1
|
Zou Z, Zhong L. Anaplastic thyroid cancer: Genetic roles, targeted therapy, and immunotherapy. Genes Dis 2025; 12:101403. [PMID: 40271195 PMCID: PMC12018003 DOI: 10.1016/j.gendis.2024.101403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/02/2024] [Accepted: 08/02/2024] [Indexed: 04/25/2025] Open
Abstract
Anaplastic thyroid cancer (ATC) stands as the most formidable form of thyroid malignancy, presenting a persistent challenge in clinical management. Recent years have witnessed a gradual unveiling of the intricate genetic underpinnings governing ATC through next-generation sequencing. The emergence of this genetic landscape has paved the way for the exploration of targeted therapies and immunotherapies in clinical trials. Despite these strides, the precise mechanisms governing ATC pathogenesis and the identification of efficacious treatments demand further investigation. Our comprehensive review stems from an extensive literature search focusing on the genetic implications, notably the pivotal MAPK and PI3K-AKT-mTOR signaling pathways, along with targeted therapies and immunotherapies in ATC. Moreover, we screen and summarize the advances and challenges in the current diagnostic approaches for ATC, including the invasive tissue sampling represented by fine needle aspiration and core needle biopsy, immunohistochemistry, and 18F-fluorodeoxyglucose positron emission tomography/computed tomography. We also investigate enormous studies on the prognosis of ATC and outline independent prognostic factors for future clinical assessment and therapy for ATC. By synthesizing this literature, we aim to encapsulate the evolving landscape of ATC oncology, potentially shedding light on novel pathogenic mechanisms and avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zhao Zou
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Linhong Zhong
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
2
|
Sun J, Wang Y, Ma S, Zhang W, Yang L, Zhang B, Tang Q. Combined Ribociclib and NU7026 administration enhances radio-sensitivity by inhibiting DNA repair in prostate cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:7075-7085. [PMID: 39718616 DOI: 10.1007/s00210-024-03673-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/24/2024] [Indexed: 12/25/2024]
Abstract
Radiosensitivity is critical for clinical outcomes and overall survival of prostate cancer patients treated with irradiation. Ribociclib and NU7026 have been reported as radiosensitizers in cancer cells, but which are inadequately understood in prostate cancer cells. The present study was performed to investigate the effects of ribociclib, NU7026, and their combination on the radiosensitivity of prostate cancer cells. Optimal combined concentrations of ribociclib and NU7026 with X-irradiation were selected. Flow cytometry, CCK-8 assays, soft agar colony formation, and γH2AX foci formation assays were performed to assess the cell cycle distribution, cell viability, cell proliferation, and DNA damage, respectively. Finally, the expression of proteins related to the cell cycle, DNA repair, and cell death was evaluated by western blot analysis. Ribociclib arrested prostate cell cycle in the G0/G1 phase. Compared with the control, X-irradiation combined with ribociclib or NU7026 significantly decreased cell proliferation, reduced colony formation, and enhanced γH2AX foci formation in PC3 and DU145 cells. Moreover, the protein expression levels of CDK4, p-Rb, cyclin D1, rad51, and DNA-PKcs were significantly decreased, whereas that of Bax was significantly increased.The combination of ribociclib and NU7026 may be a potential strategy for prostate cancer therapy by promoting radiosensitivity.
Collapse
Affiliation(s)
- Jinbo Sun
- Department of Urology, General Hospital of the Central Theater Command, Wuhan, 430070, Hubei, China
| | - Yong Wang
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Shanjin Ma
- Department of Urology, the 955th Hospital of Army, Changdu, 854000, Tibet, China
| | - Wei Zhang
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Longfei Yang
- Department of Medical Laboratory and Research Center, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Bo Zhang
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| | - Qisheng Tang
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
3
|
Mughal MJ, Zhang Y, Li Z, Zhou S, Peng C, Zhang YQ, Seto E, Shen M, Hall MD, Zhu W. TFAP2C-DDR1 axis regulates resistance to CDK4/6 inhibitor in breast cancer. Cancer Lett 2025; 610:217356. [PMID: 39603379 PMCID: PMC11783577 DOI: 10.1016/j.canlet.2024.217356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Breast cancer is the predominant malignancy with the majority of cases are characterized as HR+/HER2-subtype. Although cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) have shown remarkable efficacy in treating this subtype when combined with endocrine therapy, the development of resistance to these inhibitors remains a significant clinical obstacle. Hence, there is an urgent need to explore innovative therapies and decipher the underlying mechanisms of resistance to CDK4/6i. In this study, we employed quantitative high-throughput combination screening (qHTCS) and genomics/proteomics approaches to uncover the molecular mechanisms driving resistance to CDK4/6i (palbociclib) in breast cancer. The comprehensive analyses revealed DDR1 as a potential factor implicated in mediating resistance to CDK4/6i. Specifically, DDR1 inhibition in combination with palbociclib exhibited remarkable synergistic effects, reducing cell survival signaling and promoting apoptosis in resistant cells. In-vivo xenograft model further validated the synergistic effects, showing a significant reduction in the resistant tumor growth. Exploration into DDR1 activation uncovered TFAP2C as a key transcription factor regulating DDR1 expression in palbociclib resistant cells and inhibition of TFAP2C re-sensitized resistant cells to palbociclib. Gene set enrichment analysis (GSEA) in the NeoPalAna trial demonstrated a significant enrichment of the TFAP2C-DDR1 gene set from patitens after palbociclib treatment, suggesting the possible activation of the TFAP2C-DDR1 axis following palbociclib exposure. Overall, this study provides crucial insights into the novel molecular landscape of palbociclib resistance in breast cancer, suggesting TFAP2C-DDR1 axis inhibition as a promising strategy to overcome resistance.
Collapse
Affiliation(s)
- Muhammad Jameel Mughal
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Yi Zhang
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Zhuqing Li
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Shuyan Zhou
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Changmin Peng
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Ya-Qin Zhang
- Early Translation Branch, Division of Preclinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, 20850, USA
| | - Edward Seto
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Min Shen
- Early Translation Branch, Division of Preclinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, 20850, USA
| | - Matthew D Hall
- Early Translation Branch, Division of Preclinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, 20850, USA
| | - Wenge Zhu
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA.
| |
Collapse
|
4
|
Zamora-Zaragoza J, Klap K, Sánchez-Pérez J, Vielle-Calzada JP, Willemsen V, Scheres B. Developmental cues are encoded by the combinatorial phosphorylation of Arabidopsis RETINOBLASTOMA-RELATED protein RBR1. EMBO J 2024; 43:6656-6678. [PMID: 39468281 PMCID: PMC11649800 DOI: 10.1038/s44318-024-00282-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 08/29/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
RETINOBLASTOMA-RELATED (RBR) proteins orchestrate cell division, differentiation, and survival in response to environmental and developmental cues through protein-protein interactions that are governed by multisite phosphorylation. Here we explore, using a large collection of transgenic RBR phosphovariants to complement protein function in Arabidopsis thaliana, whether differences in the number and position of RBR phosphorylation events cause a diversification of the protein's function. While the number of point mutations influence phenotypic strength, phosphosites contribute differentially to distinct phenotypes. RBR pocket domain mutations associate primarily with cell proliferation, while mutations in the C-region are linked to stem cell maintenance. Both phospho-mimetic and a phospho-defective variants promote cell death, suggesting that distinct mechanisms can lead to similar cell fates. We observed combinatorial effects between phosphorylated T406 and phosphosites in different protein domains, suggesting that specific, additive, and combinatorial phosphorylation events fine-tune RBR function. Suppression of dominant phospho-defective RBR phenotypes with a mutation that inhibits RBR interacting with LXCXE motifs, and an exhaustive protein-protein interaction assay, not only revealed the importance of DREAM complex members in phosphorylation-regulated RBR function but also pointed to phosphorylation-independent RBR roles in environmental responses. Thus, combinatorial phosphorylation defined and separated developmental, but not environmental, functions of RBR.
Collapse
Affiliation(s)
- Jorge Zamora-Zaragoza
- Laboratory of Cell and Developmental Biology, Department of Plant Sciences, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
- Rijk Zwaan Breeding B.V., Department of Biotechnology, Eerste Kruisweg 9, 4793 RS, Fijnaart, The Netherlands
| | - Katinka Klap
- Laboratory of Cell and Developmental Biology, Department of Plant Sciences, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
| | - Jaheli Sánchez-Pérez
- Laboratorio Nacional de Genómica para la Biodiversidad, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 36824, Irapuato, Guanajuato, Mexico
| | - Jean-Philippe Vielle-Calzada
- Laboratorio Nacional de Genómica para la Biodiversidad, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 36824, Irapuato, Guanajuato, Mexico
| | - Viola Willemsen
- Laboratory of Cell and Developmental Biology, Department of Plant Sciences, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
| | - Ben Scheres
- Laboratory of Cell and Developmental Biology, Department of Plant Sciences, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands.
- Rijk Zwaan Breeding B.V., Department of Biotechnology, Eerste Kruisweg 9, 4793 RS, Fijnaart, The Netherlands.
| |
Collapse
|
5
|
Baghermanesh SS, Barati M, Hosseini A. The effect of ribociclib on the expression levels of miR-141 and CDK4/6-USP51 signaling pathway genes in MCF-7 and MDA-MB-231 cells. PLoS One 2024; 19:e0309289. [PMID: 39196911 PMCID: PMC11355560 DOI: 10.1371/journal.pone.0309289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 08/07/2024] [Indexed: 08/30/2024] Open
Abstract
INTRODUCTION Patients with breast cancer, especially triple-negative breast cancer, have a poor prognosis. There is still no effective treatment for this disease. Due to resistance to traditional treatments such as chemotherapy and radiation therapy, there is a need to discover novel treatment strategies to treat this disease. Ribociclib is a selective CDK4/6 inhibitor. Approximately 20% of patients with HR+ breast cancer developed primary resistance to CDK4/6 inhibitors, and more than 30% experienced secondary resistance. Since most patients experience resistance during CDK4/6 inhibitor treatment, managing this disease is becoming more challenging. Many malignant tumors abnormally express microRNA (miR)-141, which participates in several cellular processes, including drug resistance, proliferation, epithelial-mesenchymal transition, migration, and invasion. MATERIALS AND METHODS In the present study, we cultured MDA-MB-231 and MCF-7 cells in DMEM-F12 medium. By performing MTT assay we determined the cytotoxic effects of ribociclib on breast cancer cells, as well as determining the IC50 of it. Then, we treated the cells with ribociclib at two time points: 24 h and 72 h. After that, RNA was isolated and reverse transcribed to cDNA. Finally, we performed qRT‒PCR to evaluate how ribociclib affects the expression level of desired genes. RESULTS AND CONCLUSION We found that ribociclib can inhibit cell growth in a dose- and time-dependent manner. We examined the mRNA expression of 4 genes. After ribociclib treatment, the mRNA expression of CDK6 and MYH10 decreased (p < 0.01, p < 0.05). The mRNA expression of CDON increased (p<0.05), but no significant changes were observed in ZEB1 mRNA expression. Furthermore, the qRT‒PCR results for miR-141 showed that the expression of miR-141 increased (p<0.01) after 72 h of treatment with ribociclib.
Collapse
Affiliation(s)
- Shayeste Sadat Baghermanesh
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Barati
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Arshad Hosseini
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Pita JM, Raspé E, Coulonval K, Decaussin-Petrucci M, Tarabichi M, Dom G, Libert F, Craciun L, Andry G, Wicquart L, Leteurtre E, Trésallet C, Marlow LA, Copland JA, Durante C, Maenhaut C, Cavaco BM, Dumont JE, Costante G, Roger PP. CDK4 phosphorylation status and rational use for combining CDK4/6 and BRAF/MEK inhibition in advanced thyroid carcinomas. Front Endocrinol (Lausanne) 2023; 14:1247542. [PMID: 37964967 PMCID: PMC10641312 DOI: 10.3389/fendo.2023.1247542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/26/2023] [Indexed: 11/16/2023] Open
Abstract
Background CDK4/6 inhibitors (CDK4/6i) have been established as standard treatment against advanced Estrogen Receptor-positive breast cancers. These drugs are being tested against several cancers, including in combinations with other therapies. We identified the T172-phosphorylation of CDK4 as the step determining its activity, retinoblastoma protein (RB) inactivation, cell cycle commitment and sensitivity to CDK4/6i. Poorly differentiated (PDTC) and anaplastic (ATC) thyroid carcinomas, the latter considered one of the most lethal human malignancies, represent major clinical challenges. Several molecular evidence suggest that CDK4/6i could be considered for treating these advanced thyroid cancers. Methods We analyzed by two-dimensional gel electrophoresis the CDK4 modification profile and the presence of T172-phosphorylated CDK4 in a collection of 98 fresh-frozen tissues and in 21 cell lines. A sub-cohort of samples was characterized by RNA sequencing and immunohistochemistry. Sensitivity to CDK4/6i (palbociclib and abemaciclib) was assessed by BrdU incorporation/viability assays. Treatment of cell lines with CDK4/6i and combination with BRAF/MEK inhibitors (dabrafenib/trametinib) was comprehensively evaluated by western blot, characterization of immunoprecipitated CDK4 and CDK2 complexes and clonogenic assays. Results CDK4 phosphorylation was detected in all well-differentiated thyroid carcinomas (n=29), 19/20 PDTC, 16/23 ATC and 18/21 thyroid cancer cell lines, including 11 ATC-derived ones. Tumors and cell lines without phosphorylated CDK4 presented very high p16CDKN2A levels, which were associated with proliferative activity. Absence of CDK4 phosphorylation in cell lines was associated with CDK4/6i insensitivity. RB1 defects (the primary cause of intrinsic CDK4/6i resistance) were not found in 5/7 tumors without detectable phosphorylated CDK4. A previously developed 11-gene expression signature identified the likely unresponsive tumors, lacking CDK4 phosphorylation. In cell lines, palbociclib synergized with dabrafenib/trametinib by completely and permanently arresting proliferation. These combinations prevented resistance mechanisms induced by palbociclib, most notably Cyclin E1-CDK2 activation and a paradoxical stabilization of phosphorylated CDK4 complexes. Conclusion Our study supports further clinical evaluation of CDK4/6i and their combination with anti-BRAF/MEK therapies as a novel effective treatment against advanced thyroid tumors. Moreover, the complementary use of our 11 genes predictor with p16/KI67 evaluation could represent a prompt tool for recognizing the intrinsically CDK4/6i insensitive patients, who are potentially better candidates to immediate chemotherapy.
Collapse
Affiliation(s)
- Jaime M. Pita
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Eric Raspé
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Katia Coulonval
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Maxime Tarabichi
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Geneviève Dom
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Frederick Libert
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
- BRIGHTCore, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Ligia Craciun
- Tumor Bank of the Institut Jules Bordet Comprehensive Cancer Center – Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Guy Andry
- Department of Head & Neck and Thoracic Surgery, Institut Jules Bordet Comprehensive Cancer Center – Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Laurence Wicquart
- Tumorothèque du Groupement de Coopération Sanitaire-Centre Régional de Référence en Cancérologie (C2RC) de Lille, Lille, France
| | - Emmanuelle Leteurtre
- Department of Pathology, Univ. Lille, Centre National de la Recherche Scientifique (CNRS), Inserm, Centre Hospitalo-Universitaire (CHU) Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Christophe Trésallet
- Department of General and Endocrine Surgery - Pitié-Salpêtrière Hospital, Sorbonne University, Assistance Publique des Hôpitaux de Paris, Paris, France
- Department of Digestive, Bariatric and Endocrine Surgery - Avicenne University Hospital, Paris Nord - Sorbonne University, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Laura A. Marlow
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| | - John A. Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Cosimo Durante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Carine Maenhaut
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Branca M. Cavaco
- Molecular Endocrinology Group, Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Lisbon, Portugal
| | - Jacques E. Dumont
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Giuseppe Costante
- Departments of Endocrinology and Medical Oncology, Institut Jules Bordet Comprehensive Cancer Center – Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Pierre P. Roger
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
7
|
Bai F, Liu X, Zhang X, Mao Z, Wen H, Ma J, Pei XH. p18INK4C and BRCA1 inhibit follicular cell proliferation and dedifferentiation in thyroid cancer. Cell Cycle 2023; 22:1637-1653. [PMID: 37345432 PMCID: PMC10361144 DOI: 10.1080/15384101.2023.2225938] [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: 09/09/2022] [Revised: 04/13/2023] [Accepted: 06/09/2023] [Indexed: 06/23/2023] Open
Abstract
Only 3% of thyroid cancers are medullary thyroid carcinomas (MTCs), the rest are follicular epithelial cell derived non-MTCs (NMTCs). A dysfunctional INK4-CDK4-RB pathway is detected in most of NMTCs. DNA repair defects and genome instability are associated with NMTC dedifferentiation and aggressiveness. Whether inactivation of the INK4-CDK4-RB pathway induces NMTCs and how differentiation of NMTC cells is controlled remain elusive. In this study, we generated p18Ink4c and Brca1 singly and doubly deficient mice as well as p16Ink4a and Brca1 singly and doubly deficient mice. By using these mice and human thyroid carcinoma cell lines, we discovered that loss of p18Ink4c, not p16Ink4a, in mice stimulated follicular cell proliferation and induced NMTCs. Depletion of Brca1 alone or both p16Ink4a and Brca1 did not induce thyroid tumor. Depletion of Brca1 in p18Ink4c null mice results in poorly differentiated and aggressive NMTCs with epithelial-mesenchymal transition (EMT) features and enhanced DNA damage. Knockdown of BRCA1 in thyroid carcinoma cells activated EMT and promoted tumorigenesis whereas overexpression of BRCA1 inhibited EMT. BRCA1 and EMT marker expression were inversely related in human thyroid cancers. Our finding, for the first time, demonstrates that inactivation of INK4-CDK4-RB pathway induces NMTCs and that Brca1 deficiency promotes dedifferentiation of NMTC cells. These results suggest that BRCA1 and p18INK4C collaboratively suppress thyroid tumorigenesis and progression and CDK4 inhibitors will be effective for treatment of INK4-inactivated or cyclin D-overexpressed thyroid carcinomas.
Collapse
Affiliation(s)
- Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, China
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| | - Xu Zhang
- Department of Pathology, School of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Zhuo Mao
- Department of Physiology, Shenzhen University Health Science Center, Shenzhen, China
| | - He Wen
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Jinshan Ma
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
- Department of Thoracic Surgery, Xinjiang Uigur Autonomous Region People’s Hospital, Xinjiang, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
8
|
Cai W, Shu LZ, Liu DJ, Zhou L, Wang MM, Deng H. Targeting cyclin D1 as a therapeutic approach for papillary thyroid carcinoma. Front Oncol 2023; 13:1145082. [PMID: 37427143 PMCID: PMC10324616 DOI: 10.3389/fonc.2023.1145082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023] Open
Abstract
Cyclin D1 functions as a mitogenic sensor that specifically binds to CDK4/6, thereby integrating external mitogenic inputs and cell cycle progression. Cyclin D1 interacts with transcription factors and regulates various important cellular processes, including differentiation, proliferation, apoptosis, and DNA repair. Therefore, its dysregulation contributes to carcinogenesis. Cyclin D1 is highly expressed in papillary thyroid carcinoma (PTC). However, the particular cellular mechanisms through which abnormal cyclin D1 expression causes PTC are poorly understood. Unveiling the regulatory mechanisms of cyclin D1 and its function in PTC may help determine clinically effective strategies, and open up better opportunities for further research, leading to the development of novel PTC regimens that are clinically effective. This review explores the mechanisms underlying cyclin D1 overexpression in PTC. Furthermore, we discuss the role of cyclin D1 in PTC tumorigenesis via its interactions with other regulatory elements. Finally, recent progress in the development of therapeutic options targeting cyclin D1 in PTC is examined and summarized.
Collapse
Affiliation(s)
- Wei Cai
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lin-Zhen Shu
- Medical College, Nanchang University, Nanchang, China
| | - Ding-Jie Liu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People’s Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Lv Zhou
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Meng-Meng Wang
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
9
|
Zhao F, Zhu S, Fang J, Dong H, Zhu C. Correlation of DNA methylation and lymph node metastasis in papillary thyroid carcinoma. Head Neck 2023. [PMID: 37097909 DOI: 10.1002/hed.27377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/11/2023] [Accepted: 04/09/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer with a primarily good prognosis, and its 10-year survival rate is over 90%. However, PTC is prone to early lymph node metastasis. METHODS Thyroid cancer tissues from PTC patients with lymphatic metastasis and normal tissues were collected for DNA methylation analysis. Different methylation sites, different methylation regions, gene-enriched pathways, and protein-protein interactions (PPIs) were analyzed. RESULTS There were 1004 differentially methylated sites in the PTC group versus the control group; these involved 479 hypermethylated sites in 415 related genes, 525 hypomethylated sites in 482 related genes, 64 differentially methylated regions located in the CpG island region, 34 differentially methylated genes closely related to thyroid cancer, and 17 genes with differentially methylated genes in the DNA promoter region. CONCLUSION NDRG4 hypermethylation and FOXO3, ZEB2, and CDK6 hypomethylation were associated with PTC lymph node metastasis.
Collapse
Affiliation(s)
- Feng Zhao
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyi Zhu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Fang
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huilei Dong
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, Shenyang, China
| | - Chenfang Zhu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Li Q, Tie Y, Alu A, Ma X, Shi H. Targeted therapy for head and neck cancer: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:31. [PMID: 36646686 PMCID: PMC9842704 DOI: 10.1038/s41392-022-01297-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/27/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
Head and neck cancer (HNC) is malignant, genetically complex and difficult to treat and is the sixth most frequent cancer, with tobacco, alcohol and human papillomavirus being major risk factors. Based on epigenetic data, HNC is remarkably heterogeneous, and treatment remains challenging. There is a lack of significant improvement in survival and quality of life in patients with HNC. Over half of HNC patients experience locoregional recurrence or distal metastasis despite the current multiple traditional therapeutic strategies and immunotherapy. In addition, resistance to chemotherapy, radiotherapy and some targeted therapies is common. Therefore, it is urgent to explore more effective and tolerable targeted therapies to improve the clinical outcomes of HNC patients. Recent targeted therapy studies have focused on identifying promising biomarkers and developing more effective targeted therapies. A well understanding of the pathogenesis of HNC contributes to learning more about its inner association, which provides novel insight into the development of small molecule inhibitors. In this review, we summarized the vital signaling pathways and discussed the current potential therapeutic targets against critical molecules in HNC, as well as presenting preclinical animal models and ongoing or completed clinical studies about targeted therapy, which may contribute to a more favorable prognosis of HNC. Targeted therapy in combination with other therapies and its limitations were also discussed.
Collapse
Affiliation(s)
- Qingfang Li
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Tie
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Aqu Alu
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
11
|
Abdelmalak M, Singh R, Anwer M, Ivanchenko P, Randhawa A, Ahmed M, Ashton AW, Du Y, Jiao X, Pestell R. The Renaissance of CDK Inhibitors in Breast Cancer Therapy: An Update on Clinical Trials and Therapy Resistance. Cancers (Basel) 2022; 14:cancers14215388. [PMID: 36358806 PMCID: PMC9655989 DOI: 10.3390/cancers14215388] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Simple Summary Cyclin-dependent kinase inhibitors (palbociclib (Ibrance), ribociclib (Kisqali), and abemaciclib (Verzenio)), targeting aberrant cell-cycle activity have been evaluated extensively in clinical trials. Significant delays in progression free survival and overall survival are now documented with each agent in estrogen receptor positive and human epidermal growth factor receptor two negative advanced breast cancer including luminal B breast cancer. Therapy resistance, driven by chromosomal instability, results in genomic rearrangements, activation of cell-cycle components (cyclin E/cdk2 in Rb− tumors, cyclin D1 in growth factor activated pathways), and the immune response. Molecular analysis of therapy resistant tumors may provide the rational basis for new therapies (brivanib, CYC065, WEE1 kinase and other inhibitors). Luminal B breast cancer is enriched for cyclin D1 overexpression and the chromosomal instability gene signature. The molecular mechanisms governing chromosomal instability in luminal B breast cancer remain poorly understood. Co-targeting of chromosomal instability may potentially reduce the prevalent escape mechanisms that reduce the effectiveness of cyclin-dependent kinase inhibitors. Abstract Cyclin-dependent kinases (CDKs) govern cell-cycle checkpoint transitions necessary for cancer cell proliferation. Recent developments have illustrated nuanced important differences between mono CDK inhibitor (CDKI) treatment and the combination therapies of breast cancers. The CDKIs that are currently FDA-approved for breast cancer therapy are oral agents that selectively inhibit CDK4 and CDK6, include palbociclib (Ibrance), ribociclib (Kisqali), and abemaciclib (Verzenio). CDKI therapy is effective in hormone receptor positive (HR+), and human epidermal growth factor receptor two negative (HER2−) advanced breast cancers (ABC) malignancies, but remains susceptible due to estrogen and progesterone receptor overexpression. Adding a CDK4/6I to endocrine therapy increases efficacy and delays disease progression. Given the side effects of CDKI, identifying potential new treatments to enhance CDKI effectiveness is essential. Recent long-term studies with Palbociclib, including the PALLAS and PENELOPE B, which failed to meet their primary endpoints of influencing progression-free survival, suggest a deeper mechanistic understanding of cyclin/CDK functions is required. The impact of CDKI on the anti-tumor immune response represents an area of great promise. CDKI therapy resistance that arises provides the opportunity for specific types of new therapies currently in clinical trials.
Collapse
Affiliation(s)
- Mary Abdelmalak
- Xavier University School of Medicine, #23, Santa Helenastraat, Oranjestad, Aruba
| | - Rajanbir Singh
- Xavier University School of Medicine, #23, Santa Helenastraat, Oranjestad, Aruba
| | - Mohammed Anwer
- Xavier University School of Medicine, #23, Santa Helenastraat, Oranjestad, Aruba
| | - Pavel Ivanchenko
- Xavier University School of Medicine, #23, Santa Helenastraat, Oranjestad, Aruba
| | - Amritdeep Randhawa
- Xavier University School of Medicine, #23, Santa Helenastraat, Oranjestad, Aruba
| | - Myra Ahmed
- Xavier University School of Medicine, #23, Santa Helenastraat, Oranjestad, Aruba
| | - Anthony W. Ashton
- Xavier University School of Medicine, #23, Santa Helenastraat, Oranjestad, Aruba
- Lankenau Institute for Medical Research Philadelphia, 100 East Lancaster Ave., Wynnewood, PA 19069, USA
| | - Yanming Du
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA 18902, USA
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA 18902, USA
- Xavier University School of Medicine, #23, Santa Helenastraat, Oranjestad, Aruba
- Correspondence: (X.J.); (R.P.)
| | - Richard Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA 18902, USA
- Xavier University School of Medicine, #23, Santa Helenastraat, Oranjestad, Aruba
- The Wistar Cancer Center, Philadelphia, PA 19107, USA
- Correspondence: (X.J.); (R.P.)
| |
Collapse
|
12
|
Posttranslational Modifications in Thyroid Cancer: Implications for Pathogenesis, Diagnosis, Classification, and Treatment. Cancers (Basel) 2022; 14:cancers14071610. [PMID: 35406382 PMCID: PMC8996999 DOI: 10.3390/cancers14071610] [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] [Received: 02/25/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
There is evidence that posttranslational modifications, including phosphorylation, acetylation, methylation, ubiquitination, sumoylation, glycosylation, and succinylation, may be involved in thyroid cancer. We review recent reports supporting a role of posttranslational modifications in the tumorigenesis of thyroid cancer, sensitivity to radioiodine and other types of treatment, the identification of molecular treatment targets, and the development of molecular markers that may become useful as diagnostic tools. An increased understanding of posttranslational modifications may be an important supplement to the determination of alterations in gene expression that has gained increasing prominence in recent years.
Collapse
|
13
|
Lee WK, Cheng SY. Targeting transcriptional regulators for treatment of anaplastic thyroid cancer. JOURNAL OF CANCER METASTASIS AND TREATMENT 2021; 7. [PMID: 34761120 PMCID: PMC8577520 DOI: 10.20517/2394-4722.2021.58] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dysregulation of genes perpetuates cancer progression. During carcinogenesis, cancer cells acquire dependency of aberrant transcriptional programs (known as “transcription addiction”) to meet the high demands for uncontrolled proliferation. The needs for particular transcription programs for cancer growth could be cancer-type-selective. The dependencies of certain transcription regulators could be exploited for therapeutic benefits. Anaplastic thyroid cancer (ATC) is an extremely aggressive human cancer for which new treatment modalities are urgently needed. Its resistance to conventional treatments and the lack of therapeutic options for improving survival might have been attributed to extensive genetic heterogeneity due to subsequent evolving genetic alterations and clonal selections during carcinogenesis. Despite this genetic complexity, mounting evidence has revealed a characteristic transcriptional addiction of ATC cells resulting in evolving diverse oncogenic signaling for cancer cell survival. The transcriptional addiction has presented a huge challenge for effective targeting as shown by the failure of previous targeted therapies. However, an emerging notion is that many different oncogenic signaling pathways activated by multiple upstream driver mutations might ultimately converge on the transcriptional responses, which would provide an opportunity to target transcriptional regulators for treatment of ATC. Here, we review the current understanding of how genetic alterations in cancer distorted the transcription program, leading to acquisition of transcriptional addiction. We also highlight recent findings from studies aiming to exploit the opportunity for targeting transcription regulators as potential therapeutics for ATC.
Collapse
Affiliation(s)
- Woo Kyung Lee
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
14
|
Niciporuka R, Nazarovs J, Ozolins A, Narbuts Z, Miklasevics E, Gardovskis J. Can We Predict Differentiated Thyroid Cancer Behavior? Role of Genetic and Molecular Markers. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:1131. [PMID: 34684168 PMCID: PMC8540789 DOI: 10.3390/medicina57101131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/07/2021] [Accepted: 10/15/2021] [Indexed: 12/27/2022]
Abstract
Thyroid cancer is ranked in ninth place among all the newly diagnosed cancer cases in 2020. Differentiated thyroid cancer behavior can vary from indolent to extremely aggressive. Currently, predictions of cancer prognosis are mainly based on clinicopathological features, which are direct consequences of cell and tissue microenvironment alterations. These alterations include genetic changes, cell cycle disorders, estrogen receptor expression abnormalities, enhanced epithelial-mesenchymal transition, extracellular matrix degradation, increased hypoxia, and consecutive neovascularization. All these processes are represented by specific genetic and molecular markers, which can further predict thyroid cancer development, progression, and prognosis. In conclusion, evaluation of cancer genetic and molecular patterns, in addition to clinicopathological features, can contribute to the identification of patients with a potentially worse prognosis. It is essential since it plays a crucial role in decision-making regarding initial surgery, postoperative treatment, and follow-up. To date, there is a large diversity in methodologies used in different studies, frequently leading to contradictory results. To evaluate the true significance of predictive markers, more comparable studies should be conducted.
Collapse
Affiliation(s)
- Rita Niciporuka
- Department of Surgery, Riga Stradins University, Pilsonu Street 13, LV-1002 Riga, Latvia; (A.O.); (Z.N.); (J.G.)
- Department of Surgery, Pauls Stradins Clinical University Hospital, Pilsonu Street 13, LV-1002 Riga, Latvia
| | - Jurijs Nazarovs
- Department of Pathology, Pauls Stradins Clinical University Hospital, Pilsonu Street 13, LV-1002 Riga, Latvia;
| | - Arturs Ozolins
- Department of Surgery, Riga Stradins University, Pilsonu Street 13, LV-1002 Riga, Latvia; (A.O.); (Z.N.); (J.G.)
- Department of Surgery, Pauls Stradins Clinical University Hospital, Pilsonu Street 13, LV-1002 Riga, Latvia
| | - Zenons Narbuts
- Department of Surgery, Riga Stradins University, Pilsonu Street 13, LV-1002 Riga, Latvia; (A.O.); (Z.N.); (J.G.)
- Department of Surgery, Pauls Stradins Clinical University Hospital, Pilsonu Street 13, LV-1002 Riga, Latvia
| | - Edvins Miklasevics
- Institute of Oncology, Riga Stradins University, Pilsonu Street 13, LV-1002 Riga, Latvia;
| | - Janis Gardovskis
- Department of Surgery, Riga Stradins University, Pilsonu Street 13, LV-1002 Riga, Latvia; (A.O.); (Z.N.); (J.G.)
- Department of Surgery, Pauls Stradins Clinical University Hospital, Pilsonu Street 13, LV-1002 Riga, Latvia
| |
Collapse
|
15
|
Wang L, Chen Y, Li H, Xu Q, Liu R. The cyclin-dependent kinase inhibitor AT7519 augments cisplatin's efficacy in ovarian cancer via multiple oncogenic signaling pathways. Fundam Clin Pharmacol 2021; 36:81-88. [PMID: 34212421 DOI: 10.1111/fcp.12709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 06/22/2021] [Indexed: 11/27/2022]
Abstract
Although cisplatin is the most active drug for the treatment of ovarian cancer, majority of patients develop resistance and ultimately relapse. Enhancing the efficacy of cisplatin could represent a promising strategy to improve the clinical outcome of patients with ovarian cancer. AT7519 is a multitargeted cyclin-dependent kinase (CDK) inhibitor and displays potent anticancer activities. In this work, we show that the combination of AT7519 with cisplatin is much more superior to cisplatin alone in inhibiting ovarian cancer. AT7519 at nanomolar concentrations inhibits proliferation and migration and induces apoptosis of multiple ovarian cancer cell lines. In contrast, AT7519 at the same concentrations either does not affect survival or is significantly less effective in inhibiting proliferation and migration in normal ovarian cells and fibroblast cells. AT7519 significantly augments the inhibitory effects of cisplatin in ovarian cancer cells in a dose-dependent manner. Mechanistic studies suggest that AT7519 (i) inhibits proliferation via decreasing activities of CDK1 and 2, and via inhibiting RNA transcription; (ii) inhibits migration via suppressing epithelial-mesenchymal transition (EMT); and (iii) induces apoptosis via decreasing Mcl-1 and increasing Bim in ovarian cancer cells. Using a human ovarian cancer xenograft mouse model, we confirm the in vivo efficacy of AT7519 alone, and the synergistic effects of AT7519 and cisplatin in combination, at doses that cause minimal toxicity in mice. Our findings provide systematic preclinical evidence to support the initialization of clinical trials of the AT7519 and cisplatin combination for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Ling Wang
- Department of Obstetrics and Gynaecology, The Second Clinical Medical College, Yangtze University, Jingzhou Central Hospital, Jingzhou, China
| | - Yan Chen
- Department of Obstetrics and Gynaecology, The Second Clinical Medical College, Yangtze University, Jingzhou Central Hospital, Jingzhou, China
| | - Hui Li
- Department of Obstetrics and Gynaecology, The Second Clinical Medical College, Yangtze University, Jingzhou Central Hospital, Jingzhou, China
| | - Qiqi Xu
- Department of Obstetrics and Gynaecology, The Second Clinical Medical College, Yangtze University, Jingzhou Central Hospital, Jingzhou, China
| | - Rong Liu
- Department of Obstetrics and Gynaecology, The Second Clinical Medical College, Yangtze University, Jingzhou Central Hospital, Jingzhou, China
| |
Collapse
|
16
|
Marinelli O, Romagnoli E, Maggi F, Nabissi M, Amantini C, Morelli MB, Santoni M, Battelli N, Santoni G. Exploring treatment with Ribociclib alone or in sequence/combination with Everolimus in ER +HER2 -Rb wild-type and knock-down in breast cancer cell lines. BMC Cancer 2020; 20:1119. [PMID: 33213401 PMCID: PMC7678099 DOI: 10.1186/s12885-020-07619-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 11/06/2020] [Indexed: 11/10/2022] Open
Abstract
Background Breast cancer (BC) is the second most common type of cancer worldwide. Among targeted therapies for Hormone Receptor-positive (HR+) and Human Epidermal growth factor Receptor 2-negative (HER2−) BC, the Cyclin-Dependent Kinases (CDK4/6) are targeted by inhibitors such as Ribociclib (Rib); however, resistance to CDK4/6 inhibitors frequently develops. The aim of this work is to assess in vitro activity of Rib and Everolimus (Eve) in HR+HER2− MCF-7 and HR−HER2−BT-549 BC cell lines. Methods HR+HER2− MCF-7 and HR−HER2− BT-549 BC cell lines were treated with increasing concentration of Rib and Eve (up to 80 μg/mL) for 48–72 h. Subsequently, HR+HER2− MCF-7 cells were silenced for Retinoblastoma (Rb) gene, and thus, the effect of Rib in sequential or concurrent schedule with Eve for the treatment of both Rb wild type or Rb knock-down MCF-7 in vitro was evaluated. Cell viability of HR+HER2− MCF-7cells treated with sequential and concurrent dosing schedule was analyzed by MTT assay. Moreover, cell cycle phases, cell death and senescence were evaluated by cytofluorimetric analysis after treatment with Rib or Eve alone or in combination. Results The sequential treatment didn’t produce a significant increase of cytotoxicity, compared to Rib alone. Instead, the cotreatment synergized to increase the cytotoxicity compared to Rib alone. The cotreatment reduced the percentage of cells in S and G2/M phases and induced apoptosis. Rib triggered senescence and Eve completely reversed this effect in Rb wild type BC cells. Rib also showed Rb-independent effects as shown by results in Rb knock-down MCF-7. Conclusion Overall, the Rib/Eve concurrent therapy augmented the in vitro cytotoxic effect, compared to Rib/Eve sequential therapy or single treatments. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-020-07619-1.
Collapse
Affiliation(s)
| | | | - Federica Maggi
- School of Pharmacy, University of Camerino, 62032, Camerino, MC, Italy.,Department of Molecular Medicine, University of Rome Sapienza, Rome, Italy
| | - Massimo Nabissi
- School of Pharmacy, University of Camerino, 62032, Camerino, MC, Italy
| | - Consuelo Amantini
- School of Bioscience and Veterinary Medicine, University of Camerino, Camerino, MC, Italy
| | | | - Matteo Santoni
- Medical Oncology Unit, Hospital of Macerata, Macerata, Italy
| | - Nicola Battelli
- Medical Oncology Unit, Hospital of Macerata, Macerata, Italy
| | - Giorgio Santoni
- School of Pharmacy, University of Camerino, 62032, Camerino, MC, Italy.
| |
Collapse
|
17
|
Huang Y, Wu H, Li X. Novel sequential treatment with palbociclib enhances the effect of cisplatin in RB-proficient triple-negative breast cancer. Cancer Cell Int 2020; 20:501. [PMID: 33061853 PMCID: PMC7552520 DOI: 10.1186/s12935-020-01597-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/06/2020] [Indexed: 12/24/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a highly aggressive malignancy that lacks sensitivity to chemotherapy, endocrine therapy or targeted therapy. CDK4/6 inhibitors, combined with endocrine therapy, have been shown to be effective in postmenopausal women with HR-positive, HER2-negative advanced or metastatic breast cancer. Therefore, we investigated whether the CDK4/6 inhibitor palbociclib (PD) could enhance the effects of cisplatin (CDDP) on TNBC. Methods The effects of different drug regimens consisting of PD and CDDP on MDA-MB-231 and RB-knockdown MDA-MB-231 (sh-MDA-MB-231) cells were assessed in vitro and in vivo. MDA-MB-468 and RB-overexpressing MDA-MB-468 cells were used to assess the effect of the PD-CDDP regimens in vitro. Immunoblotting illustrated the role of the cyclin D1/RB/E2F axis signalling pathway. Results PD induced G1 phase cell cycle arrest in the MDA-MB-231 cell line. However, synchronous treatment with PD and CDDP for 24 h, treatment with PD for 24 h followed by CDDP and treatment with CDDP for 24 h followed by PD had no influence on MDA-MB-231 cell apoptosis. We further investigated the effect of PD or CDDP withdrawal on the effects of sequential treatment and found that PD treatment for 48 h followed by withdrawal for 48 h and subsequent CDDP treatment (PD-CDDP) significantly increased apoptosis and inhibited the cell viability and colony formation of MDA-MB-231 cells, while with other regimens, PD and CDDP had an additive or antagonistic response. The preferential use of PD increased DNA damage induced by CDDP, as measured through γH2AX immunofluorescence. These findings were not observed in sh-MDA-MB-231 cells, and experiments to assess cell function in MDA-MB-468 and RB-overexpressing MDA-MB-468 cells yielded similar results, which indicated that PD enhanced the sensitivity of TNBC cells to CDDP in an RB-dependent manner. In vivo, compared with single drug treatment, combination treatment inhibited tumour growth and Ki-67 expression in MDA-MB-231 xenograft models. Western blot analysis revealed that PD enhanced sensitivity to CDDP through the CDK4/6-cyclin D1-RB-E2F pathway. Conclusions Pre-treatment with PD synchronized the tumour cell cycle through the CDK4/6-cyclin D1-RB-E2F pathway, which increased the antitumour effect of CDDP. Thus, PD-CDDP might be an effective treatment for RB-proficient TNBC patients.
Collapse
Affiliation(s)
- Yajing Huang
- Department of Breast and Thyroid Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Hao Wu
- Department of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Xingrui Li
- Department of Breast and Thyroid Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| |
Collapse
|
18
|
Oshi M, Takahashi H, Tokumaru Y, Yan L, Rashid OM, Nagahashi M, Matsuyama R, Endo I, Takabe K. The E2F Pathway Score as a Predictive Biomarker of Response to Neoadjuvant Therapy in ER+/HER2- Breast Cancer. Cells 2020; 9:E1643. [PMID: 32650578 PMCID: PMC7407968 DOI: 10.3390/cells9071643] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/28/2020] [Accepted: 07/07/2020] [Indexed: 12/20/2022] Open
Abstract
E2F transcription factors play critical roles in the cell cycle. Therefore, their activity is expected to reflect tumor aggressiveness and responsiveness to therapy. We scored 3905 tumors of nine breast cancer cohorts for this activity based on their gene expression for the Hallmark E2F targets gene set. As expected, tumors with a high score had an increased expression of cell proliferation-related genes. A high score was significantly associated with shorter patient survival, greater MKI67 expression, histological grade, stage, and genomic aberrations. Furthermore, metastatic tumors had higher E2F scores than the primary tumors from which they arose. Although tumors with a high score had greater infiltration by both pro- and anti-cancerous immune cells, they had an increased expression of immune checkpoint genes. Estrogen receptor (ER)-positive/human epidermal growth factor receptor 2 (HER2)-negative cancer with a high E2F score achieved a significantly higher pathological complete response (pCR) rate to neoadjuvant chemotherapy. The E2F score was significantly associated with the expression of cyclin-dependent kinase (CDK)-related genes and strongly correlated with sensitivity to CDK inhibition in cell lines. In conclusion, the E2F score is a marker of breast cancer aggressiveness and predicts the responsiveness of ER-positive/HER2-negative patients to neoadjuvant chemotherapy and possibly to CDK and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Masanori Oshi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (H.T.); (Y.T.)
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama 2360004, Japan; (R.M.); (I.E.)
| | - Hideo Takahashi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (H.T.); (Y.T.)
| | - Yoshihisa Tokumaru
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (H.T.); (Y.T.)
- Department of Surgical Oncology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Li Yan
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Omar M. Rashid
- Department of Surgery, Holy Cross Hospital, Michael and Dianne Bienes Comprehensive Cancer Center, Fort Lauderdale, FL 33308, USA;
- Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 9518520, Japan;
| | - Ryusei Matsuyama
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama 2360004, Japan; (R.M.); (I.E.)
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama 2360004, Japan; (R.M.); (I.E.)
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (H.T.); (Y.T.)
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama 2360004, Japan; (R.M.); (I.E.)
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima 9601295, Japan
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14263, USA
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 9518510, Japan
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 1608402, Japan
| |
Collapse
|
19
|
Wen HL, Xu ZM, Wen D, Lin SY, Liang Y, Xie JP. Long noncoding RNAs SET-binding factor 2-antisense RNA1 promotes cell growth through targeting miR-431-5p/CDK14 axis in human papillary thyroid cancer. Kaohsiung J Med Sci 2020; 36:808-816. [PMID: 32602632 DOI: 10.1002/kjm2.12259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/06/2020] [Accepted: 06/03/2020] [Indexed: 12/14/2022] Open
Abstract
Papillary thyroid cancer (PTC) is a frequent thyroid malignancy. With the significant regulatory role in tumor progression, more attention has been employed to investigate mechanism of long noncoding RNAs (lncRNAs) in progression of PTC. We prospectively explored the mechanism whereby lncRNA SET-binding factor 2-antisense RNA1 (SBF2-AS1) is implicated in pathogenesis of PTC. First, differentially expressed SBF2-AS1 between PTC and normal adjacent thyroid tissues was determined, and result indicated a higher SBF2-AS1 expression in PTC tissues than adjacent normal tissues. Moreover, highly SBF2-AS1 expression predicted a poor prognosis in PTC patients. Second, SBF2-AS1 overexpression promoted cell viability and cycle of PTC, while inhibited cell apoptosis. However, SBF2-AS1 downregulation reduced viability and cycle, while promoted cell apoptosis. Moreover, SBF2-AS1 could bind with miR-431-5p and showed negative correlation with miR-431-5p in PTC patients. Furthermore, miR-431-5p bind with cyclin-dependent kinase (CDK) 14 and showed negative correlation with CDK14 in PTC patients. Finally, overexpression of CDK14 counteracted with the inhibitory role of SBF2-AS1 downregulation on cell viability, cycle, and apoptosis of PTC. In conclusion, SBF2-AS1 exhibited oncogenic property in PTC, and knockdown of SBF2-AS1 could be a therapeutic strategy for PTC.
Collapse
Affiliation(s)
- Hu-Ling Wen
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zheng-Min Xu
- Institute of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Dan Wen
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shi-Yu Lin
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yu Liang
- Department of Ultrasonography, Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Jian-Ping Xie
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
20
|
Sanjari M, Kordestani Z, Safavi M, Mashrouteh M, FekriSoofiAbadi M, Ghaseminejad Tafreshi A. Enhanced expression of Cyclin D1 and C-myc, a prognostic factor and possible mechanism for recurrence of papillary thyroid carcinoma. Sci Rep 2020; 10:5100. [PMID: 32198408 PMCID: PMC7083882 DOI: 10.1038/s41598-020-61985-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/03/2020] [Indexed: 12/13/2022] Open
Abstract
A direct association has been shown between Cyclin D1 and C-myc gene expressions and the proliferation of human thyroid tumor cells. Our previous study showed that increased β catenin led to a reduction in disease-free probability in patients with papillary thyroid cancer. This study was designed to investigate Cyclin D1 and C-myc genes as targets for β catenin function in PTC and to determine the association between genes expression and staging, recurrence, metastasis, and disease-free survival of PTC. This study was conducted via a thorough investigation of available data from medical records as well as paraffin blocks of 77 out of 400 patients over a 10-year period. Cyclin D1 and C-myc gene expression levels were measured using real-time polymerase chain reaction (RT-PCR) and the Kaplan-Meier method was used to evaluate disease-free survival. Higher levels of Cyclin D1 and C-myc gene expressions were observed in patients with recurrence by 8.5 (P = 0.004) and 19.5 (p = 0.0001) folds, respectively. A significant positive correlation was found between Cyclin D1 expression and the cumulative dose of radioactive iodine received by patients (r = −0.2, p value = 0.03). The ten-year survival rate in the patients included in this study was 98.25% while disease-free survival was 48.1%. Higher Cyclin D1 and C-myc gene expression levels were observed in patients with recurrence/distant metastasis. Inversely, lower expression of Cyclin D1 and C-myc genes were associated with better survival of patients (SD, 0.142-0.052) (Mantel-Cox test, P = 0.002). The enhancement of Cyclin D1 and C-myc gene expression may be a potential mechanism for recurrence and aggressiveness of PTC.
Collapse
|
21
|
Li T, Xiong Y, Wang Q, Chen F, Zeng Y, Yu X, Wang Y, Zhou F, Zhou Y. Ribociclib (LEE011) suppresses cell proliferation and induces apoptosis of MDA-MB-231 by inhibiting CDK4/6-cyclin D-Rb-E2F pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:4001-4011. [PMID: 31588803 DOI: 10.1080/21691401.2019.1670670] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Triple-negative breast cancer (TNBC) stands for a refractory subtype, which predicts poor prognosis and has no effective therapies yet for improving it. Given the restrictions of traditional treatments, novel therapeutic strategies need excavating to alleviate the intrinsic or acquired resistance. Ribociclib, a selective CDK4/6 inhibitor, has successfully prevented cancers from deteriorating by intervening the CDK4/6-cyclin D-Rb-E2F pathway, especially for estrogen receptor-positive (ER +) breast cancer. However, there still remains limited accessibility referring to TNBC. Performing experiments on MDA-MB-231 cells, we found that LEE011 could suppress cell proliferation, and this suppression tended to be dose-dependently. Western blotting analysis presented significant decrease with the expression of CDK4/6 after LEE011 treated, and other proteins associated with this axis such as cyclin D1, p-Rb, Rb, E2F1 showed aberrant changes. Moreover, LEE011 induced G0-G1 phase cell cycle arrest, promoted cell apoptosis, and reduced cell migration in vitro. In addition, tumor growth was remarkably impeded without obvious side-effects in MDA-MB-231 xenograft models. Our research has identified that LEE011 was not completely invalid for MDA-MB-231. Considering its pivotal status in TNBC, the CDK4/6-cyclin D-Rb-E2F pathway informed us the possibility and practicality of Ribociclib (LEE011) as pharmacological intervention, but challenges warrant further validation in prospective studies.
Collapse
Affiliation(s)
- Tianqi Li
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University , Wuhan , China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University , Wuhan , China
| | - Yudi Xiong
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University , Wuhan , China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University , Wuhan , China
| | - Qingqing Wang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University , Wuhan , China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University , Wuhan , China
| | - Fengxia Chen
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University , Wuhan , China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University , Wuhan , China
| | - Yangyang Zeng
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University , Wuhan , China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University , Wuhan , China
| | - Xiaoyan Yu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University , Wuhan , China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University , Wuhan , China
| | - Yuan Wang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University , Wuhan , China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University , Wuhan , China
| | - Fuxiang Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University , Wuhan , China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University , Wuhan , China
| | - Yunfeng Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University , Wuhan , China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University , Wuhan , China
| |
Collapse
|
22
|
Pozdeyev N, Rose MM, Bowles DW, Schweppe RE. Molecular therapeutics for anaplastic thyroid cancer. Semin Cancer Biol 2020; 61:23-29. [PMID: 31991166 DOI: 10.1016/j.semcancer.2020.01.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023]
Abstract
Anaplastic thyroid cancer (ATC) represents one of the most lethal human cancers and although this tumor type is rare, ATC accounts for the majority of deaths from thyroid cancer. Due to the rarity of ATC, a comprehensive genomic characterization of this tumor type has been challenging, and thus the development of new therapies has been lacking. To date, there is only one mutation-driven targeted therapy for BRAF-mutant ATC. Recent genomic studies have used next generation sequencing to define the genetic landscape of ATC in order to identify new therapeutic targets. Together, these studies have confirmed the role of oncogenic mutations of MAPK pathway as key drivers of differentiated thyroid cancer (BRAF, RAS), and that additional genetic alterations in the PI3K pathway, TP53, and the TERT promoter are necessary for anaplastic transformation. Recent novel findings have linked the high mutational burden associated with ATC with mutations in the Mismatch Repair (MMR) pathway and overactivity of the AID/APOBEC family of cytidine deaminases. Additional novel mutations include cell cycle genes, SWI/SNF chromatin remodeling complex, and histone modification genes. Mutations in RAC1 were also identified in ATC, which have important implications for BRAF-directed therapies. In this review, we summarize these novel findings and the new genetic landscape of ATC. We further discuss the development of therapies targeting these pathways that are being tested in clinical and preclinical studies.
Collapse
Affiliation(s)
- Nikita Pozdeyev
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, Aurora, CO, USA; Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, Aurora, CO, USA
| | - Madison M Rose
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, Aurora, CO, USA
| | - Daniel W Bowles
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rebecca E Schweppe
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, Aurora, CO, USA.
| |
Collapse
|
23
|
Fazeli S, Paal E, Maxwell JH, Burman KD, Nylen ES, Khosla SG. Salutary Response to Targeted Therapy in Anaplastic Thyroid Cancer. J Investig Med High Impact Case Rep 2019; 7:2324709619890942. [PMID: 31766881 PMCID: PMC6880026 DOI: 10.1177/2324709619890942] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Context. Anaplastic thyroid cancer (ATC) is an aggressive tumor
with a median survival of 3 to 9 months, a 1-year survival of less than 10% and
without definitive therapies. Recently, in BRAF V600E mutated
ATCs, new targeted therapy using a combination of a BRAF inhibitor, dabrafenib
(Dab), with a mitogen-activated extracellular protein kinase (MEK) inhibitor,
trametinib (Tram), has shown significant promise. Case
Description. We report a case of aggressive ATC with 5 sequence
mutations: BRAF V600E (mutation fraction [MF] 34%),
TERT E441del (MF 37%), RET N579K (MF 55%),
EZH2 D154E (MF 60%), and CDK4 S259L (MF
48%). The patient had a dramatic response to the Dab/Tram combination with near
complete resolution of his lung, bone, hepatic, and splenic lesions soon after
starting therapy. Unfortunately, intolerable side effects (grade 2-3) on this
regimen required tapering and discontinuation of the treatment. He had a quick
resurgence of disease after stopping the combination therapy. The patient died
approximately 3 months after discontinuing Dab/Tram. Autopsy revealed an
atrophic thyroid gland with microscopic subcapsular focus of well-differentiated
papillary thyroid carcinoma. There was extensive lymphatic spread of the tumor
throughout bilateral lungs with fibrosis. No other metastatic site was
identified. Conclusion. We report a unique case of ATC with 2
new mutations of EZH2 D154E and CDK S529L.
This case exemplifies the significant promise Dab/Tram therapy holds, the
potential side effects that limit their use, and autopsy findings status post
use of this combination therapy.
Collapse
Affiliation(s)
- Sasan Fazeli
- George Washington University Medical Faculty Associates, Washington, DC, USA.,Washington DC VA Medical Center, Washington, DC, USA
| | - Edina Paal
- Washington DC VA Medical Center, Washington, DC, USA.,George Washington University, Washington, DC, USA
| | - Jessica H Maxwell
- Washington DC VA Medical Center, Washington, DC, USA.,MedStar Georgetown University Medical Center, Washington, DC, USA
| | - Kenneth D Burman
- MedStar Georgetown University Medical Center, Washington, DC, USA.,MedStar Washington Hospital Center, Washington, DC, USA
| | - Eric S Nylen
- George Washington University Medical Faculty Associates, Washington, DC, USA.,Washington DC VA Medical Center, Washington, DC, USA
| | - Shikha G Khosla
- George Washington University Medical Faculty Associates, Washington, DC, USA.,Washington DC VA Medical Center, Washington, DC, USA
| |
Collapse
|
24
|
Jin D, Tran N, Thomas N, Tran DD. Combining CDK4/6 inhibitors ribociclib and palbociclib with cytotoxic agents does not enhance cytotoxicity. PLoS One 2019; 14:e0223555. [PMID: 31600301 PMCID: PMC6786609 DOI: 10.1371/journal.pone.0223555] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022] Open
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4/6) play critical roles in the G1 to S checkpoint of the cell cycle and have been shown to be overactive in several human cancers. Small-molecule inhibitors of CDK4/6 have demonstrated significant efficacy against many solid tumors. Since CDK4/6 inhibition is thought to induce cell cycle arrest at the G1/S checkpoint, much interest has been focused on combining CDK4/6 inhibitors with cytotoxic agents active against the S or M phase of the cell cycle to enhance therapeutic efficacy. However, it remains unclear how best to combine these two classes of drugs to avoid their potentially antagonistic effects. Here, we test various combinations of highly selective and potent CDK4/6 inhibitors with commonly used cytotoxic drugs in several cancer cell lines derived from lung, breast and brain cancers, for their cell-killing effects as compared to monotherapy. All combinations, either concurrent or sequential, failed to enhance cell-killing effects. Importantly, in certain schedules, especially pre-treatment with a CDK4/6 inhibitor, combining these drugs resulted in reduced cytotoxicity of cytotoxic agents. These findings urge cautions when combining these two classes of agents in clinical settings.
Collapse
Affiliation(s)
- Dan Jin
- Division of Neuro-Oncology, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, Gainesville, FL, United States of America
| | - Nguyen Tran
- Division of Neuro-Oncology, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, Gainesville, FL, United States of America
| | - Nagheme Thomas
- Division of Neuro-Oncology, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, Gainesville, FL, United States of America
| | - David D Tran
- Division of Neuro-Oncology, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, Gainesville, FL, United States of America
| |
Collapse
|
25
|
A Systematic Review of Phase II Targeted Therapy Clinical Trials in Anaplastic Thyroid Cancer. Cancers (Basel) 2019; 11:cancers11070943. [PMID: 31277524 PMCID: PMC6678800 DOI: 10.3390/cancers11070943] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 12/19/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is a rare, but devastating disease. Despite multimodal approaches combining surgery, chemotherapy and radiation therapy, ATC is associated with a dire prognosis, with a median overall survival of only three to ten months. Novel treatments are thus urgently needed. Recent efforts towards the characterization of the molecular landscape of ATC have led to the identification of pro-oncogenic targetable alterations, lending promise for novel targeted therapeutic approaches. This systematic review summarizes the results of phase II clinical trials of targeted therapy in ATC, providing an overview of efficacy and safety profiles. The majority of trials to date have consisted of small single-arm studies and have presented modest results. However, only a minority of trials have selected or stratified patients by molecular alterations. In the setting of BRAF V600E mutated ATC, dabrafenib/trametinib combination therapy and vemurafenib monotherapy have both demonstrated efficacy. Everolimus has furthermore shown promising results in patients with PI3K/mTOR/AKT pathway alterations. These studies underscore the importance of molecular profiling of tumors for appropriate patient selection and determination of genomic correlates of response. Clinical trials are underway testing additional targeted therapies as monotherapy, or as a part of multimodal treatment, and in combination with immunotherapy.
Collapse
|
26
|
Yuan Y, Wen W, Yost SE, Xing Q, Yan J, Han ES, Mortimer J, Yim JH. Combination therapy with BYL719 and LEE011 is synergistic and causes a greater suppression of p-S6 in triple negative breast cancer. Sci Rep 2019; 9:7509. [PMID: 31101835 PMCID: PMC6525251 DOI: 10.1038/s41598-019-43429-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 04/15/2019] [Indexed: 02/07/2023] Open
Abstract
A third of patients with triple negative breast cancer (TNBC) have relapsed disease within 2-5 years from initial diagnosis, leaving an unmet need for therapeutic targets. TNBC frequently harbors alterations of the PI3K/AKT/mTOR pathway, but single agent PI3K/AKT/mTOR inhibitors have not shown marked efficacy. In this study, we investigated a strategy to improve efficacy of PI3K-α inhibitor BYL719 (alpelisib) in TNBC. While BYL719 is effective at inhibiting cell proliferation in T47D, a triple positive cell line, it had limited activity in TNBC. This may be partially due to persistent phosphorylation of RB, and incomplete inhibition of p-S6 in TNBC, since the inhibitory effect of BYL719 on p-RB and p-S6 was significantly reduced in TNBC compared to T47D cells. Addition of the CDK4/6 inhibitor LEE011 to BYL719 caused a simultaneous reduction of p-RB and p-S6, and a more complete inhibition of p-S6, leading to decreased expression of the pro-survival protein MCL-1, an induction of apoptosis, and an enhanced reduction of tumor growth in a PDX model of TNBC. These findings suggest that inhibition of p-RB and p-S6 is important for an effective response to the treatment of TNBC, and provides a strong rationale for clinical development of combination therapy with BYL719 and LEE011 for treatment of metastatic TNBC with intact RB.Presentation: This study was presented in part as an abstract at the 2016 San Antonio Breast Cancer Symposium (P3-03-15) and the 2018 Cancer Research and Targeted Therapy in London.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Medical Oncology & Molecular Therapeutics, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, USA.
| | - Wei Wen
- Division of Surgical Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, USA
| | - Susan E Yost
- Department of Medical Oncology & Molecular Therapeutics, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, USA
| | - Quanhua Xing
- Division of Surgical Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, USA
| | - Jin Yan
- Division of Surgical Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, USA
| | - Ernest S Han
- Division of Gynecologic Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, USA
| | - Joanne Mortimer
- Department of Medical Oncology & Molecular Therapeutics, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, USA
| | - John H Yim
- Division of Surgical Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, USA.
| |
Collapse
|
27
|
Wong K, Di Cristofano F, Ranieri M, De Martino D, Di Cristofano A. PI3K/mTOR inhibition potentiates and extends palbociclib activity in anaplastic thyroid cancer. Endocr Relat Cancer 2019; 26:425-436. [PMID: 30699064 PMCID: PMC6602869 DOI: 10.1530/erc-19-0011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/30/2019] [Indexed: 12/28/2022]
Abstract
Anaplastic thyroid carcinoma (ATC) is the most aggressive form of thyroid cancer. Despite its low incidence, it accounts for a disproportionate number of thyroid cancer-related deaths, because of its resistance to current therapeutic approaches. Novel actionable targets are urgently needed to prolong patient survival and increase their quality of life. Loss and mutation of the RB1 tumor suppressor are rare events in ATC, which suggests that therapies directed at inhibiting the cyclin D/CDK4 complexes, responsible for RB phosphorylation and inactivation, might be effective in this tumor type. In fact, we found that the CDK4/6 inhibitor, palbociclib, strongly inhibits proliferation in all the RB1 wild type ATC cell lines tested. Efficacy was also observed in vivo, in a xenograft model. However, ATC cells rapidly developed resistance to palbociclib. Resistance was associated with increased levels of cyclin D1 and D3. To counter cyclin D overexpression, we tested the effect of combining palbociclib with the PI3K/mTOR dual inhibitor, omipalisib. Combined treatment synergistically reduced cell proliferation, even in cell lines that do not carry PI3K-activating mutations. More importantly, low-dose combination was dramatically effective in inhibiting tumor growth in a xenograft model. Thus, combined PI3K/mTOR and CDK4/6 inhibition is a highly promising novel approach for the treatment of aggressive, therapy-resistant thyroid cancer.
Collapse
Affiliation(s)
| | | | | | | | - Antonio Di Cristofano
- A. Di Cristofano, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Room 302, Bronx, NY 10461., Tel: 718-678-1137,
| |
Collapse
|
28
|
Xiong Y, Li T, Assani G, Ling H, Zhou Q, Zeng Y, Zhou F, Zhou Y. Ribociclib, a selective cyclin D kinase 4/6 inhibitor, inhibits proliferation and induces apoptosis of human cervical cancer in vitro and in vivo. Biomed Pharmacother 2019; 112:108602. [PMID: 30784916 DOI: 10.1016/j.biopha.2019.108602] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 11/20/2022] Open
Abstract
Cervical cancer remains one of the main factors leading to tumor-related death worldwide. Many strategies of cancer treatment such as chemotherapy are developed and used nowadays. However, for the cancer chemotherapy resistance, reduction of the limitation of cancer chemotherapy efficacy is one of the aims of several oncology teams. Moreover, the cyclin-dependent kinase 4/6-cyclin D-retinoblastoma protein-E2F pathway is an important mechanism for cell cycle control and its dysregulation is one of the key factors for cancers development including cervical cancer. Ribociclib is one of the selective CDK4/6 inhibitors and is a new therapeutic approach showing promise as a good strategy of therapy in many human cancers. However, there are not the studies regarding the investigation of effects of Ribociclib in cervical cancer yet. In the present study, by western blotting and immunofluorescence assay, we found respectively that CDK4, CDK6 and cyclin D1 are highly expressed and are mostly localized in the nucleus with some localized in the cytoplasm of cervical cancer cell lines. Moreover, Ribociclib induced cell cycle arrest in G0-G1 phase and cell apoptosis, and inhibited C33A cell proliferation in dose - dependent manner following by decreased expression of certain related genes such as CDK4, CDK6, E2F1, P-Rb, and increased Bax expression. In C33A xenografts, Ribociclib inhibited tumor growth associated with decreased expressions of CDK4, CDK6, cyclin D1, Rb and Ki-67, and also significantly increased tumor cell apoptosis. However, we didn't find side effect of Ribociclib concerning heart, liver and kidney perturbation and any Ribociclib anti-tumor effects on HeLa in vitro and in vivo which may be due to Hela cell infection by HPV. Based on our findings, the Rb-E2F pathway can be considered as an important factor in human cervical cancer pathogenesis and as a mechanism of Ribociclib, a potential strategy of treatment for the improvement of new therapeutic measures for the treatment of HPV-negative cervical cancer which application for HPV-positive cervical cancer is desired in further study.
Collapse
Affiliation(s)
- Yudi Xiong
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Tianqi Li
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Ganiou Assani
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Huan Ling
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Qian Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yangyang Zeng
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Fuxiang Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yunfeng Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.
| |
Collapse
|
29
|
MYC status as a determinant of synergistic response to Olaparib and Palbociclib in ovarian cancer. EBioMedicine 2019; 43:225-237. [PMID: 30898650 PMCID: PMC6557734 DOI: 10.1016/j.ebiom.2019.03.027] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 12/18/2022] Open
Abstract
Background While PARP inhibitors and CDK4/6 inhibitors, the two classes of FDA-approved agents, have shown promising clinical benefits, there is an urgent need to develop new therapeutic strategies to improve clinical response. Meanwhile, extending the utility of these inhibitors beyond their respective molecularly defined cancer types is challenging and will likely require biomarkers predictive of treatment response especially when used in a combination drug development setting. Methods The effects of PARP inhibitor Olaparib and CDK4/6 inhibitor Palbociclib on ovarian cancer cells lines including those of high-grade serous histology were examined in vitro and in vivo. We investigated the molecular mechanism underlying the synergistic effects of drug combination. Findings We show for the first time that combining PARP and CDK4/6 inhibition has synergistic effects against MYC overexpressing ovarian cancer cells both in vitro and in vivo. Mechanistically, we find that Palbociclib induces homologous recombination (HR) deficiency through downregulation of MYC-regulated HR pathway genes, causing synthetic lethality with Olaparib. We further demonstrate that MYC expression determines sensitivity to combinatorial treatment with Olaparib and Palbociclib. Interpretation Our data provide a rationale for clinical evaluation of therapeutic synergy of these two classes of inhibitors in ovarian cancer patients whose tumors show high MYC expression and who do not respond to PARP inhibitors or CDK4/6 inhibitors monotherapies. Fund This work was supported by the National Natural Science Foundation of China [81672575, 81874111, 81472447 to HC; 81572586 and 81372853 to PL], and the Liaoning Provincial Key Basic Research Program for Universities [LZ2017002 to HC].
Collapse
|
30
|
Cui D, Zhao Y, Xu J. Activation of CXCL5-CXCR2 axis promotes proliferation and accelerates G1 to S phase transition of papillary thyroid carcinoma cells and activates JNK and p38 pathways. Cancer Biol Ther 2018; 20:608-616. [PMID: 30404567 DOI: 10.1080/15384047.2018.1539289] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
C-X-C motif chemokine ligand 5 (CXCL5) is initially identified to recruit neutrophils by interacting with its receptor, C-X-C motif chemokine receptor 2 (CXCR2). Our prior work demonstrated that the expression levels of CXCL5 and CXCR2 were higher in the papillary thyroid carcinoma (PTC) tumors than that in the non-tumors. This study was performed to further investigate how this axis regulates the growth of PTC cells. B-CPAP cells (BRAFV600E) and TPC-1 cells (RET/PTC rearrangement) expressing CXCR-2 were used as in vitro cell models. Our results showed that the recombinant human CXCL5 (rhCXCL5) promoted the proliferation of PTC cells. rhCXCL5 accelerated the G1/S transition, upregulated the expression of a group of S (DNA synthesis) or M (mitosis)-promoting cyclins and cyclin-dependent kinases (CDKs), and downregulated CDK inhibitors in PTC cells. The CDS region of homo sapiens CXCL5 gene was inserted into an eukaryotic expression vector to mediate the overexpression of CXCL5 in PTC cells. The phosphorylation of c-Jun N-terminal kinases (JNK) and p38, and the nuclear translocation of c-Jun were enhanced by CXCL5 overexpression, whereas attenuated by CXCR2 antagonist SB225002. Additionally, CXCL5/CXCR2 axis, JNK and p38 pathway inhibitors, SB225002, SP600125 and SB203580, suppressed the growth of PTC cells overexpressing CXCL5 in nude mice, respectively. Collectively, our study demonstrates a growth-promoting effect of CXCL5-CXCR2 axis in PTC cells in vitro and in vivo.
Collapse
Affiliation(s)
- Dong Cui
- a Department of Thyroid Surgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , People's Republic of China
| | - Yongfu Zhao
- a Department of Thyroid Surgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , People's Republic of China
| | - Jingchao Xu
- a Department of Thyroid Surgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , People's Republic of China
| |
Collapse
|
31
|
Gahete MD, Jimenez-Vacas JM, Alors-Perez E, Herrero-Aguayo V, Fuentes-Fayos AC, Pedraza-Arevalo S, Castaño JP, Luque RM. Mouse models in endocrine tumors. J Endocrinol 2018; 240:JOE-18-0571.R1. [PMID: 30475226 DOI: 10.1530/joe-18-0571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022]
Abstract
Endocrine and neuroendocrine tumors comprise a highly heterogeneous group of neoplasms that can arise from (neuro)endocrine cells, either from endocrine glands or from the widespread diffuse neuroendocrine system, and, consequently, are widely distributed throughout the body. Due to their diversity, heterogeneity and limited incidence, studying in detail the molecular and genetic alterations that underlie their development and progression is still a highly elusive task. This, in turn, hinders the discovery of novel therapeutic options for these tumors. To circumvent these limitations, numerous mouse models of endocrine and neuroendocrine tumors have been developed, characterized and used in pre-clinical, co-clinical (implemented in mouse models and patients simultaneously) and post-clinical studies, for they represent powerful and necessary tools in basic and translational tumor biology research. Indeed, different in vivo mouse models, including cell line-based xenografts (CDXs), patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMs), have been used to delineate the development, progression and behavior of human tumors. Results gained with these in vivo models have facilitated the clinical application in patients of diverse breakthrough discoveries made in this field. Herein, we review the generation, characterization and translatability of the most prominent mouse models of endocrine and neuroendocrine tumors reported to date, as well as the most relevant clinical implications obtained for each endocrine and neuroendocrine tumor type.
Collapse
Affiliation(s)
- Manuel D Gahete
- M Gahete, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, 14011, Spain
| | - Juan M Jimenez-Vacas
- J Jimenez-Vacas, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Emilia Alors-Perez
- E Alors-Perez, Department of Cell Biology, Physiology and Inmunology, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC) / University of Cordoba, Cordoba, Spain
| | - Vicente Herrero-Aguayo
- V Herrero-Aguayo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- A Fuentes-Fayos, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Sergio Pedraza-Arevalo
- S Pedraza-Arevalo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Justo P Castaño
- J Castaño, Dpt. of Cell Biology-University of Córdoba, IMIBIC-Maimonides Biomedical Research Institute of Cordoba, Cordoba, E-14004, Spain
| | - Raul M Luque
- R Luque, Dept of Cell Biology, Phisiology and Inmunology, Section of Cell Biology, University of Cordoba, Cordoba, Spain, Cordoba, 14014, Spain
| |
Collapse
|
32
|
Baek HB, Lombard AP, Libertini SJ, Fernandez-Rubio A, Vinall R, Gandour-Edwards R, Nakagawa R, Vidallo K, Nishida K, Siddiqui S, Wettersten H, Landesman Y, Weiss RH, Ghosh PM, Mudryj M. XPO1 inhibition by selinexor induces potent cytotoxicity against high grade bladder malignancies. Oncotarget 2018; 9:34567-34581. [PMID: 30349650 PMCID: PMC6195388 DOI: 10.18632/oncotarget.26179] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/15/2018] [Indexed: 12/28/2022] Open
Abstract
Treatment options for high grade urothelial cancers are limited and have remained largely unchanged for several decades. Selinexor (KPT-330), a first in class small molecule that inhibits the nuclear export protein XPO1, has shown efficacy as a single agent treatment for numerous different malignancies, but its efficacy in limiting bladder malignancies has not been tested. In this study we assessed selinexor-dependent cytotoxicity in several bladder tumor cells and report that selinexor effectively reduced XPO1 expression and limited cell viability in a dose dependent manner. The decrease in cell viability was due to an induction of apoptosis and cell cycle arrest. These results were recapitulated in in vivo studies where selinexor decreased tumor growth. Tumors treated with selinexor expressed lower levels of XPO1, cyclin A, cyclin B, and CDK2 and increased levels of RB and CDK inhibitor p27, a result that is consistent with growth arrest. Cells expressing wildtype RB, a potent tumor suppressor that promotes growth arrest and apoptosis, were most susceptible to selinexor. Cell fractionation and immunofluorescence studies showed that selinexor treatment increased nuclear RB levels and mechanistic studies revealed that RB ablation curtailed the response to the drug. Conversely, limiting CDK4/6 dependent RB phosphorylation by palbociclib was additive with selinexor in reducing bladder tumor cell viability, confirming that RB activity has a role in the response to XPO1 inhibition. These results provide a rationale for XPO1 inhibition as a novel strategy for the treatment of bladder malignancies.
Collapse
Affiliation(s)
- Han Bit Baek
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA.,Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Alan P Lombard
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA.,Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA.,Biochemistry, Molecular, Cellular, and Developmental Biology Graduate Group and Biotechnology Program, University of California Davis, Davis, CA, USA
| | - Stephen J Libertini
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA.,Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Aleida Fernandez-Rubio
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Ruth Vinall
- California Northstate College of Pharmacy, Elk Grove, CA, USA
| | - Regina Gandour-Edwards
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, USA
| | - Rachel Nakagawa
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Kathleen Vidallo
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Kristine Nishida
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Salma Siddiqui
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA
| | - Hiromi Wettersten
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | | | - Robert H Weiss
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA.,Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Paramita M Ghosh
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA.,Department of Urology, University of California Davis, Sacramento, CA, USA
| | - Maria Mudryj
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA.,Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| |
Collapse
|
33
|
Aberrant miRNAs Regulate the Biological Hallmarks of Glioblastoma. Neuromolecular Med 2018; 20:452-474. [PMID: 30182330 DOI: 10.1007/s12017-018-8507-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 08/17/2018] [Indexed: 12/14/2022]
Abstract
GBM is the highest incidence in primary intracranial malignancy, and it remains poor prognosis even though the patient is gave standard treatment. Despite decades of intense research, the complex biology of GBM remains elusive. In view of eight hallmarks of cancer which were proposed in 2011, studies related to the eight biological capabilities in GBM have made great progress. From these studies, it can be inferred that miRs, as a mode of post-transcriptional regulation, are involved in regulating these malignant biological hallmarks of GBM. Herein, we discuss state-of-the-art research on how aberrant miRs modulate the eight hallmarks of GBM. The upregulation of 'oncomiRs' or the genetic loss of tumor suppressor miRs is associated with these eight biological capabilities acquired during GBM formation. Furthermore, we also discuss the applicable clinical potential of these research results. MiRs may aid in the diagnosis and prognosis of GBM. Moreover, miRs are also therapeutic targets of GBM. These studies will develop and improve precision medicine for GBM in the future.
Collapse
|
34
|
Ribociclib shows potential for pharmacokinetic drug-drug interactions being a substrate of ABCB1 and potent inhibitor of ABCB1, ABCG2 and CYP450 isoforms in vitro. Biochem Pharmacol 2018; 154:10-17. [DOI: 10.1016/j.bcp.2018.04.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 04/13/2018] [Indexed: 11/20/2022]
|