Next Article in Journal
Effects of Elevated Glucose on Bacterial Respiratory Infections in Cystic Fibrosis and Chronic Airway Diseases
Next Article in Special Issue
Application of Optical Genome Mapping for the Diagnosis and Risk Stratification of Myeloid and Lymphoid Malignancies
Previous Article in Journal
Patterns of Recombination in Coronaviruses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Overexpression of CDCA2 in Diffuse Large B-Cell Lymphoma Promotes Cell Proliferation and Bortezomib Sensitivity

by
Hanne Due
1,2,*,
Asta Brogaard
1,
Issa Ismail Issa
1,2,
Maja Zimmer Jakobsen
1,2,
Cathrine Sylvester
1,
Anne Krogh Nøhr
3,
Louiza Bohn Thomsen
4,
Martin Kristian Thomsen
5,
Rasmus Froberg Brøndum
3 and
Karen Dybkær
1,2,*
1
Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
2
Department of Clinical Medicine, Aalborg University, 9260 Gistrup, Denmark
3
Center for Clinical Data Science, Aalborg University Hospital, and Aalborg University, 9260 Gistrup, Denmark
4
Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark
5
Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(12), 5596; https://doi.org/10.3390/ijms26125596
Submission received: 23 April 2025 / Revised: 30 May 2025 / Accepted: 5 June 2025 / Published: 11 June 2025
(This article belongs to the Special Issue Molecular Diagnosis and Treatment of Hematological Malignancies)

Abstract

Numerous clinical trials have attempted to improve first-line R-CHOP treatment of diffuse large B-cell lymphoma (DLBCL) through the addition or substitution of drugs. The REMoDL-B trial, testing the addition of bortezomib (RB-CHOP), revealed that ABC and molecular high-grade DLBCL patients benefit from bortezomib. The aim of this study was to achieve a better understanding of the bortezomib response in DLBCL through a functional investigation of clinically identified markers. A retrospective analysis of transcriptional and clinical data from the REMoDL-B trial was conducted to identify genes associated with bortezomib response, identifying CDCA2. DLBCL patients with high expression of CDCA2 had a superior survival outcome when treated with RB-CHOP in comparison to R-CHOP, whereas no difference in outcome was observed for patients with low CDCA2. Moreover, CDCA2 was found to be overexpressed in DLBCL compared to non-malignant tissue, and to have higher levels in GCB and MYC/BCL2 double-expressor patients. Functional in vitro and in vivo studies revealed that knockout of CDCA2 decreased DLBCL cell proliferation and a bortezomib dose–response analysis showed less sensitivity in CDCA2 knockout cells compared to control cells. This study shows that DLBCL patients with high CDCA2 expression benefitted from the addition of bortezomib to R-CHOP and functional studies documented a direct impact of CDCA2 on the bortezomib response in DLBCL cells.

1. Introduction

Molecular heterogeneity is characteristic of a diffuse large B-cell lymphoma (DLBCL), causing differences in clinical outcomes depending on the cytogenetic, genetic, and transcriptional profile [1,2,3,4,5]. The initial discovery of molecular heterogeneity arose from gene expression profiling (GEP) and showed that DLBCL can be divided into cell of origin subgroups, with expression profiles resembling normal germinal-center B-cells and in vitro-activated B-cells, defined as GCB- and ABC-DLBCL, respectively. These molecular subgroups display distinct pathogenesis and clinical outcomes, with inferior prognosis in ABC-DLBCL patients [1]. In addition, DLBCL can be subclassified based on translocations of MYC, BCL2, and/or BCL6, defining high-grade B-cell lymphoma, also referred to as double-hit and triple-hit lymphoma [6]. GEP-based approaches have also been applied to identify MYC-driven subgroups of DLBCL, including the double-hit signature and molecular high-grade (MHG), both of which partially overlap with double- and triple-hit lymphomas [7,8]. More recently, comprehensive investigations of the mutational landscape of DLBCL have led to the discovery of several genetic subclasses [3,4,9,10].
Although the standard treatment with R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone) cures up to 60% of patients, recurrent and progressive diseases constitute a significant clinical challenge, where most relapsed/refractory patients die [11,12]. Multiple clinical trials have tested the addition of novel agents targeting aberrant intracellular pathways of the malignant B-cells, most with limited effect [13,14,15,16]. The REMoDL-B trial tested R-CHOP vs. R-CHOP + bortezomib (RB-CHOP) in newly diagnosed DLBCL patients stratified by cell of origin [15,16] and documented a significantly improved 5-year overall and progression-free survival for ABC- and MHG-DLBCL patients treated with bortezomib in combination with R-CHOP [16].
To achieve a better understanding of the molecular determinants of the bortezomib response we re-analyzed transcriptional and clinical data of the REMoDL-B trial and described for the first-time the aberrant expression and prognostic significance of CDCA2 in DLBCL. Through knockout of CDCA2, we showed the functional impact of CDCA2 on DLBCL cell proliferation, cell cycle progression, and response to proteasome inhibitor treatment in single and combination drug studies.

2. Results

2.1. CDCA2 Is Upregulated and Associated with Bortezomib Response in DLBCL

To identify genes displaying prognostic impact in interaction with bortezomib, the REMoDL-B data (Supplementary Table S1) was analyzed by multiple Cox proportional hazards regressions. The analysis tested all expressed genes with overall survival as the outcome and included an interaction term between gene expression and the treatment arm. Identified genes were involved in a variety of biological processes, including cell cycle, transcription, and Wnt signaling among others (Supplementary Table S2). One of the strongest interactions to bortezomib response was found in CDCA2, which displayed a different effect on the outcome in the R-CHOP and RB-CHOP treatment arm (p = 0.08). Cell Division Cycle Associated 2 (CDCA2) is a cell cycle related protein, which has been documented to be upregulated in other types of cancer [17,18,19,20]. Therefore, CDCA2 mRNA expression was examined in non-malignant and DLBCL samples, which showed higher levels of CDCA2 in DLBCL compared to normal tissue specimens (p ≤ 0.001) (Figure 1A). Consistently, the CDCA2 expression level was significantly higher in DLBCL cell lines than in normal B-cell compartments (p ≤ 0.001). CDCA2 expression was not associated with clinical factors including disease stage and international prognostic index (IPI) (Supplementary Figure S1A–D); however, GCB-DLBCL patients displayed increased CDCA2 expression in comparison to ABC and unclassified patients (p ≤ 0.05) (Figure 1B). Moreover, higher CDCA2 levels were observed in MYC/BCL2 double-expressors (p ≤ 0.01) (Figure 1C), constituting of 36% GCB subclassed patients. The difference in CDCA2 mRNA levels between ABC and GCB-DLBCL patients prompted us to examine its expression in refined B-cell subsets including normal B-cell differentiation subsets and B-cell-associated gene signature (BAGS) classification of DLBCL patients. A higher expression of CDCA2 was observed in normal centroblasts and centrocytes compared to naïve, memory, and plasmablast B-cells from healthy tonsils (p ≤ 0.01), and likewise, the BAGS-classified centrocyte and centroblast subgroups of DLBCL tumors [2] displayed higher CDCA2 levels than the other subtypes, although they were not statistically significant (Figure 1D, Supplementary Figure S1F).
A Kaplan–Meier survival analysis revealed a superior outcome in DLBCL patients with a high CDCA2 expression when treated with RB-CHOP in comparison to R-CHOP (p = 0.012), whereas no difference in survival was observed for patients with low expression of CDCA2 (p = 0.095) (Figure 2A,B). In multivariate Cox proportional hazards regression analyses, CDCA2 displayed prognostic impact independently of IPI and ABC/GCB in R-CHOP treated patients, whereas no prognostic significance was observed in RB-CHOP treated patients (Figure 2C,D, Supplementary Table S3A,B), suggesting CDCA2 as a marker of a bortezomib response. Moreover, the prognostic value of CDCA2 was independent of double-expressor status (Supplementary Table S4A,B).

2.2. Knockout of CDCA2 Suppresses DLBCL Cell Proliferation

To elucidate the biological function of CDCA2 in DLBCL, RNP and lentiviral-based CRISPR-mediated knockout (KO) of CDCA2 was performed. Indel analysis showed heterogenous indel populations with knockout scores of 72 and 82 in RNP-transfected RIVA cells and monoclonal knockouts with scores >90 in lentiviral transduced OCILY7 and RIVA cells (Figure 3A, Supplementary Figure S2A,B). Protein depletion was documented by Western blotting (Figure 3A). For verification of control, the proliferation rates of scramble control (SCR) and parental wildtype cells were compared and revealed no difference (Supplementary Figure S3A). Knockout of CDCA2 suppressed the proliferation of DLBCL cells significantly compared to SCR, with the most prominent effect observed in monoclonal knockout cells (Figure 3B). In addition, indel distribution in RNP-transfected CDCA2-KO cells changed over time with decreases in indel and knockout scores, as result of the overgrowth of cells without CDCA2 knockouts (Supplementary Figure S2C,D), supporting slower proliferation rates of CDCA2-KO cells.
Transcriptional profiling of CDCA2-KO clones and SCR was performed to explore the pathways affected upon loss of CDCA2. Gene set enrichment analysis (GSEA) indicated that CDCA2 is closely related to the G2/M checkpoint and to targets of the E2F transcription factor which play a prominent role during the G1/S transition of the cell cycle (Supplementary Figure S4). Additionally, CDCA2 is associated with the PI3K/AKT signaling pathway, which is also important for the regulation of cell cycle progression [21]. In agreement, flow cytometry analysis showed that loss of CDCA2 affected the cell cycle transition between the G1- and S-phase, resulting in a significantly increased percentage of cells in G1 compared to the control (Figure 3C, Supplementary Figure S5A). Moreover, a phospho-flow analysis showed significantly less AKT Ser473 phosphorylation in CDCA2-KO cells compared to SCR (Figure 3D, Supplementary Figure S5B), documenting the direct impact of CDCA2 on the PI3K/AKT pathway.
DLBCL xenografts were established to explore effects of CDCA2 loss in vivo. The tumor growth was significantly reduced in CDCA2-KO xenograft tumors in comparison to SCR tumors, resulting in longer survival time (Figure 3E,F). Indel analysis documented stable knockout of CDCA2 in tumors of CDCA2-KO xenografts (Supplementary Figure S6A,B). Tumors were analyzed by immunohistochemistry (IHC), and increased expression of Ki-67 was observed in CDCA2-KO tumors in comparison to SCRs (Figure 3G, Supplementary Figure S7A,B).

2.3. CDCA2 Impacts Bortezomib Response in DLBCL Cells

The findings from the analysis of REMoDL-B data prompted us to investigate the role of CDCA2 in bortezomib response in single and combination dose–response screens. No difference in response was observed between parental wildtype and SCR (Supplementary Figure S3B,C). Compared to SCR, CDCA2-KO cells demonstrated significantly less sensitivity to bortezomib over a range of concentrations with a more modest response in the polyclonal CDCA2-KO cell populations compared to the monoclonal CDCA2-KO clones (Figure 4A–C). In concordance, lower apoptotic levels were observed in CDCA2-KO cells compared to SCR upon bortezomib exposure (Figure 4D,E, Supplementary Figure S8). Moreover, CDCA2-KO cells were less sensitive to carfilzomib, a 2nd generation proteasome inhibitor (Supplementary Figure S9), showing a general effect of CDCA2 on proteasome inhibitors. Combinatory drug screens using CHOP and B-CHOP showed that SCR cells possessing physiological levels of CDCA2 were more sensitive to B-CHOP than CHOP, whereas CDCA2-KO cells displayed no difference in response to the two regimens (Figure 4F), illustrating CDCA2 to be cardinal and specifically mediating the bortezomib response.
Next, we set out to decipher the mechanism through which CDCA2 impact bortezomib response. CDCA2 did not affect the proteasomal inhibiting effect of bortezomib as no difference in chymotrypsin, trypsin, and caspase activity of the proteasome was observed upon bortezomib treatment between CDCA2-KO and SCR cells (Figure 5A). This suggests alternative mechanisms by which CDCA2 modulates bortezomib response. As CDCA2 was documented to affect PI3K/AKT signaling at the transcriptional and phosphorylation levels, the impact of this pathway was examined. Applying the PI3K inhibitor, LY294002, a reduction in cell proliferation was observed in both CDCA2-KO and SCR cells; however, it was observed to a greater extent in SCR cells with a 25% reduction compared to 16% of CDCA2-KO cells (Figure 5B). Combinatory dose–response analysis of LY294002 and bortezomib revealed that inhibition of the PI3K pathway sensitizes cells to bortezomib irrespective of presence or absence of CDCA2 (Figure 5C), suggesting that CDCA2 affects the bortezomib response through mechanisms other than PI3K/AKT. The sensitizing effect of LY294002 is equal in SCR and CDCA2-KO cells, despite having a different reducing impact on cell proliferation, showing that bortezomib response through PI3K is not driven by proliferation.

3. Discussion

In this study, we demonstrate, for the first-time, the aberrant expression of CDCA2 in DLBCL samples and the functional impact of CDCA2 in DLBCL cells. Overexpression of CDCA2 has been reported in colorectal, prostate, and liver cancer, among others [17,18,19,20], and it was reported to be positively correlated to the disease stage [18,19,20]. However, CDCA2 expression did not correlate with stage or prognostic index in our data. Interestingly, higher levels of CDCA2 were observed in normal germinal-center B-cells, indicating that CDCA2 plays a role in normal B-cell development. In line, DLBCL patients of the molecular GCB cell of origin subclass displayed higher CDCA2 expression than ABC-DLBCL patients.
The prognostic impact of CDCA2 mRNA expression was documented, suggesting CDCA2 expression as indicator of bortezomib response. Further investigation of additional clinical DLBCL cohorts could provide more information on the predictive biomarker potential of CDCA2, yet the REMoDL-B trial is, to our knowledge, the only comprehensive study of bortezomib treatment in DLBCL. Moreover, as DLBCL is a highly heterogeneous disease [1,2,3,4,5] it is unlikely that a single gene can predict the response to a given treatment and therefore further assessment of the biomarker potential is out of scope for this study. Our study should thus be considered useful for improved biological understanding of bortezomib response rather than as a single gene prognostic biomarker candidate.
The REMoDL-B trial showed subclass-specific responses to bortezomib addition, where the improved outcome of ABC-DLBCL is explained by the inhibition of the constitutive NF-ΚΒ signaling observed in this subclass [22]. This mechanism is unlikely to be the explanation for the treatment efficacy observed in MHG-DLBCL, which has been suggested to be attributed to MYC-driven proteolytic stress, sensitizing cells to bortezomib [16]. We observed higher CDCA2 expression in DLBCL patients classified as MYC/BCL2 double-expressors, who display a more aggressive disease, as it is an adverse prognostic indicator [23,24]. It could be speculated that a high CDCA2 expression is surrogate for these aggressive tumors, thus the improved clinical outcome observed upon bortezomib addition was a result of more extensive treatment of this adverse prognostic subgroup. However, double-expressor status was not of prognostic significance [16].
For functional examination of CDCA2, knockouts were established to obtain the greatest phenotypic effect in DLBCL cells, which endogenously express high levels of CDCA2. CDCA2 affected DLBCL cell proliferation with a reduction upon the loss of CDCA2. In agreement with other studies [17,25,26], we found CDCA2 to be involved in G1/S phase cell cycle transition, which has been suggested to be mediated by regulation of CCND1 [17,25,26]. In DLBCL xenograft mice, loss of CDCA2 reduced the tumor growth rate, in agreement with the slower proliferation confirmed in cell culture studies [17,25,27]. A higher Ki-67 expression was observed in CDCA2-KO tumors, suggesting genetic redundancy. Correlation between CDCA2 and Ki-67 expression have been examined in various cancer types, yet reporting contradictory findings [17,18,25]. CDCA2 and Ki-67 organize the mitotic chromosome periphery in a similar manner—by recruiting protein phosphatase 1 (PP1) to chromatin during the anaphase. The regulatory mechanisms of PP1 are identical for CDCA2 and Ki-67 [28], suggesting redundancy between these two genes.
The PI3K/AKT pathway plays a key role in modulating cell proliferation by regulating CCND1 and is crucial in DLBCL pathogenesis [29]. This study shows that the loss of CDCA2 reduces pAKT and inhibition of this pathway in SCR cells reduced the proliferation levels more when compared to CDCA2-KO cells. The link between proliferation, CDCA2, and PI3K signaling has been reported in other cancer types [17,27]. Moreover, we found that, despite this pathway influencing the bortezomib response, it is not the sole mechanism through which CDCA2 exerts its action. Inhibition of the PI3K/AKT pathway has previously been shown to confer bortezomib sensitivity in DLBCL cells [30] and while this is contradictory to our observation of reduced sensitivity upon CDCA2-KO which also diminished pAKT, it can be explained by PI3K/AKT signaling being regulated by several other genes than CDCA2.
In dose–response screens, we applied the CHOP treatment alone and in combination with bortezomib and documented CDCA2 to mediate the bortezomib response. In agreement with our findings from the analysis of the REMoDL-B data showing superior clinical outcomes of high-CDCA2-expressing patients when treated with RB-CHOP, the SCR cells possessing naturally high levels of CDCA2 displayed greater sensitivity to B-CHOP than to CHOP. In parallel to the clinical observations, the cell viability of CDCA2-KO cells was equally affected by CHOP and B-CHOP treatment in accordance with the lack of difference in outcome between the two treatment arms for DLBCL patients with low CDCA2 expressions. As CDCA2 does not affect the proteasomal activity and the PI3K/AKT pathway was disproved as sole mechanisms of action, it is reasonable to believe that the impact on bortezomib response is mediated by the high involvement of CDCA2 on proliferation. Clinically, it is the ABC and MHG molecular subclasses of DLBCL which benefit from addition of bortezomib [16], and as these subclasses constitute highly proliferative and aggressive tumors it can be assumed that high proliferation increases the susceptibility to bortezomib.
In summary, our present data are the first to demonstrate overexpression of CDCA2 in DLBCL. Functional studies confirmed a direct link between CDCA2 and DLBCL cell proliferation and bortezomib response. Moreover, we found that DLBCL patients with high expression of CDCA2 displayed superior clinical outcomes upon the addition of bortezomib to the standard R-CHOP treatment.

4. Materials and Methods

4.1. Clinical Cohorts

This study includes data from the REMoDL-B trial (R-CHOP, n = 469; RB-CHOP, n = 459) (Supplementary Table S1) and local data of normal lymph nodes (n = 6) and tonsils (n = 6), sorted B-cell subsets from tonsils, and diagnostic DLBCL biopsies (n = 86). Data summary and analysis are outlined in Supplementary Methods.

4.2. CRISPR/Cas9 Knockout

An ABC- and GCB-DLBCL cell line was selected for functional studies to reflect the REMoDL-B trial (RIVA and OCILY7, respectively). Transfection and lentiviral delivery were applied for CRISPR-mediated knockout of CDCA2 as outlined in Supplementary Methods. To validate knockout, DNA was extracted (Qiagen, #69506, Hilden, Germany) and the knockout site was PCR-amplified. Amplicons were Sanger sequenced (Eurofins Genomics, Cologne, Germany) and analyzed for indels [31]. Western blot analyses were performed as previously described [32], using 20–40 µg protein.

4.3. RNA-Sequencing

RNA-sequencing of OCILY7 SCR and CDCA2 knockout (CDCA2-KO) cells were performed in technical duplicates, as previously described [33]. GSEA was performed using hallmark gene sets from the Molecular Signatures Database for the human species [34]. DESeq2 v.1.36.0 [35] was used to estimate log2 fold changes in differentially expressed genes. Subsequently, gage v.2.46.1 [36] was used to identify enriched hallmark gene sets, considering direction of effect. Gene sets with less than 15 or more than 500 genes were excluded from the analysis.

4.4. Flow Cytometry

Analysis of cell cycle and AKT/AKT phosphorylation (Ser473, pAKT) was performed on ethanol fixed samples, whereas apoptosis was analyzed in unfixed samples using flow cytometry (SONY, SH800 Cell Sorter, Tokyo, Japan). Procedures are outlined in Supplementary Methods.

4.5. MTS Assays

Proliferation and dose response screens were performed as previously described [32,37] using seeding concentration of 0.25 × 106 cells/mL. Drug information outlined in Supplementary Methods. For combinatory drug studies, rituximab was omitted to minimize complexity and risk of bias, as addition of human serum is required for rituximab to exert its cytotoxic effect through the complement system.

4.6. Proteasome Activity Assay

Cells (15,000) were seeded in 50 µL media, in 96-well plates, prior to the addition of 10 µL of saline or bortezomib (3.5 ng/mL). After 5 h of exposure, 60 µL enzyme-specific reagent (Cell-Based Proteasome-GloTM, Promega, Singapore) was added and luminescence was measured by the Omega Fluostar plate reader (BMG LABTECH, Ortenberg, Germany). The setup included technical triplicates in biological triplicates.

4.7. Xenograft Mice

OCILY7 CDCA2-KO or SCR cells were subcutaneously injected into flanks of SCID mice. Tumor sizes were measured every 3rd-4th day and mice were sacrificed upon humane endpoint (nSCR = 6, nCDCA2-KO = 10). IHC and indel analysis was performed. Extended information provided in Supplementary Methods.

4.8. Statistical Analysis

Statistical analyses of clinical data were performed with R v.4.4. Flow data was analyzed using FlowJo v. 10.10. Cell cycle phases were defined using a univariate cell cycle model with the Watson–Pragmatic statistic. Mean fluorescence intensity (MFI) was determined with a statical feature in FlowJo. Statistical analyses of in vitro and in vivo studies were performed using GraphPad v.10.0. One-way or two-way ANOVA were performed for comparison of means between samples and p-values were adjusted using Bonferroni multiple comparison testing. * p ≤ 0.05; ** p ≤ 0.01; *** p ≤ 0.001; **** p ≤ 0.0001.

Supplementary Materials

The supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms26125596/s1. References [38,39,40,41,42] are cited in the Supplementary Materials.

Author Contributions

Conceptualization, H.D., R.F.B. and K.D.; methodology, H.D., I.I.I., L.B.T. and M.K.T.; software, R.F.B. and A.K.N.; formal analysis, H.D. and R.F.B.; investigation, H.D., A.B., I.I.I., M.Z.J., C.S., L.B.T. and M.K.T.; resources, H.D. and K.D.; data curation, H.D., R.F.B. and K.D.; writing—original draft preparation, H.D.; writing—review and editing, H.D., A.B., I.I.I., M.Z.J., C.S., A.K.N., L.B.T., M.K.T., R.F.B. and K.D.; visualization, H.D. and R.F.B.; supervision, K.D.; project administration, K.D.; funding acquisition, H.D. and K.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by The Danish Cancer Society grant number R269-A15841 and the A.P. Moeller Foundation (L-2021-00163), Denmark.

Institutional Review Board Statement

Tumor biopsies were collected in accordance with the RetroGen research protocol (Approval jr. no. N-20140099, 15 December 2014) and non-malignant tissue from healthy donors from approval N-20080062MCH, reviewed and approved by the North Denmark Region Committee on Health Research Ethics. Allowed exemption from the requirement of informed consent according to Sections 3 to 5 in Danish Act on Research Ethics Review of Health Research Projects. Informed consent was waived since this notifiable database research project did not involve any health risks and under the given conditions could not otherwise put a strain on the trial subject. Animal studies were performed in accordance with protocols approved by the Animal Experiments Inspectorate, Denmark. The maximal tumor burden of 2 cm3 permitted by the Animal Experiments Inspectorate was not exceeded.

Informed Consent Statement

Not applicable.

Data Availability Statement

The GEP datasets supporting the conclusions of this article are available in the National Center for Biotechnology Information Gene Expression Omnibus repository, GSE72648 (cell lines), GSE56315 (normal B-cell subsets), GSE109027 (local DLBCL cohort), and GSE117556 (REMoDL-B cohort).

Acknowledgments

The authors greatly appreciate the technical assistance from Helle Høholt, Department of Hematology, Aalborg University Hospital, and Hanne Krone Nielsen, Department of Health Science and Technology, Aalborg University. This study was supported by the Danish Cancer Society and the A.P. Moeller foundation.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ABCactivated B-cell
BAGSB-cell associated gene signatures
CDCA2-KOCDCA2 knockout
DLBCLdiffuse large B-cell lymphoma
GCBgerminal-center B-cell
GEPgene expression profile
GSEAgene set enrichment analysis
IHCimmunohistochemistry
IPIinternational prognostic index
KOknockout
MHGmolecular high-grade
pAKTSer473 phosphorylation of Akt
RB-CHOPrituximab, bortezomib, cyclophosphamide, doxorubicin, vincristine, prednisone
RNPribonucleoprotein
R-CHOPrituximab, cyclophosphamide, doxorubicin, vincristine, prednisone
SCRscramble
Uncunclassified

References

  1. Alizadeh, A.A.; Elsen, M.B.; Davis, R.E.; Ma, C.L.; Lossos, I.S.; Rosenwald, A.; Boldrick, J.C.; Sabet, H.; Tran, T.; Yu, X.; et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 2000, 403, 503–511. [Google Scholar] [CrossRef] [PubMed]
  2. Dybkær, K.; Bøgsted, M.; Falgreen, S.; Bødker, J.S.; Kjeldsen, M.K.; Schmitz, A.; Bilgrau, A.E.; Xu-Monette, Z.Y.; Li, L.; Bergkvist, K.; et al. Diffuse large B-cell lymphoma classification system that associates normal B-cell subset phenotypes with prognosis. J. Clin. Oncol. 2015, 33, 1379–1388. [Google Scholar] [CrossRef] [PubMed]
  3. Chapuy, B.; Stewart, C.; Dunford, A.J.; Kim, J.; Kamburov, A.; Redd, R.A.; Lawrence, M.S.; Roemer, M.G.M.; Li, A.J.; Ziepert, M.; et al. Molecular Subtypes of Diffuse Large B-cell Lymphoma are Associated with Distinct Pathogenic Mechanisms and Outcomes. Nat. Med. 2018, 24, 679–690. [Google Scholar] [CrossRef] [PubMed]
  4. Schmitz, R.; Wright, G.W.; Huang, D.W.; Johnson, C.A.; Phelan, J.D.; Wang, J.Q.; Roulland, S.; Kasbekar, M.; Young, R.M.; Shaffer, A.L.; et al. Genetics and Pathogenesis of Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2018, 378, 1396–1407. [Google Scholar] [CrossRef]
  5. Morin, R.D.; Arthur, S.E.; Hodson, D.J. Molecular profiling in diffuse large B-cell lymphoma: Why so many types of subtypes? Br. J. Haematol. 2022, 196, 814–829. [Google Scholar] [CrossRef]
  6. Copie-Bergman, C.; Cuillière-Dartigues, P.; Baia, M.; Briere, J.; Delarue, R.; Canioni, D.; Salles, G.; Parrens, M.; Balhadj, K.; Fabiani, B.; et al. MYC-IG rearrangements are negative predictors of survival in DLBCL patients treated with immunochemotherapy: A GELA/LYSA study. Blood 2015, 126, 2466–2474. [Google Scholar] [CrossRef]
  7. Sha, C.; Barrans, S.; Cucco, F.; Bentley, M.A.; Care, M.A.; Cummin, T.; Kennedy, H.; Thompson, J.S.; Uddin, R.; Worrillow, L.; et al. Molecular high-grade B-cell lymphoma: Defining a poor-risk group that requires different approaches to therapy. J. Clin. Oncol. 2019, 37, 202–212. [Google Scholar] [CrossRef]
  8. Ennishi, D.; Jiang, A.; Boyle, M.; Collinge, B.; Grande, B.M.; Ben-Neriah, S.; Rushton, C.; Tang, J.; Thomas, N.; Slack, G.W.; et al. Double-hit gene expression signature defines a distinct subgroup of germinal center B-cell-like diffuse large B-cell lymphoma. J. Clin. Oncol. 2019, 37, 190–201. [Google Scholar] [CrossRef]
  9. Wright, G.W.; Huang, D.W.; Phelan, J.D.; Coulibaly, Z.A.; Roulland, S.; Young, R.M.; Wang, J.Q.; Schmitz, R.; Morin, R.D.; Tanh, J.; et al. A Probabilistic Classification Tool for Genetic Subtypes of Diffuse Large B Cell Lymphoma with Therapeutic Implications. Cancer Cell 2020, 37, 551–568.e14. [Google Scholar] [CrossRef]
  10. Lacy, S.E.; Barrans, S.L.; Beer, P.A.; Painter, D.; Smith, A.G.; Roman, E.; Cooke, S.L.; Ruiz, C.; Glover, P.; Van Hoppe, S.J.L.; et al. Targeted sequencing in DLBCL, molecular subtypes, and outcomes: A Haematological Malignancy Research Network report. Blood 2020, 135, 1759–1771. [Google Scholar] [CrossRef]
  11. Ertrand Oiffier, B.C.; Ric Epage, E.L.; Osette Rière, J.B.; Aoul Erbrecht, R.H.; Ervé Illy, H.T.; Eda Ouabdallah, R.B.; Morel, P.; Van Den Neste, E.; Salles, G.; Gaulard, P.; et al. CHOP Chemotherapy plus Rituximab Compared with CHOP Alone in Elderly Patients with Diffuse Large-B-Cell Lymphoma. N. Engl. J. Med. 2002, 346, 235–242. [Google Scholar]
  12. Sehn, L.H.; Donaldson, J.; Chhanabhai, M.; Fitzgerald, C.; Gill, K.; Klasa, R.; MacPherson, N.; O’Reilly, S.; Spinelli, J.J.; Sutherland, J.; et al. Introduction of combined CHOP plus rituximab therapy dramatically improved outcome of diffuse large B-cell lymphoma in British Columbia. J. Clin. Oncol. 2005, 23, 5027–5033. [Google Scholar] [CrossRef] [PubMed]
  13. Nowakowski, G.S.; Chiappella, A.; Gascoyne, R.D.; Scott, D.W.; Zhang, Q.; Jurczak, W.; Özcan, M.; Hong, X.; Zhu, J.; Jin, J.; et al. ROBUST: A Phase III Study of Lenalidomide Plus R-CHOP Versus Placebo Plus R-CHOP in Previously Untreated Patients With ABC-Type Diffuse Large B-Cell Lymphoma. J. Clin. Oncol. 2021, 39, 1317–1328. [Google Scholar] [CrossRef]
  14. Younes, A.; Sehn, L.H.; Johnson, P.; Zinzani, P.L.; Hong, X.; Zhu, J.; Patti, C.; Belada, D.; Samoilova, O.; Suh, C.; et al. Randomized phase III trial of ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in non–germinal center B-cell diffuse large B-cell lymphoma. J. Clin. Oncol. 2019, 37, 1285–1295. [Google Scholar] [CrossRef]
  15. Davies, A.; Cummin, T.E.; Barrans, S.; Maishman, T.; Mamot, C.; Novak, U.; Caddy, J.; Stanton, L.; Kazmi-Stokes, S.; McMillan, A.; et al. Gene-expression profiling of bortezomib added to standard chemoimmunotherapy for diffuse large B-cell lymphoma (REMoDL-B): An open-label, randomised, phase 3 trial. Lancet Oncol. 2019, 20, 649–662. [Google Scholar] [CrossRef]
  16. Davies, A.J.; Barrans, S.; Stanton, L.; Caddy, J.; Wilding, S.; Saunders, G.; Mamot, C.; Novak, U.; McMillan, A.; Fields, P.; et al. Differential Efficacy from the Addition of Bortezomib to R-CHOP in Diffuse Large B-Cell Lymphoma According to the Molecular Subgroup in the REMoDL-B Study with a 5-Year Follow-Up. J. Clin. Oncol. 2023, 41, 2718–2723. [Google Scholar] [CrossRef]
  17. Feng, Y.; Qian, W.; Zhang, Y.; Peng, W.; Li, J.; Gu, Q.; Ji, D.; Zhang, Z.; Wang, Q.; Zhang, D.; et al. CDCA2 promotes the proliferation of colorectal cancer cells by activating the AKT/CCND1 pathway in vitro and in vivo. BMC Cancer 2019, 19, 576. [Google Scholar] [CrossRef]
  18. Zhang, Y.; Cheng, Y.; Zhang, Z.; Bai, Z.; Jin, H.; Guo, X.; Huang, X.; Li, M.; Wang, M.; Shu, X.S.; et al. CDCA2 Inhibits Apoptosis and Promotes Cell Proliferation in Prostate Cancer and Is Directly Regulated by HIF-1α Pathway. Front. Oncol. 2020, 10, 725. [Google Scholar] [CrossRef]
  19. Yu, Z.; Zhang, Y.; Shao, S.; Liu, Q.; Li, Y.; Du, X.; Zhang, K.; Zhang, M.; Yuan, H.; Yuan, Q.; et al. Identification of CDCA2 as a Diagnostic and Prognostic Marker for Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 755814. [Google Scholar] [CrossRef]
  20. Lin, X.; Zou, Z.; Zhong, J.; Wang, T.; Ma, W.; Hu, T.; Sun, W.; Xu, Y.; Eggermont, A.M.; Chen, Y. The Role of CDCA2 in tumor genesis, prognosis and future treatments. Eur. J. Cancer 2024, 211, 114308. [Google Scholar] [CrossRef]
  21. Chang, F.; Lee, J.T.; Navolanic, P.M.; Steelman, L.S.; Shelton, J.G.; Blalock, W.L.; Franklin, R.A.; McCubrey, J.A. Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation: A target for cancer chemotherapy. Leukemia 2003, 17, 590–603. [Google Scholar] [CrossRef] [PubMed]
  22. Eric Davis, R.; Brown, K.D.; Siebenlist, U.; Staudt, L.M. Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells. J. Exp. Med. 2001, 194, 1861–1874. [Google Scholar] [CrossRef] [PubMed]
  23. Johnson, N.A.; Slack, G.W.; Savage, K.J.; Connors, J.M.; Ben-Neriah, S.; Rogic, S.; Scott, D.W.; Tan, K.L.; Steidl, C.; Sehn, L.H.; et al. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2012, 30, 3452–3459. [Google Scholar] [CrossRef]
  24. Green, T.M.; Young, K.H.; Visco, C.; Xu-Monette, Z.Y.; Orazi, A.; Go, R.S.; Nielsen, O.; Gadeberg, O.V.; Mouritz-Andersen, T.; Frederiksen, M.; et al. Immunohistochemical double-hit score is a strong predictor of outcome in patients with diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2012, 30, 3460–3467. [Google Scholar] [CrossRef]
  25. Uchida, F.; Uzawa, K.; Kasamatsu, A.; Takatori, H.; Sakamoto, Y.; Ogawara, K.; Shiiba, M.; Bukawa, H.; Tanzawa, H. Overexpression of CDCA2 in Human Squamous Cell Carcinoma: Correlation with Prevention of G1 Phase Arrest and Apoptosis. PLoS ONE 2013, 8, 56381. [Google Scholar] [CrossRef]
  26. Li, F.; Zhang, H.; Li, Q.; Wu, F.; Wang, Y.; Wang, Z.; Wanh, X.; Huang, C. CDCA2 acts as an oncogene and induces proliferation of clear cell renal cell carcinoma cells. Oncol. Lett. 2020, 19, 2466–2474. [Google Scholar] [CrossRef]
  27. Wang, C.; Li, J.; Chen, Y.; Wang, X. CDCA2 triggers in vivo and in vitro proliferation of hepatocel-lular carcinoma by activating the AKT/CCND1 signaling. JBUON 2021, 26, 882–888. [Google Scholar]
  28. Kumar, G.S.; Gokhan, E.; De Munter, S.; Bollen, M.; Vagnarelli, P.; Peti, W.; Page, R. The Ki-67 and RepoMan mitotic phosphatases assemble via an identical, yet novel mechanism. Elife 2016, 5, e16539. [Google Scholar] [CrossRef]
  29. Majchrzak, A.; Witkowska, M.; Smolewski, P. Inhibition of the PI3K/Akt/mTOR Signaling Pathway in Diffuse Large B-Cell Lymphoma: Current Knowledge and Clinical Significance. Molecules 2014, 19, 14304. [Google Scholar] [CrossRef]
  30. Yuan, T.; Zhang, F.; Zhou, X.; Li, Y.; Zhang, Y.; Xu, Y.; Wang, X. Inhibition of the PI3K/AKT signaling pathway sensitizes diffuse large B-cell lymphoma cells to treatment with proteasome inhibitors via suppression of BAG3. Oncol. Lett. 2019, 17, 3719–3726. [Google Scholar] [CrossRef]
  31. Conant, D.; Hsiau, T.; Rossi, N.; Oki, J.; Maures, T.; Waite, K.; Yang, J.; Joshi, S.; Kelso, R.; Holden, K.; et al. Inference of CRISPR Edits from Sanger Trace Data. Cris. J. 2022, 5, 123–130. [Google Scholar] [CrossRef] [PubMed]
  32. Issa, I.I.; Due, H.; Brøndum, R.F.; Veeravakaran, V.; Haraldsdóttir, H.; Sylvester, C.; Brogaard, A.; Dhanjal, S.; Schmierer, B.; Dybkær, K. CRISPR-Cas9 Knockout Screens Identify DNA Damage Response Pathways and BTK as Essential for Cisplatin Response in Diffuse Large B-Cell Lymphoma. Cancers 2024, 16, 2437. [Google Scholar] [CrossRef] [PubMed]
  33. Bødker, J.S.; Sønderkær, M.; Vesteghem, C.; Schmitz, A.; Brøndum, R.F.; Sommer, M.; Rytter, A.S.; Nielsen, M.M.; Madsen, J.; Jensen, P.; et al. Development of a Precision Medicine Workflow in Hematological Cancers, Aalborg University Hospital, Denmark. Cancers 2020, 12, 312. [Google Scholar] [CrossRef]
  34. Liberzon, A.; Birger, C.; Thorvaldsdóttir, H.; Ghandi, M.; Mesirov, J.P.; Tamayo, P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015, 1, 417–425. [Google Scholar] [CrossRef]
  35. Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 1–21. [Google Scholar] [CrossRef]
  36. Luo, W.; Friedman, M.S.; Shedden, K.; Hankenson, K.D.; Woolf, P.J. GAGE: Generally applicable gene set enrichment for pathway analysis. BMC Bioinform. 2009, 10, 161. [Google Scholar] [CrossRef]
  37. Falgreen, S.; Dybkær, K.; Young, K.H.; Xu-Monette, Z.Y.; El-Galaly, T.C.; Laursen, M.B.; Bødker, J.S.; Kjeldsen, M.K.; Schmitz, A.; Nyegaard, M.; et al. Predicting response to multidrug regimens in cancer patients using cell line experiments and regularised regression models. BMC Cancer 2015, 15, 235. [Google Scholar] [CrossRef]
  38. Care, M.A.; Barrans, S.; Worrillow, L.; Jack, A.; Westhead, D.R.; Tooze, R.M. A Microarray Platform-Independent Classification Tool for Cell of Origin Class Allows Comparative Analysis of Gene Expression in Diffuse Large B-cell Lymphoma. PLoS ONE 2013, 8, e55895. [Google Scholar] [CrossRef]
  39. CRISPOR. Available online: http://crispor.tefor.net/ (accessed on 28 August 2023).
  40. CHOPCHOP. Available online: https://chopchop.cbu.uib.no/ (accessed on 28 August 2023).
  41. Sanjana, N.E.; Shalem, O.; Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 2014, 11, 783. [Google Scholar] [CrossRef]
  42. Habermann, T.M.; Weller, E.A.; Morrison, V.A.; Gascoyne, R.D.; Cassileth, P.A.; Cohn, J.B.; Dakhil, S.R.; Woda, B.; Fisher, R.I.; Peterson, B.A.; et al. Rituximab-CHOP versus CHOP alone or with maintenance rituximab in older patients with diffuse large B-cell lymphoma. J. Clin. Oncol. 2006, 24, 3121-7. [Google Scholar] [CrossRef]
Figure 1. CDCA2 expression. (A) CDCA2 mRNA expression in non-malignant lymph node (LN) and tonsil (TN) tissues, diagnostic DLBCL samples, and DLBCL cell lines. (B) CDCA2 expression in ABC/GCB classified patients of the REMoDL-B cohort. (C) CDCA2 expression in MYC/BCL2 double- and non-double-expressor patients of the REMoDL-B cohort. (D) CDCA2 expression in normal B-cell subsets FACS sorted from healthy tonsils. Wilcoxon test. * p  ≤  0.05; ** p  ≤  0.01; *** p  ≤  0.001; **** p  ≤  0.0001. ABC, activated B-cell-like; CB, centroblast; CC, centrocyte; GCB, germinal-center B-cell-like; M, memory; N, naïve; PB, plasmablast.
Figure 1. CDCA2 expression. (A) CDCA2 mRNA expression in non-malignant lymph node (LN) and tonsil (TN) tissues, diagnostic DLBCL samples, and DLBCL cell lines. (B) CDCA2 expression in ABC/GCB classified patients of the REMoDL-B cohort. (C) CDCA2 expression in MYC/BCL2 double- and non-double-expressor patients of the REMoDL-B cohort. (D) CDCA2 expression in normal B-cell subsets FACS sorted from healthy tonsils. Wilcoxon test. * p  ≤  0.05; ** p  ≤  0.01; *** p  ≤  0.001; **** p  ≤  0.0001. ABC, activated B-cell-like; CB, centroblast; CC, centrocyte; GCB, germinal-center B-cell-like; M, memory; N, naïve; PB, plasmablast.
Ijms 26 05596 g001
Figure 2. Survival analysis. Kaplan–Meier plots presenting the overall survival of DLBCL patients from the REMoDL-B cohort. Patients grouped by median split of CDCA2 mRNA expression irrespective of treatment regimen. (A) Patients with low CDCA2 expression. (B) Patients with high CDCA2 expression. (C) Patients treated with R-CHOP. (D) Patients treated with RB-CHOP.
Figure 2. Survival analysis. Kaplan–Meier plots presenting the overall survival of DLBCL patients from the REMoDL-B cohort. Patients grouped by median split of CDCA2 mRNA expression irrespective of treatment regimen. (A) Patients with low CDCA2 expression. (B) Patients with high CDCA2 expression. (C) Patients treated with R-CHOP. (D) Patients treated with RB-CHOP.
Ijms 26 05596 g002
Figure 3. Functional investigation of CDCA2. CDCA2 knockout cells were generated by RNP nucleofection (KO-1 and KO-2) and by lentiviral transduction (−/− #cloneID). (A) Knockouts validated by indel analysis and Western blotting. (B) Proliferation examined by MTS presented relative to SCR. (C) Flow cytometry assessment of cell cycle phases. Percentages of cells in the G1-phase are depicted. (D) Mean fluorescence intensity (MFI) of phosphorylated AKT (pAKT, Ser473), normalized to AKT levels. (E) Dot plot of tumor sizes (mm) for each OCILY7 xenograft mice measured every 3rd-4th day. (F) Survival (%) in days from time of injection for xenografts. (G) Ki-67 intensity normalized to DAPI. Mean value of 10 images per sample. Values are presented as mean ± standard deviation. * p  ≤  0.05; ** p  ≤  0.01; *** p  ≤  0.001; **** p  ≤  0.0001.
Figure 3. Functional investigation of CDCA2. CDCA2 knockout cells were generated by RNP nucleofection (KO-1 and KO-2) and by lentiviral transduction (−/− #cloneID). (A) Knockouts validated by indel analysis and Western blotting. (B) Proliferation examined by MTS presented relative to SCR. (C) Flow cytometry assessment of cell cycle phases. Percentages of cells in the G1-phase are depicted. (D) Mean fluorescence intensity (MFI) of phosphorylated AKT (pAKT, Ser473), normalized to AKT levels. (E) Dot plot of tumor sizes (mm) for each OCILY7 xenograft mice measured every 3rd-4th day. (F) Survival (%) in days from time of injection for xenografts. (G) Ki-67 intensity normalized to DAPI. Mean value of 10 images per sample. Values are presented as mean ± standard deviation. * p  ≤  0.05; ** p  ≤  0.01; *** p  ≤  0.001; **** p  ≤  0.0001.
Ijms 26 05596 g003
Figure 4. Drug response analysis of CDCA2-KO cells. (AC) Bortezomib dose–response screens. Cell viability assessed by MTS and presented relative to the untreated control. (D,E) Apoptosis examined by flow cytometry upon bortezomib exposure. Apoptotic cells (Apop+) including early and late apoptotic stages presented relative to the untreated control. (F) CHOP and B-CHOP dose–response analysis in OCILY7. Cell viability assessed by MTS and presented relative to untreated control. KO-1 and KO-2 are heterogenous cell populations created by RNP nucleofection. −/− #cloneID represents monoclonal populations established by lentiviral transduction followed by single cell expansion. Values are presented as mean ± standard deviation. * p  ≤  0.05; ** p  ≤  0.01; *** p  ≤  0.001; **** p  ≤  0.0001.
Figure 4. Drug response analysis of CDCA2-KO cells. (AC) Bortezomib dose–response screens. Cell viability assessed by MTS and presented relative to the untreated control. (D,E) Apoptosis examined by flow cytometry upon bortezomib exposure. Apoptotic cells (Apop+) including early and late apoptotic stages presented relative to the untreated control. (F) CHOP and B-CHOP dose–response analysis in OCILY7. Cell viability assessed by MTS and presented relative to untreated control. KO-1 and KO-2 are heterogenous cell populations created by RNP nucleofection. −/− #cloneID represents monoclonal populations established by lentiviral transduction followed by single cell expansion. Values are presented as mean ± standard deviation. * p  ≤  0.05; ** p  ≤  0.01; *** p  ≤  0.001; **** p  ≤  0.0001.
Ijms 26 05596 g004
Figure 5. (A) Proteasomal activity of SCR and CDCA2-KO OCILY7 cells upon exposure to 3.5 ng/mL bortezomib for 5 h. Caspase, chymotrypsin, and trypsin activity presented as luminescence read-out of bortezomib cells relative to untreated cells. (B) Proliferation of SCR and CDCA2-KO OCILY7 cells examined by MTS. Cells were exposed to saline or 10 nM LY294002 for 48 h. Data presented relative to SCR in untreated condition. (C) Combinatory dose–response analysis of bortezomib and LY294002 (10 nM). Cell viability assessed by MTS and presented relative to untreated. −/− #cloneID represents monoclonal populations established by lentiviral transduction followed by single cell expansion. Values are presented as mean ± standard deviation. *** p  ≤  0.001; **** p  ≤  0.0001.
Figure 5. (A) Proteasomal activity of SCR and CDCA2-KO OCILY7 cells upon exposure to 3.5 ng/mL bortezomib for 5 h. Caspase, chymotrypsin, and trypsin activity presented as luminescence read-out of bortezomib cells relative to untreated cells. (B) Proliferation of SCR and CDCA2-KO OCILY7 cells examined by MTS. Cells were exposed to saline or 10 nM LY294002 for 48 h. Data presented relative to SCR in untreated condition. (C) Combinatory dose–response analysis of bortezomib and LY294002 (10 nM). Cell viability assessed by MTS and presented relative to untreated. −/− #cloneID represents monoclonal populations established by lentiviral transduction followed by single cell expansion. Values are presented as mean ± standard deviation. *** p  ≤  0.001; **** p  ≤  0.0001.
Ijms 26 05596 g005
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

Due, H.; Brogaard, A.; Issa, I.I.; Jakobsen, M.Z.; Sylvester, C.; Nøhr, A.K.; Thomsen, L.B.; Thomsen, M.K.; Brøndum, R.F.; Dybkær, K. Overexpression of CDCA2 in Diffuse Large B-Cell Lymphoma Promotes Cell Proliferation and Bortezomib Sensitivity. Int. J. Mol. Sci. 2025, 26, 5596. https://doi.org/10.3390/ijms26125596

AMA Style

Due H, Brogaard A, Issa II, Jakobsen MZ, Sylvester C, Nøhr AK, Thomsen LB, Thomsen MK, Brøndum RF, Dybkær K. Overexpression of CDCA2 in Diffuse Large B-Cell Lymphoma Promotes Cell Proliferation and Bortezomib Sensitivity. International Journal of Molecular Sciences. 2025; 26(12):5596. https://doi.org/10.3390/ijms26125596

Chicago/Turabian Style

Due, Hanne, Asta Brogaard, Issa Ismail Issa, Maja Zimmer Jakobsen, Cathrine Sylvester, Anne Krogh Nøhr, Louiza Bohn Thomsen, Martin Kristian Thomsen, Rasmus Froberg Brøndum, and Karen Dybkær. 2025. "Overexpression of CDCA2 in Diffuse Large B-Cell Lymphoma Promotes Cell Proliferation and Bortezomib Sensitivity" International Journal of Molecular Sciences 26, no. 12: 5596. https://doi.org/10.3390/ijms26125596

APA Style

Due, H., Brogaard, A., Issa, I. I., Jakobsen, M. Z., Sylvester, C., Nøhr, A. K., Thomsen, L. B., Thomsen, M. K., Brøndum, R. F., & Dybkær, K. (2025). Overexpression of CDCA2 in Diffuse Large B-Cell Lymphoma Promotes Cell Proliferation and Bortezomib Sensitivity. International Journal of Molecular Sciences, 26(12), 5596. https://doi.org/10.3390/ijms26125596

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