Exposure Time Versus Cytotoxicity for Anticancer Agents

Total Page:16

File Type:pdf, Size:1020Kb

Exposure Time Versus Cytotoxicity for Anticancer Agents Cancer Chemotherapy and Pharmacology (2019) 84:359–371 https://doi.org/10.1007/s00280-019-03863-w ORIGINAL ARTICLE Exposure time versus cytotoxicity for anticancer agents David M. Evans1 · Jianwen Fang3 · Thomas Silvers1 · Rene Delosh1 · Julie Laudeman1 · Chad Ogle1 · Russell Reinhart1 · Michael Selby1 · Lori Bowles1 · John Connelly1 · Erik Harris1 · Julia Krushkal3 · Larry Rubinstein3 · James H. Doroshow2 · Beverly A. Teicher2,4 Received: 11 February 2019 / Accepted: 2 May 2019 / Published online: 17 May 2019 © This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply 2019 Abstract Purpose Time is a critical factor in drug action. The duration of inhibition of the target or residence time of the drug molecule on the target often guides drug scheduling. Methods The efects of time on the concentration-dependent cytotoxicity of approved and investigational agents [300 com- pounds] were examined in the NCI60 cell line panel in 2D at 2, 3, 7 and in 3D 11 days. Results There was a moderate positive linear relationship between data from the 2-day NCI60 screen and the 3-, 7- and 11-day and a strong positive linear relationship between 3-, 7- and 11-day luminescence screen IC50s by Pearson correlation analysis. Cell growth inhibition by agents selective for a specifc cell cycle phase plateaued when susceptible cells were growth inhibited or killed. As time increased the depth of cell growth inhibition increased without change in the IC 50. DNA interactive agents had decreasing IC50s with increasing exposure time. Epigenetic agents required longer exposure times; several were only cytotoxic after 11 days’ exposure. For HDAC inhibitors, time had little or no efect on concentration response. There were potency diferences amongst the three BET bromodomain inhibitors tested, and an exposure duration efect. The PARP inhibitors, rucaparib, niraparib, and veliparib reached IC50s < 10 μM in some cell lines after 11 days. Conclusions The results suggest that variations in compound exposure time may refect either mechanism of action or com- pound chemical half-life. The activity of slow-acting compounds may optimally be assessed in spheroid models that can be monitored over prolonged incubation times. Keywords CxT · NCI60 · Concentration times time · Epigenetic agents Introduction Electronic supplementary material The online version of this Frequently, the cytotoxicity of anticancer drugs can be esti- article (https ://doi.org/10.1007/s0028 0-019-03863 -w) contains mated as the product of concentration and time for direct supplementary material, which is available to authorized users. cytotoxins such as DNA-damaging agents. However, for cell cycle-specifc agents, maximal cytotoxicity is dependent on * Beverly A. Teicher [email protected]; [email protected] cellular generation/doubling time [1, 2]. Since anticancer therapeutics have moved away from cytotoxic agents to epi- 1 Molecular Pharmacology Group, Leidos Biomedical genetic modifers and targeted drugs, the efect of time on Research, Inc., Frederick National Laboratory for Cancer cellular response needs to be re-examined. Cell cycle selec- Research, Frederick, MD 21702, USA tivity is an important factor in understanding the efect of 2 Developmental Therapeutics Program, Division of Cancer time on cytotoxicity. Drugs such as methotrexate and cyto- Treatment and Diagnosis, National Cancer Institute, Rockville, MD 20852, USA sine arabinoside are specifc for cells in S-phase; while drugs such as vincristine and paclitaxel are specifcally cytotoxic 3 Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, toward cells in M-phase. Most drugs are more cytotoxic MD 20852, USA toward cells in exponential growth than toward plateau or 4 Molecular Pharmacology Branch, National Cancer Institute, stationary phase cells. RM 4-W602, MSC 9735, 9609 Medical Center Drive, Bethesda, MD 20892, USA Vol.:(0123456789)1 3 360 Cancer Chemotherapy and Pharmacology (2019) 84:359–371 Many approved drugs are relatively stable in cell culture at: https ://dtp.nci.nih.gov/branc hes/dscb/oncol ogy_drugs medium but have variable half-lives in human circulation et_expla natio n.html) and an investigational agent library, (Supplemental Table 1; [3, 4]). Doxorubicin, actinomycin acquired by synthesis or purchase. D, bleomycin, and vinblastine were stable in cell culture media for up to 10 days; however, these drugs have circulat- NCI60 screen ing half-lives in humans of 14.2, 14, 4, and 2.9 h, respec- tively. In cell culture media, etoposide lost 60% activity in The cell lines were grown in RPMI 1640 medium (Life 10 days; by contrast, the circulating half-life for etoposide Technologies, Gibco) containing 5% fetal bovine serum in humans is 3.62 h [5]. By HPLC, mitomycin C was stable (Life Technologies, HyClone, Logan, UT) and 2 mM for 7 days at 37 °C in cell culture media but doxorubicin L-glutamine. For experiments, cells were plated in 96-well had a half-life of 10–20 h under the same conditions [6]. In plates in 100 μl at cell numbers ranging from 5 × 103 to human circulation, mitomycin has a half-life of 0.81 h and 4 × 104 cells/well. The plates were incubated at 37 °C in doxorubicin of 14.2 h. Cisplatin had a half-life of 48 h in humid 5% CO2, for 24 h prior to compound addition. Com- cell culture media due to chemical reactivity and has a half- pounds in DMSO were added at fve concentrations from life of 0.44 h in human circulation [7, 8]. Decitabine, on the 100 to 0.01 μM. Following compound addition, the plates other hand, was relatively stable in cell culture media but has were incubated for 48 h at 37 °C, in humidifed incubators a plasma half-life of 20 min in humans’ due to high levels of with 5% CO 2. For adherent cells, the assay was terminated liver cytidine deaminase which metabolizes the drug to an by addition of cold TCA (fnal concentration, 10% TCA) inactive species [9]. The hydroxamic acid class of histone and incubated for 60 min at 4 °C. The plates were washed deacetylase inhibitors including vorinostat, belinostat, and with water and air-dried. Sulforhodamine B (SRB) solution panobinostat had very limited stability (1 day) in aqueous (100 μl) at 0.4% (w/v) in 1% acetic acid was added, and solution, were not stable in human plasma but had better plates were incubated for 10 min at room temperature. After stability in human serum (Supplemental Table 1; [10–12]). staining, unbound dye was removed by washing with 1% The mercaptoacetamide class of histone deacetylase inhibi- acetic acid and the plates were air dried. Bound stain was tors exemplifed by romidepsin was, in general, more stable solubilized with 10 mM trizma base, and the absorbance in aqueous solution, and plasma than hydroxamic acids [13]. read at 515 nm. For suspension cells, the methodology was The current study explores the efect of exposure time the same except that the assay was terminated by fxing set- (2–11 days) on the cytotoxicity and growth inhibition of tled cells at the bottom of the wells by gently adding 50 μl several classes of anticancer agents. of 80% TCA (fnal concentration, 16% TCA). 3‑ and 7‑day monolayer screen Material and methods 12 cell lines (11 lines and A549/ATCC) were screened per Cell lines run. Day 1, the cells were collected and suspended in 300 ml of media (RPMI 1640 supplemented with 5% FBS and NCI60 cell lines were obtained from the NCI Developmental 2 mM L-glutamine), then plated in 42 μl in 384-well plates Therapeutics Program Tumor Repository. For each lot of (CulturPlates, PE, Waltham, MA) using a Tecan Freedom cells, the Repository performed Applied Biosystems Amp- Evo. After incubation overnight, the Tecan Evo was used for FLSTR Identifler testing with PCR amplifcation to confrm compound addition. Each compound was added at 9 concen- consistency with the published. trations (1.5 nM–10 μM; DMSO concentration 0.25%), then Identifler STR profle for the given cell line [14, 15]. the plates were incubated for 3 or 7 days. The incubation was Each cell line was tested for mycoplasma when it was terminated by adding Cell Titer Glo (Promega, Madison, accepted into the repository; routine mycoplasma testing WI), and luminescence was determined. All assays were of lots was not performed. Cells were kept in continuous performed in triplicate. culture for no more than 20 passages. The optimal seed- ing densities for each of the cell lines for each time point 11‑day spheroid screen assessed were determined prior to performing the concentra- tion response studies [16]. All steps in the assay were performed using an automated high-throughput screening system consisting of two robotic Compounds arms interfaced with liquid handlers, pipettors, automated CO2 incubators, dispensers, shakers and plate readers. The Three hundred compounds were selected for the assays from screen was controlled via Momentum scheduling soft- the FDA-approved anticancer drugs (available from NCI ware (Thermo, Waltham, MA). Day 0: cells were plated in 1 3 Cancer Chemotherapy and Pharmacology (2019) 84:359–371 361 384-well Ultra Low Attachment (ULA) plates (Corning, Results NY) at densities ranging from 1 × 103 to 2 × 104 in RPMI media containing 10% fetal bovine serum in a volume of The NCI60 screen is the best known and most widely used 42 μL per well [16]. Spheroid formation and morphology compound cell-based screen to determine potential anti- were monitored using brightfeld imaging on a Cytation cancer activity [18–20]. The screen includes human tumor 3 high-content imaging system (BioTek, Winooski, VT) cell lines from nine major tumor types and uses a sulforho- equipped with a 4 × objective. Images were captured dig- damine B visible absorption endpoint.
Recommended publications
  • 214120Orig1s000
    CENTER FOR DRUG EVALUATION AND RESEARCH APPLICATION NUMBER: 214120Orig1s000 MULTI-DISCIPLINE REVIEW Summary Review Office Director Cross Discipline Team Leader Review Clinical Review Non-Clinical Review Statistical Review Clinical Pharmacology Review NDA Multidisciplinary Review and Evaluation Application Number NDA 214120 Application Type Type 3 Priority or Standard Priority Submit Date 3/3/2020 Received Date 3/3/2020 PDUFA Goal Date 9/3/2020 Office/Division OOD/DHM1 Review Completion Date 9/1/2020 Applicant Celgene Corporation Established Name Azacitidine (Proposed) Trade Name Onureg Pharmacologic Class Nucleoside metabolic inhibitor Formulations Tablet (200 mg, 300 mg) (b) (4) Applicant Proposed Indication/Population Recommendation on Regulatory Regular approval Action Recommended Indication/ For continued treatment of adult patients with acute Population myeloid leukemia who achieved first complete remission (CR) or complete remission with incomplete blood count recovery (CRi) following intensive induction chemotherapy and are not able to complete intensive curative therapy. SNOMED CT for the Recommended 91861009 Indication/Population Recommended Dosing Regimen 300 mg orally daily on Days 1 through 14 of each 28-day cycle Reference ID: 4664570 NDA Multidisciplinary Review and Evaluation NDA 214120 Onureg (azacitidine tablets) TABLE OF CONTENTS TABLE OF CONTENTS ................................................................................................................................... 2 TABLE OF TABLES .......................................................................................................................................
    [Show full text]
  • Expression of Genes by Hypomethylating Agents
    Wolff et al. Cell Communication and Signaling (2017) 15:13 DOI 10.1186/s12964-017-0168-z REVIEW Open Access The double-edged sword of (re)expression of genes by hypomethylating agents: from viral mimicry to exploitation as priming agents for targeted immune checkpoint modulation Florian Wolff1, Michael Leisch2, Richard Greil2,3,4, Angela Risch1,4 and Lisa Pleyer2,3,4* Abstract Hypomethylating agents (HMAs) have been widely used over the last decade, approved for use in myelodysplastic syndrome (MDS), chronic myelomonocytic leukemia (CMML) and acute myeloid leukemia (AML). The proposed central mechanism of action of HMAs, is the reversal of aberrant methylation in tumor cells, thus reactivating CpG-island promoters and leading to (re)expression of tumor suppressor genes. Recent investigations into the mode of action of azacitidine (AZA) and decitabine (DAC) have revealed new molecular mechanisms that impinge on tumor immunity via induction of an interferon response, through activation of endogenous retroviral elements (ERVs) that are normally epigenetically silenced. Although the global demethylation of DNA by HMAs can induce anti-tumor effects, it can also upregulate the expression of inhibitory immune checkpoint receptors and their ligands, resulting in secondary resistance to HMAs. Recent studies have, however, suggested that this could be exploited to prime or (re)sensitize tumors to immune checkpoint inhibitor therapies. In recent years, immune checkpoints have been targeted by novel therapies, with the aim of (re)activating the host immune system to specifically eliminate malignant cells. Antibodies blocking checkpoint receptors have been FDA-approved for some solid tumors and a plethora of clinical trials testing these and other checkpoint inhibitors are under way.
    [Show full text]
  • Phase I Trial of the PARP Inhibitor Olaparib and AKT Inhibitor Capivasertib in Patients with BRCA1/2- and Non–BRCA1/2-Mutant Cancers
    Published OnlineFirst June 12, 2020; DOI: 10.1158/2159-8290.CD-20-0163 RESEARCH ARTICLE Phase I Trial of the PARP Inhibitor Olaparib and AKT Inhibitor Capivasertib in Patients with BRCA1/2- and Non–BRCA1/2-Mutant Cancers Timothy A. Yap1,2, Rebecca Kristeleit3, Vasiliki Michalarea1, Stephen J. Pettitt4,5, Joline S.J. Lim1, Suzanne Carreira2, Desamparados Roda1,2, Rowan Miller3, Ruth Riisnaes2, Susana Miranda2, Ines Figueiredo2, Daniel Nava Rodrigues2, Sarah Ward1,2, Ruth Matthews1,2, Mona Parmar1,2, Alison Turner1,2, Nina Tunariu1, Neha Chopra1,4, Heidrun Gevensleben2, Nicholas C. Turner1,4, Ruth Ruddle2, Florence I. Raynaud2, Shaun Decordova2, Karen E. Swales2, Laura Finneran2, Emma Hall2, Paul Rugman6, Justin P.O. Lindemann6, Andrew Foxley6, Christopher J. Lord4,5, Udai Banerji1,2, Ruth Plummer7, Bristi Basu8, Juanita S. Lopez1,2, Yvette Drew7, and Johann S. de Bono1,2 Downloaded from cancerdiscovery.aacrjournals.org on September 30, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst June 12, 2020; DOI: 10.1158/2159-8290.CD-20-0163 ABSTRACT Preclinical studies have demonstrated synergy between PARP and PI3K/AKT path- way inhibitors in BRCA1 and BRCA2 (BRCA1/2)–deficient andBRCA1/2 -proficient tumors. We conducted an investigator-initiated phase I trial utilizing a prospective intrapatient dose- escalation design to assess two schedules of capivasertib (AKT inhibitor) with olaparib (PARP inhibi- tor) in 64 patients with advanced solid tumors. Dose expansions enrolled germline BRCA1/2-mutant tumors, or BRCA1/2 wild-type cancers harboring somatic DNA damage response (DDR) or PI3K–AKT pathway alterations. The combination was well tolerated. Recommended phase II doses for the two schedules were: olaparib 300 mg twice a day with either capivasertib 400 mg twice a day 4 days on, 3 days off, or capivasertib 640 mg twice a day 2 days on, 5 days off.
    [Show full text]
  • Epigenetics in Clinical Practice: the Examples of Azacitidine and Decitabine in Myelodysplasia and Acute Myeloid Leukemia
    Leukemia (2013) 27, 1803–1812 & 2013 Macmillan Publishers Limited All rights reserved 0887-6924/13 www.nature.com/leu SPOTLIGHT REVIEW Epigenetics in clinical practice: the examples of azacitidine and decitabine in myelodysplasia and acute myeloid leukemia EH Estey Randomized trials have clearly demonstrated that the hypomethylating agents azacitidine and decitabine are more effective than ‘best supportive care’(BSC) in reducing transfusion frequency in ‘low-risk’ myelodysplasia (MDS) and in prolonging survival compared with BSC or low-dose ara-C in ‘high-risk’ MDS or acute myeloid leukemia (AML) with 21–30% blasts. They also appear equivalent to conventional induction chemotherapy in AML with 420% blasts and as conditioning regimens before allogeneic transplant (hematopoietic cell transplant, HCT) in MDS. Although azacitidine or decitabine are thus the standard to which newer therapies should be compared, here we discuss whether the improvement they afford in overall survival is sufficient to warrant a designation as a standard in treating individual patients. We also discuss pre- and post-treatment covariates, including assays of methylation to predict response, different schedules of administration, combinations with other active agents and use in settings other than active disease, in particular post HCT. We note that rational development of this class of drugs awaits delineation of how much of their undoubted effect in fact results from hypomethylation and reactivation of gene expression. Leukemia (2013) 27, 1803–1812; doi:10.1038/leu.2013.173
    [Show full text]
  • Biological Heterogeneity of Chondrosarcoma: from (Epi) Genetics Through Stemness and Deregulated Signaling to Immunophenotype
    cancers Review Biological Heterogeneity of Chondrosarcoma: From (Epi) Genetics through Stemness and Deregulated Signaling to Immunophenotype Agnieszka Zaj ˛ac 1 , Sylwia K. Król 2 , Piotr Rutkowski 1 and Anna M. Czarnecka 1,3,* 1 Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; [email protected] (A.Z.); [email protected] (P.R.) 2 Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; [email protected] 3 Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-176 Warsaw, Poland * Correspondence: [email protected] Simple Summary: Chondrosarcoma (ChS) is the second most frequently diagnosed malignant bone tumor of cartilaginous origin and is generally resistant to standard treatment options. In this paper, we aim to review the current state of the knowledge regarding ChS. We discuss the genetic, epigenetic, and molecular abnormalities underlying its substantial biological and clinical heterogeneity. This review summarizes the critical genetic and molecular drivers of ChS development and progression, contributing to its radio- and chemotherapy resistance. We describe genomic aberrations and point mutations, as well as epigenetic modifications and deregulated signal transduction pathways. We Citation: Zaj ˛ac,A.; Król, S.K.; provide an insight into the stem-like characteristics and immunophenotype of ChS. The paper also Rutkowski, P.; Czarnecka, A.M. outlines potential diagnostic and prognostic biomarkers of ChS and recently identified novel targets Biological Heterogeneity of for future pharmacological interventions in patients. Chondrosarcoma: From (Epi) Genetics through Stemness and Abstract: Deregulated Signaling to Chondrosarcoma (ChS) is a primary malignant bone tumor.
    [Show full text]
  • Demethylation and Upregulation of an Oncogene Post Hypomethylating Treatment
    medRxiv preprint doi: https://doi.org/10.1101/2020.07.21.20157776; this version posted July 26, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Title: Demethylation and upregulation of an oncogene post hypomethylating treatment Authors: Yao-Chung Liu1,2,3,4°, Emiliano Fabiani5°, Junsu Kwon6°, Chong Gao1, Giulia Falconi5, Lia Valentini5, Carmelo Gurnari5, Yanjing V. Liu6, Adrianna I. Jones7, Junyu Yang1, Henry Yang6, Julie A. I. Thoms8, Ashwin Unnikrishnan9, John E. Pimanda8,9,10, Rongqing Pan11, Maria Teresa Voso5*, Daniel G. Tenen6,7*, Li Chai1* Affiliations: 1Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA 2Division of Hematology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan 3Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan 4Program in Molecular Medicine, School of Life Science, National Yang-Ming University, Taipei, Taiwan 5Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy 6Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore 7Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115 USA 8School of Medical Sciences and Lowy Cancer Research Centre, Faculty of Medicine, UNSW Sydney, NSW 2052, Australia 9Prince of Wales Clinical School and Lowy Cancer Research Centre, Faculty of Medicine, UNSW Sydney, NSW 2052, Australia 10Department of Haematology, Prince of Wales Hospital, Randwick, NSW 2031, Australia 11 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115 °These authors contibute equally to this work *Co-corresponding authors: [email protected], [email protected], and [email protected] The authors have declared that no conflict of interest exists.
    [Show full text]
  • The Influence of Cell Cycle Regulation on Chemotherapy
    International Journal of Molecular Sciences Review The Influence of Cell Cycle Regulation on Chemotherapy Ying Sun 1, Yang Liu 1, Xiaoli Ma 2 and Hao Hu 1,* 1 Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China; [email protected] (Y.S.); [email protected] (Y.L.) 2 Qingdao Institute of Measurement Technology, Qingdao 266000, China; [email protected] * Correspondence: [email protected] Abstract: Cell cycle regulation is orchestrated by a complex network of interactions between proteins, enzymes, cytokines, and cell cycle signaling pathways, and is vital for cell proliferation, growth, and repair. The occurrence, development, and metastasis of tumors are closely related to the cell cycle. Cell cycle regulation can be synergistic with chemotherapy in two aspects: inhibition or promotion. The sensitivity of tumor cells to chemotherapeutic drugs can be improved with the cooperation of cell cycle regulation strategies. This review presented the mechanism of the commonly used chemotherapeutic drugs and the effect of the cell cycle on tumorigenesis and development, and the interaction between chemotherapy and cell cycle regulation in cancer treatment was briefly introduced. The current collaborative strategies of chemotherapy and cell cycle regulation are discussed in detail. Finally, we outline the challenges and perspectives about the improvement of combination strategies for cancer therapy. Keywords: chemotherapy; cell cycle regulation; drug delivery systems; combination chemotherapy; cancer therapy Citation: Sun, Y.; Liu, Y.; Ma, X.; Hu, H. The Influence of Cell Cycle Regulation on Chemotherapy. Int. J. 1. Introduction Mol. Sci. 2021, 22, 6923. https:// Chemotherapy is currently one of the main methods of tumor treatment [1].
    [Show full text]
  • PARP Inhibitor Olaparib Causes No Potentiation of the Bleomycin Effect
    International Journal of Molecular Sciences Article PARP Inhibitor Olaparib Causes No Potentiation of the Bleomycin Effect in VERO Cells, Even in the Presence of Pooled ATM, DNA-PK, and LigIV Inhibitors Valentina Perini 1, Michelle Schacke 1, Pablo Liddle 1, Salomé Vilchez-Larrea 2 , Deborah J. Keszenman 3,* and Laura Lafon-Hughes 1,* 1 Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Departamento de Genética, Montevideo 11.600, Uruguay; [email protected] (V.P.); [email protected] (M.S.); [email protected] (P.L.) 2 Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires 1428, Argentina; [email protected] 3 Laboratorio de Radiobiología Médica y Ambiental, Grupo de Biofisicoquímica, Centro Universitario Regional Litoral Norte, Universidad de la República (UdelaR), Salto 50.000, Uruguay * Correspondence: [email protected] (D.J.K.); [email protected] (L.L.-H.) Received: 30 September 2020; Accepted: 19 October 2020; Published: 5 November 2020 Abstract: Poly(ADP-ribosyl)polymerase (PARP) synthesizes poly(ADP-ribose) (PAR), which is anchored to proteins. PAR facilitates multiprotein complexes’ assembly. Nuclear PAR affects chromatin’s structure and functions, including transcriptional regulation. In response to stress, particularly genotoxic stress, PARP activation facilitates DNA damage repair. The PARP inhibitor Olaparib (OLA) displays synthetic lethality with mutated homologous recombination proteins (BRCA-1/2), base excision repair proteins (XRCC1, Polβ), and canonical nonhomologous end joining (LigIV). However, the limits of synthetic lethality are not clear. On one hand, it is unknown whether any limiting factor of homologous recombination can be a synthetic PARP lethality partner.
    [Show full text]
  • A Phase II Study of Omacetaxine (OM) in Patients with Intermediate-1 and Higher Risk Myelodysplastic Syndrome (MDS) Post Hypomethylating Agent (HMA) Failure 2013-0870
    Protocol Page A Phase II Study of Omacetaxine (OM) in Patients with Intermediate-1 and Higher Risk Myelodysplastic Syndrome (MDS) post Hypomethylating Agent (HMA) Failure 2013-0870 Core Protocol Information Short Title Omacetaxine in Patients with Intermediate-1 and Higher Risk MDS post HMA Failure Study Chair: Elias Jabbour Additional Contact: Jhinelle L. Graham Vicky H. Zoeller Leukemia Protocol Review Group Additional Memo Recipients: Recipients List OPR Recipients (for OPR use only) None Study Staff Recipients None Department: Leukemia Phone: 713-792-4764 Unit: 0428 Full Title: A Phase II Study of Omacetaxine (OM) in Patients with Intermediate-1 and Higher Risk Myelodysplastic Syndrome (MDS) post Hypomethylating Agent (HMA) Failure Protocol Type: Standard Protocol Protocol Phase: Phase II Version Status: Activated -- Closed to new patient entry as of 08/05/2018 Version: 08 Document Status: Saved as "Final" Submitted by: Vicky H. Zoeller--9/11/2017 12:24:33 PM OPR Action: Accepted by: Margaret Okoloise -- 9/14/2017 12:09:50 PM Which Committee will review this protocol? The Clinical Research Committee - (CRC) Protocol Body 2013-0870 March 6, 2017 1 A Phase II Study of Omacetaxine (OM) in Patients with Intermediate-1 and Higher Risk Myelodysplastic Syndrome (MDS) post Hypomethylating Agent (HMA) Failure 2013-0870 March 6, 2017 2 Table of Contents 1.0 Objectives .................................................................................................. 3 2.0 Background ..............................................................................................
    [Show full text]
  • Immunotherapy of MDS: You Can Run, but You Can’T Hide
    Author Manuscript Published OnlineFirst on December 28, 2017; DOI: 10.1158/1078-0432.CCR-17-2960 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Immunotherapy of MDS: you can run, but you can’t hide Ephraim Joseph Fuchs, MD, MBA Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Baltimore, MD, USA e-mail: [email protected] Running title: Hypomethylating agents to enhance cancer vaccines for MDS Conflicts of Interest: The author has no conflicts of interest 1 Downloaded from clincancerres.aacrjournals.org on October 3, 2021. © 2017 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 28, 2017; DOI: 10.1158/1078-0432.CCR-17-2960 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Summary: The hypomethylating agent decitabine induces expression of the cancer testis antigen NY- ESO-1 in the myeloid cells of patients with myelodysplastic syndrome (MDS). MDS patients treated with decitabine and an NY-ESO-1 vaccine developed NY-ESO-1-specific T cell responses directed against their abnormal myeloid cells, raising hopes for combinatorial immunotherapy of this disease. In this issue of Clinical Cancer Research, Griffiths and colleagues conduct a phase I clinical trial in patients with myelodysplastic syndrome (MDS) of a combinatorial immunotherapy (Figure 1) comprising the hypomethylating agent decitabine plus a vaccine against the “cancer testis” antigen NY-ESO-1 (1). This strategy addresses a critical unmet need in cancer immunotherapy: the treatment of cancers with few available immunologic targets. The success of the immunologic checkpoint inhibitors (CIs) and of chimeric antigen receptor-modified T cells, or CAR T cells, has raised the level of enthusiasm for cancer immunotherapy to a fever pitch.
    [Show full text]
  • Commissioner Final Decisions
    Commissioner for the Department for Medicaid Services Selections for Preferred Products This is a summary of the final Preferred Drug List (PDL) selections made by the Commissioner of the Department for Medicaid Services (DMS) based on the Drug Review and Options for Consideration document prepared for the Pharmacy and Therapeutics (P&T) Advisory Committee’s review on March 21, 2019, and the resulting official Committee recommendations. New Products to Market Epidiolex™ – Non-prefer in the PDL class: Anticonvulsants: Second Generation Length of Authorization: 1 year Epidiolex™ (cannabidiol), a non-psychoactive cannabinoid receptor antagonist, is approved for the treatment of seizures associated with Lennox-Gastaut syndrome or Dravet syndrome in patients ≥ 2 years of age. The mechanism by which cannabidiol exerts its anticonvulsant effects is unknown. Cannabidiol (Epidiolex) is a Schedule V controlled substance. Criteria for Approval: Diagnosis of Lennox-Gastaut syndrome (LGS) OR Dravet syndrome (DS); AND Prescriber is, or has a consultative relationship with, a neurology/epilepsy specialist; AND Trial and failure (e.g., incomplete seizure control) of at least 2 antiepileptic drugs; AND Must be used in adjunct with ≥ 1 antiepileptic drug. Age Limit: > 2 years Drug Class Preferred Agents Non-Preferred Agents Anticonvulsants: Banzel® CC, QL Briviact® QL Second Generation Gabitril® QL Epidiolex™ AE lamotrigine chewable tablets, tablets (except dose Fycompa™ QL packs) Keppra® tablets QL, solution levetiracetam solution, tablets QL Keppra XR® QL Sabril® CC Lamictal® topiramate QL Lamictal ODT® zonisamide QL Lamictal® XR™ QL lamotrigine dose packs lamotrigine ER QL lamotrigine ODT levetiracetam ER QL Qudexy® XR QL Spritam QL tiagabine QL Topamax® QL topiramate ER QL Trokendi XR™ QL vigabatrin Vimpat® QL AE – Age Edit; CC – Clinical Criteria; MD – Medications with Maximum Duration; QL – Quantity Limit; ST – Step Therapy © 2019, Magellan Medicaid Administration, a Magellan Rx Management company.
    [Show full text]
  • New Strategies in Acute Myeloid Leukemia: Redefining Prognostic Markers to Guide Therapy
    Author Manuscript Published OnlineFirst on August 14, 2012; DOI: 10.1158/1078-0432.CCR-12-0313 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Khan et al. New Strategies in AML New Strategies in Acute Myeloid Leukemia: Redefining prognostic markers to guide therapy Irum Khan, Jessica K. Altman and Jonathan D. Licht Division of Hematology/Oncology Robert H. Lurie Comprehensive Cancer Center Northwestern University-Feinberg School of Medicine Running Title: New Strategies in AML Address Correspondence to Jonathan D. Licht, M.D. Division of Hematology/Oncology Robert H. Lurie Comprehensive Cancer Center Northwestern University Feinberg School of Medicine 303 East Superior Street, Lurie 5-123 Chicago, IL 60611 Office: (312) 503-0985 Fax: (312) 503-0189 [email protected] Supported by NCI T32 CA079447 (IK), A Leukemia and Lymphoma Society Specialized Center of Research Excellence (JDL) and a K12 award (JKA) through 8UL1TR000150. Conflict of Interest: JDL- Research Support from Epizyme, Inc. JKA- Advisory Boards for Cellgene, Astellas, Teva 1 Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 14, 2012; DOI: 10.1158/1078-0432.CCR-12-0313 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Khan et al. New Strategies in AML Abstract While the standard therapy of AML has been relatively constant over the past two decades, this may be changing with enhanced technologies allowing for the classification of acute myeloid leukemia (AML) into molecularly distinct subsets. Some specific subsets of AML have an excellent prognosis in response to standard therapy while the poor prognosis of AML associated with specific sets of mutations or chromosomal anomalies require the development of new therapies.
    [Show full text]