Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 7(11): 1306-1319, 2017 11.
Article in English | MEDLINE | ID: mdl-28801306

ABSTRACT

Adoptive immunotherapy with T cells expressing chimeric antigen receptors (CAR) has had limited success for solid tumors in early-phase clinical studies. We reasoned that introducing into CAR T cells an inducible costimulatory (iCO) molecule consisting of a chemical inducer of dimerization (CID)-binding domain and the MyD88 and CD40 signaling domains would improve and control CAR T-cell activation. In the presence of CID, T cells expressing HER2-CARζ and a MyD88/CD40-based iCO molecule (HER2ζ.iCO T cells) had superior T-cell proliferation, cytokine production, and ability to sequentially kill targets in vitro relative to HER2ζ.iCO T cells without CID and T cells expressing HER2-CAR.CD28ζ. HER2ζ.iCO T cells with CID also significantly improved survival in vivo in two xenograft models. Repeat injections of CID were able to further increase the antitumor activity of HER2ζ.iCO T cells in vivo Thus, expressing MyD88/CD40-based iCO molecules in CAR T cells has the potential to improve the efficacy of CAR T-cell therapy approaches for solid tumors.Significance: Inducible activation of MyD88 and CD40 in CAR T cells with a small-molecule drug not only enhances their effector function, resulting in potent antitumor activity in preclinical solid tumors, but also enables their remote control post infusion. Cancer Discov; 7(11); 1306-19. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1201.


Subject(s)
CD40 Antigens/genetics , Immunotherapy, Adoptive/methods , Myeloid Differentiation Factor 88/genetics , Neoplasms/therapy , Receptors, Antigen, T-Cell/therapeutic use , Animals , CD40 Antigens/immunology , CD40 Antigens/therapeutic use , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Mice , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/therapeutic use , Neoplasms/genetics , Neoplasms/immunology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Xenograft Model Antitumor Assays
2.
Neuro Oncol ; 18(8): 1120-8, 2016 08.
Article in English | MEDLINE | ID: mdl-26917236

ABSTRACT

BACKGROUND: Glioblastoma is one of the most malignant brain tumors in adults and has a dismal prognosis. In a previous report, we reported that CD40, a TNF-R-related cell surface receptor, and its ligand CD40L were associated with glioma outcomes. Here we attempted to activate CD40 signaling in the tumor and determine if it exerted therapeutic efficacy. METHODS: CD40 expression was examined in 3 mouse glioma cell lines (GL261, NSCL61, and bRiTs-G3) and 5 human glioma cell lines (U87, U251, U373, T98, and A172). NSCL61 and bRiTs-G3, as glioma stem cells, also expressed the glioma stem cell markers MELK and CD44. In vitro, we demonstrated direct antitumor effects of an anti-CD40 agonistic monoclonal antibody (FGK45) against the cell lines. The efficacy of FGK45 was examined by local convection-enhanced delivery of the monoclonal antibody against each glioma model. RESULTS: CD40 was expressed in all mouse and human cell lines tested and was found at the cell membrane of each of the 3 mouse cell lines. FGK45 administration induced significant, direct antitumor effects in vitro. The local delivery of FGK45 significantly prolonged survival compared with controls in the NSCL61 and bRiTs-G3 models, but the effect was not significant in the GL261 model. Increases in apoptosis and CD4(+) and CD8(+) T cell infiltration were observed in the bRiTs-G3 model after FGK45 treatment. CONCLUSIONS: Local delivery of FGK45 significantly prolonged survival in glioma stem cell models. Thus, local delivery of this monoclonal antibody is promising for immunotherapy against gliomas.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , CD40 Antigens/agonists , Glioma/drug therapy , Immunotherapy/methods , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Brain/drug effects , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Antigens/therapeutic use , Cell Line, Tumor , Convection , Disease Models, Animal , Female , Glioma/immunology , Glioma/metabolism , Humans , Mice , Mice, Inbred C57BL , Survival Analysis , Vaccination/methods
3.
J Leukoc Biol ; 90(2): 389-98, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21586676

ABSTRACT

HIV-1 does not significantly activate cellular immunity, which has made it difficult to use attenuated forms of HIV-1 as a vaccine. In contrast, EBV induces robust T cell responses in most infected individuals, perhaps as this virus contains LMP1, a viral mimic of CD40, which is a key activating molecule for DCs and macrophages. Consequently, studies were conducted using LMP1 and LMP1-CD40, a related construct formed by replacing the intracellular signaling domain of LMP1 with that of CD40. Upon electroporation into DCs, LMP1 and LMP1-CD40 mRNAs were sufficient to up-regulate costimulatory molecules and proinflammatory cytokines, indicating that these molecules can function in isolation as adjuvant-like molecules. As a first step toward an improved HIV vaccine, LMP1 and LMP1-CD40 were introduced into a HIV-1 construct to produce virions encoding these proteins. Transduction of DCs and macrophages with these viruses induced morphological changes and up-regulated costimulatory molecules and cytokine production by these cells. HIV-LMP1 enhanced the antigen-presenting function of DCs, as measured in an in vitro immunization assay. Taken together, these data show that LMP1 and LMP1-CD40 are portable gene cassettes with strong adjuvant properties that can be introduced into viruses such as HIV, which by themselves, are insufficient to induce protective cellular immunity.


Subject(s)
AIDS Vaccines/immunology , Dendritic Cells/immunology , Herpesvirus 4, Human/immunology , Viral Matrix Proteins/immunology , Adjuvants, Immunologic/therapeutic use , Antigen Presentation , CD40 Antigens/therapeutic use , HIV/genetics , HIV/immunology , Herpesvirus 4, Human/chemistry , Herpesvirus 4, Human/genetics , Humans , Molecular Mimicry/immunology , Transduction, Genetic , Viral Matrix Proteins/therapeutic use
4.
Antiviral Res ; 75(1): 14-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17210189

ABSTRACT

A recombinant Eimeria protozoan protein antigen (rEA) has been shown to have antitumor and antiviral activity. The purpose of this study was to determine the effect of rEA treatment alone or in combination with an agonist cocktail consisting of granulocyte macrophage colony stimulating factor (GM-CSF), interferon gamma (IFN-gamma), interleukin 4 (IL-4), and anti CD-40 antibody, in the treatment of Banzi virus (BV) disease in BALB/c mice. Treatment with rEA resulted in a significant increase in survival, weight gain, and mean day to death in BV-infected mice and resulted in a significant decrease in brain virus titer. Treatment with rEA, in combination with a 4-agonist cocktail, improved disease parameters to a greater degree than rEA treatment alone. The effect of treatment with a reduced concentration of agonist cocktail or fewer components of the agonist cocktail, in combination with rEA, on disease outcome in BV-infected mice was also investigated. Treatment with rEA, alone or in combination with agonist cocktail, 24h after virus challenge did not improve disease. Treatment with rEA, alone or in combination with an agonist cocktail, is efficacious for the prophylaxis of BV infection in mice.


Subject(s)
Eimeria/chemistry , Flavivirus Infections/prevention & control , Flavivirus/drug effects , Protozoan Proteins/agonists , Protozoan Proteins/therapeutic use , Animals , CD40 Antigens/therapeutic use , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Flavivirus/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Interferon-gamma/therapeutic use , Interleukin-4/therapeutic use , Mice , Mice, Inbred BALB C , Protozoan Proteins/genetics , Protozoan Proteins/isolation & purification , Protozoan Proteins/pharmacology , Recombinant Proteins/agonists , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Survival Rate , Time Factors
5.
Ann Plast Surg ; 55(5): 512-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16258305

ABSTRACT

To determine the efficacy of a regimen based on CD40 costimulation blockade and donor bone marrow in the limb allograft model, C57Bl/6 mice received limb allografts from Balb/c mice and either no treatment or a combination of MR1 (anti-CD40 ligand monoclonal antibody), CD4+ and CD8+ T-cell-depleting antibodies, low-dose irradiation, and bone marrow transfusion from Balb/c donors for 1 or 2 weeks. Recipients treated for 1 week showed rejection at 38.2 +/- 5.4 (mean +/- SEM) days, while those treated for 2 weeks had allograft survival of 56.5 +/- 9.9, with a range up to 91 days. Histology demonstrated rejection which was less cell-mediated and suggestive of transplant vasculopathy. Differential rejection of skin occurred first. Thus, a combined regimen based on CD40 costimulatory blockade and donor marrow significantly prolonged allograft survival. However, tolerance was not achieved, and histology suggests chronic rejection as a possible cause of allograft loss.


Subject(s)
Bone Marrow Transplantation/methods , CD40 Antigens/therapeutic use , Extremities/radiation effects , Extremities/surgery , Graft Survival , Lymphocyte Depletion/methods , Transplantation, Homologous , Animals , Antibody Formation , CD40 Antigens/immunology , Dose-Response Relationship, Radiation , Mice , Mice, Inbred C57BL , Radiation Dosage , Random Allocation
6.
Br J Haematol ; 121(4): 592-6, 2003 May.
Article in English | MEDLINE | ID: mdl-12752100

ABSTRACT

Multiple myeloma (MM) is currently incurable, and novel therapies are needed. In this study, we examined a novel recombinant humanized monoclonal antibody against CD40 (rhuCD40 mAb) and demonstrate for the first time that rhuCD40 mAb induces antibody-dependent cell-mediated cytotoxicity (ADCC) against CD40-positive MM cells. Importantly, we show that rhuCD40 mAb induces autologous ADCC against primary patient MM cells, without triggering ADCC against normal B cells. This study, therefore, both demonstrates that rhuCD40 mAb triggers autologous ADCC against patient MM cells and provides the framework for the clinical evaluation of rhuCD40 mAb immunotherapy to improve patient outcome in MM.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , CD40 Antigens/therapeutic use , Immunotherapy/methods , Multiple Myeloma/therapy , Antibodies, Monoclonal/therapeutic use , Humans , Recombinant Proteins/therapeutic use
7.
Chin Med J (Engl) ; 114(7): 685-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11780327

ABSTRACT

OBJECTIVE: To investigate the protective effects of blocking CD40/CD40L interactions with human CD40-Ig fusion protein in a murine graft-versus-host disease model. METHODS: Human CD40 gene extracellular region was inserted into plasmid pIG1, which contains genomic human IgG1 Fc gene. A transient vector containing CD40-Fc fusion gene was transfected into COS-7 cells. The CD40-Ig fusion protein was detected through enzyme-linked immunosorbent assay (ELISA). A constitutive vector was also generated by ligating the CD40-Fc fusion gene into pcDNA3.1 and transfecting it into CHO cells. CD40-Ig was purified by protein A affinity chromatography. SDS-PAGE, Western blot and ligand binding assay were used to identify the qualities of CD40-Ig. Murine acute graft-versus-host disease (GVHD) was induced by intravenous injection of C57BL/6J (H-2b) spleen cells into sub-lethally irradiated BALB/c (H-2d) mice. Protective effects against murine graft-versus-host disease by in vivo administration of CD40-Ig were evaluated. RESULTS: Mammalian expression vectors pIG/40Ig and p3.1/40Ig were constructed as described above. Chimeric proteins were expressed in COS-7 and CHO cell culture supernatant and confirmed by ELISA and Western blot. SDS-PAGE showed that fusion proteins had a disulfide-bonded dimeric structure and existed as homodimer. Purified CD40-Ig could bind to CD40L. In vivo administration of CD40-Ig could prevent the development of GVHD and significantly prolong the mean survival time of mice with graft-versus-host disease. CONCLUSIONS: These results demonstrate that CD40/CD40L interactions play an important role in the pathogenesis of graft-versus-host disease and suggest clinical potential for CD40-Ig in the prevention and treatment of human graft-versus-host disease.


Subject(s)
CD40 Antigens/therapeutic use , Graft vs Host Disease/prevention & control , Immunoglobulin G/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Acute Disease , Animals , CD40 Ligand/metabolism , Female , Graft vs Host Disease/immunology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology
8.
Cancer Immunol Immunother ; 49(8): 410-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11043847

ABSTRACT

Engagement of CD40 on the surface of antigen-presenting cells (APC) has been shown to substitute for T cell help in activating APC to stimulate cytotoxic T lymphocytes (CTL). We explored whether this powerful non-specific signal could enhance the CTL response to a self epitope from a tumor-associated antigen. We immunized mice with a lipopeptide covering the H-2Kd-restricted epitope, amino acids 232-240 of murine wild-type p53, followed by treatment with an activating anti-CD40 monoclonal antibody. Anti-CD40 antibody, given subcutaneously or intravenously, significantly enhanced effector activity against targets pulsed with non-lipidated 232-240 nonamer epitope peptide, as assessed both by a CTL lysis assay and an enzyme-linked immunospot (ELISPOT) assay for interferon-gamma-secreting cells. However, despite this enhancement, we could not detect activity against targets expressing p53 endogenously by either assay. This most likely reflects the low avidity of the effectors as determined by a titration of peptide on the target cells. The implications of this work for cancer immunotherapy based on specific responses directed against tumor-associated antigens are discussed.


Subject(s)
CD40 Antigens/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/therapy , Amino Acids/chemistry , Animals , Antibody Affinity , CD40 Antigens/immunology , CD40 Antigens/therapeutic use , Cancer Vaccines , Dose-Response Relationship, Drug , Epitopes , Female , Flow Cytometry , Genes, MHC Class I , Immunoenzyme Techniques , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Mammary Neoplasms, Animal/therapy , Mice , Mice, Inbred BALB C , Peptides/pharmacology , Peptides/therapeutic use , Spleen/cytology , Tumor Cells, Cultured
10.
Leuk Res ; 23(6): 549-57, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10374848

ABSTRACT

Induction of an optimal immune response will likely be a prerequisite for successful immunotherapy of human leukemias and other malignancies. Dendritic cells are highly effective at inducing an immune response to antigens to which the host is unresponsive, while transgenic expression of the costimulator molecule CD40 ligand (gp39/CD154) and the T cell growth factor interleukin 2 (IL2) are also able to augment immune responsiveness. We therefore investigated whether a combination of these two distinctive approaches to immunostimulation could safely increase the anti-tumor immune response compared to each stimulus alone. We injected BALB/CBYJ mice with syngeneic dendritic cells (DC) exposed to A20 lymphoblastic leukemia cell-derived peptides and proteins which had been acid-eluted from the cell surface. In additional mice, the pulsed DC were mixed with genetically modified syngeneic fibroblasts that were expressing CD40 ligand or secreting interleukin 2 (IL2). Three days after their third, weekly, vaccination, they were challenged with parental A20 cells. Tumor growth was suppressed by responses to pulsed DC alone (P < 0.02). This suppression was further enhanced when pulsed DC were coinjected with fibroblasts expressing CD40 ligand and IL2 (P < 0.0005 compared to DC alone) even though CD40 ligand and IL2-expressing fibroblasts alone offered no significant protection in this model. Mice receiving the full complement of immunostimulants either failed to develop visible tumors or developed small tumors which quickly necrosed and regressed, allowing the mice to become long term tumor-free survivors. Antibody mediated depletion of either CD4+ or CD8+ T-cell subset significantly reduced the level of protection afforded by the vaccination. However, it became evident that this intensive stimulation of the immune system lead not only to tumor eradication but also to destruction of cells bearing normal self antigens. Hence, 60 days after challenge with A20 cells all mice in the DC/IL2/CD40 ligand group developed a severe, systemic autoimmune disorder that resembled graft versus host disease and manifest itself by significant peripheral blood cytotoxicity against autologous fibroblasts, blood dyscrasias, gross hepatosplenomegaly, cachexia and fur loss. This phenomenon depended on CD8+ cytotoxic T lymphocytes. Our results therefore suggest that the most effective strategies of immunotherapy against leukemia may also exceed the threshold of anergic cells, leading to a loss of self tolerance to normal self-antigens and the induction of an CD8+ anti-self effector response.


Subject(s)
Autoimmune Diseases/immunology , Dendritic Cells/immunology , Immunization/adverse effects , Animals , Antigens, Neoplasm/therapeutic use , CD40 Antigens/therapeutic use , Cells, Cultured , Disease-Free Survival , Humans , Immunotherapy , Mice , Mice, Inbred BALB C
11.
Cancer Immunol Immunother ; 45(3-4): 174-9, 1997.
Article in English | MEDLINE | ID: mdl-9435867

ABSTRACT

T cells play a key role in the control of abnormal B cell proliferation. Factors that play a role in inadequate T cell responses include absence of expression of costimulatory and adhesion molecules by the malignant B cells and lack of cytotoxic T cells specific for tumor-associated antigens. A number of approaches have been used to enhance T cell response against malignant B cells. Agents such as soluble CD40 ligand can enhance expression of costimulatory molecules by the malignant B cells and improve their ability to activate T cells. Anti-CD3-based bispecific antibodies can retarget T cells toward the tumor cells irrespective of T cell specificity. We used the V 38C13 murine lymphoma model to assess whether the combination of soluble CD40 ligand and anti-CD3-based bispecific antibody can enhance T cell activation induced by malignant B cells more effectively than either approach alone. Expression of CD80, CD86, and ICAM-1 on lymphoma cells was up-regulated by soluble CD40 ligand. Syngeneic T cells were activated more extensively by lymphoma cells when the lymphoma cells were pre-treated with soluble CD40 ligand. Bispecific-antibody induced T cell activation was more extensive when lymphoma cells pretreated with soluble CD40 ligand were present. The combination of soluble CD40 ligand plus bispecific antibody enhanced the median survival of mice compared to mice treated with bispecific antibody alone. We conclude that pretreatment of tumor cells with agents capable of inducing costimulatory molecule expression, such as soluble CD40 ligand can enhance the ability of malignant B cells to activate T cells. This effect is enhanced by the addition of bispecific antibody. The combination of enhanced expression of costimulatory molecules and retargeting of T cells by bispecific antibody may allow for a more effective T-cell-based immunotherapy.


Subject(s)
Antibodies, Bispecific/immunology , CD3 Complex/immunology , CD40 Antigens/immunology , Lymphocyte Activation/immunology , Lymphoma, B-Cell/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , CD40 Antigens/therapeutic use , Female , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred C3H
12.
Kidney Blood Press Res ; 19(3-4): 201-4, 1996.
Article in English | MEDLINE | ID: mdl-8887261

ABSTRACT

The objective of immunosuppressive therapy in nephrology is to prevent autoimmune diseases and to suppress kidney allograft rejections while sparing other effects. Increased clarification of the underlying immune mechanism has made specific immunodulation possible using chimeric proteins in which the variable domains of an immunoglobulin are replaced by extracellular domains of cell surface molecules or cytokines. The immunosuppressive effects of fusion proteins such as CTLA-4 IgG, CD40 IgG, interleukin (IL)-10 IgG, IL-2 IgG or tumor necrosis factor (TNF)-receptor IgG have been proven in various animal models. Moreover, the application of TNF-receptor IgG successfully limited the OKT3-induced cytokine release syndrome in kidney graft recipients. It seems likely that recombinant proteins with increasingly effective suppression of specific elements of the immune response will become an essential element in clinical protocols.


Subject(s)
Graft Rejection/prevention & control , Immunoconjugates , Kidney Transplantation/immunology , Recombinant Fusion Proteins/therapeutic use , Abatacept , Animals , Antigens, CD , Antigens, Differentiation/therapeutic use , CD40 Antigens/therapeutic use , CTLA-4 Antigen , Cytokines/therapeutic use , Glomerulonephritis, Membranous/prevention & control , Graft Rejection/immunology , Humans , Immunoglobulin G/therapeutic use , Immunosuppressive Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...