J Pathol Transl Med.  2022 Jan;56(1):32-39. 10.4132/jptm.2021.10.05.

Clinicopathologic implication of PD-L1 gene alteration in primary adrenal diffuse large B cell lymphoma

Affiliations
  • 1Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
  • 2Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
  • 3Department of Pathology, Seoul National University Hospital, Seoul, Korea
  • 4Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
  • 5Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea

Abstract

Background
Primary adrenal (PA) diffuse large B cell lymphoma (DLBCL) was previously reported as an aggressive subset of DLBCL, but its genetic features were not sufficiently characterized. From our previous study of DLBCL with programmed death-ligand 1 (PD-L1) gene alterations, we focused on PD-L1 gene alterations in PA-DLBCL with clinicopathologic implications.
Methods
We performed fluorescence in situ hybridization for PD-L1 gene translocation and amplification in PA-DLBCL (n = 18) and comparatively analyzed clinicopathologic characteristics with systemic non-adrenal (NA)-DLBCL (n = 90).
Results
PA-DLBCL harbored distinctive features (vs. NADLBCL), including high international prognostic index score (3–5) (72% [13/18] vs. 38% [34/90], p = .007), poor Eastern Cooperative Oncology Group performance score (≥ 2) (47% [7/15] vs. 11% [10/90], p = .003), elevated serum lactate dehydrogenase (LDH) (78% [14/18] vs. 51% [44/87], p = .035) and MUM1 expression (87% [13/15] vs. 60% [54/90], p = .047). Moreover, PA-DLBCL showed frequent PD-L1 gene alterations (vs. NA-DLBCL) (39% [7/18] vs. 6% [5/86], p = .001), including translocation (22% [4/18] vs. 3% [3/87], p = .016) and amplification (17% [3/18] vs. 2% [2/87], p = .034). Within the PA-DLBCL group, PD-L1 gene–altered cases (vs. non-altered cases) tended to have B symptoms (p = .145) and elevated LDH (p = .119) but less frequent bulky disease (≥ 10 cm) (p = .119). In the survival analysis, PA-DLBCL had a poor prognosis for overall survival (OS) and progression-free survival (PFS) (vs. NA-DLBCL; p = .014 and p = .004). Within the PA-DLBCL group, PD-L1 translocation was associated with shorter OS and PFS (p < .001 and p = .012).
Conclusions
PA-DLBCL is a clinically aggressive and distinct subset of DLBCL with frequent PD-L1 gene alterations. PD-L1 gene translocation was associated with poor prognosis in PA-DLBCL.

Keyword

Malignant lymphoma; Diffuse large B cell lymphoma; Adrenal gland; PD-L1

Figure

  • Fig. 1 Fluorescence in situ hybridization analysis of the programmed death-ligand 1 (PD-L1) gene in diffuse large B cell lymphoma. By using a dual-color orange/green break-apart probe, non-split fused signal indicates the absence of translocation (A), while separate orange and green signals indicate the presence of translocation (B). By using a copy number analysis probe, orange (PD-L1 gene) and green (chromosome 9) signals showed a nearly 1:1 ratio in cases with no gain or amplification of the PD-L1 gene (C), in contrast to amplified orange signals indicating PD-L1 gene amplification (D).

  • Fig. 2 Kaplan-Meier curves of overall survival (OS) and progression-free survival (PFS) in diffuse large B cell lymphoma (DLBCL) patients. OS (A) and PFS (B) of primary adrenal (PA)- and non-adrenal (NA)-DLBCL patients. OS (C) and PFS (D) of PA-DLBCL patients by the presence of programmed death-ligand 1 (PD-L1) gene alterations. OS (E) and PFS (F) of PA-DLBCL patients by the presence of PD-L1 gene translocation.


Reference

References

1. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016; 127:2375–90.
Article
2. Nagasaki J, Togashi Y, Sugawara T, et al. The critical role of CD4+ T cells in PD-1 blockade against MHC-II-expressing tumors such as classic Hodgkin lymphoma. Blood Adv. 2020; 4:4069–82.
Article
3. Xu-Monette ZY, Zhou J, Young KH. PD-1 expression and clinical PD-1 blockade in B-cell lymphomas. Blood. 2018; 131:68–83.
Article
4. Roemer MG, Advani RH, Ligon AH, et al. PD-L1 and PD-L2 genetic alterations define classical Hodgkin lymphoma and predict outcome. J Clin Oncol. 2016; 34:2690–7.
Article
5. Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015; 372:311–9.
Article
6. Kwon HJ, Yang JM, Lee JO, Lee JS, Paik JH. Clinicopathologic implication of PD-L1 and phosphorylated STAT3 expression in diffuse large B cell lymphoma. J Transl Med. 2018; 16:320.
Article
7. Twa DD, Chan FC, Ben-Neriah S, et al. Genomic rearrangements involving programmed death ligands are recurrent in primary mediastinal large B-cell lymphoma. Blood. 2014; 123:2062–5.
Article
8. Chapuy B, Roemer MG, Stewart C, et al. Targetable genetic features of primary testicular and primary central nervous system lymphomas. Blood. 2016; 127:869–81.
Article
9. Cohen R, Jonchere V, De La Fouchardiere C, et al. Adrenal gland as a sanctuary site for immunotherapy in patients with microsatellite instability-high metastatic colorectal cancer. J Immunother Cancer. 2021; 9:e001903.
Article
10. Mushtaq RR, Gao D, Raeburn C, Tobin RP, Robinson W. Adrenal metastases in malignant melanoma, is it a privileged site? J Clin Oncol. 2019; 37(15 Suppl):e21016.
Article
11. Rashidi A, Fisher SI. Primary adrenal lymphoma: a systematic review. Ann Hematol. 2013; 92:1583–93.
Article
12. Kim YR, Kim JS, Min YH, et al. Prognostic factors in primary diffuse large B-cell lymphoma of adrenal gland treated with rituximab-CHOP chemotherapy from the Consortium for Improving Survival of Lymphoma (CISL). J Hematol Oncol. 2012; 5:49.
Article
13. Laurent C, Casasnovas O, Martin L, et al. Adrenal lymphoma: presentation, management and prognosis. QJM. 2017; 110:103–9.
Article
14. Mozos A, Ye H, Chuang WY, et al. Most primary adrenal lymphomas are diffuse large B-cell lymphomas with non-germinal center B-cell phenotype, BCL6 gene rearrangement and poor prognosis. Mod Pathol. 2009; 22:1210–7.
Article
15. Audrito V, Serra S, Stingi A, et al. PD-L1 up-regulation in melanoma increases disease aggressiveness and is mediated through miR-17-5p. Oncotarget. 2017; 8:15894–911.
Article
16. Gao Q, Wang XY, Qiu SJ, et al. Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin Cancer Res. 2009; 15:971–9.
Article
17. Hans CP, Weisenburger DD, Greiner TC, et al. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood. 2004; 103:275–82.
Article
18. Bardhan K, Anagnostou T, Boussiotis VA. The PD1:PD-L1/2 pathway from discovery to clinical implementation. Front Immunol. 2016; 7:550.
Article
19. Hudson K, Cross N, Jordan-Mahy N, Leyland R. The extrinsic and intrinsic roles of PD-L1 and its receptor PD-1: implications for immunotherapy treatment. Front Immunol. 2020; 11:568931.
Article
20. Pasqualucci L, Dalla-Favera R. The genetic landscape of diffuse large B-cell lymphoma. Semin Hematol. 2015; 52:67–76.
Article
21. Iqbal J, Greiner TC, Patel K, et al. Distinctive patterns of BCL6 molecular alterations and their functional consequences in different subgroups of diffuse large B-cell lymphoma. Leukemia. 2007; 21:2332–43.
Article
22. Peng C, Hu Q, Yang F, Zhang H, Li F, Huang C. BCL6-mediated silencing of PD-1 ligands in germinal center B cells maintains follicular T cell population. J Immunol. 2019; 202:704–13.
Article
Full Text Links
  • JPTM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr