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.
Cancer Immunol Res ; 6(10): 1199-1211, 2018 10.
Article in English | MEDLINE | ID: mdl-30154083

ABSTRACT

GITR is a costimulatory receptor currently undergoing phase I clinical trials. Efficacy of anti-GITR therapy in syngeneic mouse models requires regulatory T-cell depletion and CD8+ T-cell costimulation. It is increasingly appreciated that immune cell proliferation and function are dependent on cellular metabolism. Enhancement of diverse metabolic pathways leads to different immune cell fates. Little is known about the metabolic effects of GITR agonism; thus, we investigated whether costimulation via GITR altered CD8+ T-cell metabolism. We found activated, GITR-treated CD8+ T cells upregulated nutrient uptake, lipid stores, glycolysis, and oxygen consumption rate (OCR) in vitro Using MEK, PI3Kδ, and metabolic inhibitors, we show increased metabolism is required, but not sufficient, for GITR antibody (DTA-1)-induced cellular proliferation and IFNγ production. In an in vitro model of PD-L1-induced CD8+ T-cell suppression, GITR agonism alone rescued cellular metabolism and proliferation, but not IFNγ production; however, DTA-1 in combination with anti-PD-1 treatment increased IFNγ production. In the MC38 mouse tumor model, GITR agonism significantly increased OCR and IFNγ and granzyme gene expression in both tumor and draining lymph node (DLN) CD8+ T cells ex vivo, as well as basal glycolysis in DLN and spare glycolytic capacity in tumor CD8+ T cells. DLN in GITR-treated mice showed significant upregulation of proliferative gene expression compared with controls. These data show that GITR agonism increases metabolism to support CD8+ T-cell proliferation and effector function in vivo, and that understanding the mechanism of action of agonistic GITR antibodies is crucial to devising effective combination therapies. Cancer Immunol Res; 6(10); 1199-211. ©2018 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/immunology , Cytokines/immunology , Glucocorticoid-Induced TNFR-Related Protein/agonists , Glucocorticoid-Induced TNFR-Related Protein/immunology , Animals , Antibodies/pharmacology , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Mice, Inbred C57BL
2.
J Pharmacol Exp Ther ; 356(3): 574-86, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26669426

ABSTRACT

Administration of biologics to enhance T-cell function is part of a rapidly growing field of cancer immunotherapy demonstrated by the unprecedented clinical success of several immunoregulatory receptor targeting antibodies. While these biologic agents confer significant anti-tumor activity through targeted immune response modulation, they can also elicit broad immune responses potentially including the production of anti-drug antibodies (ADAs). DTA-1, an agonist monoclonal antibody against GITR, is a highly effective anti-tumor treatment in preclinical models. We demonstrate that repeated dosing with murinized DTA-1 (mDTA-1) generates ADAs with corresponding reductions in drug exposure and engagement of GITR on circulating CD3(+) CD4(+) T cells, due to rapid hepatic drug uptake and catabolism. Mice implanted with tumors after induction of preexisting mDTA-1 ADA show no anti-tumor efficacy when given 3 mg/kg mDTA-1, an efficacious dose in naive mice. Nonetheless, increasing mDTA-1 treatment to 30 mg/kg in ADA-positive mice restores mDTA-1 exposure and GITR engagement on circulating CD3(+) CD4(+) T cells, thereby partially restoring anti-tumor efficacy. Formation of anti-mDTA-1 antibodies and changes in drug exposure and disposition does not occur in GITR(-/-) mice, consistent with a role for GITR agonism in humoral immunity. Finally, the administration of muDX400, a murinized monoclonal antibody against the checkpoint inhibitor PD-1, dosed alone or combined with mDTA-1 did not result in reduced muDX400 exposure, nor did it change the nature of the anti-mDTA-1 response. This indicates that anti-GITR immunogenicity may not necessarily impact the pharmacology of coadministered monoclonal antibodies, supporting combination immunomodulatory strategies.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Drug Delivery Systems/methods , Glucocorticoid-Induced TNFR-Related Protein/agonists , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tissue Distribution/drug effects , Tissue Distribution/physiology , Xenograft Model Antitumor Assays/methods
3.
Mol Ther ; 17(3): 508-15, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19127252

ABSTRACT

Intravascular delivery (1.5 x 10(9) particles and higher) of recombinant adenovirus (rAd) induces myeloid cell mediated, self-limiting hemodynamic responses in normal mice. However, we observed anaphylactoid-type reactions and exacerbated hemodynamic events following rAd injection in mice bearing malignant 4T1 mammary carcinoma. Because 4T1 tumors induce significant CD11b(+)Gr-1(+) myeloid cell expansion and activation, we set to determine whether this causes rAd-induced exaggerated responses. When treated with a single intravenous dose (1 x 10(10) particles) of rAd, mice implanted with 4T1 carcinoma succumbed due to the anaphylactoid-type reactions. In contrast, normal mice and mice implanted with a related mammary carcinoma (66cl4) that does not induce CD11b(+)Gr-1(+) cell expansion, showed minimal responses. Depletion of phagocytic CD11b(+)Gr-1(+) cells prior to rAd delivery protected 4T1 tumor-bearing animals, whereas passive transfer of CD11b(+)Gr-1(+) cells from 4T1 tumor-bearing animals was sufficient to convey susceptibility to anaphylactoid-type reactions in normal animals. We further show that there is upregulation of nitric oxide and leukotriene signaling pathways in the 4T1 tumor-induced CD11b(+)Gr-1(+) myeloid cells and that pretreating mice with inhibitors of nitric oxide synthetase and leukotrienes can attenuate the anaphylactoid-type reactions. These data show that malignant tumor growth can alter CD11b(+)Gr-1(+) myeloid cells, rendering hosts susceptible to anaphylactoid-type reactions upon intravascular treatment with rAd.


Subject(s)
Adenoviridae/immunology , Anaphylaxis/metabolism , CD11b Antigen/immunology , Cell Differentiation , Genetic Therapy/adverse effects , Neoplasms/pathology , Receptors, Chemokine/metabolism , Adenoviridae/genetics , Anaphylaxis/physiopathology , Animals , Cardiovascular System/metabolism , Cell Line, Tumor , Disease Susceptibility , Female , Heart Rate , Humans , Leukotriene Antagonists/pharmacology , Liposomes , Mice , Myeloid Cells/metabolism , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Phagocytes , Receptors, Chemokine/immunology , Receptors, Leukotriene/metabolism
4.
Bioconjug Chem ; 17(2): 410-8, 2006.
Article in English | MEDLINE | ID: mdl-16536473

ABSTRACT

CC49 is a clinically validated antibody with specificity for TAG-72, a carbohydrate epitope that is overexpressed and exposed on the cell surface in a large fraction of solid malignancies. We constructed a single-chain fragment (scFv) based on CC49 and fused it to beta-lactamase (BLA). Following optimization of the scFv domain by combinatorial consensus mutagenesis (CCM) for increased expression and stability, we characterized the protein variant for binding, in vivo pharmacokinetics (PK), and antitumor efficacy. The fusion protein TAB2.5 possessed a similar binding specificity relative to the parent antibody CC49. TAB2.5 also showed prolonged retention (T(1/2) = 36.9 h) in tumor-bearing mice with tumor/plasma ratios of up to 1000. Preliminary evaluation of TAB2.5, in combination with a novel prodrug, GC-Mel, resulted in significant efficacy in a colorectal xenograft tumor model and supports the utility of the protein as an agent for tumor-selective prodrug activation.


Subject(s)
Antibodies, Neoplasm , Cephalosporins/chemistry , Cephalosporins/metabolism , Cephalosporins/therapeutic use , Nitrogen Mustard Compounds/chemistry , Nitrogen Mustard Compounds/metabolism , Nitrogen Mustard Compounds/therapeutic use , Prodrugs , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , beta-Lactamases , Animals , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/metabolism , Antibodies, Neoplasm/therapeutic use , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Delivery Systems , Female , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Irinotecan , Melphalan/chemistry , Melphalan/metabolism , Melphalan/therapeutic use , Mice , Mice, Nude , Molecular Structure , Neoplasm Transplantation , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/therapeutic use , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , beta-Lactamases/chemistry , beta-Lactamases/genetics , beta-Lactamases/metabolism , beta-Lactamases/therapeutic use
5.
Eur J Immunol ; 33(1): 172-82, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12594846

ABSTRACT

Linkage studies indicate close associations of certain HLA alleles with autoimmune diseases. To better understand how specific HLA alleles are related to disease pathogenesis, we have generated an HLA DR3/DQ2 transgenic mouse utilizing a 550-kb yeast artificial chromosome (YAC) construct containing the complete DRalpha, DRbeta1, DRbeta3, DQalpha, and DQbeta regions. The transgenic mouse (4D1/C2D) in an I-Abeta(o) background appears healthy with no signs of autoimmune diseases. Lymphoid tissues as well as CD4(+) T cells develop normally. Characterization of the transgene expression demonstrates that approximately 90% of B cells express high levels of DR3 and 50-70% of B cells express DQ2. CD11c(+) dendritic cells express high levels of DR and DQ. Approximately 12-18% of resting T cells are positive for DR expression, and further up-regulation to 40-50% expression is seen upon activation with anti-CD3/anti-CD28 mAb. These results suggest that the transgenic construct confers a high fidelity to the normal human temporal and spatial expression profile. Analysis of T cell receptor repertoire in transgenic mice confirms that DR3/DQ2 are able to mediate thymic selection. Furthermore, transgenic mice respond to a DR3-restricted antigen, demonstrating antigen processing and presentation by antigen-presenting cells (APC). Purified T cells from ovalbumin (OVA)-immunized 4D1 mice respond to human APC co-cultured with OVA, suggesting appropriate antigen/DR3 or DQ2 recognition by murine T cells. Immunoglobulin isotype switching is also observed, indicating functional T-B cognate interactions. Thus, the DR3/DQ2 transgenic mouse has normal lymphoid development and functionality that are mediated by HLA transgenes and can be used to investigate HLA-associated immunological questions.


Subject(s)
Autoimmune Diseases/immunology , HLA-DQ Antigens/immunology , HLA-DR3 Antigen/immunology , Models, Animal , Animals , Antigen-Presenting Cells/immunology , Antigens/immunology , Autoimmune Diseases/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Flow Cytometry , Gene Expression , HLA-DQ Antigens/genetics , HLA-DR3 Antigen/genetics , Humans , Mice , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thymus Gland/cytology , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...