Next Article in Journal
The Role of Genomic Scores in the Management of Prostate Cancer Patients: A Comprehensive Narrative Review
Previous Article in Journal
Animal Venoms as Potential Antitumor Agents Against Leukemia and Lymphoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Blood-Epigenetic Biomarker Associations with Tumor Immunophenotype in Patients with Urothelial Carcinoma from JAVELIN Bladder 100

by
Thomas Powles
1,*,
Srikala S. Sridhar
2,
Joaquim Bellmunt
3,
Cora N. Sternberg
4,
Petros Grivas
5,
Ewan Hunter
6,
Matthew Salter
6,
Ryan Powell
6,
Ann Dring
6,
Jayne Green
6,
Alexandre Akoulitchev
6,
Roy Ronen
7,
Janusz Dutkowski
7,
Robert Amezquita
8,
Chao-Hui Huang
8,
Diane Fernandez
8,
Robbin Nameki
8,
Keith A. Ching
8,
Jie Pu
9,
Michelle Saul
9,
Shibing Deng
8,
Alessandra di Pietro
10 and
Craig B. Davis
8,*
add Show full author list remove Hide full author list
1
Barts Cancer Institute, Experimental Cancer Medicine Centre, Queen Mary University of London, St. Bartholomew’s Hospital, Charterhouse Square, London EC1M 6BQ, UK
2
Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada
3
Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
4
Englander Institute for Precision Medicine, Division of Hematology and Medical Oncology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
5
Fred Hutchinson Cancer Center, University of Washington, Seattle, WA 98109, USA
6
Oxford BioDynamics Plc, Oxford OX4 2WB, UK
7
Data4Cure, Inc., Waltham, MA 02451, USA
8
Pfizer, San Diego, CA 92121, USA
9
Pfizer, Tucson, AZ 85714, USA
10
Pfizer srl, 20152 Milano, Italy
*
Authors to whom correspondence should be addressed.
Cancers 2025, 17(14), 2332; https://doi.org/10.3390/cancers17142332
Submission received: 10 April 2025 / Revised: 20 June 2025 / Accepted: 25 June 2025 / Published: 14 July 2025
(This article belongs to the Section Cancer Biomarkers)

Simple Summary

The JAVELIN Bladder 100 trial investigated avelumab maintenance treatment in people with advanced bladder cancer. In this exploratory analysis of data from the JAVELIN Bladder 100 trial, researchers investigated the association between chromatin conformation markers in peripheral blood specimens and tumor gene expression. Researchers identified a distinct set of chromatin conformation markers that were associated with high vs. low immune gene expression. The loci covered by these markers contained genes participating in multiple pathways that may have direct and indirect effects on immune responses. These findings support the use of blood chromatin conformation assays to query biological states in tissues.

Abstract

Background/Objectives: Response to immune checkpoint inhibitors (ICIs) is associated with several biological pathways, including tumor immunogenicity and antitumor immunity. Identifying host factors involved in these pathways may guide personalized ICI treatment. Methods: We describe the application of chromatin conformation assays to blood from patients with advanced urothelial carcinoma from the phase 3 JAVELIN Bladder 100 trial (NCT02603432). This trial demonstrated a significant survival benefit with avelumab maintenance plus best supportive care (BSC) vs. BSC alone following non-progression with platinum-based chemotherapy as first-line therapy. Blood-based chromatin conformation markers (CCMs) were screened for associations with high/low immune effector gene expression in tumors and for interactions with outcomes and tumor mutation burden. Results: Candidate CCMs included genes involved in several immune response pathways, such as POU2F2, which encodes a transcription factor that regulates B-cell maturation. Conclusions: Our findings suggest that polygenic host factors may affect response to ICIs and support further investigation of chromatin conformation assays.

1. Introduction

Exploratory analyses of biomarkers from clinical trials of immune checkpoint inhibitors (ICIs) have highlighted diverse biological pathways associated with response to ICI treatment; however, identifying biomarker assays that provide optimal predictive response value remains a major challenge [1,2]. In the phase 3 JAVELIN Bladder 100 trial, avelumab first-line (1L) maintenance plus best supportive care (BSC) significantly prolonged overall survival (OS) and progression-free survival (PFS) vs. BSC alone in patients with advanced urothelial carcinoma (UC) without disease progression after 1L platinum-based chemotherapy [3]. Based on these results, avelumab 1L maintenance was established as a standard of care in patients with advanced UC [4,5].
Exploratory tumor biomarker analyses from JAVELIN Bladder 100 revealed complex relationships with treatment outcomes [2]. Assays for individual biomarkers used in clinical practice, such as programmed death ligand 1 (PD-L1) and tumor mutation burden (TMB), did not optimally identify patients who were significantly more likely to benefit from avelumab 1L maintenance. Further analyses of gene mutation and expression within tumor biopsies identified 3 classes of biological pathways that may interact with avelumab treatment effect: (1) increased tumor immunogenicity, including APOBEC3-associated mutagenesis; (2) increased activity of both innate and adaptive immune pathways; and (3) decreased expression of genes associated with signaling pathways linked to tissue growth, such as Notch, angiogenesis, and TGF-β. These results suggested that optimization of the avelumab treatment effect would require deeper analysis of biomarkers capturing the functional heterogeneity within the tumor microenvironment.
As highlighted in the recently updated cancer immunity cycle [6], non-transformed cell types in the tumor microenvironment, including stromal and myeloid cells, have emerged as important modulators of anti-tumor immunity released by ICIs. These findings, made possible through the application of novel technologies, such as spatial transcriptomics, support hypotheses that response to ICI may be contingent on host factors such as germline single nucleotide polymorphisms [7], microbiomes [8], and epigenetic modification of the 3-dimensional (3D) structure of chromatin [9,10,11,12,13]. EpiSwitch is an established 3D genomic biomarker platform, based on the original chromosome conformation capture approach, that has demonstrated promising utility in various disease settings [14,15,16,17,18,19], including response to avelumab observed in a retrospective study [20].
We applied the EpiSwitch platform to peripheral blood of patients enrolled in the JAVELIN Bladder 100 trial to explore the potential interplay between epigenetic host factors and immune activity in the tumor microenvironment. A 3-stage process was used: (1) identification of chromatin conformation markers (CCMs) associated with tumor immune gene expression signature; (2) statistical modeling to rank CCMs association with TMB and OS following avelumab plus BSC vs. BSC alone; and (3) characterization of the expression and function of genes covered by the highest-ranked CCMs.

2. Materials and Methods

2.1. Description of Specimens Collected for Analysis

JAVELIN Bladder 100 (NCT02603432) was a randomized phase 3 trial evaluating avelumab 1L maintenance (administered every 2 weeks) plus BSC vs. BSC alone in patients with advanced UC whose cancer had not progressed after 4–6 cycles of 1 L platinum-based chemotherapy [3]. Patients received treatment until disease progression, unacceptable toxicity, or withdrawal of consent. OS (primary endpoint) and PFS (secondary endpoint) were significantly prolonged with avelumab plus BSC vs. BSC alone. Peripheral blood was collected from patients 4–10 weeks after the date of administration of the last dose of chemotherapy but before treatment with avelumab plus BSC or BSC alone. Blood samples from 496 patients satisfied quality and consent criteria and were subjected to further evaluation, as shown in Supplementary Figure S1A. Characteristics and outcomes in the biomarker analysis set compared with the full analysis set are shown in Supplementary Table S1 and Supplementary Figure S2, respectively. The biomarker analysis set did not differ notably from the full analysis set.

2.2. Identification of Candidate CCMs Using the EpiSwitch Platform

2.2.1. Preparation of 3D Genomic Templates

EpiSwitch 3D libraries, with chromosome conformation analytes converted to sequence-based tags, were prepared from frozen whole-blood samples using EpiSwitch protocols following the manufacturer’s instructions for EpiSwitch Explorer Array kits (Oxford BioDynamics Plc, Oxford, UK). Samples were processed on the Freedom EVO 200 robotic platform (Tecan Group Ltd., Männedorf, Switzerland). Briefly, the whole-blood sample was diluted and fixed with a formaldehyde-containing EpiSwitch buffer. Density cushion centrifugation was used to purify intact nuclei. Following a short detergent-based step to permeabilize the nuclei, restriction enzyme digestion and proximity ligation were used to generate the 3D libraries. Samples were centrifuged to pellet the intact nuclei before purification with an adapted protocol from the QIAamp DNA FFPE Tissue kit (Qiagen, Hilden, Germany), eluting in 1× TE buffer (pH 7.5). Three-dimensional libraries were quantified using the Quant-iT PicoGreen dsDNA Assay kit (Invitrogen, Waltham, MA, USA) and normalized to 5 ng/mL of input DNA prior to interrogation by PCR with Eppendorf Mastercycler Pro S (Eppendorf, Hamburg, Germany).

2.2.2. Array Design

Custom microarrays were designed using the EpiSwitch pattern recognition algorithm, which operates on Bayesian modeling and provides a probabilistic score for a region’s involvement in long-range chromatin interactions. The algorithm was used to annotate the GRCh38 human genome assembly across approximately 1.1 million sites with the potential to form long-range chromosome conformations. The most probable interactions were identified and filtered on probabilistic score and proximity to protein, long non-coding RNA, or microRNA coding sequences. Predicted interactions were limited to EpiSwitch sites that were ≥10 kb and <300 kb apart. Repeat masking and sequence analysis were used to ensure unique CCM sequences for each interaction. The EpiSwitch Explorer Array (Oxford BioDynamics Plc, Oxford, UK, Product Code X-HS-AC-02) containing 60-mer oligonucleotide probes was designed to interrogate potential 3D genomic interactions. In total, 964,631 experimental probes and 2500 control probes were added to a 1 × 1 comparative genomic hybridization microarray slide design. The experimental probes were placed on the design in singlicate, with the controls in groups of 250. The control probes consisted of 6 different EpiSwitch interactions that are generated during the extraction processes and used for monitoring library quality. An additional 4 external inline control probe designs were added to detect a non-human (Arabidopsis thaliana) spike in DNA added during the sample labeling protocol to provide a standard curve and control for labeling. The external spike DNA consists of 400 bp single-stranded DNA fragments from genomic regions of Arabidopsis thaliana. Array-based comparisons were performed as described previously, with the modification of only 1 sample being hybridized to each array slide in the Cy3 channel.

2.2.3. Translation of Array-Based 3D Genomic Markers to PCR Readouts

Stepwise diagnostic biomarker discovery process using EpiSwitch technology and data analysis: The EpiSwitch technology platform (Oxford Biodynamics, Oxford, UK) pairs high-resolution chromosome conformation capture results with regression analysis and a machine learning algorithm to develop disease classifications [21,22,23]. To select epigenetic biomarkers, patient samples for each class were screened for statistically significant differences in conditional and stable profiles of genome architecture. All samples used for the nested PCR biomarker discovery were processed per the manufacturer’s instructions using the proprietary EpiSwitch reagents and standard protocols [24]. A PCR product was detected only if the corresponding CCM was detected. To account for variable marker stability in assay conditions, 3 template concentrations (1×, 2×, and 4×) were evaluated.
Genomic mapping. Mapping was performed using the Bedtools closest function [21] for the 3 closest protein-coding loci: upstream, downstream, and within the long-range chromosome interaction (Gencode v33). All markers were visualized using the EpiSwitch analytical portal.

2.2.4. Identification of Top 25 CCMs Associated with Tumor JAV-Immuno Scores

Analyses for marker prioritization were performed using libraries developed for the R Statistical Language (R version 4.1.2). JAV-Immuno includes the following genes: CCL5, CD2, CD244, CD247, CD3E, CD3G, CD6, CD8B, CD96, CST7, EOMES, GFI1, GPR18, GRAP2, IL7R, ITK, KCNA3, KLRD1, NLRC3, PRF1, PSTPIP1, SH2D1A, SIT1, THEMIS, TRAT1, and XCL2. Candidate markers were identified using EpiSwitch whole-genome arrays comparing 20 patients whose tumors were JAV-Immunohi (greater than the median) and 20 patients whose tumors were JAV-Immunolo (less than the median). The data were normalized, quality controlled, and statistically analyzed using the EpiSwitch array pipeline powered by the LIMMA package. The top 150 EpiSwitch markers were selected based on their corrected p-value (false discovery rate) and directionality/interaction frequency for JAV-Immuno. Feature engineering with the selected EpiSwitch CCMs was performed using the same samples used in the array discovery, with an additional 20 samples each of the JAV-Immunohi and JAV-Immunolo classes. A reduced set of 25 CCMs was generated using the univariate EXACT test (contingency testing), binary weighting, multivariate analyses, permutated glmnet, VSURF, Boruta, and varSelRF. An XGBoost algorithm model was used for further refinement of the CCM list [22]. Shapley additive explanations values, used to characterize the contribution of each feature in a model to the final prediction and final model selection, were used to confirm marker prioritization [23].

2.3. Prioritization of Candidate CCMs Based on Estimated Interactions with TMB and OS

The starting feature set comprised 25 CCMs, each measured at 3 dilution levels for a total of 75 features, and was assessed in 496 patients for its association with OS.

Model Development

In step 1, features were filtered out prior to modeling if <40 subjects were included in either the present or absent categories and <15 subjects in any of the 4 subgroups (avelumab present, avelumab absent, BSC present, BSC absent). This filter removed 15 features.
In step 2, due to high correlation among CCMs measured at multiple dilution levels, further screening was performed to either select 1 dilution level per CCM or remove the CCM from subsequent multivariate modeling. The screening assessed each CCM in association with OS using 3 nested Cox proportional hazards models. The first model assessed the CCM main effect (direct association with OS); the second model included the CCM 2-way interaction with treatment and OS as well as the main effect; the third model incorporated the 3-way interaction among CCM, treatment, and TMB in addition to the main effect and 2-way interaction. For each CCM, this resulted in 3 models for each dilution level. The p-values derived from the 3-way interaction, 2-way interaction, and main effect terms were sequentially evaluated for each CCM. The dilution level with the lowest 3-way interaction p-value of <0.1 was selected. If none were <0.1, the process was repeated for the 2-way interaction and main effect p-values. If none of these p-values were <0.1, then the CCM was eliminated from downstream modeling.
Finally, in step 3, the remaining 16 features were fed into an elastic net Cox proportional hazards model-building procedure to select a final feature set. The elastic net [25], a regularized regression method using a weighted sum of the L1 and L2 penalty terms for the lasso [26] and ridge [27] regression, was used for statistical model building (R package version 4.1.2: glmnet). The hyperparameters α and λ, which control the weight applied to each of the penalty terms and the amount of shrinkage on the coefficients, were tuned using repeated 5-fold cross-validation (CV). For each round of 5-fold CV, a pair of α and λ associated with the optimal model performance (C-index) was determined (Supplementary Figure S3). The same procedure was iterated 1000 times using different random data partitions to account for potential CV error. The most frequently selected hyperparameter pairs were chosen as optimal and applied to the full dataset to obtain the final models. All but 1 of the 16 input features were retained by this process, generating the list of 15 CCMs (Supplementary Table S2).

2.4. Assessment of Potential CCM Gene Expression

Public data on genes labeled using the Human Genome Organization nomenclature have been compiled into a knowledge graph [28] by Data4Cure (www.data4cure.com; accessed on 30 March 2025). Knowledge graph associations between genes and specific biomedical entity domains (e.g., cell types or pathways) summarize evidence from thousands of publicly available datasets and millions of publications linking biomedical entities.

2.5. Visium Data Processing

SpaceRanger 2.0.1 was used to align raw reads to the GrCh38 genome and feature count generation using the Human Transcriptome probe set v2.0. Visium data resolution was enhanced 6-fold by inferring pseudospots using the BayesSpace method [29]. Briefly, using the BayesSpace R package Visium Space Ranger, the outs folder was processed with seed 102, spatialPreprocess(sceobj, platform = “Visium”, n.PCs = 15, n.HVGs = 2000, log.normalize = T), followed by spatialCluster(sceobj, q = 8, d = 15, platform = “Visium”, init.method = “mclust”, model = “t”, gamma = 1, nrep = 10,000, burn.in = 100, save.chain = F), and then spatialEnhance(sceobj, q = 8, platform = “Visium”, d = 15, model = “t”, gamma = 1, jitter_prior = 0.3, jitter_scale = 3.5, nrep = 100,000, burn.in = 100, save.chain = F). Enhanced features and spatial coordinates for the full expression matrix were computed using enhanceFeatures and output for subsequent visualization using the Data4Cure platform [30] (www.data4cure.com; accessed on 30 March 2025).
For the expression signatures of each pseudospot, the Data4Cure platform was used to compute the gene signature score as the mean normalized average expression value across the signature genes. The spot-level signature scores were plotted heat map style using the pseudospot coordinates and overlaid on the approximate corresponding region of the H&E image as a background image. An unmarked, blinded H&E image was annotated by a pathologist for lymphoid aggregates (numbered and circled in green).

3. Results

3.1. Selection of a Tumor Immune Gene Expression Signature to Screen for Candidate CCMs

The JAVELIN Bladder 100 study included 496 patients with tumor transcriptome data whose blood could be analyzed by EpiSwitch (Supplementary Figure S1A). A 26-gene signature (JAV-Immuno), identified in JAVELIN Renal 101 and validated in JAVELIN Bladder 100 [2,31], displayed modulation of the interaction between TMB, OS, and treatment (avelumab plus BSC vs. BSC alone) comparable to that of PD-L1 (Figure 1A). In the avelumab plus BSC arm, a significant OS benefit was associated with JAV-Immuno levels (high vs. low) in patients with low TMB (HR, 0.52 [95% CI, 0.351–0.773]; p = 0.0012) but not in patients with high TMB (HR, 1.22 [95% CI, 0.756–1.957]; p = 0.42). No association was found between either biomarker and OS in the BSC-alone arm. JAV-Immuno was therefore chosen as the measure of tumor immune response to probe for CCMs.

3.2. Screening and Selection of Candidate CCMs Through Assessment of Potential Interactions with Treatment Effect and TMB as Well as Tumor Phenotype

The CCM discovery process was conducted as previously described (Figure 1B; see Methods for details) [32]. Patient samples were classified as JAV-Immunohi if the corresponding tumor expression was greater than the sample median and JAV-Immunolo if it was less than or equal to the median. Nuclei templates from 20 JAV-Immunohi and 20 JAV-Immunolo specimens were hybridized to an array of >900 K oligonucleotides spanning the human genome, thereby identifying 150 loci that could be either positively or negatively associated with the JAV-Immuno classification. Nested polymerase chain reaction (PCR) assays designed to give a positive reading only if the PCR primer targets were juxtaposed by chromatin conformation were applied to 40 JAV-Immunohi and 40 JAV-Immunolo samples (Supplementary Figure S1A). These nested PCR data were used as a training set to build JAV-Immunohi/lo classifier models. PCR data from the remaining 416 samples were used to refine and test the classifier models. The models with optimal test accuracy (0.68) contained 25 CCMs (Figure 1C). Univariate analysis of CCM association with JAV-Immuno scores indicated that individual CCMs could be either positively or negatively associated with JAV-Immuno, depending on the detection titer dilution, suggesting complex relationships between the CCMs and expression of individual genes. The candidate CCMs were further evaluated for association with TMB and OS using Cox proportional hazards and elastic net models (see Section 2—Materials and Methods for details). The 15 CCMs selected in the final model were subjected to further biological annotation (Supplementary Table S2).

3.3. Potential Cell Types and Pathways Represented by the Genes Covered by the CCMs

CCMs in the final model were distributed across the human genome (Supplementary Table S2). These CCMs were detected in <20% to >70% of the patients in the analysis population (Supplementary Figure S1(B-i)), and no linkage disequilibrium was observed (Supplementary Figure S1(B-ii)). Twelve of the 15 CCMs covered distinct regions, and 3 overlapped in a ≈290 kb region on chromosome 22:20707691–20999032 (Supplementary Table S2 and Supplementary Figure S1C). Forty-four major genes in the Human Protein Atlas [33] were mapped to these 15 CCMs, of which 29 genes (66%) were covered by >1 CCM in the final model. In contrast, only 95 of the 323 genes (29%) mapping to the remaining CCMs in the original set were covered by >1 CCM. Thus, the final model was enriched for loci that were covered by multiple CCMs, consistent with possible functional relevance. Only one of the genes in the original 26-gene JAV-Immuno signature (CD6) [31] was present in the 367 genes mapped to the original set, and it was not present in the 15 CCMs from the final model. Individual genes from these 15 CCMs (Table 1) showed both positive and negative associations with JAV-Immuno (Figure 1D), suggesting that host factors may either enhance or diminish anti-tumor immunity.
We evaluated potential expression of genes mapping to the 15 CCMs using the Tabula Sapiens single-cell transcriptomic atlas [34], which contains single-cell sequencing data from almost 500,000 cells, representing over 400 cell types and 4 major cell lineages (immune, endothelial, stromal, and epithelial) assessed in approximately 22 distinct anatomical locations collected from multiple donors. Supplementary Figure S4 shows expression patterns by cell type for 6 genes mapping to chromosome 22:20707691-20999032, which was covered by 3 CCMs in the final model (Supplementary Table S2). Substantial variability in gene expression across cell types was observed, suggesting that cell-specific gene-regulatory machinery operates in conjunction with regulation of chromatin conformation.
We performed a comprehensive analysis of associations between genes and cell types reported in the literature and public databases using the Data4Cure knowledge graph and platform [30]. Genes in the JAV-Immuno signature were predominantly associated with lymphocytes, including T cells, B cells, and natural killer (NK) cells (Figure 2A), as previously described [31]. Genes covered by the selected CCMs were associated with endothelial and fibroblast cell types, in addition to lymphocytes (Figure 2B). Myeloid lineages, including monocytes, neutrophils, and macrophages, were associated with genes in both signatures (Figure 2A,B). JAV-Immuno genes were predominantly associated with Gene Ontology Biological Processes related to lymphocyte activation (Figure 2C) [35]; in contrast, genes mapping to the selected CCM set were associated with a wide range of biological processes (Figure 2D). These findings suggest that the chromatin conformations represented by the selected CCMs may exert both distal and proximal effects on lymphocyte activation.

3.4. Use of Spatial Profiling to Locate CCM Genes Within the TME

Spatial transcriptomic data were used to explore the topographic expression of genes represented by JAV-Immuno and the selected CCMs. A procured bladder cancer specimen was selected for initial evaluation due to its robust representation of immune and non-immune cell types [2]. Figure 3(A-i) shows a hematoxylin and eosin (H&E)–stained image of the specimen identifying the position of lymphoid aggregates. Spatial indexing of transcriptomic data from this specimen identified 8 clusters by BayesSpace (Supplementary Figure S5(A-i)). Similarities in the histology and distribution of predefined gene signatures (Supplementary Figure S5A–C) suggested that the 8 clusters represented 4 functional regions, labeled as epithelial, stromal, transitional, and lymphoid aggregates (Figure 3(A-ii)). Figure 3(B-i) depicts the spatial distribution of genes characteristic of tertiary lymphoid structures (TLS), originally identified in patients with melanoma treated with ICIs [36]. The highest overall expression of the component genes was observed in the functional region labeled as lymphoid aggregates (Figure 3(B-ii,B-iii)), suggesting that this region could be a locus of immune activation and maturation in the tumor microenvironment. Enrichment of gene expression signatures characteristic of T follicular helper cells, mature B cells, cytotoxic T-cells, and dendritic cell recruitment further support the lymphoid aggregate regions as a site for priming and maturation of anti-tumor lymphocytes (Supplementary Figure S5(Ci–Cv)).
The spatial distribution of genes comprising the JAV-Immuno signature was strongly correlated with those in the TLS signature, suggesting that JAV-Immuno is distinctly associated with TLS function (Figure 3(C-i–C-iii), Supplementary Figure S5D). To assess the potential generalizability of these findings, expression of JAV-Immuno genes was evaluated in a collection of specimens representing TLS from bladder, renal, and breast cancers. All 25 genes in the JAV-Immuno signature were positively associated with the presence of TLS in these regions (Figure 3(C-iv)). Overall, JAV-Immuno appears to be more closely associated with TLS than other signatures associated with ICI response [2,37,38,39,40].
The expression of 33 of the 35 major genes mapped to the 15 CCMs was assessed in the bladder specimen and appeared to be distributed broadly throughout the specimen, with slightly lower overall expression in the stromal region (Figure 3(D-i)). Hierarchical clustering of the genes by distribution between the 4 annotated regions identified subsets of genes that were prominent in the lymphoid aggregate and epithelial regions, as well as the stromal region (Figure 3(D-ii,D-iii)). Ten of the 15 selected CCMs in Supplementary Table S2 contained ≥1 gene that was more highly expressed in the lymphoid aggregate region relative to the epithelial region (Figure 3(D-ii)).
As for JAV-Immuno (Figure 3B), potential generalizability of the CCM findings was assessed by evaluating expression of the genes in the TLS specimen collection (Figure 3(D-iv)). Several genes positively associated with JAV-Immuno (Figure 1D) and expressed in the lymphoid aggregate region (Figure 3(D-ii)) were associated with TLS regions in the collection (Figure 3(D-iv)), including POU2F2, MBNL1, ABI3BP, VPS13C, and CPEB1. Other genes, including CMPK2, RSAD2, RNF144A, and SUCNR1, were positively associated with JAV-Immuno but were expressed more prominently outside the lymphoid aggregate regions (Figure 3(D-ii)) and were not as strongly associated with TLS in the collection (Figure 3(D-iv)). Participation in antiviral and antibacterial responses has been described for RSAD2 [41,42] and CMPK2 [42,43]. Reduced levels of RNF144A have been associated with PD-L1 stabilization and tumorigenesis in mouse bladder cancer models [44] and poor prognosis in patients with breast cancer [45]. SUCNR1 has been linked to altered immune infiltration of tumors in ovarian cancer [46] and renal cancer [47]. The adaptive immune processes represented by JAV-Immuno may thus be tuned by responses to stimuli in the tumor microenvironment that are outside of the lymphoid aggregates.

3.5. Specific Assessment of Associations Between a CCM-Covered Gene, TMB, and Avelumab Treatment Effect

Potential relationships between genes covered by the CCMs with TMB and OS with avelumab plus BSC were assessed using a 3-way interaction test (Figure 4A). This analysis was conducted in genes that were expressed in >50% of tumor samples and/or had a coefficient of variation of >5% (Supplementary Figure S6). The strongest positive monogenic association was seen with POU2F2, which also showed a positive association with JAV-Immuno (Figure 1D). POU2F2 is a transcription factor involved in B-cell differentiation and collaboration between B-cells and T-cells and is expressed in multiple cell types involved in antigen presentation, including B-cells and monocytes (Figure 2B) [48,49]. The presence of the corresponding CCM was associated with low POU2F2 gene expression and decreased expression of POU2F2 target genes, suggesting that the CCM negatively regulates POU2F2 and downstream processes, including generation of mature B cells and TLS formation (Figure 4B).
We then explored relationships between the POU2F2 CCM, TMB, and OS in the avelumab plus BSC and BSC arms. POU2F2 CCM interactions with TMB and OS were more pronounced in the avelumab plus BSC arm than in the BSC arm (Table 2). The absence vs. presence of the POU2F2 CCM was associated with a positive effect on OS in patients treated with avelumab plus BSC whose tumors displayed low TMB (less than the median value of 7.66 nonsynonymous single nucleotide variants per megabase); median OS was 37 vs. 18 months (HR, 0.46 [95% CI, 0.24–0.89]; p = 0.02). These findings suggest that the effect of CCM modulation on POU2F2 expression may be strongest when the treatment effect is contingent on immune responses to limited tumor immunogenicity.

4. Discussion

Large pivotal studies such as the phase 3 JAVELIN Bladder 100 trial are particularly valuable for investigating complex interactions between tumor and patient biomarkers in advanced UC, since (1) avelumab 1L maintenance was associated with significant OS and PFS benefit and (2) avelumab was compared with BSC rather than an active treatment control. Therefore, we utilized this study to explore the possible connections between tumor phenotype and peripheral blood epigenetic biomarkers detected by a clinically validated platform. These findings are subject to known caveats related to exploratory post hoc analyses, including the possibility of false discovery and the lack of statistical power to detect all meaningful associations; as such, they should be considered hypothesis-generating. Necessary next steps include (a) validation in independent datasets, (b) further investigation of potential clinical confounders such as age, sex, ECOG performance status, or tumor staging, (c) confirmation of generalizability and robustness given potential biases in the training set, and (d) prospective demonstration of utility in clinical studies before consideration for use in treating patients. Even so, this study lays a foundation for further identification and validation of CCM biomarkers for use in clinical development.
The associations between tumor phenotype, blood chromatin structure, and avelumab treatment effect are consistent with the hypothesis that systemic host chromatin structures may shape the tumor microenvironment and thereby influence the treatment effect of avelumab and other ICI therapies. Although the initial CCM screen evaluated associations with a gene signature of immune activity, including TLS formation, the emerging CCMs covered genes that are expressed by tumor cells (e.g., RSAD2, CMPK2) and stromal fibroblasts (e.g., SUCNR1) as well as immune cells (e.g., POU2F2). The observation that peripheral blood CCMs could be associated with genes in distinct regions of the TME has intriguing parallels to the observation, from tumor gene mutation and expression, that multiple pathways reflecting immune-tumor interactions were associated with avelumab treatment effect [2]. The generalizability of the distribution of these genes across immune, stromal, and tumor compartments needs to be confirmed via spatial profiling of additional representative tumor sets. Such investigations will clarify the potential contribution of systemic and tumor-specific biomarkers to ICI treatment optimization in UC, whether alone or in combination with other treatments.
The findings from these analyses open multiple avenues for further investigation and validation. The relationship between TMB, TLS formation in tumors, and POU2F2 gene expression may help to explain why the performance of TMB as an ICI treatment biomarker has been variable [2]. Similar investigations into specific contributions to anti-tumor immune function may be conducted for MBNL1, a pre-mRNA alternative splicing regulator [52,53]; ABI3BP, an extracellular matrix protein associated with tumor suppression and immune infiltration [54,55]; and VPS13C, a lipid transport protein participating in mitochondrial stability [56] and in associations between lysosomal function and innate immune activation [57]. Direct contributions of CMPK2 and RSAD2 to anti-tumor immune response induction, as opposed to serving as indirect biomarkers of interferon signaling, should be clarified. Potential contributions of other genes with currently poorly defined functions, such as ZNF family members and C2CD4A/B, remain to be fully assessed. Such investigations may further enable the appropriate interpretation and clinical validation of these biomarkers. Interactions between peripheral blood CCMs and tumor mutations or mutation burden that are also measurable in blood may provide novel opportunities for non-invasive monitoring of patient responses to ICI treatment.

5. Conclusions

A clinically validated CCM platform was applied to peripheral blood from patients with matched tumor data from a pivotal phase 3 study of ICI therapy for advanced UC. The emerging associations between host factors in peripheral blood and tumor molecular phenotype illustrate the utility of exploratory analyses from pivotal studies for biomarker-guided advances in treatment strategies for patients with advanced UC.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers17142332/s1, Table S1. Characteristics of the biomarker analysis set compared with the full analysis set. BSC, best supportive care; CR, complete response; OBD, Oxford BioDynamics; PR, partial response; SD, stable disease; St. Dev., standard deviation. Table S2. List of 15 markers passing the JAV-Immuno, TMB, and/or treatment interaction criteria. Twenty-five peripheral blood markers that met criteria for predicting JAV-Immuno levels in tumor were further screened for potential interactions with TMB and treatment based on OS outcome. The effect column indicates whether a marker/dilution pair was selected based on Cox models for its main effect (1-Main), its interaction with treatment alone (2-Way), or its interaction with treatment and TMB (3-way). The p values were based on 2-sided Wald test. (See the Methods section for details of the pre-filtering and modeling steps). Some chromosomal loci were covered by >1 marker from the original 150-marker set as indicated. Genes in bold font were more strongly expressed in the lymphoid aggregate region relative to the epithelial region of the bladder cancer specimen shown in Figure 3D. OS, overall survival; TMB, tumor mutation burden. Figure S1. Characterization of CCMs emerging from screening and modeling workflows. A. CONSORT flow diagram of patient disposition. Biomarker analyses were performed on specimens from patients who both (1) received ≥1 dose of study drug in the avelumab + BSC arm or completed cycle 1 day 1 in the BSC arm (ie, the safety set) and (2) had tumor transcriptome and chromatin marker data available. B. Prevalence and linkage of markers passing pre-filtering criteria for multivariate modeling. i. For each marker, prevalence was calculated as the proportion of patients with the marker present among all patients analyzed (N = 496). ii. Phi coefficient (measure of association between binary variables) matrix among pre-filtered markers. C. Map of genes covered by chromatin markers. Circles represent genes covered by at least 1 marker. Chromosome and starting positions of marker loci or individual genes (from JAV-Immuno) were assigned using hg38 coordinates. Selected: markers passing TMB modeling criteria. Not selected: remaining markers from original array screen. BSC, best supportive care; CCM, chromatin conformation marker; Chr, chromosome; TMB, tumor mutation burden. Figure S2. OS and PFS outcomes with the full analysis set and biomarker analysis set. OS, overall survival; PFS, progression-free survival. Figure S3. Identification of optimal hyperparameter pairs for elastic modeling to select markers interacting with TMB and/or treatment on OS outcome. For each round of 5-fold cross-validation, an optimal pair of α and λ associated with minimal model error was identified. The same procedure was iterated 1000 times to account for potential cross-validation error. The frequency distribution of the optimal hyperparameter pairs was plotted in a Hexbin plot with color indicating the number of optimal hyperparameter pairs located inside each hexagon. The most frequently selected hyperparameter pairs from the hexagon with the largest count (red arrow) was chosen as optimal and was applied to the full dataset to obtain the final models. BSC, best supportive care; OS, overall survival; PFS, progression-free survival, OBD, Oxford BioDynamics; TMB, tumor mutation burden. Figure S4. Heterogeneous expression of genes in a single marker across multiple cell types in Tabula Sapiens. A. Immune. B. Endothelial. C. Stromal. D. Epithelial. Cell types from the CURIE knowledge graph were mapped to the cell types reported [24]. The color represents aggregation by mean normalized by z-score. The size of circles is proportional to the percentage of cells with non-zero expression. NK, natural killer. Figure S5. Application of cell-type gene expression signatures to spatial indexing data. A. i. Original 8 clusters identified by BayesSpace. ii-iv. Gene expression profiles representing epithelial, stromal, endothelial, fibroblast, and myCAF cells. B. Gene expression profiles representative of (i) M1 macrophages, (ii) M2 macrophages, (iii) myeloid DCs, (iv) monocytes, (v) NK cells, and (vi) plasmacytoid DCs. C. Gene expression profiles representative of (i) T follicular helper cells, (ii) mature B cells, (iii) CD8 cytotoxic cells, (iv) cytotoxic granules, and (v) CCR7 and ligands CCL19 and CCL21, which are involved in recruitment of mature DCs to lymphoid tissue. D. Correlation of the average JAV-Immuno signature vs. TLS signature across 4587 Visium spots indicated that the JAV-Immuno–expressing spots also contain TLS. Best-fit line (blue) shows a positive correlation (R2 = 0.72; p < 5.29 × 10−244). DC, dendritic cell; myCAF, myofibroblastic-like cancer-associated fibroblast; NK, natural killer; TLS, tertiary lymphoid structures. Figure S6. Distribution of gene expression level. Genes in the selected marker set were pre-filtered to separate out genes that were expressed in ≤50% of JAVELIN Bladder 100 tumor samples and/or had a coefficient of variation of ≤5%. TPM, transcript count per million.

Author Contributions

Conceptualization: T.P. and C.B.D. Data curation: J.P., M.S. (Michelle Saul), K.A.C., R.N., E.H., R.R., A.d.P. and J.D. Formal Analysis: J.P. and E.H. Investigation: T.P., S.S.S., J.B., C.N.S., P.G., A.d.P. and A.D. Methodology: E.H., M.S. (Matthew Salter), R.P., A.D., J.G., A.A., R.N., K.A.C., J.P., S.D., C.B.D., R.R., J.D., D.F. and R.A. Project administration: C.B.D. Software: E.H., R.R. and J.D. Validation: E.H., M.S. (Matthew Salter), R.P., J.G., A.A. and C.-H.H. Writing—original draft: T.P. and C.B.D. Writing—review and editing: T.P., S.S.S., P.G., K.A.C., M.S. (Michelle Saul) and C.B.D. All authors have read and agreed to the published version of the manuscript.

Funding

This study was sponsored by Merck (CrossRef Funder ID: 10.13039/100009945) and was previously conducted under an alliance between Merck and Pfizer.

Institutional Review Board Statement

The JAVELIN Bladder 100 trial was conducted in accordance with the ethics principles of the Declaration of Helsinki and the Good Clinical Practice guidelines, defined by the International Council for Harmonization. The protocol, amendments, and informed consent forms were approved by the institutional review board or independent ethics committee at each trial site.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Any requests for data by qualified scientific and medical researchers for legitimate research purposes will be subject to Merck’s Data Sharing Policy. All requests should be submitted in writing to Merck’s data sharing portal (https://www.merckgroup.com/en/research/our-approach-to-research-and-development/healthcare/clinical-trials/commitment-responsible-data-sharing.html, accessed on 30 March 2025). When Merck has a co-research, co-development, co-marketing, or co-promotion agreement, or when the product has been out-licensed, the responsibility for disclosure might be dependent on the agreement between parties. Under these circumstances, Merck will endeavor to gain agreement to share data in response to requests.

Acknowledgments

Medical writing support was provided by Katherine Quiroz-Figueroa of Nucleus Global and was funded by Merck. Mehrnoush Dezfouli, Benedict Egan, Paulina Brajer, Catriona Williams, and Tomas Lavin conducted EpiSwitch platform sample processing for detection of CCMs.

Conflicts of Interest

T. Powles has served in consulting or advisory roles for Astellas Pharma, AstraZeneca, Bristol Myers Squibb, Eisai, Exelixis, Incyte, Ipsen, Johnson & Johnson, Merck, MSD, Novartis, Pfizer, Roche, and Seagen and has received travel and accommodation expenses from Pfizer. S. S. Sridhar has served in consulting or advisory roles for Astellas Pharma, AstraZeneca, Bayer, Bicycle Therapeutics, Bristol Myers Squibb, Eisai, Gilead, Ipsen, Janssen, MSD, Pfizer, and Seagen and has received institutional research funding from Bayer, Merck, Janssen, and Seagen. J. Bellmunt has served in consulting or advisory roles for Astellas Pharma, AstraZeneca/MedImmune, Bristol Myers Squibb, Genentech, Novartis, Pfizer, Pierre Fabre, and Merck; has received travel and accommodation expenses from Ipsen, MSD, and Pfizer; reports patents, royalties, and other intellectual property from UpToDate; reports stock and other ownership interests in Rainier Therapeutics; has received honoraria from UpToDate; and has received institutional research funding from Millennium, Pfizer, Sanofi, and Merck. C. N. Sternberg has served in consulting or advisory roles for Bayer, Merck, MSD, Pfizer, Roche, Amgen, Incyte, Bristol Myers Squibb, AstraZeneca, Medscape, UroToday, Astellas Pharma, Sanofi, Genzyme, Gilead Sciences, and Foundation Medicine. P. Grivas has served in consulting or advisory roles for Merck, Bristol Myers Squibb, AstraZeneca, Pfizer, Janssen, Roche, Astellas Pharma, Gilead Sciences, Fresenius Kabi, G1 Therapeutics, Aadi Bioscience, CG Oncology, Bicycle Therapeutics, MSD, Strata Oncology, ImmunityBio, AbbVie, and Asieris Pharmaceuticals and has received institutional research funding from Bristol Myers Squibb, Merck, QED Therapeutics, Mirati Therapeutics, MSD, G1 Therapeutics, Genentech, Gilead Sciences, Acrivon Therapeutics, and ALX Oncology. E. Hunter, M. Salter, R. Powell, A. Dring, J. Green, and A. Akoulitchev are employees of Oxford Biodynamics. A. Akoulitchev is the company director of Oxford BioDynamics. R. Ronen and J. Dutkowski are employees of Data4Cure. R. Amezquita and J. Pu were employees of Pfizer at the time of the study. C.-H. Huang, D. Fernandez, R. Nameki, K. A. Ching, M. Saul, S. Deng, and A. di Pietro are employees of and report stock or other ownership interests in Pfizer during the conduct of these analyses. C. B. Davis was an employee of Pfizer at the time of the study.

References

  1. Banchereau, R.; Leng, N.; Zill, O.; Sokol, E.; Liu, G.; Pavlick, D.; Maund, S.; Liu, L.-F.; Kadel, E.; Baldwin, N.; et al. Molecular determinants of response to PD-L1 blockade across tumor types. Nat. Commun. 2021, 12, 3969. [Google Scholar] [CrossRef]
  2. Powles, T.; Sridhar, S.S.; Loriot, Y.; Bellmunt, J.; Mu, X.J.; Ching, K.A.; Pu, J.; Sternberg, C.N.; Petrylak, D.P.; Tambaro, R.; et al. Avelumab maintenance in advanced urothelial carcinoma: Biomarker analysis of the phase 3 JAVELIN Bladder 100 trial. Nat. Med. 2021, 27, 2200–2211. [Google Scholar] [CrossRef] [PubMed]
  3. Powles, T.; Park, S.H.; Voog, E.; Caserta, C.; Valderrama, B.P.; Gurney, H.; Kalofonos, H.; Radulović, S.; Demey, W.; Ullén, A.; et al. Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N. Engl. J. Med. 2020, 383, 1218–1230. [Google Scholar] [CrossRef]
  4. Powles, T.; Bellmunt, J.; Comperat, E.; De Santis, M.; Huddart, R.; Loriot, Y.; Necchi, A.; Valderrama, B.P.; Ravaud, A.; Shariat, S.F.; et al. ESMO Clinical Practice Guideline interim update on first-line therapy in advanced urothelial carcinoma. Ann. Oncol. 2024, 35, 485–490. [Google Scholar] [CrossRef]
  5. Grivas, P.; Grande, E.; Davis, I.D.; Moon, H.H.; Grimm, M.-O.; Gupta, S.; Barthélémy, P.; Thibault, C.; Guenther, S.; Hanson, S.; et al. Avelumab first-line maintenance treatment for advanced urothelial carcinoma: Review of evidence to guide clinical practice. ESMO Open 2023, 8, 102050. [Google Scholar] [CrossRef] [PubMed]
  6. Mellman, I.; Chen, D.S.; Powles, T.; Turley, S.J. The cancer-immunity cycle: Indication, genotype, and immunotype. Immunity 2023, 56, 2188–2205. [Google Scholar] [CrossRef]
  7. Khan, Z.; Di Nucci, F.; Kwan, A.; Hammer, C.; Mariathasan, S.; Rouilly, V.; Carroll, J.; Fontes, M.; Acosta, S.L.; Guardino, E.; et al. Polygenic risk for skin autoimmunity impacts immune checkpoint blockade in bladder cancer. Proc. Natl. Acad. Sci. USA 2020, 117, 12288–12294. [Google Scholar] [CrossRef]
  8. Dizman, N.; Meza, L.; Bergerot, P.; Alcantara, M.; Dorff, T.; Lyou, Y.; Frankel, P.; Cui, Y.; Mira, V.; Llamas, M.; et al. Nivolumab plus ipilimumab with or without live bacterial supplementation in metastatic renal cell carcinoma: A randomized phase 1 trial. Nat. Med. 2022, 28, 704–712. [Google Scholar] [CrossRef]
  9. Cuartero, S.; Stik, G.; Stadhouders, R. Three-dimensional genome organization in immune cell fate and function. Nat. Rev. Immunol. 2022, 23, 206–221. [Google Scholar] [CrossRef] [PubMed]
  10. Johanson, T.M.; Chan, W.F.; Keenan, C.R.; Allan, R.S. Genome organization in immune cells: Unique challenges. Nat. Rev. Immunol. 2019, 19, 448–456. [Google Scholar] [CrossRef]
  11. Tordini, F.; Aldinucci, M.; Milanesi, L.; Liò, P.; Merelli, I. The genome conformation as an integrator of multi-omic data: The example of damage spreading in cancer. Front. Genet. 2016, 7, 194. [Google Scholar] [CrossRef]
  12. Bonev, B.; Cavalli, G. Organization and function of the 3D genome. Nat. Rev. Genet. 2016, 17, 661–678. [Google Scholar] [CrossRef]
  13. Tien, F.-M.; Lu, H.-H.; Lin, S.-Y.; Tsai, H.-C. Epigenetic remodeling of the immune landscape in cancer: Therapeutic hurdles and opportunities. J. Biomed. Sci. 2023, 30, 3. [Google Scholar] [CrossRef]
  14. Kempfer, R.; Pombo, A. Methods for mapping 3D chromosome architecture. Nat. Rev. Genet. 2019, 21, 207–226. [Google Scholar] [CrossRef]
  15. Dekker, J.; Rippe, K.; Dekker, M.; Kleckner, N. Capturing chromosome conformation. Science 2002, 295, 1306–1311. [Google Scholar] [CrossRef]
  16. Jakub, J.W.; Grotz, T.E.; Jordan, P.; Hunter, E.; Pittelkow, M.; Ramadass, A.; Akoulitchev, A.; Markovic, S. A pilot study of chromosomal aberrations and epigenetic changes in peripheral blood samples to identify patients with melanoma. Melanoma Res. 2015, 25, 406–411. [Google Scholar] [CrossRef]
  17. Carini, C.; Hunter, E.; Ramadass, A.S.; Green, J.; Akoulitchev, A.; McInnes, I.B.; Goodyear, C.S. Chromosome conformation signatures define predictive markers of inadequate response to methotrexate in early rheumatoid arthritis. J. Transl. Med. 2018, 16, 18. [Google Scholar] [CrossRef]
  18. Alshaker, H.; Mills, R.; Hunter, E.; Salter, M.; Ramadass, A.; Skinner, B.M.; Westra, W.; Green, J.; Akoulitchev, A.; Winkler, M.; et al. Chromatin conformation changes in peripheral blood can detect prostate cancer and stratify disease risk groups. J. Transl. Med. 2021, 19, 46. [Google Scholar] [CrossRef]
  19. Pchejetski, D.; Hunter, E.; Dezfouli, M.; Salter, M.; Powell, R.; Green, J.; Naithani, T.; Koutsothanasi, C.; Alshaker, H.; Jaipuria, J.; et al. Circulating chromosome conformation signatures significantly enhance PSA positive predicting value and overall accuracy for prostate cancer detection. Cancers 2023, 15, 821. [Google Scholar] [CrossRef] [PubMed]
  20. Hunter, E.; Salter, M.; Powell, R.; Dring, A.; Naithani, T.; Chatziioannou, M.E.; Gebregzabhar, A.; Issa, M.; Green, J.; Ng, S.; et al. Development and validation of blood-based predictive biomarkers for response to PD-1/PD-L1 checkpoint inhibitors: Evidence of a universal systemic core of 3D immunogenetic profiling across multiple oncological indications. Cancers 2023, 15, 2696. [Google Scholar] [CrossRef] [PubMed]
  21. Quinlan, A.R. BEDTools: The Swiss-army tool for genome feature analysis. Curr. Protoc. Bioinform. 2014, 47, 11–12. [Google Scholar] [CrossRef]
  22. Chen, T.; Guestrin, C. XGBoost: A scalable tree boosting system. In Proceedings of the 22nd ACM SIGKDD International Conference on Knowledge Discovery and Data Mining, San Francisco, CA, USA, 13–17 August 2016; Association for Computing Machinery: San Francisco, CA, USA, 2016; pp. 785–794. [Google Scholar]
  23. Biecek, P.; Burzykowski, T. Shapley. Additive Explanations (SHAP) for Average Attributions. In Explanatory Model Analysis, 1st ed.; Chapman and Hall/CRC: Boca Raton, FL, USA, 2021; pp. 95–106. [Google Scholar]
  24. Salter, M.; Corfield, E.; Ramadass, A.; Grand, F.; Green, J.; Westra, J.; Lim, C.R.; Farrimond, L.; Feneberg, E.; Scaber, J.; et al. Initial identification of a blood-based chromosome conformation signature for aiding in the diagnosis of amyotrophic lateral sclerosis. EBioMedicine 2018, 33, 169–184. [Google Scholar] [CrossRef]
  25. Zou, H.; Hastie, T. Regularization and variable selection via the elastic net. J. R. Stat. Soc. Stat. Methodol. Ser. B 2005, 67, 301–320. [Google Scholar] [CrossRef]
  26. Tibshirani, R. Regression shrinkage and selection via the Lasso. J. R. Stat. Soc. Ser. B Stat. Methodol. 1996, 58, 267–288. [Google Scholar] [CrossRef]
  27. Hoerl, A.E.; Kennard, R.W. Ridge regression: Applications to nonorthogonal problems. Technometrics 1970, 12, 69–82. [Google Scholar] [CrossRef]
  28. Tiddi, I.; Schlobach, S. Knowledge graphs as tools for explainable machine learning: A survey. Artif. Intell. 2022, 302, 103627. [Google Scholar] [CrossRef]
  29. Zhao, E.; Stone, M.R.; Ren, X.; Guenthoer, J.; Smythe, K.S.; Pulliam, T.; Williams, S.R.; Uytingco, C.R.; Taylor, S.E.B.; Nghiem, P.; et al. Spatial transcriptomics at subspot resolution with BayesSpace. Nat. Biotechnol. 2021, 39, 1375–1384. [Google Scholar] [CrossRef]
  30. Hu-Lieskovan, S.; Braiteh, F.; Grilley-Olson, J.E.; Wang, X.; Forgie, A.; Bonato, V.; Jacobs, I.A.; Chou, J.; Johnson, M.L. Association of tumor mutational burden and immune gene expression with response to PD-1 blockade by sasanlimab across tumor types and routes of administration. Target. Oncol. 2021, 16, 773–787. [Google Scholar] [CrossRef]
  31. Motzer, R.J.; Robbins, P.B.; Powles, T.; Albiges, L.; Haanen, J.B.; Larkin, J.; Mu, X.J.; Ching, K.A.; Uemura, M.; Pal, S.K.; et al. Avelumab plus axitinib versus sunitinib in advanced renal cell carcinoma: Biomarker analysis of the phase 3 JAVELIN Renal 101 trial. Nat. Med. 2020, 26, 1733–1741. [Google Scholar] [CrossRef] [PubMed]
  32. Yan, H.; Hunter, E.; Akoulitchev, A.; Park, P.; Winchester, D.J.; A Moo-Young, T.; A Prinz, R. Epigenetic chromatin conformation changes in peripheral blood can detect thyroid cancer. Surgery 2018, 165, 44–49. [Google Scholar] [CrossRef]
  33. Karlsson, M.; Zhang, C.; Méar, L.; Zhong, W.; Digre, A.; Katona, B.; Sjöstedt, E.; Butler, L.; Odeberg, J.; Dusart, P.; et al. A single-cell type transcriptomics map of human tissues. Sci. Adv. 2021, 7, eabh2169. [Google Scholar] [CrossRef]
  34. Tabula Sapiens Consortium; Jones, R.C.; Karkanias, J.; Krasnow, M.A.; Pisco, A.O.; Quake, S.R.; Salzman, J.; Yosef, N.; Bulthaup, B.; Brown, P.; et al. The Tabula Sapiens: A multiple-organ, single-cell transcriptomic atlas of humans. Science 2022, 376, eabl4896. [Google Scholar]
  35. Ashburner, M.; Ball, C.A.; Blake, J.A.; Botstein, D.; Butler, H.; Cherry, J.M.; Davis, A.P.; Dolinski, K.; Dwight, S.S.; Eppig, J.T.; et al. Gene ontology: Tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 2000, 25, 25–29. [Google Scholar] [CrossRef]
  36. Helmink, B.A.; Reddy, S.M.; Gao, J.; Zhang, S.; Basar, R.; Thakur, R.; Yizhak, K.; Sade-Feldman, M.; Blando, J.; Han, G.; et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020, 577, 549–555. [Google Scholar] [CrossRef]
  37. Higgs, B.W.; Morehouse, C.A.; Streicher, K.; Brohawn, P.Z.; Pilataxi, F.; Gupta, A.; Ranade, K. Interferon gamma messenger RNA signature in tumor biopsies predicts outcomes in patients with non-small cell lung carcinoma or urothelial cancer treated with durvalumab. Clin. Cancer Res. 2018, 24, 3857–3866. [Google Scholar] [CrossRef]
  38. Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C.; et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): A multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016, 387, 1837–1846. [Google Scholar] [CrossRef]
  39. Eckstein, M.; Strissel, P.; Strick, R.; Weyerer, V.; Wirtz, R.; Pfannstiel, C.; Wullweber, A.; Lange, F.; Erben, P.; Stoehr, R.; et al. Cytotoxic T-cell-related gene expression signature predicts improved survival in muscle-invasive urothelial bladder cancer patients after radical cystectomy and adjuvant chemotherapy. J. Immunother. Cancer 2020, 8, e000162. [Google Scholar] [CrossRef]
  40. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef]
  41. Wiedemann, G.M.; Geary, C.D.; Lau, C.M.; Sun, J.C. Cutting edge: STAT1-mediated epigenetic control of Rsad2 promotes clonal expansion of antiviral NK cells. J. Immunol. 2020, 205, 21–25. [Google Scholar] [CrossRef]
  42. Arnold, N.; Girke, T.; Sureshchandra, S.; Nguyen, C.; Rais, M.; Messaoudi, I. Genomic and functional analysis of the host response to acute simian varicella infection in the lung. Sci. Rep. 2016, 6, srep34164. [Google Scholar] [CrossRef]
  43. Larsson, I.; Uhlén, M.; Zhang, C.; Mardinoglu, A. Genome-scale metabolic modeling of glioblastoma reveals promising targets for drug development. Front. Genet. 2020, 11, 381. [Google Scholar] [CrossRef]
  44. Ho, S.-R.; Lee, Y.-C.; Ittmann, M.M.; Lin, F.-T.; Chan, K.S.; Lin, W.-C. RNF144A deficiency promotes PD-L1 protein stabilization and carcinogen-induced bladder tumorigenesis. Cancer Lett. 2021, 520, 344–360. [Google Scholar] [CrossRef] [PubMed]
  45. Yang, Y.-L.; Zhang, Y.; Li, D.-D.; Zhang, F.-L.; Liu, H.-Y.; Liao, X.-H.; Xie, H.-Y.; Lu, Q.; Zhang, L.; Hong, Q.; et al. RNF144A functions as a tumor suppressor in breast cancer through ubiquitin ligase activity-dependent regulation of stability and oncogenic functions of HSPA2. Cell Death Differ. 2019, 27, 1105–1118. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, J.; Zhang, Q.; Yang, Y.; Wang, Q. Association between succinate receptor SUCNR1 expression and immune infiltrates in ovarian cancer. Front. Mol. Biosci. 2020, 7, 150. [Google Scholar] [CrossRef]
  47. Najm, R.; Hachim, M.Y.; Kandasamy, R.K. Divulging a pleiotropic role of succinate receptor SUCNR1 in renal cell carcinoma microenvironment. Cancers 2022, 14, 6064. [Google Scholar] [CrossRef] [PubMed]
  48. Hodson, D.J.; Shaffer, A.L.; Xiao, W.; Wright, G.W.; Schmitz, R.; Phelan, J.D.; Yang, Y.; Webster, D.E.; Rui, L.; Kohlhammer, H.; et al. Regulation of normal B-cell differentiation and malignant B-cell survival by OCT2. Proc. Natl. Acad. Sci. USA 2016, 113, E2039–E2046. [Google Scholar] [CrossRef]
  49. Corcoran, L.; Emslie, D.; Kratina, T.; Shi, W.; Hirsch, S.; Taubenheim, N.; Chevrier, S. Oct2 and Obf1 as facilitators of B:T cell collaboration during a humoral immune response. Front. Immunol. 2014, 5, 108. [Google Scholar] [CrossRef]
  50. Rodriguez, A.B.; Peske, J.D.; Woods, A.N.; Leick, K.M.; Mauldin, I.S.; Meneveau, M.O.; Young, S.J.; Lindsay, R.S.; Melssen, M.M.; Cyranowski, S.; et al. Immune mechanisms orchestrate tertiary lymphoid structures in tumors via cancer-associated fibroblasts. Cell Rep. 2021, 36, 109422. [Google Scholar] [CrossRef]
  51. Hennequin, A.; Derangère, V.; Boidot, R.; Apetoh, L.; Vincent, J.; Orry, D.; Fraisse, J.; Causeret, S.; Martin, F.; Arnould, L.; et al. Tumor infiltration by Tbet+ effector T cells and CD20+ B cells is associated with survival in gastric cancer patients. OncoImmunology 2016, 5, e1054598. [Google Scholar] [CrossRef]
  52. Konieczny, P.; Stepniak-Konieczna, E.; Sobczak, K. MBNL proteins and their target RNAs, interaction and splicing regulation. Nucleic Acids Res. 2014, 42, 10873–10887. [Google Scholar] [CrossRef]
  53. Sznajder, Ł.J.; Scotti, M.M.; Shin, J.; Taylor, K.; Ivankovic, F.; Nutter, C.A.; Aslam, F.N.; Subramony, S.H.; Ranum, L.P.W.; Swanson, M.S. Loss of MBNL1 induces RNA misprocessing in the thymus and peripheral blood. Nat. Commun. 2020, 11, 2022. [Google Scholar] [CrossRef] [PubMed]
  54. Delfín, D.A.; DeAguero, J.L.; McKown, E.N. The extracellular matrix protein ABI3BP in cardiovascular health and disease. Front. Cardiovasc. Med. 2019, 6, 23. [Google Scholar] [CrossRef] [PubMed]
  55. Feng, Y.; Han, X.; Zhang, Z.; Qiao, H.; Tang, H. ABI3BP is a prognosis biomarker related with clinicopathological features and immunity infiltration of lung tumor. Front. Genet. 2023, 13, 1085785. [Google Scholar] [CrossRef] [PubMed]
  56. Lesage, S.; Drouet, V.; Majounie, E.; Deramecourt, V.; Jacoupy, M.; Nicolas, A.; Cormier-Dequaire, F.; Hassoun, S.M.; Pujol, C.; Ciura, S.; et al. Loss of VPS13C function in autosomal-recessive parkinsonism causes mitochondrial dysfunction and increases PINK1/Parkin-dependent mitophagy. Am. J. Hum. Genet. 2016, 98, 500–513. [Google Scholar] [CrossRef]
  57. Wang, X.; Xu, P.; Bentley-DeSousa, A.; Hancock-Cerutti, W.; Cai, S.; Johnson, B.T.; Tonelli, F.; Shao, L.; Talaia, G.; Alessi, D.R.; et al. The bridge-like lipid transport protein VPS13C/PARK23 mediates ER-lysosome contacts following lysosome damage. Nat. Cell Biol. 2025, 27, 776–789. [Google Scholar] [CrossRef]
Figure 1. Blood chromatin markers associated with JAV-Immuno in tumor and interactions between TMB, treatment, and OS. (A). Modulation of TMB interactions with treatment based on OS outcome by JAV-Immuno score. Kaplan-Meier plots of high baseline TMB (≥median) vs. low baseline TMB (<median) are shown for subgroups defined by treatment (avelumab plus BSC or BSC) and baseline JAV-Immuno scorehi/lo (based on median split). A Cox proportional hazards model with no adjustment for baseline covariates was used. A 2-sided Wald test was used to determine p-values. (B). Discovery of CCMs using EpiSwitch platform. i. DNA from fixed and permeabilized nuclei was digested with restriction enzymes and hybridized to an oligonucleotide array designed to detect sequences brought in proximity by chromatin conformation. ii. Nested PCR assays were designed for 150 sequences that could potentially differentiate between patients with JAV-Immunohi and JAV-Immunolo tumors. iii. PCR product indicated whether a given chromatin marker was present or absent. Predictive models based on the presence or absence of chromatin marker PCR products and built using methods such as XGBoost were used to narrow the list of candidate markers to 25. (C). Associations between blood chromatin markers and tumor JAV-Immuno levels. Univariate associations are shown between JAV-Immuno score and the status of 25 markers (present vs. absent) detected by PCR assay across 3 template dilution levels (1×, 2×, and 4×). The x-axis represents magnitude and direction of differences in mean JAV-Immuno levels. A Wilcoxon rank-sum test comparing JAV-Immuno scores between the presence and absence of each marker was performed at each dilution level. The y-axis represents the log10 of 2-sided p-values based on the test. Markers and dilutions that were selected in the final model are labeled. (D). Correlations between individual genes covered by selected CCM markers and JAV-Immuno signature scores in tumors are plotted as Pearson coefficients with log10 (p value). Genes with uncorrected p ≤ 0.001 are labeled.
Figure 1. Blood chromatin markers associated with JAV-Immuno in tumor and interactions between TMB, treatment, and OS. (A). Modulation of TMB interactions with treatment based on OS outcome by JAV-Immuno score. Kaplan-Meier plots of high baseline TMB (≥median) vs. low baseline TMB (<median) are shown for subgroups defined by treatment (avelumab plus BSC or BSC) and baseline JAV-Immuno scorehi/lo (based on median split). A Cox proportional hazards model with no adjustment for baseline covariates was used. A 2-sided Wald test was used to determine p-values. (B). Discovery of CCMs using EpiSwitch platform. i. DNA from fixed and permeabilized nuclei was digested with restriction enzymes and hybridized to an oligonucleotide array designed to detect sequences brought in proximity by chromatin conformation. ii. Nested PCR assays were designed for 150 sequences that could potentially differentiate between patients with JAV-Immunohi and JAV-Immunolo tumors. iii. PCR product indicated whether a given chromatin marker was present or absent. Predictive models based on the presence or absence of chromatin marker PCR products and built using methods such as XGBoost were used to narrow the list of candidate markers to 25. (C). Associations between blood chromatin markers and tumor JAV-Immuno levels. Univariate associations are shown between JAV-Immuno score and the status of 25 markers (present vs. absent) detected by PCR assay across 3 template dilution levels (1×, 2×, and 4×). The x-axis represents magnitude and direction of differences in mean JAV-Immuno levels. A Wilcoxon rank-sum test comparing JAV-Immuno scores between the presence and absence of each marker was performed at each dilution level. The y-axis represents the log10 of 2-sided p-values based on the test. Markers and dilutions that were selected in the final model are labeled. (D). Correlations between individual genes covered by selected CCM markers and JAV-Immuno signature scores in tumors are plotted as Pearson coefficients with log10 (p value). Genes with uncorrected p ≤ 0.001 are labeled.
Cancers 17 02332 g001aCancers 17 02332 g001b
Figure 2. Cell types and functions of genes covered by the selected EpiSwitch CCM. The heat maps display aggregated associations between genes (rows) and cell types (columns in A,B) or functions (columns in C,D) across public data represented in the Data4Cure CURIE knowledge graph. The association score summarizes data and literature evidence from publicly available datasets and publications linking biomedical entities. Hierarchical clustering was applied to associations to generate the heat map. Association between cell types and genes in the JAV-Immuno signature (A) and the selected CCM set (B). Only genes with mean scores of ≥0.1 for at least 4 cell types are included. T cells and NK cells are strongly associated with JAV-Immuno, whereas genes in the selected CCMs display more diverse associations, including with endothelial cells, fibroblasts, B cells, and monocytes, as well as NK and T cells. Comparison of Gene Ontology biological processes associated with JAV-Immuno (C) and the selected CCM set (D). JAV-Immuno displays strong connections with T-cell processes, whereas selected CCM genes are associated with more diverse processes. CCM, chromatin conformation marker; NK, natural killer.
Figure 2. Cell types and functions of genes covered by the selected EpiSwitch CCM. The heat maps display aggregated associations between genes (rows) and cell types (columns in A,B) or functions (columns in C,D) across public data represented in the Data4Cure CURIE knowledge graph. The association score summarizes data and literature evidence from publicly available datasets and publications linking biomedical entities. Hierarchical clustering was applied to associations to generate the heat map. Association between cell types and genes in the JAV-Immuno signature (A) and the selected CCM set (B). Only genes with mean scores of ≥0.1 for at least 4 cell types are included. T cells and NK cells are strongly associated with JAV-Immuno, whereas genes in the selected CCMs display more diverse associations, including with endothelial cells, fibroblasts, B cells, and monocytes, as well as NK and T cells. Comparison of Gene Ontology biological processes associated with JAV-Immuno (C) and the selected CCM set (D). JAV-Immuno displays strong connections with T-cell processes, whereas selected CCM genes are associated with more diverse processes. CCM, chromatin conformation marker; NK, natural killer.
Cancers 17 02332 g002aCancers 17 02332 g002bCancers 17 02332 g002cCancers 17 02332 g002d
Figure 3. Spatial transcriptomic analysis in bladder cancer sections of marker set genes compared with JAV-Immuno genes. (A). (i). Hematoxylin and eosin stain of an index bladder cancer section (A1-1) showing the position of lymphoid aggregates annotated by a pathologist (labeled as 1 and 2). (ii). Spatial indexing transcriptomic data (Visium) from the same specimen processed by BayesSpace. Eight regions identified by BayesSpace were manually merged into 4 functional regions and annotated by majority cell type and histology. (B). (i). The A1-1 bladder specimen was annotated according to average expression of a published TLS signature [36] per spot. (ii). Average expression level of the TLS signature over the 4 main regions. (iii). Heat map depicting individual gene expression across the 4 regions. Color represents aggregation by means of values normalized by z-score. Note that most genes in the signature are expressed predominantly in the lymphoid aggregate region. (C). (i). A1-1 specimen annotated according to average expression of JAV-Immuno per spot. (ii). Average expression level of JAV-Immuno over the 4 main regions. (iii). Heat map depicting individual gene expression across the 4 regions. Note that most genes in the JAV-Immuno signature are also expressed predominantly in the lymphoid aggregate region. (iv). Concordance between JAV-Immuno genes localized to lymphoid aggregates in the A1-1 bladder specimen and TLS-high regions in the bladder/renal/breast cancer collection. (D). (i). Average expression level of the CCM model genes over the 4 regions. (ii). Heat map depicting individual gene expression across clusters based on distribution of expression between regions. Approximately half of the marker-set genes are expressed in lymphoid aggregates. (iii). The A1-1 bladder specimen annotated according to average expression of the CCM model genes per spot. Separate spatial images are shown for the epithelial and lymphoid regions. (iv). Concordance between genes localized to lymphoid aggregates in the A1-1 bladder specimen and TLS-high regions in the bladder/renal/breast cancer collection. CCM, chromatin conformation marker; TLS, tertiary lymphoid structures.
Figure 3. Spatial transcriptomic analysis in bladder cancer sections of marker set genes compared with JAV-Immuno genes. (A). (i). Hematoxylin and eosin stain of an index bladder cancer section (A1-1) showing the position of lymphoid aggregates annotated by a pathologist (labeled as 1 and 2). (ii). Spatial indexing transcriptomic data (Visium) from the same specimen processed by BayesSpace. Eight regions identified by BayesSpace were manually merged into 4 functional regions and annotated by majority cell type and histology. (B). (i). The A1-1 bladder specimen was annotated according to average expression of a published TLS signature [36] per spot. (ii). Average expression level of the TLS signature over the 4 main regions. (iii). Heat map depicting individual gene expression across the 4 regions. Color represents aggregation by means of values normalized by z-score. Note that most genes in the signature are expressed predominantly in the lymphoid aggregate region. (C). (i). A1-1 specimen annotated according to average expression of JAV-Immuno per spot. (ii). Average expression level of JAV-Immuno over the 4 main regions. (iii). Heat map depicting individual gene expression across the 4 regions. Note that most genes in the JAV-Immuno signature are also expressed predominantly in the lymphoid aggregate region. (iv). Concordance between JAV-Immuno genes localized to lymphoid aggregates in the A1-1 bladder specimen and TLS-high regions in the bladder/renal/breast cancer collection. (D). (i). Average expression level of the CCM model genes over the 4 regions. (ii). Heat map depicting individual gene expression across clusters based on distribution of expression between regions. Approximately half of the marker-set genes are expressed in lymphoid aggregates. (iii). The A1-1 bladder specimen annotated according to average expression of the CCM model genes per spot. Separate spatial images are shown for the epithelial and lymphoid regions. (iv). Concordance between genes localized to lymphoid aggregates in the A1-1 bladder specimen and TLS-high regions in the bladder/renal/breast cancer collection. CCM, chromatin conformation marker; TLS, tertiary lymphoid structures.
Cancers 17 02332 g003aCancers 17 02332 g003bCancers 17 02332 g003c
Figure 4. Modulation of interactions between TMB and OS by POU2F2. (A). Three-way interactions between POU2F2 expression in tumor, TMB, and treatment on OS outcome. Continuous measures of TMB and gene expression (log2 TPM) were used in the analysis. Genes that were expressed in ≤50% of samples or had a coefficient of variation of ≤5 for expression were excluded from the analysis. The estimated coefficients for the 3-way interaction and the p-value on the y-axis were calculated based on a Cox model containing the gene*TMB*treatment interaction term and all the related lower-order terms. Note that POU2F2 showed the strongest monogenic effect. (B). Presence of the blood marker encompassing POU2F2 is associated with reduced expression of POU2F2, downstream target genes, and gene expression signatures characteristic of TLS. OS, overall survival; TLS, tertiary lymphoid structures; TMB, tumor mutation burden; TPM, transcript count per million [50,51].
Figure 4. Modulation of interactions between TMB and OS by POU2F2. (A). Three-way interactions between POU2F2 expression in tumor, TMB, and treatment on OS outcome. Continuous measures of TMB and gene expression (log2 TPM) were used in the analysis. Genes that were expressed in ≤50% of samples or had a coefficient of variation of ≤5 for expression were excluded from the analysis. The estimated coefficients for the 3-way interaction and the p-value on the y-axis were calculated based on a Cox model containing the gene*TMB*treatment interaction term and all the related lower-order terms. Note that POU2F2 showed the strongest monogenic effect. (B). Presence of the blood marker encompassing POU2F2 is associated with reduced expression of POU2F2, downstream target genes, and gene expression signatures characteristic of TLS. OS, overall survival; TLS, tertiary lymphoid structures; TMB, tumor mutation burden; TPM, transcript count per million [50,51].
Cancers 17 02332 g004
Table 1. Compiled relationship between genes in the selected CCM set and JAV-Immuno, or entities in the CURIE knowledge graph. Values reported in the cell type, immune compartment, and immune process columns represent aggregated association scores between the individual genes and selected entities in the Data4Cure CURIE Knowledge Graph. The association score summarizes data and literature evidence from publicly available datasets and publications linking biomedical entities. CCM, chromatin conformation marker; MHC, major histocompatibility complex.
Table 1. Compiled relationship between genes in the selected CCM set and JAV-Immuno, or entities in the CURIE knowledge graph. Values reported in the cell type, immune compartment, and immune process columns represent aggregated association scores between the individual genes and selected entities in the Data4Cure CURIE Knowledge Graph. The association score summarizes data and literature evidence from publicly available datasets and publications linking biomedical entities. CCM, chromatin conformation marker; MHC, major histocompatibility complex.
Association Between Tumor Gene Expression and JAV-Immuno ScoreCell TypeImmune CompartmentImmune Process
GeneGene DescriptionStatistic Valuep-Valueq-ValueEndothelial CellFibroblastB CellMonocyteNatural Killer CellDendritic CellCytotoxic T CellLymph NodeGerminal CenterMHC Class I Antigen PresentationMHC Class II Antigen PresentationResponse to Stress
POU2F2POU class 2 homeobox 20.81419212.75 × 10−1354.96 × 10−1340.090110.24490.25580.25630.2280.21250.028450.092610.0993100.040720.004806
MBNL1Muscle blind-like splicing regulator 10.60739722.31 × 10−582.77 × 10−570.21850.2220.21860.23250.073440.049990.02845000.0086770.0085050.1479
ABI3BPABI family member 3 binding protein0.55176242.09 × 10−461.88 × 10−450.2120.24350.22360.19640.18890.19740.028320.073820000
VPS13CVacuolar protein sorting 13 homolog C0.45954616.43 × 10−314.63 × 10−300.074220.18060.050650.065940.064240.057330.0221300000
SUCNR1Succinate receptor 10.43907134.48 × 10−282.69 × 10−270.21470.21180.19850.21080.20920.2080.150.0923900.0034510.007120.0858
RSAD2Radical S-adenosyl methionine domain containing 20.32679271.49 × 10−156.72 × 10−150.22660.21480.2340.21370.22340.2290.16520.092740.0786400.061170.1613
CMPK2Cytidine/uridine monophosphate kinase 20.27872441.47 × 10−115.89 × 10−110.20660.21940.20990.22430.18650.20630.14370.078980.0681200.04990.08855
PI4KAPhosphatidylinositol 4-kinase alpha0.1527420.0002650.0009540.21010.20870.21860.19670.1970.047040.02591000.0160300
RNF144ARing finger protein 144A0.14716350.0004440.001450.094490.20080.18470.16240.064010.14950.089420.070850.01967000
CPEB1Cytoplasmic polyadenylation element binding protein 10.14581140.0005020.00150.18940.20840.18930.18630.072790.05963000000.01358
NPY4RNeuropeptide Y receptor Y40.12017680.004190.009440.040410.14010.12730.12080.13180.034980000.00338400
ZNF573Zinc finger protein 5730.11568410.005860.01130.065590.11730.14370.15060.12120.094010.049080.0224600.00574400
SNAP29Synaptosome-associated protein 290.11542070.005980.01130.20490.19190.060760.068290.064070.041740.02845000.0068510.0053670.1535
ZNF781Zinc finger protein 7810.11249340.007390.01210.10240.08650.12350.086210.090410.043210.0053670.0485800.0039860.0026240.000162
SLC38A7Solute carrier family 38 member 70.09787010.01990.02860.19760.057220.1920.061420.052570.035220.0227700000.08463
DEDD2Death effector domain containing 20.09665330.02150.02970.081560.17770.17560.16430.15150.1460.10520.06802000.020470.06925
C2CD4BC2 calcium-dependent domain containing 4B0.08260850.04950.0660.21620.17750.1540.14310.13670.057370.015320.01780000
TMEM14ETransmembrane protein 14E, pseudogene0.06789540.1070.1370.034180.02320.03380.031110.045440.022710.00431200000
MMP16Matrix metallopeptidase 160.06395160.1290.160.21340.25230.20560.20380.20850.065260.0079030.092340000
LZTR1Leucine zipper-like transcription regulator 10.05506830.1910.2220.20210.19650.062150.17140.20340.031790.013180.0817900.0072570.0089010.006873
RPS17Ribosomal protein S170.01655150.6940.7570.20790.1830.20760.22690.20220.1790.1641000.034280.073470.1058
CNBD1Cyclic nucleotide binding domain containing 10.00405820.9230.9230.13550.11140.11510.11640.10730.095810.017410.0478200.0032080.0032640.00432
C2CD4AC2 calcium-dependent domain containing 4A−0.0047890.9090.9230.1710.15870.13310.12130.11560.056840.0048670.04788000.0039820.08443
DCAF4L2DDB1 and CUL4-associated factor 4-like 2−0.0087940.8350.8840.063240.037360.066590.098150.12820.0573500.0601600.005280.010130.00442
ZNF526Zinc finger protein 526−0.0486780.2480.2790.12910.14370.075710.065730.040320.030470.00480300000
CNOT1CCR4-NOT transcription complex subunit 1−0.0551190.190.2220.06780.210.20230.20260.034540.17210.01716000.018480.0030420.09112
ZFP30ZFP30 zinc finger protein−0.0989220.01860.02790.12190.12270.11670.13080.12570.073360.0225300000
ZNF607Zinc finger protein 607−0.1105540.008480.01330.046030.04710.069890.082440.10020.037780.03910.0227600.0044090.0033070.01889
GPRIN2G protein-regulated inducer of neurite outgrowth 2−0.1125850.007340.01210.060670.14650.15160.034620.15170.048010.002130.058960.0201300.019730.00166
TFGTrafficking from ER to golgi regulator−0.1146730.006310.01140.20940.18790.22050.20670.061150.028260.028450.0895200.0031580.004230.005551
SERPIND1Serpin family D member 1−0.1160830.005690.01130.21450.19940.1890.19050.059450.167200.09033000.0041570.1498
CRKLCRK-like proto-oncogene, adaptor protein−0.1303290.001890.004540.20090.20360.22490.19690.20290.16670.02420.0940100.023390.021360.02151
AIFM3Apoptosis-inducing factor mitochondria-associated 3−0.1389360.0009190.002360.190.17290.050.069310.06110.055620.0061260.0888700.01070.0044030.1262
ANXA8L1Annexin A8-like 1−0.144150.0005820.001610.14660.19220.13920.12120.13540.12760.026920.06070000
GOT2Glutamic-oxaloacetic transaminase 2−0.3812915.01 × 10−212.58 × 10−200.23460.22960.21880.21720.071410.19930.17280.091150.0747200.0063620.09641
Table 2. Associations between POU2F2 marker and OS.
Table 2. Associations between POU2F2 marker and OS.
TreatmentTMBPOU2F2 MarkerNo. of Patients (N = 457)No. of EventsOS, Median (95% CI), MonthsHR (POU2F2 Marker Absent vs. Present) (95% CI)p-Value
Avelumab plus BSC≤MedianAbsent221036.99 (18.17–NE)0.46 (0.240–0.894)0.0218
Avelumab plus BSC≤MedianPresent1128217.77 (13.34–22.34)
Avelumab plus BSC>MedianAbsent11719.25 (17.81–NE)1.38 (0.623–3.048)0.4281
Avelumab plus BSC>MedianPresent984835.12 (26.05–NE)
BSC alone≤MedianAbsent181313.68 (8.8–NE)1.14 (0.622–2.071)0.6788
BSC alone≤MedianPresent896016.07 (10.25–24.18)
BSC alone>MedianAbsent211414.78 (11.5–NE)1.14 (0.635–2.044)0.6608
BSC alone>MedianPresent865817.81 (13.54–26.64)
BSC, best supportive care; HR, hazard ratio; NE, not estimable; OS, overall survival; TMB, tumor mutation burden.
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

Powles, T.; Sridhar, S.S.; Bellmunt, J.; Sternberg, C.N.; Grivas, P.; Hunter, E.; Salter, M.; Powell, R.; Dring, A.; Green, J.; et al. Blood-Epigenetic Biomarker Associations with Tumor Immunophenotype in Patients with Urothelial Carcinoma from JAVELIN Bladder 100. Cancers 2025, 17, 2332. https://doi.org/10.3390/cancers17142332

AMA Style

Powles T, Sridhar SS, Bellmunt J, Sternberg CN, Grivas P, Hunter E, Salter M, Powell R, Dring A, Green J, et al. Blood-Epigenetic Biomarker Associations with Tumor Immunophenotype in Patients with Urothelial Carcinoma from JAVELIN Bladder 100. Cancers. 2025; 17(14):2332. https://doi.org/10.3390/cancers17142332

Chicago/Turabian Style

Powles, Thomas, Srikala S. Sridhar, Joaquim Bellmunt, Cora N. Sternberg, Petros Grivas, Ewan Hunter, Matthew Salter, Ryan Powell, Ann Dring, Jayne Green, and et al. 2025. "Blood-Epigenetic Biomarker Associations with Tumor Immunophenotype in Patients with Urothelial Carcinoma from JAVELIN Bladder 100" Cancers 17, no. 14: 2332. https://doi.org/10.3390/cancers17142332

APA Style

Powles, T., Sridhar, S. S., Bellmunt, J., Sternberg, C. N., Grivas, P., Hunter, E., Salter, M., Powell, R., Dring, A., Green, J., Akoulitchev, A., Ronen, R., Dutkowski, J., Amezquita, R., Huang, C.-H., Fernandez, D., Nameki, R., Ching, K. A., Pu, J., ... Davis, C. B. (2025). Blood-Epigenetic Biomarker Associations with Tumor Immunophenotype in Patients with Urothelial Carcinoma from JAVELIN Bladder 100. Cancers, 17(14), 2332. https://doi.org/10.3390/cancers17142332

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