Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Mol Cancer Ther ; 17(2): 554-564, 2018 02.
Article in English | MEDLINE | ID: mdl-29142066

ABSTRACT

Treatment choices for acute myelogenous leukemia (AML) patients resistant to conventional chemotherapies are limited and novel therapeutic agents are needed. IL3 receptor alpha (IL3Rα, or CD123) is expressed on the majority of AML blasts, and there is evidence that its expression is increased on leukemic relative to normal hematopoietic stem cells, which makes it an attractive target for antibody-based therapy. Here, we report the generation and preclinical characterization of SGN-CD123A, an antibody-drug conjugate using the pyrrolobenzodiazepine dimer (PBD) linker and a humanized CD123 antibody with engineered cysteines for site-specific conjugation. Mechanistically, SGN-CD123A induces activation of DNA damage response pathways, cell-cycle changes, and apoptosis in AML cells. In vitro, SGN-CD123A-mediated potent cytotoxicity of 11/12 CD123+ AML cell lines and 20/23 primary samples from AML patients, including those with unfavorable cytogenetic profiles or FLT3 mutations. In vivo, SGN-CD123A treatment led to AML eradication in a disseminated disease model, remission in a subcutaneous xenograft model, and significant growth delay in a multidrug resistance xenograft model. Moreover, SGN-CD123A also resulted in durable complete remission of a patient-derived xenograft AML model. When combined with a FLT3 inhibitor quizartinib, SGN-CD123A enhanced the activity of quizartinib against two FLT3-mutated xenograft models. Overall, these data demonstrate that SGN-CD123A is a potent antileukemic agent, supporting an ongoing trial to evaluate its safety and efficacy in AML patients (NCT02848248). Mol Cancer Ther; 17(2); 554-64. ©2017 AACR.


Subject(s)
Immunoconjugates/pharmacology , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/drug therapy , Animals , Antibodies, Monoclonal/immunology , CHO Cells , Cell Line, Tumor , Cricetulus , Humans , Immunoconjugates/immunology , Leukemia, Myeloid, Acute/immunology , Mice , Mice, SCID , THP-1 Cells , Xenograft Model Antitumor Assays
2.
MAbs ; 2(4): 440-8, 2010.
Article in English | MEDLINE | ID: mdl-20495353

ABSTRACT

Despite therapeutic advances, the poor prognoses for acute myeloid leukemia (AML) and intermediate and high-risk myelodysplastic syndromes (MDS) point to the need for better treatment options. AML and MDS cells express the myeloid marker CD33, making it amenable to CD33-targeted therapy. Lintuzumab (SGN-33), a humanized monoclonal anti-CD33 antibody undergoing clinical evaluation, induced meaningful responses in a Phase 1 clinical trial and demonstrated anti-leukemic activity in preclinical models. Recently, it was reported that 5-azacytidine (Vidaza™) prolonged the overall survival of a group of high risk MDS and AML patients. To determine whether the combination of lintuzumab and 5-azacytidine would be beneficial, a mouse xenograft model of disseminated AML was used to evaluate the combination.  There was a significant reduction in tumor burden and an increase in overall survival in mice treated with lintuzumab and 5-azacytidine. The effects were greater than that obtained with either agent alone. As the in vivo anti-leukemic activity of lintuzumab was dependent upon the presence of mouse effector cells including macrophages and neutrophils, in vitro effector function assays were used to assess the impact of 5-azacytidine on lintuzumab activity. The results show that 5-azacytidine significantly enhanced the ability of lintuzumab to promote tumor cell killing through antibody-dependent cellular cytotoxicity (ADCC) and phagocytic (ADCP) activities. These results suggest that lintuzumab and 5-azacytidine act in concert to promote tumor cell killing. Additionally, these findings provide the rationale to evaluate this combination in the clinic.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Azacitidine/administration & dosage , Drug Synergism , Leukemia, Myeloid, Acute/therapy , Macrophages/drug effects , Neutrophils/drug effects , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Drug Evaluation, Preclinical , Drug Therapy, Combination , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/immunology , Macrophages/immunology , Mice , Mice, SCID , Neutrophils/immunology , Phagocytosis/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
MAbs ; 1(5): 481-90, 2009.
Article in English | MEDLINE | ID: mdl-20065652

ABSTRACT

Despite therapeutic advances, the long-term survival rates for acute myeloid leukemia (AML) are estimated to be 10% or less, pointing to the need for better treatment options. AML cells express the myeloid marker CD33, making it amenable to CD33-targeted therapy. Thus, the in vitro and in vivo anti-tumor activities of lintuzumab (SGN-33), a humanized monoclonal anti-CD33 antibody undergoing clinical evaluation, were investigated. In vitro assays were used to assess the ability of lintuzumab to mediate effector functions and to decrease the production of growth factors from AML cells. SCID mice models of disseminated AML with the multi-drug resistance (MDR)-negative HL60 and the MDR(+), HEL9217 and TF1-alpha, cell lines were developed and applied to examine the in vivo antitumor activity. In vitro, lintuzumab significantly reduced the production of TNFalpha-induced pro-inflammatory cytokines and chemokines by AML cells. Lintuzumab promoted tumor cell killing through antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) activities against MDR(-) and MDR(+) AML cell lines and primary AML patient samples. At doses from 3 to 30 mg/kg, lintuzumab significantly enhanced survival and reduced tumor burden in vivo, regardless of MDR status. Survival of the mice was dependent upon the activity of resident macrophages and neutrophils. The results suggest that lintuzumab may exert its therapeutic effects by modulating the cytokine milieu in the tumor microenvironment and through effector mediated cell killing. Given that lintuzumab induced meaningful responses in a phase 1 clinical trial, the preclinical antitumor activities defined in this study may underlie its observed therapeutic efficacy in AML patients.


Subject(s)
Antibodies, Monoclonal , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Antineoplastic Agents , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Mice, SCID , Phagocytosis , Sialic Acid Binding Ig-like Lectin 3 , Treatment Outcome , U937 Cells
4.
Bone ; 35(4): 828-35, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15454089

ABSTRACT

A null mutation in the SOST gene is associated with sclerosteosis, an inherited disorder characterized by a high bone mass phenotype. The protein product of the SOST gene, sclerostin, is a bone morphogenetic protein (BMP) antagonist that decreases osteoblast activity and reduces the differentiation of osteoprogenitors. We sought to delineate the mechanism by which sclerostin modulated osteoblastic function by examining the effects of the protein on differentiating cultures of human mesenchymal stem cells (hMSC). Sclerostin significantly decreased alkaline phosphatase (ALP) activity and the proliferation of hMSC cells. In addition, hMSC cells treated with sclerostin displayed a marked increase in caspase activity. Elevated levels of fragmented histone-associated DNA in these cells were detected by ELISA and by TUNEL staining. Other BMP antagonists including noggin, Chordin, Gremlin, and Twisted gastrulation did not affect caspase activity. The sclerostin-mediated increase in caspase activity was blocked by caspase-1 and caspase-3 inhibitors. Sclerostin-induced changes in ALP activity and the survival of hMSC cells were partially restored by BMP-6, suggesting the involvement of additional growth factors. These findings show that sclerostin selectively controls the apoptosis of bone cells. The ability of sclerostin to interact with important growth factors such as BMPs likely serves as the basis by which it modulates the survival of osteoblasts. By making these growth factors unavailable for cell function, sclerostin promotes the apoptosis of bone cells, providing a novel level of control in the regulation of bone formation.


Subject(s)
Apoptosis , Bone Morphogenetic Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Adaptor Proteins, Signal Transducing , Alkaline Phosphatase/metabolism , Apoptosis/drug effects , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/genetics , Caspase Inhibitors , Caspases/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Genetic Markers/genetics , Humans , Osteoblasts/drug effects , Osteogenesis/drug effects , Osteogenesis/genetics
5.
Bone ; 35(2): 448-54, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15268896

ABSTRACT

SOST, a novel bone morphogenetic protein (BMP) antagonist and negative regulator of bone formation, is expressed in osteogenic cells. Null mutations in the SOST gene are associated with the sclerosteosis phenotype typified by high bone mass. We sought to delineate the pathways involved in the regulation of SOST expression in human osteoblastic cells. We evaluated the effects of bone growth factors and hormones on the RNA levels of SOST and the BMP antagonists, noggin and gremlin. Parathyroid hormone (PTH), transforming growth factor-beta1 (TGF-beta1), fibroblast growth factors 1 and 2 (FGF1, FGF2), and insulin-like growth factor-1 (IGF-1) had negligible effects on SOST expression in human osteoblasts. In comparison, BMPs-2, 4, and 6 induced the message levels of SOST in a time- and dose-dependent manner. The levels of noggin and, to a lesser extent, gremlin were also increased by BMPs. BMP's stimulatory effects on SOST were further enhanced by retinoic acid or 1,25-dihydroxyvitamin D3. In contrast, dexamethasone (DEX) blocked the effects of the BMPs on SOST and gremlin, but not on noggin. Retinoic acid and 1,25-dihydroxyvitamin D3 did not affect the BMP-enhanced expression of gremlin or noggin. The steroids did not affect the endogenous levels of the BMP antagonists. These findings show that the levels of SOST are modulated by BMPs and the interactions of the BMPs with steroid hormones in human osteoblasts. These effects differed markedly from that of noggin or gremlin, suggesting that there is an exquisite regulation of the expressions of BMP antagonists in cells of the osteoblast lineage.


Subject(s)
Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/physiology , Calcitriol/pharmacology , Dexamethasone/pharmacology , Gene Expression Regulation/physiology , Genetic Markers/genetics , Osteoblasts/metabolism , Adaptor Proteins, Signal Transducing , Base Sequence , DNA Primers , Gene Expression Regulation/drug effects , Humans , Reverse Transcriptase Polymerase Chain Reaction
6.
EMBO J ; 22(23): 6267-76, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14633986

ABSTRACT

There is an unmet medical need for anabolic treatments to restore lost bone. Human genetic bone disorders provide insight into bone regulatory processes. Sclerosteosis is a disease typified by high bone mass due to the loss of SOST expression. Sclerostin, the SOST gene protein product, competed with the type I and type II bone morphogenetic protein (BMP) receptors for binding to BMPs, decreased BMP signaling and suppressed mineralization of osteoblastic cells. SOST expression was detected in cultured osteoblasts and in mineralizing areas of the skeleton, but not in osteoclasts. Strong expression in osteocytes suggested that sclerostin expressed by these central regulatory cells mediates bone homeostasis. Transgenic mice overexpressing SOST exhibited low bone mass and decreased bone strength as the result of a significant reduction in osteoblast activity and subsequently, bone formation. Modulation of this osteocyte-derived negative signal is therapeutically relevant for disorders associated with bone loss.


Subject(s)
Bone Development/physiology , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/physiology , Genetic Markers/physiology , Osteocytes/physiology , Adaptor Proteins, Signal Transducing , Alkaline Phosphatase/metabolism , Animals , Base Sequence , Bone Density , Bone Diseases, Metabolic/genetics , Bone Morphogenetic Protein 6 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/pharmacology , Cell Line , Cloning, Molecular , DNA Primers , Genetic Markers/genetics , Glycoproteins , Humans , Intercellular Signaling Peptides and Proteins , Kinetics , Mesoderm/drug effects , Mesoderm/physiology , Mice , Mice, Inbred C3H , Mice, Transgenic , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
7.
Cancer Res ; 62(5): 1439-42, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11888917

ABSTRACT

We have compared the antitumor activities of SP500263, a novel next-generation selective estrogen receptor modulator (SERM), tamoxifen, and raloxifene side-by-side in in vitro and in vivo MCF-7 breast cancer models. In vitro, SP500263 acted as an antiestrogen and potently inhibited estrogen-dependent MCF-7 proliferation with IC(50) values in the nanomolar range. SP500263 also strongly inhibited MCF-7 proliferation in the absence of estrogen at all of the concentrations tested. To investigate the antitumor activity of SP500263 in animals, athymic nude mice were implanted with MCF-7 tumor in the presence of a tumor growth-supporting sustained release estrogen pellet. Treatment was initiated after tumors were established. SP500263, administered for 28 days through daily i.p. dosing, effectively reduced estrogen-stimulated tumor growth at 3 and 30 mg/kg. SP500263 was as efficacious as tamoxifen and superior to raloxifene at the corresponding doses. Maximum efficacy was reached with the 30 mg/kg dose. The observed effects were highly significant. SP500263 represents a member of a novel series of SERMs that is structurally unrelated to SERMs currently on the market or in clinical development. The experiments described herein demonstrate that SP500263 is efficacious in the MCF-7 proliferation assay and in a murine model of breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Coumarins/pharmacology , Estrogen Receptor Modulators/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Piperidines/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Animals , Breast Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous , Tumor Cells, Cultured
8.
Mol Pharmacol ; 61(3): 562-8, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11854436

ABSTRACT

We determined the differential response of a novel SERM, SP500263, on estrogen receptor (ER) alpha and the more recently cloned ER-beta. Because of the high homology of amino acid residues in the ligand-binding domain of ER-alpha and ER-beta, we were not surprised to find that SP500263 binds to both ERs equally well. In contrast, SP500263 acts as a strong estrogen agonist in a strictly ER-alpha-specific manner in U2OS osteosarcoma cell lines blocking the production of interleukin (IL) 6 and granulocyte macrophage colony-stimulating factor. SP500263 also blocked IL-6 production in primary bone cells. The mechanism of this inhibition is different from the classic estrogen stimulation involving an estrogen response element (ERE). SP500263 does not activate gene expression through an ERE. In contrast to the results observed in U2OS cells, SP500263 acts as a strong estrogen antagonist in an MCF-7 breast cancer proliferation assay. Therefore, SP500263 is a member of a series of next-generation SERMs with functional selectivity toward ER-alpha and a mixed agonist/antagonist profile in a bone cell assay versus a breast cancer assay. The panel of assays described herein allow for the development of receptor-specific ligands that may be further developed into novel pharmaceuticals with an improved profile for the treatments of osteoporosis and breast cancer.


Subject(s)
Coumarins/pharmacology , Estrogen Antagonists/pharmacology , Piperidines/pharmacology , Receptors, Estrogen/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Estrogen Receptor alpha , Estrogen Receptor beta , Gene Expression/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interleukin-6/genetics , Receptors, Estrogen/drug effects , Response Elements/drug effects , Response Elements/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL