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










Publication year range
1.
Biol Pharm Bull ; 45(1): 34-41, 2022.
Article in English | MEDLINE | ID: mdl-34980779

ABSTRACT

Imatinib mesylate is a potent tyrosine kinase inhibitor that may induce immunological effects, such as inhibition of immune suppressive cells; but, how it modulates the immune system remains to be completely elucidated. In this study, we showed that cell proliferation of CT26 colon cancer and Lewis lung carcinoma (3LL) lung cancer cells was not inhibited by imatinib in vitro, although its administration significantly suppressed the growth of CT26, but not 3LL, subcutaneous tumors, and prolonged survival in CT26 tumor-bearing mice. Further, we examined the expression of immune cell-related molecules in the tumors to elucidate the differences in imatinib-mediated antitumor effects between CT26 and 3LL tumors. The nCounter assay showed that the expression of CD8 and CD8+ T cell-recruiting chemokine genes was significantly elevated in imatinib-treated CT26 tumors than that in control tumors; however, the gene expression remained unchanged in imatinib-treated or control 3LL tumors. Furthermore, frequency of interferon-γ+ (IFN-γ+) CD8+ T cells was increased in imatinib-treated CT26 tumors than control tumors, indicating induction of antitumor immunity by imatinib. The analysis indicates that imatinib promotes infiltration of effector T cells in tumors by upregulating expression of cytokines that recruit CD8+ T cells in the tumor microenvironment, which may lead to a strong antitumor effect.


Subject(s)
CD8-Positive T-Lymphocytes , Colonic Neoplasms , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Imatinib Mesylate/metabolism , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Tumor Microenvironment
2.
Sci Rep ; 9(1): 5562, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30944344

ABSTRACT

An anti-glucocorticoid induced TNF receptor (GITR) agonistic antibody (Ab) induces an antitumor immunity with both stimulation of effector T cells and inhibition of regulatory T cell activity. To enhance GITR Ab-mediated tumor immunity, we focused on the intratumoral route, since a tumor-localized high concentration of Ab would confer activation of only tumor-infiltrating T cells. First, in a murine colon cancer model, we showed that the intratumoral delivery of Ab significantly increased the number of effector T cells infiltrated into tumors, and suppressed tumor growth more effectively than the intraperitoneal and intravenous injections did. Then, we found that the injection of Ab into the peritumoral area induced a systemic antitumor immunity at a similar level to the intratumoral injection. Therefore, we hypothesized that the transfer of locally administrated Ab into tumor-draining lymph nodes (TDLNs) plays an important role in inducing an effective immunity. In fact, intratumorally or peritumorally injected Ab was detected in TDLNs, and resection of Ab-injected TDLNs significantly reduced GITR Ab-mediated systemic tumor immunity. Intratumoral injection showed less number of auto-reactive T cells in the spleen than the intraperitoneal injection did. Intratumoral delivery of GITR Ab is a promising approach to induce an effective immunity compared to the systemic delivery.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Glucocorticoid-Induced TNFR-Related Protein/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Colonic Neoplasms/pathology , Female , Glucocorticoid-Induced TNFR-Related Protein/immunology , Injections, Intralesional , Injections, Intraperitoneal , Injections, Intravenous , Interferon-gamma/metabolism , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice, Inbred BALB C , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
3.
Cancer Gene Ther ; 26(9-10): 334-343, 2019 09.
Article in English | MEDLINE | ID: mdl-30420718

ABSTRACT

The effect of IFN-α on the immunosuppressive tumor microenvironment is not fully understood. We previously reported that intratumoral IFN-α gene transduction decreased the frequency of regulatory T cells (Tregs) in the tumor by inducing the secretion of IL-6 from dendritic cells. In this study, we examined whether IFN-α affects the trafficking of Tregs to the tumor. Since CT26 cells expressed CCL17 among Treg-attracting chemokines, we focused on its role in IFN-α-mediated Treg suppression. IFN-α directly suppressed CCL17 production from CT26 cells in vitro, and IFN-α transduction reduced CCL17 expression in tumors in vivo. Next, to investigate whether CCL17 downregulation is related to the suppression of Treg trafficking, CCL17-downregulated CT26 cells produced using short hairpin RNA (CT26-shCCL17) were inoculated into mice. The frequency of Tregs in CT26-shCCL17 tumors was reduced and tumor growth was suppressed. Finally, to examine the combinatorial effect of IFN-α expression with CCL17 downregulation, IFN-α was transduced into CT26-shCCL17 tumors. This resulted in an elevation of CT26-specific CD8+ T cells and the complete eradication of tumors. This study shows a novel mechanism of IFN-α-mediated Treg suppression, and combining IFN-α gene therapy with strong CCL17 downregulation could offer a promising strategy for the treatment of cancer.


Subject(s)
Chemokine CCL17/genetics , Interferon-alpha/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/etiology , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adenoviridae/genetics , Animals , Cell Line, Tumor , Chemokine CCL17/metabolism , Disease Models, Animal , Female , Gene Expression , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Humans , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Neoplasms/pathology , Neoplasms/therapy , RNA, Small Interfering/genetics , T-Lymphocytes, Regulatory/pathology , Tumor Microenvironment , Xenograft Model Antitumor Assays
4.
Biochem Biophys Res Commun ; 509(1): 96-101, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30579597

ABSTRACT

The lymphopenic condition following autologous hematopoietic stem cell transplantation (HSCT) enhances the proliferation of T cells by engaging tumor-associated antigens, leading to the alteration of the T-cell repertoire towards antitumor immunity. However, cure by autologous HSCT alone have rarely occurred in the clinical setting. Since tumor-reactive lymphocytes preferentially proliferate during reconstitution of the immune system, we examined whether the priming of donor lymphocytes can strengthen the antitumor effect by HSCT in a CT26 murine colon cancer model. The systemic administration of an anti-glucocorticoid-induced TNF receptor (GITR) agonistic antibody (Ab) significantly increased the number of CT26-responsive T cells but not that of auto-reactive lymphocytes in donor mice. The infusion of non-primed and GITR Ab-primed donor lymphocytes suppressed the CT26 tumor growth, and only the primed lymphocytes eliminated tumors in all the treated mice. The frequency of CT26-responsive T cells was elevated in recipient mice infused with both primed and non-primed lymphocytes until 4 weeks after transplantation, while the frequency in recipients with primed lymphocytes was markedly elevated compared with that in mice harboring non-primed lymphocytes at 2 weeks. The frequencies of regulatory T cells and myeloid-derived suppressor cells were elevated in recipient mice infused with primed and non-primed lymphocytes 2 weeks after transplantation, and returned to normal levels by week 4. The combination of autologous HSCT with pre-immunization of donor lymphocytes is a promising strategy to induce strong antitumor immunity.


Subject(s)
Antibodies/therapeutic use , Colonic Neoplasms/therapy , Glucocorticoid-Induced TNFR-Related Protein/immunology , Hematopoietic Stem Cell Transplantation/methods , Lymphocytes/immunology , Animals , Antibodies/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Female , Glucocorticoid-Induced TNFR-Related Protein/antagonists & inhibitors , Immunity , Immunization , Mice , Mice, Inbred BALB C , Transplantation, Autologous/methods
5.
Cancer Med ; 5(1): 49-60, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26589884

ABSTRACT

Autologous hematopoietic stem cell transplantation (HSCT) can induce a strong antitumor immunity by homeostatic proliferation (HP) of T cells and suppression of regulatory T cells following preconditioning-induced lymphopenia. However, the role of innate immunity including natural killer (NK) cells is still not understood. Here, first, we examined whether NK cells exert an antitumor effect after syngeneic HSCT in a murine colon cancer model. Flow cytometry showed that NK cells as well as T cells rapidly proliferated after HSCT, and the frequency of mature NK cells was increased in tumor during HP. Furthermore, NK cells undergoing HP were highly activated, which contributed to substantial tumor suppression. Then, we found that a large number of neutrophils accumulated in tumor early after syngeneic HSCT. It was recently reported that neutrophil-derived mediators modulate NK cell effector functions, and so we examined whether the neutrophils infiltrated in tumor are associated with NK cell-mediated antitumor effect. The depletion of neutrophils significantly impaired an activation of NK cells in tumor and increased the fraction of proliferative NK cells accompanied by a decrease in NK cell survival. The results suggested that neutrophils in tumor prevent NK cells from activation-induced cell death during HP, thus leading to a significant antitumor effect by NK cells. This study revealed a novel aspect of antitumor immunity induced by HSCT and may contribute to the development of an effective therapeutic strategy for cancer using HSCT.


Subject(s)
Cell Communication/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Neutrophils/immunology , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/metabolism , Lymphocyte Activation , Lymphocyte Depletion , Mice , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Neutrophil Infiltration , Neutrophils/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
6.
PLoS One ; 10(12): e0143804, 2015.
Article in English | MEDLINE | ID: mdl-26625258

ABSTRACT

Definitive chemoradiotherapy (CRT) is a less invasive therapy for esophageal squamous cell carcinoma (ESCC). Five-year survival rate of locally advanced ESCC patients by definitive CRT were 37%. We previously reported that tumor-specific cytotoxic T-lymphocyte (CTL) activation signatures were preferentially found in long-term survivors. However, it is unknown whether the CTL activation is actually driven by CRT. We compared gene expression profiles among pre- and post-treatment biopsy specimens of 30 ESCC patients and 121 pre-treatment ESCC biopsy specimens. In the complete response (CR) cases, 999 overexpressed genes including at least 234 tumor-specific CTL-activation associated genes such as IFNG, PRF1, and GZMB, were found in post-treatment biopsy specimens. Clustering analysis using expression profiles of these 234 genes allowed us to distinguish the immune-activated cases, designating them as I-type, from other cases. However, despite the better CR rate in the I-type, overall survival was not significantly better in both these 30 cases and another 121 cases. Further comparative study identified a series of epithelial to mesenchymal transition-related genes overexpressed in the early relapse cases. Importantly, the clinical outcome of CDH2-negative cases in the I-type was significantly better than that of the CDH2-positive cases in the I-type. Furthermore, NK cells, which were activated by neutrophils-producing S100A8/S100A9, and CTLs were suggested to cooperatively enhance the effect of CRT in the CDH2-negative I-type. These results suggested that CTL gene activation may provide a prognostic advantage in ESCCs with epithelial characteristics.


Subject(s)
Carcinoma, Squamous Cell/therapy , Chemoradiotherapy , Esophageal Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/prevention & control , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Esophageal Neoplasms/prevention & control , Esophageal Squamous Cell Carcinoma , Humans , Middle Aged , Secondary Prevention , Transcriptome
7.
J Immunol ; 194(11): 5539-48, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25911757

ABSTRACT

S100A8/A9, a proinflammatory protein, is upregulated in inflammatory diseases, and also has a tumor-promoting activity by the recruitment of myeloid cells and tumor cell invasion. However, whether the expression of S100A8/A9 in tumors predicts a good or poor prognosis is controversial in the clinical setting. In this study, to clarify the in vivo role of S100A8/A9 in the tumor microenvironment, we s.c. inoculated Pan02 cells stably expressing S100A8 and S100A9 proteins (Pan02-S100A8/A9) in syngeneic C57BL/6 mice. Unexpectedly, after small tumor nodules were once established, they rapidly disappeared. Flow cytometry showed that the number of NK cells in the tumors was increased, and an administration of anti-asialoGM1 Ab for NK cell depletion promoted the growth of Pan02-S100A8/A9 s.c. tumors. Although the S100A8/A9 proteins alone did not change the IFN-γ expression of NK cells in vitro, a coculture with Pan02 cells, which express Rae-1, induced IFN-γ production, and Pan02-S100A8/A9 cells further increased the number of IFN-γ(+) NK cells, suggesting that S100A8/A9 enhanced the NK group 2D ligand-mediated intracellular activation pathway in NK cells. We then examined whether NK cell activation by S100A8/A9 was via their binding to receptor of advanced glycation end product (RAGE) by using the inhibitors. RAGE antagonistic peptide and anti-RAGE Ab inhibited the IFN-γ production of NK cells induced by S100A8/A9 proteins, and an administration of FPS-ZM1, a RAGE inhibitor, significantly enhanced the in vivo growth of Pan02-S100A8/A9 tumors. We thus found a novel activation mechanism of NK cells via S100A8/A9-RAGE signaling, which may open a novel perspective on the in vivo interaction between inflammation and innate immunity.


Subject(s)
Calgranulin A/immunology , Calgranulin B/immunology , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Pancreatic Neoplasms/immunology , Receptors, Immunologic/immunology , Animals , Benzamides/pharmacology , Calgranulin A/biosynthesis , Calgranulin B/biosynthesis , Cell Line, Tumor , Cell Proliferation , Female , Inflammation/immunology , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Receptor for Advanced Glycation End Products , Receptors, Immunologic/antagonists & inhibitors , Transplantation, Isogeneic , Tumor Microenvironment/immunology
8.
J Control Release ; 192: 284-93, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25108153

ABSTRACT

The addition of a targeting strategy is necessary to enhance oncolysis and secure safety of a conditionally replicative adenovirus (CRAd). We have constructed an adenovirus library displaying random peptides on the fiber, and have successfully identified a pancreatic cancer-targeting ligand (SYENFSA). Here, the usefulness of cancer-targeted CRAd for pancreatic cancer was examined as a preclinical study. First, we constructed a survivin promoter-regulated CRAd expressing enhanced green fluorescent protein gene (EGFP), which displayed the identified targeting ligand (AdSur-SYE). The AdSur-SYE resulted in higher gene transduction efficiency and oncolytic potency than the untargeted CRAd (AdSur) in several pancreatic cancer cell lines. An intratumoral injection of AdSur-SYE significantly suppressed the growth of subcutaneous tumors, in which AdSur-SYE effectively proliferated and spread. An ectopic infection in adjacent tissues and organs of intratumorally injected AdSur-SYE was decreased compared with AdSur. Then, to examine whether the targeting ligand actually enhanced the infectivity of CRAd in human pancreatic cancer tissues, tumor cells prepared from surgical specimens were infected with viruses. The AdSur-SYE increased gene transduction efficiency 6.4-fold higher than did AdSur in single cells derived from human pancreatic cancer, whereas the infectivity of both vectors was almost the same in the pancreas and other cancers. Immunostaining showed that most EGFP(+) cells were cytokeratin-positive in the sliced tissues, indicating that pancreatic cancer cells but not stromal cells were injected with AdSur-SYE. AdSur-SYE resulted in a stronger oncolysis in the primary pancreatic cancer cells co-cultured with mouse embryonic fibroblasts than AdSur did. CRAd in combination with a tumor-targeting ligand is promising as a next-generation of oncolytic virotherapy for pancreatic cancer.


Subject(s)
Adenoviridae/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Pancreatic Neoplasms/therapy , Animals , Female , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Humans , Inhibitor of Apoptosis Proteins/genetics , Mice , Mice, Inbred BALB C , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , Survivin
9.
Mol Pharm ; 11(3): 1069-74, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24380399

ABSTRACT

Redirection of adenovirus vectors by engineering the capsid-coding region has shown limited success because proper targeting ligands are generally unknown. To overcome this limitation, we constructed an adenovirus library displaying random peptides on the fiber knob, and its screening led to successful selections of several particular targeted vectors. In the previous library construction method, the full length of an adenoviral genome was generated by a Cre-lox mediated in vitro recombination between a fiber-modified plasmid library and the enzyme-digested adenoviral DNA/terminal protein complex (DNA-TPC) before transfection to the producer cells. In this system, the procedures were complicated and time-consuming, and approximately 30% of the vectors in the library were defective with no displaying peptide. These may hinder further extensive exploration of cancer-targeting vectors. To resolve these problems, in this study, we developed a novel method with the transfection of a fiber-modified plasmid library and a fiberless adenoviral DNA-TPC in Cre-expressing 293 cells. The use of in-cell Cre recombination and fiberless adenovirus greatly simplified the library-making steps. The fiberless adenovirus was useful in suppressing the expansion of unnecessary adenovirus vectors. In addition, the complexity of the library was more than a 10(4) level in one well in a 6-well dish, which was 10-fold higher than that of the original method. The results demonstrated that this novel method is useful in producing a high quality live adenovirus library, which could facilitate the development of targeted adenovirus vectors for a variety of applications in medicine.


Subject(s)
Adenoviridae/genetics , Capsid Proteins/genetics , DNA, Viral/genetics , Genetic Vectors/genetics , Genome, Viral , Peptide Fragments/metabolism , Peptide Library , Adenocarcinoma/genetics , Adenocarcinoma/virology , Adenoviridae Infections/genetics , Adenoviridae Infections/virology , Genetic Engineering , HEK293 Cells , Humans , Integrases/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/virology , Plasmids/administration & dosage , Transduction, Genetic
10.
Cancer Sci ; 105(2): 159-67, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24289533

ABSTRACT

We have reported that interferon (IFN)-α can attack cancer cells by multiple antitumor mechanisms including the induction of direct cancer cell death and the enhancement of an immune response in several pancreatic cancer models. However, an immunotolerant microenvironment in the tumors is often responsible for the failure of the cancer immunotherapy. Here we examined whether the suppression of regulatory T cells (Tregs) within tumors can enhance an antitumor immunity induced by an intratumoral IFN-α gene transfer. First we showed that an intraperitoneal administration of an agonistic anti-glucocorticoid induced TNF receptor (GITR) monoclonal antibody (mAb), which is reported to suppress the function of Tregs, significantly inhibited subcutaneous tumor growth in a murine pancreatic cancer model. The anti-GITR mAb was then combined with the intratumoral injection of the IFN-α-adenovirus vector. The treatment with the antibody synergistically augmented the antitumor effect of IFN-α gene therapy not only in the vector-injected tumors but also in the vector-uninjected tumors. Immunostaining showed that the anti-GITR mAb decreased Foxp3(+) cells infiltrating in the tumors, while the intratumoral IFN-α gene transfer increased CD4(+) and CD8(+) T cells in the tumors. Therefore, the combination therapy strongly inclined the immune balance of the tumor microenvironment in an antitumor direction, leading to a marked systemic antitumor effect. The CCR5 expression on Tregs was downregulated in the antibody-treated mice, which may explain the decrease of tumor-infiltrating Tregs. The combination of Treg-suppression by GITR mAb and the tumor immunity induction by IFN-α gene therapy could be a promising therapeutic strategy for pancreatic cancer.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Immune Tolerance/immunology , Interferon-alpha/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Forkhead Transcription Factors/immunology , Gene Transfer Techniques , Genetic Therapy/methods , Immune Tolerance/drug effects , Immunotherapy/methods , Injections, Intralesional , Interferon-alpha/administration & dosage , Interferon-alpha/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pancreatic Neoplasms/genetics , T-Lymphocytes, Regulatory/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
11.
J Immunol ; 191(6): 3440-52, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23966628

ABSTRACT

Lymphopenia-induced homeostatic proliferation of T cells after autologous hematopoietic stem cell transplantation (HSCT) skews the T cell repertoire by engaging tumor-associated Ags, leading to an induction of antitumor immunity. However, how HSCT alters the immunosuppressive microenvironment in the tumors is unknown. In this study, we first analyzed the kinetics of regulatory T cells (Tregs) in the tumors after syngeneic HSCT. Unexpectedly, the frequency of CD4⁺ cells expressing Foxp3 was increased in the spleens, whereas the frequency was clearly decreased in the tumors after HSCT. The origin of reconstituted CD4⁺ and Foxp3⁺ cells in the tumors was mainly from the expansion of transferred splenic T cells. Then, to examine the mechanism of Treg suppression after HSCT, we isolated CD11c⁺ cells from tumors. A large amount of Treg-inhibitory cytokine IL-6 was secreted from the CD11c⁺ cells in the tumors, but not in the spleens in the recipient mice. Furthermore, to understand what factor affects the activity of CD11c⁺ cells in the tumors after HSCT, we analyzed the expression of various cytokines/chemokines with mouse cytokine Ab arrays, and noticed that VEGF-D concentration was increased in the tumors in the early period after HSCT. The CD11c⁺ cells produced IL-6 in response to VEGF-D stimulation, and an administration of VEGF receptor-3 neutralizing Ab significantly suppressed the production of IL-6 from CD11c⁺ cells accompanied with the increase of Tregs in the tumors of HSCT recipients. Autologous HSCT creates an environment that strongly supports the enhancement of antitumor immunity in reconstituted lymphopenic recipients through the suppression of Tregs.


Subject(s)
Hematopoietic Stem Cell Transplantation , Neoplasms, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Vascular Endothelial Growth Factor D/metabolism , Animals , Female , Flow Cytometry , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/metabolism
12.
Cancer Med ; 2(5): 636-45, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24403229

ABSTRACT

Lymphopenia-induced homeostatic proliferation (HP) of T cells following autologous hematopoietic stem cell transplantation (HSCT) skews the T-cell repertoire by engaging tumor-associated antigens (TAAs), leading to an induction of antitumor immunity. Here, as the tumor-reactive lymphocytes preferentially proliferate during the condition of HP, we examined whether the priming of a donor lymphocytes to TAAs could enhance HP-induced antitumor immunity in autologous HSCT recipients. First, to examine whether the tumor-bearing condition of donor influences the antitumor effect of HSCT, the lymphocytes isolated from CT26 tumor-bearing mice were infused into lethally irradiated mice. The growth of tumors was substantially suppressed in the mice that received HSCT from a tumor-bearing donor compared with a naïve donor, suggesting that a fraction of donor lymphocytes from tumor-bearing mice are primed in response to TAAs and remain responsive upon transplantation. We previously reported that type I interferon (IFN) maturates the dendritic cells and promotes the priming of T cells. We then investigated whether the further priming of donor cells by IFN-α can strengthen the antitumor effect of HSCT. The intratumoral IFN-α gene transfer significantly increased the number of IFN-γ-positive lymphocytes in response to CT26 cells but not the syngeneic lymphocytes in donor mice. The infusion of primed donor lymphocytes markedly suppressed the tumor growth in recipient mice, and cured 64% of the treated mice. Autologous HSCT with the infusion of primed donor lymphocytes is a promising strategy to induce an effective antitumor immunity for solid cancers.


Subject(s)
Colonic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Lymphocyte Transfusion/methods , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Female , Gene Transfer Techniques , Genetic Therapy/methods , Interferon-alpha/genetics , Interferon-alpha/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C
13.
Acta Cytol ; 56(3): 297-303, 2012.
Article in English | MEDLINE | ID: mdl-22555533

ABSTRACT

OBJECTIVE: Since well-differentiated adenocarcinoma cells of the lung (G1 cancer cells) show mild atypia, their differentiation from benign columnar epithelial cells (benign cells) is often difficult based on morphology. We performed discriminant analysis to distinguish benign from malignant cells by measuring 3-dimensional (3D) changes in nuclear luminance. STUDY DESIGN: Discriminant analysis of 231 atypical cells prepared by bronchial brushing cytology, which were difficult to morphologically classify as benign or malignant, was performed using 100 G1 cancer cells. One hundred benign cells of bronchial brushing cytology specimens served as controls. The number of pixels of the nucleus, the number of focus layers and the level of change in the coefficient of variation (CV) of nuclear luminance between layers (3D-CV) were used as analytic parameters, and benign cells were discriminated from malignant cells based on the Mahalanobis distance. RESULTS: As a result of discriminant analysis using a cutoff value determined in the control group, about 90% of the atypical cells difficult to classify as benign or malignant could be classified. CONCLUSION: For cells difficult to morphologically classify as benign or malignant, discriminant analysis based on 3D changes in nuclear luminance may be useful. This method can provide objective parameters for cancer diagnosis.


Subject(s)
Adenocarcinoma/pathology , Bronchi/pathology , Cell Nucleus/pathology , Epithelial Cells/pathology , Imaging, Three-Dimensional/methods , Lung Neoplasms/pathology , Adenocarcinoma/diagnosis , Cell Differentiation/physiology , Cytodiagnosis/methods , Diagnosis, Differential , Discriminant Analysis , Humans , Lung Neoplasms/diagnosis
14.
Acta Cytol ; 56(2): 189-95, 2012.
Article in English | MEDLINE | ID: mdl-22378083

ABSTRACT

OBJECTIVE: The cytological diagnosis of coelomic fluid is essential for examining malignant mesothelioma (MM). However, reactive mesothelium (RM), caused by various factors, is morphologically similar to MM and thus often complicates the differential diagnosis. Here, nuclear luminance and steric alterations were assessed for the discriminant analysis of MM and RM. STUDY DESIGN: Thirteen epithelial MM and 11 RM cases were included. One hundred alterations in the numbers of nuclear pixels and focus layers and the coefficient of variation of nuclear luminance among layers were determined for each case to conduct discriminant analysis using the Mahalanobis distance. RESULTS: A cutoff value of 0.072 allowed highly accurate discrimination of MM (89.5%) and RM (89.6%). Fifteen cells appeared in 6 agglomerates of indiscriminable MM cases. The 6 agglomerates were individually examined. Malignant cells were dominant in 3 agglomerates (50%), allowing the discrimination of malignant cases. CONCLUSION: Discrimination using nuclear luminance and steric alterations is useful for morphologically indiscriminable MM cases. Three-dimensional analysis of agglomerates will be further investigated to improve the diagnostic accuracy.


Subject(s)
Cytodiagnosis/methods , Epithelial Cells/pathology , Imaging, Three-Dimensional/methods , Liver Neoplasms/pathology , Lung Neoplasms/pathology , Mesothelioma/pathology , Adult , Aged , Aged, 80 and over , Diagnosis, Differential , Epithelium/pathology , Female , Humans , Imaging, Three-Dimensional/standards , Male , Microscopy/methods , Middle Aged , Young Adult
15.
Hum Gene Ther ; 23(2): 173-86, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21958207

ABSTRACT

Sarcoma at advanced stages remains a clinically challenging disease. Interferons (IFNs) can target cancer cells by multiple antitumor activities, including the induction of cancer cell death and enhancement of immune response. However, the development of an effective cancer immunotherapy is often difficult, because cancer generates an immunotolerant microenvironment against the host immune system. An autologous hematopoietic stem cell transplantation (HSCT) is expected to reconstitute a fresh immune system, and expand tumor-specific T cells through the process of homeostatic proliferation. Here we examined whether a combination of autologous HSCT and IFNs could induce an effective tumor-specific immune response against sarcoma. First, we found that a type I IFN gene transfer significantly suppressed the cell growth of various sarcoma cell lines, and that IFN-ß gene transfer was more effective in inducing cell death than was IFN-α in sarcoma cells. Then, to examine the antitumor effect in vivo, human sarcoma cells were inoculated in immune-deficient mice, and a lipofection of an IFN-ß-expressing plasmid was found to suppress the growth of subcutaneous tumors significantly. Finally, the IFN gene transfer was combined with syngeneic HSCT in murine osteosarcoma models. Intratumoral IFN-ß gene transfer markedly suppressed the growth of vector-injected tumors and inhibited formation of spontaneous lung and liver metastases in syngeneic HSCT mice, and an infiltration of many immune cells was recognized in metastatic tumors of the treated mice. The treated mice showed no significant adverse events. A combination of intratumoral IFN gene transfer with autologous HSCT could be a promising therapeutic strategy for patients with sarcoma.


Subject(s)
Hematopoietic Stem Cell Transplantation , Interferon-beta/genetics , Sarcoma, Experimental/therapy , Animals , Cell Line, Tumor , Female , Gene Transfer Techniques , Genetic Therapy , Humans , Immunotherapy , Interferon-beta/immunology , Lipids , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Plasmids , Sarcoma, Experimental/immunology , Sarcoma, Experimental/pathology , Xenograft Model Antitumor Assays
16.
Intern Med ; 50(11): 1239-44, 2011.
Article in English | MEDLINE | ID: mdl-21628942

ABSTRACT

Immunoglobulin G4 (IgG4)-related tubulointerstitial nephritis (TIN) is often accompanied by autoimmune pancreatitis (AIP) or chronic sclerosing dacryoadenitis and sialoadenitis. However, IgG4-related TIN without AIP or lacrimal and/or salivary gland lesions has not been well recognized. Here, we report a case of IgG4-related TIN associated with hepatic inflammatory pseudotumor without AIP or lacrimal and/or salivary gland lesions. A 58-year-old Japanese man with epigastralgia underwent contrast-enhanced computed tomography (CT), which revealed multiple low-density lesions in both kidneys and a low density hepatic mass. Laboratory tests showed an extremely high level of serum IgG4. Percutaneous renal and hepatic biopsies showed diffuse infiltration of lymphocytes and IgG4-positive plasma cells with fibrosis in both tissues. Two months after administration of oral prednisolone, both lesions decreased in size on follow-up CT, and the serum creatinine level also improved. No recurrence has been detected for two years with a maintenance dose of prednisolone.


Subject(s)
Glomerulonephritis, Membranoproliferative , Granuloma, Plasma Cell/epidemiology , Immunoglobulin G/blood , Liver Diseases/epidemiology , Nephritis, Interstitial/epidemiology , Nephritis, Interstitial/immunology , Biopsy , Comorbidity , Glucocorticoids/therapeutic use , Granuloma, Plasma Cell/diagnosis , Granuloma, Plasma Cell/drug therapy , Humans , Kidney/pathology , Liver/pathology , Liver Diseases/diagnosis , Liver Diseases/drug therapy , Male , Middle Aged , Nephritis, Interstitial/drug therapy , Prednisolone/therapeutic use , Tomography, X-Ray Computed , Treatment Outcome
17.
Int J Colorectal Dis ; 26(11): 1453-62, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21674139

ABSTRACT

BACKGROUND: Although adrenomedullin (AM) is known to ameliorate inflammatory processes, few data exist regarding the effect of AM on inflammatory colitis. Therefore, we examined the effect of AM on inflammatory response in vitro and in vivo colitis model. METHODS: In mice experimental colitis induced by 3% dextran sulfate sodium (DSS) in drinking water for 7 days, AM with 225-900 µg/kg in 0.5 ml of saline or saline alone were given intraperitoneally once a day. In the in vitro experiment, we determined the cytokine response in THP-1 cell activated by lipopolysaccharide with or without AM of 10 nM. Additionally, we performed wound healing assay in Caco-2 cell interfered by DSS with or without AM of 100 nM. RESULTS: In the colitis model, AM significantly reduced the disease activity index, histological score, and local production of inflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 in accordance with reduction of serum amyloid A levels. Secretion of TNF-α in lipopolysaccharide-stimulated THP-1 cells was significantly reduced in the presence of AM. The distance of wound healing interfered by 0.25% DSS was significantly improved in the presence of AM of 100 nM. CONCLUSIONS: These results demonstrate that AM could ameliorate DSS-induced experimental colitis possibly through suppression of systemic and local production of cytokines such as TNF-α, associated with acceleration of ulcer reepithelialization and colon tissue regeneration.


Subject(s)
Adrenomedullin/therapeutic use , Colitis/drug therapy , Inflammation/drug therapy , Adrenomedullin/pharmacology , Animals , Body Weight/drug effects , Cell Line , Cell Movement/drug effects , Colitis/chemically induced , Colitis/complications , Colon/drug effects , Colon/enzymology , Colon/pathology , Cytokines/biosynthesis , Dextran Sulfate , Epithelium/drug effects , Epithelium/pathology , Humans , Inflammation/complications , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred BALB C , Peroxidase/metabolism , Serum Amyloid A Protein/metabolism , Ulcer/complications , Ulcer/pathology , Up-Regulation/drug effects
18.
Cancer Sci ; 101(7): 1686-94, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20487262

ABSTRACT

Type I interferon (IFN) protein is a cytokine with pleiotropic biological functions that include induction of apoptosis, inhibition of angiogenesis, and immunomodulation. We have demonstrated that intratumoral injection of an IFN-alpha-expressing adenovirus effectively induces cell death of cancer cells and elicits a systemic tumor-specific immunity in several animal models. On the other hand, reports demonstrated that an elevation of IFN in the serum following an intramuscular delivery of a vector is able to activate antitumor immunity. In this study, we compared the intratumoral and systemic routes of IFN gene transfer with regard to the effect and safety of the treatment. Intratumoral injection of an IFN-alpha adenovirus effectively activated tumor-responsive lymphocytes and caused tumor suppression not only in the gene-transduced tumors but also in distant tumors, which was more effective than the intravenous administration of the same vector. The expression of co-stimulatory molecules on CD11c(+) cells isolated from regional lymph nodes was enhanced by IFN gene transfer into the tumors. Systemic toxicity such as an elevation of hepatic enzymes was much lower in mice treated by intratumoral gene transfer than in those treated by systemic gene transfer. Our data suggest that the intratumoral route of the IFN vector is superior to intravenous administration, due to the effective induction of antitumor immunity and the lower toxicity.


Subject(s)
Colonic Neoplasms/immunology , Gene Transfer Techniques , Interferon-alpha/genetics , Kidney Neoplasms/immunology , Adenoviridae/genetics , Animals , CD11c Antigen/genetics , Cell Death , Cell Line, Tumor , Colonic Neoplasms/pathology , DNA Primers , Genetic Vectors , Interferon-alpha/blood , Interferon-alpha/therapeutic use , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Safety
19.
Cancer Immunol Immunother ; 58(7): 1007-21, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18998126

ABSTRACT

One of the major challenges in the treatment of solid cancers by allogenic hematopoietic stem cell transfer (alloHSCT) is the specific enhancement of antitumor immunity. Interferon (IFN) is a cytokine with pleiotropic biological functions including an immunomoduration, and our preclinical studies have shown that an intratumoral IFN-alpha gene transfer induced strong local tumor control and systemic tumor-specific immunity. In the present study, we examined whether the IFN-alpha gene transfer could enhance recognition of tumor-associated antigens by donor T cells and augment the antitumor activity of alloHSCT. First, when a mouse IFN-alpha adenovirus vector (Ad-mIFN) was injected into subcutaneous xenografts of syngeneic renal and colon cancer cells, tumor growth was significantly suppressed in a dose-dependent manner. A significant tumor cell death and infiltration of immune cells was recognized in the Ad-mIFN-injected tumors, and the dendritic cells isolated from the tumors showed a strong Th1-oriented response. The antitumor effect of Ad-mIFN was then examined in a murine model of minor histocompatibility antigen-mismatched alloHSCT. The intratumoral IFN-alpha gene transfer caused significant tumor suppression in the alloHSCT recipients, and this suppression was evident not only in the gene-transduced tumors but also in simultaneously inoculated distant tumors which did not receive the vector injection. A cytotoxicity assay showed specific tumor cell lysis by donor T cells responding to IFN-alpha. Graft-versus-host disease was not exacerbated serologically or clinically in the mice treated with IFN-alpha. This combination strategy deserves evaluation in future clinical trials for human solid cancers.


Subject(s)
Colonic Neoplasms/immunology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Interferon-alpha/genetics , Kidney Neoplasms/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Colonic Neoplasms/therapy , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Graft vs Host Disease/immunology , Kidney Neoplasms/therapy , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphoma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA
20.
Biochem Biophys Res Commun ; 376(2): 299-304, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-18774770

ABSTRACT

We have been investigating the efficacy of an intratumoral interferon (IFN)-alpha gene transfer against solid cancers, and found that when the gene is transduced into the subcutaneous tumors, IFN-alpha concentration is markedly increased in the injected tumor but not in the serum. To explain this effective confinement of IFN-alpha to target tissues, we hypothesized that the extracellular matrix in the tumors interacts with IFN-alpha. In this study, a solid-phase-binding assay and immunoprecipitation demonstrated that the IFN-alpha binds directly to matrix proteins. Immunohistochemical staining showed a co-localization of IFN-alpha with pericellular fibronectin. In addition, matrix-bound IFN-alpha protein transduced intracellular signaling and potentiated its cytotoxic activity, suggesting that the retention of IFN-alpha protein on extracellular matrix is likely to play a role in its in vivo biological activity. The data suggest a therapeutic advantage of the intratumoral IFN-alpha gene transfer over the conventional parenteral therapy both in the safety and efficacy.


Subject(s)
Cytotoxicity, Immunologic , Extracellular Matrix/metabolism , Genetic Therapy , Interferon-alpha/genetics , Interferon-alpha/metabolism , Neoplasms/therapy , Animals , Cell Line, Tumor , Female , Fibronectins/metabolism , Humans , Immunoprecipitation , Interferon-alpha/blood , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/metabolism , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...