15.02.2017 Views

13th AOPT Scientific Meeting - Program

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>AOPT</strong><br />

TABLE OF CONTENTS<br />

Association for Ocular<br />

Pharmacology and Therapeutics<br />

13 th <strong>Scientific</strong> <strong>Meeting</strong><br />

February 16-19, 2017<br />

Florence (Italy)<br />

OCULAR THERAPEUTICS:<br />

VISION OF HOPE IN A<br />

CHANGING WORLD<br />

www.aopt.org


INDEX<br />

THANKS TO OUR SPONSOR........................................................ pag. 4<br />

WELCOME MESSAGE.................................................................. pag. 7<br />

<strong>AOPT</strong> MEMBERSHIP INFORMATION.............................................. pag. 8<br />

<strong>AOPT</strong> PAST MEETINGS................................................................ pag. 9<br />

YOUR <strong>AOPT</strong> BOARD.................................................................... pag. 10<br />

YOUR LOCAL ORGANIZING COMMITTEE....................................... pag. 12<br />

TRAVEL AWARD WINNERS........................................................... pag. 13<br />

GENERAL INFORMATION............................................................. pag. 14<br />

INFORMATION FOR PRESENTERS................................................. pag. 15<br />

PALAZZ0 DEGLI AFFARI PLAN....................................................... pag. 16<br />

PROGRAM AT-A-GLANCE............................................................. pag. 17<br />

KEYNOTE SPEAKER.................................................................... pag. 18<br />

SCIENTIFIC PROGRAM............................................................... pag. 19<br />

IT-ARVO PROGRAM.................................................................... pag. 27<br />

PLATFORM ABSTRACT................................................................ pag. 29<br />

POSTER SESSION....................................................................... pag. 89


PANTONE<br />

3015 C<br />

THANKS TO OUR SPONSORS<br />

TITLE LEVEL<br />

PLATINUM LEVEL<br />

GOLD LEVEL<br />

SILVER LEVEL


THANKS TO OUR SPONSORS<br />

BRONZE LEVEL<br />

OTHER<br />

YOUNG INVESTIGATOR TRAVEL AWARDS<br />

UNTHSC/Elena and Tom Yorio


WELCOME MESSAGE<br />

Welcome to the 13 th <strong>Scientific</strong> <strong>Meeting</strong> of the<br />

Association for Ocular Pharmacology and<br />

Therapeutics (<strong>AOPT</strong>). The topic of this year’s<br />

conference is Ocular Therapeutics: Vision of<br />

hope in a changing world.<br />

We are thankful for our Italian hosts who have<br />

put together an exciting meeting at a great<br />

venue at the Firenze Fiera Congress Center<br />

here in Florence Italy. The meeting agenda is packed with exciting<br />

new information set in thirteen sessions as well as a special treat with<br />

a visit to the Uffizi Museum.<br />

The Association for Ocular Pharmacology and Therapeutics is a<br />

global not-for-profit organization for scientists and individuals from<br />

all disciplines related to ocular pharmacology and its therapeutic<br />

applications. <strong>AOPT</strong>- has a diverse, multi-national membership<br />

composed of preclinical and clinical scientists, students, and healthcare<br />

professionals. Members are from academic institutions, pharma and<br />

biotech industries, device companies, clinics and private practice.<br />

<strong>AOPT</strong>’s mission is to serve as a global forum and network for<br />

the publication, dissemination and exchange of information and<br />

knowledge on treatments of eye diseases, from basic and clinical<br />

ocular pharmacology and therapeutics to related disciplines such as<br />

pharmacokinetics and dynamics, metabolism, translational research,<br />

safety, drug delivery, and pharmaceutics.<br />

This conference brings together those individuals at the forefront<br />

of scientific advancement related to ocular pharmacology and<br />

therapeutics. The agenda is rich in provocative presentations that we<br />

hope stimulate productive discussions.<br />

What better place to set a new vision for ocular therapeutics than in<br />

Florence the “Cradle of the Renaissance”.<br />

Tom Yorio, PhD, FARVO<br />

President, <strong>AOPT</strong><br />

7


<strong>AOPT</strong> MEMBERSHIP INFORMATION<br />

There are four classes of membership in <strong>AOPT</strong>: Regular Members,<br />

Associate Members, Contributing Members, and Emeritus Members.<br />

REGULAR MEMBERS<br />

The Regular Membership represent individuals demonstrating<br />

a genuine interest in or making significant contribution to<br />

ocular pharmacology and therapeutics. This may be evidenced<br />

by a) scientific publications; b) attendance at pharmacological,<br />

ophthalmological, optometric, or visual science meetings; c) direct<br />

involvement in research. A candidate for membership completes the<br />

online membership form and pays the appropriate membership dues.<br />

Membership is for two years. A subscription to the Journal of Ocular<br />

Pharmacology and Therapeutics is optional.<br />

ASSOCIATE MEMBERS<br />

Associate Membership is for predoctoral and postdoctoral students.<br />

A candidate for this membership must have a pre-doctoral, or postdoctoral<br />

student status, and must complete the online membership<br />

form and pay the appropriate membership dues.<br />

CONTRIBUTING MEMBERS<br />

Contributing Membership is restricted to corporations, associations,<br />

and individuals who support the objectives of <strong>AOPT</strong> but do not satisfy<br />

the requirements of Regular Membership or individuals elected<br />

to membership in any class who voluntarily choose to become<br />

Contributing Members. A candidate for contributing membership<br />

completes the online membership form and pays the appropriate<br />

membership dues.<br />

EMERITUS MEMBERS<br />

Any Regular Member may make a written request to the Treasurer<br />

that his/her membership be transferred to that of an Emeritus<br />

Member. The request is subject to approval of the membership<br />

committee. Emeritus Members have all the rights and privileges of<br />

Regular Members, except those of voting and holding elective office.<br />

8


<strong>AOPT</strong> PAST MEETINGS<br />

MEETING DATE LOCATION ORGANIZER<br />

TWELFTH MEETING<br />

ELEVENTH MEETING<br />

FEB 26 - MAR 1, 2015<br />

FEBRUARY 7-10, 2013<br />

CHARLESTON, SC<br />

ALICANTE, SPAIN<br />

DAN STAMER<br />

JUANA GALLAR MARTINEZ<br />

TENTH MEETING<br />

FEBRUARY 17-20, 2011<br />

FT. WORTH, TX<br />

TOM YORIO / ABBOT CLARK<br />

NINTH MEETING<br />

FEBRUARY 18-21, 2009<br />

SALZBURG, AUSTRIA<br />

HERBERT REITSAMER<br />

EIGHTH MEETING<br />

FEBRUARY 9-11, 2007<br />

SAN DIEGO, CA<br />

JOHN LIU / ACHIM KRAUSS<br />

SEVENTH MEETING<br />

FEBRUARY 3-5, 2005<br />

CATANIA, SICILY, ITALY<br />

FILIPPO DRAGO<br />

SIXTH MEETING<br />

FEBRUARY 1-4, 2003<br />

KONA, HI<br />

PETER KADOR<br />

FIFTH MEETING<br />

NOVEMBER 2-5, 2000<br />

BIRMINGHAM, AL<br />

JIMMY BARTLETT<br />

FOURTH MEETING<br />

JANUARY 28-31, 1999<br />

IRVINE, CA<br />

ACHIM KRAUSS<br />

THIRD MEETING<br />

OCTOBER 22-24, 1997<br />

BETHESDA, MD<br />

PETER KADOR<br />

SECOND MEETING<br />

AUGUST 15-17, 1996<br />

LOS ANGELES, CA<br />

DAVID LEE<br />

FIRST MEETING<br />

OCULAR<br />

PHARMACOLOGY<br />

SYMPOSIUM<br />

JANUARY 26-29, 1995<br />

AUGUST 8-10, 1993<br />

NEW ORLEANS, LA<br />

NOVI, MI<br />

HERB KAUFMAN<br />

HITOSHI SHICHI<br />

9


YOUR <strong>AOPT</strong> BOARD<br />

PRESIDENT<br />

DR. THOMAS YORIO<br />

VICE-PRESIDENT<br />

PRESIDENT-ELECT<br />

DR. FILIPPO DRAGO<br />

IMMEDIATE-PAST-PRESIDENT<br />

DR. ACHIM KRAUSS<br />

TRUSTEE<br />

DR. MALINDA FITZGERALD<br />

TRUSTEE<br />

DR. ASH JAYAGOPAL<br />

TRUSTEE<br />

DR. UDAY KOMPELLA<br />

TRUSTEE<br />

DR. GANESH PRASANNA<br />

TRUSTEE<br />

DR. SHUSHENG WANG<br />

10


YOUR <strong>AOPT</strong> BOARD<br />

TRUSTEE<br />

DR. CHRISTINE WILDSOET<br />

TREASURER<br />

DR. PETER KADOR<br />

SECRETARY<br />

DR. CAROL TORIS<br />

TRUSTEE<br />

DR. CAMERON MILLAR<br />

TRUSTEE<br />

DR. IOK-HOU PANG<br />

11


YOUR LOCAL ORGANIZING COMMITTEE YOUR <strong>AOPT</strong> BOARD<br />

FILIPPO DRAGO<br />

CLAUDIO BUCOLO<br />

CATERINA GAGLIANO<br />

ALESSANDRO MUGELLI<br />

ENRICA STRETTOI<br />

MARINA ZICHE<br />

12


TRAVEL AWARD WINNERS<br />

Biagioni Martina Tuscan Doctorate School in Neuroscience,<br />

CNR Neuroscience Institute, Pisa, University of Florence, Italy<br />

Cupri Sarha Department of Drug Sciences, University of Catania, Italy<br />

Daszynski Damian Department of Pharmaceutical Sciences,<br />

University of Nebraska, Omaha, Nebraska, USA<br />

Fidilio Annamaria Department of Biomedical and Biothecnological Sciences,<br />

University of Catania, Italy<br />

Harper Angelica Department of Cell Biology,<br />

University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA<br />

Johnson William Department of Ophthalmology, Duke University, Durham,<br />

North Carolina, USA<br />

Joubert Fanny Institut de la Vision, Equipe S12,<br />

UMR_S968 INSERM, UMR_7210 CNRS, UPMC, Paris<br />

Kelley Ryan Skaggs School of Pharmacy and Pharmaceutical Sciences,<br />

University of Colorado Anschutz Medical Center, Aurora, Colorado, USA<br />

Landowski Michael Department of Ophthalmology, Duke University, Durham,<br />

North Carolina, USA<br />

Lazzara Francesca Department of Biomedical and Biotechnological Sciences,<br />

University of Catania, Catania Italy<br />

Liu Yang North Texas Eye Research Institute,<br />

University of North Texas Health Science Center,Fort Worth Texas, USA<br />

Mishra Manish Kresge Eye Institute, Wayne State<br />

University School of Medicine, Detroit, Michigan, USA<br />

Mody Avani North Texas Eye Research Institute,<br />

University of North Texas Health Science Center,Fort Worth Texas, USA<br />

Patel Gaurang North Texas Eye Research Institute,<br />

University of North Texas Health Science Center,Fort Worth Texas, USA<br />

Pattabiraman Padmanabhan Department of Ophthalmology,Case Western Reserve<br />

University, Cleveland, Ohio, USA<br />

Perdices Lorena Institute for Health Research of Aragón (IIS Aragón), Zaragoza, Spain<br />

Platania Chiara B. M. BIOMETEC University of Catania,Italy<br />

Roubeix Christophe Department of Ophthalmology,<br />

Charite University Medicine Berlin, Germany<br />

Schmitt Heather Department of Ophthalmology,<br />

University of Wisconsin-Madison, Charter St. Madison, USA<br />

Smedowski Adrian Department of Physiology,<br />

Medical University of Silesia, Katowice, Poland<br />

Stefanov Antonia Institute of Neuroscience, National Research Council, Pisa, Italy<br />

Toro Mario Department of Ophthalmology, University of Catania, Italy<br />

Webber Hannah North Texas Eye Research Institute,<br />

University of North Texas Health Science Center,Fort Worth Texas, USA<br />

Yu Bo Tulane University, New Orleans, Lousiana LA, USA<br />

13


GENERAL INFORMATION<br />

Onsite registration desk<br />

Thursday february 16 th , 15.00-17:15<br />

Friday february 17 th , 08.30-10:30<br />

Fees:<br />

Regular Members: € 425,00<br />

Non-Members: € 525,00<br />

Students, Post Doc: € 250,00<br />

Name dadges<br />

All participants must wear their name badges during the meeting.<br />

Badges allow admission to all sessions, breaks, lunches, receptions and the banquet.<br />

Accompanying persons<br />

You can register as many accompanying persons as you want.<br />

Fees:<br />

Welcome reception and banquet: € 120,00<br />

Banquet only: € 80,00<br />

Accompanying persons are not allowed to attend scientific sessions and exhibition area.<br />

Welcome reception<br />

The welcome reception will be held on thursday february 16 th from 18.40 to 20.00<br />

in the exhibit area, first floor.<br />

<strong>AOPT</strong> business meeting<br />

The <strong>AOPT</strong> business meeting will be held on friday february 17 th from 16.40 to 17.40<br />

All <strong>AOPT</strong> members are encouraged to attend.<br />

<strong>AOPT</strong> banquet<br />

The <strong>AOPT</strong> banquet open to all regitered participants will be held on saturday 18th<br />

from 19.00 in the Basilica da Basso.<br />

Clothing<br />

Clothing in business casual for all occasion.<br />

Liability and personal insurance<br />

The <strong>AOPT</strong> 2017 Organizers can not accept liability for personal accidents or<br />

loss of or damage to private property of participants and accompanying persons.<br />

Safety and security<br />

We kindly request you not to leave bags, suitcases or backpacks unattended<br />

at any time during the meeting.<br />

14


INFORMATION FOR PRESENTERS<br />

Language<br />

The official language of the <strong>AOPT</strong> 2017 <strong>Meeting</strong> is english.<br />

Oral presentation<br />

Presenters using a powerpoint presentation should bring it on memory stick (usb) and<br />

load in the preview room near the conference hall, between 08,15-08,30 for morning<br />

sessions during the luch for afternoon sessions. Presenters with powerpoint and video<br />

are requested to check their presentation to be sure they work properly. Macintosh users<br />

must convert their files to powerpoint in order to be used on the pc, otherwise must<br />

bring their own computer and VGA adaptor.<br />

Poster presentation<br />

Posters (70 base x 100 height) need to be assembled by 08,00 on friday 17th and rimane<br />

on display until sunday afternoon. During the posters session, presenters must stand at<br />

their posters to present and discuss about their work. Posters left up on sunday evening<br />

will removed and discarded.<br />

Recording policy<br />

Recording any presentation or poster is prohibited, except by <strong>AOPT</strong> agent, or by authors.<br />

15


30<br />

E<br />

wc<br />

32<br />

170<br />

210<br />

wc wc wc wc<br />

122<br />

I/11<br />

24<br />

150<br />

265<br />

150<br />

265<br />

150<br />

265<br />

150<br />

265<br />

25<br />

150<br />

265<br />

36<br />

170<br />

210<br />

saletta<br />

mq:20,71<br />

H:285<br />

PALAZZO DEGLI AFFARI PLAN<br />

GROUND FLOOR<br />

1st floor<br />

Registration<br />

Exhibitors<br />

Coffee and lunch<br />

Poster area<br />

E<br />

Main entrance<br />

Conference room<br />

Preview room<br />

E<br />

FIRST FLOOR<br />

Bar<br />

Poster area<br />

E<br />

B5 SIFI<br />

JOPT<br />

B2 Allergan<br />

Registration<br />

desk<br />

F.C.B.<br />

Exhibition - Coffee and lunch area<br />

Medici<br />

senza<br />

Frontiere<br />

Nicox Santen Alfa Intes Chiesi Experimentica Angelini<br />

Cloakroom<br />

16


13:30 - 15:00<br />

15:00 - 16:30<br />

JOPT Board <strong>Meeting</strong><br />

<strong>AOPT</strong> Board <strong>Meeting</strong><br />

PROGRAM AT-A-GLANCE<br />

Thurdsay, February 16<br />

Friday, February 17 Saturday, February 18<br />

Sunday, February 19<br />

09:20 - 10:40<br />

SESSION 2<br />

NEW INSIGHTS IN THE THERAPEUTIC APPROACHES<br />

TO AMD: IS THERE A HOPE FOR AN AFFORDABLE<br />

TREATMENT?<br />

08:40-10:20<br />

SESSION 6<br />

ADVANCES IN OCULAR DRUG DELIVERY AND ITS ROLE<br />

IN PATIENT'S COMPLIANCE<br />

08:40-10:20<br />

SESSION 10<br />

GENETIC AND ORPHAN EYE DISEASES<br />

10:40-10:50 Break & Exhibits 10:20-10:40 Break & Exhibits 10:20-10:40 Break & Exhibits<br />

10:50-12:10<br />

SESSION 3<br />

DIABETIC RETINOPATHY, A DISEASE OF INCREASING<br />

CONCERN<br />

10:40-12:00<br />

SESSION 7<br />

GLAUCOMA: RESEARCH AND DRUG DISCOVERY IN THE<br />

XXI CENTURY<br />

10:40-12:00<br />

SESSION 11<br />

OCULAR BLOOD FLOW: AN ISSUE FOR DIAGNOSIS AND<br />

THERAPY<br />

12:10-13:30<br />

IT-ARVO sponsored symposium<br />

12:00-13:20<br />

Lunch-n-Learn<br />

12:00-13:00<br />

Lunch-Panel<br />

13:30 - 14:50<br />

SESSION 4<br />

REGULATORY ISSUES GOVERNING OPHTHALMIC<br />

DRUGS: PHARMACOECONOMICS AND DRUG<br />

DEVELOPMENT IN A CHANGING WORLD<br />

13:20- 14:40<br />

SESSION 8<br />

NEUROPROTECTION IN OPHTHALMOLOGY: DO WE<br />

STILL NEED IT?<br />

13:00-14:40<br />

SESSION 12<br />

TREATMENT OF REFRACTIVE ERROR DEFECTS: WHAT IS<br />

THE ROLE OF OCULAR PHARMACOLOGY?<br />

14:50 - 15:10 Break & Exhibits 14:40-15:00 Break & Exhibits 14:40-15:00 Break & Exhibits<br />

15:00 - 17:15<br />

Registration<br />

15:10 - 16:40<br />

SESSION 5<br />

YOUNG INVESTIGATOR<br />

15:00-16:20<br />

SESSION 9<br />

OCULAR IMMUNE DISORDERS IN THE TIME OF<br />

GLOBALIZED MEDICINE<br />

15:00-16:40<br />

SESSION 13<br />

OCULAR SURFACE DISEASES: AN EMERGING CONCERN<br />

IN OPHTHALMOLOGY<br />

17:15 - 17:20 Opening remarks<br />

16:40 - 17:40<br />

<strong>AOPT</strong> general business meeting<br />

16:20-17:30<br />

PANEL DISCUSSION<br />

THE ENDOVITREAL INSERTS TO REDUCE THE BURDEN<br />

OF ENDOVITREAL INJECTIONS IN AMD AND DME<br />

16:40-17:00<br />

Closing remarks<br />

17:20 - 18:40<br />

SESSION 1<br />

BIOMARKERS AND TECHNOLOGY FOR SUBTYPING EYE<br />

DISEASE: EBABLERS OF TRANSLATIONAL MEDICINE<br />

AND TARGETED THERAPIES<br />

17:40 - 19:00<br />

POSTER SESSION<br />

BREAK AND EXHIBITS<br />

17:30-18:40<br />

KEYNOTE ADDRESS<br />

FUNGAL KERATITIS, A MAJOR CAUSE OF BLINDNESS<br />

AND VISUAL IMPAIRMENT WORLDWIDE, ESPECIALLY IN<br />

DEVELOPING COUNTRIES<br />

18:40 - 20:00<br />

Welcome reception<br />

19:00-21:30<br />

BANQUET DINNER<br />

BASILICA DA BASSO<br />

17


KEYNOTE SPEAKER<br />

ERIC PEARLMAN PhD.<br />

Director of Institute for Immunology<br />

University of California, Irvine<br />

Prof. Pearlman started his studies at University of Glasgow, he earned his PhD<br />

in Microbiology at University of Texas (Health Sciences Center – San Antonio).<br />

He started its academic research studying “river blindness” – onchocerciasis.<br />

Eric Pearlman was part of the research group that found out that a bacterium,<br />

living inside the worm Onchocerca volvulus, was the real cause of inflammation<br />

and blindness, rather than the worm itself.<br />

These results have been published on Science in 2002.<br />

In 2005-2006, there was an outbreak of fungal keratitis in USA, northern Europe<br />

and UK; those corneal infections were difficult to treat and were related to contamination<br />

by Fusarium solani of a lens care product. Furthermore, this fungal<br />

keratitis is a major cause of blindness in developing world, because spores (conidia)<br />

are common in the soil and in the air of rural areas.<br />

Currently, Pearlman’s research is focused on Fusarium keratitis and his research<br />

group developed an animal model of this disease, in order to identify the biochemical<br />

mechanism of this infection and best pharmacological targets, to further<br />

develop new therapies.<br />

18


SCIENTIFIC<br />

PROGRAM<br />

under the auspices of


THURSDAY, FEBRUARY 16<br />

15:00-17:15 Registration<br />

17:15-17:20 Opening remarks<br />

SESSION 1<br />

BIOMARKERS AND TECHNOLOGY FOR SUBTYPING EYE DISEASE:<br />

ENABLERS OF TRANSLATIONAL MEDICINE AND TARGETED THERAPIES<br />

Moderators: Oliver Zeitz, Dan Stamer<br />

17:20-17:40 Outside-ophthalmology perspective: State-of-the-art<br />

for biomarkers in oncology<br />

Friedhelm Bladt<br />

17:40-18:00 Mining retinal imaging data for biomarkers of disease<br />

and therapy<br />

Sebastian Waldstein<br />

18:00-18:20 The potential of ocular exosomal biomarkers as<br />

therapeutic targets, and diagnostic and<br />

prognostic indicators<br />

Mikael Klingeborn<br />

18:20-18:40 Tearfilm biomarkers<br />

Leopold Schmetterer<br />

18:40-20:00 WELCOME RECEPTION<br />

FRIDAY, FEBRUARY 17<br />

SESSION 2<br />

NEW INSIGHTS IN THE THERAPEUTIC APPROACHES TO AMD:<br />

IS THERE A HOPE FOR AN AFFORDABLE TREATMENT?<br />

Moderators: Cathy Bowes Rickman, Chiara Eandi<br />

09:20-09:40 From AMD-associated polymorphisms to drug target identification<br />

Florian Sennlaub<br />

09:40-10:00 Complement signaling ar the RPE: Implications for AMD<br />

Olaf Strauss<br />

10:00-10:20 Placental growth factor: An additional therapeutic<br />

target for AMD<br />

Sandro De Falco<br />

10.20-10.40 Effects of Anti-C5a therapy on early and wet models of AMD<br />

Cathy Bowes Rickman<br />

10:40-10:50 BREAK AND EXHIBITS<br />

20


SESSION 3<br />

DIABETIC RETINOPATHY, A DISEASE OF INCREASING CONCERN<br />

Moderators: Marina Ziche, Ash Jayagopal<br />

10:50-11:10 iPSCs as a novel strategy for vascular repair in the retina<br />

Maria Grant<br />

11:10-11:30 Angiogenic/inflammatory activity of humor vitreous in<br />

proliferative diabetic retinopathy<br />

Marco Presta<br />

11:30-11:50 B2R signaling in neo-angiogenesis<br />

Sandra Donnini<br />

11:50-12:10 Imaging vascular dysfunction in diabetic retinopathy<br />

Ash Jayagopal<br />

12:10-13:30 LUNCH PANEL<br />

IT-ARVO sponsored symposium<br />

SESSION 4<br />

REGULATORY ISSUES GOVERNING OPHTHALMIC DRUGS:<br />

PHARMACOECONOMICS AND DRUG DEVELOPMENT IN A CHANGING WORLD<br />

Moderators: Filippo Drago, Peter Kador<br />

13:30-13:50 Challenges to the economic evaluation of interventions<br />

for retinal conditions: A review of NICE technology<br />

appraisals in retinal vein occlusion and diabetic macular edema<br />

Chrissy Almond<br />

13:50:14:10 Combination therapies in retinal diseases: Anticipated hurdles<br />

for regulatory approval and health technology assessment.<br />

Is there a risk for patient access to new innovative<br />

medicine in ophthalmology?<br />

Jean Claude Castanier<br />

14:10-14:30 Off-label drug use in ocular pharmacology<br />

Lucia Gozzo<br />

14:30-14:50 Clinical prevention of cataracts in diabetic dogs by kinostat<br />

Peter Kador<br />

14:50-15:10 BREAK AND EXHIBITS<br />

SESSION 5<br />

YOUNG INVESTIGATOR SESSION<br />

Moderators: Malinda Fitzgerald, Alessia Pascale<br />

15:10-15:25 Treatment of HDAC3 selective inhibitor prevents retinal<br />

ganglion cell nuclear atrophy and apoptosis after acute<br />

and chronic optic nerve injury<br />

Heather Schmitt<br />

21


15:25-15:40 Neuroprotection and neuroregeneration of Retinal Ganglion<br />

Cells using products of peripheral nerves predegeneration<br />

Adrian Smedowski<br />

15:40-15:55 Epigenetics as a code for mitochondrial DNA mismatch<br />

and its dysfunction in diabetic retinopathy<br />

Manish Mishra<br />

15:55-16:10 P2X7 receptor as a pharmacological target in<br />

diabetic retinopathy<br />

Chiara Platania<br />

16:10-16:25 Spleen derived monocytes in subretinal inflammation<br />

Christophe Roubeix<br />

16:25-16:40 The role of canonical Wnt signaling and K-cadherin in<br />

the maintenance of intraocular pressure<br />

Hannah Webber<br />

<strong>AOPT</strong> GENERAL BUSINESS MEETING<br />

16:40-17:40<br />

POSTER SESSION<br />

Moderators: Shusheng Wang, Julie Crider<br />

17:40-19:00<br />

SATURDAY, FEBRUARY 18<br />

SESSION 6<br />

ADVANCES IN OCULAR DRUG DELIVERY AND ITS ROLE IN PATIENT'S COMPLIANCE<br />

Moderators: Uday B. Kompella, Claudio Bucolo<br />

08:40-09:00 Biodegradable implants for sustained drug release:<br />

Manufacturing considerations, drug stability,<br />

and drug release<br />

Uday B. Kompella<br />

09:00-09:20 What delivery strategy for poorly soluble drugs?<br />

Robert Gurny<br />

09:20-09:40 Sustained release microtechnologies for the treatment of<br />

neurodegenerative diseases of the posterior segment<br />

Maria Del Rocio Herrero Vanrell<br />

09:40-10:00 Melanin binding and active transport in the RPE:<br />

Impact on ocular drug delivery and<br />

pharmacokinetics<br />

Arto Urtti<br />

22


10.00-10:20 Recent advances in the application of<br />

lipid-based nanocarriers to ocular drug delivery<br />

Rosario Pignatello<br />

10:20-10:40 BREAK AND EXHIBITS<br />

SESSION 7<br />

GLAUCOMA: RESEARCH AND DRUG DISCOVERY IN THE XXI CENTURY<br />

Moderators: Carol Toris, Padmanabhan Pattabiraman<br />

10:40-11:00 NCX 667, a lead nitric oxide (NO)-donating compound for a<br />

new class of ocular hypotensive agents<br />

Francesco Impagnatiello<br />

11:00-11:20 New glaucoma drainage device designs for lowering of IOP<br />

Carol Toris<br />

11:20-11:40 New highly effective and long-acting anti-glaucoma drug,<br />

new periorbital delivery method<br />

David Woodward<br />

11:40-12:00 Glycosylation status of clusterin, a secretory chaperone<br />

protein, regulates phagocytic activity and apoptosis in<br />

trabecular meshwork cells<br />

Padmanabhan Pattabiraman<br />

12:00-13:20 LUNCH-N-LEARN<br />

Keynote speaker Eric Pearlman<br />

Lab managing&career negotiation Carol Toris & Thomas Yorio<br />

Working in Industry Achim Krauss<br />

Starting a company/entrepreneurship Peter Kador & Robert Gurny<br />

The balancing act-Research, teaching...life in general Malinda Fitzgerald<br />

Peer Review Dan Stamer<br />

SESSION 8<br />

NEUROPROTECTION IN OPHTHALMOLOGY: DO WE STILL NEED IT?<br />

Moderators: Neville Osborne, Iok-HouPang<br />

13:20-13:40 Enhancement of mitochondrial function non-invasively as a<br />

means to provide neuroprotectionin ophthalmology?<br />

Neville Osborne<br />

13:40-14:00 The challenges in conducting neuroprotection studies<br />

Iok-Hou Pang<br />

14:00-14:20 Modulating autophagy to achieve retinal neuroprotection<br />

Rossella Russo<br />

14:20-14:40 Melatonin prevents photoreceptors death during aging<br />

Gianluca Tosini<br />

14:40-15:00 BREAK AND EXHIBITS<br />

23


SESSION 9<br />

OCULAR IMMUNE DISORDERS IN THE TIME OF GLOBALIZED MEDICINE<br />

Moderators: Pedram Hamrah, Juana Gallar<br />

15:00-15:20 Immunological basis for ocular graft versus host disease and<br />

novel therapeutic targets<br />

Sabrina N. Copsel<br />

15:20-15:40 Targeting inflammation: Pathogenesis and novel<br />

treatments for dry eye<br />

Chiara Bonzano<br />

15:40- 16:00 Rationale and mechanisms of neuro-regenerative therapy in<br />

patients with ocular surface disease<br />

Pedram Hamrah<br />

16:00- 16:20 Neuroanatomical, behavioral and electrophysiological<br />

data in a mouse model of dry eye<br />

Fanny Joubert<br />

PANEL DISCUSSION<br />

Moderators: Achim Krauss, Teresio Avitabile<br />

16:20-17:30 The endovitreal inserts to reduce the burden of endovitreal<br />

injections in AMD and DME<br />

KEYNOTE ADDRESS<br />

Moderator: Thomas Yorio<br />

17:30-18:40 Fungal keratitis, a major cause of blindness and visual<br />

impairment worldwide, especially in developing countries<br />

Eric Pearlman<br />

19:00-21:30 BANQUET DINNER<br />

SUNDAY, FEBRUARY 19<br />

SESSION 10<br />

GENETIC AND ORPHAN EYE DISEASES<br />

Moderators: Cheryl Rowe-Rendleman, Santi Spampinato<br />

08:40-09:00 Focus on retinitis pigmentosa (RP):<br />

translatability of success in animal models of orphan<br />

and genetic diseases of the eye<br />

Claire Gelfman<br />

24


09:00-09:20 Progesterone analogs as neuroprotectants in animal models<br />

of retinitis pigmentosa<br />

Tom Cotter<br />

09:20-09:40 Neuroprotection in inherited retinal degenerations:<br />

Role of antioxidants and neurotrophins to preserve or<br />

rescue cone function<br />

Benedetto Falsini<br />

09:40-10:00 The metabolic and redox signaling controlled by the<br />

rod-derived cone viability gene NXNL1<br />

Thierry Léveillard<br />

10:00-10.20 Development of investigational gene therapy for<br />

RPE65-mediated inherited retinal disease<br />

Daniel Chung<br />

10:20-10:40 BREAK AND EXHIBITS<br />

SESSION 11<br />

OCULAR BLOOD FLOW: AN ISSUE FOR DIAGNOSIS AND THERAPY<br />

Moderators: Jeffrey Kiel, Leopold Schmetterer<br />

10:40-11:00 Regional differences in blood flow as the basis for<br />

understanding retinal vascular disease<br />

Toke Bek<br />

11.00-11:20 Novel treatment for diabetic retinopathy by drug<br />

repositioning<br />

Taiji Nagaoka<br />

11:20-11:40 Retinal and choroidal vascular responses to electrical<br />

brain stem stimulation in rats<br />

Clemens Strohmaier<br />

11.40-12:00 Retinal oxygen extraction in diabetes and glaucoma<br />

Doreen Schmidl<br />

12:00-13:00 LUNCH PANEL<br />

SESSION 12<br />

TREATMENT OF REFRACTIVE ERROR DEFECTS: WHAT IS THE ROLE OF<br />

OCULAR PHARMACOLOGY?<br />

Moderators: Christine F. Wildsoet, Caterina Gagliano<br />

13:00-13:20 Can drug delivery help solve the problem of myopia?<br />

Heather Sheardown<br />

13:20 -13:40 Efficacy of atropine for progressive myopia<br />

in Europeans: two year results and comparison<br />

with results from East Asia<br />

Jan-Roelof Polling<br />

25


13:40-14:00 Orthokeratology combined with long-term instillations of<br />

very small atropine concentrations: A pre-evaluation of the<br />

myopia stabilizing effect<br />

Elena Tarutta<br />

14:00-14:20 Design, synthesis, and characterization of a selective inhibitor<br />

for retinaldehyde dehydrogenase (ALDH1A) enzymes<br />

Angelica Harper<br />

14.20-14:40 Medication crosslinking of the sclera: An experimental<br />

implementation of a technology of sclera strengthening<br />

treatment of myopia<br />

Elena Iomdina<br />

14:40-15:00 BREAK AND EXHIBITS<br />

SESSION 13<br />

OCULAR SURFACE DISEASES: AN EMERGING CONCERN IN OPHTHALMOLOGY<br />

Moderators: David Goldblum, Christophe Baudouin<br />

15:00-15:20 Corneal gene therapy: Beyond viral vectors<br />

Alexander V. Ljubimov<br />

15:20-15:40 Severe ocular allergies: From pathophysiology to future therapies<br />

Andrea Leonardi<br />

15:40-16:00 Gabapentin eye drops for the treatment of ophthalmic pain and<br />

ocular surface inflammation<br />

Dario Rusciano<br />

16:00-16:20 Altered electrical activity of corneal sensory receptor fibers<br />

during regeneration after corneal microkeratome lesion<br />

in the guinea-pig<br />

Juana Gallar<br />

16:20-16:40 Unique hydrogel technology in vitro model representing<br />

corneal layers<br />

Agne Žiniauskaitė<br />

16:40-17:00 Closing remarks<br />

26


IT-ARVO CHAPTER MEETING<br />

Florence (Italy) Palazzo degli Affari<br />

Friday 17 February - 12:10/13:30<br />

Horizon AMD: the drugs<br />

Moderators: Claudio Bucolo, Chiara Eandi<br />

12:10-12:30 Wet AMD: update on pharmacological therapy<br />

Chiara Eandi<br />

University of Torino, Eye Clinic, Torino, Italy<br />

12:30-12:50 Atrophic AMD: a panorama of new molecules with realistic future<br />

Monica Jablonski<br />

University of Tennessee Health Science Center, Hamilton Eye Institute, Memphis, TN, USA<br />

12:50-13:10 Pharmacological strategy for atrophic AMD<br />

Konstantin Petrukhin<br />

Columbia University, Eye Institute Research Annex, New York, NY, USA<br />

13:10-13:30 Liver X receptor ligands: new candidates to treat dry AMD?<br />

Goldis Malek<br />

Duke University, Albert Eye Research Institute, Durham, NC, USA<br />

Segreteria Organizzativa<br />

Medeacom s.r.l - info@medeacom.org - www.medeacom.org


PLATFORM<br />

ABSTRACT


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 1<br />

OUTSIDE-OPHTHALMOLOGY PERSPECTIVE: STATE-OF-THE-ART<br />

FOR BIOMARKERS IN ONCOLOGY<br />

FRIEDHELM BLADT<br />

Director, Oncology Biomarker Strategists, Berlin, Germany<br />

Cancer treatment has made big advances in the past decade with the introduction<br />

of so-called targeted therapies. These small molecule inhibitors or antibodies<br />

against distinct molecular features of cancer cells should specifically increase<br />

antitumor efficacy and reduce unwanted adverse drug reactions, thus increasing<br />

the benefit for patients. Successful examples are e.g. imatinib for the treatment of<br />

chronic myeloic leukemia or crizotinib for the treatment of ALK-mutated lung<br />

cancer. These drugs were usually accompanied by programs to identify patients<br />

harboring these alterations, usually so far on the genetic or genomic level. The<br />

success of such biomarker enriched drug development programs has changed the<br />

perception and understanding of how precision medicine programs should be developed<br />

in oncology. To optimize success of new drugs, key factors include the<br />

rational selection of preclinical model systems, selection of (clinically suitable)<br />

biomarkers for patient selection and also the careful development of pharmacodynamic<br />

and safety biomarkers. Emerging technologies like RNA based selection,<br />

next generation sequencing and the use of liquid biopsies and more sensitive<br />

methods allowing to analyze circulating tumor (stem) cells or free DNA/RNA/<br />

miRNA might further change the diagnostic landscape in the coming years. The<br />

current approaches and limitations of biomarker programs in oncology will be<br />

highlighted here.<br />

30


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 1<br />

MINING RETINAL IMAGING DATA FOR BIOMARKERS OF<br />

DISEASE AND THERAPY<br />

SEBASTIAN WALDSTEIN<br />

Department of Ophthalmology<br />

Medical University of Vienna, Austria<br />

The introduction of high-resolution in-vivo imaging has revolutionized diagnosis<br />

and management of retinal diseases. However, modern imaging generates a<br />

prohibitively large amount of data that remains untapped by human observers.<br />

At the same time, research in computational image analysis is advancing rapidly.<br />

Machine learning and artificial intelligence methods are starting to open a<br />

window of opportunity to capture the wealth of biomarkers provided by modern<br />

imaging: Sophisticated segmentation algorithms are capable of detecting several<br />

known retinal and choroidal layers as well as pathognomonic lesions. Moreover,<br />

discovery of hitherto unknown biomarkers with massive image datasets (“Big<br />

Data”) by unsupervised machine learning are beginning to be realized.<br />

This contribution will provide an overview of current developments in the area<br />

of computational analysis of retinal imaging biomarkers, as well as a perspective<br />

of future applications in clinical practice and research.<br />

It will include a brief overview of relevant known imaging biomarkers, summarize<br />

developments in biomarker discovery and discuss the role of machine learning<br />

in the establishment of future clinical trial endpoints.<br />

31


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 1<br />

THE POTENTIAL OF OCULAR EXOSOMAL BIOMARKERS AS THERAPEUTIC<br />

TARGETS, AND DIAGNOSTIC AND PROGNOSTIC INDICATORS<br />

MIKAEL KLINGEBORN<br />

Department of Ophthalmology<br />

Duke University Medical Center, Durham, USA<br />

Interest in utilizing 30 -150 nanometer sized exosomes and other extracellular<br />

vesicles (EVs) as biomarkers of disease has increased exponentially in recent years.<br />

EVs (including exosomes) have several unique features that define ideal biomarkers:<br />

(i) a lipid bilayer provides protection for their RNA, DNA, and proteins cargo;<br />

(ii) they contain tissue-, cell-, or disease-specific proteins and nucleic acids; and<br />

(iii) their hardiness enables a wide range of methods for isolation and enrichment<br />

from a range of body fluids (e.g. plasma, serum, urine, aqueous humor, tears and<br />

vitreous).<br />

To identify biomarkers for retinal disease, we defined the proteome of exosomes<br />

from the retinal pigmented epithelium (RPE), which forms the outer blood-retinal<br />

barrier in the eye. The RPE is a highly polarized barrier, leading to the directional<br />

secretion of proteins, lipoprotein particles and EVs. Such a division dictates<br />

directed interactions between RPE and the systemic circulation (basolateral side)<br />

and the retina (apical side). As a model, we used primary cultures of differentiated<br />

porcine RPE monolayers on permeable supports.<br />

EVs were isolated from conditioned medium bathing either apical or basolateral<br />

RPE surfaces, from which exosomes were purified and processed for proteomic<br />

profiling. In parallel, EV size distribution and concentration were determined.<br />

Using protein correlation profiling mass spectrometry, a total of 556 proteins<br />

were identified in exosome preparations, 465 of which were uniquely released<br />

apically, and 16 uniquely released from the basolateral side. Basolaterally released<br />

exosomes and EVs from RPE cells theoretically enter the systemic circulation<br />

and thus basolateral-RPE specific exosomal proteins that we identified, such as<br />

Bestrophin-1, represent targets for immunoisolation of RPE-derived exosomes<br />

from blood.<br />

These data serve as a foundation for comparative studies aimed at elucidating the<br />

molecular pathophysiology of retinal diseases and to help identify potential therapeutic<br />

targets and systemic biomarkers for such diseases.<br />

32


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 1<br />

TEARFILM BIOMARKERS<br />

LEOPOLD SCHMETTERER<br />

Singapore Eye Research Institute<br />

Dry eye disease (DED) is a multifactorial disease affecting the ocular surface.<br />

The prevalence of the disease is high and multiple risk factors including age, female-gender<br />

and environmental factors have been described. A problem in patients<br />

with DED is that signs and symptoms correlate poorly.<br />

This is a problem for clinical care and treatment monitoring as well as for approval<br />

of novel treatments, because regulatory authorities request superiority versus<br />

vehicle in both symptoms and one sign. Classical signs include tear film break<br />

up time and Schirmer test. Both techniques share problems in terms of reproducibility<br />

and subjectiveness. In the present talk novel biomarkers for DED will<br />

be discussed. An overview of pros and cons of imaging parameters, molecular<br />

parameters and tear osmolarity parameters will be provided.<br />

33


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 2<br />

FROM AMD-ASSOCIATED POLYMORPHISMS TO DRUG<br />

TARGET IDENTIFICATION<br />

FLORIAN SENNLAUB<br />

Institut de la Vision, Sorbonne Universités<br />

UPMC Univ Paris 06, INSERM, CNRS<br />

Age-related macular degeneration (AMD) is a highly heritable major cause of<br />

blindness characterized by subretinal inflammation. Of all genetic factors, variants<br />

of Complement factor H (CFH) are associated with greatest linkage to AMD.<br />

Using loss of function genetics and orthologous models of AMD, we provide<br />

mechanistic evidence that deficiency in CFH completely prevents pathogenic<br />

subretinal accumulation of mononuclear phagocytes (MP) and accelerates resolution<br />

of inflammation. We show that MP-persistence arises secondary to binding<br />

of CFH to CD11b/CD18, which obstructs physiologically-occurring thrombospsondin-1<br />

(TSP-1)-CD47-mediated elimination of MPs from the subretinal space.<br />

The AMD-associated CFH402H isoform markedly increased this inhibitory effect<br />

on microglial cells, indicating a causal link to disease etiology.<br />

Pharmacological activation of CD47 accelerated resolution of both subretinal and<br />

peritoneal inflammation, which may be exploited in the therapy for chronic inflammatory<br />

diseases, including AMD.<br />

34


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 2<br />

COMPLEMENT SIGNALING AR THE RPE: IMPLICATIONS FOR AMD<br />

OLAF STRAUSS 1 , GERHILD WILDNER 2 , CHRISTIAN HUBER 1 , CATHARINA BUSCH 1 , BÄRBEL ROHRER 3<br />

1 Experimental Ophthalmology, Dept. Ophthalmology Charite University Medicine Berlin Germany<br />

2<br />

Dept. Ophthalmology Ludwigs-Maximilians University Munich, Germany<br />

3<br />

Dept. Ophthalmology Medical University of South Carolina, Charleston SC, USA<br />

Purpose: Age-related macular degeneration involves functional changes or degeneration<br />

(AMD) of the retinal pigment epithelium (RPE). Polymorphisms in<br />

complement genes are associated with the AMD-risk. These polymorphisms lead<br />

to a less efficiently controlled alternative pathway of the complement cascade and<br />

to accumulation of active complement compounds in the outer retina. This has<br />

led so far to research about the effects of the terminal complement complex on<br />

the RPE. Thus the purpose of the study is to investigate the effects of the anaphylatoxins<br />

C3a and C5a on RPE cells.<br />

Methods: Increases in intracellular free Ca2+ in response to anaphylatoxins were<br />

investigated by Ca2+-imaging in ARPE-19 cells using fura-2 as Ca2+-sensitive fluorescence<br />

probe. Down-stream signaling was investigated by western-blot analysis<br />

of phosphorylated proteins, qPCR and multiplex analysis of secreted proteins.<br />

Results: ARPE-19 cells but also native human RPE cells express the anaphylatoxin<br />

receptors C3aR, C5aR as well as the C3 and C5. C3a and C5a led to increases<br />

of intracellular free Ca2+. Using double stimulation we detected interactive<br />

signaling between C3aR and C5aR where C5a appeared as a dominating factor.<br />

The downstream Akt-kinase, PI3-kinase are activated and the the transcription<br />

factors FOXO1 and FoxP3 are phosphorylated. The stimulation by C3a or C5a or<br />

the combination of both changed the secretion of chemokines/cytokines.<br />

Discussion: It appears that the RPE is an active player in the local regulation of<br />

complement activity. The RPE reacts to TCC but also to the anaphylatoxins and<br />

can thus react with secretion of immune modulatory factors upon complement<br />

activity.<br />

35


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 2<br />

PLACENTAL GROWTH FACTOR: AN ADDITIONAL<br />

THERAPEUTIC TARGET FOR AMD<br />

SANDRO DE FALCO<br />

Angiogenesis Lab, Institute of Genetics and Biophysics<br />

National Research Council, Napoli, Italy<br />

Placental Growth Factor (PlGF), the second member of Vascular Endothelial<br />

Growth Factor (VEGF) family discovered, is redundant in physiological process<br />

but undoubtedly involved in pathological angiogenesis. It specifically binds VEGF<br />

receptor 1 that is expressed in endothelial cells but also in many other cellular<br />

types, among which pericytes and the inflammatory cells.<br />

Indeed, PlGF/VEGFR1 axis mediates both neovessels formation and stabilization<br />

as well as the inflammation associated to pathological angiogenesis. In preclinical<br />

models of pathological angiogenesis, the genetic ablation or the biochemical<br />

inhibition of PlGF strongly impair neovessels formation. Recent evidences<br />

corroborating the view that the inhibition of PlGF function may be considered<br />

for therapeutic treatments in AMD and other ocular neovascular diseases will be<br />

presented.<br />

36


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 2<br />

EFFECTS OF ANTI-C5A THERAPY ON EARLY AND WET MODELS OF AMD<br />

CATHERINE BOWES RICKMAN 1,2, CHRISTOPHER B. TOOMEY 1,2, MICHAEL LANDOWSKI 1,<br />

HOLLY DONG 3 , MIKAEL KLINGEBORNE 1 , UNA KELLY 1 , ONS HARRABI 3 , JOHN LIN 3 , AND DANIEL R. SABAN 1,4<br />

1<br />

Department of Ophthalmology, 2 Department of Cell Biology, 4 Department of Immunology Duke<br />

University Medical Center, Durham, USA; 3 Rinat, Pfizer Inc., South San Francisco, USA<br />

Purpose: Patients with age-related macular degeneration (AMD) are grouped into<br />

three major categories: early, late “dry” and “wet” AMD. Complement activation<br />

has been strongly implicated in the AMD disease process. However, the mechanism<br />

by which chronic complement activation leads to the chorioretinal pathology<br />

seen in AMD and how to best target complement dysregulation pharmacologically<br />

remains unclear. We tested the impact of pharmacologically targeting<br />

C5a, a product of complement activation that is an immune cell chemoattractant<br />

and pro-inflammatory mediator in two mouse models of AMD.<br />

Methods: Early “dry” AMD-like pathology (sub-RPE deposit formation, RPE<br />

damage and vision loss) was quantified in aged (90 weeks) complement factor H<br />

heterozygous (Cfh+/-) mice fed a high fat, cholesterol-enriched (HFC) diet for 8<br />

weeks (Cfh+/-~HFC) ± 30 mg/kg anti-C5a therapy administered 1x/week by intraperitoneal<br />

injection over 8 weeks. Choroidal mononuclear phagocyte (MNP)<br />

populations were quantitatively assessed by intra- and extravascular flow cytometry.<br />

“Wet” AMD-like pathology was quantified using the laser choroidal neovascularization<br />

(CNV) model in C57BL/6J mice ± anti-C5a therapy administered one<br />

day prior to CNV induction and at day 5 and 11 post laser treatment.<br />

Results: Systemic anti-C5a therapy blocks MNP recruitment into the RPE/choroid,<br />

but does not appear to protect Cfh+/-~HFC mice from the early AMD-like<br />

pathology (RPE damage and visual function loss), which develops over the 8<br />

weeks of HFC diet. In contrast, anti-C5a treatment reduces CNV lesion size in the<br />

“wet” AMD-like pathology seen in the laser CNV model.<br />

Conclusions: These findings establish a role of C5a in choroidal monocyte recruitment<br />

and suggests that blockade of C5a may be a viable monotherapy for<br />

CNV in AMD.<br />

37


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 3<br />

IPSCS AS A NOVEL STRATEGY FOR VASCULAR REPAIR IN THE RETINA<br />

MARIA GRANT<br />

professor of Ophthalmology at Indiana<br />

University Indianapolis, Indiana<br />

Vascular complications due to diabetes mellitus (DM) are the result of sustained<br />

vascular injury with insufficient vascular repair. In chronic diabetes, vascular<br />

reparative mechanism can be lost resulting in development of microvascular<br />

complications (MVC), such as diabetic retinopathy (DR). We assessed the reparative<br />

function of progenitor cells that circulate in the peripheral blood of diabetic<br />

individuals and found that the vascular wall-derived progenitor cells, endothelial<br />

colony forming cells (ECFCs), were depleted in diabetics with MVC. Bone marrow-derived<br />

progenitor cells, CD45+CD34+ were dysfunctional in diabetics with<br />

MVC. We found that human inducible pluripotent stem cells (hiPSCs)-derived<br />

ECFCs displayed the ability to form functional and durable blood vessels in vivo<br />

and conferred therapeutic revascularization by connecting with and remaining<br />

integrated with host rodent vessels long term. We characterized a mesoderm<br />

subset (SSEA5-KNA+ cells) generated from hiPSCs that gives rise to ECFCs. Finally,<br />

we used hiPSCs to generate CD34+CD45+ cells and tested the impact of<br />

co-administration of these cells with ECFCs within the vitreous. The addition of<br />

CD34+CD45+ cells with ECFCs resulted in the enhanced survival, function and<br />

reparative ability of the ECFCs. This beneficial effect was mediated by reducing<br />

retinal oxidative stress and inflammation. In summary, current interventions to<br />

foster normal vascular remodeling and restoration of blood flow to the ischemic<br />

and injured retina are limited. Our findings would support that hiPSC represent<br />

a novel tool to facilitate retinal vascular restoration and that combinations of vascular<br />

progenitors work synergistically to optimize repair.<br />

38


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 3<br />

ANGIOGENIC/INFLAMMATORY ACTIVITY OF HUMOR VITREOUS IN<br />

PROLIFERATIVE DIABETIC RETINOPATHY<br />

MARCO PRESTA<br />

Department of Molecular and Translational Medicine<br />

University of Brescia, Italy<br />

Diabetic retinopathy (DR), a major complication of diabetes mellitus,is the leading<br />

cause of visual impairment in the working-age population. It begins as non-proliferative<br />

retinal abnormalities and progresses to moderate and severe proliferative<br />

diabetic retinopathy (PDR) characterized by neovascularization and a persistent<br />

grade of inflammation. Even though laser photocoagulation represents the gold<br />

standard therapy for PDR, anti-angiogenic vascular endothelial growth factor<br />

(VEGF) inhibitors are widely used. However, several limitations to anti-VEGF interventions<br />

exist, including local and systemic adverse effects and poor response<br />

in a significant percentage of patients. Furthermore, production of other angiogenic<br />

factors and pro-inflammatory mediators may nullify and/or cause resistance<br />

to anti-VEGF therapies. Indeed, angiogenesis and inflammation are closely<br />

related processes that play a pivotal role in ocular diseases associated with retinal<br />

neovascularization. Thus, a tight cross talk may exist between angiogenesis and<br />

inflammation in PDR, inflammatory responses contributing to neovessel formation<br />

and vice versa.<br />

Starting from the observation that diabetic patients treated with salicylates for<br />

rheumatoid arthritis showed a lower incidence of DR, the effect of intravitreal<br />

administration of anti-inflammatory corticosteroids (e.g. triamcinolone acetonide)<br />

has been investigated. However, beneficial effects can be transient and associated<br />

with steroid-related adverse events.<br />

This calls for a better understanding of the cross talk between angiogenesis and<br />

inflammation in PDR in order to identify novel anti-inflammatory approaches<br />

able to suppress retinal neovascularization.<br />

To this aim, the study of the biological effects exerted by PDR vitreous on endothelial<br />

cellsmay represent a useful tool to investigate the relationship between<br />

neovascular and inflammatory responses in preclinical in vitro and in vivo experimental<br />

models.<br />

39


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 3<br />

B2R SIGNALING IN NEO-ANGIOGENESIS<br />

SANDRA DONNINI<br />

Department of Life Sciences,<br />

University of Siena, Italy<br />

Purpose: Abnormal retinal vascular permeability is the leading cause of vision<br />

loss in diseases such as diabetic retinopathy, exudative macular degeneration, retinal<br />

vascular occlusions, and others. The main cytokine involved in ocular vascular<br />

permeability is vascular endothelial growth factor (VEGF). VEGF antagonists<br />

have been successfully used as new treatment for diabetic retinopathy, however,<br />

local side effects and systemic complications have been reported.<br />

New therapeutic approaches to selectively block VEGF angiogenic and permeabilizing<br />

actions, while sparing VEGF protective and trophic actions are needed.<br />

Kinins, such as bradykinin (BK) and kallidin, play a primary role in the development<br />

of diabetic retinopathy by enhancing vascular permeability, leukocytes<br />

infiltration, and other inflammatory mechanisms. These deleterious effects are<br />

mediated by kinin B1 and B2 receptors (B1R and B2R), which are expressed in<br />

diabetic human and rodent retina. In this study we assessed the contribution of<br />

B2R signaling in angiogenesis.<br />

Methods and Results: We demonstrated that BK, through the activation of its<br />

B2R, enhances vascular permeability and promotes angiogenesis in in vitro and in<br />

vivo models, which are significantly inhibited by the B2R antagonist, Fasitibant.<br />

In endothelial and circulating pro-angiogenic cells, B2R stimulation elicited NFκB<br />

activation, leading to COX-2 overexpression, PGE-2 production and VEGF<br />

output. B2R antagonist prevented the BK/NF-κB axis and the ensuing amplification<br />

of inflammatory/angiogenic responses.<br />

Conclusion: Based on our findings, BK/B2R system appears to be involved in the<br />

control of angiogenesis, and Fasitibant has the properties to be further studied as<br />

an alternative drug in treatment of diabetic retinopathy and macular degeneration.<br />

40


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 3<br />

IMAGING VASCULAR DYSFUNCTION IN DIABETIC RETINOPATHY<br />

ASHWATH JAYAGOPAL<br />

F. Hoffman-La Roche, Ltd.<br />

Basel, Switzerland<br />

Vascular inflammation and barrier inetgrity damage are associated with initiation<br />

and progression of diabetic retinopathy. Imaging strategies are continously being<br />

developed to improve clinical management of this disease, by enabling early<br />

detection, staging of disease, and assessment of therapeutic response in patients.<br />

In this presentation, emerging strategies for imaging diabetic retinal vasculature<br />

in the clinic will be presented, including instrumentation, contrast agents, and<br />

image processing technologies. Applications of these approaches in imaging hypoxia,<br />

blood-retinal barrier dysfunction, and vascular inflammation will be discussed<br />

as important examples.<br />

41


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 4<br />

CHALLENGES TO THE ECONOMIC EVALUATION OF INTERVENTIONS FOR<br />

RETINAL CONDITIONS: A REVIEW OF NICE TECHNOLOGY APPRAISALS IN<br />

RETINAL VEIN OCCLUSION AND DIABETIC MACULAR EDEMA<br />

CHRISSY A. ALMOND 1 , KARL W. PATTERSON 1 , NIC J. BRERETON 1<br />

1<br />

BresMed, Sheffield, UK<br />

Purpose: To identify the main challenges to the economic evaluation of interventions<br />

for retinal conditions as part of health technology assessment.<br />

Methods: Review ofUK National Institute for Health and Care Excellence technology<br />

appraisals for the treatment of macular oedema due to retinal vein occlusion<br />

(3) and diabetic macular oedema (4).<br />

Results: The main challenge identified was adequately to capture the quality of<br />

life (QoL) improvement provided by treatment. Patients can be treated in their<br />

best-seeing eye, worse-seeing eye or both with implications for visual acuity, and<br />

hence quality-of-life benefit. Clinical trial data are often collected for the treated<br />

eye only, or separately to the non-treated eye, whereas in practice it is the impact<br />

on the whole person that is important. Other issues included appropriatenessof<br />

valuation of QoL and the extrapolation of data beyond clinical trials.<br />

Conclusions: Over time, advances have been made in the economic evaluation<br />

of these treatments in response to feedback from previous technology appraisals.<br />

However, further advances could be made as long-term efficacy data become<br />

available and with changes to the way data are collected in clinical trials.<br />

42


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 4<br />

COMBINATION THERAPIES IN RETINAL DISEASES: ANTICIPATED HURDLES FOR<br />

REGULATORY APPROVAL AND HEALTH TECHNOLOGY ASSESSMENT IN FRANCE<br />

JEAN CLAUDE CASTANIER 1 , OLIVIER WONG 2<br />

1Independent Consultant in Pricing and Reimbursement, Bandol France<br />

2<br />

Mediqualité, Paris<br />

The French HTA process assess the value of all products irrespective of their indication<br />

or mode of action. A committee of 21 members decides for 66 million of<br />

French inhabitants and focus mainly on clinical effectiveness. There is no regional<br />

regulation or funding. The country is highly centralized.<br />

The HTA body (HAS) uses several tools to advise the payer organization and the<br />

Ministry of Health on value.<br />

These tools are:<br />

• The global medical value (SMR) which drives the reimbursement rate from<br />

0%, 15%, 30%, 65% to 100%. This decision is mainly driven by the ratio safety/<br />

efficacy and the willingness to fund from a solidarity and ethical point of view.<br />

• The additional medical benefit (ASMR) from 1 to 5 impacts heavily the price<br />

negotiation.<br />

• The target of the eligible population for setting a price volume agreement, if<br />

applicable<br />

• The conditions for prescriptions such as restrictions to specialists or hospital<br />

or prior request of multi-team assessment or prior authorization from the<br />

sick fund or a special formulary called “médicament d’exception”.<br />

The economic value is assessed by HAS only for very innovative drugs through<br />

the QALY/ICER system however the genuine French payers are more interested<br />

on the budget impact modelling.<br />

The economic committee (CEPS) negotiates the price, the volume and the funding<br />

on top of the DRG (if applicable), depending on the value assessment delivered<br />

by the French HTA body, namely HAS.<br />

In ophthalmology care, for future innovative compounds access are likely to be<br />

heavily challenged. The reason is that the HTA body focus mainly on the primary<br />

end-point. This HTA has set the bar for a substantial improvement at 10<br />

letters (2 lines) improvement, on top of the best optimized SOC. Thus it will be<br />

important to be successful in France to focus rather on refractory patients, fast<br />

progressers or increase the length the trial, to capture the relevant benefit.<br />

For dry MD and geographic atrophy an important proportion of avoidance of<br />

fovea involvement will be a must have. Non inferiority is always challenged by<br />

HAS (French HTA body) and this has been translated in no launch in France of<br />

several products in Keratitis or chronic glaucoma.<br />

Ultimately to successfully launch a product or a technology in France, there is<br />

more need for early pipeline review and more interaction between “trialists” and<br />

researchers on the one hand, and HTA and payer experts, on the other.<br />

43


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 4<br />

OFF-LABEL DRUG USE IN OCULAR PHARMACOLOGY<br />

LUCIA GOZZO 1 , LAURA LONGO 1 , SILVANA MANSUETO 1 , FILIPPO DRAGO 1,2<br />

1<br />

Regional Pharmacovigilance Centre/Clinical Pharmacology <strong>Program</strong>, University Hospital of<br />

Catania, Italy; 2 Department of Biomedical and Biotechnological Sciences, University of Catania, Italy<br />

According to the EMA, off-label use “relates to situations where the medicinal<br />

product is intentionally used for a medical purpose not in accordance with the<br />

authorised product information.” Off-label prescribing is not currently regulated<br />

at European level but some Countries adopted specific rules. From 2014, Italy<br />

permits off-label use of less costly safe and effective drugs even in presence<br />

of authorized alternatives with higher cost. Then, bevacizumab was re-listed as<br />

therapeutic option for AMD.<br />

We analyzed data from the Registries and the Pharmacovigilance Network between<br />

2014 and 2016, in order to evaluate the use of bevacizumab, ranibizumab<br />

and aflibercept and ADRs reports.<br />

We found in Sicilian Registries 122 treatments with bevacizumab for AMD, 5,542<br />

with ranibizumab (2,498 for AMD), 2,365 with aflibercept (1,839 for AMD). In<br />

Sicily, 63 ADRs were reported with ranibizumab (43 non-serious, 7 serious with<br />

1 death, 13 undefined), 5 ADRs were reported with aflibercept (3 non-serious regarding<br />

treatment failure, 2 serious with 1 death) and no ADRs were reported for<br />

bevacizumab. At national level, 153 ADRs were reported with ranibizumab (69<br />

non-serious, 65 serious with 3 deaths, 19 undefined), 26 ADRs were reported with<br />

aflibercept (9 non-serious, 17 serious with 2 deaths) and 25 ADRs were reported<br />

for bevacizumab (4 non-serious, 19 serious with 1 death, 2 undefined).<br />

A discussion regarding off-label use of intravitreal bevacizumab is still in place<br />

for the putative increased risk for patient safety and for economic consideration.<br />

An European harmonized approach would be of great value to improve off-label<br />

drug use.<br />

44


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 4<br />

CLINICAL PREVENTION OF CATARACTS IN DIABETIC DOGS BY KINOSTAT<br />

PETER F. KADOR 1,2,3 , M. WYMAN 1 , M. PAULOS 1 , LYNETTE M. SMITH 4 , KAREN BLESSING 1,2<br />

AND THE KINOSTAT TRIAL STUDY GROUP<br />

1<br />

Therapeutic Vision, Inc. Omaha, 2 College of Pharmacy, 3 Department of Ophthalmology, 4 College of<br />

Public Health, 4 Department of Ophthalmology, University of Nebraska Medical Center,<br />

Omaha, Nebraska, USA.<br />

Purpose: Bilateral cataracts develop in a majority of diabetic dogs within the first<br />

year of diabetes. A 9-month randomized, masked, multicenter, placebo controlled<br />

Proof of Efficacy Clinical Trial was conducted to determine whether the topical<br />

aldose reductase inhibitor Kinostat® can significantly reduce the clinical development<br />

of blinding cataracts.<br />

Methods: Newly diabetic dogs of all sizes, breeds, and sex with only equatorial<br />

vacuoles of less than 360o present and no other ocular disease were recruited at 11<br />

centers in the United States and evaluated by board certified veterinary ophthalmologists<br />

at the time of enrollment and then at 1, 2, 3, 6 and 9 months. The dog’s<br />

owners administered the topical formulations TID. Dogs not developing cortical<br />

cataracts during the 9-month period are then given Kinostat® with ophthalmic<br />

evaluations required at 6-month intervals.<br />

Results: Of the 179 dogs recruited, 127 successfully completed the 9 month study<br />

and were analyzed for efficacy. The results confirm that the daily administration<br />

of Kinostat® to diabetic dogs significantly (p=0.0169) prevents cataract formation<br />

with the placebo group being 2.18 times more likely to develop cataracts. Longterm<br />

administration showed prevention up to 6 years. A required toxicology<br />

study found that daily application of Kinostat® at doses of up to 5x the recommended<br />

doses did not induce any direct local or systemic toxic effects in any of<br />

the tissues examined.<br />

Conclusion: Kinostat® is the first drug to significantly reduce the clinical development<br />

of diabetic cataracts and represents an alternate treatment paradigm that<br />

reduces the need for cataract surgery.<br />

45


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 5 - YOUNG INVESTIGATOR<br />

TREATMENT OF HDAC3 SELECTIVE INHIBITOR PREVENTS RETINAL<br />

GANGLION CELL NUCLEAR ATROPHY AND APOPTOSIS AFTER ACUTE AND<br />

CHRONIC OPTIC NERVE INJURY<br />

HEATHER M. SCHMITT 1,2,4 , GUOJUN CHEN 3,4 , YUYUAN WANG 3,4 , CASSANDRA L. SCHLAMP 1,4 ,<br />

SHAOQUIN GONG 3,4 , ROBERT W. NICKELLS 1,4<br />

1<br />

Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 2 Cellular<br />

and Molecular Pathology, University of Wisconsin-Madison, Madison, WI 3 BIONATES Theme,<br />

Wisconsin Institute for Discovery, Madison, WI 4 McPherson Eye Research Institute, University of<br />

Wisconsin-Madison, Madison, WI<br />

Purpose: In retinal ganglion cells (RGCs) affected by optic nerve crush (ONC),<br />

HDAC3 regulates nuclear atrophy as an early response to axonal injury. Conditional<br />

knockout of Hdac3 and HDAC3 selective inhibition with RGFP966 prevent<br />

nuclear atrophy post ONC. Systemic dosing of RGFP966, which crosses the<br />

blood brain barrier, is necessary however for application to chronic models of<br />

optic nerve injury.<br />

Methods: Investigation of an intravitreal injection of RGFP966 was done to assess<br />

optimal dosing for prevention of nuclear atrophy and apoptosis up to 14 days after<br />

ONC. Intraperitoneal (IP) doses (range of 0-10mg/kg) were given and assessed by<br />

mass spectrometry and immunofluorescence for histone deacetylation and RGC<br />

survival after ONC. DBA/2J mice, which develop glaucoma, were treated IP between<br />

6-10 months with the most effective dose; 2mg/kg every 3 days. Sustained<br />

release of RGFP966 was investigated using intravitreal injection of drug mixed<br />

with microparticles and subcutaneous injection of drug mixed with hydrogel.<br />

Results: A 2µM intravitreal injection of RGFP966 provided transient protection<br />

after ONC, and a 2mg/kg intraperitoneal injection of RGFP966 every 3 days was<br />

optimal for protection against cell loss up to 4 weeks after ONC. Importantly, inhibition<br />

of HDAC3 activity with repeated systemic dosing of RGFP966 protected<br />

against RGC loss in aged DBA/2J mice. Preliminary results indicate that sustained<br />

release of RGFP966 from intravitreally injected microparticles or subcutaneously<br />

injected hydrogel protected against histone deacetylation induced 4 weeks later.<br />

Conclusion: Extended release of HDAC3 inhibitor RGFP966 may serve as a therapeutic<br />

for chronic neurodegenerative diseases such as glaucoma.<br />

46


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 5 - YOUNG INVESTIGATOR<br />

NEUROPROTECTION AND NEUROREGENERATION OF RETINAL GANGLION<br />

CELLS USING PRODUCTS OF PERIPHERAL NERVES PREDEGENERATION<br />

ADRIAN SMEDOWSKI 1,2 , MARITA PIETRUCHA-DUTCZAK 1 , JOANNA LEWIN-KOWALIK 1<br />

1<br />

Department of Physiology, School of Medicine in Katowice, Medical University of Silesia, Katowice,<br />

Poland; 2 Clinical Department of Ophthalmology, School of Medicine with Division of Dentistry in<br />

Zabrze, Medical University of Silesia, Katowice, Poland<br />

Purpose: To investigate neuroprotective effects of intravitreal therapy using peripheral<br />

nerve predegeneration products-sciatic nerve homogenate and activated<br />

Schwann cells-towards Retinal Ganglion Cells (RGC) in rat glaucoma model.<br />

Methods: Experimental glaucoma was induced in Wistar rats unilaterally using<br />

“the Bead Model”. The right eye served as a healthy control. Animals received<br />

either intravitreal injection of Schwann cells-isolated from injured sciatic nerveon<br />

2nd day after glaucoma induction, either sciatic nerve homogenate-on day<br />

2nd, 7th or 14th after glaucoma induction. PBS injection was used as a negative<br />

control. Animals were bred up to 6 weeks and intraocular pressure was monitored<br />

using laboratory tonometer. After 6 weeks, animals were sacrificed, eyes<br />

with optic nerves were enucleated and processed for histology and immunohistochemistry.<br />

RGC survival was compared by counting RGC bodies and optic<br />

nerve axons from control and treated eyes.<br />

Results: In group treated with sciatic nerve homogenate, injection performed on<br />

14th day following glaucoma induction was correlated with the highest RGC survival<br />

(28% RGC loss in treated group vs 40% RGC loss in control group; p


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 5 - YOUNG INVESTIGATOR<br />

EPIGENETICS AS A CODE FOR MITOCHONDRIAL DNA MISMATCH AND ITS<br />

DYSFUNCTION IN DIABETIC RETINOPATHY<br />

MANISH MISHRA 1 , RENU A. KOWLURU 1<br />

1<br />

Kresge Eye Institute, Wayne State University School of Medicine, 4717 St Antoine St, Detroit,<br />

Michigan 48201, USA<br />

Purpose: Mitochondrial dysfunction plays a significant role in the development<br />

of diabetic retinopathy, and its DNA (mtDNA) is damaged with increased mtD-<br />

NA-mismatch, fueling into a futile cycle of free radicals. Compared to the other<br />

regions, the damage is more at the displacement loop (D-loop) of the mtDNA,<br />

a non-coding region important for mtDNA transcription and replication. DNA<br />

methyltransferases (Dnmts), enzymes that methylate cytosine-base forming<br />

5-methylcytosine (5mC), are activated and 5mC can be spontaneously deaminated<br />

to thymine, causing DNA-mismatch. Our aim is to understand the role of<br />

mtDNA methylation in mitochondrial DNA-mismatch and dysfunction in the<br />

development of diabetic retinopathy.<br />

Methods: Human retinal endothelial cells incubated in high glucose, with or<br />

without Dnmt inhibitor (5-Aza-2'-deoxycytidine, 5-Aza; 1μM) were analyzed for<br />

mtDNA methylation and mismatch using methylated-DNA immunoprecipitation<br />

and surveyor-nuclease digestion kits respectively. In same cell preparations,<br />

mitochondrial function was evaluated by measuring mtDNA encoded Cytochrome<br />

b (Cytb) gene transcription and electron transport chain complex-III<br />

activity.<br />

Results: High glucose increased mtDNA-mismatch at the D-loop compared to<br />

the other mtDNA regions. At the D-loop region, DNA methylation was also<br />

increased by ~2.5-fold. Regulation of Dnmt activity by 5-Aza ameliorated glucose-induced<br />

increase in mtDNA methylation and prevented mtDNA-mismatch.<br />

In same cell preparations, 5-Aza prevented glucose-induced decrease in<br />

Cytb expression, and the activity of complex-III.<br />

Conclusions: Dnmt inhibitors, via regulating the levels of mtDNA methylation,<br />

ameliorate mtDNA-mismatch, and prevent mitochondrial dysfunction.<br />

Thus, identification of pathways leading to aberrations of mtDNA sequence<br />

could help identify novel therapeutic targets to inhibit the development of diabetic<br />

retinopathy.<br />

48


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 5 - YOUNG INVESTIGATOR<br />

P2X7 RECEPTOR AS PHARMACOLOGICAL TARGET IN<br />

DIABETIC RETINOPATHY<br />

CHIARA BIANCA MARIA PLATANIA, GIOVANNI GIURDANELLA 1 , LUISA DI PAOLA 2 ,<br />

GIAN MARCO LEGGIO 1 , SALVATORE SALOMONE 1 , FILIPPO DRAGO 1 , CLAUDIO BUCOLO 1<br />

1<br />

Section of Pharmacology, Department of Biomedical and Biotechnological Sciences,<br />

School of Medicine, University of Catania, Catania, Italy<br />

2<br />

School of Engineering, University Campus BioMedico, Roma, Italy<br />

Purpose: To build and validate an in-silico/in-vitro approach for discovery of new<br />

anti-inflammatory ligands (P2X7 inhibitors) to be used for treatment of diabetic<br />

retinopathy.<br />

Methods: Homology modeling, protein contact network analysis, molecular<br />

docking, MM-GBSA calculations were carried out in order to built and validate an<br />

in-silico approach aimed in finding new ligands, selective toward the P2X7 receptor.<br />

Several P2X7 ligands have been studied by the in-silico approach described,<br />

the most promising P2X7 inhibitor has been tested on human retinal pericytes,<br />

cultured with high glucose levels (25 mM). The effects of the P2X7 inhibitor have<br />

been assessed by cell viability, LDH and IL-1β levels.<br />

Results: We have generated and validated an in-silico/in-vitro platform to discover<br />

novel P2X7 receptor inhibitors. The in-silico platform is able to identify selective<br />

P2X7 inhibitors, with high true positive rate. The P2X7 inhibitor with best<br />

predicted ADME properties has shown anti-inflammatory and protective activity<br />

in the described in-vitro model.<br />

Conclusions: Our data suggested that P2X7 receptor can be an interesting pharmacological<br />

target for diabetic retinopathy. Furthermore, our in-silico/in-vitro<br />

screening platform is suitable for discovery of new and effective P2X7 inhibitors<br />

to be used for treatment of diabetic retinopathy.<br />

49


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 5 - YOUNG INVESTIGATOR<br />

SPLEEN DERIVED MONOCYTES IN SUBRETINAL INFLAMMATION<br />

CHRISTOPHER ROUBEIX 1,3 ,SÉBASTIEN AUGUSTIN 2 ,SERGIO CRESPO-GARCIA 1<br />

SOPHIE LAVALETTE 2 , NADINE REICHHART 1 , XAVIER GUILLONNEAU 2 , OLAF STRAUβ 1,3 , FLORIAN SENNLAUB 2,3<br />

1Department of Ophthalmology, Charite University Medicine Berlin, Berlin Germany<br />

2Institut de la Vision, UMRS 968, UPMC, Paris France<br />

3<br />

Berlin Institute of Health (BIH), Berlin, Germany<br />

Purpose: Angiotensin II type 1 receptor expressing mononuclear phagocytes<br />

(AT1+MPs) originating from the spleen have been shown to play an important<br />

role in the recruitment of circulating inflammatory CCR2+Mo in ischemic myocardial<br />

inflammation. The involvement of CCR2+Mo in Age Related Macular<br />

Degeneration (AMD) progression is well established.<br />

We examined here the role of splenic AT1+MPs in subretinal inflammation and<br />

choroidal neovascularization (CNV).<br />

Methods: Subretinal inflammation and choroidal neovascularisation (CNV) was<br />

induced by laser-injury (450mW; 250um; 50ms) in eyes of C57Bl6 or CCR2 KO<br />

mice daily treated or not by intraperitoneal AT1 antagonist Losartan (125mg/kg/<br />

day), and carrying or not subcutaneous osmotic pumps releasing systemic Angiotensin<br />

II (1ug/kg/min) with or without splenectomy. 7 days after the laser<br />

impacts IBA1+ and AT1+ subretinal MPs and CD102+CNV were quantified on<br />

immune-stained retinal and RPE/choroidal flatmounts.<br />

Results: Our immunostaining revealed 2 distinct sub-populations of subretinal<br />

MPs, Iba1+AT1--and Iba1+AT1+-MPs. Pharmacological antagonism of AT1 by<br />

losartan significantly decreased whereas AngII osmotic pumps exacerbated the<br />

number of both subretinal MP types and CNV. Interestingly, splenectomies significantly<br />

decreased subretinal MP accumulation and CNV and prevented the<br />

proinflammatory effect of systemic AngII. The deletion of CCR2 did not affect<br />

the recruitment of the AT1+MPs.<br />

Conclusion: Our study shows that Iba1+AT1+-MPs participate in subretinal inflammation,<br />

their infiltration of the subretinal space is independent to CCR2/<br />

CCL2 pathway, but strongly favored by systemic AngII.<br />

The observation that splenectomy prevented this effect suggests that splenic<br />

AT1+MPs participate importantly in the process. Our study might help explain<br />

why hypertension confers a risk to develop AMD.<br />

50


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 5 - YOUNG INVESTIGATOR<br />

THE ROLE OF CANONICAL WNT SIGNALING AND K-CADHERIN IN<br />

THE MAINTENANCE OF INTRAOCULAR PRESSURE<br />

HANNAH C. WEBBER, JACLYN Y. BERMUDEZ, CAMERON J. MILLAR, WEIMING MAO, ABBOT F. CLARK<br />

North Texas Eye Research Institute, University of North Texas Health Sciences Center,<br />

Fort Worth, Texas, USA<br />

Purpose: Primary open angle glaucoma is associated with increased intraocular<br />

pressure (IOP) and pathological changes in the trabecular meshwork (TM). Inhibition<br />

of canonical Wnt signaling in the TM raises IOP, though underlying<br />

mechanisms behind this remain unknown. We hypothesize that canonical Wnt<br />

signaling in the TM regulates IOP via cadherins junctions.<br />

Methods: NTM cells (gift from Novartis) were treated with or without 100ng/<br />

ml recombinant Wnt3a or 1ug/ml sFRP-1 for 4-48 hours. Membrane fractions or<br />

whole cell lysates were isolated for western immunoblotting (WB) and probed<br />

for cadherins and β-catenin. NTM cells were also immunostained for cadherins<br />

or β-catenin. RNA was extracted from NTM cells for cDNA synthesis and qPCR<br />

analysis of cadherins. Ad5.CMV recombinant adenoviruses encoding K-cadherin<br />

and/or sFRP-1 were injected into eyes of 4-6 month old female BALB/cJ mice<br />

(n=6/group).<br />

Conscious IOP was measured for 35 days. NTM cells were plated for cellular impedance<br />

assays using the Acea iCelligence system and transfected with 0.5nM<br />

non-targeting or K-cadherin siRNA.<br />

Results: WB showed that Wnt3a increased β-catenin and K-cadherin expression,<br />

which was inhibited with addition of sFRP-1.<br />

Immunostaining showed Wnt3a induced β-catenin accumulation on the cell<br />

membrane. qPCR showed Wnt3a significantly increased K-cadherin expression<br />

(n=3, p


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 6<br />

BIODEGRADABLE IMPLANTS FOR SUSTAINED DRUG RELEASE:<br />

MANUFACTURING CONSIDERATIONS, DRUG STABILITY AND DRUG RELEASE<br />

UDAY B. KOMPELLA<br />

University of Colorado Anschutz Medical Campus<br />

A biodegradable implant based on poly(lactic-co-glycolic) acid (PLGA) is approved<br />

by the US FDA for sustained delivery of a corticosteroid in treating back of the<br />

eye diseases. Any competing generic product for such an implant typically requires<br />

a human clinical study comparing the generic product with the reference,<br />

brand product. Such a study, while critical, is expected to be expensive and cumbersome.<br />

The long term goal of this study is to understand differences in implant<br />

physicochemical and drug release properties based on manufacturing methods.<br />

Further, it is our goal to assess batch to batch and within batch variations in the<br />

manufactured implants. This presentation will summarize our experience to date<br />

with dexamethasone-PLGA implants manufactured using melt-compression and<br />

hot-melt extrusion methods. We discovered that dexamethasone cumulative release<br />

when monitored over several weeks exhibits a rise and a fall behavior, indicative<br />

of drug degradation in the release medium.<br />

Using an LC-MS method, we identified over 10 major degradation products of<br />

dexamethasone during in vitro release studies. The extent of degradation was<br />

incorporated into the release profiles in order to estimate to actual drug release<br />

patterns. The observed degradation in vitro may not be relevant to in vivo release<br />

in the vitreous humor, where prolonged retention of either drug or degradation<br />

product in the vitreous humor is unlikely.<br />

52


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 6<br />

WHAT DELIVERY STRATEGY FOR POORLY SOLUBLE DRUGS?<br />

ROBERT GURNY 1,2 , THIBAULT MUGNIER 1 , NAOUAL DAHMANA 2 , VERENA SANTER 2 ,<br />

YOGESHWAR KALIA 2 , DORIS GABRIEL 1<br />

1<br />

Apidel SA, 29, Quai du Mont Blanc, CH 1201-Geneva, Switzerland,<br />

2<br />

University of Geneva, University of Lausanne, Switzerland, 1, Michel Servet,<br />

CH 1211-Geneva 4, Switzerland<br />

The development of aqueous eye drop formulations is challenging, because of<br />

poor water solubility and low corneal bioavailability of numerous drugs (APIs).<br />

For example natamycin (Natacyn®, Alcon) is on the market since many years as<br />

a suspension-based formulation, which contains 50 mg/mL of the drug in form<br />

of micrometer-sized particles. Cyclosporine (Ikervis®, Santen) is commercialized<br />

as an emulsion and fusidic acid (Fusithalmic®, Leo) and prednisolone acetate (Pred<br />

Mild®, Allegan) are also presented as suspensions.<br />

We will a present a novel innovative formulation approach for several poorly<br />

soluble APIs in form of a nanocarrier based system (PEGylated fruit acids, methoxy<br />

poly(ethylene) hexyl-substituted poly (lactic acid) (mPEGhexPLA)). Using<br />

this strategy, we successfully reduced the particle size of the existing product by<br />

a factor of up to 500 times with a particle size in the lower nano-range (approx.<br />

10-20nm). The formulations developed are perfectly clear solutions allowing<br />

smooth transport of the drug into corneal structures without transient blurring<br />

and/or local irritation often reported after application of suspension-based preparations.<br />

In vitro activity of mPEGhexPLA nanocarriers was found comparable<br />

or superior to free, suspended particulate formulations. Corneal penetration of<br />

the novel nanocarrier based formulations was significantly increased compared<br />

to suspension-based formulations and allowed to obtain comparable tissue levels.<br />

For example, a 100x lower formulation strength of natamycin (0.05%w/w nanocarrier<br />

based formulation) showed after 6 hrs in the pig cornea comparable tissue<br />

levels as Natacyn® (5% w/w) suspension.<br />

Given the localized nature of the infection a topical treatment is particularly attractive<br />

with lower doses. Formulation optimization of numerous suspended eye<br />

products should be envisioned in many cases in order to increase the comfort of<br />

the patient and decrease the amount of active in the formulation and therefore<br />

avoiding side effects from systemic absorption trough the nasal mucosa.<br />

53


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 6<br />

SUSTAINED RELEASE MICROTECHNOLOGIES FOR THE TREATMENT OF<br />

NEURODEGENERATIVE DISEASES OF THE POSTERIOR SEGMENT<br />

MARIA DEL ROCIO HERRERO VANRELL 1,2 , ALICIA ARRANZ-ROMERA 1,2<br />

CRISTINA GARCÍA- -CABALLERO 1 , SERGIO ESTEBAN- PÉREZ 1,2 , IRENE.T. MOLINA-MARTÍNEZ 1,2<br />

IRENE BRAVO-OSUNA 1,2<br />

1<br />

Department of Pharmacy and Pharmaceutical Technology (Research Group 920415), Faculty of<br />

Pharmacy, Complutense University, 28040 Madrid, Spain; 2 Red Temática de Investigación<br />

Cooperativa Sanitaria en Enfermedades Oculares (Oftared) e<br />

Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain<br />

Purpose: Pathologies affecting the optic nerve and the retina are the major causes<br />

of irreversible blindness in elderly population. Most of them are chronic and multifactorial<br />

and requiere mantained concentrations of the active substance in the<br />

site of action during long periods of time. In some cases, frequent injections are<br />

needed to control the disease. Administration of neuroprotective, antiapoptotic<br />

and antioxidant substances, has demonstrated to delay the degeneration. Drug<br />

Delivery Systems emerge as therapeutic tools to avoid succesive administration.<br />

Among them, microparticulate systems has gained a lot of interest as they are<br />

employed for long term delivery and multiloading purposes. The main advantage<br />

of these formulations is that they do not need surgery procedures for their administration<br />

and can be injected as a conventional injection. Furthermore, they<br />

disappear from the site of administration once the drug has been released.<br />

Methods: Application of different microtechnologies to load particles with several<br />

active substances. Characterization of the microparticles and evaluation of the<br />

efficacy in animal models of neurodegenerative diseases of the posterior segment.<br />

Results: Microparticles are effectively able to co-encapsulate biotechnological<br />

products and low molecular weight molecules. The particles release the therapeutic<br />

agents during long term. Therapeutic efficacy was demonstrated after MPs<br />

administration (intravitreal and periocular routes).<br />

Conclusions: Microparticles effectively co-encapsulate biotechnological products<br />

and low molecular weight molecules. Particles release the therapeutic agents<br />

for several months. The efficacy of the formulations have been demonstrated in<br />

animals models of optic nerve and retinal degeneration.<br />

54


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 6<br />

MELANIN BINDING AND ACTIVE TRANSPORT IN THE RPE:<br />

IMPACT ON OCULAR DRUG DELIVERY AND PHARMACOKINETICS<br />

ARTO URTTI<br />

Faculty of Pharmacy, University of Helsinki, Helsinki,<br />

Finland; School of Pharmacy, University of Eastern Finland, Kuopio, Finland<br />

Retinal pigment epithelium (RPE) is a key tissue in blood retina barrier. The RPE<br />

cells are highly pigmented and, therefore, capable of binding many drugs that<br />

bind to melanin. Drug transport in the RPE might be affected by the transporter<br />

protein, but the expression of these proteins has not been quantitated in the RPE<br />

cells. We investigated melanin binding and transporter expression in the RPE,<br />

and simulated potential interplay of these factors.<br />

Melanin was isolated from the porcine RPE and binding of more than 20 compounds<br />

to melanin was investigated.<br />

The results indicate very broad range in melanin binding. We investigated expression<br />

of drug transporters in the RPE cells, and quantitated expression of 16<br />

transporters, while 25 transporters were below the quantitation limit.<br />

We carried out also cell binding studies to the isolated RPE cells and modelled<br />

the cellular kinetics. The simulations suggest that there is a significant interplay<br />

between the melanin binding and permeability of drug in the plasma membranes.<br />

This indicates that melanin binding and active drug transport together are affecting<br />

drug distribution and accumulation to the RPE cells.<br />

55


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 6<br />

RECENT ADVANCES IN THE APPLICATION OF LIPID-BASED<br />

NANOCARRIERS TO OCULAR DRUG DELIVERY<br />

ROSARIO PIGNATELLO<br />

NANO-i – Research Center for Ocular Nanotechnology<br />

Department of Drug Sciences, University of Catania, Catania, Italy<br />

Controlled release of drugs to the eye tissues is still an important, though challenging<br />

topic of research. The eye is an organ highly protected from extraneous<br />

compounds by anatomical, functional and biochemical mechanisms. Such<br />

defense tools often limit the time of contact of the therapeutic formulation with<br />

the eye surface and lead to an insufficient bioavailability of the applied drugs, especially<br />

at the level of the posterior segment.<br />

Many nanotechnology strategies have been exploited for the diagnosis and cure<br />

of ocular diseases. Nanosized ocular drug delivery systems have given important<br />

results in the last years, both as topical applications on the eye surface or after<br />

intraocular administration. These colloidal carriers can be suitably engineered<br />

to overcome corneal and retinal barriers to drug penetration, protect the encapsulated<br />

drug, enhance compliance and safety of ophthalmic drugs, and prolong<br />

their activity by a controlled and/or prolonged site-specific release profile.<br />

Although the basic research on ophthalmic delivery systems supplies many technological<br />

approaches, very few of them have been able to reach a clinical relevance<br />

or to be translated into pre-industrial or industrial applications.<br />

The main reason lies in the complexity and specificity of the formulation parameters<br />

that ophthalmic products always require to tackle the high sensitivity of<br />

ocular tissues.<br />

The lecture will survey some of the more recent papers and patents regarding<br />

nanotechnology applications to ophthalmic controlled and targeted drug and<br />

gene delivery, with a specific attention to lipid-based nanocarriers, such as solid<br />

lipid nanoparticles (NLC), nanostructured lipid vectors (NLC), liposomal systems,<br />

micelles, etc.<br />

56


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 6<br />

NCX 667, A LEAD NITRIC OXIDE (NO)-DONATING COMPOUND FOR A NEW<br />

CLASS OF OCULAR HYPOTENSIVE AGENTS<br />

FRANCESCO IMPAGNATIELLO 1 , E. BASTIA 1 , N. ALMIRANTE 1 , C. TORIS 2 , C: LANZI 3 , E. ONGINI 1 ,<br />

E. MASINI 3 ,M.V.W BERGAMINI 4<br />

1<br />

Nicox Research Institute, Milan, Italy; 2 Department of Ophthalmology, Case Western Reserve<br />

University, Cleveland, OH; 3 Department of NEUROFARBA, University of Florence, Florence, Italy;<br />

4<br />

Nicox Ophthalmics, Inc., Fort Worth, TX, USA<br />

Primary open-angle glaucoma (POAG) is a common ocular disorder affecting ˜2%<br />

of the adult population and is the second-leading cause of blindness worldwide.<br />

The predominant risk factor for glaucoma progression is an increase in intraocular<br />

pressure (IOP), mediated via a reduction in aqueous humor outflow facility<br />

through the conventional (trabecular meshwork and Schlemm’s canal) outflow<br />

pathway.<br />

Current IOP lowering pharmacological strategies target aqueous humor production<br />

(i.e. β-blockers, carbonic anhydrase inhibitors) or drainage via the uveoscleral,<br />

nonconventional, outflow pathway (i.e. PGF2alpha agonists). Therapies<br />

targeting primarily the conventional pathway consist of older cholinomimetics<br />

and a rho kinase inhibitor recently approved in Japan.<br />

Data from a variety of experimental animal models coupled with recent clinical<br />

studies strongly support an important role of nitric oxide (NO) in lowering IOP<br />

by enhancing the facility of aqueous humor drainage via the conventional outflow<br />

route.<br />

NCX 667, a novel NO donor synthesized by Nicox, lowers IOP in rabbit and<br />

non-human primate models of ocular hypertension following single and repeated<br />

treatment schedules.<br />

As a consequence of NO donation, NCX 667 lowers IOP by 20% or more regardless<br />

of the specific model and animal species used. Furthermore, repeated acute<br />

dosing with NCX 667 elicits sustained IOP-lowering activity over time with no<br />

signs of tachyphylaxis or ocular discomfort.<br />

NCX 667 is a lead compound for further development to lower IOP in POAG<br />

patients.<br />

57


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 7<br />

NEW GLAUCOMA DRAINAGE DEVICE DESIGNS FOR LOWERING OF IOP<br />

CAROL TORIS<br />

Case Western Reserve University, Cleveland Ohio<br />

Over the past 10 years many drugs have come on the market that lower intraocular<br />

pressure (IOP) to treat glaucoma. These drugs need to be given topically<br />

one to 4 times daily. For an elderly presbyopic, arthritic patient, this can pose a<br />

challenge. Interest has turned to glaucoma drainage devices that are designed to<br />

improve aqueous humor drainage via numerous pathways. It is hoped that these<br />

devices would eliminate or reduce the need for topical drops and the compliance<br />

issues associated with their application.<br />

This presentation will describe numerous devices that are approved for human<br />

use and some designs that are in development. Devices can be categorized into<br />

snorkles that traverse the trabecular meshwork to provide direct communication<br />

between the anterior chamber and Schlemm’s canal, scaffolds and tubes that<br />

dilate Schlemm’s canal, tubes inserted into the suprachoroidal space to improve<br />

uveoscleral drainage and tubes that provide communication from the anterior<br />

chamber directly to the ocular surface or subconjunctival space.<br />

These devices reduce IOP by improving outflow facility or possibly uveoscleral<br />

outflow, or creating alternative routes that bypass the areas of greatest resistance<br />

to then allow drainage to the ocular surface. The cause of failures of these devices<br />

is predominantly clogging, erosion, or dislodging.<br />

In summary, glaucoma drainage devices (MIGS) may be the treatment of the future<br />

for glaucoma provided side effects can be avoided. These devices would not<br />

only benefit elderly patients but patients with little to no access to a pharmacy or<br />

routine doctor visits. While research continues on understanding signaling molecules<br />

and drainage pathways and developing treatments that may eventually<br />

cure glaucoma, the MIGS are proving to be important options for IOP lowering.<br />

58


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 7<br />

NEW HIGHLY EFFECTIVE AND LONG-ACTING ANTI-GLAUCOMA DRUG,<br />

NEW PERIORBITAL DELIVERY METHOD<br />

DAVID WOODWARD<br />

Dept of Bioengineering, Imperial College London, London, England<br />

Purpose: Two features define the future of glaucoma therapeutics: (1) greatly improved<br />

ocular hypotensive efficacy (2) a delivery method that improves patient<br />

convenience and compliance. These studies were intended determine whether<br />

dermal periorbital delivery of an exceptionally efficacious and potent ocular<br />

hypotensive agent 3-[(3’–fluoro-4-fluorobiphenyl-3-carbonyl) amino] phenoxyaceticacid<br />

isopropyl esterwould fulfil the required criteria for a next generation<br />

anti-glaucoma drug.<br />

Methods:Intraocular pressure was measured in ocular hypertensive and normotensive<br />

eyes of conscious monkeys, trained to accept pneumatonometry when<br />

under gentle restraint. For periorbital application the compound was formulated<br />

in polyethylene glycol and applied radially by using as roller ball device connected<br />

to a cylindrical reservoir.<br />

Results: A single 0.006% dose of3-[(3’–fluoro-4-fluorobiphenyl-3-carbonyl) amino]<br />

phenoxyaceticacid isopropyl ester,given as an eye drop, produced a profound<br />

decrease in intraocular pressure in “glaucomatous” monkeys that persisted for<br />

one-two weeks. It was not uncommon for a single 0.006% or 0.01% eyedrop to<br />

reduce intraocular pressure to 6-7 mmHg. Application to the periorbital dermis<br />

of a 0.1% dose to ocular normotensive monkeys produced a similarly profound<br />

reduction in intraocular pressure, which was well maintained.<br />

Conclusions: The compound 3-[(3’–fluoro-4-fluorobiphenyl-3-carbonyl) amino]<br />

phenoxyaceticacid isopropyl ester possesses the efficacy and duration of action<br />

properties to be considered as representative of the next generation of anti-glaucoma<br />

agents. Moreover, application to the periorbital skin using a roller ball device<br />

would be a more convenient method of ophthalmic drug delivery than eye<br />

drops and is non-invasive in contrast to other “dropless” technologies.<br />

59


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 7<br />

GLYCOSYLATION STATUS OF CLUSTERIN, A SECRETORY CHAPERONE<br />

PROTEIN, REGULATES PHAGOCYTIC ACTIVITY AND APOPTOSIS IN<br />

TRABECULAR MESHWORK CELLS<br />

PADMANABHAN PATTABIRAMAN, CAROL B. TORIS<br />

Department of Ophthalmology and Visual Sciences,<br />

Case Western Reserve University, Cleveland, Ohio<br />

Purpose: Clusterin, an N-glycosylated secretory molecular chaperone, requires<br />

glycosylation for its secretion, chaperone activity and its role in autophagy.<br />

Clusterin is expressed in trabecular meshwork(TM), found in aqueous humor,<br />

and its mRNA levels are decreased in TM of primary open angle glaucoma<br />

(POAG). Because very little is known about its functions in TM, we investigated<br />

the regulation of clusterin expression, glycosylation, secretion, and its functional<br />

role in TM.<br />

Method: Using immunoblotting and immunofluorescence analyses, we assessed-a)<br />

expression and secretion of clusterin in-primary human TM (HTM)<br />

cells, glaucomatous (GTM) and normal (NTM) TM lines, b) effects of stressors<br />

including TGFβ2 and elevated pressure (2X) on clusterin expression, c) role of<br />

clusterin glycosylation in HTM by expressing wild type secretory clusterin or<br />

mutant clusterin lacking glycosylation on–i) phagocytic activity by challenging<br />

HTM cells with pHRodo-labeled E.coli, ii) apoptosis using annexin-V-FITC and<br />

Akt signaling. All experiments had N>6.<br />

Results: Immunoblotting revealed the presence of fully glycosylated and nascent<br />

unglycosylated clusterin in HTM cells. Immunofluorescence for clusterin<br />

showed punctate staining in cytoplasm. Significant (p


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 8<br />

ENHANCEMENT OF MITOCHONDRIAL FUNCTION NON-INVASIVELY AS A<br />

MEANS TO PROVIDE NEUROPROTECTION IN OPHTHALMOLOGY?<br />

NEVILLE OSBORNE<br />

Nuffield Department of Clinical Neurosciences, University of Oxford, UK<br />

&Fundación de InvestigaciónOftalmológica,Oviedo, Spain<br />

The term neuroprotection in ophthalmology implies the use of pharmacological<br />

agents to slow-down insults to tissues like the retina and as a consequence<br />

preserve vision. A successful example of neuroprotection in ophthalmology is<br />

the use of VEGF antagonists in the treatment of age-related macular degeneration<br />

(AMD). However, challenges remain in providing credibility for the view<br />

that neuroprotection is a possibility for the successful treatment of diseases like<br />

diabetic retinopathy or glaucoma. For such diseases, laboratory studies suggest<br />

that if retinal mitochondrial functions can bepreserved this is likely to result in<br />

neuroprotection.<br />

However, for this idea to be tested agents will have to be delivered to the retina<br />

regularly with a prerequisite of having minimum side effects. Significantly, red<br />

light at wavelengths between 600 to 1000 nm isabsorbed by the mitochondrial<br />

photoacceptor moleculecytochrome c oxidase and in the process improvesmitochondrial<br />

energy metabolism thereby decreasing inflammation and enhancingcell<br />

survival. Studies on animal models with defined retinal injury, as well as<br />

retinal and optic nerve disease that mimic AMD, retinitis pigmentosa and glaucoma<br />

have now demonstrated that red light therapy attenuates cell death, protects<br />

retinal function and exerts anti-inflammatory actions.<br />

Such studies strongly suggest that neuroprotection for various retinal diseases are<br />

achievable by use of red light as a non-invasive methodology.<br />

Clinical trials are now being undertaken to determine ways of delivery of an<br />

appropriate amount of red light to the human retina for the treatment of both<br />

chronic and acute retinal diseases. One exciting possibility is to devise spectacles<br />

that increase the intensity of red light (in the range of 800-1000nm) specifically<br />

but nevertheless and have no effect on the normal wavelengths of the visual<br />

spectrum reaching the retina.<br />

61


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 8<br />

THE CHALLENGES IN CONDUCTING NEUROPROTECTION STUDIES<br />

IOK-HOU PANG<br />

Iok-Hou Pang, PhD, FARVO<br />

Professor & Chair<br />

Pharmaceutical Sciences, College of Pharmacy<br />

North Texas Eye Research Institute<br />

University of North Texas Health Science Center<br />

A critical unmet medical need in many retinal diseases, such as glaucoma, is the<br />

development of neuroprotective treatment. Unfortunately, many obstacles and<br />

challenges make this effort very difficult and currently unsuccessful.<br />

This presentation will discuss some of these challenges and propose potential<br />

solutions to reduce risk and lower budgetary hurdle for development of new<br />

therapies.<br />

62


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 8<br />

MODULATING AUTOPHAGY TO ACHIEVE RETINAL NEUROPROTECTION<br />

ROSSELLA RUSSO 1 , GIUSEPPE PASQUALE VARANO 1 , ANNAGRAZIA ADORNETTO 1 , FRANCESCA<br />

NAZIO 3 , LUIGI ANTONIO MORRONE 1,2 , MARIA TIZIANA CORASANITI 4 , FRANCESCO CECCONI 3 ,<br />

CARLO NUCCI 5 , GIACINTO BAGETTA 1,2<br />

1<br />

Department of Pharmacy, Health and Nutritional Sciences, Section of Preclinical and Translational<br />

Pharmacology, and 2 University Consortium for Adaptive Disorders and Head Pain (UCADH),<br />

Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity,<br />

University of Calabria, Arcavacata di Rende, Italy; 3 Department of Biology,<br />

University of Rome Tor Vergata, Rome, Italy; 4 Department of Health Sciences,<br />

University “Magna Graecia” of Catanzaro, Catanzaro, Italy; 5 Ophthalmology Unit, Department of<br />

Experimental Medicine and Surgery, University of Rome Tor Vergata Rome, Italy<br />

Purpose: Autophagy, the cellular process responsible for degradation and recycling<br />

of cytoplasmic components through the autophagosomal-lysosomal compartment,<br />

has been implicated in acute and chronic diseases. At variance with<br />

the latter, the role of autophagic modulation in the neurodegenerative process<br />

occurring in retinal ganglion cells (RGCs) exposed to glaucoma-related stressor<br />

stimuli is still debated.<br />

In an attempt to define autophagy efficiency as a determinant for RGC survival<br />

here we analyzed the autophagic response and the upstream regulatory mechanisms<br />

in retinas exposed to an ischemic insult.<br />

Methods: Retinal ischemia was induced in adult wild type C57BL/6J or GFP-LC3<br />

transgenic mice by transient elevation of intraocular pressure. Expression of<br />

autophagy related proteins (Atg) and upstream regulators (mTOR, AMPK) was<br />

studied by western blotting and immunofluorescence. RGCs were labeled by<br />

fluorogold and survival was assessed in AMBRA1+/- mice and upon rapamycin<br />

treatment or caloric restriction.<br />

Results: The expression of the autophagosomal-associated form of Atg8 (LC3II),<br />

was significantly reduced by ischemia, while the protein accumulated in the ganglion<br />

cell layer after 6 hours of reperfusion. A biphasic modulation of the autophagic<br />

substrate p62, characterized by a significant build up during the late phase<br />

of reperfusion that followed an earlier reduction, was also reported.<br />

Increased RGC death was observed in autophagy-deficient Ambra+/- mice subjected<br />

to retinal ischemia, while autophagy induction by rapamycin or caloric<br />

restriction improved RGC survival.<br />

Conclusion: Our results suggest that ischemic insult induces a dynamic modulation<br />

of autophagy in the retina and identify in the catabolic pathway an important<br />

endogenous neuroprotective mechanism that can be targeted to achieve<br />

neuroprotection.<br />

63


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 8<br />

MELATONIN PREVENTS PHOTORECEPTORS DEATH DURING AGING<br />

GIANLUCA TOSINI 1 , AIDA SANCHEZ-BRETANO 1 , ILARIA PIANO 1,2 , CLAUDIA GARGINI 2 ,KENKICHI BABA 1<br />

1<br />

Department of Pharmacology and Neuroscience Institute, Morehouse School of Medicine,<br />

Atlanta, USA; 2 Dipartimento di Farmacia, Universita di Pisa, Pisa, Italy<br />

Purpose: Several studies have shown that melatonin synthesis in the retina primarily<br />

occurs during the night and its levels are low during the day. Melatonin<br />

exerts its influence by binding to G protein-coupled receptors named melatonin<br />

receptor type 1 (MT1) and type 2 (MT2). MT1 and MT2 receptors activate a wide<br />

variety of signaling pathways and both receptors are present in the vertebrate<br />

photoreceptors where they form MT1/MT2 heteromers. Previous studies have<br />

shown that melatonin signaling is involved in the modulation of photoreceptor<br />

viability during aging and other studies have implicated melatonin in the pathogenesis<br />

of age-related macular degeneration.<br />

Methods: Melatonin-proficient mice (C3H-f+/+) and melatonin-proficient mice<br />

lacking melatonin receptors (MT1 or MT2) were used for the in vivo studies.<br />

Photoreceptor-like cells (661 W) were used for the in vitro. Melatonin signaling<br />

was investigated with western blotting, immunocytochemistry and Q-PCR.<br />

Results: Melatonin receptors knock-out mice showed a decrease in the number<br />

of cone photoreceptors during aging. Mice lacking melatonin receptors also<br />

showed an alteration in the daily activation of the AKT-FOXO1 cell survival<br />

pathway.<br />

In 661W melatonin activated pathways similar to what observed in rods and<br />

cones. In addition, melatonin prevented 661W cells death induced by 2 hours of<br />

exposure to H2O2 by preventing the activation of the Fas pathway.<br />

Conclusions: We believe that melatonin signaling via MT1/MT2 heteromers<br />

protects photoreceptors during aging. The neuroprotective effects of melatonin<br />

on photoreceptors cells (and possibly other retinal cells) are exerted by suppressing<br />

the activation of Fas pathway during night-time.<br />

64


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 9<br />

IMMUNOLOGICAL BASIS FOR OCULAR GRAFT VERSUS HOST<br />

DISEASE AND NOVEL THERAPEUTIC TARGETS<br />

SABRINA N. COPSEL<br />

Post Doctoral Associate , Laboratory of Robert Levy PhD<br />

Department of Microbiology and Immunology, University of Miami Miller School of Medicine<br />

Graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation<br />

(aHSCT) is a multiorgan disorder resulting from inflammatory cytokines<br />

and donor T cells which damage skin, liver, gastrointestinal tract, and<br />

the eye surface. Ocular GVHD (oGVHD) occurs in 60-90% of chronic GVHD<br />

patients and is characterized by inflammation, dry eye, Meibomian gland dysfunction,<br />

conjunctiva damage, punctate keratopathy, corneal ulceration and perforation.<br />

Our group has developed a novel pre-clinical matchedunrelated donor<br />

HSCT model that results in systemic and ocular GVHD with onset kinetics similar<br />

to what is clinically observed, enabling dissection ofoGVHD immune mechanisms.We<br />

demonstrated that the presence of donor Tcells in ocular tissue orchestrate<br />

therecruitment of inflammatory macrophages in this compartmentthat<br />

contribute to the ocular damageand recently, developed a scoring index for the<br />

ocular surface and adnexa.Therefore, regulating inflammatory cells recruited to<br />

ocular tissueis proposed as a strategy to ameliorate oGVHD. A recent approach to<br />

diminish inflammatory cytokines is targeting bromodomain and extra-terminal<br />

proteins (BET). We examined the ability of a new BET inhibitor (BETi) EP313<br />

in comparison with other BETi (JQ1or IBET-151) to regulate inflammatory cytokines<br />

using the macrophage RAW-264 cell line after LPS stimulation. EP313<br />

significantly decreased TNFα and IL-6 levels. To study the capacity of BETi to inhibit<br />

ocular inflammation, we utilized an in vivo model of corneal inflammation<br />

induced by topical LPS.Our data shows that BETi administered first systemically<br />

and then locally reduced corneal opacification and decreasedinflammatory cytokine<br />

expression.Because ocular GVHD is promoted by inflammation and donor<br />

T cells, we reason regulating both may effectively ameliorate this disorder.<br />

Transfer of expandedregulatory T cell (Tregs) is a promising therapy to suppress<br />

donor T cells and subsequently regulate GHVD. We therefore asked if BET inhibitors<br />

(BETi) could be combined with Tregs in vivowithout interfering with their<br />

phenotype/function/expansion. Strikingly, Tregs undergoing marked proliferationwith<br />

a novelprotocol (TNFRSF25 /CD25 stimulation) were not impaired in<br />

theirexpansion in the presence of EP313 and, in fact,exhibited stronger suppressive<br />

function vs Tregs expanded without EP313. Finally, we performed an aHSCT<br />

to examine the combined effect of in vivoexpanded Tregs and EP313. Importantly,<br />

the clinical GVHD score of mice receiving the combination strategy of Treg expansion+EP313<br />

was superior to all other groups and recipients of this regimen<br />

did not show ocular complications.In total,BETi treatment provides important<br />

advantages in regulating inflammation in the ocular compartment due to direct<br />

regulation of inflammatory cell function and promotion of Treg cell function.<br />

Therefore, we anticipate its combination with Treg cellular therapy willresult in<br />

blockingalloreactive cells and subsequent recruitment -and function of - infiltrating<br />

cells in ocular tissue thereby ameliorating oGVHD.<br />

65


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 9<br />

TARGETING INFLAMMATION: PATHOGENESIS AND NOVEL TREATMENTS<br />

FOR DRY EYE<br />

CHIARA BONZANO<br />

Clinica Oculistica, Università di Genova, Genoa, Italy<br />

The tear film, lacrimal gland, corneal and conjunctival epithelia and Meibomian<br />

glands work together as a functional unit to provide an efficient system recognized<br />

as the ocular surface. The integrity of this unit is necessary for the health<br />

and normal function of the eye and visual system.<br />

Recent studies show that immunological mechanisms also play a pivotal role in<br />

regulating the ocular surface environment. Our studies demonstrate how anti-inflammatory<br />

factors such as the expression of vascular endothelial growth<br />

factor receptor-3 (VEGFR-3) in corneal cells, immature corneal resident antigen-presenting<br />

cells, and regulatory T cells play an active role in protecting the<br />

ocular surface.<br />

Dry eye disease (DED) affects millions of people worldwide and negatively influences<br />

the quality of life for patients. In its most severe forms, DED may lead to<br />

blindness. The etiology and pathogenesis of DED remains largely unclear.<br />

The aim of this presentation is to summarize the role of the disruption of afferent<br />

and efferent immunoregulatory mechanisms that are responsible for the chronicity<br />

of the disease, its symptoms, and its clinical signs and to illustrate current<br />

anti-inflammatory treatments for DED.<br />

66


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 9<br />

RATIONALE AND MECHANISMS OF NEURO-REGENERATIVE THERAPY IN<br />

PATIENTS WITH OCULAR SURFACE DISEASE<br />

PEDRAM HAMRAH<br />

Cornea Service, Tufts New England Eye Center, Boston, MACenter for Translational<br />

Ocular Immunology, Department of Ophthalmology, Tufts Medical Center<br />

Tufts University School of Medicine, Boston, MA<br />

Corneal nerves, which may be involved in the pathogenesis of dry eye disease<br />

(DED), play a significant role in ocular surface health and function and are involved<br />

in corneal epithelial maintenance, tear secretion, and blinking. In vivo<br />

confocal microscopy (IVCM) studies have observed a significantly reduced subbasal<br />

nerve density in patients with DED, correlating to clinical severity and corneal<br />

sensation in these patients. Further, significant improvements in dry eye<br />

signs and symptoms after DED treatment were evident only in a subgroup with<br />

near-normal corneal SNFL, potentially explaining the variability of patients’ response<br />

to DED therapy. In addition, abnormal morphological changes of corneal<br />

nerves by IVCM have been observed in subsets of patients with DED with more<br />

severe symptoms, suggesting an underlying attempt of corneal nerves to regenerate,<br />

presumably subsequent to the nerve degeneration. Injured nerves are known<br />

to develop hypersensitivity (hyperalgesia), or become the source of spontaneous<br />

discharge (allodynia), explaining the hyperalgesia of some patients with DED. Regenerative<br />

activity is manifested by sprouting from endbulbs and the formation<br />

of microneuromas, seen as abrupt swelling of injured nerve endings and neurite<br />

sprouting. Recent evidence has demonstrated that the treatment of patients with<br />

DED and corneal neuropathic symptoms with autologous serum tears, showed<br />

restoration of nerve topography through nerve regeneration, correlating with<br />

improvement in symptoms. This supports the notion that corneal nerve damage<br />

results in alterations in afferent trigeminal pathways to, at least in part, result<br />

in patient symptoms. Thus, given the significant overlap of DED with corneal<br />

neuropathic disease, therapeutic strategies resulting in regeneration of damaged<br />

nerves may help alleviate patient signs and symptoms.<br />

regeneration of damaged nerves may help alleviate patient signs and symptoms.<br />

67


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 9<br />

NEUROANATOMICAL, BEHAVIORAL AND ELECTROPHYSIOLOGICAL<br />

DATA IN A MOUSE MODEL OF DRY EYE<br />

FANNY JOUBERT 1,2,3 , LAURENCE BODINEAU 5 , ELODIE REBOUSSIN 1,2,3 , PIERRE-SERGE LAUNAY 1,2,3 ,<br />

KARIMA KESSAL 1,2,3 , JOSÉ SAHEL 1,2,3,4 , CHRISTOPHE BAUDOUIN 1,2,3,4 , STÉPHANE MÉLIK PARSADANIANTZ 1,2,3<br />

AND ANNABELLE RÉAUX LE GOAZIGO 1,2,3<br />

1<br />

INSERM, U968, Paris, F-75012, France, 2 Sorbonne Universités, UPMC, Paris 06, UM 80, Institut<br />

de la Vision, 75012 Paris, France, 3 CNRS, UMR 7210, Paris, F-75012, France, 4 Centre Hospitalier<br />

National d’Ophtalmologie des Quinze-Vingts, Paris, F-75012, France, 5 Sorbonne Universités, UPMC<br />

Univ Paris 06, INSERM, UMR_S1158 Neurophysiologie respiratoire expérimentale et clinique, Paris,<br />

F-75013, France<br />

Purpose: Ocular surface diseases (OSDs) are among the most frequent ocular<br />

pathologies, with prevalence ranging 20% of the general population. The most<br />

frequent OSDs are dry eye disease associated with chronic ocular pain. Here we<br />

investigated the peripheral and central neuroinflammation and the ciliary nerve<br />

activity in response to dry eye induced ocular pain.<br />

Methods: RTqPCR and immunohistochemistry were used to measure the peripheral<br />

and central neuroinflammation and wiping test was used for measuring<br />

ocular sensitivity in adult male mice topically treated for 7 days with 0.2%<br />

benzalkonium chloride (BAK). For electrophysiological experiments, the mouse<br />

corneal epithelium was injured using a trephine (1.5 mm). After 24, 48 and 72<br />

hours, eye was placed in the two-compartment chamber and extracellular spontaneous<br />

impulse activity of the ciliary nerve was recorded.<br />

Results: BAK-treated animals developed severe dry eye, characterized by corneal<br />

inflammation and higher corneal sensitivity. We showed increased ATF3, FOS<br />

and Iba1 immunoreactivity and higher IL-6 and TNF-α mRNA levels in the trigeminal<br />

ganglion. Interestingly ocular inflammation induced higher FOS and<br />

Iba1 positive-cells in the sensory trigeminal complex in BAK animals.<br />

Futhermore, preliminary data showed altered basal activity of the ciliary nerve<br />

and modified response after mechanical stimulation in injured cornea.<br />

Conclusions: These works demonstrate that corneal inflammation/injury increases<br />

the corneal nociceptors activity and induced central neuroinflammatory<br />

process. Thus, altered activity in intracellular signaling might play a priming<br />

role in the central sensitization of ocular related brainstem circuits, which represents<br />

a significant factor in dry eye pain development.<br />

68


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 9<br />

FUNGAL KERATITIS, A MAJOR CAUSE OF BLINDNESS AND VISUAL<br />

IMPAIRMENT WORLDWIDE, ESPECIALLY IN DEVELOPING COUNTRIES<br />

ERIC PEARLMAN<br />

Director Of Institute For Immunology<br />

University of California, Irvine<br />

The World Health Organization estimates that 1.8 million people in developing<br />

nations are blinded annually from corneal ulcers; furthermore, in developing nations<br />

in Asia and Africa, up to 65% of total corneal ulcers are caused by fungal infection.<br />

In the USA and in industrialized nations, contact lenses are the major risk<br />

factor, and an ineffective contact lens care solution was the cause of an outbreak<br />

of Fusarium keratitis in 2005/2006.<br />

The vast majority of corneal infections are caused by filamentous molds of the<br />

Fusarium and Aspergillus species, which can also penetrate the vitreous and<br />

cause endophthalmitis.<br />

Current regimens for fungal keratitis are often ineffective, with up to 60% of<br />

fungal keratitis cases requiring corneal transplantation.<br />

Given that much of the disease manifestations leading to vision loss occur as a<br />

result of the host inflammatory response to fungal hyphae in the corneal stroma,<br />

therapies are directed not only at killing the pathogens, but also in curtailing the<br />

host immune response. Current and novel approaches to anti-fungal and anti-inflammatory<br />

therapy will be discussed.<br />

69


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 10<br />

FOCUS ON RETINITIS PIGMENTOSA (RP): TRANSLATABILITY OF SUCCESS<br />

IN ANIMAL MODELS OF ORPHAN AND GENETIC DISEASES OF THE EYE<br />

CLAIRE GELFMAN (ORA, INC), ANDY WHITLOCK (ORA, INC), MICHAEL YOUNG (RENEURON),<br />

ANTHONY VUGLER (UCL-INSTITUTE OF OPHTHALMOLOGY), SARA PATEL (RENEURON)<br />

Senior Director, Pre-Clinical and Translational Services at Ora Ophthalmic Research Associates<br />

A well-defined preclinical path forward into the clinic is every drug developer’s<br />

dream. The reality though is that even when a path has been carved, preclinical<br />

success does not always translate into success in the clinic.<br />

The issue is even more complex when the indication under consideration is a<br />

rare disease, when the disease pathology is less well-understood, the number of<br />

patients world-wide is not so prevalent, and funding is not as accessible for preclinical<br />

and clinical POC studies. Retinitis pigmentosa (RP) represents a rare ocular<br />

disorder with genetic origins. It is a heterogeneous group of diseases with<br />

a variety of mutations affecting photoreceptor function and can result in blindness.<br />

Common symptoms include difficulty with night vision as well as a loss of<br />

peripheral vision. While many animal models of RP are available resulting in a<br />

better understanding of the disease pathology, there is still no cure.<br />

This presentation will focus on animal models of RP and the types of therapeutic<br />

strategies that have been evaluated in terms of both safety and early efficacy.<br />

The information gleaned from such studies can be used to guide clinical decisions<br />

around dosing and patient population recruitment.<br />

A case study will be presented outlining recent data obtained using human retinal<br />

progenitor cells (hRPC) in rodent models of RP, and how that information was<br />

used to support a clinical study currently in progress.<br />

70


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 10<br />

PROGESTERONE ANALOGS AS NEUROPROTECTANTS IN ANIMAL MODELS<br />

OF RETINITIS PIGMENTOSA<br />

THOMAS G. COTTER 1 , SARAH ROCHE 1 , ASHLEIGH BYRNE1,<br />

ANI RUIZ-LOPEZ 1 VIOLETA GOMEZ 2 AND NICOLAS CUENCA 2<br />

1<br />

School of Biochemistry & Cell Biology, Biosciences Institute, University College Cork,<br />

Ireland and 2 University of Alicante Spain<br />

Purpose: Retinitis Pigmentosa (RP) is a condition where loss of both rod and subsequently<br />

cone photoreceptor cells leads to blindness. This research looks at the<br />

therapeutic potential of a progesterone analog (Norgestrel) as a very promising<br />

neuroprotectant molecule. Norgestrel is found in some forms of the contraceptive<br />

pill.<br />

Methods: The rd10 and light induced mouse models of RP were both used in this<br />

study with similar results. Methods used include, ergs, optomotor tests, immunohistochemistry<br />

and several other standard laboratory methods.<br />

Results: The progesterone analog Norgestrel preserves retinal morphology out to<br />

day 40 well beyond the peak of cell death at day 15 in the untreated rd10 animals.<br />

Visual acuity at day 40, in the treated animals, was remarkably the same as control<br />

C57 animals, ergs were also markedly improved.<br />

The neuroprotective properties of Norgestrel appear to operate through its effects<br />

on both photoreceptor and microglia cells. In the former it up-regulates key<br />

cell survival pathways and also induces the production of bFGF which acts as an<br />

autocrine survival factor for photoreceptors. In addition it also stimulates photoreceptors<br />

to produce and release fractalkine which prevents microglial cells from<br />

entering the photoreceptor layer. In untreated animals microglia are responsible<br />

for the destruction of photoreceptors and Norgestrel prevents this.<br />

Lastly, Norgestrel also acts directly on microglia to dampen the inflammatory<br />

phenotype of these cells.<br />

Conclusions: The progesterone analog Norgestrel is a strong neuroprotectant of<br />

photoreceptors in the rd10 and other animal models of RP.<br />

71


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 10<br />

NEUROPROTECTION IN INHERITED RETINAL DEGENERATIONS:<br />

ROLE OF ANTIOXIDANTS AND NEUROTROPHINS TO PRESERVE OR<br />

RESCUE CONE FUNCTION<br />

BENEDETTO FALSINI<br />

Institute of Ophthalmology, Fondazione Policlinico Gemelli,<br />

Universita Cattolica del S. Cuore, Rome, Italy<br />

Purpose: An important goal of neuroprotection in inherited retinal degenerations<br />

(IRD) is to preserve or rescue cone-mediated function, responsible of common<br />

daylight visual functions. Oxidative damage and loss of neurotrophic factors<br />

are major mechanisms that have been implicated in IRD-associated cone system<br />

dysfunction/degeneration. We report the results of pilot clinical trials on antioxidants<br />

and neurotrophic factors (performed at Fondazione Policlinico Gemelli,<br />

Universita Cattolica, Rome) aimed at preserving or rescuing cone function in<br />

IRDs.<br />

Methods: Single-center, IRB approved, Phase IIa clinical trials were performed to<br />

evaluate potential efficacy of 1. oral saffron antioxidant (20 mg/day, double-blind,<br />

placebo-controlled) on central cone function (acuity, focal macular electroretinogram,<br />

fERG) in ABCA4-related Stargardt’s macular dystrophy (STARSAF02,<br />

clinicaltrials.gov NCT01278277) 2. nerve growth factor (NGF) topical eye-drops<br />

(total dose of 1 mg in 5 ml of saline solution, open label, single-arm study) on<br />

central and peripheral cone function (acuity, fERG, Goldman visual field) in advanced<br />

RP patients (RP01, EudraCT n. 2008-004561-26).<br />

Results: 1. STARSAF02: fERG stabilization was found after 6 months of saffron<br />

but not placebo administration; no change in visual acuity was observed, 2. RP01:<br />

fERG amplitude improvements (above the 95% limits of test-retest variability)<br />

were found 30 days after NGF treatment in 3/9 patients. These changes were<br />

associated with Goldman isopter V/4e size improvements (10-40 degrees on the<br />

major axis), no change in acuity but improved subjective vision.<br />

Conclusions: Antioxidants and NGF show promise in the clinic as potential therapeutic<br />

strategies to preserve or rescue cone-mediated function in selected subtypes<br />

or stages of IRDs.<br />

72


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 10<br />

THE METABOLIC AND REDOX SIGNALING CONTROLLED BY<br />

THE ROD - DERIVED CONE VIABILITY GENE NXNL1<br />

THIERRY LÉVEILLARD 1 EMMANUELLE CLÉRIN 1 , NAJATE AÏT-ALI 1 , YING YANG 1 , GÉRALDINE<br />

MILLET-PUEL 1 , ERIKA T. CAMACHO 2 , DENIZ DALKARA 1 , ALAIN VAN DORSSELAER 3 , JOSÉ-ALAIN SAHEL 1<br />

1<br />

INSERM, U968, Paris, F-75012, France; UPMC Univ Paris 06, UMR_S 968, Institut de la Vision,<br />

Paris, F-75012, France; 3 CNRS, UMR_7210, Paris, F-75012, France<br />

2<br />

School of Mathematical & Natural Sciences, Arizona State University, USA<br />

3<br />

BioOrganic Mass Spectrometry Laboratory (LSMBO), IPHC, Université de Strasbourg,<br />

67087 Strasbourg, France<br />

Retinitis pigmentosa is an inherited retinal degeneration that processes from the<br />

death of rods followed by dysfunction and degeneration of cones. The nucleoredoxin-like<br />

1 gene (NXNL1) encodes by alternative splicing for the trophic factor<br />

RdCVF that stimulates aerobic glycolysis in cones by interaction with its cell surface<br />

receptor basigin-1 that is linked to the glucose transporter GLUT1. RdCVF<br />

accelerates glucose uptake by cones to sustain cone outer segment renewal. Rd-<br />

CVF prevents secondary cone degeneration in recessive and dominant animal<br />

models of retinitis pigmentosa. The second product of the NXNL1 gene, the thioredoxin<br />

RdCVFL protects the cones against hyperoxia. The administration of<br />

RdCVF or RdCVFL prevents visual loss in the rd10 mouse, a mouse model of recessive<br />

retinitis pigmentosa, using a non cell- and a cell-autonomous mechanism<br />

respectively. In order to translate this promising therapy towards the clinic, we<br />

have evaluated the therapeutic benefit of delivering both products of the NXNL1<br />

gene by subretinal injection with an AAV vector targeting retinal pigmented epithelial<br />

cells and cones.<br />

We measured the visual acuity of the rd10 mice, using optometry after subretinal<br />

injection of an AAV serotype 2 encoding for RdCVF and RdCVFL as compared to<br />

AAV2-RdCVF, AAV2-GFP and sham controls. The kinetics of the loss of visual<br />

acuity was statistically significantly retarded after injection of AAV-RdCVF-Rd-<br />

CVFL and very significantly considering the medical objective.<br />

Our results demonstrate that this metabolic and redox treatment will likely be<br />

successful in preserving central vision in patients suffering of retinitis pigmentosa<br />

independently of causative mutations.<br />

73


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 10<br />

DEVELOPMENT OF INVESTIGATIONAL GENE THERAPY FOR RPE65-MEDIATED<br />

INHERITED RETINAL DISEASE<br />

DANIEL CHUNG<br />

Clinical Ophthalmic Lead, Spark Therapeutics, Inc. Philadelphia<br />

Purpose: Several early-phase human trials provided preliminary evidence of the potential<br />

safety and efficacy of adeno-associated virus-mediated human RPE65 augmentation<br />

for RPE65-mutation-associated inherited retinal dystrophies. We report the<br />

latest results from a Phase 3, open-label, randomized, controlled trial that concluded<br />

in 2015 at Children’s Hospital of Philadelphia and the University of Iowa evaluating<br />

the safety and efficacy of AAV2-hRPE65v2 (SPK-RPE65) to treat RPE65-mediated inherited<br />

retinal dystrophies (NCT00999609).<br />

Methods: Thirty-one subjects with disease-causing biallelic RPE65 mutations were<br />

randomized 2:1 to intervention or control. Eligibility criteria included age ≥3 yearsold;<br />

bilateral visual acuity worse than 20/60 and/or visual field less than 20 degrees<br />

in any meridian; evidence of sufficient viable retinal cells by fundus photography<br />

and optical coherence tomography; ability to be evaluated on mobility testing; and<br />

willingness to provide consent or parental permission and assent, where appropriate.<br />

Subjects in the intervention group received subretinal injections of AAV2-hRPE65v2<br />

sequentially to each eye within an 18-day window. Control subjects did not receive<br />

AAV2-hRPE65v2 for at least 1 year from baseline, but completed the same testing regiment<br />

as those in the intervention arm. Using a standardized subretinal delivery procedure<br />

and under general anesthesia, 1.5E11 vector genomes/eye were delivered in a total<br />

volume of 300 µl. Standardized mobility testing under different luminance conditions<br />

was the primary efficacy endpoint, with secondary endpoints including full field light<br />

sensitivity testing, assigned first eye mobility change score and visual acuity.<br />

Results: All subjects completed Year 1 follow-up testing. Phase 3 study results include<br />

demographics, safety information, and mobility testing change score (performance at<br />

1 year compared with baseline), and secondary endpoints of full field tight sensitivity<br />

testing, assigned first eye mobility change score and visual acuity. A separate study<br />

analyzing mobility test data in untreated normal and retinal dystrophy cohorts was<br />

used to validate the mobility test’s ability to distinguish low vision from normal-sighted<br />

populations, differentiate a range of performance in low vision subjects, and confirm<br />

changes in functional vision over time. The trial of 31 subjects met with statistical<br />

significance its primary endpoint, the bilateral mobility test change score (p =<br />

0.001), as well as the first two of three secondary endpoints, specifically full-field light<br />

sensitivity threshold testing, or FST (p < 0.001), and the assigned first eye mobility<br />

test change score (p = 0.001). Statistical significance was not achieved for the third<br />

secondary endpoint, visual acuity (p = 0.17). No serious adverse events (SAEs) and no<br />

deleterious immune responses related to SPK-RPE65 were reported.<br />

Conclusions: Results of this study, the first Phase 3 gene therapy study completed for<br />

a retinal dystrophy, provides additional evidence regarding the potential efficacy and<br />

safety of gene therapy intervention by surgical subretinal administration of AAV2-<br />

hRPE65v2 (SPK-RPE65) as measured by the primary endpoint of mobility testing, and<br />

2 of the 3 secondary endpoints.<br />

74


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 11<br />

REGIONAL DIFFERENCES IN BLOOD FLOW AS THE BASIS FOR<br />

UNDERSTANDING RETINAL VASCULAR DISEASE<br />

TOKE BEK<br />

Toke Bek. Department of Ophthalmology, Aarhus University Hospital,<br />

DK-8000 Aarhus C, DENMARK<br />

The neurosensory structure of the retina shows distinct regional variations<br />

with derived effects on vascular structure. The foveal area which consists of the<br />

photoreceptor layer only, is nourished entirely from the choriocapillaris. The<br />

extrafoveal retina extending to the vascular arcades and thereby delimiting the<br />

macular area has a rich content of metabolically active cells and therefore needs<br />

three capillary layers. These layers are supplemented with a fourth capillary layer<br />

supplying the retinal nerve fiber layer in the arcuate areas where this layer is<br />

thick, peripheral from which the vascular density decreases towards the retinal<br />

periphery.<br />

The branching pattern of retinal vessels is also regionally varying with a dichotomous<br />

branching pattern in the macular area and small vessels leaving at right<br />

angle to larger irradiating vessels in the retinal midperipery.<br />

It is likely that the regional distribution of retinal vascular lesions reflects regional<br />

variations in vascular structure. Therefore, the distribution of retinal vascular<br />

lesions is a gate for understanding the pathophysiology of retinal vascular disease.<br />

Examples of the information value in regional variations in vascular lesions will<br />

be presented and suggestions for future studies of vascular function aimed at elucidating<br />

the pathophysiology of retinal vascular disease will be proposed.<br />

75


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 11<br />

NOVEL TREATMENT FOR DIABETIC RETINOPATHY BY<br />

DRUG REPOSITIONING<br />

TAIJI NAGAOKA<br />

Novel Treatment for Diabetic Retinopathy by Drug Repositioning<br />

Although diabetic retinopathy is a leading cause of blindness in Western countries,<br />

the causes of its vascular and visual pathologies are not fully understood. We<br />

have reported that the RBF may decrease in type 2 diabetics before development<br />

of retinopathy or mild retinopathy (IOVS, 2010), suggesting that the improvement<br />

of the impaired retinal blood flow may be a target for a novel treatment of<br />

diabetic retinopathy. We have examined whether any drugs that have been generally<br />

used for systemic disorders (i.e., diabetes, hyperlipidemia, hypertension)<br />

may be repositioned to treat diabetic retinopathy.<br />

For this purpose, porcine retinal arterioles (50 µm to 100 µm in diameter) were<br />

isolated and pressurized without flow for in vitro study.<br />

We found that some of these drugs can dilate retinal arterioles, which may contribute<br />

to improve retinal blood flow in patients with diabetes. My talk will focus<br />

on the possibility that “drug repositioning” may be a novel strategy for the treatment<br />

of diabetic retinopathy.<br />

76


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 11<br />

RETINAL AND CHOROIDAL VASCULAR RESPONSES TO ELECTRICAL BRAIN<br />

STEM STIMULATION IN RATS<br />

CLEMENS STROHMAIER 1,2 , KAROLINA MOTLOCH 1 , CHRISTIAN RUNGE 1 ,<br />

JEFFREY W. KIEL 2 , HERBERT A. REITSAMER 1<br />

1<br />

Ophthalmology, Paracelsus Medical University, SALK, Müllner Hauptsrasse 48,<br />

5020 Salzburg, Austria, 2 Ophthalmology, UTHSCSA, 7703 Floyd Curl Drive, 78229 San Antonio, TX<br />

Purpose: Electrical brain stem stimulation at the coordinates of the nucleus salivatorius<br />

superior (SSN) is known to increase choroidal blood flow, but not retinal<br />

blood flow. The present study investigates the retinal and choroidal vascular responses<br />

to SSN stimulation. Furthermore, data on possible neurotransmitters is<br />

presented.<br />

Methods: Sprague Dawley rats (n= 17) were anesthetized using pentobarbital<br />

sodium and paralyzed with gallamine triethiodide. Choroidal blood flow was<br />

measured using Laser Doppler flowmetry. Retinal vessel diameters were measurement<br />

with a fundus camera customized for rats. Stimulations at the SSN coordinates<br />

were performed at 20Hz, 9 µA, 1 ms pulse duration and 200 pulses. After<br />

baseline measurements with subsequent SSN stimulations, L-NAME (10 mg/<br />

kg) was applied intravenously and the stimulation protocol was repeated.<br />

Results: Stimulation at the SSN coordinates increased choroidal blood flow from<br />

248.17 ± 46.92 arbitrary units (a.u.) to 347.30 ± 60.44 a.u. (p ≤ 0.05). Stimulation<br />

at the SSN coordinates increased the retinal arterial diameter by 6.41 ± 1.65 % and<br />

the venous diameter by 3.48 ± 1.93 % (both p < 0.05). L-NAME application reduced<br />

the arterial response significantly to 2.93 ± 0.91 %.<br />

Conclusion: Electrical stimulation at the SSN coordinates yielded a significant increase<br />

in choroidal blood flow and induced retinal vasodilation, the application of<br />

L-NAME did not block the stimulation effect and thus indicates that NO is not the<br />

sole neurotransmitter.<br />

77


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 11<br />

RETINAL OXYGEN EXTRACTION IN DIABETES AND GLAUCOMA<br />

DOREEN SCHMIDL 1 , LEOPOLD SCHMETTERER 1,2,3,4 ,RENE WERKMEISTER 2 ,<br />

KLEMENS FONDI 1 , AHMED BATA 1 , GERHARD GARHÖFER 1<br />

1<br />

Department of Clinical Pharmacology, Medical University of Vienna, 2 Center of Medical Physics<br />

and Biomedical Engineering, Medical University of Vienna, 3 Singapore Eye Research Institute,<br />

4<br />

Nanyang Technological University<br />

Purpose: We have recently presented a method to measure oxygen extraction of<br />

the human retina. This technique combines measurement of total retinal blood<br />

flow using Doppler Optical Coherence Tomography (OCT) with measurement<br />

of oxygen saturation using spectroscopic reflectometry. In the present studies we<br />

investigated whether retinal oxygen extraction is altered in early diabetes and<br />

glaucoma.<br />

Methods: A total of 24 subjects with type 1 diabetes without diabetic retinopathy<br />

and 40 patients with primary open angle glaucoma (POAG) were included<br />

in these studies. In addition, we included a total of 64 healthy subjects who were<br />

age- and sex-matched to the patient groups. Retinal blood flow was measured by<br />

bi-directional Doppler OCT. Oxygen saturation was measured using reflectometry<br />

and oxygen extraction was calculated.<br />

Results: In patients with diabetes we observed increased retinal blood flow and<br />

decreased retinal oxygen extraction as compared to healthy subjects (p < 0.05<br />

each). Retinal nerve fiber layer thickness and multifocal electroretinography parameters<br />

were not different between the two groups. In patients with POAG we<br />

observed decreased retinal blood flow and decreased oxygen extraction (p < 0.05<br />

each). The decrease in oxygen extraction was correlated with visual field defect<br />

(p < 0.01).<br />

Conclusions: In type 1 diabetes we observed increased retinal blood flow and<br />

decreased retinal oxygen extraction at an early stage of the disease at which no<br />

changes in retinal function or retinal nerve fiber layer thickness could be detected.<br />

In POAG a reduction in retinal blood flow and retinal oxygen extraction was<br />

observed that correlated with the amount of visual field defect. Whether this is a<br />

cause or a consequence of the disease is unknown.<br />

78


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 12<br />

CAN DRUG DELIVERY HELP SOLVE THE PROBLEM OF MYOPIA?<br />

HEATHER SHEARDOWN<br />

McMaster University, Department of Chemical Engineering<br />

Natural barriers, including rapid tear turnover, a highly impermeable epithelial<br />

layer and isolation of delicate tissues, significantly limit the amount of drug that<br />

can be delivered to the eye.<br />

There is a need for alternative delivery methods to enhance therapies which<br />

have the potential to treat of host of ocular conditions including myopia. An understanding<br />

of the mechanisms used to control the release of the pharmacological<br />

active as well as the novel methods to enhance residence time will be discussed<br />

with a focus on the novel delivery systems developed for anterior segment<br />

drug delivery and degradable systems which have the potential to be used in the<br />

posterior eye.<br />

79


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 12<br />

EFFICACY OF ATROPINE FOR PROGRESSIVE MYOPIA IN EUROPEANS: TWO<br />

YEAR RESULTS AND COMPARISON WITH RESULTS FROM EAST ASIA<br />

JAN ROELOF POLLING 1-3 , ASTRID VAN DER SCHANS 1 , J. WILLEM L. TIDEMAN 1,3 ,<br />

CAROLINE C.W. KLAVER 1,3<br />

1<br />

Department of Ophthalmology, Erasmus MC, university medical center Rotterdam, the Netherlands;<br />

2 Department of Optometry & Orthoptics, Faculty of Health, University of Applied Sciences,<br />

Utrecht, the Netherlands; 3 Department of Epidemiology, Erasmus MC, university medical center<br />

Rotterdam, the Netherlands<br />

Purpose: Randomized controlled trials have shown the efficacy of atropine for<br />

progressive myopia, and this treatment has become the preferred practice pattern<br />

for this condition in many Asian countries. This study explores the two<br />

year effectiveness of atropine 0.5% treatment for progressive high myopia and<br />

adherence to therapy in a non-Asian country compared with studies from Asian<br />

countries.<br />

Methods: We performed an effectiveness study of atropine eye drops for progressive<br />

myopia in Rotterdam, the Netherlands. We included 205 children (mean age<br />

9.8 yrs. ± 3.3) of European (n=138; 67.3%), Asian (n=51; 24.9%) and African (n=16;<br />

7.8%) descent, performed a standardized eye examination including cycloplegic<br />

refraction and axial length at baseline, prescribed atropine eye drops 0.5% daily,<br />

and examined the children every 6 months at follow up. For the comparison with<br />

our data randomized controlled trials in Asian cohorts comparing atropine high<br />

dose (0.5-1.0%) with placebo were searched in PubMed. Primary outcome was<br />

progression of myopia in a 1 and 2 year period under atropine high dose regime.<br />

Results: Mean spherical equivalent (SE) at baseline was -6.15D (±3.59); mean annual<br />

progression before treatment -1.0D/yr; and the proportion of high myopes<br />

(≤-6.0D) was 40,5%. Median follow up was 23,6 months. The mean progression<br />

of SE diminished substantially during the first year -0.24D ± 1.1 and the second<br />

year -0.51D ±0.64. We included 4 Asian studies in our analysis who had a mean<br />

difference of -0.55D (CI 0.46-0.64) in the 2 year treatment period.<br />

Conclusion: Our study confirms that in every day patients in the western world<br />

myopia progression can be halted by atropine 0.5%. Racial differences in response<br />

to treatment could not be detected in our study neither by literature comparison.<br />

80


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 12<br />

ORTHOKERATOLOGY COMBINED WITH LONG-TERMINSTILLATIONS OF VERY<br />

SMALL ATROPINE CONCENTRATIONS: A PRE-EVALUATION OF<br />

THE STABILIZING EFFECT<br />

ELENA TARUTTA, TATYANA YU. VERZHANSKAYA<br />

Helmholtz Research Institute of Eye Diseases, Moscow<br />

Orthokeratology lenses (OKL) and long-term atropine instillations are currently<br />

considered the most effective methods of inhibiting progressive myopia.<br />

Purpose: First evaluation of effectiveness and safety of myopia inhibition by combining<br />

OKL with instillations of atropine microdoses.<br />

Material and methods: 13 children aged 7 to 11.5 with acquired low (8 eyes), moderate<br />

(8) and high myopia (10), found to have progressive myopia after a 1-2-year<br />

OKL usage (6-zone DL-ESA), daily received two drops of 0.01% atropine 3 hours<br />

before wearing OKL. Beside standard examinations, we measured axial length<br />

(AL, IOL–Master, Zeiss) relative accommodation reserves (RAR), objective accommodative<br />

response (OAR, Grand Seiko WR5100K) before adding atropine and<br />

6 months after it. Progression rate was assessed by AL increase (mm/year), also<br />

recalculated in diopters/year.<br />

Results: Before atropine instillations, average progression rate was 0.68 D/year<br />

(0.22 mm/year): respectively, 0.74, 0.8 and 0.49 D/year (0.24, 0.26 and 0.16 mm/<br />

year) for low, moderate and high myopia. 6 months after, the figures were respectively<br />

0.54, 0.69, 0.33 and 0.63 D/year (0.18, 0.23, 0.11, 0.21 mm/year). As<br />

observations were few, all differences are statistically insignificant. Moderate<br />

myopia showed the best inhibiting trend. Contrariwise, high myopia tended to<br />

progress faster with atropine. RAR were high: 4.0 D before atropine and -4.0 D<br />

6 months later, which is related to pseudoaccommodation due to aberration increase<br />

in OKL users. OAR decreased slightly (from -2.7 to -2.5 D).<br />

Conclusion: Although pre-evaluation cannot absolutely confirm the effectiveness<br />

of long-term atropine support of OKL, the observed results are positive and<br />

validate further research.<br />

81


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 12<br />

DESIGN, SYNTHESIS, AND CHARACTERIZATION OF A SELECTIVE INHIBITOR<br />

FOR RETINALDEHYDE DEHYDROGENASE (ALDH1A) ENZYMES<br />

ANGELICA HARPER 1 , ANH LE 2 , TIM MATHER 3 , ANTHONY BURGETT 2 , JODY A. SUMMERS 1<br />

1<br />

Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK,<br />

United States of America; 2 Department of Chemistry and Biochemistry, University of Oklahoma,<br />

Norman, OK, United States of America; 3 Oklahoma Medical Research Foundation, Oklahoma City,<br />

OK, United States of America<br />

Purpose: Retinaldehyde dehydrogenase 2 (RALDH2) has been identified as a potential<br />

therapeutic target for the control of postnatal ocular growth.<br />

The objective in this study was to use an intelligent-drug design approach to develop<br />

a RALDH2-selective inhibitor to further examine the role of RALDH2 in<br />

myopia.<br />

Methods: MoleGro software was used to dock the structure of dichloro-all-trans-retinone<br />

(DAR) into models of chick RALDH2 and human<br />

ALDH2. DAR was synthesized by a modified dihalomethyllithium approach.<br />

Selectivity and mechanism of inhibition was determined in vitro using NADH<br />

assays with recombinant RALDH2. The effect of DAR on retinoic acid (RA) synthesis<br />

was determined in 1) cells overexpressing RALDH2, 2) choroidal cell lysates,<br />

and 3) choroid tissue by an in vitro RA synthesis assay. Toxicity on scleral<br />

tissue was measured with a proteoglycan synthesis assay.<br />

Results: Docking suggested selectivity of DAR to RALDH2 compared to hu-AL-<br />

DH2 (MolDock score: -71.92±6.83 vs. 14.41±17.98). In vitro assays indicated that<br />

DAR inhibits RALDH2 in an irreversible and dose dependent manner (IC50=52.2<br />

nM, 20 min pre-incubation), with no significant inhibitory effect on hu-ALDH2.<br />

DAR successfully inhibited RA synthesis in 1) cells overexpressing RALDH2<br />

(p


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 12<br />

MEDICATION CROSSLINKING OF THE SCLERA:<br />

AN EXPERIMENTAL IMPLEMENTATION OF A TECHNOLOGY OF<br />

SCLERA STRENGTHENING TREATMENT OF MYOPIA<br />

ELENA IOMDINA 1 , ELENA TARUTTA 1 , ALISA SIANOSYAN 1 , INNA KHOROSHILOVA-MASLOVA 1 ,<br />

ALEXANDER KORIGODSKY 2 , IVAN ZAKHAROV 2 , NATALYA IGNATIEVA 3<br />

1<br />

Helmholtz Research Institute of Eye Diseases, Moscow, 2 OOO HiBiTech, Moscow, 3 Lomonosov<br />

Moscow State University, Russia<br />

Purpose: Experimental implementation of scleral collagen crosslinking by<br />

sub-Tenon’s capsule injections of a biologically active composition, Scleratex, in<br />

the equatorial and posterior pole areas of the eye.<br />

Material and Methods: A placebo-controlled study into the safety and effectiveness<br />

of sub-Tenon’s capsule injections of Scleratex (a solution of the basic amino<br />

acid salts in the form of succinates) was performed on 47 Chinchilla rabbits (94<br />

eyes). 0.1 ml of Scleratex or placebo solution was injected once a week under the<br />

Tenon’s capsule of the experimental and the fellow eyes, respectively. The first<br />

series (4 injections) lasted 1 month, and the second series (12 injections) took 3<br />

months. After the course of injections, all structures of 22 enucleated eyes, including<br />

retina, were studied morphologically using light microscopy, while<br />

scleral samples from the remaining 72 eyes were used to determine the elasticity<br />

modulus (on the testing machine Autograph AGS-H, SHIMADZU, Japan) and the<br />

level of collagen crosslinking (by denaturation temperature Td using differential<br />

scanning calorimetry on the calorimeter, Phoenix DSC 204, Netzsch, Germany).<br />

Results: The weekly injections of Scleratex performed for 1 month or 3 months<br />

showed no clinical or morphological signs of local irritation, damage, or toxicity.<br />

A 15 to 20% increase in collagen crosslinking and a 1.8-fold increase in the elasticity<br />

modulus of the sclera with respect to the fellow eye were detected. This increase<br />

was accompanied by an increase in the number of cells, formation of new<br />

connective tissue on the scleral surface, and appearance of additional vessels. In<br />

total, this is an evidence of an effective trophic and sclera strengthening influence<br />

of Scleratex.<br />

Conclusion: The outcome of experimental implementation of a minimally invasive<br />

technology for scleral collagen crosslinking shows it to be a plausible method<br />

of scleral strengthening and antidystrophic treatment of progressive myopia.<br />

83


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 13<br />

CORNEAL GENE THERAPY: BEYOND VIRAL VECTORS<br />

ALEXANDER V. LJUBIMOV<br />

Eye <strong>Program</strong>, Board of Governors Regenerative Medicine Institute and<br />

Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, USA<br />

Purpose: Cornea is ideal for gene therapy with easy accessibility, immune privilege,<br />

easy transgene expression monitoring, and topical drug application. Viral<br />

vehicles (adeno-associated virus, adenovirus, herpes simplex virus type 1, and<br />

lentivirus) allowing for lasting effects, high efficiency and time-controlled action<br />

have been used to successfully transfect corneal cells.<br />

However, as viruses may induce immune reactions, uncontrolled integration<br />

into the host genome, and are toxic for stem cells, new and safer non-viral vectors<br />

are being developed, with more focus on nanoparticles (NP).<br />

They are fairly easy to synthesize with low costs, can accommodate large vectors,<br />

are mostly non-inflammatory, do not cause genomic modifications, and are<br />

amenable to cell targeting. NP including poly (lactide-co-glycolide) NP loaded<br />

with antifibrotic pirfenidone, inorganically-coated all-trans retinoic acid NP,<br />

elastin-like polypeptide-based NP bearing a mitogenic protein lacritin, gold NP<br />

with BMP7 gene, cationic NP with TGF-β and CTGF siRNAs, polymeric micelles<br />

with bcl-xL gene were used to promote corneal wound healing, reduce stromal<br />

haze after photorefractive keratectomy, and cell apoptosis. NP with shRNA to<br />

VEGF-A inhibited corneal neovascularization upon alkaline burns.<br />

Methods: For corneal gene therapy we have used new nanobioconjugates (NBC)<br />

based on polymalic acid that do not have NP drawbacks: passive cellular uptake<br />

and cargo leakage leading to side effects. Diabetic corneas have wound healing<br />

alterations and impaired corneal epithelial stem cell (CESC) functions. AV gene<br />

therapy proved to be rather toxic for cultured CESC, prompting the use of nano<br />

vehicles.<br />

Results: Non-toxic NBC were engineered to increase the expression of diabetes-downregulated<br />

c-met gene and decrease diabetes-upregulated MMP-10 and<br />

cathepsin F genes. Cell-targeted NBC increased diabetes-impaired CESC wound<br />

healing and the expression of diabetes-suppressed stem cell markers.<br />

Conclusions: NBC and NP allowing customizable use of various drugs provide<br />

promising versatile nano vehicles for next-generation preclinical and clinical applications<br />

of corneal gene therapy.<br />

84


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 13<br />

SEVERE OCULAR ALLERGIES: FROM PATHOPHYSIOLOGY TO<br />

FUTURE THERAPIES<br />

ANDREA LEONARDI<br />

Department of Neuroscience, Ophthalmology Unit, University of Padova, Italy<br />

Allergic conjunctivitis is often considered an easy-to treat and self limiting allergic<br />

inflammation of the conjunctiva without long term complications and potential<br />

damage for the visual function. However, ocular allergic (OA) includes<br />

a variety of inflammatory diseases of the ocular surface affecting lids, cornea,<br />

lachrymal gland and tear film, at different levels of severity. The inflammatory<br />

mechanism of seasonal (SAC) or occasional allergic conjunctivitis is typically<br />

type I hypersensitivity IgE-mediated, whereas in chronic allergic disorders, such<br />

as vernal keratoconjunctivitis (VKC) or atopic keratoconjunctivitis (AKC), the<br />

mechanisms are more complex and probably involve both IgE and T cell-mediated<br />

responses. Nevertheless, acute and chronic diseases have in common: 1)<br />

the possible sensitization to environmental allergens; 2) the IgE-mast cell activation<br />

with subsequent mediator cascade; 3) the conjunctival inflammation with a<br />

prevalence of eosinophils; 4) the presence of lymphocytes with a Th2, Th9 and<br />

Th17 profiles of cytokine production; 5) a mucosal hyper-reactivity. Corneal involvement<br />

is common in VKC and AKC associated with neuro-inflammation, tissue<br />

remodelling and fibrosis, resulting in potential corneal damage and scarring.<br />

Therefore, multiple mediators, cytokines, chemokines, growth factors, proteases<br />

and enzymes are over-expressed in severe OA.<br />

Interestingly, transcriptomic, proteomic and gycomic techniques, reveal the<br />

presence of low abundant and high abundant proteins and glycoproteins with<br />

pro- and anti-inflammatory properties, which may represent either disease biomarker<br />

or target for new treatments. Understanding inflammation in ocular allergy<br />

may provide indication for a rational treatment of these diseases and future<br />

potential therapeutic approaches including immune-modulators, such as cyclosporine<br />

(CsA) and tacrolimus.<br />

In particular, CsA can be considered for treatment of moderate to severe VKC<br />

and AKC. It decreases signs and symptoms, and the need for steroids. Corneal<br />

complications should be carefully monitored and anti-inflammatory therapy adjusted;<br />

in these cases, steroids must be used since the pathogenesis of the ulcer<br />

is strictly immune-mediate. Corticosteroids are preferred over CsA, since they<br />

are more effective in inhibiting the inflammatory component of corneal damage<br />

(i.e., eosinophil- and neutrophil-liberated epithelial toxic mediators). If a systemic<br />

hypersensitivity to identified allergens exists, specific immunotherapy may be<br />

considered. The indications for of anti-IgE therapies are still unclear.<br />

85


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 13<br />

GABAPENTIN EYE DROPS FOR THE TREATMENT OF OPHTHALMIC<br />

PAIN AND OCULAR SURFACE INFLAMMATION<br />

DARIO RUSCIANO 1 , CLAUDIO BUCOLO 2 , GABRIELLA LUPO 2 , DANIELA C. ANFUSO 2 ,<br />

MELANIA OLIVIERI 1 , MARTINA CRISTALDI 1 , SALVATORE PEZZINO 3 , FILIPPO DRAGO 2<br />

1<br />

Sooft Italia, Catania, Italy; 2 Department of Biomedical and Biotechnology Sciences,<br />

University of Catania, Italy; 3 Bioos srl, Catania, Italy<br />

Purpose: Gabapentin is a synthetic molecule that targets voltage-dependent calcium<br />

channels and purinergic adenosine A1 receptors, thus eliciting analgesic,<br />

anti-convulsive and antinflammatory responses. To date, gabapentin has been<br />

successfully used for the treatment of neuropathic pain and epileptic convulsions.<br />

However, in most of the published studies systemic gabapentin was not adequate<br />

to control corneal neuropathic pain. Therefore, aim of this study has been to<br />

study the efficacy of gabapentin eye drops to control corneal pain and ocular surface<br />

inflammation after topical instillation.<br />

Methods: The effect of gabapentin eye drops was investigated on the inflammatory<br />

response of lipopolysaccharide (LPS)‐stimulated rabbit corneal cells (SIRC) and<br />

on endotoxin-induced uveitis (EIU) in rats. Further, a rat model of corneal pain<br />

was carried out using formaldehyde as insult.<br />

Results: A topical formulation of gabapentin was capable of reducing corneal pain<br />

and attenuate the ocular inflammatory reaction both in vitro and in vivo.<br />

Topical treatment with gabapentin significantly (p


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 13<br />

ALTERED ELECTRICAL ACTIVITY OF CORNEAL SENSORY RECEPTOR FIBERS<br />

DURING REGENERATION AFTER CORNEAL MICROKERATOME<br />

LESION IN THE GUINEA-PIG<br />

JUANA GALLAR, CAROLINA LUNA, SUSANA QUIRCE, LAURA RINCÓN-FRUTOS,<br />

CARLOS BELMONTE, M. CARMEN ACOSTA<br />

Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain<br />

Purpose: To characterize neural activity of corneal sensory receptors in the guinea-pig<br />

cornea at different times after corneal surgical lesion.<br />

Methods: Electrical activity of corneal sensory receptor fibers was recorded 1-60<br />

days after performing a mid-stromal surgical lesion (4mm-diameter incomplete<br />

circular flap) with a custom-made microkeratome in anesthetized guinea-pigs<br />

of both sexes. Nerve terminal impulse activity was recorded in vitro from the<br />

excised cornea and from single ciliary nerve fibers. Responses to thermal stimulation<br />

(changing bath temperature from 34ºC -basal temperature- to 20ºC -cooling<br />

ramp- or 50ºC -heating ramp-), mechanical (von Frey hairs) and chemical<br />

stimulation (30s-duration gas jets of 98% CO2 in air) were analyzed in intact and<br />

lesioned eyes.<br />

Results: Except peripheral to the lesion or at the flap hinge, no activity was recorded<br />

within the lesion area, suggesting postsurgical functional denervation.<br />

1-3 days after microkeratome-lesion, responses of polymodal nociceptors to chemical<br />

and heat stimulation were transiently increased and mechanical threshold<br />

decreased. Spontaneous activity and mechanical threshold of mechanonociceptors<br />

were not significantly modified after surgery.<br />

Ongoing activity at basal temperature and response to cooling ramps of cold thermoreceptors<br />

were transiently increased 7-14 days after lesion and tend control<br />

values afterwards.<br />

Conclusions: Corneal nerve fibers regenerating in lesioned corneas maintain<br />

their electrophysiological characteristics and responses to natural stimuli, albeit<br />

firing at higher frequencies the first days after injury.<br />

This may be due to sensitization induced by inflammatory mediators and/or to<br />

the well documented altered expression of sodium and potassium channels produced<br />

after nerve lesion.<br />

87


OCULAR THERAPEUTICS: VISION OF HOPE IN A CHANGING WORLD SESSION 13<br />

UNIQUE HYDROGEL TECHNOLOGY - IN VITRO MODEL<br />

REPRESENTING CORNEAL LAYERS<br />

AGNĖ ŽINIAUSKAITĖ 1 •<br />

, VYTAUTAS CEPLA 2 , RAMŪNAS VALIOKAS 3 , GIEDRIUS KALESNYKAS 1 , AND<br />

JENNI J. HAKKARAINEN 1<br />

1<br />

Experimentica Ltd., Kuopio, Finland<br />

2<br />

UAB Ferentis, Vilnius, Lithuania<br />

3<br />

Department of Nanoengineering, Center for Physical Sciences and Technology, Vilnius, Lithuania<br />

Purpose: The cornea is an effective penetration barrier to drugs applied topically<br />

onto the eye. In early drug development, it is important to evaluate the potency of<br />

a drug candidate for its ability to permeate through the cornea. The purpose of this<br />

study was to develop an in vitro model representing the corneal component layers<br />

(epithelium and stroma) using a unique hydrogel technology and human corneal<br />

epithelial cells (HCE-T).<br />

Methods: Different hydrogel components and cross-linking techniques were used.<br />

The apparent permeability coefficient (Papp) values of low and high permeability<br />

marker molecules across the blank hydrogels and hydrogels with HCE-T cells on<br />

top of hydrogels were measured. Expression and localization of tight junction proteins,<br />

ZO-1 and occludin, were assessed using immunocytochemistry.<br />

Results: Papp values were significantly lower for hydrogels with HCE-T cells cultured<br />

on top of the hydrogel than the Papp values for blank hydrogels. However,<br />

there were differences in the expression and localization of tight junction proteins<br />

depending on the hydrogel type where the cells were grown.<br />

Conclusions: The use of hydrogel technology is a promising model for the corneal<br />

stroma. However, additional development is needed to obtain an in vitro model<br />

comprising all functional corneal layers and possessing adequate epithelial barrier<br />

function.<br />

88


POSTER<br />

SESSION


POSTER BOARD<br />

TITLE AND AUTHOR<br />

1 IN VIVO COMPARISON OF THE RESIDENCE TIME OF CROSS-LINKED COMPARED<br />

TO LINEAR HYALURONIC ACID IN RABBIT EYE<br />

MIRKO MUZZI 1 ; RITA MENCUCCI 2<br />

1<br />

Department of Health Sciences, Section of Clinical Pharmacology and Oncology,<br />

University of Florence, Florence, Italy; 2 Ophthalmology Unit, Careggi Hospital, Florence, Italy<br />

2 PLACENTA GROWTH FACTOR PLAYS A ROLE IN IMMUNE RESPONSE<br />

ASSOCIATED WITH CHOROIDAL NEOVASCULARIZATION<br />

SERGIO CRESPO-GARCIA 1 , CAITLIN CORKHILL 1 , CHRISTOPHE ROUBEIX 1,2 ,<br />

NOR BERT KOCIOK 1 , OLAF STRAUSS 1 , ANTONIA M. JOUSSEN 1 , NADINE REICHHART 1<br />

1<br />

Department of Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany<br />

2<br />

Einstein Foundation, Berlin, Germany<br />

3 AUTOREGULATION OF RETINALGANGLIONCELLFUNCTION TO<br />

METABOLICCHALLENGE IN GLAUCOMA<br />

GIOVANNI LUCA ROMANO 1 , CHOU TSUNG HANG 2 ,CLAUDIO BUCOLO 1<br />

FILIPPO DRAGO 1 , VITTORIO PORCIATTI 2<br />

1<br />

Department of BiomedicalBiotechnologicalSciences (BIOMETEC), University of Catania<br />

2<br />

Bascom Palmer EyeInstitute,University of Miami, Miller School of Medicine<br />

Miami, FL, UnitedStates<br />

4 CANNABINOIDS IN OCULAR PATHOPHYSIOLOGY<br />

ANNA-MARIA SZCZESNIAK, ALEX STRAIKER<br />

Department of Pharmacology, Dalhousie University Halifax, NS., Canada<br />

5 IN SILICO PREDICTIONOF CONJUNCTIVAL DRUG PERMEABILITY<br />

EVA M. DEL AMO 1 , EVA RAMSAY 1,2 , THEO PICARDET 1 , SEPPO AURIOLA 1 ,<br />

ELISA TOROPAINEN 1 , MARIKA RUPONEN 1 , ARTO URTTI 1,2<br />

1<br />

School of Pharmacy, University of Eastern Finland, Kuopio, Finland<br />

2<br />

Faculty of Pharmacy, University of Helsinki, Finland<br />

6 INTRAVITREAL NA3 IS SUPPORTS RETINAL STRUCTURE AND<br />

FUNCTION IN THE RCS RAT<br />

MONICA M. JABLONSKI, XIANGDI WANG, YUNFENG SHI, AND SUMANA CHINTALAPUDI<br />

University of Tennessee Health Science Center, Memphis, TN 38163<br />

7 INTERACTION OF REACTIVATED ASTROCYTES AND RETINAL GANGLION CELLS<br />

FOLLOWING ENDOTHELIN ADMINISTRATION<br />

SHAOQING HE, HAI-YING MA, AND THOMAS YORIO<br />

North Texas Eye Research Institute, University of North Texas Health<br />

Science Center at Fort Worth, USA<br />

8 CHARACTERIZATION OF CALCIUM CHANNEL EXPRESSION IN PRIMARY<br />

OPTIC NERVE HEAD ASTROCYTES<br />

YULIYA NAUMCHUK 1 , VIDHYA R. RAO 1,2,3 , ALEXANDRA D. HEGEL 1,3 ,<br />

ALEXANDER ROCKWELL 1 , VICKI HUSAK 3 ,SIMON KAJA 1,2,3<br />

1<br />

Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago,<br />

Stritch School of Medicine, Maywood, IL, USA<br />

2<br />

Department of Ophthalmology, Loyola University Chicago<br />

Stritch School of Medicine, Maywood, IL, USA<br />

3<br />

Research Service, Edward Hines Jr. VA Hospital, Hines, IL, USA<br />

90


POSTER BOARD<br />

TITLE AND AUTHOR<br />

9 TARGETING OPTIC NERVE HEAD ASTROCYTES IN DRUG DISCOVERY FOR<br />

PRIMARY OPEN ANGLE GLAUCOMA<br />

SIMON KAJA 1,2,3 ,VIDHYA R. RAO 1,2,3 ,ALEXANDRA D. HEGEL 1,2,3 ,<br />

ALEXANDERJAMIE C. FLOSS 2 , VICKI HUSAK 3 , EVAN B. STUBBS JR. 1,3<br />

1<br />

Department of Ophthalmology, Loyola University Chicago, Stritch School<br />

of Medicine, Maywood, IL, USA<br />

2<br />

Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago<br />

Stritch School of Medicine, Maywood, IL, USA<br />

3<br />

Research Service, Edward Hines Jr. VA Hospital, Hines, IL, USA 4<strong>Program</strong> in<br />

Neuroscience, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA<br />

10 ALDH2 REGULATES ANGIOGENESIS<br />

GINEVRA NANNELLI 1 , ERIKA TERZUOLI 1 , MARINA ZICHE 1 AND SANDRA DONNINI 1<br />

1<br />

Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy<br />

11 PURINERGIC RECEPTOR MEDIATED INDUCTION OF INTERLEUKIN-1β IN MÜLLER<br />

AND MICROGLIAL CELLS IN RELATION TO GLAUCOMA<br />

JULIE SANDERSON 1 , MATTHEW FELGATE 1 , SOFIA HABIB 1,2 , PHILLIP WRIGHT 1 ,<br />

LEANNE STOKES 1 , NUWAN NIYADURUPOLA 2 AND DAVID C BROADWAY 1,2<br />

1<br />

School of Pharmacy, University of East Anglia, Norwich, UK<br />

2<br />

Department of Ophthalmology, Norfolk and Norwich University Hospital, Norwich, UK<br />

12 AGE-RELATED DIFFERENCES AFTER BRIGHT LIGHT EXPOSUREIN BALB/C MICE<br />

SYMANTAS RAGAUSKAS 1,3 , TAMUNA BOLKVADZE 1 , AGNE ZINIAUSKAITE 1 ,<br />

HENRI O. LEINONEN 2,4 , HEIKKI TANILA 2 , GIEDRIUS KALESNYKAS 1<br />

1<br />

R&D, Experimentica Ltd, Kuopio, Finland,<br />

2<br />

Neurobiology, University of Eastern Finland, Kuopio, Finland<br />

3<br />

State Research Institute for Innovative Medicine, Vilnius, Lithuania<br />

4<br />

Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, USA<br />

13 CORNEAL SURFACE TEMPERATURE UNDER<br />

PERFLUOROHEXYLOCTANE EYE DROPS<br />

M. CARMEN ACOSTA, CAROLINA LUNA, SUSANA QUIRCE, ENRIQUE VELASCO,<br />

ADOLFO ARACIL, JUANA GALLAR<br />

Universidad Miguel Hernandez, Valencia, Spain<br />

14 INNER RETINAL CHANGE IN NORMAL-TENSION GLAUCOMA<br />

JIE HYUN KIM, HAE-YOUNG LOPILLY PARK, CHAN KEE PARK<br />

Department of Ophthalmology and Visual Science,<br />

College of Medicine, The Catholic University of Korea, Seoul, Korea<br />

15 ANTERIOR CHAMBER VERSUS POSTERIOR CHAMBER PERFUSION IN LIVING<br />

MICE DOES NOT INFLUENCE MEASUREMENT OF AQUEOUS OUTFLOW<br />

FACILITY BY CONSTANT FLOW INFUSION<br />

J. CAMERON MILLAR, NAVITA N. LOPEZ, GAURANG C. PATEL, TIEN N. PHAN AND ABBOT F. CLARK<br />

North Texas Eye Research Institute, University of North Texas Health Science Center, 3500<br />

Camp Bowie Boulevard, Fort Worth, TX USA<br />

16 NOVEL TARGETS OF δ-OPIOID RECEPTOR AGONIST FOR RGC NEUROPROTECTION<br />

SHAHID HUSAIN<br />

Medical University of South Carolina, USA<br />

91


POSTER BOARD<br />

TITLE AND AUTHOR<br />

17 MAPRACORAT, A NOVEL SELECTIVE GLUCOCORTICOID RECEPTOR AGONIST,<br />

INHIBITS CYTOKINE SECRETION IN HUMAN MAST CELLS<br />

MONICA BAIULA 1 , SANTI M. SPAMPINATO 1<br />

1<br />

Dept. of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy<br />

18 CIPROXIFAN, AN H3 RECEPTOR INVERSE AGONIST, LOWERS IOP AND AMELIORATES<br />

OCULAR VASCULAR REACTIVITY IN NEW ZEALAND WHITE RABBIT MODELS OF GLAUCOMA<br />

CECILIA LANZI 1 , LAURA LUCARINI 1 , MARIA CONCETTA DURANTE 1 , ALESSANDRO PINI 2 ,<br />

FRANCESCO IMPAGNATIELLO 3 , ELENA BASTIA 3 , HOLGER STARK 4 AND EMANUELA MASINI 1<br />

1<br />

Department of NEUROFARBA, Section of Pharmacology, ; 2 Department of Experimental and<br />

Clinical Medicine, University of Florence, Italy; 3 Nicox Research Institute, Bresso, Milan, Italy<br />

4<br />

Heinrich-Heine Düsseldorf University, Institute of Medicinal Chemistry, Düsseldorf, Germany<br />

19 EFFICACY OF A NEW FOOD SUPPLEMENT IN A MURINE MODEL OF OPTICNEURITIS<br />

DARIO RUSCIANO 1 , MAURIZIO CAMMALLERI 2 , FILIPPO LOCRI 2 , MASSIMO DAL MONTE 2<br />

AND PAOLA BAGNOLI 2<br />

1<br />

Sooft Italia, Montegiorgio, Italy; 2 Department of Biology, University of Pisa, Italy<br />

20 THE WNT SIGNALING PATHWAY IN TRABECULAR MESHWORK CELLSCAN BE MODULATED<br />

BY EXOSOMES DERIVED FROM NON-PIGMENTED CILIARY EPITHELIAL CELLS<br />

ELIE BEIT-YANNAI 1 , SOFIA AVISSAR 1 , NATALIE LERNER 1<br />

1<br />

Clinical Biochemistry and Pharmacology, Ben-Gurion University, Beer-Sheva, Israel<br />

21 EXPRESSION OF OCULAR SURFACE MUCIN IN DRY EYE INDUCED MOUSE MODEL BY<br />

CURRENT DRY EYE TOPICAL MEDICATIONS<br />

INHEE MOON, YEO AREUM 1 , NOH HAEMI 1 , HYUN CHANG KIM 1,2 ,<br />

JONG SUK SONG 3 , HYUNG KEUN LEE1, 4<br />

Department of ophthalmology, Severance hospital, College of medicine, Yonsei University<br />

Seodaemun-gu, Seoul, Korea<br />

22 PKCβ INHIBITION IMPAIRS VEGF INDUCED OCULAR ANGIOGENESIS<br />

LUCIA MORBIDELLI, MARTINA MONTI, DARIA MOCHLY-ROSEN 1 , MARINA ZICHE<br />

Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy and<br />

1<br />

Department of Chemical and Systems Biology, Stanford University School of Medicine<br />

Stanford, CA 94305, USA<br />

23 ANTIANGIOGENIC AND ANTI-INFLAMMATORY ACTIVITY OF UPARANT IN THE RABBIT CORNEA<br />

VALERIO CICCONE, LORENZO BAZZANI, DARIO RUSCIANO 1 , VINCENZO PAVONE 2 , MARIO DE ROSA 3 ,<br />

MARINA ZICHE AND LUCIA MORBIDELLI<br />

Dept. Life Sciences, Univ. Siena, Italy; 1 Sooft Italia Spa, Montegiorgio, Italy<br />

2<br />

Department of Chemical Science, University of Naples “Federico II” via Cintia, 80126<br />

Napoli, Italy, 3 Department of Experimental Medicine, Second University of Naples, Napoli, Italy<br />

24 LIGHT-INDUCIBLE RHODOPSIN MUTANTS (TVRM4/+) MICE: CHARACTERIZATION AND<br />

THERAPEUTIC APPROACH<br />

ILARIA PIANO 1 , CLAUDIA GARGINI 1 , ELENA NOVELLI 2 , MARTINA BIAGIONI 2,4 , FABIOLA BONEZZI 3 ,<br />

GIUSEPPE CAMPISI 3 , RICCARDO GHIDONI 3 , CLAUDIA GARGINI 1 AND ENRICA STRETTOI 2 .<br />

1<br />

Department of Pharmacy, University of Pisa, Italy; 2 CNR Neuroscience Institute, Pisa, Italy<br />

3<br />

Biochemistry and Molecular Biology Laboratory, Health Sciences Department<br />

University of Milan, San Paolo Hospital Medical School, Milan, Italy<br />

4<br />

Tuscan Doctorate School in Neuroscience, University of Pisa<br />

92


POSTER BOARD<br />

TITLE AND AUTHOR<br />

25 TRPV4 STIMULATION INDUCED ARALKYLAMINE N-ACETYLTRANSFERASE (AANAT)<br />

PHOSPHORYLATION AND MELATONIN PRODUCTION VIA CA-CALMODULIN PATHWAY IN<br />

HUMAN CILIARY BODY EPITHELIAL CELLS<br />

JESÚS PINTOR, HANAN AWAD ALKOZI AND MARIA J. PEREZ DE LARA<br />

1<br />

Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry,<br />

Universidad Complutense de Madrid, Madrid, Spain<br />

26 INTRAPERITONEAL INJECTION OF AN ANTI-APOPTOTIC PEPTIDE INHIBITS RETINAL<br />

GANGLION CELL DEATH IN ANIMAL MODELS OF GLAUCOMA<br />

RAM H. NAGARAJ 1 , RAGHU R. KRISHNAMOORTHY 2 , SRUTHI SAMPATHKUMAR 3<br />

AND DOROTA L. STANKOWSKA 2<br />

1<br />

Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045<br />

2<br />

North Texas Eye Research Institute, UNT Health Science Center, Fort Worth, TX 76107 and<br />

3<br />

Department of Ophthalmology and Visual Sciences, Case Western Reserve University<br />

Cleveland, OH 44106<br />

27 MICROARRAY TRANSCRIPTOME ANALYSIS OF CORNEA AND LACRIMAL GLAND OF<br />

IL-22 KNOCK-OUT AND LYMPHATIC HYPOPLASIA TRANSGENIC MOUSE MODELS<br />

EUN YOUNG CHOI, MD 1 , HYUN GOO KANG, MD 1 , AREUM YEO1, SO YI JUNG 2 , HYUNG KEUN LEE, MD 1 ,<br />

1<br />

Department of Ophthalmology, Yonsei University College of Medicine<br />

Seoul, Republic of Korea; 2 Macrogen Inc. Seoul, Korea<br />

28 REGULATION OF INTRAOCULAR PRESSURE BY MICRORNA CLUSTER MIR-143/145<br />

JING MA 1 , XINYU LI 2 , MEI, XIN 3 , PEDRO GONZALEZ 4 AND SHUSHENG WANG 1, 5<br />

1<br />

Department of Cell and Molecular Biology,<br />

2<br />

Department of Ophthalmology, Tongji Hospital, Tongji Medical College,<br />

Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan,<br />

Hubei 430030, People's Republic of China<br />

3<br />

Cincinnati Children's Hospital Medical Center, Department of Pediatrics,<br />

University of Cincinnati, Cincinnati 45247, OH, USA<br />

4<br />

Department of Ophthalmology, Duke University, Durham, North Carolina, USA<br />

5<br />

Department of Ophthalmology, Tulane University, New Orleans, LA, 70118, USA<br />

29 ADVANCED ANTAGONIST OF RETINOL-BINDING PROTEIN 4 FOR TREATMENT OF<br />

THE ATROPHIC FORM OF AGE-RELATED MACULAR DEGENERATION<br />

KONSTANTIN PETRUKHIN<br />

Department of Ophthalmology, Columbia University<br />

New York, NY 10032, USA<br />

30 UNIQUE HYDROGEL TECHNOLOGY - IN VITRO MODEL REPRESENTING CORNEAL LAYERS<br />

AGNĖ ŽINIAUSKAITĖ, VYTAUTAS CĖPLA, RAMŪNAS VALIOKAS, GIEDRIUS KALESNYKAS AND<br />

JENNI J. HAKKARAINE<br />

Experimentica Ltd, R&D department, Microkatu, Finland<br />

31 HSP27 ADDITIONINTENSIFIES AII AMACRINE CELL AND SYNAPSE DAMAGE INDUCED<br />

BY S100BIMMUNIZATION IN AN AUTOIMMUNE GLAUCOMA MODEL<br />

STEPHANIE C. JOACHIM, SABRINA REINEHR, SANDRA KUEHN, CHRISTINA CASOLA,<br />

DENNIS KOCH, GESA STUTE, H. BURKHARD DICK<br />

Experimental Eye Research Institute, University Eye Hospital,<br />

Ruhr-University Bochum, Bochum, Germany<br />

93


POSTER BOARD<br />

TITLE AND AUTHOR<br />

32 PEA-15 PHOSPHOPROTEIN MEDIATES OPTIC NERVE ASTROCYTE PHAGOCYTOSIS<br />

YANG LIU 1,2 , GULAB ZODE 1 , ABBOT F. CLARK 1 , IOK-HOU PANG 1,2<br />

1<br />

North Texas Eye Research Institute, 2 Department of Pharmaceutical Sciences<br />

University of North Texas Health Science Center, Fort Worth, TX 76107<br />

33 EPIGALLOCATEQUINGALLATE AND MELATONIN IN ORAL ADMINISTRATION<br />

IMPROVE VISUAL FUNCTION IN A RETINAL DEGENERATION MODEL, THE P23H RAT<br />

LORENA PERDICES 1 , ISABEL PINILLA 1,2 , LORENA FUENTES-BROTO 1,3 , FRANCISCO J. SEGURA 4 ,<br />

GEMA INSA SÁNCHEZ 2 , ELVIRA ORDUNA 2 , ANA ISABEL SÁNCHEZ-CANO 5,2 , NICOLÁS CUENCA 6<br />

1<br />

Institute for Health Research of Aragón (IIS Aragón), Zaragoza, Spain<br />

2<br />

Universitary Hospital Lozano Blesa, Zaragoza, Spain;<br />

3<br />

Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain<br />

4<br />

Department of Surgery, University of Zaragoza, Zaragoza, Spain<br />

5<br />

Department of Applied Physics, Zaragoza University, Spain<br />

6<br />

Department of Physiology Genetics and Microbiology, Alicante University, Alicante, Spain<br />

34 OFF-LABEL DRUGS USE IN OCULAR PHARMACOLOGY<br />

LUCIA GOZZO 1 , LAURA LONGO 1 , SILVANA MANSUETO 1 , FILIPPO DRAGO 1,2<br />

1<br />

Regional Pharmacovigilance Centre/Clinical Pharmacology <strong>Program</strong>, University Hospital<br />

of Catania, Italy; 2 Department of Biomedical and Biotechnological Sciences<br />

University of Catania, Italy<br />

35 VITAMIN D IN SYSTEMIC SCLEROSIS PATIENTS WITH DRY EYE SYNDROME<br />

MIRIAM GALLO AFFLITTO, CARLO RAPISARDA 1 , ROBERTA AMATO 1,2 , SALVO FICILI 1,2 , DAVIDE SCOLLO 1<br />

GIOVANNI PANTA 1 , DANIELA ROCCA 1,2 , AGATA MESSINA 1,2 , ROSARIO FOTI 3 , TERESIO AVITABILE 1<br />

ELISA VISALLI 3 , CATERINA GAGLIANO 1,2<br />

1<br />

Eye Clinic, Catania University, Italy; 2 Neurovisual Science Technology (NEST), Italy<br />

3<br />

Operative Unit of Reumatolgy,A.O.U. Policlinico Vittorio Emanuele, Catania University, Italy<br />

36 DESIGN, SYNTHESIS, AND CHARACTERIZATION OF A SELECTIVE INHIBITOR FOR<br />

RETINALDEHYDE DEHYDROGENASE (ALDH1A) ENZYMES<br />

ANGELICA HARPER 1 , ANH LE 2 , TIM MATHER 3 , ANTHONY BURGETT 2 ,JODY A. SUMMERS 1<br />

1<br />

Department of Cell Biology, University of Oklahoma Health Sciences Center<br />

Oklahoma City, United States of America;<br />

2<br />

Department of Chemistry and Biochemistry<br />

University of Oklahoma, Norman, OK, United States of America<br />

3<br />

Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America<br />

37 NANOTECHNOLOGICAL SIRNA FORMULATIONS FOR THE TREATMENT OF<br />

DIABETIC RETINOPATHY<br />

SARHA CUPRI, MARIALAURA AMADIO, ALESSIA PASCALE, CECILIA OSERA, VELIA D'AGATA,<br />

AGATA GRAZIA D'AMICO, GIAN MARCO LEGGIO, BARBARA RUOZI, STEFANO GOVONI,<br />

FILIPPO DRAGO, CLAUDIO BUCOLO, ROSARIO PIGNATELLO<br />

Department of Drug Sciences, University of Catania, Italy<br />

38 PROTECTIVE EFFECT OF ID PROTEIN ON TGFβ2INDUCED FIBROSIS<br />

IN HUMAN TRABECULAR MESHWORK CELLS:<br />

IMPLICATION FOR DEVELOPING A GLAUCOMA THERAPY<br />

AVANI A. MODY, ROBERT J. WORDINGER, ABBOT F. CLARK<br />

North Texas Eye Research Institute<br />

University of North Texas Health Science Center, Fort Worth, TX USA<br />

94


POSTER BOARD<br />

TITLE AND AUTHOR<br />

39 ROLE OF GLUCOCORTICOID RECEPTOR GRβ IN GLUCOCORTICOID-INDUCED<br />

OCULAR HYPERTENSION AND GLAUCOMA IN MICE<br />

GAURANG C. PATEL, YANG LIU, J. CAMERON MILLAR, AND ABBOT F. CLARK<br />

North Texas Eye Research Institute, University of North Texas Health Science Center<br />

Fort Worth, TX-76107, USA<br />

40 HUMAN-SPECIFIC LONG NON-CODING RNAS REGULATEOCULAR ANGIOGENESIS<br />

BO YU 1 , QINBO ZHOU 1 , CHASTAIN ANDERSON 1 , JAKUB HANUS 1 , FANGKUN ZHAO 1 , JING MA 1<br />

KUN ZHANG 3 AND SHUSHENG WANG 1, 2<br />

1<br />

Department of Cell and Molecular Biology<br />

2<br />

Department of Ophthalmology, Tulane University New Orleans, LA, 70118, USA<br />

3<br />

Department of Computer Science, Xavier University, New Orleans, LA, 70125<br />

41 OCULAR TISSUE DISTRIBUTION OF ORALLY ACTIVE MULTIFUNCTIONAL ANTIOXIDANTS<br />

DAMIAN M. DASZYNSKI 1 , THEODOR A. WOOLMAN 1 , KAREN BLESSING1, AND PETER F. KADOR 1,2<br />

1College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA<br />

2<br />

Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE, USA<br />

42 TARGETING INFLAMMATION TO DELAY PHOTORECEPTOR DEGENERATION IN AN ANIMAL<br />

MODEL OF RETINITIS PIGMENTOSA<br />

MARTINA BIAGIONI 1 , VIVIANA GUADAGNI 1 , ELENA NOVELLI 1 , ENRICA STRETTOI 1<br />

1<br />

CNR Neuroscience Institute, Pisa, Italy<br />

43 AGE-RELATED MACULAR DEGENERATION AND TGF-β1: PHARMACODYNAMIC<br />

AND PHARMACOKINETIC PROFILE<br />

FRANCESCA LAZZARA, CLAUDIO BUCOLO, LEGGIO GIAN MARCO, VINCENZO FISICHELLA,<br />

GIURDANELLA GIOVANNI, CHIARA BIANCA MARIA PLATANIA, ANNAMARIA FIDILIO,<br />

FEDERICA GERACI, FILIPPO DRAGO<br />

Department of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania, Italy<br />

44 ANTIOXIDANT AND OSMOPROTECTING ACTIVITY OF TAURINE IN DRY EYE MODELS<br />

ANNAMARIA FIDILIO, CLAUDIO BUCOLO, CHIARA BIANCA MARIA PLATANIA, FRANCESCA LAZZARA<br />

FEDERICA GERACI, FILIPPO DRAGO<br />

Department of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania, Italy<br />

45 IDENTIFICATION OF A GENE SIGNATURE REGULATED BY A LNCRNA ASSOCIATED<br />

WITH EXFOLIATION GLAUCOMA<br />

WILLIAM M. JOHNSON 1 , INAS F. ABOOBAKAR 1 , LAURA K. FINNEGAN 2 , R. RAND ALLINGHAM 1<br />

MICHAEL A. HAUSER 1,3,4 , W. DANIEL STAMER 1<br />

1<br />

Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA<br />

2<br />

School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland<br />

3<br />

Department of Medicine, Duke University Medical Center, Durham, NC, USA<br />

4<br />

Singapore Eye Research Institute, Singapore National Eye Center, Singapore, Singapore<br />

Duke, National University of Singapore, Singapore, Singapore<br />

46 MIRNAS IN VITREOUS HUMOR OF PATIENTS AFFECTED BY IDIOPATHIC EPIRETINAL<br />

MEMBRANE AND MACULAR HOLE<br />

MARIO D. TORO (PRESENTER), 1 ANDREA RUSSO 1 , MARCO RAGUSA 2 , ANTONIO LONGO 1<br />

TERESIO AVITABILE 1 , CINZIA DI PIETRO 2 , DAVIDE BARBAGALLO 2 , MICHELE REIBALDI 1<br />

1<br />

Department of Ophthalmology, University of Catania<br />

2<br />

Department of Biology, University of Catania<br />

95


POSTER BOARD<br />

TITLE AND AUTHOR<br />

47 INNER RETINAL REMODELING IN AN INDUCIBLE MOUSE<br />

MODEL OF RETINITIS PIGMENTOS<br />

ANTONIA STEFANOV 1 , ELENA NOVELLI 1 , ENRICA STRETTOI 1<br />

1<br />

CNR Neuroscience Institute, Pisa, Italy<br />

48 RETINAL PK PROFILE OF CURCUMIN AFTER ORAL ADMINISTRATION IN RABBITS<br />

1<br />

CLAUDIO BUCOLO, 1 ANNAMARIA FIDILIO, 1 CHIARA BIANCA MARIA PLATANIA,<br />

1<br />

FRANCESCA LAZZARA, 1 FEDERICA GERACI, 2 CATENO PIAZZA<br />

1<br />

SALVATORE SALOMONE, 1 FILIPPO DRAGO<br />

1<br />

Department of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania, Italy<br />

2<br />

Research Centre, Unifarm, Catania, Italy<br />

49 NANOPARTICLE INTERACTION WITH VITREOUS HUMOR AND ITS EFFECT ON RETINAL<br />

PIGMENT EPITHELIAL CELL UPTAKE<br />

RYAN A. KELLEY AND UDAY B. KOMPELLA<br />

Skaggs School of Pharmacy and Pharmaceutical Sciences<br />

University of Colorado Anschutz Medical Center, Aurora, CO, USA<br />

50 TRANSPALPEBRAL RHEOOPHTHALMOGRAPHYEVALUATION OF THE IMPACT OF<br />

PROSTAGLANDIN ANALOGS ON OCULAR HEMODYNAMICS INEARLY PRIMARY<br />

OPEN-ANGLE GLAUCOMA<br />

ELENA IOMDINA 2 , ALINA KLEYMAN 1 , OLGA KISELEVA 1 , ALEXANDER BESSMERTNY 1 ,<br />

PETER LUZHNOV 3 , DMITRY SHAMAEV 3<br />

1<br />

Department of Glaucoma, Moscow Helmholtz Research Institute of<br />

Eye Diseases, Moscow, Russia<br />

2<br />

Department of Refraction Pathology, Binocular Vision Anomalies and<br />

Ophthalmoergonomics, Moscow Helmholtz Research Institute of Eye Diseases, Moscow, Russia<br />

51 PRESENCE OF MELANOPSIN IN HUMAN CRYSTALLINE LENS EPITHELIAL<br />

CELLS AND ITS ROLE IN MELATONIN SYNTHESIS<br />

HANAN AWAD ALKOZI, XIAOYU WANG, MARIA JESUS PEREZ DE LARA AND JESUS PINTOR<br />

Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry<br />

Universidad Complutense de Madrid, Madrid, Spain<br />

52 IMMUNOHISTOCHEMICAL CHARACTERIZATION OF NEUROTRANSMITTERS IN THE<br />

EPISCLERAL CIRCULATION IN RATS<br />

ANJA LADEK 1 , ANDREA TROST 1 , CHRISTIAN RUNGE 1 , FALK SCHRÖDL 1<br />

CLEMENS A. STROHMAIER 1 , HERBERT A. REITSAMER 1<br />

1<br />

Ophthalmology, Paracelsus Medical University<br />

SALK, MüllnerHauptstrasse 48, 5020 Salzburg, Austria<br />

53 ROLE OF LACTOBIONIC ACID AND HYALURONIC ACID IN PROMOTION OF CORNEAL<br />

EPITHELIAL WOUND HEALING IN VITRO AND IN VIVO<br />

MELANIA OLIVIERI 1 , DARIO RUSCIANO 1 , SALVATORE PEZZINO 2 , C. DANIELA ANFUSO 3<br />

GABRIELLA LUPO 3 , MARTINA CRISTALDI 1<br />

1<br />

Sooft Italia, University of Catania, Catania, Italy<br />

2Bioos Italia, University of Catania, Catania, Italy<br />

3<br />

Department of Biomedical and Biotechnological Sciences<br />

University of Catania, Catania, Italy<br />

96


POSTER BOARD<br />

TITLE AND AUTHOR<br />

54 EFFICACY AND SAFETY ASSESSMENT OF LACTOBIONIC ACID FOR THE TREATMENT<br />

OF DRY EYE SYNDROME<br />

ALESSANDRA PIZZO 1 , GAGLIANO C. 1,2 , AMATO R. 1,2 , FICILI S. 1,2 , MALAGUARNERA G. 1,2<br />

1<br />

Ophthalmology Department, University of Catania, Eye Clinic, Catania (Italy)<br />

2<br />

Neurovisual Science Technology (NEST), Catania (Italy)<br />

55 VEGF EXACERBATESHIGH-GLUCOSE-INDUCED DAMAGE IN HUMAN RETINAL<br />

ENDOTHELIAL CELLS<br />

GIOVANNI GIURDANELLA, FRANCESCA LAZZARA, CLAUDIO BUCOLO<br />

SALVATORE SALOMONE, FILIPPO DRAGO<br />

Dept. of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania, Italy<br />

56 SULODEXIDE PREVENTS HIGH GLUCOSE DAMAGE IN HUMAN RETINAL<br />

ENDOTHELIAL CELLS<br />

GIOVANNI GIURDANELLA 1 , FRANCESCA LAZZARA 1 , NUNZIA CAPORARELLO 1 ,<br />

GIAN MARCO LEGGIO 1 , GABRIELLA LUPO 1 , CARMELINA DANIELA ANFUSO 1 ,<br />

CLAUDIO BUCOLO 1 , SALVATORE SALOMONE 1 , FILIPPO DRAGO 1<br />

1<br />

Dept. of Biomedical and Biotechnological Sciences, School of Medicine,<br />

University of Catania, Catania, Italy<br />

57 GABAPENTIN NANOCARRIERS TO MANAGE OCULAR PAIN<br />

ROSARIO PIGNATELLO 1 , SARHA CUPRI 1 , CLAUDIO BUCOLO 2 , FILIPPO DRAGO 2 ,<br />

DARIO RUSCIANO 3 , TERESA MUSUMECI 1 , GIOVANNI PUGLISI 1<br />

1<br />

NANO-i — Research Center on Ocular Nanotechnology,<br />

Department of Drug Sciences, University of Catania, Catania, Italy<br />

2<br />

Department of Biomedical and Biotechnological Sciences School of Medicine<br />

University of Catania, Catania, Italy<br />

3<br />

SOOFT Italia SpA, Montegiorgio (Fermo), Italy<br />

58 RECENT ADVANCES IN THE APPLICATION OF LIPID-BASED NANOCARRIERS<br />

TO OCULAR DRUG DELIVERY<br />

R. PIGNATELLO, C. CARBONE , T. MUSUMECI, S. CUPRI, V. PEPE, G. PUGLISI<br />

NANO-i — Research Center on Ocular Nanotechnology<br />

Department of Drug Sciences, University of Catania, Catania, Italy<br />

59 P2X7 RECEPTOR AS PHARMACOLOGICAL TARGET IN DIABETIC RETINOPATHY<br />

CHIARA BIANCA MARIA PLATANIA, GIOVANNI GIURDANELLA 1 , LUISA DI PAOLA 2<br />

GIAN MARCO LEGGIO 1 , SALVATORE SALOMONE A , FILIPPO DRAGO A , CLAUDIO BUCOLO A<br />

1<br />

Section of Pharmacology, Department of Biomedical and<br />

Biotechnological Sciences School of Medicine,<br />

University of Catania, Catania, Italy<br />

2<br />

School of Engineering, University Campus BioMedico, Roma, Italy<br />

60 STRUCTURAL DIFFERENCES BETWEEN P2X7 AND P2X4 RECEPTORS:<br />

A PROTEIN CONTACT NETWORK ANALYSIS<br />

CHIARA BIANCA MARIA PLATANIA 1 , LUISA DI PAOLA 2 , FILIPPO DRAGO 1 , CLAUDIO BUCOLO 1<br />

1<br />

Department of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania ,<br />

2<br />

Università-Campus Biomedico, Roma<br />

97


POSTER BOARD<br />

TITLE AND AUTHOR<br />

61 EFFECT OFDIETARY CHOLESTEROL ONOCULAR PATHOLOGIES IN AN AGE-RELATED<br />

MACULAR DEGENERATION MOUSE MODEL<br />

MICHAEL LANDOWSKI 1 , UNA KELLY 1 , MARYBETH GROELLE 1 , CATHERINE BOWES RICKMAN 1,2<br />

1<br />

Department of Ophthalmology, Duke University Medical Center,<br />

Durham, NC, USA<br />

2<br />

Department of Cell Biology, Duke University Medical Center,<br />

Durham, NC, USA<br />

62 TREATMENT WITH AN HDAC3 SELECTIVE INHIBITOR PREVENTS RETINAL GANGLION CELL<br />

NUCLEAR ATROPHY AND APOPTOSIS AFTER ACUTE AND CHRONIC OPTIC NERVE INJURY<br />

HEATHER M. SCHMITT 1 , 2,4 , GUOJUN CHEN 3,4 , YUYUANWANG 3,4 , CASSANDRA L. SCHLAMP 1,4<br />

SHAOQUIN GONG 3,4 , ROBERT W. NICKELLS 1,4<br />

1<br />

Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI<br />

2<br />

Cellular and Molecular Pathology, University of Wisconsin-Madison, Madison WI<br />

3<br />

BIONATES Theme, Wisconsin Institute for Discovery, Madison WI<br />

4<br />

McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI<br />

63 B2R SIGNALINGIN NEO-ANGIOGENESIS<br />

SANDRA DONNINI, ERIKA TERZUOLI, MARINA ZICHE<br />

Department of Life Sciences<br />

University of Siena, Italy<br />

64 OCULAR PHARMACOKINETICS PROFILE OF PALMITOYLETHANOLAMIDE<br />

ENCAPSULATED IN NEW NANOSTRUCTURED LIPIDIC CARRIER<br />

FEDERICA GERACI 1 , CLAUDIO BUCOLO 1 , CHIARA BIANCA MARIA PLATANIA 1<br />

GIOVANNI LUCA ROMANO 1 , CARMELO PUGLIA 2 , ROSARIO PIGNATELLO 2 ,<br />

EDUARDO MARIA SOMMELLA 3 , PIETRO CAMPIGLIA 3 , CARMINE OSTACOLO 4 , FILIPPO DRAGO 1<br />

1<br />

Department of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania, Italy, 2 Section of Pharmaceutical Technology, Department of<br />

Drug Sciences, University of Catania Catania, Italy<br />

3<br />

Department of Pharmacy, University of Salerno, Fisciano (SA), Italy<br />

4<br />

Department of Pharmacy, School of Medicine, University Federico II of Naples, Naples, Italy<br />

98


POSTER BOARD N 1<br />

IN VIVO COMPARISON OF THE RESIDENCE TIME OF CROSS-LINKED<br />

COMPARED TO LINEAR HYALURONIC ACID IN RABBIT EYE<br />

MIRKO MUZZI 1 ; RITA MENCUCCI 2<br />

1<br />

Department of Health Sciences, Section of Clinical Pharmacology and Oncology<br />

University of Florence, Florence, Italy; 2 Ophthalmology Unit, Careggi Hospital, Florence, Italy<br />

Purpose: Dry Eye Disease is one of the most common ophthalmic disorders. Tear<br />

substitutes are commonly prescribed and hyaluronic acid is one of the most used<br />

components, requiring several administrations a day. Our aim is to evaluate the<br />

behaviour of a chemically modified and crosslinked derivative of hyaluronic acid<br />

in the attempt to find a tear substitute capable to have a longer<br />

residence time on the ocular surface<br />

Methods: Linear hyaluronic acid (HA) and cross-linked hyaluronic acid<br />

(CLHA) were derivatized by the fluorescent probe 5-dimethylamino-naphthalene-1-(2-amino-ethyl)-sulphonamide<br />

to obtain<br />

the respective fluorescent green compounds. HA and CLHA fluorescent solutions<br />

(50µl of 0.5%) were instilled onto the ocular surface of rabbit’s eyes whereas<br />

the controls received 50µl of fluorescent saline solution.<br />

The permanence of fluorescence and its intensity were evaluated using a<br />

software for images after 1,10, 30 and 60 minutes.<br />

Results: one minute after the instillation, the fluorescence was 60% higher with<br />

both HA and CLHA compared to controls.<br />

After 10 minutes, the intensity of the fluorescence signal was 33% higher for HA<br />

and 97% higher for CLHA respect to control. Notably, after further 20 minutes<br />

the fluorescence was still 64% higher than control only in CLHA treated eyes,<br />

while as for HA no difference respect to control was found.<br />

Conclusions: the present study of residence time kinetics in rabbit eye shows that<br />

CLHA has a longer residence time on the ocular surface compared to linear HA.<br />

99


POSTER BOARD N 2<br />

PLACENTA GROWTH FACTOR PLAYS A ROLE IN IMMUNE RESPONSE<br />

ASSOCIATED WITH CHOROIDAL NEOVASCULARIZATION<br />

SERGIO CRESPO-GARCIA 1 , CAITLIN CORKHILL 1 , CHRISTOPHE ROUBEIX 1,2,<br />

NOR BERT KOCIOK 1 , OLAF STRAUSS 1 , ANTONIA M. JOUSSEN 1 , NADINE REICHHART 1<br />

1<br />

Department of Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany<br />

2<br />

Einstein Foundation, Berlin, Germany<br />

Purpose: Inflammatory cells such as mononuclear phagocytes (MP) are crucial<br />

for choroidal neovascularization (CNV) progression. A switch from pure<br />

anti-VEGF-A intravitreal treatment to aflibercept, a drug with combined anti-VEGF-A<br />

and anti-placenta growth factor (PlGF) activity, has been reported to<br />

be beneficial for some patients who do not respond to anti-VEGF-A alone. Since<br />

MP express VEGFR1, we hypothesize that the interplay of PlGF/VEGFR1 in immune<br />

cells plays a critical role for CNV.<br />

Methods: Laser burns (50µm, 0.1s, 120 mW) induced CNV, and immune cells and<br />

neovascularization were analyzed both in vivo and ex vivo. Proteins were detected<br />

using immunohistochemistry. We studied differential expression of angiogenic<br />

factors and macrophage polarization markers by means of qPCR. Intravitreal<br />

injection of aflibercept or anti-PlGF was performed 1 day after laser.<br />

Results: Early after laser, Plgf but not Vegfa was significantly upregulated.<br />

VEGF-A up-regulation is limited to the scar, whereas PlGF is more widely distributed.<br />

Pro-inflammatory macrophage (M1) markers were upregulated in the early<br />

phase of CNV. However, pro-angiogenic (M2) markers showed no clear trend.<br />

Both aflibercept and anti-PlGF diminished the leakage reduction associated to<br />

CNV in vivo. Correlating these data, the overall amount of activated subretinal<br />

MPs, and especially, of those that expressed PlGF, was also reduced in the scar site.<br />

Aflibercept showed a stronger reduction in both parameters.<br />

Conclusions: The results hint at an interplay between PlGF/VEGFR1 and MPs<br />

that is important in the early inflammatory phase of CNV. Thus anti-PlGF might<br />

represent an interesting pharmacological approach in patients not responding to<br />

standard age-related macular degeneration therapies.<br />

100


POSTER BOARD N 3<br />

AUTOREGULATION OF RETINALGANGLIONCELLFUNCTION TO<br />

METABOLICCHALLENGE IN GLAUCOMA<br />

GIOVANNI LUCA ROMANO 1 , CHOU TSUNG HANG 2 ,CLAUDIO BUCOLO 1<br />

FILIPPO DRAGO 1 , VITTORIO PORCIATTI 2<br />

1<br />

Department of Biomedical Biotechnological Sciences (BIOMETEC), University of Catania<br />

2<br />

Bascom Palmer EyeInstitute,University of Miami, Miller School of Medicine, Miami, FL, UnitedStates<br />

Purpose: Failure of autoregulatory mechanisms is thought to trigger cell death in<br />

glaucoma. In a mouse model prone to glaucoma (DBA/2J) we propose to investigate<br />

the autoregulatory response of retinal ganglion cells (RGC) under flickering<br />

light to increase metabolic demand and cause vasodilation and compare it with<br />

the response of control mice that do not develop glaucoma (C57BL/6J) RGC response<br />

dynamics with and without flicker added will be assessed with pattern<br />

electroretinogram PERG, a sensitive measure of RGC function.<br />

Methods: As the PERG is the fundamental tool of this proposal, PERG methods<br />

was optimized to record robust responses simultaneously from both eyes using<br />

a common non-corneal electrode. Comparing to standard, this approach eliminates<br />

the need of corneal manipulation that may spuriously alter IOP and induce<br />

cataract. Finally, the use of a common non-corneal electrode minimizes interocular<br />

variability and test-retest variability.<br />

Results: When a 101 Hz flicker is superimposed to the PERG stimulus, a normal<br />

PERG signal is generated. During 11 Hz flicker, the PERG signal substantially decreases.<br />

Conclusions: Preliminary results indicate that while in control mice the flicker-PERG<br />

amplitude declines and the latency increases, the opposite effect is seen<br />

in DBA/2J mice. This indicates that while in control mice the flicker-induced<br />

metabolic unbalance results in an autoregulatory RGC response, this process is<br />

altered in DBA/2J mice. Results are significant as the flicker-PERG can disclose<br />

early RGC dysfunction and predict severity of glaucoma and indicates a clear autoregulatory<br />

PERG response to flickering light in healthy retina.<br />

101


POSTER BOARD N 4<br />

CANNABINOIDS IN OCULAR PATHOPHYSIOLOGY<br />

ANNA-MARIA SZCZESNIAK, ALEX STRAIKER<br />

Department of Pharmacology, Dalhousie University Halifax, NS., Canada<br />

Purpose: The pathology of glaucoma is characterized by optic nerve damage<br />

and retinal ganglion cell (RGCs) death. Intraocular pressure (IOP) is a risk factor<br />

associated with glaucoma. Components of the endocannabinoid system (ECS),<br />

including receptors, endocannabinoids, and their biosynthetic and degradative<br />

enzymes such as MAGL, are expressed in ocular tissues. Modulation of the ECS<br />

may be useful in the treatment of glaucoma. The objectives of this study were to:<br />

1. Determine the effects of exogenous and endogenous cannabinoids on IOP in a<br />

genetic ocular hypertensive model (nee mouse).<br />

2. Assess whether the modulation of the ECS is neuroprotective for RGC survival.<br />

Methods: IOP was measured by rebound tonometry in adult nee mice administered<br />

with either the MAGL enzyme inhibitor JZL184, the cannabinoid receptor<br />

agonist WIN55,212-2, or respective vehicles. RGC survival was evaluated by immunohistochemical<br />

staining with anti-Brn3a antibody following chronic cannabinoid<br />

treatments.<br />

Results: Inhibition of MAGL enzyme with JZL184 (which decreases degradation<br />

of the endogenous cannabinoid 2-AG), or treatment with WIN55,212-2, did not<br />

lower IOP acutely in nee mice. However, chronic treatment with JZL184, but<br />

not with WIN55,212-2, resulted in a significant neuroprotective effect on RGC<br />

survival.<br />

Conclusion: Unlike previous reports, ECS manipulation via WIN55,212-2 or<br />

JZL184 did not reduce IOP in nee mice, which may be a reflection in the difference<br />

between nee (angle-closure) vs other open-angle hypertensive models.<br />

Yet, the significant neuroprotection provided by chronic administration with<br />

ECS-modulating drugs suggest that they may be useful in the treatment of glaucoma,<br />

independent of an effect on IOP modulation.<br />

102


POSTER BOARD N 5<br />

IN SILICO PREDICTIONOF CONJUNCTIVAL DRUG PERMEABILITY<br />

EVA M. DEL AMO 1 , EVA RAMSAY 1,2 , THEO PICARDET 1 , SEPPO AURIOLA 1 , ELISA TOROPAINEN 1<br />

MARIKA RUPONEN 1 , ARTO URT TI 1,2<br />

1<br />

School of Pharmacy, University of Eastern Finland, Kuopio, Finland<br />

2<br />

Faculty of Pharmacy, University of Helsinki, Finland<br />

Purpose: To build a computational quantitative structure-property relationship<br />

(QSPR) model of conjunctival permeability.<br />

Methods: Generation of conjunctival permeability (Papp, conjunctiva) values from<br />

32 drugs: Ex vivo permeability of the 32 drugs-in one dose with fresh porcine conjunctiva<br />

was undertaken in Ussing/diffusion chambers and quantified with LC-<br />

MS/MS method.<br />

Generation of the physicochemical descriptors: 34 molecular descriptors were obtained<br />

using ACDlabs® software<br />

QSPR model: multivariate analysis methods were used to build the equation relating<br />

the Papp, conjunctiva with the relevant molecular descriptors.<br />

Principal component analysis (PCA) and linear partial least square (PLS) (Simca<br />

plus®) were the methods employed.<br />

Results: A predicting model for conjunctival permeability was obtained with a Q2<br />

value of 0.624<br />

Log P app, conjunctiva (cm/s)=<br />

- 4.1594 - 0.6121×LogPSA - 0.0792×HD + 3.2914×Halogen ratio<br />

The relevant descriptors were polar surface area (PSA), hydrogen bond donor (HD)<br />

capacity and halogen ratio. The model predicted accurately the internal and external<br />

test sets with a mean fold error of 1.58 and 1.30 respectively.<br />

Conclusions: The QSPR model can be used to predict the conjunctival permeation<br />

of ophthalmic topical drugs based in their chemical structure. Descriptors related to<br />

geometry of the molecule (such PSA) are more relevant than lipophilic ones.<br />

103


POSTER BOARD N 6<br />

INTRAVITREAL NA3 IS SUPPORTS RETINAL STRUCTURE AND<br />

FUNCTION IN THE RCS RAT<br />

MONICA M. JABLONSKI, XIANGDI WANG, YUNFENG SHI, AND SUMANA CHINTALAPUDI<br />

University of Tennessee Health Science Center, Memphis, TN 38163<br />

Purpose: AMD is a debilitating disease affecting as many as 25% of elderly individuals.<br />

The purpose of this investigation was to determine if asialo-triantennary<br />

(aka NA3) provides neuroprotective support to the retina of the well-characterized<br />

RCS rat, a proof of principal atrophic AMD model.<br />

Methods: P21 rats were dosed intravitreally in both eyes with NA3 in PBS either<br />

once or twice per week for two weeks. Control groups included PBS injections<br />

and no injections. ERG, OCT and visual acuity (V A) data were collected at three<br />

time points: baseline before the start of treatment; one week after first injection;<br />

and two weeks after first injection. Rats were sacrificed after two weeks of therapy<br />

and eyes were enucleated. One eye from each rat was embedded in plastic for<br />

histological analyses including measurement of retinal layer thickness.<br />

The fellow eye was prepared for GFAP immunochemistry.<br />

Results: The structure and function of the retina was positively affected by NA3.<br />

Compared to PBS-injected or no-injection control rats, the following parameters<br />

were positively affected by NA3-treatment: ERG amplitudes (a- and b-waves);<br />

VA measurements; outer nuclear layer thickness; photoreceptor outer segment<br />

organization; and GFAP immunoreactivity. Weekly NA3 injections were as or<br />

more efficacious than a twice weekly dosing schedule.<br />

Conclusion: Intravitreal NA3 treatment supports proper photoreceptor structure<br />

and retinal function in the RCS rat. It also preserved visual acuity. These data<br />

suggest that NA3 may be an effective therapy for atrophic AMD.<br />

The development of a topical extended release formulation is in progress.<br />

104


POSTER BOARD N 7<br />

INTERACTION OF REACTIVATED ASTROCYTES AND RETINAL GANGLION<br />

CELLS FOLLOWING ENDOTHELIN ADMINISTRATION<br />

SHAOQING HE, HAI-YING MA, AND THOMAS YORIO<br />

North Texas Eye Research Institute, University of North Texas Health<br />

Science Center at Fort Worth, USA<br />

Purpose: Endothelin-1(ET-1) and its receptors are involved in the etiology of glaucoma.<br />

However, ET-mediated activation of astrocytes (ASTs) and their effect on<br />

retinal ganglion cell (RGC) survival are largely unknown. This study aimed to<br />

studying the role of ETs in the interaction between RGCs and ASTs.<br />

Methods: Primary rat RGCs and ASTs isolated from rat pups using antibody-panning<br />

methods were treated with 100nM endothelin-1 or endothelin-3 for 24 hours<br />

and subsequent protein detection was performed using western blot and immunocytochemistry.<br />

ET-1-mediated intracellular calcium was monitored in RGCs,<br />

ASTs and in a co-culture of RGCs and ASTs using Fura-2 AM calcium imaging.<br />

Cell apoptosis and necrosis was detected using Annexin V and propidium iodide<br />

staining.<br />

Results: The treatment of ET-1 or ET-3 induced the upregulation of GFAP, NCAM,<br />

c-Jun, JNK and Ki67 in ASTs. Increases in ET-1 induced GFAP were attenuated by<br />

administration of SP600125, an inhibitor of JNK, but not BQ788, an antagonist of<br />

ETB receptor. In addition, ET-1 enhanced intracellular calcium ([Ca2+]i) in ASTs<br />

and RGCs respectively. Verapamil, an L-type calcium channel blocker, inhibited<br />

the influx of calcium in ASTs, but not in RGCs. ET-1-induced elevation of [Ca2+]i<br />

was significantly attenuated in co-culture, and accordingly less cell death was also<br />

observed in co-culture based on our preliminary data.<br />

Conclusions: ET-1 induced the activation of astrocytes through a mechanism that<br />

may be involved in the control of calcium-mediated signaling and JNK/c-Jun pathway.<br />

The reactivation of ASTs initially could be neuroprotective, however, longterm<br />

it could cause axon damage and RGC death.<br />

105


POSTER BOARD N 8<br />

CHARACTERIZATION OF CALCIUM CHANNEL EXPRESSION IN PRIMARY<br />

OPTIC NERVE HEAD ASTROCYTES<br />

YULIYA NAUMCHUK 1 , VIDHYA R. RAO 1,2,3 , ALEXANDRA D. HEGEL 1,3 ,<br />

ALEXANDER ROCKWELL 1 , VICKI HUSAK 3 ,SIMON KAJA 1,2,3<br />

1<br />

Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago,<br />

Stritch School of Medicine, Maywood, IL, USA;<br />

2<br />

Department of Ophthalmology, Loyola University Chicago,<br />

Stritch School of Medicine, Maywood, IL, USA<br />

3<br />

Research Service, Edward Hines Jr. VA Hospital, Hines, IL, USA<br />

Purpose: Pathological changes in optic nerve head astrocyte (ONHA) structure<br />

and function, such as glial activation and extracellular matrix remodeling, are<br />

pathological hallmarks of primary open angle glaucoma (POAG).<br />

POAG is the most common form of glaucoma, characterized by increased intraocular<br />

pressure (IOP). Despite their critical role in the pathophysiology of glaucoma,<br />

surprisingly little is known regarding the mechanosensitive calcium signaling<br />

pathways in ONHAs. The goal of this study was, therefore, to determine<br />

the expression profile of mechanosensitive calcium channels.<br />

Methods: Immunocytochemistry was performed on primary adult rat ONHAs<br />

grown on poly-L-lysine coated glass coverslips, as described by us previously<br />

(Kaja et al., 2015 Exp Eye Res 138:159-66). Images were obtained using high resolution<br />

confocal and total internal reflection fluorescence microscopy (TIRFM).<br />

Results: ONHAs showed strong immunoreactivity for all three subtypes of the<br />

group of polycystin-2 (transient receptor potential P) calcium channels. In accordance<br />

with their function as intracellular calcium channels, fluorescence appeared<br />

as cytoplasmic punctate staining, which colocalized with the endoplasmic<br />

reticulum. Furthermore, we detected strong immunoreactivity for mechanosensitive<br />

Piezo-1 cation channels. Using TIRFM, we confirmed that Piezo-1-specific<br />

immunoreactivity was present exclusively near the plasma membrane.<br />

Conclusions: Extending our previous work that identified differential intracellular<br />

signaling of inositol-1,4,5-trisphosphate and ryanodine receptors, this novel<br />

data provide tentative evidence for the presence of complex mechanosensitive<br />

calcium signaling in ONHAs.<br />

The known interaction of Piezo-1 with TRPP channels in other organ systems<br />

allows us to speculate that Piezo-1 channels may mediate activation of intracellular<br />

signaling pathways evoked by elevated IOP in POAG.<br />

106


POSTER BOARD N 9<br />

TARGETING OPTIC NERVE HEAD ASTROCYTES IN DRUG DISCOVERY FOR<br />

PRIMARY OPEN ANGLE GLAUCOMA<br />

SIMON KAJA 1,2,3 ,VIDHYA R. RAO 1,2,3 ,ALEXANDRA D. HEGEL 1,2,3 ,<br />

ALEXANDERJAMIE C. FLOSS 2 , VICKI HUSAK 3 , EVAN B. STUBBS JR. 1,3<br />

1<br />

Department of Ophthalmology, Loyola University Chicago, Stritch School<br />

of Medicine, Maywood, IL, USA<br />

2<br />

Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago<br />

Stritch School of Medicine, Maywood, IL, USA<br />

3<br />

Research Service, Edward Hines Jr. VA Hospital, Hines, IL, USA 4 <strong>Program</strong> in Neuroscience,<br />

Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA<br />

Purpose: To establish and validate a cell culture model system that assesses the cellular<br />

and molecular consequences of elevated intraocular pressure (IOP) on the<br />

efficacy of therapeutic drug candidates with glioprotective properties for the management<br />

of primary open angle glaucoma (POAG).<br />

Methods: Primary adult rat optic nerve head astrocytes (ONHAs) were exposed<br />

to control ambient pressure or elevated hydrostatic pressure (25-30 mm Hg above<br />

ambient pressure) for 16 hr using a custom-built cell culture pressure chamber.<br />

ONHAs were subsequently challenged with chemically-induced oxidative stress<br />

using tert-butylhydroperoxide (tBHP; 0-500 µM for 5h).<br />

For proof-of-concept experiments, some ONHAs cultures were pre-treated with<br />

the prototypic antioxidant Trolox (100 µM). Cell viability was measured using<br />

MTT and LDH assays; levels of oxidative stress were quantified using the fluorescent<br />

indicator dye, CellROX®.<br />

Results: Elevated hydrostatic pressure did not alter cell viability, but significantly<br />

increased sensitivity to subsequent exposure to oxidative stress (LD50 for tBHP<br />

were 179±2µM vs. 84±1µM; n=3; P


POSTER BOARD N 10<br />

ALDH2 REGULATES ANGIOGENESIS<br />

GINEVRA NANNELLI 1 , ERIKA TERZUOLI 1 , MARINA ZICHE 1 AND SANDRA DONNINI 1<br />

1<br />

Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy<br />

Purpose: Maintenance of endothelial function is essential for the prevention and<br />

control of many diseases associated with deregulation of angiogenesis.<br />

Despite their having relatively little dependence on oxidative phosphorylation<br />

for ATP production, endothelial cells contain mitochondria.<br />

However, endothelial mitochondria are centrally involved in maintaining the<br />

fine regulatory balance between mitochondrial calcium concentration, reactive<br />

oxygen species (ROS) production, and NO. Recent findings have shown a prominent<br />

role of mitochondria in angiogenesis. However, how mitochondria affect<br />

the angiogenic function of endothelial cells is unknown.<br />

Aldehyde dehydrogenases (ALDHs) are a family of NADP-dependent enzymes<br />

with common structural and functional features that catalyze the oxidation of a<br />

broad spectrum of aldehydes.<br />

Experimental data from literature demonstrate that mitochondrial ALDH2 plays<br />

a role in tubulogenesis.<br />

In this study the aim was to evaluate the role of mitochondrial ALDH2 activity<br />

on HUVEC proangiogenic functions.<br />

Materials and methods: To investigate the role of ALDH2 activity on HUVEC<br />

pro-angiogenic functions, the contribution of<br />

ALDH2 activity on HUVEC sprouting and proliferation were investigated.<br />

Cells were pretreated with Daidzin, a selective ALDH2 inhibitor, in the presence<br />

or absence of a positive stimulus, and they were tested for their ability to migrate<br />

and growth using scratch assay and cell proliferation assay.<br />

Results and conclusions: The results show that ALDH2 inhibition with Daidzin<br />

reduces cell proliferation. Similarly, Daidzin<br />

impairs cell ability to migrate. Together these data support our hypothesis that<br />

ALDH2 inhibition causes angiogenic dysfunction.<br />

Acknowledgment: This work was supported by Associazione Ricerca sul Cancro (AIRC IG 15443).<br />

108


POSTER BOARD N 11<br />

PURINERGIC RECEPTOR MEDIATED INDUCTION OF INTERLEUKIN-1β IN<br />

MÜLLER AND MICROGLIAL CELLS IN RELATION TO GLAUCOMA<br />

JULIE SANDERSON 1 , MATTHEW FELGATE 1 , SOFIA HABIB 1,2 , PHILLIP WRIGHT 1 ,<br />

LEANNE STOKES 1 , NUWAN NIYADURUPOLA 2 AND DAVID C BROADWAY 1,2<br />

1<br />

School of Pharmacy, University of East Anglia, Norwich, UK<br />

2<br />

Department of Ophthalmology, Norfolk and Norwich University Hospital, Norwich, UK<br />

Purpose: IL-1β is a target for development of neuroprotective strategies. Its regulation<br />

is associated with signalling via the P2X7 receptor and we have shown previously<br />

that, as well as mediating loss of retinal ganglion cells (RGCs), activation<br />

of the P2X7 receptor caused an upregulation and release of IL-1β in human retina.<br />

The purpose of this research was to investigate purinergic receptor-mediated induction<br />

of IL-1β in glial cells using Müller and microglial cell lines.<br />

Method: Two cell lines were used: MIO-M1 (human retinal Müller cells) and BV2<br />

(mouse brain microglial cells). Cell viability and death were evaluated using MTS<br />

and LDH assays respectively. Induction of IL-1β was evaluated using RT-PCR (expression)<br />

and ELISA (release).<br />

Results: BV2 cells exhibited a dose-dependent decrease in viability/increase in death<br />

following a 24h exposure to ATP (100μM – 3mM). The P2X7 receptor antagonist<br />

AZ10606120 (200nM) protected cells from cell death due to ATP (3mM). MIO-M1<br />

cells exhibited no significant change in cell viability or death (24h) at the ATP concentrations<br />

used. LPS (0.5µg/ml) stimulation produced a strong induction at 24h of<br />

IL-1β mRNA in BV2, but not MIO-M1 cells. In BV2 cells, ATP (300µM) increased<br />

IL-1β mRNA expression after 24h, although no significant increased in secretion of<br />

mature protein was seen. No changes in IL-1β expression or release were observed<br />

in MIO-M1 cells treated for 24h with ATP (300µM).<br />

Conclusion: Müller and microglial cells exhibited differential responses to ATP.<br />

Understanding the pathway of ATP-mediated IL-1β regulation in microglia may<br />

provide insight into the mechanisms of RGC death in glaucoma.<br />

109


POSTER BOARD N 12<br />

AGE-RELATED DIFFERENCES AFTER BRIGHT LIGHT<br />

EXPOSUREIN BALB/C MICE<br />

SYMANTAS RAGAUSKAS 1,3 , TAMUNA BOLKVADZE 1 , AGNE ZINIAUSKAITE 1 ,<br />

HENRI O. LEINONEN 2,4 , HEIKKI TANILA 2 , GIEDRIUS KALESNYKAS 1<br />

1<br />

R&D, Experimentica Ltd, Kuopio, Finland,<br />

2<br />

Neurobiology, University of Eastern Finland, Kuopio, Finland<br />

3<br />

State Research Institute for Innovative Medicine, Vilnius, Lithuania<br />

4<br />

Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, USA<br />

Purpose: Exposure of BALB/c mice to bright light is an established preclinical<br />

model for the dry form of age-related macular degeneration. Here we tested the<br />

hypothesis that aged BALB/c mice show differential functional deficits after light<br />

damage compared to young mice.<br />

Methods: Three and seven months old male BALB/c mice (n=6) were exposed to<br />

bright light (10,000 lux) for 14 hours. Age- and gender matched controls (n=6)<br />

were kept under normal light conditions. Retinal function was evaluated on day 1<br />

and on day 7 after exposure to bright light using flash electroretinogram (fERG).<br />

Outer nuclear layer (ONL) thickness was measured using in vivo optical coherence<br />

tomography (OCT; Envisu R2200 system, Bioptigen Inc., NC, USA).<br />

Results: Functional measurements of both young and aged BALB/c mice showed<br />

a decrease in the a-wave amplitude and a b-wave amplitude as compared to naïve<br />

controls on day 1 after exposure to bright light. On the follow-up day 7 retinal<br />

function fully recovered in young mice, whereas the aged mice showed only<br />

partial recovery. Similarly, the aged mice showed higher decrease of the outer<br />

nuclear layer (ONL) thickness than that in young mice.<br />

Conclusions: Aged BALB/c mice are more susceptible to functional deficits and<br />

morphological retinal damage after exposure to bright light compared with<br />

young BALB/c mice. Our data provide new evidence for differential regulation<br />

of cell death mechanisms during aging.<br />

110


POSTER BOARD N 13<br />

CORNEAL SURFACE TEMPERATURE UNDER PERFLUOROHEXYLOCTANE<br />

EYE DROPS<br />

M. CARMEN ACOSTA, CAROLINA LUNA, SUSANA QUIRCE, ENRIQUE VELASCO,<br />

ADOLFO ARACIL, JUANA GALLAR<br />

Universidad Miguel Hernandez, Valencia, Spain<br />

Purpose: To compare corneal surface temperature measured from infrared video<br />

images (IRVI) before and after topical instillation of a single 10µl-drop of perfluorohexyloctane<br />

(PFHO) in guinea pigs of both sexes.<br />

Methods: Temperature values at central cornea (CST) and temporal conjunctiva<br />

(CJST) were measured before, 2 and 10 min after topical PFHO. IRVI were recorded<br />

(IR-thermal video camera InfRec R300SR, Nippon Avionics) and analyzed using<br />

dedicated software. CST and CJST were measured immediately after eye opening<br />

(T0) and 5s (T5) and 10s (T10) afterwards. Slopes of temperature decay (T0/T5 and<br />

T0/T10) were calculated. Tearing and blinking rate were also measured.<br />

Results: PFHO evoked a transient increase of blinking and tearing rate, and a mild<br />

conjunctival hyperemia. Two min after PFHO, T0-CST (36.9±0.1ºC vs 35.2±0.3ºC,<br />

before/after, n=10, *p


POSTER BOARD N 14<br />

INNER RETINAL CHANGE IN NORMAL-TENSION GLAUCOMA<br />

JIE HYUN KIM, HAE-YOUNG LOPILLY PARK, CHAN KEE PARK<br />

Department of Ophthalmology and Visual Science,<br />

College of Medicine, The Catholic University of Korea, Seoul, Korea<br />

Purpose: Normal-tension glaucoma (NTG) is known as an optic neuropathy<br />

characterized by progressive retinal ganglion cell death and glaucomatous visual<br />

field loss. NTG is reported to be related with systemic hemodynamic factors, such<br />

as fluctuating blood pressure and systemic hypotension. However, the mechanism<br />

how these factors contribute to glaucomatous damage at the optic nerve<br />

head and retina is unknown. In this study, we investigated the role of cell death<br />

mechanism of the retina in NTG.<br />

Methods: NTG induced to have systemic hypotension. Apoptosis of RGCs were<br />

examined by Terminal deoxynucleotidyl transferase-mediated dUTP nick-end<br />

labeling (TUNEL). Expression of various markers related to RGC apoptosis and<br />

glial cell activation were analyzed by western blot analysis and immunohistochemical<br />

staining of the retina.<br />

Results: IOP elevation is not detected. We have analyzed the loss of Brn3a-positive<br />

RGCs and TUNEL-positive cells were detected in the ganglion cell layer.<br />

Expression of glial fibrillary acidic protein was increased throughout the retinal<br />

layer.<br />

Conclusions: These findings suggest that systemic hemodynamic factors may<br />

contribute to the changes of astrocyte and muller cells in retina cell death without<br />

elevated IOP. The role of this phenomenon needs further investigation.<br />

112


POSTER BOARD N 15<br />

ANTERIOR CHAMBER VERSUS POSTERIOR CHAMBER PERFUSION IN<br />

LIVING MICE DOES NOT INFLUENCE MEASUREMENT OF<br />

AQUEOUS OUTFLOW FACILITY BY CONSTANT FLOW INFUSION<br />

J. CAMERON MILLAR, NAVITA N. LOPEZ, GAURANG C. PATEL, TIEN N. PHAN AND ABBOT F. CLARK<br />

North Texas Eye Research Institute, University of North Texas Health Science Center, 3500<br />

Camp Bowie Boulevard, Fort Worth, TX USA<br />

Purpose: In ex-vivo mouse eyes, values for facility (C) are variable depending<br />

upon whether perfusate is introduced to the anterior chamber (AC) vs. the posterior<br />

chamber (PC). AC perfusion causes posterior iridial bowing, pupil block, and<br />

scleral spur traction, increasing C. PC perfusion does not yield this effect resulting<br />

in a lower value for C. We investigated if AC vs. PC perfusion of the living<br />

mouse eye similarly influences C.<br />

Methods: C57-BL/6J mice ( ) (20-24 weeks) were divided into 4 groups (4 animals/group).<br />

C was measured (constant flow infusion (OU)) from a 50 µL syringe.<br />

In Groups 1 and 2 a 30G needle was placed in the AC and PC, respectively.<br />

To investigate the effect of ciliary muscle (CM) and iridial tone on C, Groups 3 and<br />

4 were perfused (AC or PC, respectively) with tropicamide added to the perfusate<br />

(100 µM).<br />

Results: C in Groups 1 (AC) and 2 (PC) was 22.6 ± 2.4 vs. 23.1 ± 2.4 nL/min/mmHg,<br />

respectively (mean ± SEM, P = 0.869). Tropicamide induced cycloplegia (confirmed<br />

by histology) and mydriasis (confirmed visually). C in Group 3 (AC (tropicamide))<br />

was greater than C in Group 4 (PC (tropicamide)) (22.0 ± 4.0 vs. 13.6 ± 1.6<br />

nL/min/mmHg, respectively, P = 0.0341).<br />

Conclusions: C in living mice is not different when measured via AC or PC perfusion.<br />

However in cycloplegic/mydriatic mouse eyes C is greater when eyes are<br />

perfused via the AC. CM and possibly also iridial tone plays a role in establishment<br />

of C.<br />

113


POSTER BOARD N 16<br />

NOVEL TARGETS OF δ-OPIOID RECEPTOR AGONIST FOR RGC<br />

NEUROPROTECTION<br />

SHAHID HUSAIN<br />

Medical University of South Carolina, USA<br />

Purpose: This study is designed to determine the potential downstream targets of<br />

δ-opioid receptor that are involved in RGC neuroprotection against glaucomatous<br />

injury.<br />

Methods: Brown Norway rats were used to elevate intraocular pressure (IOP)<br />

by injecting 50 µL of 2M hypertonic saline into the circumferential limbal veins.<br />

IOP was recorded as the average of 6-8 consecutive measurements prior to surgery<br />

(baseline IOP) and weekly after treatment, using a calibrated Tonolab tonometer.<br />

Animals were treated with delta opioid-receptor agonist, SNC-121 (1<br />

mg/kg; i.p) daily for 7 days. Pattern electroretinograms (PERG), retinal ganglion<br />

cells (RGCs) in flat mount, and axons were counted 4-6 week post injury. The<br />

changes in the neurotrophins and cytokines were measured by 84-gene RT profilerTM<br />

PCR array kit. Additionally, the cAMP levels and phospho-CREB were<br />

measured by ELISA, Western blotting, and immunohistochemistry.<br />

Results: We have found that 7-days administration of a δ-opioid-receptor agonist<br />

resulted in significant long-term (42 days) neuroprotection in a chronic glaucoma<br />

model. This long-term neuroprotective response supports the idea that opioids<br />

induce epigenetic changes in the retina and optic nerve allowing RGCs to maintain<br />

their functional integrity under conditions that normally lead to progressive<br />

neuronal loss. We found that chronic δ-opioid administration increases the level<br />

of histone-H3 acetylation, stimulates cAMP/CREB signaling, and eventually increasing<br />

neurotrophic factors expression. The level of cAMP was decreased by<br />

32% in the ocular hypertensive eyes, but significantly increased in δ-opioid-receptor<br />

agonist (SNC-121) treated normal and ocular hypertensive eyes. It is important<br />

that chronic treatment with a δ-opioid agonist for 7 days increased the<br />

levels of cAMP at day 7 and cAMP production was further elevated significantly<br />

at days 14 and 42, while SNC-121 treatment had been stopped at day 7. The phosphorylation<br />

of CREB (a downstream target of cAMP) was also significantly reduced<br />

in ocular hypertensive eyes at day 42, which was also significantly (p


POSTER BOARD N 17<br />

MAPRACORAT, A NOVEL SELECTIVE GLUCOCORTICOID RECEPTOR<br />

AGONIST, INHIBITS CYTOKINE SECRETION IN HUMAN MAST CELLS<br />

MONICA BAIULA 1 , SANTI M. SPAMPINATO 1<br />

1<br />

Dept. of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy<br />

Purpose: Mapracorat, a glucocorticoid receptor agonist (SEGRA), has been proposed<br />

for the topical treatment of inflammatory eye disease. Data from in vitro<br />

and in vivo studies suggest an improved side-effect profile of this compound<br />

compared to classical glucocorticoids. In previous studies we demonstrated that<br />

mapracorat increased eosinophil apoptosis and reduced eosinophil migration in a<br />

model of allergic conjunctivitis. To further explore the mechanism of action of<br />

mapracorat, this study assessed its effects, in comparison with dexamethasone, on<br />

cytokine secretion in mast cells as these can greatly influence eosinophil activity<br />

in inflamed ocular tissues. In fact, a vast number of cytokines synthesized by mast<br />

cells can influence inflammatory eye diseases as well as eosinophil biology.<br />

Methods: Luminex technology was used to determine the effect of mapracorat<br />

(0.1-1-10µM) on ionomycin-induced cytokine release levels in human mast cell<br />

line (HMC-1). Dexamethasone was used as comparison. We focused on mast cell<br />

cytokines that are responsible for initiation, orchestration and support of the allergic<br />

reaction.<br />

Results: Ionomycin induced multiple cytokine release in human mast cells. Mapracorat<br />

significantly reduced ionomycin-induced inflammatory cytokine release (including<br />

IL-6, IL-8, IL-1ra, IP-10, MCP-1, MIP1α, MIP1β and TNF-α) in a dose-dependent<br />

manner. Mapracorat showed efficacy superior to dexamethasone.<br />

Conclusions: Data from this in vitro model indicate that mapracorat is efficacious<br />

and potent in blocking the release of the majority of pro-inflammatory cytokines<br />

from mast cells. This effect together with its effects on eosinophils suggests that<br />

it may provide a new option for the treatment of ophthalmic conditions with an<br />

inflammatory component.<br />

115


POSTER BOARD N 18<br />

CIPROXIFAN, AN H3 RECEPTOR INVERSE AGONIST, LOWERS IOP AND<br />

AMELIORATES OCULAR VASCULAR REACTIVITY IN NEW ZEALAND WHITE<br />

RABBIT MODELS OF GLAUCOMA<br />

CECILIA LANZI 1 , LAURA LUCARINI 1 , MARIACONCETTA DURANTE 1 , ALESSANDRO PINI 2 ,<br />

FRANCESCO IMPAGNATIELLO 3 , ELENA BASTIA 3 , HOLGER STARK 4 AND EMANUELA MASINI 1<br />

1<br />

Department of NEUROFARBA, Section of Pharmacology,<br />

2<br />

Department of Experimental and Clinical Medicine, University of Florence, Italy<br />

3<br />

Nicox Research Institute, Bresso, Milan, Italy<br />

4<br />

Heinrich-Heine Düsseldorf University, Institute of Medicinal Chemistry, Düsseldorf, Germany<br />

Previous evidences suggest that H3R receptors are expressed in ocular structures.<br />

However, their involvement in the regulation of intraocular pressure (IOP) and<br />

ocular blood flow has only sporadically been investigated.<br />

We report on the IOP-lowering effects resulting from application of ciproxifan,<br />

a well known H3R receptor inverse agonist, at different concentrations (0.1, 0.3,<br />

0.5, 1%) in rabbits with transient or stable ocular hypertension induced by the<br />

injection of hypertonic saline (100 µl, 5% tOHT-rabbits) or carbomer (100 µl, 0.1%<br />

stable OHT-rabbits), respectively.<br />

Ecocolor Doppler was used to evaluate changes in retinal artery resistance index<br />

(RI) before and after repeated dosing. Finally, we investigated the expression<br />

of histamine receptors in naïve rabbit ciliary bodies, retinae and optic nerve by<br />

Western blot as well as by immunofluorescence staining.<br />

IOP rose from 16.4±3.2 mmHg to 39.4±4.8 mmHg in tOHT-rabbits and from<br />

14.2±5.3 to 36.8±5.6 in stable-OHT-rabbits starting four days after carbomer injection.<br />

Ciproxifan dose-dependently reduced IOP at 60min in tOHT rabbits.<br />

Similarly, IOP and RI of the retinic artery were significantly reduced in animal<br />

treated with 1% and 0.5% ciproxifan in stable OHT-rabbits. Western blot analysis<br />

as well as immunofluorescence staining demonstrated the presence of histamine<br />

H1 and H3 receptors in ciliary bodies, retinae and optic nerve of naïve rabbits.<br />

The expression of these receptors was also detected in cultured trabecular meshwork<br />

cells. Ciproxifan effectively lowers IOP and ameliorates the vascular performance<br />

of the retinic artery.<br />

116


POSTER BOARD N 19<br />

EFFICACY OF A NEW FOOD SUPPLEMENT IN A MURINE MODEL<br />

OF OPTICNEURITIS<br />

DARIO RUSCIANO 1 , MAURIZIO CAMMALLERI 2 , FILIPPO LOCRI 2 , MASSIMO DAL MONTE 2<br />

AND PAOLA BAGNOLI 2<br />

1<br />

Sooft Italia, Montegiorgio, Italy; 2Department of Biology, University of Pisa, Italy<br />

Purpose: Optic neuritis is an inflammatory, demyelinating condition that causes<br />

acute visual loss. Corticosteroidsare the standard treatment for optic neuritis. Aim<br />

of this work was to investigate in a murine model of optic neuritisthe efficacy<br />

of an innovative food supplement orally administered during the course of the<br />

disease.<br />

Methods: C57BL/6 mice were immunized with myelo-oligodendrocyte glycoprotein35-55(MOG<br />

35-55) to induce an experimental autoimmune optic neuritis.<br />

A mixture of fatty acids and lycopene diluted in sugar-water was given by daily<br />

gavage to animals fed with a standard diet, starting on the day of MOG(35-55)<br />

administration and continuing until their sacrifice, 16 days later. Control animals<br />

received gavage treatment with sugar-water only. Markers of inflammation and<br />

macrophage infiltration were investigated in the retina and the optic nerve by<br />

mRNA and protein expression.<br />

Results: Sixteen days after MOG(35-55) administration a significant increase in<br />

markers of inflammation (TNFα, IL1β, IL6, IL7, IL8, GFAP, ICAM1 and iNOS)<br />

and macrophage infiltration (CD68, F4/80 and oncomodulin) was measured in<br />

the retina. In the optic nerve TNFα, IL1β, IL6 and IL8 were undetectable while<br />

the other markers showed an increase after MOG(35-55)administration. Both in<br />

the retina and in the optic nerve food supplementation resulted in a significant<br />

reduction of the inflammatory markers but not of the markers of macrophage<br />

infiltration.<br />

Conclusions: Our data show that oral administration of food supplement is able to<br />

limit the production of inflammatory markers in a mouse model of optic neuritis<br />

thus suggesting this formulation a useful tool to slow down inflammatory processes<br />

associated to the demyelination of optic nerve.<br />

117


POSTER BOARD N 20<br />

THE WNT SIGNALING PATHWAY IN TRABECULAR MESHWORK CELLSCAN<br />

BE MODULATED BY EXOSOMES DERIVED FROM NON-PIGMENTED CILIARY<br />

EPITHELIAL CELLS<br />

ELIE BEIT-YANNAI 1 , SOFIA AVISSAR 1 , NATALIE LERNER 1<br />

1<br />

Clinical Biochemistry and Pharmacology, Ben-Gurion University, Beer-Sheva, Israel<br />

Purpose: Cross talk between the ocular drainage system tissues contributes to the<br />

intraocular pressure homeostasis in health and disease. The present study aims to<br />

uncover exosomes as signaling mediators in the system<br />

Methods: Exosomes extracted from non-pigmented ciliary epithelia cell line<br />

(ODM2) were characterized for size zeta potential, miRNA and protein content<br />

using TRPS, GS-MSMS, Western Blot, microarray methods and, Image stream,<br />

confocal and electron microscopy analysis. Normal trabecular meshwork cells<br />

line (NTM5) were incubated with ODM derived exosomes for various periods of<br />

time and the modulation of Wnt signaling pathway was analyzed.<br />

Results: ODM2 derived exosomes were purified and detected as small rounded<br />

50-140 nm membrane vesicles positive for classic exosome markers, FLOT1,<br />

ICAM, CD81, CD63, ANAXA5, TSG101 and Alix. Using confocal microscopy, we<br />

demonstrated time-dependent specific accumulation of ODM2-derived exosomes<br />

in NTM5 cells. ODM2 exosomes induced significant decreased phosphorylation<br />

of GKS3β and reduced β-catenin levels expression in the NTM5 cells.<br />

Endogenous expression of Wnt-regulated genes in NTM cells, Axin2 and Lif1,<br />

were significantly reduced at 2h. Furthermore, treatment of NTM5 cells with<br />

ODM2 derived exosomes resulted in a significant decrease in pan-Cadherin expression<br />

at 12h and 24h.<br />

Conclusions: The data suggest that ODM2 cells release exosome-like vesicles and<br />

that these nanoparticles affect canonical Wnt signaling in NTM5 cells. These<br />

findings might have therapeutic relevance since canonical Wnt pathway is involved<br />

in intra-ocular pressure regulation.<br />

118


POSTER BOARD N 21<br />

EXPRESSION OF OCULAR SURFACE MUCIN IN DRY EYE INDUCED MOUSE<br />

MODEL BY CURRENT DRY EYE TOPICAL MEDICATIONS<br />

INHEE MOON 1 ,YEO AREUM 1 , NOH HAEMI 1 , HYUN CHANG KIM1, 2 , JONG SUK SONG 3 ,<br />

HYUNG KEUN LEE1, 4<br />

1<br />

Institute of Vision Research, Department of Ophthalmology,<br />

Yonsei University College of Medicine, Seoul, Korea<br />

2<br />

Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Korea<br />

3<br />

Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea<br />

4<br />

Institute of Corneal Dystrophy Research, Department of Ophthalmology,<br />

Yonsei University College of Medicine, Seoul, Korea<br />

Purpose: Mucin is a component of tear fluid and is essential to the wettability of<br />

the ocular surface. In this study, we aimed to evaluate the effects of 4 different<br />

dry eye medications on mucin subtype (MUC1, 4, 16, 5AC) levels in a dry eye-induced<br />

mouse model.<br />

Methods: C57BL/6 mice were separated into 6 groups: the control, dry eye-induced<br />

group, and four groups that respectively used cyclosporin A 0.05%, rebamipide<br />

2%, diquafusol 3%, prednisolone 1% twice a day. MUC 1, 4, 5ac, 16 and<br />

inflammatory cytokines were estimated through Q-PCR from the conjunctival<br />

cells and corneal epithelial cells. Through PAS staining, IHC, conjunctival goblet<br />

cells were analyzed. FACS staining was done for evaluating MUC1, 5ac, 16 level.<br />

Results: By Q-PCR, the expression of MUC1, 4 mRNA were significantly elevated<br />

in the diquafusol group(p


POSTER BOARD N 22<br />

PKCβ INHIBITION IMPAIRS VEGF INDUCED OCULAR ANGIOGENESIS<br />

LUCIA MORBIDELLI, MARTINA MONTI, DARIA MOCHLY-ROSEN 1 , MARINA ZICHE<br />

Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy<br />

1<br />

Department of Chemical and Systems Biology,<br />

Stanford University School of Medicine Stanford, CA 94305, USA<br />

Purpose: A crucial role for Protein Kinase C (PKC) β has been demonstrate in<br />

promoting angiogenesis, a process important for corneal neovascularization,<br />

age-related macular degeneration (AMD) and diabetic retinopathy. We aimed to<br />

evaluate the antiangiogenic potential of novel inhibitors of PKCβ1 and 2 isoforms<br />

on VEGF-induced neovascularization in vivo, in the rabbit cornea assay.<br />

Methods: VEGF at a fixed dose (200 ng) was enclosed in slow release pellets and<br />

implanted in the cornea of albino rabbits. Compounds to be tested were embedded<br />

together with VEGF in a single pellet (single pellet experiments) or separate adjacent<br />

pellets. In both experimental protocols, compounds were co-released with<br />

the angiogenic factor and delivered to the cornea simultaneously, or alternatively<br />

the inhibitor was given during neovascular growing. In vitro experiments<br />

were conducted on microvascular endothelial cells to evaluate the functional and<br />

intracellular effect of the inhibitors.<br />

Results: Our data demonstrate the efficacy of PKCβ inhibitors in reducing VEGF<br />

induced neovascularization. Both β1 and β2 antagonists were effective, being the<br />

antiangiogenic effect elicited by the β1 selective inhibitor stronger. Moreover,<br />

the addition of PKCβ inhibitor 5 days after the implant of VEGF bearing pellets<br />

was able to impair neovascular progression. Data on endothelial cell migration,<br />

proliferation and organization strengthened the antiangiogenic activity of the<br />

PKCβ1 selective inhibitor, though the impairment of VEGF induced Akt (but not<br />

ERK1/2) signalling.<br />

Conclusions: Based on these data, we propose the development of PKCβ inhibitors<br />

as antiangiogenic strategies in angiogenesis-dependent ocular diseases.<br />

120


POSTER BOARD N 23<br />

ANTIANGIOGENIC AND ANTI-INFLAMMATORY ACTIVITY OF UPARANT IN<br />

THE RABBIT CORNEA<br />

VALERIO CICCONE, LORENZO BAZZANI, DARIO RUSCIANO 1 , VINCENZO PAVONE 2 ,<br />

MARIO DE ROSA 3 , MARINA ZICHE AND LUCIA MORBIDELLI<br />

Dept. Life Sciences, Univ. Siena, Italy; 1 Sooft Italia Spa, Montegiorgio, Italy<br />

2<br />

Department of Chemical Science, University of Naples “Federico II” via Cintia, 80126<br />

Napoli, Italy, 3 Department of Experimental Medicine, Second University of Naples, Napoli, Italy<br />

Purpose: Diseases involving pathologic neovascularization of the eye are represented<br />

by proliferative retinopathies including retinopathy of prematurity, diabetic<br />

retinopathy, and age-related macular degeneration or corneal angiogenesis.<br />

The angiogenic phenotype is often associated with inflammation.<br />

During the neovascularization process the urokinase-type plasminogen activator<br />

(uPA)/uPA receptor (uPAR) system sustains the proteolytic degradation of the extracellular<br />

matrix by sprouting endothelial cells. The inhibitory activity of Ac-<br />

L-Arg-Aib-L-Arg-α(Me)Phe-NH2 (UPARANT), a modified peptide derived from<br />

uPAR and able to interfere with uPAR/integrin, has been reported on vascular<br />

endothelial growth factor (VEGF)-driven angiogenesis in different in vitro and<br />

in vivo models.<br />

The purpose of this study was to assess the antiangiogenic and anti-inflammatory<br />

activities of UPARANT in the rabbit cornea assay when given in the formulation<br />

of ocular drops.<br />

Methods: Angiogenesis was evaluated as neovascular growth induced by VEGF<br />

slow release in the cornea stroma, while an inflammatory response was induced<br />

by alkali burns. Ocular tissues were recovered to measure inflammatory markers.<br />

Results: The local administration of UPARANT significantly inhibited VEGF<br />

induced neovascularization and alkali burn associated inflammation of the conjunctiva,<br />

sclera and cornea tissues. At tissue level a reduction of cyclooxygenase-2<br />

expression and PGE2 production were associated to UPARANT treatment.<br />

Conclusions: Taken together, these data indicate that UPARANT may represent<br />

a promising therapeutic agent for local use in angiogenesis- and inflammation-dependent<br />

diseases of the eye.<br />

121


POSTER BOARD N 24<br />

LIGHT-INDUCIBLE RHODOPSIN MUTANTS (TVRM4/+) MICE:<br />

CHARACTERIZATION AND THERAPEUTIC APPROACH<br />

ILARIA PIANO 1 , CLAUDIA GARGINI 1 , ELENA NOVELLI 2 , MARTINA BIAGIONI 2,4 , FABIOLA BONEZZI 3 ,<br />

GIUSEPPE CAMPISI 3 , RICCARDO GHIDONI 3 , AND ENRICA STRET TOI 2<br />

1<br />

Department of Pharmacy, University of Pisa, Italy<br />

2<br />

CNR Neuroscience Institute, Pisa, Italy<br />

3<br />

Biochemistry and Molecular Biology Laboratory, Health Sciences Department,<br />

University of Milan, San Paolo Hospital Medical School, Milan, Italy<br />

4<br />

Tuscan Doctorate School in Neuroscience, University of Pisa<br />

Purpose: Rhodopsin (RHO) mutations are responsible for 25-40% of the dominant<br />

cases of Retinitis Pigmentosa (RP). Tvrm4/+ mice, heterozygous for a I307N<br />

dominant mutation of RHO, display a normal retinal phenotype when raised in<br />

ambient light conditions but undergo photoreceptor degeneration when briefly<br />

exposed to strong, white light.<br />

Here, we induce a retinal phenotype in Tvrm4/+ mice and treat the animals<br />

with intra-vitreal injections of Myriocin, a serine-palmitoyl-transferase inhibitor<br />

shown to delay retinal degeneration in rd10 mice. These mimick autosomal<br />

recessive RP caused by a phosphodiesterase mutation.<br />

Methods: Tvrm4/+ mice aged 2-4 months were pupil-dilated and exposed to<br />

white light pulses (12,000 lux) for 1 or 2 min. 24h post-induction, Myriocin (1.9<br />

mM) was injected intravitreally in one group of mice; controls were injected<br />

with vehicle alone. 48h after light-induction, mice were tested for scotopic and<br />

photopic ERGs, retinal morphology and LC/MS spectrometry assays on retinal<br />

samples.<br />

Results: Light-induction of Tvrm4/+ mice triggers a typical rod-cone degeneration<br />

in an area concentric to the optic disc. Rod outer segments shorten after<br />

48h. Cones degenerate more slowly. Myriocin injection a) decreased the number<br />

of apoptotic cells in the outer retina; b) lowered retinal levels of de-novo synthesized<br />

ceramide species (C18 and C21); c) improved scotopic ERG responses.<br />

Conclusions: Inhibition of ceramide de novo synthesis by ocular Myriocin is<br />

effective in counteracting photoreceptor degeneration also in a RHO dominant<br />

model of RP, disclosing mutation-independent perspectives for the treatment of<br />

this disease.<br />

Funded by: Macula Vision Research Foundation (USA) and Fondazione Roma (Italy).<br />

122


POSTER BOARD N 25<br />

TRPV4 STIMULATION INDUCED ARALKYLAMINE N-ACETYLTRANSFER-<br />

ASE (AANAT) PHOSPHORYLATION AND MELATONIN PRODUCTION VIA<br />

CA-CALMODULIN PATHWAY IN HUMAN CILIARY BODY EPITHELIAL CELLS<br />

JESÚS PINTOR, HANAN AWAD ALKOZI AND MARIA J. PEREZ DE LARA<br />

Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry,<br />

Universidad Complutense de Madrid, Madrid, Spain<br />

Purpose: To study the regulation by phosphorylation of aralkylamine N-acetyltransferase<br />

(AANAT) activating the TRPV4 channel present in human ciliary<br />

body epithelial cells, measuring also melatonin production.<br />

Methods: Non-pigmented ciliary epithelial cells were supplied by Dr. Coca-Prados.<br />

Cells were stimulated with TRPV4 agonists and antagonist (such as GS-<br />

K1016790A or RN-1734), and the expression of p-AANAT was measured by western-blot<br />

(ab3439, 1:1000, Abcam). Melatonin concentrations were quantified by<br />

HPLC equilibrating the system with 40% methanol, 60% H2O, measuring at a<br />

flow rate of 0.8 ml/min and at a wavelength of 244nm.<br />

Results: First it was determined the adequate time and dose of the TRPV4 agonist<br />

GSK1016790A to reach the maximal phosphorylation of AANAT. An increase<br />

of 36% was observed after 5 minutes incubation with 10nM GSK (**p


POSTER BOARD N 26<br />

INTRAPERITONEAL INJECTION OF AN ANTI-APOPTOTIC PEPTIDE INHIBITS<br />

RETINAL GANGLION CELL DEATH IN ANIMAL MODELS OF GLAUCOMA<br />

RAM H. NAGARAJ 1 , RAGHU R. KRISHNAMOORTHY 2 , SRUTHI SAMPATHKUMAR 3<br />

AND DOROTA L. STANKOWSKA 2<br />

1<br />

Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045<br />

2<br />

North Texas Eye Research Institute, UNT Health Science Center, Fort Worth, TX 76107 and<br />

3<br />

Department of Ophthalmology and Visual Sciences, Case Western Reserve University<br />

Cleveland, OH 44106<br />

Purpose:Axonal degeneration and death of retinal ganglion cells (RGC) are primary<br />

contributors to vision loss in glaucoma. In animal models of glaucoma,several<br />

approacheshave been shown to prevent or delay RGC apoptosis, including<br />

pharmacological agents and gene therapy. The purpose of this study was to determine<br />

ifintraperitoneal administration of the core peptide derived from small heat<br />

shock protein αB-crystallin (ABCP) could inhibit RGC death in animal models of<br />

glaucoma.<br />

Methods:Brown Norway rats were retrogradely labeled (to detect RGCs) using<br />

Fluoro-gold and IOP was elevated (150 mmHg/days) in one eye using the Morrison’s<br />

method, while the contralateral eye served as control. The rats were intraperitoneally<br />

injected with 10μg of ABCP (n=3 animals per group) three times<br />

per week for five weeks. Surviving RGCs were counted in retinal flat mounts.<br />

In the model of ischemia reperfusion (I/R) injury,C57BL/6 micewere subjected<br />

to IOP elevation of 120 mmHg for 30 min, followed by rapid reperfusion. Intraperitoneal<br />

ABCP injections (10 μg/animal) were given 3h before and immediately<br />

after the procedure and then once daily post I/R injury for 14 days. RGC apoptosis<br />

was assessed using TUNEL kit according manufacturer protocol (n=2 animals per<br />

group).Intraperitoneal administration of scrambled peptide was used as negative<br />

control in the Morrison’s model.<br />

Results:Intraperitoneal injections of ABCP significantly (p


POSTER BOARD N 27<br />

MICROARRAY TRANSCRIPTOME ANALYSIS OF CORNEA AND LACRIMAL<br />

GLAND OF IL-22 KNOCK-OUT AND LYMPHATIC HYPOPLASIA TRANSGENIC<br />

MOUSE MODELS<br />

EUN YOUNG CHOI, MD 1 , HYUN GOO KANG, MD 1 , AREUM YEO1, SO YI JUNG 2 , HYUNG KEUN LEE, MD 1 ,<br />

1<br />

Department of Ophthalmology, Yonsei University College of Medicine<br />

Seoul, Republic of Korea; 2 Macrogen Inc. Seoul, Korea<br />

Purpose: This study investigated the gene expression of corneal and lacrimal<br />

gland tissues from transgenic models with knocked out IL-22 expression and induced<br />

lymphatic hypoplasia.<br />

Method: Tissue samples from cornea and lacrimal glands of ILL-22 knock-out-<br />

(IL-22 KO: IL22 Cre ) and delta-LV(∆LV: Lyve-1 wt/Cre ;Vegfr2 flox/flox ) transgenic mice<br />

were used to compare with wild type control mice. Microarray chips (Affymetrix<br />

GeneChip® Mouse Gene 2.0 ST Array) were used for initial analysis and the results<br />

were compared with Affymetrix® Expression Console Software to obtain<br />

gene expression levels with significant fold changes(FC) greater than twice the<br />

control. Genes that were found significant were then imputed into the Database<br />

for Annotation, Visualization and Integrated Discovery(DAVID) for functional<br />

annotation and gene-enrichment analysis.<br />

Results: Comparison of IL-22 KO and control mice revealed 12 genes related to<br />

oxidation, 23 genes related to ocular development and maturation, 86 genes related<br />

to lens formation from corneal tissues, and 231 genes related to the immune response<br />

in addition to 122 genes related to signal transduction from lacrimal gland<br />

samples (|FC|≥2, p


POSTER BOARD N 28<br />

REGULATION OF INTRAOCULAR PRESSURE BY MICRORNA<br />

CLUSTER MIR-143/145<br />

JING MA 1 , XINYU LI 2 , MEI XIN 3 , PEDRO GONZALEZ 4 AND SHUSHENG WANG 1, 5<br />

1<br />

Department of Cell and Molecular Biology,<br />

2<br />

Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of<br />

Science and Technology, 1095 Jiefang Road, Wuhan, Hubei 430030, People's Republic of China<br />

3<br />

Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati,<br />

Cincinnati 45247, OH, USA<br />

4<br />

Department of Ophthalmology, Duke University, Durham, North Carolina, USA<br />

5<br />

Department of Ophthalmology, Tulane University, New Orleans, LA, 70118, USA<br />

Purpose: Elevated intraocular pressure (IOP), which causes optic nerve damage<br />

and retinal ganglion cell death, is a primary risk factor for blindness in glaucoma<br />

patients. IOP is controlled by the balance between aqueous humor secretion by<br />

the ciliary body (CB) and its drainage by trabecular meshwork (TM). The purpose<br />

of the project is to determine how microRNAs (miRNAs) regulate IOP and<br />

glaucoma in vivo.<br />

Methods: Reporter mice were used to determine the expression of miR-143/145 in<br />

the eye. IOP was measured in miR-143/145 knockout mice under normal and experimental<br />

glaucoma conditions. Moreover, the regulation of outflow facility and<br />

trabecular meshwork contractility by miR-143/145 was examined, and the target<br />

genes that potentially mediate the function of miR-143/145 were confirmed.<br />

Results: Here we provide genetic evidence that miRNAs are important IOP regulators<br />

in vivo. Specifically, we found that miR-143/145 are expressed in smooth<br />

muscle, pericytes and trabecular meshwork in the eye. miR-143/145 knockout<br />

mice showed significantly reduced IOP, consistent with ~2-fold increase in outflow<br />

facility. However, in an experimental glaucoma mouse model, the IOP increase<br />

in miR-143/145 knockout mice was similar to the wildtype control mice.<br />

Mechanistically, we found that miR-143/145 regulate the contractility of TM<br />

cells, consistent with its regulation of Actin-related protein 2/3 complex subunit<br />

2, 3 and 5 and Myosin light chain kinase (MLCK) in these cells.<br />

Conclusions: We found that miR-143/145 are important regulators of IOP by regulating<br />

the contractility of trabecular meshwork. Manipulating miR-143/145 levels<br />

may have important therapeutic implications in controlling IOP in glaucoma<br />

patients.<br />

126


POSTER BOARD N 29<br />

ADVANCED ANTAGONIST OF RETINOL-BINDING PROTEIN 4 FOR TREATMENT<br />

OF THE ATROPHIC FORM OF AGE-RELATED MACULAR DEGENERATION<br />

KONSTANTIN PETRUKHIN<br />

Department of Ophthalmology, Columbia University, New York, NY 10032, USA<br />

Purpose: Dry AMD is a multifactorial disorder with several pathogenic factors<br />

contributing to the disease progression. Here we report identification of the advanced<br />

non-retinoid RBP4 antagonist with the desired attributes of a potential<br />

clinical candidate and describe its characterization in relevant animal models<br />

mimicking important aspects of dry AMD.<br />

Methods: The effect of compound dosing on lipofuscin bisretinoid synthesis was<br />

assessed in the Abca4-/- mice as well as in the double knock-out Abca4-/-Rdh8-/-<br />

mouse model following 3 months of dosing. Normalization of complement system<br />

dysregulation was assessed in Abca4-/- mice after 3 months of compound<br />

administration using immunoblot analysis of retinal extracts as well as immun<br />

fluorescence analysis of retinal sections to determine expression levels for C3/<br />

C3b, CFH, CFD, MCP-1 as well as for C-reactive protein. The protective effect of<br />

compound dosing against combined bisretinoid and retinaldehyde toxicity was<br />

evaluated in the double knock-out Abca4-/-Rdh8-/- model by assessing ERG parameters<br />

and measuring anatomical preservation of photoreceptor cells.<br />

Results: Compound administration induced drastic inhibition of lipofuscin bisretinoid<br />

synthesis in eyecups of the Abca4-/- mice as well as in the retinas of<br />

double knock-out Abca4-/-Rdh8-/- mice. Compound administration inhibited<br />

photoreceptor degeneration and suppressed ERG attenuation in the Abca4-/-<br />

Rdh8-/- model. Compound dosing normalized the expression of dysregulated<br />

complement system components in the retinas of Abca4-/- mice.<br />

Conclusions: The in vivo efficacy profile of the advanced drug candidate indicates<br />

that the compound may be considered as a potential therapy for dry AMD<br />

and Stargardt disease.<br />

127


POSTER BOARD N 30<br />

UNIQUE HYDROGEL TECHNOLOGY - IN VITRO MODEL REPRESENTING<br />

CORNEAL LAYERS<br />

AGNĖ ŽINIAUSKAITĖ, VYTAUTAS CEPLA, RAMŪNAS VALIOKAS, GIEDRIUS KALESNYKAS<br />

AND JENNI J. HAKKARAINE<br />

Experimentica Ltd, R&D department, Microkatu, Finland<br />

Purpose: The cornea is an effective penetration barrier to drugs applied topically<br />

onto the eye. In early drug development, it is important to evaluate the potency<br />

of a drug candidate for its ability to permeate through the cornea. The purpose of<br />

this study was to develop an in vitro model representing the corneal component<br />

layers (epithelium and stroma) using a unique hydrogel technology and human<br />

corneal epithelial cells (HCE-T).<br />

Methods: Different hydrogel components and cross-linking techniques were used.<br />

The apparent permeability coefficient (Papp) values of low and high permeability<br />

marker molecules across the blank hydrogels and hydrogels with HCE-T cells on<br />

top of hydrogels were measured. Expression and localization of tight junction proteins,<br />

ZO-1 and occludin, were assessed using immunocytochemistry.<br />

Results: Papp values were significantly lower for hydrogels with HCE-T cells<br />

cultured on top of the hydrogel than the Papp values for blank hydrogels. However,<br />

there were differences in the expression and localization of tight junction<br />

proteins depending on the hydrogel type where the cells were grown.<br />

Conclusions: The use of hydrogel technology is a promising model for the corneal<br />

stroma. However, additional development is needed to obtain an in vitro<br />

model comprising all functional corneal layers and possessing adequate epithelial<br />

barrier function.<br />

128


POSTER BOARD N 31<br />

HSP27 ADDITIONINTENSIFIES AII AMACRINE CELL AND SYNAPSE DAMAGE<br />

INDUCED BY S100BIMMUNIZATION IN AN AUTOIMMUNE<br />

GLAUCOMA MODEL<br />

STEPHANIE C. JOACHIM, SABRINA REINEHR, SANDRA KUEHN, CHRISTINA CASOLA,<br />

DENNIS KOCH, GESA STUTE, H. BURKHARD DICK<br />

Experimental Eye Research Institute, University Eye Hospital,<br />

Ruhr-University Bochum Bochum, Germany<br />

Purpose: Previous studies revealed a loss of retinal ganglion cells (RGCs) and optic<br />

nerve fibers after immunization with the S100 protein. An addition of heat shock<br />

protein 27 (HSP27) lead also to a decrease of RGCs. In the current study, we aimed<br />

to analyze the effect on retinal cells.<br />

Methods: Rats were immunized with S100B or S100+HSP27, IOP was measured<br />

throughout the study. After 4 weeks, retinas were processed for immunohistology<br />

and Western Blot analysis. Retinal ganglion cells (Brn-3a), amacrine cells<br />

(ChAT), macroglia (GFAP, vimentin), and photoreceptors (rhodopsin, opsin)<br />

were quantified. Additionally, synapses were analyzed with Bassoon (presynaptic<br />

zone) and PSD95 (postsynapse).<br />

Results: No IOP alterations were noted in all groups. A 30% RGC loss was observed<br />

in both immunized groups at 4 weeks (S100 p=0.003; S100+HSP p=0.001).<br />

Cholinergic amacrine cells were also affected (S100 p=0.02; S100+HSP p=0.05),<br />

while photoreceptors remained intact. In regard to Bassoon, no changes were observable<br />

for S100, while less staining was noted in the S100+HSP group (p=0.02).<br />

Similar results were measured for PSD95, no alterations were observed in the<br />

S100 group, while a significant reduction of PSD95 signal was noted in S100+HSP<br />

animals (p=0.04). . An increase in astrocyte reactivity was noted (S100 p=0.05;<br />

S100+HSP p=0.04), while Müller glia remained unaltered.<br />

Conclusions: Findings from this study indicate that immunization with ocular<br />

antigens rather damages RGCs and amacrine cells than photoreceptors. However,<br />

the combination of S100 and HSP27 had a stronger additive effect of the<br />

retinal synapses and AII amacrine cells.<br />

129


POSTER BOARD N 32<br />

PEA-15 PHOSPHOPROTEIN MEDIATES OPTIC NERVE ASTROCYTE<br />

PHAGOCYTOSIS<br />

YANG LIU 1,2 , GULAB ZODE 1 , ABBOT F. CLARK 1 , IOK-HOU PANG 1,2<br />

1<br />

North Texas Eye Research Institute, 2 Department of Pharmaceutical Sciences<br />

University of North Texas Health Science Center, Fort Worth, TX 76107<br />

Purpose: Protein phosphorylation plays important roles in regulating cellular<br />

process. Using a phosphoproteomic approach, we detected a key phosphoprotein,<br />

phosphoprotein enriched in astrocytes (PEA-15), in retinal and optic nerve astrocytes.<br />

The role of PEA-15 in mediating astrocyte functions was further studied.<br />

Methods: Adult C57BL/6J mice were used in this study. Retinal degeneration<br />

was induced by intraorbital optic nerve crush (ONC) and ocular hypertension<br />

(OH). Dexamethasone acetate was periocularly injected weekly to induce OH<br />

and intraocular pressure was monitored using a TonoLab rebound tonometer.<br />

Phosphoproteomic study was performed using optic nerve crushed retinas.<br />

PATHER Classification System was used for bioinformatics analysis. Identified<br />

proteins were validated by western blotting and immunofluorescence staining in<br />

separate experiments (n ≥ 3). Phospho-site mutations were generated by site-directed<br />

mutagenesis. Flow cytometry-based phagocytosis assay was performed<br />

using primary cultured mouse optic nerve astrocytes.<br />

Results: ONC significantly increased phosphorylation of more than 100 retinal<br />

proteins. Among them, 53 proteins were identified. PANTHER analysis showed<br />

these proteins fall into several specific biological themes, such as metabolic processes,<br />

cellular component organization or biogenesis, anti-apoptosis and axon<br />

guidance. One of the identified proteins, PEA-15, showed 3.8 fold increase of<br />

phosphorylation in retinal and optic nerve head astrocytes at 12 hours after ONC.<br />

Similarly, ocular hypertension also increased astrocytic PEA-15 phosphorylation.<br />

PEA-15 knockdown significantly decreased optic nerve astrocyte phagocytosis by<br />

33%. Phosphomimetic mutations on S104 and S116 promoted astrocyte phagocytosis.<br />

Conclusions: PEA-15 phosphorylation mediates astrocyte phagocytosis, which<br />

likely affects retinal ganglion cell survival and regeneration after injury. This<br />

study provides potential therapeutic target for retinal neurodegeneration.<br />

130


POSTER BOARD N 33<br />

EPIGALLOCATEQUINGALLATE AND MELATONIN IN ORAL ADMINISTRATION<br />

IMPROVE VISUAL FUNCTION IN A RETINAL DEGENERATION MODEL,<br />

THE P23H RAT<br />

LORENA PERDICES 1 , ISABEL PINILLA 1,2 , LORENA FUENTES-BROTO 1,3 , FRANCISCO J. SEGURA 4 ,<br />

GEMA INSA SÁNCHEZ 2 , ELVIRA ORDUNA 2 , ANA ISABEL SÁNCHEZ-CANO 5,2 , NICOLÁS CUENCA 6<br />

1<br />

Institute for Health Research of Aragón (IIS Aragón), Zaragoza, Spain<br />

2<br />

Universitary Hospital Lozano Blesa, Zaragoza, Spain;<br />

3<br />

Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain<br />

4<br />

Department of Surgery, University of Zaragoza, Zaragoza, Spain<br />

5<br />

Department of Applied Physics, Zaragoza University, Spain<br />

6<br />

Department of Physiology Genetics and Microbiology, Alicante University, Alicante, Spain<br />

Purpose: Retinitis Pigmentosa (RP) is the most frequent cause of retinal degeneration.<br />

P23H is one of the most common rhodopsin mutations, which cause rod<br />

degeneration and oxidative stress. Melatonin and epigallocatechin gallate (EGCG)<br />

have been reported to exhibit anti-apoptosis, antioxidant and neuroprotective effects.<br />

Thus, we evaluated the synergistic effects of melatonin and EGCG in an<br />

animal model of RP, the P23H rat.<br />

Methods: 20 heterozygotes P23H line 1 rats were used and compared to 20 SD<br />

crossed with LE rats, the reference group. Four different treatment groups were<br />

established: Vehicle, 10 mg/kg/day of Melatonin and/or EGCG, administered in<br />

the drinking water from 30 to 180 postnatal days. Visual function was evaluated<br />

by a monthly optomotor test that measures visual acuity and contrast sensitivity.<br />

Results: P23HxLE rats treated with melatonin or EGCG showed better visual acuity<br />

and contrast sensitivity than those treated with vehicle in all measurements<br />

done after 30 days of treatment, slowing the disease progression.<br />

SDxLE rats treated with melatonin or EGCG increased, after 60 days of treatment<br />

(90 days old), visual function parameters even higher than young animals.<br />

Conclusions: Oral treatment of melatonin or EGCG improved vision in wild type<br />

animals and delayed vision loss in P23H rats. Furthermore, combination of melatonin<br />

and EGCG had a better effect than any of those treatments alone suggesting<br />

that both drugs have different mechanisms of action and their effects improving<br />

visual function are synergistic.<br />

131


POSTER BOARD N 34<br />

OFF-LABEL DRUGS USE IN OCULAR PHARMACOLOGY<br />

LUCIA GOZZO 1 , LAURA LONGO 1 , SILVANA MANSUETO 1 , FILIPPO DRAGO 1,2<br />

1<br />

Regional Pharmacovigilance Centre/Clinical Pharmacology <strong>Program</strong>,<br />

University Hospital of Catania, Italy;<br />

2<br />

Department of Biomedical and Biotechnological Sciences, University of Catania, Italy<br />

In compliance with national laws, Sicilian Department of Health has regulated<br />

the formal procedures that professionals must follow for off-label drug prescription.<br />

In an hospital setting, prescribers must request authorization for off-label<br />

treatment to the Health Director.<br />

Regional Pharmacovigilance Centre of Catania, within the Clinical Pharmacology<br />

<strong>Program</strong> of the Policlinic-University Hospital of Catania, supports Health<br />

Director in the assessment, approval, management and follow-up of requests for<br />

off-label drug prescriptions according to L. 94/1998 and to L. 648/96.<br />

We used a database that collect all the requests for off-label use since Clinical<br />

Pharmacology <strong>Program</strong> has been activated in 2012.<br />

We focused our attention over off-label prescriptions from Ophthalmology and<br />

we found that 80% were applied for mitomycin (MMC). This drug is an antimetabolite<br />

used for decades in order to reduce postoperative scarring during<br />

glaucoma drainage surgery (trabeculectomy) through the inhibition of fibroblast<br />

activity. MMC can be applied intra-operatively on a sponge placed for one to five<br />

minutes between the conjunctiva and sclera at the start of the operation. Evidences<br />

supporting off-label use of MMC in glaucoma surgery were considered so<br />

strong that AIFA in 2016 granted reimbursement for this unauthorized indication<br />

according to L. 648/1996.<br />

The experience of Clinical Pharmacology <strong>Program</strong> of Policlinic-University Hospital<br />

of Catania, shows that monitoring off-label prescriptions in an hospital setting<br />

could allow to detect unmet medical needs and to identify drugs with favorable<br />

risk/benefit profile which could be included in the lists of L. 648/1996 and<br />

reimbursed by the National Health System (NHS).<br />

132


POSTER BOARD N 35<br />

VITAMIN D IN SYSTEMIC SCLEROSIS PATIENTS WITH DRY EYE SYNDROME<br />

MIRIAM GALLO AFLITTO, CARLO RAPISARDA 1 , ROBERTA AMATO 1,2 , SALVO FICILI 1,2 ,<br />

DAVIDE SCOLLO 1 , GIOVANNI PANTA 1 , DANIELA ROCCA 1,2 , AGATA MESSINA 1,2 , ROSARIO FOTI 3 ,<br />

TERESIO AVITABILE 1 , ELISA VISALLI 3 , CATERINA GAGLIANO 1,2<br />

1<br />

Eye Clinic, Catania University, Italy; 2 Neurovisual Science Technology (NEST), Italy<br />

3<br />

Operative Unit of Reumatolgy,A.O.U. Policlinico Vittorio Emanuele, Catania University, Italy<br />

Purpose: The purpose of this study was to explore the clinical and pathogenic significance<br />

of vitamin D (25-hydroxyvitamin D) in systemic sclerosis (SSc) patients<br />

with dry eye syndrome (DES). The relationship between the severity of DES, SSc<br />

disease activity and levels of 25-hydroxyvitamin D was investigated.<br />

Methods: In this cross-sectional study, 32 consecutive patients with SSc and DES<br />

were enrolled. We measured blood concentrations of 25-hydroxyvitamin D (25<br />

OH D) and correlated the results with SSc disease activity calculated with modified<br />

Rodman Skin Score (mRSS), Tear osmolarity measurements (TearLab system),<br />

Tear Break-Up Time (TBUT) test, and Schirmer´s tests (type I and II).<br />

Results: Levels of 25 OH D were decreased in all SSc patients with DES as compared<br />

to normal controls and the reduced levels of 25 OH D were stable over<br />

the observed period of 2 months. Levels of 25 OH D correlated inversely with<br />

mRSS score and tear osmolarity (r = 0.610, p < 0.001), positively correlated with<br />

Schirmer values, (r = -0.231, p = 0.045), and TBUT values (r = -0.325, p = 0.007)<br />

among all patients.<br />

Conclusions: Our study demonstrated a high relationship with SSc disease acitivity,<br />

severity of DES and low levels of 25-hydroxyvitamin D.<br />

133


POSTER BOARD N 36<br />

DESIGN, SYNTHESIS, AND CHARACTERIZATION OF A SELECTIVE INHIBI-<br />

TOR FOR RETINALDEHYDE DEHYDROGENASE (ALDH1A) ENZYMES<br />

ANGELICA HARPER 1 , ANH LE 2 , TIM MATHER 3 , ANTHONY BURGET T 2 ,JODY A. SUMMERS 1<br />

1<br />

Department of Cell Biology, University of Oklahoma Health Sciences Center<br />

Oklahoma City, United States of America; 2 Department of Chemistry and Biochemistry<br />

University of Oklahoma, Norman, OK, United States of America<br />

3<br />

Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America<br />

Purpose: Retinaldehyde dehydrogenase 2 (RALDH2) has been identified as a potential<br />

therapeutic target for the control of postnatal ocular growth. The objective<br />

in this study was to use an intelligent-drug design approach to develop a<br />

RALDH2-selective inhibitor to further examine the role of RALDH2 in myopia.<br />

Methods: MoleGro software was used to dock the structure of dichloro-all-trans-retinone<br />

(DAR) into models of chick RALDH2 and human<br />

ALDH2. DAR was synthesized by a modified dihalomethyllithium approach.<br />

Selectivity and mechanism of inhibition was determined in vitro using NADH<br />

assays with recombinant RALDH2. The effect of DAR on retinoic acid (RA) synthesis<br />

was determined in 1) cells overexpressing RALDH2, 2) choroidal cell lysates,<br />

and 3) choroid tissue by an in vitro RA synthesis assay. Toxicity on scleral<br />

tissue was measured with a proteoglycan synthesis assay.<br />

Results: Docking suggested selectivity of DAR to RALDH2 compared to hu-AL-<br />

DH2 (MolDock score: -71.92±6.83 vs. 14.41±17.98). In vitro assays indicated that<br />

DAR inhibits RALDH2 in an irreversible and dose dependent manner (IC50=52.2<br />

nM, 20 min pre-incubation), with no significant inhibitory effect on hu-ALDH2.<br />

DAR successfully inhibited RA synthesis in 1) cells overexpressing RALDH2<br />

(p


POSTER BOARD N 37<br />

NANOTECHNOLOGICAL SIRNA FORMULATIONS FOR THE TREATMENT OF<br />

DIABETIC RETINOPATHY<br />

SARHA CUPRI, MARIALAURA AMADIO, ALESSIA PASCALE, CECILIA OSERA, VELIA D'AGATA,<br />

AGATA GRAZIA D'AMICO, GIAN MARCO LEGGIO, BARBARA RUOZI, STEFANO GOVONI, FILIPPO DRAGO<br />

CLAUDIO BUCOLO, ROSARIO PIGNATELLO<br />

Department of Drug Sciences, University of Catania, Italy<br />

Purpose: This study assessed whether the strategy to targeting to human antigen<br />

R (HuR), a member of the ELAV protein family, may represent a new potential<br />

therapeutic strategy to manage diabetic retinopathy 1 . We evaluated the delivery<br />

of siRNA silencing HuR expression, by different nanocarriers, consisting on cationic<br />

SLN and liposomes (as lipoplexes).<br />

Methods: The SLN carriers were prepared by a modification of QESD technique<br />

2 . Liposomes were obtained by a combination of the TLE technique and<br />

extrusion by LiposoFast® 3 . The complexation ratio (N/P), was set at 4:1 and 10:1,<br />

with 2.5 µM HuR siRNA. They have been characterized by PCS for mean size,<br />

PDI, zeta potential. The morphology was determinated by atomic force microscopy<br />

(AFM) and scanning transmission electron microscopy (STEM). Nanocarriers<br />

were injected intravitreously on Male Sprague–Dawley rats after diabetes<br />

induction ( streptozotocin), to measure the effects of naked or complexed siRNA<br />

on HuR protein expression and VEGF production. Retinal HuR and VEGF levels<br />

were detected by Western blot and ELISA, respectively.<br />

Results: Retinal HuR and VEGF are significantly increased in STZ-rats (control)<br />

and are blunted by HuR siRNA treatment. Lipoplexes with a weak positive surface<br />

charge and with a 4:1 N/P (cationic lipid nitrogen to siRNA phosphate) ratio<br />

exert a better transfection efficiency, significantly dumping retinal HuR and<br />

VEGF levels.<br />

Conclusions: These findings are the proof-of-concept that HuR represents a novel<br />

pharmacological target useful to counteract pathologies implicating VEGF deregulation,<br />

such as DR, and can be efficaciously delivered by optimized cationic<br />

lipid-based nanocarriers.<br />

1<br />

Amadio, M. et al. The PKCbeta/HuR/VEGF pathway in diabetic retinopathy. Biochem. Pharmacol. 2010,<br />

80, 1230-1237.<br />

2<br />

Pignatello, R. et al. Optimization and validation of a new method for the production of lipid nanoparticles for ophthalmic<br />

application. Int. J. Med. Nano Res. 2014, 1:006.<br />

3<br />

Pignatello, R., Sarpietro, M.G. General experimental set-up of liposomal systems for DSC. In: Drug-biomembrane<br />

interaction studies. The application of calorimetric techniques (Pignatello, R., ed.). Elsevier/Woodhead Publ. Ltd.,<br />

Cambridge, UK; pp. 363-379 (2013).<br />

135


POSTER BOARD N 38<br />

PROTECTIVE EFFECT OF ID PROTEIN ON TGFβ2INDUCED FIBROSIS IN<br />

HUMAN TRABECULAR MESHWORK CELLS: IMPLICATION FOR DEVELOPING<br />

A GLAUCOMA THERAPY<br />

AVANI A. MODY, ROBERT J. WORDINGER, ABBOT F. CLARK<br />

North Texas Eye Research Institute<br />

University of North Texas Health Science Center, Fort Worth, TX USA<br />

Purpose: Elevated transforming growth factor β2 (TGFβ2) expression in the<br />

trabecular meshwork (TM) causes increased deposition of extracellular matrix<br />

(ECM) and prevents ECM turnover by increasing expression of plasminogen activator<br />

inhibitor-I (PAI-1), leading to elevated intraocular pressure (IOP) in glaucoma<br />

patients. In cardiovascular diseases, bone morphogenetic proteins (BMP)<br />

through induction of inhibitor of DNA binding proteins (ID1, ID3); transcription<br />

regulators known to suppress bHLH promoter activity, regulate TGFβ2 induced<br />

ECM production. However, in TM cells the underlying mechanism for BMP4<br />

inhibition of TGFβ2-induced fibrosis remains undetermined. Our study will determine<br />

whether ID1and ID3 proteins are downstream targets of BMP4, which<br />

attenuates TGFβ2 induction of ECM proteins in cultured human TM cells.<br />

Method: Primary human TM (HTM) cells were treated with BMP4 for 0-48hr,<br />

and ID1 and ID3 mRNA and protein expression was determined by Q-PCR and<br />

western immunoblotting. HTM cells were treated with a BMPR inhibitor to confirm<br />

that BMP4 signaling is necessary for induction of ID1 and ID3 protein expression.<br />

GTM3 cells were transfected with ID1 or ID3 vectors to determine their<br />

inhibitory effects on TGFβ2 induced fibronectin and PAI-1 protein expression.<br />

Results: BMP4 significantly induced early expression of ID1 and ID3 mRNA<br />

(p


POSTER BOARD N 39<br />

ROLE OF GLUCOCORTICOID RECEPTOR GRβ IN GLUCOCORTICOID-IN-<br />

DUCED OCULAR HYPERTENSION AND GLAUCOMA IN MICE<br />

GAURANG C. PATEL, YANG LIU, J. CAMERON MILLAR, AND ABBOT F. CLARK<br />

North Texas Eye Research Institute, University of North Texas Health Science Center<br />

Fort Worth, TX-76107, USA<br />

Purpose: Prolonged glucocorticoid (GC) therapy causes GC-induced ocular hypertension<br />

(OHT) that can lead to iatrogenic glaucoma and permanent vision<br />

loss. Alternatively spliced isoform of the glucocorticoid receptor GRβ acts as<br />

dominant negative regulator of GC activity. Overexpressing GRβ in human trabecular<br />

meshwork (TM) cells inhibits GC-induced glaucomatous changes and<br />

damage. The purpose of this study was to determine whether increased expression<br />

of GRβ prevented as well as reversed GC-OHT in mice.<br />

Methods: To generate GC-OHT, C57BL/6J mice received weekly periocular injections<br />

of dexamethasone acetate. Prior to or several weeks after DEX-Ac administration,<br />

mouse eyes were injected intravitreally with Ad5.null or Ad5.hGRβ<br />

expression vectors to transduce TM. Intraocular pressure (IOP) was measured<br />

using TonoLab tonometer, and outflow facilities were measured in living mice<br />

using constant flow infusion technique. Fibronectin and collagen I expression<br />

were evaluated using immunoblotting from mouse anterior segment tissues.<br />

Results: DEX-Ac significantly increased IOP compared to vehicle control eyes<br />

(p


POSTER BOARD N 40<br />

HUMAN-SPECIFIC LONG NON-CODING RNAS REGULATEOCULAR<br />

ANGIOGENESIS<br />

BO YU 1 , QINBO ZHOU 1 , CHASTAIN ANDERSON 1 , JAKUB HANUS 1 , FANGKUN ZHAO 1 , JING MA 1<br />

KUN ZHANG 3 AND SHUSHENG WANG 1, 2<br />

1Department of Cell and Molecular Biology<br />

2<br />

Department of Ophthalmology, Tulane University New Orleans, LA, 70118, USA<br />

3<br />

Department of Computer Science, Xavier University, New Orleans, LA, 70125<br />

Purpose: Many animal studies failed to translate into humans, which is true for<br />

angiogenesis. The mechanisms underlying the species-specific regulation of angiogenesis<br />

is still unclear. The purpose of this project is to identify EC-enriched<br />

human-specific long non-coding RNAs (lncRNA), and study their function using<br />

human angiogenesis models.<br />

Methods: We employed expression profiling in 5 different human cell lines (3<br />

endothelial cell (EC) lines, 2 non-EC lines) to screen for EC-enriched lncRNAs.<br />

Real time PCR and bioinformatics analysis were performed to confirm their expression.<br />

Knock-down, overexpression as well as CRISPR/Cas9-based knock-in<br />

approaches were adopted to study the function of candidate lncRNAs using in<br />

vitro and ex vivo models.<br />

Results: About 500 lncRNAs were identified to be enriched in ECs (2-fold cutoff).<br />

A list of the lncRNAs show a correlated expression profile with nearby coding<br />

mRNAs that are implicated in vascular development and disease. Specifically,<br />

we found that a novel human specific lncEGFL7OS is required for proper angiogenesis<br />

in human systems. Silencing of lncEGFL7OS represses EC proliferation<br />

and migration, and impairs vascular tube formation in both Matrigel and EC/fibroblast<br />

co-culture angiogenesis assays. Mechanistically, lncEGFl7OS regulates<br />

MAPK and AKT pathways to enhance angiogenesis. RNA immunoprecipitation<br />

assays established that lncEGFL7OS function by interacting with key transcription<br />

factors for angiogenesis.<br />

Conclusions: We identified a list human-specific EC-enriched lncRNAs. We<br />

found a novel lncRNA lncEGFL7OS is required for human angiogenesis by interacting<br />

with transcription factors to regulate MAPK and AKT pathways in ECs.<br />

lncRNAs may represent key regulators of human angiogenesis-related diseases,<br />

including age-related macular degeneration.<br />

138


POSTER BOARD N 41<br />

OCULAR TISSUE DISTRIBUTION OF ORALLY ACTIVE MULTIFUNCTIONAL<br />

ANTIOXIDANTS<br />

DAMIAN M. DASZYNSKI 1 , THEODOR A. WOOLMAN 1 , KAREN BLESSING1, AND PETER F. KADOR 1,2<br />

1College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA<br />

2<br />

Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE, USA<br />

Purpose: To determine the ocular distribution of orally administered multifunctional<br />

antioxidants (MFAOs) along with their monofunctional and parent analogs<br />

in rats and compare this distribution to drug levels obtained in the brain,<br />

heart, liver, kidney, and sciatic nerve. The ultimate purpose of this study is to<br />

determine whether these drug levels can be correlated with various molecular<br />

parameters in order to predict the specific tissue targeting of these compounds.<br />

Methods: Twenty-four compounds consisting of 6 MFAOs, their monofunctional<br />

analogs and nonfunctional parent compounds were incorporated into standard<br />

rat chow (0.05%) and orally administered to Sprague Dawley rats. After 7 days<br />

of feeding, all rats were terminally perfused with PBS and the eyes along with<br />

the brain, heart, liver, kidney, and sciatic nerve were removed for subsequent<br />

quantification of drug levels by HPLC-MS. Eyes were further dissected to allow<br />

drug level analysis of the cornea, iris/ciliary processes, lens, neural retina, and<br />

retinal pigment epithelium attached to the remaining posterior segment. Drug<br />

levels were correlated with molecular parameters calculated using MOE which<br />

included atomic polarizabilities, molecular refractivity, logP, topological polar<br />

surface area, kappa shape indices, Balaban’s topological index, dipole moment,<br />

surface area, volume, shape, water accessible surface area, hydrophilic/hydrophobic<br />

volume, polar volume, critical packing parameter , hydrophilic-lipophilic<br />

descriptors, water accessible surface area of all hydrophobic/polar atoms, and<br />

positive/negative charge weighted surface areas.<br />

Results: Several correlations were observed between ocular drug concentrations<br />

versus calculated MOE molecular parameters. The HK-piperidine with<br />

R2 = methoxy and the HK-pyrrolidine with R2 = hydrogen MFAOs were generally<br />

shown to accumulate in greater quantities relative to their parent and<br />

monofunctional analogs. The JHX-series with R2 = methoxy or hydrogen, the<br />

HK-piperidine with R2 = hydrogen, and the HK-pyrrolidine with R2 = methoxy<br />

showed greater accumulation of either free radical scavenger or metal chelator<br />

monofunctional analog compared to the parent or multifunctional analogs.<br />

Conclusion: Results to date suggest QSAR studies that correlate the ocular levels<br />

of MFAOs and their analogs with in silico MOE calculated molecular parameters<br />

have merit in helping us understand the distribution of these drugs in the eye.<br />

139


POSTER BOARD N 42<br />

TARGETING INFLAMMATION TO DELAY PHOTORECEPTOR DEGENERATION<br />

IN AN ANIMAL MODEL OF RETINITIS PIGMENTOSA<br />

MARTINA BIAGIONI 1 , VIVIANA GUADAGNI 1 , ELENA NOVELLI 1 , ENRICA STRET TOI 1<br />

1<br />

CNR Neuroscience Institute, Pisa, Italy<br />

In Retinitis Pigmentosa (RP) a genetic defect causes the primary degeneration<br />

of rods, followed by the secondary death of cones, culminating into blindness.<br />

We previously showed that Environmental Enrichment (EE) effectively slows<br />

down photoreceptor degeneration in rd10 mutant mice, a well-established model<br />

of human RP. Searching for the molecular effectors of this protective response,<br />

we found that retinal degeneration results in a major inflammatory and immune<br />

response at retinal level, and that this response is effectively reduced by exposure<br />

to EE.<br />

To test the hypothesis that EE rescue effects could be mimicked by an anti-inflammatory<br />

treatment, we implemented a protocol of dexamethasone administration<br />

to rd10 mice.<br />

Groups of rd10 mice received daily Dexamethasone (Soldesam forte 4mg/ml)<br />

from P23 to P45, administered with a gavage needle; control mice received water.<br />

For histological studies, retinas were fixed in 4% PFA and stained with cell-specific<br />

antibodies for cone photoreceptors (cone opsins) and microglia (Iba1), then<br />

imaged by fluorescence and confocal microscopy for cells counts.<br />

Preliminary results demonstrate that Dexamethasone treated mice show an<br />

increased number of surviving cones compared to matched controls. The anti-inflammatory<br />

effect of the drug is confirmed by a decreased number of retinal<br />

microglial cells displaying an activated state. Molecular studies are ongoing to<br />

confirm a reduced pattern of retinal inflammation.<br />

These data open the perspective of slowing down retinal decay in human RP<br />

targeting the inflammatory response. Indeed, a sustained Dexamethason release<br />

device is already in use for macular edema and its repurposing for RP should be<br />

easily achievable.<br />

140


POSTER BOARD N 43<br />

AGE-RELATED MACULAR DEGENERATION AND TGF-β1:<br />

PHARMACODYNAMIC AND PHARMACOKINETIC PROFILE<br />

FRANCESCA LAZZARA, CLAUDIO BUCOLO, LEGGIO GIAN MARCO, VINCENZO FISICHELLA,<br />

GIURDANELLA GIOVANNI, CHIARA BIANCA MARIA PLATANIA, ANNAMARIA FIDILIO, FEDERICA GERACI,<br />

FILIPPO DRAGO<br />

Department of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania, Italy<br />

Purpose: To investigate the protective effect of TGF-β1 in an animal model of<br />

age-related macular degeneration (AMD) and to evaluate the ocular bioavailability<br />

of TGF-β1 formulation with lipidic system.<br />

Methods: Sprague-Dawley rats were used. Human Aβ1-42 oligomers were prepared<br />

and intravitreally injected (10µM) with and without recombinant human<br />

TGF-β1 (1ng/1 μl). After 48h, the animals were sacrificed and the eyes removed.<br />

The apoptotic markers Bax and Bcl-2 were assessed by Western Blot analyses.<br />

Small lipid unilamellar vesicles loaded with TGFβ1 were complemented by annexin<br />

V and Ca2+ prior topical administration to albino rabbits. TGFβ1 bioavailability<br />

was evaluated in the vitreous at different time points (30’, 60’, 120’, 180’,<br />

240’) by a commercial ELISA kit, after single topical administration of the new<br />

formulation. Ocular tolerability of TGFβ1 formulation was also assessed by a<br />

modified Draize’s test.<br />

Results: Treatment with Aβ oligomers induced a strong increase of Bax protein<br />

level (about 4fold; p


POSTER BOARD N 44<br />

ANTIOXIDANT AND OSMOPROTECTING ACTIVITY OF TAURINE<br />

IN DRY EYE MODELS<br />

ANNAMARIA FIDILIO, CLAUDIO BUCOLO, CHIARA BIANCA MARIA PLATANIA,<br />

FRANCESCA LAZZARA, FEDERICA GERACI, FILIPPO DRAGO<br />

Department of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania, Italy<br />

Purpose: To assess the efficacy of ophthalmic formulations composed of taurine<br />

(TAU) and sodium hyaluronate (SH) to manage ocular surface diseases.<br />

Methods: Rabbit corneal epithelial cells (SIRCs) were subjected to oxidative stress<br />

(1 mM H2O2) and treated with the following formulations: 0.2% SH, 0.4% SH,<br />

0.4% SH + 0.5% TAU. Reactive oxygen species (ROS) were evaluated by commercial<br />

kit (ab113851).<br />

Dry eye was induced in albino rabbits by topical application (4 times per day) of<br />

1% atropine eye drops and fifteen minutes after atropine instillation the eyes were<br />

treated with formulations (0.2% SH, 0.4% SH, 0.4% SH + 0.5% TAU). The following<br />

endpoints were evaluated: Ferning test, Schirmer’s test, tear breakup time<br />

(TBUT), tear osmolarity and MMP-9 expression in tear by Western Blotting.<br />

Results: Taurine significantly (p


POSTER BOARD N 45<br />

IDENTIFICATION OF A GENE SIGNATURE REGULATED BY A LNCRNA<br />

ASSOCIATED WITH EXFOLIATION GLAUCOMA<br />

WILLIAM M. JOHNSON 1 , INAS F. ABOOBAKAR 1 , LAURA K. FINNEGAN 2 , R. RAND ALLINGHAM 1<br />

MICHAEL A. HAUSER 1,3,4 , W. DANIEL STAMER 1<br />

1<br />

Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA<br />

2<br />

School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland<br />

3<br />

Department of Medicine, Duke University Medical Center, Durham, NC, USA<br />

4<br />

Singapore Eye Research Institute, Singapore National Eye Center, Singapore, Singapore<br />

Duke, National University of Singapore, Singapore, Singapore<br />

Purpose: Exfoliation glaucoma (XFG) is a clinically aggressive and genetically<br />

distinct form of glaucoma caused by extrafibullary material deposition in the trabecular<br />

meshwork, leading to increased intraocular pressure and death to retinal<br />

ganglion cells. Gene variants located in the promoter of a novel long non coding<br />

RNA (lncRNA), LOXL1-AS1 associate with XFG risk. Since lncRNAs can impact<br />

susceptibility to disease through alterations in gene expression, we investigated<br />

the targets of LOXL1-AS1.<br />

Methods: RNAseq was performed on cDNA derived from an immortalized lens<br />

epithelial cell line (B-3), where LOXL1-AS1 expression was knocked down. Incubation<br />

of LOXL1-AS1 or control RNA with nuclear lysates followed by SDS-<br />

PAGE, silver staining and mass spectrometry analysis, which revealed distinct<br />

LOXL1-AS1 binding partners. Recombinant protein and LOXL1-AS1 were incubated<br />

in vitro to test binding.<br />

Results: RNAseq identified 88 significantly altered RNAs, including 14 lncRNAs<br />

using parameters of logFC +/- 1.5 and P < 0.0001. Notably, several upregulated<br />

RNAs were filamentous/extracellular matrix proteins. To directly assess the<br />

mechanism by which LOXL1-AS1 alters gene expression, mass spectrometry<br />

revealed that LOXL1-AS1 interacts with two nuclear proteins involved in RNA<br />

processing, and deletion of a 14 bp region in LOXL1-AS1 eliminated binding of<br />

one candidate.<br />

Conclusions: Our data indicate for the first time that LOXL1-AS1 regulates a specific<br />

subset of target RNAs that may occur through interaction with the RNA<br />

processing machinery. These findings strengthen the hypothesis that LOXL1-<br />

AS1 is a key player in XFG pathology and that targeting of RNA processing proteins<br />

may provide novel therapeutic strategies for XFG.<br />

143


POSTER BOARD N 46<br />

MIRNAS IN VITREOUS HUMOR OF PATIENTS AFFECTED BY IDIOPATHIC<br />

EPIRETINAL MEMBRANE AND MACULAR HOLE<br />

MARIO D. TORO (PRESENTER), 1 ANDREA RUSSO 1 , MARCO RAGUSA 2 , ANTONIO LONGO 1<br />

TERESIO AVITABILE 1 , CINZIA DI PIETRO 2 , DAVIDE BARBAGALLO 2 , MICHELE REIBALDI 1<br />

1<br />

Department of Ophthalmology, University of Catania<br />

2<br />

Department of Biology, University of Catania<br />

Purpose: The aim of the present study was to assess the expression of miRNAs<br />

in the Vitreous Humor (VH) of patients with Macular Hole (MH) and Epiretinal<br />

Membrane (ERM) compared to an unaffected control (UC) group.<br />

Methods: In this prospective, comparative study, 2-ml of VH was extracted from<br />

the core of the vitreous chamber in consecutive patients that underwent standard<br />

vitrectomy for ERM and MH. RNA was extracted and TaqMan Low Density Arrays<br />

(TLDAs) was performed to profile the transcriptome of 754 miRNAs. Results<br />

were validated by single TaqMan assays. Finally we created a biological network<br />

of differentially expressed miRNA targets and their nearest neighbours.<br />

Results: Overall 10 eyes with MH, 16 eyes with idiopathic ERM and 6 UC were<br />

enrolled in the study. Profiling data identified 5 miRNAs differentially expressed<br />

in patients affected by vitreoretinal diseases (MH + ERM) with respect to UC. Four<br />

were downregulated (miR-19b, miR-24, miR-155, miR-451) and 1 was upregulated<br />

(miR-29a); TaqMan assays in VH of patients affected by MH and ERM with<br />

respect to UCs showed that the most differentially expressed were miR-19b (FC<br />

-9.13, p:


POSTER BOARD N 47<br />

INNER RETINAL REMODELING IN AN INDUCIBLE MOUSE MODEL OF<br />

RETINITIS PIGMENTOS<br />

ANTONIA STEFANOV 1 , ELENA NOVELLI 1 , ENRICA STRET TOI 1<br />

1<br />

CNR Neuroscience Institute, Pisa, Italy<br />

In Retinitis Pigmentosa (RP) a mutation in a retinal-specific gene triggers a rodto-cone<br />

degeneration ultimately leading to blindness. Studies on developmental<br />

rodent models of RP have shown that inner retinal cells also remodel regressively<br />

and might die out, limiting the possibility of therapeutic approaches relying on<br />

the preservation of the inner retina.<br />

Here we report remodeling studies carried on in the retina of a photo-inducible<br />

RP model, the Tvrm4 rhodopsin mouse.<br />

Our hypothesis is that remodeling of second order neurons in Tvrm4 mice induced<br />

in young adulthood conforms to stereotyped features previously described<br />

in developmental models of RP.<br />

Tvrm4 mice (with a dominant Rhodopsin mutation) aged 3-5 months and WT<br />

littermates were exposed to 12k lux neon light for 2 mins and harvested 3 or 6<br />

weeks afterwards. Retinal tissue was processed for immunocytochemistry to reveal<br />

rod bipolar and horizontal cells, which were analyzed and counted by confocal<br />

microscopy and image analysis. No variation in the number of rod bipolar cells<br />

was found in Tvrm4 mice with respect to WT controls. However, we observed<br />

a clear process of dendritic and axonal arborization retraction and loss of spatial<br />

order in these neurons. Horizontal cells also lost numerous dendrites while<br />

their density decreased to 63% in the central retina.<br />

As these events have been described in developmental RP models, we conclude<br />

that remarkable survival of neurons postsynaptic to rods (albeit regression of<br />

dendritic arbors) is a hallmark of RP and is not influenced by the age of disease<br />

onset and causative mutation.<br />

145


POSTER BOARD N 48<br />

RETINAL PK PROFILE OF CURCUMIN AFTER ORAL ADMINISTRATION<br />

IN RABBITS<br />

1<br />

CLAUDIO BUCOLO, 1 ANNAMARIA FIDILIO, 1 CHIARA BIANCA MARIA PLATANIA,<br />

1<br />

FRANCESCA LAZZARA, 1 FEDERICA GERACI, 2 CATENO PIAZZA<br />

1<br />

SALVATORE SALOMONE, 1 FILIPPO DRAGO<br />

1<br />

Department of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania, Italy<br />

2<br />

Research Centre, Unifarm, Catania, Italy<br />

Purpose: Although curcumin is widely available in the market as nutritional supplements<br />

claiming visual function protection, the bioavailability in retinal tissue,<br />

after oral administration, has never been investigated. The aim of the study was<br />

to assess retinal PK profile of three commercial products after single oral administration<br />

in rabbit.<br />

Methods: Commercial products used in the present study: 1) water soluble curcumin<br />

formulation (CHC; Diabec®-Alfa-Intes); curcumin-phosphatidylcholine<br />

complex (CPC; Norflo®-EyePharma); curcumin + piperine product (CPI; AVS<br />

Retina®-SIFI). Albino rabbits were used in the present study. All of the animals<br />

were treated according to the Association for Research in Vision and Ophthalmology<br />

(ARVO) Statement for the Use of Animals in Ophthalmic and Vision<br />

Research. The animals received curcumin formulations by oral gavage (60, 100<br />

and 50 mg/kg of turmeric extract CHC, CPC and CPI, respectively) retina was<br />

collected from animals at fixed time intervals after dosing. Retinal levels of curcumin<br />

were evaluated by liquid chromatography mass spectrometry (LC-MS/<br />

MS) with an LLOQ of 0.0002 ng/mg. Pharmacokinetic parameters, peak drug<br />

concentration (Cmax), time to peak value (Tmax), and area under the concentration-time<br />

curve between 0 and 24 h (AUC0-24) were determined.<br />

Results: Data generated from the three groups demonstrated that only CHC-treated<br />

group showed retinal levels of curcumin after oral administration (Cmax=0.011<br />

± 0.002 ng/mg Tmax= 6 h and AUC0-24= 6.66 ng*min/mg of retina). Retina<br />

curcumin levels after oral administration of CPC and CPI were below the LLOQ.<br />

Discussion: Taken together, these data seem to indicate that CHC formulation<br />

provides relevant curcumin levels in rabbit retina after single oral administration<br />

suggesting that this formulation may be of value in clinical practice to manage<br />

retinal conditions.<br />

146


POSTER BOARD N 49<br />

NANOPARTICLE INTERACTION WITH VITREOUS HUMOR AND ITS EFFECT<br />

ON RETINAL PIGMENT EPITHELIAL CELL UPTAKE<br />

RYAN A. KELLEY AND UDAY B. KOMPELLA<br />

Skaggs School of Pharmacy and Pharmaceutical Sciences<br />

University of Colorado Anschutz Medical Center, Aurora, CO, USA<br />

Purpose: The purpose of this study was to determine whether exposure of<br />

nanoparticles to vitreous humor alters their physicochemical properties and retinal<br />

cell uptake.<br />

Methods: Either negatively charged carboxylate modified or positively charged<br />

amine modified polystyrene nanoparticleswere incubated with rabbit vitreous<br />

humor or water for 30 min. Subsequently, particles were separated by centrifugationand<br />

washed. Particle size and zeta-potential were measured using Malvern<br />

Zetasizer Nano ZS. Uptake of nanoparticles into ARPE-19 cells was monitored<br />

using a plate-reader and fluorescent microscopy. Further, the nanoparticles were<br />

run on SDS-PAGE gels to determine the presence of interacting vitreous proteins.<br />

Results:Incubation of carboxylate and amine modified nanoparticlesin vitreous<br />

resulted in an increase in size from 203 and 295 to 1129and 3204nm, respectively.<br />

Zeta-potential of carboxylate nanoparticlesincreased from -58.4 to -26.2 mV,<br />

while that of amine nanoparticles decreased from +37.7 to -30.5mV. These shifts<br />

resulted in a ~30% decrease in carboxylate nanoparticleuptake in ARPE-19 cells.<br />

SDS-Page analysis of carboxylate nanoparticles post vitreous incubation revealed<br />

multiple distinct protein bands compared to nanoparticles incubated in water.<br />

Amine nanoparticle uptake was not quantified due to binding of nanoparticles<br />

to plate wells.<br />

Conclusions:Nanoparticle surface moieties interact with constituents of the vitreous<br />

humor, which causes a shift in both size and zeta-potential. These changes<br />

caused a decrease in uptake of carboxylate nanoparticles by ARPE-19 cells. Identifying<br />

nanoparticle-vitreous interactions will help design nanoparticlesthat have<br />

increased/decreased vitreous humor mobility and retinal uptake, aiding in delivery<br />

of both drugs and genes.<br />

147


POSTER BOARD N 50<br />

TRANSPALPEBRAL RHEOOPHTHALMOGRAPHYEVALUATION OF THE<br />

IMPACT OF PROSTAGLANDIN ANALOGS ON OCULAR HEMODYNAMICS<br />

INEARLY PRIMARY OPEN-ANGLE GLAUCOMA<br />

ELENA IOMDINA 2 , ALINA KLEYMAN 1 , OLGA KISELEVA 1 , ALEXANDER BESSMERTNY 1<br />

PETER LUZHNOV 3 , DMITRY SHAMAEV 3<br />

1<br />

Department of Glaucoma, Moscow Helmholtz Research Institute of<br />

Eye Diseases, Moscow, Russia<br />

2<br />

Department of Refraction Pathology, Binocular Vision Anomalies and<br />

Ophthalmoergonomics, Moscow Helmholtz Research Institute of Eye Diseases, Moscow, Russia<br />

Purpose: Experimental implementation of scleral collagen crosslinking by<br />

sub-Tenon’s capsule injections of a biologically active composition, Scleratex, in<br />

the equatorial and posterior pole areas of the eye.<br />

Material and Methods: A placebo-controlled study into the safety and effectiveness<br />

of sub-Tenon’s capsule injections of Scleratex (a solution of the basic amino<br />

acid salts in the form of succinates) was performed on 47 Chinchilla rabbits (94<br />

eyes). 0.1 ml of Scleratex or placebo solution was injected once a week under the<br />

Tenon’s capsule of the experimental and the fellow eyes, respectively. The first<br />

series (4 injections) lasted 1 month, and the second series (12 injections) took 3<br />

months. After the course of injections, all structures of 22 enucleated eyes, including<br />

retina, were studied morphologically using light microscopy, while<br />

scleral samples from the remaining 72 eyes were used to determine the elasticity<br />

modulus (on the testing machine Autograph AGS-H, SHIMADZU, Japan) and the<br />

level of collagen crosslinking (by denaturation temperature Td using differential<br />

scanning calorimetry on the calorimeter, Phoenix DSC 204, Netzsch, Germany).<br />

Results: The weekly injections of Scleratex performed for 1 month or 3 months<br />

showed no clinical or morphological signs of local irritation, damage, or toxicity.<br />

A 15 to 20% increase in collagen crosslinking and a 1.8-fold increase in the elasticity<br />

modulus of the sclera with respect to the fellow eye were detected. This increase<br />

was accompanied by an increase in the number of cells, formation of new<br />

connective tissue on the scleral surface, and appearance of additional vessels. In<br />

total, this is an evidence of an effective trophic and sclera strengthening influence<br />

of Scleratex.<br />

Conclusion: The outcome of experimental implementation of a minimally invasive<br />

technology for scleral collagen crosslinking shows it to be a plausible method<br />

of scleral strengthening and antidystrophic treatment of progressive myopia.<br />

148


POSTER BOARD N 51<br />

PRESENCE OF MELANOPSIN IN HUMAN CRYSTALLINE LENS EPITHELIAL<br />

CELLS AND ITS ROLE IN MELATONIN SYNTHESIS<br />

HANAN AWAD ALKOZI, XIAOYU WANG, MARIAJESUS PEREZ DE LARA AND JESUS PINTOR<br />

Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry<br />

Universidad Complutense de Madrid, Madrid, Spain<br />

Purpose: To detect the presence of melanopsin in the human lens epithelial cells<br />

and to establish a clear link between melanopsin activation and melatonin production<br />

as well as the quantification of melatonins’ synthesis key enzyme Arylalkymine<br />

N-acetyltransferase (AANAT).<br />

Methods: Human immortalized lens epithelial cells were used to detect melanopsin<br />

by means of immunocytochemistry and western blot. After that, cells were<br />

plated in multiwells and treated with white, blue (465-480nm, 400 Lux), green<br />

(520-550nm, 400 Lux), red (625-640nm, 400 Lux) and total darkness for 2, 4, 8,<br />

and 12 hours. Melanopsin activity was blocked with AA92593 at 1.5μM concentration<br />

and for second messenger inhibition a phospholipase C inhibitor U73122<br />

was used at 3μM. In all experimets, supernatants were collected for melatonin<br />

measurments using HPLC, and cell lysis was done for AANAT cuantification by<br />

means of western blot.<br />

Results: melatonin levels after submitting cells to total darkness are significantly<br />

higher to ones submitted to white or specifically blue light, melatonin cencentrations<br />

were 59.45 ± 15.71 pmol/106 cells in the darkness, and dropped to 37.61 ±<br />

6.64 pmol/106 under blue light (***p


POSTER BOARD N 52<br />

IMMUNOHISTOCHEMICAL CHARACTERIZATION OF NEUROTRANSMITTERS<br />

IN THE EPISCLERAL CIRCULATION IN RATS<br />

ANJA LADEK 1 , ANDREA TROST 1 , CHRISTIAN RUNGE 1 , FALK SCHRÖDL 1<br />

CLEMENS A. STROHMAIER 1 , HERBERT A. REITSAMER 1<br />

1<br />

Ophthalmology, Paracelsus Medical University<br />

SALK, MüllnerHauptstrasse 48, 5020 Salzburg, Austria<br />

Purpose: Episcleral venous pressure (EVP) is an important parameter of steadystate<br />

intraocular pressure and recent evidence suggests neuronal influence on<br />

EVP. Yet little is known about the innervation and more, specifically, the neurotransmitters<br />

involved. The present study aims at identifying possible neurotransmitter<br />

candidates for the episcleral circulation in rats.<br />

Methods: Four previously untreated, enucleated, immersion fixated rat eyes, taken<br />

from three animals were cut into serial sections using a cryostat, followed<br />

by standard immunohistochemistry. Additionally to the neurotransmitter under<br />

investigation, each slide was stained with antibodies against smooth muscle actin<br />

and neurofilament (200kDa).<br />

Results: Throughout the slides neurofilament positive cells could be identified<br />

in close proximity to the vascular endothelium. These cells showed synapse like<br />

structures and stained positive for synaptophysine. Furthermore, they stained<br />

positively for ChAT, Galanin, CGRP, Substance P and PGP 9.5. Staining with antibodies<br />

against nNOS, Tyrosine Hydroxylase, Serotonin (5-HT) Transporter and<br />

Alarine yielded negative results.<br />

Conclusion: These findings indicate that there is neuronal input to the episcleral<br />

circulation. However no assumption can be made whether the positively stained<br />

structures are of functional significance for the regulation of the episcleral venous<br />

pressure and thereby intraocular pressure.<br />

150


POSTER BOARD N 53<br />

ROLE OF LACTOBIONIC ACID AND HYALURONIC ACID IN PROMOTION OF<br />

CORNEAL EPITHELIAL WOUND HEALING IN VITRO AND IN VIVO<br />

MELANIA OLIVIERI 1 , DARIO RUSCIANO 1 , SALVATORE PEZZINO 2 , C. DANIELA ANFUSO 3<br />

GABRIELLA LUPO 3 , MARTINA CRISTALDI 1<br />

1<br />

Sooft Italia, University of Catania, Catania, Italy<br />

2Bioos Italia, University of Catania, Catania, Italy<br />

3<br />

Department of Biomedical and Biotechnological Sciences<br />

University of Catania, Catania, Italy<br />

Purpose: The aim of this study is to evaluate the wound-healing promoting activity<br />

of Lactobionic Acid (LA) and Hyaluronic Acid (HA) in an in vitro and in<br />

vivo model system. LA and HA are highly hygroscopic molecules, and in dermatology<br />

they are known to have anti-aging and cutaneous<br />

repair properties; moreover, LA is an efficient iron-chelator and anti-oxidant. Because<br />

the treatment of dry eye is mainly based on the topical supply of molecules<br />

endowed with lubricant properties, LA could be an ideal candidate as an ingredient<br />

of eye drops.<br />

Methods: Wound repair promoting activity of LA and HA have been investigated<br />

in vitro, on monolayers of rabbit corneal cells (SIRC), and in vivo, after disepithelization<br />

of the rabbit cornea. Inflammatory markers MMP9 and TGF-ß have been<br />

evaluated in tears and cornea tissue extracts of treated and control rabbit’s eyes.<br />

Results: Data have shown that both LA and HA promote faster wound repair<br />

in vitro. Moreover, a cooperative effect between the two compounds has been<br />

demonstrated by a cell proliferation assay.<br />

These data were confirmed in vivo: beside a faster repair of the wound, LA and<br />

HA determined a return of MMP9 and TGF-ß to normal values in tears and in<br />

corneal tissue within 48 hours.<br />

Conclusions: These results show that both LA and HA contribute to the wound<br />

closure process in vitro and in vivo. Therefore, an association of LA and HA<br />

could be an efficient treatment of dry eye<br />

and its related injuries.<br />

151


POSTER BOARD N 54<br />

EFFICACY AND SAFETY ASSESSMENT OF LACTOBIONIC ACID FOR THE<br />

TREATMENT OF DRY EYE SYNDROME<br />

ALESSANDRA PIZZO 1 , GAGLIANO C. 1,2 , AMATO R. 1,2 , FICILI S. 1,2 , MALAGUARNERA G. 1,2<br />

1<br />

Ophthalmology Department, University of Catania, Eye Clinic, Catania (Italy)<br />

2<br />

Neurovisual Science Technology (NEST), Catania (Italy)<br />

Purpose: The Dry Eye Disease (DED) is a disfunction of the ocular surface and<br />

tear film, characterized by eye dryness, light sensitivity, irritation, fluctuating vision<br />

with glare and eye fatigue.<br />

Current efforts focus on combined pharmacological strategies to restore tear film<br />

composition. Lactobionic acid has shown properties that resemble lactoferrin,<br />

which is one of the tears components with antioxidant, antimicrobial and anti-inflammatory<br />

activities.<br />

We evaluated the safety and efficacy of Lactoyal®, an eye drop containing lactobionic<br />

acid, versus hyaluronic acid (HA) in the treatment of DED.<br />

Methods: Forty adult subjects affected by DED for at least three months were<br />

selected and randomly divided into two groups. Twenty were treated with Lactoyal®,<br />

and twenty with HA. The treatment lasted five weeks and was followed<br />

by a 10-day follow up period. Subjective (VAS and SANDE scores) and objective<br />

(Osmolarity, BUT, Schirmer, Oxford staining, MMP9, Tear Lactoferrin) evaluation<br />

of dry eye parameters were conducted at enrolment and after 7 – 21 – 35 and<br />

45 days. A linear mixed model with robust standard error has been used to evaluate<br />

statistical significance of the data. A p value below 0.05 has been considered<br />

significant.<br />

Results: Both treatment groups showed an improvement of all parameters. Better<br />

results were witnessed in the Lactoyal® treatment group than in the HA one.<br />

Furthermore, we observe that the subjects older than 55 years have benefited<br />

more than younger ones.<br />

Conclusions: Treatment of DED with Lactoyal® is effective and safe, and shows<br />

an efficacy even better than HA.<br />

152


POSTER BOARD N 55<br />

VEGF EXACERBATESHIGH-GLUCOSE-INDUCED DAMAGE IN HUMAN<br />

RETINAL ENDOTHELIAL CELLS<br />

GIOVANNI GIURDANELLA, FRANCESCA LAZZARA, CLAUDIO BUCOLO,<br />

SALVATORE SALOMONE, FILIPPO DRAGO<br />

Dept. of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania, Italy<br />

Purpose: to assess the effect of VEGF in presence of high glucose (HG) in human<br />

retinal endothelial cells (HREC), an in vitro condition mimicking diabetic retinopathy.<br />

Methods: HREC were treated with normal concentration of glucose (NG, 5mM),<br />

with HG (25 mM) or with HG plus VEGF (80 ng/ml), for 48h. In some experiments,<br />

aflibercept (40 μg/ml), bevacizumab (25 μg/ml) or ranibizumab (10 μg/ml)<br />

were added to the culture medium. After treatments cells were analysed by MTT<br />

and LDH assays, Tube Formation Assay and Western Blot.<br />

Results: cell viability was reduced by about 35% (p


POSTER BOARD N 56<br />

SULODEXIDE PREVENTS HIGH GLUCOSE DAMAGE IN HUMAN RETINAL<br />

ENDOTHELIAL CELLS<br />

GIOVANNI GIURDANELLA 1 , FRANCESCA LAZZARA 1 , NUNZIA CAPORARELLO 1 ,<br />

GIAN MARCO LEGGIO 1 , GABRIELLA LUPO 1 , CARMELINA DANIELA ANFUSO 1 , CLAUDIO BUCOLO 1 ,<br />

SALVATORE SALOMONE 1 , FILIPPO DRAGO 1<br />

1<br />

Dept. of Biomedical and Biotechnological Sciences, School of Medicine,<br />

University of Catania, Catania, Italy<br />

Purpose:totest the hypothesis thatsulodexide(SDX)protectshuman retinal endothelial<br />

cells (HREC) from high glucose(HG) -induced damagethrough the suppression<br />

of inflammatory cPLA2-COX-2 pathway by blocking the effect of advanced<br />

glycation end-products (AGEs).<br />

Methods: HREC were treated with HG (25 mM) or AGEs (2 mg/ml) for 48h with<br />

or without SDX (60 μg/ml) orAflibercept (AFL, 40 μg/ml), an anti-VEGF agent.<br />

After treatments cells were analyzedwithMTT and LDH assays, Trans Endothelial<br />

Electrical Resistance (TEER) measurement, Immunohistochemistry,Tube<br />

Formation Assay, Western Blot and Real-time PCR.<br />

Results: SDX preventedcell viability reduction induced by HG or AGE (by about<br />

70% and 60% respectively, p


POSTER BOARD N 57<br />

GABAPENTIN NANOCARRIERS TO MANAGE OCULAR PAIN<br />

ROSARIO PIGNATELLO 1 , SARHA CUPRI 1 , CLAUDIO BUCOLO 2 , FILIPPO DRAGO 2 ,<br />

DARIO RUSCIANO 3 , TERESA MUSUMECI 1 , GIOVANNI PUGLISI 1<br />

1<br />

NANO-i — Research Center on Ocular Nanotechnology, Department of Drug Sciences<br />

University of Catania, Catania, Italy<br />

2<br />

Department of Biomedical and Biotechnological Sciences School of Medicine<br />

University of Catania, Catania, Italy<br />

3<br />

SOOFT Italia SpA, Montegiorgio (Fermo), Italy<br />

Purpose: Pain conditions involving the eye can arise in numerous instances, such<br />

as herpes, surgical procedures and post-surgical recovery, dry eye syndrome, inflammation,<br />

accidental trauma, neuropathic corneal pain, diabetes. Ocular pain<br />

can result from photorefractive keratotomy involving the photoablation of Bowman's<br />

membrane and the stromal layers of the cornea, or in post-herpetic neuralgia.<br />

Gabapentin (GBP) is structurally related to the neurotransmitter ƴ-aminobutyric<br />

acid (GABA) but does not bind to the GABA receptors, its mechanism of<br />

action being through binding to the α2δsubunit of calcium channels. In the recent<br />

past, oralGBP has been evaluated as an analgesic agent after various ophthalmic and<br />

non-ophthalmic surgical procedures, but with mixed results. Currently, GBPtopical<br />

formulations to treat ocular pain and/or eye inflammation has been disclosed. In<br />

this study, we evaluated some nanoparticle formulations made of Eudragit® Retard<br />

copolymers and containing GBP, to be proposed as a novel nanotechnological system<br />

for the reduction of ocular pain.<br />

Methods:The nanoparticles of Eudragit® RS100 and RL100 were prepared by a<br />

modification of the QESD technique and were characterized for their micromeritics<br />

and technological features. The nanoformulations (20 µl) were instilled in the<br />

right eye of Sprague-Dawley rats and, after 5 min, 5 µl of 0.1% aqueous formalin<br />

was applied topically in the eye. Corneal sensitivity was measured using a Cochet-Bonnet<br />

esthesiometer. The testing began with the nylon filament extended<br />

to the maximal length (6 cm). The end of the nylon filament was touched to the<br />

cornea. If the rat blinked (positive response) the length of the filament was recorded.<br />

If the rat did not blink, then the nylon filament was shortened by 0.5 cm and<br />

the test repeated until a positive response was recorded. This process was repeated<br />

for each eye three times.<br />

Results:GBP-loaded nanoparticles were obtained with a suitable size (80-250 nm,<br />

depending on the composition and preparation variables), and a net positive surface<br />

charge (Zeta potential around +30 mV), which would favor their adhesion and permanence<br />

onto the corneal surface. The nanocarriers showed to be perfectly tolerated<br />

by the animals (modified Draize test).The tested nanoformulations reduced the<br />

ocular pain with a significant extension in terms of duration of the activity (up to<br />

480 min) as compared to a GBP aqueous solution (60-120 min).<br />

Conclusions:From a technological point of view, this study demonstrated the possibility<br />

of efficiently loading small hydrophilic molecules, such as GBP, in Eudragit®<br />

Retard nanoparticles. The nanoformulationselected for in vivo evaluation was<br />

effective in prolonging the duration of the analgesic activity of GBP and can be<br />

proposed for the treatment and prevention of ocular pain, for instance associated<br />

to post-herpetic neuralgia andPRK,along with other surgical procedures, dry eye<br />

syndrome, and diabetes.<br />

155


POSTER BOARD N 58<br />

RECENT ADVANCES IN THE APPLICATION OF LIPID-BASED NANOCARRIERS<br />

TO OCULAR DRUG DELIVERY<br />

R. PIGNATELLO, C. CARBONE , T. MUSUMECI, S. CUPRI, V. PEPE, G. PUGLISI<br />

NANO-i — Research Center on Ocular Nanotechnology<br />

Department of Drug Sciences, University of Catania, Catania, Italy<br />

Controlled release of drugs to the eye tissues is still an important, though challenging<br />

topic of research. The eye is an organ highly protected from extraneous<br />

compounds by anatomical, functional and biochemical mechanisms. Such<br />

defense tools often limit the time of contact of the therapeutic formulation with<br />

the eye surface and lead to an insufficient bioavailability of the applied drugs, especially<br />

at the level of the posterior segment.<br />

Many nanotechnology strategies have been exploited for the diagnosis and cure<br />

of ocular diseases. Nanosized ocular drug delivery systems have given important<br />

results in the last years, both as topical applications on the eye surface or after<br />

intraocular administration.<br />

These colloidal carriers can be suitably engineered to overcome corneal and retinal<br />

barriers to drug penetration, protect the encapsulated drug, enhance compliance<br />

and safety of ophthalmic drugs, and prolong their activity by a controlled<br />

and/or prolonged site-specific release profile.<br />

Although the basic research on ophthalmic delivery systems supplies many technological<br />

approaches, very few of them have been able to reach a clinical relevance<br />

or to be translated into pre-industrial or industrial applications. The main<br />

reason lies in the complexity and specificity of the formulation parameters that<br />

ophthalmic products always require to tackle the high sensitivity of ocular tissues.<br />

The lecture will survey some of the more recent papers and patents regarding<br />

nanotechnology applications to ophthalmic controlled and targeted drug and<br />

gene delivery, with a specific attention to lipid-based nanocarriers, such as solid<br />

lipid nanoparticles (NLC), nanostructured lipid vectors (NLC), liposomal systems,<br />

micelles, etc.<br />

156


POSTER BOARD N 59<br />

P2X7 RECEPTOR AS PHARMACOLOGICAL TARGET IN DIABETIC<br />

RETINOPATHY<br />

CHIARA BIANCA MARIA PLATANIA, GIOVANNI GIURDANELLA 1 , LUISA DI PAOLA 2<br />

GIAN MARCO LEGGIO 1 , SALVATORE SALOMONE A , FILIPPO DRAGO A , CLAUDIO BUCOLO A<br />

1<br />

Section of Pharmacology, Department of Biomedical and Biotechnological Sciences<br />

School of Medicine, University of Catania, Catania, Italy<br />

2<br />

School of Engineering, University Campus BioMedico, Roma, Italy<br />

Purpose: To build and validate an in-silico/in-vitro approach for discovery of<br />

new antiinflammatory ligands (P2X7 inhibitors) to be used for treatment of diabetic<br />

retinopathy.<br />

Methods: Homology modeling, protein contact network analysis, molecular<br />

docking, MM-GBSA calculations were carried out in order to built and validate<br />

an in-silico approach aimed finding new ligands selective toward the P2X7 receptor.<br />

A series of P2X7 ligands have been studied by the insilico approach described,<br />

the most promising P2X7 inhibitor has been on human retinal pericytes<br />

cultured high glucose levels (25 mM). The effects of the P2X7 inhibitor have been<br />

assessed by cell viability, LDH and IL-1β levels.<br />

Results: We have generated and validated an in-silico/in-vitro platform to discover<br />

novel P2X7 receptor inhibitors. The in-silico platform is able to identify selective<br />

P2X7 inhibitors, with high true positive rate. The P2X7 inhibitor with best<br />

predicted ADME properties has shown antiinflammatory and protective activity<br />

in the described in-vitro model.<br />

Conclusions: Our data suggest that P2X7 receptor is an interesting pharmacological<br />

target for diabetic retinopathy. Furthermore, our in-silico/in-vitro screening<br />

platform is suitable for discovery of new and effective P2X7 inhibitors to be used<br />

for treatment of diabetic retinopathy.<br />

157


POSTER BOARD N 60<br />

STRUCTURAL DIFFERENCES BETWEEN P2X7 AND P2X4 RECEPTORS:<br />

A PROTEIN CONTACT NETWORK ANALYSIS<br />

CHIARA BIANCA MARIA PLATANIA 1 , LUISA DI PAOLA 2 , FILIPPO DRAGO 1, CLAUDIO BUCOLO 1<br />

1<br />

Department of Biomedical and Biotechnological Sciences, University of Catania, Italy<br />

2<br />

Università Campus Biomedico di Roma, Italy<br />

Purpose: P2X4 and P2X7 receptor share high sequence identity and homology,<br />

46% and 60%, respectively. The channel opening mechanism was previously<br />

studied only for the P2X4 receptor and this mechanism was extended to other<br />

subtypes of P2X family, even if each subtype is characterized by different electrophysiological<br />

properties. In order to analyze the structural differences between<br />

P2X4 and P2X7 receptor during can opening we analyzed the topological properties<br />

of this receptors through a protein contact network PCN analysis.<br />

Methods: Channel opening was simulated with ANM pathway http://anmpathway.lcrc.anl.gov/anmpathway.cgi/,<br />

which apply the anisotropic network model<br />

for Energy and coordinates calculation of transitino between the two conformational<br />

states open -> closed. The structural information embedded into the<br />

coordinates was the starting point to generate Protein Contact Networks (PCNs).<br />

Network nodes are the single residues, identified by their α–carbons, which are<br />

connected to each other if their Euclidean distance lies in 4-8 Å, in order to account<br />

only for significant non-covalent intra-molecular interactions. The generic<br />

element adjacency Aij of the adjacency matrix A is 1 if i-th and j-th nodes are<br />

connected by a link, otherwise is 0. Starting from A it is possible to derive several<br />

descriptors defining the wiring architecture of PCNs.<br />

Results: Correlation analysis of PCN parameters along the coordinates of transitions<br />

(t) was carried out for P2X4 and P2X7. For P2X4 transition all variables<br />

correlated with t; for P2X7 less variable showed correlation with t. The Energy<br />

of PCNs (graph Energy) overlapped the node degree (number of links of each<br />

node). Two transition states were indentified for both P2X4 and P2X7 conformational<br />

transition from closed->open state. The P2X7 receptor was characterized<br />

by higher graph energy of the transition state, compared to P2X4 receptor, 280<br />

and 100, respectively<br />

Conclusion: Overall, our data suggest that besides structure and sequence homology<br />

shared by P2X4 and P2X7; conformational transition of these receptors, from<br />

the closed to open state, was characterized by different topological parameters. In<br />

particular, P2X7 according to the theory of the transition state was characterized<br />

by slow channel opening rate, accordingly to Riedel T. Biophysical Journal 2007.<br />

158


POSTER BOARD N 61<br />

EFFECT OFDIETARY CHOLESTEROL ONOCULAR PATHOLOGIES IN AN<br />

AGE-RELATED MACULAR DEGENERATION MOUSE MODEL<br />

MICHAEL LANDOWSKI 1 , UNA KELLY 1 , MARYBETH GROELLE 1 , CATHERINE BOWES RICKMAN 1,2<br />

1<br />

Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA<br />

2<br />

Department of Cell Biology, Duke University Medical Center, Durham, NC, USA<br />

Purpose: Dietary interventions have been proposed as a therapeutic strategy for<br />

age-related macular degeneration (AMD). In fact, consumption of a Western-style<br />

diet that is high in fats and cholesterol increases an individual’s risk for AMD.<br />

Here, a multifactorial AMD mouse model based on advanced age, complement<br />

factor H deficiency (Cfh-/-) mice and exposure to a high fat, cholesterol-enriched<br />

(HFC) diet, was interrogated to determine the contribution of dietary cholesterol<br />

to production and accumulation of sub-retinal pigmented epithelial (RPE) basal<br />

deposit formation.<br />

Methods: Aged Cfh-/- mice (>90 weeks) were continued on a normal diet or<br />

switched to a HFC or high fat with no added cholesterol (HF) diet for eight<br />

weeks. Eyes were collected and processed for electron microscopy to quantify<br />

deposits. Tissue samples were collected for lipid, protein, and RNA assays.<br />

Results: Aged Cfh-/- mice fed a HF diet have decreased plasma cholesterol levels<br />

but develop basal deposits similar to those in aged Cfh-/- mice fed a HFC diet. In<br />

addition, there was a decrease in systemic dietary cholesterol-induced pathology<br />

in the livers of aged Cfh-/- mice fed a HF diet.<br />

Conclusions: Our results show that dietary cholesterol levels do not influence<br />

the development of basal deposits despite the alleviation of dietary cholesterol-induced<br />

liver damage in aged Cfh-/- mice fed a HF diet compared to those fed a<br />

HFC diet. These studies indicate lowering dietary cholesterol may not be sufficient<br />

as a therapy for AMD and suggest targeting dietary fat intake as a possible<br />

therapeutic strategy for AMD.<br />

159


POSTER BOARD N 62<br />

TREATMENT WITH AN HDAC3 SELECTIVE INHIBITOR PREVENTS<br />

RETINAL GANGLION CELL NUCLEAR ATROPHY AND APOPTOSIS<br />

AFTER ACUTE AND CHRONIC OPTIC NERVE INJURY<br />

HEATHER M. SCHMITT 1 , 2,4 , GUOJUN CHEN 3,4 , YUYUANWANG 3,4 , CASSANDRA L. SCHLAMP 1,4<br />

SHAOQUIN GONG 3,4 , ROBERT W. NICKELLS 1,4<br />

1<br />

Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI<br />

2<br />

Cellular and Molecular Pathology, University of Wisconsin-Madison, Madison WI<br />

3<br />

BIONATES Theme, Wisconsin Institute for Discovery, Madison WI<br />

4<br />

McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI<br />

Purpose: In retinal ganglion cells (RGCs) affected by optic nerve crush (ONC),<br />

HDAC3 regulates nuclear atrophy as an early response to axonal injury. Conditional<br />

knockout of Hdac3 and HDAC3 selective inhibition with RGFP966 prevent<br />

nuclear atrophy post ONC. Systemic dosing of RGFP966, which crosses the<br />

blood brain barrier, is necessary however for application to chronic models of<br />

optic nerve injury.<br />

Methods: Investigation of an intravitreal injection of RGFP966 was done to assess<br />

optimal dosing for prevention of nuclear atrophy and apoptosis up to 14 days after<br />

ONC. Intraperitoneal (IP) doses (range of 0-10mg/kg) were given and assessed by<br />

mass spectrometry and immunofluorescence for histone deacetylation and RGC<br />

survival after ONC. DBA/2J mice, which develop glaucoma, were treated IP between<br />

6-10 months with the most effective dose; 2mg/kg every 3 days. Sustained<br />

release of RGFP966 was investigated using intravitreal injection of drug mixed<br />

with microparticles and subcutaneous injection of drug mixed with hydrogel.<br />

Results: A 2µM intravitreal injection of RGFP966 provided transient protection<br />

after ONC, and a 2mg/kg intraperitoneal injection of RGFP966 every 3 days was<br />

optimal for protection against cell loss up to 4 weeks after ONC. Importantly, inhibition<br />

of HDAC3 activity with repeated systemic dosing of RGFP966 protected<br />

against RGC loss in aged DBA/2J mice. Preliminary results indicate that sustained<br />

release of RGFP966 from intravitreally injected microparticles or subcutaneously<br />

injected hydrogel protected against histone deacetylation induced 4 weeks later.<br />

Conclusion: Extended release of HDAC3 inhibitor RGFP966 may serve as a therapeutic<br />

for chronic neurodegenerative diseases such as glaucoma.<br />

160


POSTER BOARD N 63<br />

B2R SIGNALINGIN NEO-ANGIOGENESIS<br />

SANDRA DONNINI, ERIKA TERZUOLI, MARINA ZICHE<br />

Department of Life Sciences, University of Siena, Italy<br />

Purpose: Abnormal retinal vascular permeability is the leading cause of vision loss<br />

in diseases such as diabetic retinopathy, exudative macular degeneration, retinal<br />

vascular occlusions, and others. The main cytokine involved in ocular vascular<br />

permeability is vascular endothelial growth factor (VEGF). VEGF antagonists<br />

have been successfully used as new treatment for diabetic retinopathy,however,<br />

local side effectsand systemic complications have been reported. New therapeutic<br />

approaches to selectivelyblock VEGF angiogenic and permeabilizing actions,<br />

while sparing VEGF protective and trophic actions are needed.Kinins, such as<br />

bradykinin (BK) and kallidin, play a primary role in the development of diabetic<br />

retinopathy by enhancing vascular permeability, leukocytes infiltration, and<br />

other inflammatory mechanisms. These deleterious effects are mediated by kinin<br />

B1 and B2 receptors (B1R and B2R), which are expressed in diabetic human and<br />

rodent retina. In this study we assessed the contributionofB2R signalinginangiogenesis.<br />

Methods and Results: We demonstrated that BK, through the activation of its<br />

B2R, enhances vascular permeability and promotes angiogenesis in in vitro and<br />

in vivo models, which are significantly inhibited by the B2R antagonist, Fasitibant.In<br />

endothelial and circulating pro-angiogenic cells, B2R stimulation elicited<br />

NF-кB activation, leading to COX-2 overexpression, PGE-2 production and VEGF<br />

output. B2R antagonistprevented the BK/NF-кB axis and the ensuing amplification<br />

of inflammatory/angiogenic responses.<br />

Conclusion: Based on our findings,BK/B2Rsystem appears to be involvedin the<br />

controlof angiogenesis, and Fasitibanthasthe propertiestobe furtherstudiedasan<br />

alternative drug intreatmentofdiabetic retinopathyandmacular degeneration.<br />

161


POSTER BOARD N 64<br />

OCULAR PHARMACOKINETICS PROFILE OF PALMITOYLETHANOLAMIDE<br />

ENCAPSULATED IN NEW NANOSTRUCTURED LIPIDIC CARRIER<br />

FEDERICA GERACI 1 , CLAUDIO BUCOLO 1 , CHIARA BIANCA MARIA PLATANIA 1<br />

GIOVANNI LUCA ROMANO 1 , CARMELO PUGLIA 2 , ROSARIO PIGNATELLO 2 ,<br />

EDUARDO MARIA SOMMELLA 3 , PIETRO CAMPIGLIA 3 , CARMINE OSTACOLO 4 , FILIPPO DRAGO 1<br />

1<br />

Department of Biomedical and Biotechnological Sciences, School of Medicine<br />

University of Catania, Catania, Italy, 2 Section of Pharmaceutical Technology, Department of<br />

Drug Sciences, University of Catania Catania, Italy<br />

3<br />

Department of Pharmacy, University of Salerno, Fisciano (SA), Italy<br />

4<br />

Department of Pharmacy, School of Medicine, University Federico II of Naples, Naples, Italy<br />

Purpose: Palmitoylethanolamide (PEA), bearing anti-inflammatory and neuroprotective<br />

properties, is a candidate for treatment of glaucoma and diabetic retinopathy.<br />

PEA, a lipophilic compound, was encapsulated into nanostructured lipid<br />

carriers (NLC). We evaluated the ocular pharmacokinetics profile of a PEA-NLC<br />

formulation in comparison to a suspension of PEA.<br />

Methods: New Zealand rabbits were housed and treated according to the ARVO<br />

statement for the Use of Animals in Ophthalmic and Visual Research. The animals,<br />

randomly assigned to two groups (n=4 per group), received 30 μl eye drops<br />

of either PEA-NLC or PEA-SUSPENSION. Animals were sacrificed at different<br />

time points 30’,60’,120’,180’; and eyes were enucleated. HPLC-MS/MS analysis<br />

was used to measure [PEA] in lens and vitreous.<br />

Results: Loading of PEA into NLCs increased the ocular bioavailability of this lipophilic<br />

compound. PEA loaded in NLC showed higher Cmax, and AUC, both in<br />

lens and vitreous of treated animals, in comparison to PEA delivered as a suspension.<br />

HPLC analysis of lens revealed that AUC of PEA-NLC and PEA-SUSPEN-<br />

SION was 199998±1532 pmol*min/g and 101281±11484 pmol*min/g, respectively.<br />

The AUC of PEA-NLC in vitreous was 203705±5957 pmol*min/g, whereas AUC of<br />

PEA-SUSPENSION was 16046±567 pmol*min/g. PEA-NLC Cmax was 2961±485<br />

pmol/g and 5974±541 pmol/g in lens and vitreous, respectively. Cmax values of<br />

PEA-suspension were 731±135 pmol/g in lens, and 199±30 pmol/g in vitreous.<br />

Conclusions: Our pharmacokinetic data indicate that PEA-NLC formulation increased<br />

significantly the ocular bioavailability of PEA even in the back of the eye.<br />

Therefore, PEA-NLC could be potentially used for treatment of retinal neuroinflammatory<br />

diseases.<br />

162


Organizing Secreteriat<br />

www.medeacom.org

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!