Skip to main content
Springer logoLink to Springer
. 2025 Jul 8;13(1):9. doi: 10.1007/s40135-025-00332-x

Systemic Carbonic Anhydrase Inhibitors in Common Ophthalmic Diseases: A Scoping Review from A Clinical Standpoint

Shahin Hallaj 1,2,, Wesam Shamseldin Shalaby 1,3, Sapna Sinha 1, Jonathan S Myers 1, Reza Razeghinejad 1
PMCID: PMC12238189  PMID: 40642047

Abstract

Purpose of Review

This paper provides a detailed overview of systemic carbonic anhydrase inhibitors (CAIs) and their application in managing ophthalmic conditions, including glaucoma, cystoid macular edema, and idiopathic intracranial hypertension. Despite their recognized clinical benefits, the potential for severe systemic adverse effects often discourages clinicians from prescribing these agents. By examining the pharmacology, pharmacokinetics, safety profile, and clinical indications of systemic CAIs, this review highlights strategies to mitigate treatment challenges, particularly in patients with comorbidities and complex medical backgrounds.

Recent Findings

New insights into the diverse roles and distribution of carbonic anhydrase (CA) isozymes have expanded our understanding of both the mechanisms and clinical implications of systemic CAIs. In the eye, CA-II and CA-IV are key contributors to aqueous humor production and ocular blood flow modulation, with dose-dependent improvements in choroidal perfusion observed upon enzyme inhibition. Beyond mechanism of action, tissue-bound drug concentration is a key factor in achieving maximal intraocular pressure (IOP) reduction. Moreover, some CAIs exhibit activity independent of CA blockade, such as inhibiting aquaporin-4. Clinical data further indicate that despite theoretical risks of systemic toxicity—particularly in individuals with comorbidities—severe adverse events are relatively rare.

Summary

In specific ocular diseases, the use of systemic CAIs is vital for maintaining vision. Although systemic CAIs carry risk of systemic adverse events, with higher risk, the incidence of severe adverse events is low and in some studies were comparable to that of topical therapy. It's important to be aware of potential side effects and ensure the correct usage of oral CAIs, especially in individuals with other systemic diseases. A tailored evaluation of risks and benefits carried out for each individual, particularly in case of prolonged usage, should decrease the risk of adverse events.

Keywords: Acetazolamide, Carbonic Anhydrase Inhibitors, Dichlorphenamide, Ethoxzolamide, Methazolamide

Introduction

Carbonic anhydrase enzyme (CA) plays a crucial role in regulating various physiological processes, including acid–base equilibrium, ion transport, pancreatic juice production, memory, muscular and neural function, and aqueous humor production [1, 2]. Its primary function is catalyzing the conversion of carbon dioxide and water into bicarbonate and hydrogen ions, thereby maintaining fluid balance and pH levels. CA I-IV are the most abundant among the 16 isozymes of the CA enzyme in the eye [3]. Carbonic anhydrase inhibitors (CAIs) were introduced in the mid-1900 s [4], and later became available in oral, intramuscular, intravenous, and topical forms. While widely recognized for their efficacy in lowering the intraocular pressure (IOP), are also used in certain types of the macular edema and idiopathic intracranial hypertension (IIH) [1]. Despite their significant efficacy, the use of systemic CAIs can be associated with severe side effects [5], posing challenges for clinicians in the decision-making process. This review covers the pharmacology, pharmacokinetics, indications, and side effects of systemic CAIs.

Materials and Methods

A comprehensive search of Medline and PubMed databases was performed on articles published between 1960 and 2023 using the following keywords and combinations: carbonic anhydrase inhibitors, acetazolamide, methazolamide, intraocular pressure, mechanism of action, carbonic anhydrase, glaucoma, retina, macular edema, retinitis pigmentosa, idiopathic intracranial hypertension, side effects, sickling disorder, sulfa allergy, trabecular meshwork, pharmacology, pharmacokinetics, metabolites, pregnancy, epilepsy, diabetes, anticoagulants, liver disease, and renal impairment. Reference lists of the included papers were used to further guide the literature review.

Carbonic Anhydrase Isozymes

Different CA isozymes present in the eye are shown in Table 1. Inhibition of CAs in the eye results in elevated tissue carbon dioxide pressure, extracellular acidification, and an intensified Bohr effect, leading to localized vasodilation. This may contribute to their dose-dependent effect on ocular blood flow [68]. It is believed that most of the ocular effects of CAIs are mediated through CA-II and -IV inhibition [9, 10].

Table 1.

Different isoenzymes of carbonic anhydrase in the eye

Site Expressed Carbonic Anhydrase Isozymes
Cornea Endothelium I, II, IV
Ciliary Body Pigmented epithelium I, II, III, IV
Capillaries I, II, IV
Lens Epithelium and fibers I, II, III, IV
Retina Müller cells II
Cones I, II
Pigment epithelium IV
Choroid Choriocapillaris I, II, IV
Ophthalmic Artery Orbit IV

The cytoplasmic CA-II isozyme is found in the retinal pigment epithelium (RPE), lens, corneal endothelium, iris, and ciliary processes. In the ciliary body, CA-II plays a vital role in aqueous humor production by producing bicarbonate and facilitating the active transport of sodium and chloride ions, establishing an osmotic gradient, and promoting water movement into the posterior chamber. The inhibition of this enzyme is achieved when more than 99.9% of it is bound to CAIs [11, 12]. CA-II is highly concentrated in the kidneys, and systemic CAIs use is commonly associated with a moderate degree of metabolic acidosis [13], which may be one of the mechanisms involved in mediating the ocular hypotensive effects of CAI [14, 15].

CA-IV is a membrane-bound enzyme that plays a role in regulating acid–base balance by facilitating the removal of excess acid from the retina into the choriocapillaris [16]. CAIs are predominantly found in peripheral retina cones, with 91% testing positive, while rods show no CA activity. CA-negative cones, likely blue-sensitive, are absent near the foveal center and display distinct morphological traits compared to CA-positive cones [17]. CAIs enhance the pumping function of RPE cells by altering the activity of ion transport mechanisms. Additionally, CAIs can improve choroidal blood flow and cellular adhesion. These effects are partially mediated through the modulation of CA-IV activity and other enzymes such as gamma-glutamyl transpeptidase [1820].

Multiple isozymes of CA exist in the choroidal plexus and other parts of the brain [21]. CA-II is highly expressed in oligodendrocytes, choroidal plexus, myelinated tracts, myelin sheaths, and astrocytes [21]. CA-III is present in the choroidal plexus but has a lower catalytic activity compared to CA-II [21, 22]. Highly expressed CA-II contributes to cerebrospinal fluid (CSF) production by choroid plexus, affecting bicarbonate and sodium exchange. CA-XII is another isozyme involved in CSF production, which is expressed on the basolateral membrane of the choroid plexus epithelial cells in the brain [23].

Systemic Carbonic Anhydrase Inhibitors

Acetazolamide

Pharmacology

Chemical characteristics of acetazolamide (AZA) are displayed in Table 2. The reports on the ocular hypotensive effect of AZA through the reduction of aqueous humor formation date back seven decades [24, 25]. Besides its hypotensive effect through enzymatic inhibition, AZA can further decrease aqueous humor production by inducing systemic and intraocular acidosis [14, 15]. Oral AZA reduces IOP by 30% to 40% [26]. AZA is ineffective in lowering the IOP in individuals with CA-II deficiency [27]. AZA increases blood flow and velocity within the middle cerebral artery. It also reversibly inhibits aquaporin 4, demonstrating a carbonic anhydrase-independent effect on water exchange [28], which is particularly noteworthy given its expression in the brain and the retinal Müller cells [29]. However, reports on its effects on retrobulbar and retinal blood flow are mixed [38, 53, 54]. AZA also exhibits other clinical effects, including enhancing adhesion between the retina and RPE, facilitating fluid movement from retina to choroid, and subretinal fluid resorption [30, 31].

Table 2.

Chemical characteristics of systemic carbonic anhydrase inhibitors

graphic file with name 40135_2025_332_Tab2_HTML.jpg

*Two-dimensional chemical schemas were used from the https://pubchem.ncbi.nlm.nih.gov/ database

Pharmacokinetics

Pharmacokinetics of AZA is displayed in Table 3. It is well absorbed, with an oral bioavailability of 60–100%. Its maximum hypotensive effect is correlated with its tissue-bound concentration, not the plasma concentration, and is achieved when tissue-bound concentration approaches 99% of its maximum value [14]. The duration of action is inversely correlated with the drug's tissue dissociation rate. AZA has limited solubility in water, with 70–95% in a protein-bound form, mainly to albumin, limiting its distribution to the extracellular fluid. Despite its limited volume of distribution (0.2–0.3 L/kg), the drug reaches all organs, including the central nervous system [8, 32, 33]. AZA does not undergo metabolic alteration, kidneys excrete 70–100% of the administered dose. It is moderately dialyzable via hemodialysis (30% clearance with a 4-h hemodialysis). Peritoneal dialysis has limited clearance efficacy (less than 10% clearance over a 24-h dialysis).

Table 3.

Pharmacokinetic information of systemic carbonic anhydrase inhibitors

Drug Form Onset of Action Maximum Effect on IOP Return to Baseline IOP Peak plasma concentration Traceable in Plasma Serum Half-life
Acetazolamide Tablets 1–1.5 h 2–4 h 4–12 h 1–4 h 8–12 h 2–6 h
Slow-Release capsules 2–4 h 8 h 12–24 h 8–18 h 18–24 h
Intravenous Injection 5–10 m 15–30 m 4–6 h 15 m 4–5 h
Methazolamide Tablets 2–4 h 4–6 h 12–24 h 6–8 h ∼21 h 14 h
Ethoxzolamide Tablets 2 m 5 h 12 h N/A N/A 6 h
Dichlorphenamide Tablets 30 m 2–4 h 6–12 h 1.5–3 h N/A 32–68 h

IOP Intraocular pressure. h hours. N/A not available

The Dose and Route of Administration

Different formulations of AZA are displayed in Table 4. The recommended dose of AZA for children is 10–20 mg/kg/day [34], and in adults, the maximum dose of 1000 mg/day. AZA is typically prescribed at dosages of 250 mg 4 times daily or 500 mg in sustained-release preparations twice daily [35, 36]. AZA sustained-release capsules taken twice a day are better tolerated and have a comparable IOP-lowering effect of tables taken four times a day. However, the cost of sustained-release capsules may be a limiting factor.2 The ocular hypotensive effect of AZA is dose dependent. In case of an emergency, intravenous or oral doses of 250–500 mg of AZA are administered. A 45% reduction in IOP can be achieved when the serum concentration of AZA reaches 15–20 μg/ml [36]. Some studies suggest that plasma levels greater than 10 μg/ml might not provide an additional hypotensive effect [37].

Table 4.

Different formulations of systemic carbonic anhydrase inhibitors

Generic preparation Other Names Dosage Protein Binding Ka1 × 109 (M) pKa1 Solubility in water (mM) Metabolism Renal Excretion
Acetazolamide Diamox tablets

125 mg

250 mg

95% 6 7.4 3 Minimal 100%
Diamox sequels

250 mg

500 mg

Diamox parenteral 500 mg/vial
Methazolamide Neptazane

25 mg

50 mg

55% 8 7.2 5 75% 25%
GlaucTabs
MZM

Ethoxzolamide

(No more available in the market)

Cardrase

125 mg

250 mg

95% 1 8.1 4 60% 40%
Ethamide
Dichlorphenamide Daranide 50 mg 88% 18 8.3 N/A Minimal 100%
KEVEYIS
Ormalvi

Ka Absorption constant. pKa degree of ionization. N/A not available

Methazolamide

Pharmacology

Chemical characteristics of Methazolamide (MZM) are displayed in Table 2. MZM induces less metabolic acidosis, potentially explaining its milder ocular hypotensive effect [38]. At a 100 mg dose, MZM will bind to all CA-II isozymes and then to CA-I. The hypotensive effect of the drug occurs when the blood concentration reaches 2–4 μg/ml, achievable with a dosage of 50–100 mg twice daily [3941].

Pharmacokinetics

Pharmacokinetics of MZM is displayed in Table 3. MZM exhibits slower intestinal absorption and blood clearance than AZA. Approximately 95% of the drug is reversibly bound to CA in red blood cells (RBCs). MZM has lower plasma protein binding (55%) and better distribution inside the eyes and central nervous system compared to AZA. The metabolic pathway of MZM is yet to be fully investigated, but studies suggest that hepatic enzymes such as cytochrome P450, glutathione, and β‐lyase are involved in its metabolism [40, 4244]. Unlike AZA which undergoes up to 100% active renal excretion, only 25% of MZM is excreted unchanged in the urine, resulting in a longer half-life and requiring fewer doses [38, 45]. This makes MZM a suitable choice for patients with mild to moderate renal impairment and compromised glomerular filtration rate, as it is associated with fewer renal side effects and is preferable for individuals at high risk of kidney stones [39].

The Dose and Route of Administration

Different formulations of MZM are displayed in Table 4. MZM is available in 25 mg and 50 mg tablets and is used 2–3 times daily. To reduce the side effects, MZM may be started at 25 mg twice daily and then increased to 50 mg twice daily if needed [34, 38, 46]. The dosage can be escalated up to 100 mg three times daily. MZM at doses of 25, 50, and 100 mg three times daily may lead to reductions in IOP of 3.3, 4.3, and 5.6 mmHg, respectively [47]. To minimize the gastrointestinal side effects, it is recommended to take the medication after meals.

Ethoxzolamide

Pharmacology

Chemical characteristics of ethoxzolamide are displayed in Table 2. Although ethoxzolamide is a potent systemic CAI in vitro, its high degree of protein binding and rapid uptake into adipose tissue reduce its effectiveness in vivo. These factors limit its bioavailability and therapeutic potential despite its strong in vitro enzyme-inhibitory properties [48].

Pharmacokinetics

Pharmacokinetics of ethoxzolamide is displayed in Table 3. Ethoxzolamide is well-absorbed from the gastrointestinal tract after oral administration. Its high protein binding and rapid uptake into adipose tissue, limits its availability in the systemic circulation which reduces its effectiveness. Ethoxzolamide is metabolized in the liver. The metabolic pathways and the exact metabolites have not been well-documented [48]. The primary route of excretion is through the kidneys, but a high proportion of the drug will remain in the adipose tissue for several months [48, 49].

The Dose and Route of Administration

Different formulations of ethoxzolamide are displayed in Table 4. However, they are no longer available on the market. Previously, ethoxzolamide was supplied in 125 mg tablet form and was prescribed in doses ranging from 62.5 to 250 mg every 4 to 8 h. Toxicology studies indicated that high doses could cause occasional gastric petechiae and hydronephrosis in rats, as well as potassium depletion in dogs [45, 48].

Dichlorphenamide

Pharmacology

Chemical characteristics of dichlorphenamide are displayed in Table 2. As a CAI, it increases the urinary excretion of sodium, bicarbonate, and potassium, leading to metabolic acidosis. However, compared to AZA, it induces milder metabolic acidosis due to its inherent chloruretic activity [50]. Dichlorphenamide is more potent than AZA, being 30 times more effective in vitro and approximately 30% more effective in vivo for managing acute angle closure attacks [51]. One-third of the dosage of dichlorphenamide is required to achieve the same effect as AZA. But, dichlorphenamide may cause more symptoms and dose-dependent side effects compared to other CAIs [52, 53].

Pharmacokinetics

Pharmacokinetics of dichlorphenamide is displayed in Table 3, thought, there are limited studies on its pharmacokinetics. It is well absorbed orally and undergoes hepatic metabolism. Dichlorphenamide and its metabolites are primarily eliminated via the kidneys. Its effect tends to start earlier than AZA. Following oral administration of a 50 mg tablet, IOP drops within 30 min, reaches its lowest point within 2 to 4 h, then returns to baseline within 6 to 12 h [45, 51, 54]. Dichlorphenamide is a weak activator of hepatic enzymes such as cytochrome P450 [5456].

The Dose and Route of Administration

Different formulations of dichlorphenamide are displayed in Table 4. Dichlorphenamide is available in 50 mg tablets. The initial recommended dose is 50 mg every 12 h, which can be increased to every 6 to 8 h, up to a maximum daily dosage of 200 mg. Although studies on dichlorphenamide use in glaucoma management date back to 1958 [53], it is not as widely prescribed as AZA [51].

Indications In Ophthalmology

Pediatric Glaucoma

Systemic CAIs are valuable medications in congenital glaucoma cases to lower the IOP prior to surgery [57]. They effectively reduce the IOP by 20–35% and may be taken with milk or food to improve tolerability. Compared to adults, CAIs may lead to a greater IOP reduction in pediatrics, which could be attributed to a decrease in CA activity with aging [58]. The pediatric dosage of AZA is 10–20 mg/kg/day, 2–4 times daily, while MZM dose is 2 mg/kg/day, twice daily [59]. One of the listed side effects of systemic AZA is growth retardation due to metabolic acidosis [59]. Given the children's limited ability to communicate, close monitoring of adverse effects is crucial. Oral sodium citrate (1 mEq/kg/day) has been used to treat the metabolic acidosis of systemic CAIs [60]. In a clinical study involving long-term (> 3 months) AZA use, no weight changes were observed in 22 children aged 2 months to 15 years [61]. However, any loss of appetite or weight loss should be monitored for early detection of potential problems [62].

Adult Glaucoma

Acute Angle Closure

In acute angle-closure attacks, 250–500 mg of AZA is administered orally or intravenously and then every 4 to 6 h if needed. Intramuscular administration is allowed but is painful. In individuals with no prior history of renal dysfunction or electrolyte disorders, are used safely for few days/weeks [63, 64]. The use of an anti-emetic agent can enhance tolerance in cases of nausea and vomiting. After successful treatment of an acute angle-closure attack, the ciliary body responds to IOP spike by entering a transient hyposecretion phase, which can result in temporary hypotony. Consequently, systemic CAIs should be discontinued. However, patients may require continued use of systemic CAIs until receiving laser iridotomy or cataract extraction [63].

Open-Angle Glaucoma

Long-term use of systemic CAIs in open-angle glaucoma is not recommended due to their potential adverse effects. They are typically reserved for cases where topical agents fail to reduce IOP to the target level, or in patients who decline any form of surgical intervention despite having uncontrolled IOP with maximum tolerated medical therapy [6569]. Dose-dependent reduction of IOP of 15–30% has been reported with systemic CAIs. However, a small percentage of individuals may not respond to CAIs [70]. Systemic CAIs are traditionally used as one of the first-line treatments in eyes with inflammatory glaucoma. In inflammatory glaucoma associated with corneal edema, systemic CAIs may be the best choice since absorption of topical medications is limited, and the topical CAIs may impact corneal endothelial cell function [71].

In long-term treatment with systemic CAI, the patients need to be monitored closely for potential adverse effects, and involving primary care physicians seems reasonable. Reduced survival of glaucoma patients on chronic oral AZA was reported in one study from Norway that included 1147 patients with primary open-angle glaucoma or pseudo exfoliative glaucoma [72]. AZA users were defined as patients who used the drug for at least 2 years and/or had left the hospital after their last visit with a prescription for daily dose of 250–500 mg AZA (N = 492). While the average patient longevity was 14.2 ± 8.3 years, in the multivariable COX proportional hazards model adjusting for the glaucoma type, age at diagnosis, gender, and year of diagnosis, AZA use was associated with reduced survival (HR = 1.152, P = 0.021) with an unclear reason [72].

Systemic CAIs may be used to prevent IOP spikes following intravitreal injections or cataract surgery. In open-angle glaucoma or glaucoma suspects, prophylactic AZA, 60–90 min before intravitreal injection, lead to a significantly lower IOP in the AZA group (mean: 15.7 mmHg, range: 8–21 mmHg), compared to the control group (mean: 20.6 mmHg, range: 11–46 mmHg) 30 min after injection [73]. A clinical trial investigated the effect of different IOP-lowering interventions on eyes receiving intravitreal bevacizumab by comparing AZA, topical brimonidine, anterior chamber paracentesis, and no treatment. Anterior chamber paracentesis prevented the IOP elevation following injection, whereas AZA and brimonidine caused a faster return to baseline IOP than the untreated group [74]. Eyes with primary open-angle glaucoma may experience marked IOP elevation 3–7 h after cataract surgery. An oral dose of 500 mg AZA, one hour before or 3 h after cataract surgery reduces the incidence of postoperative IOP elevations [75]. Following cataract surgery, short-term oral AZA offered better IOP control than topical dorzolamide 2% in preventing IOP spikes [76]. Similarly, patients who received AZA before vitrectomy or buckling surgeries had lower post-operative IOP (22.11 mmHg vs. 36.36 mmHg), and no adverse effects were noted [77].

In allergic contact dermatitis caused by glaucoma eyedrops, a potential management approach is discontinuing the eyedrops and initiating a short-term course of systemic CAIs. After approximately 3–4 weeks, the glaucoma eye drops can be reintroduced individually to identify which one was the cause of the allergic reaction. Alternatively, preservative-free glaucoma medications may be used, but the cost and their availability may be the limiting factor. This approach allows targeted identification of the causative agent and facilitates the implementation of appropriate measures to manage the patient’s condition effectively. Figure 1 illustrates the resolution of the allergic contact dermatitis induced by topical agents 3 weeks following the discontinuation of the eyedrops and taking oral AZA. Similarly, some glaucoma surgeons replace topical glaucoma medications with oral AZA and start topical steroids before trabeculectomy to decrease the conjunctival inflammation and reduce the incidence of bleb fibrosis and failure [78, 79]. It has been shown that long-term use of topical glaucoma medications can lead to histologic changes of the conjunctiva, with increased numbers and activity of lymphocytes, monocytes, fibroblasts, and conjunctival goblet cells. These changes ultimately contribute to bleb scarring [80, 81]. However, Lorenz et al. reported that preoperative, preservative-free, fixed combination dorzolamide-timolol was equally effective as preoperative oral AZA and topical dexamethasone in terms of IOP reduction and need for bleb needling or suture lysis. Notably, more patients reported adverse events in the AZA group than in the dorzolamide-timolol fixed combination group [82].

Fig. 1.

Fig. 1

Figure 1: resolution of allergic contact dermatitis after discontinuation of the eyedrops and taking oral acetazolamide

The effect of CAIs on intracranial pressure (ICP) has been well described [83]. According to Laplace’s law and stress formula, translaminar pressure depends on IOP, ICP, and lamina cribrosa. Some studies consider low ICP as a glaucoma risk factor [84, 85]. Loiselle et al. observed a significant reduction of ICP of 4 mmHg with an oral dose of 125 mg AZA in glaucoma patients but not in healthy subjects. However, this intervention did not affect the IOP in either group, but increased translaminar pressure in glaucoma patients [86]. CAIs can affect both ICP and IOP, but how this relates to clinical management and outcomes has not yet been determined.

Retinal Diseases

CAIs are used in the management of cystoid macular edema (CME) and serous retinal detachment in patients with a variety of underlying pathologies, including retinitis pigmentosa, uveitis, Sturge-Weber syndrome, morning glory syndrome, optic disc pit, and central serous chorioretinopathy. CAIs affect CME by stimulating outward active transport and passive permeability across the blood-retinal barrier. AZA blocks the active transport of HCO3 and Cl across the RPE and also increases the resorption rate of subretinal fluid. Acidifying the subretinal space and improving fluid hydrodynamics through RPE cells is another aspect leading to resolution of CME.

A common starting dose for AZA is 500 mg daily for a duration of 4 weeks. The dosage should be adjusted based on the patient's age and response to treatment [87]. Some uveitic patients may require a maintenance dose of 125 to 500 mg daily [88]. In cross-over trials on uveitic CME, patients taking a 4-week course of AZA showed a significant decrease in CME (25%) compared to placebo; however, no significant change in the visual acuity was observed [89].

AZA at the dosage of 500 mg/day for two weeks improved visual acuity in a patient with retinitis pigmentosa and chronic macular edema. Additionally, half of the patients had decreased angiographic CME, primarily due to less leakage [90]. MZM improved the angiographic CME in 9 of 17 retinitis pigmentosa patients. Although angiographic CME improved, notable or even moderate visual acuity improvement was observed in only four subjects [91]. In Yeo et al. study, the eyes with CME extending to the outer nuclear layer or central fovea, milder initial photoreceptor damage, and early treatment were more likely to have visual improvement. No significant changes were observed in microvascular parameters with treatment in those with visual improvement [92]. A systematic review and meta-analysis of 32 studies about CME management in retinitis pigmentosa found that topical and systemic CAIs and local steroids effectively reduce central macular thickness and improve visual acuity [93].

A study on the effect of oral CAIs showed that the time for subjective and objective resolution of serous retinal detachment in central serous choroidopathy was shorter with AZA compared to sham. However, the two groups had no difference in the final visual acuity or recurrence rate [94]. Rübsam et al. compared the efficacy of mineralocorticoid-receptor antagonists, AZA, and observation in central serous chorioretinopathy. Both mineralocorticoid-receptor antagonists and AZA were effective in decreasing the macular thickness and subretinal fluid with a corresponding improvement in visual acuity [95]. There is no definite evidence regarding the use of systemic CAIs in central serous retinopathies. Case reports showed the effectiveness of systemic CAIs in treating serous retinal detachment in Sturge-Weber syndrome, morning glory syndrome, and optic disc pit [9699] Further research is needed to provide a comprehensive understanding of the optimal use of systemic CAIs in CME management [57].

Idiopathic Intracranial Hypertension

Systemic CAIs are first-line treatments for the medical management of IIH, they lower the ICP by reducing CSF secretion [22]. A 99.5% reduction in CA activity in the choroid plexus is required to decrease the CSF production [100]. AZA is the only medication with level A evidence efficacy (well-conducted randomized controlled trials or meta-analyses of multiple randomized clinical trials with consistent results) to manage IIH. The resolution of papilledema starts after an average treatment period of 40 days, and the complete resolution may take up to 90 days [101].

Early studies suggested AZA-induced weight loss was the mechanism of the improved visual field, not the ICP reduction [102]. This was later disapproved by NORDIC Idiopathic Intracranial Hypertension Study Group trials. In a randomized, double-masked, placebo-controlled trial on 165 IIH patients with mild visual loss, participants were given a low-sodium weight-reducing diet and either AZA (up to 4 g daily) or a placebo for 6 months. The primary outcome was the perimetric mean deviation change. The perimetric mean deviation improvement was greater with AZA (1.43 dB) than placebo (0.71 dB). There was also improvement in papilledema grade and vision-related quality of life with AZA. The study reported that both the placebo (3.45 kg) and the AZA groups (7.50 kg) lost weight, obviously 4.05 kg more in the AZA group. Further analysis demonstrated that the benefit of AZA on perimetric mean deviation was not via its effect on weight, suggesting that the main mechanism of effect was through reduction of sodium ion transport across the choroid plexus with subsequent reduction of CSF production [103, 104]. In a randomized trial, 40 patients received AZA or topiramate and were monitored over 1 year. Both drugs showed statistically significant improvement of visual fields, with no significant difference between the two groups [105]. MZM could be used in patients with limited AZA tolerance or with renal and electrolyte disorders [57]. In addition to the improvement in papilledema, studies have reported the resolution of the sixth nerve palsy seen in IIH patients [106108].

Side Effects

Systemic CAIs, either for short- or long-term use, could be associated with several side effects attributed to the inhibition of CA enzyme, present in most tissues ranging from RBCs, renal tubules, gastric mucosa, pancreatic cells, eye, lung, testis, brain and bone osteoclasts (Table 5).

Table 5.

Side effects of systemic carbonic anhydrase inhibitors

Ocular Changes in color vision
Induced myopia
Secondary angle closure
Choroidal effusion
Gastrointestinal Dysgeusia
Dry mouth
Reflux and peptic ulcer disease
Nausea and vomiting
Diarrhea or constipation
Abdominal cramps
Liver Injury
Renal Mild metabolic acidosis
Alkaline diuresis ± Polyuria
Nephrolithiasis
Hematologic Blood dyscrasias
Myelosuppression: thrombocytopenia, and aplastic anemia
Dermatologic Steven-Johnson syndrome and toxic epidermal necrolysis
Skin rash: Morbilliform or urticarial
Neurologic Perioral and distal extremity paresthesia
Malaise, fatigue, confusion, and drowsiness
Anorexia and depression
Headache, tinnitus, and vertigo
Irritability, delirium, and insomnia
Tremors
Decreased libido
Disturbed balance

Ocular

Ocular adverse effects of CAIs include changes in color vision, transient myopia, secondary angle closure attack, and choroidal effusion [109, 110]. Disturbance of color vision is attributed to the inhibition of CA enzymes in the cones, resulting in decreased cone sensitivity to blue-green light [110, 111]. AZA-induced ciliary body edema causes forward displacement of lens-iris diaphragm, leading to myopia and secondary angle closure [112]. The onset of transient myopia can occur as early as 5 h to 5 days after taking the medication. Myopia resolves in a few days after cessation of the AZA [113].

Gastrointestinal

CA-II enzyme is widely distributed in the epithelial layer of the gastrointestinal tract, bile duct, pancreatic duct, gall bladder, hepatocytes, and serous acinar cells of the parotid, submandibular, and gastric parietal glands, with the primary role of supplying bicarbonate ions to the secretions [114]. The vast range of gastrointestinal side effects include dysgeusia, dry mouth, reflux, nausea, vomiting, diarrhea, constipation, abdominal cramps, and peptic ulcer disease. AZA increases the odds of nausea, gastroesophageal reflux disease, and diarrhea by 2.6—4 folds [5, 115]. Oral administration of sodium acetate or sodium bicarbonate helps alleviate these symptoms in 50% of patients by partly correcting the CAI-induced metabolic acidosis [116, 117]. Epstein et al. reported that administering the drug with food relieves these gastrointestinal symptoms in about 50% of patients [116].

Renal

CA-II enzyme is the predominant isoform of renal CA. It is expressed widely by multiple segments of the nephrons, including proximal convoluted, straight tubules, Henle’s loop, and intercalated cells of the cortical medullary collecting ducts [118]. Systemic CAIs induce mild metabolic acidosis and alkaline diuresis, presenting as polyuria in some patients. In normal individuals, the amount of diuresis is limited as the distal tubules reabsorb the filtered bicarbonate, fluid, and salts. In contrast, these symptoms are exacerbated in diabetics and those with chronic kidney disease, wherein the risk of metabolic acidosis is higher due to delayed drug clearance [119].

Prolonged administration of AZA may cause nephrolithiasis due to a decreased urinary citrate, resulting in the accumulation of calcium phosphate. Furthermore, an increase in urinary oxalate contributes to the formation of calcium oxalate stones [120]. Kass et al. reported 11 times increase in the incidence of nephrolithiasis in patients on AZA. The mean interval to stone formation was 14.4 months [121]. The formation of renal stones is not dose-dependent [122]. In patients with renal stones, it is essential to assess the renal function, weigh the risks, and exercise utmost caution.

Hematologic

Systemic CAIs are occasionally associated with blood dyscrasias, myelosuppression, thrombocytopenia, and aplastic anemia [123125]. This does not seem to be due to CA inhibition, but an idiosyncratic reaction, which is not dose-dependent, unpredictable, and delayed in onset [126, 127]. Keisu et al. reported 11 cases of AZA-induced aplastic anemia in patients aged 63–85 years over a period of 17 years with an estimated incidence of 1 in 18,000 and relative risk of 13.3 (95% confidence limits: 6.8- 25.3) [128]. Patients on long-term oral AZA should be counseled regarding symptoms and signs of bone marrow failure, such as easy bruising, frequent infections, and fatigue. Some recommend obtaining complete blood counts bimonthly during the first 6 months of therapy, followed by every 6 months thereafter [129].

Dermatologic

Steven-Johnson Syndrome/Toxic Epidermal Necrolysis

SJS and TEN are rare skin diseases, presenting with extensive involvement of skin and mucus membranes triggered by certain drugs or infections. SJS/TEN forms a spectrum wherein SJS affects less than 10% of the skin, and TEN over 30% of skin (more severe and life-threatening). The estimated incidences of SJS, SJS/TEN, and TEN in the US are 9.2, 1.6, and 1.9 per million adults per year, respectively [130]. Drug-induced stimulation of cytotoxic and helper T-cells results in a type IV delayed hypersensitivity autoimmune reaction leading to epidermal necrosis. CAI-induced SJS/TEN results in more extensive cutaneous manifestations with frequent ocular complications compared to other drugs causing SJS/TEN [131].

AZA-induced SJS/TEN is associated with HLA-B59 in patients of Korean descent [132]. A similar association between HLA-B*5901 and MZM-induced SJS/TEN is seen in patients of Japanese and Korean descent [133135]. An extensive exome-wide and HLA-based association study by Jiang et al. identified HLA-B*55:02 as a novel risk allele in Han Chinese. The specific haplotype is invaluable as a genetic predictor of MZM-induced SJS/TEN with a sensitivity of 89% and specificity of 96% [136]. With studies showing increasing evidence of association between HLA-B*5901 and HLA-B*5901-Cw*0102 haplotype and MZM-induced SJS/TEN in Korean and Hans Chinese population, genetic screening prior to starting MZM would be beneficial in these populations [137]. In a study on 8 Korean patients with MZM-induced SJS/TEN, the mean duration of drug exposure was 22.0 ± 16.9 days (range: 2–58 days). The patients received MZM at a dose of 50 mg/day for glaucoma and postoperative IOP spike management. The latent period to symptoms varied from 12 to 58 days, with a mean of 23.0 ± 15.7 days [138].

Skin Rash

Skin eruptions following systemic use of AZA and MZM have two distinct patterns, namely morbilliform and urticarial. The maculopapular rash has a bilateral symmetric distribution involving the trunk and extremities, which disappears within 2–3 weeks after discontinuation. The urticarial type presents with a pruritic rash and swelling of the trunk and extremities. The diffuse erythematous rash are round edematous papules or arcuate plaques [139]. Maculopapular exanthema and acute generalized exanthematous pustulosis are reported with AZA [140].

Neurologic

CA enzyme present in neurovascular unit plays an important role in maintaining the pH, formation and distribution of CSF, and regulation of cerebral blood flow [141]. Systemic AZA causes perioral and distal extremity paresthesia which is the most common dose-dependent adverse effect. The reported incidence of paresthesia varies between 51–70%, with an increased incidence in patients on higher dosages. However, this is readily tolerated and is alleviated by oral administration of potassium chloride or reducing the dosage of AZA [5, 142].

Other systemic CAIs induced adverse neurological effects include malaise, fatigue, confusion, depression, drowsiness, dysgeusia, headache, vertigo, irritability, insomnia, tremor, tinnitus, decreased libido, and disturbed balance. AZA-induced metabolic acidosis may lead to lethargy and malaise, which improves with taking bicarbonate or acetate supplements. MZM-induced delirium presents as intermittent psychosis, lethargy, disorientation, and incontinence of bowel and bladder, which resolves within a week after discontinuation [143].

Special Situations and Contraindications

Pulmonary Diseases

AZA may be used as a respiratory stimulant in chronic obstructive pulmonary disease (COPD) with the goal of improving oxygenation, reducing carbon dioxide retention, and aiding liberation from mechanical ventilation and/or attempting to correct a metabolic alkalosis [144]. Accordingly, if a patient has chronic respiratory acidosis with metabolic compensation (mild alkalemia) at baseline, inducing additional metabolic acidosis may be acceptable and may even stimulate respiratory drive [145]. However, it is contraindicated in patients with acute COPD exacerbation as it may aggravate respiratory acidosis in the absence of metabolic compensation [145].

Liver Diseases

Systemic CAIs induced liver injury is rare, few cases have been reported [146148]. The mechanism of hepatic injury is presumed to be due to hypersensitivity reaction similar to the sulfonamides induced liver damage, and occurs days to weeks after administration. The severity of the hepatic injury ranges from mild, asymptomatic elevations of liver enzymes to a fulminant hepatitis [149]. Accordingly, systemic CAIs should not be used in patients with liver cirrhosis, while in mild liver disease, dose adjustment is necessary. Hepatic involvement and jaundice can also occur in cases of SJS but are generally overshadowed by the severe cutaneous reaction [149]. Corticosteroids’ role in altering the ultimate outcome of hepatic injury is unproven. Rechallenge should be avoided, and patients should be warned about any sulfa-containing medications in the future [149].

Kidney Diseases

Systemic CAIs may cause electrolyte imbalances and are not recommended in patients with renal impairment. Advanced renal dysfunction is a contraindication to systemic CAIs use, as AZA is eliminated exclusively by the kidneys and its use induces bicarbonaturia and metabolic acidosis [150]. Taking sodium bicarbonate to offset the systemic acidosis of AZA may increase the risk of kidney stone formations [145]. Oral AZA is excreted unchanged thought the kidneys. Therefore, creatinine clearance (CrCl) directly influences its efficacy and the duration of action. The following dosages are recommended in renal dysfunction based on the CrCl: [145, 151153] CrCl > 50 mL/min requires a full dose (250 mg four times a day or 500 mg extended-release capsule twice daily), CrCl 10 to 50 mL/min requires a half dose (250 mg twice daily), while AZA should not be used for CrCl < 10 mL/min.

The recommended dose for those on hemodialysis and peritoneal dialysis are 250 mg twice daily and 125 mg daily, respectively. If a patient is dialysis or has severe kidney disease, appropriate communication with the nephrologist is mandatory, with monitoring of central nervous system disturbances. The dose adjustment may reduce the IOP-lowering effect of AZA [145].

The metabolism of MZM (where only 25% of MZM is excreted unchanged in the urine) makes it a safer choice than AZA in patients with advanced renal disease (e.g., diabetic patients with neovascular glaucoma and expected diabetic nephropathy). Hypothetically, urolithiasis is also less likely with MZM than AZA due to less suppression of urine citrate and alkalinization, therefore, MZM is preferred to AZA in patients with a history of renal lithiasis [45].

Sickling Disorders

Systemic CAIs not only promote hemoconcentration but also induce systemic acidosis which are important factors in erythrocyte sickling. Besides lowering the aqueous humor pH, AZA increases the concentration of ascorbic acid in aqueous humor which exacerbate the sickling. MZM may be more desirable as it creates less systemic acidosis than AZA. If a systemic CAI use is necessary in treating high IOP associated with hyphema in a sickle cell patient, MZM is the preferred agent [154].

Pregnancy and Lactation

According to the US Food and Drug Administration, AZA and MZM are classified as a class C agent. This indicates observation of adverse effects on animal fetuses with lack of well-controlled studies in humans. As class C medications, AZA and MZM should be contraindicated in pregnancy, and prescribed only if the potential benefit justifies the risk to the fetus [155]. Congenital malformations, such as ectrodactyly, syndactyly, skeletal ossification disturbance, axial skeleton malformation, exencephaly, anophthalmia, microphthalmia, cleft lip/palate, and retarded incisor teeth development, have been reported in experimental animals [156159]. However, animal studies have shown teratogenicity at doses over 10 times the recommended human dose.

In humans, there are no adequate and well-controlled studies in pregnant women on systemic CAIs [160, 161]. In an observational study of 101 women with IIH, AZA use before 13 weeks of gestation was not associated with any major complications in the offspring [161]. In the retrospective study of the Collaborative Perinatal Project (CCP), 50,282 mother–child pairs were evaluated, 12 had first-trimester exposure to the drug, but no anomalies observed. In a total of 1024 patients with any exposures (at any trimester), no major or minor fetal anomalies were observed [162]. On the other hand, case reports of congenital malformations of the offspring have been described which include conditions such as teratoma [163], ectrodactyly, syndactyly, oligodontia [164], Additionally transient metabolic side effects and electrolyte abnormalities in newborns, such as hypocalcemia, hypomagnesemia, and metabolic acidosis have been described [165, 166].

AZA is excreted into human milk in minimal quantities. The benefits should outweigh the risks when used during breastfeeding. The infant's estimated dose from a maternal dose of 1000 mg a day is less than 0.7% of the maternal weight-adjusted dose [167]. American, Canadian, and French professional guidelines consider CAIs acceptable in breastfeeding [168170]. However, product manufacturers advise using with extreme caution in breastfeeding [167].

Drug Interactions

The most common drug interactions of systemic CAIs are listed in Table 6.

Table 6.

Drug interactions of systemic carbonic anhydrase inhibitors

Drug Risks of Concomitant Use
Metformin Lactic acidosis
Poor glycemic control
Aspirin Anorexia
Tachypnea
Coma
Death
Anticonvulsants

Phenytoin

Carbamazepine

Reduced metabolism and increased serum levels
Topiramate Metabolic acidosis
Kidney stones

Metformin

Metformin has emerged as the most widely prescribed antidiabetic medication in type II diabetes and also for weight reduction [171]. Lowering the hepatic glucose production by metformin may cause a shift from aerobic to anaerobic metabolism. This may increase lactate production, causing lactic acidosis [172]. Inhibition of CA enzyme is also associated with a decrease in hepatic glucose production. Significant reduction in circulating blood glucose and elevated lactate level in diabetic rats treated with AZA were observed [173]. Caution must be exercised in prescribing CAIs in patients taking metformin, as concomitant use of both medications may increase the risk of lactic acidosis and poor glucose control [145].

Aspirin

AZA inhibits CYP3 A4 enzyme which enhances the adverse effect of aspirin. Additionally, salicylates appear to competitively inhibit the plasma protein binding and renal tubular secretion of AZA. Caution is advised when AZA and salicylate are used concurrently as severe adverse effects have been reported, including anorexia, tachypnea, coma, and even death, likely as a result of acute salicylate intoxication secondary to its reduced liver metabolism [145]. The adverse events can be more severe in renal insufficiency [174, 175]. Accordingly, patients with any degree of renal impairment should avoid taking AZA concomitantly with aspirin [175].

Anticonvulsants

Systemic AZA is occasionally prescribed by neurologists as an adjunct therapy in epilepsy, as the AZA induced acidosis may reduce the spread of seizure leading to reduction of the number of attacks or even complete remission of seizures [176]. AZA may increase serum levels of anticonvulsants such as phenytoin or carbamazepine due to inhibition of CYP3 A4 metabolism [177], Consultation with the patient’s neurologist is recommended before prescribing AZA in epileptic patients. Additionally, caution should be exercised in prescribing AZA in patients taking topiramate. Because topiramate inhibits CA enzyme, and its concomitant use with CAIs increases the risk of metabolic acidosis and kidney stones [177].

Tolerability, Adherence, and Monitoring

Once the need for systemic CAIs is confirmed, oral AZA can be started as long as there are no contraindications. Because there is no antidote to AZA, it is essential to emphasize to the patient to seek immediate care if there is an overdose. Symptoms include dizziness, confusion, loss of consciousness, and convulsions. The pharmacist should be consulted on medication reconciliation to ensure there is no drug-drug interactions or have the primary care physician involved before starting the medication and monitoring the patient.

Counseling the patient is important. Encouraging the patient to drink additional water and take a high-potassium diet (banana, avocado, raisins, apricots, beans and lentils, potatoes, winter squash, spinach, and broccoli) may be helpful even with short-term use. The risk of kidney stone formation with long-term use may be reduced by dietary intake of certain foods that induce a mild metabolic alkalosis, which may theoretically increase urine citrate and possibly lower the risk (citrate inhibits crystallization of calcium salt) [145].

Blood urea nitrogen to creatinine ratio, complete blood count, and serum electrolytes may be tested at baseline before starting systemic CAIs and monitored periodically; however, testing these values for all patients is a controversial topic, and there is limited evidence on the possible benefits. For at-risk patients such as individuals with recurrent renal stones, chronic kidney disease, and diabetic patients on metformin, electrolyte levels (potassium and sodium in particular) and blood gas analysis are recommended before starting the medication and 3–6 months thereafter. Monitoring liver enzymes may be considered as well, as hepatotoxicity is a reported rare adverse event [178].

The frequent systemic side effects associated with the use of systemic CAIs may adversely affect compliance. A study evaluated the tolerability and efficacy of dorzolamide in those who were intolerant to systemic CAI. Within 4 weeks of switching from systemic CAI to dorzolamide, the mean health assessment scores improved significantly in seven of the eight categories of the SF-36 (The Short Form 36 Health Survey Questionnaire) and did not change for the rest of the study, reflecting the negative impact of systemic CAIs on health-related quality of life [179]. A systematic review evaluated the quality of life in glaucoma and 3 other systemic diseases [180]. In glaucoma patients, the lowest scores in most categories originated from the previous study discussing drug-related adverse effects of systemic CAIs [179]. These findings suggest that long-term use of systemic CAIs is discouraged if there are alternatives. On the other hand, a population-based cohort study in Canada involving 128,942 matched patients over a 25-year period, showed that the absolute risk of a severe complicated adverse event of SJS, TEN, or aplastic anemia was 2.90 per 1000 patients for systemic CAIs, and 2.08 per 1000 patients for topical CAIs. This indicates that the risk of severe adverse reactions from systemic CAIs was low and comparable with topical medications [181].

Expert Opinion

Systemic CAIs are a well-established treatment for various ocular conditions, particularly glaucoma. They are used in both pediatric and adult cases, often in combination with topical medications to temporarily decrease the elevated IOP prior to glaucoma surgery. They are also used in patients who do not tolerate or have contraindications to topical medications, refuse the surgical interventions, are at high risk of surgical complications, or have undergone multiple surgeries without achieving a satisfactory IOP control. In addition to IOP reduction, systemic CAIs are used in retinitis pigmentosa CME and to lower the ICP in IIH.

The systemic CAIs seems to be underutilized by ophthalmologist due to the concerns for severe adverse effects. Oral CAI had an absolute risk of 2.90 per 1000 patients of developing a life-threatening side effect compared to 2.08 per 1000 patients with topical CAIs. The observation of a directly proportional association in the risk of these events over time could reflects patients’ underlying baseline risk of sustaining these events [181]. Considering that oral CAIs are associated with a low incidence of serious adverse effects, their proper use and comprehensive review of systemic diseases and other medications the patient is taking should decrease the occurrence of severe adverse effects. Furthermore, the incidence of severe adverse events is low and in some studies were comparable to that of topical therapy. lack of cost‐effectiveness, limited test availability, and the rarity make widespread or “routine” testing for CA‐II deficiency impractical. Instead, clinicians may reserve CA‐II assays for those with strong clinical suspicion based on characteristic symptoms and family history.

A thorough review of the patient’s medication regimen, particularly anticonvulsants, salicylates, and metformin, is necessary to prevent potential drug-drug integrations. Additionally, initial laboratory tests are suggested to exclude the presence of co-existing conditions. It is beneficial to engage the primary care physician in monitoring and managing patients who have associated systemic comorbidities (such as lung, liver, or kidney diseases) or those who need long-term CAIs treatment to enhance comprehensive care coordination. Initiating treatment at a low dose with a gradual increase, if needed, can enhance patient adherence, and reduce the risk of side effects expected with maximum dosage.

Indeed, in situations where the potential benefits surpass the risks, such as in instances of vision-threatening elevated IOP or ICP, or CME not responding to other treatments, the patient should not be deprived of the sight saving effects of systemic CAIs. This approach aligns with a comprehensive and patient-centered care model, where individual patient factors and circumstances are paramount in decision-making. It’s important to monitor for potential side effects and adjust the treatment plan as necessary. Switching from AZA to MZM seems a reasonable approach for patients with kidney disease, instead of discontinuing systemic CAIs. In situations where patients have minor side effects, applying suitable management and adjusting the dosage might be enough, rather than discontinuing the treatment. It's crucial to perform a tailored evaluation of the advantages and disadvantages for each patient prior to and during the use of systemic CAIs treatment. The continuous monitoring ensures the therapy remains appropriate for the individual's specific situation.

Key References

  • Schmickl CN, Owens RL, Orr JE, Edwards BA, Malhotra A. Side effects of acetazolamide: a systematic review and meta-analysis assessing overall risk and dose dependence. BMJ Open Respir Res. 2020;7(1).
    • Findings from this study was crucial in understanding the side effects of carbonic anhydrase inhibitors.
  • Wall, M.; McDermott, M. P.; Kieburtz, K. D., et al. Effect of Acetazolamide on Visual Function in Patients With Idiopathic Intracranial Hypertension and Mild Visual Loss: The Idiopathic Intracranial Hypertension Treatment Trial. JAMA. 2014;311(16):1641-1651.
    • Findings from this study suggest that addition of Acetazolamide to low sodium diet may provide modest improvement in the visual field in patients with idiopathic intracranial hypertension.
  • Popovic, M. M.; Schlenker, M. B.; Thiruchelvam, D.; Redelmeier, D. A. Serious Adverse Events of Oral and Topical Carbonic Anhydrase Inhibitors. JAMA Ophthalmology. 2022;140(3):235-242.
    • This study suggests that adverse effects following systemic or topical carbonic anhydrase inhibitors use is rare and similar across different agents.

Acknowledgement

Dr. Hallaj reports funding from the National Institutes of Health (grant numbers: OT2OD032644 and T32EY026590).

Author Contributions

All authors have equally contributed to all aspects of this manuscript. All authors reviewed and approved the submission of the manuscript.

Data Availability

No datasets were generated or analysed during the current study.

Declarations

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Competing interest

The authors declare no competing interests.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Shahsuvaryan ML. Carbonic Anhydrase Inhibitors in Ophthalmology: Glaucoma and Macular Oedema. In: Chegwidden WR, Carter ND, editors. The Carbonic Anhydrases: Current and Emerging Therapeutic Targets. Cham: Springer International Publishing; 2021. p. 79–102. [Google Scholar]
  • 2.Clineschmidt CM, Williams RD, Snyder E, Adamsons IA. A randomized trial in patients inadequately controlled with timolol alone comparing the dorzolamide-timolol combination to monotherapy with timolol or dorzolamide 11Ms. Clineschmidt, Dr. Adamsons, Dr. Snyder, and Dr. Reines are employees of Merck & Co., Inc., the manufacturer of the dorzolamide-timolol combination. The other authors have no proprietary interest in the dorzolamide-timolol combination or in Merck. Ophthalmology. 1998;105(10):1952–9. [DOI] [PubMed]
  • 3.Scozzafava A, Supuran CT. Glaucoma and the Applications of Carbonic Anhydrase Inhibitors. In: Frost SC, McKenna R, editors. Carbonic Anhydrase: Mechanism, Regulation, Links to Disease, and Industrial Applications. Dordrecht: Springer, Netherlands; 2014. p. 349–59. [Google Scholar]
  • 4.Meldrum NU, Roughton FJ. Carbonic anhydrase its preparation and properties. J Physiol. 1933;80(2):113–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Schmickl CN, Owens RL, Orr JE, Edwards BA, Malhotra A. Side effects of acetazolamide: a systematic review and meta-analysis assessing overall risk and dose dependence. BMJ Open Respir Res. 2020;7(1):e000557. 10.1136/bmjresp-2020-000557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ringelstein EB, Van Eyck S, Mertens I. Evaluation of Cerebral Vasomotor Reactivity by Various Vasodilating Stimuli: Comparison of CO2 to Acetazolamide. http://dxorg/101038/jcbfm199220. 2016;12(1):162–8. [DOI] [PubMed]
  • 7.Hauge A, Nicolaysen G, Thoresen M. Acute effects of acetazolamide on cerbral blood flow in man. Acta Physiol Scand. 1983;117(2):233–9. [DOI] [PubMed] [Google Scholar]
  • 8.Severinghaus JW, Cotev S. Carbonic Acidosis and Cerebral Vasodilation after Diamox. http://dxorg/103109/00365516809168962 . 2010;21(s102). [DOI] [PubMed]
  • 9.Sugrue MF. Pharmacological and ocular hypotensive properties of topical carbonic anhydrase inhibitors. Prog Retin Eye Res. 2000;19(1):87–112. [DOI] [PubMed] [Google Scholar]
  • 10.Sugrue MF. The preclinical pharmacology of dorzolamide hydrochloride, a topical carbonic anhydrase inhibitor. J Ocul Pharmacol Ther. 1996;12(3):363–76. [DOI] [PubMed] [Google Scholar]
  • 11.Friedenwald JS. Current studies on acetazoleamide (diamox) and aqueous humor flow. Am J Ophthalmol. 1955;40(5 Pt 2):139–47. [DOI] [PubMed] [Google Scholar]
  • 12.Becker B. Misuse of acetazolamide. Am J Ophthalmol. 1957;43(5):799–801. [Google Scholar]
  • 13.Heller I, Halevy J, Cohen S, Theodor E. Significant metabolic acidosis induced by acetazolamide. Not a rare complication. Arch Intern Med. 1985;145(10):1815–7. [PubMed] [Google Scholar]
  • 14.Bietti G, Virno M, Pecori-Giraldi J, Pellegrino N. Acetazolamide, metabolic acidosis, and intraocular pressure. Am J Ophthalmol. 1975;80(3):360–9. [DOI] [PubMed] [Google Scholar]
  • 15.To CH, Do CW, Zamudio AC, Candia OA. Model of ionic transport for bovine ciliary epithelium: effects of acetazolamide and HCO3. Am J Physiol-Cell Physiol. 2001;280(6):C1521–30. [DOI] [PubMed] [Google Scholar]
  • 16.Costa VP, Harris A, Stefánsson E, Flammer J, Krieglstein GK, Orzalesi N, et al. The effects of antiglaucoma and systemic medications on ocular blood flow. Prog Retin Eye Res. 2003;22(6):769–805. [DOI] [PubMed] [Google Scholar]
  • 17.Nork TM, McCormick SA, Chao GM, Odom JV. Distribution of carbonic anhydrase among human photoreceptors. Invest Ophthalmol Vis Sci. 1990;31(8):1451–8. [PubMed] [Google Scholar]
  • 18.Nigel Cox S, Hay E, Bird AC. Treatment of chronic macular edema with acetazolamide. Arch Ophthalmol. 1988;106(9):1190–5. [DOI] [PubMed] [Google Scholar]
  • 19.Dallinger S, Bobr B, Findl O, Eichler HG, Schmetterer L. Effects of acetazolamide on choroidal blood flow. Stroke. 1998;29(5):997–1001. [DOI] [PubMed] [Google Scholar]
  • 20.Steinsapir KD, Tripathi RC, Tripathi BJ, Ernest JT. Inhibition of ocular gamma glutamyl transpeptidase by acetazolamide. Exp Eye Res. 1992;55(1):179–81. [DOI] [PubMed] [Google Scholar]
  • 21.Thiry A, Dogne J-M, Supuran C, Masereel B. Carbonic anhydrase inhibitors as anticonvulsant agents. Curr Top Med Chem. 2007;7(9):855–64. [DOI] [PubMed] [Google Scholar]
  • 22.Supuran CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discovery. 2008;7(2):168–81. [DOI] [PubMed] [Google Scholar]
  • 23.Halmi P, Parkkila S, Honkaniemi J. Expression of carbonic anhydrases II, IV, VII, VIII and XII in rat brain after kainic acid induced status epilepticus. Neurochem Int. 2006;48(1):24–30. [DOI] [PubMed] [Google Scholar]
  • 24.Becker B. Decrease in intraocular pressure in man by a carbonic anhydrase inhibitor, diamox; a preliminary report. Am J Ophthalmol. 1954;37(1):13–5. [DOI] [PubMed] [Google Scholar]
  • 25.Weinstein P, Forgacs J. Data concerning the mechanism of the eye tension-lowering effect of diamox. Ophthalmologica. 1955;130(3):159–64. [DOI] [PubMed] [Google Scholar]
  • 26.Maus TL, Larsson L-I, McLaren JW, Brubaker RF. Comparison of dorzolamide and acetazolamide as suppressors of aqueous humor flow in humans. Arch Ophthalmol. 1997;115(1):45–9. [DOI] [PubMed] [Google Scholar]
  • 27.Krupin T, Sly WS, Whyte MP, Dodgson SJ. Failure of acetazolamide to decrease intraocular pressure in patients with carbonic anhydrase II deficiency. Am J Ophthalmol. 1985;99(4):396–9. [DOI] [PubMed] [Google Scholar]
  • 28.Tanimura Y, Hiroaki Y, Fujiyoshi Y. Acetazolamide reversibly inhibits water conduction by aquaporin-4. J Struct Biol. 2009;166(1):16–21. [DOI] [PubMed] [Google Scholar]
  • 29.Li J, Patil RV, Verkman AS. Mildly abnormal retinal function in transgenic mice without Müller cell aquaporin-4 water channels. Invest Ophthalmol Vis Sci. 2002;43(2):573–9. [PubMed] [Google Scholar]
  • 30.Marmor MF, Negi A. Pharmacologic modification of subretinal fluid absorption in the rabbit eye. Arch Ophthalmol. 1986;104(11):1674–7. [DOI] [PubMed] [Google Scholar]
  • 31.Moldow B, Sander B, Larsen M, Engler C, Li B, Rosenberg T, et al. The effect of acetazolamide on passive and active transport of fluorescein across the blood-retina barrier in retinitis pigmentosa complicated by macular oedema. Graefes Arch Clin Exp Ophthalmol. 1998;236(12):881–9. [DOI] [PubMed] [Google Scholar]
  • 32.Heuser D, Astrup J, Lassen NA, Betz BE. Brain carbonic acid acidosis after acetazolamide. Acta Physiol Scand. 1975;93(3):385–90. [DOI] [PubMed] [Google Scholar]
  • 33.Ridderstråle Y, Hanson M. Histochemical study of the distribution of carbonic anhydrase in the cat brain. Acta Physiol Scand. 1985;124(4):557–64. [DOI] [PubMed] [Google Scholar]
  • 34.Mauger TF, Craig EL, Havener WH. Havener’s ocular pharmacology. 6th ed. St. Louis: Mosby; 1994. [Google Scholar]
  • 35.Stone RA, Zimmerman TJ, Shin DH, Becker B, Kass MA. Low-dose methazolamide and intraocular pressure. Am J Ophthalmol. 1977;83(5):674–9. [DOI] [PubMed] [Google Scholar]
  • 36.Berson FG, Epstein DL, Grant WM, Hutchinson BT, Dobbs PC. Acetazolamide dosage forms in the treatment of glaucoma. Arch Ophthalmol. 1980;98(6):1051–4. [DOI] [PubMed] [Google Scholar]
  • 37.Friedland BR, Mallonee J, Anderson DR. Short-term dose response characteristics of acetazolamide in man. Arch Ophthalmol. 1977;95(10):1809–12. [DOI] [PubMed] [Google Scholar]
  • 38.Maren TH, Haywood JR, Chapman SK, Zimmerman TJ. The pharmacology of methazolamide in relation to the treatment of glaucoma. Invest Ophthalmol Vis Sci. 1977;16(8):730–42. [PubMed] [Google Scholar]
  • 39.Netland PA, Tanna AP, editors. Glaucoma medical therapy. 3rd ed. Amsterdam, The Netherlands: Kugler Publications; 2020.
  • 40.Kishida K, Akaki Y, Sasabe T, Yamamoto C, Manabe R. Glutathione conjugation of methazolamide and subsequent reactions in the ciliary body in vitro. J Pharm Sci. 1990;79(7):638–42. [DOI] [PubMed] [Google Scholar]
  • 41.Kishida K, Saida N, Yamamura N, Iwai Y, Sasabe T. Cysteine conjugate of methazolamide is metabolized by beta-lyase. J Pharm Sci. 2001;90(2):224–33. [DOI] [PubMed] [Google Scholar]
  • 42.Konstantopoulos N, Molero JC, McGee SL, Spolding B, Connor T, de Vries M, et al. Methazolamide is a new hepatic insulin sensitizer that lowers blood glucose in vivo. Diabetes. 2012;61(8):2146–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Häussinger D, Kaiser S, Stehle T, Gerok W. Liver carbonic anhydrase and urea synthesis. Effect Diuretics Biochem Pharmacol. 1986;35(19):3317–22. [DOI] [PubMed] [Google Scholar]
  • 44.Krivoy N, Ben-Arieh Y, Carter A, Alroy G. Methazolamide-induced hepatitis and pure RBC aplasia. Arch Intern Med. 1981;141(9):1229–30. [PubMed] [Google Scholar]
  • 45.Stamper RL, Lieberman MF, Drake MV. CHAPTER 26 - Carbonic anhydrase inhibitors. In: Stamper RL, Lieberman MF, Drake MV, editors. Becker-Shaffer’s Diagnosis and Therapy of the Glaucomas. 8th ed. Edinburgh: Mosby; 2009. p. 407–19. [Google Scholar]
  • 46.Campbell DA. Effect of neptazane on intra-ocular pressure in relation to its systemic action and its clinical application. Br J Ophthalmol. 1960;44(7):415–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Dahlen K, Epstein DL, Grant WM, Hutchinson BT, Prien EL, Krall JM. A repeated dose-response study of methazolamide in glaucoma. Arch Ophthalmol. 1978;96(12):2214–8. [DOI] [PubMed] [Google Scholar]
  • 48.Maren TH. Carbonic anhydrase: chemistry, physiology, and inhibition. Physiol Rev. 1967;47(4):595–781. [DOI] [PubMed] [Google Scholar]
  • 49.Eller MG, Schoenwald RD. Determination of ethoxzolamide in the iris/ciliary body of the rabbit eye by high-performance liquid chromatography: comparison of tissue levels following intravenous and topical administrations. J Pharm Sci. 1984;73(9):1261–4. [DOI] [PubMed] [Google Scholar]
  • 50.Bartlett JD, Jaanus SD. Clinical ocular pharmacology. 5th ed. St. Louis, Mo: Butterworth-Heinemann/Elsevier; 2008. Print.
  • 51.Bleckmann H. Dichlorphenamide treatment in acute glaucoma (author’s transl). Albrecht Von Graefes Arch Klin Exp Ophthalmol. 1976;201(1):69–77. [DOI] [PubMed] [Google Scholar]
  • 52.Schimek RA, Balian JV, Lepley FJ, Ottum JA. Evaluation of dichlorphenamide as an ocular hypotensive agent. AMA Arch Ophthalmol. 1958;60(6):1053–60. [DOI] [PubMed] [Google Scholar]
  • 53.Gonzales-Jimenez E, Leopold IH. Effect of dichlorphenamide on the intraocular pressure of humans. AMA Arch Ophthalmol. 1958;60(3):427–36. [DOI] [PubMed] [Google Scholar]
  • 54.Cohen F. Open-label, dose-escalation, phase 1 study of safety and single and multiple-dose pharmacokinetics of dichlorphenamide in healthy volunteers. Clin Pharmacol Drug Dev. 2019;8(1):87–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.U.S. Food and Drug Administration. Highlights of prescribing information: KEVEYIS (dichlorphenamide) tablets, for oral use. Silver Spring, MD: FDA. 2019. Retrieved from https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/011366s033lbl.pdf.
  • 56.Ciafaloni E, Cohen F, Griggs R. Efficacy and safety of dichlorphenamide for primary periodic paralysis in adolescents compared with adults. Pediatr Neurol. 2019;101:43–6. [DOI] [PubMed] [Google Scholar]
  • 57.Albert DM, Miller JW, Azar D T, Young LH, editors. Albert and Jakobiec’s principles and practice of ophthalmology. Springer Cham. 2020. 10.1007/978-3-319-90495-5.
  • 58.Lysloff Skipski IA, Scott WN. Age-related changes in carbonic anhydrase of toad lens. Exp Eye Res. 1985;41(6):671–85. [DOI] [PubMed] [Google Scholar]
  • 59.deLuise VP, Anderson DR. Primary infantile glaucoma (congenital glaucoma). Surv Ophthalmol. 1983;28(1):1–19. [DOI] [PubMed] [Google Scholar]
  • 60.Beck AD. Diagnosis and management of pediatric glaucoma. Ophthalmol Clin North Am. 2001;14(3):501–12. [DOI] [PubMed] [Google Scholar]
  • 61.Sharan S, Dupuis A, Hébert D, Levin AV. The effect of oral acetazolamide on weight gain in children. Can J Ophthalmol. 2010;45(1):41–5. [DOI] [PubMed] [Google Scholar]
  • 62.Samant M, Medsinge A, Nischal KK. Pediatric glaucoma: pharmacotherapeutic options. Pediatr Drugs. 2016;18(3):209–19. [DOI] [PubMed] [Google Scholar]
  • 63.Lowe RF. Acute angle closure glaucoma: acetazolamide therapy. Aust J Opthalmol. 1973;1(1):24–6. [DOI] [PubMed] [Google Scholar]
  • 64.Hoh ST, Aung T, Chew PTK. Medical management of angle closure glaucoma. http://dxorg/101076/soph1727914717 . 2009;17(2):79–83. [DOI] [PubMed]
  • 65.Prum BE, Rosenberg LF, Gedde SJ, Mansberger SL, Stein JD, Moroi SE, et al. Primary open-angle glaucoma preferred practice pattern® guidelines. Ophthalmology. 2016;123(1):P41–111. [DOI] [PubMed] [Google Scholar]
  • 66.Sabri K, Levin AV. The additive effect of topical dorzolamide and systemic acetazolamide in pediatric glaucoma. J Am Assoc Pediatric Ophthalmol Strabismus. 2006;10(5):464–8. [DOI] [PubMed] [Google Scholar]
  • 67.Kaur IP, Smitha R, Aggarwal D, Kapil M. Acetazolamide: future perspective in topical glaucoma therapeutics. Int J Pharm. 2002;248(1–2):1–14. [DOI] [PubMed] [Google Scholar]
  • 68.Johnson KK, Green DL, Rife JP, Limon L. Correction: sulfonamide cross-reactivity: fact or fiction? http://dxorg/101345/aph1E350c . 2005;39(7–8):1373-. [DOI] [PubMed]
  • 69.Henry RP. Multiple roles of carbonic anhydrase in cellular transport and metabolism. Annu Rev Physiol. 1996;58:523–38. [DOI] [PubMed] [Google Scholar]
  • 70.Lichter PR, Musch DC, Medzihradsky F, Standardi CL. Intraocular pressure effects of carbonic anhydrase inhibitors in primary open-angle glaucoma. Am J Ophthalmol. 1989;107(1):11–7. [DOI] [PubMed] [Google Scholar]
  • 71.Kesav N, Palestine AG, Kahook MY, Pantcheva MB. Current management of uveitis-associated ocular hypertension and glaucoma. Surv Ophthalmol. 2020;65(4):397–407. [DOI] [PubMed] [Google Scholar]
  • 72.Ritland JS, Egge K, Lydersen S, Juul R, Semb SO. Comparison of survival of exfoliative glaucoma patients and primary open-angle glaucoma patients: impact of acetazolamide use. Acta Ophthalmol Scand. 2004;82(4):397–400. [DOI] [PubMed] [Google Scholar]
  • 73.Murray CD, Wood D, Allgar V, Walters G, Gale RP. Short-term intraocular pressure trends following intravitreal ranibizumab injections for neovascular age-related macular degeneration—the role of oral acetazolamide in protecting glaucoma patients. Eye. 2014;28(10):1218–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Katayama BYNY, Bonini-Filho MA, Messias AMV, Paula JS, Martin LFT, Costa R, et al. Comparison of acetazolamide, brimonidine, and anterior chamber paracentesis for ocular hypertension control after initial intravitreal bevacizumab injection: a randomized clinical trial. J Glaucoma. 2014;23(7):461–3. [DOI] [PubMed] [Google Scholar]
  • 75.Hayashi K, Yoshida M, Manabe SI, Yoshimura K. Prophylactic effect of oral acetazolamide against intraocular pressure elevation after cataract surgery in eyes with glaucoma. Ophthalmology. 2017;124(5):701–8. [DOI] [PubMed] [Google Scholar]
  • 76.Al-Barrag A, Al-Shaer M, Al-Matary N, Bamashmous M. Oral versus topical carbonic anhydrase inhibitors in ocular hypertension after scleral tunnel cataract surgery. Clin Ophthalmol. 2009;3:357–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Edmunds B, Canning CR. The effect of prophylactic acetazolamide in patients undergoing extensive retinal detachment repair. Eye. 1996;10(3):328–30. [DOI] [PubMed] [Google Scholar]
  • 78.Broadway DC, Grierson I, Sturmer J, Hitchings RA. Reversal of topical antiglaucoma medication effects on the conjunctiva. Arch Ophthalmol. 1996;114(3):262–7. [DOI] [PubMed] [Google Scholar]
  • 79.Breusegem C, Spielberg L, Van Ginderdeuren R, Vandewalle E, Renier C, Van de Veire S, et al. Preoperative nonsteroidal anti-inflammatory drug or steroid and outcomes after trabeculectomy: a randomized controlled trial. Ophthalmology. 2010;117(7):1324–30. [DOI] [PubMed] [Google Scholar]
  • 80.Baudouin C, Pisella PJ, Fillacier K, Goldschild M, Becquet F, De Saint JM, et al. Ocular surface inflammatory changes induced by topical antiglaucoma drugs: human and animal studies. Ophthalmology. 1999;106(3):556–63. [DOI] [PubMed] [Google Scholar]
  • 81.Sherwood MB, Grierson I, Millar L, Hitchings RA. Long-term morphologic effects of antiglaucoma drugs on the conjunctiva and Tenon’s capsule in glaucomatous patients. Ophthalmology. 1989;96(3):327–35. [DOI] [PubMed] [Google Scholar]
  • 82.Lorenz K, Wasielica-Poslednik J, Bell K, Renieri G, Keicher A, Ruckes C, et al. Efficacy and safety of preoperative IOP reduction using a preservative-free fixed combination of dorzolamide/timolol eye drops versus oral acetazolamide and dexamethasone eye drops and assessment of the clinical outcome of trabeculectomy in glaucoma. PLoS ONE. 2017;12(2):e0171636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Xie JS, Donaldson L, Margolin E. Papilledema: a review of etiology, pathophysiology, diagnosis, and management. Surv Ophthalmol. 2022;67(4):1135–59. [DOI] [PubMed] [Google Scholar]
  • 84.Baneke AJ, Aubry J, Viswanathan AC, Plant GT. The role of intracranial pressure in glaucoma and therapeutic implications. Eye. 2019;34(1):178–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.McCulley TJ, Chang JR, Piluek WJ. Intracranial pressure and glaucoma. J Neuroophthalmol. 2015;35:S38–44. [DOI] [PubMed] [Google Scholar]
  • 86.Loiselle AR, de Kleine E, van Dijk P, Jansonius NM. Intraocular and intracranial pressure in glaucoma patients taking acetazolamide. PloS One. 2020;15(6):e0234690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wolfensberger TJ. The role of carbonic anhydrase inhibitors in the management of macular edema. Doc Ophthalmol. 1999;97(3–4):387–97. [DOI] [PubMed] [Google Scholar]
  • 88.Koronis S, Stavrakas P, Balidis M, Kozeis N, Tranos PG. Update in treatment of uveitic macular edema. Drug Des Dev Ther. 2019;13:667–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Whitcup SM, Csaky KG, Podgor MJ, Chew EY, Perry CH, Nussenblatt RB, et al. A randomized, masked, crossover trial of acetazolamide for cystoid macular edema in patients with uveitis. Ophthalmology. 1996;103(7):1054–63. [DOI] [PubMed] [Google Scholar]
  • 90.Fishman GA, Gilbert LD, Fiscella RG, Kimura AE, Jampol LM. Acetazolamide for treatment of chronic macular edema in retinitis pigmentosa. Arch Ophthalmol. 1989;107(10):1445–52. [DOI] [PubMed] [Google Scholar]
  • 91.Fishman GA, Gilbert LD, Anderson RJ, Marmor MF, Weleber RG, Viana MAG. Effect of methazolamide on chronic macular edema in patients with retinitis pigmentosa. Ophthalmology. 1994;101(4):687–93. [DOI] [PubMed] [Google Scholar]
  • 92.Yeo JH, Min CH, Yoon YH. Effect of oral carbonic anhydrase inhibitor on cystoid macular edema associated with retinitis pigmentosa: an OCT and OCT angiography study. Retina. 2022;42(9):1796–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Chen C, Liu X, Peng X. Management of cystoid macular edema in retinitis pigmentosa: a systematic review and meta-analysis. Front Med. 2022;9:1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pikkel J, Beiran I, Ophir A, Miller B. Acetazolamide for central serous retinopathy. Ophthalmology. 2002;109(9):1723–5. [DOI] [PubMed] [Google Scholar]
  • 95.Rübsam A, Thieme CE, Schlomberg J, Winterhalter S, Müller B, Joussen AM, et al. Therapy rationale for mineralocorticoid-receptor antagonists, acetazolamide and a switch of therapy in nonresponders in central serous chorioretinopathy. J Ocul Pharmacol Ther. 2017;33(3):141–8. [DOI] [PubMed] [Google Scholar]
  • 96.Kinouchi R, Okumichi H, Sakata H, Ohara H, Hirooka K, Kiuchi Y. Effect of acetazolamide for exudative retinal detachment in sturge-weber syndrome. Med: Case Rep Study Protocols. 2021;2(9):e0129. [Google Scholar]
  • 97.Prakash P, De Salvo G, Lotery AJ. Morning glory with serous macular detachment responds to oral acetazolamide. Eye. 2010;24(11):1732–3. [DOI] [PubMed] [Google Scholar]
  • 98.Gíslason I, Jónasson F, Stefánsson E. Exudative retinal detachment in familial pulmonary hypertension. Acta Ophthalmol. 1991;69(6):805–9. [DOI] [PubMed] [Google Scholar]
  • 99.Osigian CJ, Gologorsky D, Cavuoto KM, Berrocal A, Villegas V. Oral acetazolamide as a medical adjuvant to retinal surgery in optic disc pit maculopathy in a pediatric patient. Am J Ophthalmol Case Rep. 2020;17:100599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.McCarthy KD, Reed DJ. The effect of acetazolamide and furosemide on cerebrospinal fluid production and choroid plexus carbonic anhydrase activity. J Pharmacol Exp Ther. 1974;189(1):194–201. [PubMed] [Google Scholar]
  • 101.Robert LT, Arysol SN, Bernd FR. Treatment of Pseudotumor Cerebri with Diamox (Acetazolamide) | Eccles Health Sciences Library | J. Willard Marriott Digital Library. Journal of Clinical Neuro-ophthalmology. 1988;8(2):93–8.
  • 102.Johnson LN, Krohel GB, Madsen RW, March GA. The role of weight loss and acetazolamide in the treatment of idiopathic intracranial hypertension (pseudotumor cerebri). Ophthalmology. 1998;105(12):2313–7. [DOI] [PubMed] [Google Scholar]
  • 103.Wall M, McDermott MP, Kieburtz KD, Corbett JJ, Feldon SE, Friedman DI, et al. Effect of acetazolamide on visual function in patients with idiopathic intracranial hypertension and mild visual loss: the idiopathic intracranial hypertension treatment trial. JAMA. 2014;311(16):1641–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Horton JC. Acetazolamide for pseudotumor cerebri: evidence from the NORDIC trial. JAMA. 2014;311(16):1618–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Çelebisoy N, Gökçay F, Şirin H, Akyürekli Ö. Treatment of idiopathic intracranial hypertension: topiramate vs acetazolamide, an open-label study. Acta Neurol Scand. 2007;116(5):322–7. [DOI] [PubMed] [Google Scholar]
  • 106.Kesserwani H. Isolated sixth nerve palsy: a case of pseudotumor cerebri and an overview of the evolutionary dynamic geometry of dorello’s canal. Cureus. 2021;13(5):e15340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Meshref M, Shaheen N, Swed S, Ibrahim M, Mostafa MR, Algazar MO, et al. An overview of third, fourth and sixth cranial nerve palsies in the setting of COVID-19: a case report and systematic review. Medicine. 2022;101(49):e32023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Samara A, Ghazaleh D, Berry B, Ghannam M. Idiopathic intracranial hypertension presenting with isolated unilateral facial nerve palsy: a case report. J Med Case Reports. 2019;13(1):94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Galin MA, Baras I, Zweifach P. Diamox-induced myopia. Am J Ophthalmol. 1962;54:237–40. [PubMed] [Google Scholar]
  • 110.Widengård I, Mandahl A, Törnquist P, Wistrand PJ. Colour vision and side-effects during treatment with methazolamide. Eye (Lond). 1995;9(Pt 1):130–5. [DOI] [PubMed] [Google Scholar]
  • 111.Leys MJ, van Slycken S, Nork TM, Odom JV. Acetazolamide affects performance on the Nagel II anomaloscope. Graefes Arch Clin Exp Ophthalmol. 1996;234(Suppl 1):S193–7. [DOI] [PubMed] [Google Scholar]
  • 112.Krieg PH, Schipper I. Drug-induced ciliary body oedema: a new theory. Eye (Lond). 1996;10(Pt 1):121–6. [DOI] [PubMed] [Google Scholar]
  • 113.Hill AD. Myopic changes in a climber after taking acetazolamide and the use of corrective lenses to temporize symptoms: a case report from mount Kilimanjaro. Wilderness Environ Med. 2016;27(3):397–400. [DOI] [PubMed] [Google Scholar]
  • 114.Parkkila S, Parkkila AK, Juvonen T, Rajaniemi H. Distribution of the carbonic anhydrase isoenzymes I, II, and VI in the human alimentary tract. Gut. 1994;35(5):646–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Swenson ER. Safety of carbonic anhydrase inhibitors. Expert Opin Drug Saf. 2014;13(4):459–72. [DOI] [PubMed] [Google Scholar]
  • 116.Epstein DL, Grant WM. Carbonic anhydrase inhibitor side effects. Serum Chem Anal Arch Ophthalmol. 1977;95(8):1378–82. [DOI] [PubMed] [Google Scholar]
  • 117.Arrigg CA, Epstein DL, Giovanoni R, Grant WM. The influence of supplemental sodium acetate on carbonic anhydrase inhibitor-induced side effects. Arch Ophthalmol. 1981;99(11):1969–72. [DOI] [PubMed] [Google Scholar]
  • 118.Schwartz GJ. Physiology and molecular biology of renal carbonic anhydrase. J Nephrol. 2002;15(Suppl 5):S61-74. [PubMed] [Google Scholar]
  • 119.Chapron DJ, Gomolin IH, Sweeney KR. Acetazolamide blood concentrations are excessive in the elderly: propensity for acidosis and relationship to renal function. J Clin Pharmacol. 1989;29(4):348–53. [DOI] [PubMed] [Google Scholar]
  • 120.Ahlstrand C, Tiselius HG. Urine composition and stone formation during treatment with acetazolamide. Scand J Urol Nephrol. 1987;21(3):225–8. [DOI] [PubMed] [Google Scholar]
  • 121.Kass MA, Kolker AE, Gordon M, Goldberg I, Gieser DK, Krupin T, et al. Acetazolamide and urolithiasis. Ophthalmology. 1981;88(3):261–5. [DOI] [PubMed] [Google Scholar]
  • 122.Au JN, Waslo CS, McGwin G, Huisingh C, Tanne E. Acetazolamide-induced nephrolithiasis in idiopathic intracranial hypertension patients. J Neuroophthalmol. 2016;36(2):126–30. [DOI] [PubMed] [Google Scholar]
  • 123.Kodjikian L, Durand B, Burillon C, Rouberol F, Grange JD, Renaudier P. Acetazolamide-induced thrombocytopenia. Arch Ophthalmol. 2004;122(10):1543–4. [DOI] [PubMed] [Google Scholar]
  • 124.Fraunfelder FT, Meyer SM, Bagby GC Jr, Dreis MW. Hematologic reactions to carbonic anhydrase inhibitors. Am J Ophthalmol. 1985;100(1):79–81. [DOI] [PubMed] [Google Scholar]
  • 125.Maclean R, O’Callaghan U, Lim SH. Acetazolamide-induced severe pancytopenia mimicking myelodysplasia relapse following allogeneic bone marrow transplantation. Bone Marrow Transplant. 1998;21(3):309–11. [DOI] [PubMed] [Google Scholar]
  • 126.Mogk LG, Cyrlin MN. Blood dyscrasias and carbonic anhydrase inhibitors. Ophthalmology. 1988;95(6):768–71. [DOI] [PubMed] [Google Scholar]
  • 127.Zimran A, Beutler E. Can the risk of acetazolamide-induced aplastic anemia be decreased by periodic monitoring of blood cell counts? Am J Ophthalmol. 1987;104(6):654–8. [DOI] [PubMed] [Google Scholar]
  • 128.Keisu M, Wiholm BE, Ost A, Mortimer O. Acetazolamide-associated aplastic anaemia. J Intern Med. 1990;228(6):627–32. [DOI] [PubMed] [Google Scholar]
  • 129.Aronson JK, editor. Meyler's side effects of drugs : the international encyclopedia of adverse drug reactions and interactions. 16th ed. Amsterdam: Elsevier Science; 2015.
  • 130.Hsu DY, Brieva J, Silverberg NB, Silverberg JI. Morbidity and mortality of stevens-johnson syndrome and toxic epidermal necrolysis in united states adults. J Investig Dermatol. 2016;136(7):1387–97. [DOI] [PubMed] [Google Scholar]
  • 131.Kim S, Yun J, Kang DY, Park HJ, Jo EJ, Jung JW, et al. Carbonic anhydrase inhibitor-induced Stevens-Johnson syndrome/toxic epidermal necrolysis leads to extensive cutaneous involvement. J Allergy Clin Immunol Pract. 2019;7(8):2851-3.e2. [DOI] [PubMed] [Google Scholar]
  • 132.Her Y, Kil MS, Park JH, Kim CW, Kim SS. Stevens-Johnson syndrome induced by acetazolamide. J Dermatol. 2011;38(3):272–5. [DOI] [PubMed] [Google Scholar]
  • 133.Kim SH, Kim M, Lee KW, Kang HR, Park HW, Jee YK. HLA-B*5901 is strongly associated with methazolamide-induced Stevens-Johnson syndrome/toxic epidermal necrolysis. Pharmacogenomics. 2010;11(6):879–84. [DOI] [PubMed] [Google Scholar]
  • 134.Shu C, Shu D, Tie D, Yu M, Zhang R, Wang T, et al. Toxic epidermal necrolysis induced by methazolamide in a Chinese-Korean man carrying HLA-B*59:01. Int J Dermatol. 2015;54(11):1242–5. [DOI] [PubMed] [Google Scholar]
  • 135.Flach AJ, Smith RE, Fraunfelder FT. Stevens-Johnson syndrome associated with methazolamide treatment reported in two Japanese-American women. Ophthalmology. 1995;102(11):1677–80. [DOI] [PubMed] [Google Scholar]
  • 136.Jiang M, Yang F, Zhang L, Xu D, Jia Y, Cheng Y, et al. Unique motif shared by HLA-B*59:01 and HLA-B*55:02 is associated with methazolamide-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in Han Chinese. J Eur Acad Dermatol Venereol. 2022;36(6):873–80. [DOI] [PubMed] [Google Scholar]
  • 137.Tangamornsuksan W, Lohitnavy M. Association between HLA-B*5901 and methazolamide-induced Stevens-Johnson syndrome/toxic epidermal necrolysis: a systematic review and meta-analysis. Pharmacogenomics J. 2019;19(3):286–94. [DOI] [PubMed] [Google Scholar]
  • 138.Yang F, Xuan J, Chen J, Zhong H, Luo H, Zhou P, et al. HLA-B*59:01: a marker for Stevens-Johnson syndrome/toxic epidermal necrolysis caused by methazolamide in Han Chinese. Pharmacogenomics J. 2016;16(1):83–7. [DOI] [PubMed] [Google Scholar]
  • 139.Gandham SB, Spaeth GL, Di Leonardo M, Costa VP. Methazolamide-induced skin eruptions. Arch Ophthalmol. 1993;111(3):370–2. [DOI] [PubMed] [Google Scholar]
  • 140.Jachiet M, Bellon N, Assier H, Amsler E, Gaouar H, Pecquet C, et al. Cutaneous adverse drug reaction to oral acetazolamide and skin tests. Dermatology. 2013;226(4):347–52. [DOI] [PubMed] [Google Scholar]
  • 141.Lemon N, Canepa E, Ilies MA, Fossati S. Carbonic anhydrases as potential targets against neurovascular unit dysfunction in Alzheimer’s disease and stroke. Front Aging Neurosci. 2021;13:772278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Becker B, Middleton WH. Long-term acetazoleamide (diamox) administration in therapy of glaucomas. AMA Arch Ophthalmol. 1955;54(2):187–92. [DOI] [PubMed] [Google Scholar]
  • 143.Cyr M, Laizure SC, daCunha CM. Methazolamide-induced delirium. Pharmacotherapy. 1997;17(2):387–9. [PubMed] [Google Scholar]
  • 144.Adamson R, Swenson ER. Acetazolamide use in severe chronic obstructive pulmonary disease pros and cons. Ann Am Thorac Soc. 2017;14(7):1086–93. [DOI] [PubMed] [Google Scholar]
  • 145.American Academy of Ophthalmology EyeNet Magazine. Acetazolamide: Considerations for Systemic Administration: AAO; 2015 [Available from: https://www.aao.org/eyenet/article/acetazolamide-considerations-systemic-administrati.
  • 146.Basar O, Basar N, Tuna Y, Yuksel H, Coban S. Acetazolamide induced severe hepatotoxicity. Wien Klin Wochenschr. 2013;125(7–8):223–4. [DOI] [PubMed] [Google Scholar]
  • 147.Kristinsson A. Fatal reaction to acetazolamide. Br J Ophthalmol. 1967;51(5):348–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Margo CE. Acetazolamide and advanced liver disease. Am J Ophthalmol. 1986;101(5):611–2. [DOI] [PubMed] [Google Scholar]
  • 149.National Institute of Diabetes and Digestive and Kidney Diseases. LiverTox: clinical and research information on drug‑induced liver injury [Online database]. Bethesda, MD: U.S. Department of Health and Human Services, National Institutes of Health. 2012. Retrieved June 18, 2025, from https://www.ncbi.nlm.nih.gov/books/NBK547852/. [PubMed]
  • 150.Kerber JM, de Mello JD, Moura KBA, da Silva GC, Wawrzeniak IC, Rech TH. Acetazolamide intoxication in an elderly patient with diabetes and chronic renal failure after cataract surgery. Case Rep Crit Care. 2020;2020:3764972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Schwenk MH, St Peter WL, Meese MG, Singhal PC. Acetazolamide toxicity and pharmacokinetics in patients receiving hemodialysis. Pharmacotherapy. 1995;15(4):522–7. [PubMed] [Google Scholar]
  • 152.Schwenk MH, Blaustein DA, Wagner JD. The pharmacokinetics of acetazolamide during CAPD. Adv Perit Dial. 1994;10:44–6. [PubMed] [Google Scholar]
  • 153.Izumi S, Hirata S, Koizumi M, Tanaka K, Ueno K, Kim M. 132 Pharmacokinetics of acetazolamide in patients with uremia. Ther Drug Monit. 1997;19(5):580. [Google Scholar]
  • 154.Goldberg MF. Sickled erythrocytes, hyphema, and secondary glaucoma: V. The effect of vitamin C on erythrocyte sickling in aqueous humor. Ophthalmic Surg. 1979;10(4):70–7. [PubMed] [Google Scholar]
  • 155.Meadows M. Pregnancy and the drug dilemma. FDA Consum. 2001;35(3):16–20. [PubMed] [Google Scholar]
  • 156.Tellone CI, Baldwin JK, Sofia RD. Teratogenic activity in the mouse after oral administration of acetazolamide. Drug Chem Toxicol. 1980;3(1):83–98. [DOI] [PubMed] [Google Scholar]
  • 157.Scott WJ Jr, Lane PD, Randall JL, Schreiner CM. Malformations in nonlimb structures induced by acetazolamide and other inhibitors of carbonic anhydrase. Ann N Y Acad Sci. 1984;429:447–56. [DOI] [PubMed] [Google Scholar]
  • 158.Biddle FG. Genetic differences in the frequency of acetazolamide-induced ectrodactyly in the mouse exhibit directional dominance of relative embryonic resistance. Teratology. 1988;37(4):375–88. [DOI] [PubMed] [Google Scholar]
  • 159.Kojima N, Naya M, Makita T. Effects of maternal acetazolamide treatment on body weights and incisor development of the fetal rat. J Vet Med Sci. 1999;61(2):143–7. [DOI] [PubMed] [Google Scholar]
  • 160.Lee AG, Pless M, Falardeau J, Capozzoli T, Wall M, Kardon RH. The use of acetazolamide in idiopathic intracranial hypertension during pregnancy. Am J Ophthalmol. 2005;139(5):855–9. [DOI] [PubMed] [Google Scholar]
  • 161.Falardeau J, Lobb BM, Golden S, Maxfield SD, Tanne E. The use of acetazolamide during pregnancy in intracranial hypertension patients. J Neuroophthalmol. 2013;33(1):9–12. [DOI] [PubMed] [Google Scholar]
  • 162.Klebanoff MA. The collaborative perinatal project: a 50-year retrospective. Paediatr Perinat Epidemiol. 2009;23(1):2–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Worsham F Jr, Beckman EN, Mitchell EH. Sacrococcygeal teratoma in a neonate. Association with maternal use of acetazolamide. JAMA. 1978;240(3):251–2. [PubMed] [Google Scholar]
  • 164.Al-Saleem AI, Al-Jobair AM. Possible association between acetazolamide administration during pregnancy and multiple congenital malformations. Drug Des Devel Ther. 2016;10:1471–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Merlob P, Litwin A, Mor N. Possible association between acetazolamide administration during pregnancy and metabolic disorders in the newborn. Eur J Obstet Gynecol Reprod Biol. 1990;35(1):85–8. [DOI] [PubMed] [Google Scholar]
  • 166.Mansour M, Shukla S, Cortez J. Neonatal metabolic acidosis after intrauterine exposure to acetazolamide. Glob J Ped Neonatol Car. 2020;3(1)
  • 167.Drugs.com. Acetazolamide Pregnancy and Breastfeeding Warnings: Drugs.com; 2022 [Available from: https://www.drugs.com/pregnancy/acetazolamide.html.
  • 168.Belkin A, Chen T, DeOliveria AR, Johnson SM, Ramulu PY, Buys YM, et al. A practical guide to the pregnant and breastfeeding patient with glaucoma. Ophthalmol Glaucoma. 2020;3(2):79–89. [DOI] [PubMed] [Google Scholar]
  • 169.Blumen-Ohana E, Sellem E. Pregnancy & glaucoma: SFO-SFG recommendations. J Fr Ophtalmol. 2020;43(1):63–6. [DOI] [PubMed] [Google Scholar]
  • 170.American Academy of Pediatrics Committee on Drugs. Transfer of drugs and other chemicals into human milk. Pediatrics. 2001;108(3):776–89. 10.1542/peds.108.3.776. [DOI] [PubMed] [Google Scholar]
  • 171.Seifarth C, Schehler B, Schneider HJ. Effectiveness of metformin on weight loss in non-diabetic individuals with obesity. Exp Clin Endocrinol Diabetes. 2013;121(1):27–31. [DOI] [PubMed] [Google Scholar]
  • 172.Pernicova I, Korbonits M. Metformin–mode of action and clinical implications for diabetes and cancer. Nat Rev Endocrinol. 2014;10(3):143–56. [DOI] [PubMed] [Google Scholar]
  • 173.Ismail IS. The role of carbonic anhydrase in hepatic glucose production. Curr Diabetes Rev. 2018;14(2):108–12. [DOI] [PubMed] [Google Scholar]
  • 174.Sweeney KR, Chapron DJ, Brandt JL, Gomolin IH, Feig PU, Kramer PA. Toxic interaction between acetazolamide and salicylate: case reports and a pharmacokinetic explanation. Clin Pharmacol Ther. 1986;40(5):518–24. [DOI] [PubMed] [Google Scholar]
  • 175.Chen TE, Liu SY. Severe metabolic acidosis and hyperammonemia induced by the concomitant use of acetazolamide and aspirin in a patient with impaired renal function. J Emerg Med. 2021;60(5):e115–7. [DOI] [PubMed] [Google Scholar]
  • 176.Shukralla AA, Dolan E, Delanty N. Acetazolamide: old drug, new evidence? Epilepsia Open. 2022;7(3):378–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Cambridge University Press. The Epilepsy Prescriber’s Guide to Antiepileptic Drugs: Cambridge; 2010 [Available from: https://assets.cambridge.org/97805216/87164/frontmatter/9780521687164_frontmatter.pdf.
  • 178.Aslam S, Gupta V. Carbonic Anhydrase Inhibitors. [Updated 2023 Apr 17]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025. Available from: https://www.ncbi.nlm.nih.gov/books/NBK557736/. [PubMed]
  • 179.Nesher R, Ticho U. Switching from systemic to the topical carbonic anhydrase inhibitor dorzolamide: effect on the quality of life of glaucoma patients with drug-related side effects. Isr Med Assoc J. 2003;5(4):260–3. [PubMed] [Google Scholar]
  • 180.Mills T, Law SK, Walt J, Buchholz P, Hansen J. Quality of life in glaucoma and three other chronic diseases: a systematic literature review. Drugs Aging. 2009;26(11):933–50. [DOI] [PubMed] [Google Scholar]
  • 181.Popovic MM, Schlenker MB, Thiruchelvam D, Redelmeier DA. Serious adverse events of oral and topical carbonic anhydrase inhibitors. JAMA Ophthalmol. 2022;140(3):235–42. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No datasets were generated or analysed during the current study.


Articles from Current Ophthalmology Reports are provided here courtesy of Springer

RESOURCES