Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
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
2.
Am J Pathol ; 182(1): 217-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23159947

ABSTRACT

The polypeptide hormone prolactin (PRL) stimulates breast epithelial cell growth, differentiation, and motility through its cognate receptor, PRLr. PRLr is expressed in most breast cancers; however, its exact role remains elusive. Our laboratory previously described a novel mode of PRLr signaling in which Stat5a-mediated transcription is regulated through ligand-induced phosphorylation of the PRLr transactivation domain (TAD). Herein, we used a PRLr transactivation-deficient mutant (PRLrYDmut) to identify novel TAD-specific target genes. Microarray analysis identified 120 PRL-induced genes up-regulated by wild type but not PRLrYDmut. Compared with control, PRLr expression significantly induced expression of approximately 4700 PRL-induced genes, whereas PRLrYDmut ablated induction of all but 19 of these genes. Ingenuity pathway analysis found that the PRLr TAD most profoundly affected networks involving cancer and proliferation. In support of this, PRLrYDmut expression reduced anchorage-dependent and anchorage-independent growth. In addition, pathway analysis identified a link between the PRLr TAD and the estrogen and progesterone receptors (ERα/PR). Although neither ERα nor PR was identified as a PRL target gene, a TAD mutation significantly impaired ERα/PR expression and estrogen responsiveness. TMA analysis revealed a marked increase in nuclear, but not cytoplasmic, PRLr TAD phosphorylation as a function of neoplastic progression. We propose that PRLr TAD phosphorylation contributes to breast cancer pathogenesis, in part through regulation of ERα and PR, and has potential utility as a biomarker in this disease.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/biosynthesis , Receptors, Progesterone/biosynthesis , Receptors, Prolactin/genetics , Transcriptional Activation/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Disease Progression , Down-Regulation/drug effects , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genes, Neoplasm , Humans , Mutation , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phosphorylation/genetics , Prognosis , Prolactin/pharmacology , Receptors, Progesterone/genetics , Receptors, Prolactin/biosynthesis , Tissue Array Analysis/methods , Tumor Cells, Cultured , Up-Regulation/drug effects , Up-Regulation/genetics
3.
Sci Rep ; 13(1): 8634, 2023 05 27.
Article in English | MEDLINE | ID: mdl-37244938

ABSTRACT

Radiation therapy induces immunogenic cell death in cancer cells, whereby released endogenous adjuvants are sensed by immune cells to direct adaptive immune responses. TLRs expressed on several immune subtypes recognize innate adjuvants to direct downstream inflammatory responses in part via the adapter protein MyD88. We generated Myd88 conditional knockout mice to interrogate its contribution to the immune response to radiation therapy in distinct immune populations in pancreatic cancer. Surprisingly, Myd88 deletion in Itgax (CD11c)-expressing dendritic cells had little discernable effects on response to RT in pancreatic cancer and elicited normal T cell responses using a prime/boost vaccination strategy. Myd88 deletion in Lck-expressing T cells resulted in similar or worsened responses to radiation therapy compared to wild-type mice and lacked antigen-specific CD8+ T cell responses from vaccination, similar to observations in Myd88-/- mice. Lyz2-specific loss of Myd88 in myeloid populations rendered tumors more susceptible to radiation therapy and elicited normal CD8+ T cell responses to vaccination. scRNAseq in Lyz2-Cre/Myd88fl/fl mice revealed gene signatures in macrophages and monocytes indicative of enhanced type I and II interferon responses, and improved responses to RT were dependent on CD8+ T cells and IFNAR1. Together, these data implicate MyD88 signaling in myeloid cells as a critical source of immunosuppression that hinders adaptive immune tumor control following radiation therapy.


Subject(s)
CD8-Positive T-Lymphocytes , Pancreatic Neoplasms , Mice , Animals , Myeloid Differentiation Factor 88/metabolism , Monocytes/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/radiotherapy , Mice, Knockout , Adjuvants, Immunologic/metabolism , Mice, Inbred C57BL , Pancreatic Neoplasms
4.
Sci Rep ; 13(1): 6277, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37072485

ABSTRACT

Tissue resident memory (Trm) CD8 T cells infiltrating tumors represent an enriched population of tumor antigen-specific T cells, and their presence is associated with improved outcomes in patients. Using genetically engineered mouse pancreatic tumor models we demonstrate that tumor implantation generates a Trm niche that is dependent on direct antigen presentation by cancer cells. However, we observe that initial CCR7-mediated localization of CD8 T cells to tumor draining lymph nodes is required to subsequently generate CD103+ CD8 T cells in tumors. We observe that the formation of CD103+ CD8 T cells in tumors is dependent on CD40L but independent of CD4 T cells, and using mixed chimeras we show that CD8 T cells can provide their own CD40L to permit CD103+ CD8 T cell differentiation. Finally, we show that CD40L is required to provide systemic protection against secondary tumors. These data suggest that CD103+ CD8 T cell formation in tumors can occur independent of the two-factor authentication provided by CD4 T cells and highlight CD103+ CD8 T cells as a distinct differentiation decision from CD4-dependent central memory.


Subject(s)
Immunologic Memory , Neoplasms , Animals , Mice , CD40 Ligand , Neoplasms/pathology , CD8-Positive T-Lymphocytes , Lymphocyte Activation
5.
Nat Med ; 11(5): 491-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15852018

ABSTRACT

Alveolar cell apoptosis is involved in the pathogenesis of emphysema, a prevalent disease primarily caused by cigarette smoking. We report that ceramide, a second messenger lipid, is a crucial mediator of alveolar destruction in emphysema. Inhibition of enzymes controlling de novo ceramide synthesis prevented alveolar cell apoptosis, oxidative stress and emphysema caused by blockade of the vascular endothelial growth factor (VEGF) receptors in both rats and mice. Emphysema was reproduced with intratracheal instillation of ceramide in naive mice. Excessive ceramide triggers a feed-forward mechanism mediated by activation of secretory acid sphingomyelinase, as suggested by experiments with neutralizing ceramide antibody in mice and with acid sphingomyelinase-deficient fibroblasts. Concomitant augmentation of signaling initiated by a prosurvival metabolite, sphingosine-1-phosphate, prevented lung apoptosis, implying that a balance between ceramide and sphingosine-1-phosphate is required for maintenance of alveolar septal integrity. Finally, increased lung ceramides in individuals with smoking-induced emphysema suggests that ceramide upregulation may be a crucial pathogenic element and a promising target in this disease that currently lacks effective therapies.


Subject(s)
Apoptosis/physiology , Ceramides/metabolism , Emphysema/metabolism , Lung/metabolism , Smoking/adverse effects , Sphingosine/analogs & derivatives , Up-Regulation , Acyltransferases/antagonists & inhibitors , Animals , Cells, Cultured , Ceramides/toxicity , Dose-Response Relationship, Drug , Emphysema/chemically induced , Emphysema/pathology , Fatty Acids, Monounsaturated/pharmacology , Fumonisins/pharmacology , Humans , Lung/drug effects , Lung/pathology , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidoreductases/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Serine C-Palmitoyltransferase , Sphingosine/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors
6.
Front Oncol ; 11: 667075, 2021.
Article in English | MEDLINE | ID: mdl-33816320

ABSTRACT

In the cancer literature tumors are inconsistently labeled as 'immunogenic', and experimental results are occasionally dismissed since they are only tested in known 'responsive' tumor models. The definition of immunogenicity has moved from its classical definition based on the rejection of secondary tumors to a more nebulous definition based on immune infiltrates and response to immunotherapy interventions. This review discusses the basis behind tumor immunogenicity and the variation between tumor models, then moves to discuss how these principles apply to the response to radiation therapy. In this way we can identify radioimmunogenic tumor models that are particularly responsive to immunotherapy only when combined with radiation, and identify the interventions that can convert unresponsive tumors so that they can also respond to these treatments.

7.
Oncoimmunology ; 10(1): 1900635, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33796412

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has traditionally been thought of as an immunologically quiescent tumor type presumably because of a relatively low tumor mutational burden (TMB) and poor responses to checkpoint blockade therapy. However, many PDAC tumors exhibit T cell inflamed phenotypes. The presence of tertiary lymphoid structures (TLS) has recently been shown to be predictive of checkpoint blockade response in melanomas and sarcomas, and are prognostic for survival in PDAC. In order to more comprehensively understand tumor immunity in PDAC patients with TLS, we performed RNA-seq, single and multiplex IHC, flow cytometry and predictive genomic analysis on treatment naïve, PDAC surgical specimens. Forty-six percent of tumors contained distinct T and B cell aggregates reflective of "early-stage TLS" (ES-TLS), which correlated with longer overall and progression-free survival. These tumors had greater CD8+ T cell infiltration but were not defined by previously published TLS gene-expression signatures. ES-TLS+ tumors were enriched for IgG1 class-switched memory B cells and memory CD4+ T cells, suggesting durable immunological memory persisted in these patients. We also observed the presence of active germinal centers (mature-TLS) in 31% of tumors with lymphocyte clusters, whose patients had long-term survival (median 56 months). M-TLS-positive tumors had equivalent overall T cell infiltration to ES-TLS, but were enriched for activated CD4+ memory cells, naive B cells and NK cells. Finally, using a TCGA-PDAC dataset, ES-TLS+ tumors harbored a decreased TMB, but M-TLS with germinal centers expressed significantly more MHCI-restricted neoantigens as determined by an in silico neoantigen prediction method. Interestingly, M-TLS+ tumors also had evidence of increased rates of B cell somatic hypermutation, suggesting that germinal centers form in the presence of high-quality tumor neoantigens leading to increased humoral immunity that confers improved survival for PDAC patients. AbbreviationsTLS: tertiary lymphoid structures; GC: germinal center(s); PDAC: pancreatic ductal adenocarcinoma; RNA-seq: RNA sequencing; BCRseq: B cell receptor sequencing; HEV: high endothelial venule; PNAd: peripheral node addressin; TMB: tumor mutational burden; TCGA: the cancer genome atlas; PAAD: pancreatic adenocarcinoma; FFPE: formalin fixed paraffin embedded; TIME: tumor immune microenvironment.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Tertiary Lymphoid Structures , Germinal Center , Humans , Immunity, Humoral , Pancreatic Neoplasms/genetics , Survivorship , Tumor Microenvironment
8.
PLoS One ; 15(8): e0238380, 2020.
Article in English | MEDLINE | ID: mdl-32866185

ABSTRACT

Pancreatic adenocarcinoma is characterized by a complex tumor environment with a wide diversity of infiltrating stromal and immune cell types that impact the tumor response to conventional treatments. However, even in this poorly responsive tumor the extent of T cell infiltration as determined by quantitative immunohistology is a candidate prognostic factor for patient outcome. As such, even more comprehensive immunophenotyping of the tumor environment, such as immune cell type deconvolution via inference models based on gene expression profiling, holds significant promise. We hypothesized that RNA-Seq can provide a comprehensive alternative to quantitative immunohistology for immunophenotyping pancreatic cancer. We performed RNA-Seq on a prospective cohort of pancreatic tumor specimens and compared multiple approaches for gene expression-based immunophenotyping analysis compared to quantitative immunohistology. Our analyses demonstrated that while gene expression analyses provide additional information on the complexity of the tumor immune environment, they are limited in sensitivity by the low overall immune infiltrate in pancreatic cancer. As an alternative approach, we identified a set of genes that were enriched in highly T cell infiltrated pancreatic tumors, and demonstrate that these can identify patients with improved outcome in a reference population. These data demonstrate that the poor immune infiltrate in pancreatic cancer can present problems for analyses that use gene expression-based tools; however, there remains enormous potential in using these approaches to understand the relationships between diverse patterns of infiltrating cells and their impact on patient treatment outcomes.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adult , Aged , Aged, 80 and over , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Male , Middle Aged , Prospective Studies , T-Lymphocytes/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
9.
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
10.
Am J Respir Cell Mol Biol ; 38(6): 639-46, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18192502

ABSTRACT

The de novo pathway of ceramide synthesis has been implicated in the pathogenesis of excessive lung apoptosis and murine emphysema. Intracellular and paracellular-generated ceramides may trigger apoptosis and propagate the death signals to neighboring cells, respectively. In this study we compared the sphingolipid signaling pathways triggered by the paracellular- versus intracellular-generated ceramides as they induce lung endothelial cell apoptosis, a process important in emphysema development. Intermediate-chain length (C(8:0)) extracellular ceramides, used as a surrogate of paracellular ceramides, triggered caspase-3 activation in primary mouse lung endothelial cells, similar to TNF-alpha-generated endogenous ceramides. Inhibitory siRNA against serine palmitoyl transferase subunit 1 but not acid sphingomyelinase inhibited both C(8:0) ceramide- and TNF-alpha (plus cycloheximide)-induced apoptosis, consistent with the requirement for activation of the de novo pathway of sphingolipid synthesis. Tandem mass spectrometry analysis detected increases in both relative and absolute levels of C(16:0) ceramide in response to C(8:0) and TNF-alpha treatments. These results implicate the de novo pathway of ceramide synthesis in the apoptotic effects of both paracellular ceramides and TNF-alpha-stimulated intracellular ceramides in primary lung endothelial cells. The serine palmitoyl synthase-regulated ceramides synthesis may contribute to the amplification of pulmonary vascular injury induced by excessive ceramides.


Subject(s)
Apoptosis/physiology , Ceramides/metabolism , Endothelial Cells/metabolism , Lung/cytology , Signal Transduction/physiology , Sphingolipids/metabolism , Animals , Caspase 3/metabolism , Cells, Cultured , Ceramides/chemistry , Endothelial Cells/cytology , Enzyme Activation , Humans , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Serine C-Palmitoyltransferase/genetics , Serine C-Palmitoyltransferase/metabolism , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Cancer Cell ; 34(4): 561-578.e6, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30300579

ABSTRACT

Complement is a critical component of humoral immunity implicated in cancer development; however, its biological contributions to tumorigenesis remain poorly understood. Using the K14-HPV16 transgenic mouse model of squamous carcinogenesis, we report that urokinase (uPA)+ macrophages regulate C3-independent release of C5a during premalignant progression, which in turn regulates protumorigenic properties of C5aR1+ mast cells and macrophages, including suppression of CD8+ T cell cytotoxicity. Therapeutic inhibition of C5aR1 via the peptide antagonist PMX-53 improved efficacy of paclitaxel chemotherapy associated with increased presence and cytotoxic properties of CXCR3+ effector memory CD8+ T cells in carcinomas, dependent on both macrophage transcriptional programming and IFNγ. Together, these data identify C5aR1-dependent signaling as an important immunomodulatory program in neoplastic tissue tractable for combinatorial cancer immunotherapy.


Subject(s)
Carcinogenesis/drug effects , Complement C5a/drug effects , Drug Therapy , Receptor, Anaphylatoxin C5a/drug effects , Animals , CD8-Positive T-Lymphocytes/drug effects , Carcinoma, Squamous Cell/drug therapy , Disease Models, Animal , Drug Therapy/methods , Humans , Macrophages/drug effects , Macrophages/physiology , Mice , Signal Transduction/drug effects
12.
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
13.
Cancer Discov ; 6(1): 22-35, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26552413

ABSTRACT

UNLABELLED: Skin is a highly ordered immune organ that coordinates rapid responses to external insult while maintaining self-tolerance. In healthy tissue, lymphatic vessels drain fluid and coordinate local immune responses; however, environmental factors induce lymphatic vessel dysfunction, leading to lymph stasis and perturbed regional immunity. These same environmental factors drive the formation of local malignancies, which are also influenced by local inflammation. Herein, we discuss clinical and experimental evidence supporting the tenet that lymphatic vessels participate in regulation of cutaneous inflammation and immunity, and are important contributors to malignancy and potential biomarkers and targets for immunotherapy. SIGNIFICANCE: The tumor microenvironment and tumor-associated inflammation are now appreciated not only for their role in cancer progression but also for their response to therapy. The lymphatic vasculature is a less-appreciated component of this microenvironment that coordinates local inflammation and immunity and thereby critically shapes local responses. A mechanistic understanding of the complexities of lymphatic vessel function in the unique context of skin provides a model to understand how regional immune dysfunction drives cutaneous malignancies, and as such lymphatic vessels represent a biomarker of cutaneous immunity that may provide insight into cancer prognosis and effective therapy.


Subject(s)
Lymphatic Vessels/immunology , Skin Neoplasms/pathology , Biomarkers, Tumor/immunology , Humans , Lymphatic Vessels/pathology , Skin Neoplasms/immunology , Tumor Microenvironment
14.
Mol Cancer Res ; 14(10): 994-1008, 2016 10.
Article in English | MEDLINE | ID: mdl-27358110

ABSTRACT

Stat5a is a transcription factor utilized by several cytokine/hormone receptor signaling pathways that promotes transcription of genes associated with proliferation, differentiation, and survival of cancer cells. However, there are currently no clinically approved therapies that directly target Stat5a, despite ample evidence that it contributes to breast cancer pathogenesis. Here, deacetylation of the Stat5a coactivator and chromatin-remodeling protein HMGN2 on lysine residue K2 by HDAC6 promotes Stat5a-mediated transcription and breast cancer growth. HDAC6 inhibition both in vitro and in vivo enhances HMGN2 acetylation with a concomitant reduction in Stat5a-mediated signaling, resulting in an inhibition of breast cancer growth. Furthermore, HMGN2 is highly acetylated at K2 in normal human breast tissue, but is deacetylated in primary breast tumors and lymph node metastases, suggesting that targeting HMGN2 deacetylation is a viable treatment for breast cancer. Together, these results reveal a novel mechanism by which HDAC6 activity promotes the transcription of Stat5a target genes and demonstrate utility of HDAC6 inhibition for breast cancer therapy. IMPLICATIONS: HMGN2 deacetylation enhances Stat5a transcriptional activity, thereby regulating prolactin-induced gene transcription and breast cancer growth. Mol Cancer Res; 14(10); 994-1008. ©2016 AACR.


Subject(s)
Breast Neoplasms/pathology , HMGN2 Protein/metabolism , Histone Deacetylases/metabolism , STAT5 Transcription Factor/genetics , Transcription, Genetic , Tumor Suppressor Proteins/genetics , Acetylation , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Histone Deacetylase 6 , Humans , Lysine/metabolism , MCF-7 Cells , Mice , Neoplasm Metastasis , Neoplasm Transplantation
15.
Trends Cancer ; 1(1): 66-75, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26457331

ABSTRACT

Chemotherapy and radiotherapy have been extensively used to eradicate cancer based on their direct cytocidal effects on rapidly proliferating tumor cells. Accumulating evidence indicates that these therapies also dramatically affect resident and recruited immune cells that actively support tumor growth. We now appreciate that mobilization of effector CD8+ T cells enhances efficacy of chemotherapy and radiotherapy; remarkable clinical advances have been achieved by blocking regulatory programs limiting cytotoxic CD8+ T cell activity . This review discusses immune-mediated mechanisms underlying efficacy of chemotherapy and radiotherapy, and provides a perspective on how understanding tissue-based immune mechanisms can be used to guide therapeutic approaches combining immune and cytotoxic therapies to improve outcomes for a larger subset of patients than currently achievable.

16.
Cancer J ; 21(4): 343-50, 2015.
Article in English | MEDLINE | ID: mdl-26222088

ABSTRACT

It is well established that cancer development ensues based on reciprocal interactions between genomically altered neoplastic cells and diverse populations of recruited "host" cells co-opted to support malignant progression. Among the host cells recruited into tumor microenvironments, several subtypes of myeloid cells, including macrophages, monocytes, dendritic cells, and granulocytes contribute to tumor development by providing tumor-promoting factors as well as a spectrum of molecules that suppress cytotoxic activities of T lymphocytes. Based on compelling preclinical data revealing that inhibition of critical myeloid-based programs leads to tumor suppression, novel immune-based therapies and approaches are now entering the clinic for evaluation. This review discusses mechanisms underlying protumorigenic programming of myeloid cells and discusses how targeting of these has potential to attenuate solid tumor progression via the induction and of mobilization CD8 cytotoxic T cell immunity.


Subject(s)
Myeloid Cells/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , B-Lymphocytes/immunology , Dendritic Cells/immunology , Eosinophils/immunology , Granulocytes/immunology , Humans , Macrophages/immunology , Monocytes/immunology , Neoplasms/drug therapy , Neutrophils/immunology
18.
Cancer Cell ; 25(6): 809-821, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24909985

ABSTRACT

B cells foster squamous cell carcinoma (SCC) development through deposition of immunoglobulin-containing immune complexes in premalignant tissue and Fcγ receptor-dependent activation of myeloid cells. Because human SCCs of the vulva and head and neck exhibited hallmarks of B cell infiltration, we examined B cell-deficient mice and found reduced support for SCC growth. Although ineffective as a single agent, treatment of mice bearing preexisting SCCs with B cell-depleting αCD20 monoclonal antibodies improved response to platinum- and Taxol-based chemotherapy. Improved chemoresponsiveness was dependent on altered chemokine expression by macrophages that promoted tumor infiltration of activated CD8(+) lymphocytes via CCR5-dependent mechanisms. These data reveal that B cells, and the downstream myeloid-based pathways they regulate, represent tractable targets for anticancer therapy in select tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B-Lymphocytes/immunology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/immunology , Macrophages/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CHO Cells , Carcinoma, Squamous Cell/pathology , Cricetulus , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Transgenic , Neoplasms/pathology , Organoplatinum Compounds/administration & dosage , Paclitaxel/administration & dosage , Phenotype , Tumor Microenvironment , Xenograft Model Antitumor Assays
19.
Mol Endocrinol ; 25(9): 1550-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21816901

ABSTRACT

The direct actions of transmembrane receptors within the nucleus remain enigmatic. In this report, we demonstrate that the prolactin receptor (PRLr) localizes to the nucleus where it functions as a coactivator through its interactions with the latent transcription factor signal transducer and activator of transcription 5a (Stat5a) and the high-mobility group N2 protein (HMGN2). We identify a novel transactivation domain within the PRLr that is activated by ligand-induced phosphorylation, an event coupled to HMGN2 binding. The association of the PRLr with HMGN2 enables Stat5a-responsive promoter binding, thus facilitating transcriptional activation and promoting anchorage-independent growth. We propose that HMGN2 serves as a critical regulatory factor in Stat5a-driven gene expression by facilitating the assembly of PRLr/Stat5a onto chromatin and that these events may serve to promote biological events that contribute to a tumorigenic phenotype. Our data imply that phosphorylation may be the molecular switch that activates a cell surface receptor transactivation domain, enabling it to tether chromatin-modifying factors, such as HMGN2, to target promoter regions in a sequence-specific manner.


Subject(s)
Cell Nucleus/metabolism , HMGN2 Protein/metabolism , Receptors, Prolactin/chemistry , Receptors, Prolactin/metabolism , STAT5 Transcription Factor/metabolism , Transcription, Genetic , Transcriptional Activation/genetics , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Models, Biological , Molecular Sequence Data , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Protein Transport
20.
Mol Endocrinol ; 25(12): 2054-64, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21964595

ABSTRACT

The molecular mechanisms that modulate the activity of the signal transducers and activators of transcription 5 (Stat5) during the progression of breast cancer remain elusive. Here, we present evidence that the calcineurin/nuclear factor of activated T cells (NFAT) pathway negatively regulates the activation of Stat5, and vice versa in breast cancer. NFAT1 interacts with Stat5 in breast cancer cells, and their physical association is mediated by the DNA binding and transactivation domains of Stat5. Ectopically expressed NFAT1 is capable of inhibiting Stat5-dependent functions, including Stat5 transactivation, Stat5-mediated transcription of the downstream target gene expression, and binding of Stat5a to the Stat5 target promoter. By contrast, overexpression of a selective NFAT inhibitor VIVIT reversed NFAT1-mediated suppression of Stat5-dependent gene expression, whereas silencing of NFAT1 through RNA interference enhanced prolactin-induced, Stat5-mediated gene transcription, and breast cancer cell proliferation. A reciprocal inhibitory effect of Stat5 activity on NFAT1 signaling was also observed, implying these two signaling cascades antagonize each other in breast cancer. Importantly, analysis of a matched breast cancer progression tissue microarray revealed a negative correlation between levels of NFAT1 and Stat5 (pY694) during the progression of breast cancer. Taken together, these studies highlight a novel negative cross talk between the NFAT1- and Stat5-signaling cascades that may affect breast tumor formation, growth, and metastasis.


Subject(s)
Breast Neoplasms/metabolism , NFATC Transcription Factors/metabolism , Receptor Cross-Talk , STAT5 Transcription Factor/metabolism , Signal Transduction , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Feedback, Physiological , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , NFATC Transcription Factors/chemistry , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , STAT5 Transcription Factor/chemistry , Tissue Array Analysis , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL