Fact-checked by Grok 2 weeks ago

Cone dystrophy

Cone dystrophy, also known as progressive cone dystrophy, is a rare group of inherited retinal disorders characterized by the primary degeneration of photoreceptor cells in the , which are responsible for and central . These conditions typically manifest in childhood or with progressive vision loss, distinguishing them from broader cone-rod dystrophies where rod cells are also affected early. The prevalence is estimated at approximately 1 in 20,000 to 100,000 individuals worldwide, reflecting their rarity and . Clinically, cone dystrophy presents with hallmark symptoms including reduced central , photophobia (sensitivity to light), and impaired , often leading to central scotomas (blind spots) that severely impact daily activities such as reading or recognizing faces. Unlike night blindness, which is more common in rod-affecting disorders, patients with pure cone dystrophy initially retain good peripheral and low-light vision, though rod involvement may develop later in some cases, accelerating progression to legal blindness by around age 48 on average. Diagnosis typically involves to confirm cone dysfunction, fundus examination showing macular changes, and to identify causative mutations. The underlying causes are in over 30 genes that disrupt photoreceptor function, maintenance, or development, with patterns including autosomal dominant, autosomal recessive, and rarely X-linked forms. Notable genes include GUCA1A (associated with early-onset dominant forms) and ABCA4 (linked to recessive cases with potential rod involvement). As of 2025, there is no cure, but management focuses on low-vision aids, UV-protective sunglasses to alleviate , and ongoing research into gene therapies, such as those targeting RPGR mutations in associated , as well as emerging cell therapies like OpCT-001 for primary photoreceptor diseases, offers hope for future treatments.

Overview

Definition

Cone dystrophy encompasses a group of inherited disorders characterized by the primary degeneration of cone photoreceptors, which are concentrated in the and responsible for central vision, color perception, and high-acuity tasks under bright light conditions. This leads to progressive or, in some cases, stationary impairment of these functions, with the cones in the central typically affected first. Unlike rod-cone dystrophies, such as , where photoreceptors degenerate initially, resulting in early night blindness and peripheral field loss before cone involvement, cone dystrophy spares s at onset. It is also distinct from cone-rod dystrophies, in which cone dysfunction predominates early but is followed by secondary degeneration, often leading to more widespread involvement over time. The terminology has evolved from earlier designations like "cone degeneration" or "retinal cone dystrophy," reflecting a broader recognition of both isolated cone-specific forms and early cone-dominant variants within the spectrum of inherited diseases. Core characteristics include an onset usually in childhood or early adulthood, with rod-mediated vision—such as —initially preserved, distinguishing the condition's functional profile from more generalized photoreceptor losses.

Epidemiology

Cone dystrophy is a rare inherited retinal disorder; the exact prevalence of isolated forms is unknown, but estimates for cone-dominant retinal disorders range from approximately 1 in 40,000 to 100,000 individuals worldwide. This rarity positions it as significantly less common than other retinal dystrophies, such as , which affects about 1 in 4,000 people. While exact incidence rates are not well-established due to diagnostic challenges, the condition shows higher occurrence in populations with elevated rates, primarily attributable to its autosomal recessive inheritance patterns that are more frequent in such communities. Reported cases span diverse ethnic groups without a pronounced geographic bias, including European, Middle Eastern, South Asian, and North American populations. The age of onset typically ranges from childhood to early adulthood, often between 5 and 30 years, though stationary forms—characterized by non-progressive cone dysfunction—may manifest from birth or infancy. Progression and severity can vary widely, even within families, influencing when symptoms become clinically apparent. Cone dystrophy affects males and females equally in its sporadic, autosomal dominant, and autosomal recessive forms, which constitute the majority of cases. However, X-linked variants predominantly impact males, with females often experiencing milder or status due to X-chromosome inactivation. Over 30 genetic subtypes have been identified, reflecting extensive molecular heterogeneity that contributes to underdiagnosis, particularly in non-specialized clinical settings where comprehensive is unavailable. This diversity underscores the condition's complexity and the need for targeted screening in at-risk populations.

Clinical Presentation

Symptoms

Cone dystrophy primarily manifests through subjective visual disturbances arising from dysfunction of the cone photoreceptors in the . Patients commonly report reduced central , characterized by blurred or distorted vision in the central field of view, which significantly impairs tasks such as reading, recognizing faces, and performing detailed work. Dyschromatopsia, or impaired , is another hallmark symptom, often beginning with difficulties in discriminating between red and green hues and potentially progressing to more severe color perception deficits. , an increased sensitivity to bright light, frequently accompanies these issues, leading to discomfort, squinting, or the need for tinted lenses during daylight exposure. Hemeralopia, or day blindness, further contributes to these challenges, as affected individuals experience heightened difficulty seeing in bright environments while initially retaining normal . The progression of symptoms varies by subtype: stationary forms, often congenital, remain relatively stable from , whereas forms typically onset in late childhood or early adulthood and worsen gradually over years, sometimes resulting in legal blindness ( of 20/200 or worse). Associated complaints may include and headaches due to prolonged visual effort, with rare instances of early (involuntary eye movements) in congenital cases.

Physical Signs

In cone dystrophy, fundoscopic examination typically reveals a bull's-eye maculopathy characterized by a ring of perifoveal retinal pigment epithelial surrounding a relatively preserved central fovea, reflecting early selective loss in the . The peripheral often appears normal in the initial stages, as function remains intact at onset. As the disease progresses, more extensive macular may develop, sometimes accompanied by pigment clumping or cystoid in advanced forms. Visual field perimetry commonly demonstrates central or paracentral scotomas corresponding to macular involvement, with peripheral fields spared early in the course due to preserved rod-mediated . These defects align with the reduced central reported by patients. Fine , manifesting as involuntary eye movements, can be observed in some cases of congenital or severe early-onset cone dystrophy. Color vision assessment yields abnormal results on standard tests such as Ishihara plates or the Farnsworth-Munsell 100-hue test, indicating tritan (blue-yellow) axis defects due to cone dysfunction. Pupillary light reflexes remain normal and brisk in early to moderate stages, as the rod pathways and afferent pupillary system are initially unaffected.

Pathophysiology

Cellular Mechanisms

Cone photoreceptor dysfunction in cone dystrophy primarily arises from defects in visual pigments such as opsins or supporting proteins, which impair phototransduction—the process by which light is converted into electrical signals. These defects disrupt the cyclic nucleotide-gated (CNG) channel function and cGMP metabolism, leading to abnormal depolarization and prolonged channel opening in cone cells. As a result, cellular stress accumulates, triggering programmed cell death pathways, including apoptosis via caspase-3 activation and necroptosis mediated by RIP3, as observed in animal models of cone-specific mutations. The , particularly the fovea where are densely concentrated, is preferentially affected, with early degeneration of these high-density populations initiating central impairment. Secondary processes, such as from activated and releasing pro-inflammatory cytokines like TNF-α, exacerbate loss by promoting . further accelerates damage through (ROS) accumulation, mitochondrial dysfunction, and toxic buildup, which can be mitigated by antioxidants in preclinical models. Cone dystrophies manifest in stationary or progressive forms; stationary variants, such as certain achromatopsias, involve non-progressive developmental arrests in cone maturation and function without ongoing degeneration. In contrast, progressive forms exhibit chronic metabolic failures in cone energy production and outer segment maintenance, driven by sustained cellular stress. Rod photoreceptors are initially spared due to their lower metabolic demands, peripheral location, and reliance on different trophic factors, though in cone-rod variants, rod-derived cone viability factors diminish over time, leading to secondary spillover effects. Pathological hallmarks include the accumulation of —a of incomplete —in the , contributing to , and disorganization of cone outer segments, characterized by shortened discs and impaired renewal. These features, evident in histological biopsies and disease models, reflect disrupted protein trafficking and , underscoring the metabolic vulnerability of cones.

Genetic Basis

Cone dystrophy is a genetically heterogeneous group of inherited disorders primarily affecting photoreceptors, with in over 35 s accounting for approximately % of cases. The condition exhibits diverse inheritance patterns, with autosomal recessive being the most common, comprising 43-% of genetically solved cases depending on cohort studies, followed by autosomal dominant (12-35%) and rare X-linked recessive forms (about 1%). Sporadic cases can occur due to or incomplete , contributing to the unsolved fraction of up to 44% of cases. Key genes implicated include GUCY2D for autosomal dominant early-onset forms, where mutations disrupt activity essential for phototransduction; ABCA4 for recessive cases overlapping with , involving accumulation; CRX, a gene associated with dominant cone dystrophy; and RPGR for X-linked inheritance, particularly mutations in the ORF15 region affecting ciliary function. Other notable genes are GUCA1A and CDHR1. Mutation types predominantly consist of missense, nonsense, frameshift, and splicing variants that impair cone-specific proteins, such as activators in GUCA1A or cadherin-like proteins in CDHR1. For instance, the GUCA1A p.(Tyr99Cys) exemplifies disruptions in calcium-dependent regulation of cyclic GMP. Genotype-phenotype correlations highlight variability; biallelic null mutations in ABCA4 often lead to severe early-onset cone-rod dystrophy with central atrophy, while missense variants may result in milder progression. In GUCA1A, certain mutations cause autosomal dominant cone dystrophy with variable expressivity, ranging from progressive forms to stationary macular phenotypes in some families. Conversely, CDHR1 mutations, such as truncating variants, are linked to progressive cone-rod involvement with early and central vision loss. GUCY2D mutations typically correlate with isolated cone dysfunction and early severe . Genetic heterogeneity is pronounced, with over 30 s involved across inheritance modes, and modifier s influencing phenotypic variability, such as age of onset and rod involvement extent, even within families sharing the same . This complexity underscores the role of oligogenic effects in modulating disease severity.

Diagnosis

Clinical Evaluation

The clinical evaluation of cone dystrophy begins with a detailed patient history to identify key features suggestive of the condition. Patients often report an onset of symptoms in childhood or early adulthood, though late-onset cases can occur after age 50. A family history of vision loss is frequently elicited, as cone dystrophy exhibits heterogeneous patterns including autosomal dominant, autosomal recessive, and X-linked forms. Associated symptoms such as , reduced central vision, and (day blindness) are commonly described, with progression varying from slow to more rapid loss of visual function over years. Visual acuity testing is a cornerstone of the initial , typically performed using Snellen charts or the Early Treatment Study (ETDRS) protocol to quantify central sharpness. In cone dystrophy, best-corrected is often reduced early in the disease, commonly to 20/50 or worse, reflecting foveal cone involvement. Color vision follows, employing standard tests such as Ishihara plates or Farnsworth-Munsell 100-hue to detect dyschromatopsia, which manifests as impaired red-green or blue-yellow discrimination due to cone dysfunction. This abnormality can precede significant acuity loss and aids in early suspicion of cone dystrophy. A comprehensive anterior segment examination via slit-lamp biomicroscopy is essential to rule out confounding factors like cataracts or corneal opacities that could mimic or exacerbate visual symptoms. This is followed by to evaluate the posterior segment for vascular anomalies or other non-retinal pathologies, while noting any expected macular changes consistent with the disease. Family analysis is conducted to map the inheritance pattern, which informs and risk assessment for relatives; for instance, autosomal dominant transmission suggests a 50% chance of inheritance per offspring, while recessive patterns require both parents to be carriers. considerations include distinguishing cone dystrophy from , which presents congenitally with and severe loss but preserved rod function; , characterized by yellow flecks in the fundus and accumulation; and toxic maculopathies, often linked to medication exposure like without familial patterns.

Specialized Tests

Electroretinography (ERG) serves as the gold standard for confirming cone dystrophy by objectively measuring retinal electrical responses to light stimuli. In cone dystrophy, full-field ERG typically reveals markedly reduced or absent cone-specific responses, such as those elicited by 30 Hz flicker stimuli, while rod-mediated responses remain normal or near-normal in early stages. This pattern distinguishes cone dystrophy from rod-cone dystrophies like retinitis pigmentosa, where both cone and rod responses are affected. Optical coherence tomography (OCT) provides high-resolution cross-sectional imaging of the , revealing characteristic structural changes in cone dystrophy. Spectral-domain OCT commonly shows thinning of the outer nuclear layer and disruption or loss of the ellipsoid zone in the , reflecting photoreceptor degeneration. These findings are heterogeneous, often correlating with disease severity, and may include foveal or cystic spaces in advanced cases. Fundus autofluorescence (FAF) imaging assesses the distribution of in the , aiding in the evaluation of cone dystrophy progression. In affected areas, FAF typically demonstrates hypoautofluorescence corresponding to macular due to loss of photoreceptors and reduced accumulation, often presenting as a bull's-eye pattern. Surrounding regions may show hyperautofluorescence in early or perifoveal involvement, highlighting metabolic stress in surviving cells. Genetic testing, particularly through next-generation sequencing (NGS) panels targeting genes, is essential for molecular of cone dystrophy. These panels analyze over 100 genes associated with cone and cone-rod dystrophies, identifying causative in approximately 60-70% of cases depending on the cohort and panel comprehensiveness. of variants often involves for accuracy, enabling precise diagnosis and family counseling. Visual field testing using automated perimetry, such as Humphrey 10-2 or 30-2 protocols, quantifies functional in cone dystrophy by mapping retinal . Patients commonly exhibit central or cecocentral scotomas reflecting macular involvement, with relative sparing of peripheral fields in pure cone forms. These defects correlate with photoreceptor and help delineate the extent of cone dysfunction. Adaptive optics (AO) imaging, primarily utilized in research settings, enables non-invasive visualization and quantification of individual cone photoreceptors. In cone dystrophy, AO scanning laser ophthalmoscopy reveals reduced cone density and mosaic disruption in the central , providing cellular-level insights beyond conventional imaging. This modality supports early detection of subtle photoreceptor abnormalities and monitoring of disease progression at a microstructural scale.

Management

Supportive Care

Supportive care for cone dystrophy primarily involves strategies to alleviate symptoms, optimize remaining , and address associated challenges, as there is no curative available. Optical aids play a key role in managing and low ; tinted lenses, such as dark grey or brown tints, and are commonly prescribed to reduce light sensitivity. For near tasks, magnifiers like 4× dome magnifiers and half-eye spectacle magnifiers are frequently recommended to enhance reading and detail work, while high-contrast materials help improve in daily activities. These devices are tailored during low-vision assessments to compensate for central without addressing the underlying degeneration. Low-vision programs focus on training patients to maximize and adapt to visual impairments. Techniques such as eccentric viewing, where individuals learn to use non-damaged areas for fixation, are effective for those with central scotomas, improving reading speeds and functional independence as demonstrated in studies. aids, including canes or electronic devices, and training in environmental modifications further support navigation and safety, with programs emphasizing integration of these tools into everyday routines to preserve . Genetic counseling is essential for patients and families, given the inherited nature of cone dystrophy, often involving autosomal recessive, dominant, or X-linked patterns. Counselors provide information on pre-symptomatic testing for at-risk relatives and reproductive options, such as fertilization with preimplantation genetic testing (PGT), to inform and carrier status. Testing identifies mutations in genes like ABCA4 or GUCY2D, enabling personalized without altering disease progression. Nutritional support may include antioxidant supplements like or , following guidelines for retinal health, but vitamin A derivatives are contraindicated in cases with ABCA4 mutations due to potential and accelerated degeneration. Recommendations are individualized by to avoid harm while supporting overall eye function through a rich in leafy greens and fruits. Regular monitoring is recommended, with annual visits to track , retinal changes via , and manage secondary issues like dry eyes through lubricants or environmental adjustments. This schedule allows for timely updates to aids and interventions as vision evolves. Psychosocial support addresses the emotional impact of progressive vision loss, with referrals to support groups and counseling services helping patients cope with anxiety, , and lifestyle adaptations. Organizations focused on inherited retinal diseases offer peer networks and resources to foster and community integration.

Emerging Therapies

represents a primary focus in the development of disease-modifying treatments for cone dystrophy, utilizing (AAV)-based vectors to deliver functional copies of mutated genes such as ABCA4, associated with and cone-rod dystrophy, or GUCY2D, linked to (LCA) with prominent cone involvement.01447-8/fulltext) For ABCA4-related forms, SpliceBio's SB-007, a dual-AAV vector system enabling full-length protein expression, entered Phase 1/2 testing in the (NCT06942572) in early 2025, with the first patient dosed in March to assess safety and tolerability in patients with . Similarly, for GUCY2D mutations, Atsena Therapeutics' ATSN-101 subretinal demonstrated safety and modest efficacy improvements in visual function during a Phase 1/2 for LCA, with sustained benefits observed up to 12 months post-treatment.01447-8/fulltext) Additionally, SparingVision's SPVN06, a gene-agnostic AAV therapy targeting in rod-cone dystrophies including cone-predominant forms, is advancing in the Phase 1/2 PRODYGY (NCT05748873), which began dose escalation in 2023 and continues to evaluate progression slowing as of 2025. Stem cell therapies aim to replace degenerated photoreceptors through transplantation of progenitor cells derived from induced pluripotent s (iPSCs). BlueRock Therapeutics' OpCT-001, an iPSC-derived , initiated the Phase 1/2 CLARICO trial (NCT06789445) in July 2025, treating the first patient with primary photoreceptor diseases such as cone-rod dystrophy; early data emphasize safety in replacing lost photoreceptors in advanced disease stages. Optogenetics offers a gene-agnostic strategy by introducing light-sensitive proteins into surviving retinal cells to restore photosensitivity in cone dystrophy patients with advanced photoreceptor loss. Preclinical studies from 2024-2025 have shown promise in animal models of inherited retinal degenerations, where optogenetic expression in bipolar or ganglion cells enabled light-evoked responses mimicking cone function. Clinical preparation for cone-specific optogenetics, such as the EYEconic study (NCT05294978), focused on qualifying vision restoration endpoints in 2025, paving the way for first-in-human trials targeting end-stage cone dystrophies. Pharmacological interventions target secondary cone degeneration mechanisms, such as . , an , has demonstrated potential in preserving cone function; a Phase 1 trial in patients showed improved cone-mediated vision after 6 months of oral , with applicability to cone dystrophy due to shared oxidative pathways. This led to the ongoing Phase 3 NAC Attack trial (NCT05537220), launched in 2024 and recruiting through 2025, to evaluate long-term safety and efficacy in slowing photoreceptor loss. As of November 2025, several Phase 1/2 trials for cone dystrophy therapies are actively recruiting or ongoing, including those for SB-007, OpCT-001, and SPVN06, reflecting accelerated progress in inherited diseases. Key challenges include precise subretinal delivery to the to target cone-rich areas and mitigating immune responses to AAV vectors, which have been addressed in preclinical optimizations but require further clinical validation. Future directions emphasize CRISPR-based editing for direct mutation correction in preclinical models of cone dystrophy. Studies in 2025 have reported efficient editing of ABCA4 variants in nonhuman primate retinas using adenine base editors delivered via AAV, achieving up to 75% correction in cone photoreceptors without off-target effects. These approaches hold potential for personalized therapies but remain in early-stage development.

Prognosis

Disease Progression

Cone dystrophy typically manifests in two forms: stationary and progressive. In the stationary form, which is often congenital or evident in , the condition presents with initial visual impairments that do not worsen over time. In contrast, the progressive form usually begins in late childhood or , with symptoms gradually deteriorating over years to decades, potentially leading to severe vision loss while sparing peripheral fields. The early stage of progressive cone dystrophy is characterized by mild loss of , often starting in the first or second decade of life, accompanied by subtle defects and . These initial changes reflect early cone dysfunction, with showing delayed implicit times in flicker responses, though full-field amplitudes may remain relatively preserved. Stationary cases plateau at this mild level immediately following onset, without further advancement. During the intermediate stage, which spans approximately 10-20 years, cone degeneration accelerates, leading to expansion of central scotomas and intensification of , alongside worsening color discrimination. reveals progressive loss of the ellipsoid zone in the , correlating with declining photopic electroretinogram amplitudes. This phase marks a transition where central vision becomes increasingly impaired, though the rate varies individually. In the late stage, many patients with progressive cone dystrophy reach legal blindness, defined as visual acuity of 20/200 or worse, by around age 48 on average, with some cases transitioning to cone-rod dystrophy involving secondary loss. Fundus changes include macular atrophy and pigmentary alterations, but complete blindness remains uncommon due to preserved rod function. Progression speed is influenced by , with autosomal dominant forms (e.g., due to GUCY2D mutations) often showing faster deterioration compared to autosomal recessive types (e.g., ABCA4), though intrafamilial variability occurs. Environmental factors, such as prolonged exposure to ultraviolet light, may exacerbate cone damage and hasten advancement. Monitoring involves serial assessments of , alongside and to track photoreceptor integrity. These markers help delineate the trajectory from early dysfunction to advanced .

Long-term Outcomes

In cone dystrophy, the visual endpoint typically involves severe impairment of central vision, often reducing acuity to levels equivalent to counting fingers or hand motion at best, while remains relatively preserved, enabling basic navigation but precluding fine visual tasks such as reading or recognizing faces. Legal blindness, defined as best-corrected visual acuity of 20/200 or worse, occurs in the majority of patients, with a mean age of onset around 48 years for isolated cone dystrophy cases. Quality of life is significantly affected, with patients experiencing restrictions in public life in over half of cases and anxiety about progression in about three-quarters. is reported in approximately 16% of individuals with macular and cone-rod dystrophies, and long-term exceeds 9%, with early common in those over 50. Adaptation through visual aids and low-vision rehabilitation helps maintain independence for many, though psychological support is often inadequate, affecting over one-third of patients. Complications include secondary cataracts and in some progressive cases, though rates specific to cone dystrophy are not well-quantified; is not a direct feature but may arise in syndromic forms. No systemic effects occur, but vision loss elevates fall risk due to central field defects and , mirroring increased hazards seen in other macular disorders. Survival remains unaffected, with normal lifespan expected, as vision loss constitutes the primary morbidity and source of disability. Legal blindness develops in 40-60% of cases by midlife, depending on subtype, rendering central vision tasks impossible while preserving mobility. Prognostic factors include , with ABCA4 associated with faster progression to severe vision loss, particularly in autosomal recessive forms. Early onset before age 20 also predicts accelerated decline to legal blindness. Early facilitates better through , potentially mitigating some quality-of-life impacts. Pre-2025 data indicate consistently poor long-term outcomes, with progressive vision deterioration leading to legal blindness in most untreated cases and limited stabilization options. Emerging therapies, such as replacement trials for specific mutations, show early promise for slowing progression but have not yet altered overall endpoints in established cohorts. As of November 2025, ongoing trials including SPVN06 (a -agnostic AAV-based ) and SparingVision's therapies continue to demonstrate preclinical and early clinical potential for preserving cone function.

References

  1. [1]
    Cone dystrophy | About the Disease | GARD
    Progressive cone dystrophy is a general term for a group of rare eye disorders that affect the cone cells of the retina.
  2. [2]
    Progressive cone and cone-rod dystrophies: clinical features ... - PMC
    Jan 24, 2019 · Progressive cone and cone-rod dystrophies are a clinically and genetically heterogeneous group of inherited retinal diseases characterised by cone ...
  3. [3]
    Cone Dystrophy - Symptoms, Causes, Treatment | NORD
    Jun 14, 2021 · Cone dystrophy is a general term used to describe a group of rare eye disorders that affect the cone cells of the retina.
  4. [4]
    Retinal Dystrophies - StatPearls - NCBI Bookshelf - NIH
    Retinal dystrophies or inherited retinal diseases (IRD) are a group of degenerative disorders of the retina with clinical and genetic heterogeneity.
  5. [5]
    Cone rod dystrophy - Orphanet
    Cone-rod dystrophy (CRD) should be distinguished from rod-cone dystrophy (RCD), also known as retinitis pigmentosa. Unlike RCD, which typically start with night ...
  6. [6]
    The Genetic Basis of Rod-Cone versus Cone-Rod Dystrophies | IOVS
    This study shows, in an unbiased way, which IRD genes cause more rod versus cone-specific defects, in many human subjects.
  7. [7]
    The cone dystrophies - PubMed
    The cone dystrophies are a heterogeneous group of inherited disorders that result in dysfunction of the cone photoreceptors and sometimes their post-receptoral ...
  8. [8]
    Cone Dystrophy - an overview | ScienceDirect Topics
    Cone dystrophy (CD) is defined as an inherited disease affecting cone photoreceptors, leading to various forms of stationary or progressive visual ...
  9. [9]
    Cone-rod dystrophy: MedlinePlus Genetics
    ### Summary of Cone Dystrophy Aspects from Cone-Rod Dystrophy Page
  10. [10]
    Cone rod dystrophies | Orphanet Journal of Rare Diseases | Full Text
    Feb 1, 2007 · Epidemiology. Prevalence of CRDs is estimated at 1/40,000 (thus, CRDs are ten times less frequent than RP) [1]. Clinical ...Missing: incidence | Show results with:incidence
  11. [11]
    Clinical and genetic characteristics of 251 consecutive patients with ...
    Mar 19, 2018 · Macular and cone/cone-rod dystrophies (MD/CCRD) demonstrate a broad genetic and phenotypic heterogeneity, with retinal alterations solely or ...<|control11|><|separator|>
  12. [12]
    Whole exome sequencing unveils a frameshift mutation in CNGB3 ...
    (A) Pedigree of the consanguineous Indian family affected with AR cone dystrophy. Squares present males, and circles denote females. Shaded shapes indicate ...2 Methods · 3 Results · 3.4 Discussion And...<|separator|>
  13. [13]
    Progressive cone dystrophy - Orphanet
    Progressive cone dystrophy usually presents in childhood or early adult life, and patients tend to develop rod photoreceptor dysfunction in later life. Synonym ...
  14. [14]
    Cone-rod dystrophy (Concept Id: C4085590) - NCBI
    There are more than 30 types of cone-rod dystrophy, which are distinguished by their genetic cause and their pattern of inheritance: autosomal recessive, ...Missing: subtypes | Show results with:subtypes
  15. [15]
    Cone-Rod Dystrophy: Symptoms & Causes - Cleveland Clinic
    Oct 4, 2024 · Cone-rod dystrophy is a genetic condition that causes light-sensitive cells in your retina to break down. It eventually leads to loss of color, ...Missing: definition | Show results with:definition
  16. [16]
    The cone dysfunction syndromes - British Journal of Ophthalmology
    The cone dysfunction syndromes are a heterogeneous group of inherited, predominantly stationary retinal disorders characterised by reduced central vision.
  17. [17]
  18. [18]
  19. [19]
  20. [20]
    Cone/Cone-rod dystrophy: for professionals - Gene Vision
    Jan 18, 2021 · Prevalence. 1:20,000 to 100,000. Inheritance. Autosomal dominant; Autosomal recessive in majority of cases; X-linked recessive.Missing: epidemiology incidence
  21. [21]
    GUCA1A mutation causes maculopathy in a five-generation family ...
    Jan 26, 2017 · In this study we found a novel GUCA1A mutation to be disease causative in a five-generation family affected with variable maculopathies ranging ...
  22. [22]
    CDHR1-Related Cone–Rod Dystrophy - PubMed Central - NIH
    Feb 17, 2023 · We demonstrate the clinical characteristics, retinal imaging outcomes, and genetic test results of a patient with CRD15.
  23. [23]
    Cone Dysfunction in Patients With Late-Onset Cone Dystrophy and ...
    In most patients, the first symptoms occur between the first and the third decade of life. Later onset, in the fourth or even after the sixth decade, is rare.
  24. [24]
    Cone rod dystrophies - PMC - NIH
    Cone rod dystrophies (CRDs) (prevalence 1/40000) are inherited retinal dystrophies that belong to the group of pigmentary retinopathies.
  25. [25]
    Top IRDs to Watch: Cone and Cone-Rod Dystrophies - Retina Today
    This is in contrast to retinitis pigmentosa, or rod-cone dystrophy, where the earliest symptoms are typically nyctalopia and peripheral vision loss (Table).
  26. [26]
    Cone Dystrophy in a 33-Year-Old Woman - - Modern Optometry
    Sep 3, 2024 · The external slit-lamp and dilated fundus examinations initially appeared unremarkable (Figure 1). Figure 1. Color retinal photography of ...
  27. [27]
    Cone/Cone-rod dystrophy: for patients - Gene Vision
    Nov 30, 2020 · Females inherit one X chromosome from each parent. As a result, males are usually affected in conditions inherited in an X-linked manner as they ...Missing: distribution | Show results with:distribution
  28. [28]
    A Boy Without Color Vision - American Academy of Ophthalmology
    Jul 1, 2015 · Bull's-eye changes on FAF combined with loss of color vision on exam make cone dystrophy the most likely diagnosis.Missing: pupillary | Show results with:pupillary
  29. [29]
    Cone Dystrophy - an overview | ScienceDirect Topics
    At least nine genes have been identified: CRX, GUCY2D, AIPL1, GUCA1A, GCAP1, RIMS1, and UNC119. The autosomal-recessive cone–rod dystrophy locus is 1p21–p13; ...
  30. [30]
    Cone Dystrophies: An Optical Coherence Tomography Angiography ...
    May 16, 2020 · Full-field electroretinogram (ERG) is considered the gold standard for diagnosis of CDs, revealing marked depression of cone responses with ...
  31. [31]
  32. [32]
  33. [33]
    Morphologic Characteristics of the Outer Retina in Cone Dystrophy ...
    This study described in depth and categorized the outer retinal morphologic abnormalities in cone dystrophy patients using SD-OCT.
  34. [34]
    Spectral-domain optical coherence tomography morphological ...
    SD-OCT imaging shows extensive morphologic findings in cone dystrophy, which can be utilized to evaluate/diagnose these patients and may serve as predictive ...
  35. [35]
    Fundus autofluorescence in cone dystrophy - PMC - PubMed Central
    Increased autofluorescence either in the perifoveal area or central macular area is a non-specific manifestation of cone dystrophy that can occur in other forms ...
  36. [36]
    Progressive cone and cone-rod dystrophies: clinical features ...
    Progressive cone and cone-rod dystrophies are a clinically and genetically heterogeneous group of inherited retinal diseases characterised by cone photoreceptor ...
  37. [37]
    Next-generation sequencing applied to a large French cone and ...
    Jun 24, 2015 · We applied Next-Generation Sequencing targeting a panel of 123 genes implicated in retinal diseases to 96 patients. A systematic filtering ...
  38. [38]
  39. [39]
    Peripheral Cone Dystrophy: Expanded Clinical Spectrum ...
    Jul 11, 2018 · Humphrey 30-2 Sita Standard and Goldmann threshold visual field testing confirmed bilateral paracentral scotomas in all patients that remained ...3. Results · 3.3. Visual Fields · 3.5. Fluorescein Angiography<|separator|>
  40. [40]
    High Resolution Imaging of Cone–Rod Dystrophy With Adaptive ...
    Conclusions: Here we show that adaptive optics ophthalmoscopy can be used to directly observe retinal pathology, such as photoreceptor loss, that is otherwise ...
  41. [41]
    High-Resolution Retinal Imaging of Cone–Rod Dystrophy
    Here we use adaptive optics ophthalmoscopy to image cone–rod dystrophy in vivo and compare these results with standard clinical tests. Design. Observational ...
  42. [42]
    Study Details | NCT06942572 | ClinicalTrials.gov - Clinical Trials
    This Phase 1/2 study will evaluate the safety, tolerability, and preliminary efficacy of subretinal SB-007 administration to determine dose selection in ...
  43. [43]
    SpliceBio Announces First Patient Dosed in Phase 1/2 ASTRA Study ...
    Mar 13, 2025 · SB-007 addresses the root cause of Stargardt disease with the potential to treat all patients across all ABCA4 mutations ...
  44. [44]
  45. [45]
    BlueRock Therapeutics announces first patient receives ...
    Jul 8, 2025 · “We are excited to announce the first patient in the CLARICO trial, the first-ever clinical trial for an iPSC-derived treatment in this field.”.
  46. [46]
    Optogenetic therapy for retinal degenerative diseases: A review
    Apr 21, 2025 · Retinal degeneration, a key indication for optogenetics, progresses through three main phases: rod degeneration, cone photoreceptor degeneration ...
  47. [47]
    EyeConic: Qualification for Cone-Optogenetics | MedPath
    Jul 31, 2025 · This study aims to prepare for the first-in-human clinical trial of cone optogenetics vision restoration. As a first step, this worldwide ...
  48. [48]
    Oral N-acetylcysteine improves cone function in retinitis pigmentosa ...
    A randomized, placebo-controlled trial is needed to determine if oral NAC can provide long-term stabilization and/or improvement in visual function in patients ...
  49. [49]
    Oral N-acetylcysteine for Retinitis Pigmentosa | ClinicalTrials.gov
    N-acetylcysteine (NAC) reduces oxidative stress and in animal models of RP it slowed cone degeneration. In a phase I clinical trial in patients with RP, NAC ...
  50. [50]
    Preclinical safety and biodistribution of SPVN06, a novel gene - Nature
    Aug 4, 2025 · Rod-cone dystrophies (RCD) are a group of rare, inherited retinal diseases characterized by the progressive loss of rod and cone photoreceptors ...
  51. [51]
    Advancements in CRISPR-based therapies for ocular pathologies
    An ABE strategy achieves high-level ABCA4 gene correction in nonhuman primates, with mean editing rates of 75% in cone photoreceptors and 87% in RPE cells. [147].<|control11|><|separator|>
  52. [52]
    Shining light on CRISPR/Cas9 therapeutics for inherited retinal ...
    In this review, we summarise the recent preclinical advances and further potential of CRISPR/Cas9 technologies to target RP, explore the avenues of delivering ...
  53. [53]
    Clinical Course, Genetic Etiology, and Visual Outcome in Cone and ...
    Jan 20, 2012 · Ten years after diagnosis, 35% of CD and 51% of CRD had a bull's eye maculopathy; 70% of CRD showed absolute peripheral visual field defects and ...
  54. [54]
    Patient-Reported Social Impact of Molecularly Confirmed Macular ...
    Most patients, however, experience symptoms within the first decades of life. Progressive loss of visual acuity, central visual field defects, and glare may ...Missing: prognosis | Show results with:prognosis
  55. [55]
    What is Cone-Rod Dystrophy and What Are Its Effects? - Vision Buddy
    Dec 24, 2024 · The risk of trips, falls, and collisions increases, turning a simple walk down the street into a cautious endeavor. Navigating crowded ...