Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1749 word(s) 1749 2021-08-03 06:28:30 |
2 corrected the format Meta information modification 1749 2021-09-13 03:32:51 | |
3 move out from EC Meta information modification 1749 2021-10-12 10:55:08 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ishikawa, E. EBV Positive B-Cell Lymphoproliferative Disorder. Encyclopedia. Available online: https://encyclopedia.pub/entry/14117 (accessed on 28 March 2024).
Ishikawa E. EBV Positive B-Cell Lymphoproliferative Disorder. Encyclopedia. Available at: https://encyclopedia.pub/entry/14117. Accessed March 28, 2024.
Ishikawa, Eri. "EBV Positive B-Cell Lymphoproliferative Disorder" Encyclopedia, https://encyclopedia.pub/entry/14117 (accessed March 28, 2024).
Ishikawa, E. (2021, September 12). EBV Positive B-Cell Lymphoproliferative Disorder. In Encyclopedia. https://encyclopedia.pub/entry/14117
Ishikawa, Eri. "EBV Positive B-Cell Lymphoproliferative Disorder." Encyclopedia. Web. 12 September, 2021.
EBV Positive B-Cell Lymphoproliferative Disorder
Edit

Epstein-Barr virus (EBV) is a gamma herpes virus that infects the majority of the world population. EBV induces B-cell transformation, and disruption of a finely balanced relationship between the virus and host immune system can lead to EBV+B-cell lymphoproliferative disorders (B-LPDs), which represent a wide and expanding clinicopathological spectrum ranging from indolent and self-limited disease to aggressive lymphoma.

Epstein-Barr virus (EBV) mucocutaneous ulcer diffuse large B cell lymphoma (DLBCL) lymphoproliferative gastrointestinal lymphoma gastric lymphoma intestinal lymphoma programmed cell death ligand 1 (PD-L1)

1. Introduction

Epstein-Barr virus (EBV) is a gamma herpes virus that infects the majority of the world population. EBV induces B-cell transformation, and disruption of a finely balanced relationship between the virus and host immune system can lead to EBV + B-cell lymphoproliferative disorders (B-LPDs), which represent a wide and expanding clinicopathological spectrum ranging from indolent and self-limited disease to aggressive lymphoma.

EBV + DLBCL was initially described as senile EBV-associated B-LPD by Oyama et al. in 2003 [1] and was listed as DLBCL of the elderly in the 2008 WHO classification [2][3][4]. After the original study, the development of nodal EBV + DLBCL in young patients with no evidence of immunosuppression was reported by Nicolae et al. in 2015 [5]. As a result, the 2017 WHO classification of malignant lymphoma encompassed these diverse diseases and emphasized that EBV + DLBCL, NOS often affects both young and elderly immunocompetent patients. EBV + DLBCL often presents an aggressive clinical course with frequent extranodal disease. Primary EBV + DLBCL of the GI tract (giDLBCL) accounted for 5% to 10% of consecutively diagnosed giDLBCL in a series of 62, 107, and 240 cases [6][7][8]. Although contradictory conclusions on the significance of EBV in regards to clinical outcome have been reported [5][9][10][11][12], we recently documented the negative impact of EBV in the largest series of 156 patients with gDLBCL and 51 patients with iDLBCL in the rituximab era to date [6][7].

EBVMCU is an ulcerating EBV + B-LPD with a self-limited indolent course [13]. The disease is associated with advanced age and immunosuppression, such as primary immunodeficiency, post-transplantation, and other iatrogenic causes, including methotrexate (MTX), prednisolone (PSL), azathioprine (AZA), cyclosporin A (CYA), and TNF-α antagonists [13]. The disease often involves the oropharyngeal mucosa, skin, and GI tract. In particular, EBVMCU in the GI tract (giEBVMCU) is frequently detected in patients with inflammatory bowel disease or immune-related colitis, which is the specific disease in the intestine [13][14][15][16][17][18][19]. In general, giEBVMCU responds well to conservative management, but patients with immune-related colitis are distinct from others in the high frequency of perforation requiring surgery [19].

Recent advances in immune-oncology have expanded our knowledge of immune evasion, mostly by programmed cell death ligand 1 (PD-L1) and the PD-1 pathway has become an attractive therapeutic target in various malignancies [20][21][22][23][24]. We also showed that PD-L1 immunohistochemistry aids in the differential diagnostic approach for EBV + B-LPDs and their morphological analogues [25]. EBV + DLBCL is accompanied by high PD-L1 expression but the clinicopathological significance remains to be clarified [21][26]. We further revealed that PD-L1 expression on tumor cells and non-malignant immune cells has an opposite prognostic impact in patients with giDLBCL [7][27].

2. EBV Biology and the GI Tract

EBV was the first oncogenic virus identified and it persistently infects the B cells of >95% of adults, resulting in an asymptomatic life-long carrier status [28]. The EBV life cycle is biphasic, with phases of lytic replication and latency [29]. EBV is etiologically linked to a wide range of human tumors, including gastric carcinoma, nasopharyngeal carcinoma, and lymphoma, and three EBV latency patterns have been recognized [29][30][31]. Latency I is associated with Burkitt’s lymphoma and a distinct subset of gastric carcinomas, whereas Latency II is associated with classic Hodgkin’s lymphoma, extranodal NK/T-cell lymphoma, and nasopharyngeal carcinoma [32]. Latency III is linked to immunodeficiency-associated LPDs arising in the setting of immunosuppression due to HIV infection, post-transplantation, and other iatrogenic causes, such as MTX and anti-TNF-α therapy.

Some studies have reported that the presence of EBV is restricted to lymphoid cells, not benign epithelial cells, in gastritis lesions and normal colonic mucosa [33][34]. In the stomach, a positive correlation between EBVDNA load and Helicobacter pylori positivity has been reported, which suggests that the H . pylori infection could trigger EBV to switch from the latent to lytic phase of its life cycle [35][36]. In addition, EBV infection of tumor cells is detected in approximately 9% of gastric cancer (GC) cases; it is categorized as one of the major GC types and characterized by a high frequency of amplification and elevated expression of PD-L1 [37][38][39]. Notably, EBV + GC is an attractive target for anti-PD-1/PD-L1 therapy in the current era [40][41].

In the intestine, EBV replication has been reported to be associated with severe inflammatory bowel disease and mucosal inflammation [42]. The presence of EBV-infected B lymphocytes in colonic lesions of IBD may indicate a potential role of EBV in colonic immune disturbances, which may be caused by the inflammatory process itself, immunosuppressive medication, or a combination of both. In contrast, Lopes et al. showed that mucosal EBV load does not correlate with the presence of inflammation or endoscopic severity despite a higher prevalence of EBV infection in IBD [43]. Whether EBV is involved in the pathogenesis or is an innocent bystander remains unclear, but active inflammation with intramucosal expansion of EBV-infected B-lymphocytes in IBD patients may cause local impairment. In IBD patients exposed to thiopurines or anti-TNF agents, particularly in combination, the risk of LPDs, mostly associated with EBV, is higher [44][45][46]. Thiopurines are cytotoxic for NK and cytotoxic T cells, which restrict proliferation of EBV-infected and immortalized B cells, which could be associated with lymphomagenesis [47].

3. EBV-Positive Mucocutaneous Ulcer (EBVMCU)

EBVMCU, first recognized by Dojcinov et al., is defined as an ulcerating EBV + B-LPD affecting the skin and mucosal surfaces, with a typically indolent course and spontaneous regression in some cases.

EBVMCU occurs in iatrogenic immunosuppressed patients with autoimmune disorders and inflammatory bowel disease receiving MTX, CYA, AZA, TNF-α antibody, tacrolimus (Tac), or steroid treatment, in solid organ or bone marrow transplant recipients, in HIV-positive patients, after other lymphoma or tumor treatment, and in elderly patients. Elderly subjects are markedly restricted and deficient in their epitope-specific T-cell repertoire, leading to an increased risk of infection for the host [13][48].

A handful of studies have reported PD-L1 expression in EBVMCU. Satou et al. described the lack of PD-L1 expression on tumor cells in seven cases with MTX-associated EBVMCU using two different clones (SP142 and E1J2J) [49], which is consistent with findings reported by Daroontum et al. in 13 cases of EBVMCU associated with treated lymphoma or MTX [50]. Interestingly, in their subsequent series, one possibly exceptional EBVMCU case in which multiple EBV-driven B-LPDs developed after spontaneous regression of the disease was found to have PD-L1 expression on tumor cells at the time of the initial onset of EBVMCU [51]. This PD-L1 expression detected by immunohistochemistry (clone SP142) may be related to unusual clinical behavior after the spontaneous regression of EBVMCU. In contrast, Prieto et al. reported that all three cases with EBVMCU were positive for PD-L1 on large cells and HRS-like cells when using clone 28-8 [18]. This discordance in PD-L1 expression between studies is thought to be due to the use of different anti-PD-L1 antibody clones (SP142 vs. 28-8). However, a definitive conclusion cannot be drawn because of a limited number of cases examined, and this issue is expected to be clarified in a larger study.

Six patients with EBVMCU in IBD consisted of three with Crohn’s disease, two with ulcerative colitis, and one without any available information. Except for the one with Crohn’s disease presenting with an anal lesion, all cases had rectal involvement, two of which were accompanied by synchronous involvement of the colon. Macroscopically, most cases had ulcerated lesions during treatment with immunosuppressive regimens, including AZA, IFX, MTX, 6-mercaptopurine, and CYA, whereas only one used mesalazine alone and had a non-specific erythematous lesion. Interestingly, two cases had multiple lesions, which is not typical for ordinal EBVMCU [15][17]. In most EBVMCU cases with IBD, the clinical course was benign with complete recovery by reduction of immune suppression or aggressive therapy. Two patients achieved complete remission upon reduction of immune suppression. One patient with ulcerative colitis received rituximab because colonic biopsies showed persistent necrotic ulceration with EBV-positive immunoblasts after 4 weeks, though the ulcer slightly decreased in size by reduction of immune suppression and achieved complete remission [17]. The other patient with Crohn’s disease finally underwent proctectomy with terminal colostomy due to an inability to control the rectal symptoms despite cessation of AZA and IFX [16]. The third patient using only mesalazine did not receive any therapy and presented with stable persistent disease without symptomatology for 6 months [18]. The remaining patient with Crohn’s disease treated with AZA and adalimumab in the past presented an aggressive clinical course, developing widespread classic Hodgkin lymphoma 18 months after cessation of IFX and MTX [15].

4. EBV-Positive Diffuse Large B-Cell Lymphoma (EBV+ DLBCL)

Among patients with common DLBCL, the frequency of PD-L1 expression has been reported to be 6% to 26% with different cut-off values and anti-PD-L1 monoclonal antibodies [26][52][53][54]. In general, an alteration in chromosome 9p24.1 is rarely found, and the structural variations disrupting the 3′ untranslated region of the PD-L1 gene, which correlates with PD-L1 expression, is detected in 8% of common DLBCL cases [52][55]. However, whether this PD-L1 expression on malignant tumor cells has an adverse prognostic impact is still controversial [26][53][56]. PD-L1 is also expressed on nonmalignant immune cells, such as macrophages and dendritic cells in DLBCL; thus, the prognostic significance is unproven because of the paucity of reports.

In EBV + cases, EBV-LMP1 increases PD-L1 promoter and enhancer activity [57]. EBV + DLBCL has a higher frequency of PD-L1 expression on tumor cells (19–100%) and immune cells (40–100%) compared to EBV-negative DLBCL [21][26][56]. Notably, 76% of young patients with EBV + DLBCL exhibit PD-L1 positivity in the tumor cells [5]. Takahara et al. recently reported that PD-L1 expression (clone SP142-positive staining) was present in more than 5% of tumor cells in only 6 (11%) of 57 cases (95% were >45 years old) [58], clearly contrasting the 77% reported in younger cases (<45 years old) [5]. The former also indicated that PD-L1 + cases had significantly shorter progression-free survival ( p = 0.002) and relatively short overall survival ( p = 0.26), compared to PD-L1-negative cases.

In our series of giDLBCL patients using PD-L1 (by clone SP142) immunohistochemistry, neoplastic cell staining was considered positive for PD-L1 (nPD-L1) when ≥5% of the lymphoid cells exhibited moderate to strong membrane staining. In addition, microenvironment immune cell staining was considered positive for PD-L1 (miPD-L1) when, among the total tissue cellularity, ≥20% comprised non-malignant cells with moderate or strong membrane or cytoplasmic PD-L1-specific staining.

Thus far in the English literature, researchers first documented that PD-L1 expression on non-malignant immune cells, such as macrophages and dendritic cells, contributes to better outcomes in giDLBCL patients treated with modern immunochemotherapy.

References

  1. Oyama, T.; Ichimura, K.; Suzuki, R.; Suzumiya, J.; Ohshima, K.; Yatabe, Y.; Yokoi, T.; Kojima, M.; Kamiya, Y.; Taji, H.; et al. Senile EBV plus B-cell Lymphoproliferative disorders—A clinicopathologic study of 22 patients. Am. J. Surg. Pathol. 2003, 27, 16–26.
  2. Oyama, T.; Yamamoto, K.; Asano, N.; Oshiro, A.; Suzuki, R.; Kagami, Y.; Morishima, Y.; Takeuchi, K.; Izumo, T.; Mori, S.; et al. Age-related EBV-Associated B-Cell Lymphoproliferative disorders constitute a distinct clinicopathologic group: A study of 96 patients. Clin. Cancer Res. 2007, 13, 5124–5132.
  3. Park, S.; Lee, J.; Ko, Y.H.; Han, A.; Jun, H.J.; Lee, S.C.; Hwang, I.G.; Park, Y.H.; Ahn, J.S.; Jung, C.W.; et al. The impact of Epstein-Barr virus status on clinical outcome in diffuse large B-cell lymphoma. Blood 2007, 110, 972–978.
  4. Morales, D.; Beltran, B.; De Mendoza, F.H.; Riva, L.; Yabar, A.; Quinones, P.; Butera, J.N.; Castillo, J. Epstein-Barr virus as a prognostic factor in de novo nodal diffuse large B-cell lymphoma. Leuk. Lymphoma 2010, 51, 66–72.
  5. Nicolae, A.; Pittaluga, S.; Abdullah, S.; Steinberg, S.M.; Pham, T.A.; Davies-Hill, T.; Xi, L.Q.; Raffeld, M.; Jaffe, E.S. EBV-positive large B-cell lymphomas in young patients: A nodal lymphoma with evidence for a tolerogenic immune environment. Blood 2015, 126, 863–872.
  6. Ishikawa, E.; Tanaka, T.; Shimada, K.; Kohno, K.; Satou, A.; EladI, A.E.; Sakakibara, A.; Furukawa, K.; Funasaka, K.; Miyahara, R.; et al. A prognostic model, including the EBV status of tumor cells, for primary gastric diffuse large B-cell lymphoma in the rituximab era. Cancer Med. 2018, 7, 3510–3520.
  7. Ishikawa, E.; Kato, S.; Shimada, K.; Tanaka, T.; Suzuki, Y.; Satou, A.; Kohno, K.; Sakakibara, A.; Yamamura, T.; Nakamura, M.; et al. Clinicopathological analysis of primary intestinal diffuse large B-cell lymphoma: Prognostic evaluation of CD5, PD-L1, and Epstein-Barr virus on tumor cells. Cancer Med. 2018, 7, 6051–6063.
  8. Maeshima, A.M.; Taniguchi, H.; Ito, Y.; Hatta, S.; Suzuki, T.; Yuda, S.; Makita, S.; Fukuhara, S.; Munakata, W.; Suzuki, T.; et al. Clinicopathological characteristics of diffuse large B-cell lymphoma involving small and large intestines: An analysis of 126 patients. Int. J. Hematol. 2019, 110, 340–346.
  9. Ahn, J.S.; Yang, D.H.; Choi, Y.D.; Jung, S.H.; Yhim, H.Y.; Kwak, J.Y.; Park, H.S.; Shin, M.G.; Kim, Y.K.; Kim, H.J.; et al. Clinical outcome of elderly patients with Epstein-Barr virus positive diffuse large B-cell lymphoma treated with a combination of rituximab and CHOP chemotherapy. Am. J. Hematol. 2013, 88, 774–779.
  10. Sato, A.; Nakamura, N.; Kojima, M.; Ohmachi, K.; Carreras, J.; Kikuti, Y.Y.; Numata, H.; Ohgiya, D.; Tazume, K.; Amaki, J.; et al. Clinical outcome of Epstein-Barr virus-positive diffuse large B-cell lymphoma of the elderly in the rituximab era. Cancer Sci. 2014, 105, 1170–1175.
  11. Hong, J.Y.; Yoon, D.H.; Suh, C.; Huh, J.; Do, I.G.; Sohn, I.; Jo, J.; Jung, S.H.; Hong, M.E.; Yoon, H.; et al. EBV-positive diffuse large B-cell lymphoma in young adults: Is this a distinct disease entity? Ann. Oncol. 2015, 26, 548–555.
  12. Ok, C.Y.; Ye, Q.; Li, L.; Manyam, G.C.; Deng, L.J.; Goswami, R.R.; Wang, X.X.; Montes-Moreno, S.; Visco, C.; Tzankov, A.; et al. Age cutoff for Epstein-Barr virus-positive diffuse large B-cell lymphoma—is it necessary? Oncotarget 2015, 6, 13935–13946.
  13. Dojcinov, S.D.; Venkataraman, G.; Raffeld, M.; Pittaluga, S.; Jaffe, E.S. EBV Positive Mucocutaneous Ulcer-A Study of 26 Cases Associated with Various Sources of Immunosuppression. Am. J. Surg. Pathol. 2010, 34, 405–417.
  14. Matnani, R.; Peker, D. Azathioprine induced Epstein Barr virus-positive mucocutaneous ulcer arising in perianal fistula and abscess associated with Crohn’s disease. J. Crohns Colitis 2014, 8, 1747–1748.
  15. Moran, N.R.; Webster, B.; Lee, K.M.; Trotman, J.; Kwan, Y.L.; Napoli, J.; Leong, R.W. Epstein Barr virus-positive mucocutaneous ulcer of the colon associated Hodgkin lymphoma in Crohn’s disease. World J. Gastroenterol. 2015, 21, 6072–6076.
  16. Juan, A.; Lobaton, T.; Tapia, G.; Manosa, M.; Cabre, E.; Domenech, E. Epstein-Barr virus-positive mucocutaneous ulcer in Crohn’s disease. A condition to consider in immunosuppressed IBD patients. Dig. Liver Dis. 2017, 49, 934–937.
  17. Goetgebuer, R.L.; van der Woude, C.J.; de Ridder, L.; Doukas, M.; de Vries, A.C. Clinical and endoscopic complications of Epstein-Barr virus in inflammatory bowel disease: An illustrative case series. Int. J. Colorectal Dis. 2019, 34, 923–926.
  18. Prieto-Torres, L.; Erana, I.; Gil-Redondo, R.; de la Riva, I.G.; Manso, R.; Pajares, R.; Cordoba, R.; Machan, S.; Ara, M.; Requena, L.; et al. The Spectrum of EBV-Positive Mucocutaneous Ulcer A Study of 9 Cases. Am. J. Surg. Pathol. 2019, 43, 201–210.
  19. Pugh, M.R.; Leopold, G.D.; Morgan, M.; Christian, A.D.; Hewett, R.; Durai, D.; Wagstaff, J.; Harris, D.; Dojcinov, S.D. Epstein-Barr Virus-Positive Mucocutaneous Ulcers Complicate Colitis Caused by Immune Checkpoint Regulator Therapy and Associate with Colon Perforation. Clin. Gastroenterol. Hepatol. 2020, 18, 1785.
  20. Dong, H.D.; Strome, S.E.; Salomao, D.R.; Tamura, H.; Hirano, F.; Flies, D.B.; Roche, P.C.; Lu, J.; Zhu, G.F.; Tamada, K.; et al. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat. Med. 2002, 8, 793–800.
  21. Chen, B.J.; Chapuy, B.; Jing, O.Y.; Sun, H.H.; Roemer, M.G.M.; Xu, M.L.; Yu, H.B.; Fletcher, C.D.M.; Freeman, G.J.; Shipp, M.A.; et al. PD-L1 Expression Is Characteristic of a Subset of Aggressive B-cell Lymphomas and Virus-Associated Malignancies. Clin. Cancer Res. 2013, 19, 3462–3473.
  22. Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.M.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and Activity of Anti-PD-L1 Antibody in Patients with Advanced Cancer. N. Engl. J. Med. 2012, 366, 2455–2465.
  23. Balar, A.V.; Weber, J.S. PD-1 and PD-L1 antibodies in cancer: Current status and future directions. Cancer Immunol. Immunother. 2017, 66, 551–564.
  24. Lesokhin, A.M.; Ansell, S.M.; Armand, P.; Scott, E.C.; Halwani, A.; Gutierrez, M.; Millenson, M.M.; Cohen, A.D.; Schuster, S.J.; Lebovic, D.; et al. Nivolumab in Patients with Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J. Clin. Oncol. 2016, 34, 2698.
  25. Sakakibara, A.; Kohno, K.; Ishikawa, E.; Suzuki, Y.; Shimada, S.; Eladl, A.E.; Elsayed, A.A.; Daroontum, T.; Satou, A.; Takahara, T.; et al. Age-related EBV-associated B-cell lymphoproliferative disorders and other EBV plus lymphoproliferative diseases: New insights into immune escape and immunodeficiency through staining with anti-PD-L1 antibody clone SP142. Pathol. Int. 2020, 70, 481–492.
  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, 2193–2201.
  27. Ishikawa, E.; Nakamura, M.; Shimada, K.; Tanaka, T.; Satou, A.; Kohno, K.; Sakakibara, A.; Furukawa, K.; Yamamura, T.; Miyahara, R.; et al. Prognostic impact of PD-L1 expression in primary gastric and intestinal diffuse large B-cell lymphoma. J. Gastroenterol. 2020, 55, 39–50.
  28. Thorley-Lawson, D.A.; Gross, A. Mechanisms of disease—Persistence of the Epstein-Barr virus and the origins of associated lymphomas. N. Engl. J. Med. 2004, 350, 1328–1337.
  29. Taylor, G.S.; Long, H.M.; Brooks, J.M.; Rickinson, A.B.; Hislop, A.D. The immunology of Epstein-Barr Virus Induced Disease. Annu. Rev. Immunol. 2015, 33, 787.
  30. Young, L.S.; Rickinson, A.B. Epstein-Barr virus: 40 years on. Nat. Rev. Cancer 2004, 4, 757–768.
  31. Young, L.S.; Yap, L.F.; Murray, P.G. Epstein-Barr virus: More than 50 years old and still providing surprises. Nat. Rev. Cancer 2016, 16, 789–802.
  32. Satou, A.; Asano, N.; Nakazawa, A.; Osumi, T.; Tsurusawa, M.; Ishiguro, A.; Elsayed, A.A.; Nakamura, N.; Ohshima, K.; Kinoshita, T.; et al. Epstein-Barr Virus (EBV)-positive Sporadic Burkitt Lymphoma an Age-related Lymphoproliferative Disorder? Am. J. Surg. Pathol. 2015, 39, 227–235.
  33. Ryan, J.L.; Shen, Y.J.; Morgan, D.R.; Thorne, L.B.; Kenney, S.C.; Dominguez, R.L.; Gulley, M.L. Epstein-Barr Virus Infection Is Common in Inflamed Gastrointestinal Mucosa. Dig. Dis. Sci. 2012, 57, 1887–1898.
  34. Yanai, H.; Shimizu, N.; Nagasaki, S.; Mitani, N.; Okita, K. Epstein-Barr virus infection of the colon with inflammatory bowel disease. Am. J. Gastroenterol. 1999, 94, 1582–1586.
  35. Shukla, S.K.; Prasad, K.N.; Tripathi, A.; Singh, A.; Saxena, A.; Chand Ghoshal, U.; Krishnani, N.; Husain, N. Epstein-Barr virus DNA load and its association with Helicobacter pylori infection in gastroduodenal diseases. Braz. J. Infect. Dis. 2011, 15, 583–590.
  36. Yanai, H.; Murakami, T.; Yoshiyama, H.; Takeuchi, H.; Nishikawa, J.; Nakamura, H.; Okita, K.; Miura, O.; Shimizu, N.; Takada, K. Epstein-Barr virus-associated gastric carcinoma and atrophic gastritis. J. Clin. Gastroenterol. 1999, 29, 39–43.
  37. Bass, A.J.; Thorsson, V.; Shmulevich, I.; Reynolds, S.M.; Miller, M.; Bernard, B.; Hinoue, T.; Laird, P.W.; Curtis, C.; Shen, H.; et al. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209.
  38. Saito, R.; Abe, H.; Kunita, A.; Yamashita, H.; Seto, Y.; Fukayama, M. Overexpression and gene amplification of PD-L1 in cancer cells and PD-L1(+) immune cells in Epstein-Barr virus-associated gastric cancer: The prognostic implications. Mod. Pathol. 2017, 30, 427–439.
  39. Fang, W.L.; Chen, M.H.; Huang, K.H.; Lin, C.H.; Chao, Y.; Lo, S.S.; Li, A.F.Y.; Wu, C.W.; Shyr, Y.M. The Clinicopathological Features and Genetic Alterations in Epstein-Barr Virus-Associated Gastric Cancer Patients after Curative Surgery. Cancers 2020, 12, 1517.
  40. Kim, S.T.; Cristescu, R.; Bass, A.J.; Kim, K.M.; Odegaard, J.I.; Kim, K.; Liu, X.Q.; Sher, X.W.; Jung, H.; Lee, M.; et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat. Med. 2018, 24, 1449.
  41. Kono, K.; Nakajima, S.; Mimura, K. Current status of immune checkpoint inhibitors for gastric cancer. Gastric Cancer 2020, 23, 565–578.
  42. Sankaran-Walters, S.; Ransibrahmanakul, K.; Grishina, I.; Hung, J.; Martinez, E.; Prindiville, T.; Dandekar, S. Epstein-Barr virus replication linked to B cell proliferation in inflamed areas of colonic mucosa of patients with inflammatory bowel disease. J. Clin. Virol. 2011, 50, 31–36.
  43. Lopes, S.; Andrade, P.; Conde, S.; Liberal, R.; Dias, C.C.; Fernandes, S.; Pinheiro, J.; Simoes, J.S.; Carneiro, F.; Magro, F.; et al. Looking into Enteric Virome in Patients with IBD: Defining Guilty or Innocence? Inflamm. Bowel Dis. 2017, 23, 278–284.
  44. Dayharsh, G.A.; Loftus, E.V.; Sandborn, W.J.; Tremaine, W.J.; Zinsmeister, A.R.; Witzig, T.E.; Macon, W.R.; Burgart, L.J. Epstein-Barr virus-positive lymphoma in patients with inflammatory bowel disease treated with azathioprine or 6-mercaptopurine. Gastroenterology 2002, 122, 72–77.
  45. Beaugerie, L.; Brousse, N.; Bouvier, A.M.; Colombel, J.F.; Lemann, M.; Cosnes, J.; Hebuterne, X.; Cortot, A.; Bouhnik, Y.; Gendre, J.P.; et al. Lymphoproliferative disorders in patients receiving thiopurines for inflammatory bowel disease: A prospective observational cohort study. Lancet 2009, 374, 1617–1625.
  46. Lemaitre, M.; Kirchgesner, J.; Rudnichi, A.; Carrat, F.; Zureik, M.; Carbonnel, F.; Dray-Spira, R. Association Between Use of Thiopurines or Tumor Necrosis Factor Antagonists Alone or in Combination and Risk of Lymphoma in Patients with Inflammatory Bowel Disease. JAMA J. Am. Med. Assoc. 2017, 318, 1679–1686.
  47. Rezk, S.A.; Weiss, L.M. Epstein-Barr virus-associated lymphoproliferative disorders. Hum. Pathol. 2007, 38, 1293–1304.
  48. Ghia, P.; Prato, G.; Stella, S.; Scielzo, C.; Geuna, M.; Caligaris-Cappio, F. Age-dependent accumulation of monoclonal CD4(+)CD8(+)double positive T lymphocytes in the peripheral blood of the elderly. Br. J. Haematol. 2007, 139, 780–790.
  49. Satou, A.; Banno, S.; Hanamura, I.; Takahashi, E.; Takahara, T.; Nobata, H.; Katsuno, T.; Takami, A.; Ito, Y.; Ueda, R.; et al. EBV-positive mucocutaneous ulcer arising in rheumatoid arthritis patients treated with methotrexate: Single center series of nine cases. Pathol. Int. 2019, 69, 21–28.
  50. Daroontum, T.; Kohno, K.; Eladl, A.E.; Satou, A.; Sakakibara, A.; Matsukage, S.; Yakushiji, N.; Ya-In, C.; Nakamura, S.; Asano, N.; et al. Comparison of Epstein-Barr virus-positive mucocutaneous ulcer associated with treated lymphoma or methotrexate in Japan. Histopathology 2018, 72, 1115–1127.
  51. Daroontum, T.; Kohno, K.; Inaguma, Y.; Okamoto, A.; Okamoto, M.; Kimura, Y.; Nagahama, M.; Sakakibara, A.; Satou, A.; Nakamura, S. Epstein-Barr virus (EBV)-positive diffuse large B-cell lymphoma arising in patient with a history of EBV-positive mucocutaneous ulcer and EBV-positive nodal polymorphous B-lymphoproliferative disorder. Pathol. Int. 2019, 69, 37–41.
  52. Georgiou, K.; Chen, L.Y.; Berglund, M.; Ren, W.C.; de Miranda, N.; Lisboa, S.; Fangazio, M.; Zhu, S.D.; Hou, Y.; Wu, K.; et al. Genetic basis of PD-L1 overexpression in diffuse large B-cell lymphomas. Blood 2016, 127, 3026–3034.
  53. McCord, R.; Bolen, C.R.; Koeppen, H.; Kadel, E.E.; Oestergaard, M.Z.; Nielsen, T.; Sehn, L.H.; Venstrom, J.M. PD-L1 and tumor-associated macrophages in de novo DLBCL. Blood Adv. 2019, 3, 531–540.
  54. 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, s40880.
  55. Kataoka, K.; Shiraishi, Y.; Takeda, Y.; Sakata, S.; Matsumoto, M.; Nagano, S.; Maeda, T.; Nagata, Y.; Kitanaka, A.; Mizuno, S.; et al. Aberrant PD-L1 expression through 3 ‘-UTR disruption in multiple cancers. Nature 2016, 534, 402.
  56. 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.
  57. Green, M.R.; Rodig, S.; Juszczynski, P.; Ouyang, J.; Sinha, P.; O’Donnell, E.; Neuberg, D.; Shipp, M.A. Constitutive AP-1 Activity and EBV Infection Induce PD-L1 in Hodgkin Lymphomas and Posttransplant Lymphoproliferative Disorders: Implications for Targeted Therapy. Clin. Cancer Res. 2012, 18, 1611–1618.
  58. Takahara, T.; Satou, A.; Ishikawa, E.; Kohno, K.; Kato, S.; Suzuki, Y.; Takahashi, E.; Ohashi, A.; Asano, N.; Tsuzuki, T.; et al. Clinicopathological analysis of neoplastic PD-L1-positive EBV(+)diffuse large B cell lymphoma, not otherwise specified, in a Japanese cohort. Virchows Archiv 2021, 478, 541–552.
More
Information
Subjects: Cell Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 379
Revisions: 3 times (View History)
Update Date: 12 Oct 2021
1000/1000