Open Access

MACC‑1 antibody target therapy suppresses growth and migration of non‑small cell lung cancer

Retraction in: /10.3892/mmr.2023.13130

  • Authors:
    • Woda Shi
    • Jianxiang Song
    • Wencai Wang
    • Yajun Zhang
    • Shiying Zheng
  • View Affiliations

  • Published online on: September 19, 2017     https://doi.org/10.3892/mmr.2017.7517
  • Pages: 7329-7336
  • Copyright: © Shi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Non‑small‑cell lung cancer (NSCLC) accounts for ~80% of human lung cancers that result in mortalities worldwide. Metastasis‑associated in colon cancer‑1 (MACC‑1) has been demonstrated to be significantly expressed in cases of NSCLC and promotes tumor cell migration and metastasis through transactivation of the metastasis‑inducing hepatocyte growth factor/MET proto‑gene, receptor tyrosine kinase (HGF/MET) signaling pathway. The present study constructed a chimeric antibody (Chanti‑MACC‑1) targeting MACC‑1 and investigated its potential as a molecular therapeutic target in the treatment of NSCLC therapy. The expression of MACC‑1 was detected by reverse transcription‑quantitative polymerase chain reaction and western blotting in lung cancer cell lines and tissues. MTT assay was used to detect proliferation of A549 cells treated by Chanti‑MACC‑1, whereas the functional and regulatory effects of Chanti‑MACC‑1 in the migration and metastasis of NSCLC cells was investigated by a cell invasion assay. The therapeutic effect and survival time was observed in animal models. The results demonstrated that MACC‑1 expression was increased and overexpression of MACC‑1 promoted the progression of the cell cycle, significantly promoted NSCLC cell growth and enhanced tumor migration and invasion through the HGF/MET signaling pathway. It was further demonstrated that Chanti‑MACC‑1 efficiently suppressed MACC‑1 expression and significantly inhibited NSCLC cell proliferation, migration and invasion by blocking the HGF/MET signaling pathway. The data revealed that Chanti‑MACC‑1 was not only beneficial for tumor remission, however additionally contributed to the long‑term survival of NSCLC ‑bearing mice. The findings of the present study indicated that MACC‑1 was significantly upregulated and promoted tumor cell growth and migration in NSCLC cells and tissues via transactivation of the metastasis‑inducing HGF/MET signaling pathway. However, Chanti‑MACC‑1significantly inhibited tumor growth and metastasis, which suggested that MACC‑1 may be essential for tumor initiation and progression by negatively regulating tumor suppressors.

Introduction

Target immunotherapies have been suggested to be important in the termination of tumor initiation, development, progression and deterioration, via immunologic cytotoxicity and personalized adoptive cellular immunotherapy (1,2). It has previously been demonstrated that various target molecules specific to tumor antigens may be used to suppress tumor growth, migration, invasion or metastasis via targeting metastasis-associated pivotal proteins, and may act as potential therapeutic strategies with tumor-inhibition and anti-metastatic properties in human cancer treatments (3,4). Cafarotti et al (5) reported that the personalized target therapy era ideally involves therapeutically treating each individual human disease case, including cancers, infections and hereditary diseases, in different ways that are most efficient and in accordance with the patient's unique genome.

Lung cancer is a primary public health concern and the leading cause of cancer-associated mortalities worldwide (6). Non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) are two particular variations of lung cancer, which account for ~85 and 15% of the incidence rate, respectively, in human cancer clinical statistical analysis (7). NSCLC includes large cell carcinoma, squamous cell carcinoma and adenocarcinoma that additionally present an increasing trend and incidence rate (810). Various investigations regarding therapeutic treatments for NSCLC have previously been conducted, however the overall 5-year survival rate is <15% in patients with NSCLC, and this is of primary clinical concern (9,11,12).

NSCLC is one of the most frequently occurring cancers resulting from poor air quality and high levels of air contamination (13). Migration and invasion are the predominant features of tumor metastasis and development. The migratory, invasive and metastatic capabilities of NSCLC result in the poor survival rate during treatment and subsequent recurrence of the disease in patients (14,15). Therefore, the development and identification of effective agents for inhibition of migration and invasion, and individualized medication for NSCLC, is of primary concern regarding treatment of cancer patients (16,17). Lung cancer spectral histopathology statistical analysis indicates that standard bio-therapy protocols, including cell therapy and target therapy, results in beneficial outcomes in 95% of patients, compared with traditional treatments (7). A previous study suggested that bio-therapy stiffness modulates lung cancer cell migration via focal adhesion signaling as opposed to epithelial mesenchymal transition (EMT) signaling (18).

Significant advances have been made, particularly with the discovery of targeted agents. Metastasis-associated in colon cancer-1 (MACC-1) is a protein that promotes human lung cancer cell metastasis and is associated with poor patient prognosis in NSCLC (19). Expression levels of MACC-1 have been observed to be increased in human colorectal cancer, and promote tumor migration and metastasis through transactivating the metastasis-inducing hepatocyte growth factor/MET proto-gene, receptor tyrosine kinase (HGF/MET) signaling pathway (20).

The present study investigated if targeting of MACC-1 is a reliable strategy for the inhibition of NSCLC migration and metastasis in vitro and in vivo. Furthermore, the regulatory pathway of MACC-1 and its functional role in NSCLC was investigated. Overall, the results of the present study suggest that targeting of MACC-1 may act as a promising potential therapeutic strategy for intervention in metastasis formation and treatment of NSCLC.

Materials and methods

Ethics statement

The present study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the First Affiliated Hospital of Soochow University (Suzhou, China). The study was approved by the ethics committee of the Third People's Hospital of Yancheng (Yancheng, China). All surgery and euthanasia were performed under sodium pentobarbital anesthesia, and all efforts were made to minimize suffering. The animals were anesthetized with 35 mg/kg sodium pentobarbital by tail vein injection.

Cell culture

CHO-K1SV, NCI-H520, A549, H358 and MRC-5 cells were purchased from American Type Culture Collection (Manassas, VA, USA). NCI-H520, A549 and H358 cells were cultured in Dulbecco's modified Eagle's medium, supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA). MRC-5 cells were cultured in RPMI-1640 supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.). All cells were cultured at 37°C in an environment containing 5% CO2.

Lung cancer tissues

Lung cancer tissue samples of large cell carcinoma, squamous cell carcinoma and adenocarcinoma were obtained from patients that underwent tumorectomies in the First Affiliated Hospital of Soochow University. The ethical committee members reviewed the experimental designs and protocols and gave ethical approval. A total of 9 male lung cancer patients were enrolled in the study during May 2015 to July 2015, with large cell carcinoma (n=3), squamous cell carcinoma (n=3) and adenocarcinoma (n=3). The mean age of the patients was 48.3 years old (range, 35.6–68.4). All patients agreed to participate in the present study and gave written informed consent.

Construction of full length Anti-MACC-1 antibody

The mouse anti-human MACC-1 monoclonal antibody was constructed using a conventional approach. The chimeric antibody (Chanti)-MACC-1 was constructed as previously described (21) The single chain variable fragments of the MACC-1 monoclonal antibody were cloned and inserted into the Pklight vector (Hengfei Bioscience, Inc., Shanghai China). The constant domain heavy chain (CH)-Fc and light chain (CL) fragments were subcloned into the Pklight-anti-MACC-1 vector, which were subcloned into the Peedual 12.4 vector (Hengfei Bioscience, Inc.). Subsequently, this was transfected into CHO-K1SV cells using Lipofectamine® 2000 (Sigma-Aldrich; Merck KGaA) at room temperature for 20 min. The production of Chanti-MACC-1extracted from the CHO-K1SV cells was confirmed by using SDS-PAGE western blotting.

Enzyme-linked immunosorbent assay (ELISA)

The affinity of Chanti-MACC-1 for its target antigens mouse-MACC-1 (m MACC-1) and human-MACC-1 (hMACC-1) was determined using ELISA kit (cat. no. bs-4293R; BIOSS, Beijing China). A total of 0.2–1.2 mg/ml mMACC-1 and hMACC-1 protein was added into a microplate and incubated with Chanti-MACC-1 at 4°C for 12 h. Chanti-MACC-1 (3 µg/ml) was subsequently added to the mMACC-1 and hMACC-1 microplate and incubated for 120 min at 37°C and PBS was used as a control. The protocol was conducted as previously described (22) and the results were analyzed at a wavelength of 450 nm by an ELISA plate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

MTT assay

A549 cells were cultured in 6-well plates and grown to ~90% monolayer cells. Subsequently, Chanti-MACC-1 was added into the 6-well plates for 12 h. A total of 10 µl MTT was used, and formazan crystals were subsequently dissolved in 100 µl dimethyl sulfoxide, then the procedure was conducted as previously described (23). The results were determined by a spectrophotometer (Bio-Rad Laboratories, Inc.) at a wavelength of 570 nm.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was obtained from NCI-H520, A549, H358 and MRC-5 cells prior or the tumor tissues with Chanti-MACC-1 by using RNAeasy Mini kit (24) (Qiagen Sciences, Inc., Gaithersburg, MD, USA). A total of 1 µg total RNA was then transcribed into cDNA using the PrimeScript™ RT Master mix (Perfect Real Time; Takara Biotechnology Co., Ltd.) in an ABI PRISM 7900 real time system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The quality of the synthetic cDNA was verified by electrophoresis. Subsequently, the synthetic cDNA (10 ng) was subjected to RT-qPCR using SYBR-Green Master Mix system (Bio-Rad Laboratories, Inc). The protocol of thermos cycling was as follows: Denaturation, 95°C for 2 min; annealing, 40 repetitions of 95°C for 30 sec and 60°C for 60 sec; and final extension, 72°C for 10 min. The primers used in the present study were synthesized by Shanghai Shenggong Biology Engineering Technology Service, Ltd., Shanghai, China. MACC-1 forward, 5′-AGTGGGATTGTGGAGACGGTGT-3′ and reverse, 5′-AGGTAAAAGGAACTGGCAACGC-3′; GAPDH forward, 5′-GTGGACATCCGCAAAGAC-3′ and reverse, 5′-AAAGGGTGTAACGCAACTA-3′. GAPDH was included as an internal control. Differences in mRNA expression alterations were calculated by 2−ΔΔCq (25). The results are expressed as the n-fold way compared with control.

Cell invasion assay

A549 cells were treated with Chanti-MACC-1 and non-treated cells served as control. Cultured cells were suspended at a density of 5×106 in 1,000 µl serum-free DMEM medium for 12 h in the upper chamber of a BD BioCoat Matrigel chamber (BD Biosciences, Franklin Lakes, NJ, USA) whereas the lower chamber was filled with 600 µl cultural medium supplied with 10% FBS. According to the manufacturer's protocol. Following incubation for 12 h at 37°C, cells in each group were fixed with ice-cold methanol for 10 min and stained with crystal violet for 5 min at room temperature. Images were captured under a light microscope (magnification, ×200; Nikon Corp., Tokyo, Japan). A549 cell invasion was determined in at least three randomly stained-fields using a microscope for each sample.

SDS-PAGE and western blot analysis

The supernatant of serum-free CHO-K1SV cells was harvested. Proteins were resolved by 12% SDS-PAGE under reducing conditions with β-mercaptoethanol (Yuanmu Bioscience Inc., Shanghai, China). A549 cells were treated with Chanti-MACC-1 at a concentration of 100 ng/ml for 12 h. Protein samples from colorectal tumors and A549 cells and were homogenized using radioimmunoprecipitation assay lysis buffer (Invitrogen; Thermo Fisher Scientific, Inc.) and samples were centrifuged at 7,103 × g at 4°C for 10 min. The protein concentrations of the cell extracts were then measured using Bradford protein dye reagent (Bio-Rad Laboratories, Inc., Hercules, CA, USA). A total of 30 µg/lane protein was loaded and separated by 12% SDS-PAGE and transferred to nitrocellulose membranes. The membranes were blocked with 5% skimmed milk for 1 h at room temperature, washed in Tris-buffered saline containing 0.1% Tween-20 (TBST) and incubated with the following primary antibodies at 4°C overnight: Anti-MACC-1 (1:2,000; cat. no. ab106579; Abcam, Cambridge, UK), anti-HGF (1:1,000; cat. no. ab83760; Abcam), anti-Met (1:2,000; cat. no. ab216574; Abcam), anti-Vimentin (1:2,000; cat. no. ab8978; Abcam), anti-E-cadherin (1:2,000; cat. no. ab11512; Abcam), anti-slug (1:2,000; cat. no. ab27568; Abcam), anti-MMP-1 (1:2,000; cat. no. ab52631; Abcam), anti-CT-1 (1:2,000; cat. no. ab13975; Abcam), anti-fibronectin (1:2,000; cat. no. ab2413; Abcam), A5 anti-rabbit primary IgG (1:1,500; cat. no. ab6721; Abcam) conjugated to horseradish peroxidase. The protein bands labeled with the antibodies were visualized using the SuperSignal West Pico Chemiluminescent Substrate Trial kit (Pierce; Thermo Fisher Scientific, Inc.). Images were obtained using the ChemiDoc XRS system with BandScan 5.0 software (Glyko, Inc., Novato, CA, USA).

Immunofluorescence

A549 cells or tumors from NSCLC xenograph mice were fixed by using 10% formaldehyde for 30 min at 4°C in the dark, embedded in paraffin and then the wax blocks were cut into sections (4 µm thickness). The sections were dewaxed by conventional methods and underwent microwave antigen retrieval at 95°C for 10 min. After cooling, they were washed with distilled water and blocked in normal 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) at room temperature for 30 min. A549 cells and tumor sections were incubated for 1 h at room temperature with MACC-1 antibodies (1:2,000; cat. no. ab106579; Abcam). Subsequently, the cells and sections were incubated with fluorophore-labeled secondary antibody (1:300; cat. no. ab150117; Abcam) for 15 min at room temperature. Sections were stained using 4′,6-diamidino-2-phenylindole (Hengfei Bioscience, Shanghai, China) for 2 min, and sealed using glycerol after being washed. Then the sections were placed under the fluorescence microscope (Olympus Corporation, Tokyo, Japan) for observation in the dark.

Animal studies

A total of 60 specific pathogen-free female C57BL/6 mice (6-week old and weight 16–22 g) were purchased from Shanghai SLAC Laboratory Animal Co., Ltd (Shanghai, China). All mice were housed at a specific temperature (22–24°C) in a pathogen-free room at 40–70% humidity with a 12 h light/dark cycle and free access to clean water and standard food. Mice were subcutaneously implanted with A549 tumor cells and were divided into 2 groups (n=30 per group). Treatments were started on day 6 following tumor implantation when the tumor diameter reached 5–6 mm. Xenograft mice were intravenously injected Chanti-MACC-1 (100 µg) and PBS as control. The treatment was continued for 14 days at a frequency of everyday. The tumor volumes were calculated as previously described (26).

Statistical analysis

All data were presented as the mean ± standard error of the mean of three independent replicates. Analysis was performed using SPSS software version 20.0 (IBM Corp., Armonk, NY, USA). Unpaired data was determined by Student's t-test and comparisons of data between multiple groups were analyzed by analysis of variance followed by a Student-Newman-Keuls test. A Kaplan-Meier test was used to estimate the survival rate during a 150-day observation. *P<0.05 and **P<0.01 were considered to indicate a statistically significant difference.

Results

MACC-1 expression increases in NSCLC cells and tissues

In order to investigate the role of MACC-1 in NSCLC and normal lung cells, RT-qPCR was used to analyze expression levels. The results in Fig. 1A and B demonstrated that mRNA and protein expression of MACC-1 was increased in NCI-H520, A549 and H358 cells compared with normal lung MRC-5 cells. In addition, relative mRNA and protein expression levels were additionally studied from patients with large cell carcinoma, squamous cell carcinoma and adenocarcinoma. It was observed that MACC-1 expression levels were upregulated at the mRNA and protein level in NSCLC tumor tissues (Fig. 1C and D). MACC-1 expression in A549 cells was the greatest in all cell lines. These results suggested that MACC-1 expression was upregulated in NSCLC, and expressed at a low level in MRC-5 cells.

Chanti-MACC-1 directly targets MACC-1 in NSCLC cells

In consideration of the increased expression of MACC-1 in NSCLC cells, the authors hypothesized that targeting of MACC-1 was beneficial for NSCLS tumor cell inhibition. As presented in Fig. 1, MACC-1 expression in A549 cells was the greatest among NSCLS cells. Therefore, A549 cells were used to further study the efficacy of Chanti-MACC-1in NSCLC cells. The results in Fig. 2A demonstrated that Chanti-MACC-1 efficiently decreased MACC-1 mRNA expression levels. The immunofluorescence assay revealed that MACC-1 expression was suppressed in A549 cells following treatment with Chanti-MACC-1 (Fig. 2B). In addition, the present study further investigated the efficient effects of Chanti-MACC-1 on NSCLS tumor cell growth and migration. The results in Fig. 2C demonstrated that A549 cell growth was significantly inhibited in the Chanti-MACC-1-treated group. However, it was observed that MACC-1 greatly promoted A549 cell growth compared with non-treated control. Chanti-MACC-1 treatment efficiently suppressed A549 cell migration, whereas MACC-1 significantly promoted A549 cell migration (Fig. 2D). These data indicated that Chanti-MACC-1 directly targets MACC-1 in NSCLC cells.

Chanti-MACC-1 inhibits the EMT process via the HGF/MET signaling pathway

A previous study indicated that MACC-1 is significantly associated with the EMT and tumor progression (27). The MACC-1-induced EMT signaling pathway was first analyzed, and observed to be blocked by Chanti-MACC-1. Therefore, the underlying mechanism of the effects of Chanti-MACC-1 on HGF/MET and EMT marker expression levels were analyzed, including Vimentin, E-cadherin and Slug. The results in Fig. 3A demonstrated that HGF/MET expression was downregulated in A549 cells following Chanti-MACC-1 treatment. In addition, EMT marker expression was analyzed and Fig. 3B demonstrated that Chanti-MACC-1 treatment decreased Vimentin, E-cadherin and Slug expression in A549 cells. Furthermore, migration-associated matrix metalloproteinase (MMP)-1 proteins, collagen type I (CT-I) and fibronectin were studied in A549 cells. The data presented in Fig. 3C revealed that MMP-1 proteins, CT-I and fibronectin protein expression levels appeared to be markedly decreased following treatment with Chanti-MACC-1 in A549 cells. These results suggested that Chanti-MACC-1 suppressed the migration-promoting proteins in the MACC-1-induced EMT process, which may be beneficial to treatment of cancer cell migration in the HGF/MET pathway.

Chanti-MACC-1 exhibits a therapeutic effect in tumor-bearing mice

Following confirmation of the in vitro effects and mechanism, the present study proceeded to analyze the in vivo effects of Chanti-MACC-1 in xenogeneic NSCLC in C57BL/6 mice. As presented in Fig. 4A, tumor growth was significantly decreased in Chanti-MACC-1-treated xenograft mice, compared with those treated with PBS. In addition, the results in Fig. 4B demonstrated that Chanti-MACC-1 treatment prolonged the survival of NSCLC-bearing mice in a 150-day observation, compared with control mice (n=30 in each group). Tumor metastasis was additionally observed and Fig. 4C demonstrated that Chanti-MACC-1 treatment inhibited 80% metastasis tumor-bearing mice compared to control group. Furthermore, the data in Fig. 4D indicated that MACC-1, HGF/MET expression levels were significantly downregulated in tumors following treatment with Chanti-MACC-1. These data suggested that inhibition of tumor metastasis in NSCLC-bearing mice was enhanced due to Chanti-MACC-1 therapy.

Discussion

The occurrence of lung cancer has previously been demonstrated to be associated with industrial pollution and destruction of the ecological environment in developing countries (28). NSCLC is the primary manifestation of human lung cancer (accountable for >80% of lung cancer cases) that are frequently diagnosed at an advanced stage. Therefore, numerous patients with NSCLC are informed of the limited survival time associated with the diagnosis (29). In addition, the relative increased morbidity and mortality rates of NSCLC among human cancers is currently of primary concern (28). Previous studies have demonstrated that NSCLC is exhibiting a significantly increasing trend in recent years, and is gradually becoming the focus of public opinion and a significant hazard to human health maintenance (30).

Currently, curative treatment of NSCLC is limited to surgical resection or orthotopic lung transplantation. However, not all patients with NSCLC benefit from a surgical approach and suffer from increased recurrence rates. Furthermore, conventional radiation and chemotherapy demonstrate little efficacy for NSCLC and more frequent recurrence and metastasis occurs. Patients generally exhibit a poor response to chemotherapy and surgery, and as of yet, no targeted therapy has been established. In addition, the poor survival rate of NSCLC patients is <15% following the overall 5-year census (31). Therefore, identification of novel therapeutic protocols to cure patients with NSCLC is of primary concern.

A previous study suggested that migration and metastasis are associated with a poor prognosis of patients with NSCLC (32). Numerous reports have indicated that migration and metastasis contribute to the short-term survival period and are important contributing factors to the relapse and retreatment of patients with NSCLC (33,34). Therefore, anti-tumor agents targeting metastasis-promoting factors are currently of great research interest for the treatment of patients with NSCLC.

MACC-1 is human colon cancer cell metastasis gene located on human chromosome 7 (35). Increased expression of MACC-1 is positively correlated with patient disease progression and prognosis in colorectal cancer (36). Previous biostatistics have revealed that the majority of NSCLC patients observed exhibit an increased expression of MACC-1 in tumors, that may contribute to a short-term survival period (37). Clinical studies have additionally demonstrated that MACC-1 is overexpressed in colorectal cancer (38). In addition, a previous study indicated that targeted therapy for MACC-1 in patients with NSCLC has been investigated in various human tumor cells (3944). Furthermore, MACC-1 may be useful in the identification of poor prognosis subjects with NSCLC in clinical diagnosis, and may act as a potential molecular target for intervention in metastasis formation. However, the mechanism of the signaling pathway of MACC-1 in the growth, migration and metastasis of NSCLC remains to be elucidated.

The results of the present study verified those of a previous study, identifying MACC-1 as a tumorigenesis and developmental metastasis-associated gene, which enhances NSCLC cell proliferation, migration and invasion (45). The data from the present study demonstrated that MACC-1 regulated proliferation, migration and invasion through HGF-induced scattering and the metastasis-inducing HGF/Met signaling pathway in tumor cells and xenograft models. MACC-1 was reported as a regulator of HGF/Met/mitogen-activated protein kinase pathway and induces proliferation, migration and metastasis. In the present study, migration and invasion-associated MMP-3, CT-I and fibronectin were studied and were demonstrated to be significantly decreased both in vitro and in vivo, following treatment with Chanti-MACC-1.

In conclusion, the results of the present study suggested that MACC-1 was overexpressed in NSCLC cells and induced the EMT signaling pathway to enhance growth, migration and metastasis. The data additionally indicated that MACC-1 was associated with a poor prognosis of mice with NSCLC. In addition, Chanti-MACC-1 targeting MACC-1 revealed beneficial outcomes for NSCLC-bearing mice, through inhibition of the HGF/Met/mitogen-activated protein kinase pathway. Furthermore, the results have identified the potential of Chanti-MACC-1 to suppress growth, migration and metastasis via regulating metastasis-associated genes, and to act as a novel therapeutic in anti-metastasis treatment strategies.

References

1 

Trautmann L, Said EA, Halwani R, Janbazian L, Chomont N, El-Far M, Breton G, Haddad EK and Sekaly RP: Programmed death 1: A critical regulator of T-cell function and a strong target for immunotherapies for chronic viral infections. Curr Opin HIV AIDS. 2:219–227. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Tanzarella S, Lionello I, Valentinis B, Russo V, Lollini PL and Traversari C: Rhabdomyosarcomas are potential target of MAGE-specific immunotherapies. Cancer Immunol Immunother. 53:519–524. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Selvan SR, Dowling JP, Kelly WK and Lin J: Indoleamine 2,3-dioxygenase (IDO): Biology and target in cancer immunotherapies. Curr Cancer Drug Targets. 16:755–764. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Pier GB: Rationale for development of immunotherapies that target mucoid Pseudomonas aeruginosa infection in cystic fibrosis patients. Behring Inst Mitt. 1–360. 1997.

5 

Cafarotti S, Lococo F, Froesh P, Zappa F and Andrè D: Target therapy in lung cancer. Adv Exp Med Biol. 893:127–136. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Magnuson WJ, Yeung JT, Guillod PD, Gettinger SN, Yu JB and Chiang VL: Impact of deferring radiation therapy in patients with epidermal growth factor receptor-mutant non-small cell lung cancer who develop brain metastases. Int J Radiat Oncol Biol Phys. 95:673–679. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Zhukovsky M, Varaksin A and Pakholkina O: Statistical analysis of observational study of the influence of radon and other risk factors on lung cancer incidence. Radiat Prot Dosimetry. 160:108–111. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Brody H: Lung cancer. Nature. 513 Suppl:S12014. View Article : Google Scholar : PubMed/NCBI

9 

Moro-Sibilot D, Smit E, de Castro Carpeño 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 

Barnett SA, Downey RJ, Zheng J, Plourde G, Shen R, Chaft J, Akhurst T, Park BJ and Rusch VW: Utility of routine PET imaging to predict response and survival after induction therapy for non-small cell lung cancer. Ann Thorac Surg. 101:1052–1059. 2016. View Article : Google Scholar : PubMed/NCBI

11 

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 Ther. 9:171–181. 2016. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Lupo B, Vialard J, Sassi F, Angibaud P, Puliafito A, Pupo E, Lanzetti L, Comoglio PM, Bertotti A and Trusolino L: Tankyrase inhibition impairs directional migration and invasion of lung cancer cells by affecting microtubule dynamics and polarity signals. BMC Biol. 14:52016. View Article : Google Scholar : PubMed/NCBI

14 

Müller 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

15 

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

16 

Zhang H, Zhu X, Li N, Li D, Sha Z, Zheng X and Wang H: miR-125a-3p targets MTA1 to suppress NSCLC cell proliferation, migration, and invasion. Acta Biochim Biophys Sin (Shanghai). 47:496–503. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Roth MT, Ivey JL, Esserman DA, Crisp G, Kurz J and Weinberger M: Individualized medication assessment and planning: Optimizing medication use in older adults in the primary care setting. Pharmacotherapy. 33:787–797. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Shukla VC, Higuita-Castro N, Nana-Sinkam P and Ghadiali SN: Substrate stiffness modulates lung cancer cell migration but not epithelial to mesenchymal transition. J Biomed Mater Res A. 104:1182–1193. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Shirahata A, Fan W, Sakuraba K, Yokomizo K, Goto T, Mizukami H, Saito M, Ishibashi K, Kigawa G, Nemoto H, et al: MACC 1 as a marker for vascular invasive hepatocellular carcinoma. Anticancer Res. 31:777–780. 2011.PubMed/NCBI

20 

Shirahata A, Sakata M, Kitamura Y, Sakuraba K, Yokomizo K, Goto T, Mizukami H, Saito M, Ishibashi K, Kigawa G, et al: MACC 1 as a marker for peritoneal-disseminated gastric carcinoma. Anticancer Res. 30:3441–3444. 2010.PubMed/NCBI

21 

Bai F, Tian H, Niu Z, Liu M, Ren G, Yu Y, Sun T, Li S and Li D: Chimeric anti-IL-17 full-length monoclonal antibody is a novel potential candidate for the treatment of rheumatoid arthritis. Int J Mol Med. 33:711–721. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Lanyon SR and Reichel MP: Pretreatment of serum samples to reduce interference of colostrum-derived specific antibodies with detection of Bovine viral diarrhea virus antigen by ELISA in young calves. J Vet Diagn Invest. 28:345–349. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Trino S, Iacobucci I, Erriquez D, Laurenzana I, De Luca L, Ferrari A, Di Rorà A Ghelli Luserna, Papayannidis C, Derenzini E, Simonetti G, et al: Targeting the p53-MDM2 interaction by the small-molecule MDM2 antagonist Nutlin-3a: A new challenged target therapy in adult Philadelphia positive acute lymphoblastic leukemia patients. Oncotarget. 7:12951–12961. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Ma J, Ma J, Meng Q, Zhao ZS and Xu WJ: Prognostic value and clinical pathology of MACC-1 and c-MET expression in gastric carcinoma. Pathol Oncol Res. 19:821–832. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-tie quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Zhuang T, Djemil T, Qi P, Magnelli A, Stephans K, Videtic G and Xia P: Dose calculation differences between Monte Carlo and pencil beam depend on the tumor locations and volumes for lung stereotactic body radiation therapy. J Appl Clin Med Phys. 14:40112013. View Article : Google Scholar : PubMed/NCBI

27 

Wang L, Lin L, Chen X, Sun L, Liao Y, Huang N and Liao W: Metastasis-associated in colon cancer-1 promotes vasculogenic mimicry in gastric cancer by upregulating TWIST1/2. Oncotarget. 6:11492–11506. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Lee YT, Liu CJ, Hu YW, Teng CJ, Tzeng CH, Yeh CM, Chen TJ, Lin JK, Lin CC, Lan YT, et al: Incidence of second primary malignancies following colorectal cancer: A distinct pattern of occurrence between colon and rectal cancers and association of Co-morbidity with second primary malignancies in a population-based cohort of 98,876 patients in Taiwan. Medicine (Baltimore). 94:e10792015. View Article : Google Scholar : PubMed/NCBI

29 

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

30 

Wink KC, Belderbos JS, Dieleman EM, Rossi M, Rasch CR, Damhuis RA, Houben RM and Troost EG: Improved progression free survival for patients with diabetes and locally advanced non-small cell lung cancer (NSCLC) using metformin during concurrent chemoradiotherapy. Radiother Oncol. 118:453–459. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Charvat H, Sasazuki S, Inoue M, Iwasaki M, Sawada N, Shimazu T, Yamaji T and Tsugane S: JPHC Study Group: Prediction of the 10-year probability of gastric cancer occurrence in the Japanese population: The JPHC study cohort II. Int J Cancer. 138:320–331. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Gold M, Dunn LB, Phoenix B, Paul SM, Hamolsky D, Levine JD and Miaskowski C: Co-occurrence of anxiety and depressive symptoms following breast cancer surgery and its impact on quality of life. Eur J Oncol Nurs. 20:97–105. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Arora S, Ranade AR, Tran NL, Nasser S, Sridhar S, Korn RL, Ross JT, Dhruv H, Foss KM, Sibenaller Z, et al: MicroRNA-328 is associated with (non-small) cell lung cancer (NSCLC) brain metastasis and mediates NSCLC migration. Int J Cancer. 129:2621–2631. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Han L, Liang XH, Chen LX, Bao SM and Yan ZQ: SIRT1 is highly expressed in brain metastasis tissues of non-small cell lung cancer (NSCLC) and in positive regulation of NSCLC cell migration. Int J Clin Exp Pathol. 6:2357–2365. 2013.PubMed/NCBI

35 

Sun L, Duan J, Jiang Y, Wang L, Huang N, Lin L, Liao Y and Liao W: Metastasis-associated in colon cancer-1 upregulates vascular endothelial growth factor-C/D to promote lymphangiogenesis in human gastric cancer. Cancer Lett. 357:242–253. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Lederer A, Herrmann P, Seehofer D, Dietel M, Pratschke J, Schlag P and Stein U: Metastasis-associated in colon cancer 1 is an independent prognostic biomarker for survival in Klatskin tumor patients. Hepatology. 62:841–850. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Stein U, Walther W, Arlt F, Schwabe H, Smith J, Fichtner I, Birchmeier W and Schlag PM: MACC1, a newly identified key regulator of HGF-MET signaling, predicts colon cancer metastasis. Nat Med. 15:59–67. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Boardman LA: Overexpression of MACC1 leads to downstream activation of HGF/MET and potentiates metastasis and recurrence of colorectal cancer. Genome Med. 1:362009. View Article : Google Scholar : PubMed/NCBI

39 

Shimokawa H, Uramoto H, Onitsuka T, Chundong G, Hanagiri T, Oyama T and Yasumoto K: Overexpression of MACC1 mRNA in lung adenocarcinoma is associated with postoperative recurrence. J Thorac Cardiovasc Surg. 141:895–898. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Migliore C, Martin V, Leoni VP, Restivo A, Atzori L, Petrelli A, Isella C, Zorcolo L, Sarotto I, Casula G, et al: miR-1 downregulation cooperates with MACC1 in promoting MET overexpression in human colon cancer. Clin Cancer Res. 18:737–747. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Yang T, Kong B, Kuang YQ, Cheng L, Gu JW, Zhang JH, Shu HF, Yu SX, He WQ, Xing XM and Huang HD: Overexpression of MACC1 protein and its clinical implications in patients with glioma. Tumour Biol. 35:815–819. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Wang Z, Li Z, Wu C, Wang Y, Xia Y, Chen L, Zhu Q and Chen Y: MACC1 overexpression predicts a poor prognosis for non-small cell lung cancer. Med Oncol. 31:7902014. View Article : Google Scholar : PubMed/NCBI

43 

Wang G, Fu Z and Li D: MACC1 overexpression and survival in solid tumors: A meta-analysis. Tumour Biol. 36:1055–1065. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Li H, Zhang H, Zhao S, Shi Y, Yao J, Zhang Y, Guo H and Liu X: Overexpression of MACC1 and the association with hepatocyte growth factor/c-Met in epithelial ovarian cancer. Oncol Lett. 9:1989–1996. 2015.PubMed/NCBI

45 

Nakamura K, Nozawa K, Aoyagi Y, Ishihara S, Matsuda K, Fukushima J and Watanabe T: A case report of thyroid gland metastasis associated with lung metastasis from colon cancer. Tumori. 97:229–232. 2011.PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shi W, Song J, Wang W, Zhang Y and Zheng S: MACC‑1 antibody target therapy suppresses growth and migration of non‑small cell lung cancer Retraction in /10.3892/mmr.2023.13130. Mol Med Rep 16: 7329-7336, 2017
APA
Shi, W., Song, J., Wang, W., Zhang, Y., & Zheng, S. (2017). MACC‑1 antibody target therapy suppresses growth and migration of non‑small cell lung cancer Retraction in /10.3892/mmr.2023.13130. Molecular Medicine Reports, 16, 7329-7336. https://doi.org/10.3892/mmr.2017.7517
MLA
Shi, W., Song, J., Wang, W., Zhang, Y., Zheng, S."MACC‑1 antibody target therapy suppresses growth and migration of non‑small cell lung cancer Retraction in /10.3892/mmr.2023.13130". Molecular Medicine Reports 16.5 (2017): 7329-7336.
Chicago
Shi, W., Song, J., Wang, W., Zhang, Y., Zheng, S."MACC‑1 antibody target therapy suppresses growth and migration of non‑small cell lung cancer Retraction in /10.3892/mmr.2023.13130". Molecular Medicine Reports 16, no. 5 (2017): 7329-7336. https://doi.org/10.3892/mmr.2017.7517