s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
Available online at www.sciencedirect.com
journal homepage: www.elsevier.com/locate/survophthal
Major review
Paraneoplastic and non-paraneoplastic retinopathy and optic neuropathy: Evaluation and management Ehsan Rahimy, MDa, David Sarraf, MDa,b,* a b
Jules Stein Eye Institute, UCLA, Los Angeles, California, USA Greater LA VA Healthcare Center, Los Angeles, California, USA
article info
abstract
Article history:
Paraneoplastic syndromes involving the visual system are a heterogeneous group of
Received 29 June 2012
disorders occurring in the setting of systemic malignancy. Timely recognition of one of
Received in revised form
these entities can facilitate early detection and treatment of an unsuspected, underlying
27 August 2012
malignancy, sometimes months before it would have otherwise presented, and gives the
Accepted 4 December 2012
patient an increased chance at survival. We outline the clinical features, pathogenesis, and treatment strategies for the retinal- and optic nerveebased paraneoplastic syndromes: cancer-associated retinopathy; melanoma-associated retinopathy; paraneoplastic vitelli-
Keywords:
form maculopathy; bilateral diffuse uveal melanocytic proliferation; paraneoplastic
paraneoplastic
optic neuropathy; and polyneuropathy, organomegaly, endocrinopathy, monoclonal
cancer-associated retinopathy
gammopathy, and skin changes syndrome. Distinguishing these disorders from their non-
melanoma-associated retinopathy
paraneoplastic counterparts (e.g., autoimmune-related retinopathy and optic neuropathy,
vitelliform maculopathy
and acute zonal occult outer retinopathy) and determining appropriate systemic evalua-
bilateral diffuse uveal melanocytic
tion for the responsible tumor can be challenging. In addition, we discuss the utility and
proliferation
interpretation of autoantibody testing.
optic neuropathy
Published by Elsevier Inc.
POEMS syndrome autoimmune-related retinopathy and optic neuropathy anti-retinal antibody autoantibody testing
1.
Introduction
Paraneoplastic syndromes are complexes of signs and symptoms in cancer-bearing patients resulting from dysfunction of tissues remote from the site of a malignant neoplasm or its metastases.57,58 Most of these syndromes occur as a result of ectopic tumor production of a hormone or functional growth factor acting at a distal site (e.g., Cushings syndrome,
syndrome of inappropriate antidiuretic hormone secretion, hypercalcemia secondary to parathyroid hormone-related protein, and carcinoid syndrome), whereas others are believed to involve immune-mediated cross-reactivity between tumor antigens and normal host tissues (e.g., Lambert-Eaton myasthenic syndrome, paraneoplastic cerebellar degeneration, and opsoclonus-myoclonus). The true incidence and prevalence of paraneoplastic syndromes
* Corresponding author: David Sarraf, MD, Retinal Disorders and Ophthalmic Genetics Division, Jules Stein Eye Institute, UCLA, Los Angeles, CA 90024. E-mail address:
[email protected] (D. Sarraf). 0039-6257/$ e see front matter Published by Elsevier Inc. http://dx.doi.org/10.1016/j.survophthal.2012.09.001
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
431
Fig. 1 e Proposed pathophysiologic mechanisms underlying the intraocular paraneoplastic syndromes.
remains unknown, but are estimated to occur in as many as 10% of cancer patients.149 Those involving the nervous and visual systems are considerably less common, affecting as little as 0.01% of patients with cancer evaluated at one tertiary care oncology hospital.57,58 In addition to their infrequency, these conditions can be easily overlooked given their subtle signs, making their detection all the more challenging. Here, we review the retinal- and optic nerve-based paraneoplastic syndromes: cancer-associated retinopathy (CAR); melanoma-associated retinopathy (MAR); paraneoplastic vitelliform maculopathy (PVM); bilateral diffuse uveal melanocytic proliferation (BDUMP); paraneoplastic optic neuropathy (PON); and polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes (POEMS) syndrome. The proposed pathophysiologic mechanisms underlying each syndrome vary and continue to evolve as we learn more about their complex origins (Fig. 1). Traditionally, molecular mimicry has been purported to underlie the development of the paraneoplastic retinopathies and optic neuropathies. By this mechanism, susceptible individuals mount an immune response to cancer antigens sharing homology with endogenous proteins of the retina and/or optic nerve. The resultant antibodies then inappropriately crossreact with and localize to these tissue sites, where they disrupt normal cellular function, and ultimately cause visual dysfunction. Recent research efforts, however, have revealed that tumor-expressed growth factors may also contribute to the development of these diseases, notably BDUMP and POEMS. Patients presenting with visual dysfunction of suspected paraneoplastic origin warrant a thorough systemic evaluation for underlying malignancy. We recommend diagnostic modalities, and discuss differentiating these syndromes from their non-paraneoplastic counterparts (e.g., autoimmunerelated retinopathy and optic neuropathy [ARRON], and acute zonal occult outer retinopathy [AZOOR]), which are likely more common than the paraneoplastic variety.58,101 Finally, we explore the utility and interpretation of autoantibody testing, a beneficial adjunct if correlated appropriately within the clinical context.
2.
Paraneoplastic retinopathies
2.1.
Cancer-associated retinopathy
2.1.1.
Clinical Presentation
CAR is considered the most common of the intraocular paraneoplastic syndromes. In 1976, Sawyer et al251 presented the
first three patients with retinal degeneration associated with cancer. Although there are no prevalence estimates for CAR, its incidence appears to be on the rise as the result of increased clinical awareness and improved diagnostic surveillance. As evidence, since Chan’s48 review in 2003 that examined the 55 known cases of CAR at the time, the number of reported cases has more than doubled. The primary tumor accounting for the vast majority is small-cell lung carcinoma,48 followed by gynecologic (ovarian, endometrial, and cervical)4,70,140,185,207,230,246,251,255,264 and breast17,68,182 malignancies. Less commonly reported solid tumor associations prostate,173 include non-small-cell lung,248,253 124,229,274,293 166 95 thymus, thyroid, pancreatic, colon,113 and 173 cancers. Hematological malignancies (leukemia, bladder lymphoma, myeloma) have also been linked to CAR.2,212,278 The average age of symptom onset in patients with CAR is 65 years.2,48 Historically, there has been no known sex predilection, although these demographics may be changing after Adamus2 recently reported a 2:1 female-to-male ratio. Visual loss is painless, develops over weeks to months, and precedes the diagnosis of underlying malignancy in nearly half of patients.19,48,115,247 Symptomatic complaints reflect both rod and cone photoreceptor dysfunction. Manifestations of conerelated compromise include photosensitivity, photopsias, glare, severely reduced central vision, and impaired color perception. Rod dysfunction may present as nyctalopia, impaired dark adaptation, ring scotoma, or other peripheral visual field loss. A lesser-known variant of CAR, termed cancer-associated cone dysfunction, characteristically affects only cones.41,53,75,114,215 Of note, recent evidence suggests photosensitivity may not be as characteristic for CAR as previously thought. In a case series by Ohguro et al,206 photosensitivity was documented in only 2 of 18 patients. Fundus examination in CAR varies from unremarkable early on to findings of optic nerve pallor, attenuated retinal arterioles, and retinal pigment epithelial (RPE) thinning and mottling later in the disease course. Retinal pigment dispersion is typically absent or sparse.101 A subtle vitritis may be present, which may coincide with an anterior uveitis, and there may also be retinal vasculitis with or without cystoid macular edema (CME).17,48,109,140,206,208,212,230,272 Rare presentations of CAR with associated choroidal neovascularization229 and in combination with BDUMP246 have also been described. Ancillary studies, especially anti-retinal antibody testing and electroretinogram (ERG), are helpful in establishing the diagnosis of CAR, as well as in the evaluation of any
432
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
paraneoplastic retinopathy. Identification of circulating antiretinal antibodies in patient sera is usually confirmed by either Western blot or enzyme-linked immunosorbent assay (ELISA) of a patient’s serum against normal retina extract. Immunohistochemical techniques localizing antibodies in patient sera to the photoreceptor layer of donor retinal sections can be performed. The most sensitive and specific antibody for CAR, when one is detected, is against the 23-kDa retinal antigen, recoverin. Additional cases of CAR have demonstrated antibodies against the 46-kDa antigen, aenolase. Weleber et al288 noted a more rapid onset and visual decline, often prior to the cancer diagnosis, in cases of antirecoverin CAR. In contrast, anti-enolase CAR tended to have a more subacute presentation with less severe course, often in patients with a known history of malignancy. The ERG in CAR classically depicts global retinal dysfunction, with severely reduced, if not extinguished, scotopic and photopic a- and b-waves.144,173,251 This reduction in amplitude is seen early on in the disease course, even while funduscopic exam may be entirely normal. An electronegative ERG, more typical of MAR, has also been described in three individuals with CAR.17,91,140 Visual field deficits can manifest as generalized depression, or central, paracentral, arcuate, or ring scotomas.206 While fluorescein angiography (FA) is unremarkable in the vast majority of CAR patients, it is nonetheless useful to obtain for ruling out other posterior segment pathology. Isolated cases have documented perivascular leakage and CME on FA.17,109,140,171,208,230,272 Optical coherence tomography (OCT) typically reveals severe macular atrophy, often associated with thinning of the photoreceptor layer,166,215 loss of the inner segment/outer segment (IS/OS) junction,109 and loss of the inner highly reflective layer.185,255 Lima et al157 advocated the additional use of fundus autofluorescence together with high-definition spectral domain OCT to aid in the diagnosis of this entity and monitoring for progression. In their report, four patients with autoimmunerelated retinopathy (two with small-cell lung cancer) displayed a hyperautofluorescent ring in the parafoveal region on fundus autofluorescence, and the corresponding OCT over this region demonstrated loss of the IS/OS junction and thinning of the outer nuclear layer. The increased zone of autofluorescence is suggestive of compromised RPE function due to increased metabolic demand, and may be a precursor to cell apoptosis and further visual compromise.157 Finally, systemic evaluation in cases of suspected CAR may reveal elevated levels of cerebrospinal fluid (CSF) protein and lymphocytosis.19
2.1.2.
Pathogenesis
Molecular mimicry is the generally accepted pathogenic mechanism driving CAR.15,48 By this process, tumor cells expressing antigenic epitopes with retinal photoreceptors elicit an immune response that cross-reacts with the retina, disrupting normal signal transmission. In 1983, Keltner et al128 demonstrated the presence of anti-retinal antibodies in the serum of a patient with CAR who was responsive to steroid therapy, suggesting an autoimmune basis for the disease process. This antibody was later identified as the 23-kDa CAR antigen, or recoverin, by Thirkill et al,276,277 a finding subsequently confirmed by Polans et al.222e224 To date, over 18
retinal antigens have been implicated in CAR, with the most commonly identified ones being recoverin and a-enolase (46-kDa). Recoverin is a calcium-binding protein located within retinal photoreceptor cells. Via a calcium-dependent process, it regulates phosphorylation of rhodopsin during visual transduction. The human recoverin gene has been mapped to chromosome 17p13.1, a region containing a number of other oncogenic loci, notably p53.174,175,191 McGinnis et al174 postulated that a single mutational event inactivating a copy of the p53 tumor suppressor gene may be sufficient to increase expression of the adjacent recoverin gene by tumor cells. Indeed, aberrant expression of recoverin or recoverin-like peptides has been previously detected in tumor tissue obtained from CAR patients with small-cell lung carcinoma.28,172,224,294 Maeda et al164 separately measured recoverin expression in 21 of 31 tumor cell lines. This extraocular expression of the protein has been shown to be both highly antigenic and uveogenic in a rat model, in which immunization of animals with purified recoverin led to increased antibody titers, photoreceptor degeneration, vitreous cells, perivasculitis, and retinal lesions.11 A separate rat model of CAR, whereby intravitreal injections of anti-recoverin antibodies recapitulated many features of the disease, has provided valuable insight into the potential mechanism of immune-mediated photoreceptor destruction.9,165,203 Antirecoverin antibodies have been shown to incorporate into photoreceptors where they likely block the function of endogenous recoverin. This results in unregulated rhodopsin phosphorylation, leading to increased intracellular calcium levels, which triggers activation of a caspasedependent apoptotic cascade, and ultimately, photoreceptor death.9,10,13,165,203 These findings are supported by postmortem histopathologic analyses of patients with CAR revealing patchy loss of photoreceptors in conjunction with immunohistochemical staining of antibodies localizing to the outer plexiform layer, the outer nuclear layer, and the inner and outer segments of most photoreceptors.238 By injecting anti-recoverin antibodies into the rat vitreous cavity, the blooderetinal barrier, which normally protects retinal antigens from immune surveillance, is bypassed. Autoantibodies, however, do indeed cross the blooderetinal barrier into the intraocular fluid, as evidenced by Ohguro et al,202 who detected anti-recoverin antibody within the aqueous humor of a patient with CAR. How these autoantibodies cross the blooderetinal barrier to access their intra-retinal targets in CAR remains poorly understood. Of note, intravenously administered anti-recoverin antibodies in a murine model could not pass through the blooderetinal barrier and did not induce apoptotic cell death.138 Likely, secondary factors facilitate the actions of the primary autoantibodies. Tumorderived vascular endothelial growth factor (VEGF) and placental growth factor have been postulated to induce significant retinal vascular remodeling via the VEGF receptor 1. The resulting ablation of pericytes and breakdown of the blooderetinal barrier would lead to increased vascular leakage and antibody access to the intra-retinal space.42,43 Although recoverin is the most frequently implicated antigen in CAR, other immunogenic factors may also be at play. In a recent cohort reported by Adamus and colleagues,2
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
only 65% of patients with CAR had detectable serum antiretinal autoantibodies that, when present, were not always directed against a well-characterized antigen. Antibodies to recoverin were present in only 10% of those patients.2 They also identified targets such as a-enolase (46-kDa) and transducin-a (40-kDa) in 30% and 17% of patients, respectively.2 Antibodies to retinal a-enolase appear to target the ganglion cell and inner retinal layers, whereas those of transducina localize to the outer and inner segments of photoreceptors, as well as the cytoplasm of ganglion cells. This variation in immunogenic profile, they argue, accounts for the phenotypic diversity of the clinical manifestations of CAR.7 Specifically, they propose that different retinal autoantigens can be used as biomarkers for different subtypes of CAR: 1) anti-recoverin phenotype typically presents with equal and severe loss of the rod and cone system that may rapidly progress to blindness with widespread panretinal degeneration and even an extinguished ERG, while being associated with cancer in almost all cases; 2) anti-enolase retinopathy presents with isolated cone dysfunction with or without optic atrophy (due to ganglion cell loss) that is typically more indolent (or stable) without severe vision loss, and is associated with an underlying cancer in only 40% of cases; and 3) anti-transducin-a retinopathy that presents as a primary rod degeneration that is mildly progressive and is associated with cancer only about 25% of the time.2,7,288 Numerous other potential CAR antigens continue to be described, including arrestin (48-kDa),182 carbonic anhydrase II (CAII, 30-kDa),2,97 tubby-like protein 1 (TULP1, 78-kDa),137 interphotoreceptor retinoid-binding protein (IRBP, 145kDa),229 heat shock cognate protein 70 (hsc-70, 70-kDa),204 and photoreceptor cell-specific nuclear receptor (PNR, 41-kDa).67 It is clear that CAR is an immunologically heterogeneous condition associated with numerous anti-retinal autoantibodies. The clinical utility of such antigenic targets, however, needs to be better defined.
2.1.3.
Management
Given CAR’s propensity to precede the discovery of malignancy, maintaining a high index of suspicion is of utmost importance in making a timely diagnosis. Jacobson et al115 previously proposed a clinical triad of photosensitivity, ring scotomata, and attenuated arteriole caliber. A simpler variation, offered by Sobrin,A advocates consideration for CAR in cases of unexplained vision loss and visual field deficits in the setting of a near normal examination. The detection of antiretinal antibodies, especially against recoverin, further justifies pursuit of an occult malignancy (see section IV, Systemic Evaluation in the Patient Without Known Malignancy). Although no effective treatment for CAR exists, long-term immunosuppression remains the mainstay of therapy. Treatment of the underlying malignancy alone with surgery, chemotherapy, and radiation therapy does not improve vision; thus, intervention must be targeted at the ocular disorder.48,135,260 Despite aggressive immunosuppressive therapy, the visual prognosis is poor, and rapid, relentless visual loss often occurs. Some benefit is obtained from combinations of systemic corticosteroids,128,255,273 plasmapheresis,195 and intravenous
433
immunoglobulin (IVIg)95 that presumably decrease circulating autoantibody titers and their resultant destruction of photoreceptors. Keltner et al131 reported a case of steroidresponsive CAR in a patient whose circulating antibody levels diminished in association with improved visual function. Likewise, Murphy et al195 observed a decline in antiretinal antibody (60-kDa) titers from 1:2,000 to 1:200 in a patient after treatment with oral corticosteroids in combination with plasmapheresis. Visual acuity improved from counting fingers to 20/200 in the right eye (OD), and from 20/40 to 20/25 in the left eye (OS). Steroid-sparing immunomodulatory therapy (IMT) shows some promising results. Ferreyra et al72 reported improvement of visual acuity and visual fields in six out of six patients treated for CAR with IMT. The caveat was that all patients were also on systemic corticosteroids, and no patients received the same IMT regimen, which consisted of azathioprine, cyclosporine, or mycophenolate mofetil, either alone or in combination. Unfortunately, toxicity limits the use of many of these agents. Monoclonal antibodies targeting B-cell lymphocytes offer a new avenue for future therapy. Espandar et al71 first reported on the successful use of alemtuzumab (anti-CD52, panlymphocytic marker) to treat several bouts of CAR exacerbation over an 8-year period. Previous treatment with steroids, plasmapheresis, and cyclosporine had failed, but a dramatic improvement in symptoms, visual acuity, and visual fields occurred after each infusion of alemtuzumab. Mahdi et al166 described a patient who responded to rituximab (anti-CD20, B-cell marker) after showing no response to either prednisone or IVIg. There has been concern that using IMT to reduce the antibody response may increase cancer mortality; however, this has not yet been observed.E Ohguro et al203 suggested a role for inhibiting antirecoverin-mediated photoreceptor apoptosis via pharmacologic reduction of intracellular calcium levels. In a rat model of CAR, use of the calcium channel blocker nilvadipine led to recovery of ERG function. Adamus et al13 found that use of the calcium channel blocker nifedipine inhibited the entry of calcium into rat retinal cells and protected them from undergoing apoptosis. How these findings translate to clinical practice remains to be seen, but in a patient treated with a systemic calcium channel blocker, visual loss still progressed to light perception OD and no light perception OS.185 Periocular steroid injections have been met with mixed results. A murine model of CAR treated with subconjunctival triamcinolone showed a trend toward improved survival of outer nuclear layer cell bodies, but ERG amplitudes were not significantly improved.163 Ferreyra et al72 noted a positive response to sub-Tenon methylprednisolone (40e60 mg) injections in four of their CAR patients without significant complications. Responses to treatment consisted of improvement in visual acuity and/or expansion of visual field by at least 25%. Notably, a patient was maintained on bilateral sub-Tenon steroid injections every 2 months as an alternative to systemic immunomodulation with azathioprine and mycophenolate mofetil, which she was unable to tolerate. Huynh et al109 reported a CAR patient presenting with diffuse retinal vasculitis and CME on FA refractory to various systemic interventions, including intravenous methylprednisolone, oral prednisone, mycophenolate mofetil, and IVIg,
434
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
who then received five bilateral intravitreal corticosteroid injections over 2.5 years, resulting in stable visual function until the time of death. They suggested a causal relationship between the injections and resulting visual improvement. Although they acknowledge the initial improvement may have been attributed at least in part to resolution of CME, they note that visual improvement from subsequent injections occurred in the absence of cystic changes on OCT. The authors attributed this positive response to suppressed photoreceptor inflammation, as suggested by restoration of the IS/OS junction and the highly reflective layer on follow-up OCT imaging. Local injections may be an attractive option in select cases of CAR, notably in patients refractory to systemic therapy or intolerant to their associated side effects. Additionally, it remains to be determined whether blockage of VEGF receptor 1 can augment CAR therapy by inhibiting the retinal penetration of autoantibodies.42
2.2.
Melanoma-associated retinopathy
2.2.1.
Clinical presentation
The first report of MAR came from Berson and Lessell29 in 1988, describing a 69-year-old man who developed acute night blindness and hallucinations of shimmering lights 3 years after resection of a cutaneous malignant melanoma. A previous report by Ripps et al236 of a patient who presented with signs and symptoms consistent with MAR attributed the findings to a toxic effect of vincristine, which was part of the individual’s melanoma chemotherapy regimen. By 2001, Keltner et al132 had collected 62 cases, 11 new and 51 previously published. Unlike CAR, which frequently precedes the discovery of systemic malignancy, patients presenting with MAR are far more likely to have a known history of melanoma. In the over 70 reported cases to date, MAR often heralded the presence of non-ocular metastasis.132 All but two cases of MAR had been associated with cutaneous melanomas until Lu et al159 also implicated choroidal and ciliary body melanomas, and even choroidal nevi. Machida et al162 recently presented a patient with MAR-like signs and symptoms who showed no signs of a malignant tumor after skin survey by a dermatologist and whole-body computerized tomography (CT) and positron emission tomography (PET) scans. An intranasal melanoma was discovered 5 months later. This was the first reported case of mucosal melanoma being associated with MAR. Another case of MAR preceded a diagnosis of primary intestinal melanoma by 3 months.231 These occurrences, although rare, highlight the importance of investigating preferential sites of mucosal melanoma (nasal cavity, paranasal sinuses, oropharynx, intestinal tract, vagina, urinary tract) when the primary lesion cannot be identified. Keltner et al132 found a latent period averaging 3.6 years (range, 2 months to 19 years) from diagnosis of the primary neoplasm to the onset of MAR. Affected individuals often present in their 50s, with males being preferentially affected over females by a ratio of 4.7:1, far exceeding the 5:4 sexual predilection of malignant cutaneous melanoma in the United States.49,132 Clinical features of MAR typically reflect rodmediated dysfunction, with patients experiencing sudden onset of bilateral shimmering photopsias and nyctalopia.
Visual loss is painless and typically not as pronounced as in CAR, with 82% (28 of 34 patients) maintaining a visual acuity of 20/60 or better at the time of presentation in Keltner et al’s132 review. A small minority do suffer from central visual deficits, as demonstrated by the remaining six patients who presented with visual acuities of 20/200 or worse in one or both eyes.132 Initial fundus examination, as is the case with CAR, is often normal. In more advanced cases, there may be optic disk pallor, retinal vessel attenuation, and areas of RPE atrophy.132,159 Retinal pigment dispersion is absent to minimal in MAR, a potentially helpful distinction from other causes of pigmentary retinal degenerations such as retinitis pigmentosa (RP). Although less common, there may be a vitreous cellular reaction117,126,132,139,178,226 with vasculitis193,233 and CME. Work-up, in the form of serologies, visual function tests, and various imaging modalities, increases diagnostic confidence. Western blot or ELISA to detect the presence of serum anti-retinal antibodies, or less conventional immunohistochemical staining of donor retina (especially bipolar cells), is confirmatory for MAR. Visual field testing most commonly reveals generalized constriction, but arcuate defects, central, and paracentral scotomas are also described.132 OCT may demonstrate normal retinal architecture and thickness in the early stages of disease, but progressive thinning of the inner retina around the paramacular region can be a later finding.162 The ERG pattern in MAR is classically electronegative, resembling that seen in congenital stationary night blindness (CSNB), central retinal artery occlusion, and juvenile x-linked retinoschisis. A preserved dark-adapted a-wave, indicating normal photoreceptor function, is followed by a markedly attenuated b-wave, reflecting either bipolar cell dysfunction or disruption of synaptic transmission between photoreceptors and the bipolar cells.132
2.2.2.
Pathogenesis
Historically, anti-bipolar cell antibodies have been considered the defining marker of MAR. It is postulated that these autoantibodies directed against the postsynaptic receptors of the depolarizing, or ON, bipolar cells disrupt neural transmission with associated rod photoreceptors, resulting in the predominantly rod-related visual compromise.16 The hyperpolarizing, or OFF, bipolar cells are believed to be spared. Lie et al156 demonstrated that a single intravitreal injection of sera from MAR patients suppresses the photopic and scotopic b-waves in a monkey model. Bipolar cells subserving the ON pathway were preferentially affected in their study. Efforts to elucidate a specific bipolar cell antigen in MAR have revealed a number of photoreceptor proteins, but none specific to the ON bipolar cells. For example, antibodies against a 35-kDa Muller cell protein,76 a 22-kDa neuronal antigen found in both the retina and optic nerve,129 and a novel membrane-associated 33-kDa protein205 have all been described in MAR. Because visual phototransduction proteins are known to be produced by melanoma cells, it should come as no surprise that autoantibodies against transducin-a,2 transducin-b,225 arrestin,27 and rhodopsin98 are found in MAR patients. The wide diversity of retinal antigens encountered is likely indicative of the immunologic heterogeneity of this disorder.
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
More recently, however, Kondo et al146 and Dhingra et al62 independently identified autoantibodies in the sera of four patients with MAR directed against the transient receptor potential cation channel, subfamily M, member 1 (TRPM1), which is specifically expressed in retinal ON bipolar cells. Also known as melastatin 1 (MLSN1), TRPM1 was recently identified as the cation channel that mediates the light response in ON bipolar cells.145,188,258 Interestingly, mutations in TRPM1 have been linked to human CSNB,196,282 and Trpm1 knock-out mice have displayed electronegative ERG patterns.258 These results are an encouraging step towards identifying a MARspecific antigen, but await further confirmation. Conversely, Keltner et al132 reported that although they were able to label retinal bipolar cells with patients’ sera via immunohistochemistry, they could not identify any MARspecific retinal antigens by Western blot technique. Likewise, Milam et al178 were unable to detect a specific antigen responsible for the immunostaining of rod bipolar cells by MAR sera using Western blotting techniques. They hypothesized that this absence may have been secondary to amino group modification by paraformaldehyde fixation, denaturation by sodium dodecyl sulfate, or the MAR-specific antigen being veiled by a nonspecifically stained component it comigrates with.178 Furthermore, the MAR-specific antigen may not even be a protein, but may be a ganglioside, proteoglycan, lipid, carbohydrate, or a combination of these. Histopathological analysis of postmortem retinas from MAR patients has revealed a variety of findings, ranging from unremarkable to widespread retinal degeneration. Okel et al210 initially observed considerable disruption of macular anatomy, with extensive destruction of photoreceptor elements. This was followed by Gittinger and Smith,90 who noted a marked reduction in the density of bipolar neurons in the inner nuclear layer, with preservation of the photoreceptor cell neurons in the outer nuclear layer. Finally, Keltner et al132 found normal retinal architecture by light microscopy, even in the presence of a strong antibody reaction to various retinal layers as detected by indirect immunohistochemistry. The variability of retinal pathology observed may be representative of the different stages of disease from which specimens were obtained.
2.2.3.
Management
Treatment for MAR has been largely ineffective, with successes limited to individual cases. Overall visual prognosis is guarded, and although vision is traditionally thought to remain stable, some cases progressively deteriorate despite treatment with combinations of steroids, plasma exchange, and IVIg.117,132,233,242,283 Treatment is directed at decreasing the melanoma tumor burden with cytoreductive surgery to resect metastases, followed by systemic immunotherapy.132 Seven of Keltner et al’s132 62 patients experienced some type of improvement after these various interventions. Of these, four had undergone cytoreductive surgery, either alone, or in combination with IVIg. Highlighting the importance of adjuvant immunotherapy is a case described by Subhadra et al,270 in which a patient’s visual function continued to deteriorate despite surgical resection of tumor metastases and chemotherapy. Once IVIg was instituted, best-corrected visual acuity and visual fields, but not the ERG, steadily improved over the
435
ensuing year. Yamamoto et al295 observed full recovery of both ERG function and visual fields in a patient with MAR secondary to a cutaneous melanoma treated with cytoreductive surgery and monthly local injections of interferon-b. This appears to be the first case where treatment of MAR resulted in recovery of ERG function. There has been speculation that the autoimmune response underlying the development of MAR is actually protective to the patient, helping to eradicate melanoma cells, prevent tumor spread, and prolong survival.47 Such reports advise caution prior to blunting the immune response with IMT as a treatment for the retinopathy; however, Keltner and colleagues found no difference in survival rates between MAR patients, treated or untreated, and those without MAR.E Pfohler et al218 also reported that anti-retinal antibodies are not always associated with prolonged survival in melanoma patients. As with CAR, the benefit of adjuvant local therapy in MAR has been difficult to assess secondary to the scarcity of reports. Handler et al96 treated a patient with MAR with three cycles of the oral alkylating chemotherapeutic temozolomide, an off-label treatment for metastatic melanoma. In addition, their patient had two bilateral periocular injections of triamcinolone acetonide 40 mg 3 months apart. Visual acuity remained stable at 20/25 in both eyes, and visual field function improved, with a reduction in the size and density of scotomata. Ultimately, a supplemental role may exist for peri- or intraocular corticosteroid injections, especially in cases with associated vitritis, vasculitis, or CME.
2.3.
Paraneoplastic vitelliform maculopathy
2.3.1.
Clinical presentation
A growing number of reports have documented retinopathies of presumed paraneoplastic origin with concurrent vitelliform fundus lesions. To our knowledge, 23 cases of PVM, albeit under different designations, have been reported in the literature (Table 1).14,20,33,35,69,93,120,122,136,147,148,200,214,265,271,301 All but two carried a known diagnosis of malignancy. Until recently, all known cases were associated with either choroidal or cutaneous melanomas, leading many authors to speculate these may be atypical variants of MAR; however, three recent reports demonstrated similar findings in patients with non-melanoma malignancies. The first was in a patient with primary carcinomas of the breast and lung without evidence of systemic melanoma, although the authors entertained the possibility of a simultaneous regressed cutaneous melanoma or undetectable visceral melanoma.93 The second patient had a history of clear cell sarcoma of the right big toe previously in remission, but onset of visual impairment consistent with PVM coincided with detection of metastatic clear cell carcinoma to the lung.271 The third occurrence was in a patient with newly diagnosed metastatic lung adenocarcinoma.14 In patients with a known history of malignancy, diagnosis and treatment of the primary tumor was often remote from onset of PVM. In one case, an eye with a choroidal melanoma had been enucleated 23 years prior to the onset of visual complaints in the remaining eye.301 The presence of PVM appears to correlate with systemic disease burden, as all
436
Table 1 e Clinical characteristics of patients with paraneoplastic vitelliform maculopathy (PVM) Patient Age/ No. Sex
Author, Year
Ref Known No. Malignancy at Time of Diagnosis
Metastases
Yes (brain metastases noted several months later) Yes (mediastinal)
Decreased vision, nyctalopia
OD: 20/400, OS: CF @3ft
Bilateral
Nyctalopia, shimmering lights
20/25
Bilateral
Yes (liver metastases noted 13 months later) Yes (ovarian, lymph nodes)
Blurred vision, nyctalopia
OD: 20/100
Yes (“widespread melanoma” noted 5 months later)
Halos around lights, mild eye pain, difficulty with light-dark adaptation
OD: 20/20
Yes (peritoneal cavity, omental metastases noted 1 month later) Yes (lymph nodes)
Difficulty with light-dark adaptation
20/20
Acute onset bilateral vision loss
OD: 20/125, OS: 20/50
1
47F
Borkowski et al, 2001
35
Yes
Cutaneous melanoma (face)
2
57M
Borkowski et al, 2001
35
Yes
Cutaneous melanoma (scalp)
3
69F
Zacks et al, 2001
301
Yes
Choroidal melanoma OS s/p enucleation
4
33F
Palmowski et al, 2002
214
Yes
Cutaneous melanoma
5
41F
Jampol et al, 2004
120
Yes
Choroidal melanoma OS s/p enucleation
6
41F
Jampol et al, 2004
120
No
7
59M
Sotodeh et al, 2005
265
Yes
e
Cutaneous melanoma (back)
Symptoms
VA at Laterality Presentation
Disabling glare, 20/20 nyctalopia, metamorphopsia
Macular Lesions
Oval-shaped white lesions at the outer retina or RPE
Autoantibodies (antigen)
ERG
EOG
Yes (retinal bipolar cells)
n/a
Scattered, wellcircumscribed chorioretinal atrophic lesions with diffuse loss of RPE Unilateral Two small serous (monocular) retinal detachments
Yes (retinal bipolar cells)
Abnormal n/a
Died 4 years later
Yes (retinal bipolar cells)
Abnormal n/a
Died 2 years later
Bilateral
Yes (retinal bipolar cells)
Abnormal Normal
Alive 2 years later
n/a
n/a
n/a
Died <1 year later
n/a
n/a
n/a
Died 2 months later
Yes (rod outer segment (ROS) protein)
Abnormal n/a
Multiple wellcircumscribed detachments of the RPE, each containing small, yellow-orange lesions Unilateral Serous elevation (monocular) of retina with yellow spot in the fovea, multiple areas of serous retinal detachment Bilateral Multiple yellow serous detachments
Bilateral
Serous retinal detachments with multiple scattered, nummular, yellowish-orange spots at the level of the RPE
n/a
Course
Died 2 months after brain mets noted
Died 1 year later
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
Type
56M
Sotodeh et al, 2005
265
Yes
Choroidal melanoma OD s/p enucleation
Yes (“multiple biopsyproven metastases”)
Acute onset of OS: 20/90 decreased vision, nyctalopia, shimmering colored spots
9
61F
Sotodeh et al, 2005
265
Yes
Cutaneous melanoma (neck)
Acute onset of OD: 20/200 reading OS: 20/125 difficulty, vanishing letters, color dullness
10
62F
200
Yes
11
45M
Nieuwendijk and Hooymans, 2007 Eksandh et al, 2008
69
Yes
12
65M
Javaheri et al, 2008
122
Yes
Choroidal melanoma OD s/p enucleation Choroidal melanoma OS s/p enucleation Mucosal melanoma (small bowel)
Yes (pulmonary metastases noted 1 month later) Yes (liver, lung)
13
58M
Krema et al, 2010
148
Yes
Cutaneous melanoma
14
74M
Bianciotto et al, 2010
33
Yes
Cutaneous melanoma (face, neck)
15
50M
Koreen et al, 2011
147
No
e
Gradual vision loss
Yes (liver, lung)
Fluctuations in visual acuity, nyctalopia Yes (kidneys, Nyctalopia brain, bladder)
Unilateral Numerous small, (monocular) round, curvilinear, yellowish vitelliform lesions arranged in a honeycomb pattern at the level of the RPE with mild serous macular detachment Bilateral Serous retinal detachments with multiple small, round vitelliform lesions
n/a
n/a
n/a
n/a
n/a
Abnormal Died 3 months later
OS: 20/40
Unilateral Multiple yellowish (monocular) areas of serous retinal detachment
Negative
Normal
Normal
OD: 20/160
Unilateral Central serous retinal Yes (bestrophin-1) (monocular) detachment as a “pseudohypopyon” vitelliform lesion Bilateral Multiple, Negative pseudovitelliform lesions varying in size (100e500 microns) at the level of the RPE/ outer retinal junction, coalescing inferiorly, with overlying neurosensory and RPE detachment Bilateral Multiple vitelliform Negative lesions involving the outer retina and RPE
Normal
Abnormal Died 4 months later
20/40
Yes (small bowel metastases noted 2 months later) Yes (liver, lung, brain)
Mild unilateral blurred vision
OD: 20/20, OS: 20/25
Gradual vision loss
OD: 20/70, OS: 20/60
Bilateral
Yes (axillary lymph node, liver metastatic melanoma of unknown primary)
Blurring of central vision
20/40
Bilateral
Serous retinal detachments
Abnormal n/a
Normal
Died 1 year later
Died 2 years later
Died
Abnormal n/a
Yes (interNormal n/a photoreceptor retinoid binding protein (IRBP)) Shallow subretinal fluid Yes (peroxiredoxin Abnormal Abnormal with whitish-yellow 3 (PRDX3)) round lesions in a honeycomb-like pattern at the level of the RPE
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
8
Died 3 months later
(continued on next page)
437
Alive 41 months later, with “excellent treatment response” to oral temozolomide
438
Table 1 e (continued ) Patient Age/ No. Sex
Author, Year
Ref Known No. Malignancy at Time of Diagnosis
Type
Metastases
Symptoms
VA at Laterality Presentation
Macular Lesions
58F
West Coast Retina, 2011
B
Yes
Choroidal Yes (lung) melanoma OS s/p plaque brachytherapy
Seeing bubbles
OD: 20/30, OS: 20/40
Bilateral
17
69F
Grunwald et al, 2011
93
Yes
Breast cancer, Yes (liver) Lung cancer
Progressive blurred vision
OD: 20/80, OS: 20/70
Bilateral
18
73M
Khurana et al, 2011
136
Yes
Cutaneous melanoma
Nyctalopia
20/30
Bilateral
19
46M
Suelves et al, 2012
271
Yes
Clear cell Yes (lung) sarcoma (toe)
Acute onset vision loss
20/20
Bilateral
20
80M
Aronow et al, 2012
20
Yes
Cutaneous melanoma
Yes (lungs, liver, spine)
n/a
OD: 20/30, OS: 20/25
Bilateral
21
79M
Al-Dahmash et al, 2012
14
Yes
Cutaneous melanoma (neck)
OD: 20/50, OS: 20/30
Bilateral
Multifocal serous retinal detachments with yellow-white subretinal fluid
22
60F
Al-Dahmash et al, 2012
14
Yes
Choroidal melanoma OS s/p plaque brachytherapy
Yes Gradual vision (“multiple loss systemic melanoma metastases”) Yes (liver) Gradual vision loss
OD: 20/30, OS: 20/100
Bilateral
Multifocal serous retinal detachments with yellow-white subretinal fluid
Yes (brain, noted 16 months later)
Multifocal, circumscribed, yellow-orange vitelliform lesions with serous retinal detachments Multiple small, round, vitelliform lesions (w500 microns), multiple areas of localized subretinal fluid with debris overlying flat RPE Multiple neurosensory and RPE detachments of varying size (500-5000 microns) with layered fluid-lipid interfaces inferiorly in the larger RPE detachments Multifocal neurosensory detachments, yellow lesions along superior temporal vessels, eventual “pseudohypopyon” formation Multiple nummular, flat, well-demarcated yellow-white lesions
ERG
EOG
Negative
Normal
Normal
n/a
n/a
n/a
n/a
Alive 10 months later
n/a
n/a
n/a
Died 3 years later
n/a
n/a
n/a
Died 6 months later
Yes (carbonic anhydrase II (CAII), and 35-kDa RPE protein) Yes (carbonic anhydrase II (CAII))
Abnormal Abnormal Died 1 month later
Abnormal n/a
Died 7 months later
Yes (68-kDa protein)
Normal
Died 2 months later
n/a
Course
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
16
Autoantibodies (antigen)
M ¼ male; F ¼ female; n/a ¼ not available; VA ¼ visual acuity; OD ¼ right eye; OS ¼ left eye; CF ¼ counting fingers; RPE ¼ retinal pigment epithelium; ERG ¼ electroretinopgrahy; EOG ¼ electrooculography. Comments in quotes represent information as provided by source article. Patient number assigned based on chronological appearance in the literature.
23
74M
Al-Dahmash et al, 2012
14
Yes
Lung adenocarcinoma
Yes (bone)
Painless loss of vision
OD: 20/60, OS: 20/60
Bilateral
Multifocal serous retinal detachments with yellow-white subretinal fluid
n/a
n/a
n/a
Alive 2 months later
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
439
reported cases had evidence of metastatic disease. Furthermore, PVM frequently heralded metastatic spread, often discovered within several months of a patient’s initial ophthalmologic evaluation. The average age of onset is 59 years (range, 33e80 years), with an equal sex distribution.14,20,33,35,69,93,120,122,136,147,148,200, 214,265,271,301 The most common presentation is nyctalopia, although some patients experienced blurry vision, metamorphopsia, shimmering photopsias, glare, and halos. Clinically, PVM may more closely resemble other vitelliform retinopathies, such as acute exudative polymorphous vitelliform maculopathy, or the multifocal variant of Best’s disease, than MAR or CAR. Ophthalmoscopy reveals varying degrees of multifocal yellow-orange vitelliform lesions at the level of the subneurosensory retina and RPE (Fig. 2), often accompanied by overlying serous macular detachment, with a tendency of the vitelliform material to gravitate inferiorly and layer out within the areas of subretinal fluid.14,33,69,93,120,122,136,147,148,200, 214,265,271 Involvement is typically bilateral, and although most cases associated with choroidal melanoma were in monocular patients, two treated with plaque brachytherapy alone had bilateral fundus lesions consistent with PVM.14,B FA typically demonstrates blockage of background choroidal fluorescence by the vitelliform lesions with late staining,14,20,93,122,136,148,200,265 although in some cases the lesions displayed a mild early hyperfluorescence.14,120,147, 214,265,271 There is little, if any, leakage of dye into the corresponding subretinal space. Corresponding fundus autofluorescence shows increased fluorescence of the lesions.20,93,214 OCT reveals multiple areas of localized serous neurosensory detachments containing optically dense debris overlying flat RPE.14,93 The advent of high-definition spectral domain OCT has more accurately localized this accumulation of fluid and vitelliform material to the subretinal space, not beneath the RPE as had been previously suggested, although the RPE may demonstrate thickening (Fig. 2).14,93 The absence of focal choroidal thickening on OCT also helps exclude choroidal metastases. Serological testing has identified autoantibodies directed against bipolar cells,35,214,301 rod outer segment protein (ROS, 120-kDa),265 peroxiredoxin 3 (PRDX3, 26kDa),147 bestropin-1 (68-kDa),69 carbonic anhydrase II (CAII, 30kDa),14,20 interphotoreceptor retinoid binding protein (IRBP, 145-kDa),33 and unnamed 35-kDa20 and 68-kDa14 proteins. Meanwhile, ERG and electrooculography (EOG) evaluation has yielded variable results.
2.3.2.
Pathogenesis
The mechanism underlying the formation of the vitelliform lesions in PVM remains poorly understood; individual reports uncovering the presence of certain anti-retinal and anti-RPE antibodies offers some potentially valuable insights, however. Koreen et al147 demonstrated circulating serum antibodies directed against a 26-kDa RPE protein identified as PRDX3, a mitochondrial peroxidase important in protection against cellular oxidative damage (Table 1, Patient 15). Accordingly, they hypothesized an autoimmune-mediated retinal pigment epitheliopathy resulting in the accumulation of unprocessed cellular debris and possibly lipofuscin, which manifests on a macroscopic level by the accumulation of yellowish subretinal deposits.
440
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
Fig. 2 e PVM in a 58-year-old man with history of metastatic cutaneous melanoma to the axillary lymph nodes who presented with mild blurring of vision in the left eye (20/25) and preserved acuity (20/20) in the right eye. Color fundus photography (A, only left eye pictured) was remarkable for bilateral, multiple, subretinal, vitelliform lesions of the macula. Red-free (B) images of the fundus lesions are also presented. Fluorescein angiography (C ) demonstrated predominant blockage of background fluorescence by the material, which appeared to layer and gravitate into the inferior macula. OCT (D) confirmed these infiltrates to be at the level of the RPE without associated serous fluid. Visual field and ERG were essentially normal, autoantibodies to bipolar cells were not detected, and EOG revealed only a mildly decreased Arden ratio to 1.7. The patient developed metastatic spread to his small bowel 2 months later. Reproduced from Krema et al148 with permission from Wolters Kluwer Health Publishing Group Ltd. Photos courtesy of Peter J. Kertes, MD.
Eksandh et al.69 identified serum anti-RPE autoantibodies against a separate 68-kDa protein, bestrophin-1, in a patient with a history of enucleation for choroidal melanoma who presented with a pseudohypopyon within a serous macular detachment in the remaining eye (Table 1, Patient 11). ERG was within normal limits, whereas EOG showed a pathological Arden ratio of 1.1, similar to what is elicited in Best disease; there was no family history of this and genetic analysis for VMD2 mutation was negative, however, leading the authors to postulate a paraneoplastic etiology for the autoantibodies. Aronow et al20 detected high titers of autoantibodies in a patient with PVM against a 30-kDa protein present in the RPE and retina, determined to be CAII (Table 1, Patient 20). Additionally, they also detected subtle reactivity to an unknown 35-kDa RPE protein. How these contributed to the development of the vitelliform lesions was not clear; although they postulated that by inhibiting the catalytic activity of CAIId which is localized to the outer segment of photoreceptors, inner nuclear layer, and ganglion cell layerdthe intracellular pH levels decrease, with a concurrent rise in intracellular calcium reducing retinal cell viability.8,20 Four other cases had serum autoantibodies that labeled retinal bipolar cells using immunohistochemical techniques
(Table 1, Patients 1e4).35,214,301 Such findings invite further investigation and clarification as to whether PVM falls along the spectrum as a MAR variant or a unique paraneoplastic entity. Adding to the discord are findings from the first histologic report of PVM136 that detailed a patient with bilateral vitelliform macular detachments in the setting of metastatic cutaneous melanoma. Although there was no clinical evidence of choroidal metastasis, postmortem pathologic examination of the eye revealed a flat, pigmented choroidal infiltrate composed of spindle-shaped and epithelioid cells with marked nuclear and cellular pleomorphism. This argues against PVM as a paraneoplastic entity altogether, suggesting instead local metastases with subclinical choroidal involvement. The authors did acknowledge that metastasis may have occurred independently of and well after the development of vitelliform lesions. Aronow et al20 actually described the histopathological correlate to the clinically observed vitelliform lesions. Microscopically, there was focal retinal edema as well as loss of nuclei from the inner retina, likely from bipolar cells. In addition, there was atrophy of the outer nuclear layer and splitting of the outer plexiform layer. In contrast to the previously described case,136 here, there was no evidence of active inflammation or malignant melanoma cells in either enucleated eye.20
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
441
Fig. 3 e DUMP in a 55-year-old woman with 2-year history of metastatic small cell lung carcinoma who presented with decreased visual acuity in the left eye (20/40). She was noted to have moderate nuclear sclerotic cataract in that eye. Color fundus photography (A) revealed islands of atrophic RPE separated by a reticular pattern of yellow-orange pigmentation in the left macula. Corresponding fluorescein angiography (B) was remarkable for nummular areas of early hyperfluorescence consistent with RPE atrophy. B-scan ultrasound (C ) revealed extensive choroidal thickening of the affected left eye (arrowheads). OCT of the same eye (D) demonstrated neurosensory detachment over the macula with focal areas of RPE atrophy and hypertrophy. The respective studies in the right eye were all normal. Reproduced from Reddy and Finger232 with permission from BMJ Publishing Group Ltd. Photos courtesy of Shantan Reddy, MD.
2.3.3.
Management
The presence of vitelliform fundus lesions in the appropriate clinical setting should alert the physician to the possibility of PVM. If there is any history of malignancy, a thorough investigation for systemic spread should be undertaken, regardless of how long the patient has been in remission. Treatment with various combinations of palliative surgical resection, radiotherapy, and chemotherapy is aimed at the underlying malignancy. Unfortunately, the identification of PVM is a grim prognostic indicator, with most patients succumbing to metastatic disease from several months up to 4 years after presentation. One patient had a favorable response to treatment with the chemotherapeutic agent temozolomide (Table 1, Patient 15).147 After initial empiric therapy with prednisone produced no subjective or objective improvement, the patient was diagnosed with a metastatic melanoma of unknown origin and treated with temozolomide therapy, with subsequent resolution of subretinal fluid bilaterally and return of visual acuity to baseline levels. The vitelliform lesions, however, not only persisted, but slightly increased in size. The patient demonstrated an excellent treatment response systemically, with normalized CT imaging, and at the time of publication, was alive and doing well.147
2.4.
Bilateral diffuse uveal melanocytic proliferation
2.4.1.
Clinical presentation
In 1966, Machemer161 first described a patient with presumed pancreatic cancer who developed bilateral visual loss, cataracts, and retinal detachments. Barr et al25 later named the disorder BDUMP in 1982, and at least 50 cases have since been described.22,31,40,59,65,111,142,168,177,187,190,198,201,232,246,256,257,262, 281,292,299 BDUMP usually presents with rapid-onset, bilateral, painless vision loss accompanied by various funduscopic findings. Patients are typically in their 50s through 80s with no sex predilection.201 BDUMP often precedes the diagnosis of the systemic malignancy by months to years. Affiliated tumors typically involve the reproductive tract in women (ovarian, uterine, or cervical cancer), whereas men are likely to have an underlying lung, pancreatic, or colon carcinoma.201 Though bilateral by definition, one unilateral case has been reported (Fig. 3).232 Gass et al85 described the five cardinal signs of BDUMP: 1) multiple, subtle, round, orange-red subretinal patches in the fundus; 2) multifocal early hyperfluorescence of these patches on FA; 3) focally elevated pigmented and non-pigmented uveal melanocytic tumors with diffuse choroidal thickening; 4) exudative retinal detachments; and 5) rapidly progressive
442
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
cataract formation. Other clinical features noted less frequently include iris and ciliary body cysts and angle closure.161,201,227,239,280 Useful adjunct tests include B-scan ultrasonography to demonstrate choroidal thickening and OCT to confirm subretinal fluid. A unique “giraffe pattern” fundus secondary to nummular or polygonal patches of RPE atrophy circumscribed by adjacent orange zones of aggregated and hypertrophied RPE is considered typical for BDUMP.155,292,297 The fundus appearance in these cases is dominated by RPE atrophy, as evidenced by the abundance of round or polygonal window defects seen on FA. Encircling these islands are orange wreaths of intact, hypertrophied RPE, which in turn block background fluorescence on FA. Fundus autofluorescence imaging demonstrates complete loss of the expected RPE autofluorescence pattern within the nummular patches, while showing intensified levels of autofluorescence in the preserved intervening regions.256 OCT likewise reveals alternating zones of irregular RPE thickening and complete RPE loss.31
2.4.2.
Pathogenesis
Through unknown mechanisms, the primary tumor stimulates the proliferation of uveal tract melanocytes. A popular hypothesis centers around the production of melanocytic growth factors by tumor cells with subsequent release into the circulation.201 The observation of hyperpigmentation in extraocular tissue sites such as the skin or mucous membranes in a substantial number of BDUMP patients (26% in one review)201 adds credence to this theory, which is further supported by a case with simultaneous development of systemic hyperpigmentation and BDUMP.86 Circulating anti-retinal autoantibodies have been detected in BDUMP patients,111,177,246 but the significance of the various proteins they are directed against is not known, and may rather represent epiphenomena. Recent studies by Pulido et al,179,C however, show that the serum of some patients with BDUMP cause human melanocytes to proliferate and become disorganized. In addition, this effect was specific for melanocytes and not seen with other tested cell types. The isolated IgG antibody is termed cultured melanocyte elongation and proliferation factor (CMEP). More specific characterization of CMEP and the mechanism whereby which it stimulates rapid melanocytic proliferation is currently under further investigation. In addition to melanocytic proliferation, Gass et al85 hypothesized that toxic or immunologic factors must also participate in the pathogenesis of BDUMP, contributing to the degeneration of the RPE, retina, and lens. Chahud et al46 suggested that ocular hypoxia secondary to the increased metabolic demand from replicating melanocytes creates a toxic milieu conducive to RPE, retina, and lens dysfunction.
2.4.3.
underlying neoplasm.59 Despite occasional transient improvement, most patients are non-responders and continue to deteriorate. Two new reports advocating plasmapheresis appear promising. Based on the premise that a circulating growth factor is responsible for the findings in BDUMP, plasma exchange therapy was initiated by both groups in hopes of removing the inciting agent. Jaben et al111 first used plasma exchange in two women with BDUMP secondary to gynecologic malignancies. The first presented with 3 months of decreased vision after being diagnosed with metastatic papillary serous adenocarcinoma involving the endometrium, bilateral ovaries, and left fallopian tube. Visual acuity at baseline was 20/40 bilaterally (OU), and funduscopic findings were consistent with BDUMP. A course of plasmapheresis was initiated, which consisted of seven volume exchange sessions every other day. During her post-treatment surveillance period, the pigmented fundus lesions remained stable in size, and visual acuity improved. She eventually underwent bilateral cataract extraction, and at follow-up 13 months since completing plasma exchange, her visual acuity was 20/25 OU. The second woman had a 7-month history of painless bilateral vision loss and a known history of metastatic uterine cancer.111 Her visual acuities were 20/200 OD and 20/400 OS at presentation. After BDUMP was diagnosed, she began a course of seven sessions of plasma exchange. Unfortunately, following the fifth session, a line infection necessitated discontinuation of plasmapheresis, and she was lost to follow-up. Nine months later in a telephone conversation, the patient stated that her vision remained stable. Mets et al177 described a patient with metastatic bronchogenic carcinoma who presented with bilateral decreased vision for one month and visual acuities of 20/40 OD and 20/50 OS. A diagnosis of BDUMP was made, and plasmapheresis was initiated three times per week. After 17 treatment sessions, visual symptoms had entirely resolved, and acuity had improved to 20/20 OU. There was corresponding resolution of subretinal fluid and reversal of choroidal thickening. After 7 months, the patient had become too ill to continue plasma exchange. His vision was 20/20 bilaterally at that time. Shortly afterwards, fundus abnormalities reappeared with decline in vision to 20/200 OD and 20/30 OS. He expired one month later. Ultimately, patients with BDUMP often die within a year of their diagnosis secondary to the underlying systemic tumor burden. Despite this poor prognosis, plasmapheresis may be a viable treatment option to improve and maintain visual function while a patient undergoes systemic treatment for the underlying malignancy.
3.
Paraneoplastic optic neuropathies
3.1.
Paraneoplastic optic neuropathy
3.1.1.
Clinical presentation
Management
Treatment for BDUMP has been largely futile, with eventual near complete blindness in most cases. Cataract surgery is frequently performed, but fails to improve visual function to the anticipated degree.201 Other interventions attempted include ocular radiation,25,36,61,65,161,244 drainage of subretinal fluid,237 corticosteroids,73,153,187,194,239,246 and treatment of the
Though less commonly encountered, the optic nerve may be the primary site of paraneoplastic phenomena. Early case reports by Hoogenraad et al108 and Oohira et al211
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
histologically confirmed the presence of optic neuropathy without evidence of tumor invasion. In 1992, Malik et al167 were the first to suggest an immunologic basis for this condition, identifying a serum antibody reactive with neuronal and glial cytoplasm in a 63-year-old man with subacute bilateral visual loss followed by cerebellar ataxia who subsequently was found to have a small-cell lung carcinoma (SCLC). At least 56 cases of PON have been reported, the vast majority associated with an underlying SCLC.32,55,160,170,189,241,259,275,279 Other associations include Bcell lymphoma,54,197,234 pancreatic glucagonoma,154 neuroblastoma,133,254 uterine sarcoma,272 breast,18,228 prostate,44 nasopharyngeal,105 bronchial,220 papillary thyroid,55,151 nonsmall cell lung,21,192 and renal cell carcinomas.55,108,268 Patients classically present with subacute, bilateral, painless vision loss that progresses over days to weeks. The optic nerve head may appear normal, edematous, or atrophic at the time of initial examination. The lack of diagnostic criteria has led to an open and often variable interpretation of what may constitute PON; Cross et al,55 however, described a clinical triad of optic neuritis, retinal vascular leakage, and vitreous cells. FA often displays optic disk hyperfluorescence consistent with leakage (Fig. 4). Peripheral retinal vascular leakage may
443
also be seen. Visual fields show enlarged blind spots, arcuate and altitudinal defects, paracentral scotomas, peripheral constriction, or generalized depression.55 Bitemporal hemianopsia has also been described in a patient with PON primarily affecting the optic chiasm, which was confirmed by postmortem histopathological findings of chiasmal demyelination.211 ERG abnormalities are not consistently noted, but have been described, notably prolongation of the maximal rodecone, cone, and 30-Hz flicker responses.55 Neuroimaging may reveal patchy, hyperintense T2 signal changes in the brain parenchyma, enhancement of the optic nerves, spinal cord abnormalities, or may be unremarkable.55,259 Most of these patients test positive for a serum antibody to the 62-kDa neuronal antigen, collapsin response-mediating protein-5 (CRMP-5), which has also been called CV2.107 The CSF typically reveals a lymphocytic-predominant pleocytosis, elevated protein, oligoclonal bands, and CRMP-5-IgG titers that equal or may significantly exceed serum levels.55,300 Additionally, CSF cytology shows no neoplastic cells. PON almost always coexists in the setting of multifocal neurological dysfunction, explaining why some reports classify the optic neuropathy as a component of paraneoplastic brainstem or cerebellar degeneration.55,300 Several patients have even presented with optic neuropathy and myelopathy
Fig. 4 e PON in a 62-year-old man presenting with subacute progressive bilateral vision loss (20/150 right eye, 20/400 left eye) with associated photopsias. Examination was remarkable for minimal anterior chamber reaction and vitritis bilaterally. Color fundus photography (A, only right eye pictured) was unremarkable, but corresponding fluorescein angiography (B) revealed pronounced papillary and peri-papillary leakage bilaterally. Humphrey visual field (C, D) displayed severely enlarged blind spots bilaterally. MRI of the orbits and brain failed to demonstrate any mass or infiltrative lesions. Chest X-ray did show hilar and paratracheal adenopathy, however, which was subsequently found to be secondary to biopsy-proven small cell adenocarcinoma of the lung.
444
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
strikingly similar to neuromyelitis optica.60,197 In the original publication detailing clinical findings in 116 patients with CRMP-5 seropositivity, Yu et al300 described a myriad of neurologic findings, including peripheral neuropathy (47%), autonomic neuropathy (31%), cerebellar ataxia (26%), subacute dementia (25%), cranial neuropathy (17%, subdivided into 10% loss of olfaction/taste and 7% optic neuropathy), neuromuscular junction disorders (12%), and chorea (11%). The largest cohort to date with PON is Cross et al’s55 16 cases out of 172 patients seropositive for CRMP-5-IgG. Symptomatic vision loss occurred in 15, with visual acuities ranging between 20/20 and 20/400 at presentation. Visual complaints included blurring, dimming, dazzling, or tunnel vision. Fifteeen individuals had disk edema, often with adjacent nerve fiber layer hemorrhages. Interestingly, nine patients had vitreous cells. The authors commented that the cells lacked the characteristic homogeneity, clumping, and larger size commonly seen with intraocular lymphoma. Subsequent vitreous biopsy in four of these patients revealed a pleomorphic reactive lymphocytic response without evidence of lymphoma.
3.1.2.
Pathogenesis
Collapsin response-mediating proteins are believed to regulate growth guidance cues during neurogenesis.48 Specifically, the CRMP-5 subtype is found in normal adult retina, optic nerve, and central and peripheral neurons.55,300 Aberrant expression of this protein is also detected in the cytoplasm of SCLC tumor cells.55,300 The resultant host immune response initiated by neoplastic CRMP-5 is believed to cross-react with native tissue sites, leading to the intra- and extraocular clinical manifestations of PON. Given the widespread production of CRMP-5 throughout the central and peripheral nervous system, it comes as no surprise that these patients present with such extensive neurological dysfunction. Pathologic specimens of the optic nerve show perivascular lymphocytic infiltration in conjunction with axonal demyelination, consistent with chronic optic neuritis.55,108,211,259 Immunohistochemical studies of the infiltrate have revealed an excess of CD8þ T-cells invading the optic nerve.55 Meanwhile, histologic sections from brain and spinal cord displayed widespread microglial activation with rare microglial nodules and prominent perivascular cuffs, again composed mainly of CD8þ T-cells.55
3.1.3.
Management
The most important diagnosis to exclude in the patient with known malignancy whose clinical picture is consistent with PON is metastatic optic nerve infiltration or direct external compression of the nerve. Even more challenging is securing the diagnosis of PON in the absence of any history of cancer. Indeed, a patient presenting with subacute vision loss, accompanying disk edema, and vitreous cells poses a broad differential, including vasculitic, infectious, infiltrative, or demyelinating disease. A thorough history and review of systems, especially for unexplained neurological deterioration, while maintaining a high index of suspicion for potential neoplasia, may avoid a delay in diagnosis.
A CRMP-5 autoantibody assay should be obtained whenever the diagnosis is entertained, and is available through the Mayo Clinic Laboratories by submitting a standard serum specimen.D The presence of CRMP-5-IgG in the serum or cerebrospinal fluid obviates the need for vitreous biopsy and should expedite the search for lung cancer. In rare cases, one must entertain the possibility of an extrapulmonary primary small-cell carcinoma,134 or other malignancies associated with PON as listed previously. Treatment of the primary tumor may stabilize or even improve visual outcomes in some patients,48,241 whereas others progress to severe visual loss.259 Interestingly, the longest published survival duration for PON secondary to SCLC is over 14 years in a patient treated with surgery, chemotherapy, and radiation therapy.160,216 Best-corrected visual acuity was 20/30 OD and 20/40 OS without a relative afferent pupillary defect. Early detection of the systemic malignancy thanks to timely recognition of the paraneoplastic syndrome may have contributed to this favorable outcome. In addition to managing the underlying malignancy, adjunctive systemic immunosuppression is considered a treatment option for PON based on its presumed autoimmune mechanism. Results from individual cases treated with systemic steroids (oral or intravenous), usually given to supplement cancer therapy, have been variable.105,189,279 Intraocular therapy may also be a consideration depending on the severity of vitritis and retinal vascular leakage. Pulido et al228 presented the outcomes in four eyes of two patients with PON treated with intravitreal triamcinolone (IVTA). Both patients were CRMP5-IgG positive in serum and CSF samples and demonstrated vitritis and optic disk edema. Each received 4 mg IVTA with subsequent stabilization or improvement of visual acuity. The first stabilized at the 20/50 level for 3 months, and the second patient improved to 20/200 OD and 20/60 OS from counting fingers OU at baseline by 3 months post-IVTA injection. The optic disk swelling and vitritis resolved in each patient by the final follow-up.
3.2. Polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes syndrome 3.2.1.
Clinical Presentation
POEMS syndrome is a rare paraneoplastic disorder secondary to an underlying plasma cell dyscrasia. In contrast to some of the previous entities, POEMS syndrome manifests with multisystem involvement, targeting numerous tissue sites in addition to the eyes.63,64 Affected individuals typically present in their 50s, with a slight male predominance.63 To date, several hundred cases have been reported; this is still considered to be an underdiagnosed condition, however. The acronym POEMS was initially coined in 1980 by Bardwick et al.23 Although capturing several dominant characteristics of the syndrome, this designation is limited by its exclusion of several others. Furthermore, not all of the components of the acronym are required to establish the diagnosis. The most recently proposed diagnostic classification requires the presence of three major criteria, two of which must include monoclonal plasma cell proliferative disorder and chronic progressive polyneuropathy, and at least one minor criterion (Table 2).64 The clinical picture is typically
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
445
Table 2 e Criteria for the diagnosis of pOEMS syndrome Mandatory Major Criteria Other Major Criteria (1 required)
Minor Criteria (1 required)
Other Signs and Symptoms
1. Polyneuropathy (typically demyelinating) 2. Monoclonal plasma cell-proliferative disorder 1. VEGF elevation 2. Castleman disease 3. Sclerotic bone lesions 1. Organomegaly (splenomegaly, hepatomegaly, lymphadenopathy) 2. Extravascular volume overload (edema, pleural effusion, ascites) 3. Endocrinopathy (adrenal, pituitary, gonadal, parathyroid) 4. Skin changes (hyperpigmentation, hypertrichosis, glomeruloid hemangiomata, plethora) 5. Papilledema 6. Thrombocytosis/polycythemia Clubbing, weight loss, hyperhidrosis, pulmonary hypertension/restrictive lung disease, thrombotic diatheses, diarrhea, low vitamin B12 values
The diagnosis of POEMS syndrome is confirmed when both of the mandatory major criteria, one of the three other major criteria, and one of the six minor criteria are present. Reproduced from Dispenzieri64 with permission from John Wiley and Sons.
dominated by peripheral neuropathy, which is initially sensory, but progresses to include the motor system.127 Involvement is bilateral, symmetric, and spreads proximally, resulting in profound muscle atrophy and weakness.63 Intraocular manifestations of POEMS syndrome, limited to single case reports and small case series, are nevertheless well documented, as evidence by the inclusion of “papilledema” among the minor diagnostic criteria (Table 2).64 The most common ocular finding in POEMS syndrome is bilateral optic disk edema (ODE), occurring in 29e73% of patients.34,38,45,52 Kaushik et al,125 in the largest study to date, found ODE in 17 of 33 patients (52%). Of note, five (29%) of the patients had asymptomatic ODE at the time of initial examination, leading the authors to advocate screening ophthalmologic examinations for all patients diagnosed with POEMS.125 When presenting with symptomatic ODE, patients often complained of blurry vision, ocular pain, or diplopia. In addition to ODE, there are at least five cases with associated CME52,94,110,290 and one of bilateral serous macular detachment.209 Adjunctive testing can aid in the evaluation of these patients and augment physical examination findings. Visual field testing usually demonstrates enlargement of the blind spot,125,291 although arcuate defects are also described.24,52,94 FA typically shows late optic disk leakage and may also demonstrate petaloid leakage consistent with CME. OCT may also confirm the presence of CME or macular detachment. Additionally, serological testing reveals significantly elevated VEGF levels, sometimes to over 2,000 pg/ml (normal range, 62e707 pg/ml).110,266,286 Watanabe et al287 found a mean serum VEGF level of 1,673.2 pg/ml in 10 patients with POEMS syndrome, as compared with <131 pg/ml in control subjects. Increased production of the associated cytokines interleukin6 (IL-6), IL-1 beta (IL-1b), and tumor necrosis factor-alpha (TNF-a) may also be detected.87,88,240
3.2.2.
Pathogenesis
Although significant progress has been made in our understanding of POEMS syndrome, its pathogenesis remains complex. Elevated production of proangiogenic and proinflammatory cytokines are believed to be the hallmark of this disorder. Specifically, excess VEGF produced by the abnormal
plasma cells is thought to be the major contributor to its development and correlates best with disease activity.64,180,252 VEGF is known to target endothelial cells, where it induces a rapid increase in vascular permeability while also being an important promoter of angiogenesis. As previously stated, patients with POEMS syndrome have also demonstrated higher circulating levels of IL-6, IL-1 b, and TNF-a, and both IL6 and IL-1b are known to stimulate VEGF production.88,266 The etiologies of ODE, CME, and macular detachments associated with POEMS syndrome are controversial. Current hypotheses include elevated intracranial pressure,56 vasculitis,181 infiltration of the optic nerve,37 increased VEGF levels,52 or any combination thereof. Increased vascular permeability of the optic nerve and macula secondary to raised serum VEGF levels is an appealing theory and is supported by the presence of extravascular volume overload elsewhere in POEMS syndrome (e.g., peripheral extremity edema, pleural effusion, pericardial effusion, ascites).52 Alternatively, the development of CME and macular detachment may be caused by an osmotic gradient created by deposition of high molecular weight immunoglobulins within and beneath the neurosensory retina. This pathologic mechanism has been similarly offered to explain the macular detachments observed in some patients with multiple myeloma and Waldenstro¨m macroglobulinemia.30,39,79,104,221
3.2.3.
Management
There are no randomized controlled trials evaluating the treatment of POEMS. Although directing therapy against the increased VEGF load seems logical, use of intravenous bevacizumab has yielded mixed results.123,249,269 Rather, the majority of successful outcomes are the result of targeting the underlying plasma cell dyscrasia itself. Treatment of the systemic disease process is beyond the scope of this discussion, but may include radiation therapy for localized bone lesions, high-dose chemotherapy with autologous stem cell transplantation, and systemic steroids.63,64 Overall, ODE and associated ocular symptoms respond variably to systemic treatment. Wiaux et al290 observed resolution of macular edema in two patients upon institution of systemic steroids in combination with oral acetazolamide, but
446
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
disk edema persisted in both patients. Okada et al209 reported a patient with bilateral macular detachment who was treated with autologous peripheral blood stem cell transplantation. Serum VEGF levels subsequently declined from >2,000 pg/ml to 221 pg/ml with resolution of the subretinal fluid and improvement of visual acuity. Imai et al110 injected sub-Tenon triamcinolone acetonide twice to treat CME in the more affected eye of their patient. Because of persistent CME, the eye underwent pars plana vitrectomy and additional intravitreal triamcinolone injection with a resulting decrease in macular thickness, but visual acuity remained poor. Of note, vitreous levels of VEGF obtained at the time of vitrectomy were <31 pg/ml, in stark contrast to serum levels, which consistently exceeded 2,000 pg/ml. The fellow eye was observed throughout the follow-up period, and macular thickness paralleled changes with serum VEGF levels. They concluded that the CME was a response to elevated VEGF in the circulation and not that locally produced by the retina. That being said, there are no reports to date evaluating the response to intravitreal anti-VEGF agents. Such a treatment strategy is appealing and could warrant consideration in the appropriate clinical scenario to augment systemic therapy.
4. Systemic evaluation in the patient without known malignancy The presence of a clinically suspected paraneoplastic syndrome in conjunction with autoantibodies, particularly against recoverin or CRMP-5, mandates a thorough systemic evaluation for cancer in collaboration with the primary care provider. In addition to a comprehensive general physical examination, this may also entail dermatologic skin survey, colonoscopy, prostate screening, and/or gynecologic exam as dictated by the individual case.213 Imaging studies, at bare minimum, should include a chest X-ray and CT scan of the chest as starting points. Some experts recommend CT and PET scanning of the whole body (usually orbits to thighs).58,176 In selected patients known to be seropositive for a paraneoplastic antibody, PET may be more sensitive than CT158,285,298; by doing both PET and CT, however, sensitivity is higher than with either alone.158 McKeon et al,176 in 56 consecutive patients with clinical suspicion for paraneoplastic retinopathy, but negative CT evaluation, found that 22 patients (39%) demonstrated abnormalities suggestive of cancer by PET, and 10 patients (18%) later had this confirmed histologically. In many centers, a CT scan is performed along with the PET scan (PET-CT)58 to enhance accuracy and localization so that metabolic PET abnormalities are correlated with anatomic CT abnormalities.176 However, because the CT portion of the PET-CT generally uses a lower dose of radiation, thicker sections, and no contrast, it is less sensitive than a dedicated CT. Therefore, a negative PET would still require a dedicated body CT, and conversely, a negative body CT would require a PET scan.58 Important sources of false-positive PET uptake include inflammatory lesions, trauma, benign tumors, and stray tracer that has infiltrated outside of the injection site and is picked up by an axillary lymph node.58 Sources of a false-
negative PET scan may include a tumor with low metabolic activity, as well as tumors smaller than 1 cm.50 In patients with autoantibodies predictive for SCLC, such as CRMP-5 in cases of suspected PON, more detailed thoracic imaging may be warranted when initial CT findings have been negative. This tumor is typically limited in its spread and thus more difficult to detect. Cases of PON with negative CT survey have subsequently demonstrated a tumor focus utilizing chest magnetic resonance imaging (MRI)141; MRI is generally not used as a primary oncologic screening tool, however.176 If no cancer is found, imaging may be repeated at future intervals.260 A report issued by the European Federation of Neurologic Societies task force on management of paraneoplastic neurologic syndromes recommends, in general, repeat surveillance every 6 months for 4 years.284 Darnell and Posner58 recommend closer follow-up examinations and studies at least initially (every 3 months, and then progress to every 6 months).
5.
Non-paraneoplastic retinopathies
The paraneoplastic syndromes, primarily CAR and MAR, must be differentiated from other mimicking retinal degenerations that may present with similar symptoms and signs: RP, cone dystrophy, AZOOR, and ARRON. Two of these entities, AZOOR and ARRON, warrant careful consideration.
5.1.
Acute zonal occult outer retinopathy
Originally described by Gass82 in 1992, AZOOR is an idiopathic retinal disease characterized by the acute onset of sectoral outer retinal dysfunction at the level of the photoreceptors.116,267 Like the paraneoplastic retinopathies, AZOOR is rare but increasingly recognized, with over 150 cases reported to date.186 Overlapping features among these entities can be cause for confusion. AZOOR typically affects young (average age, 36.7 years) white (89% of patients) females (3.2:1 ratio over males).186 In contrast, paraneoplastic retinopathies most commonly present during the sixth to seventh decades of life, show no racial preference, and, with the exception of MAR, which affects males more than females, have no sex predilection. Additionally, patients with AZOOR are more likely to have a history of antecedent viral-like illness (11e20%), and may have an associated systemic autoimmune disease (18e28%).84,186 Gass observed that AZOOR presents acutely as a scotoma accompanied by photopsias.82 In fact, visual acuity is minimally affected in AZOOR (20/40 or better in 74% of eyes),186 similar to MAR, and in stark contrast to CAR. Furthermore, whereas paraneoplastic retinopathies are classically bilateral at onset, AZOOR more commonly presents unilaterally (62% of patients).186 Fundus examination on presentation is usually unremarkable in AZOOR, MAR, and CAR. Over time in AZOOR, RPE mottling analogous to retinitis pigmentosa may develop in the zones of visual field loss. Retinal vascular abnormalities, however, are rare in AZOOR,186 unlike the vascular
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
attenuation in both RP and paraneoplastic retinopathy. In contrast to RP, intra-retinal bone spicule migration is rare with either AZOOR or paraneoplastic retinopathy; meanwhile, the presence of CME is more typical of RP and paraneoplastic retinopathy than AZOOR. Ancillary testing with ERG can be invaluable in differentiating among AZOOR and paraneoplastic degenerations. In AZOOR, the ERG is abnormal in the vast majority of cases. Despite the focal nature of AZOOR clinically, the ERG changes depict a more global retinal dysfunction.186 In a long-term follow-up study in 90 eyes of 51 patients with AZOOR, Gass et al84 detected abnormal scotopic and photopic ERGs in 55 eyes, photopic only in 16 eyes, and scotopic only in 8 eyes. The most consistent abnormality noted by Francis et al78 in a series of 28 patients is a markedly delayed implicit time of the 30-Hz cone flicker response. Moderate reductions in a- and b-wave amplitudes are also common, but not to the extent of the severely depressed to extinguished waveforms described in CAR. Furthermore, the electronegative ERG pattern of MAR is not usually observed with AZOOR, although Piao et al219 described one such case. Anti-retinal antibody testing may not be as valuable a distinguishing factor between AZOOR and paraneoplastic retinopathy as previously thought. Adamus3 reported that, of the 53 patients suspected of having AZOOR that underwent serological testing, more than half (53%) had circulating autoantibodies to various retinal proteins. Autoantibodies to a-enolase and CAII, which are often associated with paraneoplastic retinopathies, were detected in 14 (27%) and 9 (17%) of AZOOR patients. In 25 patients with AZOOR, Heckenlively and Wang99 likewise detected strong immunoreactivity to anti-retinal antibody bands by Western blot. Other reported retinal antigens include cellular retinaldehyde binding protein (CRALBP) and retinal soluble antigen (S-Ag) in one case by Neutzner et al199 and an unnamed 45-kDa antigen in one of four patients presented by Mkrtchyan et al.184 The term AZOOR complex has been used to describe a group of diseases including punctate inner choroidopathy, multifocal choroiditis and panuveitis, multiple evanescent white dot syndrome, acute idiopathic blind spot enlargement, and acute macular neuroretinopathy that share overlapping demographic and clinical features with AZOOR and may all have a common autoimmune mechanism.83,92,118,121 Fine et al74 described three cases of multiple evanescent white dot syndrome that atypically developed progressive multifocal zonal degeneration of the outer retina and RPE characteristic of AZOOR. Similarly, Holz et al106 identified seven patients with punctate inner choroidopathy who demonstrated widespread retinal dysfunction in excess of what would be expected from the visible fundus findings that they attributed to AZOOR. Some experts feel that AZOOR is a unique entity that may be more precisely defined by multimodal imaging: 1) autofluorescence, demonstrating characteristic zonal degeneration of the outer retina and RPE, typically beginning around the nerve and progressing to the periphery, and 2) high-definition spectral domain OCT, showing zonal or annular loss of the photoreceptor layer and IS/OS complex.81,143,184,199,263,267
447
5.2. Autoimmune-related retinopathy and optic neuropathy A patient’s clinical, electrophysiological, and immunological profile may be suggestive of paraneoplastic retinopathy, although the systemic work-up is unremarkable for malignancy (Fig. 5). The appropriate classification for this situation has been the subject of much confusion and discord, with different designations being adopted by different groups. Terms such as non-paraneoplastic autoimmune retinopathy, recoverin-associated retinopathy, and autoimmune-related retinopathy and optic neuropathy (ARRON) are variably used to describe this subgroup. Historically, the earliest reports of this condition described patients with a steroid-responsive optic neuropathy.66,80,112,150,235,243,E Although laboratory techniques at the time did not allow detection of autoantibodies, these cases were felt to represent a localized autoimmune-mediated optic neuropathy. It has since become apparent, however, that many patients with steroidresponsive optic neuropathy additionally manifested a retinal immune component. KeltnerE offered a rationale for the use of ARRON: 1) “autoimmune-related,” rather than “autoimmune” reflects the uncertainty whether the antibodies generated from the retina and optic nerve are directly responsible for the vision loss or just epiphenomena; and 2) the addition of “optic neuropathy” encompasses the full disease spectrum, ranging from cases of isolated retinal involvement, to isolated optic nerve involvement, to simultaneous retinal and optic nerve involvement. In Keltner et al’s130 initial report detailing ARRON in 12 patients, the average age of symptom onset was 50 years (range, 37e75 years), and women were preferentially affected (2:1 over men). Vision loss was painless, asymmetric at onset, and varied from 20/20 to no light perception. Only 3 of 12 patients experienced photopsias. Nonspecific retinal changes were present in 8 of 12 patients, optic disk pallor was noted in 11 of 12, and ERG abnormalities were present in 10 of 11. The presence of CME may be a distinctive feature of this retinopathy.72,103 Reported antigenic targets in presumed cases of have included a 22-kDa neuronal antigen found in retina and optic nerve,129 a 35-kDa component of Mu¨ller cells,76,217 and even recoverin in a single case.289 Adamus et al12 observed antiretinal antibodies in 41% of patients (58 of 141) with a clinical presentation resembling ARRON, most commonly directed against a-enolase and a 35-kDa protein unidentified at the time. In the same report, they were able to identify antirecoverin antibodies only in patients with paraneoplastic retinopathy. Patients with ARRON may have been previously diagnosed with simplex RP; in the absence of any family history, however, investigators alternatively proposed an autoimmune etiology for the retinal degeneration. Attempting to show causality, Heckenlively et al102 detected anti-retinal antibodies by Western blot in 90% of patients with RP and CME (27 of 30), compared with only 13% of patients with RP and no CME (3 of 30) and 6% of controls (3 of 50). Complicating matters further, a follow-up study by Heckenlively et al100 analyzing sera from 521 patients diagnosed with RP found
448
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
Fig. 5 e ARRON in a 53-year-old woman referred for rapidly declining vision in both eyes. Color fundus photography (A, only left eye pictured) revealed marked attenuation of the arterioles, and diffuse RPE changes bilaterally. Autofluorescence imaging (B) showed patchy areas of absent autofluorescence suggestive of RPE atrophy, which was supported by the hyperfluorescent window defects seen on fluorescein angiography (C ). Spectral domain OCT imaging (D) displayed CME bilaterally, which did not leak on fluorescein angiogram. Visual fields were severely constricted, and ERG was extinguished. Anti-retinal antibody testing was positive for antibodies to alpha-enolase; a comprehensive systemic workup for malignancy was negative, however. Oral acetazolamide therapy resulted in limited improvement of the CME.
anti-retinal antibodies in 51 and specifically, anti-recoverin antibodies in 10 patients in the absence of underlying malignancy. In an editorial response, Adamus1 highlighted that the patients in the study often demonstrated nonspecific antiretinal antibodies recognizing multiple retinal proteins, often up to 10, on the Western blots. Thus, in the absence of measuring titers to demonstrate higher intensity or quantity of one antibody over others, it would be impractical to assign such a high importance to the presence of any one particular antibody, let alone anti-recoverin antibodies.
Although not a suggested diagnostic criteria for ARRON (Table 3), 8 of 12 patients in Keltner et al’s130 cohort had concurrent systemic autoimmune diseases, a finding corroborated by other reports.101,183,296 Ferreyra et al72 were also able to elicit a strong family history of autoimmune disease, affecting the patient and at least one first-degree relative in 67% of cases (16 of 24). This may explain these patients’ susceptibility to developing retinal disease, and theoretically, justify treatment with steroids or other immunomodulatory therapy. Results of such interventions have been mixed and
Table 3 e Suggested criteria for the diagnosis of ARRON All four of the following
One of the following Modifiers
1. Visual loss as demonstrated either by visual acuity or visual field examination 2. No malignancy found after extensive evaluationa 3. Evidence of optic nerve or retinal abnormalities 4. No identifiable cause for optic neuropathy and/or retinopathy 1. Serum autoantibodies against retina and/or optic nerve not usually found in normal healthy individualsb 2. Response to immunomodulation demonstrated either by stabilization, slowing, or reversal of visual field deficit Type A: Associated with other autoimmune disease Type B: Not associated with other autoimmune disease
a Includes a history of remote malignancy that may better explain visual loss as being associated with CAR, PON, or MAR. b The presence of autoantibodies to retina and/or optic nerve antigens does not prove causality. Reproduced from Oyama et al213 with permission from Wolters Kluwer Health Publishing Group Ltd.
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
have generated more controversy than definitive answers.72,119 Generally speaking, if a coexisting autoimmune disease is present, it should be treated first. In the absence of systemic disease, corticosteroids are initially attempted, and depending on the response, additional aggressive immunotherapies may be used singly or in combination. Oyama et al213 more recently reported a case of ARRON in a patient resistant to conventional therapies who underwent successful autologous hematopoietic stem cell transplantation with improvement in symptoms, visual acuity, and visual fields. Whether these results can be duplicated remains to be seen. Equally important as choice of therapy is counseling to help the patient understand the goals of therapy. Recovery of visual function already lost may not occur. Rather, stabilization of vision and prevention of further deterioration may be the best that can be hoped for.E Ultimately, ARRON is a diagnosis of exclusion that is made following an extensive systemic evaluationdand often reevaluation at future intervalsdthat rules out an underlying malignancy (see section IV). Unexplained vision loss should always lead to consideration of an underlying paraneoplastic syndrome. The longest known latency period between onset of retinopathy and discovery of systemic malignancy is 11 years, in a patient with bronchioloalveolar carcinoma and associated circulating anti-recoverin antibodies.245 That case highlights the importance of diligent regular screenings in patients with presumed ARRON, as at the time of tumor diagnosis, no metastases had occurred, and the patient did well after lobectomy. Conversely, a patient with ARRON has gone 32 years without detectable malignancy (personal communication with John Keltner). Whether ARRON stands as an independent entity or is a prelude to a malignancy raises an intriguing issue concerning the practicality of serological tests and their overall contribution to the physician’s acumen.
6.
Autoantibody testing
Laboratory techniques for detecting circulating anti-retinal antibodies in patient sera include Western blot, ELISA, and immunohistochemistry, all which have limitations. In order to ensure both accurate and reproducible assays for antiretinal antibodies, Foorooghian et al77 stressed that rigorous control measures must be instituted. Furthermore, although all three laboratory assays require a scaffolding of protein/ tissue on which to detect anti-retinal antibodies, the source of that protein/tissue varies in different laboratories. Fresh human tissue obtained within 6 hours after death is optimal, because the ensuing degradation of protein and tissue may contribute to erroneous results. Whereas some investigators have not had difficulty obtaining fresh human tissue, others have, and instead utilized porcine,E rodent,51,169 monkey,289 primate,157 and bovine6,102 tissues with success. Recent progress towards streamlining serological testing for paraneoplastic and non-paraneoplastic retinopathies include the introduction of a commercially available anti-recoverin assay through Athena DiagnosticsF as well as more extensive autoantibody testing offered at several academic institutions (University of California at Davis Ophthalmology Research
449
Laboratories and Casey Eye Institute’s Ocular Immunology Lab). The presence of autoantibodies directed at recoverin should be considered highly indicative of a concurrent malignancy. Of the nearly 3,000 serum samples of patients with retinopathy analyzed at the University of California at Davis, only cancer-related cases had anti-recoverin antibodies.1 Likewise, Adamus et al1,2,12 have found anti-recoverin autoantibodies associated with cancer in almost all of their cases. Conversely, numerous authorities have advised against implying causation with any antigen other than recoverin, bringing a-enolase to the forefront of this discussion. Studies by Adamus et al2,12 have found anti-enolase antibodies to be far less predictive of an associated neoplasm. In fact, in approximately 60% of cases, no neoplasm can be found on extensive evaluation.2,12,288 The clinical utility of this antigen is further complicated by reports associating serum antienolase with numerous autoimmune diseases: autoimmune polyglandular syndrome type 1 (80%, 35 of 44 patients), primary (69%, 60 of 87) and secondary (58%, 14 of 24) membranous nephropathy, autoimmune hepatitis type 1 (60%, 12 of 20), mixed cryoglobulinemia with renal involvement (67%, 7 of 11), and endometriosis (50%, 21 of 41).89 Autoantibodies to enolase have also been noted in up to 12% of otherwise healthy individuals,89 although these antibodies in normal individuals may target a different epitope compared with diseased patients.5,288 Beyond their diagnostic value, the measurement of antiretinal antibodies has been proposed as a useful marker in monitoring the therapeutic response of a patient with these syndromes. Although careful monitoring of clinical parameters (including visual acuity, visual fields, color vision, ERG, OCT, and sometimes visual evoked potentials) remains key to assessing treatment response, anti-retinal antibody titers may be a clinically relevant surrogate marker of disease activity. Numerous investigators have documented a decline in anti-retinal antibody titers in response to their therapeutic intervention.131,195,276 With this increased accessibility to autoantibody testing, clinicians are strongly encouraged to interpret such test results with caution. Integration of these serological findings with the clinical presentation, rather than a reliance upon them, is paramount in order to make the appropriate diagnosis. Demonstration of anti-retinal antibodies, while necessary, is not sufficient alone for diagnosing paraneoplastic retinopathies, since such antibodies have been previously described in normal human sera as detailed by Shimazaki et al.261 In their study, 62% (57 of 92) of normal serum samples had detectable anti-retinal antibody activity by Western blot.261 In a separate study, Bazhin et al28 observed anti-recoverin seropositivity in 15% (15 of 99) and 20% (9 of 44) of patients with small cell and non-small cell lung cancers, respectively. None of the cumulative 143 patients developed visual compromise concerning for CAR, however. In a follow-up study by the same group,250 five patients with non-lung cancers (breast, ovary, stomach, and bladder) and anti-recoverin antibodies present in their sera were subject to ophthalmologic examination. Only one individual had narrowing of the visual field and corresponding
450
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
retinal degeneration consistent with a diagnosis of CAR. Similarly, Ladewig et al152 found that out of 77 serum samples from 51 patients with melanoma but no signs or symptoms of MAR, 69% (53 of 77) were found to contain antibodies reactive with different components of the retina by indirect immunohistochemical assay. Of the 49 patients followed over 4 years since the initial report, only one individual developed MAR.E Despite numerous advancements and ongoing research efforts in the field, our overall understanding of these syndromes on an immunologic and likely genetic basis remains in its infancy, as suggested by Foorooghian.77 What determines why some cancer-bearing patients mount an immune response to tumor-derived antigens and others don’t? Of the subset of patients that do express anti-retinal antibodies, why do some never manifest any signs or symptoms of visual compromise? Is there a predetermined genetic susceptibility that is acted upon by an environmental “trigger,” as has been hypothesized for the development of other types of autoimmune disease? These, and many more questions remain to be answered. Moving forward, newer technologies such as the Luminex analyzer (Luminex Corp., Austin, TX) may enable screening for antibodies against up to 30 antigens in over 800 human serum samples, ideal for large, screening seroepidemiologic studies.26 However, given the increasing number of retinal antigens being reported and their seemingly ubiquitous nature, Jampol119 correctly questions just how many of these anti-retinal antibodies are merely epiphenomena, coincidentally present yet not causally related to the disease pathogenesis. Our challenge moving forward will be to differentiate the pathogenic and disease causing antibodies from the benign “innocent bystanders” if we hope to improve our diagnostic yield and add clarity to an elusive group of diseases that are challenging to both diagnose and treat.
7.
Conclusion
The paraneoplastic retinopathies and optic neuropathies are a heterogenous group of disorders with numerous overlapping features. Clinically, these entities remain among the most challenging to diagnose, even for the most experienced ophthalmologist. Patients may present with no known oncologic history, and ophthalmologic exam may offer no strong clues to alert the investigator to the presence of an underlying malignancy, let alone any pathology at all. With no clear diagnostic criteria to refer to, recognition of one of these syndromes instead requires an integration of clinical intuition, examination findings when present, and results from ancillary testing. Securing the diagnosis in a timely fashion can facilitate systemic treatment of the primary malignancy at an earlier stage, often months before it would have otherwise presented, and gives the patient an increased chance at survival. As we continue to gain insight into pathogenic mechanisms, we will likely find these disorders to be more similar than different. Along with this knowledge will hopefully come improved diagnostic measures as well as more effective, targeted ocular therapies to help preserve, or even restore vision.
8.
Method of literature search
An electronic search of the literature was performed using Medline and PubMed (up to August 2012). Key search terms were cancer-associated retinopathy, CAR, melanoma-associated retinopathy, MAR, paraneoplastic retinopathy, autoimmune retinopathy, autoimmune-related retinopathy and optic neuropathy, ARRON, acute zonal occult outer retinopathy, AZOOR, paraneoplastic vitelliform maculopathy, paraneoplastic vitelliform retinopathy, bilateral diffuse uveal melanocytic proliferation, BDUMP, paraneoplastic optic neuropathy, PON, polyneuropathy organomegaly endocrinopathy M protein and skin changes, POEMS. Citations within material obtained from the Medline and PubMed searches were also used.
9.
Disclosure
The authors report no proprietary or commercial interest in any product mentioned or concept discussed in this article. Supported by a grant from the Karl Kirchgessner Foundation at the Jules Stein Eye Institute.
references
1. Adamus G. Antirecoverin antibodies and autoimmune retinopathy. Arch Ophthalmol. 2000;118:1577e8 2. Adamus G. Autoantibody targets and their cancer relationship in the pathogenicity of paraneoplastic retinopathy. Autoimmun Rev. 2009;8:410e4 3. Adamus G. Is zonal occult outer retinopathy an autoimmune disease? J Clinic Experiment Ophthalmol. 2011;2:104e 4. Adamus G, Amundson D, MacKay C, et al. Long-term persistence of antirecoverin antibodies in endometrial cancer-associated retinopathy. Arch Ophthalmol. 1998;116:251e3 5. Adamus G, Amundson D, Seigel GM, et al. Anti-enolasealpha autoantibodies in cancer-associated retinopathy: epitope mapping and cytotoxicity on retinal cells. J Autoimmun. 1998;11:671e7 6. Adamus G, Aptsiauri N, Guy J, et al. The occurrence of serum autoantibodies against enolase in cancer-associated retinopathy. Clin Immunol Immunopathol. 1996;78:120e9 7. Adamus G, Brown L, Weleber RG. Molecular biomarkers for autoimmune retinopathies: significance of anti-transducinalpha autoantibodies. Exp Mol Pathol. 2009;87:195e203 8. Adamus G, Karren L. Autoimmunity against carbonic anhydrase II affects retinal cell functions in autoimmune retinopathy. J Autoimmun. 2009;32:133e9 9. Adamus G, Machnicki M, Elerding H, et al. Antibodies to recoverin induce apoptosis of photoreceptor and bipolar cells in vivo. J Autoimmun. 1998;11:523e33 10. Adamus G, Machnicki M, Seigel GM. Apoptotic retinal cell death induced by antirecoverin autoantibodies of cancerassociated retinopathy. Invest Ophthalmol Vis Sci. 1997;38:283e91 11. Adamus G, Ortega H, Witkowska D, et al. Recoverin: a potent uveitogen for the induction of photoreceptor degeneration in Lewis rats. Exp Eye Res. 1994;59:447e55 12. Adamus G, Ren G, Weleber RG. Autoantibodies against retinal proteins in paraneoplastic and autoimmune retinopathy. BMC Ophthalmol. 2004;4:5
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
13. Adamus G, Webb S, Shiraga S, et al. Anti-recoverin antibodies induce an increase in intracellular calcium, leading to apoptosis in retinal cells. J Autoimmun. 2006;26:146e53 14. Al-Dahmash SA, Shields CL, Bianciotto CG, et al. Acute Exudative Paraneoplastic Polymorphous Vitelliform Maculopathy in Five Cases. Ophthalmic Surg Lasers Imaging. 2012;43:366e73 15. Albert LJ, Inman RD. Molecular mimicry and autoimmunity. N Engl J Med. 1999;341:2068e74 16. Alexander KR, Fishman GA, Peachey NS, et al. ’On’ response defect in paraneoplastic night blindness with cutaneous malignant melanoma. Invest Ophthalmol Vis Sci. 1992;33:477e83 17. Anastasakis A, Dick AD, Damato EM, et al. Cancer-associated retinopathy presenting as retinal vasculitis with a negative ERG suggestive of on-bipolar cell pathway dysfunction. Doc Ophthalmol. 2011;123:59e63 18. Antoine JC, Honnorat J, Vocanson C, et al. Posterior uveitis, paraneoplastic encephalomyelitis and auto-antibodies reacting with developmental protein of brain and retina. J Neurol Sci. 1993;117:215e23 19. Arnold AC, Lee AG. Systemic disease and neuroophthalmology: annual update 2000 (Part I). J Neuroophthalmol. 2001;21:46e61 20. Aronow ME, Adamus G, Abu-Asab M, et al. Paraneoplastic vitelliform retinopathy: clinicopathologic correlation and review of the literature. Surv Ophthalmol. 2012;57:558e64 21. Asproudis IC, Nikas AN, Psilas KG. Paraneoplastic optic neuropathy in a patient with a non-small cell lung carcinoma: a case report. Eur J Ophthalmol. 2005;15:420e3 22. Bahar I, Weinherger D, Kremer MR, et al. [Ocular manifestation of bronchogenic carcinoma: simultaneous occurrence of diffuse uveal melanocytic proliferation and uveal metastases]. Harefuah. 2007;146:2e3, 80. 23. Bardwick PA, Zvaifler NJ, Gill GN, et al. Plasma cell dyscrasia with polyneuropathy, organomegaly, endocrinopathy, M protein, and skin changes: the POEMS syndrome. Report on two cases and a review of the literature. Medicine (Baltimore). 1980;59:311e22 24. Barnes C, Sanchez-Dalmau BF, Vela D, et al. [Papilledema as an indicator of POEMS syndrome]. Arch Soc Esp Oftalmol. 2008;83:709e11 25. Barr CC, Zimmerman LE, Curtin VT, et al. Bilateral diffuse melanocytic uveal tumors associated with systemic malignant neoplasms. A recently recognized syndrome. Arch Ophthalmol. 1982;100:249e55 26. Bazhin AV. The need for standardization of antiretinal antibody detection and measurement. Am J Ophthalmol. 2009;147:374, author reply 375. 27. Bazhin AV, Dalke C, Willner N, et al. Cancer-retina antigens as potential paraneoplastic antigens in melanomaassociated retinopathy. Int J Cancer. 2009;124:140e9 28. Bazhin AV, Savchenko MS, Shifrina ON, et al. Recoverin as a paraneoplastic antigen in lung cancer: the occurrence of anti-recoverin autoantibodies in sera and recoverin in tumors. Lung Cancer. 2004;44:193e8 29. Berson EL, Lessell S. Paraneoplastic night blindness with malignant melanoma. Am J Ophthalmol. 1988;106:307e11 30. Berta A, Beck P, Mikita J. IgM paraprotein in the subretinal fluid of a patient with recurrent retinal detachment and Waldenstrom’s macroglobulinaemia. Acta Med Hung. 1985;42:179e86 31. Besirli CG, Comer GM. High-resolution OCT imaging of RPE degeneration in bilateral diffuse uveal melanocytic proliferation. Ophthalmic Surg Lasers Imaging. 2010;41(Suppl)):S96e100
451
32. Bhatti MT, Gres KE, Petitto VB, et al. Cells, cells, and more cells. Surv Ophthalmol. 2007;52:315e24 33. Bianciotto C, Shields CL, Thirkill CE, et al. Paraneoplastic retinopathy with multiple detachments of the neurosensory retina and autoantibodies against interphotoreceptor retinoid binding protein (IRBP) in cutaneous melanoma. Br J Ophthalmol. 2010;94:1684e5 34. Bolling JP, Brazis PW. Optic disk swelling with peripheral neuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes (POEMS syndrome). Am J Ophthalmol. 1990;109:503e10 35. Borkowski LM, Grover S, Fishman GA, et al. Retinal findings in melanoma-associated retinopathy. Am J Ophthalmol. 2001;132:273e5 36. Borruat FX, Othenin-Girard P, Uffer S, et al. Natural history of diffuse uveal melanocytic proliferation. Case report. Ophthalmology. 1992;99:1698e704 37. Bourdette DN, Rosenberg NL. Infiltrative orbitopathy, optic disk edema, and POEMS. Neurology. 1984;34:532e3 38. Brazis PW, Liesegang TJ, Bolling JP, et al. When do optic disc edema and peripheral neuropathy constitute poetry? Surv Ophthalmol. 1990;35:219e25 39. Brody JM, Butrus SI, Ashraf MF, et al. Multiple myeloma presenting with bilateral exudative macular detachments. Acta Ophthalmol Scand. 1995;73:81e2 40. Bruggemann A, von Bulow M, Finas D, et al. Bilateral diffuse uveal melanocytic proliferation associated with the recurrence of a Bartholin gland carcinoma. Br J Ophthalmol. 2011;95:888e9 41. Campo E, Brunier MN, Merino MJ. Small cell carcinoma of the endometrium with associated ocular paraneoplastic syndrome. Cancer. 1992;69:2283e8 42. Cao R, Cao Y. Cancer-associated retinopathy: a new mechanistic insight on vascular remodeling. Cell Cycle. 2010;9:1882e5 43. Cao R, Xue Y, Hedlund EM, et al. VEGFR1-mediated pericyte ablation links VEGF and PlGF to cancerassociated retinopathy. Proc Natl Acad Sci USA. 2010;107:856e61 44. Carboni G, Forma G, Bond AD, et al. Bilateral paraneoplastic optic neuropathy and unilateral retinal compromise in association with prostate cancer: a differential diagnostic challenge in a patient with unexplained visual loss. Doc Ophthalmol. 2012;125:63e70 45. Casale Turu A, Cabot Garcia J, Llanas Reus R, et al. [Papilledema and POEMS syndrome]. Ophthalmologica. 1992;205:144e8 46. Chahud F, Young RH, Remulla JF, et al. Bilateral diffuse uveal melanocytic proliferation associated with extraocular cancers: review of a process particularly associated with gynecologic cancers. Am J Surg Pathol. 2001;25:212e8 47. Chan C, O’Day J. Melanoma-associated retinopathy: does autoimmunity prolong survival? Clin Experiment Ophthalmol. 2001;29:235e8 48. Chan JW. Paraneoplastic retinopathies and optic neuropathies. Surv Ophthalmol. 2003;48:12e38 49. Chang AE, Karnell LH, Menck HR. The National Cancer Data Base report on cutaneous and noncutaneous melanoma: a summary of 84,836 cases from the past decade. The American College of Surgeons Commission on Cancer and the American Cancer Society. Cancer. 1998;83:1664e78 50. Chang JM, Lee HJ, Goo JM, et al. False positive and false negative FDG-PET scans in various thoracic diseases. Korean J Radiol. 2006;7:57e69 51. Chin MS, Caruso RC, Detrick B, et al. Autoantibodies to p75/ LEDGF, a cell survival factor, found in patients with atypical retinal degeneration. J Autoimmun. 2006;27:17e27
452
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
52. Chong DY, Comer GM, Trobe JD. Optic disc edema, cystoid macular edema, and elevated vascular endothelial growth factor in a patient with POEMS syndrome. J Neuroophthalmol. 2007;27:180e3 53. Cogan DG, Kuwabara T, Currie J, et al. [Paraneoplastic retinopathy simulating cone dystrophy with achromatopsia]. Klin Monbl Augenheilkd. 1990;197:156e8 54. Coppeto JR, Monteiro ML, Cannarozzi DB. Optic neuropathy associated with chronic lymphomatous meningitis. J Clin Neuroophthalmol. 1988;8:39e45 55. Cross SA, Salomao DR, Parisi JE, et al. Paraneoplastic autoimmune optic neuritis with retinitis defined by CRMP-5IgG. Ann Neurol. 2003;54:38e50 56. Dai RP, Dong FT, Chu J. [Ocular features of polyneuropathyorganomegaly-endocrinopathy-monoclonal gammopathyskin changes syndrome]. Zhonghua Yan Ke Za Zhi. 2005;41:917e9 57. Darnell RB, Posner JB. Paraneoplastic syndromes involving the nervous system. N Engl J Med. 2003;349:1543e54 58. Darnell RB, Posner JB. Paraneoplastic Syndromes. New York, Oxford University Press; 2011 59. De Salvo G, Prakash P, Rennie CA, et al. Long-term survival in a case of bilateral diffuse uveal melanocytic proliferation. Eye (Lond). 2011;25:1385e6 60. De Santis G, Caniatti L, De Vito A, et al. A possible paraneoplastic neuromyelitis optica associated with lung cancer. Neurol Sci. 2009;30:397e400 61. de Wolff-Rouendaal D. Bilateral diffuse benign melanocytic tumours of the uveal tract. A clinicopathological study. Int Ophthalmol. 1985;7:149e60 62. Dhingra A, Fina ME, Neinstein A, et al. Autoantibodies in melanoma-associated retinopathy target TRPM1 cation channels of retinal ON bipolar cells. J Neurosci. 2011;31:3962e7 63. Dispenzieri A. POEMS syndrome. Blood Rev. 2007;21:285e99 64. Dispenzieri A. POEMS syndrome: 2011 update on diagnosis, risk-stratification, and management. Am J Hematol. 2011;86:591e601 65. Duong HV, McLean IW, Beahm DE. Bilateral diffuse melanocytic proliferation associated with ovarian carcinoma and metastatic malignant amelanotic melanoma. Am J Ophthalmol. 2006;142:693e5 66. Dutton JJ, Burde RM, Klingele TG. Autoimmune retrobulbar optic neuritis. Am J Ophthalmol. 1982;94:11e7 67. Eichen JG, Dalmau J, Demopoulos A, et al. The photoreceptor cell-specific nuclear receptor is an autoantigen of paraneoplastic retinopathy. J Neuroophthalmol. 2001;21:168e72 68. Ejma M, Misiuk-Hojlo M, Gorczyca WA, et al. Antibodies to 46-kDa retinal antigen in a patient with breast carcinoma and cancer-associated retinopathy. Breast Cancer Res Treat. 2008;110:269e71 69. Eksandh L, Adamus G, Mosgrove L, et al. Autoantibodies against bestrophin in a patient with vitelliform paraneoplastic retinopathy and a metastatic choroidal malignant melanoma. Arch Ophthalmol. 2008;126:432e5 70. Eltabbakh GH, Hoogerland DL, Kay MC. Paraneoplastic retinopathy associated with uterine sarcoma. Gynecol Oncol. 1995;58:120e3 71. Espandar L, O’Brien S, Thirkill C, et al. Successful treatment of cancer-associated retinopathy with alemtuzumab. J Neurooncol. 2007;83:295e302 72. Ferreyra HA, Jayasundera T, Khan NW, et al. Management of autoimmune retinopathies with immunosuppression. Arch Ophthalmol. 2009;127:390e7 73. Filipic M, Ambler JS. Bilateral diffuse melanocytic uveal tumours associated with systemic malignant neoplasm. Aust N Z J Ophthalmol. 1986;14:293e9
74. Fine HF, Spaide RF, Ryan EH Jr,, et al. Acute zonal occult outer retinopathy in patients with multiple evanescent white dot syndrome. Arch Ophthalmol. 2009;127:66e70 75. Finger ML, Thirkill C, Borruat FX. Unusual paraneoplastic cause of vision loss: combined paraneoplastic cone dystrophy and optic neuropathy. Arch Ophthalmol. 2012;130:660e2 76. Flynn MF, Fisherman GA, Adamus G. Antiretinal Muller cell antibodies in patients with melanoma associated and autoimmune retinopathy [abstract]. Invest Ophthalmol Vis Sci. 2000;41:s567 77. Forooghian F, Macdonald IM, Heckenlively JR, et al. The need for standardization of antiretinal antibody detection and measurement. Am J Ophthalmol. 2008;146:489e95 78. Francis PJ, Marinescu A, Fitzke FW, et al. Acute zonal occult outer retinopathy: towards a set of diagnostic criteria. Br J Ophthalmol. 2005;89:70e3 79. Freidman AH, Marchevsky A, Odel JG, et al. Immunofluorescent studies of the eye in Waldenstrom’s macroglobulinemia. Arch Ophthalmol. 1980;98:743e6 80. Frohman LP, Cook SD, Bielory L. Dysgammaglobulinemia in steroid-dependent optic neuritis: response to gammaglobulin treatment. J Clin Neuroophthalmol. 1991;11:241e5 81. Fujiwara T, Imamura Y, Giovinazzo VJ, et al. Fundus autofluorescence and optical coherence tomographic findings in acute zonal occult outer retinopathy. Retina. 2010;30:1206e16 82. Gass JD. Acute zonal occult outer retinopathy. Donders Lecture: The Netherlands Ophthalmological Society, Maastricht, Holland, June 19, 1992. J Clin Neuroophthalmol. 1993;13:79e97 83. Gass JD. Are acute zonal occult outer retinopathy and the white spot syndromes (AZOOR complex) specific autoimmune diseases? Am J Ophthalmol. 2003;135:380e1 84. Gass JD, Agarwal A, Scott IU. Acute zonal occult outer retinopathy: a long-term follow-up study. Am J Ophthalmol. 2002;134:329e39 85. Gass JD, Gieser RG, Wilkinson CP, et al. Bilateral diffuse uveal melanocytic proliferation in patients with occult carcinoma. Arch Ophthalmol. 1990;108:527e33 86. Gass JD, Glatzer RJ. Acquired pigmentation simulating Peutz-Jeghers syndrome: initial manifestation of diffuse uveal melanocytic proliferation. Br J Ophthalmol. 1991;75:693e5 87. Gherardi RK, Belec L, Fromont G, et al. Elevated levels of interleukin-1 beta (IL-1 beta) and IL-6 in serum and increased production of IL-1 beta mRNA in lymph nodes of patients with polyneuropathy, organomegaly, endocrinopathy, M protein, and skin changes (POEMS) syndrome. Blood. 1994;83:2587e93 88. Gherardi RK, Belec L, Soubrier M, et al. Overproduction of proinflammatory cytokines imbalanced by their antagonists in POEMS syndrome. Blood. 1996;87:1458e65 89. Gitlits VM, Toh BH, Sentry JW. Disease association, origin, and clinical relevance of autoantibodies to the glycolytic enzyme enolase. J Investig Med. 2001;49:138e45 90. Gittinger JW Jr,, Smith TW. Cutaneous melanoma-associated paraneoplastic retinopathy: histopathologic observations. Am J Ophthalmol. 1999;127:612e4 91. Goetgebuer G, Kestelyn-Stevens AM, De Laey JJ, et al. Cancerassociated retinopathy (CAR) with electronegative ERG: a case report. Doc Ophthalmol. 2008;116:49e55 92. Gray RH. Unilateral enlargement of the blind spot: a diagnostic dilemma. Br J Ophthalmol. 2002;86:936e8 93. Grunwald L, Kligman BE, Shields CL. Acute exudative polymorphous paraneoplastic vitelliform maculopathy in
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
94.
95.
96.
97.
98.
99. 100.
101.
102.
103.
104. 105.
106.
107.
108.
109.
110.
111.
112.
113.
a patient with carcinoma, not melanoma. Arch Ophthalmol. 2011;129:1104e6 Gutierrez-Ortiz C, Castro-Rebollo M, Pareja J, et al. Bilateral non-simultaneous optic neuropathy and unilateral macular edema in a patient with POEMS syndrome. Eur J Ophthalmol. 2009;19:166e9 Guy J, Aptsiauri N. Treatment of paraneoplastic visual loss with intravenous immunoglobulin: report of 3 cases. Arch Ophthalmol. 1999;117:471e7 Handler MZ, Mruthyunjaya P, Nelson K. Melanomaassociated retinopathy: a presenting sign of metastatic disease. J Am Acad Dermatol. 2011;65:e9e11 Harmon JP, Purvin VA, Guy J, et al. Cancer-associated retinopathy in a patient with advanced epithelial ovarian carcinoma. Gynecol Oncol. 1999;73:430e2 Hartmann TB, Bazhin AV, Schadendorf D, et al. SEREX identification of new tumor antigens linked to melanomaassociated retinopathy. Int J Cancer. 2005;114:88e93 Heckenlively J, Wang A. AZOOR defined (acute zonal occult outer retinopathy). Acta Ophthalmol. 2011;89:s248 Heckenlively JR, Fawzi AA, Oversier J, et al. Autoimmune retinopathy: patients with antirecoverin immunoreactivity and panretinal degeneration. Arch Ophthalmol. 2000;118:1525e33 Heckenlively JR, Ferreyra HA. Autoimmune retinopathy: a review and summary. Semin Immunopathol. 2008;30:127e34 Heckenlively JR, Jordan BL, Aptsiauri N. Association of antiretinal antibodies and cystoid macular edema in patients with retinitis pigmentosa. Am J Ophthalmol. 1999;127:565e73 Heckenlively JR, Solish AM, Chant SM, et al. Autoimmunity in hereditary retinal degenerations. II. Clinical studies: antiretinal antibodies and fluorescein angiogram findings. Br J Ophthalmol. 1985;69:758e64 Ho AC, Benson WE, Wong J. Unusual immunogammopathy maculopathy. Ophthalmology. 2000;107:1099e103 Hoh ST, Teh M, Chew SJ. Paraneoplastic optic neuropathy in nasopharyngeal carcinomadreport of a case. Singapore Med J. 1991;32:170e3 Holz FG, Kim RY, Schwartz SD, et al. Acute zonal occult outer retinopathy (AZOOR) associated with multifocal choroidopathy. Eye (Lond). 1994;8(Pt 1):77e83 Honnorat J, Antoine JC, Derrington E, et al. Antibodies to a subpopulation of glial cells and a 66 kDa developmental protein in patients with paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry. 1996;61:270e8 Hoogenraad TU, Sanders EACM, Tan KEWP. Paraneoplastic optic neuropathy with histopathological verification of absence of meningeal metastases. Neuroophthalmology. 1989;9:247e50 Huynh N, Shildkrot Y, Lobo AM, et al. Intravitreal triamcinolone for cancer-associated retinopathy refractory to systemic therapy. J Ophthalmic Inflamm Infect. 2012;2:169e71 Imai H, Kusuhara S, Nakanishi Y, et al. A case of POEMS syndrome with cystoid macular edema. Am J Ophthalmol. 2005;139:563e6 Jaben EA, Pulido JS, Pittock S, et al. The potential role of plasma exchange as a treatment for bilateral diffuse uveal melanocytic proliferation: a report of two cases. J Clin Apher. 2011;26:356e61 Jabs DA, Miller NR, Newman SA, et al. Optic neuropathy in systemic lupus erythematosus. Arch Ophthalmol. 1986;104:564e8 Jacobson DM, Adamus G. Retinal anti-bipolar cell antibodies in a patient with paraneoplastic retinopathy and colon carcinoma. Am J Ophthalmol. 2001;131:806e8
453
114. Jacobson DM, Thirkill CE. Paraneoplastic cone dysfunction: an unusual visual remote effect of cancer. Arch Ophthalmol. 1995;113:1580e2 115. Jacobson DM, Thirkill CE, Tipping SJ. A clinical triad to diagnose paraneoplastic retinopathy. Ann Neurol. 1990;28:162e7 116. Jacobson SG, Morales DS, Sun XK, et al. Pattern of retinal dysfunction in acute zonal occult outer retinopathy. Ophthalmology. 1995;102:1187e98 117. Jacobzone C, Cochard-Marianowski C, Kupfer I, et al. Corticosteroid treatment for melanoma-associated retinopathy: effect on visual acuity and electrophysiologic findings. Arch Dermatol. 2004;140:1258e61 118. Jampol LM, Becker KG. White spot syndromes of the retina: a hypothesis based on the common genetic hypothesis of autoimmune/inflammatory disease. Am J Ophthalmol. 2003;135:376e9 119. Jampol LM, Fishman GA. Immunosuppression for autoimmune retinopathy. Arch Ophthalmol. 2009;127:573e5 120. Jampol LM, Kim HH, Bryar PJ, et al. Multiple serous retinal detachments and subretinal deposits as the presenting signs of metastatic melanoma. Retina. 2004;24:320e2 121. Jampol LM, Wiredu A. MEWDS, MFC, PIC, AMN, AIBSE, and AZOOR: one disease or many? Retina. 1995;15:373e8 122. Javaheri M, Khurana RN, Bhatti RA, et al. Optical coherence tomography findings in paraneoplastic pseudovitelliform lesions in melanoma-associated retinopathy. Clin Ophthalmol. 2008;2:461e3 123. Kanai K, Kuwabara S, Misawa S, et al. Failure of treatment with anti-VEGF monoclonal antibody for long-standing POEMS syndrome. Intern Med. 2007;46:311e3 124. Katsuta H, Okada M, Nakauchi T, et al. Cancer-associated retinopathy associated with invasive thymoma. Am J Ophthalmol. 2002;134:383e9 125. Kaushik M, Pulido JS, Abreu R, et al. Ocular findings in patients with polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes syndrome. Ophthalmology. 2011;118:778e82 126. Kellner U, Bornfeld N, Foerster MH. Severe course of cutaneous melanoma associated paraneoplastic retinopathy. Br J Ophthalmol. 1995;79:746e52 127. Kelly JJ Jr, Kyle RA, Miles JM, et al. Osteosclerotic myeloma and peripheral neuropathy. Neurology. 1983;33:202e10 128. Keltner JL, Roth AM, Chang RS. Photoreceptor degeneration. Possible autoimmune disorder. Arch Ophthalmol. 1983;101:564e9 129. Keltner JL, Thirkill CE. The 22-kDa antigen in optic nerve and retinal diseases. J Neuroophthalmol. 1999;19:71e83 130. Keltner JL, Thirkill CE, Huynh PM. Autoimmune-related retinopathy and optic neuropathy (ARRON) syndrome (Abstract). J Neuroophthalmol. 2002;22:160e1 131. Keltner JL, Thirkill CE, Tyler NK, et al. Management and monitoring of cancer-associated retinopathy. Arch Ophthalmol. 1992;110:48e53 132. Keltner JL, Thirkill CE, Yip PT. Clinical and immunologic characteristics of melanoma-associated retinopathy syndrome: eleven new cases and a review of 51 previously published cases. J Neuroophthalmol. 2001;21:173e87 133. Kennedy MJ, Eustace P, O’Briain DS, et al. Paraneoplastic papilloedema in neuroblastoma. Postgrad Med J. 1987;63:873e6 134. Kennelly KD, Dodick DW, Pascuzzi RM, et al. Neuronal autoantibodies and paraneoplastic neurological syndromes associated with extrapulmonary small cell carcinoma. Neurology. 1997;48(Suppl 2):A31 135. Khan N, Huang JJ, Foster CS. Cancer associated retinopathy (CAR): An autoimmune-mediated paraneoplastic syndrome. Semin Ophthalmol. 2006;21:135e41
454
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
136. Khurana RN, Wieland MR, Boldrey EE, et al. Vitelliform retinopathy in metastatic cutaneous melanoma with choroidal involvement. Arch Ophthalmol. 2011;129:1498e9 137. Kikuchi T, Arai J, Shibuki H, et al. Tubby-like protein 1 as an autoantigen in cancer-associated retinopathy. J Neuroimmunol. 2000;103:26e33 138. Kim JH, Kim JH, Kim DH, et al. Intravenously administered anti-recoverin antibody alone does not pass through the blood-retinal barrier. Korean J Ophthalmol. 2011;25:189e95 139. Kim RY, Retsas S, Fitzke FW, et al. Cutaneous melanomaassociated retinopathy. Ophthalmology. 1994;101:1837e43 140. Kim SJ, Toma HS, Thirkill CE, et al. Cancer-associated retinopathy with retinal periphlebitis in a patient with ovarian cancer. Ocul Immunol Inflamm. 2010;18:107e9 141. Kimmel DW, O’Neill BP, Lennon VA. Subacute sensory neuronopathy associated with small cell lung carcinoma: diagnosis aided by autoimmune serology. Mayo Clin Proc. 1988;63:29e32 142. Kiratli H, Erkan K. Loss of retinal pigment epithelium associated with bilateral diffuse uveal melanocytic proliferation. Ophthalmic Surg Lasers Imaging;1e4 143. Kitakawa T, Hayashi T, Takashina H, et al. Improvement of central visual function following steroid pulse therapy in acute zonal occult outer retinopathy. Doc Ophthalmol. 2012;124:249e54 144. Klingele TG, Burde RM, Rappazzo JA, et al. Paraneoplastic retinopathy. J Clin Neuroophthalmol. 1984;4:239e45 145. Koike C, Obara T, Uriu Y, et al. TRPM1 is a component of the retinal ON bipolar cell transduction channel in the mGluR6 cascade. Proc Natl Acad Sci USA. 2010;107:332e7 146. Kondo M, Sanuki R, Ueno S, et al. Identification of autoantibodies against TRPM1 in patients with paraneoplastic retinopathy associated with ON bipolar cell dysfunction. PLoS One. 2011;6:e19911 147. Koreen L, He SX, Johnson MW, et al. Anti-retinal pigment epithelium antibodies in acute exudative polymorphous vitelliform maculopathy: a new hypothesis about disease pathogenesis. Arch Ophthalmol. 2011;129:23e9 148. Krema H, Simpson R, Altomare F, et al. Paraneoplastic vitelliform retinopathy in metastatic cutaneous melanoma. Retin Cases Brief Rep. 2010;4:246e50 149. Kumar VA, Abbas A, Fausto N. Neoplasia, in Robbins and Cotran Pathologic Basis of Disease. Philadelphia, PA, Elsevier Saunders; 2005. ed 7. pp 333e5 150. Kupersmith MJ, Burde RM, Warren FA, et al. Autoimmune optic neuropathy: evaluation and treatment. J Neurol Neurosurg Psychiatry. 1988;51:1381e6 151. Kuroda Y, Miyahara M, Sakemi T, et al. Autopsy report of acute necrotizing opticomyelopathy associated with thyroid cancer. J Neurol Sci. 1993;120:29e32 152. Ladewig G, Reinhold U, Thirkill CE, et al. Incidence of antiretinal antibodies in melanoma: screening of 77 serum samples from 51 patients with American Joint Committee on Cancer stage I-IV. Br J Dermatol. 2005;152:931e8 153. Lafaut BA, Bourgoignie KB, Sallet G, et al. [Diffuse bilateral proliferation of uveal melanocytes. Apropos of an uncommon case]. J Fr Ophtalmol. 1994;17:208e13 154. Lambrecht ER, van der Loos TL, van der Eerden AH. Retrobulbar neuritis as the first sign of the glucagonoma syndrome. Int Ophthalmol. 1987;11:13e5 155. Lee JM, Seong HK, Nam WH, et al. Cancer-associated nummular loss of the retinal pigment epithelium. Korean J Ophthalmol. 2007;21:261e4 156. Lei B, Bush RA, Milam AH, et al. Human melanomaassociated retinopathy (MAR) antibodies alter the retinal ON-response of the monkey ERG in vivo. Invest Ophthalmol Vis Sci. 2000;41:262e6
157. Lima LH, Greenberg JP, Greenstein VC, et al. Hyperautofluorescent ring in autoimmune retinopathy. Retina. 2012;32:1385e94 158. Linke R, Schroeder M, Helmberger T, et al. Antibody-positive paraneoplastic neurologic syndromes: value of CT and PET for tumor diagnosis. Neurology. 2004;63:282e6 159. Lu Y, Jia L, He S, et al. Melanoma-associated retinopathy: a paraneoplastic autoimmune complication. Arch Ophthalmol. 2009;127:1572e80 160. Luiz JE, Lee AG, Keltner JL, et al. Paraneoplastic optic neuropathy and autoantibody production in small-cell carcinoma of the lung. J Neuroophthalmol. 1998;18:178e81 161. Machemer R. [On the pathogenesis of the flat malignant melanoma]. Klin Monbl Augenheilkd. 1966;148:641e52 162. Machida S, Ohguro H, Tateda M, et al. Melanoma-associated retinopathy associated with intranasal melanoma. Doc Ophthalmol. 2011;122:191e7 163. Maeda A, Maeda T, Liang Y, et al. Effects of cytotoxic T lymphocyte antigen 4 (CTLA4) signaling and locally applied steroid on retinal dysfunction by recoverin, cancerassociated retinopathy antigen. Mol Vis. 2006;12:885e91 164. Maeda A, Ohguro H, Maeda T, et al. Aberrant expression of photoreceptor-specific calcium-binding protein (recoverin) in cancer cell lines. Cancer Res. 2000;60:1914e20 165. Maeda T, Maeda A, Maruyama I, et al. Mechanisms of photoreceptor cell death in cancer-associated retinopathy. Invest Ophthalmol Vis Sci. 2001;42:705e12 166. Mahdi N, Faia LJ, Goodwin J, et al. A case of autoimmune retinopathy associated with thyroid carcinoma. Ocul Immunol Inflamm. 2010;18:322e3 167. Malik S, Furlan AJ, Sweeney PJ, et al. Optic neuropathy: a rare paraneoplastic syndrome. J Clin Neuroophthalmol. 1992;12:137e41 168. Mandelcorn E, McGuire K, Dixon W, et al. Ocular paraneoplastic syndrome: a case of bilateral diffuse uveal melanocytic proliferation. Retina. 2009;29:1375e6 169. Mantel I, Ramchand KV, Holder GE, et al. Macular and retinal dysfunction of unknown origin in adults with normal fundi: evidence for an autoimmune pathophysiology. Exp Mol Pathol. 2008;84:90e101 170. Margolin E, Flint A, Trobe JD. High-titer collapsin responsemediating protein-associated (CRMP-5) paraneoplastic optic neuropathy and Vitritis as the only clinical manifestations in a patient with small cell lung carcinoma. J Neuroophthalmol. 2008;28:17e22 171. Masaoka N, Emoto Y, Sasaoka A, et al. Fluorescein angiographic findings in a case of cancer-associated retinopathy. Retina. 1999;19:462e4 172. Matsubara S, Yamaji Y, Fujita T, et al. Cancer-associated retinopathy syndrome: a case of small cell lung cancer expressing recoverin immunoreactivity. Lung Cancer. 1996;14:265e71 173. Matsui Y, Mehta MC, Katsumi O, et al. Electrophysiological findings in paraneoplastic retinopathy. Graefes Arch Clin Exp Ophthalmol. 1992;230:324e8 174. McGinnis JF, Austin B, Klisak I, et al. Chromosomal assignment of the human gene for the cancer-associated retinopathy protein (recoverin) to chromosome 17p13.1. J Neurosci Res. 1995;40:165e8 175. McGinnis JF, Lerious V, Pazik J, et al. Chromosomal assignment of the recoverin gene and cancer-associated retinopathy. Mamm Genome. 1993;4:43e5 176. McKeon A, Apiwattanakul M, Lachance DH, et al. Positron emission tomography-computed tomography in paraneoplastic neurologic disorders: systematic analysis and review. Arch Neurol. 2010;67:322e9 177. Mets RB, Golchet P, Adamus G, et al. Bilateral diffuse uveal melanocytic proliferation with a positive ophthalmoscopic
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
178.
179.
180.
181.
182.
183.
184.
185.
186. 187.
188.
189. 190.
191.
192.
193.
194.
195.
196.
197.
and visual response to plasmapheresis. Arch Ophthalmol. 2011;129:1235e8 Milam AH, Saari JC, Jacobson SG, et al. Autoantibodies against retinal bipolar cells in cutaneous melanomaassociated retinopathy. Invest Ophthalmol Vis Sci. 1993;34:91e100 Miles SL, Niles RM, Pittock S, et al. A factor found in the IgG fraction of serum of patients with Paraneoplastic Bilateral Diffuse Uveal Melanocytic Proliferation causes proliferation of cultured human melanocytes. Retina. 2012;32:1959e66 Mineta M, Hatori M, Sano H, et al. Recurrent Crow-Fukase syndrome associated with increased serum levels of vascular endothelial growth factor: a case report and review of the literature. Tohoku J Exp Med. 2006;210:269e77 Miralles GD, O’Fallon JR, Talley NJ. Plasma-cell dyscrasia with polyneuropathy. The spectrum of POEMS syndrome. N Engl J Med. 1992;327:1919e23 Misiuk-Hojlo M, Ejma M, Gorczyca WA, et al. Cancerassociated retinopathy in patients with breast carcinoma. Arch Immunol Ther Exp (Warsz). 2007;55:261e5 Mizener JB, Kimura AE, Adamus G, et al. Autoimmune retinopathy in the absence of cancer. Am J Ophthalmol. 1997;123:607e18 Mkrtchyan M, Lujan BJ, Merino D, et al. Outer retinal structure in patients with acute zonal occult outer retinopathy. Am J Ophthalmol. 2012;153:757e68 Mohamed Q, Harper CA. Acute optical coherence tomographic findings in cancer-associated retinopathy. Arch Ophthalmol. 2007;125:1132e3 Monson DM, Smith JR. Acute zonal occult outer retinopathy. Surv Ophthalmol. 2011;56:23e35 Mora P, Gonzales S, Crafa P, et al. Peculiar findings in a case of bilateral uveal pigmented lesions. Ocul Immunol Inflamm. 2010;18:379e82 Morgans CW, Zhang J, Jeffrey BG, et al. TRPM1 is required for the depolarizing light response in retinal ON-bipolar cells. Proc Natl Acad Sci USA. 2009;106:19174e8 Moss HE, Liu GT, Dalmau J. Glazed (vision) and confused. Surv Ophthalmol. 2010;55:169e73 Mudhar HS, Scott I, Ul-Hassan A, et al. Bilateral diffuse uveal melanocytic hyperplasia: molecular characterization and novel association with bilateral renal papillary carcinoma. Histopathology. 2012;61:751e4 Murakami A, Yajima T, Inana G. Isolation of human retinal genes: recoverin cDNA and gene. Biochem Biophys Res Commun. 1992;187:234e44 Murakami Y, Yoshida S, Yoshikawa H, et al. CRMP-5-IgG in patient with paraneoplastic optic neuritis with lung adenocarcinoma. Eye (Lond). 2007;21:860e2 Murayama K, Takita H, Kiyohara Y, et al. [Melanomaassociated retinopathy with unknown primary site in a Japanese woman]. Nihon Ganka Gakkai Zasshi. 2006;110:211e7 Murphy MA, Hart WM Jr,, Olk RJ. Bilateral diffuse uveal melanocytic proliferation simulating an arteriovenous fistula. J Neuroophthalmol. 1997;17:166e9 Murphy MA, Thirkill CE, Hart WM Jr,. Paraneoplastic retinopathy: a novel autoantibody reaction associated with small-cell lung carcinoma. J Neuroophthalmol. 1997;17:77e83 Nakamura M, Sanuki R, Yasuma TR, et al. TRPM1 mutations are associated with the complete form of congenital stationary night blindness. Mol Vis. 2010;16:425e37 Nakayama-Ichiyama S, Yokote T, Hiraoka N, et al. A paraneoplastic neuromyelitis optica spectrum disorder associated with a mature B-cell neoplasm. Leuk Res. 2011;35:e111e3
455
198. Navajas EV, Simpson ER, Krema H, et al. Cancer-associated nummular loss of RPE: expanding the clinical spectrum of bilateral diffuse uveal melanocytic proliferation. Ophthalmic Surg Lasers Imaging. 2011;42:e103e6 199. Neutzner RV, Jager M, Friedburg C, et al. [Blind spot enlargement syndrome in acute zonal occult outer retinopathy with detection of autoantibodies against the retinal antigens CRALBP and S-Ag]. Ophthalmologe. 2011;108:1045e9 200. Nieuwendijk TJ, Hooymans JM. Paraneoplastic vitelliform retinopathy associated with metastatic choroidal melanoma. Eye (Lond). 2007;21:1436e7 201. O’Neal KD, Butnor KJ, Perkinson KR, et al. Bilateral diffuse uveal melanocytic proliferation associated with pancreatic carcinoma: a case report and literature review of this paraneoplastic syndrome. Surv Ophthalmol. 2003;48:613e25 202. Ohguro H, Maruyama I, Nakazawa M, et al. Antirecoverin antibody in the aqueous humor of a patient with cancerassociated retinopathy. Am J Ophthalmol. 2002;134:605e7 203. Ohguro H, Ogawa K, Maeda T, et al. Retinal dysfunction in cancer-associated retinopathy is improved by Ca(2þ) antagonist administration and dark adaptation. Invest Ophthalmol Vis Sci. 2001;42:2589e95 204. Ohguro H, Ogawa K, Nakagawa T. Recoverin and Hsc 70 are found as autoantigens in patients with cancer-associated retinopathy. Invest Ophthalmol Vis Sci. 1999;40:82e9 205. Ohguro H, Palczewski K, Milam AH. A retina bipolar cell protein is recognized by autoantibodies from patients with melanoma-associated retinopathy [abstract]. Invest Ophthalmol Vis Sci. 1999;40:s153 206. Ohguro H, Yokoi Y, Ohguro I, et al. Clinical and immunologic aspects of cancer-associated retinopathy. Am J Ophthalmol. 2004;137:1117e9 207. Ohkawa T, Kawashima H, Makino S, et al. Cancer-associated retinopathy in a patient with endometrial cancer. Am J Ophthalmol. 1996;122:740e2 208. Ohnishi Y, Ohara S, Sakamoto T, et al. Cancer-associated retinopathy with retinal phlebitis. Br J Ophthalmol. 1993;77:795e8 209. Okada K, Yamamoto S, Tsuyama Y, et al. Case of POEMS syndrome associated with bilateral macular detachment resolved by autologous peripheral blood stem cell transplantation. Jpn J Ophthalmol. 2007;51:237e8 210. Okel BB, Thirkill CE, Anderson K. An unusual case of melanoma-associated retinopathy. Ocul Immunol Inflamm. 1995;3:121e7 211. Oohira A, Inoue T, Fukuda N, et al. A case with paraneoplastic optic neuropathy presenting bitemporal hemianopsia. Neuroophthalmology. 1991;11:325e8 212. Otsuji F, Abematsu N, Nakao K, et al. [A case of cancerassociated retinopathy rapidly leading to blindness with a unique pupillary light reflex]. Nihon Ganka Gakkai Zasshi. 2011;115:924e9 213. Oyama Y, Burt RK, Thirkill C, et al. A case of autoimmunerelated retinopathy and optic neuropathy syndrome treated by autologous nonmyeloablative hematopoietic stem cell transplantation. J Neuroophthalmol. 2009;29:43e9 214. Palmowski AM, Haus AH, Pfohler C, et al. Bilateral multifocal chorioretinopathy in a woman with cutaneous malignant melanoma. Arch Ophthalmol. 2002;120:1756e61 215. Parc CE, Azan E, Bonnel S, et al. Cone dysfunction as a paraneoplastic syndrome associated with retinal antigens approximating 40 kiloDalton. Ophthalmic Genet. 2006;27:57e61 216. Pau D, Yalamanchili S, Lee AG. Long-term survivor of paraneoplastic optic neuropathy. J Neuroophthalmol. 2010;30:387
456
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
217. Peek R, Verbraak F, Coevoet HM, et al. Muller cell-specific autoantibodies in a patient with progressive loss of vision. Invest Ophthalmol Vis Sci. 1998;39:1976e9 218. Pfohler C, Schmaltz R, Graeber S, et al. ’Autoimmunity’ is not always associated with prolonged survival in melanoma patients. Br J Dermatol. 2008;159:735e6 219. Piao CH, Kondo M, Ishikawa S, et al. A case of unusual retinopathy showing features similar to acute zonal occult outer retinopathy associated with negative electroretinograms. Jpn J Ophthalmol. 2007;51:69e71 220. Pillay N, Gilbert JJ, Ebers GC, et al. Internuclear ophthalmoplegia and "optic neuritis": paraneoplastic effects of bronchial carcinoma. Neurology. 1984;34:788e91 221. Pilon AF, Rhee PS, Messner LV. Bilateral, persistent serous macular detachments with Waldenstrom’s macroglobulinemia. Optom Vis Sci. 2005;82:573e8 222. Polans AS, Buczylko J, Crabb J, et al. A photoreceptor calcium binding protein is recognized by autoantibodies obtained from patients with cancer-associated retinopathy. J Cell Biol. 1991;112:981e9 223. Polans AS, Burton MD, Haley TL, et al. Recoverin, but not visinin, is an autoantigen in the human retina identified with a cancer-associated retinopathy. Invest Ophthalmol Vis Sci. 1993;34:81e90 224. Polans AS, Witkowska D, Haley TL, et al. Recoverin, a photoreceptor-specific calcium-binding protein, is expressed by the tumor of a patient with cancerassociated retinopathy. Proc Natl Acad Sci USA. 1995;92:9176e80 225. Potter MJ, Adamus G, Szabo SM, et al. Autoantibodies to transducin in a patient with melanoma-associated retinopathy. Am J Ophthalmol. 2002;134:128e30 226. Potter MJ, Thirkill CE, Dam OM, et al. Clinical and immunocytochemical findings in a case of melanoma-associated retinopathy. Ophthalmology. 1999;106:2121e5 227. Prause JU, Jensen OA, Eisgart F, et al. Bilateral diffuse malignant melanoma of the uvea associated with large cell carcinoma, giant cell type, of the lung. Case report of a newly described syndrome. Ophthalmologica. 1984;189:221e8 228. Pulido J, Cross SA, Lennon VA, et al. Bilateral autoimmune optic neuritis and vitreitis related to CRMP-5-IgG: intravitreal triamcinolone acetonide therapy of four eyes. Eye (Lond). 2008;22:1191e3 229. Querques G, Thirkill CE, Hagege H, et al. Choroidal neovascularization associated with cancer-associated retinopathy. Acta Ophthalmol. 2010;88:571e5 230. Raghunath A, Adamus G, Bodurka DC, et al. Cancerassociated retinopathy in neuroendocrine carcinoma of the fallopian tube. J Neuroophthalmol. 2010;30:252e4 231. Rappoport D, Leiba H. Presumed melanoma-associated retinopathy (MAR): a presenting sign of primary small intestinal melanoma? Int Ophthalmol. 2012;32:387e91 232. Reddy S, Finger PT. Unilateral diffuse uveal melanocytic proliferation (DUMP). Br J Ophthalmol. 2007;91:1726e7 233. Remulla JF, Pineda R, Gaudio AR, et al. Cutaneous melanoma-associated retinopathy with retinal periphlebitis. Arch Ophthalmol. 1995;113:854e5 234. Richter RB, Moore RY. Non-invasive central nervous system disease associated with lymphoid tumors. Johns Hopkins Med J. 1968;122:271e83 235. Riedel P, Wall M, Grey A, et al. Autoimmune optic neuropathy. Arch Ophthalmol. 1998;116:1121e4 236. Ripps H, Carr RE, Siegel IM, et al. Functional abnormalities in vincristine-induced night blindness. Invest Ophthalmol Vis Sci. 1984;25:787e94
237. Ritland JS, Eide N, Tausjo J. Bilateral diffuse uveal melanocytic proliferation and uterine cancer. A case report. Acta Ophthalmol Scand. 2000;78:366e8 238. Rizzo JF 3rd,, Gittinger JW Jr,. Selective immunohistochemical staining in the paraneoplastic retinopathy syndrome. Ophthalmology. 1992;99:1286e95 239. Rohrbach JM, Roggendorf W, Thanos S, et al. Simultaneous bilateral diffuse melanocytic uveal hyperplasia. Am J Ophthalmol. 1990;110:49e56 240. Rose C, Zandecki M, Copin MC, et al. POEMS syndrome: report on six patients with unusual clinical signs, elevated levels of cytokines, macrophage involvement and chromosomal aberrations of bone marrow plasma cells. Leukemia. 1997;11:1318e23 241. Rosencher L, Maisonobe T, Lavole A, et al. [Neurologic paraneoplastic syndrome with anti-CV2/CRMP5 antibodies revealing a small cell lung cancer. Effectiveness of the lung cancer treatment]. Rev Neurol (Paris). 2012;168:371e4 242. Rush JA. Paraneoplastic retinopathy in malignant melanoma. Am J Ophthalmol. 1993;115:390e1 243. Rush JA, Kennerdell JS, Martinez AJ. Primary idiopathic inflammation of the optic nerve. Am J Ophthalmol. 1982;93:312e6 244. Ryll DL, Campbell RJ, Robertson DM, et al. 7. Pseudometastatic lesions of the choroid. Ophthalmology. 1980;87:1181e6 245. Saito W, Kase S, Ohguro H, et al. Slowly progressive cancerassociated retinopathy. Arch Ophthalmol. 2007;125:1431e3 246. Saito W, Kase S, Yoshida K, et al. Bilateral diffuse uveal melanocytic proliferation in a patient with cancerassociated retinopathy. Am J Ophthalmol. 2005;140:942e5 247. Sakamori Y, Kim YH, Okuda C, et al. Two cases of cancerassociated retinopathy combined with small-cell lung cancer. Jpn J Clin Oncol. 2011;41:669e73 248. Salgia R, Hedges TR, Rizk M, et al. Cancer-associated retinopathy in a patient with non-small-cell lung carcinoma. Lung Cancer. 1998;22:149e52 249. Samaras P, Bauer S, Stenner-Liewen F, et al. Treatment of POEMS syndrome with bevacizumab. Haematologica. 2007;92:1438e9 250. Savchenko MS, Goncharskaia MA, Skorikova EE, et al. Autoantibodies against the Ca(2þ)-binding protein recoverin in blood sera of patients with various oncological diseases. Oncol Lett. 2012;3:377e82 251. Sawyer RA, Selhorst JB, Zimmerman LE, et al. Blindness caused by photoreceptor degeneration as a remote effect of cancer. Am J Ophthalmol. 1976;81:606e13 252. Scarlato M, Previtali SC, Carpo M, et al. Polyneuropathy in POEMS syndrome: role of angiogenic factors in the pathogenesis. Brain. 2005;128:1911e20 253. Scott CL, Cher LM, O’Day J. Cancer associated retinopathy and non-small cell lung cancer. J Clin Neurosci. 1997;4:355e7 254. Scott JX, Moses PD, Somashekar HR, et al. Paraneoplastic papilloedema in a child with neuroblastoma. Indian J Cancer. 2005;42:102e3 255. Seles S, Lang GE. [CAR syndrome (carcinoma-associated retinopathy syndrome) associated with an adenocarcinoma of the cervix]. Klin Monbl Augenheilkd. 2005;222:736e9 256. Semenova EA, Chin KJ, Natesh S, et al. Fundus autofluorescence imaging of diffuse uveal melanocytic proliferation. Ophthalmic Surg Lasers Imaging;1e3 257. Sen J, Clewes AR, Quah SA, et al. Presymptomatic diagnosis of bronchogenic carcinoma associated with bilateral diffuse uveal melanocytic proliferation. Clin Experiment Ophthalmol. 2006;34:156e8 258. Shen Y, Heimel JA, Kamermans M, et al. A transient receptor potential-like channel mediates synaptic transmission in rod bipolar cells. J Neurosci. 2009;29:6088e93
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
259. Sheorajpanday R, Slabbynck H, Van De Sompel W, et al. Small cell lung carcinoma presenting as collapsin responsemediating protein (CRMP) -5 paraneoplastic optic neuropathy. J Neuroophthalmol. 2006;26:168e72 260. Shildkrot Y, Sobrin L, Gragoudas ES. Cancer-associated retinopathy: update on pathogenesis and therapy. Semin Ophthalmol. 2011;26:321e8 261. Shimazaki K, Jirawuthiworavong GV, Heckenlively JR, et al. Frequency of anti-retinal antibodies in normal human serum. J Neuroophthalmol. 2008;28:5e11 262. Shinmura M, Hamasaki D, Matsuyama T, et al. [A case of squamous cell carcinoma of the lung with bilateral diffuse uveal melanocytic proliferation (BDUMP) diagnosed by visual disturbance]. Nihon Kokyuki Gakkai Zasshi. 2011;49:187e91 263. So K, Shinoda K, Matsumoto CS, et al. Focal functional and microstructural changes of photoreceptors in eyes with acute zonal occult outer retinopathy. Case Report Ophthalmol. 2011;2:307e13 264. Sobottka B, Schlote T, Besch D, et al. [Carcinoma-associated retinopathy: a review with clinical examples]. Klin Monbl Augenheilkd. 2000;216:17e24 265. Sotodeh M, Paridaens D, Keunen J, et al. Paraneoplastic vitelliform retinopathy associated with cutaneous or uveal melanoma and metastases. Klin Monbl Augenheilkd. 2005;222:910e4 266. Soubrier M, Dubost JJ, Serre AF, et al. Growth factors in POEMS syndrome: evidence for a marked increase in circulating vascular endothelial growth factor. Arthritis Rheum. 1997;40:786e7 267. Spaide RF, Koizumi H, Freund KB. Photoreceptor outer segment abnormalities as a cause of blind spot enlargement in acute zonal occult outer retinopathy-complex diseases. Am J Ophthalmol. 2008;146:111e20 268. Srikantha N, Goverdhan S, Evans A. Paraneoplastic optic neuropathy associated with papillary renal cell carcinoma. Br J Ophthalmol. 2011;95:429 269. Straume O, Bergheim J, Ernst P. Bevacizumab therapy for POEMS syndrome. Blood. 2006;107:4972e3, author reply 4973e4. 270. Subhadra C, Dudek AZ, Rath PP, et al. Improvement in visual fields in a patient with melanoma-associated retinopathy treated with intravenous immunoglobulin. J Neuroophthalmol. 2008;28:23e6 271. Suelves AM, Garcia-Delpech S, Gallego R, et al. Diagnostic and therapeutic challenges. Retina. 2012;32:635e9 272. Suzuki T, Obara Y, Sato Y, et al. Cancer-associated retinopathy with presumed vasculitis. Am J Ophthalmol. 1996;122:125e7 273. Takahashi M, Hirami Y, Sakuma K, et al. [Case of cancerassociated retinopathy in which immediate treatment succeeded]. Nihon Ganka Gakkai Zasshi. 2008;112:806e11 274. Tanaka A, Takase H, Adamus G, et al. Cancer-associated retinopathy caused by benign thymoma. Br J Ophthalmol. 2010;94:526e8 275. Thambisetty MR, Scherzer CR, Yu Z, et al. Paraneoplastic optic neuropathy and cerebellar ataxia with small cell carcinoma of the lung. J Neuroophthalmol. 2001;21:164e7 276. Thirkill CE, FitzGerald P, Sergott RC, et al. Cancer-associated retinopathy (CAR syndrome) with antibodies reacting with retinal, optic-nerve, and cancer cells. N Engl J Med. 1989;321:1589e94 277. Thirkill CE, Roth AM, Keltner JL. Cancer-associated retinopathy. Arch Ophthalmol. 1987;105:372e5 278. To KW, Thirkill CE, Jakobiec FA, et al. Lymphoma-associated retinopathy. Ophthalmology. 2002;109:2149e53 279. Toribio-Garcia JA, Franco-Benito M, Garcia-Ruiz-deMorales JM. [Paraneoplastic optic neuritis in an unknown
280.
281.
282.
283. 284.
285.
286.
287.
288.
289.
290.
291.
292.
293. 294.
295.
296.
297. 298.
299.
300.
457
small cell lung carcinoma defined by CRMP-5-IgG]. Arch Soc Esp Oftalmol. 2007;82:777e80 Tsukahara S, Wakui K, Ohzeki S. Simultaneous bilateral primary diffuse malignant uveal melanoma: case report with pathological examination. Br J Ophthalmol. 1986;70:33e8 Ulrich JN, Garg S, Escaravage GK Jr,, et al. Bilateral diffuse uveal melanocytic proliferation presenting as small choroidal melanoma. Case Rep Ophthalmol Med. 2011;2011:1e3 van Genderen MM, Bijveld MM, Claassen YB, et al. Mutations in TRPM1 are a common cause of complete congenital stationary night blindness. Am J Hum Genet. 2009;85:730e6 Vaphiades MS, Brown H, Whitcup SM. Node way out. Surv Ophthalmol. 2000;45:77e83 Vedeler CA, Antoine JC, Giometto B, et al. Management of paraneoplastic neurological syndromes: report of an EFNS Task Force. Eur J Neurol. 2006;13:682e90 Vernino S, Tuite P, Adler CH, et al. Paraneoplastic chorea associated with CRMP-5 neuronal antibody and lung carcinoma. Ann Neurol. 2002;51:625e30 Watanabe O, Arimura K, Kitajima I, et al. Greatly raised vascular endothelial growth factor (VEGF) in POEMS syndrome. Lancet. 1996;347:702 Watanabe O, Maruyama I, Arimura K, et al. Overproduction of vascular endothelial growth factor/vascular permeability factor is causative in Crow-Fukase (POEMS) syndrome. Muscle Nerve. 1998;21:1390e7 Weleber RG, Watzke RC, Shults WT, et al. Clinical and electrophysiologic characterization of paraneoplastic and autoimmune retinopathies associated with antienolase antibodies. Am J Ophthalmol. 2005;139:780e94 Whitcup SM, Vistica BP, Milam AH, et al. Recoverinassociated retinopathy: a clinically and immunologically distinctive disease. Am J Ophthalmol. 1998;126:230e7 Wiaux C, Landau K, Borruat FX. Unusual cause of bilateral optic disc swelling: POEMS syndrome. Klin Monbl Augenheilkd. 2007;224:334e6 Wong VA, Wade NK. POEMS syndrome: an unusual cause of bilateral optic disk swelling. Am J Ophthalmol. 1998;126:452e4 Wu S, Slakter JS, Shields JA, et al. Cancer-associated nummular loss of the pigment epithelium. Am J Ophthalmol. 2005;139:933e5 Yamada G, Ohguro H, Aketa K, et al. Invasive thymoma with paraneoplastic retinopathy. Hum Pathol. 2003;34:717e9 Yamaji Y, Matsubara S, Yamadori I, et al. Characterization of a small-cell-lung-carcinoma cell line from a patient with cancer-associated retinopathy. Int J Cancer. 1996;65:671e6 Yamamoto S, Hanaya J, Mera K, et al. Recovery of visual function in patient with melanoma-associated retinopathy treated with surgical resection and interferon-beta. Doc Ophthalmol Yanagi Y, Eguchi R, Obata R, et al. Autoimmune retinopathy after chronic renal allograft rejection. Arch Ophthalmol. 2006;124:418e20 Yannuzzi LA. The Retinal Atlas. Philadelphia, PA, Elsevier; 2010. pp 689e90. Younes-Mhenni S, Janier MF, Cinotti L, et al. FDG-PET improves tumour detection in patients with paraneoplastic neurological syndromes. Brain. 2004;127:2331e8 Yu S, Ikeda T, Ikeda N, et al. Coloration of fundus lesions in bilateral diffuse uveal melanocytic proliferation. Jpn J Ophthalmol. 2003;47:612e5 Yu Z, Kryzer TJ, Griesmann GE, et al. CRMP-5 neuronal autoantibody: marker of lung cancer and thymoma-related autoimmunity. Ann Neurol. 2001;49:146e54
458
s u r v e y o f o p h t h a l m o l o g y 5 8 ( 2 0 1 3 ) 4 3 0 e4 5 8
301. Zacks DN, Pinnolis MK, Berson EL, et al. Melanomaassociated retinopathy and recurrent exudative retinal detachments in a patient with choroidal melanoma. Am J Ophthalmol. 2001;132:578e81
other cited material
A. Sobrin L. Cancer-associated retinopathy: update on diagnosis and treatment. Retina 2011 The Magical World of Retina; American Academy of Ophthalmology October 21, 2011; Orlando, Florida. B. A 58-year-old female with bubbles in her vision. Case of the month: January, 2011. West Coast Retina. Available at: http:// westcoastretina.com/WestCoastRetina/Jan-2011.html. Accessed May 20, 2012. C. Pulido JS. Paraneoplastic bilateral diffuse uveal melanocytic proliferation is caused by a factor found in the IgG fraction of
serum. 44th Annual Retina Society Scientific Meeting; The Retina Society and the Societa Italiana della Retina 2011; Rome, Italy. D. Paraneoplastic autoantibody evaluation, serum. Mayo Medical Laboratories Test Catalog. [database on the Internet]. Rochester, MN: Mayo Foundation, Mayo Medical Laboratories; 2000e2001. Available at: http://www.mayomedicallabora tories.com/test-catalog/Overview/83380. Accessed May 20, 2012. E. Keltner JL. Paraneoplastic retinopathies and optic neuropathies vs. (non-cancer) autoimmune related retinopathy and optic neuropathy (ARRON syndrome). 36th Annual North American Neuro-Ophthalmology Society (NANOS) Meeting March 6e12, 2010; Tucson, AZ. F. Recombx CAR (anti-recoverin) autoantibody test. Athena Diagnostics [database on the Internet]. Worcester, MA; 2012. Available at: http://www.athenadiagnostics.com/content/ contact/. Accessed May 20, 2012.