Bioengineered corneal tissue for minimally invasive vision restoration in advanced keratoconus in two clinical cohorts

Mehrdad Rafat, Mahmoud Jabbarvand, Namrata Sharma, Maria Xeroudaki, Shideh Tabe, Raha Omrani, Muthukumar Thangavelu, Anthony Mukwaya, Per Fagerholm, Anton Lennikov, Farshad Askarizadeh, Neil Lagali, Mehrdad Rafat, Mahmoud Jabbarvand, Namrata Sharma, Maria Xeroudaki, Shideh Tabe, Raha Omrani, Muthukumar Thangavelu, Anthony Mukwaya, Per Fagerholm, Anton Lennikov, Farshad Askarizadeh, Neil Lagali

Abstract

Visual impairment from corneal stromal disease affects millions worldwide. We describe a cell-free engineered corneal tissue, bioengineered porcine construct, double crosslinked (BPCDX) and a minimally invasive surgical method for its implantation. In a pilot feasibility study in India and Iran (clinicaltrials.gov no. NCT04653922 ), we implanted BPCDX in 20 advanced keratoconus subjects to reshape the native corneal stroma without removing existing tissue or using sutures. During 24 months of follow-up, no adverse event was observed. We document improvements in corneal thickness (mean increase of 209 ± 18 µm in India, 285 ± 99 µm in Iran), maximum keratometry (mean decrease of 13.9 ± 7.9 D in India and 11.2 ± 8.9 D in Iran) and visual acuity (to a mean contact-lens-corrected acuity of 20/26 in India and spectacle-corrected acuity of 20/58 in Iran). Fourteen of 14 initially blind subjects had a final mean best-corrected vision (spectacle or contact lens) of 20/36 and restored tolerance to contact lens wear. This work demonstrates restoration of vision using an approach that is potentially equally effective, safer, simpler and more broadly available than donor cornea transplantation.

Conflict of interest statement

M.R. holds stock and relevant patents in LinkoCare Life Sciences AB, a spin-off company from Linköping University developing BPCDX and related products. He also serves on the board of directors of the company. The terms of his arrangements have been approved by Linköping University in accordance with its policy on objectivity in research. S.T. is business development manager of LinkoCare and serves on the board of directors of LinkoCare Life Sciences AB, as an unpaid board member. N.L., M.J. and N.S. serve on the scientific advisory board of LinkoCare Life Sciences AB, as unpaid advisory board members. Other co-authors have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Biomaterial properties of BPCDX.
Fig. 1. Biomaterial properties of BPCDX.
a, Appearance of BPCDX, indicating transparency and refractive nature of the curved device. b, Light transmission through 550-µm-thick samples of BPCDX, single-crosslinked BPC and the human cornea. The human cornea contains a layer of epithelial cells which absorb UV light, whereas the bioengineered materials are cell-free. Data shown represent mean and standard deviation of measurements from three independent samples. c, Mechanical properties of BPCDX relative to single-crosslinked BPC and previously published data of bioengineered constructs made from porcine collagen,, with human cornea reference values included for comparison. Data values for BPCDX represent mean and standard deviation of measurements from 22 independent samples per test (taken across different production batches, 550-µm-thick ‘dog-bone’ specimens). d, Scanning electron microscope images of the surface and bulk (cross-section) structure of BPCDX and a porcine cornea, indicating tightly packed collagen fibrils in BPCDX with diameter slightly thicker than the native porcine cornea (representative images from three samples per cornea type with similar results). e, Degradation of BPCDX, single-crosslinked BPC and a human donor cornea in 1 mg ml−1 collagenase (data represent mean and standard deviation of measurements from three independent samples for bioengineered materials (550 µm thick, 12 mm diameter) and two independent samples of human donor cornea). f, HCE-2 human corneal epithelial cell attachment and growth on BPCDX relative to the control culture plate surface after 16 days of culture. Cells adhered to BPCDX, with NucBlue staining indicating nuclei and morphology of live, viable cells in brightfield mode. BPCDX had greater cell density than cell culture plasticware (three control samples, six BPCDX samples; error bars represent mean and standard deviation, P = 0.003, two-sided independent t-test). Scale bars, 100 µm. Source Data
Fig. 2. Results 6 months after intrastromal…
Fig. 2. Results 6 months after intrastromal BPCDX implantation in minipigs.
a, OCT images of the central 6 mm of cornea and corresponding photographs of operated eyes indicated localized thinning and loss of transparency in the access cut region in both groups (arrows in OCT scans and photographs). In the autograft group, one cornea (central image) exhibited loss of transparency, while another had partial loss of transparency (bottom image). In both cases, the implantation zone was skewed towards the limbus. The remaining three eyes were transparent with good thickness and only minor thinning at the access cut. In the BPCDX group, two eyes (second image from top, and bottom image) had a partially reduced transparency. In both cases, the implanted zone was skewed towards the limbus. In all eyes, transparency outside the access cut region was maintained. b, Pachymetry maps indicating corneal thickness 6 months post-operatively with color coding of thickness indicated by the grading scale, and mean thickness in µm indicated in each sector. BPCDX corneas exhibited similar thickness as the native porcine cornea. The native porcine cornea is shown for comparison purposes at the bottom of a,b. c, Table indicating pre-operative and post-operative mean corneal thickness and difference in corneal thickness in the central 2 mm zone as determined by OCT. Absence of positive fluorescein staining indicates complete epithelial wound healing. d, In vivo confocal microscopy images of porcine corneas at 6 months. In both groups, the epithelial cell mosaic appeared intact. Basal epithelium and sub-basal nerves (white arrows) were also observed, indicating preservation of the nerve plexus owing to minimal trauma during surgery. Anterior stromal nerves and keratocytes had normal morphology in both groups. The mid-stromal region appeared normal in autografts but BPCDX was devoid of keratocytes, except for individual cellular features (black arrows). Posterior stromal keratocytes and endothelial cells appeared normal and intact in both groups. All images in d are 400 × 400 µm2. Representative images are from five corneas per group with similar results obtained for each group. Source Data
Fig. 3. Postmortem histologic analysis of corneas…
Fig. 3. Postmortem histologic analysis of corneas in the minipig model.
a, Hematoxylin and eosin (H&E) staining revealed autografts with thickened epithelium and increased presence of anterior stromal cells relative to the native porcine cornea. Epithelial and stromal layers in BPCDX corneas were uniform, with maintenance of overall corneal structure and anatomy. Representative images shown from three corneas per group. b, Three different BPCDX-implanted corneas where host cells (arrows, left and center images) migrated into the BPCDX. The edge of the BPCDX had multiple tissue attachments (arrows, right image) with cells appearing to migrate towards the BPCDX. c, Immunohistochemical analysis indicated sub-basal nerves by the β-III-tubulin marker (immediately below the epithelium, arrows), while a preserved stromal nerve in a BPCDX cornea was apparent. Leukocyte marker CD45 indicated weak staining of stromal cells located at the BPCDX periphery (arrows), suggesting leukocyte-mediated remodeling that differed in extent in different corneas (sections from two different BPCDX corneas shown). Leukocytes were absent in native and allograft corneas. All markers are indicated by green fluorescence, while a blue DAPI counterstain indicates the presence of cell nuclei. Non-specific diffuse signal from the green channel indicated the implanted BPCDX (asterisk in all panels). All images are representative images chosen from three corneas per group. Scale bars, 100 µm (a,c) and 50 µm (b). Source Data
Fig. 4. Clinical data from subjects in…
Fig. 4. Clinical data from subjects in Iran receiving BPCDX.
a, Keratometric and corneal thickness maps from the same subject indicate the thin and steep pre-operative cornea that was substantially thickened and flattened after intrastromal implantation of a 440-µm-thick BPCDX. The corresponding OCT cross-section scans indicate corneal thickness and shape before and after BPCDX implantation, with anterior and posterior borders of the BPCDX indicated by white arrows. The subject had an initial BSCVA of 20/200 that improved to 20/50 at 24 months post-operatively. b,c, Photographs of eyes from two subjects with the BPCDX four months post-operatively, indicating maintenance of corneal transparency. d,e, In vivo confocal microscopy images obtained from a single subject confirming the presence of sub-basal nerves (d) and endothelial cell mosaic (c) 6 months post-operatively. Endothelial cell density was 2,222 ± 62 cells per mm2 in the eye. As only this single subject was imaged by in vivo confocal microscopy, it is unknown if these images are representative. Images in d,e are 400 × 400 µm2.
Fig. 5. Clinical data from a subject…
Fig. 5. Clinical data from a subject in India receiving BPCDX.
a, Slit-lamp photographs pre-operatively (left) and one day post-operative (right) with arrows indicating immediate change in thickness and curvature in the central cornea. b, OCT scans indicating sustained thickening and regularization of corneal curvature following implantation of 280-µm-thick BPCDX (anterior and posterior surfaces of BPCDX indicated by white arrows). c, Topographic maps (left, values given are keratometric power in diopters), anterior surface elevation maps (center, values are in µm displacement from a best-fit sphere) and OCT pachymetric maps (right, thickness in µm) from the same subject indicated substantial flattening of the steepest pre-operative central region (black arrow), and substantial increase in corneal thickness post-operatively. The subject initially had a best contact lens-corrected visual acuity (BCLVA) of 20/600. At 24 months BCLVA improved to 20/30.
Extended Data Fig. 1. Real-time shelf-life stability…
Extended Data Fig. 1. Real-time shelf-life stability test data for BPCDX after 24 months of storage at 7°C, relative to time zero samples.
(a) Light transmission was maintained throughout the visible range of wavelengths (10 samples per group, BPCDX thickness 300 µm, diameter 9 mm, two-sided independent t-test). (b) Enzymatic degradation was unchanged, indicating implant integrity after long-term storage (9 samples per time point from 3 separate batches, BPCDX thickness 300 µm, diameter 9 mm, two-sided independent t-test). (c) Table indicating preservation of hydration and mechanical properties after long-term storage, with no significant differences relative to non-aged samples (550µm-thick ‘dog-bone’ specimens, two-sided independent t-test for each parameter). Data values in the graphs and table represent mean and standard deviation of measurements (error bars) from the indicated number of independent samples (at time 0 and 24 months, respectively). Source data

References

    1. Jeng BH, Ahmad S. In pursuit of the elimination of corneal blindness: is establishing eye banks and training surgeons enough? Opthalmology. 2021;128:813–815. doi: 10.1016/j.ophtha.2020.06.042.
    1. Mathews PM, Lindsley K, Aldave AJ, Akpek EK. Etiology of global corneal blindness and current practices of corneal transplantation: a focused review. Cornea. 2018;37:1198–1203. doi: 10.1097/ICO.0000000000001666.
    1. Gain P, et al. Global survey of corneal transplantation and eye banking. JAMA Ophthalmol. 2016;134:167–173. doi: 10.1001/jamaophthalmol.2015.4776.
    1. Oliva MS, Schottman T, Gulati M. Turning the tide of corneal blindness. Indian J. Ophthalmol. 2012;60:423. doi: 10.4103/0301-4738.100540.
    1. Whitcher JP, Srinivasan M, Upadhyay MP. Corneal blindness: a global perspective. Bull. World Health Organ. 2001;79:214–221.
    1. Chaurasia S, Sharma N, Das S. COVID-19 and eye banking. Indian J. Ophthalmol. 2020;68:1215. doi: 10.4103/ijo.IJO_1033_20.
    1. Toro M, Choragiewicz T, Posarelli C, Figus M, Rejdak R. Early Impact of COVID-19 outbreak on the availability of cornea donors: warnings and recommendations. Clin. Ophthalmol. 2020;14:2879. doi: 10.2147/OPTH.S260960.
    1. Lagali N. Corneal stromal regeneration: current status and future therapeutic potential. Curr. Eye Res. 2020;45:278–290. doi: 10.1080/02713683.2019.1663874.
    1. Matthyssen S, Van den Bogerd B, Dhubhghaill SN, Koppen C, Zakaria N. Corneal regeneration: a review of stromal replacements. Acta Biomater. 2018;69:31–41. doi: 10.1016/j.actbio.2018.01.023.
    1. Mahdavi SS, Abdekhodaie MJ, Mashayekhan S, Baradaran-Rafii A, Djalilian AR. Bioengineering approaches for corneal regenerative medicine. J. Tissue Eng. Regen. Med. 2020;17:567–593. doi: 10.1007/s13770-020-00262-8.
    1. Romero-Jiménez M, Santodomingo-Rubido J, Wolffsohn JS. Keratoconus: a review. Cont. Lens Anterior Eye. 2010;33:157–166. doi: 10.1016/j.clae.2010.04.006.
    1. Matthaei M, et al. Changing indications in penetrating keratoplasty: a systematic review of 34 years of global reporting. Transplantation. 2017;101:1387–1399. doi: 10.1097/TP.0000000000001281.
    1. Xu L, et al. Prevalence and associations of steep cornea/keratoconus in Greater Beijing. The Beijing Eye Study. PLoS ONE. 2012;7:e39313. doi: 10.1371/journal.pone.0039313.
    1. Gokhale NS. Epidemiology of keratoconus. Indian J. Ophthalmol. 2013;61:382–383. doi: 10.4103/0301-4738.116054.
    1. Hashemi H, et al. High prevalence and familial aggregation of keratoconus in an Iranian rural population: a population-based study. Ophthalmic Physiol. Opt. 2018;38:447–455. doi: 10.1111/opo.12448.
    1. Amayem AF, Hamdi IM, Hamdi MM. Refractive and visual outcomes of penetrating keratoplasty versus deep anterior lamellar keratoplasty with hydrodissection for treatment of keratoconus. Cornea. 2013;32:e2–e5. doi: 10.1097/ICO.0b013e31825ca70b.
    1. Hamdi IM, Hamdi MM. Quality of vision after deep anterior lamellar keratoplasty (fluid dissection) compared to penetrating keratoplasty for the treatment of keratoconus. J. Ophthalmol. 2017;2017:4507989.
    1. Jonas JB, Budde WM. Loosening of single versus double running sutures in penetrating keratoplasty for keratoconus. Graefe’s Arch. Clin. Exp. Opthalmol. 1999;237:522–523. doi: 10.1007/s004170050273.
    1. Khakshoor H, Eslampoor A, Rad SS, Vejdani A. Modified deep anterior lamellar keratoplasty for the treatment of advanced keratoconus with steep corneal curvature to help in eliminating the wrinkles in the Descemet’s membrane. Indian J. Ophthalmol. 2014;62:392–395. doi: 10.4103/0301-4738.121108.
    1. Parker JS, van Dijk K, Melles GR. Treatment options for advanced keratoconus: a review. Surv. Ophthalmol. 2015;60:459–480. doi: 10.1016/j.survophthal.2015.02.004.
    1. Mastropasqua L, Nubile M, Salgari N, Mastropasqua R. Femtosecond laser-assisted stromal lenticule addition keratoplasty for the treatment of advanced keratoconus: a preliminary study. J. Refract. Surg. 2018;34:36–44. doi: 10.3928/1081597X-20171004-04.
    1. van Dijk K, et al. Bowman layer transplantation to reduce and stabilize progressive, advanced keratoconus. Ophthalmol. 2015;122:909–917. doi: 10.1016/j.ophtha.2014.12.005.
    1. van Dijk K, et al. Bowman layer transplantation: 5-year results. Graefe’s Arch. Clin. Exp. Opthalmol. 2018;256:1151–1158. doi: 10.1007/s00417-018-3927-7.
    1. Meek KM. Corneal collagen—its role in maintaining corneal shape and transparency. Biophys. Rev. 2009;1:83–93. doi: 10.1007/s12551-009-0011-x.
    1. Stephens JD, Sarkisian SR., Jr The use of collagen matrix (Ologen) as a patch graft in glaucoma tube shunt surgery, a retrospective chart review. F1000Research. 2016;5:1898. doi: 10.12688/f1000research.9232.1.
    1. Snyder, D., Sullivan, N., Margolis, D. & Schoelles, K. Skin Substitutes for Treating Chronic Wounds. (Agency for Healthcare Research and Quality (US), 2020).
    1. Fagerholm P, 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. doi: 10.1126/scitranslmed.3001022.
    1. Fagerholm P, et al. Stable corneal regeneration four years after implantation of a cell-free recombinant human collagen scaffold. Biomaterials. 2014;35:2420–2427. doi: 10.1016/j.biomaterials.2013.11.079.
    1. Koulikovska M, et al. Enhanced regeneration of corneal tissue via a bioengineered collagen construct implanted by a nondisruptive surgical technique. Tissue Eng. Part A. 2015;21:1116–1130. doi: 10.1089/ten.tea.2014.0562.
    1. Rafat M, et al. PEG-stabilized carbodiimide crosslinked collagen-chitosan hydrogels for corneal tissue engineering. Biomaterials. 2008;29:3960–3972. doi: 10.1016/j.biomaterials.2008.06.017.
    1. Rafat M, et al. Composite core-and-skirt collagen hydrogels with differential degradation for corneal therapeutic applications. Biomaterials. 2016;83:142–155. doi: 10.1016/j.biomaterials.2016.01.004.
    1. Spoerl E, Mrochen M, Sliney D, Trokel S, Seiler T. Safety of UVA-riboflavin cross-linking of the cornea. Cornea. 2007;26:385–389. doi: 10.1097/ICO.0b013e3180334f78.
    1. Ganesh S, Brar S. Femtosecond intrastromal lenticular implantation combined with accelerated collagen cross-linking for the treatment of keratoconus—initial clinical result in 6 eyes. Cornea. 2015;34:1331–1339. doi: 10.1097/ICO.0000000000000539.
    1. El Zarif M, et al. Corneal stromal regeneration therapy for advanced keratoconus: long-term outcomes at 3 years. Cornea. 2021;40:741–754. doi: 10.1097/ICO.0000000000002646.
    1. Meek KM, Leonard DW, Connon CJ, Dennis S, Khan S. Transparency, swelling and scarring in the corneal stroma. Eye (Lond) 2003;17:927–936. doi: 10.1038/sj.eye.6700574.
    1. Jue B, Maurice DM. The mechanical properties of the rabbit and human cornea. J. Biomech. 1986;19:847–853. doi: 10.1016/0021-9290(86)90135-1.
    1. Crabb RA, Chau EP, Evans MC, Barocas VH, Hubel A. Biomechanical and microstructural characteristics of a collagen film-based corneal stroma equivalent. Tissue Eng. 2006;12:1565–1575. doi: 10.1089/ten.2006.12.1565.
    1. Bancelin S, et al. Determination of collagen fibril size via absolute measurements of second-harmonic generation signals. Nat. Commun. 2014;5:4920. doi: 10.1038/ncomms5920.
    1. Holmes DF, Lu Y, Starborg T, Kadler KE. Collagen fibril assembly and function. Curr. Top. Dev. Biol. 2018;130:107–142. doi: 10.1016/bs.ctdb.2018.02.004.
    1. Al-Aqaba MA, Otri AM, Fares U, Miri A, Dua HS. Organization of the regenerated nerves in human corneal grafts. Am. J. Ophthalmol. 2012;153:29. doi: 10.1016/j.ajo.2011.06.006.
    1. Niederer RL, Perumal D, Sherwin T, McGhee CN. Corneal innervation and cellular changes after corneal transplantation: an in vivo confocal microscopy study. Invest. Ophthalmol. Vis. Sci. 2007;48:621–626. doi: 10.1167/iovs.06-0538.
    1. Müller LJ, Marfurt CF, Kruse F, Tervo TM. Corneal nerves: structure, contents and function. Exp. Eye Res. 2003;76:521–542. doi: 10.1016/S0014-4835(03)00050-2.
    1. Xeroudaki M, et al. A porous collagen-based hydrogel and implantation method for corneal stromal regeneration and sustained local drug delivery. Sci. Rep. 2020;10:1–18. doi: 10.1038/s41598-020-73730-9.
    1. Zhang C, Xu J, Chen R. Histology of corneal wound healing after deep lamellar endothelial keratoplasty. Clin. Ophthalmol. 2008;2:217.
    1. Pirhadi S, et al. Comparison of the myoring implantation depth by mechanical dissection using pocketmaker microkeratome versus melles hook via AS-OCT. BMC Ophthalmol. 2018;18:137. doi: 10.1186/s12886-018-0806-2.
    1. Studeny P, Krizova D, Kuchynka P. Use of pocketmaker microkeratome for creation of corneal pocket for foldable keratoprosthesis keraklear implantation—case series. Open Ophthalmol. J. 2015;9:126. doi: 10.2174/1874364101509010126.
    1. Ong JA, et al. 3D corneal shape after implantation of a biosynthetic corneal stromal substitute. Invest. Ophthalmol. Vis. Sci. 2016;57:2355–2365. doi: 10.1167/iovs.15-18271.
    1. Funnell C, Ball J, Noble B. Comparative cohort study of the outcomes of deep lamellar keratoplasty and penetrating keratoplasty for keratoconus. Eye (Lond.) 2006;20:527–532. doi: 10.1038/sj.eye.6701903.
    1. Nguyen NX, Seitz B, Martus P, Langenbucher A, Cursiefen C. Long-term topical steroid treatment improves graft survival following normal-risk penetrating keratoplasty. Am. J. Ophthalmol. 2007;144:318–319. doi: 10.1016/j.ajo.2007.03.028.
    1. Sung MS, Yoon KC. Evaluation of graft–host interface after penetrating keratoplasty using anterior segment optical coherence tomography. Jpn. J. Ophthalmol. 2014;58:282–289. doi: 10.1007/s10384-014-0309-5.
    1. Tomita M, et al. Astigmatism following deep anterior lamellar keratoplasty versus penetrating keratoplasty. Invest. Ophthalmol. Vis. Sci. 2007;48:4680–4680.
    1. Zhao Y, Zhuang H, Hong J, Tian L, Xu J. Malapposition of graft–host interface after penetrating keratoplasty (PK) and deep anterior lamellar keratoplasty (DALK): an optical coherence tomography study. BMC Ophthalmol. 2020;20:41. doi: 10.1186/s12886-020-1307-7.
    1. Geerards AJ, Vreugdenhil W, Khazen A. Incidence of rigid gas-permeable contact lens wear after keratoplasty for keratoconus. Eye Contact Lens. 2006;32:207–210. doi: 10.1097/01.icl.0000191953.84449.d6.
    1. Buznyk O, et al. Bioengineered corneas grafted as alternatives to human donor corneas in three high‐risk patients. Clin. Transl. Sci. 2015;8:558–562. doi: 10.1111/cts.12293.
    1. Islam MM, et al. Biomaterials-enabled cornea regeneration in patients at high risk for rejection of donor tissue transplantation. NPJ Regen. Med. 2018;3:1–10. doi: 10.1038/s41536-017-0038-8.
    1. Khodaparast M, et al. Sutureless femtosecond laser-assisted anterior lamellar keratoplasty using a bioengineered cornea as a viable alternative to human donor transplantation for superficial corneal opacities. Cornea. 2020;39:1184–1189. doi: 10.1097/ICO.0000000000002394.
    1. Zhang MC, et al. Lamellar keratoplasty treatment of fungal corneal ulcers with acellular porcine corneal stroma. Am. J. Transplant. 2015;15:1068–1075. doi: 10.1111/ajt.13096.
    1. Zheng J, et al. Short‐term results of acellular porcine corneal stroma keratoplasty for herpes simplex keratitis. Xenotransplantation. 2019;26:e12509. doi: 10.1111/xen.12509.
    1. Rico-Sánchez L, et al. Successful development and clinical translation of a novel anterior lamellar artificial cornea. J. Tissue Eng. Regen. Med. 2019;13:2142–2154. doi: 10.1002/term.2951.
    1. Mathur V, Parihar J, Srivastava V, Avasthi A. Clinical evaluation of deep anterior lamellar keratoplasty (DALK) for stromal corneal opacities. Med. J. Armed Forces India. 2013;69:21–26. doi: 10.1016/j.mjafi.2012.04.023.
    1. Kolozsvári L, Nógrádi A, Hopp BL, Bor Z. UV absorbance of the human cornea in the 240- to 400-nm range. Invest. Ophthalmol. Vis. Sci. 2002;43:2165–2168.
    1. Merrett K, et al. Tissue-engineered recombinant human collagen-based corneal substitutes for implantation: performance of type I versus type III collagen. Invest. Ophthalmol. Vis. Sci. 2008;49:3887–3894. doi: 10.1167/iovs.07-1348.
    1. Wekhof A., Trompeter, F.-J. & Franken O. Pulsed UV Disintegration (PUVD): a new sterilisation mechanism for packaging and broad medical-hospital applications. In Proc. 1st Int. Conf. on Ultraviolet Technologies (International Ozone Association, 2001).
    1. Villacís JE, et al. Efficacy of pulsed-xenon ultraviolet light for disinfection of high-touch surfaces in an Ecuadorian hospital. BMC Infect. Dis. 2019;19:575. doi: 10.1186/s12879-019-4200-3.
    1. Lagali N, Rafat M. Femtosecond laser-assisted surgery for implantation of bioengineered corneal stroma to promote corneal regeneration. Methods Mol. Biol. 2020;2145:197–214. doi: 10.1007/978-1-0716-0599-8_14.
    1. Lagali, N., Griffith, M. & Fagerholm, P. In Vivo Confocal Microscopy of the Cornea to Assess Tissue Regenerative Response After Biomaterial Implantation in Humans. In Corneal Regenerative Medicine (eds Wright, B. & Connon, C. J.) 211–223 (Springer, 2013).
    1. Thatte S, Choudhary U, Sharma B. Efficacy of amniotic membrane transplantation in refractory infective keratitis leading to stromal thinning, descematocele and perforations. JOJ Ophthalmol. 2017;3:555611.

Source: PubMed

3
Subscribe