Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Ann Surg Oncol ; 22(6): 2082-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25319581

RESUMO

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.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Recidiva Local de Neoplasia/prevenção & controle , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , Cirurgia Assistida por Computador , Animais , Peptídeos Penetradores de Células/metabolismo , Feminino , Fluorescência , Proteínas de Fluorescência Verde/metabolismo , Humanos , Metaloproteinase 2 da Matriz/química , Metaloproteinase 9 da Matriz/química , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Metástase Neoplásica , Imagem Óptica/métodos , Carga Tumoral
2.
J Am Coll Surg ; 219(1): 132-41, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24768506

RESUMO

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.


Assuntos
Anticorpos Monoclonais , Anticorpos Antineoplásicos , Antígeno Carcinoembrionário/imunologia , Laparoscopia/métodos , Imagem Óptica/métodos , Pancreatectomia/métodos , Neoplasias Pancreáticas/cirurgia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Intervalo Livre de Doença , Feminino , Corantes Fluorescentes , Fluorbenzenos , Humanos , Modelos Logísticos , Camundongos , Camundongos Nus , Recidiva Local de Neoplasia , Transplante de Neoplasias , Neoplasias Pancreáticas/imunologia , Distribuição Aleatória , Resultado do Tratamento , Carga Tumoral
3.
J Laparoendosc Adv Surg Tech A ; 24(4): 241-7, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24494971

RESUMO

BACKGROUND: Fluorescence-guided surgery (FGS) can enable successful cancer surgery where bright-light surgery often cannot. There are three important issues for FGS going forward toward the clinic: (a) proper tumor labeling, (b) a simple portable imaging system for the operating room, and (c) patient-like mouse models in which to develop the technology. The present report addresses all three. MATERIALS AND METHODS: Patient colon tumors were initially established subcutaneously in nonobese diabetic (NOD)/severe combined immune deficiency (SCID) mice immediately after surgery. The tumors were then harvested from NOD/SCID mice and passed orthotopically in nude mice to make patient-derived orthotopic xenograft (PDOX) models. Eight weeks after orthotopic implantation, a monoclonal anti-carcinoembryonic antigen (CEA) antibody conjugated with AlexaFluor 488 (Molecular Probes Inc., Eugene, OR) was delivered to the PDOX models as a single intravenous dose 24 hours before laparotomy. A hand-held portable fluorescence imaging device was used. RESULTS: The primary tumor was clearly visible at laparotomy with the portable fluorescence imaging system. Frozen section microscopy of the resected specimen demonstrated that the anti-CEA antibody selectively labeled cancer cells in the colon cancer PDOX. The tumor was completely resected under fluorescence navigation. Histologic evaluation of the resected specimen demonstrated that cancer cells were not present in the margins, indicating successful tumor resection. The FGS animals remained tumor free for over 6 months. CONCLUSIONS: The results of the present report indicate that FGS using a fluorophore-conjugated anti-CEA antibody and portable imaging system improves efficacy of resection for CEA-positive colorectal cancer. These data provide the basis for clinical trials.


Assuntos
Neoplasias do Colo/patologia , Neoplasias do Colo/cirurgia , Imagem Óptica/métodos , Cirurgia Assistida por Computador/métodos , Animais , Anticorpos Anti-Idiotípicos , Antígeno Carcinoembrionário/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Hidrazinas , Camundongos , Camundongos Nus , Camundongos SCID , Transplante Heterólogo
4.
Ann Surg Oncol ; 21(4): 1405-11, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24499827

RESUMO

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.


Assuntos
Anticorpos Monoclonais , Antígeno Carcinoembrionário/imunologia , Diagnóstico por Imagem , Imunofluorescência/métodos , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/cirurgia , Animais , Feminino , Corantes Fluorescentes , Humanos , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/diagnóstico , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Surg Oncol ; 109(5): 451-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24249594

RESUMO

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.


Assuntos
Anticorpos Monoclonais , Antígeno Carcinoembrionário/imunologia , Quimera , Neoplasias do Colo/diagnóstico , Neoplasias do Colo/cirurgia , Diagnóstico por Imagem/métodos , Imunofluorescência/métodos , Aumento da Imagem/métodos , Animais , Carbocianinas , Corantes Fluorescentes , Xenoenxertos , Humanos , Camundongos , Camundongos Nus
6.
Ann Surg Oncol ; 20 Suppl 3: S693-700, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23943029

RESUMO

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.


Assuntos
Anticorpos Monoclonais , Modelos Animais de Doenças , Corantes Fluorescentes , Tumores do Estroma Gastrointestinal/diagnóstico , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/imunologia , Animais , Feminino , Fluorescência , Tumores do Estroma Gastrointestinal/imunologia , Humanos , Técnicas Imunoenzimáticas , Laparoscopia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética
7.
J Surg Res ; 184(1): 290-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23590868

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Camundongos Nus , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/secundário , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Animais , Linhagem Celular Tumoral , Feminino , Proteínas de Fluorescência Verde/genética , Humanos , Estimativa de Kaplan-Meier , Proteínas Luminescentes/genética , Camundongos , Transplante de Neoplasias , Neoplasias Pancreáticas/mortalidade , Proteínas Proto-Oncogênicas p21(ras) , Proteína Vermelha Fluorescente
8.
Gastroenterol Res Pract ; 2013: 290634, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23533387

RESUMO

There are many challenges that face surgeons when attempting curative resection for gastrointestinal cancers. The ability to properly delineate tumor margins for complete resection is of utmost importance in achieving cure and giving the patient the best chance of prolonged survival. Targeted tumor imaging techniques have gained significant interest in recent years to enable better identification of tumor lesions to improve diagnosis and treatment of cancer from preoperative staging modalities to optimizing the surgeon's ability to visualize tumor margins at the initial operation. Using unique characteristics of the tumor to fluorescently label the tissue can delineate tumor margins from normal surrounding tissue, allowing improved precision of surgical resection. In this paper, different methods of fluorescently labeling native tumor are discussed as well as the development of fluorescence laparoscopy and the potential role for fluorescence-guided surgery in the treatment of gastrointestinal cancers.

9.
J Surg Res ; 179(1): 87-93, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23079571

RESUMO

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.


Assuntos
Neoplasias Colorretais/cirurgia , Procedimentos Cirúrgicos do Sistema Digestório/métodos , Modelos Animais de Doenças , Imagem Óptica/métodos , Transplante Heterólogo , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Intervalo Livre de Doença , Feminino , Imunofluorescência , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Nus , Prevenção Secundária , Resultado do Tratamento
10.
J Am Coll Surg ; 215(1): 126-35; discussion 135-6, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22632917

RESUMO

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.


Assuntos
Proteínas Luminescentes/análise , Pancreatectomia/métodos , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/cirurgia , Animais , Modelos Animais de Doenças , Intervalo Livre de Doença , Feminino , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Células Tumorais Cultivadas , Proteína Vermelha Fluorescente
11.
J Am Coll Surg ; 214(6): 997-1007.e2, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22542065

RESUMO

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.


Assuntos
Fluorescência , Corantes Fluorescentes , Laparoscopia/métodos , Iluminação/instrumentação , Neoplasias Experimentais/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Animais , Desenho de Equipamento , Feminino , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/secundário , Neoplasias Pancreáticas/secundário , Reprodutibilidade dos Testes , Células Tumorais Cultivadas/transplante
12.
Hepatogastroenterology ; 59(118): 1994-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22369743

RESUMO

BACKGROUND/AIMS: Laparoscopy is important in staging pancreatic cancer, but false negatives remain problematic. Making tumors fluorescent has the potential to improve the accuracy of staging laparoscopy. METHODOLOGY: Orthotopic and carcinomatosis models of pancreatic cancer were established with BxPC-3 human pancreatic cancer cells in nude mice. Alexa488-antiCEA conjugates were injected via tail vein 24 hours prior to laparoscopy. Mice were examined under bright field laparoscopic (BL) and fluorescence laparoscopic (FL) modes. Outcomes measured included time to identification of primary tumor for the orthotopic model and number of metastases identified within 2 minutes for the carcinomatosis model. RESULTS: FL enabled more rapid and accurate identification and localization of primary tumors and metastases than BL. Using BL took statistically significantly longer time than FL (p<0.0001, fold change and 95% CI for BL vs. FL: 8.12 (4.54,14.52)). More metastatic lesions were detected and localized under FL compared to BL and with greater accuracy, with sensitivities of 96% vs. 40%, respectively, when compared to control. FL was sensitive enough to detect metastatic lesions <1mm. CONCLUSIONS: The use of fluorescence laparoscopy with tumors labeled with fluorophore-conjugated anti-CEA antibody permits rapid detection and accurate localization of primary and metastatic pancreatic cancer in an orthotopic model. The results of the present report demonstrate the future clinical potential of fluorescence laparoscopy.


Assuntos
Anticorpos Monoclonais , Antígeno Carcinoembrionário/imunologia , Imunofluorescência , Laparoscopia , Estadiamento de Neoplasias/métodos , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Animais , Linhagem Celular Tumoral , Feminino , Corantes Fluorescentes , Humanos , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/secundário , Neoplasias Pancreáticas/cirurgia , Valor Preditivo dos Testes , Sensibilidade e Especificidade , Fatores de Tempo
13.
J Laparoendosc Adv Surg Tech A ; 21(6): 485-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21699431

RESUMO

BACKGROUND: Staging laparoscopy can visualize peritoneal and liver metastases in pancreatic cancer otherwise undetectable by preoperative imaging. However, false-negative rates may be as high as 18%-26%. The aim of the present study was to improve detection of metastatic pancreatic cancer with the use of fluorescence laparoscopy (FL) in a nude-mouse model with the tumors expressing green fluorescent protein (GFP). METHODS: The carcinomatosis mouse model of human pancreatic cancer was established by intraperitoneal injections of green fluorescent protein-expressing MiaPaca-2 human pancreatic cancer cells into 6-week-old female athymic mice. Two weeks later, mice underwent diagnostic laparoscopy. Laparoscopy was performed first under standard brightfield lighting, followed by fluorescent lighting. The number of metastatic foci identified within the four quadrants of the peritoneal cavity was recorded. After laparoscopy, the animals were sacrificed, opened, and imaged with the OV-100 Small Animal Imaging system as a positive control to identify metastasis. Tumors were collected and processed for histologic review. RESULTS: FL enabled visualization of pancreatic cancer metastatic foci not visualized with standard brightfield laparoscopy (BL). Under FL, in 1 representative mouse, 26 separate micrometastatic lesions were identified. In contrast, only very large tumors were seen using BL. Use of the OV-100 images, as positive controls, confirmed the presence of tumor foci. FL thus allowed identification and exact localization of submillimeter tumor foci. Such small-sized tumor foci were not distinguished from surrounding tissue under BL. All malignant lesions were histologically confirmed. CONCLUSIONS: The use of FL enables the identification of tumor foci that cannot be seen with standard laparoscopy. The technology described in this report has important potential for the clinical development of FL.


Assuntos
Carcinoma/secundário , Laparoscopia/métodos , Neoplasias Pancreáticas/patologia , Animais , Modelos Animais de Doenças , Feminino , Fluorescência , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...