1
|
Sasaki K, Takahashi S, Ouchi K, Shirota H, Sato N, Kaneko K, Masuda N, Fujishima F, Sato S, Ishioka C. Dynamic predictive power of TP53 signatures in breast cancer prognosis: Pre- and post-neoadjuvant chemotherapy insights. Transl Oncol 2025; 56:102398. [PMID: 40245753 PMCID: PMC12020852 DOI: 10.1016/j.tranon.2025.102398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND The TP53 signature determined using a biopsy specimen before neoadjuvant chemotherapy (pre-NAC biopsy specimens) predicts NAC response and prognosis in breast cancer. We aimed to compare the clinical utility of the TP53 signature determined using pre-NAC biopsy specimens and surgical specimens after NAC (post-NAC surgical specimens). METHODS This observational cohort study included patients with paired pre-NAC biopsy and post-NAC surgical specimens, analyzing the association between the TP53 signature from each specimen and prognosis (UMIN000042055). RESULTS Pre-NAC biopsy specimens classified 71 patients into those having a TP53 mutant signature (pre-mt, n = 47) and wild-type signature (pre-wt, n = 24), with the same for post-NAC surgical specimens (post-mt, n = 16 and post-wt, n = 55). Among the 47 pre-mt patients, 31 became post-wt (pre-mt/post-wt), whereas 16 remained post-mt (pre-mt/post-mt). All pre-wt patients remained post-wt (pre-wt/post-wt). Recurrence-free survival (RFS) was significantly shorter in the pre-mt group than in the pre-wt group, although no significant difference was observed between the post-mt and post-wt groups. Change in the TP53 signature following NAC did not affect predictive ability of the TP53 signature determined using pre-NAC biopsy specimens. CONCLUSIONS The TP53 signature status should be determined using pre-NAC biopsy specimens.
Collapse
Affiliation(s)
- Keiju Sasaki
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan; Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shin Takahashi
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan; Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kota Ouchi
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan; Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hidekazu Shirota
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan; Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuaki Sato
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Kouji Kaneko
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Norikazu Masuda
- Department of Surgery, Breast Oncology, National Hospital Organization Osaka National Hospital, Osaka, Japan; Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumiyoshi Fujishima
- Department of Pathology, Tohoku University Hospital, Sendai, Japan; Division of Diagnostic Pathology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Satoko Sato
- Department of Pathology, Tohoku University Hospital, Sendai, Japan
| | - Chikashi Ishioka
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan; Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Clinical Oncology Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, Japan; JR Sendai Hospital, Sendai, Japan.
| |
Collapse
|
2
|
Thomas N, Foukakis T, Willard-Gallo K. The interplay between the immune response and neoadjuvant therapy in breast cancer. Front Oncol 2025; 15:1469982. [PMID: 40421087 PMCID: PMC12104209 DOI: 10.3389/fonc.2025.1469982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 04/16/2025] [Indexed: 05/28/2025] Open
Abstract
Treatment of early breast cancer is currently experiencing a rapid evolution because of important insight into tumor subtypes and continuous development and improvement of novel therapeutics. Historically considered non-immunogenic, breast cancer has seen a paradigm shift with increased understanding of immune microenvironment, which have revealed extensive heterogeneity in tumor-associated inflammation. Notably, the more aggressive breast cancer subtypes, including triple-negative and HER2-positive, have exhibited favorable responses to combined chemo-immunotherapy protocols. Neoadjuvant therapy has emerged as the standard of care for these tumors, with pathological complete response used as a surrogate endpoint for long-term clinical outcomes and coincidently expediting new drug approval. The neoadjuvant setting affords a unique opportunity for in vivo treatment response evaluation and effects on the tumor microenvironment. In this review, the predictive and prognostic value of the tumor immune microenvironment before, during, and after treatment across various therapeutic regimens, tailored to distinct breast cancer subtypes, is carefully examined.
Collapse
Affiliation(s)
- Noémie Thomas
- Molecular Immunology Unit, Institut Jules Bordet, Brussel, Belgium
| | - Theodoros Foukakis
- Translational Breast Cancer Research, Department of Oncology-Pathology, Karolinska Institute, Stokholm, Sweden
| | | |
Collapse
|
3
|
Li Q, Li W, Wang J, Li X, Ji Y, Wu M. Non-invasive prediction of DCE-MRI radiomics model on CCR5 in breast cancer based on a machine learning algorithm. Cancer Biomark 2025; 42:18758592251332852. [PMID: 40395152 DOI: 10.1177/18758592251332852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
BackgroundNon-invasive methods with universal prognostic guidance for detecting breast cancer (BC) survival biomarkers need to be further explored.ObjectiveThis study aimed to investigate C-C motif chemokine receptor type 5 (CCR5) prognosis value in BC and develop a radiomics model for noninvasive prediction of CCR5 expression in BC.MethodsA total of 840 cases with genomic information were included and divided into CCR5 high- and low-expression groups for clinical characteristic differences exploration. Bioinformatics and survival analysis including Kaplan-Meier (KM) survival analysis, Cox regression, immunoinfiltration analysis, and tumor mutation load (TMB) were performed. For radiomics model development, 98 cases with dynamic contrast-enhancement magnetic resonance imaging (DCE-MRI) scans were used. Radiomics features extracted were using Pyradiomics and filtered by maximum-relevance minimum-redundancy (mRMR) and recursive feature elimination (REF) algorithms. Support vector machine (SVM) and logistic regression (LR) models were developed to predict CCR5 expression, with the radiomics score (Rad_score) representing the predicted probability of CCR5 expression. The models' performance was compared using the Delong test, and the model with the superior area under the curve (AUC) values was selected to analyze the correlation between CCR5 expression, Rad_score, and immune genes.ResultsThe CCR5 high-expression group exhibited better overall survival (OS) (p < 0.01). Six radiomics features were selected for model development. The AUCs of the SVM model predicting CCR5 were 0.753 and 0.748 in the training and validation sets, respectively, while the AUCs of the LR model were 0.763 and 0.762. Calibration curves and decision curve analysis (DCA) validated the models' calibration and clinical utility. The SVM_Rad_score showed a strong association with immune-related genes.ConclusionsThe DCE-MRI radiomics model presents a novel, non-invasive tool for predicting CCR5 expression in BC and provides valuable insights to inform clinical decision-making.
Collapse
Affiliation(s)
- Qingfeng Li
- First Clinical Medical College, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Cancer, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenting Li
- First Clinical Medical College, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Cancer, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianliang Wang
- Department of Radiology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Xiangyuan Li
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Yi Ji
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mianhua Wu
- First Clinical Medical College, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Cancer, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
4
|
Shenasa E, He Y, Wang Z, Tu D, Gao D, Kos Z, Thornton S, Nielsen TO. Digital Profiling of Immune Biomarkers in Breast Cancer: Relation to Anthracycline Benefit. Mod Pathol 2025; 38:100718. [PMID: 39863112 DOI: 10.1016/j.modpat.2025.100718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Assessment of the tumor-immune microenvironment can be used as a prognostic tool for improved survival and as a predictive biomarker for treatment benefit, particularly from immune-modulating treatments including cytotoxic chemotherapy. Using digital spatial profiling (DSP), we studied the tumor-immune microenvironment of 522 breast cancer cases by quantifying 35 immune biomarkers on tissue microarrays from the MA.5 phase III clinical trial. In this trial, node-positive breast cancer patients were randomized to receive either non-anthracycline chemotherapy (cyclophosphamide, methotrexate, 5'-fluorouracil [CMF]) or anthracycline-containing cytotoxic chemotherapy (CEF). Donor block hematoxylin and eosin (H&E)-stained sections were scored for the level of stromal tumor-infiltrating lymphocytes (sTILs), according to the international guidelines. We hypothesized that patients with higher levels of tumor-immune infiltration, assessed by either DSP or H&E staining, would benefit from CEF (relative to CMF) more than patients with lower immune infiltration. Unsupervised hierarchical clustering of digitally scored biomarkers revealed 2 patient clusters: immune infiltrated versus ignored. Following a prespecified statistical plan crafted to meet REMARK (REporting recommendations for tumor MARKer prognostic studies) guidelines, we found that the DSP-derived Immune Cluster assignment did not predict an improved 10-year relapse-free survival for patients receiving CEF compared with CMF. However, a secondary hypothesis revealed a significant predictive value for H&E sTILs assessed on full-faced sections for CEF benefit over CMF in the entire cohort and the human epidermal growth factor receptor 2-enriched subset. As exploratory analyses, supervised clustering of DSP-scored biomarkers suggested that low levels of T-cell immunoglobulin and mucin domain 3 TIM-3 and high levels of human leukocyte antigen HLA-DR and programmed cell death protein ligand PD-L-1 are associated with sensitivity to CEF. Although novel high-plex techniques provide a detailed insight into the tumor microenvironment, conventional H&E staining remains a powerful tool that can be applied to full-faced sections to assess the value of the immune microenvironment, particularly sTILs, in predicting benefits from immunogenic chemotherapies.
Collapse
Affiliation(s)
- Elahe Shenasa
- Interdisciplinary Oncology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ye He
- Visual Computing and Virtual Reality Key Laboratory of Sichuan Province, Sichuan Normal University, Sichuan, China
| | - Zehui Wang
- Mathematics and Statistics, Queen's University, Kingston, Ontario, Canada
| | - Dongsheng Tu
- Community Health & Epidemiology, Queen's University, Kingston, Ontario, Canada
| | - Dongxia Gao
- MAPcore, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zuzana Kos
- Pathology, BC Cancer Vancouver Centre, Vancouver, British Columbia, Canada
| | - Shelby Thornton
- MAPcore, University of British Columbia, Vancouver, British Columbia, Canada
| | - Torsten O Nielsen
- Interdisciplinary Oncology, University of British Columbia, Vancouver, British Columbia, Canada; MAPcore, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
5
|
Cha YJ, O'Connell CE, Calhoun BC, Felsheim BM, Fernandez-Martinez A, Fan C, Brueffer C, Larsson C, Borg Å, Saal LH, Perou CM. Genomic Characteristics Related to Histology-Based Immune Features in Breast Cancer. Mod Pathol 2025; 38:100736. [PMID: 39956271 PMCID: PMC12103273 DOI: 10.1016/j.modpat.2025.100736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
The immune cell component of the tumor microenvironment is an important modulator of tumor progression. In patients with breast cancer, tumor-infiltrating lymphocytes (TILs) and tertiary lymphoid structures (TLS) represent core aspects of antitumor immunity, both increasingly recognized for clinical relevance. In this study, we evaluated immune-related histology features using whole-slide hematoxylin and eosin (H&E) images of The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) data set (n = 1035) and analyzed these distinct features relative to gene expression, PAM50 subtypes, and patient survival. H&E images were evaluated for TILs, plasma cells (PCs), high-endothelial venule-associated lymphoid aggregates (HALA), and mature TLS. For HALA and TLS, location relative to the tumor (nontumor, peritumor, and intratumor) was determined. HER2-enriched (HER2E) and basal-like breast tumors exhibited the highest mean TILs and the presence of PCs. HALA were present in 35.1% of cases and TLS in 6.5% of cases, also predominantly in HER2E and basal-like tumors. We derived gene expression signatures for 10 histologically defined immune features and tested their clinical significance using transcriptomic and survival data from the Sweden Cancerome Analysis Network - Breast (SCAN-B) cohort. Signatures related to TILs, PCs, HALA/TLS, TLS, and specifically intratumor HALA and TLS were associated with better survival in HER2E and basal-like tumors. Peritumor HALA/TLS and nontumor signatures were nonsignificant or associated with worse outcomes. Furthermore, we compared the immune microenvironment of high-TIL (TILs > 10%) tumors from TCGA-BRCA by PAM50 subtype through supervised analyses of 200+ immune gene expression signatures, and unique immune features were identified for each subtype. In high-TIL luminal tumors, enriched immune signatures had little relation to prognosis. High-TIL HER2E and basal-like tumors had distinct immune signatures linked to improved survival, related to B and T cells, respectively. Overall, PAM50 subtypes of breast cancer exhibit distinct immune microenvironments, both histologically and molecularly. These differences in immune properties should be considered when developing precise treatment strategies to achieve optimal therapeutic efficacy for patients.
Collapse
Affiliation(s)
- Yoon Jin Cha
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Constandina E O'Connell
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Benjamin C Calhoun
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Brooke M Felsheim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Bioinformatics and Computational Biology Curriculum, Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Aranzazu Fernandez-Martinez
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France; Inserm, Gustave Roussy Cancer Campus, UMR981, Villejuif, France
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christian Brueffer
- Division of Oncology, Department of Clinical Sciences, Lund University, Sweden; Lund University Cancer Center, Lund University, Sweden; Skåne University Hospital Comprehensive Cancer Center, Lund, Sweden
| | - Christer Larsson
- Lund University Cancer Center, Lund University, Sweden; Skåne University Hospital Comprehensive Cancer Center, Lund, Sweden; Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Sweden
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences, Lund University, Sweden; Lund University Cancer Center, Lund University, Sweden; Skåne University Hospital Comprehensive Cancer Center, Lund, Sweden
| | - Lao H Saal
- Division of Oncology, Department of Clinical Sciences, Lund University, Sweden; Lund University Cancer Center, Lund University, Sweden; Skåne University Hospital Comprehensive Cancer Center, Lund, Sweden
| | - Charles M Perou
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
6
|
Kınıkoğlu O, Altıntaş YE, Yıldız A, Akdağ G, Bal H, Yaşar ZY, Özkerim U, Yıldız HŞ, Öksüz S, Tünbekici S, Doğan A, Işık D, Yaşar A, Başoğlu T, Sürmeli H, Odabaş H, Turan N. Tumor-infiltrating lymphocytes as predictive biomarkers in neoadjuvant treatment of HER2-positive breast cancer. Oncologist 2025; 30:oyaf054. [PMID: 40271640 PMCID: PMC12019226 DOI: 10.1093/oncolo/oyaf054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/19/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs) have emerged as predictive biomarkers in HER2-positive breast cancer, correlating with treatment response and survival outcomes. This study evaluates the impact of TIL levels and Ki67 suppression on neoadjuvant therapy efficacy in this patient population. MATERIALS AND METHODS A retrospective analysis of 136 HER2-positive breast cancer patients was conducted. Patients were stratified by TIL levels, and clinical outcomes, including Ki67 expression, pathological complete response (pCR), and disease-free survival (DFS), were assessed. RESULTS High TIL levels (≥ 40%) were significantly associated with higher pCR rates (60.32% vs. 39.73%, P = .02) and with TIL ≥ 10% greater Ki67 suppression. In patients with low TIL levels, high Ki67 expression correlated with better pCR rates (57.1% vs 30.8%, P = 0.010), while in high TIL patients, no significant difference was observed between high and low Ki67 groups (P = 0.317). A trend toward improved DFS was noted in the high TIL group, with 3-year survival rates of 91.9% vs. 80.7% in the low TIL group, though this was not statistically significant (P = .062). CONCLUSION TIL levels are robust predictors of pCR and Ki67 suppression in HER2-positive breast cancer, particularly in patients with high initial TILs. These findings highlight the potential for integrating TIL evaluation into personalized treatment strategies to optimize neoadjuvant therapy outcomes. Further research is warranted to validate these results and explore underlying mechanisms.
Collapse
Affiliation(s)
- Oğuzcan Kınıkoğlu
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Yunus Emre Altıntaş
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Anıl Yıldız
- Istanbul University Oncology Institute, Department of Medical Oncology, Istanbul, Turkey
| | - Goncagül Akdağ
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Hamit Bal
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Zeynep Yüksel Yaşar
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Uğur Özkerim
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Hacer Şahika Yıldız
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Sıla Öksüz
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Salih Tünbekici
- Ege University Faculty of Medicine, Department of Medical Oncology, Izmir, Turkey
| | - Akif Doğan
- Sancaktepe Şehit Prof. Dr. İlhan Varank City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Deniz Işık
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Alper Yaşar
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Tuğba Başoğlu
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Heves Sürmeli
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Hatice Odabaş
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| | - Nedim Turan
- Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Department of Medical Oncology, Istanbul, Turkey
| |
Collapse
|
7
|
Ren X, Zhou T, Song Y, Wu H, Chou J, Miller LD, Liang Z, Shen S. Intrinsic subtype and immunity score in identification of triple-negative breast cancer at low risk. Breast 2025; 80:103889. [PMID: 39908962 PMCID: PMC11847028 DOI: 10.1016/j.breast.2025.103889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 12/15/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is group of heterogeneity caner. Despite majority of them had the unfavorable prognosis, a subset of patients who do not receive chemotherapy exhibit a good prognosis. Biomarkers are required to recognize these group of pateints and improve the current therapeutic strategies for them. METHODS A retrospective analysis of 997 patients with TNBC from three datasets including 188 case of Peking Union Medical College Hospital (PUMCH) and two TNBC datasets from published cohort studies(279 case of Affy-set, 530 case of GSE set) was conducted. Intrinsic subtypes (basal-like, immune-enhanced, human epidermal growth factor receptor-2 [HER2]-enriched and luminal A/B) and tumor environmental immunity were evaluated using expression profiles of a 72-gene panel. Association of intrinsic subtype and immunity score with distant metastasis-free survival (DMFS) and overall survival (OS) was analyzed. RESULTS Five intrinsic subtypes were identified in the patients with TNBC, comprising 64 % basal-like, 19 % immune-enhanced, 11 % HER2-enriched, 5 % luminal A, and 2 % luminal B. In the absence of adjuvant chemotherapy (ACT), Luminal A and immune-enhanced subtypes showed better DMFS than basal-like, HER2-enriched, and luminal B subtypes(P = 0.35). Significantly good OS was observed in luminal A and immune-enhanced subtypes compared to basal-like and HER2-enriched subtypes (P < 0.05). So two subtype groups were further classified as low-risk subtypes, including luminal A and immune-enhanced, and high-risk subtypes, including basal-like, HER2-enriched, and luminal B. Except for the immune-enhanced subtype, each subtype was further sorted and grouped according to immunity score, istrong and iweak. Significant improvements in both DMFS and OS were observed in patients with istrong compared with those with iweak(P = 0.01 and 0.0051 respectively). When combining intrinsic subtype and immunity status to predict the benefit from ACT, all high-risk subtype patients demonstrated improved DMFS(P = 0.075) and OS(P < 0.0001), with istrong patients exhibiting greater benefit; low-risk subtype plus iweak patients showed marginal benefit, whereas low-risk subtype plus istrong patients demonstrated least benefit from ACT. CONCLUSION Intrinsic subtype and immunity score is good prognostic biomarkers for patients with TNBC in the absence of chemotherapy. Combined intrinsic subtype and immunity evaluation could identify patients with TNBC who do not benefit from chemotherapy.
Collapse
Affiliation(s)
- Xinyu Ren
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tong Zhou
- Department of Oncology, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Yu Song
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huanwen Wu
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jeff Chou
- Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Lance D Miller
- Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA; Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Blvd, Winston Salem, NC, 27157, USA
| | - Zhiyong Liang
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Songjie Shen
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Fernandez-Martinez A, Tanioka M, Ahn SG, Zagami P, Pascual T, Rediti M, Tang G, Hoadley KA, Venet D, Rashid NU, Spears PA, Di Cosimo S, de Azambuja E, Choudhury A, Rastogi P, Islam MN, Cortes J, Llombart-Cussac A, Swain SM, Sotiriou C, Prat A, Perou CM, Carey LA. Prognostic value of residual disease (RD) biology and gene expression changes during the neoadjuvant treatment in patients with HER2-positive early breast cancer (EBC). Ann Oncol 2025; 36:403-413. [PMID: 39706338 PMCID: PMC11949722 DOI: 10.1016/j.annonc.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND In human epidermal growth factor receptor 2 (HER2)-positive early breast cancer (EBC), we investigated tumor and immune changes during neoadjuvant treatment and their impact on residual disease (RD) biology and prognostic implications across four neoadjuvant studies of trastuzumab with or without lapatinib, and with or without chemotherapy: CALGB 40601, PAMELA, NeoALTTO, and NSABP B-41. PATIENTS AND METHODS We compared tumor and immune gene expression changes during neoadjuvant treatment and their association with event-free survival (EFS) by uni- and multivariable Cox regression models in different cohorts and timepoints: 452 RD samples at baseline including 169 with a paired RD, and biomarker changes during neoadjuvant therapy, evaluating model performance via the c-index. RESULTS Analysis of 169 paired tumor samples revealed a shift in intrinsic subtype proportions from HER2-enriched at baseline (50.3%) to normal-like (49.1%) followed by luminal A (18.9%) in RD. This luminal phenotypic change was supported by decreased correlation to the HER2-enriched centroid, ERBB2, and HER2 amplicon genes and increased correlation to the luminal A centroid (Wilcoxon test P < 0.001). Additionally, RD showed relative immune activation marked by significant increases in B-cell, CD8 T-cell, and natural killer cell signatures (Wilcoxon test P < 0.05). In multivariable Cox models, intrinsic subtypes at baseline provided more prognostic information, while immune gene expression signatures provided more prognostic information in RD. Notably, the best multivariable EFS model (c-index = 0.77) integrated the immunoglobulin G signature from RD samples (adjusted hazard ratio 0.45, 95% confidence interval 0.30-0.67, adjusted P = 0.002). CONCLUSIONS In patients with HER2-positive EBC and RD, tumor biomarkers provide more prognostic information at baseline. In contrast, immune biomarkers perform better for EFS prognosis in RD.
Collapse
Affiliation(s)
- A Fernandez-Martinez
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, USA; Department of Genetics, University of North Carolina, Chapel Hill, USA
| | - M Tanioka
- Okayama University, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Medical AI Project, Okayama, Japan
| | - S G Ahn
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - P Zagami
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, USA; University of Milan, Milan, Italy
| | - T Pascual
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; SOLTI Breast Cancer Cooperative Group, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - M Rediti
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - G Tang
- National Surgical Adjuvant Breast and Bowel Project (NSABP), Pittsburgh, USA; Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, USA
| | - K A Hoadley
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, USA; Department of Genetics, University of North Carolina, Chapel Hill, USA
| | - D Venet
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - N U Rashid
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, USA; Department of Biostatistics, University of North Carolina, Chapel Hill, USA
| | - P A Spears
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, USA
| | - S Di Cosimo
- Integrated Biology Platform, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - E de Azambuja
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), Institut Jules Bordet, Service d'Oncologie Médicale, Brussels, Belgium
| | - A Choudhury
- FTE of Novartis Healthcare Pvt. Ltd., Hyderabad, India
| | - P Rastogi
- National Surgical Adjuvant Breast and Bowel Project (NSABP), Pittsburgh, USA; UPMC Hillman Cancer Center, Pittsburgh, USA
| | - M N Islam
- Genomics and Epigenomics Shared Resource (GESR), Georgetown University Medical Center, Washington, USA
| | - J Cortes
- Oncology Department, International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Barcelona, Spain; IOB Madrid, Institute of Oncology, Hospital Beata Maria Ana, Madrid, Spain; Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Department of Medicine, Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Madrid, Spain
| | - A Llombart-Cussac
- Medical Oncology Department, Hospital Arnau de Vilanova, Valencia, Spain
| | - S M Swain
- National Surgical Adjuvant Breast and Bowel Project (NSABP), Pittsburgh, USA; Lombardi Comprehensive Cancer Center, Georgetown University Medical Center and MedStar Health, Washington, USA
| | - C Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - A Prat
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - C M Perou
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, USA; Department of Genetics, University of North Carolina, Chapel Hill, USA
| | - L A Carey
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, USA; Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, USA.
| |
Collapse
|
9
|
Hennessy MA, Cimino-Mathews A, Carter JM, Kachergus JM, Ma Y, Leal JP, Solnes LB, Denbow R, Abramson VG, Carey LA, Rimawi M, Specht J, Storniolo AM, Valero V, Vaklavas C, Winer EP, Krop IE, Wolff AC, Wahl RL, Perez EA, Huang CY, Stearns V, Thompson EA, Connolly RM. Multiplex Spatial Proteomic Analysis of HER2-Positive Breast Tumors Reveals Unique Molecular and Immunologic Features Associated With Treatment Response. JCO Precis Oncol 2025; 9:e2400546. [PMID: 40179327 PMCID: PMC11968088 DOI: 10.1200/po-24-00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/17/2024] [Accepted: 02/21/2025] [Indexed: 04/05/2025] Open
Abstract
PURPOSE Predictive biomarkers to better tailor therapy for patients with early-stage human epidermal growth factor 2 (HER2)-positive breast cancer are a priority. We hypothesized that HER2 and immune-based biomarkers would be predictive of pathologic complete response (pCR) to preoperative trastuzumab/pertuzumab (HP). MATERIALS AND METHODS Patients with stage II/III, estrogen receptor (ER)-negative, HER2-positive breast cancer received neoadjuvant HP in the TBCRC026 clinical trial. The pCR after receiving HP alone was 22% (18/83). Tumor biopsies were performed at baseline. Secondary correlative objectives were to determine the relationship between HER2-based biomarkers and immune processes with pCR. NanoString code sets BC360 and IO360 were used to compare differential gene expression in baseline tumors that underwent pCR versus no pCR. NanoString GeoMx digital spatial profiling was used to assess immune protein abundance in intraepithelial and stromal segments. Stromal tumor-infiltrating lymphocytes and Ki67 were evaluated by hematoxylin and eosin and immunohistochemistry, respectively. RESULTS Intraepithelial HER2 protein abundance was significantly associated with pCR (P = .001). Low HER2 abundance tumors were primarily basal-like, and essentially all (19/20) failed to achieve pCR. High HER2 abundance tumors that achieved pCR (14/51) exhibited a high degree of immune cell activity, whereas high HER2 abundance tumors that failed to achieve pCR tumors (37/51) were enriched for M-phase processes and epidermal growth factor receptor signaling. Baseline Ki67 was significantly higher in nonresponders (P = .04). CONCLUSION ER-negative, HER2-positive breast cancer has unique molecular and immunologic features that may predict pCR after neoadjuvant HP. Validation of these potential biomarkers and composite biomarker analyses may guide design of future clinical trials.
Collapse
Affiliation(s)
| | - Ashley Cimino-Mathews
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Jodi M. Carter
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | | | - Yaohua Ma
- Department of Quantitative Health Science, Mayo Clinic Florida, Jacksonville, FL
| | - Jeffrey P. Leal
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Lilja B. Solnes
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Rita Denbow
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | | | | | - Anna Maria Storniolo
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN
| | - Vicente Valero
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | - Antonio C. Wolff
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | | | - Edith A. Perez
- Department of Cancer Biology, Mayo Clinic Florida, Jacksonville, FL
| | - Chiung-Yu Huang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
- University of California, San Francisco, CA
| | - Vered Stearns
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | | | - Roisin M. Connolly
- Cancer Research @UCC, University College Cork, Cork, Ireland
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
10
|
Dieci MV, Bisagni G, Bartolini S, Schirone A, Cavanna L, Musolino A, Giotta F, Rimanti A, Garrone O, Bertone E, Cagossi K, Sarti S, Ferro A, Piacentini F, Orvieto E, Sanders M, Miglietta F, Massa D, Balduzzi S, Conte P, D’Amico R, Guarneri V. Tumor-Infiltrating Lymphocytes and Survival Outcomes in Early ERBB2-Positive Breast Cancer: 10-Year Analysis of the ShortHER Randomized Clinical Trial. JAMA Oncol 2025; 11:386-393. [PMID: 39946142 PMCID: PMC11826437 DOI: 10.1001/jamaoncol.2024.6872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/10/2024] [Indexed: 02/16/2025]
Abstract
Importance For patients with early ERBB2 (formerly HER2)-positive breast cancer, there is a need to identify biomarkers to guide treatment de-escalation. Objective To evaluate the association of tumor-infiltrating lymphocytes (TILs) with distant disease-free (DDFS) and overall survival (OS) for patients with ERBB2-positive early breast cancer. Design, Setting, and Participants The ShortHER randomized clinical trial was a multicentric trial in Italy that enrolled patients with ERBB2-positive breast cancer from December 2007 to October 2013. Patients received 9 weeks or 1 year of adjuvant trastuzumab combined with chemotherapy. Tumor samples were evaluated for TILs. Herein, patients were evaluated at a median follow-up of 9 years, and data were analyzed from February 2023 to August 2024. Intervention Four cycles of anthracycline-based chemotherapy followed by 4 courses of taxanes combined with trastuzumab for 1 year (long arm) or 3 courses of taxanes combined with trastuzumab for 9 weeks followed by reduced-dose anthracycline-based chemotherapy for 3 courses (short arm). Main Outcomes and Measures The association of TILs with DDFS and OS was assessed with Cox models. Results Of 1253 patients enrolled in the ShortHER trial, 866 women (median [IQR] age, 56 [48-64] years) had evaluable TILs. In Cox models with relevant factors, each 5% TIL increment was associated with improved DDFS (hazard ratio [HR], 0.87; 95% CI, 0.80-0.95; P = .001) and OS (HR, 0.89; 95% CI, 0.81-0.98; P = .01). The 10-year OS rate was 91.3% for patients with TILs 20% or higher, 93.3% for patients with TILs 30% or higher, and 98.1% for patients with TILs 50% or higher, resulting higher vs lower TIL counterparts. Patients with TILs lower than 20% showed a better outcome with the long vs short treatment (10-year DDFS, 88.7% vs 81.0%), whereas patients with TILs 20% or higher showed the opposite (10-year DDFS, 87.1% vs 92.2%; P for interaction = .01). Similarly, patients with TILs 20% or higher had a 10-year OS rate of 89.3% in the long arm vs 93.1% in the short arm (HR, 0.36; 95% CI, 0.10-1.36); patients with TILs lower than 20% had a 10-year OS rate of 91.3% in the long arm vs 86.9% in the short arm (HR, 1.36; 95% CI, 0.82-2.23; P for interaction = .06). Conclusions and Relevance This follow-up analysis of the ShortHER randomized clinical trial is, to our knowledge, the first demonstration of an independent effect of TILs in terms of OS for patients with ERBB2-positive early breast cancer treated with adjuvant chemotherapy and anti-ERBB2 therapy. Patients with TILs 20% or higher who de-escalated trastuzumab duration and chemotherapy dose were not exposed to an excess risk of distant relapse or death. Trial Registration EudraCT: 2007-004326-25.
Collapse
Affiliation(s)
- Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Giancarlo Bisagni
- Department of Oncology and Advanced Technologies, Azienda USL-IRCCS, Reggio Emilia, Italy
| | - Stefania Bartolini
- Department of Nervous System Medical Oncology, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | | | - Luigi Cavanna
- Internal Medicine and Oncology, Clinica Piacenza, Piacenza, Italy
| | - Antonino Musolino
- Medical Oncology, Breast and GYN Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori,” Meldola, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | | | - Ornella Garrone
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
| | | | - Katia Cagossi
- Breast Unit Ausl Modena, Ramazzini Hospital, Carpi, Italy
| | - Samanta Sarti
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori,” Meldola, Italy
| | - Antonella Ferro
- Rete Clinica Senologica-Oncologia Medica S. Chiara, APSS, Trento, Italy
| | - Federico Piacentini
- Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Modena, Italy
- Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | | | - Melinda Sanders
- Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Davide Massa
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Sara Balduzzi
- University of Modena and Reggio Emilia, Modena, Italy
| | - Pierfranco Conte
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - Roberto D’Amico
- Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Modena, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| |
Collapse
|
11
|
Mubarak O, Middleton GW. The impact of the immunological context on outcomes of solid cancer patients treated with genotype-matched targeted therapies: a comprehensive review. Ann Oncol 2025:S0923-7534(25)00113-9. [PMID: 40118150 DOI: 10.1016/j.annonc.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Outcomes with genotype-matched targeted therapy in solid cancer patients are heterogeneous: some have exceptional responses, whereas others have primary progression. This review explores the immunobiological features which may underlie this differential response. METHODS We conducted a literature review of studies assessing the impact of immune context following searches on Web of Science, Medline and Embase. Relevant outcomes include response, progression-free survival and overall survival. Data were extracted from multivariate analysis, univariate analysis or directly from Kaplan-Meier curves. Meta-analyses were carried out where three or more studies analysed the same immune factor for the same cancer type. The remaining studies were analysed descriptively. RESULTS In the adjuvant setting, assessment of the immune context does not highlight a group failing to derive benefit for the use of dabrafenib/trametinib after resection of BRAFV600E melanoma. Differential gene expression in exceptional responders show enrichment of genes associated with immune activation. BRAFV600E colorectal cancer patients with high cytolytic scores benefit from the addition of MEK inhibition whereas those with low scores fare better without. High programmed death-ligand 1 (PD-L1) expression is predictive of inferior outcomes to epidermal growth factor receptor (EGFR), ALK and G12C tyrosine kinase inhibitors. EGFR-mutant patients with high CD8+ T cells and PD-L1 positivity have very poor outcomes. Stromal tumour-infiltrating lymphocytes predict for efficacy of stromal-poor tumours in human epidermal growth factor receptor 2 (HER2)-positive breast cancer treated with short-course adjuvant trastuzumab. High immune metagene and single immune gene expression are predictive of benefit for chemotherapy plus trastuzumab, but not chemotherapy alone. The addition of pertuzumab or lapatanib appears to be beneficial in those with immune non-enriched microenvironments. High major histocompatibility complex (MHC)-I is negatively predictive and high MHC-II is positively predictive of outcomes with trastuzumab-based therapy. CONCLUSIONS To our knowledge, this is the first review assessing immunological context as a biomarker for targeted therapy. The results of this review represent an important resource to aid future translational studies in advancing stratified precision medicine oncology.
Collapse
Affiliation(s)
- O Mubarak
- Department of Immunology and Immunotherapy, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - G W Middleton
- Department of Immunology and Immunotherapy, College of Medicine and Health, University of Birmingham, Birmingham, UK; University Hospitals Birmingham, Birmingham, UK.
| |
Collapse
|
12
|
Martín M, Stecklein SR, Gluz O, Villacampa G, Monte-Millán M, Nitz U, Cobo S, Christgen M, Brasó-Maristany F, Álvarez EL, Echavarría I, Conte B, Kuemmel S, Bueno-Muiño C, Jerez Y, Kates R, Cebollero M, Kolberg-Liedtke C, Bueno O, García-Saenz JÁ, Moreno F, Grischke EM, Forstbauer H, Braun M, Warm M, Hackmann J, Uleer C, Aktas B, Schumacher C, Wuerstleins R, Graeser M, Zu Eulenburg C, Kreipe HH, Gómez H, Massarrah T, Herrero B, Paré L, Bohn U, López-Tarruella S, Vivancos A, Sanfeliu E, Parker JS, Perou CM, Villagrasa P, Prat A, Sharma P, Harbeck N. TNBC-DX genomic test in early-stage triple-negative breast cancer treated with neoadjuvant taxane-based therapy. Ann Oncol 2025; 36:158-171. [PMID: 39419289 DOI: 10.1016/j.annonc.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/28/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Identification of biomarkers to optimize treatment strategies for early-stage triple-negative breast cancer (TNBC) is crucial. This study presents the development and validation of TNBC-DX, a novel test aimed at predicting both short- and long-term outcomes in early-stage TNBC. The objective of this study was to evaluate the association between TNBC-DX and efficacy outcomes [pathologic complete response (pCR), distant disease-free survival (DDFS) or event-free survival (EFS), and overall survival (OS)] in the validation cohorts. METHODS Information from 1259 patients with early-stage TNBC (SCAN-B, CALGB-40603, and BrighTNess) was used to establish the TNBC-DX scores. Independent validation of TNBC-DX was carried out in three studies: (i) WSG-ADAPT-TN; (ii) MMJ-CAR-2014-01; and (iii) NeoPACT, including 527 patients with stage I-III TNBC undergoing neoadjuvant chemotherapy. In WSG-ADAPT-TN, patients were randomized to receive nab-paclitaxel plus gemcitabine or carboplatin. In MMJ-CAR-2014-01, patients received carboplatin plus docetaxel. In NeoPACT, patients received carboplatin plus docetaxel and pembrolizumab. RESULTS TNBC-DX test was created incorporating the 10-gene Core Immune Gene module, the 4-gene tumor cell proliferation signature, tumor size, and nodal staging. In the two independent validation cohorts without pembrolizumab, the TNBC-DX pCR score was significantly associated with pCR after adjustment for clinicopathological variables and treatment regimen [odds ratio per 10-unit increment 1.34, 95% confidence interval (CI) 1.20-1.52, P < 0.001]. pCR rates for the TNBC-DX pCR-high, pCR-medium, and pCR-low categories were 56.3%, 53.6%, and 22.5% respectively (odds ratio for pCR-high versus pCR-low 3.48, 95% CI 1.72-7.15, P < 0.001). In addition, the TNBC-DX risk score was significantly associated with DDFS [hazard ratio (HR) high-risk versus low-risk 0.24, 95% CI 0.15-0.41, P < 0.001] and OS (HR 0.19, 95% CI 0.11-0.35, P < 0.001). In the validation cohort with pembrolizumab, the TNBC-DX scores were significantly associated with pCR, EFS, and OS. CONCLUSIONS TNBC-DX predicts pCR to neoadjuvant taxane-carboplatin in stage I-III TNBC and helps to forecast the patient's long-term survival in the absence of neoadjuvant anthracycline-cyclophosphamide, and independent of pembrolizumab use.
Collapse
Affiliation(s)
- M Martín
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Grupo Español de Investigación en Cáncer de Mama, Madrid, Spain; Universidad Complutense de Madrid, Madrid, Spain
| | - S R Stecklein
- Department of Internal Medicine, University of Kansas Medical Center, Westwood; Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, USA; Departments of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, USA; Departments of Cancer Biology, University of Kansas Medical Center, Kansas City, USA
| | - O Gluz
- West German Study Group, Monchengladbach, Germany; Breast Center Niederrhein, Ev. Hospital Bethesda, Moenchengladbach, Germany; University Clinics Cologne, Cologne, Germany
| | - G Villacampa
- SOLTI Cancer Research Group, Barcelona, Spain; Statistics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - M Monte-Millán
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - U Nitz
- West German Study Group, Monchengladbach, Germany; Breast Center Niederrhein, Ev. Hospital Bethesda, Moenchengladbach, Germany
| | - S Cobo
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - M Christgen
- University Clinics Cologne, Cologne, Germany
| | - F Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Reveal Genomics, Barcelona, Spain
| | - E L Álvarez
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - I Echavarría
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - B Conte
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - S Kuemmel
- Medical School Hannover, Institute of Pathology, Hannover; Breast Unit, Clinics Essen Mitte, Breast Center, Essen, Germany
| | - C Bueno-Muiño
- Medical Oncology Department, Hospital Infanta Cristina (Parla), Madrid, Spain
| | - Y Jerez
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - R Kates
- West German Study Group, Monchengladbach, Germany
| | - M Cebollero
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - C Kolberg-Liedtke
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - O Bueno
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - J Á García-Saenz
- Grupo Español de Investigación en Cáncer de Mama, Madrid, Spain; Department of Medical Oncology, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), Madrid, Spain
| | - F Moreno
- Grupo Español de Investigación en Cáncer de Mama, Madrid, Spain; Department of Medical Oncology, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), Madrid, Spain
| | - E-M Grischke
- Department of Gynecology, Women's Clinic, University Clinics Tuebingen, Tuebingen, Germany
| | | | - M Braun
- Breast Center, Rotkreuz Clinics Munich, Munich, Germany
| | - M Warm
- Breast Center, City Hospital Holweide, Cologne, Germany
| | - J Hackmann
- Breast Center, Marien-Hospital, Witten, Germany
| | - C Uleer
- Practice of Gynecology and Oncology, Hildesheim, Germany
| | - B Aktas
- Women's Clinic, University Clinics Essen, Essen, Germany
| | - C Schumacher
- Breast Center, St. Elisabeth Hospital, Cologne, Germany
| | - R Wuerstleins
- West German Study Group, Monchengladbach, Germany; Breast Center, Department of Obstetrics and Gynecology and CCC Munich, LMU University Hospital, Munich, Germany
| | - M Graeser
- West German Study Group, Monchengladbach, Germany; Breast Center Niederrhein, Ev. Hospital Bethesda, Moenchengladbach, Germany; University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - C Zu Eulenburg
- West German Study Group, Monchengladbach, Germany; University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - H H Kreipe
- West German Study Group, Monchengladbach, Germany
| | - H Gómez
- Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - T Massarrah
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - B Herrero
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Grupo Español de Investigación en Cáncer de Mama, Madrid, Spain
| | - L Paré
- Reveal Genomics, Barcelona, Spain
| | - U Bohn
- Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas, Spain
| | - S López-Tarruella
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Grupo Español de Investigación en Cáncer de Mama, Madrid, Spain; Universidad Complutense de Madrid, Madrid, Spain
| | | | - E Sanfeliu
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Pathology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - J S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, USA
| | - C M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, USA
| | | | - A Prat
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Reveal Genomics, Barcelona, Spain; Cancer Institute and Blood Disorders, Hospital Clínic de Barcelona, Barcelona, Spain; Medicine Department, University of Barcelona, Barcelona, Spain; Breast Cancer Unit, IOB-QuirónSalud, Barcelona, Spain.
| | - P Sharma
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, USA
| | - N Harbeck
- West German Study Group, Monchengladbach, Germany; Breast Center, Department of Obstetrics and Gynecology and CCC Munich, LMU University Hospital, Munich, Germany
| |
Collapse
|
13
|
Schlam I, Loi S, Salgado R, Swain SM. Tumor-infiltrating lymphocytes in HER2-positive breast cancer: potential impact and challenges. ESMO Open 2025; 10:104120. [PMID: 39826475 PMCID: PMC11786075 DOI: 10.1016/j.esmoop.2024.104120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION In this review, we evaluate the role of stromal tumor-infiltrating lymphocytes (sTILs) as a biomarker in human epidermal growth factor receptor 2 (HER2)-positive breast cancer, exploring the prognostic and predictive potential in various treatment settings. METHODS Data from multiple clinical trials in the early and metastatic settings, focusing on TILs' correlation with pathologic complete response (pCR), progression-free survival (PFS), and overall survival across early and metastatic HER2-positive breast cancer were summarized. This review also discusses TILs' assessment methods, interobserver variability, and emerging technologies to assess TILs. RESULTS TILs have been identified as a highly reproducible biomarker that predicts pCR in patients receiving neoadjuvant therapy and serves as a prognostic indicator for long-term outcomes in several breast cancer subtypes, including HER2-positive. Studies indicate that higher TIL levels correlate with better recurrence-free survival rates. Despite these findings, there is no consensus on the optimal TIL threshold for clinical decision making, and further research is required on how to incorporate TILs into routine clinical practice. CONCLUSIONS TILs represent a promising biomarker in HER2-positive breast cancer, particularly in early disease settings. This assessment could guide treatment de-escalation or intensification, tailoring therapies to individual patient profiles. Due to their prognostic importance, TILs can be added to pathology reports. However, further validation in clinical trials is essential for the widespread adoption of TILs in clinical practice.
Collapse
Affiliation(s)
- I Schlam
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA. https://twitter.com/ilanaschlam
| | - S Loi
- Peter Mac Callum Cancer Centre, Melbourne, Victoria, Australia. https://twitter.com/LoiSher
| | - R Salgado
- Peter Mac Callum Cancer Centre, Melbourne, Victoria, Australia; ZAS-Hospitals, Antwerp, Belgium. https://twitter.com/TILsWorkGroup
| | - S M Swain
- Georgetown Lombardi Comprehensive Cancer Center, Washington, USA; MedStar Health, Columbia, Maryland, USA.
| |
Collapse
|
14
|
Cao F, Guo C, Guo J. Deciphering CSU pathogenesis: Network toxicologyand molecular dynamics of DOTP exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117864. [PMID: 39923559 DOI: 10.1016/j.ecoenv.2025.117864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/14/2025] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVE This study elucidated the molecular mechanisms underlying chronic spontaneous urticaria (CSU), potentially induced by the food and environmental pollutant dioctyl terephthalate (DOTP), through the application of network toxicology and molecular dynamics simulations. METHODS The structural analysis of DOTP was conducted in PubChem, with target prediction executed via Swiss Target Prediction and SuperPred, and target identification standardized using UniProt. A PPI network analysis identified core disease-related targets using STRING and Cytoscape. GO and KEGG analyses were utilized to explore target functions, particularly in inflammation and immune response pathways. AutoDock was employed for molecular docking to predict DOTP's binding to core targets, followed by molecular dynamics simulations with Gromacs to observe detailed interactions and conformational changes in the target proteins. RESULTS The study identified 38 potential targets associated with CSU and highlighted six core targets-EGFR, BCL2, NFKB1, CASP3, ERBB2, and mTOR-through PPI network analysis. GO and KEGG analyses illuminated the roles of these targets in biological processes, cellular components, molecular functions, and signaling pathways, with a particular emphasis on the PI3K-Akt signaling pathway. Molecular docking demonstrated strong binding affinities between DOTP and the core targets, while molecular dynamics simulations confirmed the stable binding of DOTP to these targets, with exceptional stability observed in its interaction with the mTOR protein. CONCLUSION This study clarifies the potential molecular mechanisms of DOTP-induced CSU and underscores the efficacy of network toxicology, molecular docking, and molecular dynamics simulations in assessing the toxicity of food and environmental pollutants and their related molecular biological mechanisms. These findings offer new insights for future research, enhance our comprehension of the potential health impacts of food and environmental pollutants, and establish a scientific foundation for the development of prevention and treatment strategies.
Collapse
Affiliation(s)
- Fang Cao
- Chengdu University of Traditional Chinese Medicine, China
| | - Cui Guo
- Chengdu University of Traditional Chinese Medicine, China
| | - Jing Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, China.
| |
Collapse
|
15
|
Angelats L, Paré L, Rubio-Perez C, Sanfeliu E, González A, Seguí E, Villacampa G, Marín-Aguilera M, Pernas S, Conte B, Albarrán-Fernández V, Martínez-Sáez O, Aguirre Á, Galván P, Fernandez-Martinez A, Cobo S, Rey M, Martínez-Romero A, Walbaum B, Schettini F, Vidal M, Buckingham W, Muñoz M, Adamo B, Agrawal Y, Guedan S, Pascual T, Agudo J, Grzelak M, Borcherding N, Heyn H, Vivancos A, Parker JS, Villagrasa P, Perou CM, Prat A, Brasó-Maristany F. Linking tumor immune infiltration to enhanced longevity in recurrence-free breast cancer. ESMO Open 2025; 10:104109. [PMID: 39765189 PMCID: PMC11758579 DOI: 10.1016/j.esmoop.2024.104109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The infiltration of tumor-infiltrating B cells and plasma cells in early-stage breast cancer has been associated with a reduced risk of distant metastasis. However, the influence of B-cell tumor infiltration on overall patient survival remains unclear. MATERIALS AND METHODS This study explored the relationship between an antitumor immune response, measured by a 14-gene B-cell/immunoglobulin (IGG) signature, and mortality risk in 9638 breast cancer patients across three datasets. Associations with tumor subtype, stage, and age were examined. IGG was characterized using spatial GeoMx profiling and single-cell RNA sequencing, and its relationship with tertiary lymphoid structures (TLSs) was evaluated. The predictive value of each of the 14 IGG genes for B-cell receptor (BCR) and T-cell receptor (TCR) clonality and longevity was also assessed, along with its association with longevity in other cancer types. RESULTS High IGG signature expression was significantly associated with a 41%-47% reduction in death risk in breast cancer survivors (P < 0.001), regardless of age, tumor stage, or subtype. Similar associations were observed in other cancers, including melanoma. In breast cancer, the IGG signature was significantly linked to overall survival without relapse in patients aged 41-70 years at diagnosis. Additionally, IGG expression correlated with the presence of TLSs and higher B- and T-cell polyclonality. A specific subset of seven IGG genes strongly correlated with BCR and TCR clonality, with predictive power for identifying clonality and improved longevity, especially when combining two of these genes. CONCLUSIONS This study uncovers a significant link between immune gene expression in tumors and extended longevity in breast cancer survivors, even in the absence of recurrence. The IGG signature, particularly its key gene subset, emerges as a powerful marker of sustained antitumor immunity and overall patient fitness. These findings pave the way for personalized treatment strategies that enhance both survival and long-term health outcomes.
Collapse
Affiliation(s)
- L Angelats
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - L Paré
- Reveal Genomics, Barcelona, Spain
| | - C Rubio-Perez
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - E Sanfeliu
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Pathology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - A González
- Department of Pathology, Hospital Clinic of Barcelona, Barcelona, Spain; Immunogenetics and Immunotherapy in Autoinflammatory and Immune Responses, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - E Seguí
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - G Villacampa
- Statistics Unit, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - S Pernas
- Institut Catala d'Oncologia, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; Institut Català d'Oncologia Medical Oncology, Breast Unit Barcelona, Barcelona, Spain
| | - B Conte
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - V Albarrán-Fernández
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - O Martínez-Sáez
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Á Aguirre
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - P Galván
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain; Reveal Genomics, Barcelona, Spain
| | | | - S Cobo
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - M Rey
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - A Martínez-Romero
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - B Walbaum
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - F Schettini
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - M Vidal
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | | | - M Muñoz
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - B Adamo
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Y Agrawal
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, USA
| | - S Guedan
- Cellular Immunotherapies for Cancer Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - T Pascual
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain; SOLTI Cancer Cooperative Group, Barcelona, Spain
| | - J Agudo
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, USA; Department of Immunology, Harvard Medical School, Boston, USA; Parker Institute for Cancer Immunotherapy at Dana-Farber Cancer Institute, Boston, USA; New York Stem Cell Foundation, Robertson Investigator, New York, USA
| | | | | | - H Heyn
- Omniscope Inc., Palo Alto, USA
| | - A Vivancos
- Vall d'Hebron Institute of Oncology (VHIO), Cancer Genomics Group, Barcelona, Spain
| | - J S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, USA
| | | | - C M Perou
- Department of Genetics, University of North Carolina, Chapel Hill, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, USA
| | - A Prat
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain; Reveal Genomics, Barcelona, Spain; Institute of Oncology (IOB)-Hospital Quirónsalud, Barcelona, Spain
| | - F Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona, Barcelona, Spain; Reveal Genomics, Barcelona, Spain.
| |
Collapse
|
16
|
Veeraraghavan J, De Angelis C, Gutierrez C, Liao FT, Sabotta C, Rimawi MF, Osborne CK, Schiff R. HER2-Positive Breast Cancer Treatment and Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:495-525. [PMID: 39821040 DOI: 10.1007/978-3-031-70875-6_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
HER2-positive (+) breast cancer is an aggressive disease with poor prognosis, a narrative that changed drastically with the advent and approval of trastuzumab, the first humanized monoclonal antibody targeting HER2. In addition to another monoclonal antibody, more classes of HER2-targeted agents, including tyrosine kinase inhibitors, and antibody-drug conjugates were developed in the years that followed. While these potent therapies have substantially improved the outcome of patients with HER2+ breast cancer, resistance has prevailed as a clinical challenge ever since the arrival of targeted agents. Efforts to develop new treatment regimens to treat/overcome resistance is futile without a primary understanding of the mechanistic underpinnings of resistance. Resistance could be attributed to mechanisms that are either specific to the tumor epithelial cells or those that emerge through changes in the tumor microenvironment. Reactivation of the HER receptor layer due to incomplete blockade of the HER receptor layer or due to alterations in the HER receptors is one of the major mechanisms. In other instances, resistance may occur due to deregulations in key downstream signaling such as the PI3K/AKT or RAS/MEK/ERK pathways or due to the emergence of compensatory pathways such as ER, other RTKs, or metabolic pathways. Potent new targeted agents and approaches to target key actionable drivers of resistance have already been identified, many of which are in early clinical development or under preclinical evaluation. Ongoing and future translational research will continue to uncover additional therapeutic vulnerabilities, as well as new targeted agents and approaches to treat and/or overcome anti-HER2 treatment resistance.
Collapse
Affiliation(s)
- Jamunarani Veeraraghavan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Carmine De Angelis
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Carolina Gutierrez
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Fu-Tien Liao
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Caroline Sabotta
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Mothaffar F Rimawi
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - C Kent Osborne
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
17
|
Wei J, Li W, Zhang P, Guo F, Liu M. Current trends in sensitizing immune checkpoint inhibitors for cancer treatment. Mol Cancer 2024; 23:279. [PMID: 39725966 DOI: 10.1186/s12943-024-02179-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically transformed the treatment landscape for various malignancies, achieving notable clinical outcomes across a wide range of indications. Despite these advances, resistance to immune checkpoint blockade (ICB) remains a critical clinical challenge, characterized by variable response rates and non-durable benefits. However, growing research into the complex intrinsic and extrinsic characteristics of tumors has advanced our understanding of the mechanisms behind ICI resistance, potentially improving treatment outcomes. Additionally, robust predictive biomarkers are crucial for optimizing patient selection and maximizing the efficacy of ICBs. Recent studies have emphasized that multiple rational combination strategies can overcome immune checkpoint resistance and enhance susceptibility to ICIs. These findings not only deepen our understanding of tumor biology but also reveal the unique mechanisms of action of sensitizing agents, extending clinical benefits in cancer immunotherapy. In this review, we will explore the underlying biology of ICIs, discuss the significance of the tumor immune microenvironment (TIME) and clinical predictive biomarkers, analyze the current mechanisms of resistance, and outline alternative combination strategies to enhance the effectiveness of ICIs, including personalized strategies for sensitizing tumors to ICIs.
Collapse
Grants
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
Collapse
Affiliation(s)
- Jing Wei
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Wenke Li
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Pengfei Zhang
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Ming Liu
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
18
|
Rediti M, Venet D, Joaquin Garcia A, Maetens M, Vincent D, Majjaj S, El-Abed S, Di Cosimo S, Ueno T, Izquierdo M, Piccart M, Pusztai L, Loi S, Salgado R, Viale G, Rothé F, Sotiriou C. Identification of HER2-positive breast cancer molecular subtypes with potential clinical implications in the ALTTO clinical trial. Nat Commun 2024; 15:10402. [PMID: 39613746 PMCID: PMC11607438 DOI: 10.1038/s41467-024-54621-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
In HER2-positive breast cancer, clinical outcome and sensitivity to HER2-targeted therapies are influenced by both tumor and microenvironment features. However, we are currently unable to depict the molecular heterogeneity of this disease with sufficient granularity. Here, by performing gene expression profiling in HER2-positive breast cancers from patients receiving adjuvant trastuzumab in the ALTTO clinical trial (NCT00490139), we identify and characterize five molecular subtypes associated with the risk of distant recurrence: immune-enriched, proliferative/metabolic-enriched, mesenchymal/stroma-enriched, luminal, and ERBB2-dependent. Additionally, we validate the biological profiles of the subtypes and explore their prognostic/predictive value in external cohorts, namely the NeoALTTO trial (NCT00553358), SCAN-B (NCT02306096), I-SPY2 (NCT01042379), METABRIC and TCGA. Immune-enriched tumors present better survival outcomes, in contrast to mesenchymal/stroma-enriched and proliferative/metabolic-enriched tumors, while luminal and ERBB2-dependent tumors are characterized by low and high rates of pathological complete response, respectively. Of note, these molecular subtypes provide the rationale for treatment approaches leveraging the heterogeneous biology of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Mattia Rediti
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - David Venet
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Andrea Joaquin Garcia
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marion Maetens
- Laboratory for Translational Breast Cancer Research, KU Leuven, Leuven, Belgium
| | - Delphine Vincent
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Samira Majjaj
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Serena Di Cosimo
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Takayuki Ueno
- Breast Surgical Oncology, Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | - Martine Piccart
- Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Lajos Pusztai
- Yale School of Medicine, Yale Cancer Center, New Haven, CT, USA
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Roberto Salgado
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
| | - Giuseppe Viale
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Françoise Rothé
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
19
|
Li YW, Dai LJ, Wu XR, Zhao S, Xu YZ, Jin X, Xiao Y, Wang Y, Lin CJ, Zhou YF, Fu T, Yang WT, Li M, Lv H, Chen S, Grigoriadis A, Jiang YZ, Ma D, Shao ZM. Molecular Characterization and Classification of HER2-Positive Breast Cancer Inform Tailored Therapeutic Strategies. Cancer Res 2024; 84:3669-3683. [PMID: 39186675 DOI: 10.1158/0008-5472.can-23-4066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/19/2024] [Accepted: 08/08/2024] [Indexed: 08/28/2024]
Abstract
HER2-positive breast cancer is an aggressive subtype that accounts for 15% to 20% of all breast cancers. Recent studies have suggested that HER2-positive breast cancer is a group of heterogeneous diseases with different sensitivities to standard treatment regimens. Revealing the molecular heterogeneity of HER2-positive breast cancer could potentially enable more precise treatment strategies. In this study, we performed multiomics profiling on a HER2-positive breast cancer cohort and identified four transcriptome-based subtypes. The classical HER2 (HER2-CLA) subtype comprised 28.3% of the samples and displayed high ERBB2 activation and significant benefit from anti-HER2 therapy. The immunomodulatory (HER2-IM) subtype (20%) featured an immune-activated microenvironment, potentially suitable for de-escalated treatment and immunotherapy. The luminal-like (HER2-LUM) subtype (30.6%) possessed similar molecular features of hormone receptor-positive HER2-negative breast cancer, suggesting endocrine therapy and CDK4/6 inhibitors as a potential therapeutic strategy. Lastly, the basal/mesenchymal-like (HER2-BM) subtype (21.1%) had a poor response to current dual HER2-targeted therapy and could potentially benefit from tyrosine kinase inhibitors. The molecular characteristics and clinical features of the subtypes were further explored across multiple cohorts, and the feasibility of the proposed treatment strategies was validated in patient-derived organoid and patient-derived tumor fragment models. This study elucidates the molecular heterogeneity of HER2-positive breast cancer and paves the way for a more tailored treatment. Significance: Illumination of the inherent heterogeneity within HER2-positive breast cancers through the delineation of distinct molecular subtypes lays the groundwork for developing more personalized treatment strategies based on specific patient characteristics.
Collapse
Affiliation(s)
- Yu-Wei Li
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei-Jie Dai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiang-Rong Wu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shen Zhao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Zheng Xu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi Jin
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Xiao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Wang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cai-Jin Lin
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Fan Zhou
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tong Fu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Tao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ming Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hong Lv
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Siyuan Chen
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Anita Grigoriadis
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ding Ma
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
20
|
García-Torralba E, Galluzzi L, Buqué A. Prognostic value of atypical B cells in breast cancer. Trends Cancer 2024; 10:990-991. [PMID: 39353814 DOI: 10.1016/j.trecan.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
The impact of tumor-infiltrating B cells on breast cancer (BRCA) outcomes remains poorly understood. Recent findings from Yang et al. identify an atypical, clonally expanded population of activated Fc receptor-like 4 (FCRL4)+ B cells that is associated with improved overall survival in patients affected by various tumor types, including BRCA.
Collapse
Affiliation(s)
- Esmeralda García-Torralba
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Medical Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain; Department of Medicine, Medical School, University of Murcia, Murcia, Spain; IMIB-Arrixaca, Murcia, Spain
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Aitziber Buqué
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Waks AG, Martínez-Sáez O, Tarantino P, Braso-Maristany F, Pascual T, Cortés J, Tolaney SM, Prat A. Dual HER2 inhibition: mechanisms of synergy, patient selection, and resistance. Nat Rev Clin Oncol 2024; 21:818-832. [PMID: 39271787 DOI: 10.1038/s41571-024-00939-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2024] [Indexed: 09/15/2024]
Abstract
HER2-targeted therapies for patients with HER2+ breast cancer are rapidly evolving, offering a range of more complex and personalized treatment options. Currently, an array of anti-HER2 monoclonal antibodies, tyrosine kinase inhibitors and antibody-drug conjugates are administered, sometimes alongside chemotherapy or endocrine therapy, both in curative and palliative contexts. However, the heterogeneous nature of HER2+ breast cancer demands a deeper understanding of disease biology and its role in responsiveness to novel HER2-targeted agents, as well as non-HER2-targeted therapies, in order to optimize patient outcomes. In this Review, we revisit the mechanisms of action of HER2-targeted agents, examine the evidence supporting the use of dual HER2 blockade in patients with HER2-amplified tumours, and explore the role of biomarkers in guiding future treatment strategies. We also discuss potential implications for the future treatment of patients with HER2+ breast cancer.
Collapse
Affiliation(s)
- Adrienne G Waks
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Olga Martínez-Sáez
- Cancer Institute, Hospital Clinic of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Paolo Tarantino
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Fara Braso-Maristany
- Cancer Institute, Hospital Clinic of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Tomás Pascual
- Cancer Institute, Hospital Clinic of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- SOLTI Cancer Research Group, Barcelona, Spain
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Barcelona, Spain
- IOB Madrid, Hospital Beata Maria Ana, Madrid, Spain
- Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
| | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aleix Prat
- Cancer Institute, Hospital Clinic of Barcelona, Barcelona, Spain.
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Department of Medicine, University of Barcelona, Barcelona, Spain.
- Breast Cancer Unit, IOB-QuirónSalud, Barcelona, Spain.
- Reveal Genomics, Barcelona, Spain.
| |
Collapse
|
22
|
Lin X, Liu J, Zhang N, Zhou D, Liu Y. Decoding the immune microenvironment: unveiling CD8 + T cell-related biomarkers and developing a prognostic signature for personalized glioma treatment. Cancer Cell Int 2024; 24:331. [PMID: 39354483 PMCID: PMC11443942 DOI: 10.1186/s12935-024-03517-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/25/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Gliomas are aggressive brain tumors with poor prognosis. Understanding the tumor immune microenvironment (TIME) in gliomas is essential for developing effective immunotherapies. This study aimed to identify TIME-related biomarkers in glioma using bioinformatic analysis of RNA-seq data. METHODS In this study, we employed weighted gene co-expression network analysis (WGCNA) on bulk RNA-seq data to identify TIME-related genes. To identify prognostic genes, we performed univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analyses. Based on these genes, we constructed a prognostic signature and delineated risk groups. To validate the prognostic signature, external validation was conducted. RESULTS CD8 + T cell infiltration was strongly correlated with glioma patient prognosis. We identified 115 CD8 + T cell-related genes through integrative analysis of bulk-seq data. CDCA5, KIF11, and KIF4A were found to be significant immune-related genes (IRGs) associated with overall survival in glioma patients and served as independent prognostic factors. We developed a prognostic nomogram that incorporated these genes, age, gender, and grade, providing a reliable tool for clinicians to predict patient survival probabilities. The nomogram's predictions were supported by calibration plots, further validating its accuracy. CONCLUSION In conclusion, our study identifies CD8 + T cell infiltration as a strong predictor of glioma patient outcomes and highlights the prognostic value of genes. The developed prognostic nomogram, incorporating these genes along with clinical factors, provides a reliable tool for predicting patient survival probabilities and has important implications for personalized treatment decisions in glioma.
Collapse
Affiliation(s)
- Xiaofang Lin
- Laboratory Department of Zengcheng Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianqiang Liu
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ni Zhang
- Department of Physical Medicine and Rehabilitation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China.
| | - Dexiang Zhou
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China.
| | - Yakang Liu
- Department of Physical Medicine and Rehabilitation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
23
|
Ultimescu F, Hudita A, Popa DE, Olinca M, Muresean HA, Ceausu M, Stanciu DI, Ginghina O, Galateanu B. Impact of Molecular Profiling on Therapy Management in Breast Cancer. J Clin Med 2024; 13:4995. [PMID: 39274207 PMCID: PMC11396537 DOI: 10.3390/jcm13174995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/16/2024] Open
Abstract
Breast cancer (BC) remains the most prevalent cancer among women and the leading cause of cancer-related mortality worldwide. The heterogeneity of BC in terms of histopathological features, genetic polymorphisms, and response to therapies necessitates a personalized approach to treatment. This review focuses on the impact of molecular profiling on therapy management in breast cancer, emphasizing recent advancements in next-generation sequencing (NGS) and liquid biopsies. These technologies enable the identification of specific molecular subtypes and the detection of blood-based biomarkers such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and tumor-educated platelets (TEPs). The integration of molecular profiling with traditional clinical and pathological data allows for more tailored and effective treatment strategies, improving patient outcomes. This review also discusses the current challenges and prospects of implementing personalized cancer therapy, highlighting the potential of molecular profiling to revolutionize BC management through more precise prognostic and therapeutic interventions.
Collapse
Affiliation(s)
- Flavia Ultimescu
- OncoTeam Diagnostic S.A., 010719 Bucharest, Romania
- Doctoral School of Medicine, "Carol Davila" University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania
| | - Ariana Hudita
- Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
- Research Institute of the University of Bucharest, University of Bucharest, 050663 Bucharest, Romania
| | - Daniela Elena Popa
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy Bucharest, 020956 Bucharest, Romania
| | - Maria Olinca
- OncoTeam Diagnostic S.A., 010719 Bucharest, Romania
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania
| | | | - Mihail Ceausu
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania
| | | | - Octav Ginghina
- Faculty of Dental Medicine, "Carol Davila" University of Medicine and Pharmacy Bucharest, 010221 Bucharest, Romania
- Department of Surgery 3, "Prof. Dr. Al. Trestioreanu" Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Bianca Galateanu
- Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| |
Collapse
|
24
|
Fu C, Ji W, Cui Q, Chen A, Weng H, Lu N, Yang W. GSDME-mediated pyroptosis promotes anti-tumor immunity of neoadjuvant chemotherapy in breast cancer. Cancer Immunol Immunother 2024; 73:177. [PMID: 38954046 PMCID: PMC11219631 DOI: 10.1007/s00262-024-03752-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/02/2024] [Indexed: 07/04/2024]
Abstract
Paclitaxel and anthracycline-based chemotherapy is one of the standard treatment options for breast cancer. However, only about 6-30% of breast cancer patients achieved a pathological complete response (pCR), and the mechanism responsible for the difference is still unclear. In this study, random forest algorithm was used to screen feature genes, and artificial neural network (ANN) algorithm was used to construct an ANN model for predicting the efficacy of neoadjuvant chemotherapy for breast cancer. Furthermore, digital pathology, cytology, and molecular biology experiments were used to verify the relationship between the efficacy of neoadjuvant chemotherapy and immune ecology. It was found that paclitaxel and doxorubicin, an anthracycline, could induce typical pyroptosis and bubbling in breast cancer cells, accompanied by gasdermin E (GSDME) cleavage. Paclitaxel with LDH release and Annexin V/PI doubule positive cell populations, and accompanied by the increased release of damage-associated molecular patterns, HMGB1 and ATP. Cell coculture experiments also demonstrated enhanced phagocytosis of macrophages and increased the levels of IFN-γ and IL-2 secretion after paclitaxel treatment. Mechanistically, GSDME may mediate paclitaxel and doxorubicin-induced pyroptosis in breast cancer cells through the caspase-9/caspase-3 pathway, activate anti-tumor immunity, and promote the efficacy of paclitaxel and anthracycline-based neoadjuvant chemotherapy. This study has practical guiding significance for the precision treatment of breast cancer, and can also provide ideas for understanding molecular mechanisms related to the chemotherapy sensitivity.
Collapse
Affiliation(s)
- Changfang Fu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, 230001, Anhui, China
| | - Wenbo Ji
- Clinical Pharmacy Department, Anhui Provincial Children's Hospital, Hefei, 230000, Anhui, China
| | - Qianwen Cui
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Anling Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Haiyan Weng
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Nannan Lu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China.
| |
Collapse
|
25
|
Leon-Ferre RA, Whitaker KR, Suman VJ, Hoskin T, Giridhar KV, Moore RM, Al-Jarrad A, McLaughlin SA, Northfelt DW, Hunt KN, Conners AL, Moyer A, Carter JM, Kalari K, Weinshilboum R, Wang L, Ingle JN, Knutson KL, Ansell SM, Boughey JC, Goetz MP, Villasboas JC. Pre-treatment peripheral blood immunophenotyping and response to neoadjuvant chemotherapy in operable breast cancer. Breast Cancer Res 2024; 26:97. [PMID: 38858721 PMCID: PMC11165781 DOI: 10.1186/s13058-024-01848-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/22/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Tumor immune infiltration and peripheral blood immune signatures have prognostic and predictive value in breast cancer. Whether distinct peripheral blood immune phenotypes are associated with response to neoadjuvant chemotherapy (NAC) remains understudied. METHODS Peripheral blood mononuclear cells from 126 breast cancer patients enrolled in a prospective clinical trial (NCT02022202) were analyzed using Cytometry by time-of-flight with a panel of 29 immune cell surface protein markers. Kruskal-Wallis tests or Wilcoxon rank-sum tests were used to evaluate differences in immune cell subpopulations according to breast cancer subtype and response to NAC. RESULTS There were 122 evaluable samples: 47 (38.5%) from patients with hormone receptor-positive, 39 (32%) triple-negative (TNBC), and 36 (29.5%) HER2-positive breast cancer. The relative abundances of pre-treatment peripheral blood T, B, myeloid, NK, and unclassified cells did not differ according to breast cancer subtype. In TNBC, higher pre-treatment myeloid cells were associated with lower pathologic complete response (pCR) rates. In hormone receptor-positive breast cancer, lower pre-treatment CD8 + naïve and CD4 + effector memory cells re-expressing CD45RA (TEMRA) T cells were associated with more extensive residual disease after NAC. In HER2 + breast cancer, the peripheral blood immune phenotype did not differ according to NAC response. CONCLUSIONS Pre-treatment peripheral blood immune cell populations (myeloid in TNBC; CD8 + naïve T cells and CD4 + TEMRA cells in luminal breast cancer) were associated with response to NAC in early-stage TNBC and hormone receptor-positive breast cancers, but not in HER2 + breast cancer. TRIAL REGISTRATION NCT02022202 . Registered 20 December 2013.
Collapse
Affiliation(s)
| | | | - Vera J Suman
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Tanya Hoskin
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Raymond M Moore
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Katie N Hunt
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Ann Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Krishna Kalari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Liewei Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | | |
Collapse
|
26
|
Chen H, Gui X, Zhou Z, Su F, Gong C, Li S, Wu W, Rao N, Liu Q, Yao H. Distinct ER and PR expression patterns significantly affect the clinical outcomes of early HER2-positive breast cancer: A real-world analysis of 871 patients treated with neoadjuvant therapy. Breast 2024; 75:103733. [PMID: 38615482 PMCID: PMC11026842 DOI: 10.1016/j.breast.2024.103733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/07/2024] [Accepted: 04/07/2024] [Indexed: 04/16/2024] Open
Abstract
INTRODUCTION The impact of distinct estrogen receptor (ER) and progesterone receptor (PR) expression patterns on tumor behavior and treatment outcomes within HER2-positive breast cancer is not fully explored. This study aimed to comprehensively examine the clinical differences among patients with HER2-positive breast cancer harboring distinct ER and PR expression patterns in the neoadjuvant setting. METHODS This retrospective analysis included 871 HER2-positive breast patients treated with neoadjuvant therapy at our hospital between 2011 and 2022. Comparisons were performed across the three hormone receptor (HR)-specific subtypes, namely the ER-negative/PR-negative/HER2-positive (ER-/PR-/HER2+), the single HR-positive (HR+)/HER2+, and the triple-positive breast cancer (TPBC) subtypes. RESULTS Of 871 patients, 21.0% had ER-/PR-/HER2+ tumors, 33.6% had single HR+/HER2+ disease, and 45.4% had TPBC. Individuals with single HR+/HER2+ tumors and TPBC cases demonstrated significantly lower pathological complete response (pCR) rates compared to those with ER-/PR-/HER2+ tumors (36.9% vs. 24.3% vs. 49.2%, p < 0.001). Multivariate analysis confirmed TPBC as significantly associated with decreased pCR likelihood (OR = 0.42, 95%CI 0.28-0.63, p < 0.001). Survival outcomes, including disease-free survival (DFS) and overall survival (OS), showed no significant differences across HR-specific subtypes in the overall patient population. However, within patients without anti-HER2 therapy, TPBC was linked to improved DFS and a trend towards better OS. CONCLUSIONS HER2-positive breast cancer exhibited three distinct HR-specific subtypes with varying clinical manifestations and treatment responses. These findings suggest personalized treatment strategies considering ER and PR expression patterns, emphasizing the need for further investigations to unravel molecular traits underlying HER2-positive breast cancer with distinct HR expression patterns.
Collapse
Affiliation(s)
- Haizhu Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiujuan Gui
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ziwei Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fengxi Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chang Gong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shunrong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nanyan Rao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
27
|
Otterlei Fjørtoft M, Huse K, Rye IH. The Tumor Immune Microenvironment in Breast Cancer Progression. Acta Oncol 2024; 63:359-367. [PMID: 38779867 PMCID: PMC11332517 DOI: 10.2340/1651-226x.2024.33008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/12/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND The tumor microenvironment significantly influences breast cancer development, progression, and metastasis. Various immune cell populations, including T cells, B cells, NK cells, and myeloid cells exhibit diverse functions in different breast cancer subtypes, contributing to both anti-tumor and pro-tumor activities. PURPOSE This review provides an overview of the predominant immune cell populations in breast cancer subtypes, elucidating their suppressive and prognostic effects. We aim to outline the role of the immune microenvironment from normal breast tissue to invasive cancer and distant metastasis. METHODS A comprehensive literature review was conducted to analyze the involvement of immune cells throughout breast cancer progression. RESULTS In breast cancer, tumors exhibit increased immune cell infiltration compared to normal tissue. Variations exist across subtypes, with higher levels observed in triple-negative and HER2+ tumors are linked to better survival. In contrast, ER+ tumors display lower immune infiltration, associated with poorer outcomes. Furthermore, metastatic sites commonly exhibit a more immunosuppressive microenvironment. CONCLUSION Understanding the complex interaction between tumor and immune cells during breast cancer progression is essential for future research and the development of immune-based strategies. This comprehensive understanding may pave the way for more effective treatment approaches and improved patients outcomes.
Collapse
Affiliation(s)
- Marit Otterlei Fjørtoft
- Department of Cancer Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Radium Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Radium Hospital, Oslo, Norway
| | - Inga Hansine Rye
- Department of Cancer Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Radium Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
28
|
Fernandez-Martinez A, Rediti M, Tang G, Pascual T, Hoadley KA, Venet D, Rashid NU, Spears PA, Islam MN, El-Abed S, Bliss J, Lambertini M, Di Cosimo S, Huobe J, Goerlitz D, Hu R, Lucas PC, Swain SM, Sotiriou C, Perou CM, Carey LA. Tumor Intrinsic Subtypes and Gene Expression Signatures in Early-Stage ERBB2/HER2-Positive Breast Cancer: A Pooled Analysis of CALGB 40601, NeoALTTO, and NSABP B-41 Trials. JAMA Oncol 2024; 10:603-611. [PMID: 38546612 PMCID: PMC10979363 DOI: 10.1001/jamaoncol.2023.7304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/08/2023] [Indexed: 04/01/2024]
Abstract
Importance Biologic features may affect pathologic complete response (pCR) and event-free survival (EFS) after neoadjuvant chemotherapy plus ERBB2/HER2 blockade in ERBB2/HER2-positive early breast cancer (EBC). Objective To define the quantitative association between pCR and EFS by intrinsic subtype and by other gene expression signatures in a pooled analysis of 3 phase 3 trials: CALGB 40601, NeoALTTO, and NSABP B-41. Design, Setting, and Participants In this retrospective pooled analysis, 1289 patients with EBC received chemotherapy plus either trastuzumab, lapatinib, or the combination, with a combined median follow-up of 5.5 years. Gene expression profiling by RNA sequencing was obtained from 758 samples, and intrinsic subtypes and 618 gene expression signatures were calculated. Data analyses were performed from June 1, 2020, to January 1, 2023. Main Outcomes and Measures The association of clinical variables and gene expression biomarkers with pCR and EFS were studied by logistic regression and Cox analyses. Results In the pooled analysis, of 758 women, median age was 49 years, 12% were Asian, 6% Black, and 75% were White. Overall, pCR results were associated with EFS in the ERBB2-enriched (hazard ratio [HR], 0.45; 95% CI, 0.29-0.70; P < .001) and basal-like (HR, 0.19; 95% CI, 0.04-0.86; P = .03) subtypes but not in luminal A or B tumors. Dual trastuzumab plus lapatinib blockade over trastuzumab alone had a trend toward EFS benefit in the intention-to-treat population; however, in the ERBB2-enriched subtype there was a significant and independent EFS benefit of trastuzumab plus lapatinib vs trastuzumab alone (HR, 0.47; 95% CI, 0.27-0.83; P = .009). Overall, 275 of 618 gene expression signatures (44.5%) were significantly associated with pCR and 9 of 618 (1.5%) with EFS. The ERBB2/HER2 amplicon and multiple immune signatures were significantly associated with pCR. Luminal-related signatures were associated with lower pCR rates but better EFS, especially among patients with residual disease and independent of hormone receptor status. There was significant adjusted HR for pCR ranging from 0.45 to 0.81 (higher pCR) and 1.21-1.94 (lower pCR rate); significant adjusted HR for EFS ranged from 0.71 to 0.94. Conclusions and relevance In patients with ERBB2/HER2-positive EBC, the association between pCR and EFS differed by tumor intrinsic subtype, and the benefit of dual ERBB2/HER2 blockade was limited to ERBB2-enriched tumors. Immune-activated signatures were concordantly associated with higher pCR rates and better EFS, whereas luminal signatures were associated with lower pCR rates.
Collapse
Affiliation(s)
- Aranzazu Fernandez-Martinez
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill
- Department of Genetics, University of North Carolina, Chapel Hill
| | - Mattia Rediti
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Gong Tang
- NSABP Foundation Inc., Pittsburgh, Pennsylvania
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tomás Pascual
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill
- Department of Medical Oncology, Hospital Clínic de Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- SOLTI Breast Cancer Cooperative Group, Barcelona, Spain
| | - Katherine A. Hoadley
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill
- Department of Genetics, University of North Carolina, Chapel Hill
| | - David Venet
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Naim U. Rashid
- Department of Biostatistics, University of North Carolina, Chapel Hill
| | - Patricia A. Spears
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill
| | - Md N. Islam
- Genomics and Epigenomics Shared Resource (GESR), Georgetown University Medical Center, Washington, DC
| | | | - Judith Bliss
- The Institute of Cancer Research, Clinical Trials & Statistics Unit, London, United Kingdom
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Serena Di Cosimo
- Integrated Biology Platform, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Jens Huobe
- Kantonsspital St. Gallen, Brustzentrum, Departement Interdisziplinäre medizinische Dienste, St. Gallen, Switzerland
| | - David Goerlitz
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Rong Hu
- Genomics and Epigenomics Shared Resource (GESR), Georgetown University Medical Center, Washington, DC
| | - Peter C. Lucas
- NSABP Foundation Inc., Pittsburgh, Pennsylvania
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sandra M. Swain
- NSABP Foundation Inc., Pittsburgh, Pennsylvania
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Charles M. Perou
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill
- Department of Genetics, University of North Carolina, Chapel Hill
| | - Lisa A. Carey
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill
- Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
29
|
Saleh RO, Ibrahim FM, Pallathadka H, Kaur I, Ahmad I, Ali SHJ, Redhee AH, Ghildiyal P, Jawad MA, Alsaadi SB. Nucleic acid vaccines-based therapy for triple-negative breast cancer: A new paradigm in tumor immunotherapy arena. Cell Biochem Funct 2024; 42:e3992. [PMID: 38551221 DOI: 10.1002/cbf.3992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/04/2024] [Accepted: 03/10/2024] [Indexed: 04/02/2024]
Abstract
Nucleic acid vaccines (NAVs) have the potential to be economical, safe, and efficacious. Furthermore, just the chosen antigen in the pathogen is the target of the immune responses brought on by NAVs. Triple-negative breast cancer (TNBC) treatment shows great promise for nucleic acid-based vaccines, such as DNA (as plasmids) and RNA (as messenger RNA [mRNA]). Moreover, cancer vaccines offer a compelling approach that can elicit targeted and long-lasting immune responses against tumor antigens. Bacterial plasmids that encode antigens and immunostimulatory molecules serve as the foundation for DNA vaccines. In the 1990s, plasmid DNA encoding the influenza A nucleoprotein triggered a protective and targeted cytotoxic T lymphocyte (CTL) response, marking the first instance of DNA vaccine-mediated immunity. Similarly, in vitro transcribed mRNA was first successfully used in animals in 1990. At that point, mice were given an injection of the gene encoding the mRNA sequence, and the researchers saw the production of a protein. We begin this review by summarizing our existing knowledge of NAVs. Next, we addressed NAV delivery, emphasizing the need to increase efficacy in TNBC.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Fatma M Ibrahim
- Community Health Nursing, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
- Geriatric Nursing, Mansoura University, Mansoura, Egypt
| | | | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Saad Hayif Jasim Ali
- Department of Medical Laboratory, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq
| | - Ahmed Huseen Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Salim B Alsaadi
- Department of Pharmaceutics, Al-Hadi University College, Baghdad, Iraq
| |
Collapse
|
30
|
Conte B, Brasó-Maristany F, Hernández AR, Pascual T, Villacampa G, Schettini F, Vidal Losada MJ, Seguí E, Angelats L, Garcia-Fructuoso I, Gómez-Bravo R, Lorman-Carbó N, Paré L, Marín-Aguilera M, Martínez-Sáez O, Adamo B, Sanfeliu E, Fratini B, Falato C, Chic N, Vivancos A, Villagrasa P, Staaf J, Parker JS, Perou CM, Prat A. A 14-gene B-cell immune signature in early-stage triple-negative breast cancer (TNBC): a pooled analysis of seven studies. EBioMedicine 2024; 102:105043. [PMID: 38447275 PMCID: PMC10924177 DOI: 10.1016/j.ebiom.2024.105043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Early-stage triple-negative breast cancer (TNBC) displays clinical and biological diversity. From a biological standpoint, immune infiltration plays a crucial role in TNBC prognosis. Currently, there is a lack of genomic tools aiding in treatment decisions for TNBC. This study aims to assess the effectiveness of a B-cell/immunoglobulin signature (IGG) alone, or in combination with tumor burden, in predicting prognosis and treatment response in patients with TNBC. METHODS Genomic and clinical data were retrieved from 7 cohorts: SCAN-B (N = 874), BrighTNess (n = 482), CALGB-40603 (n = 389), METABRIC (n = 267), TCGA (n = 118), GSE58812 (n = 107), GSE21653 (n = 67). IGG and a risk score integrating IGG with tumor/nodal staging (IGG-Clin) were assessed for event-free survival (EFS) and overall survival (OS) in each cohort. Random effects model was used to derive pooled effect sizes. Association of IGG with pathological complete response (pCR) was assessed in CALGB-40603 and BrighTNess. Immune significance of IGG was estimated through CIBERSORTx and EcoTyper. FINDINGS IGG was associated with improved EFS (pooled HR = 0.77, [95% CI = 0.70-0.85], I2 = 18%) and OS (pooled HR = 0.79, [0.73-0.85], I2 = 0%) across cohorts, and was predictive of pCR in CALGB-40603 (OR 1.25, [1.10-1.50]) and BrighTNess (OR 1.57 [1.25-1.98]). IGG-Clin was predictive of recurrence (pooled HR = 2.11, [1.75-2.55], I2 = 0%) and death (pooled HR = 1.99, 95% [0.84-4.73], I2 = 79%) across cohorts. IGG was associated with adaptive immune response at CIBERSORTx and EcoTyper analysis. INTERPRETATION IGG is linked to improved prognosis and pCR in early-stage TNBC. The integration of IGG alongside tumor and nodal staging holds promise as an approach to identify patients benefitting from intensified or de-intensified treatments. FUNDING This study received funding from: Associació Beca Marta Santamaria, European Union's Horizon 2020 research and innovation and Marie Skłodowska-Curie Actions programs, Fundación FERO, Fundación CRIS contra el cáncer, Agència de Gestó d'Ajuts Universitaris i de Recerca, Instituto de Salud Carlos III, Fundación Contigo, Asociación Cáncer de Mama Metastásico IV, Breast Cancer Research Foundation, RESCUER, Fundación científica AECC and FSEOM.
Collapse
Affiliation(s)
- Benedetta Conte
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain; Reveal Genomics, Barcelona, Spain
| | - Adela Rodríguez Hernández
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Tomás Pascual
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain
| | - Guillermo Villacampa
- Reveal Genomics, Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain; Oncology Data Science, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Francesco Schettini
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Maria J Vidal Losada
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain; Institute of Oncology (IOB)-Hospital QuirónSalud, Barcelona, Spain
| | - Elia Seguí
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain
| | - Laura Angelats
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Isabel Garcia-Fructuoso
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Raquel Gómez-Bravo
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Natàlia Lorman-Carbó
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | | | - Olga Martínez-Sáez
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Barbara Adamo
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Esther Sanfeliu
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Pathology Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Beatrice Fratini
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Claudette Falato
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain
| | - Núria Chic
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Ana Vivancos
- Reveal Genomics, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Cancer Genomics Group, Barcelona, Spain
| | | | - Johan Staaf
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Sweden
| | - Joel S Parker
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Charles M Perou
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, Spain; Reveal Genomics, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain; Institute of Oncology (IOB)-Hospital QuirónSalud, Barcelona, Spain.
| |
Collapse
|
31
|
Eun NL, Bae SJ, Youk JH, Son EJ, Ahn SG, Jeong J, Kim JH, Lee Y, Cha YJ. Tumor-Infiltrating Lymphocyte Level Consistently Correlates with Lower Stiffness Measured by Shear-Wave Elastography: Subtype-Specific Analysis of Its Implication in Breast Cancer. Cancers (Basel) 2024; 16:1254. [PMID: 38610934 PMCID: PMC11011118 DOI: 10.3390/cancers16071254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Background: We aimed to elucidate the clinical significance of tumor stiffness across breast cancer subtypes and establish its correlation with the tumor-infiltrating lymphocyte (TIL) levels using shear-wave elastography (SWE). Methods: SWE was used to measure tumor stiffness in breast cancer patients from January 2016 to August 2020. The association of tumor stiffness and clinicopathologic parameters, including the TIL levels, was analyzed in three breast cancer subtypes. Results: A total of 803 patients were evaluated. Maximal elasticity (Emax) showed a consistent positive association with an invasive size and the pT stage in all cases, while it negatively correlated with the TIL level. A subgroup-specific analysis revealed that the already known parameters for high stiffness (lymphovascular invasion, lymph node metastasis, Ki67 levels) were significant only in hormone receptor-positive and HER2-negative breast cancer (HR + HER2-BC). In the multivariate logistic regression, an invasive size and low TIL levels were significantly associated with Emax in HR + HER2-BC and HER2 + BC. In triple-negative breast cancer, only TIL levels were significantly associated with low Emax. Linear regression confirmed a consistent negative correlation between TIL and Emax in all subtypes. Conclusions: Breast cancer stiffness presents varying clinical implications dependent on the tumor subtype. Elevated stiffness indicates a more aggressive tumor biology in HR + HER2-BC, but is less significant in other subtypes. High TIL levels consistently correlate with lower tumor stiffness across all subtypes.
Collapse
Affiliation(s)
- Na Lae Eun
- Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea; (N.L.E.); (J.H.Y.); (E.J.S.)
| | - Soong June Bae
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea; (S.J.B.); (S.G.A.); (J.J.); (J.H.K.)
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Ji Hyun Youk
- Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea; (N.L.E.); (J.H.Y.); (E.J.S.)
| | - Eun Ju Son
- Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea; (N.L.E.); (J.H.Y.); (E.J.S.)
| | - Sung Gwe Ahn
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea; (S.J.B.); (S.G.A.); (J.J.); (J.H.K.)
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Joon Jeong
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea; (S.J.B.); (S.G.A.); (J.J.); (J.H.K.)
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Jee Hung Kim
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea; (S.J.B.); (S.G.A.); (J.J.); (J.H.K.)
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Yangkyu Lee
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea; (S.J.B.); (S.G.A.); (J.J.); (J.H.K.)
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Yoon Jin Cha
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea; (S.J.B.); (S.G.A.); (J.J.); (J.H.K.)
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| |
Collapse
|
32
|
Lee S, Kang BH, Lee HB, Jang BS, Han W, Kim IA. B-Cell-Mediated Immunity Predicts Survival of Patients With Estrogen Receptor-Positive Breast Cancer. JCO Precis Oncol 2024; 8:e2300263. [PMID: 38452311 DOI: 10.1200/po.23.00263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 12/21/2023] [Accepted: 01/11/2024] [Indexed: 03/09/2024] Open
Abstract
PURPOSE The estrogen receptor-positive (ER+) breast cancer (BC), which constitutes the majority of BC cases, exhibits highly heterogeneous clinical behavior. To aid precision treatments, we aimed to find molecular subtypes of ER+ BC representing the tumor microenvironment and prognosis. METHODS We analyzed RNA-seq data of 113 patients with BC and classified them according to the PAM50 intrinsic subtypes using gene expression profiles. Among them, we further focused on 44 patients with luminal-type (ER+) BC for subclassification. The Cancer Genome Atlas (TCGA) data of patients with BC were used as a validation data set to verify the new classification. We estimated the immune cell composition using CIBERSORT and further analyzed its association with clinical or molecular parameters. RESULTS Principal component analysis clearly divided the patients into two subgroups separately from the luminal A and B classification. The top differentially expressed genes between the subgroups were distinctly characterized by immunoglobulin and B-cell-related genes. We could also cluster a separate cohort of patients with luminal-type BC from TCGA into two subgroups on the basis of the expression of a B-cell-specific gene set, and patients who were predicted to have high B-cell immune activity had better prognoses than other patients. CONCLUSION Our transcriptomic approach emphasize a molecular phenotype of B-cell immunity in ER+ BC that may help to predict disease prognosis. Although further researches are required, B-cell immunity for patients with ER+ BC may be helpful for identifying patients who are good responders to chemotherapy or immunotherapy.
Collapse
Affiliation(s)
- Seungbok Lee
- Department of Genomic Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Byung-Hee Kang
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Radiation Oncology, Ewha Womans University Seoul Hospital, Seoul, Republic of Korea
| | - Han-Byoel Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Bum-Sup Jang
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - In Ah Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Radiation Oncology and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
33
|
Jääskeläinen MM, Tumelius R, Hämäläinen K, Rilla K, Oikari S, Rönkä A, Selander T, Mannermaa A, Tiainen S, Auvinen P. High Numbers of CD163+ Tumor-Associated Macrophages Predict Poor Prognosis in HER2+ Breast Cancer. Cancers (Basel) 2024; 16:634. [PMID: 38339385 PMCID: PMC10854814 DOI: 10.3390/cancers16030634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are associated with a poor outcome in breast cancer (BC), but their prognostic value in different BC subtypes has remained somewhat unclear. Here, we investigated the prognostic value of M2-like TAMs (CD163+) and all TAMs (CD68+) in a patient cohort of 278 non-metastatic BC patients, half of whom were HER2+ (n = 139). The survival endpoints investigated were overall survival (OS), breast cancer-specific survival (BCSS) and disease-free survival (DFS). In the whole patient cohort (n = 278), a high CD163+ TAM count and a high CD68+ TAM count were associated with a worse outcome (p ≤ 0.023). In HER2+ BC, a high CD163+ TAM count was an independent factor for a poor prognosis across all the investigated survival endpoints (p < 0.001). The prognostic effect was evident in both the HER2+/hormone receptor-positive (p < 0.001) and HER2+/hormone receptor-negative (p ≤ 0.012) subgroups and regardless of the provision of adjuvant trastuzumab (p ≤ 0.002). In HER2-negative BC, the CD163+ TAM count was not significantly associated with survival. These results suggest that a high CD163+ TAM count predicts an inferior outcome, especially in HER2+ BC patients, and as adjuvant trastuzumab did not overcome the poor prognostic effect, combination treatments including therapies targeting the macrophage function could represent an effective therapeutic approach in HER2+ BC.
Collapse
Affiliation(s)
- Minna M. Jääskeläinen
- Cancer Center, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Ritva Tumelius
- Kuopio Center for Gene and Cell Therapy, 70210 Kuopio, Finland
| | - Kirsi Hämäläinen
- Institute of Clinical Medicine, Clinical Pathology and Forensic Medicine, University of Eastern Finland, 70211 Kuopio, Finland
- Imaging Center, Clinical Pathology, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
- Biocenter Kuopio and Cancer Center of Eastern Finland, University of Eastern Finland, 70211 Kuopio, Finland
| | - Kirsi Rilla
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Sanna Oikari
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Aino Rönkä
- Cancer Center, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Tuomas Selander
- Science Services Center, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
| | - Arto Mannermaa
- Institute of Clinical Medicine, Clinical Pathology and Forensic Medicine, University of Eastern Finland, 70211 Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, Wellbeing Services County of North Savo, 700029 Kuopio, Finland
| | - Satu Tiainen
- Cancer Center, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Päivi Auvinen
- Cancer Center, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
34
|
Mano MS. More on treatment de-escalation, biomarkers of response in human epidermal growth factor receptor 2 (HER2)-positive breast cancer: long-term outcomes and translational research findings of the PREDIX HER2 trial. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:16. [PMID: 38304900 PMCID: PMC10777231 DOI: 10.21037/atm-23-393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 02/17/2023] [Indexed: 02/03/2024]
Affiliation(s)
- Max S Mano
- Grupo Oncoclínicas, Centro Paulista de Oncologia, Sao Paulo, Brazil
| |
Collapse
|
35
|
Quiroga D, Wesolowski R, Zelinskas S, Pinette A, Benner B, Schwarz E, Savardekar H, Johnson C, Stiff A, Yu L, Macrae E, Lustberg M, Mrozek E, Ramaswamy B, Carson WE. An Open-Label Study of Subcutaneous CpG Oligodeoxynucleotide (PF03512676) in Combination with Trastuzumab in Patients with Metastatic HER2+ Breast Cancer. Cancer Control 2024; 31:10732748241250189. [PMID: 38797949 PMCID: PMC11129578 DOI: 10.1177/10732748241250189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 03/18/2024] [Accepted: 04/10/2024] [Indexed: 05/29/2024] Open
Abstract
OBJECTIVES CpG ODN is a Toll-like receptor 9 agonist with immunotherapeutic potential for many cancer types, including aggressive breast cancers. There is strong interest in utilizing CpG ODN as an adjuvant to improve clinical efficacy of current treatments and immunogenicity of breast cancers not traditionally responsive to active immunotherapy, such as those that are human epidermal growth factor receptor 2 (HER2)-positive. This study aimed to study the efficacy and safety of combination CpG ODN plus anti-HER2 antibody trastuzumab treatment in patients with advanced/metastatic breast cancer. METHODS This single-arm, open-label phase II clinical trial treated patients (n = 6) with advanced/metastatic HER2-positive breast cancer with weekly subcutaneous CpG ODN and trastuzumab. Patients may have received any number of prior therapies to be enrolled (most enrolled at median 1 prior line of chemotherapy). Peripheral blood was collected at baseline and weeks 2, 6, 12, and 18 for immune analyses. Six patients were enrolled and 50% achieved stable disease (SD) response. RESULTS Median PFS was 8.3 months. Three of the six patients enrolled opted to stop treatment due to tolerability issues. Multiplex assay for cytokine measurements revealed significantly higher VEGF-D levels at week 2 compared to baseline. Peripheral blood mononuclear cells analyzed by flow cytometry showed a significant increase in monocytic MDSC between weeks 6 and 12. Patients with progressive disease tended to have higher levels of week 6 monocytic MDSC and PD-1+ T cells than patients with SD. NK cell populations did not significantly change throughout treatment. CONCLUSIONS CpG ODN and trastuzumab treatment of metastatic HER2 + breast cancer was safe but was not tolerable for all patients. This combination did induce potentially predictive immune profile changes in treated patients with metastatic HER2 + breast cancer, the significance of which needs to be further explored.
Collapse
Affiliation(s)
- Dionisia Quiroga
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Robert Wesolowski
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Sara Zelinskas
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ashley Pinette
- Department of Surgery, The Ohio State University, Columbus, OH, USA
- Miami Valley Hospital, Dayton, OH, USA
| | - Brooke Benner
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Emily Schwarz
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Himanshu Savardekar
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Courtney Johnson
- Department of Surgery, The Ohio State University, Columbus, OH, USA
- Miami Valley Hospital, Dayton, OH, USA
| | - Andrew Stiff
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Erin Macrae
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
- Columbus Oncology Associates, Columbus, OH, USA
| | - Maryam Lustberg
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
- Yale School of Medicine, New Haven, CN, USA
| | - Ewa Mrozek
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
- St. Rita’s Cancer Center, Lima, OH, USA
| | - Bhuvaneswari Ramaswamy
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - William E. Carson
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
36
|
Wang X, Wang Y, Chen X, He Y, Zhou X, Jiao S, Zhu Z, Wu C, Bao J. Identification of glycogene-based prognostic signature and validation of B3GNT7 as a potential biomarker and therapeutic target in breast cancer. J Cancer Res Clin Oncol 2023; 149:16957-16969. [PMID: 37740763 DOI: 10.1007/s00432-023-05345-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/24/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND Breast cancer is the most common cancer worldwide, with the fifth highest mortality rate among all cancers and high risk of metastasis. However, potential biomarkers and molecular mechanisms underlying the stratification of breast cancer in terms of clinical outcomes remain to be investigated. Therefore, we aimed to find a novel prognostic biomarker and therapeutic target for breast cancer patients. METHODS Unsupervised hierarchical clustering was used to perform comprehensive transcriptomic study of total 185 glycogenes in public datasets of breast cancer with clinicopathological and survival information. A glycogene-based signature for subtype classification was discovered using Limma packages, and relevance to four known molecular features was identified by GSVA. Experimental verification was performed and biological functions of B3GNT7 were characterized by quantitative RT-PCR, western blot, transwell assays, and lectin immunofluorescence staining in breast cancer cells. RESULTS A 23-glycogene signature was identified for the classification of breast cancer. Among the 23 glycogenes, B3GNTs showed significantly positive associations with ER-/Her2- subtype in breast cancer patients (n = 2655). Overexpressed B3GNT7 were correlated with poor prognosis in breast cancer patients based on public datasets. B3GNT7 depletion inhibited cell proliferation, migration, and invasion, and decreased global fucosylation in MDA-MB-231 and HCC1937 breast cancer cells. CONCLUSIONS Herein, we discovered a unique 23-gene signature for breast cancer patient glycogene-type classification. Among these genes, B3GNT7 was shown to be a potential biomarker for unfavorable outcomes and therapeutic target of breast cancer.
Collapse
Affiliation(s)
- Xin Wang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Yida Wang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Xuanming Chen
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Yufei He
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Xunyu Zhou
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Sitong Jiao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Zilin Zhu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Chuanfang Wu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China.
| | - Jinku Bao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China.
| |
Collapse
|
37
|
Rediti M, Fernandez-Martinez A, Venet D, Rothé F, Hoadley KA, Parker JS, Singh B, Campbell JD, Ballman KV, Hillman DW, Winer EP, El-Abed S, Piccart M, Di Cosimo S, Symmans WF, Krop IE, Salgado R, Loi S, Pusztai L, Perou CM, Carey LA, Sotiriou C. Immunological and clinicopathological features predict HER2-positive breast cancer prognosis in the neoadjuvant NeoALTTO and CALGB 40601 randomized trials. Nat Commun 2023; 14:7053. [PMID: 37923752 PMCID: PMC10624889 DOI: 10.1038/s41467-023-42635-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 10/16/2023] [Indexed: 11/06/2023] Open
Abstract
The identification of prognostic markers in patients receiving neoadjuvant therapy is crucial for treatment optimization in HER2-positive breast cancer, with the immune microenvironment being a key factor. Here, we investigate the complexity of B and T cell receptor (BCR and TCR) repertoires in the context of two phase III trials, NeoALTTO and CALGB 40601, evaluating neoadjuvant paclitaxel with trastuzumab and/or lapatinib in women with HER2-positive breast cancer. BCR features, particularly the number of reads and clones, evenness and Gini index, are heterogeneous according to hormone receptor status and PAM50 subtypes. Moreover, BCR measures describing clonal expansion, namely evenness and Gini index, are independent prognostic factors. We present a model developed in NeoALTTO and validated in CALGB 40601 that can predict event-free survival (EFS) by integrating hormone receptor and clinical nodal status, breast pathological complete response (pCR), stromal tumor-infiltrating lymphocyte levels (%) and BCR repertoire evenness. A prognostic score derived from the model and including those variables, HER2-EveNT, allows the identification of patients with 5-year EFS > 90%, and, in those not achieving pCR, of a subgroup of immune-enriched tumors with an excellent outcome despite residual disease.
Collapse
Affiliation(s)
- Mattia Rediti
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - David Venet
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Françoise Rothé
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Katherine A Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | | | - Jordan D Campbell
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| | - Karla V Ballman
- Alliance Statistics and Data Management Center, Weill Cornell Medicine, New York, NY, USA
| | - David W Hillman
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| | - Eric P Winer
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | | | - Martine Piccart
- Medical Oncology Department, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Serena Di Cosimo
- Integrated biology platform unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - William Fraser Symmans
- Department of Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ian E Krop
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA Ziekenhuizen, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Sherene Loi
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lajos Pusztai
- Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Lisa A Carey
- Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
38
|
Giffoni de Mello Morais Mata D, Chehade R, Hannouf MB, Raphael J, Blanchette P, Al-Humiqani A, Ray M. Appraisal of Systemic Treatment Strategies in Early HER2-Positive Breast Cancer-A Literature Review. Cancers (Basel) 2023; 15:4336. [PMID: 37686612 PMCID: PMC10486709 DOI: 10.3390/cancers15174336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND The overexpression of the human epidermal growth factor receptor 2 (HER2+) accounts for 15-20% of all breast cancer phenotypes. Even after the completion of the standard combination of chemotherapy and trastuzumab, relapse events occur in approximately 15% of cases. The neoadjuvant approach has multiple benefits that include the potential to downgrade staging and convert previously unresectable tumors to operable tumors. In addition, achieving a pathologic complete response (pCR) following preoperative systemic treatment is prognostic of enhanced survival outcomes. Thus, optimal evaluation among the suitable strategies is crucial in deciding which patients should be selected for the neoadjuvant approach. METHODS A literature search was conducted in the Embase, Medline, and Cochrane electronic libraries. CONCLUSION The evaluation of tumor and LN staging and, hence, stratifying BC recurrence risk are decisive factors in guiding clinicians to optimize treatment decisions between the neoadjuvant versus adjuvant approaches. For each individual case, it is important to consider the most likely postsurgical outcome, since, if the patient does not obtain pCR following neoadjuvant treatment, they are eligible for adjuvant T-DM1 in the case of residual disease. This review of HER2-positive female BC outlines suitable neoadjuvant and adjuvant systemic treatment strategies for guiding clinical decision making around the selection of an appropriate therapy.
Collapse
Affiliation(s)
- Danilo Giffoni de Mello Morais Mata
- Division of Medical Oncology, London Regional Cancer Program, London Health Sciences Centre, Western University, London, ON N6A 5W9, Canada; (J.R.); (P.B.)
| | - Rania Chehade
- Division of Medical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada; (R.C.); (A.A.-H.)
| | - Malek B. Hannouf
- Department of Internal Medicine, Western University, London, ON N6A 3K7, Canada;
| | - Jacques Raphael
- Division of Medical Oncology, London Regional Cancer Program, London Health Sciences Centre, Western University, London, ON N6A 5W9, Canada; (J.R.); (P.B.)
| | - Phillip Blanchette
- Division of Medical Oncology, London Regional Cancer Program, London Health Sciences Centre, Western University, London, ON N6A 5W9, Canada; (J.R.); (P.B.)
| | - Abdullah Al-Humiqani
- Division of Medical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada; (R.C.); (A.A.-H.)
| | - Monali Ray
- Division of Medical Oncology, Markham Stouffville Hospital, Markham, ON L3P 7P3, Canada;
| |
Collapse
|
39
|
Choi H, Ahn SG, Bae SJ, Kim JH, Eun NL, Lee Y, Nahm JH, Jeong J, Cha YJ. Comparison of Programmed Cell Death Ligand 1 Status between Core Needle Biopsy and Surgical Specimens of Triple-Negative Breast Cancer. Yonsei Med J 2023; 64:518-525. [PMID: 37488704 PMCID: PMC10375241 DOI: 10.3349/ymj.2023.0090] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/26/2023] Open
Abstract
PURPOSE Pembrolizumab is currently used to treat advanced triple-negative breast cancer (TNBC) and high-risk early TNBC with neoadjuvant chemotherapy (NAC). The tumor-infiltrating lymphocyte (TIL) level and programmed cell death ligand 1 (PD-L1) status are predictors of response to NAC and immune checkpoint inhibitor treatment. We aimed to investigate whether the PD-L1 status in core needle biopsies (CNBs) could represent the whole tumor in TNBC. MATERIALS AND METHODS A total of 49 patients diagnosed with TNBC who received upfront surgery without NAC between January 2018 and March 2021 were included. The PD-L1 expression (SP142 and 22C3 clones) and TIL were evaluated in paired CNBs and resected specimens. The concordance PD-L1 status and TIL levels between CNBs and resected specimens were analyzed. RESULTS PD-L1 positivity was more frequently observed in resected specimens. The overall reliability of TIL level in the CNB was good [intraclass correlation coefficient (ICC)=0.847, p<0.001]. The agreements of PD-L1 status were good and fair, respectively (SP142, κ=0.503, p<0.001; 22C3, κ=0.380, p=0.010). As the core number of CNB increased, the reliability and agreement also improved, especially from five tumor cores (TIL, ICC=0.911, p<0.001; PD-L1 [22C3], κ=0.750, p=0.028). Regarding PD-L1 (SP142), no further improvement was observed with ≥5 tumor cores (κ=0.600, p=0.058). CONCLUSION CNBs with ≥5 tumor cores were sufficient to represent the TIL level and PD-L1 (22C3) status in TNBC.
Collapse
Affiliation(s)
- Hyungwook Choi
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Gwe Ahn
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Soong Joon Bae
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jee Hung Kim
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Na Lae Eun
- Department of Radiology, Yonsei University College of Medicine, Seoul, Korea
| | - Yangkyu Lee
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hae Nahm
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Joon Jeong
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Jin Cha
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
40
|
Jääskeläinen MM, Tiainen S, Siiskonen H, Ahtiainen M, Kuopio T, Rönkä A, Kettunen T, Hämäläinen K, Rilla K, Harvima I, Mannermaa A, Auvinen P. The prognostic and predictive role of tumor-infiltrating lymphocytes (FoxP3 + and CD8 +) and tumor-associated macrophages in early HER2 + breast cancer. Breast Cancer Res Treat 2023:10.1007/s10549-023-07017-8. [PMID: 37428418 PMCID: PMC10361875 DOI: 10.1007/s10549-023-07017-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/19/2023] [Indexed: 07/11/2023]
Abstract
PURPOSE In HER2-positive (HER2 +) breast cancer, tumor-infiltrating lymphocytes (TILs) and tumor-associated macrophages (TAMs) may influence the efficacy of the HER2-antibody trastuzumab and the patient's outcome. In this HER2 + patient cohort, our aim was to study the numbers of FoxP3 + regulatory TILs and CD8 + cytotoxic TILs, their correlations with CD68 + and CD163 + TAMs, and the prognostic and predictive value of the studied factors. METHODS We evaluated 139 non-metastatic HER2 + breast cancer patients operated between 2001 and 2008. The FoxP3+TIL count (FoxP3+TILs) was assessed using the hotspot method, and the CD8 + TIL count (CD8+mTILs) utilizing a digital image analysis from invasive margin areas. The ratios between CD8+mTILs and FoxP3+TILs as well as CD8+mTILs and TAMs were calculated. RESULTS FoxP3 + TILs and CD8 + mTILs correlated positively with each other (p<0.001). FoxP3+TILs had a positive correlation with CD68+and CD163+TAMs (p≤0.038), while CD8 + mTILs correlated only with CD68+TAMs (p<0.001). In the HER2 + and hormone receptor-positive Luminal B subgroup, high numbers of FoxP3+TILs were associated with shorter disease-free survival (DFS) (54% vs. 79%, p = 0.040). The benefit from adjuvant trastuzumab was extremely significant among patients with a high CD8 + mTILs/CD68 + TAMs ratio, with overall survival (OS) 84% vs. 33% (p = 0.003) and breast cancer-specific survival (BCSS) 88% vs. 48% (p = 0.009) among patients treated with or without trastuzumab, respectively. CONCLUSION In the HER2 + Luminal B subgroup, high FoxP3 + TILs were associated with shorter DFS. A high CD8 + mTILs/CD68 + TAMs ratio seems to associate with impressive efficacy of trastuzumab.
Collapse
Affiliation(s)
- Minna M Jääskeläinen
- Cancer Center, Kuopio University Hospital, Northern Savonia Healthcare Municipality, P.O.Box 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Satu Tiainen
- Cancer Center, Kuopio University Hospital, Northern Savonia Healthcare Municipality, P.O.Box 100, 70029, Kuopio, Finland.
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.
| | - Hanna Siiskonen
- Imaging Center, Clinical Pathology, Kuopio University Hospital, Northern Savonia Healthcare Municipality, Kuopio, Finland
| | - Maarit Ahtiainen
- Department of Pathology, Central Finland Hospital Nova, Jyväskylä, Finland
| | - Teijo Kuopio
- Department of Pathology, Central Finland Hospital Nova, Jyväskylä, Finland
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Aino Rönkä
- Cancer Center, Kuopio University Hospital, Northern Savonia Healthcare Municipality, P.O.Box 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tiia Kettunen
- Cancer Center, Kuopio University Hospital, Northern Savonia Healthcare Municipality, P.O.Box 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Kirsi Hämäläinen
- Imaging Center, Clinical Pathology, Kuopio University Hospital, Northern Savonia Healthcare Municipality, Kuopio, Finland
- Institute of Clinical Medicine, Clinical Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Biocenter Kuopio and Cancer Center of Eastern Finland, University of Eastern Finland, Kuopio, Finland
| | - Kirsi Rilla
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Ilkka Harvima
- Department of Dermatology, Kuopio University Hospital, Northern Savonia Healthcare Municipality and University of Eastern Finland, Kuopio, Finland
| | - Arto Mannermaa
- Institute of Clinical Medicine, Clinical Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, Northern Savonia Healthcare Municipality, Kuopio, Finland
| | - Päivi Auvinen
- Cancer Center, Kuopio University Hospital, Northern Savonia Healthcare Municipality, P.O.Box 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
41
|
Fernandez-Martinez A, Pascual T, Singh B, Nuciforo P, Rashid NU, Ballman KV, Campbell JD, Hoadley KA, Spears PA, Pare L, Brasó-Maristany F, Chic N, Krop I, Partridge A, Cortés J, Llombart-Cussac A, Prat A, Perou CM, Carey LA. Prognostic and Predictive Value of Immune-Related Gene Expression Signatures vs Tumor-Infiltrating Lymphocytes in Early-Stage ERBB2/HER2-Positive Breast Cancer: A Correlative Analysis of the CALGB 40601 and PAMELA Trials. JAMA Oncol 2023; 9:490-499. [PMID: 36602784 PMCID: PMC9857319 DOI: 10.1001/jamaoncol.2022.6288] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/21/2022] [Indexed: 01/06/2023]
Abstract
Importance Both tumor-infiltrating lymphocytes (TILs) assessment and immune-related gene expression signatures by RNA profiling predict higher pathologic complete response (pCR) and improved event-free survival (EFS) in patients with early-stage ERBB2/HER2-positive breast cancer. However, whether these 2 measures of immune activation provide similar or additive prognostic value is not known. Objective To examine the prognostic ability of TILs and immune-related gene expression signatures, alone and in combination, to predict pCR and EFS in patients with early-stage ERBB2/HER2-positive breast cancer treated in 2 clinical trials. Design, Setting, and Participants In this prognostic study, a correlative analysis was performed on the Cancer and Leukemia Group B (CALGB) 40601 trial and the PAMELA trial. In the CALGB 40601 trial, 305 patients were randomly assigned to weekly paclitaxel with trastuzumab, lapatinib, or both for 16 weeks. The primary end point was pCR, with a secondary end point of EFS. In the PAMELA trial, 151 patients received neoadjuvant treatment with trastuzumab and lapatinib for 18 weeks. The primary end point was the ability of the HER2-enriched subtype to predict pCR. The studies were conducted from October 2013 to November 2015 (PAMELA) and from December 2008 to February 2012 (CALGB 40601). Data analyses were performed from June 1, 2020, to January 1, 2022. Main Outcomes and Measures Immune-related gene expression profiling by RNA sequencing and TILs were assessed on 230 CALGB 40601 trial pretreatment tumors and 138 PAMELA trial pretreatment tumors. The association of these biomarkers with pCR (CALGB 40601 and PAMELA) and EFS (CALGB 40601) was studied by logistic regression and Cox analyses. Results The median age of the patients was 50 years (IQR, 42-50 years), and 305 (100%) were women. Of 202 immune signatures tested, 166 (82.2%) were significantly correlated with TILs. In both trials combined, TILs were significantly associated with pCR (odds ratio, 1.01; 95% CI, 1.01-1.02; P = .02). In addition to TILs, 36 immune signatures were significantly associated with higher pCR rates. Seven of these signatures outperformed TILs for predicting pCR, 6 of which were B-cell related. In a multivariable Cox model adjusted for clinicopathologic factors, including PAM50 intrinsic tumor subtype, the immunoglobulin G signature, but not TILs, was independently associated with EFS (immunoglobulin G signature-adjusted hazard ratio, 0.63; 95% CI, 0.42-0.93; P = .02; TIL-adjusted hazard ratio, 1.00; 95% CI, 0.98-1.02; P = .99). Conclusions and Relevance Results of this study suggest that multiple B-cell-related signatures were more strongly associated with pCR and EFS than TILs, which largely represent T cells. When both TILs and gene expression are available, the prognostic value of immune-related signatures appears to be superior.
Collapse
Affiliation(s)
- Aranzazu Fernandez-Martinez
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
- Department of Genetics, University of North Carolina, Chapel Hill
| | - Tomás Pascual
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- SOLTI Breast Cancer Cooperative Group, Barcelona, Spain
| | - Baljit Singh
- Department of Pathology, White Plains Hospital, White Plains, New York
| | - Paolo Nuciforo
- Molecular Oncology Laboratory, Vall d’Hebron Institute of Oncology, Barcelona, Spain
| | - Naim U. Rashid
- Department of Biostatistics, University of North Carolina, Chapel Hill
| | - Karla V. Ballman
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota
| | - Jordan D. Campbell
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota
| | - Katherine A. Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
- Department of Genetics, University of North Carolina, Chapel Hill
| | - Patricia A. Spears
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
| | | | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Nuria Chic
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- SOLTI Breast Cancer Cooperative Group, Barcelona, Spain
| | - Ian Krop
- Yale Cancer Center, New Haven, Connecticut
| | - Ann Partridge
- Department of Breast Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Javier Cortés
- International Breast Cancer Center, Barcelona, Spain
| | | | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- SOLTI Breast Cancer Cooperative Group, Barcelona, Spain
- Reveal Genomics, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Breast Cancer Unit, IOB-QuirónSalud, Barcelona, Spain
| | - Charles M. Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
- Department of Genetics, University of North Carolina, Chapel Hill
| | - Lisa A. Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill
| |
Collapse
|
42
|
Nolan E, Lindeman GJ, Visvader JE. Deciphering breast cancer: from biology to the clinic. Cell 2023; 186:1708-1728. [PMID: 36931265 DOI: 10.1016/j.cell.2023.01.040] [Citation(s) in RCA: 270] [Impact Index Per Article: 135.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/14/2023] [Accepted: 01/30/2023] [Indexed: 03/17/2023]
Abstract
Breast cancer remains a leading cause of cancer-related mortality in women, reflecting profound disease heterogeneity, metastasis, and therapeutic resistance. Over the last decade, genomic and transcriptomic data have been integrated on an unprecedented scale and revealed distinct cancer subtypes, critical molecular drivers, clonal evolutionary trajectories, and prognostic signatures. Furthermore, multi-dimensional integration of high-resolution single-cell and spatial technologies has highlighted the importance of the entire breast cancer ecosystem and the presence of distinct cellular "neighborhoods." Clinically, a plethora of new targeted therapies has emerged, now being rapidly incorporated into routine care. Resistance to therapy, however, remains a crucial challenge for the field.
Collapse
Affiliation(s)
- Emma Nolan
- Auckland Cancer Society Research Centre, University of Auckland, Auckland 1023, New Zealand
| | - Geoffrey J Lindeman
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC 3050, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Jane E Visvader
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|