Cancer Letters

Cancer Letters

Volume 380, Issue 1, 28 September 2016, Pages 205-215
Cancer Letters

Mini-review
Tumor microenvironment and cancer therapy resistance

https://doi.org/10.1016/j.canlet.2015.07.044Get rights and content

Highlights

  • Cancer gains resistance via multiple mechanisms involving cell intrinsic and extrinsic factors, but the latter often remains overlooked.

  • Increasing evidence suggests that the TME plays a critical role in multiple aspects of cancer progression particularly resistance.

  • Treatment itself frequently activates the TME, and a remodeled TME can accelerate cancer cell repopulation and exacerbate disease conditions.

  • This review provides a body of updated information and delivers a balanced account of newly emerging fields in cancer research.

  • A significant guide is presented to advance basic, translational and clinical oncology in the current era of precision medicine.

Abstract

Innate resistance to various therapeutic interventions is a hallmark of cancer. In recent years, however, acquired resistance has emerged as a daunting challenge to anticancer treatments including chemotherapy, radiation and targeted therapy, which abolishes the efficacy of otherwise successful regimens. Cancer cells gain resistance through a variety of mechanisms in both primary and metastatic sites, involving cell intrinsic and extrinsic factors, but the latter often remains overlooked. Mounting evidence suggests critical roles played by the tumor microenvironment (TME) in multiple aspects of cancer progression particularly therapeutic resistance. The TME decreases drug penetration, confers proliferative and antiapoptotic advantages to surviving cells, facilitates resistance without causing genetic mutations and epigenetic changes, collectively modifying disease modality and distorting clinical indices. Recent studies have set the baseline for future investigation on the intricate relationship between cancer resistance and the TME in pathological backgrounds. This review provides an updated outline of research advances in TME biology and highlights the prospect of targeting the TME as an essential strategy to overcome cancer resistance and improve therapeutic outcomes through precise intervention. In the long run, continued inputs into translational medicine remain highly desired to achieve durable responses in the current era of personalized clinical oncology.

Introduction

The steps of tumor development implicate co-evolution of malignant cells and benign constituents of the surrounding stroma, while dynamic interactions between pathologically altered parenchyma and stroma within the TME represents a critical paradigm now considered among the typical hallmarks of cancer [1]. Histologically the association of infiltrating leukocytes and tumorigenesis was first described by Rudolf Virchow in 1863 to propose the potential relevance of chronic inflammation to neoplastic events [2]. Subsequently in 1889, Stephen Paget contributed a “seed and soil” concept to delineate the distinct patterns of recurrent metastatic sites in human breast cancer, and to plausibly interpret the tropism of tumor metastases to specific organs [3]. To date, a plethora of studies have disclosed the unique aspects of the TME, with its mystical veil removed and diverse characteristics ascertained. It is increasingly evident that individual compartments of the TME do not stay as quiet bystanders, but significantly regulate tumor initiation, disease progression, metastatic development, and more importantly, therapeutic response. Among multiple TME-implicated activities, clinical response to therapies is the major factor that directly determines the long term fate of patients who undergo anticancer interventions. In this review, an updated picture of tumor–stroma interaction is depicted, with a particular emphasis on the capacity of the TME in modifying cancer sensitivity to therapeutic agents. An appropriate, thorough and in-depth understanding of the functional roles of TME in disease evolution is essential for rational design, reasonable innovation and successful translation of novel anticancer approaches to precise medicine with substantially improved clinical outcomes.

Section snippets

The TME orchestrates disease progression and dominates therapeutic responses

As a most lethal age-related pathology that imperils human health, cancer progresses with the surrounding TME to achieve continuous outgrowth and ensuing metastasis that correlates with the majority of cancer-related death [4]. Despite considerable advancements in therapeutic concepts and techniques, disease relapse with limited response remains a major challenge and confers poor prognosis in clinical oncology. Cancer resistance involves intrinsic mechanisms that are determined by pre-existing

Damage responses of the TME offset therapy-enforced tumor regression

First introduced into clinics in 1940s for preliminary uses of nitrogen mustards and antifolate drugs, chemotherapy constitutes the mainstay of modern anticancer regimes and demonstrated remarkable potency in controlling malignancies including leukemia, lymphoma and most solid tumors [64]. However, the technical limitations of chemotherapy discovered by the early researchers still remain, and the major downside has been the low efficiency in distinguishing between normal and cancer cells.

Implications of personalized cancer therapy in an era of precision medicine

Personalized cancer therapy (PCT) takes advantage of informative clues from the tumor and its microenvironment, together with distinct conditions of the patient, to tailor therapeutic regimes and treat the disease more effectively with less toxicity. PCT delivers a similar concept with “individualized cancer therapy” (ICT) that aims to design strategies for a person diagnosed with cancer, through covering drug sensitivity testing, cancer biomarkers and bioinformatics detection,

Concluding remarks and future avenues

Next generation sequencing (NGS) enables genome-wide personalized oncology efforts with the specialty and infrastructure necessary for identification and prioritization of tumor genome variants, as piloted by the oncomine comprehensive panel (OCP) which represents a streamlined and broadly applicable targeted NGS system to advance precision oncology [102]. In contrast, biological mechanisms of resistance to conventional cytotoxic chemotherapeutics and targeted therapies designed for specific

Conflict of interest

There is no conflict of interest regarding the publication of this paper.

Acknowledgements

The author sincerely apologizes for not being able to thoroughly cite the contribution of all studies related to the current topic due to space restriction, and gratefully acknowledges Drs. Peter Nelson, Judith Campisi, Stephen Plymate and their lab members for inspiring discussion and insightful comments that made the manuscript possible. This work was supported by grants from the U.S. Department of Defense (DoD) Prostate Cancer Research Program (PCRP) (Idea Development Award PC111703),

References (106)

  • D. Hanahan et al.

    Hallmarks of cancer: the next generation

    Cell

    (2011)
  • F. Balkwill et al.

    Inflammation and cancer: back to Virchow?

    Lancet

    (2001)
  • S. Paget

    The distribution of secondary growths in cancer of the breast

    Lancet

    (1889)
  • F. Chen et al.

    New horizons in tumor microenvironment biology: challenges and opportunities

    BMC Med

    (2015)
  • M.R. Junttila et al.

    Influence of tumour micro-environment heterogeneity on therapeutic response

    Nature

    (2013)
  • Y. Sun

    Translational horizons in the tumor microenvironment: harnessing breakthroughs and targeting cures

    Med. Res. Rev

    (2015)
  • D.F. Quail et al.

    Microenvironmental regulation of tumor progression and metastasis

    Nat. Med

    (2013)
  • T. Song et al.

    TIMP-1 activated carcinoma-associated fibroblasts inhibit tumor apoptosis by activating SDF1/CXCR4 signaling in hepatocellular carcinoma

    Oncotarget

    (2015)
  • K. Tanaka et al.

    miR-27 is associated with chemoresistance in esophageal cancer through transformation of normal fibroblasts to cancer-associated fibroblasts

    Carcinogenesis

    (2015)
  • B.C. Ozdemir et al.

    Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival

    Cancer Cell

    (2014)
  • A.D. Rhim et al.

    Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma

    Cancer Cell

    (2014)
  • C. Duluc et al.

    Pharmacological targeting of the protein synthesis mTOR/4E-BP1 pathway in cancer-associated fibroblasts abrogates pancreatic tumour chemoresistance

    EMBO Mol. Med

    (2015)
  • 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)
  • N. Erez et al.

    Cancer associated fibroblasts express pro-inflammatory factors in human breast and ovarian tumors

    Biochem. Biophys. Res. Commun

    (2013)
  • T. Nagasaki et al.

    Interleukin-6 released by colon cancer-associated fibroblasts is critical for tumour angiogenesis: anti-interleukin-6 receptor antibody suppressed angiogenesis and inhibited tumour-stroma interaction

    Br. J. Cancer

    (2014)
  • X.H. Zhang et al.

    Selection of bone metastasis seeds by mesenchymal signals in the primary tumor stroma

    Cell

    (2013)
  • J. Yuan et al.

    Acquisition of epithelial-mesenchymal transition phenotype in the tamoxifen-resistant breast cancer cell: a new role for G protein-coupled estrogen receptor in mediating tamoxifen resistance through cancer-associated fibroblast-derived fibronectin and beta1-integrin signaling pathway in tumor cells

    Breast Cancer Res

    (2015)
  • M. Peiris-Pages et al.

    Chemotherapy induces the cancer-associated fibroblast phenotype, activating paracrine Hedgehog-GLI signalling in breast cancer cells

    Oncotarget

    (2015)
  • S.M. Weis et al.

    Tumor angiogenesis: molecular pathways and therapeutic targets

    Nat. Med

    (2011)
  • G.L. Semenza

    Cancer-stromal cell interactions mediated by hypoxia-inducible factors promote angiogenesis, lymphangiogenesis, and metastasis

    Oncogene

    (2013)
  • O. Tredan et al.

    Drug resistance and the solid tumor microenvironment

    J. Natl. Cancer Inst

    (2007)
  • M.M. Yuan et al.

    TLR3 expression correlates with apoptosis, proliferation and angiogenesis in hepatocellular carcinoma and predicts prognosis

    BMC Cancer

    (2015)
  • C. Ariotti et al.

    VEGFR1 and VEGFR2 in lip carcinogenesis and its association with microvessel density

    Tumour Biol

    (2015)
  • J.G. Lee et al.

    Erlotinib-cisplatin combination inhibits growth and angiogenesis through c-MYC and HIF-1 alpha in EGFR-mutated lung cancer in vitro and in vivo

    Neoplasia

    (2015)
  • P.S. Hegde et al.

    Predictive impact of circulating vascular endothelial growth factor in four phase III trials evaluating bevacizumab

    Clin. Cancer Res

    (2013)
  • K. Yamada et al.

    CXCL12-CXCR7 axis is important for tumor endothelial cell angiogenic property

    Int. J. Cancer

    (2015)
  • K.T. Flaherty et al.

    BEST: a randomized phase II study of vascular endothelial growth factor, RAF kinase, and mammalian target of rapamycin combination targeted therapy with Bevacizumab, Sorafenib, and Temsirolimus in advanced renal cell carcinoma-A trial of the ECOG-ACRIN Cancer Research Group (E2804)

    J. Clin. Oncol

    (2015)
  • M.M. Martino et al.

    Growth factors engineered for super-affinity to the extracellular matrix enhance tissue healing

    Science

    (2014)
  • F. Klemm et al.

    Microenvironmental regulation of therapeutic response in cancer

    Trends Cell Biol

    (2015)
  • P. Friedl et al.

    Cancer invasion and the microenvironment: plasticity and reciprocity

    Cell

    (2011)
  • V.M. Weaver et al.

    beta4 integrin-dependent formation of polarized three-dimensional architecture confers resistance to apoptosis in normal and malignant mammary epithelium

    Cancer Cell

    (2002)
  • H.L. Goel et al.

    beta1 integrins mediate resistance to ionizing radiation in vivo by inhibiting c-Jun amino terminal kinase 1

    J. Cell. Physiol

    (2013)
  • L. Seguin et al.

    An integrin beta(3)-KRAS-RalB complex drives tumour stemness and resistance to EGFR inhibition

    Nat. Cell Biol

    (2014)
  • K.R. Levental et al.

    Matrix crosslinking forces tumor progression by enhancing integrin signaling

    Cell

    (2009)
  • K.P. Olive et al.

    Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer

    Science

    (2009)
  • A. Bergamaschi et al.

    Extracellular matrix signature identifies breast cancer subgroups with different clinical outcome

    J. Pathol

    (2008)
  • A. Naba et al.

    The matrisome: in silico definition and in vivo characterization by proteomics of normal and tumor extracellular matrices

    Mol. Cell. Proteomics

    (2012)
  • L.R. Gomes et al.

    Three-dimensional microenvironment confers enhanced sensitivity to doxorubicin by reducing p53-dependent induction of autophagy

    Oncogene

    (2015)
  • M.X. Feng et al.

    Elevated autocrine EDIL3 protects hepatocellular carcinoma from anoikis through RGD-mediated integrin activation

    Mol. Cancer

    (2014)
  • D.M. Pardoll

    The blockade of immune checkpoints in cancer immunotherapy

    Nat. Rev. Cancer

    (2012)
  • F. Shojaei et al.

    G-CSF-initiated myeloid cell mobilization and angiogenesis mediate tumor refractoriness to anti-VEGF therapy in mouse models

    Proc. Natl. Acad. Sci. U.S.A.

    (2009)
  • L.J. Bayne et al.

    Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer

    Cancer Cell

    (2012)
  • G.T. Motz et al.

    The parallel lives of angiogenesis and immunosuppression: cancer and other tales

    Nat. Rev. Immunol

    (2011)
  • Y. Komohara et al.

    The coordinated actions of TIM-3 on cancer and myeloid cells in the regulation of tumorigenicity and clinical prognosis in clear cell renal cell carcinomas

    Cancer Immunol. Res

    (2015)
  • G. Finak et al.

    Stromal gene expression predicts clinical outcome in breast cancer

    Nat. Med

    (2008)
  • N. Weizman et al.

    Macrophages mediate gemcitabine resistance of pancreatic adenocarcinoma by upregulating cytidine deaminase

    Oncogene

    (2014)
  • C. Yang et al.

    Increased drug resistance in breast cancer by tumor-associated macrophages through IL-10/STAT3/bcl-2 signaling pathway

    Med. Oncol

    (2015)
  • S. Wan et al.

    Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells

    Gastroenterology

    (2014)
  • B. Ruffell et al.

    Macrophages and therapeutic resistance in cancer

    Cancer Cell

    (2015)
  • F. Hossain et al.

    Inhibition of fatty acid oxidation modulates immunosuppressive functions of myeloid-derived suppressor cells and enhances cancer therapies

    Cancer Immunol. Res

    (2015)
  • Cited by (244)

    View all citing articles on Scopus
    View full text