Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 9(5)2021 05.
Article in English | MEDLINE | ID: mdl-33958486

ABSTRACT

BACKGROUND: Clinical studies have linked usage of progestins (synthetic progesterone [P4]) to breast cancer risk. However, little is understood regarding the role of native P4, signaling through the progesterone receptor (PR), in breast tumor formation. Recently, we reported a link between PR and immune signaling pathways, showing that P4/PR can repress type I interferon signaling pathways. Given these findings, we sought to investigate whether P4/PR drive immunomodulation in the mammary gland and promote tumor formation. METHODS: To determine the effect of P4 on immune cell populations in the murine mammary gland, mice were treated with P4 or placebo pellets for 21 days. Immune cell populations in the mammary gland, spleen, and inguinal lymph nodes were subsequently analyzed by flow cytometry. To assess the effect of PR overexpression on mammary gland tumor development as well as immune cell populations in the mammary gland, a transgenic mouse model was used in which PR was overexpressed throughout the entire mouse. Immune cell populations were assessed in the mammary glands, spleens, and inguinal lymph nodes of 6-month-old transgenic and control mice by flow cytometry. Transgenic mice were also monitored for mammary gland tumor development over a 2-year time span. Following development of mammary gland tumors, immune cell populations in the tumors and spleens of transgenic and control mice were analyzed by flow cytometry. RESULTS: We found that mice treated with P4 exhibited changes in the mammary gland indicative of an inhibited immune response compared with placebo-treated mice. Furthermore, transgenic mice with PR overexpression demonstrated decreased numbers of immune cell populations in their mammary glands, lymph nodes, and spleens. On long-term monitoring, we determined that multiparous PR-overexpressing mice developed significantly more mammary gland tumors than control mice. Additionally, tumors from PR-overexpressing mice contained fewer infiltrating immune cells. Finally, RNA sequencing analysis of tumor samples revealed that immune-related gene signatures were lower in tumors from PR-overexpressing mice as compared with control mice. CONCLUSION: Together, these findings offer a novel mechanism of P4-driven mammary gland tumor development and provide rationale in investigating the usage of antiprogestin therapies to promote immune-mediated elimination of mammary gland tumors.


Subject(s)
Breast Neoplasms/chemically induced , Cell Transformation, Neoplastic/chemically induced , Mammary Glands, Animal/drug effects , Progesterone/administration & dosage , Receptors, Progesterone/agonists , Tumor Escape/drug effects , Tumor Microenvironment/immunology , Adaptive Immunity/drug effects , Animals , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Drug Implants , Female , Galectin 4/genetics , Galectin 4/metabolism , Immunity, Innate/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mammary Glands, Animal/immunology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice, Transgenic , Ovariectomy , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Signal Transduction , Time Factors , Tumor Burden/drug effects
2.
Mol Cancer Ther ; 17(11): 2320-2328, 2018 11.
Article in English | MEDLINE | ID: mdl-30108133

ABSTRACT

Antitumor alkyl phospholipid (APL) analogs comprise a group of structurally related molecules with remarkable tumor selectivity. Some of these compounds have shown radiosensitizing capabilities. CLR127 is a novel, clinical-grade antitumor APL ether analog, a subtype of synthetic APL broadly targeting cancer cells with limited uptake in normal tissues. The purpose of this study was to investigate the effect of CLR127 to modulate radiation response across several adult and pediatric cancer types in vitro as well as in murine xenograft models of human prostate adenocarcinoma, neuroblastoma, Ewing sarcoma, and rhabdomyosarcoma. In vitro, CLR127 demonstrated selective uptake in cancer cells compared to normal cells. In cancer cells, CLR127 treatment prior to radiation significantly decreased clonogenic survival in vitro, and led to increased radiation-induced double-stranded DNA (dsDNA) breakage compared with radiation alone, which was not observed in normal controls. In animal models, CLR127 effectively increased the antitumor response to fractionated radiotherapy and led to delayed tumor regrowth at potentially clinically achievable doses. In conclusion, our study highlights the ability of CLR127 to increase radiation response in several cancer types. Given almost universal uptake of CLR127 in malignant cells, future research should test whether the observed effects can be extended to other tumor types. Our data provide a strong rationale for clinical testing of CLR127 as a tumor-targeted radiosensitizing agent. Mol Cancer Ther; 17(11); 2320-8. ©2018 AACR.


Subject(s)
Neoplasms/pathology , Phospholipid Ethers/pharmacology , Radiation Tolerance , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Clone Cells , DNA Damage , Histones/metabolism , Humans , Mice, Nude , Radiation Tolerance/drug effects , Radiation Tolerance/radiation effects , X-Rays , Xenograft Model Antitumor Assays
3.
Cancer Immunol Res ; 6(7): 825-834, 2018 07.
Article in English | MEDLINE | ID: mdl-29748391

ABSTRACT

In situ vaccination is an emerging cancer treatment strategy that uses local therapies to stimulate a systemic antitumor immune response. We previously reported an in situ vaccination effect when combining radiation (RT) with intratumor (IT) injection of tumor-specific immunocytokine (IC), a fusion of tumor-specific antibody and IL2 cytokine. In mice bearing two tumors, we initially hypothesized that delivering RT plus IT-IC to the "primary" tumor would induce a systemic antitumor response causing regression of the "secondary" tumor. To test this, mice bearing one or two syngeneic murine tumors of B78 melanoma and/or Panc02 pancreatic cancer were treated with combined external beam RT and IT-IC to the designated "primary" tumor only. Primary and secondary tumor response as well as animal survival were monitored. Immunohistochemistry and quantitative real-time PCR were used to quantify tumor infiltration with regulatory T cells (Treg). Transgenic "DEREG" mice or IgG2a anti-CTLA-4 were used to transiently deplete tumor Tregs. Contrary to our initial hypothesis, we observed that the presence of an untreated secondary tumor antagonized the therapeutic effect of RT + IT-IC delivered to the primary tumor. We observed reciprocal tumor specificity for this effect, which was circumvented if all tumors received RT or by transient depletion of Tregs. Primary tumor treatment with RT + IT-IC together with systemic administration of Treg-depleting anti-CTLA-4 resulted in a renewed in situ vaccination effect. Our findings show that untreated tumors can exert a tumor-specific, Treg-dependent, suppressive effect on the efficacy of in situ vaccination and demonstrate clinically viable approaches to overcome this effect. Untreated tumor sites antagonize the systemic and local antitumor immune response to an in situ vaccination regimen. This effect is radiation sensitive and may be mediated by tumor-specific regulatory T cells harbored in the untreated tumor sites. Cancer Immunol Res; 6(7); 825-34. ©2018 AACR.


Subject(s)
Cancer Vaccines/immunology , Neoplasms/immunology , Animals , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Humans , Immune Tolerance , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Melanoma/therapy , Mice , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Vaccination , Xenograft Model Antitumor Assays
4.
Radiother Oncol ; 124(3): 418-426, 2017 09.
Article in English | MEDLINE | ID: mdl-28893414

ABSTRACT

BACKGROUND AND PURPOSE: We recently reported a time-sensitive, cooperative, anti-tumor effect elicited by radiation (RT) and intra-tumoral-immunocytokine injection in vivo. We hypothesized that RT triggers transcriptional-mediated changes in tumor expression of immune susceptibility markers at delayed time points, which may explain these previously observed time-dependent effects. MATERIALS AND METHODS: We examined the time course of changes in expression of immune susceptibility markers following in vitro or in vivo RT in B78 murine melanoma and A375 human melanoma using flow cytometry, immunoblotting, and qPCR. RESULTS: Flow cytometry and immunoblot revealed time-dependent increases in expression of death receptors and T cell co-stimulatory/co-inhibitory ligands following RT in murine and human melanoma. Using high-throughput qPCR, we observed comparable time courses of RT-induced transcriptional upregulation for multiple immune susceptibility markers. We confirmed analogous changes in B78 tumors irradiated in vivo. We observed upregulated expression of DNA damage response markers days prior to changes in immune markers, whereas phosphorylation of the STAT1 transcription factor occurred concurrently with changes following RT. CONCLUSION: This study highlights time-dependent, transcription-mediated changes in tumor immune susceptibility marker expression following RT. These findings may help in the design of strategies to optimize sequencing of RT and immunotherapy in translational and clinical studies.


Subject(s)
Melanoma/radiotherapy , Animals , B7-1 Antigen/biosynthesis , B7-1 Antigen/immunology , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/immunology , Cell Line, Tumor , Female , Flow Cytometry , Gene Knockdown Techniques , Humans , Immunoblotting , Melanoma/genetics , Melanoma/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/radiotherapy , Mice , Mice, Inbred C57BL , Phosphorylation , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Transcription, Genetic , Up-Regulation
5.
Clin Cancer Res ; 23(20): 6044-6053, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28659312

ABSTRACT

Purpose: Adenoid cystic carcinoma (ACC) is a rare cancer arising from the major or minor salivary gland tissues of the head and neck. There are currently no approved systemic agents or known radiosensitizers for ACC. Unlike the more common head and neck squamous cell carcinomas that frequently harbor TP53 mutations, ACCs contain TP53 mutations at a rate of <5%, rendering them an attractive target for MDM2 inhibition.Experimental Design: We report the successful establishment and detailed characterization of a TP53-WT ACC patient-derived xenograft (PDX), which retained the histologic features of the original patient tumor. We evaluated this model for response to the MDM2 inhibitor AMG 232 as monotherapy and in combination with radiotherapy.Results: AMG 232 monotherapy induced modest tumor growth inhibition, and radiation monotherapy induced a transient tumor growth delay in a dose-dependent fashion. Strikingly, combination treatment of AMG 232 with radiotherapy (including low-dose radiotherapy of 2 Gy/fraction) induced dramatic tumor response and high local tumor control rates 3 months following treatment. Posttreatment analysis revealed that although both AMG 232 and radiotherapy alone induced TP53 tumor-suppressive activities, combination therapy amplified this response with potent induction of apoptosis after combination treatment.Conclusions: These data identify that MDM2 inhibition can provide potent radiosensitization in TP53-WT ACC. In light of the absence of effective systemic agents for ACC, the powerful response profile observed here suggests that clinical trial evaluation of this drug/radiotherapy combination may be warranted to improve local control in this challenging malignancy. Clin Cancer Res; 23(20); 6044-53. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Adenoid Cystic/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Adenoid Cystic/pathology , Carcinoma, Adenoid Cystic/therapy , Disease Models, Animal , Female , Humans , Immunohistochemistry , Mice , Phosphorylation , Radiation-Sensitizing Agents/therapeutic use , Radiotherapy , Radiotherapy Dosage , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
6.
Cancer Res ; 76(13): 3929-41, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27197149

ABSTRACT

Interest in combining radiotherapy and immune checkpoint therapy is growing rapidly. In this study, we explored a novel combination of this type to augment antitumor immune responses in preclinical murine models of melanoma, neuroblastoma, and head and neck squamous cell carcinoma. Cooperative effects were observed with local radiotherapy and intratumoral injection of tumor-specific antibodies, arising in part from enhanced antibody-dependent cell-mediated cytotoxicity (ADCC). We could improve this response by combining radiation with intratumoral injection of an IL2-linked tumor-specific antibody (termed here an immunocytokine), resulting in complete regression of established tumors in most animals associated with a tumor-specific memory T-cell response. Given the T-cell response elicited by combined local radiation and intratumoral immunocytokine, we tested the potential benefit of adding this treatment to immune checkpoint blockade. In mice bearing large primary tumors or disseminated metastases, the triple-combination of intratumoral immunocytokine, radiation, and systemic anti-CTLA-4 improved primary tumor response and animal survival compared with combinations of any two of these three interventions. Taken together, our results show how combining radiation and intratumoral immunocytokine in murine tumor models can eradicate large tumors and metastases, eliciting an in situ vaccination effect that can be leveraged further by T-cell checkpoint blockade, with immediate implications for clinical evaluation. Cancer Res; 76(13); 3929-41. ©2016 AACR.


Subject(s)
Antibodies, Monoclonal/pharmacology , CTLA-4 Antigen/immunology , Interleukin-2/immunology , Lung Neoplasms/therapy , Melanoma, Experimental/therapy , Pancreatic Neoplasms/therapy , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Blotting, Western , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Chemoradiotherapy , Combined Modality Therapy , Female , Humans , Immunoenzyme Techniques , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , T-Lymphocytes/immunology , Tumor Cells, Cultured , Vaccination , X-Rays , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 22(3): 633-43, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26420857

ABSTRACT

PURPOSE: Aberrant regulation of the EGF receptor family (EGFR, HER2, HER3, HER4) contributes to tumorigenesis and metastasis in epithelial cancers. Pan-HER represents a novel molecular targeted therapeutic composed of a mixture of six monoclonal antibodies against EGFR, HER2, and HER3. EXPERIMENTAL DESIGN: In the current study, we examine the capacity of Pan-HER to augment radiation response across a series of human lung and head and neck cancers, including EGFR inhibitor-resistant cell lines and xenografts. RESULTS: Pan-HER demonstrates superior antiproliferative and radiosensitizing impact when compared with cetuximab. The mechanisms underlying these effects appear to involve attenuation of DNA damage repair, enhancement of programmed cell death, cell-cycle redistribution, and induction of cellular senescence. Combined treatment of Pan-HER with single or fractionated radiation in human tumor xenografts reveals a potent antitumor and regrowth delay impact compared with Pan-HER or radiation treatment alone. CONCLUSIONS: These data highlight the capacity of Pan-HER to augment radiation response in lung and head and neck cancer models and support investigation of Pan-HER combined with radiation as a promising clinical therapeutic strategy.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/metabolism , Lung Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Radiation Tolerance/drug effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Disease Models, Animal , ErbB Receptors/genetics , ErbB Receptors/metabolism , Head and Neck Neoplasms/pathology , Humans , Lung Neoplasms/pathology , Mice , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-3/metabolism , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 14(9): 2049-59, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26141946

ABSTRACT

Human epidermal growth factor receptor family members (EGFR, HER2, HER3, and HER4) play important roles in tumorigenesis and response to cancer therapeutics. In this study, we evaluated the capacity of the dual-target antibody MEHD7945A that simultaneously targets EGFR and HER3 to modulate radiation response in lung and head and neck cancer models. Antitumor effects of MEHD7945A in combination with radiation were evaluated in cell culture and tumor xenograft models. Mechanisms that may contribute to increased radiation killing by MEHD7945A, including DNA damage and inhibition of EGFR-HER signaling pathways, were analyzed. Immunohistochemical analysis of tumor xenografts was conducted to evaluate the effect of MEHD7945A in combination with radiation on tumor growth and microenvironment. MEHD7945A inhibited basal and radiation-induced EGFR and HER3 activation resulting in the inhibition of tumor cell growth and enhanced radiosensitivity. MEHD7945A was more effective in augmenting radiation response than treatment with individual anti-EGFR or anti-HER3 antibodies. An increase in DNA double-strand breaks associated γ-H2AX was observed in cells receiving combined treatment with MEHD7945A and radiation. Immunohistochemical staining evaluation in human tumor xenografts showed that MEHD7945A combined with radiation significantly reduced the expression of markers of tumor proliferation and tumor vasculature. These findings reveal the capacity of MEHD7945A to augment radiation response in lung and head and neck cancers. The dual EGFR/HER3-targeting action of MEHD7945A merits further investigation and clinical trial evaluation as a radiation sensitizer in cancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/metabolism , Immunoglobulin G/pharmacology , Receptor, ErbB-3/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Breaks, Double-Stranded/drug effects , Disease Models, Animal , ErbB Receptors/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Humans , Immunoglobulin G/administration & dosage , Immunohistochemistry , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/radiotherapy , Radiation Tolerance/drug effects , Radiotherapy, Adjuvant , Receptor, ErbB-3/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 14(9): 1994-2003, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26162687

ABSTRACT

MDM2-p53 interaction and downstream signaling affect cellular response to DNA damage. AMG 232 is a potent small molecule inhibitor that blocks the interaction of MDM2 and p53. We examined the capacity of AMG 232 to augment radiation response across a spectrum of human tumor cell lines and xenografts. AMG 232 effectively inhibited proliferation and enhanced radiosensitivity via inhibition of damage repair signaling. Combined AMG 232 and radiation treatment resulted in the accumulation of γH2AX-related DNA damage and induction of senescence with promotion of apoptotic and/or autophagic cell death. Several molecules involved in senescence, autophagy, and apoptosis were specifically modulated following the combined AMG 232/radiation treatment, including FoxM1, ULK-1, DRAM, and BAX. In vivo xenograft studies confirmed more potent antitumor and antiangiogenesis efficacy with combined AMG 232/radiation treatment than treatment with drug or radiation alone. Taken together, these data identify the capacity of AMG 232 to augment radiation response across a variety of tumor types harboring functional p53.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Radiation Tolerance/drug effects , Tumor Suppressor Protein p53/metabolism , Acetates/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/radiation effects , Autophagy/drug effects , Autophagy/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Disease Models, Animal , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/radiotherapy , Piperidones/pharmacology , Protein Binding/drug effects , Proto-Oncogene Proteins c-mdm2/genetics , Radiation, Ionizing , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...