Skip to main content

SPECIALTY GRAND CHALLENGE article

Front. Ophthalmol., 04 July 2022
Sec. Surgical Ophthalmology
Volume 2 - 2022 | https://doi.org/10.3389/fopht.2022.922240

Grand Challenges and Opportunities in Surgical Ophthalmology: Together for a Shared Future

  • 1Eye Center, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
  • 2Zhejiang University Eye Hospital, Hangzhou, China
  • 3Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
  • 4Department of Ophthalmology, Queen’s University, Kingston, ON, Canada
  • 5Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
  • 6Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
  • 7Department of Ophthalmology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
  • 8Department of Ophthalmology and Visual Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
  • 9Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
  • 10Center for Integrated Oncology (CIO) Aachen-Bonn-Cologne-Duesseldorf, Cologne, Germany


Introduction

Surgical Ophthalmology is one of the topical branches of ophthalmology, which strives to relieve patients’ pain through operations. It plays an integral role in modern ophthalmology, involving almost all structures of the eye and its adnexal tissues. Therefore, from every ophthalmic subspecialty, we highlighted some of the biggest challenges and most recent developments in these fields to set the tone of this specialty section, inspire researchers to conduct groundbreaking research, and stimulate ophthalmologists to improve medical practice to serve patients better.

Ocular Surface

The ocular surface is a complex and crucial component of the visual system. It comprises the cornea, conjunctiva, and adnexa tissues, such as lacrimal glands and meibomian glands. Any disorder in these structures, including corneal ulcers, keratitis, chemical and thermal burns, dry eye disease (DED), etc. falls into the ocular surface disorder (OSD) (1, 2). OSD could severely affect the quality of vision and life and, in severe cases, may lead to blindness. Though with high prevalence, unfortunately, some cases often go undertreated due to the lack of complete understanding of the underlying mechanism of these diseases.

Recent years have witnessed rapid advancement in the researches in the OSD (3, 4). The advances in research and surgical treatment of severe OSDs have shed light on the management of these diseases. Surgical interventions, such as simple limbal epithelial transplantation (SLET) (5, 6) and corneal endothelial transplantation (7), have greatly improved the management of severe OSD. Another area that has lately gained popularity is the treatment of neurotrophic keratitis with corneal neurotization, with excellent outcomes in appropriate cases. It involves the coaptation of a healthy sensory periorbital nerve to the limbal area either directly or via an interposed nerve graft (8). There is nearly complete recovery of sensation in most cases, with accompanying improvement of the clinical picture.

We are particularly interested in manuscripts that report the new tools for detecting risk factors and early diagnosis of corneal ectasia, looking to the biomechanical behavior of the cornea and the future research directions for the treatment of the ectatic corneal disorder. Furthermore, potential promising topics include, but are not limited to, discoveries in the mechanisms of different OSDs, potential treatments of different OSDs, novel therapeutic approaches in OSDs, novel surgical treatments in OSDs, consecutive surgical treatment in OSDs, comparisons of the outcomes of different surgical options in OSDs.

Cataract

Over the last two decades, many new surgery technologies dramatically advanced cataract surgical treatments (913). Besides, various molecular studies about cataractogenesis enriched the cataract formation mechanism and provided novel applications for non-surgical treatments, including breakthroughs in possible pharmacological treatments (1418).

Small incision phacoemulsification cataract surgery with the intraocular lens (IOL) implantation has been a trend in cataract surgery. With the rapid development of the laser, recently, femtosecond laser-assisted cataract surgery (FLACS) has become an important development in the history of cataract surgery. Many clinical studies showed that FLACS offered the perfection of the capsulorhexis and long-term stability of the effective lens position of FLACS, which provided more predictable refractive outcomes (12, 1921). These were the significant advantages that manual surgery cannot achieve (20). In addition to these practices, FLACS offered a greater precision and repeatability for pre-chop than manual techniques and reduced effective phaco energy (22), especially in hard nuclear cataract surgery (21). Additionally, FLACS offered more precise incisional astigmatism management. As cataract surgery procedures advance every year, patient demands become more challenging. An ‘ideal’ IOL should restore the patients’ vision without glasses or visual compromises at all distances (23). Premium IOLs, including multifocal and accommodative IOLs, were designed to provide clear vision at near and distant focal points without additional spectacle correction and toric IOLs for astigmatism correction (24, 25). With the use of different IOLs, an ‘ideal’ vision can be achieved in some cases at present (26, 27).

To date, the only remedy for cataracts is surgically removing the cloudy lens and substituting it with a suitable IOL. Aiming to find more solutions to cure or prevent cataracts, the studies about cataractogenesis are still a hot topic. Oxidative stress, excess of quinoid substances, aldose reductase activation, and deficiency of autophagy are essential in the progression of cataracts (28, 29). Significant breakthroughs in pharmacological applications appeared in 2015 (30, 31). Novel pharmacological substances, 5-cholesten-3b,25-diol and lanosterol, can reverse lens opacity via dissolving the crystallin proteins aggregates. GSH, L-cystine, and rapamycin, as potential drugs, could protect the lens from opacity or reverse the opacity but still need more in-vivo testing (18, 32). Notably, the creation and development of high-throughput drug screening platforms, such as lentoid bodies and cataractous animal models (33), allow researchers to find more small molecular applications among millions.

Glaucoma

There has been an increase in optimizing glaucoma treatment medically and surgically over the past decades, with a subsequent decrease in associated complications, thereby improving the quality of life in glaucoma patients.

For decades, Prostaglandin F2α (PGF2α) analogs have been essential in the treatment of open-angle glaucoma. However, side effects such as prostaglandin-associated periorbitopathy (PAP) seriously affect patients’ treatment persistence. Recently, a new selective prostaglandin-EP2 agonist (omidenepag isopropyl OMDI), which is hydrolyzed to OMD (Omidenepag) when it penetrates the cornea, has shown promising clinical outcomes for decreasing the adverse effects (34). Moreover, clinical trials demonstrated that OMDI was non-inferior to PGF2α analogs in reducing IOP in patients with OHT or POAG and was well tolerated (35).

Although trabeculectomy is currently the gold standard in glaucoma surgery, it has many potential complications. Hence there is a rising interest in various minimally invasive glaucoma surgeries (MIGS) with a lower risk of complications and comparable IOP lower effects with conventional surgeries. According to the different strategies of surgery design, there are two main categories of MIGS available. The first group has yielded promising results in adult and pediatric glaucoma by creating a circumferential incision into SC and reducing the resistance to aqueous outflow (3639). Another one develops new MIGS devices to provide an alternative pathway through which aqueous humor can effectively exit the anterior chamber, reducing IOP (4042). MIGS is widely becoming the front-line glaucoma surgery with respect to various advantages (4348).

Deep learning (DL), a subset of artificial intelligence based on deep neural networks, has made significant breakthroughs in glaucoma image classification and pattern recognition. Studies incorporated with DL for interpreting optical coherence tomography(OCT)or visual field (VF) data have demonstrated exemplary performance for discriminating glaucomatous eyes from normal eyes (49, 50). At the same time, there are currently some virtual reality-based visual field examinations (VR VFs) to check the visual field, which is not only more ergonomic in design but also cost-effective, suggesting that VR perimeters have the potential to examine VFs with high enough confidence, whereby reducing challenges in current perimetry test by providing a more accessible visual field test (51, 52). These high developed techs make “tele-ophthalmology” possible hence enabling patients to communicate with their attending doctors in real-time through the network, which comprehensively promotes the development of remote ophthalmic consultation (53).

Posterior Eye Segment

The posterior eye segment comprises the back two-thirds of the eye, involving the anterior hyaloid membrane and all of the optical structures behind it: the vitreous humor, retina, choroid, and optic nerve.

The rapid development of fundus imaging technology has led to an increasing range of fundus images, with more accurate picture resolution, non-invasive examination methods, and efficient treatment (54). The range of visual fundus imaging has evolved from 25° to 35°, 45°, and 55° in the early days, and the most remarkable progress has been made with the advent of laser ophthalmoscopy, which has enabled a qualitative leap in the range of fundus imaging, from 55° to over 100°. In particular, ultra-wide-field imaging has enabled fundus imaging to reach even 200° (i.e., to the extent of the vortex vein and the area before the vortex vein). This wider range helps ophthalmologists detect more peripheral fundus diseases and improves the detection rate of disease (55). That was followed by fluorescence angiography, OCT, and OCTA, with resolutions ranging from 10 µm to 5 µm to 3 µm, gradually progressing to the current level of cellular resolution. More accurate examination techniques help ophthalmologists understand the onset and progression of the disease. In recent years, OCTA has been a fascinating new technology, a non-invasive way to quantitatively assess retinal and even choroidal blood flow without invasive fluorescein angiography (56, 57). It is now widely used in fundus vascular disease.

The advent of small-molecule antibodies, represented by anti-VEGF drugs, and the introduction of intravitreal injections have been landmark advances in the treatment of fundus diseases, changing the previous treatment of fundus vascular diseases, reducing the number of vitreous procedures, and significantly improving the prognosis of diseases such as wAMD, macular edema, and diabetic retinopathy. Anti-VEGF drugs have also emerged as a first-line treatment option for many fundus vascular diseases (58). Furthermore, since the advent of standard three-channel 20G vitrectomy in 1972, sutureless vitrectomies of 23G, 25G, and 27G have been introduced, bringing vitrectomy surgery into the minimally invasive era (59, 60). Compared with the traditional 20G three-channel vitrectomy, minimally invasive vitrectomy reduces the size of the scleral incision, dramatically simplifies the surgical procedure, shortens the operating time, and reduces surgical complications. The availability of anti-VEGF drugs and innovations in minimally invasive vitrectomy techniques have improved the cure rate of fundus diseases, reduced surgical complications, and greatly improved the prognosis of patients.

In recent years, with the interdisciplinary interpenetration of molecular biology, cytogenetics, genetic engineering, stem cell biology, and artificial intelligence (61), ophthalmologists’ knowledge and understanding of fundus diseases such as wAMD, diabetic macular edema, and macular hole have gradually improved, and their diagnosis and treatment have also advanced significantly. However, we still do not have effective treatment options for fundus diseases like retinitis pigmentosa and posterior scleral staphyloma in pathological myopia. There is still a need for further clinical and basic research to be conducted in the future.

Strabismus

Unlike other eye surgery (cataract, glaucoma, and vitreoretinal surgery, among others) in which skilled techniques are the primary factors of successful operation, the key to strabismus surgery is reasonable design. In recent years, several innovations and modifications in strabismus surgery design have been brought up. The innovation in strabismus surgery focused on the merits of techniques in common strabismus and the treatment of paralytic strabismus such as abducens nerve palsy.

The bilateral lateral rectus recession (BLRc) and unilateral lateral rectus recession combined with medial rectus resection (R&R) are the most common two surgical procedures treating basic-type intermittent exotropia (IXT). The Pediatric Eye Disease Investigator Group performed a multicenter, randomized clinical trial in children of 3 to 11 years old to compare the short-term and long-term outcomes. They found no statistically significant difference in the surgical success rates by three years between children treated with both surgeries, respectively (62).

Paralytic strabismus due to cranial nerve palsy is a relatively tricky problem. Strabismus secondary to abducens nerve (CN 6) palsy is the most common paralytic type, and various surgical techniques are now used in the clinic. Hummelshein first introduced vertical rectus transposition (VRT) in 1907 (63, 64), in which full-tendon width superior and inferior rectus with tenotomy transposed to the margins of the lateral rectus. Medial rectus recession would be performed in most cases due to medial rectus contracture (65), but tenotomy of three or more rectus muscles would increase the risk of anterior segment ischemia (ASI). In order to reduce the risk of ASI and also improve abduction ability, VRT procedures were modified to superior rectus transposition (SRT) (66, 67) and vertical rectus belly transposition (VRBT) without tenotomy by Nishida (68) and also modified by Chen Zhao (mVRBT)[50]. The critical step of mVRBT was without tenotomy. The superior rectus belly and inferior rectus belly are transposed to a position 2 mm adjacent and 6–8 mm posterior to the superior and inferior pole of the lateral rectus muscle, resulting in 57.8△ of esotropia correction and 2.3 scales of abduction improvement.

The third nerve (CN 3) palsy treatments are more complicated since the affected branches and their severity vary and may accompany aberrant regeneration. The ideal goal of treating complete CN 3 palsy is to retain alignment at the primary gaze without diplopia. Although previous techniques could improve the deviations in complete CN 3 palsy cases, residual deviations at primary gaze in short-term or long-term follow-up often occur. The recent popular surgery is Gokyigit’s technique (69), The lateral rectus muscle was split with the upper half transposed to the superior border and the lower half to the inferior border of the medial rectus insertion. Since with Gokyigit’s technique, 50% of patients achieved stable alignment, and the other 50% were undercorrected, requiring a second surgery, a modification of the existing technique by force augmentation through the use of equatorial fixation sutures resulting in satisfactory primary gaze alignment in the complete CN 3 palsy (70).

Ophthalmic Plastic and Reconstructive Surgery

The subspecialty of Ophthalmic Plastic and Reconstructive Surgery has experienced a kind of renaissance for research and innovation over the last 10-15 years. As an overview, we can discuss some of these advancements in terms of medical and surgical approaches to disease management.

On the medical (pharmaceutical) side, probably the most crucial innovation came with the advent of biopharmaceuticals, better known as biologic agents, as a novel targeted treatment for inflammatory conditions of the orbit. That would likely revolutionize the treatment of thyroid eye disease (TED), with drugs such as teprotumumab and tocilizumab showing significant improvement in the condition in recent studies (71, 72). Such targeted treatment with biologics has been the first major innovation in the management of TED in many years. The same can be said for idiopathic orbital inflammatory syndromes (IOIS), where biologics also show great promise (73). Regarding IOIS, advances have also been made to reclassify some of the so-called ‘idiopathic’ inflammatory conditions into specific disease entities, the prime example being the IgG-4 group of diseases (74).

In the field of oncology, targeted molecular therapy has also made a significant leap forward as a treatment option for some cancers, such as advanced, recurrent basal cell carcinoma (BCC), squamous cell carcinoma (SCC), and malignant melanoma of the periorbital tissues, where surgery may not be indicated or feasible. Especially for BCC, vismodegib has been shown to interrupt the abnormally upregulated Hedgehog signaling pathway specifically. This results in significant regression, if not resolution, of the disease (75). Recent advances have also been made with the advent of immune checkpoint inhibitors for extensive and inoperative squamous cell carcinoma (SCC) and metastatic carcinoma. Recently, the PD-1 inhibitor (cemiplimab) has been approved for this indication in the US, and EGFR inhibitors such as cetuximab may also be helpful in some patients (76). Targeted molecular therapy has also attracted research for ocular and adnexal malignant melanomas (77), and secondary orbital tumors, such as lymphomas. In addition to the above-mentioned new treatment options, updated TNM staging protocols and indications for sentinel lymph node (SLN) biopsy/resection have also been added to our management armamentarium in treating periorbital cancers.

Despite anatomy being an old field, advances in the orbital/eyelid region have also occurred. As an example, recent work by MJ Ali on the Horner-Duverney’s muscle microanatomy has added more to our understanding of the lacrimal pump, with the potential existence of a type of canalicular peristaltic function to aid in tear drainage (78). The description of a relatively new finding of a post-aponeurotic fat pad is another example (79).

There has been a steady move towards minimally invasive orbital surgery as far as surgical advancements go (80, 81). Transconjunctival incisions have become relatively common to access anterior orbital tumors, such as the transcaruncular approach to the medial orbit or superior fornix incisions for superior lesions. More work is being done in endoscopic orbital surgery for mid-orbit and the apex (82). Often it is combined with 3D stereo navigation for accurate localization of the deep apical tumor and safer removal. Additionally, custom-made, 3D printed patient-specific implants have been described for orbital fractures and congenital craniofacial anomalies and are being further refined (83).

Minimally invasive surgery has also become the norm in eyelid and lacrimal procedures. Ptosis surgery is now commonly carried out as a minimal incision levator advancement (MILA) or a posterior approach to Muller’s muscle (MMR) and the levator to hide the wound completely. Dacryocystorhinostomies have been done endoscopically for some time, but in the last few years, transcanalicular laser-assisted lacrimal procedures have been studied and are already in use in some countries (8487).

For surface reconstruction of periorbital defects, bioengineered dermal substitutes have become more popular with advances in autograft, allograft, and xenograft laboratory preparation and the development of entirely synthetic substitutes (88).

Looking at the way forward, the future holds continued refinement and improvement in all areas mentioned above. That is specifically true in the availability and choice of biologic agents as well as the expanding list of conditions to treat. For example, it may well be that primarily thyroid orbitopathy becomes a medical disease, with surgery used only in extreme recalcitrant cases. There will be continued refinement in identifying ‘idiopathic’ inflammations as specific disease entities, which will lend themselves better to targeted systemic treatments. The same will hold for targeted biologic therapy for periorbital and orbital tumors as the primary treatment modality for advanced cases.

Minimal incision and endoscopic orbital surgery would likely become the standard of care for orbital tumors, with the likely innovative expansion to trans-orbital approaches to the middle cranial fossa for neurosurgical cases. Improvement in equipment and laser technology for transcanalicular lacrimal surgery may, in the future, result in a DCR being a 10-minute office procedure (86).

With respect to infections, a recent study described a metagenome analysis of the local lacrimal biome and coined the term ‘lacriome.’ With the aid of DNA sequencing, this technique identifies organisms present well beyond standard laboratory culture methods and may aid in future identifying and studying infectious processes more accurately (89).

Gene therapy has been studied for several years and shows promising results in inherited eye disorders, such as Leber congenital amaurosis and retinitis pigmentosa. More recently, a mouse model of oculopharyngeal muscular dystrophy (OPMD) with a faulty PABPN1 gene was successfully treated with gene therapy, which resulted in regression of muscle aggregates and fibrosis (90). That may hold promise for some inherited diseases in the field of Ophthalmic Plastic Surgery in the future.

Artificial Intelligence (AI) already plays a role in several specialties, such as radiology for routine screening of images, and the same concepts can be applied to Ophthalmic Plastic Surgery. A number of recent papers have used AI for eyelid and periorbital measurements (91, 92). A similar approach could be used to classify periorbital lesions with 3D stereophotogrammetry and apply AI analysis to aid in diagnosis and triage (9398).

Lastly, most oculoplastic conditions lend themselves nicely to photographic documentation without specialized camera equipment. This feature can easily be used for electronic medical records and also adopted for transmission via Telemedicine protocols (91). The current pandemic has reinforced the utility of e-consultations, and this trend is bound to continue and expand well beyond this period.

Author Contributions

The writing of this manuscript was a collective effort among YG, VK, HX, XS, XM, YW, WW, AR, and LH. YG, AR, and LH contributed to the conception and design of the work, the organization of the draft, and the improvement of the manuscript. YG, VK, HX, XS, XM, YW, and WW each drafted initial sections of the paper. All authors contributed to the manuscript revision and approved the submitted version.

Funding

This work was supported by National Natural Science Foundation of China (Grant No. 82102346).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Mikalauskiene L, Grzybowski A, Zemaitiene R. Ocular Surface Changes Associated With Ophthalmic Surgery. J Clin Med (2021) 10(8):1642. doi: 10.3390/jcm10081642

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Rana HS, Akella SS, Clabeaux CE, Skurski ZP, Aakalu VK. Ocular Surface Disease in Thyroid Eye Disease: A Narrative Review. Ocul Surf (2022) 24:67–73. doi: 10.1016/j.jtos.2022.02.001

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Aragona P, Baudouin C, Benitez Del Castillo JM, Messmer E, Barabino S, Merayo-Lloves J, et al. The Ocular Microbiome and Microbiota and Their Effects on Ocular Surface Pathophysiology and Disorders. Surv Ophthalmol (2021) 66(6):907–25. doi: 10.1016/j.survophthal.2021.03.010

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Jackson CJ, Gundersen KG, Tong L, Utheim TP. Dry Eye Disease and Proteomics. Ocul Surf (2022) 24:119–28. doi: 10.1016/j.jtos.2022.03.001

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Singh P, Raj A, Gupta A. Outcomes of Ipsilateral Simple Limbal Epithelial Transplantation, Tenonectomy, Mitomycin, and Amniotic Membrane Transplantation for Treatment of Recurrent Pterygium- Letter to Editor. Cornea (2021) 40(10):e19–20. doi: 10.1097/ICO.0000000000002726

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Kate A, Mudgil T, Basu S. Longitudinal Changes in Corneal Epithelial Thickness and Reflectivity Following Simple Limbal Epithelial Transplantation: An Optical Coherence Tomography-Based Study. Curr Eye Res (2022) 47(3):336–342. doi: 10.1080/02713683.2021.1988985

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Song ES, Park JH, Ha SS, Cha PH, Kang JT, Park CY, et al. Novel Corneal Endothelial Cell Carrier Couples a Biodegradable Polymer and a Mesenchymal Stem Cell-Derived Extracellular Matrix. ACS Appl Mater Interfaces (2022) 14(10):12116–29. doi: 10.1021/acsami.2c01709

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Koaik M, Baig K. Corneal Neurotization. Curr Opin Ophthalmol (2019) 30(4):292–8. doi: 10.1097/ICU.0000000000000578

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Schweitzer C, Brezin A, Cochener B, Monnet D, Germain C, Roseng S, et al. Femtosecond Laser-Assisted Versus Phacoemulsification Cataract Surgery (FEMCAT): A Multicentre Participant-Masked Randomised Superiority and Cost-Effectiveness Trial. Lancet (2020) 395(10219):212–24. doi: 10.1016/S0140-6736(19)32481-X

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Wang W, Chen X, Liu X, Zhang X, Lyu D, Yao K. Lens Capsule-Related Complications in Femtosecond Laser-Assisted Cataract Surgery: A Study Based on Video Analysis. Br J Ophthalmol (2022). doi: 10.1136/bjophthalmol-2021-320842

CrossRef Full Text | Google Scholar

11. Marcos S, Martinez-Enriquez E, Vinas M, de Castro A, Dorronsoro C, Bang SP, et al. Simulating Outcomes of Cataract Surgery: Important Advances in Ophthalmology. Annu Rev BioMed Eng (2021) 23:277–306. doi: 10.1146/annurev-bioeng-082420-035827

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Zhong Y, Zhu Y, Wang W, Wang K, Liu X, Yao K. Femtosecond Laser-Assisted Cataract Surgery Versus Conventional Phacoemulsification: Comparison of Internal Aberrations and Visual Quality. Graefes Arch Clin Exp Ophthalmol (2022) 260(3):901–11. doi: 10.1007/s00417-021-05441-4

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Wang K, Song F, Zhang L, Xu J, Zhong Y, Lu B, et al. Three-Dimensional Heads-Up Cataract Surgery Using Femtosecond Laser: Efficiency, Efficacy, Safety, and Medical Education-A Randomized Clinical Trial. Transl Vis Sci Technol (2021) 10(9):4. doi: 10.1167/tvst.10.9.4

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Morishita H, Eguchi T, Tsukamoto S, Sakamaki Y, Takahashi S, Saito C, et al. Organelle Degradation in the Lens by PLAAT Phospholipases. Nature (2021) 592(7855):634–8. doi: 10.1038/s41586-021-03439-w

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Ren L, Hu L, Zhang Y, Liu J, Xu W, Wu W, et al. Cataract-Causing S93R Mutant Destabilized Structural Conformation of βb1 Crystallin Linking With Aggregates Formation and Cellular Viability. Front Mol Biosci (2022) 9:844719. doi: 10.3389/fmolb.2022.844719

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Gu Y, Yao K, Fu Q. Lens Regeneration: Scientific Discoveries and Clinical Possibilities. Mol Biol Rep (2021) 48(5):4911–23. doi: 10.1007/s11033-021-06489-5

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Gulluni F, Prever L, Li H, Krafcikova P, Corrado I, Lo WT, et al. PI(3,4)P2-Mediated Cytokinetic Abscission Prevents Early Senescence and Cataract Formation. Science (2021) 374(6573):eabk0410. doi: 10.1126/science.abk0410

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Ping X, Liang J, Shi K, Bao J, Wu J, Yu X, et al. Rapamycin Relieves the Cataract Caused by Ablation of Gja8b Through Stimulating Autophagy in Zebrafish. Autophagy (2021) 17(11):3323–37. doi: 10.1080/15548627.2021.1872188

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Xu J, Chen X, Wang H, Yao K. Safety of Femtosecond Laser-Assisted Cataract Surgery Versus Conventional Phacoemulsification for Cataract: A Meta-Analysis and Systematic Review. Adv Ophthalmol Pract Res (2022) 2(1):100027. doi: 10.1016/j.aopr.2022.100027

CrossRef Full Text | Google Scholar

20. Conrad-Hengerer I, Al Sheikh M, Hengerer FH, Schultz T, Dick HB. Comparison of Visual Recovery and Refractive Stability Between Femtosecond Laser-Assisted Cataract Surgery and Standard Phacoemulsification: Six-Month Follow-Up. J Cataract Refract Surg (2015) 41(7):1356–64. doi: 10.1016/j.jcrs.2014.10.044

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Chen X, Yu Y, Song X, Zhu Y, Wang W, Yao K. Clinical Outcomes of Femtosecond Laser-Assisted Cataract Surgery Versus Conventional Phacoemulsification Surgery for Hard Nuclear Cataracts. J Cataract Refract Surg (2017) 43(4):486–91. doi: 10.1016/j.jcrs.2017.01.010

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Day AC, Burr JM, Bennett K, Doré CJ, Bunce C, Hunter R, et al. Femtosecond Laser-Assisted Cataract Surgery Compared With Phacoemulsification Cataract Surgery: Randomized Noninferiority Trial With 1-Year Outcomes. J Cataract Refract Surg (2020) 46(10):1360–7. doi: 10.1097/j.jcrs.0000000000000257

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Braga-Mele R, Chang D, Dewey S, Foster G, Henderson BA, Hill W, et al. Multifocal Intraocular Lenses: Relative Indications and Contraindications for Implantation. J Cataract Refract Surg (2014) 40(2):313–22. doi: 10.1016/j.jcrs.2013.12.011

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Wang SY, Stem MS, Oren G, Shtein R, Lichter PR. Patient-Centered and Visual Quality Outcomes of Premium Cataract Surgery: A Systematic Review. Eur J Ophthalmol (2017) 27(4):387–401. doi: 10.5301/ejo.5000978

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Miháltz K, Szegedi S, Steininger J, Vécsei-Marlovits PV. The Relationship Between Patient Satisfaction and Visual and Optical Outcome After Bilateral Implantation of an Extended Depth of Focus Multifocal Intraocular Lens. Adv Ophthalmol Pract Res (2022) 2(1):100043. doi: 10.1016/j.aopr.2022.100043

CrossRef Full Text | Google Scholar

26. Zvorničanin J, Zvorničanin E. Premium Intraocular Lenses: The Past, Present and Future. J Curr Ophthalmol (2018) 30(4):287–96. doi: 10.1016/j.joco.2018.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Vitalyevich PV, Astakhov SY, Vladimirovna GE, Yuan VS. Limbal Mini-Pockets for Transscleral IOL Fixation. Adv Ophthalmol Pract Res (2022) 2(1):100044.

Google Scholar

28. Morishita H, Mizushima N. Autophagy in the Lens. Exp Eye Res (2016) 144:22–8. doi: 10.1016/j.exer.2015.08.019

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Xu J, Fu Q, Chen X, Yao K. Advances in Pharmacotherapy of Cataracts. Ann Transl Med (2020) 8(22):1552. doi: 10.21037/atm-20-1960

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Makley LN, McMenimen KA, DeVree BT, Goldman JW, McGlasson BN, Rajagopal P, et al. Pharmacological Chaperone for α-Crystallin Partially Restores Transparency in Cataract Models. Science (2015) 350(6261):674–7. doi: 10.1126/science.aac9145

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Zhao L, Chen XJ, Zhu J, Xi YB, Yang X, Hu LD, et al. Lanosterol Reverses Protein Aggregation in Cataracts. Nature (2015) 523(7562):607–11. doi: 10.1038/nature14650

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Babizhayev MA. Generation of Reactive Oxygen Species in the Anterior Eye Segment. Synergistic Codrugs of N-Acetylcarnosine Lubricant Eye Drops and Mitochondria-Targeted Antioxidant Act as a Powerful Therapeutic Platform for the Treatment of Cataracts and Primary Open-Angle Glaucoma. BBA Clin (2016) 6:49–68. doi: 10.1016/j.bbacli.2016.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Lyu D, Zhang L, Qin Z, Ni S, Li J, Lu B, et al. Modeling Congenital Cataract In Vitro Using Patient-Specific Induced Pluripotent Stem Cells. NPJ Regener Med (2021) 6(1):60. doi: 10.1038/s41536-021-00171-x

CrossRef Full Text | Google Scholar

34. Oogi S, Nakakura S, Terao E, Fujisawa Y, Tabuchi H, Kiuchi Y. One-Year Follow-Up Study of Changes in Prostaglandin-Associated Periorbital Syndrome After Switch From Conventional Prostaglandin F2alfa to Omidenepag Isopropyl. Cureus (2020) 12(8):e10064. doi: 10.7759/cureus.10064

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Aihara M, Lu F, Kawata H, Iwata A, Odani-Kawabata N, Shams NK. Omidenepag Isopropyl Versus Latanoprost in Primary Open-Angle Glaucoma and Ocular Hypertension: The Phase 3 AYAME Study. Am J Ophthalmol (2020) 220:53–63. doi: 10.1016/j.ajo.2020.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Areaux RG Jr., Grajewski AL, Balasubramaniam S, Brandt JD, Jun A, Edmunds B, et al. Trabeculotomy Ab Interno With the Trab360 Device for Childhood Glaucomas. Am J Ophthalmol (2020) 209:178–86. doi: 10.1016/j.ajo.2019.10.014

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Grover DS, Smith O, Fellman RL, Godfrey DG, Butler MR, Montes de Oca I, et al. Gonioscopy Assisted Transluminal Trabeculotomy: An Ab Interno Circumferential Trabeculotomy for the Treatment of Primary Congenital Glaucoma and Juvenile Open Angle Glaucoma. Br J Ophthalmol (2015) 99(8):1092–6. doi: 10.1136/bjophthalmol-2014-306269

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Rahmatnejad K, Pruzan NL, Amanullah S, Shaukat BA, Resende AF, Waisbourd M, et al. Surgical Outcomes of Gonioscopy-Assisted Transluminal Trabeculotomy (GATT) in Patients With Open-Angle Glaucoma. J Glaucoma (2017) 26(12):1137–43. doi: 10.1097/IJG.0000000000000802

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Wang Y, Wang H, Han Y, Shi Y, Xin C, Yin P, et al. Outcomes of Gonioscopy-Assisted Transluminal Trabeculotomy in Juvenile-Onset Primary Open-Angle Glaucoma. Eye (Lond) (2021) 35(10):2848–54. doi: 10.1038/s41433-020-01320-0

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Koh V, Chew P, Triolo G, Lim KS, Barton K. Treatment Outcomes Using the PAUL Glaucoma Implant to Control Intraocular Pressure in Eyes With Refractory Glaucoma. Ophthalmol Glaucoma (2020) 3(5):350–9. doi: 10.1016/j.ogla.2020.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Pereira ICF, van de Wijdeven R, Wyss HM, Beckers HJM, den Toonder JMJ. Conventional Glaucoma Implants and the New MIGS Devices: A Comprehensive Review of Current Options and Future Directions. Eye (Lond) (2021) 35(12):3202–21. doi: 10.1038/s41433-021-01595-x

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Scheres LMJ, Kujovic-Aleksov S, Ramdas WD, de Crom R, Roelofs LCG, Berendschot T, et al. XEN(®) Gel Stent Compared to PRESERFLO™ MicroShunt Implantation for Primary Open-Angle Glaucoma: Two-Year Results. Acta Ophthalmol (2021) 99(3):e433–e40. doi: 10.1111/aos.14602

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Luebke J, Böhringer D, Evers C, Daniel MC, Reinhard T, Lang SJ. Glaucoma Treatment in German Hospitals in 2019. Klin Monbl Augenheilkd (2022).

PubMed Abstract | Google Scholar

44. Romera Romero P, Duch S, Moreno-Montañés J, Botella García J, Balboa Miró M, Loscos Arenas J. Survey of Glaucoma Surgical Preferences Among Glaucoma Specialists in Spain. Arch Soc Esp Oftalmol (Engl Ed) (2022) 97(6):310–316. doi: 10.1016/j.oftale.2022.03.004

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Gambini G, Carlà MM, Giannuzzi F, Caporossi T, De Vico U, Savastano A, et al. PreserFlo(®) MicroShunt: An Overview of This Minimally Invasive Device for Open-Angle Glaucoma. Vision (Basel) (2022) 6(1):12. doi: 10.3390/vision6010012

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Fujita A, Hashimoto Y, Matsui H, Yasunaga H, Aihara M. Recent Trends in Glaucoma Surgery: A Nationwide Database Study in Japan, 2011-2019. Jpn J Ophthalmol (2022) 66(2):183–92. doi: 10.1007/s10384-021-00898-6

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Ichhpujani P, Singla E, Kalra G, Bhartiya S, Kumar S. Surgical Trends in Glaucoma Management: The Current Indian Scenario. Int Ophthalmol (2022) 42(6):1661–1668. doi: 10.1007/s10792-021-02160-x

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Kalarn S, Le T, Rhee DJ. The Role of Trabeculectomy in the Era of Minimally Invasive Glaucoma Surgery. Curr Opin Ophthalmol (2022) 33(2):112–8. doi: 10.1097/ICU.0000000000000811

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Ran AR, Tham CC, Chan PP, Cheng CY, Tham YC, Rim TH, et al. Deep Learning in Glaucoma With Optical Coherence Tomography: A Review. Eye (Lond) (2021) 35(1):188–201. doi: 10.1038/s41433-020-01191-5

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Wang M, Shen LQ, Pasquale LR, Boland MV, Wellik SR, De Moraes CG, et al. Artificial Intelligence Classification of Central Visual Field Patterns in Glaucoma. Ophthalmology (2020) 127(6):731–8. doi: 10.1016/j.ophtha.2019.12.004

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Montelongo M, Gonzalez A, Morgenstern F, Donahue SP, Groth SL. A Virtual Reality-Based Automated Perimeter, Device, and Pilot Study. Transl Vis Sci Technol (2021) 10(3):20. doi: 10.1167/tvst.10.3.20

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Stapelfeldt J, Kucur SS, Huber N, Höhn R, Sznitman R. Virtual Reality-Based and Conventional Visual Field Examination Comparison in Healthy and Glaucoma Patients. Transl Vis Sci Technol (2021) 10(12):10. doi: 10.1167/tvst.10.12.10

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Chandrasekaran S, Kass W, Thangamathesvaran L, Mendez N, Khouri P, Szirth BC, et al. Tele-Glaucoma Versus Clinical Evaluation: The New Jersey Health Foundation Prospective Clinical Study. J Telemed Telecare (2020) 26(9):536–44. doi: 10.1177/1357633X19845273

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Soomro T, Shah N, Niestrata-Ortiz M, Yap T, Normando EM, Cordeiro MF. Recent Advances in Imaging Technologies for Assessment of Retinal Diseases. Expert Rev Med Devices (2020) 17(10):1095–108. doi: 10.1080/17434440.2020.1816167

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Ludwig CA, Moon J, Garg I, Miller JB. Ultra-Widefield Imaging for Evaluation of the Myopic Eye. Semin Ophthalmol (2021) 36(4):185–90. doi: 10.1080/08820538.2021.1887904

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Adejumo T, Kim TH, Le D, Son T, Ma G, Yao X. Depth-Resolved Vascular Profile Features for Artery-Vein Classification in OCT and OCT Angiography of Human Retina. BioMed Opt Express (2022) 13(2):1121–30. doi: 10.1364/BOE.450913

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Lal B, Alonso-Caneiro D, Read SA, Tran B, Van Bui C, Tang D, et al. Changes in Retinal Optical Coherence Tomography Angiography Indexes Over 24 Hours. Invest Ophthalmol Vis Sci (2022) 63(3):25. doi: 10.1167/iovs.63.3.25

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Tsai AS, Chou HD, Ling XC, Al-Khaled T, Valikodath N, Cole E, et al. Assessment and Management of Retinopathy of Prematurity in the Era of Anti-Vascular Endothelial Growth Factor (VEGF). Prog Retin Eye Res (2022) 88:101018. doi: 10.1016/j.preteyeres.2021.101018

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Brown GT, Pugazhendhi S, Beardsley RM, Karth JW, Karth PA, Hunter AA. 25 vs. 27-Gauge Micro-Incision Vitrectomy Surgery for Visually Significant Macular Membranes and Full-Thickness Macular Holes: A Retrospective Study. Int J Retina Vitreous (2020) 6(1):56. doi: 10.1186/s40942-020-00259-4

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Ting Yip L, Fong A, Wai Tsang C. A Novel Technique for 27-Gauge Forceps Assisted Transcleral Refixation of Dislocated CZ70BD Intraocular Lens. Retina (2021). doi: 10.1097/IAE.0000000000003225

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Li M, Wan C. The Use of Deep Learning Technology for the Detection of Optic Neuropathy. Quant Imaging Med Surg (2022) 12(3):2129–43. doi: 10.21037/qims-21-728

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Donahue SP, Chandler DL, Holmes JM, Arthur BW, Paysse EA, Wallace DK, et al. A Randomized Trial Comparing Bilateral Lateral Rectus Recession Versus Unilateral Recess and Resect for Basic-Type Intermittent Exotropia. Ophthalmology (2019) 126(2):305–17. doi: 10.1016/j.ophtha.2018.08.034

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Schillinger RJ. A New Type of Tendon Transplant Operation for Abducens Paralysis. J Int Coll Surg (1959) 31(5):593–600.

PubMed Abstract | Google Scholar

64. del Pilar González M, Kraft SP. Outcomes of Three Different Vertical Rectus Muscle Transposition Procedures for Complete Abducens Nerve Palsy. J aapos (2015) 19(2):150–6. doi: 10.1016/j.jaapos.2015.01.007

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Rosenbaum AL. Costenbader Lecture. The Efficacy of Rectus Muscle Transposition Surgery in Esotropic Duane Syndrome and VI Nerve Palsy. J aapos (2004) 8(5):409–19. doi: 10.1016/j.jaapos.2004.07.006

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Akbari MR, Masoumi A, Mirmohammadsadeghi A. Superior Rectus Transposition and Medial Rectus Recession for Treatment of Duane Retraction Syndrome and Sixth Nerve Palsy. J Binocul Vis Ocul Motil (2021) 71(2):45–9. doi: 10.1080/2576117X.2021.1879985

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Patil-Chhablani P, Kothamasu K, Kekunnaya R, Sachdeva V, Warkad V. Augmented Superior Rectus Transposition With Medial Rectus Recession in Patients With Abducens Nerve Palsy. J aapos (2016) 20(6):496–500. doi: 10.1016/j.jaapos.2016.07.227

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Nishida Y, Hayashi O, Oda S, Kakinoki M, Miyake T, Inoki Y, et al. A Simple Muscle Transposition Procedure for Abducens Palsy Without Tenotomy or Splitting Muscles. Jpn J Ophthalmol (2005) 49(2):179–80. doi: 10.1007/s10384-004-0151-2

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Gokyigit B, Akar S, Satana B, Demirok A, Yilmaz OF. Medial Transposition of a Split Lateral Rectus Muscle for Complete Oculomotor Nerve Palsy. J aapos (2013) 17(4):402–10. doi: 10.1016/j.jaapos.2013.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Saxena R, Sharma M, Singh D, Dhiman R, Sharma P. Medial Transposition of Split Lateral Rectus Augmented With Fixation Sutures in Cases of Complete Third Nerve Palsy. Br J Ophthalmol (2016) 100(5):585–7. doi: 10.1136/bjophthalmol-2015-307583

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Hwang CJ, Eftekhari K. Teprotumumab: The First Approved Biologic for Thyroid Eye Disease. Int Ophthalmol Clin (2021) 61(2):53–61. doi: 10.1097/IIO.0000000000000353

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Sy A, Eliasieh K, Silkiss RZ. Clinical Response to Tocilizumab in Severe Thyroid Eye Disease. Ophthalmic Plast Reconstr Surg (2017) 33(3):e55–e7. doi: 10.1097/IOP.0000000000000730

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Lee MJ, Planck SR, Choi D, Harrington CA, Wilson DJ, Dailey RA, et al. Non-Specific Orbital Inflammation: Current Understanding and Unmet Needs. Prog Retin Eye Res (2021) 81:100885. doi: 10.1016/j.preteyeres.2020.100885

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Sa HS, Lee JH, Woo KI, Kim YD. IgG4-Related Disease in Idiopathic Sclerosing Orbital Inflammation. Br J Ophthalmol (2015) 99(11):1493–7. doi: 10.1136/bjophthalmol-2014-305528

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Gill HS, Moscato EE, Chang AL, Soon S, Silkiss RZ. Vismodegib for Periocular and Orbital Basal Cell Carcinoma. JAMA Ophthalmol (2013) 131(12):1591–4. doi: 10.1001/jamaophthalmol.2013.5018

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Rischin D, Khushalani NI, Schmults CD, Guminski A, Chang ALS, Lewis KD, et al. Integrated Analysis of a Phase 2 Study of Cemiplimab in Advanced Cutaneous Squamous Cell Carcinoma: Extended Follow-Up of Outcomes and Quality of Life Analysis. J Immunother Cancer (2021) 9(8):e002757. doi: 10.1136/jitc-2021-002757

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Jonas RA, Rokohl AC, Heindl LM. Targeted Therapy for Malignant Ocular Melanomas. Ann Eye Sci (2020) 6:7. doi: 10.21037/aes-20-101

CrossRef Full Text | Google Scholar

78. Ali MJ, Zetzsche M, Scholz M, Hahn D, Gaffling S, Heichel J, et al. New Insights Into the Lacrimal Pump. Ocul Surf (2020) 18(4):689–98. doi: 10.1016/j.jtos.2020.07.013

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Malhotra R, Mahadevan V, Leatherbarrow B, Barrett AW. The Post-Levator Aponeurosis Fat Pad. Ophthalmic Plast Reconstr Surg (2015) 31(4):313–7. doi: 10.1097/IOP.0000000000000337

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Tu Y, Wu S, Pan Z, Hu X, Zhou G, Shi J, et al. Endoscopic Transconjunctival Deep Lateral Wall Decompression for Thyroid-Associated Orbitopathy: A Minimally Invasive Alternative: Transconjunctival Endoscopic With Wall Decompression for TAO. Am J Ophthalmol (2022) 235:71–9. doi: 10.1016/j.ajo.2021.08.013

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Hou X, Guo Y, Li S, Lin M, Jia R, Rokohl A, et al. Lateral Tarsal Strip Procedure for Involutional Ectropion: A Retrospective Analysis of 85 Cases and a Comprehensive Literature Review. Adv Ophthalmol Pract Res (2021) 1(1):100004. doi: 10.1016/j.aopr.2021.100004

CrossRef Full Text | Google Scholar

82. Balakrishnan K, Moe KS. Applications and Outcomes of Orbital and Transorbital Endoscopic Surgery. Otolaryngol Head Neck Surg (2011) 144(5):815–20. doi: 10.1177/0194599810397285

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Prabhu SS, Chung SA, Brown PJ, Runyan CM. Utilizing 3d-Printed Orbital Floor Stamps to Create Patient-Specific Implants for Orbital Floor Reconstruction. Ophthalmic Plast Reconstr Surg (2021) 37(1):81–5. doi: 10.1097/IOP.0000000000001734

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Guo Y, Koch KR, Heindl LM. Transcaruncular Laser-Assisted StopLoss Lester Jones Tube Surgery for Lacrimal Canalicular Obstructions. Graefe's Arch Clin Exp Ophthalmol = Albrecht von Graefes Archiv fur klinische und Experimentelle Ophthalmol (2019) 257(7):1569–70. doi: 10.1007/s00417-019-04331-0

CrossRef Full Text | Google Scholar

85. Guo Y, Rokohl AC, Kroth K, Li S, Lin M, Jia R, et al. Endoscopy-Guided Diode Laser-Assisted Transcaruncular StopLoss Jones Tube Implantation for Canalicular Obstructions in Primary Surgery. Graefe's Arch Clin Exp Ophthalmol = Albrecht von Graefes Archiv fur klinische und Experimentelle Ophthalmol (2020) 258(12):2809–17. doi: 10.1007/s00417-020-04942-y

CrossRef Full Text | Google Scholar

86. Mor JM, Guo Y, Koch KR, Heindl LM. Transcanalicular Diode Laser-Assisted Dacryocystorhinostomy for the Treatment of Primary Acquired Nasolacrimal Duct Obstruction. J visualized Experiments JoVE (2017) (128):55981. doi: 10.3791/55981

CrossRef Full Text | Google Scholar

87. Rokohl AC, Guo YW, Mor JM, Loreck N, Koch KR, Heindl LM. Intubation Systems in Lacrimal Drainage Surgery a Current Overview. Klinische Monatsblatter fur Augenheilkunde (2020) 237(1):20–8. doi: 10.1055/a-0992-9966

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Kopecký A, Němčanský J, Kratky V, Rokohl AC, Heindl LM. Bioengineered Dermal Substitutes for Periocular Defects. Ann Eye Sci (2021) 6:16. doi: 10.21037/aes-20-97

CrossRef Full Text | Google Scholar

89. Ali MJ. Introducing the Concept of "Lacriome". Graefes Arch Clin Exp Ophthalmol (2021) 259(5):1087–8. doi: 10.1007/s00417-020-05049-0

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Malerba A, Klein P, Bachtarzi H, Jarmin SA, Cordova G, Ferry A, et al. PABPN1 Gene Therapy for Oculopharyngeal Muscular Dystrophy. Nat Commun (2017) 8:14848. doi: 10.1038/ncomms14848

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Van Brummen A, Owen JP, Spaide T, Froines C, Lu R, Lacy M, et al. PeriorbitAI: Artificial Intelligence Automation of Eyelid and Periorbital Measurements. Am J Ophthalmol (2021) 230:285–96. doi: 10.1016/j.ajo.2021.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Lou L, Cao J, Wang Y, Gao Z, Jin K, Xu Z, et al. Deep Learning-Based Image Analysis for Automated Measurement of Eyelid Morphology Before and After Blepharoptosis Surgery. Ann Med (2021) 53(1):2278–85. doi: 10.1080/07853890.2021.2009127

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Guo Y, Schaub F, Mor JM, Jia R, Koch KR, Heindl LM. A Simple Standardized Three-Dimensional Anthropometry for the Periocular Region in a European Population. Plast Reconstr Surg (2020) 145(3):514e–23e. doi: 10.1097/PRS.0000000000006555

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Hou X, Rokohl AC, Meinke MM, Li S, Liu J, Fan W, et al. A Novel Standardized Distraction Test to Evaluate Lower Eyelid Tension Using Three-Dimensional Stereophotogrammetry. Quantitative Imaging Med Surg (2021) 11(8):3735–48. doi: 10.21037/qims-20-1016

CrossRef Full Text | Google Scholar

95. Hou X, Rokohl AC, Meinke MM, Liu J, Li S, Fan W, et al. Standardized Three-Dimensional Lateral Distraction Test: Its Reliability to Assess Medial Canthal Tendon Laxity. Aesthetic Plast Surg (2021) 45(6):2798–807. doi: 10.1007/s00266-021-02440-y

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Guo Y, Liu J, Ruan Y, Rokohl AC, Hou X, Li S, et al. A Novel Approach Quantifying the Periorbital Morphology: A Comparison of Direct, 2-Dimensional, and 3-Dimensional Technologies. J Plast Reconstr Aesthet Surg (2021) 74(8):1888–99. doi: 10.1016/j.bjps.2020.12.003

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Liu J, Guo Y, Arakelyan M, Rokohl AC, Heindl LM. Accuracy of Areal Measurement in the Periocular Region Using Stereophotogrammetry. J Oral Maxillofac Surg (2021) 79(5):1106.e1–.e9. doi: 10.1016/j.joms.2020.12.015

CrossRef Full Text | Google Scholar

98. Liu J, Rokohl AC, Guo Y, Li S, Hou X, Fan W, et al. Reliability of Stereophotogrammetry for Area Measurement in the Periocular Region. Aesthetic Plast Surg (2021) 45(4):1601–10. doi: 10.1007/s00266-020-02091-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: ocular surface and corneal disease, cataract, glaucoma, retina, strabismus, orbit, plastics, challenge

Citation: Guo Y, Kratky V, Xie H, Shentu X, Man X, Wang Y, Wen W, Rokohl AC and Heindl LM (2022) Grand Challenges and Opportunities in Surgical Ophthalmology: Together for a Shared Future. Front. Ophthalmol. 2:922240. doi: 10.3389/fopht.2022.922240

Received: 17 April 2022; Accepted: 06 June 2022;
Published: 04 July 2022.

Edited by:

Michael Yoon, Harvard Medical School, United States

Reviewed by:

Jinhua Liu, University of Cologne, Germany

Copyright © 2022 Guo, Kratky, Xie, Shentu, Man, Wang, Wen, Rokohl and Heindl. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ludwig M. Heindl, ludwig.heindl@uk-koeln.de; Yongwei Guo, yongwei-guo@zju.edu.cn

Download