Abstract 4046: Immune repertoire sequencing reveals tumor microenvironment and tracks clonally expanded B cell and T cell in blood

Author(s):  
Chen Song ◽  
Pingfang Liu ◽  
Andrew Barry ◽  
Bradley W. Langhorst ◽  
Fiona J. Stewart ◽  
...  
2019 ◽  
Author(s):  
Chen Song ◽  
Pingfang Liu ◽  
Andrew Barry ◽  
Bradley W. Langhorst ◽  
Fiona J. Stewart ◽  
...  

2021 ◽  
Vol 17 (1) ◽  
Author(s):  
Gary Kwok Cheong Lee ◽  
Dorothee Bienzle ◽  
Stefan Matthias Keller ◽  
Mei-Hua Hwang ◽  
Nikos Darzentas ◽  
...  

Abstract Background Lymphocytic neoplasms with frequent reactive lymphocytes are uncommonly reported in dogs, and can pose a diagnostic challenge. Different diagnostic modalities such as cytology, flow cytometry, histopathology, immunohistochemistry, and clonality testing, are sometimes required for a diagnosis. This report illustrates the value of using a multi-modal diagnostic approach to decipher a complex lymphocytic tumor, and introduces immune repertoire sequencing as a diagnostic adjunct. Case presentation A 10-month-old Great Dane was referred for marked ascites. Cytologic analysis of abdominal fluid and hepatic aspirates revealed a mixed lymphocyte population including numerous large lymphocytes, yielding a diagnosis of lymphoma. Flow cytometrically, abdominal fluid lymphocytes were highly positive for CD4, CD5, CD18, CD45, and MHC II, consistent with T cell lymphoma. Due to a rapidly deteriorating clinical condition, the dog was euthanized. Post mortem histologic evaluation showed effacement of the liver by aggregates of B cells surrounded by T cells, suggestive of hepatic T cell-rich large B cell lymphoma. Immune repertoire sequencing confirmed the presence of clonal B cells in the liver but not the abdominal fluid, whereas reactive T cells with shared, polyclonal immune repertoires were found in both locations. Conclusions T cell-rich large B cell lymphoma is a rare neoplasm in dogs that may be challenging to diagnose and classify due to mixed lymphocyte populations. In this case, the results of histopathology, immunohistochemistry and immune repertoire sequencing were most consistent with a hepatic B cell neoplasm and reactive T cells exfoliating into the abdominal fluid. Immune repertoire sequencing was helpful in delineating neoplastic from reactive lymphocytes and characterizing repertoire overlap in both compartments. The potential pitfalls of equating atypical cytomorphology and monotypic marker expression in neoplasia are highlighted.


2018 ◽  
Vol 102 ◽  
pp. S36
Author(s):  
Silvia Pineda ◽  
Tara K Sigdel ◽  
Juliane Liberto ◽  
Krishna Roskin ◽  
Scott Boyd ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2684-2684
Author(s):  
Nasir Bakshi ◽  
Mansoor Aljabry ◽  
Saad Akhter ◽  
Irfan Maghfoor ◽  
Ayman Mashi

Abstract Abstract 2684 NLPHL accounts for 6.5% of all Hodgkin lymphoma cases in the West. It is characterized by a nodular or a nodular & diffuse proliferation of scattered large atypical CD20+ neoplastic B-cells referred to as lymphocyte predominant (LP) cells and typically associated with small lymphocytes mainly of B-cell type. Patients with NLPHL typically have an indolent clinical course but can frequently relapse. Progression to a higher grade lymphoma, notably T-cell/Histiocyte rich B-cell lymphoma (T/HRBCL) has been described in a relatively small number of cases. Because of its rarity, limited information is available about the role of non-neoplastic lymphocytes in NLPHL. Some studies suggest that NLPHL with T-cell rich background may behave differently than the conventional type with predominance of B-cells within the nodules. The purpose of this study was to evaluate outcomes of differential tumor microenvironment namely B-cell versus T-cell rich in patients with NLPHL. We document the clinicopathologic profiles of 29 patients with biopsy proven NLPHL, consisting of 22 male & 7 female, median age 26 years (range, 13–80 years). All patients had lymphoadenopathy & 2 cases showed extranodal involvement in addition to nodal disease. Two patients had a bulky mass, and three had stage 4 disease at presentation. The pathological diagnoses was reviewed and confirmed by an expert hematopathologist in all 29 cases. The LP cells in all cases had a prototypic immunophenotype of CD20+, CD79a+, PU.1+, Bcl-6+, CD15− CD30− & Fascin−. T/HRBCL was excluded as all cases demonstrated preservation of follicular dendritic meshwork by CD21 staining. The meshwork was expanded in 20 cases & in 9 cases it was partially disrupted evincing an irregular architectural pattern. Epstein-Barr Virus encoded RNA by in situ hybridization was negative in 8/8 cases tested. 27/29 patients received systemic multi-agent chemotherapy consisting of: doxorubicin, bleomycin, vinblastine, and dacarbacin (ABVD), 24 patients; cyclophosphamide, doxorubicin, vincristin, and prednisone (CHOP), 2 patients; Rituximab + CHOP (R-CHOP), 1 patient. 9/29 (31%) cases underwent autologous stem cell transplant. One patient in stage 2A refused therapy and one patient (stage 3A) developed significantly decreased cardiac ejection fraction following initial 2 cycles of ABVD. Both of these cases did not have adequate follow-up information available. Results: Twelve of the 29 cases (42%) were designated as having T-cell rich background population, whereas 17 (58%) were considered as conventional variant with a vast predominance of non-neoplastic small lymphocytes being B-cells. A few of the cases seemed to show admixture of both B-cells & T-cells. Comparing T-cell rich & B-cell rich background NLPHL no significant differences were detected in clinical parameters: age, sex, and stage at presentation, absolute lymphocyte count, LDH & Hb. All 27 (100%) patients in this study responded to first-line treatment: 23 with complete response & 4 with partial response. 13/27 (48%) had relapse/s. Five cases had more than one relapses. No patient died within a clinical follow-up period ranging from 18 to 84 months. When the overall survival (OS) of T-cell rich NLPHL was compared with the conventional variant there was no statistical significance between the two groups (log rank p= 0.1206). However, comparison of relapse rate showed that cases with T-cell rich background had higher relapse rate as well as greater incidence of multiple relapses as compared to B-cell rich type of NLPHL even after adjusting for the type of treatment received (log rank p= 0.003). Moreover, 2/12 (17%) T-cell rich NLPHL cases showed transformation to a high grade lymphoma (both T/HRBCL) at the time of recurrence. These findings suggest that in NLPHL a tumor microenvironment rich in T-cells rather than B-cells is characterized by an unfavorable clinical course although OS appears to be similar. These cases perhaps represent a distinctive clinicopathologic variant within the framework of NLPHL. Lately, the term ‘NLPHL with nodules resembling T/HRBCL’ has been used to express the immunobiological overlap between these two entities. It is possible that such cases could be regarded as “intermediate lymphomas” treading between NLPHL and T/HRLBCL. Further studies using gene array profiling analysis may help clarify the molecular differences between these closely related entities. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1554-1554
Author(s):  
Lucy S. Hodge ◽  
Steve Ziesmer ◽  
Frank J Secreto ◽  
Zhi-Zhang Yang ◽  
Anne Novak ◽  
...  

Abstract Abstract 1554 T cells in the tumor microenvironment influence the biology of malignant cells in many hematologic malignancies, often through cytokine-mediated interactions. Recent studies involving healthy B cells and CD4+T cells identified an interplay between IL-6 and IL-21, whereby IL-6 increased IL-21 production by T cells, driving the differentiation and IL-6 secretion of nearby B cells. In addition to their known effects on healthy B cell function, IL-6 and IL-21 have also been implicated in the pathology of various lymphomas. In Waldenstrom's macroglobulinemia (WM), IL-6 is elevated in the bone marrow and is associated with increased IgM production. However, the function of IL-21 in the WM tumor microenvironment and its relationship to IL-6 is poorly understood. Our objective in this study was to characterize IL-21 production and function in WM and to examine the role of IL-6 and IL-21 in regulating interactions between malignant B cells and T cells in the tumor microenvironment. Immunohistochemistry revealed significant IL-21 staining in bone marrows of patients with WM (n=5), but the areas of infiltration by WM in the bone marrow sections appeared negative for IL-21 staining. To better understand the origin of IL-21 in in the tumor microenvironment, IL-21 expression was assessed by PCR in the CD19−CD138− fraction of cells remaining in patient bone marrow aspirates after positive selection for malignant B cells (n=5). IL-21 transcript was detected in 4/5 samples. CD19−CD138− cells activated with anti-CD3 and anti-CD28 antibodies expressed higher levels of IL-21 transcript and secreted significantly higher levels of IL-21 protein compared to unstimulated cells, suggesting that IL-21 in the WM bone marrow is derived from activated T cells. Intracellular expression of IL-21 protein was confirmed in CD4+ and CD8+ cells within the CD19−CD138− population using flow cytometry. Furthermore, dual staining of WM bone marrow sections with antibodies against IL-21 and CD3 or CD20 revealed co-staining of IL-21 with CD3+ T cells but not with CD20+ B cells. The response of WM B cells to T-cell derived IL-21 was then assessed in positively selected CD19+CD138+ WM B cells (n=5) and in the MWCL-1 cell line. Using flow cytometry, both the IL-21 receptor and the required common gamma chain subunit were detected on all patient samples as well as on MWCL-1 cells. Treatment of MWCL-1 cells with IL-21 (100 ng/mL) for 72 h increased proliferation by 35% (p<0.05) and IgM secretion by 80% (p<0.005). Similarly, in primary CD19+CD138+ WM cells (n=5), proliferation increased on average by 38% and IgM secretion by 71%. No apoptotic effects were associated with IL-21 in WM. Characterization of STAT activation in response to IL-21 revealed significant phosphorylation of STAT3 in both CD19+CD138+ WM cells and MWCL-1 cells and was associated with increases in BLIMP-1 and XBP-1 protein and decreases in PAX5. As STAT3 activation is known to regulate IL-6, we assessed the effect of IL-21 on B cell-mediated IL-6 secretion using ELISA. IL-21 significantly increased IL-6 secretion by both primary CD19+CD138+ WM cells (n=4) and MWCL-1 cells (87.9 +/− 10.9 ng/mL vs. 297.8 +/− 129.2 ng/mL, p<0.05). Treatment with IL-6 and IL-21 together had no additional effect over IL-21 alone on proliferation or IgM secretion in MWCL-1 cells, but culturing anti-CD3/anti-CD28-activated CD19−CD138−cells from WM bone marrows with IL-6 significantly increased IL-21 secretion (n=3). Overall, these data indicate that T-cell derived IL-21 significantly promotes growth and immunoglobulin production by malignant WM B cells and that subsequent IL-6 secretion by malignant B cells may enhance the secretion of IL-21 by T cells within the bone marrow microenvironment. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 35 (7_suppl) ◽  
pp. 76-76
Author(s):  
Young Kwang Chae ◽  
William Han Bae ◽  
Yeonjoo Choi ◽  
Young Suk Kim ◽  
Jonathan Forrest Anker ◽  
...  

76 Background: Compared to recent advances in our knowledge of T cell biology with success of immunotherapy, little progress has been made in understanding of the effects of B cells in tumor microenvironment and their interactions with T cells. Preclinical studies reported that B cells may have immune suppressive roles in tumor microenvironment via induction of T cell exhaustion. However, this association has not been shown in human tissues. We explored the landscape of tumor infiltrating B and T cells and their association with tumor microenvironment in various human cancers for which the FDA approved the use of immune checkpoint inhibitors. Methods: Expression patterns for 812 immune related genes from the TCGA database were utilized to define tumor infiltrating cells in 2951 patients with bladder urothelial carcinoma, renal clear cell carcinoma, skin cutaneous melanoma, lung squamous cell carcinoma, lung adenocarcinoma, and head and neck squamous cell carcinoma. Odds ratios (ORs) of the numbers of tumors with versus without activated B cell infiltration by the presence of activated CD8T cell infiltration were calculated. Results: Immune landscape of the six human cancers showed a consistent inverse association between tumor infiltrating activated B and CD8 T cells (OR = 0.18, p < 0.001). B cell infiltration was associated with increased expressions of immune checkpoints PD-L1, PD-1 and CTLA-4 and regulatory cytokines TGF-β, IL-10 and IL-35, which are known to be secreted by regulatory B cells. Angiogenic markers, such as angiopoietins, VEGF, MMP-9, CXCL10, CXCL11 and Tie2, showed differential expression patterns between B cell high and low groups. Conclusions: This is the first study that reports the inverse association between tumor infiltrating B and CD8 T cells in human tissues. The strong associations between B cell infiltration and increased expressions of suppressive cytokines and immune checkpoints suggest regulatory B cells may play a role in the T cell suppression in tumor microenvironment. Our results implicate that depleting B cells, leading to possible disinhibition of T cell activation, may be a future therapeutic option in potentiating T cell mediated immunity.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1509-1509
Author(s):  
Katsuyoshi Takata ◽  
Lauren C. Chong ◽  
Avinash Thakur ◽  
Tomohiro Aoki ◽  
Anja Mottok ◽  
...  

Background: The tumor-associated antigen PRAME is over-expressed in several types of cancer and is currently investigated as a therapeutic target for T-cell immunotherapy. Our previous integrative genomic study in diffuse large B-cell lymphoma (DLBCL) identified PRAME deletion to be correlated with patient outcome and an immunologically "cold" tumor microenvironment. However, it remains an open question whether PRAME expression significantly contributes to differential treatment outcomes and tumor microenvironment crosstalk across various B-cell lymphoma subtypes. Material and Methods: We performed an immunohistochemical (IHC) screen in a large cohort of B-cell lymphomas (de novo DLBCL; N=347, follicular lymphoma (FL); N= 166, mantle cell lymphoma (MCL); N= 180), and classical Hodgkin lymphoma (HL); N= 166) to assess PRAME expression as a prognostic biomarker. Moreover, to investigate PRAME-expression associated tumor microenvironment composition and function, we correlated PRAME IHC results with single cell RNA sequencing data of more than 127,000 cells from 22 HL tissue specimens. Results: PRAME IHC analysis revealed frequent PRAME over-expression in HL (115/166, 69%), followed by DLBCL (104/319, 33%), FL (13/166, 8%), and MCL (14/180, 8%). Interestingly, only HL showed a significant treatment outcome correlation, whereas other B-cell lymphoma subtypes did not. Specifically, using a previously published HL cohort (Steidl et al, NEJM 2010) PRAME-negative Hodgkin Reed Sternberg (HRS) cells indicated significantly shorter overall survival (P = 0.008) and disease-specific survival (P = 0.042 ). To characterize PRAME-specific microenvironment composition and function in HL, we analyzed T-, B-, NK-cell, and macrophage subsets in PRAME-positive (17 of 22 cases) vs -negative (5 of 22 cases) tumor samples using single cell RNA sequencing data. From 22 expression-based microenvironment cell clusters that were annotated and assigned to a cell type based on gene expression, all three CD4 helper T-cell clusters were de-enriched in PRAME-negative samples, and the CD4 non-Treg proportion was significantly lower in PRAME-negative samples (P = 0.049). Strikingly, when focusing on phenotypic features of cells within the CD4 non-Treg T-cell cluster, CXCL13 was identified as the most up-regulated gene in PRAME-negative samples. When interrogating published HRS cell transcriptome data (Steidl et al, Blood 2012), immune response pathways including chemokine receptors and chemokine ligands were up-regulated in PRAME-negative HRS cell samples. Of specific interest, CXCR5, the cognate receptor for CXCL13, was significantly upregulated as a member of the chemokine pathway (P = 0.0086) in PRAME-negative HRS cell samples. These results suggest that crosstalk between CXCL13 (produced in the microenvironment) and CXCR5 (expressed on HRS cells) contributes to tumor maintenance in PRAME-negative HL. Finally, to explore potential therapeutic approaches for PRAME-negative HL cells, we focused on 3 HL-derived cell lines (L540, L591, DEV) with low PRAME expression and exposed these lines to DNMT or HDAC inhibitors. DNMT inhibitor treatment showed clear restoration of PRAME expression in a dose dependent manner, but no restoration was found by HDAC inhibitor treatment. To investigate the effect of DNA methylation in transcriptional regulation of PRAME in HL cells, we performed bisulfite sequencing in the PRAME CpG promoter region in PRAME down-regulated (L540, L591, DEV) and up-regulated (HD-LM2, KMH-2, L1236) cell lines and found hypermethylation in PRAME low vs high cell lines. Moreover, the CpG promoter region was significantly demethylated by DNMT inhibitor treatment in cell lines with low PRAME expression. Conclusion: We discovered that PRAME protein expression was correlated with outcome in HL and identified specific T-cell subsets in PRAME-negative patients. PRAME restoration by DNMT inhibitors might represent a new therapeutic avenue in combination with modern immunotherapies, such as PRAME-specific T-cell therapy or PD1 inhibition. Disclosures Scott: Roche/Genentech: Research Funding; Janssen: Consultancy, Research Funding; NanoString: Patents & Royalties: Named inventor on a patent licensed to NanoSting [Institution], Research Funding; Celgene: Consultancy. Steidl:Nanostring: Patents & Royalties: Filed patent on behalf of BC Cancer; Bristol-Myers Squibb: Research Funding; Roche: Consultancy; Seattle Genetics: Consultancy; Bayer: Consultancy; Juno Therapeutics: Consultancy; Tioma: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document