リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Soluble Programmed Cell Death Ligand 1 as a Novel Biomarker for Nivolumab Therapy for Non-Small-cell Lung Cancer」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Soluble Programmed Cell Death Ligand 1 as a Novel Biomarker for Nivolumab Therapy for Non-Small-cell Lung Cancer

和久井, 大 東京慈恵会医科大学 DOI:info:doi/10.1016/j.cllc.2018.04.014

2021.08.11

概要

Biomarkers for predicting the effect of antieprogrammed cell death 1 monoclonal antibody therapy against nonesmall-cell lung cancer (NSCLC) are urgently required. We prospectively studied the baseline plasma soluble programmed cell death ligand 1 (sPD-L1) levels from 39 NSCLC patients as a predictive marker of nivolumab therapy. The clinical benefit from nivolumab therapy was significantly associated with the baseline plasma sPD-L1 levels. Plasma sPD-L1 levels could represent a novel predictive marker for nivolumab therapy against NSCLC.

Background: Biomarkers for predicting the effect of antieprogrammed cell death 1 (PD-1) monoclonal antibody against nonesmall-cell lung cancer (NSCLC) are urgently required. Although it is known that the blood levels of soluble programmed cell death ligand 1 (sPD-L1) are elevated in various malignancies, the nature of sPD-L1 has not been thoroughly elucidated. We investigated the significance of plasma sPD-L1 levels as a biomarker for antiePD-1 monoclonal antibody, nivolumab therapy.

Patients and Methods: The present prospective study included 39 NSCLC patients. The patients were treated with nivolumab at the dose of 3 mg/kg every 2 weeks, and the effects of nivolumab on NSCLC were assessed according to the change in tumor size, time to treatment failure (TTF), and overall survival (OS). The baseline plasma sPD-L1 concentration was determined using an enzyme-linked immunosorbent assay.

Results: The area under the curve of the receiver operating characteristic curve was 0.761. The calculated optimal cutoff point for sPD-L1 in the plasma samples was 3.357 ng/mL. Of the 39 patients, 59% with low plasma sPD-L1 levels achieved a complete response or partial response and 25% of those with high plasma sPD-L1 levels did so. In addition, 22% of the patients with low plasma sPD-L1 levels developed progressive disease compared with 75% of those with high plasma sPD-L1 levels. The TTF and OS were significantly longer for those patients with low plasma sPD-L1 levels compared with the TTF and OS for those with high plasma sPD-L1 levels.

Conclusion: The clinical benefit from nivolumab therapy was significantly associated with the baseline plasma sPD-L1 levels. Plasma sPD-L1 levels might represent a novel biomarker for the prediction of the efficacy of nivolumab therapy against NSCLC.

参考文献

1. Giroux Leprieur E, Dumenil C, Julie C, et al. Immunotherapy revolutionises non- small-cell lung cancer therapy: results, perspectives and new challenges. Eur J Cancer 2017; 78:16-23.

2. Valecha GK, Vennepureddy A, Ibrahim U, et al. Anti-PD-1/PD-L1 antibodies in non-small cell lung cancer: the era of immunotherapy. Expert Rev Anticancer Ther 2017; 17:47-59.

3. Ellis PM, Vella ET, Ung YC. Immune checkpoint inhibitors for patients with advanced non-small-cell lung cancer: a systematic review. Clin Lung Cancer 2017; 18:444-59.

4. Khanna P, Blais N, Gaudreau PO, et al. Immunotherapy comes of age in lung cancer. Clin Lung Cancer 2017; 18:13-22.

5. Frydenlund N, Mahalingam M. PD-L1 and immune escape—insights from melanoma and other lineage-unrelated malignancies. Hum Pathol 2017; 66:13-33.

6. Wang J, Yuan R, Song W, et al. PD-1, PD-L1 (B7-H1) and tumor-site immune modulation therapy: the historical perspective. J Hematol Oncol 2017; 10:34.

7. Qin A, Coffey DG, Warren EH, et al. Mechanisms of immune evasion and current status of checkpoint inhibitors in non-small cell lung cancer. Cancer Med 2016; 5: 2567-78.

8. Gandini S, Massi D, Mandala M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: a systematic review and meta-analysis. Crit Rev Oncol Hematol 2016; 100:88-98.

9. Aguiar PN Jr, Santoro IL, Tadokoro H, et al. A pooled analysis of nivolumab for the treatment of advanced non-small-cell lung cancer and the role of PD-L1 as a predictive biomarker. Immunotherapy 2016; 8:1011-9.

10. Grigg C, Rizvi NA. PD-L1 biomarker testing for non-small cell lung cancer: truth or fiction? J Immunother Cancer 2016; 4:48.

11. Chae YK, Pan A, Davis AA, et al. Biomarkers for PD-1/PD-L1 blockade therapy in non-small-cell lung cancer: is PD-L1 expression a good marker for patient selec- tion? Clin Lung Cancer 2016; 17:350-61.

12. Wang X, Teng F, Kong L, et al. PD-L1 expression in human cancers and its as- sociation with clinical outcomes. Onco Targets Ther 2016; 9:5023-39.

13. Ilie M, Hofman V, Dietel M, et al. Assessment of the PD-L1 status by immu- nohistochemistry: challenges and perspectives for therapeutic strategies in lung cancer patients. Virchow Arch 2016; 468:511-25.

14. Pitt JM, Vetizou M, Daillere R, et al. Resistance mechanisms to immune- checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors. Immunity 2016; 44:1255-69.

15. Kleinovink JW, Marijt KA, Schoonderwoerd MJA, et al. PD-L1 expression on malignant cells is no prerequisite for checkpoint therapy. Oncoimmunology 2017; 6: e1294299.

16. Frigola X, Inman BA, Lohse CM, et al. Identification of a soluble form of B7-H1 that retains immunosuppressive activity and is associated with aggressive renal cell carcinoma. Clin Cancer Res 2011; 17:1915-23.

17. Rossille D, Gressier M, Damotte D, et al. High level of soluble programmed cell death ligand 1 in blood impacts overall survival in aggressive diffuse large B-cell lymphoma: results from a French multicenter clinical trial. Leukemia 2014; 28: 2367-75.

18. Wang L, Wang H, Chen H, et al. Serum levels of soluble programmed death ligand 1 predict treatment response and progression free survival in multiple myeloma. Oncotarget 2015; 6:41228-36.

19. Finkelmeier F, Canli O, Tal A, et al. High levels of the soluble programmed death- ligand (sPD-L1) identify hepatocellular carcinoma patients with a poor prognosis. Eur J Cancer 2016; 59:152-9.

20. Takahashi N, Iwasa S, Sasaki Y, et al. Serum levels of soluble programmed cell death ligand 1 as a prognostic factor on the first-line treatment of metastatic or recurrent gastric cancer. J Cancer Res Clin Oncol 2016; 142:1727-38.

21. Okuma Y, Hosomi Y, Nakahara Y, et al. High plasma levels of soluble pro- grammed cell death ligand 1 are prognostic for reduced survival in advanced lung cancer. Lung Cancer 2017; 104:1-6.

22. Chen Y, Wang Q, Shi B, et al. Development of a sandwich ELISA for evaluating soluble PD-L1 (CD274) in human sera of different ages as well as supernatants of PD-L1+ cell lines. Cytokine 2011; 56:231-8.

23. Kruger S, Legenstein ML, Rosgen V, et al. Serum levels of soluble programmed death protein 1 (sPD-1) and soluble programmed death ligand 1 (sPD-L1) in advanced pancreatic cancer. Oncoimmunology 2017; 6:e1310358..

24. Zhou J, Mahoney KM, Giobbie-Hurder A, et al. Soluble PD-L1 as a biomarker in malignant melanoma treated with checkpoint blockade. Cancer Immunol Res 2017; 5:480-92.

25. Yu H, Boyle TA, Zhou C, et al. PD-L1 expression in lung cancer. J Thorac Oncol 2016; 11:964-75.

26. Peng J, Hamanishi J, Matsumura N, et al. Chemotherapy induces programmed cell death-ligand 1 overexpression via the nuclear factor-kappaB to foster an immunosuppressive tumor microenvironment in ovarian cancer. Cancer Res 2015; 75:5034-45.

27. Topalian SL, Taube JM, Anders RA, et al. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 2016; 16:275-87.

28. Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol 2016; 17:e542-51.

29. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009; 45: 228-47.

30. McShane LM, Altman DG, Sauerbrei W, et al. Reporting recommendations for tumour MARKer prognostic studies (REMARK). Br J Cancer 2005; 93:387-91.

31. Merchant N, Nagaraju GP, Rajitha B, et al. Matrix metalloproteinases: their functional role in lung cancer. Carcinogenesis 2017; 38:766-80.

32. Wang S, Bajorath J, Flies DB, et al. Molecular modeling and functional mapping of B7-H1 and B7-DC uncouple costimulatory function from PD-1 interaction. J Exp Med 2003; 197:1083-91.

33. Ni X, Song Q, Cassady K, et al. PD-L1 interacts with CD80 to regulate graft- versus-leukemia activity of donor CD8+ T cells. J Clin Invest 2017; 127:1960-77.

34. Seo YD, Pillarisetty VG. T-cell programming in pancreatic adenocarcinoma: a review. Cancer Gene Ther 2017; 24:106-13.

35. Seton-Rogers S. Pancreatic cancer: dodging immunosuppression. Nat Rev Cancer 2016; 16:480-1.

36. Frigola X, Inman BA, Krco CJ, et al. Soluble B7-H1: differences in production between dendritic cells and T cells. Immunol Lett 2012; 142:78-82.

37. Yen JH, Khayrullina T, Ganea D. PGE2-induced metalloproteinase-9 is essential for dendritic cell migration. Blood 2008; 111:260-70.

38. Kis-Toth K, Bacskai I, Gogolak P, et al. Monocyte-derived dendritic cell sub- populations use different types of matrix metalloproteinases inhibited by GM6001. Immunobiology 2013; 218:1361-9.

39. Kossakowska AE, Edwards DR, Prusinkiewicz C, et al. Interleukin-6 regulation of matrix metalloproteinase (MMP-2 and MMP-9) and tissue inhibitor of metalloproteinase (TIMP-1) expression in malignant non-Hodgkin’s lymphomas. Blood 1999; 94:2080-9.

40. Shapiro SD, Campbell EJ, Kobayashi DK, et al. Immune modulation of metal- loproteinase production in human macrophages: selective pretranslational sup- pression of interstitial collagenase and stromelysin biosynthesis by interferon- gamma. J Clin Invest 1990; 86:1204-10.

41. Pico de Coana Y, Choudhury A, Kiessling R. Checkpoint blockade for cancer therapy: revitalizing a suppressed immune system. Trends Mol Med 2015; 21:482-91.

42. Aerts JG, Hegmans JP. Tumor-specific cytotoxic T cells are crucial for efficacy of immunomodulatory antibodies in patients with lung cancer. Cancer Res 2013; 73: 2381-8.

43. Gubin MM, Zhang X, Schuster H, et al. Checkpoint blockade cancer immuno- therapy targets tumour-specific mutant antigens. Nature 2014; 515:577-81.

44. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015; 348:69-74.

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る