Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Ann Surg Oncol ; 22 Suppl 3: S1469-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25893411

ABSTRACT

BACKGROUND: Photoimmunotherapy (PIT) uses a target-specific photosensitizer based on a near-infrared (NIR) phthalocyanine dye, IR700, to induce tumor necrosis after irradiation with NIR light to kill cancer cells, such as those that remain after surgery. The purpose of the present study was to sterilize the surgical bed after pancreatic cancer resection with PIT in carcinoembryonic antigen (CEA)-expressing, patient-derived, orthotopic xenograft (PDOX) nude mouse models. METHODS: After confirmation of tumor engraftment, mice were randomized to two groups: bright light surgery (BLS)-only and BLS + PIT. Each treatment arm consisted of seven tumor-bearing mice. BLS was performed under standard bright-field with an MVX10 long-working distance, high-magnification microscope on all mice. For BLS + PIT, anti-CEA antibody conjugated with IR700 (anti-CEA-IR700) (50 µg) was injected intravenously in all mice 24 h before surgery. After the surgery, the resection bed was then irradiated with a red-light-emitting diode at 690 ± 5 nm with a power density of 150 mW/cm(2). RESULTS: Anti-CEA-IR700 labelled and illuminated the pancreatic cancer PDOX. Minimal residual cancer of the PDOX was detected by fluorescence after BLS. The local recurrence rate was 85.7 % for BLS-only and 28.6 % for BLS + PIT-treated mice (p = 0.05). The average recurrent tumor weight was 1149.0 ± 794.6 mg for BLS-only and 210.8 ± 336.9 mg for BLS + PIT-treated mice (p = 0.015). CONCLUSION: Anti-CEA-IR700 was able to label and illuminate a pancreatic cancer PDOX nude mouse model sufficiently for PIT. PIT reduced recurrence by eliminating remaining residual cancer cells after BLS.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carcinoembryonic Antigen/immunology , Immunotherapy , Neoplasm Recurrence, Local/drug therapy , Pancreatic Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Animals , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/surgery , Neoplasm, Residual/drug therapy , Neoplasm, Residual/immunology , Neoplasm, Residual/surgery , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/surgery , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
PLoS One ; 10(3): e0121989, 2015.
Article in English | MEDLINE | ID: mdl-25799218

ABSTRACT

Photoimmunotherapy (PIT) of cancer utilizes tumor-specific monoclonal antibodies conjugated to a photosensitizer phthalocyanine dye IR700 which becomes cytotoxic upon irradiation with near infrared light. In this study, we aimed to evaluate the efficacy of PIT on human pancreatic cancer cells in vitro and in vivo in an orthotopic nude mouse model. The binding capacity of anti-CEA antibody to BxPC-3 human pancreatic cancer cells was determined by FACS analysis. An in vitro cytotoxicity assay was used to determine cell death following treatment with PIT. For in vivo determination of PIT efficacy, nude mice were orthotopically implanted with BxPC-3 pancreatic tumors expressing green fluorescent protein (GFP). After tumor engraftment, the mice were divided into two groups: (1) treatment with anti-CEA-IR700 + 690 nm laser and (2) treatment with 690 nm laser only. Anti-CEA-IR700 (100 µg) was administered to group (1) via tail vein injection 24 hours prior to therapy. Tumors were then surgically exposed and treated with phototherapy at an intensity of 150 mW/cm2 for 30 minutes. Whole body imaging was done subsequently for 5 weeks using an OV-100 small animal imaging system. Anti-CEA-IR700 antibody bound to the BxPC3 cells to a high degree as shown by FACS analysis. Anti-CEA-IR700 caused extensive cancer cell killing after light activation compared to control cells in cytotoxicity assays. In the orthotopic models of pancreatic cancer, the anti-CEA-IR700 group had significantly smaller tumors than the control after 5 weeks (p<0.001). There was no significant difference in the body weights of mice in the anti-CEA-IR700 and control groups indicating that PIT was well tolerated by the mice.


Subject(s)
Carcinoembryonic Antigen/immunology , Immunotherapy , Pancreatic Neoplasms/therapy , Phototherapy , Animals , Antibodies, Monoclonal , Cell Line, Tumor , Humans , Infrared Rays , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Radiation-Sensitizing Agents/therapeutic use , Tumor Burden , Xenograft Model Antitumor Assays
3.
J Surg Res ; 197(1): 5-11, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25799527

ABSTRACT

BACKGROUND: Photoimmunotherapy (PIT) is based on the use of a monoclonal antibody specific to cancer epitopes conjugated to a photosensitizer near-infrared phthalocyanine dye (IR700). In this study, PIT with IR700 conjugated to anti-carcinoembryonic antigen (CEA) was used as an adjunct to surgery in orthotopically-implanted human pancreatic cancer in a nude mouse model to eliminate microscopic disease in the post-surgical tumor bed and prevent local as well as metastatic recurrence. MATERIALS AND METHODS: Athymic nude mice were orthotopically implanted with the human pancreatic cancer cell line BxPC3 expressing green fluorescent protein. After tumor engraftment, the mice were divided into two groups as follows: bright light surgery (BLS) + anti-CEA-IR700 + 690 nm laser (PIT); and BLS only. Anti-CEA-IR700 (100 µg) was administered to the treatment group via tail-vein injection 24 h before therapy. Tumors were resected, and the surgical bed was treated with intraoperative phototherapy at an intensity of 150 mW/cm(2) for 30 min. Mice were imaged noninvasively for 8 wk using an OV-100 small animal fluorescence imager. RESULTS: BLS + PIT reduced local recurrence to 1/7 mice from 7/7 mice with BLS-only (P = 0.001) and metastatic recurrence to 2/7 mice compared with 6/7 mice with BLS-only (P = 0.03). Local tumor growth continued at a rapid rate after BLS-only compared with BLS + PIT where almost no local growth occurred. There was a significant difference in tumor size between mice in the BLS + PIT (2.14 mm(2), 95% confidence interval [CI] [-2.06 to 6.34] and BLS-only groups (115.2 mm(2), 95% CI [88.8-141.6]) at 6 wk after surgery (P < 0.001). There was also a significant difference in tumor weight between the BLS + PIT group (6.65 mg, 95% CI [-6.35 to 19.65] and BLS-only group (1100 mg, 95% CI [794-1406] at 8 wk after surgery (P < 0.001). CONCLUSIONS: PIT holds promise in the treatment of pancreatic cancer and may serve as a useful adjunct to surgery in the eradication of microscopic residual disease that can lead to both local and metastatic recurrence. Further studies are warranted to investigate the potential toxicities of PIT, especially with regard to anastomoses, such as those involved in pancreaticoduodenectomy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Indoles/therapeutic use , Neoplasm Recurrence, Local/prevention & control , Pancreatectomy , Pancreatic Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Animals , Carcinoembryonic Antigen/immunology , Cell Line, Tumor , Chemotherapy, Adjuvant , Humans , Isoindoles , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/surgery , Treatment Outcome
4.
J Biomed Opt ; 19(10): 101504, 2014.
Article in English | MEDLINE | ID: mdl-24887695

ABSTRACT

Labeling of metastatic tumors can aid in their staging and resection of cancer. Near infrared (NIR) dyes have been used in the clinic for tumor labeling. However, there can be a nonspecific uptake of dye by the liver, lungs, and lymph nodes, which hinders detection of metastasis. In order to overcome these problems, we have used two NIR dyes (DyLight 650 and 750) conjugated to a chimeric anti-carcinoembryonic antigen antibody to evaluate how polyethylene glycol linkage (PEGylation) can improve specific tumor labeling in a nude mouse model of human pancreatic cancer. The conjugated PEGylated and non-PEGylated DyLight 650 and 750 dyes were injected intravenously into non-tumor-bearing nude mice. Serum samples were collected at various time points in order to determine serum concentrations and elimination kinetics. Conjugated PEGylated dyes had significantly higher serum dye concentrations than non-PEGylated dyes (p=0.005 for the 650 dyes and p<0.001 for the 750 dyes). Human pancreatic tumors subcutaneously implanted into nude mice were labeled with antibody-dye conjugates and serially imaged. Labeling with conjugated PEGylated dyes resulted in significantly brighter tumors compared to the non-PEGylated dyes (p<0.001 for the 650 dyes; p=0.01 for 750 dyes). PEGylation of the NIR dyes also decreased their accumulation in lymph nodes, liver, and lung. These results demonstrate enhanced selective tumor labeling by PEGylation of dyes conjugated to a tumor-specific antibody, suggesting their future clinical use in fluorescence-guided surgery.


Subject(s)
Antibodies, Monoclonal/immunology , Carcinoembryonic Antigen/immunology , Disease Models, Animal , Microscopy, Fluorescence/methods , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Polyethylene Glycols/chemistry , Animals , Cell Line, Tumor , Cross-Linking Reagents/chemistry , Drug Carriers/chemistry , Fluorescent Dyes/chemistry , Humans , Infrared Rays , Mice , Mice, Nude , Recombinant Fusion Proteins/immunology , Reproducibility of Results , Sensitivity and Specificity , Staining and Labeling/methods
5.
PLoS One ; 9(5): e97965, 2014.
Article in English | MEDLINE | ID: mdl-24859320

ABSTRACT

We report here that polyethylene glycol (PEG) linked to near infrared dyes conjugated to chimeric mouse-human anti-carcinoembryonic antigen (CEA) antibody greatly improves imaging of liver metastases in a nude mouse model of colon-cancer experimental metastases. PEGylated and non-PEGylated DyLight 650 and 750 dyes were conjugated to the chimeric anti-CEA antibody. The dyes were initially injected intravenously into nude mice without tumors. Tissue biodistribution was determined by tissue sonication and analyzing tissue dye concentration profiles over time. PEGylated dyes had significantly lower accumulation in the liver (p = 0.03 for the 650 dyes; p = 0.002 for the 750 dyes) compared to non-PEGylated dyes. In an experimental liver metastasis model of HT-29 colon cancer, PEGylated dyes conjugated to the anti-CEA antibody showed good labeling of metastatic tumors with high contrast between normal and malignant tissue which was not possible with the non-PEGylated dyes since there was so much non-specific accumulation in the liver. PEGylation of the DyLight 650 and 750 NIR dyes significantly altered tissue biodistribution, allowing brighter tissue labeling, decreased accumulation in normal organs, particularly the liver. This enabled high fidelity and high contrast imaging of liver metastases.


Subject(s)
Antibodies, Monoclonal , Carcinoembryonic Antigen/immunology , Colonic Neoplasms/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/secondary , Optical Imaging/methods , Polyethylene Glycols/chemistry , Animals , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Disease Models, Animal , Fluorescent Dyes/chemistry , HT29 Cells , Humans , Liver/metabolism , Mice, Nude , Recombinant Fusion Proteins/chemistry , Signal-To-Noise Ratio , Whole Body Imaging
6.
J Am Coll Surg ; 219(1): 132-41, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24768506

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neoplasm , Carcinoembryonic Antigen/immunology , Laparoscopy/methods , Optical Imaging/methods , Pancreatectomy/methods , Pancreatic Neoplasms/surgery , Animals , Cell Line, Tumor , Disease Models, Animal , Disease-Free Survival , Female , Fluorescent Dyes , Fluorobenzenes , Humans , Logistic Models , Mice , Mice, Nude , Neoplasm Recurrence, Local , Neoplasm Transplantation , Pancreatic Neoplasms/immunology , Random Allocation , Treatment Outcome , Tumor Burden
7.
Ann Surg Oncol ; 21(4): 1405-11, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24499827

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal , Carcinoembryonic Antigen/immunology , Diagnostic Imaging , Fluorescent Antibody Technique/methods , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Animals , Female , Fluorescent Dyes , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/diagnosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
J Surg Oncol ; 109(5): 451-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24249594

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal , Carcinoembryonic Antigen/immunology , Chimera , Colonic Neoplasms/diagnosis , Colonic Neoplasms/surgery , Diagnostic Imaging/methods , Fluorescent Antibody Technique/methods , Image Enhancement/methods , Animals , Carbocyanines , Fluorescent Dyes , Heterografts , Humans , Mice , Mice, Nude
9.
J Biomed Opt ; 18(12): 126016, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24356647

ABSTRACT

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.


Subject(s)
Antibodies , Carcinoembryonic Antigen/immunology , Carcinoembryonic Antigen/metabolism , Fluorescent Dyes , Optical Imaging/methods , Pancreatic Neoplasms/pathology , Animals , Antibodies/chemistry , Antibodies/metabolism , Cell Line, Tumor , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Hemoglobins , Histocytochemistry , Humans , Mice , Mice, Nude , Microscopy, Fluorescence , Photobleaching
10.
J Am Coll Surg ; 214(6): 997-1007.e2, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22542065

ABSTRACT

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.


Subject(s)
Fluorescence , Fluorescent Dyes , Laparoscopy/methods , Lighting/instrumentation , Neoplasms, Experimental/diagnosis , Pancreatic Neoplasms/diagnosis , Animals , Equipment Design , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/secondary , Pancreatic Neoplasms/secondary , Reproducibility of Results , Tumor Cells, Cultured/transplantation
11.
Hepatogastroenterology ; 59(118): 1994-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22369743

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal , Carcinoembryonic Antigen/immunology , Fluorescent Antibody Technique , Laparoscopy , Neoplasm Staging/methods , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Animals , Cell Line, Tumor , Female , Fluorescent Dyes , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/secondary , Pancreatic Neoplasms/surgery , Predictive Value of Tests , Sensitivity and Specificity , Time Factors
12.
Ear Nose Throat J ; 87(8): E1, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18712683

ABSTRACT

Medullary carcinoma of the thyroid requires aggressive treatment because of its potential to metastasize and because of the current limitations of preoperative localization and systemic therapy. If these tumors could be made to fluoresce in vivo with tagged fluorophore antibodies against tumor antigens, surgeons would be able to obtain additional information in the operating room to facilitate a more complete resection. Based on the success of our previous work in breast and colon cancer models, we conducted an animal study of in vivo tumor fluorescence of a human medullary thyroid cell line in which bright tumor fluorescence is visible during dissection. To accomplish this, we used an inexpensive and commercially available handheld, blue (470 nm), light-emitting diode flashlight and filtered goggles (520 nm). This procedure, which we call the fluorescent antibody-assisted surgical technique (FAAST), is easy to perform, requires no complex or expensive technical equipment, and has the potential to be applied to a wide variety of tumors. To the best of our knowledge, this is the first experiment of its kind to be reported in the literature.


Subject(s)
Carcinoma, Medullary/diagnosis , Fluorescent Antibody Technique/instrumentation , Thyroid Neoplasms/diagnosis , Animals , Carcinoma, Medullary/pathology , Carcinoma, Medullary/surgery , Mice , Pilot Projects , Prognosis , Thyroid Neoplasms/pathology , Thyroid Neoplasms/surgery
13.
J Gastrointest Surg ; 12(11): 1938-50, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18665430

ABSTRACT

INTRODUCTION: Colorectal and pancreatic cancers together comprise the third and fourth most common causes of cancer-related death in the United States. In both of these cancers, complete detection of primary and metastatic lesions at the time of surgery is critical to optimal surgical resection and appropriate patient treatment. MATERIALS AND METHODS: We have investigated the use of fluorophore-labeled anti-carcinoembryonic antigen (CEA) monoclonal antibody to aid in cancer visualization in nude mouse models of human colorectal and pancreatic cancer. Anti-CEA was conjugated with a green fluorophore. Subcutaneous, orthotopic primary and metastatic human pancreatic and colorectal tumors were easily visualized with fluorescence imaging after administration of conjugated anti-CEA. The fluorescence signal was detectable 30 min after systemic antibody delivery and remained present for 2 weeks, with minimal in vivo photobleaching after exposure to standard operating room lighting. Tumor resection techniques revealed improved ability to resect labeled tumor tissue under fluorescence guidance. Comparison of two different fluorophores revealed differences in dose-response and photobleaching in vivo. CONCLUSION: These results indicate that fluorophore-labeled anti-CEA offers a novel intraoperative imaging technique for enhanced visualization of tumors in colorectal and pancreatic cancer when CEA expression is present, and that the choice of fluorophore significantly affects the signal intensity in the labeled tumor.


Subject(s)
Antibodies, Anti-Idiotypic , Colorectal Neoplasms/diagnostic imaging , Image Enhancement/methods , Monitoring, Intraoperative/methods , Pancreatic Neoplasms/diagnostic imaging , Radioimmunodetection , Animals , Carcinoembryonic Antigen , Colorectal Neoplasms/surgery , Diagnostic Imaging/methods , Disease Models, Animal , Fluorescent Dyes , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/surgery , Random Allocation , Sensitivity and Specificity , Transplantation, Heterologous
14.
World J Surg ; 32(6): 1057-66, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18264829

ABSTRACT

BACKGROUND: Despite recent surgical advances, pancreatic cancer remains the fourth leading cause of cancer-related death in the United States. This is due to inaccurate staging and difficulty in achieving negative margins at the time of pancreaticoduodenectomy. CA19-9 is a carbohydrate tumor-associated antigen found in up to 94% of pancreatic adenocarcinomas. In this study we investigate the use of a fluorophore-labeled anti-CA19-9 monoclonal antibody to improve intraoperative visualization of both primary and metastatic tumors in a mouse model of pancreatic cancer. METHODS: A monoclonal antibody specific for CA19-9 was conjugated to a green fluorophore and delivered to tumor-bearing mice as a single intravenous (IV) dose. Intravital fluorescence imaging was used to localize tumor implants 24 h after antibody administration. RESULTS: Using fluorescence imaging, the primary tumor was clearly visible at laparotomy, as were small metastatic implants within the liver and spleen and on the peritoneum. These tumor implants, which were nearly impossible to see using standard bright-field imaging, demonstrated clear fluorescence under LED light excitation. The fluorescence signal within the tumor tissue was maintained for over 3 weeks after a single administration of the labeled antibody. Histologic evaluation of tissue from animals treated with the conjugated anti-CA19-9 antibody likewise revealed strong staining of the tumor cells with minimal background staining of the peritumoral stroma. CONCLUSIONS: Fluorophore-labeled anti-CA19-9 offers a novel intraoperative imaging technique for enhanced visualization of primary and metastatic tumors in pancreatic cancer when CA19-9 expression is present and may improve intraoperative staging and efficacy of resection.


Subject(s)
Antibodies, Monoclonal/immunology , CA-19-9 Antigen/immunology , Liver Neoplasms/diagnosis , Pancreatic Neoplasms/diagnosis , Splenic Neoplasms/diagnosis , Stereotaxic Techniques , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Liver Neoplasms/secondary , Mice , Mice, Nude , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Splenic Neoplasms/secondary
SELECTION OF CITATIONS
SEARCH DETAIL
...