1
|
Rupp B, Nagpal N, Thanasiu B, Tuck K, Herman K, Brenner DE, Colacino J, Wicha M, Nagrath S. Multiplex characterization of circulating tumor cells from ductal carcinoma in situ patients suggests early tumor dissemination. Cancer Lett 2025; 623:217703. [PMID: 40250790 DOI: 10.1016/j.canlet.2025.217703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/04/2025] [Accepted: 04/04/2025] [Indexed: 04/20/2025]
Abstract
While ducal carcinoma in situ (DCIS) is considered to be pre-invasive, some patients will develop metastatic disease after a long disease-free interval. The prevailing dogma posits that invasive local recurrence is the source of subsequent metastasis, and thus the goal of DCIS therapy is the prevention of local recurrence. Recently, this paradigm has been called into question by the observation that some women develop metastatic disease in the absence of local recurrence or even following bilateral mastectomies, suggesting early cancer dissemination in some patients. If the presence of circulating tumor cells (CTCs) can be verified on some patients with pure DCIS, then dissemination may be occurring earlier than previously thought, suggesting that these patients might require additional monitoring or treatment. Here, we present a workflow to isolate and characterize CTCs from DCIS patients. Using a high throughput size based inertial focusing microfluidic device, the Labyrinth, we isolated and identified CTCs in 66.6 % (12/18) of DCIS patients with an average of 1.337 CTCs per five mL. Immunofluorescence staining and single cell qPCR of CTCs reveal mesenchymal characteristics of the cells that may contribute to their ability to migrate and metastasize. Preliminary targeted DNA sequencing revealed single nucleotide variations previously found in DCIS samples. Overall, this data supports the hypothesis that cancer dissemination is occurring in a subset of DCIS patients earlier than previously thought. Additionally, the molecular characterization of CTC in DCIS patients may provide important information on their biological characteristics and associated clinical behavior.
Collapse
Affiliation(s)
- Brittany Rupp
- Department of Chemical Engineering, University of Michigan, USA; BioInterfaces Institute, University of Michigan, USA
| | - Neha Nagpal
- Department of Chemical Engineering, University of Michigan, USA; BioInterfaces Institute, University of Michigan, USA
| | - Brooke Thanasiu
- Department of Chemical Engineering, University of Michigan, USA
| | - Kristen Tuck
- Rogel Cancer Center, University of Michigan, USA
| | - Kirk Herman
- Rogel Cancer Center, University of Michigan, USA
| | - Dean E Brenner
- Rogel Cancer Center, University of Michigan, USA; Department of Pharmacology, University of Michigan, USA; Department of Internal Medicine, University of Michigan, USA
| | - Justin Colacino
- Rogel Cancer Center, University of Michigan, USA; Department of Environmental Health Sciences, University of Michigan, USA
| | - Max Wicha
- Rogel Cancer Center, University of Michigan, USA; Department of Internal Medicine, University of Michigan, USA.
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, USA; BioInterfaces Institute, University of Michigan, USA; Rogel Cancer Center, University of Michigan, USA.
| |
Collapse
|
2
|
Kim R, Kawai A, Wakisaka M, Shimoyama M, Yasuda N, Ito M, Kin T, Arihiro K. Outcomes in patients with non‐invasive breast carcinoma. Cancer Rep (Hoboken) 2022; 6:e1768. [PMID: 36494178 PMCID: PMC10075290 DOI: 10.1002/cnr2.1768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND AIM Non-invasive breast carcinoma is considered to be localized disease and is distinguished from invasive ductal and lobular carcinomas. The local recurrence of non-invasive carcinoma after surgery may lead to development of invasive carcinoma and promote distant metastasis, which worsens the prognosis for breast cancer mortality. The distant metastasis of non-invasive carcinoma may involve the ductal microvasculature without invasion. The outcomes of non-invasive breast carcinoma were examined in this retrospective cohort study. METHODS AND RESULTS Of 872 primary breast cancers diagnosed at a single center between May 2008 and March 2022, 93 (10.6%) were found to be non-invasive carcinomas and were examined in this study. The breast cancer recurrence and survival rates of patients with non-invasive carcinoma were analyzed retrospectively. The median follow-up period was 1891 (range, 5-4804) days. All patients underwent surgical treatment [mastectomy with sentinel lymph node biopsy (SLNB) and partial mastectomy with or without SLNB, tumorectomy, and microdochectomy]. Postoperatively, radiation therapy was administered to 73 (78.4%) of the patients and endocrine therapy was administered to 64 (81.0%) of 79 patients with hormone-receptor positivity. Of 26 patients who underwent partial mastectomy with SLNB, 24 (92.3%) showed isolated tumor cells in the SLNs on one-step nucleic acid amplification. Local recurrence was observed in three (0.3%) patients; no distant metastasis was observed. One patient died of a noncancerous disease. The overall survival rate was 98.0% and the breast cancer-specific survival rate was 100.0%. CONCLUSIONS Non-invasive breast carcinoma, like invasive breast carcinoma, causes local recurrence, but has a good prognosis without distant metastasis. The clinical significance of isolated tumor cells in the SLNs as a systemic component of non-invasive breast carcinoma remains to be elucidated.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Breast Surgery Hiroshima Mark Clinic Hiroshima Japan
| | - Ami Kawai
- Department of Breast Surgery Hiroshima Mark Clinic Hiroshima Japan
| | - Megumi Wakisaka
- Department of Breast Surgery Hiroshima Mark Clinic Hiroshima Japan
| | - Mika Shimoyama
- Department of Breast Surgery Hiroshima Mark Clinic Hiroshima Japan
| | - Naomi Yasuda
- Department of Breast Surgery Hiroshima Mark Clinic Hiroshima Japan
| | - Mitsuya Ito
- Department of Breast Surgery Hiroshima City Hospital Hiroshima Japan
| | - Takanori Kin
- Department of Breast Surgery Hiroshima City Hospital Hiroshima Japan
| | - Koji Arihiro
- Department of Anatomical Pathology Hiroshima University Hospital Hiroshima Japan
| |
Collapse
|
3
|
Signatures of Breast Cancer Progression in the Blood: What Could Be Learned from Circulating Tumor Cell Transcriptomes. Cancers (Basel) 2022; 14:cancers14225668. [PMID: 36428760 PMCID: PMC9688726 DOI: 10.3390/cancers14225668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Gene expression profiling has revolutionized our understanding of cancer biology, showing an unprecedented ability to impact patient management especially in breast cancer. The vast majority of breast cancer gene expression signatures derive from the analysis of the tumor bulk, an experimental approach that limits the possibility to dissect breast cancer heterogeneity thoroughly and might miss the message hidden in biologically and clinically relevant cell populations. During disease progression or upon selective pressures, cancer cells undergo continuous transcriptional changes, which inevitably affect tumor heterogeneity, response to therapy and tendency to disseminate. Therefore, metastasis-associated signatures and transcriptome-wide gene expression measurement at single-cell resolution hold great promise for the future of breast cancer clinical care. Seen from this perspective, transcriptomics of circulating tumor cells (CTCs) represent an attractive opportunity to bridge the knowledge gap and develop novel biomarkers. This review summarizes the current state-of-the-science on CTC gene expression analysis in breast cancer, addresses technical and clinical issues related to the application of CTC-derived signatures, and discusses potential research directions.
Collapse
|
4
|
Darlix A, Cayrefourcq L, Pouderoux S, Menjot de Champfleur N, Bievelez A, Jacot W, Leaha C, Thezenas S, Alix-Panabières C. Detection of Circulating Tumor Cells in Cerebrospinal Fluid of Patients with Suspected Breast Cancer Leptomeningeal Metastases: A Prospective Study. Clin Chem 2022; 68:1311-1322. [PMID: 35953885 DOI: 10.1093/clinchem/hvac127] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/30/2022] [Indexed: 11/14/2022]
Abstract
BACKGROUND The diagnosis of breast cancer (BC)-related leptomeningeal metastases (LM) relies on the detection of tumor cells in cerebrospinal fluid (CSF) using conventional cytology (gold standard). However, the sensitivity of this technique is low. Our goal was to evaluate whether circulating tumor cell (CTC) detection in CSF using the CellSearch® system could be used for LM diagnosis. METHODS This prospective, monocentric study included adult patients with suspected BC-related LM. The clinical sensitivity and specificity of CTC detection in CSF for LM diagnosis were calculated relative to conventional CSF cytology. RESULTS Forty-nine eligible patients were included and 40 were evaluable (CTC detection technical failure: n = 8, eligibility criteria failure: n = 1). Cytology was positive in 18/40 patients. CTCs were detected in these 18 patients (median: 5824 CTC, range: 93 to 45052) and in 5/22 patients with negative cytology (median: 2 CTC, range: 1 to 44). The detection of ≥1 CSF CTC was associated with a clinical sensitivity of 100% (95% CI, 82.4-100) and a specificity of 77.3% (95% CI, 64.3-90.3) for LM diagnosis. HER2+ CTCs were detected in the CSF of 40.6% of patients with HER2- BC (median: 500 CTC, range: 13 to 28 320). CONCLUSIONS The clinical sensitivity of CTC detection in CSF with the CellSearch® system for LM diagnosis is higher than that of CSF cytology. CTC detection in patients with negative cytology, however, must be further investigated. The finding of HER2+ CTCs in patients with HER2- BC suggests that the HER2 status of LM should be evaluated to increase the treatment opportunities for these patients.
Collapse
Affiliation(s)
- Amélie Darlix
- Department of Medical Oncology, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France.,Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, Montpellier, France
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, University of Montpellier, Montpellier, France.,CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
| | - Stéphane Pouderoux
- Department of Medical Oncology, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | | | - Alexis Bievelez
- Biometrics Unit, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | - William Jacot
- Department of Medical Oncology, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier IRCM, INSERM U1194, University of Montpellier; Montpellier, France
| | - Cristina Leaha
- Department of Pathology, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | - Simon Thezenas
- Biometrics Unit, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, University of Montpellier, Montpellier, France.,CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
| |
Collapse
|
5
|
Søiland H, Janssen EAM, Helland T, Eliassen FM, Hagland M, Nordgård O, Lunde S, Lende TH, Sagen JV, Tjensvoll K, Gilje B, Jonsdottir K, Gudlaugsson E, Lode K, Hagen KB, Gripsrud BH, Lind R, Heie A, Aas T, Austdal M, Egeland NG, Bernklev T, Lash TL, Skartveit L, Kroksveen AC, Oltedal S, Kvaløy JT, Lien EA, Sleire L, Mellgren G. Liquid biopsies and patient-reported outcome measures for integrative monitoring of patients with early-stage breast cancer: a study protocol for the longitudinal observational Prospective Breast Cancer Biobanking (PBCB) study. BMJ Open 2022; 12:e054404. [PMID: 35487718 PMCID: PMC9058781 DOI: 10.1136/bmjopen-2021-054404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
INTRODUCTION Breast cancer is still the most common malignancy among women worldwide. The Prospective Breast Cancer Biobank (PBCB) collects blood and urine from patients with breast cancer every 6 or 12 months for 11 years from 2011 to 2030 at two university hospitals in Western Norway. The project aims to identify new biomarkers that enable detection of systemic recurrences at the molecular level. As blood represents the biological interface between the primary tumour, the microenvironment and distant metastases, liquid biopsies represent the ideal medium to monitor the patient's cancer biology for identification of patients at high risk of relapse and for early detection systemic relapse.Including patient-reported outcome measures (PROMs) allows for a vast number of possibilities to compare PROM data with biological information, enabling the study of fatigue and Quality of Life in patients with breast cancer. METHODS AND ANALYSIS A total of 1455 patients with early-stage breast cancer are enrolled in the PBCB study, which has a one-armed prospective observational design. Participants consent to contribute liquid biopsies (i.e., peripheral blood and urine samples) every 6 or 12 months for 11 years. The liquid biopsies are the basis for detection of circulating tumour cells, circulating tumour DNA (ctDNA), exosomal micro-RNA (miRNA), miRNA in Tumour Educated Platelet and metabolomic profiles. In addition, participants respond to 10 PROM questionnaires collected annually. Moreover, a control group comprising 200 women without cancer aged 25-70 years will provide the same data. ETHICS AND DISSEMINATION The general research biobank PBCB was approved by the Ministry of Health and Care Services in 2007, by the Regional Ethics Committee (REK) in 2010 (#2010/1957). The PROM (#2011/2161) and the biomarker study PerMoBreCan (#2015/2010) were approved by REK in 2011 and 2015 respectively. Results will be published in international peer reviewed journals. Deidentified data will be accessible on request. TRIAL REGISTRATION NUMBER NCT04488614.
Collapse
Affiliation(s)
- Håvard Søiland
- Department of Breast and Endocrine Surgery, Stavanger University Hospital, Stavanger, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Emiel A M Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Thomas Helland
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Finn Magnus Eliassen
- Department of Breast and Endocrine Surgery, Stavanger University Hospital, Stavanger, Norway
| | - Magnus Hagland
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Oddmund Nordgård
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience, University of Stavanger, Stavanger, Norway
| | - Siri Lunde
- Department of Breast and Endocrine Surgery, Stavanger University Hospital, Stavanger, Norway
| | - Tone Hoel Lende
- Department of Breast and Endocrine Surgery, Stavanger University Hospital, Stavanger, Norway
| | - Jørn Vegard Sagen
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Kjersti Tjensvoll
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Bjørnar Gilje
- Department of Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Kristin Jonsdottir
- Department of Research, Stavanger University Hospital, Stavanger, Norway
| | - Einar Gudlaugsson
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Kirsten Lode
- Department of Research, Stavanger University Hospital, Stavanger, Norway
- Faculty of Health Sciences Department of Caring and Ethics, University of Stavanger, Stavanger, Norway
| | - Kari Britt Hagen
- Department of Breast and Endocrine Surgery, Haukeland University Hospital, Bergen, Norway
| | - Birgitta Haga Gripsrud
- Faculty of Health Sciences Department of Caring and Ethics, University of Stavanger, Stavanger, Norway
| | - Ragna Lind
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Anette Heie
- Department of Breast and Endocrine Surgery, Haukeland University Hospital, Bergen, Norway
| | - Turid Aas
- Department of Breast and Endocrine Surgery, Haukeland University Hospital, Bergen, Norway
| | - Marie Austdal
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Research, Stavanger University Hospital, Stavanger, Norway
| | - Nina Gran Egeland
- Department of Breast and Endocrine Surgery, Stavanger University Hospital, Stavanger, Norway
| | - Tomm Bernklev
- Central Hospital in Vestfold, Tønsberg, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Timothy L Lash
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
| | - Linn Skartveit
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | | | - Satu Oltedal
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Jan Terje Kvaløy
- Department of Research, Stavanger University Hospital, Stavanger, Norway
- Mathematics and Physics, Department of Mathematics and Natural Science, University of Stavanger, Stavanger, Norway
| | - Ernst A Lien
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Linda Sleire
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Laboratory Medicine and Pathology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
6
|
Functional analysis of circulating tumour cells: the KEY to understand the biology of the metastatic cascade. Br J Cancer 2022; 127:800-810. [PMID: 35484215 PMCID: PMC9427839 DOI: 10.1038/s41416-022-01819-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 03/21/2022] [Accepted: 04/01/2022] [Indexed: 02/06/2023] Open
Abstract
Metastasis formation is the main cause of cancer-related death in patients with solid tumours. At the beginning of this process, cancer cells escape from the primary tumour to the blood circulation where they become circulating tumour cells (CTCs). Only a small subgroup of CTCs will survive during the harsh journey in the blood and colonise distant sites. The in-depth analysis of these metastasis-competent CTCs is very challenging because of their extremely low concentration in peripheral blood. So far, only few groups managed to expand in vitro and in vivo CTCs to be used as models for large-scale descriptive and functional analyses of CTCs. These models have shown already the high variability and complexity of the metastatic cascade in patients with cancer, and open a new avenue for the development of new diagnostic and therapeutic approaches.
Collapse
|
7
|
The potential of liquid biopsy in the management of cancer patients. Semin Cancer Biol 2022; 84:69-79. [PMID: 35331850 DOI: 10.1016/j.semcancer.2022.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 03/06/2022] [Accepted: 03/17/2022] [Indexed: 02/07/2023]
|
8
|
Verschoor N, Deger T, Jager A, Sleijfer S, Wilting SM, Martens JW. Validity and utility of HER2/ERBB2 copy number variation assessed in liquid biopsies from breast cancer patients: a systematic review. Cancer Treat Rev 2022; 106:102384. [DOI: 10.1016/j.ctrv.2022.102384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/02/2022]
|
9
|
Cell Force-Driven Basement Membrane Disruption Fuels EGF- and Stiffness-Induced Invasive Cell Dissemination from Benign Breast Gland Acini. Int J Mol Sci 2021; 22:ijms22083962. [PMID: 33921304 PMCID: PMC8070162 DOI: 10.3390/ijms22083962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/30/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Local basement membrane (BM) disruption marks the initial step of breast cancer invasion. The activation mechanisms of force-driven BM-weakening remain elusive. We studied the mechanical response of MCF10A-derived human breast cell acini with BMs of tuneable maturation to physical and soluble tumour-like extracellular matrix (ECM) cues. Traction force microscopy (TFM) and elastic resonator interference stress microscopy (ERISM) were used to quantify pro-invasive BM stress and protrusive forces. Substrate stiffening and mechanically impaired BM scaffolds induced the invasive transition of benign acini synergistically. Robust BM scaffolds attenuated this invasive response. Additional oncogenic EGFR activation compromised the BMs’ barrier function, fuelling invasion speed and incidence. Mechanistically, EGFR-PI3-Kinase downstream signalling modulated both MMP- and force-driven BM-weakening processes. We show that breast acini form non-proteolytic and BM-piercing filopodia for continuous matrix mechanosensation, which significantly push and pull on the BM and ECM under pro-invasive conditions. Invasion-triggered acini further shear and compress their BM by contractility-based stresses that were significantly increased (3.7-fold) compared to non-invasive conditions. Overall, the highest amplitudes of protrusive and contractile forces accompanied the highest invasiveness. This work provides a mechanistic concept for tumour ECM-induced mechanically misbalanced breast glands fuelling force-driven BM disruption. Finally, this could facilitate early cell dissemination from pre-invasive lesions to metastasize eventually.
Collapse
|
10
|
RNA-Based CTC Analysis Provides Prognostic Information in Metastatic Breast Cancer. Diagnostics (Basel) 2021; 11:diagnostics11030513. [PMID: 33799422 PMCID: PMC7998407 DOI: 10.3390/diagnostics11030513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/16/2022] Open
Abstract
In metastatic breast cancer (MBC) the molecular characterization of circulating tumor cells (CTCs) provides a unique tool to understand metastasis-biology and therapy-resistance. We evaluated the prognostic significance of gene expression in EpCAM(+) CTCs in 46 MBC patients based on a long follow-up. We selected a panel consisting of stem cell markers (CD24, CD44, ALDH1), the mesenchymal marker TWIST1, receptors (ESR1, PGR, HER2, EGFR) and the epithelial marker CK-19. Singleplex RT-qPCR was used for TWIST1 and CK-19 and multiplex RT-qPCR for stem cell markers and receptors. A group of 19 healthy donors (HD) was used as control. Univariate (p = 0.001) and multivariate analysis (p = 0.002) revealed the prognostic value of combined gene expression of CK-19(+), CD44high/CD24low, ALDH1high/CD24low and HER2 over-expression for overall survival (OS). The Kaplan–Meier estimates of OS were significantly different in patients positive for CK-19 (p = 0.028), CD44high/CD24low (p = 0.002), ALDH1high/CD24low (p = 0.007) and HER2-positive (p = 0.022). Our results indicate that combined gene expression analysis in EpCAM(+) CTCs provides prognostic information in MBC.
Collapse
|
11
|
ESR1 NAPA Assay: Development and Analytical Validation of a Highly Sensitive and Specific Blood-Based Assay for the Detection of ESR1 Mutations in Liquid Biopsies. Cancers (Basel) 2021; 13:cancers13030556. [PMID: 33535614 PMCID: PMC7867152 DOI: 10.3390/cancers13030556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/15/2021] [Accepted: 01/26/2021] [Indexed: 01/04/2023] Open
Abstract
Simple Summary A considerable number of estrogen-receptor–positive (ER+) breast cancer patients develop resistance to endocrine treatment. One of the most important resistance mechanisms is the presence of ESR1 mutations. In the present study, we developed and analytically validated a novel, highly sensitive and specific nuclease-assisted minor-allele enrichment with probe-overlap (NaME-PrO)-assisted Amplification refractory mutation system (ARMS) (NAPA) assay for the detection of four ESR1 mutations (Y537S, Y537C, Y537N and D538G). The assay was further applied in 13 ER+ breast cancer (BrCa) primary tumour tissues (FFPEs), 13 non-cancerous breast tissues (mammoplasties), and 32 pairs of liquid biopsy samples [circulating tumour cells (CTCs) and paired plasma circulating tumour DNA (ctDNA)] obtained at different time points from 8 ER+ metastatic breast cancer patients. In the plasma ctDNA, the ESR1 mutations were not identified at the baseline, whereas the D538G mutation was detected during the follow-up period at five consecutive time points in one patient. In the CTCs, only the Y537C mutation was detected in one patient sample at the baseline. A direct comparison of the ESR1 NAPA assay with the drop-off ddPCR using 32 identical plasma ctDNA samples gave a concordance of 90.6%. We present a low-cost, highly specific, sensitive and robust assay for blood-based ESR1 profiling. Abstract A considerable number of estrogen receptor-positive breast cancer (ER+ BrCa) patients develop resistance to endocrine treatment. One of the most important resistance mechanisms is the presence of ESR1 mutations. We developed and analytically validated a highly sensitive and specific NaME-PrO-assisted ARMS (NAPA) assay for the detection of four ESR1 mutations (Y537S, Y537C, Y537N and D538G) in circulating tumour cells (CTCs) and paired plasma circulating tumour DNA (ctDNA) in patients with ER+ BrCa. The analytical specificity, analytical sensitivity and reproducibility of the assay were validated using synthetic oligos standards. We further applied the developed ESR1 NAPA assay in 13 ER+ BrCa primary tumour tissues, 13 non-cancerous breast tissues (mammoplasties) and 64 liquid biopsy samples: 32 EpCAM-positive cell fractions and 32 paired plasma ctDNA samples obtained at different time points from 8 ER+ metastatic breast cancer patients, during a 5-year follow-up period. Peripheral blood from 11 healthy donors (HD) was used as a control. The developed assay is highly sensitive (a detection of mutation-allelic-frequency (MAF) of 0.5% for D538G and 0.1% for Y537S, Y537C, Y537N), and highly specific (0/13 mammoplasties and 0/11 HD for all mutations). In the plasma ctDNA, ESR1 mutations were not identified at the baseline, whereas the D538G mutation was detected in five sequential ctDNA samples during the follow-up period in the same patient. In the EpCAM-isolated cell fractions, only the Y537C mutation was detected in one patient sample at the baseline. A direct comparison of the ESR1 NAPA assay with the drop-off ddPCR using 32 identical plasma ctDNA samples gave a concordance of 90.6%. We present a low cost, highly specific, sensitive and robust assay for blood-based ESR1 profiling. The clinical performance of the ESR1 NAPA assay will be prospectively evaluated in a large number of well-characterized patient cohorts.
Collapse
|
12
|
Heterogeneity of Circulating Tumor Cells in Breast Cancer: Identifying Metastatic Seeds. Int J Mol Sci 2020; 21:ijms21051696. [PMID: 32121639 PMCID: PMC7084665 DOI: 10.3390/ijms21051696] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/24/2022] Open
Abstract
Metastasis being the main cause of breast cancer (BC) mortality represents the complex and multistage process. The entrance of tumor cells into the blood vessels and the appearance of circulating tumor cells (CTCs) seeding and colonizing distant tissues and organs are one of the key stages in the metastatic cascade. Like the primary tumor, CTCs are extremely heterogeneous and presented by clusters and individual cells which consist of phenotypically and genetically distinct subpopulations. However, among this diversity, only a small number of CTCs is able to survive in the bloodstream and to form metastases. The identification of the metastasis-initiating CTCs is believed to be a critical issue in developing therapeutic strategies against metastatic disease. In this review, we summarize the available literature addressing morphological, phenotypic and genetic heterogeneity of CTCs and the molecular makeup of specific subpopulations associated with BC metastasis. Special attention is paid to the need for in vitro and in vivo studies to confirm the tumorigenic and metastatic potential of metastasis-associating CTCs. Finally, we consider treatment approaches that could be effective to eradicate metastatic CTCs and to prevent metastasis.
Collapse
|
13
|
Yang X, Kui L, Tang M, Li D, Wei K, Chen W, Miao J, Dong Y. High-Throughput Transcriptome Profiling in Drug and Biomarker Discovery. Front Genet 2020; 11:19. [PMID: 32117438 PMCID: PMC7013098 DOI: 10.3389/fgene.2020.00019] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/07/2020] [Indexed: 01/26/2023] Open
Abstract
The development of new drugs is multidisciplinary and systematic work. High-throughput techniques based on “-omics” have driven the discovery of biomarkers in diseases and therapeutic targets of drugs. A transcriptome is the complete set of all RNAs transcribed by certain tissues or cells at a specific stage of development or physiological condition. Transcriptome research can demonstrate gene functions and structures from the whole level and reveal the molecular mechanism of specific biological processes in diseases. Currently, gene expression microarray and high-throughput RNA-sequencing have been widely used in biological, medical, clinical, and drug research. The former has been applied in drug screening and biomarker detection of drugs due to its high throughput, fast detection speed, simple analysis, and relatively low price. With the further development of detection technology and the improvement of analytical methods, the detection flux of RNA-seq is much higher but the price is lower, hence it has powerful advantages in detecting biomarkers and drug discovery. Compared with the traditional RNA-seq, scRNA-seq has higher accuracy and efficiency, especially the single-cell level of gene expression pattern analysis can provide more information for drug and biomarker discovery. Therefore, (sc)RNA-seq has broader application prospects, especially in the field of drug discovery. In this overview, we will review the application of these technologies in drug, especially in natural drug and biomarker discovery and development. Emerging applications of scRNA-seq and the third generation RNA-sequencing tools are also discussed.
Collapse
Affiliation(s)
- Xiaonan Yang
- Guangxi Key Laboratory of Medicinal Resources Protection and Genetic Improvement, Guangxi Botanical Garden of Medicinal Plants, Nanning, China
| | - Ling Kui
- Dana-Farber Cancer Institute, Harvard Medical School, Brookline, MA, United States
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Dawei Li
- College of Biological Big Data, Yunnan Agricultural University, Kunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
| | - Kunhua Wei
- Guangxi Key Laboratory of Medicinal Resources Protection and Genetic Improvement, Guangxi Botanical Garden of Medicinal Plants, Nanning, China.,School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Wei Chen
- College of Biological Big Data, Yunnan Agricultural University, Kunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
| | - Jianhua Miao
- Guangxi Key Laboratory of Medicinal Resources Protection and Genetic Improvement, Guangxi Botanical Garden of Medicinal Plants, Nanning, China.,School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Yang Dong
- Guangxi Key Laboratory of Medicinal Resources Protection and Genetic Improvement, Guangxi Botanical Garden of Medicinal Plants, Nanning, China.,College of Biological Big Data, Yunnan Agricultural University, Kunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
14
|
Costa C, Dávila-Ibáñez AB. Methodology for the Isolation and Analysis of CTCs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1220:45-59. [PMID: 32304079 DOI: 10.1007/978-3-030-35805-1_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The majority of deaths related to breast cancer are caused by metastasis. Understanding the process of metastasis is key to achieve a reduction on breast cancer mortality. Currently, liquid biopsies are gaining attention in this regard. Circulating tumor cells (CTCs), an important component of liquid biopsies, are cells shed from primary tumor that disseminate to blood circulation being responsible of distal metastasis. Hence, the study CTCs is a promising alternative to monitor the progress of metastasis disease and can be used for early diagnosis of cancers as well as for earlier assessment of cancer recurrence and therapy efficacy. Despite their clinical interest, CTC analysis is not recommended by oncology guidelines so far. The main reason is that there is no gold standard technology for CTCs isolation and most of the current technologies are not yet validated for clinical use. In this chapter we will focus on the most relevant technologies for CTC isolation based on their properties and depending on whether it is a positive or negative selection. We also describe each technology based on its potential use and its relevance in breast cancer. The chapter also contains a future perspective including the challenges and requirements of CTC detection.
Collapse
Affiliation(s)
- Clotilde Costa
- Roche-Chus Joint Unit, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain. .,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain.
| | - Ana B Dávila-Ibáñez
- Roche-Chus Joint Unit, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
15
|
Advances in the Characterization of Circulating Tumor Cells in Metastatic Breast Cancer: Single Cell Analyses and Interactions, and Patient-Derived Models for Drug Testing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1220:61-80. [PMID: 32304080 DOI: 10.1007/978-3-030-35805-1_5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metastasis is the major cause of breast cancer death worldwide. In metastatic breast cancer, circulating tumor cells (CTCs) can be captured from patient blood samples sequentially over time and thereby serve as surrogates to assess the biology of surviving cancer cells that may still persist in solitary or multiple metastatic sites following treatment. CTCs may thus function as potential real-time decision-making guides for selecting appropriate therapies during the course of disease or for the development and testing of new treatments. The heterogeneous nature of CTCs warrants the use of single cell platforms to better inform our understanding of these cancer cells. Current techniques for single cell analyses and techniques for investigating interactions between cancer and immune cells are discussed. In addition, methodologies for growing patient-derived CTCs in vitro or propagating them in vivo to facilitate CTC drug testing are reviewed. We advocate the use of CTCs in appropriate microenvironments to appraise the effectiveness of cancer chemotherapies, immunotherapies, and for the development of new cancer treatments, fundamental to personalizing and improving the clinical management of metastatic breast cancer.
Collapse
|
16
|
Circulating Tumor Cells in Pancreatic Cancer: Current Perspectives. Cancers (Basel) 2019; 11:cancers11111659. [PMID: 31717773 PMCID: PMC6895979 DOI: 10.3390/cancers11111659] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related death in the USA and Europe; early symptoms and screenings are lacking, and it is usually diagnosed late with a poor prognosis. Circulating tumor cells (CTCs) have been promising new biomarkers in solid tumors. In the last twenty years (1999-2019), 140 articles have contained the key words "Circulating tumor cells, pancreatic cancer, prognosis and diagnosis." Articles were evaluated for the use of CTCs as prognostic markers and their correlation to survival in pancreatic ductal adenocarcinoma (PDAC). In the final selected 17 articles, the CTC detection rate varied greatly between different enrichment methodologies and ranged from 11% to 92%; the majority of studies used the antigen-dependent CellSearch© system for CTC detection. Fifteen of the reviewed studies showed a correlation between CTC presence and a worse overall survival. The heterogeneity of CTC-detection methods and the lack of uniform results hinder a comparison of the evaluated studies. However, CTCs can be detected in pancreatic cancer and harbor a hope to serve as an early detection tool. Larger studies are needed to corroborate CTCs as valid biomarkers in pancreatic cancer.
Collapse
|
17
|
Snow A, Chen D, Lang JE. The current status of the clinical utility of liquid biopsies in cancer. Expert Rev Mol Diagn 2019; 19:1031-1041. [PMID: 31482746 DOI: 10.1080/14737159.2019.1664290] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Liquid biopsies have attracted considerable attention as potential diagnostic, prognostic, predictive, and screening assays in oncology. The term liquid biopsies include circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) in the blood. While many liquid biopsy technologies are under active investigation, relatively few liquid biopsy assays have been proven to serve as a diagnostic surrogate for biopsies of metastatic disease as predictive biomarkers to guide the selection of therapy in the clinic. Areas covered: The objective of this review is to highlight the status of liquid biopsies in solid tumors in the oncology literature with attention to proven utility as diagnostic surrogates for macrometastases. Expert opinion: Carefully designed clinical-translational studies are needed to establish the diagnostic accuracy and clinical utility of liquid biopsy biomarkers in oncology. Investigators must fully consider relevant pre-analytical variables, assay sensitivity, bioinformatics considerations as well as the clinical utility of rare event profiling in the context of the normal blood background. Future liquid biopsy research should address the concern that not all DNA mutations are expressed and should provide the means to discover potential therapeutic targets in metastatic patients via a minimally invasive blood draw.
Collapse
Affiliation(s)
- Anson Snow
- Department of Surgery, University of Southern California Norris Comprehensive Cancer Center , Los Angeles , CA , USA
| | - Denaly Chen
- Department of Medicine, University of Southern California Norris Comprehensive Cancer Center , Los Angeles , CA , USA
| | - Julie E Lang
- Department of Surgery, University of Southern California Norris Comprehensive Cancer Center , Los Angeles , CA , USA
| |
Collapse
|
18
|
Strati A, Nikolaou M, Georgoulias V, Lianidou ES. Prognostic Significance of TWIST1, CD24, CD44, and ALDH1 Transcript Quantification in EpCAM-Positive Circulating Tumor Cells from Early Stage Breast Cancer Patients. Cells 2019; 8:cells8070652. [PMID: 31261917 PMCID: PMC6679222 DOI: 10.3390/cells8070652] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022] Open
Abstract
(1) Background: The aim of the study was to evaluate the prognostic significance of EMT-associated (TWIST1) and stem-cell (SC) transcript (CD24, CD44, ALDH1) quantification in EpCAM+ circulating tumor cells (CTCs) of early breast cancer patients. (2) Methods: 100 early stage breast cancer patients and 19 healthy donors were enrolled in the study. CD24, CD44, and ALDH1 transcripts of EpCAM+ cells were quantified using a novel highly sensitive and specific quadraplex RT-qPCR, while TWIST1 transcripts were quantified by single RT-qPCR. All patients were followed up for more than 5 years. (3) Results: A significant positive correlation between overexpression of TWIST1 and CD24−/low/CD44high profile was found. Kaplan–Meier analysis revealed that the ER/PR-negative (HR-) patients and those patients with more than 3 positive lymph nodes that overexpressed TWIST1 in EpCAM+ cells had a significant lower DFI (log rank test; p < 0.001, p < 0.001) and OS (log rank test; p = 0.006, p < 0.001). Univariate and multivariate analysis also revealed the prognostic value of TWIST1 overexpression and CD24−/low/CD44high and CD24−/low/ALDH1high profile for both DFI and OS. (4) Conclusions: Detection of TWIST1 overexpression and stem-cell (CD24, CD44, ALDH1) transcripts in EpCAM+ CTCs provides prognostic information in early stage breast cancer patients.
Collapse
Affiliation(s)
- Areti Strati
- Analysis of Circulating Tumor Cells Lab, Department of Chemistry, University of Athens, 15771 Athens, Greece
| | - Michail Nikolaou
- Medical Oncology Unit, "Elena Venizelou" Hospital, 11521 Athens, Greece
| | | | - Evi S Lianidou
- Analysis of Circulating Tumor Cells Lab, Department of Chemistry, University of Athens, 15771 Athens, Greece.
| |
Collapse
|
19
|
Tellez-Gabriel M, Heymann MF, Heymann D. Circulating Tumor Cells as a Tool for Assessing Tumor Heterogeneity. Am J Cancer Res 2019; 9:4580-4594. [PMID: 31367241 PMCID: PMC6643448 DOI: 10.7150/thno.34337] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/23/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor heterogeneity is the major cause of failure in cancer prognosis and prediction. Accurately detecting heterogeneity for the development of biomarkers and the detection of the clones resistant to therapy is one of the main goals of contemporary medicine. Metastases belong to the natural history of cancer. The present review gives an overview on the origin of tumor heterogeneity. Recent progress has made it possible to isolate and characterize circulating tumor cells (CTCs), which are the drivers of the disease between the primary sites and metastatic foci. The most recent methods for characterizing CTCs are summarized and we discuss the power of CTC profiling for analyzing tumor heterogeneity in early and advanced diseases.
Collapse
|
20
|
Advances in liquid biopsy using circulating tumor cells and circulating cell-free tumor DNA for detection and monitoring of breast cancer. Clin Exp Med 2019; 19:271-279. [PMID: 31190187 DOI: 10.1007/s10238-019-00563-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 06/03/2019] [Indexed: 12/17/2022]
Abstract
Overview the progress of liquid biopsy using circulating tumor cells (CTCs) and circulating cell-free tumor DNA (cfDNA) to detect and monitor breast cancer. Based on numerous research efforts, the potential value of CTCs and cfDNA in the clinical aspects of cancer has become clear. With the development of next-generation sequencing analysis and newly developed technologies, many technical issues have been resolved, making liquid biopsy widely used in clinical practice. They can be powerful tools for dynamic monitoring of tumor progression and therapeutic efficacy. In the field of breast cancer, liquid biopsy is a research hot spot in recent years, playing a key role in monitoring breast cancer metastasis, predicting disease recurrence and assessing clinical drug resistance. Liquid biopsy has the advantages of noninvasive, high sensitivity, high specificity and real-time dynamic monitoring. Still application is far from reality, but the research and application prospects of CTCs and cfDNA in breast cancer are still worth exploring and discovering. This article reviews the main techniques and applications of CTCs and cfDNA in breast cancer.
Collapse
|
21
|
Kim TJ, Moon HW, Kang S, Yang J, Hong SH, Lee JY, Ha US. Urovysion FISH Could Be Effective and Useful Method to Confirm the Identity of Cultured Circulating Tumor Cells from Bladder Cancer Patients. J Cancer 2019; 10:3259-3266. [PMID: 31289598 PMCID: PMC6603370 DOI: 10.7150/jca.30079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 04/28/2019] [Indexed: 12/17/2022] Open
Abstract
Objective: To explore whether cultured CTC from bladder-cancer patients originate from bladder cancer and share chromosomal abnormalities, by means of a fluorescence in situ hybridization (FISH) test. Methods: A total of 15 ml of blood was collected from the patients with bladder cancer before treatment began. Isolated CTCs were divided into 5 ml for CTC enumeration and 10 ml for CTC culture. CTCs were counted by immunofluorescent staining with vimentin, cytokeratin, CD45, and DAPI antibody. CTCs were cultured using isolated CTCs in 96-well plates of Mesenchymal Stem Cell Growth Medium for 16~18 days. The resulting cultured CTCs from 20 men with bladder cancer were analyzed by Urovysion FISH. Results: Common gains were on chromosome 3, 7, and 17 in 20 (74.1%), 14 (51.9%), and 20 (74.1%) of 27 patients, respectively. Polysomy was detected on chromosomes 3 and 7 in 9 patients (33.3%). Polysomy involving two chromosomes was observed in 16 (59.3%, chromosome 3 and 17) and 9 patients (33.3%, chromosome 7 and 17) in the same cell. Among the patients with isolated gain, 17 (63.0%) met the positive criteria for Urovysion FISH. Homozygous deletion of 9p21, 5 (18.5%) involved more than 12 cells. Among the different patient cohorts, positive results based on the Urovysion criteria were obtained in cultured CTCs derived from 19 (70.4%) patients. Conclusion: Application of FISH Urovysion to cultured CTCs from bladder cancer could be an effective first step to confirm their origin and sharing of chromosomal abnormalities.
Collapse
Affiliation(s)
- Tae-Jung Kim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyong Woo Moon
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sungmin Kang
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jonghyup Yang
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hoo Hong
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Youl Lee
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - U-Syn Ha
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
22
|
Cheng YH, Chen YC, Lin E, Brien R, Jung S, Chen YT, Lee W, Hao Z, Sahoo S, Min Kang H, Cong J, Burness M, Nagrath S, S Wicha M, Yoon E. Hydro-Seq enables contamination-free high-throughput single-cell RNA-sequencing for circulating tumor cells. Nat Commun 2019; 10:2163. [PMID: 31092822 PMCID: PMC6520360 DOI: 10.1038/s41467-019-10122-2] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 04/16/2019] [Indexed: 01/06/2023] Open
Abstract
Molecular analysis of circulating tumor cells (CTCs) at single-cell resolution offers great promise for cancer diagnostics and therapeutics from simple liquid biopsy. Recent development of massively parallel single-cell RNA-sequencing (scRNA-seq) provides a powerful method to resolve the cellular heterogeneity from gene expression and pathway regulation analysis. However, the scarcity of CTCs and the massive contamination of blood cells limit the utility of currently available technologies. Here, we present Hydro-Seq, a scalable hydrodynamic scRNA-seq barcoding technique, for high-throughput CTC analysis. High cell-capture efficiency and contamination removal capability of Hydro-Seq enables successful scRNA-seq of 666 CTCs from 21 breast cancer patient samples at high throughput. We identify breast cancer drug targets for hormone and targeted therapies and tracked individual cells that express markers of cancer stem cells (CSCs) as well as of epithelial/mesenchymal cell state transitions. Transcriptome analysis of these cells provides insights into monitoring target therapeutics and processes underlying tumor metastasis.
Collapse
Affiliation(s)
- Yu-Heng Cheng
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
| | - Yu-Chih Chen
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA.,Forbes Institute for Cancer Discovery, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA
| | - Eric Lin
- Department of Chemical Engineering, University of Michigan, 2300 Hayward St, Ann Arbor, MI, 48109, USA
| | - Riley Brien
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
| | - Seungwon Jung
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA.,Forbes Institute for Cancer Discovery, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA
| | - Yu-Ting Chen
- Computer Science Department UCLA, Boelter Hall, Los Angeles, CA, 90095-1596, USA
| | - Woncheol Lee
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
| | - Zhijian Hao
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
| | - Saswat Sahoo
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA
| | - Hyun Min Kang
- School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109-2029, USA
| | - Jason Cong
- Computer Science Department UCLA, Boelter Hall, Los Angeles, CA, 90095-1596, USA
| | - Monika Burness
- Rogel Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, 2300 Hayward St, Ann Arbor, MI, 48109, USA
| | - Max S Wicha
- Forbes Institute for Cancer Discovery, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA. .,Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA.
| |
Collapse
|
23
|
Characterization of circulating tumor cells as a reflection of the tumor heterogeneity: myth or reality? Drug Discov Today 2019; 24:763-772. [DOI: 10.1016/j.drudis.2018.11.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/04/2018] [Accepted: 11/10/2018] [Indexed: 12/25/2022]
|
24
|
Rehman Z, Fahim A, Bhatti A, Sadia H, John P. Co-expression of HIF-1α, MDR1 and LAPTM4B in peripheral blood of solid tumors. PeerJ 2019; 7:e6309. [PMID: 30746305 PMCID: PMC6368972 DOI: 10.7717/peerj.6309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/19/2018] [Indexed: 12/22/2022] Open
Abstract
The hypoxic tumor microenvironment is the major contributor of chemotherapy resistance in solid tumors. One of the key regulators of hypoxic responses within the cell is the hypoxia inducible factor-1α (HIF-1α) that is involved in transcription of genes promoting cell survival and chemotherapy resistance. Multidrug resistance gene-1 (MDR1) and Lysosome-associated protein transmembrane 4B-35 (LAPTM4B-35) are among those notable players which augment their responses to cellular hypoxia. MDR1 is the hypoxia responsive gene involved in multidrug resistance phenotype while LAPTM4B-35 is involved in chemotherapy resistance by stabilizing HIF-1α and overexpressing MDR1. Overexpression of HIF-1α, MDR1 and LAPTM4B has been associated with poor disease outcome in many cancers when studied individually at tissue level. However, accessibility of the tissues following the course of chemotherapy for ascertaining chemotherapy resistance is difficult and sometimes not clinically feasible. Therefore, indication of hypoxic biomarkers in patient’s blood can significantly alter the clinical outcome. Hence there is a need to identify a blood based marker to understand the disease progression. In the current study the expression of hypoxia associated chemotherapy resistance genes were studied in the peripheral blood lymphocytes of solid tumor patients and any potential correlation with disease progression were explored. The expression of HIF-1α, MDR1 and LAPTM4B was studied in blood of 72 breast, 42 ovarian, 32 colon and 21 prostate cancer patients through real time PCR analysis using delta cycle threshold method. The statistical scrutiny was executed through Fisher’s Exact test and the Spearman correlation method. There was 12–13 fold increased in expression of HIF-1α, two fold increased in MDR1 and 13–14 fold increased in LAPTM4B mRNA level in peripheral blood of breast, ovarian, prostate and colon cancer patients. In the current study there was an association of HIF-1α, MDR1 and LAPTM4B expression with advanced tumor stage, metastasis and chemotherapy treated group in breast, ovarian, prostate and colon cancer patients. The Spearman analysis also revealed a positive linear association among HIF-1α, MDR1 and LAPTM4B in all the studied cancer patients. The elevated expression of HIF-1α, MDR1 and LAPTM4B in peripheral blood of solid tumor patients can be a predictor of metastasis, disease progression and treatment response in these cancers. However, larger studies are needed to further strengthen their role as a potential biomarker for cancer prognosis.
Collapse
Affiliation(s)
- Zaira Rehman
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Ammad Fahim
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Attya Bhatti
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Hajra Sadia
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Peter John
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| |
Collapse
|
25
|
Zhuang J, Wu Y, Chen L, Liang S, Wu M, Zhou L, Fan C, Zhang Y. Single-Cell Mobility Analysis of Metastatic Breast Cancer Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1801158. [PMID: 30581709 PMCID: PMC6299679 DOI: 10.1002/advs.201801158] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/26/2018] [Indexed: 05/03/2023]
Abstract
Efforts have been taken to enhance the study of single-cells, however, the task remains challenging because most previous investigations cannot exclude the interactions between single cells or separately retrieved cells with specificity for further analyses. Here, a single-cell mobility analysis platform (SCM-Chip) is developed that can not only real-time monitor single-cell migration in independent niches but can also selectively recover target cells one by one. The design of each channel with a single-cell capture unit and an outlet enables the system to place single cells in different isolated niches with fluidic capture and to respectively collect target cells based on mobilities. SCM-Chip characterization of breast cancer cells reveals the presence of high- and low-migratory populations. Whole-cell transcriptome analysis establishes that monocyte chemotactic protein induced protein 1 (MCPIP1) is related with cell mobility; cells with a high expression of MCPIP1 exhibit low mobility in vitro and metastasis in vivo. The SCM platform provides a generic tool for accurate single-cell isolation and differentiation that can be readily adapted for the study of cancer and drug development.
Collapse
Affiliation(s)
- Jialang Zhuang
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Yongjian Wu
- Department of ImmunologyZhongshan School of MedicineSun Yat‐sen University74 Zhongshan 2nd RoadGuangzhou510080P. R. China
| | - Liang Chen
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Siping Liang
- Department of ImmunologyZhongshan School of MedicineSun Yat‐sen University74 Zhongshan 2nd RoadGuangzhou510080P. R. China
| | - Minhao Wu
- Department of ImmunologyZhongshan School of MedicineSun Yat‐sen University74 Zhongshan 2nd RoadGuangzhou510080P. R. China
| | - Ledu Zhou
- Department of General SurgeryXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Chunhai Fan
- Laboratory of Physical BiologyShanghai Institute of Applied PhysicsChinese Academy of SciencesShanghai201800P. R. China
| | - Yuanqing Zhang
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006P. R. China
| |
Collapse
|
26
|
Neves M, Azevedo R, Lima L, Oliveira MI, Peixoto A, Ferreira D, Soares J, Fernandes E, Gaiteiro C, Palmeira C, Cotton S, Mereiter S, Campos D, Afonso LP, Ribeiro R, Fraga A, Tavares A, Mansinho H, Monteiro E, Videira PA, Freitas PP, Reis CA, Santos LL, Dieguez L, Ferreira JA. Exploring sialyl-Tn expression in microfluidic-isolated circulating tumour cells: A novel biomarker and an analytical tool for precision oncology applications. N Biotechnol 2018; 49:77-87. [PMID: 30273682 DOI: 10.1016/j.nbt.2018.09.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 09/14/2018] [Accepted: 09/25/2018] [Indexed: 12/17/2022]
Abstract
Circulating tumour cells (CTCs) originating from a primary tumour, lymph nodes and distant metastases hold great potential for liquid biopsies by providing a molecular fingerprint for disease dissemination and its temporal evolution through the course of disease management. CTC enumeration, classically defined on the basis of surface expression of Epithelial Cell Adhesion Molecule (EpCAM) and absence of the pan-leukocyte marker CD45, has been shown to correlate with clinical outcome. However, existing approaches introduce bias into the subsets of captured CTCs, which may exclude biologically and clinically relevant subpopulations. Here we explore the overexpression of the membrane protein O-glycan sialyl-Tn (STn) antigen in advanced bladder and colorectal tumours, but not in blood cells, to propose a novel CTC isolation technology. Using a size-based microfluidic device, we show that the majority (>90%) of CTCs isolated from the blood of patients with metastatic bladder and colorectal cancers express the STn antigen, supporting a link with metastasis. STn+ CTC counts were significantly higher than EpCAM-based detection in colorectal cancer, providing a more efficient cell-surface biomarker for CTC isolation. Exploring this concept, we constructed a glycan affinity-based microfluidic device for selective isolation of STn+ CTCs and propose an enzyme-based strategy for the recovery of viable cancer cells for downstream investigations. Finally, clinically relevant cancer biomarkers (transcripts and mutations) in bladder and colorectal tumours, were identified in cells isolated by microfluidics, confirming their malignant origin and highlighting the potential of this technology in the context of precision oncology.
Collapse
Affiliation(s)
- Manuel Neves
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; International Iberian Nanotechnology Laboratory (INL), Braga, Portugal
| | - Rita Azevedo
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| | - Luís Lima
- Portuguese Institute of Oncology, Porto, Portugal; Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal
| | - Marta I Oliveira
- International Iberian Nanotechnology Laboratory (INL), Braga, Portugal
| | - Andreia Peixoto
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal; INEB-Institute for Biomedical Engineering of Porto, Portugal
| | | | - Janine Soares
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| | - Elisabete Fernandes
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal; INEB-Institute for Biomedical Engineering of Porto, Portugal
| | - Cristiana Gaiteiro
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| | | | - Sofia Cotton
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| | - Stefan Mereiter
- Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal
| | - Diana Campos
- Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal
| | | | - Ricardo Ribeiro
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal; INEB-Institute for Biomedical Engineering of Porto, Portugal
| | - Avelino Fraga
- Hospital Centre- Hospital of Santo António of Porto, Portugal
| | - Ana Tavares
- Portuguese Institute of Oncology, Porto, Portugal
| | - Hélder Mansinho
- Hemato-Oncology Clinic, Hospital Garcia de Orta, EPE, Almada, Portugal; Gupo de Investigação do Cancro Digestivo-GICD, Portugal
| | | | - Paula A Videira
- Glycoimmunology Group, UCIBIO, Departamento Ciências da Vida, Faculdade de Ciência e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Paulo P Freitas
- International Iberian Nanotechnology Laboratory (INL), Braga, Portugal; INESC - Microsistemas e Nanotecnologias, Lisboa, Lisbon, Portugal
| | - Celso A Reis
- Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal; Faculty of Medicine, University of Porto, Portugal
| | - Lúcio Lara Santos
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; UFP: School of Health Sciences, Fernando Pessoa University of Porto, Portugal; Porto Comprehensive Cancer Center (P.ccc), Porto, Portugal
| | - Lorena Dieguez
- International Iberian Nanotechnology Laboratory (INL), Braga, Portugal
| | - José Alexandre Ferreira
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; International Iberian Nanotechnology Laboratory (INL), Braga, Portugal; Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal.
| |
Collapse
|