A study protocol for a multicentre randomised clinical trial evaluating the safety and feasibility of a bioengineered human allogeneic nanostructured anterior cornea in patients with advanced corneal trophic ulcers refractory to conventional treatment

Miguel González-Andrades, Rosario Mata, María Del Carmen González-Gallardo, Santiago Medialdea, Salvador Arias-Santiago, Juliana Martínez-Atienza, Antonio Ruiz-García, Lorena Pérez-Fajardo, Antonio Lizana-Moreno, Ingrid Garzón, Antonio Campos, Miguel Alaminos, Gloria Carmona, Natividad Cuende, Miguel González-Andrades, Rosario Mata, María Del Carmen González-Gallardo, Santiago Medialdea, Salvador Arias-Santiago, Juliana Martínez-Atienza, Antonio Ruiz-García, Lorena Pérez-Fajardo, Antonio Lizana-Moreno, Ingrid Garzón, Antonio Campos, Miguel Alaminos, Gloria Carmona, Natividad Cuende

Abstract

Introduction: There is a need to find alternatives to the use of human donor corneas in transplants because of the limited availability of donor organs, the incidence of graft complications, as well as the inability to successfully perform corneal transplant in patients presenting limbal deficiency, neo-vascularized or thin corneas, etc. We have designed a clinical trial to test a nanostructured fibrin-agarose corneal substitute combining allogeneic cells that mimics the anterior human native cornea in terms of optical, mechanical and biological behaviour.

Methods and analysis: This is a phase I-II, randomised, controlled, open-label clinical trial, currently ongoing in ten Spanish hospitals, to evaluate the safety and feasibility, as well as clinical efficacy evidence, of this bioengineered human corneal substitute in adults with severe trophic corneal ulcers refractory to conventional treatment, or with sequelae of previous ulcers. In the initial phase of the trial (n=5), patients were sequentially recruited, with a safety period of 45 days, receiving the bioengineered corneal graft. In the second phase of the trial (currently ongoing), subjects are block randomised (2:1) to receive either the corneal graft (n=10), or amniotic membrane (n=5), as the control treatment. Adverse events, implant status, infection signs and induced neovascularization are evaluated as determinants of safety and feasibility of the bioengineered graft (main outcomes). Study endpoints are measured along a follow-up period of 24 months, including 27 post-implant assessment visits according to a decreasing frequency. Intention to treat, and per protocol, and safety analysis will be performed.

Ethics and dissemination: The trial protocol received written approval by the corresponding Ethics Committee and the Spanish Regulatory Authority and is currently recruiting subjects. On completion of the trial, manuscripts with the results of phases I and II of the study will be published in a peer-reviewed journal.

Trial registration: CT.gov identifier: NCT01765244 (Jan2013). EudraCT number: 2010-024290-40 (Dec2012).

Keywords: corneal transplantation; corneal ulcer; limbal stem cell deficiency; randomized controlled trial; stromal fibrosis; tissue bioengineering.

Conflict of interest statement

Competing interests: Dr. González-Andrades, Dr. Alaminos and Dr. Campos are inventors of issued patents P200930625 and P200930943, broadly relevant to the work. Remaining authors declare that they have no competing interests.

© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2017. All rights reserved. No commercial use is permitted unless otherwise expressly granted.

Figures

Figure 1
Figure 1
Trial design and stopping rules. Within phase I of the trial, the first five eligible patients were recruited sequentially with a safety period of 45 days between each other, receiving the bioengineered corneal graft (no randomisation). When all five subjects completed a 3 month post-implantation follow-up period, safety and feasibility data generated were analysed by the trial’s Data Safety Monitoring Committee (interim analysis), according to the trial’s stopping rule. Subject enrolment was re-activated within the phase II of the study, and the remaining fifteen patients that complete the study sample size are currently being recruited and randomly allocated 2:1 to receive either the bioengineered cornea (n=10), or aminotic membrane transplantation (n=5), selected as the control treatment for trophic corneal ulcers in advanced stages. All subjects recruited in the trial will complete a 24 months follow-up period.
Figure 2
Figure 2
Study visits and procedures. Clinical trial visits are structured in 31 time points, that include three pre-implantation visits (screening, randomization and pre-implantation) and 27 post-implantation evaluation visits according to a decreasing frequency rate: daily (week 1), weekly (month 1), fortnightly (months 2 and 3), monthly (months 4 to 12), and quarterly (months 13 to 24). The procedures and assessments performed are detailed for each visit.

References

    1. Resnikoff S, Pascolini D, Etya’ale D, et al. . Global data on visual impairment in the year 2002. Bull World Health Organ 2004;82:844–51.
    1. Pascolini D, Mariotti SP. Global estimates of visual impairment: 2010. Br J Ophthalmol 2012;96:614–8. 10.1136/bjophthalmol-2011-300539
    1. Oliva MS, Schottman T, Gulati M. Turning the tide of corneal blindness. Indian J Ophthalmol 2012;60:423–7. 10.4103/0301-4738.100540
    1. Garg P, Krishna PV, Stratis AK, et al. . The value of corneal transplantation in reducing blindness. Eye 2005;19:1106–14. 10.1038/sj.eye.6701968
    1. Gain P, Jullienne R, He Z, et al. . Global survey of corneal transplantation and Eye Banking. JAMA Ophthalmol 2016;134:167–73. 10.1001/jamaophthalmol.2015.4776
    1. Dua HS, Azuara-Blanco A. Corneal allograft rejection: risk factors, diagnosis, prevention, and treatment. Indian J Ophthalmol 1999;47:3–9.
    1. Brunette I, Roberts CJ, Vidal F, et al. . Alternatives to eye bank native tissue for corneal stromal replacement. Prog Retin Eye Res. In Press 2017. 10.1016/j.preteyeres.2017.04.002
    1. Fagerholm P, Lagali NS, Carlsson DJ, et al. . Corneal regeneration following implantation of a biomimetic tissue-engineered substitute. Clin Transl Sci 2009;2:162–4. 10.1111/j.1752-8062.2008.00083.x
    1. Fagerholm P, Lagali NS, Merrett K, et al. . A biosynthetic alternative to human donor tissue for inducing corneal regeneration: 24-month follow-up of a phase 1 clinical study. Sci Transl Med 2010;2:46ra61 10.1126/scitranslmed.3001022
    1. Fagerholm P, Lagali NS, Ong JA, et al. . Stable corneal regeneration four years after implantation of a cell-free recombinant human collagen scaffold. Biomaterials 2014;35:2420–7. 10.1016/j.biomaterials.2013.11.079
    1. Rama P, Matuska S, Paganoni G, et al. . Limbal stem-cell therapy and long-term corneal regeneration. N Engl J Med 2010;363:147–55. 10.1056/NEJMoa0905955
    1. Basu S, Sureka SP, Shanbhag SS, et al. . Simple limbal epithelial transplantation: long-term clinical outcomes in 125 cases of unilateral chronic ocular surface Burns. Ophthalmology 2016;123:1000–10. 10.1016/j.ophtha.2015.12.042
    1. Aravena C, Bozkurt TK, Yu F, et al. . Long-Term Outcomes of the Boston type I Keratoprosthesis in the management of corneal limbal stem cell deficiency. Cornea 2016;35:1156–64. 10.1097/ICO.0000000000000933
    1. Alaminos M, Del Carmen Sánchez-Quevedo M, Muñoz-Avila JI, et al. . Construction of a complete rabbit cornea substitute using a fibrin-agarose scaffold. Invest Ophthalmol Vis Sci 2006;47:3311–7. 10.1167/iovs.05-1647
    1. González-Andrades M, Garzón I, Gascón MI, et al. . Sequential development of intercellular junctions in bioengineered human corneas. J Tissue Eng Regen Med 2009;3:442–9. 10.1002/term.178
    1. Ionescu AM, Alaminos M, de la Cruz Cardona J, et al. . Investigating a novel nanostructured fibrin-agarose biomaterial for human cornea tissue engineering: rheological properties. J Mech Behav Biomed Mater 2011;4:1963–73. 10.1016/j.jmbbm.2011.06.013
    1. Cardona JL, Ionescu AM, Gómez-Sotomayor R, et al. . Transparency in a fibrin and fibrin-agarose corneal stroma substitute generated by tissue engineering. Cornea 2011;30:1428–35. 10.1097/ICO.0b013e31821bdfd4
    1. Fernández-Valadés-Gámez R, Garzón I, Liceras-Liceras E, et al. . Usefulness of a bioengineered oral mucosa model for preventing palate bone alterations in rabbits with a mucoperiostial defect. Biomed Mater 2016;11:015015 10.1088/1748-6041/11/1/015015
    1. Carriel V, Garzón I, Jiménez JM, et al. . Epithelial and stromal developmental patterns in a novel substitute of the human skin generated with fibrin-agarose biomaterials. Cells Tissues Organs 2012;196:1–12. 10.1159/000330682
    1. Carriel V, Garrido-Gómez J, Hernández-Cortés P, et al. . Combination of fibrin-agarose hydrogels and adipose-derived mesenchymal stem cells for peripheral nerve regeneration. J Neural Eng 2013;10:026022 10.1088/1741-2560/10/2/026022
    1. Ionescu AM, de la Cruz Cardona J, González-Andrades M, et al. . UV absorbance of a bioengineered corneal stroma substitute in the 240-400 nm range. Cornea 2010;29:895–8. 10.1097/ICO.0b013e3181ca3650
    1. Mantelli F, Nardella C, Tiberi E, et al. . Congenital corneal anesthesia and neurotrophic keratitis: diagnosis and management. Biomed Res Int 2015;2015:1–8. 10.1155/2015/805876
    1. Nishida T, Yanai R. Advances in treatment for neurotrophic keratopathy. Curr Opin Ophthalmol 2009;20:276–81. 10.1097/ICU.0b013e32832b758f
    1. Bonini S, Rama P, Olzi D, et al. . Neurotrophic keratitis. Eye 2003;17:989–95. 10.1038/sj.eye.6700616
    1. Yanai R, Nishida T, Chikama T, et al. . Potential new modes of treatment of Neurotrophic Keratopathy. Cornea 2015;34(Suppl 11):S121–S127. 10.1097/ICO.0000000000000587
    1. Prabhasawat P, Tesavibul N. Preserved amniotic membrane transplantation for Conjunctival surface reconstruction. Cell Tissue Bank 2001;2:31–9. 10.1023/A:1011597332277
    1. Krachmer JH, Mannis MJ, Holland EJ, et al. . Cornea. St Louis: Elsevier Health Sciences, 2010.
    1. Sotozono C, Ang LP, Koizumi N, et al. . New grading system for the evaluation of chronic ocular manifestations in patients with Stevens-Johnson syndrome. Ophthalmology 2007;114:1294–302. 10.1016/j.ophtha.2006.10.029
    1. Whitcher JP, Shiboski CH, Shiboski SC, et al. . A simplified quantitative method for assessing keratoconjunctivitis sicca from the Sjögren’s Syndrome International Registry. Am J Ophthalmol 2010;149:405–15. 10.1016/j.ajo.2009.09.013
    1. Seitz B, Das S, Sauer R, et al. . Amniotic membrane transplantation for persistent corneal epithelial defects in eyes after penetrating keratoplasty. Eye 2009;23:840–8. 10.1038/eye.2008.140
    1. van Essen TH, Roelen DL, Williams KA, et al. . Matching for Human Leukocyte Antigens (HLA) in corneal transplantation - to do or not to do. Prog Retin Eye Res 2015;46:84–110. 10.1016/j.preteyeres.2015.01.001
    1. Lizana-Moreno A, Guerrero-Calvo J, Fernández-González A, et al. . Optimization of the manufacturing process of an artificial human cornea under GMP guidelines. Cytotherapy 2017;19:S230–S231. 10.1016/j.jcyt.2017.02.330
    1. Mackie IA. Neuroparalytic keratitis : Fraunfelder FRF, Meyer SM, Current ocular therapy. Philadelphia, PA, USA: WB Saunders, 1995.
    1. Chan AW, Tetzlaff JM, Altman DG, et al. . SPIRIT 2013 Statement: defining standard protocol items for clinical trials. Rev Panam Salud Publica 2015;38:506–14.
    1. Schulz KF, Altman DG, Moher D. CONSORT 2010 statement: updated guidelines for reporting parallel group randomised trials. Int J Surg 2011;9:672–7. 10.1016/j.ijsu.2011.09.004

Source: PubMed

3
Abonneren