1
|
Wu Q, Siddharth S, Verma D, Parida S, Sharma D. TRIM29 upregulation contributes to chemoresistance in triple negative breast cancer via modulating S100P-β-catenin axis. Cell Commun Signal 2025; 23:244. [PMID: 40420099 DOI: 10.1186/s12964-025-02233-9] [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: 11/20/2024] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Triple negative breast cancer, an inherently aggressive disease, is further impaired by the limited therapeutic options and chemotherapy-resistance; hence, elucidating the signaling nodes underlying chemotherapy resistance is of major interest. Focusing on the differentially expressed genes in recurrent TNBC, we identified TRIM29, a ubiquitin ligase belonging to TRIM family, as a uniquely enriched protein in chemoresistant TNBC. Here, we demonstrate that chemoresistant TNBC cells are inherently aggressive, exhibiting elevated growth and migration potential compared to chemosensitive cells, and in particular, they possess higher TRIM29 expression whose expression level modulation results in altered chemosensitivity. TRIM29 overexpression reduces chemotherapy response whereas TRIM29 knockout not only increases chemosensitivity but also reduces TNBC tumor growth. Tumor-dissociated cells maintain TRIM29 knockout status as well as exhibit similar functional alterations as chemoresistant TNBC cells. Mechanistically, RNA-sequencing of parental-chemosensitive, chemoresistant-inherently overexpressing TRIM29 and chemoresistant-TRIM29 knockout TNBC cells reveals a unique set of genes (S100P, SERPINB3, SERPINB4, CEACAM5, CEACAM6 and CDH6) showing significant upregulation with the acquisition of chemoresistance and downregulation with the TRIM29 knockout. Furthermore, an enrichment of β-catenin pathway in chemoresistant TNBC cells is observed. We uncovered a functional network where S100P, a metastasis inducing secretory factor, bidirectionally interacts with TRIM29, and modulates the expression of SERPINB3, SERPINB4, CEACAM5, CEACAM6 as well as β-catenin pathway genes. Showing the functional importance, S100P inhibitor reduces the growth and mammosphere formation in chemoresistant TNBC. Moreover, combining β-catenin inhibitor with chemotherapy shows synergistic inhibition of chemoresistant TNBC cells. Indeed, higher expression of TRIM29, S100P and β-catenin associates with reduced recurrence free survival. This work proposes TRIM29 as an important node that modulates a unique gene network in chemoresistant TNBC and whose biological impact is mediated by modulation of S100P and β-catenin.
Collapse
Affiliation(s)
- Qitong Wu
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA
| | - Sumit Siddharth
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA.
| | - Deepak Verma
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA
| | - Sheetal Parida
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA
| | - Dipali Sharma
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA.
| |
Collapse
|
2
|
Jin P, Hasan MM, Pepper AGS, Mitchell S, Rahman KM, Pepper C. Design, synthesis and evaluation of pyrrolobenzodiazepine (PBD)-based PROTAC conjugates for the selective degradation of the NF-κB RelA/p65 subunit. RSC Med Chem 2025:d5md00316d. [PMID: 40443648 PMCID: PMC12117510 DOI: 10.1039/d5md00316d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Accepted: 05/08/2025] [Indexed: 06/02/2025] Open
Abstract
NF-κB signalling is frequently dysregulated in human cancers making it an attractive therapeutic target. Despite concerted efforts to generate NF-κB inhibitors, direct pharmacological inhibition of the kinases mediating canonical NF-κB has failed due to on-target toxicities in normal tissues. So, alternative strategies, designed to target specific components of the NF-κB signalling machinery, have the potential to selectively inhibit tumour cells whilst reducing the toxicities associated with broad inhibition of NF-κB in non-malignant cells. Here we present evidence that a C8-linked pyrrolobenzodiazepine (PBD) containing proteolysis-targeting chimera (PROTAC) selectively degrades the NF-κB subunit, RelA/p65, in a proteasome-dependent manner. Our lead PROTAC (JP-163-16, 15d) showed cytotoxicity with mean LC50 values of 2.9 μM in MDA-MB-231 cells, 0.14 μM in MEC-1 cells and 0.23 μM in primary chronic lymphocytic leukaemia cells. In contrast, 15d was two-logs less toxic in primary B- and T-lymphocytes (mean LD50 19.1 μM and 36.4 μM, respectively). Importantly, the development of 15d, by conjugating the C8-linked PBD with a cereblon-targeting ligand using a five-carbon linker, abolished the ability of the C8-linked PBD to bind to DNA, whilst demonstrating cytotoxicity in cancer cells associated with the degradation of RelA/p65. Mechanistically, 15d displayed PROTAC credentials through the selective degradation of NF-κB RelA/p65 in a proteasome-dependent manner and showed a five-fold reduction in potency in the cereblon deficient, lenalidomide resistant, myeloma cell line, RPMI-8226. To our knowledge, this work describes the first PROTAC capable of selective degradation of a single NF-κB subunit and highlights the therapeutic potential of our strategy for the treatment of RelA/p65-dependent tumours.
Collapse
Affiliation(s)
- Peiqin Jin
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London London SE1 9NH UK
| | - Md Mahbub Hasan
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London London SE1 9NH UK
| | - Andrea G S Pepper
- Brighton and Sussex Medical School, University of Brighton and University of Sussex Brighton BN1 9PX UK
| | - Simon Mitchell
- Brighton and Sussex Medical School, University of Brighton and University of Sussex Brighton BN1 9PX UK
| | - Khondaker Miraz Rahman
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London London SE1 9NH UK
| | - Chris Pepper
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London London SE1 9NH UK
- Brighton and Sussex Medical School, University of Brighton and University of Sussex Brighton BN1 9PX UK
| |
Collapse
|
3
|
Alaklabi S, Roy AM, Zagami P, Chakraborty A, Held N, Elijah J, George A, Attwood K, Shaikh SS, Chaudhary LN, Abdou Y, Gandhi S. Real-World Clinical Outcomes With Sacituzumab Govitecan in Metastatic Triple-Negative Breast Cancer. JCO Oncol Pract 2025; 21:620-628. [PMID: 39353151 DOI: 10.1200/op.24.00242] [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: 03/19/2024] [Revised: 06/19/2024] [Accepted: 08/14/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE Sacituzumab govitecan (SG) is approved for the treatment of metastatic triple-negative breast cancer (mTNBC). We report the real-world clinical effectiveness and toxicity data of SG in patients with mTNBC. METHODS A multi-institution retrospective study of patients with mTNBC treated with SG from January 2021 to May 2023 was conducted. Demographic and clinicopathologic variables were collected. Univariate and multivariate Cox regression models were used for survival analysis. RESULTS A total of 115 patients were included. The median age at SG initiation was 60 years (range, 31-85). All patients were female; 73 (63.5%) were White and 31 (27.0%) were Black. The median number of previous therapies in the metastatic setting was two (range, 0-8). Sixty-one (56.0%) patients had primary refractory disease. Median relative dose intensity was 92% (range, 33%-100%). Grade 3 or higher adverse events (AEs) were seen in 50.9% of patients, which included neutropenia (35.7%), anemia (27.0%), vomiting (16.5%), fatigue (8.7%), and diarrhea (7.0%). Dose reductions and treatment discontinuation due to AEs were required in 51.3% and 13.2%, respectively. The objective response rate (ORR) was 27.8%. Median overall survival was 9.6 months (95% CI, 7.8 to 12.9) and median progression-free survival (PFS) was 4.8 months (95% CI, 3.6 to 5.9). In patients with human epidermal growth factor receptor 2 (HER2)-low mTNBC who received trastuzumab deruxtecan (T-DXd) after SG, the ORR to T-DXd was 34.8% and median PFS was 7 months (95% CI, 4.6 to 10.1). CONCLUSION In a real-world cohort of heavily pretreated patients with mTNBC, SG retains its clinical activity. In a subgroup with HER2-low disease, T-DXd continues to demonstrate promising clinical activity after SG, supporting the use of sequential antibody-drug conjugates in this population.
Collapse
Affiliation(s)
- Sabah Alaklabi
- Department of Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Cancer Center of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Arya Mariam Roy
- Department of Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Paola Zagami
- Department of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Nicole Held
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Joseph Elijah
- Department of Pharmacology, Northeastern University School of Pharmacy and Pharmaceutical Sciences, Boston, PA
| | - Anthony George
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Kristopher Attwood
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Saba S Shaikh
- Department of Medicine, Division of Hematology/Oncology, University of Texas Health San Antonio, San Antonio, TX
| | - Lubna N Chaudhary
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Yara Abdou
- Department of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Shipra Gandhi
- Department of Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| |
Collapse
|
4
|
Ticha P, Northey JJ, Kersten K, Velozo HG, Ironside AJ, Zidek M, Drain A, Lakins JN, Chen YY, Tsai KK, Weaver VM. NCOR2 represses MHC class I molecule expression to drive metastatic progression of breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642060. [PMID: 40161756 PMCID: PMC11952456 DOI: 10.1101/2025.03.10.642060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Metastatic progression depends upon the ability of disseminated tumor cells to evade immune surveillance. MHC molecule expression facilitates T cell recognition and activation to permit the eradication of metastatic tumor cells. We identified nuclear corepressor 2 (NCOR2) as a key epigenetic regulator of MHC class I molecule expression on breast tumor cells. Patients with triple negative breast cancers (TNBC) that expressed high levels of NCOR2 also exhibited reduced metastasis free survival and decreased MHC class I expression, and the metastatic lesions in patients with TNBC had high nuclear NCOR2 and reduced CD8 T cell levels and activity. Genetically and experimentally reducing NCOR2 expression in tumor cells permitted interferon gamma upregulation of MHC class I, and potentiated CD8 T cell activity and induction of apoptosis to repress metastatic progression of disseminated breast cancer cells. These studies provide evidence to support NCOR2 as a targetable epigenetic regulator of metastasis towards which therapies could be developed to reduce patient mortality.
Collapse
Affiliation(s)
- Pavla Ticha
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
- Current address: Department of Plastic Surgery, 3rd Faculty of Medicine and University Hospital Kralovske Vinohrady, Charles University in Prague, Srobarova 50, 10034, Praha 10, Czech Republic
| | - Jason J. Northey
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelly Kersten
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
- Current address: Cancer Metabolism and Microenvironment Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Hugo González Velozo
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
- Laboratory of Tumor Microenvironment and Metastasis, Centro Ciencia & Vida, Santiago, Chile
| | | | - Martin Zidek
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Allison Drain
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathan N. Lakins
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yunn-Yi Chen
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelvin K. Tsai
- Laboratory of Advanced Molecular Therapeutics, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Valerie M. Weaver
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
5
|
McBean B, Abou Zeidane R, Lichtman-Mikol S, Hauk B, Speers J, Tidmore S, Flores CL, Rana PS, Pisano C, Liu M, Santola A, Montero A, Boyle AP, Speers CW. MELK as a Mediator of Stemness and Metastasis in Aggressive Subtypes of Breast Cancer. Int J Mol Sci 2025; 26:2245. [PMID: 40076867 PMCID: PMC11900306 DOI: 10.3390/ijms26052245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the breast cancer subtype with the poorest prognosis and lacks actionable molecular targets for treatment. Maternal embryonic leucine zipper kinase (MELK) is highly expressed in TNBC and has been implicated in poor clinical outcomes, though its mechanistic role in the aggressive biology of TNBC is poorly understood. Here, we demonstrate a role of MELK in TNBC progression and metastasis. Analysis of publicly available datasets revealed that high MELK expression correlates with worse overall survival, recurrence-free survival, and distant metastasis-free survival, and MELK is co-expressed with metastasis-related genes. Functional studies demonstrated that MELK inhibition, using genomic or pharmacologic inhibition, reduces mammosphere formation, migration, and invasion in high-MELK-expressing TNBC cell lines. Conversely, MELK overexpression in low-MELK-expressing cell lines significantly increased invasive capacity in vitro and metastatic potential in vivo, as evidenced by enhanced metastasis to the liver and lungs in a chorioallantoic membrane assay. These findings highlight MELK as a key regulator of TNBC aggressiveness and support its potential as a therapeutic target to mitigate metastasis and improve patient outcomes.
Collapse
Affiliation(s)
- Breanna McBean
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA; (B.M.); (A.P.B.)
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Reine Abou Zeidane
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Samuel Lichtman-Mikol
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Benjamin Hauk
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Johnathan Speers
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Savannah Tidmore
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Citlally Lopez Flores
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Priyanka S. Rana
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Courtney Pisano
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Meilan Liu
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA; (M.L.); (A.S.)
| | - Alyssa Santola
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA; (M.L.); (A.S.)
| | - Alberto Montero
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alan P. Boyle
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA; (B.M.); (A.P.B.)
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Corey W. Speers
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA; (M.L.); (A.S.)
| |
Collapse
|
6
|
Álvarez Pinheiro CG, Corral Tuesta C, Sánchez García E. [Management of triple negative metastatic breast cancer in elderly patients: treatment approach, tolerability and new target therapies. Regarding a case report]. Rev Esp Geriatr Gerontol 2025; 60:101579. [PMID: 39689440 DOI: 10.1016/j.regg.2024.101579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 12/19/2024]
Affiliation(s)
| | - Celia Corral Tuesta
- Servicio de Geriatría, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, España
| | | |
Collapse
|
7
|
Arora A, Seenu V, Parshad R, Bansal VK, Dhar A, Mathur S, Tanwar P, Mishra P, Kataria K, Suhani, Gogia A, Kumar B, Haresh KP, Mallick S, Saini SK, Mishra A, Bansal B, Sharma J, Saikia J, Rangarajan K, Dhamija E, Prasad CP, Shamin SA, Agastm S, Kalra K, Vishvam D, Mani K, Kumar A, Bakhshi S, Batra A. A phase II, randomized, open-labeled study to evaluate low-dose pembrolizumab in addition to neoadjuvant chemotherapy for triple-negative breast cancer (TNBC). Trials 2025; 26:73. [PMID: 40011935 PMCID: PMC11866667 DOI: 10.1186/s13063-025-08726-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/13/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Breast cancer is the most common malignancy diagnosed in women worldwide. Triple-negative breast cancer (TNBC) is the most aggressive subtype, accounting for nearly one third of all breast cancers in India. The addition of pembrolizumab to neoadjuvant chemotherapy improved the pathological response and event free survival in patients with TNBC. However, for most patients in low- and middle-income countries, immunotherapy remains inaccessible due to its high cost. Pharmacological and early clinical data suggest that a lower dose of pembrolizumab may be effective. However, there are no prospective clinical trials in patients with TNBC. METHODS This is a single-site phase II, randomized, open-labeled, parallel-group trial. Eligible patients will be randomized (1:1) to either of the two treatment groups. Patients in the control arm will be administered standard of care chemotherapy [4 cycles of dose-dense doxorubicin (60 mg/m2) and cyclophosphamide (600 mg/m2), followed by 4 cycles of dose-dense paclitaxel (175 mg/m2)]. Patients in the experimental arm will receive 3 doses of pembrolizumab 50 mg every 6 weeks along with neoadjuvant dose-dense chemotherapy. The primary objective of the study is to compare the pathological complete response with the addition of low-dose pembrolizumab to neoadjuvant chemotherapy in patients with TNBC. Secondary objectives include invasive disease-free survival and quality of life assessment. DISCUSSION The PLANeT trial aims to establish the efficacy of low-dose pembrolizumab in addition to neoadjuvant chemotherapy in patients with triple-negative breast cancer patients. This strategy, if found effective, will help improve the outcomes of women with TNBC who currently have limited access to pembrolizumab. TRIAL REGISTRATION Clinical Trials Registry of India-CTRI/2024/01/062088.
Collapse
Affiliation(s)
- Adhip Arora
- Department of Medical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - V Seenu
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rajinder Parshad
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - V K Bansal
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Anita Dhar
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sandeep Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Pranay Tanwar
- Department of Laboratory Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Piyush Mishra
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Kamal Kataria
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Suhani
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ajay Gogia
- Department of Medical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Brijesh Kumar
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - K P Haresh
- Department of Radiation Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Supriya Mallick
- Department of Radiation Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Surendra Kumar Saini
- Department of Radiation Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ashutosh Mishra
- Department of Surgical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Babul Bansal
- Department of Surgical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Jyoti Sharma
- Department of Surgical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Jyoutishman Saikia
- Department of Surgical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Krithika Rangarajan
- Department of Radio-diagnosis, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ekta Dhamija
- Department of Radio-diagnosis, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Chandra Prakash Prasad
- Department of Laboratory Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Shamim Ahmed Shamin
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sourabh Agastm
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Kaushal Kalra
- Department of Medical Oncology, VMMC and Safdarjung Hospital, New Delhi, 110029, India
| | - Divvay Vishvam
- Department of Medical Oncology, VMMC and Safdarjung Hospital, New Delhi, 110029, India
| | - Kalaivani Mani
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Akash Kumar
- Department of Medical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Atul Batra
- Department of Medical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
8
|
Novakova A, Morris SA, Vaiarelli L, Frank S. Manufacturing and Financial Evaluation of Peptide-Based Neoantigen Cancer Vaccines for Triple-Negative Breast Cancer in the United Kingdom: Opportunities and Challenges. Vaccines (Basel) 2025; 13:144. [PMID: 40006691 PMCID: PMC11860436 DOI: 10.3390/vaccines13020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
This review evaluates the financial burden of current treatments for triple-negative breast cancer (TNBC) and projects potential financial scenarios to assess the feasibility of introducing a peptide-based neoantigen cancer vaccine (NCV) targeting the disease, using the UK as a healthcare system model. TNBC, the most aggressive breast cancer subtype, is associated with poor prognosis, worsened by the lack of personalised treatment options. Neoantigen cancer vaccine therapies present a personalised alternative with the potential to enhance T-cell responses independently of genetic factors, unlike approved immunotherapies for TNBC. Through a systematic literature review, the underlying science and manufacturing processes of NCVs are explored, the direct medical costs of existing TNBC treatments are enumerated, and two contrasting pricing scenarios for NCV clinical adoption are evaluated. The findings indicate that limited immunogenicity is the main scientific barrier to NCV clinical advancement, alongside production inefficiencies. Financial analysis shows that the UK spends approximately GBP 230 million annually on TNBC treatments, ranging from GBP 2200 to GBP 54,000 per patient. A best-case pricing model involving government-sponsored NCV therapy appears financially viable, while a worst-case, privately funded model exceeds the National Institute for Health and Care Excellence (NICE) cost thresholds. This study concludes that while NCVs show potential clinical benefits for TNBC, uncertainties about their standalone efficacy make their widespread adoption in the UK unlikely without further clinical research.
Collapse
Affiliation(s)
| | | | - Ludovica Vaiarelli
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK; (A.N.); (S.A.M.)
| | - Stefanie Frank
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK; (A.N.); (S.A.M.)
| |
Collapse
|
9
|
Srivastava TP, Dhar R, Karmakar S. Looking beyond the ER, PR, and HER2: what's new in the ARsenal for combating breast cancer? Reprod Biol Endocrinol 2025; 23:9. [PMID: 39833837 PMCID: PMC11744844 DOI: 10.1186/s12958-024-01338-z] [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: 09/12/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Breast cancer (BrCa) is a complex and heterogeneous disease with diverse molecular subtypes, leading to varied clinical outcomes and posing significant treatment challenges. The increasing global burden of BrCa, particularly in low- and middle-income countries, underscores the urgent need for more effective therapeutic strategies. The androgen receptor (AR), expressed in a substantial proportion of breast cancer cases, has emerged as a potential biomarker and therapeutic target. In breast cancer, AR exhibits diverse functions across subtypes, often interacting with other hormone receptors, thereby influencing tumor progression and treatment responses. This intricate interplay is further complicated by the presence of constitutively expressed AR splice variants (AR-Vs) that drive resistance to AR-targeting therapies through structural rearrangements in the domains and activation of aberrant signaling pathways. Although AR-targeting drugs, initially developed for prostate cancer (PCa), have shown promise in AR-positive breast cancer, significant gaps remain in understanding AR's precise functions and therapeutic potential. The systemic management of breast cancer is guided primarily by theranostic biomarkers; ER, PR, HER2, and Ki67 which also dictate the breast cancer classification. The ubiquitous expression of AR in BrCa and the emergence of AR-Vs can assist the management of disease complementing the standard of care. This article provides a comprehensive overview of AR and its splice variants in the context of breast cancer, highlighting their prognostic and predictive value across different subtypes looking beyond the conventional ER, PR, and HER2 status. This review also raises the possibility of using AR splice variants in predicting tumor aggressiveness. From the settings of developing nations, this may provide useful insight by integrating recent advances in AR-targeted therapies and exploring their translational potential, emphasizing the critical need for further research to optimize AR-based therapeutic strategies for breast cancer management.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/therapy
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- Female
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
Collapse
Affiliation(s)
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
10
|
Ma T, Hao XM, Chen HD, Zheng MH, Chen XG, Cai SL, Zhang J. Predictive markers of rapid disease progression and chemotherapy resistance in triple-negative breast cancer patients following postoperative adjuvant therapy. Sci Rep 2025; 15:386. [PMID: 39748017 PMCID: PMC11697311 DOI: 10.1038/s41598-024-84785-3] [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: 09/30/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a diverse category with a subset that displays particularly aggressive characteristics, referred to in this study as "rapid relapse" TNBC (rrTNBC). This term is defined as the occurrence of distant metastasis or death within 24 months post-diagnosis. The paper mainly studies the clinicopathologic traits of TNBC patients experiencing rapid disease progression and chemotherapy resistance and identify predictive markers for this outcome. A retrospective evaluation was conducted on 2294 TNBC patients who underwent surgery at Tianjin Medical University Cancer Hospital. Of these, 369 were categorized as experiencing rapid relapse, while 1925 did not relapse rapidly. Logistic regression analysis was applied to determine potential markers predictive of rapid relapse post-chemotherapy. Both univariate and multivariate logistic regression analyses pinpointed several predictors of rapid relapse in TNBC patients post-chemotherapy. These include age at diagnosis (≥ 50 years, OR = 0.413, 95% CI: 0.289-0.590), postoperative pathological T staging (T2, OR = 2.557, 95% CI: 1.766-3.703; T3 + T4, OR = 3.725, 95% CI: 1.355-10.454), and N staging (N1, OR = 3.056, 95% CI: 2.021-4.619; N2, OR = 6.917, 95% CI: 3.920-12.206; N3, OR = 24.597, 95% CI: 11.875-50.948). Additionally, sTIL expression (intermediate, OR = 0.204, 95% CI: 0.139-0.300; high, OR = 0.020, 95% CI: 0.011-0.035) and Her2 expression (Her2 1+, OR = 0.470, 95% CI: 0.321-0.688) were identified as protective indicators against rapid relapse. A predictive model incorporating these predictors yielded a C-index of 0.898 in the training set and 0.938 in the validation set, with respective Brier scores of 0.079 and 0.073. The study successfully established and validated a predictive model for rapid disease progression and chemotherapy resistance in TNBC patients post-chemotherapy, demonstrating robust discrimination and accuracy.
Collapse
Affiliation(s)
- Tao Ma
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiao-Meng Hao
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Hong-Dan Chen
- First Department of Cadre Clinic, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Min-Hui Zheng
- Oral and Maxillofacial Surgery Thyroid and Hernia Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Xiao-Geng Chen
- Department of Breast Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Shuang-Long Cai
- Department of Breast Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China.
| | - Jin Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
| |
Collapse
|
11
|
Ali BM, El-Abhar HS, Mohamed G, Nassar HR, Aliedin N, Sharaky M, Shouman SA, Kamel M. A study of the role of androgen receptor and androgen receptor variant 7 in TNBC patients and the effect of their targeting by Enzalutamide and EPI-001 in MDA-MB-231. J Steroid Biochem Mol Biol 2025; 245:106636. [PMID: 39536950 DOI: 10.1016/j.jsbmb.2024.106636] [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: 08/20/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Abstract
The lack of targeted therapy for triple-negative breast cancer (TNBC) is among the mainsprings of its poor prognosis. This study aimed to elucidate the role of the androgen receptor (AR) and its splice variant 7 (ARv7) in TNBC patients. Further, the molecular impact of their blockers, Enzalutamide and EPI-001, on the TNBC cell line MDA-MB-231 was investigated. Thereby, immunohistochemical expression of AR/ARv7 was assessed for TNBC Egyptian patients. Moreover, bioinformatics analysis of AR/ARv7 RNA status was carried out on TNBC patients from The Cancer Genome Atlas Breast Carcinoma project (TCGA-BRCA). Data from both groups was correlated with patients' clinicopathological features. Besides, scratch wound healing assay and ELISA were employed to assess the effect of AR/ARv7 blockers on several metastasis markers in MDA-MB-231 cell line. In the Egyptian-TNBC patients, AR expression was associated with worse 7-year DFS (40.6 ± 18.6 %). In addition, ARv7 showed cytoplasmic and nuclear patterns, and both cytoplasmic and nuclear ARv7+ patients demonstrated a worse 7-year DFS (22.7 ± 17.7 % and 20 ± 17.9 %) and overall survival (63.6 ± 14.5 % and 40 ± 21.8 %). Importantly, 80 % of the nuclear ARv7+ patients developed distant metastasis. The data of the TCGA-TNBC patients showed a tendency for poor outcomes in the high ARv7-expressing patients. Molecularly, in MDA-MB-231, both inhibitors modulated metastasis and epithelial to mesenchymal transition (EMT) markers ROCK1, ROCK2, c-Myc, E-cadherin and N-cadherin, with EPI-001 downregulating NF-ĸB level as well. We concluded that ARv7 indicated poor prognosis in the studied cohorts and that blocking of AR/ARv7 abated metastasis and key regulators of EMT in MDA-MB-231, at least in part by targeting ROCK/NF-ĸB/c-Myc axis.
Collapse
Affiliation(s)
- Belal M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Ministry of Health and Population, Egypt.
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt (FUE), Cairo, Egypt.
| | - Ghada Mohamed
- Department of Pathology, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Hanan R Nassar
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Nelly Aliedin
- Department of Medical Statistics Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Marwa Sharaky
- Department of Cancer Biology, Unit of Pharmacology and Experimental Therapeutics, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Samia A Shouman
- Department of Cancer Biology, Unit of Pharmacology and Experimental Therapeutics, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Marwa Kamel
- Department of Cancer Biology, Unit of Pharmacology and Experimental Therapeutics, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
Collapse
|
12
|
Chaudhary A, Patil P, Raina P, Kaul-Ghanekar R. Matairesinol repolarizes M2 macrophages to M1 phenotype to induce apoptosis in triple-negative breast cancer cells. Immunopharmacol Immunotoxicol 2024:1-15. [PMID: 39722605 DOI: 10.1080/08923973.2024.2425028] [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: 06/05/2024] [Accepted: 10/27/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Triple-Negative Breast Cancer (TNBC), the most challenging subtype of Breast Cancer (BC), currently lacks targeted therapy, presenting a significant therapeutic gap in its management. Tumor Associated Macrophages (TAMs) play a significant role in TNBC progression and could be targeted by repolarizing them from M2 to M1 phenotype. Matairesinol (MAT), a plant lignan, has been shown to exhibit anticancer, anti-inflammatory and immunomodulatory activities. In this study, we explored how MAT-induced repolarization of THP-1-derived M2 macrophages towards the M1 phenotype, which could effectively target the TNBC cell line, MDA-MB-231. METHODS The differential expression of genes in THP-1-derived macrophages at mRNA levels was evaluated by RNAseq assay. An inverted microscope equipped with a CMOS camera was utilized to capture the morphological variations in THP-1 cells and THP-1-derived macrophages. Relative mRNA expression of M1 and M2 specific marker genes was quantified by qRT-PCR. Cell viability and induction of apoptosis were evaluated by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1 dye) assays, respectively. RESULTS MAT reduced the viability of M2a and M2d macrophages and repolarized them to M1 phenotype. Conditioned medium (CM) from MAT-treated M2a and M2d macrophages significantly reduced the viability of TNBC cells by apoptosis. CONCLUSION Targeting M2 macrophages is an important strategy to regulate cancer progression. Our study provides evidence that MAT may be a promising drug candidate for developing novel anti-TNBC therapy. However, further studies are warranted to thoroughly elucidate the molecular mechanism of action of MAT and evaluate its therapeutic potential in TNBC in vitro and in vivo models.
Collapse
Affiliation(s)
- Amol Chaudhary
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Prajakta Patil
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Prerna Raina
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Analytical Department (ADT), Lupin Limited, Pune, India
| | - Ruchika Kaul-Ghanekar
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Symbiosis Centre for Research and Innovation (SCRI); Symbiosis International Deemed University (SIU), Pune, India
- Cancer Research Lab, Symbiosis School of Biological Sciences (SSBS), Symbiosis International Deemed University (SIU), Pune, India
| |
Collapse
|
13
|
Zhang JN, Zhang Z, Huang ZL, Guo Q, Wu ZQ, Ke C, Lu B, Wang ZT, Ji LL. Isotoosendanin inhibits triple-negative breast cancer metastasis by reducing mitochondrial fission and lamellipodia formation regulated by the Smad2/3-GOT2-MYH9 signaling axis. Acta Pharmacol Sin 2024; 45:2672-2683. [PMID: 39009651 PMCID: PMC11579498 DOI: 10.1038/s41401-024-01335-3] [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: 01/01/2024] [Accepted: 06/04/2024] [Indexed: 07/17/2024]
Abstract
Triple-negative breast cancer (TNBC) is incurable and prone to widespread metastasis. Therefore, identification of key targets for TNBC progression is urgently needed. Our previous study revealed that isotoosendanin (ITSN) reduced TNBC metastasis by targeting TGFβR1. ITSN is currently used as an effective chemical probe to further discover the key molecules involved in TNBC metastasis downstream of TGFβR1. The results showed that GOT2 was the gene downstream of Smad2/3 and that ITSN decreased GOT2 expression by abrogating the activation of the TGF-β-Smad2/3 signaling pathway through directly binding to TGFβR1. GOT2 was highly expressed in TNBC, and its knockdown decreased TNBC metastasis. However, GOT2 overexpression reversed the inhibitory effect of ITSN on TNBC metastasis both in vitro and in vivo. GOT2 interacted with MYH9 and hindered its binding to the E3 ubiquitin ligase STUB1, thereby reducing MYH9 ubiquitination and degradation. Moreover, GOT2 also enhanced the translocation of MYH9 to mitochondria and thus induced DRP1 phosphorylation, thereby promoting mitochondrial fission and lamellipodia formation in TNBC cells. ITSN-mediated inhibition of mitochondrial fission and lamellipodia formation was associated with reduced GOT2 expression. In conclusion, ITSN prevented MYH9-regulated mitochondrial fission and lamellipodia formation in TNBC cells by enhancing MYH9 protein degradation through a reduction in GOT2 expression, thus contributing to its inhibition of TNBC metastasis.
Collapse
Affiliation(s)
- Jing-Nan Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, China
| | - Ze Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhen-Lin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qian Guo
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ze-Qi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chuang Ke
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zheng-Tao Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li-Li Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
14
|
Alajroush DR, Anderson BF, Bruce JA, Lartey CI, Mathurin DA, Washington ST, Washington TS, Diawara S, Waheed SA, Thomas KL, Beebe SJ, Holder AA. Enhancement of antitumor effects of berberine chloride with a copper(II) complex against human triple negative breast cancer: In vitro studies. RESULTS IN CHEMISTRY 2024; 12:101882. [PMID: 39802841 PMCID: PMC11720871 DOI: 10.1016/j.rechem.2024.101882] [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] [Indexed: 01/16/2025] Open
Abstract
In this study, the copper(II) complex [Cu(chromoneTSC)Cl2]•0.5H2O•0.0625C2H5OH (where chromoneTSC = (E)-N-Ethyl-2-((4-oxo-4H-chromen-3-yl)methylene)-hydrazinecarbothioamide) was synthesized and characterized; then used to carry out in vitro studies in combination with berberine chloride (BBC). The ligand and complex were characterized by elemental analysis, FTIR and NMR (1H and 13C) spectroscopy, and conductivity measurements. The cytotoxic effect was analyzed by using the CCK-8 viability assay in cancer MDA-MB-231 VIM RFP and non-cancer MCF-10A cell lines. The IC50 values for the complex and BBC were 21.2 ±1.6 and 48.3 ± 2.4 μM, respectively at 24 h incubation, while the IC50 value of the combination treatment was 9.3 ± 1.5 in cancer cells. The co-treatment group significantly increased the number of cells in G2 phase, indicating the growth arrest of cancer cells. Moreover, the combination group showed induction of both intrinsic and extrinsic apoptotic pathways. There was also a study on the effect of the combination treatment on receptor-interacting serine/threonine-protein kinase 3 (RIPK3) and mixed lineage kinase domain-like pseudokinase (MLKL) as biomarkers of necroptosis. The results showed activation of necroptosis after treatment with the combination of the copper complex and BBC via the activation of RIPK3-MLKL pathway.
Collapse
Affiliation(s)
- Duaa R. Alajroush
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, USA
| | - Brittney F. Anderson
- Department of Biological Sciences, University of the Virgin Islands, 2 John Brewers Bay, St. Thomas, VI 00802, USA
| | - Janae A. Bruce
- Department of Biological Sciences, University of the Virgin Islands, 2 John Brewers Bay, St. Thomas, VI 00802, USA
| | - Christian I. Lartey
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, USA
| | - Dazonte A. Mathurin
- Department of Biological Sciences, University of the Virgin Islands, 2 John Brewers Bay, St. Thomas, VI 00802, USA
| | - Sean T. Washington
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, USA
| | - Tanaya S. Washington
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, USA
| | - Sidy Diawara
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, USA
| | - Sakariyau A. Waheed
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, USA
| | - Kaylin L. Thomas
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, USA
| | - Stephen J. Beebe
- Frank Reidy Research center for Bioelectrics, Old Dominion University, 4211 Monarch Way, Suite 300, Norfolk, VA, 23508, USA
| | - Alvin A. Holder
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, USA
| |
Collapse
|
15
|
Zuo X, Zhao X, Zhang X, Li Q, Jiang X, Huang S, Chen X, Chen X, Jia W, Zou H, Shi D, Qian X. PTPN20 promotes metastasis through activating NF-κB signaling in triple-negative breast cancer. Breast Cancer Res 2024; 26:155. [PMID: 39506852 PMCID: PMC11542355 DOI: 10.1186/s13058-024-01910-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: 07/22/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Cancer metastasis remains a major challenge in the clinical management of triple-negative breast cancer (TNBC). The NF-κB signaling pathway has been implicated as a crucial factor in the development of metastases, but the underlying molecular mechanisms remain largely unclear. METHODS PTPN20 expression was evaluated using data from the Sweden Cancerome Analysis Network-Breast and The Cancer Genome Atlas database, as well as by western blotting and immunohistochemistry in 88 TNBC patients. The ability of PTPN20 to activate NF-κB was assessed by luciferase reporter assays. The effects of PTPN20 overexpression and knockdown via short hairpin RNA were examined in TNBC cell lines by wound healing and transwell matrix penetration assays. Additionally, we analyzed the growth and metastasis abilitiy of 4T1 xenograft tumors in nude mice. RESULTS PTPN20 levels were elevated in TNBC cell lines and patient samples compared to controls, and higher protein levels correlated with metastasis-free survival. Overexpression of PTPN20 enhanced migration and invasion in vitro, and promoted lung metastasis in vivo. Our finding revealed that PTPN20 activates NF-κB signaling by dephosphorylating p65 at Ser468, preventing its binding to COMMD1, thereby protecting p65 from degradation. Downregulation of PTPN20 effectively inhibit, while p65 S468A mutant restored the migratory and invasive abilities of TNBC cells. CONCLUSIONS Collectively, our results demonstrate that PTPN20 plays a critical role in TNBC metastasis through the activation of NF-κB signaling. We propose that PTPN20 may serve as a novel prognostic marker and potential therapeutic target for the treatment of TNBC.
Collapse
Affiliation(s)
- Xiaoxiao Zuo
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xiaohan Zhao
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Xiaofei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Qingyuan Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Xingyu Jiang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Shumei Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xuwei Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Xiangfu Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Weihua Jia
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
- Department of Biobank, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Hequn Zou
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Dongni Shi
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China.
| | - Xueke Qian
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
16
|
Miracle CE, McCallister CL, Denning KL, Russell R, Allen J, Lawrence L, Legenza M, Krutzler-Berry D, Salisbury TB. High BMI Is Associated with Changes in Peritumor Breast Adipose Tissue That Increase the Invasive Activity of Triple-Negative Breast Cancer Cells. Int J Mol Sci 2024; 25:10592. [PMID: 39408921 PMCID: PMC11476838 DOI: 10.3390/ijms251910592] [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: 08/28/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Breast cancer is the most common cancer in women with multiple risk factors including smoking, genetics, environmental factors, and obesity. Smoking and obesity are the top two risk factors for the development of breast cancer. The effect of obesity on adipose tissue mediates the pathogenesis of breast cancer in the context of obesity. Triple-negative breast cancer (TNBC) is a breast cancer subtype within which the cells lack estrogen, progesterone, and HER2 receptors. TNBC is the deadliest breast cancer subtype. The 5-year survival rates for patients with TNBC are 8-16% lower than the 5-year survival rates for patients with estrogen-receptor-positive breast tumors. In addition, TNBC patients have early relapse rates (3-5 years after diagnosis). Obesity is associated with an increased risk for TNBC, larger TNBC tumors, and increased breast cancer metastasis compared with lean women. Thus, novel therapeutic approaches are warranted to treat TNBC in the context of obesity. In this paper, we show that peritumor breast adipose-derived secretome (ADS) from patients with a high (>30) BMI is a stronger inducer of TNBC cell invasiveness and JAG1 expression than peritumor breast ADS from patients with low (<30) BMI. These findings indicate that patient BMI-associated changes in peritumor AT induce changes in peritumor ADS, which in turn acts on TNBC cells to stimulate JAG1 expression and cancer cell invasiveness.
Collapse
Affiliation(s)
- Cora E. Miracle
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (C.E.M.); (C.L.M.)
| | - Chelsea L. McCallister
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (C.E.M.); (C.L.M.)
| | - Krista L. Denning
- Cabell Huntington Hospital Laboratory, Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (K.L.D.); (R.R.); (J.A.); (L.L.)
| | - Rebecca Russell
- Cabell Huntington Hospital Laboratory, Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (K.L.D.); (R.R.); (J.A.); (L.L.)
| | - Jennifer Allen
- Cabell Huntington Hospital Laboratory, Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (K.L.D.); (R.R.); (J.A.); (L.L.)
| | - Logan Lawrence
- Cabell Huntington Hospital Laboratory, Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (K.L.D.); (R.R.); (J.A.); (L.L.)
| | - Mary Legenza
- Edwards Comprehensive Cancer Center, Department of Oncology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (M.L.); (D.K.-B.)
| | - Diane Krutzler-Berry
- Edwards Comprehensive Cancer Center, Department of Oncology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (M.L.); (D.K.-B.)
| | - Travis B. Salisbury
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (C.E.M.); (C.L.M.)
| |
Collapse
|
17
|
Fu Y, Sun X, He Q, Gu Z, Jia X, Zhuang Z. Connexin 43 controls metastatic behavior in triple negative breast cancer through TGFβ1-Smad3-intergin αV signaling axis Based on optical image diagnosis. SLAS Technol 2024; 29:100190. [PMID: 39299505 DOI: 10.1016/j.slast.2024.100190] [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: 04/09/2024] [Revised: 07/19/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
Abnormal expression of connexin 43 (Cx43) contributes to the development and progression of cancer. However, its regulation is complex and dependent on the environment. The expression of Cx43 in triple-negative cancer lesions was analyzed by immunohistochemistry and optical coherence tomography using experimental models and clinical samples. The model of TGFβ1-SMad3-in-αv signal axis was established and verified by experiments. The results show that Cx43 plays a key role in the regulation of triple-negative cancer metastasis. In vivo, over-expressed Cx43 decreased tumor volume and inhibited ITGαV, TGF-β1, Smad3 and N-cadherin expressions, but enhanced the E-cadherin. Cx43 had the lowest expression in the TNBC samples, especially in lymph node metastatic TNBC patients and had a negative correlation with ITG alpha V, TGF-β1 and Smad3.The study demonstrated Cx43 controlled metastatic behavior through TGF-β1 -Smad3-ITG αV signaling axis in MDA-MB-231 cells, providing evidence for Cx43's function in TNBC. The optical image diagnosis method can realize the identification and quantitative evaluation of early cancer triple negative, and provide a new strategy and means for the treatment of cancer triple negative.
Collapse
Affiliation(s)
- Yun Fu
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, PR China
| | - Xiaoyin Sun
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, PR China
| | - Qizhi He
- Department of Pathology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, PR China
| | - Zhangyuan Gu
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, PR China
| | - Xiaoqing Jia
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, PR China
| | - Zhigang Zhuang
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, PR China.
| |
Collapse
|
18
|
Falahi F, Akbari-Birgani S, Mortazavi Y, Johari B. Caspase-9 suppresses metastatic behavior of MDA-MB-231 cells in an adaptive organoid model. Sci Rep 2024; 14:15116. [PMID: 38956424 PMCID: PMC11219723 DOI: 10.1038/s41598-024-65711-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: 03/18/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
Caspase-9, a cysteine-aspartate protease traditionally associated with intrinsic apoptosis, has recently emerged as having non-apoptotic roles, including influencing cell migration-an aspect that has received limited attention in existing studies. In our investigation, we aimed to explore the impact of caspase-9 on the migration and invasion behaviors of MDA-MB-231, a triple-negative breast cancer (TNBC) cell line known for its metastatic properties. We established a stable cell line expressing an inducible caspase-9 (iC9) in MDA-MB-231 and assessed their metastatic behavior using both monolayer and the 3D organotypic model in co-culture with human Foreskin fibroblasts (HFF). Our findings revealed that caspase-9 had an inhibitory effect on migration and invasion in both models. In monolayer culture, caspase-9 effectively suppressed the migration and invasion of MDA-MB-231 cells, comparable to the anti-metastatic agent panitumumab (Pan). Notably, the combination of caspase-9 and Pan exhibited a significant additional effect in reducing metastatic behavior. Interestingly, caspase-9 demonstrated superior efficacy compared to Pan in the organotypic model. Molecular analysis showed down regulation of epithelial-mesenchymal transition and migratory markers, in caspase-9 activated cells. Additionally, flow cytometry analysis indicated a cell cycle arrest. Moreover, pre-treatment with activated caspase-9 sensitized cells to the chemotherapy of doxorubicin, thereby enhancing its effectiveness. In conclusion, the anti-metastatic potential of caspase-9 presents avenues for the development of novel therapeutic approaches for TNBC/metastatic breast cancer. Although more studies need to figure out the exact involving mechanisms behind this behavior.
Collapse
Affiliation(s)
- Farzaneh Falahi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 45137-66731, Iran
| | - Shiva Akbari-Birgani
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 45137-66731, Iran.
- Research Center for Basic Sciences and Modern Technologies (RBST), Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 45137-66731, Iran.
| | - Yousef Mortazavi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Behrooz Johari
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
19
|
Kimura T, Takami T, Piao Y, Ntalla I, Saji S. Treatment patterns and clinical outcomes in patients with metastatic triple-negative breast cancer: a large-scale data analysis using the Japanese claims database. Breast Cancer Res Treat 2024; 206:91-103. [PMID: 38704772 PMCID: PMC11182808 DOI: 10.1007/s10549-024-07273-2] [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] [Accepted: 01/25/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE This study evaluated treatment patterns and clinical outcomes among patients with metastatic triple-negative breast cancer (mTNBC) in real-world clinical settings in Japan. METHODS The treatment patterns, time to next treatment or death (TTNTD), time to treatment discontinuation, adverse events of interest, and medical costs of treating patients with mTNBC in first-, second-, and third-line settings were investigated using data of patients meeting the inclusion criteria between January 2017 and March 2022 in a Japanese medical claims database. The treatment regimens for mTNBC were defined according to the Japanese Breast Cancer Society Clinical Practice Guidelines. RESULTS In this study, 2236 patients with mTNBC (median age 66.0 years; 99.8% female) were included in the first-line cohort. Of these, 46.6% and 20.8% were included in the second- and third-line cohorts, respectively. The two most frequently used treatments were capecitabine (19.1%) and S-1 (tegafur-gimeracil-oteracil) (14.5%) in the first-line cohort, eribulin (18.3%) and bevacizumab/paclitaxel (14.4%) in the second-line cohort, and eribulin (19.4%) and bevacizumab/paclitaxel (17.5%) in the third-line cohort. The TTNTD shortened as the line of therapy progressed (median 8.0, 6.5, and 5.2 months for the first-, second-, and third-line treatments, respectively). Nausea/vomiting and neutropenia/leukopenia occurred in 62.8% and 18.3% of all patients, respectively. The medical total costs per day were 6.7, 10.2, and 12.9 thousand yen during the first-/second-/third-line treatments, respectively. CONCLUSION This study provides insight into current treatment patterns for mTNBC in Japan. The cost-benefit balance worsens with later-line treatment and a high unmet need for mTNBC drug treatment remains.
Collapse
Affiliation(s)
| | | | - Yi Piao
- Medical Affairs, Gilead Sciences K.K., Tokyo, Japan
| | - Ioanna Ntalla
- Real-World Evidence, Gilead Sciences Europe Ltd., Stockley Park, Uxbridge, UK
| | - Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
20
|
Oey O, Wijaya W, Redfern A. Eribulin in breast cancer: Current insights and therapeutic perspectives. World J Exp Med 2024; 14:92558. [PMID: 38948420 PMCID: PMC11212747 DOI: 10.5493/wjem.v14.i2.92558] [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: 01/29/2024] [Revised: 02/21/2024] [Accepted: 03/20/2024] [Indexed: 06/19/2024] Open
Abstract
Eribulin is a non-taxane synthetic analogue approved in many countries as third-line treatment for the treatment of patients with metastatic breast cancer. In addition to its mitotic property, eribulin has non-mitotic properties including but not limited to, its ability to induce phenotypic reversal of epithelial to mesenchymal transition, vascular remodelling, reduction in immunosuppressive tumour microenvironment. Since approval, there has been a surge in studies investigating the application of eribulin as an earlier-line treatment and also in combination with other agents such as immunotherapy and targeted therapy across all breast cancer sub-types, including hormone receptor positive, HER2 positive and triple negative breast cancer, many demonstrating promising activity. This review will focus on the application of eribulin in the treatment of metastatic breast cancer across all subtypes including its role as an earlier-line agent, its toxicity profile, and potential future directions.
Collapse
Affiliation(s)
- Oliver Oey
- Faculty of Medicine, University of Western Australia, Nedlands 6009, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands 6009, WA, Australia
| | - Wynne Wijaya
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
- Department of Internal Medicine, Universitas Gadjah Mada, Sleman 55281, Indonesia
| | - Andrew Redfern
- Department of Medical Oncology, Fiona Stanley Hospital, Murdoch 6150, WA, Australia
| |
Collapse
|
21
|
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
|
22
|
Mishra A, Mishra SK, Sharanappa V, Krishnani N, Kumari N, Agarwal G. Incidence and Prognostic Significance of Androgen Receptors (AR) in Indian Triple-Negative Breast Cancer (TNBC). Indian J Surg Oncol 2024; 15:250-257. [PMID: 38741650 PMCID: PMC11088609 DOI: 10.1007/s13193-024-01877-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 01/07/2024] [Indexed: 05/16/2024] Open
Abstract
Molecular sub-characterization of triple-negative breast cancer (TNBC) has great therapeutic and possibly prognostic implications. The primary aim of this study was to investigate the incidence of luminal androgen receptor (LAR) subtype of TNBC and secondary aims were sub-categorization and clinico-pathologic correlation of LAR breast cancers. Retrospective study (January 2008 and 31st of December 2018) consisting of 157 TNBC patients. Androgen receptor (AR) expression was measured by immunohistochemical analysis. One percent cutoff was set as a positive expression. Sub-categorization was done on the basis of EGFR (> 15% of tumor cells) and Ki-67 expression (low- < 11%, intermediate- 11-20%, and high- > 21%). AR expression was correlated with various clinico-pathologic features and outcomes of the patients. The incidence of AR expression in TNBC was 24.8%. Considering different thresholds of > 5%, > 10%, and > 20% immunostaining, the incidence of AR positivity was 18.4, 15.2, and 11.5% respectively. The incidence of Ki-67 (p = 0.89) and EGFR (p = 0.643) expression did not differ significantly in AR-positive and -negative TNBC. Based on EGFR expression 19, 67 and 14% patients were categorized as low, intermediate, and high risk respectively. Low-risk (p ≤ 0.001) and low-grade (p = 0.014) tumors were more likely to have > 10% AR expression. Clinico-pathological profile, response to neoadjuvant chemotherapy, disease-free survival (p = 0.458), and overall survival (p = 0.806) did not significantly differ between AR expressing and negative TNBC. On multivariate analysis, only tumor staging was a significant predictor of survival (p = 0.012) and AR expression of > 10% revealed a trend towards improved survival (p = 0.07). When considering only AR-positive TNBC, AR expression of > 10% (p = 0.038), distant metastases (p = 0.003), and EGFR status (p = 0.024) were significantly associated with survival. AR expression does not seem to very strongly correlate with prognosis in TNBC and further studies could focus more on its predictive role in deciding anti-androgen therapy.
Collapse
Affiliation(s)
- Anjali Mishra
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Shravan Kumar Mishra
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Vikram Sharanappa
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Narendra Krishnani
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Niraj Kumari
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Gaurav Agarwal
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| |
Collapse
|
23
|
Kesireddy M, Elsayed L, Shostrom VK, Agarwal P, Asif S, Yellala A, Krishnamurthy J. Overall Survival and Prognostic Factors in Metastatic Triple-Negative Breast Cancer: A National Cancer Database Analysis. Cancers (Basel) 2024; 16:1791. [PMID: 38791870 PMCID: PMC11120599 DOI: 10.3390/cancers16101791] [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: 04/10/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Metastatic triple-negative breast cancer (TNBC) is aggressive with poor median overall survival (OS) ranging from 8 to 13 months. There exists considerable heterogeneity in survival at the individual patient level. To better understand the survival heterogeneity and improve risk stratification, our study aims to identify the factors influencing survival, utilizing a large patient sample from the National Cancer Database (NCDB). METHODS Women diagnosed with metastatic TNBC from 2010 to 2020 in the NCDB were included. Demographic, clinicopathological, and treatment data and overall survival (OS) outcomes were collected. Kaplan-Meier curves were used to estimate OS. The log-rank test was used to identify OS differences between groups for each variable in the univariate analysis. For the multivariate analysis, the Cox proportional hazard model with backward elimination was used to identify factors affecting OS. Adjusted hazard ratios and 95% confidence intervals are presented. RESULTS In this sample, 2273 women had a median overall survival of 13.6 months. Factors associated with statistically significantly worse OS included older age, higher comorbidity scores, specific histologies, higher number of metastatic sites, presence of liver or other site metastases in those with only one metastatic site (excluding brain metastases), presence of cranial and extra-cranial metastases, lack of chemotherapy, lack of immunotherapy, lack of surgery to distant sites, lack of radiation to distant sites, and receipt of palliative treatment to alleviate symptoms. In the multivariate analysis, comorbidity score, histology, number of metastatic sites, immunotherapy, and chemotherapy had a statistically significant effect on OS. CONCLUSIONS Through NCDB analysis, we have identified prognostic factors for metastatic TNBC. These findings will help individualize prognostication at diagnosis, optimize treatment strategies, and facilitate patient stratification in future clinical trials.
Collapse
Affiliation(s)
- Meghana Kesireddy
- Division of Hematology and Medical Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA (J.K.)
| | - Lina Elsayed
- Division of Hematology and Medical Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA (J.K.)
| | - Valerie K. Shostrom
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Priyal Agarwal
- Division of Hematology and Medical Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA (J.K.)
| | - Samia Asif
- Division of Hematology and Medical Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA (J.K.)
| | - Amulya Yellala
- Division of Hematology and Medical Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA (J.K.)
| | - Jairam Krishnamurthy
- Division of Hematology and Medical Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA (J.K.)
| |
Collapse
|
24
|
Calaf GM, Crispin LA, Quisbert-Valenzuela EO. Noscapine and Apoptosis in Breast and Other Cancers. Int J Mol Sci 2024; 25:3536. [PMID: 38542508 PMCID: PMC10970989 DOI: 10.3390/ijms25063536] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/13/2024] [Accepted: 03/16/2024] [Indexed: 06/15/2024] Open
Abstract
Breast cancer is the second leading contributor to the age-standardized mortality rate, for both sexes and all ages worldwide. In Europe and the United States, it is the second leading cause of mortality, with an incidence rate of about 2.6 million cases per year. Noscapine, a well-known alkaloid used as a cough suppressant, demonstrated anti-tumor effects by triggering apoptosis in various cancer cell lines and has the potential to become another ally against breast, ovarian, colon, and gastric cancer, among other types of malignancy. Apoptosis plays a crucial role in the treatment of cancer. Noscapine affected BAX, CASP8, CASP9, NFKBIA, and RELA gene and protein expression in the MCF-7 and MDA-MB-231 cell lines. Gene expression was higher in tumor than in normal tissue, including the BAX expression levels in lung, ovary, endometrium, colon, stomach, and glioblastoma patients; BCL2L1 expression in endometrium, colon, and stomach patients; CASP8 gene expression levels in lung, endometrium, colon, stomach, and glioblastoma patients; RELA in colon, stomach, and glioblastoma patients; and NFKBIA in glioblastoma patients. It can be concluded that noscapine affected genes and proteins related to apoptosis in cancer cell lines and several types of cancer patients.
Collapse
Affiliation(s)
- Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.)
| | | | | |
Collapse
|
25
|
Alajroush DR, Smith CB, Anderson BF, Oyeyemi IT, Beebe SJ, Holder AA. A Comparison of In Vitro Studies between Cobalt(III) and Copper(II) Complexes with Thiosemicarbazone Ligands to Treat Triple Negative Breast Cancer. Inorganica Chim Acta 2024; 562:121898. [PMID: 38282819 PMCID: PMC10810091 DOI: 10.1016/j.ica.2023.121898] [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] [Indexed: 01/30/2024]
Abstract
Metal complexes have gained significant attention as potential anti-cancer agents. The anti-cancer activity of [Co(phen)2(MeATSC)](NO3)3•1.5H2O•C2H5OH 1 (where phen = 1,10-phenanthroline and MeATSC = 9-anthraldehyde-N(4)-methylthiosemicarbazone) and [Cu(acetylethTSC)Cl]Cl•0.25C2H5OH 2 (where acetylethTSC = (E)-N-ethyl-2-[1-(thiazol-2-yl)ethylidene]hydrazinecarbothioamide) was investigated by analyzing DNA cleavage activity. The cytotoxic effect was analyzed using CCK-8 viability assay. The activities of caspase 3/7, 9, and 1, reactive oxygen species (ROS) production, cell cycle arrest, and mitochondrial function were further analyzed to study the cell death mechanisms. Complex 2 induced a significant increase in nicked DNA. The IC50 values of complex 1 were 17.59 μM and 61.26 μM in cancer and non-cancer cells, respectively. The IC50 values of complex 2 were 5.63 and 12.19 μM for cancer and non-cancer cells, respectively. Complex 1 induced an increase in ROS levels, mitochondrial dysfunction, and activated caspases 3/7, 9, and 1, which indicated the induction of intrinsic apoptotic pathway and pyroptosis. Complex 2 induced cell cycle arrest in the S phase, ROS generation, and caspase 3/7 activation. Thus, complex 1 induced cell death in the breast cancer cell line via activation of oxidative stress which induced apoptosis and pyroptosis while complex 2 induced cell cycle arrest through the induction of DNA cleavage.
Collapse
Affiliation(s)
- Duaa R. Alajroush
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
| | - Chloe B. Smith
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
| | - Brittney F. Anderson
- Department of Biological Sciences, University of the Virgin Islands, 2 John Brewers Bay, St. Thomas, VI 00802, U.S.A
| | - Ifeoluwa T. Oyeyemi
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
- Department of Biological Sciences, University of Medical Sciences, Ondo City, Nigeria
| | - Stephen J. Beebe
- Frank Reidy Research center for Bioelectrics, Old Dominion University, 4211 Monarch Way, Suite 300, Norfolk, VA, 23508, U.S.A
| | - Alvin A. Holder
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
| |
Collapse
|
26
|
Fan S, Kang B, Li S, Li W, Chen C, Chen J, Deng L, Chen D, Zhou J. Exploring the multifaceted role of RASGRP1 in disease: immune, neural, metabolic, and oncogenic perspectives. Cell Cycle 2024; 23:722-746. [PMID: 38865342 PMCID: PMC11229727 DOI: 10.1080/15384101.2024.2366009] [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: 05/02/2023] [Accepted: 11/25/2023] [Indexed: 06/14/2024] Open
Abstract
RAS guanyl releasing protein 1 (RASGRP1) is a guanine nucleotide exchange factor (GEF) characterized by the presence of a RAS superfamily GEF domain. It functions as a diacylglycerol (DAG)-regulated nucleotide exchange factor, specifically activating RAS through the exchange of bound GDP for GTP. Activation of RAS by RASGRP1 has a wide range of downstream effects at the cellular level. Thus, it is not surprising that many diseases are associated with RASGRP1 disorders. Here, we present an overview of the structure and function of RASGRP1, its crucial role in the development, expression, and regulation of immune cells, and its involvement in various signaling pathways. This review comprehensively explores the relationship between RASGRP1 and various diseases, elucidates the underlying molecular mechanisms of RASGRP1 in each disease, and identifies potential therapeutic targets. This study provides novel insights into the role of RASGRP1 in insulin secretion and highlights its potential as a therapeutic target for diabetes. The limitations and challenges associated with studying RASGRP1 in disease are also discussed.
Collapse
Affiliation(s)
- Shangzhi Fan
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Bo Kang
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shaoqian Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weiyi Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Canyu Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jixiang Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lijing Deng
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Danjun Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiecan Zhou
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
27
|
Jeon Y, Lee G, Jeong H, Gong G, Kim J, Kim K, Jeong JH, Lee HJ. Proteomic analysis of breast cancer based on immune subtypes. Clin Proteomics 2024; 21:17. [PMID: 38424522 PMCID: PMC10905797 DOI: 10.1186/s12014-024-09463-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Immunotherapy is applied to breast cancer to resolve the limitations of survival gain in existing treatment modalities. With immunotherapy, a tumor can be classified into immune-inflamed, excluded and desert based on the distribution of immune cells. We assessed the clinicopathological features, each subtype's prognostic value and differentially expressed proteins between immune subtypes. METHODS Immune subtyping and proteomic analysis were performed on 56 breast cancer cases with neoadjuvant chemotherapy. The immune subtyping was based on the level of tumor-infiltrating lymphocytes (TILs) and Klintrup criteria. If the level of TILs was ≥ 10%, it was classified as immune-inflamed type without consideration of the Klintrup criteria. In cases of 1-9% TIL, Klintrup criteria 1-3 were classified as the immune-excluded subtype and Klintrup criteria not available (NA) was classified as NA. Cases of 1% TILs and Klintrup 0 were classified as the immune-desert subtype. Mass spectrometry was used to identify differentially expressed proteins in formalin-fixed paraffin-embedded biopsy tissues. RESULTS Of the 56 cases, 31 (55%) were immune-inflamed, 21 (38%) were immune-excluded, 2 (4%) were immune-desert and 2 (4%) were NA. Welch's t-test revealed two differentially expressed proteins between immune-inflamed and immune-excluded/desert subtypes. Coronin-1A was upregulated in immune-inflamed tumors (adjusted p = 0.008) and α-1-antitrypsin was upregulated in immune-excluded/desert tumors (adjusted p = 0.008). Titin was upregulated in pathologic complete response (pCR) than non-pCR among immune-inflamed tumors (adjusted p = 0.036). CONCLUSIONS Coronin-1A and α-1-antitrypsin were upregulated in immune-inflamed and immune-excluded/desert subtypes, respectively. Titin's elevated expression in pCR within the immune-inflamed subtype may indicate a favorable prognosis. Further studies involving large representative cohorts are necessary to validate these findings.
Collapse
Affiliation(s)
- Yeonjin Jeon
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - GunHee Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hwangkyo Jeong
- Prometabio Research Institute, Prometabio co., ltd, Hanam-Si, Gyeonggi-Do, Republic of Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - JiSun Kim
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyunggon Kim
- Department of Digital Medicine, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
- Biomedical Sciences, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
- NeogenTC Corp, Seoul, Korea.
| |
Collapse
|
28
|
Tenggara JB, Rachman A, Prihartono J, Rachmadi L, Panigoro SS, Heriyanto DS, Sutandyo N, Nasution IR, Rahadiati FB, Steven R, Betsy R, Juanputra S, Sudoyo AW. The relationship between high ratios of CD4/FOXP3 and CD8/CD163 and the improved survivability of metastatic triple-negative breast cancer patients: a multicenter cohort study. BMC Res Notes 2024; 17:44. [PMID: 38308298 PMCID: PMC10835864 DOI: 10.1186/s13104-024-06704-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: 07/19/2023] [Accepted: 01/24/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) has been documented as the most aggressive subtype of breast cancer. This study aimed to analyze antitumor and protumor immune activities, and their ratios as significant prognostic biomarkers in metastatic TNBC (mTNBC). METHODS A multicenter cohort study was conducted among 103 de novo mTNBC patients. The expression of CD8 and CD163 was evaluated using immunohistochemistry staining, CD4 and FOXP3 using double-staining immunohistochemistry, and PD-L1 using immunohistochemistry and RT-PCR. RESULTS Multivariate analysis revealed that high CD4/FOXP3 (HR 1.857; 95% CI 1.049-3.288; p = 0.034) and the CD8/CD163 ratio (HR 2.089; 95% CI 1.174-3.717; p = 0.012) yield significantly improved 1 year overall survival (OS). Kaplan-Meier analysis showed that high levels of CD4 (p = 0.023), CD8 (p = 0.043), CD4/FOXP3 (p = 0.016), CD8/FOXP3 (p = 0.005), CD8/CD163 (p = 0.005) ratios were significantly associated with higher rate of 1 year OS. Furthermore, 1 year OS was directly correlated with antitumor CD4 (R = 0.233; p = 0.018) and CD8 (R = 0.219; p = 0.026) and was indirectly correlated with protumor CD163 and FOXP3 through CD4/FOXP3 (R = 0.282; p = 0.006), CD4/CD163 (R = 0.239; p = 0.015), CD8/FOXP3 (R = 0.260; p = 0.008), and CD8/CD163 (R = 0.258; p = 0.009). CONCLUSION This is the first study to demonstrate that high levels of CD4/FOXP3 and CD8/CD163 significantly improved the 1 year OS in de novo mTNBC patients. Thus, we recommend the application of these markers as prognosis determination and individual treatment decision.
Collapse
Affiliation(s)
- Jeffry Beta Tenggara
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia.
- Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia.
| | - Andhika Rachman
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia
- Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia
| | - Joedo Prihartono
- Department of Community Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Lisnawati Rachmadi
- Department of Anatomical Pathology, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Sonar Soni Panigoro
- Department of Surgical Oncology, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Dr. Sardjito Hospital-Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Noorwati Sutandyo
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dharmais National Cancer Hospital, Jakarta, Indonesia
| | - Intan Russianna Nasution
- Division of Hematology and Medical Oncology, Gatot Soebroto Army Hospital Jakarta, Jakarta, Indonesia
| | - Familia Bella Rahadiati
- Department of Anatomical Pathology, Gatot Soebroto Army Hospital Jakarta, Jakarta, Indonesia
| | - Ricci Steven
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia
- Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia
| | - Rachelle Betsy
- Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Samuel Juanputra
- Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Aru Wisaksono Sudoyo
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia
- Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia
| |
Collapse
|
29
|
Jiang X, Lin J, Zhu Z. Long-chain noncoding RNA LINC01569 upregulates filamin A-interacting protein 1-like to prevent metastasis of triple-negative breast cancer via sponging miR-300. Cancer Biomark 2024; 39:79-94. [PMID: 37955081 PMCID: PMC11002719 DOI: 10.3233/cbm-230261] [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: 06/29/2023] [Accepted: 09/18/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Long-chain noncoding RNA (lncRNA), LINC01569, is important for regulating the extracellular matrix, which affects cell migration. However, its involvement in the occurrence and development of triple-negative breast cancer (TNBC) remains unclear. OBJECTIVE This study is aimed to investigate the role of LINC01569 on TNBC. METHODS Online database was used for clinical data analysis. Cell viability and migration capability were monitored using cell counting kit-8 and transwell assays, respectively. Luciferase reporter assay and RNA pull-down were used to confirm the binding capability between noncoding RNAs and filamin A-interacting protein 1-like (FILIP1L). Western blotting was used to determine the protein content. RESULTS Compared with normal breast tissue, LINC01569 was significantly reduced in patients with TNBC subtype, and LINC01569 expression gradually decreased with the progression of tumor stage. Patients with TNBC with high lncRNA LINC01569 levels had a better prognosis than did patients with low LINC01569 levels. LINC01569 overexpression inhibited the migration capability, whereas siRNA-mediated LINC01569 downregulation promoted the migration capability in TNBC cells. Using ENCORI and lncRNA SNP online databases, miR-300 was screened as the potential sponge of LINC01569. The binding of LINC01569 to miR-300 was confirmed using the dual-luciferase reporter and RNA pull-down assays. miR-300 was negatively correlated with LINC01569, and miR-300 mimics eliminated the anti-proliferation and anti-migration effects of LINC01569 on TNBC cells. Additionally, FILIP1L was further verified as the downstream target of miR-300. miR-300 mimics blocked LINC01569 upregulation-mediated elevation of FILIP1L. Importantly, the anti-tumor effects mediated by LINC01569 overexpression were abolished by miR-300 mimics and further restored by FILIP1L upregulation. CONCLUSIONS LINC01569 was expressed at a low level in TNBC and could sponge miR-300 to promote FILIP1L expression, reducing the proliferation and metastasis capability of TNBC. Thus, LINC01569 might be a useful biomarker in the diagnosis and prognosis of metastatic TNBC.
Collapse
Affiliation(s)
| | | | - Zhanlin Zhu
- Department of Breast surgery, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
30
|
Ligorio F, Lobefaro R, Fucà G, Provenzano L, Zanenga L, Nasca V, Sposetti C, Salvadori G, Ficchì A, Franza A, Martinetti A, Sottotetti E, Formisano B, Depretto C, Scaperrotta G, Belfiore A, Vingiani A, Ferraris C, Pruneri G, de Braud F, Vernieri C. Adding fasting-mimicking diet to first-line carboplatin-based chemotherapy is associated with better overall survival in advanced triple-negative breast cancer patients: A subanalysis of the NCT03340935 trial. Int J Cancer 2024; 154:114-123. [PMID: 37615485 DOI: 10.1002/ijc.34701] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/25/2023]
Abstract
Severe calorie restriction, in the form of cyclic fasting or fasting-mimicking diets (FMDs), boosts the antitumor activity of cytotoxic chemotherapy in mouse models of triple-negative breast cancer (TNBC). This effect is mostly mediated by fasting/FMD-induced reduction of plasma glucose concentration and by a boost in antitumor immunity. However, clinical evidence that cyclic FMD may impact on the outcomes of advanced TNBC (aTNBC) patients is lacking. We compared the overall survival (OS) of 14 aTNBC patients receiving first-line carboplatin-gemcitabine plus cyclic FMD in the context of the NCT03340935 trial with the OS of 76 consecutive aTNBC patients treated with carboplatin-based chemotherapy alone at Fondazione IRCCS Istituto Nazionale dei Tumori. Multivariable Cox regression models were used to adjust the prognostic impact of FMD for other prognostic variables. Patients undergoing cyclic FMD in combination with carboplatin-gemcitabine had better OS when compared to patients receiving chemotherapy alone (median OS 30.3 months, 95% CI 18-NR, vs 17.2 months, 95% CI 15.3-25.1, log-rank P value .041). Multivariable analysis confirmed an association between FMD use and better OS (HR: 0.40; 95% CI: 0.19-0.86; P = .019) also after propensity score-based matching according to patient ECOG PS and the presence of de novo metastatic disease (HR: 0.41; 95% CI: 0.21-0.83; P = .013). Cyclic FMD in combination with first-line chemotherapy may improve clinical outcomes in aTNBC patients. Our study paves the way for conducting phase II trials to investigate if cyclic FMD can increase the antitumor activity/efficacy of chemotherapy or chemoimmunotherapy in patients with early-stage TNBC or aTNBC.
Collapse
Affiliation(s)
- Francesca Ligorio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Riccardo Lobefaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Leonardo Provenzano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Lucrezia Zanenga
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Vincenzo Nasca
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Caterina Sposetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Giulia Salvadori
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Angela Ficchì
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Franza
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisa Sottotetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Barbara Formisano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Catherine Depretto
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Antonino Belfiore
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Vingiani
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Cristina Ferraris
- Breast Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giancarlo Pruneri
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| |
Collapse
|
31
|
Castellano G, Giugliano F, Curigliano G, Marra A. Clinical utility of genomic signatures for the management of early and metastatic triple-negative breast cancer. Curr Opin Oncol 2023; 35:479-490. [PMID: 37621170 DOI: 10.1097/cco.0000000000000989] [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: 08/26/2023]
Abstract
PURPOSE OF REVIEW This comprehensive review aims to provide timely and relevant insights into the current therapeutic landscape for triple-negative breast cancer (TNBC) and the molecular features underlying this subtype. It emphasizes the need for more reliable biomarkers to refine prognostication and optimize therapy, considering the aggressive nature of TNBC and its limited targeted treatment options. RECENT FINDINGS The review explores the multidisciplinary management of early TNBC, which typically involves systemic chemotherapy, surgery, and radiotherapy. It highlights the emergence of immune checkpoint inhibitors (ICIs), poly(ADP-ribose) polymerase (PARP) inhibitors, and antibody-drug conjugates (ADCs) as promising therapeutic strategies for TNBC. Recent clinical trials investigating the use of ICIs in combination with chemotherapy and the approval of pembrolizumab and atezolizumab for PD-L1-positive metastatic TNBC are discussed. The efficacy of PARP inhibitors and ADCs in treating TNBC patients with specific genetic alterations is also highlighted. SUMMARY The findings discussed in this review have significant implications for clinical practice and research in TNBC. The identification of distinct molecular subtypes through gene expression profiling has enabled a better understanding of TNBC heterogeneity and its clinical implications. This knowledge has the potential to guide treatment decisions, as different subtypes display varying responses to neoadjuvant chemotherapy. Furthermore, the review emphasizes the importance of developing reliable genomic and transcriptomic signatures as biomarkers to refine patient prognostication and optimize therapy selection in TNBC. Integrating these signatures into clinical practice may lead to more personalized treatment approaches, improving outcomes for TNBC patients.
Collapse
Affiliation(s)
- Grazia Castellano
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Federica Giugliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Antonio Marra
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS
| |
Collapse
|
32
|
Uğurluoğlu C, Yormaz S. Clinicopathological and prognostic value of TIL and PD L1 in triple negative breast carcinomas. Pathol Res Pract 2023; 250:154828. [PMID: 37778126 DOI: 10.1016/j.prp.2023.154828] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/16/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
Triple negative breast cancer (TNBC), a highly aggressive subtype of breast cancer, accounts for 15 % of all diagnosed breast cancers. This group, which has the worst clinical outcome, high recurrence rate and poor prognosis, does not benefit from specific treatment. Therefore, there is a need to develop more effective biomarker and therapeutic strategies especially for this group. A positive level of immunity has been found to be associated with patient survival in various organ cancers. More specifically, tumor infiltrating lymphocytes (TIL) have been documented to have strong prognostic value. The programmed cell death 1 (PD 1) protein on the surface of T lymphocytes is activated by the Programmed cell death ligand 1 (PD-L1) protein on the cancer cell surface. PD- L1 is thought to form a pathway that results in suppression of antitumor responses when activated. Patients with breast cancer (BC) who underwent resection without neoadjuvant chemotherapy between 2010 and 2020 were included in this study. Of the 302 BCs examined, 21 constitute the group with TNBC. In our study, the mean age of the Triple positive breast cancer (TPBC) and TNBC groups was similar (55.67 ± 12.61 vs. 53.23 ± 8.21, p = 0.384). There was no significant correlation between TPBC and TNBC and tumor size, lymph node, histological grade, and PD-L1 positivity in the center of the tumor (all p-value >.05). It was observed that tumor stage was higher in patients with TNBC than in patients with TPBC (19 % vs. 1.1 %, p = .002). The Ki 67 proliferation index was found to be higher in patients with TNBC than in patients with TPBC (90.5 % vs. 41.8 %, p .001). Although not statistically significant, clinically, CD 3 and CD 8 immune scores with high tumor margin were higher in patients with TNBC than in patients with TPBC (90.4 % vs, 9.6 % and 85.7 % vs. 14.3 %, respectively). Positive expression of PD-L1 at the tumor margin was significantly higher in patients with TNBC than patients with TPBC (20.3 % vs, 52.4 %, p = .002). By Kaplan-Meier analysis, the survival distribution of CD 3 and CD 8 immunoscore, tumor central and margin PD-L1 values were compared. Mean follow-up was 136.18 months (range, 1 - 144 months); and the 10-year Overall Survival (OS) estimate for the population was 90.9 % (95 % CI, 85.5 - 96.7). In this study, this difference was not statistically significant according to the log-rank test. In this study, we aimed to evaluate the relationship between CD 3, CD 8 T lymphocyte immune score and PD-L1 expression at the tumor center and margin in TNBC, the prognostic value and clinicopathological significance of this relationship.
Collapse
Affiliation(s)
- Ceyhan Uğurluoğlu
- Department of Patology, Faculty of Medical, Selçuk University, Konya, Turkey.
| | - Serdar Yormaz
- Department of General Surgery, Faculty of Medical, Selçuk University, Konya, Turkey
| |
Collapse
|
33
|
Reinisch M, Bruzas S, Spoenlein J, Shenoy S, Traut A, Harrach H, Chiari O, Cremer E, Ataseven B, Gubelt L, Kuemmel S. Safety and effectiveness of sacituzumab govitecan in patients with metastatic triple-negative breast cancer in real-world settings: first observations from an interdisciplinary breast cancer centre in Germany. Ther Adv Med Oncol 2023; 15:17588359231200454. [PMID: 37789989 PMCID: PMC10542232 DOI: 10.1177/17588359231200454] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/25/2023] [Indexed: 10/05/2023] Open
Abstract
Background Sacituzumab govitecan has been recently approved by the USFDA and EMA for the treatment of patients with metastatic triple-negative breast cancer (mTNBC). We report real-world safety and effectiveness in patients with mTNBC receiving sacituzumab govitecan treatment at a breast cancer centre in Germany. Methods Data from patients who had received sacituzumab govitecan as treatment for mTNBC, in both de novo and relapsed disease, at the Kliniken Essen-Mitte, Essen, Germany, were collected through institutional records. Data were analysed for safety parameters and survival outcomes and reported using descriptive statistics. Results Patients (N = 43) received a median (range) of 5 (1-28) cycles of sacituzumab govitecan and were followed up for a median of 12.9 months. The most reported adverse events (AEs) of any grade were alopecia (n = 39; 90.7%), diarrhoea (n = 16; 37.2%), fatigue (n = 15, 34.9%), anaemia (n = 15, 34.9%) and neutropenia (n = 14, 32.6%). AEs ⩾ Grade 3 with the highest incidence were neutropenia (n = 12; 27.9%) and diarrhoea (n = 8; 18.6%). In eight (18.6%) patients, dose of sacituzumab govitecan dose was reduced due to patients' clinical condition prior to commencing treatment; in further 17 (39.5%) patients, sacituzumab govitecan dose had to be reduced or treatment interrupted on account of AEs associated with the drug after treatment had commenced. Median progression-free survival and median overall survival were calculated to be 5.0and 13.1 months, respectively. Conclusion The real-world safety and effectiveness profile of sacituzumab govitecan in patients with mTNBC are in line with clinical trial data. Further studies are required to guide optimal use of sacituzumab govitecan against mTNBC, especially in context of management of accompanying AEs.
Collapse
Affiliation(s)
- Mattea Reinisch
- Interdisciplinary Breast Unit, Kliniken Essen-Mitte, Henricistrasse 92, Essen 45136, Germany
| | - Simona Bruzas
- Interdisciplinary Breast Unit, Kliniken Essen-Mitte, Essen, Germany
| | | | - Satyendra Shenoy
- Interdisciplinary Breast Unit, Kliniken Essen-Mitte, Essen, Germany
| | - Alexander Traut
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany
| | - Hakima Harrach
- Interdisciplinary Breast Unit, Kliniken Essen-Mitte, Essen, Germany
| | - Ouafaa Chiari
- Interdisciplinary Breast Unit, Kliniken Essen-Mitte, Essen, Germany
| | - Efsthatia Cremer
- Interdisciplinary Breast Unit, Kliniken Essen-Mitte, Essen, Germany
| | - Beyhan Ataseven
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Lars Gubelt
- Zentralapotheke, Kliniken Essen-Mitte, Essen, Germany
| | - Sherko Kuemmel
- Interdisciplinary Breast Unit, Kliniken Essen-Mitte, Essen, Germany
- Department of Gynecology with Breast Center, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
34
|
Zhang J, Zhang Z, Huang Z, Li M, Yang F, Wu Z, Guo Q, Mei X, Lu B, Wang C, Wang Z, Ji L. Isotoosendanin exerts inhibition on triple-negative breast cancer through abrogating TGF- β-induced epithelial-mesenchymal transition via directly targeting TGF βR1. Acta Pharm Sin B 2023; 13:2990-3007. [PMID: 37521871 PMCID: PMC10372922 DOI: 10.1016/j.apsb.2023.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/07/2023] [Accepted: 03/14/2023] [Indexed: 08/01/2023] Open
Abstract
As the most aggressive breast cancer, triple-negative breast cancer (TNBC) is still incurable and very prone to metastasis. The transform growth factor β (TGF-β)-induced epithelial-mesenchymal transition (EMT) is crucially involved in the growth and metastasis of TNBC. This study reported that a natural compound isotoosendanin (ITSN) reduced TNBC metastasis by inhibiting TGF-β-induced EMT and the formation of invadopodia. ITSN can directly interact with TGF-β receptor type-1 (TGFβR1) and abrogated the kinase activity of TGFβR1, thereby blocking the TGF-β-initiated downstream signaling pathway. Moreover, the ITSN-provided inhibition on metastasis obviously disappeared in TGFβR1-overexpressed TNBC cells in vitro as well as in mice bearing TNBC cells overexpressed TGFβR1. Furthermore, Lys232 and Asp351 residues in the kinase domain of TGFβR1 were found to be crucial for the interaction of ITSN with TGFβR1. Additionally, ITSN also improved the inhibitory efficacy of programmed cell death 1 ligand 1 (PD-L1) antibody for TNBC in vivo via inhibiting the TGF-β-mediated EMT in the tumor microenvironment. Our findings not only highlight the key role of TGFβR1 in TNBC metastasis, but also provide a leading compound targeting TGFβR1 for the treatment of TNBC metastasis. Moreover, this study also points out a potential strategy for TNBC treatment by using the combined application of anti-PD-L1 with a TGFβR1 inhibitor.
Collapse
Affiliation(s)
- Jingnan Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ze Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhenlin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Manlin Li
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fan Yang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zeqi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qian Guo
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiyu Mei
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Changhong Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhengtao Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
35
|
Greco S, Fabbri N, Spaggiari R, De Giorgi A, Fabbian F, Giovine A. Update on Classic and Novel Approaches in Metastatic Triple-Negative Breast Cancer Treatment: A Comprehensive Review. Biomedicines 2023; 11:1772. [PMID: 37371867 PMCID: PMC10296377 DOI: 10.3390/biomedicines11061772] [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: 05/15/2023] [Revised: 06/09/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for almost 15% of all diagnosed breast cancers and often presents high rates of relapses and metastases, with generally poor prognosis despite multiple lines of treatment. Immunotherapy has radically changed the approach of clinicians towards TNBC in the last two to three years, even if targeted and specific therapeutic options are still missing; this unmet need is further justified by the extreme molecular and clinical heterogeneity of this subtype of breast cancer and by the weak response to both single-agent and combined therapies. In March 2023, the National Comprehensive Cancer Network (NCCN), the main association of cancer centers in the United States, released the last clinical practice guidelines, with an update on classic and novel approaches in the field of breast cancer. The purpose of this comprehensive review is to summarize the latest findings in the setting of metastatic TNBC treatment, focusing on each category of drugs approved by the Food and Drug Administration (FDA) and included in the NCCN guidelines. We also introduce part of the latest published studies, which have reported new and promising molecules able to specifically target some of the biomarkers involved in TNBC pathogenesis. We searched the PubMed and Scopus databases for free full texts reported in the literature of the last 5 years, using the words "triple-negative breast cancer" or "TNBC" or "basal-like". The articles were analyzed by the authors independently and double-blindly, and a total of 114 articles were included in the review.
Collapse
Affiliation(s)
- Salvatore Greco
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (S.G.); (R.S.)
- Department of Internal Medicine, Delta Hospital, Via Valle Oppio 2, 44023 Ferrara, Italy;
| | - Nicolò Fabbri
- Department of General Surgery, Delta Hospital, Via Valle Oppio 2, 44023 Ferrara, Italy;
| | - Riccardo Spaggiari
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (S.G.); (R.S.)
| | - Alfredo De Giorgi
- Department of Internal Medicine, University Hospital of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy;
| | - Fabio Fabbian
- Department of Medical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Antonio Giovine
- Department of Internal Medicine, Delta Hospital, Via Valle Oppio 2, 44023 Ferrara, Italy;
| |
Collapse
|
36
|
Ahmadi SE, Shabannezhad A, Kahrizi A, Akbar A, Safdari SM, Hoseinnezhad T, Zahedi M, Sadeghi S, Mojarrad MG, Safa M. Tissue factor (coagulation factor III): a potential double-edge molecule to be targeted and re-targeted toward cancer. Biomark Res 2023; 11:60. [PMID: 37280670 DOI: 10.1186/s40364-023-00504-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/19/2023] [Indexed: 06/08/2023] Open
Abstract
Tissue factor (TF) is a protein that plays a critical role in blood clotting, but recent research has also shown its involvement in cancer development and progression. Herein, we provide an overview of the structure of TF and its involvement in signaling pathways that promote cancer cell proliferation and survival, such as the PI3K/AKT and MAPK pathways. TF overexpression is associated with increased tumor aggressiveness and poor prognosis in various cancers. The review also explores TF's role in promoting cancer cell metastasis, angiogenesis, and venous thromboembolism (VTE). Of note, various TF-targeted therapies, including monoclonal antibodies, small molecule inhibitors, and immunotherapies have been developed, and preclinical and clinical studies demonstrating the efficacy of these therapies in various cancer types are now being evaluated. The potential for re-targeting TF toward cancer cells using TF-conjugated nanoparticles, which have shown promising results in preclinical studies is another intriguing approach in the path of cancer treatment. Although there are still many challenges, TF could possibly be a potential molecule to be used for further cancer therapy as some TF-targeted therapies like Seagen and Genmab's tisotumab vedotin have gained FDA approval for treatment of cervical cancer. Overall, based on the overviewed studies, this review article provides an in-depth overview of the crucial role that TF plays in cancer development and progression, and emphasizes the potential of TF-targeted and re-targeted therapies as potential approaches for the treatment of cancer.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Departments of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ashkan Shabannezhad
- Departments of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Kahrizi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Armin Akbar
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mehrab Safdari
- Departments of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Taraneh Hoseinnezhad
- Department of Hematolog, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soroush Sadeghi
- Faculty of Science, Engineering and Computing, Kingston University, London, UK
| | - Mahsa Golizadeh Mojarrad
- Shahid Beheshti Educational and Medical Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Majid Safa
- Departments of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Avancini G, Menilli L, Visentin A, Milani C, Mastrotto F, Moret F. Mesenchymal Stem Cell Membrane-Coated TPCS 2a-Loaded Nanoparticles for Breast Cancer Photodynamic Therapy. Pharmaceutics 2023; 15:1654. [PMID: 37376102 PMCID: PMC10302938 DOI: 10.3390/pharmaceutics15061654] [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: 05/03/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Despite substantial improvements in breast cancer (BC) treatment there is still an urgent need to find alternative treatment options to improve the outcomes for patients with advanced-stage disease. Photodynamic therapy (PDT) is gaining a lot of attention as a BC therapeutic option because of its selectivity and low off-target effects. However, the hydrophobicity of photosensitizers (PSs) impairs their solubility and limits the circulation in the bloodstream, thus representing a major challenge. The use of polymeric nanoparticles (NPs) to encapsulate the PS may represent a valuable strategy to overcome these issues. Herein, we developed a novel biomimetic PDT nanoplatform (NPs) based on a polymeric core of poly(lactic-co-glycolic)acid (PLGA) loaded with the PS meso-tetraphenylchlorin disulfonate (TPCS2a). TPCS2a@NPs of 98.89 ± 18.56 nm with an encapsulation efficiency percentage (EE%) of 81.9 ± 7.92% were obtained and coated with mesenchymal stem cells-derived plasma membranes (mMSCs) (mMSC-TPCS2a@NPs, size of 139.31 ± 12.94 nm). The mMSC coating armed NPs with biomimetic features to impart long circulation times and tumor-homing capabilities. In vitro, biomimetic mMSC-TPCS2a@NPs showed a decrease in macrophage uptake of 54% to 70%, depending on the conditions applied, as compared to uncoated TPCS2a@NPs. Both NP formulations efficiently accumulated in MCF7 and MDA-MB-231 BC cells, while the uptake was significantly lower in normal breast epithelial MCF10A cells with respect to tumor cells. Moreover, encapsulation of TPCS2a in mMSC-TPCS2a@NPs effectively prevents its aggregation, ensuring efficient singlet oxygen (1O2) production after red light irradiation, which resulted in a considerable in vitro anticancer effect in both BC cell monolayers (IC50 < 0.15 µM) and three-dimensional spheroids.
Collapse
Affiliation(s)
- Greta Avancini
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| | - Luca Menilli
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| | - Adele Visentin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Celeste Milani
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Francesca Moret
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| |
Collapse
|
38
|
Sun M, Chen G, Ouyang S, Chen C, Zheng Z, Lin P, Song X, Chen H, Chen Y, You Y, Tao J, Lin B, Zhao P. Magnetic Resonance Diagnosis of Early Triple-Negative Breast Cancer Based on the Ionic Covalent Organic Framework with High Relaxivity and Long Retention Time. Anal Chem 2023; 95:8267-8276. [PMID: 37191204 DOI: 10.1021/acs.analchem.3c00307] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Patients with triple-negative breast cancer (TNBC) have dismal prognoses due to the lack of therapeutic targets and susceptibility to lymph node (LN) metastasis. Therefore, it is essential to develop more effective approaches to identify early TNBC tissues and LNs. In this work, a magnetic resonance imaging (MRI) contrast agent (Mn-iCOF) was constructed based on the Mn(II)-chelated ionic covalent organic framework (iCOF). Because of the porous structure and hydrophilicity, the Mn-iCOF has a high longitudinal relaxivity (r1) of 8.02 mM-1 s-1 at 3.0 T. For the tumor-bearing mice, a lower dose (0.02 mmol [Mn]/kg) of Mn-iCOF demonstrated a higher signal-to-noise ratio (SNR) value (1.8) and longer retention time (2 h) compared to a 10-fold dose of commercial Gd-DOTA (0.2 mmol [Gd]/kg). Moreover, the Mn-iCOF can provide continuous and significant MR contrast for the popliteal LNs within 24 h, allowing for accurate evaluation and dissection of LNs. These excellent MRI properties of the Mn-iCOF may open new avenues for designing more biocompatible MRI contrast agents with higher resolutions, particularly in the diagnosis of TNBC.
Collapse
Affiliation(s)
- Mingyan Sun
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515 Guangzhou, China
| | - Guanjun Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Sixue Ouyang
- School of Chemistry and Chemical Engineering, South China University of Technology, 510640 Guangzhou, China
| | - Chuyao Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Zhiyuan Zheng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515 Guangzhou, China
| | - Peiru Lin
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515 Guangzhou, China
| | - Xiangfei Song
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515 Guangzhou, China
| | - Huiting Chen
- School of Chemistry and Chemical Engineering, South China University of Technology, 510640 Guangzhou, China
| | - Yuying Chen
- School of Chemistry and Chemical Engineering, South China University of Technology, 510640 Guangzhou, China
| | - Yuanyuan You
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515 Guangzhou, China
| | - Jia Tao
- School of Chemistry and Chemical Engineering, South China University of Technology, 510640 Guangzhou, China
| | - Bingquan Lin
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Peng Zhao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515 Guangzhou, China
| |
Collapse
|
39
|
Geurts V, Kok M. Immunotherapy for Metastatic Triple Negative Breast Cancer: Current Paradigm and Future Approaches. Curr Treat Options Oncol 2023; 24:628-643. [PMID: 37079257 PMCID: PMC10172210 DOI: 10.1007/s11864-023-01069-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2023] [Indexed: 04/21/2023]
Abstract
OPINION STATEMENT In approximately 15-20% of the patients diagnosed with breast cancer, it comprises the triple negative (TN) subtype, which until recently lacked targets for specific treatments and is known for its aggressive clinical behavior in patients with metastatic disease. TNBC is considered the most immunogenic breast cancer subtype due to higher levels of tumor infiltrating lymphocytes (TILs), tumor mutational burden and PD-L1 expression, providing a rationale for immunotherapy. The addition of pembrolizumab to chemotherapy as first-line treatment resulted in significantly improved PFS and OS for PD-L1 positive mTNBC, leading to FDA approval. However, response rate of ICB in unselected patients is low. Ongoing (pre)clinical trials aim to further optimize ICB efficacy and widen its application beyond PD-L1 positive breast tumors. Novel immunomodulatory approaches to induce a more inflamed tumor microenvironment include dual checkpoint blockade, bispecific antibodies, immunocytokines, adoptive cell therapies, oncolytic viruses, and cancer vaccines. Preclinical data for these novel strategies seems promising, but solid clinical data to further support its application for mTNBC is awaited. Biomarkers capturing the degree of immunogenicity such as but not limited to TILs, CD8 T cell levels, and IFNg signatures could support deciding which therapeutic strategy is most appropriate for which patient. Given 1) the accumulating therapy options for patients with metastatic disease and 2) the heterogeneity of mTNBC from inflamed to immune-desert tumors, the challenge is to work towards immunomodulatory strategies for specific subgroups of patients with TNBC to enable personalized (immuno)therapy for patients with metastatic disease.
Collapse
Affiliation(s)
- Veerle Geurts
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
| | - Marleen Kok
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands.
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands.
| |
Collapse
|
40
|
Mukherjee D, Previs RA, Haines CN, Abo MA, Juras PK, Strickland KC, Chakraborty B, Artham S, Whitaker R, Hebert KL, Fontenot J, Patierno SR, Freedman JA, Lau FH, Burow M, Chang CY, McDonnell DP. Ca 2+ /Calmodulin Dependent Protein Kinase Kinase-2 (CaMKK2) promotes Protein Kinase G (PKG)-dependent actin cytoskeletal assembly to increase tumor metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.536051. [PMID: 37131673 PMCID: PMC10153149 DOI: 10.1101/2023.04.17.536051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Triple-negative breast cancers (TNBCs) tend to become highly invasive early during cancer development. Despite some successes in the initial treatment of patients diagnosed with early-stage localized TNBC, the rate of metastatic recurrence remains high with poor long-term survival outcomes. Here we show that elevated expression of the serine/threonine-kinase, Calcium/Calmodulin (CaM)-dependent protein kinase kinase-2 (CaMKK2), is highly correlated with tumor invasiveness. We determined that genetic disruption of CaMKK2 expression, or inhibition of its activity, disrupted spontaneous metastatic outgrowth from primary tumors in murine xenograft models of TNBC. High-grade serous ovarian cancer (HGSOC), a high-risk, poor-prognosis ovarian cancer subtype, shares many genetic features with TNBC, and importantly, CaMKK2 inhibition effectively blocked metastatic progression in a validated xenograft model of this disease. Probing the mechanistic links between CaMKK2 and metastasis we defined the elements of a new signaling pathway that impacts actin cytoskeletal dynamics in a manner which increases cell migration/invasion and metastasis. Notably, CaMKK2 increases the expression of the phosphodiesterase PDE1A which decreases the cGMP-dependent activity of protein kinase G1 (PKG1). This inhibition of PKG1 results in decreased phosphorylation of Vasodilator-Stimulated Phosphoprotein (VASP), which in its hypophosphorylated state binds to and regulates F-actin assembly to facilitate contraction/cell movement. Together, these data establish a targetable CaMKK2-PDE1A-PKG1-VASP signaling pathway that controls cancer cell motility and metastasis. Further, it credentials CaMKK2 as a therapeutic target that can be exploited in the discovery of agents for use in the neoadjuvant/adjuvant setting to restrict tumor invasiveness in patients diagnosed with early-stage TNBC or localized HGSOC.
Collapse
|
41
|
Saleh L, Ottewell PD, Brown JE, Wood SL, Brown NJ, Wilson C, Park C, Ali S, Holen I. The CDK4/6 Inhibitor Palbociclib Inhibits Estrogen-Positive and Triple Negative Breast Cancer Bone Metastasis In Vivo. Cancers (Basel) 2023; 15:cancers15082211. [PMID: 37190140 DOI: 10.3390/cancers15082211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/04/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
CDK 4/6 inhibitors have demonstrated significant improved survival for patients with estrogen receptor (ER) positive breast cancer (BC). However, the ability of these promising agents to inhibit bone metastasis from either ER+ve or triple negative BC (TNBC) remains to be established. We therefore investigated the effects of the CDK 4/6 inhibitor, palbociclib, using in vivo models of breast cancer bone metastasis. In an ER+ve T47D model of spontaneous breast cancer metastasis from the mammary fat pad to bone, primary tumour growth and the number of hind limb skeletal tumours were significantly lower in palbociclib treated animals compared to vehicle controls. In the TNBC MDA-MB-231 model of metastatic outgrowth in bone (intracardiac route), continuous palbociclib treatment significantly inhibited tumour growth in bone compared to vehicle. When a 7-day break was introduced after 28 days (mimicking the clinical schedule), tumour growth resumed and was not inhibited by a second cycle of palbociclib, either alone or when combined with the bone-targeted agent, zoledronic acid (Zol), or a CDK7 inhibitor. Downstream phosphoprotein analysis of the MAPK pathway identified a number of phosphoproteins, such as p38, that may contribute to drug-insensitive tumour growth. These data encourage further investigation of targeting alternative pathways in CDK 4/6-insensitive tumour growth.
Collapse
Affiliation(s)
- Lubaid Saleh
- Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - Penelope D Ottewell
- Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - Janet E Brown
- Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
- Weston Park Hospital, Whitham Road, Sheffield S10 2SJ, UK
| | - Steve L Wood
- Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - Nichola J Brown
- Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | | | - Catherine Park
- Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - Simak Ali
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Ingunn Holen
- Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| |
Collapse
|
42
|
Yuan L, Liu J, Bao L, Qu H, Xiang J, Sun P. Upregulation of the ferroptosis-related STEAP3 gene is a specific predictor of poor triple-negative breast cancer patient outcomes. Front Oncol 2023; 13:1032364. [PMID: 37064114 PMCID: PMC10102497 DOI: 10.3389/fonc.2023.1032364] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
ObjectiveThis study was designed to assess ferroptosis regulator gene (FRG) expression patterns in patients with TNBC based on data derived from The Cancer Genome Atlas (TCGA). Further, it was utilized to establish a TNBC FRG signature, after which the association between this signature and the tumor immune microenvironment (TIME) composition was assessed, and relevant prognostic factors were explored.MethodsThe TCGA database was used to obtain RNA expression datasets and clinical information about 190 TNBC patients, after which a prognostic TNBC-related FRG signature was established using a least absolute shrinkage and selection operator (LASSO) Cox regression approach. These results were validated with separate data from the Gene Expression Omnibus (GEO). The TNBC-specific prognostic gene was identified via this method. The STEAP3 was then validated through Western immunoblotting, immunohistochemical staining, and quantitative real‐time polymerase chain reaction (RT-qPCR) analyses of clinical tissue samples and TNBC cell lines. Chemotherapy interactions and predicted drug sensitivity studies were investigated to learn more about the potential clinical relevance of these observations.ResultsThese data revealed that 87 FRGs were differentially expressed when comparing TNBC tumors and healthy tissue samples (87/259, 33.59%). Seven of these genes (CA9, CISD1, STEAP3, HMOX1, DUSP1, TAZ, HBA1) are significantly related to the overall survival of TNBC patients. Kaplan-Meier analyses and established FRG signatures and nomograms identified CISD1 and STEAP3 genes of prognostic relevance. Prognostic Risk Score values were positively correlated with the infiltration of CD4+ T cells (p = 0.001) and myeloid dendritic cells (p =0.004). Further evidence showed that STEAP3 was strongly and specifically associated with TNBC patient OS (P<0.05). The results above were confirmed by additional examinations of STEAP3 expression changes in TNBC patient samples and cell lines. High STEAP3 levels were negatively correlated with half-maximal inhibitory concentration (IC50) values for GSK1904529A (IGF1R inhibitor), AS601245 (JNK inhibitor), XMD8−85 (Erk5 inhibitor), Gefitinib, Sorafenib, and 5-Fluorouracil (P < 0.05) in patients with TNBC based on information derived from the TCGA-TNBC dataset.ConclusionIn the present study, a novel FRG model was developed and used to forecast the prognosis of TNBC patients accurately. Furthermore, it was discovered that STEAP3 was highly overexpressed in people with TNBC and associated with overall survival rates, laying the groundwork for the eventually targeted therapy of individuals with this form of cancer.
Collapse
Affiliation(s)
- Lifang Yuan
- Department of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, China
- Department of Breast Oncology, Huanxing Cancer Hospital, Beijing, China
| | - Jiannan Liu
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Lei Bao
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Huajun Qu
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jinyu Xiang
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Ping Sun
- Department of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, China
- *Correspondence: Ping Sun,
| |
Collapse
|
43
|
Zhou Y, Yu S, Chen D, Li H, Xu P, Yuan C, Jiang L, Huang M. Nafamostat Mesylate in Combination with the Mouse Amino-Terminal Fragment of Urokinase-Human Serum Albumin Improves the Treatment Outcome of Triple-Negative Breast Cancer Therapy. Mol Pharm 2023; 20:905-917. [PMID: 36463525 DOI: 10.1021/acs.molpharmaceut.2c00297] [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: 12/07/2022]
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive and causes a higher proportion of metastatic cases. However, therapies directed to specific molecular targets have rarely achieved clinically meaningful improvements in the outcome of TNBC therapy. A urokinase-type plasminogen activator (uPA), one of the best-validated biomarkers of breast cancer, is an extracellular proteolytic serine protease involved in many pathological and physiological processes, including tumor cell invasion and metastasis. Nafamostat mesylate (NM) is a synthetic compound that inhibits various serine proteases and has been used as a therapeutic agent for the treatment of TNBC. Nevertheless, NM has poor specificity for serine proteases and is easy be hydrolyzed; moreover, the inhibitory mechanism of TNBC therapy is unclear. In this study, we combine NM with a macromolecular drug delivery vehicle, mouse amino-terminal fragment of urokinase-human serum albumin (mATF-HSA), to form a complex (mATF-HSA:NM) using the dilution-incubation-purification method. mATF specifically targets uPAR overexpressed on the surface of TNBC cells; moreover, HSA prevents NM from being hydrolyzed by numerous serine proteases. mATF-HSA:NM showed stronger inhibitory effects on the proliferation and metastasis of TNBC in vitro and in vivo without significant cytotoxicity on normal cells and tissues. In addition, we demonstrated that NM mediates metastasis of TNBC cells through inhibition of uPA using a stable uPA knockdown cell line (MDA-MB231 shuPA). Overall, we have developed a macromolecular complex targeted to treat high uPAR-expressing tumor types, and mATF-HSA can potentially be used to load other types of drugs with tumor-targeting specificity for mouse tumor models and is a promising tool to study tumor biology in mouse tumor models.
Collapse
Affiliation(s)
- Yang Zhou
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Shujuan Yu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Dan Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Hanlin Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China.,Fujian Key Lab Moratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| |
Collapse
|
44
|
Marini W, Wilson BE, Reedijk M. Targeting Notch-Driven Cytokine Secretion: Novel Therapies for Triple Negative Breast Cancer. DNA Cell Biol 2023; 42:73-81. [PMID: 36579947 DOI: 10.1089/dna.2022.0578] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Compared with other breast cancer subtypes, triple negative breast cancer (TNBC) is an aggressive malignancy with a high recurrence rate and reduced overall survival. Immune checkpoint inhibition (ICI) has shown modest results in this subgroup, highlighting the need for improved targeted therapeutic options. Notch is a defining feature of TNBC and drives the expression of interleukin-1 beta (IL1β) and C-C motif chemokine ligand 2 (CCL2). These cytokines are involved in the recruitment of tumor-associated macrophages (TAMs) to the tumor, resulting in immune evasion and tumor progression. Targeting Notch, IL1β or CCL2 may reduce TAM recruitment and resistance to ICI, illuminating the potential of combination immunotherapy in TNBC.
Collapse
Affiliation(s)
- Wanda Marini
- Division of General Surgery, University of Toronto, Toronto, Ontario, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Brooke E Wilson
- Department of Oncology, Queen's University, Kingston, Ontario, Canada.,Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Michael Reedijk
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
45
|
Cai SL, Liu JJ, Liu YX, Yu SH, Liu X, Lin XQ, Chen HD, Fang X, Ma T, Li YQ, Li Y, Li CY, Zhang S, Chen XG, Guo XJ, Zhang J. Characteristics of recurrence, predictors for relapse and prognosis of rapid relapse triple-negative breast cancer. Front Oncol 2023; 13:1119611. [PMID: 36874102 PMCID: PMC9978400 DOI: 10.3389/fonc.2023.1119611] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC) patients who recur at different times are associated with distinct biological characteristics and prognoses. Research on rapid-relapse TNBC (RR-TNBC) is sparse. In this study, we aimed to describe the characteristics of recurrence, predictors for relapse, and prognosis in rrTNBC patients. Methods Clinicopathological data of 1584 TNBC patients from 2014 to 2016 were retrospectively reviewed. The characteristics of recurrence were compared between patients with RR-TNBC and slow relapse TNBC(SR-TNBC). All TNBC patients were randomly divided into a training set and a validation set to find predictors for rapid relapse. The multivariate logistic regression model was used to analyze the data of the training set. C-index and brier score analysis for predicting rapid relapse in the validation set was used to evaluate the discrimination and accuracy of the multivariate logistic model. Prognostic measurements were analyzed in all TNBC patients. Results Compared with SR-TNBC patients, RR-TNBC patients tended to have a higher T staging, N staging, TNM staging, and low expression of stromal tumor-infiltrating lymphocytes (sTILs). The recurring characteristics were prone to appear as distant metastasis at the first relapse. The first metastatic site was apt to visceral metastasis and less likely to have chest wall or regional lymph node metastasis. Six predictors (postmenopausal status, metaplastic breast cancer,≥pT3 staging,≥pN1 staging, sTIL intermediate/high expression, and Her2 [1+]) were used to construct the predictive model of rapid relapse in TNBC patients. The C-index and brier score in the validation set was 0.861 and 0.095, respectively. This suggested that the predictive model had high discrimination and accuracy. The prognostic data for all TNBC patients showed that RR-TNBC patients had the worst prognosis, followed by SR-TNBC patients. Conclusion RR-TNBC patients were associated with unique biological characteristics and worse outcomes compared to non-RR-TNBC patients.
Collapse
Affiliation(s)
- Shuang-Long Cai
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Jing-Jing Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Ying-Xue Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Shao-Hong Yu
- College of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xu Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xiu-Quan Lin
- Department for Chronic and Noncommunicable Disease Control and Prevention, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Hong-Dan Chen
- First Department of Cadre Clinic, Provincial Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Xuan Fang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Tao Ma
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Ya-Qing Li
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of Oncological Surgery, Provincial Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Ying Li
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Chun-Yan Li
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Sheng Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xiao-Geng Chen
- Department of Oncological Surgery, Provincial Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Xiao-Jing Guo
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jin Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| |
Collapse
|
46
|
Huang M, O'Shaughnessy J, Haiderali A, Pan W, Hu P, Chaudhuri M, Le Bailly De Tilleghem C, Cappoen N, Fasching PA. Q-TWiST analysis of pembrolizumab combined with chemotherapy as first-line treatment of metastatic triple-negative breast cancer that expresses PD-L1. Eur J Cancer 2022; 177:45-52. [PMID: 36323052 DOI: 10.1016/j.ejca.2022.09.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 01/06/2023]
Abstract
OBJECTIVE In the KEYNOTE-355 (KN355) trial, pembrolizumab in combination with chemotherapy demonstrated superior efficacy and manageable safety compared with chemotherapy alone in patients with previously untreated locally recurrent inoperable and metastatic triple-negative breast cancer (mTNBC) with PD-L1 positive (Combined Positive Score [CPS]≥ 10) tumours. This study aimed to evaluate the clinical benefits and risks of pembrolizumab measured by quality-adjusted survival in the trial population. METHODS The study used data from the final analysis of KN355. The Quality-adjusted Time Without Symptoms of disease progression or Toxicity of treatment (Q-TWiST) analysis was used to compare treatments of pembrolizumab plus chemotherapy versus chemotherapy alone. Patients' survival time was partitioned into three health states - toxicity before disease progression (TOX), time without symptoms or toxicity before disease progression (TWiST), and relapse (REL). Utilities for these health states were estimated using EuroQol-5 Dimensions, 3 Levels (EQ-5D-3L) data collected in KN355. Q-TWiST was derived as the utility-weighted sum of the mean health state durations. RESULTS Patients randomised to pembrolizumab plus chemotherapy had 3.7 months greater Q-TWiST (relative gain of 18%; P = 0.003) compared to those randomised to chemotherapy at the median follow-up of 44 months, and 4.3 months greater Q-TWiST (relative gain of 20%; P = 0.004) at the maximum follow-up of 52 months. The Q-TWiST gain increased with longer follow-up time. CONCLUSIONS Pembrolizumab plus chemotherapy was associated with statistically significant and clinically important improvement in Q-TWiST compared to chemotherapy in previously untreated PD-L1-positive mTNBC.
Collapse
Affiliation(s)
| | - Joyce O'Shaughnessy
- Baylor University Medical Center, Texas Oncology and US Oncology, Dallas, TX, USA
| | | | | | - Peter Hu
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | | | - Peter A Fasching
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Department of Gynecology and Obstetrics, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
47
|
Han Y, Wang J, Wu Y, Xu H, Wang Y, Xu B. Optimal Choice as First-Line Therapy for Patients with Triple-Negative Breast Cancer: A Bayesian Network Meta-Analysis. Curr Oncol 2022; 29:9172-9180. [PMID: 36547132 PMCID: PMC9777258 DOI: 10.3390/curroncol29120718] [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: 10/15/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
To identify the advantageous therapy as the first-line treatment for patients with triple-negative breast cancer (TNBC). Randomized controlled trials were searched for on Medline, Embase, ClinicalTrials.gov, and the Cochrane Library between January 2001 and December 2021. The primary endpoint was progression-free survival (PFS) and secondary endpoints were overall survival (OS) and treatment-related adverse events (TRAEs). A Bayesian framework was applied to facilitate indirect comparisons, of which the outcomes were presented using cumulative ranking curve (SUCRA) values, synthesized hazard ratio, risk ratio, and 95% credible interval. A total of 3140 patients were identified. Pooled results of PFS revealed that chemotherapy plus AKT inhibitors (AKTi) was likely the most effective therapy among enrolled therapies (SUCRA = 91.6%), of which the result remained consistent in comparative analysis for OS. In addition, no significant difference was detected between PD-1/PD-L1 antibodies in patients, whereas the PD-1 inhibitors (PD-1i) regimen was advantageous over PD-L1 inhibitor (PD-L1i) therapy for PD-L1 positive TNBC. Concerning TRAEs, an apparent heterogeneity associated with safety profiles were denoted among enrolled agents. Chemotherapy plus AKTi was the most effective therapy with comparable safety profiles. Chemotherapy plus the anti-PD-1 regimen was advantageous over the combination therapy based on the PD-L1 blockade.
Collapse
Affiliation(s)
| | - Jiayu Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | | | | | | | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| |
Collapse
|
48
|
Vaid PM, Puntambekar AK, Jumle NS, Banale RA, Ansari D, Reddy RR, Unde RR, Namewar NP, Kelkar DA, Shashidhara LS, Koppiker CB, Kulkarni MD. Evaluation of tumor-infiltrating lymphocytes (TILs) in molecular subtypes of an Indian cohort of breast cancer patients. Diagn Pathol 2022; 17:91. [PMID: 36411483 PMCID: PMC9677664 DOI: 10.1186/s13000-022-01271-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/21/2022] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVES Evaluation of tumor-infiltrating lymphocytes (TILs) distribution in an Indian cohort of breast cancer patients for its prognostic significance. METHODS A retrospective cohort of breast cancer patients from a single onco-surgeon's breast cancer clinic with a uniform treatment strategy was evaluated for TILs. Tumor sections were H&E stained and scored for the spatial distribution and percent stromal TILs infiltration by a certified pathologist. The scores were analysed for association with treatment response and survival outcomes across molecular subtypes. RESULTS Total 229 breast cancer tumors were evaluated. Within spatial distribution categories, intra-tumoral TILs were observed to be associated with complete pathological response and lower recurrence frequency for the entire cohort. Subtype-wise analysis of stromal TILs (sTILs) re-enforced significantly higher infiltration in TNBC compared to HER2-positive and ER-positive tumors. A favourable association of higher stromal infiltration was observed with treatment response and disease outcomes, specifically in TNBC. CONCLUSION Intra-tumoral TILs showed a higher proportion with favourable association with better patient outcomes in an Indian cohort, unlike western cohorts where both stromal and intra-tumoral TILs show similar association with prognosis. With further validation, TILs can be developed as a cost-effective surrogate marker for treatment response, especially in a low-resource setting such as India.
Collapse
Affiliation(s)
- Pooja M. Vaid
- grid.417959.70000 0004 1764 2413Centre for Translational Cancer Research: a joint initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM), Pune, India ,grid.449178.70000 0004 5894 7096Department of Biological Sciences, Ashoka University, Sonipat, India
| | | | - Nutan S. Jumle
- grid.414967.90000 0004 1804 743XDepartment of Pathology, Jehangir Hospital, Pune, India
| | - Rituja A. Banale
- grid.417959.70000 0004 1764 2413Centre for Translational Cancer Research: a joint initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM), Pune, India ,grid.506045.20000 0004 4911 4105Prashanti Cancer Care Mission, Pune, Maharashtra India
| | - Danish Ansari
- grid.417959.70000 0004 1764 2413Centre for Translational Cancer Research: a joint initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM), Pune, India ,grid.506045.20000 0004 4911 4105Prashanti Cancer Care Mission, Pune, Maharashtra India
| | - Ruhi R. Reddy
- grid.417959.70000 0004 1764 2413Centre for Translational Cancer Research: a joint initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM), Pune, India ,grid.506045.20000 0004 4911 4105Prashanti Cancer Care Mission, Pune, Maharashtra India
| | - Rohini R. Unde
- grid.417959.70000 0004 1764 2413Centre for Translational Cancer Research: a joint initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM), Pune, India ,grid.506045.20000 0004 4911 4105Prashanti Cancer Care Mission, Pune, Maharashtra India
| | - Namrata P. Namewar
- grid.417959.70000 0004 1764 2413Centre for Translational Cancer Research: a joint initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM), Pune, India ,grid.506045.20000 0004 4911 4105Prashanti Cancer Care Mission, Pune, Maharashtra India
| | - Devaki A. Kelkar
- grid.417959.70000 0004 1764 2413Centre for Translational Cancer Research: a joint initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM), Pune, India ,grid.506045.20000 0004 4911 4105Prashanti Cancer Care Mission, Pune, Maharashtra India
| | - L. S. Shashidhara
- grid.417959.70000 0004 1764 2413Centre for Translational Cancer Research: a joint initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM), Pune, India ,grid.449178.70000 0004 5894 7096Department of Biological Sciences, Ashoka University, Sonipat, India ,grid.417959.70000 0004 1764 2413Indian Institute of Science Education and Research, Pune, India
| | - Chaitanyanand B. Koppiker
- grid.417959.70000 0004 1764 2413Centre for Translational Cancer Research: a joint initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM), Pune, India ,grid.506045.20000 0004 4911 4105Prashanti Cancer Care Mission, Pune, Maharashtra India
| | - Madhura D. Kulkarni
- grid.417959.70000 0004 1764 2413Centre for Translational Cancer Research: a joint initiative of Indian Institute of Science Education and Research (IISER) Pune and Prashanti Cancer Care Mission (PCCM), Pune, India ,grid.506045.20000 0004 4911 4105Prashanti Cancer Care Mission, Pune, Maharashtra India
| |
Collapse
|
49
|
Tumor suppressor DEAR1 regulates mammary epithelial cell fate and predicts early onset and metastasis in triple negative breast cancer. Sci Rep 2022; 12:19504. [PMID: 36376460 PMCID: PMC9663828 DOI: 10.1038/s41598-022-22417-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a disease of poor prognosis, with the majority classified as the basal-like subtype associated with epithelial-mesenchymal transition and metastasis. Because basal breast cancers originate from proliferative luminal progenitor-like cells upon dysregulation of proper luminal differentiation, genes regulating luminal-basal transition are critical to elucidate novel therapeutic targets to improve TNBC outcomes. Herein we demonstrate that the tumor suppressor DEAR1/TRIM62 is a critical regulator of luminal cell fate. DEAR1 loss in human mammary epithelial cells results in significantly enhanced mammosphere formation that is accelerated in the presence of TGF-β/SMAD3 signaling. Mammospheres formed following DEAR1 loss are enriched for ALDH1A1 and CK5 expression, EpCAM-/CD49f+ and CD44high/24low basal-like epithelial cells, indicating that DEAR1 regulates stem/progenitor cell properties and luminal-basal progenitor transition. We show that DEAR1 maintains luminal differentiation as a novel ubiquitin ligase for SNAI2/SLUG, a master regulator driving stemness and generation of basal-like progenitor populations. We also identify a significant inverse correlation between DEAR1 and SNAI2 expression in a 103 TNBC case cohort and show that low DEAR1 expression significantly correlates with young age of onset and shorter time to metastasis, suggesting DEAR1 could serve as a biomarker to stratify early onset TNBCs for targeted stem cell therapies.
Collapse
|
50
|
Yang H, Karl MN, Wang W, Starich B, Tan H, Kiemen A, Pucsek AB, Kuo YH, Russo GC, Pan T, Jaffee EM, Fertig EJ, Wirtz D, Spangler JB. Engineered bispecific antibodies targeting the interleukin-6 and -8 receptors potently inhibit cancer cell migration and tumor metastasis. Mol Ther 2022; 30:3430-3449. [PMID: 35841152 PMCID: PMC9637575 DOI: 10.1016/j.ymthe.2022.07.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 06/12/2022] [Accepted: 07/09/2022] [Indexed: 12/15/2022] Open
Abstract
Simultaneous inhibition of interleukin-6 (IL-6) and interleukin-8 (IL-8) signaling diminishes cancer cell migration, and combination therapy has recently been shown to synergistically reduce metastatic burden in a preclinical model of triple-negative breast cancer. Here, we have engineered two novel bispecific antibodies that target the IL-6 and IL-8 receptors to concurrently block the signaling activity of both ligands. We demonstrate that a first-in-class bispecific antibody design has promising therapeutic potential, with enhanced selectivity and potency compared with monoclonal antibody and small-molecule drug combinations in both cellular and animal models of metastatic triple-negative breast cancer. Mechanistic characterization revealed that our engineered bispecific antibodies have no impact on cell viability, but profoundly reduce the migratory potential of cancer cells; hence they constitute a true anti-metastatic treatment. Moreover, we demonstrate that our antibodies can be readily combined with standard-of-care anti-proliferative drugs to develop effective anti-cancer regimens. Collectively, our work establishes an innovative metastasis-focused direction for cancer drug development.
Collapse
Affiliation(s)
- Huilin Yang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Michelle N Karl
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Nano Biotechnology (INBT), the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Wentao Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Bartholomew Starich
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Nano Biotechnology (INBT), the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Haotian Tan
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Nano Biotechnology (INBT), the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ashley Kiemen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Nano Biotechnology (INBT), the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Alexandra B Pucsek
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yun-Huai Kuo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Gabriella C Russo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Nano Biotechnology (INBT), the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Tim Pan
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Nano Biotechnology (INBT), the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Elizabeth M Jaffee
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Sidney Kimmel Cancer Center, the Johns Hopkins University, Baltimore, MD 21231, USA
| | - Elana J Fertig
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Sidney Kimmel Cancer Center, the Johns Hopkins University, Baltimore, MD 21231, USA; Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD 21218, USA; Convergence Institute, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Nano Biotechnology (INBT), the Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Sidney Kimmel Cancer Center, the Johns Hopkins University, Baltimore, MD 21231, USA
| | - Jamie B Spangler
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Sidney Kimmel Cancer Center, the Johns Hopkins University, Baltimore, MD 21231, USA; Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|