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

Conditions for which IVIg has an established therapeutic role

This chapter comprises conditions for which intravenous immunoglobulin (IVIg) use is well established in Australia. There is evidence from reasonable-quality studies and clinical support for IVIg therapy in selected patients. For a number of conditions, IVIg is first-line therapy and may be the only established treatment option; for example, as replacement therapy in primary immunodeficiency disease.

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 2 Conditions for which IVIg has an established therapeutic role as immunoglobulin-replacement therapy

Conditions for which IVIg has an established therapeutic role as immunoglobulin-replacement therapy
Condition Evidence level Page
Acquired hypogammaglobulinaemia secondary to haematological malignancies (chronic lymphocytic leukaemia, multiple myeloma, non-Hodgkin lymphoma and other relevant malignancies, and post- haemopoietic stem cell transplantation) 2a 48
Primary immunodeficiency diseases with antibody deficiency 2a 55

Table 3 Conditions for which IVIg has an established therapeutic role as immunomodulation therapy

Conditions for which IVIg has an established therapeutic role as immunomodulation therapy
Condition Evidence level Page
Chronic inflammatory demyelinating polyneuropathy 1 58
Guillain–Barré syndrome 1 62
Idiopathic (autoimmune) thrombocytopenic purpura (ITP) in adults 2a 66
Inflammatory myopathies (polymyositis, dermatomyositis, inclusion body myositis) 2a 73
Kawasaki disease 1 79
Lambert–Eaton myasthenic syndrome 2a 83
Multifocal motor neuropathy 1 87
Myasthenia gravis 1 92
Neonatal haemochromatosis 2a 96
Stiff person syndrome 2a 99
Medical condition Acquired hypogammaglobulinaemia secondary to haematological malignancies chronic lymphocytic leukaemia (CLL), multiple myeloma (MM), non-Hodgkin lymphoma (NHL) and other relevant malignancies, and post-haemopoietic stem cell transplantation (HSCT)
Indication for IVIg use

Prevention of recurrent bacterial infections due to antibody failure associated with haematological malignancies.

Prevention of recurrent bacterial infections in patients undergoing HSCT for haematological malignancies.

Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria The manifestations of haematological malignancies can include a wide range of symptoms and physical and laboratory abnormalities in an individual patient. For diagnostic criteria, refer to the current World Health Organization classification criteria.
Justification for evidence category

One small crossover study of 12 patients with CLL or NHL reported that the number of serious bacterial infections was significantly decreased (p = 0.001) in the months in which patients received IgG every three weeks for one year. Serious bacterial infections showed a trend to be associated with an IgG level <6.4 g/L.

Three randomised controlled trials (RCTs) and one crossover trial of low–moderate quality reported a reduction in infection rates in CLL patients with hypogammaglobulinaemia after three to four-weekly administration of IVIg for one year.

One placebo-controlled RCT of monthly IVIg given to 82 MM patients for one year (with 22 withdrawing due to reaction) concluded that IVIg protects against life-threatening infections and significantly reduces risk of recurrent infections. The greatest benefit was seen in individuals who had a poor response to pneumococcal vaccine. A small prospective RCT with 30 multiple myeloma patients reported a possible decrease in symptoms of chronic bronchitis.

A recent systematic review and meta-analysis of patients undergoing HSCT [60 trials (>4000 patients)] reported an increased risk of veno-occlusive disease with no survival benefit particularly in studies conducted since 2000. The authors concluded that routine prophylaxis with IVIg is not supported, but suggest that its use may be considered in lymphoproliferative disorder patients with hypogammaglobulinaemia and recurrent infections, for reduction of clinically documented infections.

Qualifying criteria for IVIg therapy

Diagnosis of acquired hypogammaglobulinaemia secondary to haematological malignancies or stem cell transplantation with:

  • Recurrent or severe bacterial infection(s) and evidence of hypogammaglobulinaemia (excluding paraprotein);

OR

  • Hypogammaglobulinaemia with IgG <4 g/L (excluding paraprotein).

Note: For data tracking purposes, the type of malignancy being treated should be recorded with each request for IVIg.

Exclusion criteria

The following conditions should not be approved under this indication:

  1. HIV in children (see page 185);
  2. Transplantation-related immunomodulation (solid organ transplantation; (see page 208);
  3. Secondary hypogammaglobulinaemia (including iatrogenic immunodeficiency (see page 106).
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 an 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 <4 g/L.

Subcutaneous administration of immunoglobulin can be considered as an alternative to IVIg. A suggested dose is 0.1 g/kg lean body mass every week modified to achieve an IgG trough level of at least the lower limit of the age-specific serum IgG reference range.

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. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf.

Cordonnier, C, Chevret, S, Legrand, M, et al 2003, ‘Should immunoglobulin therapy be used in allogeneic stem cell transplantation? A randomised, double-blind dose effect, placebo-controlled, multicentre trial’, Annals of Internal Medicine, vol. 139, pp. 8–18.

Chapel, H, Dicato, M, Gamm, H, et al 1994, ‘Immunoglobulin replacement in patients with chronic lymphocytic leukaemia: a comparison of two dose regimes’, British Journal of Haematology, vol. 88, pp. 209–12.

Chapel, HM, Lee, M, Hargreaves, R, et al 1994, ‘Randomised trial of intravenous immunoglobulin as prophylaxis against infection in plateau-phase multiple myeloma. The UK Group for Immunoglobulin Replacement Therapy in Multiple Myeloma’, Lancet, vol. 343, no. 8905, pp. 1059–63.

Cooperative Group for the Study of Immunoglobulin in Chronic Lymphocytic Leukaemia 1988, ‘Intravenous immunoglobulin for the prevention of infection in chronic lymphocytic leukaemia. A randomised, controlled clinical trial’, New England Journal of Medicine, vol. 319, pp. 902–7.

Couderc, B, Dujols, JP, Mokhtari, F, et al 2000, ‘The management of adult aggressive non-Hodgkin’s lymphomas’, Critical Reviews in Oncology/Hematology, vol. 35, no. 1, pp. 33–48.

Molica, S, Musto, P, Chiurazzi, F, et al 1996, ‘Prophylaxis against infections with low dose intravenous immunoglobulins in chronic lymphocytic leukaemia. Results of a crossover study’, Haematologica, vol. 81, pp. 121–6.

Orange, JS, Hossny, EM, Weiler, CR, et al 2006, ‘Use of intravenous immunoglobulin in human disease: a review of 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.

Raanani, P, Gafter-Gvili, A, Mical, P, et al 2009, ‘Immunoglobulin prophylaxis in hematopoietic stem cell transplantation: systematic review and meta-analysis’, Journal of Clinical Oncolology, vol. 27, no. 5, pp. 770-81.

Raanani, P, Gafter-Gvili, A, Paul, M, Ben-Bassat, I, Leibovici, L & Shpilberg, O 2008, ‘Immunoglobulin prophylaxis in hematological malignancies and hematopoietic stem cell transplantation’, Cochrane Database of Systematic Reviews, Issue 4, art. no. CD006501, doi:10.1002/14651858.CD006501. pub2.

Sklenar, I, Schiffman, G, Jonsson, V, et al 1993, ‘Effect of various doses of intravenous polyclonal IgG on in vivo levels of 12 pneumococcal antibodies in patients with chronic lymphocytic leukaemia and multiple myeloma’, Oncology, vol. 50, no. 6, pp. 466–77.

Thomson, D 2005, ‘Lymphatic malignancies-non-Hodgkin’s lymphoma’, Hospital Pharmacist, vol. 12, pp. 353–8.

Winston, DJ, Antin, JH, Wolff, SN, et al 2001, ‘A multicentre randomised double-blind comparison of different doses of intravenous immunoglobulin for prevention of graft-versus- host disease and infection after allogeneic bone marrow transplantation’, Bone Marrow Transplantation, vol. 28, pp. 187–96.

Weeks, JC, Tierney, MR & Weinstein, MC 1991, ‘Cost effectiveness of prophylactic intravenous immune globulin in chronic lymphocytic leukaemia’, New England Journal of Medicine, vol. 325, pp. 61–6.

Medical condition

Primary immunodeficiency diseases (PID) with antibody deficiency

This excludes:

  1. specific antibody deficiency (see page 110);
  2. IgG subclass deficiency (not funded see page 112).
Indication for IVIg use Management of infection related to antibody deficiency.
Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria

PID comprise a group of more than 120 separate conditions. Many of these are manifest by failure of protective antibody production. Key diagnoses include common variable immunodeficiency (CVID), severe combined immunodeficiencies, transient hypogammaglobulinaemia of infancy, Wiskott Aldrich syndrome and X-linked agammaglobulinaemia. In certain conditions, such as Wiskott Aldrich syndrome, antibody failure may not be manifest as hypogammaglobulinaemia but functional antibody responses will be impaired.

Some PID does not involve antibody failure, such as chronic granulomatous disease and deficiencies of complement components. In these cases, antibody replacement therapy is not justified.

Justification for evidence category The Biotext (2004) review reported level 2a evidence for the use of IVIg in the treatment of common variable immunodeficiency and primary hypogammaglobulinaemia.
Qualifying criteria for IVIg therapy In each case, a specific PID diagnosis must be established under the supervision of a specialist clinical immunologist and the diagnosis must be advised for IVIg to be approved.
Exclusion criteria for IVIg therapy

The following conditions should not be approved under this indication:

  1. Miscellaneous hypogammaglobulinaemia (see Secondary hypogammaglobulinaemia, page 106)
  2. Specific antibody deficiency (see page 110)
  3. IgG subclass deficiency (not funded; see page 112).
Review criteria for assessing the effectiveness of IVIg use

Review criteria for primary immunodeficiency diseases with antibody deficiency are not mandated.

Nevertheless, the following may be of value to the clinician:

  • frequency of clinical episodes of infection
  • trough levels; and
  • renal function.
Dose

Maintenance dose: 0.4 g/kg every four weeks, modifying dose and schedule 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.

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. 218. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf.

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.

Cooper, MD & Schroeder, Jr HW 2005, ‘Primary immune deficiency diseases’, in DL Kasper, E Braunwald, AS Fauci, et al (eds), Harrison’s Textbook of Medicine, 16th edn, McGraw-Hill, New York, pp. 1939–47.

Orange, JS, Hossny, EM, Weiler, CR, et al 2006, ‘Use of intravenous immunoglobulin in human disease: a review of 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 Chronic inflammatory demyelinating polyneuropathy (CIDP), (including IgG and IgA paraproteinaemic neuropathies)
Indication for IVIg use First-line treatment for CIDP with treatment initiated when progression is rapid, or walking is compromised, or there is significant functional impairment.
Level of evidence Clear evidence of benefit (Category 1).
Description and diagnostic criteria

CIDP is an acquired sensorimotor polyneuropathy characterised by a progressive or relapsing/ remitting course with evidence of demyelination on electrophysiological or pathological studies and response to immunomodulating therapies.

There is no specific diagnostic test, but characteristic clinical and laboratory findings help distinguish this disorder from other immune mediated neuropathic syndromes. Serum protein electrophoresis with immunofixation may be indicated to search for monoclonal gammopathy and associated conditions.

Justification for evidence category

The Biotext (2004) review found one Cochrane review of six RCTs with a total sample size of 170. The quality of the studies was low–moderate, found IVIg improved disability in the short-term, and had comparable results to treatment with plasma exchange or prednisolone.

The Frommer and Madronino (2006) review found one low-quality RCT with a total sample size of 20, which demonstrated that more patients responded to immunoadsorption than IVIg, although the baseline disease duration was higher in the IVIg group. Differences were not significant.

Qualifying criteria for IVIg therapy
  1. Diagnosis of CIDP verified by a neurologist; AND
  2. Significant functional impairment of activities of daily living (ADL).
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. Most individuals will respond within three months unless there is significant axonal degeneration whereby a six-month course will be necessary.

If there is no benefit after three to six courses, IVIg therapy should be abandoned.

Review

Regular review by a 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, 2–6 weekly.

The amount per dose should be titrated to the individual’s response.

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

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 Inc., pp. 21–29.

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. 132–3. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf.

Frommer, M & Madronio, C 2006, The use of intravenous immunoglobulin in Australia. A report for the National Blood Authority, Part B, Sydney Health Projects Group, University of Sydney, Sydney, pp. 29–31.

Hughes, RAC, Bouche, P, Cornblath, DR, et al 2006, ‘European Federation of Neurological Societies/Peripheral Nerve Society guideline on management of chronic inflammatory demyelinating polyradiculoneuropathy: report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society’, European Journal of Neurology, pp. 326–32.

van Schaik, IN, Winer, JB, de Haan, R, et al 2002, ‘Intravenous immunoglobulin for chronic inflammatory demyelinating polyradiculoneuropathy (Cochrane Review)’, in The Cochrane Library, Issue 2, John Wiley & Sons, Ltd, Chichester, UK.

Zinman, L, Sutton, HD, Ng, E, et al 2005, ‘A pilot study to compare the use of the Excorim staphylococcal protein immunoadsorption system and IVIG in chronic inflammatory demyelinating polyneuropathy’, Transfusion and Apheresis Science, vol. 33, no. 3, pp. 317–24.

Medical condition Guillain–Barré Syndrome (GBS)
Indication for IVIg use GBS and its variants with significant disability and progression.
Level of evidence Clear evidence of benefit (Category 1).
Description and diagnostic criteria

GBS is the commonest cause of acute flaccid paralysis in the West. The syndrome typically presents with rapidly progressive, relatively symmetrical ascending limb weakness consistent with a polyradiculoneuropathy and often with associated cranial nerve involvement.

Motor signs and symptoms usually predominate over sensory signs and symptoms. Loss of tendon reflexes occurs in most cases. Major complications include respiratory failure and autonomic dysfunction.

The disease is monophasic, reaching its nadir usually within two weeks, although arbitrary definition accepts a limit of four weeks. A plateau phase of variable duration follows the nadir before gradual recovery. Although recovery is generally good or complete in the majority of patients, persistent disability has been reported to occur in about 20% and death in 4 to 15% of patients.

IVIg has been shown to have the same efficacy as plasma exchange. The choice is based on availability, practicality, convenience, cost, and ease or safety of administration (Asia–Pacific IVIg Advisory Group).

Investigations

There is no biological marker for GBS. It is diagnosed by clinical recognition of rapidly evolving paralysis with areflexia. Investigations include the following:

  • Cerebrospinal fluid (CSF) protein elevation, although the level may be normal in the first two weeks of illness. The CSF white cell count may rise transiently, but a sustained pleocytosis suggests an alternative diagnosis or association with an underlying illness (e.g. HIV).
  • Electrophysiological studies may show changes after the first or second week of the illness, including conduction block, conduction slowing or abnormalities in F waves.
Justification for evidence category

One systematic review of nine RCTs of moderate quality found IVIg hastened recovery in adults with GBS to the same degree as plasma exchange (Biotext 2004).

One low-quality RCT with a small sample size (n=21), in which the randomisation of patients to the IVIg treatment group was skewed, was identified. Children who received IVIg treatment showed earlier signs of improvement, and disability scores were lower at four weeks than the placebo group (Frommer and Madronio 2006).

Qualifying criteria for IVIg therapy

Patients with GBS (or variant) with significant disability and disease progression.

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

Review criteria for assessing the effectiveness of IVIg use

Primary outcome measures: improvement in disability grade four weeks after treatment:

  1. healthy
  2. minor symptoms or signs of neuropathy but capable of manual work
  3. able to walk without support of a stick but incapable of manual work
  4. able to walk with a stick, appliance or support
  5. confined to bed or chair bound
  6. requiring assisted ventilation
  7. dead

Secondary outcome measures:

  1. time until recovery of unaided walking
  2. time until recovery of walking with aid
  3. time until discontinuation of ventilation (for those ventilated)
  4. death or disability (inability to walk without aid after 12 months)
  5. treatment-related fluctuation
Dose

2 g/kg in 2 to 5 divided doses.

Approximately 10% of patients relapse, which may require a second treatment with IVIg. A second dose of IVIg must only be on the advice of and after assessment by a neurologist.

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

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

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.149–50. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf.

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. 32–4.

Hughes, RAC, Raphaël, J-C, Swan, AV, et al 2006, ‘Intravenous immunoglobulin for Guillain Barré syndrome (Cochrane Review)’, in The Cochrane Library, Issue 1, John Wiley & Sons, Ltd, Chichester, UK.

Korinthenberg, R, Schessl, J, Kirschner, J, et al 2005, Intravenously administered immunoglobulin in the treatment of childhood Guillain-Barré syndrome: a randomized trial’, Paediatrics, vol. 116, no. 1, pp. 8–14.

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. 14–20.

Medical condition Idiopathic (autoimmune) thrombocytopenic purpura (ITP) — adult
Indication for IVIg use

1. Refractory acute ITP on the recommendation of a clinical haematologist

Patients with severe thrombocytopenia (platelets <30x109/L) who have not responded to corticosteroid therapy.

2. ITP with life-threatening haemorrhage or the potential for life-threatening haemorrhage

Patients with severe thrombocytopenia (<30x109/L) with clinical evidence of a haemostatic defect (e.g. mucous membrane haemorrhage) or active bleeding.

3. ITP in pregnancy

a. Platelets <30x109/L

b. Impending delivery

4. Specific circumstances

a. Planned surgery

b. Other concurrent risk factors for bleeding (e.g. concurrent anti-coagulant therapy)

c. Severe ITP (platelets <30x109/L) where corticosteroids and immunosuppression are contraindicated

d. Chronic ITP under the guidance of a clinical haematologist, as adjunctive therapy or where other therapies have failed or are not appropriate

5. HIV–associated ITP

Patients with severe ITP associated with HIV infection.

Level of evidence Evidence of probable benefit (Category 2a).
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 platelet production (megakaryopoiesis) is morphologically 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. It is a common finding in patients with HIV, and while it may be found at any stage of the infection, its prevalence increases as HIV disease advances.

Around 80% of adults with ITP have the chronic form of disease. The highest incidence of chronic ITP is in women aged 15–50 years, although some reports suggest increasing incidence with age.

Chronic ITP may relapse and remit spontaneously and the course may be difficult to predict. If the platelet count can be maintained at a level that prevents spontaneous bleeding or bruising, the outlook is good.

Justification for evidence category

Five small prospective studies, including three randomised studies, demonstrated equivalent efficacy of IVIg in comparison to prednisone 1 mg/kg/ day and high-dose dexamethasone regimen. Overall, the studies found a dose response with more rapid increment in platelet counts at scheduling ≥0.8 g/kg on day one compared with 0.4 g/kg/day for three days.

A small controlled study (10 patients in each arm) of HIV-positive patients with severe thrombocytopenia reported possible benefit for the restoration and maintenance of platelet count for the duration of the haemorrhagic disorder (Biotext 2004).

An international consensus statement from January 2010 (Provan et al 2010) reported on new data and provided consensus-based recommendations relating to diagnosis and treatment of ITP in adults, in children, and during pregnancy. This statement concluded that few RCTs have been conducted and that multi-centre, prospective RCTs are required.

Qualifying criteria for IVIg therapy

1. Refractory acute ITP:

  1. Patients qualify for initial IVIg therapy when conventional doses of corticosteroids (0.5-2.0 mg/ kg prednisolone, or equivalent) have failed to improve the platelet count or stop bleeding within a clinically appropriate time frame, as assessed by a clinical haematologist. The objective of therapy is to induce a prompt increase in the platelet count (to >30x109/L) while other therapies are introduced.
  2. Patients qualify for continuing doses when splenectomy has failed or is contraindicated AND where therapy with at least one second-line agent has been unsuccessful in maintaining a platelet count >30x109/L.

With ongoing therapy, IVIg may be administered to achieve a platelet count >30x109/L. Further doses may be administered in responsive patients for up to 6 months (thereafter see Chronic refractory ITP). The frequency and dose should be titrated to maintain a platelet count of at least 30x109/L. The objective of therapy is to maintain a safe platelet count while other therapeutic options are explored.

2. ITP with life-threatening haemorrhage or the potential for life-threatening haemorrhage:

IVIg therapy may be given when conventional doses of corticosteroids have failed or in conjunction with steroids when a rapid response is required.

3. ITP in pregnancy:

  1. Platelets <30x109/L: IVIg therapy may be used to avoid corticosteroids, immunosuppressive agents and splenectomy. Further doses titrated to maintain a platelet count >30x109/L may be administered every three to four weeks throughout the pregnancy.
  2. Impending delivery: IVIg therapy may be used to achieve a platelet count considered safe for delivery (80–100x109/L).

4. Specific circumstances:

  1. Planned surgery: IVIg may be used to achieve a platelet count considered safe for surgery. The safe threshold will vary with the nature of the surgery (Recommended platelet counts for patients without concurrent risks of bleeding: minor dental work >30x109/L, minor surgery >50x109/L, major surgery >80x109/L, major neurosurgery >100x109/L.)
  2. Severe ITP: IVIg may be used where corticosteroids and immunosuppression are contraindicated.
  3. Chronic refractory ITP unresponsive to all other available therapies: These patients may be considered for long-term maintenance therapy with IVIg, subject to regular review by a haematologist.

5. HIV-associated ITP:

  1. Failure of antiretroviral therapy with platelet count <30x109/L;

OR

  1. Life-threatening haemorrhage secondary to thrombocytopenia.
Review criteria for assessing the effectiveness of IVIg use
  • In chronic refractory ITP, six-month review assessing evidence of clinical benefit;
  • Resolution of bleeding;
  • Increment in platelet count.
Dose

Initial therapy: 1–2 g/kg as a single or divided dose.

Ongoing therapy: When indicated, 1–2 g/kg in single or divided dose at 4 to 6 weekly intervals titrated to symptoms and platelet count.

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

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, pp. 42–48. 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. 13–14.

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.

Godeau, B, Caulier, MT, Decuypere, L, et al 1999, ‘Intravenous immunoglobulin for adults with autoimmune thrombocytopenic purpura: results of a randomised trial comparing 0.5 and 1 g/kg b.w.’, British Journal of Haematology, vol. 107, no. 4, pp. 716–9.

Godeau, B, Chevret, S, Varet, B, et al 2002, ’Intravenous immunoglobulin or high-dose methylprednisolone, with or without oral prednisone, for adults with untreated severe autoimmune thrombocytopenic purpura: a randomised, multicentre trial’, Lancet, vol. 359, no. 9300, pp. 23–9.

Godeau, B, Lesage, S, Divine, M, et al 1993, ‘Treatment of adult chronic autoimmune thrombocytopenic purpura with repeated high-dose intravenous immunoglobulin’, Blood, vol. 82, no. 5, pp. 1415–21.

Jacobs, P, Wood, L & Novitzky N 1994, ‘Intravenous gammaglobulin has no advantages over oral corticosteroids as primary therapy for adults with immune thrombocytopenia: a prospective randomised clinical trial’, American Journal of Medicine, vol. 97, no. 1, pp. 55–9.

Kurlander, RJ & Rosse WF 1986, ‘Efficacy of a 2-day schedule for administering intravenous immunoglobulin in treating adults with ITP’, Blood, vol. 68, p. 112A.

Perrella, O 1990, ‘Idiopathic thrombocytopenic purpura in HIV infection: therapeutic possibilities of intravenous immunoglobulins’, Journal of Chemotherapy, vol. 2, no. 6, pp. 390–3.

Provan, D, Stasi, R, Newland, AC, et al 2010, ‘International consensus report on the investigation and management of primary immune thrombocytopenia’, Blood, vol. 115, no. 2, pp. 168–86.

Unsal, C, Gurkan, E, Guvenc, B, et al 2004, ‘Anti-D and intravenous immunoglobulin treatments in chronic idiopathic thrombocytopenic purpura’, Turkish Journal of Haematology, vol. 21, no. 1, pp. 27–32.

Zell, SC & Peterson, K 1997, ‘Long-term remission of HIV- associated thrombocytopenia parallels ongoing suppression of viral replication’, Western Journal of Medicine, vol. 167,no. 6, pp. 433–35.

Medical condition Inflammatory myopathies: polymyositis (PM), dermatomyositis (DM) and inclusion body myositis (IBM)
Indication for IVIg use
  1. Patients with PM or DM with significant muscle weakness unresponsive to corticosteroids and other immunosuppressive agents.
  2. Patients with IBM who have dysphagia affecting function.
  3. Patients with rapidly progressive IBM.
Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria

The inflammatory myopathies are a group of three discrete disorders of skeletal muscle: DM, PM and IBM.

These disorders are acquired and have in common the occurrence of significant muscle weakness and the presence of an inflammatory response within the muscle.

The diagnosis of DM, PM or IBM is usually made by neurologists or rheumatologists, and relies on the combination of careful clinical evaluation, an elevated creatine kinase level, electromyography and muscle biopsy.

Justification for evidence category

PM: The Biotext (2004) review included one prospective case-series study of 35 adults with chronic refractory polymyositis. IVIg may be of benefit in these patients, improve mean muscle power and allow reduction in dose of corticosteroid. Further research is needed.

DM: The Biotext (2004) review included one double-blind, placebo-controlled trial considered of low quality of 15 patients with biopsy-confirmed, treatment-resistant dermatomyositis. IVIg treatment combined with prednisone led to significant improvement in muscle strength and neuromuscular symptoms of patients in the intervention group (n=8).

IBM: The Biotext (2004) review included three small controlled studies, two of which had a crossover design. A total sample of 77 patients diagnosed with IBM was followed for between 4 and 12 months. The three studies showed possible slight benefit in reducing endomysial inflammation, disease progression and severity of IBM. Further research is needed.

One submission reported the effectiveness of IVIg therapy for PM and DM as add-on therapy for patients who have not responded to steroids and immunosuppression (NSW IVIg User Group). A further submission confirms a role for IVIg as add-on maintenance therapy in some patients resulting in an increased chance of complete remission and reduction in corticosteroid dose. A third submission suggests that IVIg can be tried as add-on treatment for patients with PM or DM who have not responded adequately to corticosteroids and second-line immunosuppressive agents (Asia–Pacific IVIg Advisory Board 2004).

Weak evidence suggests that it may benefit patients with dysphagia associated with IBM (Asia–Pacific IVIg Advisory Board 2004).

Qualifying criteria for IVIg therapy

Diagnosis made by a neurologist, rheumatologist or immunologist of:

  1. Patients with PM or DM who have significant muscle weakness or dysphagia and have not responded to corticosteroids and other immunosuppressive agents;

OR

  1. Patients with IBM who have dysphagia affecting function;

OR

  1. Patients with rapidly progressive IBM.
Exclusion criteria for IVIg therapy Expert consensus does not recommend IVIg to treat the limb weakness of IBM
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 a neurologist, rheumatologist, or clinical immunologist is required; frequency as determined by clinical status of patient.

For stable patients on maintenance treatment, review by a specialist 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 (ADL) or quantitative muscle scores or Medical Research Council (MRC) muscle assessment;

OR

  1. Stabilisation of disease as defined by stable functional scores (ADLs) or quantitative muscle scores or MRC muscle assessment 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, 4–6 weekly.

Aim for the minimum dose to maintain optimal functional status.

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

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]

Cherin, P, Pelletier, S, Teixeira, A, et al 2002, ‘Results and long-term follow up of intravenous immunoglobulin infusions in chronic, refractory polymyositis: an open study with thirty-five adult patients’, Arthritis & Rheumatism, vol. 46, no. 2, pp. 467–74.

Choy, EHS, Hoogendijk, JE, Lecky, B, et al 2005, ‘Immunosuppressant and immunomodulatory treatment for dermatomyositis and polymyositis (Cochrane Review)’, in The Cochrane Library, Issue 3, John Wiley & Sons, Ltd, Chichester, UK.

Dalakas, MC 2004, ‘The use of intravenous immunoglobulin in the treatment of autoimmune neuromuscular diseases: evidence-based indications and safety profile’, Pharmacology & Therapeutics, vol. 102, no. 3, pp. 177–93.

Dalakas, MC 2005, ‘Polymyositis, dermatomyositis, and inclusion body myositis’, in DL Kasper, E Braunwald, AS Fauci, et al (eds), Harrison’s Textbook of Medicine, 16th edn, McGraw-Hill, New York, pp. 2540–45.

Dalakas, MC, Illa, I, Dambrosia, JM, et al 1993, ‘A controlled trial of high-dose intravenous immune globulin infusions as treatment for dermatomyositis’, New England Journal of Medicine, vol. 329, no. 27, pp. 1993–2000.

Dalakas, MC, Koffman, B, Fujii, M, et al 2001, ‘A controlled study of intravenous immunoglobulin combined with prednisone in the treatment of IBM’, Neurology, vol. 56, no. 3, pp. 323–7.

Dalakas, MC, Sonies, B, Dambrosia, J, et al 1997, ‘Treatment of inclusion body myositis with IVIg: a double-blind, placebo-controlled study’, 7, vol. 48, no. 3, pp. 712–6.

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.

Walter, MC, Lochmuller, H, Toepfer, M, et al 2000, ‘High-dose immunoglobulin therapy in sporadic inclusion body myositis: a double-blind, placebo-controlled study’, Journal of Neurolology, vol. 247, no. 1, pp. 22–8.

Wiles, CM, Brown, P, Chapel, H, et al 2002, ‘Intravenous immunoglobulin in neurological disease: a specialist review’, Journal of Neurology, Neurosurgery and Psychiatry, vol. 72, no. 4, pp. 440–8.

Medical condition Kawasaki disease (mucocutaneous lymph node syndrome)
Indication for IVIg use Early in Kawasaki disease to prevent coronary artery pathology.
Level of evidence Clear evidence of benefit (Category 1).
Description and diagnostic criteria

Kawasaki disease is an acute, febrile, multi-system disease of children and young infants often involving the coronary arteries. Coronary artery aneurysms may occur from the second week of illness during the convalescent stage.

The cause of the condition is unknown but there is evidence that the characteristic vasculitis results from an immune reaction characterised by T-cell and macrophage activation to an unknown antigen, secretion of cytokines, polyclonal B-cell hyperactivity, and the formation of autoantibodies to endothelial cells and smooth muscle cells. It is likely that in genetically susceptible individuals, one or more uncharacterised common infectious agents, possibly with super-antigen activity, may trigger the disease.

Diagnosis

A diagnosis of Kawasaki disease is generally made if fever of four or more days’ duration is associated with at least four of the following changes, which often appear sequentially:

  • bilateral (non-purulent) conjunctival injection;
  • changes of the mucous membranes of the upper respiratory tract and oropharynx, including diffuse redness of pharyngeal mucosa, dry fissured lips, red fissured lips, and/or ‘strawberry tongue’;
  • changes of the extremities, including peripheral erythema, peripheral oedema, and subsequent periungual or more generalised desquamation;
  • polymorphous rash;
  • cervical lymphadenopathy.

A diagnosis of Kawasaki disease may be made if fever and fewer than four of the changes listed above are present where there is strong clinical suspicion of Kawasaki disease (refer to Newburger 2004). Between 10% and 20% of cases, particularly in younger infants, present with fever and fewer than four of the listed criteria. Expert advice should be sought.

Data support the use of IVIg while there is ongoing inflammation (usually taken as ongoing fever or raised acute inflammatory markers). Prognosis is worse if IVIg is used 10 days post-onset, but should be used at any time if there is evidence of inflammation. Up to 15% of patients do not respond to initial IVIg therapy. Consensus is for re-treatment with 2 g/kg of IVIg before considering steroids.

Justification for evidence category One high-quality systematic review of 16 RCTs that showed that IVIg is of benefit in treating Kawasaki disease (Biotext 2004).
Qualifying criteria for IVIg therapy Clinical diagnosis of Kawasaki disease by a paediatrician or immunologist.
Dose

2 g/kg in a single dose over 10–12 hours unless cardiac function necessitates the administration of a prolonged or divided treatment dose, usually once only.

Re-treatment with 2 g/kg in a single dose may be given when there is ongoing inflammation.

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. 255–6. Available from:anba.gov.au/pubs.htm [cited 7 Dec 2007]

Burns, JC & Glode, MP 2004, ‘Kawasaki syndrome’, Lancet, vol. 364, no. 9433, pp. 533–44.

De Zorzi, A, Colan, SD, Gauvreau, K, et al 1998, ‘Coronary artery dimensions may be misclassified as normal in Kawasaki disease’, Journal of Pediatrics, vol. 133, no. 2, pp. 254–8.

Durongpisitkul, K, Soongswang, J, Laohaprasitiporn, D, et al 2003, ‘Immunoglobulin failure and retreatment in Kawasaki disease’, Paediatric Cardiology, vol. 24, no. 2, pp. 145–8.

Feigin, RD, Cecchin, F & Wissman, SD 2006, ‘Kawasaki disease’, in JA McMillan (ed.), Oski’s paediatrics: principles and practice, 4th edn, Lippincott Williams & Wilkins, Philadelphia, pp. 1015–20.

Newburger, JW, Takahashi, M, Gerber, MA, et al 2004, ‘Diagnosis, treatment, and long-term management of Kawasaki disease: a statement for health professionals from the Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease, Council on Cardiovascular Disease in the Young, American Heart Association’, Paediatrics, vol. 114, no. 6, pp. 1708–33.

Oates-Whitehead, RM, Baumer, JH, Haines, L, et al 2003, ‘Intravenous immunoglobulin for the treatment of Kawasaki disease in children (Cochrane Review)’, in The Cochrane Library, Issue 4, John Wiley & Sons, Ltd, Chichester, UK.

Rosenfeld, EA, Shulman, ST, Corydon, KE, et al 1995, ‘Comparative safety and efficacy of two immune globulin products in Kawasaki disease’, Journal of Paediatrics, vol. 126, no. 6, pp. 1000–3.

Stiehm, ER 2006, ‘Lessons from Kawasaki disease: all brands of IVIg are not equal’, Journal of Paediatrics, vol. 148, pp. 6–8.

Tsai, MH, Huang, YC, Yen, MH, et al 2006, ‘Clinical responses of patients with Kawasaki disease to different brands of intravenous immunoglobulin’, Journal of Paediatrics, vol. 148, no. 1, pp. 38–43.

Wang, CL, Wu, YT, Liu, CA, et al 2005, ‘Kawasaki disease: infection, immunity and genetics’, The Pediatric Infectious Disease Journal, vol. 24, no. 11, pp. 998–1004.

Medical condition Lambert–Eaton myasthenic syndrome (LEMS)
Indication for IVIg use Short-term therapy for severely affected nonparaneoplastic LEMS patients.
Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria

LEMS is a disorder of neuromuscular transmission first recognised clinically in association with lung cancer and subsequently in cases in which no neoplasm was detected.

Patients with LEMS have a presynaptic neuromuscular junction defect. The clinical picture is characterised by proximal muscle weakness with augmentation of strength after exercise, mild oculomotor signs, depressed deep tendon reflexes and autonomic dysfunction (dry mouth, constipation, erectile failure).

Justification for evidence category

In the Biotext (2004) review, one systematic review (containing one RCT with 9 patients) and one case series of 7 patients with a crossover design were included. IVIg appeared to provide some benefit to patients with LEMS. However, both studies only included a small number of patients.

Expert consensus states that IVIg produces temporary improvement in patients with LEMS. It therefore has a role as second line therapy (Asia–Pacific IVIg Advisory Board 2004).

One submission to the National Blood Authority reported on a randomised controlled trial that showed significant improvement in strength associated with a decline in the level of pathogenic antibodies (NSW IVIg User Group).

Qualifying criteria for IVIg therapy
  1. Mandatory assessment by a neurologist;

AND

  1. Severely affected nonparaneoplastic LEMS patients in whom other therapy (e.g. with 3,4-diaminopyridine) has failed.
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. Initial review three to six monthly.

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 (ADL) or quantitative muscle scores or Medical Research Council (MRC) muscle assessment;

OR

  1. Stabilisation of disease as defined by stable functional scores (ADLs) or quantitative muscle scores or MRC muscle assessment 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, 2–6 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

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]

Bain, PG, Motomura, M, Newsom-Davis, J, et al 1996, ‘Effects of intravenous immunoglobulin on muscle weakness and calcium- channel autoantibodies in the Lambert-Eaton myasthenic syndrome’, Neurology, vol. 47, no. 3, pp. 678–83.

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. 184–7. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf.

Dalakas, MC 2004, ‘The use of intravenous immunoglobulin in the treatment of autoimmune neuromuscular diseases: evidence- based indications and safety profile’, Pharmacology & Therapeutics, vol. 102, no. 3, pp. 177–93.

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.

Maddison, P & Newsom-Davis, J 2005, ‘Treatment for Lambert- Eaton myasthenic syndrome (Cochrane Review)’, in The Cochrane Library, Issue 2, John Wiley & Sons, Ltd, Chichester, UK.

Motomura, M, Bain, PG, et al 1995, ‘Effects of intravenous immunoglobulin treatment on anti-calcium channel antibody titres in the Lambert-Eaton myasthenic syndrome’, Journal of Neurology, vol. 242, p. S44.

Skeie, GO, Apostolski, S, Evoli, A, et al 2006, ‘Guidelines for the treatment of autoimmune neuromuscular transmission disorders’, European Journal of Neurology, vol. 13, no. 7, pp. 691–9.

Medical condition Multifocal motor neuropathy (MMN)
Indication for IVIg use First-line therapy for MMN.
Level of evidence Clear evidence of benefit (Category 1).
Description and diagnostic criteria

MMN is a relatively rare disorder characterised by slowly progressive, asymmetric, predominately distal limb weakness without sensory impairment. Weakness often begins in the arms and the combination of weakness, wasting, cramps and fasciculations may suggest a diagnosis of motor neuron disease. However, clinical examination may demonstrate that the pattern of weakness follows the distribution of individual nerves rather than a spinal segmental pattern.

Investigations will typically show conduction block on nerve conduction studies. IgM anti-GM-1 antibodies have been reported in a large number of patients with MMN and provide confirmatory evidence but are not essential for the diagnosis.

Justification for evidence category

The Biotext (2004) review found six low-quality case studies or crossover RCTs with a total sample size of 68 patients. A possible benefit of IVIg treatment in these patients was observed, although five studies were not controlled.

The Frommer and Madronio (2006) review found one high-quality systematic review (a Cochrane review) of four crossover RCTs with 34 patients. Evidence for improvement in muscle strength with IVIg and limited evidence of a reduction in disability after IVIg administration.

Consensus statements assert that IVIg is the only safe treatment demonstrated to work in patients with MMN. It is recommended in those who have significant disability. Dose and monitoring is similar to chronic inflammatory demyelinating polyneuropathy. IVIg therapy is usually long term, but the minimum effective dose for each patient should be sought.

Plasma exchange and steroids appear to cause a worsening in the condition of patients with MMN with conduction block. Regular maintenance doses of IVIg are needed.

The National Guideline Clearinghouse recommends the use of IVIg in the treatment of patients with progressive, symptomatic MMN that has been diagnosed using electrophysiology, ruling out other possible conditions that may not respond to IVIg treatment.

Qualifying criteria for IVIg therapy Patients who have multifocal motor neuropathy, with a typical clinical phenotype, with or without persistent conduction block, as diagnosed by a neurologist.
Exclusion criteria for IVIg therapy
  • Presence of upper motor neuron signs.
  • Significant sensory impairment without an adequate alternative explanation.
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. Most individuals will respond within three months unless there is significant axonal degeneration whereby a six-month course will be necessary. 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

  1. 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: 0.4–2 g/kg, 2–6 weekly. The amount per dose should be titrated to the individual’s response.

Aim for the minimum dose to maintain optimal functional status.

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

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

European Federation of Neurological Societies, Peripheral Nerve Society, van Schaik, IN, Bouche, P, et al 2006, ‘European Federation of Neurological Societies/Peripheral Nerve Society guideline on management of multifocal motor neuropathy’, European Journal of Neurology, vol. 13, pp. 802–8.

Federico, P, Zochodne, DW, Hahn, AF, et al 2000, ‘Multifocal motor neuropathy improved by IVIg: randomized, double-blind, placebo- controlled study’ (comment), Neurology, vol. 55, no. 9, pp. 1256–62.

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. 35–7.

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. 30–4.

Van den Berg-Vos, RM, Franssen, H, Wokke, JH, et al 2002, ‘Multifocal motor neuropathy: long-term clinical and electrophysiological assessment of intravenous immunoglobulin maintenance treatment’, Brain, vol. 125, pt 8, pp. 1875–86.

Van Schaik, IN, van den Berg, LH, de Haan, R, et al 2005, ‘Intravenous immunoglobulin for multifocal motor neuropathy (Cochrane Review)’, in The Cochrane Library, Issue 2, John Wiley & Sons, Ltd, Chichester, UK.

Medical condition Myasthenia gravis (MG)
Indication for IVIg use
  1. As an alternative treatment to plasma exchange in acute exacerbation (myasthenic crisis) or before surgery and/or thymectomy.
  2. As maintenance therapy for moderate to severe MG when other treatments have been ineffective or caused intolerable side effects.
Level of evidence Clear evidence of benefit (Category 1).
Description and diagnostic criteria

MG is an autoimmune disease associated with the presence of antibodies to acetylcholine receptors (AChR) or to muscle-specific tyrosine kinase (MuSK) at the neuromuscular junction. Some patients with myasthenia gravis are antibody negative.

Clinical features are characterised by fluctuating weakness and fatigability of voluntary muscles, namely levator palpebrae, extraocular, bulbar, limb and respiratory muscles. Patients usually present with unilateral or bilateral drooping of eyelid (ptosis), double vision (diplopia), difficulty in swallowing (dysphagia) and proximal muscle weakness. Weakness of respiratory muscles can result in respiratory failure in severe cases or in acute severe exacerbations (myasthenic crisis).

Diagnosis is suspected based on the clinical picture described above, without loss of reflexes or impairment of sensation. Repetitive nerve stimulation typically shows a decreasing response at 2–3 Hz, which repairs after brief exercise (exercise facilitation). Edrophonium can be used for confirmation. Other useful investigations include serum anti-AChR or MuSK antibody titre, or SFEMG (single-fibre electromyography).

Justification for evidence category

A Cochrane review of four RCTs (a total of 147 children and adult patients) found benefit but no significant difference between IVIg and plasma exchange, and no significant difference between IVIg and methylprednisolone. One of the four studies found no benefit of IVIg (i.e. no significant difference between IVIg and placebo). The individual trials were of poor design and some had small numbers of participants, so more research is needed (Biotext 2004).

Anecdotal evidence of efficacy has come from clinicians. There is insufficient placebo-controlled evidence for IVIg use as a steroid-sparing agent or before thymectomy in stable MG, although multiple case reports suggest benefit in this context.

The Asia–Pacific Advisory Group (2004) supports the use of IVIg over a single day for the treatment of myasthenia gravis exacerbations, in myasthenic crisis, or in patients with severe weakness poorly controlled with other agents. It does not support the use of IVIg for maintenance in stable moderate or severe MG unless other therapies have failed and IVIg has shown benefit.

Effectiveness of IVIg is equivalent to steroids and plasma exchange but IVIg may be easier to administer than plasma exchange and avoids the side effects of steroids.

Qualifying criteria for IVIg therapy

Mandatory diagnosis and assessment by a neurologist;

AND

  1. As an alternative treatment to plasma exchange in acute exacerbation (myasthenic crisis) or before surgery and/or thymectomy;

OR

  1. As maintenance therapy for moderate to severe MG when other treatments have been ineffective or caused intolerable side effects.
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. Initial review three to six monthly.

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 improvement in fatigability and weakness.

Various scores can be used, including:

  • forward arm abduction time (up to a full five minutes);
  • Quantitative Myasthenia gravis score (Duke);
  • respiratory function (e.g. forced vital capacity); quantitative dynamometry of proximal limb muscles;
  • variation of a myasthenic muscular score (MSS).
Dose

Maintenance: 0.4–1 g/kg, 4–6 weekly.

Induction or before surgery, or during myasthenic crisis: 1–2 g/kg in 2 to 5 divided doses.

Aim for minimum dose to maintain optimal functional status.

Note: smaller dosage may be of greater efficacy.

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. 188–9. Available from: http://www.nba.gov.au/pubs/pdf/report-lit-rev.pdf.

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.

Gajdos, P, Chevret, S & Toyka, K 2003, ‘Intravenous immunoglobulin for myasthenia gravis (Cochrane Review)’, in The Cochrane Library, Issue 2, John Wiley & Sons, Ltd, Chichester, UK.

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.

Medical condition Neonatal haemochromatosis (NH)
Indication for IVIg use Pregnant women who have had a previous pregnancy affected by neonatal haemochromatosis.
Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria

NH manifests in the foetus and newborn and is characterised by abnormal accumulation of iron in the liver and extra-hepatic tissues. Affected neonates present with fulminant liver failure, usually in the context of a history of prematurity, intrauterine growth retardation and oligohydramnios. NH differs from most other causes of neonatal liver disease, other than congenital infections, in that the condition begins in utero and fulminant liver disease is manifested in the first few days of life. The aetiology and pathogenesis remains uncertain. The NH phenotype may be the outcome of numerous disease processes. There is also evidence, however, that NH is an alloimmune disorder. First, there is an approximate 80% likelihood of NH once a woman has an affected baby. Second, mothers can have affected babies with different fathers. It has not been described that fathers can have affected half-siblings with different mothers.

Symptoms and signs

Affected neonates present with signs of liver failure, including extreme cholestasis, hypoalbuminaemia, coagulopathy, ascites and hypoglycaemia.

Diagnosis of neonatal haemochromatosis is made after other causes of neonatal liver failure have been ruled out.

In addition to extensive iron deposition (siderosis), liver biopsy would show cirrhosis with diffuse fibrosis, bile duct proliferation, and giant cells. Siderosis is also present in other tissues and viscera (e.g. epithelial tissues and the heart) but not in reticuloendothelial cells.

Occurrence

NH is a rare disease but the rate of recurrence after the index case in a sibship is up to 80%.

Prognosis

About 20% survival with medical treatment.

Justification for evidence category A trial compared the impact of IVIg on pregnancy outcome of women whose most recent pregnancy had resulted in NH with historical controls (randomly selected previously affected pregnancies). All 15 pregnancies resulted in live births. NH was diagnosed in 11 but responded to medical treatment. By contrast, there were 2 successful outcomes in controls (Biotext 2004).
Qualifying criteria for IVIg therapy Women who are pregnant or attempting to conceive and their most recent pregnancy ended in delivery of a foetus shown to have had NH.
Review criteria for assessing the effectiveness of IVIg use
  • Occurrence of NH, or evidence of liver disease (serum ferritin and a-fetoprotein levels, coagulopathy) in the offspring of women who have previously given birth to an NH-affected neonate.
  • Requirement for liver transplantation in these neonates.
  • Survival and development of infants following maternal IVIg therapy during pregnancy.
Dose

1 g/kg body weight weekly from the 18th week until the end of gestation.

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

Flynn, DM, Mohan, N, McKiernan, P, et al 2003, ‚Progress in treatment and outcome for children with neonatal haemochromatosis’, Archives of Disease in Childhood – Foetal Neonatal Edition, vol. 88, no. 2, pp. F124–7.

Knisely, AS, Mieli-Vergani, G & Whitington, PF 2003, ‘Neonatal haemochromatosis’, Gastroenterology Clinics of North America, vol. 32, no. 3, pp. 877–89, vi–vii.

Rodriguez, F, Kallas, M, Nash, R, et al 2005, ‘Neonatal haemochromatosis – medical treatment vs. transplantation: the King’s experience’, Liver Transplantation, vol. 11, no. 11, pp. 1417–24.

Schneider, BL 1996, ‘Neonatal liver failure’, Current Opinion in Paediatrics, vol. 8, pp. 495–501.

Whittington, PF & Hibbard, JU 2004, ‘High-dose immunoglobulin during pregnancy for recurrent neonatal haemochromatosis’, Lancet, vol. 364, pp. 1690–8.

Whittington, PF, Kelly, S & Ekong, UD 2005, ‘Neonatal haemochromatosis: foetal liver disease leading to liver failure in the foetus and newborn’, Paediatric Transplantation, vol. 9, pp. 640–5.

Medical condition Stiff person syndrome (Moersch–Woltmann syndrome)
Indication for IVIg use Treatment of significant functional impairment in patients who have a verified diagnosis of stiff person syndrome.
Level of evidence Evidence of probable benefit (Category 2a).
Description and diagnostic criteria

Patients with stiff person syndrome present with symptoms related to muscular rigidity and superimposed episodic spasms. The rigidity insidiously spreads involving axial muscles, primarily abdominal and thoracolumbar, as well as proximal limb muscles. Typically, co-contraction of truncal agonist and antagonistic muscles leads to a board-like appearance with hyperlordosis. Less frequently, respiratory muscle involvement leads to breathing difficulty and facial muscle involvement to a mask-like face.

Investigations that may be useful for diagnosis include auto-antibodies to GAD-65 or GAD-67, electromyography recordings from stiff muscles that may show continuous discharges of motor unit, and cerebrospinal fluid oligoclonal bands.

Justification for evidence category

The Biotext (2004) review included one randomised, double blind, placebo-controlled trial with a crossover design of 16 patients with stiff person syndrome and anti-GAD-65 antibodies. A significant treatment effect with IVIg was seen, resulting in patients’ decreased stiffness and heightened sensitivity scores.

According to expert consensus, considering the disabling progressive course of stiff person syndrome, IVIg should be offered as the first-line treatment. Although periodic infusions would be required in the majority, further studies are needed to determine optimal dosage and duration (Asia–Pacific Advisory Board 2004).

Qualifying criteria for IVIg therapy Significant functional impairment in patients who have a verified diagnosis of stiff person syndrome made by a neurologist.
Review criteria for assessing the effectiveness of IVIg use.

Review

Regular review by a 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 stiffness, including:

  • improvement or stabilisation of activities of daily living scores;
  • other specialised scoring systems, such as distribution-of-stiffness index and heightened sensitivity scale.
Dose

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

Maintenance: 1–2 g/kg, 4–6 weekly.

Aim for the minimum dose to maintain optimal functional status.

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. 190–1. Available from: www.nba.gov.au/pubs.htm [cited 7 Dec 2007]

Dalakas, MC 2005, ‘The role of IVIg in the treatment of patients with stiff person syndrome and other neurological diseases associated with anti-GAD antibodies’, Journal of Neurology, vol. 252, suppl. 1, pp. 119–25.

Dalakas, MC, Fujii, M, Li, M, et al 2001, ‘High-dose intravenous immune globulin for stiff-person syndrome’ [see comment], New England Journal of Medicine, vol. 345, no. 26, pp. 1870–6.

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. 70–2.

Rowland, LP & Layzer RB 2005, ‘Stiff man syndrome (Moersch– Woltman syndrome)‘, in LP Rowland (ed.), Merritt’s Neurology, 11th edn, Lippincott Williams & Wilkins, Philadelphia, p. 927.