Criteria for the clinical use of Intravenous Immunoglobulin in Australia - Second Edition

Conditions for which IVIg has an emerging therapeutic role

This chapter comprises conditions for which the therapeutic role of intravenous immunoglobulin (IVIg) is either emerging or uncertain. There is clinical support for IVIg use in selected patients, although the quality of evidence supporting use is variable. For many conditions, IVIg use is considered only as second-line therapy when standard therapies have proven ineffective, have become intolerable, or are contraindicated.

Many of the conditions are rare and as a result the evidence of benefit is often patchy and inconclusive. Others are more prevalent, yet the evidence of benefit is either conflicting or uncertain, requiring more research. For some conditions, the use of IVIg may represent a relatively new direction in their management and evidence of benefit is still emerging.

One exception to this is immune thrombocytopenic purpura (ITP) in children. While the effectiveness of IVIg is not disputed, clinical experts advise that most children with ITP do not require IVIg. Its use in childhood ITP is considered only for the relatively small proportion of children who do not remit spontaneously or respond to standard care.

Another example is kidney transplantation. While evidence from good quality studies exists for the effectiveness of IVIg in antibody-mediated rejection, evidence for its use in cellular rejection is still emerging.

The information provided is not intended to be a definitive reference on any of the conditions, or to be used by clinicians for actual diagnosis or management. Expert clinical opinion about treatment regimens should always be sought. In particular, dose and schedule information is provided as a guide only. The aim in each case is to find the minimal effective dose and optimise the treatment of each individual.

Table 4 Conditions for which IVIg has an emerging therapeutic role as immunoglobulin replacement therapy

Conditions for which IVIg has an emerging therapeutic role as immunoglobulin replacement therapy
Condition Evidence level Page
Secondary hypogammaglobulinaemia (including iatrogenic immunodeficiency) 4b 106
Specific antibody deficiency 4a 110

Table 5 Conditions for which IVIg has an emerging therapeutic role as immunomodulation therapy

Conditions for which IVIg has an emerging therapeutic role as immunomodulation therapy
Condition Evidence level Page
Acute disseminated encephalomyelitis 2a 115
ANCA-positive systemic necrotising vasculitis 2a 119
Autoimmune haemolytic anaemia 4a 123
Bullous pemphigoid 4a 126
Cicatricial pemphigoid 2a 129
Evans syndrome - autoimmune haemolytic anaemia with immune thrombocytopenia 4a 133
Foeto-maternal/neonatal alloimmune thrombocytopenia (FMAIT/NAIT) 4a 136
Haemophagocytic syndrome 4a 141
Idiopathic (autoimmune) thrombocytopenia purpura (ITP) in children 1 144
IgM paraproteinaemic neuropathy 2c 148
Kidney transplantation 1 152
Multiple sclerosis 2a 158
Opsoclonus-myoclonus ataxia 4a 164
Pemphigus foliaceus 4a 166
Pemphigus vulgaris 2a 168
Post-transfusion purpura 4a 171
Toxic epidermal necrolysis/Stevens–Johnson syndrome (TEN/SJS) 4a 173
Toxic shock syndrome 4a 177
Medical condition Secondary hypogammaglobulinaemia (including iatrogenic immunodeficiency)
Indication for IVIg use

Replacement therapy for life-threatening infection due to hypogammaglobulinaemia related to other diseases or medical therapy.

Note: The following secondary causes of hypogammaglobulinaemia are considered elsewhere:

  1. Acquired hypogammaglobulinaemia secondary to haematological malignancies or stem cell transplantation (see page 48)
  2. HIV in children (see page 185)
  3. Solid organ transplantation (see page 208)
Level of evidence No included studies (Category 4b).
Description and diagnostic criteria

Recurrent and/or severe bacterial infections may arise from hypogammaglobulinaemia of diverse causes. Hypogammaglobulinaemia may arise from protein losing states, malnutrition and medical immunosuppression. In most cases, successful management of the underlying condition will reverse the immunodeficiency, restoring immunocompetence. In some cases, recurrent or severe infection may arise from secondary immunodeficiency where the underlying cause cannot be reversed, or where there are unwanted effects of removing or reducing immunosuppressive therapy. New immunosuppressive regimens such as monoclonal B-cell depletion with Rituximab or similar agents do not generally induce hypogammaglobulinaemia at standard doses.

However, repeated cycles of B-cell depletion in combination with other agents used to treat life-threatening immune-mediated diseases may increase rates of infection related to hypogammaglobulinaemia.

Qualifying criteria for IVIg therapy

Hypogammaglobulinaemia secondary to underlying disease or medical therapy (including haemopoietic stem cell transplantation [HCST]) with all the following:

  1. Serum IgG less than the lower limit of the reference range on two separate occasions;

AND

  1. Underlying cause of hypogammaglobulinaemia cannot be reversed or reversal is contraindicated;

AND

  1. At least one of the following:
    1. One invasive or life-threatening bacterial infection (e.g. pneumonia, meningitis, sepsis) in the previous year; or
    2. Clinically active bronchiectasis confirmed by radiology.
Exclusion criteria for IVIg therapy

Reversible underlying cause of hypogammaglobulinaemia.

The following conditions should not be approved under this indication:

  1. Acquired hypogammaglobulinaemia secondary to haematological malignancies or stem cell transplantation (see page 48);
  2. HIV in children (see page 185); or
  3. Transplantation related immunomodulation (solid organ transplantation; see page 208).
Review criteria for assessing the effectiveness of IVIg use

Six-monthly review to assess clinical benefit.

Cessation of IVIg should be considered, at least after each 12 months of therapy, extended as required to enable cessation of therapy in September/October, with repeat clinical and/or immunological evaluation before re-commencement of therapy.

Written confirmation from the treating physician that:

  • an annual review has been undertaken;
  • the patient had demonstrated clinical benefit;
  • a trial period of cessation of IVIg for the purpose of immunological evaluation is medically contraindicated on safety grounds.

In principle, IVIg should be continued or renewed only if there is a demonstrated clinical benefit.

Dose

Maintenance dose: 0.4 g/kg every four weeks, modified to achieve IgG trough level of at least the lower limit of the age-specific serum IgG reference range.

Loading dose: One additional dose of 0.4 g/kg in the first month of therapy is permitted if the serum IgG level is markedly reduced.

Chronic suppurative lung disease: Dosing to achieve IgG trough level of up to 9 g/L is permitted if chronic suppurative lung disease is not adequately controlled at an IgG trough level at the lower limit of the age- specific serum IgG reference range.

Subcutaneous administration of immunoglobulins is a suitable alternative to IVIg in this disease.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Orange, JS, Hossny, EM, Weiler, CR, et al 2006, ’Use of intravenous immunoglobulin in human disease: A review of primary evidence by members of the Primary Immunodeficiency Committee of the American Academy of Allergy, Asthma and Immunology’, Journal of Allergy and Clinical Immunology, vol. 117, no. 4, pp. S525–53.

Medical condition Specific antibody deficiency
Indication for IVIg use Prevention of infections in individuals with frequent infections who have demonstrated failure to mount protective IgG antibody responses to vaccine antigen challenge despite normal total serum IgG levels.
Level of evidence Small case studies only, insufficient data (Category 4a).
Description and diagnostic criteria

The term ‘specific antibody deficiency’ describes failure of specific antibody response to an antigen challenge, and is most often used in the more restrictive sense of applying to polysaccharide antibody responses only.

Patients who have normal total IgG levels but impaired production of specific antibodies, including those with isolated deficient responses to numerous polysaccharide antigens after vaccination, can present a diagnostic challenge. IgG replacement therapy should be provided when there is well-documented severe polysaccharide non-responsiveness and evidence of recurrent infections with a documented requirement for antibiotic therapy and ongoing recurrent infections despite antibiotic prophylaxis (Orange et al 2006).

It is now generally agreed that IgG subclass level estimation in serum is relatively poorly predictive of infectious risk and is of limited value in the definition of those patients most likely to benefit from IVIg therapy.

Further research investigating clinical and laboratory features of this disorder is required.

Qualifying criteria for IVIg therapy

To access IVIg for a period of 12 months, the following qualifying criteria must be met:

  1. A clinical immunologist must be consulted to confirm the diagnosis;

AND

  1. Frequent bacterial infections despite oral antibiotic therapy consistent with best practice recommendations;

AND

  1. Documented failure of serum antibody response to unconjugated pneumococcal or protein vaccine challenge.
Exclusion criteria for IVIg therapy
  1. Isolated IgG subclass deficiency in the absence of evidence of specific antibody deficiency.
  2. Low total IgG. This should be considered under primary or secondary immunodeficiency.
Review criteria for assessing the effectiveness of IVIg use

Natural history of specific antibody deficiency remains poorly defined, although antibody production will improve for many patients over time, particularly children.

To be eligible to receive IVIg for a further 12 months, the following is required:

Written confirmation from the treating clinical immunologist that:

  • an annual review has been undertaken;
  • the patient had demonstrated clinical benefit;
  • a trial period of cessation of IVIg for the purpose of immunological evaluation is medically contraindicated on safety grounds.

Cessation of IVIg should be considered, at least after each 12 months of therapy extended as required to enable cessation of therapy in September/October.

This should particularly be considered in patients who do not have suppurative lung disease or bronchiectasis. An immunoglobulin washout period of four to six months is necessary to enable an accurate assessment. Prophylactic antibiotics may be considered to cover the period of IVIg cessation.

Patients may qualify for further IVIg therapy:

  • under other immunodeficiency criteria (e.g. common variable immunodeficiency [CVID]) depending on the results of subsequent immune evaluation; or
  • rarely under specific antibody deficiency following re-emergence of severe significant infection requiring hospitalisation.

In principle, IVIg should only be continued or renewed if there is a demonstrated clinical benefit.

Note that re-vaccination with pneumococcal polysaccharide vaccine is not recommended because of safety concerns, and the potential for specific hyporesponsiveness induced by repeated vaccination (O’Brien et al 2007).

IgG subclass deficiency
  1. New patients

IVIg is not funded for new patients diagnosed with IgG subclass deficiency.

  1. Patients who were receiving IVIg for IgG subclass deficiency before initial publication of the Criteria (December 2007)
  • Without clinically active bronchiectasis or suppurative lung disease:

These patients should have ceased IVIg and had their immunological status re-evaluated. Patients with a confirmed IgG deficiency have become eligible under another indication (e.g. primary immunodeficiency with antibody deficiency). Patients without a confirmed IgG deficiency have ceased IVIg therapy.

  • With clinically active bronchiectasis or suppurative lung disease over the previous 12 months: To be eligible to receive IVIg for a further 12 months, the following is required:
  1. Written confirmation from the treating clinical immunologist that :
  • an annual review has been undertaken;
  • the patient has demonstrated clinical benefit; and
  • a trial period of cessation of IVIg for the purpose of immunological evaluation is medically contraindicated on safety grounds.

AND

  1. Written confirmation by a second physician that cessation of IVIg for the purpose of immunological evaluation is medically contraindicated on safety grounds.

Cessation of IVIg should be considered, at least after each 12 months of therapy extended as required to enable cessation of therapy in September/October.

NOTE: The above criteria for initial and ongoing access to IVIg funded by all governments under the National Blood Arrangements will be reviewed in light of emerging evidence at the next review of the Criteria.

Dose

Maintenance dose: 0.4 g/kg every 4 weeks.

Loading dose: not approved.

Subcutaneous administration of immunoglobulins (SCIg) is a suitable alternative to IVIg in this setting.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Bheng, YK, Decker, PA, O’Byrne, MM, et al 2006, ‘Clinical and Laboratory characteristics of 75 patients with specific Polysaccharide antibody deficiency syndrome’, Annals of Allergy, Asthma and Immunology, vol. 97, no. 3, pp. 306–11.

Bonilla, FA, Bernstein, L, Khan, DA, et al 2005, ‘Practice parameter for the diagnosis and management of primary immunodeficiency’, Annals of Allergy, Asthma and Immunology, vol. 94, no. 5, suppl. 1, pp. S1–63.

O’Brien, KL, Hochman, M & Goldblatt, D 2007, ‘Combined schedules of pneumococcal conjugate and polysaccharide vaccines: is hyporesponsiveness an issue?’ Lancet Infectious Diseases, vol. 7, no. 9, pp. 597–606.

Orange, JS, Hossny, EM, Weiler, CR, et al 2006, ‘Use of intravenous immunoglobulin in human disease: A review of primary evidence by members of the Primary Immunodeficiency Committee of the American Academy of Allergy, Asthma and Immunology’, Journal of Allergy and Clinical Immunology, vol. 117, no. 4, pp. S525–53.

Medical condition Acute disseminated encephalomyelitis (ADEM)
Indication for IVIg use
  1. ADEM unresponsive to steroid therapy or where steroids are contraindicated (e.g. suspicion of CNS infection).
  2. Recurrent or multiphasic ADEM unresponsive to steroid therapy or where steroid therapy has become intolerable or is contraindicated.
Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria

ADEM is a monophasic inflammatory condition of the central nervous system that usually presents in children and young adults. It typically occurs following a viral prodrome with multifocal neurological disturbance and altered conscious state. ADEM usually follows a monophasic course, but patients may experience recurrence of the initial symptom complex (recurrent ADEM) or a second episode of ADEM (multiphasic ADEM). The majority make a full recovery.

ADEM is thought to have an autoimmune basis. Pathologic similarities to experimental allergic encephalomyelitis (EAE), an animal model of inflammatory demyelination, support this theory. It is postulated that a common antigen shared by an infectious agent and a myelin epitope results in an autoimmune response.

Patients show multiple demyelinating lesions on magnetic resonance imaging (MRI) in the deep and subcortical white matter. The differential diagnosis includes other inflammatory demyelinating disorders, such as multiple sclerosis, optic neuritis and transverse myelitis.

High-dose corticosteroids are first-line treatment of ADEM. IVIg has been used for patients who fail to respond to steroid therapy or in patients where steroids are contraindicated. Most patients with ADEM recover completely over a period of six weeks from onset.

There is no biological marker for ADEM. Diagnosis is by clinical recognition of the multifocal neurological disturbance and altered conscious state, with the typical MRI findings of demyelination.

Justification for evidence category On review of multiple case series of IVIg use for paediatric ADEM found that children with monophasic ADEM completely recovered after administration of IVIg or IVIg plus corticosteroids. In recurrent ADEM, children either completely recovered after IVIg, or showed improvement. Adults with monophasic or recurrent ADEM recovered after treatment with IVIg.
Qualifying criteria for IVIg therapy
  1. ADEM unresponsive to steroid therapy or where steroids are contraindicated (e.g. suspicion of CNS infection).

Note: Assessment by a neurologist is recommended, but not mandatory.

OR

  1. Recurrent or multiphasic ADEM unresponsive to steroid therapy, or where steroid therapy has become intolerable or is contraindicated, with assessment by a neurologist mandatory.
Review criteria for assessing the effectiveness of IVIg use

Objective evidence of improvement in relapse rate in comparison to pre-treatment levels.

Six-monthly review by a neurologist is required for recurrent or multiphasic ADEM.

Dose

Induction: 2 g/kg in 2 to 5 divided doses.

Maintenance dose: For recurrent or multiphasic
ADEM only: 0.4–2 g/kg, 4–6 weekly.

Aim for the minimum dose to maintain optimal functional status and prevent relapses.

In recurrent or multiphasic ADEM, assessment by a neurologist is mandatory.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Andersen, JB, Rasmussen, LH, Herning, M, et al 2001, ‘Dramatic improvement of severe acute disseminated encephalomyelitis after treatment with intravenous immunoglobulin in a three-year- old boy’, Developmental Medicine & Child Neurology, vol. 43, no. 2, pp. 136–8.

Feasby, T, Banwell, B, Benstead, T, et al 2007, ‘Guidelines on the use of intravenous immune globulin for neurologic conditions’, Transfusion Medicine Reviews, vol. 2, no. 2, suppl. 1, pp. S57–107.

Finsterer, J, Grass, R, Stollberger, C, et al 1998, ‘Immunoglobulins in acute, parainfectious, disseminated encephalomyelitis’, Clinical Neuropharmacology, vol. 21, pp. 258–61.

Hahn, JS, Siegler, DJ & Enzmann, D 1996, ‘Intravenous gammaglobulin therapy in recurrent acute disseminated encephalomyelitis’, Neurology, vol. 46, no. 4, pp. 1173–4.

Kleiman, M & Brunquell, P 1995, ‘Acute disseminated encephalomyelitis: response to intravenous immunoglobulin?’ Journal of Child Neurology, vol. 10, no. 6, pp. 481–3.

Marchioni, E, Marinou-Aktipi, K, Uggetti, C, et al 2002, ‘Effectiveness of intravenous immunoglobulin treatment in adult patients with steroid-resistant monophasic or recurrent acute disseminated encephalomyelitis’, Journal of Neurology, vol. 249, no. 1, pp. 100–4.

Mariotti, P, Batocchi, AP, Colosimo, C, et al 2003, ‘Multiphasic demyelinating disease involving central and peripheral nervous system in a child’, Neurology, vol. 60, no. 2, pp. 348–9.

Nishikawa, M, Ichiyama, T, Hayashi, T, et al 1999, ‘Intravenous immunoglobulin therapy in acute disseminated encephalomyelitis’, Paediatric Neurology, vol. 21, no. 2, pp. 583–6.

Pittock, SJ, Keir, G, Alexander, M, et al 2001, ‘Rapid clinical and CSF response to intravenous gamma globulin in acute disseminated encephalomyelitis’, European Journal of Neurology, vol. 8, no. 6, p. 725.

Pradhan, S, Gupta, RP, Shashank, S, et al 1999, ‘Intravenous immunoglobulin therapy in acute disseminated encephalomyelitis’, Journal of the Neurological Sciences, vol. 165, no. 1, pp. 56–61.

Revel-Vilk, S, Hurvitz, H, Klar, A, et al 2000, ‘Recurrent acute disseminated encephalomyelitis associated with acute cytomegalovirus and Epstein-Barr virus infection’, Journal of Child Neurology, vol. 15, no. 6, pp. 421–4.

Sahlas, DJ, Miller, SP, Guerin, M, et al 2000, ‘Treatment of acute disseminated encephalomyelitis with intravenous immunoglobulin’, Neurology, vol. 54, no. 6, pp. 1370–2.

Medical condition Anti-neutrophil cytoplasmic antibody (ANCA) [Proteinase 3 (PR3) or myeloperoxidase (MPO)]- positive systemic necrotising vasculitis
Indication for IVIg use Control of vasculitic activity in rare cases of ANCA-positive systemic necrotising vasculitis failing to respond to corticosteroids and cytotoxic immunosuppression.
Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria

ANCA associated systemic necrotising vasculitides are life-threatening immune-mediated inflammatory diseases comprising one of four clinical syndromes:

  1. Wegener granulomatosis;
  2. microscopic polyangiitis;
  3. Churg–Strauss syndrome; and
  4. ANCA (PR3 or MPO)-positive idiopathic rapidly progressive glomerulonephritis.

In these cases the ANCA specificity is directed against the neutrophil cytoplasmic antigens PR3 and MPO. ANCA that lack MPO or PR3 specificity tend to be non-specific. Biopsy of affected tissue is required to establish the diagnosis.

Standard combinations of corticosteroids and cytotoxic immunosuppression are generally effective at controlling disease, but relapses are common. IVIg has a limited role as one of several therapeutic options in relapsing disease.

Justification for evidence category The Biotext (2004) review found one randomised trial of 34 patients and one case series of 7 patients with ANCA-associated systemic vasculitis (AASV). Different AASVs were represented in the studies. The Biotext (2004) review concluded that there is possible benefit in the treatment of AASV with IVIg if disease activity persists after standard therapy.
Qualifying criteria for IVIg therapy

MPO or PR3 ANCA-positive systemic necrotising vasculitis with both of the following:

  1. Current (or within the previous six months) standard cytotoxic immunosuppressive ANCA-vasculitis regimens;

AND

  1. Persisent active disease.
Exclusion criteria for IVIg therapy Initial therapy
Review criteria for assessing the effectiveness of IVIg use
  • Six-month review assessing evidence of clinical benefit.
  • Reduction in the Birmingham vasculitis activity score of more than 50% after three months.
  • Erythrocyte sedimentation rate and C-reactive protein concentration.
  • ANCA titre.
Dose

2 g/kg in single or divided doses.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Biotext 2004, ‘Summary data on conditions and papers’, in A systematic literature review and report on the efficacy of intravenous immunoglobulin therapy and its risks, commissioned by the National Blood Authority on behalf of all Australian Governments, pp. 248–50. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf

Foster, R, Rosenthal, E, Marques, S, et al 2006, ‘Primary systemic vasculitis: treatment of difficult cases’, Lupus, vol. 15, no. 3, pp. 143–7.

Jayne, DR & Rasmussen, N 1997, ‘Treatment of antineutrophil cytoplasm autoantibody-associated systemic vasculitis: initiatives of the European Community Systemic Vasculitis Clinical Trials Study Group’, Mayo Clinic Proceedings, vol. 72, no. 8, pp. 737–47.

Jayne, DR, Chapel, H, Adu, D, et al 2000, ‘Intravenous immunoglobulin for ANCA-associated systemic vasculitis with persistent disease activity’, Quarterly Journal of Medicine, vol. 93, no. 7, pp. 433–9.

Jayne, DR, Davies, MJ, Fox, CJ, et al 1991, ‘Treatment of systemic vasculitis with pooled intravenous immunoglobulin’, Lancet, vol. 337, no. 8750, pp. 1137–9.

Jennette, JC, Falk, RJ, Andrassy, K, et al 2004, ‘Nomenclature of systemic vasculitides: proposal of an international consensus conference’, Arthritis & Rheumatism, vol. 37, no. 2, pp. 187–92 (Chapel Hill Consensus criteria).

Orange, JS, Hossny, EM, Weiler, CR, et al 2006, ‘Use of intravenous immunoglobulin in human disease: A review of primary evidence by members of the Primary Immunodeficiency Committee of the American Academy of Allergy, Asthma and Immunology’, Journal of Allergy and Clinical Immunology, vol. 117, no. 4, pp. S525–53.

Medical condition Autoimmune haemolytic anaemia (AIHA)
Indication for IVIg use To reduce haemolysis in patients not responding to corticosteroid therapy.
Level of evidence Small case studies only; insufficient data (Category 4a).
Description and diagnostic criteria

AIHA is a rare but serious autoimmune disease in which an individual’s antibodies recognise antigens on their own red blood cells. AIHA presents as an acute or chronic anaemia characterised by the occurrence of biochemical parameters of red cell destruction associated with a positive direct antiglobulin test indicating the presence of antibodies and/or complement on the red cell surface. It may be secondary to a number of underlying disorders or drugs.

Investigations

A full blood count will confirm the presence of anaemia. A peripheral blood smear may reveal evidence of spherocytes along with polychromasia due to reticulocytosis. A direct antiglobulin test is usually positive, the serum lactate dehydrogenase is raised, and there is a reduction in serum haptoglobin.

Prognosis

The prognosis of AIHA is good in most cases although severe refractory AIHA can cause cardio-respiratory problems because of severe anaemia, especially in adults.

Standard therapy

Corticosteroid administration is the cornerstone of therapy. For those with relapsing disease, splenectomy and immunosuppression are second line treatments while anti-CD20 antibodies have shown promise in individual cases of refractory disease.

Justification for evidence category An analysis of 73 patients with AIHA in 1993 based on three pilot studies and a literature review showed a 40% response to IVIg given together with corticosteroids. A lower initial haemoglobin concentration and hepatomegaly were positive correlates of response. Several small case series have suggested a benefit for IVIg in AIHA associated with lymphoproliferative diseases, especially CLL. On the basis of these findings, IVIg is not supported as standard therapy for AIHA, only in cases refractory to conventional corticosteroid therapy, as a temporising measure before splenectomy or as maintenance therapy where splenectomy or immunosuppression are not appropriate.
Qualifying criteria for IVIg therapy
  1. Symptomatic or severe AIHA (Hb <60 g/L, except patients with co-morbidities) refractory to conventional therapy with corticosteroids;

OR

  1. As a temporising measure before splenectomy;

OR

  1. As initial and maintenance therapy in AIHA in patients unsuitable for splenectomy or immunosuppression.
Exclusion criteria for IVIg therapy Patients in whom a trial of corticosteroids has not been undertaken.
Review criteria for assessing the effectiveness of IVIg use
  • Resolution of haemolytic anaemia (rising haemoglobin concentrations, falling bilirubin and LDH).
  • Clinical improvement in symptoms and signs.
Dose

Up to 2 g/kg as a single or divided dose.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Besa, EC 1988, ‘Rapid transient reversal of anaemia and long-term effects of maintenance intravenous immunoglobulin for autoimmune haemolytic anaemia in patients with lymphoproliferative disorders’, American Journal of Medicine, vol. 84, no. 4, pp. 691–8.

Flores, G, Cunningham-Rundles, C, Newland, AC, et al 1993, ‘Efficacy of intravenous immunoglobulin in the treatment of autoimmune haemolytic anaemia: results in 73 patients’, American Journal of Hematology, vol. 44, no. 4, pp. 237–42.

Majer, RV & Hyde, RD 1988, ‘High-dose intravenous immunoglobulin in the treatment of autoimmune haemolytic anaemia’, Clinical and Laboratory Haematology, vol. 10, no. 4, pp. 391–5.

Sherer, Y, Levy, Y, Fabbrizzi, F, et al 2000, ‘Treatment of hematologic disorders other than immune thrombocytopenic purpura with intravenous immunoglobulin (IVIg) – report of seven cases and review of the literature’, European Journal of Internal Medicine, vol. 11, pp. 85–8.

Medical condition Bullous pemphigoid (BP)
Indication for IVIg use BP resistant to topical and systemic glucocorticoids and immunosuppressive therapy.
Level of evidence Small case studies only; insufficient data (Category 4a).
Description and diagnostic criteria

BP is a rare disease of elderly people characterised by tense blisters and vesicles with a prominent inflammatory component. The cause is unknown. Lesions result from a failure of basal keratinocytes to adhere to the epidermal basement membrane.

The course of BP is characterised by exacerbations and remissions. Pruritis is a common feature and an increase in pruritis may herald an exacerbation.

In most patients, BP is not a life-threatening disease. The side effects of systemic immunosuppressive therapy need to be managed. In most patients, the disease spontaneously clears within six years and all medication can be stopped. In a small group, the disease recurs after treatment is stopped. Skin infection is the most common complication.

A submission by the Australasian College of Dermatologists recommends IVIg use in BP only in severe cases where improvement with conventional therapy is not readily achieved.

Justification for evidence category The 2003 Harvard consensus statement identified a small study (17 cases) where patients who were on IVIg therapy for at least three months benefited from the therapy. The same article mentioned another small study (15 cases) where patients with BP could not be controlled with high-dose systemic corticosteroids and multiple immunosuppressive agents. IVIg produced prolonged clinical remission sustained after IVIg therapy was discontinued.
Qualifying criteria for IVIg therapy

Moderate to severe disease diagnosed by a dermatologist

AND

  1. Corticosteroids or immunosuppressive agents are contraindicated;

OR

  1. Condition is unresponsive to corticosteroids and immunosuppressive agents;

OR

  1. Presenting with severe side effects of therapy.
Review criteria for assessing the effectiveness of IVIg use
  • Response demonstrated at review at six months. Improvement to be demonstrated for continuation of supply.
  • Reduction in recurrence of disease or relapse.
  • Ability to reduce dose or discontinue other therapies.
  • Improved quality of life.
  • Resolution of blisters and healing of affected skin.
  • Resolution of pruritis.
Dose

Efficacy demonstrated with doses of at least 2 g/kg per monthly treatment cycle.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Ahmed, AR & Dahl MV, for the Consensus Development Group 2003, ‘Consensus statement on the use of intravenous immunoglobulin therapy in the treatment of autoimmune mucocutaneous blistering diseases’, Archives of Dermatology, vol. 139, pp. 1051–9.

Orange, JS, Hossny, EM, Weiler, CR, et al 2006, Use of intravenous immunoglobulin in human disease: A review of primary evidence by members of the Primary Immunodeficiency Committee of the American Academy of Allergy, Asthma and Immunology’, Journal of Allergy and Clinical Immunology, vol. 117, no. 4, pp. S525–53.

Medical condition Cicatricial pemphigoid (CP) or mucous membrane pemphigoid (MMP)
Indication for IVIg use CP resistant to glucocorticoid and immunosuppressive therapy.
Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria

CP or MMP is a rare, acquired subepithelial blistering disease characterised by erosive lesions of mucous membranes and skin. Serious complications may occur due to erosions and scarring. Hoarseness, pain, tissue loss and even upper airway destruction can occur with nasopharyngeal or laryngeal involvement, and oesophageal and urogenital lesions may lead to stenosis or strictures. CP is usually a chronic, progressive disorder.

The aim of long-term treatment is cessation of the self-destructive autoimmune process. Failure to do so results in invariable progression of the disease, culminating in progressive scarring. Permanent remission is usually possible if the disease is diagnosed early and treated sufficiently for one to five years.

For the 70% of patients who have eye involvement, the disease progresses to conjunctival scarring and shrinkage, but may take 10–20 years to reach the end stage of bilateral blindness.

Justification for evidence category

Prolonged clinical remission and reduction in side effects was demonstrated in one small case series (15 cases) of patients with CP/MMP unresponsive to systemic corticosteroids and immuno-suppressive agents or presenting with multiple side effects of therapy (Biotext 2004).

A small non-randomised, non-blinded trial (16 patients) showed significant improvement in the mean time for clinical control, recurrence, disease progression and drug-related side effects among patients receiving IVIg compared to conventional immunosuppressive therapy (Frommer and Madronio 2006).

The (2003) consensus statement from the Harvard Medical School Department of Dermatology identified a study of 10 MMP patients who had progressive ocular involvement and did not respond to corticosteroids or immunosuppressants. IVIg administration as monotherapy arrested the progression and vision was maintained after IVIg was discontinued. The authors cited two other studies of oral pemphigoid in 15 and 7 patients respectively who could not be treated with dapsone; IVIg was compared to immunosuppressants. IVIg led to early and long-term remission and no disease progression.

Qualifying criteria for IVIg therapy

Moderate to severe disease diagnosed by a dermatologist;

AND

  1. Corticosteroids or immunosuppressive agents are contraindicated;

OR

  1. Condition is unresponsive to corticosteroids and immunosuppressive agents;

OR

  1. Presenting with severe side effects of therapy.
Review criteria for assessing the effectiveness of IVIg use
  • Response demonstrated at review at six months. Improvement to be demonstrated for continuation of supply.
  • Disease recurrence or relapse and duration of clinical remission.
  • Ability to reduce dose or discontinue other therapies.
  • Resolution of conjunctival inflammation.
  • Reduction of drug-related side effects.
Dose

Efficacy demonstrated with doses of at least 2 g/kg per monthly treatment cycle.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Ahmed, AR & Dahl, MV, for the Consensus Development Group 2003, ‘Consensus statement on the use of intravenous immunoglobulin therapy in the treatment of autoimmune mucocutaneous blistering diseases’, Archives of Dermatology, vol. 139, pp. 1051–9.

Daoud, YJ & Amin, KG 2006, ‘Comparison of cost of immune globulin intravenous therapy to conventional immunosuppressive therapy in treating patients with autoimmune mucocutaneous blistering diseases’, International Immunopharmacology, vol. 6, no. 4, pp. 600–6.

Letko, E, Miserocchi, E, Daoud, YJ, et al 2004, ‚A nonrandomized comparison of the clinical outcome of ocular involvement in patients with mucous membrane (cicatricial) pemphigoid between conventional immunosuppressive and intravenous immunoglobulin therapies’, Clinical Immunology, vol. 111, no. 3, pp. 303–10.

Orange, JS, Hossny, EM, Weiler, CR, et al 2006, ‚Use of intravenous immunoglobulin in human disease: A review of primary evidence by members of the Primary Immunodeficiency Committee of the American Academy of Allergy, Asthma and Immunology’, Journal of Allergy and Clinical Immunology, vol. 117, no. 4, pp. S525–53.

Medical condition Evans syndrome — autoimmune haemolytic anaemia (AIHA) with immune thrombocytopenia
Indication for IVIg use To reduce platelet destruction and improve haemolysis in patients not responding to corticosteroid therapy.
Level of evidence Small case studies only; insufficient data (Category 4a).
Description and diagnostic criteria

Evans syndrome is a rare but serious autoimmune disease defined by the simultaneous or sequential occurrence of AIHA and immune thrombocytopenia purpura (ITP) without underlying aetiology. As such, it is a diagnosis of exclusion and other disorders, such as collagen vascular diseases, especially systemic lupus erythematosus (SLE) and scleroderma should be ruled out.

The 2005 review by Norton and Roberts provided perspective on diagnosis, clinical features and management.

Justification for evidence category

A 2005 review on the management of Evans syndrome, based on Massachusetts Hospital data and a literature review, showed a transient response in all patients unless IVIg was given every three weeks (Norton and Roberts 2006). The review concluded that the data supported a role for IVIg in first-line therapy. It was not clear whether it was important for steroids to be given at the same time, although this is common practice. A total dose of 2 g/kg in divided doses appeared to be sufficient.

The review also stated that there might be a role for IVIg in preference to steroids in the acute setting in very young children.

Qualifying criteria for IVIg therapy
  1. Refractory to conventional therapy with corticosteroids;

OR

  1. Where corticosteroids are contraindicated;

OR

  1. As a temporising measure before splenectomy.
Exclusion criteria for IVIg therapy Patients in whom a trial of corticosteroids has not been undertaken (providing corticosteroids are not contra-indicated and can be tolerated at the required doses).
Review criteria for assessing the effectiveness of IVIg use
  • Maintenance therapy rarely required.
  • Resolution of haemolytic anaemia.
  • Improvement in platelet count.
  • Clinical improvement in symptoms and signs.
Dose

Up to 2 g/kg in divided dose.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Darabi, K, Abdel-Wahab, O & Dzik, WH 2006, ‘Current usage of intravenous immunoglobulin and the rationale behind it: the Massachusetts General Hospital data and review of the literature’, Transfusion, vol. 46, no. 5, pp. 741–53.

Mathew, P, Chen, G & Wang, W 1997, ‘Evans syndrome: results of a national survey’, Journal of Pediatric Hematology/Oncology, vol. 19, no. 5, pp. 433–7.

Norton, A & Roberts, I 2006, ‘Management of Evans syndrome’, British Journal of Haematology, vol. 132, no. 2, pp. 125–37.

Medical condition

Foeto-maternal/neonatal alloimmune thrombocytopenia (FMAIT/NAIT):

  • antenatal
  • neonatal
Indication for IVIg use Prevention or treatment of foetal or neonatal thrombocytopenia or haemorrhage.
Level of evidence Small case studies only; insufficient data (Category 4a).
Description and diagnostic criteria

FMAIT/NAIT develops because of maternal sensitisation to foetal platelets that possess a paternally inherited antigen. In Caucasians, the antigen is human platelet antigen (HPA) 1a in 80% of cases and HPA-5b in 15%, but other antigens are also implicated. The mother’s antibodies cross the placenta and coat the baby’s platelets, with accelerated platelet clearance leading to thrombocytopenia. This may result in serious and potentially life-threatening bleeding in the foetus or neonate. Pathogenesis is analogous to that of haemolytic disease of the newborn due to red cell antigen-antibody incompatibility.

The aim of management of the thrombocytopenic foetus or neonate is to increase the platelet count.

If foetal blood sampling reveals thrombocytopenia, IVIg may be administered weekly to the mother, with or without steroids, until delivery. Recent studies using IVIg weekly from around 20 weeks gestation, without foetal blood sampling, have shown reduced foetal and neonatal morbidity. This approach may be used for current pregnancies where the condition in a previous pregnancy was not associated with a foetal death or severe haemorrhage. Testing on maternal blood for foetal DNA or early genetic testing of the foetus (for platelet genotype) may predict the need to use IVIg.

Management of this condition is a specialised area and may include administration of HPA-compatible intrauterine and/or neonatal platelet transfusions. Further information regarding specialised platelet support is available from the Blood Service. Random (non-HPA-matched) platelets may be of benefit in the neonatal setting when matched platelets are not available (Kiefel 2006).

Justification for evidence category Evidence from randomised trials (Berkowitz et al 2006, Bussel et al 1996), case series (Kiefel et al 2006, Yinon et al 2006) and a review (Spencer and Burrows 2001) shows that IVIg modulates the course of this condition. A 2004 Cochrane review (Rayment et al 2005) reported on one randomised controlled trial (RCT) comparing IVIg plus dexamethasone with IVIg alone. This RCT was methodologically sound, but too small to detect differences among comparison groups.
Qualifying criteria for IVIg therapy

Clinical suspicion of FMAIT in antenatal or neonatal setting based on clinical and laboratory features, including:

  1. Thrombocytopenia or spontaneous haemorrhage in the foetus;

OR

  1. Thrombocytopenia with or without haemorrhage in the neonate;

OR

  1. Unexplained foetal death in a previous pregnancy and the presence of maternal platelet-specific alloantibodies that are known or suspected to cause this condition (most commonly HPA-1a or HPA-5b).
Review criteria for assessing the effectiveness of IVIg use
  • Foetal or neonatal morbidity and mortality in the context of maternal alloantibodies.
  • Occurrence and severity of thrombocytopenia in the neonate.
  • Maternal HPA-1a antibody level (if assay is available). Note that the strength/titre of maternal antibody level, even if available, is not proven clinically relevant and not able to be compared readily between laboratories at this time.
Dose

Maternal dose: 1 g/kg weekly throughout pregnancy, with starting time tailored to individual risk profile and history if relevant. Other doses and schedules have been used and some studies have used IVIg in conjunction with steroids.

Treatment of the neonate: 1 g/kg. Occasionally more than one dose is required if thrombocytopenia persists.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Berkowitz, RL, Kolb, EA, McFarland, JG, et al 2006, ‚Parallel randomized trials of risk-based therapy for fetal alloimmune thrombocytopenia’, Obstetrics & Gynecology, vol. 107, no. 1, pp. 91–6.

Bussel, JB, Berkowitz, RL, Lynch, L, et al 1996, ‘Antenatal management of alloimmune thrombocytopenia with intravenous immunoglobulin: a randomised trial of the addition of low dose steroid to intravenous gamma globulin’, American Journal of Obstetrics & Gynecology, vol. 74, no. 5, pp. 1414–23.

Kiefel, V, Bassler, D, Kroll, H, et al 2006, ‘Antigen-positive platelet transfusion in neonatal alloimmune thrombocytopenia’, Blood, vol. 107, no. 9, pp. 3761–3.

Rayment, R, Brunskill, SJ, Stanworth, S, et al 2005, ‘Antenatal interventions for fetomaternal alloimmune thrombocytopenia (Cochrane Review)’, in The Cochrane Library, Issue 1, John Wiley & Sons, Ltd, Chichester, UK.

Spencer, JA & Burrows, RF 2001, ‘Feto-maternal alloimmune thrombocytopenia: a literature review and statistical analysis’, Australia New Zealand Journal of Obstetrics and Gynaecology, vol. 41, no. 1, pp. 45–55.

Yinon, Y, Spira, M, Solomon, O, et al 2006, ‘Antenatal noninvasive treatment of patients at risk for alloimmune thrombocytopenia without a history of intracranial hemorrhage’, American Journal of Obstetrics & Gynecology, vol. 195, no. 4, pp. 1153–7.

Medical condition Haemophagocytic syndrome
Indication for IVIg use Management of severe haemophagocytic syndrome not responding to other treatments.
Description and diagnostic criteria Haemophagocytic syndrome is characterised by fever, splenomegaly, jaundice, rash and the pathologic finding of haemophagocytosis (phagocytosis by macrophages of erythrocytes, leukocytes, platelets and their precursors) in bone marrow and other tissues with peripheral blood cytopenias. Haemophagocytic syndrome has been associated with a wide range of infectious, autoimmune, malignant and other disorders (modified from Fisman 2000). Mortality is high.
Level of evidence Small case studies only; insufficient data (Category 4a).
Justification for evidence category No RCTs have been done, although many, mostly small, case series show evidence of benefit.
Qualifying criteria for IVIg therapy

Bone marrow diagnosis or other biopsy evidence of haemophagocytosis in the characteristic clinical setting.

Note: Since other therapies (cytotoxic agents) have major potential side effects, optimal therapy is not yet defined.

Review criteria for assessing the effectiveness of IVIg use

Amelioration of cytopenia(s), hepato/splenomegaly and lymphadenopathy if present.

Survival or death.

Dose

2 g/kg is the most widely published dose.

Emmenegger et al (2001) reported that better outcomes were associated with early administration of IVIg in their small case series (10 patients).

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Arlet, JB, Le, TH, Marinho, A, et al 2006, ‘Reactive haemophagocytic syndrome in adult onset Still’s disease: report of six patients and review of the literature’, Annals of the Rheumatic Diseases, vol. 65, no. 12, pp. 1596–601.

Asci, G, Toz, H, Ozkahya, M, et al 2006, ‘High-dose immunoglobulin therapy in renal transplant recipients with hemophagocytic histiocytic syndrome’, Journal of Nephrology, vol. 19, no. 3, pp. 322–6.

Chen, RL, Lin, KH, Lin, DT, et al 1995, ‘Immunomodulation treatment for childhood virus-associated haemophagocytic lymphohistiocytosis’, British Journal of Haematology, vol. 89, no. 2, pp. 282–90.

Emmenegger, U, Frey, U, Reimers, A, et al 2001, ‘Hyperferritinemia as indicator for intravenous immunoglobulin treatment in reactive macrophage activation syndromes’, American Journal of Haematology, vol. 68, no. 1, pp. 4–10.

Fisman, D, 2000, ‘Hemophagocytic syndromes and infection’, Emerging Infectious Diseases. Available from: www.cdc.gov/ncidod/eid/vol6no6/fisman.htm [cited 7 Dec 2007]

Freeman, B, Rathore, MH, Salman, E, et al 1993, ‘Intravenously administered immune globulin for the treatment of infection- associated hemophagocytic syndrome’, Journal of Pediatrics, vol. 123, no. 3, pp. 479–81.

Ostronoff, M, Ostronoff, F, Coutinho, M, et al 2006, ‘Haemophagocytic syndrome after autologous peripheral blood stem cell transplantation for multiple myeloma; successful treatment with high-dose intravenous immunoglobulin’, Bone Marrow Transplantation, vol. 37, no. 8, pp. 797–8.

Medical condition Idiopathic (autoimmune) thrombocytopenic purpura (ITP) — in children 15 years and younger
Indication for IVIg use ITP with platelet count <30x109/L with significant bleeding.
Level of evidence Clear evidence of benefit (Category 1).
Description and diagnostic criteria

ITP is a reduction in platelet count (thrombocytopenia) resulting from shortened platelet survival due to anti-platelet antibodies. When counts are very low (<30x109/L) bleeding into the skin (purpura) and mucous membranes can occur. Bone marrow morphology is normal. In some cases, there is additional impairment of platelet function related to antibody binding to glycoproteins on the platelet surface. ITP is divided into chronic and acute forms. In children, the acute form is the most common. The disease tends to present abruptly with dramatic evidence of bleeding into the skin (petechiae and purpura) and mucous membranes (gum bleeding, nose bleeds, blood blisters).

Occurrence

Girls and boys are affected equally. In 75% of patients, the episode follows vaccination or a viral infection such as varicella or infectious mononucleosis.

Prognosis

At least 80–90% of children will have spontaneous remission of their disease within 6–12 months. In 5–10% of cases, the disease may become chronic (lasting >6 months). Morbidity and mortality from acute ITP is very low.

Justification for evidence category

Category 1 classification in the Biotext (2004) review was based on four low–moderate quality RCTs.

The Frommer and Madronio (2006) review identified a good-quality systematic review/meta-analysis of RCTs to support the Category 1 classification.

Qualifying criteria for IVIg therapy

Note: While the effectiveness of IVIg is not disputed, clinical experts advise that most children with ITP do not require IVIg therapy; indeed, no treatment at all is required for many children. Corticosteroids are the alternative therapy to IVIg.

Acute ITP

  1. Life-threatening bleeding due to thrombocytopenia;

OR

  1. Thrombocytopenia with platelet count <30x109/L and moderate to severe mucosal and/or cutaneous bleeding.

Chronic ITP

  1. Life-threatening bleeding due to thrombocytopenia;

OR

  1. In responsive patients for treatment of thrombocytopenia (<30x109/L) with moderate to severe bleeding symptoms where other therapeutic options have failed or are contraindicated;

OR

  1. In responsive patients given before surgery to elevate the platelet count to haemostatically safe levels.
Exclusion criteria for IVIg therapy
  1. Platelet count >30x109/L.
  2. Absence of significant bleeding.
Review criteria for assessing the effectiveness of IVIg use
  • Platelet count at 48 hours.
  • Control or resolution of bleeding.
  • Duration of effect.
  • Progression to chronic ITP.
Dose

Acute ITP

Life-threatening bleeding: up to 2 g/kg total dose, generally given as 2 doses of 1 g/kg.

Other indications: 0.5 g/kg given as a single dose, repeated at 24–48 hours if the response is inadequate. A higher total dose of 2 g/kg may be required in 5–10% of cases.

Duration of response to initial dose is typically two to four weeks. A repeat dose may be considered if recurrent symptomatic thrombocytopenia occurs.

Chronic ITP

Life-threatening bleeding: up to 2 g/kg total dose, generally given as 2 doses of 1 g/kg.

Other indications: 0.5 to 1 g/kg at intervals generally > three weekly.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Beck, CE, Nathan, PC, Parkin, PC, et al 2005, ‘Corticosteroids versus intravenous immune globulin for the treatment of acute immune thrombocytopenic purpura in children: a systematic review and meta-analysis of randomized controlled trials’, Journal of Pediatrics, vol. 147, no. 4, pp. 521–7.

Bierling, P & Godeau, B 2005, ‘Intravenous immunoglobulin for autoimmune thrombocytopenic purpura’, Human Immunology, vol. 66, no. 4, pp. 387–94.

Biotext 2004, ‘Summary data on conditions and papers’, in A systematic literature review and report on the efficacy of intravenous immunoglobulin therapy and its risks, commissioned by the National Blood Authority on behalf of all Australian Governments. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf

British Society for Haematology General Haematology Task Force 2003, ‘Guidelines for the investigation and management of idiopathic thrombocytopenic purpura in adults, children and in pregnancy’, British Journal of Haematology, vol. 120, no. 4, pp. 574–96.

Frommer, M & Madronio, C 2006, The use of intravenous immunoglobulin in Australia. A report for the National Blood Authority, Part B: systematic literature review, Sydney Health Projects Group, University of Sydney, Sydney, pp. 11–12.

George, JN, Woolf, SH, Raskob, GE, et al 1996, ‘Idiopathic thrombocytopenic purpura: a practice guideline developed by explicit methods for The American Society of Haematology’, Blood, vol. 88, no. 1, pp. 3–40.

Warrier, I, Bussel, JB, Valdez, L, et al 1997, ‘Safety and efficacy of low dose intravenous immune globulin treatment for infants and children with immune thrombocytopenic purpura’, Journal of Pediatric Hematology/Oncology, vol. 19, no. 3, pp. 197–201.

Medical condition IgM paraproteinaemic neuropathy
Indication for IVIg use Patients with IgM paraproteinaemic neuropathy with functional impairment in whom other therapies have failed or are contraindicated or undesirable.
Level of evidence Conflicting evidence of benefit (Category 2c).
Description and diagnostic criteria

IgM paraproteinaemic neuropathy is a slowly progressive, predominantly sensory neuropathy that may eventually produce disabling motor symptoms. The condition is associated with IgM paraprotein, which is a monoclonal antibody to myelin associated glycoprotein (MAG).

IgM paraproteinaemic neuropathy is the most common subgroup of the monoclonal gammopathy of undetermined significance (MGUS) group.

It is distinguishable from chronic inflammatory demyelinating polyneuropathy (CIDP) by:

  • the presence of tremor;
  • a greater severity of sensory loss, with ataxia and relatively mild or no weakness;
  • damage tends to be permanent and the degree of improvement in IgM paraproteinaemic neuropathy is much smaller than the improvement observed in CIDP patients.

Nerve conduction studies usually show uniform symmetrical conduction slowing with prolonged distal latencies and distal attenuation (distal index is prolonged).

Test for antibodies to neural antigens (MAG or other neural antigens) may be helpful.

Justification for evidence category

The Biotext (2004) review included three low quality studies (one RCT, one case-control and one case- series) with 20 patients. No benefit from treatment with IVIg was demonstrated in the case-control study (Biotext 2004).

The Frommer and Madronio (2006) found a Cochrane systematic review of five medium-quality RCTs with 97 patients of any age with a diagnosis of MGUS. There was inadequate evidence of efficacy of IVIg in anti-myelin-associated glycoprotein paraprotein peripheral neuropathies.

Qualifying criteria for IVIg therapy

Diagnosis by a neurologist of IgM paraproteinaemic neuropathy with:

  1. Functional impairment of activities of daily living;

AND

  1. Other therapies have failed or are contraindicated or undesirable.
Review criteria for assessing the effectiveness of IVIg use

IVIg should be used for three to six months (three to six courses) before determining whether the patient has responded. If there is no benefit after three to six courses, IVIg therapy should be abandoned.

Review

Regular review by neurologist is required; frequency as determined by clinical status of patient.

For stable patients on maintenance treatment review by a neurologist is required at least annually.

Effectiveness

Clinical documentation of effectiveness is necessary for continuation of IVIg therapy.

Effectiveness can be demonstrated by objective findings of either:

  1. Improvement in functional scores (activities of daily living — ADLs) or quantitative muscle scores, or Medical Research Council (MRC) muscle assessment or neuropathy score; or
  2. Stabilisation of disease as defined by stable functional scores (ADLs) or quantitative muscle scores, or MRC muscle assessment or neuropathy score after previous evidence of deterioration in one of these scores.
Dose

Induction: 2 g/kg in 2 to 5 divided doses.

Maintenance: 0.4–1 g/kg, 2 to 6 weekly.

Maintenance treatment only with clear, objective improvement.

Aim for minimum dose to maintain optimal functional status.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Biotext 2004, ‘Summary data on conditions and papers’, in A systematic literature review and report on the efficacy of intravenous immunoglobulin therapy and its risks, commissioned by the National Blood Authority on behalf of all Australian Governments, pp. 151–154. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf.

Comi, G, Roveri, L, Swan, A, et al 2002, A randomised controlled trial of intravenous immunoglobulin in IgM paraprotein associated demyelinating neuropathy, Journal of Neurology, 249, no. 10, pp. 1370–7.

Dalakas, MC, Quarles, RH, Farrer, RG, et al 1996, A controlled study of intravenous immunoglobulin in demyelinating neuropathy with IgM gammopathy, Annals of Neurology, vol. 40, no. 5, pp. 792–5.

Frommer, M & Madronio, C 2006, The use of intravenous immunoglobulin in Australia. A report for the National Blood Authority, Part B: systematic literature review, Sydney Health Projects Group, University of Sydney, Sydney, pp. 40–1.

European Federation of Neurological Societies, Peripheral Nerve Society, Hadden, RD, Nobile-Orazio, E, et al 2006, ‘European Federation of Neurological Societies/Peripheral Nerve Society guideline on management of paraproteinaemic demyelinating neuropathies: report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society’, European Journal of Neurology, vol. 13, no. 8, pp. 809–18.

Lunn, MPT & Nobile-Orazio, E 2006, ‘Immunotherapy for IgM anti- Myelin-Associated Glycoprotein paraprotein-associated peripheral neuropathies (Cochrane Review)’, in The Cochrane Library, Issue 2, John Wiley & Sons, Ltd, Chichester, UK.

Mariette, X, Chastang, C, Clavelou, P, et al 1997, ‘A randomised clinical trial comparing interferon-alpha and intravenous immunoglobulin in polyneuropathy associated with monoclonal IgM. The IgM-associated Polyneuropathy Study Group’, Journal of Neurology, Neurosurgery & Psychiatry, vol. 63, no. 1, pp. 28–34.

Medical condition Kidney transplantation
Indication for IVIg use

Pre-transplantation

Patients in whom an antibody or antibodies prevent transplantation (donor specific anti-human leukocyte antigen (HLA) antibody/ies or anti-blood group antibody).

Post-transplantation

To treat steroid-resistant acute rejection which may be cellular or antibody mediated.

For prevention and/or treatment of rejection where other therapies are contraindicated or pose a threat to the graft or patient.

Level of evidence Clear evidence of benefit (Category 1).
Description and diagnostic criteria

Transplant rejection occurs when a recipient’s immune system attacks a transplanted organ or tissue. Despite the use of immunosuppressants, one or more episodes of rejection can occur after transplantation. Both cellular and humoral (antibody-mediated) effector mechanisms can play a role.

The presence and pattern of rejection need to be established by biopsy. Laboratory tests to assess the presence and strength of antibodies to the donor antigens can provide additional useful information. Clinical assessment, blood tests, ultrasound and nuclear imaging are used primarily to exclude other causes of organ dysfunction.

Acute cellular rejection occurs in 15–30% of renal transplants and is responsive to steroids in more than 90% of cases. When rejection is steroid resistant, IVIg is a safer therapy than anti-T cell antibody therapy with equal efficacy.

Antibody mediated rejection (AbMR) occurs in 5–10% of renal transplants that have been performed with a compatible cross match. Before the use of IVIg and plasma exchange, AbMR failed to respond adequately to therapy in most cases. Additionally, complications from therapy were severe and sometimes fatal. AbMR responds to IVIg with or without plasma exchange in more than 85% of patients.

Justification for evidence category

An RCT enrolling adult patients with end stage renal disease (ESRD) who were highly sensitised to HLA antigens found that IVIg was better than placebo in reducing anti-HLA antibody levels in highly sensitised patients with ESRD (followed for two years after transplant), and that transplant rates were improved with IVIg therapy (Jordan et al 2004).

Multiple case series have been reported in the literature, indicating efficacy in (acute) antibody mediated rejection, and recommended by a consensus conference (Takemoto et al 2004).

Jordan et al (1998) combined data from seven renal transplant recipients and three heart transplant recipients with steroid-resistant combined antibody-mediated and cellular rejection. All patients in this series were successfully treated with high-dose IVIg.

A small RCT of transplanted patients with a five-year follow-up period showed that IVIg was as effective as OKT3 monoclonal antibodies in the treatment of steroid resistant rejection (survival rate at two years was 80% in both groups) but IVIg generated fewer side effects (Casadei et al 2001).

Qualifying criteria for IVIg therapy

Pre-transplantation

Patients in whom an antibody or antibodies prevent transplantation (donor-specific anti-HLA antibody/ies or anti-blood group antibody).

Post-transplantation

  1. Biopsy proven cellular rejection unresponsive to steroids with clinical evidence of graft dysfunction;

OR

  1. Acute antibody mediated rejection with clinical evidence of graft dysfunction;

OR

  1. As treatment or prophylaxis for rejection where conventional immunosuppressive therapy is contraindicated, for example:
  • in a patient with life-threatening infection in whom conventional immunosuppression will place the patient at even greater risk;
  • when the transplant is at risk (e.g. due to BK virus infection).
Review criteria for assessing the effectiveness of IVIg use
  • Allograft organ function tests.
  • Biopsy response.
  • Laboratory monitoring of anti-HLA antibody and/or anti-blood group antibody responses.
  • Duration of graft and patient survival.
  • Reversal of clinical graft dysfunction.
Dose

IVIg with plasma exchange: 0.1 to 0.5 g/kg post exchange.

IVIg alone: 2 g/kg to a maximum of 140 g as a single dose, or 2 to 3.5 g/kg in a divided dose.

When IVIg is used alone, further doses may be warranted two to four weeks after initial therapy depending on clinical response and/or biopsy findings.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Ahsan, N & Shah, KV 2002, ‘Polyomaviruses: an overview’, Graft, vol. 5, pp. S9–18.

Biotext 2004, ‘Summary data on conditions and papers’, in A systematic literature review and report on the efficacy of intravenous immunoglobulin therapy and its risks, commissioned by the National Blood Authority on behalf of all Australian Governments, pp. 86–7. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf.

Casadei, DH, del C Rial, M, Opelz, G, et al 2001, ‘A randomised and prospective study comparing treatment with high-dose intravenous immunoglobulin with monoclonal antibodies for rescue of kidney grafts with steroid-resistant rejection’, Transplantation, vol. 71, no. 1, pp. 53–8.

Conti, DJ, Freed, BM, Gruber, SA, et al 1994, ‘Prophylaxis of primary cytomegalovirus disease in renal transplant recipients. A trial of gancyclovir vs. immunoglobulin’, Archives of Surgery, vol. 129, no. 4, pp. 443–7.

Frommer, M & Madronio, C 2006, The use of intravenous immunoglobulin in Australia. A report for the National Blood Authority, Part B: systematic literature review, Sydney Health Projects Group, University of Sydney, Sydney, pp. 18–20.

Jordan, SC, Tyan, D, Stablein, D, et al 2004, ‘Evaluation of intravenous immunoglobulin as an agent to lower allosensitisation and improve transplantation in highly sensitized adult patients with end-stage renal disease: report of the NIH IG02 trial’, Journal of the American Society of Nephrology, vol. 15, no. 12, pp. 3256–62.

Jordan, SC, Vo, A, Bunnapradist, S, et al 2003, ‘Intravenous immune globulin treatment inhibits cross match positivity and allows for successful transplantation of incompatible organs in living-donor and cadaver recipients’, Transplantation, vol. 76, no. 4, pp. 631–6.

Jordan, SC, Vo, AA, Tyan, D, et al 2005, ‘Current approaches to treatment of antibody-mediated rejection’, Pediatric Transplantation, vol. 9, no. 3, pp. 408–15.

Lian, M, Chan, W, Slavin, M, et al 2006, ‘Miliary tuberculosis in a Caucasian male transplant recipient and the role of intravenous immunoglobulin as an immunosuppressive sparing agent’, Nephrology (Carlton), vol. 11, no. 2, pp. 156–8.

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, vol. 72, no. 3, pp. 419–22.

Moger, V, Ravishankar, M, Sakhuja, V, et al 2004, ‘Intravenous immunoglobulin: a safe option for treatment of steroid-resistant rejection in the presence of infection’, Transplantation, vol. 77, no. 9, pp. 1455–6.

Orange, JS, Hossny, EM, Weiler, CR, et al 2006, ‘Use of intravenous immunoglobulin in human disease: A review of primary evidence by members of the Primary Immunodeficiency Committee of the American Academy of Allergy, Asthma and Immunology’, Journal of Allergy and Clinical Immunology, vol. 117, no. 4, pp. S525–53.

Peraldi, MN, Akposso, K, Haymann, JP, et al 1996, ‘Long-term benefit of intravenous immunoglobulins in cadaveric kidney retransplantation’, Transplantation, vol. 62, no. 11, pp. 1670–3.

Puliyanda, D, Radha, RK, Amet, N, et al 2003, ‘IVIg contains antibodies reactive with polyoma BK virus and may represent a therapeutic option for BK nephropathy’, American Journal of Transplantation, vol. 3, suppl. 4, p. 393.

Sonnenday, CJ, Warren, DS, Cooper, MC, et al 2004, ‘Plasmapheresis, CMV hyperimmune globulin, and anti-CD20 allow ABO-incompatible renal transplantation without splenectomy’, American Journal of Transplantation, vol. 4, pp. 1315–22.

Takemoto, SK, Zeevi, A, Feng, S, et al 2004, ‘National conference to assess antibody-mediated rejection in solid organ transplantation’, American Journal of Transplantation, vol. 4, no. 7, pp. 1033–41.

Tydén, G, Kumlien, G, Genberg, H, et al 2005, ‘ABO incompatible kidney transplantations without splenectomy, using antigen-specific immunoadsorption and rituximab’, American Journal of Transplantation, vol. 5, no. 1, pp. 145–8.

UK National Kidney Federation 2002, ‘Transplant’. Available from: www.kidney.org.uk/Medical-Info/transplant.html#rej [cited 7 Dec 2007]

Medical condition Multiple sclerosis (MS)
Indication for IVIg use

Short-term therapy in patients with clinically definite relapsing remitting MS in the following circumstances:

  • Pregnancy and the immediate post-partum period when other immunomodulation is contraindicated;
  • Young patients with severe relapsing remitting disease in whom other therapies have failed;
  • Severe relapse with no response to high-dose methylprednisolone.
Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria

MS is a chronic disorder of the central nervous system (CNS) characterised by a triad of inflammation, demyelination and gliosis. Lesions of MS, known as plaques, are typically disseminated in time and location throughout the brain and spinal cord.

Four clinical types of MS have been described: relapsing/remitting MS (RRMS), primary progressive MS (PPMS), secondary progressive MS (SPMS), and progressive/relapsing MS (PRMS).

Diagnosis requires two or more episodes of symptoms and two or more signs that reflect pathology in anatomically non-contiguous white matter tracts of the CNS. Symptoms must last >24 hours and occur as separate episodes at least one month apart. At least one of the two signs must be present on neurological examination, while the other may be detected by paraclinical tests such as intrathecal IgG (oligoclonal bands and visual evoked potentials).

Justification for evidence category

The Biotext (2004) literature review included one systematic review, six RCTs, three case-control studies and one case-series with a total sample size of 849. The quality of the included studies varied widely. The systematic review found some benefit. No benefit was found in two of the RCTs (IVIg did not appear to reverse established muscle weakness), and significant benefit was reported in two RCTs. The other two RCTs were identified by Biotext from the Cochrane register of trials, but no further information about the studies was obtained.

The review by Frommer and Madronio (2006) included eight high-quality RCTs and one medium-quality double-blinded controlled trial with a total of 708 patients. These studies suggested that the occurrence of relapse may be reduced by IVIg at three years, but conclusive evidence in relation to the use of IVIg in reducing relapse rates and severity of relapse in established disease could not be demonstrated. IVIg treatment for the first year from onset of the first neurological event significantly lowered the incidence of second attacks and reduced disease activity as measured by MRI.

IVIg administered in monthly pulses for up to two years appeared to reduce annual exacerbation rates in patients with RRMS and SPMS, but its effect on long-term disability was unclear.

Qualifying criteria for IVIg therapy

Clinically definite RRMS as defined by McDonald et al (2001) criteria and confirmed by a neurologist with one of the following indications:

  1. Pregnancy and immediate post partum period when other immunomodulation is contraindicated;

OR

  1. Young patients with severe relapsing remitting disease in whom other therapies have failed;

OR

  1. Severe relapse with no response to high-dose methylprednisolone.

Application for IVIg use for these indications will be considered on a case-by-case basis and may be reviewed by an expert neurologist in MS in each state

Note: There are numerous immunomodulatory therapies available for multiple sclerosis. IVIg is not available for routine ongoing treatment for patients with MS.

Exclusion criteria for IVIg therapy
  1. Primary progressive MS.
  2. Progressive phase of MS without relapses.
Review criteria for assessing the effectiveness of IVIg use
  • Six-monthly review by a neurologist is required.
  • Objective evidence of improvement in relapse rate in comparison to pre-treatment levels.
  • Other measures that may be useful include: - expanded disability status scale;

- MS functional scores;

- other functional measures.

Dose

Induction: 1–2 g/kg in 2 to 5 divided doses.

Maintenance dose for indications 1 and 2 above:

0.4–1 g/kg, 4 to 6 weekly.

Aim for minimum dose to maintain optimal functional status.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Achiron, A, Kishner, I, Sarova-Pinhas, I, et al 2004, ‘Intravenous immunoglobulin treatment following the first demyelinating event suggestive of multiple sclerosis: a randomized, double-blind, placebo-controlled trial’, Archives of Neurology, vol. 61, no. 10, pp. 1515–20.

Association of British Neurologists 2005, Guidelines for the use of intravenous immunoglobulin in neurological diseases, The Association, London. Available from: www.theabn.org/abn/userfiles/file/IVIg-guidelines-final-July05.pdf [cited 7 Dec 2007]

Barak, Y, Gabbay, U, Gilad, R, et al 1999, ‘Neuropsychiatric assessment as a secondary outcome measure in a multiple sclerosis intravenous immunoglobulin trial’, International Journal of Psychiatry in Clinical Practice, vol. 3, no. 1, pp. 31–4.

Deisenhammer, F, Fazekas, F, Strasser-Fuchs, S, et al 1999, ‘Intravenous immunoglobulins in multiple sclerosis: results of the Austrian Immunoglobulin in Multiple Sclerosis (AIMS) trial’, Infusionstherapie und Transfusionsmedizin, vol. 26, pp. 42–7.

Fazekas, F, Sorensen, PS, Filippi, M, et al 2005, ‘MRI results from the European Study on Intravenous Immunoglobulin in Secondary Progressive Multiple Sclerosis (ESIMS)’, Multiple Sclerosis, vol. 11, no. 4, pp. 433–40.

Filippi, M, Rocca, MA, Pagani, E, et al 2004, ‘European study on intravenous immunoglobulin in multiple sclerosis: results of magnetization transfer magnetic resonance imaging analysis’, Archives of Neurology, vol. 61, no. 9, pp. 1409–12.

Goodin, DS, Frohman, EM, Garmany, GP, et al 2002, ‘Disease modifying therapies in multiple sclerosis: Subcommittee of the American Academy of Neurology and the MS Council for Clinical Practice Guidelines’, Neurology, vol. 58, pp. 169–78.

Gray, OM, McDonnell, GV & Forbes, RB 2004, ‘Intravenous immunoglobulins for multiple sclerosis (Cochrane Review)’, in The Cochrane Library, Issue 2, John Wiley & Sons, Ltd, Chichester, UK.

Lewanska, M, Siger-Zajdel, M & Selmaj, K 2002, ‘No difference in efficacy of two different doses of intravenous immunoglobulins in MS: clinical and MRI assessment’, European Journal of Neurology, vol. 9, no. 6, pp. 565–72.

McDonald, WI, Compston, A, Edan, G, et al 2001, ‘Recommended diagnostic criteria for multiple sclerosis: guidelines from the International Panel on the diagnosis of multiple sclerosis’, Annals of Neurology, vol. 50, no. 1, pp. 121–7.

Noseworthy, JH, O’Brien, PC, Weinshenker, BG, et al 2000, ‘IV immunoglobulin does not reverse established weakness in MS’, Neurology, vol. 55, no. 8, pp. 1135–43.

Orange, JS, Hossny, EM, Weiler, CR, et al 2006, ‘Use of intravenous immunoglobulin in human disease: A review of primary evidence by members of the Primary Immunodeficiency Committee of the American Academy of Allergy, Asthma and Immunology’, Journal of Allergy and Clinical Immunology, vol. 117, no. 4, pp. S525–53.

Oztekin, N & Oztekin MF 1998, ‘Intravenous immunoglobulin treatment in relapsing-remitting multiple sclerosis: a double blind cross over study’, Multiple Sclerosis, vol. 4, p. 391.

Roed, HG, Langkilde, A, Sellebjerg, F, et al 2005, ‘A double-blind randomised trial of IV immunoglobulin treatment in acute optic neuritis’, Neurology, vol. 64, pp. 804–10.

Sacher, RA & IVIg Advisory Panel 2001, ‘Intravenous immunoglobulin consensus statement’, Journal of Allergy and Clinical Immunology, vol. 108, no. 4, pp. S139–46.

Soelberg-Sorensen, P, Haas, J, Sellebjerg, F, et al 2004, ‘IV immunoglobulins as add-on treatment to methylprednisolone for acute relapses in MS’, Neurology, vol. 63, no. 11, pp. 2008–33.

Soelberg-Sorensen, P, Wanscher, B, Schreiber, K, et al 1997, ‘Effect of intravenous immunoglobulin on gadolinium enhancing lesions on MRI in multiple sclerosis (MS): final results of a double-blind cross-over trial,’ Multiple Sclerosis, vol. 3, suppl., p. 268.

Sorensen, PS, Fazekas, F & Lee, M 2002, ‘Intravenous immunoglobulin G for the treatment of relapsing-remitting multiple sclerosis: a meta-analysis’, European Journal of Neurology, vol. 9, no. 6, pp. 557–63.

Stangel, M, Boegner, F, Klatt, CH, et al 2000, ‘Placebo controlled pilot trial to study the remyelinating potential of intravenous immunoglobulins in multiple sclerosis’, Journal of Neurology, Neurosurgery and Psychiatry, vol. 68, no. 1, pp. 89–92.

Strasser-Fuchs, S, Fazekas, F, Deisenhammer, F, et al 2000, ‘The Austrian Immunoglobulin in MS (AIMS) study: final analysis’, Multiple Sclerosis, vol. 6, suppl. 2, pp. S9–13.

Visser, LH, Beekman, R, Tijssen, CC, et al 2004, ‘A randomized, double-blind, placebo-controlled pilot study of i.v. immune globulins in combination with IV methylprednisolone in the treatment of relapses in patients with MS’, Multiple Sclerosis, vol. 10, no. 1, pp. 89–91.

Medical condition Opsoclonus-myoclonus ataxia (OMA)
Indication for IVIg use Long-term maintenance therapy of OMA in association with other tumour therapies.
Description and diagnostic criteria OMA is an immune-mediated monophasic or multiphasic disorder consisting of opsoclonus (conjugate chaotic eye movements), cerebellar ataxia, and arrhythmic myoclonus affecting the trunk, the head and the extremities. OMA may be either paraneoplastic or idiopathic, presumably para-infectious (e.g. post-viral). In children, OMA complicates about 2–3% of neuroblastomas. In adults, it may occur in association with several cancers, most commonly small-cell lung cancer and breast cancer.
Level of evidence Small case studies only; insufficient data (Category 4a).
Justification for evidence category The Asia–Pacific IVIg Advisory Board (2004) consensus statement summarises several case reports suggesting that IVIg is useful in idiopathic OMA and childhood paraneoplastic OMA associated with neuroblastoma.
Qualifying criteria for IVIg therapy

Diagnosis of OMA by a neurologist:

  1. In children;

OR

  1. As second-line treatment following the use of adrenocorticotrophic hormone or corticosteroids.
  1. Note: Given the rarity of OMA and its devastating effects, IVIg should be used where it is considered appropriate by a neurologist.
Exclusion criteria for IVIg therapy Adult paraneoplastic OMA.
Review criteria for assessing the effectiveness of IVIg use

Review

Regular review by neurologist is required; frequency as determined by clinical status of patient.

For stable patients on maintenance treatment, review by a neurologist is required at least annually.

Effectiveness

Objective indicators of relief of symptoms of OMA and improvement or stabilisation of scores of ADLs.

Dose

Induction: 1–2 g/kg in 2 to 5 divided doses.

Maintenance: 0.4–1 g/kg, 4 to 6 weekly.

Aim for the minimum dose to maintain optimal functional status.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Glatz, K, Meinck, HM & Wildemann, B 2003, ‘Para-infectious opsoclonus-myoclonus syndrome: high dose intravenous immunoglobulins are effective’, Journal of Neurology, Neurosurgery and Psychiatry, vol. 74, no. 2, pp. 279–80.

Kornberg, AJ, for the Asia–Pacific IVIg Advisory Board 2004, Bringing consensus to the use of IVIg in neurology. Expert consensus statements on the use of IVIg in neurology, 1st edn, Asia–Pacific IVIg Advisory Board, Melbourne, pp. 80–82.

National Institute of Neurological Disorders 2006, ‘NINDS opsoclonus myoclonus information page’, January. Available from: www.ninds.nih.gov/disorders/opsoclonus_myoclonus/opsoclonus_myoclonus.htm [cited 7 Dec 2007]

Medical condition Pemphigus foliaceus (PF)
Indication for IVIg use PF resistant to corticosteroids and immunosuppressive therapy or when these agents are contra-indicated.
Level of evidence Small case studies only; insufficient data (Category 4a).
Description and diagnostic criteria

PF is a rare autoimmune blistering skin disease characterised by loss of cohesion of cells (acantholysis) in the superficial (subcorneal) layers of the epidermis. The lesions are generally well demarcated and do not coalesce to form large eroded areas (as seen in pemphigus vulgaris). It is mediated by an autoantibody that targets desmoglein 1, a cell-to-cell protein molecule that binds the desmosomes of neighbouring keratinocytes in the epidermis.

The disease has a long-term course with patients maintaining satisfactory health. Spontaneous remissions occasionally occur.

Justification for evidence category Habif (2004) concluded that IVIg was effective as monotherapy for PF and particularly useful in patients who experienced life-threatening complications from immunosuppression. Sami et al (2002) observed that autoantibody titres to desmoglein 1 in a series of 15 PF patients declined persistently following IVIg therapy.
Qualifying criteria for IVIg therapy

Severe widespread PF, defined as disease involving 30% or more of body surface area, diagnosed by a dermatologist;

AND

  1. Corticosteroids or immunosuppressive agents are contraindicated;

OR

  1. Condition is unresponsive to corticosteroids and immunosuppressive agents;

OR

  1. Presenting with severe side effects of therapy.
Review criteria for assessing the effectiveness of IVIg use
  • Response demonstrated at review at six months. Improvement to be demonstrated for continuation of supply.
  • Clinical progression: Treatment is stopped when patients are clinically free from disease and have a negative finding on direct immunofluorescence.
  • Autoantibody titres reflect the response to systemic therapy.
Dose

Efficacy demonstrated with doses of at least 2 g/kg per monthly treatment cycle.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Habif TP. Vesicular and bullous diseases. Chapter 16 in: Clinical Dermatology [electronic resource] : A Color Guide to Diagnosis and Therapy, 4th edition. Mosby Inc, Edinburgh. 2004

Sami, N, Bhol, KC & Razzaque, A 2002, ‘Influence of IVIg therapy on autoantibody titres to desmoglein 1 in patients with pemphigus foliaceus’, Clinical Immunology, vol. 105, no. 2 pp. 192–8.

Medical condition Phemphigus vulgaris (PV)
Indication for IVIg use Moderate to severe PV as an adjuvant to prolonged corticosteroid treatment.
Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria

PV is a rare but potentially fatal condition accounting for approximately 70% of pemphigus cases. While the cause is unknown, an immuno-genetic predisposition is well established. PV may also be drug-induced. Drugs reported to be most significantly associated with PV include penicillamine, captopril and other thiol-containing compounds. Rifampicin and emotional stress have recently been reported as triggers for PV.

The oral cavity is almost always affected and erosions can be scattered and extensive, with subsequent dysphagia. Blistering and erosions secondary to the rupture of blisters may be painful and limit the patient’s daily activities.

Pemphigus may occur in patients with other autoimmune diseases, particularly myasthenia gravis and thymoma.

Prognosis

The severity and natural history of PV are variable. Before the advent of steroids, most patients with PV died. Treatment with systemic steroids has reduced the mortality rate to 5–15%. Most deaths occur during the first few years of disease and if the patient survives five years, the prognosis is good. Early disease is easier to control than widespread disease and mortality may be higher if therapy is delayed. Morbidity and mortality are related to the extent of disease, the maximum dose of corticosteroid required to induce remission, and the presence of other diseases.

Justification for evidence category In a retrospective cohort study, 15 corticosteroid- dependent patients with moderate to severe PV were treated with IVIg and followed over a mean period of 6.2 years. All 15 patients had a satisfactory clinical response to IVIg therapy. IVIg had a demonstrable corticosteroid-sparing effect and was considered a safe and effective alternative treatment in patients who were dependent on systemic corticosteroids or who developed significant adverse effects as a result of their use (Biotext 2004).
Qualifying criteria for IVIg therapy

Moderate to severe disease diagnosed by a dermatologist;

AND

  1. Corticosteroids or immunosuppressive agents are contraindicated;

OR

  1. Condition is unresponsive to corticosteroids and immunosuppressive agents;

OR

  1. Presenting with severe side effects of therapy.
Review criteria for assessing the effectiveness of IVIg use
  • Response demonstrated at review at six months. Improvement to be demonstrated for continuation of supply.
  • Titres of serum antibodies against keratinocytes.
  • Whether systemic corticosteroids can be gradually discontinued.
  • Total dose and duration of corticosteroid therapy, and number of relapses before and after the initiation of IVIg therapy.
Dose

Efficacy demonstrated with doses of at least 2 g/kg per monthly treatment cycle.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Biotext 2004, ‘Summary data on conditions and papers’, A systematic literature review and report on the efficacy of intravenous immunoglobulin therapy and its risks, commissioned by the National Blood Authority on behalf of all Australian Governments, pp. 240–1. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf.

Bystryn, JC, Jiao, D & Natow, S 2002, ‘Treatment of pemphigus with intravenous immunoglobulin’, Journal of the American Academy of Dermatology, vol. 47, no. 3, pp. 358–63.

Sami, N, Oureshi, A, Ruocco, E, et al 2002, ‘Corticosteroid-sparing effect of intravenous immunoglobulin therapy in patients with pemphigus vulgaris’, Archives of Dermatology, vol. 138, pp. 1158–62.

Medical condition Post-transfusion purpura (PTP)
Indication for IVIg use Treatment of profound thrombocytopenia associated with bleeding.
Level of evidence Small case studies only; insufficient data (Category 4a).
Description and diagnostic criteria

PTP is caused by antibodies to platelet-specific antigens, usually anti-HPA1a. PTP may result in profound thrombocytopenia with associated life- threatening bleeding. While the platelet count typically recovers spontaneously, this can take several weeks or more.

Specialised investigations (antibody screening, patient/donor genotyping) and antigen-matched platelet and/or red cell transfusion support may be required — contact the Blood Service for more information.

Justification for evidence category Mueller-Eckhardt and Kiefel (1988) evaluated the effect of high-dose IgG (HDIgG) in 11 PTP cases investigated in one institution and summarised clinical data on 8 additional reported cases. Of 17 cases, 16 had good or excellent response to HDIgG, attaining normal platelet counts within a few days; only one failure was observed. Five patients relapsed, but attained complete remission after a second course (dose) of IgG. Total IgG doses per course were in the range 52–180 g. Five different IgG preparations were used and seemed similarly effective. No adverse reactions were observed. The authors conclude that HDIgG is the treatment of choice for PTP.
Qualifying criteria for IVIg therapy

Clinical diagnosis/suspicion of PTP with thrombocytopenia associated with life-threatening bleeding.

Note: Laboratory confirmation is desirable where possible in the time frame (usually an urgent, life-threatening clinical situation).

Review criteria
  • Platelet counts in the days and weeks following IVIg.
  • Resolution of bleeding.
Dose

1 g/kg as a total dose, repeated if necessary

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Gonzalez, CE & Pengetze, YM 2005, ‘Post-transfusion purpura’, Current Haematology Reports, vol. 4, no. 2, pp. 154–9.

Mueller-Eckhardt, C & Kiefel, V 1988, ‘High-dose IgG for post- transfusion purpura – revisited’, Blut, vol. 57, no. 4, pp. 163–7.

Medical condition Toxic epidermal necrolysis (TEN; Lyell syndrome) Stevens–Johnson syndrome (SJS)
Indication for IVIg use To limit progression of TEN or SJS/TEN when administered in early stages.
Level of evidence Small case studies only; insufficient data (Category 4a).
Description and diagnostic criteria

TEN is a rare, life-threatening hypersensitivity reaction to certain medications, such as sulphonamides, antibiotics, non-steroidal anti-inflammatory drugs and anti-convulsants. Drug-induced epidermal apoptosis has been proposed as possible pathogenesis. Stevens–Johnson syndrome (SJS) is a less extensive manifestation of the same phenomenon.

TEN and SJS are characterised by severe bullous reaction with extensive destruction of the epidermis, and morphologically by ongoing apoptotic keratinocyte cell death that results in the separation of the epidermis from the dermis.

The term SJS is now used to describe patients with blistering and skin detachment involving a total body surface area of <10%. SJS/TEN describes patients with 10–30% detachment, and TEN describes patients with >30% skin detachment.

Justification for evidence category

The Biotext (2004) review identified one small cohort study (20 patients) without a control group, which found that there appeared to be no significant effect and that death rate seems to be higher than previously reported.

The Frommer and Madronio (2006) review found one small randomised study (four patients) with a control group of two patients (supportive care only). This study found that there was some improvement in epithelialisation and prominent difference in CD95 receptor between treated patients and controls. However, neither IVIg nor its comparison group could completely stop the TEN process.

Qualifying criteria for IVIg therapy

TEN or SJS/TEN overlap with all the following:

  1. Diagnosis by a dermatologist;

AND

  1. Body surface area (erythema and/or erosions) of 10% or more;

AND

  1. Evidence of rapid evolution.

Notes:

  • IVIg should be initiated as early as possible, preferably within 24 hours of diagnosis.
  • Urgent skin biopsy should be performed for confirmation but should not delay IVIg therapy if indicated.
  • The Adverse Drug Reactions Advisory Committee should be notified of the inciting medication.
Exclusion criteria for IVIg therapy SJS alone
Dose

2 g/kg, preferably as a single dose, or divided over three consecutive days.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Bachot, N, Revuz, J & Roujeau, JC 2003, ‘Intravenous immunoglobulin treatment for Stevens-Johnson syndrome and toxic epidermal necrolysis: a prospective non-comparative study showing no benefit on mortality or progression’ (comment), Archives of Dermatology, vol. 139, no. 1, pp. 85–6.

Biotext 2004, ‘Summary data on conditions and papers’, in A systematic literature review and report on the efficacy of intravenous immunoglobulin therapy and its risks, commissioned by the National Blood Authority on behalf of all Australian Governments, pp. 242–7. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf.

Campione, E, Marulli, GC, Carrozzo, AM, et al 2003, ‘High-dose intravenous immunoglobulin for severe drug reactions: efficacy in toxic epidermal necrolysis’, Acta Dermato-venereologica, vol. 83, no. 6, pp. 430–2.

Frommer, M & Madronio, C 2006, The use of intravenous immunoglobulin in Australia. A report for the National Blood Authority, Part B: systematic literature review, Sydney Health Projects Group, University of Sydney, Sydney, pp. 55–6.

Paquet, P, Jacob, E, Damas, P, et al 2005, ‘Analytical quantification of the inflammatory cell infiltrate and CD95R expression during treatment of drug-induced toxic epidermal necrolysis’, Archives of Dermatological Research, vol. 297, no. 6, pp. 266–73.

Prins, C, Vittorio, C, Padilla, RS, et al 2003, ‘Effect of high- dose intravenous immunoglobulin therapy in Stevens-Johnson syndrome: a retrospective, multicentre study’, Dermatology, vol. 207, no. 1, pp. 96–9.

Trent, J, Halem, M, French, LE, et al 2006, ‘Toxic epidermal necrolysis and intravenous immunoglobulin: a review’, Seminars in Cutaneous Medicine and Surgery, vol. 25, no. 2, pp. 91–3.

Medical condition Toxic shock syndrome (TSS)
Indication for IVIg use

Streptococcal TSS: In view of the high mortality risk, IVIg is indicated for early use in both adults and children.

Staphylococcal TSS: IVIg is indicated where rapid improvement is not obtained with fluid resuscitation and inotropes.

In both conditions IVIg is used in addition to surgical intervention, antibiotic therapy and supportive measures.

Level of evidence Small case studies only; insufficient data (Category 4a).
Description and diagnostic criteria

TSS is a life-threatening illness characterised by hypotension and multi-organ failure. It may be caused by Staphylococcus aureus (rarely isolated) or Streptococcus pyogenes that produce and release superantigenic exotoxins. The exotoxins activate T-cells on a large scale resulting in a massive release of inflammatory cytokines.

Streptococcal TSS is defined by:

I Group A Streptococci (S. pyogenes) isolated from:

  • (IA) a normally sterile site (e.g. blood, cerebrospinal fluid, pleural or peritoneal fluid, tissue biopsy, surgical wound); or
  • (IB) a non-sterile site (e.g. throat, sputum, vagina, superficial skin lesion).

IIA. Hypotension: systolic blood pressure = 90 mmHg in adults or in the 5th percentile for age in children; and

IIB. Two or more of the following:

  1. Renal impairment: serum creatinine for adults at least twice the upper limit of normal for age; in patients with existing renal disease, elevation over baseline by a factor of at least 2;
  2. Coagulopathy: platelet count of ≤100x109/L or disseminated intravascular coagulation, defined by prolonged clotting times, low fibrinogen level, and the presence of fibrin degradation products;
  3. Liver involvement: alanine aminotransferase (ALT), aspartate aminotransferase (AST), or total bilirubin level at least twice the upper limit of normal for age; in patients with existing liver disease, elevation over baseline by a factor of 2;
  4. Adult respiratory distress syndrome, defined by acute onset of diffuse pulmonary infiltrates and hypoxaemia in the absence of cardiac failure; or evidence of diffuse capillary leak manifested by acute onset or generalised oedema; or pleural or peritoneal effusions with hypoalbuminaemia;
  5. Generalised erythematous macular rash that may desquamate;
  6. Soft tissue necrosis, including necrotising fasciitis or myositis; or gangrene.

A definite case is an illness fulfilling criteria IA and II (A and B).

A probable case is an illness fulfilling criteria IB and II (A and B) where no other aetiology is identified.

(Working Group on Severe Streptococcal Infections 1993).

Staphylococcal TSS is defined by:

  1. Fever: temperature ≥38.9°C;
  2. Hypotension: systolic blood pressure ≤90 mmHg in adults or in the 5th percentile for age in children;
  3. Diffuse macular rash with subsequent desquamation one to two weeks after onset (including palms and soles);
  4. Multisystem involvement (three or more of the following):
  1. Hepatic: bilirubin or aminotransferase ≥2 times normal;
  2. Haematologic: platelet count ≤100x109/L;
  3. Renal: blood urea nitrogen or serum creatinine level ≥2 times normal;
  4. Mucous membranes: vaginal, oropharyngeal or conjunctival hyperaemia;
  5. Gastrointestinal: vomiting or diarrhoea at onset of illness;
  6. Muscular: severe myalgia or serum creatine phosphokinase level ≥2 times upper limit;
  7. Central nervous system: disorientation or alteration in consciousness without focal neurological signs and in the absence of fever or hypotension.

A confirmed case is a case with all of the manifestations described above. However, in severe cases death may occur before desquamation develops.

A probable case is an illness with all but any one of the manifestations described above (Wharton et al 1990).

Prognosis

Streptococcal TSS has a mortality rate of 30–80% in adults and 5–10% in children, with most deaths secondary to shock and respiratory failure.

Staphylococcal TSS can also be fatal but mostly has a better prognosis.

Justification for evidence category

Streptococcal TSS: A small case series (Norrby-Teglund et al 2005), a cohort study (Kaul et al 1999) and an RCT, which was terminated prematurely (Darenberg et al 2003), suggested that IVIg improves outcomes.

Staphylococcal TSS: In vitro and animal studies suggested that IVIg is effective in neutralising staphylococcal superantigens. Anecdotal reports refer to the clinical effectiveness of IVIg in staphylococcal TSS.

Qualifying criteria for IVIg therapy
  1. Diagnosis of streptococcal or staphylococcal TSS in accordance with criteria listed above, preferably with isolation of organism;

AND

  1. Failure to achieve rapid improvement with fluid resuscitation, inotropes, surgery, antibiotic therapy and other supportive measures.
Dose

2 g/kg as a single dose.

Schrage et al (2006) reported differences between various preparations of IVIg and their ability to neutralise streptococcal superantigens. They commented that ‘the variations between IVIg preparations from different manufacturers are most likely caused by the different geographical regions from which the plasma samples were collected and might reflect differences in ... group A streptococcal ... exposure.’ The clinical significance of these findings is not yet known.

Darenberg et al (2004) suggested that higher doses of IVIg might be required for staphylococcal TSS than streptococcal TSS, based on in vitro neutralisation of superantigens.

Dosing above 1 g/kg per day is contraindicated for some IVIg products.

Refer to the current product information sheet for further information.

The aim should be to use the lowest dose possible that achieves the appropriate clinical outcome for each patient.

Bibliography

Darenberg, J, Ihendyane, N, Sjoelin, J, et al 2003, ‘Intravenous immunoglobulin G therapy for streptococcal toxic shock syndrome: a European randomised double-blind placebo- controlled trial’, Clinical Infectious Diseases, vol. 37, pp. 333–40.

Darenberg, J, Söderquist, B, Normark, BH, et al 2004, ‘Differences in potency of intravenous polyspecific immunoglobulin G against streptococcal and staphylococcal superantigens; implications for therapy of toxic shock syndrome’, Clinical Infectious Diseases, vol. 38, pp. 836–42.

Kaul, R, McGeer, A, Norrby-Teglund, A, et al 1999, ‘Intravenous immunoglobulin therapy for streptococcal toxic shock syndrome – a comparative observational study’, Clinical Infectious Diseases, vol. 28, pp. 800–7.

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’, Scandinavian Journal of Infectious Diseases, vol. 37, no. 3, pp. 166–72.

Schlievert, PM 2001, ‘Use of intravenous immunoglobulin in the treatment of staphylococcal and streptococcal toxic shock syndromes and related illnesses’, Journal of Allergy and Clinical Immunology, vol. 108, no. 4, suppl., pp. S107–10.

Schrage, B, Duan, G, Yang, LP, et al 2006, ‘Different preparations of intravenous immunoglobulin vary in their efficacy to neutralise streptococcal superantigens: implications for treatment of streptococcal toxic shock syndrome’, Clinical Infectious Diseases, vol. 43, no. 6, pp. 743–6.

Stevens, DL 1998, ‘Rationale for the use of intravenous gamma globulin in the treatment of streptococcal toxic shock syndrome’, Clinical Infectious Diseases, vol. 26(3), pp. 639–41.

Working Group on Severe Streptococcal Infections 1993, ‘Defining the Group A Streptococcal toxic shock syndrome: rationale and consensus definition’, Journal of the American Medical Association, vol. 269, pp. 390–401.