You are on page 1of 109

Evidence-based Approach in Cataract

Surgery
Evidence-based Approach in
Cataract Surgery
Jay Bhopi
MS (Ophthalmology)
Ex-Fellow, Sankara Nethralaya, Chennai
Ex-Consultant, Shri Ganapati Nethralaya, Jalna

LONDON AND NEW YORK


A MARTIN DUNITZ BOOK
© 2004 Jay Bhopi

First published in India in 2004 by Jaypee Brothers Medical Publishers (P) Ltd, New Delhi,
India. EMCA House, 23/23B Ansari Road, Daryaganj, New Delhi 110 002, India Phones:
23272143, 23272703, 23282021, 23245672 m\, Fax: +91–011–23276490 e-mail:
jpmedpub@del2.vsnl.net.in, Visit our website: http://www.jaypeebrothers.com/
This edition published in the Taylor & Francis e-Library, 2005.
To purchase your own copy of this or any of Taylor & Francis or Routledge’s collection of
thousands of eBooks please go to http://www.ebookstore.tandf.co.uk/.
First published in the United Kingdom by Taylor & Francis, a member of the Taylor & Francis
Group in 2004. Exclusively distributed worldwide (excluding the Indian Subcontinent) by Martin
Dunitz, a member of the Taylor & Francis Group.
Tel.: +44 (0) 20 7583 9855 Fax.: +44 (0) 20 7842 2298 E-mail: info@dunitz.co.uk Website:
http://www.dunitz.co.uk/
This edition published in the Taylor & Francis e-Library, 2005.
To purchase your own copy of this or any of Taylor & Francis or Routledge’s collection of
thousands of eBooks please go to http://www.ebookstore.tandf.co.uk/.
All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or
transmitted, in any form or by any means, electronic, mechanical, photocopying, recording, or
otherwise, without the prior permission of the publisher or in accordance with the provisions of the
Copyright, Designs and Patents Act 1988 or under the terms of any licence permitting limited
copying issued by the Copyright Licensing Agency, 90 Tottenham Court Road, London W1P
OLP.
Although every effort has been made to ensure that all owners of copyright material have been
acknowledged in this publication, we would be glad to acknowledge in subsequent reprints or
editions any omissions brought to our attention.
Although every effort has been made to ensure that drug doses and other information are pre sented
accurately in this publication, the ultimate responsibility rests with the prescribing physi cian.
Neither the publishers nor the authors can be held responsible for errors or for any conse quences
arising from the use of information contained herein. For detailed prescribing informa tion or
instructions on the use of any product or procedure discussed herein, please consult the prescribing
information or instructional material issued by the manufacturer.
A CIP record for this book is available from the British Library.

ISBN 0-203-32029-8 Master e-book ISBN

ISBN 1 84184 465 9 (Print Edition)


Distributed in North and South America by Taylor & Francis 2000 NW Corporate Blvd Boca
Raton, FL 33431, USA
Within Continental USA Tel.: 800 272 7737; Fax.: 800 374 3401 Outside Continental USA Tel.:
561 994 0555; Fax.: 561 361 6018 E-mail: orders@crcpress.com
Distributed in the rest of the world (excluding the Indian Subcontinent) by Thomson Publishing
Services Cheriton House North Way Andover, Hampshire SP10 5BE, UK Tel.: +44 (0)1264
332424 E-mail: salesorder.tandf@thomsonpublishingservices.co.uk
This book is dedicated to my wife Suhasini who always encouraged me in writing this
book and also my son Shreyas who allowed me to work on this book daily for long hours.
Preface

There have been rapid advances relating to technology and clinical management in the
practice of ophthalmology. Who would have imagined our present abilities to remove
cataracts using topical anesthesia, through a small self-sealing incision and correcting
vision by implantation of a foldable intraocular lens?
Scientific knowledge and literature is growing exponentially and now time has come
to spread this knowledge. This book would be beneficial not only to the practicing
ophthalmologists for improving their surgical skills but also to resident doctors who are
interested in updating their knowledge. This is a book that should remain in an
ophthalmologist’s shelf for many years.
Jay Bhopi
Contents

1. Newer Anesthesia Modalities for Cataract Surgery 1


2. Newer Incision Techniques in Cataract Surgery 6
3. New Phacoemulsification Technologies 14
4. Recent Advances in Intraocular Lens Material and Design 23
5. Phacoemulsification in Difficult Situations 35
6. Recent Trends in Management of Cataract Surgery Complications 50
7. IOL Power Calculation in Special Situations 60
8. Posterior Capsule Opacification 68
9. Prophylaxis for Postoperative Endophthalmitis 77

Index 92
Introduction

EVIDENCE-GUIDED OPHTHALMOLOGY

Evidence-based medicine is the ability to provide our patients with care, that is based on
the best evidence currently available. It has become necessary for ophthalmologists to
make clinical decisions, based on valid information or evidence rather than intuition,
hearsay and peer practice.
Knowledge about new techniques and approaches collected from peer reviewed
journals provides framework for evidence-guided ophthalmology. Patients have become
very demanding and expect the health care system to offer best practice, based on the
latest evidence.
There are certain distinct advantages of practicing evidence-guided ophthalmology.
1. Evidence-based medicine helps to improve clinical practice by evaluating the quality
of clinical evidence and ensuring that only the best evidence from clinical research is
used in the management of individual patients.
2. Evidence-based medicine has contributed to make the clinicians understand the benefit
and harm of treatment as reported in the literature and it acts as an aid to clinical
decision making.

Getting the Evidence


The best sources for information are the databases maintained by the National Library of
Medicine, such as MEDLINE. After getting a list of references from the search
interfaces, the full text of the articles from peer reviewed journals is necessary to learn
the results of a study.

Evaluating the Evidence


1. The study design should be considered for validity.
2. The methods of the study should be evaluated to assess, to what degree bias,
confounding or chance could have affected the results of the study.
3. The applicability of the results to the practitioner and the patient should be assessed.
4. Finally, all the information is synthesized and assessment-based on the benefits and
risks is considered.
This approach of implementing evidence-based practice will help to maximize the
chances for good patient outcomes.
Abbreviations
1. CCC: Continuous curvilinear capsulorhexis
2. IOL: Intraocular lens
3. CTR: Capsular tension ring
4. HSM IOL: Heparin surface modified intraocular lens
5. PCO: Posterior capsular opacification
6. PcIOL: Posterior chamber intraocular lens
7. Eto: Ethylene oxide
8. ECCE: Extracapsular cataract extraction
9. CME: Cystoid macular edema
10. BSS: Balanced salt solution
11. RK: Radial keratotomy
12. PRK: Photorefractive keratectomy
13. LASIK: Laser-in-situ-keratomileusis
14. LEC: Lens epithelial cell
Chapter 1
Newer Anesthesia Modalities for Cataract
Surgery

Recently, there is a trend toward minimally invasive cataract surgery technique. This has
led to a similar evolutionary process in anesthesia for cataract surgery. In addition to
advances in different techniques of administration of local anesthetic another concept that
has evolved is choice of local anesthetic agent.
1. Lidocaine and bupivacaine: Bupivacaine is added to lidocaine to combine the
advantages of rapid onset of action with lidocaine and longer duration of action with
bupivacaine. But mixing 2% of lidocaine and 0.5% of bupivacaine in a 1:1 ratio
dilutes the anesthetic mixture to 1% lidocaine and 0.25% bupivacaine. This leads to
reduction in efficacy of both anesthetics.
2. pH adjustment of anesthetic solution: Recent studies have demonstrated that pH
adjusted solutions of local anesthetics (pH 6.6–7.0) produce more rapid onset of action
compared to unmodified commercial preparations. The baseline pH of 0.5%
bupivacaine has to be increased from 5.8–6.01 to 6.8–7.0. Hence, 0.08 ml of sodium
bicarbonate has to be mixed in 20 ml vial of 0.5% bupivacaine. This should be freshly
prepared just before giving block.
Mechanism of action: The cation form of any local anesthetic is the active form of
the drug. The local anesthetics exist in various ratios of cation to noncation
concentration, depending on the pH of the drug. Addition of sodium bicarbonate
leads to alkalization of a local anesthetic solution. This alkalization of local
anesthetic increases the availability of the noncation form. This noncation form
penetrates soft tissues and nerve sheath faster. Hence, adjustment of pH increases
nerve penetration and decreases onset time. Tarun Sharma et al1 showed that
bicarbonate buffered 0.5% solution of bupivacaine and hyaluronidase without
lignocaine improves the efficacy of block in patients undergoing vitreoretinal
surgery.
3. Hyaluronidase (5 units/ml): It helps in spreading anesthetic solution within the orbit.
Mechanism of action: It acts by hydrolyzing C1–C2 bonds between glucosamine
and glucuronic acid in ground substance.2
4. Epinephrine: Epinephrine in the concentration of 1:1,00,000 when added to local
anesthetic solution helps in many ways, i.e.
i. It diminishes bleeding from small vessels, if punctured.
ii. It prolongs periorbital akinesia
iii. It reduces orbital volume by causing vasoconstriction.
Evidence-based approach in cataract surgery 2

TOPICAL ANESTHESIA

In the past decade, the most important development in anesthesia for cataract surgery is
introduction of topical anesthesia. Cataract surgery is routinely done under local
anesthesia. It can be grossly divided into injectional and topical.
Retrobulbar, peribulbar, sub-Tenon’s and sub-conjunctival injections of local
anesthetic agent come under “injectional” anesthesia, and topical anesthetic eyedrop
application, sponge anesthesia, eyedrop plus intracameral application and gel application
come under “topical” anesthesia.
The local and systemic complication due to retrobulbar and peribulbar anesthesia are
well-known (Table 1.1). It is now accepted that clear corneal phacoemulsification can be
done under topical anesthesia. However, it is well-known that cataract surgery anesthesia
requires not only analgesia but also akinesia and amaurosis. The newer sub-Tenon’s
technique provide akinesia and amaurosis. There is no evidence in the literature that these
are essential for safe cataract surgery in a cooperative patient (Table 1.2).
Table 1.1: Complications of injectional anesthesia
1. Retrobulbar hemorrhage.
2. Optic nerve damage.
3. Accidental injection of anesthetic agent in retrobulbar blood vessels.
4 Globe perforation.
5. Conjunctival and lid hemorrhage.

The most attractive aspect of this topical anesthesia is its simplicity.


While converting to this technique certain modifications during surgery are required:
a. Preoperative surgeon-patient interaction is required which is not necessary for
conventional injectional techniques. Some surgeons call it “vocal local” and it is in
one way a surgical skill that has to be acquired.
Table 1.2: Contraindications to topical anesthesia
1. Absolute
a Nystagmus
b. Allergy to topical anesthetic eyedrops.
2. Relative
a. Uncooperative patients.
b. Deaf patients.
c. Language barrier between surgeon and patient.
d. Difficult surgery.
e. Prolonged surgery.
Newer anesthesia modalities for cataract surgery 3

b. The brightness of the operating microscope light has to kept low. It is kept at the
lowest level throughout the surgery. This will prevent retinal bleaching and will allow
rapid vision recovery.3

Advantages of Topical Anesthesia*


1. Avoidance of severe complication, though rare, that may occur due to injectional
anesthesia, i.e. globe perforation, retrobulbar hemorrhage, optic nerve damage,
intravascular injection of anesthetic agent.
2. Popularly called “no needle, no patch” technique will allow patients to return home the
same day with better vision.

Topical Eyedrop Anesthesia Alone


The recommended dose is three drops of lidocaine 4% (unpreserved) instilled every 5
minute starting 15 minutes before surgery. Bupivacaine hydrochloride 0.75% can also be
used similarly.4
Johnston et al5 compared topical with peribulbar anesthesia for clear corneal
phacoemulsification. Pain during surgery was slightly more in topical anesthesia group
but it was not statistically significant. None of the patients in both the groups required
supplemental anesthesia or sedation.
Topically applied anesthetics block the trigeminal nerve endings in the cornea and to a
lesser extent, the conjunctiva. The analgesic effect on the iris and ciliary body is very
minimal or none and it depends on penetration of anesthetic agent in the anterior
chamber.
It is documented in literature that patients may subjectively feel pain during surgery
which involve iris manipulation and globe expansion.6 This led to the concept of topical
plus intracameral anesthesia.

Topical Plus Intracameral Anesthesia*


The recommended dosage is 0.5 ml of non-preserved lidocaine 1 % given intracamerally
at the start of surgery. Also, 0.5 ml of bupivacaine hydrochloride 0.5% can be used
similarly.
Safety of intracameral injection of preservative-free lidocaine 1% or bupivacaine
0.5%.
1. Effect on corneal endothelium: There is no difference in the endothelial cell count and
morphology noted after 3 months of surgery.6 This may be due to washout effect
caused by irrigation during phacoemulsification. This limits the exposure of
endothelium to intracameral anesthetics.8

* Fichman first presented his paper on topical anesthesia in American Society of Cataract and
Refractive Surgery (ASCRS) meeting in April 1992 that the ventured back in time to revisit the old
topical anesthesia approach7
Evidence-based approach in cataract surgery 4

2. Retinal toxicity in case of posterior capsular tear: Liang et al9 showed that posterior
movement of intraocularly injected anesthetics in moderate amounts do not cause
long-term adverse effects on retina.
Advantages over topical anesthesia alone: Pain during phacoemulsification with topical
anesthesia alone may be perceived in two circumstances:
i. Deepening of the anterior chamber causes posterior displacement of the iris lens
diaphragm may be perceived as pain.

* Gills first augmented the depth of local anesthesia with intracameral lidocaine injection10
ii. IOL implantation and associated zonular stretching may be perceived as pain.
Intracameral lidocaine causes direct anesthetic effect on the iris ciliary body zonular
complex. Carino et al6 demonstrated that topical anesthesia with intracameral lidocaine is
more effective than topical anesthesia alone.

Lidocaine Gel 2%
Advantages:
i. It provides sustained release of anesthetic hence gives prolonged anesthetic effect.
ii. Single application of gel is enough instead of multiple topical anesthetic drops.
iii. Risk of systemic side effects is less since it is poorly absorbed from ocular mucosa.
iv. Anesthetic effect is comparable to topical plus intracameral lidocaine with double
application of gel.11 There is no effect on endothelial cell count and morphology.12

Sub-Tenon’s (Parabulbar) Anesthesia


Since Tenon’s capsule is an anterior extension of dura, it provides access to the
retrobulbar space.
Procedure: A dissection is made in conjunctiva and Tenon’s capsule down to bare
sclera. A blunt curved, metal cannula is used to inject local anesthetic. A few ml of local
anesthetic is forced to dissect posteriorly to get the required anesthetic effect.
Onset of action: The anesthesia is of rapid onset and it takes few minutes for the globe
akinesia to occur.13
Advantages over peribulbar anesthesia:
i. The chances of globe perforation, retrobulbar hemorrhage, optic nerve damage are
almost nil because of the blunt cannula which is used.
ii. Re-injection of local anesthetics is possible, if surgery time is prolonged as required in
vitreoretinal surgeries.
Newer anesthesia modalities for cataract surgery 5

REFERENCES

1. Sharma T, Gopal L et al: pH adjusted periocular anesthesia for primary vitreoretinal surgery.
Indian J Ophthalmol 1999, 47:223–227.
2. Khurana AK: Peribulbar anesthesia for ocular surgery. In Gupta AK (Ed): Current Topics in
Ophthalmology-IV New Delhi: B-I Churchill Livingstone Pvt Ltd. 1995.
3. Claoue C: Simplicity and complexity in topical anesthesia for cataract surgery. J Cataract Refract
Surg 1998, 24:1546–1547.
4. Risto J Uusitalo, Eeva-Liisa Maunuksela et al: Converting to topical anesthesia in cataract
surgery. J Cataract Refract Surg 1999, 25:432–440.
5. Johnston RL, Whitefield LA, Giralt J, Harrun S, Akerele T, Bryan SJ, et al: Topical versus
peribulbar anesthesia, without sedation, for clear corneal phacoemulsification. J Cataract
Refract Surg 1998, 24:407–410.
6. Carino NS, Slomovic AR, Chung F, Marcovich AL: Topical tetracaine versus topical tetracaine
plus intracameral lidocaine for cataract surgery. J Cataract Refract Surg 1998, 24:1602–1608.
7. Fichman RA: Use of topical anesthesia alone in cataract surgery. J Cataract Refract Surg 1996,
22:612.
8. Anderson NJ, Woods WD, Kim T, Rudnick DE, Edelhauser HF: Intracameral anesthesia in vitro
iris and corneal uptake and washout of 1% lidocaine hydrochloride. Arch Ophthalmol 1999,
117:225–232.
9. Liang C, Peyman GA, Sun G: Toxicity of intraocular lidocaine and bupivacaine. Am J
Ophthalmol 1998, 125:195–196.
10. Gills JP, Cherchio M et al: Unpreserved lidocaine to control discomfort during cataract surgery
using topical anesthesia. J Cataract Refract Surg 1997, 23:545–550.
11. Koch PS: Efficacy of lidocaine 2% jelly as a topical agent in cataract surgery. J Cataract
Refract Surg 1999, 25:632–634.
12. Barequet IS, Soriano ES, Green WR, O’Brien TP: Provision of anesthesia with single
application of lidocaine 2% gel. J Cataract Refract Surg 1999, 25:626–631.
13. Alan S Cradall: Anesthesia modalities for cataract surgery. Curr Opin Ophthalmol 2001, 12:9–
11.
Chapter 2
Newer Incision Techniques in Cataract
Surgery

There has been progressive decrease in size of the cataract incision along with newer
cataract surgical techniques. The size of wound has progressively decreased from 12.0
mm in intracapsular cataract surgery to 10.5 mm in early extracapsular surgery to 5.25
mm with the advent of phacoemulsification. Foldable IOLs has further reduced the size
of incision to 3.0 mm or smaller. Advantages of small size of incision are given in Table
2.1.
Table 2.1: Advantages of small incision
1. Minimal surgical trauma.
2. More stability of anterior chamber.
3. Controlled capsulorhexis and hydrodissection.
4. Healing is rapid hence short postoperative rehabilitation period.
5. Minimal postoperative surgically-induced astigmatism.
6. Management of pre-existing astigmatism is possible by cataract surgery alone.
7. Avoid suture-related complications.
8. Low risk of postoperative leakage.
9. Theoretically low risk of endophthalmitis.

TYPES OF INCISION

The two main types of incision according to anatomic location are as follows:
1. Scleral tunnel incision.
2. Clear corneal incision.

Scleral Tunnel Incision

Concept of Incisional Funnel


Surgically induced astigmatism is inversely proportional to the distance of the incision
constructed from the limbus. This has led to the concept of incisional funnel. Incisional
Newer incision techniques in cataract surgery 7

funnel is made up of two imaginary curved lines which represent the relationship
between induced astigmatism and length of incision. The two curved lines start from the
limbus. They diverge and separate as the distance from the limbus increases (Fig. 2.1).

Fig. 2.1: Incisional funnel

Incision constructed within this imaginary funnel are astigmatically neutral. Hence,
longer incisions should be made away from the limbus to make it astigmatically neutral;
small incisions can be made close to the incision.
Scleral tunnel incision can be (a) curvilinear, (b) straight, (c) frown.
Curvilinear incision: This incision is constructed along the curve of the limbus. It has
highest potential to cause against the rule astigmatism. There is lack of support to prevent
the incision edge from falling away from the superior edge. This gap is the cause for
against the rule of astigmatism.
The boundaries of curvilinear incision fall out of funnel, giving rise to an unstable
wound and greater astigmatism (Fig. 2.2).

Fig. 2.2: Curvilinear incision fall out


of funnel
Evidence-based approach in cataract surgery 8

Straight incision: In this incision, the two terminal points of incision are secured within
the sclera. Hence, the chances of sagging of inferior edge of the incision are minimized.
The straight incision constructed at the same distance as curvilinear incision falls out of
the funnel, but not as much as the curvilinear incision. Hence, the wound is more stable
and induces less astigmatism (Fig. 2.3).
Frown incision: To make the incision more stable, the ends of the incision are placed
further superiorly in the sclera. The ends of the incision are swept superiorly away

Fig. 2.3: Straight incision falls out of


the funnel

from the limbus in a curved fashion1 exactly opposite to that of curvilinear incision. The
chances of sagging of inferior edge of the incision from the superior edge are further
minimized. The frown incision at the same distance as straight incision lie entirely within
the astigmatic funnel hence induces minimal postoperative astigmatism (Fig. 2.4).

Fig. 2.4: Frown incision lie entirely


within the astigmatic funnel
Newer incision techniques in cataract surgery 9

Chevron incision (Fig. 2.5) devised by Sam Pallin2 is similar to that of frown incision
except that it consists of two straight cuts rather than single curved cut.

Fig. 2.5: Chevron incision

Internal Configuration of Scleral Tunnel Incision


Koch’s basic incision (Fig. 2.6):
i. External configuration: It is constructed 0.5 to 1.0 mm from the cornea.
ii. Internal configuration: The groove is dissected upto the limbus and only a small
distance into the cornea before entering the anterior chamber.

Fig. 2.6: Koch’s incision

Advantages:
i. This incision can be performed in transition phase of surgeon, from conventional
curvilinear incision to self-sealing incision.
Evidence-based approach in cataract surgery 10

ii. Being relatively anteriorly situated, it gives easy maneuverability of instruments


during surgery.
Kratz’s scleral pocket incision (Fig. 2.7):
i. External configuration: A vertical groove is made about 2 mm behind the blue zone of
the limbus.
ii. Internal configuration: About half thickness scleral tunnel dissected from the groove
toward the anterior chamber. The anterior chamber is entered with a keratome
according to the width of the phaco tip.
Limitations: The scleral pocket makes easy maneuverability of instruments difficult.
Advantages:
i. More secure wound as compared to Koch’s basic incision
ii. Less surgical induced astigmatism

Fig. 2.7: Kratz’s incision

Three-step corneal valve incision (Fig. 2.8):


i. External configuration: The incision is similar to Kratz’s scleral pocket incision.
ii. Internal configuration: Scleral tunnel dissected from the groove toward the anterior
chamber. This dissection is carried further into the cornea about 1 mm. The anterior
chamber is then entered with a keratome in a vertical fashion, making the third step of
the incision. This third step acts as a valve (corneal). Postoperatively as the intraocular
pressure builds up, the sealing effect of the wound increases.3

Fig. 2.8: Three-step scleral tunnel


incision
Newer incision techniques in cataract surgery 11

Clear Corneal Incision


1. Site of incision: At surgical limbus.
2. Length of incision: Usually 3.0 mm in length. Clear corneal incision of 5.0 mm induces
more surgically-induced astigmatism as compared to scleral frown incision of 5.0
mm.4 But 3.0 mm clear corneal incision induces same amount of astigmatism as with
3.0 mm scleral tunnel incision.5
3. Instrumentation: Diamond blade which has four depth settings and a sharp side cut.
4. Internal configuration: A 200 µm deep precut made, corneal tunnel is constructed
approximately 1.7 to 2.0 mm in length.6

Location of Clear Corneal Incision and Induced Astigmatism


Surgically induced astigmatism is highest after superior followed by superolateral and
lowest after a temporal 3.0 mm clear corneal incision. Corneal flattening of approx 0.7 D
occurs in 3.0 mm temporal clear corneal incision* and approximately 1.2 D in 3.0 mm
superolateral clear corneal incision by 3 months.7
This astigmatism do not extend upto the center of cornea. More corneal flattening in
superior and superolateral incisions as compared to temporal might be due to stroking of
the upper eyelid in the superior and superolateral clear corneal incision.7

Clear Corneal Incision and Management of Pre-existing Astigmatism

A 3.0 mm temporal clear corneal incision induces approx 0.53 D to 0.70 D of temporal
flattening. There is no effect on the nasal-corneal curvature. A 5.2 mm temporal clear
corneal incision can induce 0.84 D of flattening with the rule of astigmatism by one
year.8 Hence, a larger clear corneal incision induces greater flattening effect.9
Hence pre-existing astigmatism upto 1.0 D can be managed by taking clear corneal
incision on the steepest meridian to benefit from its flattening effect. Opposite clear
corneal incision has been suggested when preexisting astigmatism is more than 2.0 D
(Fig. 2.9).

* Fine IH in February 1992 first described a new concept of a planar temporal clear corneal
sutureless incision for phacoemulsification
Evidence-based approach in cataract surgery 12

Fig. 2.9: Opposite clear corneal


incision

In this, clear corneal incision taken at the steepest meridian and similar incision made
exactly opposite to the first to enhance flattening effect.10

INCISION SIZE AND SITE AND CORNEAL ENDOTHELIUM

The scleral tunnel incision causes less postoperative endothelial damage as compared to
clear corneal incision. Scleral tunnel incision being posteriorly placed as compared to
clear corneal incision, there is less direct trauma to endothelium, i.e. by phaco tip and
IOL implantation.11

INCISION SITE AND RISK OF POSTOPERATIVE INFECTION

Recent literature reported increased risk of postoperative endophthalmitis in temporal


clear corneal incision as compared to superior sclerocorneal incision. Its probably due to
bacterial invasion through a temporal corneal incision may be easier than through a
superior corneoscleral incision.12

REFERENCES

1. Singer JA: Frown incision for minimizing induced astigmatism after small incision cataract
surgery with rigid optic intraocular lens implantation. J Cataract Refract Surg 1991,
17(Suppl):677–688.
2. Pallin SL: Chevron sutureless closure: A preliminary report. J Cataract Refract Surg 1991,
17(Suppl):706–709.
3. Grover AK, Gupta AK: Wound construction and closure in modern cataract surgery. In Gupta A
K (Ed): Current Topics in Ophthalmology-IV. New Delhi: BI Churchill Livingstone Pvt Ltd,
1995.
4. Huang FC, Tseng SH: Comparison of surgically induced astigmatism after sutureless temporal
clear corneal and scleral frown incisions. J Cataract Refract Surg 1998, 24:477–481.
Newer incision techniques in cataract surgery 13

5. Oshima Y, Tsujikawa K et al: Comparative study of intraocular lens implantation through 3.0
mm temporal clear corneal and superior scleral tunnel self-sealing incisions. J Cataract Refract
Surg 1997, 23: 347–353.
6. Langerman DW: Architectural design of a self sealing corneal tunnel: Single hinge incision. J
Cataract Refract Surg 1994, 24:84–88.
7. Rainer G, Menapace R et al: Corneal Shape changes after temporal and superotemporal 3.0 mm
clear corneal incision. J Cataract Refract Surg 1999, 25:1121–1126.
8. Nielsen PJ: Prospective evaluation of surgically induced astigmatism and astigmatic keratotomy
effects of various self-sealing small incision. J Cataract Refract Surg 1995, 21:43–48.
9. Hayashi K, Hayashi H et al: The correlation between incision size and corneal shape changes in
sutureless cataract surgery. Ophthalmology 1995, 102:550–556.
10. Lever J, Dahan E: Opposite clear corneal incision to correct pre-existing astigmatism in cataract
surgery. J Cataract Refract Surg 2000, 26:803–805.
11. Beltrame G, Salvetal ML et al: Effect of incision size and site on corneal endothelial changes in
cataract surgery. J Cataract Refract Surg 2002, 28:118–125.
12. Nagaki Y, Hayasaka S et al: Bacterial endophthalmitis after small incision cataract surgery.
Effect of incision placement and intraocular lens type. J Cataract Refract Surg 2003, 29:20–26.
Chapter 3
New Phacoemulsification Technologies

Since Dr Charles Kelman* has invented phacoemulsification it has undergone plenty of


modifications. The recent trend is toward less traumatic cataract surgery and early visual
rehabilitation. This has emerged new phacoemulsification technologies for reducing the
size of incision and using less phaco energy.

CONVENTIONAL PHACOEMULSIFICATION

Conventional ultrasonic phacoemulsification is created in a phacoemulsification


handpiece when an electric current is passed through piezoelectric crystals. These crystals
convert electrical energy to mechanical vibrations at the tip of the phaco-needle. The
phaco-needle tip is used to emulsify the lens material at frequencies between 25 and 62
KHz. Heat is produced by frictional forces. The heat is removed from the eye by fluid
flowing through an irrigation sleeve and around the incision that surrounds the
phacoemulsification needle.1 The irrigation sleeve diameter is larger than the phaco-
needle diameter. In case of diminished flow, conventional ultrasonic phacoemulsification
with an irrigation sleeve has the potential for thermal injury.

MODERN PHACOEMULSIFICATION SYSTEM

Modern phacoemulsification systems has two goals for atraumatic cataract surgery.
1. Reduction of heat produced due to friction during ultrasound phacoemulsification.
2. Reduction of power required for cataract extraction.
To achieve these two goals various modifications in phacoemulsification systems have
been done (Table 3.1).
a. Sonic phacoemulsification system is a new approach for elimination of heat thus
reducing chances of thermal injury.
b. Modification of ultrasound energy through refinement of power modulation is another
route leading to decrease of heat and reduction in incision size (WhiteStar
technology).

* Dr. Charles Kelman got an inspiration for inventing phacoemulsification in the dentist’s chair
while having his teeth, ultrasonically cleaned
New phacoemulsification technologies 15

c. The introduction of innovative oscillatory tip motion in association with power


modulation allows further reduction of phaco power (NeoSonix phacoemulsification
system).
d. Erbium: YAG laser (Phacolase) and Neodymium: YAG laser such as Photon
phacolysis and Dodick photolysis offers new approach for eliminating thermal energy
and decreasing power during phacoemulsification. These two laser systems are being
clinically investigated and clinical trials are underway. And only these lasers are
commercially available.
Table 3.1: Newer phacoemulsification system
1. Sonic phacoemulsification system
2. WhiteStar technology
3. NeoSonix phacoemulsification system
4. Laser systems
a. Erbium: YAG laser system
b. Neodymium; YAG laser phacoemulsification
i. Dodick photolysis
ii. Photon laser phacolysis

Erbium: YAG Laser System


Laser phacoemulsification is one of the emerging technology for removal of cataract.

Advantages Over Ultrasound Phacoemulsification


i. Minimal energy required for removal of cataract.
ii. Decreased chances of thermal injury to the cornea.
iii. The incision size is reduced to 1.0 mm.

Limitations
i. Hard cataract with nuclear sclerosis of grade 3 or more cannot be removed.
ii. Phacoemulsification time is usually longer than the ultrasound phacoemulsification.

Instrumentation
i. Erbium: YAG laser: It has produces a wavelength of 2.94 µm, which falls in infrared
spectrum.
ii. The phacolase Er: YAG laser: It has variable pulse energy from 5 to 50 mJ.
iii. Frequency: It is available in variable frequency from 10 to 100 Hz.
iv. Irrigation/aspiration (I/A) pump: The phacolase system is coupled to a Megatron I/A
pump. The megatron has a peristaltic pump with Venturi-like effect.
Evidence-based approach in cataract surgery 16

v. Handpiece: The phacolase handpiece contains laser fiber inside the aspiration port.
vi. Footswitch: The footswitch is bidirectional which separates irrigation and aspiration
from laser energy. Foot pedal if moved laterally increases the repetition rate in linear
fashion. Foot pedal if pushed down gives control of vacuum linearly.

Mechanism of Action
Cavitation bubble formation: Cavitation is the formation of vacuoles in a liquid by a
swiftly moving solid body. The collapse of the vacuoles releases energy that vaporizes
and crushes lens material (Fig. 3.1).
These cavitation bubbles form and collapse instantaneously. But, the collapse of the
bubbles occurs more slowly in the nucleus. The laser beam travels across the first bubble
and forms a second bubble in line with the first. Similarly if the third bubble is formed,
effective range of the laser is increased to 3.0 mm.

Fig. 3.1: Cavitation buble formation


due to vacuum from
phacoemulsification needle backstroke

Direct concussive effect: Direct concussive effects of laser energy propagation wave
causes disruption of lens material. An emulsate is created which is aspirated from eye.

Neodymium: YAG Laser Phacoemulsification

Dodick Photolysis
Photolysis is a Q-switched Nd: YAG laser introduced first by Dodick in 1991.
Advantages:
i. No heat is produced at the laser tip.2
ii. Silicone sleeve for cooling is not required as no heat is produced thus cataract
extraction is possible through 1.25 mm incision.
Instrumentation: 1064 nm Nd: YAG laser system.
Mechanism of action: Pulsed Nd: YAG laser of energy transmitted through quartz
fiber strikes a titanium target. On this titanium surface there is optical breakdown and
plasma formation. This optical breakdown produces shock waves which move toward the
distal end of the probe which is open. The shock waves here come in contact with the
New phacoemulsification technologies 17

lens material. The shock waves disrupt the lens material at the distal end of the probe and
fragmented material is aspirated out.3 Plasma formation and these shock wave generation
produce photolysis of lens material (Fig. 3.2).

Fig. 3.2: Mechanism of nucleus


removal by Dodick photolysis

Surgical technique
i. Groove and crack technique with the laser.
ii. Sculpting in a bimanual fashion then cracking the nucleus methods are described.

Photon Laser Phacolysis


Instrumentation: 1064 nm Nd: YAG laser, delivered through a special tip of the probe, its
diameter being 1.8 mm. The tip has three functions, i.e. fragmentation, aspiration and
irrigation. Laser energy travels along an optical fiber and also across an open area called
photofragmentation zone. The nuclear material is aspirated into this zone (2.5 mm). The
nuclear fragments which occur are removed from the eye by aspiration and irrigation.
The distal end of the tip is bent upward to provide backstop of the laser energy. This
prevents the laser beam from damaging non-target tissues (Fig. 3.3).4
Evidence-based approach in cataract surgery 18

Fig. 3.3: Mechanism of nucleus


fragmentation using photon laser
photolysis

Sonic Phacoemulsification
Sonic technology uses sonic rather than ultrasonic technology for removing cataract. The
operating frequency is in the range of 40 to 400 Hz. Hence, there is no heat generation
and no generation of cavitational energy. The sonic tip moves back and forth without
changing its dimentional length as against ultrasonic tip motion.
The same handpiece and tip can be used for both sonic and ultrasonic modes. The
modes can be used alternately or simultaneously with varying percentages of sonic and
ultrasonic energy.

NeoSonix Phacoemulsification
NeoSonix technology consists of dual mode comprising of low frequency oscillatory
movement that may be used alone or in combination with conventional high frequency
ultrasonic phacoemulsification.

Instrumentation
In the NeoSonix mode, the phaco tip has a variable rotational oscillation upto 2 degrees at
120 Hz.

Advantage of NeoSonix Handpiece


The lower frequency, as in sonic phacoemulsification does not produce significant
thermal energy and thus minimizes the risk of thermal injury. The NeoSonix thus permits
non-thermal cataract extraction when used alone and with reduced energy when used in
conjunction with ultrasonic energy.
New phacoemulsification technologies 19

Use of NeoSonix Handpiece During Surgery


i. Softer grades of nuclear sclerosis may be completely removed by low frequency
modality and denser grades of cataracts may be removed by additional use of
ultrasound mode.
ii. The low frequency mode may be used to burrow the tip into the nucleus for
stabilization before chopping. This can be done by setting the lower limit of NeoSonix
to 0% phacopower. When straight tip is used, it acts like an apple cores to impale the
nucleus.
iii. NeoSonix handpiece may be used in conjunction with ultrasound at 10 or 20% power
level.

WhiteStar Technology
Recent advances in phacoemulsification are mainly toward decreasing the incision size
for removal of cataract. For this, concept of separating irrigation from aspiration came
forward and removing the cataract through two stab incisions. In conventional
phacoemulsification, irrigation through the same tip helps to keep the temperature down
of the phaco tip, thus preventing wound burn.
WhiteStar micropulse technology is a software modification that allows extremely
short bursts of ultrasound energy. Thus decreases heat build-up with the retained
efficiency of continuous ultrasound.5

Evolution of WhiteStar Technology


It was assessed if the ultrasound is turned on and off in millisecond (ms) and possibly
even submillisecond ranges, whether it will of any practical benefit for removing that
cataract. Experiments were conducted in rabbits. And it was apparent that ultrasound
pulses could be very short as to produce effective phacoemulsification of the nucleus. It
was then evaluated length of downtime (ultrasound off) that can be kept in relation to on
time (ultrasound on) without loosing its efficiency. It was found that down-time could go
to double the on time with minimal and often no sense of loss of efficiency (Fig. 3.4).
Evidence-based approach in cataract surgery 20

Fig. 3.4: Phacoemulsification using


WhiteStar technology showing
phacoemulsification tip without
irrigation sleeve and irrigating chopper
through side port

Advantages
Chatter and microchatter: Chatter is a phenomenon, in which ultrasound energy along
with irrigation flow pushes a nuclear fragment away from the tip. The harder the nuclear
fragment, the more evident the chatter.
Pulsing technology in WhiteStar helps to eliminate chatter. We know that “contact” is
critical in the removal of nuclear fragments. During the down-time in WhiteStar, the
aspirational flow brings the particle back to the ultrasound tip where contact again occurs
which is necessary for removal of nuclear fragment.
Jackhammer effect and cavitational energy: Ultrasound creates a mechanical
Jackhammer effect from the oscillation of the tip, and also creates cavitational energy.
Cavitation is the formation of vacuoles in a liquid by a swiftly moving solid body such as
the ultrasound tip. The collapse of the vacuoles releases energy that vaporizes and
crushes lens material. Cavitational effect is more important of the two in emulsifying
nucleus fragments.
With conventional ultrasound, cavitational energy is greatest when the ultrasound is
first turned on and drops after a few milliseconds of utilization due to air dispersal and
depletion from water (the sourse of cavitation). WhiteStar ultrapulse technology, but
turning off so fast, can minimize this decrease and more efficiently use cavitational
energy than contineous ultrasound.
Fragment followability: Increased followability wherein nuclear fragments stay with
the emulsification tip and dont bounce off, has been reported. Followability is certainly
related to a decrease of microchatter, with the off time allowing aspirational forces to
more efficiently hold the particles in place for eventual emulsification.
New phacoemulsification technologies 21

Thus extremely short bursts of ultrasound energy leads to decrease in chatter and
microchatter, increased fragment followability with improved cavitational energy. Hence,
one can have same efficiency with one-half to one-third the energy utilization in
comparison with cavitational energy.
Absence of thermal energy obviates the need for an irrigation sleeve on the phaco tip,
permitting a reduction in incision size and allowing irrigation through a second
instrument, placed through the side port.

Phaconit
The main difference from conventional phacoemulsification is that phaco tip is used
without the infusion sleeve and separating the irrigation. This allows removal of cataract
through 0.9 mm incision. The function of sleeve is to prevent thermal burns by insulating
and continuously irrigating the vibrating tip and reducing the temperature rise from
ultrasonic vibration. A constant irrigation fluid is required for sufficient cooling of the
phaco-needle.2
Various modifications are made for the following:
a. Adequate cooling of the vibrating tip and
b. To prevent destabilization of the anterior chamber, i.e. surge.
• Irrigating chopper: It has dual function, i.e. anterior chamber maintainer and
chopper.
• Assistant has to continuously pour cooled balanced salt solution over the phaco-
needle.
• Antichamber collapser is used which injects air into the infusion bottle.6 This pushes
more fluid into the eye through irrigating chopper and prevents surge.
• Pump with micropore air filter is used to push sterile air in the bottle.
• Two BSS bottles instead of one is suggested to improve irration inside the anterior
chamber. A Y-shaped infusion (transurethral resection commercially available) is
used to connect these 2 bottles to the irrigating chopper.

REFERENCES

1. Howard V Gimbel, Sandra J Sofinski, Katerina Kurteeva et al: Advantec Legacy system and the
NeoSonix handpiece. Curr Opin Ophthalmol 2003, 14(1):31–34.
2. Alzner E, Grabner G: Dodick Laser phacolysis: Thermal effects. J Cataract Refract Surg 1999,
25:800–803.
3. Dodick JM, Christiansen J: Experimental studies on the development and propagation of shock
waves created by the interaction of short Nd: YAG laser pulses with a titanium target. Possible
implications for Nd: YAG laser phacolysis of the cataractous human lens. J Cataract Refract
Surg 1991, 17:794–797.
4. Murali K Aasuri, Surendra Basti: Laser-assisted cataract surgery and other emerging
technologies for cataract removal. Indian J Ophthalmol 1999, 47:215–222.
5. Radall J Otson, Rajiv Kumar: WhiteStar technology. Curr Opin Ophthalmol 2003, 14:20–23.
6. Agrawal A, Agrawal S, Agrawal A, Lal V, Patel N: Antichamber Collapser. J Cataract Refract
Surg 2002, 28:1085–1086.
Chapter 4
Recent Advances in Intraocular Lens
Material and Design

Intraocular lens material and design is constantly evolving and improving as also
occurring with modern cataract surgery. Improvements in intraocular lens material and
design are aimed at the following:
a. Better refractive correction postoperatively.
b. Smaller incision size.
c. Minimal incidence of posterior capsular opacification.
Various intraocular lens materials have evolved in this process with varying water
content, refractive index and tensile strength. Similarly intraocular lens design has
undergone changes to restrict lens epithelial cell migration and reflection of internal and
external light.1

Sir Harold Ridley*, in 1949, first introduced an intraocular lens. The IOL which Ridley
implanted was of polymethyl-methacrylate (PMMA). Polymethyl-methacrylate is still
considered the standard with which other materials are compared (Table 4.1).

Table 4.1: Various intraocular lens materials


1. Polymethyl methacrylate (PMMA) IOL
2. Silicone IOL (Foldable)
a. Single piece plate type
i. Spherical ii. Toric
b. Three-piece
i. Monofocal ii. Multifocal
3. Acrylic IOL (Foldable)
a. Hydrophobic b. Hydrophilic

* Sir Nicholas Harold Ridley besides inventing an IOL, very few of us know he did pioneering
work in other fields of ophthalmology like definitive characterization of onchocerciasis, first
successful penetrating keratoplasty on a leper, first to televise eye operations and take fundus
photograph, did ground work on power point presentation, used for presentations in seminars and
congresses today39
Evidence-based approach in cataract surgery 24

INTRAOCULAR LENS MATERIALS

Polymethyl-Methacrylate (PMMA)
Intraocular lens made of PMMA has following characteristics:
1. Water content of <1%
2. Refractive index of 1.49, which is higher than silicone but less than some acrylic
copolymers.
3. Inert, light weight, durable and resistant to changes caused by aging.

Limitations
Biocompatibility of PMMA is already proven but with few limitations:
i. Intraocular lenses made of PMMA are rigid hence cannot be implanted through small
(3.2 mm) incision.
ii. Higher incidence of posterior capsular opicification than hydrophobic acrylic and some
silicone IOLS.2–4
However, lens design is considered an important factor for development of PCO than the
lens material.5–7

Silicone IOL
Silicone intraocular lenses are made of polymers of silicone and oxygen. The most
common silicone in IOLs are elastomers baged on the dimethylsiloxane backbone. These
IOLs are relatively inert, non-adherent to tissue and stable at a variable range of
temperatures.

Advantages
Decreased cell adhesion: Silicon’s angle of water contact is 99° which makes it more
hydrophobic when compared to hydrophobic acrylic, which has an angle of water contact
of 73.8 It is known that hydrophobicity may decrease host cell responses. It is found that
giant cell deposition on silicone IOLs is comparable to hydrophobic foldable acrylic
IOLs.9
Newer generations of silicone: They have a higher refractive index of 1.41 to 1-46.
Hence these lenses have thinner optics, resulting in smaller wound size.

Limitations
i. Incidence of posterior capsular opacification—with first generation silicone IOLs, the
incidence of PCO is more than foldable hydrophobic acrylic IOLs and is found to be
equal to that of PMMA. Second generation silicone IOLs have been found to be equal
to hydrophobic acrylic IOLS with truncated edges, for the development of PCO.10,11
Recent advances in intraocular lens material and design 25

ii. Silicone IOLs show a lower threshold for YAG laser damage as compared to
hydrophobic acrylic IOL.12,13
iii. Being slippery, it is difficult to manipulate, especially if wet. During insertion in the
bag it unfolds rapidly, which can lead to uncontrolled insertion and intraocular
damage.
iv. Not suitable for patients with vitreoretinal disease needing silicone oil implantation.
Because there is tendency for adherence of silicone oil to the silicone IOL.14

Designs
There are various designs of silicone IOLs
i. Single piece plate type (spherical and toric)
ii. Three-piece (monofocal and multifocal)

Single Piece Plate Type (Fig. 4.1)


Advantages: It can be inserted through a small incision (3.2 mm) as against PMMA IOL.
Limitations:
i. The large contact area with the anterior capsule makes these lenses more susceptible to
contraction and fibrosis of the anterior capsule and formation of anterior capsule
opacification.15
ii. These IOLs have tendency to dislocate posteriorly after Nd: YAG capsulotomy.16,17

Fig. 4.1: Single piece plate type


silicone IOL

Toric Single Piece Plate Type Silicone IOL


The major requirements for a toric IOL are rotational stability and contraction which is
provided by this IOL. Postoperative toric IOL rotation of 30° off axis still shows
astigmatism reducing effect.18 Plate haptic IOL design does not induce a specific
deviation of rotation (clock wise or anticlockwise) (Fig. 4.2).
Evidence-based approach in cataract surgery 26

Fig. 4.2: Single piece plate type toric


IOL

Factors affecting IOL rotation:


i. Large capsular bag diameter.
ii. Eyes with high axial myopia.
iii. Implantation axis: More rotation of IOL is seen when the axis of IOL implantation is
vertical.18
Preoperative and intraoperative modifications for implanting toric single piece plate type
silicone IOL:
i. Staar SRK/T toric version 1.0 software is used for calculating IOL power.
ii. Before making incision, cylindrical axis is marked on the corneal limbus with the aid
of a Mendez gauge.19

Three-piece Monofocal Silicone IOL


Silicone IOL with squared off and truncated lens edge prevents PCO formation
comparable with that of hydrophobic acrylic IOL. The squared edge is known to create
capsular bend which prohibits migration of lens epithelial cells and thus PCO.20

Multifocal IOL
A pseudophakic patient usually needs spectacle bifocal lenses postoperatively to achieve
best visual acuity for distance and near. A multifocal IOL provides clear vision both for
distance and for near without wearing bifocal spectacle lenses.
Defractive multifocal IOL: Approximately 25 concentric annular zones are cut on the
posterior surface of a conventional IOL with microscopic steps between coterminous
annuli. The stepheight is in the range of the wavelength of light (Fig. 4.3).
Recent advances in intraocular lens material and design 27

Fig. 4.3: Annular concentric zones cut


on posterior surface of IOL optic
The spacing of the rings and the step-profile of these lenses determine the near add. The
closer the rings on the IOL, the higher the add. Thus, with the defractive IOL, both
distance and near objects are simultaneously in focus. Hence, these lenses may result in
excessive glare and halos. These are very difficult to tolerate more so if the IOL is
decentered.
Refractive multifocal IOL: To overcome the problems of glare and halos due to
defractive IOLs, these IOLs have evolved. The AMO Array foldable silicone multifocal
IOL is the single refractive multifocal IOL commercially available. This IOL is a zonal
progressive IOL with five concentric zones on the anterior surface. Zones 1, 3 and 5 are
for distance, whereas zones 2 and 4 are for near. The lens has an aspherical component
and thus each zone repeats the entire refractive sequence corresponding to distance,
intermediate and near foci.21 There is no loss of light through defraction and hence no
degradation of image quality as a result of surface discontinuities.22
The optic of the lens is of silicone which is 6.0 mm in diameters. The haptics are made
of polymethyl-methacrylate and its diameter is 13.0 mm. Hence, the lens can be inserted
through clear corneal or scleral tunnel incision that is 2.8 mm wide.
Refractive multifocal IOL such as Array, is superior to defractive multifocal IOLs by
giving better contrast sensitivity and less glare.23
In conclusion, both distance and near vision correction is possible postoperatively,
decreasing the need for spectacles. Decreased contrast sensitivity and haloes at sight may
be disturbing to patients with multifocal IOLS.24–26 Defractive multifocal IOLs cause
more decrease in contrast sensitivity and haloes as compared to newer refractive
multifocal IOLs (Figs 4.4a and b).
Evidence-based approach in cataract surgery 28

Figs 4.4a and b: Multifocal IOLs. (a)


Defractive IOL, (b) Refractive array
multifocal IOL

Hydrophobic Foldable Acrylic IOL


Hydrophobic acrylic lenses are made of copolymers of acrylate and methacrylate, which
make them flexible. They have refractive index of 1.44 to 1.55 which is higher than
silicone (1.41 to 1.46) and PMMA (1.49). Hence these lenses are thinner.

Comparison with Silicone IOL


i. Hydrophobic acrylic IOLs, because of their lack of elasticity and increased stiffness,
they require larger wounds than similar silicone IOLs.
ii. The acrylic copolymers allow for a slower unfolding action hence provides a more
controlled insertion and manipulation as against silicone IOLs.
iii. The softer nature of this acrylic copolymer makes it fragile and more susceptible to
cracks, dents and scratches.
iv. Hydrophobic acrylic IOLs have bean shown to have the least amount of damage from
the Nd: YAG laser as against silicone IOL.12,13
v. In patients who had underwent vitreoretinal surgery with silicone oil implantation the
acrylic lenses have less problems with adherence with the silicone oil than silicone
IOLs.14
Newer acrylic IOLs have a squared off edge profile. This sharp, truncated edge creates a
capsular bend, preventing migration of lens epithelial cells to form PCO (Figs 4.5a and
b).6
Recent advances in intraocular lens material and design 29

Figs 4.5a and b: (a) IOL with rounded


edge. (b) Hydrophobic acrylic IOL
with sharp, truncated, squared off edge

These squared off edges, however allow for reflection of light internally and thus,
patients report a higher rate of glare, visual aberrations and pseudophakic
dysphotopsia.27,28
Newer designs with a frosted edge or rounded anterior edge may diminish these
unwanted images.

Hydrophilic Acrylic IOL


Hydrophilic acrylic IOLs are composed of a mixture of a hydroxyethyl methacrylate
(poly-HEMA) backbone and a hydrophilic acrylic monomer. These IOLs have varying
water content, and are soft and have excellent relatively hydrophilic lens surface.
1. These IOLs show little or no surface alterations or damage from folding with insertion,
because of their soft flexible surface.29 In terms of the host foreign body response,
they have a low incidence of epitheloid and giant cells deposition.30
2. If a Nd: YAG capsulotomy is needed these lenses have high threshold for Nd: YAG
laser damage.13 These IOLs have low surface energy and in patients requiring
vitreoretinal surgery, these IOLs have minimal adherence to silicone oil.14

Types
Hydrophilic acrylic lenses can be of several types depending on its water content.
Memory lens: It is a poly-HEMA acrylic mixture with a moderate water content and
polypropylene loops. Jehan FS et al31 reported delayed onset, acute, sterile, toxic anterior
segment syndrome (TASS) with these memory lens. These patients present
postoperatively with unexplained inflammation in the anterior segment. Most patients
with TASS improved with intensive topical steroids. Residual polishing compound on
IOL is considered the underlying cause.
Hydroview lenses: Werner L et al reported vision threatening granular deposits of
variable sizes within this lens optics. These granules are distributed in a line parallel to
the anterior and posterior curvatures of the optic, with a clear zone beneath the optic
Evidence-based approach in cataract surgery 30

surface. X-ray spectroscopy revealed presence of calcium within these deposits. The
mechanism of this calcification is still unclear.32

Heparin Surface Modified PMMA IOLs


The surface of the PMMA is modified with heparin. It is found to reduce inflammatory
cell adhesion to the IOL surface. These modified IOLs have better biocompatibility than
PMMA lenses.33
More inflammatory reaction is expected in patients undergoing cataract surgery with
uveitis, diabetes and traumatic cataract, due to breakdown of blood aqueous barriers. It is
found that heparin surface modified PMMA IOLs show reduced adhesion of
inflammatory cells in these cases.33
It is found in one study that metal implantation forces can damage the heparin surface
coating over IOL surface, while insertion. There is increased deposition of inflammatory
cells seen where the heparin surface coating is accidentally removed by forceps. This can
be prevented by using a forceps with silicone sleeves covering the tips.34

INTRAOCULAR LENS IMPLANTATION IN CHILDREN

Ideal IOL Material


Polymethyl-methacrylate (PMMA) has been in use since 50 years now, this long safety
record should not be ignored until similar follow-up is done with other biomaterials.
Foldable hydrophilic material is found to be more biocompatible than PMMA and
silicone. Hence, the next biomaterial which is suitable for pediatric IOLs is hydrophilic
acrylic, i.e. hydroxy ethylmethacrylate (HEMA) and its derivatives.35

Ideal IOL Size


It is not true that large sized IOL is more stable. Oversized and malpositioned IOL can
cause damage to intraocular structures. Pediatric IOLs should not exceed 12.0 mm in
overall diameter because ciliary sulcus rarely exceeds 11.5 mm in diameter.

Factors Deciding the Size of IOL


i. Size of implantation whether in the bag or ciliary sulcus.
ii. Corneal diameter
iii. Age at the time of IOL implantation.
Pediatric IOLs should be in diameters of 10.5 mm, 11 mm and 12 mm.36,37
An example of PMMA pediatric IOL is kidlens (IOL Technologies, France). Its
overall diameter is 10.75 mm, and optic diameter is 5.5 mm (Fig. 4.6a). The haptics are
encircling and contain eyelet holes.38
Another example of pediatric IOL is a palmlens, made of 34% water content HEMA.
Its overall diameter is 10.75 mm, and optic diameter is 5.75 mm (Fig. 4.6b). The flanges
Recent advances in intraocular lens material and design 31

of the lens are structured like the webs of aquatic birds feet or fish fins. These membranes
can absorbs the eventual capsular bag contraction without causing lens valuating. This
lens is easily exchangeable because HEMA material does not adhere strongly to ocular
structures.

Figs 4.6a and b: (a) Kidlens, single


piece PMMA posterior chamber IOL
of overall diameter 10.75 mm. (b)
Palmlens, a foldable posterior chamber
IOL of overall diameter 10.75 mm

Future Intraocular Lens


Thermodynamic injectable IOL (Smartlens Medennium): It is reducible at room
temperature to a rod 2.0 mm in diameter which can be inserted through a microincision.
Once inside the eye, it will unfold to a diameter of 9.5 mm with an average thickness of
2.0 to 3.0 mm. It is a biconvex lens and will fill the entire capsular bag and remain
perfectly centred without the aid of haptics. The gellike hydrophobic acrylic material of
the lens is pliable and has a high refractive index. This means that small changes in the
central thickness will result in large changes in accommodative amplitude. Since, the lens
fills the entire capsule it may prevent the migration of lens epithelial cells and thus
prevents PCO. It is impossible to decenter, no edge glare or spherical aberration.

REFERENCES

1. Richard S Haffman, Howard Fine Mark Packer: Refractive lens exchange with a multifocal
intraocular lens. Curr Opin Ophthalmol 2003, 13:24–29.
Evidence-based approach in cataract surgery 32

2. Hayashi K, Hayashi H, Nakao F, et al: Quantitative comparison of posterior capsule


opacification after polymethyl metharylate, silicone and soft acrylic intraocular lens
implantation. Arch Ophthalmol 1998, 116:1579–1582.
3. Oner FH, Gunenc U, Ferliel ST: Posterior capsular opacification after phacoemulsification
Foldable acrylic versus polymethyl methacrylate intraocular lenses. J Cataract Refract Surg
2000, 26:722–726.
4. Hollick EJ, Spalton DJ, Ursell PG, et al: The effect of polymethyl methacrylate, silicone and
polyacrylic intraocular lenses on posterior capsular opacification 3 years after cataract surgery.
Ophthalmology 1999, 106:49–55.
5. Nishi O, Nishi K: Preventing posterior capsule opacification by creating a discontinuous sharp
bend in the capsule. J Cataract Refract Surg 1999, 25:521–526.
6. Nishi O, Nishi K, Wichstrom K: Preventing lens epithelial cell migration using intraocular lenses
with sharp rectangular edge. J Cataract Refract Surg 2000, 26:1543–1549.
7. Nishi O, Nishi K, Akura J, et al: Effect of round edged acrylic intraocular lenses on preventing
posterior capsule opacification. J Cataract Refract Surg 2001, 27:608–613.
8. Cunanan CM, Ghazizdeh M, Buchen S Y, et al: Contact angle analysis of intraocular lenses. J
Cataract Refract Surg 1998, 24:341–351.
9. Samuelson TW, Chu YR, Kreiger RA: Evaluation of giant cell deposits on foldable intraocular
lenses after combined cataract and glaucoma surgery. J Cataract Refract Surg 2000, 26:817–
823.
10. Hollick EJ, Spalton DJ, Ursell P G et al: Posterior capsular opacification with hydrogel,
polymethyl methacrylate and silicone intraocular lenses: two-year results of a randomized
prospective trial. Am J Ophthalmol 2000, 129:577–584.
11. Hayashi K, Hayashi H, Nakao F, et al: Changes in posterior capsule opacification after
polymethyl methacrylate, silicone and acrylic intraocular lens and implementation. J Cataract
Refract Surg 2001, 27:817–824.
12. Newland TJ, Mc dermott ML, Eliott D, et al: Experimental neodymium: YAG laser damage to
acrylic, polymethyl methacrylate and silicone intraocular lens materials. J Cataract Refract Surg
1999, 25:72–76.
13. Trinavarat A, Atchaneeyasakul L, Udompunturak S: Neodymium: YAG laser damage threshold
of foldable intraocular lenses. J Cataract Refract Surg 2001, 27:775–780.
14. Apple DJ, Isaacs RT, Kent DG, et al: Silicone oil adhesion to intraocular lenses: An
experimental study comparing various biomaterials. J Cataract Refract Surg 1997, 23:536–544.
15. Werner L, Pandey SK, Escobar Gomez M, et al: Anterior capsule opacification: A
histopathologic study comparing different IOL styles. Ophthalmology 2000, 107:463–471.
16. Faucher A, Rootman DS: Dislocation of plate haptic silicone intraocular lens into the anterior
Chamber. J Cataract Refract Surg 2001, 27: 169–171.
17. Petersen AM, Bluth LL, Campion M: Delayed posterior dislocation of silicone plate haptic
lenses after neodymium: YAG Capsulotomy J Cataract Refract Surg 2000, 26:1827–1829.
18. Ruhswurm I, Scholz U, Zehet mayer M, et al: Astigmatism correction with a foldable toric
intraocular lens in cataract patients. J Cataract Refract Surg 2000, 26:1022–1027.
19. Sunday X, Vicary D, Montgomery P, et al: Toric intraocular lenses for correcting astigmatism
in 130 eyes. Ophthalmology 2000, 107:1776–1781.
20. Schmack WH, Gerstmeyer K: Long-term results of the foldable CeeOn Edge intraocular lens. J
Cataract Refract Surg 2000, 26:1172–1175.
21. Richard Hoffman, Howard Fine et al: Refractive lens exchange with a multifocal intraocular
lens. Curr Opin Ophthalmol 2003, 14:24–30.
22. Percival SPB, Setty SS: Prospectively randomized trial comparing the pseudo accommodation
of the AMO Array multifocal lens and a monofocal lens. J Cataract Refract Surg 1993, 19:26–
31.
Recent advances in intraocular lens material and design 33

23. Pieh S, Marvan P, et al: Quantitative performance of bifocal and multifocal intraocular lenses in
a model eye: Point spread function in multifocal intraocular lenses. Arch Ophthalmol 2002;
120:23–28.
24. Javitt JC, Brauweiller HP, Jacobi KW, et al: Cataract extraction with multifocal intraocular lens
implantation: Clinical, functional and quality of life outcome. Multicenter clinical trial in
Germany and Austria. J Cataract Refract Surg 2000, 26:1356–1366.
25. Javitt JC, Steinert RF: Cataract extraction with multifocal intraocular lens implantation:
Clinical, functional and quality of life outcomes. Ophthalmology 2000, 107:2040–2048.
26. Dick HB, Krummenauer F, Schwenn O, et al: Objective and subjective evaluation of photic
phenomena after monofocal and multifocal lens implantation. Ophthalmology 1999, 106:1878–
1886.
27. Ellis M: Sharp edged intraocular lens design as a cause of permanent glare. J Cataract Refract
Surg 2001, 27:1061–1064.
28. Davison J A: Positive and negative dysphotopsia in patients with acrylic intraocular lenses. J
Cataract Refract Surg 2000, 26:1346–1355.
29. Kohnen T. Magdowski G, Koch DD: Scanning electron microscopic analysis of foldable acrylic
and hydrogel intraocular lenses. J Cataract Refract Surg 1996, 22:1342–1350.
30. Mulner-Eidenbock A, Amon M, Schauersberger J et al: Cellular reaction on the anterior surface
of 4 types of intraocular lenses. J Cataract Refract Surg 2001, 27:734–740.
31. Jehan FS, Mamalis N, Spencer TS, et al: Postoperative sterile endophthalmitis (TASS)
associated with memory lens. J Cataract Refract Surg 2000, 26:1773–1777.
32. Werner L, Apple DJ, Kaskaloglu M, Pandey SK: Dense opacification of the optical component
of a hydrophilic acrylic intraocular lens: A clinicopathological analysis of 9 explanted lenses. J
Cataract Refract Surg 2001, 27:1485–1492.
33. Daniele Tognetto, Giuseppe Ravalico: Inflammatory cell adhesion and surface defects on
heparin surface modified polymethyl methacrylate intraocular lenses in diabetic patients. J
Cataract Refract Surg 2001, 27:239–244.
34. Wolder JR, Wenzel MR: Spontaneous breaks in proteinaceous membranes on intraocular
lenses. J Cataract Refract Surg 1991, 17: 617–621.
35. Elie Dahan: Intraocular lens implantation in children. Curr Opin Ophthalmol 2000, 11:51–55.
36. Lam DSC, Ng JSK, Fan DSP, Chua JKH, Leung ATS, et al: Short-term results of scleral
intraocular lens fixation in children. J Cataract Refract Surg 1998, 24:1474–1479.
37. Bluestein EC, Wilson ME, Wang XH, Rust PF, Apple DJ: Dimension of the pediatric
crystalline lens: Implications for intraocular lenses in children. J Pediatr Ophthalmol Strabismus
1996, 33:18–20.
38. Dahan E, Salmenson BD: Pseudophakia in children: Precautions, techniques and feasibility. J
Cataract Refract Surg 1990, 16:75–82.
39. David J Apple, et al: Sir Nicholas Harold Ridley, Kt, MD, FRCS: Contributions in addition to
the intraocular lens. Arch Ophthalmol 2002, 120:1198–1202.
Chapter 5
Phacoemulsification in Difficult Situations

In the last decade several advances have taken place in surgical techniques for removal of
cataracts in difficult situations such as posterior polar cataract, total cataract, subluxated
cataract, and cataract in patients with uveitis. Hard cataract itself can pose a challenge to
surgeons. In addition, phacoemulsification can be difficult if the structures (e.g. weak
zonular apparatus leading to subluxated cataract) and conditions around it (e.g. posterior
polar cataract) are not very favorable.1

PHACOEMULSIFICATION IN HARD CATARACTS

The fibers of a hard nucleus are described as leathery which are cohesive and tenacious.
These fibers resist all the conventional methods of nuclear division like divide and
conquer. There is also risk of hard nuclear fragments coming in contact with corneal
endothelium and damaging it.
Modifications in surgical technique for removal of hard cataracts are as follows:
1. Peribullar anesthesia is preferred over topical anesthesia.
2. There are more chances of burns during phacoemulsification of hard cataracts because
of excessive phacoenergy used. Hence wide temporal clear corneal incision and high
aspiration flow rates are recommended.
3. Double capsulorhexis is recommended, initial smaller one enables phacoemulsification
in the bag followed by larger.
4. Sculpting by Kelman tip is recommended since deep sculpting is possible with it.
5. Creation of central space in the form of trench or crater is necessary. This central space
is necessary for maneuvering the divided lens fragments.
Pure chop technique for nuclear division provides little room for the pieces to be
removed. Especially first nuclear piece is difficult to remove. Vasavada et al has
described step by step chop in situ followed by lateral separation in hard nuclei.2 The
phacoemulsification tip is buried into the cracked nuclear half at 6 O’clock. The chopper
is placed adjacent to the phacoemulsification tip. The vertical element of the chopper is
depressed posteriorly. This leads to initiation of a crack. However, this crack never
reaches the bottom of the nucleus. The chopper is positioned next in the depth of the
crack and pushed laterally. The movement extends the crack to the bottom and nuclear
piece is separated. In this way, fragments of small size are created. The left hand, with a
chopper, continues chopping and dividing the large fragments into small pieces till all the
fragments are phacoaspirated (Fig. 5.1).
Evidence-based approach in cataract surgery 36

Fig. 5.1: Step by step chop in situ and


lateral separation

Posterior Polar Cataract


Surgeons should be careful when operating eyes with posterior polar cataract because the
capsule around the opacity tends to be weak leading to posterior capsule rupture. The
incidence of posterior capsule rupture during phacoemulsification is reported as 26% by
Osher et al3 and 36% by Vasavada et al.4 Serious complications like dropped nucleus can
be avoided if appropriate alternative techniques for cataract surgery are employed.

Continuous Curvilinear Capsulorhexis (CCC) (Fig. 5.2)


Diameter of continuous curvilinear capsulorhexis should not be larger than 5.0 mm. In
the event of posterior capsular tear during phacoemulsification, an IOL can be implanted
in the ciliary sulcus with optic capture through the capsulorhexis.
Phacoemulsification in difficult situations 37

Fig. 5.2: Continuous curvilinear


capsulorhexis not larger than 5.0 mm
in posterior polar cataract

Hydrodissection and Hydrodelineation


Hydrodissection should be avoided as posterior polar opacities adhere firmly to the
posterior capsule around the opacity. If at all it is done, it should be performed gently in
multiple quadrants with minimal fluid. A fluid wave should not be allowed to pass across
the posterior capsule in the center where the posterior capsule is weak. Hydrodelineation
is also performed with minimal fluid. Not more than 0.2 cc of irrigating fluid should be
used for hydrodissection and hydrodelineation (Fig. 5.3).

Fig. 5.3: Careful hydrodelineation in


posterior polar cataract

Phacoemulsification of Nucleus
Infusion bottle should be lowered and extremely low power settings for
phacoemulsification is used.4,5 A fine Nagahara chopper is used to incise the endonucleus
in perpendicular meridians, dividing the nucleus into small quadrants. This should be
done without countertraction and embedding the phacoemulsification tip. The quadrants
are then phacoaspirated.
Evidence-based approach in cataract surgery 38

Epinucleus Removal
Epinucleus can be effectively removed using viscodissection. Viscoelastic is injected
under the anterior capsular margin in one quadrant. The epinucleus is

Fig. 5.4: Viscodissection of the


epinucleus in posterior polar cataract

elevated with viscoelastic and is injected under the floor of epinucleus. Then the central
epinucleus is elevated and aspirated using I/A handpiece (Fig. 5.4).

Cortex Aspiration
The peripheral cortex is aspirated first using I/A handpiece. To avoid fluctuations in
anterior chamber depth an attempt should be made to keep the I/A tip always occluded.
The central posterior portion of cortex is elevated with viscoelastic and aspirated. With
all these precautions, if the defect in posterior capsule is seen, conversion of posterior
defect into posterior capsulorhexis, followed by anterior vitrectomy is necessary. IOL can
be implanted in the ciliary sulcus and optic is captured in the bag.6
Sometimes the opacity comes off spontaneously due to infusion pressure during
removal of the peripheral cortex, but in some cases, part of the opacity remains adhered
firmly to the posterior capsule and could not be separated. In such cases, residual plaque
can be left in place during surgery and later removed by Nd: YAG caplulotomy.6,7 Thus,
the amount of material that can enter the vitreous is reduced.

Phacoemulsification in Subluxated Cataract


Reinforcement of zonules in eyes with zonular dehiscence is necessary before
phacoemulsification . This can be done by the following:
1. Capsular tension ring
2. Nylon hooks.

Capsular Tension Ring


Capsular tension ring is an implantation device made of polymethyl methacrylate
(PMMA) which is used for zonular reinforcement in eyes with weak zonular apparatus.
The causes for weak zonular apparatus can be hereditary like Marfan syndrome, Weill-
Phacoemulsification in difficult situations 39

Marchesani syndrome, homocystinuria, etc. Trauma either blunt or penetrating can lead
to weak zonular apparatus. Other conditions like pseudoexfoliation syndrome and high
myopia can have weak zonular apparatus.8
Designs: The capsular tension ring is composed of an open, flexible, horseshoe shaped
filament of polymethyl methacrylate (PMMA), with fixation eyelets at either end (Fig.
5.5a). It is available in three sizes 12.5, 13.5 and 14.5 mm.

Figs 5.5a and b: (a) Capsular tension


ring, (b) Modified capsular tension
ring

A modification of the ring designed by Cionni consists of a comma shaped filament of


PMMA swedged onto the main ring, emanating at an angle of 90°, at the distal end of this
filament is another eyelet (Fig. 5.5b). This CTR is used with profound zonular deficiency.
Technique of insertion:
i. The CTR can be inserted with forceps or by injectors. Capsular tension ring inserter
consists of a spring loaded plunger assembly with a hook at its distal end. Easiest way
is by Macpherson forceps or a Sinskey hook.
ii. It can be inserted through tunnel or paracentesis.
iii. In profound zonular weakness, a CTR should not be dialed into the capsular bag. In
such cases, the leading fixation hole is positioned at its desired location and then “tire-
ironed” into the capsular bag.
iv. Another method is, a CTR can be inserted above the anterior capsular rim. Eyelets can
be engaged
Evidence-based approach in cataract surgery 40

Fig. 5.6: Capsular tension ring being


“tire-ironed” into the capsular bag
with two Sinskey hooks and drawn toward one another thereby compressing the
ring and delivering it into the capsular bag (Fig. 5.6).
v. For localized zonular dehiscence, a CTR should be positioned so that the body of the
ring is coincident with the weakened zonular fibers.
vi. In profound zonular weakness, the modified ring by Cionni can be sutured to the
scleral wall. Preplaced 10–0 prolene suture taken through eyelet. Each of the two
needles is then placed through the incision and diverted over the area of maximum
zonular dialysis to exit through the ciliary sulcus and scleral wall. The ring is then
inserted in the capsular bag and the fixating element captures itself anterior to the
residual anterior capsular rim (Fig. 5.7).9

Fig. 5.7: Modified capsular tension


ring by cionni sutured to the scleral
wall
Phacoemulsification in difficult situations 41

Time of insertion during surgery: The CTR can be inserted after capsulorhexis, but there
are chances of cortex getting trapped against the capsular bag making cortical clean-up
difficult later.
To prevent this, following steps are advisable:
i. Hydrodissection should be performed .
ii. Aspiration of anterior and equatorial cortex.
iii. A viscoelastic is placed under anterior capsular rim thus displacing the remaining
cortex posteriorly and creating space for the insertion of CTR.10–13

Disposable Nylon Hooks


Disposable nylon iris hooks are placed in the capsulorhexis in the quadrant where zonules
are weak to support the lens. This can be done after capsulorhoxis is performed (Fig.
5.8).14

Fig. 5.8: Disposable nylon iris hooks


placed under the capsulorhexis in
subluxated cataract before
phacoemulsification

Hydrodissection and Hydrodelineation


One has to be extremely careful, multiple locations are used for partial cortical cleaving
hydrodissection. During hydrodissection, the cannula should depress the posterior lip of
the incision. This allows easy outflow of viscoelastic or fluid out of the eye and thus will
prevent overinflation of anterior chamber with irrigating solution.
During phacoemulsification following are extremely useful:
i. Two handed rotations of the lens nucleus is recommended because the forces can be
truely tangential and divided by using opposite sides of the same meridian.
ii. High cavitation tip (e.g. Kelman tip) have a great advantage because they can
obliterate nuclear material in advance of the tip without exerting forces on the lens or
the lens zonules. Variation of phaco sweep procedure can be performed in which
initial groove formed, then without rotating the nucleus, a lateral and rotational motion
of the phaco probe grooves is made in a lateral direction.
Evidence-based approach in cataract surgery 42

iii. Nucleus has to be stabilized during sculpting, through sideport with second
instrument.
iv. Nonrotational cracking is least traumatic method.
v. Cortical aspiration is biggest threat to the zonules. Most of it removed during flipping
and evacuation of the epinuclear shell. Viscodissection can be helpful to separate
cortex from capsule.
vi. Aspiration of residual cortex is safer after the IOL has been implanted because IOL
stabilizes the capsular bag.
vii. Foldable IOL optic with PMMA haptics sized for in-the-bag placement is ideal.
PMMA haptics helps to increase haptic resistance and attempt to prevent capsule
contraction syndrome and lens decentration.12

PHACOEMULSIFICATION IN WHITE CATARACT

In India, mature and hypermature cataracts constitute a significant proportion of


cataracts. Phacoemulsification is ideally suited for immature cataracts in which red reflex
is adequate for a continuous curvilinear capsulorhexis.
The two steps that make phacoemulsification challenging in eyes with white cataract
are as follows:
1. Continuous curvilinear capsulorhexis
2. Emulsification of the hard nucleus—the uses of vital dyes like trypan blue has made it
possible to deliver the benefits of phacoemulsification in these cases.
Continuous curvilinear capsulorhexis technique has improved significantly the safety of
phacoemulsification technique (emulsifying the nucleus safely in the bag). The coaxial
light of the operating microscope produces red fundus reflex which is necessary to
visualize the anterior capsule while performing capsulorhexis. In mature and hypermature
cataracts, this retroillumination is absent. The advancing edge of the anterior
capsulorhexis is very difficult to visualize in this situation. There is risk of peripheral
extension of the advancing edge of the capsulorhexis with its attendant complications.
This can be avoided if the anterior capsule is temporarily stained with contrasting dye
trypan blue (0.1%) that can be used to stain the anterior capsule thus helping its
visualization during CCC.

Techniques
Trypan blue dye can be injected using the following:
i. Classic air bubble technique.
ii. Dispersive viscoelastic material.
iii. Intracameral subcapsular injection.
Phacoemulsification in difficult situations 43

Classic Air Bubble Technique


Steps:
a. Air is injected in the anterior chamber through a 26 gauge needle. Single large uniform
bubble essential for homogenous staining.
b. A 0.2 ml trypan blue (0.1%) injected under air bubble through 26 gauge needle (Fig.
5.9).15 The air allows to spread the dye over the capsule and also prevents dilution of
the dye. The dye is bordered by the peripheral rim of the iris thus preventing direct
endothelial touch.16
c. Anterior chamber is thoroughly irrigated with irrigating solution after 5 to 10
seconds.17

Fig. 5.9: Trypan blue (0.1%) injected


under single large air bubble through
26 gauge needle

Using Dispersive Viscoelastic Material


Alternatively the dye can be injected under viscoat (sodium hyaluronate 3.0%—
condroitin sulfate 4.0%) instead of air bubble. It is possible, to stain the anterior capsule
with concentrations lower than 0.1% by this method.18

Intracameral Subcapsular Injection


The dye is trapped in the subcapsular space, giving sufficient time for the surgeon to
perform CCC. Posterior surface of the anterior capsule is stained hence when capsular
flap is inverted during CCC, it enhances visualization of the advancing edge.
Evidence-based approach in cataract surgery 44

Advantages of Trypan Blue Staining


1. Trypan blue does not stain the corneal endothelium because it selectively stain dead
corneal endothelial cells. Hence, it does not hamper surgical view while
phacoemulsification either by coating the endo thelium or by causing corneal edema.
2. Peripheral stained rim of anterior capsule is visible during phacoemulsification thus
avoiding damage to the capsular rim by phaco tip.

Precautions
Trypan blue being potentially carcinogenic vital dye its lowest effective concentration,
i.e. 0.1% should be used.18 Permanent blue discoloration of hydrogel IOL by
intraoperative use of trypan blue has been reported.19 Hence, hydrogel IOL should be
avoided.
Indocyanine green (ICG) dye can be as effective as trypan blue in staining the anterior
capsule. Its disadvantages being apart from expensive, it has to be reconstituted and its
shelf life is only 10 hours.

Nonstaining Techniques
Various nonstaining techniques for performing CCC in mature and hypermature cataracts
are as follows:
1. CCC using diathermy is described in case of intumescent cataract in absence of red
reflex.20 Its dis-advantage being fibrosis of margins of CCC due to development of
heat and its margins possess less strength than usual CCC margin.
2. Use of endoilluminator described for visualizing anterior capsule during CCC.21
3. Initially, Gimble reported two-step CCC method in which small CCC created first
followed by second CCC around the first. He found phacoemulsification difficult with
small CCC and there were increased chances of rupture of CCC margin during
phacoemulsification.

Emulsification of Nucleus in Total White Cataract


In white cataract, hydrodissection and hydrodelineation are not necessary because the
nuclei are mobile as the corticocapsular adhesions are minimal. Hydrodissection can
make the nucleus excessively mobile by washing out the cortical material.22 For nuclear
fragmentation and aspiration divide and conquer and phaco chop techniques are equally
effective.23 The mature and intumescent cataracts are not very hard, however
hypermature cataracts are relatively hard to emulsify.
Deep anterior chamber should be maintained everytime during phacoemulsification.
This prevents rupture of the posterior capsule as it is not protected by an epinuclear
cushion.15
Phacoemulsification in difficult situations 45

CATARACT SURGERY IN A PATIENT WITH UVEITIS

Cataract surgery in a patient with uveitis is more complex because it involves the
following:
1. Meticulous control of perioperative inflammation.
2. Appropriate surgical timing.
3. Modified surgical techniques to remove cataract in presence of posterior synechia and
undilating pupils.
4. Type and material of IOL to be used.

Preoperative Control of Intraocular Inflammation


Complete abolition of all active inflammation for at least 3 months prior to surgery is
necessary for good surgical outcome in uveitic cataract. One exception for this is protein
leaking lens (phacolytic glaucoma) in which immediate surgery is mandatory.
Supplementary perioperative anti-inflammatory therapy recommended is as follows:
1. One mg/kg/day of prednisone and a drop of 1% prednisolone acetate eight times a day
2 days before surgery.
2. Oral nonsteroidal anti-inflammatory agent twice daily and a topical nonsteroidal anti-
inflammatory agent such as flurbiprofen four times daily.24

Surgical Technique in Uveitic Cataract


A small pupil presents a challenging problem for cataract surgeons. Disposable nylon iris
hooks are most effective means of increasing the size of rigid small pupils. And these are
increasingly being used during phacoemulsification in small pupils.25 Simple stretching
of the iris is effective when pupil diameter is 4 to 5 mm (Fig. 5.10).

Figs 5.10a and b: Disposable nylon


iris hooks for increasing pupil size: (a)
Classic technique, (b) Modified
diamond technique
Evidence-based approach in cataract surgery 46

Technique
Classic technique for using iris hooks: Four flexible nylon iris retractors are used and
inserted at 90° apart from one another. The retractors are placed through 4 paracentesis
wounds at the limbus. These are used to retract the iris, resulting in a pupil of square
shape (Fig. 5.10a).
Modified diamond technique: This technique is a modified form of classic technique,
devised mainly to prevent iris prolapse during surgery through a clear corneal wound.
The phaco incision is shifted 45° to an area just anterior to one of the iris hooks. Thus,
giving the shape of diamond to the pupil instead of square (Fig. 5.10b).

Complications
The use of these iris hooks may lead to certain intraoperative problems:
i. Iris chafing and iridodialysis: In a fully dialated pupil, the iris lies anterior to its
anatomical position, which is posterior to the limbus. Between the two iris hooks there
is wide platform of iris, that is elevated. This can lead to entanglement of an
instrument and iris chafing.
ii. Iris prolapse: It can occur through clear corneal incision in classic method. Hence
subincisional iris hooks with a diamond shaped configuration of the pupil is
recommended.
iii. Damage to anterior capsule: During engagement of pupillary edge with iris hooks,
the tip of the hook may damage anterior capsule. Hence, a bolus of viscoelastic
material has to be injected between iris and capsule.
iv. During phacoemulsification: Small nuclear fragments may get stuck underneath the
iris hooks. This can be prevented by doing in -the- bag phacoemulsification.
v. Localized Descemet’s membrane detachment: This can occur during insertion and
removal of iris hooks hence one has to be gentle while doing this maneuver.26
In addition to adequate pupil dilatation, peripheral iredectomy, synechiolysis are
additional procedures needed in uveitic cataract surgeries.
The anterior capsulotomy recommended is capsulorhexis, because it allows the
surgeon to be certain that the IOL is in-the-bag. Difficulty encountered during
capsulorhexis is irregular fibrotic anterior capsular bands. These tend to inhibit consistent
tearing of the anterior capsule. The capsulorhexis may be extended through the fibrous
bands with capsulotomy scissors and then continued with forceps.
Capsule contraction syndrome27 is found frequently with capsulorhexis
postoperatively. This is frequently encountered with small capsulorhexis. Hence,
capsulorhexis should have a minimum diameter of 5.0 mm. It is found that twice as much
epithelium can be removed with 5.5 mm capsulotomy as with a 4.0 mm capsulotomy.
Some authors proposed a 6.0 mm anterior capsulotomy. It is recommended to do a well-
centered capsulorhexis which should be as circular as possible.
Another fact influencing capsule shrinkage is lens epithelial cells. The more
epithelium is left, the greater the potential for capsule contraction syndrome. Hence,
vacuuming the undersurface of the anterior capsule to remove maximum lens epithelial
cells28 is recommended.
Phacoemulsification in difficult situations 47

The advantages of phacoemulsification are small incision, shorter surgical duration


and thus less surgical trauma. All these may decrease the inflammatory response
compared to ECCE. But carefully done ECCE with complete cortex removal is likely to
achieve similar long-term results.29 In Juvenile rheumatoid arthritis, it is recommended a
combination of surgical procedures like phacoemulsification followed by pars plana
vitrectomy for total removal of lens.30 There are fewer chances of loss of lens fragments
in vitreous with pars plana lensectomy and vitrectomy, though similar results can be
achieved with this technique.

IOL Implantation
A posterior chamber IOL resting on the ciliary body or anterior chamber lens should be
avoided to minimize haptic uveal contact in eyes with uveitis. If the posterior capsule
ruptures to an extent that precludes IOL implantation in the bag, lens implantation should
be aborted. In-the-bag posterior chamber lens is only well-tolerated in patients with
uveitis. PMMA optic with polypropylene haptics should be avoided because
polypropylene haptics are known to cause complement mediated inflammation. Hence,
all polymethylmethacrylate (PMMA) IOLs are recommended.
Heparin surface modified (HSM) IOL has been recommended in patients with uveitis.
Short-term clinical evaluation revealed significantly less anterior chamber reaction and
lower IOL deposits in eyes with HSM IOL. Though, over the long-term, there was no
statistically significant difference in postoperative visual acuity, corneal edema, anterior
chamber reaction, amount of posterior synechia formation and IOL deposits.31
Hydrophobic acrylic IOL has the lowest incidence of aqueous cells in the first
postoperative week as compared with the silicone, hydrophilic acrylic, PMMA and HSM
PMMA IOLs.32 But cell reaction was similar in all these foldable IOL (Hydrophobic
acrylic, hydrophilic acrylic and silicone) after 6 months of surgery.
Hydrophobic material IOL has shown to have a good effect on capsular
biocompatibility33 and a sharp edged optic design reduces most effectively the formation
of PCO in nonuveitic uncomplicated eyes.34 However, in uveitic eyes apart from IOL
material optic edge design, the grade of postoperative inflammation is important factor in
the development of PCO.

Addition of Heparin in the Irrigating Solution


Heparin, in addition to its anticoagulant activity, is known to have anti-inflammatory and
antiproliferative effects. Heparin has been shown to induce apoptosis in human peripheral
neutrophils, which may explain its anti-inflammatory effects. One ml of heparin sodium
adding to irrigating solution which makes a diluted concentration of 10 IU/ml. Adding
heparin sodium to irrigation might help control of easily postoperative inflammation after
cataract surgery. In addition, it is safe and economical. However, it is not advocated as an
alternative to HSM IOLs because the surface modification has a longer acting effect on
flare and the cellular reaction on the IOL.35
Evidence-based approach in cataract surgery 48

REFERENCES

1. Vasavada A, et al: Surgical techniques for difficult cataracts. Curr Opin Ophthalmol 1999,
10:46–52.
2. Vasavada AR, Singh R: Step-by-step chop in situ and aspiration of very dense cataracts J
Cataract Refract Surg 1998, 24:156–159.
3. Osher RH, Yu BC-Y, Koch DD: Posterior polar cataracts: A predisposition to intraoperative
posterior capsule rupture. J Cataract Refract Surg 1990, 16:157–162.
4. Vasavada A, Singh R: Phacoemulsification in eyes with posterior polar cataract. J Cataract
Refract Surg 1999, 25:238–245.
5. Allen D, Wood C: Minimizing risk to the capsule during surgery for posterior polar cataract. J
Cataract Refract Surg 2002, 28:742–744.
7. Ken Hayashi, Hideyuki Hayashi, et al: Outcomes of surgery for posterior polar cataract. J
Cataract Refract Surg 2003, 29:45–49.
6. Howard Fine, Mark Packer, et al: Management of posterior polar cataract. J Cataract Refract
Surg 2003, 29:16–19.
8. Paolo Lanzetta, Raffaella Gortana Chiodiniet: Use of capsular tension ring in
phacoemulsification: Indications and technique. Indian J Ophthalmol 2002, 50:333–37.
9. Cionni RJ, Osher RH: Management of profound zonular dialysis or weakness with a new
endocapsular ring designed for scleral fixation. J Cataract Refract Surg 1998, 24:1299–1306.
10. Kenneth J Rosenthal: The capsular Ring: Indications and surgery. Focal points. Clinical
Modules for Ophthalmologists, September 2002, 20(7).
11. Robert J Cionni, Robert H Osher: Management of profound zonular dialysis or weakness with a
new endocapsular ring designed for scleral fixation. J Cataract refract Surg 1998, 24:1299–
1306.
12. Fine IH, Hoffman RS: Phacoemulsification in the presence of pseudoexfoliation: Challenges
and options. J Cataract Refract Surg 1997, 23:160–165.
13. Menapace R, Findl O, et al: The capsular tension ring: Designs, applications, and techniques. J
Cataract Refract Surg 2000, 26:898–912.
14. John C.Merriam, et al: Ins hooks for phacoemulsification of the subluxated lens. J Cataract
Refract Surg 1997, 23:1295–1297.
15. Jacob S, Agrawal A, et al: Trypan blue as an adjunct for safe phacoemulsification in eyes with
white cataract. J Cataract Refract Surg 2002, 28:1819–1825.
16. Melles GRJ, de Waard PWT, et al: Trypan blue capsule staining to visualize the capsulorhexis
in cataract surgery. J Cataract Refract Surg 1999, 25:7–9.
17. Kothari K, Jain S, Shah N: Anterior capsular staining with trypan blue for capsulorhexis in
mature and hypermature cataract. A preliminary study. Indian J Ophthalmol 2001, 49:177–180.
18. Yetik H, Devranoglu K, et al: Determining the lowest trypan blue concentration that
satisfactorily stains the anterior capsule. J Cataract Refract Surg 2002, 28:988–991.
19. Warner L, Apple DJ, et al: Permanent blue discoloration of a hydrogel intraocular lens by
intraoperative trypan blue. J Cataract Refract Surg 2002, 28:1279–1286.
20. Hausmann N, Richard G: Investigations on diathermy for anterior capsulotomy. Invest
Ophthalmol Vis Sci 1991, 32:155–159.
21. Mansour AM: Anterior capsulorhexis in hypermature cataracts (letter). J Cataract Refract Surg
1993, 19:116–117.
22. Chakarabati A, Singh S, et al: Phacoemulsification in eyes with white cataract J Cataract
Refract Surg 2000, 26:1041–1047.
23. Vasavada A, Singh R, et al: Phacoemulsification of white mature cataracts. J Cataract Refract
Surg 1998, 24:270–277.
Phacoemulsification in difficult situations 49

24. Foster CS, Vitale AT: Diagnosis and Treatment of Uveitis. Philadelphia: WB Saunders
Company, 2002.
25. Mhomas A Oetting, et al: Modified technique using flexible iris retractors in clear corneal
cataract surgery. J Cataract Refract Surg 2002, 28: 596–598.
26. Dadu T, Sethi HS, et al: Using nylon hooks during small pupil phacoemulsification. J Cataract
Refract Surg 2003, 29:412–413.
27. Davison J A: Capsule contraction syndrome. J Cataract Refract Surg 1993, 19:582–589.
28. Nishi O: Intercapsular cataract surgery with lens epithelial cell removal. Part II: Effect on
prevention of fibrinous reaction J Cataract Refract Surg 1989, 15:301–303.
29. Kaufman AH, Foster CS: Cataract extraction in patients with pars planitis. Ophthalmology
1993, 100:1210–1217.
30. Foster CS, Barrett F: Cataract development and cataract surgery in patients with juvenile
rheumatoid arthritis-associated iridocyclitis. Ophthalmology 1993, 100:809–817.
31. Foster RE, Lowder CY, et al: Extracapsular cataract extraction and posterior chamber
intraocular lens implantation in uveitis patients. Ophthalmology 1992, 99:1234–1240.
32. Abela Formanek C, Amon M, et al: Results of hydrophilic acrylic, hydrophobic acrylic, and
silicone intraocular lenses in uveitic eyes with cataract: Comparison to a contract group. J
Cataract Refract Surg 2002, 28:1141–1152.
33. Hayashi H, Hayashi K, et al: Quantitative comparison of posterior capsule opacification after
polymethylmethacrylate, silicon and soft acrylic intraocular lens implantation. Arch Ophthalmol
1998, 116: 1579–1582.
34. Schuersberger J, Amon M, et al: Comparison of the biocompatibility of 2 foldable intraocular
lenses with sharp optic edges. J Cataract Refract Surg 2001, 27:1579–1585.
35. Kruger A, Amon M, et al: Effect of heparin in the irrigation solution on postoperative
inflammation and cellular reaction on the intraocular lens surface J Cataract Refract Surg 2002,
28:87–92.
Chapter 6
Recent Trends in Management of Cataract
Surgery Complications

Technological advances has evolved cataract surgery dramatically over the last two
decades. This increased instrumentation and technology has lead to increased complexity
of cataract surgery and the advent of complications unique to these advances. Cataract
surgeons should be able to avoid, quickly identify and manage these complication
efficiently.

CATARACT SURGERY COMPLICATIONS

Orbital Hemorrhage
Factors that can lead to orbital hemorrhage during injectional anesthesia are as follows:
1. Excessive needle manipulation by the surgeon.
2. Patient movement during block retrobulbar (30%) and less commonly peribulbar
(0.44%)
3. Patients on anticoagulant therapy.1
In patients with bleeding disorders anticoagulation therapy should be discontinued before
surgery. Alternative forms of anesthesia such as topical anesthesia should be preferred in
whom anticoagulation therapy cannot be discontinued.

Management
i. Surgery can be safely done in cases of mild orbital hemorrhage.
ii. In severe cases, orbital hemorrhage produces positive pressure which needs canceling
the surgery.
iii. Very rarely orbital hemorrhage leads to elevation of intraocular pressure which is
enough to threaten vision.2,3
Orbital hemorrhage is considered severe, if
1. There is progressive proptosis.
2. A tight orbit.
3. Increased resistance to retropulsion.
Recent trends in management of cataract surgery complications 51

In such cases, retinal arterioles should be evaluated for flow and pulsations. If retinal
arteriolar pressure is compromised, urgent decompression should be performed. It can be
done by the followings:
i. Dissection of the periocular space with a scissor.
ii. Lateral cantholysis.4

Intraoperative Iris Prolapse with Small Incision Surgery


Intraoperative iris polapse can be very frustrating to the surgeon. Because it leads to the
followings:
i. Risk of trauma to the iris.
ii. Makes introduction of instruments into the anterior chamber difficult.
iii. Distorts the pupil leading to cosmetic havoc.

Causes
i. Increased intraocular pressure from external or internal etiologies.
ii. Incision that is too large or placed too posterior.
iii. Improper placement of speculum.

Management
i. Aspiration of fluid and viscoelastic material from the anterior chamber via the second
incision leads to lowering of intraocular pressure. Then the prolapsed iris can be
gently repositioned.
ii. Judicious use of viscoelastic materials injected in the anterior chamber to maintain the
iris within the anterior chamber.
iii. If this maneuver fails, small peripheral iridectomy should be performed at the site of
prolapse. This neutralizes the pressure gradient between anterior and posterior
chamber.
iv. At the conclusion of surgery a miotic agent is injected intracamerally. This will help
reposition the iris in the anterior chamber.
v. Wound can be secured by placing deep sutures. This reduces the possibility of
spontaneous prolapse with iris incarceration in the wound.

Positive Pressure
Positive pressure can lead to the followings:
1. Shallowing of the anterior chamber.
2. Iris prolapse.
3. Posterior capsule becomes convex, making it more susceptible to tearing.
4. Difficult to aspirate cortex and implantation of IOL.
Evidence-based approach in cataract surgery 52

Cause
1. Tight lids due to narrow palpebral fissures.
2. Inadequate lid akinesia.
3. Excessive traction from a fixation suture.
4. Misdirection of irrigating fluid into the vitreous cavity. This can occur due to a radial
tear in the anterior capsule with extension to the posterior capsule, zonular disruption
or any tear of the posterior capsule.5,6
When positive pressure exists, following maneuvers are recommended:
i. Avoid excessive hydrodissection.
ii. Use highly viscous viscoelastic agents.
iii. Low aspiration rate.
iv. Short bursts of phacoemulsification power.
v. Intravenous mannitol for dehydrating vitreous.

Posterior Capsular Tear


Posterior capsular tear during phacoemulsification is the most significant complication
for a cataract surgeon. Because, it increases the intraoperative risk of dropped nucleus or
lens fragments and postoperative risks of cystoids macular edema and retinal
detachment.7
The main objectives in management of posterior capsular tear are as follows:
1. To prevent vitreous movement into the anterior chamber.
2. To prevent loss of nuclear and cortical material in the vitreous.

Fig. 6.1: Rent in posterior capsule


during phacoemulsification

Immediate steps in case of posterior capsular tear are as follows:


1. Immediate lowering of the infusion bottle
2. Maintaining a stable anterior chamber. Before withdrawing instruments from the
anterior chamber it is necessary to stabilize and replace volume in the anterior
Recent trends in management of cataract surgery complications 53

chamber with irrigating solution or viscoelastic. Viscoelastic agent should be


introduced over the rent (Fig. 6.1).
3. A lens glide is effective in securing the nucleus in the anterior segment.
If additional phacoemulsification is required, low aspiration and irrigation rates, low
bottle height, low vacuum and short bursts of ultrasound power are used. If additional
cortical removal is required, it can be done by the followings:
1. Automated irrigation and aspiration.
2. Manual irrigation and aspiration.
If automated irrigation and aspiration is to be performed:
1. The infusion bottle should be lowered immediately. This will decrease inflow and
turbulence that may enlarge the capsule tear.
2. The irrigation and aspiration tip should be embedded into the cortex before the
initiation of vacuum. This avoids removing the viscoelastic and promoting vitreous
prolapse.
The safest way to remove the remaining cortex is by dry manual irrigation and aspiration.
Cortex, that is too difficult to remove must be left behind. But every attempt should be
made to clear the visual axis.
Surgeons’ new objective should be avoiding the extension of the tear. Hence,
conversion of tear to posterior continuous curvilinear capsulorhexis (PCCC) must be
attempted (Figs 6.2a to c).

Figs 6.2a to c: (a) Closed chamber


vitrectomy through separate incisions.
(b) Posterior CCC done by using
cystitome, (c) Posterior CCC

This has following advantages:


Evidence-based approach in cataract surgery 54

1. It strengthens posterior capsule which allows vitrectomy with decreased chance of tear
enlargement.
2. In-the-bag IOL implantation becomes possible.8

Vitrectomy
If vitreous prolapse occurs, vitrectomy is necessary. During vitrectomy, care must be
taken to preserve anterior capsulorhexis. Vitrectomy can be performed either by anterior
or by posterior approach.
i. Anterior vitrectomy: Anterior vitrectomy can be performed by one-handed or two-
handed technique.
The one-handed technique allows the surgeon to use other instruments in the
second-hand such as light pipe to better visualize the vitreous strands.
The two-handed technique gives better control of the placement of infusion and
thus allows better followability of the aspiration vitrector. In this technique,
vitrectomy cutter should be inserted in the anterior chamber through a new
paracentesis. This will create a closed system and therefore will minimize fluid
outflow through the main incision during vitrectomy.8
ii. Posterior vitrectomy: This is an alternative to anterior approach and has to performed
through pars plana.
iii. Dry vitrectomy: Alternatively, dry vitrectomy is very effective for limiting the amount
of vitreous removed. Anterior chamber is filled with viscoelastic and either an anterior
or posterior vitrectomy is performed behind the viscotemponade.
A suspension of Kenalog (triamcinolone acetonide) can be used to highlight and help
visualize vitreous in the anterior chamber. This is a method of visualizing vitreous gel in
the anterior chamber.
Method: 0.2 ml of injectable triamcinolone 40 mg/ml captured in a 5 mm filter and
rinsed with 2 ml of balanced salt solution (BSS). It is then re-suspended in 5 ml of BSS
and recaptured to thoroughly remove the preservative. The Kenalog particles are
ultimately resuspended in 2 ml of BSS and injected into the anterior chamber using 27
gauge cannula. The Kenalog particles are trapped on and within the vitreous gel, making
it clearly visible.
Thus, helps surgeon in easily identifying and removing the vitreous from the anterior
chamber. This reduces tendency for vitreous strands to remain incarcerated within the
wound after vitrectomy.9

Dropped Nucleus
A dropped nucleus is one of the cataract surgeons biggest fears because of the followings:
1. It requires additional surgery by the vitreoretinal specialist.
2. It increases the risk of postoperative cystoid macular edema, retinal detachment and
persistent ocular inflammation.10,11
Recent trends in management of cataract surgery complications 55

Anticipation of this complication before it happens and pre-emptively works to prevent it


is necessary. Immediately steps necessary on anticipating this complication are as
follows:
1. Infusion should be immediately reduced by lowering the bottle height.
2. Descent of the nucleus and cortical material should be prevented.
If the surgeon decides to convert into extracapsular cataract extraction, following
procedures have to be performed to prevent descent of the lens material.
Viscoelastic is injected behind the nucleus to establish a barrier. Hyperviscous agents
such as sodium hyaluronate* can
1. create a stable shelf to prevent dislocation of the nucleus posteriorly.
2. the use of lens glide to cover the tear may be beneficial.
3. manual removal of nucleus and cortex can be done.
If the surgeon decides to continue with phacoemulsification, Kelman’s posterior assisted
levitation (PAL) technique can be performed to save a dropped nucleus, which is an
emergency maneuver. This has to be performed quickly.

Posterior Assisted Levitation


Posterior assisted levitation is a challenge to the surgeon when during
phacoemulsification, the entire nucleus or part of it starts to sink in the vitreous cavity.
Attempts to retrieve a sinking nucleus by anterior approach usually fails. And, it may
actually push the nucleus further back. In this situation Kelman’s posterior assisted
levitation

* Andre Balaz in the mid 1970s pioneered the first successful viscoelastic sodium hyaluronate
which allowed ophthalmic surgerns to protect corneal ensotheluim during cataract surgery21

(PAL) maneuver is invaluable. Kelman advocates performing this maneuver even on the
suspicion of a posterior capsule rupture (Fig. 6.3).
Evidence-based approach in cataract surgery 56

Fig. 6.3: Posterior assisted levitation


Method: A spaluta is inserted via the pars plana and placed behind the nucleus or its
major remnant. Then it is lifted forward into the anterior chamber. Relaxing incision to
the anterior capsular rim may be required if anterior capsulorhexis is small and its rim is
intact.12
Phacoemulsification can be continued with low settings of flow, bottle height, vacuum
and aspiration. Short bursts of ultrasound energy minimizes the chance of prolonged
occlusion and surge with associated fluctuations in anterior chamber pressure and
volume.
If the lens nucleus disappears then:
i. The cortex should be removed in combination with an anterior vitrectomy.
ii. If the nucleus does not appear during vitrectomy, surgeon can direct stream of
irrigation fluid into the vitreous. If the nucleus becomes visible, it can be recovered by
PAL technique, viscoelevation or by inserting a lens loop behind it.
iii. If the nucleus does not appear, the surgeon should remove cortex as much as possible,
place an appropriate IOL over the anterior capsular rim, secure the wound with sutures
and refer the patient to a vitreoretinal specialist.

Acute Suprachoroidal Hemorrhage


Acute suprachoroidal hemorrhage is one of the most serious vision threatening
complications of cataract surgery. It can be expulsive or nonexpulsive. Expulsive, when
the bleeding pushes choroids and retina out of the wound and nonexpulsive when the
bleeding remains confined within the globe.
Acute suprachoroidal hemorrhage is more likely to occur in older patients, history of
arteriosclerosis, systemic hypertension, diabetes and patients on anticoagulant therapy.13
Recent trends in management of cataract surgery complications 57

Local factors like pre-existing uveitis, glaucoma, high myopia are predisposing factors
for development of acute suprachoroidal hemorrhage.14 An essential factor in the
development of acute suprachoroidal hemorrhage is sudden intraocular hypotension
during surgery. The sudden drop in IOP when the eye is opened, creates a gradient
between intravascular and extravascular pressure in the eye. Hence, incidence of acute
suprachoroidal hemorrhage is more in patients undergoing extracapsular cataract
extraction (0.13%) as compared to patients undergoing phacoemulsification (0.03%).14

Mechanism of Occurrence
Sudden drop in IOP when the eye is opened, causes primary diffusion of fluid from the
veins into the suprachoroidal space creating a suprachoroidal effusion,15 or a primary
hemorrhage when a weakened artery ruptures.15 Or an acute suprachoroidal effusion can
develop into a secondary hemorrhage. The arteries in the suprachoroidal space are
stretched and then rupture because of increased distance between the sclera and the
detached choroids.16

Management
Cataract surgery in patients with all these predisposing factors should be performed using
a small incision technique. If at all it occurs early recognition is important for proper
management. The early signs are as follows:
i. Sudden shallowing of anterior chamber.
ii. Loss of red reflex.
iii. Globe becomes firm, abruptly.
iv. Patient complains of pain in spite of adequate anesthesia.
Once it is detected following steps are recommended:
i. Incision should be closed as quickly as possible. This can be done with manual
tamponade using an index finger. The incision should be closed with strong sutures.17
ii. Administration of mannitol intravenously. This dehydrates the vitreous and decreases
the intraocular pressure.
iii. Creation of sclerotomies to drain the hemorrhage. This is made with stab sclerotomies
in the involved quadrant 5 to 7 mm posterior to the limbus.
iv. The surgery should be aborted once the eye is closed.
Visual activity is often permanently reduced despite aggressive treatment.18

Descemet’s Membrane Detachment


Large Descemet’s membrane detachment results in persistent postoperative corneal
edema with a severe reduction in the patient’s visual acuity.
Evidence-based approach in cataract surgery 58

Prevention
i. Corneal incision are usually triplanar, the phaco tip should be introduced in a similar
fashion. Hence, it is necessary to push the tip posterior when entering the anterior
chamber.
ii. Avoiding forcing an instrument or IOL through a tight incision.
iii. A sharp blade is used to made the incision to avoid detachment of Descemet’s
membrane.

Management
Small Descemet’s membrane detachment:
i. At the conclusion of surgery, it can be reattached by applying pressure on the posterior
lip of the incision at the site of the tear to generate an egress of fluid.
ii. A small air bubble or viscoelastic agent can be used to tamponade a Descemet’s
membrane flap into position during suturing of the wound.
Large or total detachment of Descemet’s membrane:
i. Reattachment of Descemet’s membrane is done by taking transcorneal mattress sutures
to fixate Descemet’s membrane to the cornea in combination to intracameral air
injection.19
ii. Air does not last long enough to hold totally detached Descemet’s membrane. Hence,
intracameral injection of sulfur hexafluoride (SF6) or perfluropropane (C3F8) is most
effective means of managing this complication.20

REFERENCES

1. Davis DB 11, Mandel MR: Efficacy and complication rate of 16, 224 consecutive peribullar
blocks: A prospective multicenter study. J. Cataract Refract Surg 1994, 20:327–337.
2. Wadood AC, Dhillon B, Singh J: Inadvertent ocular perforation and intravitreal injection of an
anesthetic agent during retrobulbar injection. J Cataract Refract Surg 2002, 28:562–565.
3. Bullock JD, Warwar RE, Grein WR: Ocular explosions from periocular anesthetic injections: A
clinical histopathologic, experimental and biophysical study. Ophthalmology 1999, 106:2341–
2345 ; discussion 2352–2353.
4. Goodall KL, Brahma A, Bates A, et al: Lateral canthotomy and inferior cantholysis: An effective
method of urgent orbital decompression for sight threatening acute retrobulbar haemorrhage.
Injury 1999, 30:485–490.
5. Chaudhry NA, Flynn HWJr, Murray TG, et al: Pars plana vitrectomy during cataract surgery for
prevention of aqueous misdirection in high risk follow eyes. Am J Ophthalmol 2000, 129:387–
388.
6. Yeoh R, Theng J: Capsular block syndrome and pseudoexpulsive haemorrhage. J Cataract
Refract Surg 2000, 26:1082–1084.
7. Mulhern M, Kelly G, Barry P: Effects of posterior capsule disruption on the outcome of
phacoemulsification surgery. Br J Opthalmol 1995, 79:1133–1137.
Recent trends in management of cataract surgery complications 59

8. Gimbel HV, Sun R, Ferensowicz M, et al: Intraoperative management of posterior capsule tears
in phacoemulsification and intraocular lens implantation Ophthalmology 2001, 108:2186–2189;
discussion 2190–2192.
9. Burk SE, De Mata AP, Snyder ME, Schneider S, Osher RH, Cionni RJ: Visualizing vitreous
using Kenalog suspension. J Cataract Refract Surg 2003, 29:645–651.
10. Aasuri MK, Kompella VB, Majji AB: Risk factors for and management of dropped nucleus
during phacoemulsification. J Cataract Refract Surg 2001, 27:1428–1432.
11. Lu H, Jiang YR, Grabow HB: Managing a dropped nucleus during the phacoemulsification
learning curve. J Cataract Refract Surg 1999, 25:447–450.
12. Teichmann KD: Posterior assisted levitation. Surv Opthalmol 2002, 47–78.
13. Sekine Y, Takci K, Nakano H, et al: Survey of risk factors for expulsive choroidal haemorrhage
case reports: Substantiation of the risk factors and their incidence. Ophthalmologica 1996,
210:344–347.
14. Eriksson A, Koranyi G, Seregard S, et al: Risk of acute Suprachoroidal hemorrhage with
phacoemulsification. J Cataract Refract Surg 1998, 24:793–800.
15. Maumence AE, Schwartz MF, Acute intraoperative choroidal effusion. Am J Opthalmol 1985,
100:147–154.
16. Manschot WA: The pathology of expulsive hemorrhage. Am J Ophthalmol 1955, 40:15–24.
17. Osher M: Emergency treatment of vitreous bulge and wound gaping complicating cataract
surgery. Am J Opthalmol 1957, 44:409–411.
18. Michael L Nordlund, Daniela MV Marques, et al: Techniques for managing common
complications of cataract surgery. Curr Opin Opthalmol 2003, 14:7–19.
19. Amaral CE, Palay DA: Technique for repair of Descemet’s membrane detachment. Am J
Ophthalmol 1999, 127:88–90.
20. Terry Kim, Saiyid Akbar Hasan: A new technique for repairing Descemet’s membrane
detachments using intracameral gas injection. Arch Ophthalmol 2002, 120:181–183.
21. Pape LG, Balazs EA: The use of sodium hyaluronate (Healon) in human anterior segment
surgery. Opthalmology 1980, 87:699–705.
Chapter 7
IOL Power Calculation in Special
Situations

The refractive results of cataract surgery has greatly improved due to recent advances in
biometry. Accurate measurement of preoperative axial length is required for accurate IOL
power calculation. Postoperative refractive surprises due to error in axial length and
keratometry measurements are common. A 54% of all IOL power calculation surprises
are due to wrong axial length measurements.1 Under ideal circumstances, an error of 1.0
diopter is seen in 15% of eyes.2
The established method for determining axial length is ultrasound A-scan biometry.
Partial coherence interferometry by IOL Master and corneal topography have improved
spherical correction to the point that the new standard of error is only 0.50 diopters.3
Table 7.1: Disadvantages of ultrasound A-scan
biometry for axial length measurement
1. Local anesthetic has to be used.
2. Risk of corneal epithelial abrasion and infection.
3. The resolution of ultrasound is limited to 200 µm (using 10 MHz transducer).
4. Accuracy reported is 100 to 200 µm.4

Recently, noninvasive optical biometry methods for measuring axial length has been
developed. They are based on the principle of partial coherence interferometry (PCI). It is
made commercially available as (IOL Master by Carl Zeiss (Tables 7.1 and 7.2).
Table 7.2: Advantages of partial coherence
interferometry
1. Noncontact method, hence minimal error is avoided due to indentation of cornea as in
ultrasound A-scan biometry method.
2. No topical anesthesia required.
3. Accuracy is reported to be between 5 µm and 30 µm.5

Principle: Principle is similar to conventional ultrasound A-scan which uses ultrasonic


pulse echo-imaging technique. It measures the echo-delay and intensity using infrared
light reflected back from internal tissue interfaces. Since the velocity of light is high,
IOL power calculation in special situations 61

echo-delay times cannot be measured directly and interferometric techniques have to be


used.
The IOL Master can also measure corneal radius and anterior chamber depth. But the
corneal radius and anterior chamber depth measurements are not based on the PCI
principle but on the image analysis principle, in which distances between light reflection
on cornea, iris and lens are measured.6
This IOL Master provides axial length measurements comparable to those by the
immersion method (A-scan) and provides observer independent measurement results
(Table 7.3).
Table 7.3: Limitations of partial coherence
interferometry
1. Measurements not possible in total cataract.
2. Measurements not possible in patients with associated nystagmus.

IOL POWER CALCULATION

IOL Power Calculation after Corneal Refractive Surgery


An increasing number of cataract surgeries in eyes after keratorefractive surgery is
expected within next few decades. Although cataract extraction seams to be possible
without major technical obstacles, IOL power calculation turned out to be problematic.
Feeding of measured average K-reading into standard IOL power calculation formulas
results into overestimation of keratometric diopters, hence underestimation of IOL power
leading to postoperative hyperopia.

Keratometry*
Manual keratometry analyzes only four points on two orthogonal meridians separated 3
mm on the paracentral cornea. Hencc asymmetry of corneal surface cannot be measured
with this keratometry. Automated keratometry was found to be as accurate as manual
keratometry with less variability than manual keratometry.8 A still small central area of
the cornea (2.6 mm) is taken into consideration. In post-RK eyes with optic zone less
than 3 mm, automated keratometers may be more accurate than manual keratometers.
Computerized topography system

* Hermann von Helmholtz had five major accomplishments relating to ophthalmology, i.e.
invention of an ophthalmoscope (1850), confirmation of the Thomas Young theory of color vision
(1852), elucidation of the mechanism of accommodation (1854) and development of an
ophthalmometer (1854).
Ophthalmometer was the first device to measure corneal curvature accurately. Although, his
ophthalmometer was a research tools, later modifications led to clinically useful models like
keratometer7
Evidence-based approach in cataract surgery 62

measures more than 1000 points within the central 3 mm and more than 5000 points over
the entire cornea. Hence topography analysis provides greater accuracy in determining
the corneal power with irregular astigmatism compared with keratometers.
The main reason for underestimation of IOL power after keratorefractive surgery (RK,
PRK and LASIK) lies in inaccurate determination of keratometric diopters.
Cause of inaccurate determination of keratometric diopters in PRK and LASIK:
i. Current keratometers and topography systems primarily measure radius of curvature of
anterior surface of central cornea.
ii. Keratometric diopters are derived from this radius of curvature using an
effective refractive index. Such an effective refractive index characterizes the
refraction of a fictitious single refractive lens representing single anterior and
posterior surface, (*Gullstrands model eye). This effective refractive index is
considered valid as long as radius of anterior and posterior surface of the cornea
are proportionate and resembles that of the model eye (Fig. 7.1).

Fig. 7.1: Anterior corneal curvature,


pre- and post-PRK
iii. In PRK and LASIK, the radius of curvature of anterior surface is considerably
increased and distance between both refractive surface is decreased.
iv. Hence, this method calculating keratometric diopters from anterior radius of curvature
becomes inaccurate.9,10
Causes of inaccurarate determination of keratometric diopters in RK:
i. There is mismeasurement of the anterior radius of curvature, main reason for
inadequate prediction of keratometric diopters.10

* Herman von Helmholtz in his book “Handbook of Physiologic Optics” published the first
accurate model eye. Helmholtz’s numbers were later slightly modified by his student, Alvar
Gullstrand, in the model, that is used today as “Gullstrands model eye”7
IOL power calculation in special situations 63

ii. The location of the keratometry mires which are 3 mm apart from each other fall over
an area of the paracentral “Knee” (Fig. 7.2).
iii. This results in measurement that is too steep.

Fig. 7.2: Anterior corneal curvature,


post-RK

Methods of Estimating Keratometric Power


Indirect method:
a. The clinical history method: This method requires preoperative average reading,
preoperative refraction and stable postoperative refraction. The stable postoperative
refraction should not be due to cataract.
Steps:
i. Change in spherical equivalent refraction (post-refractive surgery refraction—pre-
refractive surgery refraction), e.g. a patient of −10.00 sphere undergoes LASIK for
correction of full −10.00 sphere and has stable postoperative refraction of plano.
SEQ=OD−(−10.00 D)
0+10.00 D
+10.00 D
ii. This has to be subtracted from preoperative keratometric power, (K pre) e.g. 44 D.
K post=K pre−SEQ
K post=44.00 D−10.00 D
K post=34.00 D
iii. The contact lens over refraction method: When pre-refractive surgery data are not
available this method is used. Following parameters are needed to calculate the
postrefractive surgery keratometric diopters.
i. Power (P) and base curve (BC) of rigid contact lens.
ii. Spherical equivalent refraction without contact lens (MRx).
iii. Spherical equivalent refraction with contact lens on (ORx).
Evidence-based approach in cataract surgery 64

The post-refractive surgery keratometric diopters, K post will be


K post=BC+P+(ORx–MRx)
e.g. A plano rigid contact lens of base curve 41.00 D if used and a patient has a
spherical equivalent manifest refraction of −2.00 D and over-refraction of −3.00 D.
K post=41.00+0+[−3−(−2)]
=41.00+0+[−1]
=40.00 D

Direct Method (Ignoring Posterior Curvature Change)


Modern corneal topography machines has various algorithms to calculate estimated
keratometric diopters. They can give power of the anterior corneal surface over a
specified area, e.g. central 3 or 5 mm optic zone.
i. Sim-K: Simulated keratometric diopter or Sim-k measures more than 1000 points over
a 3 mm annular zone.11
ii. Effective refractive power (Eff R P): It is the mean refractive power of the cornea over
the central 3 mm.12
iii. Average corneal power (ACP): Maeda N et al developed the average corneal power
parameter, which looks at the corneal power within the region demarcated by the
entrance pupil of the TMS-1 tomography unit.13
The problem with each of these techniques is that none of them take into account the
posterior curvature, which is believed to be a highly variable between normal individuals.

IOL Power Calculation in Pediatric Cataract


In infants, there is rapid growth of the eye and thus increase in axial length during the
first 2 years of life. This must be taken into account when choosing the diopteric power
of the IOL in infants. In toddlers and older children, the eye continues to grow, although
at a slower pace. As against the axial length, the corneal power drops considerably during
the first 2 years of life.
Gordon RA et al showed a steep axial length growth rate, increasing by 6 mm (~20
D), from premature babies to age 2 years, while corneal power drops from 54 D to 44 D,
offsetting 10 D.14
Between the ages 2 to 5 years axial length growth slows to about 0.4 mm per year and
only increases another 1 mm from 5 to 10 years, while corneal power remains stable.
It is recommended that childrens’ eyes should be undercorrected at the time of
surgery, to offset the myopic shift that occurs in there growing eyes.15–17
Dehan E et al advices to categorize the children as children younger than 2 years and
children older than 2 years of age.15 For children under 2 years they advice to
undercorrect the IOL Power by 20%, since axial length and keratometry readings change
rapidly. For children older than 2 years they advice to undercorrect IOL power by 10%.
This helps in minimizing the need for an IOL exchange later in life, when myopic shift
occurs.

IOL Power Calculation in Patients with High Myopia


IOL power calculation in special situations 65

The various formulas for calculation of IOL power work best for eyes with normal axial
lengths.18–21
Posterior pole staphyloma temporal to the fovea is commonly present in eyes with
axial lengths longer than 30 mm. The distance from corneal vertex to fovea is 0.5 to 1.5
mm shorter than the distance from corneal vertex to the bottom of the staphyloma. The
A-scan usually finds the perpendicular axis between corneal vertex to the bottom of the
staphyloma and records the axial length.22 Hence, current third and forth generation IOL
power calculation formulas have a tendency to give the IOL power lower than what is
necessary, leaving patients with postoperative hyperopia. The use of B-scan
ultrasonography to identify the location of a posterior pole staphyloma is necessary. Few
refinements in preoperative measurement techniques helps to improve the accuracy of
IOL calculation in eyes with extreme myopia.

IOL Power Calculation of Silicone Oil Filled Eyes


This condition arises when vitreous is replaced with silicone oil as in vitreoretinal
surgeries. The refractive index of silicone oil is much less than that of vitreous. Hence, it
acts as negative lens in the eye. This silicone oil refractive effect must be offset with
more power in the IOL. The effect is dependent on the back surface of the IOL. A
biconvex IOL creates the worst problem and a concave posterior surface (no longer
available) causes practically none. Plano convex lens which is between the two is
recommended. With a plano convex lens, 2 to 3 D must be added to the IOL power to
compensate for this effect.23

REFERENCES

1. Olsen T: Sources of error in intraocular lens power calculation. J Cataract Refract Surg 1992;
18:125–129.
2. Annette Vogel, H Burkhard Dick, et al: Reproduction of optical biometry using partial coherence
interferometry: Intraobserver and interobserver reliability. J Cataract Refract Surg 2001,
27:1961–1968)
3. Connors R III, Boseman P III, et al: Accuracy and reproducibility of biometry using partial
coherence interferometry. J Cataract Refract Surg 2002, 28:235–238.
4. Schachar RA, Levy NS, et al: Accuracy of intraocular lens powers calculated from A-scan
biometry with the Echo -oculometer. Opthalmic Surg 1980, 11:856–858.
5. Drexler W, Findl O, et al: Partial coherence interferometry: A novel approach to biometry in
cataract surgery. Am J Ophthalmol 1998, 26: 524–534.
6. Wolfgang Drexler, Oliver Findl, et al: Partial coherence interferometry: A novel approach to
biometry in cataract surgery. Am J Ophthalmol 1998, 126:524–534.
7. Hermann von Helmholtz. A century later Arch Ophthalmol, editorial 1994, 112:1524–1525.
8. Manning CA, Kloess PM: Comparison of portable automated keratometry and manual
kertometry for IOL calculation. J Cataract Refract Surg 1997, 23:1213–1216.
9. Mandel RB: Corneal power correction factor for photorefractive keratometry. J Cataract Refract
Surg 1994, 10:125–128.
10. Gobbi PG, Carones F, et al: Keratometric index, video keratography, and refractive surgery. J
Cataract Refract Surg 1998, 24:202–211.
Evidence-based approach in cataract surgery 66

11. Wilson SE, Klyce SD: Quantitative descriptors of corneal topography: A clinical study. Arch
Ophthalmol 1991, 109:349–353.
12. Holladay JT: Corneal topography using the Holladay diagnostic summary. J Cataract Refract
Surg 1997, 13:209–221.
13. Maeda N, Klyce SD, et al: Disparity between keratometry style readings and corneal power
within the pupil after refractive surgery for myopia. Cornea 1997, 16:517–524.
14. Gordon RA, Donzis PB: Refractive development of the human eye. Arch Ophthalmol 1985,
103:785–789.
15. Dahan E, Drusedau MU: Choice of lens and diopteric power in pediatric psendophakia. J
Cataract Refract Surg 1997, 23:618–623.
16. Hutchinson AK, Drews Botsch C, et al: Myopic shift after intraocular lens implantation during
childhood. Ophthalmology 1997, 104:1752–1757.
17. Flitcroft DI, Knight-Nanan D, et al: Intraocular lenses in children: Changes in axial length,
corneal curvature, and refraction. Br J Ophthalmol 1999, 83:265–269.
18. Huber C. Effectiveness of intraocular lens calculation in high emmetropia. J Cataract Refract
Surg 1989, 15:667–672.
19. Olsen T: Thim K et al: Accuracy of the newer generation intraocular lens power calculation
formulas in long and short eyes. J Cataract Refract Surg 1991, 17:187–193.
20. Olsen T: Sources of error in intraocular lens power calculation. J Cataract Refract Surg 1992,
18:125–129.
21. Olsen T, Corydon L, Gimbel H: Intraocular lens power calculation with an improved anterior
chamber depth prediction algorithm. J Cataract Refract surg 1995, 21:313–315.
22. Roberto Zaldivar, Mitchell C, et al: Intraocular lens power calculations in patients with extreme
myopia. J Cataract Refract surg 2000, 26: 668–674.
23. Kenneth J Hoffer: Modern IOL power calculations Avoiding errors and planning for special
circumstance. Focal points clinical modules for ophthalmologists, 1995 volume XVII, 12.
Chapter 8
Posterior Capsule Opacification

Posterior capsule opacification (PCO) a major post-operative complication following


cataract surgery occurs in 20 to 50% of adult patients. Migration and proliferation of lens
epithelial cells (LECs) onto the posterior capsule is considered the primary step in two
major forms of PCO, posterior capsular fibrosis and Elshnig pearls. Inflammatory cells
also plays a role in formation of PCO, in patients who have significant postoperative
inflammation following cataract surgery.

INHIBITION OF POSTERIOR CAPSULE OPACIFICATION

Posterior capsule opacification being most common complication of primary cataract


surgery, it continues to stimulate important work toward understanding its causes and
thus preventing it from occurring (Table 8.1).
Table 8.1: Factors known to decrease posterior
capsule opacification
1. Trauncated squared posterior IOL edge 360°.
2. Overlap of anterior capsular rim on the anterior IOL surface 360°.
3. Thorough clean-up of cortical matter.
4. Minimum aspiration of anterior capsular lens epithelial cells.
5. Minimum postoperative posterior capsular folds.

Intraocular Lens Material

Sandwich Theory
An IOL can be biocompatible in a bioinert or bioactive way. PMMA and silicone are
bioinert and acrylic is bioactive. For an IOL which is bioactive sandwich theory of PCO
is proposed which is as follows:
i. The anterior rim of continuous curvilinear capsulorhexis forms a bond over the IOLs
bioactive (acrylic) surface. When the bond is complete, the IOL and the capsular bag
are a closed system (Fig. 8.1a).
ii. Another bioactive bond is formed between LECs, posterior capsule and bioactive IOL
(acrylic) which has 90° edge to its optic (Fig. 8.1b).
Posterior capsule opacification 69

The sandwich is formed and the cell-posterior capsule and the cell-bioactive IOL surface
junctions prevent more cells from migrating behind the IOL, thus preventing PCO.

Figs 8.1 a and b: (a) Bioactive bond is


formed between IOL anterior surface
and capsulorhexis. (b) Bioactive bond
is formed between LECs, posterior
capsule and bioactive IOL (acrylic)
which has 90° edge to its optic

Hence without this bond, LEC freely migrate and proliferate behind the IOL, causing
higher rate of PCO, as seen in PMMA and silicone IOL.1

Anterior Capsule Rim Stability and PCO


Continuous curvilinear capsulorhexis (CCC) has become common because it increases
surgical safety during phacoemulsification and allows fixation of IOL in the bag. Overlap
of the anterior capsular rim for 360 on the anterior surface of IOL optic helps in
preventing PCO. A “shrink-wrap” phenomenon is produced due to sequestration of the
IOL in the capsule.12
The relationship of the anterior capsule to the IOL is dynamic, and anterior capsule
contraction or retraction can occur. Initial large CCC≥5.5 mm tends to retract and smaller
than 5.0 mm CCC constrict. This is because, when the CCC is large, there is small area
between the IOL and anterior capsular rim causing the retraction forces to dominate.
Evidence-based approach in cataract surgery 70

With small CCC, relatively more LECs come in contact with the IOL surface and
undergoes fibrous metaplasia, causing a constriction of the capsular opening.
In acrylic IOL, the anterior capsule remains in the same position on the anterior
surface of the IOL from the immediate postoperative phase till 1 year after surgery. The
reason for this is that there is bioadhesion between the anterior capsular rim and the IOL.
This also explains clear anterior capsule and less PCO in acrylic IOL as compared to
PMMA and silicone IOL.3

Intraocular Lens Design


1. A convex plano IOL was found to be superior to the biconvex lens in inhibiting
migration of LECs. A convex plano IOL forms a firm contact between IOL and the
posterior capsule which blocks migration of LECs migration behind the IOL.4
2. The squared off edge of the acrylic IOL creates a sharp bend in the posterior lens
capsule. This prevents migration of the LECs behind the IOL.5 A typical PMMA IOL
with rounded edge do not prevent migrating LECs. But when the edges of PMMA lens
were also squared off, a similar inhibition effect is observed.6 The truncated edges is
equally effective in PMMA, hydrophobic acrylic, hydrophilic acrylic and silicone
IOLs.

Position of Intraocular Lens


The IOL haptics both in-the-bag creates a barrier effect for development of PCO. This is
the best position to place the IOL for preventing development of PCO.7

Surgical Technique
1. Polishing the undersurface of anterior capsular rim and posterior capsule removes
remaining LECs, this helps in preventing PCO. Retained cortex increases
inflammation and inflammation has been shown to increase PCO formation. The
impact of the truncated edge of an IOL is overcome by this retained cortex allowing
lens epithelial cell growth behind IOL.
Conversely, aggressive removal of lens epithelial cells on the anterior capsule
may increase PCO. The reason behind this is, there is decrease in anterior
capsular reaction which is necessary in pushing the truncated edge of IOL over
the posterior capsule thereby producing a discontinuous bend.
2. An in vitro study found no significant difference in the rate of cell growth on the
posterior capsule with the two surgical techniques, i.e. phacoemulsification and
extracapsular cataract extraction.8
3. In pediatric cataract surgeries, of the different methods for management of posterior
capsule and anterior vitreous, the only method which prevents PCO formation is
posterior continuous curvilinear capsulorhexis with anterior vitrectomy. Even
posterior capsulorhexis without vitrectomy with or without optic capture does not
prevent development of PCO.9
Posterior capsule opacification 71

4. A capsule bending ring made of PMMA which has a sharp edge is inserted in the bag.
The idea is to induce contact inhibition of the migrating LECs after cataract surgery,
thus preventing PCO. The ring is inserted

Figs 8.2a and b: (a) Slit lamp


retroillumination photograph. (b) Slit
lamp retroillumination photograph,
wedge is checked if 50% of the wedge
is opacified
Evidence-based approach in cataract surgery 72

Fig. 8.3: Software for quantification of


posterior capsular opacification
into the capsular fornix before IOL insertion. The PCO is found to be reduced in
eyes with this ring.10

Capsular Folds
Folds in the posterior capsule increase the incidence of PCO. The folds occur due to long
haptic length or if the haptics are too stiff. However, haptics that are too short and flaccid
will also result in PCO by not filling out the capsular bag.

ASSESSMENT OF PCO

Amount of PCO can be quantified. Various methods described are as follows:


Posterior capsule opacification 73

1. Photographic image analysis system: Standardized slit lamp retroillumination


photographs are analysed (Figs 8.2a and b, Plate 1). A posterior capsule opacification
score calculated by multiplying the density of the opacification graded from 0 to 4 by
the fraction of the capsule area behind the IOL optic that is opacified.11
2. High resolution digital retroillumination imaging: A system is developed that uses
coaxial illumination and imaging with a digital camera directly linked to a computer
for varification of image analysis (Fig. 8.3, Plate 2).12

Treatment of PCO
Nd: YAG laser posterior capsulotomy is the definitive treatment of PCO.
1. Elschnig pearl formation along the posterior capsulotomy margin after Nd: YAG laser
capsulotomy is a common and significant complication. It can be noted within 1 year
after capsulotomy.
Mechanism of formation: When the posterior capsule is disrupted by the Nd:
YAG laser, intravitreal growth factors can directly affect LECs. Aqueous humor
may dilute these factors to diminish their activities in aphakic eyes. Intraocular
lens implantation and CCC may promote Elschnig pearl formation. This is
because, IOL in the bag, due to compartmentalization of anterior chamber,
hinders the dilution of these factors.13
2. Spontaneous disappearance of Elschnig pearls after Nd: YAG laser posterior
capsulotomy* is described. It may take several years after capsulotomy. Probable
causes are as follows:
• Falling of pearls into the vitreous through the capsulotomy
• Phagocytosis of pearls by macrophages
• Cell death by apoptosis14

* Doctor Danielle Avon Rosa first demonstrated that Nd: YAG laser posterior capsulotomy was a
safe and effective therapy for the treatment of PCO19

Newer Treatments of PCO


Posterior capsule polishing by neodymium: YLF picosecond laser in a model eye is
studied. This eye model consisted of a latex posterior capsule facsmile to demonstrate
micron level polishing to treat posterior capsule opacification. Energy treatment levels
ranged from 5 to 15 ml. The polishing effect was seen with all energy settings. IOL was
found to be safe with energy setting below 5 ml. The authors feel it is a safe and effective
procedure in human eyes also.

Elimination of LECs at Time of Surgery


Caffeic acid phenethyl ester which is the active component of propolis produced by
Honeybees was studied in rabbit eyes.
Evidence-based approach in cataract surgery 74

i. This material shows selective toxicity to certain transformed fibroblasts in carcinoma


cell lines but not to normal cells.15
ii. A new immunotoxin (IMT) is studied for its effectiveness to inhibit PCO following
cataract surgery. The IMT is specific to human LECs and has been shown to be
cytotoxic to these cells in υitro.16
iii. Catalin was examined in rabbit eyes to test its inhibitory effects on PCO. The degree
of PCO was found to be much less as compared to placebo group in which corneal
absorption of catalin is very poor, hence the frequency of its administration to produce
these results should be more.17
iv. Carbon dioxide laser has been used in sheep and rabbits to burn the LECs in the
equatorial and anterior peripheral regions of lens capsule. A major limitation of this
technique is laser probe has to be used under air, because a viscoelastic agent plugs the
hollow probe and absorbs laser energy.18

REFERENCES

1. Linnola R: Sandwich theory: Bioactivity-based explanation for posterior capsule opacification. J


Cataract Refract Surg 1997, 23:1539–1542.
2. Randal J Olson, Nick Mamalis, et al: Cataract treatment in the beginning of the 21st century. Am
J Ophthalmol 2003, 136:146–154.
3. Ursell P, Spalton D, Pande M: Anterior capsule stability in eyes with intraocular lenses made of
polymethylmethacrylate silicone, and Acrysof. J Cataract Refract Surg 1997, 23:1532–1538.
4. Nagamoto T, Eguchi G: Effect of intraocular lens design on migration of lens epithelial cells
onto the posterior capsule. J Cataract Refract Surg 1997, 23:866–872.
5. Nishi O, Nishi K: Posterior capsule opacification, part 1: Experimental investigations. J Cataract
Refract Surg 1999, 25:106–117.
6. Nishi O, Nishi K: Preventing posterior capsule opacification by creating a discontinuous sharp
bend in the capsule. J Cataract Refract Surg 1999, 25:521–526.
7. Ram J, Apple DJ, Peng Q, Visessook N, Auffarth GV, Schoderbek RJ, Jr, et al: Update on
fixation of rigid and foldable posterior chamber intraocular lenses, Part II choosing the current
haptic fixation and intraocular lens design to help eradicate posterior capsule opacification.
Ophthalmology 1999, 106:891–900.
8. Quinlon M, Wormstone I, et al: Phacoemulsification versus extracapsular cataract extraction: A
comparative study of cell survival and growth on the human capsular bag in vitro. Br J
Ophthalmol 1997, 81:907–910.
9. Koch D, Kohnen T: Retrospective comparison of techniques to prevent secondary cataract
formation after posterior chamber intraocular lens implantation in infants and children. J
cataract refract surg 1997, 23: 657–663.
10. Nishi O, Nishik, et al: Capsular opacification a preliminary report: Ophthalmic Surg Lasers
1998, 29:749–753.
11. Tetz MR, Auffarth GU, et al: Photographic image analysis system of posterior capsule
opacification. J Cataract Refract Surg 1997, 23:1515–1520.
12. Pande M, Ursell P, et al: High-resolution digital retroillumination imaging of the posterior lens
capsule after cataract Refract surgery. J Cataract Refract Surg 1997, 23:1521–1527.
13. Kato K, Kurosaka D, et al: Elschnig pearl formation along the posterior capsulotomy. J Cataract
Refract Surg 1997, 23:1556–1560.
14. Cebellero A, Salinas M, et al: Spontaneous disapperance of Elschnig pearls after neodymium:
YAG laser posterior Capsulotomy. J Cataract Refract Surg 1997, 23:1590–1594.
Posterior capsule opacification 75

15. Hepsen I, Bayramlar H, et al: Caffeic acid phenethyl ester to inhibit posterior capsule
opacification in rabbits. J Cataract Refract Surg 1997, 23:1572–1576.
16. Clark DS, Emerg IM, et al: Inhibition of posterior capsule opacification with an immunotoxin
specific for lens epithelial cells: 24-month clinical results. J Cataract Refract Surg 1998,
24:1614–1620.
17. Biswas WR, Mongre PK, et al: Animal study on the effects of catalin on after cataract and
posterior capsule opacification. Ophthalmic Res 1999, 31:140–142.
18. Michacli-Cohen A, Belkin M, et al: Prevention of posterior capsule opacification with the CO2
laser. Ophthalmic Surg Lasers 1998, 29: 985–990.
19. Avon Rosa D, Avon JJ, et al: Use of the neodumium: YAG laser to open the posterior capsule
after lens implant surgery: A preliminary report. Am Intraocular Implant Soc J 1980, 6:352–
354.
Chapter 9
Prophylaxis for Postoperative
Endophthalmitis

MODERN OPHTHALMIC OPERATING ROOM AND INSTRUMENT


STERILIZATION TECHNIQUES

PREVENTION OF INFECTIONS IN OPERATING ROOM

Prevention of postoperative endophthalmitis is the final goal in any cataract surgery,


Hence maintenance of asepsis in operating room rather operating room complex is
important.
The prevention of surgical infection in the operating theater is a complex pursuit.
These many facets that may be divided into four main components as in Table 9.1.

Location of Operating Room Complex


Operating room complex should be located on a separate floor (top floor) of a building or
at least in a separate wing of a floor. It should be away from wards where indoor patients
are admitted to avoid cross-contamination. Operating room should be exclusively for
ophthalmic surgical procedures and should never be shared with other surgical
specialities.4 Toilets for both patients and operating room staff should not be in operating
room complex.
Window air conditioner is preferable over central air conditioning since the filter from
window air conditioner can be easily removed and cleaned regularly.5
Design of the operating room complex should be such that asepsis maintenance can be
managed easily in all its four areas, i.e. changing room, scrubbing area, transfer area and
operating room proper.
There should be a separate room for changing clothes. It should be situated at the
entrance of the operating room complex. It should have separate entrance and exit doors.
Entrance door from where one enters with usual clothes and exit through which one
enters inside the operating room complex with operation room dress, cap, mask and
slippers.
Operating room should have scrubbing area which is situated just outside. The length
of the scrubbing area should be such that one surgeon and one assistant can scrub
simultaneously without touching each other elbows. The washbasin should be at waist
height so as to avoid the arms touching the basin. The tap should have a long handle for
opening or closing with ones elbow with arms folded. Running tap water from an
overhead tank preferably through a filter (Aquaguard) should be used for hand scrubbing.
Evidence-based approach in cataract surgery 78

And stored water in a container should never be used. Opening of an overhead tank
should always be kept closed and its regular cleaning is necessary.
Use of brush during scrubbing should be discouraged. As it itself can cause minute
skin abrasions which can harbor bacteria. Washbasin should have a sufficient depth so
that water running down from scrubbed hands should not spillover it again (Fig. 9.1).
There should be separate entrance to the patient in the operating room complex. This
area contains a redline drawn on the floor. This redline demarcates the area where patient
is shifted from outer trolley to inner one. The operating room proper* should not be more
than 20×20 feet in length and breadth. It should have one entrance door and a separate
service window for receiving sterilized surgical trays and instruments and disposing them
after use.
The door should be swing door, so that it remains closed all the time. To enable proper
cleaning and disinfection, dirt resistant less porous material (granite or marble) should be
used for flooring and walls with minimum joints. The head end of the operating table
should be away from the entrance door and it should be placed in such a way that there is
enough space around it at all sides. Each operating room should not have more than one
operating table. Operating room door should be closed from inside just before starting the
surgery and opened only after completion of surgery.8 If multiple serial surgeries are to
be performed instrument tray for next surgery should be opened only after shifting
previous patient outside and next patient inside the operating room.

Sterile Corridor
This area has operating room on one side and sterilization room on other side. The
sterilized instrument trays and linen can be supplied from the sterilization room to the
operating room through the service window (Fig. 9.2).

Operating Room Environment


Circulating air entering inside the operating room should be filtered air. The high
efficiency particulate air (HEPA) filter system removes microorganisms ranging in size
from 0.5 to 5.0 µ. As sizes of most bacteria and fungi are in the range of 0.5 to 5.0 µ
diameter. Frequent opening and closing of doors should be avoided. The operating room
door should be always kept closed. Hairs of operating room personal should be properly
covered under the cap. Number of people inside the operating room should be kept
minimum.9,10 Temperature inside the operating room should be 18 to 24°C and humidity
of 55 to 80%.

Cleaning and Disinfection of the Operating Room


Disinfectants can be divided into high level, intermediate level and lower level
disinfectants. Glutaraldehyde 2%, formaldehyde 6% and hydrogen peroxide 6% are
considered as high level disinfectants. Phenol is considered as intermediate level
disinfectant. Alcohols 70 to 90%, sodium hypochlorite and quaternary ammonium
compounds are considered as lower level disinfectants.
Prophylaxis for postoperative endophthalmitis 79

Low level disinfectants are effective against bacteria, fungi, lipophilic viruses but not
against spores and tubercle bacilli. An intermediate level disinfectant destroys all these
and tubercle bacilli but not spores. A high level disinfectant destroys all these and also
spores. Rutala WA divided operating room items into critical, semicritical and noncritical
for determining which level of disinfectant needed for which item.11
High level disinfectant needed for critical items like operating instruments, also high
level disinfectant needed for semicritical items, i.e. instrument which touches mucous
membrane, e.g. suture removal forceps, Bowman’s lacrimal probe, punctum dilator and
intermediate to low level disinfectant for noncritical items which touches intact skin.11
Frequent wet mopping of all hard surfaces using a phenolic solution is recommended
to keep the operating room environment clean.

Regimen for Frequency of Decontamination in the Operating Room


In the morning, before shifting first patient inside the operating room, items
recommended for cleaning are operating table mat and sides, instrument trolleys, stools
and chairs and floor.
After each patient’s surgery is over, cleaning of operating table mat and sides,
instrument trolleys and floor is necessary. Infected cases should be operated in separate
operating room meant for the same. If the operating room is soiled or contaminated due
to any reason, recommended items for cleaning are entire operating table, chair, stools,
anesthesia equipment and walls.12 Washing of operating room walls, floor, tables and
trolleys with detergent should be done weekly. This enhance the effect of daily cleaning
and disinfection.
Operating room should be fumigated with formal-dehyde once in fortnight or after
surgery on an infected case. Samples should be taken from various areas in the operating
room for culture of microorganisms after fumigation.

Formaldehyde Fumigation
Before fumigation, adhesive tape is applied to all apertures in doors and windows of the
operating room. Consider operating room of dimension 10 feet×10 feet of length and
breadth and 10 feet in height. One litre of water is mixed with 500 ml of formaldehyde
(40%). This mixture is put into an electric boiler or a large bowl is kept over an electric
hot plate. After turning on the boiler or hot plate, the operating room is sealed.For the
next 8 to 10 hours, the operating room is kept closed. To neutralize the formaldehyde,
ammonium solution is introduced and kept in the room for another few hours. For every 1
litre of 40% formaldehyde used, one litre of water plus one litre of ammonium solution
used.13

Alternative Methods for Fumigation


Permanganate method: Operating room of 1000 cu ft space, 5 ounces (1 ounce=28.2 gm)
of potassium permangnate is placed in a jar, 10 to 15 ounces of formalin (40%) diluted
with equal amount of water is poured over it. As soon as these reagents are mixed, a
violent effervescence takes place and formaldehyde is set free.
Evidence-based approach in cataract surgery 80

Paraform method: Formalin when heated, the aldehyde changes into the solid
polymeride paraform. Gas is generated by heating paraform tablets. For 1000 cu feet
space of operating room 30 tablets are needed.
Formalin spray: In this method 250 cc formalin (40%) is mixed with 5 liters of tap
water. This makes dilution of 1:20. Aeromax vaporizer is used to fumigate an operating
room in this method. This is vaporized for 30 minutes.
Disadvantages: Spraying is not a satisfactory method as compared to vaporization of
formaldehyde by boiling because fine aerosol has poor penetration.
In the west, formalin fogging is no longer a preferred method because of its
carcinogenic nature. Manual cleaning with high level disinfectant is considered more
effective infection control measure.12

STERILIZATION PROCESSES

Sterilization processes can be divided into following heads (Table 9.2).

Steam Sterilization
Steam sterilizers are of two types (Figs 9.3a and b):
a. Downward displacement sterilizer (Gravity displacement sterilizer).
b. Prevacuum sterilizer.
The downward displacement sterilizer displaces air from the chamber and load (packed
instrument tray to be autoclaved) by gravity. Prevacuum sterilizer displaces air from the
chamber and load by vacuum pump. Hence, sterilization is much faster in prevacuum
sterilizer than gravity displacement sterilizer because speed of air removal is faster in the
prevacuum sterilizer.
Microprocessor control (i.e. automatic control of the sterilization process by computer
or microprocessor) system is preferred over manual control system. Effective sterilization
is considered when holding time of 121°C for 15 minutes is recorded.
Cleaning and drying of surgical instruments should be done before packaging.

Packaging
After cleaning and drying of surgical instruments, they should be properly packaged for
loading into the sterilizer. Since, linen or textile is reusable, cheap it is commonly used
for wrapping surgical trays. It should be used in a minimum of two thicknesses. It should
be of a good quality muslin having a specification of 140 thread count.14
The linen wrappers are being substituted by paper products for packaging recently.
The reason being linen is poorly water repellant and also not a good bacteriological
barrier. The various types of paper used are follows:
i. Medical grade Kraft paper.
ii. Laminated paper.
iii. Crepe paper.
Prophylaxis for postoperative endophthalmitis 81

iv. Non- glazed butter paper.


Plastic film cannot be used as packaging material for steam sterilization as water cannot
penetrate plastic films in liquid or vapor form. Sterilizer rack should be used inside the
sterilizer so that all the surfaces of instrument trays are directly exposed to steam.
Instrument trays should be placed upright.
The instrument trays should be perforated (Holes) at the bottom.
After sterilization, trays should be left in the sterilizer till all the steam has escaped
and it has undergone initial cooling.

Monitoring of Sterilization Process


For prevacuum sterilizer.
Bowie Dick test: It is a diagnostic test of a sterilizers’ ability to removes air from the
chamber. Indirectly, it checks vacuum pump in a high vacuum sterilizer. It should be
done daily. An adhesive tape which is printed with chemical substance (indicator) is fixed
in a shape of a cross to a piece of suitable paper. This sheet is placed at center of a folds
of multiple towels above and below. This is loaded inside the sterilizer. The color change
should occur within 3½ minutes at 135°C.14
Chemical indicators: It should be used in each pack and in every cycle. These
indicators show exposure to sterilization processes by means of physical or chemical
change. These should be used inside and outside individual backs. Internal chemical
indicator indicates whether penetration of sterilant was adequate.
Biological indicators: Bacillus stearothermophilus, a highly resistant spore forming
microorganism used in spore form on paper strips for checking effectiveness of
sterilization in steam sterilizer. Biological indicators are indicated during installation and
after major repairs.

Dry Heat Sterilization

Working Principle
Heat is distributed equally throughout the chamber which is done by forced circulation of
air with the help of fan, by which sterilization is achieved.

Efficacy
Since, hot air is a poor conductor and does not penetrate well, dry heat sterilization is less
effective than steam sterilization.

Indications
Sterilization of sharp instruments which can be damaged by moist heat in steam
sterilization method.
Evidence-based approach in cataract surgery 82

Sterilization Procedure
The sterilization of contents occurs at a temperature of 160°C for 60 minutes or 180°C
for 30 minutes. There is destructive oxidation of constituents inside the cell (Bacterial) by
sterilization. Automatically controlled dry heat sterilizers are preferred over manually
controlled.

Monitoring
i. Chemical indicator should be used with each sterilization cycle.
ii. Biological indicator (Bacillus subtilis) should be used once a weak.

Chemical Sterilization: Ethylene Oxide (Eto) Sterilization

Working Principle
Ethylene oxide alters the DNA of microorganisms by a process of alkylation thus killing
them.

Efficacy
Ethylene oxide sterilization kills all organisms including tubercle bacilli and spores.

Indications
The Eto should be used only for those instruments that are liable for damage due to heat,
e.g. phacoemulsification tubings, cryoprobes, laser probes, light pipe and vitrectomy
culters.

Sterilization Procedure
Recommended parameters are as follows:
i. Temperature 37 to 60 °C depending on items.
ii. Exposure time 105 to 300 minutes.
iii. Humidity 45 to 75%.
Specially designed plastic bags made of polyethylene are used for packing instruments.
Plastic pouches designed for Eto should have paper on one side. The plastic films should
be in the range of 1 to 3 mils (fractions of a inch) in thickness. Excess of air should be
removed before sealing the plastic bag to avoid bursting of seams during sterilization.

Advantages
i. There is minimal damage to sterilized instruments.
Prophylaxis for postoperative endophthalmitis 83

ii. Can pack the instruments before hand and once sterilized can be stored for relatively
long time.

Disadvantages
i. Eto is toxic gas hence proper training of operating persons is necessary.
ii. Eto sterilized material that absorb Eto have to be properly aerated before use.

Chemical Sterilization by Liquid Chemical Agents


Liquid chemical agents are classified according to their germicidal activity as:
1. Low level disinfectant.
2. Intermediate level disinfectant.
3. High level disinfectant.
Only high level disinfectant destroys all bacteria, fungi, lipophylic virus and spores.
Hence, surgical instruments require high level disinfectant for sterilization. A 2%
glutaraldehyde is the commonest high level disinfectant used.

Properties of 2% Glutaraldehyde
i. It is noncorrosive, hence can be used for plastic or rubber material.
ii. It has low surface tension which allows easy access to inner surfaces of instruments.
iii. Stable in alkaline solution, hence it is available commercially with separate alkaline
buffer which has to be added before its use.

Indications
For sterilization of sharp instruments which are liable to get damaged by heat. When used
for an extended time it destroys all spores and is considered sterilant.

Procedure
Instruments to be sterilized are left in the solution for few hours. Thorough rinsing of
instruments with normal saline is necessary before use because residual glutaraldehyde is
very irritating to ocular tissues.

Limitations
Sterilization by glutaraldehyde is not recommended due to:
1. Lack of adequate monitoring system.
2. Effect of several variables on the efficacy of the process.

Cleaning of Surgical Instruments


Evidence-based approach in cataract surgery 84

i. All reusable operating instruments should be cleaned thoroughly prior to sterilization.


ii. It is not possible to sterilize an instrument without cleaning it first. Because the
presence of soil on instruments inhibits the contact of a sterilant with microbial cell
thus reducing its effectiveness.
iii. Cleaning should be started as soon as possible after use.
iv. The instruments, if composed of more than one part should be disassembled before
cleaning.

Method
i. Tip of the instruments are cleaned using distilled water.
ii. Any blood or debris are removed from its surface using an instrument wipe or sponge.
iii. Then washing of instruments done in detergent solution. Alkaline detergents easily
removes protein soils and protein-enzyme dissolving solution should be used for
devices having lumens, e.g. phacoemulsification tips.
iv. Cleaning method can be either manual or mechanical.
a. Manual method: It includes rinsing the instrument with plenty of tap water to
remove detergents (deionized and distilled water preferred over tap water) followed
by drying.
b. Mechanical method: It is done in ultrasonic cleaners. In ultrasonic cleaners, sound
waves are passed at high frequency (100,000 Hz.) through liquids. Submicroscopic
bubbles are generated by these waves. When these bubbles collapse they create a
negative pressure on the particles in the suspension. By this action bacteria are
disintegrated and there is coagulation of protein matter.12,14,15
Phaco and irrigation/aspiration handpieces which contain lumen should not be
cleaned in ultrasonic cleaners. Irrigating their lumen with high pressure water
jet is recommended before sterilization.
v. Drying of instruments is necessary before packing in trays.

PROCEDURES FOR POSTCATARACT SURGERY ENDOPHTHALMITIS


PROPHYLAXIS
The surgical team and the patient are the prime sources of contamination during an
operation, as evidenced by the good matches between bacteria of infected wounds and
those of the team or the patient and by the poor matches between bacteria of infected
wounds and airborne bacteria.16
Postoperative endophthalmitis is one of the most serious conditions that can occur
after routine and otherwise uncomplicated cataract surgery. Fortunately its occurrence is
rare (5 to 10 cases/1000). This can be one of the factors that hinders investigation of
prophylactic measures.17 There are large number of variables in operating techniques
among various surgeons. Hence, carefully controlled prospective and masked studies of
systems for infection prevention have not been reported.18

SOURCES OF INFECTIONS
Prophylaxis for postoperative endophthalmitis 85

Origins for infection includes the followings:


1. Patients own ocular flora, lids and adnexa.
2. Surgical instruments, IOLs, irrigating solutions, viscoelastics.
3. Skin and respiratory flora of surgeon and assistant.
4. Operating room environment.

Causative Organisms
Ninety percent of causative organisms are gram-positive aerobic bacteria, 7% of
organisms are gram-negative bacteria and fungi constitutes only 3% of all causative
organisms.19 Staphylococcus epidermidis and Staphylo-coccus aureus are the two most
common organisms causing postoperative endophthalmitis.20,21 These organisms are the
most common organisms which can be recovered from patients eyelids.22,23
Patients own ocular flora was found to be the main source of postoperative infection.20
Various prophylactic interventions recommended for this are preoperative topical
antibiotic, preoperative use of povidone iodine, etc.

Irrigating solutions and viscoelastics are recommended to inspect before the use for intact
packing and for obvious contamination of vial in the form of particulate matter.
It is recommended to scrub hands and arms above elbow for 7–8 minutes with soap
alone. Recommended methods of hand hygiene include handwashing (washing hands
with plain soap), hygienic handwash (washing hands with medicated soap) and hygienic
hand-rub (use of antiseptic rubs).24 Of these hygienic hand-rub is the best technique.
Chlorhexidine can be used for hand disinfection system. It is found to reduce rate of
nosocomical infections more effectively than alcohol and soap.25 The 7.5% povidone
iodine scrub is another hand disinfecting agent. Scrubbing twice with 7.5% povidone
iodine scrub, for 2 minutes each is adequate (Fig. 9.4, Plate 3).
Evidence-based approach in cataract surgery 86

Fig. 9.4: 7.5% povidone iodine scrub


Prophylaxis for postoperative endophthalmitis 87

Fig. 9.5: 5% povidone iodine

PROCEDURES

A number of sources of postoperative infection may be targeted for prophylaxis.


Commonly used prophylactic interventions can be applied preoperatively, intraoperative
and postoperatively. There are a number of prophylactic techniques to decrease the risk
of postoperative endophthalmitis. But, justification of their use from the past literature is
unclear.

Preoperative Topical Antibiotics


The role of topical preoperative antibiotics in reducing postoperative infection is
documented.26 Topical antibiotics should be instilled only one day before surgery and
should never be instilled more than 6 to 8 times on that day. Instillation of topical
antibiotics for more than one day preoperatively leads to alteration of patients own
bacterial flora and replaced by more virulent organisms.27 Hence it should be
discouraged.

Preoperative Use of Povidone lodine


Topical 5% povidone iodine applied once preoperatively has been shown to reduce the
incidence of endophthalmitis by a factor of three (Fig. 9.5, Plate 3). There is no need for
irrigating it out of one eye before surgery. Povidone iodine is bactericidal in 30 seconds.28
Evidence-based approach in cataract surgery 88

There are no adverse reactions due to topical povidone iodine noted. Current literature
strongly recommends the use of preoperative povidone iodine antiseptic for postoperative
endophthalmitis prophylaxis in cataract surgery.29

Preoperative Preparation of the Eye


The regimens for preoperative antisepsis used to prepare the ocular surface are as
follows:
1. Cleaning the lids, lid margins and adjacent skin with 5% povidone iodine.
2. Eyelash trimming.
3. Saline irrigation of conjunctiva.
i. Five percent povidone iodine used for cleaning of lids, lid margins and adjacent
skin, is an effective method of eliminating microbes.30
ii. There is no evidence supporting lash trimming as a means of decreasing the ocular
surface flora.31 Schmitz S et al showed that lash trimming is not associated with
any reduction in the incidence of postoperative infection.32 Current trend is not
trimming the lashes but isolating them with sterile adhesive drapes. This prevents
to reduce passage of microorganisms into the eye.30
Recommended procedure: Lids, lid margins and surrounding skin after cleaned
with 5% povidone iodine twice and allowed to dry. The adhesive drape is
applied after opening the lids widely. The drape is cut between the lids and the
eye speculum is applied such that the drape is under the arms of speculum thus
isolating the eyelashes.27
iii. Saline irrigation of conjunctival sac, preoperatively is commonly practiced. This
intervention has not been shown to reduce ocular surface flora aqueous
contamination. Schmitz S et al found no reduction in the incidence of
endophthalmitis with this intervention.32

Antibiotics in Irrigating Solution


Another intervention for postcataract surgery endophthalmitis prophylaxis is
supplementing irrigating solutions with antibiotics. But, studies evaluating aqueous
contamination rates show mixed results. Beigi B et al found 20% positive anterior
chamber aspirate cultures in the group that received no antibiotics in irrigating solution as
compared to 2.7% cultures from group that received antibiotics in irrigating solution
(vancomycin 20 mg/L and gentamicin 8 mg/L).33 On the contrary, few literatures suggest
no beneficial effect of antibiotics in irrigating solution as postoperative endophthalmitis
antisepsis. The reasons for this being as follows.
i. Exposure to antibiotics for a short duration as in intraocular surgery has little effect on
organisms responsible for endophthalmitis.34
ii. The half-life of gentamicin in the anterior chamber after completion of
phacoemulsification is 51 minutes. It is cleared from the anterior chamber very
quickly. Hence, the bactericidal levels required for reliable antibiotic prophyaxis is
never maintained.35
Prophylaxis for postoperative endophthalmitis 89

Heparin in Irrigating Solution and Heparin Surface Modified IOL


This intervention for postcataract surgery endophthalmitis prophylaxis also has mixed
result. Bacterial strains, i.e. S. epidermidis, S. aureus and P. areuginosa were found
attached in significantly lower numbers to heparin surface modified IOLs than to PMMA
IOLs alone. Heparin in irrigating also inhibits attachment of S. epidermidis to regular
PMMA IOLs. This is due to heparin reduces adherence of those bacteria by placing a
highly hydrated layer between the bacteria and surface of IOL.36
But, there is no significant difference found in the rate of culture positive anterior
chamber aspirates between the heparinized group and the control group.37

Subconjunctival Antibiotics
Subconjunctival antibiotics after cataract surgery is found to reduce postoperative
infection but this reduction was found to be statistical insignificant. Literature is not
conclusive regarding the choice of antibiotic though gentamicin is found to be most
effective drug.38

REFERENCES

1. Laufman H: Design, devices, and discipline in operating room infcction control. Med Instrum.
1978 May–Jun;12(3):158–160.
2. Ruehl C.Insight: The value of the ORN. Ophthalmic Registered Nurse, 1997, Jun 22(2):37–38.
3. Au YK, Bynum PS, Poole T: Increasing OR efficiency with a specialty services manual. AORN
J 1996 Jul; 64(l):96–98, 100–101, 104.
4. Saravolatz LD, Arking L: Ophthalmology operating room standards and infection control
concerns. Am J Infect Control. 1984 Oct, 12(5): 271–275.
5. Heudorf U, Hentschel W, Kutzke G, Pfetzing H, Voigt K: Hygienic procedures in operation
theatres—guidelines and reality. Data obtained on hygiene control measures by public health
service at Frankfurt am Main. Gesundheitswesen. 2003 May; 65(5):312–320.
6. Haeseker B: Microsurgery: A ‘small’ surgical revolution in the medical history of the 20th
century. Ned Tijdschr Geneeskd. 1999 Apr 17;143(16):858–864.
7. Tamai S: History of microsurgery—from the beginning until the end of the 1970s. Microsurgery.
1993; 14(1):6–13.
8. Ayliffe GA: Role of the environment of the operating suite in surgical wound infection. Rev
Infect Dis. 1991 Sep–Oct; 13 Suppl 10:S800– S804.
9. Schonholtz GJ: Maintenance of aseptic barriers in the conventional operating room: General
principles. J Bone Joint Surg Am 1976 Jun; 58(4):439–445.
10. Fitzgerald RH Jr: Microbiologic environment of the conventional operating room. Arch Surg
1979 Jul; 114(7):772–775.
11. Rutala WA, Guidelines for Infection control practice in APIC guideline for selection and use of
disinfectants. Am J Inf Control 1990, 18:99–117.
12. Sharma S, Bansal AK, et al: Asepsis in Ophthalmic operating room. Indian J Ophthalmol 1996,
44:173–177.
13. Ram J, Kaushik S, et al: Prevention of postoperative Infections in ophthalmic surgery. Indian J
Ophthalmol 2001, 49:59–69.
14. Mehtra G: The sterile supply department, Guidelines for planning and quality management. A
publication from the hospital infection society-India.
Evidence-based approach in cataract surgery 90

15. Austin GC: Efficient storage of sterilized surgical instruments. Am J Ophthalmol 1982, 93:518–
519.
16. Laufman H: Airflow effects in surgery. Arch Surg 1979 Jul, 114(7): 826–830.
17. Kattan HM, Flynn HW Jr, et al: Nosocomial endophthalmitis survey. Current incidence of
infection after intraocular surgery. Ophthalmology 1991, 98:227–238.
18. Masket S: Preventing, diagnosing and treating endophthalmitis. J Cataract Refract Surg 1998,
24:725–726.
19. Olson JC, Flynn HW Jr, et al: Results in the treatment of postoperative endophthalmitis.
Ophthalmology 1983, 87:313–319.
20. Speaker MG, Milch FA, et al: Role of external bacterial flora in the pathogenesis of acute
postoperative endophthalmitis. Ophthalmology 1991, 98:639–649; discussion 650.
21. Han DP, Wisniewski SR, et al: Spectrum and susceptibilities of microbiologic isolates in the
endophthalmitis vitrectomy study [published erratum appears in Am J Ophthalmol 1996,
122:920]. Am J Ophthalmol 1996, 122:1–17.
22. Groden LR, Murphy B, et al: Lid flora in blepheritis. Cornea 1991,10 50–53.
23. Mistlberger A, Ruckhofer J, et al: Anterior chamber contamination during cataract surgery with
intraocular lens implantation. J Cataract Refract Surg 97, 23:1064–1069.
24. Wendt C: Hand hygiene—comparison of international recommendations. J Hosp Infect 2001
Aug; 48 Suppl A: S23–S28.
25. Doebbeling BN, Stanley GL, et al: Comparative efficacy of alternative hand washing agents in
reducing nosocomial infections in intensive care units. N Engl J Med 1992 Jul 9, 327:88–93.
26. Donnenfeld ED, Schrier A, et al: Penetration of topically applied ciprofloxacin, norfloxacin and
ofloxacin into the aqueous humour. Ophthalmology 1994, 101:902–905.
27. Ram J, Kaushik S, et al: Prevention of postoperative infections in ophthalmic surgery. Indian J
Ophthalmol 2001, 49:59–69.
28. Speaker MG,Meinikoff JA: Prophylaxis of endophthalmitis with povidone-iodine.
Ophthalmology 1991;98:1769–1775.
29. Thomas A Ciulla, Michael B Starr, et al: Bacterial Endophthalmitis prophylaxis for cataract
surgery. Ophthalmology 2002, 109:13–26.
30. Ram J. Reducing cataract related complications. Indian J Ophthalmol 1999, 47:153–154.
31. Perry L D, Skaggs C: Preoperative topical antibiotics and lash trimming in cataract surgery.
Ophthalmic Surg 1977, 8:44–28.
32. Schmitz S, Dick H B, et al: Endophthalmitis in cataract surgery. Results of a German Survey.
Ophthalmology 1999, 106:1869–1877.
33. Beigi B, Westlake W, et al: The effect of intracameral, per-operative antibiotics on microbial
contamination of anterior chamber aspirates during phacoemulsification. Eye 1998, 12:390–394.
34. Gritz D C, Cevallos AV, et al: Antibiotics supplementation of intraocular irrigating solutions.
An in vitro model of antibacterial action. Ophthalmology 1996, 103:1204–1208; discussion
1208–1209.
35. Lehmann OJ, Thompson JP, et al: Half-life of intracameral gentamicin after
phacoemulsification. J Cataract Refract Surg 1997, 23:883–888.
36. Portoles M, Refojo MF, et al: Reduced bacterial adhesion to heparin surface modified
intraocular lenses. J Cataract Refract Surg 1993, 19: 755–759.
37. Manners TD, Turner DPJ, et al: heparinized intraocular infusion and bacterial contamination in
cataract surgery. Br J Ophthalmol 1997, 81:949–952.
38. Starr MM: Prophylactic antibiotics for Ophthalmic surgery. Surv Ophthalmol 1983, 27:353–
373.
Index

A
A-scan biometry 104
Acrylic IOL 40
Acute suprachoroidal hemorrhage 97
Antibiotics in irrigating solution 154

C
Capsular folds 123
Capsular tension ring 66
Capsulorhexis 79
Cataract surgery 1, 11, 76, 86
complications 86
in a patient with uveitis 76
newer anesthesia modalities 1
newer incision techniques 11
Classic air bubble technique 72
Clear corneal incision 18
Continuous curvilinear capsulorhexis (CCC) 63, 120
Conventional phacoemulsification 24

D
Defractive multifocal IOL 45
Descemet’s membrane detachment 79, 99
Disposable nylon hooks 69
Dodick photolysis 28
Dropped nucleus 94

E
Erbium: YAG laser system 26
Extracapsular cataract surgery 12

F
Foldable IOLs 12
Formaldehyde fumigation 138
Future intraocular lens 53

G
Glutaraldehyde 136

H
Hard cataract 60
Index 93

Heparin surface modified (HSM) IOL 81


Heparin surface modified PMMA IOLs 51
Hydrodelineation 63
Hydrodissection 63
Hydrophilic acrylic IOL 49
Hydrophobic foldable acrylic IOL 48

I
Incision
clear corneal 12
scleral tunnel 12
Injectional anesthesia 4
Intracameral anesthesia 7
Intracameral subcapsular injection 73
Intracapsular cataract surgery 12
Intraocular inflammation 76
Intraocular lens
position of 121
Intraocular lens design 121
Intraocular lens implantation in children
ideal IOL material 51
ideal IOL size 52
Intraocular lens materials 41, 118
polymethyl-methacrylate (PMMA) 41
silicone IOL 42
IOL implantation 80
IOL power calculation 103, 111
in patients with high myopia 112
in pediatric cataract 111
of silicone oil filled eyes 113

K
Kelman tip 61
Keratometric diopters 107
Keratometry 106
Koch’s basic incision 16
Kratz’s scleral pocket incision 17

L
Laboratory microscope 133
LASIK 107
Layout of an ideal operating room complex 135

M
Modern phacoemulsification system 25
Multifocal IOL 45

N
Nd:YAG laser posterior capsulotomy 124
Neodymium: YAG laser phacoemulsification 28
Index 94

NeoSonix phacoemulsification 31
New phacoemulsification technologies 23

O
Operating room 130
cleaning 136
disinfection 136
environment 134
location 130
prevention of infections 130
Orbital hemorrhage 86

P
Partial coherence interferometry (PCI) 104
advantages 105
limitations 105
PCO
assessment of 123
newer treatments 125
treatment of 124
Peribullar anesthesia 60
Phacoemulsification 12, 59, 60, 64, 65, 71
in difficult situations 59
in hard cataracts 60
in subluxated cataract 65
in white cataract 71
of nucleus 64
Phaconit 35
Photon laser phacolysis 30
Polymethyl methacrylate (PMMA) IOL 40
Posterior assisted levitation 96
Posterior capsular tear 89
Posterior capsule opacification 117
Posterior polar cataract 62
Postoperative endophthalmitis 149
Preoperative preparation of the eye 152
Preoperative topical antibiotics 152
Preoperative use of povidone iodine 152
Prophylaxis for postoperative endophthalmitis 129

R
Recent advances in intraocular lens 39
Refractive multifocal IOL 46

S
Sandwich theory 118
Scleral tunnel incision 13
chevron incision 15
curvilinear incision 13
frown incision 14
Index 95

incisional funnel 13
internal configuration 16
straight incision 14
Silicone IOL 40
Small incision
advantages 12
Small incision surgery 87
Sonic phacoemulsification 30
Sterilization processes 139
chemical sterilization:
ethylene oxide (Eto) sterilization 144
chemical sterilization by liquid chemical agents 146
dry heat sterilization 143
steam sterilization 140
Sub-Tenon’s (Parabulbar) anesthesia 8
Subconjunctival antibiotics 155
Subluxated cataract 65

T
Thermodynamic injectable IOL 53
Three-piece monofocal silicone IOL 45
Three-step corneal valve incision 17
Topical anesthesia 3
advantages 5
contraindications 5
Topical eyedrop anesthesia 6
Toric IOL 44
Trypan blue 73
Trypan blue staining 74

U
Ultrasound phacoemulsification 26

V
Vitrectomy 92

W
White cataract 71
WhiteStar technology 32

You might also like