1
|
Yang WC, Wei MF, Lee YH, Huang CS, Kuo SH. Radiosensitizing effects of CDK4/6 inhibitors in hormone receptor-positive and HER2-negative breast cancer mediated downregulation of DNA repair mechanism and NF-κB-signaling pathway. Transl Oncol 2024; 49:102092. [PMID: 39153367 PMCID: PMC11381799 DOI: 10.1016/j.tranon.2024.102092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 07/05/2024] [Accepted: 08/11/2024] [Indexed: 08/19/2024] Open
Abstract
CDK4/6 inhibitors combined with endocrine therapy prolonged survival in hormone receptor (HR)-positive and HER2-negative advanced breast cancer. We investigated whether CDK4/6 inhibitors enhance radiosensitivity and their underlying mechanisms of this subtype of breast cancer. In vitro and in vivo experiments were conducted using two HR-positive and HER2-negative breast cancer cell lines (MCF-7 and T-47D), CDK4/6 inhibitors (ribociclib and palbociclib) and radiotherapy (RT) to assess the biological functions and mechanisms. The radiation-enhancing effect was assessed using clonogenic assays; γH2AX and 53BP1 levels were assessed by immunofluorescence to evaluate DNA damage. The levels of phospho (p)-ERK, c-Myc, and DNA-double strand break (DSB)-related molecules, p-DNA-PKcs, Rad51, and p-ATM, were assessed by western blotting. We used an NF-κB p65 transcription factor assay kit to evaluate NF-κB activity. We evaluated the antitumor effect of the combination of RT and ribociclib through the MCF-7 orthotopic xenograft model. The synergistic effects of combining RT with ribociclib and palbociclib pretreatment were demonstrated by clonogenic assay. CDK4/6 inhibitors synergistically increased the numbers of RT-induced γH2AX and 53BP1, downregulated the expression of p-DNA-PKcs, Rad51 and p-ATM activated by RT, and reduced RT-triggering p-ERK expression, NF-κB activation, and its down-streaming gene, c-Myc. Combined ribociclib and RT reduced the growth of MCF-7 cell xenograft tumors, and downregulated the immunohistochemical expression of p-ERK, p-NF-κB p65, and c-Myc compared to that in the control group. Combining CDK4/6 inhibitors enhanced radiosensitivity of HR-positive and HER2-negative breast cancer cells at least by reducing DNA-DSB repair and weakening the activation of ERK and NF-κB signaling by RT.
Collapse
Affiliation(s)
- Wen-Chi Yang
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Radiation Oncology, National Taiwan University Cancer Center and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ming-Feng Wei
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chiun-Sheng Huang
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sung-Hsin Kuo
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan; Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
2
|
Lu K, Zhang M, Qin H, Shen S, Song H, Jiang H, Zhang C, Xiao G, Tong L, Jiang Q, Chen D. Disruption of cyclin D1 degradation leads to the development of mantle cell lymphoma. Acta Pharm Sin B 2024; 14:2977-2991. [PMID: 39027231 PMCID: PMC11252481 DOI: 10.1016/j.apsb.2024.03.013] [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: 11/23/2023] [Revised: 01/12/2024] [Accepted: 02/28/2024] [Indexed: 07/20/2024] Open
Abstract
Cyclin D1 has been recognized as an oncogene due to its abnormal upregulation in different types of cancers. Here, we demonstrated that cyclin D1 is SUMOylated, and we identified Itch as a specific E3 ligase recognizing SUMOylated cyclin D1 and mediating SUMO-induced ubiquitination and proteasome degradation of cyclin D1. We generated cyclin D1 mutant mice with mutations in the SUMOylation site, phosphorylation site, or both sites of cyclin D1, and found that double mutant mice developed a Mantle cell lymphoma (MCL)-like phenotype. We showed that arsenic trioxide (ATO) enhances cyclin D1 SUMOylation-mediated degradation through inhibition of cyclin D1 deSUMOylation enzymes, leading to MCL cell apoptosis. Treatment of severe combined immunodeficiency (SCID) mice grafted with MCL cells with ATO resulted in a significant reduction in tumor growth. In this study, we provide novel insights into the mechanisms of MCL tumor development and cyclin D1 regulation and discover a new strategy for MCL treatment.
Collapse
Affiliation(s)
- Ke Lu
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen 518055, China
| | - Ming Zhang
- Oncology Department, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hongyu Qin
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen 518055, China
- Division of Spine Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Siyu Shen
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China
| | - Haiqing Song
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Hua Jiang
- Division of Spine Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Chunxiang Zhang
- Department of Cardiology, Basic Medicine Innovation Center for Cardiometabolic Diseases of Ministry of Education, Institute of Cardiovascular Research, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Guozhi Xiao
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen 518055, China
| |
Collapse
|
3
|
Huang CH, Khan P, Xu S, Cohen J, Georgakis GV, Turkman N. Development of a Radiolabeled Cyclin-Dependent Kinases 4 and 6 (CDK4/6) Inhibitor for Brain and Cancer PET Imaging. Int J Mol Sci 2024; 25:6870. [PMID: 38999983 PMCID: PMC11241330 DOI: 10.3390/ijms25136870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
The synthesis, biochemical evaluation and radiosynthesis of a cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitor and radioligand was performed. NT431, a newly synthesized 4-fluorobenzyl-abemaciclib, exhibited high potency to CDK4/6 and against four cancer cell lines with IC50 similar to that of the parent abemaciclib. We performed a two-step one-pot radiosynthesis to produce [18F]NT431 with good radiochemical yield (9.6 ± 3%, n = 3, decay uncorrected), high radiochemical purity (>95%), and high molar activity (>370 GBq/µmol (>10.0 Ci/µmol). In vitro autoradiography confirmed the specific binding of [18F]NT431 to CDK4/6 in brain tissues. Dynamic PET imaging supports that both [18F]NT431 and the parent abemaciclib crossed the BBB albeit with modest brain uptake. Therefore, we conclude that it is unlikely that NT431 or abemaciclib (FDA approved drug) can accumulate in the brain in sufficient concentrations to be potentially effective against breast cancer brain metastases or brain cancers. However, despite the modest BBB penetration, [18F]NT431 represents an important step towards the development and evaluation of a new generation of CDK4/6 inhibitors with superior BBB penetration for the treatment and visualization of CDK4/6 positive tumors in the CNS. Also, [18F]NT431 may have potential application in peripheral tumors such as breast cancer and other CDK4/6 positive tumors.
Collapse
Affiliation(s)
- Chun-Han Huang
- Stony Brook Cancer Center, Stony Brook, Long Island, NY 11794, USA
- Department of Radiology, School of Medicine, Stony Brook University, Long Island, NY 11794, USA
- Department of Biomedical Engineering, Stony Brook University, Long Island, NY 11794, USA
| | - Palwasha Khan
- Stony Brook Cancer Center, Stony Brook, Long Island, NY 11794, USA
- Department of Radiology, School of Medicine, Stony Brook University, Long Island, NY 11794, USA
| | - Sulan Xu
- Stony Brook Cancer Center, Stony Brook, Long Island, NY 11794, USA
- Department of Radiology, School of Medicine, Stony Brook University, Long Island, NY 11794, USA
| | - Jules Cohen
- Stony Brook Cancer Center, Stony Brook, Long Island, NY 11794, USA
- Department of Medicine, School of Medicine, Stony Brook University, Long Island, NY 11794, USA
| | - Georgios V Georgakis
- Stony Brook Cancer Center, Stony Brook, Long Island, NY 11794, USA
- Department of Surgery, School of Medicine, Stony Brook University, Long Island, NY 11794, USA
| | - Nashaat Turkman
- Stony Brook Cancer Center, Stony Brook, Long Island, NY 11794, USA
- Department of Radiology, School of Medicine, Stony Brook University, Long Island, NY 11794, USA
- Department of Biomedical Engineering, Stony Brook University, Long Island, NY 11794, USA
| |
Collapse
|
4
|
Saleban M, Harris EL, Poulter JA. D-Type Cyclins in Development and Disease. Genes (Basel) 2023; 14:1445. [PMID: 37510349 PMCID: PMC10378862 DOI: 10.3390/genes14071445] [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: 06/17/2023] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
D-type cyclins encode G1/S cell cycle checkpoint proteins, which play a crucial role in defining cell cycle exit and progression. Precise control of cell cycle exit is vital during embryonic development, with defects in the pathways regulating intracellular D-type cyclins resulting in abnormal initiation of stem cell differentiation in a variety of different organ systems. Furthermore, stabilisation of D-type cyclins is observed in a wide range of disorders characterized by cellular over-proliferation, including cancers and overgrowth disorders. In this review, we will summarize and compare the roles played by each D-type cyclin during development and provide examples of how their intracellular dysregulation can be an underlying cause of disease.
Collapse
Affiliation(s)
- Mostafa Saleban
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Erica L Harris
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - James A Poulter
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
5
|
Beddok A, Porte B, Cottu P, Fourquet A, Kirova Y. [Biological, preclinical and clinical aspects of the association between radiation therapy and CDK4/6 inhibitors]. Cancer Radiother 2023; 27:240-248. [PMID: 37080859 DOI: 10.1016/j.canrad.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 10/06/2022] [Accepted: 11/30/2022] [Indexed: 04/22/2023]
Abstract
Several clinical studies have shown that CDK4/6 inhibitors (CDK4/6i) improve survival in patients with metastatic or locally advanced HR-positive, HER-2-negative breast cancer (BC). The aim of this review was to synthesize the biological, preclinical and clinical aspects of the treatment of BC with CDK4/6i, with a focus on the combination of CDK4/6i and radiotherapy. The DNA damage induced after exposure of cells to ionizing radiation activates control pathways that inhibit cell progression in the G1 and G2 phases and induce a transient delay in progression in the S phase. These checkpoints are in particular mediated by cyclin-dependent kinases (CDK) 4/6 activated by cyclin D1. Several preclinical studies have shown that CDK4/6i could be used as radiosensitizers in non-small cell lung cancer, medulloblastoma, brainstem glioma and breast cancer. CDK4/6 inhibition also protected against radiation-induced intestinal toxicities by inducing redistribution of quiescent intestinal progenitor cells, making them less radiosensitive. Clinical data on the combination of CDK inhibitors and radiotherapy for both locoregional and metastatic irradiation are based on retrospective data. Nevertheless, the most optimal therapeutic sequence would be radiotherapy followed by palbociclib. Pending prospective clinical trials, the concomitant combination of the two treatments should be done under close supervision.
Collapse
Affiliation(s)
- A Beddok
- Institut Curie, PSL Research University, University Paris Saclay, Inserm LITO, 91898 Orsay, France; Institut Curie, PSL Research University, Radiation Oncology Department, Proton Therapy Centre, Centre Universitaire, 91898 Orsay, France.
| | - B Porte
- Service d'oncologie médicale, GHU hôpital européen Georges-Pompidou, Paris, France
| | - P Cottu
- Département d'oncologie médicale, Institut Curie, Paris, France
| | - A Fourquet
- Institut Curie, PSL Research University, Radiation Oncology Department, Paris, France
| | - Y Kirova
- Institut Curie, PSL Research University, Radiation Oncology Department, Paris, France
| |
Collapse
|
6
|
Galogre M, Rodin D, Pyatnitskiy M, Mackelprang M, Koman I. "A Review of HER2 overexpression and somatic mutations in cancers". Crit Rev Oncol Hematol 2023; 186:103997. [PMID: 37062337 DOI: 10.1016/j.critrevonc.2023.103997] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/14/2023] [Accepted: 04/13/2023] [Indexed: 04/18/2023] Open
Abstract
The Human Epidermal Growth Factor Receptor (HER) proteins family, which includes HER2, are membrane-bound receptors that activate many intracellular pathways associated with growth and development. When there are mutations in HER2, or when it becomes overexpressed, it can cause oncogenesis and offer differential prognosis and treatment across almost all cancer types. Both mutations in HER2 and its overexpression have distinct mechanisms by which they can cause these effects in cancers. This review outlines how HER2's normal pathway is altered in both overexpression and mutation and compiles all the well-known mechanisms by which HER2 can cause oncogenesis. Finally, this review briefly outlines how HER2 mutants and HER2 overexpression is detected, and how their detection can lead to different prognosis and treatment in cancers.
Collapse
Affiliation(s)
| | - Dmitry Rodin
- Institute of Personalised and Translational Medicine, Ariel University, Ariel, Israel Kiryat Hamada
| | - Mikhail Pyatnitskiy
- Institute of Biomedical Chemistry RAMS, Solianka st.,14, 109544, Moscow, Russia
| | | | - Igor Koman
- SmartOmica, Tērbatas iela 36 - 4, Latvia Rīga, LV-1011; Institute of Personalised and Translational Medicine, Ariel University, Ariel, Israel Kiryat Hamada
| |
Collapse
|
7
|
Wang J, Su W, Zhang T, Zhang S, Lei H, Ma F, Shi M, Shi W, Xie X, Di C. Aberrant Cyclin D1 splicing in cancer: from molecular mechanism to therapeutic modulation. Cell Death Dis 2023; 14:244. [PMID: 37024471 PMCID: PMC10079974 DOI: 10.1038/s41419-023-05763-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023]
Abstract
Cyclin D1 (CCND1), a crucial mediator of cell cycle progression, possesses many mutation types with different mutation frequencies in human cancers. The G870A mutation is the most common mutation in CCND1, which produces two isoforms: full-length CCND1a and divergent C-terminal CCND1b. The dysregulation of the CCND1 isoforms is associated with multiple human cancers. Exploring the molecular mechanism of CCND1 isoforms has offer new insight for cancer treatment. On this basis, the alterations of CCND1 gene are described, including amplification, overexpression, and mutation, especially the G870A mutation. Subsequently, we review the characteristics of CCND1 isoforms caused by G870A mutation. Additionally, we summarize cis-regulatory elements, trans-acting factors, and the splice mutation involved in splicing regulation of CCND1. Furthermore, we highlight the function of CCND1 isoforms in cell cycle, invasion, and metastasis in cancers. Importantly, the clinical role of CCND1 isoforms is also discussed, particularly concerning prognosis, chemotherapy, and radiotherapy. Last, emphasis is given to the corrective strategies that modulate the cancerous CCND1 isoforms. Thus, it is highlighting significance of aberrant isoforms of CCND1 as targets for cancer therapy.
Collapse
Affiliation(s)
- Jing Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Wei Su
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Taotao Zhang
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Shasha Zhang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Huiwen Lei
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Fengdie Ma
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Maoning Shi
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Wenjing Shi
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Xiaodong Xie
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Cuixia Di
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China.
| |
Collapse
|
8
|
Matherne MG, Phillips ES, Embrey SJ, Burke CM, Machado HL. Emerging functions of C/EBPβ in breast cancer. Front Oncol 2023; 13:1111522. [PMID: 36761942 PMCID: PMC9905667 DOI: 10.3389/fonc.2023.1111522] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023] Open
Abstract
Breast tumorigenesis relies on complex interactions between tumor cells and their surrounding microenvironment, orchestrated by tightly regulated transcriptional networks. C/EBPβ is a key transcription factor that regulates the proliferation and differentiation of multiple cell types and modulates a variety of biological processes such as tissue homeostasis and the immune response. In addition, C/EBPβ has well-established roles in mammary gland development, is overexpressed in breast cancer, and has tumor-promoting functions. In this review, we discuss context-specific roles of C/EBPβ during breast tumorigenesis, isoform-specific gene regulation, and regulation of the tumor immune response. We present challenges in C/EBPβ biology and discuss the importance of C/EBPβ isoform-specific gene regulation in devising new therapeutic strategies.
Collapse
Affiliation(s)
- Megan G. Matherne
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, United States
| | - Emily S. Phillips
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, United States
| | - Samuel J. Embrey
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, United States
| | - Caitlin M. Burke
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, United States
| | - Heather L. Machado
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, United States,Tulane Cancer Center, Louisiana Cancer Research Consortium, New Orleans, LA, United States,*Correspondence: Heather L. Machado,
| |
Collapse
|
9
|
Baker SJ, Poulikakos PI, Irie HY, Parekh S, Reddy EP. CDK4: a master regulator of the cell cycle and its role in cancer. Genes Cancer 2022; 13:21-45. [PMID: 36051751 PMCID: PMC9426627 DOI: 10.18632/genesandcancer.221] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/17/2022] [Indexed: 11/25/2022] Open
Abstract
The cell cycle is regulated in part by cyclins and their associated serine/threonine cyclin-dependent kinases, or CDKs. CDK4, in conjunction with the D-type cyclins, mediates progression through the G1 phase when the cell prepares to initiate DNA synthesis. Although Cdk4-null mutant mice are viable and cell proliferation is not significantly affected in vitro due to compensatory roles played by other CDKs, this gene plays a key role in mammalian development and cancer. This review discusses the role that CDK4 plays in cell cycle control, normal development and tumorigenesis as well as the current status and utility of approved small molecule CDK4/6 inhibitors that are currently being used as cancer therapeutics.
Collapse
Affiliation(s)
- Stacey J. Baker
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Levy Place, NY 10029, USA
| | - Poulikos I. Poulikakos
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Levy Place, NY 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, Levy Place, NY 10029, USA
| | - Hanna Y. Irie
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Levy Place, NY 10029, USA
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Levy Place, NY 10029, USA
| | - Samir Parekh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, Levy Place, NY 10029, USA
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Levy Place, NY 10029, USA
| | - E. Premkumar Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Levy Place, NY 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, Levy Place, NY 10029, USA
| |
Collapse
|
10
|
Sammons S, Moore H, Cushman J, Hamilton E. Efficacy, safety and toxicity management of adjuvant abemaciclib in early stage HR+/HER2- high-risk breast cancer. Expert Rev Anticancer Ther 2022; 22:805-814. [PMID: 35737886 DOI: 10.1080/14737140.2022.2093719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The majority of the over 250,000 new cases of invasive breast cancer diagnosed in the United States is driven by hormone receptor signaling (HR+). Since 2015, cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) have become the standard in combination with endocrine therapy (ET) for patients facing metastatic disease. AREAS COVERED There are now three approved agents in the metastatic setting; abemaciclib, ribociclib, and palbociclib. Due to the almost doubling of progression free survival (PFS) and improvement in overall survival (OS) in the metastatic setting, studies were conducted to examine the benefit of adding CDK4/6i in the adjuvant setting for those patients at high risk for recurrence. Despite negative results of PALLAS (palbociclib) in this setting, monarchE (abemaciclib) showed an improvement in invasive disease-free survival (IDFS) and distant recurrence free survival (DRFS) at the 3 year time point for patients with high risk tumor characteristics leading to its approval. Herein, we discuss the data, the population studied and the population in which abemaciclib is approved as well as safety, tolerability, and dose reductions for practical management of these patients. EXPERT OPINION Abemaciclib is appropriate and beneficial for those patients with high-risk, node-positive, hormonally-driven breast cancer.
Collapse
Affiliation(s)
- Sarah Sammons
- Duke University Medical Center.,Duke Cancer Institute, 30 Duke Medicine Circle, Durham, North Carolina 27705-3827
| | - Heather Moore
- Duke University Medical Center.,Duke Cancer Institute, 30 Duke Medicine Circle, Durham, North Carolina 27705-3827
| | - Jaycee Cushman
- Duke University Medical Center.,Duke Cancer Institute, 30 Duke Medicine Circle, Durham, North Carolina 27705-3827
| | - Erika Hamilton
- Sarah Cannon Research Institute, Nashville, Tennessee 37203-1632.,Tennessee Oncology, Nashville, TN 37203
| |
Collapse
|
11
|
Miller JL, Bartlett AP, Harman RM, Majhi PD, Jerry DJ, Van de Walle GR. Induced mammary cancer in rat models: pathogenesis, genetics, and relevance to female breast cancer. J Mammary Gland Biol Neoplasia 2022; 27:185-210. [PMID: 35904679 DOI: 10.1007/s10911-022-09522-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 10/16/2022] Open
Abstract
Mammary cancer, or breast cancer in women, is a polygenic disease with a complex etiopathogenesis. While much remains elusive regarding its origin, it is well established that chemical carcinogens and endogenous estrogens contribute significantly to the initiation and progression of this disease. Rats have been useful models to study induced mammary cancer. They develop mammary tumors with comparable histopathology to humans and exhibit differences in resistance or susceptibility to mammary cancer depending on strain. While some rat strains (e.g., Sprague-Dawley) readily form mammary tumors following treatment with the chemical carcinogen, 7,12-dimethylbenz[a]-anthracene (DMBA), other strains (e.g., Copenhagen) are resistant to DMBA-induced mammary carcinogenesis. Genetic linkage in inbred strains has identified strain-specific quantitative trait loci (QTLs) affecting mammary tumors, via mechanisms that act together to promote or attenuate, and include 24 QTLs controlling the outcome of chemical induction, 10 QTLs controlling the outcome of estrogen induction, and 4 QTLs controlling the outcome of irradiation induction. Moreover, and based on shared factors affecting mammary cancer etiopathogenesis between rats and humans, including orthologous risk regions between both species, rats have served as useful models for identifying methods for breast cancer prediction and treatment. These studies in rats, combined with alternative animal models that more closely mimic advanced stages of breast cancer and/or human lifestyles, will further improve our understanding of this complex disease.
Collapse
Affiliation(s)
- James L Miller
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 14853, Ithaca, NY, USA
| | - Arianna P Bartlett
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 14853, Ithaca, NY, USA
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 14853, Ithaca, NY, USA
| | - Prabin Dhangada Majhi
- Department of Veterinary & Animal Sciences, University of Massachusetts, 01003, Amherst, MA, USA
| | - D Joseph Jerry
- Department of Veterinary & Animal Sciences, University of Massachusetts, 01003, Amherst, MA, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 14853, Ithaca, NY, USA.
| |
Collapse
|
12
|
Valla M, Klæstad E, Ytterhus B, Bofin AM. CCND1 Amplification in Breast Cancer -associations With Proliferation, Histopathological Grade, Molecular Subtype and Prognosis. J Mammary Gland Biol Neoplasia 2022; 27:67-77. [PMID: 35459982 PMCID: PMC9135839 DOI: 10.1007/s10911-022-09516-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/05/2022] [Indexed: 11/25/2022] Open
Abstract
CCND1 is located on 11q13. Increased CCND1 copy number (CN) in breast cancer (BC) is associated with high histopathological grade, high proliferation, and Luminal B subtype. In this study of CCND1 in primary BCs and corresponding axillary lymph node metastases (LNM),we examine associations between CCND1 CN in primary BCs and proliferation status, molecular subtype, and prognosis. Furthermore, we studied associations between CCND1 CN and CNs of FGFR1 and ZNF703, both of which are located on 8p12. Fluorescence in situ hybridization probes for CCND1 and chromosome 11 centromere were used on tissue microarrays comprising 526 BCs and 123 LNM. We assessed associations between CCND1 CN and tumour characteristics using Pearson's χ2 test, and estimated cumulative risks of death from BC and hazard ratios in analysis of prognosis. We found CCND1 CN ≥ 4 < 6 in 45 (8.6%) tumours, and ≥ 6 in 42 (8.0%). CCND1 CN (≥ 6) was seen in all molecular subtypes, most frequently in Luminal B (HER2-) (20/126; 16%). Increased CCND1 CN was associated with high histopathological grade, high Ki-67, and high mitotic count, but not prognosis. CCND1 CN ≥ 6 was accompanied by CN increase of FGFR1 in 6/40 cases (15.0%) and ZNF703 in 5/38 cases (13.2%). Three cases showed CN increase of all three genes. High CCND1 CN was most frequent in Luminal B (HER2-) tumours. Good correlation between CCND1 CNs in BCs and LNM was observed. Despite associations between high CCND1 CN and aggressive tumour characteristics, the prognostic impact of CCND1 CN remains unresolved.
Collapse
Affiliation(s)
- Marit Valla
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olav's Hospital, Trondheim University Hospital, 7006, Trondheim, Norway
| | - Elise Klæstad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Borgny Ytterhus
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna M Bofin
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
13
|
Bahrami A, Jafari A, Ferns GA. The dual role of microRNA-9 in gastrointestinal cancers: oncomiR or tumor suppressor? Biomed Pharmacother 2021; 145:112394. [PMID: 34781141 DOI: 10.1016/j.biopha.2021.112394] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/26/2022] Open
Abstract
microRNA are noncoding endogenous RNAs of ∼ 25-nucleotide, involved in RNA silencing and controlling of cell function. Recent evidence has highlighted the important role of various in the biology of human cancers. miR-9 is a highly conserved microRNA and abnormal regulation of miR-9 expression has various impacts on disease pathology. miR-9 may play a dual tumor-suppressive or oncomiR activity in several cancers. There have been conflicting reports concerning the role of miR-9 in gastrointestinal cancers. Several signaling pathways including PDK/AKT, Hippo, Wnt/β-catenin and PDGFRB axes are affected by miR-9 in suppressing proliferation, invasion and metastasis of tumor cells. Oncogenic miR-9 triggers migration, metastasis and clinic-pathological characteristics of patients with gastrointestinal malignancy by targeting various enzymes and transcription factors such as E-cadherin, HK2, LMX1A, and CDX2. On the other hand, long non-coding RNAs and circular RNAs can modulate miR-9 expression in human cancers. In this review, we aimed to summarize recent findings about the potential value of miR-9 in gastrointestinal tumors, that include: screening, prognostic and treatment.
Collapse
Affiliation(s)
- Afsane Bahrami
- Clinical Research Development Unit of Akbar Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amirsajad Jafari
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| |
Collapse
|
14
|
Sharma P, Tiufekchiev S, Lising V, Chung SW, Suk JS, Chung BM. Keratin 19 interacts with GSK3β to regulate its nuclear accumulation and degradation of cyclin D3. Mol Biol Cell 2021; 32:ar21. [PMID: 34406791 PMCID: PMC8693971 DOI: 10.1091/mbc.e21-05-0255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cyclin D3 regulates the G1/S transition and is frequently overexpressed in several cancer types including breast cancer, where it promotes tumor progression. Here we show that a cytoskeletal protein keratin 19 (K19) physically interacts with a serine/threonine kinase GSK3β and prevents GSK3β-dependent degradation of cyclin D3. The absence of K19 allowed active GSK3β to accumulate in the nucleus and degrade cyclin D3. Specifically, the head (H) domain of K19 was required to sustain inhibitory phosphorylation of GSK3β Ser9, prevent nuclear accumulation of GSK3β, and maintain cyclin D3 levels and cell proliferation. K19 was found to interact with GSK3β and K19–GSK3β interaction was mapped out to require Ser10 and Ser35 residues on the H domain of K19. Unlike wildtype K19, S10A and S35A mutants failed to maintain total and nuclear cyclin D3 levels and induce cell proliferation. Finally, we show that the K19–GSK3β-cyclin D3 pathway affected sensitivity of cells toward inhibitors to cyclin-dependent kinase 4 and 6 (CDK4/6). Overall, these findings establish a role for K19 in the regulation of GSK3β-cyclin D3 pathway and demonstrate a potential strategy for overcoming resistance to CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Pooja Sharma
- Department of Biology, The Catholic University of America, Washington, DC 20064
| | - Sarah Tiufekchiev
- Department of Biology, The Catholic University of America, Washington, DC 20064
| | - Victoria Lising
- Department of Biology, The Catholic University of America, Washington, DC 20064
| | - Seung Woo Chung
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231
| | - Jung Soo Suk
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231
| | - Byung Min Chung
- Department of Biology, The Catholic University of America, Washington, DC 20064
| |
Collapse
|
15
|
Collard M, Gallagher PE, Tallant EA. A Polyphenol-Rich Extract From Muscadine Grapes Inhibits Triple-Negative Breast Tumor Growth. Integr Cancer Ther 2021; 19:1534735420917444. [PMID: 32578460 PMCID: PMC7315667 DOI: 10.1177/1534735420917444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that tends to affect young women and has a high propensity to metastasize. No targeted treatments are available for this type of breast cancer due to a lack of estrogen or progesterone receptors or overexpression of human epidermal growth factor receptor type 2 overexpression. Currently, patients have no therapeutic options once standard of care is complete, indicating a need for safe and effective therapies to slow or prevent the progression of TNBC to metastatic disease. Studies showed that isolated polyphenols or polyphenol-rich muscadine grape extracts polyphenols inhibit the proliferation of various cancer cells including breast cancer. A proprietary muscadine grape extract (MGE) was administered to nude mice with human MDA-MB-231 TNBC atumors for 4 weeks to determine the effect of the extract on tumor growth. MGE decreased tumor volume in association with a reduction in the proliferative markers Ki67 and cyclin D1. To determine the molecular mechanisms for the MGE-induced reduction in tumor growth, mouse 4T1, MDA-MB-231, or human BT-549 TNBC cells were treated with MGE, and various signaling pathways were investigated. MGE reduced c-Met, differentially abrogated ERK/MAPK and AKT signaling, and decreased a downstream targets of ERK/MAPK and AKT pathways, cyclin D1. Cyclin D1 reduction was associated with retinoblastoma activation and cell cycle arrest in MDA-MB-231 TNBC cells. MGE-regulated molecular signaling pathways were functionally associated with a dose-dependent reduction in cell proliferation. The pluripotency of MGE and high index of safety and tolerability suggest that the extract may serve as a therapeutic to reduce TNBC progression to metastatic disease.
Collapse
Affiliation(s)
| | | | - E Ann Tallant
- Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
16
|
Komini C, Theohari I, Lambrianidou A, Nakopoulou L, Trangas T. PAPOLA contributes to cyclin D1 mRNA alternative polyadenylation and promotes breast cancer cell proliferation. J Cell Sci 2021; 134:237820. [PMID: 33712453 DOI: 10.1242/jcs.252304] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 02/26/2021] [Indexed: 12/16/2022] Open
Abstract
Poly(A) polymerases add the poly(A) tail at the 3' end of nearly all eukaryotic mRNA, and are associated with proliferation and cancer. To elucidate the role of the most-studied mammalian poly(A) polymerase, poly(A) polymerase α (PAPOLA), in cancer, we assessed its expression in 221 breast cancer samples and found it to correlate strongly with the aggressive triple-negative subtype. Silencing PAPOLA in MCF-7 and MDA-MB-231 breast cancer cells reduced proliferation and anchorage-independent growth by decreasing steady-state cyclin D1 (CCND1) mRNA and protein levels. Whereas the length of the CCND1 mRNA poly(A) tail was not affected, its 3' untranslated region (3'UTR) lengthened. Overexpressing PAPOLA caused CCND1 mRNA 3'UTR shortening with a concomitant increase in the amount of corresponding transcript and protein, resulting in growth arrest in MCF-7 cells and DNA damage in HEK-293 cells. Such overexpression of PAPOLA promoted proliferation in the p53 mutant MDA-MB-231 cells. Our data suggest that PAPOLA is a possible candidate target for the control of tumor growth that is mostly relevant to triple-negative tumors, a group characterized by PAPOLA overexpression and lack of alternative targeted therapies.
Collapse
Affiliation(s)
- Chrysoula Komini
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, 45110, Greece
| | - Irini Theohari
- First Department of Pathology, Medical School, University of Athens, Athens, 11517, Greece
| | - Andromachi Lambrianidou
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, 45110, Greece
| | - Lydia Nakopoulou
- First Department of Pathology, Medical School, University of Athens, Athens, 11517, Greece
| | - Theoni Trangas
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, 45110, Greece
| |
Collapse
|
17
|
Sex-Specific Role for SLIT1 in Regulating Stress Susceptibility. Biol Psychiatry 2021; 91:81-91. [PMID: 33896623 PMCID: PMC8390577 DOI: 10.1016/j.biopsych.2021.01.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/22/2020] [Accepted: 01/06/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Major depressive disorder is a pervasive and debilitating syndrome characterized by mood disturbances, anhedonia, and alterations in cognition. While the prevalence of major depressive disorder is twice as high for women as men, little is known about the molecular mechanisms that drive sex differences in depression susceptibility. METHODS We discovered that SLIT1, a secreted protein essential for axonal navigation and molecular guidance during development, is downregulated in the adult ventromedial prefrontal cortex (vmPFC) of women with depression compared with healthy control subjects, but not in men with depression. This sex-specific downregulation of Slit1 was also observed in the vmPFC of mice exposed to chronic variable stress. To identify a causal, sex-specific role for SLIT1 in depression-related behavioral abnormalities, we performed knockdown (KD) of Slit1 expression in the vmPFC of male and female mice. RESULTS When combined with stress exposure, vmPFC Slit1 KD reflected the human condition by inducing a sex-specific increase in anxiety- and depression-related behaviors. Furthermore, we found that vmPFC Slit1 KD decreased the dendritic arborization of vmPFC pyramidal neurons and decreased the excitability of the neurons in female mice, effects not observed in males. RNA sequencing analysis of the vmPFC after Slit1 KD in female mice revealed an augmented transcriptional stress signature. CONCLUSIONS Together, our findings establish a crucial role for SLIT1 in regulating neurophysiological and transcriptional responses to stress within the female vmPFC and provide mechanistic insight into novel signaling pathways and molecular factors influencing sex differences in depression susceptibility.
Collapse
|
18
|
Liu Y, Zhang A, Bao PP, Lin L, Wang Y, Wu H, Shu XO, Liu A, Cai Q. MicroRNA-374b inhibits breast cancer progression through regulating CCND1 and TGFA genes. Carcinogenesis 2021; 42:528-536. [PMID: 33480984 DOI: 10.1093/carcin/bgab005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/11/2021] [Accepted: 01/18/2021] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence indicates that microRNAs (miRNAs) play a critical role in breast cancer development. We recently reported that a higher expression of miR-374b in tumor tissues was associated with a better disease-free survival of triple-negative breast cancer (TNBC). However, the functional significance and molecular mechanisms underlying the role of miR-374b in breast cancer are largely unknown. In this current study, we evaluated the biological functions and potential mechanisms of miR-374b in both TNBC and non-TNBC. We found that miR-374b was significantly downregulated in breast cancer tissues, compared to adjacent tissues. MiR-374b levels were also lower in breast cancer cell lines, as compared to breast epithelial cells. In vitro and in vivo studies demonstrated that miR-374b modulates the malignant behavior of breast cancer cells, such as cell proliferation in 2D and 3D, cell invasion ability, colony-forming ability and tumor growth in mice. By using bioinformatics tools, we predicted that miR-374b plays a role in breast cancer cells through negatively regulating cyclin D1 (CCND1) and transforming growth factor alpha (TGFA). We further confirmed that CCND1 and TGFA contribute to the malignant behavior of breast cancer cells in vitro and in vivo. Our rescue experiments showed that overexpressing CCND1 or TGFA reverses the phenotypes caused by miR-374b overexpression. Taken together, our studies suggest that miR-374b modulates malignant behavior of breast cancer cells by negatively regulating CCND1 and TGFA genes. The newly identified miR-374b-mediated CCND1 and TGFA gene silencing may facilitate a better understanding of the molecular mechanisms of breast cancer progression.
Collapse
Affiliation(s)
- Yan Liu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA.,Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Ai Zhang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, P.R. China
| | - Ping-Ping Bao
- Shanghai Municipal Center for Disease Control and Prevention, Shanghai 200336, P.R. China
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, P.R. China
| | - Yina Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, P.R. China
| | - Haijian Wu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA.,Department of Radiation Oncology, Qi-lu Hospital affiliated to Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Aiguo Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, P.R. China
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| |
Collapse
|
19
|
Wang C, Zhang R, Wang X, Zheng Y, Jia H, Li H, Wang J, Wang N, Xiang F, Li Y. Silencing of KIF3B Suppresses Breast Cancer Progression by Regulating EMT and Wnt/ β-Catenin Signaling. Front Oncol 2021; 10:597464. [PMID: 33542902 PMCID: PMC7851081 DOI: 10.3389/fonc.2020.597464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/30/2020] [Indexed: 01/06/2023] Open
Abstract
Breast cancer is the most common malignant tumors in women. Kinesin family member 3B (KIF3B) is a critical regulator in mitotic progression. The objective of this study was to explore the expression, regulation, and mechanism of KIF3B in 103 cases of breast cancer tissues, 35 metastatic lymph nodes and breast cancer cell lines, including MDA-MB-231, MDA-MB-453, T47D, and MCF-7. The results showed that KIF3B expression was up-regulated in breast cancer tissues and cell lines, and the expression level was correlated with tumor recurrence and lymph node metastasis, while knockdown of KIF3B suppressed cell proliferation, migration, and invasion both in vivo and in vitro. In addition, UALCAN analysis showed that KIF3B expression in breast cancer is increased, and the high expression of KIF3B in breast cancer is associated with poor prognosis. Furthermore, we found that silencing of KIF3B decreased the expression of Dvl2, phospho-GSK-3β, total and nucleus β-catenin, then subsequent down-regulation of Wnt/β-catenin signaling target genes such as CyclinD1, C-myc, MMP-2, MMP-7 and MMP-9 in breast cancer cells. In addition, KIF3B depletion inhibited epithelial mesenchymal transition (EMT) in breast cancer cells. Taken together, our results revealed that KIF3B is up-regulated in breast cancer which is potentially involved in breast cancer progression and metastasis. Silencing KIF3B might suppress the Wnt/β-catenin signaling pathway and EMT in breast cancer cells.
Collapse
Affiliation(s)
- Chengqin Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Runze Zhang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiao Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Zheng
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Huiqing Jia
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haiyan Li
- Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, Qingdao, China
| | - Jin Wang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Ning Wang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Fenggang Xiang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yujun Li
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
20
|
Gomatou G, Trontzas I, Ioannou S, Drizou M, Syrigos N, Kotteas E. Mechanisms of resistance to cyclin-dependent kinase 4/6 inhibitors. Mol Biol Rep 2021; 48:915-925. [PMID: 33409716 DOI: 10.1007/s11033-020-06100-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022]
Abstract
Cyclin-dependent kinase (CDK) 4/6 inhibitors have emerged in the treatment of metastatic hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-negative breast cancer. However, most patients will eventually present disease progression, highlighting the inevitable resistance of cancer cells to CDK4/6 inhibition. Several studies have suggested that resistance mechanisms involve aberrations of the molecules that regulate the cell cycle, and the re-wiring of the cell to escape CDK4/6 dependence and turn to alternative pathways. Loss of retinoblastoma function, overexpression of CDK 6, upregulation of cyclin E, overexpression of CDK 7, and dysregulation of several signaling pathways, notably the PI3/AKT/mTOR pathway, have been implicated in the development of resistance to CDK4/6 inhibitors. Overlap with endocrine resistance mechanisms might be possible. Combinational therapeutic strategies should be explored in order to prevent resistance and optimize the management of patients after progression under CDK 4/6 inhibition.
Collapse
Affiliation(s)
- Georgia Gomatou
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - Ioannis Trontzas
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stephanie Ioannou
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Drizou
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Syrigos
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Elias Kotteas
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
21
|
Saeidi S, Joo S, Kim SJ, Jagadeesh ASV, Surh YJ. Interaction between Peptidyl-prolyl Cis- trans Isomerase NIMA-interacting 1 and GTP-H-Ras: Implications for Aggressiveness of Human Mammary Epithelial Cells and Drug Resistance. J Cancer Prev 2020; 25:234-243. [PMID: 33409256 PMCID: PMC7783236 DOI: 10.15430/jcp.2020.25.4.234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/08/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Aberrant activation of Ras has been implicated in aggressiveness of breast cancer. Among Ras isoforms (H-, K-, and N-), H-Ras has been known to be primarily responsible for invasion and metastasis of breast cancer cells. Phosphorylation of serine (Ser) or threonine (Thr) is a key regulatory mechanism responsible for controlling activities and functions of various proteins involved in intracellular signal transduction. Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1, Pin1 changes the conformation of a subset of proteins phosphorylated on Ser/Thr that precedes proline (Pro). In this study we have found that Pin1 is highly overexpressed in human breast tumor tissues and H-Ras transformed human mammary epithelial (H-Ras MCF10A) and MDA-MB-231 breast cancer cells. Notably, Pin1 directly bound to the activated form of H-Ras harbouring a Ser/Thr-Pro motif. Pharmacologic inhibition of Pin1 reduced clonogenicity of MDA-MB-231 human breast cancer cells. Paclitaxel accelerates apoptosis in Pin1 silenced H-Ras MCF10A cells. MDR genes (MDR1 and MRP4) were significantly downregulated in MDA-MB-231 cells stably silenced for Pin1. We speculate that Pin1 interacts with GTP-H-Ras, thereby upregulating the expression of drug resistance genes, which confers survival advantage and aggressiveness of breast cancer cells under chemotherapy.
Collapse
Affiliation(s)
- Soma Saeidi
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.,Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Sihyung Joo
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Su-Jung Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Achanta Sri Venkata Jagadeesh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Young-Joon Surh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.,Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, Korea.,College of Pharmacy, Seoul National University, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
22
|
Scheiblecker L, Kollmann K, Sexl V. CDK4/6 and MAPK-Crosstalk as Opportunity for Cancer Treatment. Pharmaceuticals (Basel) 2020; 13:E418. [PMID: 33255177 PMCID: PMC7760252 DOI: 10.3390/ph13120418] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the development of targeted therapies and novel inhibitors, cancer remains an undefeated disease. Resistance mechanisms arise quickly and alternative treatment options are urgently required, which may be partially met by drug combinations. Protein kinases as signaling switchboards are frequently deregulated in cancer and signify vulnerable nodes and potential therapeutic targets. We here focus on the cell cycle kinase CDK6 and on the MAPK pathway and on their interplay. We also provide an overview on clinical studies examining the effects of combinational treatments currently explored for several cancer types.
Collapse
Affiliation(s)
| | | | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (L.S.); (K.K.)
| |
Collapse
|
23
|
Neven P, Sonke GS, Jerusalem G. Ribociclib plus fulvestrant in the treatment of breast cancer. Expert Rev Anticancer Ther 2020; 21:93-106. [PMID: 33085548 DOI: 10.1080/14737140.2021.1840360] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Endocrine therapy (ET) is a standard first-line treatment for hormone receptor-positive, human epidermal growth factor receptor 2-negative (HR+/HER2-) advanced breast cancer (ABC) Cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) have demonstrated significantly improved progression-free survival (PFS) with ET in patients with ABC. Recent reports indicate that the addition of the CDK4/6i ribociclib to ET, including fulvestrant, significantly improves PFS and overall survival (OS). AREAS COVERED This review summarizes the efficacy and safety of ribociclib plus fulvestrant in HR+/HER2- ABC and its role in clinical practice. Various post-progression strategies are discussed. EXPERT OPINION In MONALEESA-3, ribociclib +fulvestrant significantly improved PFS and OS in postmenopausal patients who received no prior chemotherapy and ≤1 prior line of ET for ABC and benefited many patient subgroups, including those with visceral metastases and ET resistance. The safety of this combination is manageable and consistent with the known safety profile of ribociclib, with myelosuppression being a common and expected toxicity; other relevant toxicities requiring monitoring that occur at a low rate include hepatobiliary toxicity, pneumonitis, and QTc prolongation. There is an important role for CDK4/6i + ET, including ribociclib + fulvestrant, in clinical practice. The optimal position of CDK4/6i in first or subsequent lines of treatment and the optimal post-CDK4/6i progression strategies are not yet elucidated.
Collapse
Affiliation(s)
- Patrick Neven
- Multidisciplinary Breast Center, Universitair Ziekenhuis Leuven , Leuven, Belgium
| | - Gabe S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute , Amsterdam, The Netherlands
| | | |
Collapse
|
24
|
Liu D, Li P, Wang X, Wang W. hsa-miR-195-5p inhibits cell proliferation of human thyroid carcinoma cells via modulation of p21/cyclin D1 axis. Transl Cancer Res 2020; 9:5190-5199. [PMID: 35117886 PMCID: PMC8799136 DOI: 10.21037/tcr-20-1083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022]
Abstract
Background Based on existing evidence, microRNAs (miRs) are gene regulators that undertake key functions in the oncogenesis and tumor progression of every single human malignant disease, such as thyroid carcinoma (TC). Previous clinical findings showed that expression of miR-195 is down-regulated in TC, which implies that miR-195 may be practically involved in TC pathogenesis. Nevertheless, the function of hsa-miR-195-5p in TC is still largely unclear. Herein, we detected the conceivable involvement of hsa-miR-195-5p in TC cell proliferation. Methods Real time PCR examination was performed to assess the expression level of hsa-miR-195-5p in TC cell lines TPC-1 and B-CPAP. TPC-1 cells were transfected with either hsa-miR-195-5p mimics or hsa-miR-195-5p inhibitor. After confirmation of transfection efficiency, the effect of hsa-miR-195-5p on proliferation and cell cycle of TPC-1 cells was assessed. The expression of cyclin D1 and p21 was simultaneously detected by western blotting. Moreover, targetScan 6.2 was used to predict hsa-miR-195-5p target genes. Subsequently, luciferase reporter was performed to examine whether there is a possible binding of hsa-miR-195-5p to 3’-UTR of cyclin D1 mRNA. Furthermore, cyclin D1 mRNA and protein levels were measured to check whether hsa-miR-195-5p exerts its function at the post-transcriptional level. In addition, to explore the function of cyclin D1 in TPC-1 cells overexpressing hsa-miR-195-5p, cyclin D1 siRNA was used to silence the expression of cyclin D1 in TPC-1 cells overexpressing hsa-miR-195-5p. Results We quantified the expression of hsa-miR-195-5p in TC cells and normal thyroid cells and found a remarkable decrease in hsa-miR-195-5p expression in TC cells. Over-expression of hsa-miR-195-5p obviously resulted in downgraded proliferation of TC cells. Moreover, hsa-miR-195-5p caused cell arrest at the GO/G1 phase. Further in silico analyses and the dual-luciferase reporter assay confirmed that 3’-UTR of cyclin D1 is a direct target of hsa-miR-195-5p. Western blot analysis uncovered that hsa-miR-195-5p over-expression led to decreased levels of cyclin D1 and p21. In mechanistic analyses, we found that silencing of cyclin D1 reversed the inhibitory effect of hsa-miR-195-5p on the proliferation of TC cells, which indicates that hsa-miR-195-5p suppresses TC cell proliferation by adversely regulating cyclin D1. Conclusions We concluded that hsa-miR-195-5p is a candidate tumor-suppressor miRNA in TC and that the hsa-miR-195-5p/p21/cyclin D1 pathway could be a potential therapeutic target for TC.
Collapse
Affiliation(s)
- Dexin Liu
- Department of Radiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Ping Li
- Department of Radiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiaodong Wang
- Department of Radiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei Wang
- Department of Radiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
25
|
Ghimire H, Garlapati C, Janssen EAM, Krishnamurti U, Qin G, Aneja R, Perera AGU. Protein Conformational Changes in Breast Cancer Sera Using Infrared Spectroscopic Analysis. Cancers (Basel) 2020; 12:E1708. [PMID: 32605072 PMCID: PMC7407230 DOI: 10.3390/cancers12071708] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 01/08/2023] Open
Abstract
Protein structural alterations, including misfolding and aggregation, are a hallmark of several diseases, including cancer. However, the possible clinical application of protein conformational analysis using infrared spectroscopy to detect cancer-associated structural changes in proteins has not been established yet. The present study investigates the applicability of Fourier transform infrared spectroscopy in distinguishing the sera of healthy individuals and breast cancer patients. The cancer-associated alterations in the protein structure were analyzed by fitting the amide I (1600-1700 cm-1) band of experimental curves, as well as by comparing the ratio of the absorbance values at the amide II and amide III bands, assigning those as the infrared spectral signatures. The snapshot of the breast cancer-associated alteration in circulating DNA and RNA was also evaluated by extending the spectral fitting protocol to the complex region of carbohydrates and nucleic acids, 1140-1000 cm-1. The sensitivity and specificity of these signatures, representing the ratio of the α-helix and β-pleated sheet in proteins, were both 90%. Likewise, the ratio of amides II and amide III (I1556/I1295) had a sensitivity and specificity of 100% and 80%, respectively. Thus, infrared spectroscopy can serve as a powerful tool to understand the protein structural alterations besides distinguishing breast cancer and healthy serum samples.
Collapse
Affiliation(s)
- Hemendra Ghimire
- Department of Physics and Astronomy, Georgia State University, Atlanta, GA 30303, USA;
| | | | - Emiel A. M. Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger NO-4068, Norway;
| | - Uma Krishnamurti
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Gengsheng Qin
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA 30303, USA;
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (C.G.); (R.A.)
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - A. G. Unil Perera
- Department of Physics and Astronomy, Georgia State University, Atlanta, GA 30303, USA;
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
26
|
Palmer N, Kaldis P. Less-well known functions of cyclin/CDK complexes. Semin Cell Dev Biol 2020; 107:54-62. [PMID: 32386818 DOI: 10.1016/j.semcdb.2020.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/31/2022]
Abstract
Cyclin-dependent kinases (CDKs) are activated by cyclins, which play important roles in dictating the actions of CDK/cyclin complexes. Cyclin binding influences the substrate specificity of these complexes in addition to their susceptibility to inhibition or degradation. CDK/cyclin complexes are best known to promote cell cycle progression in the mitotic cell cycle but are also crucial for important cellular processes not strictly associated with cellular division. This chapter primarily explores the understudied topic of CDK/cyclin complex functionality during the DNA damage response. We detail how CDK/cyclin complexes perform dual roles both as targets of DNA damage checkpoint signaling as well as effectors of DNA repair. Additionally, we discuss the potential CDK-independent roles of cyclins in these processes and the impact of such roles in human diseases such as cancer. Our goal is to place the spotlight on these important functions of cyclins either acting as independent entities or within CDK/cyclin complexes which have attracted less attention in the past. We consider that this will be important for a more complete understanding of the intricate functions of cell cycle proteins in the DNA damage response.
Collapse
Affiliation(s)
- Nathan Palmer
- Institute of Molecular and Cell Biology (IMCB), A⁎STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore; National University of Singapore (NUS), Department of Biochemistry, Singapore, 117597, Republic of Singapore
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), A⁎STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore; National University of Singapore (NUS), Department of Biochemistry, Singapore, 117597, Republic of Singapore; Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Box 50332, SE-202 13, Malmö, Sweden.
| |
Collapse
|
27
|
Ramos-Esquivel A, Hernández-Romero G, Landaverde DU. Cyclin‑dependent kinase 4/6 inhibitors in combination with fulvestrant for previously treated metastatic hormone receptor‑positive breast cancer patients: A systematic review and meta‑analysis of randomized clinical trials. Cancer Treat Res Commun 2020; 23:100175. [PMID: 32361352 DOI: 10.1016/j.ctarc.2020.100175] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 03/19/2020] [Accepted: 04/13/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE To compare the efficacy and safety profile of the combination of cyclin-dependent kinase 4/6 (CDK 4/6) inhibitors and fulvestrant versus fulvestrant alone in previously treated patients with advanced hormone-receptor positive breast cancer. METHODS Phase III randomized clinical trials (RCTs) were retrieved from a systematic review of electronic databases. A random-effect model was employed to determine the pooled hazard ratio (HR) for Progression-Free Survival (PFS) and Overall Survival (OS) using the inverse-variance method. The Mantel Haenszel method was used to calculate the pooled odds ratio (OR) for treatment-related side effects. Heterogeneity was measured using the tau-squared and I2 statistics. RESULTS Three phase III RCTs (n = 1916) were included in the systematic review. Use of abemaciclib, palbociclib, or ribociclib in combination with fulvestrant was significantly associated with longer PFS compared to use of fulvestrant alone (HR: 0.53; 95%CI: 0.47-0.60; p<0.00001), with no significant heterogeneity found among trials. Similarly, OS was significantly longer for patients who received combination therapy in comparison with those allocated to receive fulvestrant alone (HR: 0.77; 95%CI: 0.67-0.89; p<0.0004). The overall odds ratio of serious adverse events (AE) was not significantly increased with the use of the combination therapy (OR: 1.51; 95%CI: 0.74-3.08), with significant heterogeneity found among trials (tau2=0.34; I2=86%; p = 0.0006). CONCLUSION The addition of CDK 4/6 inhibitors (either abemaciclib, palbociclib, or ribociclib) to fulvestrant significantly improved PFS and OS in comparison with fulvestrant alone in patients previously treated with endocrine therapy for advanced breast cancer. No significant heterogeneity was found among CDK 4/6 inhibitors.
Collapse
Affiliation(s)
- Allan Ramos-Esquivel
- Departamento de Oncología Médica. Hospital San Juan de Dios, San José, Costa Rica; Escuela de Medicina, Universidad de Costa Rica. San José, Costa Rica.
| | | | - Denis Ulises Landaverde
- Escuela de Medicina, Universidad de Costa Rica. San José, Costa Rica; Departamento de Oncología Médica, Hospital México, San José, Costa Rica
| |
Collapse
|
28
|
Shankaraiah N, Sakla AP, Laxmikeshav K, Tokala R. Reliability of Click Chemistry on Drug Discovery: A Personal Account. CHEM REC 2020; 20:253-272. [DOI: 10.1002/tcr.201900027] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/08/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Nagula Shankaraiah
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Akash P. Sakla
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Kritika Laxmikeshav
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Ramya Tokala
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| |
Collapse
|
29
|
Zhang J, Wang Q, Wang Q, Cao J, Sun J, Zhu Z. Mechanisms of resistance to estrogen receptor modulators in ER+/HER2- advanced breast cancer. Cell Mol Life Sci 2020; 77:559-572. [PMID: 31471681 PMCID: PMC11105043 DOI: 10.1007/s00018-019-03281-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023]
Abstract
Endocrine therapy represents a mainstay adjuvant treatment of estrogen receptor-positive (ER+) breast cancer in clinical practice with an overall survival (OS) benefit. However, the emergence of resistance is inevitable over time and is present in one-third of the ER+ breast tumors. Several mechanisms of endocrine resistance in ER+/HER2- advanced breast cancers, through ERα itself, receptor tyrosine signaling, or cell cycle pathway, have been identified to be pivotal in endocrine therapy. The epigenetic alterations also contribute to ensuring tumor cells' escape from endocrine therapies. The strategy of combined hormone therapy with targeted pharmaceutical compounds has shown an improvement of progression-free survival or OS in clinical practice, including three different classes of drugs: CDK4/6 inhibitors, selective inhibitor of PI3Kα and mTOR inhibitors. Many therapeutic targets of cell cycle pathway and cell signaling and their combination strategies have recently entered clinical trials. This review focuses on Cyclin D-CDK4/6-RB axis, PI3K pathway and HDACs. Additionally, genomic evolution is complex in tumors exposed to hormonal therapy. We highlight the genomic alterations present in ESR1 and PIK3CA genes to elucidate adaptive mechanisms of endocrine resistance, and discuss how these mutations may inform novel combinations to improve clinical outcomes in the future.
Collapse
Affiliation(s)
- Jin Zhang
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qianying Wang
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qing Wang
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jiangran Cao
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jiafu Sun
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zhengmao Zhu
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
30
|
Ma X, Biswas A, Hammes SR. Paxillin regulated genomic networks in prostate cancer. Steroids 2019; 151:108463. [PMID: 31344408 PMCID: PMC6802295 DOI: 10.1016/j.steroids.2019.108463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 01/26/2023]
Abstract
Paxillin is extensively involved in focal adhesion signaling and kinase signaling throughout the plasma membrane and cytoplasm. However, recent studies in prostate cancer suggest that paxillin also plays a critical role in regulating gene expression within the nucleus, serving as a liaison between cytoplasmic and nuclear MAPK and Androgen Receptor (AR) signaling. Here we used RNA-seq to examine the paxillin-regulated transcriptome in several human prostate cancer cell lines. First, we examined paxillin effects on androgen-mediated transcription in control or paxillin-depleted AR-positive LNCaP and C4-2 human prostate cancer cells. In androgen-dependent LNCaP cells, we found over 1000 paxillin-dependent androgen-responsive genes, some of which are involved in endocrine therapy resistance. Most paxillin-dependent AR-mediated genes in LNCaP cells were no longer paxillin-dependent in androgen-sensitive, castration-resistant C4-2 cells, suggesting that castration-resistance may markedly alter paxillin effects on genomic AR signaling. To examine the paxillin-regulated transcriptome in the absence of androgen signaling, we performed RNA-seq in AR-negative PC3 human prostate cancer cells. Paxillin enhanced several pro-proliferative pathways, including the CyclinD/Rb/E2F and DNA replication/repair pathways. Additionally, paxillin suppressed pro-apoptotic genes, including CASP1 and TNFSF10. Quantitative PCR confirmed that these pathways are similarly regulated by paxillin in LNCaP and C4-2 cells. Functional studies showed that, while paxillin stimulated cell proliferation, it had minimum effect on apoptosis. Thus, paxillin appears to be an important transcriptional regulator in prostate cancer, and analysis of its transcriptome might lead to novel approaches toward the diagnosis and treatment of this important disease.
Collapse
Affiliation(s)
- Xiaoting Ma
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical School, Rochester, NY, United States
| | - Anindita Biswas
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, Lansing, MI, United States
| | - Stephen R Hammes
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical School, Rochester, NY, United States.
| |
Collapse
|
31
|
Ramos N, Baquero-Buitrago J, Ben Youss Gironda Z, Wadghiri YZ, Reiner T, Boada FE, Carlucci G. Noninvasive PET Imaging of CDK4/6 Activation in Breast Cancer. J Nucl Med 2019; 61:437-442. [PMID: 31481582 DOI: 10.2967/jnumed.119.232603] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/19/2019] [Indexed: 11/16/2022] Open
Abstract
The cell cycle is a progression of 4 distinct phases (G1, S, G2, and M), with various cycle proteins being essential in regulating this process. We aimed to develop a radiolabeled cyclin-dependent kinase 4/6 (CDK4/6) inhibitor for breast cancer imaging. Our transfluorinated analog (18F-CDKi) was evaluated and validated as a novel PET imaging agent to quantify CDK4/6 expression in estrogen receptor (ER)-positive human epidermal growth factor receptor 2 (HER2)-negative breast cancer. Methods: 18F-CDKi was synthesized and assayed against CDK4/6 kinases. 18F-CDKi was prepared with a 2-step automated synthetic strategy that yielded the final product with remarkable purity and molar activity. In vitro and in vivo biologic specificity was assessed in a MCF-7 cell line and in mice bearing MCF-7 breast tumors. Nonradioactive palbociclib was used as a blocking agent to investigate the binding specificity and selectivity of 18F-CDKi. Results: 18F-CDKi was obtained with an overall radiochemical uncorrected yield of 15% and radiochemical purity higher than 98%. The total time from the start of synthesis to the final injectable formulated tracer is 70 min. The retention time reported for 18F-CDKi and 19F-CDKi is 27.4 min as demonstrated by coinjection with 19F-CDKi in a high-pressure liquid chromatograph. In vivo blood half-life (weighted, 7.03 min) and octanol/water phase partition coefficient (1.91 ± 0.24) showed a mainly lipophilic behavior. 18F-CDKi is stable in vitro and in vivo (>98% at 4 h after injection) and maintained its potent targeting affinity to CDK4/6. Cellular uptake experiments performed on the MCF-7 breast cancer cell line (ER-positive and HER2-negative) demonstrated specific uptake with a maximum intracellular concentration of about 65% as early as 10 min after incubation. The tracer uptake was reduced to less than 5% when cells were coincubated with a molar excess of palbociclib. In vivo imaging and ex vivo biodistribution of ER-positive, HER2-negative MCF-7 breast cancer models showed a specific uptake of approximately 4% injected dose/g of tumor (reduced to ∼0.3% with a 50-fold excess of cold palbociclib). A comprehensive biodistribution analysis also revealed a significantly lower activation of CDK4/6 in nontargeting organs. Conclusion: 18F-CDKi represents the first 18F PET CDK4/6 imaging agent and a promising imaging agent for ER-positive, HER2-negative breast cancer.
Collapse
Affiliation(s)
- Nicholas Ramos
- Center for Advanced Imaging Innovation and Research (CAIR), NYU School of Medicine, New York, New York; Center for Biomedical Imaging, Department of Radiology, NYU School of Medicine, New York, New York
| | - Jairo Baquero-Buitrago
- Center for Advanced Imaging Innovation and Research (CAIR), NYU School of Medicine, New York, New York; Center for Biomedical Imaging, Department of Radiology, NYU School of Medicine, New York, New York
| | - Zakia Ben Youss Gironda
- Center for Advanced Imaging Innovation and Research (CAIR), NYU School of Medicine, New York, New York; Center for Biomedical Imaging, Department of Radiology, NYU School of Medicine, New York, New York
| | - Youssef Zaim Wadghiri
- Center for Advanced Imaging Innovation and Research (CAIR), NYU School of Medicine, New York, New York; Center for Biomedical Imaging, Department of Radiology, NYU School of Medicine, New York, New York
| | - Thomas Reiner
- Department of Radiology, Weill Cornell Medical College, New York, New York.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and.,Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fernando E Boada
- Center for Advanced Imaging Innovation and Research (CAIR), NYU School of Medicine, New York, New York; Center for Biomedical Imaging, Department of Radiology, NYU School of Medicine, New York, New York
| | - Giuseppe Carlucci
- Center for Advanced Imaging Innovation and Research (CAIR), NYU School of Medicine, New York, New York; Center for Biomedical Imaging, Department of Radiology, NYU School of Medicine, New York, New York
| |
Collapse
|
32
|
Lee KA, Shepherd ST, Johnston SR. Abemaciclib, a potent cyclin-dependent kinase 4 and 6 inhibitor, for treatment of ER-positive metastatic breast cancer. Future Oncol 2019; 15:3309-3326. [PMID: 31464525 DOI: 10.2217/fon-2019-0169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
CDK 4/6 inhibitors have given patients with estrogen receptor (ER)-positive/HER2-negative (ER+/HER2ࢤ) advanced metastatic breast cancer important new therapeutic options. Abemaciclib is different to the other two licensed and approved CDK 4/6 inhibitors, palbociclib and ribociclib, both in dosing schedule (continuous vs intermittent) and toxicity profile (less neutropenia, more diarrhea), yet the magnitude of clinical benefit seen in first- and second-line studies is very similar. One of the key issues for clinicians is when to use these therapies. Ultimately, the biggest impact of abemaciclib could be in the adjuvant setting if the current MONARCH-E trial in high-risk node-positive patients is positive. The emerging biomarker work in the early breast cancer setting (i.e., neoMONARCH) may determine which tumors are most sensitive to abemaciclib.
Collapse
Affiliation(s)
- Karla A Lee
- Breast Unit, The Royal Marsden NHS Foundation Trust, Chelsea, London SW3 6JJ, UK
| | - Scott Tc Shepherd
- Breast Unit, The Royal Marsden NHS Foundation Trust, Chelsea, London SW3 6JJ, UK
| | - Stephen Rd Johnston
- Breast Unit, The Royal Marsden NHS Foundation Trust, Chelsea, London SW3 6JJ, UK
| |
Collapse
|
33
|
Orhan C, Bulut P, Dalay N, Ersen E, Buyru N. Downregulation of TCEAL7 expression induces CCND1 expression in non-small cell lung cancer. Mol Biol Rep 2019; 46:5251-5256. [PMID: 31321645 DOI: 10.1007/s11033-019-04982-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/15/2019] [Indexed: 02/02/2023]
Abstract
Transcription Elongation Factor A-like 7 (TCEAL7) was first reported as a candidate tumor suppressor gene because of its inactivation in ovarian cancer as a result of promoter methylation. Down-regulation of the TCEAL7 gene expression was also associated with other cancers such as endometrial, breast, brain, prostate, gastric cancers, glioblastoma and linked to tumor phenotypes and clinical outcomes. However, there is no report in the literature investigating the role of TCEAL7 in non-small cell lung cancer. Cyclin D1 is an important molecule in the transition from G1 to S phase of the cell cycle, and is frequently deregulated in cancers. Cylin D1 (CCND1) gene is amplified or overexpressed in a variety of tumors. In our previous study we reported that CCND1 over-expression was not associated with amplification in non-small cell lung cancer. Recently, it has been reported that TCEAL7 regulates CCND1 expression through myc-binding E-box sequences. The aim of this study was to investigate the expression of TCEAL7 gene in non-small cell lung cancer and to determine its effect on the CCND1 expression level. For this purpose, expression levels of TCEAL7 and CCND1 genes were investigated in 50 patients with non-small cell lung cancer by quantitative real time polymerase chain reaction (qRT-PCR). TCEAL7 was under-expressed (68%) in non-small cell lung cancer tumor tissues while CCND1 was over-expressed (42%). The TCEAL7 levels negatively correlated with increased CCND1 expression (p = 0.002).
Collapse
Affiliation(s)
- Ceren Orhan
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Cerrahpasa Street, Kocamustafapasa, Fatih, 34098, Istanbul, Turkey
| | - Pelin Bulut
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Cerrahpasa Street, Kocamustafapasa, Fatih, 34098, Istanbul, Turkey
| | - Nejat Dalay
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Cerrahpasa Street, Kocamustafapasa, Fatih, 34098, Istanbul, Turkey
| | - Ezel Ersen
- Department of Chest Surgery, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Kocamustafapasa, Fatih, Istanbul, Turkey
| | - Nur Buyru
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Cerrahpasa Street, Kocamustafapasa, Fatih, 34098, Istanbul, Turkey.
| |
Collapse
|
34
|
Chi Y, Wang F, Zhang T, Xu H, Zhang Y, Shan Z, Wu S, Fan Q, Sun Y. miR-516a-3p inhibits breast cancer cell growth and EMT by blocking the Pygo2/Wnt signalling pathway. J Cell Mol Med 2019; 23:6295-6307. [PMID: 31273950 PMCID: PMC6714144 DOI: 10.1111/jcmm.14515] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 05/13/2019] [Accepted: 06/11/2019] [Indexed: 01/06/2023] Open
Abstract
miR‐516a‐3p has been reported to play a suppressive role in several types of human tumours. However, the expression level, biological function and fundamental mechanisms of miR‐516a‐3p in breast cancer remain unclear. In the present study, we found that miR‐516a‐3p expression was down‐regulated and Pygopus2 (Pygo2) expression was up‐regulated in human breast cancer tissues and cells. Through analysing the clinicopathological characteristics, we demonstrated that low miR‐516a‐3p expression or positive Pygo2 expression was a predictor of poor prognosis for patients with breast cancer. The results of a dual luciferase reporter assay and Western blot analysis indicated that Pygo2 was a target gene of miR‐516a‐3p. Moreover, overexpression of miR‐516a‐3p inhibited cell growth, migration and invasion as well as epithelial‐mesenchymal transition (EMT) of breast cancer cells, whereas reduced miR‐516a‐3p expression promoted breast cancer cell growth, migration, invasion and EMT. Furthermore, we showed that miR‐516a‐3p suppressed cell proliferation, metastasis and EMT of breast cancer cells by inhibiting Pygo2 expression. We confirmed that miR‐516a‐3p exerted an anti‐tumour effect by inhibiting the activation of the Wnt/β‐catenin pathway. Finally, xenograft tumour models were used to show that miR‐516a‐3p inhibited breast cancer cell growth and EMT via suppressing the Pygo2/Wnt signalling pathway. Taken together, these results show that miR‐516a‐3p inhibits breast cancer cell growth, metastasis and EMT by blocking the Pygo2/ Wnt/β‐catenin pathway.
Collapse
Affiliation(s)
- Yanyan Chi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tengfei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Han Xu
- Department of Breast Disease Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yana Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengzheng Shan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shaoxuan Wu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Sun
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
35
|
Makwana V, Ryan P, Patel B, Dukie SA, Rudrawar S. Essential role of O-GlcNAcylation in stabilization of oncogenic factors. Biochim Biophys Acta Gen Subj 2019; 1863:1302-1317. [PMID: 31034911 DOI: 10.1016/j.bbagen.2019.04.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 12/14/2022]
Abstract
A reversible post-translational protein modification which involves addition of N-acetylglucosamine (GlcNAc) onto hydroxyl groups of serine and/or threonine residues which is known as O-GlcNAcylation, has emerged as a potent competitor of phosphorylation. This glycosyltransfer reaction is catalyzed by the enzyme O-linked β-N-acetylglucosamine transferase (OGT). This enzyme uses uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), the end product of hexosamine biosynthetic pathway, to modify numerous nuclear and cytosolic proteins. O-GlcNAcylation influences cancer cell metabolism in such a way that hyper-O-GlcNAcylation is considered as a prominent trait of many cancers, and is proposed as a major factor enabling cancer cell proliferation and progression. Growing evidence supports a connection between O-GlcNAcylation and major oncogenic factors, including for example, c-MYC, HIF-1α, and NF-κB. A comprehensive study of the roles of O-GlcNAc modification of oncogenic factors is warranted as a thorough understanding may help drive advances in cancer diagnosis and therapy. The focus of this article is to highlight the interplay between oncogenic factors and O-GlcNAcylation along with OGT in cancer cell proliferation and survival. The prospects for OGT inhibitors will also be discussed.
Collapse
Affiliation(s)
- Vivek Makwana
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Philip Ryan
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Bhautikkumar Patel
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Shailendra-Anoopkumar Dukie
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland 4222, Australia; Quality Use of Medicines Network, Griffith University, Gold Coast 4222, Australia.
| | - Santosh Rudrawar
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland 4222, Australia; Quality Use of Medicines Network, Griffith University, Gold Coast 4222, Australia.
| |
Collapse
|
36
|
Lundberg A, Lindström LS, Li J, Harrell JC, Darai-Ramqvist E, Sifakis EG, Foukakis T, Perou CM, Czene K, Bergh J, Tobin NP. The long-term prognostic and predictive capacity of cyclin D1 gene amplification in 2305 breast tumours. Breast Cancer Res 2019; 21:34. [PMID: 30819233 PMCID: PMC6394106 DOI: 10.1186/s13058-019-1121-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/14/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Use of cyclin D1 (CCND1) gene amplification as a breast cancer biomarker has been hampered by conflicting assessments of the relationship between cyclin D1 protein levels and patient survival. Here, we aimed to clarify its prognostic and treatment predictive potential through comprehensive long-term survival analyses. METHODS CCND1 amplification was assessed using SNP arrays from two cohorts of 1965 and 340 patients with matching gene expression array and clinical follow-up data of over 15 years. Kaplan-Meier and multivariable Cox regression analyses were used to determine survival differences between CCND1 amplified vs. non-amplified tumours in clinically relevant patient sets, within PAM50 subtypes and within treatment-specific subgroups. Boxplots and differential gene expression analyses were performed to assess differences between amplified vs. non-amplified tumours within PAM50 subtypes. RESULTS When combining both cohorts, worse survival was found for patients with CCND1-amplified tumours in luminal A (HR = 1.68; 95% CI, 1.15-2.46), luminal B (1.37; 1.01-1.86) and ER+/LN-/HER2- (1.66; 1.14-2.41) subgroups. In gene expression analysis, CCND1-amplified luminal A tumours showed increased proliferation (P < 0.001) and decreased progesterone (P = 0.002) levels along with a large overlap in differentially expressed genes when comparing luminal A and B-amplified vs. non-amplified tumours. CONCLUSIONS Our results indicate that CCND1 amplification is associated with worse 15-year survival in ER+/LN-/HER2-, luminal A and luminal B patients. Moreover, luminal A CCND1-amplified tumours display gene expression changes consistent with a more aggressive phenotype. These novel findings highlight the potential of CCND1 to identify patients that could benefit from long-term treatment strategies.
Collapse
Affiliation(s)
- Arian Lundberg
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Linda S Lindström
- Department of Biosciences and Nutrition, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Jingmei Li
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
| | - J Chuck Harrell
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, USA
| | - Eva Darai-Ramqvist
- Department of Pathology and Cytology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Emmanouil G Sifakis
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Charles M Perou
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Department of Public Health, Oxford University, Oxford, UK
| | - Nicholas P Tobin
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden.
| |
Collapse
|
37
|
Scott SC, Lee SS, Abraham J. Mechanisms of therapeutic CDK4/6 inhibition in breast cancer. Semin Oncol 2019; 44:385-394. [PMID: 29935900 DOI: 10.1053/j.seminoncol.2018.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 01/21/2018] [Indexed: 12/29/2022]
Abstract
Cyclin dependent kinase (CDK) 4/6 inhibitors have advanced the treatment of metastatic breast cancer by targeting the cell cycle machinery, interrupting intracellular and mitogenic hormone signals that stimulate proliferation of malignant cells. Preclinical evidence demonstrated that derangements of cyclin D1, CDK4/6, and retinoblastoma expression are common in breast cancer, and suggested a therapeutic benefit from interrupting this axis required for cell cycle progression. Studies of cell lines and animal models of breast cancer have demonstrated the complex interplay between the cell cycle and estrogen receptor and human epidermal growth receptor 2 signaling, which informs our understanding of synergistic use of CDK4/6 inhibitors with endocrine therapy, as well as mechanisms of resistance to endocrine therapy. Interestingly, estrogen receptor activity leads to upregulation of cyclin D1 expression, but the estrogen receptor is also in turn activated by cyclin D1, independent of estrogen binding. Early CDK inhibitors were nonspecific and limited by systemic toxicities, while the current generation of CDK4/6 inhibitors have shown promise in the treatment of hormone receptor-positive breast cancer. Preclinical investigations of the three CDK4/6 inhibitors approved by the US Food and Drug Administration (palbociclib, ribociclib, and abemaciclib) lend further insight into their mechanism of action, which will hopefully inform the future use and refinement of these therapies. Finally, we summarize evidence for additional novel CDK4/6 inhibitors currently in development.
Collapse
Affiliation(s)
| | - Sarah S Lee
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | - Jame Abraham
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH.
| |
Collapse
|
38
|
Hu J, Yue X, Liu J, Kong D. Construction of an miRNA‑gene regulatory network in colorectal cancer through integrated analysis of mRNA and miRNA microarrays. Mol Med Rep 2018; 18:5109-5116. [PMID: 30272280 DOI: 10.3892/mmr.2018.9505] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 08/08/2018] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to identify potential biomarkers associated with colorectal cancer (CRC). The GSE32323 and GSE53592 mRNA and microRNA (miRNA) expression profiles were selected from the Gene Expression Omnibus database. The differentially expressed genes (DEGs) and differentially expressed miRNAs (DEMs) in CRC tissue samples compared with surrounding control tissue samples (DEGs‑CC), and DEGs in cells treated with 5‑aza‑2'‑deoxycitidine compared with untreated cells (DEGs‑TC) were identified with the Limma package. The Database for Annotation, Visualization and Integrated Discovery was used to conduct the functional and pathways enrichment analysis. Differential co‑regulation networks were constructed using the DCGL package of R. The targets of DEMs were identified using TargetScan. The overlaps between DEGs and the targets were selected. The miRNA‑gene regulatory network of the overlaps was established. There were 145 DEMs, and 1,284 DEGs in DEGs‑CC, and 101 DEGs in DEGs‑TC. DEGs‑CC were enriched in 196 Gene Ontology (GO) terms and 23 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. DEGs‑TC were enriched in 46 GO terms and two KEGG pathways. A differential co‑regulation network of the DEGs and a miRNA‑gene regulatory network between DEMs and overlapped DEGs were respectively constructed. miR‑124‑3p, miR‑145‑5p and miR‑320a may be critical in CRC, and serum/glucocorticoid regulated kinase 1 and SRY‑box 9 may be potential biomarkers for CRC tumor progression.
Collapse
Affiliation(s)
- Jun Hu
- Department of Colorectal Cancer Surgery, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Xin Yue
- Department of Colorectal Cancer Surgery, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Jianzhong Liu
- Department of Colorectal Cancer Surgery, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Dalu Kong
- Department of Colorectal Cancer Surgery, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| |
Collapse
|
39
|
Preusser M, De Mattos-Arruda L, Thill M, Criscitiello C, Bartsch R, Ruhstaller T, de Azambuja E, Zielinski CC. CDK4/6 inhibitors in the treatment of patients with breast cancer: summary of a multidisciplinary round-table discussion. ESMO Open 2018; 3:e000368. [PMID: 30167331 PMCID: PMC6109817 DOI: 10.1136/esmoopen-2018-000368] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 12/14/2022] Open
Abstract
This article is the result of a round-table discussion organised by ESMO Open in Vienna in December 2017. Its purpose is to discuss the background and advances in the evidence regarding cyclin-dependent kinase 4/6 inhibitors (palbociclib, ribociclib and abemaciclib) in the treatment of metastatic and early-stage breast cancer and to explore what the key open research questions are and next steps should be.
Collapse
Affiliation(s)
- Matthias Preusser
- Clinical Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Centre, Medical University Vienna - General Hospital, Vienna, Austria.
| | | | - Marc Thill
- Department of Gynaecology and Obstetrics, Agaplesion Markus Hospital, Frankfurt am Main, Germany
| | - Carmen Criscitiello
- Division of Experimental Therapeutics, European Institute of Oncology, Milano, Italy
| | - Rupert Bartsch
- Clinical Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; German Breast Group, Neu-Isenburg, Germany
| | - Thomas Ruhstaller
- Breast Center St. Gallen, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Evandro de Azambuja
- Medicine Department, Institut Jules Bordet and L'Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Christoph C Zielinski
- Comprehensive Cancer Centre, Medical University Vienna - General Hospital, Vienna, Austria
| |
Collapse
|
40
|
Expression of cyclin D1 correlates with p27KIP1 and regulates the degree of oral dysplasia and squamous cell carcinoma differentiation. Oral Surg Oral Med Oral Pathol Oral Radiol 2018; 126:174-183. [DOI: 10.1016/j.oooo.2018.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/16/2018] [Accepted: 01/23/2018] [Indexed: 01/30/2023]
|
41
|
Andrieu GP, Shafran JS, Deeney JT, Bharadwaj KR, Rangarajan A, Denis GV. BET proteins in abnormal metabolism, inflammation, and the breast cancer microenvironment. J Leukoc Biol 2018; 104:265-274. [PMID: 29493812 PMCID: PMC6134394 DOI: 10.1002/jlb.5ri0917-380rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 02/10/2018] [Accepted: 02/10/2018] [Indexed: 12/21/2022] Open
Abstract
Obesity and its associated pathology Type 2 diabetes are two chronic metabolic and inflammatory diseases that promote breast cancer progression, metastasis, and poor outcomes. Emerging critical opinion considers unresolved inflammation and abnormal metabolism separately from obesity; settings where they do not co-occur can inform disease mechanism. In breast cancer, the tumor microenvironment is often infiltrated with T effector and T regulatory cells programmed by metabolic signaling. The pathways by which tumor cells evade immune surveillance, immune therapies, and take advantage of antitumor immunity are poorly understood, but likely depend on metabolic inflammation in the microenvironment. Immune functions are abnormal in metabolic disease, and lessons learned from preclinical studies in lean and metabolically normal environments may not translate to patients with obesity and metabolic disease. This problem is made more urgent by the rising incidence of breast cancer among women who are not obese but who have metabolic disease and associated inflammation, a phenotype common in Asia. The somatic BET proteins, comprising BRD2, BRD3, and BRD4, are new critical regulators of metabolism, coactivate transcription of genes that encode proinflammatory cytokines in immune cell subsets infiltrating the microenvironment, and could be important targets in breast cancer immunotherapy. These transcriptional coregulators are well known to regulate tumor cell progression, but only recently identified as critical for metabolism, metastasis, and expression of immune checkpoint molecules. We consider interrelationships among metabolism, inflammation, and breast cancer aggressiveness relevant to the emerging threat of breast cancer among women with metabolic disease, but without obesity.
Collapse
Affiliation(s)
| | - Jordan S. Shafran
- Cancer Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jude T. Deeney
- Department of Medicine, Section of Endocrinology, Obesity Research Center, Evans Biomedical Research Center; Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kishan R. Bharadwaj
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Annapoorni Rangarajan
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Gerald V. Denis
- Cancer Center, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Miettinen TP, Peltier J, Härtlova A, Gierliński M, Jansen VM, Trost M, Björklund M. Thermal proteome profiling of breast cancer cells reveals proteasomal activation by CDK4/6 inhibitor palbociclib. EMBO J 2018; 37:e98359. [PMID: 29669860 PMCID: PMC5978322 DOI: 10.15252/embj.201798359] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 03/02/2018] [Accepted: 03/09/2018] [Indexed: 11/24/2022] Open
Abstract
Palbociclib is a CDK4/6 inhibitor approved for metastatic estrogen receptor-positive breast cancer. In addition to G1 cell cycle arrest, palbociclib treatment results in cell senescence, a phenotype that is not readily explained by CDK4/6 inhibition. In order to identify a molecular mechanism responsible for palbociclib-induced senescence, we performed thermal proteome profiling of MCF7 breast cancer cells. In addition to affecting known CDK4/6 targets, palbociclib induces a thermal stabilization of the 20S proteasome, despite not directly binding to it. We further show that palbociclib treatment increases proteasome activity independently of the ubiquitin pathway. This leads to cellular senescence, which can be counteracted by proteasome inhibitors. Palbociclib-induced proteasome activation and senescence is mediated by reduced proteasomal association of ECM29. Loss of ECM29 activates the proteasome, blocks cell proliferation, and induces a senescence-like phenotype. Finally, we find that ECM29 mRNA levels are predictive of relapse-free survival in breast cancer patients treated with endocrine therapy. In conclusion, thermal proteome profiling identifies the proteasome and ECM29 protein as mediators of palbociclib activity in breast cancer cells.
Collapse
Affiliation(s)
- Teemu P Miettinen
- Division of Cell and Developmental Biology, University of Dundee, Dundee, UK
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Julien Peltier
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Anetta Härtlova
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Marek Gierliński
- Division of Computational Biology, University of Dundee, Dundee, UK
| | - Valerie M Jansen
- Division of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthias Trost
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mikael Björklund
- Division of Cell and Developmental Biology, University of Dundee, Dundee, UK
| |
Collapse
|
43
|
Maharjan S, Park BK, Lee SI, Lim Y, Lee K, Kwon HJ. Gomisin G Inhibits the Growth of Triple-Negative Breast Cancer Cells by Suppressing AKT Phosphorylation and Decreasing Cyclin D1. Biomol Ther (Seoul) 2018; 26:322-327. [PMID: 29587339 PMCID: PMC5933900 DOI: 10.4062/biomolther.2017.235] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/05/2018] [Accepted: 01/09/2018] [Indexed: 12/31/2022] Open
Abstract
A type of breast cancer with a defect in three molecular markers such as the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor is called triple-negative breast cancer (TNBC). Many patients with TNBC have a lower survival rate than patients with other types due to a poor prognosis. In this study, we confirmed the anti-cancer effect of a natural compound, Gomisin G, in TNBC cancer cells. Treatment with Gomisin G suppressed the viability of two TNBC cell lines, MDA-MB-231 and MDA-MB-468 but not non-TNBC cell lines such as MCF-7, T47D, and ZR75-1. To investigate the molecular mechanism of this activity, we examined the signal transduction pathways after treatment with Gomisin G in MDA-MB-231 cells. Gomisin G did not induce apoptosis but drastically inhibited AKT phosphorylation and reduced the amount of retinoblastoma tumor suppressor protein (Rb) and phosphorylated Rb. Gomisin G induced in a proteasome-dependent manner a decrease in Cyclin D1. Consequently, Gomisin G causes cell cycle arrest in the G1 phase. In contrast, there was no significant change in T47D cells except for a mild decrease in AKT phosphorylation. These results show that Gomisin G has an anti-cancer activity by suppressing proliferation rather than inducing apoptosis in TNBC cells. Our study suggests that Gomisin G could be used as a therapeutic agent in the treatment of TNBC patients.
Collapse
Affiliation(s)
- Sony Maharjan
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Byoung Kwon Park
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Su In Lee
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Yoonho Lim
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul 05029, Republic of Korea
| | - Keunwook Lee
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyung-Joo Kwon
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea.,Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
44
|
Villegas MR, Baeza A, Vallet-Regí M. Nanotechnological Strategies for Protein Delivery. Molecules 2018; 23:E1008. [PMID: 29693640 PMCID: PMC6100203 DOI: 10.3390/molecules23051008] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 04/20/2018] [Accepted: 04/22/2018] [Indexed: 12/22/2022] Open
Abstract
The use of therapeutic proteins plays a fundamental role in the treatment of numerous diseases. The low physico-chemical stability of proteins in physiological conditions put their function at risk in the human body until they reach their target. Moreover, several proteins are unable to cross the cell membrane. All these facts strongly hinder their therapeutic effect. Nanomedicine has emerged as a powerful tool which can provide solutions to solve these limitations and improve the efficacy of treatments based on protein administration. This review discusses the advantages and limitations of different types of strategies employed for protein delivery, such as PEGylation, transport within liposomes or inorganic nanoparticles or their in situ encapsulation.
Collapse
Affiliation(s)
- María Rocío Villegas
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, UCM, 28040 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.
| | - Alejandro Baeza
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, UCM, 28040 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, UCM, 28040 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.
| |
Collapse
|
45
|
Molecular mechanisms affecting estrogen receptor levels in breast cancer. JOURNAL OF SURGERY AND MEDICINE 2018. [DOI: 10.28982/josam.412314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
46
|
Sgambato A, Flamini G, Cittadini A, Weinstein IB. Abnormalities in Cell Cycle Control in Cancer and Their Clinical Implications. TUMORI JOURNAL 2018; 84:421-33. [PMID: 9824993 DOI: 10.1177/030089169808400401] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recent studies indicate that the functions of several genes that control the cell cycle are altered during the carcinogenic process and that these changes perturb both cell proliferation and genomic stability, thus promoting cell transformation and enhancing the process of tumor progression. The purpose of this paper is to review current information on the role of cyclins and related genes in the control of the mammalian cell cycle, the types of abnormalities in these genes found in human tumors and the possible clinical implications of these findings.
Collapse
Affiliation(s)
- A Sgambato
- Centro di Ricerche Oncologiche Giovanni XXIII, Catholic University, Rome, Italy
| | | | | | | |
Collapse
|
47
|
Abstract
The tumor suppressor LKB1 is an essential serine/threonine kinase, which regulates various cellular processes such as cell metabolism, cell proliferation, cell polarity, and cell migration. Germline mutations in the STK11 gene (encoding LKB1) are the cause of the Peutz-Jeghers syndrome, which is characterized by benign polyps in the intestine and a higher risk for the patients to develop intestinal and extraintestinal tumors. Moreover, mutations and misregulation of LKB1 have been reported to occur in most types of tumors and are among the most common aberrations in lung cancer. LKB1 activates several downstream kinases of the AMPK family by direct phosphorylation in the T-loop. In particular the activation of AMPK upon energetic stress has been intensively analyzed in various diseases, including cancer to induce a metabolic switch from anabolism towards catabolism to regulate energy homeostasis and cell survival. In contrast, the regulation of LKB1 itself has long been only poorly understood. Only in the last years, several proteins and posttranslational modifications of LKB1 have been analyzed to control its localization, activity and recognition of substrates. Here, we summarize the current knowledge about the upstream regulation of LKB1, which is important for the understanding of the pathogenesis of many types of tumors.
Collapse
|
48
|
Villegas SL, Darb-Esfahani S, von Minckwitz G, Huober J, Weber K, Marmé F, Furlanetto J, Schem C, Pfitzner BM, Lederer B, Engels K, Kümmel S, Müller V, Mehta K, Denkert C, Loibl S. Expression of Cyclin D1 protein in residual tumor after neoadjuvant chemotherapy for breast cancer. Breast Cancer Res Treat 2017; 168:179-187. [PMID: 29177689 DOI: 10.1007/s10549-017-4581-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/15/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE Hormone receptor (HR)-positive breast cancer (BC) shows a poor response to neoadjuvant chemotherapy (NACT). New treatment targets like the Cyclin D1-CDK4/CDK6 complex are promising adjuvant/post-neoadjuvant therapeutic strategies. Evaluating Cyclin D1 overexpression in residual tumor could recognize those patients that benefit most from such post-neoadjuvant treatment. In this study, we determined Cyclin D1 expression in residual BC after NACT. Secondary aims were to correlate Cyclin D1 expression levels with clinicopathological parameters and to assess its prognostic value after NACT. METHODS We retrospectively assessed the nuclear expression of Cyclin D1 on tissue microarrays with residual tumor from 284 patients treated in the neoadjuvant GeparTrio (n = 186) and GeparQuattro (n = 98) trials. Evaluation was performed with a standardized immunoreactive score (IRS) after selecting a cut-off value. RESULTS A high expression level (IRS ≥ 6) of Cyclin D1 was found in 37.3% of the assessed specimens. An increased Cyclin D1 expression was observed in HR-positive tumors, compared to HR-negative tumors (p = 0.02). Low Cyclin D1 levels correlated with clinical tumor stage 1-3 (p = 0.03). Among patients with HR-positive/Her2-negative tumors and high Cyclin D1 expression, a better disease-free survival (DFS) was graphically suggested, but not significant (p = 0.21). CONCLUSION Our study demonstrates a measurable nuclear expression of Cyclin D1 in post-neoadjuvant residual tumor tissue of HR-positive BC. Cyclin D1 expression was not prognostic for DFS after NACT. Our results and defined cut-off suggest that the marker can be used to stratify tumors according to protein expression levels. Based on this, a prospective evaluation is currently performed in the ongoing Penelope-B trial.
Collapse
Affiliation(s)
- S L Villegas
- Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - S Darb-Esfahani
- Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Institute of Pathology Spandau, Evangelisches Waldkrankenhaus, Stadtrandstr. 555, 13589, Berlin, Germany
| | - G von Minckwitz
- German Breast Group (GBG Forschungs GmbH), Martin-Behaim-Str. 12, 63263, Neu-Isenburg, Germany
| | - J Huober
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - K Weber
- German Breast Group (GBG Forschungs GmbH), Martin-Behaim-Str. 12, 63263, Neu-Isenburg, Germany
| | - F Marmé
- National Center for Tumor Diseases, University-Hospital Heidelberg, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - J Furlanetto
- German Breast Group (GBG Forschungs GmbH), Martin-Behaim-Str. 12, 63263, Neu-Isenburg, Germany
| | - C Schem
- Department of Gynecology and Obstetrics, University Hospital Schleswig-Hostein, Kiel, Germany
| | - B M Pfitzner
- Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - B Lederer
- German Breast Group (GBG Forschungs GmbH), Martin-Behaim-Str. 12, 63263, Neu-Isenburg, Germany
| | - K Engels
- Zentrum für Pathologie, Zytologie und Molekularpathologie Neuss, Neuss, Germany
| | - S Kümmel
- Breast Unit Kliniken Essen-Mitte, Essen, Germany
| | - V Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - K Mehta
- German Breast Group (GBG Forschungs GmbH), Martin-Behaim-Str. 12, 63263, Neu-Isenburg, Germany
| | - C Denkert
- Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,German Cancer Consortium (DKTK), Partner Site Charité, Berlin, Germany.
| | - S Loibl
- German Breast Group (GBG Forschungs GmbH), Martin-Behaim-Str. 12, 63263, Neu-Isenburg, Germany
| |
Collapse
|
49
|
Ortiz AB, Garcia D, Vicente Y, Palka M, Bellas C, Martin P. Prognostic significance of cyclin D1 protein expression and gene amplification in invasive breast carcinoma. PLoS One 2017; 12:e0188068. [PMID: 29140993 PMCID: PMC5687747 DOI: 10.1371/journal.pone.0188068] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023] Open
Abstract
The oncogenic capacity of cyclin D1 has long been established in breast cancer. CCND1 amplification has been identified in a subset of patients with poor prognosis, but there are conflicting data regarding the predictive value of cyclin D1 protein overexpression. This study was designed to analyze the expression of cyclin D1 and its correlation with CCND1 amplification and their prognostic implications in invasive breast cancer. By using the tissue microarray technique, we performed an immunohistochemical study of ER, PR, HER2, p53, cyclin D1, Ki67 and p16 in 179 invasive breast carcinoma cases. The FISH method was performed to detect HER2/Neu and CCND1 amplification. High cyclin D1 expression was identified in 94/179 (52%) of invasive breast cancers. Cyclin D1 overexpression and CCND1 amplification were significantly associated (p = 0.010). Overexpression of cyclin D1 correlated with ER expression, PR expression and Luminal subtypes (p<0.001), with a favorable impact on overall survival in the whole series. However, in the Luminal A group, high expression of cyclin D1 correlated with shorter disease-free survival, suggesting that the prognostic role of cyclin D1 depends on the molecular subtype. CCND1 gene amplification was detected in 17 cases (9%) and correlated significantly with high tumor grade (p = 0.038), high Ki-67 protein expression (p = 0.002), and the Luminal B subtype (p = 0.002). Patients with tumors with high amplification of CCND1 had an increased risk of recurrence (HR = 2.5; 95% CI, 1.2-4.9, p = 0.01). These findings suggest that CCND1 amplification could be useful for predicting recurrence in invasive breast cancer.
Collapse
Affiliation(s)
- Angela B. Ortiz
- Pathology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM) Majadahonda, Madrid, Spain
| | - Diego Garcia
- Pathology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM) Majadahonda, Madrid, Spain
| | - Yolanda Vicente
- Pathology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM) Majadahonda, Madrid, Spain
| | - Magda Palka
- Medical Oncology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM) Majadahonda, Madrid, Spain
| | - Carmen Bellas
- Pathology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM) Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Paloma Martin
- Pathology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM) Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- * E-mail:
| |
Collapse
|
50
|
Sledge GW, Toi M, Neven P, Sohn J, Inoue K, Pivot X, Burdaeva O, Okera M, Masuda N, Kaufman PA, Koh H, Grischke EM, Frenzel M, Lin Y, Barriga S, Smith IC, Bourayou N, Llombart-Cussac A. MONARCH 2: Abemaciclib in Combination With Fulvestrant in Women With HR+/HER2− Advanced Breast Cancer Who Had Progressed While Receiving Endocrine Therapy. J Clin Oncol 2017; 35:2875-2884. [DOI: 10.1200/jco.2017.73.7585] [Citation(s) in RCA: 824] [Impact Index Per Article: 117.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Purpose MONARCH 2 ( ClinicalTrials.gov identifier: NCT02107703) compared the efficacy and safety of abemaciclib, a selective cyclin-dependent kinase 4 and 6 inhibitor, plus fulvestrant with fulvestrant alone in patients with advanced breast cancer (ABC). Patients and Methods MONARCH 2 was a global, double-blind, phase III study of women with hormone receptor-positive and human epidermal growth factor receptor 2-negative ABC who had progressed while receiving neoadjuvant or adjuvant endocrine therapy (ET), ≤ 12 months from the end of adjuvant ET, or while receiving first-line ET for metastatic disease. Patients were randomly assigned 2:1 to receive abemaciclib or placebo (150 mg twice daily) on a continuous schedule and fulvestrant (500 mg, per label). The primary end point was investigator-assessed progression-free survival (PFS), and key secondary end points included overall survival, objective response rate (ORR), duration of response, clinical benefit rate, quality of life, and safety. Results Between August 2014 and December 2015, 669 patients were randomly assigned to receive abemaciclib plus fulvestrant (n = 446) or placebo plus fulvestrant (n = 223). Abemaciclib plus fulvestrant significantly extended PFS versus fulvestrant alone (median, 16.4 v 9.3 months; hazard ratio, 0.553; 95% CI, 0.449 to 0.681; P < .001). In patients with measurable disease, abemaciclib plus fulvestrant achieved an ORR of 48.1% (95% CI, 42.6% to 53.6%) compared with 21.3% (95% CI, 15.1% to 27.6%) in the control arm. The most common adverse events in the abemaciclib versus placebo arms were diarrhea (86.4% v 24.7%), neutropenia (46.0% v 4.0%), nausea (45.1% v 22.9%), and fatigue (39.9% v 26.9%). Conclusions Abemaciclib at 150 mg twice daily plus fulvestrant was effective, significantly improving PFS and ORR and demonstrating a tolerable safety profile in women with hormone receptor-positive and human epidermal growth factor receptor 2-negative ABC who progressed while receiving ET.
Collapse
Affiliation(s)
- George W. Sledge
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Masakazu Toi
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Patrick Neven
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Joohyuk Sohn
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Kenichi Inoue
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Xavier Pivot
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Olga Burdaeva
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Meena Okera
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Norikazu Masuda
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Peter A. Kaufman
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Han Koh
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Eva-Maria Grischke
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Martin Frenzel
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Yong Lin
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Susana Barriga
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Ian C. Smith
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Nawel Bourayou
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Antonio Llombart-Cussac
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| |
Collapse
|