Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Hum Gene Ther ; 29(5): 585-601, 2018 05.
Article in English | MEDLINE | ID: mdl-29641319

ABSTRACT

B-cell maturation antigen (BCMA) expression has been proposed as a marker for the identification of malignant plasma cells in patients with multiple myeloma (MM). Nearly all MM tumor cells express BCMA, while normal tissue expression is restricted to plasma cells and a subset of mature B cells. Consistent BCMA expression was confirmed on MM biopsies (29/29 BCMA+), and it was further demonstrated that BCMA is expressed in a substantial number of lymphoma samples, as well as primary chronic lymphocytic leukemia B cells. To target BCMA using redirected autologous T cells, lentiviral vectors (LVV) encoding chimeric antigen receptors (CARs) were constructed with four unique anti-BCMA single-chain variable fragments, fused to the CD137 (4-1BB) co-stimulatory and CD3ζ signaling domains. One LVV, BB2121, was studied in detail, and BB2121 CAR-transduced T cells (bb2121) exhibited a high frequency of CAR + T cells and robust in vitro activity against MM cell lines, lymphoma cell lines, and primary chronic lymphocytic leukemia peripheral blood. Based on receptor quantification, bb2121 recognized tumor cells expressing as little as 222 BCMA molecules per cell. The in vivo pharmacology of anti-BCMA CAR T cells was studied in NSG mouse models of human MM, Burkitt lymphoma, and mantle cell lymphoma, where mice received a single intravenous administration of vehicle, control vector-transduced T cells, or anti-BCMA CAR-transduced T cells. In all models, the vehicle and control CAR T cells failed to inhibit tumor growth. In contrast, treatment with bb2121 resulted in rapid and sustained elimination of the tumors and 100% survival in all treatment models. Together, these data support the further development of anti-BCMA CAR T cells as a potential treatment for not only MM but also some lymphomas.


Subject(s)
B-Cell Maturation Antigen/antagonists & inhibitors , Hematologic Neoplasms/therapy , Multiple Myeloma/therapy , Receptors, Chimeric Antigen/immunology , Animals , B-Cell Maturation Antigen/immunology , CD3 Complex/genetics , CD3 Complex/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Immunotherapy, Adoptive , Lentivirus/genetics , Mice , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Receptors, Chimeric Antigen/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Xenograft Model Antitumor Assays
2.
Blood ; 119(26): 6307-16, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22577173

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by complement-mediated intravascular hemolysis because of the lack from erythrocyte surface of the complement regulators CD55 and CD59, with subsequent uncontrolled continuous spontaneous activation of the complement alternative pathway (CAP), and at times of the complement classic pathway. Here we investigate in an in vitro model the effect on PNH erythrocytes of a novel therapeutic strategy for membrane-targeted delivery of a CAP inhibitor. TT30 is a 65 kDa recombinant human fusion protein consisting of the iC3b/C3d-binding region of complement receptor 2 (CR2) and the inhibitory domain of the CAP regulator factor H (fH). TT30 completely inhibits in a dose-dependent manner hemolysis of PNH erythrocytes in a modified extended acidified serum assay, and also prevents C3 fragment deposition on surviving PNH erythrocytes. The efficacy of TT30 derives from its direct binding to PNH erythrocytes; if binding to the erythrocytes is disrupted, only partial inhibition of hemolysis is mediated by TT30 in solution, which is similar to that produced by the fH moiety of TT30 alone, or by intact human fH. TT30 is a membrane-targeted selective CAP inhibitor that may prevent both intravascular and C3-mediated extravascular hemolysis of PNH erythrocytes and warrants consideration for the treatment of PNH patients.


Subject(s)
Complement Factor H/chemistry , Erythrocytes/drug effects , Hemoglobinuria, Paroxysmal/blood , Hemolysis/drug effects , Oncogene Proteins, Fusion/pharmacology , Receptors, Complement 3d/chemistry , Recombinant Fusion Proteins/pharmacology , Case-Control Studies , Cells, Cultured , Complement C3/adverse effects , Complement C3/antagonists & inhibitors , Complement C3/pharmacology , Complement Factor H/metabolism , Complement Factor H/pharmacology , Complement System Proteins/adverse effects , Complement System Proteins/physiology , Cytoprotection/drug effects , Drug Evaluation, Preclinical , Erythrocytes/physiology , Hemoglobinuria, Paroxysmal/pathology , Humans , Oncogene Proteins, Fusion/metabolism , Protein Binding , Receptors, Complement 3d/metabolism , Receptors, Complement 3d/physiology , Recombinant Fusion Proteins/metabolism
3.
Blood ; 118(17): 4705-13, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21860027

ABSTRACT

To selectively modulate human complement alternative pathway (CAP) activity implicated in a wide range of acute and chronic inflammatory conditions and to provide local cell surface and tissue-based inhibition of complement-induced damage, we developed TT30, a novel therapeutic fusion protein linking the human complement receptor type 2 (CR2/CD21) C3 fragment (C3frag = iC3b, C3dg, C3d)-binding domain with the CAP inhibitory domain of human factor H (fH). TT30 efficiently blocks ex vivo CAP-dependent C3frag accumulation on activated surfaces, membrane attack complex (MAC) formation and hemolysis of RBCs in a CR2-dependent manner, and with a ∼ 150-fold potency gain over fH, without interference of C3 activation or MAC formation through the classic and lectin pathways. TT30 protects RBCs from hemolysis and remains bound and detectable for at least 24 hours. TT30 selectively inhibits CAP in cynomolgus monkeys and is bioavailable after subcutaneous injection. Using a unique combination of targeting and effector domains, TT30 controls cell surface CAP activation and has substantial potential utility for the treatment of human CAP-mediated diseases.


Subject(s)
Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3d/metabolism , Complement Factor H/therapeutic use , Complement Pathway, Alternative/immunology , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Immune System Diseases/drug therapy , Immune System Diseases/immunology , Receptors, Complement 3d/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Animals , Complement C3-C5 Convertases/metabolism , Complement Factor H/administration & dosage , Drug Design , Drug Evaluation, Preclinical , Female , Humans , Immune System Diseases/metabolism , Macaca fascicularis , Male , Models, Immunological , Molecular Targeted Therapy/methods , Rabbits , Receptors, Complement 3d/administration & dosage , Recombinant Fusion Proteins/administration & dosage
6.
Circulation ; 116(23): 2678-86, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-18025536

ABSTRACT

BACKGROUND: ARC1779 is a therapeutic aptamer antagonist of the A1 domain of von Willebrand Factor (vWF), the ligand for receptor glycoprotein 1b on platelets. ARC1779 is being developed as a novel antithrombotic agent for use in patients with acute coronary syndromes. METHODS AND RESULTS: This was a randomized, double-blind, placebo-controlled study in 47 healthy volunteers of doses of ARC1779 from 0.05 to 1.0 mg/kg. Pharmacodynamic effects were measured by an ELISA for free vWF A1 binding sites and by a platelet function analyzer. In terms of pharmacokinetics, the concentration-time profile of ARC1779 appeared monophasic. The observed concentration and area under the curve were dose proportional. The mean apparent elimination half-life was approximately 2 hours, and mean residence time was approximately 3 hours. The mean apparent volumes of distribution (at steady state and during terminal phase) were approximately one half the blood volume, suggesting that ARC1779 distribution is in the central compartment. The mean clearance ranged from approximately 10% to approximately 21% of the glomerular filtration rate, suggesting that renal filtration may not be a major mechanism of clearance of ARC1779. Inhibition of vWF A1 binding activity was achieved with an EC(90) value of 2.0 mug/mL (151 nmol/L) and of platelet function with an EC(90) value of 2.6 mug/mL (196 nmol/L). ARC1779 was generally well tolerated, and no bleeding was observed. Adverse events tended to be minor and not dose related. CONCLUSIONS: This is the first-in-human evaluation of a novel aptamer antagonist of vWF. ARC1779 produced dose- and concentration-dependent inhibition of vWF activity and platelet function with duration of effect suitable for the intended clinical use in acute coronary syndromes.


Subject(s)
Acute Coronary Syndrome/drug therapy , Aptamers, Nucleotide/pharmacokinetics , Fibrinolytic Agents/pharmacokinetics , von Willebrand Factor/antagonists & inhibitors , Adolescent , Adult , Aged , Aptamers, Nucleotide/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Female , Fibrinolytic Agents/adverse effects , Humans , Male , Middle Aged , Platelet Function Tests , Platelet Glycoprotein GPIb-IX Complex/agonists , Protein Structure, Tertiary , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...