Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
3.
Biopreserv Biobank ; 18(3): 222-227, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32302515

RESUMEN

The availability of viable human tissues is critical to support translational research focused on personalized care. Most studies have relied on fresh frozen or formalin-fixed paraffin-embedded tissues for histopathology, genomics, and proteomics. Yet, basic, translational, and clinical research downstream assays such as tumor progression/invasion, patient-derived xenograft, organoids, immunoprofiling, and vaccine development still require viable tissue, which are time-sensitive and rare commodities. We describe the generation of two-dimensional (2D) and three-dimensional (3D) cultures to validate a viable freeze cryopreservation technique as a standard method of highest quality specimen preservation. After surgical resection, specimens were minced, placed in CryoStor™ media, and frozen using a slow freezing method (-1°C/min in -80°C) for 24 hours and then stored in liquid nitrogen. After 15-18 months, the tissues were thawed, dissociated into single-cell suspensions, and evaluated for cell viability. To generate primary 2D cultures, cells were plated onto Collagen-/Matrigel-coated plates. To develop 3D cultures (organoids), the cells were plated in reduced serum RPMI media on nonadherent plates or in Matrigel matrix. The epithelial nature of the cells was confirmed by using immunohistochemistry for cytokeratins. DNA and RNA isolation was performed using QIAGEN AllPrep kits. We developed primary lines (2D and 3D) of colon, thyroid, lung, renal, and liver cancers that were positive for cytokeratin staining. 3D lines were developed from the same cohort of tumor types in both suspended media and Matrigel matrix. Multiple freeze-thaw cycles did not significantly alter the viability and growth of 2D and 3D lines. DNA/RNA recovery was similar to its fresh frozen cohort. In this study, we validated 2D and 3D tissue cultures as methods to corroborate the feasibility of viable cryopreservation of tumor tissue. This proof-of-principle study, if more widely implemented, should improve accessibility of human viable tumor tissue/cells in a time-independent manner for many basic, preclinical, and translational assays.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Criopreservación/métodos , Conservación de Tejido/métodos , Biomarcadores/metabolismo , Línea Celular/citología , Línea Celular/metabolismo , Supervivencia Celular , Marcadores Genéticos , Humanos , Organoides/citología , Organoides/metabolismo , Prueba de Estudio Conceptual , Análisis de la Célula Individual , Células Tumorales Cultivadas/citología
4.
Drug Deliv Transl Res ; 9(6): 1095-1105, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31228097

RESUMEN

Poly(lactic-co-glycolic) acid (PLGA) has been successfully used in drug delivery and biomaterial applications, but very little attention has been directed towards the potential in vivo effects of peptide-loaded PLGA nanoparticles (NPs), specifically the potency of intravenous (IV) STEAP peptide-loaded PLGA-NP (nanovaccine) dosing and whether STEAP-specific CD8+ T cells directly play a key role in tumor inhibition. To address these concerns, syngeneic prostate cancer mouse models were established and treated with either mSTEAP peptide emulsified in incomplete Freund's adjuvant (IFA) via subcutaneous (SC) injection or mSTEAP peptide nanovaccine containing the same amount of peptide via IV or SC injection. Meanwhile, mice were treated with either CD8b mAb followed by nanovaccine treatment, free mSTEAP peptide, or empty PLGA-NPs. Immune responses in these mice were examined using cytotoxicity assays at 14 days after treatment. Tumor size and survival in various treatment groups were measured and monitored. The results demonstrated that mSTEAP peptide nanovaccine resulted in tumor inhibition by eliciting a significantly stronger CD8+ T cell immune response when compared with the controls. Moreover, the survival periods of mice treated with mSTEAP nanovaccine were significantly longer than those of mice treated with mSTEAP peptide emulsified in IFA or the treatment controls. Additionally, it was observed that the peptide nanovaccine was mainly distributed in the mouse liver and lungs after IV injection. These findings suggest that the peptide nanovaccine is a promising immunotherapeutic approach and offers a new opportunity for prostate cancer therapies.


Asunto(s)
Antígenos de Neoplasias/administración & dosificación , Linfocitos T CD8-positivos/efectos de los fármacos , Vacunas contra el Cáncer/administración & dosificación , Nanopartículas/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antígenos de Neoplasias/farmacología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/farmacocinética , Línea Celular Tumoral , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Ratones Endogámicos C57BL , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacocinética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo
5.
Biopreserv Biobank ; 17(2): 139-142, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30912671

RESUMEN

Adequate preservation of biospecimens has been proven to be critical to obtain reliable and reproducible results in genomics, transcriptomics, proteomics, and many other assays. Most biological assays can be performed on specimens preserved in -80°C ultra-low freezers, but their quality can be influenced by temperature variability within storage units. Thus, regulatory standards such as those from the College of American Pathologists (CAP), the federal Clinical Laboratory Improvement Amendments, and standards from the Food and Drug Administration require temperature mapping, a standard quality assessment for accreditation when using ultra-low freezers for long-term biospecimen storage. The current mapping methods, providing annual/periodic data, may not be adequate indicators of temperature stability within the different zones of the freezers. In addition, they frequently require manual handling of biospecimens periodically, as they require freezers to be emptied or rearranged temporarily for the installation of temperature probes, risking the integrity of biospecimen quality. In this article, we describe a novel monitoring methodology based on real-time temperature reading of multiple zones by permanently installing thermocouples. An online cloud-based application records temperature variations within 1 minute intervals, and its 24/7 alert system triggers text alarm messages to a predefined set of users when temperature values increase above preset defaults. This provides an opportunity to take remedial action and to obtain a better-quality assessment. Our results indicate that real-time temperature monitoring at multiple zones of a freezer with a 1 minute resolution is a stable and sustainable methodology and, most importantly, lowers the risk of compromising the quality of the biospecimen. The design and use of the real-time monitoring system for ultra-low freezers is one of the acceptable methods by CAP to ensure the stability of biospecimen quality during long-term storage.


Asunto(s)
Criopreservación/instrumentación , Criopreservación/normas , Control de Calidad , Manejo de Especímenes/instrumentación , Manejo de Especímenes/normas , Termografía , Criopreservación/métodos , Manejo de Especímenes/métodos
6.
Nature ; 543(7643): 122-125, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28178237

RESUMEN

Human cells have twenty-three pairs of chromosomes. In cancer, however, genes can be amplified in chromosomes or in circular extrachromosomal DNA (ecDNA), although the frequency and functional importance of ecDNA are not understood. We performed whole-genome sequencing, structural modelling and cytogenetic analyses of 17 different cancer types, including analysis of the structure and function of chromosomes during metaphase of 2,572 dividing cells, and developed a software package called ECdetect to conduct unbiased, integrated ecDNA detection and analysis. Here we show that ecDNA was found in nearly half of human cancers; its frequency varied by tumour type, but it was almost never found in normal cells. Driver oncogenes were amplified most commonly in ecDNA, thereby increasing transcript level. Mathematical modelling predicted that ecDNA amplification would increase oncogene copy number and intratumoural heterogeneity more effectively than chromosomal amplification. We validated these predictions by quantitative analyses of cancer samples. The results presented here suggest that ecDNA contributes to accelerated evolution in cancer.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Evolución Molecular , Amplificación de Genes/genética , Heterogeneidad Genética , Modelos Genéticos , Neoplasias/genética , Oncogenes/genética , Cromosomas Humanos/genética , Análisis Citogenético , Análisis Mutacional de ADN , Genoma Humano/genética , Humanos , Metafase/genética , Neoplasias/clasificación , ARN Mensajero/análisis , ARN Neoplásico/genética , Reproducibilidad de los Resultados , Programas Informáticos
7.
Ann Surg Oncol ; 22(6): 2082-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25319581

RESUMEN

BACKGROUND: The aim of this study was to evaluate the efficacy of using matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9)-cleavable ratiometric activatable cell-penetrating peptides (RACPPs) conjugated to Cy5 and Cy7 fluorophores to accurately label pancreatic cancer for fluorescence-guided surgery (FGS) in an orthotopic mouse model. METHODS: Orthotopic mouse models were established using MiaPaCa-2-GFP human pancreatic cancer cells. Two weeks after implantation, tumor-bearing mice were randomized to conventional white light reflectance (WLR) surgery or FGS. FGS was performed at far-red and infrared wavelengths with a customized fluorescence-dissecting microscope 2 h after injection of MMP-2 and MMP-9-cleavable RACPPs. Green fluorescence imaging of the GFP-labeled cancer cells was used to assess the effectiveness of surgical resection and monitor recurrence. At 8 weeks, mice were sacrificed to evaluate tumor burden and metastases. RESULTS: Mice in the WLR group had larger primary tumors than mice in the FGS group at termination [1.72 g ± standard error (SE) 0.58 vs. 0.25 g ± SE 0.14; respectively, p = 0.026). Mean disease-free survival was significantly lengthened from 5.33 weeks in the WLR group to 7.38 weeks in the FGS group (p = 0.02). Recurrence rates were lower in the FGS group than in the WLR group (38 vs. 73 %; p = 0.049). This translated into lower local and distant recurrence rates for FGS compared to WLR (31 vs. 67 for local recurrence, respectively, and 25 vs. 60 % for distant recurrence, respectively). Metastatic tumor burden was significantly greater in the WLR group than in the FGS group (96.92 mm(2) ± SE 52.03 vs. 2.20 mm(2) ± SE 1.43; respectively, χ (2) = 5.455; p = 0.02). CONCLUSIONS: RACPPs can accurately and effectively label pancreatic cancer for effective FGS, resulting in better postresection outcomes than for WLR surgery.


Asunto(s)
Péptidos de Penetración Celular/administración & dosificación , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Recurrencia Local de Neoplasia/prevención & control , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Cirugía Asistida por Computador , Animales , Péptidos de Penetración Celular/metabolismo , Femenino , Fluorescencia , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/química , Metaloproteinasa 9 de la Matriz/química , Ratones , Ratones Desnudos , Microscopía Fluorescente , Metástasis de la Neoplasia , Imagen Óptica/métodos , Carga Tumoral
8.
Cancer Res ; 74(22): 6671-81, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25261239

RESUMEN

Deregulation of protein synthesis is a hallmark of cancer cell proliferation, survival, and metastatic progression. eIF5A1 and its highly related isoform eIF5A2 are translation initiation factors that have been implicated in a range of human malignancies, but how they control cancer development and disease progression is still poorly understood. Here, we investigated how eIF5A proteins regulate pancreatic ductal adenocarcinoma (PDAC) pathogenesis. eIF5A proteins are the only known proteins regulated by a distinct posttranslational modification termed hypusination, which is catalyzed by two enzymes, deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase (DOHH). The highly selective nature of the hypusine modification and its amenability to pharmacologic inhibition make eIF5A proteins attractive therapeutic targets. We found that the expression and hypusination of eIF5A proteins are upregulated in human PDAC tissues and in premalignant pancreatic intraepithelial neoplasia tissues isolated from Pdx-1-Cre: LSL-KRAS(G12D) mice. Knockdown of eIF5A proteins in PDAC cells inhibited their growth in vitro and orthotopic tumor growth in vivo, whereas amplification of eIF5A proteins increased PDAC cell growth and tumor formation in mice. Small-molecule inhibitors of DHPS and DOHH both suppressed eIF5A hypusination, preventing PDAC cell growth. Interestingly, we found that eIF5A proteins regulate PDAC cell growth by modulating the expression of PEAK1, a nonreceptor tyrosine kinase essential for PDAC cell growth and therapy resistance. Our findings suggest that eIF5A proteins utilize PEAK1 as a downstream effector to drive PDAC pathogenesis and that pharmacologic inhibition of the eIF5A-hypusine-PEAK1 axis may provide a novel therapeutic strategy to combat this deadly disease.


Asunto(s)
Carcinoma Ductal Pancreático/etiología , Lisina/análogos & derivados , Neoplasias Pancreáticas/etiología , Factores de Iniciación de Péptidos/fisiología , Proteínas Tirosina Quinasas/fisiología , Proteínas de Unión al ARN/fisiología , Animales , Carcinoma Ductal Pancreático/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Ciclopirox , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Femenino , Humanos , Lisina/fisiología , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas p21(ras) , Piridonas/farmacología , Proteínas ras/fisiología , Gemcitabina , Factor 5A Eucariótico de Iniciación de Traducción
9.
J Am Coll Surg ; 219(1): 132-41, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24768506

RESUMEN

BACKGROUND: Our laboratory has previously developed fluorescence-guided surgery of pancreatic and other cancers in orthotopic mouse models. Laparoscopic surgery is being used more extensively in surgical oncology. This report describes the efficacy of laparoscopic fluorescence-guided surgery of pancreatic cancer in an orthotopic mouse model. STUDY DESIGN: Mouse models of human pancreatic cancer were established with fragments of the BxPC-3 red fluorescent protein-expressing human pancreatic cancer using surgical orthotopic implantation. Mice were randomized to bright-light laparoscopic surgery (BLLS) or to fluorescence-guided laparoscopic surgery (FGLS). Fluorescence-guided laparoscopic surgery was performed with a light-emitting diode light source through a 495-nm emission filter in order to resect the primary tumors and any additional separate submillimeter tumor deposits within the pancreas, the latter of which was not possible with BLLS. Tumors were labeled with anti-CEA AlexaFluor 488 antibodies 24 hours before surgery with intravenous injection. Perioperative fluorescence images were obtained to evaluate tumor size. Mice were followed postoperatively to assess for recurrence and at termination to evaluate tumor burden. RESULTS: At termination, the FGLS-treated mice had less pancreatic tumor volume than the BLLS-treated mice (5.75 mm(2) vs 28.43 mm(2), respectively; p = 0.012) and lower tumor weight (21.1 mg vs 174.4 mg, respectively; p = 0.033). Fluorescence-guided laparoscopic surgery compared with BLLS also decreased local recurrence (50% vs 80%, respectively; p = 0.048) and distant recurrence (70% vs 95%, respectively; p = 0.046). More mice in the FGLS group than the BLLS group were free of tumor at termination (25% vs 5%, respectively). Median disease-free survival was lengthened from 2 weeks with BLLS (95% CI, 1.635-2.365) to 7 weeks with FGLS (95% CI, 5.955-8.045; p = 0.001). CONCLUSIONS: Fluorescence-guided laparoscopic surgery is more effective than BLLS and, therefore, has important potential for surgical oncology.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antineoplásicos , Antígeno Carcinoembrionario/inmunología , Laparoscopía/métodos , Imagen Óptica/métodos , Pancreatectomía/métodos , Neoplasias Pancreáticas/cirugía , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Supervivencia sin Enfermedad , Femenino , Colorantes Fluorescentes , Fluorobencenos , Humanos , Modelos Logísticos , Ratones , Ratones Desnudos , Recurrencia Local de Neoplasia , Trasplante de Neoplasias , Neoplasias Pancreáticas/inmunología , Distribución Aleatoria , Resultado del Tratamiento , Carga Tumoral
10.
Ann Surg Oncol ; 21(4): 1405-11, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24499827

RESUMEN

BACKGROUND: We have developed a method of distinguishing normal tissue from pancreatic cancer in vivo using fluorophore-conjugated antibody to carcinoembryonic antigen (CEA). The objective of this study was to evaluate whether fluorescence-guided surgery (FGS) with a fluorophore-conjugated antibody to CEA, to highlight the tumor, can improve surgical resection and increase disease-free survival (DFS) and overall survival (OS) in orthotopic mouse models of human pancreatic cancer. METHODS: We established nude-mouse models of human pancreatic cancer with surgical orthotopic implantation of the human BxPC-3 pancreatic cancer. Orthotopic tumors were allowed to develop for 2 weeks. Mice then underwent bright-light surgery (BLS) or FGS 24 h after intravenous injection of anti-CEA-Alexa Fluor 488. Completeness of resection was assessed from postoperative imaging. Mice were followed postoperatively until premorbid to determine DFS and OS. RESULTS: Complete resection was achieved in 92 % of mice in the FGS group compared to 45.5 % in the BLS group (p = 0.001). FGS resulted in a smaller postoperative tumor burden (p = 0.01). Cure rates with FGS compared to BLS improved from 4.5 to 40 %, respectively (p = 0.01), and 1-year postoperative survival rates increased from 0 % with BLS to 28 % with FGS (p = 0.01). Median DFS increased from 5 weeks with BLS to 11 weeks with FGS (p = 0.0003). Median OS increased from 13.5 weeks with BLS to 22 weeks with FGS (p = 0.001). CONCLUSIONS: FGS resulted in greater cure rates and longer DFS and OS using a fluorophore-conjugated anti-CEA antibody. FGS has potential to improve the surgical treatment of pancreatic cancer.


Asunto(s)
Anticuerpos Monoclonales , Antígeno Carcinoembrionario/inmunología , Diagnóstico por Imagen , Técnica del Anticuerpo Fluorescente/métodos , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/cirugía , Animales , Femenino , Colorantes Fluorescentes , Humanos , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/diagnóstico , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Surg Oncol ; 109(5): 451-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24249594

RESUMEN

BACKGROUND AND OBJECTIVES: The aim of this study was to evaluate a new fluorescently labeled chimeric anti-CEA antibody for improved detection and resection of colon cancer. METHODS: Frozen tumor and normal human tissue samples were stained with chimeric and mouse antibody-fluorophore conjugates for comparison. Mice with patient-derived orthotopic xenografts (PDOX) of colon cancer underwent fluorescence-guided surgery (FGS) or bright-light surgery (BLS) 24 hr after tail vein injection of fluorophore-conjugated chimeric anti-CEA antibody. Resection completeness was assessed using postoperative images. Mice were followed for 6 months for recurrence. RESULTS: The fluorophore conjugation efficiency (dye/mole ratio) improved from 3-4 to >5.5 with the chimeric CEA antibody compared to mouse anti-CEA antibody. CEA-expressing tumors labeled with chimeric CEA antibody provided a brighter fluorescence signal on frozen human tumor tissues (P = 0.046) and demonstrated consistently lower fluorescence signals in normal human tissues compared to mouse antibody. Chimeric CEA antibody accurately labeled PDOX colon cancer in nude mice, enabling improved detection of tumor margins for more effective FGS. The R0 resection rate increased from 86% to 96% with FGS compared to BLS. CONCLUSION: Improved conjugating efficiency and labeling with chimeric fluorophore-conjugated antibody resulted in better detection and resection of human colon cancer in an orthotopic mouse model.


Asunto(s)
Anticuerpos Monoclonales , Antígeno Carcinoembrionario/inmunología , Quimera , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/cirugía , Diagnóstico por Imagen/métodos , Técnica del Anticuerpo Fluorescente/métodos , Aumento de la Imagen/métodos , Animales , Carbocianinas , Colorantes Fluorescentes , Xenoinjertos , Humanos , Ratones , Ratones Desnudos
12.
J Biomed Opt ; 18(12): 126016, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24356647

RESUMEN

The aim of this study was to evaluate a set of visible and near-infrared dyes conjugated to a tumor-specific chimeric antibody for high-resolution tumor imaging in orthotopic models of pancreatic cancer. BxPC-3 human pancreatic cancer was orthotopically implanted into pancreata of nude mice. Mice received a single intravenous injection of a chimeric anti-carcinoembryonic antigen antibody conjugated to one of the following fluorophores: 488-nm group (Alexa Fluor 488 or DyLight 488); 550-nm group (Alexa Fluor 555 or DyLight 550); 650-nm group (Alexa Fluor 660 or DyLight 650), or the 750-nm group (Alexa Fluor 750 or DyLight 755). After 24 h, the Olympus OV100 small-animal imaging system was used for noninvasive and intravital fluorescence imaging of mice. Dyes were compared with respect to depth of imaging, resolution, tumor-to-background ratio (TBR), photobleaching, and hemoglobin quenching. The longer wavelength dyes had increased depth of penetration and ability to detect the smallest tumor deposits and provided the highest TBRs, resistance to hemoglobin quenching, and specificity. The shorter wavelength dyes were more photostable. This study showed unique advantages of each dye for specific cancer imaging in a clinically relevant orthotopic model.


Asunto(s)
Anticuerpos , Antígeno Carcinoembrionario/inmunología , Antígeno Carcinoembrionario/metabolismo , Colorantes Fluorescentes , Imagen Óptica/métodos , Neoplasias Pancreáticas/patología , Animales , Anticuerpos/química , Anticuerpos/metabolismo , Línea Celular Tumoral , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Hemoglobinas , Histocitoquímica , Humanos , Ratones , Ratones Desnudos , Microscopía Fluorescente , Fotoblanqueo
13.
Ann Surg Oncol ; 20 Suppl 3: S693-700, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23943029

RESUMEN

BACKGROUND: Gastrointestinal stromal tumors (GISTs) are frequently characterized by KIT overexpression. Tumor-free margins and complete cytoreduction of disease are mainstays of treatment. We hypothesized that fluorescently labeled anti-KIT antibodies can label GIST in vivo. METHODS: KIT K641E(+/-) transgenic mice that spontaneously develop cecal GISTs were used in this study, with C57BL/6 mice serving as controls. Alexa 488 fluorophore-conjugated anti-KIT antibodies were delivered via the tail vein 24 h prior to fluorescence imaging. Following fluorescence laparoscopy, mice were sacrificed. The gastrointestinal tracts were grossly examined for tumors followed by fluorescence imaging. Tumors were harvested for histologic confirmation. RESULTS: KIT K641E(+/-) mice and C57BL/6 control mice received anti-KIT antibody or isotope control antibody. Fluorescence laparoscopy had a high tumor signal-to-background noise ratio. Upon blinded review of intravital fluorescence and bright light images, there were 2 false-positive and 0 false-negative results. The accuracy was 92 %. The sensitivity, specificity, positive and negative predictive values were 100, 87, 85, and 100 %, respectively, for the combined modalities. CONCLUSIONS: In this study, we present a method for in vivo fluorescence labeling of GIST in a murine model. Several translatable applications include: laparoscopic staging; visualization of peritoneal metastases; assessment of margin status; endoscopic differentiation of GISTs from other benign submucosal tumors; and longitudinal surveillance of disease response. This novel approach has clear clinical applications that warrant further research and development.


Asunto(s)
Anticuerpos Monoclonales , Modelos Animales de Enfermedad , Colorantes Fluorescentes , Tumores del Estroma Gastrointestinal/diagnóstico , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/inmunología , Animales , Femenino , Fluorescencia , Tumores del Estroma Gastrointestinal/inmunología , Humanos , Técnicas para Inmunoenzimas , Laparoscopía , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética
14.
Pancreas ; 42(6): 952-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23648841

RESUMEN

OBJECTIVES: To develop a mouse model for multispectral fluorescence imaging of the pancreas and pancreatic microenvironment. METHODS: Cre/loxP technology was used to develop this model. We crossed mT/mG indicator mice, engineered to constitutively express a conditional tdTomato transgene that converts to green fluorescent protein (GFP) expression after exposure to Cre recombinase, with Pdx1-Cre transgenic mice. To characterize this model for studies of pancreas biology, we performed bright light and fluorescence imaging of body cavities and intact organs and confocal microscopy of pancreata from offspring of Pdx1-Cre and mT/mG crosses. RESULTS: Pdx1-Cre-mT/mG mice demonstrated bright GFP expression within the pancreas and duodenum and intense tdTomato expression in all other organs. Green fluorescent protein expression was mosaic in Pdx1-Cre-mT/mG pancreata, with most showing extensive conversion from tdTomato to GFP expression within the epithelial-derived elements of the pancreatic parenchyma. Because both GFP and tdTomato are membrane targeted, individual cell borders were clearly outlined in confocal images of mT/mG pancreata. CONCLUSIONS: This mouse model enables multispectral fluorescence imaging of individual cells and cell processes at the microscopic level of the pancreatic microenvironment; it should prove valuable for a variety of fluorescence imaging studies, ranging from pancreatic development to pancreatic cancer biology.


Asunto(s)
Proteínas Luminiscentes/metabolismo , Neoplasias Pancreáticas/metabolismo , Microambiente Tumoral , Animales , Diagnóstico por Imagen/métodos , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Integrasas/genética , Integrasas/metabolismo , Proteínas Luminiscentes/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Factores de Tiempo , Transactivadores/genética , Transactivadores/metabolismo , Proteína Fluorescente Roja
15.
J Surg Res ; 184(1): 290-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23590868

RESUMEN

INTRODUCTION: Kras mutations have been thought to play an important role in pancreatic cancer progression. In this study, we evaluated how serially passaging primary pancreatic tumors with and without Kras mutations, in nude mice, can generate more aggressive variants of human pancreatic cancer. MATERIALS & METHODS: Orthotopic mouse models of human pancreatic cancer were established by injecting 1 × 10(6) cells of the Kras wildtype BxPC-3 cell line, expressing red fluorescent protein or the Kras mutant Panc-1 cell line expressing green fluorescent protein, into the pancreas. Pancreatic tumors were harvested from premorbid mice to establish cell lines. One million passaged cells were then orthotopically injected into another set of mice. Serial passaging continued until decreasing lifespan of the implanted mice stabilized, which occurred by six passages. Mice harboring serially-passaged cell lines were followed with weekly imaging. RESULTS: Serially passaging generated more aggressive variants of both human pancreatic cancer cell lines, one of which was Kras wild-type (BXPC-3) and the other Kras mutant, Panc-1, which displayed faster tumor growth and shortened survival time. Overall survival decreased from 18 wk in mice with the parental cell line (passage 0) tumor to ∼6 wk in mice by passage 6. Average time to metastasis was shortened from 14 wk to ∼3 wk or less. At termination, mice with the passaged tumor demonstrated a greater extent of distant metastasis. CONCLUSIONS: Serial passaging of tumor creates more aggressive variants of human pancreatic cancer cell lines regardless of Kras mutation. The aggressive variants can be used to study the molecular basis of highly malignant pancreatic cancer and to screen for effective agents against this disease.


Asunto(s)
Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Ratones Desnudos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/secundario , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Animales , Línea Celular Tumoral , Femenino , Proteínas Fluorescentes Verdes/genética , Humanos , Estimación de Kaplan-Meier , Proteínas Luminiscentes/genética , Ratones , Trasplante de Neoplasias , Neoplasias Pancreáticas/mortalidad , Proteínas Proto-Oncogénicas p21(ras) , Proteína Fluorescente Roja
16.
J Surg Res ; 179(1): 87-93, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23079571

RESUMEN

BACKGROUND: We inquired if fluorescence-guided surgery (FGS) could improve surgical outcomes in fluorescent orthotopic nude mouse models of human colon cancer. METHODS: We established fluorescent orthotopic mouse models of human colon cancer expressing a fluorescent protein. Tumors were resected under bright light surgery (BLS) or FGS. Pre- and post-operative images with the OV-100 Small Animal Imaging System (Olympus Corp, Tokyo Japan) were obtained to assess the extent of surgical resection. RESULTS: All mice with primary tumor that had undergone FGS had complete resection compared with 58% of mice in the BLS group (P = 0.001). FGS resulted in decreased recurrence compared with BLS (33% versus 62%, P = 0.049) and lengthened disease-free median survival from 9 to >36 wk. The median overall survival increased from 16 wk in the BLS group to 31 weeks in the FGS group. FGS resulted in a cure in 67% of mice (alive without evidence of tumor at >6 mo after surgery) compared with only 37% of mice that underwent BLS (P = 0.049). CONCLUSIONS: Surgical outcomes in orthotopic nude mouse models of human colon cancer were significantly improved with FGS. The present study can be translated to the clinic by various effective methods of fluorescently labeling tumors.


Asunto(s)
Neoplasias Colorrectales/cirugía , Procedimientos Quirúrgicos del Sistema Digestivo/métodos , Modelos Animales de Enfermedad , Imagen Óptica/métodos , Trasplante Heterólogo , Animales , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Supervivencia sin Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Desnudos , Prevención Secundaria , Resultado del Tratamiento
17.
Arch Surg ; 147(9): 876-80, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22987187

RESUMEN

OBJECTIVE: To improve detection of colon cancer metastases using fluorescence laparoscopy (FL). DESIGN: An orthotopic mouse model of human colon cancer was established by intracecal injection of HCT-116 human colon cancer cells expressing green fluorescent protein into 12 mice. One group modeled early disease and the second modeled late metastatic disease. For the early-disease model, 2 weeks after implantation, 6 mice underwent 2 modalities of laparoscopy: bright field laparoscopy (BL) and FL. The number of metastases identified within each of the 4 abdominal quadrants was recorded with both laparoscopy modalities. This process was repeated in the late-metastatic disease group 4 weeks after implantation. All animals were then humanely sacrificed and imaged using open fluorescence laparoscopy (OL) as a positive control to identify metastases. SETTING: Basic science laboratory. PARTICIPANTS: Twelve female, 6-week-old nude mice. INTERVENTIONS: Detection of tumor foci by FL compared with BL. MAIN OUTCOME MEASURES: Number of tumors identified in each quadrant. RESULTS Fluorescence laparoscopy enabled superior visualization of colon cancer metastases compared with BL in the early (P = .03) and late (P = .002) models of colon cancer. Compared with OL, BL was significantly inferior in the early (P = .04) and late (P < .001) groups. Fluorescence laparoscopy was not significantly different from OL in the early (P = .85) or late (P = .46) group. Thus, FL allowed identification of micrometastases that could not be distinguished from surrounding tissue using BL. CONCLUSIONS: The use of FL enables identification of metastases that could not be visualized using standard laparoscopy. This report illustrates the important clinical potential for FL in the surgical treatment of cancer.


Asunto(s)
Neoplasias del Colon/patología , Laparoscopía/métodos , Animales , Modelos Animales de Enfermedad , Femenino , Fluorescencia , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias
18.
Cancer Res ; 72(10): 2554-64, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22589274

RESUMEN

Early biomarkers and effective therapeutic strategies are desperately needed to treat pancreatic ductal adenocarcinoma (PDAC), which has a dismal 5-year patient survival rate. Here, we report that the novel tyrosine kinase PEAK1 is upregulated in human malignancies, including human PDACs and pancreatic intraepithelial neoplasia (PanIN). Oncogenic KRas induced a PEAK1-dependent kinase amplification loop between Src, PEAK1, and ErbB2 to drive PDAC tumor growth and metastasis in vivo. Surprisingly, blockade of ErbB2 expression increased Src-dependent PEAK1 expression, PEAK1-dependent Src activation, and tumor growth in vivo, suggesting a mechanism for the observed resistance of patients with PDACs to therapeutic intervention. Importantly, PEAK1 inactivation sensitized PDAC cells to trastuzumab and gemcitabine therapy. Our findings, therefore, suggest that PEAK1 is a novel biomarker, critical signaling hub, and new therapeutic target in PDACs.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Genes erbB-2 , Proteína Oncogénica pp60(v-src)/genética , Neoplasias Pancreáticas/genética , Proteínas Tirosina Quinasas/genética , Animales , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Resistencia a Antineoplásicos , Genes ras , Humanos , Ratones , Ratones Transgénicos , Modelos Moleculares , Metástasis de la Neoplasia/genética , Trasplante de Neoplasias , Transducción de Señal/genética , Activación Transcripcional , Regulación hacia Arriba
19.
J Am Coll Surg ; 215(1): 126-35; discussion 135-6, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22632917

RESUMEN

BACKGROUND: Negative surgical margins are vital to achieve cure and prolong survival in patients with pancreatic cancer. We inquired if fluorescence-guided surgery (FGS) could improve surgical outcomes and reduce recurrence rates in orthotopic mouse models of human pancreatic cancer. STUDY DESIGN: A randomized active-control preclinical trial comparing bright light surgery (BLS) to FGS was used. Orthotopic mouse models of human pancreatic cancer were established using the BxPC-3 pancreatic cancer cell line expressing red fluorescent protein (RFP). Two weeks after orthotopic implantation, tumors were resected with BLS or FGS. Pre- and postoperative images were obtained with the OV-100 Small Animal Imaging System to assess completeness of surgical resection in real time. Postoperatively, noninvasive whole body imaging was done to assess recurrence and follow tumor progression. Six weeks postoperatively, mice were sacrificed to evaluate primary pancreatic and metastatic tumor burden at autopsy. RESULTS: A more complete resection of pancreatic cancer was achieved using FGS compared with BLS: 98.9% vs 77.1%, p = 0.005. The majority of mice undergoing BLS (63.2%) had evidence of gross disease with no complete resections; 20% of mice undergoing FGS had complete resection and an additional 75% had only minimal residual disease (p = 0.0001). The mean postoperative tumor burden was significantly less with FGS compared with BLS: 0.08 ± 0.06 mm(2) vs 2.64 ± 0.63 mm(2), p = 0.001. The primary tumor burden at termination was significantly less with FGS compared with BLS: 19.3 ± 5.3 mm(2) vs 6.2 ± 3.6 mm(2), p = 0.048. FGS resulted in significantly longer disease-free survival than BLS (p = 0.02, hazard ratio = 0.39, 95% CI 0.17, 0.88). CONCLUSIONS: Surgical outcomes were improved in pancreatic cancer using fluorescence-guidance. This novel approach has significant potential to improve surgical treatment of cancer.


Asunto(s)
Proteínas Luminiscentes/análisis , Pancreatectomía/métodos , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/cirugía , Animales , Modelos Animales de Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Células Tumorales Cultivadas , Proteína Fluorescente Roja
20.
J Am Coll Surg ; 214(6): 997-1007.e2, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22542065

RESUMEN

BACKGROUND: The aim of this study was to improve fluorescence laparoscopy of pancreatic cancer in an orthotopic mouse model with the use of a light-emitting diode (LED) light source and optimal fluorophore combinations. STUDY DESIGN: Human pancreatic cancer models were established with fluorescent FG-RFP, MiaPaca2-GFP, BxPC-3-RFP, and BxPC-3 cancer cells implanted in 6-week-old female athymic mice. Two weeks postimplantation, diagnostic laparoscopy was performed with a Stryker L9000 LED light source or a Stryker X8000 xenon light source 24 hours after tail-vein injection of CEA antibodies conjugated with Alexa 488 or Alexa 555. Cancer lesions were detected and localized under each light mode. Intravital images were also obtained with the OV-100 Olympus and Maestro CRI Small Animal Imaging Systems, serving as a positive control. Tumors were collected for histologic analysis. RESULTS: Fluorescence laparoscopy with a 495-nm emission filter and an LED light source enabled real-time visualization of the fluorescence-labeled tumor deposits in the peritoneal cavity. The simultaneous use of different fluorophores (Alexa 488 and Alexa 555), conjugated to antibodies, brightened the fluorescence signal, enhancing detection of submillimeter lesions without compromising background illumination. Adjustments to the LED light source permitted simultaneous detection of tumor lesions of different fluorescent colors and surrounding structures with minimal autofluorescence. CONCLUSIONS: Using an LED light source with adjustments to the red, blue, and green wavelengths, it is possible to simultaneously identify tumor metastases expressing fluorescent proteins of different wavelengths, which greatly enhanced the signal without compromising background illumination. Development of this fluorescence laparoscopy technology for clinical use can improve staging and resection of pancreatic cancer.


Asunto(s)
Fluorescencia , Colorantes Fluorescentes , Laparoscopía/métodos , Iluminación/instrumentación , Neoplasias Experimentales/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Animales , Diseño de Equipo , Femenino , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/secundario , Neoplasias Pancreáticas/secundario , Reproducibilidad de los Resultados , Células Tumorales Cultivadas/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...