1
|
Brozos-Vázquez EM, Rodríguez-López C, Cortegoso-Mosquera A, López-Landrove S, Muinelo-Romay L, García-González J, López-López R, León-Mateos L. Immunotherapy in patients with brain metastasis: advances and challenges for the treatment and the application of circulating biomarkers. Front Immunol 2023; 14:1221113. [PMID: 38022574 PMCID: PMC10654987 DOI: 10.3389/fimmu.2023.1221113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
The central nervous system (CNS) is one of the most frequent metastatic sites of various cancers, including lung cancer, breast cancer and melanoma. The development of brain metastases requires a specific therapeutic approach and is associated with high mortality and morbidity in cancer patients. Advances in precision medicine and the introduction in recent years of new drugs, such as immunotherapy, have made it possible to improve the prognosis of these patients by improving survival and quality of life. New diagnostic techniques such as liquid biopsy allow real-time monitoring of tumor evolution, providing molecular information on prognostic and predictive biomarkers of response to treatment in blood or other fluids. In this review, we perform an exhaustive update of the clinical trials that demonstrate the utility of immunotherapy in patients with brain metastases and the potential of circulating biomarkers to improving the results of efficacy and toxicity in this subgroup of patients.
Collapse
Affiliation(s)
- E M Brozos-Vázquez
- Medical Oncology Department, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
- Medical Oncology Department, Complexo Hospitalario Universitario de A Coruña, Santiago de Compostela, Spain
- ONCOMET, Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Spain
- CIBERONC, Madrid, Spain
| | - C Rodríguez-López
- Medical Oncology Department, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
- ONCOMET, Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Spain
- CIBERONC, Madrid, Spain
| | - A Cortegoso-Mosquera
- Medical Oncology Department, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
- ONCOMET, Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Spain
- CIBERONC, Madrid, Spain
| | - S López-Landrove
- ONCOMET, Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Spain
| | - L Muinelo-Romay
- ONCOMET, Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Spain
- CIBERONC, Madrid, Spain
| | - J García-González
- Medical Oncology Department, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
- ONCOMET, Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Spain
- CIBERONC, Madrid, Spain
| | - R López-López
- Medical Oncology Department, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
- ONCOMET, Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Spain
- CIBERONC, Madrid, Spain
| | - L León-Mateos
- Medical Oncology Department, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
- ONCOMET, Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Spain
- CIBERONC, Madrid, Spain
| |
Collapse
|
2
|
Vardas V, Ju JA, Christopoulou A, Xagara A, Georgoulias V, Kotsakis A, Alix-Panabières C, Martin SS, Kallergi G. Functional Analysis of Viable Circulating Tumor Cells from Triple-Negative Breast Cancer Patients Using TetherChip Technology. Cells 2023; 12:1940. [PMID: 37566019 PMCID: PMC10416943 DOI: 10.3390/cells12151940] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
Metastasis, rather than the growth of the primary tumor, accounts for approximately 90% of breast cancer patient deaths. Microtentacles (McTNs) formation represents an important mechanism of metastasis. Triple-negative breast cancer (TNBC) is the most aggressive subtype with limited targeted therapies. The present study aimed to isolate viable circulating tumor cells (CTCs) and functionally analyze them in response to drug treatment. CTCs from 20 TNBC patients were isolated and maintained in culture for 5 days. Biomarker expression was identified by immunofluorescence staining and VyCap analysis. Vinorelbine-induced apoptosis was evaluated based on the detection of M30-positive cells. Our findings revealed that the CTC absolute number significantly increased using TetherChips analysis compared to the number of CTCs in patients' cytospins (p = 0.006) providing enough tumor cells for drug evaluation. Vinorelbine treatment (1 h) on live CTCs led to a significant induction of apoptosis (p = 0.010). It also caused a significant reduction in Detyrosinated α-tubulin (GLU), programmed death ligand (PD-L1)-expressing CTCs (p < 0.001), and disruption of McTNs. In conclusion, this pilot study offers a useful protocol using TetherChip technology for functional analysis and evaluation of drug efficacy in live CTCs, providing important information for targeting metastatic dissemination at a patient-individualized level.
Collapse
Affiliation(s)
- Vasileios Vardas
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece;
| | - Julia A. Ju
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.A.J.); (S.S.M.)
| | | | - Anastasia Xagara
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.)
| | | | - Athanasios Kotsakis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Center of Montpellier, 34295 Montpellier, France;
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, CNRS, IRD, 34090 Montpellier, France
- European Liquid Biopsy Society (ELBS), 20246 Hamburg, Germany
| | - Stuart S. Martin
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.A.J.); (S.S.M.)
| | - Galatea Kallergi
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece;
| |
Collapse
|
3
|
Vardas V, Tolios A, Christopoulou A, Georgoulias V, Xagara A, Koinis F, Kotsakis A, Kallergi G. Immune Checkpoint and EMT-Related Molecules in Circulating Tumor Cells (CTCs) from Triple Negative Breast Cancer Patients and Their Clinical Impact. Cancers (Basel) 2023; 15:1974. [PMID: 37046635 PMCID: PMC10093450 DOI: 10.3390/cancers15071974] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Triple negative breast cancer (TNBC) is the most aggressive breast cancer subtype. There are few targeted therapies for these patients, leading to an unmet need for new biomarkers. The present study aimed to investigate the expression of PD-L1, CTLA-4, GLU, and VIM in CTCs of TNBC patients. Ninety-five patients were enrolled in this study: sixty-four TNBC and thirty-one luminal. Of these patients, 60 were in the early stage, while 35 had metastatic disease. Protein expression was identified by immunofluorescence staining experiments and VyCAP analysis. All the examined proteins were upregulated in TNBC patients. The expression of the GLU+VIM+CK+ phenotype was higher (50%) in metastatic TNBC compared to early TNBC patients (17%) (p = 0.005). Among all the BC patients, a significant correlation was found between PD-L1+CD45-CK+ and CTLA-4+CD45-CK+ phenotypes (Spearman test, p = 0.024), implying an important role of dual inhibition in BC. Finally, the phenotypes GLU+VIM+CK+ and PD-L1+CD45-CK+ were associated with shorter OS in TNBC patients (OS: log-rank p = 0.048, HR = 2.9, OS: log-rank p < 0.001, HR = 8.7, respectively). Thus, PD-L1, CTLA-4, GLU, and VIM constitute significant biomarkers in TNBC associated with patients' outcome, providing new therapeutic targets for this difficult breast cancer subtype.
Collapse
Affiliation(s)
- Vasileios Vardas
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece
| | - Anastasios Tolios
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece
| | | | | | - Anastasia Xagara
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece
| | - Filippos Koinis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Athanasios Kotsakis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Galatea Kallergi
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece
| |
Collapse
|
4
|
Rehman AU, Khan P, Maurya SK, Siddiqui JA, Santamaria-Barria JA, Batra SK, Nasser MW. Liquid biopsies to occult brain metastasis. Mol Cancer 2022; 21:113. [PMID: 35538484 PMCID: PMC9088117 DOI: 10.1186/s12943-022-01577-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/19/2022] [Indexed: 02/08/2023] Open
Abstract
Brain metastasis (BrM) is a major problem associated with cancer-related mortality, and currently, no specific biomarkers are available in clinical settings for early detection. Liquid biopsy is widely accepted as a non-invasive method for diagnosing cancer and other diseases. We have reviewed the evidence that shows how the molecular alterations are involved in BrM, majorly from breast cancer (BC), lung cancer (LC), and melanoma, with an inception in how they can be employed for biomarker development. We discussed genetic and epigenetic changes that influence cancer cells to breach the blood-brain barrier (BBB) and help to establish metastatic lesions in the uniquely distinct brain microenvironment. Keeping abreast with the recent breakthroughs in the context of various biomolecules detections and identifications, the circulating tumor cells (CTC), cell-free nucleotides, non-coding RNAs, secretory proteins, and metabolites can be pursued in human body fluids such as blood, serum, cerebrospinal fluid (CSF), and urine to obtain potential candidates for biomarker development. The liquid biopsy-based biomarkers can overlay with current imaging techniques to amplify the signal viable for improving the early detection and treatments of occult BrM.
Collapse
Affiliation(s)
- Asad Ur Rehman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Parvez Khan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Shailendra Kumar Maurya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | | | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA. .,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA.
| |
Collapse
|
5
|
PARP-1 Expression and BRCA1 Mutations in Breast Cancer Patients' CTCs. Cancers (Basel) 2022; 14:cancers14071731. [PMID: 35406503 PMCID: PMC8996866 DOI: 10.3390/cancers14071731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Recent estimates have shown that approx. 70% of individuals with BRCA1 mutations will develop breast cancer by the age of 70. To make matters worse, breast cancer patients with BRCA1 mutations are more likely to have the more aggressive triple-negative breast cancer. PARPs, belong to a family of nuclear enzymes, which are involved in many cellular processes, including DNA repair. PARP inhibitors have been approved for the treatment of BRCA-mutated breast cancer. The aim of the study was the determination of PARP-1 expression in the context of the presence of BRCA1 mutations in circulating tumor cells of breast cancer patients. PARP-1 (nuclear) expression and BRCA1 mutations were mainly detected in triple negative breast cancer patients, and the latter were correlated with decreased survival. Our data suggest that PARP-1, in conjunction with BRCA1, could potentially be used as (a) biomarker(s) for patients’ stratification. Abstract BRCA1 and PARP are involved in DNA damage repair pathways. BRCA1 mutations have been linked to higher likelihood of triple negative breast cancer (TNBC). The aim of the study was to determine PARP-1 expression and BRCA1 mutations in circulating tumor cells (CTCs) of BC patients. Fifty patients were enrolled: 23 luminal and 27 TNBC. PARP expression in CTCs was identified by immunofluorescence. Genotyping was performed by PCR-Sanger sequencing in the same samples. PARP-1 expression was higher in luminal (61%) and early BC (54%), compared to TNBC (41%) and metastatic (33%) patients. In addition, PARP-1 distribution was mostly cytoplasmic in luminal patients (p = 0.024), whereas it was mostly nuclear in TNBC patients. In cytokeratin (CK)-positive patients, those with the CK+PARP+ phenotype had longer overall survival (OS, log-rank p = 0.046). Overall, nine mutations were detected; M1 and M2 were completely new and M4, M7 and M8 were characterized as pathogenic. M7 and M8 were predominantly found in metastatic TNBC patients (p = 0.014 and p = 0.002). Thus, PARP-1 expression and increased mutagenic burden in TNBC patients’ CTCs, could be used as an indicator to stratify patients regarding therapeutic approaches.
Collapse
|
6
|
Grüntkemeier L, Khurana A, Bischoff FZ, Hoffmann O, Kimmig R, Moore M, Cotter P, Kasimir-Bauer S. Single HER2-positive tumor cells are detected in initially HER2-negative breast carcinomas using the DEPArray™-HER2-FISH workflow. Breast Cancer 2022; 29:487-497. [PMID: 35025065 PMCID: PMC9021056 DOI: 10.1007/s12282-022-01330-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/22/2021] [Indexed: 02/06/2023]
Abstract
Background In breast cancer (BC), overexpression of HER2 on the primary tumor (PT) is determined by immunohistochemistry (IHC) or fluorescence in situ hybridization (FISH) to stratify samples as negative, equivocal and positive to identify patients (pts) for anti-HER2 therapy. CAP/ASCO guidelines recommend FISH for analyzing HER2/neu (ERBB2) gene amplification and for resolving equivocal HER2 IHC results. However, pre-analytical and analytical aspects are often confounded by sample related limitations and tumor heterogeneity and HER2 expression may differ between the PT and circulating tumor cells (CTCs), the precursors of metastasis. We used a validation cohort of BC patients to establish a new DEPArray™-PT-HER2-FISH workflow for further application in a development cohort, characterized as PT-HER2-negative but CTC-HER2/neu-positive, to identify patients with PT-HER2 amplified cells not detected by routine pathology. Methods 50 µm FFPE tumor curls from the validation cohort (n = 49) and the development cohort (n = 25) underwent cutting, deparaffinization and antigen retrieval followed by dissociation into a single-cell suspension. After staining for cytokeratin, vimentin, DAPI and separation via DEPArray™, single cells were processed for HER2-FISH analysis to assess the number of chromosome 17 and HER2 loci signals for comparison, either with available IHC or conventional tissue section FISH. CTC-HER2/neu status was determined using the AdnaTest BreastCancer (QIAGEN, Hilden, Germany). Results Applying CAP/ASCO guidelines for HER2 evaluation of single PT cells, the comparison of routine pathology and DEPArray™-HER2-FISH analysis resulted in a concordance rate of 81.6% (40/49 pts) in the validation cohort and 84% (21/25 pts) in the development cohort, respectively. In the latter one, 4/25 patients had single HER2-positive tumor cells with 2/25 BC patients proven to be HER2-positive, despite being HER2-negative in routine pathology. The two other patients showed an equivocal HER2 status in the DEPArray™-HER2-FISH workflow but a negative result in routine pathology. Whereas all four patients with discordant HER2 results had already died, 17/21 patients with concordant HER2 results are still alive. Conclusions The DEPArray™ system allows pure tumor cell recovery for subsequent HER2/neu FISH analysis and is highly concordant with conventional pathology. For PT-HER2-negative patients, harboring HER2/neu-positive CTCs, this approach might allow caregivers to more effectively offer anti-HER2 treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s12282-022-01330-8.
Collapse
Affiliation(s)
- Lisa Grüntkemeier
- Department of Gynecology and Obstetrics, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | | | | | - Oliver Hoffmann
- Department of Gynecology and Obstetrics, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | | | | | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|
7
|
Duque G, Manterola C, Otzen T, Arias C, Galindo B, Mora M, Guerrero E, García N. Clinical utility of liquid biopsy in breast cancer: A systematic review. Clin Genet 2021; 101:285-295. [PMID: 34687555 DOI: 10.1111/cge.14077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022]
Abstract
Advancements in genetic sequencing techniques along with the identification of specific mutations and structural changes in multiple cancer genes, make it possible to identify circulating tumor cells and cell free nucleic acids as blood-based biomarkers, serving as a liquid biopsy (LB) with great utility for the diagnosis, treatment and follow-up of patients with neoplasms. This systematic review focuses on the clinical utility of LB in patients with breast cancer (BC). Articles published between 1990 and 2021 were included. Databases searched: Trip Database, WoS, EMBASE, PubMed, SCOPUS, and Clinical Keys. Variables studied: Publication year, country, number of cases, primary study design, LB detection methods, genes found, overall survival, disease-free survival, stage, response to treatment, clinical utility, BC molecular type, systemic treatment and methodological quality of primary studies. Of 2619 articles, 74 were retained representing 12 658 patients, mainly cohort studies (66.2%), the majority were from China (15%) and Japan (12.2%). All primary studies described clinical stage and type of systemic treatment used. Most used biomarker detection method: DNA (52.7%) and type of analysis: quantification of total cfDNA (35.1%). PIK3CA mutation was most frequent (62.9%). Evidence suggests clinically useful applications of BC. Though heterogeneous, publications suggest that LB will constitute part of the standard diagnostic-therapeutic process of BC.
Collapse
Affiliation(s)
- Galo Duque
- PhD Program in Medical Sciences, Universidad de La Frontera, Temuco, Chile.,Faculty of Medicine, Universidad del Azuay, Cuenca, Ecuador
| | - Carlos Manterola
- PhD Program in Medical Sciences, Universidad de La Frontera, Temuco, Chile.,Center of Excellence in Morphological and Surgical Studies (CEMyQ), Universidad de La Frontera, Temuco, Chile
| | - Tamara Otzen
- PhD Program in Medical Sciences, Universidad de La Frontera, Temuco, Chile.,Center of Excellence in Morphological and Surgical Studies (CEMyQ), Universidad de La Frontera, Temuco, Chile
| | - Cristina Arias
- Faculty of Medicine, Universidad del Azuay, Cuenca, Ecuador
| | - Bryan Galindo
- Faculty of Medicine, Universidad del Azuay, Cuenca, Ecuador
| | - Miriann Mora
- PhD Program in Medical Sciences, Universidad de La Frontera, Temuco, Chile.,Faculty of Medicine, Universidad del Azuay, Cuenca, Ecuador
| | - Enmanuel Guerrero
- PhD Program in Medical Sciences, Universidad de La Frontera, Temuco, Chile.,Solca Cancer Institute, Sociedad de Lucha Contra el Cáncer, Cuenca, Ecuador
| | - Nayeli García
- PhD Program in Medical Sciences, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
8
|
Schuster E, Taftaf R, Reduzzi C, Albert MK, Romero-Calvo I, Liu H. Better together: circulating tumor cell clustering in metastatic cancer. Trends Cancer 2021; 7:1020-1032. [PMID: 34481763 PMCID: PMC8541931 DOI: 10.1016/j.trecan.2021.07.001] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 01/30/2023]
Abstract
Circulating tumor cells (CTCs) are vital components of liquid biopsies for diagnosis of residual cancer, monitoring of therapy response, and prognosis of recurrence. Scientific dogma focuses on metastasis mediated by single CTCs, but advancement of CTC detection technologies has elucidated multicellular CTC clusters, which are associated with unfavorable clinical outcomes and a 20- to 100-fold greater metastatic potential than single CTCs. While the mechanistic understanding of CTC cluster formation is still in its infancy, multiple cell adhesion molecules and tight junction proteins have been identified that underlie the outperforming attributes of homotypic and heterotypic CTC clusters, such as cell survival, cancer stemness, and immune evasion. Future directions include high-resolution characterization of CTCs at multiomic levels for diagnostic/prognostic evaluations and targeted therapies.
Collapse
Affiliation(s)
- Emma Schuster
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rokana Taftaf
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Carolina Reduzzi
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mary K Albert
- Biomedical Visualization Graduate Program, Department of Biomedical and Health Information Sciences, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Isabel Romero-Calvo
- Biomedical Visualization Graduate Program, Department of Biomedical and Health Information Sciences, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Huiping Liu
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Lurie Comprehensive Cancer Center and Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
9
|
Chantzara E, Xenidis N, Kallergi G, Georgoulias V, Kotsakis A. Circulating tumor cells as prognostic biomarkers in breast cancer: current status and future prospects. Expert Rev Mol Diagn 2021; 21:1037-1048. [PMID: 34328384 DOI: 10.1080/14737159.2021.1962710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction : Despite advances in diagnostic and therapeutic techniques breast cancer is still associated with significant morbidity and mortality. CTCs play a crucial role in the metastatic process, which is the main cause of death in BC patients.Areas covered : This review discusses the prognostic and predictive value of CTCs and their prospective in management of BC patients.Expert opinion : The analysis of CTCs through improved technologies offers a new insight into the metastatic cascade. Assessment of the number and molecular profile of CTCs holds great promises for disease monitoring and therapeutic decisions. However, more research is needed until they can be used in therapeutic decisions in clinical practice.
Collapse
Affiliation(s)
- Evagelia Chantzara
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece
| | - Nikolaos Xenidis
- Department of Medical Oncology, University General Hospital of Alexandroupolis, Alexandroupolis, Thrace, Greece
| | - Galatea Kallergi
- Division of Genetics, Cell and Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Vassilis Georgoulias
- Department of Medical Oncology, Hellenic Oncology Research Group (HORG), Athens, Greece
| | - Athanasios Kotsakis
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece.,Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Thessaly, Greece
| |
Collapse
|
10
|
Strati A, Zavridou M, Kallergi G, Politaki E, Kuske A, Gorges TM, Riethdorf S, Joosse SA, Koch C, Bohnen AL, Mueller V, Koutsodontis G, Kontopodis E, Poulakaki N, Psyrri A, Mavroudis D, Georgoulias V, Pantel K, Lianidou ES. A Comprehensive Molecular Analysis of in Vivo Isolated EpCAM-Positive Circulating Tumor Cells in Breast Cancer. Clin Chem 2021; 67:1395-1405. [PMID: 34322698 DOI: 10.1093/clinchem/hvab099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/13/2021] [Indexed: 11/12/2022]
Abstract
BACKGROUND Circulating tumor cell (CTC) analysis is highly promising for liquid biopsy-based molecular diagnostics. We undertook a comprehensive molecular analysis of in vivo isolated CTCs in breast cancer (BrCa). METHODS In vivo isolated CTCs from 42 patients with early and 23 patients with metastatic breast cancer (MBC) were prospectively collected and analyzed for gene expression, DNA mutations, and DNA methylation before and after treatment. 19 healthy donor (HD) samples were analyzed as a control group. In identical blood draws, CTCs were enumerated using CellSearch® and characterized by direct IF staining. RESULTS All 19 HD samples were negative for CK8, CK18, CK19, ERBB2, TWIST1, VEGF, ESR1, PR, and EGFR expression, while CD44, CD24, ALDH1, VIM, and CDH2 expression was normalized to B2M (reference gene). At least one gene was expressed in 23/42 (54.8%) and 8/13 (61.5%) CTCs in early BrCa before and after therapy, and in 20/23 (87.0%) and 5/7 (71.4%) MBC before and after the first cycle of therapy. PIK3CA mutations were detected in 11/42 (26.2%) and 3/13 (23.1%) in vivo isolated CTCs in early BrCa before and after therapy, and in 11/23 (47.8%) and 2/7 (28.6%) MBC, respectively. ESR1 methylation was detected in 5/32 (15.7%) and 1/10 (10.0%) CTCs in early BrCa before and after therapy, and in 3/15(20.0%) MBC before the first line of therapy. The comprehensive molecular analysis of CTC revealed a higher sensitivity in relation to CellSearch or IF staining when based on creatine kinase selection. CONCLUSIONS In vivo-CTC isolation in combination with a comprehensive molecular analysis at the gene expression, DNA mutation, and DNA methylation level comprises a highly powerful approach for molecular diagnostic applications using CTCs.
Collapse
Affiliation(s)
- Areti Strati
- Department of Chemistry, Analysis of Circulating Tumor Cells Laboratory, University of Athens, Greece
| | - Martha Zavridou
- Department of Chemistry, Analysis of Circulating Tumor Cells Laboratory, University of Athens, Greece
| | - Galatea Kallergi
- Laboratory of Τumor Cell Biology, School of Medicine, University of Crete, Heraklion, Crete, Greece.,Division of Genetics, Cell, and Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Eleni Politaki
- Laboratory of Τumor Cell Biology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Andra Kuske
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias M Gorges
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Riethdorf
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Koch
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna-Lena Bohnen
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volkmar Mueller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - George Koutsodontis
- Oncology Unit, Second Department of Internal Medicine, Attikon University Hospital, Haidari, Greece
| | - Emmanouil Kontopodis
- Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece
| | | | - Amanda Psyrri
- Oncology Unit, Second Department of Internal Medicine, Attikon University Hospital, Haidari, Greece
| | - Dimitris Mavroudis
- Laboratory of Τumor Cell Biology, School of Medicine, University of Crete, Heraklion, Crete, Greece.,Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece
| | - Vasilis Georgoulias
- Laboratory of Τumor Cell Biology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Evi S Lianidou
- Department of Chemistry, Analysis of Circulating Tumor Cells Laboratory, University of Athens, Greece
| |
Collapse
|
11
|
Yu T, Wang C, Xie M, Zhu C, Shu Y, Tang J, Guan X. Heterogeneity of CTC contributes to the organotropism of breast cancer. Biomed Pharmacother 2021; 137:111314. [PMID: 33581649 DOI: 10.1016/j.biopha.2021.111314] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/11/2021] [Accepted: 01/21/2021] [Indexed: 02/08/2023] Open
Abstract
Circulating tumor cells (CTCs) are viewed as pro-metastasis precursors shed from primary tumors or metastatic sites. The phenotypic and molecular heterogeneity of CTCs is associated with breast cancer progression and prognosis. Therefore, we divided CTCs into several subtypes according to their differences in biomarker status, epithelial/mesenchymal phenotype, aggregation status, and other factors to summarize their characteristics. Considering that the organ-specific metastasis is a hallmark of breast cancer, we adopted the "seed and soil" model to further analyze the relationship between the heterogeneity of CTCs and the organotropism of breast cancer. We speculated that CTCs might not only develop their genetic potential but communicate with surroundings, including chemokine systems, hemocytes, and extracellular matrix components, to regulate the organ-specific metastases of breast cancer.
Collapse
Affiliation(s)
- Tao Yu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Cenzhu Wang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Mengyan Xie
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Chengjun Zhu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Yongqian Shu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Jinhai Tang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China.
| | - Xiaoxiang Guan
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China.
| |
Collapse
|
12
|
Spiliotaki M, Kallergi G, Nikolaou C, Xenidis N, Politaki E, Apostolaki S, Georgoulia N, Koinis F, Tsoukalas N, Hatzidaki D, Kotsakis A, Georgoulias V. Dynamic changes of CTCs in patients with metastatic HR(+)/HER2(-) breast cancer receiving salvage treatment with everolimus/exemestane. Cancer Chemother Pharmacol 2021; 87:277-287. [PMID: 33515073 DOI: 10.1007/s00280-020-04227-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 12/29/2020] [Indexed: 01/23/2023]
Abstract
PURPOSE Detection of CTCs represents a poor prognostic factor in patients with early and metastatic breast cancer (mBC) and treatment with everolimus-exemestane (E/E) is an established effective treatment in hormone receptor-positive/HER2-negative mBC patients. The effect of E/E on CTCs in mBC patients was prospectively investigated. METHODS CTCs from 50 pre-treated patients with mBC receiving E/E were analyzed using the CellSearch (CS) platform and triple immunofluorescence (IF) staining for cytokeratin, M30 and Ki67 expression to assess their proliferative and apoptotic status. RESULTS CTCs (by CS) were detected in 64% of patients before treatment and E/E administration resulted in their decreased prevalence [(n = 18; 36%, p = 0.004) and (n = 7; 19.4%, p = 0.019) post-1st and post-3rd treatment cycle, respectively] whereas it was significantly increased at disease progression (PD: 61%) compared to post-1st and post-3rd cycle (p = 0.049 and p = 0.021, respectively). Ki67-positive CTCs were detected in 60%, 60%, 17% and 50% of patients before treatment, post-1st, post-3rd cycle and at PD, respectively, while the opposite was observed for M30-positive CTCs (0% at baseline, 10% after the 1st cycle, 50% after the 3rd cycle and 0% at PD). The detection of even ≥ 1 CTC/5 ml after one cycle was associated with decreased PFS (3.3 vs 9.0 months, p = 0.025) whereas the detection of even ≥ 2 CTCs at PD was associated with decreased OS (32.4 vs 19.5 months; p = 0.009). CONCLUSIONS The combination of E/E resulted in early elimination of proliferating CTCs in mBC patients and this effect was associated with a favorable clinical outcome.
Collapse
Affiliation(s)
- Maria Spiliotaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Galatea Kallergi
- Department of Biochemistry, University of Crete Medical School, Heraklion, Crete, Greece
| | - Christos Nikolaou
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Nikolaos Xenidis
- Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece.,Department of Medical Oncology, Medical School, Democritus University of Thrace, Xanthi, Greece
| | - Eleni Politaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Stella Apostolaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Nefeli Georgoulia
- Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece
| | - Filippos Koinis
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece.,Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece.,Department of Medical Oncology, Faculty of Medicine, School of Health Sciences, University Hospital of Larissa & Laboratory of Oncology, University of Thessaly Mezourlo, Larissa, Thessaly, Greece
| | - Nikolaos Tsoukalas
- Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece.,Medical Oncology Unit, NIMITS Hospital, Athens, Greece
| | - Dora Hatzidaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece.,Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece
| | - Athanasios Kotsakis
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece.,Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece.,Department of Medical Oncology, Faculty of Medicine, School of Health Sciences, University Hospital of Larissa & Laboratory of Oncology, University of Thessaly Mezourlo, Larissa, Thessaly, Greece
| | - Vassilis Georgoulias
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece. .,Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece.
| |
Collapse
|
13
|
Amelio I, Bertolo R, Bove P, Buonomo OC, Candi E, Chiocchi M, Cipriani C, Di Daniele N, Ganini C, Juhl H, Mauriello A, Marani C, Marshall J, Montanaro M, Palmieri G, Piacentini M, Sica G, Tesauro M, Rovella V, Tisone G, Shi Y, Wang Y, Melino G. Liquid biopsies and cancer omics. Cell Death Discov 2020; 6:131. [PMID: 33298891 PMCID: PMC7691330 DOI: 10.1038/s41420-020-00373-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
The development of the sequencing technologies allowed the generation of huge amounts of molecular data from a single cancer specimen, allowing the clinical oncology to enter the era of the precision medicine. This massive amount of data is highlighting new details on cancer pathogenesis but still relies on tissue biopsies, which are unable to capture the dynamic nature of cancer through its evolution. This assumption led to the exploration of non-tissue sources of tumoral material opening the field of liquid biopsies. Blood, together with body fluids such as urines, or stool, from cancer patients, are analyzed applying the techniques used for the generation of omics data. With blood, this approach would allow to take into account tumor heterogeneity (since the circulating components such as CTCs, ctDNA, or ECVs derive from each cancer clone) in a time dependent manner, resulting in a somehow "real-time" understanding of cancer evolution. Liquid biopsies are beginning nowdays to be applied in many cancer contexts and are at the basis of many clinical trials in oncology.
Collapse
Affiliation(s)
- Ivano Amelio
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy.
- School of Life Sciences, University of Nottingham, Nottingham, UK.
| | - Riccardo Bertolo
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
- San Carlo di Nancy Hospital, Rome, Italy
| | - Pierluigi Bove
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
- San Carlo di Nancy Hospital, Rome, Italy
| | - Oreste Claudio Buonomo
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Eleonora Candi
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Marcello Chiocchi
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Chiara Cipriani
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
- San Carlo di Nancy Hospital, Rome, Italy
| | - Nicola Di Daniele
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Carlo Ganini
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | | | - Alessandro Mauriello
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Carla Marani
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
- San Carlo di Nancy Hospital, Rome, Italy
| | - John Marshall
- Medstar Georgetown University Hospital, Georgetown University, Washington, DC, USA
| | - Manuela Montanaro
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Giampiero Palmieri
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Mauro Piacentini
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Giuseppe Sica
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Manfredi Tesauro
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Valentina Rovella
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Giuseppe Tisone
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Yufang Shi
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, 200031, Shanghai, China
- The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, 215123, Suzhou, Jiangsu, China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, 200031, Shanghai, China
| | - Gerry Melino
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
14
|
Nouri Z, Fakhri S, Nouri K, Wallace CE, Farzaei MH, Bishayee A. Targeting Multiple Signaling Pathways in Cancer: The Rutin Therapeutic Approach. Cancers (Basel) 2020; 12:E2276. [PMID: 32823876 PMCID: PMC7463935 DOI: 10.3390/cancers12082276] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022] Open
Abstract
Multiple dysregulated signaling pathways are implicated in the pathogenesis of cancer. The conventional therapies used in cancer prevention/treatment suffer from low efficacy, considerable toxicity, and high cost. Hence, the discovery and development of novel multi-targeted agents to attenuate the dysregulated signaling in cancer is of great importance. In recent decades, phytochemicals from dietary and medicinal plants have been successfully introduced as alternative anticancer agents due to their ability to modulate numerous oncogenic and oncosuppressive signaling pathways. Rutin (also known as rutoside, quercetin-3-O-rutinoside and sophorin) is an active plant-derived flavonoid that is widely distributed in various vegetables, fruits, and medicinal plants, including asparagus, buckwheat, apricots, apples, cherries, grapes, grapefruit, plums, oranges, and tea. Rutin has been shown to target various inflammatory, apoptotic, autophagic, and angiogenic signaling mediators, including nuclear factor-κB, tumor necrosis factor-α, interleukins, light chain 3/Beclin, B cell lymphoma 2 (Bcl-2), Bcl-2 associated X protein, caspases, and vascular endothelial growth factor. A comprehensive and critical analysis of the anticancer potential of rutin and associated molecular targets amongst various cancer types has not been performed previously. Accordingly, the purpose of this review is to present an up-to-date and critical evaluation of multiple cellular and molecular mechanisms through which the anticancer effects of rutin are known to be exerted. The current challenges and limitations as well as future directions of research are also discussed.
Collapse
Affiliation(s)
- Zeinab Nouri
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran;
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Keyvan Nouri
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran;
| | - Carly E. Wallace
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| |
Collapse
|
15
|
Ivanova E, Ward A, Wiegmans AP, Richard DJ. Circulating Tumor Cells in Metastatic Breast Cancer: From Genome Instability to Metastasis. Front Mol Biosci 2020; 7:134. [PMID: 32766277 PMCID: PMC7378584 DOI: 10.3389/fmolb.2020.00134] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
The emergence of clinical resistance in repeatedly treated cancers extends from the primary tumor's capability to exploit genome instability to adapt, escape, and progress. Triple negative breast cancer serves as a good example of such a response demonstrating poor clinical outcome due to a high rate of cellular heterogeneity resulting in metastatic relapse. The capability to effectively track the emergence of therapeutic resistance in real-time and adapt the clinical response is the holy grail for precision medicine and has yet to be realized. In this review we present liquid biopsy using CTCs and ctDNA as a potential replacement and/or addition to the current diagnostic tests to deliver personalized therapies to patients with advanced breast cancer. We outline current uses of liquid biopsy in the metastatic breast cancer setting and discuss their limitations. In addition, we provide a detailed overview of common genome instability events in patients with metastatic breast cancer and how these can be tracked using liquid biopsy.
Collapse
Affiliation(s)
- Ekaterina Ivanova
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Woolongabba, QLD, Australia.,Centre for Tumour and Immune Biology (ZTI), Philipps University Marburg, Marburg, Germany
| | - Ambber Ward
- Tumor Microenvironment Laboratory, QIMR Berghofer, Herston, QLD, Australia
| | - Adrian P Wiegmans
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Woolongabba, QLD, Australia
| | - Derek John Richard
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Woolongabba, QLD, Australia
| |
Collapse
|
16
|
Bittner AK, Keup C, Hoffmann O, Hauch S, Kimmig R, Kasimir-Bauer S. Molecular characterization of circulating tumour cells identifies predictive markers for outcome in primary, triple-negative breast cancer patients. J Cell Mol Med 2020; 24:8405-8416. [PMID: 32558176 PMCID: PMC7412423 DOI: 10.1111/jcmm.15349] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 08/08/2019] [Accepted: 02/08/2020] [Indexed: 12/13/2022] Open
Abstract
mRNA profiles of circulating tumour cells (CTCs) were analysed in patients with triple‐negative breast cancer (TNBC) (pts) before (BT) and after therapy (AT) to identify additional treatment options. 2 × 5 mL blood of 51 TNBC pts and 24 non‐TNBC pts (HR+/HER2−; HR−/HER2+) was analysed for CTCs using the AdnaTest EMT‐2/Stem Cell Select™, followed by mRNA isolation and cDNA analysis for 17 genes by qPCR PIK3CA, AKT2, MTOR and the resistance marker AURKA and ERCC1 were predominantly expressed in all breast cancer subtypes, the latter ones especially AT. In TNBC pts, ERBB3, EGFR, SRC, NOTCH, ALK and AR were uniquely present and ERBB2+/ERBB3 + CTCs were found BT and AT in about 20% of cases. EGFR+/ERBB2+/ERBB3 + CTCs BT and ERBB2+/ERBB3 + CTCs AT significantly correlated with a shorter progression‐free survival (PFS; P = 0.01 and P = 0.02). Platinum‐based therapy resulted in a reduced PFS (P = 0.02) and an induction of PIK3CA expression in CTCs AT. In non‐TNBC pts, BT, the expression pattern in CTCs was similar. AURKA+/ERCC1 + CTCs were found in 40% of HR−/HER2 + pts BT and AT. In the latter group, NOTCH, PARP1 and SRC1 were only present AT and ERBB2 + CTCs completely disappeared AT. These findings might help to predict personalized therapy for TNBC pts in the future.
Collapse
Affiliation(s)
- Ann-Kathrin Bittner
- Department of Gynecology and Obstetrics, University Hospital of Essen, Essen, Germany
| | - Corinna Keup
- Department of Gynecology and Obstetrics, University Hospital of Essen, Essen, Germany
| | - Oliver Hoffmann
- Department of Gynecology and Obstetrics, University Hospital of Essen, Essen, Germany
| | | | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital of Essen, Essen, Germany
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital of Essen, Essen, Germany
| |
Collapse
|
17
|
Abreu M, Cabezas-Sainz P, Pereira-Veiga T, Falo C, Abalo A, Morilla I, Curiel T, Cueva J, Rodríguez C, Varela-Pose V, Lago-Lestón R, Mondelo P, Palacios P, Moreno-Bueno G, Cano A, García-Caballero T, Pujana MÁ, Sánchez-Piñón L, Costa C, López R, Muinelo-Romay L. Looking for a Better Characterization of Triple-Negative Breast Cancer by Means of Circulating Tumor Cells. J Clin Med 2020; 9:E353. [PMID: 32012729 PMCID: PMC7074553 DOI: 10.3390/jcm9020353] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
Traditionally, studies to address the characterization of mechanisms promoting tumor aggressiveness and progression have been focused only on primary tumor analyses, which could provide relevant information but have limitations to really characterize the more aggressive tumor population. To overcome these limitations, circulating tumor cells (CTCs) represent a noninvasive and valuable tool for real-time profiling of disseminated tumor cells. Therefore, the aim of the present study was to explore the value of CTC enumeration and characterization to identify markers associated with the outcome and the aggressiveness of triple-negative breast cancer (TNBC). For that aim, the CTC population from 32 patients diagnosed with TNBC was isolated and characterized. This population showed important cell plasticity in terms of expression of epithelia/mesenchymal and stemness markers, suggesting the relevance of epithelial to mesenchymal transition (EMT) intermediate phenotypes for efficient tumor dissemination. Importantly, the CTC signature demonstrated prognostic value to predict the patients' outcome and pointed to a relevant role of tissue inhibitor of metalloproteinases 1 (TIMP1) and androgen receptor (AR) for TNBC biology. Furthermore, we also analyzed the usefulness of the AR and TIMP1 blockade to target TNBC proliferation and dissemination using in vitro and in vivo zebra fish and mouse models. Overall, the molecular characterization of CTCs from advanced TNBC patients identifies highly specific biomarkers with potential applicability as noninvasive prognostic markers and reinforced the value of TIMP1 and AR as potential therapeutic targets to tackle the most aggressive breast cancer.
Collapse
Affiliation(s)
- Manuel Abreu
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.L.-L.); (P.M.); (R.L.)
| | - Pablo Cabezas-Sainz
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (L.S.-P.)
| | - Thais Pereira-Veiga
- Roche-CHUS Joint Unit, Oncomet, Health Research Institute of Santiago (IDIS), Complejo Hospitalario de Santiago de Compostela, Trav. Choupana s/n, 15706 Santiago de Compostela, Spain; (T.P.-V.); (C.C.)
| | - Catalina Falo
- Department of Medical Oncology-Breast Cancer Unit, Institut Català d’Oncologia (ICO)-Hospitalet-Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, 08007 Barcelona, Spain; (C.F.); (I.M.)
| | - Alicia Abalo
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.L.-L.); (P.M.); (R.L.)
| | - Idoia Morilla
- Department of Medical Oncology-Breast Cancer Unit, Institut Català d’Oncologia (ICO)-Hospitalet-Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, 08007 Barcelona, Spain; (C.F.); (I.M.)
| | - Teresa Curiel
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (T.C.); (J.C.); (C.R.); (V.V.-P.); (P.P.)
| | - Juan Cueva
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (T.C.); (J.C.); (C.R.); (V.V.-P.); (P.P.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos 3-5, 28029 Madrid, Spain; (G.M.-B.); (A.C.)
| | - Carmela Rodríguez
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (T.C.); (J.C.); (C.R.); (V.V.-P.); (P.P.)
| | - Vanesa Varela-Pose
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (T.C.); (J.C.); (C.R.); (V.V.-P.); (P.P.)
| | - Ramón Lago-Lestón
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.L.-L.); (P.M.); (R.L.)
| | - Patricia Mondelo
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.L.-L.); (P.M.); (R.L.)
| | - Patricia Palacios
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (T.C.); (J.C.); (C.R.); (V.V.-P.); (P.P.)
| | - Gema Moreno-Bueno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos 3-5, 28029 Madrid, Spain; (G.M.-B.); (A.C.)
- Fundación MD Anderson Internacional, C/Gómez Hemans 2, 28033 Madrid, Spain
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), IdiPaz, Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Amparo Cano
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos 3-5, 28029 Madrid, Spain; (G.M.-B.); (A.C.)
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), IdiPaz, Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Tomás García-Caballero
- Departamento de Ciencias Morfológicas, Facultad de Medicina, Universidad de Santiago, Servicio de Anatomía Patológica, Complejo Hospitalario Universitario de Santiago, 15706 Santiago de Compostela, Spain;
| | - Miquel Ángel Pujana
- ProCURE, Catalan Institute of Oncology (ICO), Bellvitge Institute of Biomedical Research (IDIBELL), 08908 Barcelona, Spain;
| | - Laura Sánchez-Piñón
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (L.S.-P.)
| | - Clotilde Costa
- Roche-CHUS Joint Unit, Oncomet, Health Research Institute of Santiago (IDIS), Complejo Hospitalario de Santiago de Compostela, Trav. Choupana s/n, 15706 Santiago de Compostela, Spain; (T.P.-V.); (C.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos 3-5, 28029 Madrid, Spain; (G.M.-B.); (A.C.)
| | - Rafael López
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.L.-L.); (P.M.); (R.L.)
- Roche-CHUS Joint Unit, Oncomet, Health Research Institute of Santiago (IDIS), Complejo Hospitalario de Santiago de Compostela, Trav. Choupana s/n, 15706 Santiago de Compostela, Spain; (T.P.-V.); (C.C.)
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (T.C.); (J.C.); (C.R.); (V.V.-P.); (P.P.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos 3-5, 28029 Madrid, Spain; (G.M.-B.); (A.C.)
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (M.A.); (A.A.); (R.L.-L.); (P.M.); (R.L.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos 3-5, 28029 Madrid, Spain; (G.M.-B.); (A.C.)
| |
Collapse
|
18
|
Jacot W, Cottu P, Berger F, Dubot C, Venat-Bouvet L, Lortholary A, Bourgeois H, Bollet M, Servent V, Luporsi E, Espié M, Guiu S, D'Hondt V, Dieras V, Sablin MP, Brain E, Neffati S, Pierga JY, Bidard FC. Actionability of HER2-amplified circulating tumor cells in HER2-negative metastatic breast cancer: the CirCe T-DM1 trial. Breast Cancer Res 2019; 21:121. [PMID: 31727113 PMCID: PMC6854749 DOI: 10.1186/s13058-019-1215-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/15/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In this prospective phase 2 trial, we assessed the efficacy of trastuzumab-emtansine (T-DM1) in HER2-negative metastatic breast cancer (MBC) patients with HER2-positive CTC. METHODS Main inclusion criteria for screening were as follows: women with HER2-negative MBC treated with ≥ 2 prior lines of chemotherapy and measurable disease. CTC with a HER2/CEP17 ratio of ≥ 2.2 by fluorescent in situ hybridization (CellSearch) were considered to be HER2-amplified (HER2amp). Patients with ≥ 1 HER2amp CTC were eligible for the treatment phase (T-DM1 monotherapy). The primary endpoint was the overall response rate. RESULTS In 154 screened patients, ≥ 1 and ≥ 5 CTC/7.5 ml of blood were detected in N = 118 (78.7%) and N = 86 (57.3%) patients, respectively. ≥1 HER2amp CTC was found in 14 patients (9.1% of patients with ≥ 1 CTC/7.5 ml). Among 11 patients treated with T-DM1, one achieved a confirmed partial response. Four patients had a stable disease as best response. Median PFS was 4.8 months while median OS was 9.5 months. CONCLUSIONS CTC with HER2 amplification can be detected in a limited subset of HER2-negative MBC patients. Treatment with T-DM1 achieved a partial response in only one patient. TRIAL REGISTRATION NCT01975142, Registered 03 November 2013.
Collapse
Affiliation(s)
- William Jacot
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France.,Montpellier University, Montpellier, France
| | - Paul Cottu
- Department of Medical Oncology, Institut Curie, PSL Research University, Saint Cloud, France
| | - Frederique Berger
- Biometry and Clinical Trial Promotion Units, Institut Curie, PSL Research University, Saint Cloud, France
| | - Coraline Dubot
- Department of Medical Oncology, Institut Curie, PSL Research University, Saint Cloud, France
| | | | - Alain Lortholary
- Department of Medical Oncology, Centre Catherine de Sienne, Nantes, France
| | - Hugues Bourgeois
- Department of Medical Oncology, Clinique Victor Hugo, Le Mans, France
| | - Marc Bollet
- Department of Radiation Therapy, Clinique Hartmann, Neuilly, France
| | | | - Elisabeth Luporsi
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Nancy, France
| | - Marc Espié
- Department of Medical Oncology, Hôpital Saint Louis, Paris, France
| | - Severine Guiu
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Veronique D'Hondt
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Veronique Dieras
- Department of Medical Oncology, Institut Curie, PSL Research University, Saint Cloud, France
| | - Marie-Paule Sablin
- Department of Medical Oncology, Institut Curie, PSL Research University, Saint Cloud, France
| | - Etienne Brain
- Department of Medical Oncology, Institut Curie, PSL Research University, Saint Cloud, France
| | - Souhir Neffati
- Biometry and Clinical Trial Promotion Units, Institut Curie, PSL Research University, Saint Cloud, France
| | - Jean-Yves Pierga
- Department of Medical Oncology, Institut Curie, PSL Research University, Saint Cloud, France.,Université Paris Descartes, Paris, France.,Laboratory of Circulating Tumor Biomarkers, Institut Curie, PSL Research University, Paris, France
| | - Francois-Clement Bidard
- Department of Medical Oncology, Institut Curie, PSL Research University, Saint Cloud, France. .,Université Paris Descartes, Paris, France. .,UVSQ, Paris Saclay University, Saint Cloud, France.
| |
Collapse
|
19
|
Fabbri F, Salvi S, Bravaccini S. Know your enemy: Genetics, aging, exposomic and inflammation in the war against triple negative breast cancer. Semin Cancer Biol 2019; 60:285-293. [PMID: 31669505 DOI: 10.1016/j.semcancer.2019.10.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 01/11/2023]
Abstract
Triple negative breast cancer (TNBC) is one of the most biologically aggressive and very often lethal breast disease. It is one of the most puzzling women malignancies, and it currently appears not to be a good candidate to a standardized, unanimously accepted and sufficiently active therapeutic strategy. Fast proliferating and poorly differentiated, it is histopathologically heterogeneous, and even more ambiguous at the molecular level, offering few recurrent actionable targets to the clinicians. It is a formidable and vicious enemy that requires a huge investigational effort to find its vital weak spots. Here, we provide a broad review of "old but gold" biological aspects that taken together may help in finding new TNBC management strategies. A better and updated knowledge of the origins, war-like tactics, refueling mechanisms and escape routes of TNBC, will help in moving the decisive steps towards its final defeat.
Collapse
Affiliation(s)
- Francesco Fabbri
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Samanta Salvi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| | - Sara Bravaccini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
20
|
Vishnoi M, Liu NH, Yin W, Boral D, Scamardo A, Hong D, Marchetti D. The identification of a TNBC liver metastasis gene signature by sequential CTC-xenograft modeling. Mol Oncol 2019; 13:1913-1926. [PMID: 31216110 PMCID: PMC6717757 DOI: 10.1002/1878-0261.12533] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 06/17/2019] [Indexed: 12/23/2022] Open
Abstract
Triple-negative breast cancer (TNBC) liver metastasis is associated with poor prognosis and low patient survival. It occurs when tumor cells disseminate from primary tumors, circulate in blood/lymph [circulating tumor cells (CTCs)], and acquire distinct characteristics during disease progression toward the metastatic phenotype. The purpose of this study was to decipher the genomic/transcriptomic properties of TNBC liver metastasis and its recurrence for potential therapeutic targeting. We employed a negative depletion strategy to isolate and interrogate CTCs from the blood of patients with TNBC, and to establish sequential generations of CTC-derived xenografts (CDXs) through injection of patient CTCs in immunodeficient mice. The isolation and validation of CDX-derived cell populations [analyses of CTCs were paired with bone marrow-resident cells (BMRTCs) and liver tissue cells obtained from the same animal] were performed by multiparametric flow cytometry, immune phenotyping, and genomic sequencing of putative CTCs. Comprehensive characterization of gene expression arrays from sequentially generated CDX-derived cell populations, online gene expression arrays, and TCGA databases were employed to discover a CTC-driven, liver metastasis-associated TNBC signature. We discovered a distinct transcriptomic signature of TNBC patient-isolated CTCs from primary TNBCs, which was consistent throughout sequential CDX modeling. We established a novel TNBC liver metastasis-specific CDX model that selectively recapitulates CTC biology for four sequential generations of mice. The evaluation of online databases and CDX-derived populations revealed 597 genes specific to the TNBC liver metastasis signatures. Further investigation of the TNBC liver metastasis signature predicted 16 hub genes, 6 biomarkers with clinically available drugs, and 22 survival genes. The sequential interrogation of CDX-CTCs is an innovative liquid biopsy-based approach for the discovery of organ metastasis-specific signatures of CTCs. This represents the first step for mechanistic and analytical validation in their application as prognostic indicators and therapeutic targets. Targeting CTC drug candidate biomarkers along with combination therapy can improve the clinical outcome of TNBC patients in general and recurrence of liver metastasis in particular.
Collapse
Affiliation(s)
- Monika Vishnoi
- Biomarker Research Program CenterHouston Methodist Research InstituteTXUSA
| | - Nikki Haowen Liu
- Biomarker Research Program CenterHouston Methodist Research InstituteTXUSA
| | - Wei Yin
- Biomarker Research Program CenterHouston Methodist Research InstituteTXUSA
| | - Debasish Boral
- Biomarker Research Program CenterHouston Methodist Research InstituteTXUSA
| | - Antonio Scamardo
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - David Hong
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Dario Marchetti
- Biomarker Research Program CenterHouston Methodist Research InstituteTXUSA
| |
Collapse
|
21
|
Kallergi G, Tsintari V, Sfakianakis S, Bei E, Lagoudaki E, Koutsopoulos A, Zacharopoulou N, Alkahtani S, Alarifi S, Stournaras C, Zervakis M, Georgoulias V. The prognostic value of JUNB-positive CTCs in metastatic breast cancer: from bioinformatics to phenotypic characterization. Breast Cancer Res 2019; 21:86. [PMID: 31370904 PMCID: PMC6676640 DOI: 10.1186/s13058-019-1166-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 07/01/2019] [Indexed: 12/19/2022] Open
Abstract
Background Circulating tumor cells (CTCs) are important for metastatic dissemination of cancer. They can provide useful information, regarding biological features and tumor heterogeneity; however, their detection and characterization are difficult due to their limited number in the bloodstream and their mesenchymal characteristics. Therefore, new biomarkers are needed to address these questions. Methods Bioinformatics functional enrichment analysis revealed a subgroup of 24 genes, potentially overexpressed in CTCs. Among these genes, the chemokine receptor CXCR4 plays a central role. After prioritization according to the CXCR4 corresponding pathways, five molecules (JUNB, YWHAB, TYROBP, NFYA, and PRDX1) were selected for further analysis in biological samples. The SKBR3, MDA-MB231, and MCF7 cell lines, as well as PBMCs from normal (n = 10) blood donors, were used as controls to define the expression pattern of all the examined molecules. Consequently, 100 previously untreated metastatic breast cancer (mBC) patients (n = 100) were analyzed using the following combinations of antibodies: CK (cytokeratin)/CXCR4/JUNB, CK/NFYA/ΥWHΑΒ (14-3-3), and CK/TYROBP/PRDX1. A threshold value for every molecule was considered the mean expression in normal PBMCs. Results Quantification of CXCR4 revealed overexpression of the receptor in SKBR3 and in CTCs, following the subsequent scale (SKBR3>CTCs>Hela>MCF7>MDA-MB231). JUNB was also overexpressed in CTCs (SKBR3>CTCs>MCF7>MDA-MB231>Hela). According to the defined threshold for each molecule, CXCR4-positive CTCs were identified in 90% of the patients with detectable tumor cells in their blood. In addition, 65%, 75%, 14.3%, and 12.5% of the patients harbored JUNB-, TYROBP-, NFYA-, and PRDX-positive CTCs, respectively. Conversely, none of the patients revealed YWHAB-positive CTCs. Interestingly, JUNB expression in CTCs was phenotypically and statistically enhanced compared to patients’ blood cells (p = 0.002) providing a possible new biomarker for CTCs. Furthermore, the detection of JUNB-positive CTCs in patients was associated with poorer PFS (p = 0.015) and OS (p = 0.002). Moreover, JUNB staining of 11 primary and 4 metastatic tumors from the same cohort of patients revealed a dramatic increase of JUNB expression in metastasis. Conclusions CXCR4, JUNB, and TYROBP were overexpressed in CTCs, but only the expression of JUNB was associated with poor prognosis, providing a new biomarker and a potential therapeutic target for the elimination of CTCs. Electronic supplementary material The online version of this article (10.1186/s13058-019-1166-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Galatea Kallergi
- Laboratory of Τumor Cell Βiology, Medical School, University of Crete, Heraklion, Greece. .,Department of Biochemistry, Medical School, University of Crete, Voutes, 70013, Heraklion, Crete, Greece. .,Hellenic Oncology Research Group (HORG), Athens, Greece.
| | - Vasileia Tsintari
- Department of Oncology, Hematology, Rheumatology, Immunology and Pulmology, University Hospital, Tübingen, Germany
| | - Stelios Sfakianakis
- Computational BioMedicine Laboratory, Institute of Computer Science, Foundation for Research and Technology, Heraklion, Greece
| | - Ekaterini Bei
- Digital Image and Signal Processing Laboratory, School of Electrical and Computer Engineering, Technical University of Crete, Chania, Greece
| | - Eleni Lagoudaki
- Department of Pathology, University General Hospital of Heraklion, Heraklion, Crete, Greece
| | | | - Nefeli Zacharopoulou
- Department of Biochemistry, Medical School, University of Crete, Voutes, 70013, Heraklion, Crete, Greece
| | - Saad Alkahtani
- Department of Biochemistry, Medical School, University of Crete, Voutes, 70013, Heraklion, Crete, Greece.,Department of Zoology, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Saud Alarifi
- Department of Biochemistry, Medical School, University of Crete, Voutes, 70013, Heraklion, Crete, Greece.,Department of Zoology, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Christos Stournaras
- Department of Biochemistry, Medical School, University of Crete, Voutes, 70013, Heraklion, Crete, Greece
| | - Michalis Zervakis
- Digital Image and Signal Processing Laboratory, School of Electrical and Computer Engineering, Technical University of Crete, Chania, Greece
| | - Vassilis Georgoulias
- Laboratory of Τumor Cell Βiology, Medical School, University of Crete, Heraklion, Greece.,Hellenic Oncology Research Group (HORG), Athens, Greece
| |
Collapse
|
22
|
Papadaki MA, Stoupis G, Theodoropoulos PA, Mavroudis D, Georgoulias V, Agelaki S. Circulating Tumor Cells with Stemness and Epithelial-to-Mesenchymal Transition Features Are Chemoresistant and Predictive of Poor Outcome in Metastatic Breast Cancer. Mol Cancer Ther 2018; 18:437-447. [PMID: 30401696 DOI: 10.1158/1535-7163.mct-18-0584] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/28/2018] [Accepted: 10/31/2018] [Indexed: 11/16/2022]
Abstract
Circulating tumor cells (CTCs) bearing phenotypes related to cancer stem cells (CSCs) and epithelial-to-mesenchymal transition (EMT) have been identified in breast cancer; however, their clinical significance is not clear. In the current study, we investigated the prognostic relevance of single CSC+/partial-EMT+ CTCs in patients with metastatic breast cancer and the effect of first-line chemotherapy on their incidence. For this purpose, triple immunofluorescence against cytokeratin, ALDH1, and TWIST1 was performed in peripheral blood mononuclear cell (PBMC) cytospins from 130 patients before and after first-line chemotherapy. CSC+/partial-EMT+ CTCs were characterized as cells co-expressing cytokeratin, high levels of ALDH1, and nuclear TWIST1. CSC+/partial-EMT+ CTCs were evident in 27.7% of patients at baseline and were correlated to lung metastases (P = 0.010) and decreased progression-free survival [PFS; median 10.2 (8.9-11.6) vs. 13.5 (11.3-15.7) months; P = 0.024]. Their detection was an independent factor predicting for increased risk of relapse [multivariate analysis; HR (95% confidence interval (CI)): 1.785 (1.171-2.720); P = 0.007]. In HER-2-negative patients, CSC+/partial-EMT+ CTCs were additionally associated with reduced overall survival (OS) [median 39 (26.2-51.9) vs. 51 (15.7-86.4) months; P = 0.020] and increased risk of death [multivariate analysis; HR (95% CI): 2.228 (1.066-4.655); P = 0.033]. Chemotherapy resulted in a significant increase in the incidence of CSC+/partial-EMT+ CTCs (mean CTC% per patient: 59.4% post vs. 39.5% pre; P = 0.018), which was subsequently confirmed only in HER2-negative patients (P = 0.040) and in non-responders at the end of treatment (P = 0.020). In conclusion, CSC+/partial-EMT+ CTCs represent a chemoresistant subpopulation, which independently predicts for unfavorable outcome in metastatic breast cancer. Efficient targeting of these CTCs could potentially increase patient survival.
Collapse
Affiliation(s)
- Maria A Papadaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Crete, Greece
| | - Giannis Stoupis
- Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece
| | | | - Dimitris Mavroudis
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Crete, Greece.,Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece
| | - Vassilis Georgoulias
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Crete, Greece.,Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece
| | - Sofia Agelaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Crete, Greece. .,Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece
| |
Collapse
|
23
|
Voutsadakis IA. HER2 in stemness and epithelial-mesenchymal plasticity of breast cancer. Clin Transl Oncol 2018; 21:539-555. [PMID: 30306401 DOI: 10.1007/s12094-018-1961-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 10/03/2018] [Indexed: 02/06/2023]
Abstract
Breast cancer had been the first non-hematologic malignancy where sub-types based on molecular characterization had entered clinical practice. HER2 over-expression, due to either gene amplification or protein up-regulation, defines one of these sub-types and is clinically exploited by addition of HER2-targeted treatments to the regimens of treatment. Nevertheless, in many occasions HER2-positive cancers are resistant or become refractory to these therapies. Several mechanisms, such as activation of alternative pathways or loss of expression of the receptor in cancer cells, have been proposed as the cause of these therapeutic failures. Cancer stem cells (CSCs, alternatively called tumor-initiating cells) comprise a small percentage of the tumor cells, but are capable of reconstituting and propagating tumors due to their superior intrinsic capacity for regeneration, survival and resistance to therapies. CSCs possess circuits enabling epigenetic plasticity which endow them with the ability to alternate between epithelial and mesenchymal states. This paper will discuss the expression and regulation of HER2 in CSCs of the different sub-types of breast cancer and relationships of the receptor with both the circuits of stemness and epithelial-mesenchymal plasticity. Therapeutic repercussions of the relationship of HER2-initiated signaling with stemness networks will also be proposed.
Collapse
Affiliation(s)
- I A Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, 750 Great Northern Road, Sault Ste. Marie, ON, P6B 0A8, Canada. .,Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada.
| |
Collapse
|
24
|
Kallergi G, Aggouraki D, Zacharopoulou N, Stournaras C, Georgoulias V, Martin SS. Evaluation of α-tubulin, detyrosinated α-tubulin, and vimentin in CTCs: identification of the interaction between CTCs and blood cells through cytoskeletal elements. Breast Cancer Res 2018; 20:67. [PMID: 29976237 PMCID: PMC6034292 DOI: 10.1186/s13058-018-0993-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 05/25/2018] [Indexed: 01/16/2023] Open
Abstract
Background Circulating tumor cells (CTCs) are the major players in the metastatic process. A potential mechanism of cell migration and invasion is the formation of microtentacles in tumor cells. These structures are supported by α-tubulin (TUB), detyrosinated α-tubulin (GLU), and vimentin (VIM). In the current study, we evaluated the expression of those cytoskeletal proteins in CTCs. Methods Forty patients with breast cancer (BC) (16 early and 24 metastatic) were enrolled in the study. CTCs were isolated using the ISET platform and stained with the following combinations of antibodies: pancytokeratin (CK)/VIM/TUB and CK/VIM/GLU. Samples were analyzed with the ARIOL platform and confocal laser scanning microscopy. Results Fluorescence quantification revealed that the ratios CK/TUB, CK/VIM, and CK/GLU were statistically increased in MCF7 compared with more aggressive cell lines (SKBR3 and MDA-MB-231). In addition, all of these ratios were statistically increased in MCF7 cells compared with metastatic BC patients’ CTCs (p = 0.0001, p = 0.0001, and p = 0.003, respectively). Interestingly, intercellular connections among CTCs and between CTCs and blood cells through cytoskeleton bridges were revealed, whereas microtentacles were increased in patients with CTC clusters. These intercellular connections were supported by TUB, VIM, and GLU. Quantification of the examined molecules revealed that the median intensity of TUB, GLU, and VIM was significantly increased in patients with metastatic BC compared with those with early disease (TUB, 62.27 vs 11.5, p = 0.0001; GLU, 6.99 vs 5.29, p = 0.029; and VIM, 8.24 vs 5.38, p = 0.0001, respectively). Conclusions CTCs from patients with BC aggregate to each other and to blood cells through cytoskeletal protrusions, supported by VIM, TUB, and GLU. Quantification of these molecules could potentially identify CTCs related to more aggressive disease. Electronic supplementary material The online version of this article (10.1186/s13058-018-0993-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- G Kallergi
- Laboratory of Τumor Cell Biology, School of Medicine, University of Crete, Heraklion, Greece. .,Department of Biochemistry, University of Crete, Greece Medical School, Heraklion, Greece.
| | - D Aggouraki
- Laboratory of Τumor Cell Biology, School of Medicine, University of Crete, Heraklion, Greece
| | - N Zacharopoulou
- Department of Biochemistry, University of Crete, Greece Medical School, Heraklion, Greece
| | - C Stournaras
- Department of Biochemistry, University of Crete, Greece Medical School, Heraklion, Greece
| | - V Georgoulias
- Laboratory of Τumor Cell Biology, School of Medicine, University of Crete, Heraklion, Greece
| | - S S Martin
- Department of Physiology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD, USA
| |
Collapse
|
25
|
Detection of HER2 Amplification in Circulating Tumor Cells of HER2-Negative Gastric Cancer Patients. Target Oncol 2018; 12:341-351. [PMID: 28508152 DOI: 10.1007/s11523-017-0493-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A key to the successful use of targeted cancer therapy is the ability to preselect patients who are likely to benefit from the treatment according to molecular markers. Assessment for predicting therapy response is mostly done using tumor biopsies. However, these might not truly represent all of the patient's malignant cells because of tumor heterogeneity and/or clonal evolution during disease progression. One potential strategy that can complement primary tumor biopsy is the analysis of circulating tumor cells (CTCs). In this study, we analyzed CTCs of patients with gastric cancer (GC) to find those who were likely to benefit from trastuzumab therapies. We developed an imaging-based method that enabled CTC identification simultaneously with evaluation of HER2 gene amplification (the 3D-IF-FISH method). Then we performed a study enrolling 101 GC patients in whom we analyzed CTCs by both 3D-IF-FISH and an FDA-approved CellSearch system. As compared with the CellSearch system, 3D-IF-FISH methods identified a higher number of patients whose primary tumors were HER2- but who had HER2+ CTCs, suggesting that the 3D-IF-FISH method is effective in preselecting patients for trastuzumab therapies. To demonstrate this, we performed an exploratory clinical study to evaluate the clinical benefits of trastuzumab treatment for advanced GC patients (n = 15) whose primary tumors were HER2-, but whose CTCs showed HER2 amplification. An interim evaluation after the first stage showed that these preselected patients had response rates comparable to those reported in the trastuzumab-plus-chemotherapy arm of the ToGA study. The present study offers a new, non-invasive strategy to select patients who are likely to benefit from trastuzumab-based therapies, despite their primary biopsy being HER2-negative. (UMIN ID: UMIN000008622).
Collapse
|
26
|
Abraham J, Singh S, Joshi S. Liquid biopsy - emergence of a new era in personalized cancer care. ACTA ACUST UNITED AC 2018. [DOI: 10.1186/s41241-018-0053-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
27
|
|