Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Life Sci Alliance ; 5(9)2022 09.
Article in English | MEDLINE | ID: mdl-35487695

ABSTRACT

Radiation therapy generates extensive cancer cell death capable of promoting tumor-specific immunity. Within the tumor, conventional dendritic cells (cDCs) are known to carry tumor-associated antigens to the draining lymph node (TdLN) where they initiate T-cell priming. How radiation influences cDC migration is poorly understood. Here, we show that immunological efficacy of radiation therapy is dependent on cDC migration in radioimmunogenic tumors. Using photoconvertible mice, we demonstrate that radiation impairs cDC migration to the TdLN in poorly radioimmunogenic tumors. Comparative transcriptional analysis revealed that cDCs in radioimmunogenic tumors express genes associated with activation of endogenous adjuvant signaling pathways when compared with poorly radioimmunogenic tumors. Moreover, an exogenous adjuvant combined with radiation increased the number of migrating cDCs in these poorly radioimmunogenic tumors. Taken together, our data demonstrate that cDC migration play a critical role in the response to radiation therapy.


Subject(s)
Dendritic Cells , Lymph Nodes , Animals , Mice , T-Lymphocytes
2.
J Hum Lact ; 37(3): 492-498, 2021 08.
Article in English | MEDLINE | ID: mdl-34297643

ABSTRACT

BACKGROUND: The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic has infected over 127 million people worldwide, with almost 2.8 million deaths at the time of writing. Since no lactating individuals were included in initial trials of vaccine safety and efficacy, research on SARS-CoV-2 vaccination in lactating women and the potential transmission of passive immunity to the infant through mother's milk is needed to guide patients, clinicians, and policy makers on whether to recommend immunization during the worldwide effort to curb the spread of this virus. RESEARCH AIMS: (1) To determine whether SARS-CoV-2 specific immunoglobins are found in human milk after vaccination, and (2) to characterize the time course and types of immunoglobulins present. METHODS: A longitudinal cohort study of lactating women (N = 7) who planned to receive both doses of the Pfizer-BioNTech or Moderna SARS-CoV-2 vaccine between December 2020 and January 2021 provided milk samples. These were collected pre-vaccination and at 11 additional timepoints, with the last sample at 14 days after the second dose of vaccine. Samples were analyzed for levels of SARS-CoV-2 specific immunoglobulins A and G (IgA and IgG). RESULTS: We observed significantly elevated levels of SARS-CoV-2 specific IgG and IgA antibodies in human milk beginning approximately 7 days after the initial vaccine dose, with an IgG-dominant response. CONCLUSIONS: Maternal vaccination results in SARS-CoV-2 specific immunoglobulins in human milk that may be protective for infants.


Subject(s)
COVID-19 , SARS-CoV-2 , Breast Feeding , COVID-19 Vaccines , Female , Humans , Infant , Lactation , Longitudinal Studies , Milk, Human , Mothers , Vaccination
3.
Oncotarget ; 12(13): 1201-1213, 2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34194619

ABSTRACT

Surgical resection of head and neck squamous-cell carcinoma (HNSCC) is associated with high rates of local and distant recurrence, partially mitigated by adjuvant therapy. A pre-existing immune response in the patient's tumor is associated with better outcomes following treatment with conventional therapies, but improved options are needed for patients with poor anti-tumor immunity. We hypothesized that local delivery of tumor antigen-specific T-cells into the resection cavity following surgery would direct T-cells to residual antigens in the margins and draining lymphatics and present a platform for T-cell-targeted immunotherapy. We loaded T-cells into a biomaterial that conformed to the resection cavity and demonstrated that it could release T-cells that retained their functional activity in-vitro, and in a HNSCC model in-vivo. Locally delivered T-cells loaded in a biomaterial were equivalent in control of established tumors to intravenous adoptive T-cell transfer, and resulted in the systemic circulation of tumor antigen-specific T-cells as well as local accumulation in the tumor. We demonstrate that adjuvant therapy with anti-PD1 following surgical resection was ineffective unless combined with local delivery of T-cells. These data demonstrate that local delivery of tumor-specific T-cells is an efficient option to convert tumors that are unresponsive to checkpoint inhibitors to permit tumor cures.

4.
J Immunol ; 204(12): 3416-3424, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32341058

ABSTRACT

Radiation therapy is capable of directing adaptive immune responses against tumors by stimulating the release of endogenous adjuvants and tumor-associated Ags. Within the tumor, conventional type 1 dendritic cells (cDC1s) are uniquely positioned to respond to these signals, uptake exogenous tumor Ags, and migrate to the tumor draining lymph node to initiate cross-priming of tumor-reactive cytotoxic CD8+ T cells. In this study, we report that radiation therapy promotes the activation of intratumoral cDC1s in radioimmunogenic murine tumors, and this process fails to occur in poorly radioimmunogenic murine tumors. In poorly radioimmunogenic tumors, the adjuvant polyinosinic-polycytidylic acid overcomes this failure following radiation and successfully drives intratumoral cDC1 maturation, ultimately resulting in durable tumor cures. Depletion studies revealed that both cDC1 and CD8+ T cells are required for tumor regression following combination therapy. We further demonstrate that treatment with radiation and polyinosinic-polycytidylic acid significantly expands the proportion of proliferating CD8+ T cells in the tumor with enhanced cytolytic potential and requires T cell migration from lymph nodes for therapeutic efficacy. Thus, we conclude that lack of endogenous adjuvant release or active suppression following radiation therapy may limit its efficacy in poorly radioimmunogenic tumors, and coadministration of exogenous adjuvants that promote cDC1 maturation and migration can overcome this limitation to improve tumor control following radiation therapy.


Subject(s)
Dendritic Cells/immunology , Neoplasms/immunology , Neoplasms/radiotherapy , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Movement/immunology , Cross-Priming/immunology , Immunotherapy, Adoptive/methods , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C/immunology , Radiotherapy/methods
5.
Int Rev Cell Mol Biol ; 344: 173-214, 2019.
Article in English | MEDLINE | ID: mdl-30798988

ABSTRACT

Nucleic acid sensing pathways have likely evolved as part of a broad pathogen sensing strategy intended to discriminate infectious agents and initiate appropriate innate and adaptive controls. However, in the absence of infectious agents, nucleic acid sensing pathways have been shown to play positive and negative roles in regulating tumorigenesis, tumor progression and metastatic spread. Understanding the normal biology behind these pathways and how they are regulated in malignant cells and in the tumor immune environment can help us devise strategies to exploit nucleic acid sensing to manipulate anti-cancer immunity.


Subject(s)
Immunity , Neoplasms/immunology , Nucleic Acids/metabolism , Animals , Carcinogenesis/pathology , DNA Damage , Humans , Neoplasms/therapy
6.
Cancer Res ; 78(21): 6308-6319, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30224374

ABSTRACT

Surgeons have unique in situ access to tumors enabling them to apply immunotherapies to resection margins as a means to prevent local recurrence. Here, we developed a surgical approach to deliver stimulator of interferon genes (STING) ligands to the site of a purposeful partial tumor resection using a gel-based biomaterial. In a range of head and neck squamous cell carcinoma (HNSCC) murine tumor models, we demonstrate that although control-treated tumors recur locally, tumors treated with STING-loaded biomaterials are cured. The mechanism of tumor control required activation of STING and induction of type I IFN in host cells, not cancer cells, and resulted in CD8 T-cell-mediated cure of residual cancer cells. In addition, we used a novel tumor explant assay to screen individual murine and human HNSCC tumor responses to therapies ex vivo We then utilized this information to personalize the biomaterial and immunotherapy applied to previously unresponsive tumors in mice. These data demonstrate that explant assays identify the diversity of tumor-specific responses to STING ligands and establish the utility of the explant assay to personalize immunotherapies according to the local response.Significance: Delivery of immunotherapy directly to resection sites via a gel-based biomaterial prevents locoregional recurrence of head and neck squamous cell carcinoma. Cancer Res; 78(21); 6308-19. ©2018 AACR.


Subject(s)
Head and Neck Neoplasms/therapy , Immunotherapy/methods , Interferons/chemistry , Squamous Cell Carcinoma of Head and Neck/therapy , Animals , Biocompatible Materials/chemistry , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/surgery , Humans , Ligands , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Neoplasm Recurrence, Local , Neoplasm Transplantation , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/surgery , Wound Healing
7.
Sci Rep ; 8(1): 7012, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29725089

ABSTRACT

Radiation therapy is a source of tumor antigen release that has the potential to serve as an endogenous tumor vaccination event. In preclinical models radiation therapy synergizes with checkpoint inhibitors to cure tumors via CD8 T cell responses. To evaluate the immune response initiated by radiation therapy, we used a range of approaches to block the pre-existing immune response artifact initiated by tumor implantation. We demonstrate that blocking immune responses at tumor implantation blocks development of a tumor-resident antigen specific T cell population and prevents tumor cure by radiation therapy combined with checkpoint immunotherapy. These data demonstrate that this treatment combination relies on a pre-existing immune response to cure tumors, and may not be a solution for patients without pre-existing immunity.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Combined Modality Therapy/methods , Immunologic Factors/administration & dosage , Immunotherapy/methods , Radiotherapy/methods , Animals , Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Disease Models, Animal , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Transplantation, Heterologous , Treatment Outcome
9.
J Immunol ; 200(1): 177-185, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29150567

ABSTRACT

Although prophylactic vaccines provide protective humoral immunity against infectious agents, vaccines that elicit potent CD8 T cell responses are valuable tools to shape and drive cellular immunity against cancer and intracellular infection. In particular, IFN-γ-polarized cytotoxic CD8 T cell immunity is considered optimal for protective immunity against intracellular Ags. Suppressor of cytokine signaling (SOCS)1 is a cross-functional negative regulator of TLR and cytokine receptor signaling via degradation of the receptor-signaling complex. We hypothesized that loss of SOCS1 in dendritic cells (DCs) would improve T cell responses by accentuating IFN-γ-directed immune responses. We tested this hypothesis using a recombinant Listeria monocytogenes vaccine platform that targets CD11c+ DCs in mice in which SOCS1 is selectively deleted in all CD11c+ cells. Unexpectedly, in mice lacking SOCS1 expression in CD11c+ cells, we observed a decrease in CD8+ T cell response to the L. monocytogenes vaccine. NK cell responses were also decreased in mice lacking SOCS1 expression in CD11c+ cells but did not explain the defect in CD8+ T cell immunity. We found that DCs lacking SOCS1 expression were functional in driving Ag-specific CD8+ T cell expansion in vitro but that this process was defective following infection in vivo. Instead, monocyte-derived innate TNF-α and inducible NO synthase-producing DCs dominated the antibacterial response. Thus, loss of SOCS1 in CD11c+ cells skewed the balance of immune response to infection by increasing innate responses while decreasing Ag-specific adaptive responses to infectious Ags.


Subject(s)
Bacterial Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Suppressor of Cytokine Signaling 1 Protein/metabolism , Adaptive Immunity , Animals , CD11c Antigen/metabolism , CD8-Positive T-Lymphocytes/microbiology , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Immunity, Innate , Interferon-gamma/metabolism , Killer Cells, Natural/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Suppressor of Cytokine Signaling 1 Protein/genetics
11.
PLoS One ; 12(11): e0187532, 2017.
Article in English | MEDLINE | ID: mdl-29135982

ABSTRACT

Human papilloma virus positive (HPV+) tumors represent a large proportion of anal, vulvar, vaginal, cervical and head and neck squamous carcinomas (HNSCC) and late stage invasive disease is thought to originate from a premalignant state. Cyclic dinucleotides that activate STimulator of INterferon Genes (STING) have been shown to cause rapid regression of a range of advanced tumors. We aimed to investigate STING ligands as a novel treatment for papilloma. We tested therapies in a spontaneous mouse model of papilloma of the face and anogenital region that histologically resembles human HPV-associated papilloma. We demonstrate that STING ligands cause rapid regression of papilloma, associated with T cell infiltration, and are significantly more effective than Imiquimod, a current immunotherapy for papilloma. In humans, we show that STING is expressed in the basal layer of normal skin and lost during keratinocyte differentiation. We found STING was expressed in all HPV-associated cervical and anal dysplasia and was strongly expressed in the cancer cells of HPV+ HNSCC but not in HPV-unrelated HNSCC. We found no strong association between STING expression and progressive disease in non-HPV oral dysplasia and oral pre-malignancies that are not HPV-related. These data demonstrate that STING is expressed in basal cells of the skin and is retained in HPV+ pre-malignancies and advanced cancers, but not in HPV-unrelated HNSCC. However, using a murine HNSCC model that does not express STING, we demonstrate that STING ligands are an effective therapy regardless of expression of STING by the cancer cells.


Subject(s)
Alphapapillomavirus/isolation & purification , Membrane Proteins/metabolism , Neoplasms/virology , Precancerous Conditions/virology , Animals , Female , Humans , Ligands , Male , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Precancerous Conditions/drug therapy , Precancerous Conditions/metabolism
12.
Int J Radiat Oncol Biol Phys ; 99(2): 362-373, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28871985

ABSTRACT

Novel ligands that target Toll-like receptors and other innate recognition pathways represent a potent strategy for modulating innate immunity to generate antitumor immunity. Although many of the current clinically successful immunotherapies target adaptive T-cell responses, both preclinical and clinical studies suggest that adjuvants have the potential to enhance the scope and efficacy of cancer immunotherapy. Radiation may be a particularly good partner to combine with innate immune therapies, because it is a highly efficient means to kill cancer cells but may fail to send the appropriate inflammatory signals needed to act as an efficient endogenous vaccine. This may explain why although radiation therapy is a highly used cancer treatment, true abscopal effects-regression of disease outside the field without additional systemic therapy-are extremely rare. This review focuses on efforts to combine innate immune stimuli as adjuvants with radiation, creating a distinct and complementary approach from T cell-targeted therapies to enhance antitumor immunity.


Subject(s)
Immunity, Innate , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/radiotherapy , Combined Modality Therapy/methods , DEAD Box Protein 58/metabolism , Humans , Membrane Proteins/metabolism , Neoplasms/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptors, Immunologic , T-Lymphocytes/immunology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
Hepatol Res ; 47(7): 702-714, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27501850

ABSTRACT

AIM: Small, solitary hepatocellular carcinoma is curable with stereotactic radiation or other methods of tumor ablation, however, regional and systemic tumor recurrence occurs in over 70% of patients. Here we describe the ability of immunoradiotherapy to induce an antitumor immune response and delay the growth of tumors in immunocompetent mice. METHODS: A syngeneic hepatocellular carcinoma cell line (Hep-55.1c) was injected directly into the livers of C57BL/6 mice using ultrasound guidance, then tumors were treated with stereotactic radiation using a Small Animal Radiation Research Platform with computed tomography guidance. RESULTS: Delivery of three doses of 250 µg anti-programmed cell death protein-1 (αPD-1) antibody concurrently with 30 Gy stereotactic body radiation therapy in three fractions reduced the growth rate of tumors and improved survival (P < 0.05). Combined treatment was associated with increased CD8+ cytotoxic T cells in the tumor; depletion of CD8 T cells eliminated the efficacy of combined treatment. Combined treatment also induced expression of programmed cell death-1 ligand expression on tumor-infiltrating macrophages, and the tumors grew rapidly after αPD-1 treatment was discontinued. CONCLUSIONS: Tumor response to stereotactic radiation can be augmented by concurrent treatment with αPD-1. The efficacy of this combination therapy was transient, however, and treatment induced markers of adaptive immune resistance. These data are promising, but also indicate that mechanisms of immune resistance will need to be durably overcome for this combination to generate lasting immunity to protect against tumor recurrence.

14.
PLoS One ; 11(6): e0157164, 2016.
Article in English | MEDLINE | ID: mdl-27281029

ABSTRACT

The anecdotal reports of promising results seen with immunotherapy and radiation in advanced malignancies have prompted several trials combining immunotherapy and radiation. However, the ideal timing of immunotherapy with radiation has not been clarified. Tumor bearing mice were treated with 20Gy radiation delivered only to the tumor combined with either anti-CTLA4 antibody or anti-OX40 agonist antibody. Immunotherapy was delivered at a single timepoint around radiation. Surprisingly, the optimal timing of these therapies varied. Anti-CTLA4 was most effective when given prior to radiation therapy, in part due to regulatory T cell depletion. Administration of anti-OX40 agonist antibody was optimal when delivered one day following radiation during the post-radiation window of increased antigen presentation. Combination treatment of anti-CTLA4, radiation, and anti-OX40 using the ideal timing in a transplanted spontaneous mammary tumor model demonstrated tumor cures. These data demonstrate that the combination of immunotherapy and radiation results in improved therapeutic efficacy, and that the ideal timing of administration with radiation is dependent on the mechanism of action of the immunotherapy utilized.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Colorectal Neoplasms/therapy , Immunotherapy , Mammary Neoplasms, Animal/therapy , Receptors, OX40/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation , CTLA-4 Antigen/metabolism , Colorectal Neoplasms/immunology , Combined Modality Therapy , Dose Fractionation, Radiation , Female , Mammary Neoplasms, Animal/immunology , Mice , Mice, Inbred BALB C , Time Factors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Cancer Res ; 76(1): 50-61, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26567136

ABSTRACT

Cytotoxic therapies prime adaptive immune responses to cancer by stimulating the release of tumor-associated antigens. However, the tumor microenvironment into which these antigens are released is typically immunosuppressed, blunting the ability to initiate immune responses. Recently, activation of the DNA sensor molecule STING by cyclic dinucleotides was shown to stimulate infection-related inflammatory pathways in tumors. In this study, we report that the inflammatory pathways activated by STING ligands generate a powerful adjuvant activity for enhancing adaptive immune responses to tumor antigens released by radiotherapy. In a murine model of pancreatic cancer, we showed that combining CT-guided radiotherapy with a novel ligand of murine and human STING could synergize to control local and distant tumors. Mechanistic investigations revealed T-cell-independent and TNFα-dependent hemorrhagic necrosis at early times, followed by later CD8 T-cell-dependent control of residual disease. Clinically, STING was found to be expressed extensively in human pancreatic tumor and stromal cells. Our findings suggest that this novel STING ligand could offer a potent adjuvant for leveraging radiotherapeutic management of pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Membrane Proteins/genetics , Oligonucleotides/pharmacology , Pancreatic Neoplasms/therapy , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/radiotherapy , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Membrane Proteins/biosynthesis , Membrane Proteins/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Targeted Therapy , Oligonucleotides/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/radiotherapy , Random Allocation , Tumor Microenvironment
16.
Oncoimmunology ; 3: e28926, 2014.
Article in English | MEDLINE | ID: mdl-25083323

ABSTRACT

A principal mechanism by which tumors evade immune-mediated elimination is through immunosuppression. Previous approaches to tumor immunotherapy have focused on modifying the immunosuppressive environment with immune checkpoint inhibitors, cytokine therapy, and other modalities with the intent to generate T-cell based anti-tumor immunity. We hypothesized that transformation of the suppressive ovarian cancer microenvironment could be achieved by introduction of the attenuated ΔactA/ΔinlB strain of Listeria monocytogenes. ΔactA/ΔinlB introduced into the microenvironment of the aggressive ID8-Defb29/Vegf-A murine ovarian carcinoma is preferentially phagocytosed by tumor-associated macrophages (TAMs) and reprograms that population from one of suppression to immunostimulation. TAMs in the peritoneum upregulated their co-stimulatory molecules CD80 and CD86, increased transcription of inflammatory cytokines, and downregulated transcription of suppressive effector molecules. Surprisingly, therapeutic benefit was not mediated by T- or NK-cell activity. ΔactA/ΔinlB-induced repolarization of TAMs activated direct tumor cell lysis via Nos2 production of nitric oxide. Modulation of the immunosuppressive nature of the ID8-Defb29/Vegf-A microenvironment, specifically by reprogramming of the TAM suppressive population from M2 to M1 polarization, is critical for our observed immune-mediated survival benefit.

17.
Cancer Immunol Res ; 2(10): 1011-22, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25047233

ABSTRACT

The immune infiltrate in colorectal cancer has been correlated with outcome, such that individuals with higher infiltrations of T cells have increased survival independent of the disease stage. For patients with lower immune infiltrates, overall survival is limited. Because the patients with colorectal cancer studied have received conventional cancer therapies, these data may indicate that the pretreatment tumor environment increases the efficacy of treatments such as chemotherapy, surgery, and radiotherapy. This study was designed to test the hypothesis that an improved immune environment in the tumor at the time of treatment will increase the efficacy of radiotherapy. We demonstrate that inhibition of TGFß using the orally available small-molecule inhibitor SM16 improved the immune environment of tumors in mice and significantly improved the efficacy of subsequent radiotherapy. This effect was not due to changes in radiosensitivity, epithelial-mesenchymal transition, or changes in vascular function in the tumor; rather, this effect was dependent on adaptive immunity and resulted in long-term protective immunity in cured mice. These data demonstrate that immunotherapy is an option to improve the immune status of patients with poor tumor infiltrates and that pretreatment improves the efficacy of radiotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Azabicyclo Compounds/therapeutic use , Colorectal Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Animals , Chemotherapy, Adjuvant/methods , Colorectal Neoplasms/immunology , Colorectal Neoplasms/radiotherapy , Drug Evaluation, Preclinical/methods , Female , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/radiotherapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoadjuvant Therapy/methods , Neoplasm Transplantation , Radiation Tolerance/drug effects , Tumor Cells, Cultured
18.
Nanomedicine ; 10(6): 1273-1285, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24566274

ABSTRACT

Combinatorial use of iron oxide nanoparticles (IONPs) and an alternating magnetic field (AMF) can induce local hyperthermia in tumors in a controlled and uniform manner. Heating B16 primary tumors at 43°C for 30 min activated dendritic cells (DCs) and subsequently CD8(+) T cells in the draining lymph node (dLN) and conferred resistance against rechallenge with B16 (but not unrelated Lewis Lung carcinoma) given 7 days post hyperthermia on both the primary tumor side and the contralateral side in a CD8(+) T cell-dependent manner. Mice with heated primary tumors also resisted rechallenge given 30 days post hyperthermia. Mice with larger heated primary tumors had greater resistance to secondary tumors. No rechallenge resistance occurred when tumors were heated at 45°C. Our results demonstrate the promising potential of local hyperthermia treatment applied to identified tumors in inducing anti-tumor immune responses that reduce the risk of recurrence and metastasis. FROM THE CLINICAL EDITOR: Local heating of tumors via iron oxide NPs and an alternating magnetic field led to activation of anti-cancer CD8 T cells, which resulted in resistance against re-challenge and greater resistance to secondary tumors. Similar local heating-based strategies may become an important weapon in enhancing tumor elimination via a naturally existing but attenuated immune response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hyperthermia, Induced/methods , Magnetite Nanoparticles/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Animals , Cell Line, Tumor , Magnetic Fields , Mice, Inbred C57BL , Neoplasms/pathology
19.
Cancer Res ; 73(13): 3842-51, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23704211

ABSTRACT

Reversing tumor-associated immunosuppression seems necessary to stimulate effective therapeutic immunity against lethal epithelial tumors. Here, we show this goal can be addressed using cps, an avirulent, nonreplicating uracil auxotroph strain of the parasite Toxoplasma gondii (T. gondii), which preferentially invades immunosuppressive CD11c(+) antigen-presenting cells in the ovarian carcinoma microenvironment. Tumor-associated CD11c(+) cells invaded by cps were converted to immunostimulatory phenotypes, which expressed increased levels of the T-cell receptor costimulatory molecules CD80 and CD86. In response to cps treatment of the immunosuppressive ovarian tumor environment, CD11c(+) cells regained the ability to efficiently cross-present antigen and prime CD8(+) T-cell responses. Correspondingly, cps treatment markedly increased tumor antigen-specific responses by CD8(+) T cells. Adoptive transfer experiments showed that these antitumor T-cell responses were effective in suppressing solid tumor development. Indeed, intraperitoneal cps treatment triggered rejection of established ID8-VegfA tumors, an aggressive xenograft model of ovarian carcinoma, also conferring a survival benefit in a related aggressive model (ID8-Defb29/Vegf-A). The therapeutic benefit of cps treatment relied on expression of IL-12, but it was unexpectedly independent of MyD88 signaling as well as immune experience with T. gondii. Taken together, our results establish that cps preferentially invades tumor-associated antigen-presenting cells and restores their ability to trigger potent antitumor CD8(+) T-cell responses. Immunochemotherapeutic applications of cps might be broadly useful to reawaken natural immunity in the highly immunosuppressive microenvironment of most solid tumors.


Subject(s)
Immune Tolerance , Ovarian Neoplasms/immunology , Toxoplasma/immunology , Tumor Microenvironment/immunology , Adoptive Transfer , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/parasitology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , B-Lymphocytes/immunology , CD11c Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Cytokines/metabolism , Female , Humans , Killer Cells, Natural/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Protozoan Vaccines , Spleen/immunology , Spleen/pathology , Tumor Burden/immunology , Tumor Escape , Vascular Endothelial Growth Factor A/metabolism
20.
Integr Biol (Camb) ; 5(1): 159-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22935885

ABSTRACT

Nanotechnology has great potential to produce novel therapeutic strategies that target malignant cells through the ability of nanoparticles to get access to and be ingested by living cells. However its specificity for accumulation in tumors, which is the key factor that determines its efficacy, has always been a challenge. Here we tested a novel strategy to target and treat ovarian cancer, a representative peritoneal cancer, using iron oxide nanoparticles (IONPs) and an alternating magnetic field (AMF). Peritoneal tumors in general are directly accessible to nanoparticles administered intraperitoneally (IP), as opposed to the more commonly attempted intravenous (IV) administration. In addition, tumor-associated immunosuppressive phagocytes, a predominant cell population in the tumor microenvironment of almost all solid tumors, and cells that are critical for tumor progression, are constantly recruited to the tumor, and therefore could possibly function to bring nanoparticles to tumors. Here we demonstrate that tumor-associated peritoneal phagocytes ingest and carry IONPs specifically to tumors and that these specifically delivered nanoparticles can damage tumor cells after IONP-mediated hyperthermia generated by AMF. This illustrates therapeutic possibilities of intraperitoneal (IP) injection of nanoparticles and subsequent ingestion by tumor-associated phagocytes, to directly impact tumors or stimulate antitumor immune responses. This approach could use IONPs combined with AMF as done here, or other nanoparticles with cytotoxic potential. Overall, the data presented here support IP injection of nanoparticles to utilize peritoneal phagocytes as a delivery vehicle in association with IONP-mediated hyperthermia as therapeutic strategies for ovarian and other peritoneal cancers.


Subject(s)
Hyperthermia, Induced/methods , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/therapy , Phagocytes/chemistry , Phagocytes/physiology , Animals , Cell Line, Tumor , Female , Magnetic Field Therapy/methods , Mice , Mice, Inbred C57BL , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...