Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 98
1.
Dtsch Med Wochenschr ; 146(15): 955-958, 2021 08.
Article De | MEDLINE | ID: mdl-34344029

Givosiran is a small synthetic double-stranded siRNA (small interfering RNA) conjugated with N-acetyl-galactosamine (GalNAc) for specific hepatocyte targeting via the asialoglycoprotein receptor. A prospective randomized multicenter study (Envision) demonstrated the clinical efficacy of monthly subcutaneous injection of Givosiran for the prevention of attacks of acute hepatic porphyria (AHP). This leads to highly selective transcriptional inhibition of the key hepatic enzyme, aminolaevulinate synthase 1, that is overexpressed in AHP. The success of the Envision study has led to the approval of Givosiran in the US and Europe for the treatment of severe AHP. This innovative guided siRNA therapy has opened up the possibility to selectively inhibit the expression of any hepatocyte gene whose overexpression that causes pathology, which can be considered a milestone development in hepatology. However, currently this treatment with givosiran is very costly. Moreover, since some patients experience worsening of kidney function and elevated aminotransferases, monthly monitoring of these parameters is necessary in the first half year of treatment.


Acetylgalactosamine/analogs & derivatives , Porphobilinogen Synthase/deficiency , Porphyrias, Hepatic/drug therapy , Pyrrolidines , RNA, Small Interfering , RNAi Therapeutics , 5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/therapeutic use , Humans , Porphobilinogen Synthase/metabolism , Porphyrias, Hepatic/metabolism , Porphyrias, Hepatic/physiopathology , Porphyrias, Hepatic/prevention & control , Pyrrolidines/administration & dosage , Pyrrolidines/therapeutic use , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , RNA, Small Interfering/ultrastructure , Randomized Controlled Trials as Topic
2.
Ann Hematol ; 100(7): 1685-1693, 2021 Jul.
Article En | MEDLINE | ID: mdl-34050373

Givosiran is a novel approach to treat patients with acute intermittent porphyrias (AIP) by silencing of ∂-ALA-synthase 1, the first enzyme of heme biosynthesis in the liver. We included two patients in the Envision study who responded clinically well to this treatment. However, in both patients, therapy had to be discontinued because of severe adverse effects: One patient (A) developed local injection reactions which continued to spread all over her body with increasing number of injections and eventually caused a severe systemic allergic reaction. Patient B was hospitalized because of a fulminant pancreatitis. Searching for possible causes, we also measured the patients plasma homocysteine (Hcy) levels in fluoride-containing collection tubes: by LC-MS/MS unexpectedly, plasma Hcy levels were 100 and 200 in patient A and between 100 and 400 µmol/l in patient B. Searching for germline mutations in 10 genes that are relevant for homocysteine metabolism only revealed hetero- and homozygous polymorphisms in the MTHFR gene. Alternatively, an acquired inhibition of cystathionine-beta-synthase which is important for homocysteine metabolism could explain the plasma homocysteine increase. This enzyme is heme-dependent: when we gave heme arginate to our patients, Hcy levels rapidly dropped. Hence, we conclude that inhibition of ∂-ALA-synthase 1 by givosiran causes a drop of free heme in the hepatocyte and therefore the excessive increase of plasma homocysteine. Hyperhomocysteinemia may contribute to the adverse effects seen in givosiran-treated patients which may be due to protein-N-homocysteinylation.


5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/analogs & derivatives , Heme/deficiency , Hyperhomocysteinemia/etiology , Porphyria, Acute Intermittent/drug therapy , Pyrrolidines/therapeutic use , Acetylgalactosamine/adverse effects , Acetylgalactosamine/therapeutic use , Adult , Arginine/therapeutic use , Colitis/etiology , Colon, Sigmoid/pathology , Controlled Clinical Trials as Topic , Drug Hypersensitivity/etiology , Female , Fibrosis , Heme/analysis , Heme/therapeutic use , Hepatocytes/drug effects , Hepatocytes/metabolism , High-Throughput Nucleotide Sequencing , Homocysteine/metabolism , Humans , Hydroxymethylbilane Synthase/blood , Hydroxymethylbilane Synthase/genetics , Male , Models, Biological , Pancreatitis/etiology , Porphyria, Acute Intermittent/blood , Porphyria, Acute Intermittent/complications , Porphyria, Acute Intermittent/genetics , Pyrrolidines/adverse effects
3.
Drug Metab Dispos ; 49(7): 572-580, 2021 07.
Article En | MEDLINE | ID: mdl-33941543

Givosiran is an N-acetylgalactosamine-conjugated RNA interference therapeutic that targets 5'-aminolevulinate synthase 1 mRNA in the liver and is currently marketed for the treatment of acute hepatic porphyria. Herein, nonclinical pharmacokinetics and absorption, distribution, metabolism, and excretion properties of givosiran were characterized. Givosiran was completely absorbed after subcutaneous administration with relatively short plasma elimination half-life (t1/2; less than 4 hours). Plasma exposure increased approximately dose proportionally with no accumulation after repeat doses. Plasma protein binding was concentration dependent across all species tested and was around 90% at clinically relevant concentration in human. Givosiran predominantly distributed to the liver by asialoglycoprotein receptor-mediated uptake, and the t1/2 in the liver was significantly longer (∼1 week). Givosiran was metabolized by nucleases, not cytochrome P450 (P450) isozymes, across species with no human unique metabolites. Givosiran metabolized to form one primary active metabolite with the loss of one nucleotide from the 3' end of antisense strand, AS(N-1)3' givosiran, which was equipotent to givosiran. Renal and fecal excretion were minor routes of elimination of givosiran as approximately 10% and 16% of the dose was recovered intact in excreta of rats and monkeys, respectively. Givosiran is not a substrate, inhibitor, or inducer of P450 isozymes, and it is not a substrate or inhibitor of uptake and most efflux transporters. Thus, givosiran has a low potential of mediating drug-drug interactions involving P450 isozymes and drug transporters. SIGNIFICANCE STATEMENT: Nonclinical pharmacokinetics and absorption, distribution, metabolism, and excretion (ADME) properties of givosiran were characterized. Givosiran shows similar pharmacokinetics and ADME properties across rats and monkeys in vivo and across human and animal matrices in vitro. Subcutaneous administration results in adequate exposure of givosiran to the target organ (liver). These studies support the interpretation of toxicology studies, help characterize the disposition of givosiran in humans, and support the clinical use of givosiran for the treatment of acute hepatic porphyria.


Acetylgalactosamine/analogs & derivatives , Pyrrolidines/pharmacokinetics , 5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/pharmacokinetics , Animals , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Female , Half-Life , Injections, Subcutaneous , Intestinal Elimination , Macaca fascicularis , Male , Models, Animal , Porphobilinogen Synthase/deficiency , Porphyrias, Hepatic/drug therapy , Pyrrolidines/administration & dosage , Rats , Renal Elimination , Tissue Distribution
4.
Sci Rep ; 11(1): 8455, 2021 04 19.
Article En | MEDLINE | ID: mdl-33875732

Filarial infections affect millions of individuals and are responsible for some notorious disabilities. Current treatment options involve repeated mass drug administrations, which have been met with several challenges despite some successes. Administration of doxycycline, an anti-Wolbachia agent, has shown clinical effectiveness but has several limitations, including long treatment durations and contraindications. We describe the use of an in silico drug repurposing approach to screening a library of over 3200 FDA-approved medications against the filarial endosymbiont, Wolbachia. We target the enzyme which catalyzes the first step of heme biosynthesis in the Wolbachia. This presents an opportunity to inhibit heme synthesis, which leads to depriving the filarial worm of heme, resulting in a subsequent macrofilaricidal effect. High throughput virtual screening, molecular docking and molecular simulations with binding energy calculations led to the identification of paritaprevir and nilotinib as potential anti-Wolbachia agents. Having higher binding affinities to the catalytic pocket than the natural substrate, these drugs have the structural potential to bind and engage active site residues of the wolbachia 5'-Aminolevulinic Acid Synthase. We hereby propose paritaprevir and nilotinib for experimental validations as anti-Wolbachia agents.


5-Aminolevulinate Synthetase/antagonists & inhibitors , Computer Simulation , Cyclopropanes/pharmacology , Drug Repositioning/methods , Enzyme Inhibitors/pharmacology , Lactams, Macrocyclic/pharmacology , Proline/analogs & derivatives , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Wolbachia/drug effects , Amino Acid Sequence , Humans , Proline/pharmacology , Sequence Homology , Wolbachia/enzymology , Wolbachia/growth & development
5.
Drugs ; 81(7): 841-848, 2021 May.
Article En | MEDLINE | ID: mdl-33871817

Givosiran (Givlaari®) is an δ-aminolevulinic acid synthase 1 (ALAS1)-directed small interfering RNA (siRNA) approved for the treatment of acute hepatic porphyria (AHP). In the phase 3 ENVISION trial, givosiran significantly reduced the annualized rate of composite porphyria attacks (i.e. attacks requiring hospitalization, urgent healthcare visit or intravenous hemin administration at home) compared with placebo in patients with recurrent acute intermittent porphyria (the most common type of AHP) attacks. Givosiran also improved several other outcomes, including hemin use and pain (the cardinal symptom of AHP). While generally well tolerated with an acceptable safety profile, the drug may increase the risk of hepatic and kidney adverse events. Givosiran offers the convenience of once-monthly subcutaneous administration. Available evidence indicates that givosiran is an important newer therapeutic option for patients with AHP and severe recurrent attacks.


Acetylgalactosamine/analogs & derivatives , Porphobilinogen Synthase/deficiency , Porphyria, Acute Intermittent/drug therapy , Porphyrias, Hepatic/drug therapy , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , 5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/adverse effects , Acetylgalactosamine/pharmacokinetics , Acetylgalactosamine/pharmacology , Acetylgalactosamine/therapeutic use , Acute Kidney Injury/chemically induced , Chemical and Drug Induced Liver Injury , Drug Interactions , Hemin/administration & dosage , Hospitalization , Humans , Pain/drug therapy , Pain/etiology , Porphyria, Acute Intermittent/complications , Porphyrias, Hepatic/complications , Pyrrolidines/adverse effects , Pyrrolidines/pharmacokinetics , RNA, Small Interfering , Randomized Controlled Trials as Topic , Severity of Illness Index
6.
Article De | MEDLINE | ID: mdl-33588480

Following the accidental feeding of a compound feed containing the coccidiostat nicarbacin in layer breeder flocks (Lohmann Brown Classic), the birds displayed distinct clinical signs within a few hours. Mortality increased during the following 5 days, whereas laying performance and hatching rate of eggs during this period decreased markedly. Egg shell discoloration was observed as early as during the first day. As a consequence, an association between feeding of the coccidiostat nicarbacin and the observed symptoms was assumed. Recent studies indicate that Nicarbacin reduces the activity of aminolevulinic acid synthase type 1 (ALAS 1), which is responsible for the synthesis of protoporphyrin IX in the shell gland as main compound of brown egg shells. Reduced laying performance and increased mortality was likely due to nicarbacin-induced deregulated body temperature homeostasis and concomitant imbalances in acid-base status of the animals. The case reveals that the accidental feeding of nicarbacin to non-target animals such as laying hens and their parents may result in acute clinical symptoms. This highlights the necessity of appropriate care in handling feed additives and their premixes for specific non-target animals and should sensitize farmers and veterinarians.


Chickens/physiology , Coccidiostats/administration & dosage , Nicarbazin/administration & dosage , Oviposition/drug effects , 5-Aminolevulinate Synthetase/antagonists & inhibitors , Acid-Base Equilibrium/drug effects , Animal Feed , Animals , Body Temperature/drug effects , Coccidiostats/adverse effects , Egg Shell/drug effects , Eggs/standards , Female , Nicarbazin/adverse effects , Protoporphyrins/biosynthesis
7.
Blood ; 136(21): 2457-2468, 2020 11 19.
Article En | MEDLINE | ID: mdl-32678895

Congenital erythropoietic porphyria (CEP) is an inborn error of heme synthesis resulting from uroporphyrinogen III synthase (UROS) deficiency and the accumulation of nonphysiological porphyrin isomer I metabolites. Clinical features are heterogeneous among patients with CEP but usually combine skin photosensitivity and chronic hemolytic anemia, the severity of which is related to porphyrin overload. Therapeutic options include symptomatic strategies only and are unsatisfactory. One promising approach to treating CEP is to reduce the erythroid production of porphyrins through substrate reduction therapy by inhibiting 5-aminolevulinate synthase 2 (ALAS2), the first and rate-limiting enzyme in the heme biosynthetic pathway. We efficiently reduced porphyrin accumulation after RNA interference-mediated downregulation of ALAS2 in human erythroid cellular models of CEP disease. Taking advantage of the physiological iron-dependent posttranscriptional regulation of ALAS2, we evaluated whether iron chelation with deferiprone could decrease ALAS2 expression and subsequent porphyrin production in vitro and in vivo in a CEP murine model. Treatment with deferiprone of UROS-deficient erythroid cell lines and peripheral blood CD34+-derived erythroid cultures from a patient with CEP inhibited iron-dependent protein ALAS2 and iron-responsive element-binding protein 2 expression and reduced porphyrin production. Furthermore, porphyrin accumulation progressively decreased in red blood cells and urine, and skin photosensitivity in CEP mice treated with deferiprone (1 or 3 mg/mL in drinking water) for 26 weeks was reversed. Hemolysis and iron overload improved upon iron chelation with full correction of anemia in CEP mice treated at the highest dose of deferiprone. Our findings highlight, in both mouse and human models, the therapeutic potential of iron restriction to modulate the phenotype in CEP.


Anemia, Hemolytic/drug therapy , Deferiprone/therapeutic use , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Photosensitivity Disorders/drug therapy , Porphyria, Erythropoietic/drug therapy , 5-Aminolevulinate Synthetase/antagonists & inhibitors , 5-Aminolevulinate Synthetase/biosynthesis , 5-Aminolevulinate Synthetase/genetics , Adult , Anemia, Hemolytic/etiology , Animals , CRISPR-Cas Systems , Cell Line , Cell Line, Tumor , Disease Models, Animal , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Female , Gene Knock-In Techniques , Humans , Iron/metabolism , Iron Overload/etiology , Leukemia, Erythroblastic, Acute/pathology , Mice , Peripheral Blood Stem Cells/drug effects , Peripheral Blood Stem Cells/metabolism , Photosensitivity Disorders/etiology , Porphyria, Acute Intermittent/metabolism , Porphyria, Erythropoietic/complications , Porphyrins/biosynthesis , RNA Interference , RNA, Small Interfering/pharmacology
8.
Drugs ; 80(3): 335-339, 2020 Feb.
Article En | MEDLINE | ID: mdl-32034693

Givosiran (Givlaari™) is an aminolevulinate synthase 1 (ALAS1)-directed small interfering RNA (siRNA) covalently linked to a ligand to enable specific delivery of the siRNA to hepatocytes. This results in downregulation of ALAS1 mRNA and prevents accumulation of neurotoxic δ-aminolevulinic acid and porphobilinogen levels that are associated with acute porphyria attacks. Givosiran is being developed by Alnylam Pharmaceuticals for the treatment of acute hepatic porphyria (AHP). In November 2019, givosiran was approved in the USA for the treatment of adults with AHP based on the positive results from the multinational, phase III ENVISION trial. In the EU, givosiran received a positive opinion in January 2020 for the treatment of AHP in adults and adolescents aged 12 years and older. This article summarizes the milestones in the development of givosiran leading to this first approval for the treatment of adults with AHP.


5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/analogs & derivatives , Drug Approval , Enzyme Inhibitors/pharmacology , Porphobilinogen Synthase/deficiency , Porphyrias, Hepatic/drug therapy , Pyrrolidines/pharmacology , 5-Aminolevulinate Synthetase/metabolism , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/pharmacology , Enzyme Inhibitors/administration & dosage , Humans , Porphobilinogen Synthase/metabolism , Porphyrias, Hepatic/metabolism , Pyrrolidines/administration & dosage , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism
9.
Med Hypotheses ; 131: 109314, 2019 Oct.
Article En | MEDLINE | ID: mdl-31443750

Metabolic targeting of liver 5-aminolevulinate synthase (5-ALAS) by inhibition of heme utilisation by tryptophan (Trp) 2,3-dioxygenase (TDO) or the use of tryptophan is proposed as a therapy of acute hepatic porphyrias. 5-ALAS, the rate-limiting enzyme of heme biosynthesis, is under negative feedback control by a small regulatory heme pool in the hepatic cytosol. Acute porphyric attacks, precipitated by fasting, certain hormones and some drugs, involve induction of 5-ALAS secondarily to depletion of the above pool, and the resultant elevation of 5-ALA levels initiates the abdominal and neurological symptoms of attacks. By utilising the regulatory heme, cytosolic TDO undermines the feedback control, thus allowing 5-ALAS induction to occur, e.g. upon glucocorticoid induction of TDO during fasting (starvation) and exogenous glucocorticoid administration. Currently, glucose therapy is the preferred strategy for reversing moderate attacks induced by fasting (calorie restriction), with more severe attacks being treated by intravenous heme preparations. Reversal of fasting-induced attacks by glucose is explained by the previously demonstrated reversal of increased heme utilisation by TDO. Inhibitors of this utilisation are therefore potential therapeutic targets in acute attacks and also for maintenance of a symptomless state. Existing TDO inhibitors other than glucose include allopurinol, nicotinamide and recently developed potent inhibitors such as LM10 used in cancer therapy. Based on studies in rats, the hypothesis predicts that the safety or otherwise of drugs in the hepatic porphyrias is determined by their ability to inhibit TDO utilisation of heme under basal conditions or after glucocorticoid induction or heme activation of TDO, in parallel with reciprocal changes in 5-ALAS induction. Tryptophan is also proposed as a potential therapy of acute attacks either alone or as an adjunct to the recently proposed 5-ALAS1 gene silencing. Trp increases heme biosynthesis by enhancing 5-ALA dehydratase activity and, based on a Trp-5-ALA model presented herein, Trp offers several advantages over heme therapy, namely rapid conversion of 5-ALA into heme, a greatly enhanced heme availability, a near complete inhibition of 5-ALAS induction, assumed rapid clearance of 5-ALA and hence accelerated resolution of symptoms of attacks, and finally provision of the neuroprotective metabolite kynurenic acid to neutralise the neurological symptoms. The hypothesis also addresses heme regulation in species lacking the TDO free apoenzyme and its glucocorticoid induction mechanism and proposes detailed assessment of heme biosynthesis in these species. Detailed proposals for testing the hypothesis are presented.


5-Aminolevulinate Synthetase/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Heme/metabolism , Molecular Targeted Therapy , Porphyrias, Hepatic/drug therapy , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan/therapeutic use , 5-Aminolevulinate Synthetase/genetics , Allopurinol/pharmacology , Allopurinol/therapeutic use , Animals , Enzyme Induction/drug effects , Fasting/metabolism , Feedback, Physiological , Gene Silencing , Glucose/metabolism , Glucose/therapeutic use , Guinea Pigs , Heme/therapeutic use , Humans , Kynurenine/metabolism , Liver/drug effects , Liver/enzymology , Models, Biological , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Porphyrias, Hepatic/chemically induced , Porphyrias, Hepatic/genetics , Porphyrias, Hepatic/metabolism , Rodentia , Species Specificity , Tryptophan/adverse effects , Tryptophan/pharmacology
10.
Mol Genet Metab ; 128(3): 309-313, 2019 11.
Article En | MEDLINE | ID: mdl-31395332

Erythropoietic protoporphyria (EPP), the most common porphyria of childhood and the third most common porphyria of adulthood, is characterized clinically by painful, non-blistering cutaneous photosensitivity. Two distinct inheritance patterns involving mutations affecting genes that encode enzymes of the heme biosynthetic pathway underlie the clinical phenotype. Aminolevulinic acid synthase 2 (ALAS2), the rate limiting enzyme of the heme pathway in the erythron, is a therapeutic target in EPP because inhibiting enzyme function would reduce downstream production of protoporphyrin IX (PPIX), preventing accumulation of the toxic molecule and thereby ameliorating symptoms. Isoniazid (INH) is widely used for treatment of latent and active M. tuberculosis (TB). Sideroblastic anemia is observed in some patients taking INH, and studies have shown that this process is a consequence of inhibition of ALAS2 by INH. Based on these observations, we postulated that INH might have therapeutic activity in patients with EPP. We challenged this hypothesis in a murine model of EPP and showed that, after 4 weeks of treatment with INH, both plasma PPIX and hepatic PPIX were significantly reduced. Next, we tested the effect of INH on patients with EPP. After eight weeks, no significant difference in plasma or red cell PPIX was observed among the 15 patients enrolled in the study. These results demonstrate that while INH can lower PPIX in an animal model of EPP, the standard dose used to treat TB is insufficient to affect levels in humans.


5-Aminolevulinate Synthetase/antagonists & inhibitors , Isoniazid/therapeutic use , Protoporphyria, Erythropoietic/drug therapy , Protoporphyrins/blood , Anemia, Sideroblastic/enzymology , Animals , Disease Models, Animal , Female , Humans , Liver/chemistry , Liver/drug effects , Male , Mice , Pilot Projects , Proof of Concept Study , Protoporphyria, Erythropoietic/genetics , Protoporphyrins/metabolism
11.
J Hepatol ; 71(2): 422-433, 2019 08.
Article En | MEDLINE | ID: mdl-31102718

Porphyrias are rare inherited disorders caused by specific enzyme dysfunctions in the haem synthesis pathway, which result in abnormal accumulation of specific pathway intermediates. The symptoms depend upon the chemical characteristics of these substances. Porphyrins are photoreactive and cause photocutaneous lesions on sunlight-exposed areas, whereas accumulation of porphyrin precursors is related to acute neurovisceral attacks. Current therapies are suboptimal and mostly address symptoms rather than underlying disease mechanisms. Advances in the understanding of the molecular bases and pathogenesis of porphyrias have paved the way for the development of new therapeutic strategies. In this Clinical Trial Watch we summarise the basic principles of these emerging approaches and what is currently known about their application to porphyrias of hepatic origin or with hepatic involvement.


Acetylgalactosamine/analogs & derivatives , Bone Marrow Transplantation/methods , Cholestyramine Resin/therapeutic use , Genetic Therapy/methods , Liver Transplantation/methods , Porphyrias, Hepatic/drug therapy , Porphyrias, Hepatic/surgery , Pyrrolidines/therapeutic use , Receptor, Melanocortin, Type 1/agonists , alpha-MSH/analogs & derivatives , 5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/pharmacology , Acetylgalactosamine/therapeutic use , Heme/biosynthesis , Humans , Liver/metabolism , Porphyrias, Hepatic/classification , Porphyrias, Hepatic/pathology , Porphyrins/metabolism , Pyrrolidines/pharmacology , alpha-MSH/therapeutic use
12.
N Engl J Med ; 380(6): 549-558, 2019 02 07.
Article En | MEDLINE | ID: mdl-30726693

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 .).


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
13.
Clin J Gastroenterol ; 10(5): 452-458, 2017 Oct.
Article En | MEDLINE | ID: mdl-28676994

A 21-year-old Japanese man was admitted to our hospital because of severe abdominal pain and jaundice. He had been suffering from abdominal pain attacks and liver dysfunction since 18 years of age. Liver histology showed amorphous brown deposits in the sinusoidal space and significant periportal fibrosis without apparent hepatitis. Increased protoporphyrin in serum and feces and ferrochelatase gene mutation confirmed the final diagnosis of erythropoietic protoporphyria (EPP). Since ursodeoxycholic acid (UDCA) intake and glucose infusion are insufficient to ameliorate jaundice and abdominal attacks, cimetidine and lactulose were added in order to suppress hepatic delta-aminolevulinic acid synthase and limit re-absorption of protoporphyrin, respectively. Afterwards, the jaundice, liver dysfunction and abdominal symptoms improved and UDCA, cimetidine, and lactulose administration was continued. A repeat biopsy at 1.5 years after adding cimetidine/lactulose revealed marked attenuation of periportal fibrosis and protoporphyrin deposits. As far as we know, this is the first demonstration of histological improvement of EPP-induced liver abnormalities due to persistent cimetidine/lactulose administration. These treatments may be useful for EPP-related liver injury.


Cimetidine/therapeutic use , Gastrointestinal Agents/therapeutic use , Lactulose/therapeutic use , Liver Cirrhosis/drug therapy , Liver Cirrhosis/etiology , Protoporphyria, Erythropoietic/complications , 5-Aminolevulinate Synthetase/antagonists & inhibitors , Humans , Liver/enzymology , Liver/physiopathology , Male , Protoporphyria, Erythropoietic/physiopathology , Protoporphyrins/antagonists & inhibitors , Young Adult
14.
Biochim Biophys Acta Mol Basis Dis ; 1863(2): 428-439, 2017 02.
Article En | MEDLINE | ID: mdl-27838491

Mutations in the C-terminus of human erythroid 5-aminolevulinate synthase (hALAS2), a pyridoxal 5'-phosphate (PLP)-dependent enzyme, are associated with two different blood disorders, X-linked sideroblastic anemia (XLSA) and X-linked protoporphyria (XLPP). XLSA-causing mutations yield hALAS2 variants with decreased activity, while XLPP-causing mutations result in a gain-of-function of hALAS2. There are no specific treatments for XLPP. Isonicotinic acid hydrazide (isoniazid, INH), an antituberculosis agent, can cause sideroblastic anemia as a side-effect, by limiting PLP availability to hALAS2, via inhibition of pyridoxal kinase or reaction with pyridoxal to form pyridoxal isonicotinoyl hydrazone. We hypothesized that INH also binds and directly inhibits hALAS2. Using fluorescence-activated cell sorting and confocal fluorescence microscopy, we demonstrate that INH reduces protoporphyrin IX levels in HeLa cells expressing either wild-type hALAS2 or XLPP variants. In addition, PLP and pyridoxamine 5'-phosphate (PMP) reversed the cellular inhibition of hALAS2 activity by INH. Steady-state kinetic analyses with purified hALAS2 indicated that INH directly inhibits the enzyme, noncompetitively or uncompetitively, with an apparent Ki of 1.2µM. Circular dichroism spectroscopy revealed that INH triggered tertiary structural changes in hALAS2 that altered the microenvironment of the PLP cofactor and hampered the association of PLP with apo-hALAS2. Treatment of four XLPP patients with INH (5mg·kg-1·day-1) over a six-month period was well tolerated but without statistically significant modification of PPIX levels. These results, taken together, permit us to further an INH inhibition kinetic mechanism for ALAS, which suggests the possible use of INH-derived drugs in treating patients with XLPP and potentially other protoporphyrin-accumulating porphyrias.


5-Aminolevulinate Synthetase/deficiency , Enzyme Inhibitors/pharmacology , Genetic Diseases, X-Linked/drug therapy , Isoniazid/pharmacology , Protoporphyria, Erythropoietic/drug therapy , 5-Aminolevulinate Synthetase/antagonists & inhibitors , 5-Aminolevulinate Synthetase/blood , 5-Aminolevulinate Synthetase/chemistry , 5-Aminolevulinate Synthetase/metabolism , Anemia, Sideroblastic/enzymology , Enzyme Inhibitors/therapeutic use , Genetic Diseases, X-Linked/blood , Genetic Diseases, X-Linked/enzymology , HeLa Cells , Humans , Isoniazid/therapeutic use , Protein Binding/drug effects , Protein Structure, Tertiary/drug effects , Protoporphyria, Erythropoietic/blood , Protoporphyria, Erythropoietic/enzymology , Protoporphyrins/blood , Pyridoxal Phosphate/metabolism , Pyridoxine/pharmacology , Vitamin B Complex/pharmacology
15.
Biochem Biophys Res Commun ; 454(1): 102-8, 2014 Nov 07.
Article En | MEDLINE | ID: mdl-25450364

Congenital sideroblastic anemia (CSA) is a hereditary disorder characterized by microcytic anemia and bone marrow sideroblasts. The most common form of CSA is attributed to mutations in the X-linked gene 5-aminolevulinic acid synthase 2 (ALAS2). ALAS2 is a mitochondrial enzyme, which utilizes glycine and succinyl-CoA to form 5-aminolevulinic acid (ALA), a crucial precursor in heme synthesis. Therefore, ALA supplementation could be an effective therapeutic strategy to restore heme synthesis in CSA caused by ALAS2 defects. In a preclinical study, we examined the effects of ALA in human erythroid cells, including K562 cells and human induced pluripotent stem cell-derived erythroid progenitor (HiDEP) cells. ALA treatment resulted in significant dose-dependent accumulation of heme in the K562 cell line. Concomitantly, the treatment substantially induced erythroid differentiation as assessed using benzidine staining. Quantitative reverse transcription polymerase chain reaction (RT-PCR) analysis confirmed significant upregulation of heme-regulated genes, such as the globin genes [hemoglobin alpha (HBA) and hemoglobin gamma (HBG)] and the heme oxygenase 1 (HMOX1) gene, in K562 cells. Next, to investigate the mechanism by which ALA is transported into erythroid cells, quantitative RT-PCR analysis was performed on previously identified ALA transporters, including solute carrier family 15 (oligopeptide transporter), member (SLC15A) 1, SLC15A2, solute carrier family 36 (proton/amino acid symporter), member (SLC36A1), and solute carrier family 6 (neurotransmitter transporter), member 13 (SLC6A13). Our analysis revealed that SLC36A1 was abundantly expressed in erythroid cells. Thus, gamma-aminobutyric acid (GABA) was added to K562 cells to competitively inhibit SLC36A1-mediated transport. GABA treatment significantly impeded the ALA-mediated increase in the number of hemoglobinized cells as well as the induction of HBG, HBA, and HMOX1. Finally, small-interfering RNA-mediated knockdown of ALAS2 in HiDEP cells considerably decreased the expression of HBA, HBG, and HMOX1, and these expression levels were rescued with ALA treatment. In summary, ALA appears to be transported into erythroid cells mainly by SLC36A1 and is utilized to generate heme. ALA may represent a novel therapeutic option for CSA treatment, particularly for cases harboring ALAS2 mutations.


Aminolevulinic Acid/pharmacology , Anemia, Sideroblastic/drug therapy , Erythropoiesis/drug effects , Genetic Diseases, X-Linked/drug therapy , 5-Aminolevulinate Synthetase/antagonists & inhibitors , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/metabolism , Animals , Drug Evaluation, Preclinical , Erythroblasts/cytology , Erythroblasts/drug effects , Erythroblasts/metabolism , Erythropoiesis/genetics , Erythropoiesis/physiology , Gene Knockdown Techniques , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Heme/biosynthesis , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hemoglobin A/genetics , Hemoglobin A/metabolism , Hemoglobins, Abnormal/genetics , Hemoglobins, Abnormal/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , K562 Cells , Mice , Symporters/genetics , Symporters/metabolism , Up-Regulation/drug effects , gamma-Aminobutyric Acid/pharmacology
16.
Biotechnol Lett ; 35(5): 763-8, 2013 May.
Article En | MEDLINE | ID: mdl-23338702

5-Aminolevulinic acid (ALA) synthase (ALAS) HemA from non-sulfur photosynthetic bacteria has been used for the ALA bioproduction, whereas the isoenzyme HemT/HemO is less studied and not used for ALA production. Two ALAS-encoding genes, hemA and hemO from Rhodopseudomonas palustris were cloned, purified and characterized. The ALASs had very high specific activity, 3.6 and 2.7 U/mg, respectively, and strong affinity for one of its substrates, succinyl-CoA, K m with values of 11 and 4.4 µM, respectively. HemO retained up to 60 % maximum activity within a broad range of concentrations of hemin, while HemA kept only 20 % at 10 µM hemin. Escherichia coli overexpressing HemA or HemO produced 5.7 and 6.3 g ALA/l, respectively, in a 5 l bioreactor.


5-Aminolevulinate Synthetase/metabolism , Aminolevulinic Acid/metabolism , Cloning, Molecular/methods , Recombinant Proteins/metabolism , Rhodopseudomonas/enzymology , 5-Aminolevulinate Synthetase/antagonists & inhibitors , 5-Aminolevulinate Synthetase/chemistry , 5-Aminolevulinate Synthetase/genetics , Aminolevulinic Acid/analysis , Electrophoresis, Polyacrylamide Gel , Hemin/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Rhodopseudomonas/genetics
19.
Cell Mol Biol (Noisy-le-grand) ; 55(2): 147-51, 2009 Jul 01.
Article En | MEDLINE | ID: mdl-19656463

Acute porphyrias are caused by enzyme defects along the heme synthesis pathway. Patients usually present with abdominal pain, impaired intestinal motility, neurological and psychiatric symptoms, hypertension, tachycardia, hyponatriemia and reddish urine. This article gives an overview over drugs that are recommended in patients with acute hepatic porphyrias and represents a compilation of four so far existing lists.


Drug-Related Side Effects and Adverse Reactions , Porphyria, Acute Intermittent/therapy , 5-Aminolevulinate Synthetase/antagonists & inhibitors , 5-Aminolevulinate Synthetase/metabolism , Arginine/therapeutic use , Heme/metabolism , Heme/therapeutic use , Humans , Liver Transplantation
...