Abstract:With the development of new therapeutic drugs and the optimization of multidisciplinary comprehensive treatment models, traditional pathological typing and detection methods are no longer sufficient to meet clinical needs. Next generation sequencing (NGS) has become a commonly used detection method by Chinese oncologists. In order to further assist clinicians to understand the annotation and interpretation of variants related to clinical targets or driver genes, sort out the logic of NGS reports, and improve the capture of key information, The Oncology Expert Group on the Interpretation of NGS Clinical Reports performed careful analysis, discussion, and summary based on the latest progress in NGS testing at home and abroad, and added some examples of NGS report interpretation, as well as interpretation of homologous recombination deficiency (HRD) and minimal/molecular/measurable residual disease (MRD) related content according to the “Guidelines for the Interpretation of Next-Generation Sequencing Clinical Reports”, and formulated the “Consensus on Interpretation of Clinical Reports for Next-Generation Sequencing of Tumors” to help clinicians make the right clinical decisions.
二代测序临床报告解读肿瘤学专家组. 肿瘤二代测序临床报告解读共识[J]. 循证医学, 2022, 22(2): 65-79.
The Oncology Expert Group on the Interpretation of Next Generation Sequencing Clinical Reports. Consensus on Interpretation of Clinical Reports for Next-Generation Sequencing of Tumors[J]. Journal of Evidence-based Medicine, 2022, 22(2): 65-79.
MORGANTI S, TARANTINO P, FERRARO E, et al.Complexity of genome sequencing and reporting: Next generation sequencing (NGS) technologies and implementation of precision medicine in real life[J]. Crit Rev OncolHematol, 2019,133:171-182.
[6]
LI M M, DATTO M, DUNCAVAGE E J, et al.Standards and guidelines for the interpretation and reporting of sequence variants in cancer: A joint consensus recommendation of the association for molecular pathology, American Society of Clinical Oncology, and College of American Pathologists[J]. J Mol Diagn, 2017, 19(1):4-23.
[7]
MATEO J,CHAKRAVARTY D,DIENSTMANN R,et al.A framework to rank genomic alterations as targets for cancer precision medicine: The ESMO Scale for Clinical Actionability of molecular Targets (ESCAT)[J]. Ann Oncol, 2018,29(9):1895-1902.
[8]
CHAKRAVARTY D, GAO J, PHILLIPS S M, et al. OncoKB: A precision oncology knowledge base[J]. JCO Precis Oncol, 2017:PO.17.00011.
[9]
ILYAS M.Next-generation sequencing in diagnostic pathology[J]. Pathobiology, 2017,84(6):292-305.
[10]
FRAMPTON G M, ALI S M, ROSENZWEIG M, et al.Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors[J]. Cancer Discov, 2015,5(8):850-859.
[11]
GOW C H, HSIEH M S, WU S G, et al.A comprehensive analysis of clinical outcomes in lung cancer patients harboring a MET exon 14 skipping mutation compared to other driver mutations in an East Asian population[J]. Lung Cancer, 2017,103:82-89.
[12]
HEIST R S, SHIM H S, GINGIPALLY S, et al.MET exon 14 skipping in non-small cell lung cancer[J]. Oncologist, 2016,21(4):481-486.
[13]
BENAYED R, OFFIN M, MULLANEY K, et al.High yield of RNA sequencing for targetable kinase fusions in lung adenocarcinomas with no mitogenic driver alteration detected by DNA sequencing and low tumor mutation burden[J]. Clin Cancer Res, 2019,25(15):4712-4722.
[14]
JURKIEWICZ M, SAQI A, et al. Efficacy of DNA versus RNA NGS-based methods in MET exon 14 skipping mutation detection[J]. J Clin Oncol, 2020 38(15S):Abstr9036.
[15]
TANG Y, LIU X, OU Z, et al.Maximum allele frequency observed in plasma: A potential indicator of liquid biopsy sensitivity[J]. Oncol Lett, 2019,18(2):2118-2124.
[16]
PÉCUCHET N, ZONTA E, DIDELOT A, et al. Base-position error rate analysis of next-generation sequencing applied to circulating tumor DNA in non-small cell lung cancer: A prospective study[J]. PLoS Med, 2016,13(12): e1002199.
[17]
WU Y L, LU S,CHENG Y, et al.Expert consensus of molecular residual disease for non-small cell lung cancer[J]. The Journal of Evidence-Base Medicine, 2021,21(3):129-135.[吴一龙, 陆舜, 程颖, 等. 非小细胞肺癌分子残留病灶专家共识[J]. 循证医学, 2021,21(3):129-135.]
[18]
LARRIBÈRE L, MARTENS U M. Advantages and challenges of using ctDNA NGS to assess the presence of minimal residual disease (MRD) in solid tumors[J]. Cancers, 2021,13(22):5698.
[19]
ZHANG J T,YANG X N,WU Y L.Minimal residual disease: Leading the cross - age biomarker for lung cancer[J]. The Journal of Evidence-Base Medicine, 2021,21(1):1-5.[张嘉涛, 杨学宁, 吴一龙. 引领肺癌治疗新时代的标志物——微小残留病灶[J]. 循证医学, 2021,21(1):1-5. ]
[20]
DETEERBECK F C, NICHOLSON A G, FRANKLIN W A, et al.The IASLC Lung Cancer Staging Project: Summary of proposals for revisions of the classification of lung cancers with multiple pulmonary sites of involvement in the forthcoming eighth edition of the TNM classification[J]. Thorac Oncol, 2016,11(5):639-650.
[21]
RICHARDS S, AZIZ N, BALE S, et al.Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology[J]. Genet Med, 2015,17(5):405-424.
[22]
ALLGÄUER M, BUDCZIES J, CHRISTOPOULOS P, et al. Implementing tumor mutational burden (TMB) analysis in routine diagnostics- A primer for molecular pathologists and clinicians[J]. Transl Lung Cancer Res, 2018,7(6):703-715.
[23]
CHALMERS Z R, CONNELLY C F, FABRIZIO D, et al.Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden[J]. Genome Med, 2017,9(1):34.
[24]
NIAN W Q, NIE Y Z, YING J M, et al.Chinese expert consensus on tumor mutational burden testing and clinical application[J]. Chinese Journal of Oncology Prevention and Treatment, 2020,12(5):485-494.[辇伟奇, 聂勇战, 应建明, 等. 肿瘤突变负荷检测及临床应用中国专家共识(2020年版)[J]. 中国癌症防治杂志, 2020,12(5):485-494. ]
[25]
MERINO D M, MCSHANE L M, et al.Establishing guidelines to harmonize tumor mutational burden (TMB): In silico assessment of variation in TMB quantification across diagnostic platforms: Phase I of the Friends of Cancer Research TMB Harmonization Project[J]. J Immunother Cancer, 2020,8(1):e000147.
[26]
STENZUNGER A, ENDRIS V, BUDCZIES J, et al.Harmonization and standardization of panel-based tumor mutational burden measurement: Real-world results and recommendations of the quality in pathology study[J]. Thorac Oncol, 2020,15(7):1177-1189.
[27]
LUCHINI C, BIBEAU F, LIGTENBERG M J L, et al.ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: A systematic review-based approach[J]. Ann Oncol, 2019,30(8):1232-1243.
[28]
BAUDRIN L G, DELEUZE J F, HOW-KIT A.Molecular and computational methods for the detection of microsatellite instability in cancer[J]. Front Oncol, 2018,8:621.
[29]
YUAN Y.Consensus on detection of microsatellite instability in colorectal cancer and other related solid tumors in China[J]. Journal of Practical Oncology, 2019,34(5):381-389.[袁瑛. 结直肠癌及其他相关实体瘤微卫星不稳定性检测中国专家共识[J].实用肿瘤杂志, 2019,34(5):381-389.]
[30]
CHEN R, JI Y, SHEN P, et al.Expert consensus on clinical detection and application of homologous recombination deficiency, 2021[J]. Chinese Journal of Oncology Prevention and Treatment, 2021,13(4):329-338.[陈锐, 纪元, 申鹏, 等. 同源重组修复缺陷临床检测与应用专家共识(2021版)[J]. 中国癌症防治杂志, 2021,13(4):329-338.]
[31]
TEW W P, LACCHETTI C, ELLIS A, et al.PARP inhibitors in the management of ovarian cancer: ASCO guideline[J]. J Clin Oncol, 2020,38(30):3468-3493.
[32]
MANSFIELD A S, PARK B H, MULLANE M P.Identification, prioritization, and treatment of mutations identified by next-generation sequencing[J]. Am Soc Clin Oncol Educ Book, 2018,38:873-880.
[33]
ROLFO C, MANCA P, SALGADO R, et al.Multidisciplinary molecular tumour board: A tool to improve clinical practice and selection accrual for clinical trials in patients with cancer[J]. ESMO Open, 2018,3(5):e000398.