Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 16(3)2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-38339316

RESUMO

For over a century, early researchers sought to study biological organisms in a laboratory setting, leading to the generation of both in vitro and in vivo model systems. Patient-derived models of cancer (PDMCs) have more recently come to the forefront of preclinical cancer models and are even finding their way into clinical practice as part of functional precision medicine programs. The PDMC Consortium, supported by the Division of Cancer Biology in the National Cancer Institute of the National Institutes of Health, seeks to understand the biological principles that govern the various PDMC behaviors, particularly in response to perturbagens, such as cancer therapeutics. Based on collective experience from the consortium groups, we provide insight regarding PDMCs established both in vitro and in vivo, with a focus on practical matters related to developing and maintaining key cancer models through a series of vignettes. Although every model has the potential to offer valuable insights, the choice of the right model should be guided by the research question. However, recognizing the inherent constraints in each model is crucial. Our objective here is to delineate the strengths and limitations of each model as established by individual vignettes. Further advances in PDMCs and the development of novel model systems will enable us to better understand human biology and improve the study of human pathology in the lab.

2.
STAR Protoc ; 4(3): 102402, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37402170

RESUMO

Patient-derived xenografts (PDXs) have clinical value but are time-, cost-, and labor-intensive and thus ill-suited for large-scale experiments. Here, we present a protocol to convert PDX tumors into PDxOs for long-term cultures amenable to moderate-throughput drug screens, including in-depth PDxO validation. We describe steps for PDxO preparation and mouse cell removal. We then detail PDxO validation and characterization and drug response assay. Our PDxO drug screening platform can predict therapy response in vivo and inform functional precision oncology for patients. For complete details on the use and execution of this protocol, please refer to Guillen et al.1.


Assuntos
Neoplasias da Mama , Humanos , Animais , Camundongos , Feminino , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Medicina de Precisão , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos/métodos
3.
Elife ; 122023 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-36876914

RESUMO

Recent studies reveal that lateral mitochondrial transfer, the movement of mitochondria from one cell to another, can affect cellular and tissue homeostasis. Most of what we know about mitochondrial transfer stems from bulk cell studies and have led to the paradigm that functional transferred mitochondria restore bioenergetics and revitalize cellular functions to recipient cells with damaged or non-functional mitochondrial networks. However, we show that mitochondrial transfer also occurs between cells with functioning endogenous mitochondrial networks, but the mechanisms underlying how transferred mitochondria can promote such sustained behavioral reprogramming remain unclear. We report that unexpectedly, transferred macrophage mitochondria are dysfunctional and accumulate reactive oxygen species in recipient cancer cells. We further discovered that reactive oxygen species accumulation activates ERK signaling, promoting cancer cell proliferation. Pro-tumorigenic macrophages exhibit fragmented mitochondrial networks, leading to higher rates of mitochondrial transfer to cancer cells. Finally, we observe that macrophage mitochondrial transfer promotes tumor cell proliferation in vivo. Collectively these results indicate that transferred macrophage mitochondria activate downstream signaling pathways in a ROS-dependent manner in cancer cells, and provide a model of how sustained behavioral reprogramming can be mediated by a relatively small amount of transferred mitochondria in vitro and in vivo.


Assuntos
Mitocôndrias , Neoplasias , Humanos , Espécies Reativas de Oxigênio/metabolismo , Mitocôndrias/metabolismo , Neoplasias/patologia , Transdução de Sinais , Proliferação de Células
4.
Commun Biol ; 5(1): 794, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35941353

RESUMO

Quantitative phase imaging (QPI) measures the growth rate of individual cells by quantifying changes in mass versus time. Here, we use the breast cancer cell lines MCF-7, BT-474, and MDA-MB-231 to validate QPI as a multiparametric approach for determining response to single-agent therapies. Our method allows for rapid determination of drug sensitivity, cytotoxicity, heterogeneity, and time of response for up to 100,000 individual cells or small clusters in a single experiment. We find that QPI EC50 values are concordant with CellTiter-Glo (CTG), a gold standard metabolic endpoint assay. In addition, we apply multiparametric QPI to characterize cytostatic/cytotoxic and rapid/slow responses and track the emergence of resistant subpopulations. Thus, QPI reveals dynamic changes in response heterogeneity in addition to average population responses, a key advantage over endpoint viability or metabolic assays. Overall, multiparametric QPI reveals a rich picture of cell growth by capturing the dynamics of single-cell responses to candidate therapies.


Assuntos
Antineoplásicos , Neoplasias da Mama , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Proliferação de Células , Avaliação Pré-Clínica de Medicamentos , Detecção Precoce de Câncer , Feminino , Humanos
5.
Nat Cancer ; 3(2): 232-250, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35221336

RESUMO

Models that recapitulate the complexity of human tumors are urgently needed to develop more effective cancer therapies. We report a bank of human patient-derived xenografts (PDXs) and matched organoid cultures from tumors that represent the greatest unmet need: endocrine-resistant, treatment-refractory and metastatic breast cancers. We leverage matched PDXs and PDX-derived organoids (PDxO) for drug screening that is feasible and cost-effective with in vivo validation. Moreover, we demonstrate the feasibility of using these models for precision oncology in real time with clinical care in a case of triple-negative breast cancer (TNBC) with early metastatic recurrence. Our results uncovered a Food and Drug Administration (FDA)-approved drug with high efficacy against the models. Treatment with this therapy resulted in a complete response for the individual and a progression-free survival (PFS) period more than three times longer than their previous therapies. This work provides valuable methods and resources for functional precision medicine and drug development for human breast cancer.


Assuntos
Organoides , Neoplasias de Mama Triplo Negativas , Descoberta de Drogas , Xenoenxertos , Humanos , Medicina de Precisão/métodos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Estados Unidos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Breast Cancer Res ; 23(1): 100, 2021 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-34717714

RESUMO

BACKGROUND: Metastatic breast cancer (MBC) is incurable, with a 5-year survival rate of 28%. In the USA, more than 42,000 patients die from MBC every year. The most common type of breast cancer is estrogen receptor-positive (ER+), and more patients die from ER+ breast cancer than from any other subtype. ER+ tumors can be successfully treated with hormone therapy, but many tumors acquire endocrine resistance, at which point treatment options are limited. There is an urgent need for model systems that better represent human ER+ MBC in vivo, where tumors can metastasize. Patient-derived xenografts (PDX) made from MBC spontaneously metastasize, but the immunodeficient host is a caveat, given the known role of the immune system in tumor progression and response to therapy. Thus, we attempted to develop an immune-humanized PDX model of ER+ MBC. METHODS: NSG-SGM3 mice were immune-humanized with CD34+ hematopoietic stem cells, followed by engraftment of human ER+ endocrine resistant MBC tumor fragments. Strategies for exogenous estrogen supplementation were compared, and immune-humanization in blood, bone marrow, spleen, and tumors was assessed by flow cytometry and tissue immunostaining. Characterization of the new model includes assessment of the human tumor microenvironment performed by immunostaining. RESULTS: We describe the development of an immune-humanized PDX model of estrogen-independent endocrine resistant ER+ MBC. Importantly, our model harbors a naturally occurring ESR1 mutation, and immune-humanization recapitulates the lymphocyte-excluded and myeloid-rich tumor microenvironment of human ER+ breast tumors. CONCLUSION: This model sets the stage for development of other clinically relevant models of human breast cancer and should allow future studies on mechanisms of endocrine resistance and tumor-immune interactions in an immune-humanized in vivo setting.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos , Receptores de Estrogênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Antígenos CD34/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Receptor alfa de Estrogênio/genética , Estrogênios/administração & dosagem , Estrogênios/farmacologia , Feminino , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Xenoenxertos/efeitos dos fármacos , Xenoenxertos/metabolismo , Xenoenxertos/patologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Mutação , Receptores de Estrogênio/genética , Microambiente Tumoral/imunologia
7.
Oncogene ; 40(47): 6494-6512, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34611309

RESUMO

Expression of the immediate-early response gene IER2 has been associated with the progression of several types of cancer, but its functional role is poorly understood. We found that increased IER2 expression in human melanoma is associated with shorter overall survival, and subsequently investigated the mechanisms through which IER2 exerts this effect. In experimental melanoma models, sustained expression of IER2 induced senescence in a subset of melanoma cells in a p53/MAPK/AKT-dependent manner. The senescent cells produced a characteristic secretome that included high levels of the extracellular phosphoglycoprotein osteopontin. Nuclear localization of the IER2 protein was critical for both the induction of senescence and osteopontin secretion. Osteopontin secreted by IER2-expressing senescent cells strongly stimulated the migration and invasion of non-senescent melanoma cells. Consistently, we observed coordinate expression of IER2, p53/p21, and osteopontin in primary human melanomas and metastases, highlighting the pathophysiological relevance of IER2-mediated senescence in melanoma progression. Together, our study reveals that sustained IER2 expression drives melanoma invasion and progression through stimulating osteopontin secretion via the stochastic induction of senescence.


Assuntos
Biomarcadores Tumorais/metabolismo , Senescência Celular , Regulação Neoplásica da Expressão Gênica , Proteínas Imediatamente Precoces/metabolismo , Melanoma/patologia , Osteopontina/metabolismo , Transativadores/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Humanos , Proteínas Imediatamente Precoces/genética , Melanoma/genética , Melanoma/metabolismo , Camundongos , Invasividade Neoplásica , Osteopontina/genética , Prognóstico , Transativadores/genética , Células Tumorais Cultivadas
8.
Cancer Res ; 80(6): 1234-1245, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-32046982

RESUMO

Estrogen signaling through estrogen receptor alpha (ER) plays a major role in endometrial cancer risk and progression, however, the molecular mechanisms underlying ER's regulatory role in endometrial cancer are poorly understood. In breast cancer cells, ER genomic binding is enabled by FOXA1 and GATA3, but the transcription factors that control ER genomic binding in endometrial cancer cells remain unknown. We previously identified ETV4 as a candidate factor controlling ER genomic binding in endometrial cancer cells, and here we explore the functional importance of ETV4. Homozygous deletion of ETV4, using CRISPR/Cas9, led to greatly reduced ER binding at the majority of loci normally bound by ER. Consistent with the dramatic loss of ER binding, the gene expression response to estradiol was dampened for most genes. ETV4 contributes to estrogen signaling in two distinct ways. ETV4 loss affects chromatin accessibility at some ER bound loci and impairs ER nuclear translocation. The diminished estrogen signaling upon ETV4 deletion led to decreased growth, particularly in 3D culture, where hollow organoids were formed and in vivo in the context of estrogen-dependent growth. These results show that ETV4 plays an important role in estrogen signaling in endometrial cancer cells. SIGNIFICANCE: Estrogen receptor alpha (ER) is a key oncogene in endometrial cancer. This study uncovers ETV4 as an important factor in controlling the activity of ER and the growth of endometrial cancer cells. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/6/1234/F1.large.jpg.


Assuntos
Neoplasias do Endométrio/genética , Receptor alfa de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas Proto-Oncogênicas c-ets/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Cromatina/metabolismo , Sequenciamento de Cromatina por Imunoprecipitação , Citoplasma/metabolismo , Neoplasias do Endométrio/patologia , Estradiol/metabolismo , Feminino , Técnicas de Inativação de Genes , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-ets/genética , RNA-Seq , Transdução de Sinais/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Nat Commun ; 11(1): 798, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-32034142

RESUMO

Multiple myeloma (MM) is a plasma cell malignancy and most patients eventually succumb to the disease. Chimeric antigen receptor (CAR) T cells targeting B-Cell Maturation Antigen (BCMA) on MM cells have shown high-response rates, but limited durability. CD229/LY9 is a cell surface receptor present on B and T lymphocytes that is universally and strongly expressed on MM plasma cells. Here, we develop CD229 CAR T cells that are highly active in vitro and in vivo against MM plasma cells, memory B cells, and MM-propagating cells. We do not observe fratricide during CD229 CAR T cell production, as CD229 is downregulated in T cells during activation. In addition, while CD229 CAR T cells target normal CD229high T cells, they spare functional CD229neg/low T cells. These findings indicate that CD229 CAR T cells may be an effective treatment for patients with MM.


Assuntos
Imunoterapia Adotiva/métodos , Mieloma Múltiplo/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Animais , Anticorpos/imunologia , Linfócitos B/metabolismo , Humanos , Células K562/imunologia , Masculino , Camundongos Endogâmicos NOD , Mieloma Múltiplo/patologia , Receptores de Antígenos de Linfócitos T/metabolismo , Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Família de Moléculas de Sinalização da Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Int J Cancer ; 147(4): 1190-1198, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31675122

RESUMO

Metastasis is a multistep process, during which circulating tumor cells traffic through diverse anatomical locations. Stable inducible marking of tumor cells in a manner that is tightly spatially and temporally controlled would allow tracking the contribution of cells passing through specific locations to metastatic dissemination. For example, tumor cells enter the lymphatic system and can form metastases in regional lymph nodes, but the relative contribution of tumor cells that traffic through the lymphatic system to the formation of distant metastases remains controversial. Here, we developed a novel genetic switch based on mild transient warming (TW) that allows cells to be marked in a defined spatiotemporal manner in vivo. Prior to warming, cells express only EGFP. Upon TW, the EGFP gene is excised and expression of mCherry is permanently turned on. We employed this system in an experimental pancreatic cancer model and used localized TW to induce the genetic switch in tumor cells trafficking through tumor-draining lymph nodes. Thereby we found that tumor cells disseminating via the lymphatics make a major contribution to the seeding of lung metastases. The inducible genetic marking system we have developed is a powerful tool for the tracking of metastasizing cells in vivo.


Assuntos
Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Células Neoplásicas Circulantes/metabolismo , Animais , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Linfonodos/patologia , Metástase Linfática , Sistema Linfático/patologia , Neoplasias/metabolismo , Neoplasias/patologia , Células Neoplásicas Circulantes/patologia , Ratos , Análise Espaço-Temporal , Proteína Vermelha Fluorescente
11.
Cell Rep ; 17(5): 1344-1356, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27783948

RESUMO

While chemotherapy strongly restricts or reverses tumor growth, the response of host tissue to therapy can counteract its anti-tumor activity by promoting tumor re-growth and/or metastases, thus limiting therapeutic efficacy. Here, we show that vascular endothelial growth factor receptor 3 (VEGFR3)-expressing macrophages infiltrating chemotherapy-treated tumors play a significant role in metastasis. They do so in part by inducing lymphangiogenesis as a result of cathepsin release, leading to VEGF-C upregulation by heparanase. We found that macrophages from chemotherapy-treated mice are sufficient to trigger lymphatic vessel activity and structure in naive tumors in a VEGFR3-dependent manner. Blocking VEGF-C/VEGFR3 axis inhibits the activity of chemotherapy-educated macrophages, leading to reduced lymphangiogenesis in treated tumors. Overall, our results suggest that disrupting the VEGF-C/VEGFR3 axis not only directly inhibits lymphangiogenesis but also blocks the pro-metastatic activity of macrophages in chemotherapy-treated mice.


Assuntos
Linfangiogênese , Macrófagos/patologia , Paclitaxel/farmacologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Catepsinas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Glucuronidase/metabolismo , Humanos , Linfangiogênese/efeitos dos fármacos , Vasos Linfáticos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Fenótipo , Regulação para Cima/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/sangue , Fator C de Crescimento do Endotélio Vascular/metabolismo
12.
PLoS One ; 11(9): e0162221, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27589056

RESUMO

In patients with low-risk breast cancer, intraoperative radiotherapy (IORT) during breast-conserving surgery is a novel and convenient treatment option for delivering a single high dose of irradiation directly to the tumour bed. However, edema and fibrosis can develop after surgery and radiotherapy, which can subsequently impair quality of life. TGF- ß is a strong inducer of the extracellular matrix component hyaluronan (HA). TGF-ß expression and HA metabolism can be modulated by irradiation experimentally, and are involved in edema and fibrosis. We therefore hypothesized that IORT may regulate these factors.Wound fluid (WF) draining from breast lumpectomy sites was collected and levels of TGF-ß1 and HA were determined by ELISA. Proliferation and marker expression was analyzed in primary lymphatic endothelial cells (LECs) treated with recombinant TGF-ß or WF. Our results show that IORT does not change TGF-ß1 or HA levels in wound fluid draining from breast lumpectomy sites, and does not lead to accumulation of sHA oligosaccharides. Nevertheless, concentrations of TGF-ß1 were high in WF from patients regardless of IORT, at concentrations well above those associated with fibrosis and the suppression of LEC identity. Consistently, we found that TGF-ß in WF is active and inhibits LEC proliferation. Furthermore, all three TGF-ß isoforms inhibited LEC proliferation and suppressed LEC marker expression at pathophysiologically relevant concentrations. Given that TGF-ß contributes to edema and plays a role in the regulation of LEC identity, we suggest that inhibition of TGF-ß directly after surgery might prevent the development of side effects such as edema and fibrosis.


Assuntos
Neoplasias da Mama/cirurgia , Mastectomia Segmentar/métodos , Fator de Crescimento Transformador beta1/análise , Idoso , Líquidos Corporais/química , Neoplasias da Mama/metabolismo , Neoplasias da Mama/radioterapia , Terapia Combinada , Feminino , Humanos , Ácido Hialurônico/análise , Cuidados Intraoperatórios/métodos , Pessoa de Meia-Idade , Radioterapia , Cicatrização/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA