Semin Liver Dis
DOI: 10.1055/s-0044-1787076
Review Article

Understanding Hepatic Porphyrias: Symptoms, Treatments, and Unmet Needs

Oluwashanu Balogun
1   Department of Experimental Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
,
Kari Nejak-Bowen
1   Department of Experimental Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
2   Pittsburgh Liver Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
› Author Affiliations
Funding This study was funded by NIH grant 1R01DK124412 to K.N-B.


Abstract

Hepatic porphyrias are a group of metabolic disorders that are characterized by overproduction and accumulation of porphyrin precursors in the liver. These porphyrins cause neurologic symptoms as well as cutaneous photosensitivity, and in some cases patients can experience life-threatening acute neurovisceral attacks. This review describes the acute hepatic porphyrias in detail, including acute intermittent porphyria, hereditary coproporphyria, and variegate porphyria, as well as the hepatic porphyrias with cutaneous manifestations such as porphyria cutanea tarda and hepatoerythropoietic porphyria. Each section will cover disease prevalence, clinical manifestations, and current therapies, including strategies to manage symptoms. Finally, we review new and emerging treatment modalities, including gene therapy through use of adeno-associated vectors and chaperone therapies such as lipid nanoparticle and small interfering RNA-based therapeutics.

Supplementary Material



Publication History

Article published online:
17 May 2024

© 2024. Thieme. All rights reserved.

Thieme Medical Publishers
333 Seventh Avenue, New York, NY 10001, USA.

 
  • References

  • 1 Puy H, Gouya L, Deybach JC. Porphyrias. Lancet 2010; 375 (9718) 924-937
  • 2 Anderson KE, Sassa SS, Bishop DF. et al. Disorders of heme biosynthesis: X-linked sideroblastic anemia and the porphyrias. In: Scriver CR, Beaudet AL, Sly WS. et al, eds, The Metabolic Basis of Inherited Disease. 8th ed. New York:: McGraw-Hill;; 2001
  • 3 Karim Z, Lyoumi S, Nicolas G, Deybach JC, Gouya L, Puy H. Porphyrias: A 2015 update. Clin Res Hepatol Gastroenterol 2015; 39 (04) 412-425
  • 4 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood 2012; 120 (23) 4496-4504
  • 5 Balogun O, Nejak-Bowen K. The hepatic porphyrias: revealing the complexities of a rare disease. Semin Liver Dis 2023; 43 (04) 446-459
  • 6 Ajioka RS, Phillips JD, Weiss RB. et al. Down-regulation of hepcidin in porphyria cutanea tarda. Blood 2008; 112 (12) 4723-4728
  • 7 Phillips JD. Heme biosynthesis and the porphyrias. Mol Genet Metab 2019; 128 (03) 164-177
  • 8 Ajioka RS, Phillips JD, Kushner JP. Biosynthesis of heme in mammals. Biochim Biophys Acta 2006; 1763 (07) 723-736
  • 9 Ogun AS, Joy NV, Valentine M. . Biochemistry, Heme Synthesis. Treasure Island (FL): StatPearls; 2024
  • 10 Dailey HA, Meissner PN. Erythroid heme biosynthesis and its disorders. Cold Spring Harb Perspect Med 2013; 3 (04) a011676
  • 11 Anderson KE. Porphyrias: an overview. In: Connor R. , ed, UpToDate. Waltham, MA: . Accessed on April 15, 2024. AT: https://www.uptodate.com/contents/porphyrias-an-overview
  • 12 Schultz IJ, Chen C, Paw BH, Hamza I. Iron and porphyrin trafficking in heme biogenesis. J Biol Chem 2010; 285 (35) 26753-26759
  • 13 Peoc’h K, Nicolas G, Schmitt C. et al. Regulation and tissue-specific expression of δ-aminolevulinic acid synthases in non-syndromic sideroblastic anemias and porphyrias. Mol Genet Metab 2019; 128 (03) 190-197
  • 14 Elbirt KK, Bonkovsky HL. Heme oxygenase: recent advances in understanding its regulation and role. Proc Assoc Am Physicians 1999; 111 (05) 438-447
  • 15 America Porphyria Foundation. Types of Porphyria. 2010–2022
  • 16 Warren MJSAG. Tetrapyrroles Birth, Life and Death. New York, NY:: Springer;; 2009
  • 17 Doss M, Look D, Henning H. et al. Hepatic porphyrins and urinary porphyrins and porphyrin precursors in liver cirrhosis. Klin Wochenschr 1972; 50 (22) 1025-1032
  • 18 Maruno M, Furuyama K, Akagi R. et al. Highly heterogeneous nature of delta-aminolevulinate dehydratase (ALAD) deficiencies in ALAD porphyria. Blood 2001; 97 (10) 2972-2978
  • 19 Ramanujam V-MS, Anderson KE. Porphyria diagnostics-Part 1: a brief overview of the porphyrias. Curr Protoc Hum Genet 2015;86:17.20.1117.20.26
  • 20 Mohan G, Madan A. Ala Dehydratase Deficiency Porphyria. Treasure Island (FL):: StatPearls Publishing;; 2023
  • 21 Kappas A. The porphyrias. In: The Metabolic and Molecular Basis of Inherited Disease. 7th ed. 1995: 2103-2159
  • 22 Akagi R, Kato N, Inoue R, Anderson KE, Jaffe EK, Sassa S. delta-Aminolevulinate dehydratase (ALAD) porphyria: the first case in North America with two novel ALAD mutations. Mol Genet Metab 2006; 87 (04) 329-336
  • 23 Meyer UA, Schuurmans MM, Lindberg RL. Acute porphyrias: pathogenesis of neurological manifestations. Semin Liver Dis 1998; 18 (01) 43-52
  • 24 Edel Y, Mamet R. Porphyria: what is it and who should be evaluated?. Rambam Maimonides Med J 2018; 9 (02) 9
  • 25 Desnick RJ, Balwani M, Anderson KE. 10 - Inherited Porphyrias. In: Pyeritz RE, Korf BR, Grody WW. eds, Emery and Rimoin's Principles and Practice of Medical Genetics and Genomics. 7th ed. Academic Press; 2021: 373-411
  • 26 Paller A, Mancini A. Photosensitivity and photoreactions. In: Hurwitz Clinical Pediatric Dermatology;. 2016: 448-466.e4
  • 27 Mustajoki P, Desnick RJ. Genetic heterogeneity in acute intermittent porphyria: characterisation and frequency of porphobilinogen deaminase mutations in Finland. Br Med J (Clin Res Ed) 1985; 291 (6494) 505-509
  • 28 Brennan MJW, Cantrill RC. δ-aminolaevulinic acid is a potent agonist for GABA autoreceptors. Nature 1979; 280 (5722) 514-515
  • 29 Mustajoki P, Koskelo P. Hereditary hepatic porphyrias in Finland. Acta Med Scand 1976; 200 (03) 171-178
  • 30 Lenglet H, Schmitt C, Grange T. et al. From a dominant to an oligogenic model of inheritance with environmental modifiers in acute intermittent porphyria. Hum Mol Genet 2018; 27 (07) 1164-1173
  • 31 Floderus Y, Shoolingin-Jordan PM, Harper P. Acute intermittent porphyria in Sweden. Molecular, functional and clinical consequences of some new mutations found in the porphobilinogen deaminase gene. Clin Genet 2002; 62 (04) 288-297
  • 32 Chen B, Solis-Villa C, Hakenberg J. et al. Acute intermittent porphyria: predicted pathogenicity of HMBS variants indicates extremely low penetrance of the autosomal dominant disease. Hum Mutat 2016; 37 (11) 1215-1222
  • 33 Whatley SD, Roberts AG, Llewellyn DH, Bennett CP, Garrett C, Elder GH. Non-erythroid form of acute intermittent porphyria caused by promoter and frameshift mutations distant from the coding sequence of exon 1 of the HMBS gene. Hum Genet 2000; 107 (03) 243-248
  • 34 Bissell DM, Wang B. Acute hepatic porphyria. J Clin Transl Hepatol 2015; 3 (01) 17-26
  • 35 Whatley SD, Badminton MN. Acute intermittent porphyria. In: Adam MP, Mirzaa GM, Pagon RA. , et al, eds, GeneReviews. Seattle (WA): University of Washington;; 1993
  • 36 Jericó D, Luis EO, Cussó L. et al. Brain ventricular enlargement in human and murine acute intermittent porphyria. Hum Mol Genet 2020; 29 (19) 3211-3223
  • 37 Gonzalez-Mosquera LF, Sonthalia S. Acute Intermittent Porphyria. Treasure Island (FL):: StatPearls Publishing;; 2023
  • 38 O'Malley R, Rao G, Stein P, Bandmann O. Porphyria: often discussed but too often missed. Pract Neurol 2018; 18 (05) 352-358
  • 39 Innala E, Andersson C. Screening for hepatocellular carcinoma in acute intermittent porphyria: a 15-year follow-up in northern Sweden. J Intern Med 2011; 269 (05) 538-545
  • 40 Wang B, Bissell DM. Hereditary coproporphyria. In: Adam MP, Everman DB, Mirzaa GM. , et al, eds, GeneReviews. Seattle (WA):: University of Washington, Seattle;; 1993
  • 41 Tchernitchko D, Tavernier Q, Lamoril J. et al. A variant of peptide transporter 2 predicts the severity of porphyria-associated kidney disease. J Am Soc Nephrol 2017; 28 (06) 1924-1932
  • 42 Gugig R, Rosenthal P. Porphyria. In: Aminoff MJ, Daroff RB. eds, Encyclopedia of the Neurological Sciences. New York:: Academic Press;; 2003: 26-34
  • 43 Anderson KE, Collins S. Open-label study of hemin for acute porphyria: clinical practice implications. Am J Med 2006; 119 (09) 801.e19-8 -01.e24
  • 44 Fontanellas A, Ávila MA, Anderson KE, Deybach JC. Current and innovative emerging therapies for porphyrias with hepatic involvement. J Hepatol 2019; 71 (02) 422-433
  • 45 Bissell DM, Anderson KE, Bonkovsky HL. Porphyria. N Engl J Med 2017; 377 (09) 862-872
  • 46 Ricci A, Ventura P. Givosiran for the treatment of acute hepatic porphyria. Expert Rev Clin Pharmacol 2022; 15 (04) 383-393
  • 47 Soonawalla ZF, Orug T, Badminton MN. et al. Liver transplantation as a cure for acute intermittent porphyria. Lancet 2004; 363 (9410) 705-706
  • 48 Wahlin S, Harper P, Sardh E, Andersson C, Andersson DE, Ericzon BG. Combined liver and kidney transplantation in acute intermittent porphyria. Transpl Int 2010; 23 (06) e18-e21
  • 49 Yasuda M, Gan L, Chen B. et al. Homozygous hydroxymethylbilane synthase knock-in mice provide pathogenic insights into the severe neurological impairments present in human homozygous dominant acute intermittent porphyria. Hum Mol Genet 2019; 28 (11) 1755-1767
  • 50 Ma E, Mar V, Varigos G, Nicoll A, Ross G. Haem arginate as effective maintenance therapy for hereditary coproporphyria. Australas J Dermatol 2011; 52 (02) 135-138
  • 51 Elder G, Harper P, Badminton M, Sandberg S, Deybach JC. The incidence of inherited porphyrias in Europe. J Inherit Metab Dis 2013; 36 (05) 849-857
  • 52 Blake D, McManus J, Cronin V, Ratnaike S. Fecal coproporphyrin isomers in hereditary coproporphyria. Clin Chem 1992; 38 (01) 96-100
  • 53 Towns C, Balakrishnan S, Florkowski C, Davies A, Barrington-Ward E. High penetrance, recurrent attacks and thrombus formation in a family with hereditary coproporphyria. JIMD Rep 2022; 63 (03) 211-215
  • 54 Kühnel A, Gross U, Doss MO. Hereditary coproporphyria in Germany: clinical-biochemical studies in 53 patients. Clin Biochem 2000; 33 (06) 465-473
  • 55 Balwani M, Sardh E, Ventura P. et al; ENVISION Investigators. Phase 3 trial of RNAi therapeutic givosiran for acute intermittent porphyria. N Engl J Med 2020; 382 (24) 2289-2301
  • 56 Logan GM, Weimer MK, Ellefson M, Pierach CA, Bloomer JR. Bile porphyrin analysis in the evaluation of variegate porphyria. N Engl J Med 1991; 324 (20) 1408-1411
  • 57 Ferrer MD, Tauler P, Sureda A, Palacín C, Tur JA, Pons A. Antioxidants restore protoporphyrinogen oxidase in variegate porphyria patients. Eur J Clin Invest 2013; 43 (07) 668-678
  • 58 Eales L, Day RS, Blekkenhorst GH. The clinical and biochemical features of variegate porphyria: an analysis of 300 cases studied at Groote Schuur Hospital, Cape Town. Int J Biochem 1980; 12 (5-6): 837-853
  • 59 von und zu Fraunberg M, Timonen K, Mustajoki P, Kauppinen R. Clinical and biochemical characteristics and genotype-phenotype correlation in Finnish variegate porphyria patients. Eur J Hum Genet 2002; 10 (10) 649-657
  • 60 Long C, Smyth SJ, Woolf J. et al. Detection of latent variegate porphyria by fluorescence emission spectroscopy of plasma. Br J Dermatol 1993; 129 (01) 9-13
  • 61 Pinder VA, Holden ST, Deshpande C. et al. Homozygous variegate porphyria presenting with developmental and language delay in childhood. Clin Exp Dermatol 2013; 38 (07) 737-740
  • 62 Meissner PN, Dailey TA, Hift RJ. et al. A R59W mutation in human protoporphyrinogen oxidase results in decreased enzyme activity and is prevalent in South Africans with variegate porphyria. Nat Genet 1996; 13 (01) 95-97
  • 63 Singal AK, Anderson KE. Variegate porphyria. In: GeneReviews [Internet]. Seattle, WA:: University of Washington;; 2013
  • 64 William DJ, Elston DM, Treat JR, Rosenbach MA. Andrews' Diseases of the Skin. 13th ed. Elsevier Inc;; 2020: 515-546
  • 65 Patterson JW. Metabolic and storage diseases. In: Weedon's Skin Pathology. 5th ed. Elsevier Limited;; 2010: 593-616
  • 66 Schulenburg-Brand D, Gardiner T, Guppy S. et al. An audit of the use of gonadorelin analogues to prevent recurrent acute symptoms in patients with acute porphyria in the United Kingdom. JIMD Rep 2017; 36: 99-107
  • 67 Marsden JT, Guppy S, Stein P. et al. Audit of the use of regular haem arginate infusions in patients with acute porphyria to prevent recurrent symptoms. JIMD Rep 2015; 22: 57-65
  • 68 Schneider-Yin X, van Tuyll van Serooskerken AM, Went P. et al. Hepatocellular carcinoma in variegate porphyria: a serious complication. Acta Derm Venereol 2010; 90 (05) 512-515
  • 69 Singal AK. Porphyria cutanea tarda: recent update. Mol Genet Metab 2019; 128 (03) 271-281
  • 70 Toussi A, Le ST, Merleev AA. et al. Porphyria cutanea tarda presenting as a lichenoid eruption. Photodermatol Photoimmunol Photomed 2021; 37 (03) 233-235
  • 71 de Verneuil H, Aitken G, Nordmann Y. Familial and sporadic porphyria cutanea: two different diseases. Hum Genet 1978; 44 (02) 145-151
  • 72 Elder GH, Lee GB, Tovey JA. Decreased activity of hepatic uroporphyrinogen decarboxylase in sporadic porphyria cutanea tarda. N Engl J Med 1978; 299 (06) 274-278
  • 73 Bonkovsky HL, Lambrecht RW, Shan Y. Iron as a co-morbid factor in nonhemochromatotic liver disease. Alcohol 2003; 30 (02) 137-144
  • 74 Bonkovsky HL, Poh-Fitzpatrick M, Pimstone N. et al. Porphyria cutanea tarda, hepatitis C, and HFE gene mutations in North America. Hepatology 1998; 27 (06) 1661-1669
  • 75 Muñoz-Santos C, Guilabert A, Moreno N. et al. Familial and sporadic porphyria cutanea tarda: clinical and biochemical features and risk factors in 152 patients. Medicine (Baltimore) 2010; 89 (02) 69-74
  • 76 Handler NS, Handler MZ, Stephany MP, Handler GA, Schwartz RA. Porphyria cutanea tarda: an intriguing genetic disease and marker. Int J Dermatol 2017; 56 (06) e106-e117
  • 77 McColl KE, Moore MR, Thompson GG, Goldberg A. Abnormal haem biosynthesis in chronic alcoholics. Eur J Clin Invest 1981; 11 (06) 461-468
  • 78 Fontanellas A, Martínez-Fresno M, Garrido-Astray MC. et al. Smoking but not homozygosity for CYP1A2 g-163A allelic variant leads to earlier disease onset in patients with sporadic porphyria cutanea tarda. Exp Dermatol 2010; 19 (08) e326-e328
  • 79 Cruz-Rojo J, Fontanellas A, Morán-Jiménez MJ. et al. Precipitating/aggravating factors of porphyria cutanea tarda in Spanish patients. Cell Mol Biol 2002; 48 (08) 845-852
  • 80 Quansah R, Cooper CJ, Said S, Bizet J, Paez D, Hernandez GT. Hepatitis C- and HIV-induced porphyria cutanea tarda. Am J Case Rep 2014; 15: 35-40
  • 81 Nishina S, Hino K, Korenaga M. et al. Hepatitis C virus-induced reactive oxygen species raise hepatic iron level in mice by reducing hepcidin transcription. Gastroenterology 2008; 134 (01) 226-238
  • 82 Cruz MJ, Alves S, Baudrier T, Azevedo F. Porphyria cutanea tarda induced by tamoxifen. Dermatol Online J 2010; 16 (09) 2
  • 83 Bulaj ZJ, Franklin MR, Phillips JD. et al. Transdermal estrogen replacement therapy in postmenopausal women previously treated for porphyria cutanea tarda. J Lab Clin Med 2000; 136 (06) 482-488
  • 84 Roenigk Jr HH, Gottlob ME. Estrogen-induced porphyria cutanea tarda. Report of three cases. Arch Dermatol 1970; 102 (03) 260-266
  • 85 Gibson GE, McEvoy MT. Coexistence of lupus erythematosus and porphyria cutanea tarda in fifteen patients. J Am Acad Dermatol 1998; 38 (04) 569-573
  • 86 Rodrigues N, Caeiro F, Santana A, Mendes T, Lopes L. Porphyria cutanea tarda in a patient with end-stage renal disease: a case of successful treatment with deferoxamine and ferric carboxymaltose. Case Rep Nephrol 2017; 2017: 4591871
  • 87 Christiansen AL, Bygum A, Hother-Nielsen O, Rasmussen LM. Diagnosing diabetes mellitus in patients with porphyria cutanea tarda. Int J Dermatol 2018; 57 (07) 763-769
  • 88 McKenna DB, Browne M, O'Donnell R, Murphy GM. Porphyria cutanea tarda and hematologic malignancy–a report of 4 cases. Photodermatol Photoimmunol Photomed 1997; 13 (04) 143-146
  • 89 Rudnick S, Phillips J, Bonkovsky H. et al. Familial porphyria cutanea tarda. In: Adam MP, Mirzaa GM, Pagon RA. , et al, eds, GeneReviews. Seattle, WA:: University of Washington;; 1993
  • 90 Ryan Caballes F, Sendi H, Bonkovsky HL. Hepatitis C, porphyria cutanea tarda and liver iron: an update. Liver Int 2012; 32 (06) 880-893
  • 91 Ellervik C, Birgens H, Tybjaerg-Hansen A, Nordestgaard BG. Hemochromatosis genotypes and risk of 31 disease endpoints: meta-analyses including 66,000 cases and 226,000 controls. Hepatology 2007; 46 (04) 1071-1080
  • 92 Berenguer J, Blasco J, Cardenal C. et al. Hepatoerythropoietic porphyria: neuroimaging findings. AJNR Am J Neuroradiol 1997; 18 (08) 1557-1560
  • 93 de Verneuil H, Beaumont C, Deybach JC, Nordmann Y, Sfar Z, Kastally R. Enzymatic and immunological studies of uroporphyrinogen decarboxylase in familial porphyria cutanea tarda and hepatoerythropoietic porphyria. Am J Hum Genet 1984; 36 (03) 613-622
  • 94 Rudnick S, Phillips J, Bonkovsky H. et al. Hepatoerythropoietic Porphyria. In: Adam MP, Mirzaa GM, Pagon RA. , et al, eds, GeneReviews. Seattle (WA):: University of Washington;; 1993
  • 95 Cantatore-Francis JL, Cohen-Pfeffer J, Balwani M. et al. Hepatoerythropoietic porphyria misdiagnosed as child abuse: cutaneous, arthritic, and hematologic manifestations in siblings with a novel UROD mutation. Arch Dermatol 2010; 146 (05) 529-533
  • 96 Elder GH. Hepatic porphyrias in children. J Inherit Metab Dis 1997; 20 (02) 237-246
  • 97 Anderson KE, Bloomer JR, Bonkovsky HL. et al. Recommendations for the diagnosis and treatment of the acute porphyrias. Ann Intern Med 2005; 142 (06) 439-450
  • 98 Marcacci M, Ricci A, Cuoghi C, Marchini S, Pietrangelo A, Ventura P. Challenges in diagnosis and management of acute hepatic porphyrias: from an uncommon pediatric onset to innovative treatments and perspectives. Orphanet J Rare Dis 2022; 17 (01) 160
  • 99 Buttery JE. Is the Watson-Schwartz screening method for porphobilinogen reliable?. Clin Chem 1995; 41 (11) 1670-1671
  • 100 Thadani H, Deacon A, Peters T. Diagnosis and management of porphyria. BMJ 2000; 320 (7250) 1647-1651
  • 101 Bonkovsky HL, Maddukuri VC, Yazici C. et al. Acute porphyrias in the USA: features of 108 subjects from porphyrias consortium. Am J Med 2014; 127 (12) 1233-1241
  • 102 Andersen J, Thomsen J, Enes AR, Sandberg S, Aarsand AK. Health-related quality of life in porphyria cutanea tarda: a cross-sectional registry based study. Health Qual Life Outcomes 2020; 18 (01) 84
  • 103 Mustajoki P, Nordmann Y. Early administration of heme arginate for acute porphyric attacks. Arch Intern Med 1993; 153 (17) 2004-2008
  • 104 Kuter DJ, Bonkovsky HL, Monroy S. et al; ENVISION Investigators. Efficacy and safety of givosiran for acute hepatic porphyria: Final results of the randomized phase III ENVISION trial. J Hepatol 2023; 79 (05) 1150-1158
  • 105 Massachi S, Epstein J, Hurd J, Bonkovsky HL. Cost savings with hemin versus givosiran for the treatment of patients with acute intermittent porphyria (AIP). J Med Econ 2020; 23 (12) 1441-1449
  • 106 Willandt B, Langendonk JG, Biermann K. et al. Liver fibrosis associated with iron accumulation due to long-term heme-arginate treatment in acute intermittent porphyria: a case series. JIMD Rep 2016; 25: 77-81
  • 107 Schmitt C, Lenglet H, Yu A. et al. Recurrent attacks of acute hepatic porphyria: major role of the chronic inflammatory response in the liver. J Intern Med 2018; 284 (01) 78-91
  • 108 To-Figueras J, Wijngaard R, García-Villoria J. et al. Dysregulation of homocysteine homeostasis in acute intermittent porphyria patients receiving heme arginate or givosiran. J Inherit Metab Dis 2021; 44 (04) 961-971
  • 109 Longo M, Paolini E, Meroni M, Dongiovanni P. Cutting-edge therapies and novel strategies for acute intermittent porphyria: step-by-step towards the solution. Biomedicines 2022; 10 (03) 10
  • 110 Yasuda M, Bishop DF, Fowkes M, Cheng SH, Gan L, Desnick RJ. AAV8-mediated gene therapy prevents induced biochemical attacks of acute intermittent porphyria and improves neuromotor function. Mol Ther 2010; 18 (01) 17-22
  • 111 Unzu C, Sampedro A, Mauleón I. et al. Sustained enzymatic correction by rAAV-mediated liver gene therapy protects against induced motor neuropathy in acute porphyria mice. Mol Ther 2011; 19 (02) 243-250
  • 112 Unzu C, Sampedro A, Mauleón I. et al. Porphobilinogen deaminase over-expression in hepatocytes, but not in erythrocytes, prevents accumulation of toxic porphyrin precursors in a mouse model of acute intermittent porphyria. J Hepatol 2010; 52 (03) 417-424
  • 113 D'Avola D, López-Franco E, Sangro B. et al. Phase I open label liver-directed gene therapy clinical trial for acute intermittent porphyria. J Hepatol 2016; 65 (04) 776-783
  • 114 Meliani A, Boisgerault F, Hardet R. et al. Antigen-selective modulation of AAV immunogenicity with tolerogenic rapamycin nanoparticles enables successful vector re-administration. Nat Commun 2018; 9 (01) 4098
  • 115 Serrano-Mendioroz I, Sampedro A, Serna N. et al. Bioengineered PBGD variant improves the therapeutic index of gene therapy vectors for acute intermittent porphyria. Hum Mol Genet 2018; 27 (21) 3688-3696
  • 116 Serrano-Mendioroz I, Sampedro A, Alegre M, Enríquez de Salamanca R, Berraondo P, Fontanellas A. An inducible promoter responsive to different porphyrinogenic stimuli improves gene therapy vectors for acute intermittent porphyria. Hum Gene Ther 2018; 29 (04) 480-491
  • 117 Bustad HJ, Toska K, Schmitt C. et al. A pharmacological chaperone therapy for acute intermittent porphyria. Mol Ther 2020; 28 (02) 677-689
  • 118 Córdoba KM, Serrano-Mendioroz I, Jericó D. et al. Recombinant porphobilinogen deaminase targeted to the liver corrects enzymopenia in a mouse model of acute intermittent porphyria. Sci Transl Med 2022; 14 (627) eabc0700
  • 119 Jiang L, Berraondo P, Jericó D. et al. Systemic messenger RNA as an etiological treatment for acute intermittent porphyria. Nat Med 2018; 24 (12) 1899-1909
  • 120 Russell JO, Monga SP. Wnt/β-catenin signaling in liver development, homeostasis, and pathobiology. Annu Rev Pathol 2018; 13: 351-378
  • 121 Saggi H, Maitra D, Jiang A. et al. Loss of hepatocyte β-catenin protects mice from experimental porphyria-associated liver injury. J Hepatol 2019; 70 (01) 108-117
  • 122 Yasuda M, Desnick RJ. Murine models of the human porphyrias: contributions toward understanding disease pathogenesis and the development of new therapies. Mol Genet Metab 2019; 128 (03) 332-341
  • 123 Sardh E, Harper P, Balwani M. et al. Phase 1 trial of an RNA interference therapy for acute intermittent porphyria. N Engl J Med 2019; 380 (06) 549-558