Anti-Programmed Cell Death (PD)-1 Immunotherapy for Malignant Tumor: A Systematic Review and Meta-Analysis

Ran Chen, Pei-Chun Peng, Bin Wen, Fu-Ying Li, Sheng Xie, Guozhong Chen, Jiefu Lu, Zhuoyu Peng, Shao-Bo Tang, Yu-Mei Liang, Xin Deng, Ran Chen, Pei-Chun Peng, Bin Wen, Fu-Ying Li, Sheng Xie, Guozhong Chen, Jiefu Lu, Zhuoyu Peng, Shao-Bo Tang, Yu-Mei Liang, Xin Deng

Abstract

This systematic review and meta-analysis evaluated anti-programmed cell death (PD)-1 immunotherapy (nivolumab or pembrolizumab) for overall efficacy, safety, and effective dose relative to standard chemotherapy or other conventional drugs in the treatment of malignant tumors. We searched the following databases, PubMed, Medline, Embase, Cochrane, Wangfang Data, Weipu, and China National Knowledge Infrastructure, and the reference lists of the selected articles for randomized controlled trials (RCTs) of anti-PD-1 therapies in humans. The outcome measures were overall survival, treatment response, and adverse events. Only four randomized controlled trials met our inclusion criteria. Three of these evaluated responses to nivolumab, whereas one tested pembrolizumab. The result of our analysis suggested that nivolumab may improve the overall response rate in treating melanoma relative to chemotherapy and has few associated adverse events. Similarly, in metastatic melanoma patients, nivolumab had a significant advantage over dacarbazine in terms of 1-year survival, progression-free survival, and objective response rate. Regarding dose levels of nivolumab for patients with metastatic renal cell carcinoma, the outcomes in response to 2 and 10 mg/kg were similar, but both had significant advantages over 0.3 mg/kg. In addition, pembrolizumab showed similar outcomes in response to 2- and 10-mg/kg treatment. Anti-PD-1 immunotherapy appears to be safe and effective for patients with melanoma or metastatic renal cell carcinoma. Our meta-analysis is limited, but additional clinical trials are warranted to verify this preliminary evidence of positive outcomes and before anti-PD-1 therapy can be recommended for routine clinical use.

Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
Flowchart representing the systematic identification of studies.
Figure 2
Figure 2
Assessment of risk bias.
Figure 3
Figure 3
Forest plot depicting the overall response rate of nivolumab compared with chemotherapy.
Figure 4
Figure 4
Forest plot depicting the comparison of adverse events associated with nivolumab and chemotherapy.
Figure 5
Figure 5
Forest plot depicting the 1-year OS rates of nivolumab and dacarbazine.
Figure 6
Figure 6
Forest plot depicting the PFS rates associated with nivolumab and dacarbazine.
Figure 7
Figure 7
Forest plot depicting the objective response rates associated with nivolumab and dacarbazine.
Figure 8
Figure 8
Forest plot depicting the comparison of adverse events associated with nivolumab and dacarbazine.
Figure 9
Figure 9
Forest plot depicting the overall response rate with different doses (2 and 10 mg/kg) of pembrolizumab.
Figure 10
Figure 10
Forest plot depicting the PFS rates associated with different doses (2 and 10 mg/kg) of pembrolizumab.
Figure 11
Figure 11
Forest plot depicting 1-year survival rates associated with different doses (2 and 10 mg/kg) of pembrolizumab.
Figure 12
Figure 12
Forest plot depicting overall response rates of different doses (0.3 and 2 mg/kg) of nivolumab.
Figure 13
Figure 13
Forest plot depicting the PFS rates associated with different doses (0.3 and 2 mg/kg) of nivolumab.
Figure 14
Figure 14
Forest plot depicting the OS rates associated with different doses (0.3 and 2 mg/kg) of nivolumab.
Figure 15
Figure 15
Forest plot depicting the overall response rates associated with different doses (0.3 and 10 mg/kg) of nivolumab.
Figure 16
Figure 16
Forest plot depicting PFS rates associated with different doses (0.3 and 10 mg/kg) of nivolumab.
Figure 17
Figure 17
Forest plot depicting the OS rates associated with different doses (0.3 and 10 mg/kg) of nivolumab.
Figure 18
Figure 18
Forest plot depicting the overall response rates associated with different doses (2 and 10 mg/kg) of nivolumab.
Figure 19
Figure 19
Forest plot depicting the OS rates associated with different doses (2 and 10 mg/kg) of nivolumab.
Figure 20
Figure 20
Forest plot depicting the PFS rates associated with different doses (2 and 10 mg/kg) of nivolumab.

References

    1. Stewart BW, Wild CP. IARC Nonserial Publication; 2014. World Cancer Report.
    1. Bryan LJ, Gordon LI. Blocking tumor escape in hematologic malignancies: the anti–PD-1 strategy. Blood Rev. 2015;29:25–32.
    1. Li R, Wang C, Liu L, Du C, Cao S, Yu J, Wang SE, Hao X, Ren X, Li H. Autologous cytokine-induced killer cell immunotherapy in lung cancer: a phase II clinical study. Cancer Immunol Immunother. 2012;61:2125–2133.
    1. Hontscha C, Borck Y, Zhou H, Messmer D, Schmidt-Wolf IG. Clinical trials on CIK cells: first report of the international registry on CIK cells (IRCC) J Cancer Res Clin Oncol. 2011;137:305–310.
    1. Mahoney KM, Freeman GJ, McDermott DF. The next immune-checkpoint inhibitors: PD-1/PD-L1 blockade in melanoma. Clin Ther. 2015;37:764–782.
    1. Tang PA, Heng DY. Programmed death 1 pathway inhibition in metastatic renal cell cancer and prostate cancer. Curr Oncol Rep. 2013;15:98–104.
    1. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.
    1. Okazaki T, Honjo T. The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 2006;27:195–201.
    1. Quezada SA, Peggs KS. Exploiting CTLA-4, PD-1 and PD-L1 to reactivate the host immune response against cancer. Br J Cancer. 2013;108:1560–1565.
    1. Higgins JPT, Green S. The Cochrane Collaboration. 2011. Cochrane Handbook for Systematic Reviews of Interventions, Version 5.2.0.
    1. Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, Kefford R, Weber JS, Joshua AM, Hwu WJ, Gangadhar TC. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014;384:1109–1117.
    1. Weber JS, Minor DR, D'Angelo S, Hodi FS, Gutzmer R, Neyns B, Hoeller C, Khushalani NI, Miller WH, Grob J. European Society for Medical Oncology 2014 Congress. 2014. A phase 3 randomized, open-label study of nivolumab (anti–PD-1; BMS-936558; ONO-4538) versus investigator's choice chemotherapy (ICC) in patients with advanced melanoma with prior anti-CTLA-4 therapy. [Madrid]
    1. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, Hassel JC, Rutkowski P, McNeil C, Kalinka-Warzocha E. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320–330.
    1. Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, Vaishampayan UN, Drabkin HA, George S, Logan TF. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J Clin Oncol. 2015;33:1430–1437.
    1. Blank C, Kuball J, Voelkl S, Wiendl H, Becker B, Walter B, Majdic O, Gajewski TF, Theobald M, Andreesen R. Blockade of PD-L1 (B7-H1) augments human tumor-specific T cell responses in vitro. Int J Cancer. 2006;119:317–327.
    1. Brown JA, Dorfman DM, Ma FR, Sullivan EL, Munoz O, Wood CR, Greenfield EA, Freeman GJ. Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J Immunol. 2003;170:1257–1266.
    1. Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112:4371–4383.
    1. Afreen S, Dermime S. The immunoinhibitory B7-H1 molecule as a potential target in cancer: killing many birds with one stone. Hematol Oncol Stem Cell Ther. 2014;7:1–17.
    1. Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, Iwai Y, Long AJ, Brown JA, Nunes R. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261–268.
    1. Yao Y, Tao R, Wang X, Wang Y, Mao Y, Zhou LF. B7-H1 is correlated with malignancy-grade gliomas but is not expressed exclusively on tumor stem-like cells. Neuro Oncol. 2009;11:757–766.
    1. Wintterle S, Schreiner B, Mitsdoerffer M, Schneider D, Chen L, Meyermann R, Weller M, Wiendl H. Expression of the B7-related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. Cancer Res. 2003;63:7462–7467.
    1. Usui Y, Okunuki Y, Hattori T, Takeuchi M, Kezuka T, Goto H, Usui M. Expression of costimulatory molecules on human retinoblastoma cells Y-79: functional expression of CD40 and B7H1. Invest Ophthalmol Vis Sci. 2006;47:4607–4613.
    1. Routh JC, Ashley RA, Sebo TJ, Lohse CM, Husmann DA, Kramer SA, Kwon ED. B7-H1 expression in Wilms tumor: correlation with tumor biology and disease recurrence. J Urol. 2008;179:1954–1959. [discussion 1959–1960]
    1. Kim JR, Moon YJ, Kwon KS, Bae JS, Wagle S, Kim KM, Park HS, Lee H, Moon WS, Chung MJ. Tumor infiltrating PD1-positive lymphocytes and the expression of PD-L1 predict poor prognosis of soft tissue sarcomas. PLoS One. 2013;8:e82870.
    1. Lussier DM, Nieves LM, McAfee MS, Hyunh TP, Holechek S. Society for Immunotherapy of Cancer 28th Annual Meeting 2013. 2013. Enhancement of T cell immunity to osteosarcoma through modulation of PD-1/PD-L1 interactions.

Source: PubMed

3
Abonnere