Circulating endothelial cells as biomarkers in clinical oncology

https://doi.org/10.1016/j.mvr.2010.02.007Get rights and content

Abstract

Circulating endothelial cells (CECs) and circulating endothelial progenitors (CEPs) play a different role in cancer development, acting as possible markers of vascular turnover/damage (CECs) and vasculogenesis (CEPs). Preclinical and clinical data suggest that CEC enumeration might be useful to define the best treatment option for patients who are candidate to anti-angiogenic therapy, while CEPs seem to have a “catalytic” role in different steps of cancer progression and recurrence after therapy. The definition of CEC and CEP phenotype and the standardization of CEC and CEP enumeration procedures are highly warranted to use these cells as biomarkers in clinical trials in oncology, and to compare results from different studies.

Introduction

The endothelial cell turnover has always been thought to be very slow compared with other tissues. This notwithstanding, cells with endothelial morphology were found to circulate in the blood more than 35 years ago (Hladovec and Rossamn, 1973). In the following years, the endothelial nature of these cells was confirmed by immunohistochemistry (IHC) studies, and their enumeration by means of positive enrichment, IHC or flow cytometry (FC) indicated that circulating endothelial cells (CECs) are increased in a very wide spectrum of disorders encompassing vascular, autoimmune, infectious and ischemic diseases (Moldovan et al., 1994, Bertolini et al., 2006). Over the past 10 years, increased CEC counts were observed in some cancer patients (Mancuso et al., 2001, Farace et al., 2007), and these cells were studied as surrogate biomarkers of angiogenesis and anti-angiogenic drug activity in preclinical models and medical oncology (Monestiroli et al., 2001, Shaked et al., 2005a, Shaked et al., 2005b, Calleri et al., 2009). These studies also indicated that the endothelial phenotype was expressed by cells displaying a wide variety of different features (Blann et al., 2005, Bertolini et al., 2006). Some CECs had a phenotype compatible with terminally differentiated endothelial cells (EC), in some cases being apoptotic or necrotic and thus most likely derived from the turnover of vessel walls. Some other cells expressed progenitor-associated antigens in addition to endothelial antigens, and were considered as circulating endothelial progenitor (CEP) candidates.

Section snippets

CEP and CEC phenotype

There is a lack of consensus regarding the surface markers that identify CEPs and CECs, due to the lack of a marker that can unambiguously identify these cells.

By means of flow cytometry, the most widely used antigens to identify CEPs are CD34, VEGFR2 and CD133. Many investigators identify and enumerate CEPs by means of the co-expression of CD34 and VEGFR2 (Rosenzweig, 2005, Bertolini et al., 2006, Asahara et al., 1997, Shi et al., 1998), but these antigens are expressed also by mature CECs.

CEC number and viability in cancer and treatment

Endothelial cell enumeration (both CECs and CEPs) has led to the observation that these cells are increased and are more viable in some types of cancer patients compared to healthy controls (Mancuso et al., 2001, Farace et al., 2007) (Fig. 2). In breast cancer, CECs and CEPs demonstrated a strong relationship with the Nottingham prognostic index (NPI), but only CECs positively predicted higher NPI scores and correlated with tumor invasiveness and size, possibly reflecting total tumor vascular

CEPs in tumor grow and metastases

There are different lines of research studying the role of CEP in early and late stage of cancer development and metastasis. In the late '90s, Asahara et al. (1997) and Shi et al. (1998) demonstrated that human peripheral blood contains cells that are able to differentiate into endothelial cells. However, the first proof of the contribution of CEPs in tumor development was reported by Lyden et al. (2001) in Id1 deficient mice. These mice have a severe CEP defect and show impaired angiogenesis,

CEC and CEP as tools to monitor minimal residual disease

A number of investigators have suggested that vessel-lining ECs in tumors might express a specific antigenic profile. The St. Croix lab compared gene expression patterns of ECs obtained from normal resting tissues, tumors, and regenerating liver (Seaman et al., 2007). They identified 25 transcripts overexpressed in tumor versus normal ECs, including 13 that were not found in the angiogenic endothelium of regenerating liver. Those EC tumor-specific antigens were primarily cell surface molecules

CEC/CEP in the clinic

It seems now possible to anticipate two separate fields of clinical investigation for CEPs and CECs. Some CEPs (along with other pro-angiogenic cells of hematopoietic and/or mesenchymal origin) seem to have a “catalytic” role (Seande et al., 2008) in promoting angiogenesis during tumor growth, in stimulating growth of micro- and macrometastases and in rebound revascularization after certain therapies (Rafii et al., 2002, Nolan et al., 2007, Gao et al., 2008, Lyden et al., 2001, Ciarrocchi et

Conclusions

In the future, a great effort should be devoted to reach a consensus in the identification and enumeration of CECs and CEPs. Multi parametric FC seems to be a good tool to reach this goal, in spite of the lack of a single specific antigen/marker able to identify these cells and of the rarity of them.

Clinical data suggest that the identification and enumeration of CECs and CEPs might be useful to select the most appropriate therapy for patients who are candidates to anti-angiogenic treatments,

Acknowledgments

We apologize to the numerous investigators whose papers could not be cited because of space limitations. This work was supported in part by AIRC (Associazione Italiana per la Ricerca sul Cancro), ISS (Istituto Superiore di Sanità), and Ministero della Salute grant RF-IMI-2006-411189.

References (55)

  • M.B. Ruzinova et al.

    Effect of angiogenesis inhibition by Id loss and the contribution of bone-marrow-derived endothelial cells in spontaneous murine tumors

    Cancer Cell

    (2003)
  • M. Schneider et al.

    A surrogate marker to monitor angiogenesis at last

    Cancer Cell

    (2005)
  • S. Seaman et al.

    Genes that distinguish physiological and pathological angiogenesis

    Cancer Cell

    (2007)
  • Y. Shaked et al.

    Genetic heterogeneity of the vasculogenic phenotype parallels angiogenesis; implications for cellular surrogate marker analysis of antiangiogenesis

    Cancer Cell

    (2005)
  • Y. Shaked et al.

    Optimal biologic dose of metronomic chemotherapy regimens is associated with maximum antiangiogenic activity

    Blood

    (2005)
  • Y. Shaked et al.

    Rapid chemotherapy-induced acute endothelial progenitor cell mobilization: implications for antiangiogenic drugs as chemosensitizing agents

    Cancer Cell

    (2008)
  • Q. Shi et al.

    Evidence for circulating bone marrow-derived endothelial cells

    Blood

    (1998)
  • M.C. Yoder et al.

    Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals

    Blood

    (2007)
  • O. Adighibe et al.

    Is nonangiogenesis a novel pathway for cancer progression? A study using 3-dimensional tumour reconstructions

    Br. J. Cancer

    (2006)
  • T. Asahara et al.

    Isolation of putative progenitor endothelial cells for angiogenesis

    Science

    (1997)
  • G. Bergers et al.

    Models of resistance to anti-angiogenic therapy

    Nat. Rev. Cancer

    (2008)
  • F. Bertolini et al.

    The multifaceted circulating endothelial cell in cancer: towards marker and target identification

    Nat. Rev. Cancer

    (2006)
  • A.D. Blann et al.

    Circulating endothelial cells. Biomarker of vascular disease

    Thromb. Haemost.

    (2005)
  • A. Calleri et al.

    Predictive potential of angiogenic growth factors and circulating endothelial cells in breast cancer patients receiving metronomic chemotherapy plus bevacizumab

    Clin. Cancer Res.

    (2009)
  • A. Ciarrocchi et al.

    Id1 restrains p21 expression to control endothelial progenitor cell formation

    PLoS ONE

    (2007)
  • M.G. Della Porta et al.

    Immunophenotypic, cytogenetic and functional characterization of circulating endothelial cells in myelodysplastic syndromes

    Leukemia

    (2008)
  • S. Dellapasqua et al.

    Metronomic cyclophosphamide and capecitabine combined with bevacizumab in advanced breast cancer

    J. Clin. Oncol.

    (2008)
  • Cited by (49)

    • Study the relationship of endothelial damage / dysfunction due to occupational exposure to low dose ionizing radiation versus high dose exposure during radiotherapy

      2020, Cancer Treatment and Research Communications
      Citation Excerpt :

      In agreement with our result, Beerepoot et al. [23] reported that the increased levels of CECs indicate progressive disease in cancer patients. In addition, Mancuso & Bertline [24] demonstrated that CECs are biomarkers of neoplastic disease. The detection of CECs in the present study in normal control individuals by flow cytometry has proven to be a good option as these cells are very rare in the general circulation which makes them difficult to be visualized by other laboratory measures such as immunohistochemistry.

    • Circulating endothelial cells and microparticles as diagnostic and prognostic biomarkers in small-cell lung cancer

      2018, Lung Cancer
      Citation Excerpt :

      It has been reported that CECs are increased and viable in cancer patients—including those with NSCLC—compared with healthy subjects [16–18]. CECs are considered as markers for endothelial damage, and have clinical relevance in cancer [19,20]. The change in CEC counts after chemotherapy was correlated with treatment response in patients with advanced NSCLC [18,21].

    • Circulating endothelial cells and their subpopulations: Role as predictive biomarkers in antiangiogenic therapy for colorectal cancer

      2015, Clinical Colorectal Cancer
      Citation Excerpt :

      In this case, the endothelial phenotype displays a variety of different features: some CECs have a phenotype compatible with terminally differentiated endothelial cells, while in other cases they are apoptotic or necrotic and thus they likely derive from the turnover of vessel walls. Other endothelial cells express progenitor-associated antigens in addition to endothelial antigens and are considered circulating endothelial progenitors (CEPs) deriving from bone marrow rather than from vessel walls.23,24 Absolute baseline number and changes in number and viability of CECs (and CEPs) have shown predictive value for response to an antiangiogenetic therapy for breast cancer.25-28

    • CD144, CD146 and VEGFR-2 properly identify circulating endothelial cell

      2015, Revista Brasileira de Hematologia e Hemoterapia
      Citation Excerpt :

      Immunohistochemistry is not a good option for the same reasons aggravated by the extreme rarity of these cells in peripheral blood, about 0.01% of mononuclear blood cells,17,27 and a lack of staining could be erroneously interpreted as a false negative result. Therefore, none of these possibilities have emerged as the best choice, and an effective comparison of results between laboratories is difficult.31 Furthermore, several technical issues must be taken into account in order to truly analyze rare cells such as CECs.

    • Metronomic chemotherapy in veterinary patients with cancer: Rethinking the targets and strategies of chemotherapy

      2014, Veterinary Clinics of North America - Small Animal Practice
      Citation Excerpt :

      In mice and humans, CECs and CEPs have also been used to identify the optimal biological activity of metronomic drug dosing.62,63 However, debate continues as to the phenotype of these rare cell populations, and their accurate detection depends on access to a flow cytometer and other sophisticated equipment.64 Canine CECs and CEPs have been identified and isolated from whole blood.65

    View all citing articles on Scopus
    View full text