg
J1560Injection, gamma globulin, intramuscular, over 10 cc
J1561Injection, immune globulin, (gamunex c/gammaked), non lyophilized (e.g., liquid), 500 mg
J1562Injection, immune globulin (vivaglobin), 100 mg
J1566Injection, immune globulin, intravenous, lyophilized (e.g., powder), not otherwise specified, 500 mg
J1568Injection, immune globulin, (octagam), intravenous, non lyophilized (e.g., liquid), 500 mg
J1569Injection, immune globulin, (gammagard liquid), non lyophilized, (e.g., liquid), 500 mg
J1572Injection, immune globulin, (flebogamma/flebogamma dif), intravenous, non lyophilized (e.g., liquid), 500 mg
J1575Injection, immune globulin/hyaluronidase, (hyqvia), 100 mg immuneglobulin
J1599Injection, immune globulin, intravenous, non lyophilized (e.g., liquid), not otherwise specified, 500 mg
J3490Unclassified drugs
J3590Unclassified biologics
J9999Not otherwise classified, antineoplastic drugs

References: Azulay JP, Blin O, Pouget J, et al.(1994) Intravenous immunoglobulin treatment in patients with motor neuron syndromes associated with anti-GM1 antibodies: a double-blind, placebo-controlled study. Neurology 44:429–432.

Schoser B, Jacob S, et al.(2009) Immune-mediated rippling muscle disease with myasthenia gravis: a report of seven patients with long-term follow-up in two. Neuromuscul Disord, 2009; 19:223-8.

0Ohlsson A, Lacy JB.(2015) Intravenous immunoglobulin for suspected or proven infection in neonates. Cochrane Database Syst Rev. 2015;3:CD001239. PMID 25815707

1998 Blue Cross Blue Shield Association Technology Evaluation Center Assessment; Tab 14.

1998 Blue Cross Blue Shield Association Technology Evaluation Center Assessment; Tab 19.

Achilli C, Duran-Retamal M, Saab W, et al.(2018) The role of immunotherapy inin vitro fertilization and recurrent pregnancy loss: a systematic review and meta-analysis. Fertil Steril. 2018 Nov;110(6):1089-1100. doi: 10.1016/j.fertnstert.2018.07.004. PMID: 30396553

Al-Mayouf SM, Laxer RM, Schneider R, et al.(2000) Intravenous immunoglobulin therapy for juvenile dermatomyositis: efficacy and safety. J Rheumatol 2000; 27(10):2498-503.

Alabdali M, Barnett C, Katzberg H, et al.(2014) Intravenous immunoglobulin as treatment for myasthenia gravis: current evidence and outcomes. Expert Rev Clin Immunol. 2014 Dec;10(12):1659-65. doi: 10.1586/1744666X.2014.971757. Epub 2014 Oct 20.

Alcock GS, Liley H.(2002) Immunoglobulin infusion for isoimmune haemolytic jaundice in neonates. Cochrane Database Syst Rev. 2002(3):CD003313. PMID 12137687

American Society for Reproductive Medicine, Birmingham, Alabama, USA.(2012) . Evaluation and treatment of recurrent pregnancy loss: a committee opinion. Fertil Steril. 2012 Nov;98(5):1103-11. doi: 10.1016/j.fertnstert.2012.06.048. Epub 2012 Jul 24.

Approval of immune globulin product called Gamunex for treatment of CIDP. http://www.fda.gov/bbs/topics/NEWS/2008/NEW01884.html.

Barron SJ, Del Vecchio MT, Aronoff SC.(2015) Intravenous immunoglobulin in the treatment of Stevens-Johnson syndrome and toxic epidermal necrolysis: a meta-analysis with meta-regression of observational studies Int J Dermatol. Jan 2015;54(1):108-115. PMID 24697283

Barth D, Nabavi NM, Ng E, Nwe P, Bril V.(2011) Comparison of IVIg and PLEX in patients with myasthenia gravis. Neurology. 2011;76(23):2017. Epub 2011 May 11.

Blade J, Rosinol L.(2007) Complications of multiple myeloma. Hematol Oncol Clin N Am, 2007; 21:1231-46.

Boletis J, Ioannidis J, Boki K, et al.(1999) Intravenous immunoglobulin compared with cyclophosphamide for proliferative lupus nephritis. Lancet. 1999;354(9178):569-570.

Boletis JN, Ioannidis JP, Boki KA, et al.(1999) Intravenous immunoglobulin compared with cyclophosphamide for proliferative lupus nephritis. Lancet 1999; 354(9178):569-70.

Branch DW, Peaceman AM, Druzin M, et al.(2000) A multicenter, placebo controlled pilot study of intravenous immune globulin treatment of antiphospholipid syndrome during pregnancy. The Pregnancy Loss Study Group. Am J Ob Gyn 2000; 182(1 Pt 1):122-7.

Bucciarelli S, Espinosa G, Cervera R, et al.(2006) Mortality in the catastrophic antiphospholipid syndrome: causes of death and prognostic factors in a series of 250 patients. Arthritis Rheum. Aug 2006;54(8):2568-2576. PMID 16868979

Bucuvalas JC, Anand R.(2009) Treatment with immunoglobulin improves outcome for pediatric liver transplant recipients. Liver Transpl 2009; 15(11):1564-9.

Bussel JB, Berkowitz RL, Lynch L, et al.(1996) Antenatal management of alloimmune thrombocytopenia with intravenous immunoglobulin: a randomized trial of the addition of low dose steroid to intravenous gammaglobulin. Am J Ob Gyn 1996; 174(5):1414-23.

Cantoni N, Weisser M, et al.(2009) Infection prevention strategies in a stem cell transplant unit: impacto of change of care in isolation practice and routine use of high dose intravenous immunoglobulins on infectious complications and transplant related mortality. Eur J Haematol, 2009:83:130-8.

Casadei DH, del C Rial M, Opelz G, et al.(2001) A randomized and prospective study comparing treatment with high dose intravenous immunoglobulin with monoclonal antibodies for rescue of kidney grafts with steroid resistant rejection. Transplantation 2001; 71(1):53-8.

Caveolinopathies.(2012) Bruno C, Sotgia F, et al. http://www.ncbi.nlm.nih.gov/books?term=Caveolinopathies+AND+gene%5Bbook%5D.

Cherin P, Pelletier S, Teixeira A, et al.(2002) Results and long-term follow-up of intravenous immunoglobulin infusions in chronic, refractory polymyositis: an open study with thirty-five adult patients. Arthrit Rheum 2002; 46(2):467-74.

Christiansen OB, Kolte AM, Krog MC, et al.(2019) Treatment with intravenous immunoglobulin in patients with recurrent pregnancy loss: An update. J Reprod Immunol. 2019 Jun;133:37-42. doi: 10.1016/j.jri.2019.06.001. Epub 2019 Jun 18. PMID: 31238263

Cordonnier C, Chevret S, et al.(2003) Should immunoglobulin therapy be used in allogeneic stem-cell transplantation? A randomized, double-blind, dose effect, placebo-controlled, multicenter trial. Ann Int Med, 2003; 139:8-18.

Cosi V, Lombardi M, Piccolo G, Erbetta A.(1991) reatment of myasthenia gravis with high-dose intravenous immunoglobulin. Acta Neurol Scand. 1991;84(2):81.

Dalakas MC, Fujii M, Li M, et al.(2001) High dose intravenous immune globulin for stiff-person syndrome. NEJM 2001; 345:1870-6.

Dalakas MC, Fujii M, Li M, et al.(2001) High-dose intravenous immune globulin for stiff-person syndrome. NEJM 2001; 345(26):1870-6.

Dalakas MC, Illa I, Dambrosia JM, et al.(1993) A controlled trial of high dose intravenous immune globulin infusions as treatment for dermatomyositis. NEJM 1993; 329(27):1993-2000.

Dalakas MC, Koffman B, Fujii M, et al.(2001) A controlled study of intravenous immunoglobulin combined with prednisone in the treatment of IBM. Neurology 2001; 56(3):323-7.

Dalakas MC, Quarles RH, Farrer RX, et al.(1996) A controlled study of intravenous immunoglobulin in demyelinating neuropathy with IgM gammopathy. Ann Neurol 1996; 40(5):792-5.

Dalakas MC, Sonies B, Dambrosia J, et al.(1997) Treatment of inclusion-body myositis with IVIg: a double-blind, placebo-controlled study. Neurology 1997; 48(3):712-6.

Dalakas MC.(2004) Intravenous immunoglobulin in autoimmune neuromuscular diseases. JAMA, 2004; 291:2367-75.

Danieli MG, Malcangi G, Palmieri C, et al.(2002) Cyclosporin A and intravenous immunoglobulin treatment in polymyositis/dermatomyositis. Ann Rheum Dis 2002; 61:37-41.

Darabi K, Abdel-Wahab O, Dzik WH.(2006) Current usage of intravenous immune globulin and the rationale behind it: the Massachusetts General Hospital data and a review of the literature. Transfusion, 2006; 46:741-53.

Darenberg J, Ihendyane N, Sjolin J, et al.(2003) Intravenous immunoglobulin G therapy in streptococcal toxic shock syndrome: a European randomized, double-blind, placebo-controlled trial. Clin Infect Dis. Aug 01 2003;37(3):333-340. PMID 12884156

Donofrio PD, Berger A, et al.(2009) Consenus Statement: the use of intravenousl immunoglobulin in the treatment of neuromuscular conditions report of the AANEM ad hoc committee. Muscle Nerve, 2009; 40:890-900.

Douzinas EE, Pitaridis MT, Louris G, et al.(2000) Prevention of infection in multiple trauma patients by high-dose intravenous immunoglobulins. Crit Care Med 2000; 28(1):8-15.

Dyck PJ, Litchy WJ, Kratz KM, et al.(1994) A plasma exchange versus immune globulin infusion trial in chronic inflammatory demyelinating polyradiculoneuropathy. Ann Neurol 1994; 36(6):838-45.

Elovaara S, Apostolski S, et al.(2008) EFNS guidelines for the use of intravenous immunoglobulin in treatment of neurological diseases. European J Neurology, 2008; 15:893-908.

Fam AG.(2001) Recent advances in the management of adult myositis. Expert Opin Investig Drugs 2001; 10:1265-77.

Feasby T, Banwell B, et al.(2007) Guidelines on the use of intravenous immune globulin for neurologic conditions. Transfus Med Pract, 2007; 21 Suppl 1:S57-S107.

Federico P, Zochodne DW, Hahn AF, et al.(2000) Multifocal motor neuropathy improved by IVIg: randomized, double-blind, placebo-controlled study. Neurology 2000; 55(9):1256-62.

Federico P, Zochodne DW, Hahn AF, et al.(2000) Multifocal motor neuropathy improved by IVIg: randomized, double-blind, placebo-controlled study. Neurology 55:1256–1262.

Ferguson D, Hutton B, et al.(2005) Use of intravenous immunoglobulin for treatment of neurologic conditions: a systematic review. Transfusion Pract, 2005; 45:1640-57.

Flores G, Cunningham-Rundles C, Newland AC, et al.(1993) Efficacy of intravenous immunoglobulin in the treatment of autoimmune hemolytic anemia: results in 73 patients. Am J Hematol. Dec 1993;44(4):237-242. PMID 8237993

Gajdos J, Chevret S, Toyka KV.(2012) Intravenous immunoglobulin for myasthenia gravis. Cochrane Database Syst Rev. 2012;12:CD002277.

Gajdos P, Chevret S, Clair B, et al.(1997) Clinical trial of plasma exchange and high-dose intravenous immunoglobulin in myasthenia gravis. Myasthenia Gravis Clinical Study Group. Ann Neurol 1997; 41(6):789-96.

Gajdos P, Tranchant C, et al.(2005) Treatment of myasthenia gravis exacerbation with intravenous immunoglobulin: a randomized double-bline clinical trial. Arch Neurol, 2005; 62:1689-93.

Genevay S, Saudan-Kister A, Guerne PA.(2001) Intravenous gammaglobulins in refractory polymyositis: lower dose for maintenance treatment is effective. Ann Rheum Dis 2001; 60:635-6.

Gold R, Stangel M, Dalakas MC.(2007) Drug Insight: the use of intravenous immunoglobulin in neurology-therapeutic considerations and practical issues. Nat Clin Prac Neurol, 2007; 3:36-44.

Gottfried I, Seeber A, Anegg B, et al.(2000) High dose intravenous immunoglobulin (IVIG) in dermatomyositis: clinical responses and effect on sIL-2R levels. Eur J Derm 2000; 10(1):29-35.

Hahn AF, Bolton CF, Zochodne D, et al.(1996) Intravenous immunoglobulin treatment in chronic inflammatory demyelinating polyneuropathy. A double-blind, placebo-controlled, cross-over study. Brain 1996; 119(Pt 4):1067-77.

Hargreaves RM, Lea JR, et al.(1995) Immunological factors and risk of infection in plateau phase myeloma. J Clin Pathol, 1995; 48(3):260-6.

Huang YH, Chen HC, Huang KW, et al.(2015) Intravenous immunoglobulin for postpolio syndrome: a systematic review and meta-analysis. BMC Neurol. 2015;15:39. PMID 25886512

Hughes RA, Donofrio P, Bril V, et al.(2008) Intravenous immune globulin (10% caprylate-chromatography purified) for treatment of chronic inflammatory demyelinating polyradiculoneuropathy (ICE study): a randomized placebo-controlled trial. Lancet Neurol, 2008; 7:136-144.

Hundt M, Manger K, Dorner T, et al.(2000) Treatment of acute exacerbation of systemic lupus erythematosus with high-dose intravenous immunoglobulin. Rheumatology (Oxford) 2000; 39(11):1301-2.

Ibernon M, Gil-Vernet S, Carrera M et al.(2005) Therapy with plasmapheresis and intravenous immunoglobulin for acute humoral rejection in kidney transplantation. Transplant Proc 2005; 37(9):3743-5.

Intravenous Immunoglobulin (IVIG) and Recurrent Spontaneous Pregnancy Loss: A Practice Committee Report; A Committee Opinion. Am Society of Reproductive Medicine. http://www.asrm.org/Media/Practice/ivig.html. 1998; Accessed October 2002.

Jablonowska B, Selbing A, Palfi M et al.(1999) Prevention of recurrent spontaneous abortion by intravenous immunoglobulin: a double-blind placebo-controlled study. Hum Reprod 1999; 14(3):838-41.

Jayne DR, Chapel H, Adu D, et al.(2000) Intravenous immunoglobulin for ANCA-associated systemic vasculitis with persistent disease activity. QJM 2000; 93(7):433-9.

Joint Task Force of the EFNS and the PNS.(2010) EFNS/PNS Guideline on management of multifocal motor neuropathy. Report of a Joint Task Force of the EFNS and the PNS - first revision. J Peripher Nerv Syst, 2010; 15:295-301.

Joint Task Force of the EFNS and the PNS.(2010) European Federation of Neurological Societies/Peripheral Nerve Society guideline on management of multifocal motor neuropathy. J Peripher Nerv Syst. 2010 Dec;15(4):295-301.

Jordan S, Tyan D, Stablein D et al.(2004) Evaluation of intravenous immunoglobulin as an agent to lower allosensitization and improve transplantation in highly sensitized adult patients with end-stage renal disease: report of the NIH IG02 trial. J Am Soc Nephrol 2004; 15(12):3256-62.

Jordan S, Vo A, Tyan D et al.(2005) Current approaches to treatment of antibody-mediated rejection. Pediatr Transplant 2005; 9(3):408-15.

Jordan SC, Quartel AW, Czer LS, et al.(1998) Posttransplant therapy using high-dose human immunoglobulin (intravenous gammaglobulin) to control acute humoral rejection in renal and cardiac allograft recipients and potential mechanism of action. Transplantation 1998; 66(6):8005.

Jordan SC, Vo AA, Nast CC et al.(2003) Use of high-dose human intravenous immunoglobulin therapy in sensitized patients awaiting transplantation: the Cedars-Sinai experience. Clin Transpl 2003:193-8.

Kaul R, McGeer A, Norrby-Teglund A, et al.(1999) Intravenous immunoglobulin therapy for streptococcal toxic shock syndrome--a comparative observational study. The Canadian Streptococcal Study Group. Clin Infect Dis. Apr 1999;28(4):800-807. PMID 10825042

Kazatchkine MD, Kaveri SV.(2001) Immunomodulation of autoimmune and inflammatory diseases with intravenous immune globulin. NEJM 2001; 345:747-755.

Kiehl MG, Stoll R, Broder M, et al.(1996) A controlled trial of intravenous immune globulin for the prevention of serious infections in adults with advanced human immunodeficiency virus infection. Arch Intern Med 1996; 156(22):2545-50.

Kishiyama JL, Valacer D, Cunningham-Rundles C, et al.(1999) A multicenter, randomized, double blind, placebo controlled trial of high dose intravenous immunoglobulin for oral corticosteroid dependent asthma. Clin Immunol 1999; 91(2):126-33.

Kress HG, Scheidewig C, Schmidt H, et al.(1999) Reduced incidence of postoperative infection after intravenous administration of an immunoglobulin A- and immunoglobulin M-enriched preparation in anergic patients undergoing cardiac surgery. Crit Care Med 1999; 27(7):1281-7.

Lancaster, E.(2016) The Diagnosis and Treatment of Autoimmune Encephalitis. J Clin Neurol. 2016 Jan;12(1):1-13. Available at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4712273/pdf/jcn-12-1.pdf.

Léger JM, Chassande B, Musset L, et al.(2001) Intravenous immunoglobulin therapy in multifocal motor neuropathy: a double-blind, placebo-controlled study. Brain 124:145–153.

Leger JM, Chassande B, Musset L, et al.(2001) Intravenous immunoglobulin therapy in multifocal motor neuropathy: a double-blind, placebo-controlled study. Brain 2001; 124(Pt 1):145-53.

LeHoang P, Cassoux N, George F, et al.(2000) Intravenous immunoglobulin (IVIg) for the treatment of bird shot retinochoroidopathy. Ocul Immunol Inflamm 2000; 8(1):49-57.

Lehrich R, Rocha P, Reinsmoen N et al.(2005) Intravenous immunoglobulin and plasmapheresis in acute humoral rejection: Experience in renal allograft transplantation. Hum Immunol 2005; 66(4):350-8.

Levy Y, Sherer Y, Ahmed A, et al.(1999) A study of 20 SLE patients with intravenous immunoglobulin clinical and serologic response. Lupus 1999; 8(9):705-12.

Liew W, Powell C, Sloan S, et al.(2014) Comparison of plasmapheresis and intravenous immunoglobulin as maintenance therapies for juvenile myasthenia gravis. JAMA Neurol. 2014 May;71(5):575-80.

Linner A, Darenberg J, Sjolin J, et al.(2014) Clinical efficacy of polyspecific intravenous immunoglobulin therapy in patients with streptococcal toxic shock syndrome: a comparative observational study. Clin Infect Dis. Sep 15 2014;59(6):851-857. PMID 24928291

Lockwood CM.(1996) New treatment strategies for systemic vasculitis: the role of intravenous immune globulin therapy. Clin Exp Immunol 1996; 104(sup 1):77-82.

Louis D, More K, Oberoi S, et al.(2014) Intravenous immunoglobulin in isoimmune haemolytic disease of newborn: an updated systematic review and meta-analysis. Arch Dis Child Fetal Neonatal Ed. Jul 2014;99(4):F325-331. PMID 24514437

Ludwig H, Zojer N.(2007) Supportive care in multiple myeloma. Best Pract Res Clin Haematol, 2007; 20(4):817-35.

Luke PP, Scantlebury VP, Jordan ML, et al.(2001) Reversal of steroid- and anti-lymphocyte antibody resistant rejection using intravenous immunoglobulin (IVIG) in renal transplant recipients. Transplantation 2001; 72(3):419-22.

Macintyre EA, Linch DC, Macey MG, et al.(1985) Successful response to intravenous immunoglobulin in autoimmune haemolytic anaemia. Br J Haematol. Jun 1985;60(2):387-388. PMID 4005186

Marie I, Hachulla E, Levesque H, et al.(1999) Intravenous immunoglobulins as treatment of life threatening esophageal involvement in polymyositis and dermatomyositis. J Rheumatol 1999; 26:2706-9.

McNamara DM, Holubkov R, Starling RC, et al.(2001) Controlled trial of intravenous immune globulin in recent-onset dilated cardiomyopathy. Circ 2001; 103(18):2254-9.

Medicare coverage policy #CAG-00109N. http://cms.hhs.gov/ coverage/8b3-kkk.asp; 2002.

Meriggioli MN.(2007) IVIG in myasthenia gravis. Getting enough "bang for the buck". Neurology, 2007; 68:803-4.

Montgomery R, Zachary A.(2004) Transplanting patients with a positive donor-specific crossmatch: a single center’s perspective. Pediatr Transplant 2004; 8(6):535-42.

Mouthon L, Lortholary O.(2003) Intravenous immunoglobulins in infectious diseases: where do we stand now? Clin Microbial Infect, 2003; 9:333-8.

Muller-Felber W, Ansevin CF, et al.(1999) Immunosuppressive treatment of rippling muscles in patients with myasthenia gravis. Neuromuscul Disord, 1999; 9:604-7.

National Comprehensive Cancer Network (NCCN).(2021) Management of Immunotherapy-Related Toxicities v.3.2021. Accessed at https://www.nccn.org/professionals/drug_compendium/content/. Accessed September 15, 2021.

Newburger JW, Takahashi M, Beiser AS, et al.(1991) A single intravenous infusion of gamma globulin compared with four infusions in the treatment of acute Kawasaki syndrome. NEJM 1991; 324(23):1633-9.

Norrby-Teglund A, Muller MP, McGeer A, et al.(2005) Successful management of severe group A streptococcal soft tissue infections using an aggressive medical regimen including intravenous polyspecific immunoglobulin together with a conservative surgical approach. Scand J Infect Dis. 2005;37(3):166-172. PMID 15849047

Noseworthy JH, O’Brien PC, Petterson TM, et al.(2001) A randomized trial of intravenous immunoglobulin in inflammatory demyelinating optic neuritis. Neurology 2001; 56(11):1514-22.

Noseworthy JH, O’Brien PC, Weinshenker BG, et al.(2000) IV immunoglobulin does not reverse established weakness in MS. Neurology 2000; 55(8):1135-43.

Nucci M, Anaissie E.(2009) Infections in patients with multiple myeloma in the era of high-dose therapy and novel agents. Clin Infect Dis, 2009; 40:1211-25.

Patwa HS, Chaudhry V, et al.(2012) Evidence-based guideline: Intravenous immunoglobulin in the treatment of neuromusculr disorders: report the the Therapeutics & Technology Assessment Subcommittee of the American Academy of Neurology. Neurology, 2012; 78:1009-15.

Plasma Exchange/Sandoglobulin Guillain-Barre Syndrome Trial Group.(1997) Randomized trial of plasma exchange, intravenous immunoglobulin, and combined treatments in Guillain-Barre syndrome. Lancet 1997; 349(9047):225-30.

Qureshi AI, Choudhry MA, Akbar MS, et al.(1999) Plasma exchange versus intravenous immunoglobulin treatment in myasthenic crisis. Neurology 1999; 52(3):629-32.

Raanani P, Gafter-Gvili A, et al.(2008) Immunoglobulin prophylaxis in hematopoietic stem cell transplantation: systematic review and meta-analysis. J Clin Oncol, 2008; 27:770-81.

Ronager J, Ravnborg M, Hermansen I, et al.(2001) Immunoglobulin treatment versus plasma exchange in patients with chronic moderate to severe myasthenia gravis. Artif Organs 2001; 25(12):967-73.

Rosenbaum JT, George RK, Gordon C.(1999) The treatment of refractory uveitis with intravenous immunoglobulin. Am J Ophthal 1999; 127(5):545-9.

Sakthiswary R, D'Cruz D.(2014) Intravenous immunoglobulin in the therapeutic armamentarium of systemic lupus erythematosus: a systematic review and meta-analysis. Medicine (Baltimore). Oct 2014;93(16):e86. PMID 25310743

Salmun LM, Barlan I, Wolf HM, et al.(1999) Effect of intravenous immunoglobulin on steroid consumption in patients with severe asthma: a double-blind, placebo-controlled, randomized trial. J Allergy Clin Immunol 1999; 103(5 Pt 1):810-5.

Scott JR.(2002) Immunotherapy for recurrent miscarriage. The Cochrane Library; Issue 3; Oxford: Update Software, 2002.

Shah SS, Hall M, Srivastava R, et al.(2009) Intravenous immunoglobulin in children with streptococcal toxic shock syndrome. Clin Infect Dis. Nov 01 2009;49(9):1369-1376. PMID 19788359

Sharma KR, Cross J, Ayyar DR, et al.(2002) Diabetic demyelinating polyneuropathy responsive to intravenous immunoglobulin therapy. Arch Neurol 2002; 59(5):751-7.

Sigra S, Hesselmark E, Bejerot S.(2018) Treatment of PANDAS and PANS: a systematic review. Neurosci Biobehav Rev 2018; 86:51.

Skeie GO, Apostolski S, et al.(2006) Guidelines for the treatment of autoimmune neuromuscular transmission disorders. Eur J Neurology, 2006; 13:691-9.

Sokos DR, Berger M, Lazarus HM.(2002) Intravenous immunoglobulin: appropriate indications and uses in hematopoietic stem cell transplantation. Biol Blood Bone Marrow Transplant, 2002; 8:117-30.

The Intravenous Immunoglobulin Collaborative Study Group.(1992) Prophylactic intravenous administration of standard immune globulin as compared with core-lipopolysaccharide immune globulin in patients at high risk of post-surgical infection. NEJM 1992; 327:234-240.

Van den Berg LH, Kerkhoff H, Oey PL, et al.(1995) Treatment of multifocal motor neuropathy with high dose intravenous immunoglobulins: a double blind, placebo controlled study. J Neurol Neurosurg Psychiatry 59:248–252.

van der Meche FG, Schmitz PI.(1992) A randomized trial comparing intravenous immune globulin and plasma exchange in Guillain-Barre syndrome. NEJM 1992; 326(17):1123-9.

van der Meche FGA, Schmitz PIM, et al.(1992) A randomized trial comparing intravenous immune globulin and plasma exchange in Guillain-Barre syndrome. NEJM 1992; 326:1123-1129.

van Schaik IN, Bril V, van Geloven N, Hartung HP, Lewis RA, Sobue G, Lawo JP, Praus M, Mielke O, Durn BL, Cornblath DR, Merkies ISJ; PATH study group.(2018) Subcutaneous immunoglobulin for maintenance treatment in chronic inflammatory demyelinating polyneuropathy (PATH): a randomised, double-blind, placebo-controlled, phase 3 trial. van Schaik IN, Bril V, van Geloven N, Hartung HP, Lewis RA, Sobue G, Lawo JP, Praus M, Mielke O, Durn BL, Cornblath DR, Merkies ISJ; PATH study group. Subcutaneous immunoglobulin for maintenance treatment in chronic inflammatory demyelinating polyneuropathy (PATH): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol. 2018 Jan;17(1):35-46. doi: 10.1016/S1474-4422(17)30378-2. Epub 2017 Nov 6. Erratum in: Lancet Neurol. 2018 Jan;17 (1):26. Erratum in: Lancet Neurol. 2018 Aug;17(8):661.

Vollmer-Conna U, Hickie I, Hadzi-Pavlovic D, et al.(1997) Intravenous immunoglobulin is ineffective in the treatment of patients with chronic fatigue syndrome. Am J Med 1997; 103(1):38-43.

Voss LM, Wilson NJ, Neutze JM, et al.(2001) Intravenous immunoglobulin in acute rheumatic fever: a randomized controlled trial. Circ 2001; 103(3):401-6.

Walter MC, Lochmuller H, Toepfer M, et al.(2000) High-dose immunoglobulin therapy in sporadic inclusion body myositis: a double-blind, placebo-controlled study. J Neurol 2000; 247(1):22-8.

Williams KA, Swedo SE, Farmer CA, et al.(2016) Randomized, Controlled Trial of Intravenous Immunoglobulin for Pediatric Autoimmune Neuropsychiatric Disorders Associated With Streptococcal Infections. J Am Acad Child Adolesc Psychiatry 2016; 55:860.

Wolfe GF, Barohn RJ, et al.(2002) Randomized, controlled trial of intravenous immunoglobulin in myasthenia gravis. Muscle Nerve, 2002; 26:549-52.

Zinman L, Ng E, Bril V.(2007) IV immunoglobulin in patients with myasthenia gravis. A randomized controlled trial. Neurology, 2007; 68:837-41.

Zuliani L, Nosadini M, Gastaldi M, et. al.(2019) Management of antibody-mediated autoimmune encephalitis in adults and children: literature review and consensus-based practical recommendations. Neurological Sciences 2019. 40:2017-2030. https://doi.org/10.1007/s10072-019-03930-3. .


Group specific policy will supersede this policy when applicable. This policy does not apply to the Wal-Mart Associates Group Health Plan participants or to the Tyson Group Health Plan participants.
CPT Codes Copyright © 2024 American Medical Association.