TARGETING WEE1 in STANDARD THERAPY RESISTANT ESTROGEN RECEPTOR POSITIVE BREAST CANCER a Dissertation Submitted to the Faculty Of

Total Page:16

File Type:pdf, Size:1020Kb

TARGETING WEE1 in STANDARD THERAPY RESISTANT ESTROGEN RECEPTOR POSITIVE BREAST CANCER a Dissertation Submitted to the Faculty Of TARGETING WEE1 IN STANDARD THERAPY RESISTANT ESTROGEN RECEPTOR POSITIVE BREAST CANCER A Dissertation submitted to the Faculty of the Graduate School of Arts and Sciences of Georgetown University in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Tumor Biology By Yassi Fallah, M.S. Washington, DC March 10, 2021 Copyright 2021 by Yassi Fallah, M.S. All Rights Reserved ii TARGETING WEE1 IN STANDARD THERAPY RESISTANT ESTROGEN RECEPTOR POSITIVE BREAST CANCER Yassi Fallah, M.S. Thesis Advisor: Ayesha N. Shajahan-Haq, Ph.D. ABSTRACT Despite the success of antiestrogens in extending overall survival of patients with estrogen receptor positive (ER+) breast tumors, resistance to these therapies is prevalent. ER+ tumors that progress on antiestrogens are treated with antiestrogens and CDK4/6 inhibitors. However, 20% of these tumors never respond to CDK4/6 inhibitors (intrinsic resistance). Here, we used endocrine sensitive ER+ MCF7 and T47D breast cancer cells to generate long-term estrogen deprived (LTED) endocrine resistant cells that are intrinsically resistant to CDK4/6 inhibitors. Since treatment with antiestrogens arrests cells in the G1 phase of the cell cycle, we hypothesized that a defective G1 checkpoint allows resistant cells to escape this arrest but increases their dependency on G2 checkpoint for DNA repair and growth, and hence, targeting the G2 checkpoint will induce cell death. Indeed, inhibition of WEE1, a crucial G2 checkpoint regulator, with AZD1775 (Adavosertib), significantly decreased cell proliferation and increased G2/M arrest, apoptosis and gamma-H2AX levels (a marker for DNA double stranded breaks) in resistant cells compared with sensitive cells. Thus, targeting WEE1 is a promising anti-cancer therapeutic strategy in endocrine resistant breast cancer. iii DEDICATION I dedicate this work to my parents. To my mother, Faranak and father, Hamid, I thank you for everything that you do for me. Thank you for your unconditional love and support. This thesis is for all the sacrifices that you have made for me. I am grateful to have you as parents. To my brother, Kayvan thank you for being the most caring human I know. You always believe in me and I thank you. May one-day cancer deaths become a tragic memory. iv ACKNOWLEDGEMENTS I would like to first express my sincere gratitude to my mentor, Dr. Ayesha N. Shajahan-Haq for her guidance throughout my studies. Your encouragement and thoughtful criticisms motivated me to continue and do better as a scientist. I am grateful for your continuous support and patience. A special thank you to my great friend, Diane Demas. I had the pleasure of being trained by you and working with you throughout these past 4 years. I will deeply miss our science and non- science discussions. Without you, this journey would not have been the same. I would like to thank my incredible thesis committee members: Dr. Marc Lippman, Dr. Anton Wellstein, Dr. Filipa Lynce and Dr. Amrita Cheema for all the clinical and preclinical knowledge and supervision. A special thank you to my co-mentor and thesis committee chair, Dr. Lippman for all of our meetings in critiquing my presentations and for your helpful guidance. Also, a warm thank you to Dr. Wellstein for your advice and support. I learned the most about cancer biology from your teachings. I am truly honored to have you all throughout this journey. Thank you to all the members of the Tumor Biology Training Program, especially Dr. Anna Riegel, Dr. Rebecca Riggins and our director, Dr. Michael Johnson for your crucial roles and leaderships. I also would like to extend my gratitude to the faculty and staff of Georgetown University Lombardi Comprehensive Cancer Center. In particular, I thank Dr. Karen Creswell, the director of the Flow Cytometry and Cell Sorting Shared Resources and Dan Xun for your expertise and assistance. v TABLE OF CONTENTS CHAPTER I: INTRODUCTION.....................................................................................................1 Hormone Receptor Positive (HR+) Breast Cancer………………………………………. 1 Development of Cyclin-Dependent Kinases 4 and 6 (CDK4/6) Inhibitors.........................5 Current Treatment of Advanced HR+ Breast Cancer…………………………………....11 CHAPTER II: DRUG RESISTANCE IN BREAST CANCER…………………………………15 Endocrine Therapy Resistance...........................................................................................15 Resistant Mechanisms of CDK4/6 Inhibitors in Breast Cancer………………………….17 CHAPTER III: CDC25A AND WEE1 AS POTENTIAL TARGETS IN BREAST CANCER...23 Regulation of the Cell Cycle by Cell Division Cycle 25A (CDC25A) and WEE1 in Breast Cancer …………………………………………………………………………………...23 Preclinical Use of WEE1 Inhibitor in Breast Cancer…………………………………….29 Research Goals…………………………………………………………………………...35 CHAPTER IV: TARGETING WEE1 INHIBITS GROWTH OF BREAST CANCER CELLS THAT ARE RESISTANT TO ENDOCRINE THERAPY AND CDK4/6 INHIBITOR……………………. ……………………………………………………………….37 Introduction………………………………………………………………………………37 Materials and Methods.......................................................................................................38 Results……………………………………………………………………………………42 Discussion………………………………………………………………………………..66 Conclusion……………………………………………………………………………….69 Supplementary Figures…………………………………………………………………..70 vi CHAPTER V: CDC25A AS A TARGET IN ENDOCRINE THERAPY AND CDK4/6 INHIBITOR RESISTANT BREAST CANCER CELLS………………………………………..76 CHAPTER VI: CLOSING REMARKS AND FUTURE DIRECTIONS………………….........82 Closing Remarks…………………………………………………………………………82 Future Directions………………………………………………………………………...88 APPENDIX: TABLES...…………………………………………………………………….…..91 REFERENCES…………………………………………………………………………………..92 vii LIST OF FIGURES Figure 1. Intrinsic subtypes of breast cancer ...................................................................................2 Figure 2. Timeline of approval of antiestrogen agents by the FDA ................................................4 Figure 3. Chemical structures of CDK4/6 inhibitors .....................................................................11 Figure 4. Four main clinical trials in HR+ metastatic patients who have progressed on endocrine therapy ............................................................................................................................................13 Figure 5. Mechanisms of resistance to CDK4/6 inhibitors ............................................................19 Figure 6. Activity of CDC25A throughout the cell cycle ..............................................................24 Figure 7. Checkpoint control by WEE1 upon DNA damage .........................................................29 Figure 8. AZD1775 (WEE1 inhibitor) clinical trials .....................................................................33 Figure 9. LTED cells are resistant to antiestrogens .......................................................................45 Figure 10. LTED cells are resistant to CDK4/6 inhibitors ............................................................46 Figure 11. LTED cells are resistant to the combination of antiestrogens and CDK4/6 inhibitors ........................................................................................................................................48 Figure 12. Differential levels of CyclinD1 and pRB(780) proteins in paternal and LTED breast cancer cells treated with combination of E2 deprivation and ribociclib ........................................51 Figure 13. Increased G1 cell cycle arrest in LTED derivative cells ..............................................52 Figure 14. AZD1775 monotherapy suppressed growth of LTED cells .........................................55 Figure 15. Inhibition of WEE1 with AZD1775 increased G2/M cell cycle arrest in LTED cells…. ...........................................................................................................................................57 Figure 16. AZD1775 induced marked increase in apoptosis in LTED cells .................................60 Figure 17. Knockdown of p53 in combination with AZD1775 enhances inhibition of growth in MCF7 and T47D cells ....................................................................................................................63 viii Figure 18. Increased WEE1 gene expression correlate with survival in LTED cells ....................65 Figure 19. Basal growth rate of LTED cells ..................................................................................70 Figure 20. LTED cells are resistant to the combination of antiestrogens and CDK4/6 inhibitors. .......................................................................................................................................71 Figure 21. AZD1775 monotherapy suppressed growth of LTED cells .........................................72 Figure 22. Increased inhibition of cell proliferation in LTED cells following siRNA mediated WEE1 knockdown .........................................................................................................................73 Figure 23. Treatment with AZD1775 induced PARP cleavage .....................................................75 Figure 24. Inhibition of CDC25 suppressed growth of LTED cells ..............................................78 Figure 25. Anti-proliferative effect of BN82002 in combination with antiestrogen and CDK4/6 inhibitor was enhanced in LTED cells ...........................................................................................79 Figure 26. Interaction
Recommended publications
  • Downloaded from Bioscientifica.Com at 09/26/2021 02:04:00PM Via Free Access
    26 1 Endocrine-Related N Portman et al. CDK4/6 inhibitor resistance in 26:1 R15–R30 Cancer breast cancer REVIEW Overcoming CDK4/6 inhibitor resistance in ER-positive breast cancer Neil Portman1,2, Sarah Alexandrou1,2, Emma Carson1,2, Shudong Wang3, Elgene Lim1,2 and C Elizabeth Caldon1,2 1The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia 2St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, New South Wales, Australia 3Centre for Drug Discovery and Development, Cancer Research Institute, University of South Australia, Adelaide, South Australia, Australia Correspondence should be addressed to C E Caldon: [email protected] Abstract Three inhibitors of CDK4/6 kinases were recently FDA approved for use in combination Key Words with endocrine therapy, and they significantly increase the progression-free survival of f CDK4/6 inhibitors patients with advanced estrogen receptor-positive (ER+) breast cancer in the first-line f estrogen receptor treatment setting. As the new standard of care in some countries, there is the clinical f breast cancer emergence of patients with breast cancer that is both CDK4/6 inhibitor and endocrine f endocrine therapy therapy resistant. The strategies to combat these cancers with resistance to multiple treatments are not yet defined and represent the next major clinical challenge in ER+ breast cancer. In this review, we discuss how the molecular landscape of endocrine therapy resistance may affect the response to CDK4/6 inhibitors, and how this intersects with biomarkers of intrinsic insensitivity. We identify the handful of pre-clinical models of acquired resistance to CDK4/6 inhibitors and discuss whether the molecular changes in these models are likely to be relevant or modified in the context of endocrine therapy resistance.
    [Show full text]
  • The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression
    International Journal of Molecular Sciences Review The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression Tingting Zou and Zhenghong Lin * School of Life Sciences, Chongqing University, Chongqing 401331, China; [email protected] * Correspondence: [email protected] Abstract: The cell cycle is a collection of events by which cellular components such as genetic materials and cytoplasmic components are accurately divided into two daughter cells. The cell cycle transition is primarily driven by the activation of cyclin-dependent kinases (CDKs), which activities are regulated by the ubiquitin-mediated proteolysis of key regulators such as cyclins, CDK inhibitors (CKIs), other kinases and phosphatases. Thus, the ubiquitin-proteasome system (UPS) plays a pivotal role in the regulation of the cell cycle progression via recognition, interaction, and ubiquitination or deubiquitination of key proteins. The illegitimate degradation of tumor suppressor or abnormally high accumulation of oncoproteins often results in deregulation of cell proliferation, genomic instability, and cancer occurrence. In this review, we demonstrate the diversity and complexity of the regulation of UPS machinery of the cell cycle. A profound understanding of the ubiquitination machinery will provide new insights into the regulation of the cell cycle transition, cancer treatment, and the development of anti-cancer drugs. Keywords: cell cycle regulation; CDKs; cyclins; CKIs; UPS; E3 ubiquitin ligases; Deubiquitinases (DUBs) Citation: Zou, T.; Lin, Z. The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression. 1. Introduction Int. J. Mol. Sci. 2021, 22, 5754. https://doi.org/10.3390/ijms22115754 The cell cycle is a ubiquitous, complex, and highly regulated process that is involved in the sequential events during which a cell duplicates its genetic materials, grows, and di- Academic Editors: Kwang-Hyun Bae vides into two daughter cells.
    [Show full text]
  • TEAD4 Ensures Postimplantation Development by Promoting Trophoblast Self-Renewal: an Implication in Early Human Pregnancy Loss
    TEAD4 ensures postimplantation development by promoting trophoblast self-renewal: An implication in early human pregnancy loss Biswarup Sahaa,1,2, Avishek Gangulya,1, Pratik Homea,b, Bhaswati Bhattacharyaa, Soma Raya, Ananya Ghosha, M. A. Karim Rumia,b, Courtney Marshb,c, Valerie A. Frenchc, Sumedha Gunewardenad, and Soumen Paula,b,c,3 aDepartment of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160; bInstitute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160; cDepartment of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160; and dDepartment of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160 Edited by R. Michael Roberts, University of Missouri, Columbia, MO, and approved June 22, 2020 (received for review February 12, 2020) Early pregnancy loss affects ∼15% of all implantation-confirmed Studies in mutant mouse models showed that failure in pla- human conceptions. However, evolutionarily conserved molecular centation often leads to in utero embryonic death (6, 7). Therefore, mechanisms that regulate self-renewal of trophoblast progenitors impaired placentation due to defective development or function of and their association with early pregnancy loss are poorly under- trophoblast cell lineages is considered one of the major underlying stood. Here, we provide evidence that transcription factor TEAD4 causes of early pregnancy loss. Disruptions of trophoblast pro- ensures survival of postimplantation mouse and human embryos genitor differentiation and defective placentation have also been by controlling self-renewal and stemness of trophoblast progeni- implicated as probable causes of pregnancy-associated compli- tors within the placenta primordium.
    [Show full text]
  • Cyclin E2, a Novel Human G1 Cyclin and Activating Partner of CDK2 and CDK3, Is Induced by Viral Oncoproteins
    Oncogene (1998) 17, 2787 ± 2798 ã 1998 Stockton Press All rights reserved 0950 ± 9232/98 $12.00 http://www.stockton-press.co.uk/onc Cyclin E2, a novel human G1 cyclin and activating partner of CDK2 and CDK3, is induced by viral oncoproteins Maimoona Zariwala1, Jidong Liu2 and Yue Xiong*,1,2,3 1Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-3280; 2Department of Biochemistry and Biophysics; University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599- 3280; 3Program in Molecular Biology and Biotechnology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-3280, USA G1 cyclin E controls the initiation of DNA synthesis by complexes with CDK4 or CDK6 to prevent the CDKs activating CDK2, and abnormally high levels of cyclin E from binding with and becoming activated by D-type expression have frequently been observed in human cyclins. The main function of CDK inhibitors is cancers. We have isolated a novel human cyclin, cyclin believed to couple diversi®ed growth inhibitory signals E2, that contains signi®cant homology to cyclin E. to the cell cycle clock. Cyclin E2 speci®cally interacts with CDK inhibitors of The decision to enter the replicative DNA synthesis the CIP/KIP family and activates both CDK2 and (S) phase or arrest in G1 is linked to diverse cellular CDK3. The expression of cyclin E2 mRNA oscillates processes such as signal transduction, cell differentia- periodically throughout the cell cycle, peaking at the tion, senescence, and oncogenic transformation G1/S transition, and exhibits a pattern of tissue (Hunter and Pines, 1994).
    [Show full text]
  • Tumour Kinome Re-Wiring Governs Resistance to Palbociclib in Oestrogen Receptor Positive Breast Cancers, Highlighting New Therapeutic Modalities
    Oncogene (2020) 39:4781–4797 https://doi.org/10.1038/s41388-020-1284-6 ARTICLE Tumour kinome re-wiring governs resistance to palbociclib in oestrogen receptor positive breast cancers, highlighting new therapeutic modalities 1 1 1 1 1 Sunil Pancholi ● Ricardo Ribas ● Nikiana Simigdala ● Eugene Schuster ● Joanna Nikitorowicz-Buniak ● 2 3 1 4 5 6 Anna Ressa ● Qiong Gao ● Mariana Ferreira Leal ● Amandeep Bhamra ● Allan Thornhill ● Ludivine Morisset ● 6 6 2 2 7 Elodie Montaudon ● Laura Sourd ● Martin Fitzpatrick ● Maarten Altelaar ● Stephen R. Johnston ● 6 1,8 1 Elisabetta Marangoni ● Mitch Dowsett ● Lesley-Ann Martin Received: 24 May 2019 / Revised: 18 March 2020 / Accepted: 24 March 2020 / Published online: 19 April 2020 © The Author(s) 2020. This article is published with open access Abstract Combination of CDK4/6 inhibitors and endocrine therapy improves clinical outcome in advanced oestrogen receptor (ER)- positive breast cancer, however relapse is inevitable. Here, we show in model systems that other than loss of RB1 few gene- copy number (CN) alterations are associated with irreversible-resistance to endocrine therapy and subsequent secondary 1234567890();,: 1234567890();,: resistance to palbociclib. Resistance to palbociclib occurred as a result of tumour cell re-wiring leading to increased expression of EGFR, MAPK, CDK4, CDK2, CDK7, CCNE1 and CCNE2. Resistance altered the ER genome wide-binding pattern, leading to decreased expression of ‘classical’ oestrogen-regulated genes and was accompanied by reduced sensitivity to fulvestrant and tamoxifen. Persistent CDK4 blockade decreased phosphorylation of tuberous sclerosis complex 2 (TSC2) enhancing EGFR signalling, leading to the re-wiring of ER. Kinome-knockdown confirmed dependency on ERBB- signalling and G2/M–checkpoint proteins such as WEE1, together with the cell cycle master regulator, CDK7.
    [Show full text]
  • Cyclin E Provides a Link Between Cell Cycle, Dna
    CYCLIN E PROVIDES A LINK BETWEEN CELL CYCLE, DNA REPAIR AND APOPTOSIS A dissertation submitted to Kent State University in collaboration with the Lerner Research Institute, Cleveland Clinic in partial fulfillment of the requirements for the degree of Doctor of Philosophy by Dragos Costin Plesca May, 2008 Dissertation written by Dragos Costin Plesca Pharm.D., University of Medicine and Pharmacy “Carol Davila”, Romania, 2002 Ph.D., Kent State University, 2008 Approved by ____________________, Chair, Doctoral Dissertation Committee Alexandru Almasan, Ph.D ____________________, Member, Doctoral Dissertation Committee James Blank, Ph.D ____________________, Member, Doctoral Dissertation Committee Gail Fraizer, Ph.D ____________________, Member, Doctoral Dissertation Committee Olena Piontkivska, Ph.D ____________________, Graduate Faculty Representative Jennifer Marcinkiewicz, Ph.D Accepted by ____________________, Director, School of Biomedical Sciences Robert V. Dorman, Ph.D ____________________, Dean, College of Arts and Sciences John R. D. Stalvey, Ph.D ii TABLE OF CONTENTS List of Figures.................................................................................................................vi List of Tables ..................................................................................................................ix Acknowledgments ............................................................................................................x Chapter I. Introduction ................................................................................................1
    [Show full text]
  • Cyclin-Dependent Kinase 2 (Cdk2) Controls Phosphatase-Regulated Signaling and Function in Platelets
    bioRxiv preprint doi: https://doi.org/10.1101/2020.05.31.126953; this version posted June 28, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Cyclin-dependent kinase 2 (Cdk2) controls phosphatase-regulated signaling and function in platelets Paul R. Woods, Jr.,1,2 Brian L. Hood5, Sruti Shiva,$4 Thomas P. Conrads5, Sarah Suchko,2 Richard Steinman, 1,2,4# Departments oF Medicine1, Hillman Cancer Center2, Vascular Medicine Institute3, Department oF Molecular Pharmacology and Chemical Biology4, University of Pittsburgh School of Medicine; The Henry M. Jackson Foundation For the Advancement of Military Medicine, Inc., Inova Women’s Service Line, Inova Health System5 #Corresponding author: Richard Steinman, MD, PhD Associate Professor of Medicine and Pharmacology Associate Dean, Director Medical Scientist Training Program Director, Physician Scientist Training Program University of Pittsburgh School of Medicine 2.26f Hillman Cancer Center 5117 Centre Avenue Pittsburgh, PA 15213 USA phone: 412 6233237 fax: 412 6234840 [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2020.05.31.126953; this version posted June 28, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract Cell cycle regulatory molecules including cyclin-dependent kinases can be recruited into non-nuclear pathways to coordinate cell cycling with the energetic state oF the cell or with Functions such as motility.
    [Show full text]
  • Inhibition of CDK4/6 As Therapeutic Approach for Ovarian Cancer Patients: Current Evidences and Future Perspectives
    cancers Review Inhibition of CDK4/6 as Therapeutic Approach for Ovarian Cancer Patients: Current Evidences and Future Perspectives Alessandra Dall’Acqua 1,†, Michele Bartoletti 2,3,† , Nastaran Masoudi-Khoram 1,‡, Roberto Sorio 2, Fabio Puglisi 2,3 , Barbara Belletti 1 and Gustavo Baldassarre 1,* 1 Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, 33081 Aviano, Italy; [email protected] (A.D.); [email protected] (N.M.-K.); [email protected] (B.B.) 2 Medical Oncology and Cancer Prevention Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, 33081 Aviano, Italy; [email protected] (M.B.); [email protected] (R.S.); [email protected] (F.P.) 3 Department of Medicine (DAME), University of Udine, 33100 Udine, Italy * Correspondence: [email protected]; Tel.: +39-0434-659779; Fax: +39-0434-659429 † These authors equally contributed to the work. ‡ Present address: Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran. Simple Summary: Altered regulation of the cell cycle is a hallmark of cancer. The recent clinical success of the inhibitors of CDK4 and CDK6 has convincingly demonstrated that targeting cell cycle components may represent an effective anti-cancer strategy, at least in some cancer types. However, possible applications of CDK4/6 inhibitors in patients with ovarian cancer is still under evaluation. Citation: Dall’Acqua, A.; Bartoletti, M.; Masoudi-Khoram, N.; Sorio, R.; Here, we describe the possible biological role of CDK4 and CDK6 complexes in ovarian cancer and Puglisi, F.; Belletti, B.; Baldassarre, G. provide the rationale for the use of CDK4/6 inhibitors in this pathology, alone or in combination Inhibition of CDK4/6 as Therapeutic with other drugs.
    [Show full text]
  • Wee1 Kinase Inhibitor AZD1775 Effectively Sensitizes Esophageal Cancer to Radiotherapy
    Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Wee1 Kinase Inhibitor AZD1775 Effectively Sensitizes Esophageal Cancer to Radiotherapy Linlin Yang1, Changxian Shen1, Cory Pettit1, Tianyun Li1, Andrew Hu1, Eric Miller1, Junran Zhang1, Steven H. Lin2, Terence M. Williams1, * 1The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio. 2The University of Texas MD Anderson Cancer Center, Houston, Texas. *Corresponding Author: Terence M. Williams, Department of Radiation Oncology, The Ohio State University, 460 W. 12th Avenue, BRT/Room 492, Columbus, OH 43210-1280. Phone: (614) 293-3244. Fax: 614-293-4044. E-mail: [email protected] Running title: Targeting Wee1 for radiosensitization of esophageal cancer Key words: Wee1, AZD1775, G2 checkpoint, mitotic catastrophe, esophageal cancer Conflicts of Interest: The authors report no potential conflicts of interest. Financial Disclosure Statements: All authors have no competing financial interests to disclose. Funding Support: This work was supported by the following grants: The Ohio State University Comprehensive Cancer Center (OSU-CCC), National Institutes of Health (P30 CA016058 and R01 CA198128), and National Center for Advancing Translational Sciences (KL2TR001068). 1 Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. ABSTRACT Purpose: Esophageal cancer (ESCA) is a deadly malignancy with a 5-year survival rate of only 5-20%, which has remained unchanged for decades.
    [Show full text]
  • Early Adaptation and Acquired Resistance to CDK4/6 Inhibition In
    Published OnlineFirst March 28, 2016; DOI: 10.1158/0008-5472.CAN-15-0728 Cancer Therapeutics, Targets, and Chemical Biology Research Early Adaptation and Acquired Resistance to CDK4/6 Inhibition in Estrogen Receptor–Positive Breast Cancer Maria Teresa Herrera-Abreu1, Marta Palafox2, Uzma Asghar1, Martín A. Rivas3, Rosalind J. Cutts1, Isaac Garcia-Murillas1, Alex Pearson1, Marta Guzman2, Olga Rodriguez2, Judit Grueso2, Meritxell Bellet4, Javier Cortes 4, Richard Elliott1, Sunil Pancholi1, Jose Baselga5, Mitch Dowsett1,6, Lesley-Ann Martin1, Nicholas C. Turner1,7, and Violeta Serra2 Abstract Small-molecule inhibitors of the CDK4/6 cell-cycle kinases apy, CDK4/6, and PI3K inhibition was more effective than have shown clinical efficacy in estrogen receptor (ER)-positive paired combinations, provoking rapid tumor regressions in a metastatic breast cancer, although their cytostatic effects are PDX model. Mechanistic investigations showed that acquired limited by primary and acquired resistance. Here we report that resistance to CDK4/6 inhibition resulted from bypass of cyclin ER-positive breast cancer cells can adapt quickly to CDK4/6 D1–CDK4/6 dependency through selection of CCNE1 ampli- inhibition and evade cytostasis, in part, via noncanonical cyclin fication or RB1 loss. Notably, although PI3K inhibitors could D1-CDK2–mediated S-phase entry. This adaptation was pre- prevent resistance to CDK4/6 inhibitors, they failed to resen- vented by cotreatment with hormone therapies or PI3K inhi- sitize cells once resistance had been acquired. However, we bitors, which reduced the levels of cyclin D1 (CCND1)and found that cells acquiring resistance to CDK4/6 inhibitors due other G1–S cyclins, abolished pRb phosphorylation, and inhib- to CCNE1 amplification could be resensitized by targeting ited activation of S-phase transcriptional programs.
    [Show full text]
  • Differential Expression of Cell Cycle Regulators in CDK5- Dependent Medullary Thyroid Carcinoma Tumorigenesis
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 6, No. 14 Differential expression of cell cycle regulators in CDK5- dependent medullary thyroid carcinoma tumorigenesis Karine Pozo1, Antje Hillmann1, Alexander Augustyn2,3, Florian Plattner1, Tao Hai4, Tanvir Singh1, Saleh Ramezani5, Xiankai Sun5, Roswitha Pfragner6, John D. Minna2,3,7, Gilbert J. Cote4, Herbert Chen8, James A. Bibb1,5,9,* and Fiemu E. Nwariaku10,* 1 Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA 2 Hamon Center for Therapeutic Oncology Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA 3 Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA 4 Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 5 Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA 6 Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria 7 Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, USA 8 Endocrine Surgery Research Laboratory, The University of Wisconsin Carbone Cancer Center, Madison, WI, USA 9 Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX, USA 10 Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX, USA * These authors have contributed equally to this work Correspondence to: James A. Bibb, email: [email protected] Correspondence to :Karine Pozo, email: [email protected] Keywords: Cdk5, retinoblastoma protein, neuroendocrine, medullary thyroid carcinoma, cell cycle Received: August 26, 2014 Accepted: March 03, 2015 Published: April 14, 2015 This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
    [Show full text]
  • CDK11 Loss Induces Cell Cycle Dysfunction and Death of BRAF and NRAS Melanoma Cells
    pharmaceuticals Article CDK11 Loss Induces Cell Cycle Dysfunction and Death of BRAF and NRAS Melanoma Cells Rehana L. Ahmed 1,2, Daniel P. Shaughnessy 3, Todd P. Knutson 4,5 , Rachel I. Vogel 2,6 , Khalil Ahmed 2,3,4,7, Betsy T. Kren 2,3 and Janeen H. Trembley 2,3,4,* 1 Department of Dermatology, University of Minnesota, Minneapolis, MN 55455, USA; [email protected] 2 Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; [email protected] (R.I.V.); [email protected] (K.A.); [email protected] (B.T.K.) 3 Research Service, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA; [email protected] 4 Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; [email protected] 5 Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA 6 Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA 7 Department of Urology, University of Minnesota, Minneapolis, MN 55455, USA * Correspondence: [email protected] Received: 15 February 2019; Accepted: 28 March 2019; Published: 2 April 2019 Abstract: Cyclin dependent kinase 11 (CDK11) is a protein kinase that regulates RNA transcription, pre-mRNA splicing, mitosis, and cell death. Targeting of CDK11 expression levels is effective in the experimental treatment of breast and other cancers, but these data are lacking in melanoma. To understand CDK11 function in melanoma, we evaluated protein and RNA levels of CDK11, Cyclin L1 and Cyclin L2 in benign melanocytes and BRAF- as well as NRAS-mutant melanoma cell lines. We investigated the effectiveness of reducing expression of this survival kinase using RNA interference on viability, clonal survival, and tumorsphere formation in melanoma cell lines.
    [Show full text]