Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
Add more filters

Publication year range
1.
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
2.
Lancet ; 400(10351): 493-501, 2022 08 13.
Article in English | MEDLINE | ID: mdl-35964609

ABSTRACT

BACKGROUND: Patients with non-transfusion-dependent thalassaemia (NTDT), although they do not require regular blood transfusions for survival, can still accrue a heavy burden of comorbidities. No approved disease-modifying therapies exist for these patients. We aimed to investigate the safety and efficacy of mitapivat (Agios Pharmaceuticals, Cambridge, MA, USA), a pyruvate kinase activator, in adults with non-transfusion-dependent (NTD) α-thalassaemia or NTD ß-thalassaemia. METHODS: In this open-label, multicentre, phase 2 study, patients were recruited from four academic clinical study sites in Oakland, CA, and Boston, MA, USA; Toronto, ON, Canada; and London, UK. Patients were eligible if they were aged 18 years or older, with NTDT (including ß-thalassaemia with or without α-globin gene mutations, haemoglobin E ß-thalassaemia, or α-thalassaemia), and a baseline haemoglobin concentration of 10·0 g/dL or lower. During a 24-week core period, mitapivat was administered orally at 50 mg twice daily for the first 6 weeks followed by an escalation to 100 mg twice daily for 18 weeks thereafter. The primary endpoint was haemoglobin response (a ≥1·0 g/dL increase in haemoglobin concentration from baseline at one or more assessments between weeks 4 and 12). Efficacy and safety were assessed in the full analysis set (ie, all patients who received at least one dose of study drug). This study is registered with ClinicalTrials.gov, NCT03692052, and is closed to accrual. FINDINGS: Between Dec 28, 2018, and Feb 6, 2020, 27 patients were screened, of whom 20 were enrolled (15 [75%] with ß-thalassaemia and five [25%] with α-thalassaemia) and received mitapivat. The median age of patients was 44 years (IQR 35-56), 15 (75%) of 20 patients were female, five (25%) were male, and ten (50%) identified as Asian. 16 (80% [90% CI 60-93]) of 20 patients had a haemoglobin response (p<0·0001), five (100%) of five with α-thalassaemia and 11 (73%) of 15 with ß-thalassaemia. 17 (85%) patients had a treatment-emergent adverse event, and 13 had a treatment-emergent event that was considered to be treatment related. One serious treatment-emergent adverse event occurred (grade 3 renal impairment), which was considered unrelated to study drug, resulting in discontinuation of treatment. The most commonly reported treatment-emergent adverse events were initial insomnia (ten [50%] patients), dizziness (six [30%]), and headache (five [25%]). No patients died during the 24-week core period. INTERPRETATION: These efficacy and safety results support the continued investigation of mitapivat for the treatment of both α-thalassaemia and ß-thalassaemia. FUNDING: Agios Pharmaceuticals.


Subject(s)
Piperazines , Quinolines , alpha-Thalassemia , beta-Thalassemia , Adult , Female , Hemoglobins , Humans , Male , Middle Aged , Piperazines/adverse effects , Pyruvate Kinase , Quinolines/adverse effects , alpha-Thalassemia/drug therapy , beta-Thalassemia/drug therapy
3.
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
4.
Blood ; 133(17): 1865-1875, 2019 04 25.
Article in English | MEDLINE | ID: mdl-30655275

ABSTRACT

New treatments directly targeting polymerization of sickle hemoglobin (HbS), the proximate event in the pathophysiology of sickle cell disease (SCD), are needed to address the severe morbidity and early mortality associated with the disease. Voxelotor (GBT440) is a first-in-class oral therapy specifically developed to treat SCD by modulating the affinity of hemoglobin (Hb) for oxygen, thus inhibiting HbS polymerization and downstream adverse effects of hemolytic anemia and vaso-occlusion. GBT440-001 was a phase 1/2 randomized, double-blind, placebo-controlled, single and multiple ascending dose study of voxelotor in adult healthy volunteers and patients with SCD, followed by a single-arm, open-label extension study. This report describes results of voxelotor (500-1000 mg per day) in patients with sickle cell anemia. The study evaluated the safety, tolerability, pharmacokinetic, and pharmacodynamic properties of voxelotor and established proof of concept by improving clinical measures of anemia, hemolysis, and sickling. Thirty-eight patients with SCD received 28 days of voxelotor 500, 700, or 1000 mg per day or placebo; 16 patients received 90 days of voxelotor 700 or 900 mg per day or placebo. Four patients from the 90-day cohort were subsequently enrolled in an extension study and treated with voxelotor 900 mg per day for 6 months. All patients who received multiple doses of voxelotor for ≥28 days experienced hematologic improvements including increased Hb and reduction in hemolysis and percentage of sickled red cells, supporting the potential of voxelotor to serve as a disease-modifying therapy for SCD. Voxelotor was well tolerated with no treatment-related serious adverse events and no evidence of tissue hypoxia. These trials were registered at www.clinicaltrials.gov as #NCT02285088 and #NCT03041909.


Subject(s)
Anemia, Sickle Cell/drug therapy , Benzaldehydes/therapeutic use , Hematologic Agents/therapeutic use , Pyrazines/therapeutic use , Pyrazoles/therapeutic use , Adolescent , Adult , Benzaldehydes/pharmacokinetics , Case-Control Studies , Cohort Studies , Double-Blind Method , Female , Follow-Up Studies , Hematologic Agents/pharmacokinetics , Humans , Male , Maximum Tolerated Dose , Middle Aged , Prognosis , Pyrazines/pharmacokinetics , Pyrazoles/pharmacokinetics , Tissue Distribution , Young Adult
5.
Br J Haematol ; 189(4): 635-639, 2020 05.
Article in English | MEDLINE | ID: mdl-32330288

ABSTRACT

With the developing COVID-19 pandemic, patients with inherited anaemias require specific advice regarding isolation and changes to usual treatment schedules. The National Haemoglobinopathy Panel (NHP) has issued guidance on the care of patients with sickle cell disease, thalassaemia, Diamond Blackfan anaemia (DBA), congenital dyserythropoietic anaemia (CDA), sideroblastic anaemia, pyruvate kinase deficiency and other red cell enzyme and membrane disorders. Cascading of accurate information for clinicians and patients is paramount to preventing adverse outcomes, such as patients who are at increased risk of fulminant bacterial infection due to their condition or its treatment erroneously self-isolating if their fever is mistakenly attributed to a viral cause, delaying potentially life-saving antibiotic therapy. Outpatient visits should be minimised for most patients, however some, such as first transcranial dopplers for children with sickle cell anaemia should not be delayed as known risk of stroke will outweigh the unknown risk from COVID-19 infection. Blood transfusion programmes should be continued, but specific changes to usual clinical pathways can be instituted to reduce risk of patient exposure to COVID-19, as well as contingency planning for possible reductions in blood available for transfusions. Bone marrow transplants for these disorders should be postponed until further notice. With the current lack of evidence on the risk and complications of COVID-19 infection in these patients, national data collection is ongoing to record outcomes and eventually to identify predictors of disease severity, particularly important if further waves of infection travel through the population.


Subject(s)
Anemia/complications , Anemia/therapy , Betacoronavirus , Coronavirus Infections/complications , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Pneumonia, Viral/complications , Pneumonia, Viral/prevention & control , Blood Transfusion , Bone Marrow Transplantation , COVID-19 , Cross Infection/prevention & control , Humans , SARS-CoV-2
6.
Br J Haematol ; 184(5): 721-734, 2019 03.
Article in English | MEDLINE | ID: mdl-30681718

ABSTRACT

Novel therapies in development have brought a new focus on pyruvate kinase deficiency (PKD), the most common congenital haemolytic anaemia due to a glycolytic enzyme deficiency. With an improved recognition of its clinical presentation and understanding of the diagnostic pathway, more patients are likely to be identified with this anaemia. Complications, including gallstones and non-transfusion-related iron overload, require monitoring for early diagnosis and management. Current management remains supportive with red cell transfusions, chelation and splenectomy. Decisions to transfuse and/or splenectomise must be individualised. Haematopoietic stem cell transplant has been pursued in a small number of patients with mixed outcomes. Novel treatment approaches, which range from a small molecule pyruvate kinase activator to gene therapy, may transform the way in which PKD is managed in the future. In this review, we discuss the pathophysiology of PKD and present our approaches to diagnosis, monitoring and management of patients with this anaemia.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/therapy , Erythrocyte Transfusion , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/therapy , Anemia, Hemolytic, Congenital Nonspherocytic/complications , Anemia, Hemolytic, Congenital Nonspherocytic/diagnosis , Gallstones/etiology , Gallstones/therapy , Humans , Iron Overload/etiology , Iron Overload/therapy , Pyruvate Metabolism, Inborn Errors/complications , Pyruvate Metabolism, Inborn Errors/diagnosis
12.
Blood ; 124(20): 3151-4, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25293775

ABSTRACT

A rare point mutation in the core promoter -270GC-rich box of PIGM, a housekeeping gene, disrupts binding of the generic transcription factor (TF) Sp1 and causes inherited glycosylphosphatidylinositol (GPI) deficiency (IGD). We show that whereas PIGM messenger RNA levels and surface GPI expression in IGD B cells are low, GPI expression is near normal in IGD erythroid cells. This divergent phenotype results from differential promoter chromatin accessibility and binding of Sp1. Specifically, whereas PIGM transcription in B cells is dependent on Sp1 binding to the -270GC-rich box and is associated with lower promoter accessibility, in erythroid cells, Sp1 activates PIGM transcription by binding upstream of (but not to) the -270GC-rich box. These findings explain intact PIGM transcription in IGD erythroid cells and the lack of clinically significant intravascular hemolysis in patients with IGD. Furthermore, they provide novel insights into tissue-specific transcriptional control of a housekeeping gene by a generic TF.


Subject(s)
Glycosylphosphatidylinositols/deficiency , Hemoglobinuria, Paroxysmal/genetics , Mannosyltransferases/genetics , Transcriptional Activation , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Erythrocytes/metabolism , Erythrocytes/pathology , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Hemoglobinuria, Paroxysmal/metabolism , Hemoglobinuria, Paroxysmal/pathology , Humans , Mutation , Phenotype , Promoter Regions, Genetic , Seizures , Sp1 Transcription Factor/metabolism
13.
Blood ; 124(1): 134-41, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24805191

ABSTRACT

HDAC inhibitors (HDACi) increase transcription of some genes through histone hyperacetylation. To test the hypothesis that HDACi-mediated enhanced transcription might be of therapeutic value for inherited enzyme deficiency disorders, we focused on the glycolytic and pentose phosphate pathways (GPPPs). We show that among the 16 genes of the GPPPs, HDACi selectively enhance transcription of glucose 6-phosphate dehydrogenase (G6PD). This requires enhanced recruitment of the generic transcription factor Sp1, with commensurate recruitment of histone acetyltransferases and deacetylases, increased histone acetylation, and polymerase II recruitment to G6PD. These G6PD-selective transcriptional and epigenetic events result in increased G6PD transcription and ultimately restored enzymatic activity in B cells and erythroid precursor cells from patients with G6PD deficiency, a disorder associated with acute or chronic hemolytic anemia. Therefore, restoration of enzymatic activity in G6PD-deficient nucleated cells is feasible through modulation of G6PD transcription. Our findings also suggest that clinical consequences of pathogenic missense mutations in proteins with enzymatic function can be overcome in some cases by enhancement of the transcriptional output of the affected gene.


Subject(s)
Epigenesis, Genetic/drug effects , Glucosephosphate Dehydrogenase Deficiency/genetics , Glucosephosphate Dehydrogenase/biosynthesis , Glucosephosphate Dehydrogenase/genetics , Histone Deacetylase Inhibitors/pharmacology , Transcription, Genetic/drug effects , Cells, Cultured , Chromatin Immunoprecipitation , Glucosephosphate Dehydrogenase Deficiency/enzymology , Humans , Real-Time Polymerase Chain Reaction
14.
Hum Mol Genet ; 22(16): 3187-94, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23591993

ABSTRACT

How the transcription repressing complex Polycomb interacts with transcriptional regulators at housekeeping genes in somatic cells is not well understood. By exploiting a CpG island (CGI) point mutation causing a Mendelian disease, we show that DNA binding of activating transcription factor (TF) determines histone acetylation and nucleosomal depletion commensurate with Polycomb exclusion from the target promoter. Lack of TF binding leads to reversible transcriptional repression imposed by nucleosomal compaction and consolidated by Polycomb recruitment and establishment of bivalent chromatin status. Thus, within a functional hierarchy of transcriptional regulators, TF binding is the main determinant of Polycomb recruitment to the CGI of a housekeeping gene in somatic cells.


Subject(s)
Activating Transcription Factors/chemistry , B-Lymphocytes/chemistry , B-Lymphocytes/metabolism , CpG Islands/genetics , Glycosylphosphatidylinositols/deficiency , Hemoglobinuria, Paroxysmal/genetics , Mannosyltransferases/genetics , Polycomb-Group Proteins/metabolism , Activating Transcription Factors/genetics , Activating Transcription Factors/metabolism , Base Sequence , Cells, Cultured , DNA Methylation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Genes, Essential , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Hemoglobinuria, Paroxysmal/metabolism , Histones/chemistry , Histones/metabolism , Humans , Mannosyltransferases/metabolism , Molecular Sequence Data , Nucleosomes/metabolism , Point Mutation , Polycomb-Group Proteins/chemistry , Polycomb-Group Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Seizures
16.
Am J Transplant ; 14(11): 2478-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25220786

ABSTRACT

Mixed hematopoietic chimerism is a powerful means of generating donor-specific tolerance, allowing long-term graft acceptance without lifelong dependence on immunosuppressive drugs. To avoid the need for whole body irradiation and associated side effects, we utilized a radiation-free minimal conditioning regime to induce long-term tolerance across major histocompatibility barriers. We found that low-dose busulfan, in combination with host T cell depletion and short-term sirolimus-based immunosuppression, facilitated efficient donor engraftment. Tolerance was achieved when mice were transplanted with whole or T cell-depleted bone marrow, or purified progenitor cells. Tolerance induction was associated with an expansion in regulatory T cells and was not abrogated in the absence of a thymus, suggesting a dominant or compensatory peripheral mode of tolerance. Importantly, we were able to generate durable chimerism and tolerance to donor skin grafts in both young and aged mice, despite age-related thymic atrophy and immune senescence. Clinically, this is especially relevant as the majority of transplant recipients are older patients whose immune recovery might be dangerously slow and would benefit from radiation-free minimal conditioning regimes that allow efficient donor engraftment without fully ablating the recipient immune system.


Subject(s)
Aging/immunology , Immune Tolerance , Transplantation Conditioning , Transplantation Immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Whole-Body Irradiation
18.
J Oral Rehabil ; 41(12): 928-40, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25195784

ABSTRACT

To explore the quality of reporting (writing and graphics) of articles that used time-to-event analyses to report dental treatment outcomes. A systematic search of the top 50 dental journals in 2008 produced the sample of articles for this analysis. Articles reporting treatment outcomes with (n = 95) and without (n = 91) time-to-event statistics were reviewed. Survival descriptive words used in the two groups were analysed (Pearson's chi-square). The quality of life tables, survival curves and time-to-event statistics were assessed (Kappa analysed agreement) and explored. Words describing dental outcomes 'over time' were more common in time-to-event compared with control articles (77%, 3%, P < 0.001). Non-specific use of 'rate' was common across both groups. Life tables and survival curves were used by 39% and 48% of the time-to-event articles, with at least one used by 82%. Construction quality was poor: 21% of life tables and 28% of survival curves achieved an acceptable standard. Time-to-event statistical reporting was poor: 3% achieved a high and 59% achieved an acceptable standard. The survival statistic, summary figure and standard error were reported in 76%, 95% and 20% of time-to-event articles. Individual statistical terms and graphic aids were common within and unique to time-to-event articles. Unfortunately, important details were regularly omitted from statistical descriptions and survival figures making the overall quality poor. It is likely this will mean such articles will be incorrectly indexed in databases, missed by searchers and unable to be understood completely if identified.


Subject(s)
Dental Research/standards , Kaplan-Meier Estimate , Periodicals as Topic , Humans , Life Tables , Quality of Life , Research Design , Statistics as Topic
19.
Nat Genet ; 37(11): 1258-63, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16227998

ABSTRACT

We identified 11 human pedigrees with dominantly inherited hemolytic anemias in both the hereditary stomatocytosis and spherocytosis classes. Affected individuals in these families had an increase in membrane permeability to Na and K that is particularly marked at 0 degrees C. We found that disease in these pedigrees was associated with a series of single amino-acid substitutions in the intramembrane domain of the erythrocyte band 3 anion exchanger, AE1. Anion movements were reduced in the abnormal red cells. The 'leak' cation fluxes were inhibited by SITS, dipyridamole and NS1652, chemically diverse inhibitors of band 3. Expression of the mutated genes in Xenopus laevis oocytes induced abnormal Na and K fluxes in the oocytes, and the induced Cl transport was low. These data are consistent with the suggestion that the substitutions convert the protein from an anion exchanger into an unregulated cation channel.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/genetics , Cations/metabolism , Chlorides/metabolism , Erythrocytes/metabolism , Potassium/metabolism , Sodium/metabolism , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid/pharmacology , Amino Acid Substitution , Anemia, Hemolytic/genetics , Anemia, Hemolytic/metabolism , Animals , Anion Exchange Protein 1, Erythrocyte/metabolism , Benzoates/pharmacology , Biological Transport , Cell Membrane Permeability , Dipyridamole/pharmacology , Humans , Molecular Sequence Data , Oocytes/cytology , Oocytes/metabolism , Pedigree , Phenylurea Compounds/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Protein Structure, Tertiary , RNA/metabolism , Spherocytosis, Hereditary/genetics , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL