肺癌放射治疗疗效及毒性相关的遗传免疫学标志物的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
肺癌的发病率和死亡率均逐年上升,虽然综合治疗在不断进步,其5年总生存率仍不足15%。临床上发现,病情相似的肺癌患者接受近乎相同的临床治疗后,其治疗疗效及副反应却差异较大,提示肺癌患者间存在较大的临床异质性。因此,区分不同的肺癌亚型而进行个体化治疗成为当今肺癌研究的热点问题之一。从而,发掘能够在治疗前评估预后、预测治疗疗效及毒性的生物标志物(Biomarker)成为当前肺癌个体化治疗研究的重要途径。
     胸部放射治疗是肺癌有效治疗手段之一,放射性肺损伤(Radiation-inducedlung injury, RILI)是肺癌胸部放疗最主要的并发症,限制了肺癌放疗的有效实施而影响放疗疗效。肺癌患者放疗后的总体预后及RILI的个体间差别较大。若在放疗前能够较准确评估肺癌患者的预后及疗效,预测重症RILI的发生,实施个体化的放疗方案,将有助于降低治疗相关死亡率,从而提高肺癌总生存。
     遗传学研究表明,单核苷酸多态性(Single nucleotide polymorphisms, SNPs)作为第三代遗传标记,代表着人类基因组中最丰富的遗传变异,其被作为候选的恶性肿瘤生物标志物而被研究多年。多个SNPs已被证实与肺癌的遗传易感性和/或治疗疗效及总体预后相关,其中DNA修复通路(DNArepair pathway)基因的SNPs作为肺癌生物标志物的研究报道最多。典型的范例是ERCC1基因多态性与肺癌的铂类化疗疗效相关。
     免疫学研究表明,外周血的肿瘤相关抗原(Tumor associate antigens, TAAs)/免疫调节分子(Immuno-regulatory molecules, IRMs)的自身抗体可在肿瘤形成实体肿块而被影像学检查发现前数月至数年即可被检测到。所以,TAAs/IRMs自身抗体作为肿瘤的生物标志物有其天然的优势。Early-CDT lung是第一个被研发出的诊断试剂盒用于肺癌的早期诊断。同时,许多研究也证实某些TAAs自身抗体与肺癌的预后及治疗疗效相关。
     在上述研究背景的基础上,本研究选择SNPs和TAAs/IRMs自身抗体作为候选的生物标志物。检测非小细胞肺癌(non-small cell lung cancer, NSCLC)患者个体间基因多态性及TAAs/IRMs自身抗体水平的差异,并与NSCLC胸部放疗后总生存及放射性肺炎做相关性分析。通过筛查阳性的生物标志物,尝试阐明NSCLC患者间的异质性。
     本研究结果显示:(1)遗传学方面:DNA修复基因的5个SNPs(ERCC2:rs238406、ERCC1:rs11615、ERCC1:rs3212948、XRCC4:rs9293329和XRCC4:rs2075685)与NSCLC患者总生存显著相关联。而且ERCC2:rs238406、ERCC1:rs11615与胸部放疗剂量对NSCLC患者总生存的影响存在交互作用。DNA修复基因的2个SNPs(XRCC4:rs1478486CC和XRCC4:rs2075685)与放射性肺炎(≥2级)显著相关联。(2)免疫学方面:在3个免疫调节分子及4个肿瘤相关抗原中未发现与NSCLC总生存相关的生物标志物。但发现CD25及OCT4II自身抗体水平与放射性肺炎(≥2级)显著相关联。
     本研究工作是NSCLC在免疫遗传学领域生物标志物研究的前沿课题,从遗传学和免疫学两个方面去筛查NSCLC胸部放疗后总生存及放射性肺炎相关的生物标志物。实验结果将有助于肺癌个体临床亚型的鉴别,为NSCLC个体化治疗临床策略的制定提供理论依据,最终将有助于提高NSCLC的总体临床疗效及总生存率。
Objectives
     Thoracic radiation therapy (TRT) is one of the most effective modalities innon-small cell lung cancer (NSCLC) management. However, the prognosis andradiation-induced lung injury (RILI) vary from patients to patients after TRT. Itindicates that one-size can not fit all in TRT. So, Personalized TRT based ondifferent subtypes of NSCLC has been proposed. Furthermore, biomarker researchhas been considered as the most important way for personalized TRT. This studytested the single nucleotide polymorphisms (SNPs) in lung cancer-related genes andthe levels of autoantibodies against tumor-associated antigens (TAAs)/immunologicregulated molecules (IRMs), and explored any associations between thesebiomarkers and overall survival (OS) and/or radiation pneumonitis (RP) in NSCLCpatients treated with definitive TRT. The aim of this study was to identify usefulbiomarkers for personalized TRT.
     Methods
     1. Genetic study (done at the University of Michigan, USA)
     This study included newly pathologically diagnosed and clinical stage I-IIINSCLC patients treated with definitive TRT. Bioinformatic methods and moleculargenetic techniques were used to test some SNPs in the TGFβ1gene and11DNArepair genes (ATM、ERCC1、ERCC2、ERCC5、MGMT、NBN、TP53、XRCC1、XRCC2、XRCC4、XPC). A total of30SNPs from these12genes were selected asgenetic markers. Matrix-assisted laser desorption/ionization time-of-flight massspectrometer (MALDI-TOF) was utilized to genotypes these SNPs in NSCLCpatients. Kaplan-Meire method, Cox proportional hazards model, receiver operating characteristic (ROC) curve analysis and genetic statistical software package(SPSS19.0and STREE TREE) were used to analyze the association between SNPsand OS/RP.
     2. Immunologic study (done at Jilin Univerisity)
     This study included newly pathologically diagnosed and clinical stage I-IIINSCLC patients treated with definitive TRT. A total of7IRMs and TAAs wereselected as antigens in this study, including CD25,FOXP3,TGF-β1,p16,P53,OCT4II and OCT4III. The bioinformatics database and software were used toanalyze their structures and characters. Based on in silico mapping of MHCrestricted epitopes derived from these proteins, linear antigen fragments were thendesigned; the linear peptide antigens were synthezied with a chemical method and anin-house ELISA test was developed to detect circulating autoantibodies in NSCLCpatients. Kaplan-Meire method, Cox proportional hazards model, logistic regressionmodel, ROC curve analysis and genetic statistical software package (SPSS19.0andSTREE TREE) were used to analyze the association between circulatingautoantibodies to TAAs/IRMs and OS/RP.
     Results
     1. Genetic analysis
     One hundred thirty-four patients who were enrolled in prospective trials withsurvival and lung toxicity data recorded prospectively were evaluated.8patientswere excluded with palliative dose (<60Gy).96patients (76%) underwentconcurrent chemo radiation therapy with the platinum-based regimen. The medianfollow-up time was45months, and the minimum follow-up time was12months.
     (1) Association between SNPs and OS of NSCLC
     In the univariate analysis of clinical characteristics, there were statisticallysignificant associations between OS and sex, tumor volume, KPS or TRT dose. Indetail, patients of female, tumor volume≤118cc, KPS≥80and TRT dose≥70Gywould survive longer. Multivariate Cox analysis in30SNPs showed that5SNPs(ERCC2:rs238406, ERCC1:rs11615, ERCC1:rs3212948, XRCC4:rs9293329,XRCC4:rs2075685) were significantly associated with OS of NSCLC. In detail, patients with genotype of ERCC2:rs238406GT/TT、 ERCC1:rs11615TT、ERCC1:rs3212948CC、XRCC4:rs9293329GA/AA and XRCC4:rs2075685TTwould survive longer. Furthermore, combined analysis of unfavorable genotypes(UFGS) from these5SNPs showed significantly cumulative effects on OS, i.e., thedeath hazard was increased with the numbers of UFGS increasing (0-5). TRT dosesubgroup analysis showed that the effect of SNPs tested on OS was observed only inthe TRT dose <70Gy group (P=0.015) rather than in the TRT dose≥70Gygroup (P=0.112). Interactive analysis by STREE TREE showed that there wasinteractive effect between TRT dose and ERCC2:rs238406or ERCC1:rs11615onOS. ERCC2:rs238406and ERCC1:rs11615could serve as genetic markers for TRTdose <70Gy group and TRT dose≥70Gy group, respectively. ROC analysisshowed that the area under ROC curve (AUC) was less than0.7when clinical factorswere used only. However, the AUC would be more than0.8when clinical factorsand5SNPs were combined together.
     (2) Association between SNPs and radiation pneumonitis
     In the univariate analysis of clinical characteristics, there were statisticallysignificant associations between RP and mean lung dose (MLD) and/or the lungvolume receiving more than20Gy (V20). In detail, patients of MLD≤16.3Gy orV20≤26%would be less likely to develop RP after TRT. Multivariate Cox analysisshowed that2out of30SNPs, XRCC4:rs1478486and XRCC4:rs2075685weresignificantly associated with RP. In detail, patients with genotypes ofXRCC4:rs1478486CC or XRCC4:rs2075685could have higher risk of RP after TRT.Furthermore, combined analysis of unfavorable genotypes (UFGS) from these2SNPs showed a significantly cumulative effect on RP, i.e. an increase in the RPhazard with the numbers of UFGS increasing (0-2). Interactive analysis showed thatthere was an interactive effect between lung dosimetric metrics (MLD/V20) andXRCC4:rs1478486CC or XRCC4:rs2075685on RP. ROC analysis showed that theAUC was0.66when MLD/V20was used only. However, the AUC was more than0.76when MLD/V20and2SNPs were combined together.
     2. Immunologic analysis
     Eighty-eight patients who were enrolled in prospective trials with survival andlung toxicity data recorded prospectively were evaluated,10of whom were excludeddue to palliative dose (<60Gy), and61(78%) underwent concurrent chemoradiation therapy with the platinum-based regimen. The median follow-up time was10.3months, and the minimum follow-up time was6months.(1) Association between circulating antibodies to TAAs/IRMs and OS of NSCLCIn the univariate analysis of clinical characteristics, there were statisticallysignificant associations between OS and age or chemotherapy. In detail, patientsaged≤60, who received systemic chemotherapy, could survive longer. MultivariateCox analysis showed that none of circulating autoantibodies to7TAAs/IRMs wassignificantly associated with OS of NSCLC (P>0.05)
     (2) Association between TAAs/IRMs and radiation pneumonitis
     In the univariate analysis of clinical characteristics, there was no clinicalvariable significantly associated with RP. Multivariate logistic analysis of antibodiesto7TAAs/IRMs showed that anti-CD25IgG and anti-OCT4II IgG weresignificantly associated with RP. In detail, patients with a low level of anti-CD25IgG or a high level of anti-OCT4II IgG could have a higher risk of RP after TRT.ROC analysis showed that the AUC was0.59when MLD/V20was used only.However, the AUC would be0.77when MLD/V20and2TAAs/IRMs werecombined together.
     Conclusion
     (1) Genetic variants in DNA repair genes are significantly associated with survivalof NSCLC patients treated with definitive TRT. Some SNPs from the DNA repairgenes can serve as biomarkers in predicting survival of NSCLC.
     (2) Higher dose TRT (≥70Gy) may improve survival in NSCLC patients withunfavorable genotypes of DNA repair genes. However, NSCLC patients carryingfavorable genotypes of DNA repair genes may not benifit from high dose TRT.
     (3) Genetic variants in DNA repair genes are significantly associated with radiationpneumonitis after TRT. Some SNPs from the DNA repair genes can serve asbiomarkers for prediction of radiation pneumonitis.
     (4) Circulating autoantibodies against CD25and OCT4II are significantlyassociated with radiation pneumonitis after TRT and can serve as biomarkers forprediction of radiation pneumonitis.
     (5) A novel genetic and immunological method can be set up based on present workto screen biomarkers for cancer.
引文
[1]郝捷,陈万青主编.中国肿瘤登记年报2012[M].北京:军事医学科学出版社2012.
    [2]蒋国梁主编.现代临床肿瘤学[M].上海:上海科学技术文献出版社2004.
    [3]Gatenby RA, Vincent TL. An evolutionary model of carcinogenesis. Cancer Res2003;63:6212-6220.
    [4] Woodard KM, Chapman CJ. Lung cancer-can autoantibodies provide an aid todiagnosis? Expert Opin Med Diagn2008;2:911-923.
    [5] Travis WD, Brambilla E, Riely GJ. New pathologic classification of lung cancer:relevance for clinical practice and clinical trials. J Clin Oncol2013;31:992-1001.
    [6] Tsim S, O'Dowd CA, Milroy R et al. Staging of non-small cell lung cancer(NSCLC): a review. Respir Med2010;104:1767-1774.
    [7] Ou SH, Zell JA, Ziogas A et al. Prognostic factors for survival of stage Inonsmall cell lung cancer patients: a population-based analysis of19,702stage Ipatients in the California Cancer Registry from1989to2003. Cancer2007;110:1532-1541.
    [8] Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies incancer treatment. Nat Rev Cancer2012;12:237-251.
    [9] Siegel R, Naishadham D, Jemal A. Cancer statistics,2012. CA Cancer J Clin2012;62:10-29.
    [10] Willers H, Azzoli CG, Santivasi WL et al. Basic mechanisms of therapeuticresistance to radiation and chemotherapy in lung cancer. Cancer J2013;19:200-207.
    [11] Herbst RS, Heymach JV, Lippman SM. Lung cancer. N Engl J Med2008;359:1367-1380.
    [12] Scheff RJ, Schneider BJ. Non-Small-Cell Lung Cancer: Treatment of LateStage Disease: Chemotherapeutics and New Frontiers. Semin Intervent Radiol2013;30:191-198.
    [13] Perez-Soler R. Individualized therapy in non-small-cell lung cancer: futureversus current clinical practice. Oncogene2009;28Suppl1: S38-45.
    [14] Miller VA, Riely GJ, Zakowski MF et al. Molecular characteristics ofbronchioloalveolar carcinoma and adenocarcinoma, bronchioloalveolar carcinomasubtype, predict response to erlotinib. J Clin Oncol2008;26:1472-1478.
    [15] Tsao MS, Sakurada A, Cutz JC et al. Erlotinib in lung cancer-molecular andclinical predictors of outcome. N Engl J Med2005;353:133-144.
    [16] Sequist LV, Martins RG, Spigel D et al. First-line gefitinib in patients withadvanced non-small-cell lung cancer harboring somatic EGFR mutations. J ClinOncol2008;26:2442-2449.
    [17] Bepler G, Kusmartseva I, Sharma S et al. RRM1modulated in vitro and in vivoefficacy of gemcitabine and platinum in non-small-cell lung cancer. J Clin Oncol2006;24:4731-4737.
    [18] Olaussen KA, Dunant A, Fouret P et al. DNA repair by ERCC1innon-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N Engl JMed2006;355:983-991.
    [19] Simon GR, Sharma S, Cantor A et al. ERCC1expression is a predictor ofsurvival in resected patients with non-small cell lung cancer. Chest2005;127:978-983.
    [20] Tsao MS, Aviel-Ronen S, Ding K et al. Prognostic and predictive importance ofp53and RAS for adjuvant chemotherapy in non small-cell lung cancer. J Clin Oncol2007;25:5240-5247.
    [21] Zheng Z, Chen T, Li X et al. DNA synthesis and repair genes RRM1andERCC1in lung cancer. N Engl J Med2007;356:800-808.
    [22] Bepler G, Sharma S, Cantor A et al. RRM1and PTEN as prognostic parametersfor overall and disease-free survival in patients with non-small-cell lung cancer. JClin Oncol2004;22:1878-1885.
    [23] Shaw AT, Yeap BY, Mino-Kenudson M et al. Clinical features and outcome ofpatients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol2009;27:4247-4253.
    [24] Rodig SJ, Shapiro GI. Crizotinib, a small-molecule dual inhibitor of the c-Metand ALK receptor tyrosine kinases. Curr Opin Investig Drugs2010;11:1477-1490.
    [25] Mino-Kenudson M, Chirieac LR, Law K et al. A novel, highly sensitiveantibody allows for the routine detection of ALK-rearranged lung adenocarcinomasby standard immunohistochemistry. Clin Cancer Res2010;16:1561-1571.
    [26] Rodig SJ, Mino-Kenudson M, Dacic S et al. Unique clinicopathologic featurescharacterize ALK-rearranged lung adenocarcinoma in the western population. ClinCancer Res2009;15:5216-5223.
    [27] Takahashi T, Sonobe M, Kobayashi M et al. Clinicopathologic features ofnon-small-cell lung cancer with EML4-ALK fusion gene. Ann Surg Oncol2010;17:889-897.
    [28] Furuse K, Fukuoka M, Kawahara M et al. Phase III study of concurrent versussequential thoracic radiotherapy in combination with mitomycin, vindesine, andcisplatin in unresectable stage III non-small-cell lung cancer. J Clin Oncol1999;17:2692-2699.
    [29] Kong FM, Lally BE, Chang JY et al. ACR Appropriateness Criteria(R)radiation therapy for small-cell lung cancer. Am J Clin Oncol2013;36:206-213.
    [30] Decker RH, Langer CJ, Rosenzweig KE et al. ACR Appropriateness Criteria(R)Postoperative Adjuvant Therapy in Non-Small Cell Lung Cancer. Am J Clin Oncol2011;34:537-544.
    [31] Gopal RS, Dubey S, Rosenzweig KE et al. ACR Appropriateness Criteria(R) onInduction and Adjuvant Therapy for Stage N2Non-Small-Cell Lung Cancer: expertpanel on radiation oncology-lung. Int J Radiat Oncol Biol Phys2010;78:969-974.
    [32] Gewanter RM, Rosenzweig KE, Chang JY et al. ACR Appropriateness Criteria:nonsurgical treatment for non-small-cell lung cancer: good performancestatus/definitive intent. Curr Probl Cancer2010;34:228-249.
    [33] Rosenzweig KE, Movsas B, Bradley J et al. ACR appropriateness criteria onnonsurgical treatment for non-small-cell lung cancer: poor performance status orpalliative intent. J Am Coll Radiol2009;6:85-95.
    [34] Timmerman R, Paulus R, Galvin J et al. Stereotactic body radiation therapy forinoperable early stage lung cancer. JAMA2010;303:1070-1076.
    [35] Baumann P, Nyman J, Hoyer M et al. Outcome in a prospective phase II trial ofmedically inoperable stage I non-small-cell lung cancer patients treated withstereotactic body radiotherapy. J Clin Oncol2009;27:3290-3296.
    [36] Bradley JD, El Naqa I, Drzymala RE et al. Stereotactic body radiation therapyfor early-stage non-small-cell lung cancer: the pattern of failure is distant. Int JRadiat Oncol Biol Phys2010;77:1146-1150.
    [37] Grutters JP, Kessels AG, Pijls-Johannesma M et al. Comparison of theeffectiveness of radiotherapy with photons, protons and carbon-ions for non-smallcell lung cancer: a meta-analysis. Radiother Oncol2010;95:32-40.
    [38] Goodman N. Biological data becomes computer literate: new advances inbioinformatics. Curr Opin Biotechnol2002;13:68-71.
    [39] Zhao LP, Aragaki C, Hsu L et al. Mapping of complex traits bysingle-nucleotide polymorphisms. Am J Hum Genet1998;63:225-240.
    [40] Thangadurai S. The Human Genome Project: the role of analytical chemists.Anal Sci2004;20:595-601.
    [41] Karp PD, Paley S, Romero P. The Pathway Tools software. Bioinformatics2002;18Suppl1: S225-232.
    [42] Kiechle FL, Zhang X. The postgenomic era: implications for the clinicallaboratory. Arch Pathol Lab Med2002;126:255-262.
    [43] Chanda SK, Caldwell JS. Fulfilling the promise: drug discovery in thepost-genomic era. Drug Discov Today2003;8:168-174.
    [44] Thorisson GA, Muilu J, Brookes AJ. Genotype-phenotype databases: challengesand solutions for the post-genomic era. Nat Rev Genet2009;10:9-18.
    [45] Papin JA, Price ND, Wiback SJ et al. Metabolic pathways in the post-genomeera. Trends Biochem Sci2003;28:250-258.
    [46] Collins FS, Patrinos A, Jordan E et al. New goals for the U.S. Human GenomeProject:1998-2003. Science1998;282:682-689.
    [47] Gabriel SB, Schaffner SF, Nguyen H et al. The structure of haplotype blocks inthe human genome. Science2002;296:2225-2229.
    [48] Shen GQ, Abdullah KG, Wang QK. The TaqMan method for SNP genotyping.Methods Mol Biol2009;578:293-306.
    [49] Petrosino JF, Highlander S, Luna RA et al. Metagenomic pyrosequencing andmicrobial identification. Clin Chem2009;55:856-866.
    [50] Yu B, Sawyer NA, Chiu C et al. DNA mutation detection using denaturinghigh-performance liquid chromatography (DHPLC). Curr Protoc Hum Genet2006;Chapter7: Unit710.
    [51] Ghatak S, Muthukumaran RB, Nachimuthu SK. A simple method of genomicDNA extraction from human samples for PCR-RFLP analysis. J Biomol Tech2013;24:224-231.
    [52] Kapuscinski J. DAPI: a DNA-specific fluorescent probe. Biotech Histochem1995;70:220-233.
    [53] Bizzini A, Greub G. Matrix-assisted laser desorption ionization time-of-flightmass spectrometry, a revolution in clinical microbial identification. Clin MicrobiolInfect2010;16:1614-1619.
    [54] Delahunty C, Ankener W, Deng Q et al. Testing the feasibility of DNA typingfor human identification by PCR and an oligonucleotide ligation assay. Am J HumGenet1996;58:1239-1246.
    [55] Brown PO, Hartwell L. Genomics and human disease--variations on variation.Nat Genet1998;18:91-93.
    [56] Housman D, Ledley FD. Why pharmacogenomics? Why now? Nat Biotechnol1998;16:492-493.
    [57] Xu TP, Shen H, Liu LX et al. Association of ERCC1-C118T and-C8092Apolymorphisms with lung cancer risk and survival of advanced-stage non-small celllung cancer patients receiving platinum-based chemotherapy: A pooled analysisbased on39reports. Gene2013.
    [58] McNeil EM, Melton DW. DNA repair endonuclease ERCC1-XPF as a noveltherapeutic target to overcome chemoresistance in cancer therapy. Nucleic Acids Res2012.
    [59] Wei HB, Hu J, Shang LH et al. A meta-analytic review of ERCC1/MDR1polymorphism and chemosensitivity to platinum in patients with advanced non-smallcell lung cancer. Chin Med J (Engl)2012;125:2902-2907.
    [60] Durrington HJ. Is ERCC1a reliable prognostic protein biomarker innon-small-cell lung cancer? Thorax2013.
    [61] Yan D, Wei P, An G et al. Prognostic potential of ERCC1protein expressionand clinicopathologic factors in stage III/N2non-small cell lung cancer. JCardiothorac Surg2013;8:149.
    [62] Maus MK, Mack PC, Astrow SH et al. Histology-related associations ofERCC1, RRM1, and TS biomarkers in patients with non-small-cell lung cancer:implications for therapy. J Thorac Oncol2013;8:582-586.
    [63] Bepler G, Williams C, Schell MJ et al. Randomized International Phase III Trialof ERCC1and RRM1Expression-Based Chemotherapy VersusGemcitabine/Carboplatin in Advanced Non-Small-Cell Lung Cancer. J Clin Oncol2013;31:2404-2412.
    [64] Bai ZL, Wang YY, Zhe H et al. ERCC1mRNA levels can predict the responseto cisplatin-based concurrent chemoradiotherapy of locally advanced cervicalsquamous cell carcinoma. Radiat Oncol2012;7:221.
    [65] Su Y, Orelli B, Madireddy A et al. Multiple DNA binding domains mediate thefunction of the ERCC1-XPF protein in nucleotide excision repair. J Biol Chem2012;287:21846-21855.
    [66] Friboulet L, Olaussen KA, Pignon JP et al. ERCC1isoform expression andDNA repair in non-small-cell lung cancer. N Engl J Med2013;368:1101-1110.
    [67] Besse B, Olaussen KA, Soria JC. ERCC1and RRM1: ready for prime time? JClin Oncol2013;31:1050-1060.
    [68] Kalikaki A, Kanaki M, Vassalou H et al. DNA repair gene polymorphismspredict favorable clinical outcome in advanced non-small-cell lung cancer. ClinLung Cancer2009;10:118-123.
    [69] Liao WY, Shih JY, Chang GC et al. Genetic polymorphism of XRCC1Arg399Gln is associated with survival in non-small-cell lung cancer patients treatedwith gemcitabine/platinum. J Thorac Oncol2012;7:973-981.
    [70] Yoon SM, Hong YC, Park HJ et al. The polymorphism and haplotypes ofXRCC1and survival of non-small-cell lung cancer after radiotherapy. Int J RadiatOncol Biol Phys2005;63:885-891.
    [71] Li Y, Huang XE, Jin GF et al. Lack of any relationship between chemotherapytoxicity in non-small cell lung cancer cases and polymorphisms in XRCC1codon399or XPD codon751. Asian Pac J Cancer Prev2011;12:739-742.
    [72] Ryu JS, Hong YC, Han HS et al. Association between polymorphisms ofERCC1and XPD and survival in non-small-cell lung cancer patients treated withcisplatin combination chemotherapy. Lung Cancer2004;44:311-316.
    [73] Wu W, Li H, Wang H et al. Effect of polymorphisms in XPD on clinicaloutcomes of platinum-based chemotherapy for Chinese non-small cell lung cancerpatients. PLoS One2012;7: e33200.
    [74] Wu W, Zhang W, Qiao R et al. Association of XPD polymorphisms with severetoxicity in non-small cell lung cancer patients in a Chinese population. Clin CancerRes2009;15:3889-3895.
    [75] Kim SH, Lee GW, Lee MJ et al. Clinical significance of ERCC2haplotype-tagging single nucleotide polymorphisms in patients with unresectablenon-small cell lung cancer treated with first-line platinum-based chemotherapy.Lung Cancer2012.
    [76] Wu X, Ye Y, Rosell R et al. Genome-wide association study of survival innon-small cell lung cancer patients receiving platinum-based chemotherapy. J NatlCancer Inst2011;103:817-825.
    [77] Andreassen CN, Alsner J, Overgaard M et al. Prediction of normal tissueradiosensitivity from polymorphisms in candidate genes. Radiother Oncol2003;69:127-135.
    [78] Andreassen CN, Alsner J, Overgaard M et al. Risk of radiation-inducedsubcutaneous fibrosis in relation to single nucleotide polymorphisms in TGFB1,SOD2, XRCC1, XRCC3, APEX and ATM--a study based on DNA from formalinfixed paraffin embedded tissue samples. Int J Radiat Biol2006;82:577-586.
    [79] Alsbeih G, Al-Harbi N, Al-Hadyan K et al. Association between normal tissuecomplications after radiotherapy and polymorphic variations in TGFB1and XRCC1genes. Radiat Res2010;173:505-511.
    [80] Talbot CJ, Tanteles GA, Barnett GC et al. A replicated association betweenpolymorphisms near TNFalpha and risk for adverse reactions to radiotherapy. Br JCancer2012;107:748-753.
    [81] Yuan X, Liao Z, Liu Z et al. Single nucleotide polymorphism atrs1982073:T869C of the TGFbeta1gene is associated with the risk of radiationpneumonitis in patients with non-small-cell lung cancer treated with definitiveradiotherapy. J Clin Oncol2009;27:3370-3378.
    [82] Wang L, Bi N. TGF-beta1gene polymorphisms for anticipatingradiation-induced pneumonitis in non-small-cell lung cancer: different ethnicassociation. J Clin Oncol2010;28: e621-622.
    [83] Kerns SL, Ostrer H, Stock R et al. Genome-wide association study to identifysingle nucleotide polymorphisms (SNPs) associated with the development of erectiledysfunction in African-American men after radiotherapy for prostate cancer. Int JRadiat Oncol Biol Phys2010;78:1292-1300.
    [84] Barnett GC, Coles CE, Elliott RM et al. Independent validation of genes andpolymorphisms reported to be associated with radiation toxicity: a prospectiveanalysis study. Lancet Oncol2012;13:65-77.
    [85] Zhang L, Yang M, Bi N et al. Association of TGF-beta1and XPDpolymorphisms with severe acute radiation-induced esophageal toxicity in locallyadvanced lung cancer patients treated with radiotherapy. Radiother Oncol2010;97:19-25.
    [86] Yang M, Zhang L, Bi N et al. Association of P53and ATM PolymorphismsWith Risk of Radiation-Induced Pneumonitis in Lung Cancer Patients Treated WithRadiotherapy. Int J Radiat Oncol Biol Phys2011;79:1402-1407.
    [87] Yin M, Liao Z, Liu Z et al. Functional Polymorphisms of Base Excision RepairGenes XRCC1and APEX1Predict Risk of Radiation Pneumonitis in Patients WithNon-Small Cell Lung Cancer Treated With Definitive Radiation Therapy. Int JRadiat Oncol Biol Phys2011;81: e67-73.
    [88] Yin M, Liao Z, Huang YJ et al. Polymorphisms of homologous recombinationgenes and clinical outcomes of non-small cell lung cancer patients treated withdefinitive radiotherapy. PLoS One2011;6: e20055.
    [89] Yin M, Liao Z, Liu Z et al. Genetic variants of the nonhomologous end joininggene LIG4and severe radiation pneumonitis in nonsmall cell lung cancer patientstreated with definitive radiotherapy. Cancer2012;118:528-535.
    [90] Kelsey CR, Jackson L, Langdon S et al. A Polymorphism Within the Promoterof the TGFbeta1Gene Is Associated With Radiation Sensitivity Using an ObjectiveRadiologic Endpoint. Int J Radiat Oncol Biol Phys2011.
    [91] Hildebrandt MA, Komaki R, Liao Z et al. Genetic variants ininflammation-related genes are associated with radiation-induced toxicity followingtreatment for non-small cell lung cancer. PLoS One2010;5: e12402.
    [92] Mak RH, Alexander BM, Asomaning K et al. A single-nucleotidepolymorphism in the methylene tetrahydrofolate reductase (MTHFR) gene isassociated with risk of radiation pneumonitis in lung cancer patients treated withthoracic radiation therapy. Cancer2012;118:3654-3665.
    [93] Yin M, Liao Z, Yuan X et al. Polymorphisms of the vascular endothelial growthfactor gene and severe radiation pneumonitis in non-small cell lung cancer patientstreated with definitive radiotherapy. Cancer Sci2012;103:945-950.
    [94] Van der Bruggen P, Traversari C, Chomez P et al. A gene encoding an antigenrecognized by cytolytic T lymphocytes on a human melanoma. J Immunol2007;178:2617-2621.
    [95]杨镇主编.肿瘤免疫学[M].武汉:湖北科学技术出版社,1998:1.
    [96] Dumitriu IE, Dunbar DR, Howie SE et al. Human dendritic cells produceTGF-beta1under the influence of lung carcinoma cells and prime the differentiationof CD4+CD25+Foxp3+regulatory T cells. J Immunol2009;182:2795-2807.
    [97] Muller AJ, Scherle PA. Targeting the mechanisms of tumoral immune tolerancewith small-molecule inhibitors. Nat Rev Cancer2006;6:613-625.
    [98] Van den Eynde BJ, van der Bruggen P. T cell defined tumor antigens. CurrOpin Immunol1997;9:684-693.
    [99] Liu W, Peng B, Lu Y et al. Autoantibodies to tumor-associated antigens asbiomarkers in cancer immunodiagnosis. Autoimmun Rev2011;10:331-335.
    [100] Sahin U, Tureci O, Schmitt H et al. Human neoplasms elicit multiple specificimmune responses in the autologous host. Proc Natl Acad Sci U S A1995;92:11810-11813.
    [101] Tong YQ, Zhang ZJ, Liu B et al. Autoantibodies as potential biomarkers fornasopharyngeal carcinoma. Proteomics2008;8:3185-3193.
    [102] Hudson ME, Pozdnyakova I, Haines K et al. Identification of differentiallyexpressed proteins in ovarian cancer using high-density protein microarrays. ProcNatl Acad Sci U S A2007;104:17494-17499.
    [103] Bui JD, Schreiber RD. Cancer immunosurveillance, immunoediting andinflammation: independent or interdependent processes? Curr Opin Immunol2007;19:203-208.
    [104] Li Y, Karjalainen A, Koskinen H et al. p53autoantibodies predict subsequentdevelopment of cancer. Int J Cancer2005;114:157-160.
    [105] Zhong L, Coe SP, Stromberg AJ et al. Profiling tumor-associated antibodiesfor early detection of non-small cell lung cancer. J Thorac Oncol2006;1:513-519.
    [106]何维主编.医学免疫学[M].8年制第2版,北京:人民卫生出版社,2010:257-280..
    [107]朱道银,吴玉章主编.免疫学实验[M].北京:科学出版社,2008,140-160.
    [108] Ralhan R, Nath N, Agarwal S et al. Circulating p53antibodies as earlymarkers of oral cancer: correlation with p53alterations. Clin Cancer Res1998;4:2147-2152.
    [109] Lubin R, Zalcman G, Bouchet L et al. Serum p53antibodies as early markersof lung cancer. Nat Med1995;1:701-702.
    [110] Kobold S, Lutkens T, Cao Y et al. Autoantibodies against tumor-relatedantigens: incidence and biologic significance. Hum Immunol2010;71:643-651.
    [111] Lam S, Boyle P, Healey GF et al. EarlyCDT-Lung: an immunobiomarker testas an aid to early detection of lung cancer. Cancer Prev Res (Phila)2011;4:1126-1134.
    [112] Chapman CJ, Healey GF, Murray A et al. EarlyCDT(R)-Lung test: improvedclinical utility through additional autoantibody assays. Tumour Biol2012;33:1319-1326.
    [113] Jett JR, Peek LJ, Fredericks L et al. Audit of the autoantibody test,EarlyCDT(R)-lung, in1600patients: an evaluation of its performance in routineclinical practice. Lung Cancer2014;83:51-55.
    [114] Wang W, Guan S, Sun S et al. Detection of circulating antibodies to linearpeptide antigens derived from ANXA1and DDX53in lung cancer. Tumour Biol2014.
    [115] Zhang C, Ye L, Guan S et al. Autoantibodies against p16protein-derivedpeptides may be a potential biomarker for non-small cell lung cancer. Tumour Biol2013.
    [116] Ye L, Li X, Sun S et al. A study of circulating anti-CD25antibodies innon-small cell lung cancer. Clin Transl Oncol2012.
    [117] Liu L, Liu N, Liu B et al. Are circulating autoantibodies to ABCC3transportera potential biomarker for lung cancer? J Cancer Res Clin Oncol2012;138:1737-1742.
    [118] Wang W, Ye L, Li X et al. Circulating IgG antibody against FOXP3may be apotential biomarker for lung cancer. Advances in Lung Cancer2013;2:79.
    [119]关松磊.肺癌和食管癌外周血免疫调节因子及肿瘤相关抗原的自身抗体研究. In.吉林大学2013.
    [120] Hirasawa Y, Kohno N, Yokoyama A et al. Natural autoantibody to MUC1is aprognostic indicator for non-small cell lung cancer. Am J Respir Crit Care Med2000;161:589-594.
    [121] Vural B, Chen LC, Saip P et al. Frequency of SOX Group B (SOX1,2,3) andZIC2antibodies in Turkish patients with small cell lung carcinoma and theircorrelation with clinical parameters. Cancer2005;103:2575-2583.
    [122] Kobold S, Luetkens T, Cao Y et al. Prognostic and diagnostic value ofspontaneous tumor-related antibodies. Clin Dev Immunol2010;2010:721531.
    [123] Zhang L, Yang M, Bi N et al. ATM polymorphisms are associated with risk ofradiation-induced pneumonitis. Int J Radiat Oncol Biol Phys2010;77:1360-1368.
    [124] Tsoutsou PG, Koukourakis MI. Radiation pneumonitis and fibrosis:mechanisms underlying its pathogenesis and implications for future research. Int JRadiat Oncol Biol Phys2006;66:1281-1293.
    [125] Hanna N, Neubauer M, Yiannoutsos C et al. Phase III study of cisplatin,etoposide, and concurrent chest radiation with or without consolidation docetaxel inpatients with inoperable stage III non-small-cell lung cancer: the Hoosier OncologyGroup and U.S. Oncology. J Clin Oncol2008;26:5755-5760.
    [126] Rosenzweig KE, Amols H, Ling CC. New radiotherapy technologies. SeminSurg Oncol2003;21:190-195.
    [127] Marks LB, Bentzen SM, Deasy JO et al. Radiation dose-volume effects in thelung. Int J Radiat Oncol Biol Phys2010;76: S70-76.
    [128] Schallenkamp JM, Miller RC, Brinkmann DH et al. Incidence of radiationpneumonitis after thoracic irradiation: Dose-volume correlates. Int J Radiat OncolBiol Phys2007;67:410-416.
    [129] The International HapMap Project. Nature2003;426:789-796.
    [130] De Silva IU, McHugh PJ, Clingen PH, Hartley JA. Defining the roles ofnucleotide excision repair and recombination in the repair of DNA interstrandcross-links in mammalian cells. Mol Cell Biol2000;20:7980-7990.
    [131] Reynolds C, Obasaju C, Schell MJ et al. Randomized phase III trial ofgemcitabine-based chemotherapy with in situ RRM1and ERCC1protein levels forresponse prediction in non-small-cell lung cancer. J Clin Oncol2009;27:5808-5815.
    [132] Rosell R, Taron M, Camps C, Lopez-Vivanco G. Influence of genetic markerson survival in non-small cell lung cancer. Drugs Today (Barc)2003;39:775-786.
    [133] Tibaldi C, Giovannetti E, Vasile E et al. Correlation of CDA, ERCC1, andXPD polymorphisms with response and survival in gemcitabine/cisplatin-treatedadvanced non-small cell lung cancer patients. Clin Cancer Res2008;14:1797-1803.
    [134] Wei HB, Lu XS, Shang LH et al. Polymorphisms of ERCC1C118T/C8092Aand MDR1C3435T predict outcome of platinum-based chemotherapies in advancednon-small cell lung cancer: a meta-analysis. Arch Med Res2011;42:412-420.
    [135] Zhang C, Ye L, Guan S et al. Autoantibodies against p16protein-derivedpeptides may be a potential biomarker for non-small cell lung cancer. TumorBiology2013;1-5.