Skip to main content

Advertisement

Log in

The Neurological Spectrum for Acetazolamide Pharmacotherapy: from Basic Science to Clinical Applications

  • Review
  • Published:
SN Comprehensive Clinical Medicine Aims and scope Submit manuscript

Abstract

Acetazolamide was developed nearly 70 years ago and continues to be used for niche indications. Mechanisms of action are relatively less well-studied than clinical applications. Several potential uses span the spectrum of neurological illnesses, but yet neurological side effects complicate short- and long-term applications for many patients. This review examines the basic science and clinical usages of acetazolamide germane to neurological and psychiatric spectrums. The existing publications were reviewed from the time of the drug’s inception to April 2021. This review utilized access as available in PubMed, EMBASE, CINAHL Plus, and the Cochrane Library. Acetazolamide is firmly established as first-line prevention for altitude sickness. Outside of declining use for glaucoma, most other medical indications are few, highly selective, and/or partially beneficial. In the past, carbonic anhydrase inhibition has been the proposed mainstay of acetazolamide’s mode of action, but more contemporary studies have focused on cellular membrane ion/water channel activity. Prolonged use of acetazolamide is nevertheless likely to be limited due to common adverse events unless more effective mitigating approaches are discovered. A better understanding of acetazolamide’s clinical impact and molecular mechanisms is sure to elaborate on both healthy physiological mechanisms and disease.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

Availability of data and material

Not required.

Code availability

Not applicable.

References

  1. Roblin RO, Clapp JW. The preparation of heterocyclic sulfonamides. J Amer Chem Soc. 1950;72:4890–2.

    CAS  Google Scholar 

  2. Friedland BR, Mallonee J, Anderson DR. Short-term dose response characteristics of acetazolamide in man. Arch Ophthalmol. 1977;95(10):1809–12.

    CAS  PubMed  Google Scholar 

  3. Chapron DJ, Gomolin IH, Sweeney KR. Acezazolamide blood concentrations are excessive in the elderly: propensity for acidosis and relationship to renal function. J Clin Pharmacol. 1989;29(4):348–53.

    CAS  PubMed  Google Scholar 

  4. Lehmann B, Linnér E, Wistrand PJ. The pharmacokinetics of acetazolamide in relation to its use in the treatment of glaucoma and to its effects as an inhibitor of carbonic anhydrase. Adv Biosciences. 1970;5:197–217.

    Google Scholar 

  5. Van Berkel MA, Elefritz JL. Evaluating off-label uses of acetazolamide. Am J Health-Syst Pharm. 2018;75(8):524–31.

    PubMed  Google Scholar 

  6. Chapron DJ, Sweeney KR, Feig PU, Kramer PA. Influence of advanced age on the disposition of acetazolamide. Br J Clin Pharmacol. 1985;19(3):363–71.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Heller I, Halevy J, Cohen S, Theodor E. Significant metabolic acidosis induced by acetazolamide – not a rare complication. Arch Intern Med. 1985;15(10):1815–7.

    Google Scholar 

  8. Schwenk MH, St Peter WL, Meese MG, Singhal PC. Acetazolamide toxicity and pharmacokinetics in patients receiving hemodialysis. Pharmacotherapy. 1995;15(4):522–7.

    CAS  PubMed  Google Scholar 

  9. Maren TH. Carbonic anhydrase: chemistry, physiology, and inhibition. Physiol Rev. 1967;47(4):595–781.

    CAS  PubMed  Google Scholar 

  10. Maren TH, Sanyal G. The activity of sulfonamides and anions against the carbonic anhydrases of animals, plants, and bacteria. Annu Rev Pharmacol Toxicol. 1983;23:439–59.

    CAS  PubMed  Google Scholar 

  11. Swenson ER. A comparative approach to carbonic anhydrase: the work of Thomas H. Maren. Comp Biochem Physiol A Mol Integr Physiol. 2003;136(2):229–41.

    PubMed  Google Scholar 

  12. Schmidt SD, Costa A, Rani B, et al. The role of carbonic anhydrases in extinction of contextual fear memory. Proc Natl Acad Sci USA. 2020;117(27):16000–8.

    PubMed  PubMed Central  Google Scholar 

  13. Blandina P, Provensi G, Passani MB, Capasso C, Supuran CT. Carbonic anhydrase modulation of emotional memory. Implications for the treatment of cognitive disorders. J Enzyme Inhim Med Chem. 2020;35(1):1206–14.

    CAS  Google Scholar 

  14. Kumar S, Rulhania S, Jaswal S, et al. Recent advances in the medicinal chemistry of carbonic anhydrase inhibitors. Eur J Med Chem. 2021;209:112923.

    CAS  PubMed  Google Scholar 

  15. Supuran CT. An update on drug interaction considerations in the therapeutic use of carbonic anhydrase inhibitors. Expert Opin Drug Metab Toxicol. 2020;16(4):297–304.

    CAS  PubMed  Google Scholar 

  16. Mazaheri M, Assadi F, Sadeghi-Bojd S. Adjunctive acetazolamide therapy for the treatment of Bartter syndrome. Int Urol Nephrol. 2020;52(1):121–8.

    CAS  PubMed  Google Scholar 

  17. Lang S-S, Ploof J, Atkin NJ, et al. Decadron, Diamox, and Zantac: a novel combination for ventricular shunt failure in pediatric neurosurgical patients. Pediatr Emerg Care 2020 Mar 12. https://doi.org/10.1097/PEC.0000000000002070

  18. Millichap JG, Woodbury DM, Goodman LS. Mechanism of the anticonvulsant action of acetazolamide, a carbonic anhydrase inhibitor. J Pharmacol Exp Ther. 1955;115(3):251–8.

    CAS  PubMed  Google Scholar 

  19. Gray WD, Maren TH, Sisson GM, Smith FH. Carbonic anhydrase inhibition. VII. Carbonic anhydrase inhibition and anticonvulsant effect. J Pharmacol Exp Ther. 1957;121(2):160–70.

    CAS  PubMed  Google Scholar 

  20. Tanimukai H, Inui M, Hariguchi S, Kaneko Z. Antiepileptic property of inhibitors of carbonic anhydrase. Biochem Pharmacol. 1965;14(6):961–70.

    CAS  PubMed  Google Scholar 

  21. Rudzik AD, Mennear JH. The mechanism of action of anticonvulsants. II Acetazolamide Life Sci. 1966;5(8):747–56.

    CAS  PubMed  Google Scholar 

  22. Kjällquist A, Messeter K, Siesjö BK. The in vivo CO2 buffer capacity of rat brain tissue under carbonic anhydrase inhibition. Acta Physiol Scand. 1970;78(1):94–102.

    PubMed  Google Scholar 

  23. Roth LJ, Schoolar JC, Barlow CF. Sulfur-35 labeled acetazolamide in cat brain. J Pharmacol Exp Ther. 1959;125(2):128–36.

    CAS  PubMed  Google Scholar 

  24. Friis ML, Paulson OB, Hertz MM. Carbon dioxide permeability of the blood-brain barrier in man. The effect of acetazolamide Microvasc Res. 1980;20(1):71–80.

    CAS  PubMed  Google Scholar 

  25. Messeter K. The effect of acetazolamide upon the regulation of the cerebrospinal fluid pH in the rat. Acta Physiol Scand. 1972;85(1):58–70.

    CAS  PubMed  Google Scholar 

  26. Heuser D, Astrup J, Lassen NA, Betz BE. Brain carbonic acid acidosis after acetazolamide. Acta Physiol Scand. 1975;93(3):385–90.

    CAS  PubMed  Google Scholar 

  27. Severinghaus JW, Hamilton FN, Cotev S. Carbonic acid production and the role of carbonic anhydrase in decarboxylation in brain. Biochem J. 1969;114(4):703–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. 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.

    CAS  PubMed  Google Scholar 

  29. Kjällquist A, Nardini M, Siesjö BK. The effect of acetazolamide upon tissue concentrations of bicarbonate, lactate, and pyruvate in the rat brain. Acta Physiol Scand. 1969;77(1):241–51.

    PubMed  Google Scholar 

  30. Saldias C, Cabieses F, Eidelberg E. Encephalographic changes induced by the intravenous administration of acetazolamide (Diamox) in epileptic patients. Electroencephalgr Clin Neurophysiol. 1957;9(2):333–6.

    CAS  Google Scholar 

  31. Maren TH, Broder LE. The role of carbonic anhydrase in anion secretion into cerebrospinal fluid. J Pharmacol Exp Ther. 1970;172(2):197–202.

    CAS  PubMed  Google Scholar 

  32. Sturdivant NM, Smith SG, Ali SF, Wolchok JC, Balachandran K. Acetazolamide mitigates astrocyte cellular edema following mild traumatic brain injury. Sci Rep. 2016;6:33330.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Duan L, Di Q. Acetazolamide suppresses multi-drug resistance-related protein 1 and P-glycoprotein expression by inhibiting aquaporins expression in a mesial temporal epilepsy rat. Model Med Sci Monit. 2017;23:5818–25.

    PubMed  Google Scholar 

  34. Uldall M, Botfield H, Jansen-Olesen I, Sinclair A, Jensen R. Acetazolamide lowers intracranial pressure and modulates the cerebrospinal fluid secretion pathway in healthy rats. Neurosci Lett. 2017;645:33–9.

    CAS  PubMed  Google Scholar 

  35. Huang JL, Zhang YL, Xiao X, et al. Bidirectional influences of acetazolamide on central nervous system oxygen toxicity of rats. Undersea Hyperb Med. 2010;37(5):271–80.

    CAS  PubMed  Google Scholar 

  36. Collins P, Morriss GM. Changes in the surface features of choroid plexus of the rat following the administration of acetazolamide and other drugs which affect CSF secretion. J Anat. 1975;120(Pt. 3):571–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Azzam NA, Choudhury SR, Donohue JM. Changes in the surface of fine structure of choroid plexus epithelium following chronic acetazolamide treatment. J Anat. 1978;127(Pt. 2):333–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Choudhury SR, Azzam NA, Donohue JM. Changes in the surface fine structure of rat third ventricular ependyma following chronic acetazolamide treatment. J Anat. 1979;129(Pt. 1):51–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Teppema LJ. Multifaceted clinical effects of acetazolamide: will the underlying mechanisms please stand up? J Appl Physiol. 2014;116(7):713–4.

    CAS  PubMed  Google Scholar 

  40. Gotoh F, Shinhara Y. Role of carbonic anhydrase in chemical control and autoregulation of cerebral circulation. In J Neurol. 1977;11(2–3):219–27.

    CAS  Google Scholar 

  41. Benga O, Huber VJ. Brain water channel proteins in health and disease. Mol Aspects Med. 2012;33(5–6):562–78.

    CAS  PubMed  Google Scholar 

  42. Abir-Awan M, Kitchen P, Salman MM, Conner MT, Conner AC, Bill RM. Inhibitors of mammalian aquaporin water channels. Int J Mol Sci. 2019;20(7):E1589.

    PubMed  Google Scholar 

  43. Vandebroek A, Yasui M. Regulation of AQP4 in the central nervous system. Int J Mol Sci. 2020;21(5):1603.

    CAS  PubMed Central  Google Scholar 

  44. Bertone NI, Groisman AI, Mazzone GL, Cano R, Tabares L Uchitel OD. Carbonic anhydrase inhibitor acetazolamide shifts synaptic vesicle recycling to a fast mode at the mouse neuromuscular junction. Synapse 2017;71(12): https://doi.org/10.1002/syn.22009.

  45. Bueno-Junior LS, Ruggiero RN, Rossignoli MT, Del Bel EA, Leite JP, Uchitel OD. Acetazolamide potentiates the afferent drive to prefrontal cortex in vivo. Physiol Rep. 2017;5(1):e13066.

    PubMed  PubMed Central  Google Scholar 

  46. Brechue WF, Koceja DM, Stager JM. Acetazolamide reduces peripheral afferent transmission in humans. Muscle Nerve. 1997;20(12):1541–8.

    CAS  PubMed  Google Scholar 

  47. Ambrosini A, Pierelli F, Schoenen J. Acetazolamide acts on neuromuscular transmission abnormalities found in some migraineurs. Cephalalgia. 2003;23(2):75–8.

    CAS  PubMed  Google Scholar 

  48. Ruff RL. Sour on the inside, calm on the outside: how acetazolamide may stabilize membrane excitability. Muscle Nerve. 2006;34(3):263–4.

    CAS  PubMed  Google Scholar 

  49. Eguchi H, Tsujino A, Kaibara M, et al. Acetazolamide acts directly on the human skeletal muscle chloride channel. Muscle Nerve. 2006;34(3):292–7.

    CAS  PubMed  Google Scholar 

  50. Kotagal V. Acetazolamide-responsive ataxia. Semin Neurol. 2012;32:533–7.

    PubMed  Google Scholar 

  51. Alam P, Amlal S, Thakar CV, Amlal H. Acetazolamide causes renal wasting but inhibits ammoniagenesis and prevents the correction of metabolic acidosis by the kidney. Am J Physiol Renal Physiol. 2020;319(3):F366–79.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Kister SJ. Carbonic anhydrase inhibition. VI. The effect of acetazolamide on cerebrospinal fluid flow. J Pharmacol Exp Ther. 1956;117(4):402–5.

    CAS  PubMed  Google Scholar 

  53. Tschirgi RD, Frost RW, Taylor JL. Inhibition of cerebrospinal fluid formation by a carbonic anhydrase inhibitor, 2-acetylamino-1,3,4-thiadiazole-5-sulfonamide (Diamox). Proc Soc Exp Biol Med. 1954;87(2):373–8.

    CAS  PubMed  Google Scholar 

  54. Reed DJ, Woodbury DM. Effect of urea and acetazolamide on brain volume and cerebrospinal fluid pressure. J Physiol. 1962;164:265–73.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Holloway LS Jr, Cassin S. Effect of acetazolamide and ouabain on CSF production rate in the newborn dog. Am J Physiol. 1972;223(3):503–6.

    CAS  PubMed  Google Scholar 

  56. Carrion E, Hertzog JH, Medlock MD, Hauser GJ, Dalton HJ. Use of acetazolamide to decrease cerebrospinal fluid production in chronically ventilated patients with ventriculopleural shunts. Arch Dis Child. 2001;84(1):68–71.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Reed DJ, Woodbury DM, Jacobs L, Squires R. Factors affecting distribution of iodide in brain and cerebrospinal fluid. Am J Physiol. 1965;209(4):757–64.

    CAS  PubMed  Google Scholar 

  58. Faraci FM, Mayhan WG, Heistad DD. Vascular effects of acetazolamide on the choroid plexus. J Pharmacol Exp Ther. 1990;254(1):23–7.

    CAS  PubMed  Google Scholar 

  59. Coppen AJ, Russell GF. Effect of intravenous acetazolamide on cerebrospinal-fluid pressure. Lancet. 1957;270(7002):926–7.

    Google Scholar 

  60. Huttenlocher PR. Treatment of hydrocephalus with acetazolamide: results in 15 cases. J Pediatr. 1965;66:1023–30.

    CAS  PubMed  Google Scholar 

  61. Libenson MH, Kaye EM, Rosman NP, Gilmore HE. Acetazolamide and furosemide for posthemorrhagic hydrocephalus of the newborn. Pediatr Neurol. 1999;20(3):185–91.

    CAS  PubMed  Google Scholar 

  62. Mealey J Jr, Barker DT. Failure of oral acetazolamide to avert hydrocephalus in infants with myelomeningocele. J Pediatr. 1968;72(2):257–9.

    PubMed  Google Scholar 

  63. Rubin RC, Henderson ES, Ommaya AK, Walker MD, Rall DP. The production of cerebrospinal fluid in man and its modification by acetazolamide. J Neurosurg. 1966;25(4):430–6.

    CAS  PubMed  Google Scholar 

  64. Watling CJ, Cairncross JG. Acetazolamide therapy for symptomatic plateau waves in patients with brain tumors. Report of three cases J Neurosurg. 2002;97(1):224–6.

    CAS  PubMed  Google Scholar 

  65. Schoeman JF. Childhood pseudotumor cerebri: clinical and intracranial pressure response to acetazolamide and furosemide treatment in a case series. J Child Neurol. 1994;9(2):130–4.

    CAS  PubMed  Google Scholar 

  66. Kattah JC, Pula JH, Mejico LJ, McDermott MP, Kupersmith MJ, Wall M. CSF pressure, papilledema grade, and response to acetazolamide in the Idiopathic Intracranial Hypertension Treatment Trial. J Neurol. 2015;212(10):2271–314.

    Google Scholar 

  67. NORDIC Idiopathic Intracranial Hypertension Study Group Writing Committee, 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.

  68. Celebisoy N, Gökçay F, Sirin H, Akyürekli O. Treatment of idiopathic intracranial hypertension: topiramate vs acetazolamide, an open-label study. Acta Neurol Scand. 2007;116(5):322–7.

    CAS  PubMed  Google Scholar 

  69. Ball AK, Howman A, Wheatley K, et al. A randomised controlled trial of treatment for idiopathic intracranial hypertension. J Neurol. 2011;258(5):874–81.

    CAS  PubMed  Google Scholar 

  70. Ahmad SR, Moss HE. Update on the diagnosis and treatment of idiopathic intracranial hypertension. Semin Neurol. 2019;39(6):682–91.

    PubMed  PubMed Central  Google Scholar 

  71. Rock NM, Demaret T, Stéphenne X, et al. Intracranial hypertension and papilledema in a large cohort of pediatric patients with Alagille Syndrome. J Pediatr Gasteroenterol Nutr. 2020;71(5):655–62.

    Google Scholar 

  72. Hady-Cohen R, Maharshak I, Michelson M, et al. Familial intracranial hypertension in 2 brothers with PTEN mutation: expansion of the phenotypic spectrum. J Child Neurol. 2019;34(9):506–10.

    PubMed  Google Scholar 

  73. Gaier ED, Heidary G. Pediatric idiopathic intracranial hypertension. Semin Neurol. 2019;39(6):704–10.

    PubMed  Google Scholar 

  74. Tilak AM, Koehn H, Mattos J, Payne SC. Preoperative management of spontaneous cerebrospinal fluid rhinorrhea with acetazolamide. Int Forum Allergy Rhinol. 2019;9(3):265–9.

    PubMed  Google Scholar 

  75. Okazawa H, Yamauchi H, Sugimoto K, Toyoda H, Kishibe Y, Takahashi M. Effects of acetazolamide on cerebral blood flow, blood volume, and oxygen metabolism: a positron emission tomography study with healthy volunteers. J Cereb Blood Flow Metab. 2001;21(12):1472–9.

    CAS  PubMed  Google Scholar 

  76. Ehrenreich DL, Burns RA, Alman RW, Fazekas JF. Influence of acetazolamide on cerebral blood flow. Arch Neurol. 1961;5:227–33.

    CAS  PubMed  Google Scholar 

  77. McDowell HA Jr, Clark LC Jr, Galbraith JG. Prevention of cerebral ischemia during carotid occlusion by acetazolamide. South Med J. 1967;60(9):940–2.

    PubMed  Google Scholar 

  78. Cotev S, Lee J, Severinghaus JW. The effects of acetazolamide on cerebral blood flow and cerebral tissue PO2. Anesthesiology. 1968;29(3):471–7.

    CAS  PubMed  Google Scholar 

  79. Kong Y, Lunzer S, Heyman A, Thompson HK Jr, Saltzman HA. Effects of acetazolamide on cerebral blood flow of dogs during hyperbaric oxygenation. Am Heart J. 1969;78(2):229–33.

    CAS  PubMed  Google Scholar 

  80. Wilson TM, Strang R, MacKenzie ET. The response of the choroidal and cerebral circulations to changing arterial PCO2 and acetazolamide in the baboon. Invest Ophthalmol Vis Sci. 1977;16(6):576–80.

    CAS  PubMed  Google Scholar 

  81. Laux BE, Raichle ME. The effect of acetazolamide on cerebral blood flow and oxygen utilization in the rhesus monkey. J Clin Invest. 1978;62(3):585–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Hauge A, Nicolaysen G, Thoresen M. Acute effects of acetazolamide on cerebral blood flow in man. Acta Physiol Scand. 1983;11(2):233–9.

    Google Scholar 

  83. Vorstrup S, Henriksen L, Paulson OB. Effect of acetazolamide on cerebral blood flow and cerebral metabolic rate for oxygen. J Clin Invest. 1984;74(5):1634–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Lassen NA, Friberg L, Kastrup J, Rizzi D, Jensen JJ. Effects of acetazolamide on cerebral blood flow and brain tissue oxygenation. Postgrad Med J. 1987;63(737):185–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Huang SY, McCullough RE, McCullough RG, et al. Usual clinical dose of acetazolamide does not alter cerebral blood flow velocity. Respir Physiol. 1988;72(3):315–26.

    CAS  PubMed  Google Scholar 

  86. Friberg L, Kastrup J, Rizzi D, Jensen JB, Lassen NA. Cerebral blood flow and end-tidal pCO2 during prolonged acetazolamide treatment in humans. Am J Physiol 1990;258( pt. 2): H954-9

  87. Cowan F, Whitelaw A. Acute effects of acetazolamide on cerebral blood flow velocity and pCO2 in the newborn infant. Acta Paediatr Scand. 1991;80(1):22–7.

    CAS  PubMed  Google Scholar 

  88. Frankel HM, Garcia E, Malik F, Weiss JK, Weiss HR. Effect of acetazolamide on cerebral blood flow and capillary patency. J Appl Physiol. 1992;73(5):756–61.

    Google Scholar 

  89. Eicke BM, Buss E, Bähr RR, Hajak G, Paulus W. Influence of acetazolamide and CO2 on extracranial flow volume and intracranial blood flow velocity. Stroke. 1999;30(1):76–80.

    CAS  PubMed  Google Scholar 

  90. Lampl Y, Kesler A, Lorberboyn M, Gadot N, Sadeh M. Cerebrovascular reactivity in patients under long-term acetazolamide treatment. Acta Neurol Scand. 2002;105(1):59–62.

    CAS  PubMed  Google Scholar 

  91. Mukherjee B, Preece M, Houston GC, et al. Mapping of the cerebral response to acetazolamide using graded asymmetric spin echo EPI. Magn Reson Imaging. 2005;23(9):907–20.

    CAS  PubMed  Google Scholar 

  92. Gückel FJ, Brix G, Schmiedek P, et al. Cerebrovascular reserve capacity in patients with occlusive cerebrovascular disease: assessment with dynamic susceptibility contrast-enhanced MR imaging and the acetazolamide stimulation test. Radiology. 1996;201(2):405–12.

    PubMed  Google Scholar 

  93. Mathew RJ, Wilson WH. Cerebral blood flow responses to CO2 and acetazolamide: the effect of anxiety. Psychiatr Res. 1989;28(2):241–2.

    CAS  Google Scholar 

  94. Bonte FJ, Devous MD, Reisch JS. The effect of acetazolamide on regional cerebral blood flow in normal human subjects as measured by single-photon emission computed tomography. Invest Radiol. 1988;23(8):564–8.

    CAS  PubMed  Google Scholar 

  95. Harris NG, Mironova YA, Chen SF, Richards HK, Pickard JD. Preventing flow-metabolism uncoupling acutely reduces axonal injury after traumatic brain injury. J Neurotrauma. 2012;29(7):1469–82.

    PubMed  PubMed Central  Google Scholar 

  96. Grossmann WM, Koeberle B. The dose-response relationship of acetazolamide on the cerebral blood flow in normal subjects. Cerebrovasc Dis. 2000;10(1):65–9.

    CAS  PubMed  Google Scholar 

  97. Kong Y, Lunzer S, Heyman A, Saltzman HA. Protective effects of acetazolamide and hyperbaric oxygenation on experimentally induced syncope. Stroke. 1970;1(2):69–76.

    CAS  PubMed  Google Scholar 

  98. Tachtsidis I, Tisdall M, Delpy DT, Smith M, Elwell CE. Measurement of cerebral tissue oxygenation in young healthy volunteers during acetazolamide provocation: a transcranial Doppler and near-intrared spectroscopy investigation. Adv Exp Med Biol. 2008;614:389–96.

    CAS  PubMed  Google Scholar 

  99. Dallinger S, Bobr B, Findl O, Eichler HG, Schmetterer L. Effects of acetazolamide on choroidal blood flow. Stroke. 1998;29(5):997–1001.

    CAS  PubMed  Google Scholar 

  100. Yamauchi H, Okazawa H, Kishibe Y, Sugimoto K, Takahashi M. The effect of acetazolamide on the changes of cerebral blood flow and oxygen metabolism during visual stimulation. Neuroimage. 2003;20(1):543–9.

    PubMed  Google Scholar 

  101. Bonte FJ, Devous MS Sr, Reisch JS, et al. The effect of acetazolamide on regional cerebral blood flow in patients with Alzheimer’s disease or stroke as measured by single-photon emission computed tomography. Invest Radiol. 1989;24(2):99–103.

    CAS  PubMed  Google Scholar 

  102. Chang CC, Kuwana N, Ito S, Ikegami T. Response of cerebral blood flow and cerebrovascular reactivity to acetazolamide in patients with dementia and idiopathic normal-pressure hydrocephalus. Neurosurg Focus. 1999;7(4):e10–6.

    CAS  PubMed  Google Scholar 

  103. Chang CC, Kuwana N, Ito S, Ikegami T. Impairment of cerebrovascular reactivity to acetazolamide in patients with normal pressure hydrocephalus. Nucl Med Commun. 2000;21(2):139–41.

    CAS  PubMed  Google Scholar 

  104. Oishi M, Mochizuki Y, Takasu T. Regional differences in cerebrovascular reactivity to acetazolamide in Alzheimer’s disease. J Clin Neurosci. 1999;6(5):380–1.

    CAS  PubMed  Google Scholar 

  105. Stoppe G, Schütze R, Kögler A, et al. Cerebrovascular reactivity to acetazolamide in (senile) dementia of Alzheimer’s type: relationship to disease severity. Dementia. 1995;6(2):73–82.

    CAS  PubMed  Google Scholar 

  106. Neu P, Schlattmann P, Schilling A, Hartmann A. Cerebrovascular reactivity in major depression: a pilot study. Psychosom Med. 2004;66(1):6–8.

    PubMed  Google Scholar 

  107. Lemke H, de Castro AG, Schlattmann P, Heuser I, Neu P. Cerebrovascular reactivity over time-course – from major depressive episode to remission. J Psychiatr Res. 2010;44(3):132–6.

    PubMed  Google Scholar 

  108. Vagal AS, Leach JL, Fernandez-Ulloa M, Zuccarello M. The acetazolamide challenge: techniques and applications in the evaluation of chronic cerebral ischemia. AJNR Am J Neuroradiol. 2009;30(5):876–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Højer-Pedersen E. Effect of acetazolamide on cerebral blood flow in subacute and chronic cerebrovascular disease. Stroke. 1987;18(5):887–91.

    PubMed  Google Scholar 

  110. Chollet F, Celsis P, Clanet M, Guiraud-Chaumeil B, Rascol A, Marc-Vergnes JP. SPECT study of cerebral blood flow reactivity after acetazolamide in patients with transient ischemic attacks. Stroke. 1989;20(4):458–64.

    CAS  PubMed  Google Scholar 

  111. Kuwabara Y, Ichiya Y, Sasaki M, Yoshida T, Masuda K. Time dependency of the acetazolamide effect on cerebral hemodynamics in patients with chronic occlusive cerebral arteries: early steal phenomenon demonstrated by [sup 15O]H sub 2O positron emission tomography. Stroke. 1995;26(10):1825–9.

    CAS  PubMed  Google Scholar 

  112. Fujimoto K, Uwano I, Sasaki M, et al. Acetazolamide-loaded dynamic 7T MR quantitative susceptibility mapping in major cerebral artery steno-occlusive disease: comparison with PET. AJNR Am J Neuroradiol. 2020;41(5):785–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Regli F, Yamaguchi T, Waltz AG. Effects of acetazolamide on cerebral ischemia and infarction after experimental occlusion of middle cerebral artery. Stroke. 1971;2(5):456–60.

    CAS  PubMed  Google Scholar 

  114. Komiyama M, Nishikawa M, Yasui T, Sakamoto H. Reversible pontine ischemia caused by acetazolamide challenge. AJNR Am J Neuroradiol. 1997;18(9):1782–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Choksi V, Hughes M, Selwa L, Hoeffner E. Transient neurologic deficit after acetazolamide challenge for computed tomography perfusion imaging. J Comput Assist Tomagr. 2005;29(2):278–80.

    Google Scholar 

  116. Katada R, Nishitani, Honmou O, et al. Expression of aquaporin-4 augments cytotoxic brain edema after traumatic brain injury during acute ethanol exposure. Am J Pathol 2012;180(1):17–23.

  117. Glober NK, Sprague S, Ahmad S, et al. Acetazolamide treatment prevents redistribution of astrocyte aquaporin 4 after murine traumatic brain injury. Neurosci J 2019; May 2;2019:2831501.

  118. Neu P, Schwertfeger N, Schlattmann P, Heuser I, Berman RM. Cerebrovascular reactivity following administration of mirtazapine in healthy probands – a randomized, placebo controlled double-blind clinical study. J Psychiatr Res. 2006;40(4):349–52.

    PubMed  Google Scholar 

  119. de Castro AG, Bajbouj M, Schlattmann P, Lemke H, Heuser I, Neu P. Cerebrovascular reactivity in depressed patients without vascular risk factors. J Psychiatr Res. 2008;42(1):78–82.

    PubMed  Google Scholar 

  120. Dahl A, Russell D, Rootwelt K, Nyberg-Hansen R, Kerty E. Cerebral vasoreactivity assessed with transcranial Doppler and regional cerebral blood flow measurements. Dose, serum concentration, and time course of the response to acetazolamide. Stroke. 1995;26(12):2302–6.

    CAS  PubMed  Google Scholar 

  121. Hamann GF, Stoll M, Jost V, Bompotti UA, Fitridge R, Schimrigk K. Time course of acetazolamide effect in normal persons. J Neuroimaging. 1996;6(1):29–31.

    CAS  PubMed  Google Scholar 

  122. Ryoo HG, Choi H, Lee DS. Deep learning-based interpretation of basal/acetazolamide brain perfusion SPECT leveraging unstructured reading reports. Eur J Nucl Med Mol Imaging. 2020;47(9):2186–96.

    CAS  PubMed  Google Scholar 

  123. Kuttner S, Knutsen Wickstrøm K, Lubberink M, et al. Cerebral blood flow measurements with 15O-water PET using a non-invasive machine-learning-derived arterial input function. J Cereb Blood Flow Metab 2021 Feb 8;271678X21991393. https://doi.org/10.1177/027168X21991393

  124. Shirai T, Meyer JS, Akiyama H, Mortel KF, Wills PM. Acetazolamide testing of cerebral vasodilator capacity provokes “vascular” but not tension headaches. Headache. 1996;32(10):589–94.

    Google Scholar 

  125. Wong TH, Shagera QA, Ryoo HG, Ha S, Le DS. Basal and acetazolamide brain perfusion SPECT in internal carotid artery stenosis. Nucl Med Mol Imaging. 2020;54(1):9–27.

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Forwand SA, Landowne M, Follansbee JN, Hansen JE. Effect of acetazolamide on acute mountain sickness. N Engl J Med. 1968;279(16):139–45.

    Google Scholar 

  127. Editorial. Acetazolamide in control of acute mountain sickness. Lancet 1981;1(8213):180–3.

  128. Greene MK, Kerr AM, McIntosh IB, Prescott RJ. Acetazolamide in prevention of acute mountain sickness: a double-blind controlled cross-over study. BMJ. 1981;283(6295):811–3.

    CAS  PubMed  PubMed Central  Google Scholar 

  129. McIntosh IB, Prescott RJ. Acetazolamide in prevention of acute mountain sickness. J Int Med Res. 1986;14(5):285–7.

    CAS  PubMed  Google Scholar 

  130. Hackett PH, Schoene RB, Winslow RM, Peters RM Jr, West JB. Acetazolamide and exercise in sojourners to 6,300 meters - a preliminary study. Med Sci Sport Exerc. 1985;17(5):590–7.

    Google Scholar 

  131. Basnyat B, Gertsch JH, Johnson EW, Castro-Marin F, Inoue Y, Yeh C. Efficacy of low-dose acetazolamide (125 mg BID) for the prophylaxis of acute mountain sickness: a prospective, double-blind, randomized, placebo-controlled trial. High Alt Med Biol. 2003;4(1):45–52.

    CAS  PubMed  Google Scholar 

  132. Carlsten C, Swenson ER, Ruoss S. A dose-response study of acetazolamide for acute mountain sickness prophylaxis in vacationing tourists at 12,000 feet (3630 m). High Alt Med Biol. 2004;5(1):33–9.

    CAS  PubMed  Google Scholar 

  133. Basnyat B, Gertsch JH, Holck PS, et al. Acetazolamide 125 mg BD is not significantly different from 375 mg BD in the prevention of acute mountain sickness: the prophylactic acetazolamide dosage comparison for efficacy (PACE) trial. High Alt Med Biol. 2006;7(1):17–27.

    CAS  PubMed  Google Scholar 

  134. Chow T, Browne V, Heileson HL, Wallace D, Anholm J, Green SM. Ginkgo biloba and acetazolamide prophylaxis for acute mountain sickness: a randomized, placebo-controlled trial. Arch Intern Med. 2005;165(3):296–301.

    CAS  PubMed  Google Scholar 

  135. Hillenbrand P, Pahari AK, Soon Y, et al. Prevention of acute mountain sickness by acetazolamide in Nepali porters: a double-blind controlled trial. Wilderness Environ Med. 2006;17(2):87–93.

    PubMed  Google Scholar 

  136. Wang J, Ke T, Zhang X, et al. Effects of acetazolamide on cognitive performance during high-altitude exposure. Neurotoxicol Teratol. 2013;35:28–33.

    PubMed  Google Scholar 

  137. Gao D, Wang W, Zhang R, Zhang Y. Efficacy of acetazolamide for the prophylaxis of acute mountain sickness: a systematic review, meta-analysis and trial sequential analysis of randomized clinical trials. Am J Med Sci. 2021;S0002–9629(21):00060–4. https://doi.org/10.1016/j.amjms.2020.12.022.

    Article  Google Scholar 

  138. Leaf DE, Goldfarb DS. Mechanisms of action of acetazolamide in the prophylaxis and treatment of acute mountain sickness. J Appl Physiol. 2007;102(4):1313–22.

    CAS  PubMed  Google Scholar 

  139. Ritchie ND, Baggott AV, Andrew Todd WT. Acetazolamide for the prevention of acute mountain sickness–a systematic review and meta-analysis. J Travel Med. 2012;19(5):298–307.

    PubMed  Google Scholar 

  140. Swenson ER, Teppema LJ. Prevention of acute mountain sickness by acetazolamide: as yet an unfinished story. J Appl Physiol. 2007;102(4):1305–7.

    PubMed  Google Scholar 

  141. Kayser B, Dumont L, Lysakowski C, Combescure C, Haller G, Tramèr MR. Reappraisal of acetazolamide for the prevention of acute mountain sickness: a systematic review and meta-analysis. High Alt Med Biol. 2012;13(2):82–92.

    CAS  PubMed  Google Scholar 

  142. van Patot MC, Leadbetter G 3rd, Keyes LE, Maakestad KM, Olson S, Hackett PH. Prophylactic low-dose acetazolamide reduces the incidence and severity of acute mountain sickness. High Alt Med Biol. 2008;9(4):289–93.

    PubMed  Google Scholar 

  143. McIntosh SE, Hemphill M, McDevitt MC, et al. Reduced acetazolamide dosing in countering altitude illness: a comparison of 62.5 vs 125 mg (the RADICAL Trial). Wilderness Environ Med. 2019;30(1):12–21.

    PubMed  Google Scholar 

  144. Lipman GS, Jurkiewicz C, Burnier A, et al. A randomized controlled trial of the lowest effective dose of acetazolamide for acute mountain sickness prevention. Am J Med 2020 May 29;S0002-9343(20)30439-3. https://doi.org/10.1016/j.amjmed.2020.05.003

  145. Lipman GS, Jurkiewicz C, Winstead-Derlega C, et al. Day of ascent dosing of acetazolamide for prevention of acute mountain sickness. High Alt Med Biol. 2019;20(3):271–8.

    CAS  PubMed  Google Scholar 

  146. Pun M. Rapid ascent to high altitude: acetazolamide or ibuprofen? Am J Med. 2021;134(3):e230.

    CAS  PubMed  Google Scholar 

  147. Davis C, Hackett P. Advances in the prevention and treatment of high altitude illness. Emerg Med Clin North Am. 2017;35(2):241–60.

    PubMed  Google Scholar 

  148. Sharma S, Gralla J, Ordonez JG, et al. Acetazolamide and N-acetylcysteine in the treatment of chronic mountain sickness (Monge’s disease). Respir Physiol Neurobiol. 2017;26:1–8.

    Google Scholar 

  149. Simancas-Racines D, Arevalo-Rodriguez I, Osorio D, Franco JV, Xu Y, Hidalgo R. Interventions for treating acute high altitude illness. Cochrane Database Syst Rev 2018;6:CD009567.

  150. Grissom CK, Roach RC, Sarnquist FH, Hackett PH. Acetazolamide in the treatment of acute mountain sickness: clinical efficacy and effect on gas exchange. Ann Intern Med. 1992;116:461–5.

    CAS  PubMed  Google Scholar 

  151. Richalet JP, Rivera M, Bouchet P, et al. Acetazolamide: a treatment for chronic mountain sickness. Am J Respir Crit Care Med. 2005;172(11):1427–33.

    PubMed  Google Scholar 

  152. Gertsch JH, Lipman GS, Holck PS, et al. Prospective, double-blind, randomized, placebo-controlled comparison of acetazolamide versus ibuprofen for prophylaxis against high altitude headache: the Headache Evaluation at Altitude Trial (HEAT). Wilderness Environ Med. 2010;21(3):236–43.

    PubMed  Google Scholar 

  153. White AJ. Cognitive impairment of acute mountain sickness and acetazolamide. Aviat Space Environ Med. 1984;55(7):598–603.

    CAS  PubMed  Google Scholar 

  154. Phillips L, Basnyat B, Chang Y, Swenson ER, Harris NS. Findings of cognitive impairment at high altitude: relationships to acetazolamide use and acute mountain sickness. High Alt Med Biol. 2017;18(2):121–3.

    CAS  PubMed  Google Scholar 

  155. Bradbury KE, Yurkevicius BR, Mitchell KM, et al. Acetazolamide does not alter endurance exercise performance at 3,500-m altitude. J Appl Physiol. 2020;128(2):390–6.

    CAS  PubMed  Google Scholar 

  156. Joyce KE, Lucas SJE, Bradwell AR. Acetazolamide can impair exercise performance; it depends upon the cohort studied. J Appl Physiol. 2020;128(5):1457.

    CAS  PubMed  Google Scholar 

  157. Yurkevicius BR, Bradbury KE, Nixon AC, et al. Influence of acetazolamide on hand strength and manual dexterity during a 30-h simulated high altitude exposure. Mil Med. 2020;185(7–8):e1161–7.

    PubMed  Google Scholar 

  158. Dawadi S, Basyal B, Subedi Y. Morbidity among athletes presenting for medical care during 3 iterations of an ultratrail race in the Himamayas. Wilderness Environ Med. 2020;31(4):437–40.

    PubMed  Google Scholar 

  159. Cain SM, Dunn JE 2nd. Low doses of acetazolamide to aid accommodation of men to altitude. J Appl Physiol. 1966;21(4):1195–2000.

    CAS  PubMed  Google Scholar 

  160. Wang K, Smith ZM, Buxton RB, Swenson ER, Dubowitz DJ. Acetazolamide during acute hypoxia improves tissue oxygenation in the human brain. J Appl Physiol. 2015;119(12):1494–500.

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Hung PH, Lin FC, Tsai HC, Chao HS, Chou CW, Chang SC. The usefulness of prophylactic use of acetazolamide in subjects with acute mountain sickness. J Chin Med Assoc. 2019;82(2):126–32.

    PubMed  Google Scholar 

  162. Swenson ER. Pharmacology of acute mountain sickness: old drugs and newer thinking. J Appl Physiol. 2016;1202:204–15.

    Google Scholar 

  163. Bergstom WH, Carzoli RF, Lombroso C, Davidson DT, Wallace WM. Observations on the metabolic and clinical effects of carbonic anhydrase inhibitors in epileptics. Am J Dis Child. 1952;84(6):771–2.

    CAS  Google Scholar 

  164. Ansell B, Clarke E. Acetazolamide in treatment of epilepsy. BMJ. 1956;1(4968):650–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Livingston S, Peterson D, Boks L. Ineffectiveness of Diamox in the treatment of childhood epilepsy. Pediatrics. 1956;17(4):541.

    CAS  PubMed  Google Scholar 

  166. Ross IP. Acetazolamide therapy in epilepsy. Lancet. 1958;2(7060):1308–9.

    CAS  PubMed  Google Scholar 

  167. Holowach J, Thurston DL. A clinical evaluation of acetazolamide (Diamox) in the treatment of epilepsy in children. J Pediatr. 1958;53(2):160–71.

    CAS  PubMed  Google Scholar 

  168. Chao DH, Plumb RL. Diamox in epilepsy. A critical review of 178 cases. J Pediatr. 1961;58:211–8.

    CAS  PubMed  Google Scholar 

  169. Resor SR Jr, Resor LD. Chronic acetazolamide monotherapy in the treatment of juvenile myoclonic epilepsy. Neurology. 1990;40(11):1677–88.

    PubMed  Google Scholar 

  170. Irahara K, Saito Y, Sugai K, et al. Effects of acetazolamide on epileptic apnea in migrating partial seizures in infancy. Epilepsy Res. 2011;96(1–2):185–9.

    CAS  PubMed  Google Scholar 

  171. Fine AL, Wirrell EC, Wong-Kisiel LC, Nickels KC. Acetazolamide for electrical status epilepticus in slow-wave sleep. Epilepsia. 2015;56(9):e134–8.

    CAS  PubMed  Google Scholar 

  172. Lim LL, Foldvary N, Mascha E, Lee J. Acetazolamide in women with catamenial epilepsy. Epilepsia. 2001;42(6):741–9.

    Google Scholar 

  173. Sethi KD, Hess DC, Huffnagle VH, Adams RJ. Acetazolamide treatment of paroxysmal dystonia in central demyelinating disease. Neurology. 1992;42(4):919–21.

    CAS  PubMed  Google Scholar 

  174. Varadkar S, Duncan JS, Cross JH. Acetazolamide and autosomal dominant nocturnal frontal lobe epilepsy. Epilepsia. 2003;4(7):986–7.

    Google Scholar 

  175. Golla FL, Hodge RS. Control of petit mal by acetazolamide. J Ment Sci. 1957;103(430):214–7.

    CAS  PubMed  Google Scholar 

  176. Wennberg R, Maurice C, Carlen PL, Garcia DL. Pilomotor seizures marked by infraslow activity and acetazolamide responsiveness. Ann Clin Transl Neurol. 2018;6(1):167–73.

    PubMed  PubMed Central  Google Scholar 

  177. Reiss WG, Oles KS. Acetazolamide in the treatment of seizures. Ann Pharmacother. 1996;30(5):514–9.

    CAS  PubMed  Google Scholar 

  178. Forsythe WI, Owens JR, Toothill C. Effectiveness of acetazolamide in the treatment of carbamazepine-resistant epilepsy in children. Dev Med Child Neurol. 1981;23(6):761–9.

    CAS  PubMed  Google Scholar 

  179. Sato J, Nioka M, Owada E, Ito K, Murata T. Effect of acetazolamide on the anticonvulsant potency of phenobarbital in mice. J Pharmacobiodyn. 1981;4(12):952–60.

    CAS  PubMed  Google Scholar 

  180. Katayama F, Miura H, Takanashi S. Long-term effectiveness and side effects of acetazolamide as an adjunct to other anticonvulsants in the treatment of refractory epilepsies. Brain Dev. 2002;24(3):150–4.

    PubMed  Google Scholar 

  181. Rowe TO. Acetazolamide delirium. Am J Psychiatry. 1977;134(5):587–8.

    CAS  PubMed  Google Scholar 

  182. Koch A, Woodbury DM. Effects of carbonic anhydrase inhibition of brain excitability. J Pharmacol Exp Ther. 1958;122(3):335–42.

    CAS  PubMed  Google Scholar 

  183. Hamada K, Song HK, Ishida S, Yagi K, Seino M. Contrasting effects of zonisamide and acetazolamide on amygdaloid kindling in rats. Epilepsia. 2001;42(11):1379–86.

    CAS  PubMed  Google Scholar 

  184. Yu H, Qi GL, Wang J, et al. Aquaporin 4 inhibition decreased synthesis of cytokines by acetazolamide in the hippocampus of rats with pentrazol-induced chronic epilepsy. Genet Mol Res 2016;15(3).

  185. Wang SJ, Fuh JL, Lu SR. Benign cough headache is responsive to acetazolamide. Neurology. 2000;55(1):149–50.

    CAS  PubMed  Google Scholar 

  186. Chalaupka FD. Therapeutic effectiveness of acetazolamide in hindbrain hernia headache. Neurol Sci. 2000;21(2):117–9.

    CAS  PubMed  Google Scholar 

  187. Sibon I, Ghorayeb I, Henry P. Successful treatment of hypnic headache syndrome with acetazolamide. Neurology. 2003;61(8):1151–8.

    Google Scholar 

  188. De Simone R, Marano E, Di Stasio E, Bonuso S, Fiorillo C, Bonavita V. Acetazolamide efficacy and tolerability in migraine with aura: a pilot study. Headache. 2005;45(4):385–6.

    PubMed  Google Scholar 

  189. Vahedi K, Taupin P, Djomby R, et al. Efficacy and tolerability of acetazolamide in migraine prophylaxis: a randomised placebo-controlled trial. J Neurol. 2002;249(2):206–11.

    CAS  PubMed  Google Scholar 

  190. Haan J, Sluis P, Sluis LH, Ferrari MD. Acetazolamide treatment for migraine aura status. Neurology. 2000;55(10):1588–9.

    CAS  PubMed  Google Scholar 

  191. Seet RC, Lim EC. A case of intermittent ataxia associated with migraine headaches. CMAJ. 2007;177(6):565–7.

    PubMed  PubMed Central  Google Scholar 

  192. Çelebisoy N, Gökçay F, Karahan C, et al. Acetazolamide in vestibular migraine prophylaxis: a retrospective study. Eur Arch Otorhinolaryngol. 2016;273(10):2947–51.

    PubMed  Google Scholar 

  193. Athwal BS, Lennox GG. Acetazolamide responsiveness in familial hemiplegic migraine. Ann Neurol. 1996;40(5):820–1.

    CAS  PubMed  Google Scholar 

  194. Suzuki M, Fujiwara K, Tsubuku T, Yabe I, Sasaki H, Fukuda S. Time course of downbeat positioning nystagmus in familial hemiplegic migraine type 1 treated with acetazolamide. J Neurol Sci. 2016;368:206–8.

    PubMed  Google Scholar 

  195. Paucar M, Granberg T, Lagerstedt-Robinson K, et al. SLC1A3 variant associated with hemiplegic migraine and acetazolamide-responsive MRS changes. Neurol Genet. 2020;6: e474. https://doi.org/10.1212/NXG.0000000000000474.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Forteza AM, Brozman B, Rabinstein AA, Romano JG, Bradley WG. Acetazolamide for the treatment of migraine with aura in CADASIL. Neurology. 2001;57(11):2144–5.

    CAS  PubMed  Google Scholar 

  197. Donnini I, Nannucci S, Valenti R, et al. Acetazolamide for the prophylaxis of migraine in CADASIL: a preliminary experience. J Headache Pain. 2012;13(4):299–302.

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Tojima H, Kunitomo F, Kimura H, Tatsumi K, Kuriyama T, Honda Y. Effects of acetazolamide in patients with the sleep apnoea syndrome. Thorax. 1988;43(2):113–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Glidewell RN, Orr WC, Imes N. Acetazolamide as an adjunct to CPAP treatment: a case of complex sleep apnea in a patient on long-acting opioid therapy. J Clin Sleep Med. 2009;5(1):63–4.

    PubMed  PubMed Central  Google Scholar 

  200. Ni Y-N, Thomas RJ. Acetazolamide for residual apnea and periodic breathing on continuous positive airway pressure therapy. Sleep Med. 2020;71:52–3.

    PubMed  Google Scholar 

  201. Brown LK. Acetazolamide for central sleep apnea: teaching an old drug new tricks? J Clin Sleep Med 2021. https://doi.org/10.5664/jcsm.9306

  202. Hoff E, Zou D, Schiza S, et al. Carbonic anhydrase, obstructive sleep apnea and hypertension: effects of intervention. J Sleep Res. 2020;29(2):e12956.

    PubMed  Google Scholar 

  203. DeBacker WA, Verbraecken J, Willemen M, Wittesaele W, DeCock W, Van deHeyning P. Central apnea index decreases after prolonged treatment with acetazolamide. Am J Respir Crit Care Med. 1995;151(1):87–91.

    CAS  PubMed  Google Scholar 

  204. Ginter G, Sankari A, Eshraghi M, et al. Effect of acetazolamide on susceptibility to central sleep apnea in chronic spinal cord injury. J Appl Physiol. 2020;128(4):960–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Javaheri S. Acetazolamide improves central sleep apnea in heart failure: a double-blind, prospective study. Am J Respir Crit Care Med. 2006;173(2):234–7.

    CAS  PubMed  Google Scholar 

  206. Javaheri S, Sands SA, Edwards BA. Acetazolamide attenuates Hunter-Cheyne-Stokes breathing but augments the hypercapnic ventilatory response in patients with heart failure. Ann Amer Thorac Soc. 2014;11(1):80–6.

    CAS  Google Scholar 

  207. Philippi H, Bieber I, Reitter B. Acetazolamide treatment for infantile central sleep apnea. J Child Neurol. 2001;16(8):600–3.

    CAS  PubMed  Google Scholar 

  208. Verbraecken J, Willemen M, De Cock W, Coen E, Van de Heyning P, De Backer W. Central sleep apnea after interrupting long term acetazolamide therapy. Respir Physiol. 1998;112(1):59–70.

    CAS  PubMed  Google Scholar 

  209. Naghan PS, Raeisi K, Khoundabi B, et al. The effect of acetazolamide on the improvement of central apnea caused by abusing opioid drugs in the clinical trial. Sleep Breath 2019. https://doi.org/10.1007/s11325-019-01968-3

  210. Schmickl CN, Landry SA, Orr JE, et al. Acetazolamide for OSA and central sleep apnea: a comprehensive systematic review and meta-analysis. Chest. 2020;S0012–3692(20):131904–8. https://doi.org/10.1016/j.chest.2020.06.078.

    Article  CAS  Google Scholar 

  211. Ni Y-N, Yang H, Thomas RJ. The role of acetazolamide in sleep apnea at sea level: a systematic review and meta-analysis. J Clin Sleep Med. 2021. https://doi.org/10.5664/jcsm.9116.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Muftic MK. Acetazolamide in Ménière’s disease. AMA Arch Otolaryngol. 1957;65(6):575–9.

    CAS  PubMed  Google Scholar 

  213. Varga G. The treatment of Menière’s disease with acetazolamide. J Laryngol. 1958;72(3):190–3.

    CAS  Google Scholar 

  214. Varga G, Ribari O. Action of acetazolamide in Ménière’s disease. J Laryngol. 1958;72(11):920–5.

    CAS  Google Scholar 

  215. Brookes GB, Hodge RA, Booth JB, Morrison AW. The immediate effects of acetazolamide in Menière’s disease. J Laryngol. 1982;96(1):57–72.

    CAS  Google Scholar 

  216. Brookes GB, Booth JB. Oral acetazolamide in Menière’s disease. J Laryngol. 1984;98(11):1087–95.

    CAS  Google Scholar 

  217. Sluch IM, Elliott MS, Dvorak J, Ding K, Farris BK. Acetazolamide: a new treatment for visual vertigo. Neuro-Ophthalmology. 2017;41(6):315–20.

    PubMed  PubMed Central  Google Scholar 

  218. Baloh RW, Winder A. Acetazolamide-responsive vestibulocerebellar syndrome: clinical and oculographic features. Neurology. 1991;41(3):29–21.

    Google Scholar 

  219. Richards CL, Bouchard JP, Dumas F, Tardif D. Quantitative evaluation of the effects of acetazolamide in Friedreich’s ataxia: a pilot study. Can J Neurol Sci. 1984;11(Suppl. 4):55–60.

    Google Scholar 

  220. Griggs RC, Moxley RT 3rd, Lafrance RA, McQuillen J. Hereditary paroxysmal ataxia: response to acetazolamide. Neurology. 1978;28(12):1259–64.

    CAS  PubMed  Google Scholar 

  221. Zasorin NL, Baloh RW, Myers LB. Acetazolamide-responsive episodic ataxia syndrome. Neurology. 1983;33(9):1212–4.

    CAS  PubMed  Google Scholar 

  222. Bouchard JP, Roberge C, van Gelder NM, Barbeau A. Familial periodic ataxia responsive to acetazolamide. Can J Neurol Sci. 1984;11(Suppl. 4):550–7.

    CAS  PubMed  Google Scholar 

  223. Koller W, Bahamon-Dussan J. Hereditary paroxysmal cerebellopathy: responsiveness to acetazolamide. Clin Neuropharmacol. 1987;10(1):65–8.

    CAS  PubMed  Google Scholar 

  224. Harno H, Hirvonen T, Kaunisto MA, et al. Acetazolamide improves neurotological abnormalities in a family with episodic ataxia type 2 (EA-2). J Neurol. 2004;251(2):232–4.

    PubMed  Google Scholar 

  225. Schwarz N, Bast T, Gaily E, et al. Clinical and genetic spectrum of SCN2A-associated episodic ataxia. Eur J Pediatr Neurol. 2019;23(3):438–47.

    CAS  Google Scholar 

  226. D’Adamo MC, Liantonio A, Rolland J-F, Pessia M, Imbrici P. KV1.1 channelopathies: pathophysiological mechanisms and therapeutic approaches. Int J Mol Sci. 2020;21(8):2935.

    PubMed Central  Google Scholar 

  227. Orsucci D, Raglione LM, Mazzoni M, Vista M. Therapy of episodic ataxias: case report and review of the literature. Drugs Context. 2019;8:212576.

    PubMed  PubMed Central  Google Scholar 

  228. Paucar M, Ågren R, Li T, et al. V374A KCND3 pathogenic variant associated with paroxysmal ataxia exacerbations. Neurol Genet. 2021;7(1):e546.

    PubMed  PubMed Central  Google Scholar 

  229. Martinez-Monseny A, Bolasell M, Callejón-Póo L, et al. AZATAX: acetazolamide safety and efficacy in cerebellar syndrome in PMM2 congenital disorder of glycosylation (PMM2-CDG). Ann Neurol. 2019;85(5):74–51.

    Google Scholar 

  230. Resnick JS, Engel WK, Griggs RC, Stam AC. Acetazolamide prophylaxis in hypokalemic periodic paralysis. N Engl J Med. 1968;278(11):582–6.

    CAS  PubMed  Google Scholar 

  231. Griggs RC, Engel WK, Resnick JS. Acetazolamide treatment of hypokalemic periodic paralysis. Prevention of attacks and improvement of persistent weakness. Ann Intern Med. 1970;73(1):39–48.

    CAS  PubMed  Google Scholar 

  232. Viskoper RJ, Licht A, Fidel J, Chaco J. Acetazolamide treatment in hypokalemic periodic paralysis: a metabolic and electromyographic study. Am J Med Sci. 1973;266(2):118–23.

    CAS  PubMed  Google Scholar 

  233. Vroom FW, Jarrell MA, Maren TH. Acetazolamide treatment of hypokalemic periodic paralysis. Probable mechanism of action Arch Neurol. 1975;32(6):385–92.

    CAS  PubMed  Google Scholar 

  234. Matthews E, Portaro S, Ke Q, et al. Acetazolamide efficacy in hypokalemic periodic paralysis and the predictive role of genotype. Neurology. 2011;77(22):1960–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  235. Sharawat IK, Suthar R, Sankhyan N, Singhi P. Primary hypokalemic periodic paralysis: long-term management and complications in a child. J Pediatr Neurosci. 2020;15(2):132–4.

    PubMed  PubMed Central  Google Scholar 

  236. Desaphy J-F, Altamura C, Vicart S, Fontaine B. Targeted therapies for skeletal muscle ion channelopathies: systematic review and steps towards precision medicine. J Neuromuscul Dis 2020. https://doi.org/10.3233/JND-200582

  237. Jarrell MA, Greer M, Maren TH. The effect of acidosis in hypokalemic periodic paralysis. Arch Neurol. 1976;33(11):791–3.

    CAS  PubMed  Google Scholar 

  238. Dejthevaporn C, Papsing C, Phakdeekitcharoen B, et al. Long-term effectiveness of acetazolamide on permanent weakness in hyperkalemic periodic paralysis. Neuromuscul Disorder. 2013;23(5):445–9.

    Google Scholar 

  239. Nagamine S, Sakoda S, Koide R, et al. A case of Andersen-Tawil syndrome presenting periodic paralysis exacerbated by acetazolamide. J Neurol Sci. 2014;347(1–2):385–6.

    PubMed  Google Scholar 

  240. Sansone VA, Burge J, McDermoott MP, et al. Randomized, placebo-controlled trials of dichlorphenamide in periodic paralysis. Neurology. 2016;86(15):1408–16.

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Heim J, Vemuri A, Lewis S, et al. Cataplexy in patients harboring the KCNMA1 p.N999S mutation. Mov Disord Clin Pract. 2020;7(7):861–2.

    PubMed  PubMed Central  Google Scholar 

  242. Busenbark K, Pahwa R, Hubble J, Koller W. The effect of acetazolamide on essential tremor: an open-label trial. Neurology. 1992;42(7):1394–5.

    CAS  PubMed  Google Scholar 

  243. Gunal DI, Afşar N, Bekiroglu N, Aktan S. New alternative agents in essential tremor therapy: double-blind placebo-controlled study of alprazolam and acetazolamide. Neurol Sci. 2000;21(5):315–8.

    CAS  PubMed  Google Scholar 

  244. Richter A, Hamann M. The carbonic anhydrase inhibitor acetazolamide exerts antidystonic effects in the dt(sz) mutant hamster. Eur J Pharmacol. 2004;502(1–2):105–8.

    CAS  PubMed  Google Scholar 

  245. Griggs RC, Moxley RT 3rd, Riggs JE, Engel WK. Effects of acetazolamide on myotonia. Ann Neurol. 1978;3(6):531–7.

    CAS  PubMed  Google Scholar 

  246. Kwieciński H. Treatment of myotonic dystrophy with acetazolamide. J Neurol. 1980;222(4):261–3.

    PubMed  Google Scholar 

  247. Markhorst JM, Stunnenberg BC, Ginjaar IB, Drost G, Erasmus CE, Sie LT. Clinical experience with long-term acetazolamide treatment in children with nondystrophic myotonias: a three-case report. Pediatr Neurol. 2014;51(4):537–41.

    PubMed  Google Scholar 

  248. Baig SM. Acetazolamide therapy improves action myoclonus in Ramsay Hunt Syndrome. J Neurol Sci. 1997;145(1):123–4.

    CAS  PubMed  Google Scholar 

  249. Olson WL Jr. Acetazolamide relieves concurrent episodic movement disorders encountered in Southern states. South Med J. 1999;92(2):228–32.

    PubMed  Google Scholar 

  250. Sanahuja J, Vazquez P, Falguera M. Paroxysmal hemicrania-tic syndrome responsive to acetazolamide. Cephalalgia. 2005;25(7):547–9.

    CAS  PubMed  Google Scholar 

  251. Anheim M, Maillart E, Vuillaumier-Barrot S, et al. Excellent response to acetazolamide in a case of paroxysmal dyskinesias due to GLUT1-deficiency. J Neurol. 2011;258(2):316–7.

    PubMed  Google Scholar 

  252. Michel V, Riant F, Tournier-Lasserve E, et al. Long-term improvement of paroxysmal dystonic choreoathetosis with acetazolamide. J Neurol. 2006;253(10):1362–4.

    CAS  PubMed  Google Scholar 

  253. Celebisoy N, Colakoglu Z, Akbaba Y, Yüceyar N. Continuous muscle fibre activity: a case treated with acetazolamide. J Neurol Neurosurg Psychiatry. 1998;64(2):256–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  254. Howell NA, Maher J, Fasano A. Acetazolamide-induced myokymia. Parkinsonism Relat Disord. 2015;21(5):540–3.

    Google Scholar 

  255. Matthews E, Hartley L, Sud R, Hanna MG, Muntoni F, Munot P. Acetazolamide can improve symptoms and signs in ion channel-related congenital myopathy. J Neurol Neurosurg Psychiatry. 2019;90(2):243–5.

    PubMed  Google Scholar 

  256. Guimarães J, Vale SJ. Paroxysmal dystonia induced by exercise and acetazolamide. Eur J Neurol. 2000;7(2):237–40.

    PubMed  Google Scholar 

  257. Hsieh PC, Chen SM, Guo YH, et al. Effect of acetazolamide for long-lasting paroxysmal dystonia in a patient with multiple sclerosis: a case report and review of literature. Neuropsychiatr Dis Treat. 2013;9:445–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  258. Voiculescu V, Pruskauer-Apostol B, Alecu C. Treatment with acetazolamide of brain-stem and spinal paroxysmal disturbances in multiple sclerosis. J Neurol Neurosurg Psychiatry. 1975;38(2):191–3.

    CAS  PubMed  PubMed Central  Google Scholar 

  259. Güleç S, Günal I, Torun S, Oztuna V. Cerebrospinal fluid pressure and acetazolamide treatment in idiopathic low back pain. Clin J Pain. 1998;14(2):176–7.

    PubMed  Google Scholar 

  260. Sacks W, Esser AH, Feitel B, Abbott K. Acetazolamide and thiamine: an ancillary therapy for chronic mental illness. Psychiatr Res. 1989;28(3):279–88.

    CAS  Google Scholar 

  261. Inoue H, Hazama H, Hamazoe K, et al. Antipsychotic and prophylactic effects of acetazolamide (Diamox) on atypical psychosis. Folia Psychiatr Neurol Jpn. 1984;38(4):425–36.

    CAS  PubMed  Google Scholar 

  262. Kakunje A, Prabhu A, Priya Es S, Karkal R, Pookoth RK, Pd R. Acetazolamide for antipsychotic-associated weight gain in schizophrenia. J Clin Psychopharmacol. 2018;38(6):652–3.

    PubMed  PubMed Central  Google Scholar 

  263. Brandt C, Grunze H, Normann C, Walden J. Acetazolamide in the treatment of acute mania. A case report Neuropsychobiology. 1998;38(3):202–7.

    CAS  PubMed  Google Scholar 

  264. Hayes SG. Acetazolamide in bipolar affective disorders. Ann Clin Psychiatry. 1994;6(2):91–8.

    CAS  PubMed  Google Scholar 

  265. Mathew RJ, Wilson WH, Tant S. Responses to hypercarbia induced by acetazolamide in panic disorder patients. Am J Psychiatry. 1989;146(8):996–1000.

    CAS  PubMed  Google Scholar 

  266. Takagi S, Watanabe Y, Imaoka T, Sakata M, Watanabe M. Treatment of psychogenic polydipsia with acetazolamide: a report of 5 cases. Clin Neuropharmacol. 2011;34(1):5–7.

    PubMed  Google Scholar 

  267. Ahmed SE, Khan AH. Acetazolamide: treatment of psychogenic polydipsia. Cureus. 2017;9(8):e1553.

    PubMed  PubMed Central  Google Scholar 

  268. Kapson B, Nayar S, Spiegel R. Treatment of Kleine-Levin syndrome with acetazolamide. J Clin Sleep Med. 2014;10(11):1153–4.

    PubMed  PubMed Central  Google Scholar 

  269. Hanley T, Platts MM. Acetazolamide (Diamox) in the treatment of congestive heart failure. Lancet. 1956;267(6919):357–9.

    Google Scholar 

  270. Ellsworth AJ, Larson EB, Strickland D. A randomized trial of dexamethasone and acetazolamide for acute mountain sickness prophylaxis. Am J Med. 1987;83(6):1024–30.

    CAS  PubMed  Google Scholar 

  271. Garner LL, Carl EF, Ferwerda JR. Advantages of sustained-release therapy with acetazolamide in glaucoma. Am J Ophthalmol. 1963;55(2):323–7.

    Google Scholar 

  272. Lichter PR, Newman LP, Wheeler NC, Beall OV. Patient tolerance to carbonic anhydrase inhibitors. Am J Ophthalmol. 1978;85(4):495–502.

    CAS  PubMed  Google Scholar 

  273. Becker B, Middleton WH. Long-term acetazolamide (Diamox) administration in therapy of glaucomas. AMA Arch Ophthalmol. 1955;54(2):187–92.

    CAS  PubMed  Google Scholar 

  274. Becker B. Diamox and the therapy of glaucoma. Am J Ophthalmol. 1954;38(11):109–11.

    CAS  PubMed  Google Scholar 

  275. Scmickl CN, Owens RL, Orr JE, Edwards BA, Malhotra A. Side effects of acetazolamide: a systematic review and meta-analysis screening overall risk and dose dependence. BMJ Open Respir Res. 2020;7(1):e000557.

    Google Scholar 

  276. Lawson C, Morris L, Wilson V, Burns B Jr. Hemorrhagic anuria with acute kidney injury after a single dose of acetazolamide: a case study of a rare side effect. Cureus. 2020;12(8):e10107.

    PubMed  PubMed Central  Google Scholar 

  277. Grant WM, Trotter RR. Diamox (acetazolamide) in treatment of glaucoma. AMA Arch Ophthalmol. 1954;51(6):735–9.

    CAS  PubMed  Google Scholar 

  278. Massumi RA, Evans JM. Studies on the continuous use of a carbonic anhydrase inhibitor (Diamox) in ambulatory patients. Am Heart J. 1955;49(4):626–32.

    CAS  PubMed  Google Scholar 

  279. De Carvalho CA, Lawrence C, Stone HH. Acetazolamide (Diamox) therapy in chronic glaucoma; a three-year follow-up study. AMA Arch Ophthalmol. 1958;59(6):840–9.

    Google Scholar 

  280. Brest AN, Onesti G, Sekine G, Kodama R, Moyer JH. Acetazolamide alone and in combination with reserpine in the treatment of hypertension. Angiology. 1961;62:589–92.

    Google Scholar 

  281. McMurdo ME, Hutchison GL, Lindsay G. Taste disturbance with acetazolamide. Lancet. 1990;336(8724):1190–1.

    CAS  PubMed  Google Scholar 

  282. Stewart WC, Halper LK, Johnson-Pratt L, Polis A, Hartenbaum D. Tolerability and efficacy of dorzolamide versus acetazolamide added to timolol. J Ocul Pharmacol Ther. 2002;18(3):211–20.

    CAS  PubMed  Google Scholar 

  283. Newton PN, le Thai H, Tip NQ, et al. A randomized, double-blind, placebo-controlled trial of acetazolamide for the treatment of elevated intracranial pressure in cryptococcal meningitis. Clin Infect Dis. 2002;35(6):369–72.

    Google Scholar 

  284. Leopold IH, Eisenberg LJ, Yasuna J. Experience with Diamox in glaucoma. Am J Ophthalmol. 1955;39(6):885–8.

    CAS  PubMed  Google Scholar 

  285. Kupfer C, Lawrence C, Linner E. Long-term administration of acetazolamide (Diamox) in the treatment of glaucoma. Am J Ophthalmol. 1955;40:673–80.

    CAS  PubMed  Google Scholar 

  286. Lichter PR. Reducing side effects of carbonic anhydrase inhibitors. Ophthalmology. 1981;88(3):266–9.

    CAS  PubMed  Google Scholar 

  287. Theeuwes F, Bayne W, McGuire J. Gastrointestinal therapeutic system for acetazolamide. Efficacy and side effects Arch Ophthalmol. 1978;96(12):2219–21.

    CAS  PubMed  Google Scholar 

  288. ten Hove MW, Friedman DI, Patel AD, et al. Safety and tolerability of acetazolamide in the Idiopathic Intracranial Hypertension Treatment Trial. J Neuroophthalmol. 2016;36(1):13–9.

    PubMed  Google Scholar 

  289. Braveman WS, Dexter RL, Rubin AL. Diamox (acetazolamide) as an oral diuretic in ambulatory cardiac patients. Am Heart J. 1957;54(2):284–90.

    CAS  PubMed  Google Scholar 

  290. Mendes TM. Eur J Case Rep Intern Med. 2018;5(5):000822.

    PubMed  PubMed Central  Google Scholar 

  291. Miller LG, Miller SM. Altered taste secondary to acetazolamide therapy. J Fam Pract. 1990;31(2):199–200.

    CAS  PubMed  Google Scholar 

  292. Lee YT, Wu JC, Chan FK. Acetazolamide-induced Gerstmann syndrome. Int J Clin Pract. 1999;53(7):560–1.

    CAS  PubMed  Google Scholar 

  293. Faull OK, Robertson J, Thomas O, et al. The effect of acetazolamide on saccadic latency at 3459 meters. Wilderness Environ Med. 2015;26(1):72–7.

    PubMed  Google Scholar 

  294. Graber M, Kelleher S. Side effects of acetazolamide: the champagne blues. Am J Med. 1988;84(5):979–80.

    CAS  PubMed  Google Scholar 

  295. Martínez-Mir I, Navarro Badenes J, Palop LV. Taste disturbance with acetazolamide. Ann Pharmacother. 1997;31(3):373.

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nevio Cimolai.

Ethics declarations

Ethics approval

Ethics approval is not required for this review.

Consent to participate

Consent is not required for the review.

Consent for publication

There is only one author.

Conflict of interest

The author declares no conflict of interest.

Additional information

Publisher's Note

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

This article is part of the Topical Collection on Medicine

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cimolai, N. The Neurological Spectrum for Acetazolamide Pharmacotherapy: from Basic Science to Clinical Applications. SN Compr. Clin. Med. 3, 2576–2592 (2021). https://doi.org/10.1007/s42399-021-01067-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s42399-021-01067-z

Keywords

Navigation