Mitochondria in Hematopoiesis and Hematological Diseases

Total Page:16

File Type:pdf, Size:1020Kb

Mitochondria in Hematopoiesis and Hematological Diseases Oncogene (2006) 25, 4757–4767 & 2006 Nature Publishing Group All rights reserved 0950-9232/06 $30.00 www.nature.com/onc REVIEW Mitochondria in hematopoiesis and hematological diseases M Fontenay1, S Cathelin2, M Amiot3, E Gyan1 and E Solary2 1Inserm U567, Institut Cochin, Department of Hematology, Paris, Cedex, France; 2Inserm U601, Biology Institute, Nantes, Cedex, France and 3Inserm U517, Faculty of Medicine, Dijon, France Mitochondria are involved in hematopoietic cell homeo- Introduction stasis through multiple ways such as oxidative phosphor- ylation, various metabolic processes and the release of As in other tissues, mitochondria play many important cytochrome c in the cytosol to trigger caspase activation roles in hematopoietic cell homeostasis, including the and cell death. In erythroid cells, the mitochondrial steps production of adenosine triphosphate (ATP) by the in heme synthesis, iron (Fe) metabolism and Fe-sulfur process of oxidative phosphorylation, the release of (Fe-S) cluster biogenesis are of particular importance. death-promoting factors upon apoptotic stimuli and a Mutations in the specific d-aminolevulinic acid synthase variety of metabolic pathways such as heme synthesis. (ALAS) 2 isoform that catalyses the first and rate-limiting Mitochondria could also play a role in specific pathways step in heme synthesis pathway in the mitochondrial of hematopoietic cell differentiation through caspase matrix, lead to ineffective erythropoiesis that charac- activation.Alterations of these mitochondrial functions terizes X-linked sideroblastic anemia (XLSA), the most play a pathophysiological role in a variety of hemato- common inherited sideroblastic anemia. Mutations in the logical diseases, especially those affecting erythroid adenosine triphosphate-binding cassette protein ABCB7, lineage such as inherited dyserythropoiesis and side- identified in XLSA with ataxia (XLSA-A), disrupt the roblastic anemias and acquired, low-grade myelodys- maturation of cytosolic (Fe-S) clusters, leading to plastic syndromes.In these diseases, compromised mitochondrial Fe accumulation. In addition, large dele- mitochondrial functions could be related to mitochon- tions in mitochondrial DNA, whose integrity depends on a drial genome mutations, alterations in iron (Fe) specific DNA polymerase, are the hallmark of Pearson’s mitochondrial metabolism or exacerbation of physio- syndrome, a rare congenital disorder with sideroblastic logical pathways involving caspases, leading to activa- anemia. In acquired myelodysplastic syndromes at early tion of the mitochondrial pathway to apoptotic cell stage, exacerbation of physiological pathways involving death.Mitochondria also plays a central role in the caspases and the mitochondria in erythroid differentiation apoptotic response of transformed hematopoietic cells leads to abnormal activation of a mitochondria-mediated when exposed to cytotoxic drugs, although oncogenesis- apoptotic cell death pathway. In contrast, oncogenesis- associated changes at the mitochondrial level can induce associated changes at the mitochondrial level can alter the resistance to apoptosis induced by a variety of these apoptotic response of transformed hematopoietic cells to drugs.Owing to their central role in cell death, chemotherapeutic agents. Recent findings in mitochondria mitochondria now appear as a potential target for metabolism and functions open new perspectives in killing malignant cells.This review will evoke several treating hematopoietic cell diseases, for example various examples in which mitochondria play a pathophysio- compounds currently developed to trigger tumor cell death logical role in hematopoietic diseases, the role of these by directly targeting the mitochondria could prove organelles in hematopoietic cell differentiation and efficient as either cytotoxic drugs or chemosensitizing response to cytotoxic agents and the various strategies agents in treating hematological malignancies. currently tested to use mitochondria as a target to treat Oncogene (2006) 25, 4757–4767. doi:10.1038/sj.onc.1209606 malignant hematopoietic diseases. Keywords: mitochondria; sideroblastic anemia; leukemia; myelodysplastic syndromes; apoptosis Mitochondrial DNA mutations in hematopoietic cell aging and diseases Together with the nucleus, mitochondria are the only organelles of animal cells that contain DNA as well as their own machinery for RNA and protein synthesis. Mitochondrial DNA (mtDNA) is a double-stranded circular molecule present as 2–10 copies in each Correspondence: Dr E Solary, UMR Inserm 517, Faculty of Medicine, mitochondrion.Cells contain thousands of mtDNA mole- 7, boulevard Jeanne d’Arc, Dijon 21000, France. cules that distinguish from nuclear DNA (nDNA) by E-mail: [email protected] several characteristics, including maternal inheritance, Mitochondria in hematopoiesis and hematological diseases M Fontenay et al 4758 a modified genetic code, a paucity of introns, the lack of Table 1 Main genetic hematological diseases related to mitochondria histone protection and the limited efficacy of the repair alterations system.These two later characteristics, together with its Disease Genetic alterations proximity to radical oxygen species (ROS) generation due to oxidative phosphorylation for ATP production, Pearson’s syndrome Large deletions in mtDNA X-linked sideroblastic anemia ALAS2 may account to the 10- to 20-fold higher mutation rate X-linked sideroblastic anemia ABCB7 gene mutation (Atm1p in mtDNA compared with nDNA (Yakes and Van with ataxia in yeast) Houten, 1997).In addition, multiple species of mtDNA Friedreich ataxia Frataxin gene mutations can coexist in a single cell, a condition called hetero- Kostmann’s syndrome Decreased Bcl-2 expression plasmy. Animal models of sideroblastic Flexed-tail mutation, SOD2 anemia deletion Somatically acquired mtDNA mutations are attribu- table to mitochondrial oxidative damage, which in- Abbreviations: mtDNA: mitochondrial DNA; ALAS: d-amino- creases with age.Single mutant mtDNA molecules have levulinic acid synthase 2; SOD2: superoxide dismutase-2. long been assumed to be lost by dilution in rapidly dividing tissues such as bone marrow.Actually, age- dependent accumulation of mtDNA mutations, and expansion of mutated mtDNA molecules from hetero- plasmy to homoplasmy (Chinnery et al., 2002), appear to be relatively common in adult CD34 þ marrow progenitors and presumably stem cells (Ogasawara et al., 2005). Thus, age-associated mtDNA mutations lead to marked heterogeneity among adult bone marrow and peripheral blood CD34 þ cells, not present in umbilical cord blood.Mitochondrial DNA variant sequences were proposed as natural genetic markers for determining the contribution of individual stem cells Figure 1 Ring sideroblasts.( a) Perls staining showing a ring to blood production.In addition, mtDNA changes fixed sideroblast due to iron (Fe) overload in the mitochondria forming a ring around the nucleus in basophilic and polychromatophilic in leukemic cells might provide a simple method for erythroblasts.( b and c) Electron microscopy analysis of apoptotic monitoring minimal residual disease (He et al., 2003). ring sideroblasts (mi: mitochondrion). Integrity of mtDNA involves a DNA polymerase whose catalytic subunit is encoded by a nuclear gene. Knock-in mice that express a proof-reading-deficient mutations were identified in the genes encoding the version of the catalytic subunit of this polymerase develop mitochondrial tRNA for leucine, the subunit I of a MtDNA mutator phenotype with a significant increase cytochrome c oxidase and the cytochrome b (Gattermann in the level of point mutations, as well as increased et al., 1996, 1997; Reddy et al., 2002). The link between amounts of deletions.These events are associated with these mutations and the disease remains highly con- reduced lifespan and premature onset of aging-related troversial as a comparative study did not demonstrate an symptoms, including anemia (Trifunovic et al., 2004). increase in the frequency of mtDNA mutations in a large Interestingly, the increase in mtDNA mutations observed cohort of myelodysplastic syndrome patients compared in these mice does not depend on an increase in oxidative to age-matched normal individuals (Shin et al., 2003). phosphorylation (Kujoth et al.,2005). A current speculation is that age-related mtDNA Lesions in mtDNA and mitochondrial dysfunction mutations in CD34 þ clones could lead to derangement contribute to numerous human diseases.Large deletions of marrow function in older individuals (Shin et al., in mtDNA are the hallmark of Pearson’s syndrome, a 2004).Whether these mutations are associated with rare congenital disorder with lactic acidosis, pancreatic abnormal oxidative phosphorylation measured in mye- insufficiency and sideroblastic anemia (Pearson et al., lodysplastic syndrome bone-marrow samples is a matter 1979; Rotig et al., 1991) (Table 1). Sideroblastic anemias of discussion (Bowen and Peddie, 2002; Matthes et al., are characterized by Fe overload in the mitochondria 2002).At least in older patients, they could also play a that are revealed by Perls staining as a ring around the role in excessive apoptosis that characterizes low-grade nucleus in basophilic and polychromatophilic erythro- myelodysplastic syndromes (Bouscary et al., 1997) as blasts (Figure 1).Transmission electron microscopy mice with defective mtDNA polymerase exhibit apop- analysis of bone-marrow cells in infants with inherited tosis in excess in several rapidly growing tissues (Kujoth sideroblastic
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]
  • (12) United States Patent (10) Patent No.: US 8,580,814 B2 Adelman Et Al
    USOO858O814B2 (12) United States Patent (10) Patent No.: US 8,580,814 B2 Adelman et al. (45) Date of Patent: *Nov. 12, 2013 (54) METHODS OF USING 5,528,823. A 6/1996 Rudy, Jr. et al. (+)-1,4-DIHYDRO-7-(3S4S)-3-METHOXY-4- 3.43wk A E. E. St.Ola tala (METHYLAMINO)-1-PYRROLIDINYL-4- 6,171,857 B1 1/2001 Hendrickson OXO-1-(2-THIAZOLYL)-1.8- 6,291643 B1 9/2001 Zou et al. NAPHTHYRIDNE-3-CARBOXYLIC ACID 6,570,002 B1 5/2003 Hardwicket al. FORTREATMENT OF CANCER 6,641,810 B2 11/2003 Gold 6,641,833 B2 * 1 1/2003 Dang ............................ 424/426 (75) Inventors: Daniel CAdelman, Redwood City, CA 6,696,4836,670,144 B2B1 12/20032/2004 CraigSingh et al. (US); Jeffrey A. Silverman, 6,723,734 B2 4/2004 Kim et al. Burlingame, CA (US) 7,211,562 B2 5/2007 Rosen et al. 7.989,468 B2 * 8/2011 Adelman et al. .............. 514,300 (73) Assignee: Sunesis Pharmaceuticals, Inc., South 3.9. A. S. idaran-Ghera Gh et al.1 San Francisco, CA (US) 2003/0232334 A1* 12/2003 Morris et al. ..................... 435/6 c - r 2004/0106605 A1 6/2004 Carboni et al. .... 514/226.8 (*) Notice: Subject to any disclaimer, the term of this 2004/0132825 A1* 7/2004 Bacopoulos et al. ......... 514/575 patent is extended or adjusted under 35 2005/0203120 A1 9, 2005 Adelman et al. U.S.C. 154(b) by 201 days. 2005/0215583 A1 9, 2005 Arkin et al. 2006, OO25437 A1 2/2006 Adelman et al.
    [Show full text]
  • Preclinical Pharmacologic Evaluation of Pralatrexate and Romidepsin
    Published OnlineFirst February 12, 2015; DOI: 10.1158/1078-0432.CCR-14-2249 Cancer Therapy: Preclinical Clinical Cancer Research Preclinical Pharmacologic Evaluation of Pralatrexate and Romidepsin Confirms Potent Synergy of the Combination in a Murine Model of Human T-cell Lymphoma Salvia Jain1, Xavier Jirau-Serrano2, Kelly M. Zullo2, Luigi Scotto2, Carmine F. Palermo3,4, Stephen A. Sastra3,4,5, Kenneth P. Olive3,4,5, Serge Cremers3, Tiffany Thomas3,YingWei6, Yuan Zhang6, Govind Bhagat3, Jennifer E. Amengual2, Changchun Deng2, Charles Karan8, Ronald Realubit8, Susan E. Bates9, and Owen A. O'Connor2 Abstract Purpose: T-cell lymphomas (TCL) are aggressive diseases, NOG mouse model of TCL were used to explore the in vitro and in which carry a poor prognosis. The emergence of new drugs for vivo activity of pralatrexate and romidepsin in combination. TCL has created a need to survey these agents in a rapid and Corresponding mass spectrometry–based pharmacokinetic and reproducible fashion, to prioritize combinations which should be immunohistochemistry-based pharmacodynamic analyses of prioritized for clinical study. Mouse models of TCL that can be xenograft tumors were performed to better understand a mech- used for screening novel agents and their combinations are anistic basis for the drug:drug interaction. lacking. Developments in noninvasive imaging modalities, such Results: In vitro, pralatrexate and romidepsin exhibited con- as surface bioluminescence (SBL) and three-dimensional ultra- centration-dependent synergism in combination against a panel sound (3D-US), are challenging conventional approaches in of TCL cell lines. In a NOG murine model of TCL, the combination xenograft modeling relying on caliper measurements. The recent of pralatrexate and romidepsin exhibited enhanced efficacy com- approval of pralatrexate and romidepsin creates an obvious pared with either drug alone across a spectrum of tumors using combination that could produce meaningful activity in TCL, complementary imaging modalities, such as SBL and 3D-US.
    [Show full text]
  • The Ferritin Fe2 Site at the Diiron Catalytic Center Controls the Reaction with O2 in the Rapid Mineralization Pathway
    The ferritin Fe2 site at the diiron catalytic center controls the reaction with O2 in the rapid mineralization pathway Takehiko Toshaa, Mohammad R. Hasana,1, and Elizabeth C. Theila,b,2 aCouncil on BioIron at Children’s Hospital Oakland Research Institute, 5700 Martin Luther King, Jr., Way, Oakland, CA 94609; and bDepartment of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720 Edited by Edward I. Solomon, Stanford University, Stanford, CA, and approved October 13, 2008 (received for review June 2, 2008) Oxidoreduction in ferritin protein nanocages occurs at sites that Oxidoreductase/ferroxidase (Fox) catalytic centers with difer- bind two Fe(II) substrate ions and O2, releasing Fe(III)2–O products, rous binding sites, Fe1 and Fe2, occur in each ferritin subunit the biomineral precursors. Diferric peroxo intermediates form in except in animals, where a second gene encodes a catalytically ferritins and in the related diiron cofactor oxygenases. Cofactor inactive subunit (ferritin L) that co-assembles with the active iron is retained at diiron sites throughout catalysis, contrasting subunits in various ratios that depend on the tissue. The ferritin with ferritin. Four of the 6 active site residues are the same in oxidoreductase sites share properties with diiron cofactor cata- ferritins and diiron oxygenases; ferritin-specific Gln137 and variable lysts that include the first detectable reaction intermediate in Asp/Ser/Ala140 substitute for Glu and His, respectively, in diiron ferritin, a diferric peroxo (DFP) complex, characterized by cofactor active sites. To understand the selective functions of UV/visible (UV/vis), resonance Raman, Mo¨ssbauer, and ex- diiron substrate and diiron cofactor active site residues, we com- tended X-ray absorption fine structure (EXAFS) spectroscopies pared oxidoreductase activity in ferritin with diiron cofactor resi- (8–14).
    [Show full text]
  • Aptamers and Antisense Oligonucleotides for Diagnosis and Treatment of Hematological Diseases
    International Journal of Molecular Sciences Review Aptamers and Antisense Oligonucleotides for Diagnosis and Treatment of Hematological Diseases Valentina Giudice 1,2,* , Francesca Mensitieri 1, Viviana Izzo 1,2 , Amelia Filippelli 1,2 and Carmine Selleri 1 1 Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; [email protected] (F.M.); [email protected] (V.I.); afi[email protected] (A.F.); [email protected] (C.S.) 2 Unit of Clinical Pharmacology, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy * Correspondence: [email protected]; Tel.: +39-(0)-89965116 Received: 30 March 2020; Accepted: 2 May 2020; Published: 4 May 2020 Abstract: Aptamers or chemical antibodies are single-stranded DNA or RNA oligonucleotides that bind proteins and small molecules with high affinity and specificity by recognizing tertiary or quaternary structures as antibodies. Aptamers can be easily produced in vitro through a process known as systemic evolution of ligands by exponential enrichment (SELEX) or a cell-based SELEX procedure. Aptamers and modified aptamers, such as slow, off-rate, modified aptamers (SOMAmers), can bind to target molecules with less polar and more hydrophobic interactions showing slower dissociation rates, higher stability, and resistance to nuclease degradation. Aptamers and SOMAmers are largely employed for multiplex high-throughput proteomics analysis with high reproducibility and reliability, for tumor cell detection by flow cytometry or microscopy for research and clinical purposes. In addition, aptamers are increasingly used for novel drug delivery systems specifically targeting tumor cells, and as new anticancer molecules. In this review, we summarize current preclinical and clinical applications of aptamers in malignant and non-malignant hematological diseases.
    [Show full text]
  • The Construction and Characterization of Mitochondrial Ferritin Overexpressing Mice
    Article The Construction and Characterization of Mitochondrial Ferritin Overexpressing Mice Xin Li 1,†, Peina Wang 1,†, Qiong Wu 1, Lide Xie 2, Yanmei Cui 1, Haiyan Li 1, Peng Yu 1 and Yan-Zhong Chang 1,* 1 Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China; [email protected] (X.L.); [email protected] (P.W.); [email protected] (Q.W.); [email protected] (Y.C.); [email protected] (H.L.); [email protected] (P.Y.) 2 Department of Biomedical Engineering, Chengde Medical University, Chengde 067000, China; [email protected] * Correspondence: [email protected]; Tel.: +86-311-8078-6311 † These authors contributed equally to this work. Received: 31 May 2017; Accepted: 10 July 2017; Published: 13 July 2017 Abstract: Mitochondrial ferritin (FtMt) is a H-ferritin-like protein which localizes to mitochondria. Previous studies have shown that this protein can protect mitochondria from iron-induced oxidative damage, while FtMt overexpression in cultured cells decreases cytosolic iron availability and protects against oxidative damage. To investigate the in vivo role of FtMt, we established FtMt overexpressing mice by pro-nucleus microinjection and examined the characteristics of the animals. We first confirmed that the protein levels of FtMt in the transgenic mice were increased compared to wild-type mice. Interestingly, we found no significant differences in the body weights or organ to body weight ratios between wild type and transgenic mice. To determine the effects of FtMt overexpression on baseline murine iron metabolism and hematological indices, we measured serum, heart, liver, spleen, kidney, testis, and brain iron concentrations, liver hepcidin expression and red blood cell parameters.
    [Show full text]
  • The Potential for Transition Metal-Mediated Neurodegeneration in Amyotrophic Lateral Sclerosis
    REVIEW ARTICLE published: 23 July 2014 AGING NEUROSCIENCE doi: 10.3389/fnagi.2014.00173 The potential for transition metal-mediated neurodegeneration in amyotrophic lateral sclerosis David B. Lovejoy* and Gilles J. Guillemin Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia Edited by: Modulations of the potentially toxic transition metals iron (Fe) and copper (Cu) are impli- Roger S. Chung, Macquarie cated in the neurodegenerative process in a variety of human disease states including University, USA amyotrophic lateral sclerosis (ALS). However, the precise role played by these metals is Reviewed by: Junming Wang, University of still very much unclear, despite considerable clinical and experimental data suggestive of Mississippi Medical Center, USA a role for these elements in the neurodegenerative process.The discovery of mutations in Ramon Santos El-Bachá, Universidade the antioxidant enzyme Cu/Zn superoxide dismutase 1 (SOD-1) in ALS patients established Federal da Bahia, Brazil the first known cause of ALS. Recent data suggest that various mutations in SOD-1 affect *Correspondence: metal-binding of Cu and Zn, in turn promoting toxic protein aggregation. Copper home- David B. Lovejoy, Macquarie University, Australian School of ostasis is also disturbed in ALS, and may be relevant to ALS pathogenesis. Another set Advanced Medicine, Motor Neuron of interesting observations in ALS patients involves the key nutrient Fe. In ALS patients, and Neurodegenerative Diseases Fe loading can be inferred by studies showing increased expression of serum ferritin, an Research Group, Building F10A, 2 Fe-storage protein, with high serum ferritin levels correlating to poor prognosis. Magnetic Technology Place, NSW, 2109, Australia resonance imaging of ALS patients shows a characteristic T2 shortening that is attributed e-mail: [email protected] to the presence of Fe in the motor cortex.
    [Show full text]
  • Venetoclax for the Treatment of Mantle Cell Lymphoma
    Review Article Page 1 of 11 Venetoclax for the treatment of mantle cell lymphoma Victor S. Lin1, Mary Ann Anderson2,3, David C. S. Huang3, Andrew W. Roberts1,2,3, John F. Seymour1,2, Constantine S. L. Tam1,2,4 1Department of Medicine, The University of Melbourne, Parkville, Australia; 2Department of Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, Australia; 3Division of Cancer and Haematology, The Walter and Eliza Hall Institute, Parkville, Australia; 4Department of Haematology, St Vincent’s Hospital, Fitzroy, Australia Contributions: (I) Conception and design: All authors; (II) Administrative support: None; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: None; (V) Data analysis and interpretation: None; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors. Correspondence to: Mary Ann Anderson. 1G, Royal Parade, Parkville, 3052 Victoria, Australia. Email: [email protected]. Abstract: While the last decade has seen an explosion in improved therapies for mantle cell lymphoma (MCL), the outlook for patients with relapsed or refractory MCL (R/R MCL) remains poor, especially for those who are older with comorbidities or harbour dysfunction in the TP53 pathway. MCL is one of the B cell malignancies in which there is a high frequency of BCL2 overexpression, and the selective BCL2 inhibitor venetoclax has shown particular promise in the treatment of patients with R/R MCL. As a single agent, the drug is associated with a close to 80% response rate in early phase studies. However, secondary progression due to the development of resistance remains a limiting factor in the usefulness of this agent.
    [Show full text]
  • Original Article Doi:10.1093/Annonc/Mdn280 Published Online 13 May 2008
    Annals of Oncology 19: 1698–1705, 2008 original article doi:10.1093/annonc/mdn280 Published online 13 May 2008 Oblimersen combined with docetaxel, adriamycin and cyclophosphamide as neo-adjuvant systemic treatment in primary breast cancer: final results of a multicentric phase I study J. Rom1*, G. von Minckwitz2,5, W. Eiermann3, M. Sievert4, B. Schlehe1, F. Marme´ 1, F. Schuetz1, A. Scharf1, M. Eichbaum1, H.-P. Sinn5, M. Kaufmann6, C. Sohn1 & A. Schneeweiss1 1Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg; 2German Breast Group, Neu Isenburg; 3Frauenklinik vom Roten Kreuz, Mu¨nchen; 4Sanofi-Aventis GmbH, Berlin; 5Department of Pathology, University of Heidelberg, Heidelberg; 6University of Frankfurt, Department of Gynecology and Obstetrics, Frankfurt, Germany Downloaded from Received 23 January 2008; revised 6 April 2008; accepted 7 April 2008 Background: Combining the Bcl-2 down-regulator oblimersen with cytotoxic treatment leads to synergistic antitumor effects in preclinical trials. This multicentric phase I study was carried out to evaluate maximum tolerated dose (MTD), safety and preliminary efficacy of oblimersen in combination with docetaxel, adriamycin and annonc.oxfordjournals.org cyclophosphamide as neo-adjuvant systemic treatment (NST) in primary breast cancer (PBC). Methods: Previously untreated patients with PBC T2–4a–c N0–3 M0 received one cycle of docetaxel 75 mg/m2, adriamycin 50 mg/m2 and cyclophosphamide 500 mg/m2 administered on day 5 combined with escalating doses of oblimersen as a 24-h continuous infusion on days 1–7 followed by five cycles of combination of docetaxel, adriamycin and cyclophosphamide (TAC) without oblimersen every 3 weeks. Prophylactic antibiotic therapy and granulocyte article original colony-stimulating factor administration were used in all six cycles.
    [Show full text]
  • Frataxin and Mitochondrial Fes Cluster Biogenesis Timothy L
    Wayne State University DigitalCommons@WayneState Biochemistry and Molecular Biology Faculty Department of Biochemistry and Molecular Biology Publications 8-27-2010 Frataxin and Mitochondrial FeS Cluster Biogenesis Timothy L. Stemmler Wayne State University, [email protected] Emmanuel Lesuisse Institut Jacques Monod Debumar Pain University of Medicine and Dentistry, New Jersey Andrew Dancis University of Pennsylvania Recommended Citation Stemmler, T. L., Lesuisse, E., Pain, D., and Dancis, A. (2010) J. Biol. Chem. 285, 26737-26743. doi:10.1074/jbc.R110.118679 Available at: http://digitalcommons.wayne.edu/med_biochem/13 This Article is brought to you for free and open access by the Department of Biochemistry and Molecular Biology at DigitalCommons@WayneState. It has been accepted for inclusion in Biochemistry and Molecular Biology Faculty Publications by an authorized administrator of DigitalCommons@WayneState. This is the author's post-print version, previously appearing in the Journal of Biological Chemistry , 2010, 285, 35, 26737-43. Available online at: http://www.jbc.org/ Frataxin and mitochondrial Fe-S cluster biogenesis | Timothy Stemmler, et. al Frataxin and mitochondrial Fe-S cluster biogenesis Timothy L. Stemmler 1, Emmanuel Lesuisse 2, Debumar Pain 3, Andrew Dancis 4 1. Department of Biochemistry and Molecular Biology, Wayne State University, School of Medicine, Detroit, Michigan 48201 2. Laboratoire Mitochondrie, Metaux et Stress Oxydant, Institut Jacques Monod, CNRS-Universite Paris Diderot, France 75205 3. Department of Pharmacology and Physiology, UMDNJ, New Jersey Medical School, Newark, New Jersey 07101 4. Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, Pennsylvania 19104 Abstract Friedreich’s ataxia is an inherited neurodegenerative disease caused by frataxin deficiency.
    [Show full text]
  • BCL-2 Inhibitors for Chronic Lymphocytic Leukemia
    L al of euk rn em u i o a J Robak, J Leuk 2015, 3:3 Journal of Leukemia DOI: 10.4172/2329-6917.1000e114 ISSN: 2329-6917 Editorial Open access BCL-2 inhibitors for Chronic Lymphocytic Leukemia Tadeusz Robak* Department of Hematology, Medical University of Lodz, 93-510 Lodz, ul, Ciolkowskiego 2, Poland *Corresponding author: Tadeusz Robak, Department of Hematology, Medical University of Lodz, Copernicus Memorial Hospital, 93-510 Lodz, Ul. Ciolkowskiego 2, Poland, Tel: +48 42 6895191; E-mail: [email protected] Rec date: August 10, 2015; Acc date: August 12, 2015; Pub date: August 24, 2015 Copyright: © 2015 Robak T. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Editorial cyclophosphamide and rituximab (FCR) or bendamustin-rituximab (BR) regimens in refractory CLL are ongoing (NCT00868413). B-cell chronic lymphocytic leukemia (CLL) is the most common adult leukemia in Europe and North America, with an annual Venetoclax ( GDC-0199, ABT-199, RG7601; Genentech, South San incidence of three to five cases per 100,000 [1]. The approval of Francisco, CA), is a first-in-class, orally bioavailable BCL-2 family rituximab-based immunochemotherapy represented a substantial protein inhibitor that binds with high affinity to BCL-2 and with lower therapeutic advance in CLL [2]. However, currently available therapies affinity to the other BCL-2 family proteins Bcl-xL, Bcl-w and Mcl-1 are only partially efficient, and there is an obvious need to develop [14].
    [Show full text]
  • Nanocarriers As Magic Bullets in the Treatment of Leukemia
    nanomaterials Review Nanocarriers as Magic Bullets in the Treatment of Leukemia 1, 2, 1 2 Mohammad Houshmand y , Francesca Garello y , Paola Circosta , Rachele Stefania , Silvio Aime 2, Giuseppe Saglio 1 and Claudia Giachino 1,* 1 Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy; [email protected] (M.H.); [email protected] (P.C.); [email protected] (G.S.) 2 Molecular and Preclinical Imaging Centres, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; [email protected] (F.G.); [email protected] (R.S.); [email protected] (S.A.) * Correspondence: [email protected] These authors contributed equally to this work. y Received: 23 December 2019; Accepted: 1 February 2020; Published: 6 February 2020 Abstract: Leukemia is a type of hematopoietic stem/progenitor cell malignancy characterized by the accumulation of immature cells in the blood and bone marrow. Treatment strategies mainly rely on the administration of chemotherapeutic agents, which, unfortunately, are known for their high toxicity and side effects. The concept of targeted therapy as magic bullet was introduced by Paul Erlich about 100 years ago, to inspire new therapies able to tackle the disadvantages of chemotherapeutic agents. Currently, nanoparticles are considered viable options in the treatment of different types of cancer, including leukemia. The main advantages associated with the use of these nanocarriers summarized as follows: i) they may be designed to target leukemic cells selectively; ii) they invariably enhance bioavailability and blood circulation half-life; iii) their mode of action is expected to reduce side effects.
    [Show full text]