New Insights Into Molecular Mechanisms of Sunitinib-Associated Side Effects

Total Page:16

File Type:pdf, Size:1020Kb

New Insights Into Molecular Mechanisms of Sunitinib-Associated Side Effects Molecular Cancer Review Therapeutics New Insights into Molecular Mechanisms of Sunitinib-Associated Side Effects Guadalupe Aparicio-Gallego1, Moises Blanco1, Angelica Figueroa1, Rosario García-Campelo1, Manuel Valladares-Ayerbes1, Enrique Grande-Pulido2, and Luis Anton-Aparicio 1 Abstract The introduction of targeted therapy represents a major advance in the treatment of tumor progression. Targeted agents are a novel therapeutic approach developed to disrupt different cellular signaling pathways. The tyrosine kinase inhibitor sunitinib specifically blocks multiple tyrosine kinase receptors that are involved in the progression of many tumors. Sunitinib is the current standard of care in first-line treatment of advanced renal cell carcinoma, and it is approved in imatinib-intolerant and imatinib-refractory gastrointestinal stromal tumors. However, it is increasingly evident that sunitinib may display collateral effects on other proteins beyond its main target receptors, eliciting undesirable and unexpected adverse events. A better understanding of the molecular mechanisms underlying these undesirable sunitinib-associated side effects will help physicians to maximize efficacy of sunitinib and minimize adverse events. Here, we focus on new insights into molecular mechanisms that may mediate sunitinib-associated adverse events. Mol Cancer Ther; 10(12); 2215–23. Ó2011 AACR. Introduction 2 (KDR), and 3 (FLT4); platelet-derived growth factor receptors A and B (PDGFRA and PDGFRB); the stem cell Tyrosine kinase inhibitors (TKI) are a novel type of factor receptor (cKIT); FMS-like tyrosine kinase 3 (FLT3); drug, designed to target signaling pathways deregulated colony-stimulating factor 1 receptor (CSF-1R); and glial cell in cancer by blocking the ATP-binding pocket of specific line–derived neurotrophic factor receptor [RE arranged receptor tyrosine kinases (RTK). TKIs compete with during transfection (RET); refs. 1–3]. The inhibition of ATP for binding to the intracellular domain of wild-type these RTKs blocks signal transduction, thereby affecting and/or mutant forms of RTKs and, consequently, inhibit various cellular processes, such as tumor growth, tumor signal transduction. progression, angiogenesis, and metastasis (Fig. 1; ref. 4). Although TKIs are designed to target specific RTKs, it is In a phase III study in treatment-na€ve patients increasingly apparent that many TKIs may display unex- with metastatic renal cell carcinoma (RCC), sunitinib pected effects on other kinases and may trigger the significantly doubled median progression-free survival appearance of undesirable side effects. The observed compared with IFN-a and extended median overall sur- incidence of side effects associated with TKIs varies vival beyond 2 years (5, 6). Sunitinib is approved for the between clinical studies, depending on treatment sched- first-line treatment of advanced or metastatic RCC and ule, dosage, patient characteristics, predisposing factors, imatinib-resistant or imatinib-intolerant advanced gastro- secondary diseases, and concurrent administration of intestinal stromal tumors (GIST). other drugs. Moreover, these agents, which do not spe- Although sunitinib therapy is associated with several cifically target tumor cells, are able to act on normal cells, well-characterized side effects, the molecular mechan- eliciting diverse side effects. isms underlying these toxicities remain unclear, but Sunitinib (sunitinib malate; Sutent; Pfizer Inc.) is a they may be related to the additional effect of sunitinib targeted TKI able to inhibit members of the RTK families on nonmalignant (normal) cells that also express containing a split-kinase domain (Fig. 1; ref. 1). These sunitinib targets, or to the cross-talk between different families include VEGF receptor (VEGFR) types 1 (FLT1), intracellular signaling pathways. In this review, we exam- ine the current knowledge about the molecular basis of Authors' Affiliations: 1Clinical Oncology Department and Biomedical sunitinib side effects, including asthenia, fatigue, hair Research Institute (INIBIC), A Coruna~ University Hospital, A Coruna;~ depigmentation, cardiotoxicity, hypothyroidism, hyper- 2 Clinical Oncology Department, Ramon y Cajal University Hospital, Madrid, tension, dermatologic adverse events, and hematologic Spain and gastrointestinal toxicities. Corresponding Author: Luis Anton-Aparicio, Clinical Oncology Depart- ment, A Coruna~ University Hospital, As Xubias 84, 15006 A Coruna,~ Spain. Phone: 34-981-178000; Fax: 34-981-178273; E-mail: Molecular Basis of Fatigue and Asthenia [email protected] doi: 10.1158/1535-7163.MCT-10-1124 Fatigue and asthenia are among the most common Ó2011 American Association for Cancer Research. symptoms experienced by cancer patients, comprising www.aacrjournals.org 2215 Downloaded from mct.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Aparicio-Gallego et al. VEGFR1 VEGFR2 VEGFR3 PDGFRα PDGFRβ KIT FLT3 RET (FLT1) (KDR) (FLT4) CH3 CH3 N CH3 O CH3 H3C N N Sunitinib malate Sunitinib malate H O F N CH H3C N H 3 H O N F N CH H H 3 O PI3K/AKT/mTOR RAS/MAPK/MAPKK PKCs N H Antitumor effects Direct effects Antiangiogenic Vascular on cancer cells effects targeting Figure 1. Specific RTKs are blocked by sunitinib (chemical structure shown). Sunitinib is able to block different signaling pathways owing to its action on different RTKs such as PDGFRa and b, KIT, FLT3, RET, and VEGFRs 1, 2, and 3. Sunitinib inhibition of signaling pathways [phosphoinositide 3-kinase (PI3K)/AKT/mTOR, mitogen-activated protein kinase (MAPK), and PKC] triggers different antitumor effects (4). pathologic tiredness, muscle weakness, poor endurance, processes within the cell. As such, AMPK functions as and impaired motor and cognitive function. Fatigue is one an energy sensor protein that is activated in response of the most commonly reported side effects of sunitinib to an increased cellular AMP-to-ATP ratio, occurr- treatment, although the degree of fatigue and its impact on ing, for example, during nutrient starvation (pooled quality of life is variable (7). In sunitinib phase III clinical data in sunitinib-treated RCC show that grade 3–4 trials, 34% to 62% of patients exhibited fatigue of any grade, anorexia occurs in approximately 3% of patients; ref. 9) with 5% to 15% reporting grade 3 to 4 fatigue. Asthenia was or vigorous muscular exercise. The serine-threonine less common, with an all-grade frequency of 12% to 25% kinase LKB1 is an important component of the AMPK and grade 3 to 4 frequency of 3% to 11% (5, 6, 8, 9). pathway and modulates AMPK activity (12). Both The precise clinical mechanisms responsible for causing the AMPK and LKB1 kinases are potential targets for fatigue and asthenia are unknown but are likely to work sunitinib (13), which may, therefore, impair the signal- through similar pathways. Cancer-related fatigue is ing pathway responsible for maintaining the correct known to involve both peripheral (originating in the energy cellular balance, resulting in fatigue and/or muscles and related tissues) and central (developing in asthenia. the CNS) mechanisms, and several possible pathophysio- logic pathways have been explored (10). Looking at Glucose transporters RCC specifically, a study in diabetic patients showed that Glucose uptake occurs through facilitative diffusion in sunitinib triggers a decrease in blood glucose levels (11), a process mediated by plasma membrane glucose trans- which may be involved in the induction of fatigue and/or porters. Contractile activity in the muscles triggers glu- asthenia. However, as both diabetic and nondiabetic cose uptake in an insulin-independent manner. Nitric patients treated with sunitinib experience fatigue and oxide synthase (NOS) is also stimulated in muscles in asthenia (11), other mechanisms may also be involved, response to contractile activity, increasing nitric oxide as discussed below. (NO) production. Some research suggests that inhibition of NO production can block the ability of exercise to AMP-activated protein kinase stimulate glucose transport (14). Other studies have AMP-activated protein kinase (AMPK) is critical shown that endothelial NOS is stimulated in a VEGFR- in ensuring a balance between anabolic and catabolic dependent manner (15). 2216 Mol Cancer Ther; 10(12) December 2011 Molecular Cancer Therapeutics Downloaded from mct.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Molecular Basis of Sunitinib Toxicity In light of these data, sunitinib-mediated inhibition of Molecular Basis of Sunitinib Cardiotoxicity VEGFR may trigger inhibition of NOS and reduced pro- duction of NO, resulting in a reduction in glucose uptake. The reported incidence of cardiotoxicity (primarily In addition, as VEGFR can convey its signal through left ventricular dysfunction) with sunitinib varies from protein kinase C (PKC), sunitinib may indirectly inhibit approximately 11% to 16% in the phase III studies in PKC activity. PKC mediates calcium-mediated glucose patients with GIST or metastatic RCC, respectively uptake in the muscle, and its inhibition could impair (4, 7, 8). The molecular mechanisms that may explain insulin-dependent glucose uptake to the muscle. sunitinib-induced cardiomyocyte cytotoxicity are reviewed below. Circadian rhythm disruption Ligands of growth factors may also play a role in Angiogenesis circadian rhythm disruption, which can lead to symptom Among the targets of sunitinib, only PDGFRs are clusters, including fatigue, appetite
Recommended publications
  • ARTICLES Fibroblast Growth Factors 1, 2, 17, and 19 Are The
    0031-3998/07/6103-0267 PEDIATRIC RESEARCH Vol. 61, No. 3, 2007 Copyright © 2007 International Pediatric Research Foundation, Inc. Printed in U.S.A. ARTICLES Fibroblast Growth Factors 1, 2, 17, and 19 Are the Predominant FGF Ligands Expressed in Human Fetal Growth Plate Cartilage PAVEL KREJCI, DEBORAH KRAKOW, PERTCHOUI B. MEKIKIAN, AND WILLIAM R. WILCOX Medical Genetics Institute [P.K., D.K., P.B.M., W.R.W.], Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Obstetrics and Gynecology [D.K.] and Department of Pediatrics [W.R.W.], UCLA School of Medicine, Los Angeles, California 90095 ABSTRACT: Fibroblast growth factors (FGF) regulate bone growth, (G380R) or TD (K650E) mutations (4–6). When expressed at but their expression in human cartilage is unclear. Here, we deter- physiologic levels, FGFR3-G380R required, like its wild-type mined the expression of entire FGF family in human fetal growth counterpart, ligand for activation (7). Similarly, in vitro cul- plate cartilage. Using reverse transcriptase PCR, the transcripts for tivated human TD chondrocytes as well as chondrocytes FGF1, 2, 5, 8–14, 16–19, and 21 were found. However, only FGF1, isolated from Fgfr3-K644M mice had an identical time course 2, 17, and 19 were detectable at the protein level. By immunohisto- of Fgfr3 activation compared with wild-type chondrocytes and chemistry, FGF17 and 19 were uniformly expressed within the showed no receptor activation in the absence of ligand (8,9). growth plate. In contrast, FGF1 was found only in proliferating and hypertrophic chondrocytes whereas FGF2 localized predominantly to Despite the importance of the FGF ligand for activation of the resting and proliferating cartilage.
    [Show full text]
  • Antitumor Reactivity in Vitro Sorafenib Differentially Affect NK Cell The
    The Kinase Inhibitors Sunitinib and Sorafenib Differentially Affect NK Cell Antitumor Reactivity In Vitro This information is current as Matthias Krusch, Julia Salih, Manuela Schlicke, Tina of September 24, 2021. Baessler, Kerstin Maria Kampa, Frank Mayer and Helmut Rainer Salih J Immunol 2009; 183:8286-8294; ; doi: 10.4049/jimmunol.0902404 http://www.jimmunol.org/content/183/12/8286 Downloaded from References This article cites 44 articles, 21 of which you can access for free at: http://www.jimmunol.org/content/183/12/8286.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication by guest on September 24, 2021 *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2009 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology The Kinase Inhibitors Sunitinib and Sorafenib Differentially Affect NK Cell Antitumor Reactivity In Vitro1 Matthias Krusch,2 Julia Salih,2 Manuela Schlicke,2 Tina Baessler, Kerstin Maria Kampa, Frank Mayer,3 and Helmut Rainer Salih3,4 Sunitinib and Sorafenib are protein kinase inhibitors (PKI) approved for treatment of patients with advanced renal cell cancer (RCC).
    [Show full text]
  • And Insulin-Like Growth Factor-I (IGF-I) in Regulating Human Erythropoiesis
    Leukemia (1998) 12, 371–381 1998 Stockton Press All rights reserved 0887-6924/98 $12.00 The role of insulin (INS) and insulin-like growth factor-I (IGF-I) in regulating human erythropoiesis. Studies in vitro under serum-free conditions – comparison to other cytokines and growth factors J Ratajczak, Q Zhang, E Pertusini, BS Wojczyk, MA Wasik and MZ Ratajczak Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA The role of insulin (INS), and insulin-like growth factor-I (IGF- has been difficult to assess. The fact that EpO alone fails to I) in the regulation of human erythropoiesis is not completely stimulate BFU-E in serum-free conditions, but does do in understood. To address this issue we employed several comp- lementary strategies including: serum free cloning of CD34؉ serum containing cultures indicates that serum contains some cells, RT-PCR, FACS analysis, and mRNA perturbation with oli- crucial growth factors necessary for the BFU-E development. godeoxynucleotides (ODN). In a serum-free culture model, both In previous studies from our laboratory, we examined the ؉ INS and IGF-I enhanced survival of CD34 cells, but neither of role of IGF-I12 and KL9,11,13 in the regulation of early human these growth factors stimulated their proliferation. The influ- erythropoiesis. Both of these growth factors are considered to ence of INS and IGF-I on erythroid colony development was be crucial for the BFU-E growth.3,6,8,14 Unexpectedly, that dependent on a combination of growth factors used for stimul- + ating BFU-E growth.
    [Show full text]
  • Imatinib-Induced Interstitial Lung Disease and Sunitinib-Associated
    CASE Imatinib-induced interstitial lung disease and REPORT sunitinib-associated intra-tumour haemorrhage Herbert H Loong 龍浩鋒 Winnie Yeo 楊明明 An ethnically Chinese patient with newly diagnosed metastatic gastro-intestinal stromal tumour initially treated with imatinib mesylate developed severe interstitial lung disease. As his condition improved after cessation of imatinib mesylate and treatment with corticosteroids, he was started on sunitinib malate. His clinical course was then unfortunately complicated with intra-tumour bleeding. This case report illustrates the dilemmas and complexities associated with treating patients with gastro-intestinal stromal tumours with the new tyrosine kinase inhibitors. Case report A 63-year-old man was referred to our department in January 2007 after being diagnosed with a recurrent gastro-intestinal stromal tumour (GIST). He was initially diagnosed with a duodenal GIST in January 2000 on presenting with symptoms of anaemia. A workup, including upper endoscopy, revealed an ulcerative growth over the third and fourth part of the duodenum. A computed tomographic (CT) scan showed a 4.5 x 5.5 cm soft tissue mass over the same area. A duodenectomy and duodeno-jejunostomy were performed and a pathological examination of tissue removed at surgery confirmed a low-grade GIST (S-100 positive; 4.5 cm in size, mitosis 6/10 high-power field, c-KIT positive). The resection margins were clear so he was managed with routine follow-up and observation. An abdominal ultrasound performed in 2003 showed no evidence of metastases. He remained well until January 2007 when hepatomegaly was found during a physical examination. An abdominal CT scan showed multiple hypervascular tumour foci with cystic changes in both liver lobes.
    [Show full text]
  • The Tyrosine-Kinase Inhibitor Sunitinib Targets Vascular Endothelial (VE)-Cadherin: a Marker of Response to Antitumoural Treatment in Metastatic Renal Cell Carcinoma
    www.nature.com/bjc ARTICLE Translational Therapeutics The tyrosine-kinase inhibitor sunitinib targets vascular endothelial (VE)-cadherin: a marker of response to antitumoural treatment in metastatic renal cell carcinoma Helena Polena1, Julie Creuzet1, Maeva Dufies2, Adama Sidibé1, Abir Khalil-Mgharbel1, Aude Salomon1, Alban Deroux3, Jean-Louis Quesada4, Caroline Roelants1, Odile Filhol1, Claude Cochet1, Ellen Blanc5, Céline Ferlay-Segura5, Delphine Borchiellini6, Jean-Marc Ferrero6, Bernard Escudier7, Sylvie Négrier5, Gilles Pages8 and Isabelle Vilgrain1 BACKGROUND: Vascular endothelial (VE)-cadherin is an endothelial cell-specific protein responsible for endothelium integrity. Its adhesive properties are regulated by post-translational processing, such as tyrosine phosphorylation at site Y685 in its cytoplasmic domain, and cleavage of its extracellular domain (sVE). In hormone-refractory metastatic breast cancer, we recently demonstrated that sVE levels correlate to poor survival. In the present study, we determine whether kidney cancer therapies had an effect on VE- cadherin structural modifications and their clinical interest to monitor patient outcome. METHODS: The effects of kidney cancer biotherapies were tested on an endothelial monolayer model mimicking the endothelium lining blood vessels and on a homotypic and heterotypic 3D cell model mimicking tumour growth. sVE was quantified by ELISA in renal cell carcinoma patients initiating sunitinib (48 patients) or bevacizumab (83 patients) in the first-line metastatic setting (SUVEGIL and TORAVA trials). RESULTS: Human VE-cadherin is a direct target for sunitinib which inhibits its VEGF-induced phosphorylation and cleavage on endothelial monolayer and endothelial cell migration in the 3D model. The tumour cell environment modulates VE-cadherin functions through MMPs and VEGF. We demonstrate the presence of soluble VE-cadherin in the sera of mRCC patients (n = 131) which level at baseline, is higher than in a healthy donor group (n = 96).
    [Show full text]
  • Factor-The Controlling Growth Factor-At a Sufficiently Low Value, While the Cell Population Has Ceased to Grow and Is in a Sort
    708 GENETICS: NO VICK A ND SZILARD PROC. N. A. S. EXPERIMENTS WITH THE CHEMOSTAT ON SPONTANEOUS MUTATIONS OF BACTERIA By AARON NOVICK AND LEO SZILARD INSTITUTRE OF RADIOBIOLOGY AND BIOPHYSICS, UNIVERSITY OF CHICAGO Communicated by H. J. Muller, October 18, 1950 Introduction.-All bacteria require for growth the presence of certain inorganic chemical components in the nutrient, such as potassium, phos- phorus, sulphur, etc., and with a few exceptions all bacteria require an energy-yielding carbon source, such as, for instance, glucose or lactate, etc. In addition to these elements or simple compounds, certain bacteria require more complex compounds, for instance an amino acid, which they are not capable of synthesizing. For the purposes of this presentation, any of the chemical compounds which a given strain of bacteria requires for its growth will be called a "growth factor." In general, the growth rate of a bacterial strain may be within very wide limits independent of the concentration of a given growth factor; but since at zero concentration the growth rate is zero, there must of necessity exist, at sufficiently low concentrations of the growth factor, a region in which the growth rate falls with falling concentration of the growth factor. It therefore should be possible to maintain a bacterial population over an indefinite period of time growing at a rate considerably lower than normal simply by maintaining the concentration of one growth factor-the controlling growth factor-at a sufficiently low value, while the concentrations of all other growth factors may at the same time be main- tained at high values.
    [Show full text]
  • Updated November 2019 KIT Mutation the KIT Gene Is Associated With
    Updated November 2019 KIT Mutation The KIT gene is associated with autosomal dominant piebaldism, gastrointestinal stromal tumors (GISTs), and familial mastocytosis.1-3 The type of mutation identified in the KIT gene determines the symptoms/cancer risks that the individual may have. Pathogenic loss-of-function (LOF) mutations in the KIT gene are generally only associated with Piebaldism. However, pathogenic gain of function mutations in the KIT gene are associated with systemic mastocytosis and gastrointestinal stromal tumors.4,5 Piebaldism: This is a rare condition characterized by the patchy absence of melanocytes in certain areas of the skin and hair. Melanocytes produce the pigment melanin, which contributes to hair, eye, and skin color. The absence of melanocytes leads to patches of skin and hair that are lighter than normal. These unpigmented areas are typically present at birth and do not increase in size or number.6-8 Gastrointestinal stromal tumors (GIST): GISTs are tumors that occur in the gastrointestinal tract, most commonly in the stomach or small intestine. Small tumors may cause no signs or symptoms. However, some people with GISTs may experience pain or swelling in the abdomen, nausea, vomiting, loss of appetite, or weight loss. These tumors can be cancerous (malignant) or noncancerous (benign). Mastocytosis: This is a blood disorder that occurs when white blood cells called mast cells accumulate in one or more tissues (most commonly in the bone marrow). Mast cells normally trigger inflammation during an allergic reaction. When an environmental trigger activates mast cells, they release proteins that signal an immune response. In systemic mastocytosis, excess mast cells mean more proteins are being released in the tissues where the cells accumulate, leading to an increased immune response.
    [Show full text]
  • B-Cell Growth Factor
    Proc. Natl Acad. Sci. USA Vol. 79, pp. 7455-7459, December 1982 Immunology B-cell growth factor: Distinction from T-cell growth factor and B-cell maturation factor (B lymphocytes/T-cell hybridomas) TOMAS LEANDERSON*, ERIK LUNDGRENt, ERIK RUUTH*, HAKAN BORGt, HAKAN PERSSONt, AND ANTONIO COUTINHOt tLaboratory for Cell and Tissue Culture Research, *Institute of Pathology, and +Department of Immunology, UmeA University, S-901 87 UmeA, Sweden Communicated by Niels K. Jerne, July 12, 1982 ABSTRACT A T-cell hybridoma was derived by somatic cell We have initiated these studies by systematically screening hybridization between concanavalin A-activated BALB/c spleen hybridoma activities in different assays that measure either cells and the AKR thymoma BW 5147. Media conditioned by hy- growth or maturation ofall B lymphocytes, regardless ofclonal bridoma cells, even at high dilutions (1:1,000) support the growth specificity. We chose to study activated rather than resting B oflipopolysaccharide-stimulated B-cell blasts but not that ofT-cell cells because of the-overwhelming evidence that initiation of growth factor (TCGF)-reactive T-cells. This activity, herein des- cooperative B-cell responses requires direct cellular interaction ignated B-cell growth factor (BCGF), has a Mr of =20,000 and it (13, 14). In this report we describe a hybridoma clone secreting can readily be separated from TCGF (Mr "30,000) by gel filtra- a factor that supports growth but not maturation of lipopoly- tion. BCGF is constitutively produced by the hybridoma cells, it saccharide (LPS)-activated B-cell blasts and is devoid of is removed from conditioned media by incubation with target cells T-cell at +4°C, and it is equally effective on B-cell blasts carrying dif- growthfactor(TCGF) andT-cellreplacingfactor(TRF) activities.
    [Show full text]
  • Sunitinib Malate)
    Prescribing Information Update for SUTENT® (sunitinib malate) July 12, 2010 Dear Health Care Provider: Pfizer Oncology is committed to providing you with up-to-date information about SUTENT® (sunitinib malate) capsules. This letter is to inform you of an important update to the SUTENT prescribing information (PI). The following boxed warning and safety information has been added to the PI for SUTENT: WARNING: HEPATOTOXICITY Hepatotoxicity has been observed in clinical trials and post-marketing experience. This hepatotoxicity may be severe and deaths have been reported. WARNINGS and PRECAUTIONS Hepatotoxicity SUTENT has been associated with hepatotoxicity, which may result in liver failure or death. Liver failure has been observed in clinical trials (7/2281 [0.3%]) and post-marketing experience. Liver failure signs include jaundice, elevated transaminases and/or hyperbilirubinemia in conjunction with encephalopathy, coagulopathy, and/or renal failure. Monitor liver function tests (ALT, AST, bilirubin) before initiation of treatment, during each cycle of treatment, and as clinically indicated. SUTENT should be interrupted for Grade 3 or 4 drug-related hepatic adverse events and discontinued if there is no resolution. Do not restart SUTENT if patients subsequently experience severe changes in liver function tests or have other signs and symptoms of liver failure. Safety in patients with ALT or AST >2.5 × ULN or, if due to liver metastases, >5.0 × ULN has not been established. In addition, the labeling includes a new Medication Guide that your patients will receive when SUTENT is dispensed. Pfizer maintains a global safety database, monitoring all clinical trials and reports of spontaneous adverse events. The incidence of liver failure referenced above is consistent with the very low rate of hepatic failure described in the clinical trials of sunitinib used to support original FDA registration in 2006.
    [Show full text]
  • Mechanism of Myeloid-Derived Suppressor Cell
    MECHANISM OF MYELOID-DERIVED SUPPRESSOR CELL ACCUMULATION IN CANCER AND SUSCEPTIBILITY TO REVERSAL BY SUNITINIB by JENNIFER SUSAN KO, M.D. Submitted in partial fulfillment of the requirements For the degree of Doctor of Philosophy Thesis Adviser: Dr. James H. Finke Department of Pathology CASE WESTERN RESERVE UNIVERSITY January, 2010 CASE WESTERN RESERVE UNIVERSITY SCHOOL OF GRADUATE STUDIES We hereby approve the thesis of Jennifer Susan Ko candidate for the Doctor of Philosophy degree*. (signed) Alan Levine Ph.D. David Kaplan M.D., Ph.D. Clark Distelhorst M.D. James Finke Ph.D. Charles Tannenbaum Ph.D. (date) October 12th, 2009 *We also certify that written approval has been obtained for any proprietary material contained therein. 2 TABLE OF CONTENTS Title Page 1 Signature Sheet 2 Table of Contents 3 List of Tables 6 List of Figures 7 Acknowledgements 9 List of Abbreviations 10 Abstract 14 Chapter 1: Introduction 16 Overview: Myeloid-derived suppressor cells in cancer: a novel therapeutic target. 16 Immunotherapy in cancer 16 Myeloid-derived suppressor cells limit immunotherapy 22 Myeloid-derived suppressor cells limit anti-angiogenic therapy 28 Multiple factors are implicated in MDSC formation 30 Vascular Endothelial Growth Factor 30 Stem Cell Factor 32 Granulocyte- and Granulocyte/Monocyte Colony Stimulating Factors 33 S100A9 and Inflammation 34 Intracellular signaling implicated in MDSC programming 36 3 Chapter 2: Sunitinib Mediates Reversal of Myeloid-Derived Suppressor Cell Accumulation in Renal Cell Carcinoma Patients 44 Statement
    [Show full text]
  • Human Transforming Growth Factor Α (TGF-Α)
    787 STOMACH Gut: first published as 10.1136/gut.51.6.787 on 1 December 2002. Downloaded from Human transforming growth factor α (TGF-α) is digested to a smaller (1–43), less biologically active, form in acidic gastric juice T Marchbank, R Boulton, H Hansen, R J Playford ............................................................................................................................. Gut 2002;51:787–792 Background: Transforming growth factor α (TGF-α) is a 50 amino acid peptide with potent prolifera- tive and cytoprotective activity present in gastric mucosa and juice. Aims: To determine the forms and biological activity of natural and recombinant TGF-α following incu- bation with acid pepsin. Patients: Human gastric juice was obtained under basal conditions from patients taking acid suppres- sants and from volunteers undergoing intragastric neutralisation. See end of article for Methods: Samples were analysed using mass spectroscopy and/or high pressure liquid chromatogra- authors’ affiliations phy with radioimmunoassay. Biological activity was determined using thymidine incorporation into rat ....................... hepatocytes and an indomethacin/restraint induced gastric damage rat model. α α Correspondence to: Results: TGF- 1–50 is cleaved to TGF- 1–43 by acid pepsin and this is the predominant form in normal Professor R J Playford, gastric juice. However, intragastric neutralisation or taking acid suppressants caused the predominant Gastroenterology Section, α α 3 form to be TGF- 1–50.TGF- 1–43 had only half of the ability to maximally stimulate [ H]thymidine incorpo- Imperial College School of α ration into primary rat hepatocytes (28 177 (1130) DPM/well for 2.16 nM TGF- 1–43 v 63 184 (3536) Medicine, Hammersmith α Hospital Campus, Du Cane DPM/well for TGF- 1–50; p<0.001).
    [Show full text]
  • (ALK) Rearrangement in Adult Renal Cell Carcinoma with Lung Metastasis: a Case Report and Literature Review
    2861 Case Report Anaplastic lymphoma kinase (ALK) rearrangement in adult renal cell carcinoma with lung metastasis: a case report and literature review Shengyu Zhou1, Guanxing Sun2, Jianwei Wang1, Hongtu Zhang1 1Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China; 2Department of Medical Oncology, Zaozhuang Municipal Hospital, Zaozhuang, China Correspondence to: Shengyu Zhou. Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China. Email: [email protected]. Abstract: Renal cell carcinoma (RCC) with anaplastic lymphoma kinase (ALK) rearrangement is rare, and the genetic profiles of the tumor have not been elucidated. Here, we report a case with recurrent papillary RCC and lung metastasis after nephrectomy for nearly 7 years. The patient first received sunitinib, whereas the drug toxicity was intolerable. Combined Immunohistology (IHC) and fluorescence in situ hybridization (FISH) revealed the patient has an ALK rearrangement, and the patient then was treated with crizotinib. The patient had good tolerance, and a partial response in the target lesions was achieved. In order to further understand the benefit of crizotinib in ALK-rearranged RCC, the patient was detected with whole exome sequencing (WES) to study her genetic profiles. Compared those of RCC cases without ALK rearrangement (nALK-RCC), the patient and nine RCC cases with ALK rearrangement (ALK-RCC) revealed unique genetic characteristics: 1) The common mutations that occurred in RCC were not found in ALK-RCC.; 2) A total of 11 co-existing mutations in ALK-RCC were found, and they occurred in nALK-RCC at a relatively low frequency.
    [Show full text]