Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Breast Cancer Res ; 25(1): 68, 2023 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-37312163

RESUMO

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.


Assuntos
Neoplasias , Receptores de Estrogênio , Animais , Camundongos , Fulvestranto/farmacologia , Imunoterapia , Estrogênios , Antagonistas de Estrogênios , Imunossupressores
2.
J Assist Reprod Genet ; 35(9): 1631-1640, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30069849

RESUMO

PURPOSE: Expanded carrier screening (ECS) is an available component of preconception and prenatal care. There is complexity around offering, administering, and following-up test results. The goal of this study is to evaluate current physicians' utilization and attitudes towards ECS in current practice. METHODS: This was a prospective qualitative survey study. A 32-question electronic survey was distributed during a 1-year period to obstetricians-gynecologists who were identified using a Qualtrics listserv database. RESULTS: While more than 90% of physicians offered ethnic-based carrier screening (CS), ECS was offered significantly less (2010, 20.6%, and 2016, 27.1%). Physicians who were not fellowship-trained in reproductive endocrinology and infertility (REI) preferred ethnic-based carrier screening (95.9 vs 16.8%; P < 0.001). REI subspecialists were more likely to offer ECS (80%) compared to 70% of maternal fetal medicine physicians (MFM). Physicians were comfortable discussing negative results (53.6%) compared to positive results (48.4%). Most physicians (56%) believed that ECS should not be offered until the significance of each disease is understood; 52% believed that testing should be restricted to those conditions important to couples; while 26% felt that testing should be done regardless of the clinical significance. CONCLUSIONS: Discussion and application of ECS has increased in clinical practice. However, lack of comfort with counseling and varying beliefs surrounding ECS continue to hinder its utilization. Further education and training programs, and subsequent evaluation are warranted.


Assuntos
Educação Médica , Triagem de Portadores Genéticos/tendências , Aconselhamento Genético/tendências , Infertilidade/epidemiologia , Adulto , Endocrinologia/educação , Feminino , Humanos , Masculino , Médicos , Cuidado Pré-Natal/tendências , Medicina Reprodutiva/educação , Inquéritos e Questionários
3.
Breast Cancer Res ; 19(1): 9, 2017 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-28103936

RESUMO

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.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Colágeno Tipo I/metabolismo , Receptor alfa de Estrogênio/metabolismo , Prolactina/metabolismo , Transdução de Sinais , Animais , Biomarcadores , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células , Colágeno Tipo I/genética , Progressão da Doença , Matriz Extracelular/metabolismo , Feminino , Imuno-Histoquímica , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Estadiamento de Neoplasias , Células Neoplásicas Circulantes , Proteínas Proto-Oncogênicas c-akt , Fator de Transcrição STAT5/metabolismo , Carga Tumoral
4.
Adv Exp Med Biol ; 846: 201-20, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25472540

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Neoplasias Mamárias Experimentais/patologia , Prolactina/fisiologia , Animais , Neoplasias da Mama/genética , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Transgênicos , Prolactina/sangue , Prolactina/genética , Ratos , Receptor Cross-Talk/fisiologia
5.
Am J Pathol ; 181(1): 294-302, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22658484

RESUMO

Epidemiological and experimental studies have revealed an important role for prolactin (PRL) in breast cancer. Cyclin D1 is a major downstream target of PRL in lobuloalveolar development during pregnancy and is amplified and/or overexpressed in many breast carcinomas. To examine the importance of cyclin D1 in PRL-induced pathogenesis, we generated transgenic mice (NRL-PRL) that overexpress PRL in mammary epithelial cells, with wild-type, heterozygous, or genetically ablated cyclin D1 in the FVB/N genetic background. Although loss of one cyclin D1 allele did not affect PRL-induced mammary lesions in nonparous females, the complete absence of cyclin D1 (D1(-/-)) markedly decreased tumor incidence. Nevertheless, NRL-PRL/D1(-/-) females developed significantly more preneoplastic lesions (eg, epithelial hyperplasias and mammary intraepithelial neoplasias) than D1(-/-) females. Moreover, although lack of cyclin D1 reduced proliferation of morphologically normal mammary epithelium, transgenic PRL restored it to rates of wild-type females. PRL posttranscriptionally increased nuclear cyclin D3 protein in D1(-/-) luminal cells, indicating one compensatory mechanism. Consistently, pregnancy induced extensive lobuloalveolar growth in the absence of cyclin D1. However, transcripts for milk proteins were reduced, and pups failed to survive, suggesting that mammary differentiation was inadequate. Together, these results indicate that cyclin D1 is an important, but not essential, mediator of PRL-induced mammary proliferation and pathology in FVB/N mice and is critical for differentiation and lactation.


Assuntos
Adenocarcinoma/fisiopatologia , Ciclina D1/fisiologia , Neoplasias Mamárias Experimentais/fisiopatologia , Prolactina/fisiologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Ciclina D1/deficiência , Ciclina D1/metabolismo , Ciclina D3/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Feminino , Lactação/fisiologia , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Gravidez , Prolactina/metabolismo
6.
Proc Natl Acad Sci U S A ; 107(31): 13570-5, 2010 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-20643955

RESUMO

Nearly 75% of in vitro fertilization (IVF) treatments do not result in live births and patients are largely guided by a generalized age-based prognostic stratification. We sought to provide personalized and validated prognosis by using available clinical and embryo data from prior, failed treatments to predict live birth probabilities in the subsequent treatment. We generated a boosted tree model, IVFBT, by training it with IVF outcomes data from 1,676 first cycles (C1s) from 2003-2006, followed by external validation with 634 cycles from 2007-2008, respectively. We tested whether this model could predict the probability of having a live birth in the subsequent treatment (C2). By using nondeterministic methods to identify prognostic factors and their relative nonredundant contribution, we generated a prediction model, IVF(BT), that was superior to the age-based control by providing over 1,000-fold improvement to fit new data (p<0.05), and increased discrimination by receiver-operative characteristic analysis (area-under-the-curve, 0.80 vs. 0.68 for C1, 0.68 vs. 0.58 for C2). IVFBT provided predictions that were more accurate for approximately 83% of C1 and approximately 60% of C2 cycles that were out of the range predicted by age. Over half of those patients were reclassified to have higher live birth probabilities. We showed that data from a prior cycle could be used effectively to provide personalized and validated live birth probabilities in a subsequent cycle. Our approach may be replicated and further validated in other IVF clinics.


Assuntos
Fertilização in vitro/estatística & dados numéricos , Nascido Vivo , Resultado da Gravidez , Adulto , Distribuição por Idade , Biometria , Criopreservação , Feminino , Humanos , Masculino , Fenótipo , Gravidez , Taxa de Gravidez , Probabilidade
7.
Front Endocrinol (Lausanne) ; 13: 910978, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35784527

RESUMO

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.


Assuntos
Neoplasias da Mama , Prolactina , Neoplasias da Mama/epidemiologia , Progressão da Doença , Feminino , Humanos , Recém-Nascido , Lactação , Estudos Prospectivos
8.
Nat Commun ; 13(1): 4948, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35999216

RESUMO

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.


Assuntos
Nanopartículas Multifuncionais , Neoplasias , Animais , Antígenos de Neoplasias , Linhagem Celular Tumoral , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia/métodos , Camundongos , Neoplasias/radioterapia , Microambiente Tumoral , Vacinação
9.
Cancer Lett ; 503: 231-239, 2021 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-33472091

RESUMO

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.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Neoplasias Mamárias Experimentais/patologia , Mutagênicos/toxicidade , Prolactina/genética , Animais , Núcleo Celular/metabolismo , Proliferação de Células , Feminino , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Transgênicos , Receptores Notch/metabolismo , Via de Sinalização Wnt
10.
BJU Int ; 104(7): 986-90, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19549262

RESUMO

OBJECTIVE: To evaluate the effect of preoperative risk factors on perioperative outcomes up to 3 months after robot-assisted radical cystectomy (RARC), as RC continues to be associated with a high rate of morbidity and mortality. PATIENTS AND METHODS: From 2005 to 2007, 66 consecutive patients had RARC at Roswell Park Cancer Institute. Patient demographics, preoperative risk factors and complications up to 3 months after RARC were reviewed from a prospective quality-assurance database. Patients were stratified into high- and low risk groups based on age, previous abdominal surgery, chronic obstructive pulmonary disease (COPD), body mass index (BMI), Revised Cardiac Risk Index (RCRI) and American Society of Anesthesiologists (ASA) score. RESULTS: Age, previous abdominal surgery, COPD, BMI, RCRI score and ASA score did not significantly influence complications during or up to 3 months following RARC (P > 0.05). Advanced age was associated with a higher RCRI score (P = 0.014) and an increased likelihood of admission to the Intensive Care Unit (P = 0.007). A higher ASA score was associated with an increased overall hospital stay (P = 0.039). Previous abdominal surgery was associated with more frequent unscheduled postoperative clinic visits (P = 0.014). Operative duration did not significantly influence complication rates (P > 0.05). Fifteen of 62 patients (24%) had a major complication, while 15 (24%) had minor complications within 3 months of surgery. The reoperation rate was 11% and the overall mortality rate was 1.6%. CONCLUSIONS: RARC appears to be well tolerated, independent of comorbid risk factors such as age, BMI, RCRI and ASA score.


Assuntos
Cistectomia/efeitos adversos , Robótica , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Perda Sanguínea Cirúrgica , Índice de Massa Corporal , Cistectomia/métodos , Humanos , Laparoscopia/efeitos adversos , Laparoscopia/métodos , Tempo de Internação , Pessoa de Meia-Idade , Complicações Pós-Operatórias/etiologia , Estudos Prospectivos , Reoperação , Fatores de Risco , Resultado do Tratamento
11.
Oncogene ; 38(43): 6913-6925, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31406251

RESUMO

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.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Matriz Extracelular/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Estradiol/metabolismo , Antagonistas de Estrogênios/farmacologia , Matriz Extracelular/efeitos dos fármacos , Feminino , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Camundongos , Tamoxifeno/farmacologia , Microambiente Tumoral/efeitos dos fármacos
12.
Cell Rep ; 28(6): 1526-1537.e4, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31390566

RESUMO

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.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Neoplasias Mamárias Experimentais/etiologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Envelhecimento/metabolismo , Animais , Carcinogênese/genética , Conjuntos de Dados como Assunto , Feminino , Perfilação da Expressão Gênica , Humanos , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Prolactina/genética , Prolactina/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Ratos , Transdução de Sinais , Transgenes
13.
Cancer Lett ; 433: 1-9, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29935374

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Colágeno/metabolismo , Receptor alfa de Estrogênio/metabolismo , Neoplasias Pulmonares/secundário , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Feminino , Fulvestranto/administração & dosagem , Fulvestranto/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Transplante de Neoplasias , Células-Tronco Neoplásicas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sirolimo/administração & dosagem , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Carga Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
14.
Cancer Res ; 78(7): 1672-1684, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29363543

RESUMO

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.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Plasticidade Celular/efeitos dos fármacos , Antagonistas do Receptor de Estrogênio/uso terapêutico , Receptor alfa de Estrogênio/antagonistas & inibidores , Neoplasias Mamárias Animais/tratamento farmacológico , Células-Tronco Neoplásicas/patologia , Adenocarcinoma/induzido quimicamente , Adenocarcinoma/patologia , Animais , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Receptor alfa de Estrogênio/metabolismo , Feminino , Fulvestranto/uso terapêutico , Humanos , Neoplasias Mamárias Animais/induzido quimicamente , Neoplasias Mamárias Animais/patologia , Camundongos , Prolactina/efeitos adversos , Células Tumorais Cultivadas , Via de Sinalização Wnt/fisiologia
15.
Mol Cell Biol ; 24(1): 186-91, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14673154

RESUMO

Tumor suppressor proteins must be exquisitely regulated since they can induce cell death while preventing cancer. For example, the p19(ARF) tumor suppressor (p14(ARF) in humans) appears to stimulate the apoptotic function of the p53 tumor suppressor to prevent lymphomagenesis and carcinogenesis induced by oncogene overexpression. Here we present a genetic approach to defining the role of p19(ARF) in regulating the apoptotic function of p53 in highly proliferating, homeostatic tissues. In contrast to our expectation, p19(ARF) did not activate the apoptotic function of p53 in lymphocytes or epithelial cells. These results demonstrate that the mechanisms that control p53 function during homeostasis differ from those that are critical for tumor suppression. Moreover, the Mdm2/p53/p19(ARF) pathway appears to exist only under very restricted conditions.


Assuntos
Homeostase/fisiologia , Proteínas Nucleares , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Peso Corporal/genética , Inibidor p16 de Quinase Dependente de Ciclina , Epitélio/metabolismo , Intestino Delgado/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2 , Proteína Supressora de Tumor p14ARF/genética
16.
Mol Cell Biol ; 23(2): 462-72, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12509446

RESUMO

The function of the p53 tumor suppressor protein must be highly regulated because p53 can cause cell death and prevent tumorigenesis. In cultured cells, the p90MDM2 protein blocks the transcriptional activation domain of p53 and also stimulates the degradation of p53. Here we provide the first conclusive demonstration that p90MDM2 constitutively regulates p53 activity in homeostatic tissues. Mice with a hypomorphic allele of mdm2 revealed a heretofore unknown role for mdm2 in lymphopoiesis and epithelial cell survival. Phenotypic analyses revealed that both the transcriptional activation and apoptotic functions of p53 were increased in these mice. However, the level of p53 protein was not coordinately increased, suggesting that p90MDM2 can inhibit the transcriptional activation and apoptotic functions of p53 in a manner independent of degradation. Cre-mediated deletion of mdm2 caused a greater accumulation of p53, demonstrating that p90MDM2 constitutively regulates both the activity and the level of p53 in homeostatic tissues. The observation that only a subset of tissues with activated p53 underwent apoptosis indicates that factors other than p90(MDM2) determine the physiological consequences of p53 activation. Furthermore, reduction of mdm2 in vivo resulted in radiosensitivity, highlighting the importance of mdm2 as a potential target for adjuvant cancer therapies.


Assuntos
Regulação Neoplásica da Expressão Gênica , Linfócitos/citologia , Proteínas Nucleares , Proteínas Proto-Oncogênicas/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Alelos , Animais , Apoptose , Northern Blotting , Western Blotting , Peso Corporal/genética , Morte Celular , Células Cultivadas , Relação Dose-Resposta à Radiação , Citometria de Fluxo , Deleção de Genes , Genótipo , Células-Tronco Hematopoéticas , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Genéticos , Testes de Precipitina , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-mdm2 , RNA Mensageiro/metabolismo , Fatores de Tempo , Distribuição Tecidual , Transcrição Gênica , Ativação Transcricional , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/fisiologia
17.
Can J Urol ; 14(6): 3753-6, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18163928

RESUMO

INTRODUCTION: To date, no study has compared postoperative pain and requirement for pain medications in open versus robot-assisted radical cystectomy. Patient reported pain and opiate use were reviewed retrospectively using prospectively collected data from postoperative day one to day of discharge. MATERIALS AND METHODS: Twenty consecutive robot-assisted radical cystectomy patients were compared to the prior 20 patients who underwent open radical cystectomy. Data was collected prospectively to determine opiate requirements and pain scores in each group. Daily opiate use was converted to morphine sulfate equivalents (MSE) to facilitate comparison. A Likert pain perception scale was used to assess perceived pain. Statistical models were used to test for differences in opiate usage and pain perception between groups of patients who underwent open versus robot-assisted surgery. RESULTS: Seven patients were excluded from the study (three from the open group, and four from the robotic group): five due to preoperative opiate usage, one due to missing pain data, and one whose procedure was aborted due to unresectable disease. All patients were similar with respect to age, body mass index and pathological parameters. Average MSE usage differed significantly between the two groups on all postoperative days (p < 0.007) whereas average pain scores were similar in the two groups. CONCLUSION: Patients who underwent robot-assisted radical cystectomy achieved similar pain control but required less opiates than those who underwent open radical cystectomy.


Assuntos
Cistectomia/métodos , Dor Pós-Operatória/tratamento farmacológico , Robótica , Idoso , Analgésicos/uso terapêutico , Humanos , Dor Pós-Operatória/epidemiologia , Estudos Retrospectivos
18.
Stem Cell Reports ; 9(4): 1167-1179, 2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-28919264

RESUMO

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.


Assuntos
Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Ovário/metabolismo , Prolactina/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Esteroides/metabolismo , Fatores Etários , Animais , Biomarcadores , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Células Epiteliais/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Feminino , Expressão Gênica , Hormônios Esteroides Gonadais/metabolismo , Camundongos , Camundongos Transgênicos , Progesterona/metabolismo , Ratos , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
19.
Urology ; 97: 124-129, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27491964

RESUMO

OBJECTIVE: To develop quality assessment tool to evaluate surgical performance for robot-assisted radical cystectomy program. METHODS: A prospectively maintained quality assurance database of 425 consecutive robot-assisted radical cystectomies performed by a single surgeon between 2005 and 2015 was retrospectively reviewed. Potentially modifiable factors, related to the management and perioperative care of patients, were used to evaluate patient care. Criteria included the following: preoperative (administration of neoadjuvant chemotherapy); operative (operative time <6.5 hours and estimated blood loss <500 cc); pathologic (negative soft tissue surgical margins and lymph node yield ≥20); and postoperative (no high-grade complications, readmission, or noncancer-related mortality within 30 days).The Quality Cystectomy Score (QCS) was developed (1 star: achieving ≤2 criteria or mortality within 30 days; 2 stars: 3 or 4 criteria met; 3 stars: 5 or 6 criteria met; and 4 stars: 7 or all criteria met). Univariate and multivariate Cox proportional hazard regression models were fitted to test for the association between QCS and survival outcomes. RESULTS: Most patients (85%) achieved at least 3 stars, and more patients achieved 4 stars with time. High QCS was associated with better recurrence-free, cancer-specific, and overall survival (P values <.05). None of the patients with 1-star were alive at 1 year. Patients with 4 stars achieved the best survival rates (recurrence-free survival [62%], cancer-specific survival [70%], and overall survival [53%] at 5 years) (log rank P < .0001). CONCLUSION: Continuous assessment for quality improvement facilitated implementation and maintenance of robot-assisted program for bladder cancer.


Assuntos
Cistectomia/métodos , Recidiva Local de Neoplasia/patologia , Garantia da Qualidade dos Cuidados de Saúde , Procedimentos Cirúrgicos Robóticos/métodos , Neoplasias da Bexiga Urinária/cirurgia , Idoso , Cistectomia/efeitos adversos , Bases de Dados Factuais , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Recidiva Local de Neoplasia/fisiopatologia , Recidiva Local de Neoplasia/cirurgia , Estadiamento de Neoplasias , Complicações Pós-Operatórias/mortalidade , Complicações Pós-Operatórias/fisiopatologia , Estudos Retrospectivos , Medição de Risco , Procedimentos Cirúrgicos Robóticos/efeitos adversos , Análise de Sobrevida , Fatores de Tempo , Resultado do Tratamento , Neoplasias da Bexiga Urinária/mortalidade , Neoplasias da Bexiga Urinária/patologia
20.
Arch Intern Med ; 164(21): 2343-8, 2004 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-15557413

RESUMO

BACKGROUND: Effective therapy for chronic illness requires daily medication adherence (DMA) for prolonged periods. Overall medical regimen complexity may represent one barrier to successful adherence. METHODS: To assess the relationship between the number of concurrently prescribed medicines and adherence to 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins), we analyzed a cohort of 5488 patients in a single health insurance plan who began statin therapy between July 1, 1999, and June 30, 2002. We assessed 2 parameters of statin adherence: (1) DMA ([total number of pills dispensed/total number of days between first and last prescription] x100) and (2) refill persistence (RP) (consecutive months of refills after initial prescription). RESULTS: The cohort was 61.6% male, with a mean +/- SD age of 52.7 +/- 9.3 years. Patients were prescribed a mean +/- SD of 2.9 +/- 2 total medicines (range, 1-13), with a mean +/- SD statin DMA of 82.1% +/- 26.5%. By 12 months, only 68% of patients continued filling statin prescriptions. After controlling for age, income level, and treatment for hypertension or ischemic heart disease, a greater number of concurrently prescribed medicines was significantly associated with better DMA (P = .005) and longer RP (P = .03). CONCLUSIONS: In this cohort, statin DMA was generally adequate, but RP was suboptimal. Patients with more concurrently prescribed medicines had higher DMA and better RP, even after adjusting for demographic factors and cardiovascular comorbidity. Physicians should not be deterred from initiating statin therapy by a patient's medical regimen complexity but should be alert for lack of therapy persistence, particularly in younger and healthier patients.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Cooperação do Paciente , Polimedicação , Adulto , Fatores Etários , Estudos de Coortes , Prescrições de Medicamentos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores Socioeconômicos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA