Intravesical Instillation of Azacitidine Suppresses Tumor Formation Through TNF-R1 and TRAIL-R2 Signaling in Genotoxic Carcinogen-Induced Bladder Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Intravesical Instillation of Azacitidine Suppresses Tumor Formation Through TNF-R1 and TRAIL-R2 Signaling in Genotoxic Carcinogen-Induced Bladder Cancer cancers Article Intravesical Instillation of Azacitidine Suppresses Tumor Formation through TNF-R1 and TRAIL-R2 Signaling in Genotoxic Carcinogen-Induced Bladder Cancer Shao-Chuan Wang 1,2,3, Ya-Chuan Chang 2, Min-You Wu 2, Chia-Ying Yu 2, Sung-Lang Chen 1,2,3 and Wen-Wei Sung 1,2,3,* 1 Department of Urology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan; [email protected] (S.-C.W.); [email protected] (S.-L.C.) 2 School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; [email protected] (Y.-C.C.); [email protected] (M.-Y.W.); [email protected] (C.-Y.Y.) 3 Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan * Correspondence: [email protected] or fl[email protected]; Tel.: +886-4-2473-9595 Simple Summary: Approximately 70% of all bladder cancer is diagnosed as non-muscle invasive bladder cancer and can be treated by transurethral resection of the bladder tumor, followed by intravesical instillation chemotherapy. Bacille Calmette-Guérin (BCG) is the first-line agent for intravesical instillation, but its accessibility has been limited for years due to a BCG shortage. Here, our aim was to evaluate the therapeutic role of intravesical instillation of azacitidine, a DNA methyltransferase inhibitor, in bladder cancer. Cell model experiments showed that azacitidine inhibited TNFR1 downstream pathways to downregulate HIF-1α, claspin, and survivin. Concomitant Citation: Wang, S.-C.; Chang, Y.-C.; upregulation of the TRAIL R2 pathway by azacitidine ultimately drove the tumor cells to apoptosis. Wu, M.-Y.; Yu, C.-Y.; Chen, S.-L.; Sung, Rats with genotoxic carcinogen-induced bladder cancer showed a significantly reduced in vivo tumor W.-W. Intravesical Instillation of burden after intravesical instillation of azacitidine. These findings might support further clinical Azacitidine Suppresses Tumor trials of azacitidine in bladder cancer. Formation through TNF-R1 and TRAIL-R2 Signaling in Genotoxic Abstract: Azacitidine, an inhibitor of DNA methylation, shows therapeutic effects against several Carcinogen-Induced Bladder Cancer. malignancies by inducing apoptosis and inhibiting tumor cell proliferation. However, the anti- Cancers 2021, 13, 3933. https:// tumor effects of azacitidine on urinary bladder urothelial carcinoma (UBUC), especially following doi.org/10.3390/cancers13163933 intravesical instillation (IVI), are not established. Here, UBUC cell lines were used to analyze the Academic Editor: Andrea Morrione in vitro therapeutic effects of azacitidine. Potential signaling pathways were investigated by antibody arrays and Western blotting. The N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced rat UBUC Received: 18 June 2021 model was used for in vivo quantitative analysis of tumor burden. Azacitidine significantly inhibited Accepted: 2 August 2021 DNMT expression in UBUC cell lines and reduced cell viability and clonogenic activity, as determined Published: 4 August 2021 by MTT and colony formation assays, while also inducing significant cytotoxic effects in the form of increased sub-G1 and Annexin V-PI populations (all p < 0.05). Antibody arrays confirmed the in vitro Publisher’s Note: MDPI stays neutral suppression of TNF-R1 and the induction of TRAIL-R2 and their downstream signaling molecules. with regard to jurisdictional claims in TNF-R1 suppression reduced claspin and survivin expression, while TRAIL-R2 activation induced published maps and institutional affil- cytochrome C and caspase 3 expression. Rats with BBN-induced bladder cancer had a significantly iations. reduced tumor burden and Ki67 index following IVI of azacitidine (p < 0.01). Our study provides evidence for a reduction in BBN-induced bladder cancer by IVI of azacitidine through alterations in the TRAIL-R2 and TNF-R1 signaling pathways. These findings might provide new insights for further clinical trials. Copyright: © 2021 by the authors. Licensee MDPI, Basel, Switzerland. Keywords: bladder cancer; azacitidine; intravesical instillation; TNF-R1; TRAIL-R2 This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https:// creativecommons.org/licenses/by/ 4.0/). Cancers 2021, 13, 3933. https://doi.org/10.3390/cancers13163933 https://www.mdpi.com/journal/cancers Cancers 2021, 13, 3933 2 of 13 1. Introduction Urinary bladder urothelial carcinoma (UBUC), also known as bladder cancer, is a highly prevalent malignancy worldwide. In 2020, the estimated new cases and deaths were 81,400 and 17,980, respectively, in the United States [1]. Non-muscle invasive bladder cancer (NMIBC) accounts for 70% of all bladder cancer diagnoses [2], and the management of NMIBC consists mostly of tumor excision via transurethral resection of the bladder tumor (TURBT). Subsequent intravesical instillation (IVI) of chemotherapeutic agents is performed within 24 h after surgery to eliminate residual cancer cells [3]. IVI therapy directly instills chemotherapy or immunotherapy agents, such as mit- omycin C and Bacille Calmette-Guérin (BCG), into the bladder [2]. BCG is the first-line treatment for NMIBC, and it significantly reduces the recurrence of malignancy [4]. BCG has several disadvantages, such as side effects that include a localized inflammatory re- sponse and BCG infection; however, a shortage of BCG agent currently limits accessibility to this treatment [5,6]. In addition, the tumor recurrence rate remains at about 40% fol- lowing chemotherapy instillation [7], indicating that any reduction in the rate of UBUC recurrence will require the exploration of alternative agents or agent combinations for IVI bladder cancer therapy. The progression of many cancers involves epigenetic abnormalities, such as aberrant DNA or histone methylation, which result in disturbances of gene expression [8,9]. Epige- netic therapy, therefore, plays an essential role in cancer treatment, and the vast majority of epigenetic drugs are histone deacetylase inhibitors (HDACi) and DNA methyltrans- ferase inhibitors (DNMTi) [10]. Ongoing or completed clinical trials of epigenetic drugs for treating bladder malignancy have mainly involved HDACi treatments, with only about 20% investigating DNMTi efficacy [11]. Similarly, very few studies have explored the intravesical instillation of DNMTi as adjuvant therapy. One promising DNMTi inhibitor is azacitidine, a type of hypomethylating agent that incorporates into DNA and RNA. It also binds to DNMT1 as an irreversible inhibitor [12], thereby preventing DNA methylation and reactivating tumor suppressor genes [13]. Azaci- tidine has been used as therapy for myelodysplastic syndromes and it improves outcomes for elderly patients diagnosed with acute myeloid leukemia [14,15]. Azacitidine also has therapeutic effects on solid tumors [16]. Currently, most clinical trials have focused on azacitidine as therapy for leukemia, but a few studies have examined the potential of intravenous injection as therapy for urologic malignancies, such as castration-resistant prostate cancer [17]. Azacitidine promotes the expression of tumor suppressor genes and inhibits the pro- liferation of bladder cancer cell lines, suggesting a potential therapeutic role in UBUC [18]. Azacitidine can also increase the sensitivity of drug-resistant bladder urothelial carcinoma cells to chemotherapy agents [19]. We hypothesized that azacitidine might show an anti- tumor effect and reduce the tumor recurrence rate in UBUC. The aim of the present study was to examine the anti-tumor effects of azacitidine on UBUC cell lines and to conduct in vivo experiments to assess the feasibility of using IVI of azacitidine as a treatment for UBUC. 2. Materials and Methods 2.1. Cell Culture Two UBUC cell lines, T24 and UMUC3, were purchased from The American Type Culture Collection (Manassas, VA, USA) and stored according to the supplier’s instructions. Cells were grown in RPMI medium containing 10% fetal bovine serum, 100 U/mL penicillin, 100 µg/mL streptomycin, 2 g/mL NaHCO3, 1 mM sodium pyruvate, and 0.1 mM non- ◦ essential amino acids. All cell lines were cultured at 37 C and supplied with 5% CO2. 2.2. MTT Assays The MTT assay was performed to detect the cytotoxicity of azacitidine. Briefly,2 × 104 cells were seeded into 96-well plates, incubated overnight, and exposed to azacitidine (0, 1, and Cancers 2021, 13, 3933 3 of 13 10 µM) for 1 or 24 h and harvested on the 72nd hour. MTT solution (0.5 mg/mL) was added to the wells and incubated for 3 h at 37 ◦C. The reaction was ended by the removal of the supernatant, followed by dissolving the formazan product in DMSO. Optical density was measured with an ELISA reader using a 570 nm filter. Each experiment was performed in triplicate and the cell viability of each group was calculated and analyzed compared to the control. 2.3. Clonogenic Assay T24 (250 cells) and UMUC3 (500 cells) treated with azacitidine (0, 1, and 10 µM) were seeded into each well of a 6-well plate and incubated for 3 or 10 days. The resulting colonies were fixed with 95% ethanol for 20 min and stained with 20% Giemsa solution for 30 min at room temperature, as previously described [20]. Each experiment was performed in triplicate and colony counts were calculated for each group. 2.4. Flow Cytometry Analysis Flow cytometry was conducted with a FACSCanto™ II Cell Analyzer (BD Biosciences, Franklin Lakes, NJ, USA) to determine the cell cycle distribution and percentage of apop- totic cells in each group, as previously described [21]. T24 and UMUC3 cells were seeded in 6-well plates and treated with azacitidine (0, 1, and 10 µM) for 72 h. For cell cycle distribution analysis, cells were fixed in pre-chilled 70% ethanol. The fixed cells were then re-suspended in PBS containing 0.4 µg/mL PI and 0.5 mg/mL RNase, and analyzed by flow cytometry. Apoptosis was analyzed using an Annexin V-FITC apoptosis detection kit (Strong Biotech Corporation, Taipei, Taiwan) according to the recommended protocol. Cells were re-suspended in binding buffer (100 µL), and 2 µL Annexin V-FITC and 2 µL PI were added. After a 15 min reaction in the dark, the cell apoptosis rate was evaluated by flow cytometry.
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]
  • LEUKEMIA CHEMOTHERAPY REGIMENS (Part 1 of 2) the Selection, Dosing, and Administration of Anti-Cancer Agents and the Management of Associated Toxicities Are Complex
    LEUKEMIA CHEMOTHERAPY REGIMENS (Part 1 of 2) The selection, dosing, and administration of anti-cancer agents and the management of associated toxicities are complex. Drug dose modifications and schedule and initiation of supportive care interventions are often necessary because of expected toxicities and because of individual patient variability, prior treatment, and comorbidities. Thus, the optimal delivery of anti-cancer agents requires a healthcare delivery team experienced in the use of such agents and the management of associated toxicities in patients with cancer. The chemotherapy regimens below may include both FDA-approved and unapproved uses/regimens and are provided as references only to the latest treatment strategies. Clinicians must choose and verify treatment options based on the individual patient. REGIMEN DOSING Acute Myeloid Leukemia (AML) Induction Therapy Cytarabine (Cytosar-U; ARA-C) + Days 1–3: An anthracycline (eg, daunorubicin at least 60mg/m2/day IV, an anthracycline idarubicin 10–12mg/m2/day IV, or mitoxantrone 10–12mg/m2/day IV), plus (daunorubicin [Cerubidine], Days 1–7: Cytarabine 100–200mg/m2/day continuous IV infusion. idarubicin [Idamycin], OR mitoxantrone [Novantrone])1, 2 Days 1–3: An anthracycline (eg, daunorubicin 45mg/m2/day IV, idarubicin 12mg/m2/day IV, or mitoxantrone 12mg/m2/day IV), plus Days 1–7: Cytarabine 100mg/m2/day continuous IV infusion. Intermediate-dose cytarabine3 Cycle 1 Days 1–7: Cytarabine 200mg/m2/day continuous IV infusion, plus Days 5–6: Idarubicin 12mg/m2/day IV. Cycle 2 Days 1–6: Cytarabine 1,000mg/m2 continuous IV infusion for 3 hrs twice daily, plus Days 3, 5 and 7: Amsacrine 120mg/m2/day.
    [Show full text]
  • Vidaza, INN-Azacitidine
    ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS 1 1. NAME OF THE MEDICINAL PRODUCT Vidaza 25 mg/ml powder for suspension for injection 2. QUALITATIVE AND QUANTITATIVE COMPOSITION Each vial contains 100 mg azacitidine. After reconstitution, each ml suspension contains 25 mg azacitidine. For a full list of excipients, see section 6.1. 3. PHARMACEUTICAL FORM Powder for suspension for injection. White lyophilised powder. 4. CLINICAL PARTICULARS 4.1 Therapeutic indications Vidaza is indicated for the treatment of adult patients who are not eligible for haematopoietic stem cell transplantation with: • intermediate-2 and high-risk myelodysplastic syndromes (MDS) according to the International Prognostic Scoring System (IPSS), • chronic myelomonocytic leukaemia (CMML) with 10-29 % marrow blasts without myeloproliferative disorder, • acute myeloid leukaemia (AML) with 20-30 % blasts and multi-lineage dysplasia, according to World Health Organisation (WHO) classification. 4.2 Posology and method of administration Vidaza treatment should be initiated and monitored under the supervision of a physician experienced in the use of chemotherapeutic agents. Patients should be premedicated with anti-emetics for nausea and vomiting. Posology The recommended starting dose for the first treatment cycle, for all patients regardless of baseline haematology laboratory values, is 75 mg/m2 of body surface area, injected subcutaneously, daily for 7 days, followed by a rest period of 21 days (28-day treatment cycle). It is recommended that patients be treated for a minimum of 6 cycles. Treatment should be continued as long as the patient continues to benefit or until disease progression. Patients should be monitored for haematologic response/toxicity and renal toxicities (see section 4.4); a delay in starting the next cycle or a dose reduction as described below may be necessary.
    [Show full text]
  • Azacytidine Sensitizes Acute Myeloid Leukemia Cells to Arsenic Trioxide By
    Chau et al. Journal of Hematology & Oncology (2015) 8:46 DOI 10.1186/s13045-015-0143-3 JOURNAL OF HEMATOLOGY & ONCOLOGY RESEARCH ARTICLE Open Access Azacytidine sensitizes acute myeloid leukemia cells to arsenic trioxide by up-regulating the arsenic transporter aquaglyceroporin 9 David Chau1†, Karen Ng1†, Thomas Sau-Yan Chan1, Yuen-Yee Cheng1,3, Bonnie Fong2, Sidney Tam2, Yok-Lam Kwong1 and Eric Tse1* Abstract Background: The therapeutic efficacy of arsenic trioxide (As2O3) in acute myeloid leukemia (AML) is modest, which is partly related to its limited intracellular uptake into the leukemic cells. As2O3 enters cells via the transmembrane protein aquaglyceroporin 9 (AQP9). Azacytidine, a demethylating agent that is approved for the treatment of AML, has been shown to have synergistic effect with As2O3. We tested the hypothesis that azacytidine might up-regulate AQP9 and enhances As2O3-mediated cytotoxicity in AML. Methods: Arsenic-induced cytotoxicity, the expression of AQP9, and the intracellular uptake of As2O3 were determined in AML cell lines and primary AML cells with or without azacytidine pre-treatment. The mechanism of AQP9 up-regulation was then investigated by examining the expression of transcription factors for AQP9 gene and the methylation status of their gene promoters. Results: As2O3-induced cytotoxicity in AML cell lines was significantly enhanced after azacytidine pre-treatment as a result of AQP9 up-regulation, leading to increased arsenic uptake and hence intracellular concentration. Blocking AQP9-mediated As2O3 uptake with mercury chloride abrogated the sensitization effect of azacytidine. AQP9 promoter does not contain CpG islands. Instead, azacytidine pre-treatment led to increased expression of HNF1A, a transcription activator of AQP9, through demethylation of HNF1A promoter.
    [Show full text]
  • Pharmacogenomic Biomarkers in Docetaxel Treatment of Prostate Cancer: from Discovery to Implementation
    G C A T T A C G G C A T genes Review Pharmacogenomic Biomarkers in Docetaxel Treatment of Prostate Cancer: From Discovery to Implementation Reka Varnai 1,2, Leena M. Koskinen 3, Laura E. Mäntylä 3, Istvan Szabo 4,5, Liesel M. FitzGerald 6 and Csilla Sipeky 3,* 1 Department of Primary Health Care, University of Pécs, Rákóczi u 2, H-7623 Pécs, Hungary 2 Faculty of Health Sciences, Doctoral School of Health Sciences, University of Pécs, Vörösmarty u 4, H-7621 Pécs, Hungary 3 Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland 4 Institute of Sport Sciences and Physical Education, University of Pécs, Ifjúság útja 6, H-7624 Pécs, Hungary 5 Faculty of Sciences, Doctoral School of Biology and Sportbiology, University of Pécs, Ifjúság útja 6, H-7624 Pécs, Hungary 6 Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia * Correspondence: csilla.sipeky@utu.fi Received: 17 June 2019; Accepted: 5 August 2019; Published: 8 August 2019 Abstract: Prostate cancer is the fifth leading cause of male cancer death worldwide. Although docetaxel chemotherapy has been used for more than fifteen years to treat metastatic castration resistant prostate cancer, the high inter-individual variability of treatment efficacy and toxicity is still not well understood. Since prostate cancer has a high heritability, inherited biomarkers of the genomic signature may be appropriate tools to guide treatment. In this review, we provide an extensive overview and discuss the current state of the art of pharmacogenomic biomarkers modulating docetaxel treatment of prostate cancer. This includes (1) research studies with a focus on germline genomic biomarkers, (2) clinical trials including a range of genetic signatures, and (3) their implementation in treatment guidelines.
    [Show full text]
  • Vidaza U.S. Full Prescribing Information
    -------------------------------CONTRAINDICATIONS------------------------------ HIGHLIGHTS OF PRESCRIBING INFORMATION Advanced Malignant Hepatic Tumors (4.1). These highlights do not include all the information needed to use Hypersensitivity to Azacitidine or Mannitol (4.2). VIDAZA safely and effectively. See full prescribing information for VIDAZA. -----------------------WARNINGS AND PRECAUTIONS------------------------ Anemia, Neutropenia and Thrombocytopenia: Monitor complete blood VIDAZA (azacitidine for injection), for subcutaneous or intravenous use counts (CBC) frequently (5.1). Initial U.S. Approval: 2004 Hepatotoxicity: Patients with severe preexisting hepatic impairment are at higher risk for toxicity (5.2). Renal Toxicity: Monitor patients with renal impairment for toxicity ----------------------------INDICATIONS AND USAGE--------------------------- since azacitidine and its metabolites are primarily excreted by the VIDAZA is a nucleoside metabolic inhibitor indicated for the treatment of kidneys (5.3). patients with the following FAB myelodysplastic syndrome (MDS) subtypes: Tumor Lysis Syndrome: VIDAZA may cause fatal or serious tumor Refractory anemia (RA) or refractory anemia with ringed sideroblasts (RARS) lysis syndrome, including in patients with MDS. Assess baseline risk (if accompanied by neutropenia or thrombocytopenia or requiring and monitor and treat as appropriate (5.4). transfusions), refractory anemia with excess blasts (RAEB), refractory anemia Embryo-Fetal Risk: VIDAZA can cause fetal harm. Advise females
    [Show full text]
  • Current Treatment of Juvenile Myelomonocytic Leukemia
    Journal of Clinical Medicine Review Current Treatment of Juvenile Myelomonocytic Leukemia Christina Mayerhofer 1 , Charlotte M. Niemeyer 1,2 and Christian Flotho 1,2,* 1 Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; [email protected] (C.M.); [email protected] (C.M.N.) 2 German Cancer Consortium (DKTK), 79106 Freiburg, Germany * Correspondence: christian.fl[email protected] Abstract: Juvenile myelomonocytic leukemia (JMML) is a rare pediatric leukemia characterized by mutations in five canonical RAS pathway genes. The diagnosis is made by typical clinical and hematological findings associated with a compatible mutation. Although this is sufficient for clinical decision-making in most JMML cases, more in-depth analysis can include DNA methylation class and panel sequencing analysis for secondary mutations. NRAS-initiated JMML is heterogeneous and adequate management ranges from watchful waiting to allogeneic hematopoietic stem cell transplan- tation (HSCT). Upfront azacitidine in KRAS patients can achieve long-term remissions without HSCT; if HSCT is required, a less toxic preparative regimen is recommended. Germline CBL patients often experience spontaneous resolution of the leukemia or exhibit stable mixed chimerism after HSCT. JMML driven by PTPN11 or NF1 is often rapidly progressive, requires swift HSCT and may benefit from pretransplant therapy with azacitidine. Because graft-versus-leukemia alloimmunity is central to cure high risk patients, the immunosuppressive regimen should be discontinued early after HSCT. Keywords: juvenile myelomonocytic leukemia; RAS signaling; hematopoietic stem cell transplantation; 5-azacitidine; myelodysplastic/myeloproliferative disorders; targeted therapy Citation: Mayerhofer, C.; Niemeyer, C.M.; Flotho, C.
    [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]
  • Sorafenib and Omacetaxine Mepesuccinate As a Safe and Effective Treatment for Acute Myeloid Leukemia Carrying Internal Tandem Du
    Original Article Sorafenib and Omacetaxine Mepesuccinate as a Safe and Effective Treatment for Acute Myeloid Leukemia Carrying Internal Tandem Duplication of Fms-Like Tyrosine Kinase 3 Chunxiao Zhang, MSc1; Stephen S. Y. Lam, MBBS, PhD1; Garret M. K. Leung, MBBS1; Sze-Pui Tsui, MSc2; Ning Yang, PhD1; Nelson K. L. Ng, PhD1; Ho-Wan Ip, MBBS2; Chun-Hang Au, PhD3; Tsun-Leung Chan, PhD3; Edmond S. K. Ma, MBBS3; Sze-Fai Yip, MBBS4; Harold K. K. Lee, MBChB5; June S. M. Lau, MBChB6; Tsan-Hei Luk, MBChB6; Wa Li, MBChB7; Yok-Lam Kwong, MD 1; and Anskar Y. H. Leung, MD, PhD 1 BACKGROUND: Omacetaxine mepesuccinate (OME) has antileukemic effects against acute myeloid leukemia (AML) carrying an internal tandem duplication of Fms-like tyrosine kinase 3 (FLT3-ITD). A phase 2 clinical trial was conducted to evaluate a combina- tion treatment of sorafenib and omacetaxine mepesuccinate (SOME). METHODS: Relapsed or refractory (R/R) or newly diagnosed patients were treated with sorafenib (200-400 mg twice daily) and OME (2 mg daily) for 7 (first course) or 5 days (second course on- ward) every 21 days until disease progression or allogeneic hematopoietic stem cell transplantation (HSCT). The primary endpoint was composite complete remission, which was defined as complete remission (CR) plus complete remission with incomplete hematologic recovery (CRi). Secondary endpoints were leukemia-free survival (LFS) and overall survival (OS). RESULTS: Thirty-nine R/R patients and 5 newly diagnosed patients were recruited. Among the R/R patients, 28 achieved CR or CRi. Two patients showed partial remission, and 9 patients did not respond.
    [Show full text]
  • Treating Acute Myeloid Leukemia (AML) If You've Been Diagnosed with Acute Myeloid Leukemia (AML), Your Cancer Care Team Will Discuss Your Treatment Options with You
    cancer.org | 1.800.227.2345 Treating Acute Myeloid Leukemia (AML) If you've been diagnosed with acute myeloid leukemia (AML), your cancer care team will discuss your treatment options with you. Your options may be affected by the AML subtype, as well as certain other prognostic factors, as well as your age and overall state of health. How is acute myeloid leukemia treated? The main treatment for most types of AML is chemotherapy, sometimes along with a targeted therapy drug. This might be followed by a stem cell transplant. Other drugs (besides standard chemotherapy drugs) may be used to treat people with acute promyelocytic leukemia (APL). Surgery and radiation therapy are not major treatments for AML, but they may be used in special circumstances. ● Chemotherapy for Acute Myeloid Leukemia (AML) ● Targeted Therapy Drugs for Acute Myeloid Leukemia (AML) ● Non-Chemo Drugs for Acute Promyelocytic Leukemia (APL) ● Surgery for Acute Myeloid Leukemia (AML) ● Radiation Therapy for Acute Myeloid Leukemia (AML) ● Stem Cell Transplant for Acute Myeloid Leukemia (AML) Common treatment approaches The typical treatment approach for AML is different from the treatment approach for acute promyelocytic leukemia (APL). The response rates for treatment can vary based on the subtype of AML, as well as other factors. Treatment options might be different if the AML doesn't respond to the initial treatment or if it comes back later on. The treatment approach for children with AML can be slightly different from that used for adults. It's discussed separately in Treatment of Children With Acute Myeloid Leukemia 1 ____________________________________________________________________________________American Cancer Society cancer.org | 1.800.227.2345 (AML).
    [Show full text]
  • Antiproliferative and Antitumor Effects of Azacitidine Against the Human Myelodysplastic Syndrome Cell Line SKM-1
    ANTICANCER RESEARCH 32: 795-798 (2012) Antiproliferative and Antitumor Effects of Azacitidine Against the Human Myelodysplastic Syndrome Cell Line SKM-1 SACHIE KIMURA, KAZUYA KURAMOTO, JUNKO HOMAN, HARUNA NARUOKA, TAKESHI EGO, MASAKI NOGAWA, SEISHI SUGAHARA and HARUNA NAITO Discovery Research Laboratories, Nippon Shinyaku Co., Ltd., Kyoto, Japan Abstract. Background: The myelodysplastic syndromes of tumor suppressor genes such as CDKN2B, which codes for (MDS) are a group of stem cell disorders characterized by cyclin-dependent kinase 4 inhibitor B (also known as multiple dysplasia of one or more hematopoietic cell lineages and a tumor suppressor 2 or p15INK4B), may play an important role risk of progression to acute myeloid leukemia. The cytidine in neoplastic progression (1). analog azacitidine (Vidaza), a hypomethylating agent, In 2004, the cytidine analog azacitidine (5-azacytidine; improves survival in patients with MDS, but its mechanism of marketed as Vidaza by Nippon Shinyaku from 2011 in Japan) action is not well understood. Materials and Methods: The was the first drug to be approved by the USA Food and Drug effects of azacitidine on the MDS-derived cell line SKM-1 Administration for the treatment of MDS, and it increases were investigated by DNA methylation assay, cell proliferation median overall survival in higher-risk patients with MDS in assay, and a subcutaneous xenograft mouse model. Results: comparison to drugs used in conventional care regimens, Azacitidine and decitabine induced hypomethylation of the including the structurally related cytarabine (5). Azacitidine tumor suppressor gene cyclin-dependent kinase 4 inhibitor B acts by the dual mechanisms of DNA hypomethylation and (CDKN2B) in SKM-1 cells, whereas the deoxycytidine analog cytotoxicity.
    [Show full text]
  • Acute Myeloid Leukemia (AML) Treatment Regimens
    Acute Myeloid Leukemia (AML) Treatment Regimens Clinical Trials: The National Comprehensive Cancer Network recommends cancer patient participation in clinical trials as the gold standard for treatment. Cancer therapy selection, dosing, administration, and the management of related adverse events can be a complex process that should be handled by an experienced healthcare team. Clinicians must choose and verify treatment options based on the individual patient; drug dose modifications and supportive care interventions should be administered accordingly. The cancer treatment regimens below may include both U.S. Food and Drug Administration-approved and unapproved indications/regimens. These regimens are only provided to supplement the latest treatment strategies. These Guidelines are a work in progress that may be refined as often as new significant data becomes available. The NCCN Guidelines® are a consensus statement of its authors regarding their views of currently accepted approaches to treatment. Any clinician seeking to apply or consult any NCCN Guidelines® is expected to use independent medical judgment in the context of individual clinical circumstances to determine any patient’s care or treatment. The NCCN makes no warranties of any kind whatsoever regarding their content, use, or application and disclaims any responsibility for their application or use in any way. uInduction Therapy1 Note: All recommendations are Category 2A unless otherwise indicated. The NCCN believes the best option for any patient with cancer is in a clinical trial and strongly encourages this option for all patients. PATIENT CRITERIA REGIMEN AND DOSING Age <60 years2-8 Days 1–3: An anthracycline (daunorubicin 60–90mg/m2 IV OR idarubicin 12mg/m2) Days 1–7: Cytarabine 100–200mg/m2 continuous IV (Category 1).
    [Show full text]