Advances in Clinical Medicine
Vol. 13  No. 05 ( 2023 ), Article ID: 65838 , 6 pages
10.12677/ACM.2023.1351169

D-二聚体检测对恶性肿瘤患者的临床意义

魏玥1,李国元2*

1青海大学研究生院,青海 西宁

2青海大学附属医院肿瘤内科,青海 西宁

收稿日期:2023年4月25日;录用日期:2023年5月19日;发布日期:2023年5月25日

摘要

D-二聚体是交联纤维蛋白被纤溶酶溶解后形成的蛋白凝块,以往有相关研究表明D-二聚体的水平与恶性肿瘤的分期、转移及化疗疗效有相关关系。通过比较D-二聚体在卵巢癌、胃癌、肺癌中的水平,并且与其他生物标志物敏感性作比较,得出D-二聚体检测是否对恶性肿瘤预后有相关临床意义,因此作下列综述。

关键词

D-二聚体,卵巢癌,胃癌,肺癌,预后

Clinical Significance of D-Dimer Detection in Patients with Malignant Tumors

Yue Wei1, Guoyuan Li2*

1Graduate School of Qinghai University, Xining Qinghai

2Department of Medical Oncology, Affiliated Hospital of Qinghai University, Xining Qinghai

Received: Apr. 25th, 2023; accepted: May 19th, 2023; published: May 25th, 2023

ABSTRACT

D-dimer is a protein clot formed after the cross-linked fibrin is dissolved by plasminase. Previous studies have shown that the level of D-dimer is related to the stage, metastasis and chemotherapy efficacy of malignant tumors. By comparing the level of D-dimer in ovarian cancer, gastric cancer and lung cancer, and comparing with the sensitivity of other biomarkers, it is concluded whether D-dimer detection has relevant clinical significance for the prognosis of malignant tumors. Therefore, the following review is made.

Keywords:D-Dimer, Ovarian Cancer, Gastric Cancer, Lung Cancer, Prognosis

Copyright © 2023 by author(s) and Hans Publishers Inc.

This work is licensed under the Creative Commons Attribution International License (CC BY 4.0).

http://creativecommons.org/licenses/by/4.0/

1. 引言

D- 二聚体的检测在临床上主要应用在静脉血栓栓塞(Venous Thrombo Embolism, VTE)、深静脉血栓形成和肺栓塞的诊断。升高的D-二聚体水平与原发性静脉血栓栓塞症(VTE)患者隐匿性癌症的发生存在关系 [1] 。在其他地方,Fei等人证明了这两个层次的D-二聚体和组织因子途径抑制剂——1TFPI-1 (Tissue Factor Pathway Inhibitor, TFPI)可预测肿瘤患者的深静脉血栓形成(Deep Venous Thrombosis, DVT) [2] 。这意味着预后不良的患者升高的D-二聚体水平可能是由于癌症患者静脉血栓形成发生率高 [3] 。此外,海特等人报道,肿瘤细胞不仅可能激活凝血系统,还可能损伤血管内皮细胞,增加血小板活性 [4] 。另一项研究报道凝血系统的激活与肿瘤的侵袭性生物学行为相关 [5] 。从机制上讲,这可能是由于肿瘤细胞可以通过释放一些细胞因子和蛋白质破坏正常细胞的功能,这些细胞因子和蛋白质破坏了抗凝和纤溶之间的平衡,导致细胞因子和凝集剂(如癌症促凝剂和组织因子)的释放 [6] 。其次,肿瘤细胞可通过分泌组织因子和一些炎性因子,如IL-1β和TNF-α,引起凝血–纤溶系统的异常激活 [7] 。第三,有报道称,血浆中D-二聚体、血管内皮生长因子(Vascular Endothelial Growth Factor, VEGF)是调节肺癌血管生成的最重要的血管生成因子 [8] 。因此,对D-二聚体与卵巢癌、胃癌、肺癌的相关关系作出下列综述。

2. D-二聚体与卵巢癌

卵巢癌在妇科恶性肿瘤中有较高的致死率 [9] 。2012年,估计有238,719名妇女被诊断患有卵巢癌,其中151,900人死亡 [10] 。在美国,大约87%的浆液性卵巢癌患者被诊断为晚期 [11] 。缺乏早期预测的生物标志物是导致高死亡率的原因。到目前为止,应用最广泛的卵巢癌监测生物标志物是糖类抗原125 (CA-125) [12] 。大约83%的晚期患者CA-125水平 > 35 U/mL [13] 。但在子宫内膜异位症、盆腔炎、妊娠、肝硬化、急性心衰、结核、胰腺癌、肺癌、肝癌等人群中,CA-125也有小部分升高 [14] [15] 。因此,它的阳性预测值相对较低,通常不被认为是一个独立的预测因子 [16] 此外,其对卵巢癌预后的预测价值仍存在争议 [17] [18] [19] 。因此,寻找其他可靠的生物标志物或与CA-125联合使用的生物标志物用于普通人群筛查,对卵巢癌进行有效的评估和预后预测是人们期待已久的。

使用PubMed、Cochrane图书馆和Web of Science图书馆,检索了截至2016年5月30日的所有关于血浆D-二聚体对卵巢癌的诊断和预后价值以及D-二聚体水平升高与卵巢癌静脉血栓栓塞(VTE)风险之间关系的相关研究。标准化平均差(SMD)、比值比(OR)、风险比(HR)和95%可信区间(CI)被适当地汇集起来。共纳入15项符合条件的研究,共涉及1437名癌症患者。高D-二聚体水平与卵巢癌患者的总生存率无显著相关性(HR 1.32,95%置信区间:0.90~1.95 P =  0.044)。然而,亚组分析表明,样本量可以解释研究之间的异质性。当样本量 ≥ 100时,D-二聚体升高可预测死亡风险增加(HR 1.800, 95% CI: 1.283~2.523, P = 0.845)。此外,恶性卵巢癌患者血浆D-二聚体水平显著高于良性对照组(SMD 0.774,95%置信区间:0.597~0.951,P = 0.39),晚期卵巢癌患者(国际妇产科联合会分类III和IV)高于早期卵巢癌患者(国际妇产科联合会分类I和II,SMD 0.611,95% CI:0.373~0.849,P = 0.442)。高D-二聚体水平表明高VTE风险(OR 4.068,95%置信区间:2.423~6.829,P = 0.629)的卵巢癌患者。

卵巢癌患者血浆D-二聚体水平可预测与疾病进展和VTE风险相关的变化。但其对卵巢癌预后的预测价值明显依赖于样本量。需要更多的设计良好的大样本量的研究来验证和更新本研究的结果。

3. D-二聚体与胃癌

当原发肿瘤释放肿瘤细胞进入循环系统成为CTC (循环肿瘤细胞,Circulating Tumor Cell, CTC)时,转移就开始了 [20] 。CTC在血管内滞留生成后,必须与血小板(platelet, PLT)、纤维蛋白原(fibrinogen, FIB)等凝血因子共同作用,形成微血栓,以帮助其在远处器官中粘附和转移 [21] 。人们普遍认为,在恶性肿瘤中常见的活化凝血为癌症转移提供了极大的便利,此外,在癌症患者中,通常观察到止血系统激活的证据,这是众所周知的现象,近年来的研究表明癌症与止血系统存在双向作用。恶性肿瘤中D-二聚体升高的潜在机制可能与CTC凝块(肿瘤栓塞)有关:肿瘤细胞最初停滞在毛细血管或器官的较大血管中后,血小板、血浆和其他成分很容易凝结并稳定地粘附到通过血栓的产生保护癌细胞增殖、粘附和通过脉管系统扩散。凝块(肿瘤栓子)以多种机制参与转移过程,包括:保护癌细胞免受免疫系统破坏、血流应激。凝血的各个组成部分已被证明会影响转移。基于以往的研究,他们认为较高的肿瘤相关促凝活性状态与肿瘤转移有密切关系 [22] 肿瘤患者凝血和纤溶系统的激活,可能部分反映了肿瘤细胞在宿主循环系统中的扩散。

纤维蛋白原(FIB)是纤维蛋白的重要来源,在循环肿瘤细胞(CTC)外渗和远处转移发展中起着至关重要的作用 [23] [24] D-二聚体是纤维蛋白的最终稳定产物,在凝血和纤溶系统增强激活后升高,广泛用于检测和排除深静脉血栓形成及相关血栓栓塞性疾病 [25] [26] 。近年来,多项研究报道恶性肿瘤患者血浆D-二聚体水平升高,其表达水平与肿瘤晚期、总生存期和治疗反应呈正相关 [27] - [35] 我们在先前对胃癌患者的研究中发现,D-二聚体比FIB和其他因素更能预测无症状内脏转移 [35] ,虽然FIB对CTCs的生存至关重要,但在临床上用于检测转移并不强,但D-二聚体在这一领域显示了其优势。它可能与CTC的出现有关,并能反映癌症患者的转移表型。

在不同的小鼠胃癌转移模型中验证了我们的假设,在三种转移免疫缺陷小鼠模型和对照组中直接检测了血浆D-二聚体和纤维蛋白原及其降解产物。接下来,我们收集并分析了41例进展期原发性胃癌(GC)患者血浆D-二聚体水平和CTCs数量的结果。对这些患者进行了随访研究。综上所述,我们讨论了D-二聚体来源于纤维蛋白的分解,在人血浆中稳定存在,是GC中CTCs必不可少的伴随物,易于测量,成本较低,可用于血行转移的检测。该检测方法对胃癌的常规检测有一定的参考价值,并能显著帮助临床医生预测胃癌患者的预后。

4. D-二聚体与肺癌

肿瘤细胞在激活凝血系统的同时还有可能破坏血管内皮细胞,使血小板的活性增强,通常来说,止血是促凝血因子和抗凝血因子之间平衡的动态过程,这可能与我们所知道的肿瘤细胞通过释放一些细胞因子和蛋白质来破坏正常细胞的功能有关,这些蛋白质有利于和抑制凝血和纤维蛋白溶解过程,这些细胞因子和蛋白质一旦破坏这种平衡,就会导致细胞因子和凝集剂(例如癌症促凝剂和组织因子)的过量失释放,以至于打破原来的动态平衡。第二,肿瘤细胞还会分泌有关的组织因子和炎症因子,如白介素、肿瘤坏死因子等,引起凝血–纤溶系统亢进。最后,血浆D-二聚体水平与血管内皮生长因子(VEGF)显着相关,VEGF是调节肺癌血管生成的最重要血管生成因子,D-二聚体的过表达也会提示肺部血管的异常生成,所以D-二聚体的异常表达值得引起注意。

恶性肿瘤的止血异常和高凝状态与许多因素有关。炎症细胞因子的释放、天然抗凝剂的抑制以及肿瘤细胞中止血蛋白的表达导致凝血系统激活和纤溶亢进。d-二聚体是纤维蛋白单体的降解产物,由活化的因子XIII交联并被纤溶酶水解。也是纤溶的标志物,其在血液中的浓度可以反映凝血系统和纤溶系统的激活程度。以往的研究已经证明,高预处理D-二聚体水平与肺癌预后不良,尤其是亚洲肺癌患者 [36] [37] [38] 。研究表明,增强凝血和纤溶功能可促进非小细胞肺癌的进展和转移 [39] [40] 。小细胞肺癌是肺癌的主要组织学类型,具有进展快、侵袭能力强、内分泌异常或类癌综合征发生率高等特点。小细胞肺癌患者的预后相当差 [41] 。因此,当务之急是找出能够准确预测患者生存期的因素,并为小细胞肺癌患者制定最佳治疗策略。预处理D-二聚体是肺癌预后的一个潜在指标,确定治疗前D-二聚体水平在预测小细胞肺癌(SCLC)患者的总生存期(OS)和无进展存活期(PFS)等临床结局中的预后意义。

在PubMed、Web of Science、EMBASE、Cochrane图书馆、中国知网、中国医学、万方、VIP等网站上进行系统检索数据库进行,以确定可用的研究。合并的风险比(HR)与95%置信区间(CI)被应用于评估预处理的相关性。D-二聚体水平与小细胞肺癌患者预后的关系。所有统计学分析均通过STATA 12.0版软件进行。本荟萃分析共纳入7项研究,涉及964例患者,所有患者均来自中国。结果表明,升高的预处理D-二聚体水平与较差的OS (HR = 1.90, 95% CI: 1.55~2.34, P ≤ 0.001)和PFS (比值比为1.52,95%可信区间为1.24~1.85,P < 0.001)显著相关。治疗前d-二聚体水平低的患者比治疗前d-二聚体水平高的患者具有更好的OS和PFS。该荟萃分析表明,治疗前血浆d-二聚体水平可能是预测SCLC患者生存的有前途的生物标志物。至于d-二聚体在SCLC中的临床意义,目前尚不清楚在癌症特异性治疗之前进行抗凝治疗是否会改善癌症患者的长期预后。此外,治疗前d-二聚体与SCLC的临床病理学特征(例如疾病的临床分期和组织学肿瘤类型)之间的关系仍然不明确,治疗期间d-二聚体水平动态变化的预后价值在接受放化疗的患者中尚不清楚。D-二聚体血药浓度是否作为预测小细胞肺癌预后的可靠指标,还需要更多设计良好的大样本前瞻性研究来验证。

5. 小结

血浆D-二聚体水平的检测在恶性肿瘤的分期、转移、预后的评估中有相对重要的价值,今后有可能成为独立的预测指标,同时,与其他生物标志物联合后,敏感性较与单个指标,有进一步提高,因此在恶性肿瘤中,临床干预的D-二聚体的阈值和治疗前后D-二聚体的水平检测对恶性肿瘤预后的影响值得进一步研究。

文章引用

魏 玥,李国元. D-二聚体检测对恶性肿瘤患者的临床意义
Clinical Significance of D-Dimer Detection in Patients with Malignant Tumors[J]. 临床医学进展, 2023, 13(05): 8361-8366. https://doi.org/10.12677/ACM.2023.1351169

参考文献

  1. 1. Han, D, ó Hartaigh, B., Lee, J.H., et al. (2016) Impact of D-Dimer for Prediction of Incident Occult Cancer in Patients with Unprovoked Venous Thromboembolism. PLOS ONE, 11, e0153514. https://doi.org/10.1371/journal.pone.0153514

  2. 2. Fei, X., Wang, H., Yuan, W., Wo, M. and Jiang, L. (2017) Tis-sue Factor Pathway Inhibitor-1 Is a Valuable Marker for the Prediction of Deep Venous Thrombosis and Tumor Metas-tasis in Patients with Lung Cancer. BioMed Research International, 2017, Article ID: 8983763. https://doi.org/10.1155/2017/8983763

  3. 3. Goldhaber, S.Z. and Bounameaux, H. (2012) Pulmonary Embolism and Deep Vein Thrombosis. The Lancet, 379, 1835-1846. https://doi.org/10.1016/S0140-6736(11)61904-1

  4. 4. Heit, J.A. (2005) Cancer and Venous Thromboembolism: Scope of the Problem. Cancer Control, 12, 5-10. https://doi.org/10.1177/1073274805012003S02

  5. 5. Beer, J.H., Haeberli, A., Vogt, A., et al. (2002) Coagulation Markers Predict Survival in Cancer Patients. Journal of Thrombosis and Haemostasis, 88, 745-749. https://doi.org/10.1055/s-0037-1613296

  6. 6. Gieseler, F., Lühr, I., Kunze, T., et al. (2007) Activated Coagulation Factors in Human Malignant Effusions and Their Contribution to Cancer Cell Metastasis and Therapy. Journal of Thrombosis and Haemostasis, 97, 1023-1030. https://doi.org/10.1160/TH06-12-0712

  7. 7. Gil-Bernabé, A.M., Ferjančič, Š., Tlalka, M., et al. (2012) Recruitment of Monocytes/Macrophages by Tissue Factor-Mediated Coagulation Is Essential for Metastatic Cell Survival and Premetastatic Niche Establishment in Mice. Blood, 119, 3164-3175. https://doi.org/10.1182/blood-2011-08-376426

  8. 8. Zhang, W.-W., Liu, K.-J., Hu, G.-L. and Liang, W.-J. (2015) Preoperative Platelet/Lymphocyte Ratio Is a Superior Prognostic Factor Compared to Other Systemic Inflammatory Re-sponse Markers in Ovarian Cancer Patients. Tumor Biology, 36. 8831-8837. https://doi.org/10.1007/s13277-015-3533-9

  9. 9. Watrowski, R. and Zeillinger, R. (2016) Simple Laboratory Score Improves the Preoperative Diagnosis of Adnexal Mass. Tumor Biology, 37, 4343-4349. https://doi.org/10.1007/s13277-015-4280-7

  10. 10. Narod, S. (2016) Can Advanced-Stage Ovarian Cancer Be Cured? Nature Reviews Clinical Oncology, 13, 255-261. https://doi.org/10.1038/nrclinonc.2015.224

  11. 11. Wei, S.U., Li, H. and Zhang, B. (2016) The Diagnostic Value of Serum HE4 and CA-125 and ROMA Index in Ovarian Cancer. Biomedical Reports, 5, 41-44. https://doi.org/10.3892/br.2016.682

  12. 12. Ogata, Y., Heppelmann, C.J., Charlesworth, M.C., et al. (2006) Elevated Levels of Phosphorylated Fibrinogen-α-Isoforms and Differential Expression of Other Post-Translationally Modified Proteins in the Plasma of Ovarian Cancer Patients. Journal of Proteome Research, 5, 3318-3325. https://doi.org/10.1021/pr060344+

  13. 13. Miralles, C., Orea, M., España, P., et al. (2003) Cancer Antigen 125 As-sociated with Multiple Benign and Malignant Athologies. Annals of Surgical Oncology, 10, 150-154. https://doi.org/10.1245/ASO.2003.05.015

  14. 14. Shiau, C.-S., Chang, M.-Y., Chiang, C.-H., et al. (2003) Ovarian Endometrioma Associated with very High Serum CA-125 Levels. Chang Gung Medical Journal, 26, 695-699.

  15. 15. Tiwari, R.K., Saha, K., Mukhopadhyay, D., et al. (2016) Evaluation of Preoperative Serum Levels of CA 125 and Expression of p53 in Ovarian Neoplasms: A Prospective Clinicopathological Study in a Tertiary Care Hospital. The Journal of Obstetrics and Gynecology of India, 66, 107-114. https://doi.org/10.1007/s13224-014-0611-7

  16. 16. Gadducci, A., Cosio, S., Tana, R. and Genazzani, A.R. (2009) Serum and Tissue Biomarkers as Predictive and Prognostic Variables in Epithelial Ovarian Cancer. Critical Reviews in Oncology/Hematology, 69, 12-27. https://doi.org/10.1016/j.critrevonc.2008.05.001

  17. 17. Zhang, Y.L., Chen, J.H., Lu, W., et al. (2016) Efficacy of Postoperative Hormone Replacement Therapy on Prognosis of Patients with Serous ovarian Carcinoma. Chinese Medical Journal, 129, 1316-1321. https://doi.org/10.4103/0366-6999.182835

  18. 18. Diaz-Gil, D., Fintelmann, F.J., Molaei, S., et al. (2016) Prediction of 5-Year Survival in Advanced-Stage Ovarian Cancer Patients Based on Computed Tomography Peritoneal Carcinoma-tosis Index. Abdominal Radiology, 41, 2196-202. https://doi.org/10.1007/s00261-016-0817-5

  19. 19. Gupta, G.P. and Massague, J. (2006) Cancer Metastasis: Building a Framework. Cell, 127, 679-695. https://doi.org/10.1016/j.cell.2006.11.001

  20. 20. Im, J.H., Fu, W., Wang, H., Bhatia, S.K., Hammer, D.A., Kowalska, M.A., et al. (2004) Coagulation Facilitates Tumor Cell Spreading in the Pulmonary Vasculature during Early Metastatic Colony Formation. Cancer Research, 64, 8613-8619. https://doi.org/10.1158/0008-5472.CAN-04-2078

  21. 21. Gay, L.J. and Felding-Habermann, B. (2011) Contribution of Platelets to Tumour Metastasis. Nature Reviews Cancer, 11, 123-134. https://doi.org/10.1038/nrc3004

  22. 22. Palumbo, J.S., Talmage, K.E., Massari, J.V., La Jeunesse, C.M., Flick, M.J., Kombrinck, K.W., et al. (2005) Platelets and Fibrin(ogen) Increase Metastatic Potential by Impeding Natural Killer Cell-Mediated Elimination of Tumor Cells. Blood, 105, 178-185. https://doi.org/10.1182/blood-2004-06-2272

  23. 23. Biggerstaff, J.P., Weidow, B., Vidosh, J., Dexheimer, J., Patel, S. and Patel, P. (2006) Soluble Fibrin Inhibits Monocyte Adherence and Cytotoxicity against Tumor Cells: Implications for Cancer Metastasis. Thrombosis Journal, 4, Article No. 12. https://doi.org/10.1186/1477-9560-4-12

  24. 24. Righini, M., Perrier, A., De Moerloose, P. and Bounameaux, H. (2008) D-Dimer for Venous Thromboembolism Diagnosis: 20 Years Later. Journal of Thrombosis and Haemostasis, 6, 1059-1071. https://doi.org/10.1111/j.1538-7836.2008.02981.x

  25. 25. Ay, C., Vormittag, R., Dunkler, D., Simanek, R., Chiriac, A.L., Drach, J., et al. (2009) D-Dimer and Prothrombin Fragment 1+2 Predict Venous Thromboembolism in Patients with Cancer: Results from the Vienna Cancer and Thrombosis Study. Journal of Clinical Oncology, 27, 4124-4129. https://doi.org/10.1200/JCO.2008.21.7752

  26. 26. Dompmartin, A., Ballieux, F., Thibon, P., Lequerrec, A., Hermans, C., Clapuyt, P., et al. (2009) Elevated D-Dimer Level in the Differential Diagnosis of Venous Malformations. Archives of Dermatology, 145, 1239-1244. https://doi.org/10.1001/archdermatol.2009.296

  27. 27. Go, S.I., Lee, M.J., Lee, W.S., Choi, H.J., Lee, U.S., Kim, R.B., et al. (2015) D-Dimer Can Serve as a Prognostic and Predictive Biomarker for Metastatic Gastric Cancer Treated by Chemotherapy. Medicine, 94, e951. https://doi.org/10.1097/MD.0000000000000951

  28. 28. Tomimaru, Y., Yano, M., Takachi, K., Kishi, K., Miyashiro, I., Ohue, M., et al. (2006) Plasma D-Dimer Levels Show Correlation with Number of Lymph Node Metastases in Pa-tients with Esophageal Cancer. Journal of the American College of Surgeons, 202, 139-145. https://doi.org/10.1016/j.jamcollsurg.2005.08.008

  29. 29. Lippi, G., Franchini, M., Biasiutti, C., Dellagiacoma, G., Salvagno, G.L. and Guidi, G.C. (2007) Increased D-Dimer Value and Occult Cancer in the Absence of Detectable Thrombosis. Haematologica, 92, 53-55. https://doi.org/10.3324/haematol.12237

  30. 30. Ay, C., Dunkler, D., Pirker, R., Thaler, J., Quehenberger, P., Wagner, O., et al. (2012) High D-Dimer Levels Are Associated with Poor Prognosis in Cancer Patients. Haematologica, 97, 1158-1164. https://doi.org/10.3324/haematol.2011.054718

  31. 31. Stender, M.T., Larsen, T.B., Sorensen, H.T. and Thor-lacius-Ussing, O. (2012) Preoperative Plasma D-Dimer Predicts 1-Year Survival in Colorectal Cancer Patients with Ab-sence of Venous Thromboembolism (VTE): A Prospective Clinical Cohort Study. Journal of Thrombosis and Haemo-stasis, 10, 2027-2031. https://doi.org/10.1111/j.1538-7836.2012.04887.x

  32. 32. Tas, F., Ciftci, R., Kilic, L., Bilgin, E., Keskin, S., Sen, F., et al. (2012) Clinical and Prognostic Significance of Coagulation Assays in Melanoma. Melanoma Research, 22, 368-375. https://doi.org/10.1097/CMR.0b013e328357be7c

  33. 33. Diao, D., Zhu, K., Wang, Z., Cheng, Y., Li, K., Pei, L., et al. (2013) Prognostic Value of the D-Dimer Test in Oesophageal Cancer during the Perioperative Period. The Journal of Surgical Oncology, 108, 34-41. https://doi.org/10.1002/jso.23339

  34. 34. Diao, D., Wang, Z., Cheng, Y., Zhang, H., Guo, Q., Song, Y., Zhu, K., Li, K., Liu, D. and Dang, C. (2014) D-Dimer: Not Just an Indicator of Venous Thrombosis but a Predictor of Asymptomatic Hematogenous Metastasis in Gastric Cancer Patients. PLOS ONE, 9, e101125. https://doi.org/10.1371/journal.pone.0101125

  35. 35. Zhou, Y.X., Yang, Z.M., Feng, J., Shan, Y.-J., Wang, W.-L. and Mei, Y.-Q. (2013) High Plasma D-Dimer Level Is Associated with Decreased Survival in Patients with Lung Cancer. Tumor Biology, 34, 3701-3704. https://doi.org/10.1007/s13277-013-0953-2

  36. 36. Li, W., Tang, Y., Song, Y., et al. (2018) Prognostic Role of Pre-treatment Plasma D-Dimer in Patients with Solid Tumors: A Systematic Review and Meta-Analysis. Cellular Physiology and Biochemistry, 45, 1663-1676. https://doi.org/10.1159/000487734

  37. 37. Ma, X., Li, Y., Zhang, J., Huang, J. and Liu, L. (2014) Prognostic Role of D-Dimer in Patients with Lung Cancer: A Meta-Analysis. Tumor Biology, 35, 2103-2109. https://doi.org/10.1007/s13277-013-1279-9

  38. 38. Zhang, P.P., Sun, J.W., Wang, X.Y., Liu, X.M. and Li, K. (2013) Preoperative Plasma D-Dimer Levels Predict Survival in Patients with Operable Non-Small Cell Lung Cancer Inde-pendently of Venous Thromboembolism. European Journal of Surgical Oncology, 39, 951-956. https://doi.org/10.1016/j.ejso.2013.06.008

  39. 39. Jiang, H.G., Li, J., Shi, S.B., Chen, P., Ge, L.-P., Jiang, Q. and Tang, X.-P. (2014) Value of Fibrinogen and D-Dimer in Predicting Recurrence and Metastasis after Radical Surgery for Non-Small Cell Lung Cancer. Medical Oncology, 31, Article No. 22. https://doi.org/10.1007/s12032-014-0022-8

  40. 40. Jemal, A., Bray, F., Center, M.M., Ferlay, J., Ward, E. and Forma, D. (2011) Global Cancer Statistics. CA, 61, 69-90. https://doi.org/10.3322/caac.20107

  41. 41. Torre, L.A., Siegel, R.L. and Jemal, A. (2016) Lung Cancer Statistics. In: Ahmad, A. and Gadgeel, S., Eds., Lung Cancer and Personalized Medicine, Advances in Experimental Medicine and Bi-ology, Vol. 893, Springer, Cham, 1-19. https://doi.org/10.1007/978-3-319-24223-1_1

期刊菜单