Next Article in Journal
Clinical and Analytical Comparison of Monoclonal and Polyclonal Immunoassays for Fecal Pancreatic Elastase
Previous Article in Journal
Private Doctors’ Perspective towards “Patient First” in TB Diagnostic Cascade, Hisar, India
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Is There an Immunohistochemical PD-L1 Cut-Off Point That Serves as a Prognostic Indicator for Large B-Cell Lymphomas? †

1
Department of Pathology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, 34098 Istanbul, Turkey
2
Division of Hematology, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, 34320 Istanbul, Turkey
*
Author to whom correspondence should be addressed.
This study was presented as oral presentation at the 30th National Congress of Pathology, online, 20–23 May 2021.
Current address: Department of Pathology, University of Health Sciences, Bagcılar Training and Research Hospital, 34212 Istanbul, Turkey.
Diagnostics 2024, 14(11), 1167; https://doi.org/10.3390/diagnostics14111167
Submission received: 21 April 2024 / Revised: 15 May 2024 / Accepted: 26 May 2024 / Published: 31 May 2024
(This article belongs to the Special Issue New Promising Diagnostic Signatures in Histopathological Diagnosis)

Abstract

:
The aim of this study is to investigate whether there is a cut-off value for PD-L1 expression in large B-cell lymphomas that predicts prognosis, and to clarify the relationship between PD-L1 expression and histopathological as well as clinical parameters. The study included a total of 130 patients who were diagnosed with large B-cell lymphoma at Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Pathology Department. Biopsy samples were assessed using the PD-L1 immunohistochemical antibody (Dako, 22C3 clone). The patients had a mean age of 54 ± 17 years, with a median age of 56 years. No statistically significant difference was observed between the groups in terms of survival when the 30% cut-off value was used. However, a noteworthy discrepancy in survival became apparent when the cut-off point was established at 70%. Among the diffuse large B-cell lymphoma-not otherwise specified (DLBCL-NOS) category, the activated B-cell-like (ABC-like) phenotype showed higher PD-L1 expression compared to the germinal center B-cell-like (GCB-like) phenotype. Immunohistochemical PD-L1 expression emerged as a prognostic factor, particularly significant in the ABC-like phenotype.

1. Introduction

Diffuse large B-cell lymphomas (DLBCLs) are the most common aggressive lymphomas among non-Hodgkin’s lymphomas, and account for about 30% of non-Hodgkin’s lymphomas [1,2,3]. Following FDA approval of rituximab in 1997, an anti-CD20 monoclonal antibody, and its inclusion in the standard treatment of DLBCL patients, a cure was attained in around 60% of these cases [4]. Apart from conventional treatments, recently developed immunotherapy can play an important role in the treatment of these patients [4,5,6]. After the discovery of PD-1 in 1992, and research on immune checkpoint regulation has led to the development of various immunotherapy agents, which have found utility in the treatment of various solid cancers. In this context, ’pembrolizumab’ was approved by the FDA for the treatment of classical Hodgkin’s lymphomas in 2016, and primary mediastinal large B-cell lymphoma in 2017 [7,8].
It is understood that the PD-1/PD-L1 pathway regulates the T-cell response to maintain autoimmunity and protect the organism from immune-mediated damage [9,10,11]. Although the PD1/PDL1-2 pathway plays an important role in the development of immune tolerance and the prevention of autoimmune diseases, extended exposure to antigens such as cancer or viral infections leads to the loss of CD8+ cytotoxic T-cell functions (T-cell exhaustion) and subsequent immune system inadequacy [12]. PD-L1, generated by tumor cells, restricts the immune response of the host by binding to PD-1 on the surface of T cells, which inhibits the release of cytokines including interleukin 2 (IL2), tumor necrosis factor alpha (TNFα), and interferon gamma (IFN γ). Consequently, such tumors elude the immune system [12,13]. On the other hand, Treg cells, a subtype of CD4+ T cells, contribute to the establishment of an immunosuppressive environment through the release of inhibitory cytokines such as interleukin 10 (IL-10), transforming growth factor beta by promoting effector T-cell apoptosis via granzyme and perforin, or by leading the depletion of IL2 in the environment via CD25 [14].
In various studies, genetic alterations associated with PD-L1 expression have been identified in around 20% of DLBCL cases [14,15]. This study investigates the potential relationship between immunohistochemical PD-L1 expression, the histopathological and clinical parameters, as well as the prognosis of large B-cell lymphoma (LBCL) cases.

2. Materials and Methods

2.1. Patients and Clinicopathological Parameters

After grouping all slides and blocks pertaining to 159 patients sourced from our archives who were diagnosed with LBCL between 2011 and 2019, they were re-evaluated. Among cases with different protocol numbers for the same patient, the most suitable one for the study (considering factors like tissue quantity and presence of artifacts) was chosen. In cases where bone marrow biopsies were available for the same patient, bone marrow biopsy was not preferred due to routine formic acid decalcification before formalin fixation. Twenty-four of the selected cases had insufficient tissue in their blocks, and in the H&E-stained sections of five cases, artifacts such as crushing or freezing were present, resulting in their exclusion from the study. Finally, a total of 130 patients diagnosed with nodal or extranodal LBCL at Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty Pathology Department, and followed by the Internal Medicine Hematology department, were included in the study. The clinical data for the cases in the study were obtained from pathology reports, patient follow-up files in the hematology department, and the hospital automation system. Survival time was calculated based on the time from first diagnosis date to the death date for deceased patients, and to 1 November 2019 for living patients.

2.2. Histopathological and Immunohistochemical Evaluation

The obtained sections with 2 μm thickness were deparaffinized and rehydrated. Then, the samples were incubated with anti-PD-L1 antibody (for 60 min incubation; 1:50 dilution, monoclonal mouse anti-human PD-L1 clone 22C3, and pharmDx antibody [Dako, Carpinteria, CA, USA]), using a semi-automatic device (Dako Autostainer Link48 and Dako PT link). In the control tissue sections taken from the tonsil, membranous staining was observed in lymphoid cells within the crypt epithelium and, to a lesser extent, in the follicles, which was considered as a positive control. The immunohistochemically PD-L1-stained slides were examined and compared with H&E slides under a light microscope (Figure 1a–c). We identified five hot spot areas where tumor cells showed complete or partial membranous staining at ×400 magnification. These areas were photographed digitally, and the photographs were divided into four equal parts using a digital ruler. The number of tumor cells in each part was counted manually, along with H&E sections, and in some cases with previously applied immunohistochemical slides (CD20, CD3, CD10, BCL2, BCL6). After evaluation of all tumor cells in these regions, regardless of whether they had PD-L1 staining or not, this process was repeated for five different areas. The PD-L1 score for the tumor (tPD-L1) was calculated by determining the ratio of PD-L1 (+) tumor cells to the total number of tumor cells (Figure 1d–f).

2.3. Statistical Analysis

Statistical assessments were conducted using 2 distinct scoring systems. In the first of these, tumor cells were categorized into three groups: those stained below 1%, those stained less than 50% above 1%, and those stained at 50% and above. Apart from this classification, a cut-off value of 30% was established, and statistical analyses were conducted based on this threshold. For an effect size of 0.5, a significance level of 0.05, and a power of 0.80, the minimum total sample size was calculated as 128. In the statistical comparison, the chi-square test, Fisher exact test, and likelihood ratio were used for categorical data. The Fisher exact and likelihood ratio test was used when the expected value of any of the evaluated event tables was <5. Survival was assessed using Kaplan–Meier analysis, and a log-rank (Mantel–Cox) analysis was performed to compare the survival rates among the groups.
For determining statistical significance, a p-value of less than 0.05 was considered significant within the 95% confidence interval. Statistical analysis was carried out using the Statistical Package for the Social Sciences (SPSS Inc, Chicago, IL, USA) program, version 21.0.

3. Results

Out of the cases, 70 were male (54%) and 60 were female (46%). The patients’ average age was 54 ± 17 years, with an age range of 18–87 and a median age of 56 years. Among the 130 cases included in our study, 59 (45.4%) were the DLBCL-NOS, ABC-like phenotype; 50 (38.5%) were the DLBCL-NOS, GCB-like phenotype; and 4 (3.1%) were DLBCL-NOS unclassified. Seventeen of cases consisted of special types of large B-cell lymphomas. The clinical characteristics and PD-L1 expressions of the cases are summarized in Table 1.
In this study, the tPD-L1 expression was calculated as 19.2%, using the cut-off value of 30%. (Figure 2 shows distribution of cases according to tPD-L1 ratio). The 5-year survival rate was found to be 69.3% for tPD-L1 < 30% cases, and 60.2% for tPD-L1 ≥ 30% cases. The mean survival time was statistically determined as 71.7 ± 3.7 (64.5–78.9) months in the tPD-L1 < 30% group and 50.4 ± 6.7 (37.2–63.5) months in the tPD-L1 ≥ 30% group. There was no significant difference between the groups in terms of survival (log-rank p: 0.261).
There were no significant differences between the groups in terms of gender, age, and B symptoms at 30% cut-off value (p > 0.05). At the 30% cut-off point, there was no statistically significant relationship between PD-L1 positivity and clinical parameters such as LDH levels, ECOG performance score, and number of extra nodal involvements (p > 0.05). When the cases were evaluated in two groups according to the Ann Arbor stage, a significant difference in PD-L1 expression was observed between the two groups (p: 0.032) (Table 2).
The study investigated the association between PD-L1 expression in the tumor and survival outcomes across different cut-off values: 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%. The survival rate was significantly decreased for cut-off values of 70% and above (p: 0.039). The 5-year survival rates were determined as 71.1% in patients with a cut-off value of tPD-L1 expression < 70%, and 48% in patients with a cut-off value of tPD-L1 expression ≥ 70%; the mean survival times were 72.14 ± 3.50 (65.2–79.01) months and 41.2 ± 8.8 (23.8–58.6) months, respectively. A significant difference in terms of survival was observed between the two groups (Figure 3).
There were no significant differences between the groups in terms of gender, age, and B symptoms at the 70% cut-off value (p > 0.05) (Table 3).
When the cases were divided into three groups as <1%, 1–49%, and ≥50%, there was no statistically significant difference between tPD-L1 staining and age, gender, LDH level, ECOG performance score, extra nodal involvement, and Ann Arbor stage (Table 4).
The variables included in the univariate analyses were age, B symptom status, LDH level, ECOG performance score, Ann Arbor stage groups, IPI score, and tPD-L1 70% cut-off value. These variables were subjected to Cox regression analysis (Cox regression squared value: 29.488; p: 0.001). Notably, tPD-L1 ≥ 70% was identified as a significant prognostic factor (Table 5).

4. Discussion

An extensive search through the literature indicates that PD-L1 antibodies have found their place in the treatment of various solid tumor types across different regions, including gastrointestinal tract, breast, and kidney carcinomas [7,16]. Numerous studies have also investigated the PD1/PD-L1 pathway in LBCLs. Upon reviewing the studies, it becomes evident that defining a precise cut-off value is not achievable. In a study conducted by Kwon D. et al. in 2015, a cut-off value of 10% was established for PD-L1 positivity, while in another study by Kiyasu et al., it was set at 30% [6,17]. Moreover, various other cut-off values, such as 5%, 25%, and 50%, have been employed in different studies.
In our study, we found that the proportion of cases exhibiting PD-L1 positivity at a cut-off value of 30% was 19.2%. It is worth noting that this ratio has been reported to vary widely in the literature, ranging from 10.5% to 61.1% [6,17,18,19,20,21,22,23]. In other studies that used the 30% cut-off value, this ratio has been reported as 10.5%, 15.6%, 16.8%, 16%, and 21%, and our findings were consistent with those reported in the literature [6,18,19,22,23]. However, differences in factors such as the PD-L1 antibody clone used, automated counting in either a light microscope or a digital environment, and interobserver variability can be considered as potential reasons for the variations observed. Furthermore, when assessing PD-L1-positive cells within the tumor, various criteria are applied. Some authors consider only membranous staining [24,25], while others deem membranous and/or cytoplasmic staining as positive [6,17,20]. In certain studies, staining intensity is used for evaluation [21,22,23], whereas in others, including our study, the focus is solely on whether tumor cells are stained or not [18,24,25]. In LBCL, cells such as histiocytes may sometimes exhibit PD-L1 staining, leading to potential misinterpretations. As a result, some researchers have embraced the utilization of ‘combined PD-L1 positivity’ (CPS). On the other hand, dual staining with PD-L1 and PAX-5 antibodies, serving as a B-cell indicator together, as demonstrated in Kiyasu et al.‘s study, can help reduce false positive results [6]. Nevertheless, in routine practice, assessing dual staining requires considerations of cost effectiveness and benefits. Due to these technical differences, reaching a consensus on PD-L1 expression appears challenging. However, as more studies are conducted, it may become possible to obtain more accurate results by standardizing the methodologies.
Varying results have been reported in studies investigating the relationship between tPD-L1 positivity and survival in large B-cell lymphomas. While some studies have linked tPD-L1 positivity to a poor prognosis and low overall survival, others have found no such correlation. In 2015, Kwon D. and colleagues conducted a study with 126 patients using a cut-off value of 10%. They observed that in patients treated with R-CHOP, PD-L1 expression in the tumor had no significant prognostic impact [17]. On the other hand, Kiyasu and colleagues, in a study involving 1253 patients with a cut-off value of 30%, demonstrated that tPD-L1 expression served as an independent prognostic factor for overall survival [6]. In another study with 76 patients, Fang X. and colleagues noted an association between PD-L1 expression and a poor prognosis, but it was not deemed an independent prognostic factor [26]. In our study, we observed no significant difference in 3-year and 5-year overall survival rates between the groups using the 30% cut-off value. However, we investigated the relationship between PD-L1 expression in the tumor and survival across a range of cut-off values, including 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, and 80%. Since all the cases were diagnosed with LBCL in our study and there was no control group to compare, we could not use the ROC analysis to determine the cut-off value. Among the binary groups formed using these cut-off values, a significant difference in survival emerged when a 70% threshold was used. In Cox regression analysis, tPD-L1 levels of 70% or higher were found to pose a 2.65 times higher risk in terms of survival. This led to the conclusion that PD-L1 expression is correlated with poor prognosis and serves as an independent prognostic factor. While there was no observed relationship between survival and tPD-L1 staining at a cut-off value of 30%, the identification of such an association at a higher cut-off value underscores the need for more comprehensive studies on this matter.
In our study, we conducted analyses not only using a single cut-off value, but also employing different grouping methods. Survival analyses, performed in three groups (<1%, 1–50%, 50% and above), did not reveal a significant difference between these groups.
In this study, a notable association was observed between the Ann Arbor stage, a critical determinant of patient prognosis, and tPD-L1 positivity for the 30% threshold. When patients with stages 3 and 4 were grouped together and those with stages 1 and 2 were grouped separately, significant tPD-L1 staining was observed in the former group. There are many publications that do not show such a correlation, but there are also studies supporting this finding [6,17,18,20,22,23,24,25,26,27]. A clear consensus is challenging to establish in studies, except for age and LDH-level parameters of the IPI score. Apart from variations in tPD-L1 evaluation methods, factors such as tumor heterogeneity, time to diagnosis, and the utilization of appropriate imaging methods to assess extranodal involvement may have contributed to these differing results.
In our study, apart from DLBCL-NOS, there were 17 cases found that encompass other large cell lymphoma subtypes, which may have caused heterogeneity in the cases. However, when we separately evaluated DLBCL-NOS cases (113 patients) and conducted our statistical analyses, we obtained similar results. The DLBCL-NOS, ABC-like phenotype exhibited a higher tPD-L1 expression rate compared to the GCB-like phenotype, and these cases had lower survival rates. This particular pattern appears consistently across numerous studies in the literature [6,17,18,20,22,23,25,27]. While our study showed no survival difference at the 30% cut-off value, a significant survival discrepancy was evident between these two groups at the 60% cut-off value. This discrepancy became even more pronounced at the 70% cut-off value (p: 0.002). It is worth noting that there is a significant survival difference, particularly at a lower tPD-L1 cut-off level (60%, not 70%), when compared to all cases of large B-cell lymphomas, specifically in the DLBCL-NOS group.

5. Limitations

The lack of a control group and a cut-off value to compare immunohistochemical PD-L1 positivity makes it difficult to compare both of the cases within this study as well as our findings with other research in the literature. Additionally, assessing the expression of PD-L1 can be difficult in some cases due to the potential expression of PD-L1 by cells other than tumor cells.
In our cohort, there are 121 patients who were under hematological follow-up, received R-CHOP therapy, and had complete data available. However, there were variations found among patients in terms of dose adjustments and delays, salvage regimens, and transplantation. Although no statistically significant difference was observed between treatment responses and PD-L1 expression, investigating whether different treatment regimens have any effect on PD-L1 expression remains a subject of inquiry.

6. Conclusions

In our study, we observed a significant difference in tPD-L1 expression and survival at the cut-off value of 70%. While there are varying results in the literature concerning the relationship between PD-L1 expression and survival rates in LBCLs, conducting multicenter studies with larger sample sizes and different subtypes can provide valuable insights. This may shed light on the mechanism of PD-L1 expression in the tumor and whether patients, especially those in treatment-resistant or relapsing disease groups, can benefit from anti-PD-L1 therapy.

Author Contributions

Conceptualization, S.C., S.H.A. and T.E; methodology, S.C. and S.H.A.; validation, S.C. and S.H.A.; formal analysis, S.C.; investigation, S.C. and S.H.A.; data curation, S.C. and S.H.A.; writing—original draft preparation, S.C., S.H.A. and T.E; writing—review and editing, S.C., S.H.A., T.E., D.O. and A.S.; visualization, S.C. and S.H.A.; supervision, S.H.A.; project administration, S.H.A. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Scientific Research Projects Coordination Unit of Istanbul University-Cerrahpasa. (project number: 34006).

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki and approved by the Istanbul University-Cerrahpasa Clinical Research Ethics Committee (number: A-42, date: 2 July 2019).

Informed Consent Statement

Archived material of the patients was used, and no additional procedures were performed. The study was retrospective.

Data Availability Statement

The original contributions presented in the study are included in the article, further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Li, S.; Young, K.H.; Medeiros, L.J. Diffuse large B-cell lymphoma. Pathology 2018, 50, 74–87. [Google Scholar] [CrossRef] [PubMed]
  2. The Non-Hodgkin’s Lymphoma Classification Project. A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin’s lymphoma. Blood 1997, 89, 3909–3918. [Google Scholar] [CrossRef]
  3. Jaffe, E.S.; Arber, D.A.; Campo, E.; Harris, N.L.; Quintanilla-Martinez, L. Hematopathology, 2nd ed.; Elsevier: Philadelphia, PA, USA, 2017; pp. 415–445. [Google Scholar]
  4. Zhang, J.; Medeiros, L.J.; Young, K.H. Cancer Immunotherapy in Diffuse Large B-Cell Lymphoma. Front. Oncol. 2018, 8, 351. [Google Scholar] [CrossRef] [PubMed]
  5. Pierpont, T.M.; Limper, C.B.; Richards, K.L. Past, Present, and Future of Rituximab—The World’s First Oncology Monoclonal Antibody Therapy. Front. Oncol. 2018, 8, 163. [Google Scholar] [CrossRef] [PubMed]
  6. Kiyasu, J.; Miyoshi, H.; Hirata, A.; Arakawa, F.; Ichikawa, A.; Nino, 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, 2193–2201. [Google Scholar] [CrossRef] [PubMed]
  7. Wu, Y.; Chen, W.; Xu, Z.P.; Gu, W. PD-L1 Distribution and Perspective for Cancer Immunotherapy—Blockade, Knockdown, or Inhibition. Front. Immunol. 2019, 10, 2022. [Google Scholar] [CrossRef]
  8. Tomassetti, S.; Chen, R.; Dandapani, S. The role of pembrolizumab in relapsed/refractory primary mediastinal large B-cell lymphoma. Ther. Adv. Hematol. 2019, 10, 2040620719841591. [Google Scholar] [CrossRef] [PubMed]
  9. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [PubMed]
  10. Yamazaki, T.; Akiba, H.; Iwai, H.; Matsuda, H.; Aoki, M.; Tanno, Y.; Shin, T.; Tsuchiya, H.; Pardoll, D.M.; Okumura, K.; et al. Expression of programmed death 1 ligands by murine T cells and APC. J. Immunol. 2002, 169, 5538–5545. [Google Scholar] [CrossRef]
  11. Gravelle, P.; Burroni, B.; Péricart, S.; Rossi, C.; Bezombes, C.; Tosolini, M.; Damotte, D.; Brousset, P.; Fournié, J.J.; Laurent, C. Mechanisms of PD-1/PD-L1 expression and prognostic relevance in non-Hodgkin lymphoma: A summary of immunohistochemical studies. Oncotarget 2017, 8, 44960–44975. [Google Scholar] [CrossRef]
  12. Salmaninejad, A.; Valilou, S.F.; Shabgah, A.G.; Aslani, S.; Alimardani, M.; Pasdar, A.; Sahebkar, A. PD-1/PD-L1 pathway: Basic biology and role in cancer immunotherapy. J. Cell. Physiol. 2019, 234, 16824–16837. [Google Scholar] [CrossRef]
  13. Riella, L.V.; Paterson, A.M.; Sharpe, A.H.; Chandraker, A. Role of the PD-1 pathway in the immune response. Am. J. Transplant. 2012, 12, 2575–2587. [Google Scholar] [CrossRef] [PubMed]
  14. Shitara, K.; Nishikawa, H. Regulatory T cells: A potential target in cancer immunotherapy. Ann. N. Y. Acad. Sci. 2018, 1417, 104–115. [Google Scholar] [CrossRef] [PubMed]
  15. Twa, D.D.; Chan, F.C.; Ben-Neriah, S.; Woolcock, B.W.; Mottok, A.; Tan, K.L.; Slack, G.W.; Gunawardana, J.; Lim, R.S.; McPherson, A.W.; et al. Genomic rearrangements involving programmed death ligands are recurrent in primary mediastinal large B-cell lymphoma. Blood 2014, 123, 2062–2065. [Google Scholar] [CrossRef]
  16. Shen, X.; Zhang, L.; Li, J.; Li, Y.; Wang, Y.; Xu, Z.X. Recent Findings in the Regulation of Programmed Death Ligand 1 Expression. Front. Immunol. 2019, 10, 1337. [Google Scholar] [CrossRef] [PubMed]
  17. Kwon, D.; Kim, S.; Kim, P.J.; Go, H.; Nam, S.J.; Paik, J.H.; Kim, Y.A.; Kim, T.M.; Heo, D.S.; Kim, C.W.; 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, 1079–1089. [Google Scholar] [CrossRef]
  18. Kwon, H.J.; Yang, J.M.; Lee, J.O.; Lee, J.S.; Paik, J.H. Clinicopathologic implication of PD-L1 and phosphorylated STAT3 expression in diffuse large B cell lymphoma. J. Transl. Med. 2018, 16, 320. [Google Scholar] [CrossRef]
  19. Chen, B.J.; Dashnamoorthy, R.; Galera, P.; Makarenko, V.; Chang, H.; Ghosh, S.; Evens, A.M. The immune checkpoint molecules PD-1, PD-L1, TIM-3 and LAG-3 in diffuse large B-cell lymphoma. Oncotarget 2019, 10, 2030–2040. [Google Scholar] [CrossRef]
  20. Hu, L.-Y.; Xu, X.-L.; Rao, H.-L.; Chen, J.; Lai, R.-C.; Huang, H.-Q.; Jiang, W.-Q.; Lin, T.-Y.; Xia, Z.-J.; Cai, Q.-Q. Expression and clinical value of programmed cell death-ligand 1 (PD-L1) in diffuse large B cell lymphoma: A retrospective study. Chin. J. Cancer 2017, 36, 94. [Google Scholar] [CrossRef]
  21. 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, 33350–33362. [Google Scholar] [CrossRef]
  22. Xing, W.; Dresser, K.; Zhang, R.; Evens, A.M.; Yu, H.; Woda, B.A.; Chen, B.J. PD-L1 expression in EBV-negative diffuse large B-cell lymphoma: Clinicopathologic features and prognostic implications. Oncotarget 2016, 7, 59976–59986. [Google Scholar] [CrossRef] [PubMed]
  23. Shi, Y.; Deng, L.; Song, Y.; Lin, D.; Lai, Y.; Zhou, L.; Yang, L.; Li, X. CD3+/CD8+ T-cell density and tumoral PD-L1 predict survival irrespective of rituximab treatment in Chinese diffuse large B-cell lymphoma patients. Int. J. Hematol. 2018, 108, 254–266. [Google Scholar] [CrossRef] [PubMed]
  24. Sun, C.; Jia, Y.; Wang, W.; Bi, R.; Wu, L.; Bai, Q.; Zhou, X. Integrative analysis of PD-L1 DNA status, mRNA status and protein status, and their clinicopathological correlation, in diffuse large B-cell lymphoma. Histopathology 2019, 74, 618–628. [Google Scholar] [CrossRef] [PubMed]
  25. Huang, S.; Nong, L.; Liang, L.; Zheng, Y.; Wang, W.; Liu, J.; Li, D.; Li, X.; Wang, Y.; Zhang, B.; et al. Comparison of PD-L1 detection assays and corresponding significance in evaluation of diffuse large B-cell lymphoma. Cancer Med. 2019, 8, 3831–3845. [Google Scholar] [CrossRef] [PubMed]
  26. 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, e6398. [Google Scholar] [CrossRef]
  27. Qiu, L.; Zheng, H.; Zhao, X. The prognostic and clinicopathological significance of PD-L1 expression in patients with diffuse large B-cell lymphoma: A meta-analysis. BMC Cancer 2019, 19, 273. [Google Scholar] [CrossRef]
Figure 1. (a) DLBCL-NOS, activated B-cell-like, H&E×400 (b) DLBCL-NOS, germinal center B-cell-like, H&E×400 (c) T-cell-rich B-cell lymphoma, H&E×400, (d) immunohistochemical tPD-L1 expression < 30%, (e) tPD-L1 expression ≥ 30%, (f) tPD-L1 expression almost 100%.
Figure 1. (a) DLBCL-NOS, activated B-cell-like, H&E×400 (b) DLBCL-NOS, germinal center B-cell-like, H&E×400 (c) T-cell-rich B-cell lymphoma, H&E×400, (d) immunohistochemical tPD-L1 expression < 30%, (e) tPD-L1 expression ≥ 30%, (f) tPD-L1 expression almost 100%.
Diagnostics 14 01167 g001
Figure 2. Distribution of cases according to tPD-L1 ratio.
Figure 2. Distribution of cases according to tPD-L1 ratio.
Diagnostics 14 01167 g002
Figure 3. Kaplan–Meier survival analysis for the cut-off value of 70% tPD-L1 staining.
Figure 3. Kaplan–Meier survival analysis for the cut-off value of 70% tPD-L1 staining.
Diagnostics 14 01167 g003
Table 1. Clinicopathologic characteristics of patients *.
Table 1. Clinicopathologic characteristics of patients *.
NumberRatio
SexMale7053.8
Female6046.2
Age<60 years7557.7
≥60 years5542.3
B SymptomsPositive8969
Negative4031
SurveyDeceased3829.2
Alive9270.8
LDH LevelNormal5341.1
Elevated7658.9
ECOG Performance Score<210581.4
≥22418.6
Extranodal Involvement<26348.8
≥26651.2
Ann Arbor StageStage 12116.3
Stage 22922.5
Stage 31914.7
Stage 46046.5
Stages 1 and 25038.8
Stages 3 and 47961.2
IPI Score01511.6
12720.9
22317.8
33325.6
42620.2
553.9
R-CHOP Treatment #RCHOP (+)1210.8
RCHOP (−)199.2
PD-L1 Positivity<1%6751.5%
1–49%4433.8%
>50%1914.6%
* Some of the clinical information of one patient could not be accessed. # Treatment information of eight patients could not be obtained.
Table 2. Relationship between PD-L1 expression and clinical parameters for the cut-off value of 30% *.
Table 2. Relationship between PD-L1 expression and clinical parameters for the cut-off value of 30% *.
tPD-L1 Staining Percentage
<%30≥%30
n%n%p
SexMale5477.11622.90.257
Female5185.0915.0
Age<60 years6384.01216.00.275
≥60 years4276.41323.6
B Symptoms+7483.11516.90.279
3075.01025.0
LDH LevelsNormal4686.8713.20.188
Elevated5977.61722.4
ECOG Performance Score<28681.91918.10.567
≥21875.0625.0
Exranodal
Involvement
<25282.51117.50.590
≥25278.81421.2
Ann Arbor StageStages 1 and 24590.0510.00.032
Stages 3 and 45974.72025.3
* Some of the clinical information of one patient could not be accessed.
Table 3. Relationship between PD-L1 expression and clinical parameters for the cut-off value of 70% *.
Table 3. Relationship between PD-L1 expression and clinical parameters for the cut-off value of 70% *.
tPD-L1 Staining Percentage
<%70≥%70
n%n%p
SexMale6085.71014.30.458
Female5490.0610.0
Age<60 years6586.71013.30.678
≥60 years4989.1610.9
LDH LevelsNormal4890.659.40.516
Elevated6686.81013.2
ECOG Performance Score<29388.61211.40.497
≥22083.3416.7
Exranodal
Involvement
<25688.9711.10.664
≥25786.4913.6
Ann Arbor StageStages 1 and 24692.048.00.227
Stages 3 and 46784.81215.2
* Some of the clinical information of one patient could not be accessed.
Table 4. Relationship between PD-L1 expression and clinical parameters for three groups *.
Table 4. Relationship between PD-L1 expression and clinical parameters for three groups *.
tPD-L1 Staining Percentage
<%1%1–49≥%50
n%n%n%p
SexMale3752.92028.61318.60.232
Female3050.02440.0610.0
Age<60 years4458.72026.71114.70.108
≥60 years2341.82443.6814.5
LDH LevelsNormal3056.61732.1611.30.627
Elevated3748.72735.51215.8
ECOG Performance Score<25451.43735.21413.30.499
≥21354.2625.0520.8
Extranodal Involvement<23454.02133.3812.70.802
≥23350.02233.31116.7
Ann Arbor StageStage 11571.4523.814.80.199
Stage 21758.6931.0310.3
Stage 3631.6842.1526.3
Stage 42948.32135.01016.7
* Some of the clinical information of one patient could not be accessed.
Table 5. Cox regression analysis.
Table 5. Cox regression analysis.
HR *%95 CI #p
Age groups<60 vs. ≥602.2672.2671.1000.027
B SymptomsAbsent vs. exist1.5930.7493.3900.227
LDH LevelsNormal vs. elevated0.5450.1781.6730.289
ECOG Performance Score<2 vs. ≥21.4270.6223.2770.401
Ann Arbor StageStage I/II vs. III/IV0.8690.2872.6280.804
IPI Score0/1/2 vs. 3/4/54.6581.07320.2110.040
tPD-L1 Staining Percentage<%70 vs. ≥702.6461.0496.6750.039
* Hazard ratio; # confidence interval.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cin, S.; Aki, S.H.; Elverdi, T.; Ozmen, D.; Salihoglu, A. Is There an Immunohistochemical PD-L1 Cut-Off Point That Serves as a Prognostic Indicator for Large B-Cell Lymphomas? Diagnostics 2024, 14, 1167. https://doi.org/10.3390/diagnostics14111167

AMA Style

Cin S, Aki SH, Elverdi T, Ozmen D, Salihoglu A. Is There an Immunohistochemical PD-L1 Cut-Off Point That Serves as a Prognostic Indicator for Large B-Cell Lymphomas? Diagnostics. 2024; 14(11):1167. https://doi.org/10.3390/diagnostics14111167

Chicago/Turabian Style

Cin, Selcuk, Suat Hilal Aki, Tugrul Elverdi, Deniz Ozmen, and Ayse Salihoglu. 2024. "Is There an Immunohistochemical PD-L1 Cut-Off Point That Serves as a Prognostic Indicator for Large B-Cell Lymphomas?" Diagnostics 14, no. 11: 1167. https://doi.org/10.3390/diagnostics14111167

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop