Zijun Xu-Monette
Overview:
My research efforts have been focused on identifying prognostic and therapeutic biomarkers in B-cell lymphoma. My research interests also include investigation of molecular and immune mechanisms underlying the poor clinical outcomes of lymphoma, the pathogenesis and evolution of drug resistant clones, and development of novel therapies for aggressive B-cell lymphoma.
Positions:
Assistant Professor in Pathology
Pathology
School of Medicine
Member of the Duke Cancer Institute
Duke Cancer Institute
School of Medicine
Education:
Ph.D. 2009
Michigan Technological University
Grants:
Publications:
Analysis of albumin as a prognostic factor in HHV-8/HIV-negative Castleman disease from a multicenter study.
As a rare lymphoproliferative disorder, many patients with HHV-8/HIV-negative Castleman disease (CD) have hypoalbuminemia. However, data is limited on whether hypoalbuminemia is an independent predictor of CD. We retrospectively collected data from 230 patients diagnosed at 12 medical centers in China and the U.S. Different classifications included 147 patients with unicentric CD (UCD) and 83 with idiopathic multicentric CD (iMCD). Adjusted smooth curve fitting showed that the relationship between albumin and all-cause death of patients with CD and iMCD was linear. Cox proportional hazards regression modeling showed a negative association between the risk of death and albumin level (hazard ratio [HR]: 0.84; 95% CI, 0.76, 0.93). Using the Kaplan-Meier method, we determined that hypoproteinemia was a risk factor for poorer prognosis in patients with CD, UCD, and iMCD. Albumin was independently and negatively associated with the risk of death in CD patients, especially those with iMCD.
Authors
Yu, T; Cai, Q-Q; Zhai, Q-L; Li, L; Fang, X; Li, J; Sun, R; Yang, H; Wang, Z; Qian, W; Xu-Monette, ZY; Young, KH; Yu, L
MLA Citation
Yu, Tiantian, et al. “Analysis of albumin as a prognostic factor in HHV-8/HIV-negative Castleman disease from a multicenter study.” Leuk Lymphoma, vol. 63, no. 13, Dec. 2022, pp. 3082–91. Pubmed, doi:10.1080/10428194.2022.2118528.
URI
https://scholars.duke.edu/individual/pub1535631
PMID
36074798
Source
pubmed
Published In
Leuk Lymphoma
Volume
63
Published Date
Start Page
3082
End Page
3091
DOI
10.1080/10428194.2022.2118528
EBV-positive DLBCL frequently harbors somatic mutations associated with clonal hematopoiesis of indeterminate potential.
Authors
Li, Y; Xu-Monette, ZY; Abramson, JS; Sohani, A; Bhagat, G; Tzankov, A; Visco, C; Zhang, S; Dybkaer, K; Pan, Z; Xu, ML; Tam, W; Zu, Y; Hsi, ED; Hagemeister, FB; Go, H; van Krieken, J; Winter, JN; Ponzoni, M; Ferreri, AJM; Møller, MB; Piris, MA; Wang, Y; Zhang, M; Young, KH
MLA Citation
Li, Yong, et al. “EBV-positive DLBCL frequently harbors somatic mutations associated with clonal hematopoiesis of indeterminate potential.” Blood Adv, Nov. 2022. Pubmed, doi:10.1182/bloodadvances.2022008550.
URI
https://scholars.duke.edu/individual/pub1556665
PMID
36399513
Source
pubmed
Published In
Blood Adv
Published Date
DOI
10.1182/bloodadvances.2022008550
IRF8 is a transcriptional activator of CD37 expression in diffuse large B-cell lymphoma.
Diffuse large B-cell lymphoma (DLBCL) represents the most common form of non-Hodgkin lymphoma (NHL) that is still incurable in a large fraction of patients. Tetraspanin CD37 is highly expressed on mature B lymphocytes, and multiple CD37-targeting therapies are under clinical development for NHL. However, CD37 expression is nondetectable in ∼50% of DLBCL patients, which correlates with inferior treatment outcome, but the underlying mechanisms for differential CD37 expression in DLBCL are still unknown. Here, we investigated the regulation of the CD37 gene in human DLBCL at the (epi-)genetic and transcriptional level. No differences were observed in DNA methylation within the CD37 promoter region between CD37-positive and CD37-negative primary DLBCL patient samples. On the contrary, CD37-negative DLBCL cells specifically lacked CD37 promoter activity, suggesting differential regulation of CD37 gene expression. Using an unbiased quantitative proteomic approach, we identified transcription factor IRF8 to be significantly higher expressed in nuclear extracts of CD37-positive as compared with CD37-negative DLBCL. Direct binding of IRF8 to the CD37 promoter region was confirmed by DNA pulldown assay combined with mass spectrometry and targeted chromatin immunoprecipitation (ChIP). Functional analysis indicated that IRF8 overexpression enhanced CD37 protein expression, while CRISPR/Cas9 knockout of IRF8 decreased CD37 levels in DLBCL cell lines. Immunohistochemical analysis in a large cohort of primary DLBCL (n = 206) revealed a significant correlation of IRF8 expression with detectable CD37 levels. Together, this study provides new insight into the molecular mechanisms underlying differential CD37 expression in human DLBCL and reveals IRF8 as a transcriptional regulator of CD37 in B-cell lymphoma.
Authors
Elfrink, S; Ter Beest, M; Janssen, L; Baltissen, MP; Jansen, PWTC; Kenyon, AN; Steen, RM; de Windt, D; Hagemann, PM; Hess, C; van Spronsen, D-J; Hoevenaars, B; van der Spek, E; Xu-Monette, ZY; Young, KH; Kaffa, C; Bervoets, S; van Heek, J; Hesius, E; de Winde, CM; Vermeulen, M; van den Brand, M; Scheijen, B; van Spriel, AB
MLA Citation
Elfrink, Suraya, et al. “IRF8 is a transcriptional activator of CD37 expression in diffuse large B-cell lymphoma.” Blood Adv, vol. 6, no. 7, Apr. 2022, pp. 2254–66. Pubmed, doi:10.1182/bloodadvances.2021004366.
URI
https://scholars.duke.edu/individual/pub1507453
PMID
35086136
Source
pubmed
Published In
Blood Adv
Volume
6
Published Date
Start Page
2254
End Page
2266
DOI
10.1182/bloodadvances.2021004366
Genetic Subtyping and Phenotypic Characterization of the Immune Microenvironment and MYC/BCL2 Double Expression Reveal Heterogeneity in Diffuse Large B-cell Lymphoma.
PURPOSE: Diffuse large B-cell lymphoma (DLBCL) is molecularly and clinically heterogeneous, and can be subtyped according to genetic alterations, cell-of-origin, or microenvironmental signatures using high-throughput genomic data at the DNA or RNA level. Although high-throughput proteomic profiling has not been available for DLBCL subtyping, MYC/BCL2 protein double expression (DE) is an established prognostic biomarker in DLBCL. The purpose of this study is to reveal the relative prognostic roles of DLBCL genetic, phenotypic, and microenvironmental biomarkers. EXPERIMENTAL DESIGN: We performed targeted next-generation sequencing; IHC for MYC, BCL2, and FN1; and fluorescent multiplex IHC for microenvironmental markers in a large cohort of DLBCL. We performed correlative and prognostic analyses within and across DLBCL genetic subtypes and MYC/BCL2 double expressors. RESULTS: We found that MYC/BCL2 double-high-expression (DhE) had significant adverse prognostic impact within the EZB genetic subtype and LymphGen-unclassified DLBCL cases but not within MCD and ST2 genetic subtypes. Conversely, KMT2D mutations significantly stratified DhE but not non-DhE DLBCL. T-cell infiltration showed favorable prognostic effects within BN2, MCD, and DhE but unfavorable effects within ST2 and LymphGen-unclassified cases. FN1 and PD-1-high expression had significant adverse prognostic effects within multiple DLBCL genetic/phenotypic subgroups. The prognostic effects of DhE and immune biomarkers within DLBCL genetic subtypes were independent although DhE and high Ki-67 were significantly associated with lower T-cell infiltration in LymphGen-unclassified cases. CONCLUSIONS: Together, these results demonstrated independent and additive prognostic effects of phenotypic MYC/BCL2 and microenvironment biomarkers and genetic subtyping in DLBCL prognostication, important for improving DLBCL classification and identifying prognostic determinants and therapeutic targets.
Authors
Xu-Monette, ZY; Wei, L; Fang, X; Au, Q; Nunns, H; Nagy, M; Tzankov, A; Zhu, F; Visco, C; Bhagat, G; Dybkaer, K; Chiu, A; Tam, W; Zu, Y; Hsi, ED; Hagemeister, FB; Sun, X; Han, X; Go, H; Ponzoni, M; Ferreri, AJM; Møller, MB; Parsons, BM; van Krieken, JH; Piris, MA; Winter, JN; Li, Y; Xu, B; Albitar, M; You, H; Young, KH
MLA Citation
Xu-Monette, Zijun Y., et al. “Genetic Subtyping and Phenotypic Characterization of the Immune Microenvironment and MYC/BCL2 Double Expression Reveal Heterogeneity in Diffuse Large B-cell Lymphoma.” Clin Cancer Res, vol. 28, no. 5, Mar. 2022, pp. 972–83. Pubmed, doi:10.1158/1078-0432.CCR-21-2949.
URI
https://scholars.duke.edu/individual/pub1505330
PMID
34980601
Source
pubmed
Published In
Clinical Cancer Research
Volume
28
Published Date
Start Page
972
End Page
983
DOI
10.1158/1078-0432.CCR-21-2949
Determining clinical course of diffuse large B-cell lymphoma using targeted transcriptome and machine learning algorithms.
Multiple studies have demonstrated that diffuse large B-cell lymphoma (DLBCL) can be divided into subgroups based on their biology; however, these biological subgroups overlap clinically. Using machine learning, we developed an approach to stratify patients with DLBCL into four subgroups based on survival characteristics. This approach uses data from the targeted transcriptome to predict these survival subgroups. Using the expression levels of 180 genes, our model reliably predicted the four survival subgroups and was validated using independent groups of patients. Multivariate analysis showed that this patient stratification strategy encompasses various biological characteristics of DLBCL, and only TP53 mutations remained an independent prognostic biomarker. This novel approach for stratifying patients with DLBCL, based on the clinical outcome of rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone therapy, can be used to identify patients who may not respond well to these types of therapy, but would otherwise benefit from alternative therapy and clinical trials.
Authors
Albitar, M; Zhang, H; Goy, A; Xu-Monette, ZY; Bhagat, G; Visco, C; Tzankov, A; Fang, X; Zhu, F; Dybkaer, K; Chiu, A; Tam, W; Zu, Y; Hsi, ED; Hagemeister, FB; Huh, J; Ponzoni, M; Ferreri, AJM; Møller, MB; Parsons, BM; van Krieken, JH; Piris, MA; Winter, JN; Li, Y; Xu, B; Young, KH
MLA Citation
Albitar, Maher, et al. “Determining clinical course of diffuse large B-cell lymphoma using targeted transcriptome and machine learning algorithms.” Blood Cancer J, vol. 12, no. 2, Feb. 2022, p. 25. Pubmed, doi:10.1038/s41408-022-00617-5.
URI
https://scholars.duke.edu/individual/pub1509429
PMID
35105854
Source
pubmed
Published In
Blood Cancer Journal
Volume
12
Published Date
Start Page
25
DOI
10.1038/s41408-022-00617-5

Assistant Professor in Pathology
Contact:
905 S Lasalle St, Snyderman Gsrb1, Room 1010, Durham, NC 27710
905 S Lasalle St, Snyderman Gsrb1, Room 1010, Durham, NC 27710