1
|
Dong Y, Zheng M, Ding W, Guan H, Xiao J, Li F. Nrf2 activators for the treatment of rare iron overload diseases: From bench to bedside. Redox Biol 2025; 81:103551. [PMID: 39965404 PMCID: PMC11876910 DOI: 10.1016/j.redox.2025.103551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 02/20/2025] Open
Abstract
Iron overload and related oxidative damage are seen in many rare diseases, due to mutation of iron homeostasis-related genes. As a core regulator on cellular antioxidant reaction, Nrf2 can also decrease systemic and cellular iron levels by regulating iron-related genes and pathways, making Nrf2 activators very good candidates for the treatment of iron overload disorders. Successful examples include the clinical use of omaveloxolone for Friedreich's Ataxia and dimethyl fumarate for relapsing-remitting multiple sclerosis. Despite these uses, the therapeutic potentials of Nrf2 activators for iron overload disorders may be overlooked in clinical practice. Therefore, this study talks about the potential use, possible mechanisms, and precautions of Nrf2 activators in treating rare iron overload diseases. In addition, a combination therapy with Nrf2 activators and iron chelators is proposed for clinical reference, aiming to facilitate the clinical use of Nrf2 activators for more iron overload disorders.
Collapse
Affiliation(s)
- Yimin Dong
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Zheng
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weizhong Ding
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hanfeng Guan
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Feng Li
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Liu J, Zhao J, He J, Li Y, Xu J, Xiao C, Zhang Y, Chen H, Hu Y, Fan C, Liu X. Hepcidin mediates the disorder of iron homeostasis and mitochondrial function in mice under hypobaric hypoxia exposure. Apoptosis 2025; 30:1076-1091. [PMID: 39987410 DOI: 10.1007/s10495-025-02079-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2025] [Indexed: 02/24/2025]
Abstract
The human body to prolonged high-altitude environments exposure can cause the decreased hepcidin and the disorder of iron homeostasis. Few animal studies have examined the molecular basis of iron metabolism disorder and the impact of hepcidin on the complex phenotype of high-altitude environments. In this study, we display reduced hepcidin level and impaired iron homeostasis upon hypobaric hypoxia, accompanied by mitochondrial dysfunction, abnormal blood cells and metabolic disorder. Importantly, mice overexpressing hepcidin show resistance to hypoxia-induced mitochondrial injury in liver and improve the body homeostasis. The comprehensive characterization of the metabolic pathways analysis demonstrates the porphyrin metabolism modulated by hepcidin may play a significant role in improving blood cell abnormalities upon high altitude hypoxia. In addition, hepcidin transcription is inhibited by histone modification and interruption of SMAD signaling pathway in hepatocytes in response to hypoxic stress to decrease hepcidin level. Our data highlight the key role of hepcidin in regulating the complex phenotype of high-altitude environments by altered metabolic and mitochondrial function. These results provide a theoretical reference to understanding the role of hepcidin in maintaining physiological function in high altitude, thus assisting in the development of strategies to better prevent and alleviate imbalance of energy homeostasis and adverse effects.
Collapse
Affiliation(s)
- Jiayao Liu
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Jialin Zhao
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Jintao He
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Yuhui Li
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Jie Xu
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Chenxi Xiao
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Yuyu Zhang
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Honghong Chen
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Yajie Hu
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Chunxiang Fan
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China.
| | - Xinhua Liu
- Department of Traditional Chinese Medicine, Pharmacophenomics Laboratory, Human Phenome Institute, Shanghai Pudong Hospital, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China.
| |
Collapse
|
3
|
Jia N, Yimin Y, Li M, Jiang L, Liu Y. Identification of a novel nonsense mutation and a recurrent missense mutation in UROS gene in a patient with congenital erythropoietic porphyria. Front Genet 2025; 16:1486595. [PMID: 40230347 PMCID: PMC11994962 DOI: 10.3389/fgene.2025.1486595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/19/2025] [Indexed: 04/16/2025] Open
Abstract
Background: Congenital erythropoietic porphyria (CEP, OMIM #263700) is a rare autosomal recessive disease characterized by skin photosensitivity, hypertrichosis, scarring in light-exposed areas, erythrodontia, and dark-reddish urine. The severity of the clinical phenotype is directly associated with the complete loss of enzymatic activity resulting from UROS mutations. Methods: To understand the genetic etiology of CEP in a 9-year-old female proband, we checked clinical data and collected peripheral blood samples from her and her parents. Genomic DNA was isolated and subjected to polymerase chain reaction (PCR) amplification. Sanger sequencing was performed to detect potential mutations. Bioinformatics analysis was performed to assess the pathogenicity of the identified variant, and 3D protein modeling was conducted to predict its impact on protein structure. Results: The proband presents with red wine-colored urine in early infancy, reddish-brown, notched incisors, and vellus hair on the forehead and trunk. Blisters develop on sun-exposed areas, leaving hyperpigmented macules after rupture. Sanger sequencing identified a previously reported missense mutation (c 0.425C > T: p.P142L) and a novel nonsense mutation in the UROS gene (c 0.325A > T: p.K109*). Bioinformatic analysis indicated that the c 0.325A > T: p.K109* variant is pathogenic. Structural modeling demonstrated that the heterozygous c.325A > T transversion in exon 6 of UROS caused a K109 termination at the protein's α6 helix chain. Conclusion: Our findings underscored the critical role of Sanger sequencing in the accurate diagnosis of atypical CEP cases and in facilitating informed genetic counseling. The identification of a UROS gene novel mutation in this case indicates a mild phenotype, further expanding the spectrum of disorders associated with UROS variants.
Collapse
Affiliation(s)
- Ning Jia
- Department of central laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yusupu Yimin
- Department of Dermatology, Kashi Prefecture Second People’s Hospital, Xinjiang, China
| | - Ming Li
- Department of Dermatology, The Children’s Hospital of Fudan University, Shanghai, China
| | - Long Jiang
- Department of Dermatologic Surgery, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yeqiang Liu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
4
|
Kaiser N, Magg J, Nägele T, Wolf N, Krägeloh-Mann I. Neurodevelopmental retardation and neurological symptoms in homozygous variegate porphyria: two new cases and a literature review. Orphanet J Rare Dis 2025; 20:139. [PMID: 40114189 PMCID: PMC11924618 DOI: 10.1186/s13023-025-03606-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Genetic porphyrias, namely in their homozygous form, may cause a neurodevelopmental disorder which may even be the clinically dominant feature. But few cases have been described so far. The majority of neurodevelopmental disorders has a genetic cause and there is a big overlap of the clinical presentations due to unspecific symptoms. Additional specific clinical symptoms may enable a phenotypically orientated biochemical and genetic diagnostic approach. Skin lesions occurring in the neonatal period or the first years of life in a child with developmental delay may hint at a genetic porphyria. METHODS We describe the clinical features, biochemical and genetic findings in two new cases, sister and brother, of biallelic resp. homozygous variegate porphyria and review all case reports published until December 2023 after systematic searches in PubMed, MEDLINE, Cochrane and Web of Science. RESULTS A total of 19 patients with biallelic, largely homozygous variegate porphyria have so far been reported of whom 16 were confirmed by genetic testing. In 11 patients, neurodevelopmental problems were reported in addition to skin lesions. Additional symptoms were nystagmus, epileptic seizures as well as sensory neuropathy. Only 2 patients received a brain MRI showing a severe deficit of myelination at the age of 2-3 years suggesting that neurodevelopmental delay in HVP may be associated to hypomyelination. This article adds two cases of a genetic porphyria with developmental delay and epilepsy as well as skin lesions. In our two cases biochemistry revealed a porphyria and consecutive molecular genetic testing showed in each case a homozygous variant in the PPOX gene, which corresponds to a variegate porphyria. Interestingly, magnetic resonance imaging of the brain revealed a severe myelin deficit suggesting hypomyelination in both children. CONCLUSIONS In children with a developmental disorder of unknown cause and early childhood epilepsy, an abnormally light-sensitive or fragile skin may indicate a primary genetic porphyria. Especially variegate porphyria with biallelic variants may present as neurodevelopmental disorder with hypomyelination.
Collapse
Affiliation(s)
- Nadja Kaiser
- Department of Neuropaediatrics, Developmental Neurology, Social Paediatrics, University Children's Hospital, Tübingen, Germany
| | - Janine Magg
- Department of Neuropaediatrics, Developmental Neurology, Social Paediatrics, University Children's Hospital, Tübingen, Germany.
| | - Thomas Nägele
- Department of Radiology, Section of Diagnostic and Interventional Neuroradiology, University Hospital Tübingen, Tübingen, Germany
| | - Nicole Wolf
- Department of Child Neurology, Emma Children's Hospital, Amsterdam UMC - Locatie VUMC and Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ingeborg Krägeloh-Mann
- Department of Neuropaediatrics, Developmental Neurology, Social Paediatrics, University Children's Hospital, Tübingen, Germany
| |
Collapse
|
5
|
Martínez-Ávila MC, Matijasevic-Arcila E, Pantoja-Chica S, Acosta-Cardozo AL. Characterizing hepatic porphyria: Insights from a quaternary care hospital in Bogotá, Colombia (2013-2023). J Int Med Res 2025; 53:3000605251325183. [PMID: 40116802 PMCID: PMC11930460 DOI: 10.1177/03000605251325183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/15/2025] [Indexed: 03/23/2025] Open
Abstract
IntroductionAcute hepatic porphyrias are rare, life-threatening genetic disorders that impair heme biosynthesis, often presenting with nonspecific symptoms that lead to misdiagnosis. This diagnostic challenge and low clinical recognition can delay targeted treatment, increasing morbidity and mortality. Although there have been advances in understanding porphyrias' biochemical pathways, improved diagnostic approaches are still needed, especially in acute care. This study examined diagnostic and clinical patterns of acute hepatic porphyria at a quaternary care hospital in Bogotá, Colombia, to support earlier detection and management.Materials and MethodsThis descriptive, observational, retrospective study reviewed patients diagnosed with acute hepatic porphyria at the Internal Medicine service of a Bogotá hospital from 2013 to 2023. Patients with confirmed diagnoses recorded in the hospital database were included. Data collected covered demographic characteristics, clinical presentation, diagnostic markers, and treatment. Key outcome measures were time to diagnosis, recurrence frequency, and hospitalization duration.ResultsTen patients were included, 80% of whom were female, with a median age of 32 years. Diagnosis was confirmed by urine porphobilinogen tests. All patients reported abdominal pain during attacks; 90% had tachycardia and paresis/weakness of extremities. Attack durations ranged from 4 to 11 days, with 90% treated with hemin. Median hospital stay was 18 days. Drug use and infections were common precipitants, and 40% of female patients had premenstrual-associated attacks.ConclusionsThis study provides a clinical profile of acute hepatic porphyria in a Colombian hospital, highlighting neurovisceral symptoms and female predominance. Findings suggest the need for early diagnostic protocols to prevent treatment delays, although larger studies are required to confirm these findings across different settings.
Collapse
Affiliation(s)
- María C Martínez-Ávila
- Internal Medicine Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
- School of Medicine, Internal Medicine Residency Program, Universidad El Bosque, Bogotá, Colombia
| | | | - Stefania Pantoja-Chica
- Internal Medicine Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
- School of Medicine, Internal Medicine Residency Program, Universidad El Bosque, Bogotá, Colombia
| | | |
Collapse
|
6
|
Balwani M, Keel S, Meissner P, Sonderup M, Stein P, Yasuda M. Case-based discussion of the acute hepatic porphyrias: Updates on pathogenesis, diagnosis and management. Liver Int 2025; 45:e15924. [PMID: 38618923 DOI: 10.1111/liv.15924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/16/2024]
Abstract
The acute hepatic porphyrias (AHPs) include three autosomal dominant disorders, acute intermittent porphyria, variegate porphyria and hereditary coproporphyria, and the ultra-rare autosomal recessive 5-aminolevulinic acid dehydratase-deficient porphyria. All four are characterized by episodic acute neurovisceral attacks that can be life-threatening if left untreated. The attacks are precipitated by factors that induce hepatic 5-aminolevulinic acid synthase 1 (ALAS1), resulting in accumulation of the porphyrin precursors, 5-aminolevulinic acid and porphobilinogen, which are believed to cause neurotoxicity. Diagnosis of these rare disorders is often delayed because the symptoms are non-specific with many common aetiologies. However, once clinical suspicion of an AHP is raised, diagnosis can be made by specialized biochemical testing, particularly during attacks. Moderate or severe attacks are treated with intravenous hemin infusions, together with supportive care to relieve pain and other symptoms. Prophylactic treatments are recommended in patients with confirmed recurrent attacks (≥4 attacks in a maximum period of 12 months), the most effective being givosiran, an RNAi therapeutic targeting hepatocyte ALAS1 mRNA. AHP patients with clinically and/or biochemically active disease are at elevated risk for developing long-term complications, including chronic kidney disease, chronic hypertension and hepatocellular carcinoma, thus, surveillance is recommended. Here, using a case-based format, we provide an update on the pathogenesis, diagnosis and treatment of the AHPs based on literature review and clinical experiences.
Collapse
Affiliation(s)
- Manisha Balwani
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Siobán Keel
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, Washington, USA
| | - Peter Meissner
- Department of Integrative Biomedical Sciences, Division of Medical Biochemistry and Structural Biology, University of Cape Town, Cape Town, South Africa
| | - Mark Sonderup
- Division of Hepatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Penelope Stein
- Department of Haematological Medicine, King's College Hospital, London, UK
| | - Makiko Yasuda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
7
|
Wagner MEU, Frost M, Frystyk J. Acute hepatic porphyria in Denmark; a retrospective study. Orphanet J Rare Dis 2025; 20:93. [PMID: 40022110 PMCID: PMC11871779 DOI: 10.1186/s13023-025-03536-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/27/2024] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Acute hepatic porphyria (AHP) constitutes a class of rare diseases caused by reduced function in enzymes of the heme-biosynthetic pathway. AHP includes acute intermittent porphyria (AIP), hereditary coproporphyria (HCP), variegate porphyria (VP) and the extremely rare δ-aminolevulinic-dehydrase deficiency porphyria (ADP). This retrospective study describes characteristics of the Danish AHP patient population. METHODS Department of Endocrinology at Odense University Hospital serves as national AHP center. We performed a 5-year retrospective description of our AHP cohort using electronic patient journals. We included general symptoms, number of acute attacks, hospitalization rates, long-term sequelae and symptoms, and grouped patients according to creatinine-adjusted urinary baseline excretion (i.e., outside attacks) of the porphyrin precursor porphobilinogen (PBG) in normal-, moderate- and high-excretion and unknown. RESULTS The cohort contained 129 AHP patients, hereof 100 AIP, 12 HCP and 17 VP. Median age was 46.3 (32.1-62.0) years, and 85 (65.9%) were female. During the 5-years, 38 (29.5%) patients experienced symptoms. Hereof, 20 patients were hospitalized with acute attacks or chronic symptoms and treated with human hemin (n = 14). Most frequently reported symptoms were abdominal pain, nausea, vomiting, and neurological disturbances. Symptoms were more common in patients with high PBG baseline excretion (n = 39) as compared to those with moderate (n = 31) or normal (n = 40) PBG excretion (p = 0.002). Furthermore, females dominated the symptomatic group (68.4%). CONCLUSION As reported internationally, AHP is more commonly diagnosed and symptomatic in women, and AIP was the most frequent AHP subtype. Those with an elevated urinary baseline PBG secretion were more likely to report AHP-related symptoms.
Collapse
Affiliation(s)
- Magnus Emil Ulrich Wagner
- Department of Endocrinology, Odense University Hospital, Kløvervænget 6, Entrance 93, Level 4, DK-5000, Odense C, Denmark
| | - Morten Frost
- Department of Endocrinology, Odense University Hospital, Kløvervænget 6, Entrance 93, Level 4, DK-5000, Odense C, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Jan Frystyk
- Department of Endocrinology, Odense University Hospital, Kløvervænget 6, Entrance 93, Level 4, DK-5000, Odense C, Denmark.
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
8
|
Stölzel U, Jost L, Seehofer D, Egger‐Heidrich K, Scheuermann U, Hölig K, Stauch T, Kunadt D, Schuppan D, Schetelig J, Wohmann N, Bornhäuser M, Stölzel F. Paternal Split-Liver Transplantation Followed by Haploidentical Hematopoietic Cell Transplantation in an Adult Patient With Protoporphyria-Induced Liver Failure. EJHAEM 2025; 6:e21092. [PMID: 39967799 PMCID: PMC11833665 DOI: 10.1002/jha2.1092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025]
Abstract
Introduction Erythropoietic Protoporphyria (EPP) caused skin light sensitivity and liver cirrhosis in a 35-year-old patient who subsequently developed liver-failure. Methods In absence of a human leukocyte antigens (HLA)-matched-unrelated donor, the father consented in donating for split liver transplantation (SLT) and allogeneic hematopoietic cell transplantation (HCT). Results After bridging therapy and successful SLT a first paternal HCT resulted in graft failure. For a second haploidentical HCT a different regimen was applied leading to engraftment while protoporphyrin (PP) blood-levels decreased to normal and skin light sensitivity skin disappeared, leading to complete remission in an immunosuppressive-free patient. Conclusion Haploidentical transplantation is a feasible treatment approach in EPP-patients. Trial Registration The authors have confirmed clinical trial registration is not needed for this submission.
Collapse
Affiliation(s)
- Ulrich Stölzel
- Department of Internal Medicine II and Porphyria CenterKlinikum ChemnitzChemnitzGermany
| | - Lasse Jost
- Division of Stem Cell Transplantation and Cellular ImmunotherapiesDepartment of Internal Medicine IIUniversity Hospital Schleswig Holstein Kiel, Kiel UniversityKielGermany
| | - Daniel Seehofer
- Department of VisceralTransplantation, Vascular and Thoracic SurgeryUniversity Hospital of LeipzigLeipzigGermany
| | - Katharina Egger‐Heidrich
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TU DresdenDresdenGermany
| | - Uwe Scheuermann
- Department of VisceralTransplantation, Vascular and Thoracic SurgeryUniversity Hospital of LeipzigLeipzigGermany
| | - Kristina Hölig
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TU DresdenDresdenGermany
| | - Thomas Stauch
- Deutsches Kompetenz‐Zentrum für Porphyriediagnostik und KonsultationMVZ Labor Volkmann und Kollegen GbRKarlsruheGermany
| | - Desiree Kunadt
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TU DresdenDresdenGermany
| | - Detlef Schuppan
- Institute of Translational ImmunologyJohannes Gutenberg University MainzMainzGermany
- Division of GastroenterologyBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Johannes Schetelig
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TU DresdenDresdenGermany
| | - Nils Wohmann
- Department of Internal Medicine II and Porphyria CenterKlinikum ChemnitzChemnitzGermany
| | - Martin Bornhäuser
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TU DresdenDresdenGermany
| | - Friedrich Stölzel
- Division of Stem Cell Transplantation and Cellular ImmunotherapiesDepartment of Internal Medicine IIUniversity Hospital Schleswig Holstein Kiel, Kiel UniversityKielGermany
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TU DresdenDresdenGermany
| |
Collapse
|
9
|
Kaya Ç, Heydari A, Kızılay O, Kızılay O, Sezgin G. Hepatoerythropoietic Porphyria with Coexisting BTD And CNGB1 Genetic Mutations: A First Case Report. Eur J Case Rep Intern Med 2025; 12:005198. [PMID: 40051752 PMCID: PMC11881998 DOI: 10.12890/2025_005198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
Introduction Hepatoerythropoietic porphyria (HEP) is an uncommon autosomal recessive disorder marked by deficiencies in enzymes involved in heme biosynthesis. This results in the build-up of porphyrins and their precursors. Here, we describe a case study of a 17-year-old male who has experienced symptoms of porphyria since early childhood. Case description The patient exhibited initial symptoms of porphyria, including dark-coloured urine, abdominal pain, constipation and cutaneous lesions. Genetic testing at age 17 confirmed a homozygous mutation in the UROD gene, diagnosing HEP. Additional mutations in the CNGB1 and BTD genes contributed to retinitis pigmentosa and biotinidase deficiency, respectively. The patient also experienced complications such as thumb amputation and finger developmental anomalies. Conclusion This case underscores the diagnostic challenges and multidisciplinary management required for patients with complex genetic profiles and rare porphyria subtypes such as HEP. Further research is essential to enhance understanding and treatment strategies for such intricate genetic conditions. LEARNING POINTS This is a first report of hepatoerythropoietic porphyria with coexisting BTD and CNGB1 genetic mutations.
Collapse
Affiliation(s)
- Çağdaş Kaya
- Niğde Ömer Halisdemir Training and Research Hospital, Niğde, Turkey
| | | | - Oğuz Kızılay
- Kocaeli Gebze Fatih State Hospital, Gebze, Turkey
| | - Ozan Kızılay
- Faculty of Medicine, Bahçeşehir University, Istanbul, Turkey
| | - Gülbüz Sezgin
- Sancaktepe State Hospital, Feriha Öz, Department of Internal Medicine, Istanbul, Turkey
| |
Collapse
|
10
|
Córdoba KM, Jericó D, Jiang L, Collantes M, Alegre M, García-Ruiz L, Manzanilla O, Sampedro A, Herranz JM, Insausti I, Martinez de la Cuesta A, Urigo F, Alcaide P, Morán M, Martín MA, Lanciego JL, Lefebvre T, Gouya L, Quincoces G, Unzu C, Hervas-Stubbs S, Falcón-Pérez JM, Alegre E, Aldaz A, Fernández-Seara MA, Peñuelas I, Berraondo P, Martini PGV, Avila MA, Fontanellas A. Systemic messenger RNA replacement therapy is effective in a novel clinically relevant model of acute intermittent porphyria developed in non-human primates. Gut 2025; 74:270-283. [PMID: 39366725 PMCID: PMC11874285 DOI: 10.1136/gutjnl-2024-332619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/16/2024] [Indexed: 10/06/2024]
Abstract
OBJECTIVE Acute intermittent porphyria (AIP) is a rare metabolic disorder caused by haploinsufficiency of hepatic porphobilinogen deaminase (PBGD), the third enzyme of the heme biosynthesis. Individuals with AIP experience neurovisceral attacks closely associated with hepatic overproduction of potentially neurotoxic heme precursors. DESIGN We replicated AIP in non-human primates (NHPs) through selective knockdown of the hepatic PBGD gene and evaluated the safety and therapeutic efficacy of human PBGD (hPBGD) mRNA rescue. RESULTS Intrahepatic administration of a recombinant adeno-associated viral vector containing short hairpin RNA against endogenous PBGD mRNA resulted in sustained PBGD activity inhibition in liver tissue for up to 7 months postinjection. The administration of porphyrinogenic drugs to NHPs induced hepatic heme synthesis, elevated urinary porphyrin precursors and reproduced acute attack symptoms in patients with AIP, including pain, motor disturbances and increased brain GABAergic activity. The model also recapitulated functional anomalies associated with AIP, such as reduced brain perfusion and cerebral glucose uptake, disturbances in hepatic TCA cycle, one-carbon metabolism, drug biotransformation, lipidomic profile and abnormal mitochondrial respiratory chain activity. Additionally, repeated systemic administrations of hPBGD mRNA in this AIP NHP model restored hepatic PBGD levels and activity, providing successful protection against acute attacks, metabolic changes in the liver and CNS disturbances. This approach demonstrated better efficacy than the current standards of care for AIP. CONCLUSION This novel model significantly expands our understanding of AIP at the molecular, biochemical and clinical levels and confirms the safety and translatability of multiple systemic administration of hPBGD mRNA as a potential aetiological AIP treatment.
Collapse
Affiliation(s)
- Karol M Córdoba
- Hepatology: Porphyrias & Carcinogenesis Lab. Solid Tumors Program, CIMA Universidad de Navarra, Pamplona, Spain
| | - Daniel Jericó
- Hepatology: Porphyrias & Carcinogenesis Lab. Solid Tumors Program, CIMA Universidad de Navarra, Pamplona, Spain
| | - Lei Jiang
- Moderna Inc, Cambridge, Massachusetts, USA
| | - María Collantes
- Translational Molecular Imaging Unit (UNIMTRA), and Nuclear Medicine-Department, Clínica Universidad de Navarra (CUN), University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Manuel Alegre
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Clinical Neurophysiology, Clinica Universitaria de Navarra, Pamplona, Spain
| | - Leyre García-Ruiz
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Radiology Department, Clinica Universitaria de Navarra, Pamplona, Spain
| | - Oscar Manzanilla
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Clinical Neurophysiology, Clinica Universitaria de Navarra, Pamplona, Spain
| | - Ana Sampedro
- Hepatology: Porphyrias & Carcinogenesis Lab. Solid Tumors Program, CIMA Universidad de Navarra, Pamplona, Spain
| | - Jose M Herranz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
- Hepatology Laboratory, Solid Tumors Program, CIMA-University of Navarra, Pamplona, Spain
| | - Iñigo Insausti
- Radiology Department, Clinica Universitaria de Navarra, Pamplona, Spain
| | | | - Francesco Urigo
- Hepatology: Porphyrias & Carcinogenesis Lab. Solid Tumors Program, CIMA Universidad de Navarra, Pamplona, Spain
| | - Patricia Alcaide
- Centro de Diagnóstico de Enfermedades Moleculares, Universidad Autónoma de Madrid, Madrid, Spain
| | - María Morán
- Mitochondrial Diseases Laboratory, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 12 de Octubre University Hospital, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel A Martín
- Mitochondrial Diseases Laboratory, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 12 de Octubre University Hospital, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - José Luis Lanciego
- Neurosciences Department, CIMA Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Thibaud Lefebvre
- APHP. Nord-Université de Paris Cité, Centre Français des Porphyries, Hôpital Louis Mourier, Paris, France
| | - Laurent Gouya
- APHP. Nord-Université de Paris Cité, Centre Français des Porphyries, Hôpital Louis Mourier, Paris, France
| | - Gemma Quincoces
- Translational Molecular Imaging Unit (UNIMTRA), and Nuclear Medicine-Department, Clínica Universidad de Navarra (CUN), University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Carmen Unzu
- Gene Therapy and Regulation of Gene Expression Program, CIMA Universidad de Navarra, Pamplona, Spain
| | - Sandra Hervas-Stubbs
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, CIMA-University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Oncológicas (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan M Falcón-Pérez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
- Exosomes Lab. & Metabolomics Platform. Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Technology Park, Derio, Spain
| | - Estíbaliz Alegre
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Service of Biochemistry, Clinica Universitaria de Navarra, Pamplona, Spain
| | - Azucena Aldaz
- Pharmacokinetics Division, Pharmacy Departement, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - María A Fernández-Seara
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Radiology Department, Clinica Universitaria de Navarra, Pamplona, Spain
| | - Iván Peñuelas
- Translational Molecular Imaging Unit (UNIMTRA), and Nuclear Medicine-Department, Clínica Universidad de Navarra (CUN), University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Pedro Berraondo
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, CIMA-University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Oncológicas (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
| | | | - Matias A Avila
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
- Hepatology Laboratory, Solid Tumors Program, CIMA-University of Navarra, Pamplona, Spain
- Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
| | - Antonio Fontanellas
- Hepatology: Porphyrias & Carcinogenesis Lab. Solid Tumors Program, CIMA Universidad de Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
- Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
| |
Collapse
|
11
|
Kuo N, Li P, Cunha JB, Chen L, Shavit JA, Omary MB. The Histamine Pathway is a Target to Treat Hepatic Experimental Erythropoietic Protoporphyria. Cell Mol Gastroenterol Hepatol 2025; 19:101463. [PMID: 39824305 PMCID: PMC12005319 DOI: 10.1016/j.jcmgh.2025.101463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/20/2025]
Abstract
BACKGROUND & AIMS Erythropoietic protoporphyria (EPP) is caused by mutations in ferrochelatase, which inserts iron into protoporphyrin-IX (PP-IX) to generate heme. EPP is characterized by PP-IX accumulation, skin photosensitivity, cholestasis, and end-stage liver disease. Despite available drugs that address photosensitivity, treatment of EPP-related liver disease remains an unmet need. METHODS We administered delta-aminolevulinic acid (ALA) and deferoxamine (DFO), which results in PP-IX overproduction and accumulation. High-throughput compound screening of ALA + DFO-treated zebrafish identified chlorcyclizine (first generation H1-antihistamine receptor blocker) as a drug that reduces zebrafish liver PP-IX levels. The effect of chlorcyclizine was validated in porphyrin-loaded primary mouse hepatocytes (PMHs), transgenic Fechm1Pas EPP mice, and mice fed the porphyrinogenic compound 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC). Plasma and tissue PP-IX were measured by fluorescence; livers were analyzed by histology, immunoblotting, and quantitative polymerase chain reaction. RESULTS Chlorcyclizine-treated zebrafish larvae, DDC-fed, and transgenic EPP mice manifested reduced hepatic PP-IX levels compared with controls. Histamine increased PP-IX accumulation in porphyrin-stressed hepatocytes, whereas H1/H2-receptor blockade decreased PP-IX levels. In both mouse models, chlorcyclizine lowered PP-IX levels in female but not male mice in liver, erythrocytes, and bone marrow; improved liver injury; decreased porphyrin-triggered protein aggregation and oxidation; and increased clearance of stool PP-IX. In PMHs, chlorcyclizine induced nuclear translocation of constitutive androstane and farnesoid X receptors, and transactivated bile acid transporter expression. Knockdown of the transporters BSEP and MRP4 led to increased detection of sequestosome-1 (p62 protein) high-molecular-weight species. Chlorcyclizine also reduced hepatic mast cell number and histamine level in EPP mice. CONCLUSIONS Histamine plays an important role in PP-IX accumulation in zebrafish and 2 experimental EPP models. Chlorcyclizine and/or other antihistamines provide a potential therapeutic strategy to treat EPP-associated liver disease via decreasing PP-IX accumulation.
Collapse
Affiliation(s)
- Ning Kuo
- Robert Wood Johnson Medical School, and Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey; Current affiliation: Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California
| | - Pei Li
- Robert Wood Johnson Medical School, and Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey
| | - Juliana Bragazzi Cunha
- Robert Wood Johnson Medical School, and Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey; Current affiliation: Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Lu Chen
- Robert Wood Johnson Medical School, and Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey
| | - Jordan A Shavit
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Michigan, Ann Arbor, Michigan; Department of Human Genetics, University of Michigan, Ann Arbor, Michigan
| | - M Bishr Omary
- Robert Wood Johnson Medical School, and Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey.
| |
Collapse
|
12
|
Bhasuran B, Schmolly K, Kapoor Y, Jayakumar NL, Doan R, Amin J, Meninger S, Cheng N, Deering R, Anderson K, Beaven SW, Wang B, Rudrapatna VA. Reducing diagnostic delays in acute hepatic porphyria using health records data and machine learning. J Am Med Inform Assoc 2025; 32:63-70. [PMID: 38946554 PMCID: PMC11648717 DOI: 10.1093/jamia/ocae141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/01/2024] [Accepted: 05/28/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND Acute hepatic porphyria (AHP) is a group of rare but treatable conditions associated with diagnostic delays of 15 years on average. The advent of electronic health records (EHR) data and machine learning (ML) may improve the timely recognition of rare diseases like AHP. However, prediction models can be difficult to train given the limited case numbers, unstructured EHR data, and selection biases intrinsic to healthcare delivery. We sought to train and characterize models for identifying patients with AHP. METHODS This diagnostic study used structured and notes-based EHR data from 2 centers at the University of California, UCSF (2012-2022) and UCLA (2019-2022). The data were split into 2 cohorts (referral and diagnosis) and used to develop models that predict (1) who will be referred for testing of acute porphyria, among those who presented with abdominal pain (a cardinal symptom of AHP), and (2) who will test positive, among those referred. The referral cohort consisted of 747 patients referred for testing and 99 849 contemporaneous patients who were not. The diagnosis cohort consisted of 72 confirmed AHP cases and 347 patients who tested negative. The case cohort was 81% female and 6-75 years old at the time of diagnosis. Candidate models used a range of architectures. Feature selection was semi-automated and incorporated publicly available data from knowledge graphs. Our primary outcome was the F-score on an outcome-stratified test set. RESULTS The best center-specific referral models achieved an F-score of 86%-91%. The best diagnosis model achieved an F-score of 92%. To further test our model, we contacted 372 current patients who lack an AHP diagnosis but were predicted by our models as potentially having it (≥10% probability of referral, ≥50% of testing positive). However, we were only able to recruit 10 of these patients for biochemical testing, all of whom were negative. Nonetheless, post hoc evaluations suggested that these models could identify 71% of cases earlier than their diagnosis date, saving 1.2 years. CONCLUSIONS ML can reduce diagnostic delays in AHP and other rare diseases. Robust recruitment strategies and multicenter coordination will be needed to validate these models before they can be deployed.
Collapse
Affiliation(s)
- Balu Bhasuran
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Katharina Schmolly
- David Geffen School of Medicine & Pfleger Liver Institute, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Yuvraaj Kapoor
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Nanditha Lakshmi Jayakumar
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Raymond Doan
- Alnylam Pharmaceuticals, Cambridge, MA 02142, United States
| | - Jigar Amin
- Alnylam Pharmaceuticals, Cambridge, MA 02142, United States
| | | | - Nathan Cheng
- Alnylam Pharmaceuticals, Cambridge, MA 02142, United States
| | - Robert Deering
- Alnylam Pharmaceuticals, Cambridge, MA 02142, United States
| | - Karl Anderson
- Division of Gastroenterology and Hepatology, University of Texas Medical Branch, School of Medicine, Galveston, TX 77555, United States
| | - Simon W Beaven
- David Geffen School of Medicine & Pfleger Liver Institute, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Bruce Wang
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Vivek A Rudrapatna
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143, United States
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, United States
| |
Collapse
|
13
|
Zhu J, Qin FY, Lei S, Gu R, Qi Q, Lu J, Anderson KE, Wipf P, Ma X. Inhibition of ABCG2 prevents phototoxicity in a mouse model of erythropoietic protoporphyria. Nat Commun 2024; 15:10557. [PMID: 39632884 PMCID: PMC11618456 DOI: 10.1038/s41467-024-54969-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Erythropoietic protoporphyria (EPP) is a genetic disease characterized by protoporphyrin IX-mediated painful phototoxicity. Currently, options for the management of EPP-associated phototoxicity are limited and no oral medication is available. Here, we investigated a novel therapy against EPP-associated phototoxicity by targeting the ATP-binding cassette subfamily G member 2 (ABCG2), the efflux transporter of protoporphyrin IX. Oral ABCG2 inhibitors were developed, and they successfully prevented EPP-associated phototoxicity in a genetically engineered EPP mouse model. Mechanistically, ABCG2 inhibitors suppress protoporphyrin IX release from erythroid cells and reduce the systemic exposure to protoporphyrin IX in EPP. In summary, our work establishes a novel strategy for EPP therapy by targeting ABCG2 and provides oral ABCG2 inhibitors that can effectively prevent protoporphyrin IX-mediated phototoxicity in mice.
Collapse
MESH Headings
- Animals
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- Protoporphyria, Erythropoietic/metabolism
- Protoporphyria, Erythropoietic/genetics
- Protoporphyrins/metabolism
- Protoporphyrins/pharmacology
- Mice
- Disease Models, Animal
- Humans
- Erythroid Cells/metabolism
- Erythroid Cells/drug effects
- Erythroid Cells/pathology
- Dermatitis, Phototoxic/metabolism
- Dermatitis, Phototoxic/prevention & control
- Male
- Female
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Junjie Zhu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fu-Ying Qin
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Saifei Lei
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ruizhi Gu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qian Qi
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jie Lu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karl E Anderson
- Porphyria Laboratory & Center, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
14
|
Kuo HC, Ro LS, Lin CN, Chu CC, Liao MF, Chang HS. Baseline urinary ALA and PBG as criteria for starting pharmacologic prophylactic treatment in acute intermittent porphyria treated with givosiran. Mol Genet Metab Rep 2024; 41:101169. [PMID: 39720736 PMCID: PMC11667067 DOI: 10.1016/j.ymgmr.2024.101169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/26/2024] Open
Abstract
Introduction For patients with acute intermittent porphyria (AIP), a true attack could be difficult to distinguish from chronic abdominal pain. This study focused on treatment responses from two patients with confirmed elevated biochemical data (delta-aminolevulinic acid (ALA), porphobilinogen (PBG)) and clinical evidence for acute attacks before starting givosiran. Methods Data from patients who participated in the phase III givosiran trial in Taiwan between May 2018 and May 2021 were reviewed. The pre-trial and post-trial biochemical data (urinary ALA/PBG), annualized attack rate (AAR), for two participants were obtained from our hospital record. Results Two patients had detailed records of biochemical evidence of acute attacks pre-trial (ALA:11.66-79.8 mg/24-h urine collection, PBG:75.45-160.11 mg/24-h). Patient Pb/Gn#1 with a disease duration of 1.6-years, had zero AAR during givosiran treatment. Patient Pb/Gn#2 had received prior hemin prophylaxis, had AIP for 6.7-years, had an AAR of 17.0 before givosiran, and an AAR of 12 at the post-trial compassionate-use period. The change in SF-12 PCS score from baseline was marginally clinical-meaningful (2.8 for Patient Pb/Gn#1 and 2.0 for Patient Pb/Gn#2). Conclusion Our data from 2 AIP patients with biochemical and clinical evidence of acute attacks suggested that patient with a shorter disease duration may respond better in terms of AAR. Further studies are necessary to understand the association between disease characteristics, treatment history, and optimal treatment response for patients with recurrent AIP in terms of both attack frequency and quality of life.
Collapse
Affiliation(s)
- Hung-Chou Kuo
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Long-Sun Ro
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Ni Lin
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Che Chu
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Feng Liao
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hong-Shiu Chang
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
15
|
Rudnick SB, West NE, Fong K, Beaven SW, VanderVeen NT. Acute Intermittent Porphyria in an Adolescent Patient: Diagnostic and Treatment Challenges. Cureus 2024; 16:e74784. [PMID: 39737264 PMCID: PMC11682870 DOI: 10.7759/cureus.74784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
Acute intermittent porphyria (AIP) is a rare inherited metabolic disorder caused by decreased activity of the enzyme porphobilinogen deaminase in the heme synthesis pathway. This leads to the accumulation of toxic porphyrin precursors, such as porphobilinogen and δ-aminolevulinic acid. Clinical manifestations typically include episodic bouts of severe neurovisceral pain and autonomic dysfunction. These events have various triggers, including fasting, dehydration, hormonal fluctuations, and certain medications, while treatment involves dextrose infusions for mild attacks and intravenous hemin for severe cases. This case highlights a 17-year-old female patient with numerous prior presentations to the healthcare system for intense bouts of abdominal pain, vomiting, and seizure-like activity who was ultimately diagnosed with AIP. Her course was complicated by diagnostic delays and challenges managing severe refractory pain requiring prolonged courses of hemin in addition to a multimodal pain plan. This case highlights the diagnostic complexities and treatment challenges that patients with AIP face when navigating this challenging clinical syndrome and identifies an opportunity for increased awareness to guide the testing and treatment for AIP.
Collapse
Affiliation(s)
- Samuel B Rudnick
- Pediatrics, University of California Los Angeles, Los Angeles, USA
| | - Nicole E West
- Pediatrics, University of California Los Angeles, Los Angeles, USA
| | - Kelly Fong
- Pediatrics, University of California Los Angeles, Los Angeles, USA
| | - Simon W Beaven
- Internal Medicine, University of California Los Angeles Vatche & Tamar Manoukian Division of Digestive Diseases, Los Angeles, USA
- Internal Medicine, University of California Los Angeles David Geffen School of Medicine, Los Angeles, USA
- Internal Medicine, University of California Los Angeles Pfleger Liver Institute, Los Angeles, USA
| | - Nathan T VanderVeen
- Internal Medicine-Pediatrics, University of California Los Angeles, Los Angeles, USA
| |
Collapse
|
16
|
Qi Q, Gu R, Zhu J, Anderson KE, Ma X. Roles of the ABCG2 Transporter in Protoporphyrin IX Distribution and Toxicity. Drug Metab Dispos 2024; 52:1201-1207. [PMID: 38351044 PMCID: PMC11495668 DOI: 10.1124/dmd.123.001582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/08/2024] [Indexed: 10/18/2024] Open
Abstract
ATP-binding cassette transporter subfamily G member 2 (ABCG2) is a membrane-bound transporter responsible for the efflux of various xenobiotics and endobiotics, including protoporphyrin IX (PPIX), an intermediate in the heme biosynthesis pathway. Certain genetic mutations and chemicals impair the conversion of PPIX to heme and/or increase PPIX production, leading to PPIX accumulation and toxicity. In mice, deficiency of ABCG2 protects against PPIX-mediated phototoxicity and hepatotoxicity by modulating PPIX distribution. In addition, in vitro studies revealed that ABCG2 inhibition increases the efficacy of PPIX-based photodynamic therapy by retaining PPIX inside target cells. In this review, we discuss the roles of ABCG2 in modulating the tissue distribution of PPIX, PPIX-mediated toxicity, and PPIX-based photodynamic therapy. SIGNIFICANCE STATEMENT: This review summarized the roles of ABCG2 in modulating PPIX distribution and highlighted the therapeutic potential of ABCG2 inhibitors for the management of PPIX-mediated toxicity.
Collapse
Affiliation(s)
- Qian Qi
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Ruizhi Gu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Junjie Zhu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Karl E Anderson
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| |
Collapse
|
17
|
Wijerathna HMSM, Shanaka KASN, Raguvaran SS, Jayamali BPMV, Kim SH, Kim MJ, Jung S, Lee J. CRISPR/Cas9-Mediated fech Knockout Zebrafish: Unraveling the Pathogenesis of Erythropoietic Protoporphyria and Facilitating Drug Screening. Int J Mol Sci 2024; 25:10819. [PMID: 39409147 PMCID: PMC11476521 DOI: 10.3390/ijms251910819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Erythropoietic protoporphyria (EPP1) results in painful photosensitivity and severe liver damage in humans due to the accumulation of fluorescent protoporphyrin IX (PPIX). While zebrafish (Danio rerio) models for porphyria exist, the utility of ferrochelatase (fech) knockout zebrafish, which exhibit EPP, for therapeutic screening and biological studies remains unexplored. This study investigated the use of clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-mediated fech-knockout zebrafish larvae as a model of EPP1 for drug screening. CRISPR/Cas9 was employed to generate fech-knockout zebrafish larvae exhibiting morphological defects without lethality prior to 9 days post-fertilization (dpf). To assess the suitability of this model for drug screening, ursodeoxycholic acid (UDCA), a common treatment for cholestatic liver disease, was employed. This treatment significantly reduced PPIX fluorescence and enhanced bile-secretion-related gene expression (abcb11a and abcc2), indicating the release of PPIX. Acridine orange staining and quantitative reverse transcription polymerase chain reaction analysis of the bax/bcl2 ratio revealed apoptosis in fech-/- larvae, and this was reduced by UDCA treatment, indicating suppression of the intrinsic apoptosis pathway. Neutral red and Sudan black staining revealed increased macrophage and neutrophil production, potentially in response to PPIX-induced cell damage. UDCA treatment effectively reduced macrophage and neutrophil production, suggesting its potential to alleviate cell damage and liver injury in EPP1. In conclusion, CRISPR/Cas9-mediated fech-/- zebrafish larvae represent a promising model for screening drugs against EPP1.
Collapse
Affiliation(s)
- Hitihami M. S. M. Wijerathna
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju 63243, Republic of Korea; (H.M.S.M.W.)
- Department of Aquaculture and Seafood Technology, Faculty of Fisheries and Ocean Sciences, Ocean University of Sri Lanka, Colombo 01500, Sri Lanka
| | - Kateepe A. S. N. Shanaka
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju 63243, Republic of Korea; (H.M.S.M.W.)
- Marine Life Research Institute, Jeju National University, Jeju 63333, Republic of Korea
| | - Sarithaa S. Raguvaran
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju 63243, Republic of Korea; (H.M.S.M.W.)
- Marine Life Research Institute, Jeju National University, Jeju 63333, Republic of Korea
| | - Bulumulle P. M. V. Jayamali
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju 63243, Republic of Korea; (H.M.S.M.W.)
| | - Seok-Hyung Kim
- Marine Life Research Institute, Jeju National University, Jeju 63333, Republic of Korea
| | - Myoung-Jin Kim
- Nakdonggang National Institute of Biological Resources, Sangju-si 37242, Republic of Korea
| | - Sumi Jung
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju 63243, Republic of Korea; (H.M.S.M.W.)
- Marine Life Research Institute, Jeju National University, Jeju 63333, Republic of Korea
- Marine Molecular Genetics Lab, Jeju National University, 102 Jejudaehakno, Jeju 63243, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju 63243, Republic of Korea; (H.M.S.M.W.)
- Marine Life Research Institute, Jeju National University, Jeju 63333, Republic of Korea
- Marine Molecular Genetics Lab, Jeju National University, 102 Jejudaehakno, Jeju 63243, Republic of Korea
| |
Collapse
|
18
|
Sardh E, Balwani M, Rees DC, Anderson KE, Jia G, Sweetser MT, Wang B. Long-term follow-up of givosiran treatment in patients with acute intermittent porphyria from a phase 1/2, 48-month open-label extension study. Orphanet J Rare Dis 2024; 19:365. [PMID: 39363243 PMCID: PMC11448181 DOI: 10.1186/s13023-024-03284-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/03/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Acute hepatic porphyria is a group of multisystem disorders of which acute intermittent porphyria is the most common subtype. Givosiran, a subcutaneously administered RNA interference therapeutic targeting liver ALAS mRNA, is approved for treating these disorders. This Phase 1/2 open-label extension study (NCT02949830) evaluated the long-term safety and efficacy of givosiran in adults with acute intermittent porphyria, with follow-up of up to 48 months, which is the longest follow-up of givosiran treatment to date. Participants were adults aged 18-65 years who completed part C of the Phase 1 givosiran study (NCT2452372). METHODS Enrollees received givosiran for up to 48 months. Primary and secondary endpoints included the incidence of adverse events, changes in urinary delta-aminolevulinic acid (ALA) and porphobilinogen (PBG) levels, annualized rate of porphyria attacks, and annualized hemin use. Quality of life was assessed using the EQ-5D-5L instrument as an exploratory endpoint. RESULTS Sixteen patients (median age: 39.5 years) participated. Common adverse events included abdominal pain, nasopharyngitis, and nausea (50% each), with injection-site erythema (38%) and injection-site pruritus (25%) noted as frequent treatment-related reactions. Givosiran therapy reduced annualized rates of porphyria attacks and hemin use by 97% and 96%, respectively. From months > 33 to 48, all patients were free from attacks requiring significant medical intervention and did not use hemin. There were substantial reductions in median urinary ALA and PBG of 95% and 98%, respectively. Additionally, a clinically meaningful improvement in quality of life was observed. CONCLUSIONS In the longest follow-up of givosiran-treated patients reported to date, the therapy maintained an acceptable safety profile and demonstrated sustained improvements in clinical outcomes over 4 years in patients with acute intermittent porphyria.
Collapse
Affiliation(s)
- Eliane Sardh
- CMMS - Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Karolinska Institutet, Solna, 171 64, Stockholm, Sweden.
| | - Manisha Balwani
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, New York, NY, USA
| | - David C Rees
- Comprehensive Cancer Centre, King's College Hospital, London, UK
| | - Karl E Anderson
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Texas Medical Branch, Galveston, TX, USA
| | - Gang Jia
- Medical Affairs Statistics, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | | | - Bruce Wang
- Department of Medicine and UCSF Liver Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
19
|
Vadivel VB, Lamba S. Acute Intermittent Porphyria: A Diagnostic Conundrum. Cureus 2024; 16:e72419. [PMID: 39463915 PMCID: PMC11512492 DOI: 10.7759/cureus.72419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2024] [Indexed: 10/29/2024] Open
Abstract
This case report presents the case of a 20-year-old female patient who sought emergency medical attention for severe abdominal pain, nausea and vomiting, tachycardia, hypertension, and discolored urine. Initial diagnostic evaluations yielded no significant abnormalities; however, subsequent analysis revealed elevated urinary porphobilinogen, corroborating a diagnosis of acute intermittent porphyria (AIP). The patient's medical history included recurrent urinary tract infections and a prior episode of syndrome of inappropriate antidiuretic hormone secretion (SIADH), in conjunction with psychiatric comorbidities of anxiety and depression. Management encompassed a multifaceted approach involving supportive therapies, such as hydration and analgesia, alongside the imperative to abstain from using contraindicated pharmacological agents. Following referral to the National Acute Porphyria Service (NAPS), the patient received intravenous Haem arginate, resulting in clinical improvement and subsequent discharge. Nonetheless, she later necessitated further hospitalization due to the recurrence of similar symptoms. This case highlights the exigency of recognizing AIP in young women presenting with nonspecific symptoms, necessitating a high index of clinical suspicion. Furthermore, it accentuates the critical importance of early specialist intervention to avert severe sequelae associated with acute episodes. The integration of targeted educational initiatives within Acute Medicine departments is paramount for fostering awareness and facilitating prompt diagnosis and management of this rare yet significant disorder.
Collapse
Affiliation(s)
- Vijay Balaji Vadivel
- Acute Medicine, Cumberland Infirmary, North Cumbria Integrated Care, NHS Trust, Carlisle, GBR
| | - Simran Lamba
- Internal Medicine, ABLE Charitable Hospital, Bahrola, IND
| |
Collapse
|
20
|
Guida CC, Nardella M, del Mar YS Perez A, Savino M, Ferrara G, Napolitano F, Crisetti A, Aucella F, Aucella F. Effectiveness and tolerability of givosiran for the management of acute hepatic porphyria: A monocenter real-life evaluation. Mol Genet Metab Rep 2024; 40:101111. [PMID: 39027010 PMCID: PMC11254832 DOI: 10.1016/j.ymgmr.2024.101111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
Acute hepatic porphyrias (AHPs) are a family of rare, autosomal, dominantly inherited conditions characterized by abnormalities in the production of heme. Advances in molecular engineering have provided new therapeutic possibilities for modifying the heme synthetic pathway in patients with porphyria. In particular, the RNA interference therapeutic givosiran was approved for the treatment of adults and adolescents with AHP aged >12 years based on the positive results of the phase III trial ENVISION. Despite the extended characterization of the activity of givosiran in clinical trials, reports on the long-term effects and effectiveness of the treatment in clinical practice are still scant. To fill this gap, this case series describes a monocentric Italian cohort of AHP patients treated with givosiran. Overall, our real-life experience supports the clinical evidence that long-term treatment with givosiran is well tolerated and able to provide sustained and continuous benefit to patients with acute intermittent porphyria, as reflected by the reduction in the frequency of attacks. In our series, givosiran treatment was also associated with improvement in assessments of quality of life, pain and fatigue.
Collapse
Affiliation(s)
- Claudio Carmine Guida
- Scientific Institut for Research and Health Care, Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Maria Nardella
- Scientific Institut for Research and Health Care, Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Aurora del Mar YS Perez
- Scientific Institut for Research and Health Care, Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Maria Savino
- Scientific Institut for Research and Health Care, Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Gaetano Ferrara
- Scientific Institut for Research and Health Care, Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Francesco Napolitano
- Scientific Institut for Research and Health Care, Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Annalisa Crisetti
- Scientific Institut for Research and Health Care, Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Francesco Aucella
- Scientific Institut for Research and Health Care, Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Filippo Aucella
- Scientific Institut for Research and Health Care, Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| |
Collapse
|
21
|
Jericó D, Córdoba KM, Urigo F, Enríquez de Salamanca R, Anderson KE, Deybach JC, Ávila MA, Fontanellas A. Exploring current and emerging therapies for porphyrias. Liver Int 2024; 44:2174-2190. [PMID: 38813953 DOI: 10.1111/liv.15979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/31/2024]
Abstract
Porphyrias are rare, mostly inherited disorders resulting from altered activity of specific enzymes in the haem synthesis pathway that lead to accumulation of pathway intermediates. Photocutaneous symptoms occur when excess amounts of photoreactive porphyrins circulate in the blood to the skin, whereas increases in potentially neurotoxic porphyrin precursors are associated with neurovisceral symptoms. Current therapies are suboptimal and their mechanisms are not well established. As described here, emerging therapies address underlying disease mechanisms by introducing a gene, RNA or other specific molecule with the potential to cure or slow progression of the disease. Recent progress in nanotechnology and nanoscience, particularly regarding particle design and formulation, is expanding disease targets. More secure and efficient drug delivery systems have extended our toolbox for transferring specific molecules, especially into hepatocytes, and led to proof-of-concept studies in animal models. Repurposing existing drugs as molecular chaperones or haem synthesis inhibitors is also promising. This review summarizes key examples of these emerging therapeutic approaches and their application for hepatic and erythropoietic porphyrias.
Collapse
Affiliation(s)
- Daniel Jericó
- Solid Tumors Program, Hepatology: Porphyrias & Carcinogenesis Laboratory, CIMA-University of Navarra, Pamplona, Spain
| | - Karol M Córdoba
- Solid Tumors Program, Hepatology: Porphyrias & Carcinogenesis Laboratory, CIMA-University of Navarra, Pamplona, Spain
| | - Francesco Urigo
- Solid Tumors Program, Hepatology: Porphyrias & Carcinogenesis Laboratory, CIMA-University of Navarra, Pamplona, Spain
| | - Rafael Enríquez de Salamanca
- Department of Internal Medicine, Reference Center for Inherited Metabolic Disease-MetabERN, University Hospital 12 de Octubre, UCM, Madrid, Spain
| | - Karl E Anderson
- Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jean-Charles Deybach
- French Porphyria Reference Center (CRMR Porphyries France), Université Paris, Paris, France
| | - Matías A Ávila
- Solid Tumors Program, Hepatology: Porphyrias & Carcinogenesis Laboratory, CIMA-University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Fontanellas
- Solid Tumors Program, Hepatology: Porphyrias & Carcinogenesis Laboratory, CIMA-University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
22
|
Ali M, Iqbal S. Human Chorionic Gonadotropin (hCG) Injections Exacerbating Acute Intermittent Porphyria in a 34-Year-Old Woman. Cureus 2024; 16:e68651. [PMID: 39371821 PMCID: PMC11452356 DOI: 10.7759/cureus.68651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
A 34-year-old Asian woman arrived at the emergency department (ED) with complaints of sharp, cramping abdominal pain followed by stabbing chest pain that radiated to her back. She also reported numbness, tingling in both hands and feet, and a burning sensation. Upon examination, she exhibited tachycardia and persistently elevated blood pressure. Her lab results revealed low potassium and sodium levels. Despite testing negative for pregnancy, normal hepatic function test, and nerve conduction study, her symptoms persisted, and she was admitted to the intensive care unit for the monitoring and correction of her electrolyte imbalances. She was later discharged but continued to experience symptoms, prompting multiple ED visits. The patient reported the symptoms following a calorie-restricted ketogenic diet and receiving human chorionic gonadotropin (hCG) injections for weight loss, which led the providers to initially diagnose her with "keto flu" due to inadequate nutrient intake. Despite receiving this diagnosis, her symptoms worsened, and she experienced pain throughout her whole body, along with muscle pain and abnormal sensations. Further assessment revealed that her urine was brown and showed abnormal levels of porphyrins, indicating acute intermittent porphyria. A genetic test confirmed the presence of a pathogenic mutation in hydroxymethylbilane synthase (HMBS), leading to a diagnosis of late-onset acute intermittent porphyria.
Collapse
Affiliation(s)
- Mahesheema Ali
- Pathology and Laboratory Medicine, MetroHealth System, Case Western Reserve University School of Medicine, Cleveland, USA
| | - Sahar Iqbal
- Pathology, MetroHealth Medical Center, Cleveland, USA
| |
Collapse
|
23
|
Poli A, Schmitt C, Puy H, Talbi N, Lefebvre T, Gouya L. Erythropoietic protoporphyrias: updates and advances. Trends Mol Med 2024; 30:863-874. [PMID: 38890030 DOI: 10.1016/j.molmed.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024]
Abstract
Protoporphyrias are caused by pathogenic variants in genes encoding enzymes involved in heme biosynthesis. They induce the accumulation of a hydrophobic phototoxic compound, protoporphyrin (PPIX), in red blood cells (RBCs). PPIX is responsible for painful cutaneous photosensitivity, which severely impairs quality of life. Hepatic elimination of PPIX increases the risk of cholestatic liver disease, requiring lifelong monitoring. Treatment options are scarce and mainly limited to supportive care such as protection from visible light. Here, we review the pathophysiology of protoporphyrias, their diagnosis, and current recommendations for medical care. We discuss new therapeutic strategies, some of which are currently undergoing clinical trials and are likely to radically alter the severity of the disease in the years to come.
Collapse
Affiliation(s)
- Antoine Poli
- Institut National de la Santé et de la Recherche Médicale U1149, Centre de Recherche sur l'Inflammation, Paris, France; Université Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Centre de Référence Maladies Rares Porphyries, Hôpital Louis Mourier, Colombes, France; Laboratory of excellence Gr-Ex, Paris, France.
| | - Caroline Schmitt
- Institut National de la Santé et de la Recherche Médicale U1149, Centre de Recherche sur l'Inflammation, Paris, France; Université Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Centre de Référence Maladies Rares Porphyries, Hôpital Louis Mourier, Colombes, France; Laboratory of excellence Gr-Ex, Paris, France
| | - Hervé Puy
- Institut National de la Santé et de la Recherche Médicale U1149, Centre de Recherche sur l'Inflammation, Paris, France; Université Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Centre de Référence Maladies Rares Porphyries, Hôpital Louis Mourier, Colombes, France; Laboratory of excellence Gr-Ex, Paris, France
| | - Neila Talbi
- Institut National de la Santé et de la Recherche Médicale U1149, Centre de Recherche sur l'Inflammation, Paris, France
| | - Thibaud Lefebvre
- Institut National de la Santé et de la Recherche Médicale U1149, Centre de Recherche sur l'Inflammation, Paris, France
| | - Laurent Gouya
- Institut National de la Santé et de la Recherche Médicale U1149, Centre de Recherche sur l'Inflammation, Paris, France; Université Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Centre de Référence Maladies Rares Porphyries, Hôpital Louis Mourier, Colombes, France; Laboratory of excellence Gr-Ex, Paris, France
| |
Collapse
|
24
|
Hwang J, Park A, Kim C, Kim CG, Kwak J, Kim B, Shin H, Ku M, Yang J, Baek A, Choi J, Lim H, No KT, Zhao X, Choi U, Kim TI, Jeong KS, Lee H, Shin SJ. Inhibition of IRP2-dependent reprogramming of iron metabolism suppresses tumor growth in colorectal cancer. Cell Commun Signal 2024; 22:412. [PMID: 39180081 PMCID: PMC11342626 DOI: 10.1186/s12964-024-01769-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/27/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Dysregulation of iron metabolism is implicated in malignant transformation, cancer progression, and therapeutic resistance. Here, we demonstrate that iron regulatory protein 2 (IRP2) preferentially regulates iron metabolism and promotes tumor growth in colorectal cancer (CRC). METHODS IRP2 knockdown and knockout cells were generated using RNA interference and clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 methodologies, respectively. Cell viability was evaluated using both CCK-8 assay and cell counting techniques. Furthermore, IRP2 inhibition was determined by surface plasmon resonance (SPR) and RNA immunoprecipitation (IP). The suppressive effects of IRP2 were also corroborated in both organoid and mouse xenograft models, providing a comprehensive validation of IRP2's role. RESULTS We have elucidated the role of IRP2 as a preferential regulator of iron metabolism, actively promoting tumorigenesis within CRC. Elevated levels of IRP2 expression in patient samples are correlated with diminished overall survival, thereby reinforcing its potential role as a prognostic biomarker. The functional suppression of IRP2 resulted in a pronounced delay in tumor growth. Building on this proof of concept, we have developed IRP2 inhibitors that significantly reduce IRP2 expression and hinder its interaction with iron-responsive elements in key iron-regulating proteins, such as ferritin heavy chain 1 (FTH1) and transferrin receptor (TFRC), culminating in iron depletion and a marked reduction in CRC cell proliferation. Furthermore, these inhibitors are shown to activate the AMPK-ULK1-Beclin1 signaling cascade, leading to cell death in CRC models. CONCLUSIONS Collectively, these findings highlight the therapeutic potential of targeting IRP2 to exploit the disruption of iron metabolism in CRC, presenting a strategic advancement in addressing a critical area of unmet clinical need.
Collapse
Affiliation(s)
- Jieon Hwang
- Department of Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Areum Park
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea
- Department of Chemistry, Yonsei University, Seoul, 03722, Korea
| | - Chinwoo Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Chang Gon Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Jaesung Kwak
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea
| | - Byungil Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea
| | - Hyunjin Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea
| | - Minhee Ku
- Department of Radiology, Yonsei University College of Medicine, Seoul, 03722, Korea
- Convergence Research Center for Systems Molecular Radiological Science, Yonsei University, Seoul, 03722, Korea
| | - Jaemoon Yang
- Department of Radiology, Yonsei University College of Medicine, Seoul, 03722, Korea
- Convergence Research Center for Systems Molecular Radiological Science, Yonsei University, Seoul, 03722, Korea
| | - Ayoung Baek
- Bioinformatics and Molecular Design Research Center, Incheon, 21983, Korea
| | - Jiwon Choi
- College of Pharmacy, Dongduk Women's University, Seoul, 02748, Korea
| | - Hocheol Lim
- Bioinformatics and Molecular Design Research Center, Incheon, 21983, Korea
| | - Kyoung Tai No
- Bioinformatics and Molecular Design Research Center, Incheon, 21983, Korea
- The Interdisciplinary Graduate Program in Integrative Biotechnology & Translational Medicine, Yonsei Unversity, Incheon, 21983, Korea
| | - Xianghua Zhao
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Uyeong Choi
- Department of Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Tae Il Kim
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Kyu-Sung Jeong
- Department of Chemistry, Yonsei University, Seoul, 03722, Korea
| | - Hyuk Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea.
| | - Sang Joon Shin
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Korea.
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 03722, Korea.
- Convergence Research Center for Systems Molecular Radiological Science, Yonsei University, Seoul, 03722, Korea.
| |
Collapse
|
25
|
Lei JJ, Li S, Dong BX, Yang J, Ren Y. Acute intermittent porphyria: a disease with low penetrance and high heterogeneity. Front Genet 2024; 15:1374965. [PMID: 39188285 PMCID: PMC11345236 DOI: 10.3389/fgene.2024.1374965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
Acute intermittent porphyria (AIP) is caused by mutations in the gene encoding hydroxymethylbilane synthase (HMBS), a key enzyme in the heme biosynthesis pathway. AIP is an autosomal dominant disorder characterized by low penetrance and a highly heterogenous clinical presentation. The estimated prevalence of AIP is 5-10 cases per 100,000 persons, with acute attacks manifesting in less than 1% of the at-risk population. This low frequency of attacks suggests significant roles for oligogenic inheritance and environmental factors in the pathogenesis of the disease. In recent years, identification of several modifier genes has advanced our understanding of the factors influencing AIP penetrance and disease severity. This review summarizes these factors including the impact of specific HMBS mutations, oligogenic inheritance, mitochondrial DNA copy number, age, sex, the influence of sex hormones, and the role of environmental factors. Further studies into the etiology of AIP disease penetrance should inform pathogenesis, potentially allowing for the development of more precise diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Jia-Jia Lei
- Department of First Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Shuang Li
- Department of First Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Bai-Xue Dong
- Department of First Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Jing Yang
- Department of Endocrinology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yi Ren
- Department of Endocrinology, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
26
|
Adapa SR, Meshram P, Sami A, Jiang RHY. Harnessing Porphyrin Accumulation in Liver Cancer: Combining Genomic Data and Drug Targeting. Biomolecules 2024; 14:959. [PMID: 39199347 PMCID: PMC11352895 DOI: 10.3390/biom14080959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
The liver, a pivotal organ in human metabolism, serves as a primary site for heme biosynthesis, alongside bone marrow. Maintaining precise control over heme production is paramount in healthy livers to meet high metabolic demands while averting potential toxicity from intermediate metabolites, notably protoporphyrin IX. Intriguingly, our recent research uncovers a disrupted heme biosynthesis process termed 'porphyrin overdrive' in cancers that fosters the accumulation of heme intermediates, potentially bolstering tumor survival. Here, we investigate heme and porphyrin metabolism in both healthy and oncogenic human livers, utilizing primary human liver transcriptomics and single-cell RNA sequencing (scRNAseq). Our investigations unveil robust gene expression patterns in heme biosynthesis in healthy livers, supporting electron transport chain (ETC) and cytochrome P450 function without intermediate accumulation. Conversely, liver cancers exhibit rewired heme biosynthesis and a massive downregulation of cytochrome P450 gene expression. Notably, despite diminished drug metabolism, gene expression analysis shows that heme supply to the ETC remains largely unaltered or even elevated with patient cancer progression, suggesting a metabolic priority shift. Liver cancers selectively accumulate intermediates, which are absent in normal tissues, implicating their role in disease advancement as inferred by expression analysis. Furthermore, our findings in genomics establish a link between the aberrant gene expression of porphyrin metabolism and inferior overall survival in aggressive cancers, indicating potential targets for clinical therapy development. We provide in vitro proof-of-concept data on targeting porphyrin overdrive with a drug synergy strategy.
Collapse
Affiliation(s)
- Swamy R. Adapa
- USF Genomics Program, Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA;
- Global and Planetary Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA;
| | - Pravin Meshram
- Global and Planetary Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA;
| | - Abdus Sami
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Rays H. Y. Jiang
- USF Genomics Program, Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA;
- Global and Planetary Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA;
| |
Collapse
|
27
|
M K, M R, J B, A BI, K IP, R W, E O, A ZK, R S, R P. Neurodevelopmental disorder in a patient with HMBS and SCN3A variants-A possibly blended phenotype further delineating autosomal recessive HMBS related disease. Am J Med Genet A 2024; 194:e63617. [PMID: 38568055 DOI: 10.1002/ajmg.a.63617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 03/04/2024] [Accepted: 03/22/2024] [Indexed: 07/05/2024]
Abstract
Monoallelic pathogenic HMBS variants are a well-established cause of acute intermittent porphyria (AIP), whereas biallelic pathogenic variants may cause HMBS-related leukoencephalopathy which remains a poorly characterized disorder. We describe an 8-year-old girl with hypotonia, hearing impairment, horizontal nystagmus, bilateral strabismus, impaired visual acuity, and optic nerve atrophy. She had no epilepsy but sleep electroencephalogram showed paroxysmal changes in the right hemisphere with secondary generalizations. Brain magnetic resonance imaging was unremarkable apart from a few small white matter hyperintensities. Exome sequencing (ES) initially prioritized a SCN3A c.3822G>A de novo variant whose sole causative role was eventually questioned as not fully compatible with symptoms. ES reanalysis revealed a homozygous c.674G>A HMBS variant. In the monoallelic form this variant is a known cause of AIP, whereas in trans with another HMBS pathogenic variant it was associated with the HMBS-related leukoencephalopathy in four individuals. Despite lack of signs/symptoms of porphyria, literature analysis suggested that HMBS c.674G>A likely contributed to the disease either as the sole cause or together with SCN3A c.3822G>A as a part of blended phenotype. Our report adds to the relatively small number of described cases of HMBS-related leukoencephalopathy and emphasizes that autosomal recessive form of HMBS disease can be present in the absence of porphyria symptoms.
Collapse
Affiliation(s)
- Kłaniewska M
- Department of Family and Pediatric Nursing, Wroclaw Medical University, Wroclaw, Poland
| | - Rydzanicz M
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Bladowska J
- Department of Radiology, Wroclaw 4th Military Clinical Hospital, Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Borys-Iwanicka A
- Department of Paediatrics, Gastroenterology and Nutrition, Wroclaw Medical University, Wroclaw, Poland
| | - Iwanicka-Pronicka K
- Department of Medical Genetics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Wasilewski R
- Department of Disorders of Hemostasis and Internal Medicine, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Odnoczko E
- Laboratory of Genetics in Hemostasis and Porphyria, Department of Hemostasis and Metabolic Disorders, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Zubkiewicz-Kucharska A
- Department of Pediatrics, Endocrinology, Diabetology and Metabolic Diseases, Medical University of Wroclaw, Wroclaw, Poland
| | - Smigiel R
- Department of Pediatrics, Endocrinology, Diabetology and Metabolic Diseases, Medical University of Wroclaw, Wroclaw, Poland
| | - Ploski R
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
28
|
Wang Q, Zhuang JL, Han B, Chen M, Zhao B. Drug-associated porphyria: a pharmacovigilance study. Orphanet J Rare Dis 2024; 19:286. [PMID: 39090656 PMCID: PMC11295309 DOI: 10.1186/s13023-024-03294-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The potentially fatal attacks experienced by porphyria carriers are triggered by various porphyrinogenic drugs. However, determining the safety of particular drugs is challenging. METHODS We retrospectively used the U.S. Food and Drug Administration's Adverse Event Reporting System (FAERS) to identify drugs associated with porphyria as an adverse event (AE) extracted from data from January 2004 to March 2022. The associated search terms included "Porphyria," "Porphyria screen," "Porphyria non-acute," "Porphyria acute," "Acquired porphyria," and "Pseudoporphyria." Signal mining analysis was performed to identify the association between drugs and AEs by four algorithms, namely the reporting odds ratio, proportional reporting ratio, Bayesian confidence propagation neural network, and multi-item gamma Poisson shrinker. RESULTS FAERS reported 1470 cases of porphyria-related AEs, and 406 drugs were screened after combining trade and generic names. All four algorithms identified 52 drugs with signals. The characteristics of all the reports and signaling drugs were analyzed. CONCLUSIONS This is the first report of drug-associated porphyria that provides critical information on drug porphyrogenicity, facilitating rational and evidence-based drug prescription and improving the accuracy of porphyrogenicity prediction based on model algorithms. Moreover, this study serves a reference for clinicians to ensure that porphyrinogenic drugs are not prescribed to carriers of porphyria genetic mutations.
Collapse
Affiliation(s)
- Qi Wang
- Department of Hematology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Ling Zhuang
- Department of Hematology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing Han
- Department of Hematology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miao Chen
- Department of Hematology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Bin Zhao
- Department of Pharmacy, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
29
|
Gallagher CJ, Bentley LA, Challenger R, Jones M, Schulenburg-Brand D. Stability of porphyrins and porphyrin precursors in urine and plasma samples: implications for sample handling and storage. J Clin Pathol 2024; 77:574-578. [PMID: 38631910 DOI: 10.1136/jcp-2024-209443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024]
Abstract
The porphyrias are rare disorders of haem biosynthesis. Diagnosis requires demonstrating increased porphyrins or porphyrin precursors in blood, urine and faeces. Patients may only be investigated once, and therefore, understanding the preanalytical factors affecting the reliability of results is crucial. Guidance for sample handling exists, but published evidence regarding the stability of porphyrins and their precursors is limited. The aim of this study was to evaluate the effect of light exposure and different storage temperatures on analyte stability for measurement of urinary aminolaevulinic acid and porphobilinogen, total urine porphyrin and plasma porphyrin. Our results confirm that all samples should be protected from light. Results from samples exposed to light for greater than 4 hours should be interpreted with caution and repeat samples requested. If transported to a specialist laboratory, samples should be stored at 4°C before transport. Transit time at ambient temperatures should be less than 24 hours.
Collapse
Affiliation(s)
- Claire J Gallagher
- Cardiff Porphyria Service, University Hospital of Wales, Cardiff, UK
- Department of Medical Biochemistry and Immunology, University Hospital of Wales, Cardiff, UK
| | - Lucy-Anne Bentley
- Cardiff Porphyria Service, University Hospital of Wales, Cardiff, UK
- Department of Medical Biochemistry and Immunology, University Hospital of Wales, Cardiff, UK
| | - Rhiannon Challenger
- Cardiff Porphyria Service, University Hospital of Wales, Cardiff, UK
- Department of Medical Biochemistry and Immunology, University Hospital of Wales, Cardiff, UK
| | - Martyn Jones
- Cardiff Porphyria Service, University Hospital of Wales, Cardiff, UK
- Department of Medical Biochemistry and Immunology, University Hospital of Wales, Cardiff, UK
| | - Danja Schulenburg-Brand
- Cardiff Porphyria Service, University Hospital of Wales, Cardiff, UK
- Department of Haematology, Immunology and Metabolic Medicine, University Hospital of Wales, Cardiff, UK
| |
Collapse
|
30
|
Abraham NS, Mishra S, Vig S. Palliative Care Aspects of Acute Intermittent Porphyria - A Case Report. Indian J Palliat Care 2024; 30:275-278. [PMID: 39371493 PMCID: PMC11450846 DOI: 10.25259/ijpc_14_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 08/05/2024] [Indexed: 10/08/2024] Open
Abstract
Acute intermitttent porphyria belongs to a rare group of diseases hallmarked by deficient biosynthesis of heme. It carries a significant symptom burden, both physical and emotional,and therefore palliative care has emerged as an essential tool in the armamentarium of porphyria management . It takes care of the patient as a whole and caters to all aspects that the disease process demands. There are many lacunae in the literature regarding the palliative management of porphyria. We are reporting a case of a 16-year-old female who presented with severe abdominal pain, lower backache and symmetrical bilateral lower limb pain to the palliative ward referred by the neurology department for supportive care. This case describes the palliative care aspects of porphyria management which was successfully provided in the palliative care unit right from referral till the last. A multidisciplinary palliative care team managed the patient, and the necessary interventions were provided to the patient and family. Palliative acre in AIP needs to be emphasized, and palliative care services need to be utilized in these cases. The unavailability of specific treatment measure, heme, in countries like India further emphasizes the need for long-term supportive care for the patient and family. The case shows the importance of palliative care throughout the disease course as it is a chronic disease with significant morbidity and carries a heavy symptom burden. This case provides the insight that rather than conventional management alone for such chronic diseases, palliative care should be incorportated. Early integration with palliative care helps in exploring all the domains of disease. This is one of the first cases reported highlighting palliative care in porphyria , bridging the gap in the literature.
Collapse
Affiliation(s)
- Neethu Susan Abraham
- Department of Palliative Medicine, All India Institute of Medical Sciences, Dr B.R. Ambedkar, Institute Rotary Cancer Hospital, New Delhi, Delhi, India
| | - Seema Mishra
- Department of Onco-Anaesthesia and Palliative Medicine, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Saurabh Vig
- Department of Onco-Anaesthesia and Palliative Medicine, All India Institute of Medical Sciences, Dr B.R. Ambedkar, Institute Rotary Cancer Hospital, New Delhi, Delhi, India
| |
Collapse
|
31
|
Zeng T, Chen SR, Liu HQ, Chong YT, Li XH. Successful treatment of severe hepatic impairment in erythropoietic protoporphyria: A case report and review of literature. World J Hepatol 2024; 16:966-972. [PMID: 38948434 PMCID: PMC11212650 DOI: 10.4254/wjh.v16.i6.966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Erythropoietic protoporphyria (EPP) is a rare genetic disorder stemming from ferrochelatase gene mutations, which leads to abnormal accumulation of protoporphyrin IX primarily in erythrocytes, skin, bone marrow and liver. Although porphyria-related severe liver damage is rare, its consequences can be severe with limited treatment options. CASE SUMMARY This case study highlights a successful intervention for a 35-year-old male with EPP-related liver impairment, employing a combination of red blood cell (RBC) exchange and therapeutic plasma exchange (TPE). The patient experienced significant symptom relief and a decrease in bilirubin levels following multiple PE sessions and an RBC exchange. CONCLUSION The findings suggest that this combined approach holds promise for managing severe hepatic impairment in EPP.
Collapse
Affiliation(s)
- Tao Zeng
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
| | - Shu-Ru Chen
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
| | - Hao-Qiang Liu
- Department of Blood Transfusion, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
| | - Yu-Tian Chong
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
| | - Xin-Hua Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China.
| |
Collapse
|
32
|
Balogun O, Nejak-Bowen K. Understanding Hepatic Porphyrias: Symptoms, Treatments, and Unmet Needs. Semin Liver Dis 2024; 44:209-225. [PMID: 38772406 PMCID: PMC11268267 DOI: 10.1055/s-0044-1787076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
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.
Collapse
Affiliation(s)
- Oluwashanu Balogun
- Department of Experimental Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Kari Nejak-Bowen
- Department of Experimental Pathology, University of Pittsburgh, Pittsburgh, PA
- Pittsburgh Liver Institute, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
33
|
Riera-Mestre A, García Morillo JS, Castelbón Fernández J, Hernández-Contreras ME, Aguilera Peiró P, Jacob J, Martínez Valle F, Guillén-Navarro E, Morales-Conejo M. PICO questions and DELPHI methodology for improving the management of patients with acute hepatic porphyria. Rev Clin Esp 2024; 224:272-280. [PMID: 38642893 DOI: 10.1016/j.rceng.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/05/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Acute hepatic porphyrias (AHPs) are a group of rare diseases that encompasses acute intermittent porphyria, variegate porphyria, hereditary coproporphyria, and 5-aminolaevulinic acid dehydratase deficiency porphyria. Symptoms of AHP are nonspecific which, together with its low prevalence, difficult the diagnosis and follow-up of these patients. MATERIAL AND METHODS This project used DELPHI methodology to answer PICO questions related to management of patients with AHPs. The objective was to reach a consensus among multidisciplinary porhyria experts providing answers to those PICO questions for improving diagnosis and follow-up of patients with AHP. RESULTS Ten PICO questions were defined and grouped in four domains: 1. Biochemical diagnosis of patients with AHP. 2. Molecular tests for patients with AHP. 3. Follow-up of patients with AHP. 4. Screening for long-term complications of patients with AHP. CONCLUSIONS PICO questions and DELPHI methodology have provided a consensus on relevant and controversial issues for improving the management of patients with AHP.
Collapse
Affiliation(s)
- A Riera-Mestre
- Servicio de Medicina Interna, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain; Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain; Departamento de Ciencias Clínicas, Facultad de Medicina y Ciencias de la Salud, Universitat de Barcelona, Barcelona, Spain.
| | - J S García Morillo
- Unidad de Enfermedades Autoinmunes y Minoritarias del Adulto, CSUR de Errores Innatos del Metabolismo del Adulto, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - J Castelbón Fernández
- Unidad de Enfermedades Minoritarias y Errores Congénitos del Metabolismo del Adulto (CSUR), Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Madrid, Spain; Instituto de Investigación Hospital 12 de Octubre (i + 12), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), unidad 723, Madrid, Spain
| | - M E Hernández-Contreras
- Servicio de Medicina Interna, CSUR de Enfermedades Metabólicas Congénitas para Niños y Adultos, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain; Departamento de Medicina Interna, Facultad de Medicina, Universidad de Murcia (UMU), Murcia, Spain
| | - P Aguilera Peiró
- Servicio de Dermatología, Hospital Clínic de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Madrid, Spain; Departamento de Medicina. Universitat de Barcelona, Barcelona, Spain; Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - J Jacob
- Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain; Departamento de Ciencias Clínicas, Facultad de Medicina y Ciencias de la Salud, Universitat de Barcelona, Barcelona, Spain; Servicio de Urgencias, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - F Martínez Valle
- Servicio de Medicina Interna, Hospital Universitari Vall d'Hebrón, Barcelona, Spain
| | - E Guillén-Navarro
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Madrid, Spain; Sección de Genética Médica y Servicio de Pediatría, CSUR de Enfermedades Metabólicas Congénitas para Niños y Adultos, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain; Departamento de Cirugía, Pediatría, Obstetricia y Ginecología, Facultad de Medicina, Universidad de Murcia (UMU), Murcia, Spain; Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia, Spain
| | - M Morales-Conejo
- Unidad de Enfermedades Minoritarias y Errores Congénitos del Metabolismo del Adulto (CSUR), Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Madrid, Spain; Instituto de Investigación Hospital 12 de Octubre (i + 12), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), unidad 723, Madrid, Spain
| |
Collapse
|
34
|
Dunaway LS, Loeb SA, Petrillo S, Tolosano E, Isakson BE. Heme metabolism in nonerythroid cells. J Biol Chem 2024; 300:107132. [PMID: 38432636 PMCID: PMC10988061 DOI: 10.1016/j.jbc.2024.107132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/31/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024] Open
Abstract
Heme is an iron-containing prosthetic group necessary for the function of several proteins termed "hemoproteins." Erythrocytes contain most of the body's heme in the form of hemoglobin and contain high concentrations of free heme. In nonerythroid cells, where cytosolic heme concentrations are 2 to 3 orders of magnitude lower, heme plays an essential and often overlooked role in a variety of cellular processes. Indeed, hemoproteins are found in almost every subcellular compartment and are integral in cellular operations such as oxidative phosphorylation, amino acid metabolism, xenobiotic metabolism, and transcriptional regulation. Growing evidence reveals the participation of heme in dynamic processes such as circadian rhythms, NO signaling, and the modulation of enzyme activity. This dynamic view of heme biology uncovers exciting possibilities as to how hemoproteins may participate in a range of physiologic systems. Here, we discuss how heme is regulated at the level of its synthesis, availability, redox state, transport, and degradation and highlight the implications for cellular function and whole organism physiology.
Collapse
Affiliation(s)
- Luke S Dunaway
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Skylar A Loeb
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA; Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Sara Petrillo
- Deptartment Molecular Biotechnology and Health Sciences and Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Emanuela Tolosano
- Deptartment Molecular Biotechnology and Health Sciences and Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA; Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA.
| |
Collapse
|
35
|
La Sala L, Carlini V, Conte C, Macas-Granizo MB, Afzalpour E, Martin-Delgado J, D'Anzeo M, Pedretti RFE, Naselli A, Pontiroli AE, Cappato R. Metabolic disorders affecting the liver and heart: Therapeutic efficacy of miRNA-based therapies? Pharmacol Res 2024; 201:107083. [PMID: 38309383 DOI: 10.1016/j.phrs.2024.107083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
Liver and heart disease are major causes of death worldwide. It is known that metabolic alteration causing type 2 diabetes (T2D) and Nonalcoholic fatty liver (NAFLD) coupled with a derangement in lipid homeostasis, may exacerbate hepatic and cardiovascular diseases. Some pharmacological treatments can mitigate organ dysfunctions but the important side effects limit their efficacy leading often to deterioration of the tissues. It needs to develop new personalized treatment approaches and recent progresses of engineered RNA molecules are becoming increasingly viable as alternative treatments. This review outlines the current use of antisense oligonucleotides (ASOs), RNA interference (RNAi) and RNA genome editing as treatment for rare metabolic disorders. However, the potential for small non-coding RNAs to serve as therapeutic agents for liver and heart diseases is yet to be fully explored. Although miRNAs are recognized as biomarkers for many diseases, they are also capable of serving as drugs for medical intervention; several clinical trials are testing miRNAs as therapeutics for type 2 diabetes, nonalcoholic fatty liver as well as cardiac diseases. Recent advances in RNA-based therapeutics may potentially facilitate a novel application of miRNAs as agents and as druggable targets. In this work, we sought to summarize the advancement and advantages of miRNA selective therapy when compared to conventional drugs. In particular, we sought to emphasise druggable miRNAs, over ASOs or other RNA therapeutics or conventional drugs. Finally, we sought to address research questions related to efficacy, side-effects, and range of use of RNA therapeutics. Additionally, we covered hurdles and examined recent advances in the use of miRNA-based RNA therapy in metabolic disorders such as diabetes, liver, and heart diseases.
Collapse
Affiliation(s)
- Lucia La Sala
- IRCCS MultiMedica, 20138 Milan, Italy; Dept. of Biomedical Sciences for Health, University of Milan, Milan, Italy.
| | | | - Caterina Conte
- IRCCS MultiMedica, 20138 Milan, Italy; Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| | | | - Elham Afzalpour
- Dept. of Biomedical Sciences and Clinic, University of Milan, Milan, Italy
| | - Jimmy Martin-Delgado
- Hospital Luis Vernaza, Junta de Beneficiencia de Guayaquil, 090603 Guayaquil, Ecuador; Instituto de Investigacion e Innovacion en Salud Integral, Universidad Catolica de Santiago de Guayaquil, Guayaquil 090603, Ecuador
| | - Marco D'Anzeo
- AUO delle Marche, SOD Medicina di Laboratorio, Ancona, Italy
| | | | | | | | | |
Collapse
|
36
|
Shrestha M, Amin S, Reggio C, Pokhrel A, Munankami S, Nypaver J, Gupta R, Donato A. Unveiling the Chameleon: A Case Report on Acute Intermittent Porphyria. Cureus 2024; 16:e56222. [PMID: 38618379 PMCID: PMC11016324 DOI: 10.7759/cureus.56222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2024] [Indexed: 04/16/2024] Open
Abstract
Acute intermittent porphyria (AIP) is a rare autosomal dominant metabolic disorder with low penetrance, often presenting with a broad spectrum of clinical manifestations. Acute neurovisceral attacks commonly occur in young women, mimicking signs and symptoms of other medical and psychiatric conditions, thus delaying the diagnosis. We present the case of an 18-year-old female college student with recurrent hospitalizations for intractable abdominal pain, now again with pain and new subjective hematuria. The patient had previously undergone an endoscopy/colonoscopy with negative biopsies and serologies for acute pathology, including celiac disease. Celiac studies were repeated, given the possibility of inadvertent gluten exposure before the onset of the latest symptoms, but were negative. Basic labs and repeat imaging, including contrast-enhanced CT, MRI, and magnetic resonance (MR) enterography of the abdomen, continued to be unremarkable, and the patient's symptoms were felt to be functional in etiology. The patient's urinalysis was normal, and pregnancy was also ruled out. The patient continued to have pain despite receiving opiate analgesics, thus prompting a psychiatry consultation. She was diagnosed with acute adjustment disorder with anxiety and was started on hydroxyzine. Due to persistent symptoms, serum and urine samples were sent, revealing low levels of porphobilinogen deaminase (PBGD) and hydroxymethylbilane synthase (HMBS) gene mutation, confirming the diagnosis of AIP. She was treated with oral glucose and outpatient IV hemin infusions with the resolution of symptoms. AIP presents a nonspecific and highly variable clinical picture, often making it a challenging diagnosis due to such a broad differential. While our patient was thought to have acute adjustment disorder due to an unremarkable initial workup, further testing revealed otherwise. This case demonstrates how clinicians must have a high suspicion of AIP when caring for young females, manifesting with neurovisceral and psychiatric signs and symptoms. Timely diagnosis improves a patient's quality of life and can decrease overutilization of healthcare resources.
Collapse
Affiliation(s)
| | - Shefali Amin
- Internal Medicine, Tower Health Medical Group, Reading, USA
| | | | - Arpan Pokhrel
- Internal Medicine, Tower Health Medical Group, Reading, USA
| | | | - Jakob Nypaver
- Medicine, Trauma Services, Hematology/Oncology, Drexel University College of Medicine, Philadelphia, USA
| | - Riju Gupta
- Internal Medicine, Tower Health Medical Group, Reading, USA
| | - Anthony Donato
- Internal Medicine, Tower Health Medical Group, Reading, USA
| |
Collapse
|
37
|
Levy C, Dickey AK, Wang B, Thapar M, Naik H, Keel SB, Saberi B, Beaven SW, Rudnick SR, Elmariah SB, Erwin AL, Goddu RJ, Hedstrom K, Leaf RK, Kazamel M, Mazepa M, Philpotts LL, Quigley J, Raef H, Ungar J, Anderson KE, Balwani M. Evidence-based consensus guidelines for the diagnosis and management of protoporphyria-related liver dysfunction in erythropoietic protoporphyria and X-linked protoporphyria. Hepatology 2024; 79:731-743. [PMID: 37505211 PMCID: PMC10818013 DOI: 10.1097/hep.0000000000000546] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/13/2023] [Indexed: 07/29/2023]
Affiliation(s)
- Cynthia Levy
- Division of Digestive Health and Liver Diseases, University of Miami Miller School of Medicine, Miami, FL
| | - Amy K. Dickey
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Bruce Wang
- Department of Medicine, University of California San Francisco Medical Center, San Francisco, CA
| | - Manish Thapar
- Division of Gastroenterology, Thomas Jefferson University Hospital, Philadelphia, PA
| | - Hetanshi Naik
- Department of Genetics, Stanford University School of Medicine, Palo Alto, CA
| | - Siobán B. Keel
- Division of Hematology, University of Washington School of Medicine, Seattle, WA
| | - Behnam Saberi
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Simon W. Beaven
- Vatche & Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles Medical Center, Los Angeles, CA
| | - Sean R. Rudnick
- Department of Internal Medicine, Section on Gastroenterology and Hepatology, Atrium Health Wake Forest Baptist, Winston-Salem, NC
| | - Sarina B. Elmariah
- Department of Dermatology, University of California San Francisco, San Francisco, CA
| | - Angelika L. Erwin
- Center for Personalized Genetic Healthcare, Cleveland Clinic, Cleveland, OH
| | - Robert J. Goddu
- Division of Continuing Education, University of Colorado Boulder, Boulder, CO
| | - Karli Hedstrom
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rebecca Karp Leaf
- Harvard Medical School, Boston, MA
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Mohamed Kazamel
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Marshall Mazepa
- Division of Hematology,Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | | | - John Quigley
- Division of Hematology/Oncology, Department of Medicine, University of Illinois Chicago, IL
| | - Haya Raef
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Division of Hematology, University of Washington School of Medicine, Seattle, WA
- Department of Dermatology, University of California San Francisco, San Francisco, CA
| | - Jonathan Ungar
- Department of Dermatology, Mount Sinai Hospital, New York, NY
| | - Karl E. Anderson
- Department of Internal Medicine (Division of Gastroenterology & Hepatology), University of Texas Medical Branch/UTMB Health, Galveston, TX
| | - Manisha Balwani
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
38
|
Castelbón Fernández FJ, Barreda Sánchez M, Arranz Canales E, Hernández Contreras ME, Solares I, Morales Conejo M, Muñoz Cuadrado Á, Casado Gómez A, Yébenes Cortés M, Guillén Navarro E. The burden of disease and quality of life in patients with acute hepatic porphyria: COPHASE study. Med Clin (Barc) 2024; 162:103-111. [PMID: 37838536 DOI: 10.1016/j.medcli.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Acute hepatic porphyria (AHP) comprises a group of rare genetic diseases characterized by neurovisceral crises that are manifested by abdominal pain and neurological and/or psychological symptoms that interfere with the ability to lead a normal life. Our objective was to determine the burden of the disease in one year and the health-related quality of life (HRQoL) in patients with AHP. RESULTS 28 patients were analyzed. The mean age was 36.6±10.2 years, 89.3% were women, and the average number of crises was 1.9±1.5. The average annual cost per patient was €38,255.40. 80.2% of the costs was direct medical costs, 17.5% was associated with loss of productivity and 2.3% was direct non-medical costs. 85.9% of the total cost corresponded to the crises. The intercrisis period accounted for the remaining 14.1%. The global index of the EQ-5D-5L (HRQoL) was 0.75±0.24. The dimensions of pain/discomfort, anxiety/depression and daily activities were the most affected. Leisure, travel/vacations and household activities were the most affected daily activities. 53.6% of patients required a caregiver due to AHP. 92.9% did not present overload and 7.1% presented extreme overload. CONCLUSIONS Patients with AHP are associated with a high economic impact and an affected HRQoL in the pain/discomfort dimension, with a negative impact on the performance of daily activities and a risk of psychiatric diseases.
Collapse
Affiliation(s)
- Francisco Javier Castelbón Fernández
- Consulta de Porfirias, CSUR de errores congénitos del metabolismo, Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - María Barreda Sánchez
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Elena Arranz Canales
- Consulta de Porfirias, CSUR de errores congénitos del metabolismo, Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Isabel Solares
- Consulta de Porfirias, CSUR de errores congénitos del metabolismo, Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Montserrat Morales Conejo
- Consulta de Porfirias, CSUR de errores congénitos del metabolismo, Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | | | - Encarna Guillén Navarro
- Sección Genética Médica, Hospital Clínico Universitario Virgen de la Arrixaca/Universidad de Murcia/IMIB-Arrixaca, Murcia, Spain
| |
Collapse
|
39
|
Wang B, Wang Y, Zhang Z, Wen X, Xi Z. Insight into the Role of an α-Helix Cluster in Protoporphyrinogen IX Oxidase. Biochemistry 2024. [PMID: 38285491 DOI: 10.1021/acs.biochem.3c00508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Protoporphyrinogen IX oxidase (PPO) is the last common enzyme in chlorophyll and heme biosynthesis pathways. In humans, point mutations on PPO are responsible for the dominantly inherited disorder disease variegate porphyria (VP). It is found that several VP-causing mutation sites are located on an α-helix cluster (consisting of α-5, α-6, and α-7 helix, named the G169 helix cluster) of human PPO, although these mutation sites are outside the active site of the human PPO. In this work, we investigated the role of the G169 helix cluster via site-directed mutagenesis, enzymatic kinetics, and computational studies. Kinetic studies showed that mutations on the G169 helix cluster affect the activity of PPO. The MD simulation showed that mutations on the G169 helix cluster reduced the activity of PPO by affecting the proper orientation of substrate protoporphyrinogen within the active site of PPO and possibly the dipole moment of the G169 helix cluster. Moreover, the mutation abolished the interaction between the mutated site and other residues, thus affecting the secondary structure and hydrogen bond interactions within the G169 helix cluster. These results indicated that the integrity of the G169 helix cluster is important for the stabilization of protoporphyrinogen within the active site of PPO to facilitate the interaction between protoporphyrinogen and cofactor FAD and provide a proper electrostatic environment for the activity of PPO. Our result provides new insight into understanding the relationship between the structure and function of PPO.
Collapse
Affiliation(s)
- Baifan Wang
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, National Pesticide Engineering Research Center (Tianjin), Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yiban Wang
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, National Pesticide Engineering Research Center (Tianjin), Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zijuan Zhang
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, National Pesticide Engineering Research Center (Tianjin), Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xin Wen
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, National Pesticide Engineering Research Center (Tianjin), Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zhen Xi
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, National Pesticide Engineering Research Center (Tianjin), Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
40
|
Yasuda M. [Acute hepatic porphyrias: pathophysiology and pathogenesis of acute attacks]. Rinsho Shinkeigaku 2024; 64:8-16. [PMID: 38092415 DOI: 10.5692/clinicalneurol.cn-001856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Heme is an iron-containing molecule essential for virtually all living organisms. However, excessive heme is cytotoxic, necessitating tight regulation of intracellular heme concentration. The acute hepatic porphyrias (AHPs) are a group of rare inborn errors of heme biosynthesis that are characterized by episodic acute neurovisceral attacks that are precipitated by various factors. The AHPs are often misdiagnosed, as the acute attack symptom are non-specific and can be attributed to other more common causes. Understanding how heme biosynthesis is dysregulated in AHP patients and the mechanism by which acute attacks are precipitated will aid in accurate and rapid diagnoses, and subsequently, appropriate treatment of these disorders. Therefore, this review article will focus on the biochemical and molecular changes that occur during an acute attack and present what is currently known regarding the underlying pathogenesis of acute attacks.
Collapse
Affiliation(s)
- Makiko Yasuda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai
| |
Collapse
|
41
|
Gerischer L, Mainert M, Wohmann N, Kubisch I, Stölzel U, Stauch T, von Wegerer S, Braun F, Weiler-Normann C, Blaschke S, Frank J, Somasundaram R, Diehl-Wiesenecker E. German Porphyria Registry (PoReGer)-Background and Setup. Healthcare (Basel) 2024; 12:111. [PMID: 38201016 PMCID: PMC10779132 DOI: 10.3390/healthcare12010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Porphyrias, as most rare diseases, are characterized by complexity and scarcity of knowledge. A national registry in one of the largest European populations that prospectively collects longitudinal clinical and laboratory data are an important and effective tool to close this gap. The German Porphyria Registry (PoReGer) was founded by four centers with longstanding expertise in the field of porphyrias and rare diseases (Charité-Universitätsmedizin Berlin, Porphyria Center Saxony Chemnitz, University Medical Center Hamburg-Eppendorf, University Medical Center Göttingen) and the German reference laboratory for porphyria, and is supported by the largest German porphyria patient organization. A specified data matrix for three subgroups (acute, chronic blistering cutaneous, acute non-blistering cutaneous) includes data on demographics, specific porphyria-related symptoms, clinical course, general medical history, necessary follow-up assessments (including laboratory and imaging results), symptomatic and disease-modifying therapies, and side-effects. Additionally, the registry includes patient-reported outcome measures on quality of life, depression, and fatigue. PoReGer aims to broaden and deepen the understanding on all porphyria-related subjects. We expect these data to significantly improve the management and care of porphyria patients. Additionally, the data can be used for educational purposes to increase awareness, for the planning of healthcare services, and for machine learning algorithms for early detection of porphyrias.
Collapse
Affiliation(s)
- Lea Gerischer
- Department of Neurology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Neuroscience Clinical Research Center, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Mona Mainert
- Department of Emergency Medicine and Porphyria Clinic, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Nils Wohmann
- Department of Internal Medicine II, Saxony Porphyria Center, Klinikum Chemnitz, gGmbH, 09116 Chemnitz, Germany
| | - Ilja Kubisch
- Department of Internal Medicine II, Saxony Porphyria Center, Klinikum Chemnitz, gGmbH, 09116 Chemnitz, Germany
| | - Ulrich Stölzel
- Department of Internal Medicine II, Saxony Porphyria Center, Klinikum Chemnitz, gGmbH, 09116 Chemnitz, Germany
| | - Thomas Stauch
- Porphyria Laboratory IPNET, MVZ Labor PD Dr. Med. Volkmann GbR, 76131 Karlsruhe, Germany
| | | | - Fabian Braun
- Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christina Weiler-Normann
- Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sabine Blaschke
- Emergency Department, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Jorge Frank
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Rajan Somasundaram
- Department of Emergency Medicine and Porphyria Clinic, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Eva Diehl-Wiesenecker
- Department of Emergency Medicine and Porphyria Clinic, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| |
Collapse
|
42
|
Petrukhina D, Musina N, Slavkina Y, Latypova V, Zemlyanaya N, Saprina T. Incidence of carbohydrate metabolism disorder in iron overload of different etiology. RUSSIAN JOURNAL OF PREVENTIVE MEDICINE 2024; 27:111. [DOI: 10.17116/profmed202427051111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Smith M, Foong KS. Cefiderocol-associated brown chromaturia. BMJ Case Rep 2023; 16:e258207. [PMID: 38103906 PMCID: PMC10728909 DOI: 10.1136/bcr-2023-258207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Cefiderocol is a novel siderophore cephalosporin antibiotic. In the present case report, a woman in her 70s presented with a 1-week history of altered mental status and progressive purulent discharge from a non-healing diabetic foot ulcer on her right heel. MRI of the right foot revealed chronic osteomyelitis of the calcaneum. Surgical debridement was performed, and the tissue cultures grew extensively drug resistant (XDR) Pseudomonas aeruginosa, XDR Acinetobacter baumannii and Enterococcus faecalis The patient received ampicillin-sulbactam and cefiderocol. The antibiotic treatment course was complicated by brown urine discolouration. Investigations were unrevealing for haemoglobinuria, myoglobinuria and bilirubinuria. A side effect from cefiderocol was suspected and subsequently discontinued. Her urine colour returned to its normal colour within 3 days of discontinuation of cefiderocol.
Collapse
|
44
|
Horie Y, Yasuoka Y, Adachi T. Clinical features of acute attacks, chronic symptoms, and long-term complications among patients with acute hepatic porphyria in Japan: a real-world claims database study. Orphanet J Rare Dis 2023; 18:384. [PMID: 38066651 PMCID: PMC10704654 DOI: 10.1186/s13023-023-02913-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/06/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Acute hepatic porphyria (AHP) is a family of rare genetic diseases, including acute intermittent porphyria, variegate porphyria, hereditary coproporphyria, and delta-aminolevulinic acid dehydratase-deficient porphyria. The objective of this retrospective cohort study was to provide information on the clinical features of AHP in Japan-including acute attacks, chronic symptoms, and long-term complications. METHODS Patients with AHP between April 2008 and June 2020 were selected from Japan's Medical Data Vision claims database. Patients with AHP were matched 1:10, by sex and age, to patients without AHP. The outcomes were evaluated overall, for patients age ≥ 55 years, and for the matched population. RESULTS A total of 391 patients with AHP were included from the Japanese Medical Data Vision database. During the observation period (April 2008-June 2020), 18.2% (71/391) of patients experienced 1 acute attack and 10.5% (41/391) experienced ≥ 2 attacks. Chronic symptoms with rates ~ 10% or higher in the AHP population compared with the matched population included neurotic, stress-related, and somatoform disorders (21.7% vs. 6.7% [15.0% difference]); sleep disorders (23.0% vs. 9.9% [13.1% difference]); other and unspecified abdominal pain (13.6% vs. 3.7% [9.9% difference]); and nausea and vomiting, excluding chemotherapy-induced emesis (17.9% vs. 8.1% [9.8% difference]). Long-term complications with higher incidence rates in the AHP population compared with the matched population included fibrosis and cirrhosis of liver (15.9% vs. 3.0% [12.9% difference]), polyneuropathies and other disorders of the peripheral nervous system (20.5% vs. 7.9% [12.6% difference]), liver cancer (16.9% vs. 4.7% [12.2% difference]), renal failure (16.4% vs. 4.3% [12.1% difference]), and hypertension (26.1% vs. 18.8% [7.3% difference]). Among AHP patients age ≥ 55 years, the most common long-term complications were hypertension, kidney failure, and liver cancer. CONCLUSIONS In Japan, patients with AHP experience a high clinical burden in terms of acute attacks, chronic symptoms, and long-term complications. The clinical burden related to chronic symptoms and long-term complications was substantially higher in Japanese patients with AHP compared with a matched population without AHP. Recognizing these signs and symptoms of AHP may aid physicians in making an earlier diagnosis, which may help patients avoid attack triggers, implement disease management, and reduce lifetime disease burden.
Collapse
Affiliation(s)
- Yutaka Horie
- Department of Gastroenterology, Saiseikai Gotsu General Hospital, Shimane, Japan
| | - Yuka Yasuoka
- Alnylam Japan KK, Pacific Century Place Marunouchi 11th Floor 1-11-1 Marunouchi, Chiyoda-ku, Tokyo, 100-6211, Japan.
| | - Tomohide Adachi
- Department of General Medicine and Neurology, Saiseikai Central Hospital, Tokyo, Japan
| |
Collapse
|
45
|
Poli A, Frieri C, Lefebvre T, Delforge J, Mirmiran A, Talbi N, Moulouel B, Six M, Paradis V, Parquet N, Puy H, Schmitt C, Aslangul E, de Fontbrune FS, Gouya L. Management of erythropoietic protoporphyria with cholestatic liver disease: A case report. Mol Genet Metab Rep 2023; 37:101018. [PMID: 38053924 PMCID: PMC10694760 DOI: 10.1016/j.ymgmr.2023.101018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 12/07/2023] Open
Abstract
Erythropoietic protoporphyria (EPP) is a rare metabolic disease of the heme biosynthetic pathway where an enzymatic dysfunction results in protoporphyrin IX (PPIX) accumulation in erythroid cells. The porphyrins are photo-reactive and are responsible for severe photosensitivity in patients, thus drastically decreasing their quality of life. The liver eliminates PPIX and as such, the main and rare complication of EPP is progressive cholestatic liver disease, which can lead to liver failure. The management of this complication is challenging, as it often requires a combination of approaches to promote PPIX elimination and suppress the patient's erythropoiesis. Here we described a 3-year follow-up of an EPP patient, with three episodes of liver involvement, aggravated by the coexistence of a factor VII deficiency. It covers all the different types of intervention available for the management of liver disease, right through to successful allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Antoine Poli
- Institut National de la Santé et de la Recherche Médicale U1149, Centre de Recherche sur l'Inflammation, Paris, France
- Université Paris Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France
- Laboratory of Excellence Gr-Ex, Paris, France
| | - Camilla Frieri
- Assistance Publique-Hôpitaux de Paris, Service d'hématologie-greffe, Hôpital Saint-Louis, Paris, France
- A.O.R.N. San Giuseppe Moscati Hospital, Avellino, Italy
| | - Thibaud Lefebvre
- Assistance Publique-Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France
| | - Juliette Delforge
- Assistance Publique-Hôpitaux de Paris, Service de médecine interne, Hôpital Louis Mourier, Colombes, France
| | - Arienne Mirmiran
- Assistance Publique-Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France
| | - Neila Talbi
- Assistance Publique-Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France
| | - Boualem Moulouel
- Assistance Publique-Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France
| | - Marion Six
- Assistance Publique-Hôpitaux de Paris, Service de médecine interne, Hôpital Louis Mourier, Colombes, France
| | - Valérie Paradis
- Assistance Publique-Hôpitaux de Paris, Département de pathologie, Hôpital Beaujon, Clichy, France
| | - Nathalie Parquet
- Assistance Publique-Hôpitaux de Paris, Unité d'Aphérèse Thérapeutique, Hôpital Saint-Louis, Paris, France
| | - Hervé Puy
- Institut National de la Santé et de la Recherche Médicale U1149, Centre de Recherche sur l'Inflammation, Paris, France
- Université Paris Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France
- Laboratory of Excellence Gr-Ex, Paris, France
| | - Caroline Schmitt
- Institut National de la Santé et de la Recherche Médicale U1149, Centre de Recherche sur l'Inflammation, Paris, France
- Université Paris Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France
- Laboratory of Excellence Gr-Ex, Paris, France
| | - Elisabeth Aslangul
- Assistance Publique-Hôpitaux de Paris, Service de médecine interne, Hôpital Louis Mourier, Colombes, France
| | - Flore Sicre de Fontbrune
- Assistance Publique-Hôpitaux de Paris, Service d'hématologie-greffe, Hôpital Saint-Louis, Paris, France
| | - Laurent Gouya
- Institut National de la Santé et de la Recherche Médicale U1149, Centre de Recherche sur l'Inflammation, Paris, France
- Université Paris Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France
- Laboratory of Excellence Gr-Ex, Paris, France
| |
Collapse
|
46
|
Yasuda M, Keel S, Balwani M. RNA interference therapy in acute hepatic porphyrias. Blood 2023; 142:1589-1599. [PMID: 37027823 PMCID: PMC10656724 DOI: 10.1182/blood.2022018662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/27/2023] [Accepted: 03/25/2023] [Indexed: 04/09/2023] Open
Abstract
The acute hepatic porphyrias (AHPs) are inherited disorders of heme biosynthesis characterized by life-threatening acute neurovisceral attacks precipitated by factors that upregulate hepatic 5-aminolevulinic acid synthase 1 (ALAS1) activity. Induction of hepatic ALAS1 leads to the accumulation of porphyrin precursors, in particular 5-aminolevulinic acid (ALA), which is thought to be the neurotoxic mediator leading to acute attack symptoms such as severe abdominal pain and autonomic dysfunction. Patients may also develop debilitating chronic symptoms and long-term medical complications, including kidney disease and an increased risk of hepatocellular carcinoma. Exogenous heme is the historical treatment for attacks and exerts its therapeutic effect by inhibiting hepatic ALAS1 activity. The pathophysiology of acute attacks provided the rationale to develop an RNA interference therapeutic that suppresses hepatic ALAS1 expression. Givosiran is a subcutaneously administered N-acetylgalactosamine-conjugated small interfering RNA against ALAS1 that is taken up nearly exclusively by hepatocytes via the asialoglycoprotein receptor. Clinical trials established that the continuous suppression of hepatic ALAS1 mRNA via monthly givosiran administration effectively reduced urinary ALA and porphobilinogen levels and acute attack rates and improved quality of life. Common side effects include injection site reactions and increases in liver enzymes and creatinine. Givosiran was approved by the US Food and Drug Administration and European Medicines Agency in 2019 and 2020, respectively, for the treatment of patients with AHP. Although givosiran has the potential to decrease the risk of chronic complications, long-term data on the safety and effects of sustained ALAS1 suppression in patients with AHP are lacking.
Collapse
Affiliation(s)
- Makiko Yasuda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Siobán Keel
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA
| | - Manisha Balwani
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
47
|
Kuter DJ, Bonkovsky HL, Monroy S, Ross G, Guillén-Navarro E, Cappellini MD, Minder AE, Hother-Nielsen O, Ventura P, Jia G, Sweetser MT, Thapar M. Efficacy and safety of givosiran for acute hepatic porphyria: Final results of the randomized phase III ENVISION trial. J Hepatol 2023; 79:1150-1158. [PMID: 37479139 DOI: 10.1016/j.jhep.2023.06.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/24/2023] [Accepted: 06/15/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND & AIMS Acute hepatic porphyria (AHP) is caused by defects in hepatic heme biosynthesis, leading to disabling acute neurovisceral attacks and chronic symptoms. In ENVISION (NCT03338816), givosiran treatment for 6 months reduced attacks and other disease manifestations compared with placebo. Herein, we report data from the 36-month final analysis of ENVISION. METHODS Ninety-four patients with AHP (age ≥12 years) and recurrent attacks were randomized 1:1 to monthly double-blind subcutaneous givosiran 2.5 mg/kg (n = 48) or placebo (n = 46) for 6 months. In the open-label extension (OLE) period, 93 patients received givosiran 2.5 or 1.25 mg/kg for 6 months or more before transitioning to 2.5 mg/kg. Endpoints were exploratory unless otherwise noted. RESULTS During givosiran treatment, the median annualized attack rate (AAR) was 0.4. Through Month 36, annualized days of hemin use remained low in the continuous givosiran group (median, 0.0 to 0.4) and decreased in the placebo crossover group (16.2 to 0.4). At end of OLE, in the continuous givosiran and placebo crossover groups, 86% and 92%, respectively, had 0 attacks. AAR was lower than historical AAR in 98% and 100%, respectively (post hoc analysis), and there were 0 days of hemin use in 88% and 90%, respectively. The 12-item short-form health survey physical and mental component summary scores increased by 8.6 and 8.1, respectively (continuous givosiran) and 9.4 and 3.2, respectively (placebo crossover). EQ-5D health-related questionnaire scores increased by 18.9 (continuous givosiran) and 9.9 (placebo crossover). Lower urinary delta-aminolevulinic acid and porphobilinogen levels were sustained. Safety findings demonstrated a continued positive risk/benefit profile for givosiran. CONCLUSIONS Long-term monthly givosiran treatment provides sustained and continued improvement in clinical manifestations of AHP. CLINICALTRIALS GOV IDENTIFIER NCT03338816. EUDRACT NUMBER 2017-002432-17. IMPACT AND IMPLICATIONS Acute hepatic porphyria (AHP) is a group of rare, chronic, multisystem disorders associated with overproduction and accumulation of neurotoxic heme intermediates (delta-aminolevulinic acid and porphobilinogen), sometimes resulting in recurrent acute attacks and long-term complications. Givosiran, a small-interfering RNA that prevents accumulation of delta-aminolevulinic acid and porphobilinogen, is approved for the treatment of AHP. These final 36-month results of ENVISION, a phase III study of givosiran in patients with AHP and recurrent attacks, show that long-term monthly treatment with givosiran leads to continuous and sustained reductions in annualized attack rate and use of hemin over time, as well as improved quality of life, with an acceptable safety profile. These results are important for physicians, patients, families, and caregivers who are grappling with this debilitating and potentially life-threatening disease with few effective and tolerable treatment options.
Collapse
Affiliation(s)
- David J Kuter
- Hematology Division, Massachusetts General Hospital, Boston, MA, USA.
| | | | - Susana Monroy
- Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Gayle Ross
- Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Encarna Guillén-Navarro
- Medical Genetics Section, Virgen de la Arrixaca University Hospital, IMIB Pascual Parrilla, University of Murcia (UMU), Murcia, Spain; CIBERER-ISCIII, Madrid, Spain
| | | | - Anna-Elisabeth Minder
- Division of Endocrinology, Diabetes and Porphyria, Stadtspital Zürich, Triemli, Zürich, Switzerland
| | | | - Paolo Ventura
- Department of Surgical and Medical Sciences for Children and Adults, Internal Medicine Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Gang Jia
- Alnylam Pharmaceuticals, Cambridge, MA, USA
| | | | | |
Collapse
|
48
|
Moghe A, Dickey A, Erwin A, Leaf RK, O'Brien A, Quigley JG, Thapar M, Anderson KE. Acute hepatic porphyrias: Recommendations for diagnosis and management with real-world examples. Mol Genet Metab 2023; 140:107670. [PMID: 37542766 DOI: 10.1016/j.ymgme.2023.107670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
Acute hepatic porphyria (AHP) is a group of four rare inherited diseases, each resulting from a deficiency in a distinct enzyme in the heme biosynthetic pathway. Characterized by acute neurovisceral symptoms that may mimic other medical and psychiatric conditions, lack of recognition of the disease often leads to a delay in diagnosis and initiation of effective treatment. Biochemical testing for pathway intermediates that accumulate when the disease is active forms the basis for screening and establishing a diagnosis. Subsequent genetic analysis identifies the pathogenic variant, supporting screening of family members and genetic counseling. Management of AHP involves avoidance of known exogenous and hormonal triggers, symptomatic treatment, and prevention of recurrent attacks. Here we describe six case studies from our own real-world experience to highlight current recommendations and challenges associated with the diagnosis and long-term management of the disease.
Collapse
Affiliation(s)
- Akshata Moghe
- Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States of America.
| | - Amy Dickey
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, United States of America
| | - Angelika Erwin
- Center for Personalized Genetic Healthcare, Cleveland Clinic Community Care, Cleveland Clinic, Cleveland, OH, United States of America
| | - Rebecca K Leaf
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Alan O'Brien
- Service de Médecine Génique, Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - John G Quigley
- Department of Medicine, University of Illinois College of Medicine, Chicago, IL, United States of America
| | - Manish Thapar
- Division of Gastroenterology, Thomas Jefferson University Hospital, Philadelphia, PA, United States of America
| | - Karl E Anderson
- Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States of America
| |
Collapse
|
49
|
Lissing M, Wester A, Vassiliou D, Floderus Y, Harper P, Sardh E, Wahlin S. Porphyrin precursors and risk of primary liver cancer in acute intermittent porphyria: A case-control study of 188 patients. J Inherit Metab Dis 2023; 46:1186-1194. [PMID: 37650859 DOI: 10.1002/jimd.12676] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/01/2023]
Abstract
Acute intermittent porphyria (AIP) is a rare hereditary metabolic disease characterized by acute attacks and accumulation of the porphyrin precursors 5-aminolevulinic acid (ALA) and porphobilinogen (PBG). Patients with AIP have a high risk of primary liver cancer (PLC). We aimed to assess the association between porphyrin precursor excretion and the risk for PLC in patients with AIP. We studied 48 patients with AIP who developed PLC between 1987 and 2015 and 140 age and sex matched controls with AIP but no PLC. Data on all available urinary PBG and ALA samples collected from 1975 until 1 year before PLC diagnosis were analyzed and compared between cases and controls using logistic regression. Porphyrin precursor excretion was higher in patients with PLC (PBG median 7.9 [IQR 4.4-21.9] mmol/mol creatinine) than in controls (3.8 [1.2-9.8]) (adjusted odds ratio 1.07, 95% confidence interval: 1.02-1.12). None of the 28 patients with all registered samples below the upper limit of normal (ULN) developed PLC, and only one of the 45 patients with all samples <2× ULN developed PLC. Among non-PLC controls, ALA and PBG levels decreased after age 50-60 while an increasing trend was observed after age 65 among those who developed PLC. Increased urinary porphyrin precursors are associated with a high risk of developing PLC. Patients with normal levels appear to have a low risk while high or increasing ALA and PBG after age 65 indicates high risk, which should be considered in surveillance decisions.
Collapse
Affiliation(s)
- Mattias Lissing
- Hepatology Division, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Axel Wester
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Daphne Vassiliou
- Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases (CMMS), Porphyria Centre Sweden, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ylva Floderus
- Centre for Inherited Metabolic Diseases (CMMS), Porphyria Centre Sweden, Karolinska University Hospital, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Pauline Harper
- Centre for Inherited Metabolic Diseases (CMMS), Porphyria Centre Sweden, Karolinska University Hospital, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Eliane Sardh
- Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases (CMMS), Porphyria Centre Sweden, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Staffan Wahlin
- Hepatology Division, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
50
|
Hill AC, Becker JP, Slominski D, Halloy F, Søndergaard C, Ravn J, Hall J. Peptide Conjugates of a 2'- O-Methoxyethyl Phosphorothioate Splice-Switching Oligonucleotide Show Increased Entrapment in Endosomes. ACS OMEGA 2023; 8:40463-40481. [PMID: 37929104 PMCID: PMC10620785 DOI: 10.1021/acsomega.3c05144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023]
Abstract
Antisense oligonucleotides (ASOs) are short, single-stranded nucleic acid molecules that alter gene expression. However, their transport into appropriate cellular compartments is a limiting factor in their potency. Here, we synthesized splice-switching oligonucleotides (SSOs) previously developed to treat the rare disease erythropoietic protoporphyria. Using chemical ligation-quantitative polymerase chain reaction (CL-qPCR), we quantified the SSOs in cells and subcellular compartments following free uptake. To drive nuclear localization, we covalently conjugated nuclear localization signal (NLS) peptides to a lead 2'-O-methoxyethyl phosphorothioate SSO using thiol-maleimide chemistry. The conjugates and parent SSO displayed similar RNA target-binding affinities. CL-qPCR quantification of the conjugates in cells and subcellular compartments following free uptake revealed one conjugate with better nuclear accumulation relative to the parent SSO. However, compared to the parent SSO, which altered the splicing of the target pre-mRNA, the conjugates were inactive at splice correction under free uptake conditions in vitro. Splice-switching activity could be conferred on the conjugates by delivering them into cells via cationic lipid-mediated transfection or by treating the cells into which the conjugates had been freely taken up with chloroquine, an endosome-disrupting agent. Our results identify the major barrier to the activity of the peptide-oligonucleotide conjugates as endosomal entrapment.
Collapse
Affiliation(s)
- Alyssa C. Hill
- Institute
of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Eidgenössische Technische Hochschule Zürich
(ETH Zürich), Zürich 8093, Switzerland
| | - J. Philipp Becker
- Institute
of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Eidgenössische Technische Hochschule Zürich
(ETH Zürich), Zürich 8093, Switzerland
| | - Daria Slominski
- Institute
of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Eidgenössische Technische Hochschule Zürich
(ETH Zürich), Zürich 8093, Switzerland
| | - François Halloy
- Institute
of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Eidgenössische Technische Hochschule Zürich
(ETH Zürich), Zürich 8093, Switzerland
| | | | - Jacob Ravn
- Roche
Innovation Center Copenhagen (RICC), Hørsholm 2970, Denmark
| | - Jonathan Hall
- Institute
of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Eidgenössische Technische Hochschule Zürich
(ETH Zürich), Zürich 8093, Switzerland
| |
Collapse
|