Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Int J Hematol ; 117(1): 100-109, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36175779

ABSTRACT

The safety, efficacy, and pharmacokinetics of copanlisib were evaluated in this phase Ib/II study in Japanese patients with relapsed/refractory indolent non-Hodgkin lymphoma (NHL). The primary endpoint was safety at the recommended dose; efficacy endpoints included objective response rate (ORR), progression-free survival (PFS), and overall survival. In phase Ib, patients received copanlisib 45 mg intravenously on days 1, 8, and 15 of a 28-day cycle, and when tolerated, consecutive patients received copanlisib 60 mg. As no dose-limiting toxicities occurred at the 45 mg (n = 3) or 60 mg (n = 7) dose in phase Ib, the recommended dose for Japanese patients was determined to be 60 mg, and this dose was used in phase II (n = 15). Although all patients experienced at least one treatment-emergent adverse event (TEAE), with hyperglycemia being the most common AE, no AE-related deaths were reported. The ORR was 68.0% (17/25 patients), median PFS was 302 (95% CI 231-484) days, and the duration of response was 330 (range 65-659) days. The pharmacokinetic properties of copanlisib were similar between Japanese and non-Japanese patients. Overall, copanlisib 60 mg had an acceptable safety profile and showed promising antitumor activity in Japanese patients with relapsed/refractory indolent NHL.


Subject(s)
Lymphoma, Non-Hodgkin , Neoplasm Recurrence, Local , Quinazolines , Humans , Antineoplastic Agents/adverse effects , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/pathology , Neoplasm Recurrence, Local/drug therapy , Quinazolines/adverse effects
2.
Tissue Eng Part A ; 24(3-4): 287-300, 2018 02.
Article in English | MEDLINE | ID: mdl-28498040

ABSTRACT

Although engineered cardiac tissues (ECTs) derived from induced pluripotent stem cells (iPSCs) are promising for myocardial regenerative therapy, the appropriate ratio of cardiomyocytes to non-cardiomyocytes is not fully understood. Here, we determined whether ECT-cell content is a key determinant of its structure/function, thereby affecting ECT therapeutic potential for advanced heart failure. Scaffold-free ECTs containing different ratios (25%, 50%, 70%, or 90%) of iPSC-derived cardiomyocytes were generated by magnetic-activated cell sorting by using cardiac-specific markers. Notably, ECTs showed synchronized spontaneous beating when cardiomyocytes constituted ≥50% of total cells, with the electrical-conduction velocity increasing depending on cardiomyocyte ratio; however, ECTs containing 90% cardiomyocytes failed to form stable structures. ECTs containing 25% or 50% cardiomyocytes predominantly expressed collagen and fibronectin, whereas ECTs containing 70% cardiomyocytes predominantly expressed laminin and exhibited the highest contractile/relaxation properties. Furthermore, transplantation of ECTs containing 50% or 70% cardiomyocytes into a rat chronic myocardial infarction model led to a more profound functional recovery as compared with controls. Notably, transplanted ECTs showed electrical synchronization with the native heart under Langendorff perfusion. Collectively, these results indicate that the quantity of non-cardiomyocytes is critical in generating functional iPSC-derived ECTs as grafts for cardiac-regeneration therapy, with ECTs containing 50-70% cardiomyocytes exhibiting stable structures and increased cardiotherapeutic potential.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myocardium/cytology , Myocytes, Cardiac/cytology , Cell Differentiation/physiology , Cells, Cultured , Electrophysiology , Flow Cytometry , Humans , Tissue Engineering/methods
6.
Sci Rep ; 6: 19464, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26763872

ABSTRACT

Transplantation of induced pluripotent stem cell-derived cardiac tissue constructs is a promising regenerative treatment for cardiac failure: however, its tumourigenic potential is concerning. We hypothesised that the tumourigenic potential may be eliminated by the host immune response after allogeneic cell transplantation. Scaffold-free iPSC-derived cardaic tissue sheets of C57BL/6 mouse origin were transplanted into the cardiac surface of syngeneic C57BL/6 mice and allogeneic BALB/c mice with or without tacrolimus injection. Syngeneic mice and tacrolimus-injected immunosuppressed allogeneic mice formed teratocarcinomas with identical phenotypes, characteristic, and time courses, as assessed by imaging tools including (18)F-fluorodeoxyglucose-positron emission tomography. In contrast, temporarily immunosuppressed allogeneic mice, following cessation of tacrolimus injection displayed diminished progression of the teratocarcinoma, accompanied by an accumulation of CD4/CD8-positive T cells, and finally achieved complete elimination of the teratocarcinoma. Our results indicated that malignant teratocarcinomas arising from induced pluripotent stem cell-derived cardiac tissue constructs provoked T cell-related host immune rejection to arrest tumour growth in murine allogeneic transplantation models.


Subject(s)
Cell Transformation, Neoplastic , Graft Rejection/immunology , Induced Pluripotent Stem Cells , Myocytes, Cardiac/pathology , Stem Cell Transplantation/adverse effects , Teratocarcinoma/etiology , Teratocarcinoma/pathology , Animals , Biopsy , Cell Line , Fluorodeoxyglucose F18 , Gene Order , Genetic Vectors/genetics , Immunocompromised Host , Immunosuppressive Agents/administration & dosage , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Transgenic , Models, Animal , Myocytes, Cardiac/cytology , Positron-Emission Tomography , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tacrolimus/administration & dosage , Teratocarcinoma/diagnosis , Transplantation, Homologous
7.
Curr Gene Ther ; 16(1): 5-13, 2016.
Article in English | MEDLINE | ID: mdl-26785736

ABSTRACT

Advanced cardiac failure is a progressive intractable disease and is the main cause of mortality and morbidity worldwide. Since this pathology is represented by a definite decrease in cardiomyocyte number, supplementation of functional cardiomyocytes into the heart would hypothetically be an ideal therapeutic option. Recently, unlimited in vitro production of human functional cardiomyocytes was established by using induced pluripotent stem cell (iPSC) technology, which avoids the use of human embryos. A number of basic studies including ours have shown that transplantation of iPSCderived cardiomyocytes (iPSC-CMs) into the damaged heart leads to recovery of cardiac function, thereby establishing "proof-of-concept" of this iPSC-transplantation therapy. However, considering clinical application of this therapy, its feasibility, safety, and therapeutic efficacy need to be further investigated in the pre-clinical stage. This review summarizes up-to-date important topics related to safety and efficacy of iPSC-CMs transplantation therapy for cardiac disease and discusses the prospects for this treatment in clinical studies.


Subject(s)
Cell Culture Techniques/methods , Heart Failure/therapy , Induced Pluripotent Stem Cells/transplantation , Myocytes, Cardiac/transplantation , Bioreactors , Cell Culture Techniques/instrumentation , Cell Differentiation , Cell- and Tissue-Based Therapy/methods , Humans , Induced Pluripotent Stem Cells/immunology , Myocytes, Cardiac/cytology
13.
Nat Commun ; 5: 4330, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24999918

ABSTRACT

Fanconi anaemia (FA) is a recessive disorder characterized by genomic instability, congenital abnormalities, cancer predisposition and bone marrow (BM) failure. However, the pathogenesis of FA is not fully understood partly due to the limitations of current disease models. Here, we derive integration free-induced pluripotent stem cells (iPSCs) from an FA patient without genetic complementation and report in situ gene correction in FA-iPSCs as well as the generation of isogenic FANCA-deficient human embryonic stem cell (ESC) lines. FA cellular phenotypes are recapitulated in iPSCs/ESCs and their adult stem/progenitor cell derivatives. By using isogenic pathogenic mutation-free controls as well as cellular and genomic tools, our model serves to facilitate the discovery of novel disease features. We validate our model as a drug-screening platform by identifying several compounds that improve hematopoietic differentiation of FA-iPSCs. These compounds are also able to rescue the hematopoietic phenotype of FA patient BM cells.


Subject(s)
Drug Evaluation, Preclinical/methods , Fanconi Anemia/etiology , Fanconi Anemia/pathology , Models, Biological , Stem Cells/pathology , Cell Differentiation , Epigenesis, Genetic , Fanconi Anemia/drug therapy , Fanconi Anemia Complementation Group A Protein/genetics , Humans , Induced Pluripotent Stem Cells , Male , Young Adult
16.
J Mol Cell Biol ; 5(5): 354-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24014886

ABSTRACT

Induced pluripotent stem cells (iPSCs) could be generated by a single gene Oct4 and chemical compounds, in which exogenous expression of Oct4 was indispensable for reprogramming. Recent advances in chemical-mediated cellular reprogramming suggest that small molecules alone (i.e. without Yamanaka factors) can successfully establish iPSCs from mouse somatic cells.


Subject(s)
Colforsin/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Transgenes/genetics , Animals , Cellular Reprogramming/drug effects , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Mice , Octamer Transcription Factor-3/metabolism
19.
Cell Res ; 23(4): 458-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23357848

ABSTRACT

Maintenance of hematopoietic stem cells (HSCs) in vitro has been believed to be difficult due to a lack of complete understanding of HSC quiescence maintained by the niche. Recent evidence suggests that in vitro maintenance of human and mouse long-term HSCs (LT-HSCs) is possible through dual inhibition (2i) of both GSK-3 and mTOR in the absence of cytokines, serum, or feeder cells.


Subject(s)
Cell Proliferation/drug effects , Hematopoietic Stem Cells/cytology , Homeostasis/physiology , TOR Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway/physiology , Animals , Humans
20.
Cell Res ; 23(2): 176-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23044804

ABSTRACT

Blood transfusion medicine requires a constant supply of platelets, which is now totally donor dependent. Recent advances in the generation of platelets in vitro through megakaryocytes (MKs) may provide protocols not only via pluripotent stem cells, including induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs), but also via induced MKs (iMKs). For the first time, mouse and human fibroblasts are successfully transdifferentiated into iMKs by the introduction of three factors, p45NF-E2, Maf G, and Maf K.


Subject(s)
Cell Transdifferentiation , Megakaryocytes/cytology , 3T3 Cells , Animals , Blood Platelets/cytology , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Megakaryocytes/metabolism , Mice , NF-E2 Transcription Factor, p45 Subunit/genetics , NF-E2 Transcription Factor, p45 Subunit/metabolism , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism , Proto-Oncogene Proteins c-myc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...