Prognostic value and clinicopathological characteristics of PD-L1 overexpression in non-Hodgkin lymphoma: a meta-analysis

DOI: https://doi.org/10.21203/rs.2.16718/v1

Abstract

Abstract Background: Programmed cell death ligand 1 (PD-L1) has already been detected in various carcinomas. In non-Hodgkin lymphoma (NHL), however, the prognostic value of PD-L1 overexpression remains unclear. Methods: A meta-analysis of 2,321 NHL patients from 12 studies was performed. HRs with 95% CIs were used to evaluate the correlation between PD-L1 overexpression and prognosis of NHL, and odds ratios (ORs) with 95% CIs were used to assess the association of PD-L1 overexpression with clinicopathological factors. Results: The result showed that no significant difference between PD-L1 positive and negative groups was detected in NHL (hazard ratio [HR]: 1.40, 95% confidence interval [CI]: 0.90-2.19; P =0.137). Nevertheless, the result indicated that PD-L1 overexpression was associated with poor prognosis in the subtype of diffuse large B cell lymphoma (DLBCL) (HR: 1.70, 95%CI: 1.05-2.74; P =0.031). We also performed subgroup analyses and meta-regression. The pooled odds ratios(ORs) showed that PD-L1 overexpression was associated with B symptoms, higher international prognostic index (IPI) score (3, 4 and 5 points) and Ann Arbor Stage III and IV. Conclusions: T he meta-analysis demonstrated that PD-L1 expression was not associated with prognosis of NHL but associated with prognosis of DLBCL. Key words: PD-L1, non-Hodgkin lymphoma, prognosis, meta-analysis

Background

Non-Hodgkin lymphoma (NHL), accounting for approximately 90% of lymphomas and comprising various subtypes, is a common hematological tumor. NHL is characterized by a series of malignant DNA repair obstacle events and activating proto-oncogene caused by viral or bacterial infection, immune dysfunction and genetic factors, resulting in a wide-ranging histological appearances and clinical features at presentation, including painless lymphadenopathy, B symptoms (weight loss>10%, night sweats, body temperature >38°C), and so on.[1] The prognoses of NHL patients remain poor while the 5-year over survival (OS) has improved.[2, 3] Therefore, we posit that maybe there are other biomarkers potentially influencing the prognosis of NHL.

Programmed cell death ligand 1 (PD-L1), a 40kDa type 1 transmembrane protein, can activate B, T cells, macrophages, and dendritic cells.[4, 5] It was first found by Chen et al in 1999.[6] It was reported that PD-L1 co-stimulated T-cell proliferation and interleukin-10 secretion, which was considered to be involved in the negative regulation of cell-mediated immune responses.[6] Under normal physiological conditions, immune checkpoints maintain self-tolerance and protect tissues from damage when the immune system is responding to pathogenic infections.[7, 8] However, PD-L1, bound to programmed cell death 1 (PD-1), inhibits effector T cell function and activates immunosuppressive regulative T-cell function, resulting in tumors escaping under pathological conditions,[9-11] which is a major mechanism of tumor recurrence and drug resistance.[12] Moreover, clinical research inferred that patients who had overexpression of PD-L1 in tumors had improved clinical outcomes after taking checkpoint blockades.[13]

Cumulative studies showed that PD-L1 or PD-1 could be used to determine prognosis in various cancers, such as melanoma, non-small cell lung cancer, kidney cancer[5], and classic Hodgkin lymphoma.[14] Some studies have also assessed the prognostic value of PD-L1 overexpression in NHL. However, the results were quite different. Thus, we aim to identify the problem through performing the meta-analysis.

Methods

Our meta-analysis was conducted based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement.[15]

Literature search

Four databases—Pubmed, Cochrane Library, Web of Science, and Embase—were used to retrieve articles that investigated the prognostic value of PD-L1 overexpression in NHL. Additionally, we used the following terms for searches: “PD-L1,” “B7-H1,” “CD274,” “programmed cell death ligand 1,” “lymphoma,” “non-Hodgkin lymphoma,” “NHL,” “prognosis,” and “survival”. Articles published before January 2019 were included in the meta-analysis. We also performed a reference search.

Selection of studies

Two independent reviewers evaluated all potential articles. All candidate articles had to meet the following criteria:(1) patients’ NHL diagnoses were histologically confirmed; (2) PD-L1 expression in lymphoid tissue was detected using immunohistochemistry (IHC); (3) hazard ratios (HRs) and 95% confidence intervals (CIs) could be directly obtained from the studies or calculated using data from the articles; and (4) the studies were full-text and written in English. Moreover, any disputes were solved via discussion.

Data extraction and quality assessment

Two investigators independently extracted the data from articles. We extracted the following data: first author’s name, study country, publication year, subtype, sample size, cut-off value of PD-L1, HRs and 95% CIs for OS, PD-L1 positive number, follow-up period, treatment, Ann Arbor Stage and IHC antibodies. Furthermore, we contacted the author for original data if we were unable to calculate the effect size through the methods provided by Tierney.[16] We assessed these studies using the Newcastle–Ottawa Scale (NOS),[17] in which the score ranges from 0 to 9 points. We considered studies that received 6 points or above eligible for our meta-analysis. Any issues were resolved via discussion.

Statistical analysis

HRs with 95% CIs were used to evaluate the correlation between PD-L1 overexpression and prognosis of NHL, and odds ratios (ORs) with 95% CIs were used to assess the association of PD-L1 overexpression with clinicopathological factors. Heterogeneity tests were performed using the I-squared statistics, and an I2>50% was considered significant. If heterogeneity was significant, we chose a random effect model to compute the pooled HR; otherwise, we selected fixed effect model. Additionally, sensitivity analysis was used to assess the robustness of the pooled result, and publication bias was evaluated using Begg’s test. Subgroup analyses and meta-regression were performed due to significant heterogeneity. All the analyses were performed by STATA 12.0 software (STATA, College, TX, USA) and Revman 5.3 (Revman the Cochrane, Collaboration, Oxford, England).

Results

Literature screening and characteristics

The literature screening process is illustrated in Figure 1. A total of 328 articles from the four databases and two articles from manual reference search were initially selected. After removing duplications, 224 studies remained. We excluded 189 articles after reviewing article abstracts. Next, seven articles were removed for failing to calculate the effect size; 14 studies were eliminated due to conference abstract; and two studies were excluded because PD-L1 was not detected through IHC. Finally, altogether 12 articles encompassing 2,321 patients were selected for the meta-analysis.

All characteristics of studies are displayed in Table 1. Four studies were performed in China,[18-21] four in Korea,[22-25] two in Japan[26, 27], and one each was in the USA[28] and Norway[29], respectively. The cut-off value was determined using the form of percentage except Cho’s, which ranged from 2% to 50%. According to the cut-off values, every article described the number of patients with PD-L1 overexpression. All studies referred to each disease stage according to the Ann Arbor Stage except Bi’s. In addition, all studies were retrospective and reported the association between PD-L1 and OS. Patients in the studies had a histologically confirmed NHL diagnosis and subtype.

Association between PD-L1 overexpression and OS in NHL

We calculated a pooled HR of 1.40 (95% CI: 0.90-2.19; P=0.137) for OS. The result indicated that PD-L1 overexpression was not associated with NHL prognosis. Significant heterogeneity, however, existed among the selected studies (I2=70.6%, P<0.001; Figure 2).

Association of PD-L1 overexpression with OS in DLBCL

DLBCL, accounting for 30-40% of NHL, is the most common subtype of NHL. There were 863 DLBCL patients from six articles in our study. A meta-analysis was performed that was designed to assess prognosis among DLBCL patients. The result showed that the pooled HR was 1.70 (95% CI: 1.05-2.74; P=0.031) with I2=47.2% (Figure 3). This indicated that PD-L1 overexpression potentially predicted a poor prognosis in DLBCL patients.

Association between PD-L1 overexpression and clinicopathological characteristics

We also investigated the association of PD-L1 overexpression with clinicopathological characteristics. The results suggested that PD-L1 overexpression was more frequent in patients with B symptoms (OR=1.91, 95% CI: 1.17-3.10; P=0.09), stage III and IV (OR=1.49, 95% CI: 1.09-2.04; P=0.01) and international prognostic index (IPI) score of 3 to 5 points (OR=1.79, 95% CI: 1.26-2.56; P=0.001). However, there was no significant difference in the subgroups of gender and age (Figure 4).

Subgroup and sensitivity analysis

Subgroup analyses were conducted by tumor type, country, sample size, cut-off value, therapy, antibody source, and type. Subgroup analysis by country showed HR of 2.86 (95% CI: 1.44-5.66; P=0.003) in China, 1.99 (95% CI: 1.29-3.08; P=0.002) in Japan, and 0.47 (95% CI: 0.29-0.77; P=.002) in Korea. In addition, when cut-off value ≥30%, HR was 2.54 (95% CI: 1.56-4.12; P<0.001) with I2=37% (Table 2). Sensitivity analyses demonstrated that our pooled results were robust even when omitting anyone of the included studies by turn in NHL and DLBCL (Figures 5 and 6).

Meta-regression analysis

Furthermore, meta-regression was performed for the source of heterogeneity in NHL. The results showed that sample size (P=0.638), treatment (P=0.229), location (P=0.107), tumor type (P=0.916), and cut-off value (P=0.058) did not contribute to the heterogeneity.

Publication bias

Begg’s test was used to assess the publication bias, which revealed no publication bias for both NHL (P=0.88) and DLBCL (P=0.92).

Discussion

This is a meta-analysis designed to investigate the relationship between PD-L1 overexpression and the prognosis of NHL. The association of PD-L1 overexpression with some clinicopathological factors was also evaluated. The pooled HR of 1.40 (95% CI: 0.90-2.19; P=0.137) was calculated for 2,321 patients from 12 studies, potentially indicating no significant correlation between PD-L1 and NHL prognosis. Nevertheless, the result suggested that PD-L1 overexpression was associated with poor prognosis in DLBCL patients. Figure 4 illustrated that patients with B symptoms, IPI score of 3 to 5 points and stage III or IV possessed overexpression of PD-L1.

Subgroup analysis and meta-regression showed no contribution to the heterogeneity in NHL. However, perhaps some problems contribute to the heterogeneity. Although IHC was used to detect PD-L1 protein in tumor cells in all studies, different studies adopted different procedures,[30] antibody clones and thresholds.[31] Vranic et al[32] suggested that anti-PD-L1 clones SP142 and SP263 exhibit an excellent concordance. Additionally, other confounding factors influence the expression of PD-L1. Studies[33, 34] indicated that anaplastic Lymphoma kinase (ALK) up-regulates PD-L1 expression. Research also suggested that STAT3 regulates PD-L1 expression, and it was demonstrated that the inhibitor of STAT3 abrogated the expression of PD-L1.[35, 36] Additionally, it was shown that tumor cells that overexpress PD-L1 protein have been frequently detected in EBV-positive lymphomas.[20, 26, 37, 38]

The response to treatment is also not associated with the level of PD-L1 expression. Currently, PD-1 blockades are mostly employed clinically. Some clinical trials[39, 40] showed that patients with B-cell NHL indeed responded well to PD-1 blockades combined with rituximab. Zinzani et al.[41] found that PD-1 blockades used alone also benefited B-cell NHL patients. Two studies[42, 43] showed that PD-1 blockades helped relapsed or refractory NHL patients increase complete response rate. However, the level of PD-L1 expression in patients was quite different, and PD-L1was not even detected in some patients. These findings indicate that the level of PD-L1 expression is not associated with the prognosis of NHL patients.

Nevertheless, recent studies have uncovered the concrete functional mechanism of PD-L1 in DLBCL. PD-L1, bound to PD-1, caused Akt phosphorylated, which urge m-TOR to activate its downstream molecules, such as P43-BP1 and P-P70S6K, finally resulting in proliferation and progression of malignant cells.[19, 44, 45] Theoretically, this explains why overexpression of PD-L1 causes short OS in DLBCL patients. Unfortunately, in other NHL subtypes, there is currently no such theory.

To the best of our knowledge, Zhao S et al [46]had already done the first meta-analysis including 9 studies to explore the relationship between PD-L1 overexpression and prognosis in NHL patients and concluded that PD-L1 overexpression has the association with poor prognosis in NHL and DLBCL but not with natural killer/T-cell (NK/T) lymphoma. We brought 12 studies including 2,321 patients into our meta-analysis and obtained conclusions that are different from Zhao’s. In DLBCL and NK/T lymphoma, we and Zhao S et al reach the same conclusion. Yet, in overall result of NHL, our conclusion is different from Zhao’s due to more three included studies. We also adopted two tools to conduct meta-analysis and did sub-analysis. Several limitations, however, must be considered in interpreting our findings. First, the total sample size of the included studies was small. Second, other clinicopathological factors—such as EBV infection, tumor size, central neutral system invasion—were not included in the analysis due to insufficient materials. Third, although we performed subgroup analysis by cut-off value, we did not know the best cut-off value for stratification of NHL patients in clinical management.

Conclusions

In conclusion, our pooled result showed that overexpression of PD-L1 was not associated with OS in NHL patients, but actually associated with the subtype of DLBCL, indicating that PD-L1 could perhaps predict prognosis of DLBCL. Furthermore, PD-L1 overexpression was associated with clinicopathological factors of B symptoms, IPI score, and Ann Arbor Stage. Nevertheless, studies on other specific NHL subtypes using standardized immunological tests are needed to further explore the relationship between PD-L1 overexpression and prognosis of NHL.

Abbreviations

Programmed cell death ligand 1: PD-L1

non-Hodgkin lymphoma: NHL

odds ratio: OR

hazard ratio: HR

confidence interval: CI

diffuse large B cell lymphoma: DLBCL

international prognostic index : IPI

programmed cell death 1: PD-1

Preferred Reporting Items for Systematic Reviews and Meta-Analyses: PRISMA

Immunohistochemistry: IHC

Newcastle–Ottawa Scale: NOS

Declarations

Ethical approval and consent to participate

This article does not contain samples and data of human participants or animals. All the data involved were obtained from published articles. Informed consent was obtained from all included participants in the study.

Consent for publication

Not applicable.

Availability of data and materials

Not applicable.

Competing interest

No potential conflicts of interest are disclosed.

Author Contributions

The study was conceived, designed and performed by QZ. QZ analyzed the data and ZL contributed to the materials and tools. QZ wrote this paper. ZL retrieved all the text articles. All the work was performed under TL’s instructions. All authors have read and approved the manuscript, and ensure that this is the case.

Acknowledgment

Not applicable.

Funding

This work was not financed by any grants

References

  1. Shankland KR, Armitage JO, Hancock BW: Non-Hodgkin lymphoma. Lancet (London, England) 2012, 380(9844):848-857.
  2. Kubuschok B, Held G, Pfreundschuh M: Management of diffuse large B-cell lymphoma (DLBCL). Cancer treatment and research 2015, 165:271-288.
  3. Al-Hamadani M, Habermann TM, Cerhan JR, Macon WR, Maurer MJ, Go RS: Non-Hodgkin lymphoma subtype distribution, geodemographic patterns, and survival in the US: A longitudinal analysis of the National Cancer Data Base from 1998 to 2011. 2015, 90(9):790-795.
  4. Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X: Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends in molecular medicine 2015, 21(1):24-33.
  5. Li Y, Li F, Jiang F, Lv X, Zhang R, Lu A, Zhang G: A Mini-Review for Cancer Immunotherapy: Molecular Understanding of PD-1/PD-L1 Pathway &amp; Translational Blockade of Immune Checkpoints. International journal of molecular sciences 2016, 17(7).
  6. Dong H, Zhu G, Tamada K, Chen L: B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nature medicine 1999, 5(12):1365-1369.
  7. Pardoll DM: The blockade of immune checkpoints in cancer immunotherapy. Nature reviews Cancer 2012, 12(4):252-264.
  8. Chakravarti N, Prieto VG: Predictive factors of activity of anti-programmed death-1/programmed death ligand-1 drugs: immunohistochemistry analysis. Translational lung cancer research 2015, 4(6):743-751.
  9. Francisco LM, Sage PT, Sharpe AH: The PD-1 pathway in tolerance and autoimmunity. Immunological reviews 2010, 236:219-242.
  10. Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Kuchroo VK, Sharpe AH: PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. The Journal of experimental medicine 2009, 206(13):3015-3029.
  11. Haxhinasto S, Mathis D, Benoist C: The AKT-mTOR axis regulates de novo differentiation of CD4+Foxp3+ cells. The Journal of experimental medicine 2008, 205(3):565-574.
  12. Finn OJ: Immuno-oncology: understanding the function and dysfunction of the immune system in cancer. Annals of oncology : official journal of the European Society for Medical Oncology 2012, 23 Suppl 8:viii6-9.
  13. Patel SP, Kurzrock R: PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy. Molecular cancer therapeutics 2015, 14(4):847-856.
  14. Pembrolizumab Approved for Hodgkin Lymphoma. Cancer discovery 2017, 7(5):Of1.
  15. Moher D, Liberati A, Tetzlaff J, Altman DG: Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS medicine 2009, 6(7):e1000097.
  16. Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR: Practical methods for incorporating summary time-to-event data into meta-analysis. Trials 2007, 8:16.
  17. Stang A: Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. European journal of epidemiology 2010, 25(9):603-605.
  18. Fang X, Xiu B, Yang Z, Qiu W, Zhang L, Zhang S, Wu Y, Zhu X, Chen X, Xie S et al: The expression and clinical relevance of PD-1, PD-L1, and TP63 in patients with diffuse large B-cell lymphoma. Medicine 2017, 96(15):e6398.
  19. Dong L, Lv H, Li W, Song Z, Li L, Zhou S, Qiu L, Qian Z, Liu X, Feng L et al: Co-expression of PD-L1 and p-AKT is associated with poor prognosis in diffuse large B-cell lymphoma via PD-1/PD-L1 axis activating intracellular AKT/mTOR pathway in tumor cells. Oncotarget 2016, 7(22):33350-33362.
  20. Bi XW, Wang H, Zhang WW, Wang JH, Liu WJ, Xia ZJ, Huang HQ, Jiang WQ, Zhang YJ, Wang L: PD-L1 is upregulated by EBV-driven LMP1 through NF-kappaB pathway and correlates with poor prognosis in natural killer/T-cell lymphoma. Journal of hematology & oncology 2016, 9(1):109.
  21. Hu LY, Xu XL, Rao HL, Chen J, Lai RC, Huang HQ, Jiang WQ, Lin TY, Xia ZJ, Cai QQ: Expression and clinical value of programmed cell death-ligand 1 (PD-L1) in diffuse large B cell lymphoma: a retrospective study. 2017, 36(1):94.
  22. Kwon D, Kim S, Kim PJ, Go H, Nam SJ, Paik JH, Kim YA, Kim TM, Heo DS, Kim CW et al: Clinicopathological analysis of programmed cell death 1 and programmed cell death ligand 1 expression in the tumour microenvironments of diffuse large B cell lymphomas. Histopathology 2016, 68(7):1079-1089.
  23. Kim WY, Jung HY, Nam SJ, Kim TM, Heo DS, Kim CW, Jeon YK: Expression of programmed cell death ligand 1 (PD-L1) in advanced stage EBV-associated extranodal NK/T cell lymphoma is associated with better prognosis. 2016, 469(5):581-590.
  24. Jo JC, Kim M, Choi Y, Kim HJ, Kim JE, Chae SW, Kim H, Cha HJ: Expression of programmed cell death 1 and programmed cell death ligand 1 in extranodal NK/T-cell lymphoma, nasal type. Annals of hematology 2017, 96(1):25-31.
  25. Cho H, Kim SH, Kim SJ, Chang JH, Yang WI, Suh CO, Kim YR, Jang JE, Cheong JW, Min YH et al: Programmed cell death 1 expression is associated with inferior survival in patients with primary central nervous system lymphoma. Oncotarget 2017, 8(50):87317-87328.
  26. Kiyasu J, Miyoshi H, Hirata A, Arakawa F, Ichikawa A, Niino D, Sugita Y, Yufu Y, Choi I, Abe Y et al: Expression of programmed cell death ligand 1 is associated with poor overall survival in patients with diffuse large B-cell lymphoma. Blood 2015, 126(19):2193-2201.
  27. Miyoshi H, Kiyasu J, Kato T, Yoshida N, Shimono J, Yokoyama S, Taniguchi H, Sasaki Y, Kurita D, Kawamoto K et al: PD-L1 expression on neoplastic or stromal cells is respectively a poor or good prognostic factor for adult T-cell leukemia/lymphoma. Blood 2016, 128(10):1374-1381.
  28. Xing W, Dresser K, Zhang R, Evens AM, Yu H, Woda BA, Chen BJ: PD-L1 expression in EBV-negative diffuse large B-cell lymphoma: clinicopathologic features and prognostic implications. Oncotarget 2016, 7(37):59976-59986.
  29. Blaker YN, Spetalen S, Brodtkorb M, Lingjaerde OC, Beiske K, Ostenstad B, Sander B, Wahlin BE, Melen CM, Myklebust JH et al: The tumour microenvironment influences survival and time to transformation in follicular lymphoma in the rituximab era. British journal of haematology 2016, 175(1):102-114.
  30. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN et al: Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515(7528):563-567.
  31. Scognamiglio G, De Chiara A, Di Bonito M, Tatangelo F, Losito NS, Anniciello A, De Cecio R, D'Alterio C, Scala S, Cantile M et al: Variability in Immunohistochemical Detection of Programmed Death Ligand 1 (PD-L1) in Cancer Tissue Types. International journal of molecular sciences 2016, 17(5).
  32. Vranic S, Ghosh N, Kimbrough J, Bilalovic N, Bender R, Arguello D, Veloso Y, Dizdarevic A, Gatalica Z: PD-L1 Status in Refractory Lymphomas. PloS one 2016, 11(11):e0166266.
  33. Roussel H, De Guillebon E, Biard L, Mandavit M, Gibault L, Fabre E, Antoine M, Hofman P, Beau-Faller M, Blons H et al: Composite biomarkers defined by multiparametric immunofluorescence analysis identify ALK-positive adenocarcinoma as a potential target for immunotherapy. Oncoimmunology 2017, 6(4):e1286437.
  34. Cha YJ, Shim HS: PD-L1 expression and CD8+ tumor-infiltrating lymphocytes are associated with ALK rearrangement and clinicopathological features in inflammatory myofibroblastic tumors. Oncotarget 2017, 8(52):89465-89474.
  35. Atsaves V, Tsesmetzis N, Chioureas D, Kis L, Leventaki V, Drakos E, Panaretakis T, Grander D, Medeiros LJ, Young KH et al: PD-L1 is commonly expressed and transcriptionally regulated by STAT3 and MYC in ALK-negative anaplastic large-cell lymphoma. Leukemia 2017, 31(7):1633-1637.
  36. Ma C, Horlad H, Pan C, Yano H, Ohnishi K, Fujiwara Y, Matsuoka M, Lee A, Niidome T, Yamanaka R et al: Stat3 inhibitor abrogates the expression of PD-1 ligands on lymphoma cell lines. Journal of clinical and experimental hematopathology : JCEH 2017, 57(1):21-25.
  37. Chen BJ, Chapuy B, Ouyang J, Sun HH, Roemer MG, Xu ML, Yu H, Fletcher CD, Freeman GJ, Shipp MA et al: PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clinical cancer research : an official journal of the American Association for Cancer Research 2013, 19(13):3462-3473.
  38. Laurent C, Fabiani B, Do C, Tchernonog E, Cartron G, Gravelle P, Amara N, Malot S, Palisoc MM, Copie-Bergman C et al: Immune-checkpoint expression in Epstein-Barr virus positive and negative plasmablastic lymphoma: a clinical and pathological study in 82 patients. Haematologica 2016, 101(8):976-984.
  39. Berger R, Rotem-Yehudar R, Slama G, Landes S, Kneller A, Leiba M, Koren-Michowitz M, Shimoni A, Nagler A: Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clinical cancer research : an official journal of the American Association for Cancer Research 2008, 14(10):3044-3051.
  40. Westin JR, Chu F, Zhang M, Fayad LE, Kwak LW, Fowler N, Romaguera J, Hagemeister F, Fanale M, Samaniego F et al: Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial. The Lancet Oncology 2014, 15(1):69-77.
  41. Zinzani PL, Ribrag V, Moskowitz CH, Michot JM, Kuruvilla J, Balakumaran A, Zhang Y, Chlosta S, Shipp MA, Armand P: Safety and tolerability of pembrolizumab in patients with relapsed/refractory primary mediastinal large B-cell lymphoma. Blood 2017, 130(3):267-270.
  42. Lesokhin AM, Ansell SM, Armand P, Scott EC, Halwani A, Gutierrez M, Millenson MM, Cohen AD, Schuster SJ, Lebovic D et al: Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2016, 34(23):2698-2704.
  43. Kwong YL, Chan TSY, Tan D, Kim SJ, Poon LM, Mow B, Khong PL, Loong F, Au-Yeung R, Iqbal J et al: PD1 blockade with pembrolizumab is highly effective in relapsed or refractory NK/T-cell lymphoma failing l-asparaginase. Blood 2017, 129(17):2437-2442.
  44. Uddin S, Hussain AR, Siraj AK, Manogaran PS, Al-Jomah NA, Moorji A, Atizado V, Al-Dayel F, Belgaumi A, El-Solh H et al: Role of phosphatidylinositol 3'-kinase/AKT pathway in diffuse large B-cell lymphoma survival. Blood 2006, 108(13):4178-4186.
  45. Xu ZZ, Xia ZG, Wang AH, Wang WF, Liu ZY, Chen LY, Li JM: Activation of the PI3K/AKT/mTOR pathway in diffuse large B cell lymphoma: clinical significance and inhibitory effect of rituximab. Annals of hematology 2013, 92(10):1351-1358.
  46. Zhao S, Zhang M, Zhang Y, Meng H, Wang Y, Liu Y, Jing J, Huang L, Sun M, Zhang Y et al: The prognostic value of programmed cell death ligand 1 expression in non-Hodgkin lymphoma: a meta-analysis. Cancer biology & medicine 2018, 15(3):290-298.

 

Tables

Table 1. Characteristics of studies

Study

Year

Sample size

Country

Tumor  type

Median follow-up

(range) (month)

Therapy

Stage

NOS

Cut-off

PD-L1+

Number

Antibody

Company

Source

Type

Clone

Kiyasu

2015

1253

Japan

DLBCL

NA

C+T+R

I-IV

7

30%

461

abcam, UK

mouse

MAB

ab52587

Xing

2016

86

USA

DLBCL

21(0.07-175)

C

I-IV

6

30%

14

Cell Signaling,USA

rabbit

MAB

E1L3N

Dong

2016

100

China

DLBCL

52.4(1.5-89.1)

C

I-IV

7

5%

54

abcam,UK

rabbit

PAB

ab153991

Bi

2016

77

China

NK/T

38.0(9.4-79.0)

C

I-II

8

38%

26

abcam,UK

rabbit

PAB

NA

Kim

2016

73

Korea

NK/T

20.6(0.2-83.2)

C+S

I-IV

7

10%

41

Cell Signaling,USA

rabbit

MAB

E1L3N

Fang

2017

74

China

DLBCL

2.4-86.4

C+S

I-IV

8

10%

20

ZSGB-BIO

rabbit

MAB

SP142

Kwon

2015

126

Korea

DLBCL

52(16-165)

C

I-IV

8

10%

77

Cell Signaling,USA

rabbit

MAB

E1L3N

Blaker

2016

38

Norway

FL

120(15.6-408)

C+T

NA

6

2%

15

Spring Bioscience,

Pleasanton,CA,USA

rabbit

MAB

SP142

Jo

2016

79

Korea

NK/T

52.4

C+R

I-IV

7

5%

63

R&D Systems,USA

mouse

MAB

NA

Hu

2017

204

China

DLBCL

52 (1-114)

C

I-IV

8

5%

100

Cell Signaling, USA

rabbit

MAB

NA

Cho

2017

76

Korea

PCNSL

20.2 (2.2-128.5)

C+T

NA

6

≥100 cells/HPF

10

Abcam, UK

rabbit

PAB

ab58810

Miyoshi

2016

135

Japan

ATLL

10.9 (0.03-114.8)

C+T+R

I-IV

8

50%

10

Abcam, UK

rabbit

MAB

ab174838

DLBCL: diffuse large B cell lymphoma; NK/T: NK/T cell lymphoma; FL: follicular lymphoma; PCNSL: primary central nervous system lymphoma; ATLL: adult T cell lymphoma/leukemia; C: Chemotherapy; T: Transplantation; R: Radiotherapy; S: Surgery; NOS: Newcastle–Ottawa Scale; MAB: monoclonal antibody; PAB: polyclonal antibody; NA: not applicable. NA: not applicable.

 

Table 2. Subgroup analysis for OS 

Subgroup

number of

studies

number of

patients

HR(95% CI)

P value

Heterogeneity

Location

China

4

455

2.86(1.44-5.66)

0.003

I2=45.1%; P=.141

Korea

4

354

0.47(0.29-0.77)

0.002

I2=0%; P=.836

USA

1

86

2.42(1.03-5.67)

0.042

/

Norway

1

38

1.08(0.64-1.81)

0.771

/

Japan

2

1,388

1.99(1.29-3.08)

0.002

I2=0%; P=.557

Cut-off value

≥30%

4

1,627

2.54(1.56-4.12)

<0.001

I2=37%; P=.19

≤10%

7

694

0.98(0.55-1.73)

0.938

I2=68.7%; P=.004

Tumor type

DLBCL

6

1,842

1.70(1.05-2.74)

0.031

I2=47.2%; P=.092

NK/T

3

229

1.07(0.21-5.59)

0.935

I2=89.3%; P<.001

FL

1

38

1.08(0.64-1.81)

0.771

/

PCNSL

1

76

0.84(0.20-3.55)

0.813

/

ATLL

1

136

2.37(1.15-4.90)

0.020

/

Therapy

Chemotherapy

5

1,573

2.16(0.85-5.49)

0.105

I2=73.6%; P=.004

Chemotherapy

+other treatments

7

748

1.12(0.69-1.84)

0.646

I2=68.5%; P=.004

Sample size

≥100

5

1,818

1.64(0.90-3.01)

0.529

I2=57.8%; P=.05

<100

7

503

1.26(0.66-2.43)

0.480

I2=76.8%; P<.001

Antibody type

MAB

9

2,068

1.17(0.75-1.83)

0.476

I2=68.4%; P=.001

PAB

3

253

3.23(0.89-11.74)

0.075

I2=64%; P=.062

Antibody source

Rabbit

10

989

1.52(0.91-2.55)

0.212

I2=70.6%; P<.001

Mouse

2

1,332

0.98(0.27-3.52)

0.978

I2=84.5%; P=.011

HR: hazard ratio; CI: confidence interval; DLBCL: diffuse large B cell lymphoma; NK/T: NK/T cell lymphoma; FL: follicular lymphoma; PCNSL: primary central nervous system lymphoma; ATLL: adult T cell lymphoma/leukemia; MAB: monoclonal antibody; PAB: polyclonal antibody.