Omacetaxine May Have a Role in Chronic Myeloid Leukaemia Eradication Through Downregulation of Mcl-1 and Induction of Apoptosis in Stem/Progenitor Cells

Total Page:16

File Type:pdf, Size:1020Kb

Omacetaxine May Have a Role in Chronic Myeloid Leukaemia Eradication Through Downregulation of Mcl-1 and Induction of Apoptosis in Stem/Progenitor Cells Leukemia (2011) 25, 985–994 & 2011 Macmillan Publishers Limited All rights reserved 0887-6924/11 www.nature.com/leu ORIGINAL ARTICLE Omacetaxine may have a role in chronic myeloid leukaemia eradication through downregulation of Mcl-1 and induction of apoptosis in stem/progenitor cells EK Allan1,2, TL Holyoake1, AR Craig3 and HG Jørgensen1 1Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK; 2Scottish National Blood Transfusion Service, Gartnavel General Hospital, Glasgow, UK and 3ChemGenex Pharmaceuticals Inc., Menlo Park, CA, USA Chronic myeloid leukaemia (CML) is maintained by a rare Therefore this product has, for a second time, become a population of tyrosine kinase inhibitor (TKI)-insensitive malig- valuable option in the treatment of resistant disease. Moreover, nant stem cells. Our long-term aim is to find a BcrAbl- the demonstration that omacetaxine can kill leukaemic stem independent drug that can be combined with a TKI to improve 4 overall disease response in chronic-phase CML. Omacetaxine cells in murine models has allowed the drug to be considered mepesuccinate, a first in class cetaxine, has been evaluated by as a therapeutic option for both persistent and resistant disease. clinical trials in TKI-insensitive/resistant CML. Omacetaxine Homoharringtonine (from here is referred to as omacetaxine) inhibits synthesis of anti-apoptotic proteins of the Bcl-2 family, is derived from various Cephalotaxus species (Chinese yew tree) including (myeloid cell leukaemia) Mcl-1, leading to cell death. and was first discovered by the Chinese to have natural anti- Omacetaxine effectively induced apoptosis in primary CML tumour and anti-leukaemic properties in the 1970s.5,6 Recent stem cells (CD34 þ 38lo) by downregulation of Mcl-1 protein. In work has elucidated a mechanism of action targeted to the contrast to our previous findings with TKIs, omacetaxine did 7 not accumulate undivided cells in vitro. Furthermore, the A-site cleft of eukaryotic ribosomes resulting in transient functionality of surviving stem cells following omacetaxine inhibition of protein synthesis of short-lived proteins, such as exposure was significantly reduced in a dose-dependant myeloid cell leukaemia (Mcl-1) protein (a highly expressed manner, as determined by colony forming cell and the more member of the Bcl-2 family of anti-apoptotic proteins), leading stringent long-term culture initiating cell colony assays. This to apoptosis.8 Further, it has been shown that Mcl-1 has a stem cell-directed activity was not limited to CML stem cells as þ critical role in the survival of leukaemia cells and its expression both normal and non-CML CD34 cells were sensitive to 9,10 inhibition. Thus, although omacetaxine is not leukaemia stem is upregulated with respect to normal. As such, omacetaxine cell specific, its ability to induce apoptosis of leukaemic stem acts independently of BcrAbl kinase activity, the causative cells distinguishes it from TKIs and creates the potential for a oncoprotein in CML. Taken together with a recently reported curative strategy for persistent disease. ability to kill stem cells,4 omacetaxine could therefore be an Leukemia (2011) 25, 985–994; doi:10.1038/leu.2011.55; interesting therapeutic modality in minimal residual disease in published online 5 April 2011 Keywords: omacetaxine; leukaemic stem cells; CML; BcrAbl; Mcl-1 CML, where the continued detection of BcrAbl transcripts is thought to originate from the stem cell compartment,11 and leukaemic stem cell survival may be BcrAbl independent.12 Therefore, the aim of our study was to assess the activity of omacetaxine against CML stem/progenitor cells in vitro in order Introduction to identify potential ways of eradicating persistent disease. Sequential clinical samples from newly diagnosed cases were Omacetaxine mepesuccinate (ChemGenex Pharmaceuticals selected for assay and were not subject to mutation analysis. We Inc., Menlo Park, CA, USA), a first in class cetaxine, is a have previously shown CML CD34 þ cells to be insensitive to TKI semisynthetic alkaloid that has demonstrable activity in clinical by virtue of their quiescence and primitive phenotype. From our trials in acute myeloid leukaemia, myelodysplastic syndrome, as on-going work and that of others, it is our belief that CML stem well as in chronic myeloid leukaemia (CML).1 However, with cells may not be oncogene addicted (that is, dependant on BcrAbl the continued success of tyrosine kinase inhibitors (TKIs), such for their survival), and thus any novel agents or novel indications as imatinib, nilotinib and dasatinib, as first- and second-line for existing molecules with targeting potential at the (stem) cell treatment for CML, the role of omacetaxine has been restricted level to complement TKI modalities is of significant interest. to niche indications, including failure of two or more TKI or with the TKI-insensitive mutation, T315I, against which all currently licensed TKI are ineffective. Omacetaxine is a semisynthetic, Materials and methods subcutaneously bioavailable form of homoharringtonine. Homoharringtonine was used in second-line therapy for those Reagents CML patients in the pre-TKI era of the 1990s who failed Omacetaxine mepesuccinate, supplied by ChemGenex 2,3 interferon-a therapy and were ineligible for a transplant Pharmaceuticals , was stored lyophilised at room temperature; once reconstituted in saline to 10 mM, the stock solution was Correspondence: Professor TL Holyoake, Paul O’Gorman Leukaemia stored at 4 1C for up to 3 months. Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Gartnavel General Hospital, 1053 Great Western Road, Glasgow G12 0YN, UK. E-mail: tessa.holyoake@ glasgow.ac.uk Cell culture Received 19 August 2010; revised 28 January 2011; accepted Primary CML cells were obtained with informed consent from 14 February 2011; published online 5 April 2011 leukapheresis samples of newly diagnosed patients with Omacetaxine induces stem cell apoptosis EK Allan et al 986 chronic-phase CML or PhÀ haematological disorders (for significant reduction in total cells (undivided plus proliferating example, lymphoma) as controls. CD34 þ cells enriched by cells), the absolute number of undivided cells (product of % positive selection (CliniMACS, Miltenyi Biotec Ltd., Bisley, Â total cell number) may remain unchanged. Surrey, UK) according to the manufacturer’s instructions to 495% purity were cryopreserved in 10% (v/v) dimethylsulph- oxide in 4% (w/v) ALBA (human albumin solution, Scottish Primitive subsets National Blood Transfusion Service, Edinburgh, UK) and stored The CD34 þ -enriched cells were stained with anti-CD34-APC in the vapour phase of liquid nitrogen until required. CD34 þ and anti-CD38-PE antibodies before cell sorting using an FACS cells were cultured in serum-free media comprising Iscove’s Aria (Becton Dickinson) into three populations: CD34lo38 þ modified Dulbecco’s medium with BIT (bovine serum albumin, (mature), CD34 þ 38 þ (progenitor) and CD34 þ 38lo (primitive). insulin, transferrin; Stem Cell Technologies, Vancouver, BC, The CD34 þ 38lo fraction approximates the most primitive Canada), 1 mmol/l glutamine, 1 mmol/l streptomycin/penicillin, quiescent stem cell pool (o 5% total CD34 þ cells). Cells were 40 ng/ml low-density lipoprotein and 0.1 mmol/l 2-mercapto- analysed using a BD FACS Canto instrument with Diva software. ethanol (all, Invitrogen Ltd, Paisley, UK), further supplemented with a 5-growth factor cocktail as previously described.13 The BcrAbl-positive human myeloid cell lines, K562 and Colony forming cell (CFC) and long-term KCL22 were maintained in RPMI 1640 (Sigma–Aldrich, Poole, culture-initiating cell (LTC-IC) Dorset, UK) supplemented with 10% (v/v) foetal calf serum After culture in omacetaxine for 24 or 72 h, CD34 þ cells containing 1 mmol/l each of streptomycin, penicillin and remaining were set up in CFC and LTC-IC assays to assess the glutamine (RPMI þ ). Total cell counts and viability were proliferative potential of cell populations remaining after drug assessed using the trypan blue dye exclusion method. treatment. For CFC assays, 2.5 Â 103 cells were added to 1.5 ml Methocult (Stem cell Technologies) in duplicate 35 mm culture dishes and cultured for 12–14 days at 37 1Cina Flow cytometry 5% CO2-humidified incubator. Apoptosis. To assess apoptosis after drug treatment, an LTC-IC assays were set up as previously described.15 Briefly, aliquot of cell suspension containing 1–2 Â 105 cells was M2-10B4 cells and SL/SL fibroblasts were established as feeder removed for staining with Annexin V-fluorescein isothiocyanate layers then irradiated at 80 Gy. CML cells remaining in culture and Viaprobe (BD Biosciences, Oxford, UK) as per the after drug treatment were washed three times and then plated in manufacturer’s instructions and analysed by flow cytometry duplicate on the irradiated feeder layers in Myelocult medium using a BD FACS Canto with Diva software. Cells undergoing supplemented with hydrocortisone at a final concentration of apoptosis initially become Annexin V positive (early apoptosis), 1 mM (Stem Cell Technologies). Cultures were maintained for and as apoptosis progresses, become positive for both Annexin V 5 weeks with weekly half medium changes. After 5 weeks, cells and Viaprobe (late apoptosis). were counted and 2.5 Â 104 cells were transferred to CFC assays and maintained in culture for a further 2 weeks in Methocult medium before the colonies were counted. Cell division tracking Upon recovery from liquid nitrogen, CML CD34 þ cells were stained with 1 mM carboxy-fluorescein diacetate succinimidyl Western blotting diester (CFSE; Molecular Probes, Invitrogen Ltd, Paisley, UK), as Following treatment with drugs, K562 and primary CML cells described previously in detail.14 Cells were then cultured in (2–5 Â 106) were pelleted by centrifugation, washed twice with serum-free media þ 5-growth factor in the presence or absence ice-cold phosphate-buffered saline, and then lysed in buffer of omacetaxine for 24 or 72 h.
Recommended publications
  • A Phase II Study of the Novel Proteasome Inhibitor Bortezomib In
    Memorial Sloan-Kettering Cance r Center IRB Protocol IRB#: 05-103 A(14) A Phase II Study of the Novel Proteas ome Inhibitor Bortezomib in Combination with Rituximab, Cyclophosphamide and Prednisone in Patients with Relapsed/Refractory I Indolent B-cell Lymphoproliferative Disorders and Mantle Cell Lymphoma (MCL) MSKCC THERAPEUTIC/DIAGNOSTIC PROTOCOL Principal Investigator: John Gerecitano, M.D., Ph.D. Co-Principal Carol Portlock, M.D. Investigator(s): IFormerly: A Phase I/II Study of the Nove l Proteasome Inhibitor Bortezomib in Combinati on with Rituximab, Cyclophosphamide and Prednisone in Patients with Relapsed/Refractory Indolent B-cell Lymphoproliferative Disorders and Mantle Cell Lymphoma (MCL) Amended: 07/25/12 Memorial Sloan-Kettering Cance r Center IRB Protocol IRB#: 05-103 A(14) Investigator(s): Paul Hamlin, M.D. Commack, NY Steven B. Horwitz, M.D. Philip Schulman, M.D. Alison Moskowitz, M.D. Stuart Lichtman, M.D Craig H. Moskowitz, M.D. Stefan Berger, M.D. Ariela Noy, M.D. Julie Fasano, M.D. M. Lia Palomba, M.D., Ph.D. John Fiore, M.D. Jonathan Schatz, M.D. Steven Sugarman, M.D David Straus, M.D. Frank Y. Tsai, M.D. Andrew D. Zelenetz, M.D., Ph.D. Matthew Matasar, M.D Rockville Center, NY Mark L. Heaney, M.D., Ph.D. Pamela Drullinksy, M.D Nicole Lamanna, M.D. Arlyn Apollo, M.D. Zoe Goldberg, M.D. Radiology Kenneth Ng, M.D. Otilia Dumitrescu, M.D. Tiffany Troso-Sandoval, M.D. Andrei Holodny, M.D. Sleepy Hollow, NY Nuclear Medicine Philip Caron, M.D. Heiko Schoder, M.D. Michelle Boyar, M.D.
    [Show full text]
  • VELCADE® (Bortezomib) for Injection
    1 VELCADE® (bortezomib) for Injection 2 PRESCRIBING INFORMATION 3 DESCRIPTION 4 VELCADE® (bortezomib) for Injection is an antineoplastic agent available for intravenous 5 injection (IV) use only. Each single dose vial contains 3.5 mg of bortezomib as a sterile 6 lyophilized powder. Inactive ingredient: 35 mg mannitol, USP. 7 Bortezomib is a modified dipeptidyl boronic acid. The product is provided as a mannitol boronic 8 ester which, in reconstituted form, consists of the mannitol ester in equilibrium with its 9 hydrolysis product, the monomeric boronic acid. The drug substance exists in its cyclic 10 anhydride form as a trimeric boroxine. 11 The chemical name for bortezomib, the monomeric boronic acid, is [(1R)-3-methyl-1-[[(2S)-1- 12 oxo-3-phenyl-2-[(pyrazinylcarbonyl) amino]propyl]amino]butyl] boronic acid. 13 Bortezomib has the following chemical structure: O OH H N N B N OH H O N 14 15 The molecular weight is 384.24. The molecular formula is C19H25BN4O4. The solubility of 16 bortezomib, as the monomeric boronic acid, in water is 3.3 to 3.8 mg/mL in a pH range of 2 to 17 6.5. 18 CLINICAL PHARMACOLOGY 19 Mechanism of Action 20 Bortezomib is a reversible inhibitor of the chymotrypsin-like activity of the 26S proteasome in 21 mammalian cells. The 26S proteasome is a large protein complex that degrades ubiquitinated 22 proteins. The ubiquitin-proteasome pathway plays an essential role in regulating the intracellular 23 concentration of specific proteins, thereby maintaining homeostasis within cells. Inhibition of 24 the 26S proteasome prevents this targeted proteolysis, which can affect multiple signaling 25 cascades within the cell.
    [Show full text]
  • Combining Milatuzumab with Bortezomib, Doxorubicin, Or Dexamethasone Improves Responses in Multiple Myeloma Cell Lines Rhona Stein,1Mitchell R
    CancerTherapy: Preclinical Combining Milatuzumab with Bortezomib, Doxorubicin, or Dexamethasone Improves Responses in Multiple Myeloma Cell Lines Rhona Stein,1Mitchell R. Smith,2 Susan Chen,1Maria Zalath,1and David M. Goldenberg1 Abstract Purpose: The humanized anti-CD74 monoclonal antibody, milatuzumab, is in clinical evaluation for the therapy of multiple myeloma (MM). The ability of milatuzumab to increase the efficacy of bortezomib, doxorubicin, and dexamethasone was examined in three human CD74+ MM cell lines, CAG, KMS11, KMS12-PE, and one CD74-MM cell line, OPM-2. Experimental Design: Activity of milatuzumab as a monotherapy and combined with the drugs was evaluated by studying in vitro cytotoxicity, signaling and apoptotic pathways, and in vivo therapeutic activity in severe combined immunodeficient (SCID) mouse models of MM. Results: Given as a monotherapy, cross-linked milatuzumab, but not milatuzumab alone, yielded sig- nificant antiproliferative effects in CD74+ cells.The combination of cross-linked milatuzumab with bortezomib, doxorubicin, or dexamethasone caused more growth inhibition than either cross-linked milatuzumab or drug alone, producing significant reductions in the IC50 of the drugs when combined. Efficacy of combined treatments was accompanied by increased levels of apoptosis measured by increases of activated caspase-3 and hypodiploid DNA. Both milatuzumab and bortezomib affect the nuclear factor-nB pathwayinCAGMMcells. In CAG-or KMS11-SCIDxenograftmodels ofdisseminated MM, milatuzumab more than doubled median survival time, compared withup to a 33% increase in median survival withbortezomib but no significant benefit with doxorubicin. Moreover, combining milatuzumabandbortezomibincreasedsurvivalsignificantlycomparedwitheither treatmentalone. Conclusions:The therapeutic efficacies of bortezomib, doxorubicin, and dexamethasone are en- hanced in MM cell lines when given in combination with milatuzumab, suggesting testing these combinations clinically.
    [Show full text]
  • Fluorouracil-Methotrexate (CMF PO)
    Chemotherapy Protocol BREAST CANCER CYCLOPHOSPHAMIDE (PO)-FLUOROURACIL-METHOTREXATE (CMF-PO) Regimen • Breast Cancer – Cyclophosphamide (PO)-Fluorouracil-Methotrexate (CMF PO) Indication • Adjuvant treatment of early breast cancer • WHO Performance status 0, 1, 2 Toxicity Drug Adverse Effect Cyclophosphamide Dysuria, haemorrhagic cystitis, taste disturbances Fluorouracil Diarrhoea, stomatitis Methotrexate Stomatitis, conjunctivitis, renal toxicity The presence of a third fluid compartment e.g. ascities or renal failure may delay methotrexate clearance hence increase toxicity. The adverse effects listed are not exhaustive. Please refer to the relevant Summary of Product Characteristics for full details. Monitoring Regimen • FBC, U&E’s and LFT’s prior to each cycle. • Patients with complete or partial dihydropyrimidine dehydrogenase (DPD) deficiency are at increased risk of severe and fatal toxicity during treatment with fluorouracil. All patients should be tested for DPD deficiency before initiation (cycle 1) to minimise the risk of these reactions Dose Modifications The dose modifications listed are for haematological, liver and renal function only. Dose adjustments may be necessary for other toxicities as well. In principle all dose reductions due to adverse drug reactions should not be re- escalated in subsequent cycles without consultant approval. It is also a general rule for chemotherapy that if a third dose reduction is necessary treatment should be stopped. Version 1.2 (November 2020) Page 1 of 6 Breast – Cyclophosphamide (PO)-Fluorouracil-Methotrexate (CMF-PO) Please discuss all dose reductions / delays with the relevant consultant before prescribing, if appropriate. The approach may be different depending on the clinical circumstances. The following is a general guide only. Haematological Prior to prescribing the following treatment criteria must be met on day 1 of treatment.
    [Show full text]
  • 5-Fluorouracil + Adriamycin + Cyclophosphamide) Combination in Differentiated H9c2 Cells
    Article Doxorubicin Is Key for the Cardiotoxicity of FAC (5-Fluorouracil + Adriamycin + Cyclophosphamide) Combination in Differentiated H9c2 Cells Maria Pereira-Oliveira, Ana Reis-Mendes, Félix Carvalho, Fernando Remião, Maria de Lourdes Bastos and Vera Marisa Costa * UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; [email protected] (M.P.-O.); [email protected] (A.R.-M.); [email protected] (F.C.); [email protected] (F.R.); [email protected] (M.L.B.) * Correspondence: [email protected] Received: 4 October 2018; Accepted: 3 January 2019; Published: 10 January 2019 Abstract: Currently, a common therapeutic approach in cancer treatment encompasses a drug combination to attain an overall better efficacy. Unfortunately, it leads to a higher incidence of severe side effects, namely cardiotoxicity. This work aimed to assess the cytotoxicity of doxorubicin (DOX, also known as Adriamycin), 5-fluorouracil (5-FU), cyclophosphamide (CYA), and their combination (5-Fluorouracil + Adriamycin + Cyclophosphamide, FAC) in H9c2 cardiac cells, for a better understanding of the contribution of each drug to FAC-induced cardiotoxicity. Differentiated H9c2 cells were exposed to pharmacological relevant concentrations of DOX (0.13–5 μM), 5-FU (0.13–5 μM), CYA (0.13–5 μM) for 24 or 48 h. Cells were also exposed to FAC mixtures (0.2, 1 or 5 μM of each drug and 50 μM 5-FU + 1 μM DOX + 50 μM CYA). DOX was the most cytotoxic drug, followed by 5-FU and lastly CYA in both cytotoxicity assays (reduction of 3-(4,5-dimethylthiazol-2- yl)-2,5-diphenyl tetrazolium bromide (MTT) and neutral red (NR) uptake).
    [Show full text]
  • Arsenic Trioxide Targets MTHFD1 and SUMO-Dependent Nuclear De Novo Thymidylate Biosynthesis
    Arsenic trioxide targets MTHFD1 and SUMO-dependent PNAS PLUS nuclear de novo thymidylate biosynthesis Elena Kamyninaa, Erica R. Lachenauera,b, Aislyn C. DiRisioa, Rebecca P. Liebenthala, Martha S. Fielda, and Patrick J. Stovera,b,c,1 aDivision of Nutritional Sciences, Cornell University, Ithaca, NY 14853; bGraduate Field of Biology and Biomedical Sciences, Cornell University, Ithaca, NY 14853; and cGraduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853 Contributed by Patrick J. Stover, February 12, 2017 (sent for review December 1, 2016; reviewed by I. David Goldman and Anne Parle-McDermott) Arsenic exposure increases risk for cancers and is teratogenic in levels. Decreased rates of de novo dTMP synthesis can be caused animal models. Here we demonstrate that small ubiquitin-like by the action of chemotherapeutic drugs (19), through inborn modifier (SUMO)- and folate-dependent nuclear de novo thymidylate errors of folate transport and metabolism (15, 18, 20, 21), by (dTMP) biosynthesis is a sensitive target of arsenic trioxide (As2O3), inhibiting translocation of the dTMP synthesis pathway enzymes leading to uracil misincorporation into DNA and genome instability. into the nucleus (2) and by dietary folate deficiency (22, 23). Im- Methylenetetrahydrofolate dehydrogenase 1 (MTHFD1) and serine paired dTMP synthesis leads to genome instability through well- hydroxymethyltransferase (SHMT) generate 5,10-methylenetetrahy- characterized mechanisms associated with uracil misincorporation drofolate for de novo dTMP biosynthesis and translocate to the nu- into nuclear DNA and subsequent futile cycles of DNA repair (24, cleus during S-phase, where they form a multienzyme complex with 25). Nuclear DNA is surveyed for the presence of uracil by a thymidylate synthase (TYMS) and dihydrofolate reductase (DHFR), as family of uracil glycosylases including: uracil N-glycolase (UNG), well as the components of the DNA replication machinery.
    [Show full text]
  • Antitumor Effects of Bortezomib (PS-341) on Primary Effusion Lymphomas
    Leukemia (2004) 18, 1699–1704 & 2004 Nature Publishing Group All rights reserved 0887-6924/04 $30.00 www.nature.com/leu Antitumor effects of bortezomib (PS-341) on primary effusion lymphomas JAn1, Y Sun1, M Fisher1 and MB Rettig1,2 1VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA; and 2Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA Primary effusion lymphomas (PELs) are a rare type of non- treatment of refractory multiple myeloma, where it has an Hodgkin’s lymphoma that are resistant to cytotoxic chemother- acceptable toxicity profile.4–6 By inhibiting the proteasome, apy. PELs manifest constitutive activation of nuclear factor kappa B (NF-jB), and inhibition of NF-jB induces apoptosis of which degrades proteins that are marked by ubiquitination, PELs and sensitizes to tumor necrosis factor-related apoptosis- bortezomib modulates the cellular concentrations of hundreds inducing ligand (TRAIL)-induced death. Bortezomib (PS-341), a of proteins that are involved in a wide variety of functions, peptidyl boronic acid inhibitor of the proteasome, is a potent including cell cycle progression, signal transduction, and agent against a wide range of hematologic malignancies and apoptosis.7,8 For example, the proteasome regulates the activity has been shown to inhibit NF-jB. Thus, we examined the of the NF-kB pathway, in that the degradation of I kappa B (IkB), cytotoxic effects of bortezomib alone and in combination with the NF-kB inhibitory protein, is dependent upon the ubiquitin– various drugs. Bortezomib potently inhibited NF-jB in PEL cells 9 in a dose-dependent manner.
    [Show full text]
  • Bortezomib Plus Docetaxel in Advanced Non-Small Cell Lung Cancer and Other Solid Tumors: a Phase I California Cancer Consortium Trial
    CORE Metadata, citation and similar papers at core.ac.uk Provided by Elsevier - Publisher Connector ORIGINAL ARTICLE Bortezomib Plus Docetaxel in Advanced Non-small Cell Lung Cancer and Other Solid Tumors: A Phase I California Cancer Consortium Trial Primo N. Lara, Jr., MD,*† Mariana Koczywas, MD,‡ David I. Quinn, MD, PhD,§ Heinz Josef Lenz, MD,§ Angela M. Davies, MD,* Derick H.M. Lau, MD, PhD,*† Paul H. Gumerlock, PhD,* Jeff Longmate, PhD,‡ James H. Doroshow, MD,‡ David Schenkein, MD,͉͉ Oscar Kashala, MD,͉͉ and David R. Gandara, MD*† n recent years, the inhibition of the 26S proteasome has Background: This phase I study was performed to determine the emerged as a rational anti-neoplastic strategy. The 26S dose-limiting toxicity and maximum tolerated dose (MTD) of do- I proteasome is a very large proteolytic complex involved in a cetaxel in combination with bortezomib in patients with advanced non-small cell lung cancer (NSCLC) or other solid tumors. significant catabolic pathway, the ubiquitin–proteasome path- way, for many intracellular regulatory proteins, including I␬B Methods: Patients were enrolled in cohorts of three over six dose ␬ ␬ ␬ levels. Each treatment cycle was 3 weeks long and consisted of one kinase/nuclear factor- B(I B/NF- B), p53, and the cyclin- docetaxel infusion (day 1) and four bortezomib injections (days 1, 4, dependent kinase inhibitors p21 and p27, which contribute to 8, and 11). Dose escalation and MTD determination were based on the regulation of the cell cycle, apoptosis, and angiogene- 1–3 the occurrence of dose-limiting toxicities in cycle 1 only.
    [Show full text]
  • Paclitaxel, Vinorelbine and 5-Fluorouracil in Breast Cancer Patients Pretreated with Adjuvant Anthracyclines
    British Journal of Cancer (2005) 92, 634 – 638 & 2005 Cancer Research UK All rights reserved 0007 – 0920/05 $30.00 www.bjcancer.com Paclitaxel, vinorelbine and 5-fluorouracil in breast cancer patients pretreated with adjuvant anthracyclines Clinical Studies 1 1 1 2 2 2 3 3 A Berruti , R Bitossi , G Gorzegno , A Bottini , D Generali , M Milani , D Katsaros , IA Rigault de la Longrais , 3 4 4 4 5 6 6 7 R Bellino , M Donadio , M Ardine , O Bertetto , S Danese , MG Sarobba , A Farris , V Lorusso and ,1 L Dogliotti* 1Oncologia Medica, Azienda Ospedaliera San Luigi, Regione Gonzole 10, 10043 Orbassano (TO), Italy; 2Breast Unit, Azienda Ospedaliera Istituti Ospitalieri, largo Priori, 26100 Cremona, Italy; 3Ginecologia Oncologica, Azienda Ospedaliera OIRM Sant’Anna, via Ventimiglia 3, 10126 Torino, Italy; 4 Oncologia Medica, Centro Oncologico Ematologico Subalpino, Azienda Ospedaliera San Giovanni Battista Molinette, corso Bramante 88, 10126 Torino, 5 6 Italy; Ginecologia Divisione A, Azienda Ospedaliera OIRM Sant’Anna, corso Spezia 60, 10126 Torino, Italy; Oncologia Medica, Istituto Clinica Medica 7 Universitaria, via San Pietro 8, 07100 Sassari, Italy; Oncologia Medica, Istituto Oncologico, via Amendola 209, 70126 Bari, Italy We investigated the activity and toxicity of a combination of vinorelbine (VNB), paclitaxel (PTX) and 5-fluorouracil (5-FU) continuous infusion administered as first-line chemotherapy in metastatic breast cancer patients pretreated with adjuvant À2 À2 anthracyclines. A total of 61 patients received a regimen consisting of VNB 25 mg m on days 1 and 15, PTX 60 mg m on days 1, 8 À2 and 15 and continuous infusion of 5-FU at 200 mg m every day.
    [Show full text]
  • Thesis Biochemistry By
    Ain shams Universirty Faculty Of Science Evaluation of anticancer effects of a novel proteasome inhibitor (Velcade), interferon (alpha-interferon) and antimyeloma antibodies on the growth of myeloma cells. Thesis Submitted for the Requirement of the Ph.D degree in Biochemistry By Hanan Mohammed Mohammed El shershaby. (M.Sc in Biochemistry) Under Supervision of Prof.Dr / Ibrahim Hassan Borai Prof.Dr/ Mohamed Taha Hussein El- Kolaly Prof. of Biochemistry Professor of Radiopharmaceutics Faculty Of Science Atomic Energy Authority Ain shams Universirty Prof.Dr/Naser Mohammed El-lahloby Prof. Dr/ Kamel Abd El.hameed Moustafa Prof. of Medical Oncology Professor of Biochemistry Cancer Institute Center Atomic Energy Authority Cairo University Dr / Ahmed Abd El.aziz sayed Assistant professor of Biochemistry Faculty Of Science Ain shams Universirty (2013) CONTENTS Introduction and Aim of the work 1 Review Of Literature 4 ◙ Multiple Myeloma 4 ◙ Cell cycle 16 ◙ Apoptosis 19 ◙ Caspases 23 ◙ Chemotherapy 25 ◙ Proteasomes 36 Bortezomib (Velcade) 38 ◙ Interferons 43 ◙ Immunity and Immune system 46 Antibodies 49 Production of Antibodies 51 ◙ Immunotherapy 57 ◙ Radioimmunotherapy ( RIT ) 61 ◙ Gene therapy 62 Materials and Methods 64 Results 92 Discussion 133 Summary 144 References 148 Arabic Summary LIST OF FIGURES Figure (1): Overview of the cell cycle. 16 Figure (2): Apoptosis – the programmed death of a cell. 20 Figure (3): The intrinsic and extrinsic pathways leading to apoptosis. 22 Figure (4): Structure of the 26S proteasome. 37 Figure (5): Chemical Structure of Bortezomib 38 Figure (6): Mechanism of action of Bortezomib 40 Figure (7): The different ways that Bortezomib can affect multiple myeloma 41 Figure (8): Antitumor actions of interferons.
    [Show full text]
  • Velcade, INN-Bortezomib
    SCIENTIFIC DISCUSSION This module reflects the initial scientific discussion for the approval of Velcade. This scientific discussion has been updated until 1 May 2004. For information on changes after this date please refer to module 8. 1. Introduction Multiple myeloma Multiple myeloma is a B-cell malignancy of the plasma cell and represents the second most common hematological malignancy, with non-Hodgkin’s lymphoma being the most common. The incidence and prevalence of multiple myeloma are similar in the US and Europe. In 2000, there were approximately 19,000 cases diagnosed in the European Economic Area (Globocan, 2000), with a 5- year prevalence of approximately 47,000 cases. The course of multiple myeloma is characterized by an asymptomatic or subclinical phase before diagnosis (possibly for several years), a chronic phase lasting several years, and an aggressive terminal phase. Multiple myeloma leads to progressive morbidity and eventual mortality by lowering resistance to infection and causing significant skeletal destruction (with bone pain, pathological fractures, and hypercalcemia), anemia, renal failure, and, less commonly, neurological complications and hyperviscosity. From the time of diagnosis, the survival without treatment is between 6 to 12 months and extends to 3 years with chemotherapy. Approximately 25% of patients survive 5 years or longer, with fewer than 5% surviving longer than 10 years. Approved anticancer agents for the treatment of myeloma in the US include melphalan (1992), carmustine (BCNU, 1977) and cyclophosphamide (1959) and in addition, in Europe, epirubicin is also approved for treatment of this disease. At the time of diagnosis, multiple myeloma is a heterogeneous disease, with a course that varies on the basis of both disease- and host-related factors (e.g., age, renal function, stage, alpha2- microglobulin, chromosomal abnormalities).
    [Show full text]
  • XELODA (Capecitabine) Is a Fluoropyrimidine Carbamate with Antineoplastic Activity
    XELODA® (capecitabine) TABLETS DESCRIPTION: XELODA (capecitabine) is a fluoropyrimidine carbamate with antineoplastic activity. It is an orally administered systemic prodrug of 5’-deoxy-5-fluorouridine (5’-DFUR) which is converted to 5-fluorouracil. The chemical name for capecitabine is 5’-deoxy-5-fluoro-N-[(pentyloxy) carbonyl]-cytidine and has a molecular weight of 359.35. Capecitabine has the following structural formula: O N NH O O O H3C N F HO OH Capecitabine is a white to off-white crystalline powder with an aqueous solubility of 26 mg/mL at 20ºC. XELODA is supplied as biconvex, oblong film-coated tablets for oral administration. Each light peach- colored tablet contains 150 mg capecitabine and each peach-colored tablet contains 500 mg capecitabine. The inactive ingredients in XELODA include: anhydrous lactose, croscarmellose sodium, hydroxypropyl methylcellulose, microcrystalline cellulose, magnesium stearate and purified water. The peach or light peach film coating contains hydroxypropyl methylcellulose, talc, titanium dioxide, and synthetic yellow and red iron oxides. CLINICAL PHARMACOLOGY: Capecitabine is relatively non-cytotoxic in vitro. This drug is enzymatically converted to 5-fluorouracil (5-FU) in vivo. Bioactivation: Capecitabine is readily absorbed from the gastrointestinal tract. In the liver, a 60 kDa carboxyesterase hydrolyzes much of the compound to 5’-deoxy-5-fluorocytidine (5’-DFCR). Cytidine deaminase, an enzyme found in most tissues, including tumors, subsequently converts 5’-DFCR to 5’- deoxy-5-fluorouridine (5’-DFUR). The enzyme, thymidine phosphorylase (dThdPase), then hydrolyzes 5’-DFUR to the active drug 5-FU. Many tissues throughout the body express thymidine phosphorylase. Some human carcinomas express this enzyme in higher concentrations than surrounding normal tissues.
    [Show full text]