循环肿瘤DNA在弥漫大B细胞淋巴瘤中的研究进展

循环肿瘤DNA在弥漫大B细胞淋巴瘤中的研究进展

李苗苗,宋玉琴,朱军(北京大学肿瘤医院暨北京市肿瘤防治研究所 淋巴肿瘤内科 恶性肿瘤发病机制及转化研究教育部重点实验室,北京 100142)

【摘要】 循环肿瘤DNA(circulating tumor DNA,ctDNA)检测作为液体活检的一种,具有非侵入性和克服肿瘤异质性的特点,已被证实在多种实体肿瘤中可以辅助诊断、评估预后、监测疾病复发等。弥漫大B细胞淋巴瘤(diffuse large B cell lymphoma,DLBCL)是最常见的非霍奇金淋巴瘤,其诊断和预后评估目前主要依赖于组织活检和影像学检查,而ctDNA检测可以真实反映基因突变图谱,并且可以反复取材进行动态监测,在辅助诊断、基因分型、评估预后、评价疗效、监测微小残留病变及预测复发等方面均具有重要作用。随着检测技术的改进和提升,研究ctDNA有助于改善DLBCL患者的预后。本文就ctDNA在DLBCL中的研究进展做一综述。

【关键词】 液体活检;循环肿瘤DNA;弥漫大B细胞淋巴瘤

弥漫大B细胞淋巴瘤(diffuse large B cell lym­phoma,DLBCL)占非霍奇金淋巴瘤的30%~40%,

按照基因分型可分为活化的B细胞样型(activated B cell like,ABC)、生发中心B细胞样型(germinal center B cell like,GCB)和第三型[1]。目前,DLBCL的标准一线化疗方案是R-CHOP联合化疗,主要药物是利妥昔单抗、环磷酰胺、表柔比星、长春新碱和泼尼松。75%~80%的患者在接受一线治疗后可达到完全缓解,但仍有约40%的患者复发/难治[2,3]。其诊断和预后评估目前主要依赖于影像学检查和组织活检,在没有病变的情况下,无法获取活组织标本来追踪疾病的发展过程,而影像学检查也存在一定的假阳性率和假阴性率,且具有辐射暴露风险。目前发现的用于预测DLBCL患者预后的指标,如国际预后指数(international prognostic index,IPI)、起源细胞分类、免疫组织化学、Myc基因重排,可在一定程度上提示预后,但无法准确预测患者属于难治性或复发性[1,4,5]。

液体活检为非侵入性检查,包括循环肿瘤细胞(circulating tumor cell,CTC)检测、循环肿瘤DNA(circulating tumor DNA,ctDNA)检测、外泌体和循环RNA检测,检测样本可以是血浆、尿液等其他体液,该方法较常规组织活检简便易行,且易于反复取材进行动态监测[6]。循环游离DNA(circulating cell-free DNA,cfDNA)由细胞裂解、凋亡或细胞外囊泡分泌而从细胞释放至外周血中[7]。ctDNA是cfDNA的一部分,由肿瘤细胞直接分泌,ctDNA检测克服了肿瘤异质性,反映了淋巴瘤的体细胞突变、免疫球蛋白重链(immunoglobulin heavy chain,IgH)基因重排等情况[8]。ctDNA已被证明可用于多种肿瘤的诊断、预后评估和复发监测,如非小细胞肺癌[9]、肝癌[10,11]、结肠癌[12]和胰腺癌[13]等。ctDNA的含量低且不稳定,检测技术的灵敏度和可靠性也在不断改进和验证中,随着数字化聚合酶链反应(polymerase chain reaction,PCR)和二代基因测序技术的发展,已经发现了ctDNA在DLBCL中作为生物标志物的潜在作用[14–20]。本文就ctDNA在DLBCL的诊断分型、预后评估和复发监测等方面的潜在作用做一综述。

1 辅助诊断和基因分型 

研究发现在血浆样本中检测到的基因突变与配对的肿瘤组织所检测出的突变具有高度一致性。无论是数字PCR技术还是测序技术,均可检测到这种相关性[21-23]。这种一致性表明,使用ctDNA进行的非侵入性诊断可能与从组织活检获得的基因分型结果几乎一样准确。此外,液体活检可以帮助克服肿瘤组织活检的局限性,包括其侵入性和淋巴瘤的克隆异质性。与从肿瘤组织活检获取体细胞突变相比,通过ctDNA进行基因分型具有非侵入性的优势。

研究使用二代测序基因芯片检测DLBCL患者ctDNA中的体细胞突变,但每项研究检测使用的基因芯片不同,虽然包含越多基因的芯片检出率越高,但目前尚未有统一的芯片用于DLBCL检测,34个高频突变基因芯片(87703bp)的可靠性和实用性较高,仍需进一步扩大样本量验证其应用价值[24,25]。检测结果显示每个患者的突变情况不同,而在诊断时平均基因突变个数为4~124个,在63%~100%患者的血浆或血清样本中发现至少有1个基因突变。研究显示其灵敏度大于90%,特异度达100%[23,26-29]。大多数突变集中于少数基因中,主要的突变基因有13个,包括STAT6、CD79B、MYD88、CD58、GNA13、MYC、CREBBP、BCL2、PIM1、TP53、EZH2、MEF2B和B2M。通过检测这13个基因,可以从91例已明确有基因突变的患者中检测出79例具有相应突变,阳性率高达86.8%[8]。ctDNA检测可以区分DLBCL的不同分子亚型,检测出的基因分型结果与临床分型具有一致性,且检测出的热点突变如EZH2、CD79B、MYD88具有较高的临床相关性[30,31]。例如MYD88突变是ABC型DLBCL的特征性突变。研究表明MYD88基因突变可激活下游核因子-κB(nuclear factor-κB,NF-κB)信号通路,该作用机制可能与布鲁顿酪氨酸激酶(Bruton's tyrosine kinase,BTK)抑制剂的耐药相关[25]。利用34个基因芯片检测215例DLBCL患者血浆中的ctDNA,可以很好地区分DLBCL分子亚型,且证实ABC、GCB和PMBL分别受到NF-κB、表观遗传学和Janus激酶/信号转导和转录激活子(Janus kinase/signal transducer and activator of transcription,JAK/STAT)通路的影响。这些研究为我们可以使用基因数量较少的芯片来获得强有力的结果提供了依据,降低检测成本以转化为临床应用,为研究DLBCL的靶向治疗和耐药机制提供理论依据。

2 预后评估和疗效评价

Kurtz等[32]研究发现ctDNA在患者治疗后变化明显,应用早期分子反应(early molecular response,EMR,即1个化疗周期后降低2个对数)和主要分子反应(major molecular response,MMR,2个化疗周期后降低2.5个对数)来评估预后,接受一线治疗和补救治疗患者达到EMR和MMR均具有较好的预后,且分子反应可作为独立预后因子。高水平cfDNA提示预后较差,初治时复发/难治性DLBCL患者的cfDNA水平高于完全缓解患者,治疗后达到完全缓解或部分缓解患者cfDNA水平有所降低[33,34]。高ctDNA水平患者的2年无事件生存率低于低ctDNA水平患者[35,36]。

Scherer等[37]利用深度测序方法分析了92例淋巴瘤患者和24例健康受试者的肿瘤组织和cfDNA样本,结果发现ctDNA可以反映肿瘤遗传异质性和克隆演变,可作为评估预后的分子标志物,诊断时ctDNA水平与临床指标密切相关,是独立的预后影响因素。研究发现,ctDNA水平与IPI、代谢肿瘤体积(metabolic tumor volume,MTV)、乳酸脱氢酶(lactic acid dehydrogenase,LDH)水平和Ann Arbor分期有关,除了联合分析MTV大小、GCB/ABC状态和BCL2/MYC表达进行预后评估外,诊断时ctDNA的水平也与DLBCL的预后分层有关[37,38]。

另外,Assouline等[26]研究发现,在治疗后的第15天,ctDNA水平升高提示患者对治疗的反应差。Rossi等[23]研究发现,只有在对治疗无反应的患者中,治疗后才能检测到ctDNA突变,并且复发时发现的突变并不总是与诊断时的突变相同。这些结果均提示ctDNA有可能作为评估预后和评价疗效的生物标志物。

3 微小残留病变监测和复发预测

Kurtz等[31]提出追踪治疗前后ctDNA的体细胞突变在微小残留病(minimal residual disease,MRD)检测方面较影像学检查更具优势,其具有实时监测肿瘤发展的作用。Scherer等[37]发现73%的患者在复发前可以检测到ctDNA,从检测到ctDNA变化再到临床复发的中位时间为188 d。DLBCL是克隆性疾病,所有肿瘤细胞是相同的IgH VDJ序列,可以在DLBCL患者的肿瘤组织和血浆中检测到这些基因重排[39-41]。研究表明,IgH基因重排可作为监测复发的生物标志物,没有复发的患者很少发现IgH基因重排,其特异度大于98%;而在临床提示复发前可检测到IgH基因重排的患者比例差异很大[35,37,39-44]。Roschewski等[44]检测了108例患者2个治疗周期结束时IgH基因重排序列,证明IgH基因重排可提示早期复发,其灵敏度和特异度分别为47%和88%。这些结果表明,ctDNA可以作为MRD监测和复发预测的生物标志物,对ctDNA的监测可以更早提示复发,从而为早期干预和治疗提供机会,例如强化化疗、自体造血干细胞移植等治疗方案。但ctDNA转化并应用于临床仍有一段距离,需要开展相应的临床试验来证明其实用性。

4 总结

液体活检中的ctDNA检测是一种可替代肿瘤组织活检的非侵入性检测方法。ctDNA检测已被证明可以用于辅助诊断、基因分型、预后评估、疗效评价、MRD监测和复发预测,在其真正用于临床之前,需要提高检测技术,但在液体活检中研究ctDNA可能有助于改善DLBCL患者预后。


参考文献

[1] Caimi PF, Hill BT, Hsi ED, et al. Clinical approach to diffuse large B cell lymphoma[J]. Blood Rev, 2016, 30(6):477-491.

[2] Coiffier B, Thieblemont C, van den Neste E, et al. Long-term outcome of patients in the LNH-98.5 trial, the first randomized study comparing rituximab-CHOP to standard CHOP chemo­therapy in DLBCL patients: a study by the Groupe d'Etudes des Lymphomes de l'Adulte[J]. Blood, 2010, 116(12):2040-2045.

[3] Miyazaki K. Treatment of Diffuse Large B-Cell Lymphoma[J]. J Clin Exp Hematop, 2016, 56(2):79-88.

[4] Chan A, Dogan A. Prognostic and Predictive Biomarkers in Diffuse Large B-cell Lymphoma[J]. Surg Pathol Clin, 2019, 12(3):699-707.

[5] Li S, Young KH, Medeiros LJ. Diffuse large B-cell lymp­homa[J]. Pathology, 2018, 50(1):74-87.

[6] Mader S, Pantel K. Liquid Biopsy: Current Status and Future Perspectives[J]. Oncol Res Treat, 2017, 40(7-8):404-408.

[7] Costa JL, Schmitt FC. Liquid Biopsy: A New Tool in Onco­logy[J]. Acta Cytol, 2019, 63(6):448.

[8] Arzuaga-Mendez J, Prieto-Fernández E, Lopez-Lopez E, et al. Cell-free DNA as a biomarker in diffuse large B-cell lymphoma: A systematic review[J]. Crit Rev Oncol Hematol, 2019, 139:7-15.

[9] Wang Z, Duan J, Cai S, et al. Assessment of Blood Tumor Mutational Burden as a Potential Biomarker for Immunotherapy in Patients With Non-Small Cell Lung Cancer With Use of a Next-Generation Sequencing Cancer Gene Panel[J]. JAMA Oncol, 2019, 5(5):696-702.

[10] Cai Z, Chen G, Zeng Y, et al. Comprehensive Liquid Profiling of Circulating Tumor DNA and Protein Biomarkers in Long-Term Follow-Up Patients with Hepatocellular Carcinoma[J]. Clin Cancer Res, 2019, 25(17):5284-5294.

[11] Qu C, Wang Y, Wang P, et al. Detection of early-stage hepato­cellular carcinoma in asymptomatic HBsAg-seropositive indiv­iduals by liquid biopsy[J]. Proc Natl Acad Sci U S A, 2019, 116(13):6308-6312.

[12] Hamfjord J, Guren TK, Dajani O, et al. Total circulating cell-free DNA as a prognostic biomarker in metastatic colorectal cancer before first-line oxaliplatin-based chemotherapy[J]. Ann Oncol, 2019, 30(7):1088-1095.

[13] Berger AW, Schwerdel D, Reinacher-Schick A, et al. A Blood-Based Multi Marker Assay Supports the Differential Diagnosis of Early-Stage Pancreatic Cancer[J]. Theranostics, 2019, 9(5):1280-1287.

[14] Eskandari M, Manoochehrabadi S, Pashaiefar H, et al. Clinical significance of cell-free DNA as a prognostic biomarker in patients with diffuse large B-cell lymphoma[J]. Blood Res, 2019, 54(2):114-119.

[15] Hossain NM, Dahiya S, Le R, et al. Circulating tumor DNA assessment in patients with diffuse large B-cell lymphoma following CAR T-cell therapy[J]. Leuk Lymphoma, 2019, 60(2):503-506.

[16] Hu R, Winter A, Hill BT. The Emerging Role of Minimal Residual Disease Testing in Diffuse Large B-Cell Lymp­homa[J]. Curr Oncol Rep, 2019, 21(5):44.

[17] Sidaway P. ctDNA predicts outcomes in DLBCL[J]. Nat Rev Clin Oncol, 2018, 15(11):655.

[18] Camus V, Jardin F, Tilly H. The value of liquid biopsy in diagnosis and monitoring of diffuse large b-cell lymphoma: recent developments and future potential[J]. Expert Rev Mol Diagn, 2017, 17(6):557-566.

[19] Kwok M, Wu SP, Mo C, et al. Circulating Tumor DNA to Monitor Therapy for Aggressive B-Cell Lymphomas[J]. Curr Treat Options Oncol, 2016, 17(9):47.

[20] Melani C, Roschewski M. Molecular Monitoring of Cell-Free Circulating Tumor DNA in Non-Hodgkin Lymphoma[J]. Oncology (Williston Park), 2016, 30(8):731-738, 744.

[21] Alcaide M, Yu S, Bushell K, et al. Multiplex Droplet Digital PCR Quantification of Recurrent Somatic Mutations in Diffuse Large B-Cell and Follicular Lymphoma[J]. Clin Chem, 2016, 62(9):1238-1247.

[22] Bohers E, Viailly PJ, Dubois S, et al. Somatic mutations of cell-free circulating DNA detected by next-generation sequencing reflect the genetic changes in both germinal center B-cell-like and activated B-cell-like diffuse large B-cell lymphomas at the time of diagnosis[J]. Haematologica, 2015, 100(7):e280-e284.

[23] Rossi D, Diop F, Spaccarotella E, et al. Diffuse large B-cell lymphoma genotyping on the liquid biopsy[J]. Blood, 2017, 129(14):1947-1957.

[24] Dubois S, Viailly PJ, Mareschal S, et al. Next-Generation Sequencing in Diffuse Large B-Cell Lymphoma Highlights Molecular Divergence and Therapeutic Opportunities: a LYSA Study[J]. Clin Cancer Res, 2016, 22(12):2919-2928.

[25] Dubois S, Viailly PJ, Bohers E, et al. Biological and Clinical Relevance of Associated Genomic Alterations in MYD88 L265P and non-L265P-Mutated Diffuse Large B-Cell Lymphoma: Analysis of 361 Cases[J]. Clin Cancer Res, 2017, 23(9):2232-2244.

[26] Assouline SE, Nielsen TH, Yu S, et al. Phase 2 study of panobinostat with or without rituximab in relapsed diffuse large B-cell lymphoma[J]. Blood, 2016, 128(2):185-194.

[27] Bohers E, Viailly PJ, Becker S, et al. Non-invasive monitoring of diffuse large B-cell lymphoma by cell-free DNA high-throughput targeted sequencing: analysis of a prospective cohort[J]. Blood Cancer J, 2018, 8(8):74.

[28] Camus V, Sarafan-Vasseur N, Bohers E, et al. Digital PCR for quanti­fication of recurrent and potentially actionable somatic muta­tions in circulating free DNA from patients with diffuse large B-cell lymphoma[J]. Leuk Lymphoma, 2016, 57(9):2171-2179.

[29] Rossi D, Diop F, Spaccarotella E, et al. Diffuse large B-cell lymphoma genotyping on the liquid biopsy[J]. Blood, 2017, 129(14):1947-1957.

[30] Bohers E, Viailly PJ, Becker S, et al. Non-invasive monitoring of diffuse large B-cell lymphoma by cell-free DNA high-throughput targeted sequencing: analysis of a prospective cohort[J]. Blood Cancer J, 2018, 8(8):74.

[31] Kurtz DM, Scherer F, Jin MC, et al. Circulating Tumor DNA Measurements As Early Outcome Predictors in Diffuse Large B-Cell Lymphoma[J]. J Clin Oncol, 2018, 36(28):2845-2853.

[32] Kurtz DM, Scherer F, Jin MC, et al. Circulating Tumor DNA Measurements As Early Outcome Predictors in Diffuse Large B-Cell Lymphoma[J]. J Clin Oncol, 2018, 36(28):2845-2853.

[33] Hohaus S, Giachelia M, Massini G, et al. Cell-free circulating DNA in Hodgkin's and non-Hodgkin's lymphomas[J]. Ann Oncol, 2009, 20(8):1408-1413.

[34] Li M, Xu C. Circulating Cell-free DNA Utility for the Surve­illance of Patients with Treated Diffuse Large B-cell Lymp­homa[J]. Clin Oncol (R Coll Radiol), 2017, 29(9):637-638.

[35] Kurtz DM, Green MR, Bratman SV, et al. Noninvasive monitoring of diffuse large B-cell lymphoma by immu­noglo­bulin high-throughput sequencing[J]. Blood, 2015, 125(24): 3679-3687.

[36] Li M, Jia Y, Xu J, et al. Assessment of the circulating cell-free DNA marker association with diagnosis and prognostic prediction in patients with lymphoma: a single-center experience[J]. Ann Hematol, 2017, 96(8):1343-1351.

[37] Scherer F, Kurtz DM, Newman AM, et al. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA[J]. Sci Transl Med, 2016, 8(364):364ra155.

[38] Bohers E, Viailly PJ, Becker S, et al. Non-invasive monitoring of diffuse large B-cell lymphoma by cell-free DNA high-throughput targeted sequencing: analysis of a prospective cohort[J]. Blood Cancer J, 2018, 8(8):74.

[39] Armand P, Oki Y, Neuberg DS, et al. Detection of circulating tumour DNA in patients with aggressive B-cell non-Hodgkin lymphoma[J]. Br J Haematol, 2013, 163(1):123-126.

[40] Bo J, Sun L, Wang W, et al. Novel diagnostic biomarker for patients with Non-Hodgkin's Lymphoma by IgH gene rearrangement[J]. J Cancer Res Ther, 2016, 12(2):903-908.

[41] Zhong L, Huang WF. Better detection of Ig heavy chain and TCRγ gene rearrangement in plasma cell-free DNA from patients with non-Hodgkin Lymphoma[J]. Neoplasma, 2010, 57(6):507-511.

[42] Herrera AF, Kim HT, Kong KA, et al. Next-generation sequencing-based detection of circulating tumour DNA After allogeneic stem cell transplantation for lymphoma[J]. Br J Haematol, 2016, 175(5):841-850.

[43] Hossain NM, Dahiya S, Le R, et al. Circulating tumor DNA assessment in patients with diffuse large B-cell lymphoma following CAR T-cell therapy[J]. Leuk Lymphoma, 2019, 60(2):503-506.

[44] Roschewski M, Dunleavy K, Pittaluga S, et al. Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: a correlative biomarker study[J]. Lancet Oncol, 2015, 16(5):541-549.


循环肿瘤DNA在弥漫大B细胞淋巴瘤中的研究进展


分享到:


相關文章: