Review
Tumor and its microenvironment: A synergistic interplay

https://doi.org/10.1016/j.semcancer.2013.08.007Get rights and content

Abstract

The mutual and interdependent interaction between tumor and its microenvironment is a crucial topic in cancer research. Recently, it was reported that targeting stromal events could improve efficacies of current therapeutics and prevent metastatic spreading. Tumor microenvironment is a “complex network” of different cell types, soluble factors, signaling molecules and extracellular matrix components, which orchestrate the fate of tumor progression. As by definition, cancer stem cells (CSCs) are proposed to be the unique cell type able to maintain tumor mass and survive outside the primary tumor at metastatic sites. Being exposed to environmental stressors, including reactive oxygen species (ROS), CSCs have developed a GSH-dependent antioxidant system to improve ROS defense capability and acquire a malignant phenotype. Nevertheless, tumor progression is dependent on extracellular matrix remodeling, fibroblasts and macrophages activation in response to oxidative stress, as well as epithelial mesenchymal transition (EMT)-inducing signals and endothelial and perivascular cells recruitment. Besides providing a survival advantage by inducing de novo angiogenesis, tumor-associated vessels contribute to successful dissemination by facilitating tumor cells entry into the circulatory system and driving the formation of pre-metastatic niche. In this review, we focus on the synergistic effect of hypoxia inducible factors (HIFs) and vascular endothelial growth factors (VEGFs) in the successful outgrowth of metastasis, integrating therefore many of the emerging models and theories in the field.

Introduction

Colorectal cancer (CRC) is the third most commonly diagnosed cancer in the world and one of the major causes of death worldwide [1]. The prevention and the early diagnosis are surely the most important approaches for reducing the burden of CRC, given the symptoms of early disease occur just in 5% of cases. A significant portion of patients who receive surgery and adjuvant therapy still develop recurrences and metastasis and this phenomenon seems to be driven in some cell subsets by the acquisition of resistance to conventional therapy, such as chemo- and radio-therapy [2].

Growing evidence indicates that a cellular subpopulation with stem cell like features, commonly referred to as cancer stem cells (CSCs), is critical for tumor generation and maintenance.

A recent study showed that within the tumor population it is possible to identify a heterogeneous population of cells with different biological roles [3]. Recent advances in stem cell biology are revealing that this cellular fraction shares many properties with normal adult stem cells, including dormancy (quiescence), active DNA repair machinery, the expression of several ABC drugs transporters and an intrinsic resistance to apoptosis [4]. As their normal counterpart, the colon CSCs reside in a specialized microarchitectonic structures or niches that respond to both local and systemic conditions providing also protection against conventional therapies [5].

Moreover, microenvironmental stimuli, such as those involved in the epithelial-mesenchymal transition (EMT) and hypoxia, indirectly contribute to chemoresistance by inducing in cancer cells a stem like-phenotype. Understanding the driving force of tumor progression and the relationship between cancer cells and microenvironment could be fundamental in developing innovative therapeutic strategies for a better and definitive response on patient treatments.

Section snippets

CRC, stem cell niche and colon CSCs

It is widely accepted that CRC progression is driven by the acquisition of 4–5 progressive mutations in oncogenes or tumor suppressor genes [6]. Some driver mutations frequently occur in the same gene sequences and are shared by most of the people affected by this cancer, whereas some mutations are different and responsible of the final cancer phenotype in individual patients [7]. Most of the information about CRC derives from the study of familial adenomatous polyposis (FAP), an autosomal

EMT, pre-metastatic niche and metastasis formation

Metastasis formation is considered a complex multi-step process with sequential molecular and cellular events that permit transformed cells to gain access to the blood stream (intravasation), survive their journey through the blood stream, and ultimately traverse through the microvasculature of target organs (extravasation) to deposit, survive, and grow in a foreign tissue environment. The EMT represents the first step of this highly regulated cascade and it is an important biological process

Status redox and hypoxia: two sides of the same coin

In the absence of an aberrant microenvironmental stimuli, genetic and epigenetic alterations in tumor cells are insufficient to induce primary tumor progression [27]. Either through structure and function-based mechanisms, including ECM remodeling, release of cytokines and growth factors, metabolic changes, or activation of stromal components, microenvironment enables tumor cells to achieve an aggressive phenotype [32].

As observed, reactive oxygen species (ROS) have emerged as an important

CSCs and vasculature cells crosstalk: a mutual convenience

Tumor cell growth and nurture require several strategies to supply the oxygen and metabolic demand, all involving new vessels formation and captivation from the surrounding stroma. Tumor neovascularization can occur through (a) sprouting from existing vessels (sprouting angiogenesis), (b) lumen invagination and splitting of vessels (intussusceptive angiogenesis), (c) enfolding of vessels by cancer cells (vessel co-option), (d) simulation of endothelial features by tumor cells (vasculogenic

Angiogenic pathways orchestrate CSCs survival and motility

Although CSCs represent a minority of tumor cells population, deregulation of pathways involved in cell self-renewal and motility contributes to cancer conversion and promotion. In addition to well established CSCs radioresistance and chemoresistance mechanisms, an increasing adaptability to antiangiogenic treatment was shown [88]. These cells can elicit resistance and increase their tumorigenic and invasive potential by exploiting an hypoxic microenvironment [89] as well as the activation of

Therapeutic advances

Quiescent cells within the stemness niche have been associated with tumor recurrence and relapse after chemotherapy. Targeting the molecular mediators and signaling pathways affecting EMT and tumor progression may provide novel therapeutic strategies to prevent CSCs-dependent distant metastasis formation.

Fighting neovascularization to counteract cancer promotion is a crucial step of the long-standing theory of Folkman [111]. Based on this hypothesis, the first antiangiogenic compound approved

Conclusions

The reviewed data emphasize the supporting role of the microenvironment in primary tumor establishment and dissemination to distant sites. The critical event of EMT depends on the complex signals produced by stromal components ensuring the generation of CSCs phenotype with increased proliferative capacity and metastatic potential in hostile milieu. In addition, perivascular, hypoxic and premetastatic niches have been proposed to enhance the resistance of CSCs to therapy. Based on this

Conflicts of interest

None declared.

Acknowledgements

This study was supported by Grants from AIRC to G. Stassi and M. Todaro. Alice Turdo and Simone Di Franco are Ph.D. students in “International Immunopharmacology Course” at University of Palermo. Special thanks go to D. Di Franco, doctor in informatics, and Di Stefano A.B. for their cooperation in the creation of graphic images.

References (119)

  • T. Ishimoto et al.

    CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth

    Cancer Cell

    (2011)
  • S. Misra et al.

    Delivery of CD44 shRNA/nanoparticles within cancer cells: perturbation of hyaluronan/CD44v6 interactions and reduction in adenoma growth in Apc Min/+ MICE

    J Biol Chem

    (2009)
  • T. Jung et al.

    CD44v6 coordinates tumor matrix-triggered motility and apoptosis resistance

    J Biol Chem

    (2011)
  • K. Hongo et al.

    Hypoxia enhances colon cancer migration and invasion through promotion of epithelial-mesenchymal transition

    J Surg Res

    (2013)
  • S. Pennacchietti et al.

    Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene

    Cancer Cell

    (2003)
  • D. Padua et al.

    TGFbeta primes breast tumors for lung metastasis seeding through angiopoietin-like 4

    Cell

    (2008)
  • S.M. Kakonen et al.

    Transforming growth factor-beta stimulates parathyroid hormone-related protein and osteolytic metastases via Smad and mitogen-activated protein kinase signaling pathways

    J Biol Chem

    (2002)
  • J. Lu et al.

    Endothelial cells promote the colorectal cancer stem cell phenotype through a soluble form of Jagged-1

    Cancer Cell

    (2013)
  • C. Calabrese et al.

    A perivascular niche for brain tumor stem cells

    Cancer Cell

    (2007)
  • L. Cheng et al.

    Glioblastoma stem cells generate vascular pericytes to support vessel function and tumor growth

    Cell

    (2013)
  • S.P. Lin et al.

    Colon cancer stem cells resist antiangiogenesis therapy-induced apoptosis

    Cancer Lett

    (2013)
  • Y. Mizukami et al.

    Hypoxic regulation of vascular endothelial growth factor through the induction of phosphatidylinositol 3-kinase/Rho/ROCK and c-Myc

    J Biol Chem

    (2006)
  • P. Saharinen et al.

    VEGF and angiopoietin signaling in tumor angiogenesis and metastasis

    Trends Mol Med

    (2011)
  • A. Jemal et al.

    Global cancer statistics CA

    Cancer J Clin

    (2011)
  • P.A. Janne et al.

    Chemoprevention of colorectal cancer

    N Engl J Med

    (2000)
  • M. Maugeri-Sacca et al.

    Cancer stem cells and chemosensitivity

    Clin Cancer Res

    (2011)
  • Y. Sun et al.

    Molecular pathways: involving microenvironment damage responses in cancer therapy resistance

    Clin Cancer Res

    (2012)
  • B. Vogelstein et al.

    Genetic alterations during colorectal-tumor development

    N Engl J Med

    (1988)
  • P. Galiatsatos et al.

    Familial adenomatous polyposis

    Am J Gastroenterol

    (2006)
  • N.S. Fearnhead et al.

    The ABC of APC

    Hum Mol Genet

    (2001)
  • E.M.F. De Sousa et al.

    Poor-prognosis colon cancer is defined by a molecularly distinct subtype and develops from serrated precursor lesions

    Nat Med

    (2013)
  • M. Dean et al.

    Tumour stem cells and drug resistance

    Nat Rev Cancer

    (2005)
  • S. Paget

    The distribution of secondary growths in cancer of the breast 1889

    Cancer Metastasis Rev

    (1989)
  • P.A. Adegboyega et al.

    Immunohistochemical study of myofibroblasts in normal colonic mucosa, hyperplastic polyps, and adenomatous colorectal polyps

    Arch Pathol Lab Med

    (2002)
  • D.W. Powell et al.

    Myofibroblasts. II. Intestinal subepithelial myofibroblasts

    Am J Physiol

    (1999)
  • L. Vermeulen et al.

    Wnt activity defines colon cancer stem cells and is regulated by the microenvironment

    Nat Cell Biol

    (2010)
  • T.C. He et al.

    Identification of c-MYC as a target of the APC pathway

    Science

    (1998)
  • M. Shtutman et al.

    The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway

    Proc Natl Acad Sci USA

    (1999)
  • O. Tetsu et al.

    Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells

    Nature

    (1999)
  • X.C. He et al.

    PTEN-deficient intestinal stem cells initiate intestinal polyposis

    Nat Genet

    (2007)
  • S. Persad et al.

    Tumor suppressor PTEN inhibits nuclear accumulation of beta-catenin and T cell/lymphoid enhancer factor 1-mediated transcriptional activation

    J Cell Biol

    (2001)
  • C.C. Rider et al.

    Bone morphogenetic protein and growth differentiation factor cytokine families and their protein antagonists

    Biochem J

    (2010)
  • J.H. van Es et al.

    Intestinal stem cells lacking the Math1 tumour suppressor are refractory to Notch inhibitors

    Nat Commun

    (2010)
  • G.V. Hegde et al.

    Targeting of sonic hedgehog-GLI signaling: a potential strategy to improve therapy for mantle cell lymphoma

    Mol Cancer Ther

    (2008)
  • J.M. Lee et al.

    The epithelial-mesenchymal transition: new insights in signaling, development, and disease

    J Cell Biol

    (2006)
  • N.H. Le et al.

    Tumour–stroma interactions in colorectal cancer: converging on beta-catenin activation and cancer stemness

    Br J Cancer

    (2008)
  • P. Gulhati et al.

    mTORC1 and mTORC2 regulate EMT, motility, and metastasis of colorectal cancer via RhoA and Rac1 signaling pathways

    Cancer Res

    (2011)
  • A. Moustakas et al.

    Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression

    Cancer Sci

    (2007)
  • P. Friedl et al.

    Proteolytic interstitial cell migration: a five-step process

    Cancer Metastasis Rev

    (2009)
  • K. Wolf et al.

    Compensation mechanism in tumor cell migration: mesenchymal-amoeboid transition after blocking of pericellular proteolysis

    J Cell Biol

    (2003)
  • Cited by (347)

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text