Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
AAPS J ; 22(2): 52, 2020 02 27.
Article in English | MEDLINE | ID: mdl-32107671

ABSTRACT

Biologic-device combination products using prefilled syringes (PFSs) and autoinjectors (AIs) are popular for biological products administered subcutaneously. Pharmacokinetic (PK) comparability studies commonly provide the scientific data to support introduction of AI presentations via bridging with PFS. A survey of biological products approved by FDA's Center for Drug Evaluation and Research identified 17 biologics license applications (BLAs) with both PFS and AI presentations for subcutaneous (SC) administration, including 16 approved on February 1, 2018, and one with AI presentation under review. A systematic review on the device parameters and the PK comparability studies bridging the two presentations was conducted. Subsequently, whether device parameters or the PK study design may have influenced the PK comparability study results was evaluated. The reported device parameters for AI and PFS are generally consistent across BLAs, whereas the approach to assess PK comparability varied, including the study design. Most PK comparability studies met bioequivalence (BE) criteria. Upon inspection of the studies that did not meet BE criteria, injection depth of AI and the injection site for either AI or PFS were identified as potential influencing factors to the outcome of PK comparability study. This study represents an initial attempt to identify the potential influencing factors on device bridging, including the characteristics of the device and the clinical pharmacology study. These findings may inform the combination product development strategy, specifically design considerations for device and PK comparability studies.


Subject(s)
Biological Products/administration & dosage , Biological Products/pharmacokinetics , Drug Delivery Systems/instrumentation , Needles , Syringes , Drug Compounding , Equipment Design , Female , Humans , Injections, Subcutaneous , Male , Therapeutic Equivalency , Tissue Distribution , Viscosity
2.
J Cell Physiol ; 233(11): 8691-8700, 2018 11.
Article in English | MEDLINE | ID: mdl-29761851

ABSTRACT

Plasma- and platelet-derived factor Va are essential for thrombin generation catalyzed by the prothrombinase complex; however, several observations demonstrate that the platelet-derived cofactor, which is formed following megakaryocyte endocytosis and modification of the plasma procofactor, factor V, is more hemostatically relevant. Factor V endocytosis, as a function of megakaryocyte differentiation and proplatelet formation, was assessed by flow cytometry and microscopy in CD34+ hematopoietic progenitor cells isolated from human umbilical cord blood and cultured for 12 days in the presence of cytokines to induce ex vivo differentiation into megakaryocytes. Expression of an early marker of megakaryocyte differentiation, CD41, endocytosis of factor V, and the percentage of CD41+ cells that endocytosed factor V increased from days 6 to 12 of differentiation. In contrast, statistically significant decreases in expression of the stem cell marker, CD34, and in the percentage of CD34+ cells that endocytosed factor V were observed. A statistically significant increase in the expression of CD42b, a late marker of megakaryocyte differentiation, was also observed over time, such that by Day 12, all CD42b+ cells endocytosed factor V and expressed CD41. This endocytosed factor V was trafficked to proplatelet extensions and was localized in a punctate pattern in the cytoplasm consistent with its storage in α-granules. In conclusion, loss of CD34 and expression of CD42b define cells capable of factor V endocytosis and trafficking to proplatelet extensions during differentiation of megakaryocytes ex vivo from progenitor cells isolated from umbilical cord blood.


Subject(s)
Antigens, CD34/genetics , Cell Differentiation/genetics , Factor V/genetics , Platelet Glycoprotein GPIb-IX Complex/genetics , Blood Platelets/metabolism , Cell Movement/genetics , Endocytosis/genetics , Fetal Blood/cytology , Fetal Blood/metabolism , Gene Expression Regulation/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Thrombopoietin/genetics
3.
Cytometry A ; 91(7): 713-720, 2017 07.
Article in English | MEDLINE | ID: mdl-28692777

ABSTRACT

Differentiating megakaryocytes undergo a unique endomitotic cell cycle leading to large polyploidal cells, which fragment to generate platelets, blood cells important for normal hemostasis. Simultaneous assessment of DNA content and cellular proteins by flow cytometry is a useful tool to study megakaryocyte differentiation and to define expression of proteins important for megakaryocyte development and platelet formation. The usefulness of zinc salt-based fixation (ZBF), a non-crosslinking method of cell fixation that permits downstream analysis of nucleic acids (Jensen et al., Cytometry A 2010;77A:798-804), in flow cytometric analysis of megakaryocyte ploidy in conjunction with extracellular and intracellular proteins was assessed. ZBF of a megakaryocyte-like cell line resulted in preservation of proteins similar to paraformaldehyde fixation, and preservation of DNA content in a manner similar to methanol fixation. This is highlighted by experiments in which polyploidal megakaryocytes were analyzed simultaneously for endocytosis of a fluorescently-labeled, endocytosed labile protein or expression of a cell surface integrin and DNA content. These studies demonstrate that ZBF will be a valuable tool to study the molecular events leading to platelet formation. © 2017 International Society for Advancement of Cytometry.


Subject(s)
Blood Platelets/cytology , Flow Cytometry , Megakaryocytes/cytology , Cell Division/physiology , Cell Membrane/metabolism , Cytoplasm/metabolism , Flow Cytometry/methods , Hematopoiesis/physiology , Humans , Zinc
4.
J Cell Biochem ; 116(10): 2121-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25800007

ABSTRACT

Factor Va serves as the nonenzymatic protein cofactor for the prothrombinase complex, which converts prothrombin to thrombin in the events leading to formation of a hemostatic plug. Several observations support the concept that platelet-derived factor V/Va is physically and functionally distinct and plays a more important role in thrombin generation at sites of vascular injury as compared to its plasma counterpart. Platelet-derived factor V/Va is generated following endocytosis of the plasma-derived molecule by the platelet precursor cells, megakaryocytes, via a two receptor system consisting of low density lipoprotein (LDL) receptor-related protein-1 (LRP-1) and an unidentified specific "binding site". More recently, it was suggested that a cell surface-expressed ß-galactoside binding protein, galectin-8, was involved in factor V endocytosis. Endocytosed factor V is trafficked through the cell and retailored prior to its storage in α-granules. Given the essential role of platelet-derived factor Va in clot formation, understanding the cellular and molecular mechanisms that regulate how platelets acquire this molecule will be important for the treatment of excessive bleeding or clotting.


Subject(s)
Blood Coagulation Factors/metabolism , Galectins/metabolism , Hemorrhage/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Blood Coagulation/genetics , Blood Coagulation Factors/genetics , Blood Platelets/metabolism , Blood Platelets/pathology , Endocytosis/genetics , Hemorrhage/metabolism , Hemorrhage/pathology , Humans , Megakaryocytes/metabolism , Thrombin/metabolism , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
5.
Methods Mol Biol ; 788: 91-100, 2012.
Article in English | MEDLINE | ID: mdl-22130702

ABSTRACT

Hemostasis is dependent upon the successful recruitment and activation of blood platelets to the site of a breach in the vasculature. Platelet activation stimulates the rapid reorganization of the cortical actin cytoskeleton, resulting in the transformation of platelets from biconcave disks to fully spread cells. During this process, platelets extend filopodia and generate lamellipodia, resulting in a dramatic increase in the platelet surface area. Kohler-illuminated Nomarski Differential Interference Contrast microscopy has proved an effective tool to characterize platelet morphological changes in real time, and provides a useful tool to identify genetic and pharmacological regulators of platelet function.


Subject(s)
Blood Platelets/cytology , Cell Shape/physiology , Blood Platelets/physiology , Cell Adhesion/physiology , Cell Separation , Humans
6.
J Virol ; 84(10): 4912-22, 2010 May.
Article in English | MEDLINE | ID: mdl-20200236

ABSTRACT

Ranaviruses such as frog virus 3 ([FV3] family Iridoviridae) are increasingly prevalent pathogens that infect reptiles, amphibians, and fish worldwide. Whereas studies in the frog Xenopus laevis have revealed the critical involvement of CD8 T-cell and antibody responses in host resistance to FV3, little is known about the role played by innate immunity to infection with this virus. We have investigated the occurrence, composition, activation status, and permissiveness to infection of peritoneal leukocytes (PLs) in Xenopus adults during FV3 infection by microscopy, flow cytometry, and reverse transcription-PCR. The total number of PLs and the relative fraction of activated mononucleated macrophage-like cells significantly increase as early as 1 day postinfection (dpi), followed by NK cells at 3 dpi, before the peak of the T-cell response at 6 dpi. FV3 infection also induces a rapid upregulation of proinflammatory genes including arginase 1, interleukin-1beta, and tumor necrosis factor alpha. Although PLs are susceptible to FV3 infection, as evidenced by apoptotic cells, active FV3 transcription, and the detection of viral particles by electron microscopy, the infection is weaker (fewer infectious particles), more transitory, and involves a smaller fraction (less than 1%) of PLs than the kidney, the main site of infection. However, viral DNA remains detectable in PLs for at least 3 weeks postinfection, past the point of viral clearance observed in the kidneys. This suggests that although PLs are actively involved in anti-FV3 immune responses, some of these cells can be permissive and harbor quiescent, asymptomatic FV3.


Subject(s)
DNA Virus Infections/veterinary , Immunity, Innate , Leukocytes/immunology , Leukocytes/virology , Ranavirus/immunology , Xenopus laevis/immunology , Xenopus laevis/virology , Animals , DNA Virus Infections/immunology , Flow Cytometry , Gene Expression Profiling , Microscopy , Peritoneum/immunology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
7.
Arterioscler Thromb Vasc Biol ; 30(3): 591-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20042710

ABSTRACT

OBJECTIVE: To investigate the presence and role of NF-kappaB proteins in megakaryocytes and platelets. The nuclear factor-kappaB (NF-kappaB) transcription factor family is well known for its role in eliciting inflammation and promoting cell survival. We discovered that human megakaryocytes and platelets express the majority of NF-kappaB family members, including the regulatory inhibitor-kappaB (I-kappaB) and I-kappa kinase (IKK) molecules. METHODS AND RESULTS: Anucleate platelets exposed to NF-kappaB inhibitors demonstrated impaired fundamental functions involved in repairing vascular injury and thrombus formation. Specifically, NF-kappaB inhibition diminished lamellapodia formation, decreased clot retraction times, and reduced thrombus stability. Moreover, inhibition of I-kappaB-alpha phosphorylation (BAY-11-7082) reverted fully spread platelets back to a spheroid morphology. Addition of recombinant IKK-beta or I-kappaB-alpha protein to BAY inhibitor-treated platelets partially restored platelet spreading in I-kappaB-alpha inhibited platelets, and addition of active IKK-beta increased endogenous I-kappaB-alpha phosphorylation levels. CONCLUSIONS: These novel findings support a crucial and nonclassical role for the NF-kappaB family in modulating platelet function and reveal that platelets are sensitive to NF-kappaB inhibitors. As NF-kappaB inhibitors are being developed as antiinflammatory and anticancer agents, they may have unintended effects on platelets. On the basis of these data, NF-kappaB is also identified as a new target to dampen unwanted platelet activation.


Subject(s)
Blood Platelets/metabolism , Megakaryocytes/metabolism , NF-kappa B/metabolism , Adult , Aged , Blood Platelets/drug effects , Blood Platelets/pathology , Cell Adhesion/drug effects , Cell Differentiation , Cell Line, Tumor , Cell Shape/drug effects , Female , Fetal Blood/cytology , Fetal Blood/metabolism , Humans , Leukemia, Megakaryoblastic, Acute/metabolism , Leukemia, Megakaryoblastic, Acute/pathology , Male , Megakaryocytes/drug effects , Megakaryocytes/pathology , Middle Aged , NF-kappa B/antagonists & inhibitors , NF-kappa B p50 Subunit/metabolism , Nitriles/pharmacology , Sulfones/pharmacology , Transcription Factor RelA/metabolism
8.
Arterioscler Thromb Vasc Biol ; 29(10): 1602-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19661487

ABSTRACT

OBJECTIVE: Factor XI (FXI) promotes hemostasis and thrombosis through enhancement of thrombin generation and has been shown to play a critical role in the formation of occlusive thrombi in arterial injury models. The aim of this study was to investigate the mechanisms governing interactions between FXI and platelets. METHODS AND RESULTS: Platelet adhesion to immobilized FXI was abrogated in the presence of the low-density lipoprotein (LDL) receptor antagonist, receptor-associated protein (RAP), soluble recombinant apolipoprotein E receptor 2 (ApoER2), or the LDL-binding domain 1 or 2 of ApoER2. FXI supported wild-type murine platelet binding; in contrast, ApoER2-deficient murine platelets did not adhere to FXI. In the presence of shear, platelet aggregates formed on FXI or activated FXI (FXIa) surfaces, whereas the presence of RAP, binding domain 1 of ApoER2, or an anti-GPIb alpha mAb blocked platelet adhesion to FXI or FXIa under shear. Soluble FXI bound to immobilized ApoER2' with an affinity of 61 nmol/L. CONCLUSIONS: This study has identified apolipoprotein E receptor 2 (ApoER2, LRP8), a member of the LDL receptor family, as a platelet receptor for FXI. The interaction of FXI with other cell types that express ApoER2 remains to be explored.


Subject(s)
Factor XI/metabolism , Receptors, Lipoprotein/metabolism , Animals , Blood Platelets/metabolism , Calcium/metabolism , Humans , Kininogen, High-Molecular-Weight/pharmacology , LDL-Receptor Related Proteins , Ligands , Mice , Mice, Inbred C57BL , Platelet Adhesiveness , Zinc/pharmacology
9.
Blood ; 112(10): 4051-60, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18755987

ABSTRACT

Thrombocytopenia is a critical problem that occurs in many hematologic diseases, as well as after cancer therapy and radiation exposure. Platelet transfusion is the most commonly used therapy but has limitations of alloimmunization, availability, and expense. Thus, the development of safe, small, molecules to enhance platelet production would be advantageous for the treatment of thrombocytopenia. Herein, we report that an important lipid mediator and a peroxisome proliferator-activated receptor gamma (PPARgamma) ligand called 15-deoxy-Delta(12,14) prostaglandin J(2) (15d-PGJ(2)), increases Meg-01 maturation and platelet production. 15d-PGJ(2) also promotes platelet formation from culture-derived mouse and human megakaryocytes and accelerates platelet recovery after in vivo radiation-induced bone marrow injury. Interestingly, the platelet-enhancing effects of 15d-PGJ(2) in Meg-01 cells are independent of PPARgamma, but dependent on reactive oxygen species (ROS) accumulation; treatment with antioxidants such as glutathione ethyl ester (GSH-EE); or N-acetylcysteine (NAC) attenuate 15d-PGJ(2)-induced platelet production. Collectively, these data support the concept that megakaryocyte redox status plays an important role in platelet generation and that small electrophilic molecules may have clinical efficacy for improving platelet numbers in thrombocytopenic patients.


Subject(s)
Antineoplastic Agents/pharmacology , Blood Platelets/metabolism , Megakaryocytes/metabolism , Prostaglandin D2/analogs & derivatives , Thrombocytopenia/metabolism , Acetylcysteine/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Antioxidants/pharmacology , Bone Marrow/metabolism , Cell Line , Female , Glutathione/analogs & derivatives , Glutathione/pharmacology , Humans , Male , Mice , PPAR gamma/metabolism , Platelet Transfusion , Prostaglandin D2/pharmacology , Prostaglandin D2/therapeutic use , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/therapy , Reactive Oxygen Species/metabolism , Thrombocytopenia/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...