Next Article in Journal
Prioritized SNP Selection from Whole-Genome Sequencing Improves Genomic Prediction Accuracy in Sturgeons Using Linear and Machine Learning Models
Previous Article in Journal
Review of Recent Medicinal Applications of Rhenium(I) Tricarbonyl Complexes
Previous Article in Special Issue
Variable Ophthalmologic Phenotypes Associated with Biallelic Loss-of-Function Variants in POMGNT1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Stargardt’s Disease: Molecular Pathogenesis and Current Therapeutic Landscape

1
Ocugen India, 5th Floor, AYDIV IT Park, Financial District, Nanakramguda, Hyderabad 500032, Telangana, India
2
Ocugen, 11 Great Valley Parkway, Malvern, PA 19355, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(14), 7006; https://doi.org/10.3390/ijms26147006
Submission received: 28 May 2025 / Revised: 7 July 2025 / Accepted: 16 July 2025 / Published: 21 July 2025
(This article belongs to the Special Issue Molecular Research in Retinal Degeneration)

Abstract

Stargardt’s disease (STGD1) is an autosomal recessive juvenile macular degeneration caused by mutations in the ABCA4 gene, impairing clearance of toxic retinoid byproducts in the retinal pigment epithelium (RPE). This leads to lipofuscin accumulation, oxidative stress, photoreceptor degeneration, and central vision loss. Over 1200 pathogenic/likely pathogenic ABCA4 variants highlight the genetic heterogeneity of STGD1, which manifests as progressive central vision loss, with phenotype influenced by deep intronic variants, modifier genes, and environmental factors like light exposure. ABCA4 variants also show variable penetrance and geographical prevalence. With no approved treatment, investigational therapies target different aspects of disease pathology. Small-molecule therapies target vitamin A dimerization (e.g., ALK-001), inhibit lipofuscin accumulation (e.g., soraprazan), or modulate the visual cycle (e.g., emixustat hydrochloride). Gene therapy trials explore ABCA4 supplementation including strategies like RNA exon editing (ACDN-01) and bioengineered ambient light-activated OPSIN. RORA gene therapy (Phase 2/3) addresses oxidative stress, inflammation, lipid metabolism, and complement system dysregulation. Trials like DRAGON (Phase 3, tinlarebant), STARLIGHT (phase 2, bioengineered OPSIN) show promise, but optimizing efficacy remains challenging. With the key problem of establishing genotype–phenotype correlations, the future of STGD1 therapy may rely on approaches targeting oxidative stress, lipid metabolism, inflammation, complement regulation, and genetic repair.

1. Introduction

Stargardt’s disease, first described by German ophthalmologist Karl Stargardt in 1909 [1], is a type of juvenile-onset macular degradation [2]. It typically presents in the first or second decade of life with progressive central vision loss and characteristic white flecks in the retinal pigment epithelium (RPE) that cover the macula, and their progressive degeneration [3,4]. Activities requiring sharp vision, like reading and identifying faces, are significantly impaired, while peripheral vision often remains mostly intact [4]. It is a heritable disorder and the ABCA4 gene, expressed at high levels in the rod photoreceptors and encoding ATP-binding cassette (ABC) transporter, has been identified as the causal gene for Stargardt’s disease and shows an autosomal recessive inheritance [5,6]. Stargardt-like disease, Stargardt 3 and Stargardt 4, are retinopathies with similar phenotype but with autosomal dominant inheritance associated with variants in ELOV4 [7] and PROM1 [8] genes, respectively. In this review, we have considered the Stargardt 1 disease, and it has been referred to as Stargardt’s disease throughout.
Robust data on the prevalence of Stargardt’s disease is lacking. A genomic database study, published in 2020, estimated the genetic prevalence of inherited retinal diseases (IRDs). The study indicated that more than one-third of the world population consists of healthy carriers of one mutation that can cause an autosomal recessive IRD. Among the 5.5 million people expected to be affected by an autosomal recessive IRD based on their genotype, about 1.98 million (33%) would have Stargardt’s disease genotype [9]. This gives an estimated prevalence of 1 in 4000 for Stargardt’s disease genotype (including late-onset disease). However, regional variations in prevalence are expected. The prevalence of Stargardt’s disease was observed to be 1 in 16,000 in Israel for 2023 [10] and 1 in 22,000 in The Netherlands for 2018 [11]. In the European Union, the prevalence of Stargardt’s disease has been reported as 1 in 10,000 in 2019 [12].
A female specific predisposition has also been observed for overall patients [13], patients aged 10–19 years [11], and for patients having mild ABCA4 allele [14] suggesting that sex may be considered as a disease-modifying variable.
Despite the quantum of research on Stargardt’s disease, particularly the extensive studies on the disease associated variants present in the ABCA4 gene, an approved therapy for this disease is lacking and promising therapy remains elusive.
Herein, we describe the etiology, pathophysiology, and epidemiology of Stargardt’s disease and dwell on the current therapeutic landscape of the disease while taking a deeper look on the candidate drugs and therapies evaluated in clinical trials.

1.1. The Visual Cycle and ABCA4 Protein

The gene product of ABCA4 gene, the ABCA4 protein, is the photoreceptor specific flippase known as ATP-binding cassette subfamily A member 4, and it is a crucial component in the retina, specifically in photoreceptor cells [15]. This protein belongs to the ATP-binding cassette (ABC) transporter superfamily, a vast group of proteins that harness ATP hydrolysis to transport different molecules across cellular membranes. The designation “A4” signifies that it is the fourth gene in the “A” subfamily within this superfamily [16].
The ABCA4, a 2273 amino acid long protein, resides in the outer segments of photoreceptor cells, actively transporting vitamin A derivatives across the disc membranes of these segments following transduction of visual signal by photoreceptors [6]. ABCA4 protein has two nucleotide-binding domains (NBDs) and two extracellular domains (ECDs) [17]. The NBD-1 and NBD-2 each contain an ABC transporter region [18]. A terminal sequence essential for ATP binding and ATPase activity has also been described [19]. Various domains of ABCA4 have been shown in Figure 1A.
The human retina contains rods and cones photoreceptors, with rods highly sensitive to dim light and cones responsible for color vision and bright light detection [20]. In the rod and cone photoreceptors, the outer segment is a specialized cellular compartment responsible for detecting light and converting it into electrical signals through phototransduction. This segment contains a well-organized stack of disc membranes, which are densely packed with visual pigments rhodopsin or cone opsin [21,22]. Opsin, when covalently bound to 11-cis-retinal (a vitamin A derivative) via a Schiff base, is photoactive and referred to as rhodopsin in rods and cone opsin in cones. Upon photon absorption, 11-cis-retinal isomerizes to all-trans-retinal, triggering a series of conformational changes in opsin to its active state. The pigment is termed as “bleached” when it undergoes hydrolysis into opsin and all-trans-retinal and is regenerated when opsin recombines with 11-cis-retinal completing the visual cycle [23].
ABCA4 localizes to the rim region and incisures of the retinal outer segment [24]. The all-trans-retinal, produced after the bleaching of rhodopsin, binds to phosphatidyl ethanolamine (PE) to form N-trans-retinylidene-phosphatidylethanolamine (NRPE) [25]. ABCA4 acts as a retinoid transporter moving NRPE, the Schiff-base adduct of retinal (all-trans-retinal) and phosphatidylethanolamine (PE), from the lumen to the cytoplasmic side of outer segment disc membranes of photoreceptors [25,26]. In the cytosolic side, all-trans retinal gets converted into all-trans-retinol by the action of retinyl dehydrogenase, which then binds to interphotoreceptor retinoid binding protein (IRBP), also known as retinol binding protein-3 (RBP3), and passes through the interphotoreceptor matrix to the retinal pigment epithelium cells [27]. Again, in the retinal pigment epithelium cells, all-trans-retinol undergoes a series of biochemical changes to eventually get converted into 11-cis-retinal [25,28]. Cellular Retinol Binding Protein (CRBP) binds the all-trans-retinol in retinal pigment epithelium cells. Subsequently, the conversion of all-trans-retinol to 11-cis-retinol occurs, which, in turn, binds to Cellular Retinaldehyde Binding Protein (CRALBP) and gets oxidized to 11-cis-retinal. The 11-cis-retinal moves to the interphotoreceptor matrix, binds to IRBP, and reaches the photoreceptor outer segment [27]. This recycled 11-cis-retinal binds to PE and enters the lumen of photoreceptor cells via ABCA4, making it available to create fresh rhodopsin and thus complete the visual cycle [28]. The retinal pigment epithelium cells also convert 11-cis retinol to all-trans-retinyl ester, which acts as a reservoir to store the retinol [29]. These events are depicted in Figure 2.

1.2. Etiology of Stargardt’s Disease

Insufficiency of ABCA4, as observed in Stargardt’s disease, leads to the accumulation of NRPE in the outer segment of photoreceptors. After the outer segments are phagocytosed by retinal pigment epithelium cells, the NRPE is hydrolyzed to N-retinylidene-N-retinylethanolamine, a component of lipofuscin, and phosphatidic acid [30]. Bisretinoids, like NRPE, also undergo photooxidation followed by degradation [31]. Progressive buildup of lipofuscin in retinal pigment epithelium cells leads to degradation of the photoreceptors and subsequent loss in central vision (Figure 3). Vitamin A byproducts may also cause delayed dark adaptation by competing with vitamin A in binding to RPE65 and retinoic acid receptor [32].
As per a comprehensive study published in November 2023, 796 pathogenic, 452 likely pathogenic, and 971 ABCA4 variants of uncertain significance were identified associated with Stargardt’s disease [33]. Another recent database study analyzing the ABCA4 gene variants in about 11,000 patients having inheritable retinal disease identified around 1200 unique variants in the ABCA4 gene that were pathogenic or likely pathogenic. Inheritance of Stargardt’s disease is autosomal recessive and compound heterozygous for Stargardt’s disease [34]. Pathogenic variants have also been observed on single allele in patients with Stargardt’s disease. Further analysis revealed deep intronic variants in some of these cases [35]. The variants in ABCA4 gene are spread across the domains of ABCA4 protein [36]. In a German multicenter cohort study of 335 patients with Stargardt’s disease, 148 mutations in the ABCA4 gene that are likely to impact disease pathology have been identified. About half of the patients (47.2%) had ≥2 mutations in the ABCA4 gene, and 45.1% had mutations that were pathogenic or likely pathogenic. Single ABCA4 mutation was observed in 28.4% of patients [37]. Similarly, in a study involving 133 Spanish patients with Stargardt’s disease, 48.1% reported ≥2 mutations and 24.8% reported single mutations in the ABCA4 gene [38].
No mutational hotspots have been observed in the ABCA4 protein and mutations are observed in the protein across various functional domains. This underscores the complex heterogeneity in the correlation of genotype to phenotype in Stargardt’s disease. Table 1 and Figure 1B highlight the frequent mutations observed in ≥5% of patients with Stargardt’s disease across studies.
A recent study has demonstrated the effect of specific ABCA4 variants on disease progression. Square root-transformed atrophy growth, assessed by definitely decreased autofluorescence (DDAF), was slower in patients with the 5882G>A (0.0821 mm/year) and 4539+2001G>A (0.0686 mm/year) variants compared to those with 768G>T (0.1299 mm/year) and 5461-10T>C (0.1565 mm/year) [43]. In another recent study, the 5461-10T>C variant induced exon skipping in Stargardt’s disease patients and was found in compound heterozygous with G1961E which is a hypomorphic variant [44].
Various studies have reported deep intronic variants in patients with Stargardt’s disease with frequency ranging from 2% to 18% in the ABCA4 gene [36,37,45,46,47,48]. This highlights the potential of these variants in disease etiology, which is further underlined by the fact that these variants are not detected in the typical variant screening involving exons. A total of 353 deep intronic variants in ABCA4 gene have been identified in patients with Stargardt’s disease [49]. A multinational study involving 1054 probands with Stargardt’s disease and Stargardt’s like disease identified c.4253+43G>A (n = 100), c.4539+2001G>A (n = 64), and c.5196+1137G>A (n = 47) as the three most common deep intronic variants [49]. A recent study in a Chinese cohort of patients with Stargardt’s disease has shown that 45% of patients with no variants in exon region have deep intronic variants in the ABCA4 gene. Interestingly, the study also showed that such patients having deep intronic variants displayed a milder disease phenotype as compared to patients having biallelic variants in the exon region of the ABCA4 gene [50]. Deep intronic variants have also been reported to occur in trans with other variants occurring in a single allele [37]. The deep intronic variant c.769-784C>T has been found to be act as a modifier variant for G1961E variant, partly explaining the low penetrance of this variant [51] exemplified with the fact that its prevalence in normal population can be as high as 20% in west Africa [52]. The disease association of deep intronic variants warrants further studies with deep sequence analysis of larger number of patients.
Hypomorphic variants in ABCA4 have been associated with distinct disease phenotype. In a European cohort study in patients with Stargardt’s disease, the allele c.5603A>T (N1868I) was associated with majority (~80%) of late-onset cases and was determined as hypomorphic variant. The phenotype associated with this variant, late-onset disease with foveal sparring, was expressed only when this variant occurred in trans conformation with a deleterious mutation [53]. It is noteworthy that a deep intronic variant, c.4253+43G>A, has also been identified as an hypomorphic variant. This variant was detected in 2.6% of patients with Stargardt’s disease and was significantly enriched (14.4%) in patients having only one pathogenic allele. The variant, when present in trans with a loss-of-function ABCA4 variant, was associated with late symptom onset and foveal sparring Stargardt’s disease [54].
Mutations in Bestrophin-1 (BEST1) and Crumbs homolog 1 (CRB1) genes have also been reported in patients with Stargardt’s disease with wildtype ABCA4 gene [55].
The root cause of Stargardt’s disease remains mutations in ABCA4 gene, but the complete manifestation of disease pathology is likely to be affected by alteration in molecular pathways resulting from the lack of a functional ABCA4. This notion is supported by the fact that the Stargardt’s disease has juvenile onset. Data supporting this theory are shown in the preclinical studies described below.

1.2.1. Role of Complement System

Dysregulation of the complement system has been observed in the outer retina of eyes of patients with Stargardt’s disease [56]. On the other hand, when the complement negative regulatory protein, complement receptor 1-like protein y (CRRY), is expressed in the retinal pigment epithelium cells using gene therapy in mice lacking ABCA4, there is a rescue of Stargardt’s disease phenotype. These mice expressing CRRY show a 2-fold reduction in bisretinoid levels [57]. Photooxidatin of NRPE, a component of lipofuscin, has also been shown to activate the complement C3 in in vitro studies [58,59]. Hence, Stargardt’s is associated with a dysregulation of the complement system on one hand, and checking the aberrant complement activation reduces the disease phenotype. These preclinical observations suggest a potential role of complement activation in the pathophysiology of Stargardt’s disease (Figure 3).

1.2.2. Role of Oxidative Stress

As discussed above, retinal pigment epithelium cells phagocytose the outer segments of photoreceptors. An in vitro study involving cultured retinal pigment epithelium cells showed that the phagocytosis results in increased production of reactive oxygen species in the retinal pigment epithelium cells and the enzyme catalase protect these cells from the increased oxidative stress [60]. Reactive oxygen species are also sourced exogenously in retinal pigment epithelium cells due to exposure to visible light and high oxygen tension (70 mm Hg) [61]. The pigment melanin has been described as an antioxidant that protects retinal pigment epithelium cells from oxidative damage [62]. In mouse-based models of Stargardt’s disease, oxidative stress has been observed, which gets augmented in the absence of melanin. Mice lacking ABCA4 in the albino background showed increased oxidation of N-retinylidene-N-retinylethanolamine, which was checked by inhibition of rhodopsin regeneration. Although the levels of N-retinylidene-N-retinylethanolamine were not higher in ABCA knock out mice lacking melanin (albino) as compared ABCA4 knockout mice with melanin, the presence of increased oxidation of retinoid in albino mice indicates the role of light exposure in Stargardt’s pathology [63]. On similar lines, increased light exposure did not increase the lipofuscin formation per se but increased light-dependent oxidation of N-retinylidene-N-retinylethanolamine [64]. In the photoreceptors of albino mice lacking ABCA4, shortening of outer segment, disordering of disk membranes, damaging of inner segments, and morphological changes at the base of outer segments were observed. These changes were not observed in mice lacking ABCA4 but having intact melanin production [63]. These observations further highlight the potential role of increased oxidative stress in augmenting Stargardt’s disease pathology (Figure 3).

1.3. Stargardt’s Disease: Pathophysiology and Clinical Presentation

The macula is a small, round area located at the center of the retina, measuring about 5 mm in diameter. Despite its small size, it plays a crucial role in central vision, allowing you to focus on details like text, faces, and colors. The macula contains a high concentration of cone photoreceptors that are responsible for processing these details, while rods, found in the peripheral retina, aid with peripheral and night vision [65]. High expression of ABCA4 protein has been observed in the retina [66]. At the core of macula is the fovea, which is densely packed with cone photoreceptors and plays a vital role in tasks such as reading and facial recognition. In Stargardt’s disease, accumulation bisretinoid derivatives lead to lipofuscin accumulation in the fovea, which is particularly harmful due to the high density of photoreceptors and the region’s key role in detailed vision. Lipofuscin fluorescence has been observed to be up to three folds higher in patients with Stargardt’s disease as compared to healthy controls [51]. Lipofuscin buildup leads to the formation of hyperreflective debris on the apical side of retinal pigment epithelial cells seen as flecks. These flecks predominantly emit short-wave autofluorescence, and imaging studies have shown the associated loss of retinal pigment epithelium cells and corresponding thinning of outer nuclear layer in patients with Stargardt’s disease [67]. Consequently, the buildup of lipofuscin in the retinal pigment epithelium leads to damage to photoreceptors, causing the characteristic loss of central vision [68]. The clinical features of Stargardt disease involve progressive central vision loss, photophobia, and impaired color vision, often starting in childhood or adolescence. Fundus examination typically reveals characteristic yellow flecks and macular atrophy, while electrophysiological testing shows reduced ERG responses. The disease progresses slowly, with eventual total loss of central vision in severe cases. However, peripheral vision is often preserved for a long time, and visual prognosis can vary significantly based on disease progression and other factors [69]. With the gradual and progressive loss of central vision in Stargardt’s disease, a visual acuity ranging from 20/40 to 20/400 has been observed in patients [70].
As discussed earlier, numerous mutations in the ABCA4 gene have been associated with the Stargardt’ disease, highlighting the genetic heterogeneity which translates into the heterogeneity of disease onset and progression. Siblings carrying the same ABCA4 variant have been reported to display phenotypic discordance, showing substantial differences in disease progression including age at onset, visual acuity loss, and time to severe visual impairment [69,71].
The age of disease onset has been described as an important marker for disease severity. Early-onset Stargardt’s disease, occurring in first or second decade of life, has been described as a more severe disease with rapid decline of visual activity with progressive retinal degradation [72]. Milder disease phenotype has been observed with greater proportions in patients with late-onset disease (mean ages around 40 years). These patients were more likely to have hypomorphic alleles, while the severe phenotypes were associated with early-onset Stargardt’s disease (mean age around 17 years) [73]. Late-onset Stargardt’s disease has also been associated with foveal sparring. In a study involving 103 patients having Stargardt’s disease from The Netherlands with an age of onset of ≥45 years, about one-third of the patients displayed foveal sparring and a median 15.4 years passed from first diagnosis to foveal involvement [74]. Late-onset Stargardt’s disease can be misdiagnosed as age-related macular degeneration with one study reporting a misdiagnosis rate of 22% [11]. Phenotypic discordance, primarily affected by age and, as per some reports, by gender, highlights complex nature of genotype–phenotype relation of this disease.

1.4. Stargardt’s Disease: Diagnosis

Clinical features of Stargardt’s disease overlap with other retinopathies, making its careful diagnosis critical to avoid misdiagnosis. The diagnosis involves clinical examinations and imaging techniques described in brief below.
The clinical examination includes visual acuity testing and ophthalmoscopy.
  • Visual acuity test: This test determines a patient’s ability to distinguish between two standard symbols or alphabets while viewing them from a standard distance (20 feet for US; 6 feet for UK). A visual acuity of x/y indicates that the patient can recognize symbols from a distance of x, whereas individuals with normal vision can identify the same symbols from a distance of y [75]. An age-related decline in the probability of maintaining a visual acuity of 20/40 has been observed in patients with Stargardt’s disease. A faster decline in visual acuity has also been reported in these patients after it dropped further from 20/40 and stabilized at 20/200 [76].
  • Ophthalmoscopy: It is a clinical assessment to observe the internal structures of the eyes, specifically the fundus, of the patient through the dilated pupil using a ophthalmoscope [77]. The characteristic fundus appearance of Stargardt disease includes a macula with “beaten bronze” look, surrounded by distinct yellow flecks, either round or pisciform, located at the level of the retinal pigment epithelium [78].
Majorly, the key imaging techniques used in the diagnosis and prognostic monitoring of Stargardt’s disease are
  • Fundus autofluorescence (FAF): This imaging technique detects the autofluorescence caused due to lipofuscin deposition in retinal pigment epithelium cells [79,80]. In the ProgStar study, which is a multinational, observational cohort study involving patients with Stargardt’s disease, around 250 patients each were analyzed retrospectively and prospectively with FAF as the primary outcome measure [81].
  • Optical coherence tomography (OCT) and Spectral-domain OCT (SD-OCT): OCT uses light in the near-infrared region to examine the tissue. The delay in the reflected light provides a measure of the depth of reflection that determines its axial resolution. SD-OCT is an improved version of OCT with increased speed and image quality [82]. SD-OCT reveals outer retinal atrophy, including loss of the ellipsoid zone and thinning of the retinal pigment epithelium [83].
  • Electroretinography (ERG): It can aid in the diagnosis and monitoring of Stargardt’s disease, but it is suggested to avoid these techniques as it involves bright light flashes that can exacerbate lipofuscin formation [84].
In 1976, Fishman classified Stargardt’s disease phenotypes on the basis of ophthalmoscopy [85]. Ophthalmoscopy imaging has the lacuna of not detecting the disease in early stages. In a study analyzing childhood-onset Stargardt’s disease, about one-fourth of patients showed no clinically detectable retinal lesions assessed by ophthalmoscopy at the time of presentation [72].
Three clinical phenotypes of Stargardt’s disease have been described based on FAF by Fujinami, et al., 2013 [86].
  • Type 1: A localized low FAF signal at the fovea, set against a uniform background, with or without perifoveal foci of increased or decreased signal.
  • Type 2: A localized low FAF signal at the macula, surrounded by a varied background, with widespread foci of increased or decreased FAF signal extending beyond the vascular arcades.
  • Type 3: Multiple areas of low FAF signal at the posterior pole, accompanied by a heterogeneous background and/or foci of increased or decreased signal.
These phenotypes differed in the rate of atrophy enlargement with disease severity increasing from type 1 to 3, highlighting the fact that patients who exhibit localized foveal atrophy tend to experience slower progression of central atrophy, whereas those with multiple atrophic lesions show a more rapid decline in central retinal structure over time [86].
FAF can be used for differential diagnosis of Stargardt’s disease along with multiple hereditary retinal disorders in children [87], Table 2.

1.5. Stargardt’s Disease: Therapeutic Landscape

No approved therapy is available for Stargardt’s disease at present. The clinical trial landscape for Stargardt’s disease is majorly dominated by small molecules followed by gene therapy and stem cell therapy. Trials that study small molecules, gene therapy, and stem cell therapy for Stargardt’s disease are listed in Table 3. To evaluate efficacy, the trials include structural assessments such as changes in lesion size, as well as functional assessments such as changes in visual acuity and retinal sensitivity.

1.5.1. Small Molecules for Stargardt’s Disease

Tinlarebant is an orally administered retinal binding protein-4 (RBP4) antagonist [88]. RBP4 is responsible for delivery of retinol from liver to extrahepatic tissues including the eye [89]. The rationale behind using tinlarebant is to reduce the serum RBP-4, which would lead to a subsequent reduction in levels of retinol in the retina, thereby checking the levels of bisretinoids [88]. Tinlarebant has received a fast-track designation from the FDA in May 2022 [90] and received the Sakigake designation, which expedites the approval for innovative treatments targeting serious ailments, in Japan in June 2024. This designation offers prioritized consultation, pre-application support, expedited review, dedicated review partners, and extended re-examination periods for expediting the approval process [91]. The 2-year phase 2 trial for tinlarebant showed that patients receiving the drug showed sustained lower lesion growth as compared to patients enrolled in ProgStar trial having similar baseline characteristics (p = 0.001) [92]. The pivotal global phase 3 DRAGON trial has showed that tinlarebant is well-tolerated and has a consistent safety profile with stabilization of visual acuity in the 1-year interim analysis [93].
Similar to tinlarebant’s approach of reducing RBP-4 activity, STG-001 reduces the plasma levels of RBP-4, leading to reduced levels of vitamin A in retina [94]. It received an orphan drug designation in April 2019 from US FDA [95]. A phase 2a study comparing two doses of STG-001 and studying safety, tolerability, PK, and reduction in plasma RBP-4 levels was completed in April 2021 [96], Table 3. Subsequent trials for STG-001 have not been reported.
Metformin has been shown to reduce the levels of N-retinylidene-N-retinylethanolamine in the retina and retinal pigment epithelium/choroid of mice that were lacking ABCA4 gene [97]. A later study showed that metformin rescues the intra-cellular lipid accumulation by targeting lysosomal and fatty acid oxidation pathways in retinal pigment epithelium cells derived from induced pluripotent cells obtained from ABCA4 knockout mice [98]. The NEI is conducting a clinical trial to assess the safety and efficacy of oral metformin in slowing the progression of Stargardt disease. Participants aged 12 and older will take metformin for 24 months, with evaluations over at least 36 months [99].
Reducing the dimerization of vitamin A, which eventually leads to the formation of bisretinoids, is also a promising therapeutic approach. ALK001 is a vitamin A molecule with hydrogen at C20 position replaced by deuterium. It has shown reduced formation of vitamin A dimers in wildtype mice and rats [100]. A phase 2 trial assessing safety and tolerability of ALK-001 along with PK and lesion size and its extension trial (NCT4239625) assessing safety, tolerability, and PK is ongoing (Table 2).
Modulating the visual cycle to reduce the availability of vitamin A derivatives that subsequently form lipofuscin is another approach followed in the case of emixustat hydrochloride. This drug inhibits retinal pigment epithelium protein 65 (RPE65), which is responsible for an intermediate step in the molecular pathway converting all-trans retinal to 11-cis-retinal [101]. Emixustat received an orphan drug designation from the US FDA in January 2017 [102]. A phase 2 trial for emixustat, which was completed in 2017, tested its pharmacodynamics and safety in 23 patients with Stargardt’s disease [103]. This trail was followed by a phase 3 trial involving 194 patients with Stargardt’s disease which, upon completion in June 2022, showed a mean rate of change in macular atrophic area as 1.28 mm2/year in patients receiving oral emixustat (placebo: 1.31 mm2/year) at the end of 24 months [104]. A post hoc analysis of the same trial results including only the patients having smaller lesion size at baseline showed a 40.8% reduction in the progression of atrophic lesion with emixustat as compared to placebo (p = 0.02) [105]. No further trials have been reported for emixustat.
Soraprazan is a reversible inhibitor of H+/K+ ATPase. It has been shown to remove existing lipofuscin in monkeys [106,107]. A single intravitreal injection of soraprazan reduced lipofuscin in retinal pigment epithelium cells without toxicity. It also preserved photoreceptors in aged mice. The drug specifically accumulated in retinal pigment epithelium pigment granules, not in choroidal melanocytes, supporting its potential as a treatment [108]. Unlike ALK-001, which prevents additional vitamin A aggregates eventually leading to lipofuscin formation, soraprazan can also reduce the existing lipofuscin [106]. Soraprazan was given the orphan drug status by FDA in 2017 [109]. A double-blinded randomized phase 2 trial assessing lipofuscin by quantitative autofluorescence was started in 2019 in The Netherlands. As of the last update in April 2024, the trial status is still “recruiting” [110]. No further trials have been reported for soraprazan.
Dietary supplementation of omega-3 fatty acids and particularly DHA/EPA is another therapeutic approach that is being tried in clinical studies for Stargardt’s disease (Table 3). In mice lacking ABCA4, supplementation with omega-3 fatty acids adjusted to achieve the arachidonic acid to eicosapentaenoic acid ratio between 1 and 1.5, reduced the levels of N-retinylidene-N-retinylethanolamine approximately fourfold compared to mice not receiving the supplement (p < 0.05). Preservation of the outer nuclear layer (p < 0.01) and a reduction in the C3 complement system protein (p < 0.05) was also observed with omega-3 fatty acid supplementation [111]. The phase 2 trial testing omega-3 fatty acid supplementation in patients (N = 21) with Stargardt’s disease showed that, at 24 weeks of supplementation, there was an increase in best-corrected visual acuity (p = 0.003) and a higher score for perceived vision and subjective mood as compared to placebo [112].
As discussed above, complement system activation is a potential contributor to Stargardt’s disease etiology. A small-molecule inhibitor of complement C5 is being tested in a randomized phase 2b trial (Table 3). This global trial is active with 121 patients who would be given intravitreous injection of the study drug avacincaptad pegol [113].

1.5.2. Gene Therapy for Stargardt’s Disease

Two kinds of gene therapy are under clinical trials for Stargardt’s disease, those supplementing wildtype ABCA4, and those aiming to modify the disease by expressing other molecules. Three phase 1/2 trials evaluating the safety and preliminary efficacy of gene therapy supplementing wildtype ABCA4 are ongoing.
ACDN-01 is an RNA exon editor delivered as a DNA construct via adeno-associated viral (AAV) vector. Once it is transcribed in the cells, ACDN-01 binds to the mutant ABCA4 pre-mRNA, via a highly specific binding domain, resulting in replacing the mutant RNA with the wildtype RNA [114]. This drug was given a fast-track designation by the FDA in January 2024 [115]. The organization developing the drug has claimed that ACDN-01 has shown durable and efficient RNA exon editing in human retinal explants and in primates [115]. A phase1/2 open-label dose ascending trial is ongoing to assess the safety and tolerability of this ACDN-01 delivered via a single subretinal injection [116].
Another AAV-based gene therapy, JWK006, which expresses ABCA4 gene, is being assessed for safety and visual acuity in an ongoing phase 1/2 trial. It is administered via subretinal route [117].
A different gene therapy approach is followed in the case of virally carried ambient-light activatable multi-characteristic opsin (vMCO-010), also known as Sonpiretigene isteparvovec. It consists of an AAV-based expression of three opsins with separate activation peaks [118]. The protein product, multi-characteristic opsin, is a bioengineered fusion protein that is highly sensitive to light with broad spectrum responsiveness and faster kinetics to activate retinal cells in expressing the vMCO-001 in ambient light [119]. Outcomes at 24 weeks from the phase 2 trial, STARLIGHT, evaluating the safety and effectiveness of intravitreal injection of vMCO-010 in 6 patients with Stargardt’s disease, show favorable safety and tolerability. There was a gain of five points as per the Early Treatment Diabetic Retinopathy Study (ETDRS) chart in visual acuity and a three decibel gain in visual field in overall patients [119].
Another ABCA4 agnostic gene therapy approach is to express retinoic acid-related orphan receptor alpha (RORA), which is a nuclear hormone receptor. In the context of Stargardt’s disease, RORA regulates inflammatory response pathways that include the proteins CD59. It gets downregulated in Stargardt’s disease and is an inhibitor of the membrane attack complex (MAC) assembled with the help of complement C3 and C5, and ABCA4. RORA expression restores the expression of CD59 in mice lacking ABCA4 to levels that are comparable to wildtype mice [120]. As discussed above, dysregulation of a complement system has been implicated in Stargardt’s disease [56]. In mice lacking ABCA4, a rescue of Stargardt’s disease phenotype was observed in mice treated with AAV5 expressing human RORA (hRORA). Lipofuscin deposits, assessed by blue autofluorescence intensity, were significantly reduced upon RORA expression. The electroretinogram showed improvement in the scotopic b-wave amplitude and increased peak percentage recovery of baseline a-wave amplitude after photobleaching [120]. The role of oxidative stress in retina in augmenting Stargardt’s disease has also been discussed above. RORA protects from oxidative stress in cardiac fibroblast [121] and in cortical neurons [122]. A phase 1/2 study is being conducted to assess the safety and efficacy of OCU410, which uses an AAV5-based vector expressing hRORA, in patients with Stargardt’s disease. The trial is expected to be completed in October 2025 [123].
Table 3. Clinical trials for Stargardt’s disease using small molecules, gene therapy, and stem cell therapy.
Table 3. Clinical trials for Stargardt’s disease using small molecules, gene therapy, and stem cell therapy.
InterventionMOAOutcomes AssessedPhaseSponsorReference
Small Molecules
TinlarebantRetinol binding protein 4 antagonistPK/PD, safety, change in atrophic lesion size1b, 2/3Mata NathanJPRN-jRCT2031240209 [124]
Change in atrophic lesion size3Belite Bio, Inc.NCT05244304 [125]
Systemic and ocular safety and tolerability; the optimal dose for Phase 21/2RBP4 Pty Ltd.NCT05266014 [126]
STG-001Reduces plasma levels of RBP4Safety and tolerability2Stargazer Pharmaceuticals, Inc.NCT04489511 [96]
Metformintargeting lysosomal and fatty acid oxidation pathways in retinal pigment epithelium cellsChange in atrophic lesion size in ellipsoid zone band1/2National Eye InstituteNCT04545736 [99]
ALK001Deuterated vitamin A; slows down the vitamin A dimer formation Safety and tolerability, change in atrophic lesion size, PK2Alkeus Pharmaceuticals, Inc.NCT02402660 [127]
Safety and tolerability, PKNCT04239625 [128]
Emixustat HydrochlorideInhibition of RPE65 and reducing 11 cis-retinal productionChange in retina electroretinogram; safety2Kubota Vision Inc.NCT03033108 [103]
Change in macular atrophic lesion3Kubota Vision Inc.NCT03772665 [104]
Soraprazanremoval of lipofuscin in retinal pigment epithelium cellsChange in quantitative fundus autofluorescence2Katairo GmbHNL-OMON48130 [110]
Omega-3 Fatty Acids SupplementationReducing complement C3 and accumulation of lipofuscin in retinal pigment epithelium cells Visual acuityN/AOphthalmos Research and Education InstituteNCT03297515 [129]
Zimura (Aptamer)Complement C5 inhibitionChange in atrophic lesion size in ellipsoid zone2Astellas Pharma Global Development, Inc.NCT03364153 [113]
Gene Therapies
ACDN-01 RNA exon editorAAV-based vector carrying a DNA construct encoding for an ABCA4 RNA exon editorSafety and tolerability1/2Ascidian Therapeutics, Inc.NCT06467344 [116]
JWK006ABCA4 gene expression via adeno-associated virusSafety and visual acuity1/2West China HospitalNCT06300476 [117]
OCU-410RORA expression using Adeno-Associated Virus serotype 5Safety and visual acuity1/2OcugenNCT05956626 [123]
SAR422459Expression of ABCA4 using recombinant equine infectious anemia virusSafety and tolerability1/2SanofiNCT01736592 [130]
vMCO-010Viral expression of Opsin activated by ambient lightSafety, visual acuity, light-guided mobility, determination of shape and optical flow2Nanoscope Therapeutics Inc.NCT05417126 [131]
Stem Cell Therapies
autologous bone marrow-derived stem cellsTreatment of retinal and optic nerve damageSafety and neuronal degradation in the eye1Pomeranian Medical University SzczecinNCT03772938 [132]
Visual acuityN/AMD Stem CellsNCT01920867 [133]
NCT03011541 [134]

1.5.3. Stem Cell Therapy for Stargardt’s Disease

The therapeutic potential of autologous bone marrow-derived stem cells in Stargardt’s disease has been assessed in two clinical trials. A phase 1 trial with an estimated enrollment of 30 patients having retinal degeneration, including those having Stargardt’s disease, started in 2018. Currently, the status of the trial is unknown [132]. Another trial using autologous bone marrow-derived stem cells in patients with retinal degeneration was a single arm, open-label study, titled as Stem Cell Ophthalmology Treatment Study (SCOTS) started in 2012 and completed in 2020 [133]. A subsequent trial Stem Cell Ophthalmology Treatment Study II (SCOTS2) started by the same organization has been started in 2016, with an expected completion in July 2026 [134]. One-year follow-up of patients with Stargardt’s disease from SCOT and SCOT2 trials showed that the disease phenotype improved or remained stable in about 75% of patients (p = 0.0004). About 94% of patients had improved vision or remained stable [135].
A pictorial summary of various therapeutic approaches that are under clinical development are shown in Figure 4. Despite ABCA4 mutations being the chief contributor in the etiology of Stargardt’s disease, gene therapies supplementing the wildtype ABCA4 are limited. The relatively large size of ABCA4 mRNA (6.6 kb) limits the use of a AAV-based gene therapy-based approach directly supplementing the wildtype ABCA4 with only one such gene therapy, JWK006, supplementing the wildtype ABCA4 (via an AAV-based vector) under clinical trial (Table 3). Supplementing wildtype ABCA4 via lentiviral or HSV-based vectors that can accommodate larger target gene payloads is limited by intrinsic problems associated with these therapies. Disease-modifying gene therapy approaches, which aim at checking disease progression, provide an alternative to ABCA4 supplementation. As described above, the disease-modifying gene therapy OCU410 that expresses RORA targets two critical aspects of Stargardt’s disease pathology—complement activation and oxidative stress. Preclinical studies in a mouse model of Stargardt’s disease indeed showed a rescue of the disease phenotype. In patients with Stargardt’s disease, OCU410ST treatment resulted in 48% slower lesion growth from baseline and a 10-letter improvement in visual function, as measured by best corrected visual acuity (BCVA), compared to untreated eyes [136]. OCU410 is also being evaluated in patients with geographic atrophy, and the preliminary results from the phase 1/2 trial show a favorable safety and tolerability profile exhibiting slower lesion growth (44%) from the baseline in treated eyes compared to untreated eyes in treated eye versus fellow eyes after single injection [137].

2. Conclusions and Future Directions

Stargardt disease was first described in 1909 by Dr. Karl Stargardt, but its genetic basis was identified only in 1997 with the discovery of ABCA4 mutations. Since the initial report of 19 variants, over 1200 pathogenic or likely pathogenic ABCA4 variants have been identified. This extensive genetic heterogeneity, often involving rare variants, complicates genotype–phenotype correlation, further challenged by variable penetrance and geographic variation in allele frequency among healthy individuals.
Only two small molecules have advanced to phase 3 trials: tinlarebant and emixustat hydrochloride. Tinlarebant showed disease stabilization in phase 2 and maintained a favorable safety profile in 1-year interim phase 3 data. Emixustat, in post hoc analysis of the completed phase 3 trial (June 2022), proved to be beneficial only in patients with small baseline lesions; with no further trials, its future remains uncertain.
Gene therapies aimed at restoring wildtype ABCA4 are in early clinical stages. A disease-modifying RORA gene therapy targeting complement activation and oxidative stress shows promise. A light-activated bioengineered opsin also offers a novel approach, with encouraging 24-week results. Stem cell therapy using autologous bone marrow-derived cells has shown positive outcomes at one-year follow-up.
RORA gene therapy, designed to regulate complement activation and oxidative stress, has shown promising results. Preclinical studies of OCU410ST demonstrated safety in cynomolgus monkeys. In the completed Phase I GARDian trial (NCT05956626), treated eyes showed a 48% reduction in atrophic lesion growth and a 2-line (10-letter) BCVA improvement at 6 months. The forthcoming results from ongoing studies will be of significant interest considering encouraging results in both structural and functional assessments. Currently Ocugen is conducting phase 2/3 pivotal confirmatory trial of OCU410ST in patients with Stargardt’s disease.
Despite progress, effective therapies for Stargardt disease remain limited. Given the complexity of the ABCA4 genotype–phenotype relationship, ABCA4-agnostic approaches, such as targeting oxidative stress (RORA gene therapy) and complement inhibition (RORA gene therapy, omega-3 supplementation, and Zimura), may offer broader therapeutic potential.

Funding

This review did not receive specific funding but was performed as part of the employment of the authors, by Ocugen Inc.

Acknowledgments

All the figures were generated by using the Biorender (www.biorender.com).

Conflicts of Interest

The authors are employees at Ocugen Inc., which is currently developing gene therapy for Stargardt disease and geographic atrophy.

References

  1. Stargardt, K. Über Familiäre, progressive degeneration in der maculagegend des auges. Graefe’s Arch. Clin. Exp. Ophthalmol. 1909, 71, 534–550. [Google Scholar] [CrossRef]
  2. Glazer, L.C.; Dryja, T.P. Understanding the etiology of Stargardt’s disease. Ophthalmol. Clin. N. Am. 2002, 15, 93–100. [Google Scholar] [CrossRef] [PubMed]
  3. Michaelides, M.; Hunt, D.M.; Moore, A.T. The genetics of inherited macular dystrophies. J. Med. Genet. 2003, 40, 641–650. [Google Scholar] [CrossRef] [PubMed]
  4. Willoughby, J.J.; Jensen, A.M. Abca4, mutated in Stargardt disease, is required for structural integrity of cone outer segments. Dis. Model. Mech. 2025, 18, DMM052052. [Google Scholar] [CrossRef] [PubMed]
  5. Allikmets, R.; Singh, N.; Sun, H.; Shroyer, N.F.; Hutchinson, A.; Chidambaram, A.; Gerrard, B.; Baird, L.; Stauffer, D.; Peiffer, A.; et al. A photoreceptor cell-specific ATP-binding transporter gene (ABCR) is mutated in recessive Stargardt macular dystrophy. Nat. Genet. 1997, 15, 236–246. [Google Scholar] [CrossRef] [PubMed]
  6. Molday, R.S.; Zhong, M.; Quazi, F. The role of the photoreceptor ABC transporter ABCA4 in lipid transport and Stargardt macular degeneration. Biochim. Biophys. Acta 2009, 1791, 573–583. [Google Scholar] [CrossRef] [PubMed]
  7. Bernstein, P.S.; Tammur, J.; Singh, N.; Hutchinson, A.; Dixon, M.; Pappas, C.M.; Zabriskie, N.A.; Zhang, K.; Petrukhin, K.; Leppert, M.; et al. Diverse macular dystrophy phenotype caused by a novel complex mutation in the ELOVL4 gene. Investig. Ophthalmol. Vis. Sci. 2001, 42, 3331–3336. [Google Scholar]
  8. Imani, S.; Cheng, J.; Dehghan Shasaltaneh, M.; Wei, C.; Yang, L.; Fu, S.; Zou, H.; Khan, M.A.; Zhang, X.; Chen, H.; et al. Genetic identification and molecular modeling characterization reveal a novel PROM1 mutation in Stargardt4-like macular dystrophy. Oncotarget 2018, 9, 122–141. [Google Scholar] [CrossRef] [PubMed]
  9. Hanany, M.; Rivolta, C.; Sharon, D. Worldwide carrier frequency and genetic prevalence of autosomal recessive inherited retinal diseases. Proc. Natl. Acad. Sci. USA 2020, 117, 2710–2716. [Google Scholar] [CrossRef] [PubMed]
  10. Shalom, S.; Ben-Yosef, T.; Sher, I.; Zag, A.; Rotenstreich, Y.; Poleg, T.; Birk, O.S.; Gradstein, L.; Ehrenberg, M.; Deitch, I.; et al. Nationwide Prevalence of Inherited Retinal Diseases in the Israeli Population. JAMA Ophthalmol. 2024, 142, 609–616. [Google Scholar] [CrossRef] [PubMed]
  11. Runhart, E.H.; Dhooge, P.; Meester-Smoor, M.; Pas, J.; Pott, J.W.R.; van Leeuwen, R.; Kroes, H.Y.; Bergen, A.A.B.; de Jong-Hesse, Y.; Thiadens, A.A.; et al. Stargardt disease: Monitoring incidence and diagnostic trends in the Netherlands using a nationwide disease registry. Acta Ophthalmol. 2022, 100, 395–402. [Google Scholar] [CrossRef] [PubMed]
  12. EMA. EU/3/19/2162-Orphan Designation for Treatment of Stargardt’s Disease. Emixustat Hydrochloride. 2025. Available online: https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu-3-19-2162 (accessed on 15 July 2025).
  13. Heath Jeffery, R.C.; Mukhtar, S.A.; McAllister, I.L.; Morgan, W.H.; Mackey, D.A.; Chen, F.K. Inherited retinal diseases are the most common cause of blindness in the working-age population in Australia. Ophthalmic Genet. 2021, 42, 431–439. [Google Scholar] [CrossRef] [PubMed]
  14. Runhart, E.H.; Khan, M.; Cornelis, S.S.; Roosing, S.; Del Pozo-Valero, M.; Lamey, T.M.; Liskova, P.; Roberts, L.; Stöhr, H.; Klaver, C.C.W.; et al. Association of Sex With Frequent and Mild ABCA4 Alleles in Stargardt Disease. JAMA Ophthalmol. 2020, 138, 1035–1042. [Google Scholar] [CrossRef] [PubMed]
  15. Sassone, F.; Estay-Ahumada, C.; Roux, M.J.; Ciocca, D.; Rossolillo, P.; Birling, M.-C.; Sparrow, J.R.; Montenegro, D.; Hicks, D. Interruption of the visual cycle in a novel animal model induces progressive vision loss resembling Stargardts Disease. Sci. Rep. 2024, 14, 30880. [Google Scholar] [CrossRef] [PubMed]
  16. Vasiliou, V.; Vasiliou, K.; Nebert, D.W. Human ATP-binding cassette (ABC) transporter family. Hum. Genom. 2009, 3, 281–290. [Google Scholar] [CrossRef] [PubMed]
  17. Biswas-Fiss, E.E.; Kurpad, D.S.; Joshi, K.; Biswas, S.B. Interaction of extracellular domain 2 of the human retina-specific ATP-binding cassette transporter (ABCA4) with all-trans-retinal. J. Biol. Chem. 2010, 285, 19372–19383. [Google Scholar] [CrossRef] [PubMed]
  18. UniProt. P78363·ABCA4_HUMAN. June 2024. Available online: https://www.uniprot.org/uniprotkb/P78363/entry (accessed on 15 July 2025).
  19. Patel, M.J.; Biswas, S.B.; Biswas-Fiss, E.E. Functional significance of the conserved C-Terminal VFVNFA motif in the retina-specific ABC transporter, ABCA4, and its role in inherited visual disease. Biochem. Biophys. Res. Commun. 2019, 519, 46–52. [Google Scholar] [CrossRef] [PubMed]
  20. Ebrey, T.; Koutalos, Y. Vertebrate photoreceptors. Prog. Retin. Eye Res. 2001, 20, 49–94. [Google Scholar] [CrossRef] [PubMed]
  21. Molday, R.S.; Moritz, O.L. Photoreceptors at a glance. J. Cell Sci. 2015, 128, 4039–4045. [Google Scholar] [CrossRef] [PubMed]
  22. Goldberg, A.F.; Moritz, O.L.; Williams, D.S. Molecular basis for photoreceptor outer segment architecture. Prog. Retin. Eye Res. 2016, 55, 52–81. [Google Scholar] [CrossRef] [PubMed]
  23. Luo, D.G.; Xue, T.; Yau, K.W. How vision begins: An odyssey. Proc. Natl. Acad. Sci. USA 2008, 105, 9855–9862. [Google Scholar] [CrossRef] [PubMed]
  24. Illing, M.; Molday, L.L.; Molday, R.S. The 220-kDa rim protein of retinal rod outer segments is a member of the ABC transporter superfamily. J. Biol. Chem. 1997, 272, 10303–10310. [Google Scholar] [CrossRef] [PubMed]
  25. Quazi, F.; Molday, R.S. ATP-binding cassette transporter ABCA4 and chemical isomerization protect photoreceptor cells from the toxic accumulation of excess 11-cis-retinal. Proc. Natl. Acad. Sci. USA 2014, 111, 5024–5029. [Google Scholar] [CrossRef] [PubMed]
  26. Quazi, F.; Lenevich, S.; Molday, R.S. ABCA4 is an N-retinylidene-phosphatidylethanolamine and phosphatidylethanolamine importer. Nat. Commun. 2012, 3, 925. [Google Scholar] [CrossRef] [PubMed]
  27. Chen, C.; Adler, L.; Goletz, P.; Gonzalez-Fernandez, F.; Thompson, D.A.; Koutalos, Y. Interphotoreceptor retinoid-binding protein removes all-trans-retinol and retinal from rod outer segments, preventing lipofuscin precursor formation. J. Biol. Chem. 2017, 292, 19356–19365. [Google Scholar] [CrossRef] [PubMed]
  28. Sears, A.E.; Bernstein, P.S.; Cideciyan, A.V.; Hoyng, C.B.; Charbel Issa, P.; Palczewski, K.; Rosenfeld, P.J.; Sadda, S.; Schraermeyer, U.; Sparrow, J.R.; et al. Towards Treatment of Stargardt Disease: Workshop Organized and Sponsored by the Foundation Fighting Blindness. Transl. Vis. Sci. Technol. 2017, 6, 6. [Google Scholar] [CrossRef] [PubMed]
  29. O’Byrne, S.M.; Blaner, W.S. Retinol and retinyl esters: Biochemistry and physiology. J. Lipid Res. 2013, 54, 1731–1743. [Google Scholar] [CrossRef] [PubMed]
  30. Molday, R.S.; Garces, F.A.; Scortecci, J.F.; Molday, L.L. Structure and function of ABCA4 and its role in the visual cycle and Stargardt macular degeneration. Prog. Retin. Eye Res. 2022, 89, 101036. [Google Scholar] [CrossRef] [PubMed]
  31. Ueda, K.; Kim, H.J.; Zhao, J.; Sparrow, J.R. Bisretinoid Photodegradation Is Likely Not a Good Thing. Adv. Exp. Med. Biol. 2018, 1074, 395–401. [Google Scholar] [PubMed]
  32. Zhang, D.; Robinson, K.; Saad, L.; Washington, I. Vitamin A cycle byproducts impede dark adaptation. J. Biol. Chem. 2021, 297, 101074. [Google Scholar] [CrossRef] [PubMed]
  33. Cornelis, S.C.; Bauwens, M.; haer-Wigman, L.; de Bruyne, M.; Pantrani, M.; de Baere, E.; Hufnagel, R.B.; Cremers, F.P.M. Compendium of Clinical Variant Classification for 2246 Unique ABCA4 Variants to Clarify Variant Pathogenicity in Stargardt Disease Using a Modified ACMG/AMP Framework. Hum. Mutat. 2023, 6815504. [Google Scholar]
  34. Lewis, R.A.; Shroyer, N.F.; Singh, N.; Allikmets, R.; Hutchinson, A.; Li, Y.; Lupski, J.R.; Leppert, M.; Dean, M. Genotype/Phenotype analysis of a photoreceptor-specific ATP-binding cassette transporter gene, ABCR, in Stargardt disease. Am. J. Hum. Genet. 1999, 64, 422–434. [Google Scholar] [CrossRef] [PubMed]
  35. Lee, W.; Zernant, J.; Nagasaki, T.; Fishman, G.A.; Tsang, S.H.; Allikmets, R. Clinical and Genetics Characterization of Hypomorphic Variants in Stargardt disease. Investig. Ophthalmol. Vis. Sci. 2021, 62, 2171. [Google Scholar]
  36. Nassisi, M.; Mohand-Saïd, S.; Dhaenens, C. -M.; Boyard, F.; Démontant, V.; Andrieu, C.; Antonio, A.; Condroyer, C.; Foussard, M.; Méjécase, C.; et al. Expanding the Mutation Spectrum in ABCA4: Sixty Novel Disease Causing Variants and Their Associated Phenotype in a Large French Stargardt Cohort. Int. J. Mol. Sci. 2018, 19, 2196. [Google Scholar] [CrossRef] [PubMed]
  37. Schulz, H.L.; Grassmann, F.; Kellner, U.; Spital, G.; Rüther, K.; Jägle, H.; Hufendiek, K.; Rating, P.; Huchzermeyer, C.; Baier, M.J.; et al. Mutation Spectrum of the ABCA4 Gene in 335 Stargardt Disease Patients From a Multicenter German Cohort-Impact of Selected Deep Intronic Variants and Common SNPs. Investig. Ophthalmol. Vis. Sci. 2017, 58, 394–403. [Google Scholar] [CrossRef] [PubMed]
  38. Riveiro-Alvarez, R.; Aguirre-Lamban, J.; Lopez-Martinez, M.A.; Trujillo-Tiebas, M.J.; Cantalapiedra, D.; Vallespin, E.; Avila-Fernandez, A.; Ramos, C.; Ayuso, C. Frequency of ABCA4 mutations in 278 Spanish controls: An insight into the prevalence of autosomal recessive Stargardt disease. Br. J. Ophthalmol. 2009, 93, 1359–1364. [Google Scholar] [CrossRef] [PubMed]
  39. Mena, M.D.; Moresco, A.A.; Vidal, S.H.; Aguilar-Cortes, D.; Obregon, M.G.; Fandiño, A.C.; Sendoya, J.M.; Llera, A.S.; Podhajcer, O.L. Clinical and Genetic Spectrum of Stargardt Disease in Argentinean Patients. Front. Genet. 2021, 12, 646058. [Google Scholar] [CrossRef] [PubMed]
  40. Passerini, I.; Sodi, A.; Giambene, B.; Mariottini, A.; Menchini, U.; Torricelli, F. Novel mutations in of the ABCR gene in Italian patients with Stargardt disease. Eye 2010, 24, 158–164. [Google Scholar] [CrossRef] [PubMed]
  41. Fujinami, K.; Strauss, R.W.; Chiang, J.P.; Audo, I.S.; Bernstein, P.S.; Birch, D.G.; Bomotti, S.M.; Cideciyan, A.V.; Ervin, A.M.; Marino, M.J.; et al. Detailed genetic characteristics of an international large cohort of patients with Stargardt disease: ProgStar study report 8. Br. J. Ophthalmol. 2019, 103, 390–397. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, X.; Liu, Z.; Cui, J.; Tan, C.; Sun, W.; Lin, Y. Clinical and Genetic Characteristics of 18 Patients from Southeast China with ABCA4-Associated Stargardt Disease. Int. J. Mol. Sci. 2025, 26, 3354. [Google Scholar] [CrossRef] [PubMed]
  43. Pas, J.; Li, C.H.Z.; Van den Broeck, F.; Dhooge, P.P.A.; De Zaeytijd, J.; Collin, R.W.J.; Leroy, B.P.; Hoyng, C.B. Progression of Atrophy as a Function of ABCA4 Variants and Age of Onset in Stargardt Disease. Investig. Ophthalmol. Vis. Sci. 2025, 66, 76. [Google Scholar] [CrossRef] [PubMed]
  44. Quinodoz, M.; Iglesias-Romero, A.B.; Cancellieri, F.; Kaminska, K.; Scholl, H.P.N.; Pfau, M.; Rivolta, C. ABCA4 c.5461-6T>C Causes Stargardt Disease Through Exon Skipping. Adv. Exp. Med. Biol. 2025, 1468, 57–62. [Google Scholar] [PubMed]
  45. Zernant, J.; Xie, Y.A.; Ayuso, C.; Riveiro-Alvarez, R.; Lopez-Martinez, M.A.; Simonelli, F.; Testa, F.; Gorin, M.B.; Strom, S.P.; Bertelsen, M.; et al. Analysis of the ABCA4 genomic locus in Stargardt disease. Hum. Mol. Genet. 2014, 23, 6797–6806. [Google Scholar] [CrossRef] [PubMed]
  46. Braun, T.A.; Mullins, R.F.; Wagner, A.H.; Andorf, J.L.; Johnston, R.M.; Bakall, B.B.; Deluca, A.P.; Fishman, G.A.; Lam, B.L.; Weleber, R.G.; et al. Non-exomic and synonymous variants in ABCA4 are an important cause of Stargardt disease. Hum. Mol. Genet. 2013, 22, 5136–5145. [Google Scholar] [CrossRef] [PubMed]
  47. Bax, N.M.; Sangermano, R.; Roosing, S.; Thiadens, A.A.H.J.; Hoefsloot, L.H.; van den Born, L.I.; Phan, M.; Klevering, B.J.; Westeneng-van Haaften, C.; Braun, T.A.; et al. Heterozygous deep-intronic variants and deletions in ABCA4 in persons with retinal dystrophies and one exonic ABCA4 variant. Hum. Mutat. 2015, 36, 43–47. [Google Scholar] [CrossRef] [PubMed]
  48. Bauwens, M.; De Zaeytijd, J.; Weisschuh, N.; Kohl, S.; Meire, F.; Dahan, K.; Depasse, F.; De Jaegere, S.; De Ravel, T.; De Rademaeker, M.; et al. An augmented ABCA4 screen targeting noncoding regions reveals a deep intronic founder variant in Belgian Stargardt patients. Hum. Mutat. 2015, 36, 39–42. [Google Scholar] [CrossRef] [PubMed]
  49. Khan, M.; Cornelis, S.S.; Del Pozo-Valero, M.; Whelan, L.; Runhart, E.H.; Mishra, K.; Bults, F.; AlSwaiti, Y.; AlTalbishi, A.; De Baere, E.; et al. Resolving the dark matter of ABCA4 for 1054 Stargardt disease probands through integrated genomics and transcriptomics. Genet. Med. 2020, 22, 1235–1246. [Google Scholar] [CrossRef] [PubMed]
  50. Wang, Y.; Wang, P.; Yi, Z.; Ouyang, J.; Jiang, Y.; Li, S.; Jia, X.; Xiao, X.; Hejtmancik, J.F.; Sun, W.; et al. ABCA4 Deep Intronic Variants Contributed to Nearly Half of Unsolved Stargardt Cases With a Milder Phenotype. Investig. Ophthalmol. Vis. Sci. 2025, 66, 65. [Google Scholar] [CrossRef] [PubMed]
  51. Lee, W.; Zernant, J.; Nagasaki, T.; Molday, L.L.; Su, P.-Y.; Fishman, G.A.; Tsang, S.H.; Molday, R.S.; Allikmets, R. Cis-acting modifiers in the ABCA4 locus contribute to the penetrance of the major disease-causing variant in Stargardt disease. Hum. Mol. Genet. 2021, 30, 1293–1304. [Google Scholar] [CrossRef] [PubMed]
  52. Guymer, R.H.; Heon, E.; Lotery, A.J.; Munier, F.L.; Schorderet, D.F.; Baird, P.N.; McNeil, R.J.; Haines, H.; Sheffield, V.C.; Stone, E.M. Variation of codons 1961 and 2177 of the Stargardt disease gene is not associated with age-related macular degeneration. Arch. Ophthalmol. 2001, 119, 745–751. [Google Scholar] [CrossRef] [PubMed]
  53. Zernant, J.; Lee, W.; Collison, F.T.; Fishman, G.A.; Sergeev, Y.V.; Schuerch, K.; Sparrow, J.R.; Tsang, S.H.; Allikmets, R. Frequent hypomorphic alleles account for a significant fraction of ABCA4 disease and distinguish it from age-related macular degeneration. J. Med. Genet. 2017, 54, 404–412. [Google Scholar] [CrossRef]
  54. Zernant, J.; Lee, W.; Nagasaki, T.; Collison, F.T.; Fishman, G.A.; Bertelsen, M.; Rosenberg, T.; Gouras, P.; Tsang, S.H.; Allikmets, R. Extremely hypomorphic and severe deep intronic variants in the ABCA4 locus result in varying Stargardt disease phenotypes. Cold. Spring. Harb. Mol. Case. Stud. 2018, 4, a002733. [Google Scholar] [CrossRef] [PubMed]
  55. Zaneveld, J.; Siddiqui, S.N.; Li, H.; Wang, X.; Wang, H.; Wang, K.; Li, H.; Ren, H.; Lopez, I.; Dorfman, A.; et al. Comprehensive analysis of patients with Stargardt macular dystrophy reveals new genotype-phenotype correlations and unexpected diagnostic revisions. Genet. Med. 2015, 17, 262–270. [Google Scholar] [CrossRef] [PubMed]
  56. Hu, J.; Pauer, G.J.; Hagstrom, S.A.; Bok, D.; DeBenedictis, M.J.; Bonilha, V.L.; Hollyfield, J.G.; Radu, R.A. Evidence of complement dysregulation in outer retina of Stargardt disease donor eyes. Redox Biol. 2020, 37, 101787. [Google Scholar] [CrossRef] [PubMed]
  57. Lenis, T.L.; Sarfare, S.; Jiang, Z.; Lloyd, M.B.; Bok, D.; Radu, R.A. Complement modulation in the retinal pigment epithelium rescues photoreceptor degeneration in a mouse model of Stargardt disease. Proc. Natl. Acad. Sci. USA 2017, 114, 3987–3992. [Google Scholar] [CrossRef] [PubMed]
  58. Zhou, J.; Jang, Y.P.; Kim, S.R.; Sparrow, J.R. Complement activation by photooxidation products of A2E, a lipofuscin constituent of the retinal pigment epithelium. Proc. Natl. Acad. Sci. USA 2006, 103, 16182–16187. [Google Scholar] [CrossRef] [PubMed]
  59. Zhou, J.; Kim, S.R.; Westlund, B.S.; Sparrow, J.R. Complement activation by bisretinoid constituents of RPE lipofuscin. Investig. Ophthalmol. Vis. Sci. 2009, 50, 1392–1399. [Google Scholar] [CrossRef] [PubMed]
  60. Miceli, M.V.; Liles, M.R.; Newsome, D.A. Evaluation of oxidative processes in human pigment epithelial cells associated with retinal outer segment phagocytosis. Exp. Cell Res. 1994, 214, 242–249. [Google Scholar] [CrossRef] [PubMed]
  61. Winkler, B.S.; Boulton, M.E.; Gottsch, J.D.; Sternberg, P. Oxidative damage and age-related macular degeneration. Mol. Vis. 1999, 5, 32. [Google Scholar] [PubMed]
  62. Strauss, O. The retinal pigment epithelium in visual function. Physiol. Rev. 2005, 85, 845–881. [Google Scholar] [CrossRef] [PubMed]
  63. Taubitz, T.; Tschulakow, A.V.; Tikhonovich, M.; Illing, B.; Fang, Y.; Biesemeier, A.; Julien-Schraermeyer, S.; Schraermeyer, U. Ultrastructural alterations in the retinal pigment epithelium and photoreceptors of a Stargardt patient and three Stargardt mouse models: Indication for the central role of RPE melanin in oxidative stress. PeerJ 2018, 6, e5215. [Google Scholar] [CrossRef] [PubMed]
  64. Radu, R.A.; Mata, N.L.; Bagla, A.; Travis, G.H. Light exposure stimulates formation of A2E oxiranes in a mouse model of Stargardt’s macular degeneration. Proc. Natl. Acad. Sci. USA 2004, 101, 5928–5933. [Google Scholar] [CrossRef] [PubMed]
  65. Clinic, C. Macula. 26 January 2025. Available online: https://my.clevelandclinic.org/health/body/23185-macula (accessed on 15 July 2025).
  66. The Human Protein Atlas. Tissue Expression of ABCA4-Summary-The Human Protein Atlas. 2025. Available online: https://www.proteinatlas.org/ENSG00000198691-ABCA4/tissue (accessed on 15 July 2025).
  67. Sparrow, J.R.; Marsiglia, M.; Allikmets, R.; Tsang, S.; Lee, W.; Duncker, T.; Zernant, J. Flecks in Recessive Stargardt Disease: Short-Wavelength Autofluorescence, Near-Infrared Autofluorescence, and Optical Coherence Tomography. Investig. Ophthalmol. Vis. Sci. 2015, 56, 5029–5039. [Google Scholar] [CrossRef] [PubMed]
  68. Zaydon, Y.A.; Tsang, S.H. The ABCs of Stargardt disease: The latest advances in precision medicine. Cell Biosci. 2024, 14, 98. [Google Scholar] [CrossRef] [PubMed]
  69. Tanna, P.; Strauss, R.W.; Fujinami, K.; Michaelides, M. Stargardt disease: Clinical features, molecular genetics, animal models and therapeutic options. Br. J. Ophthalmol. 2017, 101, 25–30. [Google Scholar] [CrossRef] [PubMed]
  70. Rotenstreich, Y.; Fishman, G.A.; Anderson, R.J. Visual acuity loss and clinical observations in a large series of patients with Stargardt disease. Ophthalmology 2003, 110, 1151–1158. [Google Scholar] [CrossRef] [PubMed]
  71. Tracewska, A.M.; Kocyła-Karczmarewicz, B.; Rafalska, A.; Murawska, J.; Jakubaszko-Jablonska, J.; Rydzanicz, M.; Stawiński, P.; Ciara, E.; Khan, M.I.; Henkes, A.; et al. Genetic Spectrum of ABCA4-Associated Retinal Degeneration in Poland. Genes 2019, 10, 959. [Google Scholar] [CrossRef] [PubMed]
  72. Lambertus, S.; van Huet, R.A.; Bax, N.M.; Hoefsloot, L.H.; Cremers, F.P.; Boon, C.J.; Klevering, B.J.; Hoyng, C.B. Early-onset stargardt disease: Phenotypic and genotypic characteristics. Ophthalmology 2015, 122, 335–344. [Google Scholar] [CrossRef] [PubMed]
  73. Lee, W.; Zernant, J.; Su, P.Y.; Nagasaki, T.; Tsang, S.H.; Allikmets, R. A genotype-phenotype correlation matrix for ABCA4 disease based on long-term prognostic outcomes. JCI Insight 2022, 7, e156154. [Google Scholar] [CrossRef]
  74. Li, C.H.Z.; Pas, J.A.A.H.; Corradi, Z.; Hitti-Malin, R.J.; Hoogstede, A.; Runhart, E.H.; Dhooge, P.P.A.; Collin, R.W.J.; Cremers, F.P.M.; Hoyng, C.B. Study of Late-Onset Stargardt Type 1 Disease: Characteristics, Genetics, and Progression. Ophthalmology 2024, 131, 87–97. [Google Scholar] [CrossRef] [PubMed]
  75. Daiber, H.F.; Gnugnoli, D.M. Visual Acuity. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  76. Fishman, G.A.; Farber, M.; Patel, B.S.; Derlacki, D.J. Visual acuity loss in patients with Stargardt’s macular dystrophy. Ophthalmology 1987, 94, 809–814. [Google Scholar] [CrossRef] [PubMed]
  77. Sutradhar, S. Direct Ophthalmoscope-Everything You Need to Know. 13 May 2024. Available online: https://smartoptometryacademy.com/direct-ophthalmoscope-all-u-need-to-know/ (accessed on 15 July 2025).
  78. Mukherjee, N.; Schuman, S. Diagnosis and Management of Stargardt Disease. EyeNet Magazine, 1 December 2014; pp. 29–31. [Google Scholar]
  79. Delori, F.C.; Dorey, C.K.; Staurenghi, G.; Arend, O.; Goger, D.G.; Weiter, J.J. In vivo fluorescence of the ocular fundus exhibits retinal pigment epithelium lipofuscin characteristics. Investig. Ophthalmol. Vis. Sci. 1995, 36, 718–729. [Google Scholar]
  80. von Ruckmann, A.; Fitzke, F.W.; Bird, A.C. Distribution of fundus autofluorescence with a scanning laser ophthalmoscope. Br. J. Ophthalmol. 1995, 79, 407–412. [Google Scholar] [CrossRef] [PubMed]
  81. Strauss, R.W.; Muñoz, B.; Ho, A.; Jha, A.; Michaelides, M.; Cideciyan, A.V.; Audo, I.; Birch, D.G.; Hariri, A.H.; Nittala, M.G.; et al. Progression of Stargardt Disease as Determined by Fundus Autofluorescence in the Retrospective Progression of Stargardt Disease Study (ProgStar Report No. 9). JAMA Ophthalmol. 2017, 135, 1232–1241. [Google Scholar] [CrossRef] [PubMed]
  82. Aumann, S.; Donner, S.; Fischer, J.; Muller, F. Optical Coherence Tomography (OCT): Principle and Technical Realization. In High Resolution Imaging in Microscopy and Ophthalmology: New Frontiers in Biomedical Optics; Bille, J.F., Ed.; Springer: Berlin/Heidelberg, Germany, 2019; pp. 59–85. [Google Scholar]
  83. Strauss, R.W.; Lang, L.; Ho, A.; Jha, A.; Ip, M.; Bernstein, P.S.; Birch, D.G.; Cideciyan, A.V.; Michaelides, M.; Audo, I.; et al. The Progression of Stargardt Disease as Determined by Spectral-Domain Optical Coherence Tomography over a 24-Month Period (ProgStar Report No. 18). Ophthalmic Res. 2024, 67, 435–447. [Google Scholar] [CrossRef] [PubMed]
  84. Giacalone, J.C.; Scruggs, B.A. The Pathology of Stargardt Disease and Future Treatment Horizons. Retin. Physician 2024, 21, 8–12. [Google Scholar]
  85. Fishman, G.A. Fundus flavimaculatus: A clinical classification. Arch. Ophthalmol. 1976, 94, 2061–2067. [Google Scholar] [CrossRef] [PubMed]
  86. Fujinami, K.; Fujinami, K.; Lois, N.; Mukherjee, R.; McBain, V.A.; Tsunoda, K.; Tsubota, K.; Stone, E.M.; Fitzke, F.W.; Bunce, C.; et al. A longitudinal study of Stargardt disease: Quantitative assessment of fundus autofluorescence, progression, and genotype correlations. Investig. Ophthalmol. Vis. Sci. 2013, 54, 8181–8190. [Google Scholar] [CrossRef] [PubMed]
  87. Wabbels, B.; Demmler, A.; Paunescu, K.; Wegscheider, E.; Preising, M.N.; Lorenz, B. Fundus autofluorescence in children and teenagers with hereditary retinal diseases. Graefes Arch. Clin. Exp. Ophthalmol. 2006, 244, 36–45. [Google Scholar] [CrossRef] [PubMed]
  88. Wu, J.; Fallon, J. Belite Bio Announces First Patient Dosed in Phase 2/3 DRAGON II Trial of Tinlarebant for the Treatment of Stargardt Disease. 10 September 2024. Available online: https://www.globenewswire.com/news-release/2024/09/10/2943626/0/en/Belite-Bio-Announces-First-Patient-Dosed-in-Phase-2-3-DRAGON-II-Trial-of-Tinlarebant-for-the-Treatment-of-Stargardt-Disease.html (accessed on 15 July 2025).
  89. Steinhoff, J.S.; Lass, A.; Schupp, M. Biological Functions of RBP4 and Its Relevance for Human Diseases. Front. Physiol. 2021, 12, 659977. [Google Scholar] [CrossRef] [PubMed]
  90. Wu, J.; McCarthy, T. Belite Bio Receives FDA Fast Track Designation for LBS-008. 3 May 2022. Available online: https://www.globenewswire.com/news-release/2022/05/03/2434325/0/en/Belite-Bio-Receives-FDA-Fast-Track-Designation-For-LBS-008.html (accessed on 15 July 2025).
  91. Wu, J.; Fallon, J. Belite Bio Receives Sakigake (Pioneer Drug) Designation of Tinlarebant for Stargardt Disease in Japan. 12 June 2024. Available online: https://www.globenewswire.com/news-release/2024/06/12/2897299/0/en/Belite-Bio-Receives-Sakigake-Pioneer-Drug-Designation-of-Tinlarebant-for-Stargardt-Disease-in-Japan.html (accessed on 15 July 2025).
  92. Belite Bio. Belite Bio Presents Results from a 24-Month, Phase 2 Study of Tinlarebant in Childhood-Onset Stargardt Disease at the AAO Annual Meeting. June 2023. Available online: https://investors.belitebio.com/news-releases/news-release-details/belite-bio-presents-results-24-month-phase-2-study-tinlarebant (accessed on 15 July 2025).
  93. Belite Bio. Belite Bio Announces Interim Analysis Results from the Pivotal Global Phase 3 DRAGON Trial of Tinlarebant in Adolescent Stargardt Disease Subjects. 27 February 2025. Available online: https://investors.belitebio.com/news-releases/news-release-details/belite-bio-announces-interim-analysis-results-pivotal-global (accessed on 15 July 2025).
  94. Retinal Degeneration Fund; Stargazer Pharmaceuticals, Inc. Announces $57 Million Series A Financing and Initiation of a Phase 2a Clinical Study of STG-001 in Stargardt Disease Patients. 9 November 2020. Available online: https://www.retinaldegenerationfund.org/news/news-posts/stargazer-pharmaceuticals-inc-announces-57-million-series-a-financing-and-initiation-of-a-phase-2a-clinical-study-of-stg-001-in-stargardt-disease-patients/ (accessed on 15 July 2025).
  95. U.S. Food and Drug Administration. Search Orphan Drug Designations and Approvals: 3-(3-(3,5-bis(trifluoromethyl)phenyl)-1H-pyrazol-1-yl) Propanoic Acid. 22 April 2019. Available online: https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=681519&utm (accessed on 15 July 2025).
  96. ClinicalTrials.gov. A Phase 2a Study of the Safety, Pharmacokinetics and Pharmacodynamics of STG-001 in Subjects With Stargardt Disease (STGD1) Caused by Autosomal Recessive Mutation in ATP Binding Cassette Subfamily A Member 4 (ABCA4) Gene. 27 April 2021. Available online: https://clinicaltrials.gov/study/NCT04489511 (accessed on 15 July 2025).
  97. Farnoodian, M.; Barone, F.; Boyle, M.; Gupta, R.; Nelson, L.M.; Bose, D.A.; Jun, B.; Gordon, W.C.; Do, K.V.; Guerin, M.A.; et al. Metformin Attenuates the Hallmarks of Stargardt Disease. Investig. Ophthalmol. Vis. Sci. 2023, 64, 476. [Google Scholar]
  98. Bose, D.A.; Farnoodian, M.; Sharma, R.; McGaughey, D.; Bharti, K. Use of Metformin as a Potential Treatment for Stargardt Maculopathy. Investig. Ophthalmol. Vis. Sci. 2024, 65, 2203. [Google Scholar]
  99. ClinicalTrials.gov. Oral Metformin for Treatment of ABCA4 Retinopathy. 15 January 2025. Available online: https://clinicaltrials.gov/study/NCT04545736 (accessed on 15 July 2025).
  100. Kaufman, Y.; Ma, L.; Washington, I. Deuterium enrichment of vitamin A at the C20 position slows the formation of detrimental vitamin A dimers in wild-type rodents. J. Biol. Chem. 2011, 286, 7958–7965. [Google Scholar] [CrossRef] [PubMed]
  101. Kubota, R.; Gregory, J.; Henry, S.; Mata, N.L. Pharmacotherapy for metabolic and cellular stress in degenerative retinal diseases. Drug Discov. Today 2020, 25, 292–304. [Google Scholar] [CrossRef] [PubMed]
  102. U.S. Food and Drug Administration. Search Orphan Drug Designations and Approvals: Emixustat. 4 January 2017. Available online: https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=548816&utm (accessed on 15 July 2025).
  103. ClinicalTrials.gov. A Phase 2a Multicenter, Randomized, Masked Study Evaluating the Pharmacodynamics of Emixustat Hydrochloride in Subjects With Macular Atrophy Secondary to Stargardt Disease. 19 May 2021. Available online: https://clinicaltrials.gov/study/NCT03033108 (accessed on 15 July 2025).
  104. ClinicalTrials.gov. A Phase 3 Multicenter, Randomized, Double-Masked Study Comparing the Efficacy and Safety of Emixustat Hydrochloride with Placebo for the Treatment of Macular Atrophy Secondary to Stargardt Disease. 30 May 2024. Available online: https://clinicaltrials.gov/study/NCT03772665 (accessed on 15 July 2025).
  105. Business Wire. Kubota Vision Announces Positive Post Hoc Analysis from Phase 3 Clinical Trial of Emixustat in Patients with Stargardt Disease. 3 October 2022. Available online: https://www.businesswire.com/news/home/20221003005323/en/Kubota-Vision-Announces-Positive-Post-Hoc-Analysis-from-Phase-3-Clinical-Trial-of-Emixustat-in-Patients-with-Stargardt-Disease (accessed on 15 July 2025).
  106. Julien-Schraermeyer, S.; Illing, B.; Tschulakow, A.; Taubitz, T.; Guezguez, J.; Burnet, M.; Schraermeyer, U. Penetration, distribution, and elimination of remofuscin/soraprazan in Stargardt mouse eyes following a single intravitreal injection using pharmacokinetics and transmission electron microscopic autoradiography: Implication for the local treatment of Stargardt’s disease and dry age-related macular degeneration. Pharmacol. Res. Perspect. 2020, 8, e00683. [Google Scholar]
  107. Julien, S.; Schraermeyer, U. Lipofuscin can be eliminated from the retinal pigment epithelium of monkeys. Neurobiol. Aging 2012, 33, 2390–2397. [Google Scholar] [CrossRef] [PubMed]
  108. Fang, Y.; Tschulakow, A.; Tikhonovich, M.; Taubitz, T.; Illing, B.; Schultheiss, S.; Schraermeyer, U.; Julien-Schraermeyer, S. Preclinical results of a new pharmacological therapy approach for Stargardt disease and dry age-related macular degeneration. Investig. Ophthalmol. Vis. Sci. 2017, 58, 256. [Google Scholar]
  109. U.S. Food and Drug Administration. Search Orphan Drug Designations and Approvals: Soraprazan. 22 December 2017. Available online: https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=617617&utm (accessed on 15 July 2025).
  110. Central Committee on Research Involving Human Subjects, T.N. A Multi-National, Multi-Centre, Double-Masked, Placebo-Controlled Proof of Concept Trial to Evaluate the Safety and Efficacy of Oral Soraprazan in Stargardt Disease. 12 April 2024. Available online: https://onderzoekmetmensen.nl/en/trial/48130 (accessed on 15 July 2025).
  111. Prokopiou, E.; Kolovos, P.; Kalogerou, M.; Neokleous, A.; Nicolaou, O.; Sokratous, K.; Kyriacou, K.; Georgiou, T. Omega-3 Fatty Acids Supplementation: Therapeutic Potential in a Mouse Model of Stargardt Disease. Investig. Ophthalmol. Vis. Sci. 2018, 59, 2757–2767. [Google Scholar] [CrossRef] [PubMed]
  112. Prokopiou, K.; Kolovos, P.; Tsangari, H.; Bandello, F.; Rossetti, L.M.; Mastropasqua, L.; Mohand-Said, S.; Georgiou, T. A prospective, multicentre, randomised, double-blind study designed to assess the potential effects of omega-3 fatty acids supplementation in dry age-related macular degeneration or Stargardt disease. Investig. Ophthalmol. Vis. Sci. 2022, 63, 377-F0208. [Google Scholar]
  113. ClinicalTrials.gov. A Phase 2b Randomized, Double-Masked, Controlled Trial to Establish the Safety and Efficacy of Zimura™ (Complement C5 Inhibitor) Compared to Sham in Subjects With Autosomal Recessive Stargardt Disease. 4 December 2024. Available online: https://clinicaltrials.gov/study/NCT03364153 (accessed on 15 July 2025).
  114. Karen, O.H.C. FDA Clears First Clinical Trial of RNA Exon Editor Developed to Treat Stargardt Disease. 31 January 2024. Available online: https://crisprmedicinenews.com/news/fda-clears-first-clinical-trial-of-rna-exon-editor-developed-to-treat-stargardt-disease/ (accessed on 15 July 2025).
  115. Ascidian. Ascidian Therapeutics Announces First-Ever IND for an RNA Exon Editor as FDA Approves Trial Plan and Fast Tracks ACDN-01 in Stargardt Disease and Other ABCA4 Retinopathies. 29 January 2024. Available online: https://ascidian-tx.com/wp-content/uploads/2024/01/Ascidian-IND-Acceptance-Release_FINAL.1.29.24.pdf (accessed on 15 July 2025).
  116. ClinicalTrials.gov. ACDN-01-001: Open-Label, Single Ascending Dose Study to Evaluate the Safety, Tolerability, and Preliminary Efficacy of Subretinal ACDN-01 in Participants With ABCA4-related Retinopathy. 22 January 2025. Available online: https://clinicaltrials.gov/study/NCT06467344 (accessed on 15 July 2025).
  117. ClinicalTrials.gov. Safety and Efficacy of a Single Subretinal Injection of JWK006 Gene Therapy in Subjects with Stargardt Disease (STGD1). 8 March 2024. Available online: https://clinicaltrials.gov/study/NCT06300476 (accessed on 15 July 2025).
  118. Bank, D. Sonpiretigene Isteparvovec. 6 March 2025. Available online: https://go.drugbank.com/drugs/DB17844?utm (accessed on 15 July 2025).
  119. Allen, C.H. MCO-010 optogenetic gene therapy for severe vision loss in Stargardt disease. Opthalmol. Times, 17 November 2023; p. 48. [Google Scholar]
  120. Akula, M.; McNamee, S.M.; Love, Z.; Nasraty, N.; Chan, N.P.M.; Whalen, M.; Avola, M.O.; Olivares, A.M.; Leehy, B.D.; Jelcick, A.S.; et al. Retinoic acid related orphan receptor alpha is a genetic modifier that rescues retinal degeneration in a mouse model of Stargardt disease and Dry AMD. Gene Ther. 2024, 31, 413–421. [Google Scholar] [CrossRef] [PubMed]
  121. San, W.; Zhou, Q.; Shen, D.; Cao, D.; Chen, Y.; Meng, G. Roles of retinoic acid-related orphan receptor alpha in high glucose-induced cardiac fibroblasts proliferation. Front. Pharmacol. 2025, 16, 1539690. [Google Scholar] [CrossRef] [PubMed]
  122. Boukhtouche, F.; Vodjdani, G.; Jarvis, C.I.; Bakouche, J.; Staels, B.; Mallet, J.; Mariani, J.; Lemaigre-Dubreuil, Y.; Brugg, B. Human retinoic acid receptor-related orphan receptor alpha1 overexpression protects neurones against oxidative stress-induced apoptosis. J. Neurochem. 2006, 96, 1778–1789. [Google Scholar] [CrossRef] [PubMed]
  123. ClinicalTrials.gov. A Phase 1/2 Study to Assess the Safety and Efficacy of OCU410ST for STARGARDT DISEASE. 12 March 2024. Available online: https://clinicaltrials.gov/study/NCT05956626 (accessed on 15 July 2025).
  124. ClinicalTrials.gov. A Phase Ib, Open-Label Study to Evaluate the Pharmacokinetics, Pharmacodynamics, Safety, and Tolerability of Tinlarebant in Japanese Patients with Stargardt Disease and a Phase II/III, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety, Tolerability, and Efficacy of Tinlarebant in Japanese Patients with Stargardt Disease. 28 November 2024. Available online: https://clinicaltrials.gov/study/NCT06388083 (accessed on 15 July 2025).
  125. ClinicalTrials.gov. Phase 3, Multicenter, Randomized, Double-Masked, Placebo-Controlled Study to Evaluate the Safety and Efficacy of Tinlarebant in the Treatment of Stargardt Disease in Adolescent Subjects. 7 June 2024. Available online: https://clinicaltrials.gov/study/NCT05244304 (accessed on 15 July 2025).
  126. ClinicalTrials.gov. Phase 1/2, Open-Label, Dose-Finding Followed by 2-Year Extension Study to Evaluate Safety and Tolerability of Tinlarebant in Adolescent Subjects With Stargardt Disease. 2024. Available online: https://clinicaltrials.gov/study/NCT05266014#study-plan (accessed on 15 July 2025).
  127. ClinicalTrials.gov. A Phase 2 Multicenter, Double-Masked, Randomized, Placebo-Controlled Study to Investigate the Long Term Safety, Tolerability, Pharmacokinetics and Effects of ALK-001 on the Progression of Stargardt Disease. 27 April 2025. Available online: https://clinicaltrials.gov/study/NCT02402660 (accessed on 15 July 2025).
  128. ClinicalTrials.gov. A Phase 2 Multicenter, Double-Masked, Randomized, Placebo-Controlled Study to Investigate the Long Term Safety, Tolerability, Pharmacokinetics and Effects of ALK-001 on the Progression of Stargardt Disease. 21 June 2024. Available online: https://clinicaltrials.gov/study/NCT04239625 (accessed on 15 July 2025).
  129. ClinicalTrials.gov. Prospective, Randomised, Double-Blind Study to Assess the Therapeutic Potential of Omega-3 Fatty Acids Supplementation in Dry Macular Degeneration and Stargardt Disease (Macular Degeneration Omega-3 Study—MADEOS. 23 February 2021. Available online: https://clinicaltrials.gov/study/NCT03297515 (accessed on 15 July 2025).
  130. ClinicalTrials.gov. An Open Label Study to Determine the Long Term Safety, Tolerability and Biological Activity of SAR422459 in Patients With Stargardt’s Macular Degeneration. 1 October 2024. Available online: https://clinicaltrials.gov/study/NCT01736592 (accessed on 15 July 2025).
  131. ClinicalTrials.gov. A Phase 2a, Open Label Multicenter Clinical Trial to Evaluate the Safety and Effects of a Single Intravitreal Injection of vMCO-010 Optogenetic Therapy in Subjects With Stargardt Disease. 11 September 2023. Available online: https://clinicaltrials.gov/study/NCT05417126 (accessed on 15 July 2025).
  132. ClinicalTrials.gov. Stem Cells Therapy in Degenerative Diseases of the Retina. 17 December 2018. Available online: https://clinicaltrials.gov/study/NCT03772938 (accessed on 15 July 2025).
  133. U.S. Food and Drug Administration. Stem Cell Ophthalmology Treatment Study (SCOTS). 23 October 2019. Available online: https://clinicaltrials.gov/study/NCT01920867 (accessed on 15 July 2025).
  134. ClinicalTrials.gov. Bone Marrow Derived Stem Cell Ophthalmology Treatment Study II. 16 April 2024. Available online: https://clinicaltrials.gov/study/NCT03011541 (accessed on 15 July 2025).
  135. Weiss, J.N.; Levy, S. Stem Cell Ophthalmology Treatment Study (SCOTS): Bone Marrow-Derived Stem Cells in the Treatment of Stargardt Disease. Medicines 2020, 7, 16. [Google Scholar] [CrossRef] [PubMed]
  136. Ocugen. Ocugen Announces Positive Opinion of EMA’s Committee for Advanced Therapies for ATMP Classification for Novel Modifier Gene Therapy Candidate OCU410 for Geographic Atrophy and OCU410ST for Stargardt Disease. Available online: https://ir.ocugen.com/news-releases/news-release-details/ocugen-announces-positive-opinion-emas-committee-advanced (accessed on 15 July 2025).
  137. Ocugen. Ocugen, Inc. Announces Dosing Completion in the Phase 2 ArMaDa Clinical Trial for OCU410—A Multifunctional Modifier Gene Therapy for the Treatment of Geographic Atrophy Secondary to Dry Age-Related Macular Degeneration. 12 February 2025. Available online: https://ir.ocugen.com/node/13586/pdf (accessed on 15 July 2025).
Figure 1. A schematic showing a linear structure of ABCA4 protein is depicted. (A) The domains of ABCA4 protein with amino acid numbers are shown. (B) ABCA4 variants that are present in at least 5% of patients are shown. a Frameshift results in a premature stop codon 6 amino acids downstream from the mutation site, b Frameshift results in a premature stop codon 13 amino acids downstream from the mutation site.
Figure 1. A schematic showing a linear structure of ABCA4 protein is depicted. (A) The domains of ABCA4 protein with amino acid numbers are shown. (B) ABCA4 variants that are present in at least 5% of patients are shown. a Frameshift results in a premature stop codon 6 amino acids downstream from the mutation site, b Frameshift results in a premature stop codon 13 amino acids downstream from the mutation site.
Ijms 26 07006 g001
Figure 2. Visual cycle and ABCA4. The schematic depicts the steps of the visual cycle and involvement of ABCA4. Opsin binds to 11-cis-retinal and becomes photoactive (rhodopsin). The light, in visible range, induces the conformational change in the bound opsin bound 11-cis-retinal to convert it into all-trans-retinal which binds to phosphatidylethanolamine (PE) to form N-trans-retinylidene-phosphatidylethanolamine (NRPE). ABCA4 flips the NRPE to the cytoplasmic side from the lumen side of photoreceptor outer segment, where the all-trans-retinal is converted into all-trans-retinol by the action of retinyl dehydrogenase. The PE is recycled to the lumen side by the action of ABCA4. The all-trans-retinol binds to interphotoreceptor retinoid binding protein (IRBP), which takes it through the interphotoreceptor matrix to the retinal pigment epithelium cells. Inside the retinal pigment epithelium cells, the all-trans-retinal, which is sourced from photoreceptor outer segments and blood circulation, is bound to Cellular Retinol Binding Protein Type I (CRBPI). Series of steps lead to conversion of all-trans-retinol to 11-cis-retinol, which binds to Cellular Retinaldehyde Binding Protein (CRALBP) and is subsequently converted into 11-cis-retinol, which enters the interphotoreceptor matrix and binds to IRBP. In the retinal pigment epithelium cells, all-trans-retinol is also converted to all-trans-retinyl ester, which is a storage form of retinol. PE binds to the recycled 11-cis-retinal to form NRPE, which is flipped by the ABCA4 to the lumen side of the photoreceptor outer segment. After the flipping, 11-cis-retinal is available for binding to the opsin to start a fresh round of visual cycle.
Figure 2. Visual cycle and ABCA4. The schematic depicts the steps of the visual cycle and involvement of ABCA4. Opsin binds to 11-cis-retinal and becomes photoactive (rhodopsin). The light, in visible range, induces the conformational change in the bound opsin bound 11-cis-retinal to convert it into all-trans-retinal which binds to phosphatidylethanolamine (PE) to form N-trans-retinylidene-phosphatidylethanolamine (NRPE). ABCA4 flips the NRPE to the cytoplasmic side from the lumen side of photoreceptor outer segment, where the all-trans-retinal is converted into all-trans-retinol by the action of retinyl dehydrogenase. The PE is recycled to the lumen side by the action of ABCA4. The all-trans-retinol binds to interphotoreceptor retinoid binding protein (IRBP), which takes it through the interphotoreceptor matrix to the retinal pigment epithelium cells. Inside the retinal pigment epithelium cells, the all-trans-retinal, which is sourced from photoreceptor outer segments and blood circulation, is bound to Cellular Retinol Binding Protein Type I (CRBPI). Series of steps lead to conversion of all-trans-retinol to 11-cis-retinol, which binds to Cellular Retinaldehyde Binding Protein (CRALBP) and is subsequently converted into 11-cis-retinol, which enters the interphotoreceptor matrix and binds to IRBP. In the retinal pigment epithelium cells, all-trans-retinol is also converted to all-trans-retinyl ester, which is a storage form of retinol. PE binds to the recycled 11-cis-retinal to form NRPE, which is flipped by the ABCA4 to the lumen side of the photoreceptor outer segment. After the flipping, 11-cis-retinal is available for binding to the opsin to start a fresh round of visual cycle.
Ijms 26 07006 g002
Figure 3. A schematic showing the general role of ABCA4 loss in Stargardt’s disease pathology. Loss of ABCA4 function results in accumulation of vitamin A derivatives in photoreceptors. Autophagy of photoreceptor outer membrane segments results in increased accumulation of lipofuscin. ABCA4 loss also results in increased complement C3 activation, which has been shown to increase lipofuscin accumulation. Also, photooxidation of lipofuscin, which gets augmented in the presence of increased oxidative stress due to loss of ABCA4, in turn activates the complement C3 in retinal pigment epithelium cells eventually leading to their cell death and decreased central vision.
Figure 3. A schematic showing the general role of ABCA4 loss in Stargardt’s disease pathology. Loss of ABCA4 function results in accumulation of vitamin A derivatives in photoreceptors. Autophagy of photoreceptor outer membrane segments results in increased accumulation of lipofuscin. ABCA4 loss also results in increased complement C3 activation, which has been shown to increase lipofuscin accumulation. Also, photooxidation of lipofuscin, which gets augmented in the presence of increased oxidative stress due to loss of ABCA4, in turn activates the complement C3 in retinal pigment epithelium cells eventually leading to their cell death and decreased central vision.
Ijms 26 07006 g003
Figure 4. Various therapeutic approaches (small molecules in green and gene therapy in blue) for Stargardt’s disease are shown.
Figure 4. Various therapeutic approaches (small molecules in green and gene therapy in blue) for Stargardt’s disease are shown.
Ijms 26 07006 g004
Table 1. Prevalence of various common (prevalence ≥ 5%) mutations across countries.
Table 1. Prevalence of various common (prevalence ≥ 5%) mutations across countries.
VariantAmino Acid ChangePrevalenceCountry
3386G>TR1129L26% [38]Spain
10% [39]Argentina
5882G>AG1961E20% [40]Italy
15% [41]USA
18% [34]USA
13% [37]Germany
10% [39]Argentina
~8% [38]Spain
3113C>TA1038V9% [37]Germany
18% [34]USA
2894A>GN965S10% [42]China
1622T>CL541P9% [37]Germany
2588G>CG863A, G863del7% [37]Germany
5461-10T>CT1821Vfs*13, T1821Dfs*66% [37]Germany
5% [41]USA
Table 2. Fundus autofluorescence (FAF) features are described for various genetic retinopathies.
Table 2. Fundus autofluorescence (FAF) features are described for various genetic retinopathies.
DiseaseFAF Features
Stargardt diseaseCentral oval area of reduced AF, often surrounded by irregular AF
Best diseaseCentral round structure with regular or irregular intense AF
Rod-cone dystrophiesCentral oval ring-shaped area of increased AF
Early-onset severe retinal dystrophy (with RPE65 mutations)Complete absence of AF
Leber congenital amaurosisAF is normal (except in EOSRD with RPE65 mutations)
X-linked retinoschisisCentral radial structures of AF
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dayma, K.; Rajanala, K.; Upadhyay, A. Stargardt’s Disease: Molecular Pathogenesis and Current Therapeutic Landscape. Int. J. Mol. Sci. 2025, 26, 7006. https://doi.org/10.3390/ijms26147006

AMA Style

Dayma K, Rajanala K, Upadhyay A. Stargardt’s Disease: Molecular Pathogenesis and Current Therapeutic Landscape. International Journal of Molecular Sciences. 2025; 26(14):7006. https://doi.org/10.3390/ijms26147006

Chicago/Turabian Style

Dayma, Kunal, Kalpana Rajanala, and Arun Upadhyay. 2025. "Stargardt’s Disease: Molecular Pathogenesis and Current Therapeutic Landscape" International Journal of Molecular Sciences 26, no. 14: 7006. https://doi.org/10.3390/ijms26147006

APA Style

Dayma, K., Rajanala, K., & Upadhyay, A. (2025). Stargardt’s Disease: Molecular Pathogenesis and Current Therapeutic Landscape. International Journal of Molecular Sciences, 26(14), 7006. https://doi.org/10.3390/ijms26147006

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop