throbber
INTRAVITREAL INJECTION OF
`THERAPEUTIC AGENTS
`
`GHOLAM A. PEYMAN, MD,*† ELEONORA M. LAD, MD, PHD,†
`DARIUS M. MOSHFEGHI, MD†
`
`Background:
`Intravitreal injection (IVI) with administration of various pharmacological
`agents is a mainstay of treatment in ophthalmology for endopthalmitis, viral retinitis,
`age-related macular degeneration, cystoid macular edema, diabetic retinopathy, uveitis,
`vascular occlusions, and retinal detachment. The indications and therapeutic agents are
`reviewed in this study.
`Methods: A search of the English, German, and Spanish language MEDLINE database
`was conducted. A total of 654 references spanning the period through early 2008 were
`individually evaluated.
`Results: The advantage of the IVI technique is the ability to maximize intraocular levels
`of medications and to avoid the toxicities associated with systemic treatment. Intravitreal
`injection has been used to deliver several types of pharmacological agents into the vitreous
`cavity: antiinfective and antiinflammatory medications, immunomodulators, anticancer
`agents, gas, antivascular endothelial growth factor, and several others. The goal of this
`review is to provide a detailed description of the properties of numerous therapeutic agents
`that can be delivered through IVI, potential complications of the technique, and recom-
`mendations to avoid side effects.
`Conclusion: The IVI technique is a valuable tool that can be tailored to the disease
`process of interest based on the pharmacological agent selected. This review provides the
`reader with a comprehensive summary of the IVI technique and its multitude of uses.
`RETINA 29:875–912, 2009
`
`Intravitreal injection (IVI) was initially performed
`
`for treatment of retinal detachment (RD) and vit-
`reous hemorrhage. In 1895, Deutschmann1 injected
`transplanted rabbit vitreous, and Ohm2 injected air
`in the vitreous cavity for the repair of RD. During
`subsequent decades, the use of IVI was limited to
`administration of saline3–7 and air.8 In the 1960s
`and 1970s, long-lasting gases were developed for
`the repair of complex RD.9 Intravitreal penicillin
`for treatment of endophthalmitis was attempted in
`the late 1940s,10 –12 but was subsequently aban-
`doned by the same authors in favor of systemic and
`subconjunctival injection.
`
`From the *Department of Ophthalmology and Vision Science,
`College of Medicine, University of Arizona, Tucson, Arizona;
`Attending Physician, Maricopa Health System, Phoenix, Arizona;
`Department of Ophthalmology, Tulane University, New Orleans,
`Louisiana; and †Department of Ophthalmology, Stanford Univer-
`sity, Stanford, California.
`The authors have no proprietary interest in any of the materials
`discussed in this article.
`Reprint requests: Gholam A. Peyman, MD, Department of Oph-
`thalmology and Vision Science, College of Medicine, University of
`Arizona, 10650 Tropicana Circle, Sun City, AZ 85351; e-mail:
`gpeyman1@ yahoo.com
`
`The modern era of IVI began in the early 1970s
`with the investigation of the blood ocular barri-
`ers.13,14 The results of these investigations stimu-
`lated the use of IVI of antibiotics for treatment of
`endophthalmitis and steroids for treatment of in-
`traocular inflammation to bypass anatomical barri-
`ers in the eye.15–17 This concept heralded the advent
`of IVI of antiinflammatory and antineoplastic com-
`pounds in the 1970s to 1980s, antivirals in the
`1980s to 1990s, triamcinolone acetonide (TA), and
`vascular endothelial growth factor (VEGF) inhibi-
`tors in the 2000s, setting the stage for a paradigm
`shift in how therapeutic agents are delivered to the
`eye.18 Medicare claims data indicated that 325,000
`IVIs (CPT 67028) codes had been filed in 2006
`alone,19 and the number is likely in excess of a
`million today. Intravitreal injection has been used to
`deliver many types of medications into the vitreous
`cavity: antiinfective (antibiotic, antifungal, and an-
`tiviral), antiinflammatory (nonsteroidal antiinflam-
`matory, steroids, and immunomodulators), antican-
`cer agents, gas, anti-VEGF, etc. With numerous
`novel therapies currently being investigated in clin-
`
`875
`
`Novartis Exhibit 2009.001
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`876
`
`RETINA, THE JOURNAL OF RETINAL AND VITREOUS DISEASES ● 2009 ● VOLUME 29 ● NUMBER 7
`
`ical trials, it is likely that the number of drugs under
`development for IVI will continue to increase.
`
`Intravitreal Injection Technique
`
`The use of preoperative topical antibiotics is not
`widespread despite evidence that this method can re-
`duce the conjunctival bacterial flora.20 –26 A recent
`report of a large series of IVI without preoperative use
`of antibiotics showed a low rate of endophthalmitis,
`indicating that the preoperative antibiotic may not be
`needed in each case.27 Another study involving more
`than 16,000 patients receiving office-based IVIs of anti-
`VEGF demonstrated a low rate of endophthalmitis
`(0.02%) despite variable preoperative prophylaxis.28
`Intravitreal injection is performed under topical an-
`esthesia, usually in an office-based setting. Clinicians
`should use aseptic techniques and use povidone–iod-
`ine to minimize the conjunctival bacterial flora.20 –24
`The type of anesthesia used with IVI varies between
`retinal specialists. Although 98% of retinal physicians
`surveyed in 2006 reported the use of anesthesia before
`IVI, the mode of anesthesia administration was divided
`between topical anesthesia (66.6%) and subconjunctival
`(33.3%).29 Anesthesia is usually accomplished with ei-
`ther cotton-tip applicators soaked in lidocaine or subcon-
`junctival injection of 2% lidocaine or 4% lidocaine jelly.
`Other options include topical anesthetics (lidocaine, pro-
`paracaine, or tetracaine) or the application of tetracaine
`jelly. Typically, a povidone–iodine (5%) solution is ap-
`plied to the eye either in solution form or povidone
`followed by thorough cleaning of the eyelashes, with
`subsequent application of a lid speculum. The anes-
`thetics used (proparacaine, lidocaine, or bupivacaine)
`have been shown to be nontoxic to intraocular struc-
`tures in rabbits. Intraocular application of lidocaine up
`to 2%, bupivacaine up to 0.75%, and a combination of
`the two anesthetics induced reversible electroretino-
`gram changes in the rabbit retina but did not result in
`any adverse clinical reactions or histologic retinal
`abnormalities.30
`After anesthesia, the pharmacological agent for IVI
`(either prepared by the manufacturer, such as ranibi-
`zumab, or prepared by the compounding pharmacist,
`such as bevacizumab) is drawn into a 1-mL tuberculin
`syringe from a sterile bottle, usually with a filter needle
`in place for the IVI itself. Thirty- to 32-gauge needles are
`most commonly used, whereas larger bore needles (27
`or 28.5 gauge) are less frequently used.31
`The injection site is usually the infero-temporal
`quadrant to avoid drug deposition in front of the visual
`axis. The pars plana is 3 mm to 4 mm posterior to the
`limbus. The needle should be aimed at the midvitreous
`with the bevel upward, and the injection should be
`
`done slowly to avoid jet formation or cavitary flow.
`The needle should not be introduced all the way to the
`hub. Using a single smooth continuous maneuver, the
`pharmacological agent is injected into the eye. The vol-
`ume of the administered compound is relatively con-
`sistent between studies, with the most commonly used
`volume being 50 ␮L to 100 ␮L. However, the studies
`using standard volumes do not account for the reduc-
`tion in size of the human vitreous that occurs with
`age.32 Generally, most clinicians believe that 100 ␮L
`is the largest safe volume, and larger volumes are
`reserved mainly for administration of gas for pneu-
`matic retinopexy (PR).31,33 The needle is removed si-
`multaneously with the application of a cotton-tipped
`applicator over the sclerotomy site to minimize reflux of
`material (particularly if the injected volume exceeded
`0.05 mL).
`The use of postoperative antibiotics is routine, re-
`ported in approximately 59% of the studies.31 A
`postinjection course of topical antibiotics typically
`lasts for 3 days to 7 days.
`
`Delivery of Antiinfective Agents
`
`Antibacterial Drugs
`
`Administration of antibiotics directly into the vitre-
`ous body through IVI is the standard of care for the
`treatment of bacterial endophthalmitis. Table 1 shows
`the recommended dosage of antibiotic for IVI, the
`half-life of each antibiotic in the vitreous body, and
`the maximum nontoxic dosage for IVI. Selection of an
`appropriate antibiotic depends on prevailing organ-
`isms in the geographic area, mechanism of endoph-
`thalmitis, strain of bacteria, sensitivity and resistance,
`as well as patient factors (e.g., allergies and sensitiv-
`ities). In the setting of acute postsurgical endoph-
`thalmitis, vancomycin in combination with either a
`third-generation cephalosporin or amikacin is typi-
`cally injected as initial empirical therapy.
`Aminoglycosides. Aminoglycosides have potent ac-
`tivity against gram-negative bacteria but are limited
`by their potential toxicity. The aminoglycoside anti-
`biotic family—streptomycin, gentamicin, kanamycin,
`tobramycin, amikacin, and netilmicin— has a chemi-
`cal composition of an organic base with amino
`sugars and are synthesized from numerous species
`of fungal organisms. Their wide spectrum of anti-
`biotic activity against both gram-positive and gram-
`negative bacteria is due to the ability to interfere
`with synthesis of ribosomal proteins. Amikacin is
`often the aminoglycoside of choice for human en-
`dophthalmitis,18 whereas gentamicin and clindamy-
`cin were shown to be effective in the prevention of
`
`Novartis Exhibit 2009.002
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`INTRAVITREAL INJECTION OF THERAPEUTIC AGENTS ● PEYMAN ET AL
`
`877
`
`Table 1. Recommended Dose and Pharmacologic
`Profile of Intravitreal Antibiotics
`
`Antibiotic
`
`Aminoglycosides
`Amikacin
`Gentamicin
`Kanamycin
`Netilmicin
`Streptomycin
`Tobramycin
`Penicillins
`Ampicillin
`Azlocillin
`Carbenicillin
`Cloxacillin
`Dicloxacillin
`Methicillin
`Mezlocillin
`Oxacillin
`Penicillin G
`Piperacillin
`Ticarcillin
`Cephalosporins
`Cefamandole
`Cefazolin
`Cefotaxime
`Cefotetan
`Cefoxitin
`Ceftazidime
`Ceftriaxone
`Cephalexin
`Cephaloridine
`Cephalothin
`Moxolactam
`Fluoroquinolones
`Ciprofloxacin
`Garenoxacin
`Gatifloxacin
`Levofloxacin
`Moxifloxacin
`Norfloxacin
`Ofloxacin
`Pefloxacin
`Travofloxacin
`Macrolides
`Erythromycin
`Clarithromycin
`Lincosamides
`Lincomycin
`Clindamycin
`Carbapenems
`Imipenem
`Meropenem
`Glycopeptides
`Vancomycin
`Teicoplanin
`Miscellaneous
`Aztreonam
`Chloramphenicol
`Cotrimoxazole
`Doxycycline
`Chloramphenicol
`
`Intravitreal
`Dose (mg)
`
`Vitreous
`Half-Life (h)
`
`0.4
`0.2
`
`0.25
`
`0.2
`
`5
`
`24
`12–35
`
`24
`
`16
`
`6
`
`0.5–2
`
`10–20
`
`3–5
`
`3
`
`0.5
`7
`
`16
`12
`3
`
`2.4
`20
`
`5
`
`1.72
`
`5.65
`3
`
`30
`2
`
`7–8
`
`30
`
`7.5
`10
`
`2
`
`0.5
`0.2–0.3
`1.5
`3
`
`0.08
`0.5–2.0
`0.4
`1
`
`2
`2
`
`2.5
`2
`1.25–2.0
`
`0.1
`0.1–2
`0.4
`0.625
`0.05–0.16
`
`0.05
`0.2
`0.025
`
`0.5
`1
`
`1
`0.5–1
`
`0.5
`
`1
`0.75
`
`0.1
`2
`1.6
`0.125
`1
`
`Antibiotic
`
`Antifungals
`Polyene antimycotics
`Amphotericin B
`Natamycin
`Nystatin
`Azoles
`Fluconazole
`Itraconazole
`Ketoconazole
`Miconazole
`Oxiconazole
`Terconazole
`Voriconazole
`Echinocandins
`Caspofungin
`Miscellaneous
`Faeriefungin
`Flucytosine
`Cilofungin
`
`Table 1.
`
`Intravitreal
`Dose (mg)
`
`Vitreous
`Half-Life (h)
`
`6.9–15.1
`
`3.1
`
`2
`
`2.5
`
`0.005–0.01
`3 X 0.025
`200 U
`
`0.1
`0.01
`0.54
`0.025–0.05
`0.1
`10
`0.05–0.1
`
`0.1
`
`0.1
`0.1
`0.32
`
`acute posttraumatic bacterial endophthalmitis.34,35
`Side effects of parenteral aminoglycosides include
`ototoxicity and neprotoxicity.18
`Toxicity of intravitreal aminoglycosides, which can
`occur with doses as low as 10 ␮g to 200 ␮g of
`gentamicin, includes the following: 1) formation of
`lysosomal cellular inclusions, resulting in ocular cell
`toxicity and loss of function; 2) cataracts following a
`dose greater than 200 ␮g of gentamicin; and 3) retinal
`infarction due to gentamicin and tobramycin.36,37 For
`this reason, some advocate the use of low-dose gen-
`tamicin (4 – 8 ␮g) in the infusion fluid.34,35 Current
`recommendations emphasize administration of IVI
`into the anterior vitreous with the needle bevel facing
`the lens, slow injection of the medication, and avoid-
`ance of repeated IVI in short intervals (less than 1
`week, except for the negative sensitivity results for the
`antibiotic used), especially in combination at
`low
`doses with vancomycin or clindamycin to limit the
`possibility of toxicity.18,34
`Cephalosporin. The third-generation cephalosporins,
`which include ceftriaxone, ceftazidime, and moxolac-
`tam, are a mainstay in the initial management of IVI.
`They have increased activity against gram-negative
`organisms but are less active against gram-positive
`organisms.38 The cephalosporins were first isolated
`from the fungus Cephalosporium acremonium. These
`semisynthetic antibiotics have a similar chemical
`structure and mechanism of action to the penicillin
`family. The first generation of cephalosporins, which
`includes cefazolin, cephalexin, and cephalothin, has a
`wide spectrum of activity against gram-positive bac-
`
`Novartis Exhibit 2009.003
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`878
`
`RETINA, THE JOURNAL OF RETINAL AND VITREOUS DISEASES ● 2009 ● VOLUME 29 ● NUMBER 7
`
`teria and some activity against gram-negative bacteria.
`The second generation, represented by cefotetan, ce-
`foxitin, and cefamandole, has more activity against
`gram-negative organisms but are less active against
`gram-positive bacteria, especially Staphylococcus. Other
`members of this family—cephaloridine, cephalothin,
`ceftazidime, and cefotaxime—have demonstrated effi-
`cacy and safety in experimental models of progression of
`endophthalmitis without toxicity to intraocular structures
`in rabbit eyes.39 – 44 Cefazolin is currently not recom-
`mended for treatment of endophthalmitis due to increase
`in resistant organisms.39,42 Intravitreal injection of moxa-
`lactam inhibited experimental Staphylococcus aureus
`endophthalmitis but resulted in retinal abnormalities in
`rabbits at doses of 2.5 mg or higher.45,46
`
`Glycopeptides
`Vancomycin.—Vancomycin is the preeminent choice
`for initial treatment of acute postoperative endoph-
`thalmitis due to gram-positive organisms. Vancomy-
`cin is an antibiotic produced by Streptomyces asianis
`and has a unique chemical structure that lends it high
`activity against gram-positive cocci.47 Vancomycin
`inhibits bacterial cell wall synthesis. Vancomycin,
`first developed in 1958, has become an extremely
`effective drug in treatment of severe gram-positive
`ocular infections (keratitis, endophthalmitis, orbital
`cellulitis) in the setting of antibiotic-resistant bacteria
`or antibiotic allergy. However, in recent years, a con-
`troversy arose over the use of this agent in ophthalmol-
`ogy, especially as a prophylaxis in elective cataract sur-
`gery.18 The identification of vancomycin resistance in
`coagulase-negative Staphylococci in 1987, followed by
`cases of resistant Enterococci in 1988, has led to calls for
`the judicious use of this agent in ophthalmology.48,49
`Intravitreal vancomycin is the drug of choice for
`endophthalmitis caused by gram-positive organisms.18
`Intravitreal injection of vancomycin was nontoxic to
`ocular structures in the clinically recommended dose
`of 1 mg/0.1 mL; added to infusion fluid for intraocular
`surgery, the nontoxic dose is 8 mg/mL to 32 mg/mL.50
`A dose of 1 mg/0.1 mL halted progression of methi-
`cillin-resistant S. aureus endophthalmitis.47 We urge
`caution in patients with silicone oil, because nontoxic
`concentrations of this drug may become toxic after
`IVI in postvitrectomy, silicone-filled rabbit eyes.51
`The threshold for ocular toxicity in rabbits decreased
`to one quarter of the nontoxic dosage in an unoperated
`eye compared with silicone-filled eyes.51 In the En-
`dophthalmitis Vitrectomy Study,52 all gram-positive
`bacteria including methicillin-resistant S. aureus were
`susceptible to vancomycin.53
`In cases of endophthalmitis due to gram-positive
`organisms, resistance to vancomycin is still rare, at
`
`only 2.1% of a reported 8,500 strains of bacteria.54
`However, ophthalmologists should use caution when
`using vancomycin, because the incidence of vanco-
`mycin-resistant S. aureus and Enterococci isolates
`continues to rise.49 New antibiotics such as quinu-
`pristin-dalfopristin, linezolid, and daptomycin may
`show promise in the treatment of vancomycin-re-
`sistant infections.55– 65 Vancomycin showed syner-
`gism with aminoglycoside antibiotics against vari-
`ous organisms, especially Enterococcus, and with
`␤-lactams against gram-positive bacteria and some
`gram-negative bacteria.54,55
`Teicoplanin.—Teicoplanin is a relatively new glyco-
`peptide antibiotic that is isolated from Actinoplanes
`teichomyceticus. It is active against most gram-posi-
`tive bacteria, especially Staphylococci. Teicoplanin in
`combination with gentamicin is effective against En-
`terococci. Intravitreal injection of teicoplanin is non-
`toxic in rabbit retinas at doses up to 750 g/0.1 mL.66,67
`Penicillins. The penicillins are rarely used for treat-
`ment of endophthalmitis because of widespread resis-
`tance. Penicillins are a group of antibiotics derived from
`6-aminopenicillic acid. They work by inhibiting bacterial
`cell wall synthesis. Intravitreal injection administration is
`currently reserved for methicillin, cloxacillin, dicloxacil-
`lin, and ampicillin. Methicillin, oxacillin, and ampicillin
`are penicillinase-resistant, semisynthetic penicillins that
`can be delivered by IVI and do not cause toxicity at
`recommended dosages.68 –70 The number of Staphylo-
`cocci species resistant to methicillin and other peni-
`cillinase-resistant penicillins has increased.18 The tox-
`icity, clearance, and efficacy of IVI cloxacillin and
`dicloxacillin for endophthalmitis management have
`not been studied.18
`Extended-spectrum penicillins are alphacarboxypeni-
`cillins (e.g., carbenicillin and ticarcillin) and acylamin-
`openicillins (e.g., azlocillin, mezlocillin, and piperacil-
`lin). Although these antibiotics are effective against
`gram-negative aerobic organisms that cause endoph-
`thalmitis, such as Enterobacteriaceae and Pseudomo-
`nas, which are currently resistant to many penicillins,
`their use is not widespread. Similarly, experimental
`evidence suggests that
`intravitreal piperacillin/ta-
`zobactam, carbenicillin,
`ticarcillin, and piperacillin
`have demonstrated efficacy and lack of toxicity in
`experimental models.71–73
`are not
`Fluoroquinolones. The fluoroquinolones
`widely used via IVI for treatment of endophthalmitis.
`The fluoroquinolones are derived from nalidixic acid
`and have a potent activity against gram-negative bac-
`teria. Ciprofloxacin is considered effective for prophy-
`laxis and treatment of intraocular infections74 and is
`not toxic to the rabbit retina.75
`
`Novartis Exhibit 2009.004
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`INTRAVITREAL INJECTION OF THERAPEUTIC AGENTS ● PEYMAN ET AL
`
`879
`
`Norfloxacin, ofloxacin, and pefloxacin are only ad-
`ministered orally for prevention or management of
`endophthalmitis76 but have not been used for IVI.
`Despite their initial efficacy, resistance to first- and
`second-generation quinolones in bacterial keratitis and
`other ophthalmologic infections is increasing.76,77
`This prompted the development of third- (levofloxa-
`cin) and fourth-generation quinolones (gatifloxacin,
`garenonoxacin, and moxifloxacin) during the past de-
`cade. New quinolones achieved an increased efficacy
`against gram-positive organisms while maintaining
`activity against gram-negative bacteria.78 – 80
`The third- and fourth-generation fluoroquinolones
`represent a good first-line antibiotic treatment for
`many ocular infections. Intravitreal injection of these
`antibiotics did not
`result
`in electroretinographic
`changes or retinal toxicity in rabbits when adminis-
`tered in doses up to 625 ␮g for levofloxacin,78 400 ␮g
`for gatifloxacin,78 4,000 ␮g for garenonoxacin,79 and
`160 ␮g for moxifloxacin.80
`Macrolides. Due to increasing resistance, erythromy-
`cin has rarely been used for IVI therapy of endoph-
`thalmitis. Erythromycin, which was first synthesized
`from Streptomyces erytheus in 1952, has activity
`against gram-positive cocci and Neisseria species. Its
`mechanism of action is inhibition of protein synthesis
`by binding the 23S rRNA molecule of the 50S subunit
`of bacterial ribosome, inhibiting peptide growth.81 De-
`spite effectiveness against experimental S. aureus en-
`dophthalmitis and a lack of ocular toxicity,82 erythro-
`mycin is not used due to widespread resistance.
`Intravitreal clarithromycin was also nontoxic to rabbit
`eyes in a dose of up to 1.0 mg.83
`Lincosamides. The lincosamides are rarely used for
`IVI therapy of endophthalmitis.
`Lincomycin.—Lincomycin is derived from the acti-
`nomycete Streptomyces lincolnensis and has a mech-
`anism of action similar to that of erythromycin.81,84 It
`is effective against Group A Streptococci, Pneumo-
`cocci, penicillinase-producing S. aureus, Corynebac-
`teria, Clostridia, and Bacteroides species.
`Clindamycin.—Clindamycin, a semisynthetic deriv-
`ative of lincomycin, has the same antibiotic activity
`and mechanism of action as lincomycin but with
`higher potency.18 Intravitreal injection of clindamycin
`has demonstrated efficacy for prophylaxis against en-
`dophthalmitis and is nontoxic to the retina.34,68 Intra-
`vitreal injection or intracameral injection of clindamy-
`cin (45 ␮g) in conjunction with IVI or intracameral
`gentamicin (40 ␮g) was superior to balanced salt
`solution IVI/intracameral injections in the prevention
`of endophthalmitis after penetrating eye injury (2.3%
`vs. 0.3%, P ⫽ 0.04).34 Additionally, it was found that
`
`IVI antibiotic administration was superior to intracam-
`eral administration.34 These data strongly suggested
`that intraocular and especially intravitreal gentamicin
`and clindamycin were effective in preventing acute
`posttraumatic bacterial endophthalmitis. Intravitreal
`injection of 1.0 mg clindamycin in 0.1 mL and 1.0 mg
`of dexamethasone in 0.1 mL was also well tolerated in
`patients with toxoplasmic retinochoroiditis.85 This
`treatment resulted in a favorable response after 2
`weeks; specifically, there was improvement in visual
`acuity and preservation of the disk and macula, sug-
`gesting that this regimen may be an additional tool in
`treatment of toxoplasmic retinochoroiditis.85 Further
`research into IVI clindamycin is warranted because
`the reported adverse drug reaction rate to clindamycin
`for ocular toxoplasmosis is 22.5%.86
`Carbapenems. Imipenem/cilastatin is a combination
`of two antibiotics, cilastatin and the ␤-lactam theina-
`mycin, and works by inhibiting cell wall synthesis. It
`has a wide spectrum of activity against gram-positive
`and gram-negative aerobic and anaerobic bacteria.
`Systemic imipenem has good ocular penetration, with
`aqueous concentration of 2.99 ␮g/mL and vitreous
`concentration of 2.53 ␮g/mL 2 hours after adminis-
`tration of 1 g ofimipenem in 25 patients undergoing
`routine cataract extraction.87 Although rarely used in
`IVI therapy, imipenem has demonstrated efficacy in
`one case of Pseudomonas endophthalmitis and was
`nontoxic to ocular structures.88 Experimental IVI of
`imipenem for
`treatment of experimental Bacillus
`cereus and Pseudomonas endophthalmitis in pigs
`demonstrated safety and efficacy.89,90 The combina-
`tion of amikacin and vancomycin was superior to IVI
`of imipenem for treatment of experimental S. aureus
`endophthalmitis in rabbits.91
`is an antibiotic
`Chloramphenicol. Chloramphenicol
`first isolated from Streptomyces venezuelae in 1947.18 It
`has a wide spectrum of action that includes many gram-
`positive and gram-negative baceria but excludes P.
`aeruginosa.18 Systemic use of this antibiotic should be
`carefully monitored, because it may result in serious
`toxic reactions such as fatal blood dyscrasias.92 Intravit-
`real injection of chloramphenicol (1 mg), in contrast, is
`nontoxic to all intraocular structures and is effective
`against experimental endophthalmitis.93 The lack of cov-
`erage of P. aeruginosa has limited the use of IVI chlor-
`amphenicol for treatment of endophthalmitis.18
`
`Antifungal Agents
`
`Fungal endophthalmitis, representing only 3% to
`13% of all reported cases of endophthalmitis, can be a
`difficult diagnosis due to the long onset of symptoms
`and the prolonged time necessary for organism iden-
`
`Novartis Exhibit 2009.005
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`880
`
`RETINA, THE JOURNAL OF RETINAL AND VITREOUS DISEASES ● 2009 ● VOLUME 29 ● NUMBER 7
`
`tification.18 McDonnell et al94 suggested that ophthal-
`mologic screening is important in patients at high risk
`for development of systemic fungal infections, be-
`cause the eye is commonly affected in fungemia.
`Empirical treatment is given based on clinical presen-
`tation and presence of associated risk factors for fun-
`gal infection. Fungal endophthalmitis is associated with
`the use of hyperalimentation, broad-spectrum antibiotics,
`indwelling catheters, hemodialysis, cancer, intravenous
`drug use, acquired immunodeficiency syndrome (AIDS),
`or other immunocompromised state.18 Premature infants
`are more prone to development of endogenous Can-
`dida endophthalmitis, especially during treatment
`with broad-spectrum antibiotics and hyperalimenta-
`tion.95–97 Finally, geographic area may play a role,
`because postoperative fungal endophthalmitis is more
`common in the Far East than in Western societies.18
`Fungal endophthalmitis is most commonly caused by
`Candida (56% of cases) and Aspergillus (24% of
`cases).98 Endogenous Candida endophthalmitis has
`slower progression but better prognosis compared
`with other fungal endophthalmitis.99,100 One study re-
`ported a mean of 61 days from onset of symptoms to
`treatment for Candida versus 5 days for Aspergil-
`lus.100 Aspergillus endophthalmitis, which occurs in
`association with pulmonary disease and intravenous
`drug use, has a more acute presentation and a more
`rapid progression than Candida endophthalmitis.100
`Aspergillus also has an increased tendency to result in
`macular scarring, which potentially can lead to visual
`loss.101 Endophthalmitis caused by other fungal or-
`ganisms is rare.102–106 There are many treatment
`options for fungal endophthalmitis including IVI
`amphotericin B (which has significantly better in-
`traocular penetration compared with the intrave-
`nous route),107,108 systemic fluoconazole,109 sys-
`temic fluocytosine for all fungal infections except
`resistant Candida species,107,108 systemic or intravit-
`real voriconazole,98,110 –112 and caspofungin.98,113–115
`
`Polyene Antimycotics
`Amphotericin B.—Amphotericin B, a fungistatic and
`fungicidal antibiotic synthesized from Streptomyces
`nodosus strains, is currently the most effective anti-
`fungal drug available. This agent acts by binding a
`sterol on the cell membrane, resulting in permeabili-
`zation and outflow of intracellular components. How-
`ever, amphotericin B can have significant side effects
`that include severe constitutional symptoms, thrombo-
`phlebitis, hepatoxicity, nephrotoxicity, neurotoxicity,
`allergic reactions, and cardiac arrest with rapid intra-
`venous infusion.116 Intravitreal injection of amphoter-
`icin B reversed experimental Candida endophthalmi-
`tis and was nontoxic in doses up to 10 ␮g.117–124
`
`Amphotericin B was effective against zygomycetes
`endophthalmitis when 10 ␮g was administered in the
`vitreous and 10 ␮g between the iris and posterior
`capsule.122 Early vitrectomy and IVI of amphotericin
`B demonstrated efficacy in treatment of fungal en-
`dophthalmitis123,124 and in a case of Aspergillus ne-
`crotizing retinitis.125 Currently, IVI of amphotericin B
`(5 ␮g in 0.05 mL) is the drug of choice for suspected
`fungal endophthalmitis. Care must be taken while
`performing IVI of amphotericin B because apposition
`to the retina can result in retinal necrosis.18,120 The
`finding that amphotericin B is synergistic with ri-
`fampin against some Candida, Aspergillus, Penicil-
`lium, and Rhizopus strains may lead to future clinical
`applications in ophthalmology.126
`Nystatin.—Nystatin is an antifungal isolated from
`Streptomyces noursei that has a similar mechanism of
`action as amphotericin B. It is fungistatic and fungi-
`cidal against Candida, Cryptococcus, Histoplasma,
`and Blastomyces species.18 Nystatin was successful
`against experimental Aspergillus endophthalmitis if
`administered 24 hours after inoculation.127 At thera-
`peutic doses (200 U or 57.14 ␮g), a transient inflam-
`matory reaction occurs, and this may explain the rel-
`ative lack of use of IVI nystatin.127
`Natamycin.—Natamycin (pimaricin) belongs to the
`same class of polyene antifungal antibiotics as ampho-
`tericin B and nystatin. It has a wide spectrum of
`activity but its clinical utility is limited by poor ocular
`penetration.128,129 Intravitreal natamycin was thera-
`peutic in rabbits with Aspergillus endophthalmitis
`only when administered in doses that resulted in irre-
`versible retinal toxicity. Some clinicians suggest that a
`useful regimen of intravitreal natamycin may use
`25-␮g doses given at 72-hour intervals130; however,
`the practicality of such a divided dose in the face of
`known retinal toxicity has severely limited the use of
`IVI natamycin. Presently, we do not recommend that
`IVI natamycin be used except in the most extraordi-
`nary situations where other IVI antifungals have failed
`to halt the progression of fungal endophthalmitis.
`Azoles. The antibiotic agents from the azole family
`are active against dermatophytes, yeasts, fungi, some
`bacteria, and protozoa and are generally less toxic
`than amphotericin B.18 These qualities rendered them
`useful in treatment of systemic and ocular fungal
`infections.
`The imidazoles (miconazole, ketoconazole, and
`oxiconazole) have a broad spectrum of activity that
`includes Candida, Cryptococcus, and Coccidioides.
`Miconazole is fungistatic at low dosages and fungi-
`cidal at higher dosages. Intravenous miconazole has
`good penetration of the vitreous body.116 Intravitreal
`
`Novartis Exhibit 2009.006
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`INTRAVITREAL INJECTION OF THERAPEUTIC AGENTS ● PEYMAN ET AL
`
`881
`
`injection of miconozale demonstrated that it may not
`be as effective as its systemic adminstration.131,132
`Ketoconazole has a lower incidence of systemic side
`effects than miconazole, can result in hepatotoxicity,
`and has poor vitreous penetration.133 Ketoconazole is
`only available as an oral tablet or powder dissolved in
`dimethyl sulfoxide. There is a paucity of literature on
`use of intravitreal ketoconazole for treatment of fungal
`endophthalmitis. Intravitreal injection of ketoconazole
`has been mainly administered after intravitreal am-
`photericin B. In 2 patients with postoperative Candida
`parapsilosis endophthalmitis treated with a combina-
`tion of intravitreal amphotericin B, removal of the
`pseudophakos, and oral ketoconazole (800 mg/day for
`12 weeks), the efficacy of oral ketoconazole could not
`be ascertained.134 Intravitreal ketoconazole in doses
`up to 540 mg was nontoxic to ocular structures.135
`Oxiconazole has a broad spectrum of activity and is
`more effective against Candida than other imida-
`zoles.18 The use of oxiconazole is currently limited
`mainly to superficial mycoses; oxiconazole was non-
`toxic to rabbit ocular structures.136
`Triazoles (itraconazole, fluconazole, and voricon-
`azole) are newer azole antifungals that contain a third
`nitrogen on the azole ring. Their mechanism of action
`is via inhibition of ergosterol biosynthesis and damage
`of phospholipid membranes at higher dosages.137 Itra-
`conazole has a broad spectrum of activity and low
`systemic toxicity. It was successful even at low doses
`when administered early in a rabbit model of endog-
`enous Candida endophthalmitis but not when admin-
`istered 7 days after the infection.138 In a mouse model,
`itraconazole or fluconazole was synergistic when given
`in combination with flucytosine in the management of
`Candida and Aspergillus infections.139 Fluconazole is
`active against a wide variety of fungal organisms and has
`one of the best pharmacokinetic properties of all antifun-
`gal drugs.139 Although fluconazole was nontoxic to rab-
`bit eyes in doses up to 100 ␮g,138,140 it was not effective
`in a rabbit model of endogenous Candida endoph-
`thalmitis when administered in high doses at 7 days
`after infection.138 Voriconazole is a second-generation
`synthetic derivative of fluconazole that is a promising
`new modality in the treatment of fungal endophthalmi-
`tis.111 This agent has a greater ability to inhibit biosyn-
`thesis of fungal cell membranes than other antifungal
`drugs. The minimal inhibitory concentration for Candida
`species, Aspergillus fumigatus, Histoplasma capsulatum,
`and Fusarium organisms is much lower than those of
`amphotericin B, fluconazole, itraconazole, ketocon-
`azole, and 5-flucytosine.112 Intravitreal injection of
`voriconazole was effective against experimental Can-
`dida albicans endophthalmitis in a rabbit model98 and
`has demonstrated efficacy against endogenous Scedos-
`
`porium apios

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket