1
|
Shen J, Ye X, Hou H, Wang Y. Efficacy and Safety of Immunochemotherapy in Advanced Triple-negative Breast Cancer: A Meta-analysis of Randomised Clinical Trials. Clin Oncol (R Coll Radiol) 2025; 40:103783. [PMID: 39955967 DOI: 10.1016/j.clon.2025.103783] [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: 07/02/2024] [Revised: 12/19/2024] [Accepted: 01/30/2025] [Indexed: 02/18/2025]
Abstract
AIMS Based on the existing controversial clinical research results, this study conducted a comprehensive meta-analysis of relevant literature to clarify the benefits of immunochemotherapy (ICT)-which combines immune checkpoint inhibitors and chemotherapy (CT)-for patients with advanced triple-negative breast cancer (aTNBC). MATERILAS AND METHODS A thorough literature search was conducted up to February 15, 2024. Subsequently, meta-analyses were performed to aggregate hazard ratios (HRs) for progression-free survival (PFS) and overall survival (OS), odds ratios (ORs) for objective response rate (ORR) and relative risks (RRs) for adverse events (AEs). RESULTS Six randomised clinical trials (RCTs) involving 3,105 patients met the inclusion criteria. In comparison with CT, ICT yielded significant enhancements in PFS (HR, 0.80; 95%CI: 0.73-0.87), OS (HR, 0.87; 95%CI: 0.80-0.96), and ORR (OR, 1.34; 95%CI: 1.15-1.55) in the intention-to-treat population. However, ICT also exhibited an increase in grade ≥3 AEs (RR, 1.11; 95%CI: 1.04-1.19) and severe AEs (RR, 1.40; 95%CI: 1.18-1.66). Subgroup analyses revealed that ICT significantly improved PFS (HR, 0.67; 95%CI: 0.58-0.77), OS (HR, 0.75; 95%CI: 0.64-0.87), and ORR (OR, 1.47; 95%CI: 1.16-1.84) within the PD-L1-positive subgroup, whereas no statistically significant differences were detected for PD-L1-negative population. CONCLUSION ICT demonstrates superior efficacy over conventional CT in the treatment of aTNBC, albeit accompanied by heightened toxicity. Notably, the assessment of PD-L1 status may serve as a valuable biomarker in discerning aTNBC patients who are particularly predisposed to derive benefit from ICT. PROSPERO NUMBER CRD42024513270.
Collapse
Affiliation(s)
- J Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - X Ye
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - H Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China.
| | - Y Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China.
| |
Collapse
|
2
|
Al-Sharabass EA, El-Houseini ME, Effat H, Ibrahim SA, Abdellateif MS. The clinical potential of PDL-1 pathway and some related micro-RNAs as promising diagnostic markers for breast cancer. Mol Med 2025; 31:106. [PMID: 40108523 PMCID: PMC11921724 DOI: 10.1186/s10020-025-01137-1] [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: 07/31/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Immune checkpoint pathways play important roles in breast cancer (BC) pathogenesis and therapy. METHODS Expression levels of programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed death-ligand 1 (PD-L1), Forkhead box P3 (FOXP3), miR-155, and miR-195 were assessed in the peripheral blood of 90 BC patients compared to 30 healthy controls using quantitative real-time PCR (qRt-PCR). The plasma level of soluble MHC class I chain related-protein B (MIC-B) protein was assessed using the enzyme linked immunosorbent assay (ELISA) technique. The data were correlated to the clinico-pathological characteristics of the patients. RESULTS There was a significant increase in the expression levels of PDL-1 [17.59 (3.24-123), p < 0.001], CTLA-4 [23.34 (1.3-1267), p = 0.006], PD-1 [10.25 (1-280), p < 0.001], FOXP3 [11.5 (1-234.8), p = 0.001], miR-155 [87.3 (1.5-910), p < 0.001] in BC patients compared to normal controls. The miR-195 was significantly downregulated in BC patients [0.23 (0-0.98, p < 0.001]. The plasma level of MIC-B was significantly increased in the BC patients [0.941 (0.204-6.38) ng/ml], compared to the control group [0.351 (0.211-0.884) ng/mL, p < 0.00]. PDL-1, CTLA-4, PD-1, and FOXP3 achieved a specificity of 100% for distinguishing BC patients, at a sensitivity of 93.3%, 82.2%, 62.2%, and 71.1% respectively. The combined expression of PDL-1 and CTLA-4 scored a 100% sensitivity and 100% specificity for diagnosing BC (p < 0.001). The sensitivity, specificity, and AUC of miR-155 were 88.9%, 96.7%, and 0.934; respectively (p < 0.001). While those of miR-195 were 73.3%, 60%, and 0.716; respectively (p = 0.001). MIC-B expression showed a 77.8% sensitivity, 80% specificity, and 0.811 AUC at a cutoff of 1.17 ng/ml (p < 0.001). Combined expression of miR-155 and miR-195 achieved a sensitivity of 91.1%, a specificity of 96.7%, and AUC of 0.926 (p < 0.001). Multivariate analysis showed that PDL-1 (OR:13.825, p = 0.004), CTLA-4 (OR: 20.958, p = 0.010), PD-1(OR:10.550, p = 0.044), MIC-B (OR: 17.89, p = 0.003), miR-155 (OR: 211.356, P < 0.001), and miR-195(OR:0.006, P < 0.001) were considered as independent risk factors for BC. CONCLUSIONS The PB levels of PDL-1, CTLA-4, PD-1, FOXP3, MIC-B, miR-155, and miR-195 could be used as promising diagnostic markers for BC patients.
Collapse
Affiliation(s)
| | - Motawa E El-Houseini
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Heba Effat
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
Collapse
|
3
|
Dent R, Cortés J, Park YH, Muñoz-Couselo E, Kim SB, Sohn J, Im SA, Holgado E, Foukakis T, Kümmel S, Yearley J, Wang A, Nebozhyn M, Huang L, Cristescu R, Jelinic P, Karantza V, Schmid P. Molecular determinants of response to neoadjuvant pembrolizumab plus chemotherapy in patients with high-risk, early-stage, triple-negative breast cancer: exploratory analysis of the open-label, multicohort phase 1b KEYNOTE-173 study. Breast Cancer Res 2025; 27:35. [PMID: 40069763 PMCID: PMC11895130 DOI: 10.1186/s13058-024-01946-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/09/2024] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The multicohort, open-label, phase 1b KEYNOTE-173 study was conducted to investigate pembrolizumab plus chemotherapy as neoadjuvant therapy for triple-negative breast cancer (TNBC). This exploratory analysis evaluated features of the tumor microenvironment that might be predictive of response. METHODS Cell fractions from 20 paired samples collected at baseline and after one cycle of neoadjuvant pembrolizumab prior to chemotherapy initiation were analyzed by spatial localization (tumor compartment, stromal compartment, or sum of tumor and stromal compartments [total tumor]) using three six-plex immunohistochemistry panels with T-cell, myeloid cell, and natural killer cell components. Area under the receiver operating characteristic curve (AUROC) was used to assess associations between immune subsets and gene expression signatures (T-cell-inflamed gene expression profile [TcellinfGEP] and 10 non-TcellinfGEP signatures using RNA sequencing) and pathologic complete response (pCR). RESULTS At baseline, six immune subsets quantitated within the tumor compartment showed AUROC with 95% CIs not crossing 0.5, including CD11c+ cells (macrophage and dendritic cell [DC]: AUROC, 0.85; 95% confidence interval [CI] 0.63-1.00), CD11c+/MHCII+/CD163-/CD68- cells (DC: 0.76; 95% CI, 0.53-0.99), CD11c+/MHCII-/CD163-/CD68- cells (nonactivated/immature DC: 0.80; 95% CI 0.54-1.00), and CD11c+/CD163+ cells (M2 macrophage: 0.77; 95% CI 0.55-0.99). Other associations with pCR included baseline CD11c+/MHCII-/CD163-/CD68- (nonactivated/immature DC) within the total tumor (AUROC, 0.76; 95% CI 0.51-1.00) and the baseline CD11c/CD3 ratio within the tumor compartment (0.75; 95% CI 0.52-0.98). Changes in immune subsets following one cycle of pembrolizumab were not strongly associated with pCR. Although T-cell associations were relatively weak, specific CD8 subsets trended toward association. The AUROC for discriminating pCR based on TcellinfGEP was 0.55 (95% CI 0.25-0.85); when detrended by TcellinfGEP, AUROC varied for the non-TcellinfGEP signatures. TcellinfGEP expression trended higher in responders than in nonresponders when evaluating pCR. CONCLUSIONS Myeloid cell populations within the tumor compartment at baseline and TcellinfGEP show a promising trend toward an association with pCR in a small subgroup of patients with early-stage TNBC treated with neoadjuvant pembrolizumab plus chemotherapy. TRIAL REGISTRATION ClinicalTrials.gov, NCT02622074; registration date, December 2, 2015.
Collapse
Affiliation(s)
- Rebecca Dent
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore, 168583, Singapore.
| | - Javier Cortés
- Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
- International Breast Cancer Center, Quironsalud Group, Barcelona, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, European University of Madrid, Madrid, Spain
| | - Yeon Hee Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eva Muñoz-Couselo
- Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron Hospital, Barcelona, Spain
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joohyuk Sohn
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Esther Holgado
- Medical Oncology Service, Ramón y Cajal University Hospital, Madrid, Spain
| | - Theodoros Foukakis
- Department of Oncology-Pathology, Karolinska Comprehensive Cancer Center, Karolinska Institute and Breast Cancer Centre, Cancer Theme, Karolinska University Hospital, Solna, Sweden
| | - Sherko Kümmel
- Interdisciplinary Breast Unit, Essen-Mitte Clinics, Essen, and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | - Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, London, UK
| |
Collapse
|
4
|
Khan Y, Rizvi S, Raza A, Khan A, Hussain S, Khan NU, Alshammari SO, Alshammari QA, Alshammari A, Ellakwa DES. Tailored therapies for triple-negative breast cancer: current landscape and future perceptions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03896-4. [PMID: 40029385 DOI: 10.1007/s00210-025-03896-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/07/2025] [Indexed: 03/05/2025]
Abstract
Triple-negative breast cancer (TNBC) has become one of the most challenging cancers to date due to its great variability in biological features, high growth rate, and rare options for treatment. This review examines several innovative strategies for tailored treatment of TNBC, focusing mainly on the most recent developments and potential directions. The molecular landscape of TNBC is covered in the first section, which keeps the focus on transcriptome and genomic profiling while highlighting key molecular targets like mutations in the BRCA1/2, PIK3CA, androgen receptors (AR), epidermal growth factor receptors (EGFR), and immunological checkpoint molecules. This review also covers novel therapies that aim to block well-defined pathways, including immune checkpoint inhibitors (ICI), EGFR inhibitors, drugs that target AR, poly ADP ribose polymerase (PARP) inhibitors, and drugs that disrupt the PI3K/AKT/mTOR pathway. Additionally, it covers novel strategies focusing on combination therapy, targeting the DNA damage response pathway, and epigenetic modulators. Conclusively, it emphasizes perspectives and directions on topics such as personalized medicine, artificial intelligence (AI), predictive biomarkers, and treatment planning with the inclusion of machine learning (ML).
Collapse
Affiliation(s)
- Yumna Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, 25130, Pakistan.
| | - Sana Rizvi
- Bakhtawar Amin Medical and Dental College, Bakhtawar Amin Trust Teaching Hospital, Multan, Pakistan
| | - Ali Raza
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Amna Khan
- Abbottabad International Medical Institute, Abbottabad, 22020, Pakistan
| | - Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248007, India
| | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, 25130, Pakistan
| | - Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, College of Pharmacy, Northern Border University, 76321, Rafha, Saudi Arabia
| | - Qamar A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Abdulkarim Alshammari
- Department of Clinical Practice, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Doha El-Sayed Ellakwa
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo, Egypt.
- Department of Biochemistry, Faculty of Pharmacy, Sinai University, Kantra Branch, Ismailia, Egypt.
| |
Collapse
|
5
|
Fu Y, Yang Q, Xu N, Zhang X. MiRNA affects the advancement of breast cancer by modulating the immune system's response. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167759. [PMID: 40037267 DOI: 10.1016/j.bbadis.2025.167759] [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: 10/29/2024] [Revised: 02/05/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Breast cancer (BC), which is the most common tumor in women, has greatly endangered women's lives and health. Currently, patients with BC receive comprehensive treatments, including surgery, chemotherapy, radiotherapy, endocrine therapy, and targeted therapy. According to the latest research, the development of BC is closely related to the inflammatory immune response, and the immunogenicity of BC has steadily been recognized. As such, immunotherapy is one of the promising and anticipated forms of treatment for BC. The potential values of miRNA in the diagnosis and prognosis of BC have been established, and aberrant expression of associated miRNA can either facilitate or inhibit progression of BC. In the tumor immune microenvironment (TME), miRNAs are considered to be an essential molecular mechanism by which tumor cells interact with immunocytes and immunologic factors. Aberrant expression of miRNAs results in reprogramming of tumor cells actively, which may suppress the generation and activation of immunocytes and immunologic factors, avoid tumor cells apoptosis, and ultimately result in uncontrolled proliferation and deterioration. Therefore, through activating and regulating the immunocytes related to tumors and associated immunologic factors, miRNA can contribute to the advancement of BC. In this review, we assessed the function of miRNA and associated immune system components in regulating the advancement of BC, as well as the potential and viability of using miRNA in immunotherapy for BC.
Collapse
Affiliation(s)
- Yeqin Fu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
| | - Qiuhui Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 310006, China
| | - Ning Xu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Xiping Zhang
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
6
|
Andour L, Hagenaars SC, Gregus B, Tőkes AM, Karancsi Z, Tollenaar RAEM, Kroep JR, Kulka J, Mesker WE. The prognostic value of the tumor-stroma ratio compared to tumor-infiltrating lymphocytes in triple-negative breast cancer: a review. Virchows Arch 2025; 486:427-444. [PMID: 39904885 PMCID: PMC11950021 DOI: 10.1007/s00428-025-04039-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: 10/21/2024] [Revised: 12/17/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Previous literature extensively explored biomarkers to personalize treatment for breast cancer patients. The clinical need is especially high in patients with triple-negative breast cancer (TNBC) due to its aggressive nature and limited treatment modalities. This review aims to evaluate the value of tumor-infiltrating lymphocytes (TILs) and tumor-stroma ratio (TSR) as prognostic biomarkers in TNBC patients and assess their clinical potential. A literature search was conducted in PubMed, Embase, Emcare, Web of Science, and Cochrane Library. Papers comparing survival outcomes of TNBC patients with low/high or negative/positive TSR and immune cells were included. The most frequently mentioned subgroups of TILs were selected and reported in this review. Data from 43 articles on TILs and eight articles on TSR were included. Among TNBC patients, high CD8 expression was generally associated with better survival. Notable, the poor survival outcomes were related to high intra-tumoral PD-L1 expression, whereas high stromal PD-L1 expression more often was correlated with favorable outcomes. For the TSR, a high amount of stroma in the primary tumor of TNBC patients was consistently associated with worse survival. This review highlights that a high number of CD8-positive T-cells is a promising prognostic factor for TNBC patients. PD-L1 expression analyzed for intra-tumoral and stromal expression separately reports strong but contrasting information. Finally, the TSR shows potential to be an important prognostic marker, especially for TNBC patients. Utilizing both biomarkers, either on itself or combined, could enhance clinical decision-making and personalization of treatment.
Collapse
Affiliation(s)
- Layla Andour
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Sophie C Hagenaars
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Barbara Gregus
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Mária Tőkes
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Zsófia Karancsi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
7
|
Guo Z, Zhu Z, Lin X, Wang S, Wen Y, Wang L, Zhi L, Zhou J. Tumor microenvironment and immunotherapy for triple-negative breast cancer. Biomark Res 2024; 12:166. [PMID: 39741315 DOI: 10.1186/s40364-024-00714-6] [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: 10/16/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer known for its high aggressiveness and poor prognosis. Conventional treatment of TNBC is challenging due to its heterogeneity and lack of clear targets. Recent advancements in immunotherapy have shown promise in treating TNBC, with immune checkpoint therapy playing a significant role in comprehensive treatment plans. The tumor microenvironment (TME), comprising immune cells, stromal cells, and various cytokines, plays a crucial role in TNBC progression and response to immunotherapy. The high presence of tumor-infiltrating lymphocytes and immune checkpoint proteins in TNBC indicates the potential of immunotherapeutic strategies. However, the complexity of the TME, while offering therapeutic targets, requires further exploration of its multiple roles in immunotherapy. In this review, we discuss the interaction mechanism between TME and TNBC immunotherapy based on the characteristics and composition of TME, and elaborate on and analyze the effect of TME on immunotherapy, the potential of TME as an immune target, and the ability of TME as a biomarker. Understanding these dynamics will offer new insights for enhancing therapeutic approaches and investigating stratification and prognostic markers for TNBC patients.
Collapse
Affiliation(s)
- Zijie Guo
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Ziyu Zhu
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Xixi Lin
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Shenkangle Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Yihong Wen
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Linbo Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Lili Zhi
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Jichun Zhou
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
8
|
Heater NK, Warrior S, Lu J. Current and future immunotherapy for breast cancer. J Hematol Oncol 2024; 17:131. [PMID: 39722028 DOI: 10.1186/s13045-024-01649-z] [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: 09/24/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Substantial therapeutic advancement has been made in the field of immunotherapy in breast cancer. The immune checkpoint inhibitor pembrolizumab in combination with chemotherapy received FDA approval for both PD-L1 positive metastatic and early-stage triple-negative breast cancer, while ongoing clinical trials seek to expand the current treatment landscape for immune checkpoint inhibitors in hormone receptor positive and HER2 positive breast cancer. Antibody drug conjugates are FDA approved for triple negative and HER2+ disease, and are being studied in combination with immune checkpoint inhibitors. Vaccines and bispecific antibodies are areas of active research. Studies of cellular therapies such as tumor infiltrating lymphocytes, chimeric antigen receptor-T cells and T cell receptor engineered cells are promising and ongoing. This review provides an update of recent major clinical trials of immunotherapy in breast cancer and discusses future directions in the treatment of breast cancer.
Collapse
Affiliation(s)
- Natalie K Heater
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, 60611, USA
| | - Surbhi Warrior
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 676 N St. Clair, Suite 850, Chicago, IL, 60611, USA
| | - Janice Lu
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 676 N St. Clair, Suite 850, Chicago, IL, 60611, USA.
| |
Collapse
|
9
|
Lin S, Fu B, Khan M. Identifying subgroups deriving the most benefit from PD-1 checkpoint inhibition plus chemotherapy in advanced metastatic triple-negative breast cancer: a systematic review and meta-analysis. World J Surg Oncol 2024; 22:346. [PMID: 39709499 DOI: 10.1186/s12957-024-03424-3] [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: 03/22/2024] [Accepted: 05/21/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND The combination of immunotherapy and chemotherapy has demonstrated an enhancement in progression-free survival (PFS) for individuals with advanced and metastatic triple-negative breast cancer (TNBC) when compared to the use of chemotherapy alone. Nevertheless, the extent to which different subgroups of metastatic TNBC patients experience this benefit remains uncertain. OBJECTIVES Our objective was to conduct subgroup analyses to more precisely identify the factors influencing these outcomes. MATERIALS AND METHODS The PubMed database was searched until Dec 2023 for studies that compared PD-1 checkpoint inhibitors plus chemotherapy (ICT) with chemotherapy (CT) alone. The primary outcome of interest was progression-free survival (PFS). Review Manager (RevMan) version 5.4. was used for the data analysis. RESULTS Four randomized controlled trials (RCTs) comprising 2468 advanced and metastatic TNBC were included in this systematic review and meta-analysis. PFS surge with combined therapy was observed in White (HR 0.80 [0.70, 0.91], p = 0.0007) and Asian ethnicities (HR 0.73 [0.58, 0.93], p = 0.01) but not in Blacks (HR 0.72 [0.42, 1.24], p = 0.24). Overall, patients with distant metastasis demonstrated to derive the PFS benefit from additional immunotherapy (HR 0.87 [0.77, 0.99], p = 0.03); however, metastasis to individual distant site was associated with failure to achieve any treatment difference (Bone: HR 0.79 [0.41, 1.52], p = 0.49; Lung: HR 0.85 [0.70, 1.04], p = 0.11; Liver: HR 0.80 [0.64, 1.01], p = 0.06). While number of metastases > 3 also showed to impact the PFS advantage (HR 0.89 [0.69, 1.16], p = 0.39). While patients, regardless of prior chemotherapy, experienced a notable enhancement in PFS with ICT (Overall: HR 0.79 [0.71, 0.88], p < 0.0001; Yes: HR 0.87 [0.76, 1.00], p = 0.05; No: HR 0.67 [0.56, 0.80], p < 0.00001), those previously exposed to chemotherapy exhibited a significantly smaller PFS advantage compared to those without prior chemotherapy, as evidenced by a significant subgroup difference (Test for subgroup difference: P = 0.02, I2 = 82.2%). Patients lacking PD-L1 expression also failed to achieve any additional benefit from immunotherapy (PD-L1-: HR 0.95 [0.81, 1.12]; p = 0.54; PD-L1+: HR 0.73 [0.64, 0.85], p < 0.0001). Age, ECOG status, and presentation with de novo metastasis/recurrent shown no impact on IT-associated PFS advantage. CONCLUSIONS Patient- and treatment- related factors such as ethnicity, distant metastases, number of metastases (> 3), previous exposure to chemotherapy and PD-L1 expression, seem to influence or restrict the advantage in progression-free survival associated with the addition of immunotherapy to chemotherapy, as opposed to chemotherapy alone, in patients with advanced and metastatic TNBC. Larger studies are warranted to validate these outcomes.
Collapse
Affiliation(s)
- Shengfa Lin
- Department of Oncology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510168, People's Republic of China
| | - Bihe Fu
- Department of Oncology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510168, People's Republic of China
| | - Muhammad Khan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou, Guangdong, 510095, People's Republic of China.
| |
Collapse
|
10
|
Wang Y, Liu L, Graff SL, Cheng L. Recent advancements in biomarkers and molecular diagnostics in hormonal receptor-positive breast cancer. Histopathology 2024. [PMID: 39687977 DOI: 10.1111/his.15395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Molecular applications have limited use in breast cancer compared to other cancer types. In recent years, with an improving appreciation of the molecular genetics of breast cancer and innovative novel targeted and immune-mediated therapeutics, opportunities have arisen for more biomarker analysis and molecular applications in the diagnosis and treatment of both locally advanced and metastatic breast cancers. In hormone receptor-positive, HER2-negative breast cancers, a growing number of revolutionized personalized therapies are in clinical use or on trials, such as CDK4/6 inhibitors and immune checkpoint inhibitors in adjuvant and neoadjuvant settings, and PIK3CA inhibitors in metastatic disease. In this review, we focus on biomarkers associated with those new therapeutic targets and molecular applications for genetic alterations associated with drug resistance or interaction from a pathology perspective for selecting and optimizing breast cancer treatment.
Collapse
Affiliation(s)
- Yihong Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Liu Liu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Stephanie L Graff
- Division of Medical Oncology, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
11
|
Pont M, Marqués M, Sorolla A. Latest Therapeutical Approaches for Triple-Negative Breast Cancer: From Preclinical to Clinical Research. Int J Mol Sci 2024; 25:13518. [PMID: 39769279 PMCID: PMC11676458 DOI: 10.3390/ijms252413518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) represents roughly one-sixth of all breast cancer patients, but accounts for 30-40% of breast cancer deaths. Due to the lack of typical biomarkers exploited clinically for breast cancer, it remains very difficult to treat. Moreover, its intrinsic high heterogeneity and proneness to become resistant to the drugs administered makes the treatment management very challenging for oncologists. Herein, we outline the different therapies used currently for TNBC and list the ongoing clinical trials to provide an overview of the most recent TNBC therapeutic landscape. In addition, we highlight the emerging therapies in the preclinical stage that hold the most promise, such as epigenetic modulators, CRISPR, miniproteins, radioconjugates, cancer vaccines, and PROTACs. Moreover, we navigate through the existing limitations and challenges which hamper the development of new and more effective treatments for TNBC. Lastly, we point to emerging new directions that may revolutionize future therapy for TNBC.
Collapse
Affiliation(s)
- Mariona Pont
- Research Group of Cancer Biomarkers, Biomedical Research Institute of Lleida (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.P.); (M.M.)
- Department of Medicine, University of Lleida, Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain
| | - Marta Marqués
- Research Group of Cancer Biomarkers, Biomedical Research Institute of Lleida (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.P.); (M.M.)
- Department of Medicine, University of Lleida, Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain
| | - Anabel Sorolla
- Research Group of Cancer Biomarkers, Biomedical Research Institute of Lleida (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198 Lleida, Spain; (M.P.); (M.M.)
| |
Collapse
|
12
|
Shi Y, Guo Z, Wang Q, Deng H. Prognostic value of tumor-infiltrating lymphocyte subtypes and microorganisms in triple-negative breast cancer. J Cancer Res Ther 2024; 20:1983-1990. [PMID: 39792407 DOI: 10.4103/jcrt.jcrt_41_24] [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: 01/05/2024] [Accepted: 09/02/2024] [Indexed: 01/12/2025]
Abstract
ABSTRACT Tumor-infiltrating lymphocytes (TILs) are key components of the tumor microenvironment (TME) and serve as prognostic markers for breast cancer. Patients with high TIL infiltration generally experience better clinical outcomes and extended survival compared to those with low TIL infiltration. However, as the TME is highly complex and TIL subtypes perform distinct biological functions, TILs may only provide an approximate indication of tumor immune status, potentially leading to biased prognostic results. Therefore, we reviewed the interactions between immune-infiltrating subtypes and tumor cells throughout the entire TME. By examining the antitumor or protumor effects of each TIL subtype, we aimed to better characterize the tumor immune landscape, offering more accurate and comprehensive insights for guiding triple-negative breast cancer (TNBC) treatment. In addition, this approach could lead to the development of new therapeutic targets, enabling tailored treatment strategies and precision medicine. Accumulating evidence suggests that the intestinal microbiome and its metabolites influence antitumor responses by modulating innate and adaptive immunity, with specific bacteria potentially serving as biomarkers for predicting clinical responses. Various studies have identified microorganisms in breast tissue, previously considered sterile, revealing differences in breast microbial composition between patients with breast cancer and controls, as well as associations between specific breast microorganisms and clinicopathologic features, including immune correlations. The aim of this review was to provide a more comprehensive set of prognostic markers for TNBC and to tap into potential-specific therapeutic targets.
Collapse
Affiliation(s)
- Yating Shi
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Zhi Guo
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Huan Deng
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| |
Collapse
|
13
|
Peng Z, Zhao T, Gao P, Zhang G, Wu X, Tian H, Qu M, Tan X, Zhang Y, Zhao X, Qi X. Tumor-Derived Extracellular Vesicles Enable Tumor Tropism Chemo-Genetherapy for Local Immune Activation in Triple-Negative Breast Cancer. ACS NANO 2024; 18:30943-30956. [PMID: 39474658 PMCID: PMC11562804 DOI: 10.1021/acsnano.3c12967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 11/13/2024]
Abstract
Triple-negative breast cancer (TNBC) is highly heterogeneous, lacks accessible therapeutic targets, and features an immunosuppressive tumor microenvironment (TME). Anthracycline-based chemotherapy remains the primary treatment method for TNBC, while the current popular immune checkpoint inhibitors persistently encounter therapeutic resistance. Therefore, there is an urgent need to explore combined therapeutic strategies to remodel the TME and improve the treatment response. Considering the highly specific homing ability of tumor cell-derived vesicles and the key role of the signal transduction and activation of the transcription factor 3 (STAT3) pathway in TNBC, we propose a synergistic therapeutic strategy that integrates gene therapy, chemotherapy, and immunotherapy based on STAT3 short interfering RNA (siSTAT3) and doxorubicin (DOX)-functionalized tumor-derived extracellular vesicles (TEVs) (siSTAT3-DOX@TEV). The in vitro and in vivo results demonstrate that siSTAT3-DOX@TEV target tumor tissues precisely, downregulate STAT3 expression, and synergistically and efficiently induce immunogenic death, thereby reversing the immunosuppressive TME. Moreover, mass cytometry and immunohistochemistry reveal the local immune activation effect of siSTAT3-DOX@TEV, with a significant increase in M1 macrophages, CD4+ T cells, and CD8+ T cells in tumor tissues. These results provide strong hints for the development of TEV-based chemo-gene therapeutic agents for TNBC treatment at the clinical level.
Collapse
Affiliation(s)
- Zaihui Peng
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tingting Zhao
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Pingping Gao
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Guozhi Zhang
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiujuan Wu
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hao Tian
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Man Qu
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xuanni Tan
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yi Zhang
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiang Zhao
- Department
of Oncology, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaowei Qi
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
14
|
Arulraj T, Wang H, Deshpande A, Varadhan R, Emens LA, Jaffee EM, Fertig EJ, Santa-Maria CA, Popel AS. Virtual patient analysis identifies strategies to improve the performance of predictive biomarkers for PD-1 blockade. Proc Natl Acad Sci U S A 2024; 121:e2410911121. [PMID: 39467131 PMCID: PMC11551325 DOI: 10.1073/pnas.2410911121] [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: 05/31/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
Patients with metastatic triple-negative breast cancer (TNBC) show variable responses to PD-1 inhibition. Efficient patient selection by predictive biomarkers would be desirable but is hindered by the limited performance of existing biomarkers. Here, we leveraged in silico patient cohorts generated using a quantitative systems pharmacology model of metastatic TNBC, informed by transcriptomic and clinical data, to explore potential ways to improve patient selection. We evaluated and quantified the performance of 90 biomarker candidates, including various cellular and molecular species, at different cutoffs by a cutoff-based biomarker testing algorithm combined with machine learning-based feature selection. Combinations of pretreatment biomarkers improved the specificity compared to single biomarkers at the cost of reduced sensitivity. On the other hand, early on-treatment biomarkers, such as the relative change in tumor diameter from baseline measured at two weeks after treatment initiation, achieved remarkably higher sensitivity and specificity. Further, blood-based biomarkers had a comparable ability to tumor- or lymph node-based biomarkers in identifying a subset of responders, potentially suggesting a less invasive way for patient selection.
Collapse
Affiliation(s)
- Theinmozhi Arulraj
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Atul Deshpande
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Ravi Varadhan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | | | - Elizabeth M. Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Elana J. Fertig
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Applied Mathematics and Statistics, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Cesar A. Santa-Maria
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
15
|
Nandi D, Sharma D. Integrating immunotherapy with conventional treatment regime for breast cancer patients- an amalgamation of armamentarium. Front Immunol 2024; 15:1477980. [PMID: 39555066 PMCID: PMC11563812 DOI: 10.3389/fimmu.2024.1477980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/08/2024] [Indexed: 11/19/2024] Open
Abstract
Immunotherapy stands as the frontrunner in treatment strategies imparting efficient remission in various types of cancer. In fact, emerging breakthroughs with immune checkpoint inhibitors (ICI) in a spectrum of cancers have evoked interest in research related to the potential effects of immunotherapy in breast cancer patients. A major challenge with breast cancer is the molecular heterogeneity that limits the efficacy of many therapeutic regimes. Clinical trials have shown favorable clinical outcomes with immunotherapeutic options in some subtypes of breast cancer. However, ICI monotherapy may not be sufficient for all breast cancer patients, emphasizing the need for combinatorial approaches. Ongoing research is focused on untangling the interplay of ICI with established as well as novel anticancer therapeutic regimens in preclinical models of breast cancer. Our review will analyze the existing research regarding the mechanisms and clinical impact of immunotherapy for the treatment of breast cancer. We shall evaluate the role of immune cell modulation for improved therapeutic response in breast cancer patients. This review will provide collated evidences about the current clinical trials that are testing out the implications of immunotherapy in conjunction with traditional treatment modalities in breast cancer and summarize the potential future research directions in the field. In addition, we shall underline the recent findings related to microbiota modulation as a key regulator of immune therapy response in cancer patients and its plausible applications in breast cancer.
Collapse
Affiliation(s)
- Deeptashree Nandi
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| |
Collapse
|
16
|
Naji O, Ghouzlani A, Rafii S, Sadiqi RU, Kone AS, Harmak Z, Choukri K, Kandoussi S, Karkouri M, Badou A. Investigating tumor immunogenicity in breast cancer: deciphering the tumor immune response to enhance therapeutic approaches. Front Immunol 2024; 15:1399754. [PMID: 39507526 PMCID: PMC11538072 DOI: 10.3389/fimmu.2024.1399754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/18/2024] [Indexed: 11/08/2024] Open
Abstract
The interplay between immune cells and malignant cells represents an essential chapter in the eradication of breast cancer. This widely distributed and diverse form of cancer represents a major threat to women worldwide. The incidence of breast cancer is related to several risk factors, notably genetic predisposition and family antecedents. Despite progress in treatment modalities varying from surgery and chemotherapy to radiotherapy and targeted therapies, persistently high rates of recurrence, metastasis, and treatment resistance underscore the urgent need for new therapeutic approaches. Immunotherapy has gained considerable ground in the treatment of breast cancer, as it takes advantage of the complex interactions within the tumor microenvironment. This dynamic interplay between immune and tumor cells has become a key point of focus in immunological research. This study investigates the role of various cancer markers, such as neoantigens and immune regulatory genes, in the diagnosis and treatment of breast tumors. Moreover, it explores the future potential of immune checkpoint inhibitors as therapeutically effective agents, as well as the challenges that prevent their efficacy, in particular tumor-induced immunosuppression and the difficulty of achieving tumor specificity.
Collapse
Affiliation(s)
- Oumayma Naji
- Immuno-Genetics and Human Pathologies Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Amina Ghouzlani
- Immuno-Genetics and Human Pathologies Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Soumaya Rafii
- Immuno-Genetics and Human Pathologies Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Rizwan ullah Sadiqi
- Faculty of Science and Technology, Middlesex University, London, United Kingdom
| | - Abdou-samad Kone
- Immuno-Genetics and Human Pathologies Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Zakia Harmak
- Immuno-Genetics and Human Pathologies Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Khalil Choukri
- Immuno-Genetics and Human Pathologies Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Sarah Kandoussi
- Immuno-Genetics and Human Pathologies Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Mehdi Karkouri
- Department of Pathological Anatomy, University Hospital Center (CHU) Ibn Rochd and Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Abdallah Badou
- Immuno-Genetics and Human Pathologies Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Mohammed VI Center for Research and Innovation, Rabat and Mohammed VI University for Sciences and Health, Casablanca, Morocco
| |
Collapse
|
17
|
Ascic E, Åkerström F, Nair MS, Rosa A, Kurochkin I, Zimmermannova O, Catena X, Rotankova N, Veser C, Rudnik M, Ballocci T, Schärer T, Huang X, de Rosa Torres M, Renaud E, Santiago MV, Met Ö, Askmyr D, Lindstedt M, Greiff L, Ligeon LA, Agarkova I, Svane IM, Pires CF, Rosa FF, Pereira CF. In vivo dendritic cell reprogramming for cancer immunotherapy. Science 2024; 386:eadn9083. [PMID: 39236156 PMCID: PMC7616765 DOI: 10.1126/science.adn9083] [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: 02/29/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
Immunotherapy can lead to long-term survival for some cancer patients, yet generalized success has been hampered by insufficient antigen presentation and exclusion of immunogenic cells from the tumor microenvironment. Here, we developed an approach to reprogram tumor cells in vivo by adenoviral delivery of the transcription factors PU.1, IRF8, and BATF3, which enabled them to present antigens as type 1 conventional dendritic cells. Reprogrammed tumor cells remodeled their tumor microenvironment, recruited, and expanded polyclonal cytotoxic T cells; induced tumor regressions; and established long-term systemic immunity in multiple mouse melanoma models. In human tumor spheroids and xenografts, reprogramming to immunogenic dendritic-like cells progressed independently of immunosuppression, which usually limits immunotherapy. Our study paves the way for human clinical trials of in vivo immune cell reprogramming for cancer immunotherapy.
Collapse
Affiliation(s)
- Ervin Ascic
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | | | - Malavika Sreekumar Nair
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | - André Rosa
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
| | - Ilia Kurochkin
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | - Olga Zimmermannova
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | - Xavier Catena
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
| | | | | | | | - Tommaso Ballocci
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | | | - Xiaoli Huang
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
| | - Maria de Rosa Torres
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | - Emilie Renaud
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
| | - Marta Velasco Santiago
- National Center of Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730Herlev, Denmark
| | - Özcan Met
- National Center of Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730Herlev, Denmark
- Department of Health Technology, Technical University of Denmark, 2800Kongens Lyngby, Denmark
| | - David Askmyr
- Department of ORL, Head & Neck Surgery, Skåne University Hospital, 221 85Lund, Sweden
- Department of Clinical Sciences, Lund University, 221 84Lund, Sweden
| | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Medicon Village, 223 81Lund, Sweden
| | - Lennart Greiff
- Department of ORL, Head & Neck Surgery, Skåne University Hospital, 221 85Lund, Sweden
- Department of Clinical Sciences, Lund University, 221 84Lund, Sweden
| | | | | | - Inge Marie Svane
- National Center of Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730Herlev, Denmark
| | | | - Fábio F. Rosa
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
| | - Carlos-Filipe Pereira
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
- Centre for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês do Pombal, 3004-517Coimbra, Portugal
| |
Collapse
|
18
|
Li G, Ma X, Sui S, Chen Y, Li H, Liu L, Zhang X, Zhang L, Hao Y, Yang Z, Yang S, He X, Wang Q, Tao W, Xu S. NAT10/ac4C/JunB facilitates TNBC malignant progression and immunosuppression by driving glycolysis addiction. J Exp Clin Cancer Res 2024; 43:278. [PMID: 39363363 PMCID: PMC11451012 DOI: 10.1186/s13046-024-03200-x] [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: 06/22/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND N4-Acetylcytidine (ac4C), a highly conserved post-transcriptional mechanism, plays a pivotal role in RNA modification and tumor progression. However, the molecular mechanism by which ac4C modification mediates tumor immunosuppression remains elusive in triple-negative breast cancer (TNBC). METHODS NAT10 expression was analyzed in TNBC samples in the level of mRNA and protein, and compared with the corresponding normal tissues. ac4C modification levels also measured in the TNBC samples. The effects of NAT10 on immune microenvironment and tumor metabolism were investigated. NAT10-mediated ac4C and its downstream regulatory mechanisms were determined in vitro and in vivo. The combination therapy of targeting NAT10 in TNBC was further explored. RESULTS The results revealed that the loss of NAT10 inhibited TNBC development and promoted T cell activation. Mechanistically, NAT10 upregulated JunB expression by increasing ac4C modification levels on its mRNA. Moreover, JunB further up-regulated LDHA expression and facilitated glycolysis. By deeply digging, remodelin, a NAT10 inhibitor, elevated the surface expression of CTLA-4 on T cells. The combination of remodelin and CTLA-4 mAb can further activate T cells and inhibite tumor progression. CONCLUSION Taken together, our study demonstrated that the NAT10-ac4C-JunB-LDHA pathway increases glycolysis levels and creates an immunosuppressive tumor microenvironment (TME). Consequently, targeting this pathway may assist in the identification of novel therapeutic strategies to improve the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Guozheng Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Xin Ma
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Shiyao Sui
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Yihai Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
- National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Hui Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Lei Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Xin Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Lei Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Yi Hao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
- National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Zihan Yang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Shuai Yang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Xu He
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
- Weihan Yu Academy, Harbin Medical University, Harbin, 150086, China
| | - Qin Wang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China.
- Key Laboratory of Tumor Biotherapy of Heilongjiang Province, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Weiyang Tao
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Shouping Xu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China.
- Key Laboratory of Tumor Biotherapy of Heilongjiang Province, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
19
|
Sabu N, Attia Hussein Mahmoud H, Salazar González JF, Naruboina N, Esteban Rojas Prieto S, Govender S, Ruthvik Phani Narayan V, Priyank Batukbhai B, Ahmadi Y. Role of Immunotherapy in Conjunction With the Surgical Treatment of Breast Cancer: Preoperative and Postoperative Applications. Cureus 2024; 16:e71441. [PMID: 39539894 PMCID: PMC11559439 DOI: 10.7759/cureus.71441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Breast cancer is one of the most common cancers in the world. Since the appearance of molecular medicine, the perspective of breast cancer treatment has changed, making it more successful in comparison with the treatment during previous years. Numerous ongoing trials are exploring the capacity of immunotherapy, mainly in immune checkpoint inhibitors (ICIs), in conjunction with conventional therapies or with antibody-drug conjugates (ADCs). The current narrative review discusses the advantages and limitations of immunotherapy in breast cancer treatment in conjunction with the surgical options available. Going through the modern capacity of surgery treatment and how the use of immunotherapy in conjunction with it has emerged as a transformative approach to breast cancer and listing the main complications and adverse effects caused by ICIs. We searched Google Scholar, PubMed, MEDLINE, and EMBASS. Fourteen different articles showed that the use of cytokines and cancer vaccines revealed new possibilities to treat breast cancer with antibodies against PD-1/PD-L1 (pembrolizumab), PI3K/Akt/mTOR (alpelisib and everolimus), CAR T-cell (chimeric antigen receptor), PARP (poly ADP-ribose polymerase), and CTLA4 (cytotoxic T-lymphocyte-associated protein 4), and with representative relevance of changing in tumor microenvironment. Immunotherapy made it possible to reduce recurrences, after radiotherapy and surgery. Estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2) targets show also a high effectivity. In recent years, the release of new strategies has become promising, for changing the microenvironment and de-escalation of therapy based on tumor biology, novel biomarkers, and tumor spread.
Collapse
Affiliation(s)
- Nagma Sabu
- Department of Surgery, University of Perpetual Help System Dalta - JONELTA Foundation School of Medicine, Las Pinas, PHL
| | | | | | | | | | - Seyanne Govender
- General Practice, American University of the Caribbean, Cupecoy, SXM
| | | | | | - Yasmin Ahmadi
- School of Medicine, Royal College of Surgeons in Ireland - Medical University of Bahrain, Muharraq, BHR
| |
Collapse
|
20
|
Wang R, Kumar P, Reda M, Wallstrum AG, Crumrine NA, Ngamcherdtrakul W, Yantasee W. Nanotechnology Applications in Breast Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308639. [PMID: 38126905 PMCID: PMC11493329 DOI: 10.1002/smll.202308639] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Next-generation cancer treatments are expected not only to target cancer cells but also to simultaneously train immune cells to combat cancer while modulating the immune-suppressive environment of tumors and hosts to ensure a robust and lasting response. Achieving this requires carriers that can codeliver multiple therapeutics to the right cancer and/or immune cells while ensuring patient safety. Nanotechnology holds great potential for addressing these challenges. This article highlights the recent advances in nanoimmunotherapeutic development, with a focus on breast cancer. While immune checkpoint inhibitors (ICIs) have achieved remarkable success and lead to cures in some cancers, their response rate in breast cancer is low. The poor response rate in solid tumors is often associated with the low infiltration of anti-cancer T cells and an immunosuppressive tumor microenvironment (TME). To enhance anti-cancer T-cell responses, nanoparticles are employed to deliver ICIs, bispecific antibodies, cytokines, and agents that induce immunogenic cancer cell death (ICD). Additionally, nanoparticles are used to manipulate various components of the TME, such as immunosuppressive myeloid cells, macrophages, dendritic cells, and fibroblasts to improve T-cell activities. Finally, this article discusses the outlook, challenges, and future directions of nanoimmunotherapeutics.
Collapse
Affiliation(s)
- Ruijie Wang
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
| | - Pramod Kumar
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
| | - Moataz Reda
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| | | | - Noah A. Crumrine
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| | | | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| |
Collapse
|
21
|
Quintana A, Saini KS, Vidal L, Peg V, Slebe F, Loibl S, Curigliano G, Schmid P, Cortes J. Window of opportunity trials with immune checkpoint inhibitors in triple-negative breast cancer. ESMO Open 2024; 9:103713. [PMID: 39357122 PMCID: PMC11480225 DOI: 10.1016/j.esmoop.2024.103713] [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: 04/11/2024] [Revised: 07/08/2024] [Accepted: 08/18/2024] [Indexed: 10/04/2024] Open
Abstract
Patients with triple-negative breast cancer (TNBC) have a relatively poor clinical outcome. The immune checkpoint inhibitor (ICI) pembrolizumab combined with chemotherapy is the current standard of care in TNBC patients with stage II and III. Monotherapy with ICIs has not been comprehensively assessed in the neoadjuvant setting in TNBC patients, given unfavorable results in metastatic trials. ICIs, however, have been tested in the window of opportunity (WOO) before surgery or standard chemotherapy-based neoadjuvant treatment. The WOO design is well suited to assess an ICI alone or in combination with other ICIs, targeted therapy, radiotherapy or cryotherapy, and measure their pharmacodynamic and clinical effect in this treatment-naive population. Some patients show a good response to ICIs in WOO studies. Biomarkers like tumor-infiltrating lymphocytes, programmed death ligand-1, and interferon-γ signature may predict activity and may identify patients likely to benefit from ICIs. Moreover, an increase in tumor-infiltrating lymphocytes, programmed death ligand-1 expression or T cell receptor expansion following administration of ICIs in the WOO setting could potentially inform of immunotherapy benefit, which would allow tailoring further treatment. This article reviews WOO trials that assessed immunotherapy in the early-stage TNBC population, and how these results could be translated to test de-escalation strategies of neoadjuvant chemotherapy and immunotherapy without compromising a patient's prognosis.
Collapse
Affiliation(s)
- A Quintana
- Breast Cancer Unit, Vall d'Hebrón Institute of Oncology, Barcelona, Spain.
| | - K S Saini
- Fortrea, Inc., Durham, USA; Addenbrooke's Hospital, Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, UK
| | | | - V Peg
- Biomedical Research Network Centre in Oncology (CIBERONC), Madrid; Department of Pathology, Vall d'Hebron University Hospital, Barcelona; Departament of Medicine, Universitat Autonoma de Barcelona, Barcelona
| | - F Slebe
- Medica Scientia Innovation Research (MedSIR), Barcelona, Spain; Oncoclínicas & Co, Jersey City, New Jersey and Sao Paulo, Brazil
| | - S Loibl
- German Breast Group, GBG Forschungs GmbH, Neu-Isenburg, Germany
| | - G Curigliano
- European Institute of Oncology, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - P Schmid
- Barts Cancer Institute, Queen Mary University London, London, UK
| | - J Cortes
- Medica Scientia Innovation Research (MedSIR), Barcelona, Spain; Oncoclínicas & Co, Jersey City, New Jersey and Sao Paulo, Brazil; Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid; International Breast Cancer Center, Pangaea Oncology, Quironsalud Group, Barcelona; IOB Madrid, Institute of Oncology, Hospital Beata Maria Ana, Madrid, Spain
| |
Collapse
|
22
|
Amir M, Khan ZA, Asad A, Shaikh TG. Role of neoadjuvant pembrolizumab in advanced melanoma. Semin Oncol 2024; 51:161-162. [PMID: 39551678 DOI: 10.1053/j.seminoncol.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 11/19/2024]
Abstract
Melanoma, a malignancy originating from melanocytes, poses a significant global health challenge, with approximately 325,000 cases and 57,000 deaths annually. Advanced melanoma (AM), categorized as stage III and IV, presents considerable treatment challenges due to its complex mutational landscape. Traditional treatment options have included checkpoint inhibitors and BRAF inhibitors, with pembrolizumab emerging as a promising agent. Approved by the FDA and EMA for various stages of melanoma, pembrolizumab is a humanized monoclonal antibody that blocks the PD-1/PD-L1 interaction, thereby enhancing immune system-mediated tumor eradication. This abstract discusses a recent phase 2 clinical trial evaluating the efficacy of neoadjuvant (presurgery) versus adjuvant (postsurgery) pembrolizumab treatment in resectable stage III or IV melanoma. The study, involving 313 patients across 90 US hospitals, found that neoadjuvant-adjuvant pembrolizumab significantly improved event-free survival (72%) compared to adjuvant-only treatment (49%) after 2 years. Treatment-related adverse events were consistent with known profiles, including fatigue, nausea, and diarrhea, without new severe adverse effects. No increase in surgery-related complications was observed with neoadjuvant treatment. These findings suggest that neoadjuvant pembrolizumab offers substantial benefits over adjuvant-only treatment, although further research is warranted. Future studies should focus on larger cohorts, diverse demographics, and extended follow-up to validate these results and potentially integrate neoadjuvant pembrolizumab into standard treatment protocols for advanced melanoma.
Collapse
Affiliation(s)
- Maheen Amir
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Zoha Ali Khan
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan.
| | - Ayza Asad
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Taha Gul Shaikh
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
23
|
Sriramulu S, Thoidingjam S, Speers C, Nyati S. Present and Future of Immunotherapy for Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:3250. [PMID: 39409871 PMCID: PMC11475478 DOI: 10.3390/cancers16193250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Triple-negative breast cancer (TNBC) lacks the expression of estrogen receptors (ERs), human epidermal growth factor receptor 2 (HER2), and progesterone receptors (PRs). TNBC has the poorest prognosis among breast cancer subtypes and is more likely to respond to immunotherapy due to its higher expression of PD-L1 and a greater percentage of tumor-infiltrating lymphocytes. Immunotherapy has revolutionized TNBC treatment, especially with the FDA's approval of pembrolizumab (Keytruda) combined with chemotherapy for advanced cases, opening new avenues for treating this deadly disease. Although immunotherapy can significantly improve patient outcomes in a subset of patients, achieving the desired response rate for all remains an unmet clinical goal. Strategies that enhance responses to immune checkpoint blockade, including combining immunotherapy with chemotherapy, molecularly targeted therapy, or radiotherapy, may improve response rates and clinical outcomes. In this review, we provide a short background on TNBC and immunotherapy and explore the different types of immunotherapy strategies that are currently being evaluated in TNBC. Additionally, we review why combination strategies may be beneficial, provide an overview of the combination strategies, and discuss the novel immunotherapeutic opportunities that may be approved in the near future for TNBC.
Collapse
Affiliation(s)
- Sushmitha Sriramulu
- Department of Radiation Oncology, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Shivani Thoidingjam
- Department of Radiation Oncology, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Corey Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Radiation Oncology, UH Seidman Cancer Center, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Shyam Nyati
- Department of Radiation Oncology, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
24
|
Im SA, Cortes J, Cescon DW, Yusof MM, Iwata H, Masuda N, Takano T, Huang CS, Chung CF, Tsugawa K, Park YH, Matsumoto K, Inoue K, Kwong A, Loi S, Fu W, Pan W, Karantza V, Rugo HS, Schmid P. Results from the randomized KEYNOTE-355 study of pembrolizumab plus chemotherapy for Asian patients with advanced TNBC. NPJ Breast Cancer 2024; 10:79. [PMID: 39266535 PMCID: PMC11393332 DOI: 10.1038/s41523-024-00679-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/19/2024] [Indexed: 09/14/2024] Open
Abstract
In the phase 3 KEYNOTE-355 study (NCT02819518), pembrolizumab plus chemotherapy demonstrated statistically significant and clinically meaningful improvements in progression-free survival (PFS) and overall survival (OS) versus placebo plus chemotherapy among patients with previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (TNBC) and programmed cell death ligand 1 (PD-L1) combined positive score (CPS) ≥ 10 tumors. We analyzed outcomes for the subgroup of patients enrolled in Asia in KEYNOTE-355. Patients received pembrolizumab 200 mg or placebo (2:1 randomization) every 3 weeks for 35 cycles plus investigator's choice chemotherapy. Primary endpoints were PFS per Response Evaluation Criteria in Solid Tumors version 1.1 and OS. Among patients enrolled in Hong Kong, Japan, Korea, Malaysia and Taiwan (pembrolizumab plus chemotherapy, n = 113; placebo plus chemotherapy, n = 47), 117 (73.1%) had PD-L1 CPS ≥ 1 and 56 (35.0%) had PD-L1 CPS ≥ 10. Median time from randomization to data cutoff (June 15, 2021) was 43.8 (range, 36.8‒53.2) months (intent-to-treat [ITT] population). Hazard ratios (HRs [95% CI]) for PFS in the CPS ≥ 10, CPS ≥ 1, and ITT populations were 0.48 (0.24‒0.98), 0.58 (0.37‒0.91), and 0.66 (0.44‒0.99), respectively. Corresponding HRs (95% CI) for OS were 0.54 (0.28‒1.04), 0.62 (0.40‒0.97), and 0.57 (0.39‒0.84). Grade 3/4 treatment-related adverse events (AEs) occurred in 77.9% versus 78.7% of patients with pembrolizumab plus chemotherapy versus placebo plus chemotherapy. No grade 5 AEs occurred. Clinically meaningful improvement in PFS and OS with manageable toxicity were observed with pembrolizumab plus chemotherapy versus placebo plus chemotherapy in patients enrolled in Asia with previously untreated, inoperable or metastatic TNBC.Trial registration: ClinicalTrials.gov, NCT02819518.
Collapse
Affiliation(s)
- Seock-Ah Im
- Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, Republic of Korea.
| | - Javier Cortes
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Madrid and Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | - David W Cescon
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Mastura Md Yusof
- Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Hiroji Iwata
- Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Norikazu Masuda
- Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toshimi Takano
- The Cancer Institute Hospital of JFCR, Tokyo, Japan
- Toranomon Hospital, Tokyo, Japan
| | - Chiun-Sheng Huang
- National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Feng Chung
- Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | | | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | - Ava Kwong
- Division of Breast Surgery, The University of Hong Kong, Queen Mary and Tung Wah Hospital, Hong Kong, China
- The University of Hong Kong-ShenZhen Hospital, Shenzhen, China
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, Australia
| | - Wei Fu
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, CA, USA
| | - Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, London, UK
| |
Collapse
|
25
|
Cao Z, Wichmann CW, Burvenich IJG, Osellame LD, Guo N, Rigopoulos A, O'Keefe GJ, Scott FE, Lorensuhewa N, Lynch KP, Scott AM. Radiolabelling and preclinical characterisation of [ 89Zr]Zr-Df-ATG-101 bispecific to PD-L1/4-1BB. Eur J Nucl Med Mol Imaging 2024; 51:3202-3214. [PMID: 38730087 PMCID: PMC11368977 DOI: 10.1007/s00259-024-06742-6] [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: 11/14/2023] [Accepted: 04/26/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE ATG-101, a bispecific antibody that simultaneously targets the immune checkpoint PD-L1 and the costimulatory receptor 4-1BB, activates exhausted T cells upon PD-L1 crosslinking. Previous studies demonstrated promising anti-tumour efficacy of ATG-101 in preclinical models. Here, we labelled ATG-101 with 89Zr to confirm its tumour targeting effect and tissue biodistribution in a preclinical model. We also evaluated the use of immuno-PET to study tumour uptake of ATG-101 in vivo. METHODS ATG-101, anti-PD-L1, and an isotype control were conjugated with p-SCN-Deferoxamine (Df). The Df-conjugated antibodies were radiolabelled with 89Zr, and their radiochemical purity, immunoreactivity, and serum stability were assessed. We conducted PET/MRI and biodistribution studies on [89Zr]Zr-Df-ATG-101 in BALB/c nude mice bearing PD-L1-expressing MDA-MB-231 breast cancer xenografts for up to 10 days after intravenous administration of [89Zr]Zr-labelled antibodies. The specificity of [89Zr]Zr-Df-ATG-101 was evaluated through a competition study with unlabelled ATG-101 and anti-PD-L1 antibodies. RESULTS The Df-conjugation and [89Zr]Zr -radiolabelling did not affect the target binding of ATG-101. Biodistribution and imaging studies demonstrated biological similarity of [89Zr]Zr-Df-ATG-101 and [89Zr]Zr-Df-anti-PD-L1. Tumour uptake of [89Zr]Zr-Df-ATG-101 was clearly visualised using small-animal PET imaging up to 7 days post-injection. Competition studies confirmed the specificity of PD-L1 targeting in vivo. CONCLUSION [89Zr]Zr-Df-ATG-101 in vivo distribution is dependent on PD-L1 expression in the MDA-MB-231 xenograft model. Immuno-PET with [89Zr]Zr-Df-ATG-101 provides real-time information about ATG-101 distribution and tumour uptake in vivo. Our data support the use of [89Zr]Zr-Df-ATG-101 to assess tumour and tissue uptake of ATG-101.
Collapse
Affiliation(s)
- Zhipeng Cao
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
| | - Christian Werner Wichmann
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
- School of Chemistry - Bio21 Institute, University of Melbourne, Melbourne, Australia
| | - Ingrid Julienne Georgette Burvenich
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Laura Danielle Osellame
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Nancy Guo
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
| | - Angela Rigopoulos
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
| | - Graeme Joseph O'Keefe
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Fiona Elizabeth Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | | | | | - Andrew Mark Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia.
- School of Cancer Medicine, La Trobe University, Melbourne, Australia.
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia.
- Department of Medicine, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
26
|
Pedroza DA, Gao Y, Zhang XHF, Rosen JM. Leveraging preclinical models of metastatic breast cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189163. [PMID: 39084494 PMCID: PMC11390310 DOI: 10.1016/j.bbcan.2024.189163] [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: 11/17/2023] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Women that present to the clinic with established breast cancer metastases have limited treatment options. Yet, the majority of preclinical studies are actually not directed at developing treatment regimens for established metastatic disease. In this review we will discuss the current state of preclinical macro-metastatic breast cancer models, including, but not limited to syngeneic GEMM, PDX and xenografts. Challenges within these models which are often overlooked include fluorophore-immunogenic neoantigens, differences in experimental vs spontaneous metastasis and tumor heterogeneity. Furthermore, due to cell plasticity in the tumor immune microenvironment (TIME) of the metastatic landscape, the treatment efficacy of newly approved immune checkpoint blockade (ICB) may differ in metastatic sites as compared to primary localized tumors.
Collapse
Affiliation(s)
- Diego A Pedroza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Yang Gao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Xiang H-F Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America.
| |
Collapse
|
27
|
Ribeiro R, Carvalho FM, Baiocchi G, Guindalini RSC, da Cunha JR, Anjos CHD, de Nadai Costa C, Gifoni ACLVC, Neto RC, Cagnacci AQC, Carneiro VCG, Calabrich A, Moretti-Marques R, Pinheiro RN, de Castro Ribeiro HS. Guidelines of the Brazilian Society of Surgical Oncology for anatomopathological, immunohistochemical, and molecular testing in female tumors. J Surg Oncol 2024; 130:882-895. [PMID: 39038206 DOI: 10.1002/jso.27717] [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: 05/01/2024] [Accepted: 05/04/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Precision medicine has revolutionized oncology, providing more personalized diagnosis, treatment, and monitoring for patients with cancer. In the context of female-specific tumors, such as breast, ovarian, endometrial, and cervical cancer, proper tissue collection and handling are essential for obtaining tissue, immunohistochemical (IHC), and molecular data to guide therapeutic decisions. OBJECTIVES To establish guidelines for the collection and handling of tumor tissue, to enhance the quality of samples for histopathological, IHC, genomic, and molecular analyses. These guidelines are fundamental in informing therapeutic decisions in cancer treatment. METHOD The guidelines were developed by a multidisciplinary panel of renowned specialists between June 12, 2013 and February 12, 2024. Initially, the panel deliberated on critical and controversial topics related to conducting precision medicine studies focusing on female tumors. Subsequently, 22 pivotal topics were identified within the framework and assigned to groups. These groups reviewed relevant literature and drafted preliminary recommendations. Following this, the recommendations were reviewed by the coordinators and received unanimous approval. Finally, the groups made the final adjustments, classified the level of evidence, and ranked the recommendations. CONCLUSION The collection of surgical samples requires minimum quality standards to enable histopathological, IHC, genomic, and molecular analyses. These analyses provide crucial data for informing therapeutic decisions, significantly impacting potential survival gains for patients with female tumors.
Collapse
Affiliation(s)
- Reitan Ribeiro
- Department of Gynecology Oncology, Erasto Gaertner Hospital, Curitiba, Paraná, Brazil
| | - Filomena Marino Carvalho
- Department of Pathology, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Glauco Baiocchi
- Department of Gynecologic Oncology, AC Camargo Cancer Center , São Paulo, São Paulo, Brazil
| | | | | | | | | | | | - Renato Cagnacci Neto
- Department of Mastology, Breast Cancer Reference Center, AC Camargo Cancer, CenterSão Paulo, São Paulo, Brazil
| | - Allyne Queiroz Carneiro Cagnacci
- Department of Oncology, Oncology Center, Hospital Alemão Oswaldo Cruz, São Paulo, São Paulo, Brazil
- Hereditary Cancer Department, Instituto do Câncer do Estado de São Paulo (ICESPSP), São Paulo, São Paulo, Brazil
| | - Vandré Cabral Gomes Carneiro
- Department of Gynecology Oncology, Instituto de Medicina Integral Professor Fernando Figueira (IMIP), Recife, Pernambuco, Brazil
- Research Department, Hospital de Câncer de Pernambuco, Recife, Brazil
- Department of Oncogenetic, Oncologia D'OR, Recife, Pernambuco, Brazil
| | - Aknar Calabrich
- Department of Oncology, Clínica AMO/DASA, Salvador, Bahia, Brazil
| | - Renato Moretti-Marques
- Department of Oncology, Albert Einstein Israelite Hospital, São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
28
|
Bischoff H, Espié M, Petit T. Neoadjuvant Therapy: Current Landscape and Future Horizons for ER-Positive/HER2-Negative and Triple-Negative Early Breast Cancer. Curr Treat Options Oncol 2024; 25:1210-1224. [PMID: 39145854 PMCID: PMC11416407 DOI: 10.1007/s11864-024-01251-y] [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] [Accepted: 07/25/2024] [Indexed: 08/16/2024]
Abstract
OPINION STATEMENT Navigating the complex landscape of breast cancer treatment involves distinct strategies for luminal and triple-negative subtypes. While neoadjuvant chemotherapy historically dominates the approach for aggressive triple-negative tumors, recent evidence highlights the transformative impact of immunotherapy, alongside chemotherapy, in reshaping treatment paradigms. In luminal cancers, endocrine therapy, notably aromatase inhibitors, demonstrates promising outcomes in postmenopausal patients with low-grade luminal A tumors. However, integrating targeted therapies like CDK4/6 inhibitors in neoadjuvant setting remains inconclusive. Identifying predictive factors for treatment response, especially in luminal tumors, poses a challenge, emphasizing the necessity for ongoing research. A multidisciplinary approach, tailored to individual patient profiles, is crucial for maximizing efficacy while minimizing toxicity. As we strive to optimize breast cancer management, a comprehensive understanding of the distinct characteristics and treatment implications of luminal and triple-negative subtypes, including the transformative role of immunotherapy, is essential for informed decision-making and personalized care.
Collapse
Affiliation(s)
- Hervé Bischoff
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, 67033, Strasbourg, France.
| | - Marc Espié
- Medical Oncology Department, Hôpital Saint Louis, Paris, France
| | - Thierry Petit
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, 67033, Strasbourg, France
| |
Collapse
|
29
|
Zhang XC, Zhou YW, Wei GX, Luo YQ, Qiu M. Locoregional therapies combined with immune checkpoint inhibitors for liver metastases. Cancer Cell Int 2024; 24:302. [PMID: 39217341 PMCID: PMC11365172 DOI: 10.1186/s12935-024-03484-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have achieved remarkable success in clinical research and practice. Notably, liver metastasis is not sensitive to ICIs. Liver locoregional therapies can cause irreversible damage to tumor cells and release tumor antigens, thereby providing a rationale for immunotherapy treatments in liver metastasis. The combination therapy of ICIs with locoregional therapies is a promising option for patients with liver metastasis. Preclinical studies have demonstrated that combining ICIs with locoregional therapies produces a significantly synergistic anti-tumor effect. However, the current evidence for the efficacy of ICIs combined with locoregional therapies remains insufficient. Therefore, we review the literature on the mechanisms of locoregional therapies in treating liver metastasis and the clinical research progress of their combination with ICIs.
Collapse
Affiliation(s)
- Xing-Chen Zhang
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Yu-Wen Zhou
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Gui-Xia Wei
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yi-Qiao Luo
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Meng Qiu
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
30
|
Hu Y, Wang C, Liang H, Li J, Yang Q. The treatment landscape of triple-negative breast cancer. Med Oncol 2024; 41:236. [PMID: 39210220 DOI: 10.1007/s12032-024-02456-9] [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] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
Triple-negative breast cancer (TNBC) tumors are biologically aggressive breast cancer. On the molecular level, TNBC is a highly heterogeneous disease; more biotechnologies are gradually being used to advance the understanding of TNBC subtypes and help establish more targeted therapies. Multiple TNBC target-related agents are already approved by the Food and Drug Administration for clinical use, including PI3K/AKT/mTOR inhibitors, PRAP inhibitors, and antibody-drug conjugates. Some innovative approaches, like peptide strategies, also promise to treat TNBC. Currently, the interplay between TNBC tumors and their tumor microenvironment provides a promising prospect for improving the efficacy of immunotherapy. In this review, we summarize the prevalent TNBC subtype methodologies, discuss the evolving therapeutic strategies, and propose new therapeutic possibilities based on existing foundational theories, with the attempt to serve as a reference to further advance tailoring treatment of TNBC.
Collapse
Affiliation(s)
- Yi Hu
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Chen Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Huishi Liang
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Jie Li
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China.
| | - Qiong Yang
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China.
| |
Collapse
|
31
|
Wen QE, Li L, Feng RQ, Li DH, Qiao C, Xu XS, Zhang YJ. Recent Advances in Immunotherapy for Breast Cancer: A Review. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:497-516. [PMID: 39220564 PMCID: PMC11365501 DOI: 10.2147/bctt.s482504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Breast cancer is one of the most common malignant tumors in women in the world, and its incidence is increasing year by year, which seriously threatens the physical and mental health of women. Triple negative breast cancer (TNBC) is a special molecular type of breast cancer in which estrogen receptor, progesterone receptor and human epidermal growth factor receptor-2 are negative. Compared with other molecular types of breast cancer, triple-negative breast cancer (TNBC) has high aggressiveness and metastasis, high recurrence rate, lack of effective therapeutic targets, and usually poor clinical treatment effect. Chemotherapy was the main therapeutic means used in the past. With the advent of the immune era, immunotherapy has made a lot of progress in the treatment of triple-negative breast cancer (TNBC), bringing new therapeutic hope for the treatment of triple-negative breast cancer. This review combines the results of cutting-edge medical research, mainly summarizes the research progress of immunotherapy, and summarizes the main treatment methods of triple-negative breast cancer (TNBC) immunotherapy, including immune checkpoint inhibitors, tumor vaccines, adoptive immunotherapy and the application of traditional Chinese and western medicine. It provides a new idea for the treatment of triple negative breast cancer (TNBC).
Collapse
Affiliation(s)
- Qian-Er Wen
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Liang Li
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Rui-Qi Feng
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, People’s Republic of China
| | - De-Hui Li
- Oncology Department II, The First Affiliated Hospital of Hebei University of Chinese Medicine (Hebei Province Hospital of Chinese Medicine), Key Laboratory of Integrated Chinese and Western Medicine for Gastroenterology Research, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Chang Qiao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Xiao-Song Xu
- Scientific research Center, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Yan-Jing Zhang
- Oncology Department II, The First Affiliated Hospital of Hebei University of Chinese Medicine (Hebei Province Hospital of Chinese Medicine), Key Laboratory of Integrated Chinese and Western Medicine for Gastroenterology Research, Shijiazhuang, Hebei Province, People’s Republic of China
| |
Collapse
|
32
|
Li Y, Yang X, Ma L. Comparative analysis of adverse event risks in breast cancer patients receiving pembrolizumab combined with paclitaxel versus paclitaxel monotherapy: insights from the FAERS database. Front Pharmacol 2024; 15:1345671. [PMID: 39234109 PMCID: PMC11372242 DOI: 10.3389/fphar.2024.1345671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
Objective This study aimed to evaluate the risk of adverse events (AEs) in breast cancer patients treated with pembrolizumab combined with paclitaxel versus those receiving pembrolizumab or paclitaxel monotherapy, using the FDA Adverse Event Reporting System (FAERS) database. Methods Data were extracted from the FAERS database for breast cancer patients treated with pembrolizumab combined with paclitaxel or with pembrolizumab or paclitaxel monotherapy from Q1 2016 to Q2 2023. Disproportionation analysis was performed by calculating the reporting odds ratio (ROR) with corresponding 95% confidence interval (95% CI), the information component (IC), and the lower bound of the information component 95% confidence interval (IC025) to identify potential safety signals. Results No significant difference in AEs was observed between the combined treatment group and the pembrolizumab monotherapy group. However, the combined treatment group exhibited a substantial increase in AE risk compared to the paclitaxel monotherapy group. The most significant increases in AE risk were adrenal insufficiency (ROR = 189.94, 95% CI 25.41-1419.7, IC = 3.37, IC025 = 1.59), hypophysitis (ROR = 99.46, 95% CI 12.72-777.4, IC = 3.31, IC025 = 1.44), and myocarditis (ROR = 69.5, 95% CI 8.55-565.23, IC = 3.25, IC025 = 1.33). The time-to-event for combined treatment was 35 (34-70) days, for pembrolizumab was 43 (35-90) days, and for paclitaxel was 42 (37-76) days. The combination therapy group demonstrated significantly shorter intervals to the onset of adrenal insufficiency (p = 0.008), myocarditis (p < 0.001), and immune-related enterocolitis (p = 0.009). Conclusion Analysis of the FAERS database indicates that combination therapy significantly elevates the risk of adrenal insufficiency, myocarditis, hypophysitis, and immune-related enterocolitis compared to paclitaxel monotherapy. These findings provide critical insights for clinicians in predicting and managing potential AEs associated with this treatment regimen.
Collapse
Affiliation(s)
- Yilun Li
- Department of Breast Disease Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, Shijiazhuang, China
| | - Xiaolu Yang
- Department of Breast Disease Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Medical University, Shijiazhuang, China
| | - Li Ma
- Department of Breast Disease Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, Shijiazhuang, China
| |
Collapse
|
33
|
Chen L, Dhoomun DK, Liu Q, Kong X, Li X, Peng S, Lan P, Wang J. A prognostic model based on CLEC6A predicts clinical outcome of breast cancer patients. Int Immunopharmacol 2024; 137:112411. [PMID: 38852520 DOI: 10.1016/j.intimp.2024.112411] [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: 11/05/2023] [Revised: 05/10/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
CLEC6A, (C-type lectin domain family 6, member A), plays a prominent role in regulating innate immunity and adaptive immunity. CLEC6A has shown great potential as a target for cancer immunotherapy. This study aims to explore the prognostic value of CLEC6A, and analyze the relationship associated with the common hematological parameters in breast cancer patients. We performed a retrospective analysis on 183 breast cancer patients data in hospital information system from January 2013 to December 2015. The expression of CLEC6A was recorded via semiquantitative immunohistochemistry in breast cancer. The association between expression of CLEC6A and relative parameters were performed by Chi-square test and Fisher's exact test. Kaplan-Meier assay and Log-rank test were performed to evaluate the survival time. The Cox proportional hazards regression analysis was applied to identify prognostic factors. Nomograms were conducted to predict 1-, 3-, and 5-year disease free survival (DFS) and overall survival (OS) for breast cancer, which could be a good reference in clinical practice. The nomogram model was estimated by calibration curve analysis for its function of discrimination. The accuracy and benefit of the nomogram model were appraised by comparing it to only CLEC6A via decision curve analysis (DCA). The prediction accuracy of CLEC6A was also determined by time-dependent receiver operating characteristics (TDROC) curves, and the area under the curve (AUC) for different survival time. There were 94 cases in the CLEC6A low-expression group and 89 cases in CLEC6A high-expression group. Compared to CLEC6A low-expression group, the CLEC6A high-expression group had better survival (DFS: 56.95 vs. 70.81 months, P = 0.0078 and OS: 67.98 vs. 79.05 months, P = 0.0089). The CLEC6A was a potential prognostic factor in multivariate analysis (DFS: P = 0.023, hazard ratio (HR): 0.454, 95 % confidence interval (CI): 0.229-0.898; OS: P = 0.020, HR: 0.504, 95 %CI: 0.284-0.897). The nomogram in accordance with these potential prognostic factors was constructed to predict survival and the calibration curve analysis had indicated that the predicted line was well-matched with reference line in 1-, 3-, and 5-year DFS and OS category. The 1-, 3-, and 5-year DCA curves have revealed that nomogram model yielded larger net benefits than CLEC6A alone. Finally, the TDROC curve indicated that CLEC6A could better predict 1-year DFS and OS than others. Furthermore, we combined these potential independent prognostic factors to analyze the relationship among these hematologic index and oxidative stress indicators, and indicated that higher CLEC6A level, higher CO2 level or low CHOL level or high HDL-CHO level would have survived longer and better prognosis. In breast cancer, high expression of CLEC6A can independently predict better survival. Our nomogram consisted of CLEC6A and other indicators has good predictive performance and can facilitate clinical decision-making.
Collapse
Affiliation(s)
- Li Chen
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China; Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Deenraj Kush Dhoomun
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Qiang Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| | - Shu Peng
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| | - Peixiang Lan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Hubei 430030, PR China.
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China.
| |
Collapse
|
34
|
Al-Ruwishan A, Amer B, Salem A, Abdi A, Chimpandu N, Esa A, Melemenis A, Saleem MZ, Mathew R, Gamallat Y. Advancements in Understanding the Hide-and-Seek Strategy of Hibernating Breast Cancer Cells and Their Implications in Oncology from a Broader Perspective: A Comprehensive Overview. Curr Issues Mol Biol 2024; 46:8340-8367. [PMID: 39194709 DOI: 10.3390/cimb46080492] [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: 06/10/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Despite recent advancements in technology, breast cancer still poses a significant threat, often resulting in fatal consequences. While early detection and treatments have shown some promise, many breast cancer patients continue to struggle with the persistent fear of the disease returning. This fear is valid, as breast cancer cells can lay dormant for years before remerging, evading traditional treatments like a game of hide and seek. The biology of these dormant breast cancer cells presents a crucial yet poorly understood challenge in clinical settings. In this review, we aim to explore the mysterious world of dormant breast cancer cells and their significant impact on patient outcomes and prognosis. We shed light on the elusive role of the G9a enzyme and many other epigenetic factors in breast cancer recurrence, highlighting its potential as a target for eliminating dormant cancer cells and preventing disease relapse. Through this comprehensive review, we not only emphasise the urgency of unravelling the dynamics of dormant breast cancer cells to improve patient outcomes and advance personalised oncology but also provide a guide for fellow researchers. By clearly outlining the clinical and research gaps surrounding dormant breast cancer cells from a molecular perspective, we aim to inspire further exploration of this critical area, ultimately leading to improved patient care and treatment strategies.
Collapse
Affiliation(s)
- Aiman Al-Ruwishan
- Space for Research Initiative, Research Horizons, London NW10 2PU, UK
| | - Bushra Amer
- Department of Family Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Ahmed Salem
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, 53210 Pardubice, Czech Republic
| | - Ahmed Abdi
- Independent Researcher, Uxbridge UB9 6JH, UK
| | | | | | | | - Muhammad Zubair Saleem
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Roselit Mathew
- Department of Oncology, Biochemistry and Molecular Biology, and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Yaser Gamallat
- Department of Oncology, Biochemistry and Molecular Biology, and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
35
|
Harris MA, Savas P, Virassamy B, O'Malley MMR, Kay J, Mueller SN, Mackay LK, Salgado R, Loi S. Towards targeting the breast cancer immune microenvironment. Nat Rev Cancer 2024; 24:554-577. [PMID: 38969810 DOI: 10.1038/s41568-024-00714-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/31/2024] [Indexed: 07/07/2024]
Abstract
The tumour immune microenvironment is shaped by the crosstalk between cancer cells, immune cells, fibroblasts, endothelial cells and other stromal components. Although the immune tumour microenvironment (TME) serves as a source of therapeutic targets, it is also considered a friend or foe to tumour-directed therapies. This is readily illustrated by the importance of T cells in triple-negative breast cancer (TNBC), culminating in the advent of immune checkpoint therapy in combination with cytotoxic chemotherapy as standard of care for both early and advanced-stage TNBC, as well as recent promising signs of efficacy in a subset of hormone receptor-positive disease. In this Review, we discuss the various components of the immune TME in breast cancer and therapies that target or impact the immune TME, as well as the complexity of host physiology.
Collapse
Affiliation(s)
- Michael A Harris
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Peter Savas
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Balaji Virassamy
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Megan M R O'Malley
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jasmine Kay
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
| | - Roberto Salgado
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Pathology, ZAS Ziekenhuizen, Antwerp, Belgium
| | - Sherene Loi
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia.
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
| |
Collapse
|
36
|
Tang Q, Li H, Zhao XT, Li ZY, Ma CX, Zhou SQ, Chen DD. Opportunities and Challenges in the Development of Antibody-Drug Conjugate for Triple-Negative Breast Cancer: The Diverse Choices and Changing Needs. World J Oncol 2024; 15:527-542. [PMID: 38993251 PMCID: PMC11236369 DOI: 10.14740/wjon1853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/11/2024] [Indexed: 07/13/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous breast cancer subtype, which is also characterized by the aggressive phenotype, high recurrence rate, and poor prognosis. Antibody-drug conjugate (ADC) is a monoclonal antibody with a cytotoxic payload connected by a linker. ADC is gaining more and more attention as a targeted anti-cancer agent. Clinical studies of emerging ADC drugs such as sacituzumab govitecan and trastuzumab deruxtecan in patients with metastatic breast cancer (including TNBC) are progressing rapidly. In view of its excellent clinical efficacy and good tolerability, Sacituzumab govitecan gained accelerated approval by the FDA for the treatment of advanced metastatic TNBC in 2020. This review discusses the treatment status and challenges in TNBC, with an emphasis on the current status of ADC development and clinical trials in TNBC and metastatic breast cancer. We also summarize the clinical experience and future exploration directions of ADC development for TNBC patients.
Collapse
Affiliation(s)
- Qi Tang
- Department of Breast Surgery, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
- These authors contributed equally to this article
| | - Hui Li
- Department of Breast Surgery, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
- These authors contributed equally to this article
| | - Xin Tong Zhao
- Department of Breast Surgery, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
- These authors contributed equally to this article
| | - Ze Ying Li
- Department of Breast Surgery, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Chun Xiao Ma
- Department of Breast Surgery, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Shao Qiang Zhou
- Department of Breast Surgery, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - De Dian Chen
- Department of Breast Surgery, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| |
Collapse
|
37
|
Wang L, Li J, Mei N, Chen H, Niu L, He J, Wang R. Identifying subtypes and developing prognostic models based on N6-methyladenosine and immune microenvironment related genes in breast cancer. Sci Rep 2024; 14:16586. [PMID: 39020010 PMCID: PMC11255230 DOI: 10.1038/s41598-024-67477-w] [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: 05/14/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024] Open
Abstract
Breast cancer (BC) is the most prevalent cancer in women globally. The tumor microenvironment (TME), comprising epithelial tumor cells and stromal elements, is vital for breast tumor development. N6-methyladenosine (m6A) modification plays a key role in RNA metabolism, influencing its various aspects such as stability and translation. There is a notable link between m6A methylation and immune cells in the TME, although this relationship is complex and not fully deciphered. In this research, BC expression and clinicopathological data from TCGA were scrutinized to assess expression profiles, mutations, and CNVs of 31 m6A genes and immune microenvironment-related genes, examining their correlations, functions, and prognostic impacts. Lasso and Cox regression identified prognostic genes for constructing a nomogram. Single-cell analyses mapped the distribution and patterns of these genes in BC cell development. We investigated associations between gene-derived risk scores and factors like immune infiltration, TME, checkpoints, TMB, CSC indices, and drug response. As a complement to computational analyses, in vitro experiments were conducted to confirm these expression patterns. We included 31 m6A regulatory genes and discovered a correlation between these genes and the extent of immune cell infiltration. Subsequently, a 7-gene risk score was generated, encompassing HSPA2, TAP1, ULBP2, CXCL1, RBP1, STC2, and FLT3. It was observed that the low-risk group exhibited better overall survival (OS) in BC, with higher immune scores but lower tumor mutational burden (TMB) and cancer stem cell (CSC) indices, as well as lower IC50 values for commonly used drugs. To enhance clinical applicability, age and stage were incorporated into the risk score, and a more comprehensive nomogram was constructed to predict OS. This nomogram was validated and demonstrated good predictive performance, with area under the curve (AUC) values for 1-year, 3-year, and 5-year OS being 0.848, 0.807, and 0.759, respectively. Our findings highlight the profound impact of prognostic-related genes on BC immune response and prognostic outcomes, suggesting that modulation of the m6A-immune pathway could offer new avenues for personalized BC treatment and potentially improve clinical outcomes.
Collapse
Affiliation(s)
- Lizhao Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jianpeng Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Nan Mei
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Heyan Chen
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Ligang Niu
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jianjun He
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| | - Ru Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
38
|
Choi Y, Kim SA, Jung H, Kim E, Kim YK, Kim S, Kim J, Lee Y, Jo MK, Woo J, Cho Y, Lee D, Choi H, Jeong C, Nam GH, Kwon M, Kim IS. Novel insights into paclitaxel's role on tumor-associated macrophages in enhancing PD-1 blockade in breast cancer treatment. J Immunother Cancer 2024; 12:e008864. [PMID: 39009452 PMCID: PMC11253755 DOI: 10.1136/jitc-2024-008864] [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] [Accepted: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) poses unique challenges due to its complex nature and the need for more effective treatments. Recent studies showed encouraging outcomes from combining paclitaxel (PTX) with programmed cell death protein-1 (PD-1) blockade in treating TNBC, although the exact mechanisms behind the improved results are unclear. METHODS We employed an integrated approach, analyzing spatial transcriptomics and single-cell RNA sequencing data from TNBC patients to understand why the combination of PTX and PD-1 blockade showed better response in TNBC patients. We focused on toll-like receptor 4 (TLR4), a receptor of PTX, and its role in modulating the cross-presentation signaling pathways in tumor-associated macrophages (TAMs) within the tumor microenvironment. Leveraging insights obtained from patient-derived data, we conducted in vitro experiments using immunosuppressive bone marrow-derived macrophages (iBMDMs) to validate if PTX could augment the cross-presentation and phagocytosis activities. Subsequently, we extended our study to an in vivo murine model of TNBC to ascertain the effects of PTX on the cross-presentation capabilities of TAMs and its downstream impact on CD8+ T cell-mediated immune responses. RESULTS Data analysis from TNBC patients revealed that the activation of TLR4 and cross-presentation signaling pathways are crucial for the antitumor efficacy of PTX. In vitro studies showed that PTX treatment enhances the cross-presentation ability of iBMDMs. In vivo experiments demonstrated that PTX activates TLR4-dependent cross-presentation in TAMs, improving CD8+ T cell-mediated antitumor responses. The efficacy of PTX in promoting antitumor immunity was elicited when combined with PD-1 blockade, suggesting a complementary interaction. CONCLUSIONS This study reveals how PTX boosts the effectiveness of PD-1 inhibitors in treating TNBC. We found that PTX activates TLR4 signaling in TAMs. This activation enhances their ability to present antigens, thereby boosting CD8+ T cell antitumor responses. These findings not only shed light on PTX's immunomodulatory role in TNBC but also underscore the potential of targeting TAMs' antigen presentation capabilities in immunotherapy approaches.
Collapse
Affiliation(s)
- Yoonjeong Choi
- SHIFTBIO INC, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hanul Jung
- SHIFTBIO INC, Seoul, Republic of Korea
- Department of Otolaryngology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eunhae Kim
- SHIFTBIO INC, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | | | | | | | - Yeji Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Min Kyoung Jo
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jiwan Woo
- Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yakdol Cho
- Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | | | - Hongyoon Choi
- Portrai Inc, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Cherlhyun Jeong
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, University of Science and Technology, Seoul, Republic of Korea
| | - Gi-Hoon Nam
- SHIFTBIO INC, Seoul, Republic of Korea
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Minsu Kwon
- Department of Otolaryngology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| |
Collapse
|
39
|
Hammershøi Madsen AM, Løvendahl Eefsen RH, Nielsen D, Kümler I. Targeted Treatment of Metastatic Triple-Negative Breast Cancer: A Systematic Review. Breast J 2024; 2024:9083055. [PMID: 39742383 PMCID: PMC11257761 DOI: 10.1155/2024/9083055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 01/03/2025]
Abstract
Introduction Triple-negative breast cancer (TNBC) is a subgroup of breast cancer characterized by the absence of estrogen and the human epidermal 2 receptor and also a lack of targeted therapy options. Chemotherapy has so far been the only approved treatment option, and patients with metastatic cancer have a dismal prognosis with a median overall survival (OS) of approximately 14 months. Identification of druggable targets for metastatic TNBC is therefore of special interest. Methods A systematic search was performed, to review the existing evidence on targeted therapies in metastatic TNBC. Results A total of 37 phase 2/3 studies were identified, evaluating 29 different targeted agents. In this review, results on progression free survival (PFS) and OS are presented. Conclusion In most of the studies included, no improvement was observed for neither PFS nor OS; however, a few studies did show improvement with targeted agents and have led to new treatment options in subgroups of patients. The antibody drug conjugate, sacituzumab govitecan, demonstrated superior PFS and OS in comparison to chemotherapy. Immunotherapy with checkpoint inhibitors such as atezolizumab and pembrolizumab is now recommended as a first-line treatment option for patients with expression a PD-L1 positive tumor. Finally, the poly adenosine diphosphate-ribose polymerase (PARP) inhibitors talazoparib and olaparib are recommended, as first-line treatment options in patients with metastatic breast cancer and a germline BRCA mutation, but an immune checkpoint inhibitor should be considered for the subset of these patients who are PD-L1 positive.
Collapse
Affiliation(s)
- Anna Martha Hammershøi Madsen
- Department of Oncology 54 B1Herlev HospitalUniversity of Copenhagen, Herlev Ringvej 75, DK-2730, Copenhagen, Denmark
| | - Rikke Helene Løvendahl Eefsen
- Department of Oncology 54 B1Herlev HospitalUniversity of Copenhagen, Herlev Ringvej 75, DK-2730, Copenhagen, Denmark
| | - Dorte Nielsen
- Department of Oncology 54 B1Herlev HospitalUniversity of Copenhagen, Herlev Ringvej 75, DK-2730, Copenhagen, Denmark
| | - Iben Kümler
- Department of Oncology 54 B1Herlev HospitalUniversity of Copenhagen, Herlev Ringvej 75, DK-2730, Copenhagen, Denmark
| |
Collapse
|
40
|
Wang Y, Sun Y, Lu F, Zhao X, Nie Z, Zhu F, He B. Efficacy and safety of a combination treatment of immune checkpoint inhibitors in metastatic breast cancer: a systematic review and meta-analysis. Clin Transl Oncol 2024; 26:1725-1737. [PMID: 38587602 DOI: 10.1007/s12094-024-03396-6] [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: 09/27/2023] [Accepted: 01/22/2024] [Indexed: 04/09/2024]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) in combination with chemotherapy have showed its benefits in clinical studies, and here we conducted a further evaluation on the safety and efficacy of this treatment strategy. METHODS A systematic literature review was conducted in PubMed, Embase and Cochrane Library to identify clinical studies on ICIs and chemotherapy for metastatic breast cancer. The primary efficacy endpoints were progression-free survival (PFS) and overall survival (OS), and adverse events (AEs) were analyzed. Random or fixed effects models were used to estimate pooled Hazard ratio (HR), odds ratio (OR) and the data of 95% confidence interval (CI) depend on the Heterogeneity. Cochrane risk assessment tool was used to assess risk of bias. We also drew forest plots and funnel plots, respectively. RESULTS Seven studies with intend-to-treat (ITT) population for 3255 patients were analyzed. ICIs pooled therapy showed clinical benefits compared with chemotherapy alone, improving PFS (HR = 0.81, 95% CI: 0.74-0.90) of patients with metastatic triple negative breast cancer (mTNBC), especially in patients with PD-L1-positive tumors. However, it had no effect on OS (HR = 0.92, 95% CI 0.85-1.01). Besides, mTNBC patients received pooled therapy were less frequently to have AEs (OR = 1.30, 95% CI: 1.09-1.54). In patients with metastatic Human Epidermal Growth Factor Receptor 2 (HER2) negative breast cancer, pooled therapy showed no benefit for PFS (HR = 0.80, 95% CI: 0.50-1.28) and OS (HR = 0.87, 95% CI: 0.48-1.58). CONCLUSION Pooled therapy had improved PFS in mTNBC patients, especially in patients with PD-L1-positive tumors, and it was less likely to cause grade ≥ 3 AEs.
Collapse
Affiliation(s)
- Ying Wang
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Yalan Sun
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Fang Lu
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Xianghong Zhao
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Zhenlin Nie
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Feng Zhu
- Department of Laboratory Medicine, Nanjing Jiangning People's Hospital, 68 Gushan Road, Jiangning District, Nanjing, Jiangsu, 211100, China.
| | - Bangshun He
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China.
| |
Collapse
|
41
|
Li L, Guo Y, Gong B, Wang S, Wang MM, Sun P, Jiang S, Yang L. Association between tertiary lymphoid structures and clinical outcomes in cancer patients treated with immune checkpoint inhibitors: an updated meta-analysis. Front Immunol 2024; 15:1385802. [PMID: 38994363 PMCID: PMC11236553 DOI: 10.3389/fimmu.2024.1385802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024] Open
Abstract
Background Although numerous studies have reported the association between tertiary lymphoid structures (TLSs) and clinical outcomes in cancer patients treated with immune checkpoint inhibitors (ICIs), there remains a lack of a newer and more comprehensive meta-analysis. The main objective of this study is to explore prognostic biomarkers in immunotherapy-related patients, through analyzing the associations between tertiary lymphoid structures (TLSs) and clinical outcomes in cancer patients treated with ICIs, so as to investigate their prognostic value in cancer patients treated with ICIs. Methods A comprehensive search was conducted until February 2024 across PubMed, Embase, Web of Science, and the Cochrane Library databases to identify relevant studies evaluating the association between tertiary lymphoid structures and clinical outcomes in cancer patients treated with ICIs. The clinical outcomes were overall survival (OS), progression-free survival (PFS), and objective response rate (ORR). Results Thirteen studies were incorporated in this meta-analysis, among which nine evaluated the prognostic value of TLSs. The results showed the high levels of TLSs predicted a significantly prolonged OS (pooled HR = 0.35, 95% CI: 0.24-0.53, p < 0.001) and PFS (pooled HR = 0.47, 95% CI: 0.31-0.72, p < 0.001), while lower ORR (pooled OR = 3.78, 95% CI: 2.26-6.33, p < 0.001) in cancer patients treated with ICIs. Conclusion Our results indicated that high levels of TLSs could predict a favorable prognosis for cancer patients treated with ICIs and have the potential to become a prognostic biomarker of immunotherapy-related patients.
Collapse
Affiliation(s)
- Lingli Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bingxin Gong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Sichen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | | | - Peng Sun
- Clinical & Technical Support, Philips Healthcare, Beijing, China
| | - Shanshan Jiang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
42
|
Pan F, Liu J, Chen Y, Zhu B, Chen W, Yang Y, Zhu C, Zhao H, Liu X, Xu Y, Xu X, Huo L, Xie L, Wang R, Gu J, Huang G. Chemotherapy-induced high expression of IL23A enhances efficacy of anti-PD-1 therapy in TNBC by co-activating the PI3K-AKT signaling pathway of CTLs. Sci Rep 2024; 14:14248. [PMID: 38902343 PMCID: PMC11189934 DOI: 10.1038/s41598-024-65129-7] [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: 02/02/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024] Open
Abstract
Treatment of advanced triple-negative breast cancer (TNBC) is a great challenge in clinical practice. The immune checkpoints are a category of immunosuppressive molecules that cancer could hijack and impede anti-tumor immunity. Targeting immune checkpoints, such as anti-programmed cell death 1 (PD-1) therapy, is a promising therapeutic strategy in TNBC. The efficacy and safety of PD-1 monoclonal antibody (mAb) with chemotherapy have been validated in TNBC patients. However, the precise mechanisms underlying the synergistic effect of chemotherapy and anti-PD-1 therapy have not been elucidated, causing the TNBC patients that might benefit from this combination regimen not to be well selected. In the present work, we found that IL-23, an immunological cytokine, is significantly upregulated after chemotherapy in TNBC cells and plays a vital role in enhancing the anti-tumor immune response of cytotoxic T cells (CTLs), especially in combination with PD-1 mAb. In addition, the combination of IL-23 and PD-1 mAb could synergistically inhibit the expression of Phosphoinositide-3-Kinase Regulatory Subunit 1 (PIK3R1), which is a regulatory subunit of PI3K and inhibit p110 activity, and promote phosphorylation of AKT in TNBC-specific CTLs. Our findings might provide a molecular marker that could be used to predict the effects of combination chemotherapy therapy and PD-1 mAb in TNBC.
Collapse
Affiliation(s)
- Fan Pan
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China
| | - Jiajing Liu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Ningbo University, Liuting Road 59#, Ningbo, 315010, China
| | - Ying Chen
- Medical School of Nanjing University, Nanjing University, Hankou Road 22#, Nanjing, 210093, China
| | - Binghan Zhu
- Department of Respiratory Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing University Medical School, Zhongshan Road 321#, Nanjing, 210008, China
| | - Weiwei Chen
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China
| | - Yuchen Yang
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China
| | - Chunyan Zhu
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China
| | - Hua Zhao
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China
| | - Xiaobei Liu
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China
| | - Yichen Xu
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China
| | - Xiaofan Xu
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China
| | - Liqun Huo
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China
| | - Li Xie
- Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321#, Nanjing, 210008, China.
| | - Rui Wang
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China.
| | - Jun Gu
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China.
| | - Guichun Huang
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Eastern Zhongshan Road 305#, Nanjing, 210002, China.
| |
Collapse
|
43
|
Ruocco MR, Gisonna A, Acampora V, D’Agostino A, Carrese B, Santoro J, Venuta A, Nasso R, Rocco N, Russo D, Cavaliere A, Altobelli GG, Masone S, Avagliano A, Arcucci A, Fiume G. Guardians and Mediators of Metastasis: Exploring T Lymphocytes, Myeloid-Derived Suppressor Cells, and Tumor-Associated Macrophages in the Breast Cancer Microenvironment. Int J Mol Sci 2024; 25:6224. [PMID: 38892411 PMCID: PMC11172575 DOI: 10.3390/ijms25116224] [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: 02/20/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Breast cancers (BCs) are solid tumors composed of heterogeneous tissues consisting of cancer cells and an ever-changing tumor microenvironment (TME). The TME includes, among other non-cancer cell types, immune cells influencing the immune context of cancer tissues. In particular, the cross talk of immune cells and their interactions with cancer cells dramatically influence BC dissemination, immunoediting, and the outcomes of cancer therapies. Tumor-infiltrating lymphocytes (TILs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) represent prominent immune cell populations of breast TMEs, and they have important roles in cancer immunoescape and dissemination. Therefore, in this article we review the features of TILs, TAMs, and MDSCs in BCs. Moreover, we highlight the mechanisms by which these immune cells remodel the immune TME and lead to breast cancer metastasis.
Collapse
Affiliation(s)
- Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.R.R.); (A.G.)
| | - Armando Gisonna
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.R.R.); (A.G.)
| | - Vittoria Acampora
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Anna D’Agostino
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Barbara Carrese
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Jessie Santoro
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Alessandro Venuta
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Rosarita Nasso
- Department of Movement Sciences and Wellness, University of Naples “Parthenope”, 80133 Naples, Italy;
| | - Nicola Rocco
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | - Daniela Russo
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | | | - Giovanna Giuseppina Altobelli
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy;
| |
Collapse
|
44
|
Luo J, Ng W, Liu Y, Wang L, Gong C, Zhou Y, Fang C, Zhu S, Yao C. Rocaglamide promotes infiltration and differentiation of T cells and coordinates with PD-1 inhibitor to overcome checkpoint resistance in multiple tumor models. Cancer Immunol Immunother 2024; 73:137. [PMID: 38833034 PMCID: PMC11150362 DOI: 10.1007/s00262-024-03706-5] [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: 11/22/2023] [Accepted: 04/19/2024] [Indexed: 06/06/2024]
Abstract
Tumor-infiltrating lymphocyte (TIL) deficiency is the most conspicuous obstacle to limit the cancer immunotherapy. Immune checkpoint inhibitors (ICIs), such as anti-PD-1 antibody, have achieved great success in clinical practice. However, due to the limitation of response rates of ICIs, some patients fail to benefit from monotherapy. Thus, novel combination therapy that could improve the response rates emerges as new strategies for cancer treatment. Here, we reported that the natural product rocaglamide (RocA) increased tumor-infiltrating T cells and promoted Th17 differentiation of CD4+ TILs. Despite RocA monotherapy upregulated PD-1 expression of TILs, which was considered as the consequence of T cell activation, combining RocA with anti-PD-1 antibody significantly downregulated the expression of PD-1 and promoted proliferation of TILs. Taken together, these findings demonstrated that RocA could fuel the T cell anti-tumor immunity and revealed the remarkable potential of RocA as a therapeutic candidate when combining with the ICIs.
Collapse
Affiliation(s)
- Jiaojiao Luo
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wanyi Ng
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yangli Liu
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lixin Wang
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chenyuan Gong
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yufu Zhou
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Cheng Fang
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shiguo Zhu
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Chao Yao
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
45
|
Downs-Canner S, Weiss A. Systemic Therapy Advances for HER2-Positive and Triple Negative Breast Cancer: What the Surgeon Needs to Know. Clin Breast Cancer 2024; 24:328-336. [PMID: 38616443 DOI: 10.1016/j.clbc.2024.03.004] [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: 12/14/2023] [Revised: 02/17/2024] [Accepted: 03/08/2024] [Indexed: 04/16/2024]
Abstract
Neoadjuvant systemic therapy (NST) was initially reserved for unresectable patients however it has been increasingly used to facilitate breast conservation, downstage the axilla, and inform adjuvant therapy decisions based on response. For patients with HER2+ and triple-negative breast cancer (TNBC), clinical trials have resulted in the ability to individualize treatment regimens. For HER2+ breast cancer, de-escalation of neoadjuvant regimens to minimize cytotoxic chemotherapy and de-escalation or escalation of adjuvant regimens based on response have been effective. For TNBC, the approval of the combination of chemotherapy plus immunotherapy in the neoadjuvant setting has resulted in a major practice shift and opened the door to many additional treatment questions including de-escalation of the chemotherapy backbone or the adjuvant regimen. For both HER2+ and TNBC, most patients are treated with NST except those with very small tumors. Efforts are also being made to optimally identify patients with T1c tumors who may benefit from more aggressive NST. For patients treated according to or enrolled in NST de-escalation trials, breast conservation (even those who become eligible based on response to NST) and sentinel lymph node biopsy when cN0 at the completion of NST are safe and feasible. Continued involvement of surgeons and multidisciplinary teams in the design and reporting of trials will streamline their adoption into clinical practice. Surgeons need to remain aware of ongoing systemic therapy trials to appropriately select patients for NST and plan for appropriate post-neoadjuvant surgical care.
Collapse
Affiliation(s)
- Stephanie Downs-Canner
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY.
| | - Anna Weiss
- Division of Surgical Oncology, Department of Surgery, University of Rochester Medical Center, Rochester, NY; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
46
|
Michaels E, Chen N, Nanda R. The Role of Immunotherapy in Triple-Negative Breast Cancer (TNBC). Clin Breast Cancer 2024; 24:263-270. [PMID: 38582617 DOI: 10.1016/j.clbc.2024.03.001] [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: 11/01/2023] [Revised: 02/15/2024] [Accepted: 03/02/2024] [Indexed: 04/08/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype, generally associated with a high risk of recurrence and poor prognosis. Our understanding of the heterogeneity of TNBC has increased over the past decade, and with it a recognition that some TNBCs are immunogenically active. This finding has led to the investigation of immunotherapy-based approaches for treatment of both early and advanced-stage TNBC. In this review, we provide an overview of the biologic rationale for immunotherapy use in TNBC, and review data from seminal trials which have culminated in the approval of immunotherapy for both early and advanced TNBC. Identification of predictive biomarkers to aid in treatment selection, development of novel treatment combinations to combat resistance, and refinement of therapeutic targets enables continued improvement in outcomes with immunotherapy for TNBC.
Collapse
Affiliation(s)
- Elena Michaels
- Department of Medicine, The University of Chicago Medicine, Chicago, IL
| | - Nan Chen
- Department of Medicine, The University of Chicago Medicine, Chicago, IL; Department of Medicine, The University of Chicago Comprehensive Cancer Center, Chicago, IL
| | - Rita Nanda
- Department of Medicine, The University of Chicago Medicine, Chicago, IL; Department of Medicine, The University of Chicago Comprehensive Cancer Center, Chicago, IL.
| |
Collapse
|
47
|
Arulraj T, Wang H, Deshpande A, Varadhan R, Emens LA, Jaffee EM, Fertig EJ, Santa-Maria CA, Popel AS. Virtual patient analysis identifies strategies to improve the performance of predictive biomarkers for PD-1 blockade. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595235. [PMID: 38826266 PMCID: PMC11142158 DOI: 10.1101/2024.05.21.595235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Patients with metastatic triple-negative breast cancer (TNBC) show variable responses to PD-1 inhibition. Efficient patient selection by predictive biomarkers would be desirable, but is hindered by the limited performance of existing biomarkers. Here, we leveraged in-silico patient cohorts generated using a quantitative systems pharmacology model of metastatic TNBC, informed by transcriptomic and clinical data, to explore potential ways to improve patient selection. We tested 90 biomarker candidates, including various cellular and molecular species, by a cutoff-based biomarker testing algorithm combined with machine learning-based feature selection. Combinations of pre-treatment biomarkers improved the specificity compared to single biomarkers at the cost of reduced sensitivity. On the other hand, early on-treatment biomarkers, such as the relative change in tumor diameter from baseline measured at two weeks after treatment initiation, achieved remarkably higher sensitivity and specificity. Further, blood-based biomarkers had a comparable ability to tumor- or lymph node-based biomarkers in identifying a subset of responders, potentially suggesting a less invasive way for patient selection.
Collapse
|
48
|
Martínez-Galán J, Jiménez-Luna C, Rodriguez I, Maza E, García-Collado C, Rodríguez-Fernández A, López-Hidalgo JL, Caba O. Metastatic pancreatic and lung cancer patient in complete remission following immunotherapy: A case report and review of literature. World J Gastrointest Oncol 2024; 16:2233-2240. [PMID: 38764840 PMCID: PMC11099454 DOI: 10.4251/wjgo.v16.i5.2233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/09/2024] [Accepted: 03/13/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND Metastatic pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with dispiriting survival data. Immunotherapy is a promising approach to many cancer types, but achieves poor outcomes in advanced PDAC due to its immunosuppressive tumor microenvironment. We describe a case of metastatic PDAC effectively treated with pembrolizumab. CASE SUMMARY We report the case of a 67-year-old woman with unresectable locally advanced PDAC, treated with gemcitabine plus nab-paclitaxel followed by radiotherapy plus capecitabine. At nine months, pancreatic tumor progression was observed at the level of the hepatic hilum with the appearance of a new pulmonary nodule suggestive of a second primary, confirmed by left lung biopsy. Systemic immunotherapy was then initiated with pembrolizumab, an immune checkpoint inhibitor targeting programmed cell death protein-1 that covers the two tumor types. The patient showed a complete metabolic response that was maintained throughout the treatment. The patient continues to be disease-free at 5.6 years since the start of immunotherapy. CONCLUSION These results suggest that the administration of pembrolizumab after chemoradiotherapy has a beneficial effect in patients with metastatic PDAC. To our knowledge, this is the first reported case of a patient with metastatic PDAC and metastatic lung cancer showing such a long-lasting complete response after pembrolizumab treatment without curative surgery. Further studies are required to determine biomarkers that identify PDAC patients most likely to benefit from this immunotherapy.
Collapse
Affiliation(s)
- Joaquina Martínez-Galán
- Department of Medical Oncology, Virgen de las Nieves University Hospital, Granada 18014, Spain
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
| | - Cristina Jiménez-Luna
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada 18016, Spain
| | - Isabel Rodriguez
- Department of Medical Oncology, Virgen de las Nieves University Hospital, Granada 18014, Spain
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
| | - Elisabeth Maza
- Department of Medical Oncology, Virgen de las Nieves University Hospital, Granada 18014, Spain
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
| | - Carlos García-Collado
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
- Pharmacogenetics Unit, Virgen de las Nieves University Hospital, Granada 18014, Spain
| | - Antonio Rodríguez-Fernández
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
- Department of Medicina Nuclear, Virgen de las Nieves University Hospital, Granada 18014, Spain
| | - Javier Luis López-Hidalgo
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
- Department of Anatomía Patológica, Virgen de las Nieves University Hospital, Granada 18014, Spain
| | - Octavio Caba
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada 18016, Spain
| |
Collapse
|
49
|
Cortes J, Winer EP, Lipatov O, Im S, Gonçalves A, Muñoz‐Couselo E, Lee KS, Schmid P, Tamura K, Testa L, Witzel I, Ohtani S, Hund S, Kulangara K, Karantza V, Mejia JA, Ma J, Jelinic P, Huang L, Pruitt SK, Emancipator K. Contribution of tumour and immune cells to PD-L1 expression as a predictive biomarker in metastatic triple-negative breast cancer: exploratory analysis from KEYNOTE-119. J Pathol Clin Res 2024; 10:e12371. [PMID: 38627977 PMCID: PMC11021797 DOI: 10.1002/2056-4538.12371] [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: 10/06/2023] [Revised: 02/27/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024]
Abstract
The efficacy of pembrolizumab monotherapy versus chemotherapy increased with increasing programmed death ligand 1 (PD-L1) expression, as quantified by combined positive score (CPS; PD-L1 expression on both tumour cells and immune cells) in patients with previously treated metastatic triple-negative breast cancer (mTNBC) in the phase 3 KEYNOTE-119 study. This exploratory analysis was conducted to determine whether the expression of PD-L1 on tumour cells contributes to the predictive value of PD-L1 CPS in mTNBC. PD-L1 expression in tumour samples was assessed using PD-L1 IHC 22C3 pharmDx and quantified using both CPS and tumour proportion score (TPS; PD-L1 expression on tumour cells alone). Calculated immune cell density (CID) was defined as CPS minus TPS. The ability of each scoring method (CPS, TPS, and CID) to predict clinical outcomes with pembrolizumab was evaluated. With pembrolizumab, the area under the receiver operating characteristic curve was 0.69 (95% CI = 0.58-0.80) for CPS, 0.55 (95% CI = 0.46-0.64) for TPS, and 0.67 (95% CI = 0.56-0.77) for CID. After correction for cutoff prevalence, CPS performed as well as, if not better than, CID with respect to predicting objective response rate, progression-free survival, and overall survival. Data from this exploratory analysis suggest that, although PD-L1 expression on immune cells alone is predictive of response to programmed death 1 blockade in mTNBC, adding tumour PD-L1 expression assessment (i.e. CPS, which combines immune cell and tumour cell PD-L1 expression) may improve prediction. PD-L1 CPS thus remains an effective and broadly applicable uniform scoring system for enriching response to programmed death 1 blockade with pembrolizumab in mTNBC as well as other tumour types.
Collapse
Affiliation(s)
- Javier Cortes
- Oncology DepartmentInternational Breast Cancer Center (BCC), Pangaea Oncology, QuirónsaludBarcelonaSpain
- Department of MedicineFaculty of Biomedical and Health Sciences, European University of MadridMadridSpain
| | - Eric P Winer
- Yale Cancer CenterYale School of MedicineNew HavenCTUSA
- Present address:
Yale Cancer CenterNew HavenCTUSA
| | - Oleg Lipatov
- Department of OncologyRepublican Clinical Oncology Dispensary of the Ministry of Public Health of Bashkortostan RepublicUfaRussia
| | - Seock‐Ah Im
- Department of Internal MedicineSeoul National University Cancer Research Institute, Seoul National University College of Medicine, Seoul National UniversitySeoulRepublic of Korea
| | - Anthony Gonçalves
- Aix Marseille University, CNRS, INSERM, Department of Medical OncologyInstitut Paoli‐Calmettes, CRCMMarseilleFrance
| | - Eva Muñoz‐Couselo
- Department of Medical OncologyVall d'Hebron University HospitalBarcelonaSpain
| | - Keun Seok Lee
- Department of Medical OncologyCenter for Breast Cancer, National Cancer CenterGoyangRepublic of Korea
| | - Peter Schmid
- Department of Cancer MedicineBarts ECMC, Barts Cancer Institute, Queen Mary University of London, and Barts Health NHS TrustLondonUK
| | - Kenji Tamura
- Department of Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Laura Testa
- Instituto do Câncer do Estado de São Paulo Octavio Frias de OliveiraHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
| | - Isabell Witzel
- Department of GynecologyUniversity Medical Center Hamburg–EppendorfHamburgGermany
- Department of GynecologyUniversity of Zurich, University Hospital ZurichZurichSwitzerland
| | - Shoichiro Ohtani
- Division of Breast SurgeryHiroshima City Hiroshima Citizens HospitalHiroshimaJapan
| | - Stephanie Hund
- Diagnostics and Genomics GroupAgilent TechnologiesCarpinteriaCAUSA
| | - Karina Kulangara
- Diagnostics and Genomics GroupAgilent TechnologiesCarpinteriaCAUSA
| | | | - Jaime A Mejia
- Department of Medical OncologyMerck & Co., Inc.RahwayNJUSA
| | - Junshui Ma
- Early Development StatisticsMerck & Co., Inc.RahwayNJUSA
| | - Petar Jelinic
- Department of Medical OncologyMerck & Co., Inc.RahwayNJUSA
| | - Lingkang Huang
- Early Development StatisticsMerck & Co., Inc.RahwayNJUSA
| | - Scott K Pruitt
- Department of Medical OncologyMerck & Co., Inc.RahwayNJUSA
| | - Kenneth Emancipator
- Early Oncology DevelopmentMerck & Co., Inc.RahwayNJUSA
- Present address:
Precision Medicine, AbbVie, Inc.North ChicagoILUSA
| |
Collapse
|
50
|
Chauhan SK, Dunn C, Andresen NK, Røssevold AH, Skorstad G, Sike A, Gilje B, Raj SX, Huse K, Naume B, Kyte JA. Peripheral immune cells in metastatic breast cancer patients display a systemic immunosuppressed signature consistent with chronic inflammation. NPJ Breast Cancer 2024; 10:30. [PMID: 38653982 DOI: 10.1038/s41523-024-00638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 04/13/2024] [Indexed: 04/25/2024] Open
Abstract
Immunotherapies blocking the PD-1/PD-L1 checkpoint show some efficacy in metastatic breast cancer (mBC) but are often hindered by immunosuppressive mechanisms. Understanding these mechanisms is crucial for personalized treatments, with peripheral blood monitoring representing a practical alternative to repeated biopsies. In the present study, we performed a comprehensive mass cytometry analysis of peripheral blood immune cells in 104 patients with HER2 negative mBC and 20 healthy donors (HD). We found that mBC patients had significantly elevated monocyte levels and reduced levels of CD4+ T cells and plasmacytoid dendritic cells, when compared to HD. Furthermore, mBC patients had more effector T cells and regulatory T cells, increased expression of immune checkpoints and other activation/exhaustion markers, and a shift to a Th2/Th17 phenotype. Furthermore, T-cell phenotypes identified by mass cytometry correlated with functionality as assessed by IFN-γ production. Additional analysis indicated that previous chemotherapy and CDK4/6 inhibition impacted the numbers and phenotype of immune cells. From 63 of the patients, fresh tumor samples were analyzed by flow cytometry. Paired PBMC-tumor analysis showed moderate correlations between peripheral CD4+ T and NK cells with their counterparts in tumors. Further, a CD4+ T cell cluster in PBMCs, that co-expressed multiple checkpoint receptors, was negatively associated with CD4+ T cell tumor infiltration. In conclusion, the identified systemic immune signatures indicate an immune-suppressed environment in mBC patients who had progressed/relapsed on standard treatments, and is consistent with ongoing chronic inflammation. These activated immuno-suppressive mechanisms may be investigated as therapeutic targets, and for use as biomarkers of response or treatment resistance.
Collapse
Affiliation(s)
- Sudhir Kumar Chauhan
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Claire Dunn
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Nikolai Kragøe Andresen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Hagen Røssevold
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gjertrud Skorstad
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Adam Sike
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bjørnar Gilje
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Sunil Xavier Raj
- Department of Oncology, St Olav University Hospital, Trondheim, Norway
| | - Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bjørn Naume
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Jon Amund Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway.
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway.
| |
Collapse
|