Abstract
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 mm2 ± SE 52.03 vs. 2.20 mm2 ± SE 1.43; respectively, χ2 = 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.
A number of different methods have been used to label tumors for fluorescence-guided surgery (FGS).1,2 Several of these methods include fluorophore-conjugated antibodies to unique surface markers expressed by individual tumor types3–9 and replication-competent viruses engineered to express the green fluorescent protein (GFP) in the presence of activated telomerase10,11 to selectively label tumors for effective FGS.
Another technique for in vivo labeling of cancer cells involves the use of activatable cell-penetrating peptides (ACPPs).12–16 Polycationic cell-penetrating peptides (CPPs) are connected via a cleavable linker to a neutralizing polyanion whose adsorption and uptake into cells are inhibited until the linker is proteolyzed. With the upregulation of matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) in most solid tumors, exposure to these proteases results in cleavage and dissociation of the linked peptide, allowing it to bind to and enter tumor cells. Conjugating CPPs to a fluorophore(s) enables the tumor to acquire a fluorescent label.
The aim of this study was to develop a method to accurately visualize primary pancreatic tumors labeled with ratiometric activatable cell-penetrating peptides (RACPPs), conjugated to Cy5 and Cy7 fluorophores, in orthotopic murine models of human pancreatic cancer for effective FGS. The advantages of the use of RACPPs over single fluorophore ACPPs for tumor imaging include imaging and quantification that do not depend on the total probe uptake, varying washout of the ACPPs, or on thresholding. Also, RACPPs have a rapid change that can be detected within 1–2 h (vs. 6 h for single fluorophore versions12,15 and 24 h for dequenching probes17), which is more applicable to rapid, intraoperative use. Finally, the RACPP dequenching mechanism improves the contrast of Cy5 alone compared with a single fluorophore ACPP,16 enhancing the detection specificity/sensitivity and making ACPPs immediately adaptable to the equipment already available in FGS clinical operating rooms.
MATERIALS AND METHODS
Cell Culture
MiaPaCa-2–green fluorescent protein (GFP)-expressing pancreatic cancer cells were maintained as previously described in RPMI 1640 medium with 10 % fetal bovine serum.18
Peptide Synthesis and Fluorophore Labeling
Cy5 and Cy7 ratiometric activatable cell-penetrating peptides (RACPPs) were synthesized on an automatic peptide synthesizer using standard protocols for fluorenylmethoxycarbonyl solid-phase synthesis as previously described.16 All reactions were monitored with analytic HPLC and mass spectrometry (MS). According to characterization with HPLC-MS, all products were >95 % pure.
Animal Care
Female athymic nu/nu nude mice were maintained in a barrier facility on high-efficiency particulate air-filtered racks. All surgical procedures were performed under anesthesia with an intramuscular injection of 100 μL of a mixture of 100 mg/kg ketamine and 10 mg/kg xylazine. A total of 32 mice were used for these experiments. All animal studies were approved by the University of California, San Diego Institutional Animal Care and Use Committee and were conducted in accordance with the principles and procedures outlined in the National Institutes of Health Guide for the Care and Use of Animals.
Subcutaneous Tumor Cell Implantation
Human MiaPaCa-2–GFP pancreatic cancer cells were harvested by trypsinization and prepared for injection. Cells (2 × 106 in 100 μL serum-free media) were injected subcutaneously within 30 min of harvesting over the right and left flanks in female nu/nu mice between 4 and 6 weeks of age. Subcutaneous tumors were allowed to grow for 2–4 weeks until large enough to supply adequate tumors for orthotopic implantation.
Orthotopic Implantation of Tumor Tissue Fragments
Surgical orthotopic implantation of single 1 mm3 tumor fragments from the fluorescent MiaPaCa-2–GFP subcutaneous tumors were used to establish orthotopic models in nude mice. The tail of the pancreas was delivered through a small 6–10 mm transverse incision made on the left flank of the mouse. The tumor fragment was sutured to the tail of the pancreas using 8-0 nylon surgical sutures. Upon completion, the pancreas was returned to the abdomen and the incision was closed in two layers using 6.0 Ethibond nonabsorbable sutures (Ethicon Inc., Somerville, NJ).19–21
Imaging and Resection of Tumors
Thirty-two tumor-bearing mice were randomized to undergo FGS or WLR. For FGS, resection of the primary pancreatic tumor using fluorescence-guided molecular navigation was performed 2 h after mice were injected with RACPPs (10 nmol, i.v.). A customized fluorescence dissecting microscope (MVX10, Olympus Corporation, Tokyo, Japan) with an exposure time of 0.5–1 s, excitation of 620/60 nm, and emission of 700/75 nm (Cy5) or 785/62 nm (Cy7) was used for intraoperative optical imaging. The ratio of Cy5 to Cy7 signal (representing cleaved, activated peptide over uncleaved, intact peptide) was imaged with custom-designed software displaying a blue (low ratio, mostly uncleaved) to red (high ratio, mostly cleaved) color scale for surgical guidance. Tissue was considered tumor and resected if it had a >20 % ratio increase over background pancreas tissue. Tumor-bearing mice randomized to the WLR arm underwent resection under standard bright light. Whole body preoperative and postoperative images (GFP, excitation of 466/44 nm, emission tuned to 520 nm) were performed on all mice using a Maestro CRI Small Animal Spectral Separation Imaging System (Cambridge Research & Instrumentation, Inc. Woburn, MA) to confirm the presence of the tumor and completeness of the resection. Postoperatively, the mice underwent weekly whole body imaging of GFP to assess for recurrence using the Maestro. Eight weeks postoperatively, the surviving mice (n = 31) were terminated for intravital and ex vivo imaging, using the OV-100 Small Animal Imaging System (Olympus Corporation) with an exposure time of 1.5–3 s, excitation of 460–490 nm, and emission of 510 nm long-pass to assess for local and distant recurrence, and overall pancreatic and metastatic tumor burden. All images were analyzed with Image J version 1.440 (National Institutes of Health, Bethesda, MD). The intensity of the fluorescence signal per pixel can be measured with this software, and the area of fluorescence can be calculated with a set scale.
Tissue Histology
Samples were collected during the experiments and at necropsy for histologic preparation with hematoxylin and eosin (H&E) staining. Fresh tissue samples were fixed in Bouin’s solution and regions of interest were embedded in paraffin prior to sectioning and staining with H&E for standard light microscopy.
Data Processing and Statistical Analysis
SAS, version 9.2 (The SAS Institute, Inc.) was used for statistical analyses. Continuous variables (tumor weight and metastatic tumor burden) are described using mean ± SE. The normality of the variables was assessed by visual inspection of histograms and normal Q-Q plots. A Welch’s t test or Wilcoxon rank sum test was used to compare groups, as appropriate. Categorical variables (complete resection, local and distant recurrence, and presence of tumor at termination) were expressed as counts and percentages, and tests of significance used Fisher’s exact test. We compared the disease-free survival (DFS) between treatment groups using a log rank test. A two-sided p value ≤0.05 was considered statistically significant for all comparisons.
RESULTS
Our primary aim was to evaluate the efficacy of using Cy5 and Cy7-conjugated RACPPs to guide resection of the fluorescently labeled tumor. Orthotopic mouse models of the human pancreatic cancer MiaPaCa-2–GFP were established in 32 mice. Tumors were permitted to grow for 2 weeks after which mice were randomized to FGS or WLR. FGS was performed with the MVX10 fluorescence-dissecting microscope 2 h after injection of ACPPs (Fig. 1).
FIG. 1.

Comparison of FGS and WLR imaging. Intraoperative images during FGS of a mouse showing a white light reflectance view (a), ratiometric fluorescence view (b), and the overlay of the two (c). A tumor that is difficult to detect with bright field imaging has clear margins with ratiometric imaging. White dashed lines indicates the pancreas, spleen, and tumor outlines. The displayed ratio scale ranges from 2 to 9. P pancreas, S spleen, T tumor *Region with motion artifact from movement of the surgical scissors; also note that the scissors were in different positions in a and b
With fluorescence guidance, all 16 mice underwent a complete resection as evident by lack of GFP fluorescence in postoperative images. However, 1 of the 16 mice in the WLR group had evidence of residual tumor postoperatively and was not included in later analysis. The WLR-treated mice had early recurrence of tumor and significant tumor growth over the postoperative period that led to premature morbidity, whereas a larger percentage of the FGS-treated mice remained tumor free and healthy (Fig. 2).
FIG. 2.

Comparison of resection with FGS or WLR. Postoperative whole body noninvasive images of the white light reflectance (WLR) and fluorescence guided surgery (FGS) of the mice. The WLR treated mouse (i–iii) had an incomplete resection that resulted in early recurrence of the tumor by 2 weeks (white arrow) and a significant tumor growth over the next 6 weeks that led to early morbidity. In contrast, the FGS mouse (iv–vi) that underwent a complete resection remained tumor free in the postoperative period. The Maestro imaging settings were as follows: excitation 466/44 nm, emission 515 nm long pass (limited to 520–530 for ii–vi), and exposure time of 1 s
All mice were followed postoperatively for 8 weeks with whole body imaging of the GFP-expressing pancreatic cancer to assess for recurrence. FGS significantly lengthened the DFS from 5.33 weeks in the WLR group to 7.38 weeks (p = 0.02) (Fig. 3a). Furthermore, with improved visualization of tumor margins and thus enhanced resection, recurrence rates were significantly decreased with FGS from 73 38 % (p = 0.049) (Fig. 3b). At 8 weeks postoperatively, all mice were terminated and their abdomens were exposed to assess primary and metastatic tumor burden and local and distant recurrence rates between the two groups. Mice in the WLR group demonstrated significantly larger primary pancreatic tumors compared to mice in the FGS group (1.72 g ± SE 0.58 vs. 0.25 g ± SE 0.14; p = 0.026) (Fig. 3c). The metastatic tumor burden was also significantly greater in the WLR group (96.92 mm2 ± SE 52.03 vs. 2.20 mm2 ± SE 1.43; χ2 = 5.455; p = 0.02) (Fig. 3d). Furthermore, mice in the FGS group experienced lower local and distant recurrence rates (31 vs. 67 and 25 vs. 60 %, respectively) compared to mice in the WLR group.
FIG. 3.
Disease free survival, tumor weight at termination, and meta static tumor burden. a Recurrence in the postoperative period was defined as the presence of a green fluorescent protein (GFP) signal on whole body noninvasive imaging. Mice from the white light reflectance (WLR) group demonstrated shorter time to recurrence and thus a significantly shortened disease free survival (DFS) (p = 0.02). Median DFS for the WLR was 4 weeks. Fewer than 50 % of mice in the fluorescence guided surgery (FGS) group had evidence of recurrence during the follow up period. b Mice were terminated at 8 weeks postoperatively. The WLR group (i) demonstrated greater metastatic burden and higher local and distant recurrence rates compared to the FGS group (ii) (31 vs. 67 % for local recurrence, respectively; and 25 vs. 60 % for distant recurrence; respectively). c Improved resection rates in the FGS group resulted in a significantly smaller primary pancreatic tumor burden than in the WLR group at 8 weeks (1.72 g ± SE 0.58 vs. 1.25 g ± SE 0.14; respectively; p 0.026). d Metastatic tumor burden was also significantly less in mice from FGS compared to those in the WLR group at 8 weeks (χ2 = 5.455; p = 0.02)
DISCUSSION
In this study, we demonstrated that FGS with RACPPs can enhance pancreatic tumor resection. RACPPs, once exposed to proteases that are characteristic of pancreatic tumors, were cleaved and absorbed by cancer cells, thereby highlighting the delineation of invasive tumor from surrounding normal tissue. RACPPs enabled high absolute tumor fluorescence and, with minimal uptake by surrounding tissues such as the skin, provided a strong tumor-to-background fluorescence contrast that allowed the surgeon to better distinguish the tumor from normal tissue.
FGS can enhance detection of tumor margins and thus provide a more objective means of identifying and ultimately resecting solid tumors. Initial studies explored the use of a near-infrared fluorescent dye, indocyanine green (ICG), to label sentinel lymph nodes in breast cancer patients.22 Other surgeons have used ICG as an aid in liver resection.23 However, indocyanine green does not specifically label cancer cells. Another method was described by Stummer et al.24 in which the metabolite 5-aminolevulinic acid, a precursor of hemoglobin, resulted in accumulation of porphyrins within malignant glioma. In a clinical trial, this method of FGS led to more complete resections and improved 6-month progression-free survival.
Numerous preclinical methods of more specifically labeling tumor cells have been described and tested in mouse models. For example, genetic labeling of tumors with green fluorescent protein in situ has been achieved. Stiles et al.25 were able to label metastatic lung tumor foci of >1 mm in a mouse model using a herpes simplex virus (NV1066) that carried the gfp gene. Kishimoto et al.10 used a telomerase-dependent adenovirus (OBP-401) also containing the gfp gene to accurately label tumors. Once labeled, these tumors could be resected under fluorescence guidance.11 Furthermore, tumors that recurred also maintained GFP expression, making detection of recurrence and future metastasis possible.26
Our laboratory has demonstrated the efficacy of using fluorescently labeled antibodies directed toward specific tumor antigens, a more clinically translatable method, to improve resection rates and thus surgical outcomes in mouse models of pancreatic and colon cancer.3,4,6–9 This method was tested in a clinical trial for patients with ovarian cancer by van Dam et al.27 in which they targeted their fluorescently labeled antibodies toward the folate receptor-α. In this study, the number of tumor deposits detected by surgeons when guided by tumor-specific fluorescence was significantly higher than with visual observation alone. However, two of five patients in this study showed weak or no fluorescence.
Although antibody approaches may be successful for individual tumor types that express unique surface markers, they are often not applicable to all tumor types.3,4 RACPPs are molecular probes with wide applicability in a variety of tumor types. In addition, RACPPs bind tightly to tumors, providing ample signal with low background illumination, as previously demonstrated.12–16 RACPPs can also be used at multiple stages in the evaluation of pancreatic cancer, such as preoperative and postoperative imaging in addition to surgical guidance. Although the RACPP labeling method is useful only in those tumors that express an appropriate protease that can cleave and activate the RACPP, MMP-2 and MMP-9 activity is reported to be upregulated in human pancreatic cancer.28–30 Also, all of more than 20 human xenograft and murine syngeneic tumor lines have sufficient MMP-2 and MMP-9 activity to be detectable with RACPPs, in our experience.
CONCLUSIONS
In this study, we demonstrated that accurately labeling pancreatic tumors with RACPPs in orthotopic mouse models of human pancreatic cancer improved surgical resection under fluorescence guidance, decreased recurrence, and increased tumor-free survival compared to bright light surgery. Future studies will determine whether RACPPs are superior to antibodies for imaging pancreatic cancer.
ACKNOWLEDGMENT
This work was supported in part by grants CA132971 and CA142669 from the National Cancer Institute (to M.B. and AntiCancer, Inc.), T32 training grant CA121938 (to C.A.M.), training grant 5R25CA153915 and fellowship grant 1F30HL118998-01 (to C.N.F.), Burrough-Wellcome Fund CAMS and 5K08EB008122 from the National Institutes of Health (to Q.T.N.), and CA158448 from the National Cancer Institute (to R.Y.T.). The authors would also like to thank Elamprakash Savariar for synthesizing the ratiometric imaging agents and Paul Steinbach for surgical imaging support.
Footnotes
This work was presented at the 67th Annual Society of Surgical Oncology Cancer Symposium in Phoenix, Arizona held on March 12 15, 2014.
REFERENCES
- 1.Bouvet M, Hoffman RM. Glowing tumors make for better detection and resection. Sci Transl Med. 2011;3(110):110fs110. doi: 10.1126/scitranslmed.3003375. [DOI] [PubMed] [Google Scholar]
- 2.Nguyen QT, Tsien RY. Fluorescence guided surgery with live molecular navigation a new cutting edge. Nat Rev Cancer. 2013;13(9):653–62. doi: 10.1038/nrc3566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kaushal S, McElroy MK, Luiken GA, et al. Fluorophore conjugated anti CEA antibody for the intraoperative imaging of pancreatic and colorectal cancer. J Gastrointest Surg. 2008;12(11):1938–50. doi: 10.1007/s11605-008-0581-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.McElroy M, Kaushal S, Luiken GA, et al. Imaging of primary and metastatic pancreatic cancer using a fluorophore conjugated anti CA19-9 antibody for surgical navigation. World J Surg. 2008;32(6):1057–66. doi: 10.1007/s00268-007-9452-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hiroshima Y, Maawy A, Metildi CA, et al. Successful fluores cence guided surgery on human colon cancer patient derived orthotopic xenograft mouse models using a fluorophore conju gated anti CEA antibody and a portable imaging system. J Laparoendosc Adv Surg Tech A. 2014;24(4):241–7. doi: 10.1089/lap.2013.0418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Metildi CA, Kaushal S, Lee C, et al. An LED light source and novel fluorophore combinations improve fluorescence laparo scopic detection of metastatic pancreatic cancer in orthotopic mouse models. J Am Coll Surg. 2012;214(6):997–1007.e2. doi: 10.1016/j.jamcollsurg.2012.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Metildi CA, Kaushal S, Luiken GA, Talamini MA, Hoffman RM, Bouvet M. Fluorescently labeled chimeric anti CEA antibody improves detection and resection of human colon cancer in a patient derived orthotopic xenograft (PDOX) nude mouse model. J Surg Oncol. 2014;109(5):451–8. doi: 10.1002/jso.23507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Metildi CA, Kaushal S, Pu M, et al. Fluorescence guided surgery with a fluorophore conjugated antibody to carcinoembryonic antigen (CEA), that highlights the tumor, improves surgical resection and increases survival in orthotopic mouse models of human pancreatic cancer. Ann Surg Oncol. 2014;21(4):1405–11. doi: 10.1245/s10434-014-3495-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Tran Cao HS, Kaushal S, Metildi CA, et al. Tumor specific fluo rescence antibody imaging enables accurate staging laparoscopy in an orthotopic model of pancreatic cancer. Hepatogastroenterology. 2012;59(118):1994–9. doi: 10.5754/hge11836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kishimoto H, Urata Y, Tanaka N, Fujiwara T, Hoffman RM. Selective metastatic tumor labeling with green fluorescent protein and killing by systemic administration of telomerase dependent adenoviruses. Mol Cancer Ther. 2009;8(11):3001–8. doi: 10.1158/1535-7163.MCT-09-0556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kishimoto H, Zhao M, Hayashi K, et al. In vivo internal tumor illumination by telomerase dependent adenoviral GFP for precise surgical navigation. Proc Natl Acad Sci USA. 2009;106(34):14514–7. doi: 10.1073/pnas.0906388106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Aguilera TA, Olson ES, Timmers MM, Jiang T, Tsien RY. Systemic in vivo distribution of activatable cell penetrating peptides is superior to that of cell penetrating peptides. Integr Biol. 2009;1(5-6):371–81. doi: 10.1039/b904878b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jiang T, Olson ES, Nguyen QT, Roy M, Jennings PA, Tsien RY. Tumor imaging by means of proteolytic activation of cell pene trating peptides. Proc Natl Acad Sci USA. 2004;101(51):17867–72. doi: 10.1073/pnas.0408191101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Nguyen QT, Olson ES, Aguilera TA, et al. Surgery with molecular fluorescence imaging using activatable cell penetrating peptides decreases residual cancer and improves survival. Proc Natl Acad Sci USA. 2010;107(9):4317–22. doi: 10.1073/pnas.0910261107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Olson ES, Aguilera TA, Jiang T, et al. In vivo characterization of activatable cell penetrating peptides for targeting protease activity in cancer. Integr Biol. 2009;1(5-6):382–93. doi: 10.1039/b904890a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Savariar EN, Felsen CN, Nashi N, et al. Real time in vivo molecular detection of primary tumors and metastases with ra tiometric activatable cell penetrating peptides. Cancer Res. 2013;73(2):855–64. doi: 10.1158/0008-5472.CAN-12-2969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Technical Data Sheet, Fluorescent Imaging Agent MMPsense 680. http://www.perkinelmer.com/CMSResources/Images/44-739-69TCH_NEV10126-MMPSense680-TD.pdf.
- 18.Katz MH, Takimoto S, Spivack D, Moossa AR, Hoffman RM, Bouvet M. A novel red fluorescent protein orthotopic pancreatic cancer model for the preclinical evaluation of chemotherapeutics. J Surg Res. 2003;113(1):151–60. doi: 10.1016/s0022-4804(03)00234-8. [DOI] [PubMed] [Google Scholar]
- 19.Bouvet M, Wang J, Nardin SR, et al. Real time optical imaging of primary tumor growth and multiple metastatic events in a pancreatic cancer orthotopic model. Cancer Res. 2002;62(5):1534–40. [PubMed] [Google Scholar]
- 20.Fu X, Guadagni F, Hoffman RM. A metastatic nude mouse model of human pancreatic cancer constructed orthotopically with histologically intact patient specimens. Proc Natl Acad Sci USA. 1992;89(12):5645–9. doi: 10.1073/pnas.89.12.5645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Furukawa T, Kubota T, Watanabe M, Kitajima M, Hoffman RM. A novel “patient like” treatment model of human pancreatic cancer constructed using orthotopic transplantation of histologi cally intact human tumor tissue in nude mice. Cancer Res. 1993;53(13):3070–2. [PubMed] [Google Scholar]
- 22.Troyan SL, Kianzad V, Gibbs Strauss SL, et al. The FLARE intraoperative near infrared fluorescence imaging system: a first in human clinical trial in breast cancer sentinel lymph node mapping. Ann Surg Oncol. 2009;16(10):2943–52. doi: 10.1245/s10434-009-0594-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ishizawa T, Fukushima N, Shibahara J, et al. Real time identifi cation of liver cancers by using indocyanine green fluorescent imaging. Cancer. 2009;115(11):2491–504. doi: 10.1002/cncr.24291. [DOI] [PubMed] [Google Scholar]
- 24.Stummer W, Pichlmeier U, Meinel T, et al. Fluorescence guided surgery with 5 aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 2006;7(5):392–401. doi: 10.1016/S1470-2045(06)70665-9. [DOI] [PubMed] [Google Scholar]
- 25.Stiles BM, Bhargava A, Adusumilli PS, et al. The replication competent oncolytic herpes simplex mutant virus NV1066 is effective in the treatment of esophageal cancer. Surgery. 2003;134(2):357–64. doi: 10.1067/msy.2003.244. [DOI] [PubMed] [Google Scholar]
- 26.Kishimoto H, Aki R, Urata Y, et al. Tumor selective, adenoviral mediated GFP genetic labeling of human cancer in the live mouse reports future recurrence after resection. Cell Cycle. 2011;10(16):2737–41. doi: 10.4161/cc.10.16.16756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.van Dam GM, Themelis G, Crane LM, et al. Intraoperative tumor specific fluorescence imaging in ovarian cancer by folate receptor alpha targeting: first in human results. Nat Med. 2011;17(10):1315–9. doi: 10.1038/nm.2472. [DOI] [PubMed] [Google Scholar]
- 28.Bloomston M, Zervos EE, Rosemurgy AS., 2nd Matrix metallo proteinases and their role in pancreatic cancer: a review of preclinical studies and clinical trials. Ann Surg Oncol. 2002;9(7):668–74. doi: 10.1007/BF02574483. [DOI] [PubMed] [Google Scholar]
- 29.Bramhall SR. The matrix metalloproteinases and their inhibitors in pancreatic cancer. From molecular science to a clinical application. Int J Pancreatol. 1997;21(1):1–2. doi: 10.1007/BF02785914. [DOI] [PubMed] [Google Scholar]
- 30.Maatta M, Soini Y, Liakka A, Autio Harmainen H. Differential expression of matrix metalloproteinase (MMP) 2, MMP 9, and membrane type 1 MMP in hepatocellular and pancreatic adeno carcinoma: implications for tumor progression and clinical prognosis. Clin Cancer Res. 2000;6(7):2726–34. [PubMed] [Google Scholar]

