51
|
Kalasin S, Browne E, Arcaro K, Santore MM. Selective Adhesive Cell Capture without Molecular Specificity: New Surfaces Exploiting Nanoscopic Polycationic Features as Discrete Adhesive Units. RSC Adv 2017; 7:13416-13425. [PMID: 28989702 PMCID: PMC5628748 DOI: 10.1039/c7ra01217a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This work explored how molecularly non-specific polycationic nanoscale features on a collecting surface control kinetic and selectivity aspects of mammalian cell capture. Key principles for selective collector design were demonstrated by comparing the capture of two closely related breast cancer cell lines: MCF-7 and TMX2-28. TMX2-28 is a tamoxifen-selected clone of MCF-7. The collector was a silica surface, negatively-charged at pH 7.4, containing isolated molecules (~ 8 nm diameter) of the cationic polymer, poly(dimethyl-aminoethylmethacrylate), pDMAEMA. Important in this work is the non-selective nature of the pDMAEMA interactions with cells: pDMAEMA generally adheres negatively charged particles and cells in solution. We show here that selectivity towards cells results from collector design: this includes competition between repulsive interactions involving the negative silica and attractions to the immobilized pDMAEMA molecules, the random pDMAEMA arrangement on the surface, and the concentration of positive charge in the vicinity of the adsorbed pDMAEMA chains. The latter act as nanoscopic cationic surface patches, each weakly attracted to negatively-charged cells. Collecting surfaces engineered with an appropriate amount pDMAEMA, exposed to mixtures of MCF-7 and TMX2-28 cells preferentially captured TMX2-28 with a selectivity of 2.5. (This means that the ratio of TMX2-28 to MCF cells on the surface was 2.5 times their compositional ratio in free solution.) The ionic strength-dependence of cell capture was shown to be similar to that of silica microparticles on the same surfaces. This suggests that the mechanism of selective cell capture involves nanoscopic differences in the contact areas of the cells with the collector, allowing discrimination of closely related cell line-based small scale features of the cell surface. This work demonstrated that even without molecular specificity, selectivity for physical cell attributes produces adhesive discrimination.
Collapse
Affiliation(s)
- S. Kalasin
- Department of Polymer Science and Engineering, 120 Governors Drive, Amherst, MA 01003
| | - E.P. Browne
- Department of Veterinary and Animal Science, 240 Thatcher Road, Amherst, MA 01003
| | - K.F. Arcaro
- Department of Veterinary and Animal Science, 240 Thatcher Road, Amherst, MA 01003
| | - M. M. Santore
- Department of Polymer Science and Engineering, 120 Governors Drive, Amherst, MA 01003
| |
Collapse
|
52
|
Pizzoni S, Sabattini S, Stefanello D, Dentini A, Ferrari R, Dacasto M, Giantin M, Laganga P, Amati M, Tortorella G, Marconato L. Features and prognostic impact of distant metastases in 45 dogs with de novo stage IV cutaneous mast cell tumours: A prospective study. Vet Comp Oncol 2017; 16:28-36. [DOI: 10.1111/vco.12306] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 01/05/2017] [Accepted: 01/19/2017] [Indexed: 12/31/2022]
Affiliation(s)
- S. Pizzoni
- Centro Oncologico Veterinario; Bologna Italy
| | - S. Sabattini
- Department of Veterinary Medical Sciences; University of Bologna; Bologna Italy
| | - D. Stefanello
- Department of Veterinary Medicine; University of Milan; Milan Italy
| | | | - R. Ferrari
- Department of Veterinary Medicine; University of Milan; Milan Italy
| | - M. Dacasto
- Department of Comparative Biomedicine and Food Science; University of Padua; Padua Italy
| | - M. Giantin
- Department of Comparative Biomedicine and Food Science; University of Padua; Padua Italy
| | - P. Laganga
- Centro Oncologico Veterinario; Bologna Italy
| | - M. Amati
- Ospedale Veterinario Città di Pavia; Pavia Italy
| | | | | |
Collapse
|
53
|
Cheng M, Liu L, Yang HS, Liu GF. Circulating tumor cells are associated with bone metastasis of lung cancer. Asian Pac J Cancer Prev 2017; 15:6369-74. [PMID: 25124627 DOI: 10.7314/apjcp.2014.15.15.6369] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Lung cancer (LC) is the leading cause of cancer mortality worldwide, predominantly due to the difficulty of early diagnosis and its high metastatic potential. Recently, increasing evidence suggests that circulating tumour cells (CTCs) are responsible for cancer metastatic relapse, and CTCs have attracted interest in cancer metastasis detection and quantification. In present study, we collected blood samples from 67 patients with bone metastasis, and 30 patients without such metastasis, and searched for CTCs. Then the association of CTC numbers with bone metastasis and other clinico-pothological variants was analyzed. Results demonstrated that when 5 or 1 was taken as a threshhold for the CTC number, there were significantly higher positivity of CTCs in the bone metastasis group than in the non-metastasis group. While the increase in CTC number was not significantly associated with any other clinicopathological factor, including age, gender, pathological type, intrapulmonary metastasis and lymph node metastasis, the CTC number in patients with positivity of the last above mentioned variants was obviously higher than in patients with negativity of the two variants. Taken together, the CTC number appears to be significantly associated with the bone metastasis from lung cancer.
Collapse
Affiliation(s)
- Min Cheng
- Department of Radiology, China-Japan Union Hospital, Jilin University, Changchun, China E-mail : ,
| | | | | | | |
Collapse
|
54
|
Naftali O, Maman S, Meshel T, Sagi-Assif O, Ginat R, Witz IP. PHOX2B is a suppressor of neuroblastoma metastasis. Oncotarget 2016; 7:10627-37. [PMID: 26840262 PMCID: PMC4891146 DOI: 10.18632/oncotarget.7056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/23/2016] [Indexed: 12/27/2022] Open
Abstract
Paired like homeobox 2B (PHOX2B) is a minimal residual disease (MRD) marker of neuroblastoma. The presence of MRD, also referred to as micro-metastases, is a powerful marker of poor prognosis in neuroblastoma. Lung metastasis is considered a terminal event in neuroblastoma. Lung micro-metastatic neuroblastoma (MicroNB) cells show high expression levels of PHOX2B and possess a less malignant and metastatic phenotype than lung macro metastatic neuroblastoma (MacroNB) cells, which hardly express PHOX2B. In vitro assays showed that PHOX2B knockdown in MicroNB cells did not affect cell viability; however it decreased the migratory capacity of the MicroNB-shPHOX2B cells. An orthotopic inoculation of MicroNB-shPHOX2B cells into the adrenal gland of nude mice resulted in significantly larger primary tumors and a heavier micro-metastatic load in the lungs and bone-marrow, than when control cells were inoculated. PHOX2B expression was found to be regulated by methylation. The PHOX2B promoter in MacroNB cells is significantly more methylated than in MicroNB cells. Demethylation assays using 5-azacytidine demonstrated that methylation can indeed inhibit PHOX2B transcription in MacroNB cells. These pre-clinical data strongly suggest that PHOX2B functions as a suppressor of neuroblastoma progression.
Collapse
Affiliation(s)
- Osnat Naftali
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel 69978
| | - Shelly Maman
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel 69978
| | - Tsipi Meshel
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel 69978
| | - Orit Sagi-Assif
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel 69978
| | - Ravit Ginat
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel 69978
| | - Isaac P Witz
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel 69978
| |
Collapse
|
55
|
Mu Z, Benali-Furet N, Uzan G, Znaty A, Ye Z, Paolillo C, Wang C, Austin L, Rossi G, Fortina P, Yang H, Cristofanilli M. Detection and Characterization of Circulating Tumor Associated Cells in Metastatic Breast Cancer. Int J Mol Sci 2016; 17:ijms17101665. [PMID: 27706044 PMCID: PMC5085698 DOI: 10.3390/ijms17101665] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 01/06/2023] Open
Abstract
The availability of blood-based diagnostic testing using a non-invasive technique holds promise for real-time monitoring of disease progression and treatment selection. Circulating tumor cells (CTCs) have been used as a prognostic biomarker for the metastatic breast cancer (MBC). The molecular characterization of CTCs is fundamental to the phenotypic identification of malignant cells and description of the relevant genetic alterations that may change according to disease progression and therapy resistance. However, the molecular characterization of CTCs remains a challenge because of the rarity and heterogeneity of CTCs and technological difficulties in the enrichment, isolation and molecular characterization of CTCs. In this pilot study, we evaluated circulating tumor associated cells in one blood draw by size exclusion technology and cytological analysis. Among 30 prospectively enrolled MBC patients, CTCs, circulating tumor cell clusters (CTC clusters), CTCs of epithelial-mesenchymal transition (EMT) and cancer associated macrophage-like cells (CAMLs) were detected and analyzed. For molecular characterization of CTCs, size-exclusion method for CTC enrichment was tested in combination with DEPArray™ technology, which allows the recovery of single CTCs or pools of CTCs as a pure CTC sample for mutation analysis. Genomic mutations of TP53 and ESR1 were analyzed by targeted sequencing on isolated 7 CTCs from a patient with MBC. The results of genomic analysis showed heterozygous TP53 R248W mutation from one single CTC and pools of three CTCs, and homozygous TP53 R248W mutation from one single CTC and pools of two CTCs. Wild-type ESR1 was detected in the same isolated CTCs. The results of this study reveal that size-exclusion method can be used to enrich and identify circulating tumor associated cells, and enriched CTCs were characterized for genetic alterations in MBC patients, respectively.
Collapse
Affiliation(s)
- Zhaomei Mu
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | - Zhong Ye
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Carmela Paolillo
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Chun Wang
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Laura Austin
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Giovanna Rossi
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Paolo Fortina
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
- Department of Molecular Medicine, University of Rome "Sapienza", Rome 00185, Italy.
| | - Hushan Yang
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Massimo Cristofanilli
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
56
|
Marulli G, Mammana M, Rea F. Impact of lymph node occult metastases in stage I non-small cell lung cancer (NSCLC): what is the evidence? J Thorac Dis 2016; 8:E809-12. [PMID: 27618780 DOI: 10.21037/jtd.2016.07.59] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Giuseppe Marulli
- Thoracic Surgery Unit, Department of Cardiologic, Thoracic and Vascular Sciences, University of Padua, Padova, Italy
| | - Marco Mammana
- Thoracic Surgery Unit, Department of Cardiologic, Thoracic and Vascular Sciences, University of Padua, Padova, Italy
| | - Federico Rea
- Thoracic Surgery Unit, Department of Cardiologic, Thoracic and Vascular Sciences, University of Padua, Padova, Italy
| |
Collapse
|
57
|
Are disseminated tumor cells in bone marrow and tumor-stroma ratio clinically applicable for patients undergoing surgical resection of primary colorectal cancer? The Leiden MRD study. Cell Oncol (Dordr) 2016; 39:537-544. [PMID: 27613548 PMCID: PMC5121180 DOI: 10.1007/s13402-016-0296-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2016] [Indexed: 02/07/2023] Open
Abstract
Purpose Current TNM staging does not appropriately identify high-risk colorectal cancer (CRC) patients. The aim of this study was to evaluate whether the presence of disseminated tumor cells (DTCs) in the bone marrow (BM) and the presence of stroma in the primary tumor, i.e., the tumor-stroma ratio (TSR), in patients undergoing surgical resection of primary CRC provides information relevant for disease outcome. Methods Patients with primary CRC (n = 125), consecutively admitted for curative resection between 2001 and 2007, were included in the study. All patients underwent BM aspiration before surgery. Detection of tumor cells was performed using immunocytochemical staining for cytokeratin (CK-ICC). The TSR was determined on diagnostic H&E stained sections of primary tumors. Results DTCs were detected in the BM of 23/125 patients (18 %). No association was found between BM status and overall survival (HR 0.97 (95 % CI 0.45–2.09), p = 0.93). Also, no significant difference was found in their 5-year survival rate (resp. 72 % and 68 % for BM-positive versus BM-negative patients). The TSR was found to be associated with a worse overall survival (HR 2.16, 95 % CI 1.02–4.57, p = 0.04) with 5-year survival rates of 84 % versus 62 % for stroma-low and stroma-high patients, respectively. No relation was found between the presence of DTCs and TSR. Conclusions Our data indicate that the presence of DTCs in the BM of CRC patients is not associated with disease outcome. The TSR was, however, found to be associated with a worse overall survival, which indicates that for CRC the tumor microenvironment plays an important role in its behavior and prognosis.
Collapse
|
58
|
Bliss SA, Sinha G, Sandiford OA, Williams LM, Engelberth DJ, Guiro K, Isenalumhe LL, Greco SJ, Ayer S, Bryan M, Kumar R, Ponzio NM, Rameshwar P. Mesenchymal Stem Cell-Derived Exosomes Stimulate Cycling Quiescence and Early Breast Cancer Dormancy in Bone Marrow. Cancer Res 2016; 76:5832-5844. [PMID: 27569215 DOI: 10.1158/0008-5472.can-16-1092] [Citation(s) in RCA: 293] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/27/2016] [Indexed: 12/13/2022]
Abstract
Dormant breast cancers resurge as metastatic disease after a long dormancy period in the bone marrow, where cancer cells interact with mesenchymal stem cells (MSC). However, the nature of early interactions between breast cancer cells and MSCs in the bone marrow microenvironment that facilitate adaptation to a quiescent state remains poorly understood. Here, we report that breast cancer cells prime MSC to release exosomes containing distinct miRNA contents, such as miR-222/223, which in turn promotes quiescence in a subset of cancer cells and confers drug resistance. Building on these results, we developed a novel, nontoxic therapeutic strategy to target dormant breast cancer cells based on systemic administration of MSC loaded with antagomiR-222/223. In an immunodeficient mouse model of dormant breast cancer, this therapy sensitized breast cancer cells to carboplatin-based therapy and increased host survival. Overall, our findings illuminate the nature of the regulatory interactions between breast cancer cells and MSCs in the evolution of tumor dormancy and resurgence in the micrometastatic microenvironment of the bone marrow. Cancer Res; 76(19); 5832-44. ©2016 AACR.
Collapse
Affiliation(s)
- Sarah A Bliss
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey. Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Garima Sinha
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey. Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Oleta A Sandiford
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Lisa M Williams
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey. Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Daniel J Engelberth
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey. Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Khadidiatou Guiro
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | | | - Steven J Greco
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Seda Ayer
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Margarette Bryan
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Rakesh Kumar
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC
| | - Nicholas M Ponzio
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Pranela Rameshwar
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey. Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Newark, New Jersey.
| |
Collapse
|
59
|
Shen F, Zhang Y, Jernigan DL, Feng X, Yan J, Garcia FU, Meucci O, Salvino JM, Fatatis A. Novel Small-Molecule CX3CR1 Antagonist Impairs Metastatic Seeding and Colonization of Breast Cancer Cells. Mol Cancer Res 2016; 14:518-27. [PMID: 27001765 PMCID: PMC5070649 DOI: 10.1158/1541-7786.mcr-16-0013] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/07/2016] [Indexed: 12/11/2022]
Abstract
UNLABELLED Recent evidence indicates that cancer cells, even in the absence of a primary tumor, recirculate from established secondary lesions to further seed and colonize skeleton and soft tissues, thus expanding metastatic dissemination and precipitating the clinical progression to terminal disease. Recently, we reported that breast cancer cells utilize the chemokine receptor CX3CR1 to exit the blood circulation and lodge to the skeleton of experimental animals. Now, we show that CX3CR1 is overexpressed in human breast tumors and skeletal metastases. To assess the clinical potential of targeting CX3CR1 in breast cancer, a functional role of CX3CR1 in metastatic seeding and progression was first validated using a neutralizing antibody for this receptor and transcriptional suppression by CRISPR interference (CRISPRi). Successively, we synthesized and characterized JMS-17-2, a potent and selective small-molecule antagonist of CX3CR1, which was used in preclinical animal models of seeding and established metastasis. Importantly, counteracting CX3CR1 activation impairs the lodging of circulating tumor cells to the skeleton and soft-tissue organs and also negatively affects further growth of established metastases. Furthermore, nine genes were identified that were similarly altered by JMS-17-2 and CRISPRi and could sustain CX3CR1 prometastatic activity. In conclusion, these data support the drug development of CX3CR1 antagonists, and promoting their clinical use will provide novel and effective tools to prevent or contain the progression of metastatic disease in breast cancer patients. IMPLICATIONS This work conclusively validates the instrumental role of CX3CR1 in the seeding of circulating cancer cells and is expected to pave the way for pairing novel inhibitors of this receptor with current standards of care for the treatment of breast cancer patients. Mol Cancer Res; 14(6); 518-27. ©2016 AACR.
Collapse
Affiliation(s)
- Fei Shen
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Yun Zhang
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Danielle L Jernigan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Xin Feng
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Jie Yan
- Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Fernando U Garcia
- Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Joseph M Salvino
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Alessandro Fatatis
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania. Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania. The Biology of Prostate Cancer Program, Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
60
|
Peng C, Chen H, Wallwiener M, Modugno C, Cuk K, Madhavan D, Trumpp A, Heil J, Marmé F, Nees J, Riethdorf S, Schott S, Sohn C, Pantel K, Schneeweiss A, Yang R, Burwinkel B. Plasma S100P level as a novel prognostic marker of metastatic breast cancer. Breast Cancer Res Treat 2016; 157:329-338. [PMID: 27146585 DOI: 10.1007/s10549-016-3776-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/30/2016] [Indexed: 01/09/2023]
Abstract
UNLABELLED Metastasis is the main cause of death in breast cancer patients. The development of reliable and cost-effective biomarker to evaluate the prognosis of metastatic breast cancer (MBC) patients is of great importance. S100P is a member of S100 family and has been proved to be associated with metastasis establishment. METHODS We investigated the plasma S100P levels in 60 healthy controls, 48 primary and 273 metastatic breast cancer patients. The MBC patients were followed-up for disease progression and death up to 3.5 years after recruitment. Radiographic response of MBC patients were also analyzed for investigation on treatment monitoring value of plasma S100P level. We found a robust association between high plasma S100P level (>7 ng/mL) and poor prognosis of metastatic breast cancer (MBC) patients (median progression-free survival time: 5.0 vs. 8.7 months, log-rank test p < 0.001; median overall survival time: 22.5 vs. 31.6 months, log-rank test p < 0.001). The plasma S100P level added additional prognostic relevance to the conventional prognostication model with clinicopathological factors and CTC enumeration. The plasma S100P level decreased significantly after treatment, while the reduction correlated with the radiographic response of the MBC patients. This finding indicates the value of plasma S100P in dynamic evaluation of treatment outcome. We hereby suggest plasma S100P level as a simple and cost-effective marker for the prognosis of metastatic breast cancer.
Collapse
Affiliation(s)
- Cike Peng
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Molecular Biology of Breast Cancer, Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany.
| | - Hongda Chen
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Markus Wallwiener
- Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Caroline Modugno
- Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Katarina Cuk
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany
| | - Dharanija Madhavan
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany
| | - Andreas Trumpp
- Hi-STEM-Heidelberg Institute for Stem Cell Technology and Experimental Medicine, GmbH, Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg Heil
- Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany
| | - Frederik Marmé
- Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany
| | - Juliane Nees
- Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Sabine Riethdorf
- Department of Tumor Biology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Schott
- Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany
| | - Christof Sohn
- Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Schneeweiss
- Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Rongxi Yang
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany.,Molecular Biology of Breast Cancer, University Women's Clinic University Heidelberg, ImNeuenheimer Feld 440, 69120, Heidelberg, Germany
| | - Barbara Burwinkel
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany.,Molecular Biology of Breast Cancer, University Women's Clinic University Heidelberg, ImNeuenheimer Feld 440, 69120, Heidelberg, Germany
| |
Collapse
|
61
|
Kim N, Cho SB, Park YL, Park SY, Myung E, Kim SH, Yu HM, Son YA, Myung DS, Lee WS, Joo YE. Effect of Recepteur d'Origine Nantais expression on chemosensitivity and tumor cell behavior in colorectal cancer. Oncol Rep 2016; 35:3331-40. [PMID: 27035413 DOI: 10.3892/or.2016.4721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/03/2015] [Indexed: 11/05/2022] Open
Abstract
Recepteur d'Origine Nantais (RON) expression is known to induce oncogenic properties including tumor cell growth, survival, motility, angiogenesis and chemoresistance. In the present study, we evaluated whether RON affects chemosensitivity and oncogenic behavior of colorectal cancer cells and investigated its prognostic value in colorectal cancer. To evaluate the impact of RON on chemosensitivity and tumor cell behavior, we treated colorectal cancer cells with small interfering RNAs specific to RON. This was followed by flow cytometric analyses and migration, Matrigel invasion and endothelial tube formation assays. The expression of RON was investigated by immunohistochemistry in colorectal cancer tissues. TUNEL assay and immunohistochemical staining for CD34 and D2-40 were deployed to determine apoptosis, angiogenesis and lymphangiogenesis. RON knockdown enhanced 5-fluorouracil (FU)-induced apoptosis by upregulating the activities of caspases and expression of proapoptotic genes. Moreover, it enhanced 5-FU-induced cell cycle arrest by decreasing the expression of cyclins and cyclin‑dependent kinases and inducing that of p21. Furthermore, RON knockdown augmented the 5-FU-induced inhibition of invasion and migration of colorectal cancer cells. The β-catenin signaling cascade was blocked by RON knockdown upon 5-FU treatment. RON knockdown also decreased endothelial tube formation and expression of VEGF-A and HIF-1α and increased angiostatin expression. Furthermore, it inhibited lymphatic endothelial cell tube formation and the expression of VEGF-C and COX-2. RON expression was observed to be associated with age, tumor size, lymphovascular and perineural invasion, tumor stage, lymph node and distant metastasis, and poor survival rate. The mean microvessel density value of RON-positive tumors was significantly higher than that of RON-negative ones. These results indicate that RON is associated with tumor progression by inhibiting chemosensitivity and enhancing angiogenesis in colorectal cancer.
Collapse
Affiliation(s)
- Nuri Kim
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Sung-Bum Cho
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Young-Lan Park
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Sun-Young Park
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Eun Myung
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Seung-Hun Kim
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Hyung-Min Yu
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Young-Ae Son
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Dae-Seong Myung
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Wan-Sik Lee
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Young-Eun Joo
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| |
Collapse
|
62
|
Analysis of tumor-derived DNA in plasma and bone marrow fluid in lung cancer patients. Med Oncol 2016; 33:29. [PMID: 26897174 DOI: 10.1007/s12032-016-0744-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 02/10/2016] [Indexed: 02/05/2023]
Abstract
Liquid biopsies such as circulating tumor DNA in plasma and disseminated tumor cells in the bone marrow are currently available. However, it is unclear which types of samples are appropriate for detecting tumor DNA in these biopsies. Here, we collected primary tumors, pulmonary venous blood, peripheral blood, and rib bone marrow fluid from 10 lung cancer patients. Targeted deep sequencing was performed to identify mutations across 70 specimens. As a result, a total of 43 mutations were identified in the primary tumors. The mutation in the tumors was also identified in circulating tumor DNA in the pulmonary venous and peripheral blood in two patients. These patients showed poor prognosis, as compared to the other patients. However, no mutation was identified in the bone marrow in any of the patients. These results demonstrated that circulating tumor DNA in plasma is more sensitive and clinically useful as a biomarker as compared to DNA in bone marrow fluid.
Collapse
|
63
|
Peng C, Wallwiener M, Rudolph A, Ćuk K, Eilber U, Celik M, Modugno C, Trumpp A, Heil J, Marmé F, Madhavan D, Nees J, Riethdorf S, Schott S, Sohn C, Pantel K, Schneeweiss A, Chang-Claude J, Yang R, Burwinkel B. Plasma hyaluronic acid level as a prognostic and monitoring marker of metastatic breast cancer. Int J Cancer 2016; 138:2499-509. [PMID: 26686298 DOI: 10.1002/ijc.29975] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 10/30/2015] [Accepted: 11/13/2015] [Indexed: 11/12/2022]
Abstract
Conventional tumor markers have limited value for prognostication and treatment monitoring in metastatic breast cancer (MBC) patients and novel circulating tumor markers therefore need to be explored. Hyaluronic acid (HA) is a major macropolysaccharide in the extracellular matrix and is reported to be associated with tumor progression. In our study, we investigated plasma HA level with respect to progression free survival (PFS) and overall survival (OS), as well as the treatment monitoring value in MBC patients. The prognostic value of plasma HA level was investigated in a discovery cohort of 212 MBC patients with 2.5-year follow-up and validated in an independent validation cohort of 334 patients with 5-year follow-up. The treatment monitoring value of plasma HA level was investigated in 61 MBC patients from discovery cohort who had been radiographically examined after first complete cycle of chemo therapy. We found a robust association between high plasma HA level and poor prognosis of MBC patients in both discovery (pPFS = 7.92 × 10(-6) and pOS = 5.27 × 10(-5)) and validation studies (pPFS = 3.66 × 10(-4) and pOS = 1.43 × 10(-4)). In the discovery cohort, the plasma HA level displayed independent prognostic value after adjusted for age and clinicopathological factors, with respect to PFS and OS. Further, the decrease of plasma HA level displayed good concordance with treatment response evaluated by radiographic examination (AUC = 0.79). Plasma HA level displays prognostic value, as well as treatment monitoring value for MBC patients.
Collapse
Affiliation(s)
- Cike Peng
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Markus Wallwiener
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Anja Rudolph
- Division of Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katarina Ćuk
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Ursula Eilber
- Division of Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Muhabbet Celik
- Division of Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Caroline Modugno
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Andreas Trumpp
- Hi-STEM-Heidelberg Institute for Stem Cell Technology and Experimental Medicine GmbH, Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg Heil
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Frederik Marmé
- Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Dharanija Madhavan
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Juliane Nees
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Sabine Riethdorf
- Department of Tumor Biology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Schott
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Christof Sohn
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Schneeweiss
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Jenny Chang-Claude
- Division of Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rongxi Yang
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Barbara Burwinkel
- Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
64
|
Disseminated and circulating tumor cells in bone marrow and blood of breast cancer patients: properties, enrichment, and potential targets. J Cancer Res Clin Oncol 2016; 142:1883-95. [DOI: 10.1007/s00432-016-2118-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/18/2016] [Indexed: 02/03/2023]
|
65
|
Danila DC, Scher HI, Fleisher M. Circulating Tumor Cells as an Analytical Tool in the Management of Patients with Cancer. MANUAL OF MOLECULAR AND CLINICAL LABORATORY IMMUNOLOGY 2016:1051-1061. [DOI: 10.1128/9781555818722.ch111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
66
|
Bakhtiaridoost S, Habibiyan H, Muhammadnejad S, Haddadi M, Ghafoorifard H, Arabalibeik H, Amanpour S. Raman spectroscopy-based label-free cell identification using wavelet transform and support vector machine. RSC Adv 2016. [DOI: 10.1039/c6ra01004k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Wavelet transform and SVM applied to Raman spectra makes a powerful and accurate tool for identification of rare cells such as CTCs.
Collapse
Affiliation(s)
- S. Bakhtiaridoost
- Photonics Engineering Group
- Amirkabir University of Technology
- Tehran
- Iran
| | - H. Habibiyan
- Photonics Engineering Group
- Amirkabir University of Technology
- Tehran
- Iran
| | - S. Muhammadnejad
- Research Center for Molecular and Cellular Imaging
- Tehran University of Medical Sciences
- Tehran
- Iran
| | - M. Haddadi
- Cancer Models Research Center
- Cancer Institute
- Tehran University of Medical Sciences
- Tehran
- Iran
| | - H. Ghafoorifard
- Department of Electrical Engineering
- Amirkabir University of Technology
- Tehran
- Iran
| | - H. Arabalibeik
- Research Center for Biomedical Technology and Robotics (RCBTR)
- Tehran University of Medical Sciences
- Tehran
- Iran
| | - S. Amanpour
- Cancer Models Research Center
- Cancer Institute
- Tehran University of Medical Sciences
- Tehran
- Iran
| |
Collapse
|
67
|
Guo YT, Qiu CZ, Huang ZX, Yu WS, Yang XF, Wang MZ. Correlational research of Golgi phosphorylation protein 3 expression in colorectal cancer. World J Gastroenterol 2015; 21:13473-9. [PMID: 26730158 PMCID: PMC4690176 DOI: 10.3748/wjg.v21.i48.13473] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/23/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effect of Golgi phosphorylation protein 3 (GOLPH3) expression on cell apoptosis, angiogenesis and prognosis in colorectal cancer (CRC). METHODS The expression of GOLPH3 in CRC tissues and normal colorectal mucosae was determined by immunohistochemistry in 62 patients. In addition, immunohistochemistry was also carried out to detect the expression of vascular endothelial growth factor (VEGF), CD34 and microvessel density (MVD). Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay was used to determine the apoptotic index (AI). The Kaplan-Meier method was used to analyze the relationship between GOLPH3 expression and survival in another 123 CRC cases. RESULTS Compared with normal colorectal mucosae, a notably higher level of GOLPH3 protein expression was identified in CRC tissues (53.2% vs 24.2%, P < 0.05). Positive GOLPH3 expression was significantly associated with tumor invasion depth, TNM stage, and lymph node metastasis (P = 0.001; P = 0.020; P = 0.020; P < 0.05, respectively), but not with tumor length, tumor site, and age (P = 0.363; P = 0.819; P = 0.599; P > 0.05, respectively). VEGF expression and MVD in GOLPH3-positive CRC was significantly higher than in GOLPH3-negative CRC (VEGF: 69.7% vs 31.0%; MVD: 21.45 ± 9.39 vs 14.24 ± 8.97; P < 0.05). GOLPH3 expression was negatively correlated with AI in CRC as shown by Spearman correlation analysis (r = -0.320, P < 0.05). The 5-year survival rate in GOLPH3-negative CRC (69.4%) was significantly higher than in GOLPH3-positive CRC (48.6%) (log-rank test, P < 0.05). CONCLUSION High expression of GOLPH3 is found in CRC tissues. GOLPH3 expression may be a novel prognostic marker for CRC patients.
Collapse
|
68
|
Hinz S, Röder C, Tepel J, Hendricks A, Schafmayer C, Becker T, Kalthoff H. Cytokeratin 20 positive circulating tumor cells are a marker for response after neoadjuvant chemoradiation but not for prognosis in patients with rectal cancer. BMC Cancer 2015; 15:953. [PMID: 26674974 PMCID: PMC4682277 DOI: 10.1186/s12885-015-1989-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 12/09/2015] [Indexed: 12/18/2022] Open
Abstract
Background Several studies have shown, that circulating tumor cells (CTC) have a negative prognostic value in colorectal cancer patients. Aim of this study was to evaluate the role of CTC in specifically rectal cancer patients regarding the influence on overall survival and to elucidate the impact of CTC in predicting response after chemoradiation (RCTX). Methods In this prospective monocentric study 267 patients with rectal cancer were included. Patients with locally advanced tumors were treated with RCTX followed by surgery. The primary endpoints were: Evaluation of CTC at the time of surgery and correlation with main tumor characteristics, response to neoadjuvant RCTX and overall survival (OS). CTC were detected in the blood using CK20 RT-PCR. Results Sixty-three patients were treated with neoadjuvant RCTX. In 46.8 % of the patients receiving neoadjuvant RCTX CTC were detected, which was significantly higher than in the group without RCTX (p = 0.002). Histopathologic regression after RCTX was evident in 27.8 % of the patients. In the subgroup of responders after RCTX we found CTC at a significantly lower rate than in non-responders (p = 0.03). No significant association was found between CTC detection and tumor characteristics and OS. The OS was significantly improved for responders compared to non-responders (p = 0.007). Conclusions Responders after neoadjuvant RCTX had a lower incidence of CTC compared to non-responders, which might be a result of effective systemic and local treatment prior to surgery. Interestingly, detection of CTC did not correlate with tumor stage and OS, which is in contrast to previous reports of patients with colon cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1989-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sebastian Hinz
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany.
| | - Christian Röder
- Division Molecular Oncology, Institute for Experimental Cancer Research, Cancer Center North, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany.
| | - Jürgen Tepel
- Clinic for General, Thoracic and Visceral Surgery, Klinikum Osnabrück, Am Finkenhügel 1, 49076, Osnabrück, Germany.
| | - Alexander Hendricks
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany.
| | - Clemens Schafmayer
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany.
| | - Thomas Becker
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany.
| | - Holger Kalthoff
- Division Molecular Oncology, Institute for Experimental Cancer Research, Cancer Center North, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany.
| |
Collapse
|
69
|
Affiliation(s)
- Young Eun Joo
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
70
|
Mu Z, Wang C, Ye Z, Austin L, Civan J, Hyslop T, Palazzo JP, Jaslow R, Li B, Myers RE, Jiang J, Xing J, Yang H, Cristofanilli M. Prospective assessment of the prognostic value of circulating tumor cells and their clusters in patients with advanced-stage breast cancer. Breast Cancer Res Treat 2015; 154:563-71. [PMID: 26573830 DOI: 10.1007/s10549-015-3636-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/07/2015] [Indexed: 12/12/2022]
Abstract
The enumeration of circulating tumor cells (CTCs) provides important prognostic values in patients with metastatic breast cancer. Recent studies indicate that individual CTCs form clusters and these CTC-clusters play an important role in tumor metastasis. We aimed to assess whether quantification of CTC-clusters provides additional prognostic value over quantification of individual CTCs alone. In 115 prospectively enrolled advanced-stage (III and IV) breast cancer patients, CTCs and CTC-clusters were counted in 7.5 ml whole blood using the CellSearch system at baseline before first-line therapy. The individual and joint effects of CTC and CTC cluster counts on patients' progression-free survival (PFS) were analyzed using Cox proportional hazards modeling. Of the 115 patients, 36 (31.3 %) had elevated baseline CTCs (≥5 CTCs/7.5 ml) and 20 (17.4 %) had CTC-clusters (≥2 CTCs/7.5 ml). Patients with elevated CTCs and CTC-clusters both had worse PFS with a hazard ratio (HR) of 2.76 [95 % confidence interval (CI) 1.57-4.86, P log-rank = 0.0005] and 2.83 (1.48-5.39, P log-rank = 0.001), respectively. In joint analysis, compared with patients with <5 CTCs and without CTC-clusters, patients with elevated CTCs but without clusters, and patients with elevated CTCs and with clusters, had an increasing trend of progression risk, with an HR of 2.21 (1.02-4.78) and 3.32 (1.68-6.55), respectively (P log-rank = 0.0006, P trend = 0.0002). The additional prognostic value of CTC-clusters appeared to be more pronounced in patients with inflammatory breast cancer (IBC), the most aggressive form of breast cancer with the poorest survival. Baseline counts of both individual CTCs and CTC-clusters were associated with PFS in advanced-stage breast cancer patients. CTC-clusters might provide additional prognostic value compared with CTC enumeration alone, in patients with elevated CTCs.
Collapse
Affiliation(s)
- Zhaomei Mu
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Chun Wang
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Zhong Ye
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Laura Austin
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Jesse Civan
- Department of Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Terry Hyslop
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, 27710, USA
| | - Juan P Palazzo
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Rebecca Jaslow
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Bingshan Li
- Department of Molecular Physiology & Biophysics, Center for Human Genetics Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ronald E Myers
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Juntao Jiang
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology and Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Hushan Yang
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Massimo Cristofanilli
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
71
|
Oh HH, Park KJ, Kim N, Park SY, Park YL, Oak CY, Myung DS, Cho SB, Lee WS, Kim KK, Joo YE. Impact of KITENIN on tumor angiogenesis and lymphangiogenesis in colorectal cancer. Oncol Rep 2015; 35:253-60. [PMID: 26496979 DOI: 10.3892/or.2015.4337] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/18/2015] [Indexed: 11/06/2022] Open
Abstract
Angiogenesis and lymphangiogenesis are involved in the dissemination of tumor cells from solid tumors to regional lymph nodes and various distant sites. KAI1 COOH-terminal interacting tetraspanin (KITENIN) contributes to tumor progression and poor clinical outcomes in various cancers including colorectal cancer. The aim of the present study was to evaluate whether KITENIN affects tumor angiogenesis and lymphangiogenesis in colorectal cancer. A KITENIN small interfering RNA vector was used to silence KITENIN expression in colorectal cancer cell lines including DLD1 and SW480 cells. To evaluate the ability of KITENIN to induce angiogenesis and lymphangiogenesis in human umbilical vein endothelial cells (HUVECs) and lymphatic endothelial cells (HLECs), we performed Matrigel invasion and tube formation assays. Immunohistochemistry was used to determine the expression of KITENIN in colorectal cancer tissues. Angiogenesis and lymphangiogenesis were evaluated by immunostaining with CD34 and D2-40 antibodies. KITENIN silencing inhibited both HUVEC invasion and tube formation in the DLD1 and SW480 cells. KITENIN silencing led to decreased expression of the angiogenic inducers vascular endothelial growth factor (VEGF)-A and hypoxia-inducible factor-1α and increased expression of the angiogenic inhibitor angiostatin. KITENIN silencing did not inhibit either HLEC invasion or tube formation in all tested cells, but it resulted in decreased expression of the lymphangiogenic inducer VEGF-C. KITENIN expression was significantly associated with tumor stage, depth of invasion, lymph node and distant metastases and poor survival. The mean microvessel density was significantly higher in the KITENIN-positive tumors than that in the KITENIN-negative tumors. However, the mean lymphatic vessel density of KITENIN-positive tumors was not significantly higher than that of the KITENIN-negative tumors. These results suggest that KITENIN promotes tumor progression by enhancing angiogenesis in colorectal cancer.
Collapse
Affiliation(s)
- Hyung-Hoon Oh
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Kang-Jin Park
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Nuri Kim
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Sun-Young Park
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Young-Lan Park
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Chan-Young Oak
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Dae-Seong Myung
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Sung-Bum Cho
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Wan-Sik Lee
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Kyung-Keun Kim
- Department of Pharmacology, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| | - Young-Eun Joo
- Department of Internal Medicine, Chonnam National University Medical School, Dong-ku, Gwangju 501-757, Republic of Korea
| |
Collapse
|
72
|
Mitchell MJ, Castellanos CA, King MR. Immobilized surfactant-nanotube complexes support selectin-mediated capture of viable circulating tumor cells in the absence of capture antibodies. J Biomed Mater Res A 2015; 103:3407-18. [PMID: 25761664 PMCID: PMC4552621 DOI: 10.1002/jbm.a.35445] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/10/2015] [Indexed: 12/21/2022]
Abstract
The metastatic spread of tumor cells from the primary site to anatomically distant organs leads to a poor patient prognosis. Increasing evidence has linked adhesive interactions between circulating tumor cells (CTCs) and endothelial cells to metastatic dissemination. Microscale biomimetic flow devices hold promise as a diagnostic tool to isolate CTCs and develop metastatic therapies, utilizing E-selectin (ES) to trigger the initial rolling adhesion of tumor cells under flow. To trigger firm adhesion and capture under flow, such devices also typically require antibodies against biomarkers thought to be expressed on CTCs. This approach is challenged by the fact that CTCs are now known to exhibit heterogeneous expression of conventional biomarkers. Here, we describe surfactant-nanotube complexes to enhance ES-mediated capture and isolation of tumor cells without the use of capture antibodies. While the majority of tumor cells exhibited weaker rolling adhesion on halloysite nanotubes (HNT) coated with ES, HNT functionalization with the sodium dodecanoate (NaL) surfactant induced a switch to firm cellular adhesion under flow. Conversely, surfactant-nanotube complexes significantly reduced the number of primary human leukocytes captured via ES-mediated adhesion under flow. The switch in tumor cell adhesion was exploited to capture and isolate tumor cells in the absence of EpCAM antibodies, commonly utilized as the gold standard for CTC isolation. Additionally, HNT-NaL complexes were shown to capture tumor cells with low to negligible EpCAM expression, that are not efficiently captured using conventional approaches.
Collapse
Affiliation(s)
- Michael J. Mitchell
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Michael R. King
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
73
|
Becker S. A historic and scientific review of breast cancer: The next global healthcare challenge. Int J Gynaecol Obstet 2015; 131 Suppl 1:S36-9. [DOI: 10.1016/j.ijgo.2015.03.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
74
|
Baek SJ, Ishii H, Tamari K, Hayashi K, Nishida N, Konno M, Kawamoto K, Koseki J, Fukusumi T, Hasegawa S, Ogawa H, Hamabe A, Miyo M, Noguchi K, Seo Y, Doki Y, Mori M, Ogawa K. Cancer stem cells: The potential of carbon ion beam radiation and new radiosensitizers (Review). Oncol Rep 2015; 34:2233-7. [PMID: 26330103 DOI: 10.3892/or.2015.4236] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 07/06/2015] [Indexed: 11/06/2022] Open
Abstract
Cancer stem cells (CSCs) are a small population of cells in cancer with stem-like properties such as cell proliferation, multiple differentiation and tumor initiation capacities. CSCs are therapy-resistant and cause cancer metastasis and recurrence. One key issue in cancer therapy is how to target and eliminate CSCs, in order to cure cancer completely without relapse and metastasis. To target CSCs, many cell surface markers, DNAs and microRNAs are considered as CSC markers. To date, the majority of the reported markers are not very specific to CSCs and are also present in non-CSCs. However, the combination of several markers is quite valuable for identifying and targeting CSCs, although more specific identification methods are needed. While CSCs are considered as critical therapeutic targets, useful treatment methods remain to be established. Epigenetic gene regulators, microRNAs, are associated with tumor initiation and progression. MicroRNAs have been recently considered as promising therapeutic targets, which can alter the therapeutic resistance of CSCs through epigenetic modification. Moreover, carbon ion beam radiotherapy is a promising treatment for CSCs. Evidence indicates that the carbon ion beam is more effective against CSCs than the conventional X-ray beam. Combination therapies of radiosensitizing microRNAs and carbon ion beam radiotherapy may be a promising cancer strategy. This review focuses on the identification and treatment resistance of CSCs and the potential of microRNAs as new radiosensitizers and carbon ion beam radiotherapy as a promising therapeutic strategy against CSCs.
Collapse
Affiliation(s)
- Sung-Jae Baek
- Department of Radiation Oncology, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hideshi Ishii
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Keisuke Tamari
- Department of Radiation Oncology, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kazuhiko Hayashi
- Department of Radiation Oncology, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Naohiro Nishida
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masamitsu Konno
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Koichi Kawamoto
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Jun Koseki
- Department of Cancer Profiling Discovery, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Takahito Fukusumi
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Shinichiro Hasegawa
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hisataka Ogawa
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Atsushi Hamabe
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masaaki Miyo
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kozo Noguchi
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yuji Seo
- Department of Radiation Oncology, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kazuhiko Ogawa
- Department of Radiation Oncology, Osaka University, Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
75
|
Abstract
Cancer metastasis is the main cause of cancer-related death, and dissemination of tumor cells through the blood circulation is an important intermediate step that also exemplifies the switch from localized to systemic disease. Early detection and characterization of circulating tumor cells (CTCs) is therefore important as a general strategy to monitor and prevent the development of overt metastatic disease. Furthermore, sequential analysis of CTCs can provide clinically relevant information on the effectiveness and progression of systemic therapies (e.g., chemo-, hormonal, or targeted therapies with antibodies or small inhibitors). Although many advances have been made regarding the detection and molecular characterization of CTCs, several challenges still exist that limit the current use of this important diagnostic approach. In this review, we discuss the biology of tumor cell dissemination, technical advances, as well as the challenges and potential clinical implications of CTC detection and characterization.
Collapse
Affiliation(s)
- Simon A Joosse
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias M Gorges
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
76
|
Mitchell MJ, Castellanos CA, King MR. Surfactant functionalization induces robust, differential adhesion of tumor cells and blood cells to charged nanotube-coated biomaterials under flow. Biomaterials 2015; 56:179-86. [PMID: 25934290 PMCID: PMC4428905 DOI: 10.1016/j.biomaterials.2015.03.045] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 03/22/2015] [Accepted: 03/27/2015] [Indexed: 12/12/2022]
Abstract
The metastatic spread of cancer cells from the primary tumor to distant sites leads to a poor prognosis in cancers originating from multiple organs. Increasing evidence has linked selectin-based adhesion between circulating tumor cells (CTCs) and endothelial cells of the microvasculature to metastatic dissemination, in a manner similar to leukocyte adhesion during inflammation. Functionalized biomaterial surfaces hold promise as a diagnostic tool to separate CTCs and potentially treat metastasis, utilizing antibody and selectin-mediated interactions for cell capture under flow. However, capture at high purity levels is challenged by the fact that CTCs and leukocytes both possess selectin ligands. Here, a straightforward technique to functionalize and alter the charge of naturally occurring halloysite nanotubes using surfactants is reported to induce robust, differential adhesion of tumor cells and blood cells to nanotube-coated surfaces under flow. Negatively charged sodium dodecanoate-functionalized nanotubes simultaneously enhanced tumor cell capture while negating leukocyte adhesion, both in the presence and absence of adhesion proteins, and can be utilized to isolate circulating tumor cells regardless of biomarker expression. Conversely, diminishing nanotube charge via functionalization with decyltrimethylammonium bromide both abolished tumor cell capture while promoting leukocyte adhesion.
Collapse
Affiliation(s)
- Michael J Mitchell
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Michael R King
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
77
|
Todenhöfer T, Hennenlotter J, Faber F, Wallwiener D, Schilling D, Kühs U, Aufderklamm S, Bier S, Mischinger J, Gakis G, Fehm T, Stenzl A, Schwentner C. Significance of apoptotic and non-apoptotic disseminated tumor cells in the bone marrow of patients with clinically localized prostate cancer. Prostate 2015; 75:637-45. [PMID: 25586166 DOI: 10.1002/pros.22947] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 11/19/2014] [Indexed: 11/11/2022]
Abstract
BACKGROUND Disseminated tumor cells (DTC) can be detected in a high proportion of patients with localized solid malignancies. In prostate cancer (PC), determination of DTCs is critically discussed as there are conflicting results on their prognostic value. The aim of the present study was to evaluate the presence and prognostic role of DTCs in PC patients with a high risk of disease recurrence. METHODS 248 patients with clinically localized PC undergoing radical prostatectomy with features of increased risk of recurrence (PSA ≥10 ng/ml or Gleason score ≥ 4 + 3 = 7 or pT ≥3) were included. All patients underwent intraoperative bone marrow (BM) aspiration biopsy. BM cells were evaluated by immunocytochemistry for cytokeratines and the apoptosis marker caspase-cleaved cytokeratin 18 (M30). Results of immunocytochemistry were correlated with clinical and pathological parameters and clinical outcome of the patients. RESULTS Of 248 patients, 47 (19.0%) had evidence of DTCs at time of radical prostatectomy. In 17 of these 47 patients (36.2%), DTCs expressed the apoptosis marker M30. We observed no correlation between the presence of DTCs and tumor stage, nodal stage, prostate-specific antigen, or Gleason score. After a median-follow-up of 58 months (23-76), no differences in rates of biochemical recurrence, development of metastases and cancer-specific death were observed between patients with and without DTCs while apoptosis markers had no role. CONCLUSIONS In a single-centre cohort of patients with increased risk for disease recurrence, the presence of DTCs at the time of prostatectomy does not influence clinical outcome. For the first time in patients with PC, DTCs were evaluated for immunocytological features indicating apoptosis. Due to conflicting results of studies on DTCs, BM biopsies at time of radical prostatectomy cannot be recommended as a standard procedure in patients with clinically localized PC.
Collapse
Affiliation(s)
- Tilman Todenhöfer
- Department of Urology, University Hospital, Tübingen, Germany; Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Maenz C, Lenfert E, Pantel K, Schumacher U, Deppert W, Wegwitz F. Epithelial-mesenchymal plasticity is a decisive feature for the metastatic outgrowth of disseminated WAP-T mouse mammary carcinoma cells. BMC Cancer 2015; 15:178. [PMID: 25886487 PMCID: PMC4381675 DOI: 10.1186/s12885-015-1165-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 03/05/2015] [Indexed: 11/10/2022] Open
Abstract
Background Experimental analysis of the metastatic cascade requires suitable model systems which allow tracing of disseminated tumor cells and the identification of factors leading to metastatic outgrowth in distant organs. Such models, especially models using immune-competent mice, are rather scarce. We here analyze tumor cell dissemination and metastasis in an immune-competent transplantable mouse mammary tumor model, based on the SV40 transgenic WAP-T mouse mammary carcinoma model. Methods We orthotopically transplanted into immune-competent WAP-T mice two tumor cell lines (H8N8, moderately metastatic, and G-2, non-metastatic), developed from primary WAP-T tumors. G-2 and H8N8 cells exhibit stem cell characteristics, form homeostatic, heterotypic tumor cell systems in vitro, and closely mimic endogenous primary tumors after orthotopic transplantation into syngeneic, immune-competent WAP-T mice. Tumor cell transgene-specific PCR allows monitoring of tumor cell dissemination into distinct organs, and immunohistochemistry for SV40 T-antigen tracing of single disseminated tumor cells (DTC). Results While only H8N8 cell-derived tumors developed metastases, tumors induced with both cell lines disseminated into a variety of organs with similar efficiency and similar organ distribution. H8N8 metastases arose only in lungs, indicating that organ-specific metastatic outgrowth depends on the ability of DTC to re-establish a tumor cell system rather than on invasion per se. Resection of small tumors (0.5 cm3) prevented metastasis of H8N8-derived tumors, most likely due to the rather short half-life of DTC, and thus to shorter exposure of the mice to DTC. In experimental metastasis by tail vein injection, G-2 and H8N8 cells both were able to form lung metastases with similar efficiency. However, after injection of sorted “mesenchymal” and “epithelial” G-2 cell subpopulations, only the “epithelial” subpopulation formed lung metastases. Conclusions We demonstrate the utility of our mouse model to analyze factors influencing tumor cell dissemination and metastasis. We suggest that the different metastatic capacity of G-2 and H8N8 cells is due to their different degrees of epithelial-mesenchymal plasticity (EMP), and thus the ability of the respective disseminated cells to revert from a “mesenchymal” to an “epithelial” differentiation state. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1165-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claudia Maenz
- Institute for Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), D-20246, Hamburg, Germany. .,Department of Tumor Virology, Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, D-20251, Hamburg, Germany.
| | - Eva Lenfert
- Institute for Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), D-20246, Hamburg, Germany. .,Department of Tumor Virology, Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, D-20251, Hamburg, Germany.
| | - Klaus Pantel
- Institute for Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), D-20246, Hamburg, Germany.
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf (UKE), D-20246, Hamburg, Germany.
| | - Wolfgang Deppert
- Institute for Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), D-20246, Hamburg, Germany. .,Department of Tumor Virology, Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, D-20251, Hamburg, Germany.
| | - Florian Wegwitz
- Institute for Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), D-20246, Hamburg, Germany. .,Department of Tumor Virology, Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, D-20251, Hamburg, Germany. .,Department of Translational Cancer Research, University Medical Center Göttingen, D-37075, Göttingen, Germany.
| |
Collapse
|
79
|
Cunningham JJ, Brown JS, Vincent TL, Gatenby RA. Divergent and convergent evolution in metastases suggest treatment strategies based on specific metastatic sites. EVOLUTION MEDICINE AND PUBLIC HEALTH 2015; 2015:76-87. [PMID: 25794501 PMCID: PMC4404930 DOI: 10.1093/emph/eov006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/10/2015] [Indexed: 01/06/2023]
Abstract
Cancer cells, although maximally fit at their primary site, typically have lower fitness on the adaptive landscapes offered by the metastatic sites due to organ-specific variations in mesenchymal properties and signaling pathways. Clinically evident metastases will exhibit time-dependent divergence from the phenotypic mean of the primary population as the tumor cells evolve and adapt to their new circumstances. In contrast, tumors from different primary sites evolving on identical metastatic adaptive landscapes exhibit phenotypic convergence so that, for example, metastases in the liver from different primary tumors will evolve toward similar adaptive phenotypes. The combination of evolutionary divergence from the primary cancer phenotype and convergence towards similar adaptive strategies in the same tissue cause significant variations in treatment responses particularly for highly targeted therapies. This suggest that optimal therapies for disseminated cancer must take into account the site(s) of metastatic growth as well as the primary organ. Background and objective: Systemic therapy for metastatic cancer is currently determined exclusively by the site of tumor origin. Yet, there is increasing evidence that the molecular characteristics of metastases significantly differ from the primary tumor. We define the evolutionary dynamics of metastases that govern this molecular divergence and examine their potential contribution to variations in response to targeted therapies. Methodology: Darwinian interactions of transformed cells with the tissue microenvironments at primary and metastatic sites are analyzed using evolutionary game theory. Computational models simulate responses to targeted therapies in different organs within the same patient. Results: Tumor cells, although maximally fit at their primary site, typically have lower fitness on the adaptive landscapes offered by the metastatic sites due to organ-specific variations in mesenchymal properties and signaling pathways. Clinically evident metastases usually exhibit time-dependent divergence from the phenotypic mean of the primary population as the tumor cells evolve and adapt to their new circumstances. In contrast, tumors from different primary sites evolving on identical metastatic adaptive landscapes exhibit phenotypic convergence. Thus, metastases in the liver from different primary tumors and even in different hosts will evolve toward similar adaptive phenotypes. The combination of evolutionary divergence from the primary cancer phenotype and convergence towards similar adaptive strategies in the same tissue cause significant variations in treatment responses particularly for highly targeted therapies. Conclusion and implications: The results suggest that optimal therapies for disseminated cancer must take into account the site(s) of metastatic growth as well as the primary organ.
Collapse
Affiliation(s)
- Jessica J Cunningham
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL 33612; Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607; Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, 85745, USA
| | - Joel S Brown
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL 33612; Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607; Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, 85745, USA
| | - Thomas L Vincent
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL 33612; Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607; Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, 85745, USA
| | - Robert A Gatenby
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL 33612; Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607; Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, 85745, USA Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL 33612; Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607; Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, 85745, USA
| |
Collapse
|
80
|
Murray NP, Miranda R, Ruiz A, Droguett E. Diagnostic Yield of Primary Circulating Tumor Cells in Women Suspected of Breast Cancer: the BEST (Breast Early Screening Test) Study. Asian Pac J Cancer Prev 2015; 16:1929-34. [DOI: 10.7314/apjcp.2015.16.5.1929] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
81
|
Lee JH, Koung FP, Cho CK, Lee YW, Yoo HS. Review of tumor dormancy therapy using traditional oriental herbal medicine. J Pharmacopuncture 2015; 16:12-20. [PMID: 25780657 PMCID: PMC4331953 DOI: 10.3831/kpi.2013.16.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 01/18/2013] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVE Standard cancer therapy prolongs survival, but can be detrimental to the quality of life, compromise the immune system, and leave residual disease that can cause recurrence years or decades in the future. Tumor dormancy therapy is a novel therapeutic approach that may improve these shortcomings, promote quality of life, and prolong survival. The aim of this study was to analyze studies on dormancy therapy, especially studies using traditional Oriental herbal medicine, so as to evaluate the efficacy of dormancy therapy with traditional oriental herbal medicine. METHODS We conducted a systematic literature review using Scientific and Technical Information Integration Services (NDSL), PubMed, and RISS. We searched for clinical reports, papers, and books related to tumor metastasis, recurrence, immunotherapy, tumor dormancy, and traditional oriental herbal medicine with anticancer effects. Seventy-nine (79) experimental and clinical articles in both Korean and English were reviewed. This study was conducted from March 1, 2012 to May 31, 2012. RESULTS This approach, Tumor dormancy therapy, rather than seeking to remove the tumor, includes combination of low-dose chemotherapy, immunotherapy, immunosurveillance, and other methods to stabilize tumor growth and to enhance the host is immunity against disseminated tumor cells and thus to manage cancer as a chronic disease while maintaining quality of life. In particular, integrative use of Oriental herbal medicine has been shown to induce or maintain tumor dormancy, increase the effectiveness of conventional chemotherapy, improve quality of life, and prolong survival. CONCLUSION Tumor dormancy therapy is a promising novel therapeutic approach that may be especially effective with Oriental herbal medicine. Further research is needed to determine its potential mechanisms and therapeutic applications.
Collapse
Affiliation(s)
- Jong-Ho Lee
- East-West Cancer Center, Dunsan Oriental Hospital of Daejeon University, Daejeon, Korea
| | - Fan-Pei Koung
- Department of General Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Chong-Kwan Cho
- East-West Cancer Center, Dunsan Oriental Hospital of Daejeon University, Daejeon, Korea
| | - Yeon-Weol Lee
- East-West Cancer Center, Dunsan Oriental Hospital of Daejeon University, Daejeon, Korea
| | - Hwa-Seung Yoo
- East-West Cancer Center, Dunsan Oriental Hospital of Daejeon University, Daejeon, Korea ; Department of General Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
82
|
Pesta M, Kulda V, Narsanska A, Fichtl J, Topolcan O. May CTC technologies promote better cancer management? EPMA J 2015; 6:1. [PMID: 25628770 PMCID: PMC4307224 DOI: 10.1186/s13167-014-0023-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/15/2014] [Indexed: 12/18/2022]
Abstract
In the case of cancer, death is usually not due to the primary tumor itself but due to dissemination. Analysis of the circulating tumor cells (CTCs), i.e., cells responsible for a formation of metastases, should provide information useful for the management of cancer patients, fulfilling the objectives of predictive, preventive, and personalized medicine (PPPM). Despite promising results, the decisions on stage of disease and how to guide the adjuvant treatment still do not include results of CTC assessment. We want to describe two major reasons why the recent diagnostic value of CTC analysis is not sufficient for clinical use. The first reason arises from the biological nature of the tumor itself and the second reason is associated with an interdisciplinary status of CTC diagnostics in the sense that it is neither a theme purely for pathologists nor for haemato-oncologists nor clinical biochemists. We anticipate that there are at least three areas where CTCs can be useful for clinical practice. The first is monitoring of treatment efficacy of cancer patients. The second is a molecular characterization of captured CTCs for targeted treatment, and the third is a cultivation of captured CTCs for drug sensitivity testing. All of these approaches allow researchers recognize and respond to changes of phenotype of cancer cells during disease progression and introduce PPPM into clinical practice.
Collapse
Affiliation(s)
- Martin Pesta
- Department of Biology, The Faculty of Medicine in Pilsen, Charles University in Prague, Karlovarska 48, 301 66 Pilsen, Czech Republic ; Biomedical Center, The Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Vlastimil Kulda
- Department of Biochemistry, The Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Andrea Narsanska
- Department of Surgery, The Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Jakub Fichtl
- Department of Surgery, The Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Ondrej Topolcan
- Department of Internal Medicine II, The Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| |
Collapse
|
83
|
|
84
|
Giuliano M, Herrera S, Christiny P, Shaw C, Creighton CJ, Mitchell T, Bhat R, Zhang X, Mao S, Dobrolecki LE, Al-rawi A, Chen F, Veneziani BM, Zhang XHF, Hilsenbeck SG, Contreras A, Gutierrez C, Jeselsohn RM, Rimawi MF, Osborne CK, Lewis MT, Schiff R, Trivedi MV. Circulating and disseminated tumor cells from breast cancer patient-derived xenograft-bearing mice as a novel model to study metastasis. Breast Cancer Res 2015; 17:3. [PMID: 25572662 PMCID: PMC4318479 DOI: 10.1186/s13058-014-0508-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 12/18/2014] [Indexed: 12/23/2022] Open
Abstract
Introduction Real-time monitoring of biologic changes in tumors may be possible by investigating the transitional cells such as circulating tumor cells (CTCs) and disseminated tumor cells in bone marrow (BM-DTCs). However, the small numbers of CTCs and the limited access to bone marrow aspirates in cancer patients pose major hurdles. The goal of this study was to determine whether breast cancer (BC) patient-derived xenograft (PDX) mice could provide a constant and renewable source of CTCs and BM-DTCs, thereby representing a unique system for the study of metastatic processes. Methods CTCs and BM-DTCs, isolated from BC PDX-bearing mice, were identified by immunostaining for human pan-cytokeratin and nuclear counterstaining of red blood cell-lysed blood and bone marrow fractions, respectively. The rate of lung metastases (LM) was previously reported in these lines. Associations between the presence of CTCs, BM-DTCs, and LM were assessed by the Fisher’s Exact and Cochran-Mantel-Haenszel tests. Two separate genetic signatures associated with the presence of CTC clusters and with lung metastatic potential were computed by using the expression arrays of primary tumors from different PDX lines and subsequently overlapped to identify common genes. Results In total, 18 BC PDX lines were evaluated. CTCs and BM-DTCs, present as either single cells or clusters, were detected in 83% (15 of 18) and 62.5% (10 to16) of the lines, respectively. A positive association was noted between the presence of CTCs and BM-DTCs within the same mice. LM was previously found in 9 of 18 (50%) lines, of which all nine had detectable CTCs. The presence of LM was strongly associated with the detection of CTC clusters but not with individual cells or detection of BM-DTCs. Overlapping of the two genetic signatures of the primary PDX tumors associated with the presence of CTC clusters and with lung metastatic potential identified four genes (HLA-DP1A, GJA1, PEG3, and XIST). This four-gene profile predicted distant metastases-free survival in publicly available datasets of early BC patients. Conclusion This study suggests that CTCs and BM-DTCs detected in BC PDX-bearing mice may represent a valuable and unique preclinical model for investigating the role of these rare cells in tumor metastases. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0508-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mario Giuliano
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy.
| | - Sabrina Herrera
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
| | - Pavel Christiny
- Department of Pharmacy Practice and Translational Research, University of Houston, Houston, TX, USA.
| | - Chad Shaw
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| | - Chad J Creighton
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Tamika Mitchell
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
| | - Raksha Bhat
- Department of Pharmacy Practice and Translational Research, University of Houston, Houston, TX, USA.
| | - Xiaomei Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
| | - Sufeng Mao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
| | - Lacey E Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
| | - Ahmed Al-rawi
- Department of Pharmacy Practice and Translational Research, University of Houston, Houston, TX, USA.
| | - Fengju Chen
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| | - Bianca M Veneziani
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy.
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Alejandro Contreras
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Pathology, Baylor College of Medicine, Houston, TX, USA.
| | - Carolina Gutierrez
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Pathology, Baylor College of Medicine, Houston, TX, USA.
| | - Rinath M Jeselsohn
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Mothaffar F Rimawi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - C Kent Osborne
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Radiology, Baylor College of Medicine, Houston, TX, USA.
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| | - Meghana V Trivedi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Pharmacy Practice and Translational Research, University of Houston, Houston, TX, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, USA. .,Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA.
| |
Collapse
|
85
|
Kantara C, O’Connell M, Luthra G, Gajjar A, Sarkar S, Ullrich R, Singh P. Methods for detecting circulating cancer stem cells (CCSCs) as a novel approach for diagnosis of colon cancer relapse/metastasis. J Transl Med 2015; 95:100-12. [PMID: 25347154 PMCID: PMC4281282 DOI: 10.1038/labinvest.2014.133] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/23/2014] [Accepted: 09/30/2014] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are believed to be resistant to currently available therapies and may be responsible for relapse of cancer in patients. Measuring circulating tumor cells (CTCs) in the blood of patients has emerged as a non-invasive diagnostic procedure for screening patients who may be at high risk for developing metastatic cancers or relapse of the cancer disease. However, accurate detection of CTCs has remained a problem, as epithelial-cell markers used to date are not always reliable for detecting CTCs, especially during epithelial-mesenchymal transition. As CSCs are required to initiate metastatic tumors, our goal was to optimize and standardize a method for identifying circulating CSCs (CCSCs) in patients, using established CSC markers. Here, we report for the first time the detection of CCSCs in the blood of athymic nude mice, bearing metastatic tumors, and in the blood of patients positive for colonic adenocarcinomas. Using a simple and non-expensive method, we isolated a relatively pure population of CSCs (CD45-/CK19+), free of red blood cells and largely free of contaminating CD45+ white blood cells. Enriched CCSCs from patients with colon adenocarcinomas had a malignant phenotype and co-expressed CSC markers (DCLK1/LGR5) with CD44/Annexin A2. CSCs were not found in the blood of non-cancer patients, free of colonic growths. Enriched CCSCs from colon cancer patients grew primary spheroids, suggesting the presence of tumor-initiating cells in the blood of these patients. In conclusion, we have developed a novel diagnostic assay for detecting CSCs in circulation, which may more accurately predict the risk of relapse or metastatic disease in patients. As CSCs can potentially initiate metastatic growths, patients positive for CCSCs can be treated with inhibitory agents that selectively target CSCs, besides conventional treatments, to reduce the risk of relapse/metastatic disease for improving clinical outcomes.
Collapse
Affiliation(s)
- Carla Kantara
- Department of Neuroscience and Cell Biology, utmbHealth, Galveston, TX
| | - Malaney O’Connell
- Department of Neuroscience and Cell Biology, utmbHealth, Galveston, TX
| | | | | | | | - Robert Ullrich
- Department of Radiation Oncology, utmbHealth, Galveston, TX
| | - Pomila Singh
- Department of Neuroscience and Cell Biology, utmbHealth, Galveston, TX
| |
Collapse
|
86
|
Bone marrow as a reservoir for disseminated tumor cells: a special source for liquid biopsy in cancer patients. BONEKEY REPORTS 2014; 3:584. [PMID: 25419458 DOI: 10.1038/bonekey.2014.79] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 09/03/2014] [Indexed: 12/13/2022]
Abstract
Besides circulating tumor cells, disseminated tumor cells (DTCs) in bone marrow (BM) might be used as a 'liquid biopsy' to obtain information helpful to steer therapies in individual patients. Moreover, the molecular characterization of DTCs may provide important insight into the biology of cancer metastasis. BM is a frequent site of metastasis in breast, prostate and lung cancer, and it might represent a sanctuary site for DTCs derived from various additional types of epithelial tumors. Highly sensitive and specific immunocytological and molecular methods enable the detection of DTCs in BM of cancer patients at the single-cell level years before the occurrence of metastases. This information might be useful to assess individual prognosis and stratify patients at risk to systemic adjuvant anti-cancer therapies. Although most data on the prognostic value of DTCs are available for breast cancer, several single institution studies including patients with colon, lung, prostate, esophageal, gastric, pancreatic, ovarian and head and neck carcinomas have also documented an association between the presence of DTCs at primary surgery and subsequent metastatic relapse. Most DTCs are in a dormant (that is, non-proliferative) stage, frequently express HER2 and display a cancer stem cell and immune escape phenotype. Here, we summarize the current knowledge about specific biological properties of DTCs in BM, and discuss the clinical relevance of DTC detection in cancer patients with regard to an improved individualized therapeutic management. This will stimulate further technical developments that may make BM sampling more acceptable for the clinical management of patients with solid tumors.
Collapse
|
87
|
Identification of HERC5 and its potential role in NSCLC progression. Int J Cancer 2014; 136:2264-72. [DOI: 10.1002/ijc.29298] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 09/30/2014] [Indexed: 12/20/2022]
|
88
|
Bork U, Grützmann R, Rahbari NN, Schölch S, Distler M, Reissfelder C, Koch M, Weitz J. Prognostic relevance of minimal residual disease in colorectal cancer. World J Gastroenterol 2014; 20:10296-10304. [PMID: 25132746 PMCID: PMC4130837 DOI: 10.3748/wjg.v20.i30.10296] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 12/18/2013] [Accepted: 04/29/2014] [Indexed: 02/06/2023] Open
Abstract
Presence of occult minimal residual disease in patients with colorectal cancer (CRC) has a strong prognostic impact on survival. Minimal residual disease plays a major role in disease relapse and formation of metastases in CRC. Analysis of circulating tumor cells (CTC) in the blood is increasingly used in clinical practice for disease monitoring of CRC patients. In this review article the role of CTC, disseminated tumor cells (DTC) in the bone marrow and micrometastases and isolated tumor cells (ITC) in the lymph nodes will be discussed, including literature published until September 2013. Occult disease is a strong prognostic marker for patient survival in CRC and defined by the presence of CTC in the blood, DTC in the bone marrow and/or micrometastases and ITC in the lymph nodes. Minimal residual disease could be used in the future to identify patient groups at risk, who might benefit from individualized treatment options.
Collapse
|
89
|
Uramoto H, Tanaka F. Recurrence after surgery in patients with NSCLC. Transl Lung Cancer Res 2014; 3:242-9. [PMID: 25806307 PMCID: PMC4367696 DOI: 10.3978/j.issn.2218-6751.2013.12.05] [Citation(s) in RCA: 250] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/30/2013] [Indexed: 12/14/2022]
Abstract
Surgery remains the only potentially curative modality for early-stage non-small cell lung cancer (NSCLC) patients and tissue availability is made possible. However, a proportion of lung cancer patients develop recurrence, even after curative resection. This review discusses the superiority of surgery, the reasons for recurrence, the timing and pattern of recurrence, the identification of factors related to recurrence, current provisions for treatment and perspectives about surgery for patients with NSCLC.
Collapse
Affiliation(s)
- Hidetaka Uramoto
- Second Department of Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Fumihiro Tanaka
- Second Department of Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
90
|
Wolfrom CM, Laurent M, Deschatrette J. Can we negotiate with a tumor? PLoS One 2014; 9:e103834. [PMID: 25084359 PMCID: PMC4118912 DOI: 10.1371/journal.pone.0103834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 07/08/2014] [Indexed: 12/18/2022] Open
Abstract
Recent progress in deciphering the molecular portraits of tumors promises an era of more personalized drug choices. However, current protocols still follow standard fixed-time schedules, which is not entirely coherent with the common observation that most tumors do not grow continuously. This unpredictability of the increases in tumor mass is not necessarily an obstacle to therapeutic efficiency, particularly if tumor dynamics could be exploited. We propose a model of tumor mass evolution as the integrated result of the dynamics of two linked complex systems, tumor cell population and tumor microenvironment, and show the practical relevance of this nonlinear approach.
Collapse
Affiliation(s)
- Claire M. Wolfrom
- Equipe « Dynamiques cellulaires et modélisation », Inserm Unité 757, Université Paris-Sud, Orsay, France
| | - Michel Laurent
- Equipe « Dynamiques cellulaires et modélisation », Inserm Unité 757, Université Paris-Sud, Orsay, France
| | - Jean Deschatrette
- Equipe « Dynamiques cellulaires et modélisation », Inserm Unité 757, Université Paris-Sud, Orsay, France
| |
Collapse
|
91
|
Kamiyama H, Noda H, Konishi F, Rikiyama T. Molecular biomarkers for the detection of metastatic colorectal cancer cells. World J Gastroenterol 2014; 20:8928-8938. [PMID: 25083065 PMCID: PMC4112864 DOI: 10.3748/wjg.v20.i27.8928] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 01/29/2014] [Accepted: 04/03/2014] [Indexed: 02/06/2023] Open
Abstract
Approximately half of all patients with colorectal cancer develop local recurrence or distant metastasis during the course of their illness. Recently, the molecular detection of metastatic cancer cells in various types of clinical samples, such as lymph nodes, bone marrow, peripheral blood, and peritoneal lavage fluid, has been investigated as a potential prognostic marker. The prognostic value of molecular tumor cell detection was independent of the type of detection method used. As assays become more sensitive and quantitative, a more thorough assessment of the cancer status of patients will be based on molecular markers alone. At present, it is difficult to conclude that one specific molecular marker is superior to others. Comparative analyses are recommended to assess the prognostic impact of molecular analyses in the same patient and determine the biomarkers that provide the most accurate prognostic information.
Collapse
|
92
|
EpCAM is overexpressed in local and metastatic prostate cancer, suppressed by chemotherapy and modulated by MET-associated miRNA-200c/205. Br J Cancer 2014; 111:955-64. [PMID: 24992580 PMCID: PMC4150273 DOI: 10.1038/bjc.2014.366] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/07/2014] [Indexed: 12/14/2022] Open
Abstract
Background: Expression of epithelial cell adhesion molecule (EpCAM) is deregulated in epithelial malignancies. Beside its role in cell adhesion, EpCAM acts as signalling molecule with tumour-promoting functions. Thus, EpCAM is part of the molecular network of oncogenic receptors and considered an interesting therapeutic target. Methods: Here, we thoroughly characterised EpCAM expression on mRNA and protein level in comprehensive tissue studies including non-cancerous prostate specimens, primary tumours of different grades and stages, metastatic lesions, and therapy-treated tumour specimens, as well as in prostate cancer cell lines. Results: Epithelial cell adhesion molecule was overexpressed at mRNA and at protein level in prostate cancer tissues and cell lines. Altered EpCAM expression was an early event in prostate carcinogenesis with an upregulation in low-grade cancers and further induction in high-grade tumours and metastatic lesions. Interestingly, EpCAM was repressed upon induction of epithelial-to-mesenchymal transition (EMT) following chemotherapeutic treatment with docetaxel. Oppositely, re-induction of the epithelial phenotype through miRNAs miR-200c and miR-205, two inducers of mesenchymal-to-epithelial transition (MET), led to re-induction of EpCAM in chemoresistant cells. Furthermore, we prove that EpCAM cleavage, the first step of EpCAM signalling takes place in prostate cancer cells but in contrast to other cancer entities, EpCAM has no measurable impact on the proliferative behaviour of prostate cells, in vitro. Conclusions: In conclusion, our data confirm that EpCAM overexpression is an early event during prostate cancer progression. Epithelial cell adhesion molecule displays a dynamic, heterogeneous expression and associates with epithelial cells rather than mesenchymal, chemoresistant cells along with processes of EMT and MET.
Collapse
|
93
|
Taieb J, Tabernero J, Mini E, Subtil F, Folprecht G, Van Laethem JL, Thaler J, Bridgewater J, Petersen LN, Blons H, Collette L, Van Cutsem E, Rougier P, Salazar R, Bedenne L, Emile JF, Laurent-Puig P, Lepage C. Oxaliplatin, fluorouracil, and leucovorin with or without cetuximab in patients with resected stage III colon cancer (PETACC-8): an open-label, randomised phase 3 trial. Lancet Oncol 2014; 15:862-73. [PMID: 24928083 DOI: 10.1016/s1470-2045(14)70227-x] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Since the 1990s, fluorouracil-based adjuvant chemotherapy has significantly reduced the risk of tumour recurrence in patients with stage III colon cancer. We aimed to assess whether the addition of cetuximab to standard adjuvant oxaliplatin, fluorouracil, and leucovorin chemotherapy (FOLFOX4) in patients with stage III colon cancer improved disease-free survival (DFS). METHODS For this open-label, randomised phase 3 study done in nine European countries, we enrolled patients through an interactive voice response system to the central randomisation centre, with a central stratified permuted block randomisation procedure. We randomly assigned patients with resected (R0) stage III disease (1:1) to receive 12 cycles of FOLFOX4 twice a week with or without cetuximab. Patients were stratified by N-status (N1 vs N2), T-status (T1-3 vs T4), and obstruction or perforation status (no obstruction and no perforation vs obstruction or perforation or both). A protocol amendment (applied in June, 2008, after 2096 patients had been randomly assigned to treatment-restricted enrolment to patients with tumours wild-type at codons 12 and 13 in exon 2 of the KRAS gene (KRAS exon 2 wild-type). The primary endpoint was DFS. Analysis was intention to treat in all patients with KRAS exon 2 wild-type tumours. The study is registered at EudraCT, number 2005-003463-23. FINDINGS Between Dec 22, 2005, and Nov 5, 2009, 2559 patients from 340 sites in Europe were randomly assigned. Of these patients, 1602 had KRAS exon 2 wild-type tumours (intention-to-treat population), 791 in the FOLFOX4 plus cetuximab group and 811 in the FOLFOX4 group. Median follow-up was 3·3 years (IQR 3·2-3·4). In the experimental and control groups, DFS was similar in the intention-to-treat population (hazard ratio [HR] 1·05; 95% CI 0·85-1·29; p=0·66), and in patients with KRAS exon 2/BRAF wild-type (n=984, HR 0·99; 95% CI 0·76-1·28) or KRAS exon 2-mutated tumours (n=742, HR 1·06; 95% CI 0·82-1·37). We noted heterogeneous responses to the addition of cetuximab in preplanned subgroup analyses. Grade 3 or 4 acne-like rash (in 209 of 785 patients [27%] vs four of 805 [<1%]), diarrhoea (113 [14%] vs 70 [9%]), mucositis (63 [8%] vs 10 [1%]), and infusion-related reactions (55 [7%] vs 30 [4%]) were more frequent in patients treated with FOLFOX4 plus cetuximab than in those patients who received FOLFOX4 alone. INTERPRETATION The addition of cetuximab to FOLFOX4 did not improve DFS compared with FOLFOX4 alone in patients with KRAS exon 2 wild-type resected stage III colon cancer. This trial cannot conclude on the benefit of cetuximab in the studied population, but the heterogeneity of response suggests that further investigation of the role of FOLFOX4 plus cetuximab in specific patient subgroups is warranted. FUNDING Fédération Francophone de Cancérologie Digestive (FFCD), Merck KGaA, and Sanofi-Aventis.
Collapse
Affiliation(s)
- Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Paris Descartes University, Hôpital Européen Georges Pompidou, Paris, France.
| | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Enrico Mini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Fabien Subtil
- Francophone Federation of Digestive Oncology, Cedex Dijon, France; University of Lyon, Lyon, France; Laboratoire de Biométrie et Biologie Evolutive, Villeurbanne, France; Hospices Civils de Lyon, Service de Biostatistique, Lyon, France
| | - Gunnar Folprecht
- 1st Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany
| | | | - Josef Thaler
- Department of Internal Medicine IV, Klinikum Kreuzschwestern Wels, Austria
| | | | | | - Hélène Blons
- Université Paris Descartes, Sorbonne Paris Cité, France; Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France; UMR-S775, INSERM, Centre Universitaire des Saints Pères, Paris, France
| | - Laurence Collette
- Statistics Department, European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Eric Van Cutsem
- Digestive Oncology, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Philippe Rougier
- Department of Gastroenterology and Digestive Oncology, Paris Descartes University, Hôpital Européen Georges Pompidou, Paris, France
| | - Ramon Salazar
- Catalan Institute of Oncology (IDIBELL), Barcelona, Spain
| | - Laurent Bedenne
- Hepato-Gastroenterology Department Dijon University Hospital and INSERM U 866, France
| | - Jean-François Emile
- EA4340 and Pathology Department, Versailles University and Ambroise Paré Hospital APHP, Boulogne, France
| | - Pierre Laurent-Puig
- Université Paris Descartes Sorbonne Paris Cité France; UMR-S775, INSERM, Bases Moléculaires de la Réponse aux Xénobiotiques, Paris France; Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France
| | - Come Lepage
- Hepato-Gastroenterology Department Dijon University Hospital and INSERM U 866, France
| | | |
Collapse
|
94
|
Schwartz AM, Nolan N. Circulating tumor cells: what goes around, comes around. J Natl Cancer Inst 2014; 106:jnci/dju108. [PMID: 24832786 DOI: 10.1093/jnci/dju108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Arnold M Schwartz
- Affiliations of authors: Department of Pathology (AMS, NN) and Department of Surgery (AMS), The George Washington University, Washington, DC.
| | - Norris Nolan
- Affiliations of authors: Department of Pathology (AMS, NN) and Department of Surgery (AMS), The George Washington University, Washington, DC
| |
Collapse
|
95
|
Bliss SA, Greco SJ, Rameshwar P. Hierarchy of breast cancer cells: key to reverse dormancy for therapeutic intervention. Stem Cells Transl Med 2014; 3:782-6. [PMID: 24833590 DOI: 10.5966/sctm.2014-0013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
An understanding of how cancer cells adapt dormancy would allow for targeted treatment. The current literature suggests that the cancer stem cells might be the major cells with the ability to become quiescent and to resist current drug treatment. The properties of cancer stem cells and healthy stem cells are functionally similar, thereby posing a challenge to target the dormant cells. The bone marrow is particularly a challenge because the dormant breast cancer cells are close to the endosteum, which is also home to the endogenous hematopoietic stem cells. Here we discuss how research studies could bring an understanding of the cellular and molecular interactions between the cancer stem cells and cells within the bone marrow microenvironment. This will allow for intervention to reverse dormancy for targeted treatment. The treatment will require studies within the normal organ functions to ensure treatment without toxicity.
Collapse
Affiliation(s)
- Sarah A Bliss
- Department of Medicine, Hematology/Oncology, New Jersey Medical School and Graduate School of Biomedical Sciences, Rutgers University, Newark, New Jersey, USA
| | - Steven J Greco
- Department of Medicine, Hematology/Oncology, New Jersey Medical School and Graduate School of Biomedical Sciences, Rutgers University, Newark, New Jersey, USA
| | - Pranela Rameshwar
- Department of Medicine, Hematology/Oncology, New Jersey Medical School and Graduate School of Biomedical Sciences, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
96
|
Leong SPL, Tseng WW. Micrometastatic cancer cells in lymph nodes, bone marrow, and blood: Clinical significance and biologic implications. CA Cancer J Clin 2014; 64:195-206. [PMID: 24500995 DOI: 10.3322/caac.21217] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 11/25/2013] [Accepted: 11/25/2013] [Indexed: 01/09/2023] Open
Abstract
Cancer metastasis may be regarded as a progressive process from its inception in the primary tumor microenvironment to distant sites by way of the lymphovascular system. Although this type of tumor dissemination often occurs in an orderly fashion via the sentinel lymph node (SLN), acting as a possible gateway to the regional lymph nodes, bone marrow, and peripheral blood and ultimately to distant metastatic sites, this is not a general rule as tumor cells may enter the blood and spread to distant sites, bypassing the SLN. Methods of detecting micrometastatic cancer cells in the SLN, bone marrow, and peripheral blood of patients have been established. Patients with cancer cells in their SLN, bone marrow, or peripheral blood have worse clinical outcomes than patients with no evidence of spread to these compartments. The presence of these cells also has important biologic implications for disease progression and the clinician's understanding of the process of cancer metastasis. Further characterization of these micrometastatic cancer cells at each stage and site of metastasis is needed to design novel selective therapies for a more "personalized" treatment.
Collapse
Affiliation(s)
- Stanley P L Leong
- Chief of Cutaneous Oncology, Associate Director of the Melanoma Program, Center for Melanoma Research and Treatment, California Pacific Medical Center and Sutter Pacific Medical Foundation, Senior Scientist, California Pacific Medical Center Research Institute, San Francisco, CA
| | | |
Collapse
|
97
|
Optimal timing of pulmonary metastasectomy--is a delayed operation beneficial or counterproductive? Eur J Surg Oncol 2014; 40:1049-55. [PMID: 24746934 DOI: 10.1016/j.ejso.2014.03.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 02/24/2014] [Accepted: 03/19/2014] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Pulmonary metastasectomy represents an established approach in the treatment of lung metastases related to several solid malignant tumors, promising the chance of long term survival. Regarding the proper timing of metastasectomy both operation promptly after diagnosis and delayed operation after an interval of 3 months are common practice. MATERIALS AND METHODS A systematic Medline search addressing the optimal timing of metastasectomy was performed. Since the search query "timing of metastasectomy" yields only a limited number of articles, the Medline search was expanded to include the main arguments for prompt metastasectomy ("metastases of metastasis", "growth rate of pulmonary metastases") and for delayed metastasectomy. RESULTS Based on the data available to date, there is no necessity to expedite the timing of the operation. On the other hand, there is no evidence that a delayed operation, for example after re-staging following an interval of 3 months, provides a benefit. CONCLUSION Therefore the timing of metastasectomy should only depend on the patient's requirements, such as general state of health and oncologic considerations, such as promising multimodal therapy concepts, extrathoracal tumor manifestations or oncologic type of the primary tumor. A delayed operation seems justified if the indication for resection is questionable due to a high risk of early multilocal recurrence.
Collapse
|
98
|
Yoon HJ, Kozminsky M, Nagrath S. Emerging role of nanomaterials in circulating tumor cell isolation and analysis. ACS NANO 2014; 8:1995-2017. [PMID: 24601556 PMCID: PMC4004319 DOI: 10.1021/nn5004277] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Circulating tumor cells (CTCs) are low frequency cells found in the bloodstream after having been shed from a primary tumor. These cells are research targets because of the information they may potentially provide about both an individual cancer as well as the mechanisms through which cancer spreads in the process of metastasis. Established technologies exist for CTC isolation, but the recent progress and future of this field lie in nanomaterials. In this review, we provide perspective into historical CTC capture as well as current research being conducted, emphasizing the significance of the materials being used to fabricate these devices. The modern investigation into CTCs initially featured techniques that have since been commercialized. A major innovation in the field was the development of a microfluidic capture device, first fabricated in silicon and followed up with glass and thermopolymer devices. We then specifically highlight the technologies incorporating magnetic nanoparticles, carbon nanotubes, nanowires, nanopillars, nanofibers, and nanoroughened surfaces, graphene oxide and their fabrication methods. The nanoscale provides a new set of tools that has the potential to overcome current limitations associated with CTC capture and analysis. We believe the current trajectory of the field is in the direction of nanomaterials, allowing the improvements necessary to further CTC research.
Collapse
|
99
|
Romero-Laorden N, Olmos D, Fehm T, Garcia-Donas J, Diaz-Padilla I. Circulating and disseminated tumor cells in ovarian cancer: a systematic review. Gynecol Oncol 2014; 133:632-9. [PMID: 24657303 DOI: 10.1016/j.ygyno.2014.03.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 03/06/2014] [Accepted: 03/09/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Detecting circulating tumor cells (CTCs) in the peripheral blood and disseminated tumor cells (DTCs) in the bone marrow of cancer patients has proven feasible and of prognostic value in different neoplasms. However, the clinical significance of CTCs and DTCs in ovarian cancer and its association with outcome remains unclear. METHODS A literature search in PubMed was performed from January 2000 to December 2013 for studies evaluating CTCs and/or DTCs and its association with clinicopathological characteristics and clinical outcome in ovarian cancer. The main outcome measures were progression-free survival (PFS) and overall survival (OS). RESULTS Fourteen studies met the inclusion criteria. Median study size was 84 patients (range 43-216). Median follow-up was 19months (range 5-52). Most studies were small case series (n<100; studies; 71%). The majority of studies used an immunophenotyping approach to identify CTCs and/or DTCs, but only 3 studies (21%) used the FDA-approved Cell Search method. Despite the differences in methodology among studies the presence of CTCs and DTCs tended to be associated with higher baseline CA-125 serum levels, higher odds of residual disease after surgery, and worse survival in ovarian cancer across studies. No consistent intra-patient correlation was observed between DTCs detected in the bone marrow and CTCs detected in the blood. CONCLUSIONS The presence of CTCs and DTCs is associated with adverse clinicopathological characteristics and poor clinical outcomes in ovarian cancer patients. Its implementation as a valuable prognostic tool in the clinical setting requires uniform methodology and prospective validation.
Collapse
Affiliation(s)
- Nuria Romero-Laorden
- Division of Medical Oncology, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, Madrid, Spain; Clinical Research Program, Centro Nacional de Investigaciones Oncologicas, Madrid, Spain
| | - David Olmos
- Division of Medical Oncology, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, Madrid, Spain; Clinical Research Program, Centro Nacional de Investigaciones Oncologicas, Madrid, Spain
| | - Tanja Fehm
- Department of Obstetrics and Gynecology, University of Düsseldorf, Düsseldorf, Germany
| | - Jesus Garcia-Donas
- Division of Medical Oncology, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, Madrid, Spain; Clinical Research Program, Centro Nacional de Investigaciones Oncologicas, Madrid, Spain
| | - Ivan Diaz-Padilla
- Division of Medical Oncology, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, Madrid, Spain.
| |
Collapse
|
100
|
Reliable single cell array CGH for clinical samples. PLoS One 2014; 9:e85907. [PMID: 24465780 PMCID: PMC3897541 DOI: 10.1371/journal.pone.0085907] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/07/2013] [Indexed: 12/19/2022] Open
Abstract
Background Disseminated cancer cells (DCCs) and circulating tumor cells (CTCs) are extremely rare, but comprise the precursors cells of distant metastases or therapy resistant cells. The detailed molecular analysis of these cells may help to identify key events of cancer cell dissemination, metastatic colony formation and systemic therapy escape. Methodology/Principal Findings Using the Ampli1™ whole genome amplification (WGA) technology and high-resolution oligonucleotide aCGH microarrays we optimized conditions for the analysis of structural copy number changes. The protocol presented here enables reliable detection of numerical genomic alterations as small as 0.1 Mb in a single cell. Analysis of single cells from well-characterized cell lines and single normal cells confirmed the stringent quantitative nature of the amplification and hybridization protocol. Importantly, fixation and staining procedures used to detect DCCs showed no significant impact on the outcome of the analysis, proving the clinical usability of our method. In a proof-of-principle study we tracked the chromosomal changes of single DCCs over a full course of high-dose chemotherapy treatment by isolating and analyzing DCCs of an individual breast cancer patient at four different time points. Conclusions/Significance The protocol enables detailed genome analysis of DCCs and thereby assessment of the clonal evolution during the natural course of the disease and under selection pressures. The results from an exemplary patient provide evidence that DCCs surviving selective therapeutic conditions may be recruited from a pool of genomically less advanced cells, which display a stable subset of specific genomic alterations.
Collapse
|