Actualización para el tratamiento de la hiperamonemia aguda en pacientes con errores innatos del metabolismo

  • Lizbeth López-Mejía Instituto Nacional de Pediatría, Secretaría de Salud, México.
  • Nuria Francisco-Revilla Estivill UNAM-Instituto Nacional de Pediatría, México.
  • Sara Guillén-López Instituto Nacional de Pediatría, México.
  • Itzel Carrillo-Nieto Instituto Nacional de Pediatría, México.
  • Cynthia Fernández-Lainez Instituto Nacional de Pediatría, México.
  • Isabel Ibarra-González Instituto Nacional de Pediatría, Secretaría de Salud, México.
  • Marcela Vela-Amieva Instituto Nacional de Pediatría, Secretaría de Salud, México.
  • Leticia Belmont-Martínez Instituto Nacional de Pediatría

Resumen

La hiperamonemia aguda (HAA) es una urgencia médica que causa daño neurológico y puede conducir a daño neurológico o incluso la muerte. En los pacientes con errores innatos del metabolismo (EIM) se presenta con mayor frecuencia, sobre todo en eventos de descompensación metabólica, ameritando atención de terapia intensiva. Los EIM que principalmente causan HAA incluyen los defectos del ciclo de la urea, acidemias orgánicas, defectos de oxidación de ácidos grasos y los defectos del ciclo de la carnitina. Ante un evento de HAA el objetivo primordial es preservar la vida y la integridad del sistema nervioso central, disminuyendo los niveles sanguíneos de amonio hasta valores normales, mediante tratamiento médico urgente, soporte vital y nutrición individualizada. El conocer los medicamentos y métodos disponibles para la remoción del amonio, así como las necesidades energéticas, proteicas, suplementos, cofactores y vitaminas, es muy importante para la correcta toma de decisiones médicas y nutricionales en cada paciente tomando en cuenta su condición clínica, bioquímica y enfermedad de base. El objetivo de este trabajo es presentar conceptos actuales sobre el tratamiento de la HAA en pacientes con EIM.

 

Biografía del autor/a

Lizbeth López-Mejía, Instituto Nacional de Pediatría, Secretaría de Salud, México.

Laboratorio de Errores Innatos del Metabolismo y Tamiz. 

Nuria Francisco-Revilla Estivill, UNAM-Instituto Nacional de Pediatría, México.

Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas.

Sara Guillén-López, Instituto Nacional de Pediatría, México.

Laboratorio de Errores Innatos del Metabolismo y Tamiz.

Itzel Carrillo-Nieto, Instituto Nacional de Pediatría, México.
Laboratorio de Errores Innatos del Metabolismo y Tamiz.
Cynthia Fernández-Lainez, Instituto Nacional de Pediatría, México.
Laboratorio de Errores Innatos del Metabolismo y Tamiz.
Isabel Ibarra-González, Instituto Nacional de Pediatría, Secretaría de Salud, México.

Servicio de Urgencias.

Marcela Vela-Amieva, Instituto Nacional de Pediatría, Secretaría de Salud, México.

Laboratorio de Errores Innatos del Metabolismo y Tamiz.

Leticia Belmont-Martínez, Instituto Nacional de Pediatría

Laboratorio de  errores innatos del metabolismo y tamiz. Investigadora en ciencias médicas "B"

Citas

Walker V. Ammonia metabolism and hyperammonemic disorders. Adv Clin Chem. 2014;67:73-150. doi:10.1016/bs.acc.2014.09.002.

Matoori S, Leroux JC. Recent advances in the treatment of hyperammonemia. Adv Drug Deliv Rev. 2015;90:55-68. doi:10.1016/j.addr.2015.04.009.

Wright G, Noiret L, Olde Damink SW, Jalan R. Interorgan ammonia metabolism in liver failure: the basis of current and future therapies. Liver Int. 2011;31(2):163-175. doi:10.1111/j.1478-3231.2010.02302.x

Ali R, Nagalli S. Hyperammonemia. 2020 Aug 10. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2020 Jan. PMID: 32491436.

Gropman AL, Summar M, Leonard JV. Neurological implications of urea cycle disorders. J Inherit Metab Dis 2007;30(6):865–79.

Summar ML, Dobbelaere D, Brusilow S, et al. Diagnosis, symptoms, frequency and mortality of 260 patients with urea cycle disorders from a 21-year, multicentre study of acute hyperammonaemic episodes. Acta Paediatr 2008;97(10):1420–5.

Summar ML, Mew NA. Inborn Errors of Metabolism with Hyperammonemia: Urea Cycle Defects and Related Disorders. Pediatr Clin North Am. 2018;65(2):231-246.

doi:10.1016/j.pcl.2017.11.004.

Savy N, Brossier D, Brunel-Guitton C, Ducharme-Crevier L, Du Pont-Thibodeau G, Jouvet P. Acute pediatric hyperammonemia: current diagnosis and management strategies. Hepat Med. 2018;10:105-115. doi:10.2147/HMER.S140711.

Khalessi N, Khosravi N, Mirjafari M, Afsharkhas L. Plasma Ammonia Levels in Newborns with Asphyxia. Iran J Child Neurol. 2016;10(1):42-46.

Häberle J. Clinical and biochemical aspects of primary and secondary hyperammonemic disorders. Arch Biochem Biophys. 2013;536(2):101-108. doi:10.1016/j.abb.2013.04.009.

Kenzaka T, Kato K, Kitao A, Kosami K, Minami K, Yahata S, et al. 2015) Hyperammonemia in Urinary Tract Infections. PLoS ONE 10(8):e0136220. doi:10.1371/journal.pone.0136220.

McGuire PJ, Lee HS; members of the Urea Cycle Disorders Consoritum, Summar ML. Infectious precipitants of acute hyperammonemia are associated with indicators of increased morbidity in patients with urea cycle disorders. J Pediatr.

;163(6):1705-1710.e1. doi:10.1016/j.jpeds.2013.08.029.

Jiménez PM, Ibarra GI, Fernández LC, et al. Hiperamonemia en la edad pediátrica. Estudio de 72 casos. Acta Pediatr Mex. 2013;34(5):268-274.

Couce ML, Bustos A, García-Alix A, Lázaro M, Martínez-Pardo M, Molina A, Sáenz de Pipao M, Serrano M, Sanjurjo P. Guía clínica de diagnóstico y tratamiento urgente de hiperamonemia neonatal. An Pediatr (Barc). 2009;70(2):183–188.

Dev SP, Hillmer MD, Ferri M. Videos in clinical medicine. Arterial puncture for blood gas analysis. N Engl J Med. 2011;364(5):e7. doi: 10.1056/NEJMvcm0803851. PMID: 21288091.

Arnold K, Olivares Z, Revilla N, Ibarra I, Belmont L, Vela M. Tratamiento de la hiperamonemia en pediatría. Acta Pediatr Mex 2005; 26(6):313-24.

Mukherjee S, John S. Lactulose. [Updated 2020 Jul 10]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2020 Jan. Available from: https://www.ncbi.nlm.nih.gov/books/NBK536930/

Dara N, Sayyari AA, Imanzadeh F. Hepatic encephalopathy: early diagnosis in pediatric patients with cirrhosis. Iran J Child Neurol. 2014;8(1):1-11.

Comité de Medicamentos de la Asociación Española de Pediatría. Pediamécum. Edición 2015. ISSN 2531-2464. Disponible en: https://www.aeped.es/comite-medicamentos/pediamecum/lactitol. Consultado el 09/09/2020.

Liu J, Lkhagva E, Chung H-J, Kim H-J, Hong S-T. The Pharmabiotic Approach to Treat Hyperammonemia. Nutrients 2018;10:140. doi:10.3390/nu10020140.

Wijdicks EFM. Lactulose: A Simple Sugar in a Complex Encephalopathy. Neurocrit Care. 2018;28(2):154-156. doi:10.1007/s12028-017-0494-4.

Taketomo CK. Manual de Prescripción pediátrica y neonatal. 18va edición. Editorial: INTERSISTEMAS; 2013. México ISBN:9786074432756.

Sakai Y, Seki N, Hamano K, et al. Prebiotic effect of two grams of lactulose in healthy Japanese women: a randomised, double-blind, placebo-controlled crossover trial. Benef Microbes. 2019;10(6):629-639.

Maydeo A. Lactitol or lactulose in the treatment of chronic constipation: result of a systematic. J Indian Med Assoc. 2010;108(11):789-792.

Kishor C, Ross RL, Blanchard H. Lactulose as a novel template for anticancer drug development targeting galectins. Chem Biol Drug Des. 2018;92(4):1801-1808. doi:10.1111/cbdd.13348.

Morgan MY. Current state of knowledge of hepatic encephalopathy (part III): non-absorbable disaccharides. Metab Brain Dis. 2016 Dec;31(6):1361-1364. doi: 10.1007/s11011-016-9910-2.

Acharya C, Bajaj JS. Current Management of Hepatic Encephalopathy. Am J Gastroenterol. 2018;113(11):1600-1612. doi:10.1038/s41395-018-0179-4.

Hadjihambi A, Khetan V, Jalan R. Pharmacotherapy for hyperammonemia. Expert Opin Pharmacother. 2014 Aug;15(12):1685-95. doi: 10.1517/14656566.2014.931372. PMID: 25032885.

Kaji K, Takaya H, Saikawa S, Furukawa M, Sato S, Kawaratani H, Kitade M, Moriya K, Namisaki T, Akahane T, Mitoro A, Yoshiji H. Rifaximin ameliorates hepatic encephalopathy and endotoxemia without affecting the gut microbiome diversity. World J Gastroenterol. 2017 Dec 21;23(47):8355-8366. doi: 10.3748/wjg.v23.i47.8355. PMID: 29307995; PMCID: PMC5743506.

Zullo A, Hassan C, Ridola L, Lorenzetti R, Campo SM, Riggio O. Rifaximin therapy and hepatic encephalopathy: Pros and cons. World J Gastrointest Pharmacol Ther. 2012 Aug 6;3(4):62-7. doi: 10.4292/wjgpt.v3.i4.62. PMID: 22966484; PMCID: PMC3437447.

Zhang, Y.J.; Li, S.; Gan, R.Y.; Zhou, T.; Xu, D.P.; Li, H.B. Impacts of gut bacteria on human health and diseases. Int. J. Mol. Sci. 2015, 16, 7493–7519.

Shawcross D, Jalan R. The pathophysiologic basis of hepatic encephalopathy: central role for ammonia and inflammation. Cell Mol Life Sci 2005;62: 2295-304.

McGee RG, Bakens A, Wiley K, Riordan SM, Webster AC. Probiotics for patients with hepatic encephalopathy. Cochrane Database Syst Rev. 2011 Nov 9;(11):CD008716. doi: 10.1002/14651858.CD008716.pub2. Update in: Cochrane Database Syst Rev. 2017 Feb 23;2:CD008716. PMID: 22071855.

Misel LM, Gish RG, Patton H, Mendler M. Sodium benzoate for treatment of hepatic encephalopathy. Gastroenterol Hepatol; 2013 :9(4): 219- 227.

Temblay GC, Qureshi IA. The biochemistry and toxicology of benzoic acid metabolism and its relationship to the elimination of waste nitrogen. Pharmacol Ther. 1993; 60(1):63-90.

Jalan AB: Treatment of inborn errors of metabolism. Molecular Cytogenetics. 2014; 7(Suppl 1): 142.

Protocolo Hispano-Luso de diagnóstico y tratamiento de las Hiperamonemias en pacientes neonatos y de más de 30 días de vida. Grupo de consenso reunido en Lisboa 2006 y Madrid 2007. 2da. edición. Ergon, 2009, Madrid.

Alfadhel M, Mutairi FA, Makhseed N, Jasmi FA, Al-Thihli K, Al-Jishi E, AlSayed M, Al-Hassnan ZN, Al-Murshedi F, Häberle J, Ben-Omran T. Guidelines for acute management of hyperammonemia in the Middle East region. Ther Clin Risk Manag. 2016 Mar 31;12:479-87. doi: 10.2147/TCRM.S93144. PMID: 27099506; PMCID: PMC4820220.

Batshaw ML, MacArthur RB, Tuchman M. Alternative pathway therapy for urea cycle disorders: twenty years later. J Pediatr. 2001; (1 Suppl):S46-54; discussion S54-5. doi: 10.1067/mpd.2001.111836.

Mawal Y, Paradis K, Qureshi IA. Developmental profile of mitocondrial glycine N-acyltransferase in human liver. J Pediatr 1997; 130: 1003-7.

Enns GM, Berry SA, Berry GT, Rhead WJ, Brusilow SW, Hamosh A. Survival after treatment with phenylacetate and benzoate for urea-cycle disorders. N Engl J Med. 2007 May 31;356(22):2282-92. doi: 10.1056/NEJMoa066596. PMID: 17538087.

Rose C, Michalak A, Rao KV, Quack G, Kircheis G, Butterworth RF. L-ornithine-L- aspartate lowers plasma and cerebrospinal fluid ammonia and prevents brain edema in rats with acute liver failure. Hepatology 1999; 30:63-40.

Goh ET, Stokes CS, Sidhu SS, Vilstrup H, Gluud LL, Morgan MY. L-ornithine L-aspartate for prevention and treatment of hepatic encephalopathy in people with cirrhosis. Cochrane Database of Systematic reviews 2018; 5: CD012410.

Butterworth RF. Beneficial effects of L-ornithine L-aspartate for prevention of overt hepatic encephalopathy in patients with cirrhosis: a systematic review with meta-analysis. Metab Brain Dis. 2020 Jan;35(1):75-81. doi: 10.1007/s11011-019-00463-8.

Sraedt U, Leweling H, Gladisch R, et al. Effects of ornithine aspartate on plasma ammonia and plasma amino acids in patients with cirrhosis. A double-blind, randomized study using a four-fold crossover design. J Hepatol 1993;19: 424-30.

P.R. Vademecum [Internet].México: [ Fecha de acceso: 21 de diciembre de 2020]. Disponible en: http:// mx.prvademecum.com/medicamento/hepa-merz-9833/.

Jalan R, Wright G, Davies NA, Hodges SJ. L-ornithine phenylacetate (OP): a novel treatment for hyperammonemia and hepatic encephalopathy. Med Hypotheses. 2007;69: 1064-1069.

Jover-Cobos M, Noiret L, Lee K, Sharma V, Habtesion A, Romero-Gómez M, et al. Ornithine phenylacetate targets alterations in the expression and activity of glutamine synthase and glutaminase to reduce ammonia levels in bile duct ligated rats. J Hepatol. 2014; 60: 545-553.

Stravitz RT, Gottfried M, Durkalski V, Fontana RJ, Hanje AJ, Koch D, et al. Safety, tolerability, and pharmacokinetics of L-ornithine phenylacetate in patients with acute liver injury/failure and hyperammonemia. Hepatology 2018; 67:1003-1013.

Daniotti M, la Marca G, Fiorini P, Filippi L. New developments in the treatment of hyperammonemia: emerging use of carglumic acid. Int J Gen Med. 2011;4:21-28. doi:10.2147/IJGM.S10490.

Matsumoto S, Häberle J, Kido J, Mitsubuchi H, Endo F, Nakamura K. Urea cycle disorders-update. J Hum Genet. 2019;64(9):833-847. doi:10.1038/s10038-019-0614-4.

Ah Mew N, Simpson KL, Gropman AL, et al. Urea Cycle Disorders Overview. 2003 Apr 29 [Updated 2017 Jun 22]. In: Adam MP, Ardinger HH, Pagon RA, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2020. Available from: https://www.ncbi.nlm.nih.gov/books/NBK1217/

Häberle J, Burlina A, Chakrapani A, Dixon M, Karall D, Lindner M, et al. Suggested guidelines for the diagnosis and management of urea cycle disorders: First revision. Journal of Inherited Metabolic Disease 2019;42:1192–230. doi:10.1002/jimd.12100.

Rosenthal MD, Carrott PW, Patel J, Kiraly L, Martindale RG. Parenteral or Enteral Arginine Supplementation Safety and Efficacy. Journal of Nutrition 2016;146:2594S–600S. doi:10.3945/jn.115.228544.

Schwedhelm E, Maas R, Freese R, Jung D, Lukacs Z, Jambrecina A, et al.. Pharmacokinetic and pharmacodynamic properties of oral L-citrulline and L-arginine: impact on nitric oxide metabolism. British Journal of Clinical Pharmacology 2008;65:51–9. doi:10.1111/j.1365-2125.2007.02990.x.

Molema F, Gleich F, Burgard P, Ploeg AT, Summar ML, Chapman KA, et al.. Evaluation of dietary treatment and amino acid supplementation in organic acidurias and urea‐cycle disorders: On the basis of information from a European multicenter registry. Journal of Inherited Metabolic Disease 2019;42:1162–75. doi:10.1002/jimd.12066.

El-Hattab AW, Almannai M, Scaglia F. Arginine and citrulline for the treatment of MELAS syndrome. J Inborn Errors Metab Screen. 2017;5:10.1177/2326409817697399. doi:10.1177/2326409817697399.

Soares JDP, Howell SL, Teixeira FJ, Pimentel GD. Dietary Amino Acids and Immunonutrition Supplementation in Cancer-Induced Skeletal Muscle Mass Depletion: A Mini-Review. Curr Pharm Des. 2020;26(9):970-978. doi:10.2174/1381612826666200218100420.

Papadia C, Osowska S, Cynober L, Forbes A. Citrulline in health and disease. Review on human studies. Clinical Nutrition 2018;37:1823–8. doi:10.1016/j.clnu.2017.10.009.

Mayo Foundation for Medical Education and Research (MFMER). L-arginina.(Consultado el 21 de diciembre de 2020). Disponible en: https://www.mayoclinic.org/es-es/drugs-supplements-l-arginine/art-20364681.

Almannai M, Alfadhel M, El-Hattab AW. Carnitine Inborn Errors of Metabolism. Molecules. 2019;24(18):3251. doi:10.3390/molecules24183251.

Harthan AA. An Introduction to Pharmacotherapy for Inborn Errors of Metabolism. J Pediatr Pharmacol Ther. 2018;23(6):432-446. doi:10.5863/1551-6776-23.6.432.

Katayama K. Zinc and protein metabolism in chronic live diseases. Nutrition Research. 2020; 74: 1-9.

Dejong CH, Deutz NE, Soeters PB. Intestinal glutamine and ammonia metabolism during chronic hyperammonaemia induced by liver insufficiency. Gut 1993; 34: 1112-1119. doi: 10.1136/gut.34.8.1112.

Katayama K, Saito M, Kawaguchi T, Endo R, Sawara K, Nishiguchi S, Kato A, Kohgo H, Suzuki K, Sakaida I, Ueno Y, Habu D, Ito T, Moriwaki H, Suzuki K. Effect of zinc on liver cirrhosis with hyperammonemia: a preliminary randomized, placebo-controlled double-blind trial. Nutrition. 2014 Nov-Dec;30(11-12):1409-14. doi: 10.1016/j.nut.2014.04.018.

Mohommad MK, Zhou Z, Cave M, Barve A, McClain C. Zinc and liver disease. Nutr Clin Pract. 2012; 27 (1): 8-20.

Haijes HA, Jans JJM, Tas SY, Verhoeven‐Duif NM, Hasselt PM. Pathophysiology of propionic and methylmalonic acidemias. Part 1: Complications. Journal of Inherited Metabolic Disease 2019;42:730–44. doi:10.1002/jimd.12129.

Häberle J, Chakrapani A, Ah Mew N, Longo N. Hyperammonaemia in classic organic acidaemias: a review of the literature and two case histories. Orphanet Journal of Rare Diseases 2018;13. doi:10.1186/s13023-018-0963-7.

Baumgartner MR, Hörster F, Dionisi-Vici C, Haliloglu G, Karall D, Chapman KA, et al.. Proposed guidelines for the diagnosis and management of methylmalonic and propionic acidemia. Orphanet J Rare Dis 2014;9:130. doi:10.1186/s13023-014-0130-8.

Ramezanpour Ahangar E, Annamaraju P. Hydroxocobalamin. [Updated 2020 Jun 13]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2020 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK557632/

Shipton MJ, Thachil J. Vitamin B12 deficiency – A 21st century perspective. Clinical Medicine 2015;15:145–50. doi:10.7861/clinmedicine.15-2-145.

Mock DM. Biotin: From Nutrition to Therapeutics. The Journal of Nutrition 2017;147:1487–92. doi:10.3945/jn.116.238956.

Bistas KG, Tadi P. Biotin. [Updated 2020 Jul 8]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2020 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK554493/

D'souza AA, Shegokar R. Polyethylene glycol (PEG): a versatile polymer for pharmaceutical applications. Expert Opin Drug Deliv. 2016;13(9):1257-1275. doi:10.1080/17425247.2016.1182485.

Koppen IJN, Broekaert IJ, Wilschanski M, Papadopoulou A, Ribes-Koninckx C, Thapar N, et al.. Role of Polyethylene Glycol in the Treatment of Functional Constipation in Children. Journal of Pediatric Gastroenterology and Nutrition 2017;65:361–3. doi:10.1097/mpg.0000000000001704.

Rahimi RS, Singal AG, Cuthbert JA, Rockey DC. Lactulose vs Polyethylene Glycol 3350-Electrolyte Solution for Treatment of Overt Hepatic Encephalopathy. JAMA Internal Medicine 2014;174:1727. doi:10.1001/jamainternmed.2014.4746.

Naderian M, Akbari H, Saeedi M, Sohrabpour AA. olyethylene Glycol and Lactulose versus Lactulose Alone in the Treatment of Hepatic Encephalopathy in Patients with Cirrhosis: A Non-Inferiority Randomized Controlled Trial. Middle East Journal of Digestive Diseases 2017;9:12–9. doi:10.15171/mejdd.2016.46.

Shehata HH, Elfert AA, Abdin AA, Soliman SM, Elkhouly RA, Hawash NI, et al. Randomized controlled trial of polyethylene glycol versus lactulose for the treatment of overt hepatic encephalopathy. European Journal of Gastroenterology & Hepatology 2018;30:1476–81. doi:10.1097/meg.0000000000001267.

Raina R, Bedoyan JK, Lichter-Konecki U, et al. Consensus guidelines for management of hyperammonaemia in paediatric patients receiving continuous kidney replacement therapy. Nat Rev Nephrol. 2020;16(8):471-482. doi:10.1038/s41581-020-0267-8.

Spinale JM, Laskin BL, Sondheimer N, Swartz SJ, Goldstein SL. High-dose continuous renal replacement therapy for neonatal hyperammonemia. Pediatr Nephrol. 2013;28(6):983-986. doi:10.1007/s00467-013-2441-8.

Demirkol D, Aktuğlu Zeybek Ç, Karacabey BN, Cesur Y, Ataman Y, Soysal E. The Role of Supportive Treatment in the Management of Hyperammonemia in Neonates and Infants. Blood Purif. 2019;48(2):150-157. doi:10.1159/000495021.

Yetimakman AF, Kesici S, Tanyildiz M, Bayrakci B. Continuous Renal Replacement Therapy for Treatment of Severe Attacks of Inborn Errors of Metabolism. J Pediatr Intensive Care. 2019;8(3):164-169. doi:10.1055/s-0039-1683991.

Aygun F, Aygun D, Erbek Alp F, Zubarıoglu T, Zeybek C, Cam H. The impact of continuous renal replacement therapy for metabolic disorders in infants. Pediatr Neonatol. 2018;59(1):85-90. doi:10.1016/j.pedneo.2017.04.004.

Robinson JR, Conroy PC, Hardison D, et al. Rapid resolution of hyperammonemia in neonates using extracorporeal membrane oxygenation as a platform to drive hemodialysis. J Perinatol. 2018;38(6):665-671. doi:10.1038/s41372-018-0084-0.

Bosoi CR, Parent-Robitaille C, Anderson K, Tremblay M, Rose CF. AST-120 (spherical carbon adsorbent) lowers ammonia levels and attenuates brain edema in bile duct-ligated rats. Hepatology. 2011;53(6):1995-2002. doi: 10.1002/hep.24273.

Bajaj JS, M.Y. Sheikh, M. Chojkier, L. Balart, A.H. Sherker, R. Vemuru, J.M. Vierling, N. Sussman, G. Morelli, K.E. Anderson, M. Scott Harris, K.D. Mullen. AST-120 (spherical carbon adsorbent) in covert hepatic encephalopathy: results of the ASTUTE trial, J. Hepatol. 58 (2013) S84.

Ampuero J, Ranchal I, Nuñez D, Díaz-Herrero M del M, Maraver M, del Campo JA, Rojas Á, Camacho I, Figueruela B, Bautista JD, Romero-Gómez M. Metformin inhibits glutaminase activity and protects against hepatic encephalopathy. PLoS One. 2012;7(11):e49279. doi: 10.1371/journal.pone.0049279.

Stödberg T, Frostell CG, and Larsson A. Unconsciousness, Coma, and Death by Neurological Criteria. In Sejersen T and Wang CH, editors. Acute Pediatric Neurology. London; 2014: Springer-Verlag. p. 3-22.

Hajiloizow SM and Riviello JJ. Coma and other states of altered awareness in children.In David RB ed. Clinical pediatric neurology. Third edition. New York; 2009. Demos medical publishing. p. 495-505.

Calligaris L, Vidoni A, Bruno I, Vidoni M, Barbi E. Efficacy of clonidine in hyperammonemia induced hyperexcitability syndrome. Paediatr Anaesth. 2013;23(2):202-4. doi: 10.1111/pan.12088.

Su PH, Chen JY, Chen YJ, Niu DM, Hsu JH, Lee IC. Electroencephalography and transcranial Doppler ultrasonography in neonatal citrullinemia. J Formos Med Assoc. 2014 Nov;113(11):857-61. doi: 10.1016/j.jfma.2010.12.004.

Schizodimos T, Soulountsi V, Iasonidou C, Kapravelos N. An overview of management of intracranial hypertension in the intensive care unit. J Anesth. 2020;34(5):741-757. doi:10.1007/s00540-020-02795-7.

Publicado
2022-10-10
Cómo citar
López-Mejía, L., Francisco-Revilla Estivill, N., Guillén-López, S., Carrillo-Nieto, I., Fernández-Lainez, C., Ibarra-González, I., Vela-Amieva, M., & Belmont-Martínez, L. (2022). Actualización para el tratamiento de la hiperamonemia aguda en pacientes con errores innatos del metabolismo. Acta Pediátrica De México, 43(5), 293-313. https://doi.org/10.18233/APM43No5pp293-3132253
Sección
Artículo de revisión