Abstract
Objective: Triple-negative breast cancer (TNBC) is a heterogeneous and aggressive cancer. Although our previous study classified primary TNBC into four subtypes, comprehensive longitudinal investigations are lacking.
Methods: We assembled a large-scale, real-world cohort comprised of 880 TNBC patients [465 early-stage TNBC (eTNBC) and 415 metastatic TNBC (mTNBC) patients] who were treated at Fudan University Shanghai Cancer Center. The longitudinal dynamics of TNBC subtypes during disease progression were elucidated in this patient cohort. Comprehensive analysis was performed to compare primary and metastatic lesions within specific TNBC subtypes.
Results: The recurrence and metastasis rates within 3 years after initial diagnosis in the eTNBC cohort were 10.1% (47/465). The median overall survival (OS) in the mTNBC cohort was 27.2 months [95% confidence interval (CI), 24.4–30.2 months], which indicated a poor prognosis. The prognostic significance of the original molecular subtypes in both eTNBC and mTNBC patients was confirmed. Consistent molecular subtypes were maintained in 77.5% of the patients throughout disease progression with the mesenchymal-like (MES) subtype demonstrating a tendency for subtype transition and brain metastasis. Additionally, a precision treatment strategy based on the metastatic MES subtype of target lesions resulted in improved progression-free survival in the FUTURE trial.
Conclusions: Our longitudinal study comprehensively revealed the clinical characteristics and survival of patients with the original TNBC subtypes and validated the consistency of most molecular subtypes throughout disease progression. However, we emphasize the major importance of repeat pathologic confirmation of the MES subtype.
keywords
Introduction
Breast cancer was the most common malignant tumor among women in 2020. There were 2.26 million new cases of breast cancer worldwide in 2020, making breast cancer the leading cancer globally1. Compared with breast cancer patients in Western countries, breast cancer patients in China tend to be younger at disease onset and exhibit a greater incidence of triple-negative breast cancer (TNBC)2,3. TNBC refers to breast cancer in which estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) are not expressed. TNBC accounts for approximately 15% of all breast cancers4. Compared with other subtypes, TNBC is characterized by increased malignancy and a poorer prognosis. The systemic treatment of TNBC currently relies on chemotherapy without effective targeted therapies, which is a well-recognized clinical challenge5. The risk of recurrence and metastasis within 1–3 years after initial diagnosis is high for patients with TNBC. Indeed, once distant metastasis occurs the median survival time is only 18–24 months6,7. Therefore, creating effective precision treatment options for TNBC based on the molecular biology remains a critical and prominent research focus8.
Genome-guided targeted therapy has laid the foundation for precision medicine in TNBC9. The poly (ADP-ribose) polymerase [PARP] inhibitor, olaparib, targets patients with BRCA1 or BRCA2 mutations and significantly improved disease-free survival (DFS) for patients in the OlympiA trial10. Additionally, the PAKT trial revealed that the protein kinase B (AKT) inhibitor, capivasertib, significantly prolongs progression-free survival (PFS) and overall survival (OS)11. Although specific DNA alterations for targeted therapies have been identified, only a small fraction of TNBCs have targetable mutations10,12–15. Notably, relevant studies have focused primarily on specific targets while overlooking the intrinsic TNBC subtypes.
With advances in molecular testing technologies, Lehmann et al.16,17 concluded that TNBC is a highly heterogeneous mixed subtype of breast cancer that consists of seven subtypes based on differences in expression profiles. We previously performed multiomic profiling of large-scale Chinese TNBCs and classified the TNBCs into the following four subtypes: (1) luminal androgen receptor (LAR) is characterized by androgen receptor signaling; (2) immunomodulatory (IM) is characterized by high immune cell signaling and cytokine signaling gene expression; (3) basal-like immune-suppressed (BLIS) is characterized by upregulation of the cell cycle, activation of DNA repair, and downregulation of immune response genes; and (4) mesenchymal-like (MES) is enriched in mammary stem cell pathways18. Furthermore, we developed a clinically accessible immunohistochemical subtyping method19 and proposed that subtype-based precision treatment strategies based on molecular characteristics significantly improve the overall prognosis of TNBC patients20,21. However, there is still a long way to go with considerable room for improvement. The current molecular classification of TNBC is based on the primary tumor cohort. Longitudinal cohort studies analyzing the stability and variability of the subtypes are lacking and there is a lack of research on the long-term clinical outcomes of the subtypes.
Is the subtype determination system developed based on primary lesions suitable for metastatic TNBC (mTNBC)? Specifically, if a patient is initially diagnosed with the MES subtype, does the subtype retain the MES characteristics when the disease spreads to other sites? Do early-stage and advanced BLIS subtypes exhibit similar biological behaviors? To address these scientific issues, we established early-stage (n = 465) and metastatic (n = 415) clinical cohorts of patients with TNBC and procured 40 paired primary and metastatic TNBC samples. Following the subtype classification of TNBC, we conducted a comprehensive longitudinal study to thoroughly investigate the clinical characteristics and assess the clinical value of our original TNBC subtyping system.
Materials and methods
Real-world cohorts of patients with early-stage TNBC (eTNBC)
A total of 465 consecutive Chinese patients who were diagnosed with malignant TNBC and treated in the Department of Breast Surgery at Fudan University Shanghai Cancer Center (FUSCC) from 1 January 2007 to 31 December 2014 were enrolled according to the following defined criteria: 1) female patients diagnosed with unilateral invasive ductal carcinoma with an ER, PR, or HER2 phenotype using < 1% positively stained cells as the cut-off value for ER/PR negativity based on immunohistochemistry (IHC)22; 2) no evidence of distant metastasis at the time of diagnosis; and 3) availability of a complete medical history for further analysis. Patients with breast carcinoma in situ or inflammatory breast cancer were excluded. Follow-up evaluations within this early-stage cohort of patients were completed on 6 December 2023 and the median follow-up was 68.1 months [interquartile range (IQR), 46.8–100.9 months].
Real-world cohorts of patients with mTNBC
We retrospectively recruited patients with mTNBC treated at FUSCC from October 2018 to November 2020. The eligibility criteria for patients were as follows: (1) female patients diagnosed with metastatic breast carcinoma with an ER, PR, or HER2 phenotype by the Department of Pathology at FUSCC; and (2) a sufficient number of tumor biopsies obtained for IHC staining (AR, CD8, and FOXC1)19 to classify the tumors into four subtypes (LAR, IM, BLIS, and MES)18 based on a central pathologic examination of tumor specimens performed by the Department of Pathology at FUSCC; and (3) complete medical history obtained for further analysis. Patients with multiple primary tumors or solitary lesions who had undergone radiotherapy treatment were excluded from this study. The clinicopathologic characteristics included age, number of metastases, treatment history, disease-free interval (DFI), duration of first-line therapy, and ER, PR, HER2, and Ki-67 status. Follow-up evaluations within this cohort of patients were completed on 6 December 2023 and the median duration of follow-up was 38.9 months (IQR, 27.1–61.1 months).
All tissue samples included in the present study were approved by the FUSCC Ethics Committee [Approval No. 2011226-17 (2010-ZZK-31)] and each patient provided written informed consent.
Immunohistochemical staining
We performed immunohistochemical staining on 4 paraffin-embedded sections (4 μm thick) of tumor specimens to evaluate the expression of AR, CD8, and FOXC1. We used the following primary antibodies: anti-AR (ab133273, 1:200 dilution; Abcam, Cambridge, UK); anti-CD8 (SP57, undiluted; Ventana, Tucson, Arizona, USA); and anti-FOXC1 (ab227977, 1:500 dilution; Abcam). All four antibodies used were rabbit monoclonal antibodies and were recommended by the manufacturers for immunohistochemical experiments involving paraffin-embedded sections. The tumors were classified into IHC-based subtypes according to the staining results. Detailed descriptions of the IHC-based classification methods were published previously19.
Statistical analysis
The data distribution was characterized by frequency tabulation and summary statistics. The relationships between the TNBC subtypes and clinicopathologic parameters were determined with Pearson’s χ2 test or Fisher’s exact test, as indicated. Relapse-free survival (RFS) was defined as the time from diagnosis to first recurrence or a diagnosis of contralateral breast cancer. OS was defined as the time from diagnosis of mTNBC to death from any cause. The objective response rate (ORR) was defined as the percentage of participants with an objective complete response (CR) or partial response (PR) according to RECIST (v1.1) among all participants. Patients without events were censored from the date of the last follow-up evaluation. OS and RFS were compared among the four TNBC subtypes using the Kaplan‒Meier method and a Cox regression model. The Wilcoxon test was used to compare continuous variables. All P values were two-tailed, and a P < 0.05 was considered to indicate statistical significance. R software (version 4.3.2; The R Project for Statistical Computing, https://www.r-project.org/) was used for calculations and analyses.
Results
Research cohorts and study design
To unravel the heterogeneity of TNBC and explore precision treatment strategies, we established molecular subtypes based on a multiomics cohort of primary TNBC tumors. The TNBC populations were divided into the following four subtypes: LAR; IM; BLIS; and MES18. To deepen our understanding of the clinical characteristics of this novel TNBC classification system and provide valuable guidance for clinical practice, we assembled a real-world clinical cohort of TNBC patients and conducted a comprehensive analysis of TNBC subtypes from a temporal perspective (Figure 1A). These cohorts included an eTNBC cohort comprised of 465 individuals and a metastatic cohort comprised of 415 patients with TNBC treated at FUSCC. Subtypes were determined based on primary tumors in the eTNBC cohort, while subtypes were determined based on metastatic lesions in the mTNBC cohort. Moreover, we retrospectively collected 40 paired samples of primary tumors and paired metastatic lesions within the mTNBC cohort.
We conducted a comprehensive analysis of eTNBC and mTNBC subtypes, which revealed important insights into the stability and variability of molecular subtypes. The clinical dataset, which consisted of 880 TNBC patients, included subtype information, clinical data (age and metastatic sites), pathologic information (Ki-67 and lymph node status), treatment history (chemotherapy and radiotherapy), and prognostic information (RFS and OS). In the current study the previously published RFS data for eTNBC subtypes were updated, and for the first time, OS data are presented for each mTNBC subtype in a real-world cohort.
Poor outcomes of primary and metastatic TNBC patients in a real-world cohort
The median duration of follow-up evaluations in the eTNBC cohort was 68.1 months (IQR, 46.8–100.9 months). Most tumors [91.6% (426/465)] were invasive ductal carcinomas (IDCs) and 40.3% (186/462) of the TNBCs were lymph node-positive (Figure 1B). As of 6 December 2023, a total of 68 patients with TNBC had recurrences (Figure 1C), 69.1% (47/68) of whom relapsed within 3 years. The most common sites of metastasis were bone [47.1% (32/68)], lung [44.1% (30/68)], and lymph nodes [42.6% (29/68); Figure 1D].
The median duration of follow-up evaluations in the mTNBC cohort was 38.9 months (IQR, 27.1–61.1 months) and the median DFI was 17.0 months [95% confidence interval (CI), 15.2–18.0; Table S1]. Of the mTNBC patients, 86.5% (359/415) were < 60 years of age and 85.8% (356/415) of the patients received adjuvant chemotherapy. Additionally, 56.4% (234/415) of the TNBC patients had a heavy tumor burden at the time of enrollment with ≥ 2 metastatic sites (Table 1). Lymph nodes were the predominant biopsy site in this cohort (Figure S1A), possibly because of the frequent involvement as the primary site for initial metastasis (Figure S1B). The most common sites of metastasis were lymph nodes [75.2% (312/415)], bones [49.6% (206/415)], and chest wall [49.6% (206/415); Figure 1E]. Detailed clinical information for the mTNBC cohort is shown in Table 1. The median DFS of patients with mTNBC was 17.0 months (95% CI, 15.2–18.1 months; Figure 1F). Importantly, the median OS in these patients was only 27.2 months (95% CI, 24.4–30.2 months; Figure 1G). The above data demonstrated that patients with TNBC have a poor prognosis, which highlights the urgent need for further research to improve patient outcomes.
Stability of molecular subtypes (excluding the MES subtype) between primary and metastatic TNBC
The molecular classification of primary TNBC tumors involves full exploration of the heterogeneity of the tumors and serves to guide clinical treatment for improving patient prognosis18,20. We conducted a comprehensive longitudinal study to determine the clinical characteristics and clinical value of TNBC molecular subtypes. According to a previously established IHC-based classification method, we performed IHC staining and classification in the TNBC primary and metastatic cohorts, followed by a comparative analysis (Figure 2A). The molecular subtypes in the eTNBC cohort were distributed as follows: LAR (25.1%); IM (19.6%); BLIS (38.2%); and MES (17.1%). Proprotions of patients in the mTNBC cohort with the LAR, IM, BLIS, and MES subtypes were 34.9%, 13.5%, 42.4%, and 9.2%, respectively. Notably, compared with patients in the primary cohort, the proportion of patients in the metastatic cohort was significantly greater for the LAR and BLIS subtypes, while the proportion of patients in the MES and IM subtypes was lower (P < 0.001; Figure 2B). These findings indicated that the original TNBC molecular subtypes were present in both primary lesions and metastases, albeit in varying proportions.
To further investigate the clinical characteristics of the different molecular subtypes involved in disease progression, we obtained paired samples of primary and metastatic lesions from 40 TNBC patients and classified the lesions (Figure 2C). The distribution of the different molecular subtypes at each biopsy site is shown in Figure S2. Overall, 77.5% (31/40) of the patients maintained consistent molecular subtypes during disease progression, with LAR, IM, and BLIS concordance rates of 80.0%, 100.0%, and 78.6%, respectively, in the paired samples (Figure 2D and 2E). Furthermore, we performed an analysis of key molecules associated with each subtype. Our findings revealed that the levels of AR, CD8, and FOXC1 (key molecules in the subtyping system) protein expression remained stable throughout disease progression and showed no significant changes (Figure 2F–H). Surprisingly, the consistency of the MES subtype was poor because most patients transitioned to other subtypes upon recurrence or metastasis. Furthermore, the transition process was unidirectional with minimal evidence of other subtypes converting to the MES subtype during disease progression (Figure 2D and 2E).
In general, molecular subtype stability was observed between primary and metastatic TNBC except for the MES subtype. The subtype of TNBC metastatic lesions can be predicted by the molecular subtype of the primary TNBC among TNBC patients with LAR, IM, or BLIS. Furthermore, additional research is warranted to determine the predisposition of patients with the MES subtype during subtype transition.
A precision treatment strategy guided by the MES subtype of target lesions improves treatment efficacy
We conducted the FUTURE trial (ClinicalTrials.gov identifier: NCT03805399), a phase II umbrella trial, to evaluate the potential efficacy of subtype-based precision strategies in patients with mTNBC. As of 31 March 2022, a total of 15 mTNBC patients with the MES subtype were enrolled in the FUTURE trial. Among the 15 patients, 12 received precision treatment specific to the MES subtype, for which efficacy data are available. Among the 12 patients, 8 were diagnosed with the MES subtype based on IHC assessment of the target lesion (matched group), while 4 patients were diagnosed with the MES subtype based on the primary tumor or non-target lesions (unmatched group; Figure 3A). Interestingly, compared to the patients in the unmatched group, the patients in the matched group had longer progression-free survival (Figure 3B) and achieved an ORR of 50% (Figure 3C). None of the patients in the unmatched group achieved an ORR (Figure 3C). Furthermore, patients in the unmatched group experienced faster disease progression and a shorter duration of response (Figure 3D and 3E). Typical imaging examples of the matched and unmatched groups are shown in Figure 3F and 3G, respectively. In general, a precision treatment strategy guided by the MES subtype of target lesions improved treatment efficacy.
The original TNBC subtypes exhibit prognostic value in eTNBC and mTNBC patients
The differences in subtype proportions between primary and metastatic TNBC cohorts imply diverse clinical outcomes linked to specific subtypes. The previously published RFS data for eTNBC subtypes were updated and the long-term OS data from a large-scale cohort of patients with mTNBC subtypes were presented for the first time. Overall, patients in the IM subgroup of the eTNBC cohort exhibited longer RFS than patients in the MES or LAR subgroup [MES vs. IM, hazard ratio (HR): 3.2, 95% CI: 1.2–8.2; P = 0.02 and LAR vs. IM, HR: 2.7, 95% CI: 1.1–7.0; P = 0.04; Figure 4A and 4B]. A significant difference in OS was detected among the four subtypes in the mTNBC cohort (P = 0.001; Figure 4C). The molecular subtype was shown to be an independent prognostic factor for mTNBC based on multivariate Cox analysis and the IM subtype exhibited a longer OS than the other subtypes (35.4 months, 95% CI: 20.9–49.9 months; Table S2). The median OS times of patients with the LAR and MES subtypes were 29.6 (95% CI: 22.6–36.5 months) and 25.4 months (95% CI: 20.3–30.5 months; Table 2), respectively. BLIS tumors exhibited the worst OS (19.9 months, 95% CI: 14.6–25.2 months; Table 2) and were associated with a significantly poorer prognosis than IM tumors (BLIS vs. IM, HR: 2.0, 95% CI: 1.3–3.1; P = 0.001; Figure 4D), which corroborated previous literature on the poor prognosis of BLIS patients18. Additionally, the discovery of the inferior prognosis associated with BLIS tumors shed light on the observation that patients with the BLIS subtype developed metastasis at a younger age (< 60 years) and had a greater tumor burden (multiple metastases) compared to patients with other subtypes (Table 1).
In addition, we examined the trends in the expression of three molecules (AR, CD8, and FOXC1) among the four subtypes in both the primary and metastatic TNBC cohorts. FOXC1 was highly expressed in the metastatic cohorts of all subtypes except the MES cohort, suggesting that high expression of FOXC1 indicates poor prognosis in patients with the LAR, IM, and BLIS subtypes (Figure S3A–D). Organotropic metastasis of patients with TNBC subtypes was then examined and an intriguing phenomenon was observed. Despite representing a relatively small proportion of patients in the overall cohort, patients in the MES subgroup exhibited a significantly greater proportion of brain metastasis, accounting for 25.0% of the population (Figure 4E and Figure S4). Furthermore, among the four TNBC subtypes, the MES subgroup had the highest incidence of brain metastasis (Figure 4F and 4G). These findings strongly suggest the propensity of the MES subtype for brain metastasis and highlight the importance of enhancing brain surveillance.
Discussion
Previous studies have classified primary TNBCs into subtypes but have lacked comprehensive longitudinal efforts to gain an in-depth understanding of subtype biology, treatment efficacy, and prognosis16,18,23. A large-scale, real-world cohort involving 880 TNBC patients was assembled to determine the longitudinal dynamics of subtypes during disease progression. Most patients maintained their molecular subtype consistently throughout disease progression, while the MES subtype showed a tendency toward subtype transition. Furthermore, the necessity of resampling and pathologically confirming the subtype of mTNBCs to maximize the benefits of subtype-guided precision treatment strategies was confirmed. Distinct differences in survival and prognostic features between primary and metastatic tumors were demonstrated within each subtype (Figure 5).
Tumor metastasis is a multistep, orderly cellular-biological process that involves invasion, dissemination, and colonization at distant metastatic sites and is known as the “invasion-metastasis cascade”24–27. Numerous studies have demonstrated that the molecular profile and microenvironment of tumors undergo changes during metastasis, and subtype switching is common28–33. Additionally, due to tumor heterogeneity, tumor evolution can occur during drug treatment, leading to drug resistance and immune evasion34,35. Alterations in tumor biological characteristics that occur during metastasis and progression imply that precision treatment strategies based on the molecular features of the primary tumor may not be suitable for metastatic patients. This underscores the potential clinical importance of conducting dynamic biopsies and even sampling from multiple sites during treatment as well as the critical importance of longitudinal cohort studies in understanding tumor metastasis and evolution.
In recent years, cross-sectional studies have demonstrated the heterogeneity of TNBC and the ability to classify TNBC into distinct molecular subtypes16,23,36. However, further exploration of subtype characteristics requires longitudinal cohort studies. Our study based on real-world TNBC cohorts confirmed the presence of the original four subtypes in the eTNBC and mTNBC cohorts. Based on updated RFS data and newly disclosed OS data, the original TNBC molecular subtypes were shown to effectively determine patient prognosis in both cohorts. In both early and late-stage TNBC cohorts, the BLIS subtype was consistently associated with poor prognosis, which is in agreement with previous research findings18. This association further elucidated the observed trends of younger age at diagnosis and a greater tumor burden (multiple sites of metastasis) among patients with the BLIS subtype in our study. Additionally, our longitudinal study indicated that 77.5% of patients have maintained consistency in the TNBC molecular subtypes during disease progression, confirming the stability and reliability of our novel classification system.
In addition the stability of the subtypes, variations in the MES subtype were also observed. Patients with the MES subtype at the primary site were shown to exhibit changes to other subtypes upon relapse or metastasis, while reverse transformation was rare. The MES subtype is known to overexpress stem cell-related genes and possess stem cell-like properties18. Tumors characterized by the MES subtype exhibit activation of the JAK/STAT3 signaling pathway, which has a crucial role in the growth and maintenance of stem cell-like breast cancer cells18. The MES subtype also has elevated expression of the tumor stem cell marker, DCLK119. Therefore, tumors of the MES subtype may possess characteristics of cancer stem cells, including self-renewal capabilities and increased differentiation potential37. The stem cell-like nature of the MES subtype possibly explains the transitional characteristics and poor prognosis. Precision treatment guided by metastatic lesion-based classification was shown to provide a longer PFS than precision treatment guided by primary lesion-based classification in advanced TNBC patients enrolled in our clinical trial, emphasizing the importance of repeat pathologic confirmation of the MES subtype. Furthermore, we identified a propensity for brain metastasis in the MES subtype, highlighting the importance of thorough brain surveillance in clinical practice. In conclusion, understanding the evolution of these TNBC subtypes, including potential changes during the course of the disease, could offer valuable insights into therapeutic response and help optimize patient outcomes.
The current study had limitations. The number of paired samples between primary and metastatic lesions was small. To consolidate and validate the conclusions, the current study was augmented by constructing two large real-world cohorts comprising 465 eTNBC patients and 415 mTNBC patients. Additionally, the current study lacked in-depth mechanistic investigations into the clinical phenomena observed, especially regarding the molecular biological differences between primary and metastatic tumors of the MES subtype, which will be a focus of corollary studies.
Conclusions
The current study underscores the importance of longitudinal studies for understanding the heterogeneity of TNBC and optimizing patient management strategies. Through a longitudinal investigation, the clinical characteristics of the original TNBC subtypes were comprehensively elucidated and the consistency of most molecular subtypes during disease progression were confirmed. Indeed, there is a critical need for repeat pathologic confirmation of the MES subtype due to the propensity for subtype switching. Additionally, the prognostic significance of the subtypes in was validated in patients with eTNBC and mTNBC.
Supporting Information
Conflict of interest statement
No potential conflicts of interest are disclosed.
Authors’ contributions
Conceived and designed the analysis: Yizhou Jiang, Zhonghua Wang.
Collected the data: Xiuzhi Zhu, Xiaohan Ying, Yin Liu, Yunyi Wang.
Contributed the data or analysis tools: Yin Liu, Yunyi Wang, Li Chen, Zhiming Shao, Xi Jin.
Performed the analysis: Xiuzhi Zhu, Xiaohan Ying.
Wrote the paper: Xiuzhi Zhu, Xiaohan Ying.
Data availability statement
The clinical datasets generated and/or analyzed during the current study are available upon request from the corresponding author for research and non-commercial purposes.
Acknowledgements
The authors would like to extend thanks to all patients, clinicians, and nurses for their efforts in this study.
Footnotes
↵*These authors contributed equally to this work.
- Received January 5, 2024.
- Accepted March 27, 2024.
- Copyright: © 2024, The Authors
This work is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License.