Leber congenital amaurosis

Last updated
Leber congenital amaurosis
Other namesLeber's congenital amaurosis
Specialty Ophthalmology   OOjs UI icon edit-ltr-progressive.svg
Symptoms Visual impairment, sensitivity to light [1]
Types> 12 types [1]
Causes Genetic (autosomal recessive) [1]
Frequency1 in 40,000 newborns [1]

Leber congenital amaurosis (LCA) is a rare inherited eye disease that appears at birth or in the first few months of life. [2]

Contents

It affects about 1 in 40,000 newborns. [1] LCA was first described by Theodor Leber in the 19th century. [3] [4] It should not be confused with Leber's hereditary optic neuropathy, which is a different disease also described by Theodor Leber.

One form of LCA was successfully treated with gene therapy in 2008. [5] [6] [7] [8]

Signs and symptoms

LCA symptoms typically begin in the first few months of life, most commonly with involuntary twitching of the eye (nystagmus). Affected infants may show misaligned eyes when looking at something (strabismus), aversion to light (photophobia), and poke or rub at their eyes (Franceschetti’s oculodigital sign). [9] Those with LCA invariably experience vision problems. Affected infants show decreased visual response to objects. Loss of visual acuity is severe, with affected individuals' vision ranging from 20/200 to 20/400. [note 1] Around a third of those affected completely lose perception of light. [10]

At an eye exam, the pupils may not respond normally to light. Some affected individuals have cloudy eyes (cataracts), and irregularly shaped corneas (keratoconus). [9] Retinal exams typically look normal, especially in the young, though retinal abnormalities can appear later in life. [10]

Aside from eye problems, children with LCA are typically healthy. [11]

Cause

LCA is a genetic disease, and can be caused by pathogenic variants in at least 28 different genes. [12] Variants in three of these genes – IMPDH1 , OTX2 , and CRX – can cause LCA in an autosomal dominant manner, meaning inheriting a single copy of a pathogenic variant can result in disease. Variants in the remaining genes associated with LCA cause disease in an autosomal recessive manner, meaning one must inherit copies of the pathogenic variant from both parents to develop LCA. [12] Genes associated with LCA have a variety of roles in the develoment of the eye:

Pathogenic variants of any of these genes cause dysfunction in those associated processes, which leads to severe vision loss. Variants in DTHD1 and NMNAT1 also cause LCA, though these genes' roles in vision development are not yet known. [13]

Among the gene variants that cause LCA, CEP290 and GUCYD variants are the most common, each causing up to 20% of LCA cases. Other common variants are in CRB1 (around 10% of cases), RPE65 (up to 10%), AIPL1 (up to 8%), RDH12 (up to 5%), and RPGRIP1 (around 5%). [14] Around 25% of people with LCA do not have any of the known LCA-causing pathogenic gene variants; the cause of their LCA is unknown. [12]

Diagnosis

LCA is diagnosed clinically, by a combination of vision loss, abnormal response of the pupils to light, and by abnormal response to electroretinography, a test that measures the electrical response of the retina to light. [10]

Treatment

One form of LCA, in patients with LCA2 bearing a mutation in the RPE65 gene, has been successfully treated in clinical trials using gene therapy. The results of three early clinical trials were published in 2008 demonstrating the safety and efficacy of using adeno-associated virus to deliver gene therapy to restore vision in LCA patients. In all three clinical trials, patients recovered functional vision without apparent side effects. [5] [6] [7] [8] These studies, which used adeno-associated virus, have spawned a number of new studies investigating gene therapy for human retinal disease.[ citation needed ] On 19 December 2017, the U.S. Food and Drug Administration approved voretigene neparvovec-rzyl (Luxturna), an adeno-associated virus vector-based gene therapy for children and adults with biallelic RPE65 gene mutations responsible for retinal dystrophy, including Leber congenital amaurosis. Patients must have viable retinal cells as a prerequisite for the intraocular administration of Luxturna. [15]

For those who cannot benefit from gene therapy, LCA treatment is supportive, meant to facilitate living with visual impairment. Some benefit from vision aids such as glasses, magnifiers, and enhancers. Other resources helpful for those with visual impairment include educational programs, special education teachers, and service animals. [16]

Epidemiology

Around 2–3 out of every 100,000 people have LCA – an estimated 180,000 people worldwide. [17] LCA is a common cause of blindness in the young; around 20% of children in schools for the blind have LCA. [18]

History

LCA was originally described as a variety retinitis pigmentosa by Theodor Leber in 1869. [17]

Notable cases

See also

Notes

  1. With 20/200 vision, one can clearly see at 20 feet what would typically be seen clearly at 200 feet.

    Related Research Articles

    <span class="mw-page-title-main">Retina</span> Part of the eye

    The retina is the innermost, light-sensitive layer of tissue of the eye of most vertebrates and some molluscs. The optics of the eye create a focused two-dimensional image of the visual world on the retina, which then processes that image within the retina and sends nerve impulses along the optic nerve to the visual cortex to create visual perception. The retina serves a function which is in many ways analogous to that of the film or image sensor in a camera.

    <span class="mw-page-title-main">Retinitis pigmentosa</span> Gradual retinal degeneration leading to progressive sight loss

    Retinitis pigmentosa (RP) is a genetic disorder of the eyes that causes loss of vision. Symptoms include trouble seeing at night and decreasing peripheral vision. As peripheral vision worsens, people may experience "tunnel vision". Complete blindness is uncommon. Onset of symptoms is generally gradual and often begins in childhood.

    <span class="mw-page-title-main">Retinoschisis</span> Eye disease involving splitting of the retina

    Retinoschisis is an eye disease characterized by the abnormal splitting of the retina's neurosensory layers, usually in the outer plexiform layer. Retinoschisis can be divided into degenerative forms which are very common and almost exclusively involve the peripheral retina and hereditary forms which are rare and involve the central retina and sometimes the peripheral retina. The degenerative forms are asymptomatic and involve the peripheral retina only and do not affect the visual acuity. Some rarer forms result in a loss of vision in the corresponding visual field.

    Amaurosis is vision loss or weakness that occurs without an apparent lesion affecting the eye. It may result from either a medical condition or excess acceleration, as in flight. The term is the same as the Latin gutta serena, which means, in Latin, clear drop. Gutta serena is a condition of partial or complete blindness with a transparent, clear pupil. This term contrasts with suffusio nigra which means, in Latin, dark suffusion, indicating partial or complete blindness with a dark pupil, e.g., a cataract. Milton, already totally blind for twelve years by the time he published Paradise Lost, refers to these terms in Book 3, lines 25–26.

    <span class="mw-page-title-main">Leber's hereditary optic neuropathy</span> Mitochondrially inherited degeneration of retinal cells in human

    Leber's hereditary optic neuropathy (LHON) is a mitochondrially inherited degeneration of retinal ganglion cells (RGCs) and their axons that leads to an acute or subacute loss of central vision; it predominantly affects young adult males. LHON is transmitted only through the mother, as it is primarily due to mutations in the mitochondrial genome, and only the egg contributes mitochondria to the embryo. Men cannot pass on the disease to their offspring. LHON is usually due to one of three pathogenic mitochondrial DNA (mtDNA) point mutations. These mutations are at nucleotide positions 11778 G to A, 3460 G to A and 14484 T to C, respectively in the ND4, ND1 and ND6 subunit genes of complex I of the oxidative phosphorylation chain in mitochondria.

    <span class="mw-page-title-main">Choroideremia</span> Medical condition

    Choroideremia is a rare, X-linked recessive form of hereditary retinal degeneration that affects roughly 1 in 50,000 males. The disease causes a gradual loss of vision, starting with childhood night blindness, followed by peripheral vision loss and progressing to loss of central vision later in life. Progression continues throughout the individual's life, but both the rate of change and the degree of visual loss are variable among those affected, even within the same family.

    The visual cycle is a process in the retina that replenishes the molecule retinal for its use in vision. Retinal is the chromophore of most visual opsins, meaning it captures the photons to begin the phototransduction cascade. When the photon is absorbed, the 11-cis retinal photoisomerizes into all-trans retinal as it is ejected from the opsin protein. Each molecule of retinal must travel from the photoreceptor cell to the RPE and back in order to be refreshed and combined with another opsin. This closed enzymatic pathway of 11-cis retinal is sometimes called Wald's visual cycle after George Wald (1906–1997), who received the Nobel Prize in 1967 for his work towards its discovery.

    <span class="mw-page-title-main">RPE65</span> Protein-coding gene in humans

    Retinal pigment epithelium-specific 65 kDa protein, also known as retinoid isomerohydrolase, is an enzyme of the vertebrate visual cycle that is encoded in humans by the RPE65 gene. RPE65 is expressed in the retinal pigment epithelium and is responsible for the conversion of all-trans-retinyl esters to 11-cis-retinol during phototransduction. 11-cis-retinol is then used in visual pigment regeneration in photoreceptor cells. RPE65 belongs to the carotenoid oxygenase family of enzymes.

    <span class="mw-page-title-main">AIPL1</span> Protein-coding gene in the species Homo sapiens

    Aryl-hydrocarbon-interacting protein-like 1 is a protein that in humans is encoded by the AIPL1 gene.

    <span class="mw-page-title-main">RDH12</span> Protein-coding gene in the species Homo sapiens

    Retinol dehydrogenase 12 is an enzyme that in humans is encoded by the RDH12 gene.

    <span class="mw-page-title-main">Foundation Fighting Blindness</span>

    The mission of the Foundation Fighting Blindness is to fund research that will lead to the prevention, treatment and cures for the entire spectrum of retinal degenerative diseases, including retinitis pigmentosa, macular degeneration, Usher syndrome, Stargardt disease and related conditions. These diseases, which affect more than 10 million Americans and millions more throughout the world, often lead to severe vision loss or complete blindness.

    Gene therapy using lentiviral vectors was being explored in early stage trials as of 2009.

    <span class="mw-page-title-main">The Llura Liggett Gund Award</span> Medical award

    The Llura Liggett Gund Award honors researchers for career achievements that have significantly advanced the research and development of preventions, treatments and cures for eye disease.

    Gene therapy for color blindness is an experimental gene therapy of the human retina aiming to grant typical trichromatic color vision to individuals with congenital color blindness by introducing typical alleles for opsin genes. Animal testing for gene therapy began in 2007 with a 2009 breakthrough in squirrel monkeys suggesting an imminent gene therapy in humans. While the research into gene therapy for red-green colorblindness has lagged since then, successful human trials are ongoing for achromatopsia. Congenital color vision deficiency affects upwards of 200 million people in the world, which represents a large demand for this gene therapy.

    Retinal gene therapy holds a promise in treating different forms of non-inherited and inherited blindness.

    <span class="mw-page-title-main">Robert MacLaren</span> British ophthalmologist

    Robert E. MacLaren FMedSci FRCOphth FRCS FACS VR is a British ophthalmologist who has led pioneering work in the treatment of blindness caused by diseases of the retina. He is Professor of Ophthalmology at the University of Oxford and Honorary Professor of Ophthalmology at the UCL Institute of Ophthalmology. He is a Consultant Ophthalmologist at the Oxford Eye Hospital. He is also an Honorary Consultant Vitreo-retinal Surgeon at the Moorfields Eye Hospital. MacLaren is an NIHR Senior Investigator, or lead researcher, for the speciality of Ophthalmology. In addition, he is a member of the research committee of Euretina: the European Society of Retina specialists, Fellow of Merton College, in Oxford and a Fellow of the Higher Education Academy.

    Voretigene neparvovec, sold under the brand name Luxturna, is a gene therapy medication for the treatment of Leber congenital amaurosis.

    <span class="mw-page-title-main">Spark Therapeutics</span> American pharmaceutical company

    Spark Therapeutics, Inc. is a developer of gene therapy treatments, which treat debilitating genetic diseases. It is a subsidiary of Hoffmann-La Roche.

    <span class="mw-page-title-main">Congenital blindness</span>

    Congenital blindness refers to blindness present at birth. Congenital blindness is sometimes used interchangeably with "Childhood Blindness." However, current literature has various definitions of both terms. Childhood blindness encompasses multiple diseases and conditions present in ages up to 16 years old, which can result in permanent blindness or severe visual impairment over time. Congenital blindness is a hereditary disease and can be treated by gene therapy. Visual loss in children or infants can occur either at the prenatal stage or postnatal stage. There are multiple possible causes of congenital blindness. In general, 60% of congenital blindness cases are contributed from prenatal stage and 40% are contributed from inherited disease. However, most of the congenital blindness cases show that it can be avoidable or preventable with early treatment.

    Jean Bennett is the F. M. Kirby Professor of Ophthalmology in the Perelman School of Medicine at the University of Pennsylvania. Her research focuses on gene therapy for retinal diseases. Her laboratory developed the first FDA approved gene therapy for use in humans, which treats a rare form of blindness. She was elected a member of the National Academy of Sciences in 2022.

    References

    1. 1 2 3 4 5 "Leber congenital amaurosis". Genetics Home Reference. August 2010. Retrieved 14 May 2017.
    2. Stone EM (December 2007). "Leber congenital amaurosis - a model for efficient genetic testing of heterogeneous disorders: LXIV Edward Jackson Memorial Lecture". American Journal of Ophthalmology. 144 (6): 791–811. doi: 10.1016/j.ajo.2007.08.022 . PMID   17964524.
    3. Leber's congenital amaurosis at Who Named It?
    4. Leber T (1869). "Über Retinitis pigmentosa und angeborene Amaurose". Archiv für Ophthalmologie (in German). 15 (3): 1–25. doi:10.1007/BF02721213. S2CID   543893.
    5. 1 2 Maguire AM, Simonelli F, Pierce EA, Pugh EN, Mingozzi F, Bennicelli J, et al. (May 2008). "Safety and efficacy of gene transfer for Leber's congenital amaurosis". The New England Journal of Medicine. 358 (21): 2240–8. doi:10.1056/NEJMoa0802315. PMC   2829748 . PMID   18441370.
    6. 1 2 Simonelli F, Maguire AM, Testa F, Pierce EA, Mingozzi F, Bennicelli JL, et al. (March 2010). "Gene therapy for Leber's congenital amaurosis is safe and effective through 1.5 years after vector administration". Molecular Therapy. 18 (3): 643–50. doi:10.1038/mt.2009.277. PMC   2839440 . PMID   19953081.
    7. 1 2 Cideciyan AV, Hauswirth WW, Aleman TS, Kaushal S, Schwartz SB, Boye SL, et al. (August 2009). "Vision 1 year after gene therapy for Leber's congenital amaurosis". The New England Journal of Medicine. 361 (7): 725–7. doi:10.1056/NEJMc0903652. PMC   2847775 . PMID   19675341.
    8. 1 2 Bainbridge JW, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K, et al. (May 2008). "Effect of gene therapy on visual function in Leber's congenital amaurosis". The New England Journal of Medicine. 358 (21): 2231–9. CiteSeerX   10.1.1.574.4003 . doi:10.1056/NEJMoa0802268. PMID   18441371.
    9. 1 2 Schmitt, Ohns & DeVries 2023, "Introduction".
    10. 1 2 3 Schmitt, Ohns & DeVries 2023, "Diagnosing LCA".
    11. Schmitt, Ohns & DeVries 2023, "Etiology of LCA".
    12. 1 2 3 4 Kondkar & Abu-Amero 2019, "Genetic basis of LCA".
    13. 1 2 Kondkar & Abu-Amero 2019, "Table 1. Overview of causal genes implicated in Leber congential amaurosis".
    14. Kondkar & Abu-Amero 2019, "Table 2. Commonly affected genes in LCA and their associated phenotypes.".
    15. "Approved Products - LUXTURNA". FDA. 2019-04-05.
    16. Schmitt, Ohns & DeVries 2023, "Treatment of LCA".
    17. 1 2 Kondkar & Abu-Amero 2019, "Epidemiological, historical and clinical perspective of LCA".
    18. "Leber Congenital Amaurosis". American Association for Pediatric Ophthalmology and Strabismus. April 2023. Retrieved 1 May 2023.
    19. "4 yr old Gavin using his white cane to navigate down a curb independently". YouTube . Archived from the original on 2021-12-21.

    Works cited

    Further reading