Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Am J Hum Genet ; 107(6): 1149-1156, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33186543

ABSTRACT

The Congenital Dyserythropoietic Anemia (CDA) Registry was established with the goal to facilitate investigations of natural history, biology, and molecular pathogenetic mechanisms of CDA. Three unrelated individuals enrolled in the registry had a syndrome characterized by CDA and severe neurodevelopmental delay. They were found to have missense mutations in VPS4A, a gene coding for an ATPase that regulates the ESCRT-III machinery in a variety of cellular processes including cell division, endosomal vesicle trafficking, and viral budding. Bone marrow studies showed binucleated erythroblasts and erythroblasts with cytoplasmic bridges indicating abnormal cytokinesis and abscission. Circulating red blood cells were found to retain transferrin receptor (CD71) in their membrane, demonstrating that VPS4A is critical for normal reticulocyte maturation. Using proband-derived induced pluripotent stem cells (iPSCs), we have successfully modeled the hematologic aspects of this syndrome in vitro, recapitulating their dyserythropoietic phenotype. Our findings demonstrate that VPS4A mutations cause cytokinesis and trafficking defects leading to a human disease with detrimental effects to erythropoiesis and neurodevelopment.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Anemia, Dyserythropoietic, Congenital/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Vacuolar Proton-Translocating ATPases/genetics , Adenosine Triphosphatases/metabolism , Anemia, Dyserythropoietic, Congenital/pathology , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Child , Child, Preschool , Cytokinesis , Endosomes/metabolism , Erythroblasts/metabolism , Erythrocytes/cytology , Erythropoiesis , Female , Humans , Induced Pluripotent Stem Cells/cytology , Male , Neurodevelopmental Disorders/metabolism , Phenotype , Protein Transport , Reticulocytes/cytology
2.
Br J Haematol ; 176(2): 300-308, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27766637

ABSTRACT

'Paradoxical' embolization via intracardiac or intrapulmonary right-to-left shunts (RLS) is an established cause of stroke. Hypercoagulable states and increased right heart pressure, which both occur in sickle cell anaemia (SCA), predispose to paradoxical embolization. We hypothesized that children with SCA and overt stroke (SCA + stroke) have an increased prevalence of potential RLS. We performed contrasted transthoracic echocardiograms on 147 children (aged 2-19 years) with SCA + stroke) mean age 12·7 ± 4·8 years, 54·4% male) and a control group without SCA or stroke (n = 123; mean age 12·1 ± 4·9 years, 53·3% male). RLS was defined as any potential RLS detected by any method, including intrapulmonary shunting. Echocardiograms were masked and adjudicated centrally. The prevalence of potential RLS was significantly higher in the SCA+stroke group than controls (45·6% vs. 23·6%, P < 0·001). The odds ratio for potential RLS in the SCA + stroke group was 2·7 (95% confidence interval: 1·6-4·6) vs controls. In post hoc analyses, the SCA + stroke group had a higher prevalence of intrapulmonary (23·8% vs. 5·7%, P < 0·001) but not intracardiac shunting (21·8% vs. 18·7%, P = 0·533). SCA patients with potential RLS were more likely to report headache at stroke onset than those without. Intrapulmonary and intracardiac shunting may be an overlooked, independent and potentially modifiable risk factor for stroke in SCA.


Subject(s)
Anemia, Sickle Cell/complications , Heart Septal Defects/diagnostic imaging , Stroke/etiology , Adolescent , Anemia, Sickle Cell/epidemiology , Child , Child, Preschool , Echocardiography , Embolism, Paradoxical/etiology , Female , Headache/etiology , Heart Septal Defects/complications , Humans , Male , Prevalence , Risk Factors , Young Adult
3.
MAbs ; 11(2): 422-433, 2019.
Article in English | MEDLINE | ID: mdl-30550367

ABSTRACT

Bispecific antibody production using single host cells has been a new advancement in the antibody engineering field. We previously showed comparable in vitro biological activity and in vivo mouse pharmacokinetics (PK) for two novel single cell variants (v10 and v11) and one traditional dual cell in vitro-assembled anti-human epidermal growth factor receptor 2/CD3 T-cell dependent bispecific (TDB) antibodies. Here, we extended our previous work to assess single cell-produced bispecific variants of a novel TDB against FcRH5, a B-cell lineage marker expressed on multiple myeloma (MM) tumor cells. An in vitro-assembled anti- FcRH5/CD3 TDB antibody was previously developed as a potential treatment option for MM. Two bispecific antibody variants (designs v10 and v11) for manufacturing anti-FcRH5/CD3 TDB in single cells were compared to in vitro-assembled TDB in a dual-cell process to understand whether differences in antibody design and production led to any major differences in their in vitro biological activity, in vivo mouse PK, and PK/pharmacodynamics (PD) or immunogenicity in cynomolgus monkeys (cynos). The binding, in vitro potencies, in vitro pharmacological activities and in vivo PK in mice and cynos of these single cell TDBs were comparable to those of the in vitro-assembled TDB. In addition, the single cell and in vitro-assembled TDBs exhibited robust PD activity and comparable immunogenicity in cynos. Overall, these studies demonstrate that single cell-produced and in vitro-assembled anti-FcRH5/CD3 T-cell dependent bispecific antibodies have similar in vitro and in vivo properties, and support further development of single-cell production method for anti-FcRH5/CD3 TDBs and other single-cell bispecifics.


Subject(s)
Antibodies, Bispecific/chemistry , Antibodies, Bispecific/pharmacokinetics , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/pharmacokinetics , Receptors, Fc/chemistry , Animals , Antibodies, Bispecific/immunology , Antibodies, Monoclonal, Humanized/immunology , CD3 Complex/immunology , Drug Design , Humans , In Vitro Techniques , Macaca fascicularis , Mice , Multiple Myeloma , T-Lymphocytes/immunology
4.
MAbs ; 8(3): 593-603, 2016.
Article in English | MEDLINE | ID: mdl-26918260

ABSTRACT

MPDL3280A is a human monoclonal antibody that targets programmed cell death-1 ligand 1 (PD-L1), and exerts anti-tumor activity mainly by blocking PD-L1 interaction with programmed cell death-1 (PD-1) and B7.1. It is being investigated as a potential therapy for locally advanced or metastatic malignancies. The purpose of the study reported here was to characterize the pharmacokinetics, pharmacodynamics, tissue distribution and tumor penetration of MPDL3280A and/or a chimeric anti-PD-L1 antibody PRO304397 to help further clinical development. The pharmacokinetics of MPDL3280A in monkeys at 0.5, 5 and 20 mg · kg(-1) and the pharmacokinetics / pharmacodynamics of PRO304397 in mice at 1, 3 10 mg · kg(-1) were determined after a single intravenous dose. Tissue distribution and tumor penetration for radiolabeled PRO304397 in tumor-bearing mouse models were determined. The pharmacokinetics of MPDL3280A and PRO304397 were nonlinear in monkeys and mice, respectively. Complete saturation of PD-L1 in blood in mice was achieved at serum concentrations of PRO304397 above ∼ 0.5 µg · mL(-1). Tissue distribution and tumor penetration studies of PRO304397 in tumor-bearing mice indicated that the minimum tumor interstitial to plasma radioactivity ratio was ∼ 0.3; saturation of target-mediated uptake in non-tumor tissues and desirable exposure in tumors were achieved at higher serum concentrations, and the distribution into tumors was dose-and time-dependent. The biodistribution data indicated that the efficacious dose is mostly likely higher than that estimated based on simple pharmacokinetics/pharmacodynamics in blood. These data also allowed for estimation of the target clinical dose for further development of MPDL3280A.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neoplasm , B7-H1 Antigen/antagonists & inhibitors , Cell Cycle Checkpoints , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , B7-H1 Antigen/immunology , CHO Cells , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/immunology , Cricetinae , Cricetulus , Drug Evaluation, Preclinical , Female , Humans , Macaca fascicularis , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasm Proteins/immunology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology
5.
Sci Transl Med ; 7(287): 287ra70, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25972002

ABSTRACT

Bispecific antibodies and antibody fragments in various formats have been explored as a means to recruit cytolytic T cells to kill tumor cells. Encouraging clinical data have been reported with molecules such as the anti-CD19/CD3 bispecific T cell engager (BiTE) blinatumomab. However, the clinical use of many reported T cell-recruiting bispecific modalities is limited by liabilities including unfavorable pharmacokinetics, potential immunogenicity, and manufacturing challenges. We describe a B cell-targeting anti-CD20/CD3 T cell-dependent bispecific antibody (CD20-TDB), which is a full-length, humanized immunoglobulin G1 molecule with near-native antibody architecture constructed using "knobs-into-holes" technology. CD20-TDB is highly active in killing CD20-expressing B cells, including primary patient leukemia and lymphoma cells both in vitro and in vivo. In cynomolgus monkeys, CD20-TDB potently depletes B cells in peripheral blood and lymphoid tissues at a single dose of 1 mg/kg while demonstrating pharmacokinetic properties similar to those of conventional monoclonal antibodies. CD20-TDB also exhibits activity in vitro and in vivo in the presence of competing CD20-targeting antibodies. These data provide rationale for the clinical testing of CD20-TDB for the treatment of CD20-expressing B cell malignancies.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antigens, CD20/immunology , CD3 Complex/immunology , Leukemia, B-Cell/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacokinetics , Humans , Leukemia, B-Cell/immunology , Macaca fascicularis , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL