151
|
Drug-induced expression of EpCAM contributes to therapy resistance in esophageal adenocarcinoma. Cell Oncol (Dordr) 2018; 41:651-662. [PMID: 30116994 PMCID: PMC6244739 DOI: 10.1007/s13402-018-0399-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2018] [Indexed: 02/06/2023] Open
Abstract
Background With a less than 5% overall survival rate, esophageal adenocarcinoma (EAC) is one of the leading causes of death in the United States. Epithelial cell adhesion molecule (EpCAM) is a cancer stem cell (CSC) marker that is expressed in various epithelial carcinomas, including EAC. Accumulating evidence indicates that CSC subpopulations can initiate cancer development and, in addition, drive metastasis, recurrence and drug resistance. It has also been reported that EpCAM up-regulation in EAC may lead to an aggressive behavior and, thus, an adverse clinical outcome. Here, we aimed to determine whether treatment with standard chemotherapeutic agents may induce EpCAM expression and, concomitantly, increases in malignant potential and drug resistance in EAC. Methods EpCAM expression was assessed in 20 primary human EAC/adjacent normal tissues, as well as in a human EAC-derived cell line (OE-19), in a pre-malignant Barrett’s Esophagus cell line (Bar-T) and in a benign esophageal cell line (HET 1-A), using immunohistochemistry, Western blotting and qRT-PCR, respectively. Drug-induced resistance was investigated in OE-19-derived spheres treated with (a combination of) adriamycin, cisplatin and 5-fluorouracil (ACF) using survival, adhesion and flow cytometric assays, respectively, and compared to drug resistance induced by standard chemotherapeutic agents (CTA). Finally, ACF treatment-surviving cells were evaluated for their tumor forming capacities both in vitro and in vivo using spheroid formation and xenograft assays, respectively. Results High EpCAM expression was observed in esophageal cancer tissues and esophageal cancer-derived cell lines, but not in adjacent benign esophageal epithelia and benign esophageal cell lines (HET 1-A and Bar-T). The OE-19 cell spheres were drug resistant and EpCAM expression was significantly induced in the OE-19 cell spheres compared to the non-sphere OE-19 cells. When OE-19 cell spheres were challenged with ACF, the EpCAM mRNA and protein levels were further up-regulated up to 48 h, whereas a decreased EpCAM expression was observed at 72 h. EpCAM down-regulation by RNA interference increased the ACF efficacy to kill OE-19 cells. Increased EpCAM expression coincided with the CSC marker CD90 and was associated with an aggressive growth pattern of OE-19 cell spheres in vivo. Conclusions From our data we conclude that an ACF-induced increase in EpCAM expression reflects the selection of a CSC subpopulation that underlies tumor development and drug resistance in EAC.
Collapse
|
152
|
Kwok ZH, Roche V, Chew XH, Fadieieva A, Tay Y. A non-canonical tumor suppressive role for the long non-coding RNA MALAT1 in colon and breast cancers. Int J Cancer 2018; 143:668-678. [PMID: 29574704 DOI: 10.1002/ijc.31386] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 02/18/2018] [Accepted: 03/01/2018] [Indexed: 12/18/2022]
Abstract
Long noncoding RNAs (lncRNAs) constitute one of the largest classes of transcripts and have been widely implicated in various diseases such as cancer. Increasing evidence suggests that several lncRNAs are dysregulated and play critical roles in tumorigenesis. LncRNAs can be regulated by key oncogenes and tumor suppressors, adding complexity to the intricate crosstalk between protein coding genes and the noncoding transcriptome. In our study, we investigated the effect that dysregulation of the key tumor suppressor PTEN has on the noncoding transcriptome. We identified the lncRNA metastasis associated lung adenocarcinoma transcript 1 (MALAT1) as a target of PTEN and find that this regulation is conserved in both human and mouse as well as with both chronic and acute PTEN dysregulation. We show that this regulation is at least in part microRNA (miRNA)-dependent, and characterize the miRNAs that may be mediating this crosstalk. In summary, we establish and characterize a non-canonical PTEN-microRNA-MALAT1 axis that regulates tumorigenesis and describe for the first time that the MALAT1 lncRNA possesses novel tumor suppressive properties in colon and breast cancers.
Collapse
Affiliation(s)
- Zhi Hao Kwok
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Veronique Roche
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Xiao Hong Chew
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Anastasiia Fadieieva
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Yvonne Tay
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
153
|
Zhou L, Zhu Y. The EpCAM overexpression is associated with clinicopathological significance and prognosis in hepatocellular carcinoma patients: A systematic review and meta-analysis. Int J Surg 2018; 56:274-280. [DOI: 10.1016/j.ijsu.2018.06.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/07/2018] [Accepted: 06/10/2018] [Indexed: 02/08/2023]
|
154
|
Huang L, Yang Y, Yang F, Liu S, Zhu Z, Lei Z, Guo J. Functions of EpCAM in physiological processes and diseases (Review). Int J Mol Med 2018; 42:1771-1785. [PMID: 30015855 PMCID: PMC6108866 DOI: 10.3892/ijmm.2018.3764] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022] Open
Abstract
EpCAM (epithelial cell adhesion molecule) is a type I transmembrane glycoprotein, which was originally identified as a tumor-associated antigen due to its high expression level in rapidly growing epithelial tumors. Germ line mutations of the human EpCAM gene have been indicated as the cause of congenital tufting enteropathy. Previous studies based on cell models have revealed that EpCAM contributes to various biological processes including cell adhesion, signaling, migration and proliferation. Due to the previous lack of genetic animal models, the in vivo functions of EpCAM remain largely unknown. However, EpCAM genetic animal models have recently been generated, and are useful for understanding the functions of EpCAM. The authors here briefly review the functions and mechanisms of EpCAM in physiological processes and different diseases.
Collapse
Affiliation(s)
- Li Huang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Yanhong Yang
- The First Affiliated Hospital, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, P.R. China
| | - Fei Yang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Shaomin Liu
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Ziqin Zhu
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Zili Lei
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
155
|
Liang ZM, Chen Y, Luo ML. Targeting Stemness: Implications for Precision Medicine in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1026:147-169. [PMID: 29282683 DOI: 10.1007/978-981-10-6020-5_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The genomic landscape of breast cancer has been delineated in recent years. Advances in molecular characterization and targeting strategies are making it feasible to integrate clinical, genome-based and phenotype-based diagnostic and therapeutic methods and apply them to individual patient in the era of precision medicine. Cancer stem cells (CSCs) are a subpopulation in the tumor which have the capability of self-renewal and differentiation. Breast CSCs have important clinical implications as they account for tumor initiation, maintenance, metastasis, therapy resistance, and relapse. In this chapter, we will introduce approaches used to characterize breast CSCs, crucial pathways involved in regulating cancer stemness, and implications of breast CSCs in the precision diagnosis and treatment of breast cancer. We will also discuss novel compounds and therapeutic strategies that selectively target breast CSCs. Integration of breast CSC-related molecular diagnosis and targeted therapy into the clinical workflow of precision medicine has the potential to deliver more effective treatment to breast cancer patients.
Collapse
Affiliation(s)
- Zhi-Mei Liang
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yang Chen
- Department of Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Man-Li Luo
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
156
|
Xu L, He XY, Liu BY, Xu C, Ai SL, Zhuo RX, Cheng SX. Aptamer-functionalized albumin-based nanoparticles for targeted drug delivery. Colloids Surf B Biointerfaces 2018; 171:24-30. [PMID: 30005287 DOI: 10.1016/j.colsurfb.2018.07.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/18/2018] [Accepted: 07/03/2018] [Indexed: 01/10/2023]
Abstract
Proteins have been extensively explored as versatile nanocarriers for drug delivery due to their complete biocompatibility, ease of surface modification, and lack of toxicity and immunogenicity. In this study, a facile strategy was used to construct aptamer-functionalized albumin-based nanoparticles for effective drug delivery and targeted cancer therapy. A hydrophobic drug, doxorubicin (DOX) was employed to trigger the self-assembly of bovine serum albumin (BSA) to from stable nanoparticles via hydrophobic interaction, and then a tumor targeting aptamer AS1411 was incorporated to the surface of DOX loaded BSA. Due to the specific recognition between AS1411 and its receptor over-expressed on tumor cells, the aptamer-modified nanoparticles show higher cellular uptake and stronger cell inhibitory efficacy against cancerous MCF-7 cells as compared with the nanoparticles without aptamer modification. In addition, DOX loaded aptamer-functionalized nanoparticles can induce more significant down-regulation of Bcl-2 and PCNA as well as up-regulation of pRB, PARP and Bax in MCF-7 cells compared with unmodified nanoparticles, indicating the aptamer modification can induce cell apoptosis more effectively. Besides, aptamer-modified nanoparticles exhibit a significantly improved capability in up-regulating p16, p21 and E-cadherin, and down-regulating EpCAM, vimentin, Snail, MMP-9, CD44 and CD133, implying the favorable effects of drug delivery on the prevention of tumor progression and metastasis.
Collapse
Affiliation(s)
- Lei Xu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| | - Xiao-Yan He
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| | - Bo-Ya Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| | - Chang Xu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| | - Shu-Lun Ai
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| | - Ren-Xi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China.
| |
Collapse
|
157
|
Gatta AK, Hariharapura RC, Udupa N, Reddy MS, Josyula VR. Strategies for improving the specificity of siRNAs for enhanced therapeutic potential. Expert Opin Drug Discov 2018; 13:709-725. [PMID: 29902093 DOI: 10.1080/17460441.2018.1480607] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION RNA interference has become a tool of choice in the development of drugs in various therapeutic areas of Post Transcriptional Gene Silencing (PTGS). The critical element in developing successful RNAi therapeutics lies in designing small interfering RNA (siRNA) using an efficient algorithm satisfying the designing criteria. Further, translation of siRNA from bench-side to bedside needs an efficient delivery system and/or chemical modification. Areas covered: This review emphasizes the importance of dicer, the criteria for efficient siRNA design, the currently available algorithms and strategies to overcome off-target effects, immune stimulatory effects and endosomal trap. Expert opinion: Specificity and stability are the primary concerns for siRNA therapeutics. The design criteria and algorithms should be chosen rationally to have a siRNA sequence that binds to the corresponding mRNA as it happens in the Watson and Crick base pairing. However, it must evade a few more hurdles (Endocytosis, Serum stability etc.) to be functional in the cytosol.
Collapse
Affiliation(s)
- Aditya Kiran Gatta
- a Cell and Molecular Biology lab, Department of Pharmaceutical Biotechnology , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Raghu Chandrashekhar Hariharapura
- a Cell and Molecular Biology lab, Department of Pharmaceutical Biotechnology , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Nayanabhirama Udupa
- b Research Directorate of Health Sciences , Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Meka Sreenivasa Reddy
- c Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences , Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Venkata Rao Josyula
- a Cell and Molecular Biology lab, Department of Pharmaceutical Biotechnology , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal , Karnataka , India
| |
Collapse
|
158
|
Chen J, Shang B, Zhang H, Zhu Z, Chen L, Wang H, Ran F, Chen Q, Chen J. Enzyme-free ultrasensitive fluorescence detection of epithelial cell adhesion molecules based on a toehold-aided DNA recycling amplification strategy. RSC Adv 2018; 8:14798-14805. [PMID: 35541343 PMCID: PMC9079946 DOI: 10.1039/c8ra01362d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/04/2018] [Indexed: 01/13/2023] Open
Abstract
Epithelial cell adhesion molecules (EpCAMs) play a significant role in tumorigenesis and tumor development. EpCAMs are considered to be tumor signaling molecules for cancer diagnosis, prognosis and therapy. Herein, an enzyme-free and highly sensitive fluorescent biosensor, with a combined aptamer-based EpCAM recognition and toehold-aided DNA recycling amplification strategy, was developed for sensitive and specific fluorescence detection of EpCAMs. Due to highly specific binding between EpCAMs and corresponding aptamers, strand a, which is released from the complex of aptamer/strand a in the presence of EpCAMs which is bound to the corresponding aptamer, triggered the toehold-mediated strand displacement process. An amplified fluorescent signal was achieved by recycling strand a for ultrasensitive EpCAM detection with a detection limit as low as 0.1 ng mL-1, which was comparable or superior to that of reported immunoassays and biosensor strategies. In addition, high selectivity towards EpCAMs was exhibited when other proteins were selected as control proteins. Finally, this strategy was successfully used for the ultrasensitive fluorescence detection of EpCAMs in human serum samples with satisfactory results. Importantly, the present strategy may be also expanded for the detection of other targets using the corresponding aptamers.
Collapse
Affiliation(s)
- Jishun Chen
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Bing Shang
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Hua Zhang
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Zhengpeng Zhu
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Long Chen
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Hongmei Wang
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Fengying Ran
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Qinhua Chen
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| | - Jun Chen
- Affiliated Dongfeng Hospital, Hubei University of Medicine Hubei Shiyan 442008 China
| |
Collapse
|
159
|
Harun SNA, Israf DA, Tham CL, Lam KW, Cheema MS, Md Hashim NF. The Molecular Targets and Anti-Invasive Effects of 2,6-bis-(4-hydroxyl-3methoxybenzylidine) cyclohexanone or BHMC in MDA-MB-231 Human Breast Cancer Cells. Molecules 2018; 23:E865. [PMID: 29642589 PMCID: PMC6017078 DOI: 10.3390/molecules23040865] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 12/31/2022] Open
Abstract
In order to metastasize, tumor cells need to migrate and invade the surrounding tissues. It is important to identify compound(s) capable of disrupting the metastasis of invasive cancer cells, especially for hindering invadopodia formation, so as to provide anti-metastasis targeted therapy. Invadopodia are thought to be specialized actin-rich protrusions formed by highly invasive cancer cells to degrade the extracellular matrix (ECM). A curcuminoid analogue known as 2,6-bis-(4-hydroxy-3-methoxybenzylidine)cyclohexanone or BHMC has shown good potential in inhibiting inflammation and hyperalgesia. It also possesses an anti-tumor effects on 4T1 murine breast cancer cells in vivo. However, there is still a lack of empirical evidence on how BHMC works in preventing human breast cancer invasion. In this study, we investigated the effect of BHMC on MDA-MB-231 breast cancer cells and its underlying mechanism of action to prevent breast cancer invasion, especially during the formation of invadopodia. All MDA-MB-231 cells, which were exposed to the non-cytotoxic concentrations of BHMC, expressed the proliferating cell nuclear antigen (PCNA), which indicate that the anti-proliferative effects of BHMC did not interfere in the subsequent experiments. By using a scratch migration assay, transwell migration and invasion assays, we determined that BHMC reduces the percentage of migration and invasion of MDA-MB-231 cells. The gelatin degradation assay showed that BHMC reduced the number of cells with invadopodia. Analysis of the proteins involved in the invasion showed that there is a significant reduction in the expressions of Rho guanine nucleotide exchange factor 7 (β-PIX), matrix metalloproteinase-9 (MMP-9), and membrane type 1 matrix metalloproteinase (MT1-MMP) in the presence of BHMC treatment at 12.5 µM. Therefore, it can be postulated that BHMC at 12.5 µM is the optimal concentration for preventing breast cancer invasion.
Collapse
Affiliation(s)
- Siti Nor Aini Harun
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Daud Ahmad Israf
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Kok Wai Lam
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia.
| | - Manraj Singh Cheema
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Nur Fariesha Md Hashim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| |
Collapse
|
160
|
Heterogeneity of Circulating Tumor Cells in Neoadjuvant Chemotherapy of Breast Cancer. Molecules 2018; 23:molecules23040727. [PMID: 29565320 PMCID: PMC6017975 DOI: 10.3390/molecules23040727] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/14/2018] [Accepted: 03/18/2018] [Indexed: 12/12/2022] Open
Abstract
The biological properties of circulating tumor cells (CTCs), and their dynamics during neoadjuvant chemotherapy are important, both for disease progression prediction and therapeutic target determination, with the aim of preventing disease progression. The aim of our study was to estimate of different CTC subsets in breast cancer during the NACT (neoadjuvant chemotherapy). The prospective study includes 27 patients with invasive breast cancer, T2-4N0-3M0, aged 32 to 60 years. Venous heparinized blood samples, taken before and after biopsy, after each courses of chemotherapy (on days 3-7), and before surgical intervention, served as the material for this study. Different subsets of circulating tumor cells were determined on the basis of the expression of EpCAM, CD45, CD44, CD24, and N-Cadherin using flow cytometry. As the result of this study, it has been observed that significant changes in the quantity of the different subsets of circulating tumor cells in patients' blood were observed after carrying out the 3rd course of NACT. NACT causes significant changes in the quantity of six CTC subsets, with various combinations of stemness and epithelial-mesenchymal transition (EMT) properties.
Collapse
|
161
|
Pindiprolu SKSS, Krishnamurthy PT, Chintamaneni PK. Pharmacological targets of breast cancer stem cells: a review. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:463-479. [PMID: 29476201 DOI: 10.1007/s00210-018-1479-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 02/13/2018] [Indexed: 02/07/2023]
Abstract
Breast cancers contain small population of tumor-initiating cells called breast cancer stem cells (BCSCs), which are spared even after chemotherapy. Recently, BCSCs are implicated to be a cause of metastasis, tumor relapse, and therapy resistance in breast cancer. BCSCs have unique molecular mechanisms, which can be targeted to eliminate them. These include surface biomarkers, proteins involved in self-renewal pathways, drug efflux transporters, apoptotic/antiapoptotic proteins, autophagy, metabolism, and microenvironment regulation. The complex molecular mechanisms behind the survival of BCSCs and pharmacological targets for elimination of BCSCs are described in this review.
Collapse
Affiliation(s)
- Sai Kiran S S Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India.
| | - Pavan Kumar Chintamaneni
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India
| |
Collapse
|
162
|
Wang H, Stoecklein NH, Lin PP, Gires O. Circulating and disseminated tumor cells: diagnostic tools and therapeutic targets in motion. Oncotarget 2018; 8:1884-1912. [PMID: 27683128 PMCID: PMC5352105 DOI: 10.18632/oncotarget.12242] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/20/2016] [Indexed: 12/16/2022] Open
Abstract
Enumeration of circulating tumor cells (CTCs) in peripheral blood with the gold standard CellSearchTM has proven prognostic value for tumor recurrence and progression of metastatic disease. Therefore, the further molecular characterization of isolated CTCs might have clinical relevance as liquid biopsy for therapeutic decision-making and to monitor disease progression. The direct analysis of systemic cancer appears particularly important in view of the known disparity in expression of therapeutic targets as well as epithelial-to-mesenchymal transition (EMT)-based heterogeneity between primary and systemic tumor cells, which all substantially complicate monitoring and therapeutic targeting at present. Since CTCs are the potential precursor cells of metastasis, their in-depth molecular profiling should also provide a useful resource for target discovery. The present review will discuss the use of systemically spread cancer cells as liquid biopsy and focus on potential target antigens.
Collapse
Affiliation(s)
- Hongxia Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Nikolas H Stoecklein
- Department of General, Visceral and Pediatric Surgery, Medical Faculty, University Hospital of the Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | - Olivier Gires
- Department of Otorhinolaryngology, Head and Neck Surgery, Grosshadern Medical Center, Ludwig-Maximilians-University of Munich, Munich, Germany.,Clinical Cooperation Group Personalized Radiotherapy of Head and Neck Tumors, Helmholtz, Germany
| |
Collapse
|
163
|
Cytometric Profiling of CD133+ Cells in Human Colon Carcinoma Cell Lines Identifies a Common core Phenotype and Cell Type-specific Mosaics. Int J Biol Markers 2018; 28:267-73. [DOI: 10.5301/jbm.5000020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2013] [Indexed: 01/06/2023]
Abstract
In colorectal cancer, CD133+ cells from fresh biopsies proved to be more tumorigenic than their CD133– counterparts. Nevertheless, the function of CD133 protein in tumorigenic cells seems only marginal. Moreover, CD133 expression alone is insufficient to isolate true cancer stem cells, since only 1 out of 262 CD133+ cells actually displays stem-cell capacity. Thus, new markers for colorectal cancer stem cells are needed. Here, we show the extensive characterization of CD133+ cells in 5 different colon carcinoma continuous cell lines (HT29, HCT116, Caco2, GEO and LS174T), each representing a different maturation level of colorectal cancer cells. Markers associated with stemness, tumorigenesis and metastatic potential were selected. We identified 6 molecules consistently present on CD133+ cells: CD9, CD29, CD49b, CD59, CD151, and CD326. By contrast, CD24, CD26, CD54, CD66c, CD81, CD90, CD99, CD112, CD164, CD166, and CD200 showed a discontinuous behavior, which led us to identify cell type-specific surface antigen mosaics. Finally, some antigens, e.g. CD227, indicated the possibility of classifying the CD133+ cells into 2 subsets likely exhibiting specific features. This study reports, for the first time, an extended characterization of the CD133+ cells in colon carcinoma cell lines and provides a “dictionary” of antigens to be used in colorectal cancer research.
Collapse
|
164
|
Xu HL, Shen BX, Lin MT, Tong MQ, Zheng YW, Jiang X, Yang WG, Yuan JD, Yao Q, Zhao YZ. Homing of ICG-loaded liposome inlaid with tumor cellular membrane to the homologous xenografts glioma eradicates the primary focus and prevents lung metastases through phototherapy. Biomater Sci 2018; 6:2410-2425. [DOI: 10.1039/c8bm00604k] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Liposomes inlaid with tumor cellular membranes may serve as an excellent nanoplatform for homologous-targeting phototherapy using ICG.
Collapse
|
165
|
Gerlach JC, Foka HG, Thompson RL, Gridelli B, Schmelzer E. Epithelial cell adhesion molecule fragments and signaling in primary human liver cells. J Cell Physiol 2017; 233:4841-4851. [PMID: 29150960 DOI: 10.1002/jcp.26286] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/14/2017] [Indexed: 01/15/2023]
Abstract
Epithelial Cell Adhesion Molecule (EpCAM), or CD326, is a trans-membrane glycoprotein expressed by multiple normal epithelia as well as carcinoma. Human hepatic stem cells and bile duct epithelium of the liver are EpCAM positive. In tumor cell lines, its intracellular domain can be released after cleavage of the extracellular domain. Within the cell nucleus, it induces cell proliferation, but cleavage depends on cell contact. Fragments of various lengths have been described in tumor cells. Despite its described important role in proliferation in tumor cells, there is not much known about the expression and role of EpCAM fragments in primary human liver cells. Here, we demonstrate that EpCAM protein fragments and function are considerable different between tumor cells, normal fetal and adult liver cells. Contrary to previously reported findings in tumor cells, gene knockdown or treatment with an inhibitor of the cleavage enzyme ADAM17 (TACE) rather increased cell numbers in primary human fetal liver-derived EpCAM-positive cells. EpCAM fragment sizes were not affected by treatment with inhibitor. Knockdown of EPCAM gene expression by siRNA in sorted cells did not significantly affect proliferation-associated genes or cell numbers. The intracellular domain could not be detected within cell nuclei of fetal and adult liver cells. In conclusion, signaling through the intracellular domain of EpCAM appears to be a mechanism that induces proliferation specifically in tumorigenic cells but not in normal primary EpCAM-positive liver cells.
Collapse
Affiliation(s)
- Jörg C Gerlach
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hubert G Foka
- University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert L Thompson
- University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bruno Gridelli
- University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Surgery, ISMETT-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione, UPMC Italy, Palermo, Italy
| | - Eva Schmelzer
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
166
|
Izumi S, Yamamura S, Hayashi N, Toma M, Tawa K. Dual-Color Fluorescence Imaging of EpCAM and EGFR in Breast Cancer Cells with a Bull's Eye-Type Plasmonic Chip. SENSORS 2017; 17:s17122942. [PMID: 29257118 PMCID: PMC5751630 DOI: 10.3390/s17122942] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/11/2017] [Accepted: 12/15/2017] [Indexed: 01/05/2023]
Abstract
Surface plasmon field-enhanced fluorescence microscopic observation of a live breast cancer cell was performed with a plasmonic chip. Two cell lines, MDA-MB-231 and Michigan Cancer Foundation-7 (MCF-7), were selected as breast cancer cells, with two kinds of membrane protein, epithelial cell adhesion molecule (EpCAM) and epidermal growth factor receptor (EGFR), observed in both cells. The membrane proteins are surface markers used to differentiate and classify breast cancer cells. EGFR and EpCAM were detected with Alexa Fluor® 488-labeled anti-EGFR antibody (488-EGFR) and allophycocyanin (APC)-labeled anti-EpCAM antibody (APC-EpCAM), respectively. In MDA-MB231 cells, three-fold plus or minus one and seven-fold plus or minus two brighter fluorescence of 488-EGFR were observed on the 480-nm pitch and the 400-nm pitch compared with that on a glass slide. Results show the 400-nm pitch is useful. Dual-color fluorescence of 488-EGFR and APC-EpCAM in MDA-MB231 was clearly observed with seven-fold plus or minus two and nine-fold plus or minus three, respectively, on the 400-nm pitch pattern of a plasmonic chip. Therefore, the 400-nm pitch contributed to the dual-color fluorescence enhancement for these wavelengths. An optimal grating pitch of a plasmonic chip improved a fluorescence image of membrane proteins with the help of the surface plasmon-enhanced field.
Collapse
Affiliation(s)
- Shota Izumi
- School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda, Hyōgo 669-1337, Japan.
| | - Shohei Yamamura
- Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14, Hayashi-cho, Takamatsu, Kagawa 761-0395, Japan.
| | - Naoko Hayashi
- Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14, Hayashi-cho, Takamatsu, Kagawa 761-0395, Japan.
| | - Mana Toma
- School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda, Hyōgo 669-1337, Japan.
| | - Keiko Tawa
- School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda, Hyōgo 669-1337, Japan.
| |
Collapse
|
167
|
Alshaer W, Ababneh N, Hatmal M, Izmirli H, Choukeife M, Shraim A, Sharar N, Abu-Shiekah A, Odeh F, Al Bawab A, Awidi A, Ismail S. Selection and targeting of EpCAM protein by ssDNA aptamer. PLoS One 2017; 12:e0189558. [PMID: 29245156 PMCID: PMC5731996 DOI: 10.1371/journal.pone.0189558] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023] Open
Abstract
Aptamers are molecules that reveal highly complex and refined molecular recognition properties. These molecules are capable of binding with high affinity and selectivity to targets, ranging from small molecules to whole living cells. Several aptamers have been selected for targeting cellular proteins and they have also used in developing therapeutics and diagnostic strategies. Epithelial cell adhesion molecule (EpCAM) is considered as a cancer stem cell (CSC) biomarker and one of the most promising targets for aptamer selection against CSCs. In this study, we have developed a ssDNA aptamer with high affinity and selectivity of targeting the EpCAM protein extracellular domain. The SELEX technique was applied and the resulted sequences were tested on EpCAM-positive human gastric cancer cell line, KATO III, and the EpCAM-negative mouse embryonic fibroblast, NIH/3T3 cells. Ep1 aptamer was successfully isolated and showed selective binding on EpCAM-positive KATO III cells when compared to EpCAM-negative NIH/3T3 cells, as observed by the flow cytometry and the confocal imaging results. Additionally, the binding of Ep1 to EpCAM protein was assessed using mobility shifting assay and aptamers-protein docking. Furthermore, the binding affinity of Ep1 was measured against EpCAM protein using EpCAM-immobilized on magnetic beads and showed apparent affinity of 118 nM. The results of this study could suggest that Ep1 aptamer can bind specifically to the cellular EpCAM protein, making it an attractive ligand for targeted drug delivery and as an imaging agent for the identification of cancer cells.
Collapse
Affiliation(s)
- Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Molecular Biology Research Laboratory, Faculty of Medicine, University of Jordan, Amma, Jordan
- * E-mail: (WA); (AA)
| | - Nida Ababneh
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Molecular Biology Research Laboratory, Faculty of Medicine, University of Jordan, Amma, Jordan
| | - Mamon Hatmal
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, Zarqa, Jordan
| | - Heba Izmirli
- Molecular Biology Research Laboratory, Faculty of Medicine, University of Jordan, Amma, Jordan
| | - Moujab Choukeife
- Molecular Biology Research Laboratory, Faculty of Medicine, University of Jordan, Amma, Jordan
| | - Alaa Shraim
- Molecular Biology Research Laboratory, Faculty of Medicine, University of Jordan, Amma, Jordan
| | - Nour Sharar
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Aya Abu-Shiekah
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Fadwa Odeh
- Department of Chemistry, School of Science, The University of Jordan, Amman, Jordan
| | - Abeer Al Bawab
- Department of Chemistry, School of Science, The University of Jordan, Amman, Jordan
- HMCSR, The University of Jordan, Amman, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- * E-mail: (WA); (AA)
| | - Said Ismail
- Molecular Biology Research Laboratory, Faculty of Medicine, University of Jordan, Amma, Jordan
| |
Collapse
|
168
|
Škovierová H, Okajčeková T, Strnádel J, Vidomanová E, Halašová E. Molecular regulation of epithelial-to-mesenchymal transition in tumorigenesis (Review). Int J Mol Med 2017; 41:1187-1200. [PMID: 29286071 PMCID: PMC5819928 DOI: 10.3892/ijmm.2017.3320] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 11/23/2017] [Indexed: 12/13/2022] Open
Abstract
Numerous studies over the past two decades have focused on the epithelial-to-mesenchymal transition (EMT) and its role in the development of metastasis. Certain studies highlighted the importance of EMT in the dissemination of tumor cells and metastasis of epithelium-derived carcinomas. Tumor metastasis is a multistep process during which tumor cells change their morphology, and start to migrate and invade distant sites. The present review discusses the current understanding of the molecular mechanisms contributing to EMT in embryogenesis, fibrosis and tumorigenesis. Additionally, the signaling pathways that initiate EMT through transcriptional factors responsible for the activation and suppression of various genes associated with cancer cell migration were investigated. Furthermore, the important role of the epigenetic modifications that regulate EMT and the reverse process, mesenchymal-to-epithelial transition (MET) are discussed. MicroRNAs are key regulators of various intracellular processes and current knowledge of EMT has significantly improved due to microRNA characterization. Their effect on signaling pathways and the ensuing events that occur during EMT at the molecular level is becoming increasingly recognized. The current review also highlights the role of circulating tumor cells (CTCs) and CTC clusters, and their ability to form metastases. In addition, the biological properties of different types of circulating cells based on their tumor-forming potential are compared.
Collapse
Affiliation(s)
- Henrieta Škovierová
- Biomedical Center Martin, Department of Molecular Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 03601, Slovakia
| | - Terézia Okajčeková
- Biomedical Center Martin, Department of Molecular Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 03601, Slovakia
| | - Ján Strnádel
- Biomedical Center Martin, Department of Molecular Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 03601, Slovakia
| | - Eva Vidomanová
- Biomedical Center Martin, Department of Molecular Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 03601, Slovakia
| | - Erika Halašová
- Biomedical Center Martin, Department of Molecular Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 03601, Slovakia
| |
Collapse
|
169
|
Hong R, Zhou Y, Tian X, Wang L, Wu X. Selective inhibition of IDO1, D-1-methyl-tryptophan (D-1MT), effectively increased EpCAM/CD3-bispecific BiTE antibody MT110 efficacy against IDO1 hibreast cancer via enhancing immune cells activity. Int Immunopharmacol 2017; 54:118-124. [PMID: 29128855 DOI: 10.1016/j.intimp.2017.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/27/2017] [Accepted: 10/06/2017] [Indexed: 12/16/2022]
Abstract
MuS110 and MT110 are BiTE antibodies bispecific for CD3 and EpCAM, which is the most frequently and highly expressed tumor-associated antigen on breast cancer. And pronounced expression of IDO1 has also been reported in breast cancer. Our study aimed to investigate whether IDO1 inhibitor D-1MT combing with MuS110/MT110 had synergistic antitumor effects on IDO expressing EpCAM-positive breast cancer cells in vitro and in vivo. Data suggested that the expression of IDO1 on Epcam-positive breast cancer 4T1 and MCF-7 decreased MuS110/MT110 antitumor efficacy by the suppression of T cells activation in vitro. Combining D-1MT with MT110 in IDO+MCF-7 cells, or with MuS110 in IDO+4T1 cells, significantly improved the antitumor efficacy of BiTE antibodies via increasing T cell cytotoxicity and contributing to cytokines releasing. In vivo assay, combination of D-1MT with MT110 in NOD/SCID mice bearing IDOhi MCF-7 xenografts or MuS110 in immune competent BALB/c mice bearing IDOhi 4T1 xenografts suggested the similar synergistic effect. Together, IDO inhibition could reverse the suppression of T cells due to IDO expressing on breast cancer, and improve the antitumor efficacy of EpCAM/CD3-bispecific BiTE antibody.
Collapse
Affiliation(s)
- Ri Hong
- Maternal and Child Health Hospital of Sanya, Sanya, Hainan 572000, China.
| | - Yuhai Zhou
- Maternal and Child Health Hospital of Sanya, Sanya, Hainan 572000, China
| | - Xiujuan Tian
- Maternal and Child Health Hospital of Sanya, Sanya, Hainan 572000, China
| | - Lijuan Wang
- Maternal and Child Health Hospital of Sanya, Sanya, Hainan 572000, China
| | - Xiaoyun Wu
- Maternal and Child Health Hospital of Sanya, Sanya, Hainan 572000, China
| |
Collapse
|
170
|
Valedkarimi Z, Nasiri H, Aghebati-Maleki L, Majidi J. Antibody-cytokine fusion proteins for improving efficacy and safety of cancer therapy. Biomed Pharmacother 2017; 95:731-742. [PMID: 28888210 DOI: 10.1016/j.biopha.2017.07.160] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/25/2017] [Accepted: 07/30/2017] [Indexed: 12/23/2022] Open
|
171
|
Zhang Y, Zhou N, Yu X, Zhang X, Li S, Lei Z, Hu R, Li H, Mao Y, Wang X, Zhang J, Li Y, Guo H, Irwin DM, Niu G, Tan H. Tumacrophage: macrophages transformed into tumor stem-like cells by virulent genetic material from tumor cells. Oncotarget 2017; 8:82326-82343. [PMID: 29137267 PMCID: PMC5669893 DOI: 10.18632/oncotarget.19320] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/20/2017] [Indexed: 02/06/2023] Open
Abstract
Tumor-associated macrophages are regarded as tumor-enhancers as they have key roles in the subversion of adaptive immunity and in inflammatory circuits that promote tumor progression. Here, we show that cancer cells can subvert macrophages yielding cells that have gained pro-tumor functions. When macrophages isolated from mice or humans are co-cultured with dead cancer cell line cells, induced to undergo apoptosis to mimic chemotherapy, up-regulation of pro-tumor gene expression was identified. Phagocytosis of apoptotic cancer cells by macrophages resulted in their transformation into tumor stem (initiating)-like cells, as indicated by the expression of epithelial markers (e.g., cytokeratin) and stem cell markers (e.g., Oct4) and their capability to differentiate in vitro and self-renew in serum-free media. Moreover, we identified a subset of monocytes/macrophages cells in the blood of cancer (breast, ovarian and colorectal) patients undergoing chemotherapy that harbor tumor transcripts. Our findings uncover a new role for macrophages in tumor development, where they can be transformed into tumor-like cells, potentially by horizontal gene transfer of tumor-derived genes, thus, by taking advantage of chemotherapy, these transformed macrophages promote tumor metastasis by escaping immune surveillance.
Collapse
Affiliation(s)
- Yizhuang Zhang
- Department of Pharmacology, Peking University, Beijing, China
| | - Na Zhou
- Department of Pharmacology, Peking University, Beijing, China
| | - Xiuyan Yu
- Department of Pharmacology, Peking University, Beijing, China
| | - Xuehui Zhang
- Department of Pharmacology, Peking University, Beijing, China
| | - Shanxin Li
- Department of Pharmacology, Peking University, Beijing, China
| | - Zhen Lei
- N & N Genetech Company, Ltd., Beijing, China
| | - Ruobi Hu
- Department of Pharmacology, Peking University, Beijing, China
| | - Hui Li
- Department of Pharmacology, Peking University, Beijing, China
| | - Yiqing Mao
- Department of Pharmacology, Peking University, Beijing, China
| | - Xi Wang
- Department of Pharmacology, Peking University, Beijing, China
| | - Jinshu Zhang
- Department of Clinical Laboratory, The 305 Hospital of People’s Liberation Army, Beijing, China
| | - Yuan Li
- Department of Gynaecology and Obstetrics, Peking University Third Hospital, Beijing, China
| | - Hongyan Guo
- Department of Gynaecology and Obstetrics, Peking University Third Hospital, Beijing, China
| | - David M. Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Gang Niu
- N & N Genetech Company, Ltd., Beijing, China
| | - Huanran Tan
- Department of Pharmacology, Peking University, Beijing, China
| |
Collapse
|
172
|
Narain A, Asawa S, Chhabria V, Patil-Sen Y. Cell membrane coated nanoparticles: next-generation therapeutics. Nanomedicine (Lond) 2017; 12:2677-2692. [PMID: 28965474 DOI: 10.2217/nnm-2017-0225] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cell membrane coated nanoparticles (NPs) is a biomimetic strategy developed to engineer therapeutic devices consisting of a NP core coated with membrane derived from natural cells such as erythrocytes, white blood cells, cancer cells, stem cells, platelets or bacterial cells. These biomimetic NPs have gained a lot of attention recently owing to their cell surface mimetic features and tailored nanomaterial characteristics. They have shown strong potential in diagnostic and therapeutic applications including those in drug delivery, immune modulation, vaccination and detoxification. Herein we review the various types of cell membrane coated NPs reported in the literature and the unique strengths of these biomimetic NPs with an emphasis on how these bioinspired camouflage strategies have led to improved therapeutic efficacy. We also highlight the recent progress made by each platform in advancing healthcare and precis the major challenges associated with these NPs.
Collapse
Affiliation(s)
- Ashwin Narain
- Department of Biotechnology, National Institute of Technology, Warangal - 506004, TS, India
| | - Simran Asawa
- Department of Biotechnology, National Institute of Technology, Warangal - 506004, TS, India.,Warsaw University of Life Sciences, Warsaw, Poland
| | - Vikesh Chhabria
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston, UK
| | - Yogita Patil-Sen
- School of Physical Sciences & Computing, University of Central Lancashire, Preston, UK
| |
Collapse
|
173
|
Prasanna P, Patel J, Partovi S, Madabhushi A, Tiwari P. Radiomic features from the peritumoral brain parenchyma on treatment-naïve multi-parametric MR imaging predict long versus short-term survival in glioblastoma multiforme: Preliminary findings. Eur Radiol 2017; 27:4188-4197. [PMID: 27778090 PMCID: PMC5403632 DOI: 10.1007/s00330-016-4637-3] [Citation(s) in RCA: 195] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/28/2016] [Accepted: 10/05/2016] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Despite 90 % of glioblastoma (GBM) recurrences occurring in the peritumoral brain zone (PBZ), its contribution in patient survival is poorly understood. The current study leverages computerized texture (i.e. radiomic) analysis to evaluate the efficacy of PBZ features from pre-operative MRI in predicting long- (>18 months) versus short-term (<7 months) survival in GBM. METHODS Sixty-five patient examinations (29 short-term, 36 long-term) with gadolinium-contrast T1w, FLAIR and T2w sequences from the Cancer Imaging Archive were employed. An expert manually segmented each study as: enhancing lesion, PBZ and tumour necrosis. 402 radiomic features (capturing co-occurrence, grey-level dependence and directional gradients) were obtained for each region. Evaluation was performed using threefold cross-validation, such that a subset of studies was used to select the most predictive features, and the remaining subset was used to evaluate their efficacy in predicting survival. RESULTS A subset of ten radiomic 'peritumoral' MRI features, suggestive of intensity heterogeneity and textural patterns, was found to be predictive of survival (p = 1.47 × 10-5) as compared to features from enhancing tumour, necrotic regions and known clinical factors. CONCLUSION Our preliminary analysis suggests that radiomic features from the PBZ on routine pre-operative MRI may be predictive of long- versus short-term survival in GBM. KEY POINTS • Radiomic features from peritumoral regions can capture glioblastoma heterogeneity to predict outcome. • Peritumoral radiomics along with clinical factors are highly predictive of glioblastoma outcome. • Identifying prognostic markers can assist in making personalized therapy decisions in glioblastoma.
Collapse
Affiliation(s)
- Prateek Prasanna
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Jay Patel
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Sasan Partovi
- Case Western Reserve School of Medicine, University Hospitals Case Medical Center, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Pallavi Tiwari
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
174
|
Jenkins SV, Nima ZA, Vang KB, Kannarpady G, Nedosekin DA, Zharov VP, Griffin RJ, Biris AS, Dings RPM. Triple-negative breast cancer targeting and killing by EpCAM-directed, plasmonically active nanodrug systems. NPJ Precis Oncol 2017; 1:27. [PMID: 29872709 PMCID: PMC5871908 DOI: 10.1038/s41698-017-0030-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 06/29/2017] [Accepted: 07/19/2017] [Indexed: 11/16/2022] Open
Abstract
An ongoing need for new cancer therapeutics exists, especially ones that specifically home and target triple-negative breast cancer. Because triple-negative breast cancer express low or are devoid of estrogen, progesterone, or Her2/Neu receptors, another target must be used for advanced drug delivery strategies. Here, we engineered a nanodrug delivery system consisting of silver-coated gold nanorods (AuNR/Ag) targeting epithelial cell adhesion/activating molecule (EpCAM) and loaded with doxorubicin. This nanodrug system, AuNR/Ag/Dox-EpCAM, was found to specifically target EpCAM-expressing tumors compared to low EpCAM-expressing tumors. Namely, the nanodrug had an effective dose (ED50) of 3 μM in inhibiting 4T1 cell viability and an ED50 of 110 μM for MDA-MD-231 cells. Flow cytometry data indicated that 4T1 cells, on average, express two orders of magnitude more EpCAM than MDA-MD-231 cells, which correlates with our ED50 findings. Moreover, due to the silver coating, the AuNR/Ag can be detected simultaneously by surface-enhanced Raman spectroscopy and photoacoustic microscopy. Analysis by these imaging detection techniques as well as by inductively coupled plasma mass spectrometry showed that the targeted nanodrug system was taken up by EpCAM-expressing cells and tumors at significantly higher rates than untargeted nanoparticles (p < 0.05). Thus, this approach establishes a plasmonically active nanodrug theranostic for triple-negative breast cancer and, potentially, a delivery platform with improved multimodal imaging capability for other clinically relevant chemotherapeutics with dose-limiting toxicities, such as platinum-based or taxane-based therapies. Silver-coated gold nanorods deliver drugs to a difficult-to-treat breast cancer by targeting an over-expressed antigen on its surface. Ruud Dings and colleagues at the University of Arkansas in the USA loaded the chemotherapeutic drug doxorubicin onto silver-coated gold nanorods that were conjugated with an antibody that specifically targets an over-expressed antigen on many types of ‘triple-negative breast cancers’ (TNBCs). Unlike other breast cancers, TNBCs lack certain receptors, making them difficult to target with cancer therapies. The team found that one of the two TNBC cell lines studied over-expressed the epithelial antigen EpCAM 100 times more than the other. Their drug-loaded silver-coated gold nanorods specifically targeted the EpCAM over-expressing cells over the low-expressing ones. The nanorods’ coatings also allowed them to be easily detected by two different imaging techniques: surfaced-enhanced Raman spectroscopy and photoacoustic microscopy.
Collapse
Affiliation(s)
- Samir V Jenkins
- 1Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Zeid A Nima
- 2Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR USA
| | - Kieng B Vang
- 2Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR USA
| | - Ganesh Kannarpady
- 2Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR USA
| | - Dmitry A Nedosekin
- 3The Phillips Classic Laser and Nanomedicine Laboratories, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Vladimir P Zharov
- 3The Phillips Classic Laser and Nanomedicine Laboratories, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Robert J Griffin
- 1Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Alexandru S Biris
- 2Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR USA
| | - Ruud P M Dings
- 1Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| |
Collapse
|
175
|
Pindiprolu SKSS, Krishnamurthy PT, Chintamaneni PK, Karri VVSR. Nanocarrier based approaches for targeting breast cancer stem cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:885-898. [PMID: 28826237 DOI: 10.1080/21691401.2017.1366337] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Breast cancer stem cells (BCSCs) are heterogeneous subpopulation of tumour initiating cells within breast tumours. They are spared even after chemotherapy and responsible for tumour relapse. Targeting BCSCs is, therefore, necessary to achieve radical cure in breast cancer. Despite the availability of agents targeting BCSCs, their clinical application is limited due to their off-target effects and bioavailability issues. Nanotechnology based drug carriers (nanocarriers) offer various advantages to deliver anti-BCSCs agents specifically to their target sites by overcoming their bioavailability issues. In this review, we describe various strategies for targeting BCSCs using nanocarriers.
Collapse
Affiliation(s)
- Sai Kiran S S Pindiprolu
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Praveen T Krishnamurthy
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Pavan Kumar Chintamaneni
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Veera Venkata Satyanarayana Reddy Karri
- b Department of Pharmaceutics , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| |
Collapse
|
176
|
Fournier C, Martin F, Zitvogel L, Kroemer G, Galluzzi L, Apetoh L. Trial Watch: Adoptively transferred cells for anticancer immunotherapy. Oncoimmunology 2017; 6:e1363139. [PMID: 29147628 DOI: 10.1080/2162402x.2017.1363139] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 07/29/2017] [Accepted: 07/30/2017] [Indexed: 12/22/2022] Open
Abstract
Immunotherapies aimed at strengthening immune effector responses against malignant cells are growing at exponential rates. Alongside, the impressive benefits obtained by patients with advanced melanoma who received adoptively transferred tumor-infiltrating lymphocytes (TILs) have encouraged the scientific community to pursue adoptive cell transfer (ACT)-based immunotherapy. ACT involves autologous or allogenic effector lymphocytes that are generally obtained from the peripheral blood or resected tumors, expanded and activated ex vivo, and administered to lymphodepleted patients. ACT may be optionally associated with chemo- and/or immunotherapeutics, with the overall aim of enhancing the proliferation, persistence and functionality of infused cells, as well as to ensure their evolution in an immunological permissive local and systemic microenvironment. In addition, isolated lymphocytes can be genetically engineered to endow them with the ability to target a specific tumor-associated antigen (TAA), to increase their lifespan, and/or to reduce their potential toxicity. The infusion of chimeric antigen receptor (CAR)-expressing cytotoxic T lymphocytes redirected against CD19 has shown promising clinical efficacy in patients with B-cell malignancies. Accordingly, the US Food and Drug Administration (FDA) has recently granted 'breakthrough therapy' designation to a CAR-based T-cell therapy (CTL019) for patients with B-cell malignancies. Considerable efforts are now being devoted to the development of efficient ACT-based immunotherapies for non-hematological neoplasms. In this Trial Watch, we summarize recent clinical advances on the use of ACT for oncological indications.
Collapse
Affiliation(s)
- Carole Fournier
- INSERM, U1231, Dijon, France.,Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France
| | - François Martin
- INSERM, U1231, Dijon, France.,Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lionel Apetoh
- INSERM, U1231, Dijon, France.,Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| |
Collapse
|
177
|
Nima ZA, Alwbari AM, Dantuluri V, Hamzah RN, Sra N, Motwani P, Arnaoutakis K, Levy RA, Bohliqa AF, Nedosekin D, Zharov VP, Makhoul I, Biris AS. Targeting nano drug delivery to cancer cells using tunable, multi-layer, silver-decorated gold nanorods. J Appl Toxicol 2017; 37:1370-1378. [PMID: 28730725 DOI: 10.1002/jat.3495] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 05/14/2017] [Accepted: 05/16/2017] [Indexed: 12/21/2022]
Abstract
Multifunctional nanoparticles have high potential as targeting delivery vehicles for cancer chemotherapy. In this study, silver-decorated gold nanorods (AuNR\Ag) have been successfully used to deliver specific, targeted chemotherapy against breast cancer (MCF7) and prostate carcinoma (PC3) cell lines. Doxorubicin, a commonly used chemotherapy, and anti-Epithelial cell adhesion molecule (anti-EpCAM) antibodies were covalently bonded to thiolated polyethylene glycol-coated AuNR\Ag, and the resultant system was used to deliver the drugs to cancer cells in vitro. Furthermore, these nanoparticles have a unique spectral signature by surface enhanced Raman spectroscopy (SERS), which enables reliable detection and monitoring of the distribution of these chemotherapy constructs inside cells. The development of interest in a plasmonic nano drugs system with unique spectroscopic signatures could result in a clinical approach to the precise targeting and visualization of cells and solid tumors while delivering molecules for the enhanced treatment of cancerous tumors.
Collapse
Affiliation(s)
- Zeid A Nima
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, 72204, USA
| | - Ahmed M Alwbari
- Department of Cancer Care, Johns Hopkins Aramco Healthcare, Dhahran, 34465, Saudi Arabia.,University of Arkansas for Medical Sciences, Department of Internal Medicine, Division of Hematology/Oncology, Little Rock, Arkansas, 72205, USA
| | - Vijayalakshmi Dantuluri
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, 72204, USA
| | - Rabab N Hamzah
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, 72204, USA
| | - Natasha Sra
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, 72204, USA
| | - Pooja Motwani
- University of Arkansas for Medical Sciences, Department of Internal Medicine, Division of Hematology/Oncology, Little Rock, Arkansas, 72205, USA
| | - Konstantinos Arnaoutakis
- University of Arkansas for Medical Sciences, Department of Internal Medicine, Division of Hematology/Oncology, Little Rock, Arkansas, 72205, USA
| | - Rebecca A Levy
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA
| | - Amani F Bohliqa
- Maternity and Children's Hospital, Department of Pharmacy, Damam, 32253, Saudi Arabia
| | - Dmitry Nedosekin
- Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA
| | - Vladimir P Zharov
- Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA
| | - Issam Makhoul
- University of Arkansas for Medical Sciences, Department of Internal Medicine, Division of Hematology/Oncology, Little Rock, Arkansas, 72205, USA
| | - Alexandru S Biris
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, 72204, USA
| |
Collapse
|
178
|
Chen S, El-Heliebi A, Kroneis T. Biological and Molecular Characterization of Circulating Tumor Cells: A Creative Strategy for Precision Medicine? Adv Clin Chem 2017; 82:71-103. [PMID: 28939214 DOI: 10.1016/bs.acc.2017.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Circulating tumor cells (CTCs) are a group of rare cells disseminated from either primary or metastatic tumors into the blood stream. CTCs are considered to be the precursor of cancer metastasis. As a critical component of liquid biopsies, CTCs are a unique tool to understand the formation of metastasis and a valuable source of information on intratumor heterogeneity. Much effort has been invested in technologies for the detection of CTCs because they are rare cells among the vast number of blood cells. Studies in various cancers have repeatedly demonstrated that increased CTC counts prior to or during treatment are significantly associated with poor outcomes. In the new era of precision medicine, the study of CTCs reaches far beyond detection and counting. The rapidly growing field of analytical platforms for rare-cell analysis allows in-depth characterization of CTCs at the bulk cell and single-cell level. Genetic profiling of CTCs may provide an insight into the real-time tumor status, may allow the monitoring and evaluation of treatment response in clinical routine, and may lead to the development of novel therapeutic targets as well.
Collapse
Affiliation(s)
- Shukun Chen
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Graz, Austria.
| | - Amin El-Heliebi
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Graz, Austria
| | - Thomas Kroneis
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Graz, Austria
| |
Collapse
|
179
|
A Novel Three-Dimensional Platform to Investigate Neoangiogenesis, Transendothelial Migration, and Metastasis of MDAMB-231 Breast Cancer Cells. Plast Reconstr Surg 2017; 138:472e-482e. [PMID: 27556622 DOI: 10.1097/prs.0000000000002470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND A crucial step in the progression of cancer involves the transendothelial migration of tumor cells into the bloodstream and invasion at distant sites. Most in vitro models of malignant cell behavior do not account for the presence of and interaction with vascular cells. Three-dimensional platforms to further explore the factors responsible for metastatic cellular behavior are under intensive investigation. METHODS Hydrogels with encapsulated MDAMB-231 breast cancer cells were fabricated with a central microchannel. The microchannel was lined with a co-culture of human umbilical vein endothelial cells and human aortic smooth muscle cells. For comparison, co-culture-seeded microchannels without breast cancer cells (MDAMB-negative) were fabricated. RESULTS After 7 and 14 days, the endoluminal lining of encapsulated MDAMB-231 co-culture-seeded microchannels demonstrated aberrant endothelial cell and smooth muscle cell organization and breast cancer cell transendothelial migration. MDAMB-231 cells performed matrix remodeling, forming tumor aggregates within the bulk, migrating preferentially toward the hydrogel "neovessel." In contrast, MDAMB-negative constructs demonstrated maintenance of an intact endoluminal lining composed of endothelial cells and smooth muscle cells that organized into discrete layers. Furthermore, the thicknesses of the endoluminal lining of MDAMB-negative constructs were significantly greater than encapsulated MDAMB-231 co-culture-seeded constructs after 7 and 14 days (p = 0.012 and p < 0.001, respectively). CONCLUSION The authors have created a powerful tool that may have tremendous impact on furthering our understanding of cancer recurrence and metastasis, shedding light on these poorly understood phenomena.
Collapse
|
180
|
|
181
|
Qu W, Meng B, Yu Y, Wang S. EpCAM antibody-conjugated mesoporous silica nanoparticles to enhance the anticancer efficacy of carboplatin in retinoblastoma. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:646-651. [DOI: 10.1016/j.msec.2017.03.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/12/2017] [Accepted: 03/04/2017] [Indexed: 11/26/2022]
|
182
|
Kang JH, Driscoll H, Mammoto A, Watters AL, Melakeberhan B, Diaz A, Super M, Ingber DE. An Engineered Human Fc‐Mannose‐Binding‐Lectin Captures Circulating Tumor Cells. ACTA ACUST UNITED AC 2017; 1:e1700094. [DOI: 10.1002/adbi.201700094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Joo H. Kang
- Wyss Institute for Biologically Inspired Engineering Harvard University CLSB5, 3 Blackfan Circle Boston MA 02115 USA
- Department of Biomedical Engineering School of Life Sciences Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 South Korea
| | - Harry Driscoll
- Wyss Institute for Biologically Inspired Engineering Harvard University CLSB5, 3 Blackfan Circle Boston MA 02115 USA
| | - Akiko Mammoto
- Vascular Biology Program Boston Children's Hospital and Harvard Medical School Boston MA 02115 USA
| | - Alexander L. Watters
- Wyss Institute for Biologically Inspired Engineering Harvard University CLSB5, 3 Blackfan Circle Boston MA 02115 USA
| | - Bissrat Melakeberhan
- Wyss Institute for Biologically Inspired Engineering Harvard University CLSB5, 3 Blackfan Circle Boston MA 02115 USA
| | - Alexander Diaz
- Wyss Institute for Biologically Inspired Engineering Harvard University CLSB5, 3 Blackfan Circle Boston MA 02115 USA
| | - Michael Super
- Wyss Institute for Biologically Inspired Engineering Harvard University CLSB5, 3 Blackfan Circle Boston MA 02115 USA
| | - Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering Harvard University CLSB5, 3 Blackfan Circle Boston MA 02115 USA
- Vascular Biology Program Boston Children's Hospital and Harvard Medical School Boston MA 02115 USA
- Harvard John A. Paulson School of Engineering and Applied Sciences Cambridge MA 02139 USA
| |
Collapse
|
183
|
Li T, Wang Z, Hou YF, Li YY. Pim-3 Regulates Stemness of Pancreatic Cancer Cells via Activating STAT3 Signaling Pathway. J Cancer 2017; 8:1530-1541. [PMID: 28775772 PMCID: PMC5535708 DOI: 10.7150/jca.18628] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/27/2017] [Indexed: 02/06/2023] Open
Abstract
Due to its aggressiveness and unusual resistance to conventional therapies, pancreatic cancer is a highly lethal gastrointestinal malignancy with poor prognosis. According to the cancer stem cell hypothesis, there exists a fraction of cancer cells, that is, cancer stem cells, responsible for tumor maintenance and therapeutic failure. Herein we investigated the involvement of proto-oncogene Pim-3 in driving the stemness properties in pancreatic cancer. Expression levels of several stemness-associated markers were examined in several pancreatic cancer cell lines. The double positive (CD24+ESA+) and double negative (CD24-ESA-) pancreatic cancer cells were isolated from PANC-1 and L3.6pl, and their self-renewal ability, tumorigenicity as well as sensitivity to gemcitabine were then evaluated. Results showed that there existed heterogeneity in expression levels of stemness-associated surface markers among pancreatic cancer cell lines. CD24+ESA+ pancreatic cancer cells exhibited increased tumorigenicity and decreased chemosensitivity to gemcitabine as compared to CD24-ESA- cells. Besides, the double positive (CD24+ESA+) subpopulation also exhibited greater expression level of Pim-3 when compared with the double negative (CD24-ESA-) ones. Furthermore, silencing of Pim-3 in pancreatic cancer cells leads to decreased proportions of both single positive (CD24+ and ESA+) and double positive (CD24+ESA+) pancreatic cancer cells. Overexpression of Pim-3 was associated with increased levels of some stemness-associated transcription factors (STAT3, etc.). Moreover, the phosphorylation level and transcriptional activity of STAT3 were decreased in Pim-3 silenced pancreatic cancer cells and restoration of its activity results in restitution of stem cell-like phenotypes. Therefore, Pim-3 maintains stemness of pancreatic cancer cells via activating STAT3 signaling pathway and might be used as a novel therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Ting Li
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Wang
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Feng Hou
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying-Yi Li
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
184
|
Xie Z, Zeng X. DNA/RNA-based formulations for treatment of breast cancer. Expert Opin Drug Deliv 2017; 14:1379-1393. [DOI: 10.1080/17425247.2017.1317744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Zhaolu Xie
- Department of Pharmacy, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing, China
| | - Xianghui Zeng
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
185
|
Hu G, Xu Y, Chen W, Wang J, Zhao C, Wang M. RNA Interference of IQ Motif Containing GTPase-Activating Protein 3 (IQGAP3) Inhibits Cell Proliferation and Invasion in Breast Carcinoma Cells. Oncol Res 2017; 24:455-461. [PMID: 28281966 PMCID: PMC7838651 DOI: 10.3727/096504016x14685034103635] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Breast cancer is a highly prevalent disease affecting women. The association of IQ motif containing GTPase-activating protein 3 (IQGAP3) and breast cancer is poorly defined. Here we reported that IQGAP3 is a key regulator of cell proliferation and metastasis during breast cancer progression. The expression of IQGAP3 was significantly increased in breast tissues compared to nontumor tissues at both protein and mRNA levels. Furthermore, IQGAP3 had a high expression level in ZR-75-30 and BT474 compared to other breast cancer cell lines. Depletion of IQGAP3 through RNA interference in ZR-75-30 and BT474 significantly inhibited cell proliferation. More importantly, IQGAP3 silencing in breast cancer cells notably repressed cell migration and invasion. Further analysis suggested that inhibition of cell proliferation and metastasis was associated with some proteins, including p53, MMP9, Snail, CDC42, p-ERK1/2, KIF2C, KIF4A, PCNA, and Twist. Since expression of IQGAP3 seems to be associated with the pathogenesis of breast cancer and suppression of it can inhibit cancer cell growth and metastasis, IQGAP3 may be a potential therapeutic target in human breast cancer.
Collapse
Affiliation(s)
- Gaowu Hu
- Department of General Surgery, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai, P.R. China
| | | | | | | | | | | |
Collapse
|
186
|
Guo Y, Feng K, Wang Y, Han W. Targeting cancer stem cells by using chimeric antigen receptor-modified T cells: a potential and curable approach for cancer treatment. Protein Cell 2017; 9:516-526. [PMID: 28290053 PMCID: PMC5966354 DOI: 10.1007/s13238-017-0394-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/23/2017] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs), a subpopulation of tumor cells, have self-renewal and multi-lineage differentiation abilities that play an important role in cancer initiation, maintenance, and metastasis. An accumulation of evidence indicates that CSCs can cause conventional therapy failure and cancer recurrence because of their treatment resistance and self-regeneration characteristics. Therefore, approaches that specifically and efficiently eliminate CSCs to achieve a durable clinical response are urgently needed. Currently, treatments with chimeric antigen receptor-modified T (CART) cells have shown successful clinical outcomes in patients with hematologic malignancies, and their safety and feasibility in solid tumors was confirmed. In this review, we will discuss in detail the possibility that CART cells inhibit CSCs by specifically targeting their cell surface markers, which will ultimately improve the clinical response for patients with various types of cancer. A number of viewpoints were summarized to promote the application of CSC-targeted CART cells in clinical cancer treatment. This review covers the key aspects of CSC-targeted CART cells against cancers in accordance with the premise of the model, from bench to bedside and back to bench.
Collapse
Affiliation(s)
- Yelei Guo
- Molecular & Immunological Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Kaichao Feng
- Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yao Wang
- Molecular & Immunological Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Weidong Han
- Molecular & Immunological Department, Chinese PLA General Hospital, Beijing, 100853, China. .,Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
187
|
A double-negative feedback loop between EpCAM and ERK contributes to the regulation of epithelial-mesenchymal transition in cancer. Oncogene 2017; 36:3706-3717. [PMID: 28192403 DOI: 10.1038/onc.2016.504] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 10/17/2016] [Accepted: 12/05/2016] [Indexed: 12/13/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is an important biological process that has been implicated in cancer metastasis. Epithelial cell adhesion molecule (EpCAM) is expressed at the basolateral membrane of most normal epithelial cells but is overexpressed in many epithelial cancers. In our studies on the role of EpCAM in cancer biology, we observed that EpCAM expression is decreased in mesenchymal-like primary cancer specimens in vivo and following induction of EMT in cancer cell lines in vitro. Extracellular signal-related kinase (ERK) is a key regulator of EMT. We observed that EpCAM expression is decreased with activation of the ERK pathway in primary cancer specimens in vivo and in cancer cell lines in vitro. In experimental models, growth factor stimulation and/or oncogene-induced ERK2 activation suppressed EpCAM expression, whereas genetic or pharmacological inhibition of the ERK pathway restored EpCAM expression. In detailed studies of the EpCAM promoter region, we observed that ERK2 suppresses EpCAM transcription directly by binding to a consensus ERK2-binding site in the EpCAM promoter and indirectly through activation of EMT-associated transcription factors SNAI1, SNAI2, TWIST1 and ZEB1, which bind to E-box sites in the EpCAM promoter. Surprisingly, EpCAM appears to modulate ERK activity. Using multiple cell lines, we demonstrated that specific ablation of EpCAM resulted in increased ERK pathway activity and SNAI2 expression, migration and invasion, whereas forced expression of EpCAM resulted in decreased ERK pathway activity and SNAI2 expression, migration and invasion. These observations provide important insights into the regulation of EpCAM expression during EMT, demonstrate an unexpected role for EpCAM in the regulation of ERK and define a novel double-negative feedback loop between EpCAM and ERK that contributes to the regulation of EMT. These studies have important translational implications as both EpCAM and ERK are currently being targeted in human clinical trials.
Collapse
|
188
|
Zhong GX, Feng SD, Shen R, Wu ZY, Chen F, Zhu X. The clinical significance of the Ezrin gene and circulating tumor cells in osteosarcoma. Onco Targets Ther 2017; 10:527-533. [PMID: 28223819 PMCID: PMC5308564 DOI: 10.2147/ott.s125589] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose The aim of this study was to investigate the clinical significance of circulating tumor cells (CTCs) in the peripheral blood of an osteosarcoma and the Ezrin gene expressed in CTCs. Patients and methods CTC enrichment was done with CanPatrol™ CTC enrichment technique in 41 patients with osteosarcoma. The characterization of CTCs was performed using a multiple messenger RNA in situ analysis (MRIA). The expression of the Ezrin gene in CTCs was detected by RNA probe technology. The correlations of CTC counts, cell type and the expression level of the Ezrin gene with clinical stage and metastasis of osteosarcoma were analyzed using SPSS 16.0 software. Results The CTC counts correlated significantly with Enneking stage (P<0.001). The ratio of mesenchymal CTCs correlated with the distant metastases (P<0.001). Ezrin gene expression in CTCs correlated significantly with distant metastases (χ2=152.51, P=0.000). Conclusion The ratio of mesenchymal CTCs in the peripheral blood of osteosarcoma correlates with distant metastases. High expression of Ezrin gene in CTCs correlates with distant metastases.
Collapse
Affiliation(s)
| | - Shao-Dan Feng
- Department of Emergency, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | | | | | | | | |
Collapse
|
189
|
Wicaksono PA, Name S, Martien R, Ismail H. Formulation and Cytotoxicity of Ribosome-Inactivating Protein Mirabilis Jalapa L. Nanoparticles Using Alginate-Low Viscosity Chitosan Conjugated with Anti-Epcam Antibodies in the T47D Breast Cancer Cell Line. Asian Pac J Cancer Prev 2017; 17:2277-84. [PMID: 27221930 DOI: 10.7314/apjcp.2016.17.4.2277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Ribosome-inactivating protein (RIP) from Mirabilis jalapa L. leaves has cytotoxic effects on breast cancer cell lines but is less toxic towards normal cells. However, it can easily be degraded after administration so it needs to be formulated into nanoparticles to increase its resistance to enzymatic degradation. The objectives of this study were to develop a protein extract of M. jalapa L. leaves (RIP-MJ) incorporated into nanoparticles conjugated with Anti-EpCAM antibodies, and to determine its cytotoxicity and selectivity in the T47D breast cancer cell line. RIP-MJ was extracted from red-flowered M. jalapa L. leaves. Nanoparticles were formulated based on polyelectrolyte complexation using low viscosity chitosan and alginate, then chemically conjugated with anti-EpCAM antibody using EDAC based on carbodiimide reaction. RIP-MJ nanoparticles were characterised for the particle size, polydispersity index, zeta potential, particle morphology, and entrapment efficiency. The cytotoxicity of RIP-MJ nanoparticles against T47D and Vero cells was then determined with MTT assay. The optimal formula of RIP-MJ nanoparticles was obtained at the concentration of RIP-MJ, low viscosity chitosan and alginate respectively 0.05%, 1%, and 0.4% (m/v). RIP-MJ nanoparticles are hexagonal with high entrapment efficiency of 98.6%, average size of 130.7 nm, polydispersity index of 0.380 and zeta potential +26.33 mV. The IC50 values of both anti-EpCAM-conjugated and non-conjugated RIP-MJ nanoparticles for T47D cells (13.3 and 14.9 μg/mL) were lower than for Vero cells (27.8 and 33.6 μg/mL). The IC50 values of conjugated and non- conjugated RIP-MJ for both cells were much lower than IC50 values of non-formulated RIP-MJ (>500 μg/mL).
Collapse
|
190
|
Salim EI, Hegazi MM, Kang JS, Helmy HM. Expression Patterns of Cancer Stem Cell Markers During Specific Celecoxib Therapy in Multistep Rat Colon Carcinogenesis Bioassays. Asian Pac J Cancer Prev 2017; 17:1023-35. [PMID: 27039721 DOI: 10.7314/apjcp.2016.17.3.1023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The purpose of this study was to investigate the role of colon cancer stem cells (CSCs) during chemicallyinduced rat multi-step colon carcinogenesis with or without the treatment with a specific cyclooxygenase-2 inhibitor drug (celecoxib). Two experiments were performed, the first, a short term 12 week colon carcinogenesis bioassay in which only surrogate markers for colon cancer, aberrant crypt foci (ACF) lesions, were formed. The other experiment was a medium term colon cancer rat assay in which tumors had developed after 32 weeks. Treatment with celecoxib lowered the numbers of ACF, as well as the tumor volumes and multiplicities after 32 weeks. Immunohistochemical proliferating cell nuclear antigen (PCNA) labeling indexes LI (%) were downregulated after treatment by celecoxib. Also different cell surface antigens known to associate with CSCs such as the epithelial cell adhesion molecule (EpCAM), CD44 and CD133 were compared between the two experiments and showed differential expression patterns depending on the stage of carcinogenesis and treatment with celecoxib. Flow cytometric analysis demonstrated that the numbers of CD133 cells were increased in the colonic epithelium after 12 weeks while those of CD44 but not CD133 cells were increased after 32 weeks. Moreover, aldehyde dehydrogenase-1 activity levels in the colonic epithelium (a known CSC marker) detected by ELISA assay were found down-regulated after 12 weeks, but were up-regulated after 32 weeks. The data have also shown that the protective effect of celecoxib on these specific markers and populations of CSCs and on other molecular processes such as apoptosis targeted by this drug may vary depending on the genetic and phenotypic stages of carcinogenesis. Therefore, uncovering these distinction roles of CSCs during different phases of carcinogenesis and during specific treatment could be useful for targeted therapy.
Collapse
Affiliation(s)
- Elsayed I Salim
- Department of Zoology, Faculty of Science, Tanta University, Research Lab. of Molecular Carcinogenesis, Tanta, Egypt E-mail : ,
| | | | | | | |
Collapse
|
191
|
Chaudhuri PK, Ebrahimi Warkiani M, Jing T, Kenry, Lim CT. Microfluidics for research and applications in oncology. Analyst 2017; 141:504-24. [PMID: 26010996 DOI: 10.1039/c5an00382b] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cancer is currently one of the top non-communicable human diseases, and continual research and developmental efforts are being made to better understand and manage this disease. More recently, with the improved understanding in cancer biology as well as the advancements made in microtechnology and rapid prototyping, microfluidics is increasingly being explored and even validated for use in the detection, diagnosis and treatment of cancer. With inherent advantages such as small sample volume, high sensitivity and fast processing time, microfluidics is well-positioned to serve as a promising platform for applications in oncology. In this review, we look at the recent advances in the use of microfluidics, from basic research such as understanding cancer cell phenotypes as well as metastatic behaviors to applications such as the detection, diagnosis, prognosis and drug screening. We then conclude with a future outlook on this promising technology.
Collapse
Affiliation(s)
| | - Majid Ebrahimi Warkiani
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602 and School of Mechanical and Manufacturing Engineering, Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Tengyang Jing
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602 and Department of Biomedical Engineering, National University of Singapore, Singapore 117575.
| | - Kenry
- Department of Biomedical Engineering, National University of Singapore, Singapore 117575. and NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore 117411 and BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602
| |
Collapse
|
192
|
Optical Aptamer Probes of Fluorescent Imaging to Rapid Monitoring of Circulating Tumor Cell. SENSORS 2016; 16:s16111909. [PMID: 27886058 PMCID: PMC5134568 DOI: 10.3390/s16111909] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/02/2016] [Accepted: 11/02/2016] [Indexed: 02/08/2023]
Abstract
Fluorescence detecting of exogenous EpCAM (epithelial cell adhesion molecule) or muc1 (mucin1) expression correlated to cancer metastasis using nanoparticles provides pivotal information on CTC (circulating tumor cell) occurrence in a noninvasive tool. In this study, we study a new skill to detect extracellular EpCAM/muc1 using quantum dot-based aptamer beacon (QD-EpCAM/muc1 ALB (aptamer linker beacon). The QD-EpCAM/muc1 ALB was designed using QDs (quantum dots) and probe. The EpCAM/muc1-targeting aptamer contains a Ep-CAM/muc1 binding sequence and BHQ1 (black hole quencher 1) or BHQ2 (black hole quencher2). In the absence of target EpCAM/muc1, the QD-EpCAM/muc1 ALB forms a partial duplex loop-like aptamer beacon and remained in quenched state because the BHQ1/2 quenches the fluorescence signal-on of the QD-EpCAM/muc1 ALB. The binding of EpCAM/muc1 of CTC to the EpCAM/muc1 binding aptamer sequence of the EpCAM/muc1-targeting oligonucleotide triggered the dissociation of the BHQ1/2 quencher and subsequent signal-on of a green/red fluorescence signal. Furthermore, acute inflammation was stimulated by trigger such as caerulein in vivo, which resulted in increased fluorescent signal of the cy5.5-EpCAM/muc1 ALB during cancer metastasis due to exogenous expression of EpCAM/muc1 in Panc02-implanted mouse model.
Collapse
|
193
|
Chen Z, Zhao P, Luo Z, Zheng M, Tian H, Gong P, Gao G, Pan H, Liu L, Ma A, Cui H, Ma Y, Cai L. Cancer Cell Membrane-Biomimetic Nanoparticles for Homologous-Targeting Dual-Modal Imaging and Photothermal Therapy. ACS NANO 2016; 10:10049-10057. [PMID: 27934074 DOI: 10.1021/acsnano.6b04695] [Citation(s) in RCA: 611] [Impact Index Per Article: 67.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
An active cell membrane-camouflaged nanoparticle, owning to membrane antigens and membrane structure, can achieve special properties such as specific recognition, long blood circulation, and immune escaping. Herein, we reported a cancer cell membrane-cloaked nanoparticle system as a theranostic nanoplatform. The biomimetic nanoparticles (indocyanine green (ICG)-loaded and cancer cell membrane-coated nanoparticles, ICNPs) exhibit a core-shell nanostructure consisting of an ICG-polymeric core and cancer cell membrane shell. ICNPs demonstrated specific homologous targeting to cancer cells with good monodispersity, preferable photothermal response, and excellent fluorescence/photoacoustic (FL/PA) imaging properties. Benefited from the functionalization of the homologous binding adhesion molecules from cancer cell membranes, ICNPs significantly promoted cell endocytosis and homologous-targeting tumor accumulation in vivo. Moreover, ICNPs were also good at disguising as cells to decrease interception by the liver and kidney. Through near-infrared (NIR)-FL/PA dual-modal imaging, ICNPs could realize real-time monitored in vivo dynamic distribution with high spatial resolution and deep penetration. Under NIR laser irradiation, ICNPs exhibited highly efficient photothermal therapy to eradicate xenografted tumor. The robust ICNPs with homologous properties of cancer cell membranes can serve as a bionic nanoplatform for cancer-targeted imaging and phototherapy.
Collapse
Affiliation(s)
- Ze Chen
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
| | - Pengfei Zhao
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
| | - Zhenyu Luo
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
- University of Chinese Academy of Sciences , Beijing 100049, People's Republic of China
| | - Mingbin Zheng
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University , Dongguan 523808, People's Republic of China
| | - Hao Tian
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University , Dongguan 523808, People's Republic of China
| | - Ping Gong
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
| | - Guanhui Gao
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
- University of Chinese Academy of Sciences , Beijing 100049, People's Republic of China
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
| | - Aiqing Ma
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University , Dongguan 523808, People's Republic of China
| | - Haodong Cui
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
| | - Yifan Ma
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, People's Republic of China
| |
Collapse
|
194
|
van Driel PBAA, Boonstra MC, Prevoo HAJM, van de Giessen M, Snoeks TJA, Tummers QRJG, Keereweer S, Cordfunke RA, Fish A, van Eendenburg JDH, Lelieveldt BPF, Dijkstra J, van de Velde CJH, Kuppen PJK, Vahrmeijer AL, Löwik CWGM, Sier CFM. EpCAM as multi-tumour target for near-infrared fluorescence guided surgery. BMC Cancer 2016; 16:884. [PMID: 27842504 PMCID: PMC5109830 DOI: 10.1186/s12885-016-2932-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 10/30/2016] [Indexed: 01/08/2023] Open
Abstract
Background Evaluation of resection margins during cancer surgery can be challenging, often resulting in incomplete tumour removal. Fluorescence-guided surgery (FGS) aims to aid the surgeon to visualize tumours and resection margins during surgery. FGS relies on a clinically applicable imaging system in combination with a specific tumour-targeting contrast agent. In this study EpCAM (epithelial cell adhesion molecule) is evaluated as target for FGS in combination with the novel Artemis imaging system. Methods The NIR fluorophore IRDye800CW was conjugated to the well-established EpCAM specific monoclonal antibody 323/A3 and an isotype IgG1 as control. The anti-EpCAM/800CW conjugate was stable in serum and showed preserved binding capacity as evaluated on EpCAM positive and negative cell lines, using flow cytometry and cell-based plate assays. Four clinically relevant orthotopic tumour models, i.e. colorectal cancer, breast cancer, head and neck cancer, and peritonitis carcinomatosa, were used to evaluate the performance of the anti-EpCAM agent with the clinically validated Artemis imaging system. The Pearl Impulse small animal imaging system was used as reference. The specificity of the NIRF signal was confirmed using bioluminescence imaging and green-fluorescent protein. Results All tumour types could clearly be delineated and resected 72 h after injection of the imaging agent. Using NIRF imaging millimetre sized tumour nodules were detected that were invisible for the naked eye. Fluorescence microscopy demonstrated the distribution and tumour specificity of the anti-EpCAM agent. Conclusions This study shows the potential of an EpCAM specific NIR-fluorescent agent in combination with a clinically validated intraoperative imaging system to visualize various tumours during surgery.
Collapse
Affiliation(s)
- P B A A van Driel
- Department of Radiology, Division of Molecular Imaging, Leiden University Medical Centre, Leiden, Netherlands.,Percuros BV, Enschede, The Netherlands
| | - M C Boonstra
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - H A J M Prevoo
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - M van de Giessen
- Department of Radiology and Division of Image Processing, Leiden University Medical Centre, Leiden, Netherlands
| | - T J A Snoeks
- Department of Radiology, Division of Molecular Imaging, Leiden University Medical Centre, Leiden, Netherlands
| | - Q R J G Tummers
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - S Keereweer
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus Medical Centre, Rotterdam, Netherlands
| | - R A Cordfunke
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, Netherlands
| | - A Fish
- Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - B P F Lelieveldt
- Department of Radiology and Division of Image Processing, Leiden University Medical Centre, Leiden, Netherlands
| | - J Dijkstra
- Department of Radiology and Division of Image Processing, Leiden University Medical Centre, Leiden, Netherlands
| | - C J H van de Velde
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - P J K Kuppen
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands.,Antibodies for Research Applications BV, Gouda, The Netherlands
| | - A L Vahrmeijer
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - C W G M Löwik
- Department of Radiology, Division of Molecular Imaging, Leiden University Medical Centre, Leiden, Netherlands
| | - C F M Sier
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands. .,Antibodies for Research Applications BV, Gouda, The Netherlands.
| |
Collapse
|
195
|
Tawa K, Yamamura S, Sasakawa C, Shibata I, Kataoka M. Sensitive Detection of Cell Surface Membrane Proteins in Living Breast Cancer Cells Using Multicolor Fluorescence Microscopy with a Plasmonic Chip. ACS APPLIED MATERIALS & INTERFACES 2016; 8:29893-29898. [PMID: 27750430 DOI: 10.1021/acsami.6b07777] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A plasmonic chip was applied to live cancer cell imaging. The epithelial cell adhesion molecule (EpCAM) is a surface marker that can be used to classify breast cancer cell lines into distinct differentiation states. EpCAM and the nuclei of two kinds of living breast cancer cells, MDA-MB231 and MCF-7, were stained with allophycocyanin (APC)-labeled anti-EpCAM antibody and 4',6-diamidino-2-phenylindole (DAPI), respectively, and the cells were scattered on either a plasmonic chip (metal-coated wavelength-scale grating substrate) or a control glass slide. Multicolor fluorescence microscopic imaging allowed fluorescence images of APC-EpCAM to be obtained on the plasmonic chip that were more than 10 times brighter compared with those on the glass slide. In contrast, in the fluorescence images of DAPI-stained nuclei, no difference in brightness was observed between substrates. The fluorescence enhancement of APC-EpCAM in the cell membrane in contact with the plasmonic chip is thought to be due to the excitation of APC molecules localized within the surface plasmon field. Analysis of the cross section of a fluorescence image revealed a distribution of EpCAM at a higher level of fluorescence in the center of the cell image because of contact between the cell membrane and the plasmonic chip. In contrast, fluorescence images of APC-EpCAM taken on a glass slide were so dark that only the outline of the cell was characterized. The plasmonic chip thus constitutes a simple and powerful tool for analyzing the distribution and kinetics of surface marker proteins in cell membranes contacting the chip.
Collapse
Affiliation(s)
- Keiko Tawa
- Kwansei Gakuin University , 2-1 Gakuen, Sanda, Hyogo 669-1337, Japan
- AIST , 1-8-31 Midorigaoka, Ikeda, Osaka 563-8377, Japan
| | - Shohei Yamamura
- AIST , 2217-14 Hayashi-cho, Takamatsu, Kagawa 761-0395, Japan
| | - Chisato Sasakawa
- Kwansei Gakuin University , 2-1 Gakuen, Sanda, Hyogo 669-1337, Japan
- AIST , 1-8-31 Midorigaoka, Ikeda, Osaka 563-8377, Japan
| | - Izumi Shibata
- AIST , 2217-14 Hayashi-cho, Takamatsu, Kagawa 761-0395, Japan
| | | |
Collapse
|
196
|
Klinger M, Benjamin J, Kischel R, Stienen S, Zugmaier G. Harnessing T cells to fight cancer with BiTE® antibody constructs--past developments and future directions. Immunol Rev 2016; 270:193-208. [PMID: 26864113 DOI: 10.1111/imr.12393] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bispecific T-cell engager (BiTE(®)) antibody constructs represent a novel immunotherapy that bridges cytotoxic T cells to tumor cells, thereby inducing target cell-dependent polyclonal T-cell activation and proliferation, and leading to apoptosis of bound tumor cells. Anti-CD19 BiTE(®) blinatumomab has demonstrated clinical activity in Philadelphia chromosome (Ph)-negative relapsed or refractory (r/r) acute lymphoblastic leukemia (ALL) eventually resulting in conditional approval by the U.S. Food and Drug Administration in 2014. This drug is currently further developed in pediatric and Ph(+) r/r, as well as in minimal residual disease-positive ALL, and might also offer clinical benefit for patients with non-Hodgkin's lymphoma, especially for those with aggressive forms like diffuse large B-cell lymphoma. Another BiTE(®) antibody construct in hemato-oncology designated AMG 330 targets CD33 on acute myeloid leukemia blast cells. After showing promising ex vivo activity, this drug candidate has recently entered phase 1 clinical development, and has further indicated potential for combination with checkpoint inhibitors. In solid tumor indications, three BiTE(®) antibody constructs have been tested in phase 1 studies so far: anti-EpCAM BiTE(®) AMG 110, anti-CEA BiTE(®) MEDI-565/AMG 211, and anti-PSMA BiTE(®) BAY2010112/AMG 212. Pertinent questions comprise how to maximize BiTE(®) penetration and T-cell infiltration of the tumor while simultaneously minimizing any adverse events, which is currently explored by a continuous intravenous infusion approach. Thus, BiTE(®) antibody constructs will hopefully provide new treatment options for patients in several indications with high unmet medical need.
Collapse
|
197
|
Lin SC, Chou YT, Jiang SS, Chang JL, Chung CH, Kao YR, Chang IS, Wu CW. Epigenetic Switch between SOX2 and SOX9 Regulates Cancer Cell Plasticity. Cancer Res 2016; 76:7036-7048. [DOI: 10.1158/0008-5472.can-15-3178] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 09/22/2016] [Accepted: 09/25/2016] [Indexed: 11/16/2022]
|
198
|
Ströhlein MA, Heiss MM, Jauch KW. The current status of immunotherapy in peritoneal carcinomatosis. Expert Rev Anticancer Ther 2016; 16:1019-27. [PMID: 27530056 DOI: 10.1080/14737140.2016.1224666] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Peritoneal carcinomatosis (PC) is a cancer disease with an urgent need for effective treatment. Conventional chemotherapy failed to show acceptable results. Cytoreductive surgery and hyperthermic chemoperfusion (HIPEC) are only beneficial in few patients with resectable peritoneal metastasis. Immunotherapy could be attractive against PC, as all requirements for immunotherapy are available in the peritoneal cavity. AREAS COVERED This review analyzes the present literature for immunotherapy of PC. Advances from immune stimulators, radionucleotide-conjugated- and bispecific antibodies to future developments like adoptive engineered T-cells with chimeric receptors are discussed. The clinical development of catumaxomab, which was the first intraperitoneal immunotherapy to be approved for clinical treatment, is discussed. The requirements for future developments are illustrated. Expert commentary: Immunotherapy of peritoneal carcinomatosis is manageable, showing striking cancer cell killing. Improved profiles of adverse events by therapy-induced cytokine release, enhanced specific killing and optimal treatment schedules within multimodal treatment will be key factors.
Collapse
Affiliation(s)
- Michael Alfred Ströhlein
- a Department of Abdominal, Vascular and Transplant Surgery, Cologne Merheim Medical Center , Witten/Herdecke University , Cologne , Germany
| | - Markus Maria Heiss
- a Department of Abdominal, Vascular and Transplant Surgery, Cologne Merheim Medical Center , Witten/Herdecke University , Cologne , Germany
| | - Karl-Walter Jauch
- b Medical Center of the Ludwig Maximilian University Munich , Munich , Germany
| |
Collapse
|
199
|
Gawlik-Rzemieniewska N, Galilejczyk A, Krawczyk M, Bednarek I. Silencing expression of the NANOG gene and changes in migration and metastasis of urinary bladder cancer cells. Arch Med Sci 2016; 12:889-97. [PMID: 27478472 PMCID: PMC4947613 DOI: 10.5114/aoms.2015.55368] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/01/2015] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION It has been proved that expression of the NANOG gene is observed not only in embryonic-derived malignancies, but also in breast cancer, ovarian cancer, cervix cancer and bladder cancer. NANOG overexpression is correlated with high activity of MMP-2 and MMP-9. The aim of the study was to evaluate the changes in the malignant phenotype of T24 bladder cancer cells with modulated expression of the NANOG gene. MATERIAL AND METHODS Human urinary bladder cancer cells T24 (HTB-4) were cultivated under standard conditions. Transfection of the cells with silencing constructions was performed with the application of Lipofectamine 2000 (Invitrogen) reagent. Evaluation of changes in the expression level of individual genes was performed using qRTPCR. Changes in the protein level were evaluated using the Human ELISA Kit (Abcam). The invasion capability of transfected cells was tested using Matrigel Invasion Chambers (BD Biosciences). The changes in cell migration were assessed with a wound-healing assay. RESULTS The qRTPCR evaluation showed that silencing the NANOG gene in T24 cells led to the decrease of mRNA for the MMP-2 gene to the level of 62.4% and the MMP-9 gene to the level of 76%. The cells with modulated expression of the NANOG gene migrated slower in the Matrigel invasion assay and in the wound-healing assay. The immunoenzymatic test showed a decrease in the protein level of MMP-9. CONCLUSIONS The transcriptional activity of the NANOG gene might be connected with some aspects of bladder cancer cell metastasis in vitro and has an influence on MMP-2 and MMP-9 expression levels.
Collapse
Affiliation(s)
- Natalia Gawlik-Rzemieniewska
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| | - Anna Galilejczyk
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| | - Michał Krawczyk
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| | - Ilona Bednarek
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
200
|
Detection of soluble EpCAM (sEpCAM) in malignant ascites predicts poor overall survival in patients treated with catumaxomab. Oncotarget 2016; 6:25017-23. [PMID: 26296970 PMCID: PMC4694811 DOI: 10.18632/oncotarget.4496] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 06/29/2015] [Indexed: 01/15/2023] Open
Abstract
EpCAM is an attractive target for cancer therapy and the EpCAM-specific antibody catumaxomab has been used for intraperitoneal treatment of EpCAM-positive cancer patients with malignant ascites. New prognostic markers are necessary to select patients that mostly benefit from catumaxomab. Recent data showed that soluble EpCAM (sEpCAM) is capable to block the effect of catumaxomab in vitro. This exploratory retrospective analysis was performed on archived ascites samples to evaluate the predictive role of sEpCAM in catumaxomab-treated patients. Sixty-six catumaxomab-treated patients with an available archived ascites sample were included in this study and tested for sEpCAM by sandwich ELISA. All probes were sampled before treatment start and all patients received at least one catumaxomab infusion. Overall survival, puncture-free survival and time to next puncture were compared between sEpCAM-positive and -negative patients. We detected sEpCAM in ascites samples of 9 patients (13.6%). These patients showed a significantly shorter overall survival. The prognostic significance of sEpCAM in ascites was particularly strong in patients with ovarian cancer. Puncture-free survival and time to next puncture were not significantly different between sEpCAM-positive and -negative patients. We propose sEpCAM in malignant ascites as a potential predictive marker in cancer patients treated with catumaxomab. Prospective studies with larger patients samples are urgently needed to confirm these findings and studies testing dose-intensified catumaxomab in patients with sEpCAM-positive ascites should be envisaged.
Collapse
|