Chapter 34
Stem Cell Transplantation for Ocular Surface Diseases
SCHEFFER C.G. TSENG, EDGAR M. ESPANA and MARIO A. DI PASCUALE
Main Menu   Table Of Contents

Search

BASIC CONCEPTS
TWO TYPES OF OCULAR SURFACE FAILURE
LIMBAL STEM CELL DEFICIENCY
KEY STRATEGIES
ACKNOWLEDGEMENT
REFERENCES

BASIC CONCEPTS

OCULAR SURFACE HEALTH IS MAINTAINED BY A STABLE TEAR FILM

Anatomically, the ocular surface encompasses the entire mucosal epithelial lining bordered by the skin at the superior and inferior eyelid margins. Histologically, this epithelial surface covers two major territories (i.e., the cornea and the conjunctiva). The primary function of the ocular surface is to provide clear vision during an open-eye state. To achieve this goal while maintaining comfort and preventing microbial invasion, the ocular surface has to be covered by a stable tear film. Therefore, the mechanism by which the ocular surface health is ensured is inherently built into the relationship between ocular surface epithelia and the preocular tear film. As recently summarized,1 compositional factors including the ocular surface epithelia, meibomian glands, and lacrimal glands and hydrodynamic factors such as eyelid blinking and closure are essential to constitute ocular surface defense that governs how preocular tear film stability is maintained (Fig. 1). These two major factors are integrated via two neuronal reflex arcs, triggered by the sensory drive of the first branch of trigeminal nerve (V1), and mediated by the parasympathetic branch and the motor branch of the facial nerve (VII) as the efferent output, respectively (Fig. 2). Such a neuroanatomic integration of ocular surface defense explains how external adnexal glands and eyelids can be integrated with ocular surface epithelia as a unit to maintain a stable tear film.

Fig. 1 Ocular surface defense. Ocular surface health is maintained by a stable tear film, which requires both compositional and hydrodynamic factors. The former includes ocular surface epithelium producing mucins, meibomian glands producing meibum, and lacrimal glands producing aqueous tears. The latter includes eyelids, which blink at desirable frequencies to allow tears to spread and clear, and close completely during the close eye state to prevent evaporation.

Fig. 2 Neuroanatomic integration of compositional and hydrodynamic factors. Two neuronal reflexes are involved to integrate both factors as an integral unit to maintain a stable tear film. Both reflexes are triggered by the sensory input of the first branch of the trigeminal nerve (V1), while the compositional one is mediated by the parasympathetic branch of the facial nerve (VII), and the hydrodynamic one is mediated by the motor branch of the facial nerve.

Based on this concept, one predicts that deficiency or alteration of any of these elements involved in such neuroanatomic integration can lead to an unstable tear film, a hallmark of various forms of dry eye.2 Therefore, investigation into dysfunction of this neuroanatomic integration is the first step toward better understanding of the pathogenesis of various ocular surface disorders, and is a prerequisite before any ocular surface reconstruction.

Back to Top
TWO TYPES OF OCULAR SURFACE FAILURE

SQUAMOUS METAPLASIA

According to the resultant epithelial phenotype defined by impression cytology 3, all severe ocular surface diseases can be classified to express one of the two major types of ocular surface failure. The first type is squamous metaplasia (Fig. 3) in which a normal nonkeratinized ocular surface epithelium transforms into a skin-like keratinized epithelium.3 Because of skin-like epithelial differentiation (skin-like), the ocular surface with squamous metaplasia becomes nonwettable, a hallmark of various dry eye disorders.2 If the underlying insults are primarily derived from poor ocular surface defense, such squamous metaplasia can be reversed to a normal state when ocular surface defense is restored, indicating that epithelial progenitor cells (i.e., stem cells) of the ocular surface are not intrinsically altered. Squamous metaplasia can be caused by insults not originating from ocular surface defense. In xerophthalmia caused by systemic vitamin A deficiency,4,5 squamous metaplasia can also be reversed by replacement of vitamin A. Nevertheless, squamous metaplasia can also be caused by various insults damaging epithelial progenitor cells, the basement membrane, and the underlying stroma leading to chronic inflammation and scarring as seen in various forms of cicatricial keratoconjunctivitis such as chemical burns, Stevens-Johnson syndrome, ocular pemphigoid, etc.6,7 Except for early active ocular cicatricial pemphigoid, in which systemic immunosuppression can reverse squamous metaplasia, the rest cannot be reversed by conventional medical treatments. The irreversibility of such abnormal epithelial differentiation strongly implies that epithelial progenitor cells (i.e., stem cells [SC]), may have been intrinsically altered.

Fig. 3 Two types of ocular surface failure. In limbal stem cell deficiency, the corneal epithelium is replaced by conjunctival epithelium, while in squamous metaplasia corneal and conjunctival epithelia transform into a keratinized (skin-like) epithelium. In addition to the change in epithelial phenotype, both types of ocular surface failure are also accompanied by the destruction of the basement membrane and the inflammation and scarring of the underlying stroma.

Back to Top
LIMBAL STEM CELL DEFICIENCY
The second type of ocular surface failure is limbal stem cell deficiency (LSCD) (Fig. 3), which is characterized by the replacement of the normal corneal epithelium by an invading conjunctival epithelium.8 This pathologic state of LSCD can experimentally be created in rabbits by damaging the limbal epithelium, which contains corneal epithelial SC.9 Corneas with LSCD manifest poor epithelialization (persistent defects or recurrent erosions), chronic stromal inflammation (keratitis mixed with scarring), corneal vascularization, and conjunctival epithelial ingrowth. Consequently, patients with LSCD experience severe irritation, photophobia, and decreased vision, and are poor candidates for conventional corneal transplantation. Because most of these features can also be found in other corneal diseases, the sine qua non criterion for diagnosing LSCD is the existence of conjunctival epithelial ingrowth onto the corneal surface (conjunctivalization). Clinically, the presence of conjunctivalization may be suggested by observing the loss of limbal Vogt's palisades under slit-lamp examination10 and by occurrence of late fluorescein staining,11 reflecting poor epithelial barrier function.12 However, the definitive diagnosis of conjunctivalization relies on the detection of conjunctival goblet cells on the corneal surface by impression cytology.8 Accurate diagnosis of LSCD is crucial to guide the choice of appropriate procedures for transplanting limbal epithelial SC.

Based on the underlying etiology, corneal diseases manifesting LSCD can be subdivided into two major categories (Table 1).8 In the first category, limbal epithelial SC are destroyed by known or recognizable offenders such as a chemical or thermal burn, Stevens-Johnson syndrome/toxic epidermal necrolysis, multiple surgeries or cryotherapies or medications (iatrogenic), contact lens, severe microbial infection, radiation, and antimetabolites including 5-fluorouracil and mitomycin C.8,13–15 A second category is characterized by a gradual loss of the SC population without known or identifiable precipitating factors. In this situation, the limbal stromal niche is presumably affected and progressively deteriorates by a variety of etiologies that include aniridia and coloboma, neoplasia, multiple hormonal deficiencies, peripheral ulcerative corneal diseases, neurotrophic keratopathy and idiopathic limbal deficiency.8,10,16–18 These diseases can also be categorized by the underlying cause being hereditary or not as summarized in Table 1. The aforementioned information explains why transplantation of epithelial SC and restoration of SC stromal environment is necessary in ocular surface reconstruction.

 

TABLE 1. Ocular Surface Diseases with Limbal Stem Cell Deficiency


Clinical Diseases Destructive Loss of Limbal Stem Cells Altered Limbal Stromal Niche
Hereditary:
Anirida X
 b. Keratitis associated with multiple endocrine deficiency X
 c. Epidermal dysplasia (ectrodactyly-ectodermal dysplasia-clefting syndrome)X 
Acquired:
 a. Chemical or thermal burnsX 
 b. Stevens-Johnson syndrome, toxic epidermal necrolysisX 
 c. Multiple surgeries or cryotherapies to limbusX 
 d. Contact lens-induced keratopathyX 
 e. Severe microbial infection extending to limbusX 
 f. Antimetabolite uses (5-FU or mitomycin C)X 
 g. RadiationXX
 h. Chronic limbitis (vernal, atopy, phlyctenular) X
 i. Peripheral ulcerative keratitis (Mooren's ulcer) X
 j. Neurotrophic keratopathy X
 k. Chronic bullous keratopathy  
 l. PterygiumXX
m. Idiopathic??

5-FU, 5-fluorouracil.

 

Back to Top
KEY STRATEGIES
Considering all of the basic concepts together, we propose that the general strategy of ocular surface reconstruction is analogous to growing a flowering plant in the garden (Fig. 4). Several key strategies are undertaken: (1) to restore ocular surface defense so that a stable tear film can be maintained, analogous to providing rain/water; (2) to restore stromal substrate that is important to support existing or transplanted epithelial SC, analogous to providing a top soil; and (3) to restore epithelial SC, analogous to providing a good seed. These strategies are similarly applied to corneal and conjunctival surface reconstruction. Two major threats to successful ocular surface reconstruction are severe deficiencies in ocular surface defense leading to the lack of tear film protection and chronic inflammation or ischemia in the stroma that continue to damage the stromal substrate.

Fig. 4 General strategies of ocular surface reconstruction. Similar to growing a flowering plant in a garden, ocular surface reconstruction requires restoration of ocular surface defense to maintain a stable tear film (like rain), restoration of epithelial stem cells (SC; like seed), and restoration of stromal substrate (like soil). (Modified from Tseng SCG, Tsubota K: Important concepts for treating ocular surface and tear disorders. Am J Ophthalmol 124:825, 1997, with permission)

FIRST STRATEGY: RESTORE OCULAR SURFACE DEFENSE

Before reconstruction, one should realize that keratinization in squamous metaplasia and vascularization in LSCD are two important pathologic mechanisms to prevent such corneal complications as epithelial defects, ulcers, or melt in eyes without an unstable tear film. Therefore, one should avoid performing conventional corneal transplantation to correct these two types of corneal failures if the ocular surface defense cannot first be restored. Because ocular surface reconstruction aims to restore a clear avascular cornea, which will require a stable tear film to maintain its health (see “Basic Concepts” section), it is of utmost important to restore a sound ocular surface defense. If not restored or unable to be restored, poor ocular surface defense in these eyes represent a contraindication for corneal surface reconstruction with epithelial SC transplantation.

Deficiencies in ocular surface defense can be recognized by history taking, and external and biomicroscopic examinations, and by the use of special tests such as dynamic tear function test, dye staining and impression cytology. Measures taken to correct severe aqueous tear deficiency dry eye include punctal occlusion and frequent application of autologous serum drops for mechanical microtrauma caused by lid margin and lash abnormalities such as trichiasis, entropion, or meibomian gland orifice metaplasia, which can be corrected by high oxygen permeability (Dk) contact lens, scleral contact lens, or appropriate plastic surgeries including mucous membrane graft. Symblepharon that causes obliteration of tear meniscus, misdirected lashes, inability of adequate blinking and closure, and motility restriction should also be corrected by fornix reconstruction before corneal surface reconstruction. Exposure problems may be corrected by Botox-induced ptosis, high DK contact lens, scleral lens, or tarsorrhaphy.

SECOND STRATEGY: RESTORE STROMAL ENVIRONMENT

The state and severity of LSCD can be graded as partial or total. In partial LSCD, a portion of the limbal SC remains intact. In partial or total LSCD, if the central cornea still preserves a normal corneal epithelial phenotype and satisfactory vision, treatments should be directed to maintaining the remaining corneal epithelium and to activating the remaining limbal SC. Besides bandage contact lens or scleral lens to shield against continuous attrition, one new way of expanding remaining limbal or conjunctival epithelial SC in vivo is to transplant preserved amniotic membrane to restore stromal environment that has been destroyed in diseases mentioned in Table 1.

Amniotic membrane (i.e., the innermost layer of the placental membrane) consists of a simple epithelium, a thick basement membrane, and an avascular stroma. When appropriately procured and processed, cryopreserved amniotic membrane can be used as a surgical graft for substrate replacement and does not elicit immunologic reactions.19 A number of studies have shown that amniotic basement membrane facilitates epithelialization and maintains epithelial phenotype, while amniotic avascular stroma exerts anti-inflammatory, antiangiogenic and antiscarring effects [for reviews see Dua and Azuara-Blanco,20 Kruse et al.,21 Sippel et al.,22 and Tseng and Tsubota23), properties important for reconstruction of epithelium-lining tissues. For partial LSCD, debridement of conjunctivalized epithelium from the corneal surface24 with or without additional amniotic membrane transplantation25 is effective to restore the corneal surface. Because of its anti-inflammatory effect, amniotic membrane transplantation as a temporary patch (bandage) is also effective in restoring ocular surfaces during the acute attack of chemical burns26 and Stevens-Johnson syndrome27.

THIRD STRATEGY: RESTORE EPITHELIAL STEM CELLS

For corneal surface reconstruction, epithelial SC can be transplanted from the limbal epithelium. For conjunctival surface reconstruction, epithelial SC can be transplanted from the bulbar conjunctival epithelium, especially close to the fornix, where conjunctival epithelial SC are enriched.28 The following section is devoted to restoring corneal surfaces with LSCD.

When LSCD is total, that is, involving the entire limbal SC of an eye, amniotic transplantation alone is ineffective, and corneal surface reconstruction requires transplantation of limbal epithelial SC. According to the type and source of tissue removed to transplant limbal epithelial SC, several surgical procedures have been devised, of which each terminology used herein adopts what has been recommended by Holland and Schwartz.29

Conjunctival Limbal Autograft

When total LSCD is unilateral, conjunctival limbal autograft (CLAU) from the uninvolved fellow eye is advised.30 The surgical procedure for CLAU is shown in Figure 5. (To view surgical video, please contact Dr. Tseng at stseng@ocularsurface.com.) In brief, the conjunctivalized pannus is removed from the corneal surface by peritomy followed by superficial keratectomy with blunt dissection in the recipient eye (Fig. 5A). The cicatrix was removed from the subconjunctival space (Fig. 5B). This invariably results in the recession of the conjunctival edge to 3 to 5 mm from the limbus (Fig. 5C). Two strips of limbal conjunctival-free grafts, each spanning 6 to 7 mm limbal arc length, are removed by superficial lamellar keratectomy at 1 mm within the limbus from the superior and inferior limbal regions (Fig. 5D) and by including 5 mm of adjacent conjunctiva. These two free grafts are transferred and secured to the recipient eye at the corresponding anatomic sites by interrupted 10-0 nylon sutures to the limbus and 8-0 vicryl sutures to the sclera. The size of the limbal removal in CLAU can be adjusted according to the visual potential of the donor eye and the extent of LSCD in the recipient eye, and the amount of the conjunctiva can be increased if the recipient eye also requires symblepharon lysis. Figure 6 illustrates how such a case with unilateral acid burn improved visual acuity, and maintained a smooth and stable corneal surface without vascularization and a clearer corneal stroma following CLAU (depicted in Fig. 5) without corneal transplantation.

Fig. 5 Surgical procedure for conjuctival limbal autograft (CLAU). The conjunctivalized pannus is removed from the corneal surface by peritomy followed by superficial keratectomy with blunt dissection in the recipient eye (A). The cicatrix was removed from the subconjunctival space (B). This invariably results in the recession of the conjunctival edge to 3 to 5 mm from the limbus from the superior and inferior limbal regions (C). Two strips of limbal conjunctival free grafts, each spanning 6 to 7 mm limbal arc length, are removed by superficial lamellar keratectomy at 1 mm within the limbus (D) and by including 5 mm of adjacent conjunctiva. These two free grafts are transferred and secured to the recipient eye at the corresponding anatomic sites by interrupted 10-0 nylon sutures to the limbus and 8-0 vicryl sutures to the sclera.

Fig. 6 Clinical result of conjunctival limbal autograft (CLAU). In this representative case (surgical procedure of CLAU is shown in Fig. 5), the left eye with unilateral acid burn manifested total limbal stem cell deficiency (LSCD) with visual acuity of 20/200, persistent corneal epithelial defect, conjunctivalization, and vascularization for 7 months (A). After CLAU from the right eye, rapid epithelialization started from superior and inferior grafts in 1 week (B) and completed in 2 weeks (C). The corneal surface was stable and improved 6 weeks after CLAU (D). The midstromal vascularization that was left 1 month after CLAU (E) became regressed 6 months later (F), resulting in visual acuity of 20/30 with a stable and smooth surface and a clearer stroma in 2 years (G). The donor eye healed well without complication at the limbal region (H).

In chemical burns, severe inflammation and ischemia in the acute stage is a threat against the success of transplanted CLAU, as noted in a number of studies,30,31 For this reason, CLAU is not recommended. Nevertheless, as aforementioned, transplantation of amniotic membrane as a temporary patch is an alternative because it can suppress the intense inflammation, facilitate epithelial wound healing, and prevent scrring in acute burns26 and Stevens-Johnson syndrome/toxic epidermal necrolysis.27 Although it is generally believed that the donor eyes with such limbal removal recover well without complication (Fig. 6H), scattered reports show that some donor eyes may become decompensated with pseudopterygium or partial LSCD, especially in eyes with subclinical LSCD. Figure 7 shows one such case. To preclude such a potential complication, one alternative is to transplant amniotic membrane as a graft to cover the defect after removal of CLAU in the donor eye and over the corneal surface before CLAU in the recipient eye so that the remaining and transplanted limbal epithelial SC can be expanded in the donor and recipient eye, respectively.32 Figure 8 illustrates how amniotic membrane transplantation can resurrect the corneal surface even after the removal over 11 clock hours of the limbus in the donor eye, and how both donor and recipient eyes can be helped after CLAU.

Fig. 7 Conjuctival limbal autograft (CLAU) complication. In this patient with Stevens-Johnson syndrome, limbal stem cell deficiency (LSCD) was thought to be limited to the right eye. Despite CLAU from the left eye, his right eye did not recover useful vision (A). Importantly, the donor left eye showed pseudopterygium (B) with irregular epithelium and reduced vision (C). Fluorescein staining showed late staining decorating an area greater than the pseudopterygium (D). Impression cytology confirmed the migration of conjunctival epithelium with goblet cells on the corneal surface (insets of C and D), indicating the development of LSCD.

Fig. 8 Conjuctival limbal autograft (CLAU) with amniotic membrane transplantation. This patient with glaucoma developed drug-induced pseudopemphigoid manifesting total limbal stem cell deficiency (LSCD) with vascularization and epithelial defect in the right eye (A). She received keratolimbal allograft transplant (KLAL) and amniotic membrane transplantation with improved surface that last for 1.5 years with systemic cyclosporin (B), but developed recurrent inflamed ocular surface, LSCD, and epithelial defect (C). Two large strips of CLAU spanning nearly 11 clock hours were removed from the left eye (without visual potential) and transplanted to the right eye over a layer of amniotic membrane (D). Five days later, rapid epithelialization was noted on the amniotic membrane covered cornea from both superior and inferior CLAU (E). This resulted in an improved vision with a stable surface 2.5 years later (F). Because of the large limbal removal, the donor cornea decompensated with surface breakdown 5 days after CLAU (G). Nevertheless, because of the covering of amniotic membrane, a stable surface was recovered 2.5 years post-CLAU. (Modified from Meallet MA, Espana EM, Grueterich M, et al: Amniotic membrane transplantation for recipient and donor eyes undergoing conjunctival limbal autograft for total limbal stem cell deficiency. Ophthalmology 110:1585, 2003, with permission)

Living-Related Conjunctival Limbal Allograft Transplant

When total LSCD is bilateral, corneal surface reconstruction relies on transplantation of allogeneic limbal epithelial SC. To do so, one option is to living-related conjuctival limbal allograft transplant (lr-CLAL). The surgical procedure of lr-CLAL is identical to CLAU. Amniotic membrane can be used similarly to eliminate the concern of removing limbal SC from the healthy donor eye and to augment the effect of CLAU in the recipient eye. Nevertheless, unless the donor and the recipient are perfectly matched, the success of lr-CLAL depends on systemic immunosuppression and allograft rejection is still the main threat.33

Keratolimbal Allograft Transplant

The other option is to perform keratolimbal allograft (KLAL) from cadaveric donors. This surgical technique may restore SC in patients with bilateral LSCD or in patients with unilateral LSCD who do not wish to jeopardize the healthy eye with any surgical procedure including CLAU. Because of allogeneic transplantation, it is mandatory to administer systemic immunosuppression in the same manner as in lr-CLAL. Despite continuous oral administration of cyclosporin A (CSA), Tsubota et al.34 and Solomon et al.35reported that the long-term success of KLAL is approximately 40% to 50% in 3 to 5 years, while Ikari and Daya36 reported a 21.2% success rate in 5 years of follow-up. The lower success rate may be contributed by serveral limiting factors. The first limiting factor is that the ocular surface defense is not fully restored in some patients (see “First Strategy” section). This concept is suggested in a study showing that severe aqueous tear deficiency dry eye is a major cause leading to the failure,37 and in another study showing that uncorrected lid abnormalities are associated with failure.35 The second limiting factor is that oral CSA alone may not be sufficient to achieve adequate immunosuppression. As recently reported,34–36,38,39 among all diseases with total LSCD, Stevens-Johnson syndrome/toxic epidermal necrolysis has the worst prognosis when treated with KLAL or lr-CLAL. One reason for such a poor prognosis is the presence of chronic inflammation, which may enhance sensitization leading to allograft rejection. Indeed, ocular surface inflammation is an important clinical finding used to grade the severity of ocular surface diseases.40 Amniotic membrane transplantation has been used to augment the success of KLAL based on the fact that it may help suppress inflammation, and restore the damaged limbal stromal environment.38,41,42 Allograft rejection remains to be the most important cause limiting the success of KLAL. Signs of allograft rejection include telangiectatic and engorged limbal blood vessels, epithelial rejection lines and epithelial breakdown in severe limbal inflammation (Fig. 9). We thus believe that a combination of several immunosuppressive agents will have to be administered for lr-CLAL or KLAL in a manner similar to what is used in other solid organ transplantations.43 Table 2 describes one such protocol of immunosuppressive regimen and monitoring. It is advised that patients who are to receive such immunosuppressive regimen be screened and monitored by qualified internists, hematologists, rheumatologists, or oncologists, and be informed about all potential side effects. Furthermore, serologic screening of prior DNA viruses, and prophylactic use of oral Acyclovir are also important. As the field of immunosuppression evolves rapidly, we believe that there are other protocols that may improve the outcome of KLAL in the future. We also believe new techniques to suppress stromal inflammation are needed in the future. These new strategies of KLAL have been summarized in a recent review.44

Fig. 9 Allograft rejection of keratolimbal allograft transplant (KLAL) and management. A. A 77-year-old male underwent KLAL elsewhere and was treated with mycophelonate (CellCept®) 1 g/d. B and C. He presented on a routine follow-up visit with perilimbal redness, vessel engorgement with hemorrhage, and progressive ulceration. D. Postoperative view of a quiescent eye after a repeat penetrating keratoplasty (PKP) and institution of mycophelonate, prednisone and cyclosporine using the regimen described in Table 2. E. A 22-year-old male with chemical burn received KLAL elsewhere and presented with reduced vision, redness, and photophobia while under oral mycophelonate alone. He was noted to have vessel engorgement and ulceration with a 3 × 4 mm corneal epithelial defect (see inset). F. Impression cytology showed lymphocyte infiltration at the border of ulceration (see arrows) and no conjunctivalization, supporting the diagnosis of immune rejection and not total limbal stem cell deficiency (LSCD). Lymphocyte infiltration 100× magnification (inset). G and H. Two weeks after addition of cyclosporine and prednisone, the limbal inflammation and redness was reduced and the ulceration ceased. (Modified from Espana EM, Di Pascuale M, Grueterich M, et al: Keratolimbal allograft for corneal surface reconstruction. Eye 110:481, 2003, with permission)

 

TABLE 2. A Recent Immunosuppressive Regimen Used for Keratolimbal Allograft Transplant


Medication Dose Monitoring Treatment
Prednisone0.5 mg/kg per day POGlucose level every monthStarting 3 days prior to surgery and discontinued 2 to 3 months later
Cyclosporin A (CsA)2.5 mg/kg per day POCreatinine and CsA level every monthStarting 3 days prior to surgery
CellCept® (Mycophelonate)2 g/d POCBC, platelets and liver profile every monthStarting 2–3 weeks prior to surgery
Prograf® (FK 506)0.2 mg/kg per day POCreatinine and blood level every monthUsed instead of CsA with less toxicity, and starting 3 days prior to surgery

*Acyclovir 200 mg 5/d PO is used as a prophylactic measure against viral activation starting three days before surgery.
CBC, complete blood cell count; PO, orally.

 

The surgical procedure of KLAL is schematically depicted in Figure 10 (To view surgical video, please contact Dr. Tseng at stseng@ocularsurface.com). It should be noted that after the pannus is removed, the remaining corneal stroma is frequently clear especially in those eyes without ulceration and endothelial dysfunction (Fig. 11), resulting in significant improvement of vision without corneal transplantation. Nevertheless, if the remaining corneal stroma still has a deep scar, visual rehabilitation will require additional corneal transplantation (Fig. 12). If corneal transplantation will have to be performed, it is best to do so as a separate procedure 3 or 4 months later when the eye is not as inflamed (Fig. 12) because there is a high rejection rate for corneal transplants when combined with KLAL at the same setting.35,36,42,45 To avoid this complication, another solution is to perform deep lamellar keratoplasty rather than penetrating keratoplasty especially if there is no corneal endothelial dysfunction.

Fig. 10 Surgical procedure of keratolimbal allograft (KLAL). In the host eye the fibrovascular pannus is completely removed leaving in most of the limbal stem cell deficiency (LSCD) cases a clear residual corneal stromal bed. One layer of amniotic membrane with basal membrane up is place over the cornea as a graft and secure with interrupted 8-0 vicryl to residual conjunctiva and scleral tissue around the limbus. The donor central corneal button is removed by trephine and the residual limbal ring is trimmed off and the underlying stroma is thinned to create a smooth and thin corneal–scleral limbal ring. The limbal tissue is then lay around cornea and secure with interrupted 10-0 nylon suture. In order to promote corneal epithelial healing another amniotic membrane is placed over the cornea as a patch and secure to the scleral with running 10-0 nylon for 1 or 2 weeks (figure not shown). If amniotic membrane is dissolved before 2 weeks, exposure and/or severe inflammation should be suspected and addressed. (Reprinted from Tsubota K, Satake Y, Kaido M, et al: Treatment of severe ocular surface disorders with corneal epithelial stem-cell transplantation. N Engl J Med 340:1697, 1999, with permission)

Fig. 11 Clinical cases of removal of corneal pannus with superficial keratectomy. Preoperative appearance of Stevens-Johnson syndrome (A), a chemical burn (C) and a Stevens Johnson syndrome (E), and their immediate postoperative appearance (B, D, and F, respectively), after the removal of pannus with superficial lamellar keratectomy.

Fig. 12 Clinical cases of keratolimbal allograft transplant (KLAL) with amniotic membrane transplantation with or without corneal transplantation. This case of Stevens-Johnson syndrome (A) received punctal occlusion and autologous serum drops, and KLAL and amniotic membrane transplantation (B). Amniotic membrane became clear and no corneal transplant was needed (C). This eye with chemical burn and band keratopathy (D) also received punctal occlusion and autologous serum drops, and KLAL and amniotic membrane transplantation (E). However, corneal transplantation with extracapsular cataract extraction (ECCE) and intraocular lens (IOL) was needed to restore a clear cornea and vision because of deep corneal scar and edema and cataract (F). This eye with chemical burn presenting with persistent corneal epithelial defect and symblepharon in the upper fornix (marked by a white star, G) first received fornix reconstruction by amniotic membrane transplantation (AMT) and intraoperative 0.04% Mitomycin C and KLAL (H). However, corneal transplantation with ECCE and IOL was needed to restore a clear cornea and vision because of deep corneal scar and edema and cataract (I). All patients were under systemic immunosuppression using regimens listed in Table 2.

Ex Vivo Expansion of Limbal Stem Cells

Another new procedure of transplanting limbal epithelial SC is via ex vivo expansion of limbal SC. This technique, first demonstrated by Pellegrini et al.46,47 using 3T3 fibroblast feeder layer, can also be achieved by amniotic membrane with or without 3T3 fibroblast feeder layers for autologous48–51 or allogeneic52,53 limbal SC transplantation for treating total LSCD. The surgical procedure is schematically shown in Figure 13. A representative case is shown in Figure 14, which has been shown to result in a limbal epithelial phenotype on the corneal surface.51 The theoretical advantage over the autologous limbal SC transplantation (i.e., CLAU) or living-related allogeneic limbal SC transplantation (i.e., lr-CLAL) is that only a small limbal biopsy is needed, thus minimizing the risk to the donor eye. The theoretical advantage over the allogeneic limbal SC transplantation (i.e., KLAL or lr-CLAL) is that the allograft rejection might be reduced because only epi-thelial cells are transplanted, and antigen-presenting Langerhan's cells are eliminated during ex vivo expansion.

Fig. 13 Schematic drawing of ex vivo expansion of limbal stem cells (SC). From the donor eye a small limbal biopsy is performed (A). One or two small explants (1 to 2 mm) are seeded on amniotic membrane in a culture medium (B). After 2 to 3 weeks, the explant yields epithelial outgrowth up to 2 to 3 cm of diameter (C). After the fibrovascular pannus is removed the explant is placed on the cornea (figures not shown). Preoperative picture shows limbal stem cell deficiency (LSCD) with central scar (D), and postoperative photo shows a smooth and clear cornea without corneal transplantation. (Reprinted from Tsai RJF, Li L-M, et al: Reconstruction of damaged corneas by transplantation of autologous limbal epithelial cells. N Eng J Med 343:86, 2000, with permission)

Fig. 14 Ex vivo expansion. A. In the acute stage, the left eye presented a total corneal and conjunctival epithelial defect except the temporal bulbar conjunctiva, stromal edema, and limbal ischemia. B. Three months after AMT as a patch resulted in total healing with superficial corneal vascularization (see arrows). C. Recurrent epithelial breakdown occurred because of total limbal stem cell deficiency (LSCD), which was diagnosed by the presence of conjunctival goblet cells (i.e., conjunctivalization, of the cornea [Fig. 14C, inset]) using im-pression cytology. D. Keratolimbal allograft transplant (KLAL) was per-formed 7.5 months after the acute insult. Nevertheless one segment of the KLAL showed irreversible rejection with dissolution (see arrows) despite continuous oral cyclosporin. E. A small biopsy measuring 3 × 2mm was removed from his healthy right eye (see arrows) and placed on AM fastened to a culture insert. After three weeks of culturing a confluent layer of approximately 400 mm2 was obtained (Fig. 14E, inset). F. One week after transplantation of the composite AM graft with expanded limbal epithelium, the ocular surface was smooth and intact, with some blood trapped underneath. G. Two months later the ocular surface remained quite and smooth without any epithelial defect (Fig. 14G, inset), and the corneal transparency had markedly improved. H: The corneal epithelium remained intact without conjunctivalization or epithelial breakdown 21 months after a penetrating keratoplasty. (Modified from Grueterich M, Espana EM, Touhami A, et al: Phenotypic study of a case with successful transplantation of ex vivo expanded human limbal epithelium for unilateral total limbal stem cell deficiency. Ophthalmology 109:1547, 2002, with permission.)

Unlike transplanting amniotic membrane, which is classified by the Food and Drug Administration (FDA) as a tissue, and hence does not require premarket approval for its use in ocular surface reconstruction, ex vivo expansion needs FDA approval for clinical uses in the United States. Dr. Tseng has obtained an Investigation New Drug (IND) No. 10313 application from the FDA to conduct a phase 1 clinical trial. Once the safety and efficacy is demonstrated in clinical trials, one may envision this new procedure may one day improve the outcome of ocular surface reconstruction.

Back to Top
ACKNOWLEDGEMENT
The basic science portion of this study was supported by Public Health Service Research Grant EY06819 (to S.C.G.T. via TissueTech, Inc.) from the Department of Health and Human Services, National Eye Institute, National Institute of Health, Bethesda, Maryland; preparation of this manuscript is supported by a grant from Ocular Surface Research & Education Foundation.
Back to Top
REFERENCES

1. Tseng SCG, Tsubota K: Important concepts for treating ocular surface and tear disorders. Am J Ophthalmol 124:825, 1997

2. Lemp MA: Report of the National Eye Institute/Industry Workshop on clinical trials in dry eyes. CLAO J 21:4, 1995

3. Tseng SCG: Staging of conjunctival squamous metaplasia by impression cytology. Ophthalmology 92:728, 1985

4. Hatchell DL, Sommer A: Detection of ocular surface abnormalities in experimental vitamin A deficiency. Arch Ophthalmol 102:1389, 1989

5. Wittpenn JR, Tseng SCG, Sommer A: Detection of early xerophthalmia by impression cytology. Arch Ophthalmol 104:237, 1986

6. Nelson JD: Ocular surface impression using cellulose acetate filter material. Ocular pemphigoid. Surv Ophthalmol 27:67, 1982

7. Nelson JD, Wright JC: Conjunctival goblet cell densities in ocular surface disorders. Arch Ophthalmol 102:1049, 1984

8. Puangsricharern V, Tseng SCG: Cytologic evidence of corneal diseases with limbal stem cell deficiency. Ophthalmology 102:1476, 1995

9. Schermer A, Galvin S, Sun T-T: Differentiation-related expression of a major 64K corneal keratin in vivo and in culture suggests limbal location of corneal epithelial stem cells. J Cell Biol 103:49, 1986

10. Nishida K, Kinoshita S, Ohashi Y, et al: Ocular surface abnormalities in aniridia. Am J Ophthalmol 120:368, 1995

11. Dua HS, Gomes JAP, Singh A: Corneal epithelial wound healing. Br J Ophthalmol 78:401, 1994

12. Huang AJW, Tseng SCG, Kenyon KR: Alteration of epithelial paracellular permeability during corneal epithelial wound healing. Invest Ophthalmol Vis Sci 31:429, 1990

13. Fujishima H, Shimazaki J, Tsubota K: Temporary corneal stem cell dysfunction after radiation therapy. Br J Ophthalmol 80:911, 1996

14. Schwartz GS, Holland EJ: Iatrogenic limbal stem cell deficiency. Cornea 7:31, 1998

15. Pires RTF, Chokshi A, Tseng SCG: Amniotic membrane transplantation or limbal conjunctival autograft for limbal stem cell deficiency induced by 5-fluorouracil in glaucoma surgeries. Cornea 19:284, 1999

16. Gass JDM: The syndrome of keratoconjunctivitis, superficial moniliasis, idiopathic hypoparathyroidism and Addison's disease. Am J Ophthalmol 54:660, 1962

17. Espana EM, Raju V, Tseng SC: Focal limbal stem cell deficiency corresponding to an iris coloboma. Br J Ophthalmol 86:1451, 2002

18. Espana EM, Grueterich M, Romano AC, et al: Idiopathic limbal stem cell deficiency. Ophthalmology 109:2004, 2002

19. Kruse FE, Joussen AM, Rohrschneider K, et al: Cryoperserved human amniotic membrane for ocular surface reconstruction. Graefe's Arch Clin Exp Ophthalmol 238:68, 2000

20. Dua HS, Azuara-Blanco A: Amniotic membrane transplantation. Br J Ophthalmol 83:748, 1999

21. Kruse FE, Voelcker HE, Rohrschneider K: Transplantation of amniotic membrane for reconstruction of the eye surface. Ophthalmologe 95:114, 1998

22. Sippel KC, Ma JJK, Foster CS: Amniotic membrane surgery. Curr Opin Ophthalmol 12:269, 2001

23. Tseng SCG, Tsubota K: Amniotic membrane transplantation for ocular surface reconstruction. In Holland EJ, Mannis MJ, eds. Ocular Surface Diseases: Medical and Surgical Management. New York: Springer, 2001:226–231

24. Dua HS: The conjunctiva in corneal epithelial wound healing. Br J Ophthalmol 82:1407, 1998

25. Anderson DF, Ellies P, Pires RTF, et al: Amniotic membrane transplantation for partial limbal stem cell deficiency: long term outcomes. Br J Ophthalmol 85:567, 2001

26. Meller D, Pires RTF, Mack RJS, et al: Amniotic membrane transplantation for acute chemical or thermal burns. Ophthalmology 107:980, 2000

27. John T, Foulks GN, John ME, et al: Amniotic membrane in the surgical management of acute toxic epidermal necrolysis. Ophthalmology 109:351, 2002

28. Wei Z-G, Wu R-L, Lavker RM, et al: In vitro growth and differentiation of rabbit bulbar, fornix, and palpebral conjunctival epithelia. Implication on conjunctival epithelial transdifferentiation and stem cells. Invest Ophthalmol Vis Sci 34:1814, 1993

29. Holland EJ, Schwartz GS: The evolution of epithelial transplantation for severe ocular surface disease and a proposed classification system. Cornea 15:549, 1996

30. Kenyon KR, Tseng SCG: Limbal autograft transplantation for ocular surface disorders. Ophthalmology 96:709, 1989

31. Ronk JF, Ruiz-Esmenjaud S, Osorio M, et al: Limbal conjunctival autograft in a subacute alkaline corneal burn. Cornea 13:465, 1994

32. Meallet MA, Espana EM, Grueterich M, et al: Amniotic membrane transplantation for recipient and donor eyes undergoing conjunctival limbal autograft for total limbal stem cell deficiency. Ophthalmology 110:1585, 2003

33. Daya SM, Ilari L: Living related conjunctival limbal allograft for the treatment of stem cell deficiency. Ophthalmology 108:126, 2001

34. Tsubota K, Satake Y, Kaido M, et al: Treatment of severe ocular surface disorders with corneal epithelial stem-cell transplantation. N Engl J Med 340:1697, 1999

35. Solomon A, Ellies P, Anderson DF, et al: Long-term outcome of kerarolimbal allograft with or without penetrating keratoplasty for total limbal stem cell deficiency. Ophthalmology 109:1159, 2002

36. Ikari L, Daya SM: Long-term outcomes of keratolimbal allograft for the treatment of severe ocular surface disorders. Ophthalmology 109:1278, 2002

37. Shimazaki J, Shimmura S, Fujishima H, et al: Association of preoperative tear function with surgical outcome in severe Stevens-Johnson syndrome. Ophthalmology 107:1518, 2000

38. Tsubota K, Satake Y, Ohyama M, et al: Surgical reconstruction of the ocular surface in advanced ocular cicatricial pemphigoid and Stevens-Johnson syndrome. Am J Ophthalmol 122:38, 1996

39. Mita T, Yamashita H, Kaji Y, et al: Functional difference of TGF-β isoforms regulating corneal wound healing after excimer laser keratectomy. Exp Eye Res 68:513, 1999

40. Schwartz GS, Gomes JAP, Holland EJ: Preoperative Staging of Disease Severity. In Holland EJ, Mannis MJ, eds. Ocular Surface Disease. New York: Springer-Verlag, 2002:158–167

41. Shimazaki J, Yang H-Y, Tsubota K: Amniotic membrane transplantation for ocular surface reconstruction in patients with chemical and thermal burns. Ophthalmology 104:2068, 1997

42. Tseng SCG, Prabhasawat P, Barton K, et al: Amniotic membrane transplantation with or without limbal allografts for corneal surface reconstruction in patients with limbal stem cell deficiency. Arch Ophthalmol 116:431, 1998

43. Holland EJ, Schwartz GS: Changing concepts in the management of severe ocular surface disease over twenty-five years. Cornea 19:688, 2000

44. Espana EM, Di Pascuale M, Grueterich M, et al: Keratolimbal allograft for corneal surface reconstruction. Eye 110:481, 2003

45. Shimazaki J, Maruyama F, Shimmura S, et al: Immunologic rejection of the central graft after limbal allograft transplantation combined with penetrating keratoplasty. Cornea 20:149, 2001

46. Pellegrini G, Traverso CE, Franzi AT, et al: Long-term restoration of damaged corneal surface with autologous cultivated corneal epithelium. Lancet 349:990, 1997

47. Rama P, Bonini S, Lambiase A, et al: Autologous fibrin-cultured limbal stem cells permanently restore the corneal surface of patients with total limbal stem cell deficiency. Transplantation 72:1478, 2001

48. Tsai RJF, Li L-M, Chen J-K: Reconstruction of damaged corneas by transplantation of autologous limbal epithelial cells. N Engl J Med 343:86, 2000

49. Schwab IR: Cultured corneal epithelia for ocular surface disease. Trans Am Ophthalmol Soc 97:891, 1999

50. Schwab IR, Reyes M, Isseroff RR: Successful transplantation of bioengineered tissue replacements in patients with ocular surface disease. Cornea 19:421, 2000

51. Grueterich M, Espana EM, Touhami A, et al: Phenotypic study of a case with successful transplantation of ex vivo expanded human limbal epithelium for unilateral total limbal stem cell deficiency. Ophthalmology 109:1547, 2002

52. Koizumi N, Inatomi T, Suzuki T, et al: Cultivated corneal epithelial transplantation for ocular surface reconstruction in acute phase of Stevens-Johnson syndrome. Arch Ophthalmol 119:298, 2001

53. Koizumi N, Inatomi T, Suzuki T, et al: Cultivated corneal epithelial stem cell transplantation in ocular surface disorders. Ophthalmology 108:1569, 2001

Back to Top