Abemaciclib, a Selective CDK4/6 Inhibitor, Enhances The

Total Page:16

File Type:pdf, Size:1020Kb

Abemaciclib, a Selective CDK4/6 Inhibitor, Enhances The Published OnlineFirst May 1, 2018; DOI: 10.1158/1078-0432.CCR-17-3575 Cancer Therapy: Preclinical Clinical Cancer Research Abemaciclib, a Selective CDK4/6 Inhibitor, Enhances the Radiosensitivity of Non–Small Cell Lung Cancer In Vitro and In Vivo Sarwat Naz, Anastasia Sowers, Rajani Choudhuri, Maria Wissler, Janet Gamson, Askale Mathias, John A. Cook, and James B. Mitchell Abstract Purpose: To characterize the ionizing radiation (IR) enhancing sensitivity was lost in cell lines deficient for functional p53 and RB effects and underlying mechanisms of the CDK4/6 inhibitor protein. After IR, abemaciclib treatment inhibited DNA damage abemaciclib in non–small cell lung cancer (NSCLC) cells in vitro repair as measured by gH2AX. Mechanistically, abemaciclib and in vivo. inhibited RB phosphorylation, leading to cell-cycle arrest. It also Experimental Design: IR enhancement by abemaciclib in a inhibited mTOR signaling and reduced intracellular amino acid variety of NSCLC cell lines was assessed by in vitro clonogenic pools, causing nutrient stress. In vivo, abemaciclib, when admin- assay, flow cytometry, and target inhibition verified by immu- istered in an adjuvant setting for the second week after fraction- noblotting. IR-induced DNA damage repair was evaluated by ated IR, further inhibited vasculogenesis and tumor regrowth, gH2AX analysis. Global metabolic alterations by abemaciclib with sustained inhibition of RB/E2F activity, mTOR pathway, and IR combination were evaluated by LC/MS mass spectrom- and HIF-1 expression. In summary, our study signifies inhibiting etry and YSI bioanalyzer. Effects of abemaciclib and IR com- the CDK4/6 pathway by abemaciclib in combination with IR as a bination in vivo were studied by xenograft tumor regrowth promising therapeutic strategy to treat NSCLC. delay, xenograft lysate immunoblotting, and tissue section Conclusions: Abemaciclib in combination with IR enhances immunohistochemistry. NSCLC radiosensitivity in preclinical models, potentially provid- Results: Abemaciclib enhanced the radiosensitivity of NSCLC ing a novel biomarker-driven combination therapeutic strategy cells independent of RAS or EGFR status. Enhancement of radio- for patients with NSCLC. Clin Cancer Res; 1–12. Ó2018 AACR. Introduction gulated pathways by small molecule tyrosine kinase inhibitors (TKI) or receptor monoclonal antibodies (7–9). Although EGFR- Over the last decade, advances in molecular translational TKIs have been useful in the treatment of EGFR-mutant NSCLC, research have heralded major breakthroughs in the understand- most responses have not proved to be durable with many ing, diagnosis, and management of lung cancer, particularly for patients progressing after 7 to 12 months (4, 8, 10). The most the more common (80%) non–small cell lung cancer frequent mechanism (50%) is concurrent acquisition of novel (NSCLC). NSCLC is subclassified by histology and driver muta- mutation in exon 20 of EGFR, encoding for T790M making tions such as mutated KRAS and activating mutations in the tumors refractory to the existing EGFR-TKI therapy (8, 11). Apart epidermal growth factor receptor (EGFR) tyrosine kinase (TK) from EGFR-TKI, radiotherapy either alone or in combination domain (1–4). The two most common EGFR-TK domain muta- with chemotherapy, remains the primary modality of treatment tions are exon 19 deletions (60%) and L858R missense sub- for patients with stage III NSCLC. For stage I and II NSCLC, stitutions resulting in constitutive activation of the receptor radiotherapy is an alternative curative option to surgery for without ligand binding (3, 5, 6). Constitutive activation of patients who are medically inoperable or refuse surgery. Overall receptors or protein kinases stimulates a complex cascade of radiotherapy is an important palliative treatment modality to cross-signaling pathways leading to uncontrolled growth, pro- treat symptoms from the primary or bone or brain metastases liferation, and survival (2, 3). Successful targeted therapies in and improve patients' quality of life (12). Despite these medical NSCLC involve the identification and inhibition of these upre- interventions, 5-year survival rates of NSCLC patients are less than 5% (4, 13). Hence, there is an urgent need to target other signaling pathways or design combination therapy that is more Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, effective than first-line single agents while balancing toxicity and Bethesda, Maryland. costs. Other than the EGFR or MEK/ERK pathway, cyclin D Note: Supplementary data for this article are available at Clinical Cancer kinase 4/6 (CDK4/6) activity is typically deregulated and over- Research Online (http://clincancerres.aacrjournals.org/). active in various cancers including NSCLC (14, 15). Corresponding Author: James B. Mitchell, Center for Cancer Research, National CDK4 and CDK6 are cyclin-dependent kinases that control the Cancer Institute, Building 10, Room B3-B69, 9000 Rockville Pike, Bethesda, MD transition between the G1 and S-phase of the cell cycle. A major 20892. Phone: 240-858-3101; Fax: 240-541-4528; E-mail: james.mitchell3@nih.gov target of CDK4 and CDK6 during cell-cycle progression is the doi: 10.1158/1078-0432.CCR-17-3575 retinoblastoma protein (RB). When RB is phosphorylated, its Ó2018 American Association for Cancer Research. growth-suppressive properties are inactivated. Selective CDK4/6 www.aacrjournals.org OF1 Downloaded from clincancerres.aacrjournals.org on September 23, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst May 1, 2018; DOI: 10.1158/1078-0432.CCR-17-3575 Naz et al. All cell lines were authenticated within the past 6 months by Translational Relevance IDEXX Bioresearch using Cell Check 9 [9 allele marker STR (short- Radiotherapy plays a significant role in the management of tandem repeat) profile and interspecies contamination test; Sup- non–small cell lung cancer (NSCLC). It is useful for all stages plementary File S1]. All cells were cultured in RPMI-1640 (Invi- of disease with or without additional treatment modalities, trogen), supplemented with 10% FBS and 1% penicillin/strepto- such as surgery and chemotherapy. However, the response rate mycin at 37 C in a humidified 5% CO2 atmosphere. All CDK4/6 of some tumors remains problematic. The present study inhibitors were purchased from Selleck Chemicals. A stock solu- evaluates the combination of a highly specific CDK4/6 inhib- tion of drug was prepared in 100% dimethyl sulfoxide (Sigma- itor, abemaciclib, with ionizing radiation (IR) to enhance Aldrich) and stored at À70C. The drug was diluted in fresh media radiation sensitivity of NSCLC in vitro and in vivo. Our data prior to each experiment. Abemaciclib was added to cells for indicate that this novel combination efficiently radiosensi- 24 hours after IR treatment at a concentration of 10 mmol/L. tized proliferative and plateau-phase tumor cells and tumor Clonogenic cell survival studies were performed as described xenografts with minimal normal cell radiosensitization. Abe- before (24, 25). Experiments were repeated three independent maciclib inhibited IR-induced DNA damage repair and caused times. Radiation dose-modifying factors (DMF) were determined RB-dependent cell-cycle arrest. Further, the study identified at 10% survival levels by dividing the radiation dose for control by possible predictive biomarkers (p53, RB, and SDF-1) to guide the radiation dose for drug treated. DMFs > 1.0 indicate enhance- treatment response and efficacy of the combination. Collec- ment of radiosensitivity. tively, this study highlights the CDK4/6 axis as a potential radiation target for NSCLC and warrants assessment of abe- Immunoblotting maciclib in clinical trials as a radiation modifier. Exponentially growing cells were washed twice with chilled PBS and cell pellet were collected as a function of time follow- ing irradiation. Total protein extraction from cultured cells and xenograft tumors were performed as previously described (25). inhibitors "turn off" these kinases and dephosphorylate RB, Acid soluble histone proteins for gH2AX were extracted in resulting in a block of cell-cycle progression in mid-G prevent- 0.2 mol/L sulfuric acid. Protein concentrations were deter- 1 fi ing proliferation of cancer cells (16). Recent studies have mined using BCA reagent (Thermo Fisher Scienti c). Protein identified poor responses to EGFR-TKIs in lung adenocarcino- samples were resolved on SDS-PAGE and immunoblotting was ma patients with the altered CDK4/6-RB pathway (17, 18). In performed as described before (24, 25). Details of antibodies preclinical mouse models, KRAS-driven lung cancer is highly used for immunoblotting are described in Supplementary dependent on CDK4, and genetic or pharmacologic ablation of Experimental Procedures. CDK4 activity has effects on both the establishment and main- fi tenance of these tumors (19, 20). Palbociclib (PD0332991), a ATP and NAD quanti cation selective CDK4/6 inhibitor, has been shown to sensitize lung ATP (catalog no. K354-100) and NAD (catalog no. K337) fl cancer cells to EGFR-TKI, gefitinib (21). In addition, the MEK colorimetric assays were performed using BioVision kit. Brie y, Â 6 inhibitor (trametinib) in combination with palbociclib has 1 10 cells were treated with abemaciclib after IR exposure. significant anti-KRAS-mutant NSCLC activity and radiosensitiz- Samples were collected 24 hours post-IR, and the assays ing effects in preclinical models (22). Silencing CDK4 in breast were performed according to
Recommended publications
  • Selective Targeting of Cyclin E1-Amplified High-Grade Serous Ovarian Cancer by Cyclin-Dependent Kinase 2 and AKT Inhibition
    Published OnlineFirst September 23, 2016; DOI: 10.1158/1078-0432.CCR-16-0620 Biology of Human Tumors Clinical Cancer Research Selective Targeting of Cyclin E1-Amplified High-Grade Serous Ovarian Cancer by Cyclin- Dependent Kinase 2 and AKT Inhibition George Au-Yeung1,2, Franziska Lang1, Walid J. Azar1, Chris Mitchell1, Kate E. Jarman3, Kurt Lackovic3,4, Diar Aziz5, Carleen Cullinane1,6, Richard B. Pearson1,2,7, Linda Mileshkin2,8, Danny Rischin2,8, Alison M. Karst9, Ronny Drapkin10, Dariush Etemadmoghadam1,2,5, and David D.L. Bowtell1,2,7,11 Abstract Purpose: Cyclin E1 (CCNE1) amplification is associated with Results: We validate CDK2 as a therapeutic target by demon- primary treatment resistance and poor outcome in high-grade strating selective sensitivity to gene suppression. However, we found serous ovarian cancer (HGSC). Here, we explore approaches to that dinaciclib did not trigger amplicon-dependent sensitivity in a target CCNE1-amplified cancers and potential strategies to over- panel of HGSC cell lines. A high-throughput compound screen come resistance to targeted agents. identified synergistic combinations in CCNE1-amplified HGSC, Experimental Design: To examine dependency on CDK2 in including dinaciclib and AKT inhibitors. Analysis of genomic data CCNE1-amplified HGSC, we utilized siRNA and conditional from TCGA demonstrated coamplification of CCNE1 and AKT2. shRNA gene suppression, and chemical inhibition using dina- Overexpression of Cyclin E1 and AKT isoforms, in addition to ciclib, a small-molecule CDK2 inhibitor. High-throughput mutant TP53, imparted malignant characteristics in untransformed compound screening was used to identify selective synergistic fallopian tube secretory cells, the dominant site of origin of HGSC.
    [Show full text]
  • A Study of Abemaciclib (LY2835219)
    A Study of Abemaciclib (LY2835219) in Combination With Temozolomide and Irinotecan and Abemaciclib in Combination With Temozolomide in Children and Young Adult Participants With Solid Tumors Status: Not yet recruiting Eligibility Criteria Sex: All Age: up to 18 Years old This study is NOT accepting healthy volunteers Inclusion Criteria: • Body weight ≥10 kilograms and body surface area (BSA) ≥0.5 meters squared. • Participants with any relapsed/refractory malignant solid tumor (excluding lymphoma), including central nervous system tumors, that have progressed on standard therapies and, in the judgment of the investigator, are appropriate candidates for the experimental therapy combination in the study part that is currently enrolling. • Participants must have at least one measurable (per Response Criteria in Solid Tumors [RECIST v1.1; [Eisenhauer et al. 2009] or Response Assessment in Neuro-Oncology (RANO) for central nervous system (CNS) tumors [Wen et al. 2010]) or evaluable lesion. • Participants must have had histologic verification of malignancy at original diagnosis or relapse, except: • Participants with extra-cranial germ-cell tumors who have elevations of serum tumor markers including alpha-fetoprotein or beta- human chorionic gonadotropin (HCG). • Participants with intrinsic brain stem tumors or participants with CNS-germ cell tumors and elevations of CSF or serum tumor markers including alpha-fetoprotein or beta-HCG. • A Lansky score ≥50 for participants ≤16 years of age or Karnofsky score ≥50 for participants >16 years of age. • Participants must have discontinued all previous treatments for cancer or investigational agents and must have recovered from the acute effects to Grade ≤1 at the time of enrollment.
    [Show full text]
  • Cancer Drug Pharmacology Table
    CANCER DRUG PHARMACOLOGY TABLE Cytotoxic Chemotherapy Drugs are classified according to the BC Cancer Drug Manual Monographs, unless otherwise specified (see asterisks). Subclassifications are in brackets where applicable. Alkylating Agents have reactive groups (usually alkyl) that attach to Antimetabolites are structural analogues of naturally occurring molecules DNA or RNA, leading to interruption in synthesis of DNA, RNA, or required for DNA and RNA synthesis. When substituted for the natural body proteins. substances, they disrupt DNA and RNA synthesis. bendamustine (nitrogen mustard) azacitidine (pyrimidine analogue) busulfan (alkyl sulfonate) capecitabine (pyrimidine analogue) carboplatin (platinum) cladribine (adenosine analogue) carmustine (nitrosurea) cytarabine (pyrimidine analogue) chlorambucil (nitrogen mustard) fludarabine (purine analogue) cisplatin (platinum) fluorouracil (pyrimidine analogue) cyclophosphamide (nitrogen mustard) gemcitabine (pyrimidine analogue) dacarbazine (triazine) mercaptopurine (purine analogue) estramustine (nitrogen mustard with 17-beta-estradiol) methotrexate (folate analogue) hydroxyurea pralatrexate (folate analogue) ifosfamide (nitrogen mustard) pemetrexed (folate analogue) lomustine (nitrosurea) pentostatin (purine analogue) mechlorethamine (nitrogen mustard) raltitrexed (folate analogue) melphalan (nitrogen mustard) thioguanine (purine analogue) oxaliplatin (platinum) trifluridine-tipiracil (pyrimidine analogue/thymidine phosphorylase procarbazine (triazine) inhibitor)
    [Show full text]
  • Efficacy and Safety of Abemaciclib, an Inhibitor of CDK4 and CDK6, for Patients with Breast Cancer, Non–Small Cell Lung Cancer, and Other Solid Tumors
    Published OnlineFirst May 23, 2016; DOI: 10.1158/2159-8290.CD-16-0095 RESEARCH ARTICLE Efficacy and Safety of Abemaciclib, an Inhibitor of CDK4 and CDK6, for Patients with Breast Cancer, Non–Small Cell Lung Cancer, and Other Solid Tumors Amita Patnaik1, Lee S. Rosen2, Sara M. Tolaney3, Anthony W. Tolcher1, Jonathan W. Goldman2, Leena Gandhi3, Kyriakos P. Papadopoulos1, Muralidhar Beeram1, Drew W. Rasco1, John F. Hilton3, Aejaz Nasir4, Richard P. Beckmann4, Andrew E. Schade4, Angie D. Fulford4, Tuan S. Nguyen4, Ricardo Martinez4, Palaniappan Kulanthaivel4, Lily Q. Li4, Martin Frenzel4, Damien M. Cronier4, Edward M. Chan4, Keith T. Flaherty5, Patrick Y. Wen3, and Geoffrey I. Shapiro3 ABSTRACT We evaluated the safety, pharmacokinetic profile, pharmacodynamic effects, and antitumor activity of abemaciclib, an orally bioavailable inhibitor of cyclin-dependent kinases (CDK) 4 and 6, in a multicenter study including phase I dose escalation followed by tumor- specific cohorts for breast cancer, non–small cell lung cancer (NSCLC), glioblastoma, melanoma, and colorectal cancer. A total of 225 patients were enrolled: 33 in dose escalation and 192 in tumor-specific cohorts. Dose-limiting toxicity was grade 3 fatigue. The maximum tolerated dose was 200 mg every 12 hours. The most common possibly related treatment-emergent adverse events involved fatigue and the gastrointestinal, renal, or hematopoietic systems. Plasma concentrations increased with dose, and pharmacodynamic effects were observed in proliferating keratinocytes and tumors. Radiographic responses were achieved in previously treated patients with breast cancer, NSCLC, and melanoma. For hormone receptor–positive breast cancer, the overall response rate was 31%; moreover, 61% of patients achieved either response or stable disease lasting ≥6 months.
    [Show full text]
  • The CDK4/6 Inhibitor LY2835219 Has Potent Activity in Combination with Mtor Inhibitor in Head and Neck Squamous Cell Carcinoma
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 12 The CDK4/6 inhibitor LY2835219 has potent activity in combination with mTOR inhibitor in head and neck squamous cell carcinoma Bo Mi Ku1,*, Seong Yoon Yi3,*, Jiae Koh1, Yeon-Hee Bae1, Jong-Mu Sun2, Se-hoon Lee2, Jin Seok Ahn2, Keunchil Park2, Myung-Ju Ahn2 1 Samsung Biomedical Research Institute, Seoul, Korea 2 Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea 3 Division of Hematology-Oncology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, Gyeonggi-do, Korea *These authors contributed equally to this work Correspondence to: Myung-Ju Ahn, e-mail: silk.ahn@samsung.com Keywords: head and neck cancer, CDK4/6 inhibitor, mTOR, cell cycle, targeted therapy Received: September 15, 2015 Accepted: January 23, 2016 Published: February 21, 2016 ABSTRACT Deletion of CDKN2A (p16) or amplification ofCCND1 (cyclin D1) occurs commonly in head and neck squamous cell carcinoma (HNSCC) and induces sustained cyclin- dependent kinase (CDK) 4/6 activation. Here, we report the antiproliferative activity of LY2835219, a selective CDK4/6 inhibitor through inhibition of CDK4/6-dependent Ser780 phosphorylation in retinoblastoma (RB) and induction of cell cycle arrest in HNSCC cells. In addition, we demonstrated the antitumor effects of HNSCC xenografts to LY2835219 in vivo. Given the limited effect in HNSCC as a single-agent treatment with LY2835219, a combinational strategy is required to enhance antitumor activity. At the molecular level, we found that LY2835219 inhibited activation of AKT and ERK, but not mTOR. The combination of LY2835219 with mTOR inhibitor was found to be more effective than either drug alone in vitro and in vivo.
    [Show full text]
  • Identification of Candidate Repurposable Drugs to Combat COVID-19 Using a Signature-Based Approach
    www.nature.com/scientificreports OPEN Identifcation of candidate repurposable drugs to combat COVID‑19 using a signature‑based approach Sinead M. O’Donovan1,10, Ali Imami1,10, Hunter Eby1, Nicholas D. Henkel1, Justin Fortune Creeden1, Sophie Asah1, Xiaolu Zhang1, Xiaojun Wu1, Rawan Alnafsah1, R. Travis Taylor2, James Reigle3,4, Alexander Thorman6, Behrouz Shamsaei4, Jarek Meller4,5,6,7,8 & Robert E. McCullumsmith1,9* The COVID‑19 pandemic caused by the novel SARS‑CoV‑2 is more contagious than other coronaviruses and has higher rates of mortality than infuenza. Identifcation of efective therapeutics is a crucial tool to treat those infected with SARS‑CoV‑2 and limit the spread of this novel disease globally. We deployed a bioinformatics workfow to identify candidate drugs for the treatment of COVID‑19. Using an “omics” repository, the Library of Integrated Network‑Based Cellular Signatures (LINCS), we simultaneously probed transcriptomic signatures of putative COVID‑19 drugs and publicly available SARS‑CoV‑2 infected cell lines to identify novel therapeutics. We identifed a shortlist of 20 candidate drugs: 8 are already under trial for the treatment of COVID‑19, the remaining 12 have antiviral properties and 6 have antiviral efcacy against coronaviruses specifcally, in vitro. All candidate drugs are either FDA approved or are under investigation. Our candidate drug fndings are discordant with (i.e., reverse) SARS‑CoV‑2 transcriptome signatures generated in vitro, and a subset are also identifed in transcriptome signatures generated from COVID‑19 patient samples, like the MEK inhibitor selumetinib. Overall, our fndings provide additional support for drugs that are already being explored as therapeutic agents for the treatment of COVID‑19 and identify promising novel targets that are worthy of further investigation.
    [Show full text]
  • A Novel CDK9 Inhibitor Increases the Efficacy of Venetoclax (ABT-199) in Multiple Models of Hematologic Malignancies
    Leukemia (2020) 34:1646–1657 https://doi.org/10.1038/s41375-019-0652-0 ARTICLE Molecular targets for therapy A novel CDK9 inhibitor increases the efficacy of venetoclax (ABT-199) in multiple models of hematologic malignancies 1 1 2,3 4 1 5 6 Darren C. Phillips ● Sha Jin ● Gareth P. Gregory ● Qi Zhang ● John Xue ● Xiaoxian Zhao ● Jun Chen ● 1 1 1 1 1 1 Yunsong Tong ● Haichao Zhang ● Morey Smith ● Stephen K. Tahir ● Rick F. Clark ● Thomas D. Penning ● 2,7 3 5 1 4 2,7 Jennifer R. Devlin ● Jake Shortt ● Eric D. Hsi ● Daniel H. Albert ● Marina Konopleva ● Ricky W. Johnstone ● 8 1 Joel D. Leverson ● Andrew J. Souers Received: 27 November 2018 / Revised: 18 October 2019 / Accepted: 13 November 2019 / Published online: 11 December 2019 © The Author(s) 2019 Abstract MCL-1 is one of the most frequently amplified genes in cancer, facilitating tumor initiation and maintenance and enabling resistance to anti-tumorigenic agents including the BCL-2 selective inhibitor venetoclax. The expression of MCL-1 is maintained via P-TEFb-mediated transcription, where the kinase CDK9 is a critical component. Consequently, we developed a series of potent small-molecule inhibitors of CDK9, exemplified by the orally active A-1592668, with CDK selectivity profiles 1234567890();,: 1234567890();,: that are distinct from related molecules that have been extensively studied clinically. Short-term treatment with A-1592668 rapidly downregulates RNA pol-II (Ser 2) phosphorylation resulting in the loss of MCL-1 protein and apoptosis in MCL-1- dependent hematologic tumor cell lines. This cell death could be attenuated by either inhibiting caspases or overexpressing BCL-2 protein.
    [Show full text]
  • Cyclacel's CYC065 CDK Inhibitor Demonstrates Synergy With
    Cyclacel’s CYC065 CDK Inhibitor Demonstrates Synergy With Venetoclax By Dual Targeting Of Chronic Lymphocytic Leukemia April 17, 2018 Suppression of both BCl-2 and Mcl-1 anti-apoptotic proteins is a novel strategy in CLL BERKELEY HEIGHTS, N.J., April 17, 2018 (GLOBE NEWSWIRE) -- Cyclacel Pharmaceuticals, Inc. (NASDAQ:CYCC) (NASDAQ:CYCCP) ("Cyclacel" or the "Company"), a biopharmaceutical company developing oral therapies that target various phases of cell cycle control for the treatment of cancer and other serious disorders, today announced the presentation by investigators led by William Plunkett, PhD, Professor and Deputy Chair, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, of preclinical data demonstrating strong synergy between Cyclacel’s CDK2/9 inhibitor, CYC065, and the Bcl-2 inhibitor, venetoclax (ABT-199, AbbVie) in chronic lymphocytic leukemia (CLL) samples obtained from patients. The data were presented at the American Association for Cancer Research (AACR) Annual Meeting being held April 14-18, 2018 in Chicago, Illinois. “The MD Anderson data show that the combination of CYC065 and venetoclax is strongly synergistic in primary CLL cells from patients, including those with 17p deletions. In addition, the combination was active in two CLL samples which were resistant to either agent alone. These findings support the hypothesis that dual targeting of the Mcl-1- and Bcl-2-dependent mechanisms could induce synergistic cell death by apoptosis,” said Spiro Rombotis, President and Chief Executive Officer of Cyclacel. “Last weekend during the same AACR conference, we reported that CYC065 durably suppresses Mcl-1, a member of the Bcl-2 family of survival proteins, in patients with advanced solid tumors1.
    [Show full text]
  • Pharmacologic Properties of AG-012986, a Pan-Cyclin- Dependent Kinase Inhibitor with Antitumor Efficacy
    818 Pharmacologic properties of AG-012986, a pan-cyclin- dependent kinase inhibitor with antitumor efficacy Cathy Zhang,1 Karen Lundgren,1 Zhengming Yan,1 optimization of AG-012986 provided guidance for select- Maria E. Arango,1 Sharon Price,1 Andrea Huber,1 ing a treatment schedule to achieve the best antitumor Joseph Higgins,1 Gabriel Troche,1 efficacy while minimizing the risk of adverse side effects. Judith Skaptason,2 Tatiana Koudriakova,2 [Mol Cancer Ther 2008;7(4):818–28] Jim Nonomiya,4 Michelle Yang,5 1 1 1 Patrick O’Connor, Steve Bender, Gerrit Los, Introduction Cristina Lewis,4 and Bart Jessen3 Cyclin-dependent kinases (CDK) and their regulatory Departments of 1Cancer Biology, 2Pharmacokinetics, Dynamics cyclin partners play critical roles in cell cycle control and and Metabolism, 3Drug Safety Research and Development, the regulation of cell transcription. Progression through 4 5 Biochemical Pharmacology, and Medicinal Chemistry, the different stages of cell cycle is governed by the Pfizer Global Research and Development, La Jolla, California activities of the CDK1, CDK2, CDK4, CDK6, and possibly CDK3. CDK4/cyclin D, CDK6/cyclin D, and CDK2/cyclin E Abstract phosphorylate the retinoblastoma (Rb) protein at multiple AG-012986 is a multitargeted cyclin-dependent kinase sites, which results in activation of the E2F family of (CDK) inhibitor active against CDK1, CDK2, CDK4/6, transcription factors and serves as a trigger for cells to CDK5, and CDK9, with selectivity over a diverse panel of advance beyond the G1 checkpoint into S phase (1–3). non-CDK kinases. Here, we report the potent antitumor During S phase, CDK2/cyclin A phosphorylates several efficacies of AG-012986 against multiple tumor lines proteins, including E2F, to regulate progression through in vitro and in vivo.
    [Show full text]
  • Anticancer and Radiosensitizing Effects of the Cyclin-Dependent Kinase Inhibitors, AT7519 and SNS‑032, on Cervical Cancer
    INTERNATIONAL JOURNAL OF ONCOLOGY 53: 703-712, 2018 Anticancer and radiosensitizing effects of the cyclin-dependent kinase inhibitors, AT7519 and SNS-032, on cervical cancer MI AE KANG1*, WONWOO KIM2*, HYE-RAM JO1,3, YOUNG-JOO SHIN4, MOON-HONG KIM5 and JAE-HOON JEONG1,3 1Division of Applied Radiation Bioscience, and 2Radiation Non-Clinic Center, Korea Institute of Radiological and Medical Science, Seoul 01812; 3Radiological and Medico-Oncological Sciences, Korea University of Science and Technology, Daejeon 34113; 4Department of Radiation Oncology, Inje University Sanggye Paik Hospital, Seoul 01757; 5Department of Obstetrics and Gynecology, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea Received January 30, 2018; Accepted May 17, 2018 DOI: 10.3892/ijo.2018.4424 Abstract. Cyclin-dependent kinases (CDK) are considered the utilization of AT7519 and SNS-032 as part of an adjuvant to be potential targets of anticancer drugs that can interrupt treatment may help control cervical cancer progression. the uncontrolled division of cancer cells. In this study, we selected two selective CDK inhibitors, AT7519 and SNS-032, Introduction from current clinical trials and examined their anticancer and radiosensitizing effects in a cervical cancer model. SNS-032 Cyclin-dependent kinases (CDKs) are present in all known was found to be more potent than AT7519, with a lower half eukaryotes, and their regulatory functions during the cell cycle maximal inhibitory concentration (IC50) value. Both AT7519 are evolutionarily conserved. Cyclin-CDK complexes phos- and SNS-032 induced the apoptosis, premature senescence and phorylate specific substrates, according to the requirements cytostasis of cervical cancer cells, which led to the attenuation of a particular cell cycle phase.
    [Show full text]
  • Journal Pre-Proof
    Journal Pre-proof 5th ESO-ESMO international consensus guidelines for advanced breast cancer (ABC † 5) F. Cardoso, S. Paluch-Shimon, E. Senkus, G. Curigliano, M.S. Aapro, F. André, C.H. Barrios, J. Bergh, G.S. Bhattacharyya, L. Biganzoli, F. Boyle, M.-J. Cardoso, L.A. Carey, J. Cortés, N.S. El Saghir, M. Elzayat, A. Eniu, L. Fallowfield, P.A. Francis, K. Gelmon, J. Gligorov, R. Haidinger, N. Harbeck, X. Hu, B. Kaufman, R. Kaur, B.E. Kiely, S.-B. Kim, N.U. Lin, S.A. Mertz, S. Neciosup, B.V. Offersen, S. Ohno, O. Pagani, A. Prat, F. Penault-Llorca, H.S. Rugo, G.W. Sledge, C. Thomssen, D.A. Vorobiof, T. Wiseman, B. Xu, L. Norton, A. Costa, E.P. Winer PII: S0923-7534(20)42460-3 DOI: https://doi.org/10.1016/j.annonc.2020.09.010 Reference: ANNONC 337 To appear in: Annals of Oncology Received Date: 21 August 2020 Revised Date: 15 September 2020 Accepted Date: 16 September 2020 Please cite this article as: Cardoso F, Paluch-Shimon S, Senkus E, Curigliano G, Aapro MS, André F, Barrios CH, Bergh J, Bhattacharyya GS, Biganzoli L, Boyle F, Cardoso MJ, Carey LA, Cortés J, El Saghir NS, Elzayat M, Eniu A, Fallowfield L, Francis PA, Gelmon K, Gligorov J, Haidinger R, Harbeck N, Hu X, Kaufman B, Kaur R, Kiely BE, Kim SB, Lin NU, Mertz SA, Neciosup S, Offersen BV, Ohno S, Pagani O, Prat A, Penault-Llorca F, Rugo HS, Sledge GW, Thomssen C, Vorobiof DA, Wiseman T, Xu B, Norton L, Costa A, Winer EP, 5th ESO-ESMO international consensus guidelines for advanced breast † cancer (ABC 5) , Annals of Oncology (2020), doi: https://doi.org/10.1016/j.annonc.2020.09.010.
    [Show full text]
  • New Drug Update: the Next Step in Personalized Medicine
    New Drug Update: The Next Step in Personalized Medicine Jordan Hill, PharmD, BCOP Clinical Pharmacy Specialist WVU Medicine Mary Babb Randolph Cancer Center Objectives • Review indications for new FDA approved anti- neoplastic medications in 2017 • Outline place in therapy of new medications • Become familiar with mechanisms of action of new medications • Describe adverse effects associated with new medications • Summarize dosing schemes and appropriate dose reductions for new medications First-in-Class Approvals • FLT3 inhibitor – midostaurin • IDH2 inhibitor – enasidenib • Anti-CD22 antibody drug conjugate – inotuzumab ozogamicin • CAR T-cell therapy - tisagenlecleucel “Me-too” Approvals • CDK 4/6 inhibitor – ribociclib and abemaciclib • PD-L1 inhibitors • Avelumab • Durvalumab • PARP inhibitor – niraparib • ALK inhibitor – brigatinib • Pan-HER inhibitor – neratinib • Liposome-encapsulated combination of daunorubicin and cytarabine • PI3K inhibitor – copanlisib Drugs by Malignancy AML Breast Bladder ALL FL Lung Ovarian 0 1 2 3 Oral versus IV 46% 54% Oral IV FIRST-IN CLASS FLT3 Inhibitor - midostaurin Journal of Hematology & Oncology 2017;10:93 Midostaurin (Rydapt®) • 717 newly diagnosed FLT3+ AML Patients • Induction and consolidation chemotherapy and placebo maintenance versus chemotherapy + midostaurin and Treatment maintenance midostaurin • OS: 74.7 mo versus 25.6 mo (P=0.009) Efficacy • EFS: 8.2 mo versus 3.0 mo (P=0.002) • Higher grade > 3 anemia, rash, and nausea Safety N Engl J Med 2017;377:454-64 Midostaurin (Rydapt®) • Approved indications: FLT3+ AML, mast cell leukemia, systemic mastocytosis • AML dose: 50 mg twice daily with food on days 8-21 • Of each induction cycle (+ daunorubicin and cytarabine) • Of each consolidation cycle (+ high dose cytarabine) • ADEs: nausea, myelosuppression, mucositis, increases in LFTs, amylase/lipase, and electrolyte abnormalities • Pharmacokinetics: hepatic metabolism, substrate of CYP 3A4; < 5% excretion in urine RYDAPT (midostaurin) [package insert].
    [Show full text]