Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Breast Cancer Res ; 25(1): 68, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37312163

ABSTRACT

BACKGROUND: Most patients with estrogen receptor positive (ER+) breast cancer do not respond to immune checkpoint inhibition (ICI); the tumor microenvironment (TME) of these cancers is generally immunosuppressive and contains few tumor-infiltrating lymphocytes. Radiation therapy (RT) can increase tumor inflammation and infiltration by lymphocytes but does not improve responses to ICIs in these patients. This may result, in part, from additional effects of RT that suppress anti-tumor immunity, including increased tumor infiltration by myeloid-derived suppressor cells and regulatory T cells. We hypothesized that anti-estrogens, which are a standard of care for ER+ breast cancer, may ameliorate these detrimental effects of RT by reducing the recruitment/ activation of suppressive immune populations in the radiated TME, increasing anti-tumor immunity and responsiveness to ICIs. METHODS: To interrogate the effect of the selective estrogen receptor downregulator, fulvestrant, on the irradiated TME in the absence of confounding growth inhibition by fulvestrant on tumor cells, we used the TC11 murine model of anti-estrogen resistant ER+ breast cancer. Tumors were orthotopically transplanted into immunocompetent syngeneic mice. Once tumors were established, we initiated treatment with fulvestrant or vehicle, followed by external beam RT one week later. We examined the number and activity of tumor infiltrating immune cells using flow cytometry, microscopy, transcript levels, and cytokine profiles. We tested whether fulvestrant improved tumor response and animal survival when added to the combination of RT and ICI. RESULTS: Despite resistance of TC11 tumors to anti-estrogen therapy alone, fulvestrant slowed tumor regrowth following RT, and significantly altered multiple immune populations in the irradiated TME. Fulvestrant reduced the influx of Ly6C+Ly6G+ cells, increased markers of pro-inflammatory myeloid cells and activated T cells, and augmented the ratio of CD8+: FOXP3+ T cells. In contrast to the minimal effects of ICIs when co-treated with either fulvestrant or RT alone, combinatorial treatment with fulvestrant, RT and ICIs significantly reduced tumor growth and prolonged survival. CONCLUSIONS: A combination of RT and fulvestrant can overcome the immunosuppressive TME in a preclinical model of ER+ breast cancer, enhancing the anti-tumor response and increasing the response to ICIs, even when growth of tumor cells is no longer estrogen sensitive.


Subject(s)
Neoplasms , Receptors, Estrogen , Animals , Mice , Fulvestrant/pharmacology , Immunotherapy , Estrogens , Estrogen Antagonists , Immunosuppressive Agents
2.
Nat Commun ; 13(1): 4948, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35999216

ABSTRACT

Radiation therapy (RT) activates an in situ vaccine effect when combined with immune checkpoint blockade (ICB), yet this effect may be limited because RT does not fully optimize tumor antigen presentation or fully overcome suppressive mechanisms in the tumor-immune microenvironment. To overcome this, we develop a multifunctional nanoparticle composed of polylysine, iron oxide, and CpG (PIC) to increase tumor antigen presentation, increase the ratio of M1:M2 tumor-associated macrophages, and enhance stimulation of a type I interferon response in conjunction with RT. In syngeneic immunologically "cold" murine tumor models, the combination of RT, PIC, and ICB significantly improves tumor response and overall survival resulting in cure of many mice and consistent activation of tumor-specific immune memory. Combining RT with PIC to elicit a robust in situ vaccine effect presents a simple and readily translatable strategy to potentiate adaptive anti-tumor immunity and augment response to ICB or potentially other immunotherapies.


Subject(s)
Multifunctional Nanoparticles , Neoplasms , Animals , Antigens, Neoplasm , Cell Line, Tumor , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Mice , Neoplasms/radiotherapy , Tumor Microenvironment , Vaccination
3.
Front Endocrinol (Lausanne) ; 13: 910978, 2022.
Article in English | MEDLINE | ID: mdl-35784527

ABSTRACT

Prolactin coordinates with the ovarian steroids to orchestrate mammary development and lactation, culminating in nourishment and an increasingly appreciated array of other benefits for neonates. Its central activities in mammary epithelial growth and differentiation suggest that it plays a role(s) in breast cancer, but it has been challenging to identify its contributions, essential for incorporation into prevention and treatment approaches. Large prospective epidemiologic studies have linked higher prolactin exposure to increased risk, particularly for ER+ breast cancer in postmenopausal women. However, it has been more difficult to determine its actions and clinical consequences in established tumors. Here we review experimental data implicating multiple mechanisms by which prolactin may increase the risk of breast cancer. We then consider the evidence for role(s) of prolactin and its downstream signaling cascades in disease progression and treatment responses, and discuss how new approaches are beginning to illuminate the biology behind the seemingly conflicting epidemiologic and experimental studies of prolactin actions across diverse breast cancers.


Subject(s)
Breast Neoplasms , Prolactin , Breast Neoplasms/epidemiology , Disease Progression , Female , Humans , Infant, Newborn , Lactation , Prospective Studies
4.
Cancers (Basel) ; 13(15)2021 Jul 24.
Article in English | MEDLINE | ID: mdl-34359625

ABSTRACT

Estrogen receptor alpha (ERα) marks heterogeneous breast cancers which display a repertoire of somatic genomic mutations and an immune environment that differs from other breast cancer subtypes. These cancers also exhibit distinct biological behaviors; despite an overall better prognosis than HER2+ or triple negative breast cancers, disseminated dormant cells can lead to disease recurrence decades after the initial diagnosis and treatment. Estrogen is the best studied driver of these cancers, and antagonism or reduction of estrogen activity is the cornerstone of therapeutic approaches. In addition to reducing proliferation of ERα+ cancer cells, these treatments also alter signals to multiple other target cells in the environment, including immune cell subpopulations, cancer-associated fibroblasts, and endothelial cells via several distinct estrogen receptors. In this review, we update progress in our understanding of the stromal cells populating the microenvironments of primary and metastatic ER+ tumors, the effects of estrogen on tumor and stromal cells to modulate immune activity and the extracellular matrix, and net outcomes in experimental and clinical studies. We highlight new approaches that will illuminate the unique biology of these cancers, provide the foundation for developing new treatment and prevention strategies, and reduce mortality of this disease.

5.
Cancer Lett ; 503: 231-239, 2021 04 10.
Article in English | MEDLINE | ID: mdl-33472091

ABSTRACT

Prolactin (PRL) cooperates with other factors to orchestrate mammary development and lactation, and is epidemiologically linked to higher risk for breast cancer. However, how PRL collaborates with oncogenes to foster tumorigenesis and influence breast cancer phenotype is not well understood. To understand its interactions with canonical Wnt signals, which elevate mammary stem cell activity, we crossed heterozygous NRL-PRL mice with ApcMin/+ mice and treated pubertal females with a single dose of mutagen. PRL in the context of ApcMin/+ fueled a dramatic increase in tumor incidence in nulliparous mice, compared to ApcMin/+ alone. Although carcinomas in both NRL-PRL/ApcMin/+ and ApcMin/+ females acquired a mutation in the remaining wildtype Apc allele and expressed abundant ß-catenin, PRL-promoted tumors displayed higher levels of Notch-driven target genes and Notch-dependent cancer stem cell activity, compared to ß-catenin-driven activity in ApcMin/+ tumors. This PRL-induced shift to dominant Notch signals was evident in preneoplastic epithelial hyperplasias at 120 days of age. In NRL-PRL/ApcMin/+ females, rapidly proliferating hyperplasias, characterized by ß-catenin at cell junctions and high NOTCH1 expression, contrasted with slower growing lesions with nuclear ß-catenin in ApcMin/+ females. These studies demonstrate that PRL can powerfully modulate the incidence and phenotype of mammary tumors, shedding light on mechanisms whereby PRL elevates risk of breast cancer.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Mammary Neoplasms, Experimental/pathology , Mutagens/toxicity , Prolactin/genetics , Animals , Cell Nucleus/metabolism , Cell Proliferation , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Receptors, Notch/metabolism , Wnt Signaling Pathway
6.
Integr Biol (Camb) ; 12(2): 21-33, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32118264

ABSTRACT

Accumulating evidence suggests that our ability to predict chemical effects on breast cancer is limited by a lack of physiologically relevant in vitro models; the typical in vitro breast cancer model consists of the cancer cell and excludes the mammary microenvironment. As the effects of the microenvironment on cancer cell behavior becomes more understood, researchers have called for the integration of the microenvironment into in vitro chemical testing systems. However, given the complexity of the microenvironment and the variety of platforms to choose from, identifying the essential parameters to include in a chemical testing platform is challenging. This review discusses the need for more complex in vitro breast cancer models and outlines different approaches used to model breast cancer in vitro. We provide examples of the microenvironment modulating breast cancer cell responses to chemicals and discuss strategies to help pinpoint what components should be included in a model.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Carcinoma, Intraductal, Noninfiltrating/diagnosis , Cell Line, Tumor , Disease Progression , Drug Screening Assays, Antitumor , Epithelium/pathology , Extracellular Matrix/metabolism , Female , Humans , Inflammation , Models, Statistical , Neoplasms , Phenotype , Tissue Engineering
7.
Oncogene ; 38(43): 6913-6925, 2019 10.
Article in English | MEDLINE | ID: mdl-31406251

ABSTRACT

Metastatic, antiestrogen resistant estrogen receptor α positive (ER+) breast cancer is the leading cause of breast cancer deaths in USA women. While studies have demonstrated the importance of the stromal tumor microenvironment in cancer progression and therapeutic responses, effects on the responses of ER+ cancers to estrogen and antiestrogens are poorly understood, particularly in the complex in vivo environment. In this study, we used an estrogen responsive syngeneic mouse model to interrogate how a COL1A1-enriched fibrotic ECM modulates integrated hormonal responses in cancer progression. We orthotopically transplanted the ER+ TC11 cell line into wild-type (WT) or collagen-dense (Col1a1tm1Jae/+, mCol1a1) syngeneic FVB/N female mice. Once tumors were established, recipients were supplemented with 17ß-estradiol (E2), tamoxifen, or left untreated. Although the dense/stiff environment in mCol1a1 recipients did not alter the rate of E2-induced proliferation of the primary tumor, it fostered the agonist activity of tamoxifen to increase proliferation and AP-1 activity. Manipulation of estrogen activity did not alter the incidence of lung lesions in either WT or mCol1a1 hosts. However, the mCol1a1 environment enabled tamoxifen-stimulated growth of pulmonary metastases and further fueled estrogen-driven growth. Moreover, E2 remodeled peritumoral ECM architecture in WT animals, modifying alignment of collagen fibers and altering synthesis of ECM components associated with increased alignment and stiffness, and increasing FN1 and POSTN expression in the pulmonary metastatic niche. These studies demonstrate dynamic interactions between ECM properties and estrogen activity in progression of ER+ breast cancer, and support the need for therapeutics that target both ER and the tumor microenvironment.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Extracellular Matrix/metabolism , Animals , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Disease Progression , Estradiol/metabolism , Estrogen Antagonists/pharmacology , Extracellular Matrix/drug effects , Female , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Tamoxifen/pharmacology , Tumor Microenvironment/drug effects
8.
Cell Rep ; 28(6): 1526-1537.e4, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31390566

ABSTRACT

The NRL-PRL murine model, defined by mammary-selective transgenic rat prolactin ligand rPrl expression, establishes spontaneous ER+ mammary tumors in nulliparous females, mimicking the association between elevated prolactin (PRL) and risk for development of ER+ breast cancer in postmenopausal women. Whole-genome and exome sequencing in a discovery cohort (n = 5) of end-stage tumors revealed canonical activating mutations and copy number amplifications of Kras. The frequent mutations in this pathway were validated in an extension cohort, identifying activating Ras alterations in 79% of tumors (23 of 29). Transcriptome analyses over the course of oncogenesis revealed marked alterations associated with Ras activity in established tumors compared with preneoplastic tissues; in cell-intrinsic processes associated with mitosis, cell adhesion, and invasion; as well as in the surrounding tumor environment. These genomic analyses suggest that PRL induces a selective bottleneck for spontaneous Ras-driven tumors that may model a subset of aggressive clinical ER+ breast cancers.


Subject(s)
Estrogen Receptor alpha/metabolism , Mammary Neoplasms, Experimental/etiology , Proto-Oncogene Proteins p21(ras)/metabolism , Aging/metabolism , Animals , Carcinogenesis/genetics , Datasets as Topic , Female , Gene Expression Profiling , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Prolactin/genetics , Prolactin/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Signal Transduction , Transgenes
9.
Cancer Lett ; 433: 1-9, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29935374

ABSTRACT

Metastatic estrogen receptor alpha positive (ERα+) cancers account for most breast cancer mortality. Cancer stem cells (CSCs) and dense/stiff extracellular matrices are implicated in aggression and therapy resistance. We examined this interplay and response to mTOR inhibition using ERα+ adenocarcinomas from NRL-PRL females in combination with Col1a1tmJae/+ (mCol1a1) mice, which accumulate collagen-I around growing tumors. Orthotopic transplantation of tumor cells to mCol1a1 but not wildtype hosts resulted in striking desmoplasia. Mammary tumors in mCol1a1 recipients displayed higher CSC activity and enhanced AKT-mTOR and YAP activation, and these animals developed more and larger lung metastases. Treatment with the mTOR inhibitor, rapamycin, with or without the anti-estrogen, ICI182780, rapidly diminished mammary tumors, which rapidly reversed when treatment ceased. In contrast, lung metastases, which exhibited lower proliferation and pS6RP, indicating lower mTOR activity, were unresponsive, and mCol1a1 hosts continued to sustain greater metastatic burdens. These findings shed light on the influence of desmoplastic tumor microenvironments on the CSC niche and metastatic behavior in ERα+ breast cancer. The differential mTOR dependence of local mammary tumors and pulmonary lesions has implications for success of mTOR inhibitors in advanced ERα+ disease.


Subject(s)
Breast Neoplasms/pathology , Collagen/metabolism , Estrogen Receptor alpha/metabolism , Lung Neoplasms/secondary , Neoplastic Stem Cells/metabolism , Signal Transduction , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Female , Fulvestrant/administration & dosage , Fulvestrant/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Signal Transduction/drug effects , Sirolimus/administration & dosage , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Tumor Microenvironment/drug effects
10.
J Endocr Soc ; 2(3): 293-309, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29594259

ABSTRACT

Prolactin (PRL) and estrogen cooperate in lobuloalveolar development of the mammary gland and jointly regulate gene expression in breast cancer cells in vitro. Canonical PRL signaling activates STAT5A/B, homologous proteins that have different target genes and functions. Although STAT5A/B are important for physiological mammary function and tumor pathophysiology, little is known about regulation of their expression, particularly of STAT5B, and the consequences for hormone action. In this study, we examined the effect of two estrogenic ligands, 17ß-estradiol (E2) and the clinical antiestrogen, ICI182,780 (ICI, fulvestrant) on expression of STAT5 isoforms and resulting crosstalk with PRL in normal and tumor murine mammary epithelial cell lines. In all cell lines, E2 and ICI significantly increased protein and corresponding nascent and mature transcripts for STAT5A and STAT5B, respectively. Transcriptional regulation of STAT5A and STAT5B by E2 and ICI, respectively, is associated with recruitment of estrogen receptor alpha and increased H3K27Ac at a common intronic enhancer 10 kb downstream of the Stat5a transcription start site. Further, E2 and ICI induced different transcripts associated with differentiation and tumor behavior. In tumor cells, E2 also significantly increased proliferation, invasion, and stem cell-like activity, whereas ICI had no effect. To evaluate the role of STAT5B in these responses, we reduced STAT5B expression using short hairpin (sh) RNA. shSTAT5B blocked ICI-induced transcripts associated with metastasis and the epithelial mesenchymal transition in both cell types. shSTAT5B also blocked E2-induced invasion of tumor epithelium without altering E2-induced transcripts. Together, these studies indicate that STAT5B mediates a subset of protumorigenic responses to both E2 and ICI, underscoring the need to understand regulation of its expression and suggesting exploration as a possible therapeutic target in breast cancer.

11.
Cancer Res ; 78(7): 1672-1684, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29363543

ABSTRACT

Although antiestrogen therapies are successful in many patients with estrogen receptor alpha-positive (ERα+) breast cancer, 25% to 40% fail to respond. Although multiple mechanisms underlie evasion of these treatments, including tumor heterogeneity and drug-resistant cancer stem cells (CSC), further investigations have been limited by the paucity of preclinical ERα+ tumor models. Here, we examined a mouse model of prolactin-induced aggressive ERα+ breast cancer, which mimics the epidemiologic link between prolactin exposure and increased risk for metastatic ERα+ tumors. Like a subset of ERα+ patient cancers, the prolactin-induced adenocarcinomas contained two major tumor subpopulations that expressed markers of normal luminal and basal epithelial cells. CSC activity was distributed equally across these two tumor subpopulations. Treatment with the selective estrogen receptor downregulator (SERD), ICI 182,780 (ICI), did not slow tumor growth, but induced adaptive responses in CSC activity, increased markers of plasticity including target gene reporters of Wnt/Notch signaling and epithelial-mesenchymal transition, and increased double-positive (K8/K5) cells. In primary tumorsphere cultures, ICI stimulated CSC self-renewal and was able to overcome the dependence of self-renewal upon Wnt or Notch signaling individually, but not together. Our findings demonstrate that treatment of aggressive mixed lineage ERα+ breast cancers with a SERD does not inhibit growth, but rather evokes tumor cell plasticity and regenerative CSC activity, predicting likely negative impacts on patient tumors with these characteristics.Significance: This study suggests that treatment of a subset of ERα+ breast cancers with antiestrogen therapies may not only fail to slow growth but also promote aggressive behavior by evoking tumor cell plasticity and regenerative CSC activity. Cancer Res; 78(7); 1672-84. ©2018 AACR.


Subject(s)
Adenocarcinoma/drug therapy , Breast Neoplasms/drug therapy , Cell Plasticity/drug effects , Estrogen Receptor Antagonists/therapeutic use , Estrogen Receptor alpha/antagonists & inhibitors , Mammary Neoplasms, Animal/drug therapy , Neoplastic Stem Cells/pathology , Adenocarcinoma/chemically induced , Adenocarcinoma/pathology , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Estrogen Receptor alpha/metabolism , Female , Fulvestrant/therapeutic use , Humans , Mammary Neoplasms, Animal/chemically induced , Mammary Neoplasms, Animal/pathology , Mice , Prolactin/adverse effects , Tumor Cells, Cultured , Wnt Signaling Pathway/physiology
12.
Stem Cell Reports ; 9(4): 1167-1179, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28919264

ABSTRACT

Hormones drive mammary development and function and play critical roles in breast cancer. Epidemiologic studies link prolactin (PRL) to increased risk for aggressive cancers that express estrogen receptor α (ERα). However, in contrast to ovarian steroids, PRL actions on the mammary gland outside of pregnancy are poorly understood. We employed the transgenic NRL-PRL model to examine the effects of PRL alone and with defined estrogen/progesterone exposure on stem/progenitor activity and regulatory networks that drive epithelial differentiation. PRL increased progenitors and modulated transcriptional programs, even without ovarian steroids, and with steroids further raised stem cell activity associated with elevated canonical Wnt signaling. However, despite facilitating some steroid actions, PRL opposed steroid-driven luminal maturation and increased CD61+ luminal cells. Our findings demonstrate that PRL can powerfully influence the epithelial hierarchy alone and temper the actions of ovarian steroids, which may underlie its role in the development of breast cancer.


Subject(s)
Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Ovary/metabolism , Prolactin/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Steroids/metabolism , Age Factors , Animals , Biomarkers , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epithelial Cells/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Female , Gene Expression , Gonadal Steroid Hormones/metabolism , Mice , Mice, Transgenic , Progesterone/metabolism , Rats , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
13.
Breast Cancer Res ; 19(1): 9, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28103936

ABSTRACT

BACKGROUND: The development and progression of estrogen receptor alpha positive (ERα+) breast cancer has been linked epidemiologically to prolactin. However, activation of the canonical mediator of prolactin, STAT5, is associated with more differentiated cancers and better prognoses. We have reported that density/stiffness of the extracellular matrix potently modulates the repertoire of prolactin signals in human ERα + breast cancer cells in vitro: stiff matrices shift the balance from the Janus kinase (JAK)2/STAT5 cascade toward pro-tumor progressive extracellular regulated kinase (ERK)1/2 signals, driving invasion. However, the consequences for behavior of ERα + cancers in vivo are not known. METHODS: In order to investigate the importance of matrix density/stiffness in progression of ERα + cancers, we examined tumor development and progression following orthotopic transplantation of two clonal green fluorescent protein (GFP) + ERα + tumor cell lines derived from prolactin-induced tumors to 8-week-old wild-type FVB/N (WT) or collagen-dense (col1a1 tm1Jae/+ ) female mice. The latter express a mutant non-cleavable allele of collagen 1a1 "knocked-in" to the col1a1 gene locus, permitting COL1A1 accumulation. We evaluated the effect of the collagen environment on tumor progression by examining circulating tumor cells and lung metastases, activated signaling pathways by immunohistochemistry analysis and immunoblotting, and collagen structure by second harmonic generation microscopy. RESULTS: ERα + primary tumors did not differ in growth rate, histologic type, ERα, or prolactin receptor (PRLR) expression between col1a1 tm1Jae/+ and WT recipients. However, the col1a1 tm1Jae/+ environment significantly increased circulating tumor cells and the number and size of lung metastases at end stage. Tumors in col1a1 tm1Jae/+ recipients displayed reduced STAT5 activation, and higher phosphorylation of ERK1/2 and AKT. Moreover, intratumoral collagen fibers in col1a1 tm1Jae/+ recipients were aligned with tumor projections into the adjacent fat pad, perpendicular to the bulk of the tumor, in contrast to the collagen fibers wrapped around the more uniformly expansive tumors in WT recipients. CONCLUSIONS: A collagen-dense extracellular matrix can potently interact with hormonal signals to drive metastasis of ERα + breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Collagen Type I/metabolism , Estrogen Receptor alpha/metabolism , Prolactin/metabolism , Signal Transduction , Animals , Biomarkers , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Collagen Type I/genetics , Disease Progression , Extracellular Matrix/metabolism , Female , Immunohistochemistry , MAP Kinase Signaling System , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Staging , Neoplastic Cells, Circulating , Proto-Oncogene Proteins c-akt , STAT5 Transcription Factor/metabolism , Tumor Burden
14.
Oncotarget ; 7(30): 48093-48106, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27344177

ABSTRACT

Estrogen receptor α positive (ERα+) breast cancer accounts for most breast cancer deaths. Both prolactin (PRL) and extracellular matrix (ECM) stiffness/density have been implicated in metastatic progression of this disease. We previously demonstrated that these factors cooperate to fuel processes involved in cancer progression. Culture of ERα+ breast cancer cells in dense/stiff 3D collagen-I matrices shifts the repertoire of PRL signals, and increases crosstalk between PRL and estrogen to promote proliferation and invasion. However, previous work did not distinguish ECM stiffness and collagen density. In order to dissect the ECM features that control PRL signals, we cultured T47D and MCF-7 cells on polyacrylamide hydrogels of varying elastic moduli (stiffness) with varying collagen-I concentrations (ligand density). Increasing stiffness from physiological to pathological significantly augmented PRL-induced phosphorylation of ERK1/2 and the SFK target, FAK-Y925, with only modest effects on pSTAT5. In contrast, higher collagen-I ligand density lowered PRL-induced pSTAT5 with no effect on pERK1/2 or pFAK-Y925. Disrupting focal adhesion signaling decreased PRL signals and PRL/estrogen-induced proliferation more efficiently in stiff, compared to compliant, extracellular environments. These data indicate that matrix stiffness shifts the balance of PRL signals from physiological (JAK2/STAT5) to pathological (FAK/SFK/ERK1/2) by increasing PRL signals through focal adhesions. Together, our studies suggest that PRL signaling to FAK and SFKs may be useful targets in clinical aggressive ERα+ breast carcinomas.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Extracellular Matrix/metabolism , Focal Adhesions/metabolism , Prolactin/metabolism , Cell Line, Tumor , Extracellular Matrix/pathology , Female , Focal Adhesions/pathology , Humans , Signal Transduction
15.
Pharmacol Ther ; 165: 79-92, 2016 09.
Article in English | MEDLINE | ID: mdl-27218886

ABSTRACT

Personalized cancer therapy focuses on characterizing the relevant phenotypes of the patient, as well as the patient's tumor, to predict the most effective cancer therapy. Historically, these methods have not proven predictive in regards to predicting therapeutic response. Emerging culture platforms are designed to better recapitulate the in vivo environment, thus, there is renewed interest in integrating patient samples into in vitro cancer models to assess therapeutic response. Successful examples of translating in vitro response to clinical relevance are limited due to issues with patient sample acquisition, variability and culture. We will review traditional and emerging in vitro models for personalized medicine, focusing on the technologies, microenvironmental components, and readouts utilized. We will then offer our perspective on how to apply a framework derived from toxicology and ecology towards designing improved personalized in vitro models of cancer. The framework serves as a tool for identifying optimal readouts and culture conditions, thus maximizing the information gained from each patient sample.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor/methods , Neoplasms/drug therapy , Precision Medicine/methods , Antineoplastic Agents/adverse effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Separation/methods , Drug Resistance, Neoplasm , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Patient Selection , Predictive Value of Tests , Primary Cell Culture , Signal Transduction/drug effects , Treatment Outcome , Tumor Cells, Cultured , Tumor Microenvironment
16.
PLoS One ; 10(1): e0116891, 2015.
Article in English | MEDLINE | ID: mdl-25607819

ABSTRACT

Breast cancers that express estrogen receptor alpha (ERα+) constitute the majority of breast tumors. Estrogen is a major driver of their growth, and targeting ER-mediated signals is a largely successful primary therapeutic strategy. Nonetheless, ERα+ tumors also result in the most breast cancer mortalities. Other factors, including altered characteristics of the extracellular matrix such as density and orientation and consequences for estrogen crosstalk with other hormones such as prolactin (PRL), may contribute to these poor outcomes. Here we employed defined three dimensional low density/compliant and high density/stiff collagen-I matrices to investigate the effects on 17ß-estradiol (E2) activity and PRL/E2 interactions in two well-characterized ERα+/PRLR+ luminal breast cancer cell lines in vitro. We demonstrate that matrix density modulated E2-induced transcripts, but did not alter the growth response. However, matrix density was a potent determinant of the behavioral outcomes of PRL/E2 crosstalk. High density/stiff matrices enhanced PRL/E2-induced growth mediated by increased activation of Src family kinases and insensitivity to the estrogen antagonist, 4-hydroxytamoxifen. It also permitted these hormones in combination to drive invasion and modify the alignment of collagen fibers. In contrast, low density/compliant matrices allowed modest if any cooperation between E2 and PRL to growth and did not permit hormone-induced invasion or collagen reorientation. Our studies demonstrate the power of matrix density to determine the outcomes of hormone actions and suggest that stiff matrices are potent collaborators of estrogen and PRL in progression of ERα+ breast cancer. Our evidence for bidirectional interactions between these hormones and the extracellular matrix provides novel insights into the regulation of the microenvironment of ERα+ breast cancer and suggests new therapeutic approaches.


Subject(s)
Breast Neoplasms/pathology , Collagen Type I/metabolism , Estradiol/metabolism , Extracellular Matrix/pathology , Prolactin/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen Type I/genetics , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Prolactin/pharmacology , Receptors, Prolactin/metabolism
17.
Adv Exp Med Biol ; 846: 201-20, 2015.
Article in English | MEDLINE | ID: mdl-25472540

ABSTRACT

Elevated exposure to prolactin (PRL) is epidemiologically associated with an increased risk of aggressive ER+ breast cancer. To understand the underlying mechanisms and crosstalk with other oncogenic factors, we developed the NRL-PRL mouse. In this model, mammary expression of a rat prolactin transgene raises local exposure to PRL without altering estrous cycling. Nulliparous females develop metastatic, histotypically diverse mammary carcinomas independent from ovarian steroids, and most are ER+. These characteristics resemble the human clinical disease, facilitating study of tumorigenesis, and identification of novel preventive and therapeutic approaches.


Subject(s)
Breast Neoplasms/pathology , Mammary Neoplasms, Experimental/pathology , Prolactin/physiology , Animals , Breast Neoplasms/genetics , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Prolactin/blood , Prolactin/genetics , Rats , Receptor Cross-Talk/physiology
18.
Proc Natl Acad Sci U S A ; 111(39): 14295-300, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25225411

ABSTRACT

The circadian clock plays a significant role in many aspects of female reproductive biology, including estrous cycling, ovulation, embryonic implantation, onset of puberty, and parturition. In an effort to link cell-specific circadian clocks to their specific roles in female reproduction, we used the promoter that controls expression of Steroidogenic Factor-1 (SF1) to drive Cre-recombinase-mediated deletion of the brain muscle arnt-like 1 (Bmal1) gene, known to encode an essential component of the circadian clock (SF1-Bmal1(-/-)). The resultant SF1-Bmal1(-/-) females display embryonic implantation failure, which is rescued by progesterone supplementation, or bilateral or unilateral transplantation of wild-type ovaries into SF1-Bmal1(-/-) dams. The observation that the central clock, and many other peripheral clocks, are fully functional in this model allows the assignment of the implantation phenotype to the clock in ovarian steroidogenic cells and distinguishes it from more general circadian related systemic pathology (e.g., early onset arthropathy, premature aging, ovulation, late onset of puberty, and abnormal estrous cycle). Our ovarian transcriptome analysis reveals that deletion of ovarian Bmal1 disrupts expression of transcripts associated with the circadian machinery and also genes critical for regulation of progesterone production, such as steroidogenic acute regulatory factor (Star). Overall, these data provide a powerful model to probe the interlocking and synergistic network of the circadian clock and reproductive systems.


Subject(s)
ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/physiology , Embryo Implantation/physiology , Ovary/cytology , Ovary/physiology , Steroids/biosynthesis , ARNTL Transcription Factors/genetics , Aging/genetics , Aging/physiology , Animals , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Embryo Implantation/drug effects , Embryo Implantation/genetics , Estrus/genetics , Estrus/physiology , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovary/transplantation , Pregnancy , Progesterone/administration & dosage , Promoter Regions, Genetic , Sexual Maturation/genetics , Sexual Maturation/physiology , Steroidogenic Factor 1/genetics
19.
Endocrinology ; 154(12): 4483-92, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24064365

ABSTRACT

Resistance of estrogen receptor positive (ERα+) breast cancers to antiestrogens is a major factor in the mortality of this disease. Although activation of ERα in the absence of ligand is hypothesized to contribute to this resistance, the potency of this mechanism in vivo is not clear. Epidemiologic studies have strongly linked prolactin (PRL) to both development of ERα+ breast cancer and resistance to endocrine therapies. Here we employed genetically modified mouse models to examine the ability of PRL and cross talk with TGFα to activate ERα, using a mutated ERα, ERα(G525L), which is refractory to endogenous estrogens. We demonstrate that PRL promotes pubertal ERα-dependent mammary ductal elongation and gene expression in the absence of estrogen, which are abrogated by the antiestrogen, ICI 182,780 (ICI). PRL and TGFα together reduce sensitivity to estrogen, and 30% of their combined stimulation of ductal proliferation is inhibited by ICI, implicating ligand-independent activation of ERα as a component of their interaction. However, PRL/TGFα-induced heterogeneous ERα+ tumors developed more rapidly in the presence of ICI and contained altered transcripts for surface markers associated with epithelial subpopulations and increased signal transducer and activator of transcription 5b expression. Together, these data support strong interactions between PRL and estrogen on multiple levels. Ligand-independent activation of ERα suggests that PRL may contribute to resistance to antiestrogen therapies. However, these studies also underscore ERα-mediated moderation of tumor phenotype. In light of the high expression of PRL receptors in ERα+ cancers, understanding the actions of PRL and cross talk with other oncogenic factors and ERα itself has important implications for therapeutic strategies.


Subject(s)
Estrogen Receptor alpha/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/metabolism , Prolactin/pharmacology , Aging , Animals , Carcinogenesis/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Female , Fulvestrant , Mice , Mice, Transgenic , Mutation , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism
20.
J Biol Chem ; 288(18): 12722-32, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23530035

ABSTRACT

Clinically, circulating prolactin levels and density of the extracellular matrix (ECM) are individual risk factors for breast cancer. As tumors develop, the surrounding stroma responds with increased deposition and cross-linking of the collagen matrix (desmoplasia). In mouse models, prolactin promotes mammary carcinomas that resemble luminal breast cancers in women, and increased collagen density promotes tumor metastasis and progression. Although the contributions of the ECM to the physiologic actions of prolactin are increasingly understood, little is known about the functional relationship between the ECM and prolactin signaling in breast cancer. Here, we examined consequences of increased ECM stiffness on prolactin signals to luminal breast cancer cells in three-dimensional collagen I matrices in vitro. We showed that matrix stiffness potently regulates a switch in prolactin signals from physiologic to protumorigenic outcomes. Compliant matrices promoted physiological prolactin actions and activation of STAT5, whereas stiff matrices promoted protumorigenic outcomes, including increased matrix metalloproteinase-dependent invasion and collagen scaffold realignment. In stiff matrices, prolactin increased SRC family kinase-dependent phosphorylation of focal adhesion kinase (FAK) at tyrosine 925, FAK association with the mitogen-activated protein kinase mediator GRB2, and pERK1/2. Stiff matrices also increased co-localization of prolactin receptors and integrin-activated FAK, implicating altered spatial relationships. Together, these results demonstrate that ECM stiffness is a powerful regulator of the spectrum of prolactin signals and that stiff matrices and prolactin interact in a feed-forward loop in breast cancer progression. Our study is the first reported evidence of altered ECM-prolactin interactions in breast cancer, suggesting the potential for new therapeutic approaches.


Subject(s)
Breast Neoplasms/metabolism , Collagen Type I/metabolism , Extracellular Matrix/metabolism , MAP Kinase Signaling System , Mammary Neoplasms, Animal/metabolism , Neoplasm Proteins/metabolism , Prolactin/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Collagen Type I/genetics , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Female , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/genetics , Phosphorylation/genetics , Prolactin/genetics , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...