Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Sci ; 109(3): 587-599, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29288540

ABSTRACT

Cancer immunotherapy with human γδ T cells expressing Vγ2Vδ2 T cell receptor (also termed Vγ9Vδ2) has shown promise because of their ability to recognize and kill most types of tumors in a major histocombatibility complex (MHC) -unrestricted fashion that is independent of the number of tumor mutations. In clinical trials, adoptive transfer of Vγ2Vδ2 T cells has been shown to be safe and does not require preconditioning. In this report, we describe a method for preparing highly enriched human Vγ2Vδ2 T cells using the bisphosphonate prodrug, tetrakis-pivaloyloxymethyl 2-(thiazole-2-ylamino)ethylidene-1,1-bisphosphonate (PTA). PTA stimulated the expansion of Vγ2Vδ2 cells to purities up to 99%. These levels were consistently higher than those observed after expansion with zoledronic acid, the most commonly used stimulator for clinical trials. Cell numbers also averaged more than those obtained with zoledronic acid and the expanded Vγ2Vδ2 cells exhibited high cytotoxicity against tumor cells. The high purity of Vγ2Vδ2 cells expanded by PTA increased engraftment success in immunodeficient NOG mice. Even low levels of contaminating αß T cells resulted in some mice with circulating human αß T cells rather than Vγ2Vδ2 cells. Vγ2Vδ2 cells from engrafted NOG mice upregulated CD25 and secreted tumor necrosis factor-α and interferon-γ in response to PTA-treated tumor cells. Thus, PTA expands Vγ2Vδ2 T cells to higher purity than zoledronic acid. The high purities allow the successful engraftment of immunodeficient mice without further purification and may speed up the development of allogeneic Vγ2Vδ2 T cell therapies derived from HLA-matched normal donors for patients with poor autologous Vγ2Vδ2 T cell responses.


Subject(s)
Breast Neoplasms/therapy , Diphosphonates/administration & dosage , Prodrugs/administration & dosage , Prostatic Neoplasms/therapy , T-Lymphocytes/transplantation , Animals , Breast Neoplasms/immunology , Diphosphonates/chemistry , Diphosphonates/pharmacology , Female , Humans , Immunotherapy, Adoptive , Male , Mice , Prodrugs/pharmacology , Prostatic Neoplasms/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/immunology , Treatment Outcome , Xenograft Model Antitumor Assays
2.
Sci Rep ; 7(1): 5987, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28729550

ABSTRACT

Bisphosphonates have benefits in breast cancer and multiple myeloma patients and have been used with adoptive immunotherapy with γδ T cells expressing Vγ2 Vδ2 TCRs. Although treatment with γδ T cells is safe, it has shown limited efficacy. Present bisphosphonates stimulate γδ T cells but were designed to inhibit bone resorption rather than treating cancer and have limited oral absorption, tumor cell entry, and cause bone side effects. The development of phosphate and phosphonate nucleotide prodrugs has led to important drugs for hepatitis C and HIV. Using a similar approach, we synthesized bisphosphonate prodrugs and found that they efficiently limit tumor cell growth. Pivoxil bisphosphonate esters enter cells where esterases convert them to their active acids. The bisphosphonate esters stimulated γδ T cells to secrete TNF-α in response to a variety of tumor cells more efficiently than their corresponding acids. The most active compound, tetrakis-pivaloyloxymethyl 2-(thiazole-2-ylamino)ethylidene-1,1- bisphosphonate (7), specifically expanded γδ T cells and stimulated them to secrete interferon-γ and kill tumor cells. In preclinical studies, combination therapy with compound 7 and γδ T cells prolonged survival of mice inoculated with either human bladder cancer or fibrosarcoma cells. Therefore, bisphosphonate prodrugs could enhance the effectiveness of adoptive cancer immunotherapy with γδ T cells.


Subject(s)
Antineoplastic Agents/pharmacology , Diphosphonates/pharmacology , Immunotherapy, Adoptive , Prodrugs/pharmacology , Animals , Cell Line, Tumor , Diphosphonates/chemistry , Esterification , Humans , Mice , Prenylation , Prodrugs/chemistry , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Tumor Necrosis Factor-alpha/metabolism , rap1 GTP-Binding Proteins/metabolism
3.
ChemMedChem ; 11(24): 2656-2663, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27786425

ABSTRACT

Nitrogen-containing bisphosphonates have antitumor activity in certain breast cancer and myeloma patients. However, these drugs have limited oral absorption, tumor cell entry and activity, and cause bone side effects. The potencies of phosphorylated antiviral drugs have been increased by administering them as prodrugs, in which the negative charges on the phosphate moieties are masked to make them lipophilic. We synthesized heterocyclic bisphosphonate (BP) prodrugs in which the phosphonate moieties are derivatized with pivaloyloxymethyl (pivoxil) groups and that lack the hydroxy "bone hook" on the geminal carbon. When the lipophilic BP prodrugs enter tumor cells, they are converted into their active forms by intracellular esterases. The most active BP prodrug, tetrakispivaloyloxymethyl 2-(thiazole-2-ylamino)ethylidene-1,1-bisphosphonate (7), was found to potently inhibit the in vitro growth of a variety of tumor cell lines, especially hematopoietic cells, at nanomolar concentrations. Consistent with this fact, compound 7 inhibited the prenylation of the RAP1A small GTPase signaling protein at concentrations as low as 1-10 nm. In preclinical studies, 7 slowed the growth of human bladder cancer cells in an immunodeficient mouse model. Thus, 7 is significantly more active than zoledronic acid, the most active FDA-approved BP, and a potential anticancer therapeutic.


Subject(s)
Diphosphonates/chemical synthesis , Diphosphonates/pharmacology , Drug Delivery Systems , Neoplasms/drug therapy , Prodrugs/chemistry , Prodrugs/pharmacology , Thiazoles/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Assay , Cell Line, Tumor , Cell Proliferation/drug effects , Diphosphonates/chemistry , Disease Models, Animal , Humans , Inhibitory Concentration 50 , Mice , Molecular Structure , Prodrugs/chemical synthesis , Thiazoles/chemistry , Thiazoles/pharmacology
4.
Cancer Sci ; 104(5): 536-42, 2013 May.
Article in English | MEDLINE | ID: mdl-23387443

ABSTRACT

Exposing human tumor cells to nitrogen-containing bisphosphonates, such as zoledronic acid (Zol), greatly increases their susceptibility to killing by γδ T cells. Based on this finding and other studies, cancer immunotherapy using γδ T cells and nitrogen-containing bisphosphonates has been studied in pilot clinical trials and has shown benefits. Although Zol treatment can render a wide variety of human tumor cells susceptible to γδ T cell killing, there has not been a systematic investigation to determine which types of tumor cells are the most susceptible to γδ T cell-mediated cytotoxicity. In this study, we determined the Zol concentrations required to stimulate half maximal tumor necrosis factor-α production by γδ T cells cultured with various tumor cell lines pretreated with Zol and compared these concentrations with those required for half maximal inhibition of farnesyl diphosphate synthase (FPPS) in the same tumor cell lines. The inhibition of tumor cell growth by Zol was also assessed. We found that FPPS inhibition strongly correlated with γδ T cell activation, confirming that the mechanism underlying γδ T cell activation by Zol is isopentenyl diphosphate (IPP) accumulation due to FPPS blockade. In addition, we showed that γδ T-cell receptor-mediated signaling correlated with γδ T cell tumor necrosis factor-α production and cytotoxicity. Some lymphoma, myeloid leukemia, and mammary carcinoma cell lines were relatively resistant to Zol treatment, suggesting that assessing tumor sensitivity to Zol may help select those patients most likely to benefit from immunotherapy with γδ T cells.


Subject(s)
Diphosphonates/pharmacology , Geranyltranstransferase/antagonists & inhibitors , Imidazoles/pharmacology , Leukemia, Myeloid/drug therapy , Lymphoma/drug therapy , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/drug effects , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Diphosphonates/immunology , Geranyltranstransferase/immunology , Geranyltranstransferase/metabolism , HL-60 Cells , Hemiterpenes/immunology , Hemiterpenes/metabolism , Humans , Imidazoles/immunology , K562 Cells , Leukemia, Myeloid/immunology , Leukemia, Myeloid/metabolism , Lymphoma/immunology , Lymphoma/metabolism , MCF-7 Cells , Organophosphorus Compounds/immunology , Organophosphorus Compounds/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , U937 Cells , Zoledronic Acid
5.
Cancer Immunol Immunother ; 62(4): 677-87, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23151944

ABSTRACT

Human γδ T cells display potent cytotoxicity against various tumor cells pretreated with zoledronic acid (Zol). Zol has shown benefits when added to adjuvant endocrine therapy for patients with early-stage breast cancer or to standard chemotherapy for patients with multiple myeloma. Although γδ T cells may contribute to this additive effect, the responsiveness of γδ T cells from early-stage breast cancer patients has not been fully investigated. In this study, we determined the number, frequency, and responsiveness of Vγ2Vδ2 T cells from early- and late-stage breast cancer patients and examined the effect of IL-18 on their ex vivo expansion. The responsiveness of Vγ2Vδ2 T cells from patients with low frequencies of Vγ2Vδ2 T cells was significantly diminished. IL-18, however, enhanced ex vivo proliferative responses of Vγ2Vδ2 T cells and helper NK cells from patients with either low or high frequencies of Vγ2Vδ2 T cells. Treatment of breast cancer patients with Zol alone decreased the number of Vγ2Vδ2 T cells and reduced their ex vivo responsiveness. These results demonstrate that Zol can elicit immunological responses by γδ T cells from early-stage breast cancer patients, but that frequent in vivo treatment reduces Vγ2Vδ2 T cell numbers and their responsiveness to stimulation.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/therapy , Diphosphonates/therapeutic use , Imidazoles/therapeutic use , Interleukin-18/therapeutic use , Killer Cells, Natural/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes, Helper-Inducer/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Clinical Trials as Topic , Combined Modality Therapy , Female , Humans , Immunotherapy, Adoptive/methods , Interleukin-18/immunology , Killer Cells, Natural/drug effects , Leukocytes, Mononuclear/immunology , Middle Aged , Neoplasm Staging , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , T-Lymphocytes, Helper-Inducer/drug effects , Zoledronic Acid
6.
Eur J Immunol ; 41(2): 345-55, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268005

ABSTRACT

Programmed cell death-1 (PD-1) is an inhibitory receptor and plays an important role in the regulation of αß T cells. Little is known, however, about the role of PD-1 in γδ T cells. In this study, we investigated the expression and function of PD-1 in human γδ T cells. Expression of PD-1 was rapidly induced in primary γδ T cells following antigenic stimulation, and the PD-1(+) γδ T cells produced IL-2. When PD-1(+) γδ T cells were stimulated with Daudi cells with and without programmed cell death ligand-1 (PD-L1) expression, the levels of IFN-γ production and cytotoxicity in response to PD-L1(+) Daudi cells were diminished compared to the levels seen in response to PD-L1(-) Daudi cells. The attenuated effector functions were reversed by anti-PD-L1 mAb. When PD-1(+) γδ T cells were challenged by PD-L1(+) tumors pretreated with zoledronate (Zol), which induced γδ TCR-mediated signaling, the resulting reduction in cytokine production was only slight to moderate compared to the reduction seen when PD-1(+) γδ T cells were challenged by PD-L1(-) tumors. In addition, cytotoxic activity of PD-1(+) γδ T cells against Zol-treated PD-L1(+) tumors was comparable to that against Zol-treated PD-L1(-) tumors. These results suggest that TCR triggering may partially overcome the inhibitory effect of PD-1 in γδ T cells.


Subject(s)
Antigens, CD/metabolism , Antigens/immunology , Apoptosis Regulatory Proteins/metabolism , Organophosphorus Compounds/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Antigens, CD/genetics , Antigens, Surface/metabolism , B7-H1 Antigen , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Diphosphates/immunology , Diphosphonates/immunology , Diphosphonates/pharmacology , Humans , Imidazoles/immunology , Imidazoles/pharmacology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Jurkat Cells , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Organophosphorus Compounds/pharmacology , Programmed Cell Death 1 Receptor , Zoledronic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...