1
|
van Kampen F, Clark A, Soul J, Kanhere A, Glenn MA, Pettitt AR, Kalakonda N, Slupsky JR. Deletion of 17p in cancers: Guilt by (p53) association. Oncogene 2025; 44:637-651. [PMID: 39966556 PMCID: PMC11876076 DOI: 10.1038/s41388-025-03300-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 01/17/2025] [Accepted: 02/04/2025] [Indexed: 02/20/2025]
Abstract
Monoallelic deletion of the short arm of chromosome 17 (del17p) is a recurrent abnormality in cancers with poor outcomes. Best studied in relation to haematological malignancies, associated functional outcomes are attributed mainly to loss and/or dysfunction of TP53, which is located at 17p13.1, but the wider impact of deletion of other genes located on 17p is poorly understood. 17p is one of the most gene-dense regions of the genome and includes tumour suppressor genes additional to TP53, genes essential for cell survival and proliferation, as well as small and long non-coding RNAs. In this review we utilise a data-driven approach to demarcate the extent of 17p deletion in multiple cancers and identify a common loss-of-function gene signature. We discuss how the resultant loss of heterozygosity (LOH) and haploinsufficiency may influence cell behaviour but also identify vulnerabilities that can potentially be exploited therapeutically. Finally, we highlight how emerging animal and isogenic cell line models of del17p can provide critical biological insights for cancer cell behaviour.
Collapse
Affiliation(s)
- Francisca van Kampen
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Abigail Clark
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Jamie Soul
- Computational Biology Facility, University of Liverpool, Liverpool, UK
| | - Aditi Kanhere
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Mark A Glenn
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Andrew R Pettitt
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Nagesh Kalakonda
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Joseph R Slupsky
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
2
|
Liu X, Zhao K, Zhang Z, Liu M, Chu H, Zou X. Clinicopathological characteristics and long-term prognosis of triple-negative breast cancer patients with HER2-Low expression: a retrospective propensity score-matched cohort study. J Cancer Res Clin Oncol 2024; 151:24. [PMID: 39729247 PMCID: PMC11680652 DOI: 10.1007/s00432-024-06069-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
PURPOSE The objective of the current research was to assess the clinicopathological characteristics and long-term prognosis of triple-negative breast cancer (TNBC) patients with human epidermal growth factor receptor 2 (HER2)-low status following breast surgery. METHODS A total of 202 TNBC patients treated at Qingdao Central Hospital from January 2010 to December 2019 were included, comprising 71 HER2-low and 131 HER2-zero patients. Propensity score matching (PSM) was applied to minimize differences between the cohorts. RESULTS HER2-low TNBC patients had lower histological grade, lower Ki-67 expression levels, and a higher prevalence of hypertension compared to HER2-zero TNBC patients. Before and after PSM, the HER2-low group consistently exhibited a lower recurrence rate and longer RFS compared to HER2-zero TNBC patients. HER2-low status was validated as an independent low-risk factor for RFS both pre-PSM (HR 0.354, 95% CI 0.178-0.706, p = 0.003) and post-PSM (HR 0.405, 95% CI 0.185-0.886, p = 0.024). No statistically significant differences in mortality rate and OS were observed, both before and after PSM. CONCLUSIONS HER2-low and HER2-zero TNBC patients show significant clinicopathological differences. Compared to HER2-zero, HER2-low status is linked to better long-term prognosis and serves as an independent low-risk factor for RFS in TNBC patients.
Collapse
Affiliation(s)
- Xin Liu
- Qingdao Medical College, Qingdao University, Qingdao, 266071, Shandong, China
- Department of Breast Surgery, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, 266042, Shandong, China
| | - Kaihua Zhao
- Department of Breast Surgery, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, 266042, Shandong, China
| | - Ziyan Zhang
- Department of Breast Surgery, Women and Children's Hospital, Qingdao University, Qingdao, 266034, Shandong, China
| | - Meiyan Liu
- Qingdao Medical College, Qingdao University, Qingdao, 266071, Shandong, China
| | - Hongwu Chu
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Xiao Zou
- Department of Breast Surgery, Xiangdong Hospital Affiliated to Hunan Normal University, Liling, 412200, Hunan, China.
| |
Collapse
|
3
|
Li H, Huang HQ, Huang ZG, He RQ, Fang YY, Song R, Luo JY, Zeng DT, Qin K, Wei DM, Chen G. Potential regulatory mechanism and clinical significance of synaptotagmin binding cytoplasmic RNA interacting protein in colorectal cancer. World J Clin Oncol 2024; 15:1412-1427. [PMID: 39582611 PMCID: PMC11514426 DOI: 10.5306/wjco.v15.i11.1412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/17/2024] [Accepted: 09/09/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) causes many deaths worldwide. Synaptotagmin binding cytoplasmic RNA interacting protein (SYNCRIP) is an RNA-binding protein that plays an important role in multiple cancers by epigenetically targeting some genes. Our study will examine the expression, potential effect, biological function and clinical value of SYNCRIP in CRC. AIM To examine the expression, potential effect, biological function and clinical value of SYNCRIP in CRC. METHODS The expression of SYNCRIP was examined by immunohistochemistry arrays and high-throughput data. The effect of SYNCRIP gene in CRC cell growth was evaluated by CRISPR-Cas9 technology. The target genes of SYNCRIP were calculated using various algorithms, and the molecular mechanism of SYNCRIP in CRC was explored by mutation analysis and pathway analysis. The clinical value of SYNCRIP in prognosis and radiotherapy was revealed via evidence-based medicine methods. RESULTS The protein and mRNA levels of SYNCRIP were both highly expressed in CRC samples compared to nontumorous tissue based on 330 immunohistochemistry arrays and 3640 CRC samples. Cells grew more slowly in eleven CRC cell lines after knocking out the SYNCRIP gene. SYNCRIP could epigenetically target genes to promote the occurrence and development of CRC by boosting the cell cycle and affecting the tumor microenvironment. In addition, CRC patients with high SYNCRIP expression are more sensitive to radiotherapy. CONCLUSION SYNCRIP is upregulated in CRC, and highly expressed SYNCRIP can accelerate CRC cell division by exerting its epigenetic regulatory effects. In addition, SYNCRIP is expected to become a potential biomarker to predict the effect of radiotherapy.
Collapse
Affiliation(s)
- Hui Li
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - He-Qing Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Guang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Ye-Ying Fang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Rui Song
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jia-Yuan Luo
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Da-Tong Zeng
- Department of Pathology, Redcross Hospital of Yulin City, Yulin 537000, Guangxi Zhuang Autonomous Region, China
| | - Kai Qin
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Dan-Ming Wei
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
4
|
Zou Y, Yang A, Chen B, Deng X, Xie J, Dai D, Zhang J, Tang H, Wu T, Zhou Z, Xie X, Wang J. crVDAC3 alleviates ferroptosis by impeding HSPB1 ubiquitination and confers trastuzumab deruxtecan resistance in HER2-low breast cancer. Drug Resist Updat 2024; 77:101126. [PMID: 39243601 DOI: 10.1016/j.drup.2024.101126] [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: 01/06/2024] [Revised: 07/24/2024] [Accepted: 08/05/2024] [Indexed: 09/09/2024]
Abstract
AIMS With the wide application of trastuzumab deruxtecan (T-DXd), the survival of HER2-low breast cancer patients is dramatically improved. However, resistance to T-DXd still exists in a subset of patients, and the molecular mechanism remains unclear. METHODS An in vivo shRNA lentiviral library functional screening was performed to identify potential circular RNA (crRNA) that mediates T-DXd resistance. RNA pull-down, mass spectrometry, RNA immunoprecipitation, and co-immunoprecipitation assays were conducted to investigate the molecular mechanism. Ferroptosis was detected using C11-BODIPY, Liperfluo, FerroOrange staining, glutathione quantification, malondialdehyde quantification, and transmission electron microscopy. Molecular docking, virtual screening, and patient-derived xenograft (PDX) models were used to validate therapeutic agents. RESULTS VDAC3-derived crRNA (crVDAC3) ranked first in functional shRNA library screening. Knockdown of crVDAC3 increased the sensitivity of HER2-low breast cancer cells to T-DXd treatment. Further mechanistic research revealed that crVDAC3 specifically binds to HSPB1 protein and inhibits its ubiquitination degradation, leading to intracellular accumulation and increased levels of HSPB1 protein. Notably, suppression of crVDAC3 dramatically increases excessive ROS levels and labile iron pool accumulation. Inhibition of crVDAC3 induces ferroptosis in breast cancer cells by reducing HSPB1 expression, thereby mediating T-DXd resistance. Through virtual screening and experimental validation, we identified that paritaprevir could effectively bind to crVDAC3 and prevent its interaction with HSPB1 protein, thereby increasing ubiquitination degradation of HSPB1 protein to overcome T-DXd resistance. Finally, we validated the enhanced therapeutic efficacy of T-DXd by paritaprevir in a HER2-low PDX model. CONCLUSION This finding reveals the molecular mechanisms underlying T-DXd resistance in HER2-low breast cancer. Our study provides a new strategy to overcome T-DXd resistance by inhibiting the interaction between crVDAC3 and HSPB1 protein.
Collapse
Affiliation(s)
- Yutian Zou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Anli Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Bo Chen
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xinpei Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jindong Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Danian Dai
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Jinhui Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Tao Wu
- Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde, China.
| | - Zhigang Zhou
- Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde, China.
| | - Xiaoming Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Jin Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
5
|
Wang Y, Yang R, Xie Y, Zhou XQ, Yang JF, Shi YY, Liu S. Comprehensive review of drug-mediated ICD inhibition of breast cancer: mechanism, status, and prospects. Clin Exp Med 2024; 24:230. [PMID: 39325106 PMCID: PMC11427550 DOI: 10.1007/s10238-024-01482-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/01/2024] [Indexed: 09/27/2024]
Abstract
The escalating incidence of breast cancer (BC) in women underscores its grave health threat. Current molecular insights into BC's post-adjuvant therapy cure remain elusive, necessitating active treatment explorations. Immunotherapy, notably chemotherapy-induced immunogenic cell death (ICD), has emerged as a promising BC therapy. ICD harnesses chemotherapeutics to activate anti-tumor immunity via DAMPs, fostering long-term T-cell memory and primary BC cure. Besides chemotherapy drugs, Nanodrugs, traditional Chinese medicine (TCM) and ICIs also induce ICD, boosting immune response. ICIs, like PD-1/PD-L1 inhibitors, revolutionize cancer treatment but face limited success in cold tumors. Thus, ICD induction combined with ICIs is studied extensively for BC immunotherapy. This article reviews the mechanism of ICD related drugs in BC and provides reference for the research and development of BC treatment, in order to explore more effective clinical treatment of BC, we hope to explore more ICD inducers and make ICIs more effective vaccines.
Collapse
Affiliation(s)
- Yang Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China
- China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Rui Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China
- Shanxi Province Cancer Hospital/Shanxi Hospital Afiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital, Afiliated to Shanxi Medical University, 030013, Shanxi, China
| | - Ying Xie
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China
| | - Xi-Qiu Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China
| | - Jian-Feng Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| | - You-Yang Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| | - Sheng Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
- Graduate School, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, China.
| |
Collapse
|
6
|
Dai C, Man Y, Zhang L, Zhang X, Xie C, Wang S, Zhang Y, Guo Q, Zou L, Hong H, Jiang L, Shi Y. Identifying SLC2A6 as the novel protective factor in breast cancer by TP53-related genes affecting M1 macrophage infiltration. Apoptosis 2024; 29:1211-1231. [PMID: 38622369 DOI: 10.1007/s10495-024-01964-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 04/17/2024]
Abstract
The high heterogeneity of breast cancer (BC) caused by pathogenic gene mutations poses a challenge to immunotherapy, but the underlying mechanism remains unknown. The difference in the infiltration of M1 macrophages induced by TP53 mutations has a significant impact on BC immunotherapy. The aim of this study was to develop a TP53-related M1 macrophage infiltration molecular typing risk signature in BC and evaluate the biological functions of the key gene to find new immunotherapy biomarkers. Weighted correlation network analysis (WGCNA) and negative matrix factorization (NMF) were used for distinguishing BC subtypes. The signature and the nomogram were both constructed and evaluated. Biological functions of the novel signature gene SLC2A6 were confirmed through in vitro and in vivo experiments. RNA-Sequencing and protein profiling were used for detecting the possible mechanism of SLC2A6. The results suggested that four BC subtypes were distinguished by TP53-related genes that affect M1 macrophage infiltration. The signature constructed by molecular typing characteristics could evaluate BC's clinical features and tumor microenvironment. The nomogram could accurately predict the prognosis. The signature gene SLC2A6 was found to have an abnormally low expression in tumor tissues. Overexpression of SLC2A6 could inhibit proliferation, promote mitochondrial damage, and result in apoptosis of tumor cells. The HSP70 family member protein HSPA6 could bind with SLC2A6 and increase with the increased expression of SLC2A6. In summary, the risk signature provides a reference for BC risk assessment, and the signature gene SLC2A6 could act as a tumor suppressor in BC.
Collapse
Affiliation(s)
- Chao Dai
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yuxin Man
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Luhan Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Xiao Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Chunbao Xie
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Shan Wang
- National Center for Integrated Traditional and Western Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yinjie Zhang
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Qian Guo
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Liang Zou
- School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Huangming Hong
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| | - Lingxi Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
7
|
El Haddad G, Diab E, Hajjar M, Aoun M, Mallat F, Zalaquett Z, Kourie HR. Insights Into the Emerging Entity of HER2-Low Breast Cancer. Int J Breast Cancer 2024; 2024:2853007. [PMID: 38962672 PMCID: PMC11221987 DOI: 10.1155/2024/2853007] [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: 02/22/2024] [Revised: 04/25/2024] [Accepted: 05/10/2024] [Indexed: 07/05/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2)-low breast cancer (BC) is a subtype of BC that has been recently recognized as a separate clinical entity with distinct clinical and molecular characteristics. It is defined by a low level of HER2 protein expression, which distinguishes it from other more aggressive BC subtypes. Early studies suggest that it may have a more favorable prognosis than HER2-positive BC, as it is less likely to spread to other parts of the body and may be more responsive to standard BC treatments such as chemotherapy, radiation therapy, and hormone therapy. Given the relative new emergence of HER2-low BC, there is still much to be learned about this subtype; ongoing research is focused on identifying the underlying genetic mutations that contribute to HER2-low BC as well as developing targeted therapies that can improve outcomes for patients with this disease. This review is aimed at summarizing the current clinical knowledge on HER2-low BC, with the aim of creating a better understanding of this entity and paving the way for potential interventions and a new standard of care.
Collapse
Affiliation(s)
- Georges El Haddad
- Hematology-Oncology DepartmentHôtel-Dieu de France University HospitalSaint Joseph University of Beirut, Beirut, Lebanon
| | - Ernest Diab
- Hematology-Oncology DepartmentHôtel-Dieu de France University HospitalSaint Joseph University of Beirut, Beirut, Lebanon
| | - Michel Hajjar
- Hematology-Oncology DepartmentHôtel-Dieu de France University HospitalSaint Joseph University of Beirut, Beirut, Lebanon
| | - Maroun Aoun
- Hematology-Oncology DepartmentHôtel-Dieu de France University HospitalSaint Joseph University of Beirut, Beirut, Lebanon
| | - Farid Mallat
- Hematology-Oncology DepartmentHôtel-Dieu de France University HospitalSaint Joseph University of Beirut, Beirut, Lebanon
| | - Ziad Zalaquett
- Hematology-Oncology DepartmentHôtel-Dieu de France University HospitalSaint Joseph University of Beirut, Beirut, Lebanon
| | - Hampig-Raphael Kourie
- Hematology-Oncology DepartmentHôtel-Dieu de France University HospitalSaint Joseph University of Beirut, Beirut, Lebanon
| |
Collapse
|
8
|
Ning D, Xue J, Lou X, Shao R, Liu Y, Chen G. Transforming toxins into treatments: the revolutionary role of α-amanitin in cancer therapy. Arch Toxicol 2024; 98:1705-1716. [PMID: 38555326 DOI: 10.1007/s00204-024-03727-0] [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: 01/17/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Amanita phalloides is the primary species responsible for fatal mushroom poisoning, as its main toxin, α-amanitin, irreversibly and potently inhibits eukaryotic RNA polymerase II (RNAP II), leading to cell death. There is no specific antidote for α-amanitin, which hinders its clinical application. However, with the advancement of precision medicine in oncology, including the development of antibody-drug conjugates (ADCs), the potential value of various toxic small molecules has been explored. These ADCs ingeniously combine the targeting precision of antibodies with the cytotoxicity of small-molecule payloads to precisely kill tumor cells. We searched PubMed for studies in this area using these MeSH terms "Amanitins, Alpha-Amanitin, Therapeutic use, Immunotherapy, Immunoconjugates, Antibodies" and did not limit the time interval. Recent studies have conducted preclinical experiments on ADCs based on α-amanitin, showing promising therapeutic effects and good tolerance in primates. The current challenges include the not fully understood toxicological mechanism of α-amanitin and the lack of clinical studies to evaluate the therapeutic efficacy of ADCs developed based on α-amanitin. In this article, we will discuss the role and therapeutic efficacy of α-amanitin as an effective payload in ADCs for the treatment of various cancers, providing background information for the research and application strategies of current and future drugs.
Collapse
Affiliation(s)
- Deyuan Ning
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jinfang Xue
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Xiran Lou
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Ruifei Shao
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yu Liu
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Guobing Chen
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, No 157 Jinbi Road, Xishan District, Kunming, 650032, China.
| |
Collapse
|
9
|
Reshetnyak YK, Andreev OA, Engelman DM. Aiming the magic bullet: targeted delivery of imaging and therapeutic agents to solid tumors by pHLIP peptides. Front Pharmacol 2024; 15:1355893. [PMID: 38545547 PMCID: PMC10965573 DOI: 10.3389/fphar.2024.1355893] [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: 12/14/2023] [Accepted: 01/05/2024] [Indexed: 11/11/2024] Open
Abstract
The family of pH (Low) Insertion Peptides (pHLIP) comprises a tumor-agnostic technology that uses the low pH (or high acidity) at the surfaces of cells within the tumor microenvironment (TME) as a targeted biomarker. pHLIPs can be used for extracellular and intracellular delivery of a variety of imaging and therapeutic payloads. Unlike therapeutic delivery targeted to specific receptors on the surfaces of particular cells, pHLIP targets cancer, stromal and some immune cells all at once. Since the TME exhibits complex cellular crosstalk interactions, simultaneous targeting and delivery to different cell types leads to a significant synergistic effect for many agents. pHLIPs can also be positioned on the surfaces of various nanoparticles (NPs) for the targeted intracellular delivery of encapsulated payloads. The pHLIP technology is currently advancing in pre-clinical and clinical applications for tumor imaging and treatment.
Collapse
Affiliation(s)
- Yana K. Reshetnyak
- Physics Department, University of Rhode Island, Kingston, RI, United States
| | - Oleg A. Andreev
- Physics Department, University of Rhode Island, Kingston, RI, United States
| | - Donald M. Engelman
- Molecular Biophysics and Biochemistry Department, Yale, New Haven, CT, United States
| |
Collapse
|
10
|
Liu CC, Wolf M, Ortego R, Grencewicz D, Sadler T, Eng C. Characterization of immunomodulating agents from Staphylococcus aureus for priming immunotherapy in triple-negative breast cancers. Sci Rep 2024; 14:756. [PMID: 38191648 PMCID: PMC10774339 DOI: 10.1038/s41598-024-51361-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/03/2024] [Indexed: 01/10/2024] Open
Abstract
Immunotherapy, specifically immune checkpoint blockade (ICB), has revolutionized the treatment paradigm of triple-negative breast cancers (TNBCs). However, a subset of TNBCs devoid of tumor-infiltrating T cells (TILs) or PD-L1 expression generally has a poor response to immunotherapy. In this study, we aimed to sensitize TNBCs to ICB by harnessing the immunomodulating potential of S. aureus, a breast-resident bacterium. We show that intratumoral injection of spent culture media from S. aureus recruits TILs and suppresses tumor growth in a preclinical TNBC model. We further demonstrate that α-hemolysin (HLA), an S. aureus-produced molecule, increases the levels of CD8+ T cells and PD-L1 expression in tumors, delays tumor growth, and triggers tumor necrosis. Mechanistically, while tumor cells treated with HLA display Gasdermin E (GSDME) cleavage and a cellular phenotype resembling pyroptosis, splenic T cells incubated with HLA lead to selective expansion of CD8+ T cells. Notably, intratumoral HLA injection prior to ICB augments the therapeutic efficacy compared to ICB alone. This study uncovers novel immunomodulatory properties of HLA and suggests that intratumoral administration of HLA could be a potential priming strategy to expand the population of TNBC patients who may respond to ICB.
Collapse
Affiliation(s)
- Chin-Chih Liu
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA
| | - Matthew Wolf
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA
| | - Ruth Ortego
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA
| | - Dennis Grencewicz
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA
| | - Tammy Sadler
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA
| | - Charis Eng
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.
- Cleveland Clinic, Center for Personalized Genetic Healthcare, Medical Specialties Institute, Cleveland, OH, 44195, USA.
- Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, 44195, USA.
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Germline High Risk Cancer Focus Group, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
11
|
Jiang M, Liu J, Li Q, Xu B. The trichotomy of HER2 expression confers new insights into the understanding and managing for breast cancer stratified by HER2 status. Int J Cancer 2023; 153:1324-1336. [PMID: 37314204 DOI: 10.1002/ijc.34570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 06/15/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a tyrosine kinase receptor that plays a carcinogenic role in breast cancer (BC) through gene amplification, mutation, or overexpression. Traditional methods of HER2 detection were divided into positive (immunohistochemistry (IHC) 3+/fluorescence in situ hybridization (FISH) amplification) and negative (IHC 2+/FISH-, IHC 1+, IHC 0) according to the dichotomy method. Anti-HER2-targeted therapies, such as trastuzumab and pertuzumab, have significantly improved the prognosis of HER2-positive patients. However, up to 75% to 85% of patients remain HER2-negative. In recent years, with the rapid development of molecular biology, gene detection technology, targeted therapy, and immunotherapy, researchers have actively explored the clinicopathological characteristics, molecular biological characteristics, treatment methods, and HER2 detection methods of HER2-low/zero breast cancer. With the clinical efficacy of new anti-HER2 targeted drugs, accurate classification of breast cancer is very important for the treatment choice. Therefore, the following review summarizes the necessity of developing HER2 detection methods, and the clinicopathological and drug treatment characteristics of patients with HER2-low/zero, to light the dawn of the treatment of breast cancer patients with HER2-low/zero expression.
Collapse
Affiliation(s)
- Mingxia Jiang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaxuan Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiao Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Li Z, Guo S, Xue H, Li L, Guo Y, Duan S, Zhu H. Efficacy and safety of trastuzumab deruxtecan in the treatment of HER2-low/positive advanced breast cancer: a single-arm meta-analysis. Front Pharmacol 2023; 14:1183514. [PMID: 37426807 PMCID: PMC10324614 DOI: 10.3389/fphar.2023.1183514] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Background: Clinical trials have shown that the use of trastuzumab deruxtecan (DS-8201) alone is expected to provide novel therapeutic options for HER2-low/positive patients. Nevertheless, there are some variations in the efficacy of trial results, with potential risks at the safety level. Most DS-8201 trials in HER2 advanced breast cancer (ABC) have been conducted in the form of small-sample nonrandomized controlled studies, resulting in a lack of validated indicators to evaluate the efficacy and safety of DS-8201. Thus, this meta-analysis aimed to pool the results of various trials of DS-8201 alone to explore the efficacy and safety of DS-8201 in patients with HER2-low/positive advanced breast cancer. Methods: Relevant studies were searched in seven databases, including Embase, PubMed, Web of Science, Cochrane Library, CNKI, VIP database and WanFang data, to collect single-arm studies on DS-8201 for HER2-low/positive ABC. MINORS was adopted for quality assessment and STATA 16.0 for data analysis. Results: Ten studies involving 1,108 patients were included in this meta-analysis. As for the tumor response rate, the pooled ORR and DCR of all studies reached 57% (95% CI: 47%-67%) and 92% (95% CI: 89%-96%) respectively, and the pooled ORRs of the HER2-low expression group and the HER2-positive expression group were 46% (95% CI: 35%-56%) and 64% (95% CI: 54%-74%). Only the low expression group achieved median survival time, with a pooled median PFS and median OS of 9.24 (95% CI: 7.54-10.94) months and 23.87 (95% CI: 21.56-26.17) months, respectively. The most common treatment-related adverse events from DS-8201 were nausea (all grades: 62%; ≥ grade III: 5%), fatigue (all grade: 44%; ≥ grade III: 6%), and alopecia (all grades: 38%; ≥ grade III: 0.5%). Drug-related interstitial lung disease or pneumonitis occurred in 13% of the 1,108 patients, with only a 1% incidence of AE ≥ grade III. Conclusion: The present study suggests that DS-8201 is effective and safe in the treatment of ABC with low or positive HER2 expression, providing additional relevant information for its clinical application. However, further strengthening of the pairs is needed, as well as more clinical studies to support individualized treatment. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42023390316.
Collapse
Affiliation(s)
- Zongyu Li
- Clinical Medical Research Institute, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - Shangwen Guo
- Clinical Medical Research Institute, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - Haoyi Xue
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - Luying Li
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - Yuyuan Guo
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - Sinuo Duan
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - He Zhu
- Clinical Medical Research Institute, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
13
|
Han Y, Wang J, Xu B. Cytotoxic Lymphocyte-Related Gene Signature in Triple-Negative Breast Cancer. J Pers Med 2023; 13:457. [PMID: 36983639 PMCID: PMC10054905 DOI: 10.3390/jpm13030457] [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: 01/12/2023] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023] Open
Abstract
To curate the signature genes of cytotoxic lymphocytes (CLs) and explore the heterogeneity based on the CL-related (CLR) gene signature, we analyzed the gene expression of 592 patients with histologically diagnosed triple-negative breast cancer. Based on the 13-gene panel, CLR signatures were curated and associated with the stage of tumor size. Patients in the CLR-low group exhibited the worse overall survival (OS) (median OS, 75.23 months vs. 292.66 months, p < 0.0001) and were characterized by the upregulation of the NF-κB, Wnt, and p53 pathways, the positive regulation of angiogenesis, and a higher expression of immune checkpoints including CTLA4, LAG3, CD86, ICOS, ICOSLG, and TNFSF9. In cancer immunotherapy cohorts (GSE157284, GSE35640, IMvigor210), a higher CLR signature score was remarkably associated with greater tumor shrinkage and immune characteristics consisting of higher PD-L1 and neoantigen expression, as well as an inflamed tumor microenvironment. In the pan-cancer atlas, the CLR signature was notably associated with patient survival and revealed a profound heterogeneity across the malignancy types. In sum, the CLR signature is a promising indicator for immune characteristics, tumor shrinkage, and survival outcomes following cancer immunotherapy in addition to the prognostic heterogeneity in the pan-cancer atlas.
Collapse
Affiliation(s)
| | - Jiayu Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research, Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China; (Y.H.); (B.X.)
| | | |
Collapse
|
14
|
van der Noord VE, van de Water B, Le Dévédec SE. Targeting the Heterogeneous Genomic Landscape in Triple-Negative Breast Cancer through Inhibitors of the Transcriptional Machinery. Cancers (Basel) 2022; 14:4353. [PMID: 36139513 PMCID: PMC9496798 DOI: 10.3390/cancers14184353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer defined by lack of the estrogen, progesterone and human epidermal growth factor receptor 2. Although TNBC tumors contain a wide variety of oncogenic mutations and copy number alterations, the direct targeting of these alterations has failed to substantially improve therapeutic efficacy. This efficacy is strongly limited by interpatient and intratumor heterogeneity, and thereby a lack in uniformity of targetable drivers. Most of these genetic abnormalities eventually drive specific transcriptional programs, which may be a general underlying vulnerability. Currently, there are multiple selective inhibitors, which target the transcriptional machinery through transcriptional cyclin-dependent kinases (CDKs) 7, 8, 9, 12 and 13 and bromodomain extra-terminal motif (BET) proteins, including BRD4. In this review, we discuss how inhibitors of the transcriptional machinery can effectively target genetic abnormalities in TNBC, and how these abnormalities can influence sensitivity to these inhibitors. These inhibitors target the genomic landscape in TNBC by specifically suppressing MYC-driven transcription, inducing further DNA damage, improving anti-cancer immunity, and preventing drug resistance against MAPK and PI3K-targeted therapies. Because the transcriptional machinery enables transcription and propagation of multiple cancer drivers, it may be a promising target for (combination) treatment, especially of heterogeneous malignancies, including TNBC.
Collapse
Affiliation(s)
| | | | - Sylvia E. Le Dévédec
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
15
|
Moshnikova A, Golijanin B, Amin A, Doyle J, Kott O, Gershman B, DuPont M, Li Y, Lu X, Engelman DM, Andreev OA, Reshetnyak YK, Golijanin D. Targeting Bladder Urothelial Carcinoma with pHLIP-ICG and Inhibition of Urothelial Cancer Cell Proliferation by pHLIP-amanitin. FRONTIERS IN UROLOGY 2022; 2:868919. [PMID: 36439552 PMCID: PMC9691284 DOI: 10.3389/fruro.2022.868919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Acidity is a useful biomarker for the targeting of metabolically active-cells in tumors. pH Low Insertion Peptides (pHLIPs) sense the pH at the surfaces of tumor cells and can facilitate intracellular delivery of cell-permeable and cell-impermeable cargo molecules. In this study we have shown the targeting of malignant lesions in human bladders by fluorescent pHLIP agents, intracellular delivery of amanitin toxin by pHLIP for the inhibition of urothelial cancer cell proliferation, and enhanced potency of pHLIP-amanitin for cancer cells with 17p loss, a mutation frequently present in urothelial cancers. Twenty-eight ex-vivo bladder specimens, from patients undergoing robotic assisted laparoscopic radical cystectomy for bladder cancer, were treated via intravesical incubation for 15-60 minutes with pHLIP conjugated to indocyanine green (ICG) or IR-800 near infrared fluorescent (NIRF) dyes at concentrations of 4-8 μM. White light cystoscopy identified 47/58 (81%) and NIRF pHLIP cystoscopy identified 57/58 (98.3%) of malignant lesions of different subtypes and stages selected for histopathological processing. pHLIP NIRF imaging improved diagnosis by 17.3% (p < 0.05). All carcinoma-in-situ cases missed by white light cystoscopy were targeted by pHLIP agents and were diagnosed by NIRF imaging. We also investigated the interactions of pHLIP-amanitin with urothelial cancer cells of different grades. pHLIP-amanitin produced concentration- and pH-dependent inhibition of the proliferation of urothelial cancer cells treated for 2 hrs at concentrations up to 4 μM. A 3-4x enhanced cytotoxicity of pHLIP-amanitin was observed for cells with a 17p loss after 2 hrs of treatment at pH6. Potentially, pHLIP technology may improve the management of urothelial cancers, including imaging of malignant lesions using pHLIP-ICG for diagnosis and surgery, and the use of pHLIP-amanitin for treatment of superficial bladder cancers via intravesical instillation.
Collapse
Affiliation(s)
- Anna Moshnikova
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | - Borivoj Golijanin
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, The Miriam Hospital, Providence, RI, USA
- Division of Urology, Department of Surgery, Brown University, The Miriam Hospital, Providence, RI, USA
| | - Ali Amin
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, The Miriam Hospital, Providence, RI, USA
| | - Joshua Doyle
- Physics Department, University of Rhode Island, Kingston, RI, USA
- Current address: Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ohad Kott
- Division of Urology, Department of Surgery, Brown University, The Miriam Hospital, Providence, RI, USA
| | - Boris Gershman
- Division of Urology, Department of Surgery, Brown University, The Miriam Hospital, Providence, RI, USA
- Current address: Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michael DuPont
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | - Yujing Li
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Melvin & Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Donald M. Engelman
- Department of Molecular Biophysics and Biochemistry, Yale, New Haven, CT, USA
| | - Oleg A. Andreev
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | | | - Dragan Golijanin
- Division of Urology, Department of Surgery, Brown University, The Miriam Hospital, Providence, RI, USA
| |
Collapse
|
16
|
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
|
17
|
Todorovic M, Rivollier P, Wong AAWL, Wang Z, Pryyma A, Nguyen TT, Newell KC, Froelich J, Perrin DM. Rationally Designed Amanitins Achieve Enhanced Cytotoxicity. J Med Chem 2022; 65:10357-10376. [PMID: 35696491 DOI: 10.1021/acs.jmedchem.1c02226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
For 70 years, α-amanitin, the most cytotoxic peptide in its class, has been without a synthetic rival; through synthesis, we address the structure-activity relationships to inform the design of new amatoxins and disclose analogues that are more cytotoxic than the natural product when evaluated on CHO, HEK293, and HeLa cells, whereas on liver-derived HepG2 cells, the same toxins show diminished cytotoxicity.
Collapse
Affiliation(s)
- Mihajlo Todorovic
- Chemistry Department, 2036 Main Mall, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T-1Z1, Canada
| | - Paul Rivollier
- Chemistry Department, 2036 Main Mall, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T-1Z1, Canada
| | - Antonio A W L Wong
- Chemistry Department, 2036 Main Mall, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T-1Z1, Canada
| | - Zhou Wang
- Chemistry Department, 2036 Main Mall, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T-1Z1, Canada
| | - Alla Pryyma
- Chemistry Department, 2036 Main Mall, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T-1Z1, Canada
| | - Tuan Trung Nguyen
- Chemistry Department, 2036 Main Mall, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T-1Z1, Canada
| | - Kayla C Newell
- Chemistry Department, 2036 Main Mall, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T-1Z1, Canada
| | - Juliette Froelich
- Chemistry Department, 2036 Main Mall, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T-1Z1, Canada
| | - David M Perrin
- Chemistry Department, 2036 Main Mall, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T-1Z1, Canada
| |
Collapse
|
18
|
Zhao W, Liu J, Li Y, Chen Z, Qi D, Zhang Z. Immune Effect of Active Components of Traditional Chinese Medicine on Triple-Negative Breast Cancer. Front Pharmacol 2021; 12:731741. [PMID: 34925002 PMCID: PMC8678494 DOI: 10.3389/fphar.2021.731741] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/18/2021] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancers are heterogeneous, poorly prognostic, and metastatic malignancies that result in a high risk of death for patients. Targeted therapy for triple-negative breast cancer has been extremely challenging due to the lack of expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. Clinical treatment regimens for triple-negative breast cancer are often based on paclitaxel and platinum drugs, but drug resistance and side effects from the drugs frequently lead to treatment failure, thus requiring the development of new therapeutic platforms. In recent years, research on traditional Chinese medicine in modulating the immune function of the body has shown that it has the potential to be an effective treatment option against triple-negative breast cancer. Active components of herbal medicines such as alkaloids, flavonoids, polyphenols, saponins, and polysaccharides have been shown to inhibit cancer cell proliferation and metastasis by activating inflammatory immune responses and can modulate tumor-related signaling pathways to further inhibit the invasion of triple-negative breast cancer. This paper reviews the immunomodulatory mechanisms of different herbal active ingredients against triple-negative breast cancer and provides an outlook on the challenges and directions of development for the treatment of triple-negative breast cancer with herbal active ingredients.
Collapse
Affiliation(s)
- Wenjie Zhao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinhua Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yaqun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zichao Chen
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dongmei Qi
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
19
|
Bianchini G, De Angelis C, Licata L, Gianni L. Treatment landscape of triple-negative breast cancer - expanded options, evolving needs. Nat Rev Clin Oncol 2021; 19:91-113. [PMID: 34754128 DOI: 10.1038/s41571-021-00565-2] [Citation(s) in RCA: 640] [Impact Index Per Article: 160.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 12/13/2022]
Abstract
Tumour heterogeneity and a long-standing paucity of effective therapies other than chemotherapy have contributed to triple-negative breast cancer (TNBC) being the subtype with the least favourable outcomes. In the past few years, advances in omics technologies have shed light on the relevance of the TNBC microenvironment heterogeneity, unveiling a close dynamic relationship with cancer cell features. An improved understanding of tumour-immune system co-evolution supports the need to adopt a more comprehensive view of TNBC as an ecosystem that encompasses the intrinsic and extrinsic features of cancer cells. This new appreciation of the biology of TNBC has already led to the development of novel targeted agents, including PARP inhibitors, antibody-drug conjugates and immune-checkpoint inhibitors, which are revolutionizing the therapeutic landscape and providing new opportunities both for patients with early-stage TNBC and for those with advanced-stage disease. The current therapeutic scenario is only the tip of the iceberg, as hundreds of new compounds and combinations are in development. The translation of these experimental therapies into clinical benefit is a welcome and ongoing challenge. In this Review, we describe the current and upcoming therapeutic landscape of TNBC and discuss how an integrated view of the TNBC ecosystem can define different levels of risk and provide improved opportunities for tailoring treatment.
Collapse
Affiliation(s)
- Giampaolo Bianchini
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy. .,Università Vita-Salute San Raffaele, Milan, Italy.
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy.,Laster and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Luca Licata
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | |
Collapse
|
20
|
Kobayashi D, Kohmura Y, Sugiki T, Kuraoka E, Denda M, Fujiwara T, Otaka A. Peptide Cyclization Mediated by Metal-Free S-Arylation: S-Protected Cysteine Sulfoxide as an Umpolung of the Cysteine Nucleophile. Chemistry 2021; 27:14092-14099. [PMID: 34302308 DOI: 10.1002/chem.202102420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Indexed: 11/07/2022]
Abstract
Covalent linking of side chains provides a method to produce cyclic or stapled peptides that are important in developing peptide-based drugs. A variety of crosslinking formats contribute to fixing the active conformer and prolonging its biological activity under physiological conditions. One format uses the cysteine thiol to participate in crosslinking through nucleophilic thiolate anions or thiyl radicals to form thioether and disulfide bonds. Removal of the S-protection from an S-protected Cys derivative generates the thiol, which functions as a nucleophile. S-Oxidation of a protected Cys allows the formation of a sulfoxide that operates as an umpolung electrophile. Herein, the applicability of S-p-methoxybenzyl Cys sulfoxide (Cys(MBzl)(O)) to the formation of a thioether linkage between tryptophan and Cys has been investigated. The reaction of peptides containing Cys(MBzl)(O) and Trp with trifluoromethanesulfonic acid (TFMSA) or methanesulfonic acid (MSA) in TFA in the presence of guanidine hydrochloride (Gn ⋅ HCl) proceeded to give cyclic or stapled peptides possessing the Cys-Trp thioether linkage. In this reaction, strong acids such as TFMSA or MSA are necessary to activate the sulfoxide. Additionally, Gn ⋅ HCl plays a critical role in producing an electrophilic Cys derivative that combines with the indole by aromatic electrophilic substitution. The findings led us to conclude that the less-electrophilic Cys(MBzl)(O) serves as an acid-activated umpolung of a Cys nucleophile and is useful for S-arylation-mediated peptide cyclization.
Collapse
Affiliation(s)
- Daishiro Kobayashi
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Shomachi, Tokushima, 770-8505, Japan
| | - Yutaka Kohmura
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Shomachi, Tokushima, 770-8505, Japan
| | - Toshihiko Sugiki
- Institute of Protein Research, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Eisuke Kuraoka
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Shomachi, Tokushima, 770-8505, Japan
| | - Masaya Denda
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Shomachi, Tokushima, 770-8505, Japan
| | - Toshimichi Fujiwara
- Institute of Protein Research, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Akira Otaka
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Shomachi, Tokushima, 770-8505, Japan
| |
Collapse
|