1,109
Views
0
CrossRef citations to date
0
Altmetric
Editorial

What’s next in gene therapy for Crigler-Najjar syndrome?

ORCID Icon, ORCID Icon & ORCID Icon
Pages 119-121 | Received 27 Sep 2022, Accepted 15 Dec 2022, Published online: 29 Dec 2022

1. Crigler-Najjar syndrome: background and current management

Crigler-Najjar syndrome (CN) is a recessively inherited error of bilirubin metabolism characterized by unconjugated hyperbilirubinemia, due to impaired function of uridine diphosphoglucuronosyl transferase 1A1 (UGT1A1) [Citation1,Citation2]. This enzyme deficiency restricts glucuronidation and subsequent elimination of unconjugated bilirubin (UCB) via bile. In patients with the most severe phenotype, where UGT1A1 activity is completely absent, UCB accumulates in the body to serum levels > 340 uM leading, without adequate treatment, to bilirubin-induced encephalopathy (kernicterus) and death [Citation1,Citation3].

Dependent on phototherapy (PT) for up to 14 hour a day, patients with severe CN can reach adulthood without neurological damage [Citation4]. The evident impact of this burdensome therapy and the stigma that comes with chronic jaundice has a major impact on the quality of life in this patient population, although data to support this is scarce. Currently, liver transplantation is the only curative treatment available for CN. A donor liver fully restores hepatic UGT1A1 activity, normalizing serum bilirubin levels. With a 5-year survival rate of CN patients after liver transplantation in Europe of 92–95% the overall outcome is good [Citation5]. It is mainly the lifetime burden of immunosuppressive therapy and the limited availability of donor livers that warrants the development of novel therapeutic strategies.

2. Gene addition therapy for Crigler-Najjar syndrome

Gene addition therapy aims to correct the dysfunction of a mutated gene by achieving durable expression of a functional copy of the affected gene. Efficient delivery of this so-called transgene to the hepatocyte is a critical step to provide efficient and sustained correction of the disorder in a clinical setting. So far this has only been achieved using viral vectors.

The most successful viral vectors for in vivo liver-directed gene therapy are derived from adeno-associated virus (AAV). Effective AAV-mediated gene therapy has been demonstrated in pre-clinical models and in a clinical setting for hemophilia A and B with positive results which led to the recent conditional approval given by EMA to Roctavian (BioMarin), an AAV5 gene therapy approach for Hemophilia A [Citation6].

In both preclinical models for CN syndrome, a natural mutant rat, and a Ugt1a knockout mouse, AAV-mediated gene addition resulted in life-long correction of serum bilirubin [Citation7–10]. Currently, this approach is being tested in adult CN patients (Anonymized) demonstrating prolonged correction of bilirubin to levels far below those causing neurotoxicity and allowing patients of the high-dose cohort to stop phototherapy (NCT03466463) [Citation11].

3. Gene therapy for Crigler-Najjar disease and rare metabolic diseases in general: what’s next?

The promising therapeutic correction seen after AAV-mediated gene transfer in CN patients supports further development of this treatment strategy. The next step is to overcome the remaining challenges that currently limits its application to pre-selected patients.

A major limitation is the humoral immune response toward AAV vectors. In CN patients, one third of the potential candidates had to be excluded because of the presence of neutralizing antibodies in serum possibly due to natural exposure to wild-type AAV in early childhood [Citation12]. Even the presence of low neutralizing antibody (NAb) titers results in a dramatic reduction of transduction efficacy making this the major exclusion criterion. Depletion of anti-AAV antibodies from serum as well as capsid engineering appeared effective in circumventing preexisting humoral immunity although clinical demonstration is yet to be provided [Citation13]. Recently, a different approach demonstrated feasibility in vivo to create a window of opportunity for AAV transduction in seropositive individuals. Treatment of non-human primates positive for NAb toward AAV with IdeS, a bacterial IgG-cleaving enzyme, prior to AAV administration, resulted in efficient liver transduction [Citation14].

A second limitation to be addressed is the use of AAV-mediated gene therapy in neonates or children suffering from this disease. The rapid loss of episomal AAV vectors upon hepatocyte proliferation hampers long-term treatment efficacy in the fast-growing liver of neonatal and juvenile animals [Citation10]. In addition, treatment with AAV vectors results in the formation of a strong, neutralizing antibody response, rendering subsequent hepatocyte transduction ineffective. This underscores that treatment early after birth may not provide long-term correction. In contrast to rodents, in which the liver growth occurs in a very limited period of 6 weeks, the slower liver growth in humans may lead to lower vector dilution. Interestingly, a recent report indicated persistent expression of the transgene in a 4-year-old MPS VI patient treated with an AAV8 vector expressing arylsulfatase B, suggesting that AAV treatment of young children may provide sustained correction [Citation15].

As stated above, post-treatment humoral immunity also represents an important limitation as it precludes AAV vector re-administration in case of insufficient correction due to under-dosing or loss overtime. Transient immune suppression during the period of exposure of the vector to the immune system, prevented the induction of NAb in naïve Ugt1a1 deficient rats resulting in effective correction of serum bilirubin level upon re-administration of the vector [Citation16].

4. Expert opinion

As the clinical development of gene addition therapy for CN syndrome progresses, preliminary data show safety and efficacy in treated patients. On the other hand, challenges have been identified that currently limits its use to pre-selected patients. Striving to make gene therapy available for all patients suffering from CN syndrome, and other inherited liver disorders alike, the following has to be addressed.

4.1. Limitations caused by neutralizing antibodies toward AAV

IdeS treatment represents the best option to allow effective treatment of CN patients now excluded because of the presence of anti-AAV NAb in serum. Blocking the induction of NAb to the vector capsid upon administration by immune suppression appeared effective in naïve rats but upon re-injection NAb were induced regardless of the immune suppression [Citation16]. Due to prior exposure, most human subjects will not be naïve to AAV. Although immune suppression may not completely prevent NAbs, it can significantly reduce the NAb titers thus making the complete removal by plasmapheresis or by IdeS treatment feasible. However, the combination of IgG depletion and immune suppression may cause vulnerability to infections and the risk-benefit ratio of these combined approaches has to be taken into account.

4.2. Optimal age for of gene therapy

In the first two cohorts of the current gene therapy trial for CN only adult patients were included. Poor adherence to the PT treatment especially during adolescence may increase the risk for bilirubin-induced brain damage before adulthood. Lowering the inclusion age to younger patients in the next cohort therefore is very relevant. In several countries liver transplant at 8 or 9 year is the standard treatment for severe CN syndrome. Performing gene therapy at that age or earlier will improve the life quality of children having CN and their families. Little experience exists in liver directed gene transfer in pediatric populations. Although we cannot exclude the need to administer a higher vector dose to compensate for an eventual vector dilution, a single case report seems to indicate efficacy and stable expression overtime in a 4 year-old patient, supporting a direct translation of the adult treatment in the pediatric population.

4.3. Treating patients with a milder phenotype

Patients receiving a daily dose of PT of >6 hours are currently included. Upon demonstrating long-term safety and efficacy in the current trial, patients suffering from a milder form but dependent on PT may be included in the near future. Since these patients do have residual UGT1A1 activity, it is to be expected that the current vector will result in a more effective correction of serum bilirubin levels in the absence of phototherapy.

4.4. Cost effectiveness of gene therapy

Severely affected CN patients are initially treated by PT, a burdensome but inexpensive treatment. Most of them do need a liver transplant at some point in their life with an estimated cost of about €800.000, which increases rapidly when complications occur. Replacing OLT by AAV gene therapy can be cost effective. Some severely affected patients treated only with PT have reached adulthood without overt brain damage [Citation3]. Although in these patients cost effectiveness will be questionable, it will allow them to have a more normal and productive life.

4.5. Demonstrating long-term efficacy

A single treatment has allowed CN patients to safely stop PT. It is too early to say if this correction will persist life-long but for instance, hemophilia B trials showed persistent correction for more than 10 years. Long-term data for the current ultimate treatment, a liver transplant, also show graft failure in about 20% of patients requiring a second OLT. In contrast, upon loss of correction, patients treated by gene therapy can restart PT to prevent toxic serum bilirubin levels. Most likely, due to the presence of residual UGT1A1 activity a lower PT dose may be sufficient initially.

4.6. Impact on quality of life

Life-long jaundice and the stigma that comes with it, combined with the burden of daily phototherapy has a major impact on the quality of life (QoL) of CN patients. So far, QoL in this patient population has been under-reported and deserves prompt recognition. Improvement of QoL, evaluated using appropriate questionnaires, will be a cornerstone efficacy endpoint in the development of any future treatment for CN.

5. Conclusion

The development of gene addition therapy for Crigler-Najjar syndrome is promising and under way. In the near future this treatment strategy will find its place alongside the curative option of liver transplantation or even replace it when its advantages exceed its limitations. Current efforts are focused on showing safety and efficacy of gene therapy in the first clinical trials as well as resolving challenges that limit its application in a part of the Crigler-Najjar patient population.

Declaration of interest

G Ronzitti is author of patents related to the development of gene therapies for rare metabolic diseases and in particular, of one patent regarding AAV gene therapy for CN syndrome. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was not funded.

References

  • Crigler JF Jr., Najjar VA. Congenital familial nonhemolytic jaundice with kernicterus. Pediatrics. 1952;10(2):169–180.
  • Bosma PJ, Seppen J, Goldhoorn B, et al. Bilirubin UDP-glucuronosyltransferase 1 is the only relevant bilirubin glucuronidating isoform in man. J Biol Chem. 1994;269(27):17960–17964.
  • Aronson SJ, Junge N, Trabelsi M, et al. Disease burden and management of Crigler-Najjar syndrome: report of a world registry. Liver Int. 2022;42(7):1593–1604.
  • Yohannan MD, Terry HJ, Littlewood JM. Long term phototherapy in Crigler-Najjar syndrome. Arch Dis Child. 1983;58(6):460–462.
  • Adam R, Karam V, Delvart V, et al. Evolution of indications and results of liver transplantation in Europe. A report from the European Liver Transplant Registry (ELTR). J Hepatol. 2012;57(3):675–688.
  • Pasi KJ, Rangarajan S, Mitchell N, et al. Multiyear Follow-up of AAV5-hFVIII-SQ gene therapy for hemophilia A. N Engl J Med. 2020;382(1):29–40.
  • Seppen J, Bakker C, de Jong B, et al. Adeno-associated virus vector serotypes mediate sustained correction of bilirubin UDP glucuronosyltransferase deficiency in rats. Mol Ther. 2006;13(6):1085–1092.
  • Ronzitti G, Bortolussi G, van Dijk R, et al. A translationally optimized AAV-UGT1A1 vector drives safe and long-lasting correction of Crigler-Najjar syndrome. Mol Ther Methods Clin Dev. 2016;3:16049.
  • Collaud F, Bortolussi G, Guianvarc’h L, et al. Preclinical development of an AAV8-hUGT1A1 vector for the treatment of Crigler-Najjar syndrome. Mol Ther Methods Clin Dev. 2019;12:157–174.
  • Bortolussi G, Zentillin L, Vanikova J, et al. Life-long correction of hyperbilirubinemia with a neonatal liver-specific AAV-mediated gene transfer in a lethal mouse model of Crigler-Najjar syndrome. Hum Gene Ther. 2014;25(9):844–855.
  • D’Antiga UB L, Brunetti-Pierri N, Baumann U, et al. Adeno-associated virus vector mediated gene therapy for Crigler Najjar syndrome: preliminary report of safety and efficacy from the CareCN clinical trial. J Hepatol. 2021;75:S202.
  • Aronson SJ, Veron P, Collaud F, et al. Prevalence and relevance of pre-existing anti-adeno-associated virus immunity in the context of gene Therapy for Crigler-Najjar syndrome. Hum Gene Ther. 2019;30(10):1297–1305.
  • Gross DA, Tedesco N, Leborgne C, et al. Overcoming the challenges imposed by humoral immunity to AAV Vectors to achieve safe and efficient gene transfer in seropositive patients. Front Immunol. 2022;13:857276.
  • Leborgne C, Barbon E, Alexander JM, et al. IgG-cleaving endopeptidase enables in vivo gene therapy in the presence of anti-AAV neutralizing antibodies. Nat Med. 2020;26(7):1096–1101.
  • Brunetti-Pierri N, Ferla R, Ginocchio VM, et al. Liver-directed adeno-associated virus mediated gene therapy for mucopolysaccharidosis type VI. NEJM Evid. 2022;1(7).
  • Shi X, Aronson SJ, Ten Bloemendaal L, et al. Efficacy of AAV8-hUGT1A1 with Rapamycin in neonatal, suckling, and juvenile rats to model treatment in pediatric CNs patients. Mol Ther Methods Clin Dev. 2021;20:287–297.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.