Key Points
Regulatory regions undergoing de novo chromatin activation in CLL are associated with both progressive and indolent disease.
A balance score between proprogression and proindolence chromatin signatures is a powerful independent prognostic factor in CLL.
Visual Abstract
Previous studies have reported that chronic lymphocytic leukemia (CLL) shows a de novo chromatin activation pattern compared with normal B cells. Here, we explored whether the level of chromatin activation is related to the clinical behavior of CLL. We identified that, in some regulatory regions, increased de novo chromatin activation is linked to clinical progression, whereas, in other regions, it is associated with an indolent course. We next developed 2 prognostic scores for progressive and indolent disease, respectively, calculated a single score representing the balance between them, and further generated surrogate scores based on gene and protein expression of the target genes. The balance score outperformed the clinical impact of the 2 individual scores, because it seemed to capture the prognostic information provided by each of them. Biologically, CLLs with higher balance score showed increased activation of tumor necrosis factor alpha (TNF-α)/NF-κB and mTOR signaling pathways. Regulatory programs related to progression were predominantly activated in the lymph node microenvironment, whereas those linked to indolent disease appeared to be microenvironment independent. Finally, we thoroughly validated the balance score as a powerful and independent quantitative prognostic factor for time to first treatment across independent CLL cohorts and data modalities, such as chromatin, transcriptome, or proteome data. Our findings support the concept that de novo acquisition of chromatin changes in CLL cells plays a dual biological role, and the balance between proprogression and proindolence is a strong independent determinant of CLL prognosis.
Introduction
Chronic lymphocytic leukemia (CLL) is the most prevalent leukemia in Western countries and is characterized by extensive genetic diversity and a wide spectrum of clinical outcomes.1-3 The mutational load within the immunoglobulin heavy chain variable (IGHV) genes is the most recognized and validated prognostic marker, classifying patients into unmutated CLL (U-CLL), which lacks or shows low IGHV somatic hypermutation, and CLLs with mutated IGHV (M-CLL). The former clearly shows a worse clinical behavior than the latter.4,5 Furthermore, TP53 alterations are widely described as markers of poor prognosis.3 However, despite the identification of genetic driver alterations and transcriptional programs,6-10 the precise molecular mechanisms underlying CLL development and progression are not fully understood.
Over the last decades, epigenetics has emerged as a pivotal feature to understand cancer, including pathogenesis, evolution, clinical behavior, and therapy of several tumor types.11 In CLL, DNA methylation signatures with clinical impact have been linked with the cell of origin, proliferative history, evolutionary dynamics, and deregulated pathways.12-17 In particular, 3 epigenetic subtypes were identified based on DNA methylation imprints of normal B cells at different maturation states (from less to more aggressive; memory-like/high-programmed, intermediate, and naïve-like/low-programmed CLLs).12,13 More recently, the regulatory chromatin landscape of CLL and mature B cells has been described,18-21 identifying a signature of regulatory regions activated exclusively in CLL cells.18 Therefore, this signature was interpreted as a molecular feature related to all CLL cases, which contrasts with the overall biological heterogeneity of the disease. However, the possible link between the level of chromatin activation and clinical outcome in CLL has not been explored in any of the published chromatin profiling studies.
In this study, we evaluated whether the magnitude of de novo chromatin activation, based on chromatin immunoprecipitation with massively parallel DNA sequencing (ChIP-seq) profiles of histone 3 lysine 27 acetylation (H3K27ac), was linked with the clinical features of patients with CLL. Our findings unveil an unexpected dual role of chromatin activation, promoting either progressive disease (PD) or indolent disease (ID) clinical course. These signatures were integrated into a single balance score with strong clinical impact that was orthogonally validated with transcriptome and proteome data in independent series.
Methods
An extended version is provided in the supplemental Methods, available on the Blood website.
Patients
We studied a total of 650 patients with CLL from 7 independent cohorts, spanning 3 data modalities (chromatin activation, gene and protein expression, and generated on sorted CLL cells). These cohorts, named ES6,18,22-24 (n = 280) for the Spanish cohort, DE110 (n = 140) and DE225 (n = 45) for the German cohorts 1 and 2, US126 (n = 28), US227 (n = 21) and US38 (n = 91) for the United States cohorts 1, 2 and 3, and UK28 (n = 45) for the United Kingdom cohort, are detailed in supplemental Tables 1 and 2. Most of the data were mined from the source references. To validate CLL prognosis, we focused on 439 patients with available clinical information. All patients provided written informed consent, and the study was performed in accordance with the Declaration of Helsinki and was approved by the local ethics review boards.
Bioinformatic analyses
H3K27ac ChIP-seq profiles for CLL and B cells were obtained from Beekman et al18 and reanalyzed (supplemental Methods). CLL raw FASTQ files or gene-level counts data from bulk and single-cell RNA sequencing (RNA-seq) were obtained from previous publications6,8,10,18,22-28 and mined using state-of-the-art computational tools (supplemental Methods).
Definition of PD, ID, and balance scores
Using normalized log-transformed data of chromatin activity, gene expression, or protein expression, we initially estimated 2 values per sample: the PD and ID scores from the PD and ID signatures, respectively. Specifically, we calculated the fold change of each chromatin region/gene relative to the median of the cohort (ie, in log-transformed data, the fold change is estimated by subtraction). We next computed the median of the fold changes of all the regions/genes per signature to derive the PD and ID scores, respectively. Then, we calculated the balance score by subtracting the ID score from the PD score, which, because the data are log-transformed, effectively represents the PD-to-ID ratio.29 Finally, we calculated the z score of the balance score to study clinical associations and enable comparability across Cox regression models, irrespective of the data set (supplemental Figure 1).
Survival analyses
The primary end point for survival analysis was time to first treatment (TTFT). We included all cases with available clinical data across cohorts with sufficient treatment-naïve samples (n > 30), sourced from 5 previous studies.6,8,10,18,25 H3K27ac peak signal association with TTFT was determined using Cox scores with the SAM function from the samr30 R package (v3.0). Log-rank tests were used to compare Kaplan-Meier curves. Likelihood ratio tests evaluated the fit of Cox regression models. Prognostic variables were included in the analysis only if they had a minimum of 5 events. Permutation tests were used to test the association of the balance score with TTFT in the discovery series (supplemental Methods).
Results
Characterization of de novo chromatin activation in CLL and its clinical impact
We initially mined H3K27ac ChIP-seq data from 104 CLLs and 15 mature B-cell subpopulations.18 Within the 498 predefined de novo active regulatory regions,18 we identified 434 individual H3K27ac peaks de novo acquired in CLL (Figure 1A; supplemental Table 3). Although these regions seem to represent a signature of CLLness, we observed differences in the mean H3K27ac signal and the percentage of individual peaks detected for each sample (Figure 1A), indicating that the level of de novo chromatin activation varies among individual CLL samples.
De novo enrichment in H3K27ac in CLL. (A) Heat map depicting the H3K27ac signal intensities of genomic regions de novo enriched in H3K27ac in CLL. Signal intensities are presented as z scores (calculated per row) of the variance stabilizing transformation (VST) signal for CLL and B-cell samples. The mean log2 signal and the proportion of peaks detected in each sample are indicated below. The right bar shows the 2 signatures derived from hierarchical clustering. (B-C) Heat maps illustrating the H3K27ac signal intensities in signature S1 (B) and signature S2 (C). Signal intensities are represented as row z scores of VST signal. A box plot (right) compares the mean z scores between U-CLL and M-CLL samples in each cluster. Statistical significance was assessed using 2-tailed Student t tests. (D) Volcano plot showing hazard ratios (HRs) and P values for associations between H3K27ac enrichment in peaks and TTFT. Peaks from signatures S1 and S2 are differentiated by color. (E-F) The number of H3K27ac peaks (left) whose enrichment is associated with shorter or longer TTFT using univariate Cox scores in signature S1 (E) and signature S2 (F). Peaks’ association with shorter or longer TTFT was defined by the estimated HR >1 or <1, respectively. Kaplan-Meier curves (right) show the groups based on the H3K27ac enrichment levels of representative regions from each cluster; chr13:62.209.280-62.210.122 (E) and chr1:203.468.258-203-470.929 (F). Higher and lower H3K27ac groups were defined using the maxstat rank statistic–based cutoff. P values were obtained with log-rank tests. Note, HR and P values in panel D were computed with the coxph function from the survival31 R package, whereas the analyses in panels E-F used the samr30 R package for greater statistical power, potentially causing minor discrepancies between these analyses. ∗∗∗∗P < .0001. GCBC, germinal center B cell; MBC, memory B cell; ns, not significant; NBC-PB, naïve B cell from PB; NBC-T, naïve B cell from tonsil; PCT, plasma cell from tonsil.
De novo enrichment in H3K27ac in CLL. (A) Heat map depicting the H3K27ac signal intensities of genomic regions de novo enriched in H3K27ac in CLL. Signal intensities are presented as z scores (calculated per row) of the variance stabilizing transformation (VST) signal for CLL and B-cell samples. The mean log2 signal and the proportion of peaks detected in each sample are indicated below. The right bar shows the 2 signatures derived from hierarchical clustering. (B-C) Heat maps illustrating the H3K27ac signal intensities in signature S1 (B) and signature S2 (C). Signal intensities are represented as row z scores of VST signal. A box plot (right) compares the mean z scores between U-CLL and M-CLL samples in each cluster. Statistical significance was assessed using 2-tailed Student t tests. (D) Volcano plot showing hazard ratios (HRs) and P values for associations between H3K27ac enrichment in peaks and TTFT. Peaks from signatures S1 and S2 are differentiated by color. (E-F) The number of H3K27ac peaks (left) whose enrichment is associated with shorter or longer TTFT using univariate Cox scores in signature S1 (E) and signature S2 (F). Peaks’ association with shorter or longer TTFT was defined by the estimated HR >1 or <1, respectively. Kaplan-Meier curves (right) show the groups based on the H3K27ac enrichment levels of representative regions from each cluster; chr13:62.209.280-62.210.122 (E) and chr1:203.468.258-203-470.929 (F). Higher and lower H3K27ac groups were defined using the maxstat rank statistic–based cutoff. P values were obtained with log-rank tests. Note, HR and P values in panel D were computed with the coxph function from the survival31 R package, whereas the analyses in panels E-F used the samr30 R package for greater statistical power, potentially causing minor discrepancies between these analyses. ∗∗∗∗P < .0001. GCBC, germinal center B cell; MBC, memory B cell; ns, not significant; NBC-PB, naïve B cell from PB; NBC-T, naïve B cell from tonsil; PCT, plasma cell from tonsil.
Next, we applied unsupervised hierarchical clustering and detected 2 main sets of de novo active chromatin regions: signature S1 comprising 130 peaks and signature S2 comprising 304 peaks (Figure 1A; supplemental Table 3). Interestingly, this analysis showed that the H3K27ac signal in S1 is clearly related to the IGHV mutational status (higher in U-CLL than in M-CLL; P < .0001; Figure 1B). In contrast, the regions of S2 did not show global differences between IGHV groups (P = .3), although a subtle clustering of cases based on IGHV status could be observed in the heat map (Figure 1C). Because some of the de novo active chromatin regions undergo a higher level of activation in U-CLL than in M-CLL, we postulated that chromatin activation levels may correlate with clinical behavior. Therefore, we associated each region with TTFT as a surrogate for CLL progression32 using univariate Cox scores33 (Figure 1D; supplemental Figure 2). As expected, due to the higher levels in U-CLL, the magnitude of chromatin activation in regions from S1 was linked to worse clinical behavior (66/130 regions; hazard ratio >1; Figure 1E). In the case of S2, we unexpectedly observe that, in spite of not being significantly related to the IGHV status (Figure 1C; right), higher H3K27ac signal was associated with a more indolent clinical behavior (139/304 regions, hazard ratio <1; Figure 1F). Interestingly, compared with S1, regions of the S2 signature were enriched in enhancer elements distant from promoters (P < .001; supplemental Figure 3).
These initial analyses suggest a dual role for de novo chromatin activation in CLL. In addition to the expected link between increased chromatin alterations and worse clinical behavior, we identify that indolent clinical behavior also seems to be an active process linked to de novo programming of the chromatin landscape.
The balance between PD and ID chromatin signatures improves the estimation of prognosis
The 2 chromatin signatures associated with shorter or longer TTFT exhibit a gradient of chromatin activation levels (Figure 2A-B). Consequently, we generated 2 scores that summarize the clinically relevant chromatin activation level in each sample and signature: SPD for PD (n = 66 regions; Figure 2A; supplemental Table 3) and SID for ID (n = 139 regions; Figure 2B; supplemental Table 3). The SPD score was clearly increased in U-CLL compared with M-CLL (P < .0001; Figure 2A). Although the initial S2 signature was unrelated to the IGHV status, the refined SID, selecting clinically relevant regions, showed a significant increase in M-CLL, consistent with its better clinical behavior (P = .008; Figure 2B). However, the SPD score showed a much stronger association with the IGHV status, as determined by the magnitude of the Cohen d statistic between U-CLL and M-CLL (1.64 for SPD and –0.56 for SID). In the context of the 3 CLL epigenetic subtypes, the magnitude of the PD and ID scores was overall aligned with their reported clinical behavior (supplemental Figure 4). Interestingly, multivariate Cox analyses demonstrated that each score, taken as a continuous variable, independently predicted TTFT in opposite directions (Figure 2C).
The clinical impact of the balance between PD and ID signatures. (A-B) A heat map (left) depicting the H3K27ac PD (A) and ID (B) signatures. Signal intensities are presented as row z scores. The SPD and SID signature enrichment scores for each sample are indicated below. A box plot (right) compares the SPD or SID values between U-CLL and M-CLL. Statistical significance was assessed using 2-tailed Student t tests. (C) HRs for the multivariate Cox regression model including SPD and SID as predictors of TTFT. Both scores were standardized as z scores for easier comparison and interpretation. (D) Definition of the balance score SB. To illustrate the concept, this is represented as a quotient, despite the actual formula being SPD – SID, effectively representing the PD-to-ID ratio, because these scores are determined from log-transformed data.29 (E) Permutation test evaluating the χ2 test statistic of the balance score when rearranging the labels in the clinical data. The histogram of the null distribution and a red vertical line indicating the observed statistic with the source data are plotted. (F) Comparison of Cox regression models using TTFT as the end point variable. The χ2 test statistic and P value from the likelihood ratio test (LRT) are presented for each comparison. ∗∗∗∗P < .0001; ∗∗P < .01. CI, confidence intervals.
The clinical impact of the balance between PD and ID signatures. (A-B) A heat map (left) depicting the H3K27ac PD (A) and ID (B) signatures. Signal intensities are presented as row z scores. The SPD and SID signature enrichment scores for each sample are indicated below. A box plot (right) compares the SPD or SID values between U-CLL and M-CLL. Statistical significance was assessed using 2-tailed Student t tests. (C) HRs for the multivariate Cox regression model including SPD and SID as predictors of TTFT. Both scores were standardized as z scores for easier comparison and interpretation. (D) Definition of the balance score SB. To illustrate the concept, this is represented as a quotient, despite the actual formula being SPD – SID, effectively representing the PD-to-ID ratio, because these scores are determined from log-transformed data.29 (E) Permutation test evaluating the χ2 test statistic of the balance score when rearranging the labels in the clinical data. The histogram of the null distribution and a red vertical line indicating the observed statistic with the source data are plotted. (F) Comparison of Cox regression models using TTFT as the end point variable. The χ2 test statistic and P value from the likelihood ratio test (LRT) are presented for each comparison. ∗∗∗∗P < .0001; ∗∗P < .01. CI, confidence intervals.
We next postulated that the balance between these 2 forces34 could be a better determinant of overall CLL behavior. Thus, we defined the balance score (SB) as the SPD-to-SID ratio (ie, SPD minus SID in log-transformed values; Figure 2D). A permutation test revealed a strong association between the balance score and TTFT (P < .0001; Figure 2E). Notably, as a quantitative variable, the balance score outperformed the clinical impact of the 2 individual scores and seemed to encapsulate all prognostic information provided by each of them (Figure 2F). Moreover, a permutation test for TTFT demonstrated the independent prognostic value of the balance score compared with IGHV (P < .001).
Identification of gene and protein expression signatures as surrogates of de novo chromatin activation in CLL
Considering the well-established correlation between H3K27ac and gene expression,35 we next selected 66 CLL samples from the H3K27ac ChIP-seq ES series with available RNA-seq data to identify a surrogate expression signature of chromatin activation levels. Of the potential target genes of the de novo active regions,18 we selected those with strong positive correlation (false discovery rate [FDR] 10–5; Figure 3A). This analysis led to the identification of 30 and 38 genes associated with PD and ID, respectively (Figure 3B-C; supplemental Tables 4-5). Interestingly, these PD and ID signatures contain several genes (ie, FMOD, KSR2, CLNK, FILIP1L, and CTLA4) whose messenger RNA (mRNA) seems to be overexpressed in CLL compared with other lymphoproliferative disorders.36
Surrogate PD and ID gene expression signatures. (A) A schematic representation of the methodology used to define the surrogate gene expression signatures. (B-C) A heat map (left) depicting the gene expression of PD (B) and ID (C) signatures. Gene expression levels are presented as row z scores. The SPD and SID signature enrichment scores for each sample are indicated below. Genes listed from top to bottom and left to right correspond to the rows from the heat map from top to bottom. A box plot (right) compares the SPD or SID values between U-CLL and M-CLL. Statistical significance was assessed using 2-tailed Student t tests. (D) HRs and P values for the multivariate Cox regression model, including SPD and SID as predictors of TTFT. Independent cases from the H3K27ac series were used for this analysis. Both scores were standardized as z scores for easier comparison and interpretation. P values were calculated using the Wald test. (E) Box plots depicting the PD and ID scores in mature healthy B cells and CLL in 2 independent cohorts. Statistical significance was determined using 2-tailed Student t tests. (F) A scatterplot showing the correlation between the balance scores obtained from H3K27ac ChIP-seq and RNA-seq. Pearson correlation coefficient was calculated, and the P value was determined using a 2-tailed test, assuming the coefficient to follow a t distribution with 64 degrees of freedom. (G) HR and P value for the univariate Cox regression model, including SB as a predictor of TTFT. Independent cases from the H3K27ac series were used for this analysis. The balance score was standardized as a z score for easier comparison and interpretation. The P value was calculated using the Wald test. (H) Cox regression model assessing Binet A stage CLL cases, using balance score and IGHV mutational status as predictors and TTFT as the end point variable. Independent cases from the H3K27ac series were analyzed, with P values calculated via the Wald test. (I) Box plots comparing the balance score between CLL groups in the external UK cohort.28 Statistical significance was determined using the Tukey's Honest Significant Difference post hoc test. ∗∗∗∗P < .0001; ∗∗∗P < .001; ∗∗P < .01; ∗P < .05; ns, not significant.
Surrogate PD and ID gene expression signatures. (A) A schematic representation of the methodology used to define the surrogate gene expression signatures. (B-C) A heat map (left) depicting the gene expression of PD (B) and ID (C) signatures. Gene expression levels are presented as row z scores. The SPD and SID signature enrichment scores for each sample are indicated below. Genes listed from top to bottom and left to right correspond to the rows from the heat map from top to bottom. A box plot (right) compares the SPD or SID values between U-CLL and M-CLL. Statistical significance was assessed using 2-tailed Student t tests. (D) HRs and P values for the multivariate Cox regression model, including SPD and SID as predictors of TTFT. Independent cases from the H3K27ac series were used for this analysis. Both scores were standardized as z scores for easier comparison and interpretation. P values were calculated using the Wald test. (E) Box plots depicting the PD and ID scores in mature healthy B cells and CLL in 2 independent cohorts. Statistical significance was determined using 2-tailed Student t tests. (F) A scatterplot showing the correlation between the balance scores obtained from H3K27ac ChIP-seq and RNA-seq. Pearson correlation coefficient was calculated, and the P value was determined using a 2-tailed test, assuming the coefficient to follow a t distribution with 64 degrees of freedom. (G) HR and P value for the univariate Cox regression model, including SB as a predictor of TTFT. Independent cases from the H3K27ac series were used for this analysis. The balance score was standardized as a z score for easier comparison and interpretation. The P value was calculated using the Wald test. (H) Cox regression model assessing Binet A stage CLL cases, using balance score and IGHV mutational status as predictors and TTFT as the end point variable. Independent cases from the H3K27ac series were analyzed, with P values calculated via the Wald test. (I) Box plots comparing the balance score between CLL groups in the external UK cohort.28 Statistical significance was determined using the Tukey's Honest Significant Difference post hoc test. ∗∗∗∗P < .0001; ∗∗∗P < .001; ∗∗P < .01; ∗P < .05; ns, not significant.
Subsequently, we generated 2 expression scores for each sample, that is SPD and SID (Figure 3B-C), which were highly correlated with the analog chromatin scores (r = 0.87 for PD; r = 0.90 for ID; both P < .0001; supplemental Figure 5A-B). As expected, we observed higher levels of SPD in U-CLL (P < .0001; Figure 3B) and higher levels of SID in M-CLL (P < .001; Figure 3C), with SPD showing a more pronounced difference between the 2 IGHV subtypes than SID (Figure 3B-C; Cohen d = 1.82 for SPD; Cohen d = –0.56 for SID). Classifying cases according to 3 CLL epigenetic subtypes also led to the expected magnitudes of the scores (supplemental Figure 5C-D). Multivariate Cox analysis on independent cases of the ES cohort (n = 110) confirmed the independent prognostic value of the SPD and SID as continuous variables (Figure 3D). Stratification by maxstat (maximally selected rank statistics) further revealed a risk gradient by score combinations (supplemental Figure 6); comparisons of Cox regression models with and without scores interaction (likelihood ratio tests; P = .58) underscored their independence as prognostic factors of TTFT. Furthermore, using RNA-seq data from 2 independent cohorts,18,28 we observed that healthy mature B cells displayed lower SPD and SID than CLL, consistent with their lack of active regulatory chromatin enhancing target gene expression (P < .001; Figure 3E).
We next calculated the balance score SB as the SPD-to-SID ratio (calculated by subtraction in log-transformed data), which was strongly correlated with the original chromatin-based SB (r = 0.86; P < .0001; Figure 3F). Importantly, the clinical impact of the SB was confirmed on a set of independent cases (Figure 3G). Similar to the chromatin-based score, the SB displays, as a continuous variable, a stronger association with TTFT than SPD or SID individually (supplemental Table 6). These results were validated in a multivariate model with IGHV status, restricting the analysis to cases in Binet A (Figure 3H). Interestingly, we observed that spontaneously regressing CLLs described by Kwok et al 28 had the lowest SB, followed by indolent M-CLL, progressive M-CLL, and U-CLLs (Figure 3I).
Finally, as an additional layer of orthogonal validation, we mined proteome data of an independent series of 45 CLLs, of which 39 cases were also profiled by RNA-seq.25 We identified 11 and 20 proteins whose expression levels positively correlated (1-tailed test; P < .05) with those of the associated coding genes of the PD and ID signatures, respectively (Figure 4A-B). The subsequent SPD, SID, and final SB scores showed the same clinical associations as those observed for chromatin and gene expression data (Figure 4A-D).
Surrogate PD and ID protein expression signatures. (A-B) A heat map (left) depicting the protein abundance PD signature (A) and ID signature (B). Protein abundance levels are presented as row z scores. The SPD and SID signature enrichment scores for each sample are indicated below. Proteins listed from top to bottom and left to right correspond to the rows from the heat map from top to bottom. A box plot (right) comparing the SPD or SID values between U-CLL and M-CLL. Statistical significance was assessed using 2-tailed Student t tests. (C) HRs and P values for the multivariate Cox regression model, including SPD and SID as predictors of TTFT. Both scores were standardized as z scores for easier comparison and interpretation. P values were calculated using the Wald test. (D) Comparison of Cox regression models using TTFT as the end point variable. The χ2 test statistic and P value from the LRT are presented for each comparison. ∗∗P < .01; ns, not significant.
Surrogate PD and ID protein expression signatures. (A-B) A heat map (left) depicting the protein abundance PD signature (A) and ID signature (B). Protein abundance levels are presented as row z scores. The SPD and SID signature enrichment scores for each sample are indicated below. Proteins listed from top to bottom and left to right correspond to the rows from the heat map from top to bottom. A box plot (right) comparing the SPD or SID values between U-CLL and M-CLL. Statistical significance was assessed using 2-tailed Student t tests. (C) HRs and P values for the multivariate Cox regression model, including SPD and SID as predictors of TTFT. Both scores were standardized as z scores for easier comparison and interpretation. P values were calculated using the Wald test. (D) Comparison of Cox regression models using TTFT as the end point variable. The χ2 test statistic and P value from the LRT are presented for each comparison. ∗∗P < .01; ns, not significant.
Genes and pathways associated with the ID and PD signatures
To gain insight into the potential mechanisms underlying the antagonistic prognostic impact of the ID and PD scores, we initially explored the functions of individual genes within the signatures. We identified that 7 intragenic H3K27ac peaks of the ID signature were associated with ATXN1 expression levels (Figure 5A; supplemental Table 5). This gene has been reported to inhibit NOTCH signaling,37 a known oncogene in CLL.38,39 Therefore, we further analyzed the association between ATXN1 and NOTCH1 signaling in lymph node (LNs) samples of CLL, because it has been reported that NOTCH1 signaling is operative in the LN niche and becomes downregulated in blood.38,40-43 We observed a negative correlation of the mRNA abundance between ATXN1 and most of the canonical NOTCH1 signaling target genes in 2 series of CLLs sampled from unpurified and purified LNs (Figure 5B; supplemental Figure 7A). Moreover, comparing the expression of cases with ID high (ATXN1 high) vs ID low (ATXN1 low) in CLL cells from peripheral blood (PB), we observed a strong enrichment in NOTCH signaling (supplemental Figure 7B-C). Another region of interest of the ID signature comprised 9 H3K27ac peaks and 3 genes in 1q32. These genes included PRELP, a gene uniquely expressed in CLL, which reportedly binds and inhibits NF-κB activity44; ATP2B4 (PMCA4), which plays a role in B-cell receptor (BCR)-induced calcium efflux45; and FMOD, whose specific role in CLL remains elusive, but it has been recognized as a CLL-specific gene36,46 (supplemental Figure 8A; supplemental Tables 4-5). Additionally, another gene present in the region, LAX1, still exhibited a positive association (although weaker than the other genes) with H3K27ac peak activation (FDR, 0.01). Noticeably, LAX1 has been identified as a negative regulator in lymphocyte signaling in B cells,47 and its protein has been found to be upregulated in CLL.48
Genes and pathways associated with the activation of ID and PD signatures. (A) Epigenomic landscape of ATXN1 gene in CLL and mature B cells. The genes and genomic regions (upper) belonging to the PD or ID H3K27ac signatures and the chromatin states in 7 biologically independent CLLs, along with representative samples of normal B-cell subpopulations. No genes or peaks from the PD signatures are found within the displayed area. The median RNA-seq levels (lower) of the 7 biologically independent CLLs and 15 biologically independent normal B cells. (B) Pearson correlation coefficient between ATXN1 and NOTCH1 signaling target genes VST mRNA abundance in 5 CLL samples from the lymph nodes. P values were determined using 1-tailed tests, assuming the coefficient to follow a t distribution with 3 degrees of freedom. FDR was estimated with the Benjamini-Hochberg procedure. (C) GSEA of ES and DE1 cohorts analyzing each IGHV subtype independently. Hallmark gene sets with a normalized enrichment score (NES) absolute value >1 and adjusted P value <.1 in at least 2 analyses are depicted. GSEA results are annotated with the significance level of the P value resulting from permutation tests adjusted for multiple testing with the Benjamini-Hochberg procedure. The gene sets are ordered according to the accumulated NES across the 4 analyses, shown in the right bar. (D) Analysis of transcription factor (TF) activities in ES and DE1 cohorts, studied independently for each IGHV subtype. TFs presenting a normalized weighted mean (NWMEAN) absolute value >1.5 and P value <.01 in at least 2 analyses are displayed. The results are annotated with the significance level of the P value resulting from permutation tests. TFs are ordered according to the accumulated NWMEAN across the 4 analyses, illustrated in the right bar. (E) A panel (left) showing the association of genetic driver alterations with the balance score analyzed in the whole cohort, U-CLL and M-CLL. Point estimates with 95% confidence intervals are calculated using 2-sided t tests and controlling for FDR using the Benjamini-Hochberg method. The point estimates represent the difference between the mean balance score in individuals with CLL with and without each corresponding alteration. They are color-coded based on FDR. Genetic alterations associated with the balance score with a P value <.05 in any of the analyses are displayed. Oncoprint representation (right) of the selected genetic driver alterations. For each sample is annotated the number of genetic driver alterations, the IGHV mutational status, TTFT or right censoring time, an indicator of patient treatment after sampling, and the balance score. Samples are ordered from lower to higher balance score. The number of samples with mutations, as well as the percentage of mutated samples over the whole cohort, is shown on the right. Genetic alterations are color-coded according to their type. +FDR, 0.1; ∗∗∗∗P < .0001; ∗∗∗P < .001; ∗∗P < .01; ∗P < .05; · P < .1. Acc., accumulated; ActProm, active promoter; GCBC, germinal center B cell; H3K27me3_Repr, H3K27me3 repressed; H3K9me3_Repr, H3K9me3 repressed; Het;LowSign, Heterochromatin;Low Signal; MBC, memory B cell; NBC-PB, naïve B cell from PB; NBC-T, naïve B cell from tonsil; PCT, plasma cell from tonsil; PoisProm, poised promoter; sign., signaling; StrEnh1, Strong Enhancer 1; StrEnh2, Strong Enhancer 2; RCT, right censoring time; Txn_Elong, transcription elongation; Txn_Trans, transcription transition; Wk_Txn, weak transcription; WkEnh, weak enhancer; WkProm, weak promoter.
Genes and pathways associated with the activation of ID and PD signatures. (A) Epigenomic landscape of ATXN1 gene in CLL and mature B cells. The genes and genomic regions (upper) belonging to the PD or ID H3K27ac signatures and the chromatin states in 7 biologically independent CLLs, along with representative samples of normal B-cell subpopulations. No genes or peaks from the PD signatures are found within the displayed area. The median RNA-seq levels (lower) of the 7 biologically independent CLLs and 15 biologically independent normal B cells. (B) Pearson correlation coefficient between ATXN1 and NOTCH1 signaling target genes VST mRNA abundance in 5 CLL samples from the lymph nodes. P values were determined using 1-tailed tests, assuming the coefficient to follow a t distribution with 3 degrees of freedom. FDR was estimated with the Benjamini-Hochberg procedure. (C) GSEA of ES and DE1 cohorts analyzing each IGHV subtype independently. Hallmark gene sets with a normalized enrichment score (NES) absolute value >1 and adjusted P value <.1 in at least 2 analyses are depicted. GSEA results are annotated with the significance level of the P value resulting from permutation tests adjusted for multiple testing with the Benjamini-Hochberg procedure. The gene sets are ordered according to the accumulated NES across the 4 analyses, shown in the right bar. (D) Analysis of transcription factor (TF) activities in ES and DE1 cohorts, studied independently for each IGHV subtype. TFs presenting a normalized weighted mean (NWMEAN) absolute value >1.5 and P value <.01 in at least 2 analyses are displayed. The results are annotated with the significance level of the P value resulting from permutation tests. TFs are ordered according to the accumulated NWMEAN across the 4 analyses, illustrated in the right bar. (E) A panel (left) showing the association of genetic driver alterations with the balance score analyzed in the whole cohort, U-CLL and M-CLL. Point estimates with 95% confidence intervals are calculated using 2-sided t tests and controlling for FDR using the Benjamini-Hochberg method. The point estimates represent the difference between the mean balance score in individuals with CLL with and without each corresponding alteration. They are color-coded based on FDR. Genetic alterations associated with the balance score with a P value <.05 in any of the analyses are displayed. Oncoprint representation (right) of the selected genetic driver alterations. For each sample is annotated the number of genetic driver alterations, the IGHV mutational status, TTFT or right censoring time, an indicator of patient treatment after sampling, and the balance score. Samples are ordered from lower to higher balance score. The number of samples with mutations, as well as the percentage of mutated samples over the whole cohort, is shown on the right. Genetic alterations are color-coded according to their type. +FDR, 0.1; ∗∗∗∗P < .0001; ∗∗∗P < .001; ∗∗P < .01; ∗P < .05; · P < .1. Acc., accumulated; ActProm, active promoter; GCBC, germinal center B cell; H3K27me3_Repr, H3K27me3 repressed; H3K9me3_Repr, H3K9me3 repressed; Het;LowSign, Heterochromatin;Low Signal; MBC, memory B cell; NBC-PB, naïve B cell from PB; NBC-T, naïve B cell from tonsil; PCT, plasma cell from tonsil; PoisProm, poised promoter; sign., signaling; StrEnh1, Strong Enhancer 1; StrEnh2, Strong Enhancer 2; RCT, right censoring time; Txn_Elong, transcription elongation; Txn_Trans, transcription transition; Wk_Txn, weak transcription; WkEnh, weak enhancer; WkProm, weak promoter.
Within the PD signature, we highlight a H3K27ac peak in 1p36 associated with the expression of TNFRSF1B, a tumor necrosis factor alpha (TNF-α) receptor49 (supplemental Figure 8B; supplemental Tables 4-5); and 2 peaks in 1p22 targeting TGFBR3, a TGF-β receptor previously linked to CLL50-52 (supplemental Figure 8C; supplemental Tables 4-5). Notably, TGFBR3 expression exhibits the highest correlation with the PD score and shows prognostic value for TTFT independently of IGHV mutational status (supplemental Figure 8D-E).
Next, beyond the individual ID and PD signatures, we investigated pathways and transcription factors (TFs) correlated with the magnitude of the balance score in U-CLL and M-CLL individually (supplemental Figure 9). To increase robustness, we explored 2 independent cohorts10,18 and detected that the balance score was enriched in 2 main pathways (Figure 5C-D). First is a gene set composed of genes regulated by NF-κB in response to TNF-α signaling, which is supported by both the gene set enrichment analysis (GSEA) and the enrichment of NFKB153-55 and RELA54,56 regulons. Second is mTOR signaling, recently linked to a proliferative drive in CLL,10 which could be further supported by enrichment in the ATF6,57,58,HIF1A,59-61,MYC62 and ATF463 regulons.
Finally, we evaluated the relationship between the balance score and genetic drivers in 170 CLLs.6 The results revealed positive associations with 22 drivers (Figure 5E). Although most of these associations were related to IGHV status, we identified specific associations within each CLL subtype. In M-CLL, we observed a strong association between higher balance score and trisomy 12, IGLV3-21R110 mutation, deletion of 11q22.3, and mutations in SF3B1 and MYD88. In U-CLL, mutations in NOTCH1 and ATM were associated with a higher balance score. Notably, trisomy 12 and genetic alterations in ATM have been previously implicated in TNF-α signaling via NF-κB and/or mTOR signaling.64,65 Additionally, we studied in detail the magnitude of the PD, ID, and balance scores in cases with the IGLV3-21R110 mutation, which is related to autonomous BCR signaling and aggressive clinical behavior in M-CLL with moderate IGHV mutation levels and mostly overlaps with the intermediate CLL epigenetic subtype.12,24,66 M-CLLs and intermediate CLLs with the IGLV3-21R110 mutation showed higher PD, lower ID, and higher balance score than cases lacking this mutation (supplemental Figure 10A-B). Intermediate CLLs with IGLV3-21R110 mutation showed similar scores to naïve-like CLLs, whereas intermediate cases without the mutation were similar to memory-like CLLs, which is expected based on their clinical behavior24 (supplemental Figure 10C-E).
The magnitude of the PD score is modulated within the LN microenvironment
CLL cells recirculate between PB and lymphoid tissues, with LNs being a niche where CLL cells are stimulated to proliferate.39,42,67-70 The LN imprint can be detected in circulating CLL cells, in which 2 major subpopulations are found: the "proliferative" fraction, comprising lymphoid tissue emigrants enriched in recently divided cells, and the "resting" fraction, containing CLL cells primed to home back to the LN.69,71 Given this context, we explored whether our signatures are modulated in LNs using 4 cohorts; that is, paired LN and PB samples,26 PB samples with sorted proliferative and resting fractions,27 and 2 single-cell RNA-seq experiments22,23 (supplemental Table 7), in which CXCR4 and CD2771 levels were used to identify proliferative and resting fractions (refer to supplemental Methods; supplemental Figure 11A-B).
We observed that the PD score significantly increased both in the LN, compared with PB, and in the proliferative fraction, compared with the resting fraction, using bulk and single-cell data (P ≤ .002; Figure 6A-C). In contrast, the ID remained relatively constant (P ≥ .15), with only a few cases showing changes in either direction between LN/proliferative fraction and PB/resting fraction. Consequently, the balance score tended to be higher in the LN or proliferative fraction (P ≤ .056), reflecting the strong increase in the PD score.
Enrichment of the PD, ID, and balance scores during CLL trafficking. (A-C) PD, ID, and B score comparisons between paired samples extracted from PB and the LN (A); between paired samples of the sorted proliferative and resting CLL fractions (B); and between CLL proliferative and resting fractions identified at the single-cell level (C). P values were obtained using paired 2-tailed Student t tests; (D) HRs and P values for univariate Cox regression models using the different scores as predictors of TTFT in the proliferative and resting CLL fractions. The scores were standardized as a z score for easier comparison and interpretation. P values were calculated using the Wald test. Prolif., proliferative.
Enrichment of the PD, ID, and balance scores during CLL trafficking. (A-C) PD, ID, and B score comparisons between paired samples extracted from PB and the LN (A); between paired samples of the sorted proliferative and resting CLL fractions (B); and between CLL proliferative and resting fractions identified at the single-cell level (C). P values were obtained using paired 2-tailed Student t tests; (D) HRs and P values for univariate Cox regression models using the different scores as predictors of TTFT in the proliferative and resting CLL fractions. The scores were standardized as a z score for easier comparison and interpretation. P values were calculated using the Wald test. Prolif., proliferative.
Beyond the comparison between proliferative and resting conditions, the data shown in Figure 6 also point to different degrees of PD, ID, and balance scores in different cases. Therefore, we next explored the association of the PD, ID, and balance scores with TTFT in proliferative and resting fractions from 21 cases27 (Figure 6D). The balance score evidenced a stronger association with disease progression in the proliferative fraction (P = .012), while showing a trend in the resting fraction (P = .065). In line with our previous observations (Figures 2E and 3G), the balance score presented a stronger association with TTFT than the PD and ID scores in each cellular fraction.
We next studied the relationship between the balance score and the proliferative potential of CLL cells. We mined CLL samples treated with cytosine-phosphorothioate-guanine oligodeoxynucleotides (CpG-ODN),10 a class of Toll-like receptor 972 agonists known to induce a proliferative response in CLL cells and whose magnitude has been suggested to be linked to clinical outcomes.73 Our analysis revealed the expected positive and inverse correlation between the PD and ID scores and the percentage of Ki-67+ cells, respectively (supplemental Figure 12). Notably, the balance score exhibited a more significant association (ρ = 0.88; P < .0001) than the individual scores.
The balance score as a powerful independent prognostic factor in CLL
We conducted comprehensive multivariate analyses encompassing 439 CLL cases across independent cohorts and data modalities, including gene expression (n = 399 across 3 cohorts) and protein expression (n = 40). Whenever available and significant at the univariate level, we evaluated the balance score alongside other clinically relevant variables such as IGHV mutational status, TP53 alterations, clinical stage, lymphocyte count, lymphadenopathy, age, and sex (supplemental Figure 13).
Across all studied cohorts, the balance score maintained a strong prognostic value when analyzed alongside key clinical variables. Specifically, in validation cases from the ES cohort profiled with gene expression arrays, patient stratification by balance score quartiles was highly significant (P < .0001; Figure 7A). In line with this finding, the balance score as a quantitative variable showed prognostic power when adjusted for Binet stage, IGHV status, age, and sex (n = 165; Figure 7B). These data were further validated using RNA-seq in the same ES cohort (n = 81; Figure 7C). In this ES cohort, the balance score was also independent of lymphocyte count and lymphadenopathy (supplemental Figure 14). In the DE1 cohort profiled with RNA-seq, the score was an independent prognostic marker, alongside IGHV status and TP53 genetic alterations (n = 140; Figure 7D). These results were further replicated in the US3 cohort (RNA-seq, n = 91; Figure 7E) and the DE2 cohort (proteomics, n = 40; Figure 7F). Finally, we integrated the balance score from the previous individual cohorts and technologies into a single Cox regression model, confirming its role as an IGHV-independent prognostic marker (supplemental Figure 15).
Clinical impact of the balance score in CLL. (A-B) Survival analyses in the ES cohort using validation cases profiled with expression arrays: Kaplan-Meier curves for TTFT, stratifying patients into 4 groups based on balance score quartiles, with P value from the log-rank test (A); multivariate Cox regression incorporating the balance score, Binet stage, IGHV mutational status, age, and sex as predictors, with TTFT as the end point (B). (C) Multivariate Cox regression on ES cohort cases profiled with RNA-seq (78 overlapping with expression arrays), using the balance score, Binet stage, and IGHV mutational status as predictors of TTFT. (D) Analysis of the DE1 cohort profiled with RNA-seq, using multivariate Cox regression with balance score, IGHV status, and TP53 alterations as predictors of TTFT. (E) Multivariate Cox regression on the US3 cohort profiled with RNA-seq, with balance score and IGHV status as predictors of TTFT. (F) Multivariate Cox regression on the DE2 cohort profiled for protein abundance, using balance score and IGHV mutational status as TTFT predictors. P values in panels B-F were calculated with the Wald test. The balance score was standardized as a z score for consistent comparison and interpretation across all analyses.
Clinical impact of the balance score in CLL. (A-B) Survival analyses in the ES cohort using validation cases profiled with expression arrays: Kaplan-Meier curves for TTFT, stratifying patients into 4 groups based on balance score quartiles, with P value from the log-rank test (A); multivariate Cox regression incorporating the balance score, Binet stage, IGHV mutational status, age, and sex as predictors, with TTFT as the end point (B). (C) Multivariate Cox regression on ES cohort cases profiled with RNA-seq (78 overlapping with expression arrays), using the balance score, Binet stage, and IGHV mutational status as predictors of TTFT. (D) Analysis of the DE1 cohort profiled with RNA-seq, using multivariate Cox regression with balance score, IGHV status, and TP53 alterations as predictors of TTFT. (E) Multivariate Cox regression on the US3 cohort profiled with RNA-seq, with balance score and IGHV status as predictors of TTFT. (F) Multivariate Cox regression on the DE2 cohort profiled for protein abundance, using balance score and IGHV mutational status as TTFT predictors. P values in panels B-F were calculated with the Wald test. The balance score was standardized as a z score for consistent comparison and interpretation across all analyses.
Notably, although IGHV status is a well-established prognostic factor in CLL,4,5,74 the balance score consistently outperformed it. We further explored this unexpected finding by comparing univariate Cox regression models incorporating the IGHV mutational status or the balance score and a multivariate Cox regression model including both variables in all the cohorts (supplemental Table 8). Consistent with our previous findings, these comparisons confirmed that including the balance score, but not the IGHV status, improved the model fit. Additionally, epigenetic subtypes also reduce their prognostic power when analyzed alongside the balance score (supplemental Figure 16).
Discussion
This study provides evidence suggesting that the biological behavior of CLL cells and, consequently, the natural progression of the disease seem to be associated with the balance between 2 antagonistic molecular forces mediated by the same epigenetic mechanism of de novo chromatin activation. A similar concept was proposed in 2014 by Packham et al34 from the University of Southampton, but it centered on the balance between proliferation or anergy triggered by the BCR. The target genes involved in our chromatin signatures not only provide links to the BCR but also to other functions. Genes of the ID score such as ATP2B4 and LAX1 may negatively modulate BCR signaling, but other genes such as ATXN1 and PRELP seem to inhibit NOTCH and NF-κB activity, respectively,44,45,47,75 which altogether seem to induce cellular quiescence through downregulating the response to proliferative stimuli. The PD score contains genes such as TNFRSF1B, which is a receptor involved in TNF-α signaling, a pathway that can be linked with a more proliferative phenotype in CLL.76 Most remarkably, the magnitude of the balance between PD and ID seems to be related to transcriptional states, leading to higher proliferation, as demonstrated by the correlation with the percentage of Ki67+ cells and specific pathways such as TNF-α via NF-κB and mTOR signaling. Previous studies associated higher levels of TNF-α with a poorer prognosis,77 and TNF-α via NF-κB was identified as a driver of CLL.76 Additionally, mTOR signaling has been linked to a proliferative drive in CLL.10 Interestingly, CLLs with trisomy 12, which have a higher balance score, show a specific enrichment of these 2 pathways.65,78 These findings suggest TNF-α via NF-κB and mTOR signaling as potential therapeutical targets in CLL.
Our examination of the enrichment of the signatures in relation to CLL trafficking suggests that the PD can be extrinsically modulated in the LN niche, and such imprint gradually decreases in circulating CLL cells. In contrast, the ID is not consistently affected by CLL trafficking and seems to be independent of microenvironmental stimuli.
From the translational perspective, we validated the independent prognostic impact of the balance score using various modalities (chromatin, RNA-seq, microarrays, and proteomics) across several independent cohorts. We observed that the balance score surpasses the prognostic power of the IGHV mutational status, which seems to lose any independent prognostic value in multivariate models. Considering that the balance score is constructed as the ratio of the PD, which is strongly influenced by the IGHV status, and the ID, which is only slightly related to IGHV mutational status, it contains both IGHV-related and independent information.
From the translational perspective, we are aware that using ChIP-seq or RNA-seq–based assays may still be impractical. Therefore, we envision that the balance score could be implemented in the clinical setting through the development of dedicated gene expression assays based on standardized technologies, such as nanoString nCounter.79 Furthermore, additional studies shall also evaluate the potential of the SB as a predictive biomarker of response and overall survival in the context of current treatments.
Overall, we believe that this study provides compelling evidence that the balance score reflects CLL proliferative capacity through the integration of multiple elements involved in CLL pathogenesis. Due to its strong independent prognostic value, the incorporation of the balance score into current clinical models could serve as a new valuable asset in the clinical management of CLL.
Acknowledgments
This research was primarily funded by an accelerator award Cancer Research UK/Associazione Italiana per la Ricerca sul Cancro (AIRC)/Asociación Española contra el Cáncer (AECC) joint funder-partnership (J.I.M.-S.; grant C355/A26819), the Fundación Científica AECC (FC AECC) (J.I.M.-S.; grant PRYGN234766MART), and the European Research Council under the European Union’s Horizon 2020 research and innovation programme (J.I.M.-S. and E.C.; grant 810287). Additionally, the authors obtained funding for this project from La Caixa Foundation (CLLEvolution LCF/PR/HR17/52150017 [grant HR17-00221LCF] and CLLSYSTEMS-LCF/PR/HR22/52420015 [grant HR22-00172] Health Research 2017 and 2022 Programs to E.C.), Agència de Gestió d'Ajuts Universitaris i de Recerca (AGAUR) of the Generalitat de Catalunya (grant 2021-SGR-01343 to J.I.M.-S. and grant 2021-SGR-01172 to E.C.), and CIBERONC (group numbers CB24/12/00009, CB16/12/00225 and CB16/12/00334). This work was partially developed at the Centre Esther Koplowitz (Barcelona, Spain).
Authorship
Contribution: V.C. performed H3K27ac ChIP-seq data analysis; V.C. and G.C. conducted statistical analyses; N.R., F.N., and A.V. contributed to sample collection; N.R. and F.N. contributed to bulk transcriptome data generation; V.C., F.N., and R.R. conducted bulk transcriptome data analysis; V.C. and R.M.-B. conducted single-cell RNA-seq data analysis; F.N., T.Z., J.L., H.H., S.D., S.A.H., D.J.B., and J.C.S. contributed with essential data resources; F.N., P.M., M.D.-F., J.A.P., N.V., A.L.-G., J.D., E.C., S.D., and S.A.H. contributed to biological and/or clinical annotation; V.C., J.I.M.-S., A.M.-D., J.M.-J., G.C., F.N., M.D.-F., L.L.-C., S.C., and E.C. participated in data interpretation; V.C. and J.I.M.-S. designed the study and wrote the manuscript; J.I.M.-S. directed the research; and all the authors read, commented on, and approved the manuscript.
Conflict-of-interest disclosure: V.C. and J.I.M.-S. hold the author rights of the prognostic index CLL Balance Score described in this study. The remaining authors declare no competing financial interests.
Correspondence: Jose I. Martin-Subero, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Carrer del Rosselló 149-153, Barcelona 08036, Spain; email: imartins@recerca.clinic.cat.
References
Author notes
Most of the data processed in this study were obtained from previous publications properly referenced in the manuscript. The few newly generated data are available through the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus data repository (accession number GSE283876).
The CLL Balance Score can be used by nonprofit organizations and for noncommercial applications without seeking further permission. For profit organizations and commercial purposes, however, we kindly request that individuals or organizations contact the Knowledge and Technology Transfer Unit of the Institut d'Investigacions Biomèdiques August Pi i Sunyer (innova@recerca.clinic.cat).
The online version of this article contains a data supplement.
There is a Blood Commentary on this article in this issue.
The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.
This feature is available to Subscribers Only
Sign In or Create an Account Close Modal