1
|
Cui R, Wang G, Liu F, Wang Y, Zhao Z, Mutailipu M, Mu H, Jiang X, Le W, Yang L, Chen B. Neurturin-induced activation of GFRA2-RET axis potentiates pancreatic cancer glycolysis via phosphorylated hexokinase 2. Cancer Lett 2025; 621:217583. [PMID: 39988080 DOI: 10.1016/j.canlet.2025.217583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Pancreatic cancer, characterized by its insidious onset, high invasiveness, resistance to chemotherapy, and a grim prognosis, with a five-year survival rate hovering below 10 %. The identification of novel therapeutic targets addressing tumor progression is therefore critically important. While perineural invasion (PNI) is recognized as a pathological hallmark and key driver of pancreatic cancer progression, its role in metabolic reprogramming of malignant cells has not been fully elucidated. Using integrated metabolomics approaches, we found perineural invasion in pancreatic cancer significantly enhancing glycolytic flux of pancreatic cancer. Our data delineate a neuroendocrine-paracrine signaling axis in which neurturin secreted by neuronal cells binds to the GFRA2 receptor on pancreatic cancer cells, inducing RET kinase recruitment and subsequent heterodimer assembly. This receptor tyrosine kinase complex phosphorylates hexokinase 2 (HK2) at the evolutionarily conserved Ser122 residue, augmenting its hexokinase activity, ultimately driving aerobic glycolysis flux and fueling pancreatic cancer growth. In vivo experiments corroborate our findings, revealing that neurturin blockade effectively halts pancreatic cancer progression and synergizes with RET inhibitors. Our research underscores neurturin as a promising therapeutic target for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Ran Cui
- Department of Hepatopancreatobiliary Surgery, Frontier Science Center for Stem Cell Research & Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China
| | - Gaoming Wang
- Department of Hepatopancreatobiliary Surgery, Frontier Science Center for Stem Cell Research & Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China
| | - Fuguo Liu
- Department of Hepatopancreatobiliary Surgery, Frontier Science Center for Stem Cell Research & Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China
| | - Yongkun Wang
- Department of Hepatopancreatobiliary Surgery, Frontier Science Center for Stem Cell Research & Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China
| | - Zinan Zhao
- Department of Hepatopancreatobiliary Surgery, Frontier Science Center for Stem Cell Research & Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China
| | - Muladili Mutailipu
- Department of Hepatopancreatobiliary Surgery, Frontier Science Center for Stem Cell Research & Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China
| | - Huiling Mu
- Department of Hepatopancreatobiliary Surgery, Frontier Science Center for Stem Cell Research & Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China; Department of Biobank, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China
| | - Xiaohua Jiang
- Department of Hepatopancreatobiliary Surgery, Frontier Science Center for Stem Cell Research & Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China
| | - Wenjun Le
- Department of Hepatopancreatobiliary Surgery, Frontier Science Center for Stem Cell Research & Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China.
| | - Ludi Yang
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China.
| | - Bo Chen
- Department of Hepatopancreatobiliary Surgery, Frontier Science Center for Stem Cell Research & Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China.
| |
Collapse
|
2
|
Uemoto Y, Lin CCA, Wang B, Ye D, Fang YV, Bikorimana E, Napolitano F, Chica-Parrado MR, Li C, Mendiratta S, Chen C, Hanker AB, Arteaga CL. Selective degradation of FGFR1/2 overcomes antiestrogen resistance in ER+ breast cancer with FGFR1/2 alterations. Cancer Lett 2025; 619:217668. [PMID: 40127812 DOI: 10.1016/j.canlet.2025.217668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/25/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
FGFR1 amplification and FGFR1/2 activating mutations have been associated with antiestrogen resistance in estrogen receptor-positive (ER+) breast cancer. However, there are no approved FGFR1-targeted therapies for breast cancers harboring these alterations. In this study, we investigated the selective degradation of FGFR1/2 using the proteolysis-targeting chimera (PROTAC) DGY-09-192 as a novel therapeutic strategy in ER + breast cancers harboring FGFR1/2 somatic alterations. Treatment of ER+/FGFR1-amplified breast cancer cells and patient-derived xenografts with DGY-09-192 resulted in sustained degradation of FGFR1 in a proteasome-dependent manner and suppressed downstream signal transduction. The combination of DGY-09-192 and the ERα degrader fulvestrant resulted in complete cell growth arrest and tumor regression of ER+/FGFR1-amplified patients-derived xenografts. In addition, we tested the effect of DGY-09-192 on breast cancer cells expressing FGFR1N546K and FGFR2K659E hotspot kinase domain mutations as well as ER-negative breast cancer cells harboring FGFR2 gene amplification. Treatment with DGY-09-192 resulted in the degradation of mutant FGFR1/2 and blocked mutant receptor-induced signal transduction and antiestrogen resistance. Collectively, our study suggests that degradation of FGFR1/2, in combination with antiestrogens, can be leveraged as a therapeutic strategy in ER + breast cancers harboring FGFR1/2 driver alterations.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Female
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Animals
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Xenograft Model Antitumor Assays
- Proteolysis/drug effects
- Mice
- Fulvestrant/pharmacology
- Cell Line, Tumor
- Receptors, Estrogen/metabolism
- Signal Transduction/drug effects
- Mutation
- Cell Proliferation/drug effects
- Estrogen Receptor Modulators/pharmacology
- Estrogen Receptor alpha/metabolism
- MCF-7 Cells
Collapse
Affiliation(s)
- Yasuaki Uemoto
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Chang-Ching A Lin
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Bingnan Wang
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Dan Ye
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Yisheng V Fang
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX, USA; Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | - Cheung Li
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | | | - Chuo Chen
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ariella B Hanker
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX, USA.
| | - Carlos L Arteaga
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX, USA.
| |
Collapse
|
3
|
Hao Q, Zhao W, Li Z, Lai Y, Wang Y, Yang Q, Zhang L. Combination therapy and dual-target inhibitors based on cyclin-dependent kinases (CDKs): Emerging strategies for cancer therapy. Eur J Med Chem 2025; 289:117465. [PMID: 40037064 DOI: 10.1016/j.ejmech.2025.117465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/22/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Cyclin-dependent kinases (CDKs) are pivotal regulators of the cell cycle and transcriptional machinery, making them attractive targets for cancer therapy. While CDK inhibitors have demonstrated promising clinical outcomes, they also face challenges in enhancing efficacy, particularly in overcoming drug resistance. Combination therapies have emerged as a key strategy to augment the effectiveness of CDK inhibitors when used alongside other kinase inhibitors or non-kinase-targeted agents. Dual-target inhibitors that simultaneously inhibit CDKs and other oncogenic drivers are gaining attention, offering novel avenues to optimize cancer therapy. Based on the structural characterization and biological functions of CDKs, this review comprehensively examines the structure-activity relationship (SAR) of existing dual-target CDK inhibitors from a drug design perspective. We also thoroughly investigate the preclinical studies and clinical translational potential of combination therapies and dual-target inhibitors. Tailoring CDK inhibitors to specific cancer subtypes and therapeutic settings will inspire innovative approaches for the next generation of CDK-related therapies, ultimately improving patient survival.
Collapse
Affiliation(s)
- Qi Hao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Wenzhe Zhao
- 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
| | - Yue Lai
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yan Wang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Qianqian Yang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China; Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
4
|
Guo Z, Dong RW, Wu Y, Dong S, Alahari SK. Cyclin-dependent kinase 4 and 6 inhibitors in breast cancer treatment. Oncogene 2025; 44:1135-1152. [PMID: 40200094 DOI: 10.1038/s41388-025-03378-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Breast cancer is the second largest cancer in the world, and it has highest mortality rate in women worldwide. The aberrant activation of the cyclin-dependent kinase 4 and 6 (CDK4/6) pathway plays an important role in uncontrolled breast cancer cell proliferation. Therefore, targeting CDK4/6 to improve overall survival rates has been a strong interest in breast cancer therapeutics. Till date, four CDK4/6 inhibitors have been developed and approved for hormone receptor-positive and human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer therapies with great success. However, acquired resistance to CDK4/6 inhibitors has emerged and limits their effectiveness in breast cancer. In this review, we systematically discussed the mechanisms of resistance to CDK4/6 inhibitors including the cell cycle-specific and cell cycle-nonspecific mechanisms. Also, we analyzed combination strategies with other signaling inhibitors in clinical and preclinical settings that further expand the clinical application of CDK4/6 inhibitors in future breast cancer therapies.
Collapse
Affiliation(s)
- Zhengfei Guo
- TYK Medicines, Inc., Huzhou, Zhejiang, 313100, China
| | - Richard W Dong
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Yusheng Wu
- TYK Medicines, Inc., Huzhou, Zhejiang, 313100, China
| | - Shengli Dong
- TYK Medicines, Inc., Huzhou, Zhejiang, 313100, China.
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
5
|
Dommer AP, Kumarasamy V, Wang J, O’Connor TN, Roti M, Mahan S, McLean K, Knudsen ES, Witkiewicz AK. Tumor Suppressors Condition Differential Responses to the Selective CDK2 Inhibitor BLU-222. Cancer Res 2025; 85:1310-1326. [PMID: 39945638 PMCID: PMC11977231 DOI: 10.1158/0008-5472.can-24-2244] [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: 07/02/2024] [Revised: 11/15/2024] [Accepted: 01/22/2025] [Indexed: 03/23/2025]
Abstract
Cyclin-dependent kinase 2 (CDK2) inhibitors have recently been developed and have entered clinical trials. Combination approaches can help broaden the use of therapeutic agents and establish more effective treatments. Here, we evaluated the selective CDK2 inhibitor BLU-222 for mechanisms of response in the context of ovarian and breast cancer models. Sensors of cellular CDK activity indicated that sensitivity to either CDK4/6 or CDK2 inhibition was related to the differential dependence on a single CDK for G1-S transition. Unlike CDK4/6 inhibitors, BLU-222 was able to robustly inhibit proliferation through cell-cycle inhibition in both G1 and G2 phases. However, it remained possible for cells to reenter the cell cycle upon drug withdrawal. The antiproliferative strength and impact on G1-S versus G2-M accumulation was mediated by the RB tumor suppressor. To broaden the sensitivity to CDK2 inhibition, combinatorial drug screens were performed that identified both synergistic (e.g., CDK4/6 inhibitors) and antagonistic (e.g., WEE1 inhibitors) relationships. Models that were exceptionally sensitive to CDK2 inhibition displayed coordinate expression of cyclin E1 and P16INK4A, an endogenous CDK4/6 inhibitor. Functional studies demonstrated that P16INK4A and CDK4/6 activity were key mediators of sensitivity to BLU-222. Clinical gene and protein expression analyses revealed a positive correlation between cyclin E1 and P16INK4A and identified that ∼25% of ovarian cancers exhibited coordinate expression of cyclin E, P16INK4A, and RB, indicative of strong sensitivity to CDK2 inhibition. Together, this work advances a precision strategy for the use of CDK2 inhibitors in the context of ovarian and breast cancers. Significance: The CDK2-specific inhibitor BLU-222 shows preclinical efficacy in breast and ovarian cancer with select determinants of response and holds promise in combinatorial strategies. See related article by House and colleagues, p. 1297.
Collapse
Affiliation(s)
- Adam P. Dommer
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Vishnu Kumarasamy
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jianxin Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Thomas N. O’Connor
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Michelle Roti
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sidney Mahan
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Karen McLean
- Department of Gynecology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Erik S. Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Agnieszka K. Witkiewicz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
6
|
Yin R, Gao J, Liu Y, Guo C. Functional analysis of the effects of propofol on tamoxifen‑resistant breast cancer cells: Insights into transcriptional regulation. Oncol Lett 2025; 29:194. [PMID: 40041408 PMCID: PMC11878209 DOI: 10.3892/ol.2025.14940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/06/2025] [Indexed: 03/06/2025] Open
Abstract
Although 70% of patients with estrogen receptor-positive breast cancer benefit from tamoxifen (TAM) therapy, the development of resistance to TAM leads to high rates of metastasis and a poor prognosis. Propofol, a commonly used anesthetic, can inhibit the occurrence and progression of breast cancer. In the present study, the effects of propofol on TAM-resistant (TR) breast cancer cells were evaluated. MCF7-TR cells were treated with or without propofol. Subsequently, cell cycle progression and the induction of apoptosis were detected by flow cytometry, whereas cell proliferation was assessed using Cell Counting Kit-8 and colony formation assays. Furthermore, the potential transcriptional regulatory effects of propofol on MCF7-TR cells were investigated using RNA sequencing. The results indicated that propofol significantly promoted cell cycle arrest, induced apoptosis, and inhibited proliferation and colony formation in MCF7-TR cells. Furthermore, transcriptome sequencing analysis revealed 1,065 differentially expressed genes between propofol-treated MCF7-TR and untreated MCF7-TR cells. Gene Ontology annotation enrichment analysis, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis and Gene Set Enrichment Analysis indicated that propofol affected the expression levels of genes located on the 'plasma membrane' and 'cell periphery', while mainly regulating signals involved in cancer biology, immune response and metabolic pathways. These results identified the potential effects of propofol on TR breast cancer cells and provided a theoretical basis for clinical treatment, particularly for individuals with TAM resistance.
Collapse
Affiliation(s)
- Runyang Yin
- Department of Anesthesiology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Jing Gao
- First Clinical Medical College, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Yang Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Chunyan Guo
- Department of Anesthesiology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| |
Collapse
|
7
|
Zhang W, Bradburn D, Heidebrink G, Liu Y, Jang H, Nussinov R, Kõivomägi M. Distinct Allosteric Networks in CDK4 and CDK6 in the Cell Cycle and in Drug Resistance. J Mol Biol 2025:169121. [PMID: 40174666 DOI: 10.1016/j.jmb.2025.169121] [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: 01/31/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4 and CDK6) are key regulators of the G1-S phase transition in the cell cycle. In cancer cells, CDK6 overexpression often outcompetes CDK4 in driving cell cycle progression, contributing to resistance against CDK4/6 inhibitors (CDK4/6i). This suggests distinct functional and conformational differences between these two kinases, despite their striking structural and sequence similarities. Understanding the mechanisms that differentiate CDK4 and CDK6 is crucial, as resistance to CDK4/6i-frequently linked to CDK6 overexpression-remains a significant therapeutic challenge. Notably, CDK6 is often upregulated in CDK4/6i-resistant cancers and rapidly proliferating hematopoietic stem cells, underscoring its unique regulatory roles. We hypothesize that their distinct conformational dynamics explain their differences in phosphorylation of retinoblastoma protein, Rb, inhibitor efficacy, and cell cycle control. This leads us to question how their dissimilar conformational dynamics encode their distinct actions. To elucidate their differential activities, molecular mechanisms, and inhibitor binding, we combine biochemical assays and molecular dynamics (MD) simulations. We discover that CDK4 and CDK6 have distinct allosteric networks connecting the β3-αC loop and the G-loop. CDK6 exhibits stronger coupling and shorter path lengths between these regions, resulting in higher kinase activity upon cyclin binding and impacting inhibitor specificity. We also discover an unrecognized role of the unstructured CDK6 C-terminus, which allosterically connects and stabilizes the R-spine, facilitating slightly higher activity. Our findings bridge the gap between the structural similarity and functional divergence of CDK4 and CDK6, advancing the understanding of kinase regulation in cancer biology.
Collapse
Affiliation(s)
- Wengang Zhang
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Devin Bradburn
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, Bethesda, MD 20892, USA
| | - Gretchen Heidebrink
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yonglan Liu
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA; Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Ruth Nussinov
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA; Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Mardo Kõivomägi
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
8
|
Zhang W, Bradburn D, Heidebrink G, Liu Y, Jang H, Nussinov R, Kõivomägi M. Distinct allosteric networks in CDK4 and CDK6 in the cell cycle and in drug resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640857. [PMID: 40093074 PMCID: PMC11908124 DOI: 10.1101/2025.02.28.640857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4 and CDK6) are key regulators of the G1-S phase transition in the cell cycle. In cancer cells, CDK6 overexpression often outcompetes CDK4 in driving cell cycle progression, contributing to resistance against CDK4/6 inhibitors (CDK4/6i). This suggests distinct functional and conformational differences between these two kinases, despite their striking structural and sequence similarities. Understanding the mechanisms that differentiate CDK4 and CDK6 is crucial, as resistance to CDK4/6i-frequently linked to CDK6 overexpression-remains a significant therapeutic challenge. Notably, CDK6 is often upregulated in CDK4/6i-resistant cancers and rapidly proliferating hematopoietic stem cells, underscoring its unique regulatory roles. We hypothesize that their distinct conformational dynamics explain their differences in phosphorylation of retinoblastoma protein, Rb, inhibitor efficacy, and cell cycle control. This leads us to question how their dissimilar conformational dynamics encode their distinct actions. To elucidate their differential activities, molecular mechanisms, and inhibitor binding, we combine biochemical assays and molecular dynamics (MD) simulations. We discover that CDK4 and CDK6 have distinct allosteric networks connecting the β3-αC loop and the G-loop. CDK6 exhibits stronger coupling and shorter path lengths between these regions, resulting in higher kinase activity upon cyclin binding and impacting inhibitor specificity. We also discover an unrecognized role of the unstructured CDK6 C-terminus, which allosterically connects and stabilizes the R-spine, facilitating slightly higher activity. Our findings bridge the gap between the structural similarity and functional divergence of CDK4 and CDK6, advancing the understanding of kinase regulation in cancer biology.
Collapse
Affiliation(s)
- Wengang Zhang
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, U.S.A
| | - Devin Bradburn
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, Bethesda, MD 20892, U.S.A
| | - Gretchen Heidebrink
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, Bethesda, MD 20892, U.S.A
| | - Yonglan Liu
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, U.S.A
| | - Hyunbum Jang
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, U.S.A
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, U.S.A
| | - Ruth Nussinov
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, U.S.A
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, U.S.A
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Mardo Kõivomägi
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, Bethesda, MD 20892, U.S.A
| |
Collapse
|
9
|
Venetis K, Frascarelli C, Bielo LB, Cursano G, Adorisio R, Ivanova M, Mane E, Peruzzo V, Concardi A, Negrelli M, D'Ercole M, Porta FM, Zhan Y, Marra A, Trapani D, Criscitiello C, Curigliano G, Guerini-Rocco E, Fusco N. Mismatch repair (MMR) and microsatellite instability (MSI) phenotypes across solid tumors: A comprehensive cBioPortal study on prevalence and prognostic impact. Eur J Cancer 2025; 217:115233. [PMID: 39827722 DOI: 10.1016/j.ejca.2025.115233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Mismatch repair deficiency (MMR-d) and microsatellite instability (MSI) are prognostic and predictive biomarkers in oncology. Current testing for MMR/MSI relies on immunohistochemistry (IHC) for MMR proteins and molecular assays for MSI detection. This combined diagnostic strategy, however, lacks tumor specificity and does not account for gene variants. This study provides an in-depth analysis of MMR mutations frequency, spectrum, and distribution in solid tumors. Data from 23,893 patients across 11 tumor types, using 66 publicly available studies, were analyzed. MMR-mutated (MMR-m) status was defined by alterations in MLH1, PMS2, MSH2, and/or MSH6; MSI was assessed by MSIsensor. Cases with indeterminate labelling were excluded. Survival was analyzed using the Kaplan-Meier method. Among 19,353 tumors, 949 MMR variants were identified, comprising 432 pathogenic and 517 variants of unknown significance (VUS), as defined by OncoKB. MSH6 mutations were the most frequent (n = 279, 29.4 %), followed by MSH2 (n = 198, 20.9 %), MLH1 (n = 187, 19.7 %), and PMS2 (n = 161, 16.9 %). MMR-m cases were more frequent in endometrial (EC, 20.5 %), colorectal (CRC, 8.2 %), bladder (BLCA, 8.7 %), and gastroesophageal cancers (GEC, 5.4 %). Pathogenic mutations were more common than non-pathogenic in EC, CRC, and GEC (p < 0.001, p = 0.01, p = 0.32, respectively). MMR-m status was not associated with MSI in 247 (48.9 %) cases, including 67 (13.2 %) with pathogenic mutations. The highest concordance between MMR-m and MSI was observed in CRC (65.7 %), EC (91.2 %), and GEC (69.6 %), while the lowest in pancreatic (0.2 %) and lung cancers (0.1 %). MMR-m GECs showed improved overall survival compared to MMR-wt (p = 0.009), a relationship not observed in other tumor types. This study demonstrates that the MMR spectrum is extremely hetoerogeneous in solid tumors, highliting the need for comprehensive and tumor-specific testing strategies.
Collapse
Affiliation(s)
| | - Chiara Frascarelli
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Giulia Cursano
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Riccardo Adorisio
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Eltjona Mane
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Virginia Peruzzo
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Alberto Concardi
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Marianna D'Ercole
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Yinxiu Zhan
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Antonio Marra
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Dario Trapani
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
10
|
Kumarasamy V, Wang J, Roti M, Wan Y, Dommer AP, Rosenheck H, Putta S, Trub A, Bisi J, Strum J, Roberts P, Rubin SM, Frangou C, McLean K, Witkiewicz AK, Knudsen ES. Discrete vulnerability to pharmacological CDK2 inhibition is governed by heterogeneity of the cancer cell cycle. Nat Commun 2025; 16:1476. [PMID: 39924553 PMCID: PMC11808123 DOI: 10.1038/s41467-025-56674-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/27/2025] [Indexed: 02/11/2025] Open
Abstract
Cyclin dependent kinase 2 (CDK2) regulates cell cycle and is an emerging target for cancer therapy. There are relatively small numbers of tumor models that exhibit strong dependence on CDK2 and undergo G1 cell cycle arrest following CDK2 inhibition. The expression of P16INK4A and cyclin E1 determines this sensitivity to CDK2 inhibition. The co-expression of these genes occurs in breast cancer patients highlighting their clinical significance as predictive biomarkers for CDK2-targeted therapies. In cancer models that are genetically independent of CDK2, pharmacological inhibitors suppress cell proliferation by inducing 4N cell cycle arrest and increasing the expressions of phospho-CDK1 (Y15) and cyclin B1. CRISPR screens identify CDK2 loss as a mediator of resistance to a CDK2 inhibitor, INX-315. Furthermore, CDK2 deletion reverses the G2/M block induced by CDK2 inhibitors and restores cell proliferation. Complementary drug screens define multiple means to cooperate with CDK2 inhibition beyond G1/S. These include the depletion of mitotic regulators as well as CDK4/6 inhibitors cooperate with CDK2 inhibition in multiple phases of the cell cycle. Overall, this study underscores two fundamentally distinct features of response to CDK2 inhibitors that are conditioned by tumor context and could serve as the basis for differential therapeutic strategies in a wide range of cancers.
Collapse
Affiliation(s)
- Vishnu Kumarasamy
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jianxin Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Michelle Roti
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Yin Wan
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Adam P Dommer
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Hanna Rosenheck
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sivasankar Putta
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | | | | | | | | | - Seth M Rubin
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Costakis Frangou
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Karen McLean
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Agnieszka K Witkiewicz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| | - Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
11
|
Shanabag A, Armand J, Son E, Yang HW. Targeting CDK4/6 in breast cancer. Exp Mol Med 2025; 57:312-322. [PMID: 39930131 PMCID: PMC11873051 DOI: 10.1038/s12276-025-01395-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 03/04/2025] Open
Abstract
Dysregulation of the cell cycle machinery, particularly the overactivation of cyclin-dependent kinases 4 and 6 (CDK4/6), is a hallmark of breast cancer pathogenesis. The introduction of CDK4/6 inhibitors has transformed the treatment landscape for hormone receptor-positive breast cancer by effectively targeting abnormal cell cycle progression. However, despite their initial clinical success, drug resistance remains a significant challenge, with no reliable biomarkers available to predict treatment response or guide strategies for managing resistant populations. Consequently, numerous studies have sought to investigate the mechanisms driving resistance to optimize the therapeutic use of CDK4/6 inhibitors and improve patient outcomes. Here we examine the molecular mechanisms regulating the cell cycle, current clinical applications of CDK4/6 inhibitors in breast cancer, and key mechanisms contributing to drug resistance. Furthermore, we discuss emerging predictive biomarkers and highlight potential directions for overcoming resistance and enhancing therapeutic efficacy.
Collapse
Affiliation(s)
- Anusha Shanabag
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Jessica Armand
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Eugene Son
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Hee Won Yang
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.
| |
Collapse
|
12
|
Foffano L, Cucciniello L, Nicolò E, Migliaccio I, Noto C, Reduzzi C, Malorni L, Cristofanilli M, Gerratana L, Puglisi F. Cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i): Mechanisms of resistance and where to find them. Breast 2025; 79:103863. [PMID: 39718288 PMCID: PMC11872392 DOI: 10.1016/j.breast.2024.103863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024] Open
Abstract
CDK4/6 inhibitors (CDK4/6i) have significantly impacted on the treatment of HR + HER2 negative (HER2-) metastatic breast cancer (BC) when combined with endocrine therapy. Nonetheless, despite significant research efforts, the mechanisms of de novo and acquired resistance to CDK4/6i have not yet been fully elucidated, highlighting the need for a deeper understanding of these process. Additionally, the importance of dissecting CDK4/6i resistance from endocrine resistance for personalized treatment is increasingly recognized. Liquid biopsy has emerged as a minimally invasive tool for identifying circulating biomarkers of resistance through the integration of multiparametric and dynamic assessments that encompass ctDNA, CTCs, exosomes, and epigenetic ctDNA alterations, representing a promising perspective for the clinical characterization of treatment resistance and guiding post-progression strategies to improve patient outcomes. Aim of this review is summarize potential mechanisms of CDK4/6i resistance, along with the advantages of using liquid biopsy to identify resistance biomarkers in HR+/HER2- MBC patients treated with CDK 4/6 inhibitors.
Collapse
Affiliation(s)
- L Foffano
- Department of Medical Oncology. CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy; Department of Medicine, University of Udine, Udine, Italy
| | - L Cucciniello
- Department of Medical Oncology. CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy; Department of Medicine, University of Udine, Udine, Italy
| | - E Nicolò
- Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
| | - I Migliaccio
- "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, 59100, Prato, Italy
| | - C Noto
- Department of Medical Oncology. CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy; Department of Medicine, University of Udine, Udine, Italy
| | - C Reduzzi
- Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - L Malorni
- "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, 59100, Prato, Italy; "Sandro Pitigliani" Medical Oncology Department, Hospital of Prato, 59100, Prato, Italy
| | - M Cristofanilli
- Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - L Gerratana
- Department of Medical Oncology. CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy.
| | - F Puglisi
- Department of Medical Oncology. CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy; Department of Medicine, University of Udine, Udine, Italy
| |
Collapse
|
13
|
Würth R, Donato E, Michel LL, Saini M, Becker L, Cheytan T, Doncevic D, Messmer T, Gutjahr E, Weber R, Klein C, Alborzinia H, Yildiz U, Vogel V, Hlevnjak M, Kozyulina P, Neuberth SJ, Schwerd-Kleine P, Jakab S, Pfarr N, Angeles AK, Laut AK, Karpova D, Falcone M, Hardt O, Theek B, Wagner CV, Becker M, Wagner S, Haselmayr M, Schmitt A, Müller-Tidow C, Riethdorf S, Pantel K, Zapatka M, Sültmann H, Herrmann C, Thewes V, Lichter P, Schneeweiss A, Sprick MR, Trumpp A. Circulating tumor cell plasticity determines breast cancer therapy resistance via neuregulin 1-HER3 signaling. NATURE CANCER 2025; 6:67-85. [PMID: 39753722 PMCID: PMC11779641 DOI: 10.1038/s43018-024-00882-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/18/2024] [Indexed: 01/31/2025]
Abstract
Circulating tumor cells (CTCs) drive metastasis, the leading cause of death in individuals with breast cancer. Due to their low abundance in the circulation, robust CTC expansion protocols are urgently needed to effectively study disease progression and therapy responses. Here we present the establishment of long-term CTC-derived organoids from female individuals with metastatic breast cancer. Multiomics analysis of CTC-derived organoids along with preclinical modeling with xenografts identified neuregulin 1 (NRG1)-ERBB2 receptor tyrosine kinase 3 (ERBB3/HER3) signaling as a key pathway required for CTC survival, growth and dissemination. Genome-wide CRISPR activation screens revealed that fibroblast growth factor receptor 1 (FGFR1) signaling serves a compensatory function to the NRG1-HER3 axis and rescues NRG1 deficiency in CTCs. Conversely, NRG1-HER3 activation induced resistance to FGFR1 inhibition, whereas combinatorial blockade impaired CTC growth. The dynamic interplay between NRG1-HER3 and FGFR1 signaling reveals the molecular basis of cancer cell plasticity and clinically relevant strategies to target it. Our CTC organoid platform enables the identification and validation of patient-specific vulnerabilities and represents an innovative tool for precision medicine.
Collapse
MESH Headings
- Humans
- Neuregulin-1/metabolism
- Neuregulin-1/genetics
- Receptor, ErbB-3/metabolism
- Receptor, ErbB-3/genetics
- Female
- Breast Neoplasms/pathology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/genetics
- Signal Transduction
- Animals
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- Neoplastic Cells, Circulating/drug effects
- Drug Resistance, Neoplasm
- Mice
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Organoids/metabolism
- Organoids/pathology
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Roberto Würth
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany.
| | - Elisa Donato
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Laura L Michel
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Massimo Saini
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Lisa Becker
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Tasneem Cheytan
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Daria Doncevic
- Department of Bioinformatics, Institute of Pharmacy and Molecular Biotechnology & BioQuant, Heidelberg University, Heidelberg, Germany
| | - Tobias Messmer
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Ewgenija Gutjahr
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Rebecca Weber
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Corinna Klein
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Hamed Alborzinia
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Umut Yildiz
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Vanessa Vogel
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Mario Hlevnjak
- Computational Oncology, Molecular Precision Oncology Program, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Polina Kozyulina
- Computational Oncology, Molecular Precision Oncology Program, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Sarah-Jane Neuberth
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Paul Schwerd-Kleine
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Sevinç Jakab
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Nicole Pfarr
- Institute of Pathology, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Arlou Kristina Angeles
- Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
| | - Astrid K Laut
- Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
| | - Darja Karpova
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Mattia Falcone
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Olaf Hardt
- Miltenyi Biotec, Bergisch Gladbach, Germany
| | | | - Celina V Wagner
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mirjam Becker
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Wagner
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Haselmayr
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Sabine Riethdorf
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Zapatka
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Carl Herrmann
- Department of Bioinformatics, Institute of Pharmacy and Molecular Biotechnology & BioQuant, Heidelberg University, Heidelberg, Germany
| | - Verena Thewes
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University, Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University, Heidelberg, Germany
| | - Andreas Schneeweiss
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin R Sprick
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
14
|
Asciolla JJ, Wu X, Adamopoulos C, Gavathiotis E, Poulikakos PI. Resistance mechanisms and therapeutic strategies of CDK4 and CDK6 kinase targeting in cancer. NATURE CANCER 2025; 6:24-40. [PMID: 39885369 DOI: 10.1038/s43018-024-00893-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/09/2024] [Indexed: 02/01/2025]
Abstract
Cyclin-dependent kinases (CDKs) 4 and 6 (CDK4/6) are important regulators of the cell cycle. Selective CDK4/6 small-molecule inhibitors have shown clinical activity in hormonal receptor-positive (HR+) metastatic breast cancer, but their effectiveness remains limited in other cancer types. CDK4/6 degradation and improved selectivity across CDK paralogs are approaches that could expand the effectiveness of CDK4/6 targeting. Recent studies also suggest the use of CDK4/6-targeting agents in cancer immunotherapy. In this Review, we highlight recent advancements in the mechanistic understanding and development of pharmacological approaches targeting CDK4/6. Collectively, these developments pose new challenges and opportunities for rationally designing more effective treatments.
Collapse
Affiliation(s)
- James J Asciolla
- Department of Oncological Sciences, Precision Immunology Institute, the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xuewei Wu
- Department of Oncological Sciences, Precision Immunology Institute, the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- China Innovation Center of Roche, Shanghai, China
| | - Christos Adamopoulos
- Department of Oncological Sciences, Precision Immunology Institute, the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evripidis Gavathiotis
- Department of Biochemistry, Department of Medicine, Department of Oncology, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, New York, NY, USA
| | - Poulikos I Poulikakos
- Department of Oncological Sciences, Precision Immunology Institute, the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
15
|
Takeuchi R, Kuwahara N, Amino Y, Hayashi S, Taguchi C, Suzuki I, Suzuki H, Nagashima T, Arikawa K, Okada Y, Nomoto T, Hiratsuka K. Cyclosporine A Causes Gingival Overgrowth by Promoting Entry into the S Phase at the G1/S Cell Cycle Checkpoint in Gingival Fibroblasts Exposed to Lipopolysaccharide. Diseases 2024; 12:322. [PMID: 39727652 PMCID: PMC11727098 DOI: 10.3390/diseases12120322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVES Cyclosporine A promotes gingival fibrosis by enhancing the proliferation of gingival fibroblasts, leading to gingival overgrowth. The population of gingival fibroblasts is regulated by cell cycle machinery, which balances cell growth and inhibition. Cells that detect DNA damage pause at the G1/S checkpoint to repair the damage instead of progressing to the S phase. Previous studies have linked drug-induced gingival overgrowth to the response of fibroblasts to lipopolysaccharide (LPS) and cyclosporine A. This research investigates the effects of cyclosporine A on the G1/S checkpoint and its mediators in LPS-treated gingival fibroblasts to clarify the mechanisms behind cyclosporine-A-induced gingival overgrowth. METHODS Semi-confluent human gingival fibroblasts were treated with LPS or cyclosporine A in DMEM. Cell proliferation was evaluated by counting the total number of cells. The distribution of the cell cycle phases was analyzed using flow cytometry. Additionally, the expression levels of mRNAs and proteins related to cell cycle regulators were quantified by reverse-transcription quantitative PCR and Western blotting, respectively. RESULTS Cyclosporine A treatment significantly enhanced cell proliferation and the G1-S cell cycle transition. It increased the mRNA levels of CDC25A and CYCLIN D while decreasing those of RB1, SMAD3, and SMAD4. Additionally, it upregulated the protein levels of CDC25A, CYCLIN D, CDK4, CDK6, and pRB and downregulated the protein levels of SMAD3 and SMAD4. CONCLUSIONS Gingival overgrowth induced by cyclosporine A could be attributed to these alterations.
Collapse
Affiliation(s)
- Reiri Takeuchi
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (N.K.); (K.H.)
| | - Noriko Kuwahara
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (N.K.); (K.H.)
| | - Yuta Amino
- Department of Oral Implantology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan;
| | - Sachiyo Hayashi
- Department of Special Needs Dentistry, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (S.H.); (T.N.)
| | - Chieko Taguchi
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (C.T.); (I.S.); (H.S.); (K.A.)
| | - Itaru Suzuki
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (C.T.); (I.S.); (H.S.); (K.A.)
| | - Haruka Suzuki
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (C.T.); (I.S.); (H.S.); (K.A.)
| | - Teruaki Nagashima
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (C.T.); (I.S.); (H.S.); (K.A.)
- Department of Community Oral Health, Nihon University Graduate School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan
| | - Kazumune Arikawa
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (C.T.); (I.S.); (H.S.); (K.A.)
| | - Yuichiro Okada
- Department of Histology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan
| | - Takato Nomoto
- Department of Special Needs Dentistry, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (S.H.); (T.N.)
| | - Koichi Hiratsuka
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (N.K.); (K.H.)
| |
Collapse
|
16
|
Armand J, Kim S, Kim K, Son E, Kim M, Yang HW. Therapeutic benefits of maintaining CDK4/6 inhibitors and incorporating CDK2 inhibitors beyond progression in breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.623139. [PMID: 39605351 PMCID: PMC11601343 DOI: 10.1101/2024.11.11.623139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The combination of CDK4/6 inhibitors (CDK4/6i) and endocrine therapy has revolutionized treatment for hormone receptor-positive (HR+) metastatic breast cancer. However, the emergence of resistance in most patients often leads to treatment discontinuation with no consensus on effective second-line therapies. The therapeutic benefits of maintaining CDK4/6i or incorporating CDK2 inhibitors (CDK2i) after disease progression remain unclear. Here, we demonstrate that sustained CDK4/6i therapy, either alone or combined with CDK2i, significantly suppresses the growth of drug-resistant HR+ breast cancer. Continued CDK4/6i treatment induces a non-canonical pathway for retinoblastoma protein (Rb) inactivation via post-translational degradation, resulting in diminished E2F activity and delayed G1 progression. Importantly, our data highlight that CDK2i should be combined with CDK4/6i to effectively suppress CDK2 activity and overcome resistance. We also identify cyclin E overexpression as a key driver of resistance to CDK4/6 and CDK2 inhibition. These findings provide crucial insights into overcoming resistance in HR+ breast cancer, supporting the continued use of CDK4/6i and the strategic incorporation of CDK2i to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Jessica Armand
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sungsoo Kim
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Kibum Kim
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Eugene Son
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Minah Kim
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Hee Won Yang
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| |
Collapse
|
17
|
Kudo R, Safonov A, Jones C, Moiso E, Dry JR, Shao H, Nag S, da Silva EM, Yildirim SY, Li Q, O'Connell E, Patel P, Will M, Fushimi A, Benitez M, Bradic M, Fan L, Nakshatri H, Sudhan DR, Denz CR, Huerga Sanchez I, Reis-Filho JS, Goel S, Koff A, Weigelt B, Khan QJ, Razavi P, Chandarlapaty S. Long-term breast cancer response to CDK4/6 inhibition defined by TP53-mediated geroconversion. Cancer Cell 2024; 42:1919-1935.e9. [PMID: 39393354 DOI: 10.1016/j.ccell.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 07/02/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
Inhibition of CDK4/6 kinases has led to improved outcomes in breast cancer. Nevertheless, only a minority of patients experience long-term disease control. Using a large, clinically annotated cohort of patients with metastatic hormone receptor-positive (HR+) breast cancer, we identify TP53 loss (27.6%) and MDM2 amplification (6.4%) to be associated with lack of long-term disease control. Human breast cancer models reveal that p53 loss does not alter CDK4/6 activity or G1 blockade but instead promotes drug-insensitive p130 phosphorylation by CDK2. The persistence of phospho-p130 prevents DREAM complex assembly, enabling cell-cycle re-entry and tumor progression. Inhibitors of CDK2 can overcome p53 loss, leading to geroconversion and manifestation of senescence phenotypes. Complete inhibition of both CDK4/6 and CDK2 kinases appears to be necessary to facilitate long-term response across genomically diverse HR+ breast cancers.
Collapse
Affiliation(s)
- Rei Kudo
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Department of Surgery, The Jikei University School of Medicine, Tokyo 1058461, Japan
| | - Anton Safonov
- Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Catherine Jones
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Enrico Moiso
- Department of Medicine, MSK, New York, NY 10065, USA; Department of Epidemiology and Biostatistics, MSK, New York, NY 10065, USA
| | | | - Hong Shao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Sharanya Nag
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Edaise M da Silva
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Selma Yeni Yildirim
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Qing Li
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Elizabeth O'Connell
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Payal Patel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Marie Will
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Clinical Genetics Service, Department of Medicine, MSK, New York, NY 10065, USA
| | - Atsushi Fushimi
- Department of Surgery, The Jikei University School of Medicine, Tokyo 1058461, Japan
| | - Marimar Benitez
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Martina Bradic
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Shom Goel
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Andrew Koff
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Qamar J Khan
- Division of Medical Oncology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Pedram Razavi
- Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
18
|
Prasad D, Baldelli E, Blais EM, Davis J, El Gazzah E, Mueller C, Gomeiz A, Ibrahim A, Newrekar AV, Corgiat BA, Dunetz R, Petricoin Iii EF, Wei Q, Pierobon M. Functional activation of the AKT-mTOR signalling axis in a real-world metastatic breast cancer cohort. Br J Cancer 2024; 131:1543-1554. [PMID: 39322687 PMCID: PMC11519601 DOI: 10.1038/s41416-024-02852-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Mutations of the PIK3CA/AKT/mTOR axis are common events in metastatic breast cancers (MBCs). This study was designed to evaluate the extent to which genetic alterations of the PIK3CA/AKT/mTOR can predict protein activation of this signalling axis in MBCs. METHODS Molecular profiles were generated by CLIA-certified laboratories from a real-world evidence cohort of 171 MBC patients. Genetic alterations of the PIK3CA pathway were measured using next-generation sequencing. Activation levels of AKT and downstream signalling molecules were quantified using two orthogonal proteomic methods. Protein activity was correlated with underlying genomic profiles and response to CDK4/6 inhibition in combination with endocrine treatment (ET). RESULTS Oncogenic alterations of the PIK3CA/AKT/PTEN pathway were identified in 49.7% of cases. Genomic profiles emerged as poor predictors of protein activity (AUC:0.69), and AKT phosphorylation levels mimicked those of mutant lesions in 76.9% of wild-type tumours. High phosphorylation levels of the PI3K/AKT/mTOR downstream target p70S6 Kinase (T389) were associated with shorter PFS in patients treated with CDK4/6 inhibitors in combination with ET (HR:4.18 95%CI:1.19-14.63); this association was not seen when patients were classified by mutational status. CONCLUSIONS Phosphoprotein-based measurements of drug targets and downstream substrates should be captured along with genomic information to identify MBCs driven by the PI3K/AKT/mTOR signalling.
Collapse
Affiliation(s)
- Deepika Prasad
- School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Elisa Baldelli
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | | | | | - Emna El Gazzah
- School of Systems Biology, George Mason University, Manassas, VA, USA
| | | | - Alison Gomeiz
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Aisha Ibrahim
- School of Systems Biology, George Mason University, Manassas, VA, USA
| | | | | | | | - Emanuel F Petricoin Iii
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Qi Wei
- Department of Bioengineering, George Mason University, Fairfax, VA, USA
| | - Mariaelena Pierobon
- School of Systems Biology, George Mason University, Manassas, VA, USA.
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA.
| |
Collapse
|
19
|
Lloyd MR, Jhaveri K, Kalinsky K, Bardia A, Wander SA. Precision therapeutics and emerging strategies for HR-positive metastatic breast cancer. Nat Rev Clin Oncol 2024; 21:743-761. [PMID: 39179659 DOI: 10.1038/s41571-024-00935-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
Anti-oestrogen-based therapies, often combined with a CDK4/6 inhibitor, are the current standard-of-care first-line therapy for patients with advanced-stage hormone receptor-positive (HR+) breast cancer. Resistance to anti-oestrogen agents inevitably occurs, mediated by oestrogen receptor (ER)-dependent or ER-independent mechanisms that drive tumour progression. Emerging endocrine therapies include, but are not limited to, next-generation oral ER degraders and proteolysis targeting chimeras, which might be particularly effective in patients with ESR1-mutant breast cancer. Furthermore, cancers harbouring driver alterations in oncogenic signalling pathways, including AKT and PI3K, might be susceptible to novel combination strategies involving targeted inhibitors. Next-generation CDK2/4 inhibitors are an area of active clinical investigation, and efforts are ongoing to evaluate the role of sequential CDK inhibition. Approved and emerging antibody-drug conjugates exploiting novel target antigens have also demonstrated promising clinical activity. These novel agents, as well as further identification and characterization of predictive biomarkers, will hopefully continue to improve clinical outcomes, reduce the incidence of toxicities, and limit the extent of overtreatment in this population. In this Review, we describe the evolving treatment paradigm for patients with metastatic HR+ breast cancer in light of the growing armamentarium of drugs and biomarkers that will help to shape the future therapeutic landscape. These strategies are expected to involve tumour molecular profiling to enable the delivery of precision medicine.
Collapse
Affiliation(s)
| | - Komal Jhaveri
- Memorial Sloan Kettering Evelyn H. Lauder Breast Center, New York, NY, USA
| | - Kevin Kalinsky
- Winship Cancer Institute at Emory University, Atlanta, GA, USA
| | - Aditya Bardia
- UCLA Health Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Seth A Wander
- Massachusetts General Hospital Cancer Center, Boston, MA, USA.
| |
Collapse
|
20
|
Chen A, Kim BJ, Mitra A, Vollert CT, Lei JT, Fandino D, Anurag M, Holt MV, Gou X, Pilcher JB, Goetz MP, Northfelt DW, Hilsenbeck SG, Marshall CG, Hyer ML, Papp R, Yin SY, De Angelis C, Schiff R, Fuqua SAW, Ma CX, Foulds CE, Ellis MJ. PKMYT1 Is a Marker of Treatment Response and a Therapeutic Target for CDK4/6 Inhibitor-Resistance in ER+ Breast Cancer. Mol Cancer Ther 2024; 23:1494-1510. [PMID: 38781103 PMCID: PMC11443213 DOI: 10.1158/1535-7163.mct-23-0564] [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: 08/26/2023] [Revised: 03/25/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Endocrine therapies (ET) with cyclin-dependent kinase 4/6 (CDK4/6) inhibition are the standard treatment for estrogen receptor-α-positive (ER+) breast cancer, however drug resistance is common. In this study, proteogenomic analyses of patient-derived xenografts (PDXs) from patients with 22 ER+ breast cancer demonstrated that protein kinase, membrane-associated tyrosine/threonine one (PKMYT1), a WEE1 homolog, is estradiol (E2) regulated in E2-dependent PDXs and constitutively expressed when growth is E2-independent. In clinical samples, high PKMYT1 mRNA levels associated with resistance to both ET and CDK4/6 inhibition. The PKMYT1 inhibitor lunresertib (RP-6306) with gemcitabine selectively and synergistically reduced the viability of ET and palbociclib-resistant ER+ breast cancer cells without functional p53. In vitro the combination increased DNA damage and apoptosis. In palbociclib-resistant, TP53 mutant PDX-derived organoids and PDXs, RP-6306 with low-dose gemcitabine induced greater tumor volume reduction compared to treatment with either single agent. Our study demonstrates the clinical potential of RP-6306 in combination with gemcitabine for ET and CDK4/6 inhibitor resistant TP53 mutant ER+ breast cancer.
Collapse
Affiliation(s)
- Anran Chen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, Texas
- Repare Therapeutics, Cambridge, Massachusetts
| | - Beom-Jun Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Aparna Mitra
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Craig T Vollert
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Employee of Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas
| | - Diana Fandino
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Meenakshi Anurag
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Matthew V Holt
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Xuxu Gou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Jacob B Pilcher
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | | | - Donald W Northfelt
- Division of Hematology and Medical Oncology at Mayo Clinic, Phoenix, Arizona
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | | | - Marc L Hyer
- Repare Therapeutics, Cambridge, Massachusetts
| | - Robert Papp
- Repare Therapeutics, Saint-Laurent, Quebec, Canada
| | - Shou-Yun Yin
- Repare Therapeutics, Saint-Laurent, Quebec, Canada
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Suzanne A W Fuqua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Cynthia X Ma
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Charles E Foulds
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
21
|
Phillips AH, Kriwacki RW. The role of intrinsic protein disorder in regulation of cyclin-dependent kinases. Curr Opin Struct Biol 2024; 88:102906. [PMID: 39142260 DOI: 10.1016/j.sbi.2024.102906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024]
Abstract
While the structure/function paradigm for folded domains was established decades ago, our understanding of how intrinsically disordered regions (IDRs) contribute to biological function is still evolving. IDRs exist as conformational ensembles that can range from highly compact to highly extended depending on their sequence composition. IDR sequences are less conserved than those of folded domains, but often display short, conserved segments termed short linear motifs (SLiMs), that often mediate protein-protein interactions and are often regulated by posttranslational modifications, giving rise to complex functionality when multiple, differently regulated SLiMs are combined. This combinatorial functionality was associated with signaling and regulation soon after IDRs were first recognized as functional elements within proteins. Here, we discuss roles for disorder in proteins that regulate cyclin-dependent kinases, the master timekeepers of the eukaryotic cell cycle. We illustrate the importance of intrinsic flexibility in the transmission of regulatory signals by these entirely disordered proteins.
Collapse
Affiliation(s)
- Aaron H Phillips
- Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Richard W Kriwacki
- Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
22
|
Lee JY, Lee JW, Chung MS, Choi JG, Sim SH, Kim HJ, Kim JE, Lee KE, Park YH, Kang MJ, Ahn MS, Chae YS, Park JH, Kim JH, Kim GM, Byun JH, Park KU, Kim JW, Jung SP, Lee JH, An JS, Jang B, Yoon D, Kim J, Hong J, Koo H, Cho KR, Kim CY, Sa JK, Park KH. Age- and ethnic-driven molecular and clinical disparity of East Asian breast cancers. BMC Med 2024; 22:422. [PMID: 39334392 PMCID: PMC11438198 DOI: 10.1186/s12916-024-03638-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is a complex disease with profound genomic aberrations. However, the underlying molecular disparity influenced by age and ethnicity remains elusive. METHODS In this study, we aimed to investigate the molecular properties of 843 primary and metastatic BC patients enrolled in the K-MASTER program. By categorizing patients into two distinct age subgroups, we explored their unique molecular properties. Additionally, we leveraged large-scale genomic data from the TCGA and MSK-IMPACT studies to examine the ethnic-driven molecular and clinical disparities. RESULTS We observed a high prevalence of PI3KCA mutations in K-MASTER HER2 + tumors, particularly in older patients. Moreover, we identified increased mutation rates in DNA damage response molecules, including ARID1A, MSH6, and MLH1. The K-MASTER patients were mainly comprised of triple-negative breast cancer (TNBC) and HER2-positive tumors, while the TCGA and MSK-IMPACT cohorts exhibited a predominance of hormone receptor-positive (HR +) subtype tumors. Importantly, GATA3 mutations were less frequently observed in East Asian patients, which correlated with poor clinical outcomes. In addition to characterizing the molecular disparities, we developed a gradient-boosting multivariable model to identify a new molecular signature that could predict the therapeutic response to platinum-based chemotherapy. CONCLUSIONS Our findings collectively provide unprecedented insights into the significance of age and ethnicity on the molecular and clinical characteristics of BC patients.
Collapse
Affiliation(s)
- Ji Yoon Lee
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ji Won Lee
- Department of Internal Medicine, Division of Medical Oncology, Korea University Anam Hospital, Seoul, Korea
| | - Min Sung Chung
- Department of Surgery, College of Medicine, Hanyang University, Seoul, 04763, Korea
| | - Jong Gwon Choi
- Department of Oncology-Hematology, Konyang University Hospital, Daejeon, Korea
| | - Sung Hoon Sim
- Center for Breast Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Hyo Jeong Kim
- Department of Internal Medicine, Division of Hematology-Oncology, School of Medicine, Medical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Jeong Eun Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyoung Eun Lee
- Department of Hematology and Oncology, Ewha Womans University Hospital, Seoul, 07985, Republic of Korea
| | - Yeon Hee Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Myoung Joo Kang
- Department of Internal Medicine, Inje University Haeundae Paik Hospital, Busan, Korea
| | - Mi Sun Ahn
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Yee Soo Chae
- Department of Hematology and Oncology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Ji Hyun Park
- Department of Hematology-Oncology, Division of Internal Medicine, KonKuk University Medical Center, Seoul, Republic of Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Bundang Hospital, 166 Gumi-Ro, Bundang-Gu, Seongnam, 463-707, Korea
| | - Gun Min Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, Korea
| | - Jae Ho Byun
- Department of Internal Medicine, Division of Oncology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Keon Uk Park
- Department of Internal Medicine, Division of Hematology-Oncology, Keimyung University Dongsan Hospital, Keimyung University College of Medicine, Daegu, Republic of Korea
| | - Ju Won Kim
- Department of Internal Medicine, Division of Medical Oncology, Korea University Anam Hospital, Seoul, Korea
| | - Seung Pil Jung
- Department of Surgery, Department of Breast Surgery, Division of Breast and Endocrine, Korea University Anam Hospital, Seoul, Korea
| | - Jung Hyun Lee
- Department of Pathology, Korea University Anam Hospital, Seoul, Korea
| | - Jung Seok An
- Department of Pathology, Korea University Anam Hospital, Seoul, Korea
| | - Byunghyun Jang
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Dayoung Yoon
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Jiwon Kim
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jisoo Hong
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Harim Koo
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, South Korea
| | - Kyu Ran Cho
- Department of Radiology, Korea University Anam Hospital, Seoul, Korea
| | - Cheol Yong Kim
- Department of Radiology, Korea University Anam Hospital, Seoul, Korea
| | - Jason K Sa
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Republic of Korea.
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.
| | - Kyong Hwa Park
- Department of Internal Medicine, Division of Medical Oncology, Korea University Anam Hospital, Seoul, Korea.
| |
Collapse
|
23
|
Wu F, Li N, Wu X, Chen M, Huang W, Chen X, Hong Y, Wang L, Chen K, Lin L, You M, Liu J. EZH2 mutation is associated with the development of visceral metastasis by enhancing proliferation and invasion and inhibiting apoptosis in breast cancer cells. BMC Cancer 2024; 24:1166. [PMID: 39300407 DOI: 10.1186/s12885-024-12950-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The prognosis of breast cancer patients with visceral metastasis (VM) is significantly worse than that of patients without VM. We aimed to evaluate EZH2 (enhancer of zeste homolog 2) mutation as a biomarker associated with VM. METHODS Data from forty-nine patients with metastatic breast cancer (MBC) pathologically confirmed at our hospital between March 2016 and September 2018 were collected. Metastatic tissue samples were obtained via ultrasound-guided needle biopsy, and paired peripheral blood samples were also collected. Tissue and blood samples were subjected to targeted next-generation sequencing via a 247-gene panel. Stably transfected MDA-MB-231 cells expressing wild-type EZH2 (EZH2WT) or a mutant form of EZH2 (EZH2K515R) were generated. Cell proliferation, colony formation ability, migration and invasion abilities and apoptosis were assessed using CCK-8 assays, plate colony formation assays, Transwell chamber assays and flow cytometry. RESULTS The incidence of EZH2 mutations in the VM subgroup was greater than that in the non-VM subgroup in the entire cohort (n = 49, 42.3% vs. 13.0%, p = 0.024) and in the triple-negative breast cancer (TNBC) subgroup (n = 20, 50.0% vs. 10.0%, p = 0.05). Patients carrying EZH2 mutations had a significantly greater risk of developing VM than did those in the non-EZH2 mutation group in the entire cohort (HR 2.9) and in the TNBC subgroup (HR 6.45). Multivariate analysis revealed that EZH2 mutation was an independent prognostic factor for VM (HR 2.99, p = 0.009) in the entire cohort and in the TNBC subgroup (HR 10.1, p = 0.006). Data from cBioPortal also showed that patients with EZH2 mutations had a significantly greater risk of developing VM (HR 3.1), and the time to develop VM was significantly earlier in the EZH2 mutation group (31.5 months vs. 109.7 months, p = 0.008). Multivariate analysis revealed that EZH2 mutation (HR 2.73, p = 0.026) was an independent factor for VM after breast cancer surgery. There was no correlation between EZH2 mutations and BRCA1/2 mutations. Most of the patients (81.8%) in our cohort who developed VM carried the "c.1544A > G (p.K515R)" mutation. Compared with EZH2WT MDA-MB-231 cells, EZH2K515R MDA-MB-231 cells had greater colony formation rates (p < 0.01), greater migration and invasion rates (p < 0.001), and lower apoptosis rates (p < 0.01). The proportion of S + G2/M phase cells in the EZH2K515R group was significantly greater than that in the EZH2WT group. CONCLUSIONS EZH2 mutation is associated with VM development in breast cancer patients. The EZH2K515R mutation leads to VM and a poor prognosis by enhancing proliferation and invasion and inhibiting apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Fan Wu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, China
| | - Nani Li
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Xiufeng Wu
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Mulan Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Weiwei Huang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, China
| | - Xinhua Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Yi Hong
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Lili Wang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Kan Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Lin Lin
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Minjin You
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Jian Liu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China.
| |
Collapse
|
24
|
Glaviano A, Wander SA, Baird RD, Yap KCH, Lam HY, Toi M, Carbone D, Geoerger B, Serra V, Jones RH, Ngeow J, Toska E, Stebbing J, Crasta K, Finn RS, Diana P, Vuina K, de Bruin RAM, Surana U, Bardia A, Kumar AP. Mechanisms of sensitivity and resistance to CDK4/CDK6 inhibitors in hormone receptor-positive breast cancer treatment. Drug Resist Updat 2024; 76:101103. [PMID: 38943828 DOI: 10.1016/j.drup.2024.101103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/17/2024] [Accepted: 06/10/2024] [Indexed: 07/01/2024]
Abstract
Cell cycle dysregulation is a hallmark of cancer that promotes eccessive cell division. Cyclin-dependent kinase 4 (CDK4) and cyclin-dependent kinase 6 (CDK6) are key molecules in the G1-to-S phase cell cycle transition and are crucial for the onset, survival, and progression of breast cancer (BC). Small-molecule CDK4/CDK6 inhibitors (CDK4/6i) block phosphorylation of tumor suppressor Rb and thus restrain susceptible BC cells in G1 phase. Three CDK4/6i are approved for the first-line treatment of patients with advanced/metastatic hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) BC in combination with endocrine therapy (ET). Though this has improved the clinical outcomes for survival of BC patients, there is no established standard next-line treatment to tackle drug resistance. Recent studies suggest that CDK4/6i can modulate other distinct effects in both BC and breast stromal compartments, which may provide new insights into aspects of their clinical activity. This review describes the biochemistry of the CDK4/6-Rb-E2F pathway in HR+ BC, then discusses how CDK4/6i can trigger other effects in BC/breast stromal compartments, and finally outlines the mechanisms of CDK4/6i resistance that have emerged in recent preclinical studies and clinical cohorts, emphasizing the impact of these findings on novel therapeutic opportunities in BC.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo 90123, Italy
| | - Seth A Wander
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Richard D Baird
- Cancer Research UK Cambridge Centre, Hills Road, Cambridge CB2 0QQ, UK
| | - Kenneth C-H Yap
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Masakazu Toi
- School of Medicine, Kyoto University, Kyoto, Japan
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo 90123, Italy
| | - Birgit Geoerger
- Gustave Roussy Cancer Center, Department of Pediatric and Adolescent Oncology, Inserm U1015, Université Paris-Saclay, Villejuif, France
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Robert H Jones
- Cardiff University and Velindre Cancer Centre, Museum Avenue, Cardiff CF10 3AX, UK
| | - Joanne Ngeow
- Lee Kong Chian School of Medicine (LKCMedicine), Nanyang Technological University, Experimental Medicine Building, 636921, Singapore; Cancer Genetics Service (CGS), National Cancer Centre Singapore, 168583, Singapore
| | - Eneda Toska
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Justin Stebbing
- School of Life Sciences, Anglia Ruskin University, Cambridge, UK; Division of Cancer, Imperial College London, Hammersmith Campus, London, UK
| | - Karen Crasta
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593, Singapore; Healthy Longetivity Translational Program, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Richard S Finn
- Department of Oncology, Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo 90123, Italy
| | - Karla Vuina
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Robertus A M de Bruin
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Uttam Surana
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; SiNOPSEE Therapeutics Pte Ltd, A⁎STARTCentral, 139955, Singapore
| | - Aditya Bardia
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore.
| |
Collapse
|
25
|
Lv JL, Ren YS, Tan YJ, Chu T, Cao XY, Liu HY, Ma R, Zhang H, Zheng QS, Dong GC, Li J. Hernandezine acts as a CDK4 suppressor inhibiting tumor growth by the CDK4/PKM2/NRF2 axis in colon cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155775. [PMID: 38838401 DOI: 10.1016/j.phymed.2024.155775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/11/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND The cyclin-dependent kinase 4 (CDK4) interacts with its canonical and non-canonical substrates modulating the cell cycle in tumor cells. However, the potential substrates and the beyond-cell-cycle-regulated functions of CDK4 in colon cancer (CC) are still unknown. Hernandezine (HER) is previously verified to induce G0/G1 phase arrest and autophagic cell death in human cancer cells, which implies that HER might target G0/G1 phase-related proteins, including CDK4. PURPOSE The present study tried to investigate the glycolytic metabolism and oxidative stress functions of CDK4 in colon cancer. Furthermore, the inhibitory effects and potential binding sites of HER on CDK4, as well as its anti-tumor activity were investigated in CC cells. METHODS The mass spectrometry assay was performed to identify potential endogenous substrates of CDK4 and the correlation between glycolytic metabolic rate and CDK4 level in COAD patient tissues. Meanwhile, after inhibiting the activity or the expression of CDK4, the binding capacity of CDK4 to PKM2 and NRF2 and the latter two protein distributions in cytoplasm and nucleus were detected in CC cells. In vitro, the regulatory effects of the CDK4-PKM2-NRF2 axis on glycolysis and oxidative stress were performed by ECAR, OCR, and ROS assay. The inhibitory effect of HER on CDK4 activity was explored in CC cells and the potential binding sites were predicted and testified in vitro. Furthermore, tumor growth inhibition of HER by suppressing the CDK4-PKM2-NRF2 axis was also investigated in vitro and in vivo. RESULTS PKM2 and NRF2 were identified as endogenous substrates of CDK4 and, high-expressed CDK4 was associated with low-level glycolysis in COAD. In vitro, inactivated CDK4 facilitated CDK4-PKM2-NRF2 complex formation which resulted in 1) inhibited PKM2 activity and retarded the glycolytic rate; 2) cytoplasm-detained NRF2 failed to transcript anti-oxidative gene expressions and induced oxidant stress. Additionally, as a CDK4 inhibitor, HER developed triple anti-tumor effects including induced G0/G1 phase arrest, suppressed glycolysis, and disrupted the anti-oxidative capacity of CC cells. CONCLUSION The results first time revealed that CDK4 modulated glycolytic and anti-oxidative capacity of CC cells via bound to its endogenous substrates, PKM2 and NRF2. Additionally, 140Asp145Asn amino acid sites of CDK4 were potential targets of HER. HER exerts anti-tumor activity by inhibited the activity of CDK4, promoted the CDK4-PKM2-NRF2 complex formation in the CC cells.
Collapse
Affiliation(s)
- Jun-Lin Lv
- School of Traditional Chinese Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Yu-Shan Ren
- Department of Immunology, Medicine & Pharmacy Research Center, Binzhou Medical University, 264003 Yantai, China
| | - Yu-Jun Tan
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd, 276000 Linyi, China
| | - Ting Chu
- School of Traditional Chinese Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Xin-Yue Cao
- School of Traditional Chinese Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Huai-Yuan Liu
- School of Traditional Chinese Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Ru Ma
- School of Traditional Chinese Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Han Zhang
- School of Traditional Chinese Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Qiu-Sheng Zheng
- School of Traditional Chinese Medicine, Binzhou Medical University, 264003, Yantai, China.
| | - Gui-Cheng Dong
- College of Life Sciences, Inner Mongolia Agricultural University, 010011, Hohhot, China.
| | - Jie Li
- School of Traditional Chinese Medicine, Binzhou Medical University, 264003, Yantai, China.
| |
Collapse
|
26
|
Knudsen ES, Witkiewicz AK, Rubin SM. Cancer takes many paths through G1/S. Trends Cell Biol 2024; 34:636-645. [PMID: 37953123 PMCID: PMC11082069 DOI: 10.1016/j.tcb.2023.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023]
Abstract
In the commonly accepted paradigm for control of the mammalian cell cycle, sequential cyclin-dependent kinase (CDK) and cyclin activities drive the orderly transition from G1 to S phase. However, recent studies using different technological approaches and examining a broad range of cancer cell types are challenging this established paradigm. An alternative model is evolving in which cell cycles utilize different drivers and take different trajectories through the G1/S transition. We are discovering that cancer cells in particular can adapt their drivers and trajectories, which has important implications for antiproliferative therapies. These studies have helped to refine an understanding of how CDK inhibition impinges on proliferation and have significance for understanding fundamental features of cell biology and cancer.
Collapse
Affiliation(s)
- Erik S Knudsen
- Molecular and Cellular Biology, Roswell Park Cancer Center, Buffalo, NY, USA.
| | - Agnieszka K Witkiewicz
- Molecular and Cellular Biology, Roswell Park Cancer Center, Buffalo, NY, USA; Department of Pathology, Roswell Park Cancer Center, Buffalo, NY, USA
| | - Seth M Rubin
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA.
| |
Collapse
|
27
|
Morgan JE, Jaferi N, Shonibare Z, Huang GS. ARID1A in Gynecologic Precancers and Cancers. Reprod Sci 2024; 31:2150-2162. [PMID: 38740655 DOI: 10.1007/s43032-024-01585-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
The highest frequency of genetic alterations in the tumor suppressor ARID1A occurs in malignancies of the female reproductive tract. The prevalence of ARID1A alterations in gynecologic precancers and cancers is summarized from the literature, and the putative mechanisms of tumor suppressive action examined both in benign/precursor lesions including endometriosis and atypical hyperplasia and in malignancies of the ovary, uterus, cervix and vagina. ARID1A alterations in gynecologic cancers are usually loss-of-function mutations, resulting in diminished or absent protein expression. ARID1A deficiency results in pleiotropic downstream effects related not only to its role in transcriptional regulation as a SWI/SNF complex subunit, but also related to the functions of ARID1A in DNA replication and repair, immune modulation, cell cycle progression, endoplasmic reticulum (ER) stress and oxidative stress. The most promising actionable signaling pathway interactions and therapeutic vulnerabilities of ARID1A mutated cancers are presented with a critical review of the currently available experimental and clinical evidence. The role of ARID1A in response to chemotherapeutic agents, radiation therapy and immunotherapy is also addressed. In summary, the multi-faceted role of ARID1A mutation in precancer and cancer is examined through a clinical lens focused on development of novel preventive and therapeutic interventions for gynecological cancers.
Collapse
Affiliation(s)
- Jaida E Morgan
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Nishah Jaferi
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Zainab Shonibare
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Gloria S Huang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, USA.
- Department of Obstetrics, Gynecology & Reproductive Sciences, Division of Gynecologic Oncology, Yale School of Medicine, Yale Cancer Center, Yale University, PO Box 208063, New Haven, CT, 06520-8063, USA.
| |
Collapse
|
28
|
Chew SM, Ferraro E, Safonov A, Chen Y, Kelly D, Razavi P, Robson M, Seidman AD. Impact of cyclin dependent kinase 4/6 inhibitors on breast cancer brain metastasis outcomes. Eur J Cancer 2024; 207:114175. [PMID: 38896996 DOI: 10.1016/j.ejca.2024.114175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Cyclin dependent kinase 4/6 inhibitors (CDK4/6i) are recommended 1st line treatments in HR+HER2- metastatic breast cancer. However, the impact of prior CDK4/6i on the natural history of brain metastases (BM) is not well described. MATERIALS AND METHODS We reviewed retrospective data for 363 patients with HR+HER2- BM who received a CDK4/6i (CDK-Y) between 1 Jan 2015 to 31 July 2021 and 299 patients with HR+HER2- BM who did not receive a CDK4/6i (CDK-N) between 1 Jan 2010 to 31 Dec 2014. CNS PFS and OS were assessed in patients who received CDK4/6i after BM. OS from the time of BM development was assessed between patients who received CDK4/6i before BM and the CDK-N cohort RESULTS: In the CDK-Y cohort of 363 patients, 203 (56 %) received a CDK4/6i before BM, 133 (37 %) received a CDK4/6i only after BM and 27 (7 %) received a CDK4/6i both before and after BM. Median CNS PFS was 21.4 months for patients receiving a CDK4/6i only after BM and 9.4 months for patients who received CDK4/6i both before and after BM (p = 0.006). Median OS was 24.9 months for patients receiving a CDK4/6i only after BM and 12.1 months for patients who received CDK4/6i both before and after BM (p = 0.0098). Median OS from time of BM development for patients receiving a CDK4/6i before BM versus the CDK-N cohort was 4.3 months and 7.7 months respectively (p = 0.0082). CONCLUSIONS CDK4/6i exposure prior to BM may lead to development of resistance mechanisms which in turn reduces CNS PFS and OS upon rechallenging with a CDK4/6i after BM development. This motivates investigation of biomarkers for patient selection.
Collapse
Affiliation(s)
- Sonya M Chew
- Medical Oncology Service, University Hospital Galway, Galway, Ireland
| | - Emanuela Ferraro
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Anton Safonov
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Yuan Chen
- Epidemiology-Biostatistics Service, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Daniel Kelly
- Technology Division, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Pedram Razavi
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - Mark Robson
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA
| | - Andrew D Seidman
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, USA.
| |
Collapse
|
29
|
Xue Y, Zhai J. Strategy of combining CDK4/6 inhibitors with other therapies and mechanisms of resistance. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2024; 17:189-207. [PMID: 39114502 PMCID: PMC11301413 DOI: 10.62347/hgni4903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/29/2023] [Indexed: 08/10/2024]
Abstract
Cell cycle-dependent protein kinase 4/6 (CDK4/6) is a crucial kinase that regulates the cell cycle, essential for cell division and proliferation. Hence, combining CDK4/6 inhibitors with other anti-tumor drugs is a pivotal clinical strategy. This strategy can efficiently inhibit the growth and division of tumor cells, reduce the side effects, and improve the quality of life of patients by reducing the dosage of combined anticancer drugs. Furthermore, the combination therapy strategy of CDK4/6 inhibitors could ameliorate the drug resistance of combined drugs and overcome the CDK4/6 resistance caused by CDK4/6 inhibitors. Various tumor treatment strategies combined with CDK4/6 inhibitors have entered the clinical trial stage, demonstrating their substantial clinical potential. This study reviews the research progress of CDK4/6 inhibitors from 2018 to 2022, the related resistance mechanism of CDK4/6 inhibitors, and the strategy of combination medication.
Collapse
Affiliation(s)
- Yingfei Xue
- Tianjin University, School of Pharmaceutical Science and Technology (SPST)Tianjin 300072, China
| | - Jie Zhai
- Department of Breast Surgical Oncology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of SciencesHangzhou 310022, Zhejiang, China
| |
Collapse
|
30
|
Cheng X. A Comprehensive Review of HER2 in Cancer Biology and Therapeutics. Genes (Basel) 2024; 15:903. [PMID: 39062682 PMCID: PMC11275319 DOI: 10.3390/genes15070903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2), a targetable transmembrane glycoprotein receptor of the epidermal growth factor receptor (EGFR) family, plays a crucial role in cell proliferation, survival, and differentiation. Aberrant HER2 signaling is implicated in various cancers, particularly in breast and gastric cancers, where HER2 overexpression or amplification correlates with aggressive tumor behavior and poor prognosis. HER2-activating mutations contribute to accelerated tumorigenesis and metastasis. This review provides an overview of HER2 biology, signaling pathways, mechanisms of dysregulation, and diagnostic approaches, as well as therapeutic strategies targeting HER2 in cancer. Understanding the intricate details of HER2 regulation is essential for developing effective targeted therapies and improving patient outcomes.
Collapse
Affiliation(s)
- Xiaoqing Cheng
- Department of Oncology, School of Medicine, Washington University in Saint Louis, Saint Louis, MO 63108, USA
| |
Collapse
|
31
|
Patel V, Casimiro S, Abreu C, Barroso T, de Sousa RT, Torres S, Ribeiro LA, Nogueira-Costa G, Pais HL, Pinto C, Costa L, Costa L. DNA damage targeted therapy for advanced breast cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:678-698. [PMID: 38966174 PMCID: PMC11220312 DOI: 10.37349/etat.2024.00241] [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: 08/30/2023] [Accepted: 01/04/2024] [Indexed: 07/06/2024] Open
Abstract
Breast cancer (BC) is the most prevalent malignancy affecting women worldwide, including Portugal. While the majority of BC cases are sporadic, hereditary forms account for 5-10% of cases. The most common inherited mutations associated with BC are germline mutations in the BReast CAncer (BRCA) 1/2 gene (gBRCA1/2). They are found in approximately 5-6% of BC patients and are inherited in an autosomal dominant manner, primarily affecting younger women. Pathogenic variants within BRCA1/2 genes elevate the risk of both breast and ovarian cancers and give rise to distinct clinical phenotypes. BRCA proteins play a key role in maintaining genome integrity by facilitating the repair of double-strand breaks through the homologous recombination (HR) pathway. Therefore, any mutation that impairs the function of BRCA proteins can result in the accumulation of DNA damage, genomic instability, and potentially contribute to cancer development and progression. Testing for gBRCA1/2 status is relevant for treatment planning, as it can provide insights into the likely response to therapy involving platinum-based chemotherapy and poly[adenosine diphosphate (ADP)-ribose] polymerase inhibitors (PARPi). The aim of this review was to investigate the impact of HR deficiency in BC, focusing on BRCA mutations and their impact on the modulation of responses to platinum and PARPi therapy, and to share the experience of Unidade Local de Saúde Santa Maria in the management of metastatic BC patients with DNA damage targeted therapy, including those with the Portuguese c.156_157insAlu BRCA2 founder mutation.
Collapse
Affiliation(s)
- Vanessa Patel
- Oncology Division, Unidade Local de Saúde Santa Maria, 1649-028 Lisboa, Portugal
| | - Sandra Casimiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Catarina Abreu
- Oncology Division, Unidade Local de Saúde Santa Maria, 1649-028 Lisboa, Portugal
| | - Tiago Barroso
- Oncology Division, Unidade Local de Saúde Santa Maria, 1649-028 Lisboa, Portugal
| | | | - Sofia Torres
- Oncology Division, Unidade Local de Saúde Santa Maria, 1649-028 Lisboa, Portugal
| | - Leonor Abreu Ribeiro
- Oncology Division, Unidade Local de Saúde Santa Maria, 1649-028 Lisboa, Portugal
| | | | - Helena Luna Pais
- Oncology Division, Unidade Local de Saúde Santa Maria, 1649-028 Lisboa, Portugal
| | - Conceição Pinto
- Oncology Division, Unidade Local de Saúde Santa Maria, 1649-028 Lisboa, Portugal
| | - Leila Costa
- Pharmacy Department, Unidade Local de Saúde Santa Maria, 1649-028 Lisboa, Portugal
| | - Luís Costa
- Oncology Division, Unidade Local de Saúde Santa Maria, 1649-028 Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
32
|
Wang S, Xiong Y, Luo Y, Shen Y, Zhang F, Lan H, Pang Y, Wang X, Li X, Zheng X, Lu X, Liu X, Cheng Y, Wu T, Dong Y, Lu Y, Cui J, Jia X, Yang S, Wang L, Wang Y. Genome-wide CRISPR screens identify PKMYT1 as a therapeutic target in pancreatic ductal adenocarcinoma. EMBO Mol Med 2024; 16:1115-1142. [PMID: 38570712 PMCID: PMC11099189 DOI: 10.1038/s44321-024-00060-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/10/2024] [Accepted: 03/14/2024] [Indexed: 04/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with an overall 5-year survival rate of <12% due to the lack of effective treatments. Novel treatment strategies are urgently needed. Here, PKMYT1 is identified through genome-wide CRISPR screens as a non-mutant, genetic vulnerability of PDAC. Higher PKMYT1 expression levels indicate poor prognosis in PDAC patients. PKMYT1 ablation inhibits tumor growth and proliferation in vitro and in vivo by regulating cell cycle progression and inducing apoptosis. Moreover, pharmacological inhibition of PKMYT1 shows efficacy in multiple PDAC cell models and effectively induces tumor regression without overt toxicity in PDAC cell line-derived xenograft and in more clinically relevant patient-derived xenograft models. Mechanistically, in addition to its canonical function of phosphorylating CDK1, PKMYT1 functions as an oncogene to promote PDAC tumorigenesis by regulating PLK1 expression and phosphorylation. Finally, TP53 function and PRKDC activation are shown to modulate the sensitivity to PKMYT1 inhibition. These results define PKMYT1 dependency in PDAC and identify potential therapeutic strategies for clinical translation.
Collapse
Affiliation(s)
- Simin Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yangjie Xiong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yuxiang Luo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yanying Shen
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Fengrui Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 200011, Shanghai, China
| | - Haoqi Lan
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yuzhi Pang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaofang Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaoqi Li
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Xufen Zheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaojing Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaoxiao Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yumei Cheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Tanwen Wu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yue Dong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yuan Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 200011, Shanghai, China
| | - Jiujie Cui
- Department of Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Xiaona Jia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Sheng Yang
- Department of Oncology, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Liwei Wang
- Department of Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China.
| | - Yuexiang Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
33
|
Sun J, Du R, Li X, Liu C, Wang D, He X, Li G, Zhang K, Wang S, Hao Q, Zhang Y, Li M, Gao Y, Zhang C. CD63 + cancer-associated fibroblasts confer CDK4/6 inhibitor resistance to breast cancer cells by exosomal miR-20. Cancer Lett 2024; 588:216747. [PMID: 38403110 DOI: 10.1016/j.canlet.2024.216747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
Cyclin-dependent kinase 4 (CDK4) and CDK6 inhibitors (CDK4/6i) have rapidly received Food and Drug Administration (FDA) approval as a new type of therapy for patients with advanced hormone receptor-positive breast cancer. However, with the widespread application of CDK4/6i, drug resistance has become a new challenge for clinical practice and has greatly limited the treatment effect. Here, the whole microenvironment landscape of ER+ breast cancer tumors was revealed through single-cell RNA sequencing, and a specific subset of cancer-associated fibroblasts (CD63+ CAFs) was identified as highly enriched in CDK4/6i resistant tumor tissues. Then, we found that CD63+ CAFs can distinctly promote resistance to CDK4/6i in breast cancer cells and tumor xenografts. In addition, it was discovered that miR-20 is markedly enriched in the CD63+ CAFs-derived exosomes, which are used to communicate with ER+ breast cancer cells, leading to CDK4/6i resistance. Furthermore, exosomal miR-20 could directly target the RB1 mRNA 3'UTR and negatively regulate RB1 expression to decrease CDK4/6i sensitivity in breast cancer cells. Most importantly, we designed and synthesized cRGD-miR-20 sponge nanoparticles and found that they can enhance the therapeutic effect of CDK4/6i in breast cancer. In summary, our findings reveal that CD63+ CAFs can promote CDK4/6i resistance via exosomal miR-20, which induces the downregulation of RB1 in breast cancer cells, and suggest that CD63+ CAFs may be a novel therapeutic target to enhance CDK4/6i sensitivity.
Collapse
Affiliation(s)
- Jiahui Sun
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Ruoxin Du
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Xiaoju Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China; Bioinformatics Center of AMMS, Beijing, 100850, PR China
| | - Chenlin Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Donghui Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Xiangmei He
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Guodong Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Kuo Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Shuning Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Qiang Hao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Yingqi Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Meng Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China.
| | - Yuan Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China.
| | - Cun Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China.
| |
Collapse
|
34
|
Asghar A, Chohan TA, Khurshid U, Saleem H, Mustafa MW, Khursheed A, Alafnan A, Batul R, Bin Break MK, Almansour K, Anwar S. A systematic review on understanding the mechanistic pathways and clinical aspects of natural CDK inhibitors on cancer progression.: Unlocking cellular and biochemical mechanisms. Chem Biol Interact 2024; 393:110940. [PMID: 38467339 DOI: 10.1016/j.cbi.2024.110940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/13/2024]
Abstract
Cell division, differentiation, and controlled cell death are all regulated by phosphorylation, a key biological function. This mechanism is controlled by a variety of enzymes, with cyclin-dependent kinases (CDKs) being particularly important in phosphorylating proteins at serine and threonine sites. CDKs, which contain 20 unique components, serve an important role in regulating vital physiological functions such as cell cycle progression and gene transcription. Methodologically, an extensive literature search was performed using reputable databases such as PubMed, Google Scholar, Scopus, and Web of Science. Keywords encompassed "cyclin kinase," "cyclin dependent kinase inhibitors," "CDK inhibitors," "natural products," and "cancer therapy." The inclusion criteria, focused on relevance, publication date, and language, ensured a thorough representation of the most recent research in the field, encompassing articles published from January 2015 to September 2023. Categorization of CDKs into those regulating transcription and those orchestrating cell cycle phases provides a comprehensive understanding of their diverse functions. Ongoing clinical trials featuring CDK inhibitors, notably CDK7 and CDK4/6 inhibitors, illuminate their promising potential in various cancer treatments. This review undertakes a thorough investigation of CDK inhibitors derived from natural (marine, terrestrial, and peptide) sources. The aim of this study is to provide a comprehensive comprehension of the chemical classifications, origins, target CDKs, associated cancer types, and therapeutic applications.
Collapse
Affiliation(s)
- Andleeb Asghar
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary and Animal Sciences (UVAS), Lahore, Pakistan
| | - Tahir Ali Chohan
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary and Animal Sciences (UVAS), Lahore, Pakistan.
| | - Umair Khurshid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100 Bahawalpur, Pakistan
| | - Hammad Saleem
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary and Animal Sciences (UVAS), Lahore, Pakistan.
| | - Mian Waqar Mustafa
- Department of Pharmacy, Forman Christian College University, Lahore, Pakistan
| | - Anjum Khursheed
- Department of Pharmacy, Grand Asian University, Sialkot, Pakistan
| | - Ahmed Alafnan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Saudi Arabia
| | - Rahila Batul
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
| | - Mohammed Khaled Bin Break
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
| | - Khaled Almansour
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
| | - Sirajudheen Anwar
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Saudi Arabia
| |
Collapse
|
35
|
Mikheeva AM, Bogomolov MA, Gasca VA, Sementsov MV, Spirin PV, Prassolov VS, Lebedev TD. Improving the power of drug toxicity measurements by quantitative nuclei imaging. Cell Death Discov 2024; 10:181. [PMID: 38637526 PMCID: PMC11026393 DOI: 10.1038/s41420-024-01950-3] [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: 01/16/2024] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
Imaging-based anticancer drug screens are becoming more prevalent due to development of automated fluorescent microscopes and imaging stations, as well as rapid advancements in image processing software. Automated cell imaging provides many benefits such as their ability to provide high-content data, modularity, dynamics recording and the fact that imaging is the most direct way to access cell viability and cell proliferation. However, currently most publicly available large-scale anticancer drugs screens, such as GDSC, CTRP and NCI-60, provide cell viability data measured by assays based on colorimetric or luminometric measurements of NADH or ATP levels. Although such datasets provide valuable data, it is unclear how well drug toxicity measurements can be integrated with imaging data. Here we explored the relations between drug toxicity data obtained by XTT assay, two quantitative nuclei imaging methods and trypan blue dye exclusion assay using a set of four cancer cell lines with different morphologies and 30 drugs with different mechanisms of action. We show that imaging-based approaches provide high accuracy and the differences between results obtained by different methods highly depend on drug mechanism of action. Selecting AUC metrics over IC50 or comparing data where significantly drugs reduced cell numbers noticeably improves consistency between methods. Using automated cell segmentation protocols we analyzed mitochondria activity in more than 11 thousand drug-treated cells and showed that XTT assay produces unreliable data for CDK4/6, Aurora A, VEGFR and PARP inhibitors due induced cell size growth and increase in individual mitochondria activity. We also explored several benefits of image-based analysis such as ability to monitor cell number dynamics, dissect changes in total and individual mitochondria activity from cell proliferation, and ability to identify chromatin remodeling drugs. Finally, we provide a web tool that allows comparing results obtained by different methods.
Collapse
Affiliation(s)
- Alesya M Mikheeva
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy per., Dolgoprudny, Moscow Region, 141701, Russia
| | - Mikhail A Bogomolov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy per., Dolgoprudny, Moscow Region, 141701, Russia
| | - Valentina A Gasca
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy per., Dolgoprudny, Moscow Region, 141701, Russia
| | - Mikhail V Sementsov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy per., Dolgoprudny, Moscow Region, 141701, Russia
| | - Pavel V Spirin
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
| | - Vladimir S Prassolov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
| | - Timofey D Lebedev
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia.
| |
Collapse
|
36
|
Ye M, Xu H, Ding J, Jiang L. Therapy for Hormone Receptor-Positive, Human Epidermal Growth Receptor 2-Negative Metastatic Breast Cancer Following Treatment Progression via CDK4/6 Inhibitors: A Literature Review. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:181-197. [PMID: 38617842 PMCID: PMC11016260 DOI: 10.2147/bctt.s438366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 01/16/2024] [Indexed: 04/16/2024]
Abstract
Endocrine therapy (ET) with a cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) is currently the first-line standard treatment for most patients with hormone receptor-positive (HR+) and human epidermal growth receptor 2-negative (HER2-) metastatic or advanced breast cancer. However, the majority of tumors response to and eventually develop resistance to CDK4/6is. The mechanisms of resistance are poorly understood, and the optimal postprogression treatment regimens and their sequences continue to evolve in the rapidly changing treatment landscape. In this review, we generally summarize the mechanisms of resistance to CDK4/6is and ET, and describe the findings from clinical trials using small molecule inhibitors, antibody-drug conjugates and immunotherapy, providing insights into how these novel strategies may reverse treatment resistance, and discussing how some have not translated into clinical benefit. Finally, we provide rational treatment strategies based on the current emerging evidence.
Collapse
Affiliation(s)
- Meixi Ye
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, 315040, People’s Republic of China
| | - Hao Xu
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, 315040, People’s Republic of China
| | - Jinhua Ding
- Department of Breast and Thyroid Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, 315040, People’s Republic of China
| | - Li Jiang
- Department of General Practice, Ningbo Medical Center Lihuili Hospital, Ningbo, 315040, People’s Republic of China
| |
Collapse
|
37
|
Csergeová L, Krbušek D, Janoštiak R. CIP/KIP and INK4 families as hostages of oncogenic signaling. Cell Div 2024; 19:11. [PMID: 38561743 PMCID: PMC10985988 DOI: 10.1186/s13008-024-00115-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
CIP/KIP and INK4 families of Cyclin-dependent kinase inhibitors (CKIs) are well-established cell cycle regulatory proteins whose canonical function is binding to Cyclin-CDK complexes and altering their function. Initial experiments showed that these proteins negatively regulate cell cycle progression and thus are tumor suppressors in the context of molecular oncology. However, expanded research into the functions of these proteins showed that most of them have non-canonical functions, both cell cycle-dependent and independent, and can even act as tumor enhancers depending on their posttranslational modifications, subcellular localization, and cell state context. This review aims to provide an overview of canonical as well as non-canonical functions of CIP/KIP and INK4 families of CKIs, discuss the potential avenues to promote their tumor suppressor functions instead of tumor enhancing ones, and how they could be utilized to design improved treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Lucia Csergeová
- BIOCEV-First Faculty of Medicine, Charles University, Prague, Czechia
| | - David Krbušek
- BIOCEV-First Faculty of Medicine, Charles University, Prague, Czechia
| | | |
Collapse
|
38
|
Gómez Tejeda Zañudo J, Barroso-Sousa R, Jain E, Jin Q, Li T, Buendia-Buendia JE, Pereslete A, Abravanel DL, Ferreira AR, Wrabel E, Helvie K, Hughes ME, Partridge AH, Overmoyer B, Lin NU, Tayob N, Tolaney SM, Wagle N. Exemestane plus everolimus and palbociclib in metastatic breast cancer: clinical response and genomic/transcriptomic determinants of resistance in a phase I/II trial. Nat Commun 2024; 15:2446. [PMID: 38503755 PMCID: PMC10951222 DOI: 10.1038/s41467-024-45835-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/02/2024] [Indexed: 03/21/2024] Open
Abstract
The landscape of cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) resistance is still being elucidated and the optimal subsequent therapy to overcome resistance remains uncertain. Here we present the final results of a phase Ib/IIa, open-label trial (NCT02871791) of exemestane plus everolimus and palbociclib for CDK4/6i-resistant metastatic breast cancer. The primary objective of phase Ib was to evaluate safety and tolerability and determine the maximum tolerated dose/recommended phase II dose (100 mg palbociclib, 5 mg everolimus, 25 mg exemestane). The primary objective of phase IIa was to determine the clinical benefit rate (18.8%, n = 6/32), which did not meet the predefined endpoint (65%). Secondary objectives included pharmacokinetic profiling (phase Ib), objective response rate, disease control rate, duration of response, and progression free survival (phase IIa), and correlative multi-omics analysis to investigate biomarkers of resistance to CDK4/6i. All participants were female. Multi-omics data from the phase IIa patients (n = 24 tumor/17 blood biopsy exomes; n = 27 tumor transcriptomes) showed potential mechanisms of resistance (convergent evolution of HER2 activation, BRAFV600E), identified joint genomic/transcriptomic resistance features (ESR1 mutations, high estrogen receptor pathway activity, and a Luminal A/B subtype; ERBB2/BRAF mutations, high RTK/MAPK pathway activity, and a HER2-E subtype), and provided hypothesis-generating results suggesting that mTOR pathway activation correlates with response to the trial's therapy. Our results illustrate how genome and transcriptome sequencing may help better identify patients likely to respond to CDK4/6i therapies.
Collapse
Affiliation(s)
- Jorge Gómez Tejeda Zañudo
- Cancer Program, Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Romualdo Barroso-Sousa
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Oncology Center, Hospital Sírio-Libanês, Brasília, Brazil
| | - Esha Jain
- Cancer Program, Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Repare Therapeutics, Cambridge, MA, USA
| | - Qingchun Jin
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, MA, USA
| | - Tianyu Li
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, MA, USA
| | - Jorge E Buendia-Buendia
- Cancer Program, Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cellarity, Somerville, MA, USA
| | | | - Daniel L Abravanel
- Cancer Program, Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Arlindo R Ferreira
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Unit, Champalimaud Clinical Centre, Champalimaud Foundation, Lisbon, Portugal
| | - Eileen Wrabel
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Karla Helvie
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Ann H Partridge
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Beth Overmoyer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nancy U Lin
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nabihah Tayob
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, MA, USA
| | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nikhil Wagle
- Cancer Program, Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Genentech, South San Francisco, CA, USA.
| |
Collapse
|
39
|
Foy R, Lew KX, Saurin AT. The search for CDK4/6 inhibitor biomarkers has been hampered by inappropriate proliferation assays. NPJ Breast Cancer 2024; 10:19. [PMID: 38438376 PMCID: PMC10912267 DOI: 10.1038/s41523-024-00624-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 02/16/2024] [Indexed: 03/06/2024] Open
Abstract
CDK4/6 inhibitors are effective at treating advanced HR+ /HER2- breast cancer, however biomarkers that can predict response are urgently needed. We demonstrate here that previous large-scale screens designed to identify which tumour types or genotypes are most sensitive to CDK4/6 inhibitors have misrepresented the responsive cell lines because of a reliance on metabolic proliferation assays. CDK4/6-inhibited cells arrest in G1 but continue to grow in size, thereby producing more mitochondria. We show that this growth obscures the arrest using ATP-based proliferation assays but not if DNA-based assays are used instead. Furthermore, lymphoma lines, previously identified as the most sensitive, simply appear to respond the best using ATP-based assays because they fail to overgrow during the G1 arrest. Similarly, the CDK4/6 inhibitor abemaciclib appears to inhibit proliferation better than palbociclib because it also restricts cellular overgrowth through off-target effects. DepMap analysis of screening data using reliable assay types, demonstrates that palbociclib-sensitive cell types are also sensitive to Cyclin D1, CDK4 and CDK6 knockout/knockdown, whereas the palbociclib-resistant lines are sensitive to Cyclin E1, CDK2 and SKP2 knockout/knockdown. Potential biomarkers of palbociclib-sensitive cells are increased expression of CCND1 and RB1, and reduced expression of CCNE1 and CDKN2A. Probing DepMap with similar data from metabolic assays fails to reveal these associations. Together, this demonstrates why CDK4/6 inhibitors, and any other anti-cancer drugs that arrest the cell cycle but permit continued cell growth, must now be re-screened against a wide-range of cell types using an appropriate proliferation assay. This would help to better inform clinical trials and to identify much needed biomarkers of response.
Collapse
Affiliation(s)
- Reece Foy
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Kah Xin Lew
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Adrian T Saurin
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
40
|
Sugiyama N, Uehara O, Kawano Y, Paudel D, Morikawa T, Nakamoto N, Kato S, Takayama T, Nagasawa T, Miura H, Abiko Y, Furuichi Y. Ingenuity pathway analysis of gingival epithelial cells stimulated with estradiol and progesterone. J Oral Biosci 2024; 66:26-34. [PMID: 37949170 DOI: 10.1016/j.job.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE Periodontal disease is a risk factor for preterm delivery, and elevated female hormone levels during pregnancy promote hormone-dependent periodontopathogenic bacterial growth and gingivitis. Although the saliva of pregnant women contains female hormones at elevated levels, their effects on the gingiva are poorly understood. Therefore, in this study, we investigated the effects of estradiol and progesterone stimulation on gingival epithelial cells via ingenuity pathway analysis. METHODS Human gingival epithelial progenitors were cultured in a CnT-Prime medium; 17β-estradiol (E2) and progesterone (P4) were used as the reagents. Cells treated with dimethyl sulfoxide alone were used as the control group. Cells in the control and experimental groups were incubated for 12 h. RNA was extracted from the cultured cells, RNA-Seq was performed, and pathway analysis was conducted. RESULTS Differentially expressed genes were detected for 699 (over 2-fold increase) and 348 (decrease) genes in group E2 and for 1448 (increase) and 924 (decrease) genes in group P4 compared with those in the control group (FDR <0.05, n = 4). The z-scores of the pathways suggest that E2 and P4 increased the activity of the wound healing signaling pathway. The activation of this pathway was higher in the E2 and P4 groups than that in the control group. CONCLUSIONS The results of this study suggest that estradiol and progesterone may affect gingival homeostasis and wound healing.
Collapse
Affiliation(s)
- Nodoka Sugiyama
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health, Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan.
| | - Osamu Uehara
- Division of Disease Controlrol and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061- 0293, Japan
| | - Yutaka Kawano
- Department of Gastroenterology and Oncology Tokushima University Graduate School of Biomedical Sciences, Tokushima, 3-18-15, Kuramoto-cho, Tokushima City, Tokushima, 770-8503, Japan
| | - Durga Paudel
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, 1757, Ishikari- Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| | - Tetsuro Morikawa
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| | - Norihiro Nakamoto
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health, Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| | - Satsuki Kato
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health, Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology Tokushima University Graduate School of Biomedical Sciences, Tokushima, 3-18-15, Kuramoto-cho, Tokushima City, Tokushima, 770-8503, Japan
| | - Toshiyuki Nagasawa
- Division of Advanced Clinical Education, Department of Integrated Dental Education, School of Dentistry, Health, Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| | - Hiroko Miura
- Division of Disease Controlrol and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061- 0293, Japan
| | - Yoshihiro Abiko
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan.
| | - Yasushi Furuichi
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health, Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| |
Collapse
|
41
|
Ou Y, Wang M, Xu Q, Sun B, Jia Y. Small molecule agents for triple negative breast cancer: Current status and future prospects. Transl Oncol 2024; 41:101893. [PMID: 38290250 PMCID: PMC10840364 DOI: 10.1016/j.tranon.2024.101893] [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: 11/16/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with poor prognosis. The number of cases increased by 2.26 million in 2020, making it the most commonly diagnosed cancer type in the world. TNBCs lack hormone receptor (HR) and human epidermal growth factor 2 (HER2), which limits treatment options. Currently, paclitaxel-based drugs combined with other chemotherapeutics remain the main treatment for TNBC. There is currently no consensus on the best therapeutic regimen for TNBC. However, there have been successful clinical trials exploring large-molecule monoclonal antibodies, small-molecule targeted drugs, and novel antibody-drug conjugate (ADC). Although monoclonal antibodies have produced clinical success, their large molecular weight can limit therapeutic benefits. It is worth noting that in the past 30 years, the FDA has approved small molecule drugs for HER2-positive breast cancers. The lack of effective targets and the occurrence of drug resistance pose significant challenges in the treatment of TNBC. To improve the prognosis of TNBC, it is crucial to search for effective targets and to overcome drug resistance. This review examines the clinical efficacy, adverse effects, resistance mechanisms, and potential solutions of targeted small molecule drugs in both monotherapies and combination therapies. New therapeutic targets, including nuclear export protein 1 (XPO1) and hedgehog (Hh), are emerging as potential options for researchers and become integrated into clinical trials for TNBC. Additionally, there is growing interest in the potential of targeted protein degradation chimeras (PROTACs), degraders of rogue proteins, as a future therapy direction. This review provides potentially valuable insights with clinical implications.
Collapse
Affiliation(s)
- Yan Ou
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Mengchao Wang
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qian Xu
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Binxu Sun
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yingjie Jia
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
42
|
Zhu K, Yang X, Tai H, Zhong X, Luo T, Zheng H. HER2-targeted therapies in cancer: a systematic review. Biomark Res 2024; 12:16. [PMID: 38308374 PMCID: PMC10835834 DOI: 10.1186/s40364-024-00565-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 01/15/2024] [Indexed: 02/04/2024] Open
Abstract
Abnormal alterations in human epidermal growth factor receptor 2 (HER2, neu, and erbB2) are associated with the development of many tumors. It is currently a crucial treatment for multiple cancers. Advanced in molecular biology and further exploration of the HER2-mediated pathway have promoted the development of medicine design and combination drug regimens. An increasing number of HER2-targeted drugs including specific monoclonal antibodies, tyrosine kinase inhibitors (TKIs), and antibody-drug conjugates (ADCs) have been approved by the U.S. Food and Drug Administration. The emergence of ADCs, has significantly transformed the treatment landscape for various tumors, such as breast, gastric, and bladder cancer. Classic monoclonal antibodies and novel TKIs have not only demonstrated remarkable efficacy, but also expanded their indications, with ADCs in particular exhibiting profound clinical applications. Moreover the concept of low HER2 expression signifies a breakthrough in HER2-targeted therapy, indicating that an increasing number of tumors and patients will benefit from this approach. This article, provides a comprehensive review of the underlying mechanism of action, representative drugs, corresponding clinical trials, recent advancements, and future research directions pertaining to HER2-targeted therapy.
Collapse
Affiliation(s)
- Kunrui Zhu
- Institute for Breast Health Medicine, Cance Center, Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xinyi Yang
- College of Clinical Medical, Guizhou Medical University, Guiyang, 550000, Guizhou Province, China
| | - Hebei Tai
- College of Clinical Medical, Guizhou Medical University, Guiyang, 550000, Guizhou Province, China
| | - Xiaorong Zhong
- Institute for Breast Health Medicine, Cance Center, Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ting Luo
- Institute for Breast Health Medicine, Cance Center, Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Hong Zheng
- Institute for Breast Health Medicine, Cance Center, Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
43
|
Morrison L, Loibl S, Turner NC. The CDK4/6 inhibitor revolution - a game-changing era for breast cancer treatment. Nat Rev Clin Oncol 2024; 21:89-105. [PMID: 38082107 DOI: 10.1038/s41571-023-00840-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 01/27/2024]
Abstract
Cyclin-dependent kinase (CDK) 4/6 inhibition in combination with endocrine therapy is the standard-of-care treatment for patients with advanced-stage hormone receptor-positive, HER2 non-amplified (HR+HER2-) breast cancer. These agents can also be administered as adjuvant therapy to patients with higher-risk early stage disease. Nonetheless, the clinical success of these agents has created several challenges, such as how to address acquired resistance, identifying which patients are most likely to benefit from therapy prior to treatment, and understanding the optimal timing of administration and sequencing of these agents. In this Review, we describe the rationale for targeting CDK4/6 in patients with breast cancer, including a summary of updated clinical evidence and how this should inform clinical practice. We also discuss ongoing research efforts that are attempting to address the various challenges created by the widespread implementation of these agents.
Collapse
Affiliation(s)
- Laura Morrison
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
- Breast Unit, The Royal Marsden Hospital, London, UK
| | - Sibylle Loibl
- German Breast Group, Goethe University, Frankfurt, Germany
| | - Nicholas C Turner
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK.
- Breast Unit, The Royal Marsden Hospital, London, UK.
| |
Collapse
|
44
|
Zheng M, Zhang XY, Chen W, Xia F, Yang H, Yuan K, Yang P. Molecules inducing specific cyclin-dependent kinase degradation and their possible use in cancer therapy. Future Med Chem 2024; 16:369-388. [PMID: 38288571 DOI: 10.4155/fmc-2023-0259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/12/2024] [Indexed: 02/07/2024] Open
Abstract
Cyclin-dependent kinases (CDKs) play an important role in the regulation of cell proliferation, and many CDK inhibitors were developed. However, pan-CDK inhibitors failed to be approved due to intolerant toxicity or low efficacy and the use of selective CDK4/6 inhibitors is limited by resistance. Protein degraders have the potential to increase selectivity, efficacy and overcome resistance, which provides a novel strategy for regulating CDKs. In this review, we summarized the function of CDKs in regulating the cell cycle and transcription, and introduced the representative CDK inhibitors. Then we made a detailed introduction about four types of CDKs degraders, including their action mechanisms, research status and application prospects, which could help the development of novel CDKs degraders.
Collapse
Affiliation(s)
- Mingming Zheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiao-Yu Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Weijiao Chen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Fei Xia
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Huanaoyu Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
45
|
Tsai CJ, Yang JT, Shaverdian N, Patel J, Shepherd AF, Eng J, Guttmann D, Yeh R, Gelblum DY, Namakydoust A, Preeshagul I, Modi S, Seidman A, Traina T, Drullinsky P, Flynn J, Zhang Z, Rimner A, Gillespie EF, Gomez DR, Lee NY, Berger M, Robson ME, Reis-Filho JS, Riaz N, Rudin CM, Powell SN. Standard-of-care systemic therapy with or without stereotactic body radiotherapy in patients with oligoprogressive breast cancer or non-small-cell lung cancer (Consolidative Use of Radiotherapy to Block [CURB] oligoprogression): an open-label, randomised, controlled, phase 2 study. Lancet 2024; 403:171-182. [PMID: 38104577 PMCID: PMC10880046 DOI: 10.1016/s0140-6736(23)01857-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/27/2023] [Accepted: 08/31/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Most patients with metastatic cancer eventually develop resistance to systemic therapy, with some having limited disease progression (ie, oligoprogression). We aimed to assess whether stereotactic body radiotherapy (SBRT) targeting oligoprogressive sites could improve patient outcomes. METHODS We did a phase 2, open-label, randomised controlled trial of SBRT in patients with oligoprogressive metastatic breast cancer or non-small-cell lung cancer (NSCLC) after having received at least first-line systemic therapy, with oligoprogression defined as five or less progressive lesions on PET-CT or CT. Patients aged 18 years or older were enrolled from a tertiary cancer centre in New York, NY, USA, and six affiliated regional centres in the states of New York and New Jersey, with a 1:1 randomisation between standard of care (standard-of-care group) and SBRT plus standard of care (SBRT group). Randomisation was done with a computer-based algorithm with stratification by number of progressive sites of metastasis, receptor or driver genetic alteration status, primary site, and type of systemic therapy previously received. Patients and investigators were not masked to treatment allocation. The primary endpoint was progression-free survival, measured up to 12 months. We did a prespecified subgroup analysis of the primary endpoint by disease site. All analyses were done in the intention-to-treat population. The study is registered with ClinicalTrials.gov, NCT03808662, and is complete. FINDINGS From Jan 1, 2019, to July 31, 2021, 106 patients were randomly assigned to standard of care (n=51; 23 patients with breast cancer and 28 patients with NSCLC) or SBRT plus standard of care (n=55; 24 patients with breast cancer and 31 patients with NSCLC). 16 (34%) of 47 patients with breast cancer had triple-negative disease, and 51 (86%) of 59 patients with NSCLC had no actionable driver mutation. The study was closed to accrual before reaching the targeted sample size, after the primary efficacy endpoint was met during a preplanned interim analysis. The median follow-up was 11·6 months for patients in the standard-of-care group and 12·1 months for patients in the SBRT group. The median progression-free survival was 3·2 months (95% CI 2·0-4·5) for patients in the standard-of-care group versus 7·2 months (4·5-10·0) for patients in the SBRT group (hazard ratio [HR] 0·53, 95% CI 0·35-0·81; p=0·0035). The median progression-free survival was higher for patients with NSCLC in the SBRT group than for those with NSCLC in the standard-of-care group (10·0 months [7·2-not reached] vs 2·2 months [95% CI 2·0-4·5]; HR 0·41, 95% CI 0·22-0·75; p=0·0039), but no difference was found for patients with breast cancer (4·4 months [2·5-8·7] vs 4·2 months [1·8-5·5]; 0·78, 0·43-1·43; p=0·43). Grade 2 or worse adverse events occurred in 21 (41%) patients in the standard-of-care group and 34 (62%) patients in the SBRT group. Nine (16%) patients in the SBRT group had grade 2 or worse toxicities related to SBRT, including gastrointestinal reflux disease, pain exacerbation, radiation pneumonitis, brachial plexopathy, and low blood counts. INTERPRETATION The trial showed that progression-free survival was increased in the SBRT plus standard-of-care group compared with standard of care only. Oligoprogression in patients with metastatic NSCLC could be effectively treated with SBRT plus standard of care, leading to more than a four-times increase in progression-free survival compared with standard of care only. By contrast, no benefit was observed in patients with oligoprogressive breast cancer. Further studies to validate these findings and understand the differential benefits are warranted. FUNDING National Cancer Institute.
Collapse
Affiliation(s)
- Chiaojung Jillian Tsai
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Jonathan T Yang
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - Narek Shaverdian
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juber Patel
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Annemarie F Shepherd
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juliana Eng
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Guttmann
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Randy Yeh
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daphna Y Gelblum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Azadeh Namakydoust
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Isabel Preeshagul
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Seidman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tiffany Traina
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pamela Drullinsky
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhigang Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Erin F Gillespie
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - Daniel R Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Berger
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark E Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simon N Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
46
|
Grote I, Poppe A, Lehmann U, Christgen M, Kreipe H, Bartels S. Frequency of genetic alterations differs in advanced breast cancer between metastatic sites. Genes Chromosomes Cancer 2024; 63:e23199. [PMID: 37672607 DOI: 10.1002/gcc.23199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/15/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023] Open
Abstract
About 20%-30% of breast cancer (BC) patients will develop distant metastases, preferentially in bones, liver, lung, and brain. BCs with different intrinsic subtypes prefer different sites for metastasis. These subtypes vary in the abundance of genetic alterations which may influence the localization of metastases. Currently, information about the relation between metastatic site and mutational profile of BC is limited. In this study, n = 521 BC metastases of the most frequently affected sites (bone, brain, liver, and lung) were investigated for the frequency of AKT1, ERBB2, ESR1, PIK3CA, and TP53 mutations via NGS and pyrosequencing. Somatic mutations were present in 64% cases. PIK3CA and TP53 were the most frequently mutated genes under study. We provide an analysis of the mutational profile of BCs and the affected metastatic site. Genetic alterations differed significantly depending on the organ site affected by metastases. TP53 mutations were mostly observed in brain metastases (51.0%), metastases outside of the brain revealed a much lower proportion of TP53 mutated samples. PIK3CA mutations are frequent in liver (40.6%), lung (36.8%), and bone metastases (35.7%), whereas less common in brain metastases (18.4%). The highest percentage of ESR1 mutations was observed in liver and lung metastases (about 30% each), whereas metastatic lesions in the brain showed significantly less ESR1 mutations, only in 2.0% of the cases. In summary, we found significant differences of mutational status in mBC depending on the affected organ and intrinsic subtype. Organotropism of metastatic cancer spread may be influenced by the mutational profile of individual BCs.
Collapse
Affiliation(s)
- Isabel Grote
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Alexandra Poppe
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Ulrich Lehmann
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | | | - Hans Kreipe
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Stephan Bartels
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
47
|
Xia T, Dai X, Sang M, Zhang X, Xu F, Wu J, Shi L, Wei J, Ding Q. IGF2BP2 Drives Cell Cycle Progression in Triple-Negative Breast Cancer by Recruiting EIF4A1 to Promote the m6A-Modified CDK6 Translation Initiation Process. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305142. [PMID: 37983610 PMCID: PMC10767445 DOI: 10.1002/advs.202305142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/02/2023] [Indexed: 11/22/2023]
Abstract
IGF2BP2 is a new identified N6-methyladenosine (m6A) reader and associated with poor prognosis in many tumors. However, its role and related mechanism in breast cancer, especially in triple-negative breast cancer (TNBC), remains unclear. In this study, it is found that IGF2BP2 is highly expressed in TNBC due to the lower methylation level in its promoter region. Functional and mechanical studies displayed that IGF2BP2 could promote TNBC proliferation and the G1/S phase transition of the cell cycle via directly regulating CDK6 in an m6A-dependent manner. Surprising, the regulation of protein levels of CDK6 by IGF2BP2 is related to the changes in translation rate during translation initiation, rather than mRNA stability. Moreover, EIF4A1 is found to be recruited by IGF2BP2 to promote the translation output of CDK6, providing new evidence for a regulatory role of IGF2BP2 between m6A methylation and translation. Downregulation of IGF2BP2 in TNBC cell could enhance the sensitivity to abemaciclib, a CDK4/6 inhibitor. To sum up, our study revealed IGF2BP2 could facilitate the translation output of CDK6 via recruiting EIF4A1 and also provided a potential therapeutic target for the diagnosis and treatment of TNBC, as well as a new strategy for broadening the drug indications for CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Tian Xia
- Jiangsu Breast Disease CenterThe First Affiliated Hospital with Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Xin‐Yuan Dai
- Jiangsu Breast Disease CenterThe First Affiliated Hospital with Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Ming‐Yi Sang
- Jiangsu Breast Disease CenterThe First Affiliated Hospital with Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Xu Zhang
- Jiangsu Breast Disease CenterThe First Affiliated Hospital with Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Feng Xu
- Jiangsu Breast Disease CenterThe First Affiliated Hospital with Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Jing Wu
- Jiangsu Breast Disease CenterThe First Affiliated Hospital with Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Liang Shi
- Jiangsu Breast Disease CenterThe First Affiliated Hospital with Nanjing Medical University300 Guangzhou RoadNanjing210029China
| | - Ji‐Fu Wei
- Department of PharmacyJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical UniversityNanjing210029China
| | - Qiang Ding
- Jiangsu Breast Disease CenterThe First Affiliated Hospital with Nanjing Medical University300 Guangzhou RoadNanjing210029China
| |
Collapse
|
48
|
Zheng L, Luthra R, Alvarez HA, San Lucas FA, Duose DY, Wistuba II, Fuller GN, Ballester LY, Roy-Chowdhuri S, Sweeney KJ, Rashid A, Yang RK, Chen W, Liu A, Wu Y, Albarracin C, Patel KP, Routbort MJ, Sahin AA, Ding Q, Chen H. Intragenic EGFR::EGFR.E1E8 Fusion (EGFRvIII) in 4331 Solid Tumors. Cancers (Basel) 2023; 16:6. [PMID: 38201434 PMCID: PMC10778229 DOI: 10.3390/cancers16010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024] Open
Abstract
Epidermal growth factor receptor variant III (EGFRvIII, the deletion of exons 2-7) is a recurrent intragenic EGFR::EGFR.E1E8 fusion that occurs in high-grade gliomas. The presence of EGFRvIII in other solid tumors has not been well characterized. We retrospectively reviewed advanced malignant solid tumor cases tested by a custom hybrid capture 610-gene next-generation sequencing platform from 2021 to 2022. EGFRvIII was identified in 17 of 4331 (0.4%) cases, including 16 of 238 (7%) brain tumors and 1/301 (0.3%) breast tumors. EGFRvIII-positive brain tumors were all glioblastoma IDH-wildtype, most with concurrent TERT promoter mutation (14 of 16), EGFR amplification (13 of 16), and EGFR mutation (8 of 16). The only EGFRvIII-positive breast lesion was a sarcomatoid neoplasm in a young female patient. A separate breast case tested outside our institution with reported EGFRvIII was noted in a young female patient with a malignant phyllodes tumor with stromal overgrowth. Microscopically, both EGFRvIII-positive breast tumors showed high-grade sarcomatoid morphology with brisk mitotic activity. In summary, EGFRvIII is rare, occurring primarily in glioblastoma and rarely in breast sarcomatoid neoplasm, with no instances identified in other tumor types in our series. This select group of patients may benefit from chemotherapy and/or targeted anti-EGFR therapy.
Collapse
Affiliation(s)
- Lan Zheng
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (H.A.A.)
| | - Hector A. Alvarez
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (H.A.A.)
| | - F. Anthony San Lucas
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (H.A.A.)
| | - Dzifa Y. Duose
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (I.I.W.)
| | - Ignacio I. Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (I.I.W.)
| | - Gregory N. Fuller
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Leomar Y. Ballester
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Sinchita Roy-Chowdhuri
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Keith J. Sweeney
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Asif Rashid
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Richard K. Yang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Wei Chen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (H.A.A.)
| | - Audrey Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Yun Wu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Constance Albarracin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Keyur P. Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (H.A.A.)
| | - Mark J. Routbort
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (H.A.A.)
| | - Aysegul A. Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Qingqing Ding
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| | - Hui Chen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.); (C.A.); (A.A.S.)
| |
Collapse
|
49
|
Miller PG, Sperling AS, Mayerhofer C, McConkey ME, Ellegast JM, Da Silva C, Cohen DN, Wang C, Sharda A, Yan N, Saha S, Schluter C, Schechter I, Słabicki M, Sandoval B, Kahn J, Boettcher S, Gibson CJ, Scadden DT, Stegmaier K, Bhatt S, Lindsley RC, Ebert BL. PPM1D modulates hematopoietic cell fitness and response to DNA damage and is a therapeutic target in myeloid malignancy. Blood 2023; 142:2079-2091. [PMID: 37595362 PMCID: PMC10733824 DOI: 10.1182/blood.2023020331] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/05/2023] [Accepted: 07/20/2023] [Indexed: 08/20/2023] Open
Abstract
PPM1D encodes a phosphatase that is recurrently activated across cancer, most notably in therapy-related myeloid neoplasms. However, the function of PPM1D in hematopoiesis and its contribution to tumor cell growth remain incompletely understood. Using conditional mouse models, we uncover a central role for Ppm1d in hematopoiesis and validate its potential as a therapeutic target. We find that Ppm1d regulates the competitive fitness and self-renewal of hematopoietic stem cells (HSCs) with and without exogenous genotoxic stresses. We also show that although Ppm1d activation confers cellular resistance to cytotoxic therapy, it does so to a lesser degree than p53 loss, informing the clonal competition phenotypes often observed in human studies. Notably, loss of Ppm1d sensitizes leukemias to cytotoxic therapies in vitro and in vivo, even in the absence of a Ppm1d mutation. Vulnerability to PPM1D inhibition is observed across many cancer types and dependent on p53 activity. Importantly, organism-wide loss of Ppm1d in adult mice is well tolerated, supporting the tolerability of pharmacologically targeting PPM1D. Our data link PPM1D gain-of-function mutations to the clonal expansion of HSCs, inform human genetic observations, and support the therapeutic targeting of PPM1D in cancer.
Collapse
Affiliation(s)
- Peter G. Miller
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Adam S. Sperling
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Christina Mayerhofer
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| | - Marie E. McConkey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Jana M. Ellegast
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Carmen Da Silva
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Drew N. Cohen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Chuqi Wang
- National University of Singapore, Singapore
| | - Azeem Sharda
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Ni Yan
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Subha Saha
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Cameron Schluter
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Ilexa Schechter
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Mikołaj Słabicki
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Brittany Sandoval
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Josephine Kahn
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Steffen Boettcher
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Christopher J. Gibson
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - David T. Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA
- Department of Stem Cell and Regenerative Biology, Harvard University, Boston, MA
- Ludwig Center at Harvard, Boston, MA
| | - Kimberly Stegmaier
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - R. Coleman Lindsley
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Benjamin L. Ebert
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Howard Hughes Medical Institute, Bethesda, MD
| |
Collapse
|
50
|
Shah OS, Chen F, Wedn A, Kashiparekh A, Knapick B, Chen J, Savariau L, Clifford B, Hooda J, Christgen M, Xavier J, Oesterreich S, Lee AV. Multi-omic characterization of ILC and ILC-like cell lines as part of ILC cell line encyclopedia (ICLE) defines new models to study potential biomarkers and explore therapeutic opportunities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559548. [PMID: 37808708 PMCID: PMC10557671 DOI: 10.1101/2023.09.26.559548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Invasive lobular carcinoma (ILC), the most common histological "special type", accounts for ∼10-15% of all BC diagnoses, is characterized by unique features such as E-cadherin loss/deficiency, lower grade, hormone receptor positivity, larger diffuse tumors, and specific metastatic patterns. Despite ILC being acknowledged as a disease with distinct biology that necessitates specialized and precision medicine treatments, the further exploration of its molecular alterations with the goal of discovering new treatments has been hindered due to the scarcity of well-characterized cell line models for studying this disease. To address this, we generated the ILC Cell Line Encyclopedia (ICLE), providing a comprehensive multi-omic characterization of ILC and ILC-like cell lines. Using consensus multi-omic subtyping, we confirmed luminal status of previously established ILC cell lines and uncovered additional ILC/ILC-like cell lines with luminal features for modeling ILC disease. Furthermore, most of these luminal ILC/ILC-like cell lines also showed RNA and copy number similarity to ILC patient tumors. Similarly, ILC/ILC-like cell lines also retained molecular alterations in key ILC genes at similar frequency to both primary and metastatic ILC tumors. Importantly, ILC/ILC-like cell lines recapitulated the CDH1 alteration landscape of ILC patient tumors including enrichment of truncating mutations in and biallelic inactivation of CDH1 gene. Using whole-genome optical mapping, we uncovered novel genomic-rearrangements including novel structural variations in CDH1 and functional gene fusions and characterized breast cancer specific patterns of chromothripsis in chromosomes 8, 11 and 17. In addition, we systematically analyzed aberrant DNAm events and integrative analysis with RNA expression revealed epigenetic activation of TFAP2B - an emerging biomarker of lobular disease that is preferentially expressed in lobular disease. Finally, towards the goal of identifying novel druggable vulnerabilities in ILC, we analyzed publicly available RNAi loss of function breast cancer cell line datasets and revealed numerous putative vulnerabilities cytoskeletal components, focal adhesion and PI3K/AKT pathway in ILC/ILC-like vs NST cell lines. In summary, we addressed the lack of suitable models to study E-cadherin deficient breast cancers by first collecting both established and putative ILC models, then characterizing them comprehensively to show their molecular similarity to patient tumors along with uncovering their novel multi-omic features as well as highlighting putative novel druggable vulnerabilities. Not only we expand the array of suitable E-cadherin deficient cell lines available for modelling human-ILC disease but also employ them for studying epigenetic activation of a putative lobular biomarker as well as identifying potential druggable vulnerabilities for this disease towards enabling precision medicine research for human-ILC.
Collapse
|