1
|
Tretschock LM, Clemente H, Smetanay K, Fremd C, Thewes V, Haßdenteufel K, Scholz AS, Pantel K, Riethdorf S, Trumpp A, Schneeweiss A, Michel L, Deutsch TM. HER2(-Low) Expression on Circulating Tumor Cells and Corresponding Metastatic Tissue in Metastatic Breast Cancer. Oncol Res Treat 2024; 48:161-173. [PMID: 39608321 DOI: 10.1159/000542830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/23/2024] [Indexed: 11/30/2024]
Abstract
INTRODUCTION Significant progress has been made in the targeted therapy of metastatic breast cancer (mBC) in recent years. In this context, new biomarkers enable personalized therapy management and individualized therapy monitoring. Therefore, the systemic treatment is based increasingly on the biological characteristics of the tumor disease. Given the challenges of obtaining fresh tumor tissue through biopsies, the significance of liquid biopsies for assessing circulating tumor cells (CTCs) or circulating tumor DNA is of growing importance for the detection of prognostic and predictive biomarkers. Multiple studies have shown that the number of CTCs decreases under therapy, especially under anti-HER2-targeted therapy, and that the expression of the HER2 status on CTCs could play a role in predicting therapy response and therapeutic monitoring. The aim of this study was to analyze the HER2 status of CTCs in mBC patients before and after 3 months of systemic therapy to evaluate changes in the number of HER2-positive CTCs. The study focuses on HER2-low, which plays an increasingly important role in clinical practice due to new developments of HER2 targeting antibody-drug conjugates. In this context, temporal and spatial heterogeneity of the disease represent a major diagnostic challenge. METHODS A total of 324 patients with complete immunohistochemistry of biopsied metastases were divided into five groups: HER2 negative (-)/hormone receptor (HR) negative (-), HER2 -/HR positive (+), HER2 +/HR±, HER2-low/HR+, and HER2-low/HR-. Before and after 3 months of a new therapeutic line for mBC, CTCs were enumerated and analyzed for HER2 expression using the CellSearch® system. Overall survival of all subgroups was calculated. RESULTS The analyses revealed a discrepancy between the HER2 status of CTCs and corresponding tumor tissues in 98 patients (30.2%). The number of CTCs in general and the number of HER2+ CTCs decreased during systemic treatment, mainly in HER2+ tumors, but also in the other subgroups. CONCLUSIONS Discrepancy in the HER2 status of the metastases and of CTCs was observed in approximately one-third of patients. Measuring HER2 on CTCs could potentially offer a means to longitudinally monitor HER2 status during therapy and simultaneously address challenges such as tumor heterogeneity. Therefore, the predictive value of HER2 on CTCs should be further investigated in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Carlo Fremd
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Verena Thewes
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | | | - Anna S Scholz
- Heidelberg University Women's Hospital, Heidelberg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Riethdorf
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Trumpp
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Schneeweiss
- Heidelberg University Women's Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura Michel
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | | |
Collapse
|
2
|
Lin H, Wu W, Lou X, Wang Y, Sun H, Cai J, Liu S, Wei X. Is circulating tumor cell count-driven cost-effective for first-line therapy choice in HR+/HER2- metastatic breast cancer in the United States? Breast 2024; 74:103680. [PMID: 38359563 PMCID: PMC10877627 DOI: 10.1016/j.breast.2024.103680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/10/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Circulating tumor cell (CTC) counting may be a useful non-invasive biomarker that helps patients choose first-line treatment options. Nevertheless, the cost of CTC inspection may impose an economic burden on patients, necessitating the simultaneous consideration of both its clinical effectiveness and cost. We evaluated the cost-effectiveness of CTC count-guided chemotherapy and endocrine therapy as first-line therapy for HR+/HER2-metastatic breast cancer (MBC) from the perspective of US payers. METHODS Based on the STIC CTC trial, a Markov model was constructed for three health states, and health outcomes were measured in quality-adjusted life years (QALYs) and incremental cost-effectiveness ratio (ICER). One-way and probabilistic sensitivity analyses were performed to assess the robustness of the incremental cost per QALY. RESULTS The base-case analysis revealed that CTC count-driven treatment was associated with improved effectiveness by 0.07 QALYs and increased the overall cost by $9187.05 compared with clinician-driven first-line treatment choices, leading to an ICER of $138 354.15 per QALY. One-way sensitivity analysis indicated that the model was most sensitive to the cost of treatment for neutropenia and the utility for PFS; probability sensitivity analysis indicated that CTC count-driven treatment choices would be considered the cost-effective option at a willingness-to-pay threshold of $150 000 per QALY. CONCLUSIONS The findings of this cost-effectiveness analysis suggest that, at the current price of CTC enumeration, choosing first-line treatment options based on CTC count is a cost-effectiveness approach for treating patients with HR+/HER2- MBC in the US.
Collapse
Affiliation(s)
- Huiting Lin
- Fujian Medical University, School of Pharmacy, Fuzhou, Fujian, 350004, PR China; Shengli Clinical Medical College of Fujian Medical University, Fujian Province Hospital, Department of Pharmacy, Fuzhou, Fujian, 350001, PR China
| | - Wenhua Wu
- Fujian Medical University, School of Pharmacy, Fuzhou, Fujian, 350004, PR China; Shengli Clinical Medical College of Fujian Medical University, Fujian Province Hospital, Department of Pharmacy, Fuzhou, Fujian, 350001, PR China
| | - Xiaoya Lou
- The First Hospital of Changsha City, Affiliated Changsha Hospital of XiangYa School of Medicine, CSU, Changsha, Hunan, 410000, PR China
| | - Yiming Wang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Province Hospital, Department of Pharmacy, Fuzhou, Fujian, 350001, PR China; Fujian University of Traditional Chinese Medicine, School of Pharmacy, Fuzhou, Fujian, 350122, PR China
| | - Hong Sun
- Shengli Clinical Medical College of Fujian Medical University, Fujian Province Hospital, Department of Pharmacy, Fuzhou, Fujian, 350001, PR China
| | - Jiaqin Cai
- Shengli Clinical Medical College of Fujian Medical University, Fujian Province Hospital, Department of Pharmacy, Fuzhou, Fujian, 350001, PR China
| | - Suyan Liu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Province Hospital, Department of Pharmacy, Fuzhou, Fujian, 350001, PR China; Fujian University of Traditional Chinese Medicine, School of Pharmacy, Fuzhou, Fujian, 350122, PR China
| | - Xiaoxia Wei
- Shengli Clinical Medical College of Fujian Medical University, Fujian Province Hospital, Department of Pharmacy, Fuzhou, Fujian, 350001, PR China.
| |
Collapse
|
3
|
Secer Celik F, Eroglu Gunes C, Kurar E. Cardiac Glycoside Oleandrin Suppresses EMT Ability in Endometrial Carcinoma Cells. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2023; 12:220-228. [PMID: 38751655 PMCID: PMC11092899 DOI: 10.22088/ijmcm.bums.12.3.220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 01/25/2024] [Accepted: 04/06/2024] [Indexed: 05/18/2024]
Abstract
Endometrial carcinoma is one of the most common types of cancer among women. The progression of cancer occurs via the Epithelial- Mesenchymal Transition (EMT) pathway. Cells lose their epithelial properties and become mobile. For this reason, the EMT process is one of the most important step to be targeted in cancer treatment. Oleandrin is a cardiac glycoside and its use is limited due to its narrow therapeutic index. In this study, we aimed to evaluate effects of lower level Oleandrin doses on EMT process in endometrial carcinoma. Oleandrin was administrated to Ishikawa endometrial adenocarcinoma cells at different doses and times. IC50 dose was determined by XTT proliferation test. Expression analysis of EMT-related genes was then performed by qRT-PCR. Invasion and colony formation abilities of cells were examined microscopically. Finally, the migration analysis of cancer cells was determined by the Wound Healing Assay. The IC50 dose of Oleandrin applied to Ishikawa cells was determined as 75.3 nM at the 48 h. According to qRT-PCR analysis, expression levels of ZEB1, FN1, ITGB1, VIM, SMAD2, SNAI1, SNAI2, SNAI3, and TGFB3 genes significantly decreased, but TIMP2, TIMP3, ITGAV and GSK3B genes significantly increased. In addition, Oleandrin significantly reduced colony formation and invasion of Ishikawa cells. According to the Wound Healing analysis, the migratory abilities of the Oleandrin-treated cells were reduced compared to the control. Low dose Oleandrin suppresses the EMT pathway in Ishikawa cells. It has been shown that Oleandrin significantly suppresses the cell's colony formation, invasion and migration ability both in gene expression analyzes and microscopically.
Collapse
Affiliation(s)
- Fatma Secer Celik
- Department of Medical Biology and Genetics, Faculty of Medicine, Ankara Medipol University, Ankara, Turkey.
| | - Canan Eroglu Gunes
- Department of Medical Biology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey.
| | - Ercan Kurar
- Department of Medical Biology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey.
| |
Collapse
|
4
|
Borriello L, Karagiannis GS, Duran CL, Coste A, Oktay MH, Entenberg D, Condeelis JS. The role of the tumor microenvironment in tumor cell intravasation and dissemination. Eur J Cell Biol 2020; 99:151098. [PMID: 32800278 DOI: 10.1016/j.ejcb.2020.151098] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/15/2020] [Accepted: 06/29/2020] [Indexed: 01/11/2023] Open
Abstract
Metastasis, a process that requires tumor cell dissemination followed by tumor growth, is the primary cause of death in cancer patients. An essential step of tumor cell dissemination is intravasation, a process by which tumor cells cross the blood vessel endothelium and disseminate to distant sites. Studying this process is of utmost importance given that intravasation in the primary tumor, as well as the secondary and tertiary metastases, is the key step in the systemic spread of tumor cells, and that this process continues even after removal of the primary tumor. High-resolution intravital imaging of the tumor microenvironment of breast carcinoma has revealed that tumor cell intravasation exclusively occurs at doorways, termed "Tumor MicroEnvironment of Metastasis" (TMEM), composed of three different cell types: a Tie2high/VEGFhigh perivascular macrophage, a Mena overexpressing tumor cell, and an endothelial cell, all in direct contact. In this review article, we discuss the interactions between these cell types, the subsequent signaling events which lead to tumor cell intravasation, and the role of invadopodia in supporting tumor cell invasion and dissemination. We end our review by discussing how the knowledge acquired from the use of intravital imaging is now leading to new clinical trials targeting tumor cell dissemination and preventing metastatic progression.
Collapse
Affiliation(s)
- Lucia Borriello
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| | - George S Karagiannis
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Camille L Duran
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Anouchka Coste
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Department of Surgery, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Department of Pathology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - David Entenberg
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Department of Surgery, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
5
|
Cocco S, Piezzo M, Calabrese A, Cianniello D, Caputo R, Di Lauro V, Fusco G, di Gioia G, Licenziato M, de Laurentiis M. Biomarkers in Triple-Negative Breast Cancer: State-of-the-Art and Future Perspectives. Int J Mol Sci 2020; 21:E4579. [PMID: 32605126 PMCID: PMC7369987 DOI: 10.3390/ijms21134579] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous group of tumors characterized by aggressive behavior, high risk of distant recurrence, and poor survival. Chemotherapy is still the main therapeutic approach for this subgroup of patients, therefore, progress in the treatment of TNBC remains an important challenge. Data derived from molecular technologies have identified TNBCs with different gene expression and mutation profiles that may help developing targeted therapies. So far, however, only a few of these have shown to improve the prognosis and outcomes of TNBC patients. Robust predictive biomarkers to accelerate clinical progress are needed. Herein, we review prognostic and predictive biomarkers in TNBC, discuss the current evidence supporting their use, and look at the future of this research field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Michelino de Laurentiis
- Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola, 53, 80131 Napoli NA, Italy; (S.C.); (M.P.); (A.C.); (D.C.); (R.C.); (V.D.L.); (G.F.); (G.d.G.); (M.L.)
| |
Collapse
|
6
|
Singh A, Denu RA, Wolfe SK, Sperger JM, Schehr J, Witkowsky T, Esbona K, Chappell RJ, Weaver BA, Burkard ME, Lang JM. Centrosome amplification is a frequent event in circulating tumor cells from subjects with metastatic breast cancer. Mol Oncol 2020; 14:1898-1909. [PMID: 32255253 PMCID: PMC7400789 DOI: 10.1002/1878-0261.12687] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/03/2020] [Accepted: 03/07/2020] [Indexed: 01/05/2023] Open
Abstract
Centrosome amplification (CA) is a common phenomenon in cancer, promotes genomic stability and cancer evolution, and has been reported to promote metastasis. CA promotes a stochastic gain/loss of chromosomes during cell division, known as chromosomal instability (CIN). However, it is unclear whether CA is present in circulating tumor cells (CTCs), the seeds for metastasis. Here, we surveyed CA in CTCs from human subjects with metastatic breast cancer. CTCs were captured by CD45 exclusion and selection of EpCAM‐positive cells using an exclusion‐based sample preparation technology platform known as VERSA (versatile exclusion‐based rare sample analysis). Centriole amplification (centrin foci> 4) is the definitive assay for CA. However, determination of centrin foci is technically challenging and incompatible with automated analysis. To test if the more technically accessible centrosome marker pericentrin could serve as a surrogate for centriole amplification in CTCs, cells were stained with pericentrin and centrin antibodies to evaluate CA. This assay was first validated using breast cancer cell lines and a nontransformed epithelial cell line model of inducible CA, then translated to CTCs. Pericentrin area and pericentrin area x intensity correlate well with centrin foci, validating pericentrin as a surrogate marker of CA. CA is found in CTCs from 75% of subjects, with variability in the percentage and extent of CA in individual circulating cells in a given subject, similar to the variability previously seen in primary tumors and cell lines. In summary, we created, validated, and implemented a novel method to assess CA in CTCs from subjects with metastatic breast cancer. Such an assay will be useful for longitudinal monitoring of CA in cancer patients and in prospective clinical trials for assessing the impact of CA on response to therapy.
Collapse
Affiliation(s)
- Ashok Singh
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Ryan A Denu
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison, WI, USA
| | - Serena K Wolfe
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Jamie M Sperger
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Jennifer Schehr
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Tessa Witkowsky
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Karla Esbona
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Richard J Chappell
- Departments of Statistics and of Biostatistics & Medical Informatics, University of Wisconsin-Madison, WI, USA
| | - Beth A Weaver
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA.,Department of Cell and Regenerative Biology and Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, WI, USA
| | - Mark E Burkard
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA.,Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison, WI, USA
| | - Joshua M Lang
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA.,Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison, WI, USA
| |
Collapse
|
7
|
Liu MC, Janni W, Georgoulias V, Yardley DA, Harbeck N, Wei X, McGovern D, Beck R. First-Line Doublet Chemotherapy for Metastatic Triple-Negative Breast Cancer: Circulating Tumor Cell Analysis of the tnAcity Trial. Cancer Manag Res 2019; 11:10427-10433. [PMID: 31849532 PMCID: PMC6913260 DOI: 10.2147/cmar.s208712] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 11/14/2019] [Indexed: 12/16/2022] Open
Abstract
Purpose Circulating tumor cells (CTCs) are prognostic biomarkers in metastatic breast cancer, but their role in predicting treatment outcomes in metastatic triple-negative breast cancer (mTNBC) is less clear. The tnAcity trial demonstrated a significant progression-free survival (PFS) benefit with nab-paclitaxel (nab-P)/carboplatin (C) over nab-P/gemcitabine (G) or G/C in patients with mTNBC. We assessed the correlation between CTC dynamics and clinical benefit in all patients and by treatment arm. Methods CTC enumeration, performed using CELLSEARCH technology (Menarini Silicon Biosystems, Huntingdon Valley, PA, USA), was a prespecified exploratory endpoint in the tnAcity trial. Patients with TNBC were categorized based on pre- and post-treatment CTC levels: Group 1 (+ + +; elevated CTCs at baseline and postbaseline), Group 2 (+ ± ±; CTCs elevated at baseline and cleared postbaseline [cycle 3 and/or cycle 5]), or Group 3 (−; no CTCs detected at baseline). The baseline cutoff was ≥1 CTC/7.5 mL for the main analysis; cutoffs of ≥2 and ≥5 CTCs were used for supporting analyses. Results The main analysis included 126 patients (Group 1, n = 24; Group 2, n = 54; and Group 3, n = 48). The median PFS was longer in Group 2 vs Group 1 (8.5 vs 4.7 months; HR, 0.30 [95% CI, 0.17–0.54]). These results were supported by the ≥2- and ≥5-CTC cutoff analyses. The median overall survival rates were 17.8, 16.0, and 9.8 months in Groups 2, 3, and 1, respectively. The overall response rates were 79.6%, 43.8%, and 29.2%, respectively. A numerically higher percentage of patients had CTC clearance during nab-P/C treatment vs nab-P/G or G/C. Conclusion Efficacy outcomes trended positively with chemotherapy-induced elimination of CTCs, suggesting that CTC clearance may predict the chemosensitivity of mTNBC tumors. Trial registration EudraCT Number: 2013-000113-20; ClinicalTrials.gov number: NCT01881230.
Collapse
Affiliation(s)
- Minetta C Liu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Wolfgang Janni
- Department of Obstetrics and Gynecology, University Clinic Ulm, Ulm, Germany
| | - Vassilis Georgoulias
- Department of Medical Oncology, School of Medicine, University of Crete, Crete, Greece
| | - Denise A Yardley
- Department of Medical Oncology, Breast Cancer Research Program, Sarah Cannon Research Institute, Nashville, TN, USA
| | - Nadia Harbeck
- Department of Breast Center, Oncological Therapy and Clinical Trials Unit, University of Munich (LMU), Munich, Germany
| | - Xin Wei
- Department of Translational/Clinical Development, Celgene Corporation, Summit, NJ, USA
| | - Desmond McGovern
- Department of Clinical Research, Celgene Corporation, Summit, NJ, USA
| | - Robert Beck
- Department of Clinical Research, Celgene Corporation, Summit, NJ, USA
| |
Collapse
|
8
|
Lende TH, Austdal M, Bathen TF, Varhaugvik AE, Skaland I, Gudlaugsson E, Egeland NG, Lunde S, Akslen LA, Jonsdottir K, Janssen EAM, Søiland H, Baak JPA. Metabolic consequences of perioperative oral carbohydrates in breast cancer patients - an explorative study. BMC Cancer 2019; 19:1183. [PMID: 31801490 PMCID: PMC6894229 DOI: 10.1186/s12885-019-6393-7] [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: 09/06/2019] [Accepted: 11/21/2019] [Indexed: 12/21/2022] Open
Abstract
Background The metabolic consequences of preoperative carbohydrate load in breast cancer patients are not known. The present explorative study investigated the systemic and tumor metabolic changes after preoperative per-oral carbohydrate load and their influence on tumor characteristics and survival. Methods The study setting was on university hospital level with primary and secondary care functions in south-west Norway. Serum and tumor tissue were sampled from a population-based cohort of 60 patients with operable breast cancer who were randomized to either per-oral carbohydrate load (preOp™; n = 25) or standard pre-operative fasting (n = 35) before surgery. Magnetic resonance (MR) metabolomics was performed on serum samples from all patients and high-resolution magic angle spinning (HR-MAS) MR analysis on 13 tumor samples available from the fasting group and 16 tumor samples from the carbohydrate group. Results Fourteen of 28 metabolites were differently expressed between fasting and carbohydrate groups. Partial least squares discriminant analysis showed a significant difference in the metabolic profile between the fasting and carbohydrate groups, compatible with the endocrine effects of insulin (i.e., increased serum-lactate and pyruvate and decreased ketone bodies and amino acids in the carbohydrate group). Among ER-positive tumors (n = 18), glutathione was significantly elevated in the carbohydrate group compared to the fasting group (p = 0.002), with a positive correlation between preoperative S-insulin levels and the glutathione content in tumors (r = 0.680; p = 0.002). In all tumors (n = 29), glutamate was increased in tumors with high proliferation (t-test; p = 0.009), independent of intervention group. Moreover, there was a positive correlation between tumor size and proliferation markers in the carbohydrate group only. Patients with ER-positive / T2 tumors and high tumor glutathione (≥1.09), high S-lactate (≥56.9), and high S-pyruvate (≥12.5) had inferior clinical outcomes regarding relapse-free survival, breast cancer-specific survival, and overall survival. Moreover, Integrated Pathway Analysis (IPA) in serum revealed activation of five major anabolic metabolic networks contributing to proliferation and growth. Conclusions Preoperative carbohydrate load increases systemic levels of lactate and pyruvate and tumor levels of glutathione and glutamate in ER-positive patients. These biological changes may contribute to the inferior clinical outcomes observed in luminal T2 breast cancer patients. Trial of registration ClinicalTrials.gov; NCT03886389. Retrospectively registered March 22, 2019.
Collapse
Affiliation(s)
- Tone Hoel Lende
- Department of Breast & Endocrine Surgery, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway. .,Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, Jonas Lies vei 87, N-5012, Bergen, Norway.
| | - Marie Austdal
- Department of Research, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway
| | - Tone Frost Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne Elin Varhaugvik
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Department of Pathology, Helse Møre og Romsdal, Ålesund, Norway
| | - Ivar Skaland
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway
| | - Einar Gudlaugsson
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway
| | - Nina G Egeland
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Department of Chemistry, Bioscience and Environmental Technology, University of Stavanger, P.O. Box 8600 Forus, N-4036, Stavanger, Norway
| | - Siri Lunde
- Department of Breast & Endocrine Surgery, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway
| | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, Jonas Lies vei 87, N-5012, Bergen, Norway
| | - Kristin Jonsdottir
- Department of Research, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway
| | - Emiel A M Janssen
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Department of Chemistry, Bioscience and Environmental Technology, University of Stavanger, P.O. Box 8600 Forus, N-4036, Stavanger, Norway
| | - Håvard Søiland
- Department of Breast & Endocrine Surgery, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Department of Clinical Science, University of Bergen, Jonas Lies vei 87, N-5012, Bergen, Norway
| | - Jan P A Baak
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Dr. Med. Jan Baak AS, Risavegen 66, N-4056, Tananger, Norway
| |
Collapse
|
9
|
Malinzi J, Amima I. Mathematical analysis of a tumour-immune interaction model: A moving boundary problem. Math Biosci 2018; 308:8-19. [PMID: 30537482 DOI: 10.1016/j.mbs.2018.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/06/2018] [Accepted: 12/07/2018] [Indexed: 01/21/2023]
Abstract
A spatio-temporal mathematical model, in the form of a moving boundary problem, to explain cancer dormancy is developed. Analysis of the model is carried out for both temporal and spatio-temporal cases. Stability analysis and numerical simulations of the temporal model replicate experimental observations of immune-induced tumour dormancy. Travelling wave solutions of the spatio-temporal model are determined using the hyperbolic tangent method and minimum wave speeds of invasion are calculated. Travelling wave analysis depicts that cell invasion dynamics are mainly driven by their motion and growth rates. A stability analysis of the spatio-temporal model shows a possibility of dynamical stabilization of the tumour-free steady state. Simulation results reveal that the tumour swells to a dormant level.
Collapse
Affiliation(s)
- Joseph Malinzi
- Department of Mathematics and Applied Mathematics, University of Pretoria, Private Bag X 20, Hatfield, Pretoria 0028, South Africa.
| | - Innocenter Amima
- Department of Mathematical Sciences, Stellenbosch University, Private Bag X1 Matieland, 7602, South Africa.
| |
Collapse
|
10
|
Sheng Y, Wang T, Li H, Zhang Z, Chen J, He C, Li Y, Lv Y, Zhang J, Xu C, Wang Z, Huang C, Wang L. Comparison of analytic performances of Cellsearch and iFISH approach in detecting circulating tumor cells. Oncotarget 2018; 8:8801-8806. [PMID: 28187533 PMCID: PMC5352443 DOI: 10.18632/oncotarget.6688] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 12/10/2015] [Indexed: 01/06/2023] Open
Abstract
Circulating tumor cells (CTCs) have been widely used to predict the prognosis of breast cancer patients. The aim of the present study was to compare the performances of Cellsearch and immunostaining-fluorescence in situ hybridization (iFISH) in detecting CTCs in breast cancer patients. Forty-five newly diagnosed breast cancer patients and 14 healthy donors were recruited and their CTCs were detected by both Cellsearch and iFISH. Correlation between clinicopathological features and CTCs was investigated. We found that the positive rate of CTC detected by iFISH was significantly higher than by Cellsearch system (91% vs 38%). The CTC count, detected either by iFISH or Cellsearch, was not significantly associated with clinical pictures of patients with breast cancer. Therefore, we concluded that, compared to conventional Cellsearch CTC detection, in situ karyotypic identification performed by iFISH had higher detection rate. Therefore, iFISH may be more clinically useful than Cellsearch system.
Collapse
Affiliation(s)
- Yuan Sheng
- Department of Thyroid and Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Ting Wang
- Department of Vascular and Endocrine Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hengyu Li
- Department of Thyroid and Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Zhenzhen Zhang
- Biotecan Medical Diagnostics Co., Ltd, Zhangjiang Center for Translational Medicine, Shanghai, China
| | - Jianghao Chen
- Department of Vascular and Endocrine Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chenyang He
- Department of Vascular and Endocrine Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yongping Li
- Department of Vascular and Endocrine Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yonggang Lv
- Department of Vascular and Endocrine Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Juliang Zhang
- Department of Vascular and Endocrine Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Cheng Xu
- Biotecan Medical Diagnostics Co., Ltd, Zhangjiang Center for Translational Medicine, Shanghai,China
| | - Zhen Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chen Huang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ling Wang
- Department of Vascular and Endocrine Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
11
|
Zhang W, Li Z, Wang Z, Yue C, Zheng H, Li R, Zhou M, Hu Z, Wei Z, Li Q. Generation of a monoclonal antibody recognizing the heavily glycosylated CD45 protein and its application on identifying circulating tumor cells. PLoS One 2018; 13:e0192506. [PMID: 29425242 PMCID: PMC5806877 DOI: 10.1371/journal.pone.0192506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 01/24/2018] [Indexed: 12/31/2022] Open
Abstract
Here, we provide direct evidence that using recombinant proteins expressed in eukaryotic cells as antigen is a practical way to generate monoclonal antibodies (mAbs) against heavily glycosylated proteins. Heavily glycosylated proteins are typically difficult targets for mAb generation, being limited by unsatisfactory affinity and low specificity. Using the heavily glycosylated CD45 protein as an example, we demonstrate the entire process of expressing the protein in eukaryotic cells and using it as an antigen to generate CD45-targeting mAbs in mice. The mAbs generated showed robust affinity and specificity, which are crucial factors for differentiate circulating tumor cells from white blood cells in human breast cancer patient samples. Only 1 cell fusion and 2 cyclic sub-cloning steps were necessary before mAbs with satisfactory performance were obtained.
Collapse
Affiliation(s)
- Weikai Zhang
- Medical College, Henan University of Science and Technology, Luoyang, P. R. China
- Department of Biomedical Engineering, School of Life Science, Beijing Institute of Technology, Beijing, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, P. R. China
| | - Zhitao Li
- Medical College, Henan University of Science and Technology, Luoyang, P. R. China
| | - Zihua Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, P. R. China
| | - Chunyan Yue
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, P. R. China
| | - Hui Zheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, P. R. China
| | - Ren Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, P. R. China
| | - Mingxing Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, P. R. China
| | - Zhiyuan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, P. R. China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, P. R. China
- Yangtze River Delta Academy of Nanotechnology and Industry Development Research, Jiaxing, Zhejiang Province, P. R. China
| | - Zewen Wei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, P. R. China
| | - Qin Li
- Department of Biomedical Engineering, School of Life Science, Beijing Institute of Technology, Beijing, P. R. China
| |
Collapse
|
12
|
Guo YX, Neoh KH, Chang XH, Sun Y, Cheng HY, Ye X, Ma RQ, Han RPS, Cui H. Diagnostic value of HE4+ circulating tumor cells in patients with suspicious ovarian cancer. Oncotarget 2018; 9:7522-7533. [PMID: 29484129 PMCID: PMC5800921 DOI: 10.18632/oncotarget.23943] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/27/2017] [Indexed: 12/31/2022] Open
Abstract
Lacking a satisfactory screening test, ovarian cancer is frequently diagnosed at a late stage, leading to poor patient outcomes. This study investigated the diagnostic value of circulating tumor cells (CTCs) in peripheral blood from patients with suspected ovarian tumors. Sixty-one women suspected of having an ovarian mass were prospectively enrolled in this study. CTCs were identified and counted using microfluidic isolation and immunofluorescent staining of CD45, HE4, and epithelial and mesenchymal (E&M) markers (epithelial cell adhesion molecule, cytokeratins, and vimentin). Thirty (49%) of the patients were diagnosed with ovarian cancer. DAPI+/E&M+/CD45-/HE4+ CTC counts were higher in these patients than in patients with benign tumors (p = 0.016). The receiver operating characteristic (ROC) curve showed that the sensitivity of CTCs was 73.3%, which was superior to that of CA125 (56.7%). In patients with elevated CA125 levels (≥35 U/ml), CTC counts still showed good specificity (86.7%). Our findings suggest the DAPI+/E&M+/CD45-/HE4+ CTC count is a useful diagnostic indicator in patients with suspected ovarian cancer.
Collapse
Affiliation(s)
- Yan-Xiu Guo
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing, China
| | | | - Xiao-Hong Chang
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing, China
| | - Yukun Sun
- College of Engineering, Peking University, Beijing, China
| | - Hong-Yan Cheng
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing, China
| | - Xue Ye
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing, China
| | - Rui-Qiong Ma
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing, China
| | - Ray P S Han
- College of Engineering, Peking University, Beijing, China
| | - Heng Cui
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
13
|
Cordone I, Masi S, Summa V, Carosi M, Vidiri A, Fabi A, Pasquale A, Conti L, Rosito I, Carapella CM, Villani V, Pace A. Overexpression of syndecan-1, MUC-1, and putative stem cell markers in breast cancer leptomeningeal metastasis: a cerebrospinal fluid flow cytometry study. Breast Cancer Res 2017; 19:46. [PMID: 28399903 PMCID: PMC5387324 DOI: 10.1186/s13058-017-0827-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 03/03/2017] [Indexed: 01/09/2023] Open
Abstract
Background Cancer is a mosaic of tumor cell subpopulations, where only a minority is responsible for disease recurrence and cancer invasiveness. We focused on one of the most aggressive circulating tumor cells (CTCs) which, from the primitive tumor, spreads to the central nervous system (CNS), evaluating the expression of prognostic and putative cancer stem cell markers in breast cancer (BC) leptomeningeal metastasis (LM). Methods Flow cytometry immunophenotypic analysis of cerebrospinal fluid (CSF) samples (4.5 ml) was performed in 13 consecutive cases of BCLM. Syndecan-1 (CD138), MUC-1 (CD227) CD45, CD34, and the putative cancer stem cell markers CD15, CD24, CD44, and CD133 surface expression were evaluated on CSF floating tumor cells. The tumor-associated leukocyte population was also characterized. Results Despite a low absolute cell number (8 cell/μl, range 1–86), the flow cytometry characterization was successfully conducted in all the samples. Syndecan-1 and MUC-1 overexpression was documented on BC cells in all the samples analyzed; CD44, CD24, CD15, and CD133 in 77%, 75%, 70%, and 45% of cases, respectively. A strong syndecan-1 and MUC-1 expression was also documented by immunohistochemistry on primary breast cancer tissues, performed in four patients. The CSF tumor population was flanked by T lymphocytes, with a different immunophenotype between the CSF and peripheral blood samples (P ≤ 0.02). Conclusions Flow cytometry can be successfully employed for solid tumor LM characterization even in CSF samples with low cell count. This in vivo study documents that CSF floating BC cells overexpress prognostic and putative cancer stem cell biomarkers related to tumor invasiveness, potentially representing a molecular target for circulating tumor cell detection and LM treatment monitoring, as well as a primary target for innovative treatment strategies. The T lymphocyte infiltration, documented in all CSF samples, suggests a possible involvement of the CNS lymphatic system in both lymphoid and cancer cell migration into and out of the meninges, supporting the extension of a new form of cellular immunotherapy to LM. Due to the small number of cases, validation on large cohorts of patients are warranted to confirm these findings and to evaluate the impact and value of these results for diagnosis and management of LM. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0827-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Iole Cordone
- Regina Elena National Cancer Institute, Clinical Pathology Division, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Serena Masi
- Regina Elena National Cancer Institute, Clinical Pathology Division, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Valentina Summa
- Regina Elena National Cancer Institute, Clinical Pathology Division, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Mariantonia Carosi
- Regina Elena National Cancer Institute, Histopathology Department, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Antonello Vidiri
- Regina Elena National Cancer Institute, Radiology Department, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Alessandra Fabi
- Regina Elena National Cancer Institute, Medical Oncology Department, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Alessia Pasquale
- Regina Elena National Cancer Institute, Clinical Pathology Division, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Laura Conti
- Regina Elena National Cancer Institute, Clinical Pathology Division, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Immacolata Rosito
- Regina Elena National Cancer Institute, Clinical Pathology Division, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Carmine Maria Carapella
- Regina Elena National Cancer Institute, Neuro-Surgery Department, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Veronica Villani
- Regina Elena National Cancer Institute, Neuro-Oncology Division, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Andrea Pace
- Regina Elena National Cancer Institute, Neuro-Oncology Division, Via Elio Chianesi 53, 00144, Rome, Italy
| |
Collapse
|
14
|
Lee SJ, Lee CH, Choi SH, Ahn SH, Son BH, Lee JW, Yu JH, Kwon NJ, Lee WC, Yang KS, Lee DH, Han DY, Choi MS, Park PS, Lee HK, Kim MS, Lee J, Jeon BH. Evaluation of a novel approach to circulating tumor cell isolation for cancer gene panel analysis in patients with breast cancer. Oncol Lett 2017; 13:3025-3031. [PMID: 28521409 PMCID: PMC5431305 DOI: 10.3892/ol.2017.5807] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 11/11/2016] [Indexed: 01/03/2023] Open
Abstract
Liquid biopsy isolation of circulating tumor cells (CTCs) allows the genomic analysis of CTCs, which is useful in the determination of personalized cancer therapy. In the present study, CTCs from patients with breast cancer were enriched and successfully analyzed using cancer gene panel analysis. Blood samples from 11 patients with breast cancer were collected and CTCs enriched for using size-based filtration. The enriched CTCs were analyzed using immunofluorescence staining with antibodies directed against epithelial cell adhesion molecule (EpCAM) and cluster of differentiation 45. The genomic DNA of CTCs was extracted, amplified and 50 genes screened for mutations using the Ion AmpliSeq™ Cancer Hotspot Panel v2. EpCAM staining detected CTCs in 10/11 patients and the average CTC count was 3.9 in 5 ml blood. The average purity of enriched CTCs was 14.2±29.4% and the average amount of amplified DNA was 28.6±11.9 µg. Catalogue Of Somatic Mutations In Cancer mutations were detected in the CTCs and included IDH2, TP53, NRAS, IDH1, PDGFRA, HRAS, STK11, EGFR, PTEN, MLH1, PIK3CA, CDKN2A, KIT and SMARCB1. In conclusion, a novel size-based filtration approach for the isolation of CTCs was evaluated and successfully applied for the genomic analysis of CTCs from patients with breast cancer.
Collapse
Affiliation(s)
- Soo Jeong Lee
- Cytogen, Inc., Songpa-gu, Seoul 138-961, Republic of Korea
| | - Cham Han Lee
- Cytogen, Inc., Songpa-gu, Seoul 138-961, Republic of Korea
| | - Sung Ho Choi
- Cytogen, Inc., Songpa-gu, Seoul 138-961, Republic of Korea
| | - Sei Hyun Ahn
- Department of Surgery, Asan Medical Center Affiliated to The University of Uslan College of Medicine, Songpa-gu, Seoul 138-736, Republic of Korea
| | - Byung Ho Son
- Department of Surgery, Asan Medical Center Affiliated to The University of Uslan College of Medicine, Songpa-gu, Seoul 138-736, Republic of Korea
| | - Jong Won Lee
- Department of Surgery, Asan Medical Center Affiliated to The University of Uslan College of Medicine, Songpa-gu, Seoul 138-736, Republic of Korea
| | - Jong Han Yu
- Department of Surgery, Asan Medical Center Affiliated to The University of Uslan College of Medicine, Songpa-gu, Seoul 138-736, Republic of Korea
| | - Nak-Jung Kwon
- Macrogen, Inc., Geumcheon-gu, Seoul 08511, Republic of Korea
| | - Woo Chung Lee
- Macrogen, Inc., Geumcheon-gu, Seoul 08511, Republic of Korea
| | - Kap-Seok Yang
- Macrogen, Inc., Geumcheon-gu, Seoul 08511, Republic of Korea
| | | | - Du Yeol Han
- Cytogen, Inc., Songpa-gu, Seoul 138-961, Republic of Korea
| | - Mi So Choi
- Cytogen, Inc., Songpa-gu, Seoul 138-961, Republic of Korea
| | | | - Hyun Kyung Lee
- Cytogen, Inc., Songpa-gu, Seoul 138-961, Republic of Korea
| | | | - Jinseon Lee
- Cytogen, Inc., Songpa-gu, Seoul 138-961, Republic of Korea
| | - Byung Hee Jeon
- Cytogen, Inc., Songpa-gu, Seoul 138-961, Republic of Korea
| |
Collapse
|
15
|
Karagiannis GS, Goswami S, Jones JG, Oktay MH, Condeelis JS. Signatures of breast cancer metastasis at a glance. J Cell Sci 2016; 129:1751-8. [PMID: 27084578 DOI: 10.1242/jcs.183129] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gene expression profiling has yielded expression signatures from which prognostic tests can be derived to facilitate clinical decision making in breast cancer patients. Some of these signatures are based on profiling of whole tumor tissue (tissue signatures), which includes all tumor and stromal cells. Prognostic markers have also been derived from the profiling of metastasizing tumor cells, including circulating tumor cells (CTCs) and migratory-disseminating tumor cells within the primary tumor. The metastasis signatures based on CTCs and migratory-disseminating tumor cells have greater potential for unraveling cell biology insights and mechanistic underpinnings of tumor cell dissemination and metastasis. Of clinical interest is the promise that stratification of patients into high or low metastatic risk, as well as assessing the need for cytotoxic therapy, might be improved if prognostics derived from these two types of signatures are used in a combined way. The aim of this Cell Science at a Glance article and accompanying poster is to navigate through both types of signatures and their derived prognostics, as well as to highlight biological insights and clinical applications that could be derived from them, especially when they are used in combination.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sumanta Goswami
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joan G Jones
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
16
|
Lv Q, Gong L, Zhang T, Ye J, Chai L, Ni C, Mao Y. Prognostic value of circulating tumor cells in metastatic breast cancer: a systemic review and meta-analysis. Clin Transl Oncol 2016; 18:322-30. [PMID: 26260915 DOI: 10.1007/s12094-015-1372-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 07/28/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Metastatic breast cancer (MBC) remains the main cause of cancer-related death, and the clinical significance and prognostic role of circulating tumor cells (CTCs) in metastatic breast cancer are still controversial. Here, we conducted a meta-analysis to clarify the correlation between CTCs and the clinicopathological features and prognosis of MBC. METHODS We performed a comprehensive search of Pubmed and the ISI Web of Science through December 2014. Only articles that focused on MBC patients and detected CTCs using the CellSearch system were included. The associations between CTCs and survival rate and clinicopathological parameters, including molecular pattern, metastatic region and treatment response, were evaluated. RESULTS This meta-analysis included 24 studies (3701 MBC patients), 13 prospective studies and 11 retrospective studies. We found that CTCs were more frequently detected with HER2 + primary tumors (pooled RR = 0.73, 95 % CI = 0.63-0.84). Additionally, higher CTC numbers indicated a worse treatment response (RR = 0.56, 95 % CI = 0.40-0.79), poorer PFS (RR = 0.64, 95 % CI = 0.56-0.73) and poorer OS (RR = 0.69, 95 % CI = 0.64-0.75) in MBC patients. CONCLUSION Based on these results, we propose that HER2 positivity could be a significant risk factor for the presence of CTCs. Additionally, CTCs have a significant prognostic value for MBC patients. Therefore, CTCs should be continually monitored to guide the treatment of MBC patients, especially those with HER2 + primary tumors.
Collapse
Affiliation(s)
- Q Lv
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - L Gong
- Department of Otorhinolaryngology, Affiliated Cixi Hospital of Wenzhou Medical College, Cixi, 315300, China
| | - T Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - J Ye
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - L Chai
- Department of Otorhinolaryngology, First Affiliated Hospital, Medical School, Zhejiang University, Hangzhou, 310003, China
| | - C Ni
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, 310004, China
| | - Y Mao
- Department of Otorhinolaryngology, First Affiliated Hospital, Medical School, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
17
|
Gasparri ML, Savone D, Besharat RA, Farooqi AA, Bellati F, Ruscito I, Panici PB, Papadia A. Circulating tumor cells as trigger to hematogenous spreads and potential biomarkers to predict the prognosis in ovarian cancer. Tumour Biol 2015. [PMID: 26500096 DOI: 10.1007/s13277-015-4299-9] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Despite several improvements in the surgical field and in the systemic treatment, ovarian cancer (OC) is still characterized by high recurrence rates and consequently poor survival. In OC, there is still a great lack of knowledge with regard to cancer behavior and mechanisms of recurrence, progression, and drug resistance. The OC metastatization process mostly occurs via intracoelomatic spread. Recent evidences show that tumor cells generate a favorable microenvironment consisting in T regulatory cells, T infiltrating lymphocytes, and cytokines which are able to establish an "immuno-tolerance mileau" in which a tumor cell can become a resistant clone. When the disease responds to treatment, immunoediting processes and cancer progression have been stopped. A similar inhibition of the immunosuppressive microenvironment has been observed after optimal cytoreductive surgery as well. In this scenario, the early identification of circulating tumor cells could represent a precocious signal of loss of the immune balance that precedes cancer immunoediting and relapse. Supporting this hypothesis, circulating tumor cells have been demonstrated to be a prognostic factor in several solid tumors such as colorectal, pancreatic, gastric, breast, and genitourinary cancer. In OC, the role of circulating tumor cells is still to be defined. However, as opposed to healthy women, circulating tumor cells have been demonstrated in peripheral blood of OC patients, opening a new research field in OC diagnosis, treatment monitoring, and follow-up.
Collapse
Affiliation(s)
- Maria Luisa Gasparri
- Department of Gynecology Obstetrics and Urology, "Sapienza" University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy.
| | - Delia Savone
- Department of Gynecology Obstetrics and Urology, "Sapienza" University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy
| | - Raad Aris Besharat
- Department of Gynecology Obstetrics and Urology, "Sapienza" University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, Pakistan
| | - Filippo Bellati
- Department of Gynecology Obstetrics and Urology, "Sapienza" University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy
| | - Ilary Ruscito
- Department of Gynecology Obstetrics and Urology, "Sapienza" University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy
| | - Pierluigi Benedetti Panici
- Department of Gynecology Obstetrics and Urology, "Sapienza" University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy
| | - Andrea Papadia
- Department of Obstetrics and Gynecology, Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
18
|
Gasparri ML, Savone D, Besharat RA, Farooqi AA, Bellati F, Ruscito I, Panici PB, Papadia A. Circulating tumor cells as trigger to hematogenous spreads and potential biomarkers to predict the prognosis in ovarian cancer. Tumour Biol 2015; 37:71-5. [DOI: 10.1007/s13277-015-4299-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/20/2015] [Indexed: 12/19/2022] Open
|
19
|
Jo H, Her J, Ban C. Dual aptamer-functionalized silica nanoparticles for the highly sensitive detection of breast cancer. Biosens Bioelectron 2015; 71:129-136. [PMID: 25897882 DOI: 10.1016/j.bios.2015.04.030] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/26/2015] [Accepted: 04/12/2015] [Indexed: 02/07/2023]
Abstract
In this study, we synthesized dual aptamer-modified silica nanoparticles that simultaneously target two types of breast cancer cells: the mucin 1 (MUC1)(+) and human epidermal growth factor receptor 2 (HER2)(+) cell lines. Dual aptamer system enables a broad diagnosis for breast cancer in comparison with the single aptamer system. The dye-doped silica nanoparticles offer great stability with respect to photobleaching and enable the accurate quantification of breast cancer cells. The morphological and spectroscopic characteristics of the designed Dual-SiNPs were demonstrated via diverse methods such as DLS, zeta potential measurements, UV-vis spectroscopy, and fluorescence spectroscopy. Negatively charged Dual-SiNPs with a homogeneous size distribution showed robust and strong fluorescence. In addition, Dual-SiNPs did not affect cell viability, implying that this probe might be readily available for use in an in vivo system. Through ratio optimization of the MUC1 and HER2 aptamers, the binding capacities of the Dual-SiNPs to both cell lines were maximized. Based on Dual-SiNPs, a highly sensitive quantification of breast cancer cells was performed, resulting in a detection limit of 1 cell/100 μL, which is significantly lower compared with those reported in other studies. Moreover, the developed detection platform displayed high selectivity for only the MUC1(+) and HER2(+) cell lines. It is expected that this valuable diagnostic probe will be a noteworthy platform for the diagnosis and prognosis of breast cancer.
Collapse
Affiliation(s)
- Hunho Jo
- Department of Chemistry, Pohang University of Science and Technology, 77, Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk 790-784, South Korea.
| | - Jin Her
- Department of Chemistry, Pohang University of Science and Technology, 77, Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk 790-784, South Korea.
| | - Changill Ban
- Department of Chemistry, Pohang University of Science and Technology, 77, Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk 790-784, South Korea.
| |
Collapse
|
20
|
Zhu YY, Yuan Z. Pancreatic cancer stem cells. Am J Cancer Res 2015; 5:894-906. [PMID: 26045976 PMCID: PMC4449425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 02/10/2015] [Indexed: 06/04/2023] Open
Abstract
Studies are emerging in support of the cancer stem cells (CSCs) theory which considers that a tiny subset of cancer cells is exclusively responsible for the initiation and malignant behavior of a cancer. This cell population, also termed CSCs, possesses the capacity both to self-renew, producing progeny that have the identical tumorigenic potential, and to differentiate into the bulk of cancer cells, helping serve the formation of the tumor entities, which, altogether, build the hierarchically organized structure of a cancer. In this review, we try to articulate the complicated signaling pathways regulating the retention of the characteristics of pancreatic CSCs, and in the wake of which, we seek to offer insights into the CSCs-relevant targeted therapeutics which are, in the meantime, confronted with bigger challenges than ever.
Collapse
Affiliation(s)
- Ya-Yun Zhu
- Department of Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital Shanghai, 200233, China
| | - Zhou Yuan
- Department of Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital Shanghai, 200233, China
| |
Collapse
|
21
|
Stock C, Schwab A. Ion channels and transporters in metastasis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2638-46. [PMID: 25445667 DOI: 10.1016/j.bbamem.2014.11.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 11/03/2014] [Accepted: 11/07/2014] [Indexed: 12/23/2022]
Abstract
An elaborate interplay between ion channels and transporters, components of the cytoskeleton, adhesion molecules, and signaling cascades provides the basis for each major step of the metastatic cascade. Ion channels and transporters contribute to cell motility by letting through or transporting ions essential for local Ca2+, pH and--in cooperation with water permeable aquaporins--volume homeostasis. Moreover, in addition to the actual ion transport they, or their auxiliary subunits, can display non-conducting activities. They can exert kinase activity in order to phosphorylate cytoskeletal constituents or their associates. They can become part of signaling processes by permeating Ca2+, by generating local pH-nanodomains or by being final downstream effectors. A number of channels and transporters are found at focal adhesions, interacting directly or indirectly with proteins of the extracellular matrix, with integrins or with components of the cytoskeleton. We also include the role of aquaporins in cell motility. They drive the outgrowth of lamellipodia/invadopodia or control the number of β1 integrins in the plasma membrane. The multitude of interacting ion channels and transporters (called transportome) including the associated signaling events holds great potential as therapeutic target(s) for anticancer agents that are aimed at preventing metastasis. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Christian Stock
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany.
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, D-48149 Münster, Germany
| |
Collapse
|