Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Blood ; 143(10): 866-871, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38118071

ABSTRACT

ABSTRACT: Pyruvate kinase (PK) is a key enzyme in glycolysis, the sole source of adenosine triphosphate, which is essential for all energy-dependent activities of red blood cells. Activating PK shows great potential for treating a broad range of hemolytic anemias beyond PK deficiency, because they also enhance activity of wild-type PK. Motivated by observations of sickle-cell complications in sickle-trait individuals with concomitant PK deficiency, activating endogenous PK offers a novel and promising approach for treating patients with sickle-cell disease.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Anemia, Sickle Cell , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors , Humans , Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , Erythrocytes , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/complications
2.
Drugs ; 83(17): 1613-1620, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37991635

ABSTRACT

Mitapivat (Pyrukynd®), an oral, allosteric activator of pyruvate kinase (PK), is approved in the USA for the treatment of haemolytic anaemia in adults with PK deficiency and in the EU and UK for the treatment of PK deficiency in adults. Mitapivat acts by restoring activity of the red blood cell (RBC) PK enzyme, which is dysfunctional due to genetic mutations in the PKLR gene in patients with PK deficiency. In the double-blind placebo-controlled phase III ACTIVATE trial in adults with PK deficiency who were not regularly RBC transfused, mitapivat was superior to placebo in improving haemoglobin levels. In the single-arm phase III ACTIVATE-T trial in adults with PK deficiency who were regularly RBC transfused, a reduction in RBC transfusion burden was observed with mitapivat. In both trials, mitapivat improved other clinical parameters of haemolysis and patient-reported health-related quality of life. At the approved twice-daily dosage range, mitapivat was generally well tolerated, with adverse events generally being mild to moderate in severity. Results from an ongoing extension study in previously enrolled phase III trial patients will be of interest. Currently available data indicate that mitapivat, the first approved disease-modifying drug for PK deficiency in adults, is a valuable treatment option for this rare disease.


Pyruvate kinase (PK) deficiency is a rare hereditary disease caused by mutations affecting the function of the PK enzyme in red blood cells (RBCs) and is characterized by chronic haemolytic anaemia. Treatment options for adults with PK deficiency have historically been limited to supportive care and are themselves associated with potentially serious complications. Oral mitapivat (Pyrukynd®) is the first disease-modifying drug to be approved for use in adults with PK deficiency and acts by restoring activity of the dysfunctional RBC PK enzyme. Based on findings from two phase III clinical trials, twice-daily oral mitapivat provided clinical benefit in adults with PK deficiency, both in patients not requiring, and requiring, regular RBC transfusions. Improvements in disease-specific health-related quality of life were observed in adults with PK deficiency treated with mitapivat. Mitapivat was generally well tolerated, with most adverse events being mild to moderate in severity. In conclusion, current evidence indicates that mitapivat is a valuable treatment option for adults with PK deficiency.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Pyruvate Kinase , Humans , Adult , Pyruvate Kinase/genetics , Quality of Life , Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Randomized Controlled Trials as Topic
3.
Am J Ther ; 30(5): e433-e438, 2023.
Article in English | MEDLINE | ID: mdl-37713687

ABSTRACT

BACKGROUND: Pyruvate kinase (PK) deficiency is a rare enzyme-linked glycolytic defect resulting in mild-to-severe chronic persistent erythrocyte hemolysis. The disease is an autosomal recessive trait caused by mutations in the PK liver and red blood cell gene characterized by insufficient erythrocyte PK activity. PK deficiency is most diagnosed in persons of northern European descent and managed with packed red blood cell transfusions, chelation, and splenectomy with cholecystectomy. Mitapivat is the first approved therapy indicated for hemolytic anemia in adults with PK deficiency with the potential for delaying splenectomy in mild-moderate disease. MECHANISM OF ACTION, PHARMACODYNAMICS, AND PHARMACOKINETICS: Mitapivat is a PK activator that acts by allosterically binding to the PK tetramer and increases PK activity. The red blood cell form of PK is mutated in PK deficiency, which leads to reduced adenosine triphosphate, shortened red blood cell lifespan, and chronic hemolysis. The half-life of elimination is 3-5 hours, with 73% bioavailability, 98% plasma protein binding, and a median duration of response of 7 months. CLINICAL TRIALS: Mitapivat has been investigated through various clinical trials for different therapeutic indications. Pivotal trials that serve the primary focus throughout this article are ACTIVATE, ACTIVATE-T, and RISE. ACTIVATE is a phase 3, randomized, double-blind, placebo-controlled study that evaluated the efficacy and safety of mitapivat in adult patients who were not receiving regular blood transfusions. Contrarily, ACTIVATE-T explored the safety and efficacy of mitapivat in adults with PK deficiency who received regular blood transfusions. Both trials demonstrated favorable use of mitapivat in PK deficiency. Focusing on another indication, the ongoing RISE trial investigates the optimal dosage of mitapivat in sickle cell disease. THERAPEUTIC ADVANCE: Mitapivat is an appropriate treatment for adults with PK deficiency requiring transfusions and may be considered for patients with symptomatic anemia who do not require transfusions and/or PK deficiency with compensated hemolysis without overt anemia.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Anemia, Hemolytic , Anemia, Sickle Cell , Quinolones , Humans , Adult , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Hemolysis , Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Anemia, Hemolytic, Congenital Nonspherocytic/diagnosis , Anemia, Hemolytic/drug therapy , Anemia, Hemolytic/etiology , Anemia, Sickle Cell/complications
4.
Sci Rep ; 13(1): 4395, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36927785

ABSTRACT

Iron homeostasis and dyserythropoiesis are poorly investigated in pyruvate kinase deficiency (PKD), the most common glycolytic defect of erythrocytes. Herein, we studied the main regulators of iron balance and erythropoiesis, as soluble transferrin receptor (sTfR), hepcidin, erythroferrone (ERFE), and erythropoietin (EPO), in a cohort of 41 PKD patients, compared with 42 affected by congenital dyserythropoietic anemia type II (CDAII) and 50 with hereditary spherocytosis (HS). PKD patients showed intermediate values of hepcidin and ERFE between CDAII and HS, and clear negative correlations between log-transformed hepcidin and log-EPO (Person's r correlation coefficient = - 0.34), log-hepcidin and log-ERFE (r = - 0.47), and log-hepcidin and sTfR (r = - 0.44). sTfR was significantly higher in PKD; EPO levels were similar in PKD and CDAII, both higher than in HS. Finally, genotype-phenotype correlation in PKD showed that more severe patients, carrying non-missense/non-missense genotypes, had lower hepcidin and increased ERFE, EPO, and sTFR compared with the others (missense/missense and missense/non-missense), suggesting a higher rate of ineffective erythropoiesis. We herein investigated the main regulators of systemic iron homeostasis in the largest cohort of PKD patients described so far, opening new perspectives on the molecular basis and therapeutic approaches of this disease.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Anemia , Erythropoietin , Humans , Hepcidins/metabolism , Iron/metabolism , Anemia/drug therapy , Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Erythropoiesis/genetics , Receptors, Transferrin
6.
N Engl J Med ; 386(15): 1432-1442, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35417638

ABSTRACT

BACKGROUND: Pyruvate kinase deficiency is a rare, hereditary, chronic condition that is associated with hemolytic anemia. In a phase 2 study, mitapivat, an oral, first-in-class activator of erythrocyte pyruvate kinase, increased the hemoglobin level in patients with pyruvate kinase deficiency. METHODS: In this global, phase 3, randomized, placebo-controlled trial, we evaluated the efficacy and safety of mitapivat in adults with pyruvate kinase deficiency who were not receiving regular red-cell transfusions. The patients were assigned to receive either mitapivat (5 mg twice daily, with potential escalation to 20 or 50 mg twice daily) or placebo for 24 weeks. The primary end point was a hemoglobin response (an increase from baseline of ≥1.5 g per deciliter in the hemoglobin level) that was sustained at two or more scheduled assessments at weeks 16, 20, and 24. Secondary efficacy end points were the average change from baseline in the hemoglobin level, markers of hemolysis and hematopoiesis, and the change from baseline at week 24 in two pyruvate kinase deficiency-specific patient-reported outcome measures. RESULTS: Sixteen of the 40 patients (40%) in the mitapivat group had a hemoglobin response, as compared with none of the 40 patients in the placebo group (adjusted difference, 39.3 percentage points; 95% confidence interval, 24.1 to 54.6; two-sided P<0.001). Patients who received mitapivat had a greater response than those who received placebo with respect to each secondary end point, including the average change from baseline in the hemoglobin level. The most common adverse events were nausea (in 7 patients [18%] in the mitapivat group and 9 patients [23%] in the placebo group) and headache (in 6 patients [15%] and 13 patients [33%], respectively). Adverse events of grade 3 or higher occurred in 10 patients (25%) who received mitapivat and 5 patients (13%) who received placebo. CONCLUSIONS: In patients with pyruvate kinase deficiency, mitapivat significantly increased the hemoglobin level, decreased hemolysis, and improved patient-reported outcomes. No new safety signals were identified in the patients who received mitapivat. (Funded by Agios Pharmaceuticals; ACTIVATE ClinicalTrials.gov number, NCT03548220.).


Subject(s)
Piperazines , Pyruvate Kinase , Quinolines , Adult , Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Double-Blind Method , Hemoglobins/analysis , Hemoglobins/drug effects , Hemolysis/drug effects , Humans , Piperazines/pharmacology , Piperazines/therapeutic use , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/drug therapy , Quinolines/pharmacology , Quinolines/therapeutic use
7.
N Engl J Med ; 381(10): 933-944, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31483964

ABSTRACT

BACKGROUND: Pyruvate kinase deficiency is caused by mutations in PKLR and leads to congenital hemolytic anemia. Mitapivat is an oral, small-molecule allosteric activator of pyruvate kinase in red cells. METHODS: In this uncontrolled, phase 2 study, we evaluated the safety and efficacy of mitapivat in 52 adults with pyruvate kinase deficiency who were not receiving red-cell transfusions. The patients were randomly assigned to receive either 50 mg or 300 mg of mitapivat twice daily for a 24-week core period; eligible patients could continue treatment in an ongoing extension phase. RESULTS: Common adverse events, including headache and insomnia, occurred at the time of drug initiation and were transient; 92% of the episodes of headache and 47% of the episodes of insomnia resolved within 7 days. The most common serious adverse events, hemolytic anemia and pharyngitis, each occurred in 2 patients (4%). A total of 26 patients (50%) had an increase of more than 1.0 g per deciliter in the hemoglobin level. Among these patients, the mean maximum increase was 3.4 g per deciliter (range, 1.1 to 5.8), and the median time until the first increase of more than 1.0 g per deciliter was 10 days (range, 7 to 187); 20 patients (77%) had an increase of more than 1.0 g per deciliter in the hemoglobin level at more than 50% of visits during the core study period, with improvement in markers of hemolysis. The response was sustained in all 19 patients remaining in the extension phase, with a median follow-up of 29 months (range, 22 to 35). Hemoglobin responses were observed only in patients who had at least one missense PKLR mutation and were associated with the red-cell pyruvate kinase protein level at baseline. CONCLUSIONS: The administration of mitapivat was associated with a rapid increase in the hemoglobin level in 50% of adults with pyruvate kinase deficiency, with a sustained response during a median follow-up of 29 months during the extension phase. Adverse effects were mainly low-grade and transient. (Funded by Agios Pharmaceuticals; ClinicalTrials.gov number, NCT02476916.).


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Hemoglobins/metabolism , Piperazines/administration & dosage , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/drug therapy , Quinolines/administration & dosage , Administration, Oral , Adolescent , Adult , Anemia, Hemolytic, Congenital Nonspherocytic/blood , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Catechols , Drug Administration Schedule , Female , Follow-Up Studies , Headache/chemically induced , Humans , Male , Mutation , Piperazines/adverse effects , Pyruvate Kinase/blood , Pyruvate Kinase/genetics , Pyruvate Metabolism, Inborn Errors/blood , Pyruvate Metabolism, Inborn Errors/genetics , Quinolines/adverse effects , Sleep Initiation and Maintenance Disorders/chemically induced , Tyrphostins , Young Adult
8.
Curr Protein Pept Sci ; 20(4): 304-315, 2019.
Article in English | MEDLINE | ID: mdl-30370845

ABSTRACT

Triosephosphate isomerase is the fifth enzyme in glycolysis and its canonical function is the reversible isomerization of glyceraldehyde-3-phosphate and dihydroxyacetone phosphate. Within the last decade multiple other functions, that may not necessarily always involve catalysis, have been described. These include variations in the degree of its expression in many types of cancer and participation in the regulation of the cell cycle. Triosephosphate isomerase may function as an auto-antigen and in the evasion of the immune response, as a factor of virulence of some organisms, and also as an important allergen, mainly in a variety of seafoods. It is an important factor to consider in the cryopreservation of semen and seems to play a major role in some aspects of the development of Alzheimer's disease. It also seems to be responsible for neurodegenerative alterations in a few cases of human triosephosphate isomerase deficiency. Thus, triosephosphate isomerase is an excellent example of a moonlighting protein.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/veterinary , Animal Diseases/enzymology , Carbohydrate Metabolism, Inborn Errors/veterinary , Triose-Phosphate Isomerase/deficiency , Triose-Phosphate Isomerase/metabolism , Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Anemia, Hemolytic, Congenital Nonspherocytic/metabolism , Animal Diseases/drug therapy , Animals , Carbohydrate Metabolism, Inborn Errors/drug therapy , Carbohydrate Metabolism, Inborn Errors/metabolism , Dihydroxyacetone Phosphate/metabolism , Glyceraldehyde 3-Phosphate/metabolism , Glycolysis , Humans
10.
Wien Klin Wochenschr ; 90(19): 690-5, 1978 Oct 13.
Article in German | MEDLINE | ID: mdl-695663

ABSTRACT

The effect of 6-methylprednisolone (GCC) was studied on erythropoietin (ESF) levels and on the metabolic functions of erythrocytes (RBC). GCC (U mg/kg/day for 15 days) was administered to 6 patients with the haemolytic-uraemic syndrome (group B) and to 6 patients with non-spherocytic haemolytic anaemia due to hereditary pyruvate kinase enzyme deficiency (group C). 6 healthy persons served as control (group A). The metabolic functions of RBC were investigated by assaying HMPS activity, GSH/GSSG and lactate/pyruvate ratios, relevant glycolytic intermediates, 2,3-DPG, ATP, and key enzymes. A significant increase in ESF was observed in group B patients after GCC therapy, correlating with an improvement in the haemolytic state, and consequent rectification of the secondary disturbances of RBC metabolism. Group C patients already had raised ESF levels before GCC therapy; no further increase occured in response to treatment and no other clinical or haematological change was recorded. Hence, no harmonal influence of GCC on the disturbed RBC metabolic process was detectable in the cases.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/blood , Erythropoietin/blood , Hemolytic-Uremic Syndrome/blood , Methylprednisolone/administration & dosage , Adult , Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Deficiency Diseases/genetics , Erythrocytes/metabolism , Female , Hemolytic-Uremic Syndrome/drug therapy , Humans , Male , Methylprednisolone/therapeutic use , Pyruvate Kinase/deficiency
11.
Clin Chim Acta ; 71(2): 331-4, 1976 Sep 06.
Article in English | MEDLINE | ID: mdl-963898

ABSTRACT

The effect of riboflavin on six patients with hemolytic anemia due to hereditary pyruvate kinase deficiency was studied. Treatment over a period of four to six months produced no significant hematologic or enzymologic changes in the variants investigated.


Subject(s)
Erythrocytes/enzymology , Pyruvate Kinase/deficiency , Riboflavin/therapeutic use , Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Glutathione Reductase/metabolism , Humans , Pyruvate Kinase/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...