Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Feb 15, 2016; 8(2): 165-172
Published online Feb 15, 2016. doi: 10.4251/wjgo.v8.i2.165
Advancement in treatment and diagnosis of pancreatic cancer with radiopharmaceuticals
Yu-Ping Xu, Min Yang, Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu Province, China
Author contributions: Xu YP collected the material, prepared the manuscript and approved the final version; Yang M prepared the manuscript and approved the final version.
Supported by National Natural Science Foundation, Nos. 81171399, 51473071, 81101077, 21401084, 81401450 and 81472749; Jiangsu Province Foundation, Nos. BE2014609, BE2012622, BL2012031 and BM2012066; and Wuxi Foundation, No. CSZ0N1320.
Conflict-of-interest statement: The authors do not have any possible conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Min Yang, Professor, Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, 20 Qianrong Road, Wuxi 214063, Jiangsu Province, China. yangmin@jsinm.org
Telephone: +86-510-85508862 Fax: +86-510-85513113
Received: April 28, 2015
Peer-review started: May 5, 2015
First decision: July 21, 2015
Revised: November 26, 2015
Accepted: December 17, 2015
Article in press: December 18, 2015
Published online: February 15, 2016

Abstract

Pancreatic cancer (PC) is a major health problem. Conventional imaging modalities show limited accuracy for reliable assessment of the tumor. Recent researches suggest that molecular imaging techniques with tracers provide more biologically relevant information and are benefit for the diagnosis of the cancer. In addition, radiopharmaceuticals also play more important roles in treatment of the disease. This review summaries the advancement of the radiolabeled compounds in the theranostics of PC.

Key Words: Pancreatic cancer, Diagnosis, Therapy, Radiopharmaceuticals, Positron emission tomography

Core tip: This review describes the development of radiopharmaceuticals in diagnosis and therapy of pancreatic cancer. We herein discuss the role of the radiolabeled compounds in the preoperative diagnosis, staging, post-therapeutic monitoring, prognosis and the treatment of the disease.



INTRODUCTION

Pancreatic cancer (PC) is a major health problem due to low 5-year survival rate[1-3]. Surgery is the only curative treatment but less than 20% of cases are suitable to be respectable during diagnosis for the late onset of the symptoms[4-6]. Therefore, suitable diagnosis and staging is essential for management of the disease.

Computed tomography (CT), magnetic resonance imaging (MRI), and endoscopic ultrasound (EUS), etc., provide information regarding tumor size, location, and morphology, which can be used for initial staging, tumor evaluation and follow-up. However, it also remain suboptimal in the preoperative diagnosis and may hamper the treatment. The discrimination between benign and malignant lesions are still challenging with these methods[7,8].

Molecular imaging techniques are important tool capable of providing high sensitive non invasive and quantitative images of various cancer[9-11]. Radiopharmaceuticals is a key factor in the non-invasive molecular imaging technique which enables specific cellular and molecular processes to be functionally visualized. The development of molecular imaging agents target for specific biomarkers could provide more sensitive and specific cancer detection.

Meanwhile, a number of compounds labeled with therapy radionuclides have been employed for cancer treatment through intratumoral administration[12-15]. Compared with traditional high-dose external radiation, intratumoral administration delivers more radioactivity to the tumor than the normal structure[16].

Here, we review the pertinent literatures and the advancement in treatment and diagnosis of PC with radiopharmaceuticals was discussed.

SMALL MOLECULE TRACERS FOR TUMOR IMAGING
18F-fluorodeoxyglucose

Over the past decade, positron emission tomography (PET) is an important molecular imaging methods in various malignancies[17-20]. 18F-fluorodeoxyglucose (18F-FDG) is an analogue of glucose. After injected into the body, it is actively transported via glucose transporters (GLUT) into cells, then phosphorylated by hexokinase in the same pathway as glucose. However, unlike normal glucose, the reactions of 18F-FDG do not proceed further and the corresponding product remains in the cells[21,22]. Overexpression of GLUT-1 and hexokinase-II has been reported in PC[23]. In patients with PC, several studies have demonstrated that 18F-FDG PET/CT was an important key factor for in staging, detecting postoperative recurrence, and evaluating the response to treatment[24-28]. The recent typical researches and interest findings were listed in the follow.

Preoperative diagnosis: Ergul et al[29] compared the values of 18F-FDG PET/CT, multidetector row computed tomography (MDCT), MRI and EUS in the diagnosis and management of the tumor. It revealed that sensitivity of PET/CT were equal to EUS (100%) and higher than those of MDCT and MRI. Meanwhile, Specificity of MDCT was significantly lower than PET/CT. It suggested that 18F-FDG PET/CT is an useful imaging techniques for management of the disease[29].

Maximum standardized uptake value (SUVmax) reflects tumor aggressiveness as a marker of tumor glucose metabolism. Hu et al[30] found that the SUVmax of benign lesions significantly lower than that of malignant tumors (2.9 ± 2.0 vs 6.3 ± 2.4 respectively). A positive correlation between the SUVmax and Ki-67 was existed. It suggested that the SUVmax of 18F-FDG can be applied in the differential diagnosis and can also benefit for monitoring the proliferative status of PC[30].

Nagamachi et al[31] compared 18F-FDG PET/CT and 18F-FDG PET/MRI fusion image in diagnosing tumor. 18FDG-PET/MRI fusion image significantly improved accuracy. Results showed that this image technique was useful in differentiating diagnosis[31].

Zhang et al[32] reviewed 116 patients with pancreatic cystic tumors who had been treated with different imaging modalities. Compared with CT and EUS, PET had the best sensitivity, specificity and accuracy for detecting malignant cystic tumors[32].

When the conventional imaging modalities or biopsies are unavailable, PET also plays an important role in diagnosis of PC. Based on the 18F-FDG uptake pattern, sensitivity, specificity, positive predictive value, negative predictive value, and accuracy for FDG-PET/CT in differentiating benign and malignant lesions were all greater than 85% respectively[33].

Diagnostic performance of diffusion-weighted MRI and 18F-FDG PET/CT in the detection of pancreatic malignancy was also obtained by Wu et al[34]. When diagnosing patients with pancreatic malignancy, the sensitivity of PET/CT was higher than MRI but the specificity of the former was lower than the latter[34].

Staging: Wang et al[35] evaluate the value of 18F-FDG PET/CT on the pre-operative staging of the disease. The sensitivity and accuracy of the imaging modality to detect distant metastasis especially metastatic lymph nodes are significantly higher than those of MDCT. It showed that the extra staging information PET/CT provided could be helpful for screen of surgery[35].

18F-FDG PET/CT scans were performed at 17 patients in baseline and six weeks post-CRT. SUVmax significantly decreased during CRT (median pre- 8.0 and post- 3.6). It revealed that the baseline 18F-FDG PET was benefit for definition of the biological target volume for non-uniform dose prescriptions[36].

Topkan et al[37] evaluated the impact of 18F-FDG PET/CT restaging on management decisions and outcomes in patients with LAPC scheduled for concurrent CRT. According with PET/CT before therapy, these individuals were classified into non-metastatic (M0) and metastatic (M1) groups then received different treatment. Twenty-six point eight percent of distant metastases were detected via PET/CT not by conventional staging. Three additional regional lymph nodes were found by PET/CT restaging and the volumes of the tumors were larger than CT-defined borders. The initial management decisions of 26 patients were changed through PET/CT.

Median overall survival (OS) and progression-free survival (PFS) of M0 patients were greater than those of M1 patients. These findings conformed that PET/CT-based restaging may benefit for screening patients suitable for CRT[37].

Post-therapeutic monitoring: Picchio et al[38] evaluated the role of 18F-FDG PET/CT in screening patients with locally advanced PC for suitable treatment and monitoring the efficacy. Results showed that PET/CT play more important factors in designing the treatment plans for individual patient than conventional CT[38].

Kittaka et al[39] performed 18F-FDG PET in patients classified as responders and nonresponders before and after preoperative CRT. A pre-CRT SUV > 4.7 was seen in 15 (71%) of 21 responders and in 6 (32%) of 19 nonresponders. A regression index > 0.46 was observed in 15 (71%) responders and 5 (26%) nonresponders. It showed that the SUV based on FDG-PET/CT is a useful implement for predicting the response of treatment[39].

To study whether FDG-PET parameters can predict relatively long-term survival in patients, Chang et al[40] assess the effect of coregistered 18F-FDG PET in monitoring radiographically occult distant metastasis (DM) in patients with LAPC. Patients with a baseline standardized uptake value (SUV) < 3.5 and/or SUV decline ≥ 60% had significantly better OS and PFS than those having none, even after adjustment for all potential confounding variables. 18F-FDG PET can spare one-third of patients with occult DM from the potentially toxic therapy. 18F-FDG PET parameters including baseline SUV and SUV changes may serve as useful clinical markers for predicting the prognosis in LAPC patients[40].

Prognosis: Several prognostic factors for PC recurrence have previously been reported including tumor size, T stage, lymph node metastasis, tumor differentiation, lymphovascular invasion, involvement of the surgical margin, and serum carbohydrate antigen 19-9 (CA19-9) level. Yamamoto et al[41] evaluated whether preoperative 18F-FDG PET can predict the resectable PC. Among the patients, 34 cases with an SUVmax ≥ 6.0 developed recurrence within half year, however only 3 patients with an SUVmax < 6.0 exhibited early recurrence. The median OS time of patients with a SUVmax < 6.0 was significantly greater than those of patients with an SUVmax ≥ 6.0. Therefore, an SUVmax ≥ 6.0 maybe a significant predictor of recurrence of PC[41].

The histopathological grade of differentiation is also one of the significant prognostic factors in the disease, especially in the patients with unresectable PC. It was found that a significant correlation of SUVs and pathologic grades existed by 18F-FDG PET scans in 102 patients with histologically proven pancreas adenocarcinoma. It showed that 18F-FDG SUV is related with histologic grade and might be competitive predictor for survival[42].

Xi et al[43] determined 18F-FDG SUVmax in patients with PC at 1 h and 2 h post injection, and the retention index (RI) was defined as the percentage change between the values of two time points. It was found that there existed a significant positive correlation among RI and the tumor, node, and metastasis stage[43].

Shinoto et al[44] evaluated whether 18F-FDG PET can be used as an indicator of preoperative carbon-ion radiotherapy (CIRT) for PC patients. SUVmax was significantly correlated with DMFS and OS. The DMFS and OS in high-SUVmax group were significantly lower than those in low SUVmax group. 18F-FDG PET might be suitable for determining the indication of preoperative short-course CIRT for patients with resectable PC[44].

The prognostic role of 18F-FDG PET/CT in the prediction of PFS and chemotherapeutic response in patients with locally advanced or metastatic PC was also investigated by Moon et al[45] PFS of the low SUVmax (< 6.8) group was significantly longer than those of the high SUVmax (≥ 6.8) group. Resulted showed that SUVmax may be useful in independent predicting PFS of PC[45].

The prognostic value of volumetric parameters on preoperative 18F-FDG PET/CT was assessed. Results revealed that metabolic tumor volume and total lesion glycolysis are independent prognostic factors for predicting RFS and OS. Thus, 18F-FDG PET/CT can provide useful prognostic information for patients undergoing resection of PC with curative intent irrespective of neoadjuvant treatment[46].

Choi et al[47] evaluated the prognostic value of 18F-FDG PET in patients with resectable PC. The OS and DFS were significantly longer in the low SUVmax group than those of high SUVmax group[47].

Hwang et al[48] reviewed retrospectively the medical records of 165 patients with a diagnosis of PC. Patients were allocated to high (> 4.1) and low (≤ 4.1) SUV groups, and median survivals of these patients were 229 d and 610 d, respectively. Furthermore, SUVmax was found to be significantly related to survival in each stage. The median survival was also found to be significantly related to tumor size, site, serum level of CA19-9, distant metastasis, and type of treatment[48].

Epelbaum et al[49] evaluated the possibility of dynamic 18F-FDG PET/CT parameters used as an indicator in the tumor. The OS of patients with a high 18F-FDG influx was significantly lower than that of patients with a low 18F-FDG influx (5 and 6 mo vs 15 and 19 mo respectively). Quantitative 18F-FDG kinetic parameters in newly diagnosed PC correlated with the aggressiveness of disease[49].

Limitation: Although significant advances have been achieved in 18F-FDG PET diagnostic technologies, it has some limitations in detecting cancer. Due to increased glycolytic metabolism, 18F-FDG can also accumulate in the inflammatory cells[50]. As a result, it often yields false positive interpretations for PET. Kato et al[51] evaluated the efficacy of 18F-FDG PET/CT for the differential diagnosis in 47 individuals. It showed that differentiation is difficult by18F-FDG PET/CT due to overlapping in SUVmax between the two diseases. In addition, elevated serum glucose levels may decrease the uptake in tumors for competitive inhibition, which decreased the sensitivity of 18F-FDG PET in hyperglycemic patients[51]. Therefore, a numbers of other small molecule-based tracers were designed and developed for PET imaging of PC.

3-Deoxy-3-18F-fluorothymidine

A surrogate marker of DNA synthesis, 3-Deoxy-3-18F-fluorothymidine (18F-FLT), is another potential tracer for visualization of proliferating tissues[52-55]. For differentiation of pancreatic tumors, 18F-FLT PET showed a lower sensitivity but higher specificity than18F-FDG PET/CT (70% vs 91% and 75% vs 50% respectively)[56].

RADIOLABELED PEPTIDES FOR PC IMAGING

Peptides and their derivatives have been successfully developed for the tracer due to favorable characteristics such as low antigenicity, high specificity, fast clearance from blood and rapid tissue penetration. Radiolabelled receptor-binding peptides have become important radiopharmaceuticals for diagnosis and therapy in tumor[57-61]. Recently, a few radiolabeled peptides have been successfully used for PC imaging. It may be a promising imaging strategy for PC diagnosis and treatment.

Radiolabeled RGD analogs

Angiogenesis is necessary for tumor growth and metastasis, and the integrin αvβ3 receptor plays an important role in promoting, sustaining, and regulating the angiogenesis[62]. In vitro analysis demonstrated that integrin αvβ3 receptor was expressed in 60% of invasive pancreatic ductal carcinomas and would be an excellent target for the early detection of malignant PC[63]. Radiolabeled Arg-Gly-Asp (RGD) peptides are widely used as integrin αvβ3 receptor imaging agents in various types of tumors[63]. Yoshimoto et al[64] employed 111In-DOTA-c(RGDfK) for the early detection of PC in pancreatic carcinogenesis model. PC lesions as small as 3 mm in diameter as clearly were visualized after injection with the tracer. High tumor-to-normal pancreatic tissue radioactivity ratios were found by ARG analysis. There existed a significant relationship between the uptake of 111In-DOTA-c(RGDfK) and αvβ3-integrin expression. It also found that the false-positive rate of 111In-DOTA-c(RGDfK) was lower than that of 18F-FDG. It revealed that SPECT with 111In-DOTA-c(RGDfK) was benefit for the early accurate diagnosis of PC[64].

Trajkovic-Arsic et al[65] used 68Ga-NODAGA-RGD PET for αvβ3 integrin receptor in vivo imaging of spontaneous pancreatic ductal adenocarcinoma (PDAC) occurring in mice. It showed that αvβ3 integrin is expressed in human and murine PDAC and can be detected by molecular imaging technologies in PDAC. This strategy can further be exploited for identification of patients with αvβ3 integrin positive and application of αvβ3 targeted therapies[65].

Aung et al[66] performed a preclinical evaluation of 64Cu-RAFT-RGD in a clinically relevant orthotopic xenotransplantation model of PC. It was confirmed that the uptakes of 64Cu-RAFT-RGD in tumor was greater than those of normal tissues. Meanwhile, the tumor to background uptake ratios of the tracers was higher than those of 18F-FDG. It suggested that 64Cu-RAFT-RGD PET imaging might be useful in the diagnosis of PC[66].

Radiolabeled exendin-4 analogs

Insulinomas are the most frequent hormone-active tumors of the pancreas arising from pancreatic β cells[67-69]. Recently, glucagon-like peptide-1 receptor (GLP-1R) was found to be massively overexpressed in gut and lung neuroendocrine tumors, especially insulinomas. It provides an attractive target for the cancers[70-72].

Several radioligands towards GLP-1 receptor have been developed for GLP-1R-positive tumor imaging. At first, the analog of native receptor ligand, GLP-1(7–36) amide, was labeled with 123I and used for GLP-1R imaging. Although preclinical data showed 123I-GLP-1(7–36) amide possessed high accumulation in a RINm5F insulinoma tumor, the low stability of the peptide due to rapid degrading of GLP-1 by the enzyme dipeptidyl peptidase IV (DPIV) limited its clinical use[73].

Exendin-4 arised from the salivary gland of the gila monster lizard and has a 53% amino acid homology with GLP-1. It is more resistant to the DPIV digestion and binds with great affinity to the GLP-1R[73]. 111In- and 99mTc-labeled exendin-4 analogs have been evaluated for SPECT imaging of GLP-1R in rodents and humans, respectively, and promising results were obtained[74-77].

The sensitivity, imaging contrast and spatial resolution of PET was significantly higher than SPECT. In the past few years, exendin-4 analogs have been labeled with PET radionuclides for preclinical insulinomas imaging. Exendin-4 labeled with radio metals (68Ga, 64Cu) showed significant uptake in INS-1 insulinoma xenografts[78,79]. However, the substantial kidney uptake may limit their use in clinical practice due to high radiation exposure to the organs.

18F is the commonly used isotope. It has nearly optimal nuclear decay characteristics and chemical properties for peptide-based receptor imaging studies. In the past few years, exendin-4 analogs have been modified with either a C-terminal or N-terminal cysteine to allow site-specific labeling with a maleimide-selective prosthetic reagent, 18F-FBEM[80]. In vivo study showed that the INS-1 tumor uptake of 18F-FBEM-Cys40-exendin-4 was higher than that of 18F-FBEM-Cys0-exendin-4[80]. Based on the above results, other Cys40-exendin-4 analogs were developed for GLP-1R imaging[81,82].

In vitro receptor competitive binding study confirmed that the nine amino acid sequence at C-terminal of exendin-4 was not key for the biological activity or binding to the receptor. Meanwhile, serine is almost same as cysteine except for the difference in hydroxy and sulfhydryl group. Thus, replacing Ser39 with Cys39 could provide a unique site for attachment of a radiolabeling thiol-reactive group (such as 18F-FBEM) and may have less impact on the binding affinity of the peptide to the receptor[83]. Xu et al[83] synthesized a novel 18F-labeled exendin-4 analog, 18F-FBEM–Cys39-exendin-4. The tracer showed specific binding to GLP-1R and had better tumor to background radioactivity ratio and lower abdominal backgrounds than those of 18F-FBEM-Cys40-exendin-4[83]. It suggested that 18F-FBEM–Cys39-exendin-4 may be a potential probe for insulinomas imaging[83].

Despite the encouraging results, the tedious radiosynthesis would hinder the tracer to widespread use. Recently, a one-step simple procedure for preparing 18F-labeled peptides via chelating 18FAl with NOTA has been reported[84]. Xu et al[84] conjugated Cys39-exendin-4 with NOTA-MAL and obtained NOTA-MAL-Cys39-exendin-4. The compound was simply radiolabeled with 18FAl complex by one step in 30 min[85]. 18FAl-NOTA-MAL-Cys39-exendin-4 shows favorable characteristics for insulinoma imaging in mice bearing INS-1 tumor and may be translated to clinical studies[85].

THERAPY WITH RADIOPHARMACEUTICALS

Recently, only few patients have resectable disease. High-dose external radiation to the pancreas may damage the surrounding organs. The intratumoral administration of radiopharmaceuticals delivers the maximum amount of radioactivity to the tumor with limiting side effects[86-88].

During the past several decades, implantation of radioactive isotopes for the treatment has been used. Some basic research indicated that 125I seed with continuous low dose rate irradiation may be beneficial to PC[86-88]. Zhongmin et al[89] implanted 125I seeds into PC under CT guidance in thirty-one patients with inoperable PC. It was found that overall responding rate was greater than 60% and median survival time was about 10 mo[89]. The efficacy of intraoperative ultrasound-guided implantation of 125I seeds was also assessed for the treatment of unresectable PC by Wang et al[90]. Most of the patients achieved favorable pain relief. These studies revealed that 125I seeds implantation was benefit for the treatment of PC patients[90].

Phosphorus 32 is another ideal unsealed therapeutic radionuclide. Colloid 32P has been applied for the treatment of intracavitary malignancies[91-93]. Preclinical study showed that 32P-chromic phosphate colloid (32P-CP) through intratumoral injection mainly accumulated in the BXPC-3 human tumor and retained for a long time[94]. The safety and efficacy of the therapy to PC was also confirmed[94].

Poly (L-lactic acid) (PLLA) has been widely used as a drug delivery system due to excellent biocompatibility and biodegradability[95-99]. 32P-CP-PLLA microparticle was successfully prepared and used for brachytherapy in several tumor models[95-99]. Yang et al[100] evaluated its biodistribution, bioelimination, and therapeutic effect in mice bearing BxPC-3 human PC. Results showed that 32P-CP-PLLA was mostly remained at the tumor (> 95% ID) and almost no radioactivity excretion was observed in urine and feces. As compared, some radioactivity (over 5% ID) of 32P-CP colloid was found in the normal organs[100]. Meanwhile, the tumor volumes was significantly decreased after treatment with 32P-CP-PLLA microparticle[100]. It showed that 32P-CP-PLLA microparticle might be benefit for the management of PC[100].

CONCLUSION

Radiopharmaceuticals are favorable diagnostic and therapy facility for PC. The development of new tracers may be beneficial to personalized management of the disease.

Footnotes

P- Reviewer: Yalcin S S- Editor: Ji FF L- Editor: A E- Editor: Li D

References
1.  Hidalgo M. Pancreatic cancer. N Engl J Med. 2010;362:1605-1617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2078]  [Cited by in F6Publishing: 2117]  [Article Influence: 151.2]  [Reference Citation Analysis (2)]
2.  Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013;63:11-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9215]  [Cited by in F6Publishing: 9719]  [Article Influence: 883.5]  [Reference Citation Analysis (3)]
3.  Teague A, Lim KH, Wang-Gillam A. Advanced pancreatic adenocarcinoma: a review of current treatment strategies and developing therapies. Ther Adv Med Oncol. 2015;7:68-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 109]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
4.  Puleo F, Maréchal R, Demetter P, Bali MA, Calomme A, Closset J, Bachet JB, Deviere J, Van Laethem JL. New challenges in perioperative management of pancreatic cancer. World J Gastroenterol. 2015;21:2281-2293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
5.  Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet. 2004;363:1049-1057.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Mendieta Zerón H, García Flores JR, Romero Prieto ML. Limitations in improving detection of pancreatic adenocarcinoma. Future Oncol. 2009;5:657-668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
7.  Kinney T. Evidence-based imaging of pancreatic malignancies. Surg Clin North Am. 2010;90:235-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 59]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
8.  Katz MH, Savides TJ, Moossa AR, Bouvet M. An evidence-based approach to the diagnosis and staging of pancreatic cancer. Pancreatology. 2005;5:576-590.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Jung KH, Lee KH. Molecular imaging in the era of personalized medicine. J Pathol Transl Med. 2015;49:5-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
10.  Cunha L, Szigeti K, Mathé D, Metello LF. The role of molecular imaging in modern drug development. Drug Discov Today. 2014;19:936-948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
11.  Willmann JK, van Bruggen N, Dinkelborg LM, Gambhir SS. Molecular imaging in drug development. Nat Rev Drug Discov. 2008;7:591-607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 836]  [Cited by in F6Publishing: 736]  [Article Influence: 46.0]  [Reference Citation Analysis (0)]
12.  Bult W, Kroeze SG, Elschot M, Seevinck PR, Beekman FJ, de Jong HW, Uges DR, Kosterink JG, Luijten PR, Hennink WE. Intratumoral administration of holmium-166 acetylacetonate microspheres: antitumor efficacy and feasibility of multimodality imaging in renal cancer. PLoS One. 2013;8:e52178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
13.  Phillips WT, Bao A, Brenner AJ, Goins BA. Image-guided interventional therapy for cancer with radiotherapeutic nanoparticles. Adv Drug Deliv Rev. 2014;76:39-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 66]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
14.  Li CC, Chi JL, Ma Y, Li JH, Xia CQ, Li L, Chen Z, Chen XL. Interventional therapy for human breast cancer in nude mice with 131I gelatin microspheres (¹³¹I-GMSs) following intratumoral injection. Radiat Oncol. 2014;9:144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
15.  Chi JL, Li CC, Xia CQ, Li L, Ma Y, Li JH, Chen Z, Chen XL. Effect of (131)I gelatin microspheres on hepatocellular carcinoma in nude mice and its distribution after intratumoral injection. Radiat Res. 2014;181:416-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
16.  McCready VR, Cornes P. The potential of intratumoural unsealed radioactive source therapy. Eur J Nucl Med. 2001;28:567-569.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Farwell MD, Pryma DA, Mankoff DA. PET/CT imaging in cancer: current applications and future directions. Cancer. 2014;120:3433-3445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 133]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
18.  Gallamini A, Zwarthoed C, Borra A. Positron Emission Tomography (PET) in Oncology. Cancers (Basel). 2014;6:1821-1889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in F6Publishing: 198]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
19.  Kartalis N, Mucelli RM, Sundin A. Recent developments in imaging of pancreatic neuroendocrine tumors. Ann Gastroenterol. 2015;28:193-202.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Rijkers AP, Valkema R, Duivenvoorden HJ, van Eijck CH. Usefulness of F-18-fluorodeoxyglucose positron emission tomography to confirm suspected pancreatic cancer: a meta-analysis. Eur J Surg Oncol. 2014;40:794-804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 85]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
21.  Hong H, Zhang Y, Sun J, Cai W. Positron emission tomography imaging of prostate cancer. Amino Acids. 2010;39:11-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 46]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
22.  Macheda ML, Rogers S, Best JD. Molecular and cellular regulation of glucose transporter (GLUT) proteins in cancer. J Cell Physiol. 2005;202:654-662.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Basturk O, Singh R, Kaygusuz E, Balci S, Dursun N, Culhaci N, Adsay NV. GLUT-1 expression in pancreatic neoplasia: implications in pathogenesis, diagnosis, and prognosis. Pancreas. 2011;40:187-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 58]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
24.  Wang Z, Chen JQ, Liu JL, Qin XG, Huang Y. FDG-PET in diagnosis, staging and prognosis of pancreatic carcinoma: a meta-analysis. World J Gastroenterol. 2013;19:4808-4817.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 93]  [Cited by in F6Publishing: 81]  [Article Influence: 7.4]  [Reference Citation Analysis (1)]
25.  Murakami K. FDG-PET for hepatobiliary and pancreatic cancer: Advances and current limitations. World J Clin Oncol. 2011;2:229-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 25]  [Cited by in F6Publishing: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
26.  Rao M, Chen Y, Zhu Y, Huang Z, Zhang L. Primary pancreatic choriocarcinoma revealed on FDG PET/CT. Clin Nucl Med. 2015;40:76-78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
27.  Yoshioka M, Uchinami H, Watanabe G, Sato T, Shibata S, Kume M, Ishiyama K, Takahashi S, Hashimoto M, Yamamoto Y. F-18 fluorodeoxyglucose positron emission tomography for differential diagnosis of pancreatic tumors. Springerplus. 2015;4:154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
28.  Dibble EH, Karantanis D, Mercier G, Peller PJ, Kachnic LA, Subramaniam RM. PET/CT of cancer patients: part 1, pancreatic neoplasms. AJR Am J Roentgenol. 2012;199:952-967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 86]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
29.  Ergul N, Gundogan C, Tozlu M, Toprak H, Kadıoglu H, Aydin M, Cermik TF. Role of (18)F-fluorodeoxyglucose positron emission tomography/computed tomography in diagnosis and management of pancreatic cancer; comparison with multidetector row computed tomography, magnetic resonance imaging and endoscopic ultrasonography. Rev Esp Med Nucl Imagen Mol. 2014;33:159-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 11]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
30.  Hu SL, Yang ZY, Zhou ZR, Yu XJ, Ping B, Zhang YJ. Role of SUV(max) obtained by 18F-FDG PET/CT in patients with a solitary pancreatic lesion: predicting malignant potential and proliferation. Nucl Med Commun. 2013;34:533-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
31.  Nagamachi S, Nishii R, Wakamatsu H, Mizutani Y, Kiyohara S, Fujita S, Futami S, Sakae T, Furukoji E, Tamura S. The usefulness of (18)F-FDG PET/MRI fusion image in diagnosing pancreatic tumor: comparison with (18)F-FDG PET/CT. Ann Nucl Med. 2013;27:554-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
32.  Zhang Y, Frampton AE, Martin JL, Kyriakides C, Bong JJ, Habib NA, Vlavianos P, Jiao LR. 18F-fluorodeoxyglucose positron emission tomography in management of pancreatic cystic tumors. Nucl Med Biol. 2012;39:982-985.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
33.  Santhosh S, Mittal BR, Bhasin D, Srinivasan R, Rana S, Das A, Nada R, Bhattacharya A, Gupta R, Kapoor R. Role of (18)F-fluorodeoxyglucose positron emission tomography/computed tomography in the characterization of pancreatic masses: experience from tropics. J Gastroenterol Hepatol. 2013;28:255-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
34.  Wu LM, Hu JN, Hua J, Liu MJ, Chen J, Xu JR. Diagnostic value of diffusion-weighted magnetic resonance imaging compared with fluorodeoxyglucose positron emission tomography/computed tomography for pancreatic malignancy: a meta-analysis using a hierarchical regression model. J Gastroenterol Hepatol. 2012;27:1027-1035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
35.  Wang XY, Yang F, Jin C, Guan YH, Zhang HW, Fu DL. The value of 18F-FDG positron emission tomography/computed tomography on the pre-operative staging and the management of patients with pancreatic carcinoma. Hepatogastroenterology. 2014;61:2102-2109.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Wilson JM, Mukherjee S, Chu KY, Brunner TB, Partridge M, Hawkins M. Challenges in using 18F-fluorodeoxyglucose-PET-CT to define a biological radiotherapy boost volume in locally advanced pancreatic cancer. Radiat Oncol. 2014;9:146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
37.  Topkan E, Parlak C, Yapar AF. FDG-PET/CT-based restaging may alter initial management decisions and clinical outcomes in patients with locally advanced pancreatic carcinoma planned to undergo chemoradiotherapy. Cancer Imaging. 2013;13:423-428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
38.  Picchio M, Giovannini E, Passoni P, Busnardo E, Landoni C, Giovacchini G, Bettinardi V, Crivellaro C, Gianolli L, Di Muzio N. Role of PET/CT in the clinical management of locally advanced pancreatic cancer. Tumori. 2012;98:643-651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
39.  Kittaka H, Takahashi H, Ohigashi H, Gotoh K, Yamada T, Tomita Y, Hasegawa Y, Yano M, Ishikawa O. Role of (18)F-fluorodeoxyglucose positron emission tomography/computed tomography in predicting the pathologic response to preoperative chemoradiation therapy in patients with resectable T3 pancreatic cancer. World J Surg. 2013;37:169-178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
40.  Chang JS, Choi SH, Lee Y, Kim KH, Park JY, Song SY, Cho A, Yun M, Lee JD, Seong J. Clinical usefulness of 18F-fluorodeoxyglucose-positron emission tomography in patients with locally advanced pancreatic cancer planned to undergo concurrent chemoradiation therapy. Int J Radiat Oncol Biol Phys. 2014;90:126-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
41.  Yamamoto T, Sugiura T, Mizuno T, Okamura Y, Aramaki T, Endo M, Uesaka K. Preoperative FDG-PET predicts early recurrence and a poor prognosis after resection of pancreatic adenocarcinoma. Ann Surg Oncol. 2015;22:677-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 55]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
42.  Ahn SJ, Park MS, Lee JD, Kang WJ. Correlation between 18F-fluorodeoxyglucose positron emission tomography and pathologic differentiation in pancreatic cancer. Ann Nucl Med. 2014;28:430-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
43.  Xi Y, Guo R, Hu J, Zhang M, Zhang X, Li B. 18F-fluoro-2-deoxy-D-glucose retention index as a prognostic parameter in patients with pancreatic cancer. Nucl Med Commun. 2014;35:1112-1118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
44.  Shinoto M, Yamada S, Yoshikawa K, Yasuda S, Shioyama Y, Honda H, Kamada T, Tsujii H. Usefulness of 18F-fluorodeoxyglucose positron emission tomography as predictor of distant metastasis in preoperative carbon-ion radiotherapy for pancreatic cancer. Anticancer Res. 2013;33:5579-5584.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Moon SY, Joo KR, So YR, Lim JU, Cha JM, Shin HP, Yang YJ. Predictive value of maximum standardized uptake value (SUVmax) on 18F-FDG PET/CT in patients with locally advanced or metastatic pancreatic cancer. Clin Nucl Med. 2013;38:778-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
46.  Lee JW, Kang CM, Choi HJ, Lee WJ, Song SY, Lee JH, Lee JD. Prognostic Value of Metabolic Tumor Volume and Total Lesion Glycolysis on Preoperative 18F-FDG PET/CT in Patients with Pancreatic Cancer. J Nucl Med. 2014;55:898-904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 151]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
47.  Choi HJ, Kang CM, Lee WJ, Song SY, Cho A, Yun M, Lee JD, Kim JH, Lee JH. Prognostic value of 18F-fluorodeoxyglucose positron emission tomography in patients with resectable pancreatic cancer. Yonsei Med J. 2013;54:1377-1383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
48.  Hwang JP, Lim I, Chang KJ, Kim BI, Choi CW, Lim SM. Prognostic value of SUVmax measured by Fluorine-18 Fluorodeoxyglucose Positron Emission Tomography with Computed Tomography in Patients with Pancreatic Cancer. Nucl Med Mol Imaging. 2012;46:207-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
49.  Epelbaum R, Frenkel A, Haddad R, Sikorski N, Strauss LG, Israel O, Dimitrakopoulou-Strauss A. Tumor aggressiveness and patient outcome in cancer of the pancreas assessed by dynamic 18F-FDG PET/CT. J Nucl Med. 2013;54:12-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
50.  Buck AC, Schirrmeister HH, Guhlmann CA, Diederichs CG, Shen C, Buchmann I, Kotzerke J, Birk D, Mattfeldt T, Reske SN. Ki-67 immunostaining in pancreatic cancer and chronic active pancreatitis: does in vivo FDG uptake correlate with proliferative activity? J Nucl Med. 2001;42:721-725.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Kato K, Nihashi T, Ikeda M, Abe S, Iwano S, Itoh S, Shimamoto K, Naganawa S. Limited efficacy of (18)F-FDG PET/CT for differentiation between metastasis-free pancreatic cancer and mass-forming pancreatitis. Clin Nucl Med. 2013;38:417-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
52.  van Waarde A, Cobben DC, Suurmeijer AJ, Maas B, Vaalburg W, de Vries EF, Jager PL, Hoekstra HJ, Elsinga PH. Selectivity of 18F-FLT and 18F-FDG for differentiating tumor from inflammation in a rodent model. J Nucl Med. 2004;45:695-700.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Barwick T, Bencherif B, Mountz JM, Avril N. Molecular PET and PET/CT imaging of tumour cell proliferation using F-18 fluoro-L-thymidine: a comprehensive evaluation. Nucl Med Commun. 2009;30:908-917.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 66]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
54.  Lütje S, Boerman OC, van Rij CM, Sedelaar M, Helfrich W, Oyen WJ, Mulders PF. Prospects in radionuclide imaging of prostate cancer. Prostate. 2012;72:1262-1272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
55.  Challapalli A, Barwick T, Pearson RA, Merchant S, Mauri F, Howell EC, Sumpter K, Maxwell RJ, Aboagye EO, Sharma R. 3’-Deoxy-3’-18F-fluorothymidine positron emission tomography as an early predictor of disease progression in patients with advanced and metastatic pancreatic cancer. Eur J Nucl Med Mol Imaging. 2015;42:831-840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
56.  Herrmann K, Erkan M, Dobritz M, Schuster T, Siveke JT, Beer AJ, Wester HJ, Schmid RM, Friess H, Schwaiger M. Comparison of 3‘-deoxy-3‘-[18F]fluorothymidine positron emission tomography (FLT PET) and FDG PET/CT for the detection and characterization of pancreatic tumours. Eur J Nucl Med Mol Imaging. 2012;39:846-851.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 47]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
57.  Graham MM, Menda Y. Radiopeptide imaging and therapy in the United States. J Nucl Med. 2011;52 Suppl 2:56S-63S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 64]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
58.  Koopmans KP, Glaudemans AW. Rationale for the use of radiolabelled peptides in diagnosis and therapy. Eur J Nucl Med Mol Imaging. 2012;39 Suppl 1:S4-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
59.  Pan D, Yan Y, Yang R, Xu YP, Chen F, Wang L, Luo S, Yang M. PET imaging of prostate tumors with 18F-Al-NOTA-MATBBN. Contrast Media Mol Imaging. 2014;9:342-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 25]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
60.  Xu Y, Pan D, Zhu C, Xu Q, Wang L, Chen F, Yang R, Luo S, Yang M, Yan Y. Pilot study of a novel (18)F-labeled FSHR probe for tumor imaging. Mol Imaging Biol. 2014;16:578-585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
61.  Pan D, Xu YP, Yang RH, Wang L, Chen F, Luo S, Yang M, Yan Y. A new (68)Ga-labeled BBN peptide with a hydrophilic linker for GRPR-targeted tumor imaging. Amino Acids. 2014;46:1481-1489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
62.  Wan W, Guo N, Pan D, Yu C, Weng Y, Luo S, Ding H, Xu Y, Wang L, Lang L. First experience of 18F-alfatide in lung cancer patients using a new lyophilized kit for rapid radiofluorination. J Nucl Med. 2013;54:691-698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 143]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
63.  Hosotani R, Kawaguchi M, Masui T, Koshiba T, Ida J, Fujimoto K, Wada M, Doi R, Imamura M. Expression of integrin alphaVbeta3 in pancreatic carcinoma: relation to MMP-2 activation and lymph node metastasis. Pancreas. 2002;25:e30-e35.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Yoshimoto M, Hayakawa T, Mutoh M, Imai T, Tsuda K, Kimura S, Umeda IO, Fujii H, Wakabayashi K. In vivo SPECT imaging with 111In-DOTA-c(RGDfK) to detect early pancreatic cancer in a hamster pancreatic carcinogenesis model. J Nucl Med. 2012;53:765-771.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
65.  Trajkovic-Arsic M, Mohajerani P, Sarantopoulos A, Kalideris E, Steiger K, Esposito I, Ma X, Themelis G, Burton N, Michalski CW. Multimodal molecular imaging of integrin αvβ3 for in vivo detection of pancreatic cancer. J Nucl Med. 2014;55:446-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
66.  Aung W, Jin ZH, Furukawa T, Claron M, Boturyn D, Sogawa C, Tsuji AB, Wakizaka H, Fukumura T, Fujibayashi Y. Micro-positron emission tomography/contrast-enhanced computed tomography imaging of orthotopic pancreatic tumor-bearing mice using the αvβ3 integrin tracer 64Cu-labeled cyclam-RAFT-c(-RGDfK-)4. Mol Imaging. 2013;12:376-387.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Chatziioannou A, Kehagias D, Mourikis D, Antoniou A, Limouris G, Kaponis A, Kavatzas N, Tseleni S, Vlachos L. Imaging and localization of pancreatic insulinomas. Clin Imaging. 2001;25:275-283.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Grant CS. Insulinoma. Best Pract Res Clin Gastroenterol. 2005;19:783-798.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Wild D, Christ E, Caplin ME, Kurzawinski TR, Forrer F, Brändle M, Seufert J, Weber WA, Bomanji J, Perren A. Glucagon-like peptide-1 versus somatostatin receptor targeting reveals 2 distinct forms of malignant insulinomas. J Nucl Med. 2011;52:1073-1078.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 107]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
70.  Reubi JC, Maecke HR. Peptide-based probes for cancer imaging. J Nucl Med. 2008;49:1735-1738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 204]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
71.  Reubi JC, Waser B. Concomitant expression of several peptide receptors in neuroendocrine tumours: molecular basis for in vivo multireceptor tumour targeting. Eur J Nucl Med Mol Imaging. 2003;30:781-793.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Körner M, Stöckli M, Waser B, Reubi JC. GLP-1 receptor expression in human tumors and human normal tissues: potential for in vivo targeting. J Nucl Med. 2007;48:736-743.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Gotthardt M, Fischer M, Naeher I, Holz JB, Jungclas H, Fritsch HW, Béhé M, Göke B, Joseph K, Behr TM. Use of the incretin hormone glucagon-like peptide-1 (GLP-1) for the detection of insulinomas: initial experimental results. Eur J Nucl Med Mol Imaging. 2002;29:597-606.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Wild D, Béhé M, Wicki A, Storch D, Waser B, Gotthardt M, Keil B, Christofori G, Reubi JC, Mäcke HR. [Lys40(Ahx-DTPA-111In)NH2]exendin-4, a very promising ligand for glucagon-like peptide-1 (GLP-1) receptor targeting. J Nucl Med. 2006;47:2025-2033.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Wicki A, Wild D, Storch D, Seemayer C, Gotthardt M, Behe M, Kneifel S, Mihatsch MJ, Reubi JC, Mäcke HR. [Lys40(Ahx-DTPA-111In)NH2]-Exendin-4 is a highly efficient radiotherapeutic for glucagon-like peptide-1 receptor-targeted therapy for insulinoma. Clin Cancer Res. 2007;13:3696-3705.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Christ E, Wild D, Forrer F, Brändle M, Sahli R, Clerici T, Gloor B, Martius F, Maecke H, Reubi JC. Glucagon-like peptide-1 receptor imaging for localization of insulinomas. J Clin Endocrinol Metab. 2009;94:4398-4405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 196]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
77.  Sowa-Staszczak A, Pach D, Mikołajczak R, Mäcke H, Jabrocka-Hybel A, Stefańska A, Tomaszuk M, Janota B, Gilis-Januszewska A, Małecki M. Glucagon-like peptide-1 receptor imaging with [Lys40(Ahx-HYNIC- 99mTc/EDDA)NH2]-exendin-4 for the detection of insulinoma. Eur J Nucl Med Mol Imaging. 2013;40:524-531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 80]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
78.  Luo Y, Yu M, Pan Q, Wu W, Zhang T, Kiesewetter DO, Zhu Z, Li F, Chen X, Zhao Y. 68Ga-NOTA-exendin-4 PET/CT in detection of occult insulinoma and evaluation of physiological uptake. Eur J Nucl Med Mol Imaging. 2015;42:531-532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
79.  Wu Z, Liu S, Nair I, Omori K, Scott S, Todorov I, Shively JE, Conti PS, Li Z, Kandeel F. (64)Cu labeled sarcophagine exendin-4 for microPET imaging of glucagon like peptide-1 receptor expression. Theranostics. 2014;4:770-777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
80.  Kiesewetter DO, Gao H, Ma Y, Niu G, Quan Q, Guo N, Chen X. 18F-radiolabeled analogs of exendin-4 for PET imaging of GLP-1 in insulinoma. Eur J Nucl Med Mol Imaging. 2012;39:463-473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 86]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
81.  Wu Z, Liu S, Hassink M, Nair I, Park R, Li L, Todorov I, Fox JM, Li Z, Shively JE. Development and evaluation of 18F-TTCO-Cys40-Exendin-4: a PET probe for imaging transplanted islets. J Nucl Med. 2013;54:244-251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 90]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
82.  Yue X, Kiesewetter DO, Guo J, Sun Z, Zhang X, Zhu L, Niu G, Ma Y, Lang L, Chen X. Development of a new thiol site-specific prosthetic group and its conjugation with [Cys(40)]-exendin-4 for in vivo targeting of insulinomas. Bioconjug Chem. 2013;24:1191-1200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
83.  Xu Y, Pan D, Xu Q, Zhu C, Wang L, Chen F, Yang R, Luo S, Yang M. Insulinoma imaging with glucagon-like peptide-1 receptor targeting probe (18)F-FBEM-Cys (39)-exendin-4. J Cancer Res Clin Oncol. 2014;140:1479-1488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
84.  Xu Q, Zhu C, Xu Y, Pan D, Liu P, Yang R, Wang L, Chen F, Sun X, Luo S. Preliminary evaluation of [(18)F]AlF-NOTA-MAL-Cys(39)-exendin-4 in insulinoma with PET. J Drug Target. 2015;23:813-820.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
85.  Kiesewetter DO, Guo N, Guo J, Gao H, Zhu L, Ma Y, Niu G, Chen X. Evaluation of an [(18)F]AlF-NOTA Analog of Exendin-4 for Imaging of GLP-1 Receptor in Insulinoma. Theranostics. 2012;2:999-1009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 54]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
86.  Liu K, Ji B, Zhang W, Liu S, Wang Y, Liu Y. Comparison of iodine-125 seed implantation and pancreaticoduodenectomy in the treatment of pancreatic cancer. Int J Med Sci. 2014;11:893-896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
87.  Ma JX, Jin ZD, Si PR, Liu Y, Lu Z, Wu HY, Pan X, Wang LW, Gong YF, Gao J. Continuous and low-energy 125I seed irradiation changes DNA methyltransferases expression patterns and inhibits pancreatic cancer tumor growth. J Exp Clin Cancer Res. 2011;30:35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 36]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
88.  Huang ZM, Pan CC, Wu PH, Zhao M, Li W, Huang ZL, Yi RY. Efficacy of minimally invasive therapies on unresectable pancreatic cancer. Chin J Cancer. 2013;32:334-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
89.  Zhongmin W, Yu L, Fenju L, Kemin C, Gang H. Clinical efficacy of CT-guided iodine-125 seed implantation therapy in patients with advanced pancreatic cancer. Eur Radiol. 2010;20:1786-1791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 67]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
90.  Wang H, Wang J, Jiang Y, Li J, Tian S, Ran W, Xiu D, Gao Y. The investigation of 125I seed implantation as a salvage modality for unresectable pancreatic carcinoma. J Exp Clin Cancer Res. 2013;32:106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
91.  Montijo IJ, Khurana V, Alazmi WM, Order SE, Barkin JS. Vascular pancreatic gastric fistula: a complication of colloidal 32P injection for nonresectable pancreatic cancer. Dig Dis Sci. 2003;48:1758-1759.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Gao W, Liu L, Teng GJ, Feng GS, Tong GS, Gao NR. Internal radiotherapy using 32P colloid or microsphere for refractory solid tumors. Ann Nucl Med. 2008;22:653-660.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
93.  Rosemurgy A, Luzardo G, Cooper J, Bowers C, Zervos E, Bloomston M, Al-Saadi S, Carroll R, Chheda H, Carey L. 32P as an adjunct to standard therapy for locally advanced unresectable pancreatic cancer: a randomized trial. J Gastrointest Surg. 2008;12:682-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
94.  Gao W, Liu L, Liu ZY, Wang Y, Jiang B, Liu XN. Intratumoral injection of 32P-chromic phosphate in the treatment of implanted pancreatic carcinoma. Cancer Biother Radiopharm. 2010;25:215-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
95.  Sun L, Zhu X, Xu L, Wang Z, Shao G, Zhao J. Antitumor effects of (32)P-chromic-poly (L-lactide) brachytherapy in nude mice with human prostate cancer. Oncol Lett. 2013;6:687-692.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Zhao J, Du G, Su Y, Shao G, Wang Z, Xu L. Preliminary study of the biodegradation and the correlation between in vitro and in vivo release of (32)P-chromic phosphate-poly(L-lactide) seeds. Cancer Biother Radiopharm. 2013;28:703-708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
97.  Liu L, Huang P, Nie Q, Qi B, Wu Q, Gao H, Yang Z, Chen D. Safety evaluation of 32P-chromic phosphate-poly L lactic acid particles interstitially implanted into livers of Beagle dogs. Cancer Biother Radiopharm. 2012;27:156-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
98.  Xu Yp, Yang M, Pan Dh, Wang Lz, Liu L, Huang P, Shao G. Bioevaluation study of 32P-CP-PLLA particle brachytherapy in a rabbit VX2 lung tumor model. Appl Radiat Isot. 2012;70:583-588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
99.  He XJ, Jia RP, Shao GQ, Xu LW, Wang ZZ, Huang PL, Wu JP, Wang J. [Implantation brachytherapy with 32P-chromic phosphate-poly (L-lactide) delayed-release particles for prostate cancer in nude mice]. Zhonghua Nankexue. 2010;16:872-876.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Yang M, Xu YP, Pan DH, Wang LZ, Luo SN, Shao GQ, Liu L, Huang PL. Bioevaluation of a novel [32P]-CP-PLLA microparticle for pancreatic cancer treatment. Drug Dev Res. 2010;71:364-370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]