Key points from the evidence

The content of this evidence summary was up-to-date in December 2015. See summaries of product characteristics (SPCs), British national formulary (BNF), BNF for children (BNFc) or the MHRA or NICE websites for up-to-date information.

Summary

It is difficult to perform studies in rare diseases; therefore, the evidence for using eculizumab to treat C3 glomerulopathy in people with their own, native kidneys is limited. This evidence summary outlines 20 cases that were reported in the literature. Eculizumab improved or stabilised signs of C3 glomerulopathy in 15 cases. An initial response followed by subsequent deterioration was seen in 3 cases, and treatment was ineffective in 2 cases.

Case reports provide only low quality evidence for interventions. Also, it is possible that cases in which eculizumab was unsuccessful are under reported in the literature (publication bias). More evidence is needed to better assess the safety and efficacy of eculizumab in this heterogeneous condition and to determine which patients are most likely to respond treatment.

Regulatory status: Eculizumab (Soliris) is licensed in the UK for treating adults and children with atypical haemolytic uraemic syndrome (aHUS) or paroxysmal nocturnal haemoglobinuria (PNH). Use of eculizumab to treat people with C3 glomerulopathy is off‑label.

Effectiveness

In people with C3 glomerulopathy in their native kidneys:

  • Eculizumab was found to improve or stabilise measures of renal function and/or findings on renal biopsy in 1 person in a small open‑label study (n=6, 3 with native kidneys) and 14 cases. Kidney function did not always return to normal but 4 people stopped dialysis.

  • Renal function improved over 24 weeks but then deteriorated, and biopsy results worsened in 1 person taking eculizumab in the small open‑label study. Similarly, in 1 case an initial response to treatment was seen but not sustained after 2 months and, in another case, mycophenolate had to be reintroduced after 16 weeks.

  • Two cases were reported where eculizumab was ineffective, including 1 in the small open‑label study.

Safety

  • Eculizumab was generally well‑tolerated from the reports of the 20 cases in the literature.

  • According to the summary of product characteristics, the most common adverse effect of eculizumab is headache. The most serious adverse reaction is meningococcal sepsis.

  • Other common adverse effects include aspergillus infection, bacterial and viral infection, thrombocytopenia, leukopenia, haemolysis and anaphylaxis.

Patient factors

  • Vaccination against meningococcal infection is essential before and during treatment with eculizumab. However, vaccination may not be sufficient to prevent infection.

  • Eculizumab is administered by intravenous infusion. The optimal dosage regimen for C3 glomerulopathy is unclear.

  • Assuming eculizumab is found to be effective, long‑term treatment may be required, as recommended for aHUS and PNH.

Resource implications

  • The cost of 1 vial of eculizumab 300 mg concentrate for solution for infusion is £3150.00 excluding VAT (MIMS, November 2015).

  • Based on the dosing regimen used in most of the cases with C3 glomerulopathy, the cost of 5‑week initiation treatment is £50,400 excluding VAT. The annual cost of maintenance treatment with eculizumab 1200 mg is £327,600 excluding VAT.

Introduction

C3 glomerulopathy is a kidney condition characterised by abnormal deposits of complement protein C3 within glomeruli. It is caused by dysregulation of the alternative complement system pathway, which is part of the body's immune system. C3 glomerulopathy is subdivided into C3 glomerulonephritis (C3GN) and dense deposit disease (DDD).

The incidence of C3 glomerulopathy is 1–2 per million population per year. Renal prognosis is poor, with a 30% risk of end stage renal disease at 2 years. After kidney transplantation, the risk of recurrence in the transplanted kidney is over 70%, with more than a 50% chance of graft loss.

The optimal management of people with C3 glomerulopathy is uncertain and agents have not been tested in robust clinical trials, probably because performing randomised controlled trials (RCTs) is difficult in rare diseases. Non-specific immunomodulatory agents such as cyclophosphamide and mycophenolate mofetil are sometimes used. Other treatments which have been used include plasma exchange, rituximab (with or without plasma exchange) and eculizumab.

Full text of introduction.

Product overview

Eculizumab (Soliris, Alexion Pharma UK) is a recombinant humanised monoclonal antibody that binds to complement protein C5, inhibiting its cleavage to C5a (a proinflammatory anaphylatoxin) and C5b and preventing the generation of the membrane attack complex, C5b‑9, which causes cell lysis and death in pathogens.

Full text of product overview.

Evidence review

  • This evidence summary considers the best available evidence for using eculizumab to treat people with C3 glomerulopathy in their own, native kidneys. Use of eculizumab to treat C3 glomerulopathy in people who have had a transplant is considered in the associated evidence summary on prevention of recurrence of C3 glomerulopathy post-transplant: eculizumab.

  • The searches performed for this evidence summary found no RCTs assessing eculizumab for treating people with C3 glomerulopathy in their native kidneys. Limited evidence was found from a small open‑label study (Bomback et al. 2012, n=6 [3 with native kidneys]) and 12 full case reports of 17 single cases. A further 10 reports (15 cases) were identified but are available only as conference abstracts and have been excluded from the evidence summary due to significant limitations in the reporting of findings.

  • Bomback et al. (2012) performed a prospective single‑arm open‑label pilot study of eculizumab for treating 6 people with C3 glomerulopathy, of whom 3 (2 with DDD and 1 with C3GN) had their native kidneys. No statistical analysis was performed because of the small number of participants. Results were mixed. In the first person with DDD, proteinuria improved and remained consistently low throughout treatment, and improvements were seen on renal biopsy. Renal function did not improve in the second person with DDD, who subsequently withdrew from treatment at 40 weeks. In the third person, who had C3GN, renal function improved with eculizumab over the first 24 weeks; however, it worsened again over the second 24 weeks. Deterioration was also seen on biopsy in this person.

  • An initial response to treatment was also seen in a case with DDD reported by Berthe-Aucejo et al. (2014), with improvement in serum albumin and normal sC5b‑9 (soluble membrane attack complex, an indicator of activation of the terminal complement cascade). However, these subsequently deteriorated after 2 months' treatment and proteinuria was unchanged after 3 months' treatment. Similarly, Haffner et al. (2015), reported improvements in serum albumin and proteinuria and normalisation of eGFR in a person with C3GN, but mycophenolate subsequently needed to be reintroduced after 16 weeks following a further increase in proteinuria. Follow‑up biopsy was not reported in these cases.

  • A patient with C3GN described by Besbas et al. (2014) saw no improvements on treatment and still had proteinuria after receiving eculizumab for 10 months. Follow‑up biopsy was not reported.

  • Proteinuria improved and other measures of renal function such as serum creatinine, serum albumin or estimated glomerular filtration rate improved or were stable in 5 cases with C3GN reported by Inman et al. (2015), Kerns et al. (2013), Le Quintrec et al. (2015) (2 cases) and Payette et al. (2015). The case described by Inman et al. (2015) discontinued dialysis. Improvements or stability were seen on renal biopsy in 3 of the cases (Le Quintrec et al. 2015 [2 cases] and Payette et al. 2015). Follow‑up biopsy was not reported in the other 2 cases (Inman et al. 2015 and Kerns et al. 2013).

  • Similarly, proteinuria improved and other measures of renal function improved or stabilised in 9 cases with DDD reported by Daina et al. (2012), Oosterveld et al. (2015) (5 cases), Ozkaya et al. (2014), Rousset-Rouviere et al. (2014) and Vivarelli et al. (2012). Kidney function did not always return to normal, with various degrees of chronic kidney disease remaining. However, 3 cases no longer needed dialysis (Rousset-Rouviere et al. 2014 and Oosterveld et al. 2015 [2 cases]). Improvements were seen on biopsy in the case reported by Vivarelli et al. (2012). No improvements were seen in 1 case in Oosterveld et al. (2015). Follow‑up biopsy was not reported for the other 7 cases.

  • Renal function deteriorated in 6 cases (5 with DDD and 1 with C3GN) when eculizumab was stopped and subsequently improved again when treatment was restarted (Bomback et al. 2012, Oosterveld et al. 2015 [3 cases] Payette et al. 2015 and Vivarelli et al. 2012).

  • The number of case reports was too small to assess adverse effects; however, none were reported in any of the cases.

  • According to the summary of product characteristics, the most common adverse reaction reported in 302 people with PNH and aHUS (the licensed indications) in clinical trials and in postmarketing reports was headache (occurring in more than 1 in 10 people, mostly in the initiation phase). The most serious adverse reaction was meningococcal sepsis (occurring in between 1 in 10 and 1 in 100 people). To reduce the risk of meningococcal infection, all patients must be vaccinated before receiving eculizumab and revaccinated according to current medical guidelines. However, vaccination may not be sufficient to prevent infection. Other common adverse effects seen in between 1 in 10 and 1 in 100 people include aspergillus infection, bacterial and viral infection, thrombocytopenia, leukopenia, haemolysis and anaphylaxis.

  • This evidence review includes information on only 20 people. Case reports are subject to bias and confounding and provide only low quality evidence for interventions. In addition, it is possible that cases in which eculizumab was unsuccessful are under reported in the literature (publication bias).

  • C3 glomerulopathy is a heterogeneous condition associated with many different abnormalities in the alternative complement system pathway, and the degree of C5 convertase dysregulation varies between individuals. Therefore, people may not universally respond to eculizumab therapy. It is currently unclear whether it is possible to identify who will respond to eculizumab treatment using genetic and antibody testing for complement abnormalities. It has been proposed that elevated soluble C5b‑9 levels, an increase in or appearance of C5b‑9 deposits in the kidney, and the presence of marked inflammatory changes on biopsy might be predictors of response to treatment. Longer, larger, statistically powered and adequately controlled studies are needed to better evaluate eculizumab for treating C3 glomerulopathy, in terms of patient‑oriented outcomes such as the need for dialysis or kidney transplantation, adverse effects and quality of life. However, rare diseases present challenges in optimal study design.

  • In people in whom eculizumab is effective, long‑term treatment may be necessary (as recommended in aHUS and PNH) because eculizumab does not address the underlying complement abnormality, but merely prevents downstream formation of C5b‑9. Bomback (2014a) notes that whether the drug is considered lifelong therapy is influenced by the high cost of eculizumab treatment and the potential for infection with prolonged use. Different doses of eculizumab were used in the cases and the optimal regimen for people with recurrence of C3 glomerulopathy is unclear.

Full text of evidence review.

Context and estimated impact for the NHS

The dose of eculizumab used in the majority of cases with C3 glomerulopathy was 900 mg weekly for 4 weeks, then 1200 mg for 1 week and subsequently every 2 weeks.

Based on this dosing regimen, the cost of the 5‑week initiation phase is £50,400 and the cost of 4 weeks' maintenance treatment is £25,200 (excluding VAT, MIMS, November 2015), not including any other costs incurred when eculizumab is, for example, diluted and administered. The annual cost of treatment in the maintenance phase is £327,600 (excluding VAT).

Full text of context and estimated impact for the NHS.

About this evidence summary

'Evidence summaries: unlicensed or off‑label medicines' summarise the published evidence for selected unlicensed or off‑label medicines that are considered to be of significance to the NHS, where there are no clinically appropriate licensed alternatives. The summaries provide information for clinicians and patients to inform their decision‑making and support the construction and updating of local formularies.

The summaries support decision‑making on the use of an unlicensed or off‑label medicine for an individual patient, where there are good clinical reasons for its use, usually when there is no licensed medicine for the condition requiring treatment, or the licensed medicine is not appropriate for that individual.

The strengths and weaknesses of the relevant evidence are critically reviewed within this summary, but this summary is not NICE guidance.