Elsevier

Biochemical Pharmacology

Volume 80, Issue 12, 15 December 2010, Pages 1904-1914
Biochemical Pharmacology

Review
Targeting inflammatory pathways for tumor radiosensitization

https://doi.org/10.1016/j.bcp.2010.06.039Get rights and content

Abstract

Although radiation therapy (RT) is an integral component of treatment of patients with many types of cancer, inherent and/or acquired resistance to the cytotoxic effects of RT is increasingly recognized as a significant impediment to effective cancer treatment. Inherent resistance is mediated by constitutively activated oncogenic, proliferative and anti-apoptotic proteins/pathways whereas acquired resistance refers to transient induction of proteins/pathways following radiation exposure. To realize the full potential of RT, it is essential to understand the signaling pathways that mediate inducible radiation resistance, a poorly characterized phenomenon, and identify druggable targets for radiosensitization. Ionizing radiation induces a multilayered signaling response in mammalian cells by activating many pro-survival pathways that converge to transiently activate a few important transcription factors (TFs), including nuclear factor kappa B (NF-κB) and signal transducers and activators of transcription (STATs), the central mediators of inflammatory and carcinogenic signaling. Together, these TFs activate a wide spectrum of pro-survival genes regulating inflammation, anti-apoptosis, invasion and angiogenesis pathways, which confer tumor cell radioresistance. Equally, radiation-induced activation of pro-inflammatory cytokine network (including interleukin (IL)-1β, IL-6 and tumor necrosis factor-α) has been shown to mediate symptom burden (pain, fatigue, local inflammation) in cancer patients. Thus, targeting radiation-induced inflammatory pathways may exert a dual effect of accentuating the tumor radioresponse and reducing normal tissue side-effects, thereby increasing the therapeutic window of cancer treatment. We review recent data demonstrating the pivotal role played by inflammatory pathways in cancer progression and modulation of radiation response.

Introduction

The functional link between inflammation and cancer was postulated many years ago. In 1863, Virchow [1] noted the presence of leukocytes in the neoplastic tissue and hypothesized that cancers originated from the sites of chronic inflammation. In 1986, Dvorak illustrated that wound healing and tumor stroma formation share several salient features [2]. Since then, the notion that chronic inflammation provides a favorable environment for cancer formation and progression has been widely accepted. Whereas the critical correlation between tumor progression and inflammation is increasingly confirmed by evolving data, it is only recently that the potential significance of these associations has become apparent. The rapid expansion of our understanding of oncogenic molecular signaling pathways has revealed the intricate functional relationships that exist among the inflammatory pathways, tumor progression, invasion and metastasis and resistance of tumors to therapy [3]. These insights have led to the development of new anti-inflammatory therapeutic approaches for cancer treatment [4], [5].

This review portrays the convergence of multiple lines of evidence that inflammatory signaling pathways modulate the response of tumors to radiation therapy (RT). The scope of this review is, however, not to detail the fundamental relationship between inflammatory pathways and tumor progression which has been thoroughly reviewed elsewhere [1], [3], [6], [7] or to elaborate on tumor immunology [8] and/or tumor immunotherapy [9], [10]. Instead, focusing on the intricate interplay between tumor responses to radiation and inflammation, this review offers insights into potential druggable targets to sensitize tumors to RT without significant collateral damage to normal tissue. It is now known that exposure to clinically relevant doses of ionizing radiation (IR) not only induces DNA damage in the nucleus, but also triggers a large network of intracellular signaling events. These include transient activation of pro-survival pathways [receptor tyrosine kinase (RTK) pathways such as the epidermal growth factor receptor (EGFR) pathway and the downstream Ras and phosphoinositide 3-kinase/atypical kinase (PI3K/Akt) signaling], activation of transcription factors [p53, nuclear factor kappa B (NF-κB), activator protein-1 (AP-1)], upregulation of the expression of chemokine receptors (CXC motif receptor 4; CXCR4) [11] and upregulation of the levels of a variety of cytokines [tumor necrosis factor alpha (TNF-α), IL-6, IL-8, urokinase-type plasminogen activator (uPA), transforming growth factor (TGF)-β] [12], [13], [14], [15], all of which are not only crucial mediators of inflammation but also control post-irradiation cell survival responses [16], [17], [18], [19], [20]. Whereas many of these inflammatory downstream responses to radiation are detrimental to normal tissue, they confer a survival advantage to tumor cells. Therefore, unlike many radiosensitization strategies that equally sensitize the tumor cells and the adjacent collaterally irradiated normal cells to RT [21], [22], [23], specific targeting of downstream components of this inducible inflammatory signaling response offer the possibility of seamlessly and simultaneously abrogating radioresistance within tumor cells and blocking deleterious inflammatory responses of normal tissues to RT (Fig. 1).

Section snippets

Radiation therapy in current cancer management

RT remains an integral part of modern cancer management in both benign and malignant diseases. More than 50% of the newly diagnosed cancer patients worldwide receive RT (alone or in combination with chemotherapy or surgery) at some point in the course of their treatment [24]. Compared to both surgery and chemotherapy, RT is unique in that it is generally non-invasive and devoid of intense systemic toxicity, and therefore an integral component of organ preserving and adjuvant treatment

Molecular biology of tumor radioresistance – a brief overview

The concept that the intrinsic tumor radiosensitivity is governed by the balance between DNA damage and DNA repair following irradiation has dominated the conceptual framework of radiobiology for decades. Although there is an undisputable cause–function relationship between the extent of radiation-induced DNA damage and the cellular consequences, recent data indicate that this may not be the sole contributor to defining tumor radiosensitivity. Instead, the fate of damaged DNA as well as the

Targeting pro-inflammatory signaling pathways for tumor radiosensitization

Among the plethora of proliferative signaling pathways triggered by sublethal doses of radiation within tumor cells (inducible signaling), inflammatory signaling cascades are unique in that they promote tumor cell survival but, when unchecked, tend to have detrimental effects in normal tissues. Therefore, not only is such an inflammatory signaling pathway inducible (as opposed to constitutive), but may also confer some degree of selectivity for the tumor cell. Consequently, targeting this

Pro-inflammatory cytokines

Radiation is known to induce multiple biological responses at the cell and tissue level via the early activation of cytokine cascades [187]. Elevations in cancer-related and treatment-induced circulating inflammatory cytokines may be partially responsible for the development of clusters of symptoms (e.g., pain, fatigue, distress, and disturbed sleep; the symptom equivalent of tumor burden that is often referred to as ‘symptom burden’) during RT. For example, a positive correlation was found

Conclusions

Despite the technological advances in the field of radiation oncology, overcoming intrinsic and inducible tumor radioresistance still remains a major conceptual and therapeutic challenge. This is particularly true when the resistance mechanism is similar in tumor cells as well as in normal cells. Targeting inflammatory signaling pathways induced by radiation offers the promise of seamless integration of a means of enhancing the intrinsic sensitivity of tumors to radiation and a means of

References (201)

  • B.B. Zhou et al.

    Caffeine abolishes the mammalian G(2)/M DNA damage checkpoint by inhibiting ataxia-telangiectasia-mutated kinase activity

    J Biol Chem

    (2000)
  • S. Burma et al.

    ATM phosphorylates histone H2AX in response to DNA double-strand breaks

    J Biol Chem

    (2001)
  • S. Burma et al.

    Role of non-homologous end joining (NHEJ) in maintaining genomic integrity

    DNA Repair (Amst)

    (2006)
  • P. Chiarugi et al.

    Redox regulation of protein tyrosine phosphatases during receptor tyrosine kinase signal transduction

    Trends Biochem Sci

    (2003)
  • J. Schlessinger et al.

    Nuclear signaling by receptor tyrosine kinases: the first robin of spring

    Cell

    (2006)
  • R. Ishizawar et al.

    c-Src and cooperating partners in human cancer

    Cancer Cell

    (2004)
  • B.B. Aggarwal

    Nuclear factor-kappaB: the enemy within

    Cancer Cell

    (2004)
  • M.A. Brach et al.

    Ionizing radiation induces expression of interleukin 6 by human fibroblasts involving activation of nuclear factor-κB

    J Biol Chem

    (1993)
  • D.E. Hallahan et al.

    Mechanisms of X-ray-mediated protooncogene c-jun expression in radiation-induced human sarcoma cell lines

    Int J Radiat Oncol Biol Phys

    (1991)
  • F. Pacifico et al.

    NF-κB in solid tumors

    Biochem Pharmacol

    (2006)
  • Q. Lin et al.

    Constitutive activation of JAK3/STAT3 in colon carcinoma tumors and cell lines: inhibition of JAK3/STAT3 signaling induces apoptosis and cell cycle arrest of colon carcinoma cells

    Am J Pathol

    (2005)
  • M. Huang et al.

    Constitutive activation of stat 3 oncogene product in human ovarian carcinoma cells

    Gynecol Oncol

    (2000)
  • F.R. Greten et al.

    Stat3 and NF-κB activation prevents apoptosis in pancreatic carcinogenesis

    Gastroenterology

    (2002)
  • J.C. Betts et al.

    The role of NF-kappa B and NF-IL6 transactivating factors in the synergistic activation of human serum amyloid A gene expression by interleukin-1 and interleukin-6

    J Biol Chem

    (1993)
  • H. Harant et al.

    Synergistic activation of interleukin-8 gene transcription by all-trans-retinoic acid and tumor necrosis factor-α involves the transcription factor NF-κB

    J Biol Chem

    (1996)
  • J.F. Schmedtje et al.

    Hypoxia induces cyclooxygenase-2 via the NF-κB p65 transcription factor in human vascular endothelial cells

    J Biol Chem

    (1997)
  • H.F. Dvorak

    Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing

    N Engl J Med

    (1986)
  • F. Balkwill et al.

    Cancer and inflammation: implications for pharmacology and therapeutics

    Clin Pharmacol Ther

    (2010)
  • L.M. Coussens et al.

    Inflammation and cancer

    Nature

    (2002)
  • A. Mantovani et al.

    Cancer-related inflammation

    Nature

    (2008)
  • S. Mocellin et al.

    Tumor immunology

    Adv Exp Med Biol

    (2007)
  • D.M. Pardoll

    Spinning molecular immunology into successful immunotherapy

    Nat Rev Immunol

    (2002)
  • J. Begley et al.

    Targeted therapies to improve tumor immunotherapy

    Clin Cancer Res

    (2008)
  • D. Chendil et al.

    Curcumin confers radiosensitizing effect in prostate cancer cell line PC-3

    Oncogene

    (2004)
  • A. Meirovitz et al.

    Cytokines levels, severity of acute mucositis and the need of PEG tube installation during chemo-radiation for head and neck cancer—a prospective pilot study

    Radiat Oncol

    (2010)
  • O. Kargiotis et al.

    uPA/uPAR downregulation inhibits radiation-induced migration, invasion and angiogenesis in IOMM-Lee meningioma cells and decreases tumor growth in vivo

    Int J Oncol

    (2008)
  • D.H. Xia et al.

    The protective effects of ambroxol on radiation lung injury and influence on production of transforming growth factor β(1) and tumor necrosis factor α

    Med Oncol

    (2009)
  • H. Eichholtz-Wirth et al.

    Altered signaling of TNFα-TNFR1 and SODD/BAG4 is responsible for radioresistance in human HT-R15 cells

    Anticancer Res

    (2002)
  • Z. Ma et al.

    Endogenously produced urokinase-type plasminogen activator is a major determinant of the basal level of activated ERK/MAP kinase and prevents apoptosis in MDA-MB-231 breast cancer cells

    J Cell Sci

    (2001)
  • Y. Miyamoto et al.

    Interleukin-6 inhibits radiation induced apoptosis in pancreatic cancer cells

    Anticancer Res

    (2001)
  • D. Zhou et al.

    Effects of NF-kappaB1 (p50) targeted gene disruption on ionizing radiation-induced NF-κB activation and TNFα, IL-1α, IL-1β and IL-6 mRNA expression in vivo

    Int J Radiat Biol

    (2001)
  • K. Valerie et al.

    Radiation-induced cell signaling: inside–out and outside–in

    Mol Cancer Ther

    (2007)
  • U. Ringborg et al.

    The Swedish Council on Technology Assessment in Health Care (SBU) systematic overview of radiotherapy for cancer including a prospective survey of radiotherapy practice in Sweden 2001—summary and conclusions

    Acta Oncol

    (2003)
  • M. Elshaikh et al.

    Advances in radiation oncology

    Annu Rev Med

    (2006)
  • D. James et al.

    Radiation oncology: rationale, technique. Results

    (2010)
  • C. Oehler et al.

    Current concepts for the combined treatment modality of ionizing radiation with anticancer agents

    Curr Pharm Des

    (2007)
  • S.M. Bentzen et al.

    Exploitable mechanisms for combining drugs with radiation: concepts, achievements and future directions

    Nat Clin Pract Oncol

    (2007)
  • N. Jaffe et al.

    Improved outlook for Ewing's sarcoma with combination chemotherapy (vincristine, actinomycin D and cyclophosphamide) and radiation therapy

    Cancer

    (1976)
  • J. Bernier

    Alteration of radiotherapy fractionation and concurrent chemotherapy: a new frontier in head and neck oncology?

    Nat Clin Pract Oncol

    (2005)
  • Cited by (134)

    • Targeted delivery and reprogramming of myeloid-derived suppressor cells (MDSCs) in cancer

      2021, Systemic Drug Delivery Strategies: Volume 2 of Delivery Strategies and Engineering Technologies in Cancer Immunotherapy
    • Small-molecule drug repurposing to target DNA damage repair and response pathways

      2021, Seminars in Cancer Biology
      Citation Excerpt :

      Opposing p53, nuclear factor-kappa B (NF-KB) transcription factor regulate genes which participate in DNA damage responses to promote cell survival. As such, diverse small molecules that target NF-KB, ranging from nutraceuticals to approved drugs, also affect DNA-damaged induced apoptosis [85,86]. As an example, the proteasome inhibitor bortezomib has been tested in the clinic with glioblastoma patients receiving standard radiation treatment.

    View all citing articles on Scopus
    View full text