Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
J Clin Pathol ; 77(8): 574-578, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-38631910

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.


Subject(s)
Porphyrins , Specimen Handling , Temperature , Humans , Specimen Handling/methods , Porphyrins/urine , Porphyrins/chemistry , Porphyrins/blood , Light , Time Factors , Porphyrias/diagnosis , Porphyrias/urine , Aminolevulinic Acid/urine , Porphobilinogen/urine , Porphobilinogen/blood , Reproducibility of Results , Urinalysis/methods
2.
N Engl J Med ; 380(6): 549-558, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30726693

ABSTRACT

BACKGROUND: Induction of delta aminolevulinic acid synthase 1 ( ALAS1) gene expression and accumulation of neurotoxic intermediates result in neurovisceral attacks and disease manifestations in patients with acute intermittent porphyria, a rare inherited disease of heme biosynthesis. Givosiran is an investigational RNA interference therapeutic agent that inhibits hepatic ALAS1 synthesis. METHODS: We conducted a phase 1 trial of givosiran in patients with acute intermittent porphyria. In part A of the trial, patients without recent porphyria attacks (i.e., no attacks in the 6 months before baseline) were randomly assigned to receive a single subcutaneous injection of one of five ascending doses of givosiran (0.035, 0.10, 0.35, 1.0, or 2.5 mg per kilogram of body weight) or placebo. In part B, patients without recent attacks were randomly assigned to receive once-monthly injections of one of two doses of givosiran (0.35 or 1.0 mg per kilogram) or placebo (total of two injections 28 days apart). In part C, patients who had recurrent attacks were randomly assigned to receive injections of one of two doses of givosiran (2.5 or 5.0 mg per kilogram) or placebo once monthly (total of four injections) or once quarterly (total of two injections) during a 12-week period, starting on day 0. Safety, pharmacokinetic, pharmacodynamic, and exploratory efficacy outcomes were evaluated. RESULTS: A total of 23 patients in parts A and B and 17 patients in part C underwent randomization. Common adverse events included nasopharyngitis, abdominal pain, and diarrhea. Serious adverse events occurred in 6 patients who received givosiran in parts A through C combined. In part C, all 6 patients who were assigned to receive once-monthly injections of givosiran had sustained reductions in ALAS1 messenger RNA (mRNA), delta aminolevulinic acid, and porphobilinogen levels to near normal. These reductions were associated with a 79% lower mean annualized attack rate than that observed with placebo (exploratory efficacy end point). CONCLUSIONS: Once-monthly injections of givosiran in patients who had recurrent porphyria attacks resulted in mainly low-grade adverse events, reductions in induced ALAS1 mRNA levels, nearly normalized levels of the neurotoxic intermediates delta aminolevulinic acid and porphobilinogen, and a lower attack rate than that observed with placebo. (Funded by Alnylam Pharmaceuticals; ClinicalTrials.gov number, NCT02452372 .).


Subject(s)
5-Aminolevulinate Synthetase/antagonists & inhibitors , Amides/administration & dosage , Porphyria, Acute Intermittent/drug therapy , RNAi Therapeutics , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Acetylgalactosamine/analogs & derivatives , Adult , Amides/adverse effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Liver/metabolism , Male , Middle Aged , Molecular Targeted Therapy , Porphobilinogen/blood , Pyrrolidines , RNA, Messenger/metabolism , RNA, Messenger/urine
3.
Hum Mol Genet ; 28(11): 1755-1767, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30615115

ABSTRACT

Acute intermittent porphyria (AIP) is an inborn error of heme biosynthesis due to the deficiency of hydroxymethylbilane synthase (HMBS) activity. Human AIP heterozygotes have episodic acute neurovisceral attacks that typically start after puberty, whereas patients with homozygous dominant AIP (HD-AIP) have early-onset chronic neurological impairment, including ataxia and psychomotor retardation. To investigate the dramatically different manifestations, knock-in mice with human HD-AIP missense mutations c.500G>A (p.Arg167Glu) or c.518_519GC>AG (p.Arg173Glu), designated R167Q or R173Q mice, respectively, were generated and compared with the previously established T1/T2 mice with ~30% residual HMBS activity and the heterozygous AIP phenotype. Homozygous R173Q mice were embryonic lethal, while R167Q homozygous mice (R167Q+/+) had ~5% of normal HMBS activity, constitutively elevated plasma and urinary 5-aminolevulinic acid (ALA) and porphobilinogen (PBG), profound early-onset ataxia, delayed motor development and markedly impaired rotarod performance. Central nervous system (CNS) histology was grossly intact, but CNS myelination was delayed and overall myelin volume was decreased. Heme concentrations in liver and brain were similar to those of T1/T2 mice. Notably, ALA and PBG concentrations in the cerebral spinal fluid and CNS regions were markedly elevated in R167Q+/+ mice compared with T1/T2 mice. When the T1/T2 mice were administered phenobarbital, ALA and PBG markedly accumulated in their liver and plasma, but not in the CNS, indicating that ALA and PBG do not readily cross the blood-brain barrier. Taken together, these studies suggest that the severe HD-AIP neurological phenotype results from decreased myelination and the accumulation of locally produced neurotoxic porphyrin precursors within the CNS.


Subject(s)
Hydroxymethylbilane Synthase/genetics , Nervous System Diseases/genetics , Porphyria, Acute Intermittent/genetics , Psychomotor Disorders/genetics , Aminolevulinic Acid/blood , Aminolevulinic Acid/urine , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Gene Knock-In Techniques , Genes, Dominant , Homozygote , Humans , Hydroxymethylbilane Synthase/metabolism , Liver/metabolism , Mice , Mutation, Missense/genetics , Myelin Sheath/genetics , Myelin Sheath/metabolism , Nervous System Diseases/blood , Nervous System Diseases/pathology , Nervous System Diseases/urine , Phenobarbital/pharmacology , Porphobilinogen/blood , Porphobilinogen/urine , Porphyria, Acute Intermittent/blood , Porphyria, Acute Intermittent/pathology , Porphyria, Acute Intermittent/urine , Psychomotor Disorders/blood , Psychomotor Disorders/pathology , Psychomotor Disorders/urine
4.
Mol Genet Metab ; 128(3): 254-270, 2019 11.
Article in English | MEDLINE | ID: mdl-30583995

ABSTRACT

BACKGROUND: Lifestyle factors, including a low intake of carbohydrates, dieting, alcohol consumption, cigarette smoking and stress are some of the possible triggers of attacks in acute intermittent porphyria (AIP). The influence of lifestyle factors, including energy intake, diet and alcohol consumption on the biochemical disease activity in AIP and biochemical nutritional markers were examined. METHODS: A case-control study with 50 AIP cases and 50 controls matched for age, sex and place of residence was performed. Dietary intake was registered using a food diary in 46 matched pairs. Symptoms, alcohol intake, stress and other triggering factors of the last AIP attack were recorded on questionnaires. Porphyrin precursors, liver and kidney function markers, vitamins, diabetogenic hormones and other nutritional biomarkers were analyzed by routine methods. The Wilcoxon matched-pairs signed rank test was used to compare the cases vs. controls. The Spearman's rank correlation coefficient was used on the cases. RESULTS: Increasing total energy intake was negatively correlated with the biochemical disease activity. The intake of carbohydrates was lower than recommended, i.e., 40 and 39% of total energy intake in the AIP cases and controls, respectively. The plasma resistin level was significantly higher (p = .03) in the symptomatic than asymptomatic cases. Plasma insulin was lower in those with high porphobilinogen levels. The intake of sugar and candies were higher in the AIP cases with low U-delta aminolevulinic acid (ALA) levels (p = .04). Attacks were triggered by psychological stress (62%), physical strain (38%), food items (24%) and alcohol (32%) in the 34 symptomatic cases. Alcohol was used regularly by 88% of the cases (3.2 g ethanol/day) and 90% of the controls (6.3 g/day), but the intake was significantly lower in symptomatic than in asymptomatic cases (p = .045). CONCLUSION: A high intake of energy, sugar and candies and a higher insulin level were associated with a lower biochemical disease activity. The resistin level was higher in the symptomatic than the asymptomatic cases. AIP patients drink alcohol regularly, but the intake was significantly lower in the symptomatic cases. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT01617642.


Subject(s)
Diet , Life Style , Porphyria, Acute Intermittent/etiology , Acute Disease , Adult , Aged , Aminolevulinic Acid/urine , Biomarkers/blood , Case-Control Studies , Cross-Sectional Studies , Female , Humans , Insulin/blood , Male , Middle Aged , Norway , Porphobilinogen/blood , Porphyria, Acute Intermittent/diagnosis , Resistin/blood , Surveys and Questionnaires
6.
Mol Med ; 21: 487-95, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-26062020

ABSTRACT

Acute intermittent porphyria (AIP) is an autosomal-dominant hepatic disorder caused by the half-normal activity of hydroxymethylbilane (HMB) synthase. Symptomatic individuals experience life-threatening acute neurovisceral attacks that are precipitated by factors that induce the hepatic expression of 5-aminolevulinic acid synthase 1 (ALAS1), resulting in the marked accumulation of the putative neurotoxic porphyrin precursors 5-aminolevulinic acid (ALA) and porphobilinogen (PBG). Here, we provide the first detailed description of the biochemical and pathologic alterations in the explanted liver of an AIP patient who underwent orthotopic liver transplantation (OLT) due to untreatable and debilitating chronic attacks. After OLT, the recipient's plasma and urinary ALA and PBG rapidly normalized, and her attacks immediately stopped. In the explanted liver, (a) ALAS1 mRNA and activity were elevated approximately ~3- and 5-fold, and ALA and PBG concentrations were increased ~3- and 1,760-fold, respectively; (b) uroporphyrin III concentration was elevated; (c) microsomal heme content was sufficient, and representative cytochrome P450 activities were essentially normal; (d) HMB synthase activity was approximately half-normal (~42%); (e) iron concentration was slightly elevated; and (f) heme oxygenase I mRNA was increased approximately three-fold. Notable pathologic findings included nodular regenerative hyperplasia, previously not reported in AIP livers, and minimal iron deposition, despite the large number of hemin infusions received before OLT. These findings suggest that the neurovisceral symptoms of AIP are not associated with generalized hepatic heme deficiency and support the neurotoxicity of ALA and/or PBG. Additionally, they indicate that substrate inhibition of hepatic HMB synthase activity by PBG is not a pathogenic mechanism in acute attacks.


Subject(s)
5-Aminolevulinate Synthetase/genetics , Hydroxymethylbilane Synthase/biosynthesis , Liver/metabolism , Porphyria, Acute Intermittent/genetics , 5-Aminolevulinate Synthetase/biosynthesis , Adult , Aminolevulinic Acid/blood , Aminolevulinic Acid/urine , Female , Heme/metabolism , Humans , Hydroxymethylbilane Synthase/antagonists & inhibitors , Liver/pathology , Liver Transplantation , Porphobilinogen/blood , Porphobilinogen/urine , Porphyria, Acute Intermittent/enzymology , Porphyria, Acute Intermittent/pathology , RNA, Messenger/biosynthesis , Uroporphyrins/metabolism
7.
J Assoc Physicians India ; 62(5): 432-4, 2014 May.
Article in English | MEDLINE | ID: mdl-25438495

ABSTRACT

Acute intermittent porphyria is a rare disorder, characterised clinically by variable patterns of neurological and metabolic disturbances. We report a rare presentation with sudden onset painless bilateral reversible vision loss of cerebral origin along with a brief review of the underlying pathophysiology.


Subject(s)
Blindness/etiology , Porphyria, Acute Intermittent/complications , Porphyria, Acute Intermittent/diagnosis , Abdomen, Acute/etiology , Brain/pathology , Epilepsy, Tonic-Clonic/diagnosis , Epilepsy, Tonic-Clonic/etiology , Female , Follow-Up Studies , Humans , India , Magnetic Resonance Imaging , Papilledema/diagnosis , Papilledema/etiology , Porphobilinogen/blood , Young Adult
8.
Cell Transplant ; 23(9): 1153-62, 2014.
Article in English | MEDLINE | ID: mdl-23582197

ABSTRACT

Acute intermittent porphyria (AIP) is an autosomal dominant disorder characterized by insufficient porphobilinogen deaminase (PBGD) activity. When hepatic heme synthesis is induced, porphobilinogen (PBG) and 5-aminolevulinic acid (ALA) accumulate, which causes clinical symptoms such as abdominal pain, neuropathy, and psychiatric disturbances. Our aim was to investigate if hepatocyte transplantation can prevent or minimize the metabolic alterations in an AIP mouse model. We transplanted wild-type hepatocytes into PBGD-deficient mice and induced heme synthesis with phenobarbital. ALA and PBG concentrations in plasma were monitored, and the gene transcriptions of hepatic enzymes ALAS1, PBGD, and CYP2A5 were analyzed. Results were compared with controls and correlated to the percentage of engrafted hepatocytes. The accumulation of ALA and PBG was reduced by approximately 50% after the second hepatocyte transplantation. We detected no difference in mRNA levels of PBGD, ALAS1, or CYP2A5. Engraftment corresponding to 2.7% of the total hepatocyte mass was achieved following two hepatocyte transplantations. A lack of precursor production in less than 3% of the hepatocytes resulted in a 50% reduction in plasma precursor concentrations. This disproportional finding suggests that ALA and PBG produced in PBGD-deficient hepatocytes crossed cellular membranes and was metabolized by transplanted cells. The lack of effect on enzyme mRNA levels suggests that no significant efflux of heme from normal to PBGD-deficient hepatocytes takes place. Further studies are needed to establish the minimal number of engrafted hepatocytes needed to completely correct the metabolic abnormality in AIP and whether amelioration of the metabolic defect by partial restoration of PBGD enzyme activity translates into a clinical effect in human AIP.


Subject(s)
Hepatocytes/transplantation , Porphyria, Acute Intermittent/therapy , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Aminolevulinic Acid/blood , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Cells, Cultured , Cytochrome P450 Family 2 , Disease Models, Animal , Hepatocytes/cytology , Hepatocytes/metabolism , Hydroxymethylbilane Synthase/genetics , Hydroxymethylbilane Synthase/metabolism , Male , Mice , Mice, Inbred C57BL , Porphobilinogen/blood , RNA, Messenger/metabolism , Transplantation, Homologous
9.
J Pediatr Hematol Oncol ; 36(4): 281-92, 2014 May.
Article in English | MEDLINE | ID: mdl-23887025

ABSTRACT

Iron overload in hereditary hemochromatosis and hematologic malignancy has unfavorable effects on morbidity. Herein, 53 children (age 108.4±58.3 mo, 25 girls and 28 boys) with acute myeloblastic and lymphoblastic leukemia, who received 4 different chemotherapy protocols, were evaluated for iron overload throughout chemotherapy. Iron overload arose: (1) before chemotherapy, which was dependent on neither chemotherapy nor packed red blood cell transfusions and (2) after chemotherapy, which was dependent on the duration and nature of chemotherapy and partially on transfusion of packed red blood cells. Iron overload was documented in 75% of patients with a ferritin level >1000 ng/mL, by liver and heart magnetic resonance imaging, and they were administered iron-chelation therapy with success. Three of 10 radiologically iron-overloaded patients were heterozygous for H63D mutation. Aminolevulinic acid and porphobilinogen levels were normal. Light microscopic examination of the bone marrow revealed increased iron granules in erythroblasts, platelets, and megakaryocytes, iron-laden macrophages, free iron in the matrix, dyshematopoiesis, and apoptotic cells. Electron microscopic examination revealed iron-laden secondary lysosomes and autolysosomes in normoblasts and iron-laden primary granules in promyelocytes, irrelevant to the ferritin level, implying autophagia due to chemotherapy as a source of the excess iron. We think that iron overload, which is an important complication of acute leukemia, should be evaluated separately from "transfusion overload," and the management principles specific to leukemia should be implemented.


Subject(s)
Bone Marrow Cells , Bone Marrow , Hemochromatosis , Iron Chelating Agents/administration & dosage , Leukemia, Myeloid, Acute , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adolescent , Aminolevulinic Acid/blood , Bone Marrow/metabolism , Bone Marrow/ultrastructure , Bone Marrow Cells/metabolism , Bone Marrow Cells/ultrastructure , Child , Female , Ferritins/blood , Hemochromatosis/blood , Hemochromatosis/complications , Hemochromatosis/drug therapy , Hemochromatosis/genetics , Hemochromatosis/pathology , Humans , Iron/blood , Iron Chelating Agents/adverse effects , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Lysosomes/metabolism , Lysosomes/ultrastructure , Male , Mutation, Missense , Porphobilinogen/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
10.
Eur J Clin Invest ; 43(7): 727-39, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23650938

ABSTRACT

BACKGROUND: Acute intermittent porphyria (AIP) is an inherited disorder of haem synthesis wherein a partial deficiency of porphobilinogen (PBG) deaminase (PBGD) with other factors may give rise to biochemical and clinical manifestations of disease. The biochemical hallmarks of active AIP are relative hepatic haem deficiency and uncontrolled up-regulation of hepatic 5-aminolevulinic acid (ALA) synthase-1 (ALAS1) with over-production of ALA and PBG. The treatment of choice is intravenous haem, which restores the deficient regulatory haem pool of the liver and represses ALAS1. Recently, haem has been shown to influence circadian rhythms by controlling their negative feedback loops. We evaluated whether subjects with AIP exhibited an altered circadian profile. MATERIALS AND METHODS: Over a 21-h period, we measured levels of serum cortisol, melatonin, ALA, PBG and mRNA levels (in peripheral blood mononuclear cells) of selected clock-controlled genes and genes involved in haem synthesis in 10 Caucasian (European-American) women who were either postmenopausal or had been receiving female hormone therapy, six of whom have AIP and four do not and are considered controls. RESULTS: Four AIP subjects with biochemical activity exhibited higher levels of PBG and lower levels and dampened oscillation of serum cortisol, and a trend for lower levels of serum melatonin, than controls or AIP subjects without biochemical activity. Levels of clock-controlled gene mRNAs showed significant increases over baseline in all subjects at 5 a.m. and 11 p.m., whereas mRNA levels of ALAS1, ALAS2 and PBGD were increased only at 11 p.m. in subjects with active AIP. CONCLUSIONS: This pilot study provides evidence for disturbances of circadian markers in women with active AIP that may trigger or sustain some common clinical features of AIP.


Subject(s)
Circadian Rhythm/physiology , Porphyria, Acute Intermittent/metabolism , 5-Aminolevulinate Synthetase/blood , Adult , Aged , Case-Control Studies , Circadian Clocks/genetics , Female , Heme/biosynthesis , Heme/genetics , Humans , Hydrocortisone/blood , Melatonin/blood , Middle Aged , Pilot Projects , Porphobilinogen/blood , Porphyria, Acute Intermittent/genetics , RNA, Messenger/blood
11.
Biomed Chromatogr ; 27(2): 267-72, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23180457

ABSTRACT

Serum/plasma concentrations of 5-aminolaevulinic acid (ALA) and porphobilinogen (PBG) are elevated in patients with acute hepatic porphyrias, especially during acute attacks. Current assays require lengthy sample pre-treatment and derivatization steps. We report here a rapid, sensitive and specific hydrophilic interaction liquid chromatography-tandem mass spectrometry method for the direct and simultaneous quantitation of ALA and PBG in serum or plasma following simple protein precipitation with acetonitrile and centrifugation prior to injection. ALA and PBG were detected using selected reaction monitoring mode, following positive atmospheric pressure chemical ionization. Calibration was linear from 0.05 to 50 µmol/L for ALA and PBG. For both analytes, imprecision (relative standard deviation) was <13% and accuracy (percentage nominal concentrations) was between 92 and 107%. The method was successfully applied to the measurement of ALA and PBG in serum or plasma samples for the screening, biochemical diagnosis and treatment monitoring of patients with acute hepatic porphyrias.


Subject(s)
Aminolevulinic Acid/blood , Chromatography, Liquid/methods , Porphobilinogen/blood , Tandem Mass Spectrometry/methods , Aminolevulinic Acid/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Least-Squares Analysis , Porphobilinogen/chemistry , Porphyria, Acute Intermittent/blood , Reproducibility of Results , Sensitivity and Specificity
12.
J Chromatogr B Analyt Technol Biomed Life Sci ; 879(24): 2389-96, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21783436

ABSTRACT

Accurate determinations of 5-aminolevulinic acid (ALA) and porphobilinogen (PBG) in physiologic fluids are required for the diagnosis and therapeutic monitoring of acute porphyrias. Current colorimetric methods are insensitive and over-estimate ALA and PBG due to poor specificity, while LC-MS/MS methods increase sensitivity, but have limited matrices. An LC-MS/MS method was developed to simultaneously determine ALA and PBG concentrations in fluids or tissues which were solid phase extracted, butanol derivatized, and quantitated by selective reaction monitoring using (13)C(5), (15)N-ALA and 2,4-(13)C(2)-PBG internal standards. ALA was separated from interfering compounds on a reverse phase C8-column. For ALA and PBG, the matrix effects (87.3-105%) and process efficiencies (77.6-97.8% and 37.2-41.6%, respectively) were acceptable in plasma and urine matrices. The assay was highly sensitive for ALA and PBG (LLOQ=0.05 µM with 25 µL urine or 100 µL plasma), and required ∼4 h from extraction to results. ALA and PBG accuracy ranged from 88.2 to 110% (n=10); intra- and inter-assay coefficients of variations were <10% for urine and plasma. In clinical applications, patients with mutation-confirmed acute porphyrias had normal to slightly increased urinary ALA and PBG levels when asymptomatic, and high levels during acute attacks, which decreased with hemin therapy. In AIP mice, baseline ALA and PBG levels in urine, plasma, and liver were increased after phenobarbital induction 28-/63-, 42-/266-, and 13-/316-fold, respectively. This LC-MS/MS method is rapid, specific, highly sensitive, accurate, and simultaneously measures ALA and PBG in urine, plasma, and tissues permitting porphyria clinical diagnoses, therapeutic monitoring, and research.


Subject(s)
Aminolevulinic Acid/blood , Aminolevulinic Acid/urine , Chromatography, Liquid/methods , Porphobilinogen/blood , Porphobilinogen/urine , Porphyria, Acute Intermittent/diagnosis , Tandem Mass Spectrometry/methods , Adolescent , Adult , Animals , Child , Child, Preschool , Disease Models, Animal , Female , Humans , Male , Middle Aged , Porphyria, Acute Intermittent/blood , Porphyria, Acute Intermittent/urine , Sensitivity and Specificity , Young Adult
13.
Australas J Dermatol ; 52(2): 135-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21605099

ABSTRACT

A 35-year-old woman presented with skin fragility and photosensitivity with blisters affecting her face and hands. Other symptoms included intermittent headache, fatigue, abdominal pain and nausea. Porphyrin studies were markedly raised, with features consistent with hereditary coproporphyria (HCP). Despite strict precautions, symptoms remained significantly problematic. Regular haem arginate infusions of 3 mg/kg per day over 4 days on a monthly basis were commenced and resulted in significant improvement of the patient's symptoms and a reduction in urinary porphobilinogen. Although haem arginate infusion is known as a treatment for severe acute attacks of HCP, the effectiveness of regular infusions as maintenance therapy has not been established. This is the first report of effective symptom control correlating with normalization of biochemical markers in a patient receiving regular haem arginate infusions for the treatment of HCP.


Subject(s)
Arginine/therapeutic use , Coproporphyria, Hereditary/drug therapy , Heme/therapeutic use , Abdominal Pain/diagnosis , Abdominal Pain/drug therapy , Adult , Coproporphyria, Hereditary/diagnosis , Female , Humans , Photosensitivity Disorders/diagnosis , Photosensitivity Disorders/drug therapy , Porphobilinogen/blood , Porphobilinogen/urine , Severity of Illness Index , Treatment Outcome
14.
Transpl Int ; 23(6): e18-21, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20028496

ABSTRACT

We report two patients with acute intermittent porphyria (AIP) who were successfully treated with combined liver and kidney transplantation. Both had a very poor quality of life as a result of years of frequent acute porphyria symptoms, chronic peripheral neuropathy and renal failure requiring dialysis. After transplantation, clinical and biochemical signs of porphyria disappeared. The excretion pattern of porphyrin precursors normalized within the first day and plasma porphyrins returned to normal within a week. These and other recent cases have clarified previous concerns and have helped to formulate the indications for and the timing of transplantation in AIP.


Subject(s)
Kidney Transplantation , Liver Transplantation , Porphyria, Acute Intermittent/surgery , Female , Humans , Hydroxymethylbilane Synthase/blood , Middle Aged , Porphobilinogen/blood , Treatment Outcome , Young Adult
15.
Cell Mol Biol (Noisy-le-grand) ; 55(1): 66-71, 2009 Feb 16.
Article in English | MEDLINE | ID: mdl-19268004

ABSTRACT

Porphyrin precursors and porphyrins were measured in three patients with recurrent attacks of acute intermittent porphyria and end-stage renal disease (ESRD): two patients on hemodialysis and one on peritoneal dialysis. Plasma porphobilinogen (PBG) and porphyrins were considerably increased in the three patients. In a previous study, the mean urinary and plasma PBG/ALA ratio in biochemically active AIP patients with conserved renal function was 2.0 (normal 0.3) and plasma porphyrin levels were normal (< 10 nmol/L). In this study we show that the progression to ESRD was paralleled by an increase in urinary and plasma PBG/ALA ratio reaching levels above 6 and higher. Plasma porphyrin increased to levels above 1000 nmol/L causing cutaneous lesions resembling porphyria cutanea tarda. The porphyrin precursors were readily filtered by dialysis membranes but not the porphyrins. The development of kidney failure was a devastating complication in these AIP patients with chronic active disease, leading to unavoidable deterioration of peripheral veins, progression of peripheral neuropathy, dialysis treatment and secondary cutaneous lesions. The clinical course could not be reversed by medical treatment in any of the cases. Today, combined liver and kidney transplantation should be considered as a therapeutic option.


Subject(s)
Kidney Failure, Chronic/blood , Kidney Failure, Chronic/therapy , Porphyria, Acute Intermittent/blood , Porphyria, Acute Intermittent/therapy , Porphyrins/blood , Adult , Aminolevulinic Acid/blood , Aminolevulinic Acid/urine , Female , Humans , Kidney Failure, Chronic/urine , Middle Aged , Peritoneal Dialysis , Porphobilinogen/blood , Porphobilinogen/urine , Porphyria, Acute Intermittent/urine , Renal Dialysis
16.
Eur J Intern Med ; 20(2): 201-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19327613

ABSTRACT

BACKGROUND: Acute intermittent porphyria (AIP) is a metabolic disease affecting hepatic heme biosynthesis. The clinical course in overt disease is characterized by acute attacks of neurovisceral symptoms. Treatment is based on symptomatic relief together with carbohydrate loading and in more severe attacks heme therapy. During an acute attack the heme precursors porphobilinogen (PBG) and 5-aminolevulinic acid (ALA) are produced in high amounts by the liver and are found in high concentrations in plasma and urine. These metabolites represent the acute phase reactants confirming an ongoing attack and are used to evaluate therapeutic measures. The aim of this study was to measure PBG and ALA in plasma and urine during an acute attack and to match the biochemical pattern with the clinical and therapeutical course. METHODS: Three consecutive AIP patients were included during four acute attacks. Plasma PBG and ALA were measured by a LC-MS method and in urine by ion-exchange chromatography. The patients received symptomatic and glucose treatment at admission to hospital, and four days later, if necessary, heme therapy. RESULTS: In the three attacks that required heme therapy, plasma PBG concentrations had further increased after admission (p=0.01). In the patient that did not require heme therapy, plasma PBG had decreased after admission. CONCLUSIONS: Biochemical monitoring of an acute attack was more accurately reflected by plasma PBG than plasma ALA or urinary PBG and ALA. Glucose administration, in contrast to heme therapy, was not sufficient to achieve clinical and biochemical remission in the more serious attacks.


Subject(s)
Drug Monitoring/methods , Porphobilinogen/blood , Porphobilinogen/urine , Porphyria, Acute Intermittent/blood , Porphyria, Acute Intermittent/urine , Acute Disease , Adult , Aminolevulinic Acid/blood , Arginine/administration & dosage , Biomarkers/blood , Biomarkers/urine , Chromatography, Ion Exchange , Female , Glucose/administration & dosage , Heme/administration & dosage , Humans , Male , Porphyria, Acute Intermittent/drug therapy , Sensitivity and Specificity
17.
Clin Pharmacokinet ; 46(4): 335-49, 2007.
Article in English | MEDLINE | ID: mdl-17375984

ABSTRACT

BACKGROUND AND OBJECTIVE: Acute intermittent porphyria is an autosomal dominant disorder caused by deficient activity of the third enzyme in the haem biosynthetic pathway, porphobilinogen deaminase. It is characterised by acute, potentially life-threatening neurological attacks that are precipitated by various drugs, reproductive hormones and other factors. During acute attacks, the porphyrin precursors 5-aminolevulinic acid and porphobilinogen accumulate and are excreted at high concentrations in the urine. Current treatment is based on glucose loading and parenteral haem replenishment, which reduce the accumulation of 5-aminolevulinic acid and porphobilinogen. Recently, a new form of treatment based on porphobilinogen deaminase enzyme replacement therapy has been shown to be effective in an acute intermittent porphyria mouse model which, during phenobarbital (phenobarbitone) induction of haem biosynthesis, mimics the biochemical pattern of acute porphyric attacks. The objective of the present study was to investigate the safety, pharmacokinetics and pharmacodynamics of recombinant human porphobilinogen deaminase (P 9808), administered to healthy subjects and asymptomatic porphobilinogen deaminase-deficient subjects with high concentrations of porphobilinogen, the substrate of porphobilinogen deaminase. STUDY DESIGN: Forty individuals participated in this two-part study: 20 asymptomatic porphobilinogen deaminase-deficient subjects (both male and female) with > or =4 times the upper reference urinary porphobilinogen level, and 20 healthy male subjects. Four different doses of recombinant human porphobilinogen deaminase were studied (0.5, 1, 2 and 4 mg/kg bodyweight). Part A included 12 asymptomatic porphobilinogen deaminase-deficient subjects, and the enzyme was administered in an open-label, single-dose design. Part B included 20 asymptomatic porphobilinogen deaminase-deficient subjects and 20 healthy subjects. The same enzyme dosages were administered as divided doses every 12 hours for 4 consecutive days in a randomised, double-blinded, placebo-controlled design. The washout period between Parts A and B was 2 weeks. METHODS: The concentrations of recombinant human porphobilinogen deaminase and titres of antibodies against recombinant human porphobilinogen deaminase were analysed by ELISA. Plasma porphobilinogen and 5-aminolevulinic acid concentrations were analysed using a novel liquid chromatography-tandem mass spectrometry method. Urinary porphobilinogen, 5-aminolevulinic acid and porphyrin concentrations, as well as plasma porphyrin concentrations, were analysed using standard methods. The pharmacodynamic effect of the enzyme was studied through changes in plasma porphobilinogen concentrations. RESULTS: No serious adverse events were observed. Seven subjects (four healthy men and three asymptomatic porphobilinogen deaminase-deficient subjects) developed antibodies against recombinant human porphobilinogen deaminase but did not experience allergic manifestations. The mean elimination half-lives of the highest doses of recombinant human porphobilinogen deaminase ranged between 1.7 and 2.5 hours for both healthy men and asymptomatic porphobilinogen deaminase-deficient subjects. The area under the plasma concentration-time curve was proportional to the respective dose. In asymptomatic porphobilinogen deaminase-deficient subjects, plasma porphobilinogen concentrations decreased below measurable levels almost instantaneously after administration of any dose of the enzyme. The effect lasted for approximately 2 hours, after which the plasma porphobilinogen concentration slowly increased, reaching about 70% of the initial values 12 hours after administration. There was no effect on plasma 5-aminolevulinic acid concentrations, and there was a transitory increment in porphyrin concentrations. The corresponding concentrations of metabolites in the urine reflected the pattern observed in the plasma. CONCLUSIONS: The recombinant human porphobilinogen deaminase enzyme preparation was found to be safe to administer and effective for removal of the accumulated metabolite porphobilinogen from plasma and urine. The pharmacokinetic profile of recombinant human porphobilinogen deaminase showed dose proportionality, and the elimination half-life was about 2.0 hours for the two highest doses. Thus, clinical grounds were established for investigation of the therapeutic efficacy of the enzyme during periods of overt disease in patients with acute intermittent porphyria.


Subject(s)
Aminolevulinic Acid , Heterozygote , Hydroxymethylbilane Synthase/pharmacology , Porphobilinogen , Porphyria, Acute Intermittent/genetics , Adult , Aged , Aminolevulinic Acid/blood , Aminolevulinic Acid/urine , Antibody Formation/drug effects , Area Under Curve , Chromatography, Liquid , Dose-Response Relationship, Drug , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hydroxymethylbilane Synthase/administration & dosage , Hydroxymethylbilane Synthase/adverse effects , Hydroxymethylbilane Synthase/pharmacokinetics , Male , Mass Spectrometry , Middle Aged , Porphobilinogen/blood , Porphobilinogen/urine
18.
Clin Chem ; 52(4): 701-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16497943

ABSTRACT

BACKGROUND: The heme precursors porphobilinogen (PBG) and 5-aminolevulinic acid (ALA) accumulate during overt crises of acute intermittent porphyria (AIP), and high excretion of these metabolites often continues in the asymptomatic phase. METHODS: We measured concentrations of PBG and ALA and investigated the correlation between these metabolites in plasma and urine in 10 asymptomatic AIP carriers with high excretion and in 5 healthy individuals. We quantified plasma concentrations with an HPLC-mass spectrometric method and urine concentrations with ion-exchange chromatography. RESULTS: The mean (SD) plasma concentrations of PBG and ALA in the AIP carriers were 3.1 (1.0) and 1.7 (0.7) micromol/L, respectively. The mean 8-h urinary excretion amounts of PBG and ALA in the AIP carriers were 102 (25) and 56 (18) micromol, respectively, whereas the corresponding values for healthy individuals were 2.9 (0.7) and 9.3 (1.2) micromol. The correlations between PBG and ALA values in plasma and urine of the AIP carriers were 0.678 and 0.856, respectively. The mean PBG/ALA ratio was approximately 2.0 in both plasma and urine for the AIP carriers and 0.3 in urine for the healthy individuals. The renal clearance rates for PBG and ALA were 71 (15) and 70 (13) mL/min, respectively. CONCLUSIONS: The described HPLC-mass spectrometric method enabled characterization of variations in plasma PBG and ALA in AIP carriers during an 8-h period. The renal clearances were similar for both metabolites. This method could be used to monitor AIP patients during treatment.


Subject(s)
Aminolevulinic Acid/blood , Aminolevulinic Acid/urine , Heterozygote , Porphobilinogen/blood , Porphobilinogen/urine , Porphyria, Acute Intermittent/genetics , Adult , Chromatography, High Pressure Liquid , Female , Humans , Male , Mass Spectrometry , Middle Aged
19.
Br J Dermatol ; 149(6): 1266-9, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14674906

ABSTRACT

BACKGROUND: Porphyrinogens are the obligate intracellular precursors of haem. These compounds are, however, unstable and are easily oxidized to the corresponding porphyrins, which are the form in which they are usually measured in the laboratory. A substantial enterohepatic cycling of porphyrins has been shown. Administration of oral activated charcoal, by interrupting this cycle, may reduce plasma and urine porphyrin levels in patients with some forms of porphyria. The effect of charcoal in subjects with variegate porphyria (VP) has not been reported. OBJECTIVES: To determine the clinical and biochemical effects of the administration of oral activated charcoal in patients with VP. METHODS: Oral activated charcoal was administered to eight subjects with VP. Clinical activity was assessed by skin lesion counts fortnightly for 6 weeks, 6 weeks after cessation of therapy, and during a subsequent 6-week control period during which no charcoal was taken. Urine and plasma porphyrins and urine precursors were measured by standard techniques. RESULTS: Treatment resulted in a significant increase in skin disease, urine and plasma porphyrins. CONCLUSIONS: Oral charcoal administration results in a paradoxical aggravation of VP, suggesting a complex and as yet undefined interaction of hepatic porphyrin metabolism and bowel porphyrin reabsorption. Oral sorbents should not be prescribed to subjects with VP.


Subject(s)
Charcoal/administration & dosage , Porphyrias, Hepatic/drug therapy , Administration, Oral , Adolescent , Adult , Aminolevulinic Acid/blood , Aminolevulinic Acid/urine , Analysis of Variance , Female , Humans , Male , Porphobilinogen/blood , Porphobilinogen/urine , Porphyrias, Hepatic/metabolism , Porphyrias, Hepatic/pathology , Porphyrins/blood , Porphyrins/urine , Prospective Studies , South Africa , Treatment Failure
20.
Photochem Photobiol ; 78(1): 82-7, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12929753

ABSTRACT

Photodynamic therapy (PDT) based on the photosensitive protoporphyrin IX (PpIX) may prevent restenosis after transluminal angioplasty. PpIX is synthesized in mitochondria, which differ in number and activity among various tissues. Therefore, we questioned whether the course of PpIX concentration after systemic aminolaevulinic acid (ALA) administration differed among various arteries. ALA was administered intravenously (200 mg/kg) to male Wistar rats (n = 21). At varying time intervals (0, 1, 2, 3, 6, 12 and 24 h) both central and peripheral arteries were isolated and homogenized, and the concentration of the various heme intermediates was determined by a fluorometric extraction method. The maximal PpIX concentration was more than two-fold higher in peripheral arteries (20.49 +/- 3.0 to 24.0 +/- 7.5 pmol/mg protein) than in central arteries (0-9.46 +/- 0.01 pmol/mg protein) (P < 0.004). However, the amount of citrate synthase, reflecting the mitochondrial mass, was lower (0.14-0.61 and 1.87-2.32 U/mg protein, respectively). Apparently, the level of PpIX cannot simply be explained by the mitochondrial content of the arteries. The time interval of maximal PpIX accumulation was similar in peripheral and central arteries (2 h and 27 min vs. 2 h and 8 min) (P = 0.13). Thus, if the efficacy of PDT in vivo is directly related to the tissue concentration of PpIX, more effect can be expected in peripheral arteries than in central arteries.


Subject(s)
Aminolevulinic Acid/pharmacology , Arteries/metabolism , Protoporphyrins/pharmacokinetics , Aminolevulinic Acid/analysis , Aminolevulinic Acid/blood , Animals , Arteries/ultrastructure , Male , Mitochondria/chemistry , Porphobilinogen/analysis , Porphobilinogen/blood , Protoporphyrins/blood , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...