Contrast‑enhanced computerized tomography combined with a targeted nanoparticle contrast agent for screening for early‑phase non‑small cell lung cancer

  • Authors:
    • Ninglu Yuan
    • Xiaohe Zhang
    • Yonghui Cao
    • Xiaojie Jiang
    • Si Zhao
    • Yingying Feng
    • Yimeng Fan
    • Zhitao Lu
    • Hongmei Gao
  • View Affiliations

  • Published online on: September 19, 2017     https://doi.org/10.3892/etm.2017.5140
  • Pages: 5063-5068
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Non‑small cell lung cancer (NSCLC) is a major cause of morbidity and mortality, and patients with NSCLC are frequently diagnosed at an advanced stage. This is primarily due to a lack of advanced and sensitive protocols for the detection of early stage NSCLC. Therefore, methods for the accurate diagnosis of early stage NSCLC are urgently required to improve survival rates. The present study investigated the use of contrast‑enhanced computerized tomography (CECT) combined with a targeted nanoparticle contrast agent (TNCA) to diagnose early‑stage NSCLC in a mice xenograft model. The TNCA used was lenvatinib, a multi‑target tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor 1‑3, fibroblast growth factor receptor 1‑4, platelet‑derived growth factor receptor β, proto‑oncogene tyrosine‑protein kinase receptor Ret and mast/stem cell growth factor receptor Kit. Xenograft NSCLC mice were established and used to analyze the efficacy of CECT‑TNCA compared with CT scanning alone. The TNCA was inhaled with the use of an atomizer. The results demonstrated that CECT‑TNCA improved the sensitivity of the diagnosis of early stage NSCLC. In addition, imaging using the TNCA enabled the visualization of nodules in the lung in mice with early stage NSCLC. In addition, lung nodule signal enhancement was increased in CECT‑TNCA compared with CT, suggesting a high accurate accumulation of the TNCA in tumor nodules. Mice diagnosed with early stage NSCLC exhibited a higher eradication rate of NSCLC after treatment with cisplatin compared with mice with advanced stage NSCLC. These data indicate that the sensitivity and accuracy of CT imaging for the diagnosis of early stage NSCLC was improved through combination with the liposome‑encapsulated TNCA.

Introduction

Non-small cell lung cancer (NSCLC) is a major cause of morbidity and mortality (1,2). DeCotiis et al (3) reported that NSCLC is typically an aggressive cancer. The occurrence of NSCLC has increased in developing countries due to rising industrial pollution (4,5). The types of NSCLC include adenocarcinoma, large cell carcinoma and squamous cell carcinoma, which together account for ~80% of all lung cancer cases (6,7). Despite advancements in the therapeutic regimens for lung cancer, the survival rate of patients with NSCLC remains poor (810). This is due to NSCLC typically being diagnosed at an advanced stage (11,12). In addition, the aggressiveness of NSCLC contributes to tumor recurrence and poor survival rates for patients (13,14). These reports suggest that the early diagnosis of NSCLC would be a best way to improve the survival of patients with NSCLC.

The majority of patients with NSCLC have an advanced stage tumor at the time of diagnosis (15). In order to increase the efficacy of anticancer drugs and decrease mortality rates, the focus of lung cancer management has shifted to early diagnosis and personalized anticancer therapy (16,17). Hence, different diagnostics tools for patients with NSCLC have been explored (11,18). Clinically, computerized tomography (CT) scanning is frequently used to diagnose NSCLC (19). Subsequently, histopathology is applied to confirm the final diagnosis (20). Aberle et al (21) reported that CT screening had altered the landscape of lung-cancer screening and decreased lung cancer-associated mortalities by 20%, indicating the potential efficacy of screening for early stage lung cancer using CT imaging. However, CT used alone is not sensitive enough for the detection of early stage NSCLC (22,23). Seigneurin et al indicated that the presence of positron emission tomography (PET) in the work-up protocol is associated with higher recall rates, detection rates and positive predictive values of CT screening for lung cancer (24).

The aforementioned diagnostic methods typically diagnose patients with NSCLC at an advanced clinical stage, losing the optimal time frame for treatment and thus shortening survival times. Therefore, modified and optimized CT screening techniques should be developed for the diagnosis of early stage NSCLC. Novel imaging techniques could enhance the sensitivity of the detection of early stage tumor morphology. A previous study has evaluated the application of dynamic contrast-enhanced CT imaging in the study of the differentiation of benign and malignant tumors, observed by tumor vessel and permeability nodule perfusion (25). However, conventional contrast agents present lower efficacy for tumor analysis due to their rapid diffusion away from the lungs (26). Furthermore, previous reports have demonstrated that iodinated contrast agents are less sensitive to changes in cell morphology (27,28).

Nanoparticle-based imaging contrast agents exhibit potential in diagnosing early-stage cancer through providing a more accurate and sensitive detection of tumor nodules (29,30). Cho et al (31) investigated inorganic nanoparticles containing semiconductor quantum dots, iron oxide and gold, which revealed their potential for use as contrast agents combined with CT for diagnostics.

The present study investigated the use of a nanoscale microbubble contrast agent for contrast-enhanced (CE) CT to detect early stage NSCLC. This highlighted the potential application of CECT-targeted nanoparticle contrast agent (TNCA) imaging in the diagnosis of NSCLC. The results revealed the advantages of CECT-TNCA compared with CT alone in the early diagnosis and final confirmation of NSCLC. CECT-TNCA, with the contrast agent inhaled by nebulization, led to lesions being augmented and amplified in the lung during imaging, resulting in a reliable and sensitive assessment for the clinical diagnosis of NSCLC.

Materials and methods

Ethics statement

The present study was performed in accordance with the recommendations for the Guide for the Care and Use of Laboratory Animals of The First Hospital of Shijiazhuang (Shijiazhuang, China), and all animal work was approved by the Ethics Committee of The First Hospital of Shijiazhuang. All surgery and euthanasia were performed under sodium pentobarbital anesthesia, and all efforts were made to minimize suffering.

Scanning protocol

A CECT diagnosis system (MX4000, Philips Medical Systems, Inc., Bothell, WA, USA) was used to analyze the lungs using a preprogrammed setting, which was optimized to obtain the best image. Details of the settings used are described in previous study (32).

TNCA

A novel liposome-encapsulated contrast agent comprising lenvatinib-bound nanoparticles was used in the present study. The lenvatinib was bound to superparamagnetic iron oxide nanoparticles via a covalent bond, as described in a previous study (33). The route of administration for the TNCA or Optison™ (GE Healthcare Life Sciences, Little Chalfont, UK; used as a control) was inhalation using an atomizer. The microbubbles containing lenvatinib could reach the lesion through the pulmonary circulation due to their small diameter. After 30 min, the TNCA could be visualized using the CECT system. No side effects were observed from the nanoparticle-lenvatinib contrast agent.

Animal study

NSCLC tumors were established in mice as described previously (34). Briefly, C57BL/6 mice (age, 6–8 weeks) were purchased from Shanghai Laboratory Animal Co., Ltd. (Shanghai, China) and underwent intranasal infection with NSCLC cells (H358, NCI-H1755 or NCI-H661). NSCLC mice included adenocarcinoma, large cell carcinoma and squamous cell carcinoma (n=40 in each group). These mice were used for the imaging study 7 days after infection. All mice were housed in a temperature-controlled facility at 23±1°C and relative humidity of 50±5% with a 12-h light/dark cycle. All animals had free access to food and water. A total of 120 animals (60 male, 60 female; tumor free, n=34; tumor-bearing, n=86) were used to examine the efficacy of CECT-TNCA. Subsequently, the liposomal contrast agent (1,400 mg/kg) was inhaled for 20 min via an atomizer. Post-contrast scans were then conducted immediately (0 h), and 72 and 120 h after inhalation of the liposomal contrast agent. The experimental animals were then sacrificed, and 4-µm tumor specimens were fixed in 10% formaldehyde for 15 min at 37°C and underwent hematoxylin and eosin staining (2 h at 37°C) to confirm the tumor phenotype and morphology using a light microscope. All mice with early or late phase NSCLC were treated with cisplatin (1.2 mg/day) for a total of 10 days. Ten mice in each group were housed to observe the survival rate in 120 days.

Image data analysis

Data from the CECT-TNCA images was analyzed using the CECT system, including the volume of the tumors.

Statistical analysis

All data are presented as the mean ± standard deviation of triplicate experiments. Data was analyzed by SPSS 19.0 software (IBM Corp., Armonk, NY, USA) using one-way analysis of variance with Tukey's multiple comparison test. The Kaplan-Meier estimator was used to produce survival curves for the mice over the 120-day long treatment period. P<0.05 was considered to indicate a statistically significant difference.

Results

CECT-TNCA improves the sensitivity of the diagnosis of early stage NSCLC

A total of 120 mice with early stage NSCLC (adenocarcinoma, large cell carcinoma and squamous cell carcinoma; n=40/group) were used to analyze the efficacy of CECT-TNCA on mice with early stage NSCLC. Tumor metastasis was not observed in any of the mice. CECT-TNCA identified significantly more of the mice with NSCLC compared with CECT (32/120 vs. 7/120, respectively; P<0.001; Fig. 1). The characteristics of mice with NSCLC are detailed in Table I. These data suggest that, compared with CECT, CECT-TNCA is more sensitive for the diagnosis of early NSCLC.

Table I.

Characteristics of mice with NSCLC.

Table I.

Characteristics of mice with NSCLC.

Gender, no. of mice

CharacteristicMaleFemale
Number6060
NSCLC6060
Adenocarcinoma2020
Large cell carcinoma2020
Squamous cell carcinoma2020

[i] All mice were 6–8 weeks old. NSCLC, non-small cell lung cancer; CECT, contrast-enhanced computerized tomography; TNCA, targeted nanoparticle contrast agent.

CECT-TNCA improves the signal intensity fed back by lesions

In order to analyze the efficacy of CECT-TNCA, signal intensity was detected after administration of the liposomal contrast agent. This revealed that the nanoscale microbubble contrast agent significantly enhanced the signal intensity compared with CECT (P<0.05; Fig. 2A). The nanoscale microbubble contrast agent was cleared from systemic circulation in ~120 h (Fig. 2B). Dynamic analysis of lung tumor nodules revealed an enhanced signal after inhalation of the TNCA (Fig. 2C). These results suggest that the TNCA significantly improved signal intensity in lung tumor nodules.

Histological analysis of the phenotype of NSCLC

The phenotype of NSCLC was further analyzed by histological analysis (Fig. 3). Adenocarcinoma, large cell carcinoma and squamous cell carcinoma were identified by histological analysis. In addition, mice diagnosed by CECT-TNCA and CT were compared (Fig. 4). CECT-TNCA identified a higher number of mice with NSCLC compared with CT. These results highlight the accuracy of CECT-TNCA in diagnosing NSCLC.

Efficacy of CECT-TNCA

The pulmonary tumor nodules were further analyzed by CECT-TNCA and CECT, with results revealing a positive association between diameter and signal intensity (Fig. 5). Imaging analysis included NSCLC volume and fractional blood volume as a function of nodule diameter. In addition, signal intensity fed back by lesions in the NSCLC nodules was improved after administration of the targeted nanoscale microbubble contrast agent. These factors enabled the visualization of tiny tumor lesions in the lung (Fig. 6). Furthermore, the longitudinal aspect of CECT-TNCA allowed for the imaging of early-stage NSCLC tumors in mice (Fig. 7). In addition, the data demonstrated that survival after treatment was improved in mice with early-stage NSCLC compared with late-stage NSCLC (Fig. 8). These results indicate that CECT-TNCA-diagnosed early-stage tumors exhibited relatively higher signal enhancement compared with CT tumors in xenograft mice.

Discussion

NSCLC is one of the most common of types of respiratory cancer and a leading cause of cancer-associated mortality worldwide (35). The incidence of NSCLC and the number of NSCLC-associated mortalities is growing (36). Notably, the majority of newly diagnosed patients with NSCLC are already in a late phase, which decreases the probability of recovery and shortens survival time (37). Therapeutic regimens for advanced NSCLC include targeted intervention, chemotherapy, radiotherapy and immunotherapy (38).

Late-phase NSCLC is more aggressive, contributing to a poor survival rate (39,40). Patients with advanced stage NSCLC more frequently exhibit drug resistance and side effects (41). Therefore, diagnosis protocols for early stage NSCLC are required to improve patient survival. The early detection of NSCLC could improve patient survival due to earlier surgery, conventional treatments and comprehensive therapy (38). Clinically, the following imaging technologies are the most commonly used to diagnose tumors: Ultrasound, X-ray, CT scanning, PET scanning and magnetic resonance imaging (MRI) (42). CT scanning is relatively economic and accurate compared with the aforementioned imaging technologies (43,44). CT scanning and MRI are the most commonly used techniques to provide high spatial resolution imaging to assess tumors. Despite PET and single positron emission computerized tomography providing high-contrast sensitivity for NSCLC diagnosis, they provide a relatively low spatial resolution (45). Although CT scanning provides high spatial resolution, it is not efficient at diagnosing patients with early stage lung cancer (46,47).

The present study investigated a comprehensive approach of CECT combined with a TNCA in mice, in order to improve the accuracy of early stage NSCLC diagnosis. Lenvatinib encapsulated by liposomes was used as the TNCA. Lenvatinib is a multi-target tyrosine kinase inhibitor of vascular endothelial growth factor receptor (VEGFR) 1–3, fibroblast growth factor receptor (FGFR) 1–4, platelet-derived growth factor receptor (PDGFR) β, proto-oncogene tyrosine-protein kinase receptor Ret (Ret) and mast/stem cell growth factor receptor Kit (Kit). VEGF1-3, FGFR1-4, PDGFR-β, Ret and Kit-mediated angiogenesis have been identified as key factors in the development of human cancer (48,49). Lenvatinib has potent antitumor activity against a number of human tumors (50). A previous report has suggested targeting the receptor of lenvatinib in NSCLC (51). The results of the present study demonstrated that liposome-encapsulated lenvatinib has a potential application as a TNCA to improve the accuracy of early-stage NSCLC diagnosis. This technique enhanced the signal intensity in lesions in the lung, improving the spatial resolution of CECT.

Previously, liposomal and iodinated contrast agents have provided methods to detect solid tumors (52,53). The feasibility of spectral CT imaging for the detection of tumors with target-based contrast agents was evidenced in a previous study. Contrast agents have improved CT diagnosis in terms of image quality, as observed in a prospective randomized trial (54). However, the development of a non-invasive assay for the accurate diagnosis of NSCLC remains a challenge. The present study investigated the efficacy of a liposome-encapsulated lenvatinib TNCA for the diagnosis of early stage NSCLC in a xenograft mice model. The results revealed that CECT-TNCA-diagnosed early stage NSCLC tumors exhibited increased signal enhancement compared with CT-diagnosed tumors. This suggests that the liposome-encapsulated targeted contrast agent enables high-resolution imaging of early phase NSCLC.

In conclusion, the current study provided insights into improving CT imaging resolution using a liposome-encapsulated targeted contrast agent. The preclinical application of thus imaging procedure would provide novel opportunities to assess the efficacy of early-stage NSCLC diagnosis and determine whether nanoparticles exhibit absorbability without affecting the respiratory system. The results of the present study indicate that CECT-TNCA may be of significant value in the diagnosis of NSCLC. Further studies are required to validate these results.

References

1 

van der Wekken AJ, Saber A, Hiltermann TJ, Kok K, van den Berg A and Groen HJ: Resistance mechanisms after tyrosine kinase inhibitors afatinib and crizotinib in non-small cell lung cancer, a review of the literature. Crit Rev Oncol Hematol. 100:107–116. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Joseph SS, Yentz SE, Mikkilineni S, Nelson C and Kalemkerian GP: Eyelid metastasis in non-small cell lung cancer: Diagnosis and management. Am J Med. 129:e169–e172. 2016. View Article : Google Scholar : PubMed/NCBI

3 

DeCotiis C, Hu Y, Greenberg AK, Huie M, Tsay JC, Pass H, Goldberg JD and Rom WN: Inflammatory cytokines and non-small cell lung cancer in a CT-scan screening cohort: Background review of the literature. Cancer Biomark. 16:219–233. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Kepka L and Socha J: PET-CT use and the occurrence of elective nodal failure in involved field radiotherapy for non-small cell lung cancer: A systematic review. Radiother Oncol. 115:151–156. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Morinaga R, Okamoto I, Furuta K, Kawano Y, Sekijima M, Dote K, Satou T, Nishio K, Fukuoka M and Nakagawa K: Sequential occurrence of non-small cell and small cell lung cancer with the same EGFR mutation. Lung Cancer. 58:411–413. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Khreish F, Hellwig D, Mathews J, Bücker A, Kirsch CM and Grgic A: Simultaneous occurrence of typical carcinoid and non-small-cell lung cancer in the same lung lobe: Value of nuclear medicine. Clin Nucl Med. 36:481–483. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Targowski T, Janda P, Owczarek W, Raczka A, Jahnz-Rózyk K and Plusa T: Evaluation of occurrence frequency of circulating p53 protein in serum of patients with chronic obstructive pulmonary diseases and non-small cell lung cancer. Pol Merkur Lekarski. 28:265–267. 2010.(In Polish). PubMed/NCBI

8 

Xie FJ, Lu HY, Zheng QQ, Qin J, Gao Y, Zhang YP, Hu X and Mao WM: The clinical pathological characteristics and prognosis of FGFR1 gene amplification in non-small-cell lung cancer: A meta-analysis. Onco Targets and Ther. 9:171–181. 2016. View Article : Google Scholar

9 

Moro-Sibilot D, Smit E, de Castro Carpeno J, Lesniewski-Kmak K, Aerts JG, Villatoro R, Kraaij K, Nacerddine K, Dyachkova Y, Smith KT, et al: Non-small cell lung cancer patients with brain metastases treated with first-line platinum-doublet chemotherapy: Analysis from the European FRAME study. Lung Cancer. 90:427–432. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Lim SH, Sun JM, Lee SH, Ahn JS, Park K and Ahn MJ: Pembrolizumab for the treatment of non-small cell lung cancer. Expert Opin Biol Ther. 16:397–406. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Ulivi P, Mercatali L, Casoni GL, Scarpi E, Bucchi L, Silvestrini R, Sanna S, Monteverde M, Amadori D, Poletti V and Zoli W: Multiple marker detection in peripheral blood for NSCLC diagnosis. PloS One. 8:e574012013. View Article : Google Scholar : PubMed/NCBI

12 

Mroczko B, Szmitkowski M and Czygier M: Granulocyte colony stimulating factor (G-CSF) in diagnosis and monitoring of non-small-cell lung cancer (NSCLC). Pol Arch Med Wewn. 103:163–168. 2000.(In Polish). PubMed/NCBI

13 

Muller B, Bovet M, Yin Y, Stichel D, Malz M, González-Vallinas M, Middleton A, Ehemann V, Schmitt J, Muley T, et al: Concomitant expression of far upstream element (FUSE) binding protein (FBP) interacting repressor (FIR) and its splice variants induce migration and invasion of non-small cell lung cancer (NSCLC) cells. J Pathol. 237:390–401. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Zhao Q, Yue J, Zhang C, Gu X, Chen H and Xu L: Inactivation of M2 AChR/NF-κB signaling axis reverses epithelial-mesenchymal transition (EMT) and suppresses migration and invasion in non-small cell lung cancer (NSCLC). Oncotarget. 6:29335–29346. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Kim DS, Park KM, Won YS, Kim JY, Lee JK, Kim JG, Oh ST, Jung SS and Kang WK: Occurrence and prognosis of symptomatic venous thromboembolism in colorectal cancer surgery patients. Vasc Specialist Int. 30:49–55. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Thunnissen E, Kerr KM, Herth FJ, Lantuejoul S, Papotti M, Rintoul RC, Rossi G, Skov BG, Weynand B, Bubendorf L, et al: The challenge of NSCLC diagnosis and predictive analysis on small samples. Practical approach of a working group. Lung Cancer. 76:1–18. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Geng J, Sun J, Lin Q, Gu J, Zhao Y, Zhang H, Feng X, He Y, Wang W, Zhou X and Yu J: Methylation status of NEUROG2 and NID2 improves the diagnosis of stage I NSCLC. Oncol Lett. 3:901–906. 2012.PubMed/NCBI

18 

Peters S, Adjei AA, Gridelli C, Reck M, Kerr K and Felip E: ESMO Guidelines Working Group: Metastatic non-small-cell lung cancer (NSCLC): ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 23 Suppl 7:vii56–vii64. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Jafri SH, Shi R and Mills G: Advance lung cancer inflammation index (ALI) at diagnosis is a prognostic marker in patients with metastatic non-small cell lung cancer (NSCLC): A retrospective review. BMC Cancer. 13:1582013. View Article : Google Scholar : PubMed/NCBI

20 

Vilmar AC, Santoni-Rugiu E and Sørensen JB: ERCC1 and histopathology in advanced NSCLC patients randomized in a large multicenter phase III trial. Ann Oncol. 21:1817–1824. 2010. View Article : Google Scholar : PubMed/NCBI

21 

National Lung Screening Trial Research Team, . Aberle DR, Adams AM, Berg CD, Black WC, Clapp JD, Fagerstrom RM, Gareen IF, Gatsonis C, Marcus PM and Sicks JD: Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 365:395–409. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Saldias PF, Díaz PJ, Rain MC, Illanes CP, Díaz TR and Díaz PO: Early detection of lung cancer using computed tomography among patients with chronic obstructive pulmonary disease. Rev Med Chil. 144:202–210. 2016.(In Spanish). PubMed/NCBI

23 

Rubin GD: Lung nodule and cancer detection in computed tomography screening. J Thorac Imaging. 30:130–138. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Seigneurin A, Field JK, Gachet A and Duffy SW: A systematic review of the characteristics associated with recall rates, detection rates and positive predictive values of computed tomography screening for lung cancer. Ann Oncol. 25:781–791. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Sudarski S, Henzler T and Schoenberg SO: Post-therapeutic positron emission tomography/computed tomography for early detection of non-small cell lung cancer recurrence. Transl Lung Cancer Res. 2:295–303. 2013.PubMed/NCBI

26 

Hagberg GE, Mamedov I, Power A, Beyerlein M, Merkle H, Kiselev VG, Dhingra K, Kubìček V, Angelovski G and Logothetis NK: Diffusion properties of conventional and calcium-sensitive MRI contrast agents in the rat cerebral cortex. Contrast Media Mol Imaging. 9:71–82. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Turetschek K, Preda A, Novikov V, Brasch RC, Weinmann HJ, Wunderbaldinger P and Roberts TP: Tumor microvascular changes in antiangiogenic treatment: Assessment by magnetic resonance contrast media of different molecular weights. J Magn Reson Imaging. 20:138–144. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Samei E, Saunders RS, Badea CT, Ghaghada KB, Hedlund LW, Qi Y, Yuan H, Bentley RC and Mukundan S Jr: Micro-CT imaging of breast tumors in rodents using a liposomal, nanoparticle contrast agent. Int J Nanomedicine. 4:277–282. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Palekar RU, Jallouk AP, Lanza GM, Pan H and Wickline SA: Molecular imaging of atherosclerosis with nanoparticle-based fluorinated MRI contrast agents. Nanomedicine (Lond). 10:1817–1832. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Oghabian MA and Farahbakhsh NM: Potential use of nanoparticle based contrast agents in MRI: A molecular imaging perspective. J Biomed Nanotechnol. 6:203–213. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Cho EC, Glaus C, Chen J, Welch MJ and Xia Y: Inorganic nanoparticle-based contrast agents for molecular imaging. Trends Mol Med. 16:561–573. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Nakamoto Y, Ishimori T, Sano K, Temma T, Ueda M, Saji H and Togashi K: Clinical efficacy of dual-phase scanning using (68)Ga-DOTATOC-PET/CT in the detection of neuroendocrine tumours. Clin Radiol. 71:1069.e1–e5. 2016. View Article : Google Scholar

33 

Chen CL, Hu GY, Mei Q, Qiu H, Long GX and Hu GQ: Epidermal growth factor receptor-targeted ultra-small superparamagnetic iron oxide particles for magnetic resonance molecular imaging of lung cancer cells in vitro. Chin Med J (Engl). 125:2322–2328. 2012.PubMed/NCBI

34 

Yasugi M, Takigawa N, Ochi N, Ohashi K, Harada D, Ninomiya T, Murakami T, Honda Y, Ichihara E, Tanimoto M and Kiura K: Everolimus prolonged survival in transgenic mice with EGFR-driven lung tumors. Exp Cell Res. 326:201–209. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Fenton-Ambrose L and Kazerooni EA: Preventative care: Lung-cancer screens now worth the cost. Nature. 514:352014. View Article : Google Scholar : PubMed/NCBI

36 

Kulkarni S, Vella ET, Coakley N, Cheng S, Gregg R, Ung YC and Ellis PM: The use of systemic treatment in the maintenance of patients with non-small cell lung cancer: A systematic review. J Thorac Oncol. 11:989–1002. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Yoshiba S, Jansen M, Matsushima N, Chen S and Mendell J: Population pharmacokinetic analysis of patritumab, a HER3 inhibitor, in subjects with advanced non-small cell lung cancer (NSCLC) or solid tumors. Cancer Chemother Pharmacol. 77:987–996. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Weller A, O'Brien ME, Ahmed M, Popat S, Bhosle J, McDonald F, Yap TA, Du Y, Vlahos I and deSouza NM: Mechanism and non-mechanism based imaging biomarkers for assessing biological response to treatment in non-small cell lung cancer. Eur J Cancer. 59:65–78. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Kim SH, Cho BC, Choi HJ, Chung KY, Kim DJ, Park MS, Kim SK, Chang J, Shin SJ, Sohn JH and Kim JH: The number of residual metastatic lymph nodes following neoadjuvant chemotherapy predicts survival in patients with stage III NSCLC. Lung Cancer. 60:393–400. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Satouchi M, Negoro S, Funada Y, Urata Y, Shimada T, Yoshimura S, Kotani Y, Sakuma T, Watanabe H, Adachi S, et al: Predictive factors associated with prolonged survival in patients with advanced non-small-cell lung cancer (NSCLC) treated with gefitinib. Br J Cancer. 96:1191–1196. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Soldan K, Pooley FD, Hansen J, Andersen A, Chang-Claude J, Ferro G, Ohgaki H, Skov BG, Cherrie JW, Saracci R and Boffetta P: Lung fibre burden in lung cancer cases employed in the rock and slag wool industry. Ann Occup Hyg. 50:241–248. 2006.PubMed/NCBI

42 

Garcia Duenas OF, Villanueva Kerckoff H, Olvera Rico H and Plascencia Lira J: Benign peritoneal cystic mesothelioma as differential diagnose of an ovarian dependant tumor. Case report and review of the literature. Ginecol Obstet Mex. 75:111–114. 2007.(In Spanish). PubMed/NCBI

43 

Garg PK, Deo SV, Kumar R, Shukla NK, Thulkar S, Gogia A, Sharma DN and Mathur SR: Staging PET-CT scanning provides superior detection of lymph nodes and distant metastases than traditional imaging in locally advanced breast cancer. World J Surg. 40:2036–2042. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Nogami Y, Banno K, Irie H, Iida M, Masugi Y, Murakami K and Aoki D: Efficacy of 18-FDG PET-CT dual-phase scanning for detection of lymph node metastasis in gynecological cancer. Anticancer Res. 35:2247–2253. 2015.PubMed/NCBI

45 

Davison CA, Chapman SE, Sasser TA, Wathen C, Diener J, Schafer ZT and Leevy WM: Multimodal optical, X-ray CT and SPECT imaging of a mouse model of breast cancer lung metastasis. Curr Mol Med. 13:368–376. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Genestreti G, Burgio MA, Matteucci F, Piciucchi S, Scarpi E, Monti M, Bucchi L, Parisi E, Crociani L, Gurioli C, et al: Endobronchial/Endoesophageal Ultrasound (EBUS/EUS) guided fine needle aspiration (FNA) and 18F-FDG PET/CT scanning in restaging of locally advanced non-small cell lung cancer (NSCLC) Treated with Chemo-radiotherapy: A Mono-institutional pilot experience. Technol Cancer Res Treat. 14:721–727. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Manowitz A, Sedlar M, Griffon M, Miller A, Miller J and Markowitz S: Use of BMI guidelines and individual dose tracking to minimize radiation exposure from low-dose helical chest CT scanning in a lung cancer screening program. Acad Radiol. 19:84–88. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Rini BI and Atkins MB: Resistance to targeted therapy in renal-cell carcinoma. Lancet Oncol. 10:992–1000. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Eichelberg C, Junker K, Ljungberg B and Moch H: Diagnostic and prognostic molecular markers for renal cell carcinoma: A critical appraisal of the current state of research and clinical applicability. Eur Urol. 55:851–863. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Hutson TE: Targeted therapies for the treatment of metastatic renal cell carcinoma: Clinical evidence. Oncologist. 16 Suppl 2:S14–S22. 2011. View Article : Google Scholar

51 

Nishio M, Horai T, Horiike A, Nokihara H, Yamamoto N, Takahashi T, Murakami H, Yamamoto N, Koizumi F, Nishio K, et al: Phase 1 study of lenvatinib combined with carboplatin and paclitaxel in patients with non-small-cell lung cancer. Br J Cancer. 109:538–544. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Achenbach S, Paul JF, Laurent F, Becker HC, Rengo M, Caudron J, Leschka S, Vignaux O, Knobloch G, Benea G, et al: Erratum to: Comparative assessment of image quality for coronary CT angiography with iobitridol and two contrast agents with higher iodine concentrations: Iopromide and iomeprol. A multicentre randomized double-blind trial. Eur Radiol. 27:8312017. View Article : Google Scholar : PubMed/NCBI

53 

Mannheim JG, Schlichthaerle T, Kuebler L, Quintanilla-Martinez L, Kohlhofer U, Kneilling M and Pichler BJ: Comparison of small animal CT contrast agents. Contrast Media Mol Imaging. 11:272–284. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Honoris L, Zhong Y, Chu E, Rosenthal D, Li D, Lam F and Budoff MJ: Comparison of contrast enhancement, image quality and tolerability in Coronary CT angiography using 4 contrast agents: A prospective randomized trial. Int J Cardiol. 186:126–128. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 14 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yuan N, Zhang X, Cao Y, Jiang X, Zhao S, Feng Y, Fan Y, Lu Z and Gao H: Contrast‑enhanced computerized tomography combined with a targeted nanoparticle contrast agent for screening for early‑phase non‑small cell lung cancer. Exp Ther Med 14: 5063-5068, 2017
APA
Yuan, N., Zhang, X., Cao, Y., Jiang, X., Zhao, S., Feng, Y. ... Gao, H. (2017). Contrast‑enhanced computerized tomography combined with a targeted nanoparticle contrast agent for screening for early‑phase non‑small cell lung cancer. Experimental and Therapeutic Medicine, 14, 5063-5068. https://doi.org/10.3892/etm.2017.5140
MLA
Yuan, N., Zhang, X., Cao, Y., Jiang, X., Zhao, S., Feng, Y., Fan, Y., Lu, Z., Gao, H."Contrast‑enhanced computerized tomography combined with a targeted nanoparticle contrast agent for screening for early‑phase non‑small cell lung cancer". Experimental and Therapeutic Medicine 14.5 (2017): 5063-5068.
Chicago
Yuan, N., Zhang, X., Cao, Y., Jiang, X., Zhao, S., Feng, Y., Fan, Y., Lu, Z., Gao, H."Contrast‑enhanced computerized tomography combined with a targeted nanoparticle contrast agent for screening for early‑phase non‑small cell lung cancer". Experimental and Therapeutic Medicine 14, no. 5 (2017): 5063-5068. https://doi.org/10.3892/etm.2017.5140