Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-06-01T04:36:15.483Z Has data issue: false hasContentIssue false

Section III - Hepatitis and Immune Disorders

Published online by Cambridge University Press:  19 January 2021

Frederick J. Suchy
Affiliation:
University of Colorado, Children’s Hospital Colorado, Aurora
Ronald J. Sokol
Affiliation:
University of Colorado, Children’s Hospital Colorado, Aurora
William F. Balistreri
Affiliation:
Cincinnati Children’s Hospital Medical Center, Cincinnati
Jorge A. Bezerra
Affiliation:
Cincinnati Children’s Hospital Medical Center, Cincinnati
Cara L. Mack
Affiliation:
University of Colorado, Children’s Hospital Colorado, Aurora
Benjamin L. Shneider
Affiliation:
Texas Children’s Hospital, Houston
Get access
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2021

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Rezende, G, Roque-Afonso, AM, Samuel, D, et al. Viral and clinical factors associated with the fulminant course of hepatitis A infection. Hepatology 2003;38:613–18.Google Scholar
Robertson, BH, Jansen, RW, Khanna, B, et al. Genetic relatedness of hepatitis A virus strains recovered from different geographical regions. J Gen Virol 1992;73:1365–77.Google Scholar
Fujiwara, K, Yokosuka, O, Fukai, K, et al. Analysis of full-length hepatitis A virus genome in sera from patients with fulminant and self-limited acute type A hepatitis. J Hepatol 2001;35:112–19.Google Scholar
Taylor, GM, Goldin, RD, Karayiannis, P, et al. In situ hybridization studies in hepatitis A infection. Hepatology 1992;16:642–8.CrossRefGoogle ScholarPubMed
Ticehurst, JR, Racaniello, VR, Baroudy, BM, et al. Molecular cloning and characterization of hepatitis A virus cDNA. Proc Natl Acad Sci USA 1983;80:5885–9.CrossRefGoogle ScholarPubMed
Koff, RS. Hepatitis, A. Lancet 1998;351:1643–9.CrossRefGoogle Scholar
Craig, AS, Schaffner, W. Prevention of hepatitis A with the hepatitis A vaccine. N Engl J Med 2004;350:476–81.Google Scholar
Nelson, N. Hepatitis, A outbreaks. Presented at: Advisory Committee on Immunization Practices; October 25, 2017; Atlanta, GA. Available at: www.mdedge.com/familymedicine/article/155079/vaccines/cdc-provides-advice-recent-hepatitis-outbreaks [last accessed June 12, 2020].Google Scholar
Centers for Disease Control and Prevention. Hepatitis Surveillance Report 59. Atlanta, GA: US Department of Health and Human Services, Centers for Disease Control and Prevention, 2004:160.Google Scholar
Centers for Disease Control and Prevention. Hepatitis A outbreak associated with green onions at a restaurant – Monaca, Pennsylvania, MMWR Morb Mortal Wkly Rep 2003;52:1155–7.Google Scholar
Wheeler, C, Vogt, TM, Armstrong, GL, et al. An outbreak of hepatitis A associated with green onions. N Engl J Med 2005;353:890–7.Google Scholar
Hutin, YJF, Pool, V, Cramer, EH, et al. A multistate, foodborne outbreak of hepatitis A. N Engl J Med 1999;340:595602.CrossRefGoogle ScholarPubMed
Weinberg, M, Hopkins, J, Farrington, L, et al. Hepatitis A in Hispanic children who live along the United States– Mexico border: the role of international travel and food-borne exposures. Pediatrics 2004;114:e6873.CrossRefGoogle ScholarPubMed
Hadler, SC, Erben, JJ, Francis, DP, et al. Risk factors for hepatitis A in day-care centers. J Infect Dis 1982;145:255–61.Google Scholar
Benenson, MW, Takafuji, ET, Bancroft, WH, et al. A military community outbreak of hepatitis type A related to transmission in a child care facility. Am J Epidemiol 1980;112:471–81.Google Scholar
Vernon, AA, Schable, C, Francis, D. A large outbreak of hepatitis A in a daycare center: association with non-toilet-trained children and persistence of IgM antibody to hepatitis A virus. Am J Epidemiol 1982;115:325–31.Google Scholar
Hadler, SC, Webster, HM, Erber, JJ, et al. Hepatitis A in day care centers: a community-wide assessment. N Engl J Med 1980;302:1222–7.Google Scholar
Centers for Disease Control and Prevention. Viral hepatitis outbreaks. Atlanta, GA: Centers for Disease Control and Prevention 2019. Available at: www.cdc.gov/hepatitis/outbreaks/hepatitisaoutbreaks.htm [last accessed June 12, 2020].Google Scholar
Balistreri, WF. Viral hepatitis. Emerg Clin North Am 1991;9:365–99.CrossRefGoogle Scholar
Leach, C. Hepatitis A in the United States. Pediatr Infect Dis J 2004;23:551–2.CrossRefGoogle ScholarPubMed
Jung, YM, Park, SJ, Kim, JS, et al. Atypical manifestations of hepatitis A infection: a prospective, multicenter study in Korea. J Med Virol 2010;82:1318–26.CrossRefGoogle ScholarPubMed
Rachima, CM, Cohen, E, Garty, M. Acute hepatitis A: combination of relapsing and the cholestatic forms, two rare variants. Am J Med Sci 2000;319: 417–19.CrossRefGoogle ScholarPubMed
Schiff, ER. Atypical clinical manifestations of hepatitis A. Vaccine 1992;10(Suppl1):S18S20.Google Scholar
Ozcay, F, Canan, O, Akcan, B, et al. Hepatitis A super infection as a cause of liver failure in a child with Wilson’s disease. Turk J Pediatr 2007;49:199202.Google Scholar
Ciocca, M, Moreira-Silva, SF, Alegria, S, et al. Hepatitis A as an etiologic agent of acute liver failure in Latin America. Pediatr Infect Dis J 2007;26:711–15.Google Scholar
Centers for Disease Control and Prevention. Prevention of hepatitis A through active or passive immunization: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Morb Mortal Wkly Rep 1999;48:139.Google Scholar
Kim, HY, Eyheramonho, MB, Pichavant, M, et al. A polymorphism in TIM1 is associated with susceptibility to severe hepatitis A virus infection in humans. J Clin Invest 2011;121:1111–18.CrossRefGoogle ScholarPubMed
Arora, NK, Mathur, P, Ahuja, A, Oberoi, A. Acute liver failure. Indian J Pediatr 2003;70:73–9.Google Scholar
Amon, JJ, Darling, N, Fiore, AE, et al. Factors associated with hepatitis A vaccination among children 24 to 35 months of age: United States, 2003. Pediatrics 2006;117:30–3.Google Scholar
Centers for Disease Control and Prevention. Vaccines and Immunizations. Atlanta, GA: Centers for Disease Control and Prevention 2019. Available at: www.cdc.gov/vaccines [last accessed June 12, 2020].Google Scholar
Centers for Disease Control and Prevention. Vaccine Recommendations of the Advisory Committee for Immunization Practices. Atlanta, GA: Centers for Disease Control and Prevention 2019. Available at: www.cdc.gov/vaccines/pubs/ACIP-list.htm [last accessed June 12, 2019].Google Scholar
Landry, P, Tremblay, S, Darioli, R, Genton, B. Inactivated hepatitis vaccine booster given 24 months after the primary doses. Vaccine 2000;19:399402.Google Scholar
McMahon, BJ, Williams, J, Bulkow, L, et al. Immunogenicity of an inactivated hepatitis A vaccine in Alaska native children and native and non-native adults. J Infect Dis 1995;17:676–9.Google Scholar
Horng, YC, Chang, MH, Lee, CY, et al. Safety and immunogenicity of hepatitis A vaccine in healthy children. Pediatr Infect Dis J 1993;12:359–62.Google Scholar
Balcarek, KB, Bagley, MR, Pass, RF, Schiff, ER, Krause, DS. Safety and immunogenicity of an inactivated hepatitis A vaccine in preschool children. J Infect Dis 1995;171(Suppl1): S70S72.CrossRefGoogle ScholarPubMed
Ashur, Y, Adler, R, Rowe, M, Shouval, D. Comparison of immunogenicity of two hepatitis A vaccines – VAQTA and HAVRIX – in young adults. Vaccine 1999;17:2290–6.CrossRefGoogle ScholarPubMed
Letson, GW, Shapiro, CN, Kuehn, D, et al. Effect of maternal antibody on immunogenicity of hepatitis A vaccine in infants. J Pediatr 2004;144:327–32.Google Scholar
Das, A. An economic analysis of different strategies of immunization against hepatitis A virus in developed countries. Hepatology 1999;29:548–52.CrossRefGoogle ScholarPubMed
Werzberger, A, Mensch, B, Kuter, B, et al. A controlled trial of a formalin-inactivated hepatitis A vaccine in healthy children. N Engl J Med 1992;327:453–7.CrossRefGoogle ScholarPubMed
Innis, BL, Snithhan, R, Kunasol, P, et al. Protection against hepatitis A by an inactivated vaccine. JAMA 1994;271:1328–34.CrossRefGoogle ScholarPubMed
Gouvea, AF, De Moraes-pinto, MI, Ono, E, et al. Immunogenicity and tolerability of hepatitis A vaccine in HIV-infected children. Clin Infect Dis 2005;41:544–8.CrossRefGoogle ScholarPubMed
Siberry, GK, Coller, RJ, Henkle, E, et al. Antibody response to hepatitis A immunization among human immunodeficiency virus-infected children and adolescents. Pediatr Infect Dis J 2008;27:465–8.Google Scholar
Weinberg, A, Gona, P, Nachman, SA, et al. Antibody-response to hepatitis A virus vaccine in HIV-infected children with evidence of immunologic reconstitution while receiving highly active retroviral therapy. J Infect Dis 2006;193:302–11.CrossRefGoogle Scholar
Arslan, M, Wiesner, RH, Poterucha, JJ, Zein, NN. Safety and efficacy of hepatitis A vaccination in liver transplantation recipients. Transplantation 2001;72:272–6.Google Scholar
Diana, A, Posfay-Barbe, KM, Belli, DC, Siegrist, CA. Vaccine-induced immunity in children after orthotopic liver transplantation: a 12-yr review of the Swiss national reference center. Pediatr Transplant 2007;11:31–7.CrossRefGoogle ScholarPubMed
Lee, SD, Chan, CY, Yu, MI, et al. Safety and immunogenicity of inactivated hepatitis A vaccine in patients with chronic liver disease. J Med Virol 1997;52:215–18.Google Scholar
Keeffe, EB, Iwarson, S, McMahon, BJ, et al. Safety and immunogenicity of hepatitis A vaccine in patients with chronic liver disease. Hepatology 1998;27:881–6.Google Scholar
Ferreira, CT, da Silviera, TR, Viera, SM, et al. Immunogenicity and safety of hepatitis A vaccine in children with chronic liver disease. J Pediatr Gastroenterol Nutr 2003;37:258–61.CrossRefGoogle ScholarPubMed
El-Karaksy, HM, El-Hawary, MI, El- koofy, NM, et al. Safety and efficacy of hepatitis A vaccine in children with chronic liver disease. World J Gastroenterol 2006;12:7337–40.Google Scholar
Majda-Stanislawska, E, Bednarek, M, Kuydowicz, J. Immunogenicity of inactivated hepatitis A vaccine in children with chronic liver disease. Pediatr Infect Dis J 2004;23:571–4.Google Scholar
Troisi, CL, Hollinger, FB, Krause, DS, et al. Immunization of seronegative infants with hepatitis A vaccine (HAVRIX®; SKB): a comparative study of two dosing schedules. Vaccine 1997;15:1613–17.Google Scholar
Piazza, M, Safary, A, Vegnente, A, et al. Safety and immunogenicity of hepatitis A vaccine in infants: a candidate for inclusion in the childhood vaccination programme. Vaccine 1999;17:585–8.Google Scholar
Dagan, R, Amir, J, Mijalovsky, A, et al. Immunization against hepatitis A in the first year of life: priming despite the presence of maternal antibody. Pediatr Infect Dis J 2000;19:1045–52.Google Scholar
Fiore, AE, Shapiro, CN, Sabin, K, et al. Hepatitis A vaccination of infants: effect of maternal antibody status on antibody persistence and response to a booster dose. Pediatr Infect Dis J 2003;22:354–9.Google Scholar
Hammitt, LL, Bulkow, L, Hennessy, TW, et al. Persistence of antibody to hepatitis A virus 10 years after vaccination among children and adults. J Infect Dis 2008;198:1776–82.CrossRefGoogle ScholarPubMed
Werzberger, A, Mensch, B, Nalin, DR, Kuter, BJ. Effectiveness of hepatitis A vaccine in a former frequently affected community: 9 years’ follow-up after the Monroe field trial of VAQTA. Vaccine 2002;20:16991701.Google Scholar
Chang, CY, Lee, SD, Yu, MI, et al. Long-term follow-up of hepatitis A vaccination in children. Vaccine 1999;17:369–72.Google Scholar
Van Damme, P, Banatvala, J, Fay, O, et al. Hepatitis A booster vaccination: is there a need? Lancet 2003;362:1065–71.Google Scholar
Van Herckk, K, Van Damme, P. Inactivated hepatitis A vaccine-induced antibodies: follow-up and estimates of long-term persistence. J Med Virol 2001;63:17.Google Scholar
Centers for Disease Control and Prevention. Prevention of hepatitis A through active or passive immunization: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Morb Mortal Wkly Rep 2006;55(RR-7):123.Google Scholar
Rosenthal, P. Cost-effectiveness of hepatitis A vaccination in children, adolescents, and adults. Hepatology 2003;37:4451.Google Scholar
Ansari, IH, Nanda, SK, Durgapal, H, et al. Cloning sequencing, and expression of the hepatitis E virus (HEV) nonstructural open reading frame 1. J Med Virol 2000;60:275–83.Google Scholar
Krawczynski, K, Aggarwal, R, Kamili, S. Infections of the liver. Infect Dis Clin N Am 2000;14:118.Google Scholar
Worm, HC, van der Poel, WHM, Brandstatter, G. Hepatitis E: an overview. Microbes Infect 2002;4:657–66.CrossRefGoogle ScholarPubMed
Aggarwal, R, Krawczynski, K. Hepatitis E: an overview and recent advances in clinical and laboratory research. J Gastroenterol Hepatol 2000;15:920.Google Scholar
Ahn, J-M, Kang, S-G, Lee, D-Y, et al. Identification of novel human hepatitis E virus (HEV) isolates and determination of the seroprevalence of HEV in Korea. J Clin Microbiol 2005;3042–8. Doi: 10.1128/JCM.43.7.3042-3048.2005Google Scholar
Tam, AW, Smith, MM, Guerra, ME, et al. Hepatitis E virus (HEV): molecular cloning and sequencing of the full-length viral genome. Virology 1991;185:120–31.Google Scholar
Reyes, GR, Purdy, MA, Kim, JP, et al. Isolation of a cDNA from the virus responsible for enterically transmitted non-A, non-B hepatitis. Science 1990;247:1335–9.Google Scholar
Shrestha, SM, Srestha, S, Tsuda, F, et al. Genetic changes in hepatitis E virus of subtype 1a in patients with sporadic acute hepatitis E in Kathmandu, Nepal, from 1997 to 2002. J Gen Virol 2004;85:97104.Google Scholar
Mansuy, JM, Peron, JM, Abravanel, F, et al. Hepatitis E in the south west of France in individuals who have never visited an endemic area. J Med Virol 2004;74:419–24.Google Scholar
Redlinger, T, O’Rourke, K, Nickey, L, Martinez, G. Elevated hepatitis A and E seroprevalence rates in a Texas/Mexico border community. Tex Med 1998;94:6871.Google Scholar
Aggarwal, R, Naik, SR. Faecal excretion of hepatitis-E virus. Lancet 1992;340:787.Google Scholar
DeCock, KM, Bradley, DW, Sandford, NL, et al. Epidemic non-A, non-B hepatitis in patients from Pakistan. Ann Intern Med 1987;106:227–30.Google Scholar
Ray, R, Aggarwal, R, Salunke, PN, et al. Hepatitis E virus genome in stools of hepatitis patients during large epidemic in North India. Lancet 1991;338:783–4.CrossRefGoogle ScholarPubMed
Skidmore, SJ, Yarbough, PO, Gabor, KA, et al. Hepatitis E virus: the cause of waterborne hepatitis outbreak. J Med Virol 1992;37:5860.Google Scholar
Ticehurst, J, Popkin, TJ, Bryan, JP, et al. Association of hepatitis E virus with an outbreak of hepatitis in Pakistan: serologic responses and pattern of virus excretion. J Med Virol 1992;36:8492.Google Scholar
Chauhan, A, Dilawari, JB, Jameel, S, et al. Common etiological agent for epidemic and sporadic non-A non-B hepatitis. Lancet 1992;339:1509–10CrossRefGoogle ScholarPubMed
Hau, CH, Hien, TT, Tien, NT, et al. Prevalence of enteric hepatitis A and E viruses in the Mekong delta region of Vietnam. Am J Trop Med Hyg 1998;60:277–80.Google Scholar
Goto, K, Ito, K, Sugiura, T, et al. Prevalence of hepatitis E virus infection in Japanese children. J Pediatr Gastroenterol Nutr 2005;42:8992.CrossRefGoogle Scholar
Emerson, SU, Purcell, RH. Running like water: the omnipresence of hepatitis E. N Engl J Med 2004;351: 2367–8.Google Scholar
Bader, TF, Krawczynski, K, Polish, LB, et al. Hepatitis E in a US traveler to Mexico. N Engl J Med 1991;325:1659.Google Scholar
De Groen, PC. Hepatitis E in the United States: a case of “hog fever?Mayo Clin Proc 1997;72:1197–8.Google Scholar
Goldsmith, R, Yarbough, PO, Reyes, GR, et al. Enzyme-linked immunosorbent assay for diagnosis of acute sporadic hepatitis E in Egyptian children. Lancet 1992;339:328–32.Google Scholar
Hyams, KC, Purdy, MA, Kaur, M, et al. Acute sporadic hepatitis E in Sudanese children: analysis based on a new Western blot assay. J Infect Dis 1992;165:1001–5.Google Scholar
Daniel, HDJ, Warier, A, Abraham, P, Sridharan, G. Age-wise exposure rates to hepatitis E virus in a southern Indian patient population without liver disease. Am J Top Med Hyg 2004;71:675–8.Google Scholar
Khuroo, MS, Kamili, S, Yattoo, GN. Hepatitis E virus infection may be transmitted through blood transfusions in an endemic area. J Gastroenterol Hepatol 2004;19:778–84.Google Scholar
Singh, S, Mohanty, A, Joshi, YK, et al. Mother-to-child transmission of hepatitis E virus infection. Indian J Pediatr 2003;70:37–9, 597.CrossRefGoogle ScholarPubMed
Khuroo, MS, Kamili, S, Jameel, S. Vertical transmission of hepatitis E virus. Lancet 1995;345:1025–6.Google Scholar
Kumar, RM, Uduman, S, Rana, S, et al. Sero-prevalence and mother-to-infant transmission of hepatitis E virus among pregnant women in the United Arab Emirates. Eur J Obstet Gynecol Reprod Biol 2001;100:915.Google Scholar
Arora, NK, Panda, SK, Nanda, SK, et al. Hepatitis E infection in children: study of an outbreak. J Gastroenterol Hepatol 1999;14:572–7.CrossRefGoogle ScholarPubMed
Worm, HC, van der Poel, WHM, Brandstatter, G. Hepatitis E: an overview. Microbes Infect 2002;4:657–66.Google Scholar
Panda, SK, Jameel, S. Hepatitis E virus: from epidemiology to molecular biology. Viral Hepat 1997;3:227–51.Google Scholar
Kamar, N, Mansuy, JM, Cointault, O, et al. Hepatitis E virus-related cirrhosis in kidney- and kidney-pancreas-transplant recipients. Am J Transplant 2008;8:1744–8.Google Scholar
Kamar, N, Garrouste, C, Haagsma, EB, et al. Factors associated with chronic hepatitis in patients with hepatitis E virus infection who have received solid organ transplants. Gastroenterology 2011;140:1481–9.CrossRefGoogle ScholarPubMed
Legrand-Abravanel, F, Kamar, N, Sandres-Saune, K, et al. Hepatitis E virus infection without reactivation in solid-organ transplant recipients, France. Emerg Infect Dis 2011;17:30.Google Scholar
Hamid, SS, Atiq, M, Shehzad, F, et al. Hepatitis E virus superinfection in patients with chronic liver disease. Hepatology 2002;36:474–8.Google Scholar
Kumar, A, Aggarwal, R, Naik, SR, et al. Hepatitis E virus is responsible for decompensation of chronic liver disease in an endemic region. Indian J Gastroenterol 2004;23:5962.Google Scholar
Ramachandran, J, Eapen, CE, Kang, G, et al. Hepatitis E superinfection produces severe decompensation in patients with chronic liver disease. J Gastroenterol Hepatol 2004;19:134–8.Google Scholar
Davern, TJ, Chalasani, N, Fontana, RJ, et al. Acute hepatitis E infection accounts for some cases of suspected drug-induced liver injury. Gastroenterology 2011;14:1665–72.Google Scholar
Bryan, JP, Tsarev, SA, Iqbal, M, et al. Epidemic hepatitis E in Pakistan: patterns of serologic response and evidence that antibody to hepatitis E virus protects against disease. J Infect Dis 1994;170:517.Google Scholar
Bile, K, Isse, A, Mohamud, O, et al. Contrasting roles of rivers and wells as sources of drinking water on attack and fatality rates in a hepatitis E epidemic in Somalia. Am J Trop Med Hyg 1994;51:466–74.Google Scholar
Corwin, AL, Khiem, HB, Clayson, ET, et al. A waterborne outbreak of hepatitis E virus transmission in southwestern Vietnam. Am J Trop Med Hyg 1996;54:559–62.Google Scholar
Khuroo, MS, Dar, MY. Hepatitis E: evidence for person-to-person transmission and inability of low dose immune serum globulin from an Indian source to prevent it. Indian J Gastroenterol 1992;3:113–16.Google Scholar
Tsarev, SA, Tsareva, TS, Emerson, SU, et al. Successful passive and active immunization of cynomolgus monkeys against hepatitis E. Proc Natl Acad Sci USA 1994;91:10198–202.Google Scholar
Tsarev, SA, Tsareva, TS, Emerson, SU, et al. Recombinant vaccine against hepatitis E: dose response and protection against heterologous challenge. Vaccine 1997;15:1834–8.Google Scholar
Kamili, S. Toward the development of a hepatitis E vaccine. Virus Res 2011;161:93100.Google Scholar
Alric, L, Bonnet, D, Laurent, G, et al. Chronic hepatitis E virus infection: successful virologic response to pegylated interferon-alpha therapy. Ann Intern Med 2010;153:135–6.Google Scholar
Kamar, N, Rostaing, L, Abravanel, F, et al. Ribavirin therapy inhibits viral replication in patients with chronic hepatitis E virus infection. Gastroenterology 2010;139:1612–18.Google Scholar
Centers for Disease Control and Prevention. Viral hepatitis surveillance, United States, 2016. Atlanta, GA: Centers for Disease Control and Prevention 2016. Available at: www.cdc.gov/hepatitis/statistics/2016surveillance/pdfs/2016HepSurveillanceRpt.pdf [last accessed June 12, 2020].Google Scholar

References

Ganem, D, Prince, AM. Hepatitis B virus infection natural history and clinical consequences. N Engl J Med 2004;350:1118–29.Google Scholar
World Health Organization Hepatitis B fact-sheets. www.who.int/news-room/fact-sheets/detail/hepatitis-b [last accessed June 15, 2020].Google Scholar
Tiollais, P, Pourcel, C, Dejean, A. The hepatitis B virus. Nature 1985;317:489–95.CrossRefGoogle ScholarPubMed
Nassal, M. Hepatitis B viruses: reverse transcription a different way. Virus Research 2008;134:235–49.Google Scholar
Huang, HC, Chen, CC, Chang, WC, et al. Entry of hepatitis B virus into immortalized human primary hepatocytes by clathrin-dependent endocytosis. J Virol 2012;86(17):9443–53.Google Scholar
Murray, JM, Purcell, R, Wieland, SF. The half-life of hepatitis B virons. Hepatology 2006;44:1117–21.Google Scholar
Balistreri, WF. Viral hepatitis. Emerg Clin North Am 1991;9:365–99.Google Scholar
Hoofnagle, JH, DiBisceglie, AM. Serologic diagnosis of acute and chronic viral hepatitis. Semin Liver Dis 1991;11:7383.Google Scholar
Andersson, KL, Raymond, T, Chung, RT. Monitoring during and after antiviral therapy for hepatitis B. Hepatology 2009;49:S166S173.CrossRefGoogle ScholarPubMed
McQuillan, GM, Coleman, PJ, Kruszon-Moran, D, et al. Prevalence of hepatitis B virus infection in the United States: the National Health and Nutrition Examination Surveys, 1976 through 1994. Am J Public Health. 1999; 89:48.Google Scholar
Roberts, H, Kruszon-Moran, D, Ly, KN, et al. Prevalence of chronic hepatitis B virus (HBV) infection in US households: National Health and Nutrition Examination Survey (NHANES), 1988–2012. Hepatology 2016;63(2):388–97.Google Scholar
Kim, HS, Rotundo, L, Yang, JD, et al. Racial/ethnic disparities in the prevalence and awareness of hepatitis B virus infection and immunity in the United States. J Viral Hepat 2017;24:1052–66.Google Scholar
World Health Organization Hepatitis B fact-sheets. www.who.int/news-room/fact-sheets/detail/hepatitis-b [last accessed June 15, 2020].Google Scholar
Hsu, HY Chang, MH, Chen, DS, et al. Baseline seroepidemiology of hepatitis B virus infection in children in Taipei, 1984: a study just before mass hepatitis B vaccination program in Taiwan. J Med Virol 1986;18:301–7.Google Scholar
Lemoine, M, Thursz, MR. Battle field against hepatitis B infection and HCC in Africa. J Hepatol 2017;66:645–54.Google Scholar
Beasley, RP. Rocks along the road to the control of HBV and HCC. Ann Epidemiol 2009;19:231–4.Google Scholar
Hsu, HY, Chang, MH, Hsieh, KH, et al. Cellular immune response to hepatitis B core antigen in maternal infant transmission of hepatitis B virus. Hepatology 1992;15:770–6.Google Scholar
Lee, SD, Lo, KJ, Tsai, YT, et al. Role of caesarean section in prevention of mother-infant transmission of hepatitis B virus. Lancet 1988;2:833–4.Google Scholar
Tang, JR, Hsu, HY, Lin, HH, et al. Hepatitis B surface antigenemia at birth: a long-term follow-up study. J Pediatr 1998;133:374–7.Google Scholar
Mavilia, MG, Wu, GY. Mechanisms and prevention of vertical transmission in chronic viral hepatitis. J Clin Transl Hepatol 2017;5:119–29.Google Scholar
Beasley, RP, Stevens, CE, Shiao, IS, et al. Evidence against breast-feeding as a mechanism for vertical transmission of hepatitis B. Lancet 1975;2:740–1.Google Scholar
Gartner, LM, Morton, J, Lawrence, RA, et al. Breastfeeding and the use of human milk. Pediatrics 2005;115:496506.Google Scholar
Bhaduri, BR, Mieli-Vergani, G.. Fulminant hepatic failure: pediatric aspects. Semin Liver Dis 1996;16:349–55.Google Scholar
Chang, MH, Lee, CY, Chen, DS, et al. Fulminant hepatitis in children in Taiwan: the important role of hepatitis B virus. J Pediatr 1987;111:34–9.Google Scholar
Hsu, HY, Chang, MH, Lee, CY, et al. Pre-core mutant of hepatitis B virus in childhood fulminant hepatitis B: an infrequent association. J Infect Dis 1995;171:776–81.Google Scholar
Chang, MH, Hsu, HY, Hsu, HC, et al. The significance of spontaneous HBeAg seroconversion in childhood: with special emphasis on the clearance of HBeAg before three years of age. Hepatology 1995;22:1387–92.Google Scholar
Chu, CM, Karayiannis, P, Fowler, MJ, et al. Natural history of chronic hepatitis B virus infection in Taiwan: studies of hepatitis B virus DNA in serum. Hepatology 1985;5:431–4.Google Scholar
Chang, MH, Hwang, LY, Hsu, HC, et al. Prospective study of asymptomatic HBsAg carrier children infected in the perinatal period: clinical and liver histologic studies. Hepatology 1988;8:374–7.Google Scholar
Bortolotti, F, Cadrobbi, M, Crivellaro, C, et al. Long-term outcome of chronic type B hepatitis in patients who acquire hepatitis B virus in infection in childhood. Gastroenterology 1990;99:805–10.CrossRefGoogle ScholarPubMed
Chang, MH, Sung, JL, Lee, CY, et al. Factors affecting the clearance of hepatitis B e antigen in hepatitis B surface antigen carrier children. J Pediatr 1989;115:385–90.Google Scholar
Wu, JF, Su, YR, Chen, CH, et al. Predictive effect of serial serum alanine aminotransferase levels on spontaneous HBeAg seroconversion in chronic genotype B and C HBV-infected children. J Pediatr Gastroenterol Nutr 2012;54:97100.Google Scholar
Ni, YH, Chang, MH, Chen, PJ, et al. Viremia profiles in children with chronic hepatitis B virus infection and spontaneous e antigen seroconversion. Gastroenterology 2007;132:2340–5.Google Scholar
Bortolotti, F, Guido, M, Bartolacci, S, et al. Chronic hepatitis B in children after e antigen seroclearance: final report of a 29-year longitudinal study. Hepatology 2006;43:556–62.Google Scholar
Bortolotti, F, Wirth, S, Crivellaro, C, et al. Long-term persistence of hepatitis B virus DNA in the serum of children with chronic hepatitis B after hepatitis B e antigen to antibody seroconversion. J Pediatr Gastroenterol Nutr 1996;22:270–4.Google Scholar
Wu, JF, Chiu, YC, Chang, KC, et al. Predictors of hepatitis B e antigen-negative hepatitis in chronic hepatitis B infected patients from childhood to adulthood. Hepatology 2016;63:7482.Google Scholar
Liaw, YF. Natural history of chronic hepatitis B virus infection and long-term outcome under treatment. Liver International 2009;29(S1):100–7.Google Scholar
Chiu, YC, Liao, SF, Wu, JF, et al. Factors affecting the natural decay of hepatitis B surface antigen in children with chronic hepatitis B virus infection during long-term follow-up. J Pediatr 2014;165:767–72.Google Scholar
Chu, CM, Liaw, YF. HBsAg seroclearance in asymptomatic carriers of high endemic areas: appreciably high rates during a long-term follow-up. Hepatology 2007;45:1187–92.Google Scholar
Hung, WL, Wu, JF, Ni, YH, et al. Occult hepatitis B virus and surface antigen mutant infection in healthy vaccinated cohorts and children with various forms of hepatitis and multiple transfusions. Liver Int 2019;39:1052–61.Google Scholar
Hsu, HC, Lin, YH, Chang, MH, et al. Pathology of chronic hepatitis B virus infection in children: with special reference to the intrahepatic expression of hepatitis B virus antigens. Hepatology 1988;8:378–82.Google Scholar
Tseng, YR, Wu, JF, Ni, YH, et al. Long-term effect of maternal HBeAg on delayed HBeAg seroconversion in offspring with chronic hepatitis B infection. Liver Int 2011;31:1373–80.Google Scholar
Wu, JF, Wu, TC, Chen, CH, et al. Serum levels of interleukin-12 predict early, spontaneous hepatitis B virus e antigen seroconversiston. Gastroenterology 2010;138:165–72.Google Scholar
Lin, CL, Kao, JH. The clinical implications of hepatitis B virus genotype: recent advances. J Gastroenterol Hepatol 2011;26(Suppl. 1):123–30.Google Scholar
Ni, YH, Chang, MH, Wang, KJ, et al. Clinical relevance of hepatitis B virus genotype in children with chronic hepatitis B and hepatocellular carcinoma. Gastroenterology 2004;127:1733–8.Google Scholar
Chen, CJ, Yang, HI, Iloeje, UH; REVEAL-HBV Study Group Hepatitis B virus DNA levels and outcomes in chronic hepatitis B. Hepatology 2009;49(Suppl. 5):S7284.Google Scholar
Chang, MH, Hsu, HY, Ni, YH, et al. Precore stop codon mutant in chronic hepatitis B virus infection in children: its relation to hepatitis B e seroconversion and natural hepatitis B surface antigen. J Hepatol 1998;28:915–22.Google Scholar
Ni, YH, Chang, MH, Hsu, HY, Tsuei, DJ. Longitudinal study on mutation profiles of core promoter and precore regions of the hepatitis B virus genome in children. Pediatr Res 2004;56:396–9.Google Scholar
Ni, YH, Chang, MH, Hsu, HY, Tsuei, DJ. Different hepatitis B virus core gene mutations in children with chronic infection versus hepatocellular carcinoma. Gut 2003; 52:122–5.CrossRefGoogle Scholar
Jonas, MM, Block, JM, Haber, BA, et al. Treatment of children with chronic hepatitis B virus infection in the United States: patient selection and therapeutic options. Hepatology 2010;52:2192–205.Google Scholar
Terrault, NA, Lok, ASF, McMahon, BJ, et al. Update of prevention, diagnosis, and treatment of chronic hepatitis B: AASLD 2018 hepatitis B guidance. Hepatology 2018;67:1560–99.Google Scholar
Sokal, EM, Paganelli, M, Wirth, S, et al. Management of chronic hepatitis B in childhood: ESPGHAN clinical practice guidelines: consensus of an expert panel on behalf of the European Society of Pediatric Gastroenterology, Hepatology and Nutrition. J Hepatol 2013;59:814–29.Google Scholar
Bisceglie, AM, Lok, AS, Martin, P, et al. Recent US Food and Drug Administration warnings on hepatitis B reactivation with immune-suppressing and anti-cancer drugs: just the tip of the iceberg? Hepatology 2015;61:703–11.Google Scholar
Torre, D, Tambini, R. Interferon-a therapy for chronic hepatitis B in children: a meta-analysis. Clin Infect Dis 1996;23:131–7.Google Scholar
Sokal, EM, Conjeevaram, HS, Roberts, EA, et al. Interferon alfa therapy for chronic hepatitis B in children: a multinational randomized controlled trial. Gastroenterology 1998;114:988–95.Google Scholar
Bortolotti, F, Jara, P, Barbera, C, et al. Long term effect of alpha interferon in children with chronic hepatitis B. Gut 2000;46:715–18.Google Scholar
Hsu, HY, Tsai, HY, Wu, TC, et al. Interferon-alpha treatment in children and young adults with chronic hepatitis B: a long-term follow-up study in Taiwan. Liver Int 2008;28:1288–97.Google Scholar
Sokal, EM, Pagnelli, M, Wirth, S, et al. Management of chronic hepatitis B in childhood: ESPGHAN clinical practice guidelines. J Hepatol 2013;59:814–29.Google Scholar
Wirth, S, Zhang, H, Hardikar, W, et al. Efficacy and safety of peginterferon alfa-2a (40 KD) in children with chronic hepatitis B: the PEG-B-ACTIVE Study. Hepatology 2018;68:1681–94.Google Scholar
Jonas, M, Kelly, DA, Mizerski, J, et al. Clinical trial of lamivudine in children with chronic hepatitis B. N Engl J Med 2002;346:1706–13.Google Scholar
Sokal, EM, Kelly, DA, Badia, IB, et al. Long-term lamivudine therapy for children with HBeAg-positive chronic hepatitis B. Hepatology 2006;43:225–32.Google Scholar
European Association for the Study of the Liver. EASL clinical practice guidelines on the management of chronic hepatitis B virus infection. J Hepatol 2012;57:167–85.Google Scholar
Jonas, MM, Kelly, D, Pollack, H, Mizerski, J, Sorbel, J, Frederick, D, Mondou, E, Rousseau, F, Sokal, E. Safety, efficacy, and pharmacokinetics of adefovir dipivoxil in children and adolescents (age 2 to <8 years) with chronic hepatitis B. Hepatology 2008;47:1863–71.Google Scholar
Jonas, MM, Kelly, D, Pollack, H, et al. Efficacy and safety of long-term adfovir dipivoxil therapy in children with chronic hepatitis B infection. Pediatr Infect Dis J 2012;31:578–82.Google Scholar
Chang, TT, Gish, RG, de Man, R, et al. A comparison of entecavir and lamivudine for HBeAg-posiitve chronic hepatitis B.N Engl J Med 2006;354:1001–10.Google Scholar
Lai, CL, Shouval, D, Lok, AS, et al. Entecavir versus lamivudine for patients with HBeAg-negative chronic Hepatitis B. N Engl J Med 2006;354:1011–20.Google Scholar
Tenney, DJ, Rose, RE, Baldick, CJ, et al. Long-term monitoring shows hepatitis B virus resistance to entecavir in nucleoside-naïve patients is rare through 5 years of therapy. Hepatology 2009;49:1503–14.Google Scholar
Jonas, MM, Chang, MH, Sokal, E, et al. Randomized controlled trial of entecavir vs placebo in children with hepatitis B e antigen positive chronic hepatitis B. Hepatology 2016;63:377–89.Google Scholar
Lai, CL, Gane, E, Liaw, YF, et al. Telbivudine versus lamivudine in patients with chronic hepatitis B. N Engl J Med 2007;357:2576–88.CrossRefGoogle ScholarPubMed
Seto, WK, Lai, CL, Fung, J, Wong, DK, Yuen, JC, Hung, IF, Yuen, MF. Significance of HBV DNA levels at 12 weeks of telbivudine treatment and the 3 years treatment outcome. J Hepatol 2011;55:522–8.Google Scholar
Han, GR, Cao, MK, Zhao, W, et al. A prospective and open-label study for the efficacy and safety of telbivudine in pregnancy for prevention of perinatal transmission of hepatitis B virus infection. J Hepatol 2011;55:1215–21.Google Scholar
Marcellin, P, Heathcote, EJ, Buti, M, et al. Tenofovir disoproxil fumarate versus adefovir dipivoxil for chronic hepatitis B. N Engl J Med 2008;359:2442–55.Google Scholar
Liu, Y, Corsa, AC, Buti, M, et al. No detectable resistance to tenofivir disoproxil fumarate in HBeAg+ and HBeAg- patients with chronic hepatitis B after 8 years of treatment. J Viral Hepat 2017;24:6874.Google Scholar
Murray, KF, Szenborn, L, Wysocki, J, et al. Randomized, placebo-controlled trial of tenofovir disoproxil fumarate in adolescents with chronic hepatitis B. Hepatology 2012;56:2018–26.Google Scholar
D’Antiga, L, Aw, M, Atkins, M, et al. Combined lamivudine/interferon-alpha treatment in “immunotolerant” children perinatally infected with hepatitis B: a pilot study. J Pediatr 2006;148:228–33.Google Scholar
Zhu, S, Zhang, H, Dong, Y, et al. Antiviral therapy in hepatitis B virus-infected children with immune-tolerant characteristics: a pilot open-label randomized study. J Hepatol 2018;68:1123–8.Google Scholar
Rosenthal, P, Ling, SC, Belle, SH, et al. Combination of entecavir/peginterferon alfa-2a in children with HBeAg-positive immune tolerant chronic hepatitis B virus infection. Hepatology 2019;69:2326–37.Google Scholar
Bertoletti, A, Bert, NL. Immunotherapy for chronic hepatitis B virus infection. Gut Liver 2018;12:497507.Google Scholar
Boni, C, Vecchi, A, Rossi, M, et al. TLR7 agonist increases responses of hepatitis B virus-specific T cells and natural killer cells in patients with chronic hepatitis B treated with nucleos(t)ide analogues. Gastroenterology 2018;154:1764–77.Google Scholar
Ji, C, Sastry, KS, Tiefenthaler, G, et al. Targeted delivery of interferon-alpha to hepatitis B virus-infected cells using T-cell receptor-like antibodies. Hepatology 2012;56:2027–38.Google Scholar
Fisicaro, P, Valdatta, C, Massari, M, et al. Antiviral intrahepatic T-cell responses can be restored by blocking programmed death-1 pathway in chronic hepatitis B. Gastroenterology 2010;138:682–93.Google Scholar
Serigado, JM, Izzy, M, Kalia, H. Novel therapies and potential therapeutic targets in the management of chronic hepatitis B. Europ J Gastro Hepatol 2017;29:987–93.Google Scholar
Chen, HL, Lin, LH, Hu, FC, et al. Effects of maternal screening and universal immunization to prevent mother-to-infant transmission of HBV. Gastroenterology 2012;142:773–81.Google Scholar
Chen, DS, Hsu, NH, Sung, JL, et al. A mass vaccination program in Taiwan against hepatitis B virus infection in infants of hepatitis B surface antigen-carrier mothers. JAMA 1987;257:2597–603.Google Scholar
MacGregor, A. WHO: World Health Assembly. Lancet 1992;339(8804):1287.Google Scholar
World Health Organization Hepatitis B fact-sheets. www.who.int/news-room/fact-sheets/detail/hepatitis-b [last accessed June 15, 2020].Google Scholar
Sagnelli, E, Sagnelli, C, Pisaturo, M, et al. Epidemiology of acute and chronic hepatitis B and delta over the last 5 decades in Italy. World J Gastroenterol 2014;20(24):7635–43.Google Scholar
Schillie, S, Vellozzi, C, Reingold, A, et al. Prevention of hepatitis B virus infection in the United States: recommendations of the Advisory Committee on Immunization Practices. MMWR 2018;67(1):131.Google Scholar
Ni, YH, Chang, MH, Jan, CF, et al. Continuing decrease in hepatitis B virus infection 30 years after initiation of infant vaccination program in Taiwan. Clin Gastro Hepatol 2016;14:1324–30.Google Scholar
Hsu, HY, Chang, MH, Ni, YH, et al. Universal infant immunization and occult hepatitis B virus infection in children and adolescents: a population-based study. Hepatology 2015;61:1183–91.Google Scholar
Peto, TJ, Mendy, ME, Lowe, Y, et al. Efficacy and effectiveness of infant vaccination against chronic hepatitis B in the Gambia Hepatitis Intervention Study (1986–90) and in the nationwide immunization program. BMC Infect Dis 2014;14:7.Google Scholar
Chen, HL, Chang, CJ, Kong, MS, et al. Fulminant hepatitis failure in children in endemic areas of hepatitis B virus infection: 15 years after universal hepatitis B vaccination. Hepatology 2004;39:5863.Google Scholar
Chang, MH, Chen, CJ, Lai, MS, et al. Universal hepatitis B vaccination in Taiwan and the incidence of hepatocellular carcinoma in children. N Engl J Med 1997;336:1855–9.CrossRefGoogle ScholarPubMed
Chang, MH, You, SL, Chen, CJ, et al. Decreased incidence of hepatocellular carcinoma in hepatitis B vaccines: a 20-year follow-up study. J Natl Cancer Inst 2009;101:1348–55.Google Scholar
Chang, MH, You, SL, Chen, CJ, et al. Long-term effect of hepatitis B vaccination of infants in preventing liver cancer. Gastroenterology 2016;151:472–80.Google Scholar
Su, WJ, Ho, MC, Ni, YH, et al. High-titer antibody to hepatitis B surface antigen before liver transplantation can prevent de novo hepatitis B infection. J Pediatr Gastroenterol Nutr 2009;48:203–8.Google Scholar
Wen, WH, Chang, MH, Zhao, LL, et al. Mother-to-infant transmission of hepatitis B virus infection: significance of maternal viral load and strategies for investigation. J Hepatol 2013;59:2430.Google Scholar
Chen, HL, Lee, CN, Chang, CH, et al. Efficacy of maternal tenofivir disoproxil fumarate in interrupting mother-to-infant transmission of hepatitis B virus. Hepatology 2015;62:375–86.Google Scholar
Pan, CQ, Duan, Z, Dai, E, et al. Tenofovir to prevent hepatitis B virus transmission in mothers with high viral load. N Eng J Med 2016;374:2324–34.Google Scholar
Hsu, HY, Chang, MH, Ni, YH, et al. Chronologic changes in serum hepatitis virus DNA, genotypes, surface antigen mutants and transcriptase mutants during 25-year nationwide immunization in Taiwan. J Viral Hepat 2017;24:645–53.Google Scholar
Thio, CL, Guo, N, Xie, C, et al. Global elimination of mother-to-child transmission of hepatitis B: revisiting the current strategies. Lancet Infect Dis 2015;15:981–5.Google Scholar
Feret, E, Larouze, B, Diop, B, et al. Epidemiology of hepatitis B virus infection in the rural community of Tip, Senegal. Am J Epidemiol. 1987;125:140–9.Google Scholar
Schweitzer, A, Horn, J, Mikolajczyk, RT, Krause, G, and Ott, JJ. Estimations of worldwide prevalence of chronic hepatitis B virus infection: a systematic review of data published between 1965 and 2013. Lancet 2015;386:1546–55.Google Scholar

References

Mack, CL, Gonzalez-Peralta, RP, Gupta, N, Leung, D, Narkewicz, MR, Roberts, EA, Rosenthal, P, et al. NASPGHAN practice guidelines: diagnosis and management of hepatitis C infection in infants, children, and adolescents. J Pediatr Gastroenterol Nutr 2012;54:838–55.Google Scholar
Bukh, J. The history of hepatitis C virus (HCV): basic research reveals unique features in phylogeny, evolution and the viral life cycle with new perspectives for epidemic control. J Hepatol 2016;65:S2S21.Google Scholar
Bartlett, C, Curd, A, Peckham, M, Harris, M. Visualisation and analysis of hepatitis C virus non-structural proteins using super-resolution microscopy. Sci Rep 2018;8:13604.Google Scholar
Pileri, P, Uematsu, Y, Campagnoli, S, Galli, G, Falugi, F, Petracca, R, Weiner, AJ, et al. Binding of hepatitis C virus to CD81. Science 1998;282:938–41.Google Scholar
Urbanowicz, A, Zagozdzon, R, Ciszek, M. Modulation of the immune system in chronic hepatitis C and during antiviral interferon-free therapy. Arch Immunol Ther Exp (Warsz) 2019;67(2):7988.Google Scholar
Patra, T, Ray, RB, Ray, R. Strategies to circumvent host innate immune response by hepatitis C virus. Cells 2019;8(3):274.Google Scholar
Ciuffreda, D, Kim, AY. Update on hepatitis C virus-specific immunity. Curr Opin HIV AIDS 2011;6:559–65.Google Scholar
Heim, MH. Innate immunity and HCV. J Hepatol 2013;58:564–74.Google Scholar
Dolganiuc, A, Chang, S, Kodys, K, Mandrekar, P, Bakis, G, Cormier, M, Szabo, G. Hepatitis C virus (HCV) core protein-induced, monocyte-mediated mechanisms of reduced IFN-alpha and plasmacytoid dendritic cell loss in chronic HCV infection. J Immunol 2006;177:6758–68.Google Scholar
Shiina, M, Rehermann, B. Cell culture-produced hepatitis C virus impairs plasmacytoid dendritic cell function. Hepatology 2008;47:385–95.Google Scholar
Tseng, CT, Klimpel, GR. Binding of the hepatitis C virus envelope protein E2 to CD81 inhibits natural killer cell functions. J Exp Med 2002;195:43–9.Google Scholar
Ali, OS, Abo-Shadi, MA, Hammad, LN. The biological significance of serum complements C3 and C4 in HCV-related chronic liver diseases and hepatocellular carcinoma. Egypt J Immunol 2005;12:91–9.Google Scholar
Dumestre-Perard, C, Ponard, D, Drouet, C, Leroy, V, Zarski, JP, Dutertre, N, Colomb, MG. Complement C4 monitoring in the follow-up of chronic hepatitis C treatment. Clin Exp Immunol 2002;127:131–6.Google Scholar
Mazumdar, B, Kim, H, Meyer, K, Bose, SK, Di Bisceglie, AM, Ray, RB, Diamond, MS, et al. Hepatitis C virus infection upregulates CD55 expression on the hepatocyte surface and promotes association with virus particles. J Virol 2013;87:7902–10.Google Scholar
Gower, E, Estes, C, Blach, S, Razavi-Shearer, K, Razavi, H. Global epidemiology and genotype distribution of the hepatitis C virus infection. J Hepatol 2014;61:S4557.Google Scholar
El-Shabrawi, MH, Kamal, NM. Burden of pediatric hepatitis C. World J Gastroenterol 2013;19:7880–8.Google Scholar
Delgado-Borrego, A, Smith, L, Jonas, MM, Hall, CA, Negre, B, Jordan, SH, Ogrodowicz, M, et al. Expected and actual case ascertainment and treatment rates for children infected with hepatitis C in Florida and the United States: epidemiologic evidence from statewide and nationwide surveys. J Pediatr 2012;161:915–21.Google Scholar
Raynes-Greenow, C, Polis, S, Elliott, E, Hardikar, W, Kesson, A, Kaldor, J, Jones, CA. Childhood hepatitis C virus infection: an Australian national surveillance study of incident cases over five years. J Paediatr Child Health 2015;51:1115–20.Google Scholar
Razavi, H, Elkhoury, AC, Elbasha, E, Estes, C, Pasini, K, Poynard, T, Kumar, R. Chronic hepatitis C virus (HCV) disease burden and cost in the United States. Hepatology 2013;57:2164–70.Google Scholar
Kuiken, C, Simmonds, P. Nomenclature and numbering of the hepatitis C virus. Methods Mol Biol 2009;510:3353.Google Scholar
Liakina, V, Hamid, S, Tanaka, J, Olafsson, S, Sharara, AI, Alavian, SM, Gheorghe, L, et al. Historical epidemiology of hepatitis C virus (HCV) in select countries – volume 3. J Viral Hepat 2015;22( Suppl 4):420.Google Scholar
Lowe, NJ, Rizk, D, Grimes, P, Billips, M, Pincus, S. Azelaic acid 20% cream in the treatment of facial hyperpigmentation in darker-skinned patients. Clin Ther 1998;20:945–59.Google Scholar
Schwarz, KB, Gonzalez-Peralta, RP, Murray, KF, Molleston, JP, Haber, BA, Jonas, MM, Rosenthal, P, et al. The combination of ribavirin and peginterferon is superior to peginterferon and placebo for children and adolescents with chronic hepatitis C. Gastroenterology 2011;140:450–8 e451.Google Scholar
Prevention CfDCa. Viral hepatitis statistics and surveillance. 2013.Google Scholar
Lalloum, F, Souied, EH, Bastuji-Garin, S, Puche, N, Querques, G, Glacet-Bernard, A, Coscas, G, et al. Intravitreal ranibizumab for choroidal neovascularization complicating pathologic myopia. Retina 2010;30:399406.Google Scholar
Benova, L, Mohamoud, YA, Calvert, C, Abu-Raddad, LJ. Vertical transmission of hepatitis C virus: systematic review and meta-analysis. Clin Infect Dis 2014;59:765–73.Google Scholar
Dal Molin, G, D’Agaro, P, Ansaldi, F, Ciana, G, Fertz, C, Alberico, S, Campello, C. Mother-to-infant transmission of hepatitis C virus: rate of infection and assessment of viral load and IgM anti-HCV as risk factors. J Med Virol 2002;67:137–42.Google Scholar
European Paediatric Hepatitis CVN. A significant sex–but not elective cesarean section–effect on mother-to-child transmission of hepatitis C virus infection.J Infect Dis 2005;192:1872–9.Google Scholar
Mast, EE, Hwang, LY, Seto, DS, Nolte, FS, Nainan, OV, Wurtzel, H, Alter, MJ. Risk factors for perinatal transmission of hepatitis C virus (HCV) and the natural history of HCV infection acquired in infancy. J Infect Dis 2005;192:1880–9.Google Scholar
Wen, JW, Haber, BA. Maternal-fetal transmission of hepatitis C infection: what is so special about babies? J Pediatr Gastroenterol Nutr 2014;58:278–82.Google Scholar
Garcia-Tejedor, A, Maiques-Montesinos, V, Diago-Almela, VJ, Pereda-Perez, A, Alberola-Cunat, V, Lopez-Hontangas, JL, Perales-Puchalt, A, et al. Risk factors for vertical transmission of hepatitis C virus: a single center experience with 710 HCV-infected mothers. Eur J Obstet Gynecol Reprod Biol 2015;194:173–7.Google Scholar
Conte, D, Fraquelli, M, Prati, D, Colucci, A, Minola, E. Prevalence and clinical course of chronic hepatitis C virus (HCV) infection and rate of HCV vertical transmission in a cohort of 15,250 pregnant women. Hepatology 2000;31:751–5.Google Scholar
Resti, M, Azzari, C, Mannelli, F, Moriondo, M, Novembre, E, de Martino, M, Vierucci, A. Mother to child transmission of hepatitis C virus: prospective study of risk factors and timing of infection in children born to women seronegative for HIV-1. Tuscany Study Group on Hepatitis C Virus Infection. BMJ 1998;317:437–41.Google Scholar
Fauteux-Daniel, S, Larouche, A, Calderon, V, Boulais, J, Beland, C, Ransy, DG, Boucher, M, et al. Vertical transmission of hepatitis C virus: variable transmission bottleneck and evidence of midgestation in utero infection. J Virol 2017;91(23):e01372–17.Google Scholar
Suryaprasad, AG, White, JZ, Xu, F, Eichler, BA, Hamilton, J, Patel, A, Hamdounia, SB, et al. Emerging epidemic of hepatitis C virus infections among young nonurban persons who inject drugs in the United States, 2006–2012. Clin Infect Dis 2014;59:1411–19.Google Scholar
Kuncio, DE, Newbern, EC, Johnson, CC, Viner, KM. Failure to test and identify perinatally infected children born to hepatitis C virus-infected women. Clin Infect Dis 2016;62:980–5.Google Scholar
Chappell, CA, Hillier, SL, Crowe, D, Meyn, LA, Bogen, DL, Krans, EE. Hepatitis C virus screening among children exposed during pregnancy. Pediatrics 2018;141(6):e20173273.Google Scholar
American College of Gynecologists. ACOG Practice Bulletin No. 86: Viral hepatitis in pregnancy. Obstet Gynecol 2007;110:941–56.Google Scholar
Kanninen, TT, Dieterich, D, Asciutti, S. HCV vertical transmission in pregnancy: new horizons in the era of DAAs. Hepatology 2015;62:1656–8.Google Scholar
Selvapatt, N, Ward, T, Bailey, H, Bennett, H, Thorne, C, See, LM, Tudor-Williams, G, et al. Is antenatal screening for hepatitis C virus cost-effective? A decade’s experience at a London centre. J Hepatol 2015;63:797804.Google Scholar
Tasillo, A, Eftekhari Yazdi, G, Nolen, S, Schillie, S, Vellozzi, C, Epstein, R, Randall, L, et al. Short-term effects and long-term cost-effectiveness of universal hepatitis C testing in prenatal care. Obstet Gynecol 2019;133:289300.Google Scholar
Centers for Disease Control and Prevention. Hepatitis C virus infection among adolescents and young adults: Massachusetts, 2002–2009. MMWR Morb Mortal Wkly Rep 2011;60:537–41.Google Scholar
Zibbell, JE, Iqbal, K, Patel, RC, Suryaprasad, A, Sanders, KJ, Moore-Moravian, L, Serrecchia, J, et al. Increases in hepatitis C virus infection related to injection drug use among persons aged <30 years – Kentucky, Tennessee, Virginia, and West Virginia, 2006–2012. MMWR Morb Mortal Wkly Rep 2015;64:453–8.Google Scholar
Barritt, ASt, Lee, B, Runge, T, Schmidt, M, Jhaveri, R. Increasing prevalence of hepatitis C among hospitalized children is associated with an increase in substance abuse. J Pediatr 2018;192:159–64.Google Scholar
Abutaleb, A, Sherman, KE. A changing paradigm: management and treatment of the HCV/HIV-co-infected patient. Hepatol Int 2018;12:500–9.Google Scholar
Mallet, V, Vallet-Pichard, A, Pol, S. The impact of human immunodeficiency virus on viral hepatitis. Liver Int 2011;31(Suppl 1):135–9.Google Scholar
Indolfi, G, Bartolini, E, Serranti, D, Azzari, C, Resti, M. Hepatitis C in children co-infected with human immunodeficiency virus. J Pediatr Gastroenterol Nutr 2015;61:393–9.Google Scholar
Ciotti, M, D’Agostini, C, Marrone, A. Advances in the diagnosis and monitoring of hepatitis C virus infection. Gastroenterology Res 2013;6:161–70.Google Scholar
de Leuw, P, Sarrazin, C, Zeuzem, S. How to use virological tools for the optimal management of chronic hepatitis C. Liver Int 2011;31(Suppl 1):312.Google Scholar
Polywka, S, Pembrey, L, Tovo, PA, Newell, ML. Accuracy of HCV-RNA PCR tests for diagnosis or exclusion of vertically acquired HCV infection. J Med Virol 2006;78:305–10.Google Scholar
Easterbrook, PJ, Group WHOGD. Who to test and how to test for chronic hepatitis C infection – 2016 WHO testing guidance for low- and middle-income countries. J Hepatol 2016;65:S4666.Google Scholar
Jonas, MM, Baron, MJ, Bresee, JS, Schneider, LC. Clinical and virologic features of hepatitis C virus infection associated with intravenous immunoglobulin. Pediatrics 1996;98:211–15.Google Scholar
Squires, RH Jr., Shneider, BL, Bucuvalas, J, Alonso, E, Sokol, RJ, Narkewicz, MR, Dhawan, A, et al. Acute liver failure in children: the first 348 patients in the Pediatric Acute Liver Failure Study Group. J Pediatr 2006;148:652–8.Google Scholar
El-Guindi, MA. Hepatitis C viral infection in children: updated review. Pediatr Gastroenterol Hepatol Nutr 2016;19:8395.Google Scholar
Santantonio, T, Sinisi, E, Guastadisegni, A, Casalino, C, Mazzola, M, Gentile, A, Leandro, G, et al. Natural course of acute hepatitis C: a long-term prospective study. Dig Liver Dis 2003;35:104–13.Google Scholar
Santantonio, T, Wiegand, J, Gerlach, JT. Acute hepatitis C: current status and remaining challenges. J Hepatol 2008;49:625–33.Google Scholar
Narkewicz, MR, Horslen, S, Hardison, RM, Shneider, BL, Rodriguez-Baez, N, Alonso, EM, Ng, VL, et al. A learning collaborative approach increases specificity of diagnosis of acute liver failure in pediatric patients. Clin Gastroenterol Hepatol 2018;16:1801–10 e1803.Google Scholar
Davison, SM, Mieli-Vergani, G, Sira, J, Kelly, DA. Perinatal hepatitis C virus infection: diagnosis and management. Arch Dis Child 2006;91:781–5.Google Scholar
Mohan, N, Gonzalez-Peralta, RP, Fujisawa, T, Chang, MH, Heller, S, Jara, P, Kelly, D, et al. Chronic hepatitis C virus infection in children. J Pediatr Gastroenterol Nutr 2010;50:123–31.Google Scholar
European Paediatric Hepatitis CVN. Three broad modalities in the natural history of vertically acquired hepatitis C virus infection. Clin Infect Dis 2005;41:4551.Google Scholar
Goodman, ZD, Makhlouf, HR, Liu, L, Balistreri, W, Gonzalez-Peralta, RP, Haber, B, Jonas, MM, et al. Pathology of chronic hepatitis C in children: liver biopsy findings in the Peds-C Trial. Hepatology 2008;47:836–43.Google Scholar
Turkova, A, Volynets, GV, Crichton, S, Skvortsova, TA, Panfilova, VN, Rogozina, NV, Khavkin, AI, et al. Advanced liver disease in Russian children and adolescents with chronic hepatitis C. J Viral Hepat 2019;26(7):881–92.Google Scholar
Mizuochi, T, Takano, T, Yanagi, T, Ushijima, K, Suzuki, M, Miyoshi, Y, Ito, Y, et al. Epidemiologic features of 348 children with hepatitis C virus infection over a 30-year period: a nationwide survey in Japan. J Gastroenterol 2018;53:419–26.Google Scholar
Squires, JE, Balistreri, WF. Hepatitis C virus infection in children and adolescents. Hepatol Commun 2017;1:8798.Google Scholar
Bunchorntavakul, C, Jones, LM, Kikuchi, M, Valiga, ME, Kaplan, DE, Nunes, FA, Aytaman, A, et al. Distinct features in natural history and outcomes of acute hepatitis C. J Clin Gastroenterol 2015;49:e3140.Google Scholar
Yeung, LT, To, T, King, SM, Roberts, EA. Spontaneous clearance of childhood hepatitis C virus infection. J Viral Hepat 2007;14:797805.Google Scholar
Loomba, R, Rivera, MM, McBurney, R, Park, Y, Haynes-Williams, V, Rehermann, B, Alter, HJ, et al. The natural history of acute hepatitis C: clinical presentation, laboratory findings and treatment outcomes. Aliment Pharmacol Ther 2011;33:559–65.Google Scholar
Maheshwari, A, Ray, S, Thuluvath, PJ. Acute hepatitis C. Lancet 2008;372:321–32.Google Scholar
Resti, M, Bortolotti, F, Vajro, P, Maggiore, G. The Committee of Hepatology of the Italian Society of Pediatric Gastroenterology and Hepatology. Guidelines for the screening and follow-up of infants born to anti-HCV positive mothers. Dig Liver Dis 2003;35:453–7.Google Scholar
Thomas, DL, Astemborski, J, Rai, RM, Anania, FA, Schaeffer, M, Galai, N, Nolt, K, et al. The natural history of hepatitis C virus infection: host, viral, and environmental factors. JAMA 2000;284:450–6.Google Scholar
Villano, SA, Vlahov, D, Nelson, KE, Cohn, S, Thomas, DL. Persistence of viremia and the importance of long-term follow-up after acute hepatitis C infection. Hepatology 1999;29:908–14.Google Scholar
Wang, CC, Krantz, E, Klarquist, J, Krows, M, McBride, L, Scott, EP, Shaw-Stiffel, T, et al. Acute hepatitis C in a contemporary US cohort: modes of acquisition and factors influencing viral clearance. J Infect Dis 2007;196:1474–82.Google Scholar
Indolfi, G, Mangone, G, Bartolini, E, Moriondo, M, Azzari, C, Resti, M. Hepatitis C viraemia after apparent spontaneous clearance in a vertically infected child. Lancet 2016;387:1967–8.Google Scholar
Iorio, R, Giannattasio, A, Sepe, A, Terracciano, LM, Vecchione, R, Vegnente, A. Chronic hepatitis C in childhood: an 18-year experience. Clin Infect Dis 2005;41:1431–7.Google Scholar
Sheiko, MA, Golden-Mason, L, Giugliano, S, Hurtado, CW, Mack, CL, Narkewicz, MR, Rosen, HR. CD4+ and CD8+ T cell activation in children with hepatitis C. J Pediatr 2016;170:142–8 e141.Google Scholar
Garazzino, S, Calitri, C, Versace, A, Alfarano, A, Scolfaro, C, Bertaina, C, Vatrano, S, et al. Natural history of vertically acquired HCV infection and associated autoimmune phenomena. Eur J Pediatr 2014;173:1025–31.Google Scholar
Tovo, PA, Calitri, C, Scolfaro, C, Gabiano, C, Garazzino, S. Vertically acquired hepatitis C virus infection: correlates of transmission and disease progression. World J Gastroenterol 2016;22:1382–92.Google Scholar
Arafa, N, El Hoseiny, M, Rekacewicz, C, Bakr, I, El-Kafrawy, S, El Daly, M, Aoun, S, et al. Changing pattern of hepatitis C virus spread in rural areas of Egypt. J Hepatol 2005;43:418–24.Google Scholar
Farmand, S, Wirth, S, Loffler, H, Woltering, T, Kenzel, S, Lainka, E, Henneke, P. Spontaneous clearance of hepatitis C virus in vertically infected children. Eur J Pediatr 2012;171:253–8.Google Scholar
Serra, MA, Escudero, A, Rodriguez, F, del Olmo, JA, Rodrigo, JM. Effect of hepatitis C virus infection and abstinence from alcohol on survival in patients with alcoholic cirrhosis. J Clin Gastroenterol 2003;36:170–4.Google Scholar
Pearlman, BL, Traub, N. Sustained virologic response to antiviral therapy for chronic hepatitis C virus infection: a cure and so much more. Clin Infect Dis 2011;52:889900.Google Scholar
Indolfi, G, Mangone, G, Bartolini, E, Nebbia, G, Calvo, PL, Moriondo, M, Tovo, PA, et al. Comparative analysis of rs12979860 SNP of the IFNL3 gene in children with hepatitis C and ethnic matched controls using 1000 Genomes Project data. PLoS One 2014;9:e85899.Google Scholar
Indolfi, G, Mangone, G, Calvo, PL, Bartolini, E, Regoli, M, Serranti, D, Calitri, C, et al. Interleukin 28B rs12979860 single-nucleotide polymorphism predicts spontaneous clearance of hepatitis C virus in children. J Pediatr Gastroenterol Nutr 2014;58:666–8.Google Scholar
Ruiz-Extremera, A, Pavon-Castillero, EJ, Florido, M, Munoz de Rueda, P, Munoz-Gamez, JA, Casado, J, Carazo, A, et al. Influence of HLA class I, HLA class II and KIRs on vertical transmission and chronicity of hepatitis C virus in children. PLoS One 2017;12:e0172527.Google Scholar
Nafady, A, Nafady-Hego, H, Abdelwahab, NM, Eltellawy, RHN, Abu Faddan, NH. Peripheral lymphocytes analyses in children with chronic hepatitis C virus infection. Eur J Clin Invest 2018;48:e13004.Google Scholar
Bortolotti, F, Verucchi, G, Camma, C, Cabibbo, G, Zancan, L, Indolfi, G, Giacchino, R, et al. Long-term course of chronic hepatitis C in children: from viral clearance to end-stage liver disease. Gastroenterology 2008;134:1900–7.Google Scholar
Jonas, MM. Children with hepatitis C. Hepatology 2002;36:S173–8.Google Scholar
Lee, CK, Jonas, MM. Hepatitis C: issues in children. Gastroenterol Clin North Am 2015;44:901–9.Google Scholar
Zein, NN. Hepatitis C in children: recent advances. Curr Opin Pediatr 2007;19:570–4.Google Scholar
Casiraghi, MA, De Paschale, M, Romano, L, Biffi, R, Assi, A, Binelli, G, Zanetti, AR. Long-term outcome (35 years) of hepatitis C after acquisition of infection through mini transfusions of blood given at birth. Hepatology 2004;39:90–6.Google Scholar
Modin, L, Arshad, A, Wilkes, B, Benselin, J, Lloyd, C, Irving, WL, Kelly, DA. Epidemiology and natural history of hepatitis C virus infection among children and young people. J Hepatol 2019;70:371–8.Google Scholar
Birnbaum, AH, Shneider, BL, Moy, L. Hepatitis C in children. N Engl J Med 2000;342:290–1.Google Scholar
Guido, M, Bortolotti, F, Leandro, G, Jara, P, Hierro, L, Larrauri, J, Barbera, C, et al. Fibrosis in chronic hepatitis C acquired in infancy: is it only a matter of time? Am J Gastroenterol 2003;98:660–3.Google Scholar
Matsuoka, S, Tatara, K, Hayabuchi, Y, Taguchi, Y, Mori, K, Honda, H, Itou, S, et al. Serologic, virologic, and histologic characteristics of chronic phase hepatitis C virus disease in children infected by transfusion. Pediatrics 1994;94:919–22.Google Scholar
Minola, E, Prati, D, Suter, F, Maggiolo, F, Caprioli, F, Sonzogni, A, Fraquelli, M, et al. Age at infection affects the long-term outcome of transfusion-associated chronic hepatitis C. Blood 2002;99:4588–91.Google Scholar
Stallings-Smith, S, Krull, KR, Brinkman, TM, Hudson, MM, Ojha, RP. Long-term follow-up for incident cirrhosis among pediatric cancer survivors with hepatitis C virus infection. J Clin Virol 2015;71:1821.Google Scholar
Maasoumy, B, Wiegand, SB, Jaroszewicz, J, Bremer, B, Lehmann, P, Deterding, K, Taranta, A, et al. Hepatitis B core-related antigen (HBcrAg) levels in the natural history of hepatitis B virus infection in a large European cohort predominantly infected with genotypes A and D. Clin Microbiol Infect 2015;21:606 e601610.Google Scholar
Cesaro, S, Bortolotti, F, Petris, MG, Brugiolo, A, Guido, M, Carli, M. An updated follow-up of chronic hepatitis C after three decades of observation in pediatric patients cured of malignancy. Pediatr Blood Cancer 2010;55:108–12.Google Scholar
Dyal, HK, Aguilar, M, Bhuket, T, Liu, B, Holt, EW, Torres, S, Cheung, R, et al. Concurrent obesity, diabetes, and steatosis increase risk of advanced fibrosis among HCV patients: a systematic review. Dig Dis Sci 2015;60:2813–24.Google Scholar
Beech, BM, Myers, L, Beech, DJ. Hepatitis B and C infections among homeless adolescents. Fam Community Health 2002;25:2836.Google Scholar
Murray, KF, Richardson, LP, Morishima, C, Owens, JW, Gretch, DR. Prevalence of hepatitis C virus infection and risk factors in an incarcerated juvenile population: a pilot study. Pediatrics 2003;111:153–7.Google Scholar
Page, K, Hahn, JA, Evans, J, Shiboski, S, Lum, P, Delwart, E, Tobler, L, et al. Acute hepatitis C virus infection in young adult injection drug users: a prospective study of incident infection, resolution, and reinfection. J Infect Dis 2009;200:1216–26.Google Scholar
Wise, M, Finelli, L, Sorvillo, F. Prognostic factors associated with hepatitis C disease: a case-control study utilizing U.S. multiple-cause-of-death data. Public Health Rep 2010;125:414–22.Google Scholar
Anon. Hepatitis C: global ambition, national realities. Lancet 2016;387:1970.Google Scholar
Gonzalez-Peralta, RP, Langham, MR Jr., Andres, JM, Mohan, P, Colombani, PM, Alford, MK, Schwarz, KB. Hepatocellular carcinoma in 2 young adolescents with chronic hepatitis C. J Pediatr Gastroenterol Nutr 2009;48:630–5.Google Scholar
Jara, P, Resti, M, Hierro, L, Giacchino, R, Barbera, C, Zancan, L, Crivellaro, C, et al. Chronic hepatitis C virus infection in childhood: clinical patterns and evolution in 224 white children. Clin Infect Dis 2003;36:275–80.Google Scholar
Rumbo, C, Fawaz, RL, Emre, SH, Suchy, FJ, Kerkar, N, Morotti, RA, Shneider, BL. Hepatitis C in children: a quaternary referral center perspective. J Pediatr Gastroenterol Nutr 2006;43:209–16.Google Scholar
Badizadegan, K, Jonas, MM, Ott, MJ, Nelson, SP, Perez-Atayde, AR. Histopathology of the liver in children with chronic hepatitis C viral infection. Hepatology 1998;28:1416–23.Google Scholar
Cacoub, P, Buggisch, P, Carrion, JA, Cooke, GS, Zignego, AL, Beckerman, R, Younossi, Z. Direct medical costs associated with the extrahepatic manifestations of hepatitis C infection in Europe. J Viral Hepat 2018;25:811–17.Google Scholar
Younossi, Z, Park, H, Henry, L, Adeyemi, A, Stepanova, M. Extrahepatic manifestations of hepatitis C: a meta-analysis of prevalence, quality of life, and economic burden. Gastroenterology 2016;150:15991608.Google Scholar
Zignego, AL, Ferri, C, Pileri, SA, Caini, P, Bianchi, FB. Italian Association of the Study of Liver Commission on Extrahepatic Manifestations of HCVi. Extrahepatic manifestations of hepatitis C virus infection: a general overview and guidelines for a clinical approach. Dig Liver Dis 2007;39:217.Google Scholar
Matsumoto, S, Nakajima, S, Nakamura, K, Etani, Y, Hirai, H, Shimizu, N, Yokoyama, H, et al. Interferon treatment on glomerulonephritis associated with hepatitis C virus. Pediatr Nephrol 2000;15:271–3.Google Scholar
Romas, E, Power, DA, Machet, D, Powell, H, d’Apice, AJ. Membranous glomerulonephritis associated with hepatitis C virus infection in an adolescent. Pathology 1994;26:399402.Google Scholar
Sugiura, T, Yamada, T, Kimpara, Y, Fujita, N, Goto, K, Koyama, N. Effects of pegylated interferon alpha-2a on hepatitis-C-virus-associated glomerulonephritis. Pediatr Nephrol 2009;24:199202.Google Scholar
Gehring, S, Kullmer, U, Koeppelmann, S, Gerner, P, Wintermeyer, P, Wirth, S. Prevalence of autoantibodies and the risk of autoimmune thyroid disease in children with chronic hepatitis C virus infection treated with interferon-alpha. World J Gastroenterol 2006;12:5787–92.Google Scholar
Indolfi, G, Bartolini, E, Olivito, B, Azzari, C, Resti, M. Autoimmunity and extrahepatic manifestations in treatment-naive children with chronic hepatitis C virus infection. Clin Dev Immunol 2012;2012:785627.Google Scholar
Indolfi, G, Stagi, S, Bartolini, E, Salti, R, de Martino, M, Azzari, C, Resti, M. Thyroid function and anti-thyroid autoantibodies in untreated children with vertically acquired chronic hepatitis C virus infection. Clin Endocrinol (Oxf) 2008;68:117–21.Google Scholar
Bortolotti, F, Vajro, P, Balli, F, Giacchino, R, Crivellaro, C, Barbera, C, Cataleta, M, et al. Non-organ specific autoantibodies in children with chronic hepatitis C. J Hepatol 1996;25:614–20.Google Scholar
Gregorio, GV, Pensati, P, Iorio, R, Vegnente, A, Mieli-Vergani, G, Vergani, D. Autoantibody prevalence in children with liver disease due to chronic hepatitis C virus (HCV) infection. Clin Exp Immunol 1998;112:471–6.Google Scholar
Muratori, P, Muratori, L, Verucchi, G, Attard, L, Bianchi, FB, Lenzi, M. Non-organ-specific autoantibodies in children with chronic hepatitis C: clinical significance and impact on interferon treatment. Clin Infect Dis 2003;37:1320–6.Google Scholar
Bogdanos, DP, Mieli-Vergani, G, Vergani, D. Non-organ-specific autoantibodies in hepatitis C virus infection: do they matter? Clin Infect Dis 2005;40:508–10.Google Scholar
Delgado-Borrego, A, Jordan, SH, Negre, B, Healey, D, Lin, W, Kamegaya, Y, Christofi, M, et al. Reduction of insulin resistance with effective clearance of hepatitis C infection: results from the HALT-C trial. Clin Gastroenterol Hepatol 2010;8:458–62.Google Scholar
Akhter, A, Said, A. Cutaneous manifestations of viral hepatitis. Curr Infect Dis Rep 2015;17:452.Google Scholar
Nydegger, A, Srivastava, A, Wake, M, Smith, AL, Hardikar, W. Health-related quality of life in children with hepatitis C acquired in the first year of life. J Gastroenterol Hepatol 2008;23:226–30.Google Scholar
Rodrigue, JR, Balistreri, W, Haber, B, Jonas, MM, Mohan, P, Molleston, JP, Murray, KF, et al. Impact of hepatitis C virus infection on children and their caregivers: quality of life, cognitive, and emotional outcomes. J Pediatr Gastroenterol Nutr 2009;48:341–7.Google Scholar
Boerekamps, A, De Weggheleire, A, van den Berk, GE, Lauw, FN, Claassen, MAA, Posthouwer, D, Bierman, WF, et al. Treatment of acute hepatitis C genotypes 1 and 4 with 8 weeks of grazoprevir plus elbasvir (DAHHS2): an open-label, multicentre, single-arm, phase 3b trial. Lancet Gastroenterol Hepatol 2019;4:269–77.Google Scholar
Misra, S, Dieterich, DT, Saberi, B, Kushner, T. Direct-acting antiviral treatment of acute hepatitis C virus infections. Expert Rev Anti Infect Ther 2018;16:599610.Google Scholar
Rehermann, B. HCV in 2015: advances in hepatitis C research and treatment. Nat Rev Gastroenterol Hepatol 2016;13:70–2.Google Scholar
Kwo, PY, Mantry, PS, Coakley, E, Te, HS, Vargas, HE, Brown, R Jr., Gordon, F, et al. An interferon-free antiviral regimen for HCV after liver transplantation. N Engl J Med 2014;371:2375–82.Google Scholar
Lawitz, E, Gane, E, Pearlman, B, Tam, E, Ghesquiere, W, Guyader, D, Alric, L, et al. Efficacy and safety of 12 weeks versus 18 weeks of treatment with grazoprevir (MK-5172) and elbasvir (MK-8742) with or without ribavirin for hepatitis C virus genotype 1 infection in previously untreated patients with cirrhosis and patients with previous null response with or without cirrhosis (C-WORTHY): a randomised, open-label phase 2 trial. Lancet 2015;385:1075–86.Google Scholar
Poordad, F, Hezode, C, Trinh, R, Kowdley, KV, Zeuzem, S, Agarwal, K, Shiffman, ML, et al. ABT-450/r-ombitasvir and dasabuvir with ribavirin for hepatitis C with cirrhosis. N Engl J Med 2014;370:1973–82.Google Scholar
Sulkowski, M, Hezode, C, Gerstoft, J, Vierling, JM, Mallolas, J, Pol, S, Kugelmas, M, et al. Efficacy and safety of 8 weeks versus 12 weeks of treatment with grazoprevir (MK-5172) and elbasvir (MK-8742) with or without ribavirin in patients with hepatitis C virus genotype 1 mono-infection and HIV/hepatitis C virus co-infection (C-WORTHY): a randomised, open-label phase 2 trial. Lancet 2015;385:1087–97.Google Scholar
Zeuzem, S, Jacobson, IM, Baykal, T, Marinho, RT, Poordad, F, Bourliere, M, Sulkowski, MS, et al. Retreatment of HCV with ABT-450/r-ombitasvir and dasabuvir with ribavirin. N Engl J Med 2014;370:1604–14.Google Scholar
Abdel-Hady, M, Bansal, S, Davison, SM, Brown, M, Tizzard, SA, Mulla, S, Barnes, E, et al. Treatment of chronic viral hepatitis C in children and adolescents: UK experience. Arch Dis Child 2014;99:505–10.Google Scholar
Afdhal, N, Zeuzem, S, Kwo, P, Chojkier, M, Gitlin, N, Puoti, M, Romero-Gomez, M, et al. Ledipasvir and sofosbuvir for untreated HCV genotype 1 infection. N Engl J Med 2014;370:1889–98.Google Scholar
Curry, MP, O’Leary, JG, Bzowej, N, Muir, AJ, Korenblat, KM, Fenkel, JM, Reddy, KR, et al. Sofosbuvir and velpatasvir for HCV in patients with decompensated cirrhosis. N Engl J Med 2015;373:2618–28.Google Scholar
Druyts, E, Thorlund, K, Wu, P, Kanters, S, Yaya, S, Cooper, CL, Mills, EJ. Efficacy and safety of pegylated interferon alfa-2a or alfa-2b plus ribavirin for the treatment of chronic hepatitis C in children and adolescents: a systematic review and meta-analysis. Clin Infect Dis 2013;56:961–7.Google Scholar
Feld, JJ, Jacobson, IM, Hezode, C, Asselah, T, Ruane, PJ, Gruener, N, Abergel, A, et al. Sofosbuvir and velpatasvir for HCV genotype 1, 2, 4, 5, and 6 infection. N Engl J Med 2015;373:2599–607.Google Scholar
Foster, GR, Afdhal, N, Roberts, SK, Brau, N, Gane, EJ, Pianko, S, Lawitz, E, et al. Sofosbuvir and velpatasvir for HCV genotype 2 and 3 infection. N Engl J Med 2015;373:2608–17.Google Scholar
Gane, EJ, Stedman, CA, Hyland, RH, Ding, X, Svarovskaia, E, Symonds, WT, Hindes, RG, et al. Nucleotide polymerase inhibitor sofosbuvir plus ribavirin for hepatitis C. N Engl J Med 2013;368:3444.Google Scholar
Jara, P, Hierro, L, de la Vega, A, Diaz, C, Camarena, C, Frauca, E, Minos-Bartolo, G, et al. Efficacy and safety of peginterferon-alpha2b and ribavirin combination therapy in children with chronic hepatitis C infection. Pediatr Infect Dis J 2008;27:142–8.Google Scholar
Lawitz, E, Poordad, FF, Pang, PS, Hyland, RH, Ding, X, Mo, H, Symonds, WT, et al. Sofosbuvir and ledipasvir fixed-dose combination with and without ribavirin in treatment-naive and previously treated patients with genotype 1 hepatitis C virus infection (LONESTAR): an open-label, randomised, phase 2 trial. Lancet 2014;383:515–23.Google Scholar
Pawlotsky, JM. New hepatitis C therapies: the toolbox, strategies, and challenges. Gastroenterology 2014;146:1176–92.Google Scholar
Wirth, S, Ribes-Koninckx, C, Calzado, MA, Bortolotti, F, Zancan, L, Jara, P, Shelton, M, et al. High sustained virologic response rates in children with chronic hepatitis C receiving peginterferon alfa-2b plus ribavirin. J Hepatol 2010;52:501–7.Google Scholar
Wisniewska-Ligier, M, Pawlowska, M, Pilarczyk, M, Halota, W, Wozniakowska-Gesicka, T. Efficacy of pegylated interferon alpha-2b and ribavirin in chronic hepatitis C virus (genotypes 1 and 4) infection. J Pediatr Gastroenterol Nutr 2013;57:694–9.Google Scholar
Gonzalez-Peralta, RP, Kelly, DA, Haber, B, Molleston, J, Murray, KF, Jonas, MM, Shelton, M, et al. Interferon alfa-2b in combination with ribavirin for the treatment of chronic hepatitis C in children: efficacy, safety, and pharmacokinetics. Hepatology 2005;42:1010–18.Google Scholar
Sokal, EM, Bourgois, A, Stephenne, X, Silveira, T, Porta, G, Gardovska, D, Fischler, B, et al. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection in children and adolescents. J Hepatol 2010;52:827–31.Google Scholar
Suzuki, M, Tajiri, H, Tanaka, Y, Takano, T, Miyoshi, Y, Murakami, J, Shimizu, T, et al. Peginterferon therapy in children with chronic hepatitis C: a nationwide, multicenter study in Japan, 2004–2013. J Pediatr Gastroenterol Nutr 2016;63:8893.Google Scholar
Wirth, S, Lang, T, Gehring, S, Gerner, P. Recombinant alfa-interferon plus ribavirin therapy in children and adolescents with chronic hepatitis C. Hepatology 2002;36:1280–4.Google Scholar
Wirth, S, Pieper-Boustani, H, Lang, T, Ballauff, A, Kullmer, U, Gerner, P, Wintermeyer, P, et al. Peginterferon alfa-2b plus ribavirin treatment in children and adolescents with chronic hepatitis C. Hepatology 2005;41:1013–18.Google Scholar
Jonas, MM, Balistreri, W, Gonzalez-Peralta, RP, Haber, B, Lobritto, S, Mohan, P, Molleston, JP, et al. Pegylated interferon for chronic hepatitis C in children affects growth and body composition: results from the pediatric study of hepatitis C (PEDS-C) trial. Hepatology 2012;56:523–31.Google Scholar
Jonas, MM, Schwarz, KB, Gonzalez-Peralta, R, Lobritto, S, Molleston, JP, Murray, KF, Rosenthal, P, et al. Long-term growth outcomes in children treated for chronic hepatitis C. J Pediatr 2014;165:1252–4.Google Scholar
Granot, E, Sokal, EM. Hepatitis C virus in children: deferring treatment in expectation of direct-acting antiviral agents. Isr Med Assoc J 2015;17:707–11.Google Scholar
Schwarz, KB, Molleston, JP, Jonas, MM, Wen, J, Murray, KF, Rosenthal, P, Gonzalez-Peralta, RP, et al. Durability of response in children treated with pegylated interferon alfa [corrected] 2a +/- ribavirin for chronic hepatitis C. J Pediatr Gastroenterol Nutr 2016;62:93–6.Google Scholar
Zhong, YW, Zhang, HF, Shi, YM, Li, YL, Chu, F, Xu, ZQ, Chen, DW, et al. IL28B SNP rs12979860 is the critical predictor for sustained viral response in Chinese children aged 1 to 6 years with chronic hepatitis C. Int J Biol Sci 2016;12:1357–62.Google Scholar
Zhu, SS, Zeng, QL, Dong, Y, Xu, ZQ, Wang, LM, Chen, DW, Gan, Y, et al. Interferon-alpha plus ribavirin yields 98% sustained virologic response in children aged 1–5 years with iatrogenic chronic hepatitis C. Hepatol Int 2015;9:578–85.Google Scholar
Balistreri, WF, Murray, KF, Rosenthal, P, Bansal, S, Chuan-Hao, L, Kersey, K, Massetto, B, et al. The safety and effectiveness of ledipasvir-sofosbuvir in adolescents 12 to 17 years old with hepatitis C virus genotype 1 infection. Hepatology 2017;66(2):371–8.Google Scholar
Wirth, S, Rosenthal, P, Gonzalez-Peralta, RP, Jonas, MM, Balistreri, WF, Lin, CH, Hardikar, W, et al. Sofosbuvir and ribavirin in adolescents 12–17 years old with hepatitis C virus genotype 2 or 3 infection. Hepatology 2017;66:1102–10.Google Scholar
Indolfi, G, Hierro, L, Dezsofi, A, Jahnel, J, Debray, D, Hadzic, N, Czubkowski, P, et al. Treatment of chronic hepatitis C virus infection in children: a position paper by the Hepatology Committee of the European Society of Paediatric Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol Nutr 2018;66:505–15.Google Scholar
Nguyen, J, Barritt, ASt, Jhaveri, R. Cost effectiveness of early treatment with direct-acting antiviral therapy in adolescent patients with hepatitis C virus infection. J Pediatr 2019;207:90–6.Google Scholar
Leung, DH, Squires, JE, Jhaveri, R, Kerkar, N, Lin, CH, Mohan, P, Murray, KF, Gonzalez-Peralta, RP, Roberts, EA, Sundaram, SS. Hepatitis C in 2020: A North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition Position Paper.J Pediatr Gastroenterol Nutr. 2020 Sep;71(3):407–417.Google Scholar
The European Association for the Study of the Liver. EASL Clinical Practice Guidelines: management of hepatitis C virus infection. J Hepatol 2014;60:392420.Google Scholar
Panel, AIHG. Hepatitis C guidance: AASLD-IDSA recommendations for testing, managing, and treating adults infected with hepatitis C virus. Hepatology 2015;62:932–54.Google Scholar
Poynard, T, Morra, R, Halfon, P, Castera, L, Ratziu, V, Imbert-Bismut, F, Naveau, S, et al. Meta-analyses of FibroTest diagnostic value in chronic liver disease. BMC Gastroenterol 2007;7:40.Google Scholar
Steadman, R, Myers, RP, Leggett, L, Lorenzetti, D, Noseworthy, T, Rose, S, Sutherland, L, et al. A health technology assessment of transient elastography in adult liver disease. Can J Gastroenterol 2013;27:149–58.Google Scholar
Tsochatzis, EA, Gurusamy, KS, Ntaoula, S, Cholongitas, E, Davidson, BR, Burroughs, AK. Elastography for the diagnosis of severity of fibrosis in chronic liver disease: a meta-analysis of diagnostic accuracy. J Hepatol 2011;54:650–9.Google Scholar
Poynard, T, Moussalli, J, Munteanu, M, Thabut, D, Lebray, P, Rudler, M, Ngo, Y, et al. Slow regression of liver fibrosis presumed by repeated biomarkers after virological cure in patients with chronic hepatitis C. J Hepatol 2013;59:675–83.Google Scholar
Poynard, T, Munteanu, M, Deckmyn, O, Ngo, Y, Drane, F, Castille, JM, Housset, C, et al. Validation of liver fibrosis biomarker (FibroTest) for assessing liver fibrosis progression: proof of concept and first application in a large population. J Hepatol 2012;57:541–8.Google Scholar
Poynard, T, Ngo, Y, Munteanu, M, Thabut, D, Massard, J, Moussalli, J, Varaud, A, et al. Biomarkers of liver injury for hepatitis clinical trials: a meta-analysis of longitudinal studies. Antivir Ther 2010;15:617–31.Google Scholar
Poynard, T, Vergniol, J, Ngo, Y, Foucher, J, Munteanu, M, Merrouche, W, Colombo, M, et al. Staging chronic hepatitis C in seven categories using fibrosis biomarker (FibroTest) and transient elastography (FibroScan(R)). J Hepatol 2014;60:706–14.Google Scholar
Vergniol, J, Foucher, J, Terrebonne, E, Bernard, PH, le Bail, B, Merrouche, W, Couzigou, P, et al. Noninvasive tests for fibrosis and liver stiffness predict 5-year outcomes of patients with chronic hepatitis C. Gastroenterology 2011;140:1970–9, e1971–3.Google Scholar
Gupta, M, Bahirwani, R, Levine, MH, Malik, S, Goldberg, D, Reddy, KR, Shaked, A. Outcomes in pediatric hepatitis C transplant recipients: analysis of the UNOS database. Pediatr Transplant 2015;19:153–63.Google Scholar
Smith, BD, Morgan, RL, Beckett, GA, Falck-Ytter, Y, Holtzman, D, Teo, CG, Jewett, A, et al. Recommendations for the identification of chronic hepatitis C virus infection among persons born during 1945–1965. MMWR Recomm Rep 2012;61:132.Google Scholar
El-Kamary, SS, Serwint, JR, Joffe, A, Santosham, M, Duggan, AK. Prevalence of hepatitis C virus infection in urban children. J Pediatr 2003;143:54–9.Google Scholar
Trucchi, C, Orsi, A, Alicino, C, Sticchi, L, Icardi, G, Ansaldi, F. State of the art, unresolved issues, and future research directions in the fight against hepatitis C virus: perspectives for screening, diagnostics of resistances, and immunization. J Immunol Res 2016;2016:1412840.Google Scholar
Snijdewind, IJ, Smit, C, Schutten, M, Nellen, FJ, Kroon, FP, Reiss, P, van der Ende, ME. Low mother-to-child-transmission rate of hepatitis C virus in cART treated HIV-1 infected mothers. J Clin Virol 2015;68:1115.Google Scholar
Brown, RS Jr., McMahon, BJ, Lok, AS, Wong, JB, Ahmed, AT, Mouchli, MA, Wang, Z, et al. Antiviral therapy in chronic hepatitis B viral infection during pregnancy: a systematic review and meta-analysis. Hepatology 2016;63:319–33.Google Scholar
Moradpour, D, Grakoui, A, Manns, MP. Future landscape of hepatitis C research – basic, translational and clinical perspectives. J Hepatol 2016;65:S143–55.Google Scholar
Pierce, BG, Keck, ZY, Lau, P, Fauvelle, C, Gowthaman, R, Baumert, TF, Fuerst, TR, et al. Global mapping of antibody recognition of the hepatitis C virus E2 glycoprotein: implications for vaccine design. Proc Natl Acad Sci U S A 2016;113(45):E6946–54..Google Scholar
Folgori, A, Capone, S, Ruggeri, L, Meola, A, Sporeno, E, Ercole, BB, Pezzanera, M, et al. A T-cell HCV vaccine eliciting effective immunity against heterologous virus challenge in chimpanzees. Nat Med 2006;12:190–7.Google Scholar
Yokokawa, H, Higashino, A, Suzuki, S, Moriyama, M, Nakamura, N, Suzuki, T, Suzuki, R, et al. Induction of humoural and cellular immunity by immunisation with HCV particle vaccine in a non-human primate model. Gut 2018;67(2):372–9.Google Scholar
Swadling, L, Capone, S, Antrobus, RD, Brown, A, Richardson, R, Newell, EW, Halliday, J, et al. A human vaccine strategy based on chimpanzee adenoviral and MVA vectors that primes, boosts, and sustains functional HCV-specific T cell memory. Sci Transl Med 2014;6:261ra153.Google Scholar
Graf Einsiedel, H, Christiansen, H, Wiegand, J. Eight weeks treatment with sofosbuvir/ledipasvir in a 4-year-old child with chronic hepatitis C virus genotype 1 infection. Pediatr Infect Dis J 2016;35:1373.Google Scholar
Hashmi, MA, Cheema, HA. Effectiveness and safety of sofosbuvir in treatment-naive children with hepatitis C infection. J Coll Physicians Surg Pak 2017;27:423–6.Google Scholar
Murray, KF, Balistreri, WF, Bansal, S, Whitworth, S, Evans, HM, Gonzalez-Peralta, RP, Wen, J, et al. Safety and efficacy of ledipasvir-sofosbuvir with or without ribavirin for chronic hepatitis C in children aged 6–11. Hepatology 2018;68:2158–66.Google Scholar
El-Karaksy, H, Mogahed, EA, Abdullatif, H, Ghobrial, C, El-Raziky, MS, El-Koofy, N, El-Shabrawi, M, et al. Sustained viral response in genotype 4 chronic hepatitis C virus-infected children and adolescents treated with sofosbuvir/ledipasvir. J Pediatr Gastroenterol Nutr 2018;67:626–30.Google Scholar
Leung, DH, Wirth, S, Yao, BB, Viani, RM, Gonzalez-Peralta, RP, Jonas, MM, Lobritto, SJ, et al. Ombitasvir/paritaprevir/ritonavir with or without dasabuvir and with or without ribavirin for adolescents with HCV genotype 1 or 4. Hepatol Commun 2018;2:1311–19.Google Scholar
Alkaaby, BA, Al-Ethawi, AE. The effectiveness of oral antiviral (sofosbuvir/ledipasvir) in treating children with HCV infection. Pak J Med Sci 2018;34:1353–6.Google Scholar
Tucci, F, Calbi, V, Barzaghi, F, Migliavacca, M, Ferrua, F, Bernardo, ME, Canarutto, D, et al. Successful treatment with ledipasvir/sofosbuvir in an infant with severe combined immunodeficiency caused by adenosine deaminase deficiency with HCV allowed gene therapy with strimvelis. Hepatology 2018;68:2434–7.Google Scholar
El-Shabrawi, MHF, Kamal, NM, El-Khayat, HR, Kamal, EM, Abdel Gawad, M, Yakoot, M. A pilot single arm observational study of sofosbuvir/ledipasvir (200 + 45 mg) in 6- to 12-year old children. Aliment Pharmacol Ther 2018;47:1699–704.Google Scholar
Quintero, J, Juamperez, J, Julio, E, Cabello, V, Mercadal-Hally, M, Soler-Palacin, P, Segarra, O, et al. Ledipasvir/sofosbuvir combination for chronic hepatitis C infection in children and adolescents. An Pediatr 2019;90:141–7.Google Scholar
Abdel Ghaffar, TY, El Naghi, S, Abdel Gawad, M, Helmy, S, Abdel Ghaffar, A, Yousef, M, Moafy, M. Safety and efficacy of combined sofosbuvir/daclatasvir treatment of children and adolescents with chronic hepatitis C genotype 4. J Viral Hepat 2019;26:263–70.Google Scholar
Fouad, HM, Ahmed Mohamed, A, Sabry, M, Abdel Aziz, H, Eysa, B, Rabea, M. The effectiveness of ledipasvir/sofosbuvir in youth with genotype 4 hepatitis C virus: a single Egyptian center study. Pediatr Infect Dis J 2019;38:22–5.Google Scholar
Sofosbuvir, and Ribavirin, Therapy for Children Aged 3 to <12 Years With Hepatitis C Virus Genotype 2 or 3 Infection. Rosenthal P, Schwarz KB, Gonzalez-Peralta RP, Lin CH, Kelly DA, Nightingale S, Balistreri WF, Bansal S, Jonas MM, Massetto B, Brainard DM, Hsueh CH, Shao J, Parhy B, Davison S, Feiterna-Sperling C, Gillis LA, Indolfi G, Sokal EM, Murray KF, Wirth S.Hepatology. 2020 71(1):31–43.Google Scholar
Schwarz, KB RP, Murray, KF, Honegger, JR, Hardikar, W, Hague, R, Mittal, N, Massetto, B, Brainard, D, Begley, R, Parhy, B, Shao, J, Narkewicz, MR, Rao, GS, Whitworth, S, Bansal, S, Balistreri, WB. Ledipasvir/sofosbuvir for 12 weeks is safe and effective in children 3 to &lt;6 years old with chronic hepatitis C virus infection. Hepatology 2020;71(2):422–30.Google Scholar
Guyatt, GH, Oxman, AD, Vist, GE, Kunz, R, Falck-Ytter, Y, Alonso-Coello, P, Schunemann, HJ, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008;336:924–6.Google Scholar

References

Mieli-Vergani, G, Vergani, D. Autoimmune hepatitis. Nat Rev Gastroenterol Hepatol 2011;8(6):320–9.Google Scholar
Whittingham, S, et al. Smooth muscle autoantibody in “autoimmune” hepatitis. Gastroenterology 1966;51(4):499505.Google Scholar
De Groote, J, et al. A classification of chronic hepatitis. Lancet 1968;2(7568):626–8.Google Scholar
Johnson, PJ, McFarlane, IG. Meeting report: International Autoimmune Hepatitis Group. Hepatology 1993;18(4):9981005.Google Scholar
Palle, SK, et al. Racial disparities in presentation and outcomes of paediatric autoimmune hepatitis. Liver Int 2019;39(5):976–84.Google Scholar
Czaja, AJ. Global disparities and their implications in the occurrence and outcome of autoimmune hepatitis. Dig Dis Sci 2017;62(9):2277–92.Google Scholar
Mieli-Vergani, G, et al. Autoimmune hepatitis. Nat Rev Dis Primers 2018;4:18017.Google Scholar
Webb, GJ, et al. Cellular and molecular mechanisms of autoimmune hepatitis. Annu Rev Pathol 2018;13:247–92.Google Scholar
Longhi, MS, et al. Aetiopathogenesis of autoimmune hepatitis. J Autoimmun 2010;34(1):714.Google Scholar
Than, NN, Jeffery, HC, Oo, YH. Autoimmune hepatitis: progress from global immunosuppression to personalised regulatory T cell therapy. Can J Gastroenterol Hepatol 2016;2016:7181685.Google Scholar
Mieli-Vergani, G, Vergani, D. Autoimmune hepatitis in children: what is different from adult AIH? Semin Liver Dis 2009;29(3):297306.Google Scholar
Invernizzi, P, Lleo, A, Podda, M. Interpreting serological tests in diagnosing autoimmune liver diseases. Semin Liver Dis 2007;27(2):161–72.Google Scholar
Czaja, AJ. Autoantibodies as prognostic markers in autoimmune liver disease. Dig Dis Sci 2010;55(8):2144–61.Google Scholar
Karlsen, TH, Chung, BK. Genetic risk and the development of autoimmune liver disease. Dig Dis 2015;33(Suppl 2):1324.Google Scholar
Kerkar, N. (2019). Autoimmune hepatitis in special populations: in pediatrics and across different ethnicities/races, in Clinical Liver Disease (pp. 3740). Oxford: Wiley.Google Scholar
Jaeckel, E, Hardtke-Wolenski, M, Fischer, K. The benefit of animal models for autoimmune hepatitis. Best Pract Res Clin Gastroenterol 2011;25(6):643–51.Google Scholar
Lapierre, P, et al. A murine model of type 2 autoimmune hepatitis: xenoimmunization with human antigens. Hepatology 2004;39(4):1066–74.Google Scholar
Christen, U, Hintermann, E. Immunopathogenic mechanisms of autoimmune hepatitis: how much do we know from animal models? Int J Mol Sci 2016;17(12). https://doi.org/10.3390/ijms17122007Google Scholar
Hennes, EM, et al. Simplified criteria for the diagnosis of autoimmune hepatitis. Hepatology 2008;48(1):169–76.Google Scholar
Arcos-Machancoses, JV, et al. Accuracy of the simplified criteria for autoimmune hepatitis in children: systematic review and decision analysis. J Clin Exp Hepatol 2019;9(2):147–55.Google Scholar
Czaja, AJ. Drug-induced autoimmune-like hepatitis. Dig Dis Sci 2011;56(4):958–76.Google Scholar
Kerkar, N, Miloh, T. Sclerosing cholangitis: pediatric perspective. Curr Gastroenterol Rep 2010;12(3):195202.Google Scholar
Weiler-Normann, C, Schramm, C. Drug-induced liver injury and its relationship to autoimmune hepatitis. J Hepatol 2011;55(4):747–9.Google Scholar
Zen, Y, Yeh, MM. Hepatotoxicity of immune checkpoint inhibitors: a histology study of seven cases in comparison with autoimmune hepatitis and idiosyncratic drug-induced liver injury. Mod Pathol 2018;31(6):965–73.Google Scholar
Mathis, D, Benoist, C. Aire. Annu Rev Immunol 2009;27:287312.Google Scholar
Davies, EG, Thrasher, AJ. Update on the hyper immunoglobulin M syndromes. Br J Haematol 2010;149(2):167–80.Google Scholar
Cook, GC, Mulligan, R, Sherlock, S. Controlled prospective trial of corticosteroid therapy in active chronic hepatitis. Q J Med 1971;40(158):159–85.Google Scholar
Rumbo, C, et al. Azathioprine metabolite measurements in the treatment of autoimmune hepatitis in pediatric patients: a preliminary report. J Pediatr Gastroenterol Nutr 2002;35(3):391–8.Google Scholar
Sheiko, MA, et al. Outcomes in pediatric autoimmune hepatitis and significance of azathioprine metabolites. J Pediatr Gastroenterol Nutr 2017;65(1):80–5.Google Scholar
Dunkin, D, et al. Allopurinol salvage therapy in pediatric overlap autoimmune hepatitis-primary sclerosing cholangitis with 6-MMP toxicity. J Pediatr Gastroenterol Nutr 2010;51(4):524–6.Google Scholar
Pratt, DS, Flavin, DP, Kaplan, MM. The successful treatment of autoimmune hepatitis with 6-mercaptopurine after failure with azathioprine. Gastroenterology 1996;110(1):271–4.Google Scholar
Gregorio, GV, et al. Organ and non-organ specific autoantibody titres and IgG levels as markers of disease activity: a longitudinal study in childhood autoimmune liver disease. Autoimmunity 2002;35(8):515–19.Google Scholar
Czaja, AJ, Manns, MP. Advances in the diagnosis, pathogenesis, and management of autoimmune hepatitis. Gastroenterology 2010;139(1):5872 e4.Google Scholar
Lohse, AW, Gil, H. Reactivation of autoimmune hepatitis during budesonide monotherapy, and response to standard treatment. J Hepatol 2011;54(4):837–9.Google Scholar
Manns, MP, et al. Budesonide induces remission more effectively than prednisone in a controlled trial of patients with autoimmune hepatitis. Gastroenterology 2010;139(4):1198–206.Google Scholar
Woynarowski, M, et al. Budesonide versus prednisone with azathioprine for the treatment of autoimmune hepatitis in children and adolescents. J Pediatr 2013;163(5):1347–53 e1.Google Scholar
Zizzo, AN, et al. Second-line agents in pediatric patients with autoimmune hepatitis: a systematic review and meta-analysis. J Pediatr Gastroenterol Nutr 2017;65(1):615.Google Scholar
Kerkar, N. Pediatric autoimmune hepatitis: what do you do when first-line therapy fails? J Pediatr Gastroenterol Nutr 2017;65(1):24.Google Scholar
Richardson, PD, James, PD, Ryder, SD. Mycophenolate mofetil for maintenance of remission in autoimmune hepatitis in patients resistant to or intolerant of azathioprine. J Hepatol 2000;33(3):371–5.Google Scholar
Aw, MM, et al. Mycophenolate mofetil as rescue treatment for autoimmune liver disease in children: a 5-year follow-up. J Hepatol 2009;51(1):156–60.Google Scholar
Yu, ZJ, et al. Comparison of mycophenolate mofetil with standard treatment for autoimmune hepatitis: a meta-analysis. Eur J Gastroenterol Hepatol 2019;31(7):873–7.Google Scholar
Sherman, KE, Narkewicz, M, Pinto, PC. Cyclosporine in the management of corticosteroid-resistant type I autoimmune chronic active hepatitis. J Hepatol 1994;21(6):1040–7.Google Scholar
Hyams, JS, Ballow, M, Leichtner, AM. Cyclosporine treatment of autoimmune chronic active hepatitis. Gastroenterology 1987;93(4):890–3.Google Scholar
De Lemos-Bonotto, M, et al. A systematic review and meta-analysis of second-line immunosuppressants for autoimmune hepatitis treatment. Eur J Gastroenterol Hepatol 2018;30(2):212–16.Google Scholar
Hanouneh, M, et al. A review of the utility of tacrolimus in the management of adults with autoimmune hepatitis. Scand J Gastroenterol 2019;54(1):7680.Google Scholar
Marlaka, JR, et al. Tacrolimus without or with the addition of conventional immunosuppressive treatment in juvenile autoimmune hepatitis. Acta Paediatr 2012;101(9):993–9.Google Scholar
Efe, C, et al. Tacrolimus and mycophenolate mofetil as second-line therapies for pediatric patients with autoimmune hepatitis. Dig Dis Sci 2018;63(5):1348–54.Google Scholar
Mieli-Vergani, G, et al. Diagnosis and management of pediatric autoimmune liver disease: ESPGHAN Hepatology Committee Position Statement. J Pediatr Gastroenterol Nutr 2018;66(2):345–60.Google Scholar
Cuarterolo, ML, et al. Immunosuppressive therapy allows recovery from liver failure in children with autoimmune hepatitis. Clin Gastroenterol Hepatol 2011;9(2):145–9.Google Scholar
Rahim, MN, et al. Acute severe autoimmune hepatitis: corticosteroids or liver transplantation? Liver Transpl 2019;25(6):946–59.Google Scholar
Ichai, P, et al. Usefulness of corticosteroids for the treatment of severe and fulminant forms of autoimmune hepatitis. Liver Transpl 2007;13(7):9961003.Google Scholar
Di Giorgio, A, et al. Fulminant hepatic failure of autoimmune aetiology in children. J Pediatr Gastroenterol Nutr 2015;60(2):159–64.Google Scholar
Greene, MT, Whitington, PF. Outcomes in pediatric autoimmune hepatitis. Curr Gastroenterol Rep 2009;11(3):248–51.Google Scholar
Montano-Loza, AJ, Carpenter, HA, Czaja, AJ. Improving the end point of corticosteroid therapy in type 1 autoimmune hepatitis to reduce the frequency of relapse. Am J Gastroenterol 2007;102(5):1005–12.Google Scholar
Al-Chalabi, T, et al. Impact of gender on the long-term outcome and survival of patients with autoimmune hepatitis. J Hepatol 2008;48(1):140–7.Google Scholar
Radhakrishnan, KR, et al. Autoimmune hepatitis in children–impact of cirrhosis at presentation on natural history and long-term outcome. Dig Liver Dis 2010;42(10):724–8.Google Scholar
Yeoman, AD, et al. Evaluation of risk factors in the development of hepatocellular carcinoma in autoimmune hepatitis: implications for follow-up and screening. Hepatology 2008;48(3):863–70.Google Scholar
Kerkar, N, et al. De-novo autoimmune hepatitis after liver transplantation. Lancet 1998;351(9100):409–13.Google Scholar
Demetris, AJ, et al. Comprehensive Update of the Banff Working Group on Liver Allograft Pathology: Introduction of Antibody-Mediated Rejection. Am J Transplant 2016;16(10):2816–35.Google Scholar
Kerkar, N, Vergani, D. De novo autoimmune hepatitis: is this different in adults compared to children? J Autoimmun 2018;95:2633.Google Scholar
Kerkar, N, et al. Rapamycin successfully treats post-transplant autoimmune hepatitis. Am J Transplant 2005;5(5):1085–9.Google Scholar

References

Molodecky, NA, Kareemi, H, Parab, R, Barkema, HW, Quan, H, Myers, RP, et al. Incidence of primary sclerosing cholangitis: a systematic review and meta-analysis. Hepatology 2011;53(5):1590–9.Google Scholar
Tanaka, A, Takikawa, H. Geoepidemiology of primary sclerosing cholangitis: a critical review. J Autoimmun 2013;46:3540.Google Scholar
Lindkvist, B, Benito de Valle, M, Gullberg, B, Bjornsson, E. Incidence and prevalence of primary sclerosing cholangitis in a defined adult population in Sweden. Hepatology 2010;52(2):571–7.Google Scholar
Card, TR, Solaymani-Dodaran, M, West, J. Incidence and mortality of primary sclerosing cholangitis in the UK: a population-based cohort study. J Hepatol 2008;48(6):939–44.Google Scholar
Escorsell, A, Pares, A, Rodes, J, Solis-Herruzo, JA, Miras, M, de la Morena, E. Epidemiology of primary sclerosing cholangitis in Spain. Spanish Association for the Study of the Liver. J Hepatol 1994;21(5):787–91.Google Scholar
Kaplan, GG, Laupland, KB, Butzner, D, Urbanski, SJ, Lee, SS. The burden of large and small duct primary sclerosing cholangitis in adults and children: a population-based analysis. Am J Gastroenterol 2007;102(5):1042–9.Google Scholar
Deneau, M, Jensen, MK, Holmen, J, Williams, MS, Book, LS, Guthery, SL. Primary sclerosing cholangitis, autoimmune hepatitis, and overlap in Utah children: epidemiology and natural history. Hepatology 2013;58(4):1392–400.Google Scholar
Deneau, MR, El-Matary, W, Valentino, PL, Abdou, R, Alqoaer, K, Amin, M, et al. The natural history of primary sclerosing cholangitis in 781 children: a multicenter, international collaboration. Hepatology 2017;66(2):518–27.Google Scholar
Kaya, M, Angulo, P, Lindor, KD. Overlap of autoimmune hepatitis and primary sclerosing cholangitis: an evaluation of a modified scoring system. J Hepatol 2000;33(4):537–42.Google Scholar
Abdalian, R, Dhar, P, Jhaveri, K, Haider, M, Guindi, M, Heathcote, EJ. Prevalence of sclerosing cholangitis in adults with autoimmune hepatitis: evaluating the role of routine magnetic resonance imaging. Hepatology 2008;47(3):949–57.Google Scholar
Gregorio, GV, Portmann, B, Karani, J, Harrison, P, Donaldson, PT, Vergani, D, et al. Autoimmune hepatitis/sclerosing cholangitis overlap syndrome in childhood: a 16-year prospective study. Hepatology 2001;33(3):544–53.Google Scholar
Bergquist, A, Montgomery, SM, Bahmanyar, S, Olsson, R, Danielsson, A, Lindgren, S, et al. Increased risk of primary sclerosing cholangitis and ulcerative colitis in first-degree relatives of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol 2008;6(8):939–43.Google Scholar
Folseraas, T, Melum, E, Rausch, P, Juran, BD, Ellinghaus, E, Shiryaev, A, et al. Extended analysis of a genome-wide association study in primary sclerosing cholangitis detects multiple novel risk loci. J Hepatol 2012;57(2):366–75.Google Scholar
Melum, E, Franke, A, Schramm, C, Weismuller, TJ, Gotthardt, DN, Offner, FA, et al. Genome-wide association analysis in primary sclerosing cholangitis identifies two non-HLA susceptibility loci. Nat Genet 2011;43(1):1719.Google Scholar
Liu, JZ, Hov, JR, Folseraas, T, Ellinghaus, E, Rushbrook, SM, Doncheva, NT, et al. Dense genotyping of immune-related disease regions identifies nine new risk loci for primary sclerosing cholangitis. Nat Genet 2013;45(6):670–5.Google Scholar
Ellinghaus, D, Folseraas, T, Holm, K, Ellinghaus, E, Melum, E, Balschun, T, et al. Genome-wide association analysis in primary sclerosing cholangitis and ulcerative colitis identifies risk loci at GPR35 and TCF4. Hepatology 2013;58(3):1074–83.Google Scholar
Ellinghaus, D, Jostins, L, Spain, SL, Cortes, A, Bethune, J, Han, B, et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat Genet 2016;48(5):510–18.Google Scholar
Ji, SG, Juran, BD, Mucha, S, Folseraas, T, Jostins, L, Melum, E, et al. Genome-wide association study of primary sclerosing cholangitis identifies new risk loci and quantifies the genetic relationship with inflammatory bowel disease. Nat Genet 2017;49(2):269–73.Google Scholar
Srivastava, B, Mells, GF, Cordell, HJ, Muriithi, A, Brown, M, Ellinghaus, E, et al. Fine mapping and replication of genetic risk loci in primary sclerosing cholangitis. Scand J Gastroenterol 2012;47(7):820–6.Google Scholar
Andersen, IM, Tengesdal, G, Lie, BA, Boberg, KM, Karlsen, TH, Hov, JR. Effects of coffee consumption, smoking, and hormones on risk for primary sclerosing cholangitis. Clin Gastroenterol Hepatol 2014;12(6):1019–28.Google Scholar
Eaton, JE, Juran, BD, Atkinson, EJ, Schlicht, EM, Xie, X, de Andrade, M, et al. A comprehensive assessment of environmental exposures among 1000 North American patients with primary sclerosing cholangitis, with and without inflammatory bowel disease. Aliment Pharmacol Ther 2015;41(10):980–90.Google Scholar
Sabino, J, Vieira-Silva, S, Machiels, K, Joossens, M, Falony, G, Ballet, V, et al. Primary sclerosing cholangitis is characterised by intestinal dysbiosis independent from IBD. Gut 2016;65(10):1681–9.Google Scholar
Rossen, NG, Fuentes, S, Boonstra, K, D’Haens, GR, Heilig, HG, Zoetendal, EG, et al. The mucosa-associated microbiota of PSC patients is characterized by low diversity and low abundance of uncultured Clostridiales II. J Crohns Colitis 2015;9(4):342–8.Google Scholar
Iwasawa, K, Suda, W, Tsunoda, T, Oikawa-Kawamoto, M, Umetsu, S, Inui, A, et al. Characterisation of the faecal microbiota in Japanese patients with paediatric-onset primary sclerosing cholangitis. Gut 2017;66(7):1344–6.Google Scholar
Kummen, M, Holm, K, Anmarkrud, JA, Nygard, S, Vesterhus, M, Hoivik, ML, et al. The gut microbial profile in patients with primary sclerosing cholangitis is distinct from patients with ulcerative colitis without biliary disease and healthy controls. Gut 2017;66(4):611–19.Google Scholar
Hov, JR, Karlsen, TH. The microbiome in primary sclerosing cholangitis: current evidence and potential concepts. Semin Liver Dis 2017;37(4):314–31.Google Scholar
Quraishi, MN, Sergeant, M, Kay, G, Iqbal, T, Chan, J, Constantinidou, C, et al. The gut-adherent microbiota of PSC-IBD is distinct to that of IBD. Gut 2017;66(2):386–8.Google Scholar
Nakamoto, N, Sasaki, N, Aoki, R, Miyamoto, K, Suda, W, Teratani, T, et al. Gut pathobionts underlie intestinal barrier dysfunction and liver T helper 17 cell immune response in primary sclerosing cholangitis. Nat Microbiol 2019;4(3):492503.Google Scholar
Grant, AJ, Lalor, PF, Salmi, M, Jalkanen, S, Adams, DH. Homing of mucosal lymphocytes to the liver in the pathogenesis of hepatic complications of inflammatory bowel disease. Lancet 2002;359(9301):150–7.Google Scholar
Eksteen, B, Grant, AJ, Miles, A, Curbishley, SM, Lalor, PF, Hubscher, SG, et al. Hepatic endothelial CCL25 mediates the recruitment of CCR9+ gut-homing lymphocytes to the liver in primary sclerosing cholangitis. J Exp Med 2004;200(11):1511–17.Google Scholar
McNab, G, Reeves, JL, Salmi, M, Hubscher, S, Jalkanen, S, Adams, DH. Vascular adhesion protein 1 mediates binding of T cells to human hepatic endothelium. Gastroenterology 1996;110(2):522–8.Google Scholar
Deutschmann, K, Reich, M, Klindt, C, Droge, C, Spomer, L, Haussinger, D, et al. Bile acid receptors in the biliary tree: TGR5 in physiology and disease. Biochim Biophys Acta Mol Basis Dis 2018;1864(4 Pt B):1319–25.Google Scholar
Masyuk, TV, Masyuk, AI, Lorenzo Pisarello, M, Howard, BN, Huang, BQ, Lee, PY, et al. TGR5 contributes to hepatic cystogenesis in rodents with polycystic liver diseases through cyclic adenosine monophosphate/Galphas signaling. Hepatology 2017;66(4):1197–218.Google Scholar
Meng, L, Quezada, M, Levine, P, Han, Y, McDaniel, K, Zhou, T, et al. Functional role of cellular senescence in biliary injury. Am J Pathol 2015;185(3):602–9.Google Scholar
Lazaridis, KN, LaRusso, NF. Primary sclerosing cholangitis. N Engl J Med 2016;375(12):1161–70.Google Scholar
Lunder, AK, Hov, JR, Borthne, A, Gleditsch, J, Johannesen, G, Tveit, K, et al. Prevalence of sclerosing cholangitis detected by magnetic resonance cholangiography in patients with long-term inflammatory bowel disease. Gastroenterology 2016;151(4):660–9 e4.Google Scholar
Tischendorf, JJ, Hecker, H, Kruger, M, Manns, MP, Meier, PN. Characterization, outcome, and prognosis in 273 patients with primary sclerosing cholangitis: a single center study. Am J Gastroenterol 2007;102(1):107–14.Google Scholar
Feldstein, AE, Perrault, J, El-Youssif, M, Lindor, KD, Freese, DK, Angulo, P. Primary sclerosing cholangitis in children: a long-term follow-up study. Hepatology 2003;38(1):210–17.Google Scholar
Miloh, T, Arnon, R, Shneider, B, Suchy, F, Kerkar, N. A retrospective single-center review of primary sclerosing cholangitis in children. Clin Gastroenterol Hepatol 2009;7(2):239–45.Google Scholar
Loftus, EV Jr., Harewood, GC, Loftus, CG, Tremaine, WJ, Harmsen, WS, Zinsmeister, AR, et al. PSC-IBD: a unique form of inflammatory bowel disease associated with primary sclerosing cholangitis. Gut 2005;54(1):91–6.Google Scholar
Boonstra, K, van Erpecum, KJ, van Nieuwkerk, KM, Drenth, JP, Poen, AC, Witteman, BJ, et al. Primary sclerosing cholangitis is associated with a distinct phenotype of inflammatory bowel disease. Inflamm Bowel Dis 2012;18(12):2270–6.Google Scholar
Lundqvist, K, Broome, U. Differences in colonic disease activity in patients with ulcerative colitis with and without primary sclerosing cholangitis: a case control study. Dis Colon Rectum 1997;40(4):451–6.Google Scholar
Aitola, P, Matikainen, M, Mattila, J, Tomminen, T, Hiltunen, KM. Chronic inflammatory changes in the pouch mucosa are associated with cholangitis found on perioperative liver biopsy specimens at restorative proctocolectomy for ulcerative colitis. Scand J Gastroenterol 1998;33(3):289–93.Google Scholar
Shiau, H, Ihekweazu, FD, Amin, M, Fofanova, T, Miloh, T, Kellermayer, R. Unique inflammatory bowel disease phenotype of pediatric primary sclerosing cholangitis: a single-center study. J Pediatr Gastroenterol Nutr 2017;65(4):404–9.Google Scholar
Faubion, WA Jr., Loftus, EV, Sandborn, WJ, Freese, DK, Pediatric, Perrault J. “PSC-IBD”: a descriptive report of associated inflammatory bowel disease among pediatric patients with PSC. J Pediatr Gastroenterol Nutr 2001;33(3):296300.Google Scholar
Ricciuto, A, Fish, J, Carman, N, Walters, TD, Church, PC, Hansen, BE, et al. Symptoms do not correlate with findings from colonoscopy in children with inflammatory bowel disease and primary sclerosing cholangitis. Clin Gastroenterol Hepatol 2018;16(7):1098–105 e1.Google Scholar
Stanich, PP, Bjornsson, E, Gossard, AA, Enders, F, Jorgensen, R, Lindor, KD. Alkaline phosphatase normalization is associated with better prognosis in primary sclerosing cholangitis. Dig Liver Dis 2011;43(4):309–13.Google Scholar
el-Shabrawi, M, Wilkinson, ML, Portmann, B, Mieli-Vergani, G, Chong, SK, Williams, R, et al. Primary sclerosing cholangitis in childhood. Gastroenterology 1987;92(5 Pt 1):1226–35.Google Scholar
Debray, D, Pariente, D, Urvoas, E, Hadchouel, M, Bernard, O. Sclerosing cholangitis in children. J Pediatr 1994;124(1):4956.Google Scholar
Wilschanski, M, Chait, P, Wade, JA, Davis, L, Corey, M, St Louis, P, et al. Primary sclerosing cholangitis in 32 children: clinical, laboratory, and radiographic features, with survival analysis. Hepatology 1995;22(5):1415–22.Google Scholar
Batres, LA, Russo, P, Mathews, M, Piccoli, DA, Chuang, E, Ruchelli, E. Primary sclerosing cholangitis in children: a histologic follow-up study. Pediatr Dev Pathol 2005;8(5):568–76.Google Scholar
Hov, JR, Boberg, KM, Karlsen, TH. Autoantibodies in primary sclerosing cholangitis. World J Gastroenterol 2008;14(24):3781–91.Google Scholar
Dave, M, Elmunzer, BJ, Dwamena, BA, Higgins, PD. Primary sclerosing cholangitis: meta-analysis of diagnostic performance of MR cholangiopancreatography. Radiology 2010;256(2):387–96.Google Scholar
Ferrara, C, Valeri, G, Salvolini, L, Giovagnoni, A. Magnetic resonance cholangiopancreatography in primary sclerosing cholangitis in children. Pediatr Radiol 2002;32(6):413–17.Google Scholar
Majoie, CB, Reeders, JW, Sanders, JB, Huibregtse, K, Jansen, PL. Primary sclerosing cholangitis: a modified classification of cholangiographic findings. AJR Am J Roentgenol 1991;157(3):495–7.Google Scholar
Ponsioen, CY, Vrouenraets, SM, Prawirodirdjo, W, Rajaram, R, Rauws, EA, Mulder, CJ, et al. Natural history of primary sclerosing cholangitis and prognostic value of cholangiography in a Dutch population. Gut 2002;51(4):562–6.Google Scholar
Cotter, JM, Browne, LP, Capocelli, KE, McCoy, A, Mack, CL. Lack of correlation of liver tests with fibrosis stage at diagnosis in pediatric primary sclerosing cholangitis. J Pediatr Gastroenterol Nutr 2018;66(2):227–33.Google Scholar
Nakanuma, Y, Harada, K, Katayanagi, K, Tsuneyama, K, Sasaki, M. Definition and pathology of primary sclerosing cholangitis. J Hepatobiliary Pancreat Surg 1999;6(4):333–42.Google Scholar
Wiesner, RH, Grambsch, PM, Dickson, ER, Ludwig, J, MacCarty, RL, Hunter, EB, et al. Primary sclerosing cholangitis: natural history, prognostic factors and survival analysis. Hepatology 1989;10(4):430–6.Google Scholar
Ludwig, J. Surgical pathology of the syndrome of primary sclerosing cholangitis. Am J Surg Pathol 1989;13(Suppl 1):43–9.Google Scholar
Ishak, K, Baptista, A, Bianchi, L, Callea, F, De Groote, J, Gudat, F, et al. Histological grading and staging of chronic hepatitis. J Hepatol 1995;22(6):696–9.Google Scholar
Nakanuma, Y, Zen, Y, Harada, K, Sasaki, M, Nonomura, A, Uehara, T, et al. Application of a new histological staging and grading system for primary biliary cirrhosis to liver biopsy specimens: interobserver agreement. Pathol Int 2010;60(3):167–74.Google Scholar
de Vries, EM, Verheij, J, Hubscher, SG, Leeflang, MM, Boonstra, K, Beuers, U, et al. Applicability and prognostic value of histologic scoring systems in primary sclerosing cholangitis. J Hepatol 2015;63(5):1212–19.Google Scholar
de Vries, EM, de Krijger, M, Farkkila, M, Arola, J, Schirmacher, P, Gotthardt, D, et al. Validation of the prognostic value of histologic scoring systems in primary sclerosing cholangitis: an international cohort study. Hepatology 2017;65(3):907–19.Google Scholar
Aadland, E, Schrumpf, E, Fausa, O, Elgjo, K, Heilo, A, Aakhus, T, et al. Primary sclerosing cholangitis: a long-term follow-up study. Scand J Gastroenterol 1987;22(6):655–64.Google Scholar
Farrant, JM, Hayllar, KM, Wilkinson, ML, Karani, J, Portmann, BC, Westaby, D, et al. Natural history and prognostic variables in primary sclerosing cholangitis. Gastroenterology 1991;100(6):1710–17.Google Scholar
Broome, U, Olsson, R, Loof, L, Bodemar, G, Hultcrantz, R, Danielsson, A, et al. Natural history and prognostic factors in 305 Swedish patients with primary sclerosing cholangitis. Gut 1996;38(4):610–15.Google Scholar
Chapman, RW, Arborgh, BA, Rhodes, JM, Summerfield, JA, Dick, R, Scheuer, PJ, et al. Primary sclerosing cholangitis: a review of its clinical features, cholangiography, and hepatic histology. Gut 1980;21(10):870–7.Google Scholar
Weismuller, TJ, Trivedi, PJ, Bergquist, A, Imam, M, Lenzen, H, Ponsioen, CY, et al. Patient age, sex, and inflammatory bowel disease phenotype associate with course of primary sclerosing cholangitis. Gastroenterology 2017;152(8):1975–84 e8.Google Scholar
Bjornsson, E, Boberg, KM, Cullen, S, Fleming, K, Clausen, OP, Fausa, O, et al. Patients with small duct primary sclerosing cholangitis have a favourable long term prognosis. Gut 2002;51(5):731–5.Google Scholar
Scalori, A, Heneghon, MA, Hadzic, ND, Vergani, D, Mieli-Vergani, G. Outcome and survival in childhood onset autoimmune sclerosing cholangitis and autoimmune hepatitis; a 13 years follow-up study. Hepatology 2007;95:1525.Google Scholar
Lindor, KD. Ursodiol for primary sclerosing cholangitis. Mayo Primary Sclerosing Cholangitis-Ursodeoxycholic Acid Study Group. N Engl J Med 1997;336(10):691–5.Google Scholar
van Hoogstraten, HJ, Wolfhagen, FH, van de Meeberg, PC, Kuiper, H, Nix, GA, Becx, MC, et al. Ursodeoxycholic acid therapy for primary sclerosing cholangitis: results of a 2-year randomized controlled trial to evaluate single versus multiple daily doses. J Hepatol 1998;29(3):417–23.Google Scholar
Lindor, KD, Kowdley, KV, Luketic, VA, Harrison, ME, McCashland, T, Befeler, AS, et al. High-dose ursodeoxycholic acid for the treatment of primary sclerosing cholangitis. Hepatology 2009;50(3):808–14.Google Scholar
Benedetti, A, Alvaro, D, Bassotti, C, Gigliozzi, A, Ferretti, G, La Rosa, T, et al. Cytotoxicity of bile salts against biliary epithelium: a study in isolated bile ductule fragments and isolated perfused rat liver. Hepatology 1997;26(1):921.Google Scholar
Fickert, P, Zollner, G, Fuchsbichler, A, Stumptner, C, Weiglein, AH, Lammert, F, et al. Ursodeoxycholic acid aggravates bile infarcts in bile duct-ligated and Mdr2 knockout mice via disruption of cholangioles. Gastroenterology 2002;123(4):1238–51.Google Scholar
Rodrigues, CM, Fan, G, Ma, X, Kren, BT, Steer, CJ. A novel role for ursodeoxycholic acid in inhibiting apoptosis by modulating mitochondrial membrane perturbation. J Clin Invest 1998;101(12):2790–9.Google Scholar
Triantos, CK, Koukias, NM, Nikolopoulou, VN, Burroughs, AK. Meta-analysis: ursodeoxycholic acid for primary sclerosing cholangitis. Aliment Pharmacol Ther 2011;34(8):901–10.Google Scholar
Poropat, G, Giljaca, V, Stimac, D, Gluud, C. Bile acids for primary sclerosing cholangitis. Cochrane Database Syst Rev 2011;1:CD003626.Google Scholar
Gilger, MA, Gann, ME, Opekun, AR, Gleason, WA Jr. Efficacy of ursodeoxycholic acid in the treatment of primary sclerosing cholangitis in children. J Pediatr Gastroenterol Nutr 2000;31(2):136–41.Google Scholar
Deneau, MR, Mack, C, Abdou, R, Amin, M, Amir, A, Auth, M, et al. Gamma glutamyltransferase reduction is associated with favorable outcomes in pediatric primary sclerosing cholangitis. Hepatol Commun 2018;2(11):1369–78.Google Scholar
Cox, KL, Cox, KM. Oral vancomycin: treatment of primary sclerosing cholangitis in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr 1998;27(5):580–3.Google Scholar
Davies, YK, Cox, KM, Abdullah, BA, Safta, A, Terry, AB, Cox, KL. Long-term treatment of primary sclerosing cholangitis in children with oral vancomycin: an immunomodulating antibiotic. J Pediatr Gastroenterol Nutr 2008;47(1):61–7.Google Scholar
Tabibian, JH, Weeding, E, Jorgensen, RA, Petz, JL, Keach, JC, Talwalkar, JA, et al. Randomised clinical trial: vancomycin or metronidazole in patients with primary sclerosing cholangitis – a pilot study. Aliment Pharmacol Ther 2013;37(6):604–12.Google Scholar
Rahimpour, S, Nasiri-Toosi, M, Khalili, H, Ebrahimi-Daryani, N, Nouri-Taromlou, MK, Azizi, Z. A triple blinded, randomized, placebo-controlled clinical trial to evaluate the efficacy and safety of oral vancomycin in primary sclerosing cholangitis: a pilot study. J Gastrointestin Liver Dis 2016;25(4):457–64.Google Scholar
Abarbanel, DN, Seki, SM, Davies, Y, Marlen, N, Benavides, JA, Cox, K, et al. Immunomodulatory effect of vancomycin on Treg in pediatric inflammatory bowel disease and primary sclerosing cholangitis. J Clin Immunol 2013;33(2):397406.Google Scholar
Lindor, KD, Wiesner, RH, Colwell, LJ, Steiner, B, Beaver, S, LaRusso, NF. The combination of prednisone and colchicine in patients with primary sclerosing cholangitis. Am J Gastroenterol 1991;86(1):5761.Google Scholar
Van Thiel, DH, Carroll, P, Abu-Elmagd, K, Rodriguez-Rilo, H, Irish, W, McMichael, J, et al. Tacrolimus (FK 506), a treatment for primary sclerosing cholangitis: results of an open-label preliminary trial. Am J Gastroenterol 1995;90(3):455–9.Google Scholar
Angulo, P, Batts, KP, Jorgensen, RA, LaRusso, NA, Lindor, KD. Oral budesonide in the treatment of primary sclerosing cholangitis. Am J Gastroenterol 2000;95(9):2333–7.Google Scholar
Epstein, MP, Kaplan, MM. A pilot study of etanercept in the treatment of primary sclerosing cholangitis. Dig Dis Sci 2004;49(1):14.Google Scholar
Talwalkar, JA, Angulo, P, Keach, JC, Petz, JL, Jorgensen, RA, Lindor, KD. Mycophenolate mofetil for the treatment of primary sclerosing cholangitis. Am J Gastroenterol 2005;100(2):308–12.Google Scholar
Peng, X, Luo, X, Hou, JY, Wu, SY, Li, LZ, Zheng, MH, et al. Immunosuppressive agents for the treatment of primary sclerosing cholangitis: a systematic review and meta-analysis. Dig Dis 2017;35(5):478–85.Google Scholar
Lynch, KD, Chapman, RW, Keshav, S, Montano-Loza, AJ, Mason, AL, Kremer, AE, et al. Effects of vedolizumab in patients with primary sclerosing cholangitis and inflammatory bowel diseases. Clin Gastroenterol Hepatol 2019; S1542–3565(19)30523-3. Doi: 10.1016/j.cgh.2019.05.013 [Epub ahead of print].Google Scholar
Smolka, V, Karaskova, E, Tkachyk, O, Aiglova, K, Ehrmann, J, Michalkova, K, et al. Long-term follow-up of children and adolescents with primary sclerosing cholangitis and autoimmune sclerosing cholangitis. Hepatobiliary Pancreat Dis Int 2016;15(4):412–18.Google Scholar
Muir, AJ, Levy, C, Janssen, HLA, Montano-Loza, AJ, Shiffman, ML, Caldwell, S, et al. Simtuzumab for primary sclerosing cholangitis: phase 2 study results with insights on the natural history of the disease. Hepatology 2019;69(2):684–98.Google Scholar
Graziadei, IW, Wiesner, RH, Marotta, PJ, Porayko, MK, Hay, JE, Charlton, MR, et al. Long-term results of patients undergoing liver transplantation for primary sclerosing cholangitis. Hepatology 1999;30(5):1121–7.Google Scholar
Miloh, T, Anand, R, Yin, W, Vos, M, Kerkar, N, Alonso, E, et al. Pediatric liver transplantation for primary sclerosing cholangitis. Liver Transpl 2011;17(8):925–33.Google Scholar
Steenstraten, IC, Sebib Korkmaz, K, Trivedi, PJ, Inderson, A, van Hoek, B, Rodriguez Girondo, MDM, et al. Systematic review with meta-analysis: risk factors for recurrent primary sclerosing cholangitis after liver transplantation. Aliment Pharmacol Ther 2019;49(6):636–43.Google Scholar
Trivedi, PJ, Reece, J, Laing, RW, Slaney, E, Cooney, R, Gunson, BK, et al. The impact of ileal pouch-anal anastomosis on graft survival following liver transplantation for primary sclerosing cholangitis. Aliment Pharmacol Ther 2018;48(3):322–32.Google Scholar
Soufi, N, Bazerbachi, F, Deneau, M. Post-transplant disease recurrence in pediatric PSC. Curr Gastroenterol Rep 2018;20(9):44.Google Scholar
Gossard, AA, Angulo, P, Lindor, KD. Secondary sclerosing cholangitis: a comparison to primary sclerosing cholangitis. Am J Gastroenterol 2005;100(6):1330–3.Google Scholar
Suskind, DL, Finn, L, Wahbeh, G, Christie, D, Horslen, S. A child with Kabuki syndrome and primary sclerosing cholangitis successfully treated with ursodiol and cholestryamine. J Pediatr Gastroenterol Nutr 2006;43(4):542–4.Google Scholar
Hadj-Rabia, S, Baala, L, Vabres, P, Hamel-Teillac, D, Jacquemin, E, Fabre, M, et al. Claudin-1 gene mutations in neonatal sclerosing cholangitis associated with ichthyosis: a tight junction disease. Gastroenterology 2004;127(5):1386–90.Google Scholar
Grammatikopoulos, T, Sambrotta, M, Strautnieks, S, Foskett, P, Knisely, AS, Wagner, B, et al. Mutations in DCDC2 (doublecortin domain containing protein 2) in neonatal sclerosing cholangitis. J Hepatol 2016;65(6):1179–87.Google Scholar

References

Ferrajolo, C, Capuano, A, Verhamme, KM, et al. Drug-induced hepatic injury in children: a case/non-case study of suspected adverse drug reactions in VigiBase. Br J Clin Pharmacol 2010;70:721–8.Google Scholar
Clarkson, A, Choonara, I. Surveillance for fatal suspected adverse drug reactions in the UK. Arch Dis Child 2002;87:462–7.Google Scholar
Squires, RH Jr., Shneider, BL, Bucuvalas, J, et al. Acute liver failure in children: the first 348 patients in the pediatric acute liver failure study group. J Pediatr 2006;148:652–8.Google Scholar
Aithal, GP, Watkins, PB, Andrade, RJ, et al. Case definition and phenotype standardization in drug-induced liver injury. Clin Pharmacol Ther 2011;89:806–15.Google Scholar
Zimmerman, HJ. (1999). Hepatotoxicity: The Adverse Effects of Drugs and Other Chemicals on the Liver, 2nd edn. Philadelphia: Lippincott Williams & Wilkins.Google Scholar
Kaplowitz, N, DeLeve, L. (2013). Drug-Induced Liver Disease, 3rd edn. (p. 776). Waltham, MA: Academic Press.Google Scholar
Ingelman-Sundberg, M. Human drug metabolising cytochrome P450 enzymes: properties and polymorphisms. Naunyn Schmiedebergs Arch Pharmacol 2004;369:89104.Google Scholar
McGraw, J, Waller, D. Cytochrome P450 variations in different ethnic populations. Expert Opin Drug Metab Toxicol 2012;8:371–82.Google Scholar
Weinshilboum, R. Inheritance and drug response. N Engl J Med 2003;348:529–37.Google Scholar
Cascorbi, I. Genetic basis of toxic reactions to drugs and chemicals. Toxicol Lett 2006;162:1628.Google Scholar
Russmann, S, Jetter, A, Kullak-Ublick, GA. Pharmacogenetics of drug-induced liver injury. Hepatology 2010;52:748–61.Google Scholar
Ehmer, U, Kalthoff, S, Fakundiny, B, et al. Gilbert syndrome redefined: a complex genetic haplotype influences the regulation of glucuronidation. Hepatology 2012;55:1912–21.Google Scholar
Burchell, B, Soars, M, Monaghan, G, et al. Drug-mediated toxicity caused by genetic deficiency of UDP-glucuronosyltransferases. Toxicol Lett 2000;112 –13:333–40.Google Scholar
Pessayre, D, Fromenty, B, Berson, A, et al. Central role of mitochondria in drug-induced liver injury. Drug Metab Rev 2012;44:3487.Google Scholar
Lucena, MI, Garcia-Martin, E, Andrade, RJ, et al. Mitochondrial superoxide dismutase and glutathione peroxidase in idiosyncratic drug-induced liver injury. Hepatology 2010;52:303–12.Google Scholar
Bessone, F, Dirchwolf, M, Rodil, MA, et al. Review article: drug-induced liver injury in the context of nonalcoholic fatty liver disease – a physiopathological and clinical integrated view. Aliment Pharmacol Ther 2018;48:892913.Google Scholar
He, K, Cai, L, Shi, Q, et al. Inhibition of MDR3 activity in human hepatocytes by drugs associated with liver injury. Chem Res Toxicol 2015;28:1987–90.Google Scholar
Hines, RN, McCarver, DG. The ontogeny of human drug-metabolizing enzymes: phase I oxidative enzymes. J Pharmacol Exp Ther 2002;300:355–60.Google Scholar
Kearns, GL, Abdel-Rahman, SM, Alander, SW, et al. Developmental pharmacology–drug disposition, action, and therapy in infants and children. N Engl J Med 2003;349:1157–67.Google Scholar
Tateishi, T, Nakura, H, Asoh, M, et al. A comparison of hepatic cytochrome P450 protein expression between infancy and postinfancy. Life Sci 1997;61:2567–74.Google Scholar
Aranda, JV, Collinge, JM, Zinman, R, et al. Maturation of caffeine elimination in infancy. Arch Dis Child 1979;54:946–9.Google Scholar
Treluyer, JM, Gueret, G, Cheron, G, et al. Developmental expression of CYP2C and CYP2C-dependent activities in the human liver: in-vivo/in-vitro correlation and inducibility. Pharmacogenetics 1997;7:441–52.Google Scholar
Lang, C, Meier, Y, Stieger, B, et al. Mutations and polymorphisms in the bile salt export pump and the multidrug resistance protein 3 associated with drug-induced liver injury. Pharmacogenet Genomics 2007;17:4760.Google Scholar
Molleston, JP, Fontana, RJ, Lopez, MJ, et al. Characteristics of idiosyncratic drug-induced liver injury in children: results from the DILIN prospective study. J Pediatr Gastroenterol Nutr 2011;53:182–9.Google Scholar
Walsh, SA, Creamer, D. Drug reaction with eosinophilia and systemic symptoms (DRESS): a clinical update and review of current thinking. Clin Exp Dermatol 2011;36:611.Google Scholar
Cacoub, P, Musette, P, Descamps, V, et al. The DRESS syndrome: a literature review. Am J Med 2011;124:588–97.Google Scholar
Devarbhavi, H, Karanth, D, Prasanna, KS, et al. Drug-induced liver injury with hypersensitivity features has a better outcome: a single-center experience of 39 children and adolescents. Hepatology 2011;54:1344–50.Google Scholar
Gudnason, HO, Bjornsson, HK, Gardarsdottir, M, et al. Secondary sclerosing cholangitis in patients with drug-induced liver injury. Dig Liver Dis 2015;47:502–7.Google Scholar
Jaeschke, H, Gores, GJ, Cederbaum, AI, et al. Mechanisms of hepatotoxicity. Toxicol Sci 2002;65:166–76.Google Scholar
James, LP, Farrar, HC, Darville, TL, et al. Elevation of serum interleukin 8 levels in acetaminophen overdose in children and adolescents. Clin Pharmacol Ther 2001;70:280–6.Google Scholar
Ju, C, Reilly, TP, Bourdi, M, et al. Protective role of Kupffer cells in acetaminophen-induced hepatic injury in mice. Chem Res Toxicol 2002;15:1504–13.Google Scholar
Aithal, GP, Ramsay, L, Daly, AK, et al. Hepatic adducts, circulating antibodies, and cytokine polymorphisms in patients with diclofenac hepatotoxicity. Hepatology 2004;39:1430–40.Google Scholar
Kaddurah-Daouk, R, Weinshilboum, R, Pharmacometabolomics Research. Metabolomic signatures for drug response phenotypes: pharmacometabolomics enables precision medicine. Clin Pharmacol Ther 2015;98:71–5.Google Scholar
Enright, EF, Gahan, CG, Joyce, SA, et al. The impact of the gut microbiota on drug metabolism and clinical outcome. Yale J Biol Med 2016;89:375–82.Google Scholar
Massart, J, Begriche, K, Moreau, C, et al. Role of nonalcoholic fatty liver disease as risk factor for drug-induced hepatotoxicity. J Clin Transl Res 2017;3:212–32.Google Scholar
DiPaola, F, Molleston, JP, Gu, J, et al. Antimicrobials and antiepileptics are the leading causes of idiosyncratic drug-induced liver injury in American children. J Pediatr Gastroenterol Nutr 2019;69:152–9.Google Scholar
Mahadevan, SB, McKiernan, PJ, Davies, P, et al. Paracetamol induced hepatotoxicity. Arch Dis Child 2006;91:598603.Google Scholar
Makin, AJ, Wendon, J, Williams, R. A 7-year experience of severe acetaminophen-induced hepatotoxicity (1987–1993). Gastroenterology 1995;109:1907–16.Google Scholar
Smilkstein, MJ, Knapp, GL, Kulig, KW, et al. Efficacy of oral N-acetylcysteine in the treatment of acetaminophen overdose. N Engl J Med 1988;319:1557–62.Google Scholar
Harrison, PM, Keays, R, Bray, GP, et al. Improved outcome of paracetamol-induced fulminant hepatic failure by late administration of acetylcysteine. Lancet 1990;335:1572–3.Google Scholar
Keays, R, Harrison, PM, Wendon, JA, et al. Intravenous acetylcysteine in paracetamol induced fulminant hepatic failure: a prospective controlled trial. BMJ 1991;303:1026–9.Google Scholar
Yarema, MC, Johnson, DW, Berlin, RJ, et al. Comparison of the 20-hour intravenous and 72-hour oral acetylcysteine protocols for the treatment of acute acetaminophen poisoning. Ann Emerg Med 2009;54:606–14.Google Scholar
Gosselin, S, Juurlink, DN, Kielstein, JT, et al. Extracorporeal treatment for acetaminophen poisoning: recommendations from the EXTRIP workgroup. Clin Toxicol (Phila) 2014;52:856–67.Google Scholar
James, LP, Wells, E, Beard, RH, et al. Predictors of outcome after acetaminophen poisoning in children and adolescents. J Pediatr 2002;140:522–6.Google Scholar
Heubi, JE, Barbacci, MB, Zimmerman, HJ. Therapeutic misadventures with acetaminophen: hepatoxicity after multiple doses in children. J Pediatr 1998;132:22–7.Google Scholar
Leonis, MA, Alonso, EM, Im, K, et al. Chronic acetaminophen exposure in pediatric acute liver failure. Pediatrics 2013;131:e740–6.Google Scholar
Rajanayagam, J, Bishop, JR, Lewindon, PJ, et al. Paracetamol-associated acute liver failure in Australian and New Zealand children: high rate of medication errors. Arch Dis Child 2015;100:7780.Google Scholar
Acheampong, P, Thomas, SH. Determinants of hepatotoxicity after repeated supratherapeutic paracetamol ingestion: systematic review of reported cases. Br J Clin Pharmacol 2016;82:923–31.Google Scholar
Watkins, PB, Kaplowitz, N, Slattery, JT, et al. Aminotransferase elevations in healthy adults receiving 4 grams of acetaminophen daily: a randomized controlled trial. JAMA 2006;296:8793.Google Scholar
Savino, F, Lupica, MM, Tarasco, V, et al. Fulminant hepatitis after 10 days of acetaminophen treatment at recommended dosage in an infant. Pediatrics 2011;127:e494–7.Google Scholar
Iorio, ML, Cheerharan, M, Kaufman, SS, et al. Acute liver failure following cleft palate repair: a case of therapeutic acetaminophen toxicity. Cleft Palate Craniofac J 2013;50:747–50.Google Scholar
Webster, PA, Roberts, DW, Benson, RW, et al. Acetaminophen toxicity in children: diagnostic confirmation using a specific antigenic biomarker. J Clin Pharmacol 1996;36:397402.Google Scholar
Bhattacharyya, S, Yan, K, Pence, L, et al. Targeted liquid chromatography-mass spectrometry analysis of serum acylcarnitines in acetaminophen toxicity in children. Biomark Med 2014;8:147–59.Google Scholar
Yang, X, Salminen, WF, Shi, Q, et al. Potential of extracellular microRNAs as biomarkers of acetaminophen toxicity in children. Toxicol Appl Pharmacol 2015;284:180–7.Google Scholar
Jaeschke, H, Knight, TR, Bajt, ML. The role of oxidant stress and reactive nitrogen species in acetaminophen hepatotoxicity. Toxicol Lett 2003;144:279–88.Google Scholar
Fannin, RD, Russo, M, O’Connell, TM, et al. Acetaminophen dosing of humans results in blood transcriptome and metabolome changes consistent with impaired oxidative phosphorylation. Hepatology 2010;51:227–36.Google Scholar
Goldring, CE, Kitteringham, NR, Elsby, R, et al. Activation of hepatic Nrf2 in vivo by acetaminophen in CD-1 mice. Hepatology 2004;39:1267–76.Google Scholar
Liu, HH, Lu, P, Guo, Y, et al. An integrative genomic analysis identifies Bhmt2 as a diet-dependent genetic factor protecting against acetaminophen-induced liver toxicity. Genome Res 2010;20:2835.Google Scholar
Harrill, AH, Watkins, PB, Su, S, et al. Mouse population-guided resequencing reveals that variants in CD44 contribute to acetaminophen-induced liver injury in humans. Genome Res 2009;19:1507–15.Google Scholar
Zamora, R, Barclay, D, Yin, J, et al. HMGB1 is a central driver of dynamic pro-inflammatory networks in pediatric acute liver failure induced by acetaminophen. Sci Rep 2019;9:5971.Google Scholar
Bourdi, M, Masubuchi, Y, Reilly, TP, et al. Protection against acetaminophen-induced liver injury and lethality by interleukin 10: role of inducible nitric oxide synthase. Hepatology 2002;35:289–98.Google Scholar
Connolly, MK, Ayo, D, Malhotra, A, et al. Dendritic cell depletion exacerbates acetaminophen hepatotoxicity. Hepatology 2011;54:959–68.Google Scholar
Peterson, RG, Rumack, BH. Age as a variable in acetaminophen overdose. Arch Intern Med 1981;141:390–3.Google Scholar
Rumack, BH. Acetaminophen overdose in young children. Treatment and effects of alcohol and other additional ingestants in 417 cases. Am J Dis Child 1984;138:428–33.Google Scholar
Peterson, RG, Rumack, BH. Pharmacokinetics of acetaminophen in children. Pediatrics 1978;62:877–9.Google Scholar
Hickson, GB, Altemeier, WA, Martin, ED, et al. Parental administration of chemical agents: a cause of apparent life threatening events. J Pediatr 1989;83:772–6.Google Scholar
Pacifici, GM, Allegaert, K. Clinical pharmacology of paracetamol in neonates: a review. Curr Ther Res Clin Exp 2015;77:2430.Google Scholar
Cook, SF, Stockmann, C, Samiee-Zafarghandy, S, et al. Neonatal maturation of paracetamol (acetaminophen) glucuronidation, sulfation, and oxidation based on a parent-metabolite population pharmacokinetic model. Clin Pharmacokinet 2016;55:13951411.Google Scholar
Flint, RB, Roofthooft, DW, van Rongen, A, et al. Exposure to acetaminophen and all its metabolites upon 10, 15, and 20 mg/kg intravenous acetaminophen in very-preterm infants. Pediatr Res 2017;82:678–84.Google Scholar
Walls, L, Baker, CF, Sarkar, S. Acetaminophen-induced hepatic failure with encephalopathy in a newborn. J Perinatol 2007;27:133–5.Google Scholar
Ogilvie, JD, Rieder, MJ, Lim, R. Acetaminophen overdose in children. CMAJ 2012;184:1492–6.Google Scholar
Palmer, GM, Atkins, M, Anderson, BJ, et al. I.V. acetaminophen pharmacokinetics in neonates after multiple doses. Br J Anaesth 2008;101:523–30.Google Scholar
Rumack, BH. Acetaminophen misconceptions. Hepatology 2004;40:1015.Google Scholar
Al-Sinani, S, Al-Rawas, A, Dhawan, A. Mercury as a cause of fulminant hepatic failure in a child: case report and literature review. Clin Res Hepatol Gastroenterol 2011;35:580–2.Google Scholar
Zimmerman, HJ, Maddrey, WC. Acetaminophen (paracetamol) hepatotoxicity with regular intake of alcohol: analysis of instances of therapeutic misadventure. Hepatology 1995;22:767–73.Google Scholar
Bruun, LS, Elkjaer, S, Bitsch-Larsen, D, et al. Hepatic failure in a child after acetaminophen and sevoflurane exposure. Anesth Analg 2001;92:1446–8.Google Scholar
Ceschi, A, Hofer, KE, Rauber-Luthy, C, et al. Paracetamol orodispersible tablets: a risk for severe poisoning in children? Eur J Clin Pharmacol 2011;67:97–9.Google Scholar
Dart, RC, Rumack, BH. Intravenous acetaminophen in the United States: iatrogenic dosing errors. Pediatrics 2012;129:349–53.Google Scholar
Berling, I, Anscombe, M, Isbister, GK. Intravenous paracetamol toxicity in a malnourished child. Clin Toxicol (Phila) 2012;50:74–6.Google Scholar
Yagupsky, P, Gazala, E, Sofer, S. Fatal hepatic failure and encephalopathy associated with amiodarone therapy. J Pediatr 1985;107:967–70.Google Scholar
Floyd, J, Mirza, I, Sachs, B, et al. Hepatotoxicity of chemotherapy. Semin Oncol 2006;33:5067.Google Scholar
Pratibha, R, Sameer, R, Rataboli, PV, et al. Enzymatic studies of cisplatin induced oxidative stress in hepatic tissue of rats. Eur J Pharmacol 2006;532:290–3.Google Scholar
Lu, Y, Cederbaum, AI. Cisplatin-induced hepatotoxicity is enhanced by elevated expression of cytochrome P450 2E1. Toxicol Sci 2006;89:515–23.Google Scholar
Hruban, RH, Sternberg, SS, Meyers, P, et al. Fatal thrombocytopenia and liver failure associated with carboplatin therapy. Cancer Invest 1991;9:263–8.Google Scholar
Honjo, I, Suou, T, Hirayama, C. Hepatotoxicity of cyclophosphamide in man: pharmacokinetic analysis. Res Commun Chem Pathol Pharmacol 1988;61:149–65.Google Scholar
Berkovitch, M, Matsui, D, Zipursky, A, et al. Hepatotoxicity of 6-mercaptopurine in childhood acute lymphocytic leukemia: pharmacokinetic characteristics. Med Pediatr Oncol 1996;26:85–9.Google Scholar
Hazar, V, Kutluk, T, Akyuz, C, et al. Veno-occlusive disease-like hepatotoxicity in two children receiving chemotherapy for Wilms’ tumor and clear cell sarcoma of kidney. Pediatr Hematol Oncol 1998;15:85–9.Google Scholar
Shi, Q, Yang, X, Greenhaw, JJ, et al. Drug-induced liver injury in children: clinical observations, animal models, and regulatory status. Int J Toxicol 2017;36:365–79.Google Scholar
Pratt, CB, Johnson, WW. Duration and severity of fatty metamorphosis of the liver following L-asparaginase therapy. Cancer 1971;28:361–4.Google Scholar
Sahoo, S, Hart, J. Histopathological features of L-asparaginase-induced liver disease. Semin Liver Dis 2003;23:295–9.Google Scholar
Hijiya, N, van der Sluis, IM. Asparaginase-associated toxicity in children with acute lymphoblastic leukemia. Leuk Lymphoma 2016;57:748–57.Google Scholar
Topley, J, Benson, J, Squier, MV, et al. Hepatotoxicity in the treatment of acute lymphoblastic leukemia. Med Pediatr Oncol 1979;7:393–9.Google Scholar
Stoneham, S, Lennard, L, Coen, P, et al. Veno-occlusive disease in patients receiving thiopurines during maintenance therapy for childhood acute lymphoblastic leukaemia. Br J Haematol 2003;123:100–2.Google Scholar
Broxson, EH, Dole, M, Wong, R, et al. Portal hypertension develops in a subset of children with standard risk acute lymphoblastic leukemia treated with oral 6-thioguanine during maintenance therapy. Pediatr Blood Cancer 2005;44:226–31.Google Scholar
Rawat, D, Gillett, PM, Devadason, D, et al. Long-term follow-up of children with 6-thioguanine-related chronic hepatotoxicity following treatment for acute lymphoblastic leukaemia. J Pediatr Gastroenterol Nutr 2011;53:478–9.Google Scholar
Geller, SA, Dubinsky, MC, Poordad, FF, et al. Early hepatic nodular hyperplasia and submicroscopic fibrosis associated with 6-thioguanine therapy in inflammatory bowel disease. Am J Surg Pathol 2004;28:1204–11.Google Scholar
DeLeve, LD, Shulman, HM, McDonald, GB. Toxic injury to hepatic sinusoids: sinusoidal obstruction syndrome (veno-occlusive disease). Semin Liver Dis 2002;22:2742.Google Scholar
D’Antiga, L, Baker, A, Pritchard, J, et al. Veno-occlusive disease with multi-organ involvement following actinomycin-D. Eur J Cancer 2001;37:1141–8.Google Scholar
Barker, CC, Anderson, RA, Sauve, RS, et al. GI complications in pediatric patients post-BMT. Bone Marrow Transplant 2005;36:51–8.Google Scholar
Sulis, ML, Bessmertny, O, Granowetter, L, et al. Veno-occlusive disease in pediatric patients receiving actinomycin D and vincristine only for the treatment of rhabdomyosarcoma. J Pediatr Hematol Oncol 2004;26:843–6.Google Scholar
Elli, M, Pinarli, FG, Dagdemir, A, et al. Veno-occlusive disease of the liver in a child after chemotherapy for brain tumor. Pediatr Blood Cancer 2006;46:521–3.Google Scholar
McDonald, GB, Sharma, P, Matthews, DE, et al. The clinical course of 53 patients with veno-occlusive disease of the liver after marrow transplantation. Transplantation 1985;39:603–8.Google Scholar
McDonald, GB, Slattery, JT, Bouvier, ME, et al. Cyclophosphamide metabolism, liver toxicity, and mortality following hematopoietic stem cell transplantation. Blood 2003;101:2043–8.Google Scholar
El-Sayed, MH, El-Haddad, A, Fahmy, OA, et al. Liver disease is a major cause of mortality following allogeneic bone-marrow transplantation. Eur J Gastroenterol Hepatol 2004;16:1347–54.Google Scholar
Srivastava, A, Poonkuzhali, B, Shaji, RV, et al. Glutathione S-transferase M1 polymorphism: a risk factor for hepatic venoocclusive disease in bone marrow transplantation. Blood 2004;104:1574–7.Google Scholar
Reiss, U, Cowan, M, McMillan, A, et al. Hepatic venoocclusive disease in blood and bone marrow transplantation in children and young adults: incidence, risk factors, and outcome in a cohort of 241 patients. J Pediatr Hematol Oncol 2002;24:746–50.Google Scholar
Ravikumara, M, Hill, FG, Wilson, DC, et al. 6-Thioguanine-related chronic hepatotoxicity and variceal haemorrhage in children treated for acute lymphoblastic leukaemia–a dual-centre experience. J Pediatr Gastroenterol Nutr 2006;42:535–8.Google Scholar
Corbacioglu, S, Greil, J, Peters, C, et al. Defibrotide in the treatment of children with veno-occlusive disease (VOD): a retrospective multicentre study demonstrates therapeutic efficacy upon early intervention. Bone Marrow Transplant 2004;33:189–95.Google Scholar
Shin-Nakai, N, Ishida, H, Yoshihara, T, et al. Control of hepatic veno-occlusive disease with an antithrombin-III concentrate-based therapy. Pediatr Int 2006;48:85–7.Google Scholar
Ringden, O, Remberger, M, Lehmann, S, et al. N-acetylcysteine for hepatic veno-occlusive disease after allogeneic stem cell transplantation. Bone Marrow Transplant 2000;25:993–6.Google Scholar
Lee, AC, Goh, PY. Dactinomycin-induced hepatic sinusoidal obstruction syndrome responding to treatment with N-acetylcysteine. J Cancer 2011;2:527–31.Google Scholar
DeLeve, LD. Dacarbazine toxicity in murine liver cells: a model of hepatic endothelial injury and glutathione defense. J Pharmacol Exp Ther 1994;268:1261–70.Google Scholar
DeLeve, LD. Cellular target of cyclophosphamide toxicity in the murine liver: role of glutathione and site of metabolic activation. Hepatology 1996;24:830–7.Google Scholar
DeLeve, LD, Wang, X, Kanel, GC, et al. Decreased hepatic nitric oxide production contributes to the development of rat sinusoidal obstruction syndrome. Hepatology 2003;38:900–8.Google Scholar
Iguchi, A, Kobayashi, R, Yoshida, M, et al. Vascular endothelial growth factor (VEGF) is one of the cytokines causative and predictive of hepatic veno-occlusive disease (VOD) in stem cell transplantation. Bone Marrow Transplant 2001;27:1173–80.Google Scholar
Suzman, DL, Pelosof, L, Rosenberg, A, et al. Hepatotoxicity of immune checkpoint inhibitors: an evolving picture of risk associated with a vital class of immunotherapy agents. Liver Int 2018;38:976–87.Google Scholar
Chen, TC, Ng, KF, Jeng, LB, et al. Aspirin-related hepatotoxicity in a child after liver transplant. Dig Dis Sci 2001;46:486–8.Google Scholar
Hamdan, JA, Manasra, K, Ahmed, M. Salicylate-induced hepatitis in rheumatic fever. Am J Dis Child 1985;139:453–5.Google Scholar
Dinakaran, D, Bristow, E, Armanious, H, et al. Co-ingestion of willow bark tea and acetaminophen associated with fatal infantile fulminant liver failure. Pediatr Int 2017;59:743–5.Google Scholar
Raza, A, Vierling, J, Hussain, KB. Genetics of drug-induced hepatotoxicity toxicity in Gilbert’s syndrome. Am J Gastroenterol 2013;108:1936–7.Google Scholar
Dinh, JC, Pearce, RE, Van Haandel, L, et al. Characterization of atomoxetine biotransformation and implications for development of PBPK models for dose individualization in children. Drug Metab Dispos 2016;44:1070–9.Google Scholar
Stojanovski, SD, Casavant, MJ, Mousa, HM, et al. Atomoxetine-induced hepatitis in a child. Clin Toxicol (Phila) 2007;45:51–5.Google Scholar
Erdogan, A, Ozcay, F, Piskin, E, et al. Idiosyncratic liver failure probably associated with atomoxetine: a case report. J Child Adolesc Psychopharmacol 2011;21:295–7.Google Scholar
Lim, JR, Faught, PR, Chalasani, NP, et al. Severe liver injury after initiating therapy with atomoxetine in two children. J Pediatr 2006;148:831–4.Google Scholar
Brown, JT, Abdel-Rahman, SM, van Haandel, L, et al. Single dose, CYP2D6 genotype-stratified pharmacokinetic study of atomoxetine in children with ADHD. Clin Pharmacol Ther 2016;99:642–50.Google Scholar
DePinho, RA, Goldberg, CS, Lefkowitch, JH. Azathioprine and the liver. Evidence favoring idiosyncratic, mixed cholestatic-hepatocellular injury in humans. Gastroenterology 1984;86:162–5.Google Scholar
Sterneck, M, Wiesner, R, Ascher, N, et al. Azathioprine hepatotoxicity after liver transplantation. Hepatology 1991;14:806–10.Google Scholar
Seiderer, J, Zech, CJ, Diebold, J, et al. Nodular regenerative hyperplasia: a reversible entity associated with azathioprine therapy. Eur J Gastroenterol Hepatol 2006;18:553–5.Google Scholar
Bjornsson, ES, Gu, J, Kleiner, DE, et al. Azathioprine and 6-mercaptopurine-induced liver injury: clinical features and outcomes. J Clin Gastroenterol 2017;51:63–9.Google Scholar
Yang, SK, Hong, M, Baek, J, et al. A common missense variant in NUDT15 confers susceptibility to thiopurine-induced leukopenia. Nat Genet 2014;46:1017–20.Google Scholar
Wang, CH, Chi, MH, Lin, JY, et al. NUDT15 polymorphism identified in a patient with azathioprine hypersensitivity syndrome presenting as erythema nodosum and hepatotoxicity. Br J Dermatol 2019;181:631–2.Google Scholar
Lennard, L. The clinical pharmacology of 6-mercaptopurine. Eur J Clin Pharmacol 1992;43:329–39.Google Scholar
Adam de Beaumais, T, Fakhoury, M, Medard, Y, et al. Determinants of mercaptopurine toxicity in paediatric acute lymphoblastic leukemia maintenance therapy. Br J Clin Pharmacol 2011;71:575–84.Google Scholar
Rosh, JR, Gross, T, Mamula, P, et al. Hepatosplenic T-cell lymphoma in adolescents and young adults with Crohn’s disease: a cautionary tale? Inflamm Bowel Dis 2007;13:1024–30.Google Scholar
Ochenrider, MG, Patterson, DJ, Aboulafia, DM. Hepatosplenic T-cell lymphoma in a young man with Crohn’s disease: case report and literature review. Clin Lymphoma Myeloma Leuk 2010;10:144–8.Google Scholar
Deepak, P, Sifuentes, H, Sherid, M, et al. T-cell non-Hodgkin’s lymphomas reported to the FDA AERS with tumor necrosis factor-alpha (TNF-alpha) inhibitors: results of the REFURBISH study. Am J Gastroenterol 2013;108:99105.Google Scholar
Bedan, M, Grimm, D, Wehland, M, et al. A focus on macitentan in the treatment of pulmonary arterial hypertension. Basic Clin Pharmacol Toxicol 2018;123:103–13.Google Scholar
Fattinger, K, Funk, C, Pantze, M, et al. The endothelin antagonist bosentan inhibits the canalicular bile salt export pump: a potential mechanism for hepatic adverse reactions. Clin Pharmacol Ther 2001;69:223–31.Google Scholar
Wei, A, Gu, Z, Li, J, et al. Clinical adverse effects of endothelin receptor antagonists: insights from the meta-analysis of 4894 patients from 24 randomized double-blind placebo-controlled clinical trials. J Am Heart Assoc 2016;5(11):e003896.Google Scholar
Eriksson, C, Gustavsson, A, Kronvall, T, et al. Hepatotoxicity by bosentan in a patient with portopulmonary hypertension: a case-report and review of the literature. J Gastrointestin Liver Dis 2011;20:7780.Google Scholar
Beghetti, M, Hoeper, MM, Kiely, DG, et al. Safety experience with bosentan in 146 children 2–11 years old with pulmonary arterial hypertension: results from the European Postmarketing Surveillance Program. Pediatr Res 2008;64:200–4.Google Scholar
Berger, RM, Haworth, SG, Bonnet, D, et al. FUTURE-2: results from an open-label, long-term safety and tolerability extension study using the pediatric formulation of bosentan in pulmonary arterial hypertension. Int J Cardiol 2016;202:52–8.Google Scholar
Shear, NH, Spielberg, SP. Anticonvulsant hypersensitivity syndrome. In vitro assessment of risk. J Clin Invest 1988;82:1826–32.Google Scholar
Zucker, P, Daum, F, Cohen, MI. Fatal carbamazepine hepatitis. J Pediatr 1977;91:667–8.Google Scholar
Smith, DW, Cullity, GJ, Silberstein, EP. Fatal hepatic necrosis associated with multiple anticonvulsant therapy. Aust N Z J Med 1988;18:575–81.Google Scholar
Hadzic, N, Portmann, B, Davies, ET, et al. Acute liver failure induced by carbamazepine. Arch Dis Child 1990;65:315–17.Google Scholar
Morales-Diaz, M, Pinilla-Roa, E, Ruiz, I. Suspected carbamazepine-induced hepatotoxicity. Pharmacotherapy 1999;19:252–5.Google Scholar
Sierra, NM, Garcia, B, Marco, J, et al. Cross hypersensitivity syndrome between phenytoin and carbamazepine. Pharm World Sci 2005;27:170–4.Google Scholar
Aouam, K, Ben Romdhane, F, Loussaief, C, et al. Hypersensitivity syndrome induced by anticonvulsants: possible cross-reactivity between carbamazepine and lamotrigine. J Clin Pharmacol 2009;49:1488–91.Google Scholar
Yip, VLM, Meng, X, Maggs, JL, et al. Mass spectrometric characterization of circulating covalent protein adducts derived from epoxide metabolites of carbamazepine in patients. Chem Res Toxicol 2017;30:1419–35.Google Scholar
Ju, C, Uetrecht, J. Detection of 2-hydroxyiminostilbene in the urine of patients taking carbamazepine and its oxidation to a reactive iminequinone intermediate. J Pharmacol Exp Ther 1999;288:51–6.Google Scholar
McCormack, M, Alfirevic, A, Bourgeois, S, et al. HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans. N Engl J Med 2011;364:1134–43.Google Scholar
Phillips, EJ, Sukasem, C, Whirl-Carrillo, M, et al. Clinical Pharmacogenetics Implementation Consortium guideline for HLA genotype and use of carbamazepine and oxcarbazepine: 2017 update. Clin Pharmacol Ther 2018;103:574–81.Google Scholar
D’Orazio, JL. Oxcarbazepine-induced Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS). Clin Toxicol (Phila) 2008;46:1093–4.Google Scholar
Bosdure, E, Cano, A, Roquelaure, B, et al. Oxcarbazepine and DRESS syndrome: a paediatric cause of acute liver failure. Arch Pediatr 2004;11:1073–7.Google Scholar
Wanless, IR, Dore, S, Gopinath, G, et al. Histopathology of cocaine hepatotoxicity. Report of four cases. Gastroenterology 1990;98:497501.Google Scholar
Kassianides, C, Nussenblatt, R, Palestine, AG, et al. Liver injury from cyclosporine A. Dig Dis Sci 1990;35:693–7.Google Scholar
Brauer, RB, Heidecke, CD, Nathrath, W, et al. Liver transplantation for the treatment of fulminant hepatic failure induced by the ingestion of ecstasy. Transpl Int 1997;10:229–33.Google Scholar
Andreu, V, Mas, A, Bruguera, M, et al. Ecstasy: a common cause of severe acute hepatotoxicity. J Hepatol 1998;29:394–7.Google Scholar
Greene, SL, Dargan, PI, O’Connor, N, et al. Multiple toxicity from 3,4-methylenedioxymethamphetamine (“Ecstasy”). Am J Emerg Med 2003;21:121–4.Google Scholar
Smith, ID, Simpson, KJ, Garden, OJ, et al. Non-paracetamol drug-induced fulminant hepatic failure among adults in Scotland. Eur J Gastroenterol Hepatol 2005;17:161–7.Google Scholar
Antolino-Lobo, I, Meulenbelt, J, van den Berg, M, et al. A mechanistic insight into 3,4-methylenedioxymethamphetamine (“Ecstasy”)-mediated hepatotoxicity. Vet Q 2011;31:193205.Google Scholar
Ellis, AJ, Wendon, JA, Portmann, B, et al. Acute liver damage and ecstasy ingestion. Gut 1996;38:454–8.Google Scholar
Munoz, P, Drobinska, A, Bianchi, L, et al. Acute giant cell hepatitis in a 17-year-old man. Schweiz Rundsch Med Prax 2004;93:2109–12.Google Scholar
Duffy, MR, Swart, M. Severe Ecstasy poisoning in a toddler. Anaesthesia 2006;61:498501.Google Scholar
Yin, S. Adolescents and drug abuse: 21st century synthetic substances. Clin Pedaitr Emerg Med 2019;20:1724.Google Scholar
Hood, B, Nowicki, MJ. Eosinophilic hepatitis in an adolescent during lisdexamfetamine dimesylate treatment for ADHD. Pediatrics 2010;125:e1510–13.Google Scholar
Zafrani, ES, Ishak, KG, Rudzki, C. Cholestatic and hepatocellular injury associated with erythromycin esters. Report of nine cases. Dig Dis Sci 1979;24:385–96.Google Scholar
Phillips, KG. Hepatotoxicity of erythromycin ethylsuccinate in a child. Can Med Assoc J 1983;129:411–12.Google Scholar
Principi, N, Esposito, S. Comparative tolerability of erythromycin and newer macrolide antibacterials in paediatric patients. Drug Saf 1999;20:2541.Google Scholar
Karthik, SV, Casson, D. Erythromycin-associated cholestatic hepatitis and liver dysfunction in children: the British experience. J Clin Gastroenterol 2005;39:743–4.Google Scholar
Woodhead, JL, Yang, K, Oldach, D, et al. Analyzing the mechanisms behind macrolide antibiotic-induced liver injury using quantitative systems toxicology modeling. Pharm Res 2019;36:48.Google Scholar
Martinez, MA, Vuppalanchi, R, Fontana, RJ, et al. Clinical and histologic features of azithromycin-induced liver injury. Clin Gastroenterol Hepatol 2015;13:369–76 e363.Google Scholar
Giannattasio, A, D’Ambrosi, M, Volpicelli, M, et al. Steroid therapy for a case of severe drug-induced cholestasis. Ann Pharmacother 2006;40:1196–9.Google Scholar
Fox, JC, Szyjkowski, RS, Sanderson, SO, et al. Progressive cholestatic liver disease associated with clarithromycin treatment. J Clin Pharmacol 2002;42:676–80.Google Scholar
Brinker, AD, Wassel, RT, Lyndly, J, et al. Telithromycin-associated hepatotoxicity: clinical spectrum and causality assessment of 42 cases. Hepatology 2009;49:250–7.Google Scholar
Neuberger, J, Nunnerley, HB, Davis, M, et al. Oral-contraceptive-associated liver tumours: occurrence of malignancy and difficulties in diagnosis. Lancet 1980;i:273–6.Google Scholar
Nault, JC, Bioulac-Sage, P, Zucman-Rossi, J. Hepatocellular benign tumors: from molecular classification to personalized clinical care. Gastroenterology 2013;144:888902.Google Scholar
Jeannot, E, Poussin, K, Chiche, L, et al. Association of CYP1B1 germ line mutations with hepatocyte nuclear factor 1alpha-mutated hepatocellular adenoma. Cancer Res 2007;67:2611–16.Google Scholar
Pellock, JM, Faught, E, Leppik, IE, et al. Felbamate: consensus of current clinical experience. Epilepsy Res 2006;71:89101.Google Scholar
Popovic, M, Nierkens, S, Pieters, R, et al. Investigating the role of 2-phenylpropenal in felbamate-induced idiosyncratic drug reactions. Chem Res Toxicol 2004;17:1568–76.Google Scholar
Gaertner, I, Altendorf, K, Batra, A, et al. Relevance of liver enzyme elevations with four different neuroleptics: a retrospective review of 7,263 treatment courses. J Clin Psychopharmacol 2001;21:215–22.Google Scholar
Lo, SK, Wendon, J, Mieli-Vergani, G, et al. Halothane-induced acute liver failure: continuing occurrence and use of liver transplantation. Eur J Gastroenterol Hepatol 1998;10:635–9.Google Scholar
Kenna, JG, Newberger, J, Mieli-Vergani, G, et al. Halothane hepatitis in children. Br Med J 1987;294:1209–11.Google Scholar
Hassall, E, Israel, DM, Gunasekaran, T, et al. Halothane hepatitis in children. J Pediatr Gastroenterol Nutr 1990;11:553–7.Google Scholar
Wark, HJ. Postoperative jaundice in children. The influence of halothane. Anaesthesia 1983;38:237–42.Google Scholar
Warner, LO, Beach, TP, Gariss, JP, et al. Halothane and children: the first quarter century. Anesth Analg 1984;63:838–40.Google Scholar
Psacharopoulos, HJ, Mowat, AP, Davies, M, et al. Fulminant hepatic failure in childhood: an analysis of 31 cases. Arch Dis Child 1980;55:252–8.Google Scholar
Farrell, GC. Mechanism of halothane-induced liver injury: is it immune or metabolic idiosyncrasy? J Gastroenterol Hepatol 1988;3:465–82.Google Scholar
Pohl, LR, Satoh, H, Christ, DD, et al. The immunologic and metabolic basis of drug hypersensitivities. Annu Rev Pharmacol Toxicol 1988;28:367–87.Google Scholar
Kenna, JG, Satoh, H, Christ, DD, et al. Metabolic basis for a drug hypersensitivity: antibodies in sera from patients with halothane hepatitis recognize liver neoantigens that contain the trifluoroacetyl group derived from halothane. J Pharmacol Exp Ther 1988;245:1103–9.Google Scholar
Kenna, JG, Neuberger, J, Williams, R. Evidence for expression in human liver of halothane-induced neoantigens recognized by antibodies in sera from patients with halothane hepatitis. Hepatology 1988;8:1635–41.Google Scholar
Satoh, H, Martin, BM, Schulick, AH, et al. Human anti-endoplasmic reticulum antibodies in sera of patients with halothane-induced hepatitis are directed against a trifluoroacetylated carboxylesterase. Proc Natl Acad Sci (USA) 1989;86:322–6.Google Scholar
Cote, G, Bouchard, S. Hepatotoxicity after desflurane anesthesia in a 15-month-old child with Mobius syndrome after previous exposure to isoflurane. Anesthesiology 2007;107:843–5.Google Scholar
Her, C. Acetaminophen-induced, not desflurane-induced, hepatotoxicity. Anesthesiology 2008;109:570–1.Google Scholar
Hausmann, R, Schmidt, B, Schellmann, B, et al. Differential diagnosis of postoperative liver failure in a 12-year-old child. Int J Legal Med 1996;109:210–12.Google Scholar
Jang, Y, Kim, I. Severe hepatotoxicity after sevoflurane anesthesia in a child with mild renal dysfunction. Paediatr Anaesth 2005;15:1140–4.Google Scholar
Reich, A, Everding, AS, Bulla, M, et al. Hepatitis after sevoflurane exposure in an infant suffering from primary hyperoxaluria type 1. Anesth Analg 2004;99:370–2.Google Scholar
Abdualmjid, RJ, Sergi, C. Hepatotoxic botanicals – an evidence-based systematic review. J Pharm Pharm Sci 2013;16:376404.Google Scholar
Navarro, VJ, Khan, I, Bjornsson, E, et al. Liver injury from herbal and dietary supplements. Hepatology 2017;65:363–73.Google Scholar
Rode, D. Comfrey toxicity revisited. Trends Pharmacol Sci 2002;23:497–9.Google Scholar
Zuckerman, M, Steenkamp, V, Stewart, MJ. Hepatic veno-occlusive disease as a result of a traditional remedy: confirmation of toxic pyrrolizidine alkaloids as the cause, using an in vitro technique. J Clin Pathol 2002;55:676–9.Google Scholar
Weston, CFM, Cooper, BT, Davies, JD, et al. Veno-occlusive disease of the liver secondary to ingestion of comfrey. Br Med J 1987;295:183.Google Scholar
Roulet, M, Laurini, R, Rivier, L, et al. Hepatic veno-occlusive disease in newborn infant of a woman drinking herbal tea. J Pediatr 1988;112:433–6.Google Scholar
Sperl, W, Stuppner, H, Gassner, I, et al. Reversible hepatic veno-occlusive disease in an infant after consumption of pyrrolizidine-containing herbal tea. Eur J Pediatr 1995;154:112–16.Google Scholar
Clouatre, DL. Kava kava: examining new reports of toxicity. Toxicol Lett 2004;150:8596.Google Scholar
Campo, JV, McNabb, J, Perel, JM, et al. Kava-induced fulminant hepatic failure. J Am Acad Child Adolesc Psychiatry 2002;41:631–2.Google Scholar
Anke, J, Ramzan, I. Pharmacokinetic and pharmacodynamic drug interactions with Kava (Piper methysticum Forst. f.). J Ethnopharmacol 2004;93:153–60.Google Scholar
Batchelor, WB, Heathcote, J, Wanless, IR. Chaparral-induced hepatic injury. Am J Gastroenterol 1995;90:831–3.Google Scholar
Sheikh, NM, Philen, RM, Love, LA. Chaparral-associated hepatotoxicity. Arch Intern Med 1997;157:913–19.Google Scholar
Horowitz, RS, Feldhaus, K, Dart, RC, et al. The clinical spectrum of Jin Bu Huan toxicity. Arch Intern Med 1996;156:899903.Google Scholar
Skoulidis, F, Alexander, GJ, Davies, SE. Ma huang associated acute liver failure requiring liver transplantation. Eur J Gastroenterol Hepatol 2005;17:581–4.Google Scholar
Patel, SS, Beer, S, Kearney, DL, et al. Green tea extract: a potential cause of acute liver failure. World J Gastroenterol 2013;19:5174–7.Google Scholar
Laliberte, L, Villeneuve, JP. Hepatitis after the use of germander, a herbal remedy. CMAJ 1996;154:1689–92.Google Scholar
Lekehal, M, Pessayre, D, Lereau, JM, et al. Hepatotoxicity of the herbal medicine germander: metabolic activation of its furano diterpenoids by cytochrome P450 3 A Depletes cytoskeleton-associated protein thiols and forms plasma membrane blebs in rat hepatocytes. Hepatology 1996;24:212–18.Google Scholar
Fau, D, Lekehal, M, Farrell, G, et al. Diterpenoids from germander, an herbal medicine, induce apoptosis in isolated rat hepatocytes. Gastroenterology 1997;113:1334–46.Google Scholar
Lawrenson, JA, Walls, T, Day, AS. Echinacea-induced acute liver failure in a child. J Paediatr Child Health 2014;50:841.Google Scholar
Webb, N, Hardikar, W, Cranswick, NE, et al. Probable herbal medication induced fulminant hepatic failure. J Paediatr Child Health 2005;41:530–1.Google Scholar
Zhu, Y, Li, YG, Wang, JB, et al. Causes, features, and outcomes of drug-induced liver injury in 69 children from China. Gut Liver 2015;9:525–33.Google Scholar
Goldfeld, DA, Verna, EC, Lefkowitch, J, et al. Infliximab-induced autoimmune hepatitis with successful switch to adalimumab in a patient with Crohn’s disease: the index case. Dig Dis Sci 2011;56:3386–8.Google Scholar
Ghabril, M, Bonkovsky, HL, Kum, C, et al. Liver injury from tumor necrosis factor-alpha antagonists: analysis of thirty-four cases. Clin Gastroenterol Hepatol 2013;11:558–64 e553.Google Scholar
Bjornsson, ES, Gunnarsson, BI, Grondal, G, et al. Risk of drug-induced liver injury from tumor necrosis factor antagonists. Clin Gastroenterol Hepatol 2015;13:602–8.Google Scholar
French, JB, Bonacini, M, Ghabril, M, et al. Hepatotoxicity associated with the use of anti-TNF-alpha agents. Drug Saf 2016;39:199208.Google Scholar
Mostamand, S, Schroeder, S, Schenkein, J, et al. Infliximab-associated immunomediated hepatitis in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2016;63:94–7.Google Scholar
Ricciuto, A, Kamath, BM, Walters, TD, et al. New onset autoimmune hepatitis during anti-tumor necrosis factor-alpha treatment in children. J Pediatr 2018;194:128–35 e121.Google Scholar
Fathalla, BM, Goldsmith, DP, Pascasio, JM, et al. Development of autoimmune hepatitis in a child with systemic-onset juvenile idiopathic arthritis during therapy with etanercept. J Clin Rheumatol 2008;14:297–8.Google Scholar
Zimmerman, HJ. Update of hepatotoxicity due to classes of drugs in common clinical use: non-steroidal drugs, anti-inflammatory drugs, antibiotics, antihypertensives, and cardiac and psychotropic drugs. Sem Liver Dis 1990;10:322–38.Google Scholar
Donald, PR. Antituberculosis drug-induced hepatotoxicity in children. Pediatr Rep 2011;3:e16.Google Scholar
Beaudry, P, Brickman, H, Wise, M, et al. Liver enzyme disturbances during isoniazid chemoprophylaxis in children. Am Rev Resp Dis 1974;110:581–4.Google Scholar
Spyridis, P, Sinantios, C, Papadea, I, et al. Isoniazid liver injury during chemoprophylaxis in children. Arch Dis Child 1979;54:65–7.Google Scholar
Palusci, VJ, O’Hare, D, Lawrence, RM. Hepatotoxicity and transaminase measurement during isoniazid chemoprophylaxis in children. Pediatr Infect Dis J 1995;14:144–8.Google Scholar
Wu, SS, Chao, CS, Vargas, JH, et al. Isoniazid-related hepatic failure in children: a survey of liver transplantation centers. Transplantation 2007;84:173–9.Google Scholar
Frydenberg, AR, Graham, SM. Toxicity of first-line drugs for treatment of tuberculosis in children: review. Trop Med Int Health 2009;14:1329–37.Google Scholar
Chang, SH, Nahid, P, Eitzman, SR. Hepatotoxicity in children receiving isoniazid therapy for latent tuberculosis infection. J Pediatric Infect Dis Soc 2014;3:221–7.Google Scholar
Campos-Franco, J, Gonzalez-Quintela, A, Alende-Sixto, MR. Isoniazid-induced hyperacute liver failure in a young patient receiving carbamazepine. Eur J Intern Med 2004;15:396–7.Google Scholar
Indumathi, CK, Sethuraman, A, Jain, S, et al. Revised antituberculosis drug doses and hepatotoxicity in HIV negative children. Indian J Pediatr 2019;86:229–32.Google Scholar
Metushi, IG, Cai, P, Zhu, X, et al. A fresh look at the mechanism of isoniazid-induced hepatotoxicity. Clin Pharmacol Ther 2011;89:911–14.Google Scholar
Wang, P, Pradhan, K, Zhong, XB, et al. Isoniazid metabolism and hepatotoxicity. Acta Pharm Sin B 2016;6:384–92.Google Scholar
Vuilleumier, N, Rossier, MF, Chiappe, A, et al. CYP2E1 genotype and isoniazid-induced hepatotoxicity in patients treated for latent tuberculosis. Eur J Clin Pharmacol 2006;62:423–9.Google Scholar
Kyriakidis, I, Tragiannidis, A, Munchen, S, et al. Clinical hepatotoxicity associated with antifungal agents. Expert Opin Drug Saf 2017;16:149–65.Google Scholar
Lewis, JH, Zimmerman, HJ, Benson, GD, et al. Hepatic injury associated with ketoconazole therapy. Analysis of 33 cases. Gastroenterology 1984;86:503–13.Google Scholar
Egunsola, O, Adefurin, A, Fakis, A, et al. Safety of fluconazole in paediatrics: a systematic review. Eur J Clin Pharmacol 2013;69:1211–21.Google Scholar
Aghai, ZH, Mudduluru, M, Nakhla, TA, et al. Fluconazole prophylaxis in extremely low birth weight infants: association with cholestasis. J Perinatol 2006;26:550–5.Google Scholar
Benjamin, DK Jr., Hudak, ML, Duara, S, et al. Effect of fluconazole prophylaxis on candidiasis and mortality in premature infants: a randomized clinical trial. JAMA 2014;311:1742–9.Google Scholar
Schlienger, RG, Knowles, SR, Shear, NH. Lamotrigine-associated anticonvulsant hypersensitivity syndrome. Neurology 1998;51:1172–5.Google Scholar
Fayad, M, Choueiri, R, Mikati, M. Potential hepatotoxicity of lamotrigine. Pediatr Neurol 2000;22:4952.Google Scholar
Overstreet, K, Costanza, C, Behling, C, et al. Fatal progressive hepatic necrosis associated with lamotrigine treatment: a case report and literature review. Dig Dis Sci 2002;47:1921–5.Google Scholar
Arnon, R, DeVivo, D, Defelice, AR, et al. Acute hepatic failure in a child treated with lamotrigine. Pediatr Neurol 1998;18:251–2.Google Scholar
Bhayana, H, Appasani, S, Thapa, BR, et al. Lamotrigine-induced vanishing bile duct syndrome in a child. J Pediatr Gastroenterol Nutr 2012;55:e147–8.Google Scholar
Zidd, AG, Hack, JB. Pediatric ingestion of lamotrigine. Pediatr Neurol 2004;31:71–2.Google Scholar
Kremer, JM, Lee, RG, Tolman, KG. Liver histology in rheumatoid arthritis patients receiving long-term methotrexate therapy. A prospective study with baseline and sequential biopsy samples. Arthritis & Rheumatism 1989;32:121–7.Google Scholar
Kremer, JM, Furst, DE, Weinblatt, ME, et al. Significant changes in serum AST across hepatic histological grades: prospective analysis of 3 cohorts receiving methotrxate therapy for rheumatoid arthritis. J Rheumatol 1996;23:459–61.Google Scholar
Hashkes, PJ, Balistreri, WF, Bove, KE, et al. The relationship of hepatotoxic risk factors and liver histology in methotrexate therapy for juvenile rheumatoid arthritis. J Pediatr 1999;134:4752.Google Scholar
Graham, LD, Myones, BL, Rivas-Chacon, RF, et al. Morbidity associated with long-term methotrexate therapy in juvenile rheumatoid arthritis. J Pediatr 1992;120:468–73.Google Scholar
Kugathasam, S, Newman, AJ, Dahms, BB, et al. Liver biopsy findings in patients with juvenile rheumatoid arthritis receiving long-term, weekly methotrexate therapy. J Pediatr 1996;128:149–51.Google Scholar
Keim, D, Ragsdale, C, Heidelberger, K, et al. Hepatic fibrosis with the use of methotrexate for juvenile rheumatoid arthritis. J Rheumatol 1990;17:846–8.Google Scholar
Valentino, PL, Church, PC, Shah, PS, et al. Hepatotoxicity caused by methotrexate therapy in children with inflammatory bowel disease: a systematic review and meta-analysis. Inflamm Bowel Dis 2014;20:4759.Google Scholar
Locasciulli, A, Mura, R, Fraschini, D, et al. High-dose methotrexate administration and acute liver damage in children treated for acute lymphoblastic leukemia. A prospective study. Haematologica 1992;77:4953.Google Scholar
Fisher, MC, Cronstein, BN. Metaanalysis of methylenetetrahydrofolate reductase (MTHFR) polymorphisms affecting methotrexate toxicity. J Rheumatol 2009;36:539–45.Google Scholar
Malcolm, A, Heap, TR, Eckstein, RP, et al. Minocycline-induced liver injury. Am J Gastroenterol 1996;91:1641–3.Google Scholar
Gough, A, Chapman, S, Wagstaff, K, et al. Minocycline induced autoimmune hepatitis and systemic lupus erythematosus-like syndrome. BMJ 1996;312:169–72.Google Scholar
Bhat, G, Jordan, J Jr., Sokalski, S, et al. Minocycline-induced hepatitis with autoimmune features and neutropenia. J Clin Gastroenterol 1998;27:74–5.Google Scholar
Vo, HD, Jimenez-Rivera, C, Critch, J, et al. Distinctive features of minocycline-induced autoimmune hepatitis in children. Gastroenterology 2015;148:S996.Google Scholar
Urban, TJ, Nicoletti, P, Chalasani, N, et al. Minocycline hepatotoxicity: clinical characterization and identification of HLA-B*35:02 as a risk factor. J Hepatol 2017;67:137–44.Google Scholar
Davies, MG, Kersey, PJW. Acute hepatitis and exfoliative dermatitis associated with minocycline. BMJ 1989;298:1523–4.Google Scholar
Boudreaux, JP, Hayes, DH, Mizrahi, S, et al. Fulminant hepatic failure, hepatorenal syndrome, and necrotizing pancreatitis after minocycline hepatotoxicity. Transplant Proc 1993;25:1873.Google Scholar
Iveli, P, Noguera-Julian, A, Soler-Palacin, P, et al. [Hepatotoxicity in healthy infants exposed to nevirapine during pregnancy. Enferm Infecc Microbiol Clin 2016;34:3944.Google Scholar
McKoy, JM, Bennett, CL, Scheetz, MH, et al. Hepatotoxicity associated with long- versus short-course HIV-prophylactic nevirapine use: a systematic review and meta-analysis from the Research on Adverse Drug events And Reports (RADAR) project. Drug Saf 2009;32:147–58.Google Scholar
Ciccacci, C, Borgiani, P, Ceffa, S, et al. Nevirapine-induced hepatotoxicity and pharmacogenetics: a retrospective study in a population from Mozambique. Pharmacogenomics 2010;11:2331.Google Scholar
Ciccacci, C, Di Fusco, D, Marazzi, MC, et al. Association between CYP2B6 polymorphisms and nevirapine-induced SJS/TEN: a pharmacogenetics study. Eur J Clin Pharmacol 2013;69:1909–16.Google Scholar
Singh, H, Lata, S, Dhole, TN, et al. Occurrence of CYP2B6 516G&gt;T polymorphism in patients with ARV-associated hepatotoxicity. Mol Genet Genomic Med 2019;7:e00598.Google Scholar
Agundez, JA, Lucena, MI, Martinez, C, et al. Assessment of nonsteroidal anti-inflammatory drug-induced hepatotoxicity. Expert Opin Drug Metab Toxicol 2011;7:817–28.Google Scholar
Titchen, T, Cranswick, N, Beggs, S. Adverse drug reactions to nonsteroidal anti-inflammatory drugs, COX-2 inhibitors and paracetamol in a paediatric hospital. Br J Clin Pharmacol 2005;59:718–23.Google Scholar
Whittaker, SJ, Amar, JN, Wanless, IR, et al. Sulindac hepatotoxicity. Gut 1982;23:875–7.Google Scholar
Boelsterli, UA. Diclofenac-induced liver injury: a paradigm of idiosyncratic drug toxicity. Toxicol Appl Pharmacol 2003;192:307–22.Google Scholar
Merlani, G, Fox, M, Oehen, HP, et al. Fatal hepatoxicity secondary to nimesulide. Eur J Clin Pharmacol 2001;57:321–6.Google Scholar
Bessone, F, Colombato, L, Fassio, E, et al. The spectrum of nimesulide-induced-hepatotoxicity. An overview. Antiinflamm Antiallergy Agents Med Chem 2010;9:355–65.Google Scholar
Marotta, PJ, Roberts, EA. Pemoline hepatotoxicity in children. J Pediatr 1998;132:894–7.Google Scholar
Rosh, JR, Dellert, SF, Narkewicz, M, et al. Four cases of severe hepatotoxicity associated with pemoline: possible autoimmune pathogenesis. Pediatrics 1998;101:921–3.Google Scholar
Gresser, U. Amoxicillin-clavulanic acid therapy may be associated with severe side effects – review of the literature. Eur J Med Res 2001;6:139–49.Google Scholar
deLemos, AS, Ghabril, M, Rockey, DC, et al. Amoxicillin-clavulanate-induced liver injury. Dig Dis Sci 2016;61:2406–16.Google Scholar
Stricker, BH, Van den Broek, JW, Keuning, J, et al. Cholestatic hepatitis due to antibacterial combination of amoxicillin and clavulanic acid (augmentin). Dig Dis Sci 1989;34:1576–80.Google Scholar
Chawla, A, Kahn, E, Yunis, EJ, et al. Rapidly progressive cholstasis: an unusual reaction to amoxicillin/clavulinic acid in a child. J Pediatr 2000;136:121–3.Google Scholar
Yu, MK, Yu, MC, Lee, F. Association of DRESS syndrome with chylous ascites. Nephrol Dial Transplant 2006;21:3301–3.Google Scholar
Kumar, A, Sood, V, Khanna, R, et al. Clinical spectrum and outcome of pediatric drug induced liver injury. Indian J Pediatr 2018;85:676–8.Google Scholar
Fontana, RJ, Shakil, AO, Greenson, JK, et al. Acute liver failure due to amoxicillin and amoxicillin/clavulanate. Dig Dis Sci 2005;50:1785–90.Google Scholar
Daly, AK, Donaldson, PT, Bhatnagar, P, et al. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat Genet 2009;41:816–19.Google Scholar
Lucena, MI, Molokhia, M, Shen, Y, et al. Susceptibility to amoxicillin-clavulanate-induced liver injury is influenced by multiple HLA class I and II alleles. Gastroenterology 2011;141:338–47.Google Scholar
Roberts, EA, Spielberg, SP, Goldbach, M, et al. Phenobarbital hepatotoxicity in an 8-month-old infant. J Hepatol 1990;10:235–9.Google Scholar
Pinna, AP, Locci, G, Furno, M, et al. DILI (drug induced liver injury) in a 9-month-old infant: a rare case of phenobarbital-induced hepatotoxicity. J Pediatr Neonat Individ Med 2013;2:93–5.Google Scholar
Li, AM, Nelson, EA, Hon, EK, et al. Hepatic failure in a child with anti-epileptic hypersensitivity syndrome. J Paediatr Child Health 2005;41:218–20.Google Scholar
Bessmertny, O, Hatton, RC, Gonzalez-Peralta, RP. Antiepileptic hypersensitivity syndrome in children. Ann Pharmacother 2001;35:533–8.Google Scholar
Mullick, FG, Ishak, KG. Hepatic injury associated with diphenylhydantoin therapy. Am J Clin Pathol 1980;74:442–52.Google Scholar
Spielberg, SP, Gordon, GB, Blake, DA, et al. Anticonvulsant toxicity in vitro: possible role of arene oxides. J Pharmacol Exp Ther 1981;217:386–9.Google Scholar
Spielberg, SP, Gordon, GB, Blake, DA, et al. Predisposition to phenytoin hepatotoxicity assessed in vitro. N Engl J Med 1981;305:722–7.Google Scholar
Akmal, A, Kung, J. Propylthiouracil, methimazole, and carbimazole-related hepatotoxicity. Expert Opin Drug Saf 2014;13:13971406.Google Scholar
Rivkees, SA, Mattison, DR. Propylthiouracil (PTU) hepatotoxicity in children and recommendations for discontinuation of use. Int J Pediatr Endocrinol 2009;2009:132041.Google Scholar
Rivkees, SA, Szarfman, A. Dissimilar hepatotoxicity profiles of propylthiouracil and methimazole in children. J Clin Endocrinol Metab 2010;95:3260–7.Google Scholar
Maggiore, G, Larizza, D, Lorini, R, et al. PTU hepatotoxicity mimicking autoimmune chronic active hepatitis in a girl. J Pediatr Gastroenterol Nutr 1989;8:547–8.Google Scholar
Hayashida, CY, Duarte, AJ, Sato, AE, et al. Neonatal hepatitis and lymphocyte sensitization by placental transfer of propylthiouracil. J Endocrinol Invest 1990;13:937–41.Google Scholar
Loomba-Albrecht, LA, Bremer, AA, Wong, A, et al. Neonatal cholestasis due to hyperthyroidism. J Pediatr Gastroenterol Nutr 2012;54:433–4.Google Scholar
Zane, LT, Leyden, WA, Marqueling, AL, et al. A population-based analysis of laboratory abnormalities during isotretinoin therapy for acne vulgaris. Arch Dermatol 2006;142:1016–22.Google Scholar
Guzman Rojas, P, Gallegos Lopez, R, Ciliotta Chehade, A, et al. Autoimmune hepatitis induced by isotretionine. Rev Gastroenterol Peru 2016;36:86–9.Google Scholar
Fallon, MB, Boyer, JL. Hepatic toxicity of vitamin A and synthetic retinoids. J Gastroenterol Hepatol 1990;5:334–42.Google Scholar
Kumra, S, Herion, D, Jacobsen, LK, et al. Case study: risperidone-induced hepatotoxicity in pediatric patients. J Am Acad Child Adolesc Psychiatry 1997;36:701–5.Google Scholar
Krebs, S, Dormann, H, Muth-Selbach, U, et al. Risperidone-induced cholestatic hepatitis. Eur J Gastroenterol Hepatol 2001;13:67–9.Google Scholar
Copur, M, Erdogan, A. Risperidone rechallenge for marked liver function test abnormalities in an autistic child. Recent Pat Endocr Metab Immune Drug Discov 2011;5:237–9.Google Scholar
Bjornsson, ES. Hepatotoxicity of statins and other lipid-lowering agents. Liver Int 2017;37:173–8.Google Scholar
Desai, NK, Mendelson, MM, Baker, A, et al. Hepatotoxicity of statins as determined by serum alanine aminotransferase in a pediatric cohort with dyslipidemia. J Pediatr Gastroenterol Nutr 2019;68:175–81.Google Scholar
Olah, AV, Szabo, GP, Varga, J, et al. Relation between biomarkers and clinical severity in patients with Smith-Lemli-Opitz syndrome. Eur J Pediatr 2013;172:623–30.Google Scholar
Shear, NH, Spielberg, SP, Grant, DM, et al. Differences in metabolism of sulfonamides predisposing to idiosyncratic toxicity. Ann Intern Med 1986;105:179–84.Google Scholar
Besnard, M, Debray, D, Durand, P, et al. Fulminant hepatitis in two children treated with sulfasalazine for Crohn disease. Arch Pediatr 1999;6:643–6.Google Scholar
Karpman, E, Kurzrock, EA. Adverse reactions of nitrofurantoin, trimethoprim and sulfamethoxazole in children. J Urol 2004;172:448–53.Google Scholar
Bucaretchi, F, Vicente, DC, Pereira, RM, et al. Dapsone hypersensitivity syndrome in an adolescent during treatment of leprosy. Rev Inst Med Trop Sao Paulo 2004;46:331–4.Google Scholar
Rieder, MJ, Uetrecht, J, Shear, NH, et al. Diagnosis of sulfonamide hypersensitivity reactions by in-vitro “rechallenge” with hydroxylamine metabolites. Ann Intern Med 1989;110:286–9.Google Scholar
Cribb, AE, Spielberg, SP. Hepatic microsomal metabolism of sulfamethoxazole to the hydroxylamine. Drug Metab Dispos 1990;18:784–7.Google Scholar
Sanderson, JP, Naisbitt, DJ, Park, BK. Role of bioactivation in drug-induced hypersensitivity reactions. AAPS J 2006;8:E5564.Google Scholar
Ogese, MO, Faulkner, L, Jenkins, RE, et al. Characterization of drug-specific signaling between primary human hepatocytes and immune cells. Toxicol Sci 2017;158:7689.Google Scholar
Ogese, MO, Jenkins, RE, Adair, K, et al. Exosomal transport of hepatocyte-derived drug-modified proteins to the immune system. Hepatology 2019;70(5):1732–49.Google Scholar
Sztajnkrycer, MD. Valproic acid toxicity: overview and management. J Toxiocl Clin Toxicol 2002;40:789801.Google Scholar
Suchy, FJ, Balistreri, WF, Buchino, J, et al. Acute hepatic failure associated with the use of sodium valproate. Report of two fatal cases. N Engl J Med 1979;300:962–6.Google Scholar
Zimmerman, HJ, Ishak, KG. Valproate-induced hepatic injury: analysis of 23 fatal cases. Hepatology 1982;2:591–7.Google Scholar
Koenig, SA, Siemes, H, Blaker, F, et al. Severe hepatotoxicity during valproate therapy: an update and report of eight new fatalities. Epilepsia 1994;35:1005–15.Google Scholar
Star, K, Edwards, IR, Choonara, I. Valproic acid and fatalities in children: a review of individual case safety reports in VigiBase. PLoS One 2014;9:e108970.Google Scholar
Price, KE, Pearce, RE, Garg, UC, et al. Effects of valproic acid on organic acid metabolism in children: a metabolic profiling study. Clin Pharmacol Ther 2011;89:867–74.Google Scholar
McCall, M, Bourgeois, JA. Valproic acid-induced hyperammonemia: a case report. J Clin Psychopharmacol 2004;24:521–6.Google Scholar
Gerstner, T, Buesing, D, Longin, E, et al. Valproic acid induced encephalopathy – 19 new cases in Germany from 1994 to 2003 – A side effect associated to VPA-therapy not only in young children. Seizure 2006;15:443–8.Google Scholar
Ghodke-Puranik, Y, Thorn, CF, Lamba, JK, et al. Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenet Genomics 2013;23:236–41.Google Scholar
Li, X, Norwood, DL, Mao, L-F, et al. Mitochondrial metabolism of valproic acid. Biochemistry 1991;30:388–94.Google Scholar
Kiang, TK, Ho, PC, Anari, MR, et al. Contribution of CYP2C9, CYP2A6, and CYP2B6 to valproic acid metabolism in hepatic microsomes from individuals with the CYP2C9*1/*1 genotype. Toxicol Sci 2006;94:261–71.Google Scholar
Gopaul, S, Farrell, K, Abbott, F. Effects of age and polytherapy, risk factors of valproic acid (VPA) hepatotoxicity, on the excretion of thiol conjugates of (E)-2,4-diene VPA in people with epilepsy taking VPA. Epilepsia 2003;44:322–8.Google Scholar
Eadie, MJ, McKinnon, GE, Dunstan, PR, et al. Valproate metabolism during hepatotoxicity associated with the drug. Quart J Med 1990;77:1229–40.Google Scholar
Kossak, BD, Schmidt-Sommerfeld, E, Schoeller, DA, et al. Impaired fatty acid oxidation in children on valproic acid and the effect of L-carnitine. Neurology 1993;43:2362–8.Google Scholar
Tong, V, Teng, XW, Chang, TK, et al. Valproic acid I: time course of lipid peroxidation biomarkers, liver toxicity, and valproic acid metabolite levels in rats. Toxicol Sci 2005;86:427–35.Google Scholar
Tong, V, Teng, XW, Chang, TK, et al. Valproic acid II: effects on oxidative stress, mitochondrial membrane potential, and cytotoxicity in glutathione-depleted rat hepatocytes. Toxicol Sci 2005;86:436–43.Google Scholar
McCarver, DG, Hines, RN. The ontogeny of human drug-metabolizing enzymes: phase II conjugation enzymes and regulatory mechanisms. J Pharmacol Exp Ther 2002;300:361–6.Google Scholar
Xing, Y, Yang, L, Wang, L, et al. Systematic screening for polymorphisms within the UGT1A6 gene in three Chinese populations and function prediction through structural modeling. Pharmacogenomics 2009;10:741–52.Google Scholar
Konig, SA, Schenk, M, Sick, C, et al. Fatal liver failure associated with valproate therapy in a patient with Friedreich’s disease: review of valproate hepatotoxicity in adults. Epilepsia 1999;40:1036–40.Google Scholar
Schwabe, MJ, Dobyns, WB, Burke, B, et al. Valproate-induced liver failure in one of two siblings with Alpers disease. Pediatr Neurol 1997;16:337–43.Google Scholar
Kayihan, N, Nennesmo, I, Ericzon, BG, et al. Fatal deterioration of neurological disease after orthotopic liver transplantation for valproic acid-induced liver damage. Pediatr Transplant 2000;4:211–14.Google Scholar
Vantroys, E, Smet, J, Vanlander, AV, et al. Severe hepatopathy and neurological deterioration after start of valproate treatment in a 6-year-old child with mitochondrial tryptophanyl-tRNA synthetase deficiency. Orphanet J Rare Dis 2018;13:80.Google Scholar
Stewart, JD, Horvath, R, Baruffini, E, et al. Polymerase gamma gene POLG determines the risk of sodium valproate-induced liver toxicity. Hepatology 2010;52:1791–6.Google Scholar
Li, S, Guo, J, Ying, Z, et al. Valproic acid-induced hepatotoxicity in Alpers syndrome is associated with mitochondrial permeability transition pore opening-dependent apoptotic sensitivity in an induced pluripotent stem cell model. Hepatology 2015;61:1730–9.Google Scholar
Saneto, RP, Lee, IC, Koenig, MK, et al. POLG DNA testing as an emerging standard of care before instituting valproic acid therapy for pediatric seizure disorders. Seizure 2010;19:140–6.Google Scholar
Beghi, E, Bizzi, A, Codegoni, AM, et al. Valproate, carnitine metabolism, and biochemical indicators of liver function. Epilepsia 1990;31:346–52.Google Scholar
Lheureux, PE, Hantson, P. Carnitine in the treatment of valproic acid-induced toxicity. Clin Toxicol (Phila) 2009;47:101–11.Google Scholar
Bohan, TP, Helton, E, McDonald, I, et al. Effect of L-carnitine treatment for valproate-induced hepatotoxicity. Neurology 2001;56:1405–9.Google Scholar
Squires, RH, Dhawan, A, Alonso, E, et al. Intravenous N-acetylcysteine in pediatric patients with nonacetaminophen acute liver failure: a placebo-controlled clinical trial. Hepatology 2013;57:1542–9.Google Scholar
Suzuki, A, Brunt, EM, Kleiner, DE, et al. The use of liver biopsy evaluation in discrimination of idiopathic autoimmune hepatitis versus drug-induced liver injury. Hepatology 2011;54:931–9.Google Scholar
Naranjo, CA, Busto, U, Sellers, EM, et al. A method for estimating the probability of adverse drug reactions. Clin Pharmacol Ther 1981;30:239–45.Google Scholar
Garcia-Cortes, M, Stephens, C, Lucena, MI, et al. Causality assessment methods in drug induced liver injury: strengths and weaknesses. J Hepatol 2011;55:683–91.Google Scholar
Gallagher, RM, Kirkham, JJ, Mason, JR, et al. Development and inter-rater reliability of the Liverpool adverse drug reaction causality assessment tool. PLoS One 2011;6:e28096.Google Scholar

References

Mackay, IR. Hepatoimmunology: a perspective. Immunol Cell Biol 2002;80:3644.Google Scholar
Crispe, IN, Dao, T, Klugewitz, K, et al. The liver as a site of T-cell apoptosis: graveyard, or killing field? Immunol Rev 2000;174:4762.Google Scholar
Weiler-Normann, C, Rehermann, B. The liver as an immunological organ. J Gastroenterol Hepatol 2004;19:S279S283.Google Scholar
Rolando, N, Wade, J, Davalos, M, et al. The systemic inflammatory response syndrome in acute liver failure. Hepatology 2000;32:734–9.Google Scholar
Tilg, H, Wilmer, A, Vogel, W, et al. Serum levels of cytokines in chronic liver diseases. Gastroenterology 1992;103:264–74.Google Scholar
Izumi, S, Hughes, RD, Langley, PG, et al. Extent of the acute phase response in fulminant hepatic failure. Gut 1994;35:982–6.Google Scholar
Notarangelo, L. Primary immunodeficiencies. J Allergy Clin Immunol 2010;125:S182S194.Google Scholar
Bousfiha, A, Jeddane, L, Picard, C, et al. The 2017 IUIS Phenotypic Classification for Primary Immunodeficiencies. J Clin Immunol 2018;38:129–43.Google Scholar
Hammer, SM. Management of newly diagnosed HIV infection. N Engl J Med 2005;353:1702–10.Google Scholar
Davies, EG, Thrasher, AJ. Update on the hyper immunoglobulin M syndromes. Br J Haematol 2010;149:167–80.Google Scholar
Rodrigues, F, Davies, ED, Harrison, P, et al. Liver disease in primary immunodeficiencies. J Pediatr 2004;145:333–9.Google Scholar
Record, CO, Shilkin, KB, Eddleston, ALWF, Williams, R. Intrahepatic sclerosing cholangitis associated with a familial immunodeficiency syndrome. Lancet 1973;ii:1820.Google Scholar
Davis, JJ, Heyman, MB, Ferrell, L, et al. Sclerosing cholangitis associated with chronic cryptosporidiosis in a child with a congenital immunodeficiency disorder. Am J Gastroenterol 1987;82:1196–202.Google Scholar
Hayward, AR, Levy, J, Facchetti, F, et al. Cholangiopathy and tumours of the pancreas, liver and biliary tree in boys with X-linked immunodeficiency with hyper-IgM. J Immunol 1997;158:977–83.Google Scholar
McLauchlin, J, Amar, CFL, Pedraza-Diaz, S, et al. Polymerase chain reaction-based diagnosis of infection with Cryptosporidium in children with primary immunodeficiencies. Pediatr Inf Dis J 2003;22:329–34.Google Scholar
Stephens, J, Cosyns, M, Jones, M, Hayward, A. Liver and bile duct pathology following Cryptosporidium parvum infection of the immunodeficient mice. Hepatology 1999;30:2735.Google Scholar
Chen, XM, Levine, SA, Tietz, P, et al. Cryptosporidium parvum is cytopathic for cultured human biliary epithelia via an apoptotic mechanism. Hepatology 1998;28:906–13.Google Scholar
Cello, JP. Acquired immunodeficiency syndrome cholangiopathy: spectrum of disease. Am J Med 1989;86:539–46.Google Scholar
Gerber, DA, Green, M, Jaffe, R, Greenberg, D, et al. Cryptosporidial infections after solid organ transplantation in children. Pediatr Transplant 2000;4:50–5.Google Scholar
Hadzic, N, Pagliuca, A, Rela, M, et al. Correction of the hyper IgM-syndrome after liver and bone marrow transplantation.N Engl J Med 2000;342:320–4.Google Scholar
Winkelstein, JA, Marino, MC, Ochs, H, et al. The X-linked hyper-IgM syndrome. Clinical and immunological features of 79 patients. Medicine 2003;82:373–84.Google Scholar
Jain, A, Kovacs, JA, Nelson, DL, et al. Partial immune reconstitution of X-linked hyper IgM syndrome with recombinant CD40 ligand. Blood 2011;118:3811–17.Google Scholar
Martinez Ibanez, V, Espanol, T, Matamoros, N, et al. Relapse of sclerosing cholangitis after liver transplantation in patients with hyper-IgM syndrome. Transplant Proc 1997;29:432–3.Google Scholar
Thomas, C, De Saint, BG, Le Deist, F, et al. Brief report: correction of X-linked hyper-IgM syndrome by allogeneic bone marrow transplantation. N Engl J Med 1995;333:426–9.Google Scholar
Khawaja, K, Gennery, AR, Flood, TJ, et al. Bone marrow transplantation for CD40 ligand deficiency: a single centre experience. Arch Dis Child 2001;84:508–11.Google Scholar
Amrolia, P, Gaspar, HB, Hassan, A, et al. Nonmyeloablative stem cell transplantation for congenital immunodeficiencies. Blood 2000;96:1239–46.Google Scholar
Jacobsohn, DA, Emerick, KM, Scholl, P, et al. Nonmyeloablative hematopoietic stem cell transplant for X-linked hyper-immunoglobulin M syndrome with cholangiopathy. Pediatrics 2004;113:122–7.Google Scholar
Kahn, K, Sharp, H, Hunter, D, Kerzner, B, et al. Primary sclerosing cholangitis in Wiskott–Aldrich syndrome. J Pediatr Gastroenterol Nutr 2001;32:95–9.Google Scholar
Blanshard, C, Shanson, DC, Gazzard, BG. Pilot studies of azithromycin, letrazuril and paromomycin in the treatment of Cryptosporidiosis. Int J STD AIDS 1997;8:124–9.Google Scholar
Nachbaur, D, Kropshofer, G, Feichtinger, H, et al. Cryptosporidiosis after CD34- selected autologous peripheral blood stem cell transplantation (PBSCT). Treatment with paromomycin, azithromycin and recombinant human interleukin-2. Bone Marrow Transplant 1997;19:1261–3.Google Scholar
Rosignol, JF, Ayoub, A, Ayers, MS. Treatment of diarrhea caused by Cryptoporidium parvum: a prospective randomized, double-blind, placebo-controlled study of nitazoxanide. J Infect Dis 2001;184:103–6.Google Scholar
Gilger, MA, Gann, ME, Opekun, AR, Gleason, WA Jr. Efficacy of ursodeoxycholic acid in the treatment of primary sclerosing cholangitis in children. J Pediatr Gastroenterol Nutr 2000;31:136–41.Google Scholar
Bollinger, ME, Arredongo-Vega, FX, Santisteban, I, et al. Brief report: hepatic dysfunction as a complication of adenosine deaminase deficiency. N Engl J Med 1996;334:1367–71.Google Scholar
Bader-Meunier, B, Parez, N, Muller, S. Treatment of hemophagocytic lymphohistiocytosis with cyclosporin A and steroids in a boy with lysinuric protein intolerance. J Pediatr 2000;136:134.Google Scholar
Raby, RB, Ward, RB, Herrod, HG. Propionic academia and immunodeficiency. J Inherit Metab Dis 1994;17:250–1.Google Scholar
Moreno, LA, Gottrand, F, Turck, D, et al. Severe combined immunodeficiency syndrome associated with autosomal recessive familial multiple gastrointestinal atresias: study of a family. Am J Med Genet 1990;37:143–6.Google Scholar
Gilroy, RK, Coccia, PF, Talmadge, JE, et al. Donor immune reconstitution after liver-small bowel transplantation for multiple intestinal atresia with immunodeficiency. Blood 2004;103:1171–4.Google Scholar
Malamut, G, Ziol, M, Suarez, F, et al. Nodular regenerative hyperplasia: the main liver disease in patients with primary hypogammaglobulinemia and hepatic abnormalities. J Hepatol 2008;48:7482.Google Scholar
Fiore, M, Ammendola, R, Gaetaniello, L, et al. Chronic unexplained liver disease in children with primary immunodeficiency syndromes. J Clin Gastroenterol 1998;26:187–92.Google Scholar
Selik, RM, Lindegren, ML. Changes in deaths reported with human immunodeficiency virus infection among United States children less than thirteen years old, 1987 through 1999. Pediatr Infect Dis J 2003;22:635641.Google Scholar
Fontana, M, Boldorini, R, Zuin, G, et al. Ultrastructural changes in duodenal mucosa of HIV-infected children J Pediatr Gastroenterol Nutr 1993;17:255–9.Google Scholar
Duffy, LF, Daum, F, Kahn, E, et al. Hepatitis in children with acquired immune deficiency syndrome: histopathologic and immunocytologic features. Gastroenterology 1986;90:173–81.Google Scholar
Morotti, RA, Tata, M, Drut, R, et al. Liver pathology in children with AIDS: a comparison between the South American and North American population. Pediatr Pathol Mol Med 2001;20:537–45.Google Scholar
Lacaille, F, Fournet, JC, Blanche, S. Clinical utility of liver biopsy in children with acquired immunodeficiency syndrome. Pediatr Infect Dis J 1999;18:143–7.Google Scholar
Bouche, H, Housset, C, Dumont, JL, et al. AIDS-related cholangitis: diagnostic features and course in 15 patients.J Hepatol 1993;17:34–9.Google Scholar
Chung, CJ, Sivit, CJ, Rakusan, TA, et al. Hepatobiliary abnormalities on sonography in children with HIV infection. J Ultrasound Med 1994;13:205–10.Google Scholar
Clark, SJ, Creighton, S, Portmann, B, et al. Acute liver failure associated with antiretroviral treatment for HIV: a report of six cases. J Hepatol 2002;36:295301.Google Scholar
Church, JA, Mitchell, WG, Gonzales-Gomes, I, et al. Mitochondrial DNA depletion, near-fatal metabolic acidosis, and liver failure in an HIV-infected child treated with combination antiretroviral therapy. J Pediatr 2001;138:748–51.Google Scholar
Neff, GW, Bonham, A, Tzakis, AG, et al. Orthotopic liver transplantation in patients with human immunodeficiency virus and end-stage liver disease. Liver Transplant 2003;9:239–47.Google Scholar
Martin, P, DiBisceglie, AM, Kassianides, C, et al. Rapidly progressive non-A non-B hepatitis in patients with human immunodeficiency virus infection. Gastroenterology 1989;97:1559–61.Google Scholar
Housset, C, Pol, S, Carnot, F, et al. Interactions between human immunodeficiency virus-1, hepatitis delta virus and hepatitis B virus infections in 260 chronic carriers of hepatitis B virus. Hepatology 1992;15:578–83.Google Scholar
Resti, M, Azzari, C, Bortolotti, F. Hepatitis C virus infection in children co-infected with HIV: epidemiology and management. Pediatr Drugs 2002;4:571–80.Google Scholar
Puoti, M, Torti, C, Bruno, R, et al. Natural history of chronic hepatitis B in co-infected patients. J Hepatol 2006;44:S65S70.Google Scholar
Benhamou, Y. Treatment algorithm for chronic hepatitis B in HIV-infected patients. J Hepatol 2006;44:S90S94.Google Scholar
Lau, G, Suri, D, Liang, R, et al. Resolution of chronic hepatitis B and anti-HBs seroconversion in humans by adoptive transfer of immunity to hepatitis B core antigen. Gastroenterology 2002;122:614–24.Google Scholar
Quinti, I, Pandolfi, F, Paganelli, R, et al. HCV infection in patients with primary defects of immunoglobulin production. Clin Exp Immunol 1995;102:1116.Google Scholar
Bjoro, K, Froland, SS, Yun, Z, et al. Hepatitis C infection in patients with primary hypogammaglobulinemia after treatment with contaminated immune globulin. N Engl J Med 1994;331:1607–11.Google Scholar
Smith, MSH, Webster, DB, Dhillon, AP, et al. Orthotopic liver transplantation for chronic hepatitis in two patients with common variable immunodeficiency. Gastroenterology 1995;108:879–84.Google Scholar
Gow, PJ, Mutimer, D. Successful outcome of liver transplantation in a patient with hepatitis C and common variable immune deficiency. Transplant Int 2002;15:380383.Google Scholar
Filipovich, AH. Hemophagocytic lymphohistiocytosis and other hemophagocytic disorders. Immunol Allergy Clin N Am 2008;28:293313.Google Scholar
Freeman, HR, Ramanan, AV. Review of haemophagocytic lymphohistiocytosis. Arch Dis Child 2011;96:688–93.Google Scholar
Hirst, WJ, Layton, DM, Singh, S, et al. Haemophagocytic lymphohistiocytosis: experience at two U.K. centres. BrJ Haematol 1994;88:731–9.Google Scholar
Henter, JI, Horne, A, Arico, M, et al. HLH-2004: diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer 2007;48:124–31.Google Scholar
Farquhar, JW, Claireaux, AE. Familial haemophagocytic reticulosis. Arch Dis Child 1952;27:519–25.Google Scholar
Arico, M, Danesino, C, Pende, D, Moretta, L. Pathogenesis of haemophagocytic lymphohistiocytosis. Br J Haematol 2001;114:761–9.Google Scholar
Chisuwa, H, Hashikura, Y, Nakazawa, Y, et al. Fatal hemophagocytic syndrome after living-related liver transplantation: a report of two cases. Transplantation 2001;72:1843–6.Google Scholar
Ohadi, M, Lalloz, MR, Sham, P, et al. Localization of a gene for familial hemophagocytic lymphohistiocytosis at chromosome 9q21.3–22 by homozygosity mapping. Am J Hum Genet 1999;64:165–71.Google Scholar
Dufourcq-Lagelouse, R, Jabado, N, Le Deist, F, et al. Linkage of familial hemophagocytic lymphohistiocytosis to 10q21–22 and evidence for heterogeneity. Am J Hum Genet 1999;64:172–9.Google Scholar
Feldmann, J, Callebaut, I, Raposo, G, et al. Munc13-4 is essential for cytolytic granules fusion and is mutated in a form of familial hemophagocytic lymphohistiocytosis (FHL3). Cell 2003;115:461–73.Google Scholar
zur Stadt, U, Schmidt, S, Kasper, B, et al. Linkage of familial hemophagocytic lymphohistiocytosis (FHL) type-4 to chromosome 6q24 and identification of mutations in syntaxin 11. Hum Mol Gen 2005;14:827–34.Google Scholar
Côte, M, Ménager, MM, Burgess, A, et al. Munc18-2 deficiency causes familial hemophagocytic lymphohistiocytosis type 5 and impairs cytotoxic granule exocytosis in patient NK cells. J Clin Invest 2009;119:3765–73.Google Scholar
Kogawa, K, Lee, SM, Villanueva, J, et al. Perforin expression in cytotoxic lymphocytes from patients with hemophagocytic lymphohistiocytosis and their family members. Blood 2002;99:61–6.Google Scholar
Clementi, R, Locatelli, F, Dupre, L, et al. A proportion of patients with lymphoma may harbor mutations of the perforin gene. Blood 2005;105:4424–8.Google Scholar
Stephan, JL, Donadieu, J, Ledeist, F, et al. Treatment of familial hemophagocytic lymphohistiocytosis with antithymocyte globulins, steroids, and cyclosporin A. Blood 1993;82:2319–23.Google Scholar
Fischer, A, Cerf-Bensussan, N, Blanche, S, et al. Allogeneic bone marrow transplantation for erythrophagocytic lymphohistiocytosis. J Pediatr 1986;108:267–70.Google Scholar
Durken, M, Horstmann, M, Bieling, P, et al. Improved outcome in haemophagocytic lymphohistiocytosis after bone marrow transplantation from related and unrelated donors: a single-centre experience of 12 patients. Br J Haematol 1999;106:1052–8.Google Scholar
Cooper, N, Rao, K, Gilmour, K, et al. Stem cell transplantation with reduced intensity conditioning for haemophagocytic lymphohistiocytosis. Blood 2006;107:1233–6.Google Scholar
Matthes-Martin, S, Peters, C, Koningsrainer, A, et al. Successful stem cell transplantation following liver transplantation from the same haploidentical family donor in a girl with hemophagocytic lymphohistiocytosis. Blood 2000;96:3997–9.Google Scholar
Purtilo, DT, Cassel, CK, Yang, JPS, et al. X-linked recessive progressive combined variable immunodeficiency (Duncan’s disease). Lancet 1975;i:935–41.Google Scholar
Howie, D, Sayos, J, Terhorst, C, Morra, M. The gene defective in X-linked lymphoproliferative disease controls T cell dependent immune surveillance against Epstein–Barr virus. Curr Opin Immunol 2000;12:474–8.Google Scholar
Nichols, KE, Ma, CS, Cannons, JL, et al. Molecular and cellular pathogenesis of X-linked lymphoproliferative disease. Immunol Rev 2005;203:180–99.Google Scholar
Sayos, J, Wu, C, Morra, M, et al. The X linked lymphoproliferative: disease gene product SAP regulates signals induced through the co-receptor SLAM. Nature 1998;395:462–9.Google Scholar
Skare, JC, Sullivan, JL, Milunsky, A. Mapping the mutation causing the X- linked lymphoproliferative syndrome in relation to restriction fragment length polymorphisms on Xq. Hum Genet 1989;82:349–53.Google Scholar
Rigaud, S, Fondaneche, MC, Lambert, N, et al. XIAP deficiency in humans causes an X-linked lymphoproliferative syndrome. Nature 2006;444:110–14.Google Scholar
Paclopnik Schmid, J, Canioni, D, Moshous, D, et al. Clinical similarities and differences of patients with X-linked lymphoproliferative syndrome type 1 (XLP-1/SAP deficiency) versus type 2 (XLP-2/XIAP deficiency). Blood 2011;117:1522–9.Google Scholar
Milone, MC, Tsai, DE, Hodinka, RL, et al. Treatment of primary Epstein–Barr virus infection in patients with X-linked lymphoproliferative disease using B-cell- directed therapy. Blood 2005;105:994–6.Google Scholar
Finn, A, Hadzic, N, Morgan, G, Strobel, S, Levinsky, RJ. Prognosis of chronic granulomatous disease. Arch Dis Child 1990;65:942–5.Google Scholar
Muorah, M, Hinds, R, Verma, A, et al. Liver abscesses in children in the developed world: a single centre experience. J Pediatr Gastroenterol Nutr 2006;42:201–6.Google Scholar
Yonkof, JR, Gupta, A, Fu, P, Garabedian, E, Dalal, J; and the United States Immunodeficiency Network Consortium. Role of Allogeneic Hematopoietic Stem Cell Transplant for Chronic Granulomatous Disease (CGD): a Report of the United States Immunodeficiency Network. J Clin Immunol 2019;39:448–58.Google Scholar
Rand, EB, Karpen, SJ, Kelly, S, et al. Treatment of neonatal hemochromatosis with exchange transfusion and intravenous immunoglobulin. J Pediatr 2009;155:566–71.Google Scholar
Tung, J, Hadzic, N, Layton, M, et al. Bone marrow failure in children with acute liver failure. J Pediatr Gastroenterology Nutr 2000;31:557–61.Google Scholar
Maggiore, G, Sciveres, M, Fabre, M, et al. Giant cell hepatitis with autoimmune hemolytic anemia in early childhood: long-term outcome in 16 children.J Pediatr 2011;159:127–32.Google Scholar
Whitington, PF, Kelly, S. Outcome of pregnancies at risk for neonatal hemochromatosis is improved by treatment with high-dose intravenous immunoglobulin. Pediatrics 2008;121:e1615e1621.Google Scholar
INIS Collaborative Group. Treatment of neonatal sepsis with intravenous immune globulin. N Engl J Med 2011;365:1201–11.Google Scholar
Bucuvalas, JC, Anand, R. Studies of the Pediatric Liver Transplantation Research Group. Treatment with immunoglobulin improves outcome for pediatric liver transplant recipients. Liver Transplant 2009;15:1564–9.Google Scholar
Krischock, L, Marks, SD. Induction therapy: why, when, and which agent? Pediatr Transplant 2010;14:298313.Google Scholar
Soliman, T, Hetz, H, Burghuber, C, et al. Short-term induction therapy with anti- thymocyte globulin and delayed use of calcineurin inhibitors in orthotopic liver transplantation. Liver Transplant 2007;13:1039–44.Google Scholar
Kerkar, N, Morotti, RA, Iyer, K, et al. Anti-lymphocyte therapy successfully controls late “cholestatic” rejection in pediatric liver transplant recipients. Clin Transplant 2011;25:E584E591.Google Scholar
Hadzić, N, Height, S, Ball, S, et al. Evolution in the management of acute liver failure-associated aplastic anaemia in children: a single centre experience. J Hepatol 2008;48:6873.Google Scholar
Norin, S, Kimby, E, Ericzon, BG, et al. Posttransplant lymphoma: a single-center experience of 500 liver transplantations. Med Oncol 2004;21:273–84.Google Scholar
Hale, DA. Biological effects of induction immunosuppression. Curr Opin Immunol 2004;16:565–70.Google Scholar
Spada, M, Petz, W, Bertani, A, et al. Randomized trial of basiliximab induction versus steroid therapy in ediatric liver allograft recipients under tacrolimus immunosuppression.Am J Transplant 2006;6:1913–21.Google Scholar
Rovelli, A, Corti, P, Beretta, C, et al. Alemtuzumab for giant cell hepatitis with autoimmune hemolytic anemia. J Pediatr Gastroenterol Nutr 2007;45:596–9.Google Scholar
Schub, N, Günther, A, Schrauder, A, et al. Therapy of steroid-refractory acute GVHD with CD52 antibody alemtuzumab is effective. Bone Marrow Transplant 2011;46:143–7.Google Scholar
Gutierrez-Aguirre, CH, Cantu’-Rodriguez, OG, Borjas-Almaguer, OD, et al. Effectiveness of subcutaneous low-dose alemtuzumab and rituximab combination therapy for steroid-resistant chronic graft-versus-host disease. Haematologica 2012;97:717–22.Google Scholar
Kato, T, Selvaggi, G, Panagiotis, T, et al. Pediatric liver transplant with campath 1 H induction: preliminary report. Transplant Proc 2006;38:3609–11.Google Scholar
Levitsky, J, Thudi, K, Ison, MG, et al. Alemtuzumab induction in non-hepatitis C positive liver transplant recipients. Liver Transplant 2011;17:32–7.Google Scholar
Taylor, RP, Lindorfer, MA. Immunotherapeutic mechanisms of anti-CD20 monoclonal antibodies. Curr Opin Immunol 2008;20:444–9.Google Scholar
Cooper, N, Arnold, DM. The effect of rituximab on humoral and cell mediated immunity and infection in the treatment of autoimmune diseases. Br J Haematol 2010;149:313.Google Scholar
Gobert, D, Bussel, JB, Cunningham-Rundles, C, et al. Efficacy and safety of rituximab in common variable immunodeficiency-associated immune cytopenias: a retrospective multicentre study on 33 patients. Br J Haematol 2011;155:498508.Google Scholar
Messahel, B, Taj, MM, Hobson, R, et al. Single agent efficacy of rituximab in childhood immunosuppression related lymphoproliferative disease: a United Kingdom Children’s Cancer Study Group (UKCCSG) retrospective review. Leuk Lymphoma 2006;47:2584–9.Google Scholar
Miloh, T, Manwani, D, Morotti, R, et al. Giant cell hepatitis and autoimmune hemolytic anemia successfully treated with rituximab. J Pediatr Gastroenterol Nutr 2007;44:634–6.Google Scholar
Czubkowski, P, Williams, M, Bagia, S, Kelly, D, Gupte, G. Immune-mediated hemolytic anemia in children after liver and small bowel transplantation. Liver Transplant 2011;17:921–4.Google Scholar
Shores, D, Kobak, G, Pegram, LD, Whitington, PF, Shneider, BL. Giant cell hepatitis and immune thrombocytopenic purpura: reversal of liver failure with rituximab therapy. J Pediatr Gastroenterol Nutr 2012;55(4):e128e130.Google Scholar
Miloh, T, Arnon, R, Roman, E, et al. Autoimmune hemolytic anemia and idiopathic thrombocytopenic purpura in pediatric solid organ transplant recipients, report of five cases and review of the literature. Pediatr Transplant 2011;15:870–8.Google Scholar
Siebold, L, Dick, AA, Thompson, R, et al. Recurrent low gamma-glutamyl transpeptidase cholestasis following liver transplantation for bile salt export pump (BSEP) disease (posttransplant recurrent BSEP disease). Liver Transplant 2010;16:856–63.Google Scholar
Tsuda, M, Moritoki, Y, Lian, ZX, et al. Biochemical and immunologic effects of rituximab in primary biliary cirrhosis patients with an incomplete response to ursodeoxycholic acid. Hepatology 2012;55:512–21.Google Scholar
Wolach, O, Shpilberg, O, Lahav, M. Neutropenia after rituximab treatment: new insights on a late complication. Curr Opin Hematol 2012;19(1):32–8.Google Scholar
Cambridge, G, Leandro, MJ, Teodorescu, M, et al. B cell depletion therapy in systemic lupus erythematosus: effect on autoantibody and antimicrobial antibody profiles. Arthritis Rheum 2006;54:3612–22.Google Scholar
Kolstad, A, Holte, H, Fossa, A, Lauritzsen, GF, Gaustad, P, Torfoss, D. Pneumocystis jirovecii pneumonia in B-cell lymphoma patients treated with the rituximab-CHOEP-14 regimen. Haematologica 2007;92:139–40.Google Scholar
Green, M, Michaels, MG, Katz, BZ, et al. CMV-IVIG for prevention of Epstein–Barr virus disease and posttransplant lymphoproliferative disease in pediatric liver transplant recipients. Am J Transplant 2006;6:1906–12.Google Scholar
Patterson, SJ, Angus, PW. Post-liver transplant hepatitis B prophylaxis: the role of oral nucleos(t)ide analogues. Curr Opin Organ Transplant 2009;14:225–30.Google Scholar
Grunebaum, E, Avitzur, Y. Liver-associated immune abnormalities. Autoimmun Rev 2019;18:1520.Google Scholar
Shah, T, Cale, C, Hadzic, N, Jones, A. Dedicator of cytokinesis 8 deficiency: a predisposition to sclerosing cholangitis. Clin Immunol 2014;155: 71–3.Google Scholar
Yazdani, R, Fekrvand, S, Shahkarami, S, et al.The hyper IgM syndromes: epidemiology, pathogenesis, clinical manifestations, diagnosis and management. Clin Immunol 2019;198:1930.Google Scholar
de la Morena, MT, Leonard, D, Torgerson, TR, et al. Long-term outcomes of 176 patients with X-linked hyper-IgM syndrome treated with or without hematopoietic cell transplantation. J Allergy Clin Immunol 2017;139:1282–92.Google Scholar
Azzu, V, Kennard, L, Morillo-Gutierrez, B, et al. Liver disease predicts mortality in patients with X-linked immunodeficiency with hyper-IgM but can be prevented by early hematopoietic stem cell transplantation. J Allergy Clin Immunol 2018;141:405–8.e7.Google Scholar
Barbouche, MR, Chen, Q, Carbone, M, et al. Comprehensive review of autoantibodies in patients with hyper-IgM syndrome. Cell Mol Immunol 2018;15:610–17.Google Scholar
Quarello, P, Tandoi, F, Carraro, F, et al. Successful sequential liver and hematopoietic stem cell transplantation in a child with CD40 ligand deficiency and cryptosporidium-induced liver cirrhosis. Transplantation 2018;102:823–8.Google Scholar
Hadzic, N. Is nodular regenerative hyperplasia an immune-mediated phenomenon ? Hepatology 2019;69(6):2717–18.Google Scholar
Bucciol, G, Nicholas, SK, Calvo, PL, et al. Combined liver and hematopoietic stem cell transplantation in patients with X-linked hyper-IgM syndrome. J Allergy Clin Immunol 2019;143:1952–6.e6.Google Scholar
Chavez, MA, White, AC Jr. Novel treatment strategies and drugs in development for cryptosporidiosis. Expert Rev Anti Infect Ther 2018;16:655–61.Google Scholar
Hartleif, S, Lang, P, Handgretinger, R, et al. Outcomes of pediatric identical living-donor liver and hematopoietic stem cell transplantation. Pediatr Transplant 2016;20(7):888–97.Google Scholar
Fischler, B, Baumann, U, Dezsofi, A, et al.Hepatitis E in Children: A Position Paper by the ESPGHAN Hepatology Committee. J Pediatr Gastroenterol Nutr 2016;63(2):288–94.Google Scholar
Westhölter, D, Hiller, J, Denzer, U, et al.HEV-positive blood donations represent a relevant infection risk for immunosuppressed recipients. J Hepatol 2018;69(1):3642.Google Scholar
Acharya, C, Dharel, N, Sterling, RK. Chronic liver disease in the human immunodeficiency virus patient. Clin Liver Dis 2015;19(1):122.Google Scholar
Blumberg, EA, Rogers, CC. Solid organ transplantation in the HIV-infected patient: Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant 2019;17:e13499.Google Scholar
Rowley, MW, Patel, A, Zhou, W, Wong, M, Seetharam, AB. Immune reconstitution syndrome with initiation of treatment of HBV/HIV co-infection: activity flare associated with E antigen seroconversion. Ann Hepatol 2019;18(1):220–4.Google Scholar
Sikavi, C, Chen, PH, Lee, AD, Saab, EG, Choi, G, Saab, S. Hepatitis C and human immunodeficiency virus coinfection in the era of direct-acting antiviral agents: no longer a difficult-to-treat population. Hepatology 2018;67(3):847–57.Google Scholar
Saxena, V, Khungar, V, Verna, EC, et al. Safety and efficacy of current direct-acting antiviral regimens in kidney and liver transplant recipients with hepatitis C: results from the HCV-TARGET study. Hepatology 2017;66(4):1090–101Google Scholar
Minoia, F, Bovis, F, Davì, S, et al. Development and initial validation of the macrophage activation syndrome/primary hemophagocytic lymphohistiocytosis score, a diagnostic tool that differentiates primary hemophagocytic lymphohistiocytosis from macrophage activation syndrome. J Pediatr 2017;189:72–8.e3.Google Scholar
Wozniak, LJ, Naini, BV, Hickey, MJ, et al. Acute antibody-mediated rejection in ABO-compatible pediatric liver transplant recipients: case series and review of the literature. Pediatr Transplant 2017;21(1):e12869.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×