Review Article Ocular Toxoplasmosis: Controversies in Primary and Secondary Prevention

Similar documents
Ocular Toxoplasmosis MAJOR REVIEW. Acquired toxoplasmosis. Congenital toxoplasmosis. June 2007 Kerala Journal of Ophthalmology 141

Rare Presentation of Ocular Toxoplasmosis

Toxoplasma gondii. Definitive Host adult forms sexual reproduction. Intermediate Host immature forms asexual reproduction

Ocular Toxoplasmosis Uveitis course Antalya Miles Stanford Medical Eye Unit St Thomas Hospital London

Lecture-7- Hazem Al-Khafaji 2016

Retina Grand Rounds. Stephen Huddleston MD Charles Retina Institute University of Tennessee Hamilton Eye Institute

Toxoplasma gondii. Jarmila Kliescikova, MD 1. LF UK

Interventions for Toxoplasma Retinochoroiditis

Cerebral Toxoplasmosis in HIV-Infected Patients. Ahmed Saad,MD,FACP

Review Article Current Treatment of Toxoplasma Retinochoroiditis: An Evidence-Based Review

Moncef Khairallah, MD

Coccidia. Eucoccidioside

Ahmed M. Bawazeer MD, FRCSC 1 Nizamuddin SHM MD, FRCS(Glasg), FMRF 2

Acute Central Vision Loss in an IV Drug User

Scholars Journal of Applied Medical Sciences (SJAMS)

Toxoplasmosis Objective :

Treatment of ocular toxoplasmosis in pregnancy

Azithromycin versus Sulfadiazine and Pyrimethamine for non-vision-threatening toxoplasmic retinochoroiditis: A pilot study

Short Video. shows/monsters-inside- me/videos/toxoplasma-parasite/

GUIDELINE FOR THE MANAGEMENT OF TOXOPLASMOSIS ENCEPHALITIS

Clinical Policy: Pyrimethamine (Daraprim) Reference Number: ERX.NPA.44 Effective Date:

Extra-intestinal coccidians

Acute Retinal Necrosis Secondary to Varicella Zoster Virus in an Immunosuppressed Post-Kidney Transplant Patient

Toxoplasmosis. Life cycle Infective stages Congenital Toxoplasmosis Lab.Diagnosis Immunity to Toxoplasmosis Toxoplasmosis & Pregnancy

Various presentations of herpes simplex retinochoroiditis A case series

See Important Reminder at the end of this policy for important regulatory and legal information.

Antibiotics versus no treatment for toxoplasma retinochoroiditis(review)

Intravitreal Triamcinolone Acetonide for Macular Edema in HLA-B27 Negative Ankylosing Spondylitis

Misdiagnosed Vogt-Koyanagi-Harada (VKH) disease and atypical central serous chorioretinopathy (CSC)

Serologic Tests of IgG and IgM Antibodies and IgG Avidity for Diagnosis of Ocular Toxoplasmosis

Optical coherence tomography findings in a child with posterior scleritis

Management of uveitis

Toxoplasmosis. Seminar. For personal use. Only reproduce with permission from The Lancet Publishing Group.

How and Why Should we Care? Muge R. Kesen, MD Retina Specialty Institute (RSI)

INTERQUAL PROCEDURES CRITERIA BIBLIOGRAPHY: OPHTHALMOLOGY

Wales Neonatal Network Guideline

Human toxoplasmosis in Europe. Parasitology, Paris Descartes University, Cochin Hospital, Paris, France

Abbreviated Drug Evaluation: Fluocinolone acetonide intravitreal implant (Retisert )

A 39 years old HIV-positive black African woman with previously treated cerebral

Surgery in patients with uveitis. Lyndell Lim and Anthony Hall

Clinically Significant Macular Edema (CSME)

Bilateral acute retinal necrosis in a patient with multiple sclerosis on natalizumab

Common Causes of Vision Loss

Choroidal Neovascularization in Sympathetic Ophthalmia

International Journal of Medical and Health Sciences

Zika and the Eyes. William May, M.D. Associate Professor Co-director, Johns Hopkins Zika Center

UNDERSTAND MORE ABOUT UVEITIS UVEITIS

Case Study: Fuzz April 18th

Although photocoagulation and photodynamic PROCEEDINGS PEGAPTANIB SODIUM FOR THE TREATMENT OF AGE-RELATED MACULAR DEGENERATION *

Disseminated Nocardiosis with subretinal abscess in a patient with nephrotic syndrome-a case report

Vanderbilt Eye Institute Clinical Trials

Opportunistic infections

OCCLUSIVE VASCULAR DISORDERS OF THE RETINA

Recurrent intraocular hemorrhage secondary to cataract wound neovascularization (Swan Syndrome)

Re-emerging infections: Syphilis & Tuberculosis

ISPUB.COM. Photopsia post flu: A case of MEWDS. S Baisakhiya, S Dulani, S Lele INTRODUCTION CASE HISTORY

Uveitis unplugged: systemic therapy

Test Bank for Medical Surgical Nursing An Integrated Approach 3rd Edition by White

Diabetic maculopathy 11/ An update on. Miss Vasuki Sivagnanavel

International Journal of Case Reports in Medicine

NEPTUNE RED BANK BRICK

Eyes on Diabetics: How to Avoid Blindness in Diabetic Patient

Optic nerve changes in ocular toxoplasmosis

o White dot syndromes pattern recognition o Activity and damage o Quality of life o Key points o Idiopathic o Sarcoidosis o Multiple sclerosis

HLA-B27-related anterior Uveitis

Adventures in Posterior Segment Grand Rounds 46 W F. 46 W F central blindness. The not so fine print.

Diabetic Retinopathy Screening Program in the Cree Region of James Bay of Quebec

Study of clinical significance of optical coherence tomography in diagnosis & management of diabetic macular edema

Toxoplasmosis in the fetus and newborn: an update on prevalence, diagnosis and treatment

Double trouble: a patient with both HLA-B27 anterior uveitis and HLA-A29 birdshot chorioretinitis

PRECISION PROGRAM. Injection Technique Quick-Reference Guide. Companion booklet for the Video Guide to Injection Technique

Cases CFEH. CFEH Facebook Case #4

Shedding Light on Pediatric Cataracts. Kimberly G. Yen, MD Associate Professor of Ophthalmology Texas Children s Hospital

GLAUCOMA SUMMARY BENCHMARKS FOR PREFERRED PRACTICE PATTERN GUIDELINES

Vascular Disease Ocular Manifestations of Systemic Hypertension

Imaging in uveitis. Anthony Hall

ISPUB.COM. An Atypical Presentation of Posterior Scleritis. A Ramanathan, A Gaur CASE REPORT

2009 REIMBURSEMENT GUIDE, VISUCAM and VISUCAM NM/FA

2019 COLLECTION TYPE: MIPS CLINICAL QUALITY MEASURES (CQMS) MEASURE TYPE: Outcome High Priority


Nausheen Khuddus, MD Melissa Elder, MD, PhD

Note: This is an outcome measure and can be calculated solely using registry data.

You can C-ME after Uveitis

2018 OPTIONS FOR INDIVIDUAL MEASURES: REGISTRY ONLY. MEASURE TYPE: Outcome

Optometric Postoperative Cataract Surgery Management

PROFILE OF PATIENTS WITH FLOATERS IN SAIFUL ANWAR HOSPITAL MALANG

Congenital Toxoplasmosis

Measure #191: Cataracts: 20/40 or Better Visual Acuity within 90 Days Following Cataract Surgery

Seroprevalence of Toxoplasmosis in High School Girls in Fasa District, Iran

MANAGEMENT OF NEOVASCULAR GLAUCOMA

Differential diagnosis of posterior uveitis

A Patient s Guide to Diabetic Retinopathy

White-Spot Syndromes of the Retina Lee Jampol, M.D. Chicago, IL

Facts About Diabetic Eye Disease

Case Report Stalagmite-like preretinal inflammatory deposits in vitrectomized eyes with posterior uveitis

Uveitis. Pt Info Brochure. Q: What is Uvea?

Retina Conference. Janelle Fassbender, MD, PhD University of Louisville Department of Ophthalmology and Visual Sciences 09/04/2014

Update on management of Anterior Uveitis

DNB Question Paper. December 1

Transcription:

Ashdin Publishing Journal of Neuroinfectious Diseases Vol. 4 (2013), Article ID 235689, 5 pages doi:10.4303/jnd/235689 ASHDIN publishing Review Article Ocular Toxoplasmosis: Controversies in Primary and Secondary Prevention Norman A. Saffra, 1,2 Carly J. Seidman, 1,2 and Louis M. Weiss 3 1 Division of Ophthalmology, Maimonides Medical Center, Brooklyn, NY 11219, USA 2 Department of Ophthalmology, New York University, New York, NY 10016, USA 3 Departments of Medicine and Pathology, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY 10461, USA Address correspondence to Norman A. Saffra, eyesitemd@gmail.com Received 6 January 2013; Accepted 23 February 2013 Copyright 2013 Norman A. Saffra et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Ocular toxoplasmosis is the most common cause of posterior uveitis in the United States and worldwide. It commonly follows a relapsing course with the potential for multiple adverse visual sequelae and, rarely, blindness. Both immune-competent and immune-suppressed populations can develop severe relapsing disease. There is considerable debate regarding the initial treatment of ocular toxoplasmosis in both groups, with multiple antiparasitic agents commonly used with little evidence to support any particular regimen. The precise role of corticosteroids in treatment also has yet to be rigorously defined. Secondary prevention is commonly implemented in the immune suppressed population, but there is a lack of level one evidence to support its role in immune-competent patients, though it warrants future investigation. Keywords antiparasitic therapy; chemoprophylaxis; infectious retinal disease; ocular toxoplasmosis; retinochoroiditis; secondary prevention In humans, this parasite can be acquired in utero or postnatally from environmental exposure. This can be through contact with cat feces or litter or ingestion from a variety of routes, including infected water, produce, or undercooked meat (pregnant seronegative women should therefore avoid these exposures). It can also be acquired via blood transfusion or organ transplantation. T. gondii tachyzoites can invade any host cells in the body, including those of the retina and choroid, where it transforms to bradyzoites (tissue cysts) and becomes dormant, but can reactivate and cause infection at any time. Toxoplasma retinochoroiditis may be the result of the immune system reaction to parasite antigens [15]. 1. Epidemiology Toxoplasma gondii is the most common agent causing retinal infection in the United States, with over one million individuals infected with the intracellular parasite. Of this number, over 21,000 individuals develop ocular toxoplasmosis, and nearly 5,000 manifest visual symptoms [9]. Surprisingly, given its ubiquity, the treatment of ocular toxoplasmosis is the subject of much controversy even among uveitis experts [2,8,21]. 2. Pathophysiology Ocular toxoplasmosis can take on a prolonged and relapsing course in humans. T. gondii exists in three forms: oocyst (sexual life cycle stage), tachyzoite (rapidly replicating form), and bradyzoite (latent tissue cyst life cycle stage) (Figure 1). Cats are the definitive host the parasite reproduces in feline intestines and is shed in the feces in large numbers. Transmission occurs when oocysts (which contain sporozoites) or tissue cysts (which contain bradyzoites) are ingested (Figure 2). Figure 1: SAG1 antibody-stained T. gondii tachyzoites.

2 Journal of Neuroinfectious Diseases Figure 2: H&E stain demonstrating T. gondii cyst in brain tissue. Figure 4: Color fundus photo of same patient midway through therapy with Bactrim and prednisone, showing decrease in size of active lesion and resolution of vitritis. Figure 3: Color fundus photo from a 34-year-old immunocompetent female with multiple recurrences in the posterior pole. A. Weiss ring. B. Area of vitritis overlying active lesion. C. Chronic scarring from prior episodes. 3. Signs and symptoms The ophthalmic presentation of ocular toxoplasmosis typically depends on the retinal location of the lesion (central or peripheral retina) and the associated degree of inflammation (Figures 3, 4, and 5). Patients classically present with a unilateral decrease in vision with associated floaters. Pain, redness, and photophobia (symptoms of acute anterior uveitis) can be present as well. The classic presentation of congenital toxoplasmosis includes large macular retinochoroiditis, hydrocephalus, and intracranial calcification. Recurrent disease is marked by the presence of active lesions in the setting of old pigmented retinal scars in either eye. Figure 5: Color fundus photo following eight weeks of therapy with Bactrim DS and prednisone, demonstrating resolution of the active lesion, clearing of vitritis, and increased scarring in the posterior pole. 4. Diagnosis The diagnosis of toxoplasmosis infection is usually made by physical examination, via a dilated funduscopic examination with the use of serology as a confirmation. The retinitis is most often located in the posterior pole and occasionally adjacent to or involving the optic nerve. Serologic testing via ELISA and indirect fluorescent antibody testing is

Journal of Neuroinfectious Diseases 3 commonly used to confirm the diagnosis of ocular toxoplasmosis. The presence of IgG antibodies to T. gondii (which appear two weeks after exposure) confirms past exposure and titers remain detectable for life. The presence of IgM or IgA antibodies signal the acute phase and remain elevated for approximately one year after exposure. The presence of IgM antibodies, therefore, likely confirms the presence of active disease [7]. PCR amplification of T. gondii DNA is more sensitive and specific and can also be used as confirmatory. However, serologic testing is not prognostic as there is no correlation between antibody titers, DNA load, and severity of ocular disease [4]. 5. Disease course The impact of ocular toxoplasmosis is far ranging with the potential for cataract, glaucoma, vitreous opacification or hemorrhage, macular scarring, macular cystoid edema, choroidal neovascular membrane, retinal detachment, vascular occlusion, and optic nerve involvement [15]. Punctate outer retinal toxoplasmosis (PORT), characterized by small, deep multifocal lesions with overlying vitreous inflammation, can also occur. The development of these various sequelae often depends upon the patient s immune status and age. The immune compromised and elderly are at higher risk for severe adverse outcomes and bilateral involvement over time [5]. Although infection in the immune-competent host most commonly results in an acute, self-limited chorioretinitis (lasting 6 to 8 weeks), with the likely formation of a retinal scar, these patients are still at risk for recurrence of infection, causing additive damage each time. The factors that predispose certain individuals to recurrent infection are likely multifactorial and may be related to the strain of parasite and age of the patient, but there are no studies to support this. Treatment of ocular toxoplasmosis is controversial, as multiple regimens are used, and there is little evidence in support of their relative efficacy and in which populations to implement them [6]. Whereas secondary prevention is commonly implemented in immune-compromised patients, there is debate as to whether immune-competent patients should be candidates for long-term prophylaxis against recurrence [8,21]. 6. Therapy The goal of treating toxoplasmosis is to limit the duration of active parasitic infection so that cicatrization can occur more quickly, and the extent of structural complications is limited. It is known that a combination of glucocorticoids and antibiotics treating immune-competent patients, in whom the infection is self-limited, is likely effective in altering the short-term natural course of the disease. More evidence is needed to evaluate longer therapies. In considering whether to treat, the benefits of treatment must be weighed against the potential risks associated with antibiotic therapy. Such adverse effects range from gastrointestinal distress to rash, thrombocytopenia, leucopenia, and drug allergy/anaphylaxis. Due to these rare but possible side effects, some experts opt against treating low-risk cases in immune-competent patients who exhibit only small peripheral lesions [2, 21]. However, it is agreed that the immune-suppressed state, pregnancy, and congenital toxoplasmosis (diagnosed within the first year of life) are absolute indications for therapy [2,21]. In choosing a therapeutic regimen, multiple choices are available, although no evidence-based standards are available. Among members of the American Uveitis Society, 9 different antiparastic agents were used in 24 different combinations [8]. There have only been three randomized placebo-controlled trials investigating the efficacy of different antibiotic therapies, and these are methodologically weak and outdated [6]. Most therapeutic regimens are, thus, chosen based upon a limited number of case studies and individual ophthalmologists preferences. Classic therapy, triple therapy, is the most commonly implemented regimen and includes pyrimethamine (loading dose of 50 to 100 mg then 25 to 50 mg QD), sulfadiazine (2 to 4 g loading dose then 1 g QID), and oral corticosteroids (0.5 to 1.0 mg/kg QD). Sulfadiazine is known to cause a rare but severe systemic anaphylactic reaction. Pyrimethaimine is known to cause pancytopenia and requires weekly monitoring of blood counts; folinic acid is usually coadministered with the drug to prevent bone marrow suppression. Quadruple therapy also includes clindamycin (300 mg QID) [8]. Bactrim (trimethoprim-sulfamethoxazole) (160 mg/80 mg BID) has been successfully substituted in place of pyrimethamine and sulfadiazine [19]. Other newer agents, including macrolides, tetracyclines, atovaquone, and antiparasitic agents, have also been used when there is intolerance to classic therapy [1,16]. Duration of therapy typically spans from four to six weeks or in the immunosuppressed as long as one year [2, 21]. Newborns are usually treated with pyrimethamine and sulfonamides for one year. Local therapy utilizing intravitreal injections of clindamycin is an emerging regimen that can be potentially used as an adjunct to local therapy or as an option for patients with contraindications to systemic therapy (or pregnant women to reduce teratogenicity). It is known to achieve good intracellular concentration in the intraocular tissues, thus targeting the parasite [18] is less expensive than most antiparasitic agents, and compliance is less of an issue than with oral regimens. The recommended dose ranges from 1.0 mg/0.1 ml to 1.5 mg/0.1 ml and is administered between one to four times (given 2 weeks apart), or until complete clinical improvement is achieved [21]. In one study, intravitreal clindamycin in conjunction with local dexamethasone injections were shown to be

4 Journal of Neuroinfectious Diseases effective in controlling toxoplasma retinochoroiditis and improving visual acuity at 11 to 26 months follow-up; on average, two to four injections were required [11]. Another clinical trial compared intravitreal clindamycin with local dexamethasone injections to classic therapy and found them comparable in terms of reduction of lesion size, decreased vitritis, and improvement in visual acuity. Classic therapy was noted to be more effective in IgM positive patients, however, suggesting that intravitreal therapy might be less appropriate in patients with reactivation toxoplasmosis [18]. As the sample sizes were small, however, more adequately powered studies are needed to assess the efficacy of intravitreal therapy. Regimens are often altered in the setting of pregnancy so as to avoid teratogenic effects. Some experts avoid systemic therapy and recommend intravitreal clindamycin instead [13, 18, 21]. If systemic therapy is to be used, spiramycin is considered the preferred agent in the first trimester, although it does not cross the placenta or treat the fetus. Classic triple therapy is considered to be acceptable beginning in the second trimester, however, and some advocate earlier use of these agents. Corticosteroids (topical, oral, and intravitreal) also commonly play a role in the treatment of ocular toxoplasmosis, but as with antimicrobial therapy, there are no evidence based standards to quantify or support their use. They are thought to reduce intraocular inflammation, but do not treat the infectious component of the disease [21]. Monotherapy with corticosteroids is, therefore, not recommended. Without concomitant antibiotic use, steroids can actually cause tissue destruction, especially in immune compromised patients. Moreover, corticosteroid monotherapy does not prevent disease recurrence [3]. Many experts recommend adding corticosteroids to antiparasitic therapy in the presence of vitritis, macular edema, lesions close to the fovea, optic neuritis, and in monocular patients [21]. Most experts also agree on prescribing topical corticosteroids in the presence of anterior uveitis [21]. Concerning the time frame relative to antiparasitic therapy, some experts initiate corticosteroids simultaneously, whereas others wait anywhere from 1 day up until 1 week. There is a lack of consensus as to duration of corticosteroid therapy [8]. Of note, surgical options are also occasionally implemented in the treatment of ocular toxoplasmosis. These include photocoagulation, cryotherapy, and vitrectomy; but their efficacy has not been proven. Use of such modalities on an actively inflamed retinal can be dangerous [15]. 7. Secondary prevention It has been acknowledged that the short-term treatment of ocular toxoplasmosis does not prevent the long-term recurrence of disease. Recurrence of ocular toxoplasmosis is common; in treated patients, rates have ranged from 40% [20] to 79%, although studies have been limited by follow-up [20]. Therefore, there is a question as to whether to initiate chemoprophylaxis in certain patients as secondary prevention; the long-term impact on relapse, scarring, and visual acuity has not been studied. In T. gondii-seropositive immune-suppressed patients with signs of ocular disease, chemoprophylaxis is common practice and may reduce disease occurrence [10]. There are no evidence-based recommendations regarding therapy, but some recommendations state that following resolution of active disease that the original treatment be continued at one half of the initial dose for the lifetime of the patient or until the immune-suppressive condition has ceased. In patients with HIV, prophylaxis should be continued until the CD4 count rises above 200 [14]. Less clear is whether immune-competent patients should receive chemoprophylaxis. It has been recommended in patients with more severe disease, history of more frequent recurrences (i.e., greater than one per year or three in two years) and those at high risk of vision loss (i.e., those with significant scarring in the posterior pole or adjacent to the fovea). Postoperative prophylaxis has also been recommended in patients with a history of toxoplasmosis following cataract extraction or vitrectomy [13]. Some experts actually recommend a course of chemoprophylaxis in all patients treated for active disease, as it has been hypothesized that recurrence risk is greatest early on but decreases over time [17]. A commonly described regimen consists of macrolide therapy to avoid eliciting an allergy to sulfonamides during chemoprophylaxis. Silviera used one tablet of trimethoprimsulfamethoxazole (TMP-SMX) (160/800) every three days, although other regimens have also been used this is often chosen for its convenience, cost effectiveness, and low rate of side effects. Silviera et al. [17] found that this regimen was associated with significantly fewer recurrences of disease, and other case studies also report success with TMP- SMX [12], but studies have been limited by study bias, sample size, and duration of follow-up. It should be noted that TMPSMX is not as effective as pyrimethamine and sulfadiazine in the treatment of acute disease; therefore, although it has fewer side effects, it might not offer the best protection against recurrence over time. More information on TMP- SMX as well as other prophylactic regimens is needed. 8. Conclusions Despite the ubiquity of ocular toxoplasmosis, there remains a lack of standardized evidence regarding its treatment and prevention. However, in general, the trend does appear to be more aggressive treatment, even of milder disease [8]. There is a need for well-controlled trials to determine the relative efficacy of initial therapies as well as to explore chemoprophylactic regimens and their sight-saving impact.

Journal of Neuroinfectious Diseases 5 Acknowledgments The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper. Louis M. Weiss is supported by NIH-NIAID grant AI094094-02. References [1] K. Balaskas, J. Vaudaux, N. Boillat-Blanco, and G.-C. Y., Azithromycin versus sulfadiazine and pyrimethamine for nonvision-threatening toxoplasmic retinochoroiditis: a pilot study, Med Sci Monit, 18 (2012), 296 302. [2] A. A. Bonfioli and F. Orefice, Toxoplasmosis, Semin Ophthalmol, 20 (2005), 129 141. [3] L. E. Bosch-Driessen, T. T. Berendschot, J. V. Ongkosuwito, and A. Rothova, Ocular toxoplasmosis: clinical features and prognosis of 154 patients, Ophthalmology, 109 (2002), 869 878. [4] G. Bou, M. S. Figueroa, P. Martí-Belda, E. Navas, and A. Guerrero, Value of PCR for detection of Toxoplasma gondii in aqueous humor and blood samples from immunocompetent patients with ocular toxoplasmosis, J Clin Microbiol, 37 (1999), 3465 3468. [5] A.G.Commodaro,R.N.Belfort,L.V.Rizzo,C.Muccioli, C. Silveira, M. N. Burnier Jr., et al., Ocular toxoplasmosis: an update and review of the literature, Mem Inst Oswaldo Cruz, 104 (2009), 345 350. [6] R. E. Gilbert, S. E. See, L. V. Jones, and M. S. Stanford, Antibiotics versus control for toxoplasma retinochoroiditis, Cochrane Database Syst Rev, (2002), CD002218. [7] G. N. Holland, Ocular toxoplasmosis: a global reassessment. Part II: disease manifestations and management, Am J Ophthalmol, 137 (2004), 1 17. [8] G.N.HollandandK.G.Lewis,An update on current practices in the management of ocular toxoplasmosis, Am J Ophthalmol, 134 (2002), 102 114. [9] J. L. Jones and G. N. Holland, Annual burden of ocular toxoplasmosis in the United States, Am J Trop Med Hyg, 82 (2010), 464 465. [10] S. J. Kim, I. U. Scott, G. C. Brown, M. M. Brown, A. C. Ho, M. S. Ip, et al., Interventions for toxoplasma retinochoroiditis: a report by the American Academy of Ophthalmology, Ophthalmology, 120 (2013), 371 378. [11] K. Kishore, M. D. Conway, and G. A. Peyman, Intravitreal clindamycin and dexamethasone for toxoplasmic retinochoroiditis, Ophthalmic Surg Lasers, 32 (2001), 183 192. [12] R. Kopec, G. De Caro, E. Chapnick, M. Ghitan, and N. Saffra, Prophylaxis for ocular toxoplasmosis, Clin Infect Dis, 37 (2003), e147 e148. [13] C. Martinez, D. Zhang, M. Conway, and G. Peyman, Successful management of ocular toxoplasmosis during pregnancy using combined intraocular clindamycin and dexamethasone with systemic sulfadiazine, Int Ophthalmol, 22 (1998), 85 88. [14] J. G. Montoya and L. O., Toxoplasmosis, Lancet, 363 (2004), 1965 1976. [15] S. Pineles, S. M. Rikkers, and R. D. Levinson, Ocular toxoplasmosis, in Ophthalmology, M. Yanoff and J. S. Duker, eds., Mosby, Maryland Heights, MO, 3rd ed., 2009. [16] A. Rothova, L. Bosch-Driessen, N. van Loon, and W. Treffers, Azithromycin for ocular toxoplasmosis, Br J Ophthalmol, 82 (1998), 1306 1308. [17] C. Silveira, R. Belfort Jr., C. Muccioli, G. N. Holland, C. G. Victora, B. L. Horta, et al., The effect of long-term intermittent trimethoprim/sulfamethoxazole treatment on recurrences of toxoplasmic retinochoroiditis, Am J Ophthalmol, 134 (2002), 41 46. [18] M. Soheilian, A. Ramezani, A. Azimzadeh, M. M. Sadoughi, M. H. Dehghan, R. Shahghadami, et al., Randomized trial of intravitreal clindamycin and dexamethasone versus pyrimethamine, sulfadiazine, and prednisolone in treatment of ocular toxoplasmosis, Ophthalmology, 118 (2011), 134 141. [19] M. Soheilian, M. M. Sadoughi, M. Ghajarnia, M. H. Dehghan, S. Yazdani, H. Behboudi, et al., Prospective randomized trial of trimethoprim-sulfamethoxazole versus pyrmethamine and sulfadiazine in the treatment of ocular toxoplasmosis, Ophthalmology, 112 (2005), 1876 1882. [20] J. C. Timsit and E. Bloch-Michel, Efficacy of specific chemotherapy in the prevention of recurrences of toxoplasmic chorioretinitis during the 4 years following treatment, J Fr Ophtalmol, 10 (1987), 15 23. [21] D. Wakefield, E. T. Cunningham Jr., C. Pavesio, J. G. Garweg, and M. Zierhut, Controversies in ocular toxoplasmosis, Ocul Immunol Inflamm, 19 (2011), 2 9.