Epigenetic Cancer Therapy: Rationales, Targets and Drugs

Total Page:16

File Type:pdf, Size:1020Kb

Epigenetic Cancer Therapy: Rationales, Targets and Drugs Oncogene (2012) 31, 4257–4265 & 2012 Macmillan Publishers Limited All rights reserved 0950-9232/12 www.nature.com/onc REVIEW Epigenetic cancer therapy: rationales, targets and drugs M Rius and F Lyko Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, Heidelberg, Germany The fundamental role of altered epigenetic modification many tumors show an aberrant epigenetic modification patterns in tumorigenesis establishes epigenetic regulatory pattern. These tumor-specific epigenetic alterations or enzymes as important targets for cancer therapy. Over the epigenetic mutations, affect a variety of cancer-related past few years, several drugs with an epigenetic activity genes. The most prominent example for epimutations is have received approval for the treatment of cancer provided by the widely observed epigenetic silencing of patients, which has led to a detailed characterization of tumor suppressor genes that is closely associated with their modes of action. The results showed that both promoter hypermethylation (Esteller, 2007; Jones and established drug classes, the histone deacetylase (HDAC) Baylin, 2007). It has long been known that DNA inhibitors and the DNA methyltransferase inhibitors, show methyltransferase inhibitors can effectively induce DNA substantial limitations in their epigenetic specificity. HDAC demethylation and phenotypic changes related to the inhibitors are highly specific drugs, but the enzymes have a reactivation of epigenetically silenced genes (Jones and broad substrate specificity and deacetylate numerous Taylor, 1980). These findings were later adapted to the proteins that are not associated with epigenetic regulation. targeting of epigenetic mutations in cancer and thus Similarly, the induction of global DNA demethylation by established the fundamental concept of epigenetic non-specific inhibition of DNA methyltransferases shows cancer therapy (Egger et al., 2004). pleiotropic effects on epigenetic regulation with no apparent Research over the past decade has shown that tumor-specificity. Second-generation azanucleoside drugs epigenetic regulation occurs on several mechanistic have integrated the knowledge about the cellular uptake and levels, including DNA methylation, covalent histone metabolization pathways, but do not show any increased modifications and small regulatory RNAs (Esteller, specificity for cancer epigenotypes. As such, the traditional 2007; Jones and Baylin, 2007). Thus, in addition to rationale of epigenetic cancer therapy appears to be in need DNA methyltransferase (DNMT) inhibitors, histone of refinement, as we move from the global inhibition of deacetylase (HDAC) inhibitors have also contributed to epigenetic modifications toward the identification and shape epigenetic cancer therapy. Altogether, there are targeting of tumor-specific epigenetic programs. Recent currently four FDA-approved drugs with an epigenetic studies have identified epigenetic mechanisms that promote mode of action: the DNA methyltransferase inhibitors self-renewal and developmental plasticity in cancer cells. 5-azacytidine (Vidaza) and decitabine (20-deoxy-5-aza- Druggable somatic mutations in the corresponding epige- cytidine, Dacogen) and the HDAC inhibitors suberoy- netic regulators are beginning to be identified and should lanilide hydroxamic acid (SAHA, Zolinza) and facilitate the development of epigenetic therapy approaches romidepsin (Istodax). Various additional DNMT and with improved tumor specificity. HDAC inhibitors are currently being evaluated in Oncogene (2012) 31, 4257–4265; doi:10.1038/onc.2011.601; preclinical studies and in clinical trials. However, there published online 19 December 2011 are currently no clinical trials with drug candidates directed against other epigenetic targets. This situation Keywords: DNA methylation; chromatin; DNA methyl- warrants a detailed discussion of DNMT and HDAC transferase inhibitors; histone deacetylase inhibitors; inhibitors before novel approaches towards an epigenetic metabolic activation; stem cells therapy of cancer are considered. 5-Azacytidine and decitabine have shown significant clinical benefit in the treatment of myelodysplastic syndrome (Silverman et al., 2002; Kantarjian et al., 2006) and are also used for the treatment of myeloid Introduction leukemias. Both drugs are particularly effective when administered at low doses, which are sometimes below The traditional rationale for the development of 10% of their maximum tolerated doses (Issa and epigenetic cancer drugs is based on the observation that Kantarjian, 2009). These conditions are generally assumed to maximize DNA demethylation responses, and several studies have confirmed that drug-induced Correspondence: Dr F Lyko, Deutsches Krebsforschungszentrum, Im DNA demethylation occurs in patients (Issa et al., 2005; Neuenheimer Feld 580, 69120 Heidelberg, Germany. Mund et al., 2005; Soriano et al., 2007; Stresemann and E-mail: [email protected] Received 14 September 2011; revised and accepted 21 November 2011; Lyko, 2008). However, a direct link between DNA published online 19 December 2011 demethylation and clinical responses has not been Epigenetic cancer therapy M Rius and F Lyko 4258 demonstrated yet and there are currently no established required the presence of HR23B. These data suggest DNA methylation biomarkers that accurately predict that HDAC inhibitors target aberrant proteasome patient responses (Fandy et al., 2009). This remains as activity in cancer cells, which is possibly mediated by an important challenge that is related to the complex hyperacetylation of proteasome-associated non-histone mode of action of azanucleosides (see below) and that proteins. will have to be addressed in future studies to firmly Moreover, when yeast cells were incubated in the establish clinical proof-of-concept for epigenetic therapy presence of the HDAC inhibitor valproic acid, the drug with these drugs. specifically counteracted the activation of the ATR- The HDAC inhibitors SAHA and romidepsin are mediated DNA damage checkpoint response (Robert approved for the treatment of cutaneous T-cell lympho- et al., 2011). Surprisingly, the effect on DNA damage ma (Olsen et al., 2007; Piekarz et al., 2009). Numerous repair involved the degradation of key recombination additional HDAC inhibitors are currently undergoing proteins by autophagy (Robert et al., 2011). These clinical testing. The high number of developmental observations were explained by a model where protein drugs is related to the excellent druggability of the acetylation marks DNA recombination proteins for HDAC enzyme family, which permits the development transport to the vacuole and subsequent degradation, of potent isoenzyme-specific inhibitors (Bolden et al., and where deacetylation of these factors is required for 2006). It is important to notice that histone deacetyla- their stabilization in subnuclear compartments with tion has been shown to synergistically interact with severely damaged DNA. In this model, HDAC inhibi- DNA methylation in the epigenetic silencing of cancer tors would affect major oncological pathways (DNA genes (Cameron et al., 1999). This observation has also damage response and autophagy) without invoking any provided the scientific rationale for various clinical trials effects on epigenetic gene regulation. Altogether, these with combinations of DNMT and HDAC inhibitors. findings further illustrate why HDAC inhibitors should Somewhat surprisingly, these studies have yet to reveal not be considered as specific epigenetic drugs. It is synergistic clinical effects, which might again be related possible (although not proven) that targeting of other to complexities in the drugs’ modes of action that are histone modifying enzymes, like histone methyltrans- only beginning to become elucidated (see below). ferases and histone demethylases, might permit a higher degree of epigenetic specificity (see below). Are HDAC inhibitors epigenetic drugs? Epigenetic cancer therapy with DNMT inhibitors The important role of HDAC inhibitors in current concepts of epigenetic cancer therapy is often inter- The most frequently used epigenetic drugs are the preted to reflect a high specificity of HDACs for histone cytosine derivatives 5-azacytidine and decitabine. Both substrates. However, similar to the situation with DNA drugs are undergoing a series of metabolic activation methyltransferase inhibitors, a direct link between drug- steps before they can become incorporated into nucleic induced histone hyperacetylation and clinical responses acids (Figure 1). Incorporation of azacytosine bases into remains to be established. It is now becoming increas- DNA and RNA triggers a multitude of stress and ingly clear that the enzymatic activity of HDACs is not damage signaling pathways (Hollenbach et al., 2010), restricted to histone proteins, and treatment of human but also induces the formation of covalent adducts cancer cell lines with highly specific HDAC inhibitors between DNA and DNA methyltransferases. This can induce hyperacetylation of 1750 proteins (Choudh- covalent trapping results in the depletion of the enzymes ary et al., 2009). This finding strongly suggests that non- and thus causes a global reduction of DNA methylation histone proteins constitute the large majority of HDAC (Stresemann and Lyko, 2008). A global reduction of substrates. In addition, these studies also suggest that DNA methylation has been shown to have antitumoral the complexity and functional significance of the human effects in a mouse model for intestinal tumorigenesis
Recommended publications
  • The Regulation of Carbamoyl Phosphate Synthetase-Aspartate Transcarbamoylase-Dihydroorotase (Cad) by Phosphorylation and Protein-Protein Interactions
    THE REGULATION OF CARBAMOYL PHOSPHATE SYNTHETASE-ASPARTATE TRANSCARBAMOYLASE-DIHYDROOROTASE (CAD) BY PHOSPHORYLATION AND PROTEIN-PROTEIN INTERACTIONS Eric M. Wauson A dissertation submitted to the faculty of the University of North Carolina at Chapel Hill in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Pharmacology. Chapel Hill 2007 Approved by: Lee M. Graves, Ph.D. T. Kendall Harden, Ph.D. Gary L. Johnson, Ph.D. Aziz Sancar M.D., Ph.D. Beverly S. Mitchell, M.D. 2007 Eric M. Wauson ALL RIGHTS RESERVED ii ABSTRACT Eric M. Wauson: The Regulation of Carbamoyl Phosphate Synthetase-Aspartate Transcarbamoylase-Dihydroorotase (CAD) by Phosphorylation and Protein-Protein Interactions (Under the direction of Lee M. Graves, Ph.D.) Pyrimidines have many important roles in cellular physiology, as they are used in the formation of DNA, RNA, phospholipids, and pyrimidine sugars. The first rate- limiting step in the de novo pyrimidine synthesis pathway is catalyzed by the carbamoyl phosphate synthetase II (CPSase II) part of the multienzymatic complex Carbamoyl phosphate synthetase, Aspartate transcarbamoylase, Dihydroorotase (CAD). CAD gene induction is highly correlated to cell proliferation. Additionally, CAD is allosterically inhibited or activated by uridine triphosphate (UTP) or phosphoribosyl pyrophosphate (PRPP), respectively. The phosphorylation of CAD by PKA and ERK has been reported to modulate the response of CAD to allosteric modulators. While there has been much speculation on the identity of CAD phosphorylation sites, no definitive identification of in vivo CAD phosphorylation sites has been performed. Therefore, we sought to determine the specific CAD residues phosphorylated by ERK and PKA in intact cells.
    [Show full text]
  • Hodgkin Lymphoma Treatment Regimens
    HODGKIN LYMPHOMA TREATMENT REGIMENS (Part 1 of 5) 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 health care 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 provided only to supplement the latest treatment strategies. These Guidelines are a work in progress that may be refined as often as new significant data become 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. Classical Hodgkin Lymphoma1 Note: All recommendations are Category 2A unless otherwise indicated. Primary Treatment Stage IA, IIA Favorable (No Bulky Disease, <3 Sites of Disease, ESR <50, and No E-lesions) REGIMEN DOSING Doxorubicin + Bleomycin + Days 1 and 15: Doxorubicin 25mg/m2 IV push + bleomycin 10units/m2 IV push + Vinblastine + Dacarbazine vinblastine 6mg/m2 IV over 5–10 minutes + dacarbazine 375mg/m2 IV over (ABVD) (Category 1)2-5 60 minutes.
    [Show full text]
  • (DAC) Followed by Clofarabine, Idarubicin, and Cytarabine (CIA) in Acute Leukemia 2012-1064
    2012-1064 September 02, 2014 Page 1 Protocol Page Phase I/II Study of Decitabine (DAC) followed by Clofarabine, Idarubicin, and Cytarabine (CIA) in Acute Leukemia 2012-1064 Core Protocol Information Short Title Decitabine followed by Clofarabine, Idarubicin, and Cytarabine in Acute Leukemia Study Chair: Nitin Jain Additional Contact: Allison Pike Jeannice Y. Theriot Leukemia Protocol Review Group Department: Leukemia Phone: 713-745-6080 Unit: 428 Full Title: Phase I/II Study of Decitabine (DAC) followed by Clofarabine, Idarubicin, and Cytarabine (CIA) in Acute Leukemia Protocol Type: Standard Protocol Protocol Phase: Phase I/Phase II Version Status: Terminated 01/12/2018 Version: 12 Submitted by: Jeannice Y. Theriot--4/26/2017 2:13:38 PM OPR Action: Accepted by: Melinda E. Gordon -- 5/1/2017 7:55:15 AM Which Committee will review this protocol? The Clinical Research Committee - (CRC) 2012-1064 September 02, 2014 Page 2 Protocol Body Phase I/II Study of Decitabine (DAC) followed by Clofarabine, Idarubicin, and Cytarabine (CIA) in Acute Leukemia 1. OBJECTIVES Phase I Primary: To determine the maximal tolerated dose (MTD) of clofarabine to be used in portion II of the study Phase II Primary: To determine the response rate of the DAC-CIA regimen Secondary: A) To determine the toxicity of the combination regimen B) To determine the disease-free survival (DFS) and overall survival (OS) rates 2. RATIONALE 2.1 Acute Myelogenous Leukemia Acute myelogenous leukemia (AML) is the most common acute leukemia in adults. It is estimated that 13,780 men and women will be diagnosed with and 10,200 men and women will die of acute myeloid leukemia in the year 2012.1 AML is a disease with a poor prognosis with a 5-year survival of only around 30%.2,3 Certain subgroups of AML have a particularly worse Page 1 of 34 outcome such as patients with relapsed and/or refractory AML and AML arising from antecedent myelodysplastic syndrome (MDS) or myeloproliferative neoplasms (MPNs).
    [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]
  • A Phase I Trial of Pemetrexed Plus Gemcitabine Given Biweekly with B-Vitamin Support in Solid Tumor Malignancies Or Advanced Epithelial Ovarian Cancer Martee L
    Cancer Therapy: Clinical A Phase I Trial of Pemetrexed Plus Gemcitabine Given Biweekly with B-Vitamin Support in Solid Tumor Malignancies or Advanced Epithelial Ovarian Cancer Martee L. Hensley,1Joseph Larkin,1Matthew Fury,1Scott Gerst,1D. Fritz Tai,2 Paul Sabbatini,1 Jason Konner,1Mauro Orlando,2 Tiana L. Goss,2 and Carol A. Aghajanian1 Abstract Purpose: To determine the maximally tolerated dose (MTD) of biweekly pemetrexed with gemcitabine plus B12 and folate supplementation in patients with advanced solid tumors and ovarian cancer. Experimental Design: Patients with no prior pemetrexed or gemcitabine therapy enrolled in cohorts of three, expanding to six if dose-limiting toxicity (DLT) was observed. Pemetrexed, escalated from to 700 mg/m2, was given before gemcitabine 1,500 mg/m2 every 14 days. DLTs were grade 4 neutropenia lasting >7 days or febrile neutropenia, grade 4 or 3 thrombocytopenia (with bleeding), grade z3 nonhematologic toxicity, or treatment delay of z1 week due to unresolved toxicity. Results: The ovarian cancer cohort enrolled 24 patients with unlimited prior cytotoxic chemo- therapies. MTD was observed at pemetrexed 600 mg/m2, with 2 of 9 patients experiencing DLT. Most common grade 3 to 4 toxicities per patient were neutropenia (83%), leukopenia (67%), lymphopenia (73%), and febrile neutropenia (12%). Median cycle per patient was 8 (range, 1-16). Six of 21 (28%) patients had confirmed partial responses. Study protocol was modified for the solid tumor cohort (n = 30) to enroll patients with two or more prior cytotoxic regimens. MTD was observed at pemetrexed 500 mg/m2, with 1of 9 patients experiencing DLT.
    [Show full text]
  • Decitabine Plus CLAG Chemotherapy As a Bridge to Haploidentical
    Jin et al. BMC Cancer (2019) 19:242 https://doi.org/10.1186/s12885-019-5464-0 CASEREPORT Open Access Decitabine plus CLAG chemotherapy as a bridge to haploidentical transplantation in the setting of acute myeloid leukemia relapse after HLA-matched sibling transplantation: a case report Mengqi Jin, Yongxian Hu, Wenjun Wu, Yi Luo, Yamin Tan, Jian Yu, Aiyun Jin, Luxin Yang, He Huang* and Guoqing Wei* Abstract Background: Patients with relapsed/refractory acute myeloid leukemia after hematopoietic stem cell transplantation (HSCT) have a poor prognosis, with a 2-year survival rate of 14%. The optimal treatment for these patients remains unclear. To treat these patients, we designed a new salvage regimen consisting of decitabine, cladribine, cytarabine, and granulocyte-stimulating factor (D-CLAG). Case presentation: Here, we describe a case of acute monocytic leukemia with a complex karyotype in a 38-year-old female patient who relapsed after her first HSCT, which was performed using a matched sibling donor. The patient did not respond to standard induction chemotherapy and subsequently achieved complete remission with the D-CLAG regimen. No severe hematological or extramedullary toxicity was observed. Subsequently, the patient received a second D-CLAG regimen as a bridge therapy and directly underwent haploidentical related HSCT. Following HSCT, the marrow showed complete hematologic and cytogenetic remission. Currently, 1 year after transplantation, the patient’s general condition remains good. Conclusions: This case suggests that the D-CLAG regimen can be an option for reinduction in relapsed refractory AML patients as a bridge to transplantation. Nevertheless, further research will be required in the future as this report describes only a single case.
    [Show full text]
  • GEMCITABINE Hcl) for INJECTION
    FDA REVISED LABEL – VERSION 082598 Page 1 PA 1634 AMP GEMZAR® (GEMCITABINE HCl) FOR INJECTION DESCRIPTION Gemzar® (gemcitabine HCl) is a nucleoside analogue that exhibits antitumor activity. Gemcitabine HCl is 2'-deoxy-2',2'-difluorocytidine monohydrochloride (ß-isomer). The structural formula is as follows: NH 2•HCl N HO N O O F H OH F The empirical formula for gemcitabine HCl is C9H11F2N3O4 • HCl. It has a molecular weight of 299.66. Gemcitabine HCl is a white to off-white solid. It is soluble in water, slightly soluble in methanol, and practically insoluble in ethanol and polar organic solvents. The clinical formulation is supplied in a sterile form for intravenous use only. Vials of Gemzar contain either 200 mg or 1 g of gemcitabine HCl (expressed as free base) formulated with mannitol (200 mg or 1 g, respectively) and sodium acetate (12.5 mg or 62.5 mg, respectively) as a sterile lyophilized powder. Hydrochloric acid and/or sodium hydroxide may have been added for pH adjustment. CLINICAL PHARMACOLOGY Gemcitabine exhibits cell phase specificity, primarily killing cells undergoing DNA synthesis (S-phase) and also blocking the progression of cells through the G1/S-phase boundary. Gemcitabine is metabolized intracellularly by nucleoside kinases to the active diphosphate (dFdCDP) and triphosphate (dFdCTP) nucleosides. The cytotoxic effect of gemcitabine is attributed to a combination of two actions of the diphosphate and the triphosphate nucleosides, which leads to inhibition of DNA synthesis. First, gemcitabine diphosphate inhibits ribonucleotide reductase, which is responsible for catalyzing the reactions that generate the deoxynucleoside triphosphates for DNA synthesis.
    [Show full text]
  • Intravesical Administration of Therapeutic Medication for the Treatment of Bladder Cancer Jointly Developed with the Society of Urologic Nurses and Associates (SUNA)
    Intravesical Administration of Therapeutic Medication for the Treatment of Bladder Cancer Jointly developed with the Society of Urologic Nurses and Associates (SUNA) Revised: June 2020 Workgroup Members: AUA: Roxy Baumgartner, RN, APN-BC; Sam Chang, MD; Susan Flick, CNP; Howard Goldman, MD, FACS; Jim Kovarik, MS, PA-C; Yair Lotan, MD; Elspeth McDougall, MD, FRCSC, MHPE; Arthur Sagalowsky, MD; Edouard Trabulsi, MD SUNA: Debbie Hensley, RN; Christy Krieg, MSN, CUNP; Leanne Schimke, MSN, CUNP I. Statement of Purpose: To define the performance guidance surrounding the instillation of intravesical cytotoxic, immunotherapeutic, and/or therapeutic drugs via sterile technique catheterization for patients with non-muscle invasive bladder cancer (NMIBC, urothelial carcinoma). II. Population: Adult Urology III. Definition: Intravesical therapy involves instillation of a therapeutic agent directly into the bladder via insertion of a urethral catheter. IV. Indications: For administration of medication directly into the bladder via catheterization utilizing sterile technique for NMIBC treatment. V. Guidelines and Principles: Health care personnel (MD, NP, PA, RN, LPN, or MA) performing intravesical therapy must be educated, demonstrate competency, and understand the implications of non-muscle invasive bladder cancer. (Scope of practice for health care personnel listed may vary based on state or institution). This should include associated health and safety issues regarding handling of cytotoxic, and immunotherapeutic agents; and documented competency of safe practical skills. At a minimum, each institution or office practice setting should implement an established, annual competency program to review safety work practices and guidelines regarding storage, receiving, handling/ transportation, administration, disposal, and handling a spill of hazardous drugs. (Mellinger, 2010) VI.
    [Show full text]
  • Comparison Between 5-Day Decitabine and 7-Day Azacitidine For
    www.nature.com/scientificreports OPEN Comparison between 5-day decitabine and 7-day azacitidine for lower-risk myelodysplastic syndromes with poor prognostic features: a retrospective multicentre cohort study Byung-Hyun Lee 1, Ka-Won Kang 1, Min Ji Jeon2, Eun Sang Yu2, Dae Sik Kim2, Hojoon Choi 3, Se Ryeon Lee3, Hwa Jung Sung3, Byung Soo Kim1, Chul Won Choi2* & Yong Park1* Numerous studies have analysed the clinical efcacies of hypomethylating agents (HMAs) in patients with myelodysplastic syndromes (MDS). However, reports that compare the two HMAs, decitabine and azacitidine, in patients with lower-risk (low and intermediate-1) MDS are limited. We compared 5-day decitabine and 7-day azacitidine regimens in terms of treatment responses, survival outcomes, and adverse events in patients with lower-risk MDS with poor prognostic features. The overall response rates (ORRs) were 67.2% and 44.0% in the patients treated with decitabine and azacitidine, respectively (P = 0.014). While the median progression-free survival (PFS) was signifcantly better in the patients treated with decitabine than in those treated with azacitidine (P = 0.019), no signifcant diferences in event-free and overall survival rates were observed between the two groups. Multivariate analysis revealed that compared with azacitidine treatment, decitabine treatment is signifcantly associated with a higher ORR (P = 0.026) and longer PFS (P = 0.037). No signifcant diferences were observed in the incidence of grade 3 or higher haematologic adverse events in response to the two HMAs. In conclusion, in lower-risk MDS, especially with poor prognostic features, ORR and PFS were signifcantly better with 5-day decitabine treatment than with 7-day azacitidine treatment, with comparable safety.
    [Show full text]
  • Download Download
    Robyn A Lindley. Medical Research Archives vol 8 issue 8. Medical Research Archives REVIEW ARTICLE Review of the mutational role of deaminases and the generation of a cognate molecular model to explain cancer mutation spectra Author Robyn A Lindley1,2 1Department of Clinical Pathology 2GMDx Genomics Ltd, The Victorian Comprehensive Cancer Centre Level 3 162 Collins Street, Faculty of Medicine, Dentistry & Health Sciences Melbourne VIC3000, AUSTRALIA University of Melbourne, Email: [email protected] 305 Gratton Street, Melbourne, VIC 3000, AUSTRALIA Email: [email protected] Correspondence: Robyn A Lindley, Department of Clinical Pathology, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, 305 Gratton Street, Melbourne VIC 3000 AUSTRALIA Mobile: +61 (0) 414209132 Email: [email protected] Abstract Recent developments in somatic mutation analyses have led to the discovery of codon-context targeted somatic mutation (TSM) signatures in cancer genomes: it is now known that deaminase mutation target sites are far more specific than previously thought. As this research provides novel insights into the deaminase origin of most of the somatic point mutations arising in cancer, a clear understanding of the mechanisms and processes involved will be valuable for molecular scientists as well as oncologists and cancer specialists in the clinic. This review will describe the basic research into the mechanism of antigen-driven somatic hypermutation of immunoglobulin variable genes (Ig SHM) that lead to the discovery of TSM signatures, and it will show that an Ig SHM-like signature is ubiquitous in the cancer exome. Most importantly, the data discussed in this review show that Ig SHM-like cancer-associated signatures are highly targeted to cytosine (C) and adenosine (A) nucleotides in a characteristic codon-context fashion.
    [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]
  • Cytostatics in Dutch Surface Water: Use, Presence and Risks To
    Cytostatics in Dutch surface water Use, presence and risks to the aquatic environment RIVM Letter report 2018-0067 C. Moermond et al. Cytostatics in Dutch surface water Use, presence and risks to the aquatic environment RIVM Letter report 2018-0067 C. Moermond et al. RIVM Letter report 2018-0067 Colophon © RIVM 2018 Parts of this publication may be reproduced, provided acknowledgement is given to: National Institute for Public Health and the Environment, along with the title and year of publication. DOI 10.21945/RIVM-2018-0067 C. Moermond (auteur/coördinator), RIVM B. Venhuis (auteur/coördinator),RIVM M. van Elk (auteur), RIVM A. Oostlander (auteur), RIVM P. van Vlaardingen (auteur), RIVM M. Marinković (auteur), RIVM J. van Dijk (stagiair; auteur) RIVM Contact: Caroline Moermond VSP-MSP [email protected] This investigation has been performed by order and for the account of the Ministry of Infrastructure and Water management (IenW), within the framework of Green Deal Zorg en Ketenaanpak medicijnresten uit water. This is a publication of: National Institute for Public Health and the Environment P.O. Box 1 | 3720 BA Bilthoven The Netherlands www.rivm.nl/en Page 2 of 140 RIVM Letter report 2018-0067 Synopsis Cytostatics in Dutch surface water Cytostatics are important medicines to treat cancer patients. Via urine, cytostatic residues end up in waste water that is treated in waste water treatment plants and subsequently discharged into surface waters. Research from RIVM shows that for most cytostatics, their residues do not pose a risk to the environment. They are sufficiently metabolised in the human body and removed in waste water treatment plants.
    [Show full text]