Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncol Lett ; 25(6): 232, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37153058

RESUMO

Natural killer (NK) cells play a crucial role in early immune defenses against transformed cells and are used in the therapeutic management of cancer. However, it is difficult to sufficiently obtain high purity activated NK cells for clinical application. The function of NK cells is dependent on the balance of activating and inhibitory signals. Strong and diverse stimuli are required to increase the function of NK cells. Radiotherapy modulates the expression of various immunomodulatory molecules that recruit and activate NK cells. NK cell-mediated antibody-dependent cellular cytotoxicity is one of the most potent cytotoxic effects of NK cells against target cancer cells. To generate activated and irradiated autologous peripheral blood mononuclear cells (PBMCs), cytokine and monoclonal antibody stimulation followed by ionizing radiation was performed in the present study. The expanded NK cells were cultured for 21 days using activated/irradiated autologous PBMCs. Colorectal cancer cells (SW480 and HT-29) were used to analyze the expression of NK group 2D ligands and EGFR by radiation. The cytotoxicity of radiation plus NK cell-based targeted therapy against colorectal cancer cell lines was analyzed using flow cytometry. Activated and irradiated PBMCs exhibited significantly increased expression of various activating ligands that stimulated NK cells. In total, >10,000-fold high-purity activated NK cells were obtained, with negligible T-cell contamination. To confirm the antitumor activity of the NK cells expanded by this method, the expanded NK cells were treated with cetuximab, radiotherapy, or a combination of cetuximab and radiotherapy in the presence of human colorectal cancer cells. Expanded NK cells were effective at targeting human colorectal cancer cells, particularly when combined with cetuximab and radiotherapy. Thus, in the present study, a novel method for high-purity activated NK cell expansion was developed using activated and irradiated PBMCs. In addition, combined radiotherapy and antibody-based immunotherapy with expanded NK cells may be an effective strategy to enhance the efficiency of treatment against colorectal cancer.

2.
Sci Rep ; 13(1): 7656, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37169953

RESUMO

Pancreatic cancer is difficult to diagnose at the initial stage and is often discovered after metastasis to nearby organs. Gemcitabine is currently used as a standard treatment for pancreatic cancer. However, since chemotherapy for pancreatic cancer has not yet reached satisfactory therapeutic results, adjuvant chemotherapy methods are attempted. It can be expected that combining immune cell therapy with existing anticancer drug combination treatment will prevent cancer recurrence and increase survival rates. We isolated natural killer (NK) cells and co-cultured them with strongly activated autologous peripheral blood mononuclear cells (PBMCs) as feeder cells, activated using CD3 antibody, IFN-r, IL-2, and γ-radiation. NK cells expanded in this method showed greater cytotoxicity than resting NK cells, when co-cultured with pancreatic cancer cell lines. Tumor growth was effectively inhibited in a pancreatic cancer mouse xenograft model. Therapeutic efficacy was increased by using gemcitabine and erlotinib in combination. These findings suggest that NK cells cultured by the method proposed here have excellent anti-tumor activity. We demonstrate that activated NK cells can efficiently inhibit pancreatic tumors when used in combination with gemcitabine-based therapy.


Assuntos
Gencitabina , Neoplasias Pancreáticas , Humanos , Animais , Camundongos , Leucócitos Mononucleares , Recidiva Local de Neoplasia/metabolismo , Células Matadoras Naturais , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/metabolismo , Imunoterapia/métodos , Linhagem Celular Tumoral , Neoplasias Pancreáticas
3.
Molecules ; 26(13)2021 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-34203519

RESUMO

Histone acetylation is an epigenetic mechanism that regulates the expression of various genes, such as natural killer group 2, member D (NKG2D) ligands. These NKG2D ligands are the key molecules that activate immune cells expressing the NKG2D receptor. It has been observed that cancer cells overexpress histone deacetylases (HDACs) and show reduced acetylation of nuclear histones. Furthermore, HDAC inhibitors are known to upregulate the expression of NKG2D ligands. Humans have 18 known HDAC enzymes that are divided into four classes. At present, it is not clear which types of HDAC are involved in the expression of NKG2D ligands. We hypothesized that specific types of HDAC genes might be responsible for altering the expression of NKG2D ligands. In this study, we monitored the expression of NKG2D ligands and major histocompatibility complex (MHC) class I molecules in lung cancer cells which were treated with six selective HDAC inhibitors and specific small interfering RNAs (siRNAs). We observed that treatment with FK228, which is a selective HDAC1/2 inhibitor, also known as Romidepsin, induced NKG2D ligand expression at the transcriptional and proteomic levels in two different lung cancer cell lines. It also caused an increase in the susceptibility of NCI-H23 cells to NK cells. Silencing HDAC1 or HDAC2 using specific siRNAs increased NKG2D ligand expression. In conclusion, it appears that HDAC1 and HDAC2 might be the key molecules regulating the expression of NKG2D ligands. These results imply that specifically inhibiting HDAC1 and HDAC2 could induce the expression of NKG2D ligands and improve the NK cell-mediated anti-cancer immunity.


Assuntos
Regulação Neoplásica da Expressão Gênica/imunologia , Histona Desacetilase 1/imunologia , Histona Desacetilase 2/imunologia , Imunidade Celular/imunologia , Neoplasias Pulmonares/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Proteínas de Neoplasias/imunologia , Células A549 , Histona Desacetilase 1/genética , Histona Desacetilase 2/genética , Humanos , Células Matadoras Naturais , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Proteínas de Neoplasias/genética
4.
BMC Immunol ; 22(1): 44, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34253166

RESUMO

BACKGROUND: Transforming growth factor beta (TGF-ß) is a typical immuno-inhibitory cytokine and highly secreted by lung cancer cells. It was supposed that its immunosuppressive effects to NK cell might be related with the altered expression of activating and inhibitory molecules in lung cancer cells. In this study, we examined the expression of NKG2DLs, PD-L1 and PD-L2 in lung cancer cells after treatment of TGF-ß and a TGF-ß inhibitor, Galunisertib (LY2157299). RESULTS: TGF-ß reduced the level of surface proteins of five NKG2DLs without altered transcription levels in lung cancer cells. Galunisertib reversed the effect of TGF-ß on the expression of NKG2DLs. Since MMP inhibitors, MMPi III and MMP2 inhibitor I, restored the reduced expression of NKG2DLs after treatment of TGF-ß, it was thought that TGF-ß induced the expression of MMP2 which facilitated the shedding of the NKG2DLs in cancer cells. However, the expression of PD-L1, L2 were not changed by treatment with TGF-ß or Galunisertib. CONCLUSIONS: Therefore, inhibition of TGF-ß might reverse the immunosuppressive status on immune cells and restore NK cell mediated anticancer immune responses by upregulation of NKG2DLs in cancer cells.


Assuntos
Adenocarcinoma Bronquioloalveolar/imunologia , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/imunologia , Fator de Crescimento Transformador beta/metabolismo , Células A549 , Adenocarcinoma Bronquioloalveolar/tratamento farmacológico , Citotoxicidade Imunológica , Regulação para Baixo , Proteínas Ligadas por GPI/metabolismo , Humanos , Tolerância Imunológica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Pirazóis/farmacologia , Quinolinas/farmacologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Evasão Tumoral
5.
PLoS One ; 16(4): e0248870, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33793576

RESUMO

Since ionizing radiation has showed the dramatic effect to kill the cancer cells through direct DNA damage as well as triggering anti-cancer immune responses including induction of NKG2D ligands, it has used for long time to treat many cancer patients. However, it has been known that radiotherapy might promote the remnant cancer cells to escape immune system and metastasis. One of the suggested ways of immune evasion is induction of a ligand for programmed death-1 (PD-L1) in head and neck cancer, bladder cancer and lung cancer cells which engages the receptor, programmed death-1 (PD-1) in immune cells. PD-1/PD-L1 axis transduces the inhibitory signal and suppresses the adaptive immunity. However, their role in innate immunity remains poorly understood. Therefore, we investigated whether ionizing radiation could change the expression of PD-L1 in malignant melanoma cells and the receptor, programmed death-1 (PD-1), in NK-92 cells. Surface PD-L1 levels on melanoma cells were increased by ionizing radiation in a dose-independent manner but the level of PD-L1 was not changed significantly in NK-92 cells. Radiation-induced PD-L1 suppressed the activity of the NK-92 cells against melanoma cells despite of upregulation of NKG2D ligands. Furthermore, activated NK cells had high level of PD-1 and could not kill PD-L1+ melanoma cells effectively. When we used PD-L1 inhibitor or silenced PD-L1 gene, inhibited PD-1/PD-L1 axis reversed the activity of the suppressed NK cells. Through these results, we supposed that PD-1/PD-L1 blockade could enhance the immune responses of NK cells against melanoma cells after radiotherapy and might overcome the PD-L1 mediated radioresistance of cancer cells.


Assuntos
Antígeno B7-H1/metabolismo , Melanoma , Receptor de Morte Celular Programada 1/metabolismo , Linhagem Celular Tumoral , Humanos , Imunidade Celular , Células Matadoras Naturais , Melanoma/imunologia , Melanoma/radioterapia , Tolerância a Radiação
6.
Mol Med Rep ; 20(4): 3301-3307, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31432134

RESUMO

c­Myc is a characteristic oncogene with dual functions in cell proliferation and apoptosis. Since the overexpression of the c­Myc proto­oncogene is a common event in the development and growth of various human types of cancer, the present study investigated whether oncogenic c­Myc can alter natural killer (NK) cell­mediated immunity through the expression of associated genes, using PCR, western blotting and flow cytometry assays. Furthermore, whether c­Myc could influence the expression levels of natural killer group 2 member D (NKG2D) ligands, which are well known NK activation molecules, as well as NK cell­mediated immunity, was investigated. c­Myc was inhibited by 10058­F4 treatment and small interfering RNA transfection. Upregulation of c­Myc was achieved by transfection with a pCMV6­myc vector. The inhibition of c­Myc increased MHC class I polyeptide­related sequence B and UL16 binding protein 1 expressions among NKG2D ligands, and the overexpression of c­Myc suppressed the expression of all NKG2D ligands, except MHC class I polyeptide­related sequence A. Furthermore, the alteration of c­Myc activity altered the susceptibility of K562 cells to NK cells. These results suggested that the overexpression of c­Myc may contribute to the immune escape of cancer cells and cell proliferation. Combined treatment with NK­based cancer immunotherapy and inhibition of c­Myc may achieve improved therapeutic results.


Assuntos
Regulação Leucêmica da Expressão Gênica/imunologia , Imunidade Celular , Células Matadoras Naturais/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Proteínas Proto-Oncogênicas c-myc/imunologia , Evasão Tumoral , Regulação para Cima/imunologia , Humanos , Células K562 , Células Matadoras Naturais/patologia
7.
Cancer Res Treat ; 51(2): 464-473, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29879758

RESUMO

PURPOSE: The purpose of this study was to investigate the efficacy of stereotactic body radiation therapy (SBRT) as a tumor-associated antigen (TAA) presentation method for dendritic cell (DC) sensitization and evaluate its effect in combination with immunotherapy using an intratumoral injection of immature DCs (iDCs). METHODS AND MATERIALS: CT-26 colon carcinoma cell was used as a cancer cell line. Annexin V staining and phagocytosis assays were performed to determine the appropriate radiation dose and incubation time to generate TAAs. BALB/c mice were used for in vivo experiments. Cancer cells were injected into the right legs and left flanks to generate primary and metastatic tumors, respectively. The mice were subjected to radiation therapy (RT) alone, intradermal injection of electroporated DCs alone, or RT in combination with iDC intratumoral injection (RT/iDC). Tumor growth measurement and survival rate analysis were performed. Enzyme-linked immunospot and cytotoxicity assays were performed to observe the effect of different treatments on the immune system. RESULTS: Annexin V staining and phagocytosis assays showed that 15 Gy radiation dose and 48 hours of incubation was appropriate for subsequent experiments. Maximum DC sensitization and T-cell stimulation was observed with RT as compared to other TAA preparation methods. In vivo assays revealed statistically significant delay in the growth of both primary and metastatic tumors in the RT/iDC group. The overall survival rate was the highest in the RT/iDC group. CONCLUSION: The combination of SBRT and iDC vaccination may enhance treatment effects. Clinical trials and further studies are warranted in the future.


Assuntos
Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Radiocirurgia , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Linhagem Celular Tumoral , Terapia Combinada , Citocinas/metabolismo , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Humanos , Imunoterapia , Estimativa de Kaplan-Meier , Camundongos , Neoplasias/mortalidade , Neoplasias/patologia , Radiocirurgia/métodos , Carga Tumoral
8.
PLoS One ; 13(1): e0190638, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29298343

RESUMO

Cancer stem-like cells (CSCs) may play a key role in tumor initiation, self-renewal, differentiation, and resistance to current treatments. Dendritic cells (DCs) play a vital role in host immune reactions as well as antigen presentation. In this study, we explored the suitability of using CSC peptides as antigen sources for DC vaccination against human breast cancer and hepatocellular carcinoma (HCC) with the aim of achieving CSC targeting and enhancing anti-tumor immunity. CD44 is used as a CSC marker for breast cancer and EpCAM is used as a CSC marker for HCC. We selected CD44 and EpCAM peptides that bind to HLA-A2 molecules on the basis of their binding affinity, as determined by a peptide-T2 binding assay. Our data showed that CSCs express high levels of tumor-associated antigens (TAAs) as well as major histocompatibility complex (MHC) molecules. Pulsing DCs with CD44 and EpCAM peptides resulted in the efficient generation of mature DCs (mDCs), thus enhancing T cell stimulation and generating potent cytotoxic T lymphocytes (CTLs). The activation of CSC peptide-specific immune responses by the DC vaccine in combination with standard chemotherapy may provide better clinical outcomes in advanced carcinomas.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma Hepatocelular/terapia , Células Dendríticas/imunologia , Molécula de Adesão da Célula Epitelial/administração & dosagem , Neoplasias Hepáticas/terapia , Fragmentos de Peptídeos/administração & dosagem , Animais , Vacinas Anticâncer/imunologia , Carcinoma Hepatocelular/imunologia , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Molécula de Adesão da Célula Epitelial/química , Feminino , Antígeno HLA-A2/imunologia , Xenoenxertos , Humanos , Receptores de Hialuronatos/imunologia , Neoplasias Hepáticas/imunologia , Camundongos , Camundongos Endogâmicos BALB C
9.
Sci Rep ; 7(1): 11075, 2017 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-28894091

RESUMO

Natural killer (NK) cells are considered a promising strategy for cancer treatment. Various methods for large-scale NK cell expansion have been developed, but they should guarantee that no viable cells are mixed with the expanded NK cells because most methods involve cancer cells or genetically modified cells as feeder cells. We used an anti-CD16 monoclonal antibody (mAb) and irradiated autologous peripheral blood mononuclear cells (PBMCs) (IrAPs) to provide a suitable environment (activating receptor-ligand interactions) for the NK cell expansion. This method more potently expanded NK cells, and the final product was composed of highly purified NK cells with lesser T-cell contamination. The expanded NK cells showed greater upregulation of various activation receptors, CD107a, and secreted larger amounts of interferon gamma. IrAPs expressed NKG2D ligands and CD48, and coengagement of CD16 with NKG2D and 2B4 caused potent NK cell activation and proliferation. The expanded NK cells were cytotoxic toward various cancer cells in vitro and in vivo. Moreover, irradiation or a chemotherapeutic drug further enhanced this antitumor effect. Therefore, we developed an effective in vitro culture method for large-scale expansion of highly purified cytotoxic NK cells with potent antitumor activity using IrAPs instead of cancer cell-based feeder cells.


Assuntos
Anticorpos Monoclonais/farmacologia , Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Receptores de IgG/antagonistas & inibidores , Animais , Biomarcadores , Antígeno CD48/metabolismo , Degranulação Celular/efeitos dos fármacos , Degranulação Celular/imunologia , Degranulação Celular/efeitos da radiação , Linhagem Celular Tumoral , Citocinas/biossíntese , Citometria de Fluxo , Xenoenxertos , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/efeitos da radiação , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Ativação Linfocitária/efeitos da radiação , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Ligação Proteica
10.
Oncol Lett ; 13(5): 3781-3786, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28521478

RESUMO

Cytotoxic T lymphocyte antigen-4 (CTLA-4) is expressed during cluster of differentiation (CD)4+ T-cell activation and terminates immune responses by interrupting CD28-enhanced activation. In addition, CTLA-4 is known to be constitutively expressed in regulatory T-cells (Tregs) and to contribute to immune suppression by enhancing the suppressive function of Tregs. However, the molecular mechanisms underlying CTLA-4-mediated Treg suppression remains incompletely understood. Furthermore, it is uncertain whether the in vivo immune suppressive functions of CTLA-4 are mediated only by a reduction in the level of conventional T-cell activity, or enhancement of Treg function. The present study demonstrated that combination therapy with an anti-CTLA-4 monoclonal antibody and dendritic cell-mediated radioimmunotherapy (IR/DC) was able to promote an antitumor response and influence Treg function in a mouse model of lung cancer. Cell surface markers, including CTLA-4, CD25 and CD4, were analyzed using flow cytometry, and T-cell activities were measured using ELISPOT and cytotoxicity assays. It was revealed that anti-CTLA-4 combined treatment with IR/DC immunotherapy may execute a more powerful and effective anti-tumor immunity through the inhibition of Treg function.

11.
Immunobiology ; 222(1): 55-65, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26705936

RESUMO

Hypoxia and infiltration of tumor-associated macrophages (TAM) are intrinsic features of the tumor microenvironment. Tumor cells that remain viable in hypoxic conditions often possess an increased survival potential and tend to grow aggressively. TAM also respond to a variety of signals in the hypoxic tumor microenvironment and express a more M2-like phenotype. In this study, the established mouse tumor tissues showed a dense infiltration of CD206+ macrophages at the junctions between the normoxic and hypoxic regions and an increased IL-6 receptor (IL-6R) expression of tumor cells in the areas of CD206+ TAM accumulation, which indicates a role of M2 phenotype TAM in survival adaptation of tumor cells preparing for an impending hypoxic injury before changes in oxygen availability. Cocultured mouse FM3A or human MCF-7 tumor cells with tumor infiltrating macrophages isolated from mouse tumor tissues and M2-polarized macrophages generated from human THP-1 cells, respectively, showed significantly decreased rate of cell death in cultures exposed to hypoxia. The acquisition of survival resistance was attributed to increased IL-6 production by M2 TAM and increased expression of IL-6R in tumor cells in the coculture system. MCF-7 cells cocultured with M2 TAM showed activated JAK1/STAT3 and Raf/MEK/JNK pathways contributing to tyrosine and serine phophorylation of STAT3, respectively. However, only tyrosine phosphorylated STAT3 was detected in the nucleus, which induced upregulation of Bcl-2 and downregulation of Bax and Bak. Finally, knockdown of IL-6R by small interfering RNA significantly counteracted coculture-induced signals and completely abolished the survival resistance to hypoxic injury. Thus, we present evidence for the role of M2 phenotype TAM in IL-6 receptor-mediated signals, particularly tyrosine phosphorylation of STAT3, responsible for the prosurvival adaptation of tumor cells to hypoxia.


Assuntos
Hipóxia/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Receptores de Interleucina-6/metabolismo , Transdução de Sinais , Microambiente Tumoral/imunologia , Animais , Linhagem Celular , Sobrevivência Celular/imunologia , Técnicas de Cocultura , Citocinas/biossíntese , Feminino , Humanos , Células MCF-7 , Camundongos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Fator de Transcrição STAT3/metabolismo
12.
Sci Rep ; 6: 32470, 2016 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-27671170

RESUMO

Decitabine has been found to have anti-metabolic and anti-tumor activities in various tumor cells. Recently, the use of decitabine in combination with other conventional therapies reportedly resulted in improved anti-tumor activity against various tumors. Ionizing radiation (IR) is widely used as a cancer treatment. Decitabine and IR improve immunogenicity and susceptibility of tumor cells to immune cells by up-regulating the expression of various molecules such as major histocompatibility complex (MHC) class I; natural-killer group 2, member D (NKG2D) ligands; and co-stimulatory molecules. However, the effects of combining decitabine and IR therapies are largely unknown. Our results indicate that decitabine or IR treatment upregulates MHC class I, along with various co-stimulatory molecules in target tumor cells. Furthermore, decitabine and IR combination treatment further upregulates MHC class I, along with the co-stimulatory molecules, when compared to the effect of each treatment alone. Importantly, decitabine treatment further enhanced T cell-mediated cytotoxicity and release of IFN- γ against target tumor cells which is induced by IR. Interestingly, decitabine did not affect NKG2D ligand expression or NK cell-mediated cytotoxicity in target tumor cells. These observations suggest that decitabine may be used as a useful immunomodulator to sensitize tumor cells in combination with other tumor therapies.

13.
Int J Radiat Oncol Biol Phys ; 92(2): 390-8, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25754628

RESUMO

PURPOSE: To investigate the potential of low-dose cyclophosphamide (LD-CTX) and anti-CD25 antibody to prevent activation of regulatory T cells (Tregs) during radiation therapy. METHODS AND MATERIALS: We used LD-CTX and anti-CD25 monoclonal antibody as a means to inhibit Tregs and improve the therapeutic effect of radiation in a mouse model of lung and colon cancer. Mice were irradiated on the tumor mass of the right leg and treated with LD-CTX and anti-CD25 antibody once per week for 3 weeks. RESULTS: Combined treatment of LD-CTX or anti-CD25 antibody with radiation significantly decreased Tregs in the spleen and tumor compared with control and irradiation only in both lung and colon cancer. Combinatorial treatments resulted in a significant increase in the effector T cells, longer survival rate, and suppressed irradiated and distal nonirradiated tumor growth. Specifically, the combinatorial treatment of LD-CTX with radiation resulted in outstanding regression of local and distant tumors in colon cancer, and almost all mice in this group survived until the end of the study. CONCLUSIONS: Our results suggest that Treg depletion strategies may enhance radiation-mediated antitumor immunity and further improve outcomes after radiation therapy.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Neoplasias do Colo/radioterapia , Ciclofosfamida/administração & dosagem , Imunossupressores/administração & dosagem , Subunidade alfa de Receptor de Interleucina-2/antagonistas & inibidores , Neoplasias Pulmonares/radioterapia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/efeitos da radiação , Animais , Neoplasias do Colo/imunologia , Neoplasias do Colo/mortalidade , Terapia Combinada , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/mortalidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C
14.
J Immunother ; 38(3): 107-15, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25751500

RESUMO

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.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Antineoplásicos/administração & dosagem , Carcinoma Pulmonar de Lewis/imunologia , Ciclofosfamida/administração & dosagem , Células Dendríticas/imunologia , Subunidade alfa de Receptor de Interleucina-2/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma Pulmonar de Lewis/mortalidade , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/terapia , Linhagem Celular Tumoral , Terapia Combinada , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos da radiação , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Imunofenotipagem , Imunossupressores/administração & dosagem , Imunoterapia , Masculino , Camundongos , Fenótipo , Radiação , Baço/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/efeitos da radiação , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/efeitos da radiação , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação
15.
Mol Med Rep ; 11(3): 1800-6, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25385045

RESUMO

Radiotherapy has been used to treat cancer for >100 years and is required by numerous patients with cancer. Ionizing radiation effectively inhibits the growth of cancer cells by inducing cell death and increasing anticancer immunity, through the induction of natural killer group 2 member D ligands (NKG2DLs); however, adverse effects have also been reported, including the promotion of metastasis. Matrix metalloproteinases (MMPs) are induced by ionizing radiation and have an important role in the invasion and metastasis of cancer cells. Previously, MMPs were demonstrated to increase the shedding of NKG2DLs, which may reduce the surface expression of NKG2DLs on cancer cells. As a consequence, the cancer cells may escape natural killer (NK)­mediated anticancer immunity. In the present study, NCI­H23 human non­small cell lung cancer cells were used to investigate the combined effects of ionizing radiation and MMP inhibitors on the expression levels of NKG2DLs. Ionizing radiation increased the expression of MMP2 and ADAM metalloproteinase domain 10 protease, as well as NKG2DLs. The combined treatment of ionizing radiation and MMP inhibitors increased the surface expression levels of NKG2DLs and resulted in the increased susceptibility of the cancer cells to NK­92 natural killer cells. Furthermore, soluble NKG2DLs were increased in the media by ionizing radiation and blocked by MMP inhibitors. The present study suggests that radiotherapy may result in the shedding of soluble NKG2DLs, through the induction of MMP2, and combined treatment with MMP inhibitors may minimize the adverse effects of radiotherapy.


Assuntos
Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Metaloproteinases da Matriz/metabolismo , Proteínas ADAM/genética , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/genética , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/genética , Relação Dose-Resposta à Radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Neoplasias Pulmonares/genética , Metaloproteinase 2 da Matriz/genética , Inibidores de Metaloproteinases de Matriz/farmacologia , Metaloproteinases da Matriz/genética , Proteínas de Membrana/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , RNA Mensageiro/genética , Radiação Ionizante
16.
Exp Cell Res ; 330(2): 451-459, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25218028

RESUMO

In the present study, we investigated whether celecoxib could induce the expression of NKG2D ligands in clonogenic colon cancer cells, and increase their susceptibility to NK cell-mediated cell death. Celecoxib and its non-coxib analog, 2,5-dimethyl celecoxib, induced ULBP-1 and DR5 in both COX-2 negative HCT-15 cells and COX-2 positive HT-29 cells. Celecoxib increased their susceptibility to NK92 cells in both DELFIA assay and soft agar colony forming assay. The inducibility of ULBP-1 and DR5 by celecoxib was not different between CD44- and CD44+ HCT-15 cells, and CD133- and CD133+ HT-29 cells. Celecoxib increased the susceptibility of highly clonogenic CD44+ HCT-15 and CD133+ HT-29 cells to NK92 cells, at least comparable to less clonogenic CD44- HCT-15 and CD133- HT-29 cells, respectively. In addition, celecoxib induced CHOP, and thapsigargin, an inducer of ER (endoplasmic reticulum) stress, induced DR5 but not ULBP1 in HCT-15. Taken together, these findings suggest that celecoxib induces the expression of ULBP-1 as well as DR5 in clonogenic colon cancer cells via COX-2 and ER stress-independent pathways, and increases their susceptibility to NK cells.


Assuntos
Neoplasias do Colo/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Células Matadoras Naturais/imunologia , Pirazóis/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Sulfonamidas/farmacologia , Antígeno AC133 , Antígenos CD/biossíntese , Celecoxib , Ciclo-Oxigenase 2/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Proteínas Ligadas por GPI/biossíntese , Glicoproteínas/biossíntese , Células HT29 , Humanos , Receptores de Hialuronatos/biossíntese , Peptídeos , Tapsigargina/farmacologia , Fator de Transcrição CHOP/biossíntese
17.
Biochem Pharmacol ; 92(2): 251-65, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25268843

RESUMO

Recently, the importance of platelet activation in cancer metastasis has become generally accepted. As a result, the development of new platelet inhibitors with minimal adverse effects is now a promising area of targeted cancer therapy. Baicalein is a functional ingredient derived from the root of Scutellaria baicalensis Georgi, a plant used intraditional medicine. The pharmacological effects of this compound including anti-oxidative and anti-inflammatory activities have already been demonstrated. However, its effects on platelet activation are unknown. We therefore investigated the effects of baicalein on ligand-induced platelet aggregation and pulmonary cancer metastasis. In the present study, baicalein inhibited agonist-induced platelet aggregation, granule secretion markers (P-selectin expression and ATP release), [Ca(2+)]i mobilization, and integrin αIIbß3 expression. Additionally, baicalein attenuated ERK2, p38, and Akt activation, and enhanced VASP phosphorylation. Indeed, baicalein was shown to directly inhibit PI3K kinase activity. Moreover, baicalein attenuated the platelet aggregation induced by C6 rat glioma tumor cells in vitro and suppressed CT26 colon cancer metastasis in mice. These features indicate that baicalein is a potential therapeutic drug for the prevention of cancer metastasis.


Assuntos
Moléculas de Adesão Celular/metabolismo , Flavanonas/farmacologia , Proteínas dos Microfilamentos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Agregação Plaquetária/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Moléculas de Adesão Celular/antagonistas & inibidores , Linhagem Celular Tumoral , AMP Cíclico/fisiologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Fosfoproteínas/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Toxicol Res ; 30(3): 211-20, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25343016

RESUMO

Resveratrol has received considerable attention as a polyphenol with various biological effects such as anti-inflammatory, anti-oxidant, anti-mutagenic, anti-carcinogenic, and cardioprotective properties. As part of the overall safety assessment of HS-1793, a novel resveratrol analogue free from the restriction of metabolic instability and the high dose requirement of resveratrol, we assessed genotoxicity in three in vitro assays: a bacterial mutation assay, a comet assay, and a chromosomal aberration assay. In the bacterial reverse mutation assay, HS-1793 did not increase revertant colony numbers in S. typhimurium strains (TA98, TA100, TA1535 and TA1537) or an E. coli strain (WP2 uvrA) regardless of metabolic activation. HS-1793 showed no evidence of genotoxic activity such as DNA damage on L5178Y Tk(+/-) mouse lymphoma cells with or without the S9 mix in the in vitro comet assay. No statistically significant differences in the incidence of chromosomal aberrations following HS-1793 treatment was observed on Chinese hamster lung cells exposed with or without the S9 mix. These results provide additional evidence that HS-1793 is non-genotoxic at the dose tested in three standard tests and further supports the generally recognized as safe determination of HS-1793 during early drug development.

19.
Int J Mol Med ; 34(5): 1349-57, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25176413

RESUMO

Radiation is an important component of therapy for a wide range of malignant conditions. However, it triggers DNA damage and cell death in normal cells and results in adverse side-effects. Cordyceps militaris (C. militaris), a traditional medicinal mushroom, produces the bioactive compound, cordycepin (3'-deoxyadenosine) and has multiple pharmacological activities, such as antitumor, antimetastatic, antioxidant and immunomodulatory effects. The present study was undertaken to investigate whether CM-AE, an extract obtained from C. militaris exerts protective effects against radiation-induced DNA damage. The protective effects of CM-AE were compared with those of cordycepin. CM-AE effectively increased free radical scavenging activity and decreased radiation-induced plasmid DNA strand breaks in in vitro assays. CM-AE significantly inhibited the generation of reactive oxygen species (ROS) and cellular DNA damage in 2 Gy irradiated Chinese hamster ovary (CHO)-K1 cells. Moreover, treatment with CM-AE induced similar levels of phosphorylated H2AX in the cells, which reflects the initial DNA double-strand breaks in the irradiated cells compared with the non-irradiated CHO-K1 cells. However, cordycepin did not show free radical scavenging activity and did not protect against radiation-induced plasmid DNA or cellular DNA damage. These results suggest that the free radical scavenging activity of CM-AE contributes towards its DNA radioprotective effects and that the protective effects of CM-AE are much more potent to those of cordycepin. The data presented in this study may provide useful information for the screening of potent radioprotective materials.


Assuntos
Cordyceps/química , Protetores contra Radiação/farmacologia , Agaricales/química , Animais , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Ensaio Cometa , Cricetinae , Cricetulus , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Desoxiadenosinas/farmacologia , Relação Dose-Resposta à Radiação , Raios gama/efeitos adversos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo
20.
Int Immunopharmacol ; 22(2): 303-10, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25042796

RESUMO

Macrophages are capable of both inhibiting and promoting the growth and spread of cancers, depending on their activation state. Tumor-associated macrophages (TAM) are a kind of alternatively activated M2 macrophage, which may contribute to tumor progression. Following our previous study to evaluate the anti-tumor effect of a synthetic resveratrol analog HS-1793, the current study demonstrated that HS-1793 treatment significantly increased IFN-γ secreting cells in splenocytes and decreased CD206+ macrophage infiltration compared to CD68+ cells in the tumor site with a higher expression of IFN-γ. As these results suggested that IFN-γ increased locally at the tumor sites could modulate the status of TAM, we designed an in vitro model to study macrophage morphology and functions in relation to the tumor microenvironment. Human monocytic cell line THP-1 cells stimulated with phorbol-12-myristate-13-acetate (PMA) differentiated to macrophages with M2-like phenotypes. TAM-like properties of CD206(high), CD204(high), IL-10(high), TGF-ß(high), IL-6(low), IL-12(low), VEGF(high), and MMP-9(high) and promotion of tumor cell invasion were more pronounced in M-2-polarized THP-1 macrophages generated by differentiating THP-1 cells with PMA and subsequently polarizing them with Th2 cytokines (IL-4/IL-13). Upon IFN-γ exposure, THP-1-derived TAM changed their phenotypes to the M-1-like morphology and intracellular granular pattern with an expression of an increased level of proinflammatory and immunostimulatory cytokines and a reduced level of immunosuppressive and tumor progressive mediators. These results explain the underlying mechanism of the anti-tumor activity of HS-1793. The elevated level of IFN-γ production after HS-1793 treatment evoked reprogramming of M-2 phenotype TAM, which efficiently countered the immunosuppressive and tumor progressive influences of TAM.


Assuntos
Interferon gama/imunologia , Macrófagos/efeitos dos fármacos , Naftóis/farmacologia , Neoplasias/imunologia , Resorcinóis/farmacologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Feminino , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C3H , Invasividade Neoplásica , Neoplasias/patologia , Resveratrol , Estilbenos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...