101
|
Oropeza E, Seker S, Carrel S, Mazumder A, Lozano D, Jimenez A, VandenHeuvel SN, Noltensmeyer DA, Punturi NB, Lei JT, Lim B, Waltz SE, Raghavan SA, Bainbridge MN, Haricharan S. Molecular portraits of cell cycle checkpoint kinases in cancer evolution, progression, and treatment responsiveness. SCIENCE ADVANCES 2023; 9:eadf2860. [PMID: 37390209 PMCID: PMC10313178 DOI: 10.1126/sciadv.adf2860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 05/26/2023] [Indexed: 07/02/2023]
Abstract
Cell cycle dysregulation is prerequisite for cancer formation. However, it is unknown whether the mode of dysregulation affects disease characteristics. Here, we conduct comprehensive analyses of cell cycle checkpoint dysregulation using patient data and experimental investigations. We find that ATM mutation predisposes the diagnosis of primary estrogen receptor (ER)+/human epidermal growth factor (HER)2- cancer in older women. Conversely, CHK2 dysregulation induces formation of metastatic, premenopausal ER+/HER2- breast cancer (P = 0.001) that is treatment-resistant (HR = 6.15, P = 0.01). Lastly, while mutations in ATR alone are rare, ATR/TP53 co-mutation is 12-fold enriched over expected in ER+/HER2- disease (P = 0.002) and associates with metastatic progression (HR = 2.01, P = 0.006). Concordantly, ATR dysregulation induces metastatic phenotypes in TP53 mutant, not wild-type, cells. Overall, we identify mode of cell cycle dysregulation as a distinct event that determines subtype, metastatic potential, and treatment responsiveness, providing rationale for reconsidering diagnostic classification through the lens of the mode of cell cycle dysregulation..
Collapse
Affiliation(s)
- Elena Oropeza
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sinem Seker
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sabrina Carrel
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Aloran Mazumder
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Daniel Lozano
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Athena Jimenez
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | | | - Nindo B. Punturi
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jonathan T. Lei
- Lester and Sue Smith Breast Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Bora Lim
- Lester and Sue Smith Breast Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Oncology/Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
- Research Service, Cincinnati Veteran's Affairs Medical Center, 3200 Vine St., Cincinnati, OH, USA
| | | | | | - Svasti Haricharan
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
102
|
El Hejjioui B, Lamrabet S, Amrani Joutei S, Senhaji N, Bouhafa T, Malhouf MA, Bennis S, Bouguenouch L. New Biomarkers and Treatment Advances in Triple-Negative Breast Cancer. Diagnostics (Basel) 2023; 13:diagnostics13111949. [PMID: 37296801 DOI: 10.3390/diagnostics13111949] [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: 12/31/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 06/12/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a specific subtype of breast cancer lacking hormone receptor expression and HER2 gene amplification. TNBC represents a heterogeneous subtype of breast cancer, characterized by poor prognosis, high invasiveness, high metastatic potential, and a tendency to relapse. In this review, the specific molecular subtypes and pathological aspects of triple-negative breast cancer are illustrated, with particular attention to the biomarker characteristics of TNBC, namely: regulators of cell proliferation and migration and angiogenesis, apoptosis-regulating proteins, regulators of DNA damage response, immune checkpoints, and epigenetic modifications. This paper also focuses on omics approaches to exploring TNBC, such as genomics to identify cancer-specific mutations, epigenomics to identify altered epigenetic landscapes in cancer cells, and transcriptomics to explore differential mRNA and protein expression. Moreover, updated neoadjuvant treatments for TNBC are also mentioned, underlining the role of immunotherapy and novel and targeted agents in the treatment of TNBC.
Collapse
Affiliation(s)
- Brahim El Hejjioui
- Biomedical and Translational Research Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
- Department of Medical Genetics and Oncogenetics, HASSAN II University Hospital, Fez 30050, Morocco
| | - Salma Lamrabet
- Biomedical and Translational Research Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Sarah Amrani Joutei
- Department of Radiotherapy, HASSAN II University Hospital, Fez 30050, Morocco
| | - Nadia Senhaji
- Faculty of Sciences, Moulay Ismail University, Meknès 50000, Morocco
| | - Touria Bouhafa
- Department of Radiotherapy, HASSAN II University Hospital, Fez 30050, Morocco
| | | | - Sanae Bennis
- Biomedical and Translational Research Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Laila Bouguenouch
- Department of Medical Genetics and Oncogenetics, HASSAN II University Hospital, Fez 30050, Morocco
| |
Collapse
|
103
|
Colomer R, Miranda J, Romero-Laorden N, Hornedo J, González-Cortijo L, Mouron S, Bueno MJ, Mondéjar R, Quintela-Fandino M. Usefulness and real-world outcomes of next generation sequencing testing in patients with cancer: an observational study on the impact of selection based on clinical judgement. EClinicalMedicine 2023; 60:102029. [PMID: 37304496 PMCID: PMC10248077 DOI: 10.1016/j.eclinm.2023.102029] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Background Next Generation Sequencing (NGS) panels are increasingly used in advanced patients with cancer to guide therapy. There is, however, controversy about when should these panels be used, and about their impact on the clinical course. Methods In an observational study of 139 patients with cancer having an NGS test [from January 1st, 2017 to December 30th, 2020, in two hospitals (Hospital Universitario de La Princesa and Hospital Universitario Quironsalud Madrid) from Spain], we evaluated whether the clinical course (progression-free survival, PFS) was influenced by drug-based criteria [druggable alterations, receiving a recommended drug, having a favourable ESCAT category (ESMO Scale for Clinical Actionability of molecular Targets)] or clinical judgement criteria. Findings In 111 of 139 cases that were successfully profiled, PFS was not significantly influenced by either having druggable alterations [median PFS for patients with druggable alterations was 170 (95% C.I.: 139-200) days compared to 299 (95% C.I.: 114-483) for those without; p = 0.37], receiving a proposed matching agent [median PFS for patients receiving a genomics-informed drug was 195 days (95% C.I.: 144-245), compared with 156 days for those that did not (95% C.I.: 85-226); p = 0.50], or having favourable ESCAT categories [median PFS for patients with ESCAT I-III was 183 days (95% C.I.: 104-261), compared with 180 (95% C.I.:144-215) for patients with ESCAT IV-X; p = 0.87]. In contrast, NGS testing performed within clinical judgement showed a significantly improved PFS [median PFS for patients that were profiled under the recommended scenarios was 319 days (95% C.I.: 0-658), compared to 123 days (95% C.I.: 89-156) in the non-recommended categories; p = 0.0020]. Interpretation According to our data, real-world outcomes after NGS testing provide evidence of the benefit of clinical judgement in patients with either advanced cancers that routinely need multiple genetic markers, patients with advanced rare cancers, or patients that are screened for molecular clinical trials. By contrast, NGS does not seem to be valuable when performed in cases with a poor PS, rapidly progressing cancer, short expected lifetime, or cases with no standard therapeutic options. Funding RC, NR-L and MQF are recipients of the PMP22/00032 grant, funded by the ISCIII and co-funded by the European Regional Development Fund (ERDF). The study also received funds from the CRIS Contra el Cancer Foundation.
Collapse
Affiliation(s)
- Ramon Colomer
- Department of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Chair of Personalised Precision Medicine, Universidad Autonoma de Madrid (UAM – Fundación Instituto Roche), Madrid, Spain
- Medical Oncology Division, Hospital Universitario La Princesa, Madrid, Spain
- Breast Cancer Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
| | - Jesús Miranda
- University Hospital Quironsalud Madrid, Madrid, Spain
| | | | | | | | - Silvana Mouron
- Breast Cancer Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
| | - Maria J. Bueno
- Breast Cancer Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
| | - Rebeca Mondéjar
- Department of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Chair of Personalised Precision Medicine, Universidad Autonoma de Madrid (UAM – Fundación Instituto Roche), Madrid, Spain
- Medical Oncology Division, Hospital Universitario La Princesa, Madrid, Spain
| | - Miguel Quintela-Fandino
- Department of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Chair of Personalised Precision Medicine, Universidad Autonoma de Madrid (UAM – Fundación Instituto Roche), Madrid, Spain
- Breast Cancer Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
| |
Collapse
|
104
|
Kurimoto M, Rockenbach Y, Kato A, Natsume A. Prediction of Tumor Development and Urine-Based Liquid Biopsy for Molecule-Targeted Therapy of Gliomas. Genes (Basel) 2023; 14:1201. [PMID: 37372381 DOI: 10.3390/genes14061201] [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: 12/30/2022] [Revised: 05/21/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
The timing of the acquisition of tumor-specific gene mutations and the systems by which these gene mutations are acquired during tumorigenesis were clarified. Advances in our understanding of tumorigenesis are being made every day, and therapies targeting fundamental genetic alterations have great potential for cancer treatment. Moreover, our research team successfully estimated tumor progression using mathematical modeling and attempted early diagnosis of brain tumors. We developed a nanodevice that enables urinary genetic diagnosis in a simple and noninvasive manner. Mainly on the basis of our research and experience, this review article presents novel therapies being developed for central nervous system cancers and six molecules, which upon mutation cause tumorigenesis and tumor progression. Further understanding of the genetic characteristics of brain tumors will lead to the development of precise drugs and improve individual treatment outcomes.
Collapse
Affiliation(s)
- Michihiro Kurimoto
- Department of Neurosurgery, Aichi Children's Health and Medical Center, Obu 464-8710, Japan
| | - Yumi Rockenbach
- Institute of Innovation for Future Society, Nagoya University, Nagoya 464-8601, Japan
| | - Akira Kato
- Institute of Innovation for Future Society, Nagoya University, Nagoya 464-8601, Japan
| | - Atsushi Natsume
- Institute of Innovation for Future Society, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
105
|
Salemme V, Centonze G, Avalle L, Natalini D, Piccolantonio A, Arina P, Morellato A, Ala U, Taverna D, Turco E, Defilippi P. The role of tumor microenvironment in drug resistance: emerging technologies to unravel breast cancer heterogeneity. Front Oncol 2023; 13:1170264. [PMID: 37265795 PMCID: PMC10229846 DOI: 10.3389/fonc.2023.1170264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Breast cancer is a highly heterogeneous disease, at both inter- and intra-tumor levels, and this heterogeneity is a crucial determinant of malignant progression and response to treatments. In addition to genetic diversity and plasticity of cancer cells, the tumor microenvironment contributes to tumor heterogeneity shaping the physical and biological surroundings of the tumor. The activity of certain types of immune, endothelial or mesenchymal cells in the microenvironment can change the effectiveness of cancer therapies via a plethora of different mechanisms. Therefore, deciphering the interactions between the distinct cell types, their spatial organization and their specific contribution to tumor growth and drug sensitivity is still a major challenge. Dissecting intra-tumor heterogeneity is currently an urgent need to better define breast cancer biology and to develop therapeutic strategies targeting the microenvironment as helpful tools for combined and personalized treatment. In this review, we analyze the mechanisms by which the tumor microenvironment affects the characteristics of tumor heterogeneity that ultimately result in drug resistance, and we outline state of the art preclinical models and emerging technologies that will be instrumental in unraveling the impact of the tumor microenvironment on resistance to therapies.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Pietro Arina
- UCL, Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| |
Collapse
|
106
|
Adams CM, Mitra R, Xiao Y, Michener P, Palazzo J, Chao A, Gour J, Cassel J, Salvino JM, Eischen CM. Targeted MDM2 Degradation Reveals a New Vulnerability for p53-Inactivated Triple-Negative Breast Cancer. Cancer Discov 2023; 13:1210-1229. [PMID: 36734633 PMCID: PMC10164114 DOI: 10.1158/2159-8290.cd-22-1131] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/29/2022] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
Triple-negative breast cancers (TNBC) frequently inactivate p53, increasing their aggressiveness and therapy resistance. We identified an unexpected protein vulnerability in p53-inactivated TNBC and designed a new PROteolysis TArgeting Chimera (PROTAC) to target it. Our PROTAC selectively targets MDM2 for proteasome-mediated degradation with high-affinity binding and VHL recruitment. MDM2 loss in p53 mutant/deleted TNBC cells in two-dimensional/three-dimensional culture and TNBC patient explants, including relapsed tumors, causes apoptosis while sparing normal cells. Our MDM2-PROTAC is stable in vivo, and treatment of TNBC xenograft-bearing mice demonstrates tumor on-target efficacy with no toxicity to normal cells, significantly extending survival. Transcriptomic analyses revealed upregulation of p53 family target genes. Investigations showed activation and a required role for TAp73 to mediate MDM2-PROTAC-induced apoptosis. Our data, challenging the current MDM2/p53 paradigm, show MDM2 is required for p53-inactivated TNBC cell survival, and PROTAC-targeted MDM2 degradation is an innovative potential therapeutic strategy for TNBC and superior to existing MDM2 inhibitors. SIGNIFICANCE p53-inactivated TNBC is an aggressive, therapy-resistant, and lethal breast cancer subtype. We designed a new compound targeting an unexpected vulnerability we identified in TNBC. Our MDM2-targeted degrader kills p53-inactivated TNBC cells, highlighting the requirement for MDM2 in TNBC cell survival and as a new therapeutic target for this disease. See related commentary by Peuget and Selivanova, p. 1043. This article is highlighted in the In This Issue feature, p. 1027.
Collapse
Affiliation(s)
- Clare M. Adams
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ramkrishna Mitra
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Peter Michener
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Juan Palazzo
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Allen Chao
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | | | - Christine M. Eischen
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
107
|
Qian K, Liu Q. Narrative review on the role of immunotherapy in early triple negative breast cancer: unveiling opportunities and overcoming challenges. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2023; 4:16. [PMID: 38751461 PMCID: PMC11093071 DOI: 10.21037/tbcr-23-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/20/2023] [Indexed: 05/18/2024]
Abstract
Background and Objective Triple negative breast cancer (TNBC) represents a highly aggressive breast cancer subtype, historically managed with chemotherapy regimens predominantly involving anthracyclines and taxanes, yielding unfavorable prognoses. This review endeavors to offer a thorough examination of the present state of treatment strategies for early stage triple negative breast cancer (eTNBC), with a particular emphasis on immunotherapy modalities, combination therapies, predictive biomarkers, and ongoing clinical trials. The principal aim of this review is to meticulously assess the available literature, ascertain significant discoveries, and engage in discussions regarding their potential implications for future research endeavors, clinical applications, and policy formulation. Methods This review was conducted using PubMed and Google Scholar databases, with the latest update performed in March 2023. The search strategy was designed to ensure a comprehensive analysis of the literature, with a focus on recent advancements. Key Content and Findings We critically assess the current eTNBC treatment landscape, covering efficacy and limitations of monotherapy, combination therapies, and predictive biomarkers. We highlight promising results from recent trials, address controversies surrounding chemotherapy, and explore optimal approaches for adjuvant and neoadjuvant therapy (NAT). Insights into personalized treatment strategies, ongoing trials, and future perspectives are provided, advancing our understanding of therapeutic options for eTNBC. Conclusions Through a comprehensive analysis of the literature, this review highlights the potential of immunotherapy, particularly in combination with chemotherapy, as a promising approach for treating eTNBC. However, further research is warranted to optimize treatment strategies, refine patient selection criteria, and identify reliable biomarkers for predicting response to immune checkpoint inhibitors (ICIs). The findings of this review hold significant implications for future research, clinical practice, and policy-making, offering valuable insights into the current challenges and advancements in eTNBC treatment. Ultimately, this knowledge can contribute to improved patient outcomes, enhanced quality of life, and the development of more effective therapeutic approaches for eTNBC.
Collapse
Affiliation(s)
- Keyang Qian
- Department of Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
108
|
Comaills V, Castellano-Pozo M. Chromosomal Instability in Genome Evolution: From Cancer to Macroevolution. BIOLOGY 2023; 12:671. [PMID: 37237485 PMCID: PMC10215859 DOI: 10.3390/biology12050671] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023]
Abstract
The integrity of the genome is crucial for the survival of all living organisms. However, genomes need to adapt to survive certain pressures, and for this purpose use several mechanisms to diversify. Chromosomal instability (CIN) is one of the main mechanisms leading to the creation of genomic heterogeneity by altering the number of chromosomes and changing their structures. In this review, we will discuss the different chromosomal patterns and changes observed in speciation, in evolutional biology as well as during tumor progression. By nature, the human genome shows an induction of diversity during gametogenesis but as well during tumorigenesis that can conclude in drastic changes such as the whole genome doubling to more discrete changes as the complex chromosomal rearrangement chromothripsis. More importantly, changes observed during speciation are strikingly similar to the genomic evolution observed during tumor progression and resistance to therapy. The different origins of CIN will be treated as the importance of double-strand breaks (DSBs) or the consequences of micronuclei. We will also explain the mechanisms behind the controlled DSBs, and recombination of homologous chromosomes observed during meiosis, to explain how errors lead to similar patterns observed during tumorigenesis. Then, we will also list several diseases associated with CIN, resulting in fertility issues, miscarriage, rare genetic diseases, and cancer. Understanding better chromosomal instability as a whole is primordial for the understanding of mechanisms leading to tumor progression.
Collapse
Affiliation(s)
- Valentine Comaills
- Andalusian Center for Molecular Biology and Regenerative Medicine—CABIMER, University of Pablo de Olavide—University of Seville—CSIC, Junta de Andalucía, 41092 Seville, Spain
| | - Maikel Castellano-Pozo
- Andalusian Center for Molecular Biology and Regenerative Medicine—CABIMER, University of Pablo de Olavide—University of Seville—CSIC, Junta de Andalucía, 41092 Seville, Spain
- Genetic Department, Faculty of Biology, University of Seville, 41080 Seville, Spain
| |
Collapse
|
109
|
Takahashi H, Yasui T, Hirano M, Shinjo K, Miyazaki Y, Shinoda W, Hasegawa T, Natsume A, Kitano Y, Ida M, Zhang M, Shimada T, Paisrisarn P, Zhu Z, Ohka F, Aoki K, Rahong S, Nagashima K, Yanagida T, Baba Y. Mutation detection of urinary cell-free DNA via catch-and-release isolation on nanowires for liquid biopsy. Biosens Bioelectron 2023; 234:115318. [PMID: 37172361 DOI: 10.1016/j.bios.2023.115318] [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: 01/17/2023] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/14/2023]
Abstract
Cell-free DNA (cfDNA) and extracellular vesicles (EVs) are molecular biomarkers in liquid biopsies that can be applied for cancer detection, which are known to carry information on the necessary conditions for oncogenesis and cancer cell-specific activities after oncogenesis, respectively. Analyses for both cfDNA and EVs from the same body fluid can provide insights into screening and identifying the molecular subtypes of cancer; however, a major bottleneck is the lack of efficient and standardized techniques for the isolation of cfDNA and EVs from clinical specimens. Here, we achieved catch-and-release isolation by hydrogen bond-mediated binding of cfDNA in urine to zinc oxide (ZnO) nanowires, which also capture EVs by surface charge, and subsequently we identified genetic mutations in urinary cfDNA. The binding strength of hydrogen bonds between single-crystal ZnO nanowires and DNA was found to be equal to or larger than that of conventional hydrophobic interactions, suggesting the possibility of isolating trace amounts of cfDNA. Our results demonstrated that nanowire-based cancer screening assay can screen cancer and can identify the molecular subtypes of cancer in urine from brain tumor patients through EV analysis and cfDNA mutation analysis. We anticipate our method to be a starting point for more sophisticated diagnostic models of cancer screening and identification.
Collapse
Affiliation(s)
- Hiromi Takahashi
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
| | - Takao Yasui
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; Japan Science and Technology Agency (JST), PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan.
| | - Masaki Hirano
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Kanokoden, Chikusa-ku, Nagoya, 464-0021, Japan
| | - Keiko Shinjo
- Division of Cancer Biology, Graduate School of Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Yusuke Miyazaki
- Research Institute for Interdisciplinary Science, Okayama University, Okayama, 700-8530, Japan
| | - Wataru Shinoda
- Research Institute for Interdisciplinary Science, Okayama University, Okayama, 700-8530, Japan
| | - Takeshi Hasegawa
- Institute for Chemical Research, Kyoto University, Uji, Kyoto, 611-0011, Japan
| | - Atsushi Natsume
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Yotaro Kitano
- Department of Neurosurgery, Graduate School of Medicine, Nagoya University, Tsurumai-cho 65, Showa-ku, Nagoya, 466-8550, Japan
| | - Mikiko Ida
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Min Zhang
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Taisuke Shimada
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Piyawan Paisrisarn
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Zetao Zhu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Fumiharu Ohka
- Department of Neurosurgery, Graduate School of Medicine, Nagoya University, Tsurumai-cho 65, Showa-ku, Nagoya, 466-8550, Japan
| | - Kosuke Aoki
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Sakon Rahong
- College of Materials Innovation and Technology, King Mongkut's Institute of Technology Ladkrabang, Chalongkrung Rd., Ladkrabang, Bangkok, 10520, Thailand
| | - Kazuki Nagashima
- Japan Science and Technology Agency (JST), PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan; Department of Applied Chemistry, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Takeshi Yanagida
- Department of Applied Chemistry, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka-cho, Ibaraki, Osaka, 567-0047, Japan; Institute for Materials Chemistry and Engineering, Kyushu University, 6-1 Kasuga-Koen, Kasuga, Fukuoka, 816-8580, Japan
| | - Yoshinobu Baba
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; Institute for Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba, 263-8555, Japan.
| |
Collapse
|
110
|
Zhong Y, Li F, Zhang S, Yang Z, Ren X, Cao X, Xu Y, Guo D, Zhou Y, Mao F, Shen S, Sun Q. Syndecan-1 as an immunogene in Triple-negative breast cancer: regulation tumor-infiltrating lymphocyte in the tumor microenviroment and EMT by TGFb1/Smad pathway. Cancer Cell Int 2023; 23:76. [PMID: 37069585 PMCID: PMC10111802 DOI: 10.1186/s12935-023-02917-7] [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: 01/17/2023] [Accepted: 04/03/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors are the most studied forms of immunotherapy for triple-negative breast cancer (TNBC). The Cancer Genome Map (TCGA) and METABRIC project provide large-scale cancer samples that can be used for comprehensive and reliable immunity-related gene research. METHODS We analyzed data from TCGA and METABRIC and established an immunity-related gene prognosis model for breast cancer. The SDC1 expression in tumor and cancer associated fibroblasts (CAFs) was then observed in 282 TNBC patients by immunohistochemistry. The effects of SDC1 on MDA-MB-231 proliferation, migration and invasion were evaluated. Qualitative real-time PCR and western blotting were performed to identify mRNA and protein expression, respectively. RESULTS SDC1, as a key immunity-related gene, was significantly correlated with survival in the TCGA and METABRIC databases, while SDC1 was found to be highly expressed in TNBC in the METABRIC database. In the TNBC cohort, patients with high SDC1 expression in tumor cells and low expression in CAFs had significantly lower disease-free survival (DFS) and fewer tumor-infiltrating lymphocytes (TILs). The downregulation of SDC1 decreased the proliferation of MDA-MB-231, while promoting the migration of MDA-MB-231 cells by reducing the gene expression of E-cadherin and TGFb1 and activating p-Smad2 and p-Smad3 expression. CONCLUSION SDC1 is a key immunity-related gene that is highly expressed TNBC patients. Patients with high SDC1 expression in tumors and low expression in CAFs had poor prognoses and low TILs. Our findings also suggest that SDC1 regulates the migration of MDA-MB-231 breast cancer cells through a TGFb1-Smad and E-cadherin-dependent mechanism.
Collapse
Affiliation(s)
- Ying Zhong
- Department of Breast Disease, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Fangyuan Li
- Medical Research Central, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
- Clinical Biobank, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Sumei Zhang
- Medical Research Central, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
- Clinical Biobank, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Zhenli Yang
- Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & School of Basic Medicine, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng, Beijing, 100730, China
| | - Xinyu Ren
- Department of Pathology, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Xi Cao
- Department of Breast Disease, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Yali Xu
- Department of Breast Disease, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Dan Guo
- Medical Research Central, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
- Clinical Biobank, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Yidong Zhou
- Department of Breast Disease, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Feng Mao
- Department of Breast Disease, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Songjie Shen
- Department of Breast Disease, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Qiang Sun
- Department of Breast Disease, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| |
Collapse
|
111
|
Abuhadra N, Sun R, Bassett RL, Huo L, Chang JT, Teshome M, Clayborn AR, White JB, Ravenberg EE, Adrada BE, Candelaria RP, Yang W, Ding Q, Symmans WF, Arun B, Damodaran S, Koenig KB, Layman RM, Lim B, Litton JK, Thompson A, Ueno NT, Piwnica-Worms H, Hortobagyi GN, Valero V, Tripathy D, Rauch GM, Moulder S, Yam C. Targeting chemotherapy resistance in mesenchymal triple-negative breast cancer: a phase II trial of neoadjuvant angiogenic and mTOR inhibition with chemotherapy. Invest New Drugs 2023:10.1007/s10637-023-01357-4. [PMID: 37043123 DOI: 10.1007/s10637-023-01357-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/27/2023] [Indexed: 04/13/2023]
Affiliation(s)
- Nour Abuhadra
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Ryan Sun
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roland L Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mediget Teshome
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alyson R Clayborn
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jason B White
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Elizabeth E Ravenberg
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Beatriz E Adrada
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rosalind P Candelaria
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Yang
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qingqing Ding
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W Fraser Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Senthil Damodaran
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Kimberly B Koenig
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Rachel M Layman
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Alastair Thompson
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel N Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Gaiane M Rauch
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stacy Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Clinton Yam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
112
|
|
113
|
Yuan L, Liu J, Bao L, Qu H, Xiang J, Sun P. Upregulation of the ferroptosis-related STEAP3 gene is a specific predictor of poor triple-negative breast cancer patient outcomes. Front Oncol 2023; 13:1032364. [PMID: 37064114 PMCID: PMC10102497 DOI: 10.3389/fonc.2023.1032364] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
ObjectiveThis study was designed to assess ferroptosis regulator gene (FRG) expression patterns in patients with TNBC based on data derived from The Cancer Genome Atlas (TCGA). Further, it was utilized to establish a TNBC FRG signature, after which the association between this signature and the tumor immune microenvironment (TIME) composition was assessed, and relevant prognostic factors were explored.MethodsThe TCGA database was used to obtain RNA expression datasets and clinical information about 190 TNBC patients, after which a prognostic TNBC-related FRG signature was established using a least absolute shrinkage and selection operator (LASSO) Cox regression approach. These results were validated with separate data from the Gene Expression Omnibus (GEO). The TNBC-specific prognostic gene was identified via this method. The STEAP3 was then validated through Western immunoblotting, immunohistochemical staining, and quantitative real‐time polymerase chain reaction (RT-qPCR) analyses of clinical tissue samples and TNBC cell lines. Chemotherapy interactions and predicted drug sensitivity studies were investigated to learn more about the potential clinical relevance of these observations.ResultsThese data revealed that 87 FRGs were differentially expressed when comparing TNBC tumors and healthy tissue samples (87/259, 33.59%). Seven of these genes (CA9, CISD1, STEAP3, HMOX1, DUSP1, TAZ, HBA1) are significantly related to the overall survival of TNBC patients. Kaplan-Meier analyses and established FRG signatures and nomograms identified CISD1 and STEAP3 genes of prognostic relevance. Prognostic Risk Score values were positively correlated with the infiltration of CD4+ T cells (p = 0.001) and myeloid dendritic cells (p =0.004). Further evidence showed that STEAP3 was strongly and specifically associated with TNBC patient OS (P<0.05). The results above were confirmed by additional examinations of STEAP3 expression changes in TNBC patient samples and cell lines. High STEAP3 levels were negatively correlated with half-maximal inhibitory concentration (IC50) values for GSK1904529A (IGF1R inhibitor), AS601245 (JNK inhibitor), XMD8−85 (Erk5 inhibitor), Gefitinib, Sorafenib, and 5-Fluorouracil (P < 0.05) in patients with TNBC based on information derived from the TCGA-TNBC dataset.ConclusionIn the present study, a novel FRG model was developed and used to forecast the prognosis of TNBC patients accurately. Furthermore, it was discovered that STEAP3 was highly overexpressed in people with TNBC and associated with overall survival rates, laying the groundwork for the eventually targeted therapy of individuals with this form of cancer.
Collapse
Affiliation(s)
- Lifang Yuan
- Department of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, China
- Department of Breast Oncology, Huanxing Cancer Hospital, Beijing, China
| | - Jiannan Liu
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Lei Bao
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Huajun Qu
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jinyu Xiang
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Ping Sun
- Department of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, China
- *Correspondence: Ping Sun,
| |
Collapse
|
114
|
Kan Y, Paung Y, Kim Y, Seeliger MA, Miller WT. Biochemical Studies of Systemic Lupus Erythematosus-Associated Mutations in Nonreceptor Tyrosine Kinases Ack1 and Brk. Biochemistry 2023; 62:1124-1137. [PMID: 36854171 PMCID: PMC10052838 DOI: 10.1021/acs.biochem.2c00685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Tyrosine kinases (TKs) play essential roles in signaling processes that regulate cell survival, migration, and proliferation. Dysregulation of tyrosine kinases underlies many disorders, including cancer, cardiovascular and developmental diseases, as well as pathologies of the immune system. Ack1 and Brk are nonreceptor tyrosine kinases (NRTKs) best known for their roles in cancer. Here, we have biochemically characterized novel Ack1 and Brk mutations identified in patients with systemic lupus erythematosus (SLE). These mutations are the first SLE-linked polymorphisms found among NRTKs. We show that two of the mutants are catalytically inactive, while the other three have reduced activity. To understand the structural changes associated with the loss-of-function phenotype, we solved the crystal structure of one of the Ack1 kinase mutants, K161Q. Furthermore, two of the mutated residues (Ack1 A156 and K161) critical for catalytic activity are highly conserved among other TKs, and their substitution in other members of the kinase family could have implications in cancer. In contrast to canonical gain-of-function mutations in TKs observed in many cancers, we report loss-of-function mutations in Ack1 and Brk, highlighting the complexity of TK involvement in human diseases.
Collapse
Affiliation(s)
- Yagmur Kan
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York 11794-8661, United States
| | - YiTing Paung
- Department of Pharmacology, School of Medicine, Stony Brook University, Stony Brook, New York 11794-8661, United States
| | - Yunyoung Kim
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York 11794-8661, United States
| | - Markus A Seeliger
- Department of Pharmacology, School of Medicine, Stony Brook University, Stony Brook, New York 11794-8661, United States
| | - W Todd Miller
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York 11794-8661, United States
- Department of Veterans Affairs Medical Center, Northport, New York 11768, United States
| |
Collapse
|
115
|
Nolan E, Lindeman GJ, Visvader JE. Deciphering breast cancer: from biology to the clinic. Cell 2023; 186:1708-1728. [PMID: 36931265 DOI: 10.1016/j.cell.2023.01.040] [Citation(s) in RCA: 174] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/14/2023] [Accepted: 01/30/2023] [Indexed: 03/17/2023]
Abstract
Breast cancer remains a leading cause of cancer-related mortality in women, reflecting profound disease heterogeneity, metastasis, and therapeutic resistance. Over the last decade, genomic and transcriptomic data have been integrated on an unprecedented scale and revealed distinct cancer subtypes, critical molecular drivers, clonal evolutionary trajectories, and prognostic signatures. Furthermore, multi-dimensional integration of high-resolution single-cell and spatial technologies has highlighted the importance of the entire breast cancer ecosystem and the presence of distinct cellular "neighborhoods." Clinically, a plethora of new targeted therapies has emerged, now being rapidly incorporated into routine care. Resistance to therapy, however, remains a crucial challenge for the field.
Collapse
Affiliation(s)
- Emma Nolan
- Auckland Cancer Society Research Centre, University of Auckland, Auckland 1023, New Zealand
| | - Geoffrey J Lindeman
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC 3050, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Jane E Visvader
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
116
|
Zhao X, Fu L, Zhai L, Yang X, Gao R. MiR-526b targets lncRNA SLC16A1-AS1 to suppress cell proliferation in triple-negative breast cancer. J Biochem Mol Toxicol 2023; 37:e23247. [PMID: 36654514 DOI: 10.1002/jbt.23247] [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: 09/22/2021] [Revised: 07/31/2022] [Accepted: 10/12/2022] [Indexed: 01/20/2023]
Abstract
The present study investigated the potential interaction between miR-526b and lncRNA SLC16A1-AS1 in triple-negative breast cancer (TNBC). Expression of miR-526b and SLC16A1-AS1 in TNBC tumor tissues and paired nontumor tissues from 60 TNBC patients was detected by real-time polymerase chain reaction (RT-qPCR). The interaction between miR-526b and SLC16A1-AS1 was evaluated with overexpression experiments, followed by RT-qPCR. The proliferation and migration of cells were detected with cell counting kit-8 assay and Transwell assay, respectively. Apoptosis of cells was assessed by cell apoptosis assay. The expression of apoptosis-related proteins was quantified by Western blot analysis. MiR-526b was predicted to bind with SLC16A1-AS1. Overexpression of miR-526b in TNBC cells decreased the expression levels of SLC16A1-AS1, while overexpression of SLC16A1-AS1 did not affect the expression of miR-526b. In TNBC tissues, miR-526b was downregulated in TNBC tissues, while SLC16A1-AS1 was upregulated in TNBC tissues compared to that in nontumor tissues. The expression of SLC16A1-AS1 and miR-526b were inversely correlated. In vitro experiments showed that overexpression of SLC16A1-AS1 promoted cell proliferation and invasion but suppressed cell apoptosis. MiR-526b played an opposite role and suppressed the function of SLC16A1-AS1. MiR-526b is downregulated in TNBC and it targets SLC16A1-AS1 to regulate proliferation, apoptosis, and invasion of TNBC cells.
Collapse
Affiliation(s)
- Xingjuan Zhao
- Department of Mammary Gland, Shanxi People's Hospital, Taiyuan, Shanxi, China
| | - Lei Fu
- Department of Surgery, Shanxi Provincial General Team Hospital of the Chinese People's Armed Police Force, Taiyuan, Shanxi, China
| | - Liqin Zhai
- Department of Pathology, Shanxi People's Hospital, Shanxi, Taiyuan, China
| | - Xuan Yang
- Department of Mammary Gland, Shanxi People's Hospital, Taiyuan, Shanxi, China
| | - Runfang Gao
- Department of Mammary Gland, Shanxi People's Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
117
|
Identification of Kinase Targets for Enhancing the Antitumor Activity of Eribulin in Triple-Negative Breast Cell Lines. Biomedicines 2023; 11:biomedicines11030735. [PMID: 36979714 PMCID: PMC10045293 DOI: 10.3390/biomedicines11030735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Background: Triple-negative breast cancer (TNBC) is the most aggressive molecular subtype of breast cancer, and current treatments are only partially effective in disease control. More effective combination approaches are needed to improve the survival of TNBC patients. Eribulin mesylate, a non-taxane microtubule dynamics inhibitor, is approved by the U.S. Food and Drug Administration to treat metastatic breast cancer after at least two previous chemotherapeutic regimens. However, eribulin as a single agent has limited therapeutic efficacy against TNBC. Methods: High-throughput kinome library RNAi screening, Ingenuity Pathway Analysis, and STRING analysis were performed to identify target kinases for combination with eribulin. The identified combinations were validated using in vivo and ex vivo proliferation assays. Results: We identified 135 potential kinase targets whose inhibition enhanced the antiproliferation effect of eribulin in TNBC cells, with the PI3K/Akt/mTOR and the MAPK/JNK pathways emerging as the top candidates. Indeed, copanlisib (pan-class I PI3K inhibitor), everolimus (mTOR inhibitor), trametinib (MEK inhibitor), and JNK-IN-8 (pan-JNK inhibitor) produced strong synergistic antiproliferative effects when combined with eribulin, and the PI3K and mTOR inhibitors had the most potent effects in vitro. Conclusions: Our data suggest a new strategy of combining eribulin with PI3K or mTOR inhibitors to treat TNBC.
Collapse
|
118
|
Alves CL, Ditzel HJ. Drugging the PI3K/AKT/mTOR Pathway in ER+ Breast Cancer. Int J Mol Sci 2023; 24:4522. [PMID: 36901954 PMCID: PMC10003259 DOI: 10.3390/ijms24054522] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
The frequent activation of the PI3K/AKT/mTOR pathway and its crucial role in estrogen receptor-positive (ER+) breast cancer tumorigenesis and drug resistance has made it a highly attractive therapeutic target in this breast cancer subtype. Consequently, the number of new inhibitors in clinical development targeting this pathway has drastically increased. Among these, the PIK3CA isoform-specific inhibitor alpelisib and the pan-AKT inhibitor capivasertib were recently approved in combination with the estrogen receptor degrader fulvestrant for the treatment of ER+ advanced breast cancer after progression on an aromatase inhibitor. Nevertheless, the clinical development of multiple inhibitors of the PI3K/AKT/mTOR pathway, in parallel with the incorporation of CDK4/6 inhibitors into the standard of care treatment in ER+ advanced breast cancer, has led to a multitude of available therapeutic agents and many possible combined strategies which complicate personalizing treatment. Here, we review the role of the PI3K/AKT/mTOR pathway in ER+ advanced breast cancer, highlighting the genomic contexts in which the various inhibitors of this pathway may have superior activity. We also discuss selected trials with agents targeting the PI3K/AKT/mTOR and related pathways as well as the rationale supporting the clinical development of triple combination therapy targeting ER, CDK4/6 and PI3K/AKT/mTOR in ER+ advanced breast cancer.
Collapse
Affiliation(s)
- Carla L. Alves
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Henrik J. Ditzel
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- Department of Oncology, Institute of Clinical Research, Odense University Hospital, 5000 Odense, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, 5000 Odense, Denmark
| |
Collapse
|
119
|
Scarpetti L, Oturkar CC, Juric D, Shellock M, Malvarosa G, Post K, Isakoff S, Wang N, Nahed B, Oh K, Das GM, Bardia A. Therapeutic Role of Tamoxifen for Triple-Negative Breast Cancer: Leveraging the Interaction Between ERβ and Mutant p53. Oncologist 2023; 28:358-363. [PMID: 36772966 PMCID: PMC10078911 DOI: 10.1093/oncolo/oyac281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/30/2022] [Indexed: 02/12/2023] Open
Abstract
The absence of effective therapeutic targets and aggressive nature of triple-negative breast cancer (TNBC) renders this disease subset difficult to treat. Although estrogen receptor beta (ERβ) is expressed in TNBC, studies on its functional role have yielded inconsistent results. However, recently, our preclinical studies, along with other observations, have shown the potential therapeutic utility of ERβ in the context of mutant p53 expression. The current case study examines the efficacy of the selective estrogen receptor modulator tamoxifen in p53-mutant TNBC with brain metastases. Significant increase in ERβ protein expression and anti-proliferative interaction between mutant p53 and ERβ were observed after cessation of tamoxifen therapy, with significant regression of brain metastases. This case study provides supporting evidence for the use of tamoxifen in p53-mutant, ERβ+TNBC, especially in the setting of brain metastasis.
Collapse
Affiliation(s)
- Lauren Scarpetti
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | - Dejan Juric
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Maria Shellock
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Giuliana Malvarosa
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Kathryn Post
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Steven Isakoff
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Nancy Wang
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Brian Nahed
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Kevin Oh
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Gokul M Das
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
120
|
Wang K, Zheng C, Xue L, Deng D, Zeng L, Li M, Deng X. A bibliometric analysis of 16,826 triple-negative breast cancer publications using multiple machine learning algorithms: Progress in the past 17 years. Front Med (Lausanne) 2023; 10:999312. [PMID: 36844225 PMCID: PMC9945529 DOI: 10.3389/fmed.2023.999312] [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: 07/20/2022] [Accepted: 01/16/2023] [Indexed: 02/11/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC) is proposed at the beginning of this century, which is still the most challenging breast cancer subtype due to its aggressive behavior, including early relapse, metastatic spread, and poor survival. This study uses machine learning methods to explore the current research status and deficiencies from a macro perspective on TNBC publications. Methods PubMed publications under "triple-negative breast cancer" were searched and downloaded between January 2005 and 2022. R and Python extracted MeSH terms, geographic information, and other abstracts from metadata. The Latent Dirichlet Allocation (LDA) algorithm was applied to identify specific research topics. The Louvain algorithm established a topic network, identifying the topic's relationship. Results A total of 16,826 publications were identified, with an average annual growth rate of 74.7%. Ninety-eight countries and regions in the world participated in TNBC research. Molecular pathogenesis and medication are most studied in TNBC research. The publications mainly focused on three aspects: Therapeutic target research, Prognostic research, and Mechanism research. The algorithm and citation suggested that TNBC research is based on technology that advances TNBC subtyping, new drug development, and clinical trials. Conclusion This study quantitatively analyzes the current status of TNBC research from a macro perspective and will aid in redirecting basic and clinical research toward a better outcome for TNBC. Therapeutic target research and Nanoparticle research are the present research focus. There may be a lack of research on TNBC from a patient perspective, health economics, and end-of-life care perspectives. The research direction of TNBC may require the intervention of new technologies.
Collapse
Affiliation(s)
- Kangtao Wang
- Department of General Surgery, The Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chanjuan Zheng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan, Department of Pathophysiology, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Lian Xue
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan, Department of Pathophysiology, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Dexin Deng
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Liang Zeng
- Department of Pathology, Guangzhou Women and Children’s Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China,*Correspondence: Liang Zeng,
| | - Ming Li
- Department of Immunology, College of Basic Medical Sciences, Central South University, Changsha, Hunan, China,Ming Li,
| | - Xiyun Deng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan, Department of Pathophysiology, School of Medicine, Hunan Normal University, Changsha, Hunan, China,Xiyun Deng,
| |
Collapse
|
121
|
Banerjee M, Devi Rajeswari V. Inhibition of WNT signaling by conjugated microRNA nano-carriers: A new therapeutic approach for treating triple-negative breast cancer a perspective review. Crit Rev Oncol Hematol 2023; 182:103901. [PMID: 36584723 DOI: 10.1016/j.critrevonc.2022.103901] [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: 12/19/2021] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Triple-Negative Breast Cancer is the most aggressive form and accounts the 15%-25% of all breast cancer. Receptors are absent in triple-negative breast cancer, which makes them unresponsive to the current hormonal therapies. The patients with TNBC are left with the option of cytotoxic chemotherapy. The Wnt pathways are connected to cancer, and when activated, they result in mammary hyperplasia and tumors. The tumor suppressor microRNAs can block tumor cell proliferation, invasion, and migration, lead to cancer cell death, and are also known to down-regulate the WNT signaling. Nanoparticles with microRNA have been seen to be more effective when compared with their single release. In this review, we have tried to understand how Wnt signaling plays a crucial role in TNBC, EMT, metastasis, anti-drug resistance, and regulation of Wnt by microRNA. The role of nano-carriers in delivering micro-RNA. The clinical biomarkers, including the present state-of-the-art, involve novel pathways of Wnt.
Collapse
Affiliation(s)
- Manosi Banerjee
- Department of Biomedical Sciences, School of Bioscience and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - V Devi Rajeswari
- Department of Biomedical Sciences, School of Bioscience and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
122
|
Aggarwal M. 2,2-Diphenethyl Isothiocyanate Enhances Topoisomerase Inhibitor-Induced Cell Death and Suppresses Multi-Drug Resistance 1 in Breast Cancer Cells. Cancers (Basel) 2023; 15:cancers15030928. [PMID: 36765888 PMCID: PMC9913484 DOI: 10.3390/cancers15030928] [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: 01/12/2023] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
We previously reported that phenethyl isothiocyanate (PEITC), a dietary-related compound, can rescue mutant p53. A structure-activity relationships study showed that the synthetic analog 2,2-diphenylethyl isothiocyanate (DPEITC) is a more potent inducer of apoptosis than natural or synthetic ITCs. Here, we showed that DPEITC inhibited the growth of triple-negative breast cancer cells (MDA-MB-231, MDA-MB-468, and Hs578T) expressing "hotspot" p53 mutants, structural (p53R280K, p53R273H) or contact (p53V157F), at IC50 values significantly lower than PEITC. DPEITC inhibited the growth of HER2+ (p53R175H SK-BR-3, p53R175H AU565) and Luminal A (p53L194F T47D) breast cancer (BC) cells harboring a p53 structural mutant. DPEITC induced apoptosis, irrespective of BC subtypes, by rescuing p53 mutants. Accordingly, the rescued p53 mutants induced apoptosis by activating canonical WT p53 targets and delaying the cell cycle. DPEITC acted synergistically with doxorubicin and camptothecin to inhibit proliferation and induce apoptosis. Under these conditions, DPEITC delayed BC cells in the G1 phase, activated p53 canonical targets, and enhanced pS1981-ATM. DPEITC reduced the expression of MDR1 and ETS1. These findings are the first report of synergism between a synthetic ITC and a chemotherapy drug via mutant p53 rescue. Furthermore, our data demonstrate that ITCs suppress the expression of cellular proteins that play a role in chemoresistance.
Collapse
Affiliation(s)
- Monika Aggarwal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| |
Collapse
|
123
|
The role of MEOX1 in non-neoplastic and neoplastic diseases. Biomed Pharmacother 2023; 158:114068. [PMID: 36495659 DOI: 10.1016/j.biopha.2022.114068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Targeted gene therapy has shown durable efficacy in non-neoplastic and neoplastic patients. Therefore, finding a suitable target has become a key area of research. Mesenchyme homeobox 1 (MEOX1) is a transcriptional factor that plays a significant role in regulation of somite development. Evidence indicates that abnormalities in MEOX1 expression and function are associated with a variety of pathologies, including non-neoplastic and neoplastic diseases. MEOX1 expression is upregulated during progression of most diseases and plays a critical role in maintenance of the cellular phenotypes such as cell differentiation, cell cycle arrest and senescence, migration, and proliferation. Therefore, MEOX1 may become an important molecular target and therapeutic target. This review will discuss the current state of knowledge on the role of MEOX1 in different diseases.
Collapse
|
124
|
Tanshinone IIA promotes apoptosis by downregulating BCL2 and upregulating TP53 in triple-negative breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:365-374. [PMID: 36374307 DOI: 10.1007/s00210-022-02316-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022]
Abstract
Tanshinone IIA (Tan IIA) was mainly used for cardiovascular disease treatment. Recent studies have demonstrated the role of Tan IIA for tumor treatment, but its mechanism remains unclear. At the first, the inhibitory effect of Tan IIA on 4T1 breast cancer cells was determined by CCK8 and colony formation assay. Then, a 4T1 BALB/c model of breast cancer was established to evaluate the anti-cancer effect of Tan IIA in vivo. Flow cytometry analysis and the TUNEL test were used to detect cell apoptosis in vitro and in vivo, respectively. The related targets and mechanisms of Tan IIA were predicted through network-based systems biology. At last, molecular docking and the molecular biological techniques were used to evaluate the predicted targets. Tan IIA displayed encouraging inhibitory influences on 4T1 cells after incubation for 24 h and showed a half-maximal inhibitory concentration (IC50) of 49.78 μM after 48-h incubation. After 23 days of treatment, the relative tumor volumes in the Tan IIA group were 65.53% inhibited compared with the control group. Furthermore, Tan IIA induced 4T1 cell apoptosis both in vivo and in vitro. The possible targets of Tan IIA for TNBC treatment were predicted with network-based systems biology, and results showed that TP53, NF-κB, AKT, MYC, and BCL-2 were the hub targets. The mechanism against breast cancer may be based on the P53 signaling pathway, the PI3K/Akt pathway, the MAPK signaling pathway, and the mTOR signaling pathways. Molecular docking analysis reveals that Tan IIA has a high affinity for p53, Bcl-2, and NF-κB1; the binding energies were - 6.92, - 6.07, and - 6.28 kcal/mol, respectively. The predicted proteins were further validated using Western blotting. Increased expression of phosphorylated p53 and p53 and decreased expression of Bcl-2 were found in Tan IIA-treated 4T1 cells. Tan IIA is potentially effective for the treatment of 4T1 breast cancer, and the molecular mechanism may be through enhancing the activity of p53 and decreasing Bcl-2 to suppress proliferation and promote apoptosis.
Collapse
|
125
|
Predictive Biomarkers for Response to Immunotherapy in Triple Negative Breast Cancer: Promises and Challenges. J Clin Med 2023; 12:jcm12030953. [PMID: 36769602 PMCID: PMC9917763 DOI: 10.3390/jcm12030953] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a highly heterogeneous disease with a poor prognosis and a paucity of therapeutic options. In recent years, immunotherapy has emerged as a new treatment option for patients with TNBC. However, this therapeutic evolution is paralleled by a growing need for biomarkers which allow for a better selection of patients who are most likely to benefit from this immune checkpoint inhibitor (ICI)-based regimen. These biomarkers will not only facilitate a better optimization of treatment strategies, but they will also avoid unnecessary side effects in non-responders, and limit the increasing financial toxicity linked to the use of these agents. Huge efforts have been deployed to identify predictive biomarkers for the ICI, but until now, the fruits of this labor remained largely unsatisfactory. Among clinically validated biomarkers, only programmed death-ligand 1 protein (PD-L1) expression has been prospectively assessed in TNBC trials. In addition to this, microsatellite instability and a high tumor mutational burden are approved as tumor agnostic biomarkers, but only a small percentage of TNBC fits this category. Furthermore, TNBC should no longer be approached as a single biological entity, but rather as a complex disease with different molecular, clinicopathological, and tumor microenvironment subgroups. This review provides an overview of the validated and evolving predictive biomarkers for a response to ICI in TNBC.
Collapse
|
126
|
Abstract
The HER2+ subtype of human breast cancer is associated with the malignant transformation of luminal ductal cells of the mammary epithelium. The sequence analysis of tumor DNA identifies loss of function mutations and deletions of the MAP2K4 and MAP2K7 genes that encode direct activators of the JUN NH2-terminal kinase (JNK). We report that in vitro studies of human mammary epithelial cells with CRISPR-induced mutations in the MAPK and MAP2K components of the JNK pathway caused no change in growth in 2D culture, but these mutations promoted epithelial cell proliferation in 3D culture. Analysis of gene expression signatures in 3D culture demonstrated similar changes caused by HER2 activation and JNK pathway loss. The mechanism of signal transduction cross-talk may be mediated, in part, by JNK-suppressed expression of integrin α6β4 that binds HER2 and amplifies HER2 signaling. These data suggest that HER2 activation and JNK pathway loss may synergize to promote breast cancer. To test this hypothesis, we performed in vivo studies using a mouse model of HER2+ breast cancer with Cre/loxP-mediated ablation of genes encoding JNK (Mapk8 and Mapk9) and the MAP2K (Map2k4 and Map2k7) that activate JNK in mammary epithelial cells. Kaplan-Meier analysis of tumor development demonstrated that JNK pathway deficiency promotes HER2+-driven breast cancer. Collectively, these data identify JNK pathway genes as potential suppressors for HER2+ breast cancer.
Collapse
|
127
|
Huang T, Li J, Zhao H, Ngamphiw C, Tongsima S, Kantaputra P, Kittitharaphan W, Wang SM. Core promoter in TNBC is highly mutated with rich ethnic signature. Brief Funct Genomics 2023; 22:9-19. [PMID: 36307127 PMCID: PMC9853936 DOI: 10.1093/bfgp/elac035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 01/25/2023] Open
Abstract
The core promoter plays an essential role in regulating transcription initiation by controlling the interaction between transcriptional factors and sequence motifs in the core promoter. Although mutation in core promoter sequences is expected to cause abnormal gene expression leading to pathogenic consequences, limited supporting evidence showed the involvement of core promoter mutation in diseases. Our previous study showed that the core promoter is highly polymorphic in worldwide human ethnic populations in reflecting human history and adaptation. Our recent characterization of the core promoter in triple-negative breast cancer (TNBC), a subtype of breast cancer, in a Chinese TNBC cohort revealed the wide presence of core promoter mutation in TNBC. In the current study, we analyzed the core promoter in a Thai TNBC cohort. We also observed rich core promoter mutation in the Thai TNBC patients. We compared the core promoter mutations between Chinese and Thai TNBC cohorts. We observed substantial differences of core promoter mutation in TNBC between the two cohorts, as reflected by the mutation spectrum, mutation-effected gene and functional category, and altered gene expression. Our study confirmed that the core promoter in TNBC is highly mutable, and is highly ethnic-specific.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - San Ming Wang
- Corresponding author: S.M. Wang, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China. Tel.: +(853) 8822-4836; E-mail:
| |
Collapse
|
128
|
Roesler AS, Malasi S, Koslosky L, Hartmayer P, Naab TJ, Carter JM, Zahrieh D, Hillman D, Leon-Ferre RA, Couch FJ, Goetz MP, Anderson KS, Pockaj BA, Barrett MT. PDJ amplicon in triple negative breast cancer. Sci Rep 2023; 13:618. [PMID: 36635351 PMCID: PMC9837184 DOI: 10.1038/s41598-023-27887-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
Amplification of chromosome 9p24.1 targeting PD-L1, PD-L2, and JAK2 (PDJ amplicon) is present in subsets of triple negative breast cancers (TNBCs) and is associated with poor clinical outcomes. However, the prevalence of PDJ+ TNBCs varies extensively across studies applying different methods for interrogating samples of interest. To rigorously assess the prevalence of PDJ amplicons in TNBC, its prognostic value and whether it is enriched by chemotherapy, we interrogated 360 TNBC samples including 74 surgical resections from patients treated in the neoadjuvant setting, and tissue microarrays (TMAs) with 31 cases from African American women and 255 resected non-metastatic cases, with a 3 color fluorescence in situ hybridization (FISH) assay targeting the 9p24.1 PDJ amplicon, 9q24.3, and 9q34.1. Samples with mean PDJ signal of > 4.5 copies, and ratios of PDJ/9q24 ≥ 2 and/or PDJ/9q34.1 ≥ 2 were called amplified (PDJ+). Correlative analyses included the association of tumor infiltrating lymphocytes (TILs) with PDJ amplicons in TNBCs. In addition, we investigated intratumor copy number of PDJ amplicons in PDJ+ and PDJ- TNBCs. Matched pre- and post-neoadjuvant treatment biopsies were available from patients (n = 6) to evaluate the effects of therapy on PDJ status. Our study provides a rigorous analysis of the prevalence, distribution, and clinical correlatives of the PDJ amplicon in TNBC.
Collapse
Affiliation(s)
- Alexander S Roesler
- Department of Research, Mayo Clinic in Arizona, Scottsdale, AZ, USA
- School of Medicine, Duke University, Durham, NC, USA
| | - Smriti Malasi
- Department of Research, Mayo Clinic in Arizona, Scottsdale, AZ, USA
| | | | | | - Tammey J Naab
- Department of Pathology, Howard University Hospital, Washington, DC, USA
| | - Jodi M Carter
- Departments of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Departments of Surgery, Mayo Clinic, Rochester, MN, USA
| | - David Zahrieh
- Departments of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - David Hillman
- Departments of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Fergus J Couch
- Departments of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Karen S Anderson
- Division of Hematology-Oncology, Mayo Clinic in Arizona, Scottsdale, AZ, USA
- Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Barbara A Pockaj
- Division of General Surgery, Section of Surgical Oncology, Mayo Clinic in Arizona, Phoenix, AZ, USA
| | - Michael T Barrett
- Department of Research, Mayo Clinic in Arizona, Scottsdale, AZ, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic in Arizona, Scottsdale, AZ, USA.
| |
Collapse
|
129
|
Leighton J, Hu M, Sei E, Meric-Bernstam F, Navin NE. Reconstructing mutational lineages in breast cancer by multi-patient-targeted single-cell DNA sequencing. CELL GENOMICS 2023; 3:100215. [PMID: 36777188 PMCID: PMC9903705 DOI: 10.1016/j.xgen.2022.100215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/21/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
Single-cell DNA sequencing (scDNA-seq) methods are powerful tools for profiling mutations in cancer cells; however, most genomic regions sequenced in single cells are non-informative. To overcome this issue, we developed a multi-patient-targeted (MPT) scDNA-seq method. MPT involves first performing bulk exome sequencing across a cohort of cancer patients to identify somatic mutations, which are then pooled together to develop a single custom targeted panel for high-throughput scDNA-seq using a microfluidics platform. We applied MPT to profile 330 mutations across 23,500 cells from 5 patients with triple negative-breast cancer (TNBC), which showed that 3 tumors were monoclonal and 2 tumors were polyclonal. From these data, we reconstructed mutational lineages and identified early mutational and copy-number events, including early TP53 mutations that occurred in all five patients. Collectively, our data suggest that MPT can overcome a major technical obstacle for studying tumor evolution using scDNA-seq by profiling information-rich mutation sites.
Collapse
Affiliation(s)
- Jake Leighton
- Department of Genetics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biological Sciences, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Min Hu
- Department of Genetics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Emi Sei
- Department of Genetics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Funda Meric-Bernstam
- Graduate School of Biological Sciences, UT MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Precision Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicholas E. Navin
- Department of Genetics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biological Sciences, UT MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
130
|
Cancer Vaccines for Triple-Negative Breast Cancer: A Systematic Review. Vaccines (Basel) 2023; 11:vaccines11010146. [PMID: 36679991 PMCID: PMC9866612 DOI: 10.3390/vaccines11010146] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the subtype of breast cancer with the poorest outcomes, and is associated with a high risk of relapse and metastasis. The treatment choices for this malignancy have been confined to conventional chemotherapeutic agents, due to a lack of expression of the canonical molecular targets. Immunotherapy has been recently changing the treatment paradigm for many types of tumors, and the approach of evoking active immune responses in the milieu of breast tumors through cancer vaccines has been introduced as one of the most novel immunotherapeutic approaches. Accordingly, a number of vaccines for the treatment or prevention of recurrence have been developed and are currently being studied in TNBC patients, while none have yet received any approvals. To elucidate the efficacy and safety of these vaccines, we performed a systematic review of the available literature on the topic. After searching the PubMed, Scopus, Web of Science, Embase, Cochrane CENTRAL, and Google Scholar databases, a total of 5701 results were obtained, from which 42 clinical studies were eventually included based on the predefined criteria. The overall quality of the included studies was acceptable. However, due to a lack of reporting outcomes of survival or progression in some studies (which were presented as conference abstracts) as well as the heterogeneity of the reported outcomes and study designs, we were not able to carry out a meta-analysis. A total of 32 different vaccines have so far been evaluated in TNBC patients, with the majority belonging to the peptide-based vaccine type. The other vaccines were in the cell or nucleic acid (RNA/DNA)-based categories. Most vaccines proved to be safe with low-grade, local adverse events and could efficiently evoke cellular immune responses; however, most trials were not able to demonstrate significant improvements in clinical indices of efficacy. This is in part due to the limited number of randomized studies, as well as the limited TNBC population of each trial. However, due to the encouraging results of the currently published trials, we anticipate that this strategy could show its potential through larger, phase III randomized studies in the near future.
Collapse
|
131
|
Bassiouni R, Idowu MO, Gibbs LD, Robila V, Grizzard PJ, Webb MG, Song J, Noriega A, Craig DW, Carpten JD. Spatial Transcriptomic Analysis of a Diverse Patient Cohort Reveals a Conserved Architecture in Triple-Negative Breast Cancer. Cancer Res 2023; 83:34-48. [PMID: 36283023 PMCID: PMC9812886 DOI: 10.1158/0008-5472.can-22-2682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 02/03/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive disease that disproportionately affects African American (AA) women. Limited targeted therapeutic options exist for patients with TNBC. Here, we employ spatial transcriptomics to interrogate tissue from a racially diverse TNBC cohort to comprehensively annotate the transcriptional states of spatially resolved cellular populations. A total of 38,706 spatial features from a cohort of 28 sections from 14 patients were analyzed. Intratumoral analysis of spatial features from individual sections revealed heterogeneous transcriptional substructures. However, integrated analysis of all samples resulted in nine transcriptionally distinct clusters that mapped across all individual sections. Furthermore, novel use of join count analysis demonstrated nonrandom directional spatial dependencies of the transcriptionally defined shared clusters, supporting a conserved spatio-transcriptional architecture in TNBC. These findings were substantiated in an independent validation cohort comprising 17,861 spatial features representing 15 samples from 8 patients. Stratification of samples by race revealed race-associated differences in hypoxic tumor content and regions of immune-rich infiltrate. Overall, this study combined spatial and functional molecular analyses to define the tumor architecture of TNBC, with potential implications in understanding TNBC disparities. SIGNIFICANCE Spatial transcriptomics profiling of a diverse cohort of triple-negative breast cancers and innovative informatics approaches reveal a conserved cellular architecture across cancers and identify proportional differences in tumor cell composition by race.
Collapse
Affiliation(s)
- Rania Bassiouni
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
| | - Michael O. Idowu
- Department of Pathology, Virginia Commonwealth University; Richmond, VA
| | - Lee D. Gibbs
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
| | - Valentina Robila
- Department of Pathology, Virginia Commonwealth University; Richmond, VA
| | | | - Michelle G. Webb
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
| | - Jiarong Song
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
| | - Ashley Noriega
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
| | - David W. Craig
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
- Translational and Clinical Sciences Program, Norris Comprehensive Cancer Center, University of Southern California; Los Angeles, CA
| | - John D. Carpten
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
- Translational and Clinical Sciences Program, Norris Comprehensive Cancer Center, University of Southern California; Los Angeles, CA
| |
Collapse
|
132
|
Balkrishna A, Umar Zango U, Kauser Nasir S, Arya V. A Clinical Cognizance of Molecular and Pathological Diagnostic Approach of TNBC. THERAPEUTIC DRUG TARGETS AND PHYTOMEDICINE FOR TRIPLE NEGATIVE BREAST CANCER 2023:26-46. [DOI: 10.2174/9789815079784123010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Genetic, transcriptional, and clinical heterogeneity of disease has remained
to be a prominent obstacle to the development of a targeted therapeutic approach
against TNBC. So far, based on tumor size, lymph node status, and histologic features
TNBC subtypes were stratified. Insights into inter and intratumoral heterogeneity of
TNBC were gained by next-generation sequencing, genomic, transcriptomic,
proteomic, and clinicopathological characterization. To depict tumor response to
neoadjuvant chemotherapy, radiological characterization may also a play significant
role. Biomarkers for subtyping TNBC were highly needed to depict the survival
outcome. This chapter discussed the available and possible molecular and pathological
diagnostic approaches to TNBC. Furthermore, the integration of morphological and
genomic data may emerge as a promising approach for the identification of new
therapeutic and prognostic markers to predict the likely outcome of the disease. This
chapter aims to highlight the molecular and pathological diagnostic approaches to
depict both metastatic and non-metastatic TNBC. <br>
Collapse
Affiliation(s)
- Acharya Balkrishna
- Patanjali Research Institute,Patanjali Herbal Research Department,Haridwar,India,
| | - Usman Umar Zango
- Sa'adatu Rimi College of Education,Department of Biology,Kumbotso,Nigeria,
| | - Saima Kauser Nasir
- Indian Institute of Science Bangalore,Department of Microbiology and Cell Biology (MCB),Bangalore,India,
| | - Vedpriya Arya
- Patanjali Research Institute,Patanjali Herbal Research Department,Haridwar,India,
| |
Collapse
|
133
|
Spínola-Lasso E, Montero JC, Jiménez-Monzón R, Estévez F, Quintana J, Guerra B, Elokely KM, León F, del Rosario H, Fernández-Pérez L, López MR, Díaz-Chico BN, McNaughton-Smith G, Pandiella A, Díaz-Chico JC. Chemical-proteomics Identify Peroxiredoxin-1 as an Actionable Target in Triple-negative Breast Cancer. Int J Biol Sci 2023; 19:1731-1747. [PMID: 37063429 PMCID: PMC10092761 DOI: 10.7150/ijbs.78554] [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: 09/01/2022] [Accepted: 02/22/2023] [Indexed: 04/18/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is difficult to treat; therefore, the development of drugs directed against its oncogenic vulnerabilities is a desirable goal. Herein, we report the antitumor effects of CM728, a novel quinone-fused oxazepine, against this malignancy. CM728 potently inhibited TNBC cell viability and decreased the growth of MDA-MB-231-induced orthotopic tumors. Furthermore, CM728 exerted a strong synergistic antiproliferative effect with docetaxel in vitro and this combination was more effective than the individual treatments in vivo. Chemical proteomic approaches revealed that CM728 bound to peroxiredoxin-1 (Prdx1), thereby inducing its oxidation. Molecular docking corroborated these findings. CM728 induced oxidative stress and a multi-signal response, including JNK/p38 MAPK activation and STAT3 inhibition. Interestingly, Prdx1 downregulation mimicked these effects. Finally, CM728 led to DNA damage, cell cycle blockage at the S and G2/M phases, and the activation of caspase-dependent apoptosis. Taken together, our results identify a novel compound with antitumoral properties against TNBC. In addition, we describe the mechanism of action of this drug and provide a rationale for the use of Prdx1 inhibitors, such as CM728, alone or in combination with other drugs, for the treatment of TNBC.
Collapse
Affiliation(s)
- Elena Spínola-Lasso
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología, Universidad de Las Palmas de Gran Canaria, The Canary Islands, Spain
| | - Juan Carlos Montero
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer-CSIC and CIBERONC, Salamanca, Spain
| | | | - Francisco Estévez
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología, Universidad de Las Palmas de Gran Canaria, The Canary Islands, Spain
| | - José Quintana
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología, Universidad de Las Palmas de Gran Canaria, The Canary Islands, Spain
| | - Borja Guerra
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Departamento de Ciencias Clínicas, Universidad de Las Palmas de Gran Canaria, The Canary Islands, Spain
- Unidad de Biomedicina asociada al CSIC, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, The Canary Islands, Spain and Instituto de Investigaciones Biomédicas “Alberto Sols'' CSIC - Universidad Autónoma de Madrid, Madrid, Spain
| | - Khaled M. Elokely
- Institute for Computational Molecular Science and Department of Chemistry, Temple University, Philadelphia, USA
| | - Francisco León
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, USA
| | - Henoc del Rosario
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología, Universidad de Las Palmas de Gran Canaria, The Canary Islands, Spain
| | - Leandro Fernández-Pérez
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Departamento de Ciencias Clínicas, Universidad de Las Palmas de Gran Canaria, The Canary Islands, Spain
- Unidad de Biomedicina asociada al CSIC, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, The Canary Islands, Spain and Instituto de Investigaciones Biomédicas “Alberto Sols'' CSIC - Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Rodríguez López
- Centro Atlántico del Medicamento S.A. (CEAMED S.A), La Laguna, The Canary Islands, Spain
| | - Bonifacio Nicolás Díaz-Chico
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología, Universidad de Las Palmas de Gran Canaria, The Canary Islands, Spain
- Instituto Canario de Investigación del Cáncer (ICIC), The Canary Islands, Spain
- Centro Atlántico del Medicamento S.A. (CEAMED S.A), La Laguna, The Canary Islands, Spain
| | - Grant McNaughton-Smith
- Centro Atlántico del Medicamento S.A. (CEAMED S.A), La Laguna, The Canary Islands, Spain
| | - Atanasio Pandiella
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer-CSIC and CIBERONC, Salamanca, Spain
- ✉ Corresponding authors: Atanasio Pandiella, Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), CSIC and CIBERONC. Campus Miguel de Unamuno, 37007, Salamanca, Spain. Tel./Fax: +34 923294815. E-mail: . ORCID: 0000-0002-4704-8971. Juan Carlos Díaz-Chico, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS). Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología. Universidad de Las Palmas de Gran Canaria. Paseo Blas Cabrera Felipe “Físico”, s/n, 35016, Las Palmas de Gran Canaria, Spain. Tel.: +34 928451445. E-mail: . ORCID: 0000-0002-0944-990X
| | - Juan Carlos Díaz-Chico
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología, Universidad de Las Palmas de Gran Canaria, The Canary Islands, Spain
- ✉ Corresponding authors: Atanasio Pandiella, Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), CSIC and CIBERONC. Campus Miguel de Unamuno, 37007, Salamanca, Spain. Tel./Fax: +34 923294815. E-mail: . ORCID: 0000-0002-4704-8971. Juan Carlos Díaz-Chico, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS). Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología. Universidad de Las Palmas de Gran Canaria. Paseo Blas Cabrera Felipe “Físico”, s/n, 35016, Las Palmas de Gran Canaria, Spain. Tel.: +34 928451445. E-mail: . ORCID: 0000-0002-0944-990X
| |
Collapse
|
134
|
Lu Y, Li Z, Zhang S, Zhang T, Liu Y, Zhang L. Cellular mitophagy: Mechanism, roles in diseases and small molecule pharmacological regulation. Theranostics 2023; 13:736-766. [PMID: 36632220 PMCID: PMC9830443 DOI: 10.7150/thno.79876] [Citation(s) in RCA: 149] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023] Open
Abstract
Cellular mitophagy means that cells selectively wrap and degrade damaged mitochondria through an autophagy mechanism, thus maintaining mitochondria and intracellular homeostasis. In recent years, mitophagy has received increasing attention as a research hotspot related to the pathogenesis of clinical diseases, such as neurodegenerative diseases, cardiovascular diseases, cancer, metabolic diseases, and so on. It has been found that the regulation of mitophagy may become a new direction for the treatment of some diseases. In addition, numerous small molecule modulators of mitophagy have also been reported, which provides new opportunities to comprehend the procedure and potential of therapeutic development. Taken together, in this review, we summarize current understanding of the mechanism of mitophagy, discuss the roles of mitophagy and its relationship with diseases, introduce the existing small-molecule pharmacological modulators of mitophagy and further highlight the significance of their development.
Collapse
Affiliation(s)
- Yingying Lu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Shuangqian Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Tongtong Zhang
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China,Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China
| | - Yanjun Liu
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China,Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China,✉ Corresponding authors: Yanjun Liu, E-mail: ; Lan Zhang, E-mail:
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China,✉ Corresponding authors: Yanjun Liu, E-mail: ; Lan Zhang, E-mail:
| |
Collapse
|
135
|
Jarroudi OA, Bairi KE, Curigliano G, Afqir S. Immune-Checkpoint Inhibitors: A New Line of Attack in Triple-Negative Breast Cancer. Cancer Treat Res 2023; 188:29-62. [PMID: 38175341 DOI: 10.1007/978-3-031-33602-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Poor prognosis is a distinctive feature of triple-negative breast cancer (TNBC). Chemotherapy has long represented the main and unique treatment for patients with TNBC. Recently, immune checkpoint inhibitors (ICIs) were investigated in several clinical trials and were approved for clinical use in TNBC patients that express programmed cell death protein-1 (PD-1) in combination with chemotherapy in the first-line setting. ICIs are also being investigated in the neoadjuvant and adjuvant settings for TNBC. This chapter aims to discuss different ICIs used to treat all TNBC stages to date.
Collapse
Affiliation(s)
- Ouissam Al Jarroudi
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco.
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco.
| | - Khalid El Bairi
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
| | - Giuseppe Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Said Afqir
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
| |
Collapse
|
136
|
Ademuyiwa FO, Gao F, Street CR, Chen I, Northfelt DW, Wesolowski R, Arora M, Brufsky A, Dees EC, Santa-Maria CA, Connolly RM, Force J, Moreno-Aspitia A, Herndon JM, Carmody M, Davies SR, Larson S, Pfaff KL, Jones SM, Weirather JL, Giobbie-Hurder A, Rodig SJ, Liu Z, Hagemann IS, Sharon E, Gillanders WE. A randomized phase 2 study of neoadjuvant carboplatin and paclitaxel with or without atezolizumab in triple negative breast cancer (TNBC) - NCI 10013. NPJ Breast Cancer 2022; 8:134. [PMID: 36585404 PMCID: PMC9803651 DOI: 10.1038/s41523-022-00500-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Atezolizumab with chemotherapy has shown improved progression-free and overall survival in patients with metastatic PD-L1 positive triple negative breast cancer (TNBC). Atezolizumab with anthracycline- and taxane-based neoadjuvant chemotherapy has also shown increased pathological complete response (pCR) rates in early TNBC. This trial evaluated neoadjuvant carboplatin and paclitaxel with or without atezolizumab in patients with clinical stages II-III TNBC. The co-primary objectives were to evaluate if chemotherapy and atezolizumab increase pCR rate and tumor infiltrating lymphocyte (TIL) percentage compared to chemotherapy alone in the mITT population. Sixty-seven patients (ages 25-78 years; median, 52 years) were randomly assigned - 22 patients to Arm A, and 45 to Arm B. Median follow up was 6.6 months. In the modified intent to treat population (all patients evaluable for the primary endpoints who received at least one dose of combination therapy), the pCR rate was 18.8% (95% CI 4.0-45.6%) in Arm A, and 55.6% (95% CI 40.0-70.4%) in Arm B (estimated treatment difference: 36.8%, 95% CI 8.5-56.6%; p = 0.018). Grade 3 or higher treatment-related adverse events occurred in 62.5% of patients in Arm A, and 57.8% of patients in Arm B. One patient in Arm B died from recurrent disease during the follow-up period. TIL percentage increased slightly from baseline to cycle 1 in both Arm A (mean ± SD: 0.6% ± 21.0%) and Arm B (5.7% ± 15.8%) (p = 0.36). Patients with pCR had higher median TIL percentages (24.8%) than those with non-pCR (14.2%) (p = 0.02). Although subgroup analyses were limited by the small sample size, PD-L1-positive patients treated with chemotherapy and atezolizumab had a pCR rate of 75% (12/16). The addition of atezolizumab to neoadjuvant carboplatin and paclitaxel resulted in a statistically significant and clinically relevant increased pCR rate in patients with clinical stages II and III TNBC. (Funded by National Cancer Institute).
Collapse
Affiliation(s)
| | - Feng Gao
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | | | - Ina Chen
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | | | - Robert Wesolowski
- Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Mili Arora
- UC Davis Comprehensive Cancer Center, Sacramento, CA, 95817, USA
| | - Adam Brufsky
- University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - E Claire Dees
- University of North Carolina School of Medicine, Chapel Hill, NC, 27514, USA
| | - Cesar A Santa-Maria
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, 21287, USA
| | | | - Jeremy Force
- Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - John M Herndon
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Madelyn Carmody
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Sherri R Davies
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Sarah Larson
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Kathleen L Pfaff
- Cancer Immune Monitoring and Analysis Center, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Stephanie M Jones
- Cancer Immune Monitoring and Analysis Center, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Jason L Weirather
- Cancer Immune Monitoring and Analysis Center, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Anita Giobbie-Hurder
- Cancer Immune Monitoring and Analysis Center, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Scott J Rodig
- Cancer Immune Monitoring and Analysis Center, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Zheng Liu
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Ian S Hagemann
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Elad Sharon
- National Cancer Institute, Bethesda, MD, 20892, USA
| | | |
Collapse
|
137
|
Li M, Yan T, Wang M, Cai Y, Wei Y. Advances in Single-Cell Sequencing Technology and Its Applications in Triple-Negative Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2022; 14:465-474. [PMID: 36540278 PMCID: PMC9760048 DOI: 10.2147/bctt.s388534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/07/2022] [Indexed: 09/10/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer and is mainly treated with chemotherapy-based combination therapy. In recent years, with the increasing development of global precision medicine, single-cell sequencing (SCS) has become one of the most promising technologies in the field of biotechnology. Moreover, the related application of this technology in TNBC has been applied and developed. By using SCS to study the heterogeneity of TNBC tumor cells, metastasis, drug resistance mechanisms, mutations, and cloning; it can further guide clinical chemotherapy, targeted therapy, and immunotherapy. To further reflect the importance of SCS in TNBC, this paper elaborated on and summarized the research and application progress of SCS in TNBC.
Collapse
Affiliation(s)
- Meng Li
- Graduate School of Qinghai University, Qinghai University, Xining, Qinghai Province, People’s Republic of China
| | - Tingting Yan
- Graduate School of Qinghai University, Qinghai University, Xining, Qinghai Province, People’s Republic of China
| | - Miaozhou Wang
- Graduate School of Qinghai University, Qinghai University, Xining, Qinghai Province, People’s Republic of China
| | - Yanqiu Cai
- Graduate School of Qinghai University, Qinghai University, Xining, Qinghai Province, People’s Republic of China
| | - Yingyuan Wei
- Graduate School of Qinghai University, Qinghai University, Xining, Qinghai Province, People’s Republic of China
| |
Collapse
|
138
|
Kilroy MK, Park S, Feroz W, Patel H, Mishra R, Alanazi S, Garrett JT. HER3 Alterations in Cancer and Potential Clinical Implications. Cancers (Basel) 2022; 14:cancers14246174. [PMID: 36551663 PMCID: PMC9776947 DOI: 10.3390/cancers14246174] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
In recent years, the third member of the HER family, kinase impaired HER3, has become a target of interest in cancer as there is accumulating evidence that HER3 plays a role in tumor growth and progression. This review focuses on HER3 activation in bladder, breast, colorectal, and lung cancer disease progression. HER3 mutations occur at a rate up to ~10% of tumors dependent on the tumor type. With patient tumors routinely sequenced for gene alterations in recent years, we have focused on HER3 mutations in bladder, breast, colon, and lung cancers particularly in response to targeted therapies and the potential to become a resistance mechanism. There are currently several HER3 targeting drugs in the pipeline, possibly improving outcomes for cancer patients with tumors containing HER3 activation and/or alterations.
Collapse
Affiliation(s)
- Mary Kate Kilroy
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | - SoYoung Park
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
- Cancer Research Scholars Program, College of Allied Health Sciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Wasim Feroz
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Hima Patel
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rosalin Mishra
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Samar Alanazi
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Joan T. Garrett
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
- Correspondence:
| |
Collapse
|
139
|
Monteiro FL, Stepanauskaite L, Williams C, Helguero LA. SETD7 Expression Is Associated with Breast Cancer Survival Outcomes for Specific Molecular Subtypes: A Systematic Analysis of Publicly Available Datasets. Cancers (Basel) 2022; 14:cancers14246029. [PMID: 36551516 PMCID: PMC9775934 DOI: 10.3390/cancers14246029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
SETD7 is a lysine N-methyltransferase that targets many proteins important in breast cancer (BC). However, its role and clinical significance remain unclear. Here, we used online tools and multiple public datasets to explore the predictive potential of SETD7 expression (high or low quartile) considering BC subtype, grade, stage, and therapy. We also investigated overrepresented biological processes associated with its expression using TCGA-BRCA data. SETD7 expression was highest in the Her2 (ERBB2)-enriched molecular subtype and lowest in the basal-like subtype. For the basal-like subtype specifically, higher SETD7 was consistently correlated with worse recurrence-free survival (p < 0.009). High SETD7-expressing tumours further exhibited a higher rate of ERBB2 mutation (20% vs. 5%) along with a poorer response to anti-Her2 therapy. Overall, high SETD7-expressing tumours showed higher stromal and lower immune scores. This was specifically related to higher counts of cancer-associated fibroblasts and endothelial cells, but lower B and T cell signatures, especially in the luminal A subtype. Genes significantly associated with SETD7 expression were accordingly overrepresented in immune response processes, with distinct subtype characteristics. We conclude that the prognostic value of SETD7 depends on the BC subtype and that SETD7 may be further explored as a potential treatment-predictive marker for immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Fátima Liliana Monteiro
- Department of Medical Sciences, Institute of Biomedicine—iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Lina Stepanauskaite
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, 141 83 Stockholm, Sweden
| | - Cecilia Williams
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, 141 83 Stockholm, Sweden
| | - Luisa A. Helguero
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
- Correspondence:
| |
Collapse
|
140
|
Mouron S, Bueno MJ, Lluch A, Manso L, Calvo I, Cortes J, Garcia-Saenz JA, Gil-Gil M, Martinez-Janez N, Apala JV, Caleiras E, Ximénez-Embún P, Muñoz J, Gonzalez-Cortijo L, Murillo R, Sánchez-Bayona R, Cejalvo JM, Gómez-López G, Fustero-Torre C, Sabroso-Lasa S, Malats N, Martinez M, Moreno A, Megias D, Malumbres M, Colomer R, Quintela-Fandino M. Phosphoproteomic analysis of neoadjuvant breast cancer suggests that increased sensitivity to paclitaxel is driven by CDK4 and filamin A. Nat Commun 2022; 13:7529. [PMID: 36477027 PMCID: PMC9729295 DOI: 10.1038/s41467-022-35065-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022] Open
Abstract
Precision oncology research is challenging outside the contexts of oncogenic addiction and/or targeted therapies. We previously showed that phosphoproteomics is a powerful approach to reveal patient subsets of interest characterized by the activity of a few kinases where the underlying genomics is complex. Here, we conduct a phosphoproteomic screening of samples from HER2-negative female breast cancer receiving neoadjuvant paclitaxel (N = 130), aiming to find candidate biomarkers of paclitaxel sensitivity. Filtering 11 candidate biomarkers through 2 independent patient sets (N = 218) allowed the identification of a subgroup of patients characterized by high levels of CDK4 and filamin-A who had a 90% chance of achieving a pCR in response to paclitaxel. Mechanistically, CDK4 regulates filamin-A transcription, which in turn forms a complex with tubulin and CLIP-170, which elicits increased binding of paclitaxel to microtubules, microtubule acetylation and stabilization, and mitotic catastrophe. Thus, phosphoproteomics allows the identification of explainable factors for predicting response to paclitaxel.
Collapse
Affiliation(s)
- S Mouron
- Breast Cancer Clinical Research Unit Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - M J Bueno
- Breast Cancer Clinical Research Unit Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - A Lluch
- Medical Oncology Department, Hospital Clínico Universitario, Valencia, Spain
| | - L Manso
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - I Calvo
- Medical Oncology Department MD, Anderson Cancer Center Madrid, Madrid, Spain
| | - J Cortes
- International Breast Cancer Center Quiron Group, Barcelona, Spain
- Vall d'Hebron Institute of Oncology, Vall d'Hebron Hospital, Barcelona, Spain
| | - J A Garcia-Saenz
- Medical Oncology Department, Hospital Clinico San Carlos, Madrid, Spain
| | - M Gil-Gil
- Medical Oncoogy Department Institut, Catala d'Oncologia-IDIBELL L'Hospitalet de, Llobregat, Spain
| | - N Martinez-Janez
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - J V Apala
- Breast Cancer Clinical Research Unit Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - E Caleiras
- Histopathology Unit Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - Pilar Ximénez-Embún
- Proteomics Unit Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - J Muñoz
- Proteomics Unit Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - L Gonzalez-Cortijo
- Medical Oncology Department, Hospital Universitario Quironsalud, Madrid, Spain
| | - R Murillo
- Pathology Department, Hospital Universitario Quironsalud, Madrid, Spain
| | - R Sánchez-Bayona
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - J M Cejalvo
- Medical Oncology Department, Hospital Clínico Universitario, Valencia, Spain
| | - G Gómez-López
- Bioinformatics Unit Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - C Fustero-Torre
- Bioinformatics Unit Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - S Sabroso-Lasa
- Genetic & Molecular Epidemiology Group Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - N Malats
- Genetic & Molecular Epidemiology Group Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - M Martinez
- Pathology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - A Moreno
- Pathology Department, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - D Megias
- Confocal Microscopy Unit Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - M Malumbres
- Cell Division and Cancer Group Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain
| | - R Colomer
- Medical Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
- Endowed Chair of Personalized Precision Medicine Universidad Autonoma de Madrid (UAM) - Fundacion Instituto Roche, Madrid, Spain
| | - M Quintela-Fandino
- Breast Cancer Clinical Research Unit Centro Nacional de Investigaciones Oncológicas - CNIO, Madrid, Spain.
- Endowed Chair of Personalized Precision Medicine Universidad Autonoma de Madrid (UAM) - Fundacion Instituto Roche, Madrid, Spain.
| |
Collapse
|
141
|
Doebley AL, Ko M, Liao H, Cruikshank AE, Santos K, Kikawa C, Hiatt JB, Patton RD, De Sarkar N, Collier KA, Hoge ACH, Chen K, Zimmer A, Weber ZT, Adil M, Reichel JB, Polak P, Adalsteinsson VA, Nelson PS, MacPherson D, Parsons HA, Stover DG, Ha G. A framework for clinical cancer subtyping from nucleosome profiling of cell-free DNA. Nat Commun 2022; 13:7475. [PMID: 36463275 PMCID: PMC9719521 DOI: 10.1038/s41467-022-35076-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
Cell-free DNA (cfDNA) has the potential to inform tumor subtype classification and help guide clinical precision oncology. Here we develop Griffin, a framework for profiling nucleosome protection and accessibility from cfDNA to study the phenotype of tumors using as low as 0.1x coverage whole genome sequencing data. Griffin employs a GC correction procedure tailored to variable cfDNA fragment sizes, which generates a better representation of chromatin accessibility and improves the accuracy of cancer detection and tumor subtype classification. We demonstrate estrogen receptor subtyping from cfDNA in metastatic breast cancer. We predict estrogen receptor subtype in 139 patients with at least 5% detectable circulating tumor DNA with an area under the receive operator characteristic curve (AUC) of 0.89 and validate performance in independent cohorts (AUC = 0.96). In summary, Griffin is a framework for accurate tumor subtyping and can be generalizable to other cancer types for precision oncology applications.
Collapse
Affiliation(s)
- Anna-Lisa Doebley
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Minjeong Ko
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hanna Liao
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - A Eden Cruikshank
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
| | | | - Caroline Kikawa
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Joseph B Hiatt
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Robert D Patton
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Navonil De Sarkar
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Anna C H Hoge
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Katharine Chen
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
| | - Anat Zimmer
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Zachary T Weber
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Mohamed Adil
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Jonathan B Reichel
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Paz Polak
- Department of Oncological Sciences, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | | | - Peter S Nelson
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - David MacPherson
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Daniel G Stover
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gavin Ha
- Division of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
| |
Collapse
|
142
|
Kesharwani P, Chadar R, Shukla R, Jain GK, Aggarwal G, Abourehab MAS, Sahebkar A. Recent advances in multifunctional dendrimer-based nanoprobes for breast cancer theranostics. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:2433-2471. [PMID: 35848467 DOI: 10.1080/09205063.2022.2103627] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Breast cancer (BC) undoubtedly is one of the most common type of cancers amongst women, which causes about 5 million deaths annually. The treatments and diagnostic therapy choices currently available for Breast Cancer is very much limited . Advancements in novel nanocarrier could be a promising strategy for diagnosis and treatments of this deadly disease. Dendrimer nanoformulation could be functionalized and explored for efficient targeting of overexpressed receptors on Breast Cancer cells to achieve targeted drug delivery, for diagnostics and to overcome the resistance of the cells towards particular chemotherapeutic. Additionally, the dendrimer have shown promising potential in the improvement of therapeutic value for Breast Cancer therapy by achieving synergistic co-delivery of chemotherapeutics and genetic materials for multidirectional treatment. In this review, we have highlighted the application of dendrimer as novel multifunctional nanoplatforms for the treatment and diagnosis of Breast Cancer.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.,University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Rahul Chadar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P, India
| | - Gaurav K Jain
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Geeta Aggarwal
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.,Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia, Egypt
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
143
|
Funnell T, O’Flanagan CH, Williams MJ, McPherson A, McKinney S, Kabeer F, Lee H, Salehi S, Vázquez-García I, Shi H, Leventhal E, Masud T, Eirew P, Yap D, Zhang AW, Lim JLP, Wang B, Brimhall J, Biele J, Ting J, Au V, Van Vliet M, Liu YF, Beatty S, Lai D, Pham J, Grewal D, Abrams D, Havasov E, Leung S, Bojilova V, Moore RA, Rusk N, Uhlitz F, Ceglia N, Weiner AC, Zaikova E, Douglas JM, Zamarin D, Weigelt B, Kim SH, Da Cruz Paula A, Reis-Filho JS, Martin SD, Li Y, Xu H, de Algara TR, Lee SR, Llanos VC, Huntsman DG, McAlpine JN, Shah SP, Aparicio S, Cannell IG, Casbolt H, Jauset C, Kovačević T, Mulvey CM, Nugent F, Ribes MP, Pearson I, Qosaj F, Sawicka K, Wild SA, Williams E, Laks E, Smith A, Lai D, Roth A, Balasubramanian S, Lee M, Bodenmiller B, Burger M, Kuett L, Tietscher S, Windhager J, Boyden ES, Alon S, Cui Y, Emenari A, Goodwin DR, Karagiannis ED, Sinha A, Wassie AT, Caldas C, Bruna A, Callari M, Greenwood W, Lerda G, Eyal-Lubling Y, Rueda OM, Shea A, Harris O, Becker R, Grimaldo F, Harris S, Vogl SL, Joyce JA, Watson SS, Tavare S, Dinh KN, Fisher E, Kunes R, Walton NA, Al Sa’d M, Chornay N, Dariush A, González-Solares EA, González-Fernández C, Yoldaş AK, Miller N, Zhuang X, Fan J, Lee H, Sepúlveda LA, Xia C, Zheng P, Shah SP, Aparicio S. Single-cell genomic variation induced by mutational processes in cancer. Nature 2022; 612:106-115. [PMID: 36289342 PMCID: PMC9712114 DOI: 10.1038/s41586-022-05249-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/17/2022] [Indexed: 12/15/2022]
Abstract
How cell-to-cell copy number alterations that underpin genomic instability1 in human cancers drive genomic and phenotypic variation, and consequently the evolution of cancer2, remains understudied. Here, by applying scaled single-cell whole-genome sequencing3 to wild-type, TP53-deficient and TP53-deficient;BRCA1-deficient or TP53-deficient;BRCA2-deficient mammary epithelial cells (13,818 genomes), and to primary triple-negative breast cancer (TNBC) and high-grade serous ovarian cancer (HGSC) cells (22,057 genomes), we identify three distinct 'foreground' mutational patterns that are defined by cell-to-cell structural variation. Cell- and clone-specific high-level amplifications, parallel haplotype-specific copy number alterations and copy number segment length variation (serrate structural variations) had measurable phenotypic and evolutionary consequences. In TNBC and HGSC, clone-specific high-level amplifications in known oncogenes were highly prevalent in tumours bearing fold-back inversions, relative to tumours with homologous recombination deficiency, and were associated with increased clone-to-clone phenotypic variation. Parallel haplotype-specific alterations were also commonly observed, leading to phylogenetic evolutionary diversity and clone-specific mono-allelic expression. Serrate variants were increased in tumours with fold-back inversions and were highly correlated with increased genomic diversity of cellular populations. Together, our findings show that cell-to-cell structural variation contributes to the origins of phenotypic and evolutionary diversity in TNBC and HGSC, and provide insight into the genomic and mutational states of individual cancer cells.
Collapse
Affiliation(s)
- Tyler Funnell
- grid.5386.8000000041936877XTri-Institutional PhD Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, NY USA ,grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Ciara H. O’Flanagan
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Marc J. Williams
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Andrew McPherson
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Steven McKinney
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Farhia Kabeer
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia Canada
| | - Hakwoo Lee
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia Canada
| | - Sohrab Salehi
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Ignacio Vázquez-García
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Hongyu Shi
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Emily Leventhal
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Tehmina Masud
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Peter Eirew
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Damian Yap
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Allen W. Zhang
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Jamie L. P. Lim
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Beixi Wang
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Jazmine Brimhall
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Justina Biele
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Jerome Ting
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Vinci Au
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Michael Van Vliet
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Yi Fei Liu
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Sean Beatty
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Daniel Lai
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia Canada
| | - Jenifer Pham
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Diljot Grewal
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Douglas Abrams
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Eliyahu Havasov
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Samantha Leung
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Viktoria Bojilova
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Richard A. Moore
- grid.434706.20000 0004 0410 5424Michael Smith Genome Sciences Centre, Vancouver, British Columbia Canada
| | - Nicole Rusk
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Florian Uhlitz
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Nicholas Ceglia
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Adam C. Weiner
- grid.5386.8000000041936877XTri-Institutional PhD Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, NY USA ,grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Elena Zaikova
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - J. Maxwell Douglas
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Dmitriy Zamarin
- grid.51462.340000 0001 2171 9952GYN Medical Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Britta Weigelt
- grid.51462.340000 0001 2171 9952Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Sarah H. Kim
- grid.51462.340000 0001 2171 9952Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Arnaud Da Cruz Paula
- grid.51462.340000 0001 2171 9952Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Jorge S. Reis-Filho
- grid.51462.340000 0001 2171 9952Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Spencer D. Martin
- grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia Canada
| | - Yangguang Li
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Hong Xu
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Teresa Ruiz de Algara
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - So Ra Lee
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - Viviana Cerda Llanos
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada
| | - David G. Huntsman
- grid.248762.d0000 0001 0702 3000Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia Canada
| | - Jessica N. McAlpine
- grid.17091.3e0000 0001 2288 9830Department of Gynecology and Obstetrics, University of British Columbia, Vancouver, British Columbia Canada
| | | | - Sohrab P. Shah
- grid.51462.340000 0001 2171 9952Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Samuel Aparicio
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada. .,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Kang S, Borgsmüller N, Valecha M, Kuipers J, Alves JM, Prado-López S, Chantada D, Beerenwinkel N, Posada D, Szczurek E. SIEVE: joint inference of single-nucleotide variants and cell phylogeny from single-cell DNA sequencing data. Genome Biol 2022; 23:248. [PMID: 36451239 PMCID: PMC9714196 DOI: 10.1186/s13059-022-02813-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 11/08/2022] [Indexed: 12/02/2022] Open
Abstract
We present SIEVE, a statistical method for the joint inference of somatic variants and cell phylogeny under the finite-sites assumption from single-cell DNA sequencing. SIEVE leverages raw read counts for all nucleotides and corrects the acquisition bias of branch lengths. In our simulations, SIEVE outperforms other methods in phylogenetic reconstruction and variant calling accuracy, especially in the inference of homozygous variants. Applying SIEVE to three datasets, one for triple-negative breast (TNBC), and two for colorectal cancer (CRC), we find that double mutant genotypes are rare in CRC but unexpectedly frequent in the TNBC samples.
Collapse
Affiliation(s)
- Senbai Kang
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
| | - Nico Borgsmüller
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, 4058 Basel, Switzerland
| | - Monica Valecha
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Jack Kuipers
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, 4058 Basel, Switzerland
| | - Joao M. Alves
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Sonia Prado-López
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Institute of Solid State Electronics E362, Technische Universität Wien, Vienna, Austria
| | - Débora Chantada
- Department of Pathology, Hospital Álvaro Cunqueiro, Vigo, Spain
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, 4058 Basel, Switzerland
| | - David Posada
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Department of Biochemistry, Genetics, and Immunology, Universidade de Vigo, 36310 Vigo, Spain
| | - Ewa Szczurek
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
| |
Collapse
|
145
|
Kudelova E, Smolar M, Holubekova V, Hornakova A, Dvorska D, Lucansky V, Koklesova L, Kudela E, Kubatka P. Genetic Heterogeneity, Tumor Microenvironment and Immunotherapy in Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms232314937. [PMID: 36499265 PMCID: PMC9735793 DOI: 10.3390/ijms232314937] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
Heterogeneity of triple-negative breast cancer is well known at clinical, histopathological, and molecular levels. Genomic instability and greater mutation rates, which may result in the creation of neoantigens and enhanced immunogenicity, are additional characteristics of this breast cancer type. Clinical outcome is poor due to early age of onset, high metastatic potential, and increased likelihood of distant recurrence. Consequently, efforts to elucidate molecular mechanisms of breast cancer development, progression, and metastatic spread have been initiated to improve treatment options and improve outcomes for these patients. The extremely complex and heterogeneous tumor immune microenvironment is made up of several cell types and commonly possesses disorganized gene expression. Altered signaling pathways are mainly associated with mutated genes including p53, PIK3CA, and MAPK, and which are positively correlated with genes regulating immune response. Of note, particular immunity-associated genes could be used in prognostic indexes to assess the most effective management. Recent findings highlight the fact that long non-coding RNAs also play an important role in shaping tumor microenvironment formation, and can mediate tumor immune evasion. Identification of molecular signatures, through the use of multi-omics approaches, and effector pathways that drive early stages of the carcinogenic process are important steps in developing new strategies for targeted cancer treatment and prevention. Advances in immunotherapy by remodeling the host immune system to eradicate tumor cells have great promise to lead to novel therapeutic strategies. Current research is focused on combining immune checkpoint inhibition with chemotherapy, PARP inhibitors, cancer vaccines, or natural killer cell therapy. Targeted therapies may improve therapeutic response, eliminate therapeutic resistance, and improve overall patient survival. In the future, these evolving advancements should be implemented for personalized medicine and state-of-art management of cancer patients.
Collapse
Affiliation(s)
- Eva Kudelova
- Clinic of Surgery and Transplant Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Marek Smolar
- Clinic of Surgery and Transplant Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Veronika Holubekova
- Biomedical Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Andrea Hornakova
- Biomedical Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Dana Dvorska
- Biomedical Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Vincent Lucansky
- Biomedical Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Gynecology and Obstetrics, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Erik Kudela
- Clinic of Gynecology and Obstetrics, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
- Correspondence:
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
146
|
Lin S, Qin HZ, Li ZY, Zhu H, Long L, Xu LB. Gallic acid suppresses the progression of triple-negative breast cancer HCC1806 cells via modulating PI3K/AKT/EGFR and MAPK signaling pathways. Front Pharmacol 2022; 13:1049117. [PMID: 36523491 PMCID: PMC9744937 DOI: 10.3389/fphar.2022.1049117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/15/2022] [Indexed: 11/04/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a severe threat to women's health because of its aggressive nature, early age of onset, and high recurrence rate. Therefore, in this study, we aimed to evaluate the anti-tumor effects of Gallic acid (GA) on the TNBC HCC1806 cells in vitro. The cell proliferation was detected by MTT and plate clone formation assays, cell apoptosis, cell cycle, and mitochondrial membrane potential (MMP) were analyzed by flow cytometry and Hoechst 33258 staining assays, and the intracellular reactive oxygen species (ROS) accumulation were also investigated. Real-Time PCR and western blot were examined to explore the mechanism of action. The results indicated that GA suppressed HCC1806 cells proliferation and promoted HCC1806 cells apoptosis. Meanwhile, GA treatment changed the morphology of the HCC1806 cells. In addition, GA blocked the HCC1806 cells cycle in the S phase, and it induced cells apoptosis accompanied by ROS accumulation and MMP depolarization. Real-Time PCR results suggested that GA increased Bax, Caspase-3, Caspase-9, P53, JINK and P38 mRNA expression, and decreased Bcl-2, PI3K, AKT and EGFR mRNA expression. Western blotting results suggested that GA increased Bax, cleaved-Caspase-3, cleaved-Caspase-9, P53, P-ERK1/2, P-JNK, P-P38 proteins expression, and decreased Bcl-2, P-PI3K, P-AKT, P-EGFR proteins expression. Furthermore, molecular docking suggested that GA has the high affinity for PI3K, AKT, EGFR, ERK1/2, JNK, and P38. In conclusion, GA could suppress HCC1806 cells proliferation and promote HCC1806 cells apoptosis through the mitochondrial apoptosis pathway and induces ROS generation which further inhibits PI3K/AKT/EGFR and activates MAPK signaling pathways. Our study will provide some new references for using GA in the treatment of TNBC.
Collapse
Affiliation(s)
- Si Lin
- Guangxi Key Laboratory of Zhuang and Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Hui-Zhen Qin
- Guangxi Key Laboratory of Zhuang and Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Ze-Yu Li
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Hua Zhu
- Guangxi Key Laboratory of Zhuang and Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Li Long
- Guangxi International Zhuang Medicine Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Li-Ba Xu
- Guangxi Key Laboratory of Zhuang and Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
147
|
Lv Y, Bai Z, Wang X, Liu J, Li Y, Zhang X, Shan Y. Comprehensive evaluation of breast cancer immunotherapy and tumor microenvironment characterization based on interleukin genes-related risk model. Sci Rep 2022; 12:20524. [PMID: 36443508 PMCID: PMC9705306 DOI: 10.1038/s41598-022-25059-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Breast cancer (BRCA) is the most prevalent malignancy and the leading cause of death in women. Interleukin (IL) genes are critical in tumor initiation and control. Nevertheless, the prognosis value of the IL in BRCA remains unclear. We collected data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), and 94 IL genes were identified from GeneCard. Based on the random forest (RF), least absolute shrinkage and selection operator (LASSO) analysis, and multivariate Cox regression analysis, we constructed an IL signature. GSE22219, GSE25065, and GSE21653 were derived as validation sets. The expression differences in the tumor microenvironment (TME), immunotherapy, and chemosensitivity of BRCA between the high- and low-risk groups were evaluated. Overall, 21 IL genes were selected to construct an IL risk model, of which IL18BP, IL17D, and IL23A were the first time identified as prognostic genes in BRCA. IL score could distinguish BRCA patients with inferior outcomes, and AUC of it was 0.70, 0.76, and 0.72 for 1-,3- and 5- years, respectively, which was also verified in GSE22219, GSE25065, and GSE21653 cohorts. Meanwhile, compared to luminal A and luminal B, HER2-positive and TNBC had significantly higher IL score. Besides, the high-risk group had a significantly higher prevalence of TP53 and TTN but a lower prevalence of PIK3CA, as well as higher tumor mutation burden (TMB) and neoantigen level. High- and low-risk groups exhibited notable differences in immunomodulators and tumor infiltrates immune cells (TIICs), and the high-risk group had significantly lower Tumor Immune Dysfunction and Exclusion (TIDE) score. Additionally, the high-risk group has more responders to immune or anti-HER2 combination therapy, whereas the low-risk group has higher sensitivity to docetaxel and paclitaxel. Consequently, we constructed a reliable risk model based on the IL genes, which can provide more information on both the risk stratification and personalizing management strategies for BRCA.
Collapse
Affiliation(s)
- Yalei Lv
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, China
| | - Zihe Bai
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, China
| | - Xiaoyan Wang
- The Fifth Ward of Medical Oncology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Jiayin Liu
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, China
| | - Yuntao Li
- Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaolin Zhang
- Department of Epidemiology and Statistics, Hebei Medical University, Shijiazhuang, China
| | - Yujie Shan
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, China.
| |
Collapse
|
148
|
Ke L, Li S, Cui H. The prognostic role of tumor mutation burden on survival of breast cancer: a systematic review and meta-analysis. BMC Cancer 2022; 22:1185. [PMID: 36397030 PMCID: PMC9673350 DOI: 10.1186/s12885-022-10284-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Background As a potential genetic biomarker, tumor mutation burden (TMB) has made progress in numerous tumors. There are limited data regarding TMB and its prognostic role is controversial in breast cancer. This systematic review and meta-analysis were conducted to assess the prognostic value of TMB on survival of breast cancer. Methods The databases PubMed, Embase, Web of Science, and Cochrane Library were searched for articles published through May 31, 2022. Moreover, effective data were extracted from included studies and calculated pooled effects of hazard ratio (HR) for overall survival (OS) and progression-free survival (PFS) by STATA 16.0. Heterogeneity was conducted by the I2 statistic and p-value. Using publication bias evaluation, sensitivity analysis, and subgroup analysis, the origin of heterogeneity was further investigated. Results They were up to 1,722 patients collected from sixteen cohorts for this analysis. The pooled effects of HR for both OS (HR: 1.14, 95% CI: 0.83,1.58, p > 0.01) and PFS (HR: 0.96, 95% CI: 0.53,1.71, p > 0.01) indicated no statistically significant difference in the high TMB and low TMB group. In immune checkpoint inhibitors (ICIs) subgroup, high TMB patients demonstrated benefit of OS (HR: 0.72, 95% CI: 0.59,0.87, p = 0.001) and PFS (HR: 0.52, 95% CI: 0.35,0.77, p < 0.001), whereas difference was not statistically significant in the non-ICIs subgroup (OS, HR:1.76, 95% CI: 0.97,3.20, p = 0.062; PFS, HR:2.31, 95% CI: 0.89,5.97, p = 0.086). In addition, sensitivity analysis revealed that the pooled effects were stable. The funnel plot and Begg's test suggested the absence of publication bias. Conclusion Meta-analysis revealed that the prognostic relevance of TMB in breast cancer is limited in scope. High TMB may be associated with longer survival only in ICIs-based treatment, but the association is not evident in non-ICIs-based treatment. Trial registration [https://www.crd.york.ac.uk/PROSPERO], Prospective Register of Systematic Reviews (PROSPERO), identifier: CRD42022342488. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10284-1.
Collapse
|
149
|
Kolyvas EA, Caldas C, Kelly K, Ahmad SS. Androgen receptor function and targeted therapeutics across breast cancer subtypes. Breast Cancer Res 2022; 24:79. [PMID: 36376977 PMCID: PMC9664788 DOI: 10.1186/s13058-022-01574-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Despite significant progress in breast cancer (BC) therapy, it is globally the most commonly diagnosed cancer and leads to the death of over 650,000 women annually. Androgen receptor (AR) is emerging as a potential new therapeutic target in BC. While the role of AR is well established in prostate cancer (PCa), its function in BC remains incompletely understood. Emerging data show that AR's role in BC is dependent on several factors including, but not limited to, disease subtype, tumour microenvironment, and levels of circulating oestrogens and androgens. While targeting AR in PCa is becoming increasingly effective, these advances have yet to make any significant impact on the care of BC patients. However, this approach is increasingly being evaluated in BC and it is clear that improvements in our understanding of AR's role in BC will increase the likelihood of success for AR-targeted therapies. This review summarizes our current understanding of the function of AR across BC subtypes. We highlight limitations in our current knowledge and demonstrate the importance of categorizing BC subtypes effectively, in relation to determining AR activity. Further, we describe the current state of the art regarding AR-targeted approaches for BC as monotherapy or in combination with radiotherapy.
Collapse
Affiliation(s)
- Emily A Kolyvas
- Cancer Research UK Cambridge Institute, Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- NIH-Oxford-Cambridge Scholars Program, Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Breast Cancer Programme, CRUK Cambridge Centre, Cambridge, CB2 0RE, UK
- Cambridge Breast Cancer Research Unit, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Kathleen Kelly
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Saif S Ahmad
- Cancer Research UK Cambridge Institute, Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK.
- Department of Oncology, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SP, UK.
| |
Collapse
|
150
|
Tumor suppressor DEAR1 regulates mammary epithelial cell fate and predicts early onset and metastasis in triple negative breast cancer. Sci Rep 2022; 12:19504. [PMID: 36376460 PMCID: PMC9663828 DOI: 10.1038/s41598-022-22417-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a disease of poor prognosis, with the majority classified as the basal-like subtype associated with epithelial-mesenchymal transition and metastasis. Because basal breast cancers originate from proliferative luminal progenitor-like cells upon dysregulation of proper luminal differentiation, genes regulating luminal-basal transition are critical to elucidate novel therapeutic targets to improve TNBC outcomes. Herein we demonstrate that the tumor suppressor DEAR1/TRIM62 is a critical regulator of luminal cell fate. DEAR1 loss in human mammary epithelial cells results in significantly enhanced mammosphere formation that is accelerated in the presence of TGF-β/SMAD3 signaling. Mammospheres formed following DEAR1 loss are enriched for ALDH1A1 and CK5 expression, EpCAM-/CD49f+ and CD44high/24low basal-like epithelial cells, indicating that DEAR1 regulates stem/progenitor cell properties and luminal-basal progenitor transition. We show that DEAR1 maintains luminal differentiation as a novel ubiquitin ligase for SNAI2/SLUG, a master regulator driving stemness and generation of basal-like progenitor populations. We also identify a significant inverse correlation between DEAR1 and SNAI2 expression in a 103 TNBC case cohort and show that low DEAR1 expression significantly correlates with young age of onset and shorter time to metastasis, suggesting DEAR1 could serve as a biomarker to stratify early onset TNBCs for targeted stem cell therapies.
Collapse
|