Introduction
Diffuse Large B-cell Lymphoma (DLBCL) is the most common lymphoma, presenting as a highly heterogenous disease with diverse clinical outcomes and features [
1,
2]. The standard-of-care R-CHOP (rituximab with cyclophosphamide, vincristine, doxorubicin, and prednisone) immunochemotherapy or the emerging Polatuzumab Vedotin combination result in long-term remission in more than 60% of patients [
3,
4]. However, patients with refractory or relapsing disease (rrDLBCL) face a particularly dismal outlook, specifically those experiencing an event within the first 24 months after diagnosis, despite the success of autologous stem cell transplant (ASCT) [
5‐
7]. The extremely promising advent of tumor-targeting Chimeric Antigen Receptor Cells (CAR-T) has vastly improved the prospects of this population, significantly increasing rates of progression-free survival [
8‐
13]. Nevertheless, genomic identification of patients that would not benefit from CAR-T therapy remains a crucial need given the substantial biological and financial toxicity associated with the procedure, allowing clinicians to explore personalized treatment avenues for these patients [
14,
15].
A multitude of studies have explored the genomic landscape of DLBCL, each highlighting specific gene expression, DNA, or tumor microenvironment (TME) features. DLBCL was originally classified on the basis of the Cell of Origin (COO) classification, designating two groups: Germinal Center B-cell (GCB) and Activated B-cell (ABC), with the latter presenting more aggressively [
16,
17]. Next, studies used DNA alterations to classify distinct groups of DLBCL [
18‐
21]. Future DNA-focused studies were able to elucidate the rrDLBCL landscape, providing further clarity towards disease evolution [
22‐
26]. Notable DNA alterations predictive of CAR-T response include those to
TP53,
SMARCA4,
TMEM30A, and
MYC, among others [
27‐
32]. Intrinsic gene expression and characteristics of CAR-T products serve as another determinant of success [
33,
34]. Lastly, gene expression and single-cell-RNA-seq were utilized to explore and classify DLBCL tumors on the basis of their TME “Ecotype” [
35‐
37]. These highlighted the inferior outlooks faced by patients with a “depleted” or “cold” TME devoid of helpful immune activity, even when treated with CAR-T [
38]. The absence of native CD8 T-cells specifically portends poor CAR-T response, alongside the infiltration of M2 macrophages and T-regs [
39‐
44]. The loss of CD19 itself represents a viable resistance pathway, but alterations to key T-cell modulators such as ILR7, CD58, and the IFR1-STAT1 are also suspected to interfere [
45‐
48]. Applying these classification components to better forecast patient CAR-T response may improve precision approaches when combined with established predictive clinical data such as age, staging, and previous treatment [
49,
50].
DNA alterations to the MYC oncogene, notably mutations or translocation events capable of boosting gene expression, have long been predictive of inferior de-novo DLBCL survival and have been implicated with CD19-directed CAR-T failure in early studies [
29‐
32,
51‐
54]. MYC activation engineers a wide array of cellular responses that promote proliferation, survival, and notably immune evasion [
55‐
57]. MYC activation is often designated as a key event in most DNA classification or risk clusters within DLBCL, specifically GCB/EZB/C3 [
18,
20,
21,
58]. MYC’s association or facilitation with poor TME landscapes that favor the tumor represent an additional barrier of successful CAR-T application [
59,
60]. Further exploration of these networks, notably within the scRNA (single-cell RNA) landscape in rrDLBCL, is warranted to continue improving our understanding of CAR-T biology and interactions.
Identification of biological landscapes predictive of poor CAR-T response demands the application of genomically-targeted drug therapies to corner deleterious pathways within patients that would not be expected to have successful CAR-T outcomes. Direct inhibition of MYC remains elusive, but emerging therapeutics target its precursors and after-effects. Inhibition of the XPO1 exportin gene responsible for MYC RNA export from the nucleus has established itself as an exciting avenue, with its expression alone a poor sign for clinical outcomes [
61‐
63]. Equally exciting is the recent development of the efficacious CDK9 inhibitor AZD4573, which aggressively mutes many of the downstream proliferation effects of MYC gain [
64,
65]. Critically, few studies have examined the associations between inferior genomic CAR-T determinants, how they interact de-novo and then in rrDLBCL, and how they may be addressed. Patients with a poor CAR-T and ASCT response phenotype may represent excellent candidates for targeted therapy combinations if these molecular partnerships were better understood.
Herein, we present integrative analyses detailing the partnerships between MYC alterations, subsequent loss of CD8 T-cell presence, and an examination of a novel targeted therapeutic combination. This research provides valuable insights into the molecular pathogenesis of DLCBL and rrDLBCL and provides preliminary evidence for exploration of potentially effective personalized treatments.
Materials and methods
De-novo DLBCL dataset assemblies
A pooled dataset from 4 publicly-available studies was assembled to measure CAR-T progression vs.
MYC status. One study (Sworder et al.) relied on ctDNA sequencing while the others utilized biopsies [
27,
29,
30,
32,
66]. Next, A publicly-available de-novo DLBCL dataset containing 418 patient profiles complete with COO, Overall Survival (OS), and Progression-free Survival (PFS) was integrated alongside matched data for 70 DNA alterations and 1397 genes with expression values (Xu-Monette 2020) [
67]. This approach mirrors that of our previous work [
68]. Within the cohort, 121 patients experienced EFS24 failure (28.95%), 109 were absent CD8 T-cell deconvolution signals (26.08%), 202 were classified as GCB COO (48.33%), and 49 harbored
MYC alterations or had high expression (+ 1 standard deviation; 11.72%). Gene expression targets were filtered out if they did not contain at least 3 non-zero values. Gene expression values were normalized based on UHR FPKM NS and UHR MS values. CAR-T positivity scores and gene expression changes based on response were assembled from publicly-available supplemental material [
39,
69].
ScRNA DLBCL dataset assemblies and processing
To further investigate the relationships between high MYC expression and biological pathways of both the malignant B-cells and the immune cells of the tumor microenvironment, we analyzed single-cell RNA (scRNA) data of seven DLBCL patients from two publicly available datasets. Four patients were taken from Steen et al. (GEO: GSE182436) and three were from Roider et al. (Table
1) [
36,
70].
Table 1
Statistics for datasets included in ScRNA analyses
Cells from both datasets were filtered based on total counts, total number of genes per cell, percent counts ribosomal, and percent counts mitochondrial. The filtering was conducted by patient. A given cell was excluded if it fell outside the range of the median ± 5 standard deviations for total counts, total number of genes per cell, percent ribosomal counts, or percent mitochondrial counts. This pre-processing was performed to remove potential doublets and dead cells. From seven total patients, we filtered out 1,511 cells, leaving us with 26,938 cells to analyze. After filtering, any cells that were without a cell type annotation were annotated by K-Nearest Neighbors (KNN) labeling.
ScRNA gene set analyses
To evaluate which biological pathways/functions were active in our scRNA data, we employed a rank-based gene set scoring method. In total, we computed scores for 7,608 gene sets from the GO (C5) collection of the Molecular Signatures Database (MSigDB) [
71,
72]. Scores for all gene sets were computed for each cell in our analyzed dataset. For a cell containing N expressed genes, ranked from 1 (lowest) to N (highest) based on total counts, the enrichment score for a given gene set S was computed as.
$$\eqalign{& {\rm{Enrichment}}\,{\rm{Score}} \cr & {\rm{ = }}\,{{\sum\limits_{i \in S} {rank\,(i)} } \over {\sum\limits_{j = 1}^{|S|} {(N -|S| + j)} }} \cr} $$
where rank(i) is the rank of gene i within the particular cell, and |S| is the total number of genes in the gene set. This methodology is similar to single-sample gene set enrichment analysis (ssGSEA) [
73], but is simplified to be self-contained (independent of comparisons to a background genes outside the gene set or other gene sets) and more interpretable. Under this method, enrichment scores are guaranteed to fall within the range of 0 to 1. Finally, for all analyzed gene sets, we computed the spearman correlation coefficient between the gene set and MYC expression in the malignant B cells across all seven patients.
Differential expression, gene ontology, immune Deconvolution analyses, and ecotyping
Differential gene expression was performed using the Broad Institute’s GenePattern suite, specifically Morpheus [
74,
75]. Marker Selection analysis was performed across the 1397-gene panel vs. specific annotations to obtain FDR significance and t-values for each. FDR values below 0.05 were considered significant. Nearest Neighbor analysis was utilized to obtain gene-specific Pearson values and create similarity matrices. Gene pathway ontology and gene-gene interaction network analyses were completed using ToppFunn functional enrichment tools [
76]. UCLA’s Gene Expression Deconvolution Interactive Tool (GEDIT) was utilized to infer and display the relative presence of 22 immune components from the de-novo 418-patient Xu-Monette et al. cohort and the CAR-T-treated Scholler et al. cohort [
77]. Next, the Lymphoma Ecotyper tool developed from the Steen et al. study was utilized to designate patient cell state and ecotype in the Xu-Monette cohort [
36].
Cell lines
The DHL16 (CVCL_1890), Karpas-422 (CVCL_1325), Ly3 (CVCL_8800), and U2932 (CVCL_1896) cell lines were gifted by Dr. Javed Iqbal and Dr. Alyssa Bouska from the University of Nebraska Medical Center. The DHL6 (CVCL_2206) cell line was gifted by Dr. Anne Novak from the Mayo Clinic, Rochester. The DHL4 (CVCL_0539) cell line was obtained from ATCC (CRL-295). All cell lines were known to be STR-authenticated. Cells were incubated at 37˚ (C) with 5% CO2. DHL4, DHL6, DHL16, Karpas-422, and U2932 cell lines were maintained in RPMI-1640 media with 10% FBS and 1% penicillin/streptomycin. Ly3 was maintained in IMDM media with 20% FBS and 1% penicillin/streptomycin. Known DNA variants were confirmed via DepMap. Cell Line RNA expression values were acquired from the Hardee et al. 2013 study (GSE50721) [
78].
Small molecule treatments
The XPO1 inhibitor Selinexor (KPT-330) was obtained from SelleckChem. The CDK9 inhibitor AZD4573 was obtained from Aobious. Single-molecule ED50 assays were conducted across 3 triplicate wells in a 96-well plate for each of the following treatments (in nM): 20,000, 10,000, 5000, 2500, 1250, 625, 313, 156, 78, and 0.4% DMSO control. Each well contained approximately 10,000 tumor cells, at a concentration of 200,000 cells/mL. Plates were incubated for 96 h. Alamar Blue viability reagent was added to each well at 10% concentrations and allowed to incubate for 3 h. Fluorescence results were read using a SpectraMax iD5 multi-mode microplate reader. Each cell vs. treatment value has between 3 and 9 replicates across all assays. Synergy combination assays were performed using a similar protocol but adjusted for customized 4 × 4 dose grids. Synergy results were averaged among 2 replicates on each plate.
Microscopy
Cells treated with specific control, single, and combination doses were visualized using a Leica Brightfield Stereoscope. Z-layer images were stacked to create 3D representations.
Statistical analyses and figure creation
GraphPad Prism was utilized for the following analyses: Fisher’s Exact test, Kaplan-Meier survival analysis, Welch’s t-tests, and multiple-comparison-corrected t-tests and One-way ANOVAs. FDR and p values were considered statistically significant below 0.05. Cell viability results were normalized to average blank Alamar Blue border wells and then to the DMSO control averages. Single-molecule treatment assay ED50 values were calculated using GraphPad Prism, constraining values between 1.0 and 0. Bliss synergy combination values were calculated using the Synergy Finder tool [
79]. GraphPad Prism and Adobe Illustrator software were used to create and annotate figures, respectively.
Discussion
The advent of CAR-T therapeutics represents a paradigm shift in the treatment of DLBCL, having ushered in dramatically improved rates of progression-free survival for those refracting or relapsing from first-line regimens. Nonetheless, the molecular and microenvironmental bases of poor CAR-T response remain unclear due to the heterogenous presentation of the disease. In particular, the assignment of targeted therapies for patients with tumor profiles predictive of failure remain a critical need. Emerging characteristics associated with treatment failure continue to emerge, such as TP53 or MYC alterations, depleted or cold TMEs, protumoral immune infiltration, and increased metabolomic potential. We sought to provide a preliminary yet substantial overview of the partnerships between MYC alteration and expression, CD8 T-cell absence, and other CAR-T-informing genomic features leveraging multiple sources of data alongside in-vitro validation and translation that may be combined with other key predictive clinical data such as age, staging, and previous treatment.
As
MYC alterations within rrDLBCL patients are all but confirmed to portend inferior patient response, even against CAR-T treatment (Fig.
1a), we sought to elucidate more about this landscape, specifically the transition between de-novo presentation and CAR-T rejection [
29‐
31]. Known were the aggressive gains in proliferation and survival potential associated with greater MYC expression (Fig.
1b), but less was known about MYC’s role facilitating a “depleted” or “cold” immune phenotype within the rrDLBCL TME [
60]. Upon integrating our de-novo dataset with the rrDLBCL post-CAR-T treatment dataset from Scholler et al., we observed a striking triple association between these factors and EFS24 failure shared by a wide and powerful array of immune and T-cell facilitation genes (Fig.
1c). These losses suggest that a substantial portion of the poor CAR-T rrDLBCL phenotype may be carried over from the molecular landscape at disease presentation.
With key T-cell partners such as IL7R, CD58, ITK, STAT1, and IRF1 lost in de-novo tumors bearing greater MYC and in CAR-T failures, we next sought to measure proportions of T-cells, among other immune components, and interrogate treatment failure associations. Using deconvolution of both the de-novo and rrDLBCL gene expression datasets, CD8 T-cell loss was inferred as the most substantial collective loss between de-novo patients with increased MYC and CAR-T failure patients (Fig.
2a). Unexpectedly, Neutrophils were also lost on all three fronts, representing an intriguing future direction for study. With 26.1% of the de-novo cohort absent of CD8 T-cell deconvolution signal, it would suggest that Depleted TME phenotypes could be more common at onset than assumed (Fig.
2b). CD8-absence (
N = 109) partnered with increased MYC presence may provide the proliferation and survival strength needed for these tumors to escape first-line regimens, especially alongside increases with key oncogenic and DNA repair genes (Fig.
2d). Indeed, when isolating the 19 High-MYC/CD8-Absent patients, key gains in oncogenic interactions manifest alongside T-cell activation losses (Fig.
3a-b). Most importantly, truly inferior de-novo patient survival is only observed when High/Altered MYC status and CD8 T-cell absence co-occur (Fig.
3b), as patients bearing only one of the associations mirror typical survival patterns within the dataset. These results suggest that de-novo tumors presenting High/Altered MYC combined with CD8 T-cell loss essentially synergize and may carry a poor CAR-T phenotype from start, thus emerging as candidates for targeted therapies.
Our correlation analysis reinforces MYC’s well-known role as a central regulator of tumorigenesis in DLBCL by promoting both intrinsic tumor growth mechanisms and tumor microenvironment regulating factors that support disease progression (Fig.
4a). Enhanced expression of growth and inflammatory pathways indicates a more active and aggressive disease phenotype, which explains the worsened prognosis observed in high MYC patients. Moreover, the increased antigen processing and presentation by MYC expressing B cells suggests the immunosuppressive strategy of pro-tumor immune cells such as T-regs and M2 macrophages. However, this mechanism may have a dual effect in patients with competent CD8 T cell populations, and may also lead to recruitment and proliferation of these anti-tumor cells, This idea is further supported by the association between MYC and production of T cell promoting interleukins IL-2 and IL-12. And so, the notion of a dual effect in the tumor microenvironment from high MYC expression lends a possible explanation to why patients that are high MYC/CD8-present show the best prognosis compared to all other groups, whereas high MYC/CD8-absent patients fare the worst.
We also characterized patient DLBCL1 (from the Steen dataset) as a “high MYC” patient as the average MYC count of their cells (6.32) was significantly higher than the six other patients (average MYC counts ranged between 0.10 and 2.60) (Fig.
4b). Of the seven patients, only the high MYC patient showed exceptionally low T cell counts in the scRNA data (of the analyzed 3,832 cells from patient DLBCL1, only 5 were labeled as T cells) as all other patients in the dataset had at minimum 200 T cells. It is unclear whether this observation is a product of technical artifacts, such as differences in cell capture efficiency, sequencing depth, or flawed annotations, or is a true biological phenomenon, potentially reflecting an immunosuppressive tumor microenvironment unique to this patient. Nonetheless, this observation is worth mentioning, especially due to the known relationship between high MYC and CD8 absence. On final note, the two patients with the lowest average MYC counts in their malignant B cells (DLBCL111 and DLBCL2) were the two patients with the lowest average enrichment scores for key immune response pathways Activation of Immune Response (GO:0002253), Inflammatory Response to Antigenic Stimulus (GO:0002437), and Positive Regulation of Cytokine Production (GO:0001819) in their T cell distributions. Thus, there is evidence to suggest that MYC expression of the B cells may drive immune system recognition and activation, at least in the scenario of present T cell populations.
Seeking to provide a genomically-targeted second-line regimen for these patients, we applied the CDK9 inhibitor AZD4573 and XPO1 inhibitor Selinexor in cell line models (Fig.
5a). This combination was rationalized by the significantly increased CDK9 and XPO1 interactions associated with greater differentially-expressed genes in the High-MYC/CD8-Absent patients (Fig.
3a). Targeting MYC downstream and before its translation to protein were both observed to substantially diminish DLBCL cell viability as single agents, although these effects could be expected based on previous successful in-vitro applications [
61,
65]. We followed these results by combining the agents with the intention of cornering the effects of this unfavorable phenotype. We observed synergistic results across all 6 cell lines (Fig.
5c-d), making us the first to report the synergistic combination of AZD4573 and Selinexor vs. DLBCL cell lines. Notably, the strongest synergy was felt by the cell line expressing the highest amount of CDK9, Ly3, a particularly aggressive cell line, and our weakest synergy was observed in cell lines with lower-than-average CDK9 (DHL4, U2932), perhaps indicating that CDK9 itself represents the best biomarker for proper application of this combination. These preliminary results support the exploration of this combination in further studies in order to provide rrDLBCL patients expressing greater MYC (and potentially a shared absence of CD8 T-cells) a personalized approach.
Forthcoming studies should address specific caveats encountered in our report. To begin, validation among other large de-novo and rrDLBCL datasets (specifically additional scRNA datasets) with integrated DNA alteration and gene expression remains an important follow up for supporting these results. That, and our data were not able to integrate key clinical factors such as patient age, staging, and if treatment differed from R-CHOP (although all cases were documented to receive it). Equally, cell line results are preliminary, and additional in-vitro studies would augment our findings, perhaps leading to in-vivo studies with animal models. Similarly, both molecules require further molecular validation of their patient safety and efficacy profiles given their unique mechanisms of action, ideally utilizing knockout and induction models. CDK9 and XPO1 inhibition may also reduce the viable T-cell populations, so this treatment could only be applied if the TME already represented a Depleted Ecotype. Next, Immune deconvolution represents a suitable tool for interrogating the presence of specific components, but single-cell RNA seq or flow cytometry of live biopsies would provide the most clarity. Lastly, these analyses are primarily integrated from other DLBCL informatics studies and sources, denying the potential for a fully in-house experiment.
In conclusion, we report a concerning network portending poor CAR-T responses between increased MYC expression and a depleted immune presence, most notably CD8 T-cell absence, that may be addressed by genomically-targeted therapeutic molecules. These results provide novel informatic insights within a genomic network of importance for CAR-T application and may therefore be used to identify high-risk phenotypes and better inform future treatment teams prior to CAR-T administration in combination with clinical prognostic factors.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.