Development and validation of a prognostic index for efficacy evaluation and prognosis of first-line chemotherapy in stage III-IV lung squamous cell carcinoma

Jiangdian Song, Jie Tian, Lina Zhang, Xiujuan Qu, Wei Qian, Bin Zheng, Lina Zhang, Jia Zhao, Meng Niu, Mu Zhou, Lei Cui, Yunpeng Liu, Mingfang Zhao, Jiangdian Song, Jie Tian, Lina Zhang, Xiujuan Qu, Wei Qian, Bin Zheng, Lina Zhang, Jia Zhao, Meng Niu, Mu Zhou, Lei Cui, Yunpeng Liu, Mingfang Zhao

Abstract

Objectives: To establish a pre-therapy prognostic index model (PIM) of the first-line chemotherapy aiming to achieve accurate prediction of time to progression (TTP) and overall survival among the patients diagnosed with locally advanced (stage III) or distant metastasis (stage IV) lung squamous cell carcinoma (LSCC).

Methods: Ninety-six LSCC patients treated with first-line chemotherapy were retrospectively enrolled to build the model. Fourteen epidermal growth factor receptor (EGFR)-mutant LSCC patients treated with first-line EGFR-tyrosine kinase inhibitor (TKI) therapy were enrolled for validation dataset. From CT images, 56,000 phenotype features were initially computed. PIM was constructed by integrating a CT phenotype signature selected by the least absolute shrinkage and selection operator and the significant blood-based biomarkers selected by multivariate Cox regression. PIM was then compared with other four prognostic models constructed by the CT phenotype signature, clinical factors, post-therapy tumor response, and Glasgow Prognostic Score.

Results: The signature includes eight optimal features extracted from co-occurrence, run length, and Gabor features. By using PIM, chemotherapy efficacy of patients categorized in the low-risk, intermediate-risk, and high-risk progression subgroups (median TTP = 7.2 months, 3.4 months, and 1.8 months, respectively) was significantly different (p < 0.0001, log-rank test). Chemotherapy efficacy of the low-risk progression subgroup was comparable with EGFR-TKI therapy (p = 0.835, log-rank test). Prognostic prediction of chemotherapy efficacy by PIM was significantly higher than other models (p < 0.05, z test).

Conclusion: The study demonstrated that the PIM yielded significantly higher performance to identify individual stage III-IV LSCC patients who can potentially benefit most from first-line chemotherapy, and predict the risk of failure from chemotherapy for individual patients.

Key points: • TTP and OS of first-line chemotherapy in individual stage III-IV LSCC patients could be predicted by pre-therapy blood-based biomarkers and image-based signatures. • Risk status of pre-therapy indicators affected the efficacy of first-line chemotherapy in stage III-IV LSCC patients. • Those stage III-IV LSCC patients who were able to achieve similar efficacy to EGFR-TKI therapy through chemotherapy were identified.

Keywords: Biomarkers; Carcinoma; Prognosis; Squamous cell; Tumor.

Conflict of interest statement

Guarantor

The scientific guarantor of this publication is Prof. Mingfang Zhao.

Conflict of interest

The authors of this manuscript declare no relationships with any companies, whose products or services may be related to the subject matter of the article.

Statistics and biometry

Prof. Cui is an expert in the field of medical statistics. We consulted him about the statistics analysis in this study.

Informed consent

Written informed consent was waived by the Institutional Review Board.

Ethical approval

Institutional Review Board approval was obtained.

Methodology

• retrospective

• diagnostic or prognostic study

• performed at one institution

Figures

Fig. 1
Fig. 1
Flowchart of this study. The first step was model construction, and based on the constructed model, model validation and comparison were performed. LSCC, lung squamous cell carcinoma; TTP, time to progression; OS, overall survival. NRI net reclassification improvement, IDI integrated discrimination improvement
Fig. 2
Fig. 2
The diagram of manual segmentation by using ITK-SNAP. The subgraph in the upper left corner indicates that the manually segmented region of interest (ROI) by the radiologist from cross section. The subgraphs in the upper right and lower right corners represent the manual segmentation result of the tumor which is displayed from the sagittal and coronal planes, respectively. The tumor is then reconstructed in a view of three dimensions, which is represented in the subgraph in the lower left corner. Each of the subgraphs could be scaled to ensure accurate segmentation
Fig. 3
Fig. 3
Results of progression risk prediction. a The Kaplan-Meier curves of groups classified by the signature, and all patients were stratified into good time to progression (TTP) group and poor TTP group according to the signature. b, c The progression risk prediction of the prognostic index model (PIM). b The result of low-risk (yellow line), intermediate-risk (blue line), and high-risk (pink line) progression subgroups by the PIM. c The comparison between the stage III–IV EGFR-mutant LSCC patients treated with first-line EGFR-TKI therapy (red line) and the different risk subgroups of chemotherapy patients stratified by the PIM. d The Kaplan-Meier curves of the patients with partial response (PR), stable disease (SD), and progressive disease (PD)
Fig. 4
Fig. 4
The comparison of the models. a The plots depict the calibration of the model in terms of the agreement between predicted and observed TTP (time.inc = 6 months). Performances of the models are shown on the plots relative to the 45° line, which represents perfect prediction. b Decision curve analysis of the PIM (red line), clinical factor-based model (green line), post-treatment tumor response-based model (cyan line), and the intra-tumor heterogeneity signature-based model (blue line). The orange line represents the assumption that all patients were treated. The black line represents the assumption that no patient was treated. The x-axis represents the risk of progression (Pt). The y-axis measures the net benefit. The net benefit was calculated by subtracting the proportion of all patients who are false positive from the proportion who are true positive, weighting by (Pt/(1 − Pt)). The decision curve showed that if the threshold probability of a patient or doctor is > 26%, using the PIM to predict progression risk adds more benefit than the treat-all-patients scheme or the treat-none scheme, or other prognostic models. c The clinical impact curve of the PIM; the red line (number of high risk) represents the patients with a high risk of progression predicted by the PIM at each threshold (with 95% CI), and the green line (number of high risk with outcome) represents the patients with actual progression at each threshold (with 95% CI)
Fig. 5
Fig. 5
Prognostication of overall survival, a further exploration of the proposed PIM. By applying the PIM on overall survival, the Kaplan-Meier survival curves are the stratified high-risk (pink), intermediate-risk (blue), and low-risk (yellow) chemotherapy patient subgroups

References

    1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A (2015) Global cancer statistics, 2012. CA Cancer J Clin 65:87–108
    1. Choi M, Kadara H, Zhang J, et al. Mutation profiles in early-stage lung squamous cell carcinoma with clinical follow-up and correlation with markers of immune function. Ann Oncol. 2017;28:83–89. doi: 10.1093/annonc/mdx369.037.
    1. Shukuya T, Yamanaka T, Seto T, et al. Nedaplatin plus docetaxel versus cisplatin plus docetaxel for advanced or relapsed squamous cell carcinoma of the lung (WJOG5208L): a randomised, open-label, phase 3 trial. Lancet Oncol. 2015;16:1630–1638. doi: 10.1016/S1470-2045(15)00305-8.
    1. Soria JC, Felip E, Cobo M, et al. Afatinib versus erlotinib as second-line treatment of patients with advanced squamous cell carcinoma of the lung (LUX-Lung 8): an open-label randomised controlled phase 3 trial. Lancet Oncol. 2015;16:897–907. doi: 10.1016/S1470-2045(15)00006-6.
    1. Ettinger DS, Wood DE, Aisner DL, et al. Non-small cell lung cancer, version 5.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Cancer Netw. 2017;15:504–535. doi: 10.6004/jnccn.2017.0050.
    1. Thatcher N, Hirsch FR, Luft AV, et al. Necitumumab plus gemcitabine and cisplatin versus gemcitabine and cisplatin alone as first-line therapy in patients with stage IV squamous non-small-cell lung cancer (SQUIRE): an open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2015;16:763–774. doi: 10.1016/S1470-2045(15)00021-2.
    1. Rizvi NA, Mazières J, Planchard D, et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol. 2015;16:257–265. doi: 10.1016/S1470-2045(15)70054-9.
    1. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non–small-cell lung cancer. N Engl J Med. 2015;373:123–135. doi: 10.1056/NEJMoa1504627.
    1. Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–892. doi: 10.1056/NEJMoa1113205.
    1. Longo DL. Tumor heterogeneity and personalized medicine. N Engl J Med. 2012;366:956–957. doi: 10.1056/NEJMe1200656.
    1. Aerts HJ, Velazquez ER, Leijenaar RT, et al. Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nat Commun. 2014;5:4006. doi: 10.1038/ncomms5006.
    1. Rizzo S, Botta F, Raimondi S et al (2018) Radiomics of high-grade serous ovarian cancer: association between quantitative CT features, residual tumour and disease progression within 12 months. Eur Radiol. 10.1007/s00330-018-5389-z
    1. Zhou M, Leung A, Echegaray S, et al. Non-small cell lung cancer radiogenomics map identifies relationships between molecular and imaging phenotypes with prognostic implications. Radiology. 2018;286:307–315. doi: 10.1148/radiol.2017161845.
    1. Suh HB, Choi YS, Bae S et al (2018) Primary central nervous system lymphoma and atypical glioblastoma: differentiation using radiomics approach. Eur Radiol. 10.1007/s00330-018-5368-4
    1. Huang YQ, Liang CH, He L, et al. Development and validation of a radiomics nomogram for preoperative prediction of lymph node metastasis in colorectal cancer. J Clin Oncol. 2016;34:2157–2164. doi: 10.1200/JCO.2015.65.9128.
    1. Kim SJ, Yoon DH, Jaccard A, et al. A prognostic index for natural killer cell lymphoma after non-anthracycline-based treatment: a multicentre, retrospective analysis. Lancet Oncol. 2016;17:389–400. doi: 10.1016/S1470-2045(15)00533-1.
    1. Wu J, Aguilera T, Shultz D, et al. Early-stage non–small cell lung cancer: quantitative imaging characteristics of 18 F fluorodeoxyglucose PET/CT allow prediction of distant metastasis. Radiology. 2016;281:270–278. doi: 10.1148/radiol.2016151829.
    1. Aerts HJ. The potential of radiomic-based phenotyping in precision medicine: a review. JAMA Oncol. 2016;2:1636–1642. doi: 10.1001/jamaoncol.2016.2631.
    1. Rios Velazquez E, Parmar C, Liu Y, et al. Somatic mutations drive distinct imaging phenotypes in lung cancer. Cancer Res. 2017;77:3922–3930. doi: 10.1158/0008-5472.CAN-17-0122.
    1. Coroller TP, Agrawal V, Huynh E, et al. Radiomic-based pathological response prediction from primary tumors and lymph nodes in NSCLC. J Thorac Oncol. 2017;12:467–476. doi: 10.1016/j.jtho.2016.11.2226.
    1. Guinney J, Wang T, Laajala TD, et al. Prediction of overall survival for patients with metastatic castration-resistant prostate cancer: development of a prognostic model through a crowdsourced challenge with open clinical trial data. Lancet Oncol. 2017;18:132–142. doi: 10.1016/S1470-2045(16)30560-5.
    1. Rawson NS, Peto J. An overview of prognostic factors in small cell lung cancer. A report from the Subcommittee for the Management of Lung Cancer of the United Kingdom Coordinating Committee on Cancer Research. Br J Cancer. 1990;61:597–604. doi: 10.1038/bjc.1990.133.
    1. Xu L, Peng ZW, Chen MS, et al. Prognostic nomogram for patients with unresectable hepatocellular carcinoma after transcatheter arterial chemoembolization. J Hepatol. 2015;63:122–130. doi: 10.1016/j.jhep.2015.02.034.
    1. Margalit O, Mamtani R, Yang YX, et al. Assessing the prognostic value of carcinoembryonic antigen levels in stage I and II colon cancer. Eur J Cancer. 2018;94:1–5. doi: 10.1016/j.ejca.2018.01.112.
    1. Okamura K, Takayama K, Izumi M, Harada T, Furuyama K, Nakanishi Y (2013) Diagnostic value of CEA and CYFRA 21-1 tumor markers in primary lung cancer. Lung Cancer 80:45–49
    1. Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 2004;10:7252–7259. doi: 10.1158/1078-0432.CCR-04-0713.
    1. Kratz A, Ferraro M, Sluss PM, Lewandrowski KB. Case records of the Massachusetts General Hospital. Weekly clinicopathological exercises. Laboratory reference values. N Engl J Med. 2004;351:1548–1563. doi: 10.1056/NEJMcpc049016.
    1. Nozoe T, Matono R, Ijichi H, Ohga T, Ezaki T (2014) Glasgow prognostic score (GPS) can be a useful indicator to determine prognosis of patients with colorectal carcinoma. Int Surg 99:512–517
    1. Cui Y, Ren S, Tha KK, Wu J, Shirato H, Li R (2017) Volume of high-risk intratumoral subregions at multi-parametric MR imaging predicts overall survival and complements molecular analysis of glioblastoma. Eur Radiol 27:3583–3592
    1. Hu H, Han H, Han XK, Wang WP, Ding H. Nomogram for individualised prediction of liver failure risk after hepatectomy in patients with resectable hepatocellular carcinoma: the evidence from ultrasound data. Eur Radiol. 2018;28:877–885. doi: 10.1007/s00330-017-4900-2.
    1. Reisæter LAR, Fütterer JJ, Losnegård A, et al. Optimising preoperative risk stratification tools for prostate cancer using mpMRI. Eur Radiol. 2018;28:1016–1026. doi: 10.1007/s00330-017-5031-5.
    1. Besson FL, Henry T, Meyer C, et al. Rapid contour-based segmentation for (18)F-FDG PET imaging of lung tumors by using ITK-SNAP: comparison to expert-based segmentation. Radiology. 2018;288:277–284. doi: 10.1148/radiol.2018171756.
    1. O’Connor JP, Aboagye EO, Adams JE, et al. Imaging biomarker roadmap for cancer studies. Nat Rev Clin Oncol. 2017;14:169–186. doi: 10.1038/nrclinonc.2016.162.
    1. Song J, Shi J, Dong D, et al. A new approach to predict progression-free survival in stage IV EGFR-mutant NSCLC patients with EGFR-TKI therapy. Clin Cancer Res. 2018;24:3583–3592. doi: 10.1158/1078-0432.CCR-17-2507.
    1. Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012;13:239–246. doi: 10.1016/S1470-2045(11)70393-X.
    1. Soria JC, Wu YL, Nakagawa K, et al. Gefitinib plus chemotherapy versus placebo plus chemotherapy in EGFR-mutation-positive non-small-cell lung cancer after progression on first-line gefitinib (IMPRESS): a phase 3 randomised trial. Lancet Oncol. 2015;16:990–998. doi: 10.1016/S1470-2045(15)00121-7.
    1. Yan H, Li Q, Wang W, Zhen H, Cao B (2015) Systems assessment of intercalated combination of chemotherapy and EGFR TKIs versus chemotherapy or EGFR TKIs alone in advanced NSCLC patients. Sci Rep 5. 10.1038/srep15355
    1. Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389:255–265. doi: 10.1016/S0140-6736(16)32517-X.
    1. Pajares MJ, Agorreta J, Salvo E, et al. TGFBI expression is an independent predictor of survival in adjuvant-treated lung squamous cell carcinoma patients. Br J Cancer. 2014;110:1545–1551. doi: 10.1038/bjc.2014.33.
    1. Arrieta O, Campos-Parra AD, Zuloaga C, et al. Clinical and pathological characteristics, outcome and mutational profiles regarding non-small-cell lung cancer related to wood-smoke exposure. J Thorac Oncol. 2012;7:1228–1234. doi: 10.1097/JTO.0b013e3182582a93.
    1. Rosell R, Carcereny E, Gervais R et al (2012) Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 13:239–246
    1. Rosell R, Moran T, Queralt C, et al. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. 2009;361:958–967. doi: 10.1056/NEJMoa0904554.
    1. NSCLC Meta-analysis Collaborative Group (2014) Preoperative chemotherapy for non-small-cell lung cancer: a systematic review and meta-analysis of individual participant data. Lancet 383:1561–1571
    1. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin–paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947–957. doi: 10.1056/NEJMoa0810699.
    1. Riihimäki M, Hemminki A, Fallah M, et al. Metastatic sites and survival in lung cancer. Lung Cancer. 2014;86:78–84. doi: 10.1016/j.lungcan.2014.07.020.
    1. Wu KL, Tsai MJ, Yang CJ, et al. Liver metastasis predicts poorer prognosis in stage IV lung adenocarcinoma patients receiving first-line gefitinib. Lung Cancer. 2015;88:187–194. doi: 10.1016/j.lungcan.2015.02.012.

Source: PubMed

3
S'abonner