Molecular Functions of the Rad9a-Rad1-Hus1 Dna Damage

Total Page:16

File Type:pdf, Size:1020Kb

Molecular Functions of the Rad9a-Rad1-Hus1 Dna Damage MOLECULAR FUNCTIONS OF THE RAD9A-RAD1-HUS1 DNA DAMAGE CHECKPOINT CLAMP IN GENOME MAINTENANCE AND TUMORIGENESIS A Dissertation Presented to the Faculty of the Graduate School of Cornell University In Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy by Darshil R Patel August 2018 © 2018 Darshil R Patel MOLECULAR FUNCTIONS OF THE RAD9A-RAD1-HUS1 DNA DAMAGE CHECKPOINT CLAMP IN GENOME MAINTENANCE AND TUMORIGENESIS Darshil R Patel, Ph.D. Cornell University 2018 DNA damage checkpoint pathways are part of the broader cellular DNA damage response (DDR) that promotes genome maintenance when cells experience DNA damage. The RAD9A-HUS1-RAD1 (9-1-1) complex is a DDR component that functions as a heterotrimeric DNA binding clamp that promotes checkpoint signaling and DNA repair. Loss of 9-1-1 function has severe consequences, including embryonic lethality, genomic instability, subfertility and hypersensitivity to replication stress. To fully understand the role of the 9-1-1 complex in DNA repair, we first utilized a targeted mutation approach to identify functionally important residues of HUS1 that drive clamp formation, DNA interactions, and downstream effector functions. These studies revealed that both checkpoint signaling and DNA repair by the 9-1-1 complex separably promote genome maintenance. Next, a proteome-wide screen identified novel HUS1interactors and uncovered a new role for the 9-1-1 complex in regulating protein neddylation. As Hus1 loss results in radial chromosome formation and hypersensitivity to inter-strand crosslinking (ICL) agents, phenotypes also seen in the genome instability syndrome Fanconi Anemia (FA), we investigated the relationship between the 9-1-1 ii complex and the FA DNA repair pathway. Our studies suggested that the 9-1-1 complex was essential for recruitment of several FA proteins during ICL repair. Furthermore, the 9-1-1 complex also protected stalled replication forks against MRE11-dependent fork degradation. Overall, these data suggest a role for the 9-1-1 complex in coordinating multiple signaling and repair proteins required for accurate ICL repair. DDR proteins are known to protect against cancer-initiating mutations in normal cells; however they also support tumor growth at later stages, allowing cancers to tolerate elevated stress levels associated with malignant transformation. We explored the role of HUS1 in cancer, and discovered that Hus1 deficiency decreased cell transformation in cultured cells and decreased tumorigenesis in mouse models of lung and skin cancer. Expression analysis in multiple cancer cell lines revealed that HUS1 levels in cancer can increase through a mechanism involving alternative polyadenylation. In sum, this dissertation decodes distinct functions for the 9-1-1 complex in DNA repair and tumorigenesis, and identifies novel interactions between HUS1 and effectors important for genomic integrity and cell survival. iii BIOGRAPHICAL SKETCH Darshil Patel was born on March 26, 1990 in Vadodara, India. In 2008, he received his high school diploma from Corona High School in California. Thereafter, he enrolled into Riverside community college to continue his education. During his time at the community college, he was awarded Beckman Coulter summer internship in 2009, which allowed him to work as a research intern at Ambryx Biotechnology Incorporation, a cancer research-based company. He continued working at Ambryx Biotechnology until 2011. In 2010, after completing two years of general education at the community college, he transferred to the University of California Los Angeles (UCLA) in California. At UCLA, he majored in Molecular, Cell and Developmental Biology (MCDB) and minored in Anthropology. During his first year, he worked in Dr. Elaine Tobin’s Lab to understand the regulatory functions of the circadian clock proteins, CCA1 and LYH in Arabidopsis Thaliana. Thereafter, he joined Dr. Samuel French Lab in the department of Pathology at UCLA, where his research was focused at developing a proteomic based program to study the development of liver cancer induced by hepatitis C viral infection. He also actively volunteered at UCLA Santa Clarita Cancer Center in Valencia, California as a student intern. During his tenure at both labs, he learnt essential skills in basic molecular and biochemistry techniques, as well as exposed him to basic and translational research. After completing his B.S. degree in 2012, Darshil decided to join the Biochemistry, Molecular and Cell Biology (BMCB) program at Cornell University for his doctorate. He completed his dissertation work with Dr. Robert Weiss, where he focused on understanding the distinct roles for the DNA damage checkpoint clamp subunit HUS1 in genome maintenance. iv I would like to dedicate this thesis to my lovely and ever-supporting parents, Renu & Rohit v ACKNOWLEDGMENTS It would not have been possible to complete my doctoral dissertation work without the support of several individuals, who stood by me throughout this time, supporting and encouraging my work. I would like to start by thanking my advisor, Dr. Robert Weiss for his unqualified support and assistance during my stay in his laboratory. As a student, I could not have asked for a better mentor. He provided me with all the guidance as well as the freedom to successfully complete my work. From him, I have learnt several important technical and intellectual skills needed to be an independent researcher. I feel my great privilege and honor to have worked under him, as he has prepared me for the next phase of my scientific life. Along with Dr. Weiss, I would like to thank the members of the Weiss lab for their support and guidance. Dr. Pei Xin Lim provided great support and guidance during my early days in the lab. I was able to work with him and learn a great deal about the scientific tenacity required in research. I would also like to thank former members of the lab, Dr. Amy Lyndaker, Dr. Tim Pierpont, Dr. Yashira Abril Negron, and Dr. April Blong for providing a collaborative and healthy lab environment. I also had the privilege to work with excellent lab members and co-authors, Dr. Joanna Mleczko, and Catalina Pereira, who made substantial contribution to my dissertation work. Finally, I would like to thank the other current members of the Weiss lab, Dr. Elizabeth Moore, and Irma Fernandez for their continued support, guidance and their friendship; it was a great pleasure working with you all. I would also like to thank undergraduate mentees that I work with Ellen Hong, Charlton Tsai, Brenna Remick, and Cindy Luan for their contribution to my work. My committee members, Dr. John Schimenti and Dr. Marcus Smolka were vi also instrumental for my success as they continuously provided great scientific input and suggestions for shaping my work. I would like to thank my collaborators for their help with the project, Dr. Marcus Smolka, Dr. John Schimenti, Dr. Andrew White, Dr. Jeff Pleiss, members of the Smolka Lab, and several members of the R3 group. I also appreciate the support and guidance that I received from my class members and my funding sources. Finally, I would like to thank my friends and family, especially my wife, Bhumika, for showering me with selfless love, encouragement and support throughout my journey. vii TABLE OF CONTENTS CHAPTER 1: Literature review 1 1.1. DNA damage 1 1.2. DNA damage response (DDR) 4 1.3. Checkpoint signaling: DDR Kinases 7 1.3.1. ATM: Master regulator of DSBs 8 1.3.2. ATR: Mediator of DNA replication stress 11 1.4. DNA Repair 14 1.4.1. Direct reversal of DNA damage 15 1.4.2. Mismatch repair (MMR) 15 1.4.3. Base excision repair (BER) 16 1.4.4. Nucleotide excision repair (NER) 20 1.4.5. Translesion synthesis (TLS) 21 1.4.6. DSB repair – Non homologous end joining (NHEJ) and 22 homologous recombination (HR) 1.4.6.1.Non-homologous end-joining 22 1.4.6.2.Homologous recombination 23 1.4.6.3.Relationship between BRCA1 and 53BP1 in repair 25 choice 1.4.6.4.Other DSB repair pathways 27 1.4.7. Fanconi anemia pathway mediated DNA ICL repair 27 1.5. The RAD9A-HUS1-RAD1 (9-1-1) complex 29 1.5.1. Identification of the 9-1-1 complex 30 1.5.2. Structure and loading of the 9-1-1 complex 30 1.5.3. Role for the 9-1-1 complex in checkpoint signaling and DNA 32 repair 1.5.4. Physiological relevance of the 9-1-1 complex 35 1.6. Crosstalk between replication fork stability and DNA repair machinery 39 for genome maintenance 1.7. Role of DDR proteins in tumorigenesis and anti-cancer therapeutics 49 1.8. Summary 52 CHAPTER 2: Genome protection by the 9-1-1 complex subunit HUS1 requires 55 clamp formation, DNA contacts, and ATR signaling-independent effector functions 2.1 Abstract 55 2.2 Introduction 56 2.3 Materials and methods 58 2.4 Results 72 2.5 Discussion 93 viii 2.6. Acknowledgement 97 CHAPTER 3: The 9-1-1 DNA damage response complex acts upstream of 98 BRCA1, FANCD2, and RAD51 during DNA interstrand crosslink repair to maintain genomic stability 98 3.1 Abstract 99 3.2 Introduction 102 3.3 Materials and methods 111 3.4 Results 155 3.5 Discussion 162 3.6. Acknowledgement CHAPTER 4: A novel role for the 9-1-1 complex in regulating neddylation for 164 genome maintenance 4.1 Abstract 164 4.2 Introduction 165 4.3 Materials and methods 168 4.4 Results 175 4.5 Discussion 188 4.6. Acknowledgement 195 CHAPTER 5: Tumor promoting functions for the DNA damage response 196 protein HUS1 5.1 Abstract 196 5.2 Introduction 197 5.3 Materials and methods 199 5.4 Results 206 5.5 Discussion 240 5.6.
Recommended publications
  • A Family of Mammalian E3 Ubiquitin Ligases That Contain the UBR Box Motif and Recognize N-Degrons Takafumi Tasaki,1 Lubbertus C
    MOLECULAR AND CELLULAR BIOLOGY, Aug. 2005, p. 7120–7136 Vol. 25, No. 16 0270-7306/05/$08.00ϩ0 doi:10.1128/MCB.25.16.7120–7136.2005 Copyright © 2005, American Society for Microbiology. All Rights Reserved. A Family of Mammalian E3 Ubiquitin Ligases That Contain the UBR Box Motif and Recognize N-Degrons Takafumi Tasaki,1 Lubbertus C. F. Mulder,2 Akihiro Iwamatsu,3 Min Jae Lee,1 Ilia V. Davydov,4† Alexander Varshavsky,4 Mark Muesing,2 and Yong Tae Kwon1* Center for Pharmacogenetics and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 152611; Aaron Diamond AIDS Research Center, The Rockefeller University, New York, New York 100162; Protein Research Network, Inc., Yokohama, Kanagawa 236-0004, Japan3; and Division of Biology, California Institute of Technology, Pasadena, California 911254 Received 15 March 2005/Returned for modification 27 April 2005/Accepted 13 May 2005 A subset of proteins targeted by the N-end rule pathway bear degradation signals called N-degrons, whose determinants include destabilizing N-terminal residues. Our previous work identified mouse UBR1 and UBR2 as E3 ubiquitin ligases that recognize N-degrons. Such E3s are called N-recognins. We report here that while double-mutant UBR1؊/؊ UBR2؊/؊ mice die as early embryos, the rescued UBR1؊/؊ UBR2؊/؊ fibroblasts still retain the N-end rule pathway, albeit of lower activity than that of wild-type fibroblasts. An affinity assay for proteins that bind to destabilizing N-terminal residues has identified, in addition to UBR1 and UBR2, a huge (570 kDa) mouse protein, termed UBR4, and also the 300-kDa UBR5, a previously characterized mammalian E3 known as EDD/hHYD.
    [Show full text]
  • Blockage of Neddylation Modification Stimulates Tumor Sphere Formation
    Blockage of neddylation modification stimulates tumor PNAS PLUS sphere formation in vitro and stem cell differentiation and wound healing in vivo Xiaochen Zhoua,b,1, Mingjia Tanb,1, Mukesh K. Nyatib, Yongchao Zhaoc,d, Gongxian Wanga,2, and Yi Sunb,c,e,2 aDepartment of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; bDivision of Radiation and Cancer Biology, Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109; cInstitute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; dKey Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; and eCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310058, China Edited by Vishva M. Dixit, Genentech, San Francisco, CA, and approved March 10, 2016 (received for review November 13, 2015) MLN4924, also known as pevonedistat, is the first-in-class inhibitor acting alone or in combination with current chemotherapy of NEDD8-activating enzyme, which blocks the entire neddylation and/or radiation (6, 11). One of the seven clinical trials of MLN4924 modification of proteins. Previous preclinical studies and current (NCT00911066) was published recently, concluding a modest effect clinical trials have been exclusively focused on its anticancer property. of MLN4924 against acute myeloid leukemia (AML) (12). Unexpectedly, we show here, to our knowledge for the first time, To further elucidate the role of blocking neddylation in cancer that MLN4924, when applied at nanomolar concentrations, signif- treatment, we thought to study the effect of MLN4924 on cancer icantly stimulates in vitro tumor sphere formation and in vivo stem cells (CSCs) or tumor-initiating cells (TICs), a small group tumorigenesis and differentiation of human cancer cells and mouse of tumor cells with stem cell properties that have been claimed to embryonic stem cells.
    [Show full text]
  • The Anti-Tumor Activity of the NEDD8 Inhibitor Pevonedistat in Neuroblastoma
    International Journal of Molecular Sciences Article The Anti-Tumor Activity of the NEDD8 Inhibitor Pevonedistat in Neuroblastoma Jennifer H. Foster 1,* , Eveline Barbieri 1, Linna Zhang 1, Kathleen A. Scorsone 1, Myrthala Moreno-Smith 1, Peter Zage 2,3 and Terzah M. Horton 1,* 1 Texas Children’s Cancer and Hematology Centers, Department of Pediatrics, Section of Hematology and Oncology, Baylor College of Medicine, Houston, TX 77030, USA; [email protected] (E.B.); [email protected] (L.Z.); [email protected] (K.A.S.); [email protected] (M.M.-S.) 2 Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, CA 92024, USA; [email protected] 3 Peckham Center for Cancer and Blood Disorders, Rady Children’s Hospital, San Diego, CA 92123, USA * Correspondence: [email protected] (J.H.F.); [email protected] (T.M.H.); Tel.: +1-832-824-4646 (J.H.F.); +1-832-824-4269 (T.M.H.) Abstract: Pevonedistat is a neddylation inhibitor that blocks proteasomal degradation of cullin–RING ligase (CRL) proteins involved in the degradation of short-lived regulatory proteins, including those involved with cell-cycle regulation. We determined the sensitivity and mechanism of action of pevonedistat cytotoxicity in neuroblastoma. Pevonedistat cytotoxicity was assessed using cell viability assays and apoptosis. We examined mechanisms of action using flow cytometry, bromod- eoxyuridine (BrDU) and immunoblots. Orthotopic mouse xenografts of human neuroblastoma were generated to assess in vivo anti-tumor activity. Neuroblastoma cell lines were very sensitive to pevonedistat (IC50 136–400 nM). The mechanism of pevonedistat cytotoxicity depended on p53 Citation: Foster, J.H.; Barbieri, E.; status.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Hypermethylation of RAD9A Intron 2 in Childhood Cancer Patients, Leukemia and Tumor Cell Lines Suggest a Role for Oncogenic Transformation
    Hypermethylation of RAD9A intron 2 in childhood cancer patients, leukemia and tumor cell lines suggest a role for oncogenic transformation Danuta Galetzka ( [email protected] ) Johannes Gutenberg Universitat Mainz https://orcid.org/0000-0003-1825-9136 Julia Boeck Institute of Human Genetics, Julius Maximilians University Würzburg Marcus Dittrich Bioinformatics Department Julius-Maximilians-Universitat Wurzburg Olesja Sinizyn Department of Radiation Oncology and Radiations Therapy , University Medical Centre, Mainz Marco Ludwig DRK Medical Centre Alzey Heidi Rossmann Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre, Mainz Claudia Spix German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Centre, Mainz Markus Radsak Departement of Hematology, University Medical Centre, Mainz Peter Scholz-Kreisel Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Centre, Mainz Johanna Mirsch Radiation Biology and DNA Repair, Technical University, Darmstadt Matthias Linke Institute of Human Genetics, University Medical Centre, Mainz Walburgis Brenner Departement of Obsterics and Womans Health, University Medical Centre, Mainz Manuela Marron Leibniz Institute for Prevention Research and Epidemiology, BIPS, Bremen Alicia Poplawski Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Centre, Mainz Dirk Prawitt Center for Pediatrics and Adolescent Medicine, University Medical Centre, Mainz Thomas Haaf Institute of Human Genetics, Julius Maximilians University, Würzburg Heinz Schmidberger Department of Radiation Oncology and Radiation Therapy, University Centre, Mainz Research article Keywords: RAD9A, childhood cancer, hypermethylation, normal body cells, somatic mosaicism, leukemia Posted Date: August 12th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-55470/v1 Page 1/22 License: This work is licensed under a Creative Commons Attribution 4.0 International License.
    [Show full text]
  • Neddylation: a Novel Modulator of the Tumor Microenvironment Lisha Zhou1,2*†, Yanyu Jiang3†, Qin Luo1, Lihui Li1 and Lijun Jia1*
    Zhou et al. Molecular Cancer (2019) 18:77 https://doi.org/10.1186/s12943-019-0979-1 REVIEW Open Access Neddylation: a novel modulator of the tumor microenvironment Lisha Zhou1,2*†, Yanyu Jiang3†, Qin Luo1, Lihui Li1 and Lijun Jia1* Abstract Neddylation, a post-translational modification that adds an ubiquitin-like protein NEDD8 to substrate proteins, modulates many important biological processes, including tumorigenesis. The process of protein neddylation is overactivated in multiple human cancers, providing a sound rationale for its targeting as an attractive anticancer therapeutic strategy, as evidence by the development of NEDD8-activating enzyme (NAE) inhibitor MLN4924 (also known as pevonedistat). Neddylation inhibition by MLN4924 exerts significantly anticancer effects mainly by triggering cell apoptosis, senescence and autophagy. Recently, intensive evidences reveal that inhibition of neddylation pathway, in addition to acting on tumor cells, also influences the functions of multiple important components of the tumor microenvironment (TME), including immune cells, cancer-associated fibroblasts (CAFs), cancer-associated endothelial cells (CAEs) and some factors, all of which are crucial for tumorigenesis. Here, we briefly summarize the latest progresses in this field to clarify the roles of neddylation in the TME, thus highlighting the overall anticancer efficacy of neddylaton inhibition. Keywords: Neddylation, Tumor microenvironment, Tumor-derived factors, Cancer-associated fibroblasts, Cancer- associated endothelial cells, Immune cells Introduction Overall, binding of NEDD8 molecules to target proteins Neddylation is a reversible covalent conjugation of an can affect their stability, subcellular localization, conform- ubiquitin-like molecule NEDD8 (neuronal precursor ation and function [4]. The best-characterized substrates cell-expressed developmentally down-regulated protein of neddylation are the cullin subunits of Cullin-RING li- 8) to a lysine residue of the substrate protein [1, 2].
    [Show full text]
  • Complex Interacts with WRN and Is Crucial to Regulate Its Response to Replication Fork Stalling
    Oncogene (2012) 31, 2809–2823 & 2012 Macmillan Publishers Limited All rights reserved 0950-9232/12 www.nature.com/onc ORIGINAL ARTICLE The RAD9–RAD1–HUS1 (9.1.1) complex interacts with WRN and is crucial to regulate its response to replication fork stalling P Pichierri, S Nicolai, L Cignolo1, M Bignami and A Franchitto Department of Environment and Primary Prevention, Istituto Superiore di Sanita`, Rome, Italy The WRN protein belongs to the RecQ family of DNA preference toward substrates that mimic structures helicases and is implicated in replication fork restart, but associated with stalled replication forks (Brosh et al., how its function is regulated remains unknown. We show 2002; Machwe et al., 2006) and WS cells exhibit that WRN interacts with the 9.1.1 complex, one of enhanced instability at common fragile sites, chromo- the central factors of the replication checkpoint. This somal regions especially prone to replication fork interaction is mediated by the binding of the RAD1 stalling (Pirzio et al., 2008). How WRN favors recovery subunit to the N-terminal region of WRN and is of stalled forks and prevents DNA breakage upon instrumental for WRN relocalization in nuclear foci and replication perturbation is not fully understood. It has its phosphorylation in response to replication arrest. We been suggested that WRN might facilitate replication also find that ATR-dependent WRN phosphorylation restart by either promoting recombination or processing depends on TopBP1, which is recruited by the 9.1.1 intermediates at stalled forks in a way that counteracts complex in response to replication arrest. Finally, we unscheduled recombination (Franchitto and Pichierri, provide evidence for a cooperation between WRN and 2004; Pichierri, 2007; Sidorova, 2008).
    [Show full text]
  • Phase Ib Study of Pevonedistat, a NEDD8-Activating Enzyme Inhibitor
    Investigational New Drugs (2019) 37:87–97 https://doi.org/10.1007/s10637-018-0610-0 PHASE I STUDIES Phase Ib study of pevonedistat, a NEDD8-activating enzyme inhibitor, in combination with docetaxel, carboplatin and paclitaxel, or gemcitabine, in patients with advanced solid tumors A Craig Lockhart1 & Todd M. Bauer2 & Charu Aggarwal3 & Carrie B. Lee4 & RDonaldHarvey5 & Roger B. Cohen6 & Farhad Sedarati7 & Tsz Keung Nip8 & Hélène Faessel9 & Ajeeta B. Dash10 & Bruce J. Dezube7 & Douglas V. Faller7 & Afshin Dowlati11 Received: 1 March 2018 /Accepted: 11 May 2018 /Published online: 21 May 2018 # The Author(s) 2018 Summary Purpose This phase Ib study (NCT01862328) evaluated the maximum tolerated dose (MTD), safety, and efficacy of pevonedistat in combination with standard-of-care chemotherapies in patients with solid tumors. Methods Patients received pevonedistat with docetaxel (arm 1, n = 22), carboplatin plus paclitaxel (arm 2, n = 26), or gemcitabine (arm 3, n = 10) in 21-days (arms 1 and 2) or 28-days (arm 3) cycles. A lead-in cohort (arm 2a, n = 6) determined the arm 2 carboplatin dose. Dose escalation proceeded via continual modified reassessment. Results Pevonedistat MTD was 25 mg/m2 (arm 1) or 20 mg/m2 (arm 2); arm 3 was discontinued due to poor tolerability. Fifteen (23%) patients experienced dose-limiting toxicities during cycle 1 (grade ≥3 liver enzyme elevations, febrile neutropenia, and thrombocytopenia), managed with dose holds or reductions. Drug-related adverse events (AEs) occurred in 95% of patients. Most common AEs included fatigue (56%) and nausea (50%). One drug-related death occurred in arm 3 (febrile neutropenia). Pevonedistat exposure increased when co-administered with carboplatin plus paclitaxel; no obvious changes were observed when co-administered with docetaxel or gemcitabine.
    [Show full text]
  • Pevonedistat (MLN4924), a First-In-Class NEDD8-Activating Enzyme Inhibitor, in Patients with Acute Myeloid Leukaemia and Myelodysplastic Syndromes: a Phase 1 Study
    research paper Pevonedistat (MLN4924), a First-in-Class NEDD8-activating enzyme inhibitor, in patients with acute myeloid leukaemia and myelodysplastic syndromes: a phase 1 study Ronan T. Swords,1 Harry P. Erba,2 Summary Daniel J. DeAngelo,3 Dale L. Bixby,2 This trial was conducted to determine the dose-limiting toxicities (DLTs) Jessica K. Altman,4 Michael Maris,5 Zhaowei Hua,6 Stephen J. Blakemore,6 and maximum tolerated dose (MTD) of the first in class NEDD8-activating Helene Faessel,6 Farhad Sedarati,6 enzyme (NAE) inhibitor, pevonedistat, and to investigate pevonedistat Bruce J. Dezube,6 Francis J. Giles4 and pharmacokinetics and pharmacodynamics in patients with acute myeloid Bruno C. Medeiros7 leukaemia (AML) and myelodysplastic syndromes (MDS). Pevonedistat was 1Leukemia Program, Sylvester Comprehensive administered via a 60-min intravenous infusion on days 1, 3 and 5 (sche- Cancer Center, Miami, FL, 2Division of Hema- dule A, n = 27), or days 1, 4, 8 and 11 (schedule B, n = 26) every 21-days. tology/Oncology, University of Michigan, Ann Dose escalation proceeded using a standard ‘3 + 3’ design. Responses were 3 Arbor, MI, Department of Medical Oncology, assessed according to published guidelines. The MTD for schedules A and Dana-Farber Cancer Institute, Boston, MA, B were 59 and 83 mg/m2, respectively. On schedule A, hepatotoxicity was 4 Northwestern Medicine Developmental Thera- dose limiting. Multi-organ failure (MOF) was dose limiting on schedule B. peutics Institute, Northwestern University, The overall complete (CR) and partial (PR) response rate in patients trea- Chicago, IL, 5Colorado Blood Cancer Institute, ted at or below the MTD was 17% (4/23, 2 CRs, 2 PRs) for schedule A Denver, CO, 6Takeda Pharmaceuticals Interna- tional Co., Cambridge, MA, and 7Division of and 10% (2/19, 2 PRs) for schedule B.
    [Show full text]
  • HUS1 Regulates in Vivo Responses to Genotoxic Chemotherapies
    Oncogene (2016) 35, 662–669 © 2016 Macmillan Publishers Limited All rights reserved 0950-9232/16 www.nature.com/onc SHORT COMMUNICATION HUS1 regulates in vivo responses to genotoxic chemotherapies G Balmus1, PX Lim1, A Oswald1, KR Hume1,2, A Cassano1, J Pierre1, A Hill1, W Huang3, A August3, T Stokol4, T Southard1 and RS Weiss1 Cells are under constant attack from genotoxins and rely on a multifaceted DNA damage response (DDR) network to maintain genomic integrity. Central to the DDR are the ATM and ATR kinases, which respond primarily to double-strand DNA breaks (DSBs) and replication stress, respectively. Optimal ATR signaling requires the RAD9A-RAD1-HUS1 (9-1-1) complex, a toroidal clamp that is loaded at damage sites and scaffolds signaling and repair factors. Whereas complete ATR pathway inactivation causes embryonic lethality, partial Hus1 impairment has been accomplished in adult mice using hypomorphic (Hus1neo) and null (Hus1Δ1) Hus1 alleles, and here we use this system to define the tissue- and cell type-specific actions of the HUS1-mediated DDR in vivo. Hus1neo/Δ1 mice showed hypersensitivity to agents that cause replication stress, including the crosslinking agent mitomycin C (MMC) and the replication inhibitor hydroxyurea, but not the DSB inducer ionizing radiation. Analysis of tissue morphology, genomic instability, cell proliferation and apoptosis revealed that MMC treatment caused severe damage in highly replicating tissues of mice with partial Hus1 inactivation. The role of the 9-1-1 complex in responding to MMC was partially ATR-independent, as a HUS1 mutant that was proficient for ATR-induced checkpoint kinase 1 phosphorylation nevertheless conferred MMC hypersensitivity.
    [Show full text]
  • Structure and Functional Implications of the Human Rad9-Hus1-Rad1
    Supplemental Material can be found at: http://www.jbc.org/content/suppl/2009/06/17/C109.022384.DC1.html ACCELERATED PUBLICATION This paper is available online at www.jbc.org THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 284, NO. 31, pp. 20457–20461, July 31, 2009 © 2009 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. Structure and Functional onto DNA lesion sites by Rad17-RFC2–5 (which consists of one large subunit, Rad17, and four small subunits, RFC2–5), where Implications of the Human it triggers the activation of the cell cycle checkpoint (3, 4). Rad9-Hus1-Rad1 Cell Cycle Moreover, the 9-1-1 complex can also directly participate in DNA repair via physical association with many factors involved *□S Checkpoint Complex in base excision repair (BER), translesion synthesis, homolo- Received for publication, May 18, 2009, and in revised form, June 5, 2009 gous recombination, and mismatch repair pathways (5–9). Published, JBC Papers in Press, June 17, 2009, DOI 10.1074/jbc.C109.022384 Although both the 9-1-1 and the PCNA complexes perform Min Xu‡§, Lin Bai‡§, Yong Gong‡, Wei Xie‡§, Haiying Hang‡1, ‡2 critical functions in eukaryotic cells with predicted similar and Tao Jiang structures (10), their specific roles are distinct. First, the 9-1-1 ‡ From the National Laboratory of Biomacromolecules, Institute of complex is a DNA damage sensor in the cell cycle checkpoint Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101 and the §Graduate University of Chinese Academy but does not function as a scaffold for the major DNA replica- of Sciences, 19A Yuquan Road, Shijingshan District, Beijing 100039, China tion factors; however, PCNA plays exactly the opposite role (1, 11).
    [Show full text]
  • Rabbit Anti-Phospho-C-Abl-SL4086R-FITC
    SunLong Biotech Co.,LTD Tel: 0086-571- 56623320 Fax:0086-571- 56623318 E-mail:[email protected] www.sunlongbiotech.com Rabbit Anti-phospho-c-Abl SL4086R-FITC Product Name: Anti-phospho-c-Abl(Tyr276)/FITC Chinese Name: FITC标记的磷酸化非受体酪氨酸激酶c-Abl抗体 Abelson Murine Leukemia Viral Oncogene Homolog 1; Abelson murine leukemia viral v abl oncogene homolog 1; Abl 1; ABL; Abl protein; Abl1; Bcr/c abl oncogene protein; Alias: JTK 7; JTK7; p150 ; Proto oncogene tyrosine protein kinase ABL1; Transformation gene oncogene ABL; v abl Abelson murine leukemia viral oncogene homolog 1; v abl. Organism Species: Rabbit Clonality: Polyclonal React Species: Human,Mouse,Rat,Dog,Pig,Cow,Horse,Rabbit, IF=1:50-200 Applications: not yet tested in other applications. optimal dilutions/concentrations should be determined by the end user. Molecular weight: 126kDa Cellular localization: The cell membrane Form: Lyophilized or Liquid Concentration: 1mg/ml KLHwww.sunlongbiotech.com conjugated Synthesised phosphopeptide derived from human c-Abl isoform b immunogen: around the phosphorylation site of Tyr276 Lsotype: IgG Purification: affinity purified by Protein A Storage Buffer: 0.01M TBS(pH7.4) with 1% BSA, 0.03% Proclin300 and 50% Glycerol. Store at -20 °C for one year. Avoid repeated freeze/thaw cycles. The lyophilized antibody is stable at room temperature for at least one month and for greater than a year Storage: when kept at -20°C. When reconstituted in sterile pH 7.4 0.01M PBS or diluent of antibody the antibody is stable for at least two weeks at 2-4 °C. background: The c Abl proto oncogene encodes a protein tyrosine kinase that is located in the Product Detail: cytoplasm and nucleus.
    [Show full text]