Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 304
1.
JCI Insight ; 6(13)2021 07 08.
Article En | MEDLINE | ID: mdl-34081628

Existing patient-derived xenograft (PDX) mouse models of solid tumors lack a fully tumor donor-matched, syngeneic, and functional immune system. We developed a model that overcomes these limitations by engrafting lymphopenic recipient mice with a fresh, undisrupted piece of solid tumor, whereby tumor-infiltrating lymphocytes (TILs) persisted in the recipient mice for several weeks. Successful tumor engraftment was achieved in 83% to 89% of TIL-PDX mice, and these were seen to harbor exhausted immuno-effector as well as functional immunoregulatory cells persisting for at least 6 months postengraftment. Combined treatment with interleukin-15 stimulation and immune checkpoint inhibition resulted in complete or partial tumor response in this model. Further, depletion of cytotoxic T lymphocytes and/or natural killer cells before combined immunotherapy revealed that both cell types were required for maximal tumor regression. Our TIL-PDX model provides a valuable resource for powerful mechanistic and therapeutic studies in solid tumors.


Heterografts , Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasm Transplantation , Neoplasms , T-Lymphocytes, Cytotoxic/immunology , Adjuvants, Immunologic/pharmacology , Animals , Disease Models, Animal , Heterografts/immunology , Heterografts/pathology , Humans , Immune Checkpoint Inhibitors/pharmacology , Interleukin-15/metabolism , Mice , Neoplasm Transplantation/immunology , Neoplasm Transplantation/methods , Neoplasms/immunology , Neoplasms/therapy , Transplantation, Heterologous/methods , Xenograft Model Antitumor Assays/methods
2.
Cancer Res ; 80(15): 3088-3100, 2020 08 01.
Article En | MEDLINE | ID: mdl-32366475

IL26 is a unique amphipathic member of the IL10 family of cytokines that participates in inflammatory signaling through a canonical receptor pathway. It also directly binds DNA to facilitate cellular transduction and intracellular inflammatory signaling. Although IL26 has almost no described role in cancer, our in vivo screen of inflammatory and cytokine pathway genes revealed IL26 to be one of the most significant inflammatory mediators of mammary engraftment and lung metastatic growth in triple-negative breast cancer (TNBC). Examination of human breast cancers demonstrated elevated IL26 transcripts in TNBC specimens, specifically in tumor cells as well as in Th17 CD4+ T cells within clinical TNBC specimens. IL26 did not have an autocrine effect on human TNBC cells, but rather its effect on engraftment and growth in vivo required neutrophils. IL26 enhanced mouse-derived DNA induction of inflammatory cytokines, which were collectively important for mammary and metastatic lung engraftment. To neutralize this effect, we developed a novel IL26 vaccine to stimulate antibody production and suppress IL26-enhanced engraftment in vivo, suggesting that targeting this inflammatory amplifier could be a unique means to control cancer-promoting inflammation in TNBC and other autoimmune diseases. Thus, we identified IL26 as a novel key modulator of TNBC metastasis and a potential therapeutic target in TNBC as well as other diseases reliant upon IL26-mediated inflammatory stimulation. SIGNIFICANCE: These findings identify IL26 as a unique, clinically relevant, inflammatory amplifier that enhances TNBC engraftment and dissemination in association with neutrophils, which has potential as a therapeutic target. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/15/3088/F1.large.jpg.


Cell Adhesion , Interleukins/physiology , Neoplasm Transplantation , Neutrophils/physiology , Triple Negative Breast Neoplasms/pathology , Animals , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cells, Cultured , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Disease Progression , Extracellular Traps/drug effects , Extracellular Traps/metabolism , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Inflammation Mediators/pharmacology , Inflammation Mediators/physiology , Interleukins/genetics , Interleukins/pharmacology , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasm Transplantation/immunology , Neoplasm Transplantation/pathology , Neutrophils/pathology , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
4.
Mol Cancer Res ; 17(9): 1801-1814, 2019 09.
Article En | MEDLINE | ID: mdl-31147393

Disseminating epithelial ovarian cancer cells often become assembled into spheroids prior to their arrival at metastatic sites within the peritoneal cavity. Although epithelial ovarian carcinoma (EOC) is the deadliest gynecologic malignancy, the mechanisms regulating formation and metastatic potential of spheroids are poorly understood. We show that expression of a cell surface glycoprotein CD44 is an important contributing factor for spheroid formation and spheroid adhesion to mesothelial cells, and its loss impairs mesenteric metastasis. In contrast, loss of CD44 resulted in significant increase of tumor burden at several locoregional sites, including liver, and unleashed distant metastases to the thoracic cavity. Altogether our studies suggest that CD44 regulates metastatic progression of EOC in an organ-specific manner. IMPLICATIONS: Expression of CD44 promotes spheroid formation, mesothelial adhesion, and formation of mesenteric metastasis, but it suppresses development of metastasis to several peritoneal sites, including liver, and the thoracic cavity.


Carcinoma, Ovarian Epithelial/pathology , Hyaluronan Receptors/metabolism , Neoplasm Transplantation/pathology , Spheroids, Cellular/transplantation , Animals , Carcinoma, Ovarian Epithelial/immunology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Metastasis , Neoplasm Transplantation/immunology , Organ Specificity , Ovarian Neoplasms , Spheroids, Cellular/cytology , Spheroids, Cellular/immunology , Up-Regulation
5.
Oncotarget ; 8(2): 2053-2068, 2017 Jan 10.
Article En | MEDLINE | ID: mdl-28008146

Correlative studies from checkpoint inhibitor trials have indicated that better understanding of human leukocytic trafficking into the human tumor microenvironment can expedite the translation of future immune-oncologic agents. In order to directly characterize signaling pathways that can regulate human leukocytic trafficking into the tumor, we have developed a completely autologous xenotransplantation method to reconstitute the human tumor immune microenvironment in vivo. We were able to genetically mark the engrafted CD34+ bone marrow cells as well as the tumor cells, and follow the endogenous leukocytic infiltration into the autologous tumor. To investigate human tumor intrinsic factors that can potentially regulate the immune cells in our system, we silenced STAT3 signaling in the tumor compartment. As expected, STAT3 signaling suppression in the tumor compartment in these autologously reconstituted humanized mice showed increased tumor infiltrating lymphocytes and reduction of arginase-1 in the stroma, which were associated with slower tumor growth rate. We also used this novel system to characterize human myeloid suppressor cells as well as to screen novel agents that can alter endogenous leukocytic infiltration into the tumor. Taken together, we present a valuable method to study individualized human tumor microenvironments in vivo without confounding allogeneic responses.


Lymphocytes, Tumor-Infiltrating/pathology , Neoplasm Transplantation/immunology , Neoplasm Transplantation/pathology , Neoplasms/immunology , Neoplasms/pathology , Tumor Microenvironment/immunology , Animals , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , HLA-A2 Antigen/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Heterografts , Humans , Lymphocytes, Tumor-Infiltrating/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Squamous Cell Carcinoma of Head and Neck , Transgenes , Transplantation, Autologous
6.
Med Sci Monit ; 22: 3113-23, 2016 Sep 02.
Article En | MEDLINE | ID: mdl-27587310

BACKGROUND Transplantation with allogeneic cells has become a promising modality for cancer therapy, which can induce graft-versus-tumor (GVT) effect. This study was aimed at assessing the safety, efficacy, and tissue type GVT (tGVT) response of transplantation with allogeneic skin tumors to treat chemically-induced skin tumors in mice. MATERIAL AND METHODS FVB/N and ICR mice were exposed topically to chemicals to induce skin tumors. Healthy ICR mice were transplanted with allogeneic skin tumors from FVB/N mice to test the safety. The tumor-bearing ICR mice were transplanted with, or without, allogeneic skin tumors to test the efficacy. The body weights (BW), body condition scores (BCS), tumor volumes in situ, metastasis tumors, overall survival, and serum cytokines were measured longitudinally. RESULTS Transplantation with no more than 0.03 g allogeneic skin tumors from FVB/N mice to healthy ICR mice was safe. After transplantation with allogeneic skin tumors to treat tumor-bearing mice, it inhibited the growth of tumors slightly at early stage, accompanied by fewer metastatic tumors at 24 days after transplantation (21.05% vs. 47.37%), while there were no statistically significant differences in the values of BW, BCS, tumor volumes in situ, metastasis tumors, and overall survival between the transplanted and non-transplanted groups. The levels of serum interleukin (IL)-2 were significantly reduced in the controls (P<0.05), but not in the recipients, which may be associated with the tGVT response. CONCLUSIONS Our results suggest that transplantation with allogeneic skin tumors is a safe treatment in mice, which can induce short-term tGVT response mediated by IL-2.


Graft vs Tumor Effect/immunology , Neoplasm Transplantation/immunology , Neoplasm Transplantation/methods , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Animals , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/therapy , Female , Interleukin-2/metabolism , Mice , Mice, Inbred ICR , Random Allocation , Transplantation, Homologous/methods
7.
Hum Cell ; 29(2): 58-66, 2016 Apr.
Article En | MEDLINE | ID: mdl-26857856

Cancer vaccines serve as a promising clinical immunotherapeutic strategy that help to trigger an effective and specific antitumor immune response compared to conventional therapies. However, poor immunogenicity of tumor cells remains a major obstacle for clinical application, and developing new methods to modify the immunogenicity of tumor cells may help to improve the clinical outcome of cancer vaccines. 4T1 mouse breast cancer cell line has been known as poorly immunogenic and highly metastatic cell line. Using this model, we identified a sub cell line of 4T1-designated as 4T1-Sapporo (4T1-S)-which shows immunogenic properties when used as a vaccine against the same line. In 4T1-S-vaccinated mice, subcutaneous injection of 4T1-S resulted in an antitumor inflammatory response represented by significant enlargement of draining lymph nodes, accompanied with increased frequencies of activated CD8 T cells and a subpopulation of myeloid cells. Additionally, 4T1-S vaccine was ineffective to induce tumor rejection in nude mice, which importantly indicate that 4T1-S vaccine rely on T cell response to induce tumor rejection. Further analysis to identify mechanisms that control tumor immunogenicity in this model may help to develop new methods for improving the efficacies of clinical cancer vaccines.


Breast Neoplasms/immunology , Animals , Breast Neoplasms/pathology , Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Female , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation/immunology
8.
FEBS Lett ; 590(1): 68-75, 2016 Jan.
Article En | MEDLINE | ID: mdl-26787462

Mammalian ste-20 like kinase Mst1 plays important roles during apoptosis, proliferation, cell polarity, and migration. Here, we report a novel role of Mst1 for cytotoxic T-cell responses and tumor suppression. The defect of Mst1 caused decreased levels of FoxO, and promoted cytotoxicity in vitro. Mst1(-/-) cytotoxic T cells also exhibited enhanced T-bet expression that was associated with elevated expression levels of IFNγ and granzyme B. Moreover, Mst1(-/-) cytotoxic T cells suppressed tumor growth in vivo. The data suggest that Mst1 inhibits cytotoxicity via T-bet suppression by FoxO1 and FoxO3a. Thus, Mst1 is a potential therapeutic target for tumor immunotherapy.


Gene Expression Regulation, Neoplastic , Immunologic Surveillance , Lymphocyte Activation , Lymphoma, T-Cell/metabolism , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Transplantation/immunology , Neoplasm Transplantation/pathology , Protein Serine-Threonine Kinases/genetics , Serine-Threonine Kinase 3 , Specific Pathogen-Free Organisms , Survival Analysis , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Tumor Burden
9.
Nature ; 521(7550): 99-104, 2015 May 07.
Article En | MEDLINE | ID: mdl-25924063

Whereas cancers grow within host tissues and evade host immunity through immune-editing and immunosuppression, tumours are rarely transmissible between individuals. Much like transplanted allogeneic organs, allogeneic tumours are reliably rejected by host T cells, even when the tumour and host share the same major histocompatibility complex alleles, the most potent determinants of transplant rejection. How such tumour-eradicating immunity is initiated remains unknown, although elucidating this process could provide the basis for inducing similar responses against naturally arising tumours. Here we find that allogeneic tumour rejection is initiated in mice by naturally occurring tumour-binding IgG antibodies, which enable dendritic cells (DCs) to internalize tumour antigens and subsequently activate tumour-reactive T cells. We exploited this mechanism to treat autologous and autochthonous tumours successfully. Either systemic administration of DCs loaded with allogeneic-IgG-coated tumour cells or intratumoral injection of allogeneic IgG in combination with DC stimuli induced potent T-cell-mediated antitumour immune responses, resulting in tumour eradication in mouse models of melanoma, pancreas, lung and breast cancer. Moreover, this strategy led to eradication of distant tumours and metastases, as well as the injected primary tumours. To assess the clinical relevance of these findings, we studied antibodies and cells from patients with lung cancer. T cells from these patients responded vigorously to autologous tumour antigens after culture with allogeneic-IgG-loaded DCs, recapitulating our findings in mice. These results reveal that tumour-binding allogeneic IgG can induce powerful antitumour immunity that can be exploited for cancer immunotherapy.


Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Immunoglobulin G/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Neoplasm/administration & dosage , CD40 Antigens/metabolism , Disease Models, Animal , Female , Immunoglobulin G/administration & dosage , Isoantibodies/administration & dosage , Isoantibodies/immunology , Lymphocyte Activation/immunology , Male , Mice , Neoplasm Metastasis , Neoplasm Transplantation/immunology , Neoplasms/pathology , Receptors, IgG/immunology , Tumor Necrosis Factor-alpha/immunology
11.
Exp Anim ; 64(2): 181-90, 2015.
Article En | MEDLINE | ID: mdl-25740629

Patient-derived xenografts (PDXs) of tumors are increasingly becoming important tools for translational research in oncology. The NOD.Cg-Prkdc(scid) Il2rg(tm1Sug)/Jic (NOG) mouse is an efficient host for PDXs. Thus as a basis for future development of methods to obtain PDXs from various disease types, we have studied the factors that affect the outcome of transplantation of human colorectal cancer in NOG mice. Of the original donor cases examined, 73% had successful engraftment. The outcome of donor-matched tissues was consistent in most cases, and was thought to show that the condition of the host did not affect engraftment. Next we analyzed the tumor aggressiveness in terms of histology grade of the original tumor and found that they were related to engraftment. Detailed histopathological examination of the transplanted tissues strongly indicated that lymphocytes engrafted with the tumor cells affect engraftment. As a factor related to transplantation of lymphocytes, we studied the human IgG concentration in the serum of tumor-bearing mice, but there was no tendency for higher concentrations to result in unsuccessful engraftment. Finally, we studied the type, density and location of T cells in the original donor tissue to determine the immune contexture and found that the unsuccessful engraftment cases tended to have an adequate or coordinated immune contexture compared to successful engraftment cases. From these results, we concluded that the aggressiveness and the T cell infiltration of the original tumor affect the outcome of transplantation in the NOG mouse.


Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Heterografts , Neoplasm Transplantation , T-Lymphocytes/immunology , Tissue Donors , Animals , Cell Transformation, Neoplastic , Heterografts/immunology , Humans , Immunoglobulin G/blood , Lymphocyte Transfusion , Lymphocytes/pathology , Mice, Transgenic , Neoplasm Transplantation/immunology
12.
Cancer Biother Radiopharm ; 30(4): 182-6, 2015 May.
Article En | MEDLINE | ID: mdl-25763836

C3Hf/HZgr mice were transplanted with SCCVII carcinoma cells and treated with Newcastle disease virus (NDV). The treatment slows down the growth of transplanted tumor. Furthermore, by using specific monoclonal antibodies, the frequencies of CD4+, CD8+, and CD4+CD25+ T lymphocytes were determined in the spleen of tumorous mice at particular times following tumor transplantation and/or NDV application. The incidence of lymphocytes CD4+ and CD8+ decreased and of CD4+CD25+ increased in the spleen of mice during the time following tumor transplantation. However, the frequency of regulatory CD4+CD25+ T lymphocytes in the spleen is very low, while CD4+ and CD8+ increased to normal level following intraperitoneal (i.p.) NDV injection in tumor-bearing mice. Thus, besides directly destroying transplanted tumor, NDV seems to be involved against growing tumor by reducing the frequency of regulatory T lymphocytes maintaining the frequency of CD4+ and CD8+ T lymphocytes within the control values pointing to its role in immunomodulation.


CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasm Transplantation/immunology , Newcastle disease virus/immunology , Spleen/immunology , Spleen/virology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/virology , Incidence , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C3H , T-Lymphocytes, Regulatory/virology
13.
Methods Mol Biol ; 1267: 439-56, 2015.
Article En | MEDLINE | ID: mdl-25636483

When a tumor evolves, there is constant crosstalk between the transformed cells and cells of the immune system. Transplantation of well-established tumor cell lines into genetically engineered mice is a valuable tool to study the contribution of a gene of interest to tumor surveillance. These methods bear several advantages: first, such cell lines are well characterized; second, much data for reference exist; and third, the impact of the immune system can be separated from tumor cell intrinsic effects. Here, we provide protocols for tumor cell transplantations to address the role of a specific gene product in tumor surveillance. We furthermore describe several approaches to define the impact of natural killer cells and T cells, such as cell depletion and adoptive transfer experiments or use of different genetically modified mice.


Cell Transplantation/methods , Immunologic Surveillance , Neoplasm Transplantation/immunology , Neoplasm Transplantation/methods , Neoplasms/immunology , Neoplasms/pathology , Adoptive Transfer , Animals , Biomarkers/metabolism , Bone Marrow/metabolism , Cell Line, Tumor , Gene Knockout Techniques , Mice , Mice, Transgenic , Neoplasm Transplantation/pathology , Neoplasms/genetics
14.
Mol Ther ; 22(3): 634-644, 2014 Mar.
Article En | MEDLINE | ID: mdl-24445938

Cell therapy based on alloreactivity has completed clinical proof of concept against hematological malignancies. However, the efficacy of alloreactivity as a therapeutic approach to treat solid tumors is unknown. Using cell culture and animal models, we aimed to investigate the efficacy and safety of allogeneic suicide gene-modified killer cells as a cell-based therapy for hepatocellular carcinoma (HCC), for which treatment options are limited. Allogeneic killer cells from healthy donors were isolated, expanded, and phenotypically characterized. Antitumor cytotoxic activity and safety were studied using a panel of human or murine HCC cell lines engrafted in immunodeficient or immunocompetent mouse models. Human allogeneic suicide gene-modified killer cells (aSGMKCs) exhibit a high, rapid, interleukin-2-dependent, and non-major histocompatibility complex class I-restricted in vitro cytotoxicity toward human hepatoma cells, mainly mediated by natural killer (NK) and NK-like T cells. In vivo evaluation of this cell therapy product demonstrates a marked, rapid, and sustained regression of HCC. Preferential liver homing of effector cells contributed to its marked efficacy. Calcineurin inhibitors allowed preventing rejection of allogeneic lymphocytes by the host immune system without impairing their antitumor activity. Our results demonstrate proof of concept for aSGMKCs as immunotherapy for HCC and open perspectives for the clinical development of this approach.


Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Cytotoxicity, Immunologic , Liver Neoplasms/immunology , Neoplasm Transplantation/immunology , T-Lymphocytes/immunology , Transplantation, Homologous/methods , Animals , Cell Line, Tumor , HeLa Cells , Humans , Immunotherapy, Adoptive , Liver Neoplasms/therapy , Mice , Mice, Inbred C57BL , Mice, SCID , T-Lymphocytes/transplantation
15.
J Clin Invest ; 123(12): 5351-60, 2013 Dec.
Article En | MEDLINE | ID: mdl-24231354

Ionizing radiation (IR) and germline mutations in the retinoblastoma tumor suppressor gene (RB1) are the strongest risk factors for developing osteosarcoma. Recapitulating the human predisposition, we found that Rb1+/- mice exhibited accelerated development of IR-induced osteosarcoma, with a latency of 39 weeks. Initial exposure of osteoblasts to carcinogenic doses of IR in vitro and in vivo induced RB1-dependent senescence and the expression of a panel of proteins known as senescence-associated secretory phenotype (SASP), dominated by IL-6. RB1 expression closely correlated with that of the SASP cassette in human osteosarcomas, and low expression of both RB1 and the SASP genes was associated with poor prognosis. In vivo, IL-6 was required for IR-induced senescence, which elicited NKT cell infiltration and a host inflammatory response. Mice lacking IL-6 or NKT cells had accelerated development of IR-induced osteosarcomas. These data elucidate an important link between senescence, which is a cell-autonomous tumor suppressor response, and the activation of host-dependent cancer immunosurveillance. Our findings indicate that overcoming the immune response to senescence is a rate-limiting step in the formation of IR-induced osteosarcoma.


Bone Neoplasms/immunology , Cellular Senescence/physiology , Natural Killer T-Cells/immunology , Neoplasms, Radiation-Induced/immunology , Osteosarcoma/immunology , Retinoblastoma Protein/physiology , Animals , Bone Neoplasms/etiology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Calcium Radioisotopes/toxicity , Cytokines/physiology , Genes, Retinoblastoma , Humans , Immunologic Surveillance , Intercellular Signaling Peptides and Proteins/physiology , Interleukin-6/deficiency , Interleukin-6/physiology , Mice , Mice, Inbred C57BL , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Neoplasm Transplantation/immunology , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Osteoblasts/pathology , Osteosarcoma/etiology , Osteosarcoma/genetics , Osteosarcoma/pathology , Phenotype , Prognosis , RNA Interference , Retinoblastoma Protein/antagonists & inhibitors
16.
J Invest Dermatol ; 133(1): 27-30, 2013 Jan.
Article En | MEDLINE | ID: mdl-22854621

Photoimmunology evolved from experiments carried out in the 1970s on the immunology of cancer. In studying the antigenic properties of skin cancers induced in mice by UV radiation, I found that most of these tumors failed to grow when transplanted into normal, syngeneic mice but grew progressively in immunosuppressed mice. Thus, these UV-induced skin cancers were highly antigenic. The critical question was, how can these antigenic skin cancers escape immune rejection in their primary host? The answer was that exposing their skin to UV radiation prevented mice from triggering an immune response against their tumors. The failure to reject these tumors was owing to the development of UV tumor-specific regulatory T cells during the course of irradiation. In unraveling the mechanisms of this effect of UV, much has been learned about the immunology of the skin, including the function of Langerhans cells, the migration of immune cells in skin, the role of antigen-presenting cells in directing the immune response, and the role of keratinocytes as producers of immunological mediators. Thus, photoimmunology helped demonstrate that skin is an important immunological organ, and that the immune system can be influenced by the external environment via the skin.


Neoplasms, Radiation-Induced/immunology , Skin Neoplasms/immunology , Skin/immunology , Skin/radiation effects , Ultraviolet Rays/adverse effects , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/radiation effects , Graft Rejection/immunology , Humans , Immunocompromised Host/immunology , Immunocompromised Host/radiation effects , Keratinocytes/immunology , Keratinocytes/radiation effects , Mice , Neoplasm Transplantation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects
17.
Autoimmunity ; 46(5): 317-22, 2013 Aug.
Article En | MEDLINE | ID: mdl-23194071

The presence of a solid tumor is the result of a complex balance between rejection, tolerance and regeneration in which the interactions of tumor cells with cells of the host immune system contribute strongly to the final outcome. Here we report on a model where lethally UVB-irradiated cells cause accelerated growth of viable tumor cells in vitro and in allogeneic immune competent mice. UVB-irradiated tumor cells alone did not form tumors and failed to induce tolerance for a second challenge with the same allogeneic tumor. Our data show an important role for dying cells in promoting accelerated tumor cell growth of a small number of viable tumor cells in a large inoculum of UVB-irradiated tumor cells. This occurs when viable and dying/dead tumor cells are in close proximity, suggesting that mobile factors contribute to growth promotion. The anti-inflammatory and growth promoting properties of apoptotic cells are based on several independent effects. UVB-irradiated apoptotic cells directly release a growth promoting activity and clearance by macrophages of apoptotic cells is accompanied by the secretion of IL10, TGFß, and PGE2. Growth promotion is even observed with dying heterologous cells implying a conserved mechanism. Future experiments should focus on the effects of dying tumor cells generated in vivo on the outgrowth of surviving tumor cells which is prone to have implications for cancer therapy.


Apoptosis/immunology , Immunocompetence/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Neoplastic Stem Cells/transplantation , Ultraviolet Rays , Animals , Apoptosis/radiation effects , Cell Death/immunology , Cell Death/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Disease Models, Animal , Immunocompetence/radiation effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation/immunology , Neoplasm Transplantation/pathology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/radiation effects
18.
Exp Mol Pathol ; 93(3): 294-301, 2012 Dec.
Article En | MEDLINE | ID: mdl-23018130

Antigen-specific CD8 suppressor T cells (CD8(+) Ts) are adaptive regulatory T cells that are induced in vivo and in vitro by chronic antigenic stimulation of human T cells. CD8(+) Ts induce the upregulation of the inhibitory receptors ILT3 and ILT4 on monocytes and dendritic cells rendering these antigen presenting cells (APCs) tolerogenic. Tolerogenic APCs induce CD4(+) T helper anergy and elicit the differentiation of CD4(+) and CD8(+) T regulatory/suppressor cells. Overexpression of membrane ILT3 in APC results in inhibition of NF-κB activation, transcription of inflammatory cytokines and costimulatory molecules. Soluble ILT3-Fc which contains only the extracellular, Ig-like domain linked to mutated IgG1 Fc, is strongly immunosuppressive. ILT3-Fc, induces the differentiation of human CD8(+) Ts which inhibit CD4(+) Th and CD8(+) CTL effector function both in vitro and in vivo. The acquisition of Ts' function by primed CD8(+) T cells treated with ILT3-Fc was demonstrated to be the effect of the significant upregulation of BCL6, a transcriptional repressor of IL-2, IFN-gamma, IL-5 and granzyme B. The upregulated expression of BCL6, SOCS1 and DUSP10 is integral to the signature of ILT3-Fc-induced CD8(+) Ts. These genes are known inhibitors of cytokine production and TCR signaling and are targeted by miRNAs which are suppressed by ILT3-Fc. ILT3-Fc induces tolerance to allogeneic human islets and reverses rejection after its onset in a humanized NOD/SCID mouse model. Based on these findings we postulate that ILT3-Fc may become an important new agent for treatment of autoimmunity and transplant rejection.


CD8-Positive T-Lymphocytes/immunology , Cell Membrane/immunology , Receptors, Cell Surface/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Differentiation/immunology , Cells, Cultured , Clonal Anergy/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Graft Rejection/drug therapy , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/pharmacology , Membrane Glycoproteins , Mice , Mice, Inbred NOD , Mice, SCID , Models, Animal , Monocytes/drug effects , Monocytes/immunology , Neoplasm Transplantation/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-bcl-6 , Receptors, Cell Surface/genetics , Receptors, Immunologic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Repressor Proteins/genetics , Repressor Proteins/immunology , Signal Transduction , Transplantation Tolerance/immunology
19.
J Immunol ; 189(7): 3759-66, 2012 Oct 01.
Article En | MEDLINE | ID: mdl-22956585

CTLA-4 is a surface receptor on activated T cells that delivers an inhibitory signal, serving as an immune checkpoint. Treatment with anti-CTLA-4 Abs can induce clinical responses to different malignancies, but the nature of the induced Ag-specific recognition is largely unknown. Using microarrays spotted with >8000 human proteins, we assessed the diversity of Ab responses modulated by treatment with CTLA-4 blockade and GM-CSF. We find that advanced prostate cancer patients who clinically respond to treatment also develop enhanced Ab responses to a higher number of Ags than nonresponders. These induced Ab responses targeted Ags to which preexisting Abs are more likely to be present in the clinical responders compared with nonresponders. The majority of Ab responses are patient-specific, but immune responses against Ags shared among clinical responders are also detected. One of these shared Ags is PAK6, which is expressed in prostate cancer and to which CD4(+) T cell responses were also induced. Moreover, immunization with PAK6 can be both immunogenic and protective in mouse tumor models. These results demonstrate that immune checkpoint blockade modulates Ag-specific responses to both individualized and shared Ags, some of which can mediate anti-tumor responses.


Antibody Diversity , Autoantigens/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Autoantibodies/biosynthesis , Autoantibodies/therapeutic use , Feasibility Studies , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Ipilimumab , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation/immunology , Neoplasm Transplantation/pathology , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
20.
J Immunol ; 189(5): 2110-7, 2012 Sep 01.
Article En | MEDLINE | ID: mdl-22815287

Cancer-induced immunosuppression is a major problem reducing antitumor effects of immunotherapies, but its molecular mechanism has not been well understood. We evaluated immunosuppressive roles of activated Wnt/ß-catenin pathways in human melanoma for dendritic cells (DCs) and CTLs. IL-10 expression was associated with ß-catenin accumulation in human melanoma cell lines and tissues and was induced by direct ß-catenin/TCF binding to the IL-10 promoter. Culture supernatants from ß-catenin-accumulated melanoma have activities to impair DC maturation and to induce possible regulatory DCs. Those immunosuppressive culture supernatant activities were reduced by knocking down ß-catenin in melanoma cells, partly owing to downregulation of IL-10. Murine splenic and tumor-infiltrating DCs obtained from nude mice implanted with human mutant ß-catenin-overexpressed melanoma cells had less ability to activate T cells than did DCs from mice with control melanoma cells, showing in vivo suppression of DCs by activated Wnt/ß-catenin signaling in human melanoma. This in vivo DC suppression was restored by the administration of a ß-catenin inhibitor, PKF115-584. ß-catenin-overexpressed melanoma inhibited IFN-γ production by melanoma-specific CTLs in an IL-10-independent manner and is more resistant to CTL lysis in vitro and in vivo. These results indicate that Wnt/ß-catenin pathways in human melanoma may be involved in immunosuppression and immunoresistance in both induction and effector phases of antitumor immunoresponses partly through IL-10 production, and they may be attractive targets for restoring immunocompetence in patients with Wnt/ß-catenin-activated melanoma.


Immune Tolerance , Melanoma/immunology , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/immunology , beta Catenin/genetics , Animals , Cells, Cultured , Coculture Techniques , Disease Resistance/genetics , Disease Resistance/immunology , HEK293 Cells , HeLa Cells , Humans , Immune Tolerance/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Mutation , Neoplasm Transplantation/immunology , Tumor Cells, Cultured , beta Catenin/deficiency
...