Reactive stroma as a prognostic biomarker of metastasis in patients with breast cancer: integration of histopathology and transcriptomic profiling
A
A
Daniela P. Barrera 1,2
Muriel A. Núñez 1
Valentina Cerda 1
I. 1
J. Sebastián Contreras-Riquelme 1
Jenny Henríquez 1
Guillermo Carrasco 1
Alejandra Pereira 1
Vania Figueroa 1
Verónica Toledo 4
Badir Chahuan 4
Jorge Sapunar-Zenteno 4,5
Ximena Rodríguez 6
Daniel Moreno 3,7
José Tomás Larach 3,7
Benjamín Prieto 1
Patricia García 1,8
Leonor Moyano 9
José Peña 3,7
Javier Cerda-Infante
Ph.D.
1,3,10✉
Phone+56971408741 Email
Environ 10
Pontificia Universidad 10
1 Environ Santiago Chile
2 Facultad de Medicina y Ciencia Universidad San Sebastián Santiago Chile
3 Pontificia Universidad Católica de Chile Santiago Chile
4 Instituto Oncológico Fundación Arturo López Pérez Santiago Chile
5 Centro de Excelencia CIGES, Facultad de Medicina Universidad de la Frontera Temuco Chile
6 Hospital San José Santiago Chile
7 Red Salud UC-Christus Santiago Chile
8 Department of Pathology, School of Medicine Center for Cancer Prevention and Control (CECAN), Pontificia Universidad Católica de Chile Santiago Chile
9 Instituto Nacional del Cáncer Santiago Chile
10 Católica de Chile Santiago Chile
Daniela P. Barrera 1,2, Muriel A. Núñez1, Valentina Cerda I.1,2, J. Sebastián Contreras-Riquelme1, Jenny Henríquez1, Guillermo Carrasco1, Alejandra Pereira1, Vania Figueroa1, Verónica Toledo4, Badir Chahuan4, Jorge Sapunar-Zenteno4,5, Ximena Rodríguez6, Daniel Moreno3,7, José Tomás Larach3,7, Benjamín Prieto1, Patricia García1,8, Leonor Moyano9, José Peña3,7 and Javier Cerda-Infante1,3*
1 Environ, Santiago, Chile
2 Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
3 Pontificia Universidad Católica de Chile, Santiago, Chile
4 Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
5 Centro de Excelencia CIGES, Facultad de Medicina, Universidad de la Frontera, Temuco, Chile
6 Hospital San José, Santiago, Chile
7 Red Salud UC-Christus, Santiago, Chile
8 Department of Pathology, School of Medicine; Center for Cancer Prevention and Control (CECAN), Pontificia Universidad Católica de Chile, Santiago, Chile
9 Instituto Nacional del Cáncer, Santiago, Chile
Corresponding Author: Javier Cerda-Infante, Ph.D. Environ, Pontificia Universidad Católica de Chile, Santiago, Chile
Tel.: +56971408741, E-mail: javier@environ.bio
Running title
Reactive Stroma as a Metastasis Biomarker
ABSTRACT
Background
The tumor stroma plays a pivotal role in cancer progression, but its prognostic relevance remains underexplored in clinical settings. This study evaluated whether histologically defined reactive stroma, a fibrotic and collagen-rich microenvironment, serves as a prognostic marker for overall survival (OS) and metastasis-free survival (MFS) in patients with breast cancer, aiming to establish it as a clinically actionable biomarker.
Methods
A
We retrospectively analyzed 182 formalin-fixed, paraffin-embedded (FFPE) breast cancer samples from patients diagnosed between 2006 and 2020 at three Chilean institutions. Stromal content was digitally quantified on H&E- and Masson’s trichrome-stained sections using QuPath software and validated by expert pathologists. Optimal cutoffs for total and reactive stroma were identified via maximally selected rank statistics. Survival outcomes were assessed using Kaplan‒Meier and Cox regression models adjusted for clinical covariates. A subset of samples was subjected to RNA sequencing to explore transcriptomic programs associated with stromal phenotypes.
Results
A lower total stromal content (< 71.8%) was significantly associated with poor OS in the univariate analysis but not in the multivariate model. Conversely, a high reactive stroma content (> 53.2%) was independently associated with poor MFS (HR = 3.76, p < 0.001), regardless of the molecular subtype or other clinicopathological variables. Transcriptomic profiling revealed the overexpression of ECM-related and protumorigenic genes (e.g., FN1, OLR1, and EDN2) and enrichment of genes related to collagen remodeling and the TGF-β signaling pathway in reactive stroma-high tumors.
Conclusions
Reactive stroma, defined through routine histology and digital pathology, is a robust and independent prognostic marker for metastasis in patients with breast cancer. Its clinical integration could enhance risk stratification and support novel stroma-targeted therapeutic strategies.
Keywords:
Reactive Stroma
Tumor Microenvironment
Metastasis
Digital Pathology
Cancer Prognosis
A
1. INTRODUCTION
Breast cancer (BC) is one of the leading causes of cancer-related mortality worldwide, with distant metastasis accounting for most deaths [1]. In 2022, GLOBOCAN reported 2.3 million new cases of BC and more than 685,000 related deaths globally. Despite therapeutic advances, both incidence and mortality continue to rise, largely owing to metastatic progression, even in patients treated with curative intent [2].
Current prognostic biomarkers in BC are primarily based on histopathological and molecular features, such as hormone receptor status, HER2 expression, tumor size, and lymph node involvement [3, 4]. Additional markers, such as the Ki-67 proliferation index, support treatment decisions but fail to accurately prognosticate the risk of metastasis [5, 6]. This represents a critical gap in the clinical management of patients with BC, especially at the time of diagnosis.
The tumor microenvironment (TME), particularly its stromal compartment, plays an active role in tumor progression [7, 8]. The stroma consists of cancer-associated fibroblasts (CAF), immune cells, and extracellular matrix (ECM) proteins, which dynamically interact with tumor cells [916]. CAF are key mediators of TME remodeling and promote invasion and metastasis through excessive ECM deposition and structural alterations, leading to the development of reactive stroma [14, 1719].
The reactive stroma is defined as a fibrotic and proinflammatory microenvironment that emerges early during carcinogenesis. It is characterized by the activation of fibroblasts into a myofibroblast phenotype, with increased synthesis of collagen type I, tenascin-C, fibronectin, and other ECM proteins, as well as the expression of mesenchymal markers such as vimentin and FAP [20, 21]. This response resembles chronic wound repair and co-evolves with tumor progression, promoting angiogenesis, tissue remodeling, and cancer cell invasion. Despite growing evidence of its biological and clinical relevance, the prognostic role of reactive stroma in breast cancer remains poorly understood. Moreover, the cellular and molecular processes triggered by this stromal remodeling response and how they influence tumor behavior are still largely unknown.
To address this gap, we combined the digital histopathology of FFPE breast cancer samples with bulk transcriptomic profiling. This strategy allowed us to evaluate the association between stromal composition and metastatic progression in a clinically relevant and scalable manner [11, 15, 20, 22].
This study aimed to determine whether reactive stroma can serve as a metastasis-specific prognostic biomarker in BC. We digitally quantified total and reactive stromal contents in FFPE tumor samples from a retrospective cohort of 182 patients using hematoxylin‒eosin (H&E)- and Masson’s trichrome-stained slides analyzed with QuPath software. To ensure methodological robustness, automated quantification was validated against manual annotations by expert pathologists. We then correlated stromal characteristics with overall survival (OS) and metastasis-free survival (MFS) and analyzed the expression of genes previously associated with CAF and ECM-related pathways to explore their relationship with stromal content and prognostic value. By integrating digital pathology with transcriptomic analysis of routine clinical samples, this study proposes a clinically feasible approach to refine metastatic risk assessment and guide personalized treatment in patients with breast cancer.
2. MATERIAL AND METHODS
Table 1
Clinicopathological characteristics of the breast cancer cohort (n = 182)
Characteristic
n (%)
Age
 
Mean (range)
CALCULAR
< 55
97 (53.3%)
55–65
52 (28.6%)
> 65
31 (17.0%)
Unknown
2 (1.1%)
Menopausal status
 
Premenopausal
21 (11.5%)
Postmenopausal
44 (24.2%)
Unknown
117 (64.3%)
ER status
 
Positive
129 (70.9%)
Negative
52 (28.6%)
Unknown
1 (0.5%)
PR status
 
Positive
112 (61.5%)
Negative
65 (35.7%)
Unknown
5 (2.7%)
HER2 status
 
Positive
49 (26.9%)
Negative
98 (53.8%)
Unknown
35 (19.2%)
Histological grade
 
Well differentiated
19 (19.4%)
Moderately differentiated
51 (28.0%)
Poorly differentiated
50 (27.5%)
Unknown
62 (34.1%)
Tumor size (pT)
 
pT0
12 (6.6%)
pT1
58 (31.9%)
pT2
26 (14.3%)
pT3
6 (3.3%)
pT4
3 (1.6%)
Unknown
77 (42.3%)
Nodal Status (pN)
 
Positive
32 (17.6%)
Negative
71 (39.0%)
Unknown
79 (43.3%)
Total Stromal Content and Association with Survival
To evaluate whether the amount of total stroma present in tumor tissue was associated with clinical outcomes, the percentage of stromal area was quantified in FFPE sections stained with H&E via QuPath image analysis software. A supervised learning approach was applied in which manual annotations were used to train a pixel classifier capable of discriminating between stromal, tumor, and non-stromal regions. This enabled automated segmentation and precise quantification of each tissue compartment. QuPath uses pixel-level classification on the basis of specific visual characteristics defined by initial annotations. This process allowed the identification and delineation of relevant tissue components, including the stromal area (green) and tumor or other structures (red), in each digital sample (Fig. 1A). The total stromal content was calculated as the ratio of the stromal area to the total tissue area (Fig. 1A–B).
The automated method was validated against manual segmentation performed by two independent pathologists in a subset of 17 cases, which showed high concordance (r = 0.902; p < 0.001) (Fig. 1C).
Table 2
Multivariate Cox regression analysis of reactive stromal content. Multivariate Cox regression analysis for overall survival (OS) and metastasis-free survival (MFS) was performed based on the reactive stromal content. The models were adjusted for age, pT stage, pN stage, molecular subtype and histological grade. HR and 95% CI are presented. Reference values are indicated where applicable.
   
Overall Survival
Metastasis-Free Survival
Variables
 
HR (95% CI)
p value
HR (95% CI)
p value
Age
         
 
< 55 years
Reference value
 
Reference value
 
 
55–65 years
2.6 (1.18–5.72)
0.018
1.22 (0.6–2.49)
0.577
 
> 65 years
2.87 (1.07–7.75)
0.037
4.61 (1.45–14.69)
0.010
ER Status
         
 
Positive
Reference value
 
Reference value
 
 
Negative
2.55 (0.74–8.83)
0.140
1.46 (0.39–5.41)
0.575
PR Status
         
 
Positive
Reference value
 
Reference value
 
 
Negative
1.14 (0.33–3.98)
0.838
1.27 (0.34–4.71)
0.721
Reactive Stroma
         
 
High
2.44 (0.93–6.4)
0.069
3.76 (1.91–7.39)
< 0.001
 
Low
Reference value
 
Reference value
 
Ethics approval and consent to participate
A
The study was approved by the Research Ethics Committees of the Fundación Arturo López Pérez (FALP; breast cancer protocol ID: 2022-0232-RES-CRC-MUL), Instituto Nacional del Cancer (INC; project number CRI20050, under the framework of UC project 201016011, as the INC does not have its own ethics committee), and Red Salud UC-CHRISTUS (project ID 201016011).
A
A
A waiver of informed consent was granted by all committees, as the study fulfilled the requirements for minimal risk, impracticability, and the projection of participants’ rights and welfare.
A
All procedures complied with relevant institutional guidelines and regulations.
Study design and patient characteristics
This retrospective study aimed to evaluate the prognostic value of the stromal content and reactive stroma in patients with breast cancer. Patients were selected from the pathology archives of FALP, INC and UC-CHRISTUS. The inclusion criteria were histologically confirmed invasive breast carcinoma, availability of formalin-fixed paraffin-embedded tissue, complete clinical follow-up, and no neoadjuvant therapy prior to biopsy or surgery. A total of 182 eligible patients were enrolled. Follow-up time was defined from the date of diagnosis to the date of last follow-up or death. The clinical data included age, tumor size (pT), lymph node status (pN), molecular subtype (ER, PR, HER2), and histological grade.
For each case, one representative tumor block was selected by a certified pathologist. Sections (4 µm) were stained with hematoxylin and eosin (H&E) and Masson's trichrome. In total, 182 H&E-stained and 146 Masson-stained slides were obtained and scanned using the Aperio Digital Pathology System (Aperio Technologies Inc., Vista, CA, USA) at a spatial resolution of 0.47 µm per pixel and a magnification of ×20.
The total and reactive stroma were quantified using open-source QuPath software.
Digital image analysis was performed using QuPath (v.0.5.0) [23]. The analysis workflow included the following steps: (1) import of .scn images and assignment of image type as "brightfield H&E" or "other" (Masson); (2) manual annotation of tissue compartments, including tumor, stroma or reactive stroma, immune infiltrate, red blood cells, necrosis and background; (3) training of a Random Trees (RTrees) pixel classifier to automate tissue segmentation; and (4) quantification of stromal areas as follows: total stroma was calculated as the percentage of stromal area identified in H&E-stained slides relative to the total tissue area. Reactive stroma was quantified as the area corresponding to collagen deposition in Masson's trichrome staining slides, expressed as a percentage of the stromal area previously defined on the corresponding H&E slide.
To validate the reproducibility of this approach, 60 randomly selected cases were independently reviewed by two experienced pathologists. The percentages of total and reactive stroma were manually estimated and compared to the QuPath-based quantification. Agreement between manual and automated measurements was assessed via Pearson’s correlation coefficient. All the statistical analyses were conducted via R (v4.4.0).
Stromal stratification and Kaplan‒Meier survival analysis
Optimal cutoff thresholds for total and reactive stromal percentages were identified via the maximally selected rank statistics method implemented in the Maxstat R package (v0.7–25). Cutoff points were determined to maximize survival differences between groups. Kaplan‒Meier curves were generated via the survival (v3.7–0) and ggplot2 (v3.5.1) packages, with p values adjusted via the condMC method.
RNA sequencing, gene expression analysis and stromal association
Total RNA was extracted from ten 10 µm-thick FFPE sections per sample via the PureLink™ FFPE Total RNA Isolation Kit (Invitrogen, Carlsbad, CA, USA), followed by DNase I treatment to eliminate genomic DNA contamination. The RNA integrity and concentration were assessed via an Agilent TapeStation and an Epoch Microplate Spectrophotometer. RNA sequencing was performed on the Illumina NovaSeq 6000 platform with paired-end 150 bp reads, targeting an average depth of ~ 30 million reads per sample.
The sequencing reads were aligned to the GRCh38 human reference genome via STAR aligner (v2.7.11b) [24], and gene-level expression was quantified via FeatureCounts (V2.1.0) [25]. Gene expression values were normalized, and differential expression was subsequently assessed with DESeq2 (V1.46.0) [26]. Genes showing an adjusted p value ≤ 0.05 and an absolute log₂(fold change) > 1 were considered differentially expressed genes (DEGs).
Downstream analyses were performed as follows: heatmaps and volcano plots for DEGs were generated with the pheatmap (v1.0.12) and EnhancedVolcano (v1.22.0) packages, whereas gene set enrichment analysis (GSEA) was computed through clusterProfiler (v4.14.6) and visualized via gseaplot2 from the enrichplot package (v1.26.6) [2729]. The tumor microenvironment cell populations were quantified with an MCP-counter (v 1.2.0) [30], which was applied to the DESeq2 variance-stabilized expression matrix and specified Ensembl gene identifiers as the feature type. To investigate the relationship between gene expression and stromal content, expression values were compared between the Stroma-High and Stroma-Low groups via Student’s t-test (ggpubr). Pearson’s correlation analysis (cor.test, dplyr) was used to assess associations between gene expression and the percentage of the stromal area. Finally, KEGG and Reactome pathway enrichment analyses of the significant DEGs were conducted via the clusterProfiler and ReactomePA (v1.50.0) [28, 31] packages.
Statistical analysis
Survival analyses for overall survival (OS) and metastasis-free survival (MFS) were performed via Kaplan‒Meier estimators, and survival differences between groups were evaluated via the log-rank test. Univariate and multivariate Cox proportional hazards regression models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for the effects of stromal variables and clinical covariates on survival outcomes. The multivariate models included the following covariates: age, tumor size (pT), lymph node status (pN), molecular subtype (ER, PR, HER2) and histological grade. Comparisons of clinicopathological characteristics between stromal groups were conducted via Pearson’s χ² test or Fisher’s exact test, as appropriate.
To analyze gene expression differences between stromal groups, gene-wise comparisons were conducted using Student’s t-test or the Wilcoxon rank-sum test with the Shapiro‒Wilk normality test. Multiple testing correction was applied via the Benjamini‒Hochberg method (false discovery rate, FDR). Violin and box plots were generated to visualize expression distributions across groups, and adjusted p-values were annotated accordingly. For the selected genes, the correlation between gene expression and stromal percentage was assessed separately within the Stroma-High and Stroma-Low groups via Pearson’s correlation test. Scatter plots with linear regression lines and annotated correlation coefficients (r) and p-values were generated for each condition.
All statistical analyses and visualizations were performed via R (v4.4.0) with packages including ggplot2, dplyr, tidyr, ggsignif, ggpubr, and GraphPad Prism (v10.0.2, GraphPad Software, San Diego, CA, USA).
3.
RESULTS
Clinical characteristics of the cohort
To characterize the patient cohort prior to molecular and clinical analyses, 182 patients with invasive breast cancer were included, with a median follow-up of 60 months. All patients met the following inclusion criteria: available FFPE tumor samples, complete clinical data, and no history of neoadjuvant therapy prior to biopsy. Patients with a previous diagnosis of cancer or hereditary cancer syndromes were excluded from the study.
H&E-stained sections were obtained for all 182 patients, with overall survival (OS) data available for all patients and metastasis-free survival (MFS) data available for 134 patients. A total of 146 cases were analyzed via Masson’s trichrome staining, with complete OS and MFS data available for 85 patients. Clinicopathological characteristics—including age, tumor size (pT), nodal status (pN), molecular subtypes (ER, PR, HER2), histological grade, and adjuvant treatment—are summarized in Table 1.
A
Fig. 1
Quantification of the Total Stroma Percentage in BC. (A) Flowchart illustrating the methodology used to quantify the total stroma percentage in FFPE breast tumor tissue samples. The samples were stained with H&E and analyzed via QuPath software to identify the stroma and tumor areas, with validation performed by a pathologist. (B) Representative images of H&E staining (top) and corresponding QuPath predictions (bottom) in samples with low, medium, and high total stroma percentages. (C) Correlation between total stroma quantification by QuPath and pathologist validation (n = 17).
Maximally selected rank statistical analysis identified an optimal cutoff of 71.8% total stromal content for overall survival (OS) stratification (Fig. 2A). In the study cohort, the total stromal content ranged from 23.9–96.7%, with a mean of 67.8% and a standard deviation of 16.5%. On the basis of this threshold, 102 patients were classified as having low total stroma, and 80 patients were classified as having high total stroma, corresponding to 56.0% and 44.0% of the cohort, respectively.
Click here to Correct
Patients in the low total stroma group (≤ 71.8%) exhibited significantly shorter OS in the univariate analysis (HR = 3.49; 95% CI: 1.84–6.62; p < 0.001) (Fig. 2B; Supplementary Table 1). However, this association was no longer statistically significant after adjusting for age, tumor size (pT), nodal status (pN), molecular subtype, or histological grade (HR = 2.49; 95% CI: 0.60–10.35; p = 0.209) (Supplementary Table 2), indicating that its prognostic effect may be influenced by other clinicopathological variables. Given that distant metastasis is the leading cause of breast cancer-related mortality, we examined whether the total stromal content was associated with metastasis-free survival (MFS).
For MFS, the maximally selected rank statistical analysis identified an optimal cutoff of 80.8% total stroma content (Fig. 2C). In this subset, the total stromal content ranged from 23.9–96.7%, with a mean of 69.9% and a standard deviation of 16.3%. On the basis of this threshold, 96 patients were classified as having low total stroma (≤ 80.8%), and 38 were classified as having high total stroma (> 80.8%), accounting for 71.6% and 28.4% of the MFS cohort, respectively.
Patients in the low total stroma group exhibited a significantly shorter MFS in the univariate analysis (HR = 4.11; 95% CI: 1.62–10.40; p < 0.001) (Fig. 2D, Supplementary Table 1), and this trend persisted in the multivariate model, although it did not reach statistical significance (HR = 4.08; 95% CI: 0.88–18.94; p = 0.073) (Supplementary Table 2).
Although the association between low total stromal content and poor prognosis was significant in the univariate analyses for both OS and MFS, it did not remain statistically significant in the multivariate models after adjusting for age, tumor size, nodal status, molecular subtype, and histological grade.
To determine whether the prognostic relevance of total stromal content might be influenced by molecular subtype, we analyzed its distribution across ER+/PR+, HER2+, and triple-negative tumors. Quantitative analysis of H&E-stained images via QuPath revealed no statistically significant differences in stromal content among subtypes (Fig. 2E–F), indicating that total stromal content is not associated with molecular subtype and constitutes a consistent feature of the tumor microenvironment across different biological contexts.
A
Fig. 2
Associations of total stromal content with clinical outcomes and molecular subtypes in patients with breast cancer. (A–B) Maximally selected rank statistics and Kaplan–Meier survival analysis for overall survival (OS) in patients with breast cancer stratified by total stromal content (n = 182). (A) Determination of the optimal cutoff value (71.8%) via maximally selected rank statistics. (B) Kaplan–Meier curve showing significantly shorter OS in patients with low stromal content (HR 3.49; 95% CI: 1.84–6.62; p < 0.001). (C–D) Analysis of metastasis-free survival (MFS) in a subset of patients (n = 134). (C) The optimal cutoff value for MFS was 80.8%. (D) Kaplan–Meier curve indicating significantly worse MFS in the Stroma-Low group (HR 4.11; 95% CI: 1.62–10.4; p < 0.001). (E) Representative H&E-stained images and corresponding QuPath-based segmentation of stromal and tumor areas in tumors classified as ER+/PR+, HER2+, or triple-negative breast cancer (TNBC). Stromal areas are highlighted in green and tumor regions in red. (F) Percentage of total stromal content across molecular subtypes (ER+/PR+, HER2+, and TNBC) quantified from H&E-stained sections via QuPath. No statistically significant differences were observed among the subtypes (ns, Kruskal–Wallis test).
Given the lack of significant associations in multivariate models and the observation that total stromal content does not vary across molecular subtypes, we investigated the biological nature of the stromal compartment itself. While total stroma provides a quantitative measure of tissue microarchitecture, it may not fully capture the functional activity of stromal cells involved in tumor progression. To address this, we evaluated the expression of genes commonly expressed by CAF and other stromal components. Specifically, we assessed the expression of VIM, PDGFRA, PDGFRB, and ACTA2, which are genes predominantly expressed by stromal cells, particularly CAF, and are involved in mesenchymal activation and extracellular matrix remodeling, via RNA sequencing of a representative subset of the same FFPE samples used for stromal quantification.
Among these genes, PDGFRA and PDGFRB were significantly upregulated in tumors classified as total stroma-high (p < 0.05) (Fig. 3A–D). Although VIM and ACTA2 were more highly expressed in the total stroma-high group, these differences were not statistically significant. Moreover, correlation analysis revealed that gene expression levels were strongly positively correlated with the stromal percentage in the total stroma-low group (Fig. 3E–H), suggesting a complex relationship between stromal abundance and transcriptional activity. In addition, MCP-counter analysis revealed higher fibroblast scores in tumors with high total stromal content (Fig. 3I), reflecting increased infiltration of stromal cell populations in these samples.
Although low total stromal content was significantly associated with poorer OS and MFS, these results indicate that the total stromal percentage does not necessarily reflect the abundance or activation state of CAF. Given that CAF play a pivotal role in tumor progression, we hypothesized that the total stromal content may fail to capture the functionally active stromal compartment. Therefore, we quantified the reactive stroma, defined as the fraction of stroma characterized by a densely remodeled ECM enriched in activated CAF with a myofibroblastic phenotype and high secretory activity of protumorigenic factors, including collagen types I and II. This component represents the biologically active portion of the tumor stroma and is directly involved in invasion, angiogenesis, immunosuppression, and therapeutic resistance. Its quantification enabled a more specific assessment of its prognostic relevance.
A
Fig. 3
Expression of CAF-Associated Genes and the Abundance of Tumor Microenvironment Cell Populations According to the Total Stromal Content in Breast Cancer. (A–D) Box plots showing the variance-stabilized expression levels of the CAF markers VIM (A), PDGFRA (B), PDGFRB (C), and ACTA2 (D) in tumors classified as Stroma-High and Stroma-Low based on the total stromal percentage. (E–H) Scatter plots illustrating the correlation between VST-normalized gene expression and total stromal area across samples. Pearson’s r and p-values are shown for each stromal group. (I) MCP-counter analysis showing the estimated abundance of tumor microenvironment cell populations in patients with high and low total stromal content.
Reactive Stroma and Its Impact on Breast Cancer Survival
To quantify the reactive stroma, we analyzed 146 Masson’s trichrome–stained samples, measuring the area corresponding to collagen I and III relative to the total stromal area previously identified in H&E images. This quantification was performed via QuPath with supervised pixel classification, allowing the identification of reactive stroma, tumor, and other tissue structures (Fig. 4A-B). The method was validated in a subset of 14 cases by comparison with manual annotations from expert pathologists, which revealed a significant correlation (r = 0.577; p < 0.05) (Fig. 4C).
A
Fig. 4
Quantification of Reactive Stroma in BC. (A) Flowchart illustrating the methodology used to quantify the percentage of reactive stroma relative to total stroma in FFPE breast tumor tissue samples. The samples were stained with Masson’s trichrome and analyzed via QuPath software to identify the tumor and reactive stroma areas, followed by clinical validation by a pathologist. (B) Representative images of Masson’s trichrome staining (top) and corresponding QuPath predictions (bottom) in samples with low, medium, and high percentages of reactive stroma. (C) Correlation between reactive stroma quantification by QuPath and validation by pathologists (n = 14).
Maximally selected rank statistical analysis identified optimal cutoffs of 42.5% and 53.2% reactive stromal content for OS and MFS, respectively (Figs. 5A and 5C). In the OS cohort (n = 146), the percentage of reactive stromal content ranged from 3.7–100%, with a mean of 55.2% and a standard deviation of 21.9%. Based on the 42.5% cutoff, 42 patients were classified as reactive stroma-low, and 104 patients as reactive stroma-high. For the MFS analysis, a subset of 85 patients from the same cohort for whom time to metastatic progression was available was included. In this subgroup, the reactive stromal content ranged from 14.5–100%, with a mean of 48.7% and a standard deviation of 21.0%. According to the 53.2% threshold, 58 patients were assigned to the Reactive Stroma-Low group, and 27 were assigned to the Reactive Stroma-High group. This stratification enabled a more specific evaluation of the prognostic impact of the biologically active stromal compartment, offering additional insights beyond total stromal quantification.
Click here to Correct
Click here to Correct
Click here to Correct
According to the univariate analysis, patients in the Reactive Stroma-High group tended to have shorter OS (HR = 2.51; 95% CI: 0.97–6.49; p = 0.058) (Fig. 5B, Supplementary Table 3) and significantly shorter MFS (HR = 3.75; 95% CI: 1.98–7.09; p < 0.01) (Fig. 5D, Supplementary Table 3). These findings support the hypothesis that CAF-enriched reactive stroma, rather than stromal quantity alone, is a biologically relevant factor in tumor progression.
We also evaluated whether the reactive stromal content varied among different molecular subtypes of breast cancer. No statistically significant differences were observed among the ER+/PR+, HER2+, and triple-negative breast cancer (TNBC) groups (Fig. 5E–F), supporting the notion that reactive stroma is a consistent and subtype-independent feature of the breast cancer TME.
Importantly, multivariate Cox regression analysis confirmed that high reactive stromal content remained an independent prognostic factor for MFS after adjusting for age, ER status, and PR status (HR = 3.76; 95% CI: 1.91–7.39; p < 0.001), whereas its association with OS showed a non-significant trend (HR = 2.44; 95% CI: 0.93–6.4; p = 0.069) (Figs. 5G–H, Table 2). These results provide robust evidence that the reactive stroma captures biologically and clinically meaningful features of the tumor microenvironment that are not reflected by conventional molecular markers. The strong and independent association with metastatic risk highlights its potential utility as a prognostic biomarker in breast cancer and supports the incorporation of stromal biology into future risk-stratification strategies.
A
Fig. 5
Associations of the Reactive Stromal Content with Clinical Outcomes, Molecular Subtypes, and Multivariate Prognostic Value in Breast Cancer. (A–B) Maximally selected rank statistics and Kaplan–Meier analysis for overall survival (OS) based on the percentage of reactive stroma (n = 146). (A) Optimal cutoff point identified at 42.5% reactive stroma. (B) Kaplan–Meier curve showing a trend toward worse OS in patients with highly reactive stroma (HR = 2.51; 95% CI: 0.97–6.49; p = 0.058). (C–D) Analysis of metastasis-free survival (MFS) in a subset of patients (n = 85). (C) Optimal cutoff point determined at 53.2% reactive stroma. (D) Kaplan–Meier curve demonstrating a significantly lower MFS in the reactive stroma-high group (HR = 3.75; 95% CI: 1.98–7.09; p < 0.01). (E) Representative Masson’s trichrome-stained images and corresponding QuPath segmentation of reactive stroma in ER+/PR+, HER2+, and triple-negative breast cancer (TNBC) tumors. (F) Quantitative comparison of the reactive stromal content across molecular subtypes revealed no significant differences, suggesting that the reactive stroma is a subtype-independent feature. (G–H) Forest plots of multivariate Cox regression analyses for OS (G) and MFS (H).
Transcriptomic Profile Associated with the Reactive Stromal Content
Given the strong prognostic value of histologically defined reactive stroma, we aimed to investigate whether this phenotype is associated with specific biological programs. To this end, RNA sequencing (RNA-Seq) was performed on 62 tumor samples with available FFPE material to assess their transcriptomic profile. From these, a representative subset of 12 tumors—previously analyzed histologically and selected on the basis of high or low reactive stromal content—was used for differential gene expression (DEG) analysis, followed by gene set enrichment analysis (GSEA) and functional annotation via the KEGG and Reactome databases, with the aim of identifying signaling pathways differentially regulated between tumors with high and low reactive stromal content.
To visualize overall transcriptomic differences, a heatmap was generated using representative differentially expressed genes between tumors stratified by reactive stromal content (Fig. 6A). A clear separation was observed between the high and low groups, indicating that distinct transcriptomic profiles are associated with the stromal phenotype. The volcano plot highlights differentially expressed genes between the two groups, with several showing high magnitude changes and statistical significance (adjusted p-value < 0.05; log2-fold change > |1|) (Fig. 6B). Among the genes upregulated in tumors with high reactive stromal content were FN1, OLR1, EDN2, and MSR1, which are involved in key processes such as extracellular matrix (ECM) remodeling, inflammatory signaling, and modulation of the tumor microenvironment—all mechanisms associated with tumor progression and metastasis.
Pathway enrichment analysis via the Reactome database revealed an overrepresentation of ECM-related and immune-associated processes, including collagen degradation, ECM activation, and ECM organization, in tumors with high reactive stromal content (Fig. 6C). Consistently, functional analysis via the Kyoto Encyclopedia of Genes and Genomes (KEGG) database confirmed these findings, revealing significant enrichment of ECM-related and immune signaling pathways, such as ECM-receptor interactions, cell adhesion molecules, NF-kappa B signaling, and B cell receptor signaling, in the highly reactive stroma group (Fig. 6D).
To validate these findings at the gene set level, we performed GSEA focused on ECM-related processes. Three key gene sets—extracellular matrix disassembly (normalized enrichment score, NES = 1.68), extracellular structure organization (NES = 1.74), and extracellular matrix organization (NES = 1.75)—were significantly positively enriched in tumors with high reactive stromal content (Fig. 6E–G). Additionally, positive enrichment was observed in three gene sets associated with TGF-β signaling, including the regulation of the TGF-β signaling pathway (NES = 1.66), the cellular response to the TGF-β stimulus (NES = 1.60), and the transforming growth factor beta receptor superfamily signaling pathway (NES = 1.49) (Figs. 6H–J), supporting the activation of profibrotic and tissue remodeling programs in these patients.
Click here to Correct
Conversely, significant negative enrichment of multiple immune-related gene sets, including the T-cell receptor signaling pathway (NES = − 1.85), positive regulation of T-cell activation (NES = − 2.02), positive regulation of leukocyte cell–cell adhesion (NES = − 2.08), and positive regulation of T-cell proliferation (NES = − 1.95), was observed in tumors with high reactive stromal content (Supplementary Fig. 1), suggesting potential suppression of the adaptive immune component in these tumors. These findings reinforce the transcriptional activation of stromal remodeling programs and provide a mechanistic basis for their association with adverse clinical outcomes.
A
Fig. 6
Gene expression and pathway enrichment analyses associated with reactive stromal content. (A) Heatmap of DEGs between tumors with high and low reactive stromal content. The samples are clustered according to the stromal phenotype. (B) Volcano plot from DEG analysis. Genes with statistically significant differences (adjusted p-value < 0.05) and | log2(fold change) | > 1 are highlighted. (C) Reactome pathway enrichment analysis revealed that tumors with high reactive stromal content were enriched in pathways related to Fc gamma receptor-mediated phagocytosis, collagen degradation, platelet activation, and extracellular matrix organization. (D) KEGG pathway enrichment analysis showing immune-related and extracellular matrix-associated pathways enriched in tumors with high reactive stromal content. (E–G) Gene set enrichment analysis (GSEA) plots for the terms extracellular matrix disassembly (E), extracellular matrix organization (F), and extracellular structure organization (G), indicating significant upregulation of these processes in tumors with highly reactive stroma. (H–J) GSEA plots for the terms regulation of the transforming growth factor beta receptor signaling pathway (H), cellular response to transforming growth factor beta stimulus (I), and the transforming growth factor beta receptor superfamily signaling pathway (J), suggesting transcriptional upregulation of TGF-β–associated programs in tumors with high reactive stromal content.
DISCUSSION
The TME has emerged as a key driver of cancer progression [32, 33]. In this context, our study demonstrated that the proportion of reactive stroma represents an independent prognostic variable in patients with breast cancer. Tumors enriched in reactive stroma exhibited significantly shorter metastasis-free survival (MFS) than those enriched with lower stromal content, a finding that remained robust after adjustment for age and hormone receptor status. These results indicate that the reactive stroma is a biologically relevant feature of the TME, independent of conventional clinicopathological factors.
Reactive stroma, originally described in prostate cancer [20], is characterized by the activation of fibroblasts and the deposition of ECM proteins such as type I collagen, tenascin, and fibronectin. Indeed, abundant ECM accumulation not only provides structural support but also actively promotes tumor progression. Excessive ECM accumulation has been associated with poor prognosis across multiple solid tumors [3436], emphasizing the functional relevance of stromal remodeling in cancer biology.
However, desmoplasia, or reactive stroma, is often acellular or composed predominantly of the extracellular matrix [37, 38]. This characteristic may limit the detection of specific cellular transcripts in bulk RNA-Seq analyses. Therefore, transcriptional profiles derived from tumors with extensive acellular stroma should be interpreted cautiously, particularly when assessing cell-specific gene expression.
In our study, samples were objectively quantified via Masson's trichrome staining, which enabling the precise identification of collagen-rich tissue components. By applying digital image analysis, we achieved accurate segmentation of the fibrous ECM fraction, allowing standardized assessment of the reactive stroma content across tumor samples. This approach provides a robust histological basis for linking stromal content to clinical outcomes and molecular alterations.
Several studies have shown that collagen-enriched ECM promotes tumor progression and metastatic dissemination [39, 40]. Although previous studies have suggested that luminal tumors (ER+/HER2−) present greater stromal content than triple-negative breast cancer (TNBC) [41], our analysis did not reveal significant differences in total or reactive stroma among molecular subtypes. This lack of association underscores the possibility that reactive stroma operates as a transversal feature across breast cancer subtypes, reinforcing its prognostic significance.
Further examination of the reactive stromal compartment revealed significant associations with MFS. These findings suggest that the reactive stroma is not merely a subtype-specific phenomenon but represents a functionally active component of the TME broadly implicated in metastatic risk.
The biological basis for the prognostic role of the reactive stroma is supported by the functional properties of its major components, particularly type I collagen. In addition to providing mechanical support, accumulated collagen promotes cell migration through the activation of integrins and receptors, such as DDR1, leading to cytoskeletal reorganization and acquisition of an invasive phenotype [4245]. Increased collagen density induces tissue stiffness, which modulates cellular behavior through mechanotransduction pathways, notably via focal adhesion kinase (FAK) activation [46, 47]. This process triggers downstream cascades, including ERK activation, Rho-GTPase signaling, and integrin clustering, promoting cellular contractility, invasion, and morphological plasticity [46, 48]. These dynamic interactions between tumor cells and the ECM contribute to tissue remodeling, altered cell–cell and cell–ECM adhesion, and ultimately drive tumor cells toward an invasive phenotype capable of penetrating the matrix and initiating the metastatic cascade [4649]. Our findings reinforce the concept that, in addition to its architectural role, collagen content constitutes a biologically active component of the tumor stroma that is associated with poor prognosis.
Consistent with observations from other solid tumors, our results highlight the clinical relevance of stromal features. In prostate cancer, Ruder et al. developed a quantitative reactive stroma (qRS) biomarker independently associated with increased disease-specific mortality and biochemical recurrence [50]. In breast cancer, Sharma et al. validated Stratipath Breast, which has strong prognostic performance in ER+/HER2 − patients (HR = 2.76; p < 0.001) [51]. While stratification focuses on predicting general disease progression, our study specifically addresses metastasis, demonstrating that reactive stroma ≥ 53.2% was associated with a significantly shorter MFS (HR = 3.75; p < 0.01), and reactive stroma ≥ 42.5% was associated with a trend toward worse OS (HR = 2.51; p = 0.058).
Importantly, assessing reactive stroma with routine histological methods, such as Masson's trichrome staining combined with digital image analysis, provides a simple, reproducible, and clinically feasible strategy. Unlike complex molecular assays, incorporating reactive stroma evaluation into standard pathology workflows entails minimal modification while offering valuable prognostic information in breast cancer.
A key strength of our study is the integration of digital pathology with transcriptomic profiling of FFPE tumor tissue. Although transcriptomic data validated the activation of fibroblast-related and ECM remodeling pathways, they remain complementary to histological quantification.
By applying RNA-Seq to a subset of histologically characterized tumors, we identified a transcriptional signature enriched in biological programs associated with fibroblast activation and ECM remodeling. Notably, tumors with high reactive stroma content exhibited significant overexpression of genes such as FN1, OLR1, EDN2, and MSR1. FN1, which encodes fibronectin, is a glycoprotein expressed in the ECM as a dimer or polymer [52, 53]. This protein plays a pivotal role in physiological processes, such as wound healing, and pathological processes, such as tumor progression, facilitating cell adhesion, migration, and invasion [54, 55]. In line with our histological findings, fibronectin is deposited as fibrillar complexes that reorganize the ECM, generating a dense and stiff matrix enriched in type I collagen. This structural reorganization promotes mechanotransduction, activates pro-oncogenic signaling pathways such as FAK and YAP, and enhances cell migration, immune evasion, and therapy resistance [56, 57].
The correlation between FN1 overexpression and high reactive stroma content supports the concept that the ECM is not merely a static scaffold but also an active participant in tumor progression. Specifically, we demonstrated that tumors with abundant type I and III collagen synthesized by CAF presented a greater risk of metastasis, underscoring the functional importance of the stromal compartment in disease dissemination.
Consistently, we also observed the overexpression of OLR1, a receptor for oxidized lipoproteins (oxLDLs), which has previously been implicated in promoting a protumoral CAF phenotype. OLR1 activation stimulates fibroblast activation, collagen synthesis, and ECM remodeling, creating a dense and mechanically active microenvironment that facilitates tumor cell migration and immune evasion [58]. Additionally, OLR1 expression has been associated with increased infiltration of M2 macrophages and upregulation of immune checkpoint molecules such as PD-L1 [59], further reinforcing its role in establishing an immunosuppressive TME.
Similarly, the overexpression of EDN2, a member of the endothelin family and an adverse prognostic marker in breast cancer, was observed. EDN2 promotes tumor microenvironment remodeling through proliferation, migration, and activation of pro-oncogenic pathways such as the STAT3 pathway [60].
In addition to individual genes, functional pathway enrichment analyses provided broader insights into stromal activity. Reactome and KEGG analyses revealed significant enrichment of ECM-related pathways—including those related to collagen degradation, ECM organization, and ECM-receptor interactions—emphasizing the functional involvement of the stroma in poor-prognosis tumors. Notably, the TGF-β signaling pathway was also enriched in tumors with highly reactive stroma. TGF-β is a well-established driver of fibroblast-to-myofibroblast transition and ECM deposition, and its persistent activation within the TME has been linked to epithelial‒mesenchymal transition (EMT), immunosuppression, and increased metastatic potential [6165].
Conversely, we observed negative enrichment of immune-related gene sets, including those related to T-cell receptor signaling, T-cell proliferation, and leukocyte adhesion [66, 67]. These findings suggest that CAF-mediated ECM remodeling not only promotes invasion but also restricts immune cell infiltration, fostering an immunosuppressive and immune-excluded microenvironment that facilitates metastatic dissemination.
Future studies could also leverage digital pathology and machine learning algorithms to incorporate additional stromal parameters such as fiber density, length, linearity, and thickness. The quantitative assessment of these features could refine risk stratification and enhance the prognostic utility of the reactive stroma.
In summary, our transcriptomic results reinforce the concept that the reactive stroma is a biologically active compartment fostering an invasive and immune-suppressive tumor phenotype. The integration of molecular findings with histological quantification provides a mechanistic rationale for its prognostic value and highlights its potential as a clinical biomarker in breast cancer.
While our study benefits from a robust cohort (n = 182) and a median follow-up of 60 months, future validation in independent, multi-institutional series will further strengthen its generalizability. Our use of Masson’s trichrome staining with digital segmentation offers precise, reproducible measurements of the ECM and could be complemented by multiplex immunohistochemistry or spatial transcriptomics to refine the cellular resolution within the stroma. Finally, as treatment paradigms have evolved over the 2006–2020 period, ongoing investigations incorporating contemporary therapeutic regimens will ensure continued relevance to current clinical practice.
Taken together, our findings provide a comprehensive characterization of the reactive stromal compartment in breast cancer, highlighting its dual role as a structural and signaling component that promotes metastasis. By combining histological and transcriptomic evidence, we establish the reactive stroma as an integral feature of the tumor microenvironment with both prognostic and biological significance. These insights offer a compelling rationale for incorporating stromal assessment into routine diagnostic workflows and for the development of stromal-targeted therapies. As the field moves toward more personalized oncology, evaluating stromal dynamics may refine risk stratification and guide treatment decisions beyond conventional tumor cell–centric models.
CONCLUSIONS
This study advances our understanding of tumor–stroma interactions in breast cancer by establishing the reactive stroma as a clinically relevant prognostic marker of metastasis, independent of molecular subtype. By integrating pathology and transcriptomics from FFPE tissue, we propose a clinically feasible strategy to assess stromal activity, complementing existing molecular classifiers. Future research should aim to validate these findings prospectively and explore therapeutic opportunities targeting the reactive stromal compartment.
Declarations
Ethics approval and consent to participate: The study was approved by the following ethics committees: the Research Ethics Committee of Fundación Arturo López Pérez (FALP; breast cancer protocol ID: 2022-0232-RES-CRC-MUL), Instituto Nacional del Cáncer (INC; project number CRI20050, under the framework of UC project 201016011, as the INC does not have its own ethics committee), and Red Salud UC-CHRISTUS (project ID: 201016011). A waiver of informed consent was granted by all committees, as the study met the criteria for minimal risk, impracticability, and protection of participants’ rights and welfare. All procedures complied with relevant institutional guidelines and regulations.
Consent for publication: Not applicable.
Availability of data and materials:
The scanned histological images (H&E and Masson's trichrome) are available from the corresponding author upon reasonable request due to file size limitations and institutional storage restrictions. The RNA-Seq data will be made publicly available in the Gene Expression Omnibus (GEO) repository upon acceptance of the manuscript. The accession numbers and corresponding links are included in the final published version.
Click here to Correct
A
Competing Interests
D.P.B., M.A.N., V.C.I., J.C.C-R, J.H., G.C., A.P., V.F., B.P., and J.C-I. report funding provided by Production Development Corporation. B.P. and J.C.I. report a relationship with Environ that includes equity or stocks. D.P.B. and J.C.I. have a patent pending to Environ. The authors affiliated with Environ (D.P.B., M.A.N., V.C.I., T.Z., J.H., G.C., A.P., V.F., B.P., and J.C.I.) declare conflicts of interest as employees of the organization. The other authors declare that they have no competing interests. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
A
Funding:
This study was funded by CORFO through the following projects: BreastMets (22CVID-206707), EVA (22CVC2-218129), and ALTA TECH (20IAT130292). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.
A
A
Author Contribution
DPB: Conceptualization, methodology, software, validation, formal analysis, investigation, data curation, writing – original draft, writing – review and editing, visualization, project administration; VT, BC, JSZ, XR, DM, JTL, LM, JP: Conceptualization, investigation, and resources; GC: Methodology, software, formal analysis, data curation; JSCR: Methodology, formal analysis, writing – original draft; MAN, VCI, JH, AP, VF: Validation, investigation, writing – original draft; PG: Conceptualization, validation, investigation, writing – original draft, writing – review and editing, supervision; JCI: Conceptualization, methodology, resources, writing – original draft, writing – review and editing, supervision, project administration; BP: Supervision, project administration, and funding acquisition. All the authors read and approved the final manuscript.
Acknowledgments:
Not applicable.
Electronic Supplementary Material
Below is the link to the electronic supplementary material
A
Data Availability
The scanned histological images (H&E and Masson's trichrome) are available from the corresponding author upon reasonable request due to file size limitations and institutional storage restrictions. The RNA-Seq data will be made publicly available in the Gene Expression Omnibus (GEO) repository upon acceptance of the manuscript. The accession numbers and corresponding links are included in the final published version.
REFERENCES
1.
Mani K, Deng D, Lin C, Wang M, Hsu ML, Zaorsky NG. Causes of death among people living with metastatic cancer. Nat Commun. 2024;15.
2.
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229–63.
3.
Harbeck N, Gnant M. Breast cancer. The Lancet. Lancet Publishing Group; 2017. pp. 1134–50.
4.
Giuliano AE, Connolly JL, Edge SB, Mittendorf EA, Rugo HS, Solin LJ, et al. Breast Cancer—Major changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J Clin. 2017;67:290–303.
5.
Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA et al. Molecular portraits of human breast tumors. Nature [Internet]. 2000;406:747–52. Available from: https://doi.org/10.1038/35021093
6.
De Azambuja E, Cardoso F, De Castro G, Colozza M, Mano MS, Durbecq V, et al. Ki-67 as prognostic marker in early breast cancer: A meta-analysis of published studies involving 12 155 patients. Br J Cancer. 2007;96:1504–13.
7.
Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011. pp. 646–74.
8.
de Visser KE, Joyce JA. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell. Cell Press; 2023. pp. 374–403.
9.
Zhang H, Yue X, Chen Z, Liu C, Wu W, Zhang N, et al. Define cancer-associated fibroblasts (CAFs) in the tumor microenvironment: new opportunities in cancer immunotherapy and advances in clinical trials. Mol Cancer. BioMed Central Ltd; 2023.
10.
Yang D, Liu J, Qian H, Zhuang Q. Cancer-associated fibroblasts: from basic science to anticancer therapy. Exp Mol Med. Springer Nature; 2023. pp. 1322–32.
11.
Yang D, Guo P, He T, Powell CA. Role of endothelial cells in tumor microenvironment. Clin Transl Med. 2021;11.
12.
Fang J, Lu Y, Zheng J, Jiang X, Shen H, Shang X, et al. Exploring the crosstalk between endothelial cells, immune cells, and immune checkpoints in the tumor microenvironment: new insights and therapeutic implications. Cell Death Dis. Springer Nature; 2023.
13.
Zhang Q, Wang Y, Liu F. Cancer-associated fibroblasts: Versatile mediators in remodeling the tumor microenvironment. Cell Signal. 2023;103.
14.
Barron DA, Rowley DR. The reactive stroma microenvironment and prostate cancer progression. Endocr Relat Cancer. 2012.
15.
Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer Nat Publishing Group; 2016. pp. 582–98.
16.
Pickup MW, Mouw JK, Weaver VM. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 2014;15:1243–53.
17.
Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer Nat Res; 2020. pp. 174–86.
18.
Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006. pp. 392–401.
19.
DeClerck YA. Desmoplasia: A response or a niche? Cancer Discov. 2012;2:772–4.
20.
Tuxhorn JA, Ayala GE, Smith MJ, Smith VC, Dang TD, Rowley DR et al. Reactive Stroma in Human Prostate Cancer: Induction of Myofibroblast Phenotype and Extracellular Matrix Remodeling 1. Clinical Cancer Research [Internet]. 2002;2912–23. Available from: http://aacrjournals.org/clincancerres/article-pdf/8/9/2912/2083989/df0902002912.pdf
21.
Barrón-Gallardo CA, Garcia-Chagollán M, Morán-Mendoza AJ, Delgadillo-Cristerna R, Martínez-Silva MG, Villaseñor-García MM et al. A gene expression signature in HER2 + breast cancer patients related to neoadjuvant chemotherapy resistance, overall survival, and disease-free survival. Front Genet. 2022;13.
22.
Ayala G, Tuxhorn JA, Wheeler TM, Frolov A, Scardino PT, Ohori M et al. Reactive Stroma as a Predictor of Biochemical-Free Recurrence in Prostate Cancer. Clinical Cancer Research [Internet]. 2003;9. Available from: http://aacrjournals.org/clincancerres/article-pdf/9/13/4792/2087308/df1303004792.pdf
23.
Bankhead P, Loughrey MB, Fernández JA, Dombrowski Y, McArt DG, Dunne PD et al. QuPath: Open source software for digital pathology image analysis. Sci Rep. 2017;7.
24.
Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.
25.
Liao Y, Smyth GK, Shi W, FeatureCounts. An efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–30.
26.
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15.
27.
Blighe K, Rana S, Lewis M, EnhancedVolcano. Publication-ready volcano plots with enhanced coloring and labeling [Internet]. 2025. Available from: https://bioconductor.org/packages/EnhancedVolcano
28.
Yu G, Wang L-G, Han Y, He Q-Y, clusterProfiler. An R package for comparing biological themes among gene clusters. OMICS [Internet]. 2012;16:284–7. Available from: https://www.liebertpub.com/doi/10.1089/omi.2011.0118
29.
Yu G, enrichplot. Visualization of Functional Enrichment Result [Internet]. 2025. Available from: https://bioconductor.org/packages/enrichplot
30.
Becht E, Giraldo NA, Lacroix L, Buttard B, Elarouci N, Petitprez F et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 2016;17.
31.
Yu G, He QY. ReactomePA: An R/Bioconductor package for reactome pathway analysis and visualization. Mol Biosyst. 2016;12:477–9.
32.
Sabit H, Arneth B, Abdel-Ghany S, Madyan EF, Ghaleb AH, Selvaraj P et al. Beyond Cancer Cells: How the Tumor Microenvironment Drives Cancer Progression. Cells. 2024.
33.
Mayer S, Milo T, Isaacson A, Halperin C, Miyara S, Stein Y et al. The tumor microenvironment shows a hierarchy of cell–cell interactions dominated by fibroblasts. Nat Commun. 2023;14.
34.
Zhang H, Lei Z, Tian R, Wang Z. Polyamidoamine starburst dendrimer-activated chromatography paper-based assay for sensitive detection of telomerase activity. Talanta. 2018;178:116–21.
35.
Huang H, Brekken RA. Beyond Stiffness: Collagen Signaling in Pancreatic Cancer and Pancreas Regeneration. Am J Pathol. 2020;190:1622–4.
36.
Myllyharju J, Kivirikko KI. Collagens, modifying enzymes and their mutations in humans, flies and worms. Trends in Genetics. Elsevier Ltd; 2004. pp. 33–43.
37.
Walker RA. The complexities of breast cancer desmoplasia. Breast Cancer Research [Internet]. 2001;3:143–5. Available from: http://breast-cancer-research.com/content/3/3/143
38.
Lo A, Wang LCS, Scholler J, Monslow J, Avery D, Newick K, et al. Tumor-promoting desmoplasia is disrupted by depleting FAP-expressing stromal cells. Cancer Res. 2015;75:2800–10.
39.
Slattery ML, John E, Torres-Mejia G, Stern M, Lundgreen A, Hines L et al. Matrix Metalloproteinase Genes Are Associated with Breast Cancer Risk and Survival: The Breast Cancer Health Disparities Study. PLoS ONE. 2013;8.
40.
Hanley CJ, Noble F, Ward M, Bullock M, Drifka C, Mellone M et al. A subset of myofibroblastic cancer-associated fibroblasts regulate collagen fiber elongation, which is prognostic in multiple cancers [Internet]. Available from: www.impactjournals.com/oncotarget
41.
Olsson LT, Williams LA, Midkiff BR, Kirk EL, Troester MA, Calhoun BC. Quantitative analysis of breast cancer tissue composition and associations with tumor subtype. Hum Pathol. 2022;123:84–92.
42.
Sirvent A, Espie K, Papadopoulou E, Naim D, Roche S. New functions of DDR1 collagen receptor in tumor dormancy, immune exclusion and therapeutic resistance. Front Oncol. Frontiers Media S.A.; 2022.
43.
Su H, Yang F, Fu R, Trinh B, Sun N, Liu J, et al. Collagenolysis-dependent DDR1 signaling dictates pancreatic cancer outcome. Nature. 2022;610:366–72.
44.
Elkamhawy A, Lu Q, Nada H, Woo J, Quan G, Lee K. The journey of ddr1 and ddr2 kinase inhibitors as rising stars in the fight against cancer. Int J Mol Sci MDPI; 2021.
45.
Sun X, Wu B, Chiang HC, Deng H, Zhang X, Xiong W, et al. Tumor DDR1 promotes collagen fiber alignment to instigate immune exclusion. Nature. 2021;599:673–8.
46.
Agrawal A, Javanmardi Y, Watson SA, Serwinski B, Djordjevic B, Li W et al. Mechanical signatures in cancer metastasis. npj Biological Physics and Mechanics [Internet]. 2025;2:3. Available from: https://www.nature.com/articles/s44341-024-00007-x
47.
Northcott JM, Dean IS, Mouw JK, Weaver VM. Feeling stress: The mechanics of cancer progression and aggression. Front Cell Dev Biol. Frontiers Media S.A.; 2018.
48.
Kumar S, Weaver VM. Mechanics, malignancy, and metastasis: The force journey of a tumor cell. Cancer Metastasis Rev. 2009. pp. 113–27.
49.
Montagner M, DuPont S. Mechanical forces as determinants of disseminated metastatic cell fate. Cells. Multidisciplinary Digital Publishing Institute (MDPI); 2020.
50.
Ruder S, Gao Y, Ding Y, Bu P, Miles B, De Marzo A, et al. Development and validation of a quantitative reactive stroma biomarker (qRS) for prostate cancer prognosis. Hum Pathol. 2022;122:84–91.
51.
Sharma A, Lövgren SK, Eriksson KL, Wang Y, Robertson S, Hartman J et al. Validation of an AI-based solution for breast cancer risk stratification using routine digital histopathology images. Breast Cancer Res. 2024;26.
52.
Li J, Chen C, Chen B, Guo T. High FN1 expression correlates with gastric cancer progression. Pathol Res Pract. 2022;239.
53.
Chen C, Ye L, Yi J, Liu T, Li Z. FN1 mediated activation of aspartate metabolism promotes the progression of triple-negative and luminal a breast cancer. Breast Cancer Res Treat. 2023;201:515–33.
54.
Zhang XX, Luo JH, Wu LQ. FN1 overexpression is correlated with unfavorable prognosis and immune infiltrates in breast cancer. Front Genet. 2022;13.
55.
Wang H, Zhang J, Li H, Yu H, Chen S, Liu S et al. FN1 is a prognostic biomarker and correlated with immune infiltrates in gastric cancers. Front Oncol. 2022;12.
56.
Pan H, Luo Z, Lin F, Zhang J, Xiong T, Hong Y et al. FN1, a reliable prognostic biomarker for thyroid cancer, is associated with tumor immunity and an unfavorable prognosis. Oncol Lett. 2024;28.
57.
Ashok G, Miryala SK, Saju MT, Anbarasu A, Ramaiah S. FN1 encoding fibronectin as a pivotal signaling gene for therapeutic intervention against pancreatic cancer. Mol Genet Genomics. 2022;297:1565–80.
58.
Sun Y, Ying K, Sun J, Wang Y, Qiu L, Ji M et al. PRRX1-OLR1 axis supports CAFs-mediated lung cancer progression and immune suppression. Cancer Cell Int. 2024;24.
59.
Sun X, Fu X, Xu S, Qiu P, Lv Z, Cui M et al. OLR1 is a prognostic factor and correlated with immune infiltration in breast cancer. Int Immunopharmacol. 2021;101.
60.
Wang X, Li C, Chen T, Li W, Zhang H, Zhang D et al. Identification and Validation of a Five-Gene Signature Associated With Overall Survival in Breast Cancer Patients. Front Oncol. 2021;11.
61.
Huang M, Fu M, Wang J, Xia C, Zhang H, Xiong Y et al. TGF-β1-activated cancer-associated fibroblasts promote breast cancer invasion, metastasis and epithelial–mesenchymal transition by autophagy or overexpression of FAP-α. Biochem Pharmacol. 2021;188.
62.
Ping Q, Wang C, Cheng X, Zhong Y, Yan R, Yang M et al. TGF-β1 dominates stromal fibroblast-mediated EMT via the FAP/VCAN axis in bladder cancer cells. J Transl Med. 2023;21.
63.
Chung JYF, Chan MKK, Li JSF, Chan ASW, Tang PCT, Leung KT et al. Tgf-β signaling: From tissue fibrosis to tumor microenvironment. Int J Mol Sci MDPI; 2021.
64.
Yoon H, Tang CM, Banerjee S, Delgado AL, Yebra M, Davis J et al. TGF-β1-mediated transition of resident fibroblasts to cancer-associated fibroblasts promotes cancer metastasis in gastrointestinal stromal tumor. Oncogenesis. 2021;10.
65.
Joshi RS, Kanugula SS, Sudhir S, Pereira MP, Jain S, Aghi MK. The role of cancer-associated fibroblasts in tumor progression. Cancers (Basel). MDPI AG; 2021. pp. 1–27.
66.
Dominguez CX, Müller S, Keerthivasan S, Koeppen H, Hung J, Gierke S, et al. Single-cell RNA sequencing reveals stromal evolution into LRRC15 + myofibroblasts as a determinant of patient response to cancer immunotherapy. Cancer Discov. 2020;10:232–53.
67.
Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFβ attenuates tumor response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554:544–8.
Total words in MS: 6817
Total words in Title: 19
Total words in Abstract: 262
Total Keyword count: 5
Total Images in MS: 6
Total Tables in MS: 2
Total Reference count: 67