Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 319
Filter
1.
BMC Immunol ; 24(1): 23, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37559025

ABSTRACT

BACKGROUND: New combinations based on standard therapeutic modalities and immunotherapy require understanding the immunomodulatory properties of traditional treatments. The objective was to evaluate the impact of brachytherapy (BT) on the immune system of cervical cancer and to identify the best modality, High-dose-rate brachytherapy (HDR-BT) vs. Pulsed-dose-rate (PDR-BT), to target it. METHODS: Nineteen patients enrolled in a prospective study received chemoradiation (CRT) and subsequently HDR-BT or PDR-BT. Peripheral blood samples were obtained for immunophenotyping analysis by flow-cytometry before CRT, BT, and two and four weeks after BT. The Friedman one-way ANOVA, Conover post hoc test, and the Wilcoxon signed-rank test were used to compare changes in cell populations at different periods, perform multiple pairwise comparisons and assess differences between treatment groups (PDR and HDR). RESULTS: Natural killer cells (NKs) were the best target for BT. Patients receiving HDR-BT achieved significantly higher values ​​and longer time of the CD56dimCD16 + NK cells with greater cytotoxic capacity than the PDR-BT group, which presented their highest elevation of CD56-CD16 + NK cells. Furthermore, both BT modalities were associated with an increase in myeloid-derived suppressor cells (MDSCs), related to a worse clinical prognosis. However, there was a decrease in the percentage of CD4 + CD25 + Foxp3 + CD45RA + regulatory T cells (Tregs) in patients receiving HDR-BT, although there were no significant differences between BT. CONCLUSIONS: Immune biomarkers are important predictive determinants in cervical cancer. Higher cytotoxic NK cells and a trend toward lower values of Tregs might support the use of HDR-BT to the detriment of PDR-BT and help develop effective combinations with immunotherapy.


Subject(s)
Brachytherapy , Uterine Cervical Neoplasms , Humans , Female , Middle Aged , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/radiotherapy , Myeloid-Derived Suppressor Cells/radiation effects , Killer Cells, Natural/radiation effects , T-Lymphocytes, Regulatory/immunology , Prospective Studies
2.
Cells ; 10(9)2021 09 17.
Article in English | MEDLINE | ID: mdl-34572108

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) in endemic regions and younger patients is characterized by a prominent lymphomononuclear infiltration. Radiation is the principal therapeutic modality for patients with NPC. Recent data suggest that the efficacy of radiotherapy in various cancers can be augmented when combined with immune checkpoint blockade. Here, we investigate the effect of radiotherapy on the killing of NPC cells by Natural Killer (NK) cells. METHODS: NPC cell lines and a patient-derived xenograft were exposed to NK cells in the context of radiotherapy. Cytotoxicity was measured using the calcein-release assay. The contribution of the PD-L1/PD-1 checkpoint and signaling pathways to killing were analyzed using specific inhibitors. RESULTS: Radiotherapy sensitized NPC cells to NK cell killing and upregulated expression of PD-1 ligand (PD-L1) in NPC cells and PD-1 receptor (PD-1) in NK cells. Blocking of the PD-L1/PD-1 checkpoint further increased the killing of NPC cells by NK cells in the context of radiotherapy. CONCLUSION: Radiation boosts the killing of NPC cells by NK cells. Killing can be further augmented by blockade of the PD-L1/PD-1 checkpoint. The combination of radiotherapy with PD-L1/PD-1 checkpoint blockade could therefore increase the efficacy of radiotherapy in NPC tumors.


Subject(s)
B7-H1 Antigen/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Killer Cells, Natural/pathology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Radiotherapy/methods , Animals , Apoptosis , Cell Proliferation , Chemoradiotherapy , Female , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/radiation effects , Mice , Mice, Nude , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/therapy , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Signal Transduct Target Ther ; 6(1): 330, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34471091

ABSTRACT

Surgery is the common treatment for early lung cancer with multiple pulmonary nodules, but it is often accompanied by the problem of significant malignancy of other nodules in non-therapeutic areas. In this study, we found that a combined treatment of local radiofrequency ablation (RFA) and melatonin (MLT) greatly improved clinical outcomes for early lung cancer patients with multiple pulmonary nodules by minimizing lung function injury and reducing the probability of malignant transformation or enlargement of nodules in non-ablated areas. Mechanically, as demonstrated in an associated mouse lung tumor model, RFA not only effectively remove treated tumors but also stimulate antitumor immunity, which could inhibit tumor growth in non-ablated areas. MLT enhanced RFA-stimulated NK activity and exerted synergistic antitumor effects with RFA. Transcriptomics and proteomics analyses of residual tumor tissues revealed enhanced oxidative phosphorylation and reduced acidification as well as hypoxia in the tumor microenvironment, which suggests reprogrammed tumor metabolism after combined treatment with RFA and MLT. Analysis of residual tumor further revealed the depressed activity of MAPK, NF-kappa B, Wnt, and Hedgehog pathways and upregulated P53 pathway in tumors, which was in line with the inhibited tumor growth. Combined RFA and MLT treatment also reversed the Warburg effect and decreased tumor malignancy. These findings thus demonstrated that combined treatment of RFA and MLT effectively inhibited the malignancy of non-ablated nodules and provided an innovative non-invasive strategy for treating early lung tumors with multiple pulmonary nodules. Trial registration: www.chictr.org.cn , identifier ChiCTR2100042695, http://www.chictr.org.cn/showproj.aspx?proj=120931 .


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Melatonin/administration & dosage , Multiple Pulmonary Nodules/drug therapy , Multiple Pulmonary Nodules/radiotherapy , Adult , Aged , Aged, 80 and over , Animals , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy , Female , Hedgehog Proteins/genetics , Heterografts , Humans , Kaplan-Meier Estimate , Killer Cells, Natural/drug effects , Killer Cells, Natural/radiation effects , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinase Kinases/genetics , Multiple Pulmonary Nodules/genetics , Multiple Pulmonary Nodules/pathology , NF-kappa B/genetics , Neoplasm, Residual/drug therapy , Neoplasm, Residual/genetics , Neoplasm, Residual/pathology , Neoplasm, Residual/radiotherapy , Progression-Free Survival , Radiofrequency Ablation/adverse effects , Treatment Outcome , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/radiation effects
4.
Int J Mol Sci ; 22(14)2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34298925

ABSTRACT

The Linear-No-Threshold (LNT) model predicts a dose-dependent linear increase in cancer risk. This has been supported by biological and epidemiological studies at high-dose exposures. However, at low-doses (LDR ≤ 0.1 Gy), the effects are more elusive and demonstrate a deviation from linearity. In this study, the effects of LDR on the development and progression of mammary cancer in FVB/N-Tg(MMTVneu)202Mul/J mice were investigated. Animals were chronically exposed to total doses of 10, 100, and 2000 mGy via tritiated drinking water, and were assessed at 3.5, 6, and 8 months of age. Results indicated an increased proportion of NK cells in various organs of LDR exposed mice. LDR significantly influenced NK and T cell function and activation, despite diminishing cell proliferation. Notably, the expression of NKG2D receptor on NK cells was dramatically reduced at 3.5 months but was upregulated at later time-points, while the expression of NKG2D ligand followed the opposite trend, with an increase at 3.5 months and a decrease thereafter. No noticeable impact was observed on mammary cancer development, as measured by tumor load. Our results demonstrated that LDR significantly influenced the proportion, proliferation, activation, and function of immune cells. Importantly, to the best of our knowledge, this is the first report demonstrating that LDR modulates the cross-talk between the NKG2D receptor and its ligands.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/radiotherapy , Immunity/radiation effects , Animals , Breast Neoplasms/metabolism , Cell Proliferation/radiation effects , Female , Killer Cells, Natural/metabolism , Killer Cells, Natural/radiation effects , Ligands , Mice , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Radiation Dosage , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Up-Regulation/radiation effects
5.
Front Immunol ; 12: 684052, 2021.
Article in English | MEDLINE | ID: mdl-34149724

ABSTRACT

Background: With increasing clinical use of NK-92 cells and their CAR-modified derivatives in cancer immunotherapy, there is a growing demand for efficient production processes of these "off-the-shelf" therapeutics. In order to ensure safety and prevent the occurrence of secondary tumors, (CAR-)NK-92 cell proliferation has to be inactivated before transfusion. This is commonly achieved by gamma irradiation. Recently, we showed proof of concept that low energy electron irradiation (LEEI) is a new method for NK-92 inactivation. LEEI has several advantages over gamma irradiation, including a faster reaction time, a more reproducible dose rate and much less requirements on radiation shielding. Here, LEEI was further evaluated as a promising alternative to gamma irradiation yielding cells with highly maintained cytotoxic effector function. Methods: Effectiveness and efficiency of LEEI and gamma irradiation were analyzed using NK-92 and CD123-directed CAR-NK-92 cells. LEE-irradiated cells were extensively characterized and compared to gamma-irradiated cells via flow cytometry, cytotoxicity assays, and comet assays, amongst others. Results: Our results show that both irradiation methods caused a progressive decrease in cell viability and are, therefore, suitable for inhibition of cell proliferation. Notably, the NK-mediated specific lysis of tumor cells was maintained at stable levels for three days post-irradiation, with a trend towards higher activities after LEEI treatment as compared to gamma irradiation. Both gamma irradiation as well as LEEI led to substantial DNA damage and an accumulation of irradiated cells in the G2/M cell cycle phases. In addition, transcriptomic analysis of irradiated cells revealed approximately 12-fold more differentially expressed genes two hours after gamma irradiation, compared to LEEI. Analysis of surface molecules revealed an irradiation-induced decrease in surface expression of CD56, but no changes in the levels of the activating receptors NKp46, NKG2D, or NKp30. Conclusions: The presented data show that LEEI inactivates (CAR-)NK-92 cells as efficiently as gamma irradiation, but with less impact on the overall gene expression. Due to logistic advantages, LEEI might provide a superior alternative for the manufacture of (CAR-)NK-92 cells for clinical application.


Subject(s)
Cell Proliferation/radiation effects , DNA Damage , Gamma Rays , Killer Cells, Natural/cytology , Killer Cells, Natural/radiation effects , Cell Line, Tumor , Cell Survival , Electrons , Flow Cytometry , Humans
6.
Front Immunol ; 12: 694086, 2021.
Article in English | MEDLINE | ID: mdl-34177957

ABSTRACT

Cells of the skin and circulation are in constant two-way communication. Following exposure of humans to sunlight or to phototherapy, there are alterations in the number, phenotype and function of circulating blood cells. In this review, only data obtained from human studies are considered, with changes induced by UV radiation (UVR) exposure described for phagocytic leukocytes and peripheral blood mononuclear cells plus their component T and B cells, natural killer cells and dendritic cells. These immune modulations illustrate the potential of UVR to have therapeutic effects beyond the skin, and that sunlight exposure is an important environmental influence on human health.


Subject(s)
Dendritic Cells/radiation effects , Leukocytes/radiation effects , Phototherapy/adverse effects , Radiation Exposure/adverse effects , Sunlight/adverse effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/radiation effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/radiation effects , Leukocytes/immunology , Leukocytes/metabolism , Seasons , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Ultraviolet Rays/adverse effects , Ultraviolet Therapy/adverse effects
7.
Clin Cancer Res ; 26(20): 5368-5379, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32873573

ABSTRACT

PURPOSE: Non-small cell lung cancer (NSCLC) is a fatal disease with poor prognosis. A membrane-bound form of Hsp70 (mHsp70) which is selectively expressed on high-risk tumors serves as a target for mHsp70-targeting natural killer (NK) cells. Patients with advanced mHsp70-positive NSCLC may therefore benefit from a therapeutic intervention involving mHsp70-targeting NK cells. The randomized phase II clinical trial (EudraCT2008-002130-30) explores tolerability and efficacy of ex vivo-activated NK cells in patients with NSCLC after radiochemotherapy (RCT). PATIENTS AND METHODS: Patients with unresectable, mHsp70-positive NSCLC (stage IIIa/b) received 4 cycles of autologous NK cells activated ex vivo with TKD/IL2 [interventional arm (INT)] after RCT (60-70 Gy, platinum-based chemotherapy) or RCT alone [control arm (CTRL)]. The primary objective was progression-free survival (PFS), and secondary objectives were the assessment of quality of life (QoL, QLQ-LC13), toxicity, and immunobiological responses. RESULTS: The NK-cell therapy after RCT was well tolerated, and no differences in QoL parameters between the two study arms were detected. Estimated 1-year probabilities for PFS were 67% [95% confidence interval (CI), 19%-90%] for the INT arm and 33% (95% CI, 5%-68%) for the CTRL arm (P = 0.36, 1-sided log-rank test). Clinical responses in the INT group were associated with an increase in the prevalence of activated NK cells in their peripheral blood. CONCLUSIONS: Ex vivo TKD/IL2-activated, autologous NK cells are well tolerated and deliver positive clinical responses in patients with advanced NSCLC after RCT.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Chemoradiotherapy , HSP70 Heat-Shock Proteins/blood , Platinum/administration & dosage , Adult , Aged , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/pathology , Combined Modality Therapy , Female , Humans , Immunotherapy, Adoptive/adverse effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/radiation effects , Male , Middle Aged , Neoplasm Staging , Platinum/adverse effects , Progression-Free Survival
8.
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
9.
Int J Radiat Oncol Biol Phys ; 108(1): 70-80, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32544576

ABSTRACT

PURPOSE: Improved antitumor responses have been observed in patients after combination radiation therapy (RT) and immune checkpoint blockade (ICB). Whether these clinical responses are linked to the host systemic immune system has not been elucidated. METHODS AND MATERIALS: In this single-institution prospective observational study, peripheral blood was longitudinally collected from 10 patients with metastatic disease who had responded to anti-PD-1/anti-PD-L1 ICB and received RT (8-50 Gy in 1-5 fractions) upon disease progression at the following timepoints: baseline (pre-RT), 1 to 2 weeks post-RT, and post-ICB (cycle 1) on reintroduction post-RT. To thoroughly characterize the interaction between combined RT-ICB and the host immune system, we performed high-dimensional, mass cytometry-based immunophenotyping of circulating lymphocytes using a 40-marker panel addressing lineage, differentiation, activation, trafficking, cytotoxicity, and costimulatory and inhibitory functions. Phenotypic expression of circulating lymphocytes was compared across patients and time points and correlated with post-RT tumor responses. RESULTS: Foremost, we demonstrated excellent posttreatment clinical responses, including 4 local responses with >50% reduction in radiated tumor size, 1 out-of-field response, and 4 patients who resumed ICB for >1 year. Baseline and post-RT immune states were highly heterogeneous among patients. Despite this interindividual heterogeneity in baseline immune states, we observed a systemic immune reaction to RT-ICB common across patients, histology, and radiation sites; a subset of pre-existing Ki-67+ CD8+ T cells were increased post-RT and further expanded upon reintroduction of ICB post-RT (2.3-fold increase, P = .02). Importantly, RT did not alter the phenotypic profile of these Ki-67+ CD8+ T cells, which was characterized by a distinct activated and differentiated effector phenotype. CONCLUSIONS: Collectively, these findings point toward a sustained reinvigoration of host antitumor immunity after RT-ICB and suggest an expansion in activated Ki-67+ CD8+ T cells as a possible demonstration of this synergy, thereby providing new insights that may support the development of optimal sequencing strategies.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Radiotherapy , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cell Survival/immunology , Cell Survival/radiation effects , Combined Modality Therapy , Humans , Immunophenotyping , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/radiation effects , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/radiation effects
10.
J Immunol Res ; 2020: 8459496, 2020.
Article in English | MEDLINE | ID: mdl-32411806

ABSTRACT

NK cells are lymphocytes with antitumor properties and can directly lyse tumor cells in a non-MHC-restricted manner. However, the tumor microenvironment affects the immune function of NK cells, which leads to immune evasion. This may be related to the pathogenesis of some diseases. Therefore, great efforts have been made to improve the immunotherapy effect of natural killer cells. NK cells from different sources can meet different clinical needs, in order to minimize the inhibition of NK cells and maximize the response potential of NK cells, for example, modification of NK cells can increase the number of NK cells in tumor target area, change the direction of NK cells, and improve their targeting ability to malignant cells. Checkpoint blocking is also a promising strategy for NK cells to kill tumor cells. Combination therapy is another strategy for improving antitumor ability, especially in combination with oncolytic viruses and nanomaterials. In this paper, the mechanisms affecting the activity of NK cells were reviewed, and the therapeutic potential of different basic NK cell strategies in tumor therapy was focused on. The main strategies for improving the immune function of NK cells were described, and some new strategies were proposed.


Subject(s)
Immunotherapy/methods , Killer Cells, Natural/transplantation , Neoplasms/therapy , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/administration & dosage , Combined Modality Therapy/methods , Disease Models, Animal , Drug Delivery Systems/methods , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Evasion/drug effects , Immunologic Memory , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/radiation effects , Magnetic Field Therapy , Mice , Nanomedicine/methods , Nanoparticles/administration & dosage , Neoplasms/immunology , Oncolytic Viruses/immunology , Receptors, Chimeric Antigen/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
11.
Clin Cancer Res ; 26(4): 945-956, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31694834

ABSTRACT

PURPOSE: There is growing interest in combinations of immunogenic radiotherapy (RT) and immune checkpoint blockade, but clinical responses are still limited. Therefore, we tested the triple therapy with an inhibitor of the indoleamine 2,3-dioxygenase pathway, which like immune checkpoints, downregulates the antitumor immune response. EXPERIMENTAL DESIGN: Triple treatment with hypofractionated RT (hRT) + anti-PD-1 antibody (αPD1) + indoximod was compared with the respective mono- and dual therapies in two syngeneic mouse models. RESULTS: The tumors did not regress following treatment with hRT + αPD1. The αPD1/indoximod combination was not effective at all. In contrast, triple treatment induced rapid, marked tumor regression, even in mice with a large tumor. The effects strongly depended on CD8+ T cells and partly on natural killer (NK) cells. Numbers and functionality of tumor-specific CD8+ T cells and NK cells were increased, particularly early during treatment. However, after 2.5-3 weeks, all large tumors relapsed, which was accompanied by increased apoptosis of tumor-infiltrating lymphocytes associated with a non-reprogrammable state of exhaustion, terminal differentiation, and increased activation-induced cell death, which could not be prevented by indoximod in these aggressive tumor models. Some mice with a smaller tumor were cured. Reirradiation during late regression (day 12), but not after relapse, cured almost all mice with a large B16-CD133 tumor, and strongly delayed relapse in the less immunogenic 4T1 model, depending on CD8+ T cells. CONCLUSIONS: Our findings may serve as a rationale for the clinical evaluation of this triple-combination therapy in patients with solitary or oligometastatic tumors in the neoadjuvant or the definitive setting.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/radiotherapy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/radiotherapy , Tryptophan/analogs & derivatives , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/radiation effects , Cell Line, Tumor , Chemoradiotherapy , Female , Killer Cells, Natural/drug effects , Killer Cells, Natural/radiation effects , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Radiation Dose Hypofractionation , Survival Rate , Tryptophan/pharmacology
12.
Sci Rep ; 9(1): 7980, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138860

ABSTRACT

Clinically isolated syndrome (CIS) is the earliest clinical episode in multiple sclerosis (MS). Low environmental exposure to UV radiation is implicated in risk of developing MS, and therefore, narrowband UVB phototherapy might delay progression to MS in people with CIS. Twenty individuals with CIS were recruited, and half were randomised to receive 24 sessions of narrowband UVB phototherapy over a period of 8 weeks. Here, the effects of narrowband UVB phototherapy on the frequencies of circulating immune cells and immunoglobulin levels after phototherapy are reported. Peripheral blood samples for all participants were collected at baseline, and 1, 2, 3, 6 and 12 months after enrolment. An extensive panel of leukocyte populations, including subsets of T cells, B cells, monocytes, dendritic cells, and natural killer cells were examined in phototherapy-treated and control participants, and immunoglobulin levels measured in serum. There were significant short-term increases in the frequency of naïve B cells, intermediate monocytes, and fraction III FoxP3+ T regulatory cells, and decreases in switched memory B cells and classical monocytes in phototherapy-treated individuals. Since B cells are increasingly targeted by MS therapies, the effects of narrowband UVB phototherapy in people with MS should be investigated further.


Subject(s)
B-Lymphocyte Subsets/radiation effects , Demyelinating Diseases/therapy , Dendritic Cells/radiation effects , Killer Cells, Natural/radiation effects , Monocytes/radiation effects , T-Lymphocyte Subsets/radiation effects , Adult , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Calcifediol/blood , Demyelinating Diseases/complications , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Progression , Female , Flow Cytometry , Humans , Immunoglobulins/blood , Immunologic Memory/radiation effects , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Longitudinal Studies , Male , Middle Aged , Monocytes/immunology , Monocytes/pathology , Multiple Sclerosis/etiology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Multiple Sclerosis/prevention & control , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Ultraviolet Rays , Ultraviolet Therapy/methods
13.
Am J Transplant ; 19(2): 591-596, 2019 02.
Article in English | MEDLINE | ID: mdl-30346652

ABSTRACT

Resistance to parental bone marrow (BM) grafts in F1 hybrid recipients is due to natural killer (NK) cell-mediated rejection triggered through "missing self" recognition. "Hybrid resistance" has usually been investigated in lethally irradiated F1 recipients in conjunction with pharmacological activation of NK cells. Here, we investigated BM-directed NK-cell alloreactivity in settings of reduced conditioning. Nonlethally irradiated (1-3 Gy) or nonirradiated F1 (C57BL6 × BALB/c) recipient mice received titrated doses (5-20 x 106 ) of unseparated parental BALB/c BM without pharmacological NK cell activation. BM successfully engrafted in all mice and multilineage donor chimerism persisted long-term (24 weeks), even in the absence of irradiation. Chimerism was associated with the rearrangement of the NK-cell receptor repertoire suggestive of reduced reactivity to BALB/c. Chimerism levels were lower after transplantation with parental BALB/c than with syngeneic F1 BM, indicating partial NK-mediated rejection of parental BM. Activation of NK cells with polyinosinic-polycytidylic acid sodium salt poly(I:C), reduced parental chimerism in nonirradiated BM recipients but did not prevent hematopoietic stem cell engraftment. In contrast, equal numbers of parental lymph node cells were completely rejected. Hence, hybrid resistance leads to incomplete rejection of parental BM under reduced conditioning settings.


Subject(s)
Bone Marrow Transplantation/methods , Bone Marrow/immunology , Graft Rejection/immunology , Immune Tolerance/immunology , Killer Cells, Natural/immunology , Transplantation Chimera/immunology , Animals , Bone Marrow/radiation effects , Female , Killer Cells, Natural/radiation effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
14.
Radiat Prot Dosimetry ; 186(2-3): 181-185, 2019 Dec 31.
Article in English | MEDLINE | ID: mdl-31943099

ABSTRACT

The aim of the present study was to evaluate the biodosimetric potential of peripheral blood lymphocytes, particularly of T-cell subsets (null and T helper) and natural killer cells (NK), upon exposure to gamma irradiation (60Co) in vivo. For this purpose, the change in relative numbers of NK cells and T-lymphocyte subsets, as well as in the H2AX phosphorylation rate, were evaluated as potential early markers of the lymphocytic response to irradiation in vivo. These experiments were performed on a Large White Pig model. As a result, significant but not dose-dependent changes in the proportion of lymphocyte subpopulations (NK cells, null and T helper cells) were found after exposure to ionising radiation in vivo. On the other hand, circulating NK cells showed relatively higher radioresistance capacity when compared to the T-lymphocyte subsets; however, gamma-H2AX expression showed no significant difference between the evaluated lymphocyte subsets.


Subject(s)
Killer Cells, Natural/radiation effects , Radiometry/methods , T-Lymphocyte Subsets/radiation effects , Animals , Cobalt Radioisotopes/pharmacology , DNA Damage , Gamma Rays , Histones/metabolism , Immunophenotyping , Lymphocytes/cytology , Phenotype , Phosphorylation , Radiation, Ionizing , Swine
15.
Int J Mol Sci ; 19(11)2018 Nov 13.
Article in English | MEDLINE | ID: mdl-30428512

ABSTRACT

In cancer treatments, especially high-dose radiotherapy (HDRT) is applied. Patients suffering from chronic inflammatory diseases benefit from low-dose radiation therapy (LDRT), but exposure to very low radiation doses can still steadily increase for diagnostic purposes. Yet, little is known about how radiation impacts on forms of cell death in human immune cells. In this study, the radiosensitivity of human immune cells of the peripheral blood was examined in a dose range from 0.01 to 60 Gy with regard to induction of apoptosis, primary necrosis, and secondary necrosis. Results showed that immune cells differed in their radiosensitivity, with monocytes being the most radioresistant. T cells mainly died by necrosis and were moderately radiosensitive. This was followed by B and natural killer (NK) cells, which died mainly by apoptosis. X-radiation had no impact on cell death in immune cells at very low doses (≤0.1 Gy). Radiation doses of LDRT (0.3⁻0.7 Gy) impacted on the more radiosensitive NK and B cells, which might contribute to attenuation of inflammation. Even single doses applied during RT of tumors did not erase the immune cells completely. These in vitro studies can be considered as the basis to optimize individual radiation therapy schemes in multimodal settings and to define suited time points for further inclusion of immunotherapies.


Subject(s)
Adaptive Immunity/radiation effects , Immunity, Innate/radiation effects , Apoptosis/radiation effects , B-Lymphocytes/radiation effects , Cell Death/radiation effects , Dose-Response Relationship, Radiation , Humans , Killer Cells, Natural/radiation effects , Monocytes/radiation effects , Radiation Exposure/adverse effects , Radiotherapy
16.
Medicine (Baltimore) ; 97(42): e12866, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30334999

ABSTRACT

RATIONALE: Hepatocellular carcinomas (HCCs) with metastases to the right atrium (RA) and lungs are rare, with a poor prognosis. Furthermore, the treatment outcomes in patients with advanced HCCs remain unsatisfactory. PATIENT CONCERNS: A 46-year-old man presented to our hospital for dyspnea on exertion and abdominal pain. DIAGNOSES: HCC and extra-hepatic metastases to the lung and RA. INTERVENTIONS: Multidisciplinary treatment including radiotherapy (RT), transarterial chemoembolization (TACE), and sorafenib. During a follow-up evaluation computed tomography, he experienced a radio-contrast-induced anaphylaxis. After the event, treatment such as RT, TACE, and sorafenib were continued. OUTCOMES: His tumor burden decreased, finally leading to a complete response as per the modified Response Evaluation Criteria in Solid Tumors. The patient is still alive, 30 months after the episode. Subsequent blood tests showed increased natural killer (NK) cell activity, which was significantly higher than that seen in other age-matched HCC patients with an identical stage of the tumor, receiving sorafenib. This suggests that the increase in NK cells induced by anaphylaxis influenced the tumor burden. LESSONS: We report here a rare case of long-term survival of an HCC patient with multiple metastases treated with multidisciplinary modalities, in which high NK cell activity was observed after a radio-contrast-induced anaphylactic reaction during follow-up investigations.


Subject(s)
Antineoplastic Protocols , Carcinoma, Hepatocellular/therapy , Heart Neoplasms/therapy , Liver Neoplasms/therapy , Lung Neoplasms/therapy , Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/secondary , Chemoembolization, Therapeutic/methods , Combined Modality Therapy/methods , Heart Atria/pathology , Heart Neoplasms/pathology , Heart Neoplasms/secondary , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/radiation effects , Liver Neoplasms/pathology , Lung/pathology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Middle Aged , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Radiotherapy, Adjuvant/methods , Remission Induction/methods , Response Evaluation Criteria in Solid Tumors , Sorafenib , Tumor Burden
17.
Anticancer Res ; 38(10): 5767-5772, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30275198

ABSTRACT

BACKGROUND/AIM: γ-Irradiation has been proven to be the most effective method to inactivate K562 cells, but γ-irradiators are not available in some institutes. This study was designed to compare the effects of X-ray and γ-irradiation on K562 cells in natural killer (NK) cell expansion. MATERIALS AND METHODS: To expand NK cells, isolated peripheral blood mononuclear cells (PBMCs) were co-cultured with γ-irradiated or X-ray-treated K562 cells plus IL-2 and IL-15. Characteristics of expanded NK cells were identified by flow cytometry. RESULTS: NK cell expansion rate tended be to lower in the X-ray-treated group (68.9±32.6) than the γ-irradiated group (78±28.7), but the difference was not significant (p=0.39). Furthermore, NK cell functions or receptor expression were similar in the two groups. CONCLUSION: Our results suggest that X-ray treatment can be used as an alternative to γ-irradiation for K562 cells inactivation in human NK cell expansion.


Subject(s)
Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Feeder Cells/cytology , Killer Cells, Natural/cytology , Coculture Techniques , Feeder Cells/radiation effects , Humans , K562 Cells , Killer Cells, Natural/radiation effects , X-Rays
18.
PLoS One ; 13(6): e0198904, 2018.
Article in English | MEDLINE | ID: mdl-29897952

ABSTRACT

Hepatic irradiation for the treatment of hepatobiliary malignancies often indirectly damages liver tissue and promotes the development of liver fibrosis. However, little is known concerning the effects of hepatic irradiation on the liver immune system, including natural killer (NK) cells. The aim of this study was therefore to investigate how hepatic irradiation influences the functions and characteristics of liver resident NK cells. An established murine hepatic irradiation model was used to examine the specific effects of hepatic irradiation on immune cell populations and metastasis. This analysis demonstrated that hepatic irradiation decreased the number of liver resident NK cells (DX5-TRAIL+), but did not affect the total NK number or proportions of NK cells in the liver or spleen. This effect was correlated with the hepatic irradiation dose. Surprisingly, the liver resident NK population had not recovered by two months after hepatic irradiation. We also found that hepatic irradiation limited the cytotoxic effects of liver-derived lymphocytes against a mouse hepatoma cell line and promoted hepatic metastases in an in vivo model, although adoptive transfer of activated NK cells could alleviate metastatic growth. Finally, we demonstrated that hepatic irradiation disrupted the development of liver-resident NK cells, even after the adoptive transfer of precursor cells from the bone marrow, liver, and spleen, suggesting that irradiation had altered the developmental environment of the liver. In summary, our data demonstrated that hepatic irradiation abolished the DX5-TRAIL+ liver-resident NK cell population and dampened antitumor activities in the liver for at least two months. Additionally, hepatic irradiation prevented differentiation of precursor cells into liver-resident NK cells.


Subject(s)
Gamma Rays , Killer Cells, Natural/cytology , Liver/radiation effects , Adoptive Transfer , Animals , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/radiation effects , Liver/cytology , Liver/pathology , Lymphocytes/cytology , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Spleen/cytology , Spleen/immunology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transplantation, Homologous
19.
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
20.
J Radiat Res ; 59(3): 245-252, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29518205

ABSTRACT

Bortezomib, which is a potent proteasome inhibitor, has been used as a first-line drugs to treat multiple myeloma for a few decades, and radiotherapy has frequently been applied to manage acute bone lesions in the patients. Therefore, it was necessary to investigate what the benefits might be if the two therapies were applied simultaneously in the treatment of multiple myeloma. Since it was known that radiotherapy and proteasome inhibitors could increase the expression of NKG2D ligands through induction of protein synthesis and suppression of protein degradation of NKG2D ligands, respectively, we supposed that the combined treatment might further enhance the expression of NKG2D ligands. In this study, we analyzed the expression level of NKG2D ligands using multiplex PCR and flow cytometry after treatment of IM-9 and RPMI-8226 myeloma cells with bortezomib and ionizing radiation; we then assayed the susceptibility to NK-92 cells. Although the expression of only some kinds of NKG2D ligands were increased by treatment with bortezomib alone, five kinds of NKG2D ligands that we assayed were further induced at the surface protein level after combined treatment with ionizing radiation and bortezomib. Furthermore, combined treatment made myeloma cells more susceptible to NK-92 cells, compared with treatment with bortezomib alone. In conclusion, the combination therapy of ionizing radiation plus the proteasome inhibitor bortezomib is a promising therapeutical strategy for enhancing NK cell-mediated anticancer immune responses.


Subject(s)
Bortezomib/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/radiotherapy , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Proteasome Inhibitors/therapeutic use , Radiation, Ionizing , Bortezomib/pharmacology , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Killer Cells, Natural/radiation effects , Ligands , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Proteasome Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL