Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 224
Filter
1.
Cytotherapy ; 22(10): 552-562, 2020 10.
Article in English | MEDLINE | ID: mdl-32747298

ABSTRACT

BACKGROUND AIMS: Anti-CD19 chimeric antigen receptor (CAR)-modified T cells have shown dramatic cytotoxicity against B-cell malignancies. Currently, autologous T cells are conventionally used to manufacture CAR T cells. Low quality or insufficient quantity of autologous T cells may lead to failure of CAR T preparations. Moreover, CAR T preparation usually takes 1-2 weeks, which is too long for patients with rapid disease progression to successfully infuse CAR T cells. Thus, the development of a ready-to-use CAR immunotherapy strategy is needed. NK-92, a natural killer (NK) cell line derived from an NK lymphoma patient, has been gradually applied as a CAR-modified effector cell. To avoid the potential development of secondary NK lymphoma in patients, large doses of radiation are used to treat NK-92 cells before clinical application, which ensures the safety but reduces the cytotoxicity of NK-92 cells. Therefore, it is crucial to explore a suitable radiation dose that ensures short life span and good cytotoxicity of CAR NK-92 cells. METHODS: NK-92MI, a modified IL-2-independent NK-92 cell line, was used to establish an anti-CD19 CAR NK. The suitable radiation dose of CAR NK was then explored in vitro and validated in vivo, and the specific cytotoxicity of irradiated and unirradiated CAR NK against CD19+ malignant cells was assessed. RESULTS: CAR NK exhibited specific cytotoxicity against CD19+ malignant cells. Irradiation ensured a short life span of CAR NK in vitro and in vivo. Encouragingly, irradiated CAR NK displayed an anti-CD19+ malignancy capacity similar to that of unirradiated CAR NK. CONCLUSIONS: Five Gy is a suitable radiation dose to ensure the safety and effectiveness of CD19 CAR NK-92MI cells.


Subject(s)
Antigens, CD19/metabolism , Cytotoxicity, Immunologic , Receptors, Chimeric Antigen/metabolism , Adult , Aged , Animals , B-Lymphocytes/immunology , B-Lymphocytes/radiation effects , Cell Line, Tumor , Cell Proliferation , Cytotoxicity, Immunologic/radiation effects , Disease Models, Animal , Dose-Response Relationship, Radiation , Female , Humans , Immunotherapy, Adoptive , Killer Cells, Natural/immunology , Killer Cells, Natural/radiation effects , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Young Adult
2.
Sci Adv ; 6(13): eaay9789, 2020 03.
Article in English | MEDLINE | ID: mdl-32232155

ABSTRACT

Radiotherapy (RT) is routinely used in cancer treatment, but expansion of its clinical indications remains challenging. The mechanism underlying the radiation-induced bystander effect (RIBE) is not understood and not therapeutically exploited. We suggest that the RIBE is predominantly mediated by irradiated tumor cell-released microparticles (RT-MPs), which induce broad antitumor effects and cause immunogenic death mainly through ferroptosis. Using a mouse model of malignant pleural effusion (MPE), we demonstrated that RT-MPs polarized microenvironmental M2 tumor-associated macrophages (M2-TAMs) to M1-TAMs and modulated antitumor interactions between TAMs and tumor cells. Following internalization of RT-MPs, TAMs displayed increased programmed cell death ligand 1 (PD-L1) expression, enhancing follow-up combined anti-PD-1 therapy that confers an ablative effect against MPE and cisplatin-resistant MPE mouse models. Immunological memory effects were induced.


Subject(s)
Cell-Derived Microparticles/metabolism , Cellular Reprogramming/immunology , Cytotoxicity, Immunologic , Neoplasms/immunology , Neoplasms/metabolism , Radiation, Ionizing , Animals , Biomarkers , Biomarkers, Tumor , Bystander Effect/immunology , Bystander Effect/radiation effects , Cell Line, Tumor , Cellular Reprogramming/radiation effects , Cytotoxicity, Immunologic/radiation effects , Disease Models, Animal , Humans , Immunologic Memory , Janus Kinases/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Neoplasms/pathology , Neoplasms/therapy , STAT Transcription Factors/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
3.
Epigenomics ; 12(5): 397-408, 2020 03.
Article in English | MEDLINE | ID: mdl-32267172

ABSTRACT

Aim: We here hypothesized that tumor-derived exosomal miRNA (TexomiR) released from irradiated tumors may play a role in the tumor cells escape to natural killer (NK) cells. Materials & methods: Our study included the use of different cancer cell lines, blood biopsies of xenograph mice model and patients treated with radiotherapy. Results: The irradiation of cancer cells promotes the TET2-mediated demethylation of miR-378 promoter, miR-378a-3p overexpression and its loading in exosomes, inducing the decrease of granzyme-B (GZMB) secretion by NK cells. An inverse correlation between TexomiR-378a-3p and GZMB was observed in murine and human blood samples. Conclusion: Our work identifies TexomiR-378a-3p as a molecular signature associated with the loss of NK cells cytotoxicity via the decrease of GZMB expression upon radiotherapy.


Subject(s)
Exosomes/metabolism , Gene Expression Regulation, Neoplastic/radiation effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , MicroRNAs/genetics , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/radiation effects , DNA Methylation , DNA-Binding Proteins/metabolism , Dioxygenases , Gene Expression , Granzymes/metabolism , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Radiotherapy
4.
Sci Adv ; 6(8): eaay9209, 2020 02.
Article in English | MEDLINE | ID: mdl-32128416

ABSTRACT

T cells engineered to express chimeric antigen receptors (CARs) can recognize and engage with target cancer cells with redirected specificity for cancer immunotherapy. However, there is a lack of ideal CARs for solid tumor antigens, which may lead to severe adverse effects. Here, we developed a light-inducible nuclear translocation and dimerization (LINTAD) system for gene regulation to control CAR T activation. We first demonstrated light-controllable gene expression and functional modulation in human embryonic kidney 293T and Jurkat T cell lines. We then improved the LINTAD system to achieve optimal efficiency in primary human T cells. The results showed that pulsed light stimulations can activate LINTAD CAR T cells with strong cytotoxicity against target cancer cells, both in vitro and in vivo. Therefore, our LINTAD system can serve as an efficient tool to noninvasively control gene activation and activate inducible CAR T cells for precision cancer immunotherapy.


Subject(s)
Immunotherapy, Adoptive , Light , Neoplasms/immunology , Neoplasms/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Animals , Antigens, CD19/immunology , Antigens, Neoplasm/immunology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , Cryptochromes/genetics , Cryptochromes/metabolism , Cytotoxicity, Immunologic/immunology , Cytotoxicity, Immunologic/radiation effects , Humans , Immunotherapy, Adoptive/methods , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Activation/radiation effects , Mice , Models, Biological , Protein Binding , Protein Multimerization , Receptors, Chimeric Antigen/genetics , Transcriptional Activation/radiation effects , Xenograft Model Antitumor Assays
5.
J Clin Invest ; 129(10): 4224-4238, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31483286

ABSTRACT

Treatment of tumors with ionizing radiation stimulates an antitumor immune response partly dependent on induction of IFNs. These IFNs directly enhance dendritic cell and CD8+ T cell activity. Here we show that resistance to an effective antitumor immune response is also a result of IFN signaling in a different cellular compartment of the tumor, the cancer cells themselves. We abolished type I IFN signaling in cancer cells by genetic elimination of its receptor, IFNAR1. Pronounced immune responses were provoked after ionizing radiation of tumors from 4 mouse cancer cell lines with Ifnar1 knockout. This enhanced response depended on CD8+ T cells and was mediated by enhanced susceptibility to T cell-mediated killing. Induction of Serpinb9 proved to be the mechanism underlying control of susceptibility to T cell killing after radiation. Ifnar1-deficient tumors had an augmented response to anti-PD-L1 immunotherapy with or without radiation. We conclude that type I IFN can protect cancer cells from T cell-mediated cytotoxicity through regulation of Serpinb9. This result helps explain why radiation of tumors can stimulate antitumor immunity yet also result in resistance. It further suggests potential targets for intervention to improve therapy and to predict responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Interferon Type I/immunology , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/radiotherapy , Cell Line, Tumor , Cytotoxicity, Immunologic/radiation effects , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/radiotherapy , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/radiotherapy , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Serpins/genetics , Serpins/immunology , Signal Transduction/immunology , Signal Transduction/radiation effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
6.
Cytotherapy ; 20(5): 715-727, 2018 05.
Article in English | MEDLINE | ID: mdl-29572116

ABSTRACT

BACKGROUND AIMS: Irradiation enhances the adhesion between natural killer (NK) cells and target cells by up-regulating intercellular adhesion molecule-1 (ICAM-1) on target cells. Therefore, we investigated the effect of irradiation-induced ICAM-1 expression on human cancer cells on NK cell-mediated cytotoxicity. METHODS: Expression levels of ICAM-1 on the target cell surface before and after irradiation of six human cancer cell lines (HL60, SKBR-3, T47D, HCT-116, U937 and U251) were analyzed by flow cytometry. Ex vivo expansion of NK cells from human peripheral blood mononuclear cells was performed by co-culture with irradiated K562 cells. The related adhesion molecule lymphocyte function-associated antigen 1 (LFA-1) on NK cells was analyzed by flow cytometry. An enzyme-linked immunosorbent assay was used to detect interferon-γ (IFN-γ), and WST-8 assays were performed to check NK cell cytotoxicity. Finally, blocking assays were performed using monoclonal antibodies against ICAM-1 or LFA-1. RESULTS: LFA-1 expression increased on NK cells after expansion (P <0.001). The expression of ICAM-1 was significantly upregulated by irradiation after 24 h in various cell lines, including HL60 (P <0.001), SKBR-3 (P <0.001), T47D (P <0.001) and U937 (P <0.001), although the level of expression depended on the cell line. ICAM-1 expression was extremely low before and after irradiation in U251 cells. NK cell-mediated cytotoxicity increased after irradiation of HL60 (P <0.001), SKBR-3 (P <0.001), T47D (P = 0.003), and U937 (P = 0.004) cells, in which ICAM-1 expression was significantly increased after irradiation. IFN-γ production by NK cells in response to HL60 (P <0.001) and T47D (P = 0.011) cells significantly increased after irradiation. NK cell-mediated cytotoxicity against irradiated SKBR-3 (P <0.001) and irradiated T47D cells (P = 0.035) significantly decreased after blocking of ICAM-1. Blocking of LFA-1 on NK cells resulted in reduced cytotoxicity against irradiated HL60 (P <0.001) and irradiated SKBR-3 (P <0.001). CONCLUSIONS: Irradiation upregulates ICAM-1 expression on the surface of human cancer cells and enhances activated NK cell-mediated cytotoxicity. Therefore, irradiation combined with NK cell therapy may improve the antitumor effects of NK cells.


Subject(s)
Cytotoxicity, Immunologic/radiation effects , Intercellular Adhesion Molecule-1/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/radiation effects , Neoplasms/immunology , Neoplasms/metabolism , Radiation, Ionizing , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Cytotoxicity, Immunologic/drug effects , Humans , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Kinetics , Lymphocyte Function-Associated Antigen-1/metabolism , Up-Regulation/drug effects
7.
Tumour Biol ; 39(3): 1010428317695961, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28349820

ABSTRACT

Recurrence of breast cancer after radiotherapy may be partly explained by the presence of radioresistant cells. Thus, it would be desirable to develop an effective therapy against radioresistant cells. In this study, we demonstrated the intense antitumor activity of cytokine-induced killer cells against MCF-7 and radioresistant MCF-7 cells, as revealed by cytokine-induced killer-mediated cytotoxicity, tumor cell proliferation, and tumor invasion. Radioresistant MCF-7 cells were more susceptible to cytokine-induced killer cell killing. The stronger cytotoxicity of cytokine-induced killer cells against radioresistant MCF-7 cells was dependent on the expression of major histocompatibility complex class I polypeptide-related sequence A/B on radioresistant MCF-7 cells after exposure of cytokine-induced killer cells to sensitized targets. In addition, we demonstrated that cytokine-induced killer cell treatment sensitized breast cancer cells to chemotherapy via the downregulation of TK1, TYMS, and MDR1. These results indicate that cytokine-induced killer cell treatment in combination with radiotherapy and/or chemotherapy may induce synergistic antitumor activities and represent a novel strategy for breast cancer.


Subject(s)
Breast Neoplasms/radiotherapy , Cell- and Tissue-Based Therapy , Cytokine-Induced Killer Cells/metabolism , Neoplasm Recurrence, Local/radiotherapy , ATP Binding Cassette Transporter, Subfamily B/biosynthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cytokine-Induced Killer Cells/immunology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/radiation effects , Female , Humans , MCF-7 Cells , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Radiation Tolerance , Thymidine Kinase/biosynthesis , Thymidylate Synthase/biosynthesis
8.
Oncotarget ; 7(52): 86937-86947, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27893426

ABSTRACT

Radium-223 dichloride (Xofigo®; 223Ra) is an alpha-emitting radiopharmaceutical FDA-approved for the treatment of bone metastases in patients with advanced castration-resistant prostate cancer. It is also being examined clinically in patients with breast and lung carcinoma and patients with multiple myeloma. As with other forms of radiation, the aim of 223Ra is to reduce tumor burden by directly killing tumor cells. External beam (photon) and proton radiation have been shown to augment tumor sensitivity to antigen-specific CD8+ cytotoxic T lymphocytes (CTLs). However, little is known about whether treatment with 223Ra can also induce such immunogenic modulation in tumor cells that survive irradiation. We examined these effects in vitro by exposing human prostate, breast, and lung carcinoma cells to sublethal doses of 223Ra. 223Ra significantly enhanced T cell-mediated lysis of each tumor type by CD8+ CTLs specific for MUC-1, brachyury, and CEA tumor antigens. Immunofluorescence analysis revealed that the increase in CTL killing was accompanied by augmented protein expression of MHC-I and calreticulin in each tumor type, molecules that are essential for efficient antigen presentation. Enhanced tumor-cell lysis was facilitated by calreticulin surface translocation following 223Ra exposure. The phenotypic changes observed after treatment appear to be mediated by induction of the endoplasmic reticulum stress response pathway. By rendering tumor cells more susceptible to T cell-mediated lysis, 223Ra may potentially be effective in combination with various immunotherapies, particularly cancer vaccines that are designed to generate and expand patients' endogenous antigen-specific T-cell populations against specific tumor antigens.


Subject(s)
Alpha Particles/therapeutic use , Calreticulin/physiology , Cytotoxicity, Immunologic/radiation effects , Neoplasms/radiotherapy , Radium/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Calreticulin/analysis , Cell Line, Tumor , Cell Proliferation/radiation effects , Dose-Response Relationship, Radiation , Endoplasmic Reticulum Stress/radiation effects , HLA Antigens/analysis , Humans , Neoplasms/immunology
9.
Cell Immunol ; 303: 16-23, 2016 May.
Article in English | MEDLINE | ID: mdl-27113815

ABSTRACT

Radiotherapy has been employed for the treatment of oncological patients for nearly a century, and together with surgery and chemotherapy, radiation oncology constitutes one of the three pillars of cancer therapy. Ionizing radiation has complex effects on neoplastic cells and on tumor microenvironment: beyond its action as a direct cytotoxic agent, tumor irradiation triggers a series of alterations in tumoral cells, which includes the de novo synthesis of particular proteins and the up/down-regulation of cell surface molecules. Additionally, ionizing radiation may induce the release of "danger signals" which may, in turn lead to cellular and molecular responses by the immune system. This immunomodulatory action of ionizing radiation highlights the importance of the combined use (radiotherapy plus immunotherapy) for cancer healing. Major histocompatibility complex antigens (also called Human Leukocyte Antigens, HLA in humans) are one of those molecules whose expression is modulated after irradiation. This review summarizes the modulatory properties of ionizing radiation on the expression of HLA class I (classical and non-classical) and class II molecules, with special emphasis in non-classical HLA-I molecules.


Subject(s)
HLA Antigens/metabolism , Killer Cells, Natural/immunology , Neoplasms/radiotherapy , Radiation, Ionizing , T-Lymphocytes, Cytotoxic/immunology , Cytotoxicity, Immunologic/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , HLA Antigens/genetics , HLA-G Antigens/genetics , HLA-G Antigens/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immunity/radiation effects , Immunomodulation , Neoplasms/genetics , Neoplasms/metabolism , Radioimmunotherapy , Tumor Microenvironment/radiation effects , HLA-E Antigens
10.
Lancet Oncol ; 16(13): e498-509, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26433823

ABSTRACT

Checkpoint blockade immunotherapy has received mainstream attention as a result of striking and durable clinical responses in some patients with metastatic disease and a reasonable response rate in many tumour types. The activity of checkpoint blockade immunotherapy is not restricted to melanoma or lung cancer, and additional indications are expected in the future, with responses already reported in renal cancer, bladder cancer, and Hodgkin's lymphoma among many others. Additionally, the interactions between radiation and the immune system have been investigated, with several studies describing the synergistic effects on local and distant tumour control when radiation therapy is combined with immunotherapy. Clinical enthusiasm for this approach is strengthened by the many ongoing trials combining immunotherapy with definitive and palliative radiation. Herein, we discuss the biological and mechanistic rationale behind combining radiation with checkpoint blockade immunotherapy, with a focus on the preclinical data supporting this potentially synergistic combination. We explore potential hypotheses and important considerations for clinical trial designs. Finally, we reintroduce the notion of radiosensitising immunotherapy, akin to radiosensitising chemotherapy, as a potential definitive therapeutic modality.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Chemoradiotherapy/methods , Immunotherapy/methods , Neoplasms/therapy , Radiation Tolerance , Radiation-Sensitizing Agents/therapeutic use , Signal Transduction/drug effects , Signal Transduction/radiation effects , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/radiation effects , Dendritic Cells/diagnostic imaging , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/radiation effects , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/pathology , Radiation Dosage , Radiography , Treatment Outcome
11.
J Immunother ; 38(3): 107-15, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751500

ABSTRACT

Regulatory T cells (Tregs) is one of the main obstacles to the success of cancer immunotherapy. The effect of dendritic cell (DC)-based immunotherapy can be attenuated by immune suppressive functions of Tregs. We used a CD25-targeted antibody and low-dose cyclophosphamide (CTX) as immunomodulators to increase the antitumor effect of intratumoral injection of immature DCs into the irradiated tumor cells (IR/iDC). CTX or CD25-targeted antibody alone showed a significant reduction in the number of Tregs within the tumor microenvironment. When they are combined with IR/iDC, the number of Tregs was further reduced. Although IR/IDC showed strong antitumor effects such as reduction in tumor growth, increase in Th1 immune response, and improvement of survival, the therapeutic effect was further improved by combining treatments with immunomodulators. CTX and CD25-targeted antibody showed no significant difference in tumor growth when combined with IR/iDC, but CTX further increased the number of interferon (IFN)-γ-secreting T cells, cytotoxicity, and survival rate. Although irradiation induced depletion of T lymphocytes, administration of DCs recovered this depletion. Particularly, the lymphocytes were more significantly increased when CTX and IR/iDC were combined. Low-dose CTX has already been used as an immunomodulator in clinical trials, and it offers several advantages, including convenience, low-cost, and familiarity to clinicians. However, CD25-targeted antibody cannot only deplete Tregs, but also may affect IL-2-dependent effector T lymphocytes. Therefore, CTX is an effective means to inhibit Tregs, and an effective immunomodulatory agent for multimodality therapy such as combination treatment of conventional cancer therapy and immunotherapy.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Carcinoma, Lewis Lung/immunology , Cyclophosphamide/administration & dosage , Dendritic Cells/immunology , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Lewis Lung/mortality , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/therapy , Cell Line, Tumor , Combined Modality Therapy , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/radiation effects , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Immunophenotyping , Immunosuppressive Agents/administration & dosage , Immunotherapy , Male , Mice , Phenotype , Radiation , Spleen/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/radiation effects , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
12.
Mol Med Rep ; 11(3): 1813-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25420581

ABSTRACT

Ionizing irradiation is a well­established therapeutic modality for malignant gliomas. Due to its high cellular uptake, 5­aminolevulinic acid (ALA) is used for fluorescence­guided resection of malignant gliomas. We have previously shown that 5­ALA sensitizes glioma cells to irradiation in vitro. The aim of the present study was to assess whether 5­ALA acts as a radiosensitizer in experimental glioma in vivo. Rats were subcutaneously injected with 9L gliosarcoma cells and administered 5­ALA. The accumulation of 5­ALA­induced protoporphyrin IX was confirmed by high­performance liquid chromatography (HPLC) analysis. Subcutaneous (s.c.) tumors were subsequently irradiated with 2 Gy/day for five consecutive days. In the experimental glioma model, high­performance liquid chromatography analysis revealed a high level of accumulation of 5­ALA­induced protoporphyrin IX in s.c. tumors 3 h after 5­ALA administration. Multi­dose ionizing irradiation induced greater inhibition of tumor growth in rats that were administered 5­ALA than in the non­5­ALA­treated animals. Immunohistochemical analysis of the s.c. tumors revealed that numerous ionized calcium­binding adapter molecule 1 (Iba1)­positive macrophages gathered at the surface of and within the s.c. tumors following multi­dose ionizing irradiation in combination with 5­ALA administration. By contrast, the s.c. tumors in the control group scarcely showed aggregation of Iba1­positive macrophages. These results suggested that multi­dose ionizing irradiation with 5­ALA induced not only a direct cytotoxic effect but also enhanced the host antitumor immune response and thus caused high inhibition of tumor growth in experimental glioma.


Subject(s)
Aminolevulinic Acid/pharmacology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/radiation effects , Glioma/immunology , Glioma/pathology , Protoporphyrins/biosynthesis , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Disease Models, Animal , Glioma/therapy , Male , Necrosis , Photochemotherapy , Protoporphyrins/chemistry , Radiation Dosage , Rats , Tumor Burden/drug effects , Tumor Burden/radiation effects
13.
J Immunol ; 192(7): 3101-10, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24600032

ABSTRACT

As a side effect of cancer radiotherapy, immune cells receive varying doses of radiation. Whereas high doses of radiation (>10 Gy) can lead to lymphopenia, lower radiation doses (2-4 Gy) represent a valid treatment option in some hematological cancers, triggering clinically relevant immunological changes. Based on our earlier observations, we hypothesized that lower radiation doses have a direct positive effect on T cells. In this study, we show that 0.6-2.4 Gy radiation enhances proliferation and IFN-γ production of PBMC or purified T cells induced by stimulation via the TCR. Radiation with 1.2 Gy also lowered T cell activation threshold and broadened the Th1 cytokine profile. Although radiation alone did not activate T cells, when followed by TCR stimulation, ERK1/2 and Akt phosphorylation increased above that induced by stimulation alone. These changes were followed by an early increase in glucose uptake. Naive (CD45RA(+)) or memory (CD45RA(-)) T cell responses to stimulation were boosted at similar rates by radiation. Whereas increased Ag-specific cytotoxic activity of a CD8(+) T cell line manifested in a 4-h assay (10-20% increase), highly significant (5- to 10-fold) differences in cytokine production were detected in 6-d Ag-stimulation assays of PBMC, probably as a net outcome of death of nonstimulated and enhanced response of Ag-stimulated T cells. T cells from patients receiving pelvic radiation (2.2-2.75 Gy) also displayed increased cytokine production when stimulated in vitro. We report in this study enhanced T cell function induced by synergistic radiation treatment, with potential physiological significance in a wide range of T cell responses.


Subject(s)
Cell Proliferation/radiation effects , Peptides/immunology , T-Lymphocytes/immunology , T-Lymphocytes/radiation effects , Amino Acid Sequence , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Cytotoxicity, Immunologic/radiation effects , Dose-Response Relationship, Radiation , Epitopes, T-Lymphocyte/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Glucose/immunology , Glucose/pharmacokinetics , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/radiation effects , Lymphocyte Activation/immunology , Lymphocyte Activation/radiation effects , Male , Phosphorylation/immunology , Phosphorylation/radiation effects , Prostatic Neoplasms/blood , Prostatic Neoplasms/radiotherapy , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
14.
Radiat Oncol ; 9: 49, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24512718

ABSTRACT

BACKGROUND: The overexpression of histone deacetylase (HDAC) and a subsequent decrease in the acetylation levels of nuclear histones are frequently observed in cancer cells. Generally it was accepted that the deacetylation of histones suppressed expression of the attached genes. Therefore, it has been suggested that HDAC might contribute to the survival of cancer cells by altering the NKG2D ligands transcripts. By the way, the translational regulation of NKG2D ligands remains unclear in cancer cells. It appears the modulation of this unclear mechanism could enhance NKG2D ligand expressions and the susceptibility of cancer cells to NK cells. Previously, it was reported that irradiation can increase the surface expressions of NKG2D ligands on several cancer cell types without increasing the levels of NKG2D ligand transcripts via ataxia telangiectasia mutated and ataxia telangiectasia and Rad3 related (ATM-ATR) pathway, and suggested that radiation therapy might be used to increase the translation of NKG2D ligands. METHODS: Two NSCLC cell lines, that is, A549 and NCI-H23 cells, were used to investigate the combined effects of ionizing radiation and HDAC inhibitors on the expressions of five NKG2D ligands. The mRNA expressions of the NKG2D ligands were quantitated by multiplex reverse transcription-PCR. Surface protein expressions were measured by flow cytometry, and the susceptibilities of cancer cells to NK cells were assayed by time-resolved fluorometry using the DELFIA® EuTDA cytotoxicity kit and by flow cytometry. RESULTS: The expressions of NKG2D ligands were found to be regulated at the transcription and translation levels. Ionizing radiation and HDAC inhibitors in combination synergistically increased the expressions of NKG2D ligands. Furthermore, treatment with ATM-ATR inhibitors efficiently blocked the increased translations of NKG2D ligands induced by ionizing radiation but did not block the increased ligand translations induced by HDAC inhibitors. The study confirms that increased NKG2D ligand levels by ionizing radiation and HDAC inhibitors could synergistically enhance the susceptibilities of cancer cells to NK-92 cells. CONCLUSIONS: This study suggests that the expressions of NKG2D ligands are regulated in a complex manner at the multilevel of gene expression, and that their expressions can be induced by combinatorial treatments in lung cancer cells.


Subject(s)
Cytotoxicity, Immunologic , Histone Deacetylase Inhibitors/pharmacology , Killer Cells, Natural/immunology , Radiation, Ionizing , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Cells, Cultured , Combined Modality Therapy , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/radiation effects , Drug Synergism , Gene Expression/drug effects , Gene Expression/radiation effects , Histone Deacetylase Inhibitors/therapeutic use , Humans , Ligands , Lung Neoplasms/pathology , Lung Neoplasms/therapy , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Up-Regulation/drug effects , Up-Regulation/radiation effects
15.
Cancer Immunol Immunother ; 63(3): 259-71, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24357146

ABSTRACT

The need for an intact immune system for cancer radiation therapy to be effective suggests that radiation not only acts directly on the tumor but also indirectly, through the activation of host immune components. Recent studies demonstrated that endogenous type I interferons (type I IFNs) play a role in radiation-mediated anti-tumor immunity by enhancing the ability of dendritic cells to cross-prime CD8(+) T cells. However, it is still unclear to what extent endogenous type I IFNs contribute to the recruitment and function of CD8(+) T cells. Little is also known about the effects of type I IFNs on myeloid cells. In the current study, we demonstrate that type I and type II IFNs (IFN-γ) are both required for the increased production of CXCL10 (IP-10) chemokine by myeloid cells within the tumor after radiation treatment. Radiation-induced intratumoral IP-10 levels in turn correlate with tumor-infiltrating CD8(+) T cell numbers. Moreover, type I IFNs promote potent tumor-reactive CD8(+) T cells by directly affecting the phenotype, effector molecule production, and enhancing cytolytic activity. Using a unique inducible expression system to increase local levels of IFN-α exogenously, we show here that the capacity of radiation therapy to result in tumor control can be enhanced. Our preclinical approach to study the effects of local increase in IFN-α levels can be used to further optimize the combination therapy strategy in terms of dosing and scheduling, which may lead to better clinical outcome.


Subject(s)
CD8-Positive T-Lymphocytes/radiation effects , Interferon-alpha/metabolism , Mammary Neoplasms, Animal/radiotherapy , Melanoma, Experimental/immunology , Melanoma, Experimental/radiotherapy , Myeloid Cells/drug effects , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/radiation effects , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/radiation effects , Gene Expression Regulation/genetics , Gene Expression Regulation/radiation effects , Humans , Interferon-alpha/genetics , Interferon-alpha/pharmacology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lymphocyte Count , Mammary Neoplasms, Animal/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Neoplasm Transplantation
16.
Int J Cancer ; 134(10): 2383-92, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24154990

ABSTRACT

Radiation therapy (RT) continues to be a cornerstone in the treatment for many cancers. Unfortunately, not all individuals respond effectively to RT resulting clinically in two groups consisting of nonresponders (progressive disease) and responders (tumor control/cure). The mechanisms that govern the outcome of radiotherapy are poorly understood. Interestingly, a new paradigm has emerged demonstrating that the immune system mediates many of the antitumor effects of RT. Therefore, we hypothesized that the immune response following RT may dictate the efficacy of treatment. To examine this, we developed a tumor model that mirrors this clinically relevant phenomenon in which mice bearing Colon38, a colon adenocarcinoma, were treated locally with 15Gy RT resulting in both nonresponders and responders. More importantly, we were able to distinguish responders from nonresponders as early as 4 days post-RT allowing for the unique opportunity to identify critical events that ultimately determined the effectiveness of therapy. Intratumoral immune cells and interferon-gamma were increased in responsive tumors and licensed CD8 T cells to exhibit lytic activity against tumor cells, a response that was diminished in tumors refractory to RT. Combinatorial treatment with RT and the immunomodulatory cytokine IL-12 resulted in complete remission of cancer in 100% of cases compared to a cure rate of only 12% with RT alone. Similar data were obtained when IL-12 was delivered by microspheres. Therefore, the efficacy of RT may depend on the strength of the immune response induced after radiotherapy. Additionally, immunotherapy that further stimulates the immune cells may enhance the effectiveness of RT.


Subject(s)
Adenocarcinoma/radiotherapy , CD8-Positive T-Lymphocytes/radiation effects , Colonic Neoplasms/radiotherapy , Cytotoxicity, Immunologic/radiation effects , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Analysis of Variance , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Chemoradiotherapy , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Cytotoxicity, Immunologic/drug effects , Immune System/drug effects , Immune System/pathology , Immune System/radiation effects , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12/pharmacology , Mice , Treatment Outcome
17.
Am J Pathol ; 182(6): 2345-54, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23583648

ABSTRACT

Cancer treatments using ionizing radiation (IR) therapy are thought to act primarily through the induction of tumor cell damage at a molecular level. However, a new concept has recently emerged, suggesting that the immune system is required for effective IR therapy. Our work here has identified interferon gamma (IFN-γ) as an essential cytokine for the efficacy of IR therapy. Local IR (15 Gy) to mice bearing Colon38, a colon adenocarcinoma, decreases tumor burden in wild-type animals. Interestingly, IR therapy had no effect on tumor burden in IFNγKO mice. We further determined that intratumoral levels of IFN-γ increased 2 days following IR, which directly correlated with a decrease in tumor burden that was not a result of direct cytotoxic effects of IFN-γ on tumor cells. T cells from IR-treated tumors exhibited a far greater capacity to lyse tumor cells in a (51)Cr release assay, a process that was dependent on IFN-γ. CD8(+) T cells were the predominant producers of IFN-γ, as demonstrated by IFN-γ intracellular staining and studies in IFN-γ reporter mice. Elimination of CD8(+) T cells by antibody treatment reduced the intratumoral levels of IFN-γ by over 90%. More importantly, elimination of CD8(+) T cells completely abrogated the effects of radiation therapy. Our data suggest that IFN-γ plays a pivotal role in mediating the antitumor effects of IR therapy.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/radiotherapy , Colonic Neoplasms/immunology , Colonic Neoplasms/radiotherapy , Interferon-gamma/immunology , Adenocarcinoma/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/pathology , Cytotoxicity, Immunologic/radiation effects , Interferon-gamma/biosynthesis , Interferon-gamma/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation
18.
J Interferon Cytokine Res ; 33(6): 308-18, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23421371

ABSTRACT

It is well known that ionizing radiations induce a marked downregulation of antigen-dependent and natural immunity for a prolonged period of time. This is due, at least in part, to radiation-induced apoptosis of different lymphocyte subpopulations, including natural killer (NK) cells. Aim of this study was to investigate the capability of Beta Interferon (ß-IFN) and Interleukin-2 (IL2), alone or in combination, to restore the functional activity of the natural immune system. Mononuclear cells (MNCs) obtained from intact or in vitro irradiated human peripheral blood were treated in vitro with ß-IFN immediately before or at the end of the 4-day treatment with IL2. Time-course analysis was performed on the NK activity, the total number and the apoptotic fraction of CD16+ and CD56+ cells, the 2 main NK effector cell subpopulations. The results indicate that radiation-induced impairment of natural cytotoxicity of MNC could be successfully antagonized by the ß-IFN+IL2 combination, mainly when exposure to ß-IFN preceded IL2 treatment. This radioprotective effect is paralleled by lower levels of radiation-induced apoptosis and increased expression of the antiapoptotic Bcl-2 protein. Since natural immunity can play a significant role in antitumor host's resistance, these results could provide the rational basis for a cytokine-based pharmacological strategy able to restore immune responsiveness and to afford possible therapeutic benefits in cancer patients undergoing radiotherapy.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/radiation effects , Immunity, Innate/drug effects , Immunity, Innate/radiation effects , Interferon-beta/pharmacology , Interleukin-2/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , Apoptosis/radiation effects , CD56 Antigen/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , GPI-Linked Proteins/immunology , Gamma Rays , Humans , Immunity, Innate/immunology , Interferon-beta/immunology , Interleukin-2/immunology , K562 Cells , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/radiation effects , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/radiation effects , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Lymphocyte Subsets/radiation effects , Proto-Oncogene Proteins c-bcl-2/immunology , Receptors, IgG/immunology
19.
J Immunol ; 190(4): 1873-81, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23315072

ABSTRACT

Adoptive cell transfer (ACT) of ex vivo-activated autologous tumor-reactive T cells is currently one of the most promising approaches for cancer immunotherapy. Recent studies provided some evidence that IL-17-producing CD8(+) (Tc17) cells may exhibit potent antitumor activity, but the specific mechanisms have not been completely defined. In this study, we used a murine melanoma lung-metastasis model and tested the therapeutic effects of gp100-specific polarized type I CD8(+) cytotoxic T (Tc1) or Tc17 cells combined with autologous bone marrow transplantation after total body irradiation. Bone marrow transplantation combined with ACT of antitumor (gp100-specific) Tc17 cells significantly suppressed the growth of established melanoma, whereas Tc1 cells induced long-term tumor regression. After ACT, Tc1 cells maintained their phenotype to produce IFN-γ, but not IL-17. However, although Tc17 cells largely preserved their ability to produce IL-17, a subset secreted IFN-γ or both IFN-γ and IL-17, indicating the plasticity of Tc17 cells in vivo. Furthermore, after ACT, the Tc17 cells had a long-lived effector T cell phenotype (CD127(hi)/KLRG-1(low)) as compared with Tc1 cells. Mechanistically, Tc1 cells mediated antitumor immunity primarily through the direct effect of IFN-γ on tumor cells. In contrast, despite the fact that some Tc17 cells also secreted IFN-γ, Tc17-mediated antitumor immunity was independent of the direct effects of IFN-γ on the tumor. Nevertheless, IFN-γ played a critical role by creating a microenvironment that promoted Tc17-mediated antitumor activity. Taken together, these studies demonstrate that both Tc1 and Tc17 cells can mediate effective antitumor immunity through distinct effector mechanisms, but Tc1 cells are superior to Tc17 cells in mediating tumor regression.


Subject(s)
Adoptive Transfer/methods , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Interleukin-17/biosynthesis , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Animals , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/methods , Bone Marrow Transplantation/pathology , CD8-Positive T-Lymphocytes/radiation effects , Cytotoxicity, Immunologic/radiation effects , Interleukin-17/radiation effects , Lung Neoplasms/pathology , Lymphocyte Activation/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Whole-Body Irradiation/methods , gp100 Melanoma Antigen/biosynthesis , gp100 Melanoma Antigen/immunology
20.
Int J Radiat Biol ; 89(3): 139-46, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23078404

ABSTRACT

PURPOSE: A study of irradiated (0.25-2 Gy) murine bone marrow has investigated the relationships between apoptotic responses of cells exposed in vivo and in vitro and between in vivo apoptosis and tissue cytotoxicity. MATERIALS AND METHODS: The time course of reduction in bone marrow cellularity in vivo was determined by femoral cell counts and apoptosis measurements obtained using three commonly used assays. Inflammatory pro-apoptotic cytokine production at 24 h post-exposure in vivo was investigated using a bystander protocol. RESULTS: In vivo, there is a dose- and time-dependent non-linear reduction in bone marrow cellularity up to 24 h post- irradiation not directly represented by apoptosis measurements. The majority of cells are killed within 6 h but there is on-going cell loss in vivo up to 24 h post-irradiation in the absence of elevated levels of apoptosis and associated with the induction of cytokines produced in response to the initial tumor protein 53 (p53)-dependent apoptosis. CONCLUSION: The results demonstrate that small increases in measured apoptosis can reflect significant intramedullary cell death and with apoptotic processes being responsible for pro-inflammatory mechanisms that can contribute to additional on-going cell death. The findings demonstrate the importance of studying tissue responses when considering the mechanisms underlying the consequences of radiation exposures.


Subject(s)
Apoptosis/radiation effects , Bone Marrow Cells/pathology , Bone Marrow Cells/radiation effects , Animals , Bone Marrow Cells/immunology , Bystander Effect/radiation effects , Cytokines/biosynthesis , Cytotoxicity, Immunologic/radiation effects , Dose-Response Relationship, Radiation , Gamma Rays/adverse effects , Genes, p53 , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Signal Transduction/immunology , Signal Transduction/radiation effects , Translational Research, Biomedical
SELECTION OF CITATIONS
SEARCH DETAIL
...