Cancer Letters

Cancer Letters

Volume 382, Issue 1, 1 November 2016, Pages 95-109
Cancer Letters

Mini-review
Radiogenomics: A systems biology approach to understanding genetic risk factors for radiotherapy toxicity?

https://doi.org/10.1016/j.canlet.2016.02.035Get rights and content

Highlights

  • Different pathways are involved in normal-tissue reaction after radiotherapy.

  • Combining different ‘omics’ techniques may identify critical pathways.

  • Functional assays may identify patient subgroups with different mechanisms.

  • A systems biology approach promises synergy from ‘omics’ and functional assays.

Abstract

Adverse reactions in normal tissue after radiotherapy (RT) limit the dose that can be given to tumour cells. Since 80% of individual variation in clinical response is estimated to be caused by patient-related factors, identifying these factors might allow prediction of patients with increased risk of developing severe reactions. While inactivation of cell renewal is considered a major cause of toxicity in early-reacting normal tissues, complex interactions involving multiple cell types, cytokines, and hypoxia seem important for late reactions. Here, we review ‘omics’ approaches such as screening of genetic polymorphisms or gene expression analysis, and assess the potential of epigenetic factors, posttranslational modification, signal transduction, and metabolism. Furthermore, functional assays have suggested possible associations with clinical risk of adverse reaction. Pathway analysis incorporating different ‘omics’ approaches may be more efficient in identifying critical pathways than pathway analysis based on single ‘omics’ data sets. Integrating these pathways with functional assays may be powerful in identifying multiple subgroups of RT patients characterised by different mechanisms. Thus ‘omics’ and functional approaches may synergise if they are integrated into radiogenomics ‘systems biology’ to facilitate the goal of individualised radiotherapy.

Introduction

Radiation therapy (RT) is an important component of modern multimodality tumour therapy and is part of the treatment in approximately 60% of cancer patients treated with curative intent [1]. Although technological advances in the delivery of RT has reduced the volume of normal tissue receiving critical radiation doses, the tumour dose is still limited by adverse effects in adjacent late-reacting normal tissue. Previous studies on RT-induced telangiectasiaof the skin suggested that after considering the effects of absorbed dose and dose per fraction, up to 80% of the observed variation in risk was associated with individual patient-related factors [2], [3], [4]. The identification of patients' individual susceptibility for the development of adverse effects from RT is an important prerequisite for individualising tumour treatment. Thus the therapeutic window might be widened by increasing the dose to the tumour in patients with relatively radioresistant normal tissue. On the other hand, patients with high risk of developing severe normal-tissue reaction might be candidates for either altered radiotherapy regimens (alternative fractionation schemes, treatment planning, or modalities), changes to surgery (e.g. in breast cancer, mastectomy rather than wide local excision plus RT) or pharmacologic interventions to ameliorate symptoms. Therefore, several approaches to develop a predictive assay for normal-tissue toxicity have been pursued in the past two decades. Here we review the novel high-throughput ‘omics’ technologies and more classical functional assays and discuss how the two approaches may interact synergistically to facilitate the identification of subgroups with different risks of RT-induced toxicity.

Section snippets

Clinical end points, mechanisms and hypotheses

Before reviewing ‘omics’ approaches and functional assays for radiation-induced normal-tissue reaction, a brief overview of the clinical end points and possible mechanisms is given. This is intended to present the context and basic principles of early and late RT-toxicities based on selected representative examples. For a more detailed discussion of mechanistic aspects, the reader is encouraged to consult comprehensive reviews on the special topics.

In tumour therapy, the aim is to prevent

Candidate gene studies of single nucleotide polymorphisms (SNPs)

Earlier studies attempted correlations of genetic variants with the risk of developing normal-tissue reaction after RT using candidate gene SNPs. The majority of the candidate genes were selected on the basis of their relation with the DNA damage response (DDR), mostly related to DNA repair, or genes involved in inflammatory or pro-fibrotic processes. Various single-centre studies suggested correlations with specific polymorphisms in TGFB1, XRCC3, XRCC1, ATM, GSTP1, GSTA1, or the cumulated

Functional assays

A large number of studies have been performed with the purpose of establishing associations between normal-tissue reaction after RT and the functional response of individual patients' cells to irradiation. End points have been either clonogenic cell survival or processes considered to be related to cellular radiosensitivity, such as DSB repair, chromosome aberrations, or apoptosis.

Normal skin fibroblasts can be readily grown in vitro and are functional cells of connective tissue. Therefore,

Promise of systems biology for identification of pathways and subgroups

The ‘omics’ approaches used so far focus on identifying genetic factors and changes in the transcriptional activity of genes involved in the development of normal-tissue reaction after RT. However, cell and tissue function is also influenced by posttranslational modification, cell signalling networks, and the microenvironment. Thus cell/tissue function depends on the interaction of multiple biological components from the cell to the systemic level. A direct correlation between the level of mRNA

Conclusions

Candidate SNP studies and gene expression studies found some associations with patients' adverse reactions after radiotherapy, although few have been validated for risk prediction. Recent large GWAS and candidate SNP studies including 1000–3000 patients identified significant associations, but heterogeneity with respect to tissues and end points limits success. However, the notion of multiple low-effect genes showing additive effects assumes that the effect of each gene is independent. An

Conflict of interest

All authors declare no conflict of interest.

Acknowledgements

This work was supported by funding from the European Union Seventh Framework Programme for research, technological development and demonstration under grant agreement no 601826 (“REQUITE”). The funding source had no involvement in the writing of this review.

References (223)

  • C. Herskind et al.

    Differentiation state of skin fibroblast cultures versus risk of subcutaneous fibrosis after radiotherapy

    Radiother. Oncol

    (1998)
  • H.P. Rodemann et al.

    Selective enrichment and biochemical characterization of seven human skin fibroblasts cell types in vitro

    Exp. Cell Res

    (1989)
  • S. Delanian et al.

    Abnormal phenotype of cultured fibroblasts in human skin with chronic radiotherapy damage

    Radiother. Oncol

    (1998)
  • C. Herskind et al.

    Spontaneous and radiation-induced differentiation of fibroblasts

    Exp. Gerontol

    (2000)
  • S.M. Bentzen et al.

    Clinical correlations between late normal tissue endpoints after radiotherapy: implications for predictive assays of radiosensitivity

    Eur. J. Cancer

    (1993)
  • G.C. Barnett et al.

    No association between SNPs regulating TGF-beta1 secretion and late radiotherapy toxicity to the breast: results from the RAPPER study

    Radiother. Oncol

    (2010)
  • H.P. Rodemann et al.

    Cellular basis of radiation-induced fibrosis

    Radiother. Oncol

    (1995)
  • C.B. Westbury et al.

    Radiation fibrosis – current clinical and therapeutic perspectives

    Clin. Oncol

    (2012)
  • J. Yarnold et al.

    Pathogenetic mechanisms in radiation fibrosis

    Radiother. Oncol

    (2010)
  • Z. Vujaskovic et al.

    Radiation-induced hypoxia may perpetuate late normal tissue injury

    Int. J. Radiat. Oncol. Biol. Phys

    (2001)
  • M. Martin et al.

    TGF-beta1 and radiation fibrosis: a master switch and a specific therapeutic target?

    Int. J. Radiat. Oncol. Biol. Phys

    (2000)
  • D. Pohlers et al.

    TGF-beta and fibrosis in different organs – molecular pathway imprints

    Biochim. Biophys. Acta

    (2009)
  • T. Grantzau et al.

    Risk of second non-breast cancer after radiotherapy for breast cancer: a systematic review and meta-analysis of 762,468 patients

    Radiother. Oncol

    (2015)
  • C.N. Andreassen et al.

    TGFB1 polymorphisms are associated with risk of late normal tissue complications in the breast after radiotherapy for early breast cancer

    Radiother. Oncol

    (2005)
  • C.N. Andreassen et al.

    Prediction of normal tissue radiosensitivity from polymorphisms in candidate genes

    Radiother. Oncol

    (2003)
  • C.N. Andreassen et al.

    ATM sequence variants and risk of radiation-induced subcutaneous fibrosis after postmastectomy radiotherapy

    Int. J. Radiat. Oncol. Biol. Phys

    (2006)
  • S. Terrazzino et al.

    Common variants of GSTP1, GSTA1, and TGFbeta1 are associated with the risk of radiation-induced fibrosis in breast cancer patients

    Int. J. Radiat. Oncol. Biol. Phys

    (2012)
  • K. De Ruyck et al.

    TGFbeta1 polymorphisms and late clinical radiosensitivity in patients treated for gynecologic tumors

    Int. J. Radiat. Oncol. Biol. Phys

    (2006)
  • C.N. Andreassen et al.

    Genetic variants and normal tissue toxicity after radiotherapy: a systematic review

    Radiother. Oncol

    (2009)
  • G.C. Barnett et al.

    Independent validation of genes and polymorphisms reported to be associated with radiation toxicity: a prospective analysis study

    Lancet Oncol

    (2012)
  • G.C. Barnett et al.

    Individual patient data meta-analysis shows no association between the SNP rs1800469 in TGFB and late radiotherapy toxicity

    Radiother. Oncol

    (2012)
  • A.M. Voets et al.

    No association between TGF-beta1 polymorphisms and radiation-induced lung toxicity in a European cohort of lung cancer patients

    Radiother. Oncol

    (2012)
  • G.C. Barnett et al.

    Standardized total average toxicity score: a scale- and grade-independent measure of late radiotherapy toxicity to facilitate pooling of data from different studies

    Int. J. Radiat. Oncol. Biol. Phys

    (2012)
  • H. Edvardsen et al.

    SNP in TXNRD2 associated with radiation-induced fibrosis: a study of genetic variation in reactive oxygen species metabolism and signaling

    Int. J. Radiat. Oncol. Biol. Phys

    (2013)
  • P. Seibold et al.

    XRCC1 polymorphism associated with late toxicity after radiation therapy in breast cancer patients

    Int. J. Radiat. Oncol. Biol. Phys

    (2015)
  • J.L. Guerra et al.

    Association between single nucleotide polymorphisms of the transforming growth factor beta1 gene and the risk of severe radiation esophagitis in patients with lung cancer

    Radiother. Oncol

    (2012)
  • J.L. Lopez Guerra et al.

    Functional promoter rs2868371 variant of HSPB1 associates with radiation-induced esophageal toxicity in patients with non-small-cell lung cancer treated with radio(chemo)therapy

    Radiother. Oncol

    (2011)
  • Q. Pang et al.

    Functional promoter variant rs2868371 of HSPB1 is associated with risk of radiation pneumonitis after chemoradiation for non-small cell lung cancer

    Int. J. Radiat. Oncol. Biol. Phys

    (2013)
  • S.L. Kerns et al.

    Genome-wide association study to identify single nucleotide polymorphisms (SNPs) associated with the development of erectile dysfunction in African-American men after radiotherapy for prostate cancer

    Int. J. Radiat. Oncol. Biol. Phys

    (2010)
  • S.L. Kerns et al.

    A 2-stage genome-wide association study to identify single nucleotide polymorphisms associated with development of erectile dysfunction following radiation therapy for prostate cancer

    Int. J. Radiat. Oncol. Biol. Phys

    (2013)
  • S.L. Kerns et al.

    A 2-stage genome-wide association study to identify single nucleotide polymorphisms associated with development of urinary symptoms after radiotherapy for prostate cancer

    J. Urol

    (2013)
  • S.L. Kerns et al.

    Genome-wide association study identifies a region on chromosome 11q14.3 associated with late rectal bleeding following radiation therapy for prostate cancer

    Radiother. Oncol

    (2013)
  • G.C. Barnett et al.

    A genome wide association study (GWAS) providing evidence of an association between common genetic variants and late radiotherapy toxicity

    Radiother. Oncol

    (2014)
  • M. Someya et al.

    Relation between Ku80 and microRNA-99a expression and late rectal bleeding after radiotherapy for prostate cancer

    Radiother. Oncol

    (2015)
  • J. Alsner et al.

    Differential gene expression before and after ionizing radiation of subcutaneous fibroblasts identifies breast cancer patients resistant to radiation-induced fibrosis

    Radiother. Oncol

    (2007)
  • O.K. Rodningen et al.

    Radiation-induced gene expression in human subcutaneous fibroblasts is predictive of radiation-induced fibrosis

    Radiother. Oncol

    (2008)
  • C.N. Andreassen et al.

    Independent prospective validation of a predictive test for risk of radiation induced fibrosis based on the gene expression pattern in fibroblasts irradiated in vitro

    Radiother. Oncol

    (2013)
  • C. Mayer et al.

    A radiation-induced gene expression signature as a tool to predict acute radiotherapy-induced adverse side effects

    Cancer Lett

    (2011)
  • A. Jemal et al.

    Global cancer statistics

    CA Cancer J. Clin

    (2011)
  • C.S. Potten

    Cell cycles in cell hierarchies

    Int. J. Radiat. Biol. Relat. Stud. Phys. Chem. Med

    (1986)
  • Cited by (64)

    View all citing articles on Scopus
    View full text