Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Haematologica ; 104(3): 477-484, 2019 03.
Article in English | MEDLINE | ID: mdl-30337358

ABSTRACT

ß-thalassemia, a hereditary blood disorder caused by defective synthesis of hemoglobin ß globin chains, leads to ineffective erythropoiesis and chronic anemia that may require blood transfusions. Sotatercept (ACE-011) acts as a ligand trap to inhibit negative regulators of late-stage erythropoiesis in the transforming growth factor ß superfamily, correcting ineffective erythropoiesis. In this phase II, open-label, dose-finding study, 16 patients with transfusion-dependent ß -thalassemia and 30 patients with non-transfusion-dependent ß-thalassemia were enrolled at seven centers in four countries between November 2012 and November 2014. Patients were treated with sotatercept at doses of 0.1, 0.3, 0.5, 0.75, or 1.0 mg/kg to determine a safe and effective dose. Doses were administered by subcutaneous injection every 3 weeks. Patients were treated for ≤22 months. Response was assessed as a ≥20% reduction in transfusion burden sustained for 24 weeks in transfusion-dependent ß-thalassemia patients, and an increase in hemoglobin level of ≥1.0 g/dL sustained for 12 weeks in non-transfusion-dependent ß-thalassemia patients. Sotatercept was well tolerated. After a median treatment duration of 14.4 months (range 0.6-35.9), no severe life-threatening adverse events were observed. Thirteen percent of patients reported serious but manageable adverse events. The active dose of sotatercept was ≥0.3 mg/kg for patients with non-transfusion-dependent ß-thalassemia and ≥0.5 mg/kg for those with transfusion-dependent ß-thalassemia. Of 30 non-transfusion-dependent ß-thalassemia patients treated with ≥0.1 mg/kg sotatercept, 18 (60%) achieved a mean hemoglobin increase ≥1.0 g/dL, and 11 (37%) an increase ≥1.5 g/dL, sustained for ≥12 weeks. Four (100%) transfusion-dependent ß-thalassemia patients treated with 1.0 mg/kg sotatercept achieved a transfusion-burden reduction of ≥20%. Sotatercept was effective and well tolerated in patients with ß-thalassemia. Most patients with non-transfusion-dependent ß-thalassemia treated with higher doses achieved sustained increases in hemoglobin level. Transfusion-dependent ß-thalassemia patients treated with higher doses of sotatercept achieved notable reductions in transfusion requirements. This trial was registered at ClinicalTrials.gov with the number NCT01571635.


Subject(s)
Anemia/drug therapy , Anemia/etiology , Recombinant Fusion Proteins/administration & dosage , beta-Thalassemia/complications , Adult , Anemia/blood , Anemia/diagnosis , Biomarkers , Blood Transfusion , Combined Modality Therapy , Erythrocyte Indices , Erythropoiesis/drug effects , Female , Hemoglobins , Humans , Ligands , Male , Middle Aged , Recombinant Fusion Proteins/adverse effects , Transforming Growth Factor beta/metabolism , Treatment Outcome , beta-Thalassemia/diagnosis , beta-Thalassemia/drug therapy
2.
Kidney Int ; 91(1): 86-95, 2017 01.
Article in English | MEDLINE | ID: mdl-27666759

ABSTRACT

Dysregulation of skeletal remodeling is a component of renal osteodystrophy. Previously, we showed that activin receptor signaling is differentially affected in various tissues in chronic kidney disease (CKD). We tested whether a ligand trap for the activin receptor type 2A (RAP-011) is an effective treatment of the osteodystrophy of the CKD-mineral bone disorder. With a 70% reduction in the glomerular filtration rate, CKD was induced at 14 weeks of age in the ldlr-/- high fat-fed mouse model of atherosclerotic vascular calcification and diabetes. Twenty mice with CKD, hyperphosphatemia, hyperparathyroidism, and elevated activin A were treated with RAP-011, wherease 19 mice were given vehicle twice weekly from week 22 until the mice were killed at 28 weeks of age. The animals were then evaluated by skeletal histomorphometry, micro-computed tomography, mechanical strength testing, and ex vivo bone cell culture. Results in the CKD groups were compared with those of the 16 sham-operated ldlr-/- high fat-fed mice. Sham-operated mice had low-turnover osteodystrophy and skeletal frailty. CKD stimulated bone remodeling with significant increases in osteoclast and osteoblast numbers and bone resorption. Compared with mice with CKD and sham-operated mice, RAP-011 treatment eliminated the CKD-induced increase in these histomorphometric parameters and increased trabecular bone fraction. RAP-011 significantly increased cortical bone area and thickness. Activin A-enhanced osteoclastogenesis was mediated through p-Smad2 association with c-fos and activation of nuclear factor of activated T cells c1 (NFATc1). Thus, an ActRIIA ligand trap reversed CKD-stimulated bone remodeling, likely through inhibition of activin-A induced osteoclastogenesis.


Subject(s)
Activins/metabolism , Bone Remodeling/drug effects , Chronic Kidney Disease-Mineral and Bone Disorder/drug therapy , Diabetes Mellitus, Experimental/complications , Osteoclasts/drug effects , Recombinant Fusion Proteins/therapeutic use , Renal Insufficiency, Chronic/complications , Animals , Cells, Cultured , Chronic Kidney Disease-Mineral and Bone Disorder/etiology , Disease Models, Animal , Glomerular Filtration Rate , Hyperphosphatemia/etiology , Male , Mice , Mice, Knockout , Osteoblasts/drug effects , Receptors, LDL/genetics , Vascular Calcification/etiology , X-Ray Microtomography
3.
J Genet Genomics ; 43(5): 307-18, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27216296

ABSTRACT

5q-syndrome is a distinct form of myelodysplastic syndrome (MDS) where a deletion on chromosome 5 is the underlying cause. MDS is characterized by bone marrow failures, including macrocytic anemia. Genetic mapping and studies using various models support the notion that ribosomal protein S14 (RPS14) is the candidate gene for the erythroid failure. Targeted disruption of RPS14 causes an increase in p53 activity and p53-mediated apoptosis, similar to what is observed with other ribosomal proteins. However, due to the higher risk for cancer development in patients with ribosome deficiency, targeting the p53 pathway is not a viable treatment option. To better understand the pathology of RPS14 deficiency in 5q-deletion, we generated a zebrafish model harboring a mutation in the RPS14 gene. This model mirrors the anemic phenotype seen in 5q-syndrome. Moreover, the anemia is due to a late-stage erythropoietic defect, where the erythropoietic defect is initially p53-independent and then becomes p53-dependent. Finally, we demonstrate the versatility of this model to test various pharmacological agents, such as RAP-011, L-leucine, and dexamethasone in order to identify molecules that can reverse the anemic phenotype.


Subject(s)
Anemia, Macrocytic/genetics , CRISPR-Cas Systems/genetics , Erythroid Cells/metabolism , Gene Editing , Ribosomal Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish , Anemia/complications , Anemia, Macrocytic/blood , Anemia, Macrocytic/complications , Animals , Base Sequence , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Disease Models, Animal , Mutation , Ribosomal Proteins/deficiency
4.
Blood ; 126(7): 880-90, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26109203

ABSTRACT

Diamond-Blackfan Anemia (DBA) is a bone marrow failure disorder characterized by low red blood cell count. Mutations in ribosomal protein genes have been identified in approximately half of all DBA cases. Corticosteriod therapy and bone marrow transplantation are common treatment options for patients; however, significant risks and complications are associated with these treatment options. Therefore, novel therapeutic approaches are needed for treating DBA. Sotatercept (ACE-011, and its murine ortholog RAP-011) acts as an activin receptor type IIA ligand trap, increasing hemoglobin and hematocrit in pharmacologic models, in healthy volunteers, and in patients with ß-thalassemia, by expanding late-stage erythroblasts through a mechanism distinct from erythropoietin. Here, we evaluated the effects of RAP-011 in zebrafish models of RPL11 ribosome deficiency. Treatment with RAP-011 dramatically restored hemoglobin levels caused by ribosome stress. In zebrafish embryos, RAP-011 likely stimulates erythropoietic activity by sequestering lefty1 from erythroid cells. These findings identify lefty1 as a signaling component in the development of erythroid cells and rationalize the use of sotatercept in DBA patients.


Subject(s)
Anemia, Diamond-Blackfan/drug therapy , Erythropoiesis/drug effects , Left-Right Determination Factors/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Zebrafish Proteins/antagonists & inhibitors , Activin Receptors, Type II/antagonists & inhibitors , Activin Receptors, Type II/blood , Anemia, Diamond-Blackfan/blood , Anemia, Diamond-Blackfan/genetics , Animals , Disease Models, Animal , Erythropoiesis/genetics , Gene Knockdown Techniques , Genes, p53 , Humans , Left-Right Determination Factors/blood , Left-Right Determination Factors/genetics , Ligands , Ribosomal Proteins/blood , Ribosomal Proteins/deficiency , Ribosomal Proteins/genetics , Signal Transduction/drug effects , Zebrafish , Zebrafish Proteins/blood , Zebrafish Proteins/genetics , beta-Thalassemia/blood , beta-Thalassemia/drug therapy
5.
Am J Hematol ; 90(1): 8-14, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25236856

ABSTRACT

Over expression of hepcidin antimicrobial peptide is a common feature of iron-restricted anemia in humans. We investigated the erythroid response to either erythropoietin or RAP-011, a "murinized" ortholog of sotatercept, in C57BL/6 mice and in hepcidin antimicrobial peptide 1 over expressing mice. Sotatercept, a soluble, activin receptor type IIA ligand trap, is currently being evaluated for the treatment of anemias associated with chronic renal disease, myelodysplastic syndrome, ß-thalassemia, and Diamond Blackfan anemia and acts by inhibiting signaling downstream of activin and other Transforming Growth Factor-ß superfamily members. We found that erythropoietin and RAP-011 increased hemoglobin concentration in C57BL/6 mice and in hepcidin antimicrobial peptide 1 over expressing mice. While erythropoietin treatment depleted splenic iron stores in C57BL/6 mice, RAP-011 treatment did not deplete splenic iron stores in mice of either genotype. Bone marrow erythroid progenitors from erythropoietin-treated mice exhibited iron-restricted erythropoiesis, as indicated by increased median fluorescence intensity of transferrin receptor immunostaining by flow cytometry. In contrast, RAP-011-treated mice did not exhibit the same degree of iron-restricted erythropoiesis. In conclusion, we have demonstrated that RAP-011 can improve hemoglobin concentration in hepcidin antimicrobial peptide 1 transgenic mice. Our data support the hypothesis that RAP-011 has unique biologic effects which prevent or circumvent depletion of mouse splenic iron stores. RAP-011 may, therefore, be an appropriate therapeutic for trials in human anemias characterized by increased expression of hepcidin antimicrobial peptide and iron-restricted erythropoiesis.


Subject(s)
Erythropoiesis/drug effects , Hemoglobins/analysis , Hepcidins/genetics , Iron/metabolism , Recombinant Fusion Proteins/pharmacology , Activin Receptors, Type II/chemistry , Animals , Biological Transport , Blood Cell Count , Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacology , Female , Immunoglobulin G/chemistry , Iron/blood , Ligands , Mice, Inbred C57BL , Mice, Transgenic , Spleen/metabolism
6.
Nat Med ; 20(4): 398-407, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24658077

ABSTRACT

The pathophysiology of ineffective erythropoiesis in ß-thalassemia is poorly understood. We report that RAP-011, an activin receptor IIA (ActRIIA) ligand trap, improved ineffective erythropoiesis, corrected anemia and limited iron overload in a mouse model of ß-thalassemia intermedia. Expression of growth differentiation factor 11 (GDF11), an ActRIIA ligand, was increased in splenic erythroblasts from thalassemic mice and in erythroblasts and sera from subjects with ß-thalassemia. Inactivation of GDF11 decreased oxidative stress and the amount of α-globin membrane precipitates, resulting in increased terminal erythroid differentiation. Abnormal GDF11 expression was dependent on reactive oxygen species, suggesting the existence of an autocrine amplification loop in ß-thalassemia. GDF11 inactivation also corrected the abnormal ratio of immature/mature erythroblasts by inducing apoptosis of immature erythroblasts through the Fas-Fas ligand pathway. Taken together, these observations suggest that ActRIIA ligand traps may have therapeutic relevance in ß-thalassemia by suppressing the deleterious effects of GDF11, a cytokine which blocks terminal erythroid maturation through an autocrine amplification loop involving oxidative stress and α-globin precipitation.


Subject(s)
Activin Receptors, Type II/metabolism , Bone Morphogenetic Proteins/antagonists & inhibitors , Erythroblasts/metabolism , Erythropoiesis/drug effects , Growth Differentiation Factors/antagonists & inhibitors , Hematinics/pharmacology , Recombinant Fusion Proteins/pharmacology , beta-Thalassemia/metabolism , Animals , Apoptosis/physiology , Autocrine Communication/physiology , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Disease Models, Animal , Fas Ligand Protein , Gene Amplification/physiology , Growth Differentiation Factors/metabolism , Ligands , Mice , Oxidative Stress/physiology , Reactive Oxygen Species , Signal Transduction , fas Receptor
7.
Br J Haematol ; 165(6): 870-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24635723

ABSTRACT

Sotatercept (ACE-011), a recombinant human fusion protein containing the extracellular domain of the human Activin receptor IIA, binds to and inhibits activin and other members of the transforming growth factor -ß (TGF-ß) superfamily. Administration of sotatercept led to a rapid and sustained increase in red blood cell (RBC) count and haemoglobin (Hb) in healthy volunteers (phase I clinical trials), but the mechanism is not fully understood. Mice treated with RAP-011 (murine ortholog of ACE-011) respond with a rapid (within 24 h) increase in haematocrit, Hb, and RBC count. These effects are accompanied by an equally rapid stimulation of late-stage erythroid precursors in the bone marrow (BM). RAP-011 also induces a significant increase in erythroid burst-forming units and erythropoietin, which could contribute to additional, sustained effects on RBC production. Further in vitro co-culture studies demonstrate that BM accessory cells are required for RAP-011 effects. To better understand which TGF-ß family ligand(s) mediate RAP-011 effects, we evaluated the impact of several of these ligands on erythroid differentiation. Our data suggest that RAP-011 may act to rescue growth differentiation factor 11/Activin A-induced inhibition of late-stage erythropoiesis. These data define the mechanism of action of a novel agent that regulates RBC differentiation and provide the rationale to develop sotatercept for the treatment of anaemia and ineffective erythropoiesis.


Subject(s)
Activin Receptors, Type II/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythropoiesis/drug effects , Erythropoiesis/physiology , Hemoglobins/biosynthesis , Recombinant Fusion Proteins/pharmacology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cellular Microenvironment/physiology , Colony-Forming Units Assay , Erythrocyte Indices/drug effects , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoietin/biosynthesis , Female , Humans , Ligands , Mice , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism
8.
Exp Hematol ; 41(2): 155-166.e17, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23261964

ABSTRACT

Red cell production is primarily determined by the action of erythropoietin. Additional erythropoiesis-regulatory factors include molecules and cellular interactions occurring within the bone marrow (BM) microenvironment. Sotatercept (ACE-011) is an activin receptor ligand trap that binds several members of the TGF-ß superfamily. Treatment with ACE-011 reverses bone loss and reduces the degree of osteoporosis, but it is accompanied by elevated hemoglobin and hematocrit levels. The mechanisms underlying the beneficial effects of ACE-011 on red cell production remain unknown. This study explores the means by which ACE-011 promotes erythropoiesis. We showed that ACE-011 does not directly affect erythroid differentiation of human CD34(+) cells in vitro. We next tested whether ACE-011 acts indirectly by affecting BM accessory cells. Conditioned media produced by BM stromal cells (SCs) inhibited erythroid differentiation of CD34(+) cells while maintained their ability to proliferate. However, conditioned media from SCs treated with ACE-011 partially restored erythropoiesis, coinciding with changes in the molecular and secretory profile of SCs, including the expression and secretion of erythropoiesis-modulatory factors. We conclude that inhibitory factors produced by BM SCs in vitro might control erythropoiesis in vivo and that agents that reverse these microenvironmental signals could provide an approach to attenuate anemia in clinical conditions.


Subject(s)
Activin Receptors, Type II/antagonists & inhibitors , Erythroid Precursor Cells/drug effects , Erythropoiesis/drug effects , Stromal Cells/drug effects , Adult , Blood Cells/cytology , Blood Cells/drug effects , Blood Cells/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Colony-Forming Units Assay , Culture Media, Conditioned/chemistry , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Drug Evaluation, Preclinical , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoiesis/physiology , Fetal Blood/cytology , Gene Expression Profiling , Humans , In Vitro Techniques , K562 Cells/cytology , K562 Cells/drug effects , K562 Cells/metabolism , Ligands , Organ Specificity , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins , Stromal Cells/physiology
9.
Expert Opin Investig Drugs ; 22(1): 87-101, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23127248

ABSTRACT

INTRODUCTION: Antagonists of activin receptor signaling may be beneficial for cancer-related anemia and bone disease caused by malignancies such as multiple myeloma and solid tumors. AREAS COVERED: We review evidence of dysregulated signaling by activin receptor pathways in anemia, myeloma-associated osteolysis, and metastatic bone disease, as well as potential involvement in carcinogenesis. We then review properties of activin receptor antagonists in clinical development. EXPERT OPINION: Sotatercept is a novel receptor fusion protein that functions as a soluble trap to sequester ligands of activin receptor type IIA (ActRIIA). Preclinically, the murine version of sotatercept increased red blood cells (RBC) in a model of chemotherapy-induced anemia, inhibited tumor growth and metastasis, and exerted anabolic effects on bone in diverse models of multiple myeloma. Clinically, sotatercept increases RBC markedly in healthy volunteers and patients with multiple myeloma. With a rapid onset of action differing from erythropoietin, sotatercept is in clinical development as a potential first-in-class therapeutic for cancer-related anemia, including those characterized by ineffective erythropoiesis as in myelodysplastic syndromes. Anabolic bone activity in early clinical studies and potential antitumor effects make sotatercept a promising therapeutic candidate for multiple myeloma and malignant bone diseases. Antitumor activity has been observed preclinically with small-molecule inhibitors of transforming growth factor-ß receptor type I (ALK5) that also antagonize the closely related activin receptors ALK4 and ALK7. LY-2157299, the first such inhibitor to enter clinical studies, has shown an acceptable safety profile so far in patients with advanced cancer. Together, these data identify activin receptor antagonists as attractive therapeutic candidates for multiple diseases.


Subject(s)
Activin Receptors/antagonists & inhibitors , Anemia/drug therapy , Bone Diseases/drug therapy , Neoplasms/drug therapy , Activin Receptors/metabolism , Anemia/etiology , Anemia/metabolism , Animals , Bone Diseases/etiology , Bone Diseases/metabolism , Humans , Neoplasms/complications , Neoplasms/metabolism
10.
J Pharmacol Exp Ther ; 341(2): 464-73, 2012 May.
Article in English | MEDLINE | ID: mdl-22338033

ABSTRACT

Antitumor anthracyclines such as doxorubicin and epirubicin are known to cause cardiotoxicity that correlates with anthracycline accumulation in the heart. The anthracycline amrubicin [(7S,9S)-9-acetyl-9-amino-7-[(2-deoxy-ß-d-erythro-pentopyranosyl)oxy]-7,8,9,10-tetrahydro-6,11-dihydroxy-5,12-napthacenedione hydrochloride] has not shown cardiotoxicity in laboratory animals or patients in approved or investigational settings; therefore, we conducted preclinical work to characterize whether amrubicin attained lower levels than doxorubicin or epirubicin in the heart. Anthracyclines were evaluated in ex vivo human myocardial strips incubated in plasma to which anthracycline concentrations of 3 or 10 µM were added. Four-hour incubations were performed to characterize myocardial anthracycline accumulation derived from anthracycline uptake in equilibrium with anthracycline clearance. Short-term incubations followed by multiple washouts were performed to obtain independent measurements of anthracycline uptake or clearance. In comparison with doxorubicin or epirubicin, amrubicin attained very low levels in the soluble and membrane fractions of human myocardial strips. This occurred at both 3 and 10 µM anthracycline concentrations and was caused primarily by a highly favorable clearance of amrubicin. Amrubicin clearance was facilitated by formation and elimination of sizeable levels of 9-deaminoamrubicin and 9-deaminoamrubicinol. Amrubicin clearance was not mediated by P glycoprotein or other drug efflux pumps, as judged from the lack of effect of verapamil on the partitioning of amrubicin and its deaminated metabolites across myocardial strips and plasma. Limited accumulation of amrubicin in an ex vivo human myocardial strip model may therefore correlate with the improved cardiac tolerability observed with the use of amrubicin in preclinical or clinical settings.


Subject(s)
Anthracyclines/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Myocardium/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Aged , Aged, 80 and over , Anthracyclines/pharmacology , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Epirubicin/pharmacokinetics , Epirubicin/pharmacology , Female , Humans , In Vitro Techniques , Male , Middle Aged , Verapamil/pharmacology
11.
J Pharmacol Exp Ther ; 341(2): 474-83, 2012 May.
Article in English | MEDLINE | ID: mdl-22338034

ABSTRACT

Anthracycline-related cardiotoxicity correlates with cardiac anthracycline accumulation and bioactivation to secondary alcohol metabolites or reactive oxygen species (ROS), such as superoxide anion (O2·â») and hydrogen peroxide H2O2). We reported that in an ex vivo human myocardial strip model, 3 or 10 µM amrubicin [(7S,9S)-9-acetyl-9-amino-7-[(2-deoxy-ß-D-erythro-pentopyranosyl)oxy]-7,8,9,10-tetrahydro-6,11-dihydroxy-5,12-napthacenedione hydrochloride] accumulated to a lower level compared with equimolar doxorubicin or epirubicin (J Pharmacol Exp Ther 341:464-473, 2012). We have characterized how amrubicin converted to ROS or secondary alcohol metabolite in comparison with doxorubicin (that formed both toxic species) or epirubicin (that lacked ROS formation and showed an impaired conversion to alcohol metabolite). Amrubicin and doxorubicin partitioned to mitochondria and caused similar elevations of H2O2, but the mechanisms of H2O2 formation were different. Amrubicin produced H2O2 by enzymatic reduction-oxidation of its quinone moiety, whereas doxorubicin acted by inducing mitochondrial uncoupling. Moreover, mitochondrial aconitase assays showed that 3 µM amrubicin caused an O2·â»-dependent reversible inactivation, whereas doxorubicin always caused an irreversible inactivation. Low concentrations of amrubicin therefore proved similar to epirubicin in sparing mitochondrial aconitase from irreversible inactivation. The soluble fraction of human myocardial strips converted doxorubicin and epirubicin to secondary alcohol metabolites that irreversibly inactivated cytoplasmic aconitase; in contrast, strips exposed to amrubicin failed to generate its secondary alcohol metabolite, amrubicinol, and only occasionally exhibited an irreversible inactivation of cytoplasmic aconitase. This was caused by competing pathways that favored formation and complete or near-to-complete elimination of 9-deaminoamrubicinol. These results characterize amrubicin metabolic advantages over doxorubicin and epirubicin, which may correlate with amrubicin cardiac safety in preclinical or clinical settings.


Subject(s)
Anthracyclines/metabolism , Anthracyclines/pharmacokinetics , Doxorubicin/metabolism , Doxorubicin/pharmacokinetics , Epirubicin/metabolism , Epirubicin/pharmacokinetics , Myocardium/metabolism , Aconitate Hydratase/metabolism , Alcohols/metabolism , Anthracyclines/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cytoplasm/metabolism , Doxorubicin/pharmacology , Epirubicin/pharmacology , Humans , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Troponin I/metabolism
12.
Cancer Chemother Pharmacol ; 69(4): 965-76, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22120960

ABSTRACT

PURPOSE: Multi-drug resistance and cumulative cardiotoxicity are major limitations for the clinical use of anthracyclines. Here, we evaluated and compared the cross-resistance of amrubicin, a third-generation synthetic anthracycline and potent topoisomerase (topo)-II inhibitor with little or no observed cardiotoxicity to other anthracyclines and the topo-II inhibitor etoposide in drug-resistant tumor models in order to elucidate its potential mechanisms of action. METHODS: Amrubicin activity was assessed in multi-drug-resistant cell lines and human tumor explants using cytotoxicity assays, confocal microscopy, fluorescence time-lapse imaging, flow cytometry, immunoblotting, and gene expression profiling techniques. RESULTS: We demonstrate that both doxorubicin-resistant tumor cell lines and several drug-resistant human ovarian and breast tumor explants retain sensitivity to amrubicin. In addition, we observed similar levels of amrubicin uptake and accumulation in doxorubicin-sensitive versus doxorubicin-resistant cell lines. Although amrubicin is a weak P-glycoprotein substrate, transport and retention of amrubicin were not solely modulated by P-glycoprotein in the resistant cell lines overexpressing drug efflux pumps. The cellular retention of amrubicin is likely to be a result of rapid influx due to its high intrinsic permeability and lipophilic properties, and this may explain why amrubicin overcomes pleiotropic drug resistance. Consistent with drug accumulation studies, amrubicin induced DNA damage, G(2)-M cell cycle arrest, and apoptosis in both doxorubicin-sensitive and doxorubicin-resistant lines. Using gene expression profiling studies, several classes of genes were significantly and uniquely regulated following amrubicin, but not doxorubicin or etoposide, treatment. CONCLUSIONS: Amrubicin appears to have a distinct mode of action that overcomes typical anthracycline resistance mechanisms. Therefore, amrubicin may be useful in the treatment of anthracycline-refractory or anthracycline-resistant tumors.


Subject(s)
Anthracyclines/pharmacology , Anthracyclines/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Caspase 3/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , DNA Damage , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Epirubicin/pharmacology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Topoisomerase II Inhibitors/pharmacokinetics , Topoisomerase II Inhibitors/pharmacology , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
13.
Cancer Sci ; 102(6): 1201-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21375679

ABSTRACT

Histone deacetylase inhibitors are a group of recently developed compounds that modulate cell growth and survival. We evaluated the effects of the histone deacetylase inhibitor MGCD0103 on growth of pancreatic carcinoma models following single agent treatment and in combination with gemcitabine. MGCD0103 inhibited tumor cell growth and acted synergistically with gemcitabine to enhance its cytotoxic effects. Gene expression analysis identified the cell cycle pathway as one of the most highly modulated gene groups. Our data suggest that MGCD0103 + gemcitabine might be an effective treatment for gemcitabine-refractory pancreatic cancer.


Subject(s)
Benzamides/pharmacology , Deoxycytidine/analogs & derivatives , Histone Deacetylase Inhibitors/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pyrimidines/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Drug Synergism , Gene Expression , Humans , Immunoblotting , Pancreas/enzymology , Gemcitabine
14.
Cancer Res ; 63(12): 3356-63, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12810671

ABSTRACT

MST4, a member of the Sterile 20 serine/threonine kinase family, was found to be expressed in prostate carcinoma tumor samples and cell lines. In addition, expression levels appeared to correlate with tumorigenicity and androgen receptor status of the cells. Ectopic expression of wild-type and kinase-inactive MST4 was used to alter cellular MST4 activity levels in three widely studied human prostate tumor cell lines: LNCaP, DU 145, and PC-3. Overexpression of wild-type MST4 induced anchorage-independent growth of the LNCaP cell line, and increased both in vitro proliferation and in vivo tumorigenesis of the DU 145 cell line. On the other hand, expression of a kinase-inactive form reverted the anchorage-independent growth phenotype and highly tumorigenic behavior of the PC-3 cell line. MST4 kinase activity was stimulated significantly by epidermal growth factor receptor ligands, which are known to promote growth of prostate cancer cells. Together, our studies suggest a potential role for MST4 in the signal transduction pathways involved in prostate cancer progression.


Subject(s)
Adenocarcinoma/enzymology , Neoplasm Proteins/physiology , Prostatic Neoplasms/enzymology , Protein Serine-Threonine Kinases/physiology , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Adult , Cell Adhesion , Cell Division , Disease Progression , Enzyme Induction , Epidermal Growth Factor/pharmacology , Extracellular Matrix/metabolism , Humans , Lymphatic Metastasis , Male , Neoplasm Proteins/genetics , Organ Specificity , Prostate/enzymology , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/physiology , Transfection , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...