Pentosan Polysulfate

Pentosan Polysulfate Maculopathy: What Urologists Should Know in 2020

Jan Alberto Paredes Mogica* Anahuac University

 * Corresponding Author

Key Words: Pentosan Sulfuric Polyester, Macular Degeneration, Drug-Related Side Effects and Adverse Reactions
Abstract

Objective: To conduct a review of current literature to assess whether an association exists between Pentosan Polysulfate Sodium and the development of macular disease, as it is the only oral medication approved by the Food and Drug Administration for the management of interstitial cystitis.
Materials and Methods: A systematic review was conducted by the authors separately, with review methods established prior to the conduct of the review. Databases searched included PubMed, Ovid, Medline, EBSCO, and Google Scholar. A search was conducted for the terms “Pentosan Polysulfate Maculopathy”, “Pentosan Polysulfate Retinopathy” and “Interstitial Cystitis Maculopathy”. All papers reporting on primary data were included.
Results: A total of 14 papers reporting on primary data were identified. Most papers reported on the development of macular disease in the setting of chronic Pentosan Polysulfate Sodium

 

1
exposure. No randomized controlled trials have been performed to date and data was insufficient to perform a meta-analysis. Nevertheless, patients with interstitial cystitis were more likely to receive a diagnosis of maculopathy after several years of the medication use.
Conclusions: Although the nature of the published studies renders them prone to confounders, currently available data suggest an increased risk for developing maculopathy after years of Pentosan Polysulfate Sodium use. In light of this, and the marginal effectiveness of the medication for the average individual, we suggest that education be provided as to the possible association and that regular ophthalmic evaluation be recommended for patients who are continued on chronic Pentosan Polysulfate Sodium.

 

 

Introduction
Pentosan Polysulfate Sodium (PPS), commercially known as Elmiron®, is a semisynthetic pentasaccharide heparinoid.1 It was used in the 1950’s as an anticoagulant due to its thrombolytic properties.2,3 In 1996, PPS was approved by the Food and Drug Administration (FDA) for the treatment of interstitial cystitis/bladder pain syndrome (IC/BPS). As of today, PPS remains the only oral treatment modality approved by the FDA for the management of IC/BPS.4
Historically, some trials have reported the potential adverse reactions of PPS; the most common being diarrhea, headache and nausea. Liver function abnormalities and thrombocytopenia have also been described.5-8 More recently, in 2018, Pearce et al. identified 6 patients with a unique pigmentary maculopathy (Figure 1) in the setting of chronic PPS exposure (median exposure of 186 months and 2263 cumulative grams [heretofore abbreviated as g]).9 This maculopathy was

 

2
characterized by difficulty reading and prolonged dark adaptation. Upon retinal imaging, paracentral hyperpigmentation with surrounding vitelliform deposits (round, yellowish deposits) were found at the retinal pigment epithelium.9,10 It was later theorized that this toxicity could be the result of PPS antagonism of Fibroblast Growth Factor (FGF) signaling in the retina.11 Since then, several other authors have investigated this potential association and have found conflicting results. Some have suggested the retinopathy findings are a manifestation of IC/BPS itself.12,13 Since the potential association is a novel finding, most available data as of June 2020 is in the form of case reports and retrospective studies that may be prone to confounders.
In this review we aim to compile all available data on the question as to whether chronic PPS use puts patients at increased risk for the development of retinopathy. If the association exists, urologists in particular and providers in general should be aware of the implications for initiating and maintaining patients on PPS.
Materials and Methods

The study design was a systematic review planned in accordance with AMSTAR criteria.14 Review methods were established prior to the conduct of the review, as follows, and there were no deviations from the protocol. A comprehensive search of five databases was performed by two independent reviewers: Pubmed, Ovid, Medline, EBSCO, and Google Scholar. The latter was chosen given the infancy of the diagnosis in order to cull for publications in non-standard journals. There were no language exclusions. The terms used for the search included “Pentosan Polysulfate Maculopathy”, “Pentosan Polysulfate Retinopathy” and “Interstitial Cystitis Maculopathy”. The search strategy for each database was identical and is outlined in Appendix 1. Inclusion criteria for the review consisted of every paper that had reported on retinopathy

 

3
cases related to PPS use or associated with IC itself. Due to the novel nature of the subject, all papers with reports on primary data were selected, including case reports, case series and case- control studies (no randomized controlled trials were available). Both published and as of yet unpublished papers were included. The exclusion criteria were any articles that lacked report on primary data regarding PPS or IC associated retinopathy, e.g. letters to the editor or news pieces.

 

 

Results

Of 19 publications, 5 were excluded for lacking primary data; a total of 14 articles containing original data were 1) identified by both reviewers separately and 2) met all established criteria. Of these papers, 4 were case reports, 4 were case series and 6 were cross sectional studies. All papers were included in the review. A lowchart describing the literature search and included studies is presented in Figure 2.

All included papers are summarized in Table 1. Risk of bias and assessment of study quality is presented in Table 2. A total of 4 case reports were found in the literature. All case reports described a female patient with a longstanding diagnosis of IC/BPS managed with PPS. Among these 4 cases, patients had a mean age of 50 years and had been exposed to PPS for an average of 201 months (range 120-252 months), with a mean cumulative dose of 1,461 g (range 1150-2300 g). Three out of 4 patients presented with symptoms of blurry vision and trouble adapting to low light conditions; the 4th patient was asymptomatic. Upon dilated fundus examination, all four patients were found to have retinal pigment epithelium (RPE) abnormalities; the most prominent finding was hyperpigmented deposits within the macula. One patient demonstrated a choroidal neovascular membrane (new, damaging blood vessels that grow beneath the retina) via optical

 

4
coherence tomography (OCT). Genetic testing for inherited retinal disease was performed in 3 of the 4 patients, and all tested negative. Of note, one patient continued to develop retinal changes 7 years after discontinuing PPS (it had been stopped before undergoing sacral stimulation).15-18

A total of 4 case series were identified, including the original case series reported by Pearce et al. These series reported on a total of 68 patients with maculopathy in the setting of PPS use. All patients had a previous diagnosis of IC/BPS and were treated with PPS for at least 3 years. The mean period of time patients were exposed to PPS was 177 months (range 141-204 months) and received a mean cumulative exposure of 1,818 g (range 1610-2482 g). The mean age of the 68 patients was 56.5 years. Although one case series didn’t report on the sex of patients, females accounted for the majority of reported cases (93% of the remaining population). The case series reported by Hadad et al. screened asymptomatic patients directly regarding vision changes; only one patient reported trouble with dark adaptation. For the remaining of the case series, difficulty with dark adaptation was the most commonly reported symptom with 30 out of 51 (58.8%) being affected, followed by reading difficulty (54.9%) and metamorphopsia (9.8%). Regardless of symptoms, nearly all patients had an imaging examination consistent with the presence of bilateral paracentral pigmentary changes at the level of the RPE, either by dilated fundus examination, OCT or autofluorescence imaging. 9,10,19,20

Of the remaining reports, all 6 were cross sectional studies, 2 being large matched cohort studies. Hanif et al. examined 219 patients with the diagnosis of IC/BPS. These patients were being treated with several medications: 80 (36.5%) of the group had taken PPS for a mean duration and cumulative exposure of 219.6 months (range 36-262.8 months) and 2,300 g (range 580-2980 g) respectively. Most patients were female (89%), with a mean age of 60.8 years old. Of the 80 patients who were on PPS therapy, 20 patients (25%) had developed an unspecified pigmentary

 

5
maculopathy on exam compared to 4 out of the 139 patients (2.8%) who had never been under PPS treatment. Of the affected 20 patients in the exposed group, 14 had the classic features of RPE abnormalities originally described by Pearce et al. After analysis, it was found that PPS exposure was the sole predictor for the presence of an unspecified pigmentary maculopathy, with an odds ratio (OR) of 11.25 (95% confidence interval, 3.69-34.33, P < 0.0001). The strength of association for the development of retinal disease was not statistically significant for age, sex, smoking status, race and other medications. 10

Vora et al. screened 117 patients who were prescribed at least 500 g of PPS over a 20 year period and who were still taking the medication. Subjects were 84.6% female with a mean age of 64.4 years. These patients were screened with OCT and fundus photography, in which 27 patients (23.1%) were found to have definite signs of PPS associated maculopathy, as originally described by Pearce et al. Multivariable regression analysis found the cumulative PPS dose to be the sole predicting factor for the presence of PPS related maculopathy. Patients with maculopathy had a mean cumulative dose of 1350 g compared to 1040 g in the unexposed group (t test, P < 0.01). Within the affected group, the presence of maculopathy tended to increase among patients with the highest cumulative dose; patients with a cumulative dose of 500-999 g had a 12.7% chance of having PPS maculopathy compared to 41.7% in patients who had a
>1,500 g cumulative dose.21
Wang et al. screened 50 patients who had been prescribed PPS, of whom 46 (92%) were female and were a mean age of 60 years. Of the 50, 48 had been prescribed PPS for IC/BPS with the remaining 2 for irritable bowel syndrome and pelvic pain syndrome respectively. Patients were exposed to the medication for a mean duration of 109.2 months and a cumulative dose averaging 1228.4 g. All patients were screened via color fundus photography, OCT and fundus

 

6
autofluorescence (FAF). A total of 10 subjects (20%) were found to have the characteristic findings of PPS maculopathy, of whom 5 complained of nyctalopia (difficulty with seeing night). No statistically significant differences were found when comparing the affected and unaffected group regarding sex, age, BMI or tobacco use. The only statistically significant differences were duration of treatment, daily dose, and total cumulative dose of PPS. The unaffected group had a mean duration and cumulative dose of 75.6 months and 691.7 g, whereas the affected group had a mean duration and cumulative dose of 243.6 months and 3375.4 g respectively (p < 0.001). No patients were found to have PPS associated maculopathy with a cumulative dose lower than 1,500 g.22

Barnes et al. conducted a study to answer the question of whether ophthalmologists would be able to distinguish PPS maculopathy from hereditary causes of maculopathy based on imaging alone. A total of 1,131 patients with a diagnosis of maculopathy were analyzed by 2 blinded reviewers. Analysis was performed via imaging, including fundus color imaging, OCT and FAF. Prior to the review, 10 patients were previously identified by the researchers as demonstrating the classic features of PPS associated maculopathy as originally described by Pearce et al. (i.e. macular hyperpigmented spots and patchy RPE atrophy, along with autofluorescence findings centered on the fovea). The blinded reviewers were then tasked with assigning the subjects to 3 groups according to the likelihood of having PPS maculopathy. All 10 patients with PPS macular disease were correctly identified and placed in the correct group. An additional 5 were misclassified as having PPS maculopathy. This accounted for the 100% sensitivity and 99.6% specificity estimated by the authors for distinguishing PPS maculopathy from hereditary conditions based on imaging alone (blind to history of PPS use or symptoms). Distinguishing features were peripapillary autofluorescence and spaced hyper-autofluorescent macular spots. 23

 

7
Two large retrospective cross-sectional studies were conducted to assess the relationship between PPS and macular disease. Jain et al. conducted a retrospective, matched cohort study by pooling data from Clinformatics Data Mart Database.24 A group exposed to PPS was matched to an unexposed group by age, sex, race and insurance start date. The match ratio was 1 exposed subject to 5 unexposed subjects (1:5). Exposure (as measured by PPS prescription coverage) averaged 10-13 months. Both exposed and unexposed groups had a diagnosis of IC/BPS. These cohorts were retrospectively compared at 5 (n=18,072) and at 7 years (n=9621) after starting PPS. Assessment was made for the development of atypical maculopathy, drusen or age-related macular degeneration (AMD). At the 5 year follow-up, 103 (3.4%) of exposed subjects developed a type of maculopathy compared with the 440 (2.9%) of unexposed subjects, but it was found to not be statistically significant (OR=1.20, 95% CI 0.95 to 1.51, p=0.13). At the 7 year follow-up, 87 (5.4%) of exposed subjects developed a type of maculopathy compared with the 328 (4.1%) unexposed subjects, this time being statistically significant via multivariate analysis (OR=1.41, 95% CI 1.09 to 1.83, p=0.009). At the 7 year follow-up, patients had a mean age of 52.8 years and 83% were females. 13

Ludwig et al. conducted a multicenter, retrospective cohort study of 49,899 patients with a diagnosis of IC/BPS. Data was taken from the MarketScan database.25 Of the cohort, 89.9% were women and most were between 50 and 60 years of age when they received the diagnosis of IC/BPS. A PPS prescription was filled by 23% of subjects, averaging an exposure time of 1230 days (41 months). Follow-up was made retrospectively for 5 years, and groups were compared for the development of macular disease. Upon unadjusted analysis, exposed patients had a similar probability of being diagnosed with a maculopathy (2.37%; n=277) when compared to unexposed patients (2.77%; n=1058), demonstrating no significant associations between PPS

 

8
exposure and any maculopathy. After excluding patients with a diagnosis of diabetes (17.2% of the total population), a hazard ratio of 1.24 (95% CI, 1.05 to 1.47) was found for the development of any maculopathy in patients exposed to PPS, albeit not statistically significant (p=0.011). A hazard ratio of 14.41 was found for the diagnosis of hereditary dystrophy in patients exposed to PPS for at least 4 years; however, the number of patients with the diagnosis was deemed to be low (n=20) and it had broad confidence intervals (95% CI, 1.73 to 119.92, p=0.014).12

Risk of Bias and Study Quality: Risk of bias was assessed according to the techniques outlined by Aaron et al. and the “RoB 2: A revised Cochrane risk of bias tool for randomized trials”.26, 27 Study quality was assessed by applying the Newcastle Ottawa Scale where applicable.28 Due to the nature of the published literature on this subject, most papers received a high overall risk of bias. This means both reporting bias and evidence selection bias might be present in this systematic review. Assessment of quality and risk of bias for each study is summarized in Table 2.
Source of funding was assessed in each study and no conflicts of interest were reported or suspected.

 

 

Discussion
After analysis, most reports and studies seem to suggest an association exists between long term PPS exposure and the development of maculopathy. This association seems to increase the longer patients are exposed to the medication, as demonstrated in the studies conducted by Jain,

 

9

Wang and Vora.13,21,22 The prevalence of maculopathy in PPS users varied widely, but seemed

to be related to the cumulative amount of medication to which patients were exposed: most affected patients with PPS maculopathy were found to have a cumulative exposure of over 1000 g. The two large cohort studies conducted by Jain and Ludwig demonstrated a probability of being diagnosed with a maculopathy of 3.4% and 2.3% respectively, after 5 years of PPS use.12,13 In the Jain study, this number was significantly higher than the comparison group at 7 years follow-up (5.4% versus 4.1% in the comparison group). despite relatively short length of exposure to PPS (10-13 months). The range of exposure – and the effect – was greater in most other studies (120 to 243 months). The highest prevalence was reported by Vora et al. at 41.7% of all patients with a cumulative dose higher than 1,500 g.21 The Ludwig study, on the other hand, reported a relatively short term follow up (5 years) and relatively short duration of PPS use at 41 months (albeit longer than the Jain report), but failed to show any strong associations between PPS and maculopathy. The Ludwig data was limited by the retrospective use of coding data for maculopathy and IC, but reported on a large number of patients (227,325 patients with IC). Due to some weak associations with hereditary dystrophy and increased risk of maculopathy in their non-diabetic groups on PPS, the Ludwig group still concluded that prescribers of PPS should provide patients the appropriate warnings of the potential risk of drug-induced maculopathy.
Females comprised most of the affected patients, but this is explained by the predominance of IC/BPS diagnosis in the female population.29 The symptoms predominantly described by patients were nyctalopia (difficulty seeing in dim or night light), difficulty reading and metamorphopsia (distorted vision in which a set of straight lines appears wavy). Imaging findings were also found

 

10
to be consistent across patients, with bilateral perimacular pigment abnormalities at the level of the RPE being described in all cases. Furthermore, the study conducted by Barnes et al. suggests that this type of maculopathy might be differentiated from other forms of maculopathy by imaging alone, by ophthalmologists blinded to history of PPS use and IC.23
The efficacy of PPS is an important factor in the decision to maintain long term usage. Its therapeutic benefit was brought into question by a double blind placebo study conducted in 2015: no treatment effect was found for PPS when compared to placebo.30 A subsequent meta analysis conducted in 2019 showed a 12.4% difference in a global response assessment between PPS and placebo (95% CI: 6.4%–18.3%, p<0.001).4 For those who have success with the medication, there is potential to stay on it for years. The limited data on PPS retinal toxicity suggests that it is dependent on cumulative dosing. Of concern, is also the apparent progression of the condition even after discontinuation of the drug, as reported by Huckfeldt R. et al.18 Recently, Shah R. et al. also reported worsening maculopathy in 11 patients even after they stopped PPS ingestion; these patients had a mean exposure of 1,970 g and had a mean follow up of 12 months after PPS discontinuation.31 Metabolism of PPS involves partial desulfation in the liver and spleen. The kidney is also involved in its metabolism, by partial depolymerization of several metabolites. It has been reported that continuing dosing of PPS can result in the saturation of both desulfation and depolymerization pathways. 32 Slow saturation of these pathways after years of PPS use could partially explain its delayed toxicity The potential risk for maculopathy, progression after cessation, and reevaluation of efficacy are important considerations in patients with IC/PBS who are on chronic PPS.

 

11
.

 

Even though most literature reports on the association between PPS use and maculopathy, most are single center reports or case series. No study has been able to definitively answer the question as to whether a significant association exists between prolonged PPS use and the development of maculopathy. We agree with the recommendation of Doiron, Bona and Nickel (CUAJ 2020;14:10-11) that the question would best be answered by a prospective, properly powered, multi-center screening study of patients with IC/BPS, including a chronically exposed cohort as well as a cohort of those unexposed to the drug.33
In October 2019, Health Canada, the Canadian government health regulatory authority, published key messages for healthcare professionals stating that in the post market setting, pigmentary maculopathy has been reported with the chronic use of PPS, including difficulty reading and prolonged dark adaptation.34 It recommended patients have regular ophthalmic examinations for early detection and that discontinuation of the medication should be considered if identified. In addition, pigmentary maculopathy was added to the “warnings and precautions, “post-market adverse drug reactions”, and “consumer information” sections of the Canadian monograph.
As of June 2020, there are warnings on the label of PPS as marketed in the United States, under the brand name Elmiron®. It includes warnings regarding pigmentary changes in the retina.30 It references symptoms of difficulty reading, slow adjustment to low or reduced light environments, and blurred vision. It recommends baseline ophthalmologic evaluation within 6

 

12
months of starting therapy, including a good ophthalmologic history, a baseline retinal examination (including OCT and auto-fluorescence imaging) and posterior evaluations while continuing treatment. Cessation should be considered if retinal changes develop. Other adverse effects mentioned include liver function abnormalities, increased risk for rectal hemorrhage, alopecia, diarrhea, nausea, headache, rash, dyspepsia, and abdominal pain.

Conclusion

Existing data suggest there could be an association of PPS with pigmentary retinal maculopathy. More research is needed to characterize and confirm the association. Given the marginal efficacy of the drug, providers should exercise caution when prescribing the medication, by carefully weighing risks and benefits. At the current time, especially in light of the new package insert on PPS as of June 2020, urologists and health care providers should inform their patients of this concern as part of shared decision making, assess the therapeutic effect of the medication and indications for use. In patients who plan chronic PPS use, regular ophthalmic evaluation should be recommended.

 

 

 

 

References

1.Swain SM, Parker B, Wellstein A, Lippman ME, Steakley C, DeLap R. Phase I trial of pentosan polysulfate. Invest New Drugs. 1995;13(1):55‐62. doi:10.1007/BF02614221
2.FRILEUX C. Un nouvel anticoagulant de synthèse le thrombocid [Thrombocid: a new synthetic anticoagulant]. Presse Med. 1951;59(8):159.

 

13
3.KONCZ J, BUCHERL E. Beitrag zur Wirkungsweise des Anticoagulans Thrombocid [On the mode of action of thrombocid]. Klin Wochenschr. 1951;29(37-38):650. doi:10.1007/BF01490174
4.Arndt van Ophoven, Kirsten Vonde, Winfried Koch, Günter Auerbach & Klaus P. Maag. Efficacy of pentosan polysulfate for the treatment of interstitial cystitis/bladder pain syndrome: results of a systematic review of randomized controlled trials, Current Medical Research and Opinion, 2019; 35:9, 1495-1503, DOI: 10.1080/03007995.2019.1586401
5.Nickel JC, Barkin J, Forrest J, et al. Randomized, double-blind, dose-ranging study of pentosan polysulfate sodium for interstitial cystitis. Urology. 2005;65(4):654‐658. doi:10.1016/j.urology.2004.10.071
6.Lush RM, Figg WD, Pluda JM, et al. A phase I study of pentosan polysulfate sodium in patients with advanced malignancies. Ann Oncol. 1996;7(9):939‐944. doi:10.1093/oxfordjournals.annonc.a010797
7.Jepsen, J. V., Sall, M., Rhodes, P. R., Schmidt, D., Messing, E., & Bruskewitz, R. C. Long-term experience with pentosan polysulfate in interstitial cystitis. Urology, 1998; 51(3), 381–387. doi:10.1016/s0090-4295(97)00714-0
8.Hanno, P. M. (1997). Analysis of long-term Elmiron therapy for interstitial cystitis. Urology, 1997; 49(5), 93–99. doi:10.1016/s0090-4295(97)00179-9
9.Pearce WA, Chen R, Jain N. Pigmentary Maculopathy Associated with Chronic Exposure to Pentosan Polysulfate Sodium. Ophthalmology. 2018;125(11):1793‐1802. doi:10.1016/j.ophtha.2018.04.026
10.Hanif AM, Armenti ST, Taylor SC, et al. Phenotypic Spectrum of Pentosan Polysulfate Sodium-Associated Maculopathy: A Multicenter Study [published online ahead of print,
2019 Sep 5]. JAMA Ophthalmology. 2019;137(11):1275‐1282.
doi:10.1001/jamaophthalmol.2019.3392
11.Greenlee T, Hom G, Conti T, Babiuch AS, Singh R. Re: Pearce et al.: Pigmentary maculopathy associated with chronic exposure to pentosan polysulfate sodium
(Ophthalmology. 2018;125:1793-1802). Ophthalmology. 2019;126(7):e51.
doi:10.1016/j.ophtha.2018.12.037
12.Ludwig CA, Vail D, Callaway NF, Pasricha MV, Moshfeghi DM. Pentosan Polysulfate Sodium Exposure and Drug-Induced Maculopathy in Commercially Insured Patients in

 

14
the United States. Ophthalmology. 2020;127(4):535‐543.
doi:10.1016/j.ophtha.2019.10.036
13.Jain N, Li AL, Yu Y, VanderBeek BL. Association of macular disease with long-term use of pentosan polysulfate sodium: findings from a US cohort [published online ahead of print, 2019 Nov 6]. Br J Ophthalmology. 2019; bjophthalmol-2019-314765. doi:10.1136/bjophthalmol-2019-314765
14.Shea BJ, Reeves BC, Wells G, Thuku M, Hamel C, Moran J, Moher D, Tugwell P, Welch V, Kristjansson E, Henry DA. AMSTAR 2: a critical appraisal tool for systematic reviews that include randomised or non-randomised studies of healthcare interventions, or both. BMJ. 2017 Sep 21;358:j4008.
15.Vora, R. A., Patel, A. P., Yang, S. S., & Melles, R. A case of pentosan polysulfate maculopathy originally diagnosed as stargardt disease. American Journal of Ophthalmology Case Reports, 2020; 17, 100604. doi:10.1016/j.ajoc.2020.100604
16.Mishra K, Patel TP, Singh MS. Choroidal Neovascularization Associated with Pentosan Polysulfate Toxicity. Ophthalmol Retina, 2019 Volume 4, Issue 1, 111 – 113. doi: https://doi.org/10.1016/j.oret.2019.08.006
17.Wingelaar MJ, Raevis JJ, Conlin KA, Stepien KE. Retinal Toxicity in a Patient on
Pentosan Polysulfate Sodium [published online ahead of print, 2020 Apr 17]. Urology.
2020;S0090-4295(20)30384-8. doi:10.1016/j.urology.2020.04.006
18.Huckfeldt RM, Vavvas DG. Progressive Maculopathy After Discontinuation of Pentosan Polysulfate Sodium. Ophthalmic Surg Lasers Imaging Retina. 2019;50(10):656-659. doi:10.3928/23258160-20191009-10
19.Hadad A, Helmy O, Leeman S, Schaal S. A Novel Multimethod Image Analysis to Quantify Pentosan Polysulfate Sodium Retinal Toxicity. Ophthalmology. 2020;127(3):429-431. doi:10.1016/j.ophtha.2019.10.013
20.Foote J, Hanif AM, Jain N. Chronic Exposure to Pentosan Polysulfate Sodium is Associated with Retinal Pigmentary Changes and Vision Loss. AUA 2019 Abstract MP47-03.
21.Vora RA, Patel AP, Melles R. Prevalence of Maculopathy Associated with Long-Term
Pentosan Polysulfate Therapy. Ophthalmology. 2020;127(6):835-836.
doi:10.1016/j.ophtha.2020.01.017

 

15
22.Derrick Wang, Adrian Au, Frederic Gunnemann, Assaf Hilely, Jackson Scharf, Khoi Tran, Michel Sun, Ja-Hong Kim, David Sarraf, Pentosan-associated maculopathy: prevalence, screening guidelines, and spectrum of findings based on prospective multimodal analysis, Canadian Journal of Ophthalmology, Volume 55, Issue 2, 2020, Pages 116-125, ISSN 0008-4182, https://doi.org/10.1016/j.jcjo.2019.12.001.
23.Barnes AC, Hanif AM, Jain N. Pentosan polysulfate maculopathy versus inherited macular dystrophies: comparative assessment with multimodal imaging [published online ahead of print, 2020 May 21]. Ophthalmol Retina. 2020;S2468-6530(20)30200-1. doi:10.1016/j.oret.2020.05.008
24.Optum Clinformatics Data Mart Database. Available online at: https://web.uri.edu/optum/ Accessed 06/30/20
25.IBM MarketScan Research Databases. Available online at: https://www.ibm.com/products/marketscan-research-databases Accessed 06/30/20
26.Aaron M. Drucker1, Patrick Fleming2 and An-Wen Chan. Research Techniques Made Simple: Assessing Risk of Bias in Systematic Reviews. Journal of Investigative Dermatology (2016) 136, e109ee114; doi:10.1016/j.jid.2016.08.021
27.Sterne JAC, Savović J, Page MJ, Elbers RG, Blencowe NS, Boutron I, Cates CJ, Cheng H-Y, Corbett MS, Eldridge SM, Hernán MA, Hopewell S, Hróbjartsson A, Junqueira DR, Jüni P, Kirkham JJ, Lasserson T, Li T, McAleenan A, Reeves BC, Shepperd S, Shrier I, Stewart LA, Tilling K, White IR, Whiting PF, Higgins JPT. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ 2019; 366: l4898.
28.GA Wells, B Shea, D O’Connell, J Peterson, V Welch, M Losos, P Tugwel. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses. Ottawa Hospital Research Institute. Accessed June 6th, 2020. Available at: http://www.ohri.ca/home.asp
29.Jones CA, Nyberg L. Epidemiology of interstitial cystitis. Urology. 1997;49(5A Suppl):2-9. doi:10.1016/s0090-4295(99)80327-6
30.Nickel JC, Herschorn S, Whitmore KE, et al. Pentosan polysulfate sodium for treatment of interstitial cystitis/bladder pain syndrome: insights from a randomized, double-blind, placebo-controlled study. J Urol. 2015;193:857–862.

 

16
31.Disease Course in Patients With Pentosan Polysulfate Sodium-Associated Maculopathy After Drug Cessation. Shah R; Simonett JM; Lyons RJ; Rao RC; Pennesi ME; Jain N. JAMA Ophthalmology. 2020 Jul 09.
32.Ortho-McNeil-Janssen Pharmaceuticals, Elmiron (Pentosan Polysulfate sodium). U.S.
Food and Drug Administration website. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/020193s009lbl.pdf. Revised June 2020. Accessed June 30, 2020.
33.Doiron RC, Bona M, Nickel JC. Possible drug-induced, vision-threatening maculopathy secondary to chronic pentosan polysulfate sodium (Elmiron®) exposure. Can Urol Assoc J. 2020;14(2):10-11. doi:10.5489/cuaj.6401
34.Elmiron (pentosan polysulfate sodium) [product monograph]. Toronto (ON): Janssen Inc.; 2019. Updated: October 2019 Accessed; Accessed June 6, 2020.
Figure and Table legends
Figure 1: Images obtained from patient over a 2-year period: (Top row) serial near- infrared reflectance and (Bottom row) OCT B scan demonstrating the progressive nature of the patchy retinal pigment epithelium atrophy in more severe cases. Reprinted from Ophthalmology: Journal of the American Academy of Ophthalmology, Volume 125, William A. Pearce, Rui Chen, Nieraj Jain, Pigmentary Maculopathy Associated with Chronic Exposure to Pentosan Polysulfate Sodium, pp. 1793-1802, Copyright (2020), with permission from Elsevier.

 

Figure 2: Flow chart outlining search strategy and included studies regarding pigmentary maculopathy associated with PPS Exposure.

 

17

 

 
Database
Date of Search
# of Independent Reviewers
Search Terms

OVID Pubmed Medline EBSCO Google Scholar
June 6th, 2020
2
“Pentosan Polysulfate Maculopathy”, OR “Pentosan Polysulfate Retinopathy” OR “Interstitial Cystitis Maculopathy”

 
Appendix 1: Table outlining search strategy employed for each database.

 

18

 

19

 

20

 

Type
of
Paper
Auth
ors
and
Year
Study Objecti ve
Total
Popu lation
PPS
Expo
sed
Popu lation
PPS
expos
ed popula tion with macul opathy
Con
trol
Gro
up
Contro l
group
with
macul opathy
Follo w Up
Findings

Case Report
Robi n A.
To report
1
1
1
N/A N/A
N/A
Originally identified

Vora,
2020
on a case of maculo pathy associa ted
with chronic PPS Intake.
as Stargardt disease. RPE abnormali ties found in both eyes. Cumulativ e exposure
of 1,150 g.

Case Report
Max
well
To report
1
1
1
N/A N/A
N/A
Retinal toxicity

Wing
elar,
2020
on a case of maculo pathy associa ted
with chronic PPS Intake.
develope
dafter 10 years of use. Patient was asympto matic. Cumulativ
edose of 1,095 g.

Case Report
Kapil
Mish
To report
1
1
1
N/A N/A
N/A
Choroidal Neovascu

ra,
2020
on a case of maculo pathy associa ted
with chronic
larization and RPE abnormali ties identified. Cumulativ e exposure

 

21

 

PPS Intake.
of 2,300g

Case Report
Rach
el
To report
1
1
1
N/A N/A
N/A
RPE abnormali

Huck feldt, 2019
on a case of maculo pathy associa ted
with chronic PPS Intake.
ties. Retinal changes continued to
develop after cessation of the drug. Cumulativ e exposure
of 1,300 g.

Case Series
Willia m Pear ce, 2018
To describ e the clinical feature
s of maculo pathy
in the setting of chronic exposu
re to PPS,
38
38
6
N/A N/A
N/A
Patients develope d changes
in the macula and RPE. Mean cumulativ e exposure
of 2,263 g.

Case Series
Jenel
le
Foot
e,
2019
To evaluat e charact eristics of patient s with PPS maculo pathy.
166+ Not Repor ted
10
156 0
N/A
10 patients with self reported PPS use develope
dretinal toxicity. 156 unexpose
dpatients with

 

22
IC/BPS did not develop toxicity. Mean cumulativ e exposure of 2062 g.

Case Series
Aviel
Hada
d,
2019
To
draw
mathe matical correlat ions betwee n PPS exposu re and morpho logic change s in the retina.
17 17 17 N/A N/A N/A All patients had findings
of retinal toxicity; seemingl
y related to dose amount. Affected patients
had a cumulativ e exposure of at least 20 g per
kg of body weight.

Retros pective Cohort Study.
Ada
m
Hanif
,
2019
To evaluat
ethe risk factors for develo pment of maculo pathy among patient s diagno sed
219 80 14 139 0 N/A PPS was found to
be the sole predictor
for the presence
of an unspecifi ed pigmentar y maculopa thy. Cumulativ
edose of

 

23
with IC. 2,300 g.

Multi- instituti onal Case series
Ada
m
Hanif
,
2019
To charact erize the exposu re charact eristics of PPS–
associa ted maculo pathy.
404
404
35
N/A N/A
N/A
Symptom atic patients found to have
RPE changes. Mean cumulativ e exposure of 1,610 g.

Case Control Study
Alex
ande
r
Barn
es,
2020
To evaluat e whethe
rPPS maculo pathy can be differen tiated from other maculo pathies
1,131 Not Repor ted
10
Not
Rep
orte
d
Not Report ed
N/A
PPS maculopa thy was reliably differentia ted from other maculopa
thies by imaging alone.

Cross Sectio nal Study
Robi n A. Vora 2020
To evaluat e prevale nce
and
risk factors for maculo pathy
in patient
swith long- term exposu
117
117
27
N/A N/A
N/A
One quarter of patients with PPS intake
>500 g develope
dretinal changes.

 

24
re to PPS

Cross Sectio nal Study
Derri
ck
Wan
g,
2020
To describ
ethe prevale nce
and spectru m of multim odal imagin g finding
s and recom mend screeni ng guideli nes
50 50 10 N/A N/A N/A Affected group was notable for
significant ly longer exposure s and higher dosing of PPS compared
to the unaffecte d
patients. 3375.4 g mean cumulativ e dose in the affected group.

Retros pective Match ed Cohort Study
Niera
j
Jain,
2019
Determ ine if an associa tion exists betwee n PPS use
and macula r
diseas e
18,07 2 at 5 years follow up

9,621 at 7 years follow up
3012 at 5 years follow up

1604 at 7 years follow up
103 at 5 years follow up

87 at 7 years follow up
15,0
60
at 5 year s follo w up

801 7 at 7 year s follo w up
440 at 5 years follow up

328 at 7 years follow up
5- and 7- years retros pectiv e
follow- up.
Significan t associatio n
between PPS exposure and a new diagnosis of
macular disease
at 7 years.

Retros pective cohort study
Cass
ie
Ludw
ig,
2019
To determi ne the associa tion
49,89
9
11,68
0
277
38,2
19
1,058 5 years retros pectiv e
An increase in hazard ratio is
found in

 

25

 

and cumula tive dose respon se pattern betwee n PPS and Maculo pathy
follow up.
PPS for the diagnosis of hereditary dystrophy
. However,
it is not statisticall y significant
.

Exclud ed Studie s Reporti ng on Primar y Data
NON
E
N/A
N/A
N/A
N/A
N/A N/A
N/A
N/A
Table 1: Literature reporting on pentosan polysulfate maculopathy. Search methodology: Pentosan polysulfate maculopathy, pentosan polysulfate retinopathy. Databases: PubMed, Medline, OVID, EBSCO, Google Scholar, by 2 reviewers, all languages. N/A= Not Applicable, g=grams, RPE= Retinal Pigment Epithelium. IC/BPS= Interstitial Cystitis/ Bladder Pain Syndrome. PPS= Pentosan Polysulfate Sodium.

 

26

 

 

Type
Paper
of
Author and Year
Newcastle Ottawa Scale
Risk of Bias
Conflicts
of Interest Identified

Case Report Robin A. Vora, 2020
N/A
Overall Risk of Bias: High
No

Case Report Maxwell Wingelar, 2020
N/A
Overall Risk of Bias: High
No

Case Report Kapil Mishra, 2020
N/A
Overall Risk of Bias: High
No

Case Report Rachel Huckfeldt, 2019
N/A
Overall Risk of Bias: High
No

Case Series
William Pearce, 2018
N/A
Overall Risk of Bias: High
No

Case Series
Jenelle Foote, 2019
N/A
Overall Risk of Bias: High
No

Case Series
Aviel Hadad, 2019
N/A
Overall Risk of Bias: High
No

Multi- institutional Case series
Adam Hanif, 2019
N/A
Overall Risk of Bias: High
No

Cross Sectional Study
Robin A. Vora 2020
N/A
- Randomization process: High
- Missing outcome data: Low
- Deviations from intended interventions: N/A
- Measurement of the outcome: Low
- Selection of the reported result: High
No

Cross Sectional Study
Derrick Wang, 2020
N/A
- Randomization process: High
- Missing outcome data: Low
- Deviations from intended interventions: N/A
- Measurement of
No

 

27
the outcome: Low
- Selection of the reported result: Low

Retrospective Cohort
Adam Hanif, 2019
Selection:**** Comparability:** Outcome:***
- Randomization process: Low
- Missing outcome data: Low
- Deviations from intended interventions: N/A
- Measurement of the outcome: Low
- Selection of the reported result: High
No

Case Control Study
Alexander Barnes, 2020
Selection: * Comparability:* Exposure:**
- Randomization process: Low
- Missing outcome data: Low
- Deviations from intended interventions: N/A
- Measurement of the outcome: High
- Selection of the reported result: Low
No

Retrospective Matched Cohort Study
Nieraj Jain, 2019
Selection:**** Comparability:** Outcome:***
- Randomization process: Low
- Missing outcome data: Low
- Deviations from intended interventions: N/A
- Measurement of the outcome: Some Concerns
- Selection of the reported result: High
No

Retrospective cohort study
Cassie Ludwig, 2019
Selection:**** Comparability:** Outcome:**
- Randomization process: Low
- Missing outcome data: Low
No

 

28
- Deviations from intended interventions: N/A
- Measurement of the outcome: High
- Selection of the reported result: High

Table 2: Table assessing each paper’s risk of bias and study quality. Risk of Bias for each study was assessed using the “RoB 2: A revised Cochrane risk of bias tool for randomized trials”. Study quality was assessed using the Newcastle Ottawa scale
where applicable (cohort and case control studies). Conflicts of interests were also assessed. Note: All case reports and case series received a high overall risk of bias due to the nature of that type of publications.

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>