Skip to main content
  • Letter to the Editor
  • Open access
  • Published:

Neutralizing anti-drug antibodies in Fabry disease have no obvious clinical impact?

Abstract

Fabry disease (FD) is a rare X-linked disorder caused by a deficiency of lysosomal α-galactosidase A activity. Treatment with recombinant enzyme replacement therapy is available since 2001 and the effects of anti-drug antibodies (ADA) on therapy efficacy and disease outcome in affected patients have been controversially reported. In this letter we discuss the importance of adequate measurements of neutralizing ADAs and appropriate longitudinal analysis to determine therapy efficiency and clinical outcome in patients with FD.

Letter to the editor

With great interest we read the recent paper by Mauhin and colleagues entitled “Deep characterization of the anti-drug antibodies developed in Fabry disease patients, a prospective analysis from the French multicenter cohort FFABRY” [1]. The authors presented a detailed characterization of anti-drug antibodies (ADAs) in patients with Fabry disease, confirmed their frequency of 40% in men treated with agalsidase and complete cross reactivity against agalsidase alfa and beta and an unambiguous association with lyso-Gb3 considered as a hallmark of disease severity [1]. Nevertheless, they came to the conclusion that “anti-agalsidase antibodies have no obvious clinical impact”. By using IgG isotype-specific ELISA-techniques, the authors demonstrated the humoral IgG-dependent response to ERT. They investigated the associations between IgG subtypes and neutralizing activity, and showed that IgG1, IgG2 and IgG4 subtypes were capable of ERT neutralization in vitro. In our opinion to further substantiate these associations it would have been mandatory to use purified IgG subtypes from patients’ sera and test these fractions separately for ERT neutralization as recently demonstrated for IgG4 [2]. This approach would also have been important since it is known that not all ADAs possess neutralizing activity. Thus, ELISA-based determination of an ADA-positive status is not sufficient to define the clinically relevant impact of ERT neutralization. Of note, the clinical use of neutralization assays to assess the effect of ADAs on clinical and biochemical outcome has repeatedly been demonstrated during the last 15 years [2,3,4,5,6]. Future studies need to be performed to analyze a potential impact of neutralizing and non-neutralizing ADAs on endothelial ERT-uptake.

Furthermore, we agree with Mauhin et al. that the presence of planted ERT-ADA immune complexes in kidneys as reported for other lysosomal storage disease (LSDs) [7,8,9] has not been addressed adequately in recent studies and needs further attention [1]. From the biochemical sight of view the formation of these complexes is probably a rare event, since according to the precipitation curve (Heidelberger curve) of antigen-antibody interaction large non-soluble immune complexes will form when antigens and antibodies are in a balanced state.

In a recent prospective study including longitudinal data, a significant 2.8-fold increased risk for the formation of neutralizing ADA in male patients with FD when treated with agalsidase-beta (1.0 mg/kg every other week) compared to agalsidase-alfa (0.2 mg/kg every other week) was demonstrated [10]. A subgroup analysis of these patients with neutralizing ADAs also revealed a better biochemical response to agalsidase-beta at 1.0 mg/kg in terms of decreasing lyso-Gb3 levels over time [10]. To this respect, we recently demonstrated that antibodies can be saturated and appropriate enzyme dosages can overcome ADA titers during infusions (antigen excess), which resulted in decreased plasma lyso-Gb3 levels [2]. In their cohort, Mauhin and colleagues [1] found no differences between antibody-positive and antibody-negative in non-renal-transplanted patients. Of note, these data are difficult to interpret since the neutralizing impact of the antibodies was not considered, individual slopes were not calculated (only one time-point), and patients with severe renal impairment were excluded from the analysis. The latter point is of specific interest since antibody-positive compared to -negative patients presented with a significantly higher frequency for dialysis and renal transplantation (6 [33.3%] vs 1 [3.7%]; p = 0.012) [1].

Moreover, significantly more antibody-positive patients were treated with agalsidase-beta (n = 14; 77.8%) than with agalsidase-alfa (n = 4; 22.2%; p = 0.0012) [1]. In combination with the missing information on ERT-neutralization status, the 5-fold higher dosage due to agalsidase-beta treatment might have led to the observed effect in that no clinical differences between patients with and without ADAs had been observed.

Therefore, we conclude that neutralizing ADAs affect clinical outcomes and independent of the general assay used for neutralizing ADA measurements and titer determination, the adequate longitudinal analysis of the impact of ADAs on therapy efficiency and clinical outcome in patients with FD can only be performed when analyzed patients are stratified according to supersaturated ADA titers during infusion.

Abbreviations

ADA:

anti-drug antibody

eGFR:

estimated glomerular filtration rate

ELISA:

enzyme-linked immunosorbent assay

ERT:

enzyme replacement therapy

FD:

Fabry disease

GLA:

alpha-galactosidase A

LSD:

lysosomal storage disease

References

  1. Mauhin W, Lidove O, Amelin D, Lamari F, Caillaud C, Mingozzi F, et al. Deep characterization of the anti-drug antibodies developed in Fabry disease patients, a prospective analysis from the French multicenter cohort FFABRY. Orphanet J Rare Dis. 2018;13:127.

    Article  Google Scholar 

  2. Lenders M, Schmitz B, Brand SM, Foell D, Brand E. Characterization of drug-neutralizing antibodies in patients with Fabry disease during infusion. J Allergy Clin Immunol. 2018;141:2289–92.

    Article  Google Scholar 

  3. Linthorst GE, Hollak CE, Donker-Koopman WE, Strijland A, Aerts JM. Enzyme therapy for Fabry disease: neutralizing antibodies toward agalsidase alpha and beta. Kidney Int. 2004;66:1589–95.

    Article  CAS  Google Scholar 

  4. Aerts JM, Groener JE, Kuiper S, Donker-Koopman WE, Strijland A, Ottenhoff R, et al. Elevated globotriaosylsphingosine is a hallmark of Fabry disease. Proc Natl Acad Sci U S A. 2008;105:2812–7.

    Article  CAS  Google Scholar 

  5. Rombach SM, Aerts JM, Poorthuis BJ, Groener JE, Donker-Koopman W, Hendriks E, et al. Long-term effect of antibodies against infused alpha-galactosidase a in Fabry disease on plasma and urinary (lyso)Gb3 reduction and treatment outcome. PLoS One. 2012;e47805:7.

    Google Scholar 

  6. Lenders M, Stypmann J, Duning T, Schmitz B, Brand SM, Brand E. Serum-mediated inhibition of enzyme replacement therapy in Fabry disease. J Am Soc Nephrol. 2016;27:256–64.

    Article  Google Scholar 

  7. Hunley TE. Corzo D, DudekM, Kishnani P, Amalfitano a, Vhen YT, et al. Nephrotic syndrome complicating alpha-glucosidase replacement therapy for Pompe disease. Pediatrics. 2004;114:e532–e5.

    Article  Google Scholar 

  8. Debiec H, Valayannopoulos V, Boyer O, Nöel LH, Callard P, Sarda H, et al. Allo-immune membranous nephropathy and recombinant aryl sulfatase replacement therapy: a need for tolerance induction therapy. J Am Soc Nephrol. 2014;25:675–80.

    Article  CAS  Google Scholar 

  9. Ronco P, Cebiec H. Pathophysical advances in membranous nephropathy: time for a shift in patient’s care. Lancet. 2015;385:1983–92.

    Article  Google Scholar 

  10. Arends M, Biegstraaten M, Wanner C, Sirrs S, Mehta A, Elliott PM, et al. Agalsidase alfa versus agalsidase beta for the treatment of Fabry disease: an international cohort study. J Med Genet. 2018;55:351–8.

    Article  Google Scholar 

Download references

Acknowledgements

We acknowledge support by the Deutsche Forschungsgemeinschaft and Open Access Publication Fund of the University of Muenster.

Funding

There was no specific funding for this manuscript.

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analyzed during the current study.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed equally, ML, BS: design, literature search, discussion, first Draft; SMB, EB: literature search, discussion, critical comments. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Malte Lenders.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

ML received speaker honoraria, travel funding and research grants from Sanofi, Shire, and Amicus Therapeutics. EB received research grants and speaker honoraria from Sanofi, Shire, and Amicus Therapeutics. SMB received speaker honoraria and research grants from Shire. BS has nothing to declare.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lenders, M., Schmitz, B., Brand, SM. et al. Neutralizing anti-drug antibodies in Fabry disease have no obvious clinical impact?. Orphanet J Rare Dis 13, 171 (2018). https://doi.org/10.1186/s13023-018-0916-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13023-018-0916-1

Keywords