Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
IMPROVING TRABECULECTOMY OUTCOME
Document Type and Number:
WIPO Patent Application WO/2013/079713
Kind Code:
A1
Abstract:
The current invention relates to the improvement of trabeculectomy surgery. The improvement more specifically resides in an extended lifetime of the sclera-corneal drainage channel created by trabeculectomy surgery. The improvement is obtained by post-surgical administration of an anti-PlGF antibody or fragment thereof.

Inventors:
JONCKX BART (BE)
VAN BERGEN TINE (BE)
STALMANS INGEBORG (BE)
Application Number:
PCT/EP2012/074195
Publication Date:
June 06, 2013
Filing Date:
December 03, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
THROMBOGENICS NV (BE)
JONCKX BART (BE)
VAN BERGEN TINE (BE)
STALMANS INGEBORG (BE)
International Classes:
A61K39/395; C07K16/22; A61P27/00
Domestic Patent References:
WO2011023805A12011-03-03
WO2010097800A12010-09-02
WO2004052228A22004-06-24
WO2009073457A12009-06-11
WO2009067407A22009-05-28
WO2011023805A12011-03-03
WO2001085796A22001-11-15
WO2006099698A22006-09-28
WO2001004269A12001-01-18
WO2005016455A22005-02-24
Foreign References:
US5304118A1994-04-19
Other References:
LI Z ET AL: "Role of vascular endothelial growth factor and placental growth factor in glaucoma and scar formation after glaucoma filtration surgery", 5 October 2006 (2006-10-05), XP008159543, Retrieved from the Internet [retrieved on 20130123]
VAN BERGEN T ET AL: "Role of placental growth factor (PLGF) in wound healing after glaucoma filtration surgery.", BULLETIN DE LA SOCIÉTÉ BELGE D'OPHTALMOLOGIE 2011, no. 317, 1 January 2011 (2011-01-01), pages 65 - 66, XP002690835, ISSN: 0081-0746
TINE VAN BERGEN ET AL: "The role of different VEGF isoforms in scar formation after glaucoma filtration surgery", EXPERIMENTAL EYE RESEARCH, ACADEMIC PRESS LTD, LONDON, vol. 93, no. 5, 29 August 2011 (2011-08-29), pages 689 - 699, XP028115139, ISSN: 0014-4835, [retrieved on 20110901], DOI: 10.1016/J.EXER.2011.08.016
QUIGLEY, BR J OPHTHALMOL, vol. 80, 1996, pages 389 - 393
"Collaborative Normal-Tension Glaucoma Study Group", AM J OPTHALMOL, vol. 126, 1998, pages 487 - 497
BURR ET AL., COCHRANE DATABASE SYST REV, vol. 18, no. 2, 2005, pages CD004399
HITCHINGS, ARCH OPHTHALMOL, vol. 116, 1998, pages 241 - 242
ADDICKS ET AL., ARCH OPHTHALMOL, vol. 101, 1983, pages 795 - 798
LEE ET AL., J OCUL PHARMACOL THER, vol. 11, 1995, pages 227 - 232
LAMA; FECHTNER, SURV OPHTHALMOL, vol. 48, 2003, pages 314 - 346
HITCHINGS; GRIERSON, TRANS OPHTHALMOL SOC UK, vol. 103, 1983, pages 84 - 88
SKUTA; PARRISH, SURV OPHTHALMOL, vol. 32, 1987, pages 149 - 170
JAMPEL ET AL., ARCH OPHTHALMOL, vol. 106, 1988, pages 89 - 94
GILLIES; SU, AUST NZ J OPHTHALMOL, vol. 19, 1991, pages 299 - 304
KATZ ET AL., OPHTHALMOL, vol. 102, 1995, pages 1263 - 1269
GREENFIELD ET AL., ARCH OPHTHALMOL, vol. 116, 1998, pages 443 - 447
CORDEIRO ET AL., GENE THER, vol. 10, 2003, pages 59 - 71
KWAH; GREHN, OPHTHALMOL, vol. 114, 2007, pages 1822 - 1830
KING ET AL., BR J OPHTHALMOL, vol. 91, 2007, pages 873 - 877
STALMANS ET AL., N ENGL J MED, vol. 367, 2012, pages 606 - 615
VAN BERGEN ET AL., EXP EYE RES, vol. 93, 2011, pages 689 - 699
VEGF-A. LI ET AL., INVEST OPHTHALMOL VIS SCI, vol. 50, 2009, pages 5217 - 5225
LI ET AL., INVEST OPHTHALMOL VIS SCI, vol. 50, 2009, pages 5217 - 5225
"Remington's Pharmaceutical Sciences", 2000, LIPPINCOTT, WILLIAMS & WILKINS
Download PDF:
Claims:
An anti-PlGF antibody or fragment thereof for enhancing the success rate of trabeculectomy surgery of an eye, for treating filtration failure after trabeculectomy surgery of an eye, or for preventing, reducing or retarding the occurrence of filtration failure after trabeculectomy surgery of an eye.

The anti-PlGF antibody or fragment thereof according to claim 1 which is in a pharmaceutically acceptable formulation capable of being administered to an eye.

The anti-PlGF antibody or fragment thereof according to claim 2 which is in a pharmaceutically acceptable formulation capable of being administered into the anterior chamber of an eye.

The anti-PlGF antibody or fragment thereof according to claim 2 which is in a pharmaceutically acceptable formulation capable of being administered into the bleb created by the trabeculectomy surgery.

The anti-PlGF antibody or fragment thereof according to claim 1 wherein said treating of filtration failure after trabeculectomy surgery of an eye, or said preventing, reducing or retarding of the occurrence of filtration failure after trabeculectomy surgery of an eye results from administering to said eye at least a single dose of an effective amount of said anti-PlGF antibody or fragment thereof.

The anti-PlGF antibody or fragment thereof according to any one of claims 1 to 5 which is in a pharmaceutically acceptable formulation further comprising one or more of an agent for controlling the intra-ocular pressure, an anti-inflammatory agent, an antiviral agent, an antibacterial agent, an antiviral agent, an anti-angiogenic agent, an anti-mitotic agent, an antihistamine, an anesthetic, an agent to induce mydriasis and an agent to induce cycloplegia.

7. The anti-PlGF antibody or fragment thereof according to claim 5 wherein said eye is contacted further with one or more agents chosen from an agent for controlling the intraocular pressure, an anti-inflammatory agent, an antiviral agent, an antibacterial agent, an antiviral agent, an anti-angiogenic agent, an anti-mitotic agent, an antihistamine, an anesthetic, an agent to induce mydriasis and an agent to induce cycloplegia.

8. The anti-PlGF antibody or fragment thereof according to claim 7 wherein said further contacting is occurring prior to, concurrent with, or after administering the anti-PlGF antibody or fragment thereof.

9. The anti-PlGF antibody or fragment thereof according to claim 1 wherein said treating of filtration failure after trabeculectomy surgery of an eye, or said preventing, reducing or retarding of the occurrence of filtration failure after trabeculectomy surgery of an eye results from administering into the anterior chamber of an eye multiple doses of an effective amount of said anti-PlGF antibody or fragment thereof.

10. The anti-PlGF antibody or fragment thereof according to claim 9 wherein said multiple doses are administered with at least 6-hour time intervals.

Description:
IMPROVING TRABECULECTOMY OUTCOME

FIELD OF THE INVENTION

The current invention relates to the improvement of trabeculectomy surgery. The improvement more specifically resides in an extended lifetime of the sclera-corneal drainage channel created by trabeculectomy surgery. The improvement is obtained by administration of an antibody, or antigen-binding fragment thereof, binding to placental growth factor (P1GF) and inhibiting P1GF activity. BACKGROUND OF THE INVENTION

Glaucoma is a multifactorial, neurodegenerative disease and the second most important cause of irreversible blindness (Quigley, 1996, Br J Ophthalmol 80, 389-393). This disease is characterized by progressive retinal ganglion cell apoptosis, resulting in visual field loss. Current treatment of this disease is directed towards the reduction of intraocular pressure (IOP), which is the main risk factor for glaucoma (Collaborative Normal-Tension Glaucoma Study Group, 1998, Am J Opthalmol 126, 487-497).

Of all currently used treatments to lower IOP, glaucoma filtration surgery (trabeculectomy), or shortly filtration surgery, was shown to be the most effective (Burr et al, 2005, Cochrane Database Syst Rev 18(2):CD004399; Hitchings, 1998, Arch Ophthalmol 116, 241-242). A trabeculectomy creates a "controlled" leak of fluid (aqueous humor) from the eye, which percolates under the conjunctiva. During the operation a piece of trabecular meshwork in the drainage angle of the eye is removed, creating an opening. The opening is partially covered with a flap of tissue from the sclera and conjunctiva. A small conjunctival "bleb" (bubble) appears at the junction of the cornea and the sclera (limbus) where this surgically produced valve is made.

In 30% of the cases, however, the constructed channel closes due to excessive scar tissue formation, resulting in surgical failure (Addicks et al, 1983, Arch Ophthalmol 101, 795-798). The 4 important processes contributing to post-operative conjunctival scarring are: clot formation, inflammation, angiogenesis and fibrosis (Lee et al, 1995, J Ocul Pharmacol Ther 11, 227-232; Lama & Fechtner, 2003, Surv Ophthalmol 48, 314-346). Indeed, increased conjunctival infiltration of inflammatory cells and Tenon fibroblasts (Hitchings & Grierson, 1983, Trans Ophthalmol Soc UK 103, 84-88; Skuta & Parrish, 1987, Surv Ophthalmol 32, 149-170), and higher levels of bleb vascularisation (Jampel et al, 1988, Arch Ophthalmol 106, 89-94) are associated with surgical failure. These processes are mediated by various cytokines (e.g. IL-1 and INF-a2b) and growth factors (e.g. PDGF, FGF, TGF-βΙ and VEGF (Lama & Fechtner, 2003; Gillies & Su, 1991, Aust NZ J Ophthalmol 19, 299-304)). Peroperative anti-mitotics, such as mitomycin-C and 5-Fluorouracyl can improve surgical outcome (Quigley, 1996; Katz et al, 1995, Ophthalmol 102, 1263-1269). However, these antimetabolites carry a risk of vision-threatening complications such as scleral thinning and infections (Lama & Fechtner, 2003; Hitchings & Grierson, 1983; Skuta & Parrish, 1987; Jampel et al, 1988; Gillies & Su, 1991; Katz et al, 1995; Greenfield et al, 1998, Arch Ophthalmol 116, 443-447). Furthermore, blocking TGF-β seemed promising in animal models (Cordeiro et al, 2003, Gene Ther 10, 59-71), but was not efficient in a clinical study (CAT-152 0102 Trabeculectomy Study Group, Kwah, Grehn, 2007, Ophthalmol 114, 1822- 1830). The number of post-trabeculectomy interventions expressed as the incidence of post- surgery "bleb manipulations" was reported to be as high as 78% (King et al, 2007, Br J Ophthalmol 91, 873-877). Therefore, there is still a need for alternative strategies to prevent filtration failure and, thus, to reduce the incidence of bleb manipulations.

Microplasmin is a recombinant protein that dissolves blood clots by degrading fibrin. Recently, microplasmin has been shown to be efficient, well tolerated and safe for intraocular use (WO 2004/052228) and was approved by FDA in October 2012 for treating vitreomacular adhesion (JETREA ®; non-proprietary name: ocriplasmin). Results of the phase III clinical trials leading to this approval were published by Stalmans et al. (2012, N Engl J Med 367, 606-615). Plasmin was previously shown to be able to induce PVD as well (e.g. US 5,304,118). The mechanism by which PVD is induced by plasmin or microplasmin is currently not fully understood. Unsupported by any or any conclusive experimental data, WO 2009/073457 and WO 2009/067407 propose subconjunctival plasmin injection for rescuing filtering blebs and the use of matrix metalloproteinase activating proteases for reducing IOP, respectively. WO 2011/023805 provides the evidence that anterior chamber injection of microplasmin was effective in prolonging bleb survival, i.e., the mode of administration of microplasmin in this indication is determining success. Pegaptanib is a pegylated anti-VEGF aptamer (VEGF = vascular endothelial growth factor), a single strand of nucleic acid (50 kDa). It specifically binds the VEGFi 6 5 isoform, thereby preventing the binding to the heparin binding domain. Van Bergen et al. 2011 (Exp Eye Res 93, 689-699) showed that single or repeated injection of pegaptanib after glaucoma filtration surgery (in a rabbit model) had marginal effect on bleb area and bleb survival. Bevacizumab is an antibody inhibiting all forms of VEGF-A. Li et al. 2009 (Invest Ophthalmol Vis Sci 50, 5217-5225) disclosed the effect of bevacizumab on glaucoma filtration surgery (in a rabbit model) which was, judging from the reported effect on bleb area, limited.

SUMMARY OF THE INVENTION

The invention relates to an anti-PlGF (placental growth factor) antibody or fragment thereof for improving or enhancing the success rate of trabeculectomy surgery (or glaucoma filtration surgery) of an eye, or for preventing, reducing or retarding the occurrence of filtration failure after trabeculectomy surgery of an eye.

Alternatively, the invention relates to the use of an anti-PlGF antibody or fragment thereof for the manufacture of a medicament for, or in a method of, improving or enhancing the success rate of trabeculectomy surgery (or glaucoma filtration surgery) of an eye, or for/of preventing, reducing or retarding the occurrence of filtration failure after trabeculectomy surgery of an eye.

The anti-PlGF antibody or fragment thereof as described above may be in a pharmaceutically acceptable formulation capable of being administered to an eye. In particular, said pharmaceutically acceptable formulation is compatible with administration into the anterior chamber of an eye or with in-bleb administration.

Said treating of filtration failure after trabeculectomy surgery of an eye, or said preventing, reducing or retarding of the occurrence of filtration failure after trabeculectomy surgery of an eye with an anti-PlGF antibody or fragment thereof as described above in particular results from administering to said eye at least a single dose, or, alternatively, multiple doses, of an effective amount of said anti-PlGF antibody or fragment thereof. When multiple doses are administered to an eye, these may be administered with at least 6-hour time intervals. Said eye may be contacted further with one or more agents chosen from an agent for controlling the intra-ocular pressure, an anti-inflammatory agent, an antiviral agent, an antibacterial agent, an antiviral agent, an anti-angiogenic agent, an anti-mitotic agent, an antihistamine, an anesthetic, an agent to induce mydriasis and an agent to induce cycloplegia. Said further contacting may be occurring prior to, concurrent with, or after administering the anti-PlGF antibody or fragment thereof.

The anti-PlGF antibody or fragment thereof as described above may be in a pharmaceutically acceptable formulation further comprising one or more of an agent for controlling the intra- ocular pressure, an anti-inflammatory agent, an antiviral agent, an antibacterial agent, an antiviral agent, an anti-angiogenic agent, an anti-mitotic agent, an antihistamine, an anesthetic, an agent to induce mydriasis and an agent to induce cycloplegia.

FIGURE LEGENDS

FIGURE 1. Fig. 1 A shows result of measurement of the intra-ocular pressure (IOP) in two groups (group size n=10) that had undergone glaucoma filtration surgery (GFS). One group was treated with 1C8, an irrelevant mouse IgG antibody (4.8 mg/ml) and the other group was treated with DC 101, an anti-murine VEGF-R2 antibody (6.2 mg/ml). IOPs were measured with a Tonolab (Technop®). Results are represented as mean ±SEM. IOP was not found to be significantly different in the 2 groups (p>0.05). Fig. IB shows in a similar way the result of measurement of in two groups (group size n=10) that had undergone GFS. One group was treated with 1C8, an irrelevant mouse IgG antibody (4.8 mg/ml) and the other group was treated with 5D11D4, an anti-murine P1GF antibody (5.2 mg/ml). IOP was not found to be significantly different in the 2 groups (p>0.05).

FIGURE 2. Fig. 2A shows measurements of bleb area in two groups (group size n=10) that had undergone (GFS). One group was treated with 1C8, an irrelevant mouse IgG antibody (4.8 mg/ml) and the other group was treated with DC 101 , an anti-murine VEGF-R2 antibody (6.2 mg/ml). Results are represented as mean ±SEM. DC101 significantly improved bleb area as compared to 1C8 (p=0.05). Fig. 2B is similar to Fig. 2A except that both groups were larger in size (n=20) and were followed during a longer time period. Fig. 2C shows similar results as Fig. 2A and 2B except that the antibodies were injected repeatedly at days 0, 4 and 10 after surgery.

FIGURE 3. Fig. 3 A shows bleb survival in two groups (group size n=10) that had undergone GFS. One group was treated with 1C8, an irrelevant mouse IgG antibody (4.8 mg/ml) and the other group was treated with DC 101, an anti-murine VEGF-R2 antibody (6.2 mg/ml). Bleb survival was not found to be significantly different in the 2 groups (p=0.23). After longer follow-up of two larger groups (n=20; Fig. 3B), significantly increased bleb survival was observed in the DClOl-treated group vs the lC8-treated group (p=0.06). Repeated injection of DClOl (group size n=10) at days 0, 4 and 10 after surgery further increased the bleb survival compared to single injections (Fig. 3C).

FIGURE 4. Fig. 4 A shows measurements of bleb area in two groups (group size n=10) that had undergone GFS. One group was treated with 1C8, an irrelevant mouse IgG antibody (4.8 mg/ml) and the other group was treated with 5D1 1D4, an anti-murine P1GF antibody (5.2 mg/ml). Results are represented as mean ±SEM. 5D11D4 significantly improved bleb area as compared to 1C8 (p=0.01). This effect extended till the end of a longer follow-up period of larger groups (n=20; Fig. 4B) and was markedly enhanced by repeat injections of 5D11D4 on days 0, 4 and 10 after surgery (group size n=10; Fig. 4C). The latter effect was significantly more pronounced compared to repeat injections of anti-murine VEGF-R2 antibody DClOl (Fig. 2C / Fig. 6 C)

FIGURE 5. Fig. 5 A shows bleb survival in two groups (group size n=10) that had undergone GFS. One group was treated with 1C8, an irrelevant mouse IgG antibody (4.8 mg/ml) and the other group was treated with 5D11D4, an anti-murine P1GF antibody (5.2 mg/ml). Results are represented as mean ±SEM. 5D11D4 significantly improved bleb survival as compared to 1C8 (p=0.04). This effect extended till the end of a longer follow-up period of larger groups (n=20; Fig. 5B) which was significantly more pronounced compared to the anti-murine VEGF-R2 antibody DClOl (Fig. 3B), and was markedly enhanced by repeat injections of 5D11D4 on days 0, 4 and 10 after surgery (group size n=10; Fig. 5C).

FIGURE 6. Fig. 6 A shows measurements of bleb area in two groups (group size n=10) that had undergone GFS. One group was treated with DClOl, an anti-murine VEGF-R2 antibody (6.2 mg/ml) and the other group was treated with 5D1 1D4, an anti-murine P1GF antibody (5.2 mg/ml). Results are represented as mean ±SEM. 5D11D4 significantly improved bleb area as compared to 1C8 (Fig. 1). A trend towards an increased bleb area after 5D11D4 administration was observed compared to DClOl delivery (p=0.07). The latter was confirmed and strengthened when observing two groups (group size n=20) for a longer time period (Fig. 6B). The stronger effect of 5D11D4 administration over DClOl administration was moreover clearly obviated when comparing the effect of multiple administrations (at days 0, 4 and 10 after surgery; group size n=10) of the antibodies as depicted in Fig. 6C. DETAILED DESCRIPTION OF THE INVENTION

As known from clinical practice, each subject or patient undergoing trabeculectomy surgery is at significant risk to develop filtration failure. The present invention is based on the effect of administration of an anti-PlGF antibody (antibody inhibiting the activity of placental growth factor, P1GF) on the clinical outcome of trabeculectomy surgery, said effect being positive and resulting in the prevention, reduction or retardation of the occurrence of filtration failure. The effects obtained with an anti-PlGF antibody are moreover markedly and unexpectedly more pronounced than the effects obtained with an inhibitor of VEGF-R2 (vascular endothelial growth factor receptor 2, known to bind VEGF) or obtained with inhibitors of VEGFi 6 5 (pegaptanib; Van Bergen et al. 201 1, Exp Eye Res 93, 689-699) or of VEGF (bevacizumab; Li et al. 2009, Invest Ophthalmol Vis Sci 50, 5217-5225).

Therefore, the invention relates to an anti-PlGF antibody or fragment thereof for treating filtration failure after trabeculectomy surgery of an eye, or for improving the success rate of trabeculectomy surgery, or for increasing or enhancing the success rate of trabeculectomy surgery, or for preventing, reducing or retarding the occurrence of filtration failure after trabeculectomy surgery of an eye; all evidently as compared to trabeculectomy surgery performed without using an anti-PlGF antibody or fragment thereof. Alternatively, the invention relates to the use of anti-PlGF antibody or fragment thereof for the manufacture of a medicament for, or in a method of, treating filtration failure after trabeculectomy surgery of an eye, or for/of improving the success rate of trabeculectomy surgery, or for/of increasing or enhancing the success rate of trabeculectomy surgery, or for/of preventing, reducing or retarding the occurrence of filtration failure after trabeculectomy surgery of an eye; all evidently as compared to trabeculectomy surgery performed without using an anti-PlGF antibody or fragment thereof. The terms "glaucoma filtration surgery", "filtration surgery" and "trabeculectomy surgery" are used herein interchangeably.

The "trabecular meshwork (TM)" is a mesh-like structure inside the eye at the iris-scleral junction of the anterior chamber angle. The TM filters the aqueous fluid and controls its flow into the canal of Schlemm prior to its leaving the anterior chamber. Increased resistance in the TM leads to reduced aqueous fluid outflow and thus increased intra-ocular pressure (IOP). When left untreated, this elevated IOP leads to glaucomatous damage to the optic nerve and retinal nerve fibers, and leads to loss of vision. This vision loss can be prevented or halted by administering medication, an "agent for controlling the intra-ocular pressure", which controls the intra-ocular pressure. Such medicaments include adrenergic blocking agents (beta blockers or sympatholytic drugs such as betaxolol, carteolol, levobunolol, metipanolol and timolol), adrenergic stimulating agents (sympathomimetic drugs such as aproclonidine, epinephrine, hydroxyamphetamine, phenylephrine, naphazoline and tetrahydrozaline), carbonic anhydrase inhibitors (such as systemic acetozolamide, and topical brinzolamide and dorzolamide), miotics (cholinergic stimulating agents, parasympathomimetic drugs such as carbachol and pilocarpine), osmotic agents (such as glycerin and mannitol), prostaglandin and prostaglandin analogues (prostamides, bimatoprost, unoprostone isopropyl, travoprost, latanoprost, natural prostaglandin, prostaglandin F2a, and FP prostanoid receptor agonists). When such medicaments are not efficient (or not anymore), then filtration surgery is a viable treatment.

"Trabeculectomy", "trabeculectomy surgery" or "filtration surgery", or "glaucoma filtration surgery", is defined as a surgical procedure on the eye wherein part of the trabecular meshwork is removed whereby a filtration site (a sclera-corneal drainage channel) is created that increases the outflow of aqueous fluid from the eye; this type of filtering procedure is commonly used in the treatment of glaucoma, and more specifically to reduce the IOP in an eye subject to/suffering from glaucoma.

"Filtration failure" is a condition reversing the clinically desired effect of trabeculectomy surgery, i.e., reversing the desired drop in IOP. The initial post-operative time is crucial in the sense that eye-healing activities are highest in this period. This period of high eye-healing capacity is dependent upon the species and spans about 2 weeks for rabbits and up to 1- to 2- months in humans. Upon contacting an anti-PlGF antibody or fragment thereof with an eye according to the current invention, the frequency of occurrence of filtration failure over a given period of time is lowered. The anti-PlGF antibody or fragment thereof used according to the current invention thus results in the prevention, reduction or retarding of the occurrence of filtration failure, or in an improvement, enhancement or increase of the success rate of trabeculectomy surgery (compared to trabeculectomy surgery without administering or using an anti-PlGF antibody or fragment thereof). The anti-PlGF antibody or fragment thereof, or a medicament comprising it, may be in a pharmaceutically acceptable formulation (or composition or solution) capable of being administered to an eye. In particular, said formulation (or composition or solution) is capable of being administered into the anterior chamber of the eye or compatible with administration into the anterior chamber of the eye. Alternatively, said formulation (or composition or solution) is capable of being administered into the surgically created bleb (i.e., in-bleb administration) or compatible with administration into bleb. Said administration may e.g. be by injection of the formulation (or composition or solution) or medicament comprising an anti-PlGF antibody or fragment thereof. Although not required, there may be an additional advantage in said formulation being a slow-release formulation such as a gel-like formulation.

The improvement, enhancement or increase of the success rate of trabeculectomy surgery (or glaucoma filtration surgery) of an eye, or of the prevention, reduction or retardation of the occurrence of filtration failure after trabeculectomy surgery of an eye may result from introduction into the eye of an effective amount of at least a single dose of an anti-PlGF antibody (or fragment thereof) or of a formulation (or composition or solution) or medicament comprising it. In particular, said administration is into the anterior chamber of an eye, such as by injection. Alternatively, multiple doses of an effective amount of said anti- P1GF antibody or fragment thereof (or of a formulation (or composition or solution) or medicament comprising it) may be administered, such as to increase efficacy. When multiple doses are administered to an eye, these may be administered with at least 6-hour time intervals, with about 12-hour time intervals, with about 18-hr time intervals, with about 1-day time intervals, with about 2-day time intervals, with about 3-day time intervals, with about 4- day time intervals, with about 7-day time intervals, with about 2-week time intervals with about 1 -month time intervals, with about 2-month time intervals or with about 3-month time intervals. When multiple doses are administered to an eye with time intervals, the time interval between two subsequent doses may change during the treatment depending on the evolution of the clinical result. For example, time intervals between subsequent doses may be short immediately after the trabeculectomy surgery and may increase with increasing time after the trabeculectomy surgery. In any of the above, said anti-PlGF antibody may be any type of antibody or any fragment of any thereof that is capable of binding to PIGF and of inhibiting the activity of PIGF. In particular, said anti-PlGF antibody or fragment thereof may be neutralizing the activity of PIGF, thus may be a neutralizing anti-PlGF antibody or neutralizing anti-PlGF antibody fragment. Such antibodies include all types of antibodies known in the art, such as human or humanized antibodies, cameloid antibodies, nanobodies, domain antibodies, mono- or plural- specific antibodies, etc., and any fragment of any thereof. Examples of anti-PlGF antibodies are described in WO 01/85796 and WO 2006/099698. In particular, an anti-PlGF antibody for use as described herein is effective in inhibiting the activity of placental growth factor as present in the subject undergoing trabeculectomy. In particular, said subject is a mammal, more in particular a human.

The invention further covers an anti-PlGF antibody (or any fragment thereof) as described above for improving, enhancing or increasing the success rate of trabeculectomy surgery, or for treating filtration failure after trabeculectomy surgery of an eye, or for preventing, reducing or retarding the occurrence of filtration failure after trabeculectomy surgery of an eye, wherein the anti-PlGF antibody or fragment thereof is in a pharmaceutically acceptable formulation (or composition or solution) that may further comprise one or more of an agent for controlling the intra-ocular pressure, an anti-inflammatory agent, an antiviral agent, an antibacterial agent, an antiviral agent, an anti-angiogenic agent, an anti-mitotic agent, an antihistamine, an anesthetic, an agent to induce mydriasis and an agent to induce cycloplegia. Alternatively, when said further agent is, or said further agents are, not included in the pharmaceutically acceptable formulation (or composition or solution) containing said anti- P1GF antibody (or any fragment thereof), said eye may be contacted further with one or more agents chosen from an agent for controlling the intra-ocular pressure, an anti-inflammatory agent, an antiviral agent, an antibacterial agent, an antiviral agent, an anti-angiogenic agent, an anti-mitotic agent, an antihistamine, an anesthetic, an agent to induce mydriasis and an agent to induce cycloplegia. Such further contacting may be prior to, concurrent with, or after the administration of an anti-PlGF antibody or any fragment thereof (or of a formulation, composition, solution, or medicament comprising it).

The invention further covers the use of an anti-PlGF antibody (or any fragment thereof) as described above for the manufacture of a medicament for improving, enhancing or increasing the success rate of trabeculectomy surgery, or for treating filtration failure after trabeculectomy surgery of an eye, or for preventing, reducing or retarding the occurrence of filtration failure after trabeculectomy surgery of an eye, wherein the anti-PlGF antibody or fragment thereof is in a pharmaceutically acceptable composition that may further comprise one or more of an agent for controlling the intra-ocular pressure, an anti-inflammatory agent, an antiviral agent, an antibacterial agent, an antiviral agent, an anti-angiogenic agent, an antimitotic agent, an antihistamine, an anesthetic, an agent to induce mydriasis and an agent to induce cycloplegia. Alternatively, when said further agent is, or said further agents are, not included in the pharmaceutically acceptable formulation (or composition or solution) or medicament containing said an anti-PlGF antibody (or any fragment thereof), said eye may be contacted further with one or more agents chosen from an agent for controlling the intraocular pressure, an anti-inflammatory agent, an antiviral agent, an antibacterial agent, an antiviral agent, an anti-angiogenic agent, an anti-mitotic agent, an antihistamine, an anesthetic, an agent to induce mydriasis and an agent to induce cycloplegia. Such further contacting may be prior to, concurrent with, or after the administration of an anti-PlGF antibody or any fragment thereof (or of a formulation, composition, solution, or medicament comprising it).

Methods of improving, increasing or enhancing the success rate of trabeculectomy surgery of an eye, of treatment of filtration failure after trabeculectomy surgery of an eye, and methods of preventing, reducing or retarding the occurrence of filtration failure after trabeculectomy surgery of an eye are also envisaged. These methods comprise the step of contacting said eye after trabeculectomy surgery with an effective amount of an anti-PlGF antibody (or a fragment thereof) wherein said contacting results in said improvement, increase or enhancement of the success rate of trabeculectomy surgery treatment of an eye, in said treatment of filtration failure after trabeculectomy surgery of an eye, or in said preventing, reducing or retarding of the occurrence of filtration failure after trabeculectomy surgery of an eye. In such methods, the eye may further be contacted with an agent for controlling the intraocular pressure, an anti-inflammatory agent, an antiviral agent, an antibacterial agent, an antiviral agent, an anti-angiogenic agent, an anti-mitotic agent, an antihistamine, an anesthetic, an agent to induce mydriasis and an agent to induce cycloplegia. Such further agent different from an anti-PlGF antibody (or fragment thereof) may be co-administered with the anti-PlGF antibody in the same formulation (or composition or solution) or medicament, or concurrently in a separate formulation (or composition or solution) or medicament. Such further agent different from an anti-PlGF antibody (or fragment thereof) may alternatively be administered prior to or after administration of the anti-PlGF antibody (or fragment thereof).

"Contacting" means any mode of administration that results in interaction between an agent or composition such as a medicament and an object (such as conjunctiva or subconjunctival tissue) with which said agent or composition is contacted. The interaction between the agent or composition and the object can occur starting immediately or nearly immediately with the administration of the agent or composition, can occur over an extended time period (starting immediately or nearly immediately with the administration of the agent or composition), or can be delayed relative to the time of administration of the agent or composition. More specifically the "contacting" results in delivering an effective amount of the agent, composition or medicament to the object.

The term "effective amount" refers to the dosing regimen of the agent, composition or medicament according to the invention, in particular of the active ingredient of the medicament according to the invention, i.e., an anti-PlGF antibody or a fragment thereof. The effective amount will generally depend on and will need adjustment to the mode of contacting or administration. The effective amount of the agent, composition or medicament, more particular its active ingredient, is the amount required to obtain the desired clinical outcome or therapeutic or prophylactic effect without causing significant or unnecessary toxic effects. To obtain or maintain the effective amount, the agent, composition or medicament may be administered as a single dose or in multiple doses. The effective amount may further vary depending on the severity of the condition that needs to be treated or the expected severity of the condition that needs to be prevented or treated; this may depend on the overall health and physical condition of the patient and usually the treating doctor's or physician's assessment will be required to establish what is the effective amount. The effective amount may further be obtained by a combination of different types of contacting or administration. In the context of the present invention the effective amount may more particularly be obtained by either one or more of administration of topical eye drops, administration by injection into the anterior chamber of an eye or administration by subconjunctival injection. A typical dose of a single administration of the agent, composition or medicament of the invention may comprise 10 μ to 20 mg of the active compound, or alternatively may comprise 10 μ§/1¾ body weight to 20 mg/kg body weight of the active compound. Administration of the medicament of the invention by means of injection typically is kept to a minimum, i.e., the frequency of repeat injections is kept to a minimum. As the first weeks or months post-trabeculectomy (species dependent as described higher) are crucial in the sense that eye-healing activities are highest in this period, the duration of treatment with an agent, composition or medicament according to the present invention should be adjusted to this period. In general, the formulation (or composition or solution) or medicament of the invention comprising an anti-PlGF antibody or fragment thereof according to the invention may, depending on its ultimate use and mode of administration, comprise one or more further active ingredients such as an agent controlling the intra-ocular pressure (see higher), an anticoagulant, a thrombolytic agent, an anti-inflammatory agent, an antiviral agent, an antibacterial agent, an antifungal agent, an anti-angiogenic agent, an anti-mitotic agent, an antihistamine or anesthetic.

"Anticoagulants" include hirudins, heparins, coumarins, low-molecular weight heparin, thrombin inhibitors, platelet inhibitors, platelet aggregation inhibitors, coagulation factor inhibitors, anti-fibrin antibodies and factor Vlll-inhibitors (such as those described in WO 01/04269 and WO 2005/016455).

"Thrombolytic agents" include urokinase, streptokinase, tissue-type plasminogen activator (tPA), urokinase-type plasminogen activator (uPA) and staphylokinase or any variant or derivative of any thereof such as APSAC (anisoylated plasminogen streptokinase activator complex), alteplase, reteplase, tenecteplase, and scuPA (single chain uPA), plasmin or any truncated variant thereof such as midiplasmin, miniplasmin, deltaplasmin and microplasmin.

"Anti-inflammatory agents" include steroids (e.g. prednisolone, methylprednisolone, cortisone, hydrocortisone, prednisone, triamcinolone, dexamethasone) and non-steroidal antiinflammatory agents (NSAIDs; e.g. acetaminophren, ibuprofen, aspirin). "Antiviral agents" include trifluridine, vidarabine, acyclovir, valacyclovir, famciclovir, and doxuridine.

"Antibacterial agents" or antibiotics include ampicillin, penicillin, tetracycline, oxytetracycline, framycetin, gatifloxacin, gentamicin, tobramycin, bacitracin, neomycin and polymyxin.

"Anti-mycotic/fungistatic/antifungal agents" include fluconazole, amphotericin, clotrimazole, econazole, itraconazole, miconazole, 5-fluorocytosine, ketoconazole and natamycin.

"Anti-angiogenic agents" include antibodies (or fragments thereof) such as anti-VEGF (vascular endothelial growth factor) or anti-PlGF (placental growth factor) antibodies and agents such as macugen (pegaptanib sodium), trypthophanyl-tRNA synthetase (TrpRS), anecortave acetate, combrestatin A4 prodrug, AdPEDF (adeno vector capable of expressing pigment epithelium-derived factor), VEGF-trap, inhibitor of VEGF receptor-2, inhibitors of VEGF, P1GF or TGF-β, Sirolimus (rapamycin) and endostatin.

"Anti-mitotic agents" include mitomycin C and 5-fluorouracyl. "Antihistamine" includes ketitofen fumarate and pheniramine maleate.

"Anesthetics" include benzocaine, butamben, dibucaine, lidocaine, oxybuprocaine, pramoxine, proparacaine, proxymetacaine, tetracaine and amethocaine. Other adjunct agents or drugs that can be used in conjunction with the anti-PlGF antibody or fragment thereof according to the invention include scopoloamine, atropine or tropicamide, to induce mydriasis (pupillary dilation) and/or cycloplegia (paralysis of the eye focusing muscle). In addition to the anti-PlGF antibody or fragment thereof, each of the above listed agents as well as antihistamine and anesthetics is to be considered as an "active ingredient". A "pharmaceutically acceptable formulation" is, in the context of the current invention more particular an "ophthalmologically acceptable formulation". A formulation in general is a composition comprising a carrier, diluent or adjunvant compatible with the one or more active ingredients to be formulated, the whole formulation being compatible with the intended use in the intended tissue or organ, etc. Examples of pharmaceutically acceptable formulations as well as methods for making them can be found, e.g., in Remington's Pharmaceutical Sciences (e.g. 20 th Edition; Lippincott, Williams & Wilkins, 2000) or in any Pharmacopeia handbook (e.g. US-, European- or International Pharmacopeia).

"Lubricants" include propylene glycerol, glycerin, carboxymethylcellulose, hydroxypropylmethylcellulose, soy lecithin, polyvinyl alcohol, white petrolatum, mineral oil, povidone, carbopol 980, polysorbate 80, dextran 70.

EXAMPLES

The Examples included hereafter demonstrate the invention and are not construed to be limiting the scope of the invention in any way.

PURPOSE. Excessive postoperative wound healing with subsequent inflammation and scarring frequently leads to surgical failure of glaucoma filtration surgery (GFS). The hypothesis was checked that placental growth factor (PIGF) plays a role in scar formation after GFS, and that it may be a target for improvement of the outcome of this surgery.

METHODS. Aqueous humor and plasma samples of glaucoma and control patients (n=10) were collected and PIGF levels were determined by ELISA. The effect of the anti-murine PIGF-antibody (5D11D4) was investigated in a mouse model of GFS in C75B1/6 mice. In the single-injection setting, 5D11D4 (Ι μΐ; 5.2mg/ml; antibody described in detail in WO 01/85796) or 1C8, an irrelevant mouse IgG antibody against human tissue plasminogen activator (Ιμΐ; 4.8 mg/ml; antibody available at ThromboGenics), were injected in the anterior chamber (n=10 eyes or n=20 eyes for both groups) immediately after surgery ("day 0"). An anti-murine VEGF-R2 antibody (DC101) was used as a positive control (Ιμΐ; 6.2 mg/ml; n=10). Mice were killed on post-operative day 8. In the multiple- or repeated- injection setting, the antibodies were administered as above, albeit it on days 0, 4 and 10 after surgery; groups of n=10 eyes were assessed; and mice were killed on post-operative day 13. Treatment outcome was studied by clinical investigation of intra-ocular pressure (IOP), bleb area and bleb survival every other day. RESULTS. PIGF levels in aqueous humor were found to be significantly upregulated in glaucoma compared to control patients (17 ± 2 pg/ml versus 12 ± 0.75 pg/ml, p=0.03). No significant differences were found in plasma concentrations of PIGF. In the mouse model of GFS, single administration of the anti-PlGF antibody (5D11D4) significantly improved surgical outcome by increasing bleb survival (p=0.04) and bleb area (p=0.01) with 29% compared to negative control (1C8). A single administration of anti-VEGF-R2 (DClOl) also significantly improved bleb area with 7% as compared to 1C8 (p=0.05), but had no effect on bleb survival (p=0.23). A trend towards an increased bleb area after 5D11D4 administration was observed compared to DClOl delivery (p=0.07). IOP was not found to be significantly different in any of the groups (p>0.05). Results of single administration of the antibodies are depicted in Figure 1 (IOP); Figures 2A, 2B, 4A, 4B, 6A and 6B (all bleb area); and Figures 3A, 3B, 5A and 5B (bleb survival).

Multiple- or repeated administrations of the antibodies (at days 0, 4 and 10 after surgery) led to a more pronounced improvement of surgical outcome with both 5D11D4 and DClOl separately compared to 1C8, and with a more pronounced positive outcome with 5D1 1D4 compared to DClOl . Furthermore, the trend towards an increased bleb area after 5D11D4 administration compared to DClOl upon single administration (p=0.07) was converted into a significant difference. The latter further proves that an anti-PlGF antibody (5D11D4) is more efficient in improving surgical outcome of GFS than an anti-VEGF-R2 antibody (DClOl) (p=0.005). Results of repeated administrations of the antibodies are depicted in Figures 2C, 4C and 6C (all bleb area); and Figures 3C and 5C (bleb survival).

CONCLUSIONS. Local production of PIGF in the eye may indicate an important role for this growth factor in wound healing after GFS. Indeed, targeting PIGF with an inhibitory monoclonal antibody is efficacious in improving GFS outcome, even more efficacious than inhibition of VEGF-R2 as described herein, and more efficacious than inhibition of VEGF- 165 (Van Bergen et al. 2011, Exp Eye Res 93, 689-699) or VEGF-A (Li et al. 2009, Invest Ophthalmol Vis Sci 50, 5217-5225). This effect is seen with single administration of an anti- PIGF antibody and is significantly enhanced upon multiple administrations of an anti-PlGF antibody. These results render PIGF an validated target for ocular wound healing and point to the therapeutic benefits of PIGF-inhibition in this setting.