Review
Sieving through the cancer secretome

https://doi.org/10.1016/j.bbapap.2013.01.030Get rights and content

Abstract

Cancer is among the most prevalent and serious health problems worldwide. Therefore, there is an urgent need for novel cancer biomarkers with high sensitivity and specificity for early detection and management of the disease. The cancer secretome, encompassing all the proteins that are secreted by cancer cells, is a promising source of biomarkers as the secreted proteins are most likely to enter the blood circulation. Moreover, since secreted proteins are responsible for signaling and communication with the tumor microenvironment, studying the cancer secretome would further the understanding of cancer biology. Latest developments in proteomics technologies have significantly advanced the study of the cancer secretome. In this review, we will present an overview of the secretome sample preparation process and summarize the data from recent secretome studies of six common cancers with high mortality (breast, colorectal, gastric, liver, lung and prostate cancers). In particular, we will focus on the various platforms that were employed and discuss the clinical applicability of the key findings in these studies. This article is part of a Special Issue entitled: An Updated Secretome.

Highlights

► Cancer is among the most prevalent and serious health problems worldwide. ► Urgent need for novel cancer biomarkers with high sensitivity and specificity ► Cancer secretome is a promising source of biomarkers. ► Overview of secretome preparation process and various analytical platforms used ► Key findings of recent secretome studies from six common cancers

Introduction

With the advances in health care, cancer has emerged as among the top diseases in many parts of the world, with global estimates of 12.7 million new cases and 7.6 million cancer deaths each year [1], [2]. Cancers of the breast, colorectum, and lung are some of the most frequently diagnosed cancers that occur anywhere regardless of human development. Unfortunately, most of the clinically useful cancer biomarkers available currently, such as carcinoembryonic antigen (CEA) and alpha-fetoprotein (AFP), are still lacking in specificity and sensitivity [3]. There is thus an urgent need for more novel and useful biomarkers, not only for early diagnosis and prognosis but as therapeutic targets as well.

Serum- or plasma-based assays remain the most popular choice for clinical screening and diagnosis. The blood circulation extends throughout the entire human body and is thus a repository of signals released from any tissue within the body. Furthermore, obtaining blood samples is simple and non-invasive. Ironically, biomarker discovery using serum or plasma samples is anything but simple. These studies are often confounded by the broad dynamic range of serum and plasma protein concentrations. The presence of high abundance proteins such as albumin, haptoglobin, transferrins and immunoglobulins hinders the detection of tumor-specific biomarkers, which are usually at very low concentration in the ranges of nanograms per milliliter [4]. As a result, biomarker discovery studies have been slowly shifting towards proteomics analyses of other proximal biological fluids and in particular, the “secretome”: proteins secreted from cancer tissue specimens and cell lines.

The term “secretome” was originally used by Tjalsma and colleagues to describe the total proteins that are released by a cell, tissue or organism [5]. These secreted proteins constitute about 10–15% of the proteins encoded in the human genome and are involved in important physiological processes such as immune defense, blood coagulation, matrix remodeling and cell signaling [6]. Proteins can be released into the extracellular space through two mechanisms: the classical secretory pathway and the non-classical secretory pathways. In the classical secretory pathway, proteins targeted for extracellular release are synthesized as protein precursors which usually contain signal peptides located at the N-terminus. These signal peptides direct the proteins to the rough endoplasmic reticulum (ER) and subsequently to the Golgi apparatus, following which the proteins would then be released into the extracellular environment in Golgi-derived secretory vesicles. Alternatively, proteins can be exported through ER/Golgi-independent mechanisms which are also known as the non-classical secretory pathways. In these non-classical secretory pathways, proteins may be exported by targeting endosomes recycling back to the plasma membrane, directly translocating across the plasma membrane, or through exosomal secretion [7]. Exosomes are intralumenal vesicles (ILVs) which are formed by inward budding from the limiting membrane of multivesicular bodies (MVBs). During the formation process, some cytosolic proteins may be incorporated into the invaginating membrane and become engulfed in the ILVs. As a result, when a MVB eventually fuses with the plasma membrane, the ILVs within are released extracellularly together with their cargo of cytosolic proteins [8].

The neoplasm is by no means a stand-alone entity. Tumor cells constantly interact with their extracellular environment to create favorable conditions for tumor progression, such as inducing angiogenesis or degrading the extracellular matrix to facilitate metastasis. These interactions are mediated by a variety of proteins secreted by the tumor cells, including growth factors, chemokines, cytokines, adhesion molecules, proteases and shed receptors. In the same way, surrounding stromal cells are also recruited by tumor cells to actively release proteins which further the progression of the neoplasia [6]. Thus, the cancer secretome can be described as constituting of proteins released from cancer-associated stromal cells, as well as all proteins that are secreted by cancer cells through classical or non-classical secretory pathways [6], or shed from the cell surface [9]. These secreted proteins present a promising source of serological biomarkers, because they are most likely to enter the blood circulation.

Section snippets

Secretome sample preparation and associated challenges

Although the cancer secretome should by definition include all proteins in the interstitial fluid or any other proximal biological fluids of the tumor tissue, it is more commonly associated with the conditioned media (CM) samples of cancer cell lines [6]. Many cancer secretome discovery studies choose to study CM from cell lines due to the difficulties in analysis of biological fluids as alluded to earlier. Although it is undeniable that cancer cell lines are unable to completely represent the

Proteomics techniques for analyzing secretomes

Advances in proteomics technology have paved the way for increasing the numbers of secreted proteins being identified in secretome studies. In addition, proteomics approaches have progressed beyond simple protein identification and moved towards reliable quantitation of protein abundance in any sample at any given time. Traditionally, 2-DE has been used to compare protein abundance by determining differences in staining intensity and has been employed in a variety of secretome studies of

Recent advances in cancer secretomics

In the earlier sections, we have briefly covered the various preparation strategies and proteomics technologies that have been developed to study the cancer secretome from CM samples. In the following sections, we report some of the secretome studies on six cancer types, viz. breast, colorectal, gastric, liver, lung and prostate cancer, that are among the most common death-causing cancers in the world, and describe the various platforms that were employed to discover secreted proteins

Perspective

The typical workflow in many of the publications presented here involves the identification of one or several most promising candidate biomarkers through comparative analyses between cancer cell lines of differing properties. Unfortunately, only a portion of these studies are taken one step further to validate these potential biomarkers in clinical samples such as patient serum, plasma or tissue. Even so, the sample sizes of the clinical specimens in majority of these studies are mostly modest

Conclusion

The cancer secretome is a valuable source for biomarkers, not only in that secreted proteins are indicators of the physiological conditions of the cells at any time point, but also because they have the highest likelihood of entering the blood circulation. Considering that cancer will likely persist as one of the top diseases in the world, there is an urgent need for highly sensitive and specific biomarkers to assist clinicians in early detection, disease staging, monitoring of treatment and

Acknowledgments

Q.L. acknowledges support from a National University of Singapore Research Scholarship. The authors declare no conflict of interest.

References (112)

  • S. Minner et al.

    Low activated leukocyte cell adhesion molecule expression is associated with advanced tumor stage and early prostate-specific antigen relapse in prostate cancer

    Hum. Pathol.

    (2011)
  • M. Tachezy et al.

    Activated leukocyte cell adhesion molecule (CD166)—its prognostic power for colorectal cancer patients

    J. Surg. Res.

    (2012)
  • S. Mathivanan et al.

    Identifying mutated proteins secreted by colon cancer cell lines using mass spectrometry

    J. Proteomics

    (2012)
  • F. Bray et al.

    Global estimates of cancer prevalence for 27 sites in the adult population in 2008

    Int. J. Cancer

    (2012)
  • A. Jemal et al.

    Global cancer statistics

    CA Cancer J. Clin.

    (2011)
  • H. Zhang et al.

    Cancer biomarker discovery in plasma using a tissue-targeted proteomic approach

    Cancer Epidemiol. Biomark. Prev.

    (2007)
  • H. Tjalsma et al.

    Signal peptide-dependent protein transport in Bacillus subtilis: a genome-based survey of the secretome

    Microbiol. Mol. Biol. Rev.

    (2000)
  • W. Nickel

    The mystery of nonclassical protein secretion. A current view on cargo proteins and potential export routes

    Eur. J. Biochem.

    (2003)
  • R.J. Simpson et al.

    Proteomic profiling of exosomes: current perspectives

    Proteomics

    (2008)
  • S. Cooper

    Reappraisal of serum starvation, the restriction point, G0, and G1 phase arrest points

    FASEB J.

    (2003)
  • J.S. Shin et al.

    Serum starvation induces G1 arrest through suppression of Skp2-CDK2 and CDK4 in SK-OV-3 cells

    Int. J. Oncol.

    (2008)
  • N.M. Hasan et al.

    Effect of serum starvation on expression and phosphorylation of PKC-alpha and p53 in V79 cells: implications for cell death

    Int. J. Cancer

    (1999)
  • L. Zander et al.

    Identification of genes deregulated during serum-free medium adaptation of a Burkitt's lymphoma cell line

    Cell Prolif.

    (2008)
  • M. Colzani et al.

    Metabolic labeling and protein linearization technology allow the study of proteins secreted by cultured cells in serum-containing media

    J. Proteome Res.

    (2009)
  • K. Eichelbaum et al.

    Selective enrichment of newly synthesized proteins for quantitative secretome analysis

    Nat. Biotechnol.

    (2012)
  • P. Dowling et al.

    Conditioned media from cell lines: a complementary model to clinical specimens for the discovery of disease-specific biomarkers

    Proteomics

    (2011)
  • F. Mbeunkui et al.

    Secretory protein enrichment and analysis: an optimized approach applied on cancer cell lines using 2D LC-MS/MS

    J. Proteome Res.

    (2006)
  • C.G. Gunawardana et al.

    Comprehensive analysis of conditioned media from ovarian cancer cell lines identifies novel candidate markers of epithelial ovarian cancer

    J. Proteome Res.

    (2009)
  • G. Sardana et al.

    Proteomic analysis of conditioned media from the PC3, LNCaP, and 22Rv1 prostate cancer cell lines: discovery and validation of candidate prostate cancer biomarkers

    J. Proteome Res.

    (2008)
  • H. Zwickl et al.

    A novel technique to specifically analyze the secretome of cells and tissues

    Electrophoresis

    (2005)
  • H. Loei et al.

    Mining the gastric cancer secretome: identification of GRN as a potential diagnostic marker for early gastric cancer

    J. Proteome Res.

    (2012)
  • M. Ashburner et al.

    Gene ontology: tool for the unification of biology. The Gene Ontology Consortium

    Nat. Genet.

    (2000)
  • V.N. Bhatia et al.

    Software tool for researching annotations of proteins: open-source protein annotation software with data visualization

    Anal. Chem.

    (2009)
  • H. Mi et al.

    The PANTHER database of protein families, subfamilies, functions and pathways

    Nucleic Acids Res.

    (2005)
  • C. Guda et al.

    pTARGET [corrected] a new method for predicting protein subcellular localization in eukaryotes

    Bioinformatics

    (2005)
  • C. Guda

    pTARGET: a web server for predicting protein subcellular localization

    Nucleic Acids Res.

    (2006)
  • S. Briesemeister et al.

    SherLoc2: a high-accuracy hybrid method for predicting subcellular localization of proteins

    J. Proteome Res.

    (2009)
  • H.N. Lin et al.

    Protein subcellular localization prediction of eukaryotes using a knowledge-based approach

    BMC Bioinforma.

    (2009)
  • T.N. Petersen et al.

    SignalP 4.0: discriminating signal peptides from transmembrane regions

    Nat. Methods

    (2011)
  • M. Boden et al.

    Prediction of subcellular localization using sequence-biased recurrent networks

    Bioinformatics

    (2005)
  • Y. Chen et al.

    SPD—a web-based secreted protein database

    Nucleic Acids Res.

    (2005)
  • J.D. Bendtsen et al.

    Feature-based prediction of non-classical and leaderless protein secretion

    Protein Eng. Des. Sel.

    (2004)
  • S. Mathivanan et al.

    ExoCarta 2012: database of exosomal proteins, RNA and lipids

    Nucleic Acids Res.

    (2012)
  • B. Muthusamy et al.

    Plasma Proteome Database as a resource for proteomics research

    Proteomics

    (2005)
  • T. Farrah et al.

    A high-confidence human plasma proteome reference set with estimated concentrations in PeptideAtlas

    Mol. Cell. Proteomics

    (2011)
  • J. Cao et al.

    Nanozeolite-driven approach for enrichment of secretory proteins in human hepatocellular carcinoma cells

    Proteomics

    (2009)
  • H.Y. Wu et al.

    Proteomics analysis of nasopharyngeal carcinoma cell secretome using a hollow fiber culture system and mass spectrometry

    J. Proteome Res.

    (2009)
  • Y.H. Chang et al.

    Cell secretome analysis using hollow fiber culture system leads to the discovery of CLIC1 protein as a novel plasma marker for nasopharyngeal carcinoma

    J. Proteome Res.

    (2009)
  • M. Stastna et al.

    Secreted proteins as a fundamental source for biomarker discovery

    Proteomics

    (2012)
  • C.M. Huang et al.

    Mass spectrometric proteomics profiles of in vivo tumor secretomes: capillary ultrafiltration sampling of regressive tumor masses

    Proteomics

    (2006)
  • Cited by (31)

    • Analysis of ovarian cancer cell secretome during epithelial to mesenchymal transition reveals a protein signature associated with advanced stages of ovarian tumors

      2021, Biochimica et Biophysica Acta - Proteins and Proteomics
      Citation Excerpt :

      In this scenario, mass-spectrometry based proteomics has been recognized as a powerful technology to tackle all the challenges in terms of protein identification and has already demonstrated its importance to discover relevant proteins and mechanisms associated with EMT or biological mechanisms associated with metastasis [10]. Special model development, sample preparation, protein fractionation and quantitative strategies allows robust generation of hypothesis and protein candidates for further validation in more complex biological systems, including clinical samples [16,22–24]. To further enrich and prioritize large lists of proteins uncovered from in vitro models of a specific biological event, we currently benefit from a large amount of data generated from a great diversity of consortia that explored genomics as well as proteomics from large cohorts of patients.

    • Integrated omics profiling identifies hypoxia-regulated genes in HCT116 colon cancer cells

      2018, Journal of Proteomics
      Citation Excerpt :

      A comprehensive analysis of the hypoxic cancer cell secretome may provide a valuable source for biomarker discovery and putative therapeutic targets for cancer treatment [36,47]. Metabolic labeling (SILAC) was applied not only for proteome analysis, but also for secretome analysis in several reports [48,49]. The main reason for using SILAC with 10% FBS is to encounter culture conditions that are similar to those of transcriptome and translatome datasets.

    • Proteomic approaches to decipher cancer cell secretome

      2018, Seminars in Cell and Developmental Biology
      Citation Excerpt :

      In particular, the classical secreted proteins are synthesized as precursors containing a N-terminal signal peptide (SP). The SP of eukaryotes is typically 15–30 amino acids long and directs the protein first to the rough endoplasmic reticulum (ER), then to the Golgi apparatus and finally to a secretory vesicle system that allows the secretion [20]. Signal peptides may have a different primary amino acid sequence but all are characterized by a hydrophobic core flanked by hydrophilic N- and C-terminal regions, with conserved amino acids at the −3 and −1 positions relative to the cleavage site [69].

    View all citing articles on Scopus

    This article is part of a Special Issue entitled: An Updated Secretome.

    View full text