Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38746175

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) poses significant challenges for effective treatment, with systemic chemotherapy often proving inadequate due to poor drug delivery and the tumor's immunosuppressive microenvironment. Engineered bacteria present a novel approach to target PDAC, leveraging their ability to colonize tumors and deliver therapeutic payloads. Here, we engineered probiotic Escherichia coli Nissle 1917 (EcN) to produce the pore-forming Theta toxin (Nis-Theta) and evaluated its efficacy in a preclinical model of PDAC. Probiotic administration resulted in selective colonization of tumor tissue, leading to improved overall survival compared to standard chemotherapy. Moreover, this strain exhibited cytotoxic effects on both primary and distant tumor lesions while sparing normal tissues. Importantly, treatment also modulated the tumor microenvironment by increasing anti-tumor immune cell populations and reducing immunosuppressive markers. These findings demonstrate the potential of engineered probiotic bacteria as a safe and effective therapeutic approach for PDAC, offering promise for improved patient outcomes.

2.
Nature ; 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38588697

RESUMEN

Broad-spectrum RAS inhibition holds the potential to benefit roughly a quarter of human cancer patients whose tumors are driven by RAS mutations1,2. RMC-7977 is a highly selective inhibitor of the active GTP-bound forms of KRAS, HRAS, and NRAS, with affinity for both mutant and wild type (WT) variants (RAS(ON) multi-selective)3. As >90% of human pancreatic ductal adenocarcinoma (PDAC) cases are driven by activating mutations in KRAS4, we assessed the therapeutic potential of the RAS(ON) multi-selective inhibitor RMC-7977 in a comprehensive range of PDAC models. We observed broad and pronounced anti-tumor activity across models following direct RAS inhibition at exposures that were well-tolerated in vivo. Pharmacological analyses revealed divergent responses to RMC-7977 in tumor versus normal tissues. Treated tumors exhibited waves of apoptosis along with sustained proliferative arrest whereas normal tissues underwent only transient decreases in proliferation, with no evidence of apoptosis. In the autochthonous KPC model, RMC-7977 treatment resulted in a profound extension of survival followed by on-treatment relapse. Analysis of relapsed tumors identified Myc copy number gain as a prevalent candidate resistance mechanism, which could be overcome by combinatorial TEAD inhibition in vitro. Together, these data establish a strong preclinical rationale for the use of broad-spectrum RAS-GTP inhibition in the setting of PDAC and identify a promising candidate combination therapeutic regimen to overcome monotherapy resistance.

3.
Cancer Discov ; 14(2): 348-361, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-37966260

RESUMEN

The sparse vascularity of pancreatic ductal adenocarcinoma (PDAC) presents a mystery: What prevents this aggressive malignancy from undergoing neoangiogenesis to counteract hypoxia and better support growth? An incidental finding from prior work on paracrine communication between malignant PDAC cells and fibroblasts revealed that inhibition of the Hedgehog (HH) pathway partially relieved angiosuppression, increasing tumor vascularity through unknown mechanisms. Initial efforts to study this phenotype were hindered by difficulties replicating the complex interactions of multiple cell types in vitro. Here we identify a cascade of paracrine signals between multiple cell types that act sequentially to suppress angiogenesis in PDAC. Malignant epithelial cells promote HH signaling in fibroblasts, leading to inhibition of noncanonical WNT signaling in fibroblasts and epithelial cells, thereby limiting VEGFR2-dependent activation of endothelial hypersprouting. This cascade was elucidated using human and murine PDAC explant models, which effectively retain the complex cellular interactions of native tumor tissues. SIGNIFICANCE: We present a key mechanism of tumor angiosuppression, a process that sculpts the physiologic, cellular, and metabolic environment of PDAC. We further present a computational and experimental framework for the dissection of complex signaling cascades that propagate among multiple cell types in the tissue environment. This article is featured in Selected Articles from This Issue, p. 201.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Humanos , Ratones , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular , Proteínas Hedgehog/genética , Neoplasias Pancreáticas/patología , Factor A de Crecimiento Endotelial Vascular
4.
Gastroenterology ; 166(2): 298-312.e14, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37913894

RESUMEN

BACKGROUND & AIMS: The highly heterogeneous cellular and molecular makeup of pancreatic ductal adenocarcinoma (PDAC) not only fosters exceptionally aggressive tumor biology, but contradicts the current concept of one-size-fits-all therapeutic strategies to combat PDAC. Therefore, we aimed to exploit the tumor biological implication and therapeutic vulnerabilities of a clinically relevant molecular PDAC subgroup characterized by SMAD4 deficiency and high expression of the nuclear factor of activated T cells (SMAD4-/-/NFATc1High). METHODS: Transcriptomic and clinical data were analyzed to determine the prognostic relevance of SMAD4-/-/NFATc1High cancers. In vitro and in vivo oncogenic transcription factor complex formation was studied by immunoprecipitation, proximity ligation assays, and validated cross model and species. The impact of SMAD4 status on therapeutically targeting canonical KRAS signaling was mechanistically deciphered and corroborated by genome-wide gene expression analysis and genetic perturbation experiments, respectively. Validation of a novel tailored therapeutic option was conducted in patient-derived organoids and cells and transgenic as well as orthotopic PDAC models. RESULTS: Our findings determined the tumor biology of an aggressive and chemotherapy-resistant SMAD4-/-/NFATc1High subgroup. Mechanistically, we identify SMAD4 deficiency as a molecular prerequisite for the formation of an oncogenic NFATc1/SMAD3/cJUN transcription factor complex, which drives the expression of RRM1/2. RRM1/2 replenishes nucleoside pools that directly compete with metabolized gemcitabine for DNA strand incorporation. Disassembly of the NFATc1/SMAD3/cJUN complex by mitogen-activated protein kinase signaling inhibition normalizes RRM1/2 expression and synergizes with gemcitabine treatment in vivo to reduce the proliferative index. CONCLUSIONS: Our results suggest that PDAC characterized by SMAD4 deficiency and oncogenic NFATc1/SMAD3/cJUN complex formation exposes sensitivity to a mitogen-activated protein kinase signaling inhibition and gemcitabine combination therapy.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Gemcitabina , Línea Celular Tumoral , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína smad3/metabolismo
5.
bioRxiv ; 2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-38105998

RESUMEN

Broad-spectrum RAS inhibition holds the potential to benefit roughly a quarter of human cancer patients whose tumors are driven by RAS mutations. However, the impact of inhibiting RAS functions in normal tissues is not known. RMC-7977 is a highly selective inhibitor of the active (GTP-bound) forms of KRAS, HRAS, and NRAS, with affinity for both mutant and wild type (WT) variants. As >90% of human pancreatic ductal adenocarcinoma (PDAC) cases are driven by activating mutations in KRAS, we assessed the therapeutic potential of RMC-7977 in a comprehensive range of PDAC models, including human and murine cell lines, human patient-derived organoids, human PDAC explants, subcutaneous and orthotopic cell-line or patient derived xenografts, syngeneic allografts, and genetically engineered mouse models. We observed broad and pronounced anti-tumor activity across these models following direct RAS inhibition at doses and concentrations that were well-tolerated in vivo. Pharmacological analyses revealed divergent responses to RMC-7977 in tumor versus normal tissues. Treated tumors exhibited waves of apoptosis along with sustained proliferative arrest whereas normal tissues underwent only transient decreases in proliferation, with no evidence of apoptosis. Together, these data establish a strong preclinical rationale for the use of broad-spectrum RAS inhibition in the setting of PDAC.

6.
STAR Protoc ; 3(4): 101899, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36595938

RESUMEN

Orthotopic patient-derived xenograft models recapitulate the genomic complexity of the original tumor and some aspects of local microenvironment, useful for rapid development and validation of personalized treatment strategies. Here, we precisely describe a protocol for generating tumor slices from human or murine-derived pancreatic cancer. They are then implanted directly into the murine pancreas, monitored using ultrasound, with a 90% success rate. This assay creates a clinically relevant in vivo model facilitating personalized treatment development.


Asunto(s)
Neoplasias Pancreáticas , Humanos , Animales , Ratones , Xenoinjertos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Páncreas/patología , Microambiente Tumoral , Neoplasias Pancreáticas
7.
Nat Commun ; 12(1): 4860, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34381026

RESUMEN

Cancer metabolism is rewired to support cell survival in response to intrinsic and environmental stressors. Identification of strategies to target these adaptions is an area of active research. We previously described a cytosolic aspartate aminotransaminase (GOT1)-driven pathway in pancreatic cancer used to maintain redox balance. Here, we sought to identify metabolic dependencies following GOT1 inhibition to exploit this feature of pancreatic cancer and to provide additional insight into regulation of redox metabolism. Using pharmacological methods, we identify cysteine, glutathione, and lipid antioxidant function as metabolic vulnerabilities following GOT1 withdrawal. We demonstrate that targeting any of these pathways triggers ferroptosis, an oxidative, iron-dependent form of cell death, in GOT1 knockdown cells. Mechanistically, we reveal that GOT1 inhibition represses mitochondrial metabolism and promotes a catabolic state. Consequently, we find that this enhances labile iron availability through autophagy, which potentiates the activity of ferroptotic stimuli. Overall, our study identifies a biochemical connection between GOT1, iron regulation, and ferroptosis.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/antagonistas & inhibidores , Ferroptosis , Neoplasias Pancreáticas/metabolismo , Animales , Antioxidantes/farmacología , Aspartato Aminotransferasa Citoplasmática/genética , Aspartato Aminotransferasa Citoplasmática/metabolismo , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Cistina/metabolismo , Ferroptosis/efectos de los fármacos , Glutatión/biosíntesis , Humanos , Hierro/metabolismo , Ratones , Mitocondrias/metabolismo , Neoplasias Pancreáticas/patología
8.
Theranostics ; 10(10): 4614-4626, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32292518

RESUMEN

Background and aims: Poor specificity and predictive values of current cross-sectional radiological imaging methods in evaluation of pancreatic adenocarcinoma (PDAC) limit the clinical capability to accurately stage the tumor pre-operatively and provide optimal surgical treatment and improve patient outcomes. Methods: In this study, we applied Harmonic Motion Elastography (HME), a quantitative ultrasound-based imaging method to calculate Young's modulus (YM) in PDAC mouse models (n = 30) and human pancreatic resection specimens of PDAC (n=32). We compared the YM to the collagen assessment by Picrosirius red (PSR) stain on corresponding histologic sections. Results: HME is capable of differentiating between different levels of fibrosis in transgenic mice. In mice without pancreatic fibrosis, the measured YM was 4.2 ± 1.3 kPa, in fibrotic murine pancreata, YM was 5.5 ± 2.0 kPa and in murine PDAC tumors, YM was 11.3 ± 1.7 kPa. The corresponding PSR values were 2.0 ± 0.8 %, 9.8 ± 3.4 %, and 13.2 ± 1.2%, respectively. In addition, three regions within each human surgical PDAC specimen were assessed: tumor, which had both the highest Young's modulus (YM > 40 kPa) and collagen density (PSR > 40 %); non-neoplastic adjacent pancreas, which had the lowest Young's modulus (YM < 15 kPa) and collagen density (PSR < 10%) and a transitional peri-lesional region between the tumor and non-neoplastic pancreas with an intermediate value of measured Young's modulus (15 kPa < YM < 40 kPa) and collagen density (15% < PSR < 35 %). Conclusion: In conclusion, a non-invasive, quantitative imaging tool for detecting, staging and delineating PDAC tumor margins based on the change in collagen density was developed.


Asunto(s)
Carcinoma Ductal Pancreático/diagnóstico por imagen , Módulo de Elasticidad , Diagnóstico por Imagen de Elasticidad/métodos , Páncreas , Neoplasias Pancreáticas/diagnóstico por imagen , Adulto , Anciano , Anciano de 80 o más Años , Animales , Progresión de la Enfermedad , Femenino , Fibrosis/diagnóstico por imagen , Humanos , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Páncreas/diagnóstico por imagen , Páncreas/patología
9.
Clin Cancer Res ; 26(6): 1297-1308, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31831559

RESUMEN

PURPOSE: Pancreatic ductal adenocarcinoma (PDA) is a common, deadly cancer that is challenging both to diagnose and to manage. Its hallmark is an expansive, desmoplastic stroma characterized by high mechanical stiffness. In this study, we sought to leverage this feature of PDA for two purposes: differential diagnosis and monitoring of response to treatment. EXPERIMENTAL DESIGN: Harmonic motion imaging (HMI) is a functional ultrasound technique that yields a quantitative relative measurement of stiffness suitable for comparisons between individuals and over time. We used HMI to quantify pancreatic stiffness in mouse models of pancreatitis and PDA as well as in a series of freshly resected human pancreatic cancer specimens. RESULTS: In mice, we learned that stiffness increased during progression from preneoplasia to adenocarcinoma and also effectively distinguished PDA from several forms of pancreatitis. In human specimens, the distinction of tumors versus adjacent pancreatitis or normal pancreas tissue was even more stark. Moreover, in both mice and humans, stiffness increased in proportion to tumor size, indicating that tuning of mechanical stiffness is an ongoing process during tumor progression. Finally, using a brca2-mutant mouse model of PDA that is sensitive to cisplatin, we found that tissue stiffness decreases when tumors respond successfully to chemotherapy. Consistent with this observation, we found that tumor tissues from patients who had undergone neoadjuvant therapy were less stiff than those of untreated patients. CONCLUSIONS: These findings support further development of HMI for clinical applications in disease staging and treatment response assessment in PDA.


Asunto(s)
Diagnóstico por Imagen de Elasticidad/métodos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Fantasmas de Imagen , Procesamiento de Señales Asistido por Computador/instrumentación , Ultrasonografía/métodos , Anciano , Anciano de 80 o más Años , Animales , Diagnóstico Diferencial , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Persona de Mediana Edad , Movimiento (Física) , Estadificación de Neoplasias , Neoplasias Pancreáticas/diagnóstico por imagen , Resultado del Tratamiento
10.
Clin Cancer Res ; 25(18): 5548-5560, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31175095

RESUMEN

PURPOSE: Pancreatic ductal adenocarcinoma (PDA) is a deadly cancer that is broadly chemoresistant, due in part to biophysical properties of tumor stroma, which serves as a barrier to drug delivery for most classical chemotherapeutic drugs. The goal of this work is to evaluate the preclinical efficacy and mechanisms of PTC596, a novel agent with potent anticancer properties in vitro and desirable pharmacologic properties in vivo.Experimental Design: We assessed the pharmacology, mechanism, and preclinical efficacy of PTC596 in combination with standards of care, using multiple preclinical models of PDA. RESULTS: We found that PTC596 has pharmacologic properties that overcome the barrier to drug delivery in PDA, including a long circulating half-life, lack of P-glycoprotein substrate activity, and high systemic tolerability. We also found that PTC596 combined synergistically with standard clinical regimens to improve efficacy in multiple model systems, including the chemoresistant genetically engineered "KPC" model of PDA. Through mechanistic studies, we learned that PTC596 functions as a direct microtubule polymerization inhibitor, yet a prior clinical trial found that it lacks peripheral neurotoxicity, in contrast to other such agents. Strikingly, we found that PTC596 synergized with the standard clinical backbone regimen gemcitabine/nab-paclitaxel, yielding potent, durable regressions in a PDX model. Moreover, similar efficacy was achieved in combination with nab-paclitaxel alone, highlighting a specific synergistic interaction between two different microtubule-targeted agents in the setting of pancreatic ductal adenocarcinoma. CONCLUSIONS: These data demonstrate clear rationale for the development of PTC596 in combination with standard-of-care chemotherapy for PDA.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/metabolismo , Microtúbulos/metabolismo , Neoplasias Pancreáticas/metabolismo , Multimerización de Proteína/efectos de los fármacos , Moduladores de Tubulina/farmacología , Albúminas/farmacología , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/mortalidad , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Inmunohistoquímica , Ratones , Microtúbulos/química , Paclitaxel/farmacología , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/mortalidad , Moduladores de Tubulina/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
11.
Phys Med Biol ; 61(15): 5741-54, 2016 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-27401609

RESUMEN

Recently, ultrasonic imaging of soft tissue mechanics has been increasingly studied to image otherwise undetectable pathologies. However, many underlying mechanisms of tissue stiffening remain unknown, requiring small animal studies and adapted elasticity mapping techniques. Harmonic motion imaging (HMI) assesses tissue viscoelasticity by inducing localized oscillation from a periodic acoustic radiation force. The objective of this study was to evaluate the feasibility of HMI for in vivo elasticity mapping of abdominal organs in small animals. Pathological cases, i.e. chronic pancreatitis and pancreatic cancer, were also studied in vivo to assess the capability of HMI for detection of the change in mechanical properties. A 4.5 MHz focused ultrasound transducer (FUS) generated an amplitude-modulated beam resulting in 50 Hz harmonic tissue oscillations at its focus. Axial tissue displacement was estimated using 1D-cross-correlation of RF signals acquired with a 7.8 MHz diagnostic transducer confocally aligned with the FUS. In vitro results in canine liver and kidney showed the correlation between HMI displacement and Young's moduli measured by rheometry compression testing. HMI was capable of providing reproducible elasticity maps of the mouse abdominal region in vivo allowing the identification of, from stiffest to softest, the murine kidney, pancreas, liver, and spleen. Finally, pancreata affected by pancreatitis and pancreatic cancer showed HMI displacements 1.7 and 2.2 times lower than in the control case, respectively, indicating higher stiffness. The HMI displacement amplitude was correlated with the extent of fibrosis as well as detecting the very onset of stiffening even before fibrosis could be detected on H&E. This work shows that HMI can produce reliable elasticity maps of mouse abdominal region in vivo, thus providing a potentially critical tool to assess pathologies affecting organ elasticity.


Asunto(s)
Abdomen/diagnóstico por imagen , Diagnóstico por Imagen de Elasticidad/métodos , Animales , Perros , Módulo de Elasticidad , Diagnóstico por Imagen de Elasticidad/instrumentación , Riñón/diagnóstico por imagen , Hígado/diagnóstico por imagen , Tracto Gastrointestinal Inferior/diagnóstico por imagen , Ratones , Movimiento (Física) , Transductores
12.
Clin Cancer Res ; 21(9): 2096-106, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25677697

RESUMEN

PURPOSE: T-cell lymphomas (TCL) are aggressive diseases, which carry a poor prognosis. The emergence of new drugs for TCL has created a need to survey these agents in a rapid and reproducible fashion, to prioritize combinations which should be prioritized for clinical study. Mouse models of TCL that can be used for screening novel agents and their combinations are lacking. Developments in noninvasive imaging modalities, such as surface bioluminescence (SBL) and three-dimensional ultrasound (3D-US), are challenging conventional approaches in xenograft modeling relying on caliper measurements. The recent approval of pralatrexate and romidepsin creates an obvious combination that could produce meaningful activity in TCL, which is yet to be studied in combination. EXPERIMENTAL DESIGN: High-throughput screening and multimodality imaging approach of SBL and 3D-US in a xenograft NOG mouse model of TCL were used to explore the in vitro and in vivo activity of pralatrexate and romidepsin in combination. Corresponding mass spectrometry-based pharmacokinetic and immunohistochemistry-based pharmacodynamic analyses of xenograft tumors were performed to better understand a mechanistic basis for the drug:drug interaction. RESULTS: In vitro, pralatrexate and romidepsin exhibited concentration-dependent synergism in combination against a panel of TCL cell lines. In a NOG murine model of TCL, the combination of pralatrexate and romidepsin exhibited enhanced efficacy compared with either drug alone across a spectrum of tumors using complementary imaging modalities, such as SBL and 3D-US. CONCLUSIONS: Collectively, these data strongly suggest that the combination of pralatrexate and romidepsin merits clinical study in patients with TCLs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Modelos Animales de Enfermedad , Linfoma de Células T/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Aminopterina/administración & dosificación , Aminopterina/análogos & derivados , Animales , Línea Celular Tumoral , Depsipéptidos/administración & dosificación , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Mediciones Luminiscentes , Ratones , Transfección
13.
Sci Transl Med ; 7(271): 271ra7, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25609167

RESUMEN

The inability to visualize the true extent of cancers represents a significant challenge in many areas of oncology. The margins of most cancer types are not well demarcated because the cancer diffusely infiltrates the surrounding tissues. Furthermore, cancers may be multifocal and characterized by the presence of microscopic satellite lesions. Such microscopic foci represent a major reason for persistence of cancer, local recurrences, and metastatic spread, and are usually impossible to visualize with currently available imaging technologies. An imaging method to reveal the true extent of tumors is desired clinically and surgically. We show the precise visualization of tumor margins, microscopic tumor invasion, and multifocal locoregional tumor spread using a new generation of surface-enhanced resonance Raman scattering (SERRS) nanoparticles, which are termed SERRS nanostars. The SERRS nanostars feature a star-shaped gold core, a Raman reporter resonant in the near-infrared spectrum, and a primer-free silication method. In genetically engineered mouse models of pancreatic cancer, breast cancer, prostate cancer, and sarcoma, and in one human sarcoma xenograft model, SERRS nanostars enabled accurate detection of macroscopic malignant lesions, as well as microscopic disease, without the need for a targeting moiety. Moreover, the sensitivity (1.5 fM limit of detection) of SERRS nanostars allowed imaging of premalignant lesions of pancreatic and prostatic neoplasias. High sensitivity and broad applicability, in conjunction with their inert gold-silica composition, render SERRS nanostars a promising imaging agent for more precise cancer imaging and resection.


Asunto(s)
Diagnóstico por Imagen/métodos , Nanopartículas , Neoplasias/diagnóstico , Espectrometría Raman/métodos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Metástasis de la Neoplasia , Pinocitosis , Lesiones Precancerosas/patología , Distribución Tisular
14.
Cancer Cell ; 25(6): 735-47, 2014 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-24856585

RESUMEN

Sonic hedgehog (Shh), a soluble ligand overexpressed by neoplastic cells in pancreatic ductal adenocarcinoma (PDAC), drives formation of a fibroblast-rich desmoplastic stroma. To better understand its role in malignant progression, we deleted Shh in a well-defined mouse model of PDAC. As predicted, Shh-deficient tumors had reduced stromal content. Surprisingly, such tumors were more aggressive and exhibited undifferentiated histology, increased vascularity, and heightened proliferation--features that were fully recapitulated in control mice treated with a Smoothened inhibitor. Furthermore, administration of VEGFR blocking antibody selectively improved survival of Shh-deficient tumors, indicating that Hedgehog-driven stroma suppresses tumor growth in part by restraining tumor angiogenesis. Together, these data demonstrate that some components of the tumor stroma can act to restrain tumor growth.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/patología , Células del Estroma/patología , Animales , Anticuerpos Monoclonales/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Proteínas Hedgehog/deficiencia , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Inmunoglobulina G/farmacología , Ratones , Ratones Transgénicos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Distribución Aleatoria , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Transducción de Señal , Células del Estroma/metabolismo
15.
Cold Spring Harb Protoc ; 2014(1): 47-56, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24371318

RESUMEN

The static analysis of tumor tissues at a single, terminal end point has been the mainstay of studies in mouse models of cancer. However, with the development of model systems that reproduce the intra- and intertumoral heterogeneity of human tumors, such studies are limited by the need for large numbers of animals to overcome increased intragroup variance. The shortcomings of a single-timepoint approach to molecular analysis are especially apparent in the context of therapeutic studies, in which the dynamic response to treatment is of particular consequence. To mitigate the effects of intertumoral heterogeneity, multiple tissue samples may be harvested from the same tumor at different time points through the use of surgical biopsies. For abdominal tumors, preprocedure imaging may be used to assess the suitability of tumors for biopsy. Sterile surgical techniques are used to access the abdominal cavity, and customized instruments facilitate the immobilization and retrieval of tissue samples from hard or fibrous tumors. Thermoregulation, hemostasis, and wound closure techniques are critical to successful surgical outcomes, whereas appropriate anesthetic, analgesic, and postoperative recovery regimens are important for maintenance of animal welfare. Using a mouse model of pancreatic ductal adenocarcinoma, we present a comprehensive protocol suitable for the routine acquisition of abdominal tumor biopsies.


Asunto(s)
Neoplasias Abdominales/patología , Biopsia/métodos , Laparotomía/métodos , Animales , Modelos Animales de Enfermedad , Humanos , Ratones
16.
Methods Mol Biol ; 980: 249-66, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23359158

RESUMEN

Ultrasonography is a powerful imaging modality that enables noninvasive, real-time visualization of abdominal organs and tissues. This technology may be adapted for use in mice through the utilization of higher frequency transducers, allowing for extremely high-resolution imaging of the mouse pancreas. This technique is particularly well suited to pancreas imaging due to the ultrasonographic properties of the normal mouse pancreas, easily accessible imaging planes for the head and tail of the mouse pancreas, and the comparative difficulty in imaging the mouse pancreas with other technologies. A suite of measurement tools is available to characterize the normal and diseased states of tissues. Of particular utility for cancer applications is the ability to use tomography to construct a 3D tumor volume, enabling longitudinal imaging studies to track tumor development, or response to therapies. Here, we describe a detailed method for performing high-resolution ultrasound to detect and measure pancreatic lesions in a genetically engineered mouse model of pancreatic ductal using the VisualSonics Vevo2100 High Resolution Ultrasound System. The method includes preparation of the animal for imaging, 2D and 3D image acquisition, and post-acquisition analysis of tumor volumes. The combined procedure has been utilized extensively by our group and others for the preclinical evaluation of novel therapeutic agents in the treatment of pancreatic ductal adenocarcinoma (Olive et al., Science 324:1457-1461, 2009; Cook et al., Methods Enzymol 439:73-85, 2008; Singh et al., Nat Biotechnol 28:585-593, 2010; Beatty et al., Science 331:1612-1616, 2011).


Asunto(s)
Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/patología , Animales , Modelos Animales de Enfermedad , Ratones , Carga Tumoral , Ultrasonografía
17.
Int J Stroke ; 8(3): 180-5, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22759525

RESUMEN

BACKGROUND: Hypothermia is a promising experimental treatment for acute ischemic stroke. Human trials are still at an early stage, with the focus now on using hypothermia in awake patients. Pethidine (meperidine) is the principle agent used to control shivering in humans; however, whether it has any modulating effects on the neuroprotective efficacy of hypothermia is unknown. AIM: The aim of this study was to determine if pethidine influences the neuroprotective effect of hypothermia in experimental stroke. METHODS: Seventy-two male spontaneously hypertensive rats were anesthetized with isoflurane and randomly assigned to either normothermia (37. 4 °C rectal temperature); hypothermia (33 °C maintained for 130 mins); normothermia plus pethidine (2.5 mg/kg); or hypothermia plus pethidine. Temporary (90 mins) endovascular occlusion of the middle cerebral artery was induced blinded to treatment allocation and was confirmed with laser Doppler flowmetry. Pethidine and cooling were started immediately after vessel occlusion. Animals in the normothermia group had active temperature management using a heat lamp and fan. Assessments of outcome were carried out 24 after the induction of injury. RESULTS: Thirteen animals met our prespecified criteria for exclusion, and data for 59 rats were presented here. Hypothermia was associated with a 63% reduction in infarct size, and pethidine had no significant impact on the efficacy of hypothermia. No effects were observed in neurobehavioral outcome or edema volume across experimental groups. CONCLUSIONS: The effects of hypothermia in a model of focal ischemia are not affected by administration of pethidine.


Asunto(s)
Infarto Encefálico/terapia , Hipotermia Inducida/métodos , Meperidina/farmacología , Fármacos Neuroprotectores/farmacología , Accidente Cerebrovascular/terapia , Animales , Conducta Animal/efectos de los fármacos , Infarto Encefálico/patología , Circulación Cerebrovascular/fisiología , Masculino , Distribución Aleatoria , Ratas , Ratas Endogámicas SHR , Accidente Cerebrovascular/patología
18.
J Endocrinol ; 206(1): 93-103, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20395380

RESUMEN

We used our genomic androgen receptor (AR) knockout (ARKO) mouse model, in which the AR is unable to bind DNA to: 1) document gender differences between males and females; 2) identify the genomic (DNA-binding-dependent) AR-mediated actions in males; 3) determine the contribution of genomic AR-mediated actions to these gender differences; and 4) identify physiological genomic AR-mediated actions in females. At 9 weeks of age, control males had higher body, heart and kidney mass, lower spleen mass, and longer and larger bones compared to control females. Compared to control males, ARKO males had lower body and kidney mass, higher splenic mass, and reductions in cortical and trabecular bone. Deletion of the AR in ARKO males abolished the gender differences in heart and cortical bone. Compared with control females, ARKO females had normal body weight, but 14% lower heart mass and heart weight/body weight ratio. Relative kidney mass was also reduced, and relative spleen mass was increased. ARKO females had a significant reduction in cortical bone growth and changes in trabecular architecture, although with no net change in trabecular bone volume. In conclusion, we have shown that androgens acting via the genomic AR signaling pathway mediate, at least in part, the gender differences in body mass, heart, kidney, spleen, and bone, and play a physiological role in the regulation of cardiac, kidney and splenic size, cortical bone growth, and trabecular bone architecture in females.


Asunto(s)
ADN/metabolismo , Receptores Androgénicos/fisiología , Caracteres Sexuales , Transducción de Señal/fisiología , Andrógenos/fisiología , Animales , Peso Corporal , Desarrollo Óseo , Huesos/anatomía & histología , Calcificación Fisiológica , Femenino , Corazón/anatomía & histología , Riñón/anatomía & histología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Receptores Androgénicos/deficiencia , Bazo/anatomía & histología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA