Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Front Immunol ; 13: 960329, 2022.
Article in English | MEDLINE | ID: mdl-36420263

ABSTRACT

Graft-versus-host disease (GvHD) is a major complication after allogeneic hematopoietic cell transplantation (HCT). Current strategies to prevent GvHD with immunosuppressive drugs carry significant morbidity and may affect the graft-versus-tumor (GVT) effect. Inflammatory bowel disease (IBD) is an intestinal inflammatory condition that affects more than 2 million people in the United States. Current strategies to prevent colitis with immunosuppressive drugs carry significant morbidity. Recently, Repulsive Guidance Molecule b (RGMb) has been identified as part of a signaling hub with neogenin and BMP receptors in mice and humans. In addition, RGMb binds BMP-2/4 in mice and humans as well as PD-L2 in mice. RGMb is expressed in the gut epithelium and by antigen presenting cells, and we found significantly increased expression in mouse small intestine after total body irradiation HCT conditioning. We hypothesized that RGMb may play a role in GvHD and IBD pathogenesis by contributing to mucosal inflammation. Using major-mismatched HCT mouse models, treatment with an anti-RGMb monoclonal antibody (mAb) that blocks the interaction with BMP-2/4 and neogenin prevented GvHD and improved survival compared to isotype control (75% versus 30% survival at 60 days after transplantation). The GVT effect was retained in tumor models. Using an inflammatory bowel disease dextran sulfate sodium model, treatment with anti-RGMb blocking monoclonal antibody but not isotype control prevented colitis and improved survival compared to control (73% versus 33% at 21 days after treatment) restoring gut homeostasis. Anti-RGMb mAb (9D1) treatment decreased IFN-γ and significantly increased IL-5 and IL-10 in the gut of the treated mice compared to the isotype control treated mice.


Subject(s)
Colitis , Graft vs Host Disease , Inflammatory Bowel Diseases , Humans , Mice , Animals , Inflammation , Inflammatory Bowel Diseases/therapy , Colitis/chemically induced , Immunosuppressive Agents , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cell Adhesion Molecules, Neuronal
2.
Toxicol Sci ; 189(1): 32-50, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35583313

ABSTRACT

Bispecific T-cell engager (BiTE) molecules have great potential to treat cancer. Nevertheless, dependent on the targeted tumor antigen, the mechanism of action that drives efficacy may also contribute to on-target/off-tumor toxicities. In this study, we characterize an anti-CD70 half-life extended BiTE molecule (termed N6P) which targets CD70, a TNF family protein detected in several cancers. First, the therapeutic potential of N6P was demonstrated using in vitro cytotoxicity assays and an orthotopic xenograft mouse study resulting in potent killing of CD70+ cancer cells. Next, in vitro characterization demonstrated specificity for CD70 and equipotent activity against human and cynomolgus monkey CD70+ cells. To understand the potential for on-target toxicity, a tissue expression analysis was performed and indicated CD70 is primarily restricted to lymphocytes in normal healthy tissues and cells. Therefore, no on-target toxicity was expected to be associated with N6P. However, in a repeat-dose toxicology study using cynomolgus monkeys, adverse N6P-mediated inflammation was identified in multiple tissues frequently involving the mesothelium and epithelium. Follow-up immunohistochemistry analysis revealed CD70 expression in mesothelial and epithelial cells in some tissues with N6P-mediated injury, but not in control tissues or those without injury. Collectively, the data indicate that for some target antigens such as CD70, BiTE molecules may exhibit activity in tissues with very low antigen expression or the antigen may be upregulated under stress enabling molecule activity. This work illustrates how a thorough understanding of expression and upregulation is needed to fully address putative liabilities associated with on-target/off-tumor activity of CD3 bispecific molecules.


Subject(s)
Antibodies, Bispecific , Neoplasms , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , CD3 Complex/metabolism , Half-Life , Humans , Macaca fascicularis , Mice , Neoplasms/metabolism , T-Lymphocytes
3.
Blood Adv ; 3(21): 3419-3431, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31714958

ABSTRACT

Acute graft-versus-host disease (GVHD) is a leading cause of mortality after allogeneic hematopoietic cell transplantation (HCT) mediated by dysregulated T-cell immune reconstitution. Given the role of the T-cell immunoglobulin and mucin 1 (TIM-1) surface protein in many immune processes, including organ transplantation tolerance, we asked if TIM-1 might drive post-transplant inflammation and acute GVHD. TIM-1 binds to phosphatidylserine (PtdSer), and agonism of TIM1 on immune cells is proinflammatory. HCT conditioning results in a significant supply of PtdSer from apoptosis and cellular debris. Using murine models, treatment with an antagonistic anti-TIM-1 monoclonal antibody (mAb) protects against acute GVHD while maintaining graft-versus-tumor effects. In contrast, the addition of exogenous free PtdSer worsened GVHD in a TIM-1-dependent manner. Importantly, TIM-1 blockade did not alter the expansion of donor T cells in vitro or in vivo. Instead, TIM-1 blockade reduces proinflammatory cytokines and promotes anti-inflammatory factors like carbonic anhydrase 1 and serum amyloid A1 in the gut tissue. This is mediated by TIM-1 on donor cells, as HCT of wild-type (WT) bone marrow (BM) and conventional T (Tcon) cells into TIM-1-/- knockout (KO) recipient mice showed little survival advantage compared with WT recipients, whereas WT recipients of TIM-1-/- KO Tcon cells or TIM1-/- KO BM had improved survival, in part due to the expression of TIM-1 on donor invariant natural killer T cells, which drives inflammation. Finally, in a humanized mouse xenograft GVHD model, treatment with anti-human TIM-1 antagonist mAb reduced GVHD disease burden and mortality. This supports TIM-1 as important for GVHD pathogenesis and as a target for the prevention of GVHD.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hepatitis A Virus Cellular Receptor 1/antagonists & inhibitors , Animals , Antibodies, Blocking/therapeutic use , Biomarkers , Disease Models, Animal , Gene Expression , Graft vs Host Disease/diagnosis , Graft vs Host Disease/mortality , Hematopoietic Stem Cell Transplantation/methods , Hepatitis A Virus Cellular Receptor 1/genetics , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Immune Reconstitution , Immunohistochemistry , Immunophenotyping , Inflammation Mediators/metabolism , Lymphocyte Count , Mice , Mice, Knockout , Severity of Illness Index , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
4.
JCI Insight ; 2(20)2017 10 19.
Article in English | MEDLINE | ID: mdl-29046484

ABSTRACT

Cellular therapies based on permanent genetic modification of conventional T cells have emerged as a promising strategy for cancer. However, it remains unknown if modification of T cell subsets, such as Tregs, could be useful in other settings, such as allograft transplantation. Here, we use a modular system based on a chimeric antigen receptor (CAR) that binds covalently modified mAbs to control Treg activation in vivo. Transient expression of this mAb-directed CAR (mAbCAR) in Tregs permitted Treg targeting to specific tissue sites and mitigated allograft responses, such as graft-versus-host disease. mAbCAR Tregs targeted to MHC class I proteins on allografts prolonged islet allograft survival and also prolonged the survival of secondary skin grafts specifically matched to the original islet allograft. Thus, transient genetic modification to produce mAbCAR T cells led to durable immune modulation, suggesting therapeutic targeting strategies for controlling alloreactivity in settings such as organ or tissue transplantation.


Subject(s)
Immune Tolerance/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , Animals , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Culture Techniques , Disease Models, Animal , Graft Rejection/prevention & control , Graft Survival/immunology , Histocompatibility Antigens Class I , Mice, Inbred BALB C , Mice, Inbred C57BL , Organ Transplantation , Receptors, Chimeric Antigen/genetics , STAT5 Transcription Factor , T-Lymphocytes, Regulatory/immunology , Tissue Transplantation , Transplantation Tolerance/immunology , Transplantation, Homologous
5.
Oncotarget ; 7(8): 8653-62, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26840025

ABSTRACT

The transcription factor CREB (cAMP Response Element Binding Protein) is an important determinant in the growth of Acute Myeloid Leukemia (AML) cells. CREB overexpression increases AML cell growth by driving the expression of key regulators of apoptosis and the cell cycle. Conversely, CREB knockdown inhibits proliferation and survival of AML cells but not normal hematopoietic cells. Thus, CREB represents a promising drug target for the treatment of AML, which carries a poor prognosis. In this study, we performed a high-throughput small molecule screen to identify compounds that disrupt CREB function in AML cells. We screened ~114,000 candidate compounds from Stanford University's small molecule library, and identified 5 molecules that inhibit CREB function at micromolar concentrations, but are non-toxic to normal hematopoietic cells. This study suggests that targeting CREB function using small molecules could provide alternative approaches to treat AML.


Subject(s)
Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , High-Throughput Screening Assays/methods , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Promoter Regions, Genetic/genetics , Response Elements/genetics , Small Molecule Libraries/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Luciferases/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL