1
|
Baboudjian M, Peyrottes A, Dariane C, Fromont G, Denis JA, Fiard G, Kassab D, Ladoire S, Lehmann-Che J, Ploussard G, Rouprêt M, Barthélémy P, Roubaud G, Lamy PJ. Circulating Biomarkers Predictive of Treatment Response in Patients with Hormone-sensitive or Castration-resistant Metastatic Prostate Cancer: A Systematic Review. Eur Urol Oncol 2024; 7:1228-1245. [PMID: 38824003 DOI: 10.1016/j.euo.2024.05.003] [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: 03/08/2024] [Revised: 04/26/2024] [Accepted: 05/09/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND AND OBJECTIVE Metastatic prostate cancer (mPCa) harbors genomic alterations that may predict targeted therapy efficacy. These alterations can be identified not only in tissue but also directly in biologic fluids (ie, liquid biopsies), mainly blood. Liquid biopsies may represent a safer and less invasive alternative for monitoring patients treated for mPCa. Current research focuses on the description and validation of novel predictive biomarkers to improve precision medicine in mPCa. Our aim was to systematically review the current evidence on liquid biopsy biomarkers for predicting treatment response in mPCa. METHODS We systematically searched Medline, Web of Science, and evidence-based websites for publications on circulating biomarkers in mPCa between March 2013 and February 2024 for review. Endpoints were: prediction of overall survival, biochemical or radiographic progression-free survival after treatment (chemotherapy, androgen deprivation therapy, androgen receptor pathway inhibitors [ARPIs], immunotherapy, or PARP inhibitors [PARPIs]). For each biomarker, the level of evidence (LOE) for clinical validity was attributed: LOE IA and IB, high level of evidence; LOE IIB and IIC, intermediate level; and LOE IIIC and LOE IV-VD, weak level. KEY FINDINGS AND LIMITATIONS The predictive value of each biomarker for the response to several therapies was evaluated in both metastatic hormone-sensitive (mHSPC) and castration-resistant prostate cancer (mCRPC). In patients with mCRPC, BRCA1/2 or ATM mutations predicted response to ARPIs (LOE IB) and PARPIs (LOE IIB), while AR-V7 transcripts or AR-V7 protein levels in circulating tumor cells (CTCs) predicted response to ARPIs and taxanes (LOE IB). CTC quantification predicted response to cabazitaxel, abiraterone, and radium-223 (LOE IIB), while TP53 alterations predicted response to 177Lu prostate-specific membrane antigen radioligand treatment (LOE IIB). AR copy number in circulating tumor DNA before the first treatment line and before subsequent lines predicted response to docetaxel, cabazitaxel, and ARPIs (LOE IIB). In mHSPC, DNA damage in lymphocytes was predictive of the response to radium-223 (LOE IIB). CONCLUSIONS AND CLINICAL IMPLICATIONS BRCA1/2, ATM, and AR alterations detected in liquid biopsies may help clinicians in management of patients with mPCa. The other circulating biomarkers did not reach the LOE required for routine clinical use and should be validated in prospective independent studies. PATIENT SUMMARY We reviewed studies assessing the value of biomarkers in blood or urine for management of metastatic prostate cancer. The evidence indicates that some biomarkers could help in selecting patients eligible for specific treatments.
Collapse
Affiliation(s)
- Michael Baboudjian
- Department of Urology, North Academic Hospital, AP-HM, Marseille, France
| | - Arthur Peyrottes
- Service d'Urologie et de Transplantation Rénale, Hôpital Saint-Louis, AP-HP, Université de Paris, Paris, France
| | - Charles Dariane
- Department of Urology, European Hospital Georges-Pompidou, University Paris Cité, Paris, France; UMR-S1151, CNRS UMR-S8253 Institut Necker Enfants Malades, Paris, France
| | - Gaëlle Fromont
- INSERM UMR1069, Nutrition Croissance et Cancer, University of Tours, Tours, France; Department of Pathology, CHRU de Tours, Tours, France
| | - Jérôme Alexandre Denis
- INSERM UMR_S938, CRSA, Biologie et Thérapeutiques du Cancer, Saint-Antoine University Hospital, Sorbonne Université, Paris, France; Service de Biochimie Endocrinienne et Oncologique, Oncobiologie Cellulaire et Moléculaire, GH Pitié-Salpêtrière, AP-HP, Paris, France
| | - Gaëlle Fiard
- Department of Urology, CHU Grenoble Alpes, University of Grenoble Alpes CNRS, Grenoble INP, TIMC, Grenoble, France
| | | | - Sylvain Ladoire
- Department of Medical Oncology, Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center, Dijon, France; University of Burgundy-Franche Comté, Dijon, France; INSERM U1231, Dijon, France
| | - Jacqueline Lehmann-Che
- INSERM U976, Immunologie Humaine, Pathophysiologie, Immunothérapie, Université Paris Cité, Paris, France; UF Oncologie Moléculaire, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Guillaume Ploussard
- Department of Urology, La Croix du Sud Hospital, Quint-Fonsegrives, France; Department of Urology, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Morgan Rouprêt
- Department of Urology, University Hospital Pitié-Salpêtrière, Paris, France; Faculty of Medicine, Sorbonne University, Paris, France
| | - Philippe Barthélémy
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Guilhem Roubaud
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Pierre-Jean Lamy
- Biopathologie et Génétique des Cancers, Institut Médical d'Analyse Génomique, Imagenome, Inovie, Montpellier, France; Unité de Recherche Clinique, Clinique Beausoleil, Montpellier, France.
| |
Collapse
|
2
|
Wang Q, Tan L. Advances in the role of circulating tumor cell heterogeneity in metastatic small cell lung cancer. CANCER INNOVATION 2024; 3:e98. [PMID: 38946931 PMCID: PMC11212323 DOI: 10.1002/cai2.98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/03/2023] [Accepted: 09/11/2023] [Indexed: 07/02/2024]
Abstract
Small cell lung cancer (SCLC), a highly aggressive malignancy, is rapidly at an extensive stage once diagnosed and is one of the leading causes of death from malignancy. In the past decade, the treatment of SCLC has largely remained unchanged, and chemotherapy remains the cornerstone of SCLC treatment. The therapeutic value of adding immune checkpoint inhibitors to chemotherapy for SCLC is low, and only a few SCLC patients have shown a response to immune checkpoint inhibitors. Circulating tumor cells (CTCs) are tumor cells shed from solid tumor masses into the peripheral circulation and are key to tumor metastasis. Single-cell sequencing has revealed that the genetic profiles of individual CTCs are highly heterogeneous and contribute to the poor outcome and prognosis of SCLC patients. Theoretically, phenotypic analysis of CTCs may be able to predict the diagnostic significance of new potential targets for metastatic tumors. In this paper, we will discuss in depth the heterogeneity of CTCs in SCLC and the value of CTCs for the diagnosis and prognosis of SCLC and as relevant tumor markers in metastatic SCLC.
Collapse
Affiliation(s)
- Qunxia Wang
- Department of Laboratory Medicine, Jiangxi Province's Key Laboratory of Laboratory MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Li‐Ming Tan
- Department of Laboratory Medicine, Jiangxi Province's Key Laboratory of Laboratory MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| |
Collapse
|
3
|
Chu PY, Nguyen TNA, Wu AY, Huang PS, Huang KL, Liao CJ, Hsieh CH, Wu MH. The Utilization of Optically Induced Dielectrophoresis (ODEP)-Based Cell Manipulation in a Microfluidic System for the Purification and Sorting of Circulating Tumor Cells (CTCs) with Different Sizes. MICROMACHINES 2023; 14:2170. [PMID: 38138338 PMCID: PMC10745986 DOI: 10.3390/mi14122170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023]
Abstract
The analysis of circulating tumor cells (CTCs) at the molecular level holds great promise for several clinical applications. For this goal, the harvest of high-purity, size-sorted CTCs with different subtypes from a blood sample are important. For this purpose, a two-step CTC isolation protocol was proposed, by which the immunomagnetic beads-based cell separation was first utilized to remove the majority of blood cells. After that, an optically induced dielectrophoresis (ODEP) microfluidic system was developed to (1) purify the CTCs from the remaining magnetic microbeads-bound blood cells and to (2) sort and separate the CTCs with different sizes. In this study, the ODEP microfluidic system was designed and fabricated. Moreover, its optimum operation conditions and performance were explored. The results exhibited that the presented technique was able to purify and sort the cancer cells with two different sizes from a tested cell suspension in a high-purity (93.5% and 90.1% for the OECM 1 and HA22T cancer cells, respectively) manner. Overall, this study presented a technique for the purification and sorting of cancer cells with different sizes. Apart from this application, the technique is also useful for other applications in which the high-purity and label-free purification and sorting of cells with different sizes is required.
Collapse
Affiliation(s)
- Po-Yu Chu
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (P.-Y.C.); (T.N.A.N.); (A.-Y.W.); (P.-S.H.); (K.-L.H.)
| | - Thi Ngoc Anh Nguyen
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (P.-Y.C.); (T.N.A.N.); (A.-Y.W.); (P.-S.H.); (K.-L.H.)
| | - Ai-Yun Wu
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (P.-Y.C.); (T.N.A.N.); (A.-Y.W.); (P.-S.H.); (K.-L.H.)
| | - Po-Shuan Huang
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (P.-Y.C.); (T.N.A.N.); (A.-Y.W.); (P.-S.H.); (K.-L.H.)
| | - Kai-Lin Huang
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (P.-Y.C.); (T.N.A.N.); (A.-Y.W.); (P.-S.H.); (K.-L.H.)
| | - Chia-Jung Liao
- Department of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan;
| | - Chia-Hsun Hsieh
- Division of Hematology-Oncology, Department of Internal Medicine, New Taipei City Municipal Tucheng Hospital, New Taipei City 23652, Taiwan;
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33302, Taiwan
| | - Min-Hsien Wu
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (P.-Y.C.); (T.N.A.N.); (A.-Y.W.); (P.-S.H.); (K.-L.H.)
- Division of Hematology-Oncology, Department of Internal Medicine, New Taipei City Municipal Tucheng Hospital, New Taipei City 23652, Taiwan;
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33302, Taiwan
| |
Collapse
|
4
|
Tabor JK, Onoichenco A, Narayan V, Wernicke AG, D’Amico RS, Vojnic M. Brain metastasis screening in the molecular age. Neurooncol Adv 2023; 5:vdad080. [PMID: 37484759 PMCID: PMC10358433 DOI: 10.1093/noajnl/vdad080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023] Open
Abstract
The incidence of brain metastases (BM) amongst cancer patients has been increasing due to improvements in therapeutic options and an increase in overall survival. Molecular characterization of tumors has provided insights into the biology and oncogenic drivers of BM and molecular subtype-based screening. Though there are currently some screening and surveillance guidelines for BM, they remain limited. In this comprehensive review, we review and present epidemiological data on BM, their molecular characterization, and current screening guidelines. The molecular subtypes with the highest BM incidence are epithelial growth factor receptor-mutated non-small cell lung cancer (NSCLC), BRCA1, triple-negative (TN), and HER2+ breast cancers, and BRAF-mutated melanoma. Furthermore, BMs are more likely to present asymptomatically at diagnosis in oncogene-addicted NSCLC and BRAF-mutated melanoma. European screening standards recommend more frequent screening for oncogene-addicted NSCLC patients, and clinical trials are investigating screening for BM in hormone receptor+, HER2+, and TN breast cancers. However, more work is needed to determine optimal screening guidelines for other primary cancer molecular subtypes. With the advent of personalized medicine, molecular characterization of tumors has revolutionized the landscape of cancer treatment and prognostication. Incorporating molecular characterization into BM screening guidelines may allow physicians to better identify patients at high risk for BM development and improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Vinayak Narayan
- Department of Neurological Surgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| | - A Gabriella Wernicke
- Department of Radiation Medicine, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| | - Randy S D’Amico
- Department of Neurological Surgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| | - Morana Vojnic
- Corresponding Author: Morana Vojnic, MD, MBA, 210 East 64th Street, Floor 4, New York, NY 10065, USA ()
| |
Collapse
|
5
|
Patel DA, Blay J. Seeding metastases: The role and clinical utility of circulating tumour cells. Tumour Biol 2021; 43:285-306. [PMID: 34690152 DOI: 10.3233/tub-210001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Peripheral human blood is a readily-accessible source of patient material in which circulating tumour cells (CTCs) can be found. Their isolation and characterization holds the potential to provide prognostic value for various solid cancers. Enumeration of CTCs from blood is becoming a common practice in informing prognosis and may guide therapy decisions. It is further recognized that enumeration alone does not capture perspective on the heterogeneity of tumours and varying functional abilities of the CTCs to interact with the secondary microenvironment. Characterizing the isolated CTCs further, in particular assessing their functional abilities, can track molecular changes in the disease progress. As a step towards identifying a suite of functional features of CTCs that could aid in clinical decisions, developing a CTC isolation technique based on extracellular matrix (ECM) interactions may provide a more solid foundation for isolating the cells of interest. Techniques based on size, charge, density, and single biomarkers are not sufficient as they underutilize other characteristics of cancer cells. The ability of cancer cells to interact with ECM proteins presents an opportunity to utilize their full character in capturing, and also allows assessment of the features that reveal how cells might behave at secondary sites during metastasis. This article will review some common techniques and recent advances in CTC capture technologies. It will further explore the heterogeneity of the CTC population, challenges they experience in their metastatic journey, and the advantages of utilizing an ECM-based platform for CTC capture. Lastly, we will discuss how tailored ECM approaches may present an optimal platform to capture an influential heterogeneous population of CTCs.
Collapse
Affiliation(s)
- Deep A Patel
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Jonathan Blay
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.,Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
6
|
Chen Y, Chen X, Li M, Fan P, Wang B, Zhao S, Yu W, Zhang S, Tang Y, Gao T. A new analytical platform for potential point-of-care testing of circulating tumor cells. Biosens Bioelectron 2020; 171:112718. [PMID: 33059165 DOI: 10.1016/j.bios.2020.112718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 01/21/2023]
Abstract
It is of significance to detect circulating tumor cells (CTCs) in whole blood using transportable instruments at the point of care to assist evaluating chemotherapeutic efficacy and recurrence risk of cancer patients. However, the current widely used detection methods either require expensive and complex equipments, need complicated enrichment steps, or produce high rates of false positive and/or negative results. Aiming for solving the two critical challenges involved in instrumentation miniaturization and simplification of sample preparation for POCT of CTCs without sacrificing the detection sensitivity and accuracy, this work reports a custom-built, automatic, large field-of-view microscopic CTC cytometer and a novel enrichment strategy based on a synthesized peptide ligand discovered from One-Bead One-Compound library screening. The custom-built microscope has compact size, low weight and efficient cost while still maintaining a detection limit of as low as 5 target objects. The simplified sample preparation utilized a novel peptide LXW7 functionalized to magnetic beads and allows for rapid, highly selective and sensitive detection of CTCs. This analytical platform may fulfill the unmet need for possible point-of-care CTC counting, and provide a new option for early diagnosis of cancers and convenient evaluation of chemotherapeutic efficacy and cancer recurrence.
Collapse
Affiliation(s)
- Yangfei Chen
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Xuqi Chen
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Mengna Li
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Pengwei Fan
- Clinical Laboratory Center, Wuhan No. 7 Hospital, Wuhan, 430071, Hubei, China
| | - Bin Wang
- Clinical Laboratory Center, Wuhan No. 7 Hospital, Wuhan, 430071, Hubei, China
| | - Sheng Zhao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Shaohua Zhang
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Yuchen Tang
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Tingjuan Gao
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China.
| |
Collapse
|
7
|
Wei T, Zhu D, Yang Y, Yuan G, Xie H, Shen R. The application of nano-enrichment in CTC detection and the clinical significance of CTCs in non-small cell lung cancer (NSCLC) treatment. PLoS One 2019; 14:e0219129. [PMID: 31344053 PMCID: PMC6657845 DOI: 10.1371/journal.pone.0219129] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 06/17/2019] [Indexed: 12/20/2022] Open
Abstract
Circulating tumor cells (CTCs) are an independent prognostic marker in non-small cell lung cancer (NSCLC). CTC numbers are closely related to early diagnosis, clinical stage, therapy surveillance, and prognosis of NSCLC. We used a more efficient nano-enrichment method to detect CTCs in NSCLC patients and explored the clinical value of CTCs. The results showed that CTC numbers in stage IV cases were significantly higher than those in stage I, II or III cases. The number of CTCs in poorly-differentiated cases was significantly higher than that in well-differentiated cases. During six chemotherapy cycles, the average CTC number decreased from 5.8/7.5 ml in cycle #1 to 2.4/7.5 ml in cycle #4 and remained at almost the same level from 4 to 6 cycles. CTC numbers in patients with EGFR mutations was significantly higher than those in patients with no mutations. The average progression free survival (PFS) in the favorable group (CTC ≤ 5/7.5 ml) was 11.3 months, which was longer than that in the unfavorable group (CTC > 5/7.5 ml, 7.2 months). In conclusion, the assessment of NSCLC cannot be performed using a single CTC analysis. The clinical value is more significant in the continuous analysis of CTC data, as well as the cross-validation of other indexes and imaging results.
Collapse
Affiliation(s)
- Tengteng Wei
- Department of Thoracic Cardiovascular Surgery, Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Donglin Zhu
- Department of Thoracic Cardiovascular Surgery, Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yong Yang
- Department of Thoracic Cardiovascular Surgery, Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Guangda Yuan
- Department of Thoracic Cardiovascular Surgery, Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Hongya Xie
- Department of Thoracic Cardiovascular Surgery, Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Rongming Shen
- Department of Thoracic Cardiovascular Surgery, Affiliated Suzhou Municipal Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
- * E-mail:
| |
Collapse
|
8
|
Chistiakov DA, Chekhonin VP. Circulating tumor cells and their advances to promote cancer metastasis and relapse, with focus on glioblastoma multiforme. Exp Mol Pathol 2018; 105:166-174. [DOI: 10.1016/j.yexmp.2018.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/01/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022]
|
9
|
Neves M, Azevedo R, Lima L, Oliveira MI, Peixoto A, Ferreira D, Soares J, Fernandes E, Gaiteiro C, Palmeira C, Cotton S, Mereiter S, Campos D, Afonso LP, Ribeiro R, Fraga A, Tavares A, Mansinho H, Monteiro E, Videira PA, Freitas PP, Reis CA, Santos LL, Dieguez L, Ferreira JA. Exploring sialyl-Tn expression in microfluidic-isolated circulating tumour cells: A novel biomarker and an analytical tool for precision oncology applications. N Biotechnol 2018; 49:77-87. [PMID: 30273682 DOI: 10.1016/j.nbt.2018.09.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 09/14/2018] [Accepted: 09/25/2018] [Indexed: 12/17/2022]
Abstract
Circulating tumour cells (CTCs) originating from a primary tumour, lymph nodes and distant metastases hold great potential for liquid biopsies by providing a molecular fingerprint for disease dissemination and its temporal evolution through the course of disease management. CTC enumeration, classically defined on the basis of surface expression of Epithelial Cell Adhesion Molecule (EpCAM) and absence of the pan-leukocyte marker CD45, has been shown to correlate with clinical outcome. However, existing approaches introduce bias into the subsets of captured CTCs, which may exclude biologically and clinically relevant subpopulations. Here we explore the overexpression of the membrane protein O-glycan sialyl-Tn (STn) antigen in advanced bladder and colorectal tumours, but not in blood cells, to propose a novel CTC isolation technology. Using a size-based microfluidic device, we show that the majority (>90%) of CTCs isolated from the blood of patients with metastatic bladder and colorectal cancers express the STn antigen, supporting a link with metastasis. STn+ CTC counts were significantly higher than EpCAM-based detection in colorectal cancer, providing a more efficient cell-surface biomarker for CTC isolation. Exploring this concept, we constructed a glycan affinity-based microfluidic device for selective isolation of STn+ CTCs and propose an enzyme-based strategy for the recovery of viable cancer cells for downstream investigations. Finally, clinically relevant cancer biomarkers (transcripts and mutations) in bladder and colorectal tumours, were identified in cells isolated by microfluidics, confirming their malignant origin and highlighting the potential of this technology in the context of precision oncology.
Collapse
Affiliation(s)
- Manuel Neves
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; International Iberian Nanotechnology Laboratory (INL), Braga, Portugal
| | - Rita Azevedo
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| | - Luís Lima
- Portuguese Institute of Oncology, Porto, Portugal; Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal
| | - Marta I Oliveira
- International Iberian Nanotechnology Laboratory (INL), Braga, Portugal
| | - Andreia Peixoto
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal; INEB-Institute for Biomedical Engineering of Porto, Portugal
| | | | - Janine Soares
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| | - Elisabete Fernandes
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal; INEB-Institute for Biomedical Engineering of Porto, Portugal
| | - Cristiana Gaiteiro
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| | | | - Sofia Cotton
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| | - Stefan Mereiter
- Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal
| | - Diana Campos
- Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal
| | | | - Ricardo Ribeiro
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal; INEB-Institute for Biomedical Engineering of Porto, Portugal
| | - Avelino Fraga
- Hospital Centre- Hospital of Santo António of Porto, Portugal
| | - Ana Tavares
- Portuguese Institute of Oncology, Porto, Portugal
| | - Hélder Mansinho
- Hemato-Oncology Clinic, Hospital Garcia de Orta, EPE, Almada, Portugal; Gupo de Investigação do Cancro Digestivo-GICD, Portugal
| | | | - Paula A Videira
- Glycoimmunology Group, UCIBIO, Departamento Ciências da Vida, Faculdade de Ciência e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Paulo P Freitas
- International Iberian Nanotechnology Laboratory (INL), Braga, Portugal; INESC - Microsistemas e Nanotecnologias, Lisboa, Lisbon, Portugal
| | - Celso A Reis
- Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal; Faculty of Medicine, University of Porto, Portugal
| | - Lúcio Lara Santos
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; UFP: School of Health Sciences, Fernando Pessoa University of Porto, Portugal; Porto Comprehensive Cancer Center (P.ccc), Porto, Portugal
| | - Lorena Dieguez
- International Iberian Nanotechnology Laboratory (INL), Braga, Portugal
| | - José Alexandre Ferreira
- Portuguese Institute of Oncology, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal; International Iberian Nanotechnology Laboratory (INL), Braga, Portugal; Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Portugal.
| |
Collapse
|
10
|
TFF1 and TFF3 mRNAs Are Higher in Blood from Breast Cancer Patients with Metastatic Disease than Those without. JOURNAL OF ONCOLOGY 2018; 2018:4793498. [PMID: 29977293 PMCID: PMC6011051 DOI: 10.1155/2018/4793498] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 03/03/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022]
Abstract
Introduction Breast cancer metastasis occurs when tumor cells dissociate from the primary tumor and migrate to distant organs through the peripheral bloodstream or lymphatic drainage. Circulating tumor cells (CTCs) originate from primary sites or metastases and circulate in the patients' bloodstream. Molecular assays for the detection and molecular characterization of CTCs can serve as a liquid biopsy and can represent an alternative to invasive biopsies as a source of tumor tissue in the metastatic patients. Patients and Methods We analyzed the presence of CTCs in the peripheral blood of 50 breast cancer patients by quantitative real-time reverse transcriptase polymerase chain reaction (RT-qPCR) to detect trefoil factor family (TFF) 1 and 3 genes. Results We found significant difference in the level of both TFF1 and TFF3 mRNA in the blood of nonmetastatic versus metastatic breast cancer patients (p= 0.001 and p= 0.038, respectively). TFF1 mRNA was detected at higher levels in 34.6% of metastatic breast cancer patients as compared to 0% of nonmetastatic (p= 0.002). As regards TFF3 mRNA, it was detected at higher levels in 46.2% of metastatic breast cancer patients as compared to 4% of nonmetastatic (p= 0.026). Moreover, we found that the high level of both TFF1 and TFF3 mRNA was related to estrogen status of the patients. The detection of high level of TFF1 mRNA in CTCs was associated with bone metastases (77.8%), while that of TFF3 was related to lymph node involvement (75%) and lung metastases (68.8%). Conclusion The combined measurement of both TFF1 and TFF3 mRNA level for differentiation of metastatic from nonmetastatic breast cancer gave 57.69% sensitivity and 83.3% specificity.
Collapse
|
11
|
Yadav DK, Bai X, Yadav RK, Singh A, Li G, Ma T, Chen W, Liang T. Liquid biopsy in pancreatic cancer: the beginning of a new era. Oncotarget 2018; 9:26900-26933. [PMID: 29928492 PMCID: PMC6003564 DOI: 10.18632/oncotarget.24809] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/25/2018] [Indexed: 12/21/2022] Open
Abstract
With dismal survival rate pancreatic cancer remains one of the most aggressive and devastating malignancy. Predominantly, due to the absence of a dependable methodology for early identification and limited therapeutic options for advanced disease. However, it takes over 17 years to develop pancreatic cancer from initiation of mutation to metastatic cancer; therefore, if diagnosed early; it may increase overall survival dramatically, thus, providing a window of opportunity for early detection. Recently, genomic expression analysis defined 4 subtypes of pancreatic cancer based on mutated genes. Hence, we need simple and standard, minimally invasive test that can monitor those altered genes or their associated pathways in time for the success of precision medicine, and liquid biopsy seems to be one answer to all these questions. Again, liquid biopsy has an ability to pair with genomic tests. Additionally, liquid biopsy based development of circulating tumor cells derived xenografts, 3D organoids system, real-time monitoring of genetic mutations by circulating tumor DNA and exosome as the targeted drug delivery vehicle holds lots of potential for the treatment and cure of pancreatic cancer. At present, diagnosis of pancreatic cancer is frantically done on the premise of CA19-9 and radiological features only, which doesn't give a picture of genetic mutations and epigenetic alteration involved. In this manner, the current diagnostic paradigm for pancreatic cancer diagnosis experiences low diagnostic accuracy. This review article discusses the current state of liquid biopsy in pancreatic cancer as diagnostic and therapeutic tools and future perspectives of research in the light of circulating tumor cells, circulating tumor DNA and exosomes.
Collapse
Affiliation(s)
- Dipesh Kumar Yadav
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Rajesh Kumar Yadav
- Department of Pharmacology, Gandaki Medical College, Tribhuwan University, Institute of Medicine, Pokhara 33700, Nepal
| | - Alina Singh
- Department of Surgery, Bir Hospital, National Academy of Medical Science, Kanti Path, Kathmandu 44600, Nepal
| | - Guogang Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Tao Ma
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
12
|
Opoku-Damoah Y, Assanhou AG, Sooro MA, Baduweh CA, Sun C, Ding Y. Functional Diagnostic and Therapeutic Nanoconstructs for Efficient Probing of Circulating Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2018; 10:14231-14247. [PMID: 29557165 DOI: 10.1021/acsami.7b17896] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The circulation of tumor cells in peripheral blood is mostly recognized as a prerequisite for cancer progression or systemic invasion, and it correlates with the pivotal hallmark of malignancies known as metastasis. Multiple detection schemes for circulating tumor cells (CTCs) have emerged as the most discerning criteria for monitoring the outcome of anticancer therapy. Therefore, there has been a tremendous increase in the use of robust nanostructured platforms for observation of these mobile tumor cells through various simultaneous diagnosis and treatment regimens developed from conventional techniques. This review seeks to give detailed information about the nature of CTCs as well as techniques for exploiting specific biomarkers to help monitor cancer via detection, capturing, and analysis of unstable tumor cells. We will further discuss nanobased diagnostic interventions and novel platforms which have recently been developed from versatile nanomaterials such as polymer nanocomposites, metal organic frameworks, bioderived nanomaterials and other physically responsive particles with desirable intrinsic and external properties. Herein, we will also include in vivo nanotheranostic platforms which have received a lot of attention because of their enormous clinical potential. In all, this review sums up the general potential of key promising nanoinspired systems as well as other advanced strategies under research and those in clinical use.
Collapse
Affiliation(s)
- Yaw Opoku-Damoah
- Australian Institute for Bioengineering & Nanotechnology , The University of Queensland , St. Lucia , Brisbane, QLD 4072
| | - Assogba G Assanhou
- UFR Pharmacie, Falculté des Sciences de la Santé , Université d'Abomey-Calavi , 01BP188 Cotonou , Benin
| | | | | | | | | |
Collapse
|
13
|
Kitz J, Lowes LE, Goodale D, Allan AL. Circulating Tumor Cell Analysis in Preclinical Mouse Models of Metastasis. Diagnostics (Basel) 2018; 8:E30. [PMID: 29710776 PMCID: PMC6023422 DOI: 10.3390/diagnostics8020030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 01/24/2023] Open
Abstract
The majority of cancer deaths occur because of metastasis since current therapies are largely non-curative in the metastatic setting. The use of in vivo preclinical mouse models for assessing metastasis is, therefore, critical for developing effective new cancer biomarkers and therapies. Although a number of quantitative tools have been previously developed to study in vivo metastasis, the detection and quantification of rare metastatic events has remained challenging. This review will discuss the use of circulating tumor cell (CTC) analysis as an effective means of tracking and characterizing metastatic disease progression in preclinical mouse models of breast and prostate cancer and the resulting lessons learned about CTC and metastasis biology. We will also discuss how the use of clinically-relevant CTC technologies such as the CellSearch® and Parsortix™ platforms for preclinical CTC studies can serve to enhance the study of cancer biology, new biomarkers, and novel therapies from the bench to the bedside.
Collapse
Affiliation(s)
- Jenna Kitz
- London Regional Cancer Program, London Health Sciences Centre, Department of Anatomy & Cell Biology, Western University, London, ON N6A 5W9, Canada.
| | - Lori E Lowes
- Flow Cytometry and Special Hematology, London Health Sciences Centre, London, ON N6A 5W9, Canada.
| | - David Goodale
- London Regional Cancer Program, London Health Sciences Centre, London, ON N6A 5W9, Canada.
| | - Alison L Allan
- London Regional Cancer Program, London Health Sciences Centre, Departments of Anatomy & Cell Biology and Oncology, Lawson Health Research Institute, Western University, London, ON N6A 5W9, Canada.
| |
Collapse
|
14
|
Azevedo R, Soares J, Peixoto A, Cotton S, Lima L, Santos LL, Ferreira JA. Circulating tumor cells in bladder cancer: Emerging technologies and clinical implications foreseeing precision oncology. Urol Oncol 2018. [PMID: 29530466 DOI: 10.1016/j.urolonc.2018.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CONTEXT Circulating tumor cells (CTC) in peripheral blood of cancer patients provide an opportunity for real-time liquid biopsies capable of aiding early intervention, therapeutic decision, response to therapy, and prognostication. Nevertheless, the rare and potentially heterogeneous molecular nature of CTC has delayed the standardization of robust high-throughput capture/enrichment and characterization technologies. OBJECTIVE This review aims to systematize emerging solutions for CTC analysis in bladder cancer (BC), their opportunities and limitations, while providing key insights on specific technologic aspects that may ultimately guide molecular studies and clinical implementation. EVIDENCE ACQUISITION State-of-the-art screening for CTC technologies and clinical applications in BC was conducted in MEDLINE through PubMed. EVIDENCE SYNTHESIS From 200 records identified by the search query, 25 original studies and 1 meta-analysis met the full criteria for selection. A significant myriad of CTC technological platforms, including immunoaffinity, biophysical, and direct CTC detection by molecular methods have been presented. Despite their preliminary nature and irrespective of the applied technology, most studies concluded that CTC counts in peripheral blood correlated with metastasis. Associations with advanced tumor stage and grade and worst prognosis have been suggested. However, the unspecific nature, low sensitivity, and the lack of standardization of current methods still constitutes a major drawback. Moreover, few comprehensive molecular studies have been conducted on these poorly known class of malignant cells. CONCLUSION The current rationale supports the importance of moving the CTC field beyond proof of concept studies toward molecular-based solutions capable of improving disease management. The road has been paved for identification of highly specific CTC biomarkers and novel targeted approaches, foreseeing successful clinical applications.
Collapse
Affiliation(s)
- Rita Azevedo
- Experimental Pathology and Therapeutics Group, Research Centre, Portuguese Oncology Institute of Porto (IPO-Porto), R. Dr. António Bernardino de Almeida 62, 4200-162 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, R. Jorge de Viterbo Ferreira 228, 4050-013 Porto, Portugal
| | - Janine Soares
- Experimental Pathology and Therapeutics Group, Research Centre, Portuguese Oncology Institute of Porto (IPO-Porto), R. Dr. António Bernardino de Almeida 62, 4200-162 Porto, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Research Centre, Portuguese Oncology Institute of Porto (IPO-Porto), R. Dr. António Bernardino de Almeida 62, 4200-162 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, R. Jorge de Viterbo Ferreira 228, 4050-013 Porto, Portugal; Institute for Research and Innovation in Health (i3S), University of Porto, R. Alfredo Allen, 4200-135 Porto, Portugal
| | - Sofia Cotton
- Experimental Pathology and Therapeutics Group, Research Centre, Portuguese Oncology Institute of Porto (IPO-Porto), R. Dr. António Bernardino de Almeida 62, 4200-162 Porto, Portugal
| | - Luís Lima
- Experimental Pathology and Therapeutics Group, Research Centre, Portuguese Oncology Institute of Porto (IPO-Porto), R. Dr. António Bernardino de Almeida 62, 4200-162 Porto, Portugal; Institute for Research and Innovation in Health (i3S), University of Porto, R. Alfredo Allen, 4200-135 Porto, Portugal; Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), R. Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal; Porto Comprehensive Cancer Centre (P.ccc), R. Dr. António Bernardino de Almeida 62, 4200-162 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Centre, Portuguese Oncology Institute of Porto (IPO-Porto), R. Dr. António Bernardino de Almeida 62, 4200-162 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, R. Jorge de Viterbo Ferreira 228, 4050-013 Porto, Portugal; Health School of University Fernando Pessoa, Praça de 9 de Abril 349, 4249-004 Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology (IPO-Porto), R. Dr. António Bernardino de Almeida 62, 4200-162 Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Research Centre, Portuguese Oncology Institute of Porto (IPO-Porto), R. Dr. António Bernardino de Almeida 62, 4200-162 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, R. Jorge de Viterbo Ferreira 228, 4050-013 Porto, Portugal; Institute for Research and Innovation in Health (i3S), University of Porto, R. Alfredo Allen, 4200-135 Porto, Portugal; Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), R. Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal; Porto Comprehensive Cancer Centre (P.ccc), R. Dr. António Bernardino de Almeida 62, 4200-162 Porto, Portugal; International Iberian Nanotechnology Laboratory (INL), Avda. Mestre José Veiga, 4715 Braga, Portugal.
| |
Collapse
|
15
|
Lowes LE, Goodale D, Xia Y, Postenka C, Piaseczny MM, Paczkowski F, Allan AL. Epithelial-to-mesenchymal transition leads to disease-stage differences in circulating tumor cell detection and metastasis in pre-clinical models of prostate cancer. Oncotarget 2018; 7:76125-76139. [PMID: 27764810 PMCID: PMC5342801 DOI: 10.18632/oncotarget.12682] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/29/2016] [Indexed: 12/20/2022] Open
Abstract
Metastasis is the cause of most prostate cancer (PCa) deaths and has been associated with circulating tumor cells (CTCs). The presence of ≥5 CTCs/7.5mL of blood is a poor prognosis indicator in metastatic PCa when assessed by the CellSearch® system, the “gold standard” clinical platform. However, ~35% of metastatic PCa patients assessed by CellSearch® have undetectable CTCs. We hypothesize that this is due to epithelial-to-mesenchymal transition (EMT) and subsequent loss of necessary CTC detection markers, with important implications for PCa metastasis. Two pre-clinical assays were developed to assess human CTCs in xenograft models; one comparable to CellSearch® (EpCAM-based) and one detecting CTCs semi-independent of EMT status via combined staining with EpCAM/HLA (human leukocyte antigen). In vivo differences in CTC generation, kinetics, metastasis and EMT status were determined using 4 PCa models with progressive epithelial (LNCaP, LNCaP-C42B) to mesenchymal (PC-3, PC-3M) phenotypes. Assay validation demonstrated that the CellSearch®-based assay failed to detect a significant number (~40-50%) of mesenchymal CTCs. In vivo, PCa with an increasingly mesenchymal phenotype shed greater numbers of CTCs more quickly and with greater metastatic capacity than PCa with an epithelial phenotype. Notably, the CellSearch®-based assay captured the majority of CTCs shed during early-stage disease in vivo, and only after establishment of metastases were a significant number of undetectable CTCs present. This study provides important insight into the influence of EMT on CTC generation and subsequent metastasis, and highlights that novel technologies aimed at capturing mesenchymal CTCs may only be useful in the setting of advanced metastatic disease.
Collapse
Affiliation(s)
- Lori E Lowes
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London ON, Canada
| | - David Goodale
- London Regional Cancer Program, London Health Sciences Centre, London ON, Canada
| | - Ying Xia
- London Regional Cancer Program, London Health Sciences Centre, London ON, Canada
| | - Carl Postenka
- London Regional Cancer Program, London Health Sciences Centre, London ON, Canada
| | - Matthew M Piaseczny
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London ON, Canada
| | - Freeman Paczkowski
- London Regional Cancer Program, London Health Sciences Centre, London ON, Canada
| | - Alison L Allan
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London ON, Canada.,Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London ON, Canada.,London Regional Cancer Program, London Health Sciences Centre, London ON, Canada.,Lawson Health Research Institute, London ON, Canada
| |
Collapse
|
16
|
Bredemeier M, Edimiris P, Tewes M, Mach P, Aktas B, Schellbach D, Wagner J, Kimmig R, Kasimir-Bauer S. Establishment of a multimarker qPCR panel for the molecular characterization of circulating tumor cells in blood samples of metastatic breast cancer patients during the course of palliative treatment. Oncotarget 2018; 7:41677-41690. [PMID: 27223437 PMCID: PMC5173087 DOI: 10.18632/oncotarget.9528] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 04/16/2016] [Indexed: 12/16/2022] Open
Abstract
Background Circulating tumor cells (CTC) are discussed to be an ideal surrogate marker for individualized treatment in metastatic breast cancer (MBC) since metastatic tissue is often difficult to obtain for repeated analysis. We established a nine gene qPCR panel to characterize the heterogeneous CTC population in MBC patients including epithelial CTC, their receptors (EPCAM, ERBB2, ERBB3, EGFR) CTC in Epithelial-Mesenchymal-Transition [(EMT); PIK3CA, AKT2), stem cell-like CTC (ALDH1) as well as resistant CTC (ERCC1, AURKA] to identify individual therapeutic targets. Results At TP0, at least one marker was detected in 84%, at TP1 in 74% and at TP2 in 79% of the patients, respectively. The expression of ERBB2, ERBB3 and ERCC1 alone or in combination with AURKA was significantly associated with therapy failure. ERBB2 + CTC were only detected in patients not receiving ERBB2 targeted therapies which correlated with no response. Furthermore, patients responding at TP2 had a significantly prolonged overall-survival than patients never responding (p = 0.0090). Patients and Methods 2 × 5 ml blood of 62 MBC patients was collected at the time of disease progression (TP0) and at two clinical staging time points (TP1 and TP2) after 8–12 weeks of chemo-, hormone or antibody therapy for the detection of CTC (AdnaTest EMT-2/StemCell Select™, QIAGEN Hannover GmbH, Germany). After pre-amplification, multiplex qPCR was performed. Establishment was performed using various cancer cell lines. PTPRC (Protein tyrosine phosphatase receptor type C) and GAPDH served as controls. Conclusions Monitoring MBC patients using a multimarker qPCR panel for the characterization of CTC might help to treat patients accordingly in the future.
Collapse
Affiliation(s)
- Maren Bredemeier
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Philippos Edimiris
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mitra Tewes
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Pawel Mach
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bahriye Aktas
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | | | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
17
|
Messaritakis I, Kotsakis A, Georgoulias V. Association of epithelial-to-mesenchymal transition circulating tumor cells in non-small cell lung cancer (NSCLC) molecular subgroups. J Thorac Dis 2018; 9:4817-4820. [PMID: 29312665 DOI: 10.21037/jtd.2017.11.106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ippokratis Messaritakis
- Laboratory of Tumor Cell Biology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Athanasios Kotsakis
- Laboratory of Tumor Cell Biology, Medical School, University of Crete, Heraklion, Crete, Greece.,Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece
| | - Vassilis Georgoulias
- Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece
| |
Collapse
|
18
|
Benini S, Gamberi G, Cocchi S, Garbetta J, Alberti L, Righi A, Gambarotti M, Picci P, Ferrari S. Detection of circulating tumor cells in liquid biopsy from Ewing sarcoma patients. Cancer Manag Res 2018; 10:49-60. [PMID: 29386915 PMCID: PMC5765973 DOI: 10.2147/cmar.s141623] [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] [Indexed: 02/05/2023] Open
Abstract
Background Circulating tumor cells (CTCs) analysis is a promising new diagnostic field to estimate risk and monitor treatment efficacy, metastatic relapse, and progression in cancer patients. The study aim was to isolate and characterize CTCs in blood samples of Ewing sarcoma (ES) patients exploiting two main characteristics: CD99 expression and presence of chromosomal translocations. Materials and methods The method isolated CTCs from peripheral blood (PB) of ES patients. Cell-surface CD99 was a useful marker for CTCs determined using immunomagnetic separation with microbeads and CD99 monoclonal antibody. We tested sensitivity and specificity by detecting CTCs in blood collected from healthy donors and randomly during therapy from 18 ES patients. Evidence of CTCs was confirmed by detection of specific molecular markers using quantitative and digital reverse transcription-polymerase chain reaction targeting EWSR1/FLI1 type 1 and type 2 or EWSR1/ETS-related gene transcripts type 1 and type 9e. Results Feasibility of finding CTCs in PB of ES patients by immunoseparation with CD99 antibody and magnetic microbeads was demonstrated for the first time. At molecular analysis, three PB specimens tested positive for chimeric EWSR1/FLI1 type 2 and one PB for chimeric EWSR1/FLI1 type 2. CTCs detection was found above a limit of detection of 1 cell/mL of PB. Conclusion CTCs in PB of ES patients can be identified by this method and in ES CTCs analysis can be used as a liquid biopsy approach for prognostic and predictive purposes. The potential clinical implications of CTCs in PB samples detected by the platform for CTC isolation with molecular confirmation during therapy require further evaluation.
Collapse
Affiliation(s)
| | - Gabriella Gamberi
- Department of Pathology, Rizzoli Institute, Bologna.,Department of Biomedical and Neuromotor Science, University of Bologna, Bologna, Italy
| | | | | | | | | | | | - Piero Picci
- Department of Pathology, Rizzoli Institute, Bologna
| | - Stefano Ferrari
- Department of Chemotherapy, Rizzoli Institute, Bologna, Italy
| |
Collapse
|
19
|
Abstract
The majority of cancer-related deaths result from metastasis, the process by which cancer cells escape the primary tumor site and enter into the blood circulation in order to disseminate to secondary locations throughout the body. Tumor cells found within the circulation are referred to as circulating tumor cells (CTCs), and their detection and enumeration correlate with poor prognosis. The epithelial-to-mesenchymal transition (EMT) is a dynamic process that imparts epithelial cells with mesenchymal-like properties, thus facilitating tumor cell dissemination and contributing to metastasis. However, EMT also results in the downregulation of various epithelial proteins typically utilized by CTC technologies for enrichment and detection of these rare cells, resulting in reduced detection of some CTCs, potentially those with a more metastatic phenotype. In addition to the current clinical role of CTCs as a prognostic biomarker, they also have potential as a predictive biomarker via CTC characterization. However, CTC characterization is complicated by the unknown biological significance of CTCs possessing an EMT-like phenotype, and the ability to capture and understand this CTC subpopulation is an essential step in the utilization of CTCs for patient management. This chapter will review the process of EMT and its contribution to metastasis; discusses current and future clinical applications of CTCs; and describes both traditional and novel methods for CTC enrichment, detection, and characterization with a specific focus on CTCs with an EMT phenotype.
Collapse
|
20
|
Tartarone A, Lerose R, Rodriquenz MG, Mambella G, Calderoni G, Bozza G, Aieta M. Molecular characterization and prognostic significance of circulating tumor cells in patients with non-small cell lung cancer. J Thorac Dis 2017; 9:S1359-S1363. [PMID: 29184674 DOI: 10.21037/jtd.2017.07.80] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Circulating tumor cells (CTCs) are rare epithelial cells that can be found in the peripheral blood of cancer patients. A growing body of evidence indicates that CTCs may play a role in non-small cell lung cancer (NSCLC) for diagnosis, therapy monitoring and prognostic purposes. CTCs evaluation could be particularly relevant in this clinical setting, considering that physicians often have difficulty in obtaining an adequate tumor tissue and that patients are not always suitable to receive a re-biopsy. In the current review, we will focus on the molecular characterization and prognostic significance of CTCs in NSCLC patients.
Collapse
Affiliation(s)
- Alfredo Tartarone
- Department of Onco-Hematology, Division of Medical Oncology, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Rosa Lerose
- Hospital Pharmacy, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Maria Grazia Rodriquenz
- Department of Onco-Hematology, Division of Medical Oncology, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Giuseppina Mambella
- Department of Onco-Hematology, Division of Medical Oncology, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Giuseppe Calderoni
- Department of Onco-Hematology, Division of Medical Oncology, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Giovanni Bozza
- Department of Onco-Hematology, Division of Medical Oncology, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Michele Aieta
- Department of Onco-Hematology, Division of Medical Oncology, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| |
Collapse
|
21
|
Shields Iv CW, Wang JL, Ohiri KA, Essoyan ED, Yellen BB, Armstrong AJ, López GP. Magnetic separation of acoustically focused cancer cells from blood for magnetographic templating and analysis. LAB ON A CHIP 2016; 16:3833-3844. [PMID: 27713979 DOI: 10.1039/c6lc00719h] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Liquid biopsies hold enormous promise for the next generation of medical diagnoses. At the forefront of this effort, many are seeking to capture, enumerate and analyze circulating tumor cells (CTCs) as a means to prognosticate and develop individualized treatments for cancer. Capturing these rare cells, however, represents a major engineering challenge due to their low abundance, morphology and heterogeneity. A variety of microfluidic tools have been developed to isolate CTCs from drawn blood samples; however, few of these approaches offer a means to separate and analyze cells in an integrated system. We have developed a microfluidic platform comprised of three modules that offers high throughput separation of cancer cells from blood and on-chip organization of those cells for streamlined analyses. The first module uses an acoustic standing wave to rapidly align cells in a contact-free manner. The second module then separates magnetically labeled cells from unlabeled cells, offering purities exceeding 85% for cells and 90% for binary mixtures of synthetic particles. Finally, the third module contains a spatially periodic array of microwells with underlying micromagnets to capture individual cells for on-chip analyses (e.g., staining, imaging and quantification). This array is capable of capturing with accuracies exceeding 80% for magnetically labeled cells and 95% for magnetic particles. Overall, by virtue of its holistic processing of complex biological samples, this system has promise for the isolation and evaluation of rare cancer cells and can be readily extended to address a variety of applications across single cell biology and immunology.
Collapse
Affiliation(s)
- C Wyatt Shields Iv
- NSF Research Triangle Materials Research Science and Engineering Center, Duke University, Durham, NC 27708, USA and Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Jeffrey L Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Korine A Ohiri
- NSF Research Triangle Materials Research Science and Engineering Center, Duke University, Durham, NC 27708, USA and Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Eric D Essoyan
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Benjamin B Yellen
- NSF Research Triangle Materials Research Science and Engineering Center, Duke University, Durham, NC 27708, USA and Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA and Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | | | - Gabriel P López
- NSF Research Triangle Materials Research Science and Engineering Center, Duke University, Durham, NC 27708, USA and Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA and Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA and Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|
22
|
Bole AL, Manesiotis P. Advanced Materials for the Recognition and Capture of Whole Cells and Microorganisms. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5349-5366. [PMID: 26662854 DOI: 10.1002/adma.201503962] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/23/2015] [Indexed: 06/05/2023]
Abstract
Selective cell recognition and capture has recently attracted significant interest due to its potential importance for clinical, diagnostic, environmental, and security applications. Current methods for cell isolation from complex samples are largely dependent on cell size and density, with limited application scope as many of the target cells do not exhibit appreciable differences in this respect. The most recent and forthcoming developments in the area of selective recognition and capture of whole cells, based on natural receptors, as well as synthetic materials utilising physical and chemical properties of the target cell or microorganism, are highlighted. Particular focus is given to the development of cell complementary surfaces using the cells themselves as templating agents, by means of molecular imprinting, and their combination with sensing platforms for rapid cell detection in complex media. The benefits and challenges of each approach are discussed and a perspective of the future of this research area is given.
Collapse
Affiliation(s)
- Amanda L Bole
- School of Chemistry and Chemical Engineering, Queen's University Belfast, David Keir Building, Stranmillis Road, Belfast, BT9 5AG, N. Ireland, UK
| | - Panagiotis Manesiotis
- School of Chemistry and Chemical Engineering, Queen's University Belfast, David Keir Building, Stranmillis Road, Belfast, BT9 5AG, N. Ireland, UK
| |
Collapse
|
23
|
Forte VA, Barrak DK, Elhodaky M, Tung L, Snow A, Lang JE. The potential for liquid biopsies in the precision medical treatment of breast cancer. Cancer Biol Med 2016; 13:19-40. [PMID: 27144060 PMCID: PMC4850125 DOI: 10.28092/j.issn.2095-3941.2016.0007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Currently the clinical management of breast cancer relies on relatively few prognostic/predictive clinical markers (estrogen receptor, progesterone receptor, HER2), based on primary tumor biology. Circulating biomarkers, such as circulating tumor DNA (ctDNA) or circulating tumor cells (CTCs) may enhance our treatment options by focusing on the very cells that are the direct precursors of distant metastatic disease, and probably inherently different than the primary tumor's biology. To shift the current clinical paradigm, assessing tumor biology in real time by molecularly profiling CTCs or ctDNA may serve to discover therapeutic targets, detect minimal residual disease and predict response to treatment. This review serves to elucidate the detection, characterization, and clinical application of CTCs and ctDNA with the goal of precision treatment of breast cancer.
Collapse
Affiliation(s)
- Victoria A Forte
- Department of Medicine, Division of Medical Oncology, University of Southern California (USC), Los Angeles, CA 90033, USA; USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Dany K Barrak
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Surgery, Division of Breast, Endocrine and Soft Tissue Surgery, USC, Los Angeles, CA 90033, USA
| | - Mostafa Elhodaky
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Stem Cell and Regenerative Medicine, USC, Los Angeles, CA 90033, USA
| | - Lily Tung
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Surgery, Division of Breast, Endocrine and Soft Tissue Surgery, USC, Los Angeles, CA 90033, USA
| | - Anson Snow
- Department of Medicine, Division of Medical Oncology, University of Southern California (USC), Los Angeles, CA 90033, USA; USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Julie E Lang
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Surgery, Division of Breast, Endocrine and Soft Tissue Surgery, USC, Los Angeles, CA 90033, USA
| |
Collapse
|
24
|
Smolkova B, Mego M, Horvathova Kajabova V, Cierna Z, Danihel L, Sedlackova T, Minarik G, Zmetakova I, Krivulcik T, Gronesova P, Karaba M, Benca J, Pindak D, Mardiak J, Reuben JM, Fridrichova I. Expression of SOCS1 and CXCL12 Proteins in Primary Breast Cancer Are Associated with Presence of Circulating Tumor Cells in Peripheral Blood. Transl Oncol 2016; 9:184-90. [PMID: 27267835 PMCID: PMC4856862 DOI: 10.1016/j.tranon.2016.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/16/2016] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cells (CTCs) are independent prognostic factors in the primary and metastatic breast cancer patients and play crucial role in hematogenous tumor dissemination. The aim of this study was to correlate the presence of CTCs in peripheral blood with the expression of proteins in tumor tissue that have a putative role in regulation of cell growth and metastatic potential. This prospective study included 203 primary breast cancer patients treated by definitive surgery. CTCs were detected by quantitative real-time PCR for the expression of epithelial (CK19) or epithelial-to-mesenchymal transition–inducing transcription factor genes (TWIST1, SNAIL1, SLUG, and ZEB1). Expression of APC, ADAM23, CXCL12, E-cadherin, RASSF1, SYK, TIMP3, BRMS1, and SOCS1 proteins in primary breast tumor tissue was evaluated by immunohistochemistry. CTCs with epithelial markers were found in 17 (9.2%) patients. Their occurrence was associated with inhibition of SOCS1 expression (odds ratio [OR] = 0.07; 95% confidence interval [CI], 0.03-0.13; P < .001). CTCs with positive epithelial-to-mesenchymal transition markers were detected in 30 (15.8%) patients; however, no association with analyzed protein expressions was found. Overall, CTCs were detected in 44 (22.9%) patients. Presence of any CTC marker was significantly associated with positive CXCL12 expression (OR = 3.08; 95% CI, 1.15-8.26; P = .025) and lack of SOCS1 expression (OR = 0.10; 95% CI, 0.04-0.25; P < .001) in patient’s tumor tissues. As both CXCL12 and SOCS1 proteins are involved in cytokine signaling, our results provide support for the hypothesis that aberrant signaling cross talk between cytokine and chemokine responses could have an important role in hematogenous dissemination of tumor cells in breast cancer.
Collapse
Affiliation(s)
- Bozena Smolkova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia.
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, National Cancer Institute, Klenova 1, Bratislava, Slovakia.
| | - Viera Horvathova Kajabova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia.
| | - Zuzana Cierna
- Institute of Pathological Anatomy, Faculty of Medicine, Comenius University, University Hospital, Sasinkova 4, Bratislava, Slovakia.
| | - Ludovit Danihel
- Institute of Pathological Anatomy, Faculty of Medicine, Comenius University, University Hospital, Sasinkova 4, Bratislava, Slovakia; Pathological-Anatomical Workplace, Health Care Surveillance Authority, Sasinkova 4, Bratislava, Slovakia.
| | - Tatiana Sedlackova
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | - Gabriel Minarik
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | - Iveta Zmetakova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia.
| | - Tomas Krivulcik
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia.
| | - Paulina Gronesova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia.
| | - Marian Karaba
- Department of Surgical Oncology, National Cancer Institute, Klenova 1, Bratislava, Slovakia.
| | - Juraj Benca
- Department of Surgical Oncology, National Cancer Institute, Klenova 1, Bratislava, Slovakia.
| | - Daniel Pindak
- Department of Surgical Oncology, National Cancer Institute, Klenova 1, Bratislava, Slovakia.
| | - Jozef Mardiak
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, National Cancer Institute, Klenova 1, Bratislava, Slovakia.
| | - James M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ivana Fridrichova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia.
| |
Collapse
|
25
|
Caceres G, Puskas JA, Magliocco AM. Circulating Tumor Cells: A Window Into Tumor Development and Therapeutic Effectiveness. Cancer Control 2016; 22:167-76. [PMID: 26068761 DOI: 10.1177/107327481502200207] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are an important diagnostic tool for understanding the metastatic process and the development of cancer. METHODS This review covers the background, relevance, and potential limitations of CTCs as a measurement of cancer progression and how information derived from CTCs may affect treatment efficacy. It also highlights the difficulties of characterizing these rare cells due to the limited cell surface molecules unique to CTCs and each particular type of cancer. RESULTS The analysis of cancer in real time, through the measure of the number of CTCs in a " liquid" biopsy specimen, gives us the ability to monitor the therapeutic efficacy of treatments and possibly the metastatic potential of a tumor. CONCLUSIONS Through novel and innovative techniques yielding encouraging results, including microfluidic techniques, isolating and molecularly analyzing CTCs are becoming a reality. CTCs hold promise for understanding how tumors work and potentially aiding in their demise.
Collapse
Affiliation(s)
- Gisela Caceres
- Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | | | | |
Collapse
|
26
|
ANDERGASSEN ULRICH, KÖLBL ALEXANDRAC, MAHNER SVEN, JESCHKE UDO. Real-time RT-PCR systems for CTC detection from blood samples of breast cancer and gynaecological tumour patients (Review). Oncol Rep 2016; 35:1905-15. [DOI: 10.3892/or.2016.4608] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/15/2015] [Indexed: 11/06/2022] Open
|
27
|
Rossi E, Facchinetti A, Zamarchi R. Notes for developing a molecular test for the full characterization of circulating tumor cells. Chin J Cancer Res 2015; 27:471-8. [PMID: 26543333 DOI: 10.3978/j.issn.1000-9604.2015.09.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The proved association between the circulating tumor cell (CTC) levels and the patients' survival parameters has been growing interest to investigate the molecular profile of these neoplastic cells among which hide out precursors capable of initiating a new distant metastatic lesion. The full characterization of the tumor cells in peripheral blood of cancer patients is expected to be of help for understanding and (prospectively) for counteracting the metastatic process. The major hitch that is hampering the successful gaining of this result is the lack of a consensus onto standard operating procedures (SOPs) for performing what we generally define as the "liquid biopsy". Here we review the more recent acquisitions in the analysis of CTCs and tumor related nucleic acids, looking to the main open questions that are hampering their definitive employ in the routine clinical practice.
Collapse
Affiliation(s)
- Elisabetta Rossi
- 1 Department of Surgery, Oncology and Gastroenterology, Oncology Section, University of Padova, Padova, Italy ; 2 IOV-IRCCS, Padova, Italy
| | - Antonella Facchinetti
- 1 Department of Surgery, Oncology and Gastroenterology, Oncology Section, University of Padova, Padova, Italy ; 2 IOV-IRCCS, Padova, Italy
| | - Rita Zamarchi
- 1 Department of Surgery, Oncology and Gastroenterology, Oncology Section, University of Padova, Padova, Italy ; 2 IOV-IRCCS, Padova, Italy
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Yang KR, Mooney SM, Zarif JC, Coffey DS, Taichman RS, Pienta KJ. Niche inheritance: a cooperative pathway to enhance cancer cell fitness through ecosystem engineering. J Cell Biochem 2015; 115:1478-85. [PMID: 24700698 PMCID: PMC4143896 DOI: 10.1002/jcb.24813] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 04/01/2014] [Indexed: 02/03/2023]
Abstract
Cancer cells can be described as an invasive species that is able to establish itself in a new environment. The concept of niche construction can be utilized to describe the process by which cancer cells terraform their environment, thereby engineering an ecosystem that promotes the genetic fitness of the species. Ecological dispersion theory can then be utilized to describe and model the steps and barriers involved in a successful diaspora as the cancer cells leave the original host organ and migrate to new host organs to successfully establish a new metastatic community. These ecological concepts can be further utilized to define new diagnostic and therapeutic areas for lethal cancers. 115: 1478–1485, 2014. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kimberline R Yang
- Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | | | | | | | | |
Collapse
|
31
|
Rodriguez-Torres M, Allan AL. Aldehyde dehydrogenase as a marker and functional mediator of metastasis in solid tumors. Clin Exp Metastasis 2015; 33:97-113. [PMID: 26445849 PMCID: PMC4740561 DOI: 10.1007/s10585-015-9755-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 10/01/2015] [Indexed: 12/16/2022]
Abstract
There is accumulating evidence indicating that aldehyde dehydrogenase (ALDH) activity selects for cancer cells with increased aggressiveness, capacity for sustained proliferation, and plasticity in primary tumors. However, emerging data also suggests an important mechanistic role for the ALDH family of isoenzymes in the metastatic activity of tumor cells. Recent studies indicate that ALDH correlates with either increased or decreased metastatic capacity in a cellular context-dependent manner. Importantly, it appears that different ALDH isoforms support increased metastatic capacity in different tumor types. This review assesses the potential of ALDH as biological marker and mechanistic mediator of metastasis in solid tumors. In many malignancies, most notably in breast cancer, ALDH activity and expression appears to be a promising marker and potential therapeutic target for treating metastasis in the clinical setting.
Collapse
Affiliation(s)
- Mauricio Rodriguez-Torres
- London Regional Cancer Program, London Health Sciences Centre, London, ON, Canada.,Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Alison L Allan
- London Regional Cancer Program, London Health Sciences Centre, London, ON, Canada. .,Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Lawson Health Research Institute, London, ON, Canada. .,London Regional Cancer Program, Room A4-132, 790 Commissioners Road East, London, ON, N6A 4L6, Canada.
| |
Collapse
|
32
|
Adamczyk LA, Williams H, Frankow A, Ellis HP, Haynes HR, Perks C, Holly JMP, Kurian KM. Current Understanding of Circulating Tumor Cells - Potential Value in Malignancies of the Central Nervous System. Front Neurol 2015; 6:174. [PMID: 26322014 PMCID: PMC4530310 DOI: 10.3389/fneur.2015.00174] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/24/2015] [Indexed: 12/23/2022] Open
Abstract
Detection of circulating tumor cells (CTCs) in the blood via so-called "liquid biopsies" carries enormous clinical potential in malignancies of the central nervous system (CNS) because of the potential to follow disease evolution with a blood test, without the need for repeat neurosurgical procedures with their inherent risk of patient morbidity. To date, studies in non-CNS malignancies, particularly in breast cancer, show increasing reproducibility of detection methods for these rare tumor cells in the circulation. However, no method has yet received full recommendation to use in clinical practice, in part because of lack of a sufficient evidence base regarding clinical utility. In CNS malignancies, one of the main challenges is finding a suitable biomarker for identification of these cells, because automated systems, such as the widely used Cell Search system, are reliant on markers, such as the epithelial cell adhesion molecule, which are not present in CNS tumors. This review examines methods for CTC enrichment and detection, and reviews the progress in non-CNS tumors and the potential for using this technique in human brain tumors.
Collapse
Affiliation(s)
- Lukasz A Adamczyk
- Department of Cellular Pathology, North Bristol NHS Trust , Bristol , UK
| | - Hannah Williams
- Brain Tumor Research Group, Institute of Clinical Neuroscience, North Bristol NHS Trust , Bristol , UK
| | - Aleksandra Frankow
- IGF and Metabolic Endocrinology Group, School of Clinical Sciences, North Bristol NHS Trust, University of Bristol , Bristol , UK
| | - Hayley Patricia Ellis
- Brain Tumor Research Group, Institute of Clinical Neuroscience, North Bristol NHS Trust , Bristol , UK
| | - Harry R Haynes
- Brain Tumor Research Group, Institute of Clinical Neuroscience, North Bristol NHS Trust , Bristol , UK
| | - Claire Perks
- IGF and Metabolic Endocrinology Group, School of Clinical Sciences, North Bristol NHS Trust, University of Bristol , Bristol , UK
| | - Jeff M P Holly
- IGF and Metabolic Endocrinology Group, School of Clinical Sciences, North Bristol NHS Trust, University of Bristol , Bristol , UK
| | - Kathreena M Kurian
- Brain Tumor Research Group, Institute of Clinical Neuroscience, North Bristol NHS Trust , Bristol , UK
| |
Collapse
|
33
|
Kolostova K, Spicka J, Matkowski R, Bobek V. Isolation, primary culture, morphological and molecular characterization of circulating tumor cells in gynecological cancers. Am J Transl Res 2015; 7:1203-1213. [PMID: 26328005 PMCID: PMC4548313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 07/10/2015] [Indexed: 06/04/2023]
Abstract
The focus of the study was to implement a new workflow for circulating tumor cells (CTCs) characterization that would allow the analysis of CTCs on a cytomorphological and molecular level in patients with diagnosed gynecological cancer. Our findings may be useful in future cancer patient management. The study introduces a size-based enrichment (MetaCell(®)) method for the separation of viable CTCs, followed by CTCs culturing in vitro and gene expression characterization. It is based on the observation of CTCs and DTCs (Disseminated Tumor Cells) in several case studies of ovarian, endometrial and cervical cancer by means of cytomorphology and gene expression profiling. The viability of the enriched CTCs was estimated using vital and lethal fluorescence nuclear staining. This type of staining may be predictive for the success rate of subsequent CTC growth in vitro. To identify CTCs in the enriched CTC fraction, cytomorphological evaluations based on vital fluorescence staining were followed by gene expression analysis of tumor-associated (TA) genes. Cytokeratin expression (KRT7, KRT19) was analyzed in combination with MUC1, MUC16, CD24, CD44 and ALDH1. Gene expression analysis has shown that short-term in vitro culture enhanced the differentiation process of the captured CTCs growing on a membrane. On the other hand, redundant white blood cells captured on the membrane were eliminated during a short-term culture. The most frequently elevated genes in ovarian cancer (serous type) are EPCAM, KRT19 and MUC1. It has been demonstrated that CTC presence revealed by cytomorphological evaluation may be usefully complemented by TA-gene expression analysis, to increase the sensitivity of the analysis.
Collapse
Affiliation(s)
- Katarina Kolostova
- Department of Laboratory Genetics, Institute of Laboratory Diagnostics, University Hospital Kralovske VinohradySrobarova 50, Prague, Czech Republic
| | - Jan Spicka
- Department of Biochemistry, Institute of Laboratory Diagnostics, University Hospital Kralovske VinohradySrobarova 50, Prague, Czech Republic
| | - Rafal Matkowski
- Division of Surgical Oncology and Department of Oncology, Wroclaw Medical UniversityPlac Hirszfelda 12, 53-413 Wrocław, Poland
- Lower Silesian Oncology Centrepl. Hirszfelda 12, 53-413 Wroclaw, Poland
| | - Vladimir Bobek
- Department of Laboratory Genetics, Institute of Laboratory Diagnostics, University Hospital Kralovske VinohradySrobarova 50, Prague, Czech Republic
- 3rd Department of Surgery, First Faculty of Medicine Charles University in Prague and University Hospital MotolPrague, Czech Republic
- Department of Histology and Embryology, Wroclaw Medical UniversityChalubinskiego 6a, 50-368 Wroclaw, Poland
| |
Collapse
|
34
|
Polzer B, Medoro G, Pasch S, Fontana F, Zorzino L, Pestka A, Andergassen U, Meier-Stiegen F, Czyz ZT, Alberter B, Treitschke S, Schamberger T, Sergio M, Bregola G, Doffini A, Gianni S, Calanca A, Signorini G, Bolognesi C, Hartmann A, Fasching PA, Sandri MT, Rack B, Fehm T, Giorgini G, Manaresi N, Klein CA. Molecular profiling of single circulating tumor cells with diagnostic intention. EMBO Mol Med 2015; 6:1371-86. [PMID: 25358515 PMCID: PMC4237466 DOI: 10.15252/emmm.201404033] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Several hundred clinical trials currently explore the role of circulating tumor cell (CTC) analysis for therapy decisions, but assays are lacking for comprehensive molecular characterization of CTCs with diagnostic precision. We therefore combined a workflow for enrichment and isolation of pure CTCs with a non-random whole genome amplification method for single cells and applied it to 510 single CTCs and 189 leukocytes of 66 CTC-positive breast cancer patients. We defined a genome integrity index (GII) to identify single cells suited for molecular characterization by different molecular assays, such as diagnostic profiling of point mutations, gene amplifications and whole genomes of single cells. The reliability of > 90% for successful molecular analysis of high-quality clinical samples selected by the GII enabled assessing the molecular heterogeneity of single CTCs of metastatic breast cancer patients. We readily identified genomic disparity of potentially high relevance between primary tumors and CTCs. Microheterogeneity analysis among individual CTCs uncovered pre-existing cells resistant to ERBB2-targeted therapies suggesting ongoing microevolution at late-stage disease whose exploration may provide essential information for personalized treatment decisions and shed light into mechanisms of acquired drug resistance.
Collapse
Affiliation(s)
- Bernhard Polzer
- Project Group "Personalized Tumor Therapy", Fraunhofer Institute for Toxicology und Experimental Medicine, Regensburg, Germany
| | | | - Sophie Pasch
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | | | - Laura Zorzino
- Division of Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Aurelia Pestka
- Department of Gynecology and Obstetrics, University Munich, Munich, Germany
| | - Ulrich Andergassen
- Department of Gynecology and Obstetrics, University Munich, Munich, Germany
| | | | - Zbigniew T Czyz
- Project Group "Personalized Tumor Therapy", Fraunhofer Institute for Toxicology und Experimental Medicine, Regensburg, Germany Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Barbara Alberter
- Project Group "Personalized Tumor Therapy", Fraunhofer Institute for Toxicology und Experimental Medicine, Regensburg, Germany
| | - Steffi Treitschke
- Project Group "Personalized Tumor Therapy", Fraunhofer Institute for Toxicology und Experimental Medicine, Regensburg, Germany
| | - Thomas Schamberger
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | - Arndt Hartmann
- Department of Pathology, University Erlangen, Erlangen, Germany
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, University Erlangen, Erlangen, Germany
| | - Maria T Sandri
- Division of Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Brigitte Rack
- Department of Gynecology and Obstetrics, University Munich, Munich, Germany
| | - Tanja Fehm
- Department of Gynecology and Obstetrics, University of Düsseldorf, Düsseldorf, Germany
| | | | | | - Christoph A Klein
- Project Group "Personalized Tumor Therapy", Fraunhofer Institute for Toxicology und Experimental Medicine, Regensburg, Germany Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| |
Collapse
|
35
|
Frithiof H, Welinder C, Larsson AM, Rydén L, Aaltonen K. A novel method for downstream characterization of breast cancer circulating tumor cells following CellSearch isolation. J Transl Med 2015; 13:126. [PMID: 25896421 PMCID: PMC4409738 DOI: 10.1186/s12967-015-0493-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 04/15/2015] [Indexed: 01/05/2023] Open
Abstract
Background Enumeration of circulating tumor cells (CTCs) obtained from minimally invasive blood samples has been well established as a valuable monitoring tool in metastatic and early breast cancer, as well as in several other cancer types. The gold standard technology for detecting CTCs in blood against a backdrop of millions of leukocytes is the FDA-approved CellSearch system (Janssen Diagnostics), which relies on EpCAM-based immunomagnetic separation. Secondary characterization of these cells could enable treatment selection based on specific targets in these cells, as well as providing a real time window into the metastatic process and offering unique insights into tumor heterogeneity. The objective of this study was to develop a method for downstream characterization of CTCs following isolation with the CellSearch system. Methods An in vitro CTC model system focusing on clinically useful treatment predictive biomarkers in breast cancer, specifically the estrogen receptor α (ERα) and the human epidermal growth factor receptor 2 (HER2), was established using healthy donor blood spiked with breast cancer cell lines MCF7 (ERα+/HER2−) and SKBr3 (ERα−/HER2+). Following CTC isolation by CellSearch, the captured CTCs were further enriched and fixed on a microscope slide using the in-house-developed CTC-DropMount technique. Results The recovery rate of CTCs after CellSearch Profile analysis and CTC-DropMount was 87%. A selective and consistent triple-immunostaining protocol was optimized. Cells positive for DAPI, cytokeratin (CK) 8, 18 and 19, but negative for the leukocyte-specific marker CD45, were classified as CTCs and subsequently analyzed for ERα and HER2 expression. The method was verified in breast cancer patient samples, thus demonstrating its clinical relevance. Conclusions Our results show that it is possible to ascertain the status of important predictive biomarkers expressed in breast cancer CTCs using the newly developed CTC-DropMount technique. Downstream characterization of multiple biomarkers using a standard fluorescence microscope demonstrates that important clinical and biological information may be obtained from a single patient blood sample following either CellSearch epithelial or profile analyses. Trial registration Clinical Trials NCT01322893
Collapse
Affiliation(s)
- Henrik Frithiof
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
| | - Charlotte Welinder
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
| | - Anna-Maria Larsson
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden. .,Skåne Department of Oncology, Skåne University Hospital, Lund, Sweden.
| | - Lisa Rydén
- Division of Surgery, Department of Clinical Sciences, Lund University, Lund, Sweden. .,Department of Surgery, Skåne University Hospital, Lund, Sweden.
| | - Kristina Aaltonen
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
| |
Collapse
|
36
|
Circulating Tumor Cells: Who is the Killer? CANCER MICROENVIRONMENT 2014; 7:161-76. [PMID: 25527469 PMCID: PMC4275541 DOI: 10.1007/s12307-014-0164-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 11/27/2014] [Indexed: 01/05/2023]
Abstract
This article is a critical note on the subject of Circulating Tumor Cells (CTC). It takes into account the tumor identity of Circulating Tumor Cells as cancer seeds in transit from primary to secondary soils, rather than as a “biomarker”, and considers the help this field could bring to cancer patients. It is not meant to duplicate information already available in a large number of reviews, but to stimulate considerations, further studies and development helping the clinical use of tumor cells isolated from blood as a modern personalized, non-invasive, predictive test to improve cancer patients’ life. The analysis of CTC challenges, methodological bias and critical issues points out to the need of referring to tumor cells extracted from blood without any bias and identified by cytopathological diagnosis as Circulating Cancer Cells (CCC). Finally, this article highlights recent developments and identifies burning questions which should be addressed to improve our understanding of the domain of CCC and their potential to change the clinical practice.
Collapse
|
37
|
Bobek V, Gurlich R, Eliasova P, Kolostova K. Circulating tumor cells in pancreatic cancer patients: Enrichment and cultivation. World J Gastroenterol 2014; 20:17163-17170. [PMID: 25493031 PMCID: PMC4258587 DOI: 10.3748/wjg.v20.i45.17163] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/20/2014] [Accepted: 07/16/2014] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate the feasibility of separation and cultivation of circulating tumor cells (CTCs) in pancreatic cancer (PaC) using a filtration device.
METHODS: In total, 24 PaC patients who were candidates for surgical treatment were enrolled into the study. Peripheral blood samples were collected before an indicated surgery. For each patient, approximately 8 mL of venous blood was drawn from the antecubital veins. A new size-based separation MetaCell® technology was used for enrichment and cultivation of CTCs in vitro. (Separated CTCs were cultured on a membrane in FBS enriched RPMI media and observed by inverted microscope. The cultured cells were analyzed by means of histochemistry and immunohistochemistry using the specific antibodies to identify the cell origin.
RESULTS: CTCs were detected in 16 patients (66.7%) of the 24 evaluable patients. The CTC positivity did not reflect the disease stage, tumor size, or lymph node involvement. The same percentage of CTC positivity was observed in the metastatic and non-metastatic patients (66.7% vs 66.7%). We report a successful isolation of CTCs in PaC patients capturing proliferating cells. The cells were captured by a capillary action driven size-based filtration approach that enabled cells cultures from the viable CTCs to be unaffected by any antibodies or lysing solutions. The captured cancer cells displayed plasticity which enabled some cells to invade the separating membrane. Further, the cancer cells in the “bottom fraction”, may represent a more invasive CTC-fraction. The CTCs were cultured in vitro for further downstream applications.
CONCLUSION: The presented size-based filtration method enables culture of CTCs in vitro for possible downstream applications.
Collapse
|
38
|
Satelli A, Brownlee Z, Mitra A, Meng QH, Li S. Circulating tumor cell enumeration with a combination of epithelial cell adhesion molecule- and cell-surface vimentin-based methods for monitoring breast cancer therapeutic response. Clin Chem 2014; 61:259-66. [PMID: 25336717 DOI: 10.1373/clinchem.2014.228122] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Detection, isolation, and enumeration of circulating tumor cells (CTCs) from cancer patients has become an important modality in clinical management of patients with breast cancer. Although CellSearch, an epithelial cell adhesion molecule (EpCAM)-based method that is used to isolate epithelial CTCs, has gained prominence, its inability to detect mesenchymal CTCs from breast cancer patients raises concerns regarding its utility in clinical management. METHODS To address this gap in technology, we recently discovered the utility of cell-surface vimentin (CSV) as a marker for detecting mesenchymal CTCs from sarcoma tumors. In the present study, we tested the sensitivity and specificity of detecting CTCs from blood collected at a random time during therapy from each of 58 patients with metastatic breast cancer by use of 84-1 (a monoclonal antibody against CSV to detect epithelial/mesenchymal-transition CTCs) and CellSearch methods. Additionally, we tested the possibility of improving the sensitivity and specificity of detection by use of additional parameters including nuclear EpCAM localization and epithelial mesenchymal ratios. RESULTS CTC counts with CSV were significant (P = 0.0053) in differentiating populations responsive and nonresponsive to treatment compared with CTC counts with CellSearch (P = 0.0564). The specificity of CTC detection was found to be highest when the sum of CTC counts from the 2 methods was above a threshold of 8 CTCs/7.5 mL. CONCLUSIONS The sum of CTC counts from the CellSearch and CSV methods appears to provide new insights for assessment of therapeutic response and thus provides a new approach to personalized medicine in breast cancer patients.
Collapse
Affiliation(s)
| | | | | | - Qing H Meng
- Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shulin Li
- Department of Pediatrics and The University of Texas Graduate School of Biomedical Sciences, Houston, TX.
| |
Collapse
|
39
|
Kros JM, Mustafa DM, Dekker LJM, Sillevis Smitt PAE, Luider TM, Zheng PP. Circulating glioma biomarkers. Neuro Oncol 2014; 17:343-60. [PMID: 25253418 DOI: 10.1093/neuonc/nou207] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 07/13/2014] [Indexed: 02/06/2023] Open
Abstract
Validated biomarkers for patients suffering from gliomas are urgently needed for standardizing measurements of the effects of treatment in daily clinical practice and trials. Circulating body fluids offer easily accessible sources for such markers. This review highlights various categories of tumor-associated circulating biomarkers identified in blood and cerebrospinal fluid of glioma patients, including circulating tumor cells, exosomes, nucleic acids, proteins, and oncometabolites. The validation and potential clinical utility of these biomarkers is briefly discussed. Although many candidate circulating protein biomarkers were reported, none of these have reached the required validation to be introduced for clinical practice. Recent developments in tracing circulating tumor cells and their derivatives as exosomes and circulating nuclear acids may become more successful in providing useful biomarkers. It is to be expected that current technical developments will contribute to the finding and validation of circulating biomarkers.
Collapse
Affiliation(s)
- Johan M Kros
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., P.-P.Z.); Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (L.J.M.D., P.A.E.S.S., T.M.L.); Brain Tumor Center Rotterdam, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., L.J.M.D., P.A.E.S.S., T.M.L., P.-P.Z.)
| | - Dana M Mustafa
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., P.-P.Z.); Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (L.J.M.D., P.A.E.S.S., T.M.L.); Brain Tumor Center Rotterdam, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., L.J.M.D., P.A.E.S.S., T.M.L., P.-P.Z.)
| | - Lennard J M Dekker
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., P.-P.Z.); Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (L.J.M.D., P.A.E.S.S., T.M.L.); Brain Tumor Center Rotterdam, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., L.J.M.D., P.A.E.S.S., T.M.L., P.-P.Z.)
| | - Peter A E Sillevis Smitt
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., P.-P.Z.); Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (L.J.M.D., P.A.E.S.S., T.M.L.); Brain Tumor Center Rotterdam, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., L.J.M.D., P.A.E.S.S., T.M.L., P.-P.Z.)
| | - Theo M Luider
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., P.-P.Z.); Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (L.J.M.D., P.A.E.S.S., T.M.L.); Brain Tumor Center Rotterdam, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., L.J.M.D., P.A.E.S.S., T.M.L., P.-P.Z.)
| | - Ping-Pin Zheng
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., P.-P.Z.); Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (L.J.M.D., P.A.E.S.S., T.M.L.); Brain Tumor Center Rotterdam, Erasmus Medical Center, Rotterdam, The Netherlands (J.M.K., D.M.M., L.J.M.D., P.A.E.S.S., T.M.L., P.-P.Z.)
| |
Collapse
|
40
|
Single cell mutational analysis of PIK3CA in circulating tumor cells and metastases in breast cancer reveals heterogeneity, discordance, and mutation persistence in cultured disseminated tumor cells from bone marrow. BMC Cancer 2014; 14:456. [PMID: 24947048 PMCID: PMC4071027 DOI: 10.1186/1471-2407-14-456] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 05/28/2014] [Indexed: 12/30/2022] Open
Abstract
Background Therapeutic decisions in cancer are generally guided by molecular biomarkers or, for some newer therapeutics, primary tumor genotype. However, because biomarkers or genotypes may change as new metastases emerge, circulating tumor cells (CTCs) from blood are being investigated for a role in guiding real-time drug selection during disease progression, expecting that CTCs will comprehensively represent the full spectrum of genomic changes in metastases. However, information is limited regarding mutational heterogeneity among CTCs and metastases in breast cancer as discerned by single cell analysis. The presence of disseminated tumor cells (DTCs) in bone marrow also carry prognostic significance in breast cancer, but with variability between CTC and DTC detection. Here we analyze a series of single tumor cells, CTCs, and DTCs for PIK3CA mutations and report CTC and corresponding metastatic genotypes. Methods We used the MagSweeper, an immunomagnetic separation device, to capture live single tumor cells from breast cancer patients’ primary and metastatic tissues, blood, and bone marrow. Single cells were screened for mutations in exons 9 and 20 of the PIK3CA gene. Captured DTCs grown in cell culture were also sequenced for PIK3CA mutations. Results Among 242 individual tumor cells isolated from 17 patients and tested for mutations, 48 mutated tumor cells were identified in three patients. Single cell analyses revealed mutational heterogeneity among CTCs and tumor cells in tissues. In a patient followed serially, there was mutational discordance between CTCs, DTCs, and metastases, and among CTCs isolated at different time points. DTCs from this patient propagated in vitro contained a PIK3CA mutation, which was maintained despite morphological changes during 21 days of cell culture. Conclusions Single cell analysis of CTCs can demonstrate genotypic heterogeneity, changes over time, and discordance from DTCs and distant metastases. We present a cautionary case showing that CTCs from any single blood draw do not always reflect metastatic genotype, and that CTC and DTC analyses may provide independent clinical information. Isolated DTCs remain viable and can be propagated in culture while maintaining their original mutational status, potentially serving as a future resource for investigating new drug therapies.
Collapse
|