Вы находитесь на странице: 1из 11

Carcinogenesis, 2015, Vol. 36, No.

3, 307–317

doi:10.1093/carcin/bgv007
Advance Access publication January 20, 2015
Review

review
Radiogenomics helps to achieve personalized therapy
by evaluating patient responses to radiation treatment

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


Zhen Guo1, Yan Shu2, Honghao Zhou1, Wei Zhang1,* and Hui Wang3,*
1
Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology,
Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, P.R. China; 2Department of
Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA and, 3Department of
Radiation Oncology, Hunan Provincial Tumor Hospital & Affiliated Tumor Hospital of Xiangya Medical School, Central South
University, Changsha 410013, P.R. China
*To whom correspondence should be addressed. Tel: +86 13973168774; Fax: +86 731 8235 4476; Email: yjsd2003@163.com. Correspondence may also be
addressed to Hui Wang. Tel: +86 13973135460; Fax: +86 731 8865 1999; Email: wanghui710327@163.com

Abstract
Radiogenomics is the whole genome application of radiogenetics, which focuses on uncovering the underlying genetic causes
of individual variation in sensitivity to radiation. There is a growing consensus that radiosensitivity is a complex, inherited
polygenic trait, dependent on the interaction of many genes involved in multiple cell processes. An understanding of the
genes involved in processes such as DNA damage response and oxidative stress response, has evolved toward examination of
how genetic variants, most often, single nucleotide polymorphisms (SNPs), may influence interindividual radioresponse. Many
experimental approaches, such as candidate SNP association studies, genome-wide association studies and massively parallel
sequencing are being proposed to address these questions. We present a review focusing on recent advances in association
studies of SNPs to radiotherapy response and discuss challenges and opportunities for further studies. We also highlight the
clinical perspective of radiogenomics in the future of personalized treatment in radiation oncology.

Introduction
Radiation therapy is a key modality in the treatment of cancer. field (4). Previous interest has primarily focused on tumor radio-
Moreover, when radiotherapy is combined with chemotherapy, curability, but a shift toward cancer survivorship in recent years
surgery or other targeted therapies, treatment efficiency is has seen growing interest in understanding radiation-induced
improved and recurrence and cancer death rates are reduced complications in normal tissues (5).
(1). The main aim of radiotherapy is to maximize local control A large patient-to-patient variability exists in radiotherapy
of the tumor while minimizing the damage to patient’s normal response despite uniform treatment protocols (6). Although
tissues. There is a spectrum of side effects (toxicities) in the some of this may be ascribed to comorbidities, body habitus and
surrounding normal tissues associated with radiotherapy (2). stochastic factors (7–9), it has become increasingly believed that
Acute/early toxicities, usually occur within 90 days after treat- genetic background takes a leading role in the interindividual
ment, are transient and healing. However, late toxicities are variation in radiosensitivity (10–13). Heritability analysis sug-
more persistent and troublesome, which may include chronic gested that 58–78% of the variance can be attributed to genetic
inflammatory processes, vascular changes, fibrosis and atrophy factors (14,15). The initial evidence for the genetic basis of radio-
(3). Tumor radiation response is the determining factor of the sensitivity came from the observation that certain individuals
radiotherapeutic effect. How to elevate radiation response in with specific genetic disorders, such as ataxia telangiectasia,
tumor cells while decrease radiosensitivity in adjacent normal Nijmegen breakage syndrome, Fanconi anemia and DNA ligase
tissues has become a core issue in the tumor radiotherapeutic IV deficiency, were hypersensitive to radiation (16). Around the

Received: September 15, 2014; Revised: December 21, 2014; Accepted: January 15, 2015

© The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.

307
308 | Carcinogenesis, 2015, Vol. 36, No. 3

Abbreviations thus contributing to radioresistance (29,30). Multiple DNA repair


pathways, including single-strand break repair, nonhomologous
BC breast cancer
end joining repair, homologous recombination repair, base exci-
GWAS genome-wide association study
sion repair, nucleotide excision repair, have evolved to deal with
HNC head and neck cancer
various DNA lesions (31,32). Blocking these repair pathways will
mRNA messenger RNA
likely result in increased radiosensitivity. DNA damage levels
NPC nasopharyngeal carcinoma
that exceed the repair capacity of cells lead to the activation
OS overall survival
of apoptosis, which is controlled by a number of proapoptotic
RP radiation pneumonitis
factors and antiapoptotic factors. Details of the DNA dam-
SNP single nucleotide polymorphism
age response network are illustrated in Figure  1. As radiation
results in free radical formation and inflammatory responses
turn of the century, there was a growing consensus that the
in exposed tissues, genes involved in oxidative stress reactions
genetics behind individual radiosensitivity is more compli-
and inflammation processes also exert a considerable influence
cated and must be considered as a so-called polygenic or com-
on the response to ionizing radiation (33,34).
plex trait dependent on the combined influence of risk alleles

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


with different frequencies and different relative risks (10). It is
of importance to increase our understanding of the molecular Candidate gene approach
pathogenesis of radiotherapy toxicity, find ways of predicting Genetic association studies have been employed to identify
those patients likely to suffer with long­term side effects and causal functional SNPs in normal tissue radiotoxicities. Most
develop new approaches for their amelioration. association studies have based on a candidate gene approach
With the evolution of DNA sequencing and bioinformatics, and investigated SNPs in genes involved in the complex radi-
radiogenomics has emerged as a new research field which stud- oresponse pathways as mentioned above (Figure  2). Through
ies the influence of genetic variations on radiation response a number of case–control studies, in which SNPs at candidate
(17–20). One aim of radiogenomics studies is to develop a risk loci were genotyped across patients with or without radiother-
model including genetic factors and clinical covariants capable apy side effects, many genetic markers have been investigated.
of predicting individual radiosensitivity and likelihood of devel- Studies into the most frequently investigated candidate genes
oping side effects after radiotherapy. Another aim is to improve within each pathway are highlighted below (Table 1).
the radiotherapeutic effect with individualized radiotherapy,
through prevention or intervention of radiation-induced mor- DNA damage repair genes
bidities, by increasing our understanding of the biological mech- XRCC1, XRCC2, XRCC3, XRCC4, XRCC5
anisms behind them (4,21–24). Many cancer patients achieve Among the DNA damage repair genes, XRCC1 is the most fre-
survivorship at the cost of treatment complications occurring quently researched candidate. Contrasting results have been
in normal tissues. The ability to predict a predisposition for reported on the impact of XRCC1 on radiation-induced normal
severe radiotherapy-induced adverse effects in normal tissues tissue toxicities. Evidence from lung cancer patients showed
could potentially aid treatment decision making and improve that XRCC1 rs25487 (G > A) may influence radiation sensitiv-
therapy effects. Avoiding or reducing radiation in these patients ity (35) and contribute to the development of grade ≥2 radia-
could lessen the likelihood of radiotoxicity-related morbidities tion pneumonitis (RP) (36). As for nasopharyngeal carcinoma
and may also potentially reduce the burden of healthcare costs (NPC) patients, rs25487 A  allele was conferred a reduced risk
incurred for the supportive care required for these conditions of late normal tissue complications after radiotherapy (37–39).
(25). There has been intense interest in the phenomenon of inter- Nevertheless, another independent study demonstrated that
individual variation in normal tissues and tumor responses to head and neck cancer (HNC) patients with the rs25487 AA/
radiotherapy in the postgenomics era. Experimental approaches, AG genotypes have a higher probability of radiation-induced
such as candidate single nucleotide polymorphism (SNP) asso- mucositis (40). When it comes to breast cancer (BC) patients, no
ciation studies, genome-wide association studies (GWASs) and positive association has been found between rs25487 and acute
massively parallel sequencing are being proposed to address toxicity (41).
these questions. Evidence of genetic polymorphisms underlying Previous findings demonstrated that XRCC2 rs3218536 (G > A)
interindividual differences in radiotherapy responses is rapidly was correlated with overall survival (OS) in non-small-cell lung
increasing. In this review, we will present a systematic summary cancer (NSCLC) patients receiving radiotherapy (42). In addition,
of the progress in radiogenomics studies. results from classification and regression tree analysis displayed
male patients with TT genotype of XRCC4 rs6869366 (G > T) and
Biological pathways associated with female patients with AG/AA genotypes of XRCC5 rs3835 (G > A)
radiosensitivity were also at increased risk of severe RP (43). Alsbeih et al. (37,38)
Radiosensitivity can be defined as the relative susceptibility of have initially indicated the role of XRCC5 rs1051677 (T > C) C allele
cells, tissues or organs to the effects of radiation (4). To preserve in radiation toxicity among NPC patients. More recently, another
genomic stability, cells have developed elaborate response path- study in Chinese NPC patients demonstrated a trend of possible
ways to handle DNA lesions caused by ionizing radiation. The association between rs861539 (C > T) of XRCC3 and radiation-
DNA damage response is a core determining factor of tumor induced fibrosis, but not rs25487 of XRCC1 (44). Additionally, BC
radioresistance or radiosensitivity which decides the cell’s fate patients with the variant allele of XRCC3 rs861539 were more
either to repair DNA damage or to undergo apoptosis if there is likely to experience skin toxicity (45).
too much damage by manipulating cell cycle checkpoints, DNA
damage repair, apoptosis and other responses via multiple sig- RAD51
nal transduction pathways (26–28). DNA damage­induced activa- The RAD51 gene family consists of several proteins that show
tion of cell cycle checkpoints at either G1/S or G2/M give cells DNA-stimulated ATPase activity and play a central role in the
time to employ DNA repair machineries to resolve DNA lesions, homologous recombination repair activation. A functional SNP
Guo et al. | 309

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


Figure 1.  Highly simplified schematic of the radiation-triggered DNA damage response (DDR). The ionizing radiation-triggered DDR is a signaling network initiated by
lesion recognition and amplified by multiple signaling transducers, which eventually activate downstream effectors to modulate cell fate by manipulating DNA damage
repair, cell cycle progression and apoptosis. Four classical signal transduction pathways, including PI3K/AKT, MAPK/ERK, NF-κB, TGF-β, participate in the regulation of
tumor radiation response. The PI3K/AKT and MAPK/ERK pathways can be activated either through ionizing radiation or through the receptor tyrosine kinases (RTK) of
EGFR and IGFR, leading to the activation of DNA-PKcs and apoptotic-associated proteins, thus regulating the NHEJ process and apoptosis. TGF-β signaling is initiated
by the binding of TGF-β to its receptors, TGFBR1 and TGFBR2, and RTK activation which then phosphorylates SMAD2/3. Activated SMAD2/3 together with SMAD4 trans-
locate into the nucleus to regulate transcription factors and gene expression. TGF-β pathway is necessary for the full activation of ATM in response to DNA damage,
affecting DNA repair by NHEJ and HR. Besides, TGF-β promotes activation of NF-κB pathway via TAK1, a TGF-β activated kinase. Ionizing radiation also activates the
NF­κB pathway by ATM. With the help of NEMO and RIP1, ATM transfer the signal to the cytoplasm and mediate the activation of TAK1 and IκB, which finally promote
NF-κB activation. All these four pathways will finally affect the expression of crucial genes in nucleus which take part in the processes of DNA damage repair, cell cycle
progression and apoptosis. In terms of DNA damage repair, γH2AX, MDC1, 53BP1 and MRE11-RAD50-NBS1 complex act as the core sensors and could directly bind to
the damaged ends of DNA fragments. ATM and ATR are primary transducers in the center of the entire DDR, which can detect various forms of damaged DNA and
recruit multiple downstream effectors, including DNA-PKcs, Ku70/Ku80, LIG4, XRCC4, BRCA1, BRCA2, RAD51 and RAD52, in order to complete two types of DNA damage
repair (NHEJ and HR) processes. Meanwhile, ATM and ATR can separately interplay with CHK2 and CHK1 and activate other cell cycle regulators like CDC25, P53, P21,
so as to disturb the function of cell cycle checkpoints and interfere with the G1/S and G2/M phase progression. 53BP1, tumor protein p53 binding protein 1; ATM, ataxia
telangiectasia mutated kinase; ATR, ATR serine/threonine kinase; BRCA1, breast cancer 1; BRCA2, breast cancer 2; CDC25, cell division cycle 25C; CHK1, checkpoint
kinase 1; CHK2, checkpoint kinase 2; DNA-PKcs, catalytic subunit of DNA-dependent protein kinase; EGFR, epidermal growth factor receptor; H2AX, H2A histone family,
member X; HDM2, HDM2 proto-oncogene; HR, homologous recombination; IGFR, insulin-like growth factor receptor; IκB, inhibitor of κB; Ku70/Ku80, X-ray repair com-
plementing defective repair in Chinese hamster cells 6/5 (XRCC6/5); LIG4, ligase IV; MAPK/ERK, mitogen-activated protein kinase/extracellular signal-regulated kinase;
MDC1, mediator of DNA damage checkpoint 1; MRE11, DNA double-strand break repair protein Mre11; NBS1, nibrin; NEMO, NF-κB essential modulator; NF-κB, nuclear
factor-kappaB; NHEJ, nonhomologous end joining repair; P21, cyclin-dependent kinase inhibitor 1A (CDKN1A); P53, tumor protein p53; PI3K/AKT, phosphatidylinositol
3-kinase/protein kinase B; RAD50, RAD50 homolog; RAD51, RAD51 recombinase; RAD52, RAD52 homolog; RIP1, receptor-interacting protein 1; SMAD2/3, SMAD family
member 2/3; TAK1, TGF-beta-activated kinase 1; TF, transcription factors; TGF-β, transforming growth factor-β; TGFBR1, transforming growth factor, beta receptor 1;
TGFBR2, transforming growth factor, beta receptor 2; XRCC4, X-ray repair complementing defective repair in Chinese hamster cells 4.

rs1801320 (G > C), located in the promoter region of RAD51, result- progression (47). A number of studies have evaluated the role of
ing in upregulated gene expression level through an increased ATM polymorphisms in RP developing for NSCLC patients upon
promoter activity by substituting G for C allele, displayed a pre- radiotherapy. Two independent studies have consistently con-
dictive value for RP development in NSCLC patients and dyspha- firmed the rs189037 (G > A) A allele as a risk allele for RP (48–50).
gia among HNC patients after radiotherapy (40), which is also Both in vitro and in vivo studies demonstrated that rs189037
an independent prognostic factor for OS in NSCLC patients (42). may affect ATM expression by reducing transcriptional activity
Moreover, a recent study firstly confirmed that RAD51 rs1801321 and interfering nuclear protein binding (49). Another two SNPs
(G > T) T allele carriers may have a better OS as for cervical can- of ATM [rs373759 (G > A) and rs228590 (C > T)] were also found
cer patients upon radiotherapy (46). to be correlated with RP (49,50). In addition, haplotype analysis
showed that the rs189037/rs228590/rs1801516 (G-T-G) haplotype
ATM have a significantly lower risk of severe RP (hazard ratio = 0.52,
ATM protein presents kinase activity induced by ionizing radia- 95% confidence interval: 0.35–0.79, P  =  0.002) (50). The variant
tion, and it is involved in DNA damage detection and cell cycle A allele of ATM rs1801516 (G > A) was correlated with increased
310 | Carcinogenesis, 2015, Vol. 36, No. 3

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


Figure 2.  Strategies for candidate gene selection. DNA repair pathway, cell cycle progression and apoptosis mediated via four radiation-induced signaling pathways
construct a DNA damage response (DDR) signaling network, which can influence cell radiation response by determining cell’s fate either to repair DNA damage or to
undergo apoptosis. Besides DDR, inflammatory responses and oxidative stress reactions may have a considerable effect on radiosensitivity by regulating the tumor
microenvironment. Genes involved in these pathways may serve as good candidates for radiogenomics study. BIM, BCL-2-interacting mediator of cell death; EGF,
endothelial growth factor; FAS, Fas cell surface death receptor; GST, glutathionine S-transferase; HIF-1, hypoxia-inducible factor-1; IL, interleukin; MCL1, myeloid cell
leukemia sequence 1; NFE2L2, nuclear factor, erythroid 2-like 2; SOD2, superoxide dismutase 2; TNF-α, tumor necrosis factor- α.

risk of severe fibrosis in NPC patients (37,38). For prostate cancer that is stimulated by binding of death ligands of the tumor
patients (51) and BC patients (52), however, there seems to be no necrosis factor superfamily to their cell surface death receptors.
association between rs1801516 and radiation toxicity. FASL, a member of the tumor necrosis factor superfamily, by
interacting with its receptor FAS, can initiate the extrinsic apop-
NBN totic pathway. The expression of FAS and FASL is increased after
NBN is a component of MRE complex (MRE11-RAD50-NBN) irradiation and has been associated with radiation-induced cell
which is involved in damage sensing, signaling and responding death (60). FASL  rs763110 (C > T) is located in the binding motif
to DNA double-strand breaks (53). HNC patients experiencing for the transcription factor CAAT/enhancer-binding protein β
severe oral mucositis after radiotherapy were correlated to NBN and has been shown to affect the biological activity of FASL pro-
rs1805794 (G > C) with an odds radio of 4.72 (54). However, nega- moter and FASL expression. Thurner et al. (61) have provided the
tive results have also been found between rs1805794 and radia- first evidence that the variant allele of rs763110 may have a pro-
tion toxicity in BC patients (55) and NSCLC patients (42). tective effect against the development of high-grade late rectal
and/or urinary side effects after prostate cancer radiotherapy.
Oxidative stress response genes Polymorphisms of TP53 gene have been investigated as pos-
Radiation treatment exert its antineoplastic effects either directly sible causes of normal tissue radiation sensitivity in numer-
by attacking cellular macromolecules, including DNA, or indi- ous cancers. rs1042522 (G > C), the most frequently studied
rectly by generating reactive oxygen species and their by-prod- SNP of TP53 to date, is associated with the ability to induce cell
ucts. TXNRD2 is a key player in the defense against oxidative apoptosis (62). Mountainous evidence have demonstrated that
damage, by reducing the oxidoreductase enzyme thioredoxin rs1042522 has a potential in predicting radiation responses, such
involved in reactive oxygen species scavenging (56). rs1139793 as radiation-induced telangiectasia for BC patients (63), RP for
(C > T) was found significantly associated with messenger RNA lung cancer patients (48) and disease progression (local recur-
(mRNA) expression level of TXNRD2 in blood. A recent study with rence and distant metastases) for NPC patients (64). A  study
a relatively long median follow-up time of >17 years has reported with a relatively small sample size of 48 prostate cancer patients
the association between radiation-induced subcutaneous fibrosis noted a new SNP of TP53, rs35117667 (C > T), which was reported
and rs1139793 in a cohort of 92 BC survivors, which was then con- for the first time and has a predictive value for developing acute
firmed in an independent cohort of 283 BC survivors (57). Subjects skin adverse effects (51). Evidence also showed that the intronic
carrying polymorphic variant rs1695 (A > G) of GSTP1, an antioxi- polymorphism at TP53 rs17883323 (C > A) was linked to high-
dant enzyme, have shown a higher significant risk of developing grade urinary toxicity among prostate cancer patients (51).
fibrosis or fat necrosis, but not acute skin toxicity after radiation Additionally, TP73 rs3765701 (A > G) was associated with survival
treatment in BC patients (58,59). The endothelial nitric oxide syn- in stage III–IV NSCLC patients receiving chemoradiation therapy
thase participates in the oxidative stress response by catalyz- (65).
ing the production of the free radical nitric oxide. An increased HDM2 is an essential negative regulator in the p53 path-
risk of acute skin toxicity has been observed in BC patients with way that directly binds to the p53 transactivation domain and
endothelial nitric oxide synthase rs1799983 (G > T) (41). inhibits its transcriptional activity. Initial results from Alsbeih
et  al. (37,38) have indicated that the variant allele of HDM2
Apoptosis-related genes rs2279744 (T > G) and rs1196333 (T > A) were associated with
Apoptosis occurs through two main pathways, the intrinsic reduced risk to develop late normal tissues complications
pathway that is triggered by various intracellular stimuli, includ- among NPC patients. The functional polymorphic variant in
ing DNA damage and oxidative stress, and the extrinsic pathway the promoter at rs2279744 has been suggested to increase the
Guo et al. | 311

Table 1.  Studies addressing the associations between candidate genes with radiation toxicities

Gene investigated SNP site Endpoint (tumor type, N) OR/HR References

XRCC1 rs25487 (G > A) Fibrosis (NPC, 155) OR = 0.41 (35–41)


Radiation pneumonitis (NSCLC, 165) HR = 0.48
Mucositis (HNC, 101) OR = 4.02
Acute skin toxicity (BC, 286) No associa-
tion
XRCC2 rs3218536 (G > A) OS (NSCLC, 228) HR = 1.70 (42)
XRCC3 rs861539 (C > T) Fibrosis (NPC, 120) OR = 3.88 (44,45)
Acute skin toxicity (BC, 87) HR: unquan-
tifiably
high
XRCC4 rs6869366 (G > T) Radiation pneumonitis (NSCLC, 261) HR = 8.67 (43)
XRCC5 rs3835 (G > A) Radiation pneumonitis (NSCLC, 261) HR = 4.69 (43)

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


rs1051677 (T > C) Fibrosis (NPC, 155) OR = 0.39 (37)
RAD51 rs1801320 (G > C) Dysphagia (HNC, 101) HR = 4.00 (40,42)
Radiation pneumonitis (NSCLC, 228) HR = 0.52
rs1801321 (G > T) OS (cervical cancer, 193) HR = 0.373 (46)
ATM rs189037 (G > A) Radiation pneumonitis (lung cancer, 253) HR = 2.49 (48,50)
Radiation pneumonitis (NSCLC, 362) HR = 0.49
rs1801516 (G > A) Fibrosis (NPC, 155) OR = 2.86 (37,38,51,52)
Acute and chronic radiation toxicities (prostate cancer, 48) No associa-
Erythema (BC, 83) tion
No associa-
tion
NBN rs1805794 (G > C) Mucositis (HNC, 183) OR = 4.728 (42,54,55)
OS (NSCLC, 228) No associa-
Acute skin reactions (BC, 446) tion
No associa-
tion
TXNRD2 rs1139793 (C > T) Fibrosis (BC, 375) OR = 2.38 (57)
GSTP1 rs1695 (A > G) Necrosis (BC, 57) OR = 2.90 (58,59)
Fibrosis (BC, 257) OR = 2.756
Endothelial nitric rs1799983 (G > T) Acute skin toxicity (BC, 286) OR = 2.473 (41)
oxide synthase
FAS rs763110 (C > T) Late rectal or urinary side effects (prostate cancer, 607) HR = 0.585 (61)
TP53 rs1042522 (G > C) Local recurrence and distant metastases (NPC, 75) HR = 0.30 (48,63,64)
Telangiectasia (BC, 409) OR = 1.66
Radiation pneumonitis (lung cancer, 253) HR = 4.53
rs35117667 (C > T) Acute skin toxicity (prostate cancer, 48) OR = 0.007 (51)
rs17883323 (C > A) Chronic toxicity of urinary tract (prostate cancer, 48) OR = 0.071 (51)
TP73 rs3765701 (A > G) OS (NSCLC, 598) HR = 1.87 (65)
HDM2 rs2279744 (T > G) Fibrosis (NPC, 155) OR = 0.49 (38)
rs1196333 (T > A) Fibrosis (NPC, 155) OR = 0.13 (38)
TGFB1 rs1800469 (C > T) Erythema (BC, 83) OR = 3.10 (38,52,68,69,74,75,79–82)
Erectile dysfunction (prostate cancer, 141) OR = 4.00
Esophageal toxicity (lung cancer, 213) HR = 3.86
Fibrosis (NPC, 155) HR = 0.57
OS (NSCLC, 109) HR = 0.46
Esophagitis (NSCLC, 198) HR = 2.53
Late adverse effects (BC, 1716) No associa-
Overall toxicity (BC, 2782) tion
Late radiation toxicity (BC, 778) No associa-
Gastrointestinal and genitourinary toxicities (prostate cancer, tion
413) No associa-
tion
No associa-
tion
rs1982073 (T > C) Fibrosis (BC, 257) OR = 0.295 (59,70)
Radiation pneumonitis (NSCLC, 164) HR = 0.39

HR, hazard ratio; N, number of sample size; OR, odds ratio.

transcriptional activator SP1 binding, resulting in higher lev- (51) noted that rs2279744 did not play a role in the incidence
els of HDM2 mRNA and protein and the subsequent attenua- of radiotherapy-induced acute and chronic effects in prostate
tion of the p53 pathway (66). On the other hand, Cintra et  al. cancer patients.
312 | Carcinogenesis, 2015, Vol. 36, No. 3

Fibroblast proliferation gene signaling, have been addressed to be correlated with erectile


dysfunction (87). Meanwhile, they demonstrated that a region
TGFB1 is a versatile cytokine involved in inflammation, cell pro-
on chromosome 11q14.3 was associated with late rectal bleed-
liferation and fibrosis, which has been convincingly linked to the
ing following radiotherapy for prostate cancer patients (88).
development of radiation-induced fibrosis (67). Multiple studies
Cell line-based GWAS to identify genes associated with radi-
have observed an association among TGFB1 SNPs and radiation
ation sensitivity also yields certain novel biomarkers. Through
toxicities, including erythema (52), fibrosis (59), erectile dys-
the initial screening in 227 human lymphoblastoid cell lines and
function (68), esophageal toxicity (69), radiation pneumonitis
further functional validation using small interfering RNA knock-
(70) and miscellaneous severe complications (38). Among them,
down in multiple tumor cell lines, it has been demonstrated that
the two most commonly studied SNPs are rs1800469 (C > T) and
C13orf34, MAD2L1, PLK4, TPD52, DEPDC1B, were all significantly
rs1982073 (T > C), which have been reported to be correlated
linked with radiosensitivity (89). More recently, Barnett et  al.
with serum level of TGFB1 (71,72). Unlike the typical studies that
(90) have performed a phase-designed GWAS (1850 discovery +
use the clinical radiation toxicities as research endpoint, Kelsey
1733 replication) with a replication phase in three independent
et al. (73) applied an objective radiologic endpoint, the slope of
cohorts who had radiotherapy for prostate or BC. Interestingly,
the dose–response curve, and confirmed that rs1800469 may
none of the SNPs with potential associations to radiotherapy

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


affect radiation sensitivity. In addition, the rs1800469 might act
toxicity in this study have previously considered to be candi-
as an independent predictor for radiotherapy outcomes as the
dates. It is noteworthy to point that the study provided strong
CC genotype carriers showed better OS, whereas CT/TT geno-
evidence that the identified SNPs were correlated with tumor
types carriers displayed a higher probability of severe radiation
site-specific toxicity (90).
esophagitis in lung cancer patients (74,75). Meanwhile, certain
As mentioned above, a striking finding in most GWASs is
attention should be given to ethnic differences as the evidence
that the identified SNPs are not involved in any of the genes
noted that rs1982073, associated with RP risk in whites, was not
related to the major pathways assumed to be of importance
correlated with RP in Chinese patients (76,77).
for the investigated phenotype. Many of the identified SNPs
Although a large number of exploratory studies and a small
are located in noncoding regions without any known function
number of confirmatory studies have claimed the positive asso-
(87,90). This experience seriously questions the value of the can-
ciations between TGFB1 SNPs and the probability of develop-
didate gene approach and broadens our understanding of the
ing late adverse effects of radiotherapy, it is remarkable that
mechanisms underlying radiation-induced normal tissue com-
few studies with relatively large sample size failed to find any
plications at the same time. The ‘intronic’ genetics and biology
association. Strikingly, results from 124 BC patients’ lympho-
have renewed enthusiasm for exploring functional implications
cytes and 15 human fibroblast strains clearly demonstrated that
as it may lead to alternative splicing or microRNAs regulation
rs1800469 genotype has no effect on cellular radiosensitivity,
(91,92). Noncoding mutations with roles in radiosensitivity will
TGFB1 protein secretion or TGFB1 expression (78). This is of par-
likely open new doors to understand genome biology and gene
ticular importance, because it implies that none of the SNPs or
regulation in radiation-induced side effects.
SNP networks that affect TGFB1 expression seem to be involved
Even though GWASs have certainly provided several interest-
in the regulation of cellular radiosensitivity. Consistently, the
ing insights and proven their worth, they do at the same time
absence of a link with radiation toxicity has just been confirmed
display a number of challenges. First of all, the SNPs detected
for rs1800469 in BC patients and prostate cancer patients from
through GWASs are mostly limited to ‘index SNPs’, which are a
different research groups with quite large sample size (79–82).
subset of common variants derived from haplotype blocks, and
So, even for TGFB1 polymorphisms which are among the most
that will typically be inefficient to capture the impact of rare var-
extensively examined SNPs in normal tissue radiobiology, it is
iants with minor allele frequency below 5% (93). In other words,
difficult to draw any definite conclusion.
it is unable to uncover the genetics of a complex polygenic trait
once and for all. This is an issue of major importance since the
Genome-wide approach genetic background of normal tissue radiosensitivity is made
The recent development of GWASs has provided a radical alter- up by a spectrum ranging from rare highly penetrant alleles to
native to the candidate gene approach. GWASs offer the oppor- common low-risk alleles (93). Deeper sequencing-based charac-
tunity to conduct a hypothesis-free survey for SNP associations terization of genomic variation, fine mapping, imputation and
without any need for a prior understanding of the biology denser SNP array are needed as they can extend the reach of
underlying the phenotype of interest. For at least two reasons GWAS to ever lower ranges of minor allele frequency (94,95).
GWASs represent an important breakthrough in the attempts to Although GWAS arrays are extraordinarily efficient at genotyp-
unravel the genetics of complex traits. Above all, GWASs have ing SNPs, they are less effective at capturing structural varia-
dramatically increased the number of compelling SNP associa- tions, such as insertions, deletions, inversions and copy number
tions reported in the study of various phenotypes (83). Secondly, variants, which may also have a role in interindividual varia-
they shed important new light on the overall properties of the tion in radiation response (96,97). In addition, GWAS is a double-
allelic architecture presumably underlying most complex phe- edged sword as it facilitates the initial identification of a region
notypes (84,85). but makes it difficult to distinguish causal mutation(s), of which
The first GWAS in radiogenomics aimed at identifying SNPs the challenge will be to separate true associations from the bliz-
associated with erectile dysfunction following radiotherapy for zard of false positives (91). Moreover, in order to keep the risk of
prostate cancer in a specific African-American ancestry with a false positives at a reasonable level and still obtain acceptable
modest sample size and noted a significant SNP rs2268363 (86). statistical power, most GWASs have been based on very large
With a two-stage design (discovery and replication), the same patient cohorts (98). For example, achieving 90% power to detect
research group expanded the sample size and further con- an allele with 5% frequency and a factor of 1.2 effect at a sta-
ducted the GWAS in a cohort composed predominantly of men tistical significance of 10−8 requires 24 994 samples. For further
of European ancestry. Twelve SNPs, which lie in or near genes discovery studies, larger GWAS will be necessary to include rare
involved in controlling erectile function or cell adhesion and sequence variants through next generation sequencing. Besides,
Guo et al. | 313

rigorous re-testing in an independent validation set is also of individual radiation toxicity associations with candidate gene
utmost importance. SNPs, implying that the published associations were either false
positives or true weak positives. It is worth to highlight that a
Challenge and opportunity three-staged internal replication study, involving 1938 patients
from three independent cohorts, has positively addressed that
Though many positive results have been reported, the findings polymorphisms near tumor necrosis factor-α were associated
have been always inconsistent and lack the ability to replicate. with overall radiotherapy toxicities in BC patients (103).
Thus, none of the associations reported so far can by any means Many instances have arisen in which initial findings have
be regarded as unambiguously proven. This can presumably be not been reproduced in follow-up studies (104). Considering that
attributed to certain methodological shortcomings. Based on small sample size can provide imprecise or incorrect estimate
theoretical considerations and experiences from other scientific of the magnitude of the observed effect, sufficient sample size
fields, we address a number of critical points in order to suc- is necessary in replication study to convincingly distinguish the
cessfully unravel the genetics of normal tissue radiosensitivity. proposed effect from no effect (105). Heterogeneity in clinical
confounding factors and endpoint phenotypes between ini-
Clearly defined endpoint
tial and replication studies can undermine the opportunity to

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


The search for clinical and biologic biomarkers associated with compare among them. Because many initial studies have been
late radiotherapy toxicity is hindered by the use of multiple and reported in populations of European descent, the challenge
different endpoints from a variety of scoring systems, which remains to extend the studies to other populations (106,107).
make it tanglesome for comparisons across studies and pool- Data ‘dredging’ is also a major problem, especially when criteria
ing of data. In addition, although simple and useful in clini- for defining phenotypes are altered to achieve statistical signifi-
cal, subjective scoring systems (e.g. mild, moderate, severe) cance worthy of publication (108). Efforts are underway to pool
for mixed endpoints (like breast appearance or urinary quality homogenous patient cohorts with well-documented radiotox-
of life) cannot sufficiently distinguish the underlying biologi- icity endpoints to provide sufficient statistical power to detect
cal mechanisms, which probably are determined by different what will probably be modest functional effects. The National
genetic components. Thus, more quantifiable, biologically rel- Cancer Institute-National Human Genome Research Institute
evant phenotypes must be clearly and specifically defined, both working group on replication in association studies has pub-
to pinpoint the variable genetic components and to compare lished a comprehensive set of guidelines, providing a number
data across institutions. Barnett et  al. (99) have proposed such of essential criteria for establishing positive replication studies
a novel metric, the Standardized Total Average Toxicity score, to (108).
try to overcome these difficulties.
Predictive models construction
Confounding factors
Development of user-friendly tools for the prediction of radia-
Another major issue relating to the phenotypes is confounding tion-induced complications support the patient and the phy-
factors. Briefly, not all studies pay equal attention to the potential sician in selecting the best therapeutic approach and avoid
confounding effects of differences in tumor site, target volume, unnecessary worsening of quality of life. In the last years,
radiotherapy technique, radiation dose, juxtaposed skin surface, numerous efforts have been performed to construct well-cali-
overall treatment time, concomitant chemotherapy, body habi- brated, integrated predictive models by combining genotyping
tus and comorbidities (8,9). For example, Barnett et al. (100) have profiles, clinical data and treatment parameters, which are suit-
noted that a larger breast volume was the greatest nongenetic able in clinical practice to identify patients at risk for developing
risk factor for the development of late toxicity in a cohort of radiotherapy-induced toxicities.
1014 BC patients. A  decision tree considered clinicopathologi- Multimetric normal tissue complication probability models,
cal variables and two SNPs demonstrated that mean lung dose such as Lyman-Kutcher-Burman model (109), nearest-neighbor
was the strongest risk factor for RP among NSCLC patients (36). risk prediction model (110), relative seriality model (111) and
For late morbidities, the length of follow-up is a critical issue Logit-equivalent uniform dose model (111), have been proposed
because the frequency and severity of late reactions tend to to evaluate radiation-induced toxicities based on clinical and
progress over time (101). Additionally, the differences between dosimetric parameters. By incorporating SNP information into
clinical specimen sources (e.g. blood, tissue, plasma), the time the Lyman-Kutcher-Burman model based on mean lung dose,
of specimen collection (e.g. before, during, after treatment) and the predictive ability for severe radiation pneumonitis among
the assays used for analysis (e.g. genomics, transcriptomics, patients with NSCLC has been significantly improved (109).
proteomics) may also have subtle impact on outcomes (22). It is It is worth to note that a robust statistical method, the
essential to account for all dosimetric-, patient- and treatment- EMLasso technique, has been increasingly used for model
related factors that may predispose to increased late effects in selection in predictive radiogenetics. The EMLasso is a penal-
order to estimate the residual or unexplained toxicity likely due ized logistic regression method with a stochastic expectation–
to genetics. As individualized radiotherapy is increasingly used, maximization algorithm handling missing data and 10-fold
many centers have made great efforts to develop standardized cross-validation avoiding overfit (112). In light of this method, a
procedures for storing and evaluating treatment plans and clini- multicomponent prediction model involving clinical, treatment
cal outcomes. and genetic parameters for grade >2 acute esophagitis postra-
diotherapy toxicity in lung cancer patients with a sensitivity
The need for replication study of 84% and a specificity of 75.3% has been constructed by De
A very important aspect of any association study, whether it Ruyck et al. (113). Clinical usefulness assessment of the model
is derived from a candidate gene approach or whole genome demonstrated that 69.4% patients predicted to suffer from acute
analysis, is the need for replication studies validating the initial esophagitis will actually develop this toxicity and will benefit
findings. With a large (N = 1613), well-powered prospective study, from therapy modification. Following this, a number of predic-
Barnett et  al. (102) have failed to replicate previously reported tion models achieved by EMLasso have been developed recently,
314 | Carcinogenesis, 2015, Vol. 36, No. 3

for example, late genitourinary complaints predictive model for to fully exploit these new possibilities, international coopera-
prostate cancer (114), and dysphagia predictive model for HNC tion is essential. Several national and international cooperation
(115). genetic association studies have been initiated, such as Gene-
The development of EMLasso is appealing in constructing PARE (128), RadGenomics (129), RAPPER (130) and REQUITE pro-
predictive models as it selects models with the smallest number ject (131). The International Radiogenomics Consortium has
of parameters while preserving predictive value, which really been established in 2009 in order to foster large-scale collabora-
benefits clinical practice in consideration of time and cost effi- tive research projects (132). The future of the field is to create
ciency. Cross-validation is another fortunate property of the large patient cohorts for multiple cancer types, to validate the
EMlasso that avoids over/under-fitting. Besides, EMLasso is very genetic loci and build reliable predictive models.
suited for datasets with missing values which are common in
radiogenetics studies due to the use of high-throughput geno- Conclusion
typing techniques lacking a 100% call rate (115). Nevertheless,
validation and assessment of clinical usefulness are necessary The identified predictors of radiosensitivity are aimed to ulti-
before implementing these models in the clinic for routine use. mately contribute to an algorithm for guiding oncology treatment
Clinical usefulness can be quantified by decision analytic meth- decisions. Patients at high risk of developing radiation-induced

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


ods, such as net benefit and decision curve analysis (116,117). toxicities may be offered an alternative treatment approach, or,
for patients who have received radiotherapy, advanced planning
The biological mechanism of genotype–phenotype corrections can be introduced to better individualized radia-
associations tion treatment. In addition to predictive and prognostic testing,
the products of the identified genes could become targets for
The strong association between a genotype and a phenotype
innovative therapies in susceptible individuals. Although we
does not necessarily imply that the genetic variation is itself
are still at the foot of the mountain, we have faith to believe
the cause. The true variants affecting a given phenotype can be
that research on radiogenomics will help us achieve individual-
located within genes, or they can be located in regions that have
ized radiotherapy protocols and broadly personalized medicine,
not yet been assigned any functional elements. Thus, under-
leading to safer and more effective outcomes.
standing the biological mechanism is of utmost importance in
genotype–phenotype association studies.
Since expression levels can be regulated by genetic variants Funding
in regulatory elements, examining the gene expression patterns National Natural Science Fund (81273595); National High
after irradiation in vitro or in vivo with microarray to map the Technology Research and Development Program (2012AA02A518,
complex traits may shed new light on the pathways activated 2012AA02A517).
by ionizing radiation and help to identify novel candidate genes
(118,119). For example, by comparing the expression profiles of
radiation-resistant and radiation-sensitive NPC patient biopsy
Acknowledgement
specimens with DNA microarray, Yang et  al. (118) have dem- We thank F.He, M.Liu and Z.Luo for helpful comments on the
onstrated that 26 pathways (such as cell ion homeostasis, cell manuscript.
proliferation, receptor protein signaling, membrane system, Conflict of Interest Statement: None declared.
humoral immune response, as well as cytokines and inflamma-
tion) were probably linked to radiation resistance with biosta- References
tistics and bioinformatics analysis. Although most studies have
1. De Ruysscher, D. et al. (2012) Individualised isotoxic accelerated radio-
focused on mRNA profiles, proteomic studies will also be impor-
therapy and chemotherapy are associated with improved long-term
tant because mRNA expression is not always correlated with survival of patients with stage III NSCLC: a prospective population-
protein expression or posttranslational modification (120,121). based study. Radiother. Oncol., 102, 228–233.
Experiments based on animal models, gene knockout and small 2. Ghazali, N. et al. (2012) Genomic determinants of normal tissue toxicity
interfering RNAs techniques could provide interesting insights after radiotherapy for head and neck malignancy: a systematic review.
into functional candidates (122,123). Besides SNPs, structural Oral Oncol., 48, 1090–1100.
variations, such as insertions, deletions, inversions and copy 3. Parliament, M.B. et al. (2010) Single nucleotide polymorphisms of DNA
number variants, may also have important implications on repair genes as predictors of radioresponse. Semin. Radiat. Oncol., 20,
232–240.
radiosensitivity (92). Furthermore, epigenetic changes such as
4. West, C.M. et al. (2011) Genetics and genomics of radiotherapy toxicity:
DNA methylation, microRNA and histone modification, may
towards prediction. Genome Med., 3, 52.
represent an additional layer of complexity to understanding
5. Aziz, N.M. et  al. (2003) Trends and advances in cancer survivorship
cancer cell biology and response to radiation treatment by post- research: challenge and opportunity. Semin. Radiat. Oncol., 13, 248–
translational modifications (124–126). A recent study analyzing 266.
DNA methylation changes at >450 000 loci after ionizing radia- 6. Bentzen, S.M. et al. (1994) Patient-to-patient variability in the expres-
tion has identified that the most differentially methylated genes sion of radiation-induced normal tissue injury. Semin. Radiat. Oncol.,
were enriched in gene ontology categories relating to cell cycle, 4, 68–80.
DNA repair and apoptosis pathways, with a radiation dose and 7. Barnett, G.C. et  al. (2011) The impact of clinical factors on the devel-
postirradiation time-dependent manner (127). Evaluating the opment of late radiation toxicity: results from the Medical Research
Council RT01 trial (ISRCTN47772397). Clin. Oncol. (R. Coll. Radiol)., 23,
link among genetic variants, epigenetic modifications and dis-
613–624.
ease predispositions is currently a very active area in research.
8. Vogelius, I.R. et  al. (2012) A literature-based meta-analysis of clinical
risk factors for development of radiation induced pneumonitis. Acta
Large cohort, multicentered collaboration
Oncol., 51, 975–983.
Recent advances in technology and emerging insights into genetic 9. Qin, Q. et al. (2013) Clinical risk factors for late intestinal toxicity after
variations provide unprecedented opportunities. Nonetheless, radiotherapy: a systematic review protocol. Syst. Rev., 2, 39.
Guo et al. | 315

10. Andreassen, C.N. et  al. (2002) Does variability in normal tissue reac- 38. Alsbeih, G. et al. (2014) Among 45 variants in 11 genes, HDM2 promoter
tions after radiotherapy have a genetic basis–where and how to look polymorphisms emerge as new candidate biomarker associated with
for it? Radiother. Oncol., 64, 131–140. radiation toxicity. 3 Biotech., 4, 137–148.
11. Andreassen, C.N. (2005) Can risk of radiotherapy-induced normal tis- 39. Alsbeih, G. et al. (2010) Association between normal tissue complica-
sue complications be predicted from genetic profiles? Acta Oncol., 44, tions after radiotherapy and polymorphic variations in TGFB1 and
801–815. XRCC1 genes. Radiat. Res., 173, 505–511.
12. Barnett, G.C. et  al. (2009) Normal tissue reactions to radiotherapy:
40. Pratesi, N. et al. (2011) Association between single nucleotide polymor-
towards tailoring treatment dose by genotype. Nat. Rev. Cancer, 9, 134– phisms in the XRCC1 and RAD51 genes and clinical radiosensitivity in
142. head and neck cancer. Radiother. Oncol., 99, 356–361.
13. Becze, E. (2014) Radiogenomics offers promise of personalized radia- 41. Terrazzino, S. et al. (2012) Common variants of eNOS and XRCC1 genes
tion cancer therapy. ONS Connect, 29, 38–39. may predict acute skin toxicity in breast cancer patients receiving
14. Finnon, P. et al. (2008) Evidence for significant heritability of apoptotic radiotherapy after breast conserving surgery. Radiother. Oncol., 103,
and cell cycle responses to ionising radiation. Hum. Genet., 123, 485– 199–205.
493. 42. Yin, M. et  al. (2011) Polymorphisms of homologous recombination

15. Roberts, S.A. et  al. (1999) Heritability of cellular radiosensitivity: a
genes and clinical outcomes of non-small cell lung cancer patients
marker of low-penetrance predisposition genes in breast cancer? Am. treated with definitive radiotherapy. PLoS One, 6, e20055.

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


J. Hum. Genet., 65, 784–794. 43. Yin, M. et al. (2012) Genetic variants of the nonhomologous end joining
16. Gatti, R.A. (2001) The inherited basis of human radiosensitivity. Acta gene LIG4 and severe radiation pneumonitis in nonsmall cell lung can-
Oncol., 40, 702–711. cer patients treated with definitive radiotherapy. Cancer, 118, 528–535.
17. Proud, C. (2014) Radiogenomics: the promise of personalized treatment 44. Cheuk, I.W. et al. (2014) Association of XRCC1 and XRCC3 gene haplo-
in radiation oncology? Clin. J. Oncol. Nurs., 18, 185–189. types with the development of radiation-induced fibrosis in patients
18. Andreassen, C.N. (2014) The future has begun in radiogenomics! Radio- with nasopharyngeal carcinoma. Mol. Clin. Oncol., 2, 553–558.
ther. Oncol., 111, 165–167. 45. Mangoni, M. et al. (2011) Association between genetic polymorphisms
19. Rosenstein, B.S. et  al. (2014) Radiogenomics: radiobiology enters the in the XRCC1, XRCC3, XPD, GSTM1, GSTT1, MSH2, MLH1, MSH3, and
era of big data and team science. Int. J.  Radiat. Oncol. Biol. Phys., 89, MGMT genes and radiosensitivity in breast cancer patients. Int.
709–713. J. Radiat. Oncol. Biol. Phys., 81, 52–58.
20. Parliament, M.B. (2012) Radiogenomics: associations in all the wrong 46. Nogueira, A. et  al. (2012) Role of the RAD51 G172T polymorphism in
places? Lancet. Oncol., 13, 7–8. the clinical outcome of cervical cancer patients under concomitant
21. Ivanov, S.D. (2012) [Biochemical markers predicting response to radia- chemoradiotherapy. Gene, 504, 279–283.
tion- and radiochemo-therapy in cancer patients]. Biomed. Khim., 58, 47. Canman, C.E. et  al. (2012) Activation of the ATM kinase by ionizing
635–650. radiation and phosphorylation of p53. Science, 281, 1677–1679.
22. West, C.M. et al. (2007) The genomics revolution and radiotherapy. Clin. 48. Yang, M. et al. (2011) Association of P53 and ATM polymorphisms with
Oncol. (R. Coll. Radiol)., 19, 470–480. risk of radiation-induced pneumonitis in lung cancer patients treated
23. Kerns, S.L. et al. (2014) Radiogenomics: using genetics to identify can- with radiotherapy. Int. J. Radiat. Oncol. Biol. Phys., 79, 1402–1407.
cer patients at risk for development of adverse effects following radio- 49. Zhang, L. et  al. (2010) ATM polymorphisms are associated with risk
therapy. Cancer Discov., 4, 155–165. of radiation-induced pneumonitis. Int. J. Radiat. Oncol. Biol. Phys., 77,
24. Norman, A. et al. (1988) The importance of genetics for the optimiza- 1360–1368.
tion of radiation therapy. A hypothesis. Am. J. Clin. Oncol., 11, 84–88. 50. Xiong, H. et  al. (2013) ATM polymorphisms predict severe radiation
25. Bekiroglu, F. et  al. (2011) Adjuvant radiotherapy and health-related
pneumonitis in patients with non-small cell lung cancer treated with
quality of life of patients at intermediate risk of recurrence follow- definitive radiation therapy. Int. J. Radiat. Oncol. Biol. Phys., 85, 1066–
ing primary surgery for oral squamous cell carcinoma. Oral Oncol., 47, 1073.
967–973. 51. Cintra, H.S. et al. (2013) Investigation of genetic polymorphisms related
26. Lord, C.J. et al. (2012) The DNA damage response and cancer therapy. to the outcome of radiotherapy for prostate cancer patients. Dis. Mark-
Nature, 481, 287–294. ers, 35, 701–710.
27. Hosoya, N. et al. (2014) Targeting DNA damage response in cancer ther- 52. Raabe, A. et al. (2012) Association of single nucleotide polymorphisms
apy. Cancer Sci., 105, 370–388. in the genes ATM, GSTP1, SOD2, TGFB1, XPD and XRCC1 with risk of
28. Zhao, L. et  al. (2013) MicroRNA and signal transduction pathways in severe erythema after breast conserving radiotherapy. Radiat. Oncol.,
tumor radiation response. Cell. Signal., 25, 1625–1634. 7, 65.
29. Smith, J. et al. (2010) The ATM-Chk2 and ATR-Chk1 pathways in DNA 53. Silva, J. et  al. (2012) DNA repair system and prostate cancer progres-
damage signaling and cancer. Adv. Cancer Res., 108, 73–112. sion: the role of NBS1 polymorphism (rs1805794). DNA Cell Biol., 31,
30. Garrett, M.D. et  al. (2011) Anticancer therapy with checkpoint inhibi- 1182–1186.
tors: what, where and when? Trends Pharmacol. Sci., 32, 308–316. 54. Venkatesh, G.H. et al. (2014) Polymorphisms in radio-responsive genes
31. Furgason, J.M. et al. (2013) Targeting DNA repair mechanisms in cancer. and its association with acute toxicity among head and neck cancer
Pharmacol. Ther., 137, 298–308. patients. PLoS One, 9, e89079.
32. Mladenov, E. et  al. (2011) Induction and repair of DNA double strand 55. Popanda, O. et  al. (2006) Genetic polymorphisms in the DNA double-
breaks: the increasing spectrum of non-homologous end joining path- strand break repair genes XRCC3, XRCC2, and NBS1 are not associated
ways. Mutat. Res., 711, 61–72. with acute side effects of radiotherapy in breast cancer patients. Can-
33. Karar, J. et  al. (2009) Modulating the tumor microenvironment to
cer Epidemiol. Biomarkers Prev., 15, 1048–1050.
increase radiation responsiveness. Cancer Biol. Ther., 8, 1994–2001. 56. Sugiyama, T. et al. (2010) Thiol-metabolizing proteins and endothelial
34. Chen, B.P. et al. (2012) New insights into the roles of ATM and DNA-PKcs redox state: differential modulation of eNOS and biopterin pathways.
in the cellular response to oxidative stress. Cancer Lett., 327, 103–110. Am. J. Physiol. Heart Circ. Physiol., 298, H194–H201.
35. Chris, R. et al. (2013) Analysis of single nucleotide polymorphisms and 57. Edvardsen, H. et al. (2013) SNP in TXNRD2 associated with radiation-
radiation sensitivity of the lung assessed with an objective radiologic induced fibrosis: a study of genetic variation in reactive oxygen spe-
endpoint. Clin. Lung Cancer, 14, 267–274. cies metabolism and signaling. Int. J.  Radiat. Oncol. Biol. Phys., 86,
36. Yin, M. et al. (2011) Functional polymorphisms of base excision repair 791–799.
genes XRCC1 and APEX1 predict risk of radiation pneumonitis in 58. Falvo, E. et al. (2012) SNPs in DNA repair or oxidative stress genes and
patients with non-small cell lung cancer treated with definitive radia- late subcutaneous fibrosis in patients following single shot partial
tion therapy. Int. J. Radiat. Oncol. Biol. Phys., 81, e67–e73. breast irradiation. J. Exp. Clin. Cancer Res., 31, 7.
37. Alsbeih, G. et al. (2013) SNPs in genes implicated in radiation response 59. Terrazzino, S. et  al. (2012) Common variants of GSTP1, GSTA1, and
are associated with radiotoxicity and evoke roles as predictive and TGFβ1 are associated with the risk of radiation-induced fibrosis in
prognostic biomarkers. Radiat. Oncol., 8, 125. breast cancer patients. Int. J. Radiat. Oncol. Biol. Phys., 83, 504–511.
316 | Carcinogenesis, 2015, Vol. 36, No. 3

60. Nishioka, A. et al. (1999) An augmentation of Fas (CD95/APO-1) antigen 82. Fachal, L. et al. (2012) TGFβ1 SNPs and radio-induced toxicity in pros-
induced by radiation: flow cytometry analysis of lymphoma and leuke- tate cancer patients. Radiother. Oncol., 103, 206–209.
mia cell lines. Int. J. Mol. Med., 3, 275–278. 83. Altshuler, D. et  al. (2008) Genetic mapping in human disease. Sci-
61. Thurner, E.M. et al. (2014) Association of genetic variants in apoptosis ence, 322, 881–888.
genes FAS and FASL with radiation-induced late toxicity after prostate 84. Manolio, T.A. et al. (2009) Finding the missing heritability of complex
cancer radiotherapy. Strahlenther. Onkol., 190, 304–309. diseases. Nature, 461, 747–753.
62. Dumont, P. et al. (2003) The codon 72 polymorphic variants of p53 have 85. Cirulli, E.T. et al. (2010) Uncovering the roles of rare variants in com-
markedly different apoptotic potential. Nat. Genet., 33, 357–365. mon disease through whole-genome sequencing. Nat. Rev. Genet.,
63. Chang-Claude, J. et  al. (2009) Genetic polymorphisms in DNA repair 11, 415–425.
and damage response genes and late normal tissue complications of 86. Kerns, S.L. et  al. (2010) Genome-wide association study to identify
radiotherapy for breast cancer. Br. J. Cancer, 100, 1680–1686. single nucleotide polymorphisms (SNPs) associated with the devel-
64. Xie, X. et al. (2013) Expression of pAkt affects p53 codon 72 polymor- opment of erectile dysfunction in African-American men after
phism-based prediction of response to radiotherapy in nasopharyn- radiotherapy for prostate cancer. Int. J. Radiat. Oncol. Biol. Phys., 78,
geal carcinoma. Radiat. Oncol., 8, 117. 1292–1300.
65. Yin, J. et al. (2011) Common genetic variants in cell cycle pathway are 87. Kerns, S.L. et al. (2013) A 2-stage genome-wide association study to
associated with survival in stage III-IV non-small-cell lung cancer. Car- identify single nucleotide polymorphisms associated with develop-

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


cinogenesis, 32, 1867–1871. ment of erectile dysfunction following radiation therapy for prostate
66. Bond, G.L. et al. (2004) A single nucleotide polymorphism in the MDM2 cancer. Int. J. Radiat. Oncol. Biol. Phys., 85, e21–e28.
promoter attenuates the p53 tumor suppressor pathway and acceler- 88. Kerns, S.L. et al. (2013) Genome-wide association study identifies a
ates tumor formation in humans. Cell, 119, 591–602. region on chromosome 11q14.3 associated with late rectal bleeding
67. Yarnold, J. et al. (2010) Pathogenetic mechanisms in radiation fibrosis. following radiation therapy for prostate cancer. Radiother. Oncol.,
Radiother. Oncol., 97, 149–161. 107, 372–376.
68. Peters, C.A. et  al. (2008) TGFB1 single nucleotide polymorphisms are 89. Niu, N. et  al. (2010) Radiation pharmacogenomics: a genome-wide
associated with adverse quality of life in prostate cancer patients association approach to identify radiation response biomarkers
treated with radiotherapy. Int. J. Radiat. Oncol. Biol. Phys., 70, 752–759. using human lymphoblastoid cell lines. Genome Res., 20, 1482–1492.
69. Zhang, L. et al. (2010) Association of TGF-β1 and XPD polymorphisms 90. Barnett, G.C. et al. (2014) A genome wide association study (GWAS)
with severe acute radiation-induced esophageal toxicity in locally providing evidence of an association between common genetic vari-
advanced lung cancer patients treated with radiotherapy. Radiother. ants and late radiotherapy toxicity. Radiother. Oncol., 111, 178–185.
Oncol., 97, 19–25. 91. Edwards, S.L. et al. (2013) Beyond GWASs: illuminating the dark road
70.
Yuan, X. et  al. (2009) Single nucleotide polymorphism at from association to function. Am. J. Hum. Genet., 93, 779–797.
rs1982073:T869C of the TGFbeta 1 gene is associated with the risk of 92. Abbaszadeh, F. et al. (2010) A novel splice variant of the DNA-PKcs
radiation pneumonitis in patients with non-small-cell lung cancer gene is associated with clinical and cellular radiosensitivity in a
treated with definitive radiotherapy. J. Clin. Oncol., 27, 3370–3378. patient with xeroderma pigmentosum. J. Med. Genet., 47, 176–181.
71. Grainger, D.J. et al. (1999) Genetic control of the circulating concentra- 93. Andreassen, C.N. et al. (2009) Genetic variants and normal tissue tox-
tion of transforming growth factor type beta1. Hum. Mol. Genet., 8, icity after radiotherapy: a systematic review. Radiother. Oncol., 92,
93–97. 299–309.
72. Shah, R. et al. (2006) A molecular mechanism for the differential reg- 94. 1000 Genomes Project Consortium. et  al. (2010) A map of human
ulation of TGF-beta1 expression due to the common SNP -509C-T (c. genome variation from population-scale sequencing. Nature, 467,
-1347C > T). Hum. Genet., 120, 461–469. 1061–1073.
73. Kelsey, C.R. et al. (2012) A polymorphism within the promoter of the 95. Tennessen, J.A. et al. (2012) Evolution and functional impact of rare
TGFβ1 gene is associated with radiation sensitivity using an objec- coding variation from deep sequencing of human exomes. Science,
tive radiologic endpoint. Int. J.  Radiat. Oncol. Biol. Phys., 82, e247– 337, 64–69.
e255. 96. Tuzun, E. et  al. (2005) Fine-scale structural variation of the human
74. Xue, S.L. et al. (2013) Association between single nucleotide polymor- genome. Nat. Genet., 37, 727–732.
phisms of the transforming growth factor-β1 gene and overall survival 97. Manolio, T.A. (2013) Bringing genome-wide association findings into
in unresectable locally advanced non-small-cell lung cancer patients clinical use. Nat. Rev. Genet., 14, 549–558.
treated with radio(chemo)therapy in a Chinese population. Med. 98. Chung, C.C. et al. (2011) Current status of genome-wide association
Oncol., 30, 512. studies in cancer. Hum. Genet., 130, 59–78.
75. Guerra, J.L. et  al. (2012) Association between single nucleotide poly- 99. Barnett, G.C. et al. (2012) Standardized Total Average Toxicity score:
morphisms of the transforming growth factor β1 gene and the risk of a scale- and grade-independent measure of late radiotherapy toxic-
severe radiation esophagitis in patients with lung cancer. Radiother. ity to facilitate pooling of data from different studies. Int. J. Radiat.
Oncol., 105, 299–304. Oncol. Biol. Phys., 82, 1065–1074.
76. Niu, X. et al. (2012) A study of ethnic differences in TGFβ1 gene poly- 100. Barnett, G.C. et al. (2011) The Cambridge Breast Intensity-modulated
morphisms and effects on the risk of radiation pneumonitis in non- Radiotherapy Trial: patient- and treatment-related factors that influ-
small-cell lung cancer. J. Thorac. Oncol., 7, 1668–1675. ence late toxicity. Clin. Oncol. (R. Coll. Radiol)., 23, 662–673.
77. Wang, L. et al. (2010) TGF-beta1 gene polymorphisms for anticipating 101. Bentzen, S.M. et al. (1989) Latent-time estimation for late cutaneous
radiation-induced pneumonitis in non-small-cell lung cancer: differ- and subcutaneous radiation reactions in a single-follow-up clinical
ent ethnic association. J. Clin. Oncol., 28, e621–e622. study. Radiother. Oncol., 15, 267–274.
78. Reuther, S. et al. (2013) No effect of the transforming growth factor β1 102. Barnett, G.C. et al. (2012) Independent validation of genes and poly-
promoter polymorphism C-509T on TGFB1 gene expression, protein morphisms reported to be associated with radiation toxicity: a pro-
secretion, or cellular radiosensitivity. Int. J.  Radiat. Oncol. Biol. Phys., spective analysis study. Lancet. Oncol., 13, 65–77.
85, 460–465. 103. Talbot, C.J. et  al. (2012) A replicated association between polymor-
79. Martin, S. et al. (2010) Test of association between variant tgβ1 alleles phisms near TNFα and risk for adverse reactions to radiotherapy. Br.
and late adverse effects of breast radiotherapy. Radiother. Oncol., 97, J. Cancer, 107, 748–753.
15–18. 104. Kerns, S.L. et al. (2014) STROGAR - STrengthening the Reporting Of
80. Barnett, G.C. et al. (2012) Individual patient data meta-analysis shows Genetic Association studies in Radiogenomics. Radiother. Oncol.,
no association between the SNP rs1800469 in TGFB and late radiother- 110, 182–188.
apy toxicity. Radiother. Oncol., 105, 289–295. 105. Neale, B.M. et al. (2004) The future of association studies: gene-based
81. Barnett, G.C. et al. (2010) No association between SNPs regulating TGF- analysis and replication. Am. J. Hum. Genet., 75, 353–362.
β1 secretion and late radiotherapy toxicity to the breast: results from 106. Ioannidis, J.P. et al. (2001) Replication validity of genetic association
the RAPPER study. Radiother. Oncol., 97, 9–14. studies. Nat. Genet., 29, 306–309.
Guo et al. | 317

107. Vieland, V.J. (2001) The replication requirement. Nat. Genet., 29, 244– 120. Feng, X.P. et  al. (2010) Identification of biomarkers for predicting

245. nasopharyngeal carcinoma response to radiotherapy by proteomics.
108. Chanock, S.J. et  al. (2007) Replicating genotype-phenotype associa- Cancer Res., 70, 3450–3462.
tions. Nature, 447, 655–660. 121. Guo, Y. et  al. (2012) Identification of genes involved in radioresist-
109. Tucker, S.L. et  al. (2013) Incorporating single-nucleotide polymor-
ance of nasopharyngeal carcinoma by integrating gene ontology and
phisms into the Lyman model to improve prediction of radiation protein-protein interaction networks. Int. J. Oncol., 40, 85–92.
pneumonitis. Int. J. Radiat. Oncol. Biol. Phys., 85, 251–257. 122. Fabre, K.M. et al. (2011) Murine Prkdc polymorphisms impact DNA-
110. Saligan, L.N. et  al. (2014) Supervised classification by filter methods PKcs function. Radiat. Res., 175, 493–500.
and recursive feature elimination predicts risk of radiotherapy-related 123. Shang, Z.F. et al. (2010) Inactivation of DNA-dependent protein kinase
fatigue in patients with prostate cancer. Cancer Inform., 13, 141–152. leads to spindle disruption and mitotic catastrophe with attenuated
111. Bakhshandeh, M. et al. (2013) Normal tissue complication probability checkpoint protein 2 phosphorylation in response to DNA damage.
modeling of radiation-induced hypothyroidism after head-and-neck Cancer Res., 70, 3657–3666.
radiation therapy. Int. J. Radiat. Oncol. Biol. Phys., 85, 514–521. 1 24. Metheetrairut, C. et  al. (2013) MicroRNAs in the ionizing radia-
112. Sabbe, N. et al. (2013) EMLasso: logistic lasso with missing data. Stat. tion response and in radiotherapy. Curr. Opin. Genet. Dev., 23,
Med., 32, 3143–3157. 12–19.
113. De Ruyck, K. et al. (2011) Development of a multicomponent predic- 125. Yang, L.H. et al. (2013) Axin gene methylation status correlates with

Downloaded from https://academic.oup.com/carcin/article-abstract/36/3/307/315471 by guest on 27 May 2019


tion model for acute esophagitis in lung cancer patients receiving radiosensitivity of lung cancer cells. BMC Cancer, 13, 368.
chemoradiotherapy. Int. J. Radiat. Oncol. Biol. Phys., 81, 537–544. 126. Liang, L. et  al. (2014) MicroRNA-223 enhances radiation sensitivity
114. De Langhe, S. et  al. (2014) Integrated models for the prediction of of U87MG cells in vitro and in vivo by targeting ataxia telangiectasia
late genitourinary complaints after high-dose intensity modulated mutated. Int. J. Radiat. Oncol. Biol. Phys., 88, 955–960.
radiotherapy for prostate cancer: making informed decisions. Radio- 127. Antwih, D.A. et al. (2013) Radiation-induced epigenetic DNA meth-
ther. Oncol., 112, 95–99. ylation modification of radiation-response pathways. Epigenetics, 8,
115. De Ruyck, K. et al. (2013) A predictive model for dysphagia following 839–848.
IMRT for head and neck cancer: introduction of the EMLasso tech- 128. Ho, A.Y. et  al. (2006) Genetic predictors of adverse radiotherapy

nique. Radiother. Oncol., 107, 295–299. effects: the Gene-PARE project. Int. J.  Radiat. Oncol. Biol. Phys., 65,
116. Van Calster, B. et  al. (2013) Evaluation of markers and risk predic- 646–655.
tion models: overview of relationships between NRI and decision- 129. Iwakawa, M. et  al. (2002) [RadGenomics project]. Nihon Igaku

analytic measures. Med. Decis. Making, 33, 490–501. Hoshasen. Gakkai Zasshi., 62, 484–489.
117. Steyerberg, E.W. et al. (2010) Assessing the performance of prediction 130. Burnet, N.G. et al.; RAPPER Investigators. (2013) RAPPER: the radiog-
models: a framework for traditional and novel measures. Epidemiol- enomics of radiation toxicity. Clin. Oncol. (R. Coll. Radiol)., 25, 431–
ogy, 21, 128–138. 434.
118. Yang, S. et  al. (2012) Identification of prognostic biomarkers for
131. West, C. et al. (2014) The REQUITE Project: validating predictive mod-
response to radiotherapy by DNA microarray in nasopharyngeal car- els and biomarkers of radiotherapy toxicity to reduce side-effects
cinoma patients. Int. J. Oncol., 40, 1590–1600. and improve quality of life in cancer survivors. Clin. Oncol. (R. Coll.
119. Li, L. et  al. (2013) Identification of radioresistance-associated pro- Radiol)., 26, 739–742.
teins in human nasopharyngeal carcinoma cell lines by proteomic 132. West, C. et al. (2010) Establishment of a radiogenomics consortium.
analysis. Cancer Biother. Radiopharm., 28, 380–384. Radiother. Oncol., 94, 117–118.

Вам также может понравиться