1
|
Wang Y, Liu Z, Lv Y, Long J, Lu Y, Huang P. Mechanisms of radioresistance and radiosensitization strategies for Triple Negative Breast Cancer. Transl Oncol 2025; 55:102351. [PMID: 40112501 PMCID: PMC11964565 DOI: 10.1016/j.tranon.2025.102351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
Breast cancer is one of the most common malignant tumors in women. Triple-negative breast cancer (TNBC) is a molecular subtype of breast cancer that is characterized by a high risk of recurrence and poor prognosis. With the increasingly prominent role of radiotherapy in TNBC treatment, patient resistance to radiotherapy is an attractive area of clinical research. Gene expression changes induced by multiple mechanisms can affect the radiosensitivity of TNBC cells to radiotherapy through a variety of ways, and the enhancement of radioresistance is an important factor in the malignant progression of TNBC. The above pathways mainly include DNA damage repair, programmed cell death, cancer stem cells (CSC), antioxidant function, tumor microenvironment, and epithelial-mesenchymal transition (EMT) pathway. Tumor cells can reduce the damage of radiotherapy to themselves through the above ways, resulting in radioresistance. Therefore, in this review, we aim to summarize the strategies for immunotherapy combined with radiotherapy, targeted therapy combined with radiotherapy, and epigenetic therapy combined with radiotherapy to identify the best treatment for TNBC and improve the cure and survival rates of patients with TNBC. This review will provide important guidance and inspiration for the clinical practice of radiotherapy for TNBC, which will help deepen our understanding of this field and promote its development.
Collapse
Affiliation(s)
- Yuxuan Wang
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Zhiwei Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Yulu Lv
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Jiayang Long
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Yao Lu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Panpan Huang
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| |
Collapse
|
2
|
Emara HM, Allam NK, Youness RA. A comprehensive review on targeted therapies for triple negative breast cancer: an evidence-based treatment guideline. Discov Oncol 2025; 16:547. [PMID: 40244488 PMCID: PMC12006628 DOI: 10.1007/s12672-025-02227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive malignancy characterized by limited therapeutic options and poor prognosis. Despite advancements in precision oncology, conventional chemotherapy remains the cornerstone of TNBC treatment, often accompanied by debilitating side effects and suboptimal outcomes. This review presents a comprehensive analysis of clinical trials on targeted therapies, aiming to establish a novel, evidence-based treatment strategy exclusively leveraging molecularly targeted agents. By integrating patient-specific genetic profiles with therapeutic responses observed across various clinical trial phases, this approach seeks to optimize efficacy while minimizing toxicity. The proposed targeted therapy combinations hold significant potential to revolutionize TNBC treatment, offering a paradigm shift toward precision medicine and improved patient outcomes.
Collapse
Affiliation(s)
- Hadir M Emara
- Nanotechnology Program, School of Sciences & Engineering, The American University in Cairo, New Cairo, 11835, Egypt.
| | - Nageh K Allam
- Nanotechnology Program, School of Sciences & Engineering, The American University in Cairo, New Cairo, 11835, Egypt.
- Energy Materials Laboratory, Physics Department, School of Sciences & Engineering, The American University in Cairo, New Cairo, 11835, Egypt.
| | - Rana A Youness
- Department of Molecular Biology and Biochemistry, Faculty of Biotechnology, German International University, New Administrative Capital, Cairo, Egypt.
| |
Collapse
|
3
|
Zhu M, Liu Y, Wen Z, Tan H, Li S, Yu X, Luo H, Li D, Wang J, Qin F. Exploration of Traditional Chinese Medicine Comprehensive Treatment of Triple Negative Breast Cancer Based on Molecular Pathological Mechanism. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:289-304. [PMID: 40236879 PMCID: PMC11998019 DOI: 10.2147/bctt.s511059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/20/2025] [Indexed: 04/17/2025]
Abstract
Triple-negative breast cancer (TNBC) is recognized as the most aggressive subtype of breast cancer and is associated with poor prognosis. Clinically, TNBC is associated with significant invasiveness, high propensity for metastasis, frequent recurrence, and unfavorable outcomes. The absence of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2 (HER2) in TNBC renders it unresponsive to endocrine therapies and treatments that target HER2. Consequently, the current therapeutic options are primarily confined to surgical intervention, adjuvant chemotherapy, and radiotherapy. Given the considerable heterogeneity of TNBC, targeted therapies have emerged as promising avenues for treatment. Furthermore, immunotherapy has demonstrated the potential to enhance overall survival and therapeutic response in patients with TNBC. Additionally, research indicates that traditional Chinese medicine (TCM) may yield beneficial effects in the management of this cancer subtype. This review aims to consolidate recent advancements in treatment strategies for TNBC, particularly those based on molecular subtypes.
Collapse
Affiliation(s)
- Mingya Zhu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Yongqin Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Zhu Wen
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Hao Tan
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Siman Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Xinkang Yu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Hongping Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Delin Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Jinyan Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Fangyan Qin
- The Second People’s Hospital of Jiangjin Chongqing, Chongqing, People’s Republic of China
| |
Collapse
|
4
|
Shaik R, Mounika V, Begum S, Rajkumar A, Mallikarjun B, Sri Harshini V, Kolure R, Sreevani B, Thakur S. Monoclonal Antibodies in Clinical Trials for Breast Cancer Treatment. Monoclon Antib Immunodiagn Immunother 2025; 44:17-39. [PMID: 40171653 DOI: 10.1089/mab.2024.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025] Open
Abstract
One of the most potent therapeutic and diagnostic agents in contemporary medicine is the monoclonal antibody (mAb). mAbs can perform a variety of tasks in breast cancer (BC), including identifying and delivering therapeutic medications to targets, preventing cell development, and suppressing immune system inhibitors including directly attacking cancer cells. mAbs are one of the most effective therapeutic options, particularly for HER2, but they have not been well studied for their use in treating other forms of BC, particularly triple negative breast tumors. Bispecific and trispecific mAbs have created new opportunities for more targeted specific efficacy, which has a positive impact on the viability of antigen specificity. They are more versatile and effective than other forms of treatment, emerging as most popular option for treating BC. However, mAbs have a limit in treatment due to certain adverse effects, including fever, shaking, exhaustion, headache, nausea, and vomiting, as well as rashes, bleeding, and difficulty breathing. To examine the current and prospective future capacities of mAbs with regard to the detection and treatment of BC, the present review highlights advantages and disadvantages of mAb approach.
Collapse
Affiliation(s)
- Rahaman Shaik
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Varikuppala Mounika
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Shireen Begum
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Agolapu Rajkumar
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Bathurasi Mallikarjun
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Vollala Sri Harshini
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Rajini Kolure
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | | | - Sneha Thakur
- Department of Pharmacognosy, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| |
Collapse
|
5
|
Khan Y, Rizvi S, Raza A, Khan A, Hussain S, Khan NU, Alshammari SO, Alshammari QA, Alshammari A, Ellakwa DES. Tailored therapies for triple-negative breast cancer: current landscape and future perceptions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03896-4. [PMID: 40029385 DOI: 10.1007/s00210-025-03896-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/07/2025] [Indexed: 03/05/2025]
Abstract
Triple-negative breast cancer (TNBC) has become one of the most challenging cancers to date due to its great variability in biological features, high growth rate, and rare options for treatment. This review examines several innovative strategies for tailored treatment of TNBC, focusing mainly on the most recent developments and potential directions. The molecular landscape of TNBC is covered in the first section, which keeps the focus on transcriptome and genomic profiling while highlighting key molecular targets like mutations in the BRCA1/2, PIK3CA, androgen receptors (AR), epidermal growth factor receptors (EGFR), and immunological checkpoint molecules. This review also covers novel therapies that aim to block well-defined pathways, including immune checkpoint inhibitors (ICI), EGFR inhibitors, drugs that target AR, poly ADP ribose polymerase (PARP) inhibitors, and drugs that disrupt the PI3K/AKT/mTOR pathway. Additionally, it covers novel strategies focusing on combination therapy, targeting the DNA damage response pathway, and epigenetic modulators. Conclusively, it emphasizes perspectives and directions on topics such as personalized medicine, artificial intelligence (AI), predictive biomarkers, and treatment planning with the inclusion of machine learning (ML).
Collapse
Affiliation(s)
- Yumna Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, 25130, Pakistan.
| | - Sana Rizvi
- Bakhtawar Amin Medical and Dental College, Bakhtawar Amin Trust Teaching Hospital, Multan, Pakistan
| | - Ali Raza
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Amna Khan
- Abbottabad International Medical Institute, Abbottabad, 22020, Pakistan
| | - Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248007, India
| | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, 25130, Pakistan
| | - Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, College of Pharmacy, Northern Border University, 76321, Rafha, Saudi Arabia
| | - Qamar A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Abdulkarim Alshammari
- Department of Clinical Practice, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Doha El-Sayed Ellakwa
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo, Egypt.
- Department of Biochemistry, Faculty of Pharmacy, Sinai University, Kantra Branch, Ismailia, Egypt.
| |
Collapse
|
6
|
Yang X, Huang K, Wu XN, Zhang C, Sun Y, Gao Y, Zhou J, Tao L, Zhang H, Wu Y, Luo HB, Wang H. Discovery of a Novel Selective and Cell-Active N 6-Methyladenosine RNA Demethylase ALKBH5 Inhibitor. J Med Chem 2025; 68:4133-4147. [PMID: 39925002 DOI: 10.1021/acs.jmedchem.4c01542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
N6-methyladenosine (m6A), the most abundant methylation on mRNA, plays pivotal roles in regulating mRNA biological functions, which affect cell functions. ALKBH5, an m6A demethylase, was found to be an oncogene in several cancer types, including triple-negative breast cancer (TNBC). Here, we report a novel and selective ALKBH5 covalent inhibitor, W23-1006, through virtual screening and structure optimization. It covalently bonds to the ALKBH5 C200 residue with an IC50 value of 3.848 μM, representing roughly 30- and 8-fold stronger inhibitory activity than that against FTO and ALKBH3, respectively. Cellular experiments demonstrated that W23-1006 could efficiently enhance the m6A level on fibronectin 1 (FN1) mRNA, leading to strong suppression of TNBC cell proliferation and migration in vitro as well as tumor growth and metastasis in vivo. Collectively, our study developed a novel, selective, and cell-active ALKBH5 covalent inhibitor, W23-1006, which could be a potential therapeutic option for cancer, such as TNBC treatment.
Collapse
Affiliation(s)
- Xianyuan Yang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Kaitao Huang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xu-Nian Wu
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Chen Zhang
- School of Chemistry and Chemical Engineering and Guangdong Cosmetics Engineering and Technology Research Center, Guangdong Pharmaceutical University, Zhongshan, Guangdong 528458, China
| | - Yixuan Sun
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen Campus, Shenzhen 518107, China
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen Campus, Shenzhen 518107, China
| | - Jiawang Zhou
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Lijun Tao
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Haisheng Zhang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yinuo Wu
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hai-Bin Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education and Hainan Engineering Research Center for Drug Screening and Evaluation, School of Pharmaceutical Sciences, Hainan University, Haikou, Hainan 570228, China
| | - Hongsheng Wang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
7
|
Madorsky Rowdo FP, Martini R, Ackermann SE, Tang CP, Tranquille M, Irizarry A, Us I, Alawa O, Moyer JE, Sigouros M, Nguyen J, Assaad MA, Cheng E, Ginter PS, Manohar J, Stonaker B, Boateng R, Oppong JK, Adjei EK, Awuah B, Kyei I, Aitpillah FS, Adinku MO, Ankomah K, Osei-Bonsu EB, Gyan KK, Hoda S, Newman L, Mosquera JM, Sboner A, Elemento O, Dow LE, Davis MB, Martin ML. Kinome-Focused CRISPR-Cas9 Screens in African Ancestry Patient-Derived Breast Cancer Organoids Identify Essential Kinases and Synergy of EGFR and FGFR1 Inhibition. Cancer Res 2025; 85:551-566. [PMID: 39891928 PMCID: PMC11790258 DOI: 10.1158/0008-5472.can-24-0775] [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: 03/08/2024] [Revised: 08/10/2024] [Accepted: 11/20/2024] [Indexed: 02/03/2025]
Abstract
Precision medicine approaches to cancer treatment aim to exploit genomic alterations that are specific to individual patients to tailor therapeutic strategies. Yet, some targetable genes and pathways are essential for tumor cell viability even in the absence of direct genomic alterations. In underrepresented populations, the mutational landscape and determinants of response to existing therapies are poorly characterized because of limited inclusion in clinical trials and studies. One way to reveal tumor essential genes is with genetic screens. Most screens are conducted on cell lines that bear little resemblance to patient tumors, after years of culture under nonphysiologic conditions. To address this problem, we aimed to develop a CRISPR screening pipeline in three-dimensionally grown patient-derived tumor organoid (PDTO) models. A breast cancer PDTO biobank that focused on underrepresented populations, including West African patients, was established and used to conduct a negative-selection kinome-focused CRISPR screen to identify kinases essential for organoid growth and potential targets for combination therapy with EGFR or MEK inhibitors. The screen identified several previously unidentified kinase targets, and the combination of FGFR1 and EGFR inhibitors synergized to block organoid proliferation. Together, these data demonstrate the feasibility of CRISPR-based genetic screens in patient-derived tumor models, including PDTOs from underrepresented patients with cancer, and identify targets for cancer therapy. Significance: Generation of a breast cancer patient-derived tumor organoid biobank focused on underrepresented populations enabled kinome-focused CRISPR screening that identified essential kinases and potential targets for combination therapy with EGFR or MEK inhibitors. See related commentary by Trembath and Spanheimer, p. 407.
Collapse
Affiliation(s)
| | - Rachel Martini
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
- Institute of Translational Genomic Medicine, Morehouse School of Medicine, GA, USA
| | - Sarah E. Ackermann
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Colin P. Tang
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Marvel Tranquille
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Adriana Irizarry
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ilkay Us
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Omar Alawa
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jenna E. Moyer
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Michael Sigouros
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - John Nguyen
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Majd Al Assaad
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Esther Cheng
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Paula S. Ginter
- Department of Pathology, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Jyothi Manohar
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Brian Stonaker
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | | | | | | | | | - Ishmael Kyei
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | | | | | | | | | - Kofi K. Gyan
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Syed Hoda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lisa Newman
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Juan Miguel Mosquera
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Andrea Sboner
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Lukas E. Dow
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY, USA
| | - Melissa B. Davis
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
- Institute of Translational Genomic Medicine, Morehouse School of Medicine, GA, USA
| | - M. Laura Martin
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
8
|
Escoto A, Hecksel R, Parkinson C, Crane S, Atwell B, King S, Ortiz Chavez D, Jannuzi A, Sands B, Bitler BG, Fehniger TA, Paek AL, Padi M, Schroeder J. Nuclear EGFR in breast cancer suppresses NK cell recruitment and cytotoxicity. Oncogene 2025; 44:288-295. [PMID: 39521886 PMCID: PMC11779631 DOI: 10.1038/s41388-024-03211-0] [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: 04/30/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Natural Killer (NK) cells can target and destroy cancer cells, yet tumor microenvironments typically suppress NK cell recruitment and cytotoxicity. The epidermal growth factor receptor (EGFR) is a potent oncogene that can activate survival, migration, and proliferation pathways, and clinical data suggests it may also play an immunomodulating role in cancers. Recent work has demonstrated a novel role for nuclear EGFR (nEGFR) in regulating transcriptional events unique from the kinase domain. Using a novel peptide therapeutic (cSNX1.3) that inhibits retrograde trafficking of EGFR and an EGFR nuclear localization mutant, we discovered that nEGFR suppresses NK cell recruitment and cytotoxicity. RNA-Seq analysis of breast cancer cells treated with cSNX1.3 or modified to lack a nuclear localization sequence (EGFRΔNLS) revealed the EGF-dependent induction of NK activating receptor ligands, while kinase inhibition by erlotinib did not impact these genes. NanoString analysis of tumor-bearing WAP-TGFα transgenic mice treated with cSNX1.3 demonstrated an increase in immune cell populations and activating genes. Additionally, immunohistochemistry confirmed an increase in NK cells upon cSNX1.3 treatment. Finally, cSNX1.3 treatment was found to enhance NK cell recruitment and cytotoxicity in vitro. Together, the data demonstrate a unique immunomodulatory role for nEGFR.
Collapse
Affiliation(s)
- Angelica Escoto
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Ryan Hecksel
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Chance Parkinson
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Sara Crane
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Benjamin Atwell
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Shyanne King
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Daniela Ortiz Chavez
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Alison Jannuzi
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Barbara Sands
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Benjamin G Bitler
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Andrew L Paek
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
- University of Arizona Cancer Center, Tucson, AZ, 85721, USA
| | - Megha Padi
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA.
- University of Arizona Cancer Center, Tucson, AZ, 85721, USA.
| | - Joyce Schroeder
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA.
- University of Arizona Cancer Center, Tucson, AZ, 85721, USA.
- BIO5 Institute, Tucson, AZ, 85721, USA.
| |
Collapse
|
9
|
Anjum F, Kaushik K, Salam A, Nandi CK. Chromatin Marks H3K4me3 and H3K9me3 in Triple-Negative Breast Cancer Cell Lines. Adv Biol (Weinh) 2025:e2400752. [PMID: 39841002 DOI: 10.1002/adbi.202400752] [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: 11/27/2024] [Revised: 01/03/2025] [Indexed: 01/23/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most lethal and aggressive breast cancer among all the breast cancer subtypes. Despite several attempts, to date, there is an extensive lack of therapeutic intervention. Hence, there is a dire need for an effective biomarker to timely diagnose TNBC. Here, utilizing super-resolution microscopy, the remodeling structural aspects of euchromatin and heterochromatin in TNBC are studied and the results are compared with non-cancerous and non-TNBC cell lines. The nanoscopic visualization reveals a distinct difference in chromatin remodeling in TNBC in comparison to the other two cell lines. While the euchromatin density is found to increase, the heterochromatin is found to decrease. A complete switching of the heterochromatin-euchromatin ratio is observed in TNBC cells thus proposing that chromatin remodeling and chromatin morphological changes can be pursued as one of the targets for diagnostic purposes. Increased expression of structure specific recognition protein-1(SSRP-1) protein supports the increased rate of chromatin remodeling in breast cancer cell lines. The results may lead to developing a new strategy for diagnosing TNBC patients.
Collapse
Affiliation(s)
- Farhan Anjum
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| | - Kush Kaushik
- School of Chemical Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| | - Abdul Salam
- School of Chemical Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| | - Chayan Kanti Nandi
- School of Chemical Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| |
Collapse
|
10
|
Sharma N, Bhati A, Aggarwal S, Shah K, Dewangan HK. PARP Pioneers: Using BRCA1/2 Mutation-targeted Inhibition to Revolutionize Breast Cancer Treatment. Curr Pharm Des 2025; 31:663-673. [PMID: 39421986 DOI: 10.2174/0113816128322894241004051814] [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: 05/06/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Breast cancer stands on the second position in the world in being common and women happen to have it with high rate of about five-folds around the world. The causes of occurrence can matter with different humans be it external factors or the internal genetic ones. Breast cancer is primarily driven by mutations in the BRCA1 and BRCA2 susceptibility genes. These BC susceptibility genes encode proteins critical for DNA homologous recombination repair (HRR). Poly (ADP ribose) polymerases (PARP) are the essential enzymes involved in the repairing of the damaged DNA. So the inhibition of these inhibitors can be considered as the promising strategy for targeting cancers with defective damage in the deoxyribonucleic acid. Olaparib and talazoparib are PARP inhibitors (PARPi) are being employed for the monotherapies in case of the deleterious germline HER2-negative and BRCA-mutated breast cancer. The potency of PARP for trapping on DNA and causes cytotoxicity may have difference in the safety and efficacy with the PARPi. The PARPi have been found its place in the all different types of breast cancers and have shown potential benefits. The purpose of this review is to provide an update on the oral poly (ADP-ribose) polymerase (PARP) inhibitors for the improvement in the treatment and management of breast cancer.
Collapse
Affiliation(s)
- Navneet Sharma
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| | - Akash Bhati
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| | - Shagun Aggarwal
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| | - Kamal Shah
- Institute of Pharmaceutical Research (IPR), GLA University, NH-2, Delhi Mathura Road, PO-Chaumuhan, Mathura, Uttar Pradesh, India
| | - Hitesh Kumar Dewangan
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| |
Collapse
|
11
|
Vishnubalaji R, Alajez NM. Disrupted Lipid Metabolism, Cytokine Signaling, and Dormancy: Hallmarks of Doxorubicin-Resistant Triple-Negative Breast Cancer Models. Cancers (Basel) 2024; 16:4273. [PMID: 39766172 PMCID: PMC11674486 DOI: 10.3390/cancers16244273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/30/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chemoresistance in triple-negative breast cancer (TNBC) presents a significant clinical hurdle, limiting the efficacy of treatments like doxorubicin. This study aimed to explore the molecular changes associated with doxorubicin resistance and identify potential therapeutic targets to overcome this resistance, thereby improving treatment outcomes for TNBC patients. METHODS Doxorubicin-resistant (DoxR) TNBC models (MDA-MB-231 and BT-549) were generated by exposing cells to increasing concentrations of doxorubicin. RNA sequencing (RNA-Seq) was performed using the Illumina platform, followed by bioinformatics analysis with CLC Genomics Workbench and iDEP. Functional assays assessed proliferation, sphere formation, migration, and cell cycle changes. Protein expression and phosphorylation were confirmed via Western blotting. Pathway and network analyses were conducted using Ingenuity Pathway Analysis (IPA) and STRING, while survival analysis was performed using Kaplan-Meier Plotter database. RESULTS DoxR cells exhibited reduced proliferation, sphere formation, and migration, but showed enhanced tolerance to doxorubicin. Increased CHK2 and p53 phosphorylation indicated cellular dormancy as a resistance mechanism. RNA-Seq analysis revealed upregulation of cytokine signaling and stress-response pathways, while cholesterol and lipid biosynthesis were suppressed. Activation of the IL1β cytokine network was prominent in DoxR cells, and CRISPR-Cas9 screens data identified dependencies on genes involved in rRNA biogenesis and metabolism. A 27-gene signature associated with doxorubicin resistance was linked to worse clinical outcomes in a large breast cancer cohort (HR = 1.76, FDR p < 2.0 × 10-13). CONCLUSIONS This study uncovers potential therapeutic strategies for overcoming TNBC resistance, including dormancy reversal and targeting onco-ribosomal pathways and cytokine signaling networks, to improve the efficacy of doxorubicin-based treatments.
Collapse
Affiliation(s)
- Radhakrishnan Vishnubalaji
- Translational Oncology Research Center (TORC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar;
| | - Nehad M. Alajez
- Translational Oncology Research Center (TORC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar;
- College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| |
Collapse
|
12
|
Mehta K, Hegde M, Girisa S, Vishwa R, Alqahtani MS, Abbas M, Shakibaei M, Sethi G, Kunnumakkara AB. Targeting RTKs/nRTKs as promising therapeutic strategies for the treatment of triple-negative breast cancer: evidence from clinical trials. Mil Med Res 2024; 11:76. [PMID: 39668367 PMCID: PMC11636053 DOI: 10.1186/s40779-024-00582-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: 10/19/2023] [Accepted: 11/08/2024] [Indexed: 12/14/2024] Open
Abstract
The extensive heterogeneity and the limited availability of effective targeted therapies contribute to the challenging prognosis and restricted survival observed in triple-negative breast cancer (TNBC). Recent research indicates the aberrant expression of diverse tyrosine kinases (TKs) within this cancer, contributing significantly to tumor cell proliferation, survival, invasion, and migration. The contemporary paradigm shift towards precision medicine has highlighted TKs and their receptors as promising targets for pharmacotherapy against a range of malignancies, given their pivotal roles in tumor initiation, progression, and advancement. Intensive investigations have focused on various monoclonal antibodies (mAbs) and small molecule inhibitors that specifically target proteins such as epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor (VEGFR), cellular mesenchymal-epithelial transition factor (c-MET), human epidermal growth factor receptor 2 (HER2), among others, for combating TNBC. These agents have been studied both in monotherapy and in combination with other chemotherapeutic agents. Despite these advances, a substantial terrain of unexplored potential lies within the realm of TK targeted therapeutics, which hold promise in reshaping the therapeutic landscape. This review summarizes the various TK targeted therapeutics that have undergone scrutiny as potential therapeutic interventions for TNBC, dissecting the outcomes and revelations stemming from diverse clinical investigations. A key conclusion from the umbrella clinical trials evidences the necessity for in-depth molecular characterization of TNBCs for the maximum efficiency of TK targeted therapeutics, either as standalone treatments or a combination. Moreover, our observation highlights that the outcomes of TK targeted therapeutics in TNBC are substantially influenced by the diversity of the patient cohort, emphasizing the prioritization of individual patient genetic/molecular profiles for precise TNBC patient stratification for clinical studies.
Collapse
Affiliation(s)
- Kasshish Mehta
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Ravichandran Vishwa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, 61421, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, 61421, Abha, Saudi Arabia
| | - Mehdi Shakibaei
- Department of Human-Anatomy, Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Ludwig-Maximilian-University, 80336, Munich, Germany
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India.
| |
Collapse
|
13
|
Jin LL, Lu HJ, Shao JK, Wang Y, Lu SP, Huang BF, Hu GN, Jin HC, Wang CQ. Relevance and mechanism of STAT3/miR-221-3p/Fascin-1 axis in EGFR TKI resistance of triple-negative breast cancer. Mol Cell Biochem 2024; 479:3037-3047. [PMID: 38145448 DOI: 10.1007/s11010-023-04907-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/25/2023] [Indexed: 12/26/2023]
Abstract
The epidermal growth factor receptor 1 (EGFR) plays a crucial role in the progression of various malignant tumors and is considered a potential target for treating triple-negative breast cancer (TNBC). However, the effectiveness of representative tyrosine kinase inhibitors (TKIs) used in EGFR-targeted therapy is limited in TNBC patients. In our study, we observed that the TNBC cell lines MDA-MB-231 and MDA-MB-468 exhibited resistance to Gefitinib. Treatment with Gefitinib caused an upregulation of Fascin-1 (FSCN1) protein expression and a downregulation of miR-221-3p in these cell lines. However, sensitivity to Gefitinib was significantly improved in both cell lines with either inhibition of FSCN1 expression or overexpression of miR-221-3p. Our luciferase reporter assay confirmed that FSCN1 is a target of miR-221-3p. Moreover, Gefitinib treatment resulted in an upregulation of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) in MDA-MB-231 cells. Using Stattic, a small-molecule inhibitor of STAT3, we observed a significant enhancement in the inhibitory effect of Gefitinib on the growth, migration, and invasion of MDA-MB-231 cells. Additionally, Stattic treatment upregulated miR-221-3p expression and downregulated FSCN1 mRNA and protein expression. A strong positive correlation was noted between the expression of STAT3 and FSCN1 in breast cancer tissues. Furthermore, patients with high expression levels of both STAT3 and FSCN1 had a worse prognosis. Our findings suggest that elevated FSCN1 expression is linked to primary resistance to EGFR TKIs in TNBC. Moreover, we propose that STAT3 regulates the expression of miR-221-3p/FSCN1 and therefore modulates resistance to EGFR TKI therapy in TNBC. Combining EGFR TKI therapy with inhibition of FSCN1 or STAT3 may offer a promising new therapeutic option for TNBC.
Collapse
Affiliation(s)
- Lu-Lu Jin
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Hua-Jun Lu
- Department of Oncological Radiotherapy, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Jun-Kang Shao
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, 60 Wu Ning Xi Road, Dongyang, Zhejiang, China
| | - Yan Wang
- Department of Medical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Shi-Ping Lu
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, 60 Wu Ning Xi Road, Dongyang, Zhejiang, China
| | - Bi-Fei Huang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, 60 Wu Ning Xi Road, Dongyang, Zhejiang, China
| | - Gui-Nv Hu
- Department of Surgical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Hong-Chuan Jin
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang Province, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, Zhejiang, China
| | - Chao-Qun Wang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, 60 Wu Ning Xi Road, Dongyang, Zhejiang, China.
| |
Collapse
|
14
|
Kang JH, Uddin N, Kim S, Zhao Y, Yoo KC, Kim MJ, Hong SA, Bae S, Lee JY, Shin I, Jin YW, O'Hagan HM, Yi JM, Lee SJ. Tumor-intrinsic role of ICAM-1 in driving metastatic progression of triple-negative breast cancer through direct interaction with EGFR. Mol Cancer 2024; 23:230. [PMID: 39415210 PMCID: PMC11481280 DOI: 10.1186/s12943-024-02150-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
Triple-negative breast cancer (TNBC), the most aggressive subtype, presents a critical challenge due to the absence of approved targeted therapies. Hence, there is an urgent need to identify effective therapeutic targets for this condition. While epidermal growth factor receptor (EGFR) is prominently expressed in TNBC and recognized as a therapeutic target, anti-EGFR therapies have yet to gain approval for breast cancer treatment due to their associated side effects and limited efficacy. Here, we discovered that intercellular adhesion molecule-1 (ICAM-1) exhibits elevated expression levels in metastatic breast cancer and serves as a pivotal binding adaptor for EGFR activation, playing a crucial role in malignant progression. The activation of EGFR by tumor-expressed ICAM-1 initiates biased signaling within the JAK1/STAT3 pathway, consequently driving epithelial-to-mesenchymal transition and facilitating heightened metastasis without influencing tumor growth. Remarkably, ICAM-1-neutralizing antibody treatment significantly suppressed cancer metastasis in a breast cancer orthotopic xenograft mouse model. In conclusion, our identification of ICAM-1 as a novel tumor intrinsic regulator of EGFR activation offers valuable insights for the development of TNBC-specific anti-EGFR therapies.
Collapse
Affiliation(s)
- Jae-Hyeok Kang
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Nizam Uddin
- Center for Cell Analysis & Modeling, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Seungmo Kim
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Yi Zhao
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Ki-Chun Yoo
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Min-Jung Kim
- Fibrosis and Cancer Targeting Biotechnology (FNCT BIOTECH), Toegye-Ro 36 Gil, Seoul, 04626, South Korea
| | - Sung-Ah Hong
- Genomic Medicine Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Sangsu Bae
- Department of Biochemistry and Molecular Biology, College of Medicine, Seoul National University, Seoul, 03080, South Korea
| | - Jeong-Yeon Lee
- Department of Pathology, College of Medicine, Hanyang University, Seoul, 04763, South Korea
| | - Incheol Shin
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Young Woo Jin
- Fibrosis and Cancer Targeting Biotechnology (FNCT BIOTECH), Toegye-Ro 36 Gil, Seoul, 04626, South Korea
| | - Heather M O'Hagan
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Joo Mi Yi
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Bloomington, IN, 47405, USA.
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan, 47392, South Korea.
| | - Su-Jae Lee
- Fibrosis and Cancer Targeting Biotechnology (FNCT BIOTECH), Toegye-Ro 36 Gil, Seoul, 04626, South Korea.
| |
Collapse
|
15
|
Hong L, Tanaka M, Yasui M, Hara-Chikuma M. HSP90 promotes tumor associated macrophage differentiation during triple-negative breast cancer progression. Sci Rep 2024; 14:22541. [PMID: 39341960 PMCID: PMC11438890 DOI: 10.1038/s41598-024-73394-9] [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: 04/04/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Tumor-associated macrophages (TAMs) originating from monocytes are crucial for cancer progression; however, the mechanism of TAM differentiation is unclear. We investigated factors involved in the differentiation of monocytes into TAMs within the tumor microenvironment of triple-negative breast cancer (TNBC). We screened 172 compounds and found that a heat shock protein 90 (HSP90) inhibitor blocked TNBC-induced monocyte-to-TAM differentiation in human monocytes THP-1. TNBC-derived conditional medium (CM) activated cell signaling pathways, including MAP kinase, AKT and STAT3, and increased the expression of TAM-related genes and proteins. These inductions were suppressed by HSP90 inhibition or by knockdown of HSP90 in TNBC. Additionally, we confirmed that TNBC secreted HSP90 extracellularly and that HSP90 itself promoted TAM differentiation. In a mouse tumor model, treatment with an HSP90 inhibitor suppressed tumor growth and reduced TAMs in the tumor microenvironment. Our findings demonstrate the role of HSP90 in TAM differentiation, suggesting HSP90 as a potential target for TNBC immunotherapy due to its regulatory role in monocyte-to-TAM differentiation.
Collapse
Affiliation(s)
- Lingjia Hong
- Department of Pharmacology, School of Medicine, Keio University, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Manami Tanaka
- Department of Pharmacology, School of Medicine, Keio University, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masato Yasui
- Department of Pharmacology, School of Medicine, Keio University, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Mariko Hara-Chikuma
- Department of Pharmacology, School of Medicine, Keio University, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
16
|
R RS. Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer: Impact on Early Stage and Outcomes-Comprehensive Review. Indian J Surg Oncol 2024; 15:501-508. [PMID: 39328729 PMCID: PMC11422321 DOI: 10.1007/s13193-024-02032-7] [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: 12/26/2023] [Accepted: 07/16/2024] [Indexed: 09/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) poses a significant challenge in clinical oncology due to its aggressive nature and limited targeted therapeutic options. Neoadjuvant chemotherapy (NACT) has emerged as a promising strategy in the management of early-stage TNBC. This literature review aims to provide an in-depth analysis of the role of NACT in TNBC, focusing on its impact on early-stage disease and associated outcomes. The review synthesizes evidence from recent studies, clinical trials, and meta-analyses to present a comprehensive overview of the current landscape of NACT in early-stage TNBC.
Collapse
Affiliation(s)
- Raxith Sringeri R
- Department of Surgical Oncology and Robotic Surgery, HCG Bharath Cancer Hospital, Mysore, India
| |
Collapse
|
17
|
Dai S, Liu Y, Liu Z, Li R, Luo F, Li Y, Dai L, Peng X. Cancer-associated fibroblasts mediate resistance to anti-EGFR therapies in cancer. Pharmacol Res 2024; 206:107304. [PMID: 39002870 DOI: 10.1016/j.phrs.2024.107304] [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: 05/16/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Over the last decade, epidermal growth factor receptor (EGFR)-targeted therapies have transformed the treatment landscape for patients with advanced solid tumors. Despite these advances, resistance to anti-EGFR therapies is still a significant clinical challenge. While cell-autonomous mechanisms of resistance are well-documented, they do not fully elucidate the complexity of drug resistance. Cancer-associated fibroblasts (CAFs), key mediators within the tumor microenvironment (TME), have emerged as pivotal players in cancer progression and chemoresistance. Recent evidence implicates CAFs in resistance to anti-EGFR therapies, suggesting they may undermine treatment efficacy. This review synthesizes current data, highlighting the critical role of CAFs in resistance pathogenesis and summarizing recent therapeutic strategies targeting CAFs. We underscore the challenges and advocate for the exploration of CAFs as a potential dual-targeted approach.
Collapse
Affiliation(s)
- Shuang Dai
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingtong Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610041, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu Sichuan, China
| | - Ruidan Li
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu Sichuan, China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu Sichuan, China.
| |
Collapse
|
18
|
Hammershøi Madsen AM, Løvendahl Eefsen RH, Nielsen D, Kümler I. Targeted Treatment of Metastatic Triple-Negative Breast Cancer: A Systematic Review. Breast J 2024; 2024:9083055. [PMID: 39742383 PMCID: PMC11257761 DOI: 10.1155/2024/9083055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 01/03/2025]
Abstract
Introduction Triple-negative breast cancer (TNBC) is a subgroup of breast cancer characterized by the absence of estrogen and the human epidermal 2 receptor and also a lack of targeted therapy options. Chemotherapy has so far been the only approved treatment option, and patients with metastatic cancer have a dismal prognosis with a median overall survival (OS) of approximately 14 months. Identification of druggable targets for metastatic TNBC is therefore of special interest. Methods A systematic search was performed, to review the existing evidence on targeted therapies in metastatic TNBC. Results A total of 37 phase 2/3 studies were identified, evaluating 29 different targeted agents. In this review, results on progression free survival (PFS) and OS are presented. Conclusion In most of the studies included, no improvement was observed for neither PFS nor OS; however, a few studies did show improvement with targeted agents and have led to new treatment options in subgroups of patients. The antibody drug conjugate, sacituzumab govitecan, demonstrated superior PFS and OS in comparison to chemotherapy. Immunotherapy with checkpoint inhibitors such as atezolizumab and pembrolizumab is now recommended as a first-line treatment option for patients with expression a PD-L1 positive tumor. Finally, the poly adenosine diphosphate-ribose polymerase (PARP) inhibitors talazoparib and olaparib are recommended, as first-line treatment options in patients with metastatic breast cancer and a germline BRCA mutation, but an immune checkpoint inhibitor should be considered for the subset of these patients who are PD-L1 positive.
Collapse
Affiliation(s)
- Anna Martha Hammershøi Madsen
- Department of Oncology 54 B1Herlev HospitalUniversity of Copenhagen, Herlev Ringvej 75, DK-2730, Copenhagen, Denmark
| | - Rikke Helene Løvendahl Eefsen
- Department of Oncology 54 B1Herlev HospitalUniversity of Copenhagen, Herlev Ringvej 75, DK-2730, Copenhagen, Denmark
| | - Dorte Nielsen
- Department of Oncology 54 B1Herlev HospitalUniversity of Copenhagen, Herlev Ringvej 75, DK-2730, Copenhagen, Denmark
| | - Iben Kümler
- Department of Oncology 54 B1Herlev HospitalUniversity of Copenhagen, Herlev Ringvej 75, DK-2730, Copenhagen, Denmark
| |
Collapse
|
19
|
Towner RA, Dissanayake R, Ahmed M. Clinical Advances in Triple Negative Breast Cancer Treatment: Focus on Poly (L-lactide-coglycolide) Nanoparticles. J Pharmacol Exp Ther 2024; 390:53-64. [PMID: 38580448 DOI: 10.1124/jpet.123.002016] [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: 11/10/2023] [Revised: 03/16/2024] [Accepted: 03/27/2024] [Indexed: 04/07/2024] Open
Abstract
Triple negative breast cancer (TNBC) is the most aggressive type of breast cancer and is associated with high probability of metastasis and poor prognosis. Chemotherapeutics and surgery remain the most common options for TNBC patients; however, chemotherapeutic resistance and relapse of tumors limit the progression free survival and patient life span. This review provides an overview of recent chemotherapeutics that are in clinical trial, and the combination of drugs that are being investigated to overcome the drug resistance and to improve patient survival in different molecular subtypes of TNBCs. Nanotherapeutics have emerged as a promising platform for TNBC treatment and aim to improve the selectivity and solubility of drugs, reduce systemic side effects, and overcome multi-drug resistance. The study explores the role of nanoparticles for TNBC treatment and summarizes the types of nanoparticles that are in clinical trials. Poly(L-lactide-co-glycolide) (PLGA) is the most studied polymeric carrier for drug delivery and for TNBC treatment in research and in clinics. This review is about providing recent advancements in PLGA nanotherapeutic formulations and their application to help treat TNBC. Some background on current chemotherapies and pathway inhibitors is provided so that the readers are aware of what is currently considered for TNBC. Some of the pathway inhibitors may also be of importance for nanotherapeutics development. SIGNIFICANCE STATEMENT: This minireview summarizes the progress on chemotherapeutics and nanoparticle delivery for treatment of TNBC and specifically highlights the lead compounds that are in clinical trials.
Collapse
Affiliation(s)
- Rheal A Towner
- Department of Chemistry (R.A.T., R.D., M.A.) and Faculty of Sustainable Design Engineering (M.A.), University of Prince Edward Island, Charlottetown, Canada
| | - Ranga Dissanayake
- Department of Chemistry (R.A.T., R.D., M.A.) and Faculty of Sustainable Design Engineering (M.A.), University of Prince Edward Island, Charlottetown, Canada
| | - Marya Ahmed
- Department of Chemistry (R.A.T., R.D., M.A.) and Faculty of Sustainable Design Engineering (M.A.), University of Prince Edward Island, Charlottetown, Canada
| |
Collapse
|
20
|
Long L, Fei X, Chen L, Yao L, Lei X. Potential therapeutic targets of the JAK2/STAT3 signaling pathway in triple-negative breast cancer. Front Oncol 2024; 14:1381251. [PMID: 38699644 PMCID: PMC11063389 DOI: 10.3389/fonc.2024.1381251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Triple-negative breast cancer (TNBC) poses a significant clinical challenge due to its propensity for metastasis and poor prognosis. TNBC evades the body's immune system recognition and attack through various mechanisms, including the Janus Kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. This pathway, characterized by heightened activity in numerous solid tumors, exhibits pronounced activation in specific TNBC subtypes. Consequently, targeting the JAK2/STAT3 signaling pathway emerges as a promising and precise therapeutic strategy for TNBC. The signal transduction cascade of the JAK2/STAT3 pathway predominantly involves receptor tyrosine kinases, the tyrosine kinase JAK2, and the transcription factor STAT3. Ongoing preclinical studies and clinical research are actively investigating this pathway as a potential therapeutic target for TNBC treatment. This article comprehensively reviews preclinical and clinical investigations into TNBC treatment by targeting the JAK2/STAT3 signaling pathway using small molecule compounds. The review explores the role of the JAK2/STAT3 pathway in TNBC therapeutics, evaluating the benefits and limitations of active inhibitors and proteolysis-targeting chimeras in TNBC treatment. The aim is to facilitate the development of novel small-molecule compounds that target TNBC effectively. Ultimately, this work seeks to contribute to enhancing therapeutic efficacy for patients with TNBC.
Collapse
Affiliation(s)
- Lin Long
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiangyu Fei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Liucui Chen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Liang Yao
- Department of Pharmacy, Central Hospital of Hengyang, Hengyang, China
| | - Xiaoyong Lei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
21
|
Ma Y, Fang Z, Zhang H, Qi Y, Mao Y, Zheng J. PDZK1 suppresses TNBC development and sensitizes TNBC cells to erlotinib via the EGFR pathway. Cell Death Dis 2024; 15:199. [PMID: 38604999 PMCID: PMC11009252 DOI: 10.1038/s41419-024-06502-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 04/13/2024]
Abstract
Epidermal growth factor receptor (EGFR)-targeted drugs (erlotinib, etc.) are used to treat multiple types of tumours. EGFR is highly expressed in most triple-negative breast cancer (TNBC) patients. However, only a small proportion of TNBC patients benefit from EGFR-targeted drugs in clinical trials, and the resistance mechanism is unclear. Here, we found that PDZ domain containing 1 (PDZK1) is downregulated in erlotinib-resistant TNBC cells, suggesting that PDZK1 downregulation is related to erlotinib resistance in TNBC. PDZK1 binds to EGFR. Through this interaction, PDZK1 promotes EGFR degradation by enhancing the binding of EGFR to c-Cbl and inhibits EGFR phosphorylation by hindering EGFR dimerisation. We also found that PDZK1 is specifically downregulated in TNBC tissues and correlated with a poor prognosis in TNBC patients. In vitro and in vivo functional assays showed that PDZK1 suppressed TNBC development. Restoration of EGFR expression or kinase inhibitor treatment reversed the degree of cell malignancy induced by PDZK1 overexpression or knockdown, respectively. PDZK1 overexpression sensitised TNBC cells to erlotinib both in vitro and in vivo. In conclusion, PDZK1 is a significant prognostic factor for TNBC and a potential molecular therapeutic target for reversing erlotinib resistance in TNBC cells.
Collapse
Affiliation(s)
- Yuanzhen Ma
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Zhiyu Fang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Hongning Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Yijun Qi
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Yuke Mao
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Junfang Zheng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China.
| |
Collapse
|
22
|
Pellecchia S, Franchini M, Viscido G, Arnese R, Gambardella G. Single cell lineage tracing reveals clonal dynamics of anti-EGFR therapy resistance in triple negative breast cancer. Genome Med 2024; 16:55. [PMID: 38605363 PMCID: PMC11008053 DOI: 10.1186/s13073-024-01327-2] [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: 05/02/2023] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Most primary Triple Negative Breast Cancers (TNBCs) show amplification of the Epidermal Growth Factor Receptor (EGFR) gene, leading to increased protein expression. However, unlike other EGFR-driven cancers, targeting this receptor in TNBC yields inconsistent therapeutic responses. METHODS To elucidate the underlying mechanisms of this variability, we employ cellular barcoding and single-cell transcriptomics to reconstruct the subclonal dynamics of EGFR-amplified TNBC cells in response to afatinib, a tyrosine kinase inhibitor (TKI) that irreversibly inhibits EGFR. RESULTS Integrated lineage tracing analysis revealed a rare pre-existing subpopulation of cells with distinct biological signature, including elevated expression levels of Insulin-Like Growth Factor Binding Protein 2 (IGFBP2). We show that IGFBP2 overexpression is sufficient to render TNBC cells tolerant to afatinib treatment by activating the compensatory insulin-like growth factor I receptor (IGF1-R) signalling pathway. Finally, based on reconstructed mechanisms of resistance, we employ deep learning techniques to predict the afatinib sensitivity of TNBC cells. CONCLUSIONS Our strategy proved effective in reconstructing the complex signalling network driving EGFR-targeted therapy resistance, offering new insights for the development of individualized treatment strategies in TNBC.
Collapse
Affiliation(s)
- Simona Pellecchia
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Scuola Superiore Meridionale, Genomics and Experimental Medicine Program, Naples, Italy
| | - Melania Franchini
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | - Gaetano Viscido
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Chemical, Materials and Industrial Engineering , University of Naples Federico II, Naples, Italy
| | - Riccardo Arnese
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
23
|
Wang H, Wang Z, Wang Z, Li X, Li Y, Yan N, Wu L, Liang Y, Wu J, Song H, Qu Q, Huang J, Chang C, Shen K, Chen X, Lu M. Decitabine induces IRF7-mediated immune responses in p53-mutated triple-negative breast cancer: a clinical and translational study. Front Med 2024; 18:357-374. [PMID: 38157193 DOI: 10.1007/s11684-023-1016-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/24/2023] [Indexed: 01/03/2024]
Abstract
p53 is mutated in half of cancer cases. However, no p53-targeting drugs have been approved. Here, we reposition decitabine for triple-negative breast cancer (TNBC), a subtype with frequent p53 mutations and extremely poor prognosis. In a retrospective study on tissue microarrays with 132 TNBC cases, DNMT1 overexpression was associated with p53 mutations (P = 0.037) and poor overall survival (OS) (P = 0.010). In a prospective DEciTabinE and Carboplatin in TNBC (DETECT) trial (NCT03295552), decitabine with carboplatin produced an objective response rate (ORR) of 42% in 12 patients with stage IV TNBC. Among the 9 trialed patients with available TP53 sequencing results, the 6 patients with p53 mutations had higher ORR (3/6 vs. 0/3) and better OS (16.0 vs. 4.0 months) than the patients with wild-type p53. In a mechanistic study, isogenic TNBC cell lines harboring DETECT-derived p53 mutations exhibited higher DNMT1 expression and decitabine sensitivity than the cell line with wild-type p53. In the DETECT trial, decitabine induced strong immune responses featuring the striking upregulation of the innate immune player IRF7 in the p53-mutated TNBC cell line (upregulation by 16-fold) and the most responsive patient with TNBC. Our integrative studies reveal the potential of repurposing decitabine for the treatment of p53-mutated TNBC and suggest IRF7 as a potential biomarker for decitabine-based treatments.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhengyuan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyang Li
- Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuntong Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ni Yan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lili Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Liang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiale Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huaxin Song
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qing Qu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiahui Huang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chunkang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200025, China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Min Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
24
|
Wang P, Zhou R, Zhou R, Feng S, Zhao L, Li W, Lin J, Rajapakse A, Lee CH, Furnari FB, Burgess AW, Gunter JH, Liu G, Ostrikov KK, Richard DJ, Simpson F, Dai X, Thompson EW. Epidermal growth factor potentiates EGFR(Y992/1173)-mediated therapeutic response of triple negative breast cancer cells to cold atmospheric plasma-activated medium. Redox Biol 2024; 69:102976. [PMID: 38052106 PMCID: PMC10746566 DOI: 10.1016/j.redox.2023.102976] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/24/2023] [Indexed: 12/07/2023] Open
Abstract
Cold atmospheric plasma (CAP) holds promise as a cancer-specific treatment that selectively kills various types of malignant cells. We used CAP-activated media (PAM) to utilize a range of the generated short- and long-lived reactive species. Specific antibodies, small molecule inhibitors and CRISPR/Cas9 gene-editing approaches showed an essential role for receptor tyrosine kinases, especially epidermal growth factor (EGF) receptor, in mediating triple negative breast cancer (TNBC) cell responses to PAM. EGF also dramatically enhanced the sensitivity and specificity of PAM against TNBC cells. Site-specific phospho-EGFR analysis, signal transduction inhibitors and reconstitution of EGFR-depleted cells with EGFR-mutants confirmed the role of phospho-tyrosines 992/1173 and phospholipase C gamma signaling in up-regulating levels of reactive oxygen species above the apoptotic threshold. EGF-triggered EGFR activation enhanced the sensitivity and selectivity of PAM effects on TNBC cells. The proposed approach based on the synergy of CAP and EGFR-targeted therapy may provide new opportunities to improve the clinical management of TNBC.
Collapse
Affiliation(s)
- Peiyu Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia; State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Renwu Zhou
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, School of Electrical Engineering, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Rusen Zhou
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Shuo Feng
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Liqian Zhao
- Department of Neurosurgery, Institute of Brain Disease, Nanfang Hospital of Southern Medical University, Guangzhou 510515, PR China
| | - Wenshao Li
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Jinyong Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Aleksandra Rajapakse
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Chia-Hwa Lee
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Frank B Furnari
- Department of Medicine, University of California San Diego, California 92093, USA
| | - Antony W Burgess
- Walter and Elisa Hall Institute, Melbourne, Victoria 3052, Australia
| | - Jennifer H Gunter
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Kostya Ken Ostrikov
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Derek J Richard
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia; Cancer and Ageing Research Program, Woolloongabba, Queensland 4102, Australia
| | - Fiona Simpson
- Frazer Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Erik W Thompson
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| |
Collapse
|
25
|
Alkhatib H, Conage-Pough J, Roy Chowdhury S, Shian D, Zaid D, Rubinstein AM, Sonnenblick A, Peretz-Yablonsky T, Granit A, Carmon E, Kohale IN, Boughey JC, Goetz MP, Wang L, White FM, Kravchenko-Balasha N. Patient-specific signaling signatures predict optimal therapeutic combinations for triple negative breast cancer. Mol Cancer 2024; 23:17. [PMID: 38229082 PMCID: PMC10790458 DOI: 10.1186/s12943-023-01921-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/20/2023] [Indexed: 01/18/2024] Open
Abstract
Triple negative breast cancer (TNBC) is a heterogeneous group of tumors which lack estrogen receptor, progesterone receptor, and HER2 expression. Targeted therapies have limited success in treating TNBC, thus a strategy enabling effective targeted combinations is an unmet need. To tackle these challenges and discover individualized targeted combination therapies for TNBC, we integrated phosphoproteomic analysis of altered signaling networks with patient-specific signaling signature (PaSSS) analysis using an information-theoretic, thermodynamic-based approach. Using this method on a large number of TNBC patient-derived tumors (PDX), we were able to thoroughly characterize each PDX by computing a patient-specific set of unbalanced signaling processes and assigning a personalized therapy based on them. We discovered that each tumor has an average of two separate processes, and that, consistent with prior research, EGFR is a major core target in at least one of them in half of the tumors analyzed. However, anti-EGFR monotherapies were predicted to be ineffective, thus we developed personalized combination treatments based on PaSSS. These were predicted to induce anti-EGFR responses or to be used to develop an alternative therapy if EGFR was not present.In-vivo experimental validation of the predicted therapy showed that PaSSS predictions were more accurate than other therapies. Thus, we suggest that a detailed identification of molecular imbalances is necessary to tailor therapy for each TNBC. In summary, we propose a new strategy to design personalized therapy for TNBC using pY proteomics and PaSSS analysis. This method can be applied to different cancer types to improve response to the biomarker-based treatment.
Collapse
Affiliation(s)
- Heba Alkhatib
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, 9103401, Jerusalem, Israel
| | - Jason Conage-Pough
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sangita Roy Chowdhury
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, 9103401, Jerusalem, Israel
| | - Denen Shian
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, 9103401, Jerusalem, Israel
| | - Deema Zaid
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, 9103401, Jerusalem, Israel
| | - Ariel M Rubinstein
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, 9103401, Jerusalem, Israel
| | - Amir Sonnenblick
- Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Peretz-Yablonsky
- Sharett Institute of Oncology, Hebrew University-Hadassah Medical Center, 9103401, Jerusalem, Israel
| | - Avital Granit
- Sharett Institute of Oncology, Hebrew University-Hadassah Medical Center, 9103401, Jerusalem, Israel
| | - Einat Carmon
- Department of Surgery, Samson Assuta Ashdod University Hospital, Ashdod, Israel
| | - Ishwar N Kohale
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Matthew P Goetz
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Forest M White
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, 9103401, Jerusalem, Israel.
| |
Collapse
|
26
|
Li Y, Liang X, Li H, Chen X. Reconstruction of unreported subgroup survival data with PD-L1-low expression in advanced/metastatic triple-negative breast cancer using innovative KMSubtraction workflow. J Immunother Cancer 2024; 12:e007931. [PMID: 38212119 PMCID: PMC10806559 DOI: 10.1136/jitc-2023-007931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Among patients with advanced/metastatic triple-negative breast cancer (TNBC) with high/positive programmed death-ligand 1 (PD-L1) expression, a superior survival outcome has been demonstrated with immune checkpoint inhibitors (ICIs). However, it remains unclear whether ICIs are beneficial for patients with low PD-L1 levels. Here, we derived survival data for subgroups with low PD-L1-expressing and conducted a pooled analysis. METHODS After a systematic search of Embase, PubMed, MEDLINE, and CENTRAL from inception until May 18, 2023, randomized controlled trials (RCTs) reporting progression-free survival (PFS), overall survival (OS), or duration of response (DOR) for metastatic TNBC treated with ICI-based regimens were included. Kaplan-Meier curves were extracted for the intention-to-treat population and high PD-L1 subgroups. KMSubtraction was used when survival curves were not provided for subgroups with low PD-L1 expression. A pooled analysis of survival data was then conducted. RESULTS A total of 3022 patients were included in four RCTs: Impassion130, Impassion131, KEYNOTE-119, and KEYNOTE-355. Unreported low PD-L1-expressing subgroups were identified, including PD-L1 immune cell (IC)<1%, combined positive score (CPS)<1, and 1≤CPS<10. Compared with chemotherapy, ICI-chemotherapy combinations did not significantly differ in OS, PFS, or DOR in the Impassion PD-L1<1%, KEYNOTE-355 PD-L1 CPS<1, and KEYNOTE-355 1≤CPS<10 subgroups. In the KEYNOTE-119 CPS<1 subgroup, the risk of tumor progression was increased with pembrolizumab (HR, 2.23; 95% CI, 1.62 to 3.08; p<0.001), as well as in the 1≤CPS<10 subgroup (HR, 1.64; 95% CI, 1.22 to 2.20; p<0.001). A pooled analysis using a scoring system found no significant difference in OS and PFS among the subgroups with an IC of <1% between immunochemotherapy and chemotherapy. OS (HR, 1.07; 95% CI, 0.91 to 1.26), PFS (HR, 0.96; 95% CI, 0.84 to 1.10), and DOR were also not significantly different in pooled analysis of first-line trials for those with low PD-L1 expression. CONCLUSION ICI-based regimens are not associated with a survival benefit versus chemotherapy in subgroups of advanced/metastatic TNBC that express low PD-L1 levels.
Collapse
Affiliation(s)
- Yan Li
- Department of Clinical Pharmacy, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xueyan Liang
- Phase 1 Clinical Trial Laboratory, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Huijuan Li
- Phase 1 Clinical Trial Laboratory, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiaoyu Chen
- Department of Clinical Pharmacy, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Phase 1 Clinical Trial Laboratory, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
27
|
Tan YQ, Chiou YS, Guo H, Zhang S, Huang X, Dukanya D, Kumar AM, Basappa S, Liu S, Zhu T, Basappa B, Pandey V, Lobie PE. Vertical pathway inhibition of receptor tyrosine kinases and BAD with synergistic efficacy in triple negative breast cancer. NPJ Precis Oncol 2024; 8:8. [PMID: 38200104 PMCID: PMC10781691 DOI: 10.1038/s41698-023-00489-3] [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: 04/23/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Aberrant activation of the PI3K/AKT signaling axis along with the sustained phosphorylation of downstream BAD is associated with a poor outcome of TNBC. Herein, the phosphorylated to non-phosphorylated ratio of BAD, an effector of PI3K/AKT promoting cell survival, was observed to be correlated with worse clinicopathologic indicators of outcome, including higher grade, higher proliferative index and lymph node metastasis. The structural optimization of a previously reported inhibitor of BAD-Ser99 phosphorylation was therefore achieved to generate a small molecule inhibiting the phosphorylation of BAD at Ser99 with enhanced potency and improved oral bioavailability. The molecule 2-((4-(2,3-dichlorophenyl)piperazin-1-yl)(pyridin-3-yl)methyl) phenol (NCK) displayed no toxicity at supra-therapeutic doses and was therefore assessed for utility in TNBC. NCK promoted apoptosis and G0/G1 cell cycle arrest of TNBC cell lines in vitro, concordant with gene expression analyses, and reduced in vivo xenograft growth and metastatic burden, demonstrating efficacy as a single agent. Additionally, combinatorial oncology compound library screening demonstrated that NCK synergized with tyrosine kinase inhibitors (TKIs), specifically OSI-930 or Crizotinib in reducing cell viability and promoting apoptosis of TNBC cells. The synergistic effects of NCK and TKIs were also observed in vivo with complete regression of a percentage of TNBC cell line derived xenografts and prevention of metastatic spread. In patient-derived TNBC xenograft models, NCK prolonged survival times of host animals, and in combination with TKIs generated superior survival outcomes to single agent treatment. Hence, this study provides proof of concept to further develop rational and mechanistic based therapeutic strategies to ameliorate the outcome of TNBC.
Collapse
Grants
- This research was supported by the National Natural Science Foundation of China (82172618 to P.E.L. and 82102768 to Y.Q.T.), China; the Shenzhen Key Laboratory of Innovative Oncotherapeutics (ZDSYS20200820165400003 to P.E.L.) (Shenzhen Science and Technology Innovation Commission), China; Shenzhen Development and Reform Commission Subject Construction Project ([2017]1434 to P.E.L.), China; Universities Stable Funding Key Projects (WDZC20200821150704001 to P.E.L.), China; Guangdong Basic and Applied Basic Research Foundation (2020A1515111064 to Y.Q.T.), China; The Shenzhen Bay Laboratory, Oncotherapeutics (21310031 to P.E.L.), China; Overseas Research Cooperation Project (HW2020008 to V.P.) (Tsinghua Shenzhen International Graduate School), China; Research Fund, Kaohsiung Medical University (KMU-Q112002 to Y.C.), Taiwan and China Postdoctoral Science Foundation (2022M721894 to X.H.), China.
Collapse
Affiliation(s)
- Yan Qin Tan
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, People's Republic of China
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Yi-Shiou Chiou
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China
- Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Shenzhen Bay Laboratory, Shenzhen, 518055, Guangdong, People's Republic of China
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Hui Guo
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Shuwei Zhang
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Xiaoming Huang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, People's Republic of China
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China
- Shenzhen Bay Laboratory, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Dukanya Dukanya
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, 570006, Mysore, India
| | - Arun M Kumar
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, 570006, Mysore, India
| | - Shreeja Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, 570006, Mysore, India
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Shanghai Medical College, Key Laboratory of Breast Cancer in Shanghai, Innovation Center for Cell Signaling Network, Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Tao Zhu
- Shenzhen Bay Laboratory, Shenzhen, 518055, Guangdong, People's Republic of China
- Department of Oncology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
- Hefei National Laboratory for Physical Sciences, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, 570006, Mysore, India.
| | - Vijay Pandey
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, People's Republic of China.
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China.
| | - Peter E Lobie
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, People's Republic of China.
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, Guangdong, People's Republic of China.
| |
Collapse
|
28
|
Kumar H, Gupta NV, Jain R, Madhunapantula SV, Babu CS, Kesharwani SS, Dey S, Jain V. A review of biological targets and therapeutic approaches in the management of triple-negative breast cancer. J Adv Res 2023; 54:271-292. [PMID: 36791960 DOI: 10.1016/j.jare.2023.02.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a heterogeneous, aggressive phenotype of breast cancer with associated chemoresistance. The development of chemo- or radioresistance could be attributed to diverse tumor microenvironments, overexpression of membrane proteins (transporters), epigenetic changes, and alteration of the cell signaling pathways/genes associated with the development of cancer stem cells (CSCs). AIM OF REVIEW Due to the diverse and heterogeneous nature of TNBC, therapeutic response to the existing modalities offers limited scope and thus results in reccurance after therapy. To establish landmark therapeutic efficacy, a number of novel therapeutic modalities have been proposed. In addition, reversal of the resistance that developed during treatment may be altered by employing appropriate therapeutic modalities. This review aims to discuss the plethora of investigations carried out, which will help readers understand and make an appropriate choice of therapy directed toward complete elimination of TNBC. KEY SCIENTIFIC CONCEPTS OF REVIEW This manuscript addresses the major contributory factors from the tumor microenvironment that are responsible for the development of chemoresistance and poor prognosis. The associated cellular events and molecular mechanism-based therapeutic interventions have been explained in detail. Inhibition of ABC transporters, cell signaling pathways associated with CSCs, and epigenetic modification offers promising results in this regard. TNBC progression, invasion, metastasis and recurrence can also be inhibited by blocking multiple cell signaling pathways, targeting specific receptors/epigenetic targets, disrupting bioenergetics and generating reactive oxygen species (ROS).
Collapse
Affiliation(s)
- Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - N Vishal Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - SubbaRao V Madhunapantula
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - C Saravana Babu
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | | | - Surajit Dey
- Roseman University of Health Sciences, College of Pharmacy, Henderson, NV, USA
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India.
| |
Collapse
|
29
|
Zia T, Bangfan L, Nadeem A, Hussain A, Abdel-Maksoud MA, Zakri AM, Bashir MK, Ali M, Jabeen N, Jamil M, Al-Qahtani WH, Almanaa TN. Comprehensive multi-level expression profiling of key biomarkers in breast cancer patients. Am J Transl Res 2023; 15:6058-6070. [PMID: 37969199 PMCID: PMC10641354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 10/11/2023] [Indexed: 11/17/2023]
Abstract
OBJECTIVES In this comprehensive breast cancer (BC) study, we aimed to identify, validate, and characterize key biomarkers with significant implications in BC diagnosis, prognosis, and as therapeutic targets. METHODS Our research strategy involved a multi-level methodology, combining bioinformatic analysis with experimental validation. RESULTS Initially, we conducted an extensive literature search to identify BC biomarkers, selecting those with reported accuracies exceeding 20% in specificity and sensitivity. This yielded nine candidate biomarkers, which we subsequently analyzed using Cytoscape to identify a few key biomarkers. Based on the degree method, we denoted four key biomarkers, including progesterone receptor (PGR), epidermal growth factor receptor (EGFR), estrogen receptor 1 (ESR1), and Erb-B2 Receptor Tyrosine Kinase 2 (ERBB2). Expression analysis using The Cancer Genome Atlas (TCGA) dataset revealed that PGR and EGFR exhibited significant (p-value < 0.05) down-regulation in BC samples when compared to controls, while ESR1 and ERBB2 showed up-regulation. To strengthen our findings, we collected clinical BC tissue samples from Pakistani patients and performed expression verification using real-time quantitative polymerase chain reaction (RT-qPCR). The results aligned with our initial TCGA dataset analysis, further validating the differential expression of these key biomarkers in BC. Furthermore, we utilized receiver operating characteristic (ROC) curves to demonstrate the diagnostic use of these biomarkers. Our analysis underscored their accuracy and sensitivity as diagnostic markers for BC. Survival analysis using the Kaplan-Meier Plotter tool revealed a prognostic significance of PGR, ESR1, EGFR, and ERBB2. Their expression levels were associated with poor overall survival (OS) of BC patients, shedding light on their roles as prognostic indicators in BC. Lastly, we explored DrugBank to identify drugs that may reverse the expression patterns , and estradiol, decitabine, and carbamazepine were singled out. CONCLUSION Our study gives valuable insight into BC biomarkers, for diagnosis and prognosis. These findings have implications for BC management using personalized and targeted therapeutic approaches for BC patients.
Collapse
Affiliation(s)
- Tayyaba Zia
- School of Public Management, Yanshan UniversityQinhuangdao 066000, Hebei, China
| | - Liu Bangfan
- School of Public Administration, Yanshan UniversityQinhuangdao 066000, Hebei, China
| | - Amun Nadeem
- Department of Pathology, Gujranwala Medical College Teaching Hospital and DHQ Teaching HospitalGujranwala 52250, Punjab, Pakistan
| | - Abid Hussain
- APMO Nishter HospitalMultan 60000, Punjab, Pakistan
| | - Mostafa A Abdel-Maksoud
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Adel M Zakri
- Department of Plant Production, College of Food and Agricultural Sciences, King Saud UniversitySaudi Arabia
| | - Muhammad Kazim Bashir
- Shaukat Khanum Memorial Cancer Hospital and Research CentreLahore 13014, Punjab, Pakistan
| | - Mubarak Ali
- Animal Science Institute, National Agricultural Research CenterIslamabad 54000, Pakistan
| | - Norina Jabeen
- Department of Rural Sociology, University of Agriculture FaisalabadFaisalabad 38000, Punjab, Pakistan
| | - Muhammad Jamil
- PARC Arid Zone Research CentreDera Ismail Khan 29050, Pakistan
| | - Wahidah H Al-Qahtani
- Department of Food Sciences & Nutrition, College of Food and Agricultural Sciences, King Saud UniversitySaudi Arabia
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
30
|
Das K, Paul S, Ghosh A, Gupta S, Mukherjee T, Shankar P, Sharma A, Keshava S, Chauhan SC, Kashyap VK, Parashar D. Extracellular Vesicles in Triple-Negative Breast Cancer: Immune Regulation, Biomarkers, and Immunotherapeutic Potential. Cancers (Basel) 2023; 15:4879. [PMID: 37835573 PMCID: PMC10571545 DOI: 10.3390/cancers15194879] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype accounting for ~10-20% of all human BC and is characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) amplification. Owing to its unique molecular profile and limited targeted therapies, TNBC treatment poses significant challenges. Unlike other BC subtypes, TNBC lacks specific molecular targets, rendering endocrine therapies and HER2-targeted treatments ineffective. The chemotherapeutic regimen is the predominant systemic treatment modality for TNBC in current clinical practice. However, the efficacy of chemotherapy in TNBC is variable, with response rates varying between a wide range of patients, and the emerging resistance further adds to the difficulties. Furthermore, TNBC exhibits a higher mutational burden and is acknowledged as the most immunogenic of all BC subtypes. Consequently, the application of immune checkpoint inhibition has been investigated in TNBC, yielding promising outcomes. Recent evidence identified extracellular vesicles (EVs) as an important contributor in the context of TNBC immunotherapy. In view of the extraordinary ability of EVs to transfer bioactive molecules, such as proteins, lipids, DNA, mRNAs, and small miRNAs, between the cells, EVs are considered a promising diagnostic biomarker and novel drug delivery system among the prospects for immunotherapy. The present review provides an in-depth understanding of how EVs influence TNBC progression, its immune regulation, and their contribution as a predictive biomarker for TNBC. The final part of the review focuses on the recent key advances in immunotherapeutic strategies for better understanding the complex interplay between EVs and the immune system in TNBC and further developing EV-based targeted immunotherapies.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Subhojit Paul
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700012, India; (S.P.); (A.G.)
| | - Arnab Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700012, India; (S.P.); (A.G.)
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura 281406, India;
| | - Tanmoy Mukherjee
- School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Prem Shankar
- Department of Neurobiology, The University of Texas Medical Branch, Galveston, TX 77555, USA or
| | - Anshul Sharma
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (S.C.C.); (V.K.K.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Vivek Kumar Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (S.C.C.); (V.K.K.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Deepak Parashar
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
31
|
Ang HX, Sutiman N, Deng XL, Liu A, Cerda-Smith CG, Hutchinson HM, Kim H, Bartelt LC, Chen Q, Barrera A, Lin J, Sheng Z, McDowell IC, Reddy TE, Nicchitta CV, Wood KC. Cooperative regulation of coupled oncoprotein synthesis and stability in triple-negative breast cancer by EGFR and CDK12/13. Proc Natl Acad Sci U S A 2023; 120:e2221448120. [PMID: 37695916 PMCID: PMC10515179 DOI: 10.1073/pnas.2221448120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/19/2023] [Indexed: 09/13/2023] Open
Abstract
Evidence has long suggested that epidermal growth factor receptor (EGFR) may play a prominent role in triple-negative breast cancer (TNBC) pathogenesis, but clinical trials of EGFR inhibitors have yielded disappointing results. Using a candidate drug screen, we identified that inhibition of cyclin-dependent kinases 12 and 13 (CDK12/13) dramatically sensitizes diverse models of TNBC to EGFR blockade. This combination therapy drives cell death through the 4E-BP1-dependent suppression of the translation and translation-linked turnover of driver oncoproteins, including MYC. A genome-wide CRISPR/Cas9 screen identified the CCR4-NOT complex as a major determinant of sensitivity to the combination therapy whose loss renders 4E-BP1 unresponsive to drug-induced dephosphorylation, thereby rescuing MYC translational suppression and promoting MYC stability. The central roles of CCR4-NOT and 4E-BP1 in response to the combination therapy were further underscored by the observation of CNOT1 loss and rescue of 4E-BP1 phosphorylation in TNBC cells that naturally evolved therapy resistance. Thus, pharmacological inhibition of CDK12/13 reveals a long-proposed EGFR dependence in TNBC that functions through the cooperative regulation of translation-coupled oncoprotein stability.
Collapse
Affiliation(s)
- Hazel X. Ang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC22710
| | - Natalia Sutiman
- Duke-National University of Singapore Medical School,Singapore169857, Singapore
| | - Xinyue L. Deng
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC22710
| | - Annie Liu
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC22710
- Department of Surgery, Duke University School of Medicine, Durham, NC22710
| | - Christian G. Cerda-Smith
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC22710
| | - Haley M. Hutchinson
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC22710
| | - Holly Kim
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC22710
| | - Luke C. Bartelt
- Duke Center for Genomic and Computational Biology, Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC27708
| | - Qiang Chen
- Department of Cell Biology, Duke University School of Medicine, Durham, NC22710
| | - Alejandro Barrera
- Duke Center for Genomic and Computational Biology, Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC27708
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC27708
| | - Jiaxing Lin
- Bioinformatics Shared Resources, Duke Cancer Institute, Duke University Medical Center, Durham, NC27705
| | - Zhecheng Sheng
- Bioinformatics Shared Resources, Duke Cancer Institute, Duke University Medical Center, Durham, NC27705
| | - Ian C. McDowell
- Duke Center for Genomic and Computational Biology, Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC27708
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC27708
| | - Timothy E. Reddy
- Duke Center for Genomic and Computational Biology, Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC27708
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC27708
| | | | - Kris C. Wood
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC22710
| |
Collapse
|
32
|
Atwell B, Chalasani P, Schroeder J. Nuclear epidermal growth factor receptor as a therapeutic target. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:616-629. [PMID: 37720348 PMCID: PMC10501894 DOI: 10.37349/etat.2023.00156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/09/2023] [Indexed: 09/19/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) is one of the most well-studied oncogenes with roles in proliferation, growth, metastasis, and therapeutic resistance. This intense study has led to the development of a range of targeted therapeutics including small-molecule tyrosine kinase inhibitors (TKIs), monoclonal antibodies, and nanobodies. These drugs are excellent at blocking the activation and kinase function of wild-type EGFR (wtEGFR) and several common EGFR mutants. These drugs have significantly improved outcomes for patients with cancers including head and neck, glioblastoma, colorectal, and non-small cell lung cancer (NSCLC). However, therapeutic resistance is often seen, resulting from acquired mutations or activation of compensatory signaling pathways. Additionally, these therapies are ineffective in tumors where EGFR is found predominantly in the nucleus, as can be found in triple negative breast cancer (TNBC). In TNBC, EGFR is subjected to alternative trafficking which drives the nuclear localization of the receptor. In the nucleus, EGFR interacts with several proteins to activate transcription, DNA repair, migration, and chemoresistance. Nuclear EGFR (nEGFR) correlates with metastatic disease and worse patient prognosis yet targeting its nuclear localization has proved difficult. This review provides an overview of current EGFR-targeted therapies and novel peptide-based therapies that block nEGFR, as well as their clinical applications and potential for use in oncology.
Collapse
Affiliation(s)
- Benjamin Atwell
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Pavani Chalasani
- Department of Medicine, University of Arizona, Tucson, AZ 85721, USA
- University of Arizona Cancer Center, Tucson, AZ 85721, USA
| | - Joyce Schroeder
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
- University of Arizona Cancer Center, Tucson, AZ 85721, USA
- Bio5 Institute, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
33
|
Zhu S, Wu Y, Song B, Yi M, Yan Y, Mei Q, Wu K. Recent advances in targeted strategies for triple-negative breast cancer. J Hematol Oncol 2023; 16:100. [PMID: 37641116 PMCID: PMC10464091 DOI: 10.1186/s13045-023-01497-3] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
Triple-negative breast cancer (TNBC), a highly aggressive subtype of breast cancer, negatively expresses estrogen receptor, progesterone receptor, and the human epidermal growth factor receptor 2 (HER2). Although chemotherapy is the main form of treatment for patients with TNBC, the effectiveness of chemotherapy for TNBC is still limited. The search for more effective therapies is urgent. Multiple targeted therapeutic strategies have emerged according to the specific molecules and signaling pathways expressed in TNBC. These include PI3K/AKT/mTOR inhibitors, epidermal growth factor receptor inhibitors, Notch inhibitors, poly ADP-ribose polymerase inhibitors, and antibody-drug conjugates. Moreover, immune checkpoint inhibitors, for example, pembrolizumab, atezolizumab, and durvalumab, are widely explored in the clinic. We summarize recent advances in targeted therapy and immunotherapy in TNBC, with the aim of serving as a reference for the development of individualized treatment of patients with TNBC in the future.
Collapse
Affiliation(s)
- Shuangli Zhu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bin Song
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Yuheng Yan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qi Mei
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
34
|
Chaturvedi S, Biswas M, Sadhukhan S, Sonawane A. Role of EGFR and FASN in breast cancer progression. J Cell Commun Signal 2023:10.1007/s12079-023-00771-w. [PMID: 37490191 DOI: 10.1007/s12079-023-00771-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/22/2023] [Indexed: 07/26/2023] Open
Abstract
Breast cancer (BC) emerged as one of the life-threatening diseases among females. Despite notable improvements made in cancer detection and treatment worldwide, according to GLOBACAN 2020, BC is the fifth leading cancer, with an estimated 1 in 6 cancer deaths, in a majority of countries. However, the exact cause that leads to BC progression still needs to be determined. Here, we reviewed the role of two novel biomarkers responsible for 50-70% of BC progression. The first one is epidermal growth factor receptor (EGFR) which belongs to the ErbB tyrosine kinases family, signalling pathways associated with it play a significant role in regulating cell proliferation and division. Another one is fatty acid synthase (FASN), a key enzyme responsible for the de novo lipid synthesis required for cancer cell development. This review presents a rationale for the EGFR-mediated pathways, their interaction with FASN, communion of these two biomarkers with BC, and improvements to overcome drug resistance caused by them.
Collapse
Affiliation(s)
- Suchi Chaturvedi
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Madhya Pradesh, 453552, India
| | - Mainak Biswas
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, 751024, India
| | - Sushabhan Sadhukhan
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala, 678623, India.
- Physical & Chemical Biology Laboratory and Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Palakkad, Kerala, 678623, India.
| | - Avinash Sonawane
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Madhya Pradesh, 453552, India.
| |
Collapse
|
35
|
Valero-Cantero I, Casals C, Espinar-Toledo M, Barón-López FJ, Martínez-Valero FJ, Vázquez-Sánchez MÁ. Cancer Patients' Satisfaction with In-Home Palliative Care and Its Impact on Disease Symptoms. Healthcare (Basel) 2023; 11:healthcare11091272. [PMID: 37174814 PMCID: PMC10178555 DOI: 10.3390/healthcare11091272] [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: 04/04/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
The aim of the study was to determine whether the satisfaction of cancer patients with in-home palliative care is associated with the impact of disease symptoms and with self-perceived quality of life. This was a cross-sectional descriptive study, conducted in the primary health care sector in six clinical management units, where 72 patients were recruited over a period of six months. The severity of symptoms was determined by the Edmonton Symptom Assessment System (ESAS). Quality of life was evaluated with the EORTC QLQ-C30 (version 3) questionnaire, and patients' satisfaction with the care received was evaluated by the Client Satisfaction Questionnaire (CSQ-8). The patients' satisfaction with the health care received was represented by an average score of 6, on a scale of 1-10; thus, there is room for improvement in patient satisfaction. Moreover, it was found that more intense symptoms and lower quality of life are associated with lower satisfaction with health care received (p = 0.001). Similarly, when symptoms are more severe, the quality of life is lower (p < 0.001). The identification of fatigue, reduced well-being, pain, drowsiness, and depression as the symptoms experienced with the highest intensity by our patients provides valuable information for health care providers in developing individualized symptom management plans for patients with advanced cancer.
Collapse
Affiliation(s)
| | - Cristina Casals
- ExPhy Research Group, Department of Physical Education, Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Universidad de Cádiz, 11519 Puerto Real, Spain
| | - Milagrosa Espinar-Toledo
- Rincón de la Victoria Clinical Management Unit, Malaga-Guadalhorce Health District, 29730 Malaga, Spain
| | - Francisco Javier Barón-López
- Faculty of Health Sciences, Institute of Biomedical Research in Málaga (IBIMA), University of Malaga, 29016 Malaga, Spain
| | | | - María Ángeles Vázquez-Sánchez
- Department of Nursing, Faculty of Health Sciences, PASOS Research Group and UMA REDIAS Network of Law and Artificial Intelligence Applied to Health and Biotechnology, University of Malaga, 29071 Malaga, Spain
| |
Collapse
|
36
|
Bi J, Wu Z, Zhang X, Zeng T, Dai W, Qiu N, Xu M, Qiao Y, Ke L, Zhao J, Cao X, Lin Q, Chen XL, Xie L, Ouyang Z, Guo J, Zheng L, Ma C, Guo S, Chen K, Mo W, Fu G, Zhao TJ, Wang HR. TMEM25 inhibits monomeric EGFR-mediated STAT3 activation in basal state to suppress triple-negative breast cancer progression. Nat Commun 2023; 14:2342. [PMID: 37095176 PMCID: PMC10126118 DOI: 10.1038/s41467-023-38115-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/17/2023] [Indexed: 04/26/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with poor outcome and lacks of approved targeted therapy. Overexpression of epidermal growth factor receptor (EGFR) is found in more than 50% TNBC and is suggested as a driving force in progression of TNBC; however, targeting EGFR using antibodies to prevent its dimerization and activation shows no significant benefits for TNBC patients. Here we report that EGFR monomer may activate signal transducer activator of transcription-3 (STAT3) in the absence of transmembrane protein TMEM25, whose expression is frequently decreased in human TNBC. Deficiency of TMEM25 allows EGFR monomer to phosphorylate STAT3 independent of ligand binding, and thus enhances basal STAT3 activation to promote TNBC progression in female mice. Moreover, supplying TMEM25 by adeno-associated virus strongly suppresses STAT3 activation and TNBC progression. Hence, our study reveals a role of monomeric-EGFR/STAT3 signaling pathway in TNBC progression and points out a potential targeted therapy for TNBC.
Collapse
Affiliation(s)
- Jing Bi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Zhihui Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Xin Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China
| | - Taoling Zeng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Wanjun Dai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Ningyuan Qiu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Mingfeng Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Yikai Qiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Lang Ke
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Jiayi Zhao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Xinyu Cao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Qi Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Xiao Lei Chen
- Cancer Research Center of Xiamen University, 361102, Xiamen, Fujian, China
- School of Medicine, Xiamen University, 361102, Fujian, China
| | - Liping Xie
- School of Medicine, Xiamen University, 361102, Fujian, China
| | - Zhong Ouyang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, 361003, Xiamen, Fujian, China
| | - Jujiang Guo
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, 361003, Xiamen, Fujian, China
| | - Liangkai Zheng
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, 361003, Xiamen, Fujian, China
| | - Chao Ma
- Medical School of Chinese PLA, 100853, Beijing, China
| | - Shiying Guo
- GemPharmatech Co., Ltd., 210000, Nanjing, Jiangsu, China
| | - Kangmei Chen
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China
| | - Wei Mo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Guo Fu
- Cancer Research Center of Xiamen University, 361102, Xiamen, Fujian, China.
- School of Medicine, Xiamen University, 361102, Fujian, China.
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, 361003, Xiamen, Fujian, China.
| | - Tong-Jin Zhao
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, 200438, Shanghai, China.
| | - Hong-Rui Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China.
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, 361003, Xiamen, Fujian, China.
| |
Collapse
|
37
|
Mamot C, Wicki A, Hasler-Strub U, Riniker S, Li Q, Holer L, Bärtschi D, Zaman K, von Moos R, Dedes KJ, Boos LA, Novak U, Bodmer A, Ritschard R, Obermann EC, Tzankov A, Ackermann C, Membrez-Antonioli V, Zürrer-Härdi U, Caspar CB, Deuster S, Senn M, Winterhalder R, Rochlitz C. A multicenter phase II trial of anti-EGFR-immunoliposomes loaded with doxorubicin in patients with advanced triple negative breast cancer. Sci Rep 2023; 13:3705. [PMID: 36879012 PMCID: PMC9988854 DOI: 10.1038/s41598-023-30950-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Advanced triple negative breast cancer (TNBC) is an aggressive, but initially chemo-sensitive disease. The prognosis is poor and more than three quarters of patients experience progression 12 months after the initiation of conventional first-line chemotherapy. Approximately two thirds of TNBC express epidermal growth factor receptor 1 (EGFR). We have developed an anti-EGFR targeted nanocontainer drug by inserting anti-EGFR antibody fragments into the membrane of pegylated liposomes (anti-EGFR-ILs-dox). The payload consists of doxorubicin, a standard drug for TNBC. In a first-in-human phase I trial in 26 patients with various advanced solid malignancies, anti-EGFR-ILs-dox has shown little toxicity and encouraging efficacy. In this single-arm phase II trial, we assessed the efficacy of anti-EGFR-ILs-dox as first-line therapy in patients with advanced, EGFR + TNBC. The primary endpoint was progression-free survival at 12 months (PFS12m). Secondary endpoints included overall response rate (ORR), duration of response (DOR), time to progression (TTP), overall survival (OS) and adverse events (AEs). 48 patients received anti-EGFR-ILs-dox 50 mg/m2 iv, on day one of a 28 days-cycle until progression. The Kaplan-Meier estimate for PFS12m was 13% (one-sided 90% CI 7%, 95% CI [5%, 25%]), median PFS was 3.5 months (95% CI 1.9, 5.4). The trial has not reached its primary endpoint. There were no new toxicity signals. Based on these results, anti-EGFR-ILs-dox should not be further developed for TNBC. It remains an open question whether anti-EGFR-ILs-dox would offer more opportunities in other EGFR-expressing malignancies, where targeting this receptor has already shown anticancer effects.Trial registration: This trial was registered at clinicaltrials.gov: NCT02833766. Registered 14/07/2016.
Collapse
Affiliation(s)
- Christoph Mamot
- Cantonal Hospital Aarau, Tellstrasse 25, 5001, Aarau, Switzerland.
| | - Andreas Wicki
- University and University Hospital Zurich, Rämistrasse 100, 8091, Zürich, Switzerland.
| | | | | | - Qiyu Li
- Competence Center of the Swiss Group for Clinical Cancer Research (SAKK), Bern, Switzerland
| | - Lisa Holer
- Competence Center of the Swiss Group for Clinical Cancer Research (SAKK), Bern, Switzerland
| | - Daniela Bärtschi
- Competence Center of the Swiss Group for Clinical Cancer Research (SAKK), Bern, Switzerland
| | - Khalil Zaman
- University Hospital Lausanne, Lausanne, Switzerland
| | | | | | - Laura A Boos
- University and University Hospital Zurich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Urban Novak
- Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | | | | | | | | - Martin Senn
- University Hospital Basel, Basel, Switzerland
| | | | | |
Collapse
|
38
|
Song WM, Chia PL, Zhou X, Walsh M, Silva J, Zhang B. Pseudo-temporal dynamics of chemoresistant triple negative breast cancer cells reveal EGFR/HER2 inhibition as synthetic lethal during mid-neoadjuvant chemotherapy. iScience 2023; 26:106064. [PMID: 36824282 PMCID: PMC9942122 DOI: 10.1016/j.isci.2023.106064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/17/2022] [Accepted: 01/23/2023] [Indexed: 01/29/2023] Open
Abstract
In the absence of targetable hormonal axes, chemoresistance for triple-negative breast cancer (TNBC) often compromises patient outcomes. To investigate the underlying tumor dynamics, we performed trajectory analysis on the single-nuclei RNA-seq (snRNA-seq) of chemoresistant tumor clones during neoadjuvant chemotherapy (NAC). It revealed a common tumor trajectory across multiple patients with HER2-like expansions during NAC. Genome-wide CRISPR-Cas9 knock-out on mammary epithelial cells revealed chemosensitivity-promoting knock-outs were up-regulated along the tumor trajectory. Furthermore, we derived a consensus gene signature of TNBC chemoresistance by comparing the trajectory transcriptome with chemoresistant transcriptomes from TNBC cell lines and poor prognosis patient samples to predict FDA-approved drugs, including afatinib (pan-HER inhibitor), targeting the consensus signature. We validated the synergistic efficacy of afatinib and paclitaxel in chemoresistant TNBC cells and confirmed pharmacological suppression of the consensus signature. The study provides a dynamic model of chemoresistant tumor transcriptome, and computational framework for pharmacological intervention.
Collapse
Affiliation(s)
- Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Pei-Ling Chia
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Martin Walsh
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jose Silva
- Department of Pathology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
39
|
Subham S, Jeppson JD, Worcester C, Schatmeyer B, Zhao J, Madan R, Lakis NS, Kimler BF, McGuirk JP, Chen RC, Stecklein SR, Akhavan D. EGFR as a potent CAR T target in triple negative breast cancer brain metastases. Breast Cancer Res Treat 2023; 197:57-69. [PMID: 36318382 PMCID: PMC10987173 DOI: 10.1007/s10549-022-06783-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/23/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE There is currently no curative treatment for patients diagnosed with triple-negative breast cancer brain metastases (TNBC-BM). CAR T cells hold potential for curative treatment given they retain the cytolytic activity of a T cell combined with the specificity of an antibody. In this proposal we evaluated the potential of EGFR re-directed CAR T cells as a therapeutic treatment against TNBC cells in vitro and in vivo. METHODS We leveraged a TNBC-BM tissue microarray and a large panel of TNBC cell lines and identified elevated epidermal growth factor receptor (EGFR) expression. Next, we designed a second-generation anti-EGFR CAR T construct incorporating a clinically relevant mAb806 tumor specific single-chain variable fragment (scFv) and intracellular 4-1BB costimulatory domain and CD3ζ using a lentivirus system and evaluated in vitro and in vivo anti-tumor activity. RESULTS We demonstrate EGFR is enriched in TNBC-BM patient tissue after neurosurgical resection, with six of 13 brain metastases demonstrating both membranous and cytoplasmic EGFR. Eleven of 13 TNBC cell lines have EGFR surface expression ≥ 85% by flow cytometry. EGFR806 CAR T treated mice effectively eradicated TNBC-BM and enhanced mouse survival (log rank p < 0.004). CONCLUSION Our results demonstrates anti-tumor activity of EGFR806 CAR T cells against TNBC cells in vitro and in vivo. Given EGFR806 CAR T cells are currently undergoing clinical trials in primary brain tumor patients without obvious toxicity, our results are immediately actionable against the TNBC-BM patient population.
Collapse
Affiliation(s)
- Siddharth Subham
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS, USA
- Department of Cancer Biology, University of Kansas Cancer Center, Kansas City, KS, USA
- BioEngineering Program, University of Kansas, Lawrence, KS, USA
| | - John D Jeppson
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS, USA
| | - Colette Worcester
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Bryan Schatmeyer
- Department of Neurosurgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jie Zhao
- Department of Cancer Biology, University of Kansas Cancer Center, Kansas City, KS, USA
| | - Rashna Madan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nelli S Lakis
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Bruce F Kimler
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS, USA
| | - Joseph P McGuirk
- Department of Hematology and Stem Cell Transplantation, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ronald C Chen
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS, USA
| | - Shane R Stecklein
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS, USA
- Department of Cancer Biology, University of Kansas Cancer Center, Kansas City, KS, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - David Akhavan
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS, USA.
- Department of Cancer Biology, University of Kansas Cancer Center, Kansas City, KS, USA.
- BioEngineering Program, University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
40
|
Rashid NS, Boyd DC, Olex AL, Grible JM, Duong AK, Alzubi MA, Altman JE, Leftwich TJ, Valentine AD, Hairr NS, Zboril EK, Smith TM, Pfefferle AD, Dozmorov MG, Harrell JC. Transcriptomic changes underlying EGFR inhibitor resistance in human and mouse models of basal-like breast cancer. Sci Rep 2022; 12:21248. [PMID: 36482068 PMCID: PMC9731984 DOI: 10.1038/s41598-022-25541-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
The goals of this study were to identify transcriptomic changes that arise in basal-like breast cancer cells during the development of resistance to epidermal growth factor receptor inhibitors (EGFRi) and to identify drugs that are cytotoxic once EGFRi resistance occurs. Human patient-derived xenografts (PDXs) were grown in immunodeficient mice and treated with a set of EGFRi; the EGFRi erlotinib was selected for more expansive in vivo studies. Single-cell RNA sequencing was performed on mammary tumors from the basal-like PDX WHIM2 that was treated with vehicle or erlotinib for 9 weeks. The PDX was then subjected to long-term erlotinib treatment in vivo. Through serial passaging, an erlotinib-resistant subline of WHIM2 was generated. Bulk RNA-sequencing was performed on parental and erlotinib-resistant tumors. In vitro high-throughput drug screening with > 500 clinically used compounds was performed on parental and erlotinib-resistant cells. Previously published bulk gene expression microarray data from MMTV-Wnt1 tumors were contrasted with the WHIM2 PDX data. Erlotinib effectively inhibited WHIM2 tumor growth for approximately 4 weeks. Compared to untreated cells, single-cell RNA sequencing revealed that a greater proportion of erlotinib-treated cells were in the G1 phase of the cell cycle. Comparison of WHIM2 and MMTV-Wnt1 gene expression data revealed a set of 38 overlapping genes that were differentially expressed in the erlotinib-resistant WHIM2 and MMTV-Wnt1 tumors. Comparison of all three data types revealed five genes that were upregulated across all erlotinib-resistant samples: IL19, KLK7, LCN2, SAA1, and SAA2. Of these five genes, LCN2 was most abundantly expressed in triple-negative breast cancers, and its knockdown restored erlotinib sensitivity in vitro. Despite transcriptomic differences, parental and erlotinib-resistant WHIM2 displayed similar responses to the majority of drugs assessed for cytotoxicity in vitro. This study identified transcriptomic changes arising in erlotinib-resistant basal-like breast cancer. These data could be used to identify a biomarker or develop a gene signature predictive of patient response to EGFRi. Future studies should explore the predictive capacity of these gene signatures as well as how LCN2 contributes to the development of EGFRi resistance.
Collapse
Affiliation(s)
- Narmeen S Rashid
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
- Department of Biology, University of Richmond, Richmond, VA, 23173, USA
| | - David C Boyd
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
- Program in Integrative Life Sciences, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Amy L Olex
- C. Kenneth and Diane Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Jacqueline M Grible
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Alex K Duong
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Mohammad A Alzubi
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
- Oncology Center-Division of Pediatric Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Julia E Altman
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Tess J Leftwich
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Aaron D Valentine
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Nicole S Hairr
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Emily K Zboril
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Timothy M Smith
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Adam D Pfefferle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Mikhail G Dozmorov
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - J Chuck Harrell
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA.
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23220, USA.
| |
Collapse
|
41
|
Passalacqua MI, Rizzo G, Santarpia M, Curigliano G. 'Why is survival with triple negative breast cancer so low? insights and talking points from preclinical and clinical research'. Expert Opin Investig Drugs 2022; 31:1291-1310. [PMID: 36522800 DOI: 10.1080/13543784.2022.2159805] [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] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Triple negative breast cancer is typically related to poor prognosis, early metastasis, and high recurrence rate. Intrinsic and extrinsic biological features of TNBC and resistance mechanisms to conventional therapies can support its aggressive behavior, characterizing TNBC how extremely heterogeneous. Novel combination strategies are under investigation, including immunotherapeutic agents, anti-drug conjugates, PARP inhibitors, and various targeting agents, exploring, in the meanwhile, possible predictive biomarkers to correctly select patients for the optimal treatment for their specific subtype. AREAS COVERED This article examines the main malignity characteristics across different subtype, both histological and molecular, and the resistance mechanisms, both primary and acquired, to different drugs explored in the landscape of TNBC treatment, that lead TNBC to still has high mortality rate. EXPERT OPINION The complexity of TNBC is not only the main reason of its aggressivity, but its heterogeneity should be exploited in terms of therapeutics opportunities, combining agents with different mechanism of action, after a correct selection by biologic or molecular biomarkers. The main goal is to understand what TNBC really is and to act selectively on its characteristics, with a personalized anticancer treatment.
Collapse
Affiliation(s)
- Maria Ilenia Passalacqua
- Division of Early Drug Development for Innovative Therapies, Ieo, European Institute of Oncology Irccs, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy.,Medical Oncology Unit, Department of Human Pathology G Barresi, University of Messina, Messina, Italy
| | - Graziella Rizzo
- Division of Early Drug Development for Innovative Therapies, Ieo, European Institute of Oncology Irccs, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy.,Medical Oncology Unit, Department of Human Pathology G Barresi, University of Messina, Messina, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology G Barresi, University of Messina, Messina, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, Ieo, European Institute of Oncology Irccs, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| |
Collapse
|
42
|
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
|
43
|
Antibody-Drug Conjugates in Breast Cancer: What Is Beyond HER2? Cancer J 2022; 28:436-445. [DOI: 10.1097/ppo.0000000000000629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
44
|
Williams SD, Smith TM, Stewart LV, Sakwe AM. Hypoxia-Inducible Expression of Annexin A6 Enhances the Resistance of Triple-Negative Breast Cancer Cells to EGFR and AR Antagonists. Cells 2022; 11:3007. [PMID: 36230969 PMCID: PMC9564279 DOI: 10.3390/cells11193007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
Physiological changes such as hypoxia in the tumor microenvironment (TME) endow cancer cells with malignant properties, leading to tumor recurrence and rapid progression. Here, we assessed the effect of hypoxia (1% Oxygen) on the tumor suppressor Annexin A6 (AnxA6) and the response of triple-negative breast cancer (TNBC) cells to epidermal growth factor receptor (EGFR) and androgen receptor (AR) targeted therapies. We demonstrate that brief exposure of TNBC cells to hypoxia (within 24 h) is associated with down regulation of AnxA6 while > 24 h exposure cell type dependently stimulated the expression of AnxA6. Hypoxia depicted by the expression and stability of HIF-1/2α led to up regulation of the HIF target genes SLC2A1, PGK1 as well as AR and the AR target genes FABP-4 and PPAR-γ, but the cellular levels of AnxA6 protein decreased under prolonged hypoxia. Down regulation of AnxA6 in TNBC cells inhibited, while AnxA6 over expression enhanced the expression and cellular levels of HIF-1/2α, SLC2A1 and PGK1. RNAi mediated inhibition of hypoxia induced AnxA6 expression also strongly inhibited glucose uptake and ROS production in AnxA6 expressing TNBC cells. Using a luciferase reporter assay, we confirm that short-term exposure of cells to hypoxia inhibits while prolonged exposure of cells to hypoxia enhances AnxA6 promoter activity in HEK293T cells. Compared to cells cultured under normoxia, TNBC cells were more resistant to lapatinib under hypoxic conditions, and the downregulation of AnxA6 sensitized the cells to EGFR as well as AR antagonists. These data suggest that AnxA6 is a hypoxia inducible gene and that targeting AnxA6 upregulation may be beneficial in overcoming TNBC resistance to EGFR and/or AR targeted therapies.
Collapse
Affiliation(s)
- Stephen D. Williams
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
| | - Tunde M. Smith
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
| | - LaMonica V. Stewart
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
- School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Amos M. Sakwe
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
45
|
Zhang J, Xia Y, Zhou X, Yu H, Tan Y, Du Y, Zhang Q, Wu Y. Current landscape of personalized clinical treatments for triple-negative breast cancer. Front Pharmacol 2022; 13:977660. [PMID: 36188535 PMCID: PMC9523914 DOI: 10.3389/fphar.2022.977660] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/05/2022] [Indexed: 11/30/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly malignant subtype of breast cancer (BC) with vicious behaviors. TNBC is usually associated with relatively poor clinical outcomes, earlier recurrence, and high propensity for visceral metastases than other BC types. TNBC has been increasingly recognized to constitute a very molecular heterogeneous subtype, which may offer additional therapeutic opportunities due to newly discovered cancer-causing drivers and targets. At present, there are multiple novel targeted therapeutic drugs in preclinical researches, clinical trial designs, and clinical practices, such as platinum drugs, poly ADP-ribose polymerase (PARP) inhibitors, immunocheckpoint inhibitors, androgen receptor inhibitors as well as PI3K/AKT/mTOR targeted inhibitors. These personalized, single, or combinational therapies based on molecular heterogeneity are currently showing positive results. The scope of this review is to highlight the latest knowledge about these potential TNBC therapeutic drugs, which will provide comprehensive insights into the personalized therapeutic strategies and options for combating TNBC.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Yu Xia
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomei Zhou
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Honghao Yu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufang Tan
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaying Du
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Ni Z, Xu S, Yu Z, Ye Z, Li R, Chen C, Yang J, Liu H, Zhou Z, Zhang X. Comparison of dual mTORC1/2 inhibitor AZD8055 and mTORC1 inhibitor rapamycin on the metabolism of breast cancer cells using proton nuclear magnetic resonance spectroscopy metabolomics. Invest New Drugs 2022; 40:1206-1215. [PMID: 36063263 DOI: 10.1007/s10637-022-01268-w] [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: 05/02/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022]
Abstract
Dual mTORC1/2 inhibitors may be more effective than mTORC1 inhibitor rapamycin. Nevertheless, their metabolic effects on breast cancer cells have not been reported. We compared the anti-proliferative capacity of rapamycin and a novel mTORC1/2 dual inhibitor (AZD8055) in two breast cancer cell lines (MDA-MB-231 and MDA-MB-453) and analyzed their metabolic effects using proton nuclear magnetic resonance (1H NMR) spectroscopy-based metabolomics. We found that AZD8055 more strongly inhibited breast cancer cell proliferation than rapamycin. The half-inhibitory concentration of AZD8055 in breast cancer cells was almost one-tenth that of rapamycin. We identified 22 and 23 metabolites from the 1H NMR spectra of MDA-MB-231 and MDA-MB-453 cells. The patterns of AZD8055- and rapamycin-treated breast cancer cells differed significantly; we then selected the metabolites that contributed to these differences. For inhibiting glycolysis and reducing glucose consumption, AZD8055 was likely to be more potent than rapamycin. For amino acids metabolism, although AZD8055 has a broad effect as rapamycin, their effects in degrees were not exactly the same. AZD8055 and rapamycin displayed cell-specific metabolic effects on breast cancer cells, a finding that deserves further study. These findings help fill the knowledge gap concerning dual mTORC1/2 inhibitors and provide a theoretical basis for their development.
Collapse
Affiliation(s)
- Zhitao Ni
- Clinical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shaolin Xu
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zheng Yu
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhongjiang Ye
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rongqi Li
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chuang Chen
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianhui Yang
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huamin Liu
- College of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziye Zhou
- Clinical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Xiuhua Zhang
- Clinical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
47
|
Drago JZ, Ferraro E, Abuhadra N, Modi S. Beyond HER2: Targeting the ErbB receptor family in breast cancer. Cancer Treat Rev 2022; 109:102436. [PMID: 35870237 PMCID: PMC10478787 DOI: 10.1016/j.ctrv.2022.102436] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 11/28/2022]
Abstract
Targeting the HER2 oncogene represents one of the greatest advances in the treatment of breast cancer. HER2 is one member of the ERBB-receptor family, which includes EGFR (HER1), HER3 and HER4. In the presence or absence of underling genomic aberrations such as mutations or amplification events, intricate interactions between these proteins on the cell membrane lead to downstream signaling that encourages cancer growth and proliferation. In this Review, we contextualize efforts to pharmacologically target the ErbB receptor family beyond HER2, with a focus on EGFR and HER3. Preclinical and clinical efforts are synthesized. We discuss successes and failures of this approach to date, summarize lessons learned, and propose a way forward that invokes new therapeutic modalities such as antibody drug conjugates (ADCs), combination strategies, and patient selection through rational biomarkers.
Collapse
Affiliation(s)
- Joshua Z Drago
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA.
| | - Emanuela Ferraro
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nour Abuhadra
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA
| | - Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA
| |
Collapse
|
48
|
Kohale IN, Yu J, Zhuang Y, Fan X, Reddy RJ, Sinnwell J, Kalari KR, Boughey JC, Carter JM, Goetz MP, Wang L, White FM. Identification of Src Family Kinases as Potential Therapeutic Targets for Chemotherapy-Resistant Triple Negative Breast Cancer. Cancers (Basel) 2022; 14:cancers14174220. [PMID: 36077757 PMCID: PMC9454481 DOI: 10.3390/cancers14174220] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/16/2022] [Accepted: 08/28/2022] [Indexed: 01/19/2023] Open
Abstract
Neoadjuvant chemotherapy (NAC) remains the cornerstone of the treatment for triple negative breast cancer (TNBC), with the goal of complete eradication of disease. However, for patients with residual disease after NAC, recurrence and mortality rates are high and the identification of novel therapeutic targets is urgently needed. We quantified tyrosine phosphorylation (pTyr)-mediated signaling networks in chemotherapy sensitive (CS) and resistant (CR) TNBC patient-derived xenografts (PDX), to gain novel therapeutic insights. The antitumor activity of SFK inhibition was examined in vivo. Treated tumors were further subjected to phosphoproteomic and RNAseq analysis, to identify the mechanism of actions of the drug. We identified Src Family Kinases (SFKs) as potential therapeutic targets in CR TNBC PDXs. Treatment with dasatinib, an FDA approved SFK inhibitor, led to inhibition of tumor growth in vivo. Further analysis of post-treatment PDXs revealed multiple mechanisms of actions of the drug, confirming the multi-target inhibition of dasatinib. Analysis of pTyr in tumor specimens suggested a low prevalence of SFK-driven tumors, which may provide insight into prior clinical trial results demonstrating a lack of dasatinib antitumor activity in unselected breast cancer patients. Taken together, these results underscore the importance of pTyr characterization of tumors, in identifying new targets, as well as stratifying patients based on their activated signaling networks for therapeutic options. Our data provide a strong rationale for studying SFK inhibitors in biomarker-selected SFK-driven TNBC.
Collapse
Affiliation(s)
- Ishwar N. Kohale
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jia Yu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Yongxian Zhuang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaoyang Fan
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Raven J. Reddy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jason Sinnwell
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Krishna R. Kalari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Judy C. Boughey
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Jodi M. Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew P. Goetz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Forest M. White
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Correspondence: ; Tel.: +617-258-8949
| |
Collapse
|
49
|
Abstract
Triple negative breast cancer (TNBC) is characterized by the lack of estrogen and progesterone receptor expression and lacks HER2 overexpression or gene amplification. It accounts for 10–15% of incident breast cancers and carries the worst prognosis. TNBC is overrepresented among Black and pre-menopausal women and is associated with significant psychological and treatment-related burdens, including financial toxicity. Like other breast cancers, TNBC is biologically heterogeneous, leading to diverse clinical and epidemiological behaviors, however, unlike the other clinical subtypes, in TNBC we still lack tumor-specific targeted therapy. Early TNBC outcomes have improved due to the intensification of therapies, including improvements in polychemotherapy and the addition of immunotherapy. Future efforts are needed to identify targetable aberrations for specific drug therapy, prevent immune evasion, and increase social-economic support. Given that the name TNBC illustrates its lack of specifically targeted and effective therapy, we look forward to being able to retire the name in favor of a group of targetable entities within what is now called “TNBC”.
Collapse
|
50
|
Zhou Y, Che Y, Fu Z, Zhang H, Wu H. Triple-Negative Breast Cancer Analysis Based on Metabolic Gene Classification and Immunotherapy. Front Public Health 2022; 10:902378. [PMID: 35875026 PMCID: PMC9296841 DOI: 10.3389/fpubh.2022.902378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/23/2022] [Indexed: 12/24/2022] Open
Abstract
Triple negative breast cancer (TNBC) has negative expression of ER, PR and HER-2. TNBC shows high histological grade and positive rate of lymph node metastasis, easy recurrence and distant metastasis. Molecular typing based on metabolic genes can reflect deeper characteristics of breast cancer and provide support for prognostic evaluation and individualized treatment. Metabolic subtypes of TNBC samples based on metabolic genes were determined by consensus clustering. CIBERSORT method was applied to evaluate the score distribution and differential expression of 22 immune cells in the TNBC samples. Linear discriminant analysis (LDA) established a subtype classification feature index. Kaplan-Meier (KM) and receiver operating characteristic (ROC) curves were generated to validate the performance of prognostic metabolic subtypes in different datasets. Finally, we used weighted correlation network analysis (WGCNA) to cluster the TCGA expression profile dataset and screen the co-expression modules of metabolic genes. Consensus clustering of the TCGA cohort/dataset obtained three metabolic subtypes (MC1, MC2, and MC3). The ROC analysis showed a high prognostic performance of the three clusters in different datasets. Specifically, MC1 had the optimal prognosis, MC3 had a poor prognosis, and the three metabolic subtypes had different prognosis. Consistently, the immune characteristic index established based on metabolic subtypes demonstrated that compared with the other two subtypes, MC1 had a higher IFNγ score, T cell lytic activity and lower angiogenesis score, T cell dysfunction and rejection score. TIDE analysis showed that MC1 patients were more likely to benefit from immunotherapy. MC1 patients were more sensitive to immune checkpoint inhibitors and traditional chemotherapy drugs Cisplatin, Paclitaxel, Embelin, and Sorafenib. Multiclass AUC based on RNASeq and GSE datasets were 0.85 and 0.85, respectively. Finally, based on co-expression network analysis, we screened 7 potential gene markers related to metabolic characteristic index, of which CLCA2, REEP6, SPDEF, and CRAT can be used to indicate breast cancer prognosis. Molecular classification related to TNBC metabolism was of great significance for comprehensive understanding of the molecular pathological characteristics of TNBC, contributing to the exploration of reliable markers for early diagnosis of TNBC and predicting metastasis and recurrence, improvement of the TNBC staging system, guiding individualized treatment.
Collapse
Affiliation(s)
- Yu Zhou
- Oncology Department, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Yingqi Che
- Hematology-Oncology Department, Long Nan Hospital, Daqing, China
| | - Zhongze Fu
- Gastroenterology Department, The First Hospital of Qiqihar, Qiqihar, China
| | - Henan Zhang
- Oncology Department, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Huiyu Wu
- Third Department of Oncology, People's Hospital of Daqing, Daqing, China
- *Correspondence: Huiyu Wu
| |
Collapse
|