Associate editor: B. Teicher
Tumor stroma as targets for cancer therapy

https://doi.org/10.1016/j.pharmthera.2012.10.003Get rights and content

Abstract

Cancer is not only composed malignant epithelial component but also stromal components such as fibroblasts, endothelial cells, and inflammatory cells, by which an appropriate tumor microenvironment (TME) is formed to promote tumorigenesis, progression, and metastasis. As the most abundant component in the TME, cancer-associated fibroblasts (CAFs) are involved in multifaceted mechanistic details including remodeling the extracellular matrix, suppressing immune responses, and secreting growth factors and cytokines that mediate signaling pathways to extensively affect tumor cell growth and invasiveness, differentiation, angiogenesis, and chronic inflammatory milieu. Today, more and more therapeutic strategies are purposefully designed to target the TME as well as tumor cells. This review will focus on the role of CAFs in tumor development and the novel strategies to target this component to inhibit the tumor growth.

Introduction

Despite continued development of surgical techniques and meticulously designed chemotherapy or radiotherapy regimens, cancer remains the second leading cause of deaths after cardiovascular disease. The American Cancer Society estimates that in 2012, cancer will affect more than 1,600,000 patients and result in nearly 580,000 deaths (Siegel et al., 2012). Cancer has long been believed to be a cell-autonomous process, in which consecutive mutations in oncogenes and tumor suppressors lead to infinite tumor cell proliferation (Kenny et al., 2007). Understandably, most research focuses on these tumor cells. However, mounting evidence suggests that tumor genesis and progression are determined not only by tumor cells, but also by a favorable tumor microenvironment (TME) (Hu et al., 2009, Tyan et al., 2011), which supports the “seed [tumor cells] and soil [tumor stroma]” hypothesis proposed by Paget more than 100 years ago (Paget, 1989).

Tumor is considered a complicated “organ” with dysregulated growth (Bissell & Radisky, 2001) that consists of a highly heterogeneous population of cell including tumor cells, fibroblasts, inflammatory cells (e.g., lymphocytes and macrophages), endothelial cells, pericytes, and smooth muscle cells. Existing data clearly reveal that the TME differs distinctly from the corresponding normal stroma (Fig. 1). Rather than a “bystander”, the TME acts as an important “accomplice” of tumorigenesis and development.

Evidence suggests that there is close link between tumor cells and their TME: the basement membrane (BM) is degraded and the extracellular matrix (ECM) is overproduced; epithelial to mesenchymal transition (EMT) may occur; activated fibroblasts and inflammatory cells release plentiful growth factors, cytokines, and ECM-degrading proteases; newly formed vasculatures provide nutritional support to tumor cells (Petersen et al., 2003, Radisky and Radisky, 2007). All of the above changes in turn act on tumor cells and stroma to further alter and facilitate tumor progression and metastasis (Stuelten et al., 2010). On the basis of these data, it has been suggested that targeting the TME is a promising therapeutic approaches for diverse tumor types and disease stages (Chuaysri et al., 2009). The targets are also rapidly expanding from endothelial cells to other stromal components including fibroblasts, inflammatory cells, and the ECM.

In this review, we focus in particular on the role of the major cellular component in tumor milieu, cancer-associated fibroblasts (CAFs). We then discuss several crucial signaling pathways between the tumor and CAFs. Finally, we highlight the current advances in the development of novel therapeutic strategies against CAFs. Alone or in combination with conventional therapies, some target agents have been approved by Food and Drug Administration (FDA), but many more are currently being explored.

Section snippets

Cancer-associated fibroblasts (CAFs)

Fibroblasts represent the principal cellular component in the TME. Normal fibroblasts are in an inactive quiescent state and are embedded within the ECM consisting of collagen type I, laminin, fibronectin, and proteoglycans (Bremnes et al., 2011); they play a role in suppressing inappropriate or preneoplastic epithelial proliferation (Schauer et al., 2011). Once they are recruited, activated, and accumulated to the tumor area, these cells which are called CAFs, myofibroblasts, reactive stromal

Functional roles of CAFs in cancer progression

CAFs play a critical role in cancer-stromal interaction. They contribute to the growth and invasion of tumor cells through remodeling of the ECM, suppression of immune responses, and secretion of various soluble factors (growth factors and cytokines), to name a few. In many cancer types, fibroblasts with high α-SMA ( Tsujino et al., 2007, Chuaysri et al., 2009), FAP (Henry et al., 2007), or PDGFRβ (Paulsson et al., 2009) expression help cancer cells exhibit an aggressive malignant behavior

Therapeutic strategies designed to target the TME

Traditionally, the main treatment of patients with cancer is chemotherapy with cytotoxic agents, radiotherapy, debulking surgery, or any combination of these three methods to reduce tumor cells. With increasing understanding that alterations of the tumor milieu also play pivotal roles in tumorigenesis and progression, it is believed that targeting therapy against the TME, particularly CAFs, open up the new opportunities for cancer therapy. In general, these novel perspective treatment

Conclusions and future direction

Cancer is considered one of the most challenging diseases because of its resistance to treatment. As knowledge accumulates on the crucial roles of the tumor milieu, the consensus is that the TME is an integral part of the tumor that not only provides tumor architectural support but also affects its physiology and function. In particular, CAFs, the most abundant component of TME, play multiple roles in tumor development and therefore offer attractive targets for therapeutic intervention.

Conflict of interest statement

The authors declare that there are no conflicts of interest.

Acknowledgments

The authors thank for MD Anderson Cancer Center's Department of Scientific Publication for insightful manuscript editing and revision and Xueqin Cai for preparing our figures. J.L. was supported by an R01 grant (R01CA131183-01A2) and ovarian cancer Specialized Programs of Research Excellence (SPORE) grant (IP50CA83638) from the National Institutes of Health and Ovarian Cancer Research Fund. This work was also supported, in part, by the National Institutes of Health through MD Anderson's Cancer

References (202)

  • K. Dass et al.

    Evolving role of uPA/uPAR system in human cancers

    Cancer Treat Rev

    (2008)
  • J.F. DiPersio et al.

    Plerixafor and G-CSF versus placebo and G-CSF to mobilize hematopoietic stem cells for autologous stem cell transplantation in patients with multiple myeloma

    Blood

    (2009)
  • S. Duriseti et al.

    Antagonistic anti-urokinase plasminogen activator receptor (uPAR) antibodies significantly inhibit uPAR-mediated cellular signaling and migration

    J Biol Chem

    (2010)
  • R.A. El-Awady et al.

    Interaction of celecoxib with different anti-cancer drugs is antagonistic in breast but not in other cancer cells

    Toxicol Appl Pharmacol

    (2011)
  • N. Erez et al.

    Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner

    Cancer Cell

    (2010)
  • J. Fuxe et al.

    Pericyte requirement for anti-leak action of angiopoietin-1 and vascular remodeling in sustained inflammation

    Am J Pathol

    (2011)
  • A. Gaya et al.

    A preclinical and clinical review of aflibercept for the management of cancer

    Cancer Treat Rev

    (2012)
  • S.A. Ghamande et al.

    A phase 2, randomized, double-blind, placebo-controlled trial of clinical activity and safety of subcutaneous A6 in women with asymptomatic CA125 progression after first-line chemotherapy of epithelial ovarian cancer

    Gynecol Oncol

    (2008)
  • G. Giannelli et al.

    Transforming growth factor-β1 triggers hepatocellular carcinoma invasiveness via alpha3beta1 integrin

    Am J Pathol

    (2002)
  • G. Gokalp et al.

    Malignant spiculated breast masses: dynamic contrast enhanced MR (DCE-MR) imaging enhancement characteristics and histopathological correlation

    Eur J Radiol

    (2012)
  • T.A. Gonda et al.

    Molecular biology of cancer-associated fibroblasts: can these cells be targeted in anti-cancer therapy?

    Semin Cell Dev Biol

    (2010)
  • H. Hirte et al.

    A phase III randomized trial of BAY 12–9566 (tanomastat) as maintenance therapy in patients with advanced ovarian cancer responsive to primary surgery and paclitaxel/platinum containing chemotherapy: a National Cancer Institute of Canada Clinical Trials Group Study

    Gynecol Oncol

    (2006)
  • Y. Itoh et al.

    IL-8 promotes cell proliferation and migration through metalloproteinase-cleavage proHB-EGF in human colon carcinoma cells

    Cytokine

    (2005)
  • H. Katoh et al.

    COX-2 and prostaglandin EP3/EP4 signaling regulate the tumor stromal proangiogenic microenvironment via CXCL12–CXCR4 chemokine systems

    Am J Pathol

    (2010)
  • T. Kelly

    Fibroblast activation protein-alpha and dipeptidyl peptidase IV (CD26): cell-surface proteases that activate cell signaling and are potential targets for cancer therapy

    Drug Resist Updat

    (2005)
  • Y. Koshida et al.

    Interaction between stromal fibroblasts and colorectal cancer cells in the expression of vascular endothelial growth factor

    J Surg Res

    (2006)
  • M.I. Lin et al.

    Antiangiogenic therapy: creating a unique “window” of opportunity

    Cancer Cell

    (2004)
  • C. Anderberg et al.

    Paracrine signaling by platelet-derived growth factor-CC promotes tumor growth by recruitment of cancer-associated fibroblasts

    Cancer Res

    (2009)
  • P.A. Andreasen et al.

    The plasminogen activation system in tumor growth, invasion, and metastasis

    Cell Mol Life Sci

    (2000)
  • M. Augsten et al.

    CXCL14 is an autocrine growth factor for fibroblasts and acts as a multi-modal stimulator of prostate tumor growth

    Proc Natl Acad Sci U S A

    (2009)
  • M. Augsten et al.

    A digest on the role of the tumor microenvironment in gastrointestinal cancers

    Cancer Microenviron

    (2010)
  • M. Balsamo et al.

    Melanoma-associated fibroblasts modulate NK cell phenotype and antitumor cytotoxicity

    Proc Natl Acad Sci U S A

    (2009)
  • V. Barry-Hamilton et al.

    Allosteric inhibition of lysyl oxidase-like-2 impedes the development of a pathologic microenvironment

    Nat Med

    (2010)
  • M.M. Bertagnolli et al.

    Celecoxib for the prevention of sporadic colorectal adenomas

    N Engl J Med

    (2006)
  • P.M. Biselli-Chicote et al.

    VEGF gene alternative splicing: pro- and anti-angiogenic isoforms in cancer

    J Cancer Res Clin Oncol

    (2012)
  • M.J. Bissell et al.

    Putting tumours in context

    Nat Rev Cancer

    (2001)
  • C. Bisson et al.

    Restricted expression of membrane type 1-matrix metalloproteinase by myofibroblasts adjacent to human breast cancer cells

    Int J Cancer

    (2003)
  • U. Bogdahn et al.

    Targeted therapy for high-grade glioma with the TGF-β2 inhibitor trabedersen: results of a randomized and controlled phase IIb study

    Neuro Oncol

    (2011)
  • M.A. Buerkle et al.

    Inhibition of the alpha-nu integrins with a cyclic RGD peptide impairs angiogenesis, growth and metastasis of solid tumours in vivo

    Br J Cancer

    (2002)
  • S. Cannito et al.

    Epithelial–mesenchymal transition: from molecular mechanisms, redox regulation to implications in human health and disease

    Antioxid Redox Signal

    (2010)
  • R. Cao et al.

    Comparative evaluation of FGF-2-, VEGF-A-, and VEGF-C-induced angiogenesis, lymphangiogenesis, vascular fenestrations, and permeability

    Circ Res

    (2004)
  • A.R. Cardones et al.

    CXCR4 enhances adhesion of B16 tumor cells to endothelial cells in vitro and in vivo via beta(1) integrin

    Cancer Res

    (2003)
  • A. Carter

    Integrins as target: first phase III trial launches, but questions remain

    J Natl Cancer Inst

    (2010)
  • C. Chuaysri et al.

    Alpha-smooth muscle actin-positive fibroblasts promote biliary cell proliferation and correlate with poor survival in cholangiocarcinoma

    Oncol Rep

    (2009)
  • P. Cirri et al.

    Cancer-associated-fibroblasts and tumour cells: a diabolic liaison driving cancer progression

    Cancer Metastasis Rev

    (2012)
  • E.C. Connolly et al.

    Outgrowth of drug-resistant carcinomas expressing markers of tumor aggression after long-term TβRI/II kinase inhibition with LY2109761

    Cancer Res

    (2011)
  • J.F. DiPersio et al.

    Phase III prospective randomized double-blind placebo-controlled trial of plerixafor plus granulocyte colony-stimulating factor compared with placebo plus granulocyte colony-stimulating factor for autologous stem-cell mobilization and transplantation for patients with non-Hodgkin's lymphoma

    J Clin Oncol

    (2009)
  • N.C. Direkze et al.

    Bone marrow contribution to tumor-associated myofibroblasts and fibroblasts

    Cancer Res

    (2004)
  • J. Dong et al.

    VEGF-null cells require PDGFR alpha signaling-mediated stromal fibroblast recruitment for tumorigenesis

    EMBO J

    (2004)
  • D.G. Duda et al.

    CXCL12 (SDF1alpha)-CXCR4/CXCR7 pathway inhibition: an emerging sensitizer for anticancer therapies?

    Clin Cancer Res

    (2011)
  • Cited by (148)

    View all citing articles on Scopus
    View full text