chemosensitivity vol 1

Upload: pmendez00

Post on 13-Apr-2018

236 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/26/2019 Chemosensitivity Vol 1

    1/247

    M E T H O D S I N M O L E C U L A R M E D I C I N ETM

    Chemosensitivity

    Edited by

    Rosalyn D. Blumenthal

    Volume 1

    In Vitro Assays

    Chemosensitivity

    Edited by

    Rosalyn D. Blumenthal

    Volume 1

    In Vitro Assays

  • 7/26/2019 Chemosensitivity Vol 1

    2/247

    Chemosensitivity

  • 7/26/2019 Chemosensitivity Vol 1

    3/247

    M E T H O D S I N M O L E C U L A R M E D I C I N E

    John M. Walker, SERIESEDITOR

    118. Antifungal Agents:Methods and Protocols,edited byErika J. Ernst and P. David Rogers,2005

    117. Fibrosis Research:Methods and Protocols,edited byJohn Varga, David A. Brenner,and Sem H. Phan, 2005

    116. Inteferon Methods and Protocols, edited byDaniel J. J. Carr, 2005

    115. Lymphoma:Methods and Protocols, edited byTimothy Illidge and Peter W. M. Johnson,2005

    114. Microarrays in Clinical Diagnostics, edited byThomas Joos and Paolo Fortina, 2005

    113. Multiple Myeloma:Methods andProtocols,edited byRoss D. Brown and P. Joy Ho, 2005

    112. Molecular Cardiology:Methods andProtocols,edited byZhongjie Sun, 2005

    111. Chemosensitivity: Volume 2, In Vivo Mod-els, Imaging, and Molecular Regulators,editedbyRosalyn D. Blumethal, 2005

    110. Chemosensitivity: Volume 1, In Vitro Assays,edited byRosalyn D. Blumethal, 2005

    109. Adoptive Immunotherapy:Methods andProtocols,edited byBurkhard Ludewig and

    Matthias W. Hoffman, 2005

    108. Hypertension:Methods and Protocols,edited byJrme P. Fennell and Andrew

    H. Baker, 2005

    107. Human Cell Culture Protocols, SecondEdition,edited byJoanna Picot, 2005

    106. Antisense Therapeutics, Second Edition,edited byM. Ian Phillips, 2005

    105. Developmental Hematopoiesis:Methodsand Protocols,edited byMargaret H. Baron,2005

    104. Stroke Genomics:Methods and Reviews,editedby Simon J. Read and David Virley, 2004

    103. Pancreatic Cancer:Methods and Protocols,edited by Gloria H. Su, 2004

    102. Autoimmunity:Methods and Protocols,edited byAndras Perl, 2004

    101. Cartilage and Osteoarthritis: Volume 2,Structure and In Vivo Analysis,edited byFrdric De Ceuninck, Massimo Sabatini, and

    Philippe Pastoureau, 2004100. Cartilage and Osteoarthritis: Volume 1,

    Cellular and Molecular Tools,edited byMassimo Sabatini, Philippe Pastoureau, andFrdric De Ceuninck, 2004

    99. Pain Research:Methods and Protocols,edited byDavid Z. Luo, 2004

    98. Tumor Necrosis Factor:Methods andProtocols,edited byAngelo Corti and PietroGhezzi, 2004

    97. Molecular Diagnosis of Cancer:Methods andProtocols, Second Edition,edited byJoseph E.

    Roulston and John M. S. Bartlett, 2004

    96. Hepatitis B and D Protocols: Volume 2,Immunology, Model Systems, and ClinicalStudies,edited byRobert K. Hamatake and

    Johnson Y. N. Lau, 200495. Hepatitis B and D Protocols: Volume 1,

    Detection, Genotypes, and Characterization,edited byRobert K. Hamatake and Johnson Y.

    N. Lau, 2004

    94. Molecular Diagnosis of Infectious Diseases,Second Edition,edited byJochen Decker andUdo Reischl, 2004

    93. Anticoagulants, Antiplatelets, andThrombolytics, edited by Shaker A. Mousa,2004

    92. Molecular Diagnosis of Genetic Diseases,Second Edition, edited byRob Elles and

    Roger Mountford, 2004

    91. Pediatric Hematology:Methods andProtocols, edited byNicholas J. Gouldenand Colin G. Steward, 2003

    90. Suicide Gene Therapy:Methods and Reviews,edited by Caroline J. Springer, 2004

    89. The BloodBrain Barrier:Biology andResearch Protocols, edited by Sukriti Nag, 2003

    88. Cancer Cell Culture:Methods and Protocols,

    edited bySimon P. Langdon, 200387. Vaccine Protocols, Second Edition, edited by

    Andrew Robinson, Michael J. Hudson, andMartin P. Cranage, 2003

    86. Renal Disease: Techniques and Protocols,edited byMichael S. Goligorsky, 2003

    85. Novel Anticancer Drug Protocols, edited byJohn K. Buolamwiniand Alex A. Adjei, 2003

    84. Opioid Research:Methods and Protocols,edited byZhizhong Z. Pan, 2003

    83. Diabetes Mellitus:Methods and Protocols,edited by Sabire zcan, 2003

    82. Hemoglobin Disorders:Molecular Methodsand Protocols, edited byRonald L. Nagel,2003

  • 7/26/2019 Chemosensitivity Vol 1

    4/247

    Chemosensitivity

    Volume 1

    In Vitro Assays

    Edited by

    Rosalyn D. BlumenthalGarden State Cancer Center, Belleville, NJ

    M E T H O D S I N M O L E C U L A R M E D I C I N E

  • 7/26/2019 Chemosensitivity Vol 1

    5/247

    2005 Humana Press Inc.999 Riverview Drive, Suite 208Totowa, New Jersey 07512

    www.humanapress.com

    All rights reserved. No part of this book may be reproduced, stored in a retrieval system, or transmitted in any form orby any means, electronic, mechanical, photocopying, microfilming, recording, or otherwise without written permissionfrom the Publisher. Methods in Molecular Medicine is a trademark of The Humana Press Inc.

    The content and opinions expressed in this book are the sole work of the authors and editors, who havewarranted due diligence in the creation and issuance of their work. The publisher, editors, and authors arenot responsible for errors or omissions or for any consequences arising from the information or opinionspresented in this book and make no warranty, express or implied, with respect to its contents.

    This publication is printed on acid-free paper.

    ANSI Z39.48-1984 (American Standards Institute)

    Permanence of Paper for Printed Library Materials.

    Cover illustrations: Foreground illustration: Figure 3, from Chapter 10, Chemosensitivity Testing UsingMicroplateAdenosine TriphosphateBased Luminescence Measurements, by Christian M. Kurbacher andIan A. Cree. Background illustration: Figure 4, from Chapter 22 (Volume 2), Assessing Growth andResponse to Therapy in Murine Tumor Models, by C. P. Reynolds et al.

    Cover design by Patricia F. Cleary.

    For additional copies, pricing for bulk purchases, and/or information about other Humana titles, contactHumana at the above address or at any of the following numbers: Tel.: 973-256-1699; Fax: 973-256-8341;E-mail: [email protected]; or visit our Website: www.humanapress.com

    Photocopy Authorization Policy:

    Authorization to photocopy items for internal or personal use, or the internal or personal use of specificclients, is granted by Humana Press Inc., provided that the base fee of US $30.00 per copy is paid directlyto the Copyright Clearance Center at 222 Rosewood Drive, Danvers, MA 01923. For those organizationsthat have been granted a photocopy license from the CCC, a separate system of payment has been arrangedand is acceptable to Humana Press Inc. The fee code for users of the Transactional Reporting Service is:[1-58829-345-9/05 $30.00].

    Printed in the United States of America. 10 9 8 7 6 5 4 3 2 1

    E-ISBN 1-59259-869-2Library of Congress Cataloging in Publication DataChemosensitivity / edited by Rosalyn D. Blumenthal.

    v. ; cm. (Methods in molecular medicine ; 110-111)Includes bibliographical references and index.Contents: v. 1. In vitro assays v. 2 In vivo models, imaging, andmolecular regulators.ISBN 1-58829-345-9 (hardcover : alk. paper)1. CancerChemotherapyLaboratory manuals. 2. AntineoplasticagentsEffectivenessLaboratory manuals. 3. Cancer cellsLaboratorymanuals. 4. Cancer--Molecular aspectsLaboratory manuals.

    [DNLM: 1. Antineoplastic Agentspharmacology. 2. Drug Screening Assays,Antitumormethods. 3. Drug Resistance, Neoplasm. 4. Models, Animal. 5.Neoplasmsdrug therapy. QV 269 C5177 2005] I. Blumenthal, Rosalyn D. II.Series.

    RC271.C5C396 2005

    616.994061dc22 2004012494

  • 7/26/2019 Chemosensitivity Vol 1

    6/247

    v

    Preface

    Chemotherapy is used to treat many types of cancer. A large number of drug

    classes are in use, including the vinca alkaloids, taxanes, antibiotics, anthracy-

    clines, DNA alkylators, other DNA damaging agents, hormones, and interfer-

    ons. More potent analogs of existing drugs and novel agents directed at new

    targets are continuously being developed. Over the last few years, agents that

    affect COX-2, PPAR, and various signal transduction pathways have received

    much attention. To identify which agents are effective for which types of tumors,

    it is important to develop accurate in vitro and preclinical in vivo screening sys-

    tems that can identify the cytotoxic and/or cytostatic potential of an agent on

    established tumor cell lines or cells isolated from individual fresh cancer biopsy

    specimens removed from cancer patients. Chemosensitivity testing allows the

    selection of drugs that appear sensitive in the laboratory, thus offering patients a

    better chance of response.

    One of the main problems associated with chemotherapy has been that

    patient tumors with the same histology do not necessarily respond identically

    to the same agent or dose schedule of multiple agents. Identifying the presence

    of resistance mechanisms and other determinants for drug sensitivity in orderto classify tumors into response categories has been an ongoing research

    effort. Advances in our understanding of the genetic and protein fingerprints of

    primary tumors and their metastases has opened a door to the possibility of

    customizing therapy to individuals. There is accumulating evidence suggest-

    ing that laboratory screening of samples from a patients tumor may help select

    the appropriate treatment(s) to administer, thereby avoiding ineffective drugs,

    and sparing patients the side effects normally associated with these agents.

    The aim of these two volumes on Chemosensitivity of theMethods in Molecu-lar Medicineseries, is to comprehensively present protocols that can be used to

    (a) assess chemosensitivity in vitro and in vivo, and (b) assess parameters that

    modulate chemosensitivity in individual tumors. Volume I presents an overview

    in Chapter 1 and then covers In Vitro Measures of Chemosensitivity, includes

    clonogenic, colorimetric, fluorometric, and histochemical approaches. Volume

    II, Part I,Measurements of DNA Damage, Cell Death, and Regulators of Cyto-

    toxicity, includes methods to detect chromosome loss and breakage, changes in

    cell cycle, expression of members of the bcl-2 family of proteins, expression of

    caspases and PARP cleavage, metabolic factors influencing sensitivity, measure-

    ments of drug retention, expression of drug resistance proteins, and measure-

    ments of ceramide and sphingolipids associated with drug sensitivity. Volume

  • 7/26/2019 Chemosensitivity Vol 1

    7/247

    vi Preface

    II, Part II, Genomics, Proteomics, and Chemosensitivity, addresses DNA

    microarrays for gene profiling, genetic manipulation to identify genes regu-

    lating chemosensitivity, proteomics using 2D-PAGE and mass spectrometry,and bioinformatics approaches. The last part, In Vivo Animal Modeling of

    Chemosensitivity,covers protocols to establish clinically meaningful metastatic

    and orthotropic models of solid and liquid tumors, statistical approaches to ana-

    lyze preclinical data, and animal imaging approaches that can be used to assess

    chemosensitivity such as GFP-tagged genes, SPECT using 99mTc-annexin, PET

    imaging with 18FDG, and magnetic resonance imaging.

    Each chapter is written by someone experienced with the methodology and

    contains a detailed introductory section with references of how the technique

    has been used in the past, a list of materials and equipment needed to perform

    the assay, and a step-by-step set of instructions for each method. At the end of

    each chapter a Notes section is included with useful information, helpful

    hints, and problems and pitfalls to be aware of, in order to make the assay run

    smoothly and allow for easy interpretation of data.

    Rosalyn D. Blumenthal

  • 7/26/2019 Chemosensitivity Vol 1

    8/247

    Contents

    Preface ..............................................................................................................v

    Contributors ..................................................................................................... ix

    Contents of Volume 2 ......................................................................................xi

    PARTI. OVERVIEW

    1 An Overview of Chemosensitivity Testing

    Rosalyn D. Blumenthal ......................................................................... 3

    PARTII. INVITROMEASURESOFCHEMOSENSITIVITY

    2 Clonogenic Cell Survival Assay

    Anupama Munshi, Marvette Hobbs, and Raymond E. Meyn.............. 21

    3 High-Sensitivity Cytotoxicity Assays for Nonadherent Cells

    M. Jules Mattes ................................................................................... 29

    4 Sulforhodamine B Assay and Chemosensitivity

    Wieland Voight ................................................................................... 39

    5 Use of the Differential Staining Cytotoxicity

    Assay to Predict ChemosensitivityGertjan J. L. Kaspers ........................................................................... 49

    6 Collagen Gel Droplet Culture Methodto Examine In Vitro Chemosensitivity

    Hisayuki Kobayashi ............................................................................. 59

    7 The MTT Assay to Evaluate Chemosensitivity

    Jack D. Burton .................................................................................... 69

    8 Histoculture Drug Response Assay to Monitor Chemoresponse

    Shinji Ohie, Yasuhiro Udagawa, Daisuke Aoki,and Shiro Nozawa .......................................................................... 79

    9 In Vitro Testing of Chemosensitivity in Physiological Hypoxia

    Rita Grigoryan, Nino Keshelava, Clarke Anderson,and C. Patrick Reynolds.................................................................. 87

    10 Chemosensitivity Testing Using Microplate AdenosineTriphosphateBased Luminescence Measurements

    Christian M. Kurbacher and Ian A. Cree .......................................... 101

    11 High-Throughput Technology:Green Fluorescent Protein to Monitor Cell Death

    Marylne Fortin, Ann-Muriel Steff, and Patrice Hugo ..................... 121

    vii

  • 7/26/2019 Chemosensitivity Vol 1

    9/247

    viii Contents

    12 DIMSCAN: A Microcomputer Fluorescence-Based CytotoxicityAssay for Preclinical Testing of Combination Chemotherapy

    Nino Keshelava, Tom

    s

    Frgala, Jir

    Krejsa, Ondrej Kalous,and C. Patrick Reynolds ................................................................ 139

    13 The ChemoFxAssay: An Ex Vivo Cell Culture Assayfor Predicting Anticancer Drug Responses

    Robert L. Ochs, Dennis Burholt, and Paul Kornblith ....................... 155

    14 Evaluating Response to Antineoplastic DrugCombinations in Tissue Culture Models

    C. Patrick Reynolds and Barry J. Maurer .......................................... 173

    15 Image Analysis Using the Fluochromasia Assayto Quantify Tumor Drug Sensitivity

    John F. Gibbs, Youcef M. Rustum, and Harry K. Slocum ................. 185

    16 Immunohistochemical Detection of Ornithine Decarboxylaseas a Measure of Chemosensitivity

    Uriel Bachrach .................................................................................. 197

    17 Immunohistochemistry of p53, Bcl-2 and Ki-67as Predictors of Chemosensitivity

    Mitsuyoshi Itaya, Jiro Yoshimoto, Kuniaki Kojima,

    and Seiji Kawasaki ........................................................................ 213

    Index ............................................................................................................ 229

  • 7/26/2019 Chemosensitivity Vol 1

    10/247

    ix

    Contributors

    CLARKE ANDERSONDivision Hematology-Oncology, Keck School of Medicine,

    University of Southern California, Los Angeles, CA, USA

    DAISUKEAOKIDepartment of Obstetrics-Gynecology, Keio University

    School of Medicine, Keio, Japan

    URIELBACHRACHDepartment of Molecular Biology, Hebrew University

    Hadassah Medical School, Jerusalem, Israel

    ROSALYND. BLUMENTHAL Garden State Cancer Center, Belleville, NJ, USA

    DENNISBURHOLT Precision Therapeutics, Pittsburgh, PA, USA

    JACKD. BURTON Garden State Cancer Center, Belleville, NJ, USA

    IANA. CREEDepartment of Histopathology, Queen Alexandria Hospital,

    Portsmouth, UK

    MARYLENEFORTIN Topigen Pharmaceuticals, Montreal, Quebec, Canada

    TOMS FRGALA USC-CHLA Institute for Pediatric Clinical Research,

    University of Southern California and Childrens Hospital Los Angeles,

    Los Angeles, CA, USA

    JOHNF. GIBBSDepartment of Surgical Oncology, Roswell Park Cancer

    Institute, Buffalo, NY, USA

    RITAGRIGORYANDevelopmental Therapeutics Section, Childrens Hospital

    of Los Angeles, Los Angeles, CA, USA

    MARVETTEHOBBSDepartment of Experimental Radiology, University of

    Texas M.D. Anderson Cancer Center, Houston, TX, USA

    PATRICE HUGO Caprion Pharmaceuticals Inc., Saint Laurent, QC, Canada

    MITSUYOSHIITAYADepartment of Hepato-Biliary-Pancreatic Surgery,

    Juntendo University, Tokyo, Japan

    ONDREJ KALOUS USC-CHLA Institute for Pediatric Clinical Research,University of Southern California and Childrens Hospital Los Angeles,

    Los Angeles, CA, USA

    GERTJANJ. L. KASPERSDepartment of Pediatric Hematology Oncology, VU

    University Medical Center, Amsterdam, Netherlands

    SEIJIKAWASAKIDepartment of Hepato-Biliary-Pancreatic Surgery,

    Juntendo University, Tokyo, Japan

    NINOKESHELAVA USC-CHLA Institute for Pediatric Clinical Research,

    University of Southern California and Childrens Hospital Los Angeles,Los Angeles, CA, USA

    HISAYUKIKOBAYASHI Biochemical Laboratory, Nitta Gelatin Inc.,

    Futamata, Yao-City, Japan

  • 7/26/2019 Chemosensitivity Vol 1

    11/247

    KUNIAKIKOJIMADepartment of Breast and Endocrine Surgery, Juntendo

    University, Tokyo, Japan

    PAULKORNBLITHPrecision Therapeutics, Pittsburgh, PA, USA

    JIR KREJSA USC-CHLA Institute for Pediatric Clinical Research, University of

    Southern California and Childrens Hospital Los Angeles, Los Angeles, CA,

    USA

    CHRISTIANM. KURBACHERDepartment of Gynecology & Obstetrics, University

    of Cologne, Cologne, Germany

    M. JULESMATTES Center for Molecular Medicine and Immunology,

    Belleville, NJ, USA

    BARRYJ. MAURER USC-CHLA Institute for Pediatric Clinical Research,

    University of Southern California and Childrens Hospital Los Angeles,

    Los Angeles, CA, USA

    RAYMONDE. MEYNDepartment of Experimental Radiology, University of

    Texas M.D. Anderson Cancer Center, Houston, TX, USA

    ANUPAMAMUNSHIDepartment of Experimental Radiology, University of

    Texas M.D. Anderson Cancer Center, Houston, TX, USA

    SHIRONOZAWADepartment of Obstetrics-Gynecology, Keio University

    School of Medicine, Keio, Japan

    ROBERTL. OCHS Precision Therapeutics, Pittsburgh, PA, USA

    SHINJIOHIE Cancer Research Laboratory, Hanno Research Center, Taiho

    Pharmaceutical Co., Saitama, Japan

    C. PATRICKREYNOLDS USC-CHLA Institute for Pediatric Clinical Research,

    University of Southern California and Childrens Hospital Los Angeles,

    Los Angeles, CA, USA

    YOUCEFM. RUSTUMDepartment of Experimental Therapeutics, Roswell

    Park Cancer Institute, Buffalo, NY, USA

    HARRYK. SLOCUMDepartment of Experimental Therapeutics, RoswellPark Cancer Institute, Buffalo, NY, USA

    ANN-MURIELSTEFF World Anti-Doping Agency, Montreal, Quebec, Canada

    YASUHIROUDAGAWADepartment of Obstetrics-Gynecology, Fujita Health

    University School of Medicine, Japan

    WIELANDVOIGHT Klinik fr Innere Medizin IVMartin Luther Universitt

    Halle, Halle/Saale, Germany

    JIROYOSHIMOTODepartment of Hepato-Biliary-Pancreatic Surgery,

    Juntendo University, Tokyo, Japan

    x Contributors

  • 7/26/2019 Chemosensitivity Vol 1

    12/247

    xi

    Contents of Volume 2

    Preface ..............................................................................................................v

    Color Plate .......................................................................................................xi

    Contributors .................................................................................................. xiii

    Contents of Volume 1 ...................................................................................xvii

    PARTI. MEASUREMENTSOFDNA DAMAGE, CELLDEATH,ANDREGULATORSOFCYTOTOXICITY

    1 In Vitro Micronucleus Technique to Predict ChemosensitivityMichael Fenech ..................................................................................... 3

    2 Cell Cycle and Drug Sensitivity

    Aslamuzzaman Kazi and Q. Ping Dou ................................................ 33

    3 TUNEL Assay as a Measure of Chemotherapy-Induced Apoptosis

    Robert Wieder..................................................................................... 43

    4 Apoptosis Assessment by the DNA Diffusion Assay

    Narendra P. Singh ............................................................................... 55

    5 PARP Cleavage and Caspase Activity to Assess ChemosensitivityAlok C. Bharti, Yasunari Takada, and Bharat B. Aggarwal ................ 69

    6 Diphenylamine Assay of DNA Fragmentationfor Chemosensitivity Testing

    Cicek Gercel-Taylor ............................................................................ 79

    7 Immunodetecting Members of the Bcl-2 Family of Proteins

    Richard B. Lock and Kathleen M. Murphy.......................................... 83

    8 Correlation of Telomerase Activity

    and Telomere Length to ChemosensitivityYasuhiko Kiyozuka .............................................................................. 97

    9 Application of Silicon Sensor Technologies to Tumor Tissue In Vitro:Detection of Metabolic Correlates of Chemosensitivity

    Pedro Mestres-Ventura, Andrea Morguet, Anette Schofer,Michael Laue, and Werner Schmidt ............................................. 109

    10 Overview of Tumor Cell Chemoresistance Mechanisms

    Laura Gatti and Franco Zunino ........................................................ 127

    11 Flow Cytometric Monitoring of Fluorescent DrugRetention and Efflux

    Awtar Krishan and Ronald M. Hamelik ............................................ 149

  • 7/26/2019 Chemosensitivity Vol 1

    13/247

    12 Flow Cytometric Measurement of Functionaland Phenotypic P-Glycoprotein

    Monica Pallis and Emma Das-Gupta ................................................ 16713 Measurement of Ceramide and Sphingolipid Metabolism in Tumors:Potential Modulation of Chemosensitivity

    David E. Modrak ............................................................................... 183

    PARTII. GENOMICS, PROTEOMICS, ANDCHEMOSENSITIVITY14 Gene Expression Profiling to Characterize Anticancer

    Drug Sensitivity

    James K. Breaux and Gerrit Los ........................................................ 197

    15 Identifying Genes Related to ChemosensitivityUsing Support Vector Machine

    Lei Bao .............................................................................................. 233

    16 Genetic Manipulation of Yeast to Identify GenesInvolved in Regulation of Chemosensitivity

    Giovanni L. Beretta and Paola Perego .............................................. 241

    17 Real-Time RT-PCR (Taqman) of Tumor mRNAto Predict Sensitivity of Specimens to 5-Fluorouracil

    Tetsuro Kubota .................................................................................. 25718 Use of Proteomics to Study Chemosensitivity

    Julia Poland, Silke Wandschneider, Andrea Urbani,Sergio Bernardini, Giorgio Federici, and Pranav Sinha ............... 267

    PARTIII. INVIVOANIMALMODELINGOFCHEMOSENSITIVITY19 Clinically Relevant Metastatic Breast Cancer

    Models to Study Chemosensitivity

    Lee Su Kim and Janet E. Price ........................................................... 285

    20 Orthotopic Metastatic (MetaMouse) Modelsfor Discovery and Development of Novel Chemotherapy

    Robert M. Hoffman ........................................................................... 297

    21 Preclinical Testing of Antileukemic DrugsUsing an In Vivo Model of Systemic Disease

    Richard B. Lock, Natalia L. Liem, and Rachael A. Papa ................... 323

    22 Assessing Growth and Responseto Therapy in Murine Tumor Models

    C. Patrick Reynolds, Bee-Chun Sun, Yves A. DeClerck,and Rex A. Moats .......................................................................... 335

    xii Contents of Vol. 2

  • 7/26/2019 Chemosensitivity Vol 1

    14/247

    23 Evaluation of Chemosensitivity of Micrometastatseswith Green Fluorescent ProteinGene-Tagged Tumor Models in Mice

    Hayao Nakanishi, Seiji Ito, Yoshinari Mochizuki,and Masae Tatematsu ................................................................... 351

    24 99mTc-Annexin A5 Uptake and Imaging to Monitor Chemosensitivity

    Tarik Z. Belhocine and Francis G. Blankenberg ............................... 363

    25 Magnetic Resonance Imagingof Tumor Response to Chemotherapy

    Richard Mazurchuk and Joseph A. Spernyak ................................... 381

    26 Metabolic Monitoring of Chemosensitivity with 18FDG PET

    Guy Jerusalem and Tarik Z. Belhocine ............................................. 417

    Index ............................................................................................................ 441

    Contents of Vol. 2 xiii

  • 7/26/2019 Chemosensitivity Vol 1

    15/247

  • 7/26/2019 Chemosensitivity Vol 1

    16/247

    I

    OVERVIEW

  • 7/26/2019 Chemosensitivity Vol 1

    17/247

  • 7/26/2019 Chemosensitivity Vol 1

    18/247

    3

    1

    An Overview of Chemosensitivity Testing

    Rosalyn D. Blumenthal

    SummaryThis overview chapter presents the importance of chemosensitivity testing for screening new

    therapeutic agents, identifying patterns of chemosensitivity for different types of tumors, estab-

    lishing patterns of cross-resistance and sensitivity in treatment naive and relapsing tumors; iden-

    tifying genomic and proteomic profiles associated with sensitivity; correlating in vitro response,

    preclinical in vivo effect, and clinical outcome associated with a particular therapeutic agent,

    and tailoring chemotherapy regimens to individual patients. Various assays are available to

    achieve these end points, including several in vitro clonogenic and proliferation assays, cell

    metabolic activity assays, molecular assays to monitor expression of markers for responsiveness,

    development of drug resistance and induction of apoptosis, in vivo tumor growth and survival

    assays in metastatic and orthotopic models, and in vivo imaging assays. The advantages and dis-

    advantages of the specific assays are discussed. A summary of research areas related to chemo-

    sensitivity testing is also included.

    Key Words

    Dose-response curve; IC50 values; imaging; metabolic assays; molecular markers; proliferation

    assays.

    1. Introduction

    Chemosensitivity testing is an ex vivo means of determining the cytotoxicand/or cytostatic, or apoptosis-inducing effect of anticancer drugs. The empha-

    sis on screening new agents derived from synthetic compound archives, and

    from pure natural products and their extracts, for antitumor activity necessi-

    tates in vitro evaluation in cell culture and then in vivo in appropriate tumor-

    bearing animal models. If the agent appears effective in this system, then the

    drug may be further evaluated in clinical trials. This paradigm seeks to identify

    the single best treatment to administer to the average patient with a given form

    of cancer through the use of prospective, randomized trials.

    From: Methods in Molecular Medicine, vol. 110: Chemosensitivity: Vol. 1: In Vitro AssaysEdited by: R. D. Blumenthal Humana Press Inc., Totowa, NJ

  • 7/26/2019 Chemosensitivity Vol 1

    19/247

    In addition to the standard, DNA-damaging and metabolic inhibiting agents

    in use, many new target classes now exist, such as kinase and nonkinase

    enzymes, transcriptional regulators, growth factor receptors, chemokines, angio-

    genic regulators, and proteinprotein interactions. Identifying which types of

    tumors (e.g., breast, lung, pancreas) and which subtypes (e.g., based on mor-

    phology, differentiation status, growth rate, drug resistance status, p53 expres-

    sion) within each tumor category will respond to each new agent is essential.

    Experience has shown that individual patients with a similar tumor histology do

    not necessarily respond identically to a given agent or set of agents. Defining

    the best treatment options, combinations, and schedules using standard agents

    alone or combined with new agents for an individual patient is an area of active

    investigation that requires both in vitro and in vivo testing. Chemosensitivityassays can potentially facilitate the individualization of patient treatment plans.

    Many retrospective and prospective studies for leukemia and for solid tumors

    have shown that drug resistance/sensitivity can be determined accurately by in

    vitro drug-response assays. As a consequence, knowing the drug sensitivity of

    a given tumor for a particular agent can significantly impact decision making

    and treatment planning. By identifying inactive drugs, patients can be spared

    standard chemotherapy regimens and their associated toxicities. In cases in

    which the patients tumor is unresponsive to chemotherapy, the oncologist maybe able to offer alternative or experimental treatments much sooner, when they

    may have a better chance of succeeding. Cell culture drug resistance testing

    refers to testing the resistance of a patients own cancer cells in the laboratory

    to drugs that may be used to treat the patients cancer (1). Table 1 summarizes

    the potential value of predictive chemosensitivity assays.

    When addressing the usefulness of an assay method, several questions must

    be addressed: (1) Are the drug sensitivity patterns observed in vitro with tumors

    of a particular histological type similar to those observed clinically for the sametumor type? (2) Can chemotherapy, which is selected on the basis of in vitro

    studies, improve patient survival? Efforts have been made to correlate in vitro

    drug sensitivity at initial diagnosis with 4-yr survival results, or in vitro resis-

    tance and early relapse. If this can be achieved, it may allow one to select more

    intensive regimens. (3) Do the results obtained in vitro predict those obtained

    in vivo? Since the process of tumor resection, transport, and processing for

    culture (mechanical or enzymatic disaggregation) introduces tissue stress and

    damage, which can perturb cell function and potentiate drug sensitivity, obtain-

    ing good correlations can be problematic. Another important consideration isthat tumor cell growth rate in vitro is likely to be much faster than in vivo.

    Therefore, the in vitro assay might indicate chemosensitivity in a situation

    of rapid cell division, which may be a false positive result, when attempting

    to translate results in vivo. Test systems using single-cell suspensions may

    4 Blumenthal

  • 7/26/2019 Chemosensitivity Vol 1

    20/247

    overestimate sensitivity. Finally, the ability to shrink tumors in vivo with a given

    treatment does not necessarily translate into a significant survival benefit.

    Prediction of chemosensitivity in the clinic is particularly challenging

    because drug responses reflect not only properties intrinsic to the target cell, but

    also host metabolic properties. Pharmacokinetic and pharmacodynamic vari-

    ables that affect drug action in vivo are not considered by in vitro assays.

    Because each patient has a unique pharmacogenetic makeup, leading to sig-nificant interpatient variations in drug half-life, volume of distribution, types of

    metabolites formed, and routes of elimination, correlating in vitro and in vivo

    results is often not a straightforward process. Furthermore, because some ther-

    apeutic agents (e.g., cyclophosphamide or CPT-11) are prodrugs that require

    metabolic activation, the in vitro modeling of in vivo tumor cell drug exposure

    becomes even more complex. However, by using in vitro models to address

    questions of chemosensitivity, one limits the study to cell-intrinsic properties

    found in cultured cells, which simplifies the system and focuses the initialinvestigation on tumor cell responsiveness.

    Many different methods are available for assessing chemosensitivity (24).

    In general, all assays generate dose-response curves where the dose of the drug

    is related to the percentage effect, such as tumor cell kill (Fig. 1). Determining

    the molar concentration that results in a 50% reduction in cell survival (IC50)

    can be used to compare the efficacy of different drugs in one tumor cell system

    or the same drug in different cell systems.

    The most common in vitro assays can be divided into one of three cate-

    gories: (1) clonogenic/proliferation assays, (2) assessment of cell metabolicactivity, and (3) measurement of cell membrane integrity. There has been much

    debate as to the characteristics of the best chemosensitivity assay. For exam-

    ple, should the assay measure colony formation or tumor cell proliferation?

    Should the assay be short term (hours to days) vs long term (days to weeks)?

    Chemosensitivity Testing 5

    Table 1

    Summary of Potential Value of Chemosensitivity Assays

    Conducting an initial screening of new therapeutic agentsTailoring chemotherapy regimens to the individual patient: determining tumors that

    are likely to respond to a particular agent and eliminating ineffective drugs

    Identifying patterns of chemosensitivity for different types and subtypes of tumors

    Establishing patterns of cross-resistance and sensitivity in treatment-naive and in

    relapsing patients

    Identifying genomic and proteomic profiles associated with sensitivity

    Correlating in vitro, preclinical in vivo, and clinical response to a therapeutic agent

  • 7/26/2019 Chemosensitivity Vol 1

    21/247

    Should the assay measure cytostatic or cytotoxic end points? Should the assay

    use a cell suspension or tumor microorgans? Should metabolic or morpholog-

    ical end points be used? Various assays measure different end points of cellu-

    lar damage. Morphological appearance is generally considered too insensitive.

    Measurements of biochemical parameters or reproductive capacity are likelyto be more reliable. Short-term assays may suggest that an agent is cytotoxic,

    but cells may recover. Thus, early indicators of drug-induced cell damage may

    provide misleading results. For an assay to be useful, the results must correlate

    with clinical response and survival; the end point of the assay must detect

    effects on cancer cells exclusive of other cellular elements such as fibroblasts,

    mesothelial cells, and endothelial cells; the turnaround time must meet clinical

    requirements; the test information must be easily interpreted and applied; and

    the test must be cost-effective.

    2. Clonogenic and Proliferation Assays

    The clonogenic method such as the human tumor colony-forming assay (5)

    is analogous to antibiotic sensitivity testing in bacteria. Single untreated or

    treated tumor cells are grown in Petri dishes in a soft agar system and colonies

    are counted after about 2 to 3 wk. Automated image analyzers now make this a

    much faster procedure. The use of agar allows most tumor cells to grow but

    prevents fibroblast proliferation. A reduction in colony number in treated groups

    reflects cytotoxicity of the agent toward the tumor cells. This is the gold stan-dard to which all other predictive assays have been compared for positive pre-

    dictive reliability (predicting patient response). Controlling the number of

    colonies per plate and including only colonies with at least 4050 cells is

    6 Blumenthal

    Fig. 1. Theoretical dose response curve.

  • 7/26/2019 Chemosensitivity Vol 1

    22/247

    important for obtaining accurate information from this assay. However, the

    method is complicated by the ability to obtain a single cell tumor suspension,

    adequate plating efficiency, proper growth in agar, and sufficient cell numbers

    to test multiple concentrations of drug. Variations on this assay include micro-

    clonogenic assays on tumor cells grown in a 96-well plate or in suspension

    (6). IC50 values are determined based on dose-response curves that fit the data

    to a linear quadratic equation.

    Several other in vitro short-term growth inhibition assays are in use, which

    are based on survival of tumor cell populations that have been in contact with

    a chemotherapeutic agent. In these assays, which are experimentally simpler

    than clonogenic assays, growth inhibition might not reflect true cell kill and can

    result in higher false positive results. However, a measure of chemosensitivitycan be obtained even when plating efficiencies are low. One such assay is the

    differential staining cytotoxicity assay (DiSC; [7]), which consists of incubat-

    ing dissociated cells from biopsy specimens in the presence or absence of a

    drug for 46 d, and using a dye such as fastgreen, which permeates only

    through dead cells. The ratio of dead cells to total cells is a measure of cell kill.

    In general, there has been good qualitative agreement between the DiSC assay

    and the clonogenic assay. Duration of the assay is relatively short, and the assay

    can be used on the majority of tumor specimens but is labor intensive and sub-ject to individual interpretation. Another method, the Kern assay (8), relies on

    uptake of radiolabeled precursor such as 3H-thymidine into the DNA of prolif-

    erating cells and is an example of similar assays that measure drug-induced

    inhibition of radioactive precursor incorporation into cellular macromole-

    cules (DNA, RNA, or protein) of single-cell suspensions or tumor slices. The

    assumption is that thymidine incorporation into DNA reflects cell division.

    There is a difference in view as to whether the assay is considered a reliable

    means of quantifying drug sensitivity. Some believe that it is not an accuratemeasure in biopsies because of contaminating nontumor cells and, in general,

    may be problematic because the effect on thymidine incorporation is not the

    same as measuring cell kill or growth inhibition (2). Others have reported sim-

    ilar results with the Kern assay to that obtained with clonogenic assays and

    significant correlations between clinical responses and depression of DNA syn-

    thesis in vitro (9). In general, this assay excels at negative predictive reliability

    (predicting drug resistance). A third approach based on proliferation is the col-

    lagen gel droplet drug sensitivity test, which is a quick and simple colorimet-

    ric quantitative approach using neutral red staining within collagen gel dropsthat are imaged on a videomicroscope (10). It affords the advantage of being

    able to eliminate the influence of fibroblasts in a mixed tumor/stroma popula-

    tion derived from a biopsy specimen.

    Chemosensitivity Testing 7

  • 7/26/2019 Chemosensitivity Vol 1

    23/247

    3. Cell Metabolic Activity Assays

    Assays that use various surrogate markers of cell number and viability have

    been used to measure cell survival in response to a therapeutic. These methodsinclude bioluminescence of adenosine triphosphate (ATP) (ATP-based tumor

    chemosensitivity assay [ATP-TCA]) (11) using the firefly luciferin-luciferase

    reagent; fluorometric microculture cytotoxicity assay (FMCA), which measures

    fluorescence generated from cellular hydrolysis of nonfluorescent fluorescein

    diacetate to fluorescein by viable cells in microtiter plates (12); coloration with

    sulforhodamine B (SRB), a general aminoxanthene dye for proteins, which

    binds electrostatically to basic amino acids; and coloration with reduction of a

    yellow tetrazolium dye such as MTT or MTS (13), substrates for mitochon-

    drial dehydrogenases, resulting in a blue-purple formazan product that reflects

    metabolic activity of living cells.

    The ATP-TCA is extremely sensitive because ATP levels are linear with the

    number of viable cells and correlate with cytotoxicity to a number of antineo-

    plastic agents in vitro. Assay results have also been used to predict clinical

    responses (14). This assay is done in a 96-well microplate and can use cell lines,

    or specimens from surgical needle biopsies, pleural or ascites fluid, and requires

    only 10,00020,000 cells/well (15). The FMCA is also a useful assay in that it

    is simple, is rapid, and seems to report clinically relevant cytotoxic drug sensi-tivity data. An alternative high-throughput fluorescence assay using enhanced

    green fluorescent protein (GFP) is available that can be analyzed by flow cytom-

    etry or by fluorescence microplate reader (16). It is a sensitive and rapid method

    to detect drug-induced cell death that provides results comparable to those

    obtained by other traditional apoptosis assays such as annexin-V binding, pro-

    pidium iodide incorporation, or reactive oxygen species production.

    The SRB assay provides a sensitive index of cellular protein content that is

    linear over a cell density range of two orders of magnitude, has a stable endpoint that does not require immediate range, and compares favorably with the

    MTT assay. However, the MTT assay is able to discriminate live cells from

    cellular debris, which the SRB assay cannot do. The drawbacks of assays such

    as the MTT are the sensitivity to the pH and glucose content of the media;

    chemical interferences with dye reduction; the quality of reagents used to sol-

    ubilize the formazan crystals; the need to read the assay immediately before the

    color fades; and the limited dynamic range, with a sensitivity of only 1-log cell

    kill (17). In addition, such assays do not differentiate between cytostatic and

    cytotoxic effects. In spite of these limitations, the MTT assay has been effec-

    tive at predicting the sensitivity of patients who have attained remission and of

    patient resistance (18). Both SRB and MTT assays provide rapid results and

    hold more promise in the screening and evaluation of potential new agents in

    8 Blumenthal

  • 7/26/2019 Chemosensitivity Vol 1

    24/247

    established tumor cell lines than in evaluating chemosensitivity of primary

    tumor specimens.

    Some researchers believe that results derived from single-cell preparations

    are not as accurate as those obtained from tumor fragments in which the three-

    dimensional structure of the tumor tissue is maintained (19). Two methods to

    assess chemosensitivity using tumor fragments in place of single cells have

    been utilized. The first is a variation on the MTT assay, called the histoculture

    drug response assay (20). The second approach, the fluorescent cytoprint assay

    (FCA), can be directly used on biopsy tissue without cell dissociation (21). In

    this assay, the tumor fragments are maintained in a collagen matrix during drug

    exposure. They are then treated with a fluorescent dye, which is visualized and

    photographed as cytoprints before and after drug treatment. This method hasbeen shown to have a very high positive and negative predictive accuracy.

    Another marketed in vitro assay that has been reported to have a high pre-

    dictive value is the extreme drug resistance assay (22). Tumor cells are exposed

    to suprapharmacological drug concentrations and either precursor incorporation

    or colony formation is measured. A drug resistance profile (extreme, intermedi-

    ate, or low) can be determined based on statistical comparison to a historical

    database of tumor specimens tested against the same panel of chemotherapeutic

    agents (23).An alternative approach to measuring cell proliferation or metabolic viabil-

    ity is to measure drug-induced DNA damage at the chromosome level using

    morphological criteria. The cytokinesis block micronucleus assay can reliably

    measure chromosome loss, chromosome breakage, chromosome rearrangement

    (nucleoplasmic bridges), cell division inhibition, necrosis, and apoptosis (24).

    All the previously summarized in vitro assays, and which are detailed in the

    chapters that follow, must ultimately provide a drug sensitivity index to put the

    results into the context of other similar assay results, or calculate a probability

    of response. Table 2 provides a summary of correlations of in vitro results with

    patient response (25). The most sensitive assays are the DiSc and the FCA,

    and the most specific are the clonogenic, MTT, and ATP assays. The ATP assay

    also gives the highest positive prediction, and the clonogenic assay, thymidine

    incorporation assay, DiSc and FCA are all highly accurate.

    4. In Vivo Assays

    In vitro assays in general are limited in that they do not take into account insuf-

    ficient drug absorption, inadequate drug distribution to the tumor owing to poor

    vascularization, pharmacological barriers, the need for drug activation, detoxifi-

    cation of drug by general metabolic pathways, differences in tumor growth rate in

    vitro and in vivo, or the problem of tumor heterogeneity. Therefore, assessing

    Chemosensitivity Testing 9

  • 7/26/2019 Chemosensitivity Vol 1

    25/247

    chemosensitivity in preclinical tumor-bearing animal models is essential. Pri-mary tumor models that have been utilized include human tumors grown sub-

    cutaneously in athymic nude mice, severe combined immunodeficient mice, or

    triple-deficient mice (bg/nu/xid mutations; [26]). Examples of metastasis

    models of human tumors include those grown orthotopically (27) or introduced

    systemically via iv, ip, intracardiac, intrasplenic, or intrahepatic injection or via

    inoculation of a cell suspension or tumor fragment into the mammary pad (28)

    or subrenal capsule (29). For primary growth, tumor size is followed by caliper

    measurement and tumor growth curves are constructed. Various formulas tomeasure tumor volume, area, and diameter have been used (30). Absolute tumor

    size, change in tumor size, percentage of growth inhibition, and area under the

    tumor growth curve have been reported. Using appropriate statistical methods

    to analyze tumor growth experiments that have sufficient power and low type

    I error rates is essential when performing in vivo chemosensitivity experiments

    (31). For metastasis models, median animal survival time, number and/or size

    of metastases after a fixed time, and weight of an organ containing metastases

    have been used as measures of response to a therapeutic agent. In addition todirect measurements of tumor growth, imaging approaches have been devel-

    oped to assess tumor chemosensitivity (32). These tools include the use of18FDG positron emission tomography (33), 99mTc-annexin-V scintigraphy (34),

    magnetic resonance imaging (35) and GFP (36).

    10 Blumenthal

    Table 2

    Correlation of In Vitro Results with Patient Responsea

    Type of Total Positive Accuracy Sensitivity Specificityassay (N) TP TN FP FN prediction (%) (%) (%) (%)

    Clonogenic 2300 512 1427 226 135 69 91 79 86

    Thymidine 494 123 232 119 20 51 92 86 66

    DiSC 510 247 175 72 16 77 92 94 71

    MTT 326 187 74 37 28 83 73 86 86

    ATP 129 74 37 6 12 93 76 86 86

    FCA 333 154 116 52 11 75 91 93 69

    Total 4092 1297 2061 512 222 75 86 87 74

    aSummary of clinical correlations from Table 7 in ref. 25. TP = true positivepatients who are

    sensitive in vitro and respond to therapy; TN = true negativepatients who are resistant in vitro and

    do not respond to therapy; FP = false positivepatients who are sensitive in vitro but resistant clin-

    ically; FN = false negativepatients who are resistant in vitro but respond clinically; positive pre-

    diction = TP/(TP + FP); accuracy = TN/(TN + FN); sensitivity = tests ability to detect clinically

    responsive patients = TP/(TP + FN); specificity = tests ability to detect clinically unresponsive

    patients = TN/(TN + FP).

  • 7/26/2019 Chemosensitivity Vol 1

    26/247

    5. Molecular Assays of Chemosensitivity

    The range of response to a particular cytotoxic agent can be quite substantial,

    and studies to understand the molecular parameters at the gene and at the pro-tein level that regulate chemosensitivity or resistance are an active area of inves-

    tigation (37). The molecular basis of sensitivity to chemotherapy is a complex

    product of cellular and tissue factors (3841). These include expression of the

    P-glycoprotein family of membrane transporters (e.g., MDR1, MRP, LRP), which

    decrease the intracellular accumulation of drug; changes in cellular proteins

    involved in detoxification (e.g., glutathione S-transferase , metallothioneins,

    NADP cytochrome P-450 reductase); changes in expression of molecules

    involved in DNA repair (e.g., O6-methylguanine DNA methyltransferase, DNA

    topoisomerase II, hMLH1, p21WAF1/CIP1); and activation oncogenes such as

    Her-2/neu, bcl-2, bcl-XL, c-myc, ras, c-jun, c-fos, MDM2, p210, BCR-abl, or

    mutant p53. In addition, expression of growth factors and receptors, proliferation

    markers (42), telomerase (43), enzymes that regulate intracellular ceramide

    levels (44), positive and negative regulators of apoptosis, and cell-cycle check-

    point controls are all important (45,46). For example, the form of p53 expression

    (null, wild type, or mutant and the location of the mutation) is known to affect

    chemosensitivity (47). Drug resistance can occur at the onset of the disease or

    can be acquired after previous chemotherapy. Methods that identify and detectexpression of drug resistance genes (48) or proteins (49) by flow cytometry

    (50), Western blotting, or immunohistochemistry (IHC) (51); or by induction of

    apoptosis by the DNA diffusion assay (52), diphenylamine assay to evaluate

    DNA fragmentation (53), TUNEL assay (54), flow cytometry with fluorescein

    isothiocyanate-annexin-V (34), PARP assay (55), or expression of BCl-2 family

    proteins including Bcl-2, Bcl-X(L), Bax, Bad, and Bak (56); or by changes in

    cell cycle (57) can be used as indirect measures of chemosensitivity.

    6. Genomic and Proteomic Approachesto Understand Chemosensitivity

    Previous efforts to use genetic information to predict drug sensitivity primar-

    ily have focused on individual genes that have broad effects, such as the mul-

    tidrug resistance genes mdr1 and mrp1 (58). There has been an effort to develop

    a genomics- and proteomics-based approach for the prediction of drug response

    (5961). An example of the clinical application of single-gene analysis as a pre-

    dictor of chemosensitivity is the evaluation of thymidylate synthase mRNAexpression from tumor core needle biopsies using real-time polymerase chain

    reaction analysis. Those patients with a clinical response to 5-fluorouracil (5-FU)

    therapy had a significantly lower level of expression of this gene. Expanding

    beyond single-gene effects on treatment response, the recent development of

    Chemosensitivity Testing 11

  • 7/26/2019 Chemosensitivity Vol 1

    27/247

    DNA microarrays permits large-scale screening of genes and the simultaneous

    measurement of the expression levels of thousands of genes and raises the pos-

    sibility of an unbiased, genomewide approach to understand the genetic basis

    of drug response (62). Algorithms have been developed for the classification

    of cell line chemosensitivity based on gene expression profiles. Using oligonu-

    cleotide microarrays, the expression levels of genes can be measured in a panel

    of cancer cell lines with known chemosensitivity profiles for various chemical

    compounds. Genes associated with repair processes, cell-cycle checkpoints,

    apoptosis, signal transduction, and metabolism can all be studied. Microarray

    analysis of baseline gene expression (63,64) and drug-induced changes in gene

    expression (65) have been successfully applied to predicting chemotherapy

    response. Further development of this technology will enable those responsiblefor treatment planning not only to predict chemosensitivity prior to therapy, but

    to answer whether the classifiers are dependent on tumor type or tumor class.

    Toward that goal, chemosensitivity prediction studies are being extended beyond

    cell line models to include the analysis of primary patient material.

    The addition of proteomic studies to genomic studies will further facilitate

    the ability to identify a priori sensitive and resistant tumors (66). In the past,

    protein analysis of formalin-fixed paraffin-embedded tumor tissue or Western

    blotting was used to assess prediction of therapeutic response. Techniques suchas IHC provide semiquantitative information on the level of expression of key

    proteins. Similar to mRNA results, thymidylate synthase expression at the pro-

    tein level has been a consistent predictor of response to 5-FU-based chemother-

    apy of metastatic colorectal cancer (67). Other protein markers that have been

    useful for predicting chemosensitivity include ornithine decarboxylase (68),

    HER-2/neu (69), the p27 cell-cycle regulatory protein (70), and Bcl-2 (71).

    Other proteins that have been studied but have not been predictive include p53

    and cerbB-2(72)

    . The use of tissue arrays allows IHC to be performed on0.6-mm core tissue sections from a large panel of tumor samples of varying his-

    totypes (73). The field is now expanding beyond detection and quantification of

    single proteins, to include evaluation of a sizable panel of proteins by combin-

    ing the tools of laser capture detection of tumor cells with silver stained two-

    dimensional gel electrophoresis and generate a tumor phenotype (74). Much

    of this effort has focused on the mass spectral identification of the thousands of

    proteins that populate complex biosystems. Protein patterns can be analyzed

    in hundreds of clinical samples per day utilizing this technology.

    7. Future Directions

    The availability of predictive in vitro and in vivo assays provides a mean of

    addressing many important issues in tumor biology. Several areas of investiga-

    12 Blumenthal

  • 7/26/2019 Chemosensitivity Vol 1

    28/247

    tion related to chemosensitivity testing are worthy of consideration. First, work

    is required to establish gene and protein profiles associated with drug sensitiv-

    ity (59). Studies are needed to address whether baseline gene/protein expression

    or drug-induced changes in expression are most predictive of response to treat-

    ment. Second, developing a better understanding of how to use drug combina-

    tions and schedules is essential. In vitro (75) and in vivo (76) models have

    been developed to assess interactions between multiple therapeutic agents.

    Toward that end, studies to determine how the use of one agent influences the

    biology of the surviving tumor cells and environmental milieu, and thus affects

    sensitivity to the second agent, are needed. Third, how chemosensitivity can

    be modulated, such as by using compounds that affect DNA repair (77); phar-

    macological agents (e.g., small molecule [78]); or genetic methods such as anti-sense, ribozyme, or RNA interference technology to overcome drug resistance

    (79) or antiapoptotic states must be determined. Fourth, the role that circadian

    rhythms play in chronotherapy (80) is just beginning to receive attention. In

    vivo designs to address how light/dark cycles impact the optimal time of day to

    administer a particular drug to achieve maximal efficacy are under investiga-

    tion. Fifth, tumor heterogeneity within a patient is important. Clinically, this is

    seen when a tumor mass in one area responds to chemotherapy, while a sepa-

    rate site may remain stable or progress. Intrapatient response variations havebeen evaluated by comparing the in vitro drug response of primary vs metasta-

    tic sites from the same patient. Thus, in vitro test results for solid tumors from

    one site may not be representative for other sites within the same patient. Addi-

    tional research to understand the molecular differences in chemosensitivity

    between primary and secondary sites is needed. Finally, developing approaches

    to individualize drug treatment as a function of tumor cell sensitivity (as has

    been done in infectious disease) requires further attention.

    References

    1. Kaspers, G., Zwaan, C., Pieters, R., and Veerman, A. (1999) Cellular drug resis-

    tance in childhood acute myeloid leukemia: a mini-review with emphasis on cell

    culture assays.Adv. Exp. Med. Biol. 457, 415421.

    2. Robert, J. (1999) Chemosensitivity testing: prediction of response to anticancer

    drugs using in vitro assays.Electronic J. Oncol. 2, 198210.

    3. Bellamy, W. (1992) Prediction of response to drug therapy of cancer: a review of

    in vitro assays.Drugs 44, 690708.

    4. Gercel-Taylor, C., Ackermann, M., and Taylor, D. (2001) Evaluation of cell pro-liferation and cell death based assays in chemosensitivity testing. Anticancer Res.

    21, 27612768.

    5. Von Hoff, D., Clark, G., Stogdill, B., et al. (1983) Prospective clinical trial of a

    human tumor cloning system. Cancer Res. 43, 19261936.

    Chemosensitivity Testing 13

  • 7/26/2019 Chemosensitivity Vol 1

    29/247

    6. Engblom, P., Rantanen, V., Kulmala, J., and Grenman, S. (1996) Paclitaxel and

    cisplatin sensitivity of ovarian carcinoma cell lines tested with a 96-well plate

    clonogenic assay.Anticancer Res. 16, 17431747.

    7. Weisenthal, L., Marsden, J., Dill, P., and Macaluso, C. (1983) A novel dye exclu-sion method for testing in vitro chemosensitivity of human tumors. Cancer Res.43,

    749757.

    8. Kern, D., Drozemuller, C., Kennedy, M., et al. (1985) Development of a minia-

    turized improved nucleic acid precursor incorporation assay for chemosensitivity

    testing of human solid tumors. Cancer Res. 45, 54365441.

    9. Sondak, V., Bertelson, C., Tanigawa, N., et al. (1984) Clinical correlations with

    chemosensitivities measured in a rapid thymidine incorporation assay. Cancer Res.

    46, 17251728.

    10. Kobayashi, H., Higashiyami, M., Minamigawa, K., et al. (2001) Examination of invitro chemosensitivity test using collagen gel droplet culture method with colori-

    metric endpoint quantitation.Jpn. J. Cancer Res. 92, 203210.

    11. Kangas, L., Gronroos, M., and Nieminen, A. (1984) Bioluminesence of cellular

    ATP: a new method for evaluating cytotoxic agents in vitro. Med. Biol. 62,

    338343.

    12. Csoka, K., Larsson, R., Tholander, B., Gerdin, E., de la Torre, M., and Nygren, P.

    (1994) Cytotoxic drug sensitivity testing of tumor cells from patients with ovarian

    carcinoma using the fluorometric microculture cytotoxicity assay (FMCA).

    Gynecol. Oncol. 54, 163170.13. Rubinstein, L., Shoemaker, R., Pacell, K., Simon, R., and Tosini, S. (1990) Com-

    parison of in vitro anticancer-drug-screening data generated with a tetrazolium

    assay versus a protein assay against a diverse panel of human tumor cells lines.

    J. Natl. Cancer Inst. 82, 11131118.

    14. Sevin, B., Peng, Z., Perras, J., Panalver, G., and Averette, H. Application of an

    ATP bioluminescence assay in human tumor chemosensitivity testing. Gynecol.

    Oncol. 31, 191204.

    15. Andreotti, P., Cree, I., Kurbacher, C., et al. (1995) Chemosensitivity testing of

    human tumors using a microplate adenosine triphosphate luminescence assay: clin-ical correlation for cisplatin resistance of ovarian carcinoma. Cancer Res., 55,

    52765282.

    16. Steff, A., Fortin, M., Arguin, C., and Hugo, P. (2001) Detection of a decrease in

    green fluorescent protein fluorescence for the monitoring of cell death: an assay

    amenable to high-throughput screening technologies. Cytometry 45, 237243.

    17. Vistica, D., Skehan, P., Scudiero, D., et al. (1991) Tetrazolium-based assays for

    cellular viability: a critical examination of selected parameters affecting formazan

    production. Cancer Res. 51, 25152520.

    18. Sargent, J. and Taylor, C. Appraisal of the MTT assay as a rapid test of chemosen-sitivity in acute myeloid leukemia.Br. J. Cancer60, 206210.

    19. Hoffman, R. (1991) Three-dimensional histoculture: origins and applications in

    cancer research. Cancer Cells 3, 8692.

    14 Blumenthal

  • 7/26/2019 Chemosensitivity Vol 1

    30/247

    20. Singh, B., Li, R., Xyu, L., et al. (2002) Prediction of survival in patients with head

    and neck cancer using the histoculture drug response assay.Head Neck24, 437442.

    21. Meitner, P. (1991) The fluorescence cytoprint assay: a new approach to in vitro

    chemosensitivity testing. Oncology 5, 7581.22. Kern, D. and Weisenthal, L. (1990) Highly specific prediction of antineoplastic

    drug resistance with an in vitro assay using supra-pharmacologic drug exposures.

    J. Natl. Cancer Inst. 82, 582588.

    23. Haroun, R., Clatterbuck, R., Gibbons, M., et al. (2002) Extreme drug resistance in

    primary brain tumors: in vitro analysis of 64 resection specimens.J. Neurooncol.

    58, 115123.

    24. Fenech, M. (2000) The in vitro micronucleus technique.Mutation Res.455, 8195.

    25. Fruhauf, J. and Bosanquet, A. (1993) In vitro determination of drug response: a

    discussion of clinical applications. PPO Updates 7, 121.26. Clarke, R. (1996) Human breast cancer cell line xenografts as models of breast

    cancer: the immunobiologies of recipient mice and the characteristics of several

    tumorigenic lines.Breast Cancer Res. Treat. 39, 6986.

    27. Hoffman, R. (1999) Orthotopic metastatic mouse models for anticancer drug dis-

    covery and evaluation: a bridge to the clinic.Invest. New Drugs 17, 343359.

    28. Price, J. (1996) Metastasis from human breast cancer cell lines. Breast Cancer

    Res. Treat. 39, 93102.

    29. Konovalova, N., Diatchkovskaya, R., Ganieva, L., Volkova, L., Lapshin, I.,

    Rudakov, B., Shaposhnikov, Y., and Shapiro, A. (1991) Subrenal capsule assay ofhuman tumor chemosensitivity.Neoplasma 38, 275284.

    30. Tomayko, M. and Reynolds, C. (1989) Determination of subcutaneous tumor size

    in athymic (nude) mice. Cancer Chemother. Pharmacol. 24, 148154.

    31. Heitjan, D., Manni, A., and Santen, R. (1993) Statistical analysis of in vivo tumor

    growth experiments. Cancer Res. 53, 60426050.

    32. Gibbs, J., Slocum, H., Cao, S., and Rustum, Y. (1999) Image analysis for quanti-

    tation of solid tumor drug sensitivity. Int. J. Surg. Invest.1, 133138.

    33. Kubota, K. (2001) From tumor biology to clinical PET: a review of positron

    emission tomography (PET) in oncology. Ann. Nucl. Med. 15, 471486.34. Belhocine, T., Steinmetz, N., Hustinx, R., et al. (2002) Increased uptake of the

    apoptosis imaging agent (99m)Tc recombinant human Annexin V in human tumors

    after one course of chemotherapy as a predictor of tumor response and patient

    prognosis. Clin. Cancer Res. 8, 27662774.

    35. Mazurchuk, R., Glaves, D., and Raghavan, D. (1997) Magnetic resonance imaging

    of response to chemotherapy in orthotopic xenografts of human bladder cancer.

    Clin. Cancer Res. 3, 16351641.

    36. Nakanishi, H., Mochizuki, Y., Kodera, Y., et al. (2003) Chemosensitivity of peri-

    toneal micrometastases as evaluated using green fluorescence protein(GFP)tagged human gastric cancer cell line. Cancer Sci. 94, 112118.

    37. Tewari, K. and Manetta, A. (1999) In vitro chemosensitivity testing and mecha-

    nisms of drug resistance. Curr. Oncol. Rep. 1, 7784.

    Chemosensitivity Testing 15

  • 7/26/2019 Chemosensitivity Vol 1

    31/247

    38. el-Deiry, W. S. (1997) Role of oncogenes in resistance and killing by cancer ther-

    apeutic agents. Curr. Opin. Oncol. 9, 7987.

    39. Fan, S., el-Deiry, W., Bae, I., et al. (1994) p53 gene mutations are associated with

    decreased sensitivity of human lymphoma cells to DNA damaging agents. CancerRes. 54, 58245830.

    40. Bosken, C., Wei, Q., Amos, C., and Spitz, M. (2002) An analysis of DNA repair as

    a determinant of survival in patients with non-small-cell lung cancer. J. Natl.

    Cancer Inst. 94, 10911099.

    41. Zunino, F., Perego, P., Pilotti, S., and Pratesi, G. (1997) Role of apoptotic response

    in cellular resistance to cytotoxic agents. Pharmacol. Ther. 76, 177185.

    42. Wang, Y., Ashkenazi, Y., and Bachrach, U. In vitro chemosensitivity of hemato-

    logical cancers: immunohistochemical detection of ornithine decarboxylase.Anti-

    cancer Drugs 10, 797805.43. Park, K., Rha, S., Kim, C., et al. (1998) Telomerase activity and telomere lengths

    in various cell lines: changes of telomerase activity can be another method for

    chemosensitivity evaluation.Int. J. Oncol. 13, 489495.

    44. Modrak, D., Rodriguez, M., Goldenberg, D., Lew, W., and Blumenthal, R. (2002)

    Sphingomyelin enhances chemotherapy efficacy and increases apoptosis in human

    colonic tumor xenografts.Int. J. Oncol. 20, 379384.

    45. Epstein, R. (1990) Drug-induced DNA damage and tumor chemosensitivity.

    J. Clin. Oncol. 8, 20622084.

    46. Xia, F. and Powell, S. (2002) The molecular basis of radiosensitivity andchemosensitivity in the treatment of breast cancer. Semin. Radiat. Oncol. 12,

    296304.

    47. Manahan, K., Taylor, D., and Gercel-Taylor, C. (2001) Clonal heterogeneity of

    p53 mutations in ovarian cancer. Int. J. Oncol. 19, 387394.

    48. Granjean, F., Bremaud, L., Verdier, M., Robert, J., and Ratinaud, M. (2001)

    Sequential gene expression of P-glycoprotein (P-gp), multidrug resistance associ-

    ated protein (MRP) and lung resistance protein: functional activity of P-gp and

    MRP present in the doxorubicin-resistant human K562 cell lines.Anticancer Drugs

    12, 247258.49. Michieli, M., Damiani, D., Ermacora, A., et al. (2000) P-glycoprotein (PGP), lung

    resistancerelated protein (LRP) and multidrug resistanceassociated protein

    (MRP) expression in acute promyelocytic leukemia. Br. J. Haematol. 108,

    703709.

    50. Pallis, M., Turzanski, J., Langabeer, S., and Russell, N. (1999) Reproducible flow

    cytometric methodology for measuring multidrug resistance in leukaemic blasts.

    Adv. Exp. Med. Biol. 457, 7788.

    51. Pall, G., Spitaler, M., Hofmann, J., Thaler, J., and Ludescher, C. (1997) Multidrug

    resistance in acute leukemia: a comparison of different diagnostic methods.Leukemia 11, 10671072.

    52. Singh, N. (2000) A simple method for accurate estimation of apoptotic cells.Exp.

    Cell. Res. 256, 328337.

    16 Blumenthal

  • 7/26/2019 Chemosensitivity Vol 1

    32/247

    53. Munshi, A., McDonnell, T., and Meyn, R. (2002) Chemotherapeutic agents

    enhance TRAIL-induced apoptosis in prostate cancer cells. Cancer Chemother.

    Pharmacol. 50, 4652.

    54. Maciorowski, Z., Klijanienko, J., Padoy, E., et al. (2001) Comparative image andflow cytometric TUNEL analysis of fine needle samples of breast carcinoma.

    Cytometry 46, 150156.

    55. Ogata, S., Okumura, K., and Taguchi, H. (2000) A simple and rapid method for the

    detection of poly(ADP-ribose) by flow cytometry. Biosci. Biotechnol. Biochem.

    64, 510515.

    56. Nita, M., Nagawa, H., Tominago, O., et al. (1998) 5-Fluorouracil induces apopto-

    sis in human colon cancer cell lines with modulation of Bcl-2 family proteins.

    Br. J. Cancer78, 986992.

    57. Smith, D., Gao, G., Zhang, X., Wang, G., and Dou, Q. (2000) Regulation of tumorcell apoptotic sensitivity during the cell cycle [review].Int. J. Mol. Med.6, 503507.

    58. Sonneveld, P. (2000) Multidrug resistance in hematological malignancies.J. Intern.

    Med. 247, 521534.

    59. Staunton, J., Slonim, D., Coller, H., et al. (2001) Chemosensitivity prediction by

    transcriptional profiling. Proc. Natl. Acad. Sci. USA 98, 10,78710,792.

    60. Bao, L. and Sun, Z. (2002) Identifying genes related to drug anticancer mecha-

    nisms using support vector machine. FEBS Lett. 521, 109114.

    61. McLeod, H. Individualized cancer therapy: molecular approaches to the predic-

    tion of tumor response.Expert Rev. Anticancer Ther. 2, 113119.62. Amundson, S., Myers, T., Scudiero, D., Kitada, S., Reed, J., and Fornace, A.

    (2000) An informatics approach identifying markers of chemosensitivity in human

    cancer cell lines. Cancer Res. 60, 61016110.

    63. Alizadeh, A., Eisen, M., and Dasvis, R. (2000) Distinct types of diffuse large

    B-cell lympoma identified by gene expression profiling.Nature403, 503511.

    64. Kihara, C., Tsunoda, T., Tanaka, T., et al. (2001) Prediction of sensitivity of

    esophageal tumors to adjuvant chemotherapy by cDNA microarray analysis of

    gene-expression profiles. Cancer Res. 61, 64746479.

    65. Sotirious, C., Powles, T., Dowsett, M., et al. (2002) Gene expression profilesderived from fine needle aspiration correlate with response to systemic chemother-

    apy in breast cancer.Breast Cancer Res. 4, R3.

    66. Poland, J., Schadendorf, D., Lage, H., Schnolzer, M., Celis, J., and Siha, P. (2002)

    Study of therapy resistance in cancer cells with functional proteome analysis. Clin.

    Chem. Lab. Med. 40, 221234.

    67. Aschele, C., Debernardis, D., Casazza, S., et al. (1999) Immunohistochemical

    quantitation of thymidylate synthase expression in colorectal cancer metastases

    predicts for clinical outcome to fluorouracil-based chemotherapy. J. Clin. Oncol.

    17, 17601770.68. Bachrach, U. and Wang, Y. (2003) In vitro chemosensitivity testing of hematolog-

    ical cancer patients: detection of ornithine decarboxylase.Rec. Results Cancer Res.

    161, 6270.

    Chemosensitivity Testing 17

  • 7/26/2019 Chemosensitivity Vol 1

    33/247

    69. Konecny, G., Fritz, M., Untch, M., et al. (2001) Her-2/neu overexpression and in

    vitro chemosensitivity to CMF and FEC in primary breast cancer. Breast Cancer

    Res. Treat. 69, 5363.

    70. Yang, Q., Sakurai, T., Yoshimura, G., et al. (2000) Overexpression of p27 proteinin human breast cancer correlates with in vitro resistance to doxorubicin and mit-

    omycin C.Anticancer Res. 20, 43194322.

    71. Yang, Q., Sakurai, T., Yoshimura, G., et al. (2000) Expression of Bcl-2 but not

    Bax or p53 correlates with in vitro resistance to a series of anticancer drugs in

    breast carcinoma.Breast Cancer Res. Treat. 61, 211216.

    72. Rozan, S., Vincent-Salomon, A., Zafrani, B., et al. (1998) No significant predictive

    value of c-erbB-2 or p53 expression regarding sensitivity to primary chemotherapy

    or radiotherapy in breast cancer. Int. J. Cancer79, 2733.

    73. Ginestier, C., Charafe-Jauffret, E., Bertucci, F., et al. (2002) Distinct and comple-mentary information provided by use of tissue and DNA microarrays in the study

    of breast tumor markers. Am. J. Pathol. 161, 12231233.

    74. Jones, M., Krutzsch, H., Shu, H., et al. (2002) Proteomic analysis and identifica-

    tion of new biomarkers and therapeutic targets for invasive ovarian cancer. Pro-

    teomics 2, 7684.

    75. Konecny, G., Untch, M., Slamon, D., et al. (2001) Drug interactions and cytotoxic

    effects of paclitaxel in combination with carboplatin, epirubicin, gemcitabine, or

    vinorelbine in breast cancer cell lines and tumor samples. Breast Cancer Res.

    Treat. 67, 223233.76. Lopez, A., Pegram, M., Slamon, D., and Landaw, E. (1999) A model-based

    approach for assessing in vivo combination therapy interactions. Proc. Natl. Acad.

    Sci. USA 96, 13,02313,028.

    77. Heim, M., Eberhardt, W., Seeber, S., and Muller, M. (2000) Differential modula-

    tion of chemosensitivity to alkylating agents and platinum compounds by DNA

    repair modulators in human lung cancer cell lines. J. Cancer Res. Clin. Oncol.

    126, 198204.

    78. Kondratov, R., Komarov, P., Becker, Y., Ewenson, A., and Gudkov, A. (2001)

    Small molecules that dramatically alter multidrug resistance phenotype by modu-lating the substrate specificity of P-glycoprotein. Proc. Natl. Acad. Sci. USA 98,

    14,07814,083.

    79. Henewisch-Becker, S. (1996) MDR1 reversal: criteria for clinical trials designed to

    overcome the multidrug resistance phenotype. Leukemia 10, S32S38.

    80. Levi, F., Giacchetti, S., Zidani, R., et al. (2001) Chronotherapy of colorectal cancer

    metastases.Hepatogastroenterology 48, 320322.

    18 Blumenthal

  • 7/26/2019 Chemosensitivity Vol 1

    34/247

    II

    IN VITRO MEASURES OF CHEMOSENSITIVITY

  • 7/26/2019 Chemosensitivity Vol 1

    35/247

  • 7/26/2019 Chemosensitivity Vol 1

    36/247

    21

    2

    Clonogenic Cell Survival Assay

    Anupama Munshi, Marvette Hobbs, and Raymond E. Meyn

    SummaryThe clonogenic cell survival assay determines the ability of a cell to proliferate indefinitely,

    thereby retaining its reproductive ability to form a large colony or a clone. This cell is then said

    to be clonogenic. A cell survival curve is therefore defined as a relationship between the dose

    of the agent used to produce an insult and the fraction of cells retaining their ability to reproduce.

    Although clonogenic cell survival assays were initially described for studying the effects of radi-

    ation on cells and have played an essential role in radiobiology, they are now widely used to

    examine the effects of agents with potential applications in the clinic. These include, in addition

    to ionizing radiation, chemotherapy agents such as etoposide and cisplatin, antiangiogenic agents

    such as endostatin and angiostatin, and cytokines and their receptors, either alone or in combi-

    nation therapy. Survival curves have been generated for many established cell lines growing in

    culture. One can use cell lines from various origins including humans and rodents; these cells can

    be neoplastic or normal. Because survival curves have wide application in evaluating the repro-

    ductive integrity of different cells, we provide here the steps involved in setting up a typical

    experiment using an established cell line in culture.

    Key Words

    Survival curve; cell survival; plating efficiency; radiation.

    1. IntroductionClonogenic cell survival is a basic tool that was described in the 1950s for

    the study of radiation effects. Much of the information that has been generated

    on the effect of radiation on mammalian cells has been obtained from clono-

    genic cell survival assays.

    Various mechanisms have been described for cell death; however, loss

    of reproductive integrity and the inability to proliferate indefinitely are the

    most common features. Therefore, a cell that retains its ability to synthesize pro-

    teins and DNA and go through one or two mitoses, but is unable to divide and

    From: Methods in Molecular Medicine, vol. 110: Chemosensitivity: Vol. 1: In Vitro AssaysEdited by: R. D. Blumenthal Humana Press Inc., Totowa, NJ

  • 7/26/2019 Chemosensitivity Vol 1

    37/247

    produce a large number of progeny is considered dead. This is very commonly

    referred to as loss of reproductive integrity or reproductive death and is the

    end point measured with cells in culture. On the other hand, a cell that is not

    reproductively dead and has retained the capacity to divide and proliferate

    indefinitely can produce a large clone or a large colony of cells and is then

    referred to as clonogenic. A cell survival curve describes a relationship

    between the insult-producing agent and the proportion of cells that survive.

    The ability of a single cell to grow into a large colony that can be visualized

    with the naked eye is proof that it has retained its capacity to reproduce. The

    loss of this ability as a function of dose of radiation or chemotherapy agent is

    described by the dose-survival curve. Most laboratories now extensively use

    established cell lines for studying the effects of various agents either alone orin combination. Therefore, the aim of this chapter is to go through the steps

    involved in setting up a typical clonogenic cell survival experiment using estab-

    lished cells lines growing as monolayer cultures.

    In brief, cells from an actively growing stock culture in monolayer are pre-

    pared in a suspension by the use of trypsin, which causes the cells to detach

    from the substratum. The number of cells per milliliter in this suspension is

    then counted using a hemocytometer or a Coulter counter. From this stock cul-

    ture, if 50 cells are seeded into a dish, e.g., and the dish is incubated for approx2 wk, each single cell divides many times and forms a colony that is easily

    visible with the naked eye, especially if it is fixed and stained (the steps

    involved in this process are briefly outlined in Fig. 1). All the cells that make

    up the colony are the progeny of a single cell. For the 50 cells seeded into

    the dish, the number of colonies counted may be anywhere from 0 to 50. One

    would ideally expect the number to be 50, but that is rarely the case for several

    possible reasons, including suboptimal growth medium, errors in counting the

    number of cells initially plated, and the loss of cells by trypsinization and gen-eral handling. The term plating efficiency (PE) indicates the percentage of cells

    seeded into a dish that finally grow to form a colony. Therefore, in the previ-

    ous example, if there are 25 colonies in the dish, then the PE becomes 50%. If

    a parallel dish is seeded with cells exposed to a dose of 6 Gy of gamma rays

    and incubated for approx 2 wk before being fixed and stained, then the fol-

    lowing may be observed:

    1. Some cells may remain single, not divide, and, in some cases, may show evi-

    dence of nuclear deterioration as they die by apoptosis. These cells would bescored as dead.

    2. Some cells may go through one or two divisions and form small colonies of

    just a few cells. These cells would be scored as dead.

    3. Some cells may form large colonies, indicating that the cells have survived the

    treatment and have retained the ability to reproduce indefinitely.

    22 Munshi et al.

  • 7/26/2019 Chemosensitivity Vol 1

    38/247

    2. Materials

    2.1. Preparation of Cell Lines Prior to Setting Up Clonogenic Assays

    1. Cell lines that need to be tested for their ability to form colonies.

    2. Complete growth medium as recommended by the manufacturer typically con-

    taining 10% fetal bovine serum plus antibiotics (penicillinstreptomycin) and glu-

    tamine. For every 500 mL of medium, add 55 mL of serum, 5 mL of 200 mM

    L-glutamine, and 5 mL of 10,000 U/mL penicillinstreptomycin solution. Medium

    should be stored at 4C but warmed to 37C prior to use.

    3. Trypsin-EDTA, to make single-cell suspensions from monolayer cultures. Store

    at 4C.

    4. Plasticware, for carrying out tissue culture including flasks (T-25 and T-75);

    100-mm dishes; and 5-, 10-, and 25-mL pipets.

    5. Micropipets and corresponding tips.

    6. 70% ethanol, for wiping the surface of the hood as well as the surface of all

    medium bottles prior to bringing them into the hood.7. Cidecon (detergent disinfectant with bactericidal and virucidal properties), to wipe

    the surface of the shelf on which the dishes will be incubated.

    8. Phosphate-buffered saline (PBS) (calcium magnesium free). Store at 4C.

    9. Isoton II (diluent for counting cells using a Coulter counter). Store at room

    temperature.

    Clonogenic Cell Survival Assay 23

    Fig. 1. Schematic representation of steps involved in setting up a clonogenic cell sur-

    vival assay.

  • 7/26/2019 Chemosensitivity Vol 1

    39/247

    2.2 Staining of Plates

    1. 0.5% Gentian Violet (made up in methanol). Store at room temperature in a dark

    bottle. Do not pour it down the sink.

    3. Methods

    3.1. Cells Growing in Monolayers or Attached Cells

    The procedure that we outline in this chapter is a basic protocol for setting

    up radiation clonogenic assays. However, this protocol can be modified to test

    the effect of different agents on the cell line of interest, either alone or in com-

    bination. These could include gene therapy vectors, chemotherapy agents, tyro-

    sine kinase inhibitors, antiangiogenic agentsbasically any agent that has to betested for its ability to affect reproductive cell death.

    3.2. Preparation of Cell Lines Prior to Setting UpClonogenic Cell Survival

    1. Label six T-25 flasks in preparation for setting each flask with a known number

    of cells as 0, 2, 4, and 6 Gy (depending on the experiment) for the various doses

    of radiation to be given. Add 5 mL of growth medium to the flasks and keep them

    aside in a hood.

    2. Trypsinize the stock flask of cells containing the cells that have to be tested fortheir radiosensitivity. Make sure that the cells are in single-cell suspension and

    obtain an accurate cell count. We use a Coulter counter to obtain a cell count. If

    a Coulter counter is not available, cells can be counted using a hemocytometer.

    Using a Pipettman, add 250,000 cells (the cell number can vary depending on the

    cell type) to the 5 mL of medium in each T-25 flask. Shake gently to distribute the

    cells evenly.

    3. Place the flasks in a 37C incubator set at 5% CO2 and be sure to leave the cap

    one thread loose so as to allow CO2 exchange (see Note 1).

    4. Allow the cells to settle and attach as a monolayer.

    3.3. Irradiation of Flasks and Performance of Plating Experimentfor Clonogenic Assay

    1. Prepare the hood and clean an incubator shelf. Because these cells are going to be

    left untouched in an incubator for up to 2 wk, and the possibility of contamination

    is high, clean the shelf thoroughly with Cidecon and 70% ethanol. Keep the

    cleaned shelf in the hood. Make sure that more than one bottle of complete

    medium is available; this experiment may require >500 mL of medium.

    2. Prepare the 100-mm dishes and 15-mL tubes in advance. One will need 100-mm

    dishes in triplicate, and two cell numbers will be plated for each dose of radiation.

    Because cells will be exposed to six doses of radiation, 36 dishes will be needed.

    Label the bottom of each dish as the lid, for the dishes will be loose during stain-

    ing, and the bottom is where the colonies will form, which is what will actually

    24 Munshi et al.

  • 7/26/2019 Chemosensitivity Vol 1

    40/247

    be counted. Label the first set of triplicate dishes as 0 Gy A and 0 Gy B (A and

    B for the two cell numbers to be used). Repeat this for all dose levels: 2, 4, 6, 8,

    and 10 Gy. Place 10 mL of complete medium in each dish. Place all dishes,

    stacked in threes, on the incubator shelf and put back in the incubator until readyfor plating.

    3. Place 18, 15-mL tubes on a clean rack three deep. Label the first tube 0Gy, 11;

    the next one 0 Gy 110; and the next 0 Gy 1100. Repeat this for all dose levels.

    Place 4.5 mL of complete medium in the back two tubes but not in the 11 dilu-

    tion tubes.

    4. Put the flasks on ice. Clean a rectangular tub and fill it halfway with ice. Remove

    the flasks from the incubator and close the caps tightly. Place the flasks on ice,

    and insert half depth into the ice. Tilt the flasks to the bottom so that the medium

    does not rest against the cap. Start a timer for 20 min.5. While waiting, prepare the Coulter counting vials. Label each as 0, 2, 4, and 6 Gy.

    Place 9.9 mL of Isoton (to be used for counting cells if using a Coulter counter)

    into each vial. Place Isoton in a control vial and run through the counter to get a

    background measurement; repeat until a satisfactory low background is obtained.

    Make sure that the Coulter is set to the appropriate size parameter for the cell line.

    6. When the 20-min time is up, irradiate the flasks using an appropriate irradiator

    according to the desired dose.

    7. Return to the hood, keeping the flasks on ice outside the hood. Begin the

    trypsinization procedure for each flask. Start with the 0-Gy flask. Aspirate themedium, rinse the cells gently with PBS, and then trypsinize. Place the harvested

    cells in the 15-mL tube labeled 0 Gy, 11. Make sure that you have a good single-

    cell suspension. From this cell suspension, take 100 L and place in the appro-

    priate counting vial for 0 Gy. Now trypsinize the next flask. While the flask is on

    the warming tray, count the previous counting vial and record the counts on a

    dilution sheet (see Fig. 2 for a setup of a dilution sheet that we commonly use in

    the laboratory). This expedites the experiments and lessens the chance of cell divi-

    sions taking place unequally during the time of trypsinization and counting.

    8. Continue this procedure until all the flasks have been trypsinized and counted.There should now be cells in each of the 11 dilution tubes, with a known number

    of cells/milliliter, all documented on the dilution sheet.

    9. Perform serial dilutions for each radiation dose so that the desired number of cells

    will be obtained by adding between 100 and 1000 L of volume to the dishes. If

    the number of cells needed requires a volume exceeding 1000 L, use a more

    concentrated dilution. Plate a number of cells consistent with obtaining a colony

    count of 50100. This may require only 100 cells for the control plate whereas at

    6 Gy this may require 4000 cells or more. Remember that the larger the insult to

    the flask (i.e., increasing radiation dose or increasing drug concentration if usingchemotherapy agents), the lower the plating efficiency and the more cells are

    needed to obtain the desired colony count (see Note 2).

    10. It is important to resuspend the cell pellet, which has probably settled to the bottom

    of the tube by the time all the flasks have been counted. Place 0.5 mL of the 11

    Clonogenic Cell Survival Assay 25

  • 7/26/2019 Chemosensitivity Vol 1

    41/247

    dilution into the 110 dilution tube (contai