ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Age-related macular degeneration

Age-related macular degeneration
Literature review current through: Jan 2024.
This topic last updated: Jan 10, 2024.

INTRODUCTION — Age-related macular degeneration (AMD) is a common cause of severe central vision loss and legal blindness in adults. The etiology, diagnosis, and treatment of AMD will be reviewed here.

DEFINITION AND CLASSIFICATION — AMD is a degenerative disease of the photoreceptors of the central portion of the retina (the macula) and the supporting retinal pigment epithelium [1-3]. It is characterized by loss of central vision.

For clinical purposes, AMD is classified as either "dry" or "wet". The more common form (affecting approximately 75 percent of patients) is dry AMD (also known as nonexudative or nonneovascular AMD). The less common form is wet AMD (also known as exudative or neovascular AMD).

Both forms of the disease are characterized by the presence of lipid-rich extracellular deposits under the retinal pigment epithelium called drusen, as well as retinal pigmentary and atrophic changes [4,5]. Wet AMD is characterized by new vessel formation in and under the retina [3,5]. These abnormal blood vessels have a tendency to leak, leading to collections of fluid and/or blood in and/or beneath the retina.

Dry AMD progresses to wet AMD in a minority of patients. The risk of developing wet AMD in people with bilateral, early, dry AMD (bilateral soft drusen) was estimated at approximately 3 per 100 person-years if both eyes have early- or intermediate-stage AMD [6].

Dry AMD can also progress to an advanced stage called geographic atrophy in approximately 15 percent of cases. Geographic atrophy and wet AMD are considered advanced forms of the disease and are responsible for most of the severe vision loss in AMD.

There is no consensus for classification and staging of AMD. Several classification schemes have been developed based on the appearance of the fundus. The most common one comes from the Age-Related Eye Disease Study (AREDS) [7], ranging from zero to four, with points assigned based on patient factors such as the presence of large drusen or pigment abnormalities [8,9]. Another classification scheme comes from the Beckman Initiative for Macular Research Classification Committee, with three stages of the disease and one for a normal, aging phenotype [10]. In clinical practice, the early/intermediate stage is clearly distinct from the universally accepted advanced wet AMD or geographic atrophy. However, the distinction between early and intermediate is less formal and could be based on patient symptoms and/or examination findings. (See 'Staging' below.)

EPIDEMIOLOGY — AMD is the most common cause of severe central vision loss and legal blindness among adults in the Western world [11]. The estimated worldwide prevalence of AMD is approximately 8 to 9 percent, affecting approximately 190 million people (18 million in the United States) [12-15]. It is predicted that worldwide, almost a quarter billion people will be affected by this disease by 2040 [12]. The incidence and prevalence increase with age. A study from 2004 of the United States population showed an overall prevalence of 1.5 percent that increases by tenfold (15 percent) for those over the age of 80 [16,17]. A study among European patients showed prevalence of early AMD increasing from 3.5 percent among people aged 55 to 59 to over 17 percent for those over the age of 85 [18].

PATHOGENESIS — The pathogenesis of AMD remains poorly understood. It is clearly a multifactorial, polygenic disease [1,2,19-23]. Aging remains the single most important risk factor [24-26]. Whole-genome sequencing has identified a set of 52 common variants mapped to 34 loci, with most notable changes in chromosome 1 (complement factor H locus) [27-31] and chromosome 10 (ARMS2/HTRA1 locus) [19]. Other factors identified to be involved in AMD pathogenesis include lipid dyshomeostasis [4,32], impaired autophagy [33-36], increased oxidative stress [37,38], inflammation/para-inflammation [39-41], local iron overload/ferroptosis [42], and accumulation of toxic drusen-associated materials [43-46].

RISK FACTORS — Important nonmodifiable risk factors include older age and genetic risk, while modifiable risk factors include smoking and type of diet (low dietary intake of antioxidants) [47,48]. Epidemiologic studies have shown that current smokers are twice as likely to have AMD compared with nonsmokers, whereas ex-smokers had significantly less risk and people who quit smoking >20 years previously were not at increased risk [49]. A Mediterranean diet that includes fruits, vegetables, and fish is associated with a lower risk of AMD [50-57].

The data on the relationship between blood pressure and AMD are not strong or reproducible [58,59]. Similarly, the relationship between elevated lipid or other cardiovascular risk factors and AMD remains controversial [60].

CLINICAL PRESENTATION — AMD may be asymptomatic in early stages. Patients tend to present with problems in contrast sensitivity and adaptation to dark environments. They may complain that they need brighter lighting and more time to read and that their visual function is worse in situations with low contrast like rainy or overcast days. They will also be affected on bright days when they go from a bright to a dark environment (ie, when driving and entering a tunnel) or when they go from a brightly lit room to a darkened one. Eventually, as the disease progresses, their central visual acuity declines and they have trouble not only in reading but in recognizing faces and facial expressions of other people. In almost all patients, the peripheral visual function will remain very similar to that of unaffected individuals. Some patients may also notice distortion of straight edges such as doors or window blinds. Sudden onset of this later symptom requires urgent examination within few days to a week since it could herald progression to the advanced wet form.

It is estimated that approximately 15 percent of patients will become legally blind (central visual acuity less than 20/200), but complete blindness is not a typical outcome of this disease. The disease affects both eyes, but it can be asymmetric. If one eye converts to the advanced stage of the disease, the other eye has a 50 percent chance of advancing to that stage within five years [8,9].

EVALUATION

Primary care evaluation

History — In any patient presenting with symptoms of visual disturbance, the history should include age and the rate of vision loss; whether one or both eyes are involved; and whether the vision loss is for distance vision, near vision, or both. Patients with an acute distortion or loss of central vision may have an initial presentation of wet AMD. The evaluation of the patient with acute vision loss is discussed separately. (See "Amaurosis fugax (transient monocular or binocular visual loss)" and "Approach to the adult with acute persistent visual loss".)

Further questions should inquire about other symptoms suggestive of AMD:

Is there a need for greater contrast sensitivity? (Does the patient require brighter sources of light to read?)

Are there difficulties with dark adaptation? (Does the patient have issues going from a bright environment to a dark environment?)

Is greater time needed to process visual information? (Patients will complain that they are not able to see the highway signs until they are very close to them or need more time to discern reading material.)

A family history of eye problems should be taken into account, including age of onset. Earlier onset in family members may suggest greater likelihood or progressive disease for the patient.

Eye examination — Eye examination by the primary care physician is limited to central visual acuity with either a near card or a distance chart, if available. Vision should always be checked by having the patient wear their glasses for the appropriate distance. Additional examination of distortion or scotomas with an Amsler grid can be useful. (figure 1) (see 'Amsler grid' below). An undilated funduscopic examination may reveal drusen or macula hemorrhage.

Primary care providers may be able to send photographs and rarely optical coherence tomography (OCT) images to a specialist. However, images are sometimes of limited quality and there may be little utility of such an approach if a patient will eventually be referred to an eye doctor.

Ophthalmologic evaluation and diagnosis — AMD is a clinical diagnosis based on the presence of characteristic findings on dilated eye examination using a slit-lamp instrument (biomicroscopy):

In dry AMD, drusen are visible on dilated eye examination. Patches of geographic atrophy of the retina may be evident as areas of depigmentation; increased pigmentation may be seen with retinal pigment epithelium pigmentary mottling.

In wet AMD, dilated examination may reveal subretinal fluid and/or hemorrhage. Wet AMD appears as a grayish-green discoloration in the macular area. The presence of subretinal hemorrhage or a gray subretinal membrane is strongly suggestive of wet AMD. These patients require an urgent assessment by an eye care specialist trained to treat wet AMD where a thorough evaluation and appropriate management can be instituted.

Additional specialized imaging of the retina includes color photograph of the retina, autofluorescence imaging of the retina, and an OCT. Depending on the suspected stage (intermediate to advanced wet AMD), a fluorescein angiogram or an indocyanine green angiogram, as well as OCT angiography may also be obtained to evaluate for the presence of wet AMD. Functional tests such as contrast sensitivity, dark adaptation, and microperimetry are not commonly available in a clinical setting.

Staging — Staging may be based on visual symptoms, as follows:

Early stage:

Mild vague visual complaints

Issues with low contrast

Issues with adapting from bright to dark environments

More light needed for reading

Intermediate stage:

Distortion

Blurriness

Mild to moderate decreased visual acuity

More significant loss of contrast sensitivity

More prolonged dark adaptation times

Advanced stage:

Loss of central visual acuity

Significant distortion

Significant central/paracentral scotomas

Legal blindness

ADVICE FOR ALL PATIENTS

Smoking cessation — All patients should be encouraged to quit smoking or to avoid initiating smoking due to the increased risk of development or progression of AMD [49,58].

Diet/alcohol — Increasing evidence suggests that a Mediterranean diet may decrease the risk of developing advanced AMD [50-57]. In one of the largest studies, the association between adherence to a Mediterranean diet over the prior 12 months and prevalence of AMD (evaluated by eye examination and digital retinal color photography) was assessed among 4753 older adults in seven European countries [55]. Participants' diets were categorized according to the Mediterranean Diet Score, scored from zero to nine, with higher scores indicating greater adherence. Compared with those who scored four or less, participants with scores of six or greater had lower odds of wet AMD (odds ratio 0.53, 95% CI 0.27-1.04). The Mediterranean diet is described in detail separately. (See "Healthy diet in adults", section on 'Mediterranean diet'.)

Moderate consumption of alcohol lowers AMD progression risk; however, high alcohol intake may worsen advanced AMD [51,58,61].

Limited role for antioxidant vitamins — There is lack of conclusive evidence about the benefit of multivitamin supplements in AMD. Although multivitamin supplements are heavily marketed for people with AMD, evidence for effect on the progression of AMD is equivocal and they may have harmful effects [62-65].

Vitamin supplements are not appropriate for people with early stages of the disease or bilateral advanced disease.

We suggest daily vitamin supplements, consistent with the Age-Related Eye Disease Study 2 (AREDS2) formula, can be considered for patients with intermediate dry AMD or advanced AMD in one eye. However, it is also reasonable not to use these supplements based on limited benefit. Further, if a patient's disease progresses to the point of bilateral advanced disease with end-stage vision, we discontinue vitamin supplements due to lack of benefit.

In 2001, a formula containing high doses of vitamin C, vitamin E, beta-carotene, zinc oxide, and cupric oxide (also known as the AREDS formula) was shown in post hoc subgroup analysis of a phase 3 trial to reduce the risk of progression to wet AMD; however, there was controversy surrounding the results of this study and findings have not been replicated [7,62-64].

Further, the AREDS formula had some safety concerns due to the inclusion of high levels of vitamin A and zinc. High vitamin A is associated with increased risk of lung cancer in smokers [66-71], and the 100 mg/day of supplemental zinc is associated with advanced prostate cancer in the Health Professionals Follow-up Study [72]. Thus, a new formula (AREDS2) was developed with reduced zinc content (25 mg) and no vitamin A (lutein + zeaxanthin as carotenoid replacement).

In the AREDS2 trial, 4203 participants at risk for progression to advanced AMD were randomized to a placebo control, lutein and zeaxanthin only, omega 3 fatty acids only, and lutein/zeaxanthin plus omega 3 fatty acids in addition to various modifications of the original AREDS formulation [73]. After a five-year follow-up, none of the treatment options resulted in a greater probability of reduction in progression to advanced AMD when compared with placebo (29, 31, 30, and 31 percent, respectively). However, in a further randomization, eliminating beta-carotene from the AREDS formulation resulted in fewer cases of lung cancer (11 [0.9 percent] versus 23 [2 percent]).

In a systematic review of trials, antioxidant vitamin and mineral supplementation did not prevent, but might delay, the onset of AMD and there was little effect from lutein and zeaxanthin alone [65].

WET AGE-RELATED MACULAR DEGENERATION TREATMENT — Early treatment of wet AMD is associated with better outcomes. In the early stage of the disease, the chance of conversion to wet AMD is less than 5 percent over five years. The chance of conversion to wet AMD increases to approximately 50 percent over five years when the disease is in the "advanced intermediate" stage [74-76].

Our approach — For most patients with wet AMD, we initiate treatment with vascular endothelial growth factor (VEGF) inhibitors or VEGF-angiopoietin-2 (VEGF-Ang2) inhibitors, for example intravitreal bevacizumab, ranibizumab, aflibercept, or faricimab. We do not routinely offer brolucizumab, due to the risk of occlusive vasculitis. In rare cases, we may use photodynamic therapy (PDT; polypoidal variety of wet AMD).

VEGF inhibitors — VEGF is a potent mitogen and vascular permeability factor that plays a pivotal role in wet AMD. Anti-VEGF treatments are the main therapies for wet AMD and have revolutionized management due to their ability to limit progression of wet AMD and stabilize, or reverse, visual loss [3,77].

Treatment with these agents should be initiated as soon after the diagnosis of wet AMD is made. Delay in the initiation of VEGF therapy (greater than 21 weeks compared with less than seven weeks) after first symptoms of wet AMD has been associated with poorer vision outcomes [78]. In practice, most retina specialists would prefer to treat patients within a week of symptom initiation.

Several anti-VEGF molecules have been developed that can limit the destructive effects of neovascularization in patients with AMD. The best studied include intravitreal ranibizumab, aflibercept, faricimab, and bevacizumab [79-82].

In a meta-analysis of six trials (2667 patients), patients treated with anti-VEGF therapy (bevacizumab or ranibizumab) were more likely to have improvement in visual acuity (defined as a gain of ≥15 letters on the vision chart) after one year compared with those in the placebo or control group (18 versus 4 percent, respectively; risk ratio 4.19, 95% CI 2.3-7.6) [77]. Patients treated with anti-VEGF also had greater improvements in vision-related quality-of-life scores (mean difference 6.7 points on a 100-point scale). Rates of serious adverse ocular events in these trials were low, the most serious of them being an infection or retinal detachment, which occurred at a rate of 1/4000 injections or less. Systemic serious adverse events were closer to one to two percent and very similar to the control group.

Ranibizumab, aflibercept, and bevacizumab have similar efficacy. For example, ranibizumab and bevacizumab have similar visual and safety outcomes [77,83-86] and aflibercept has been found to be noninferior to ranibizumab [82]. Intravitreal injection of ranibizumab every four weeks, bevacizumab (off label) every four weeks, or aflibercept every eight weeks have similar efficacy [87]. Since bevacizumab is approved for treatment of patients with metastatic colon cancer, its ophthalmic use is off label and it is significantly cheaper than ranibizumab or aflibercept [88,89]. Faricimab (Vabysmo) is a bispecific anti-VEGF and anti-ANG2 molecule found to be noninferior in efficacy to aflibercept [90].

Another anti-VEGF agent to obtain US Food and Drug Administration (FDA) approval is a small single-chain antibody fragment called brolucizumab (Beovu), which was found not to be noninferior in efficacy to aflibercept, with up to one-third of the patients being able to be dosed at intervals of up to 12 weeks. Unfortunately, brolucizumab was found to cause in 2 percent of the people serious occlusive vasculitis and, thus, its adoption in real-world practice has been minimal [91-93].

Biosimilars — The cost of FDA-approved anti-VEGF medications for wet AMD is high (approximately $1000 to $2000 per injection in the United States). Biosimilar drugs promise to decrease cost, increase adherence, and potentially improve outcomes, especially in low-socioeconomic populations [94]. It is important to know that biosimilars are not automatically interchangeable with the original product. Interchangeable status can be granted by the FDA after additional evidence from studies in patients evaluating multiple switches between biosimilar and original product.

As of December 2022 the following biosimilars are approved in the United States for use in the eye:

Ranibizumab-nuna (Byooviz), a biosimilar product interchangeable with ranibizumab (Lucentis)

Ranibizumab-eqrn (Cimerli), a biosimilar product interchangeable with ranibizumab (Lucentis)

The naming of biosimilars is governed by distinct and differing process in the European Union and United States. In Europe, the unique brand name of a biosimilar is paired with the standard international nonproprietary name (eg, ranibizumab [Byooviz]). In the United States, a meaningless, random distinguishing four-letter lowercase suffix by hyphen is added to the standard international nonproprietary name (eg, ranibizumab-nuna [Byooviz]).

Risks of anti-VEGF injections — Most risks of anti-VEGF therapy are transient and related to the injections itself. Serious risks include infection and retinal detachment. Risks per injection should be interpreted in light of the fact that patients receive multiple injections over long periods of time.

Systemic exposure to anti-VEGF is limited due to its local administration, although there is some concern of increased risk of vascular events. There appears to be a slight increased risk of stroke with ranibizumab, which is likely true with other agents as well. Further studies are needed, but clinicians should be aware of this potential risk when using VEGF inhibitors in patients who are at increased risk for hemorrhagic stroke or other serious bleeding or thrombotic events.

Significant risks of anti-VEGF injections:

Endophthalmitis (1/2000 to 1/10,000) [95-98]

Retinal detachment (1/2000 to 1/10,000) [99,100]

Cerebrovascular events (1 to 2 percent versus 1 percent in the control) [85,101]

Safety in patients taking anticoagulants or antiplatelet drugs — The bleeding risk of intravitreal injections in patients taking anticoagulants is minimal, and anticoagulation should not be discontinued prior to injection [102,103].

Treatment schedules for VEGF inhibitors — The optimal frequency for injections of VEGF inhibitors is not known. After initial treatment and disease stabilization, there are strategies designed to limit subsequent treatments.

One common approach is discontinuous treatment, with injections given on an as-needed basis, guided by results of optical coherence tomography (OCT). This approach has been evaluated with ranibizumab and bevacizumab and found to result in fewer injections and equivalent visual outcomes compared with monthly injections [104-108]. However, patients treated in this fashion should have indefinite close follow-up so as not to miss disease reoccurrence [109].

Another dosing approach is a "treat-and-extend" regimen in which, after initial treatment and stabilization, the intervals for patient follow-up and treatment are adjusted based on clinical findings. Intervals are extended if neovascular activity is not present and shortened if either fluid or hemorrhage is seen; anti-VEGF treatment is administered at every visit, regardless of disease activity, so there is no prolonged lapse in treatment. This approach can decrease the number of injections and clinic visits while maintaining comparable visual improvement compared with fixed-interval dosing [110-113].

Ranibizumab and bevacizumab are usually given monthly for three to six months and then on an as-needed basis after assessment of disease activity or on "treat and extend" [114]. The on-label schedule for aflibercept is three doses (2 mg) at four-week intervals, followed by 2 mg every eight weeks [115]. However, most physicians use the treat-and-extend paradigm or on an as-needed basis. Similar approaches are used for faricimab and brolucizumab.

Photodynamic therapy — The role for photodynamic therapy (PDT) has decreased with the increasing use of anti-VEGF therapy. We now rarely use PDT (with or without intravitreal anti-VEGF) only for unique situations involving wet AMD [116,117] or central serous chorioretinopathy [118-120].

PDT utilizes the photosensitizing dye verteporfin (Visudyne), and laser activation [121-125]. Treatment involves intravenous injection of verteporfin, which accumulates in the abnormal blood vessels of wet AMD and, when stimulated by a low-power nonthermal red laser (689 nm) in the presence of oxygen, produces highly reactive, short-lived singlet oxygen and other reactive oxygen radicals, resulting in local damage to the endothelium and blockage of the vessels. Not all patients with wet AMD are eligible for PDT therapy (mostly patients with a subtype called "classic wet AMD" are eligible). Furthermore, treatment with PDT usually does not lead to vision gains but it does reduce the rate of vision decline in wet AMD. Dosing can be repeated for up to four times a year. As it is a photosensitizer, patients must avoid sun exposure for two to three days after the injection. Rare adverse reactions include severe vision loss from occlusion of normal vessels and reversible idiopathic low back pain.

Experimental and surgical treatments — Experimental submacular surgery with translocation of the retina [126] as well as radiation therapy for wet AMD [127,128] have also been tried, and some patients who have been recipients of these older approaches are still alive and may display late sequelae related to these interventions. Although these interventions have been mostly abandoned, submacular surgery for evacuation for large hemorrhages from wet AMD with or without tissue plasminogen activator and/or air are performed occasionally [129,130]. The optimal management of large, central submacular hemorrhages due to wet AMD is unknown, and outcomes remain mostly poor [130].

Geographic atrophy — Geographic atrophy is a form of advanced dry AMD, affecting 15 percent of AMD patients, characterized of loss of photoreceptors, retinal pigment epithelium, and choriocapillaris.

In 2023, the FDA approved the first two drugs aimed at this disease (intravitreal injection of pegcetacoplan and intravitreal injection of avacincaptad pegol) based on surrogate endpoint trial results that demonstrated a calculated reduction in lesion size [131-134]. However, approval of these drug has been controversial due to lack of demonstration of functional benefit and significant side effects [135]. Early after pegcetacoplan approval, reports of rare but serious vasculitis events with significant vision loss were reported by the American Society of Retina Specialists [136]. Due to lack of demonstration of functional benefit to any subgroup of patients, and the associated risks of vision loss, we do not endorse the use of these injections.

PREVENTION AND MONITORING — Adherence to a healthy lifestyle by quitting smoking and adopting a diet rich in plants, vegetables, and fish (eg, Mediterranean) is the best way to reduce risk of developing and progression of AMD. Regular annual visits to an eye doctor after the age of 55 may identify early AMD and become an opportunity for earlier counseling regarding the modifiable risk factors.

Amsler grid — Once AMD is detected, self-monitoring at home with an Amsler grid weekly is one of the best ways to detect, at the earliest phase, conversion from dry to wet AMD (figure 1).

Instructions for how to use an Amsler grid are as follows:

The chart should measure 10 cm × 10 cm in size (approximately 4 × 4 inches), should be well illuminated, and is used to measure central 20° visual field when kept at a distance of 33 cm (13 inches) from the eye.

Testing should be done with glasses if the patients need them.

In a well-illuminated room, ask the patient to hold the grid approximately 13 inches away from the face and test one eye at a time. (Ask to cover one eye.)

While looking directly at the center dot, the patient should observe the grid without redirecting the focus from the central point. Determine whether the patient can see all corners and sides of the grid. If any lines or areas look blurry, wavy, dark, or blank, mark that area in a chart note and schedule an urgent eye consult.

Home monitoring technology is improving and is becoming more common as both a way to detect the disease earlier and to manage the disease in a personalized fashion [137-145].

Retina specialists tend to monitor patients with dry AMD at least once a year. This periodic monitoring serves the dual purpose of identifying patients who progress from dry to wet AMD without having symptoms and to also educate the patient about the disease.

VISUAL AIDS — Being diagnosed with AMD can lead to significant central vision loss for approximately 15 to 30 percent of patients and can affect many more to a lesser degree. Having AMD and impaired vision does not need to mean losing control of one's life [146,147]. There is plenty of support available from several sources and professional services as well as a large array of equipment called "low-vision aids" including magnifiers [148], products that use color or contrast to make things easier to see and use, as well as merchandise designed for people with impaired vision. Many items such as reading stands, antiglare spectacles, and task lights can be found useful by individuals, though solid evidence is lacking for many of them [146].

Most electronic devices have special operating system modes made for the visually impaired, and these can have a significant impact on the quality of life of patients [149-153].

Low-vision devices for patients with AMD affecting their vision include:

Reading handheld optical magnifiers.

Portable electronic magnifiers.

High-power reading glasses (affects reading distance; patient needs to be very close to the book).

Video magnifiers (higher cost than handheld optical magnifiers).

Telemicroscopic glasses (restricted visual field).

Increase in contrast (increase lighting or darker print on brighter background). Certain computer monitors and electronic devices have special settings to accommodate need for higher contrast. White/black reverse modes for monitors.

Increase lighting (especially coming from behind the patient to decrease glare such as with goose neck lamps).

Large-print books.

Useful sites providing more information on visual aids and rehabilitation services available:

Macular Society; low-vision aids

National Library Service for the Blind and Print Disabled, Library of Congress

American Foundation for the Blind; resources for adults new to vision loss

American Academy of Ophthalmology; links and resources for people with low vision

VisionAware; locate vision rehabilitation services

United States Department of Veterans Affairs (844-698-2311); veterans can receive services and devices free of charge

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: Age-related macular degeneration (The Basics)" and "Patient education: Age-related vision loss (The Basics)")

SUMMARY AND RECOMMENDATIONS

Definition and classification – Age-related macular degeneration (AMD) is a degenerative disease of the photoreceptors of the central portion of the retina (the macula) and the supporting retinal pigment epithelium. AMD is classified as either dry or wet. The more common form is dry AMD (also known as nonexudative or nonneovascular AMD). The less common form is wet AMD (also known as exudative or neovascular AMD). (See 'Definition and classification' above.)

Epidemiology and risk factors – AMD is the most common cause of severe central vision loss and legal blindness among adults in the Western world. Risk factors include older age, genetic risk, smoking, and low dietary intake of antioxidants. (See 'Epidemiology' above and 'Risk factors' above.)

Clinical presentation – AMD may be asymptomatic in early stages. Patients tend to present with problems in contrast sensitivity and adaptation to dark environments. As the disease progresses, central visual acuity declines. (See 'Clinical presentation' above.)

Evaluation and diagnosis – Primary care evaluation includes a history and evaluation of visual acuity. Diagnosis requires ophthalmologic evaluation and is based on the presence of characteristic findings on dilated eye examination using a slit-lamp instrument (biomicroscopy). (See 'Ophthalmologic evaluation and diagnosis' above.)

Advice for all patients – We advise all patients quit smoking or avoid initiating smoking. We suggest that patients adhere to a Mediterranean diet (Grade 2C). (See 'Advice for all patients' above.)

Antioxidant vitamins – For patients with intermediate dry AMD in both eyes or severe AMD in one eye, we suggest daily vitamin supplements consistent with the Age-Related Eye Disease Study 2 (AREDS2) formula (Grade 2C).

However, it is also reasonable not to use these supplements based on demonstration of only modest benefit in these patients. Further, once disease has progressed beyond the stages described above, the supplements can be stopped due to lack of benefit.

The AREDS2 formulation contains a daily dose of vitamin C 500 mg, vitamin E 400 international units, lutein 10 mg, zeaxanthin 2 mg, zinc 80 mg (as zinc oxide), and copper 2 mg (as cupric oxide). (See 'Limited role for antioxidant vitamins' above.)

Treatment for wet AMD – For patients with AMD and neovascularization, we recommend as first-line treatment an intravitreal vascular endothelial growth factor (VEGF) inhibitor (eg, bevacizumab, ranibizumab, aflibercept) (Grade 1B). (See 'VEGF inhibitors' above.)

Monitoring – Once AMD is detected, self-monitoring with an Amsler grid weekly is one of the best ways to detect, at the earliest phase, conversion to wet AMD. (See 'Amsler grid' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Jorge Arroyo, MD, MPH, who contributed to earlier versions of this topic review.

  1. Miller JW. Beyond VEGF-The Weisenfeld Lecture. Invest Ophthalmol Vis Sci 2016; 57:6911.
  2. Miller JW. Developing Therapies for Age-related Macular Degeneration: The Art and Science of Problem-solving: The 2018 Charles L. Schepens, MD, Lecture. Ophthalmol Retina 2019; 3:900.
  3. Miller JW, Bagheri S, Vavvas DG. Advances in Age-related Macular Degeneration Understanding and Therapy. US Ophthalmic Rev 2017; 10:119.
  4. Curcio CA. Soft Drusen in Age-Related Macular Degeneration: Biology and Targeting Via the Oil Spill Strategies. Invest Ophthalmol Vis Sci 2018; 59:AMD160.
  5. Apte RS. Age-Related Macular Degeneration. N Engl J Med 2021; 385:539.
  6. Chakravarthy U, Bailey CC, Scanlon PH, et al. Progression from Early/Intermediate to Advanced Forms of Age-Related Macular Degeneration in a Large UK Cohort: Rates and Risk Factors. Ophthalmol Retina 2020; 4:662.
  7. Age-Related Eye Disease Study Research Group. A randomized, placebo-controlled, clinical trial of high-dose supplementation with vitamins C and E, beta carotene, and zinc for age-related macular degeneration and vision loss: AREDS report no. 8. Arch Ophthalmol 2001; 119:1417.
  8. Ferris FL, Davis MD, Clemons TE, et al. A simplified severity scale for age-related macular degeneration: AREDS Report No. 18. Arch Ophthalmol 2005; 123:1570.
  9. Liew G, Joachim N, Mitchell P, et al. Validating the AREDS Simplified Severity Scale of Age-Related Macular Degeneration with 5- and 10-Year Incident Data in a Population-Based Sample. Ophthalmology 2016; 123:1874.
  10. Ferris FL 3rd, Wilkinson CP, Bird A, et al. Clinical classification of age-related macular degeneration. Ophthalmology 2013; 120:844.
  11. Koh AH, Ang CL. Age-related macular degeneration: what's new. Ann Acad Med Singap 2002; 31:399.
  12. Wong WL, Su X, Li X, et al. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: a systematic review and meta-analysis. Lancet Glob Health 2014; 2:e106.
  13. Rein DB, Wittenborn JS, Burke-Conte Z, et al. Prevalence of Age-Related Macular Degeneration in the US in 2019. JAMA Ophthalmol 2022; 140:1202.
  14. Klein R, Klein BE, Cruickshanks KJ. The prevalence of age-related maculopathy by geographic region and ethnicity. Prog Retin Eye Res 1999; 18:371.
  15. Kawasaki R, Yasuda M, Song SJ, et al. The prevalence of age-related macular degeneration in Asians: a systematic review and meta-analysis. Ophthalmology 2010; 117:921.
  16. Friedman DS, O'Colmain BJ, Muñoz B, et al. Prevalence of age-related macular degeneration in the United States. Arch Ophthalmol 2004; 122:564.
  17. Klein R, Klein BE, Linton KL. Prevalence of age-related maculopathy. The Beaver Dam Eye Study. Ophthalmology 1992; 99:933.
  18. Colijn JM, Buitendijk GHS, Prokofyeva E, et al. Prevalence of Age-Related Macular Degeneration in Europe: The Past and the Future. Ophthalmology 2017; 124:1753.
  19. Fritsche LG, Igl W, Bailey JN, et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat Genet 2016; 48:134.
  20. Strunz T, Lauwen S, Kiel C, et al. A transcriptome-wide association study based on 27 tissues identifies 106 genes potentially relevant for disease pathology in age-related macular degeneration. Sci Rep 2020; 10:1584.
  21. Xue Z, Yuan J, Chen F, et al. Genome-wide association meta-analysis of 88,250 individuals highlights pleiotropic mechanisms of five ocular diseases in UK Biobank. EBioMedicine 2022; 82:104161.
  22. Huan T, Cheng SY, Tian B, et al. Identifying Novel Genes and Variants in Immune and Coagulation Pathways Associated with Macular Degeneration. Ophthalmol Sci 2023; 3:100206.
  23. van Asten F, Simmons M, Singhal A, et al. A Deep Phenotype Association Study Reveals Specific Phenotype Associations with Genetic Variants in Age-related Macular Degeneration: Age-Related Eye Disease Study 2 (AREDS2) Report No. 14. Ophthalmology 2018; 125:559.
  24. Lee KS, Lin S, Copland DA, et al. Cellular senescence in the aging retina and developments of senotherapies for age-related macular degeneration. J Neuroinflammation 2021; 18:32.
  25. Blasiak J. Senescence in the pathogenesis of age-related macular degeneration. Cell Mol Life Sci 2020; 77:789.
  26. Blasiak J, Pawlowska E, Sobczuk A, et al. The Aging Stress Response and Its Implication for AMD Pathogenesis. Int J Mol Sci 2020; 21.
  27. Despriet DD, Klaver CC, Witteman JC, et al. Complement factor H polymorphism, complement activators, and risk of age-related macular degeneration. JAMA 2006; 296:301.
  28. Hageman GS, Anderson DH, Johnson LV, et al. A common haplotype in the complement regulatory gene factor H (HF1/CFH) predisposes individuals to age-related macular degeneration. Proc Natl Acad Sci U S A 2005; 102:7227.
  29. Tzoumas N, Hallam D, Harris CL, et al. Revisiting the role of factor H in age-related macular degeneration: Insights from complement-mediated renal disease and rare genetic variants. Surv Ophthalmol 2021; 66:378.
  30. Klein RJ, Zeiss C, Chew EY, et al. Complement factor H polymorphism in age-related macular degeneration. Science 2005; 308:385.
  31. Whitmore SS, Sohn EH, Chirco KR, et al. Complement activation and choriocapillaris loss in early AMD: implications for pathophysiology and therapy. Prog Retin Eye Res 2015; 45:1.
  32. Vavvas DG, Daniels AB, Kapsala ZG, et al. Regression of Some High-risk Features of Age-related Macular Degeneration (AMD) in Patients Receiving Intensive Statin Treatment. EBioMedicine 2016; 5:198.
  33. Notomi S, Ishihara K, Efstathiou NE, et al. Genetic LAMP2 deficiency accelerates the age-associated formation of basal laminar deposits in the retina. Proc Natl Acad Sci U S A 2019; 116:23724.
  34. Lee JJ, Ishihara K, Notomi S, et al. Lysosome-associated membrane protein-2 deficiency increases the risk of reactive oxygen species-induced ferroptosis in retinal pigment epithelial cells. Biochem Biophys Res Commun 2020; 521:414.
  35. Wang AL, Lukas TJ, Yuan M, et al. Autophagy, exosomes and drusen formation in age-related macular degeneration. Autophagy 2009; 5:563.
  36. Wang AL, Lukas TJ, Yuan M, et al. Autophagy and exosomes in the aged retinal pigment epithelium: possible relevance to drusen formation and age-related macular degeneration. PLoS One 2009; 4:e4160.
  37. Solberg Y, Rosner M, Belkin M. The association between cigarette smoking and ocular diseases. Surv Ophthalmol 1998; 42:535.
  38. Shen JK, Dong A, Hackett SF, et al. Oxidative damage in age-related macular degeneration. Histol Histopathol 2007; 22:1301.
  39. Penfold PL, Madigan MC, Gillies MC, Provis JM. Immunological and aetiological aspects of macular degeneration. Prog Retin Eye Res 2001; 20:385.
  40. Wang JC, Cao S, Wang A, et al. CFH Y402H polymorphism is associated with elevated vitreal GM-CSF and choroidal macrophages in the postmortem human eye. Mol Vis 2015; 21:264.
  41. Lad EM, Cousins SW, Van Arnam JS, Proia AD. Abundance of infiltrating CD163+ cells in the retina of postmortem eyes with dry and neovascular age-related macular degeneration. Graefes Arch Clin Exp Ophthalmol 2015; 253:1941.
  42. He X, Hahn P, Iacovelli J, et al. Iron homeostasis and toxicity in retinal degeneration. Prog Retin Eye Res 2007; 26:649.
  43. Ding JD, Johnson LV, Herrmann R, et al. Anti-amyloid therapy protects against retinal pigmented epithelium damage and vision loss in a model of age-related macular degeneration. Proc Natl Acad Sci U S A 2011; 108:E279.
  44. Isas JM, Luibl V, Johnson LV, et al. Soluble and mature amyloid fibrils in drusen deposits. Invest Ophthalmol Vis Sci 2010; 51:1304.
  45. Luibl V, Isas JM, Kayed R, et al. Drusen deposits associated with aging and age-related macular degeneration contain nonfibrillar amyloid oligomers. J Clin Invest 2006; 116:378.
  46. Johnson LV, Leitner WP, Rivest AJ, et al. The Alzheimer's A beta -peptide is deposited at sites of complement activation in pathologic deposits associated with aging and age-related macular degeneration. Proc Natl Acad Sci U S A 2002; 99:11830.
  47. Chapman NA, Jacobs RJ, Braakhuis AJ. Role of diet and food intake in age-related macular degeneration: a systematic review. Clin Exp Ophthalmol 2019; 47:106.
  48. Mitchell P, Liew G, Gopinath B, Wong TY. Age-related macular degeneration. Lancet 2018; 392:1147.
  49. Evans JR, Fletcher AE, Wormald RP. 28,000 Cases of age related macular degeneration causing visual loss in people aged 75 years and above in the United Kingdom may be attributable to smoking. Br J Ophthalmol 2005; 89:550.
  50. Gastaldello A, Giampieri F, Quiles JL, et al. Adherence to the Mediterranean-Style Eating Pattern and Macular Degeneration: A Systematic Review of Observational Studies. Nutrients 2022; 14.
  51. Agrón E, Mares J, Clemons TE, et al. Dietary Nutrient Intake and Progression to Late Age-Related Macular Degeneration in the Age-Related Eye Disease Studies 1 and 2. Ophthalmology 2021; 128:425.
  52. Keenan TD, Agrón E, Mares J, et al. Adherence to the Mediterranean Diet and Progression to Late Age-Related Macular Degeneration in the Age-Related Eye Disease Studies 1 and 2. Ophthalmology 2020; 127:1515.
  53. Merle BMJ, Colijn JM, Cougnard-Grégoire A, et al. Mediterranean Diet and Incidence of Advanced Age-Related Macular Degeneration: The EYE-RISK Consortium. Ophthalmology 2019; 126:381.
  54. Zhu W, Wu Y, Meng YF, et al. Fish Consumption and Age-Related Macular Degeneration Incidence: A Meta-Analysis and Systematic Review of Prospective Cohort Studies. Nutrients 2016; 8.
  55. Hogg RE, Woodside JV, McGrath A, et al. Mediterranean Diet Score and Its Association with Age-Related Macular Degeneration: The European Eye Study. Ophthalmology 2017; 124:82.
  56. Nunes S, Alves D, Barreto P, et al. Adherence to a Mediterranean diet and its association with age-related macular degeneration. The Coimbra Eye Study-Report 4. Nutrition 2018; 51-52:6.
  57. Pameijer EM, Heus P, Damen JAA, et al. What did we learn in 35 years of research on nutrition and supplements for age-related macular degeneration: a systematic review. Acta Ophthalmol 2022; 100:e1541.
  58. Kuan V, Warwick A, Hingorani A, et al. Association of Smoking, Alcohol Consumption, Blood Pressure, Body Mass Index, and Glycemic Risk Factors With Age-Related Macular Degeneration: A Mendelian Randomization Study. JAMA Ophthalmol 2021; 139:1299.
  59. Cougnard-Grégoire A, Delyfer MN, Korobelnik JF, et al. Long-term blood pressure and age-related macular degeneration: the ALIENOR study. Invest Ophthalmol Vis Sci 2013; 54:1905.
  60. Pennington KL, DeAngelis MM. Epidemiology of age-related macular degeneration (AMD): associations with cardiovascular disease phenotypes and lipid factors. Eye Vis (Lond) 2016; 3:34.
  61. Dinu M, Pagliai G, Casini A, Sofi F. Food groups and risk of age-related macular degeneration: a systematic review with meta-analysis. Eur J Nutr 2019; 58:2123.
  62. Gaynes BI. AREDS misses on safety. Arch Ophthalmol 2003; 121:416.
  63. Seigel D. AREDS investigators distort findings. Arch Ophthalmol 2002; 120:100.
  64. Ambati J, Ambati BK. Age-related eye disease study caveats. Arch Ophthalmol 2002; 120:997; author reply 997.
  65. Evans JR, Lawrenson JG. Antioxidant vitamin and mineral supplements for slowing the progression of age-related macular degeneration. Cochrane Database Syst Rev 2023; 9:CD000254.
  66. Omenn GS, Goodman GE, Thornquist MD, et al. Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease. N Engl J Med 1996; 334:1150.
  67. Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N Engl J Med 1994; 330:1029.
  68. Tanvetyanon T, Bepler G. Beta-carotene in multivitamins and the possible risk of lung cancer among smokers versus former smokers: a meta-analysis and evaluation of national brands. Cancer 2008; 113:150.
  69. Cortés-Jofré M, Rueda JR, Asenjo-Lobos C, et al. Drugs for preventing lung cancer in healthy people. Cochrane Database Syst Rev 2020; 3:CD002141.
  70. Caraballoso M, Sacristan M, Serra C, Bonfill X. Drugs for preventing lung cancer in healthy people. Cochrane Database Syst Rev 2003; :CD002141.
  71. Yu N, Su X, Wang Z, et al. Association of Dietary Vitamin A and β-Carotene Intake with the Risk of Lung Cancer: A Meta-Analysis of 19 Publications. Nutrients 2015; 7:9309.
  72. Leitzmann MF, Stampfer MJ, Wu K, et al. Zinc supplement use and risk of prostate cancer. J Natl Cancer Inst 2003; 95:1004.
  73. Age-Related Eye Disease Study 2 Research Group. Lutein + zeaxanthin and omega-3 fatty acids for age-related macular degeneration: the Age-Related Eye Disease Study 2 (AREDS2) randomized clinical trial. JAMA 2013; 309:2005.
  74. Klein R, Klein BE, Jensen SC, Meuer SM. The five-year incidence and progression of age-related maculopathy: the Beaver Dam Eye Study. Ophthalmology 1997; 104:7.
  75. Klein R, Klein BE, Tomany SC, et al. Ten-year incidence and progression of age-related maculopathy: The Beaver Dam eye study. Ophthalmology 2002; 109:1767.
  76. Barbazetto IA, Saroj N, Shapiro H, et al. Incidence of new choroidal neovascularization in fellow eyes of patients treated in the MARINA and ANCHOR trials. Am J Ophthalmol 2010; 149:939.
  77. Solomon SD, Lindsley K, Vedula SS, et al. Anti-vascular endothelial growth factor for neovascular age-related macular degeneration. Cochrane Database Syst Rev 2019; 3:CD005139.
  78. Lim JH, Wickremasinghe SS, Xie J, et al. Delay to treatment and visual outcomes in patients treated with anti-vascular endothelial growth factor for age-related macular degeneration. Am J Ophthalmol 2012; 153:678.
  79. Rofagha S, Bhisitkul RB, Boyer DS, et al. Seven-year outcomes in ranibizumab-treated patients in ANCHOR, MARINA, and HORIZON: a multicenter cohort study (SEVEN-UP). Ophthalmology 2013; 120:2292.
  80. Moja L, Lucenteforte E, Kwag KH, et al. Systemic safety of bevacizumab versus ranibizumab for neovascular age-related macular degeneration. Cochrane Database Syst Rev 2014; :CD011230.
  81. Rosenfeld PJ, Brown DM, Heier JS, et al. Ranibizumab for neovascular age-related macular degeneration. N Engl J Med 2006; 355:1419.
  82. Heier JS, Brown DM, Chong V, et al. Intravitreal aflibercept (VEGF trap-eye) in wet age-related macular degeneration. Ophthalmology 2012; 119:2537.
  83. Chakravarthy U, Harding SP, Rogers CA, et al. Alternative treatments to inhibit VEGF in age-related choroidal neovascularisation: 2-year findings of the IVAN randomised controlled trial. Lancet 2013; 382:1258.
  84. Kodjikian L, Souied EH, Mimoun G, et al. Ranibizumab versus Bevacizumab for Neovascular Age-related Macular Degeneration: Results from the GEFAL Noninferiority Randomized Trial. Ophthalmology 2013; 120:2300.
  85. CATT Research Group, Martin DF, Maguire MG, et al. Ranibizumab and bevacizumab for neovascular age-related macular degeneration. N Engl J Med 2011; 364:1897.
  86. Comparison of Age-related Macular Degeneration Treatments Trials (CATT) Research Group, Martin DF, Maguire MG, et al. Ranibizumab and bevacizumab for treatment of neovascular age-related macular degeneration: two-year results. Ophthalmology 2012; 119:1388.
  87. VEGF inhibitors for AMD and diabetic macular edema. Med Lett Drugs Ther 2015; 57:41.
  88. Rich RM, Rosenfeld PJ, Puliafito CA, et al. Short-term safety and efficacy of intravitreal bevacizumab (Avastin) for neovascular age-related macular degeneration. Retina 2006; 26:495.
  89. Rosenfeld PJ. Intravitreal avastin: the low cost alternative to lucentis? Am J Ophthalmol 2006; 142:141.
  90. Heier JS, Khanani AM, Quezada Ruiz C, et al. Efficacy, durability, and safety of intravitreal faricimab up to every 16 weeks for neovascular age-related macular degeneration (TENAYA and LUCERNE): two randomised, double-masked, phase 3, non-inferiority trials. Lancet 2022; 399:729.
  91. Witkin AJ, Hahn P, Murray TG, et al. Occlusive Retinal Vasculitis Following Intravitreal Brolucizumab. J Vitreoretin Dis 2020; 4:269.
  92. Haug SJ, Hien DL, Uludag G, et al. Retinal arterial occlusive vasculitis following intravitreal brolucizumab administration. Am J Ophthalmol Case Rep 2020; 18:100680.
  93. Monés J, Srivastava SK, Jaffe GJ, et al. Risk of Inflammation, Retinal Vasculitis, and Retinal Occlusion-Related Events with Brolucizumab: Post Hoc Review of HAWK and HARRIER. Ophthalmology 2021; 128:1050.
  94. Polat O, İnan S, Özcan S, et al. Factors Affecting Compliance to Intravitreal Anti-Vascular Endothelial Growth Factor Therapy in Patients with Age-Related Macular Degeneration. Turk J Ophthalmol 2017; 47:205.
  95. Chen Y, Wei W, Vavvas DG, et al. Incidence of Endophthalmitis after Intravitreal Anti-Vascular Endothelial Growth Factor Injections in an Operating Room in China. J Ophthalmol 2020; 2020:5163484.
  96. Dossarps D, Bron AM, Koehrer P, et al. Endophthalmitis After Intravitreal Injections: Incidence, Presentation, Management, and Visual Outcome. Am J Ophthalmol 2015; 160:17.
  97. Gregori NZ, Flynn HW Jr, Schwartz SG, et al. Current Infectious Endophthalmitis Rates After Intravitreal Injections of Anti-Vascular Endothelial Growth Factor Agents and Outcomes of Treatment. Ophthalmic Surg Lasers Imaging Retina 2015; 46:643.
  98. Rayess N, Rahimy E, Storey P, et al. Postinjection Endophthalmitis Rates and Characteristics Following Intravitreal Bevacizumab, Ranibizumab, and Aflibercept. Am J Ophthalmol 2016; 165:88.
  99. Storey PP, Pancholy M, Wibbelsman TD, et al. Rhegmatogenous Retinal Detachment after Intravitreal Injection of Anti-Vascular Endothelial Growth Factor. Ophthalmology 2019; 126:1424.
  100. Mammo DA, Ringeisen AL, Parke DW 3rd. Frequency of Rhegmatogenous Retinal Detachment after Intravitreal Therapy in Neovascular Age-Related Macular Degeneration. Ophthalmol Retina 2020; 4:973.
  101. Brown DM, Kaiser PK, Michels M, et al. Ranibizumab versus verteporfin for neovascular age-related macular degeneration. N Engl J Med 2006; 355:1432.
  102. Charles S, Rosenfeld PJ, Gayer S. Medical consequences of stopping anticoagulant therapy before intraocular surgery or intravitreal injections. Retina 2007; 27:813.
  103. Mason JO 3rd, Frederick PA, Neimkin MG, et al. Incidence of hemorrhagic complications after intravitreal bevacizumab (avastin) or ranibizumab (lucentis) injections on systemically anticoagulated patients. Retina 2010; 30:1386.
  104. Brown DM, Regillo CD. Anti-VEGF agents in the treatment of neovascular age-related macular degeneration: applying clinical trial results to the treatment of everyday patients. Am J Ophthalmol 2007; 144:627.
  105. Lalwani GA, Rosenfeld PJ, Fung AE, et al. A variable-dosing regimen with intravitreal ranibizumab for neovascular age-related macular degeneration: year 2 of the PrONTO Study. Am J Ophthalmol 2009; 148:43.
  106. Bashshur ZF, Haddad ZA, Schakal AR, et al. Intravitreal bevacizumab for treatment of neovascular age-related macular degeneration: the second year of a prospective study. Am J Ophthalmol 2009; 148:59.
  107. El-Mollayess GM, Mahfoud Z, Schakal AR, et al. Fixed-interval versus OCT-guided variable dosing of intravitreal bevacizumab in the management of neovascular age-related macular degeneration: a 12-month randomized prospective study. Am J Ophthalmol 2012; 153:481.
  108. Boyer DS, Nguyen QD, Brown DM, et al. Outcomes with As-Needed Ranibizumab after Initial Monthly Therapy: Long-Term Outcomes of the Phase III RIDE and RISE Trials. Ophthalmology 2015; 122:2504.
  109. Rasmussen A, Bloch SB, Fuchs J, et al. A 4-year longitudinal study of 555 patients treated with ranibizumab for neovascular age-related macular degeneration. Ophthalmology 2013; 120:2630.
  110. Rayess N, Houston SK 3rd, Gupta OP, et al. Treatment outcomes after 3 years in neovascular age-related macular degeneration using a treat-and-extend regimen. Am J Ophthalmol 2015; 159:3.
  111. Arnold JJ, Campain A, Barthelmes D, et al. Two-year outcomes of "treat and extend" intravitreal therapy for neovascular age-related macular degeneration. Ophthalmology 2015; 122:1212.
  112. Wykoff CC, Croft DE, Brown DM, et al. Prospective Trial of Treat-and-Extend versus Monthly Dosing for Neovascular Age-Related Macular Degeneration: TREX-AMD 1-Year Results. Ophthalmology 2015; 122:2514.
  113. Silva R, Berta A, Larsen M, et al. Treat-and-Extend versus Monthly Regimen in Neovascular Age-Related Macular Degeneration: Results with Ranibizumab from the TREND Study. Ophthalmology 2018; 125:57.
  114. Skelly A, Bezlyak V, Liew G, et al. Treat and Extend Treatment Interval Patterns with Anti-VEGF Therapy in nAMD Patients. Vision (Basel) 2019; 3.
  115. Aflibercept (eylea) for age-related macular degeneration. Med Lett Drugs Ther 2012; 54:9.
  116. Koh AH, Expert PCV Panel, Chen LJ, et al. Polypoidal choroidal vasculopathy: evidence-based guidelines for clinical diagnosis and treatment. Retina 2013; 33:686.
  117. Koh A, Lee WK, Chen LJ, et al. EVEREST study: efficacy and safety of verteporfin photodynamic therapy in combination with ranibizumab or alone versus ranibizumab monotherapy in patients with symptomatic macular polypoidal choroidal vasculopathy. Retina 2012; 32:1453.
  118. van Rijssen TJ, van Dijk EHC, Tsonaka R, et al. Half-Dose Photodynamic Therapy Versus Eplerenone in Chronic Central Serous Chorioretinopathy (SPECTRA): A Randomized Controlled Trial. Am J Ophthalmol 2022; 233:101.
  119. Chan WM, Lai TY, Lai RY, et al. Half-dose verteporfin photodynamic therapy for acute central serous chorioretinopathy: one-year results of a randomized controlled trial. Ophthalmology 2008; 115:1756.
  120. Yannuzzi LA, Slakter JS, Gross NE, et al. Indocyanine green angiography-guided photodynamic therapy for treatment of chronic central serous chorioretinopathy: a pilot study. Retina 2003; 23:288.
  121. Miller JW, Walsh AW, Kramer M, et al. Photodynamic therapy of experimental choroidal neovascularization using lipoprotein-delivered benzoporphyrin. Arch Ophthalmol 1995; 113:810.
  122. Miller JW, Schmidt-Erfurth U, Sickenberg M, et al. Photodynamic therapy with verteporfin for choroidal neovascularization caused by age-related macular degeneration: results of a single treatment in a phase 1 and 2 study. Arch Ophthalmol 1999; 117:1161.
  123. Blumenkranz MS, Bressler NM, Bressler SB, et al. Verteporfin therapy for subfoveal choroidal neovascularization in age-related macular degeneration: three-year results of an open-label extension of 2 randomized clinical trials--TAP Report no. 5. Arch Ophthalmol 2002; 120:1307.
  124. Blinder KJ, Bradley S, Bressler NM, et al. Effect of lesion size, visual acuity, and lesion composition on visual acuity change with and without verteporfin therapy for choroidal neovascularization secondary to age-related macular degeneration: TAP and VIP report no. 1. Am J Ophthalmol 2003; 136:407.
  125. Miller JW. Higher irradiance and photodynamic therapy for age-related macular degeneration (an AOS thesis). Trans Am Ophthalmol Soc 2008; 106:357.
  126. Falkner CI, Leitich H, Frommlet F, et al. The end of submacular surgery for age-related macular degeneration? A meta-analysis. Graefes Arch Clin Exp Ophthalmol 2007; 245:490.
  127. Chen L, Kim IK, Lane AM, et al. Proton beam irradiation for non-AMD CNV: 2-year results of a randomised clinical trial. Br J Ophthalmol 2014; 98:1212.
  128. Zambarakji HJ, Lane AM, Ezra E, et al. Proton beam irradiation for neovascular age-related macular degeneration. Ophthalmology 2006; 113:2012.
  129. Grohmann C, Dimopoulos S, Bartz-Schmidt KU, et al. Surgical management of submacular hemorrhage due to n-AMD: a comparison of three surgical methods. Int J Retina Vitreous 2020; 6:27.
  130. Tranos P, Tsiropoulos GN, Koronis S, et al. Comparison of subretinal versus intravitreal injection of recombinant tissue plasminogen activator with gas for submacular hemorrhage secondary to wet age-related macular degeneration: treatment outcomes and brief literature review. Int Ophthalmol 2021; 41:4037.
  131. SYFOVRE (pegcetacoplan injection), for intravitreal use. United States Prescribing Information. US Food and Drug Administration. Available at: https://pi.apellis.com/files/PI_SYFOVRE.pdf. (Accessed on May 16, 2023).
  132. IZERVAY™ (avacincaptad pegol intravitreal solution) prescribing information. Available at: https://ivericbio.com/wp-content/uploads/IZERVAY-avacincaptad-pegol-intravitreal-solution-PI_Final_8.4.23.pdf (Accessed on September 01, 2023).
  133. Khanani AM, Patel SS, Staurenghi G, et al. Efficacy and safety of avacincaptad pegol in patients with geographic atrophy (GATHER2): 12-month results from a randomised, double-masked, phase 3 trial. Lancet 2023; 402:1449.
  134. Heier JS, Lad EM, Holz FG, et al. Pegcetacoplan for the treatment of geographic atrophy secondary to age-related macular degeneration (OAKS and DERBY): two multicentre, randomised, double-masked, sham-controlled, phase 3 trials. Lancet 2023; 402:1434.
  135. Spaide RF, Vavvas DG. COMPLEMENT INHIBITION FOR GEOGRAPHIC ATROPHY: Review of Salient Functional Outcomes and Perspective. Retina 2023; 43:1064.
  136. American Society of Retina Specialists (ASRS) Research and Safety in Therapeutics (ReST) committee update on intraocular inflammation (IOI). Available at: https://www.asrs.org/content/documents/asrs-rest-committee-update-on-intraocular-inflammation-after-ivi-2023.pdf (Accessed on September 01, 2023).
  137. Fawzi A. Re: Bontzos et al.: Nonresponders to Ranibizumab Anti-VEGF Treatment Are Actually Short-term Responders: A Prospective Spectral-Domain OCT Study (Ophthalmol Retina. 2020;4:1138-1145). Ophthalmol Retina 2021; 5:e3.
  138. Bontzos G, Bagheri S, Ioanidi L, et al. Nonresponders to Ranibizumab Anti-VEGF Treatment Are Actually Short-term Responders: A Prospective Spectral-Domain OCT Study. Ophthalmol Retina 2020; 4:1138.
  139. Bontzos GC, Bagheri S, Ioanidi LD, et al. Reply. Ophthalmol Retina 2021; 5:e4.
  140. Yu HJ, Kiernan DF, Eichenbaum D, et al. Home Monitoring of Age-Related Macular Degeneration: Utility of the ForeseeHome Device for Detection of Neovascularization. Ophthalmol Retina 2021; 5:348.
  141. Busquets MA, Sabbagh O. Current status of home monitoring technology for age-related macular degeneration. Curr Opin Ophthalmol 2021; 32:240.
  142. Armstrong GW, Miller JB. Telemedicine for the Diagnosis and Management of Age-Related Macular Degeneration: A Review. J Clin Med 2022; 11.
  143. Faes L, Golla K, Islam M, et al. System usability, user satisfaction and long-term adherence to mobile hyperacuity home monitoring-prospective follow-up study. Eye (Lond) 2023; 37:650.
  144. Kim JE, Tomkins-Netzer O, Elman MJ, et al. Evaluation of a self-imaging SD-OCT system designed for remote home monitoring. BMC Ophthalmol 2022; 22:261.
  145. Chen E, Mills M, Gallagher T, et al. Remote patient monitoring of central retinal function with MACUSTAT®: A multi-modal macular function scan. Digit Health 2022; 8:20552076221132105.
  146. Virgili G, Acosta R, Bentley SA, et al. Reading aids for adults with low vision. Cochrane Database Syst Rev 2018; 4:CD003303.
  147. Christoforidis JB, Tecce N, Dell'Omo R, et al. Age related macular degeneration and visual disability. Curr Drug Targets 2011; 12:221.
  148. Virtanen P, Laatikainen L. Low-vision aids in age-related macular degeneration. Curr Opin Ophthalmol 1993; 4:33.
  149. Suo L, Ke X, Zhang D, et al. Use of Mobile Apps for Visual Acuity Assessment: Systematic Review and Meta-analysis. JMIR Mhealth Uhealth 2022; 10:e26275.
  150. Wittich W, Jarry J, Morrice E, Johnson A. Effectiveness of the Apple iPad as a Spot-reading Magnifier. Optom Vis Sci 2018; 95:704.
  151. Haji SA, Sambhav K, Grover S, Chalam KV. Evaluation of the iPad as a low vision aid for improving reading ability. Clin Ophthalmol 2015; 9:17.
  152. Robinson JL, Braimah Avery V, Chun R, et al. Usage of Accessibility Options for the iPhone and iPad in a Visually Impaired Population. Semin Ophthalmol 2017; 32:163.
  153. Mednick Z, Jaidka A, Nesdole R, Bona M. Assessing the iPad as a tool for low-vision rehabilitation. Can J Ophthalmol 2017; 52:13.
Topic 6901 Version 65.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟