Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-4hhp2 Total loading time: 0 Render date: 2024-05-01T09:13:27.652Z Has data issue: false hasContentIssue false

57 - Brain tumors

from Part 3.2 - Molecular pathology: cancers of the nervous system

Published online by Cambridge University Press:  05 February 2015

Chang-Hyuk Kwon
Affiliation:
Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Solid Tumor Program and Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
Dennis K. Burns
Affiliation:
Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
Luis F. Parada
Affiliation:
Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Primary brain tumors are neoplasms that originate from brain cells. In 2005, approximately 44 000 primary brain tumors were newly diagnosed and about 13 000 deaths were attributed to these types of tumors in the United States (1). They account for 1.4% of all cancers and 2.4% of all cancer-related deaths.

Our knowledge of brain tumors has expanded in recent years with new findings in tumor molecular genetics and biology. A greater appreciation of the singular nature of glioma in particular has become better appreciated, and new and important questions have arisen and begun to be addressed. For example, there is now compelling data supporting the concept of cancer stem cells in glioma and ideas concerning the identity of the tumor cell of origin have emerged. Physiologically relevant animal models have been developed that may reveal new insights into the etiology and development of these tumors. Innovative improvements in live imaging, neurosurgery, radiation, and chemotherapy have significantly extended survival time for patients with particular tumor types. In this chapter, we will briefly compare the characteristics of major brain neoplasms and describe recent advances in brain tumor research with an emphasis on potential therapies for malignant astrocytomas, the most common and most lethal brain tumor type.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 641 - 651
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

CBTRUS, Statistical Report: Primary Brain Tumors in the United States, 1998–2002. 2005: Central Brain Tumor Registry of the United States.
Zhu, Y, Parada, LF. The molecular and genetic basis of neurological tumours. Nature Reviews Cancer 2002;2:616–26.CrossRef
Maher, EA, Furnari, FB, Bachoo, RM, et al. Malignant glioma: genetics and biology of a grave matter. Genes and Development 2001;15:1311–33.CrossRef
Kleihues, P, Cavenee, WK. Pathology and genetics of tumours of the nervous system. In Kleihues P and Sobin LH, editors, World Health Organization Classification of Tumors. IARC Press: Lyon; 2000.
Stupp, R, Mason, WP, van den, Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. New England Journal of Medicine 2005;352:987–96.CrossRefGoogle ScholarPubMed
Ohgaki, H, Dessen, P, Jourde, B, et al. Genetic pathways to glioblastoma: a population-based study. Cancer Research 2004;64:6892–9.CrossRef
Maher, EA, Brennan, C, Wen, PY, et al. Marked genomic differences characterize primary and secondary glioblastoma subtypes and identify two distinct molecular and clinical secondary glioblastoma entities. Cancer Research 2006;66:11 502–13.
Yan, H, Parsons, DW, Jin, G, et al. IDH1 and IDH2 mutations in gliomas. New England Journal of Medicine 2009;360:765–73.CrossRefGoogle ScholarPubMed
TCGA Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 2008;455:1061–8.CrossRef
Parsons, DW, Jones, S, Zhang, X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008;321:1807–12.CrossRef
Shete, S, Hosking, FJ, Robertson, LB, et al. Genome-wide association study identifies five susceptibility loci for glioma. Nature Genetics 2009;41:899–904.CrossRef
Thiel, G, Marczinek, K, Neumann, R, et al. Somatic mutations in the neurofibromatosis 1 gene in gliomas and primitive neuroectodermal tumours. Anticancer Research 1995;15:2495–9.
Jensen, S, Paderanga, DC, Chen, P, et al. Molecular analysis at the NF1 locus in astrocytic brain tumors. Cancer 1995;76:674–7.3.0.CO;2-4>CrossRef
Reilly, KM, Loisel, DA, Bronson, RT, McLaughlin, ME, Jacks, T. Nf1;Trp53 mutant mice develop glioblastoma with evidence of strain-specific effects. Nature Genetics 2000;26:109–13.CrossRef
Zhu, Y, Guignard, F, Zhao, D, et al. Early inactivation of p53 tumor suppressor gene cooperating with NF1 loss induces malignant astrocytoma. Cancer Cell 2005;8:119–30.CrossRef
Kwon, CH, Zhao, D, Chen, J, et al. PTEN haploinsufficiency accelerates formation of high grade astrocytomas. Cancer Research 2008;68:1–9.
Alcantara, Llaguno S, Chen, J, Kwon, CH, et al. Malignant astrocytomas originate from neural stem/progenitor cells in a somatic tumor suppressor mouse model. Cancer Cell 2009;15:45–56.CrossRef
Freeman, CR, Farmer, JP. Pediatric brain stem gliomas: a review. International Journal of Radiation Oncology and Biological Physics 1998;40:265–71.CrossRefGoogle ScholarPubMed
Rutka, JT, Kuo, JS, Carter, M, et al. Advances in the treatment of pediatric brain tumors. Expert Review of Neurotherapeutics 2004;4:879–93.CrossRef
Kluwe, L, Hagel, C, Tatagiba, M, et al. Loss of NF1 alleles distinguish sporadic from NF1-associated pilocytic astrocytomas. Journal of Neuropathology and Experimental Neurology 2001;60:917–20.CrossRefGoogle ScholarPubMed
Lau, N, Feldkamp, MM, Roncari, L, et al. Loss of neurofibromin is associated with activation of RAS/MAPK and PI3-K/AKT signaling in a neurofibromatosis 1 astrocytoma. Journal of Neuropathology and Experimental Neurology 2000;59:759–67.CrossRefGoogle Scholar
Gutmann, DH, Donahoe, J, Brown, T, James, CD, Perry, A. Loss of neurofibromatosis 1 (NF1) gene expression in NF1-associated pilocytic astrocytomas. Neuropathology and Applied Neurobiology 2000;26:361–7.CrossRef
Hamilton, RL, Pollack, IF. The molecular biology of ependymomas. Brain Pathology 1997;7:807–22.CrossRef
Smyth, MD, Horn, BN, Russo, C, Berger, MS. Intracranial ependymomas of childhood: current management strategies. Pediatric Neurosurgery 2000;33:138–50.CrossRef
Reni, M, Gatta, G, Mazza, E, Vecht, C. Ependymoma. Critical Reviews in Oncology/Hematology 2007;63:81–9.CrossRef
Merchant, TE, Fouladi, M. Ependymoma: new therapeutic approaches including radiation and chemotherapy. Journal of Neurooncology 2005;75:287–99.CrossRefGoogle ScholarPubMed
Shu, HK, Sall, WF, Maity, A, et al. Childhood intracranial ependymoma: twenty-year experience from a single institution. Cancer 2007;110:432–41.CrossRef
Gilbertson, RJ, Bentley, L, Herna+n R, et al. ERBB receptor signaling promotes ependymoma cell proliferation and represents a potential novel therapeutic target for this disease. Clinical Cancer Research 2002;8:3054–64.
Suzuki, SO, Iwaki, T. Amplification and overexpression of mdm2 gene in ependymomas. Modern Pathology 2000;13:548–53.CrossRef
Polkinghorn, WR, Tarbell, NJ. Medulloblastoma: tumorigenesis, current clinical paradigm, and efforts to improve risk stratification. Nature Clinical Practice Oncology 2007;4:295–304.CrossRef
Fogarty, MP, Kessler, JD, Wechsler-Reya, RJ. Morphing into cancer: the role of developmental signaling pathways in brain tumor formation. Journal of Neurobiology 2005;64:458–75.CrossRefGoogle ScholarPubMed
Marino, S. Medulloblastoma: developmental mechanisms out of control. Trends in Molecular Medicine 2005;11:17–22.CrossRef
Knoepfler, PS, Kenney, AM. Neural precursor cycling at sonic speed: N-Myc pedals, GSK-3 brakes. Cell Cycle 2006;5:47–52.CrossRef
Kenney, AM, Cole, MD, Rowitch, DH. Nmyc upregulation by sonic hedgehog signaling promotes proliferation in developing cerebellar granule neuron precursors. Development 2003;130:15–28.CrossRef
Oliver, TG, Grasfeder, LL, Carroll, AL, et al. Transcriptional profiling of the Sonic hedgehog response: a critical role for N-myc in proliferation of neuronal precursors. Proceedings of the National Academy of Sciences USA 2003;100:7331–6.CrossRef
Rao, G, Pedone, CA, Valle, LD, et al. Sonic hedgehog and insulin-like growth factor signaling synergize to induce medulloblastoma formation from nestin-expressing neural progenitors in mice. Oncogene 2004;23:6156–62.CrossRef
Browd, SR, Kenney, AM, Gottfried, ON, et al. N-myc can substitute for insulin-like growth factor signaling in a mouse model of sonic hedgehog-induced medulloblastoma. Cancer Research 2006;66:2666–72.CrossRef
Read, TA, Fogarty, MP, Markant, SL, et al. Identification of CD15 as a marker for tumor-propagating cells in a mouse model of medulloblastoma. Cancer Cell 2009;15:135–47.CrossRef
Lee, A, Kessler, JD, Read, TA, et al. Isolation of neural stem cells from the postnatal cerebellum. Nature Neuroscience 2005;8:723–9.CrossRef
Singh, SK, Clarke, ID, Terasaki, M, et al. Identification of a cancer stem cell in human brain tumors. Cancer Research 2003;63:5821–8.
Singh, SK, Hawkins, C, Clarke, ID, et al. Identification of human brain tumour initiating cells. Nature 2004;432:396–401.CrossRef
Jeuken, JW, von Deimling, A, Wesseling, P. Molecular pathogenesis of oligodendroglial tumors. Journal of Neurooncology 2004;70:161–81.CrossRefGoogle ScholarPubMed
Baehring, JM. An update on oligodendroglial neoplasms. Current Opinion in Neurology 2005;18:639–44.CrossRef
Ueki, K. Oligodendroglioma: impact of molecular biology on its definition, diagnosis and management. Neuropathology 2005;25:247–53.CrossRef
Cairncross, JG, Macdonald, DR. Successful chemotherapy for recurrent malignant oligodendroglioma. Annals of Neurology 1988;23:360–4.CrossRef
Mason, WP, Krol, GS, DeAngelis, LM. Low-grade oligodendroglioma responds to chemotherapy. Neurology 1996;46:203–7.CrossRef
Olson, JD, Riedel, E, DeAngelis, LM. Long-term outcome of low-grade oligodendroglioma and mixed glioma. Neurology 2000;54:1442–8.CrossRef
Jaeckle, KA, Ballman, KV, Rao, RD, Jenkins, RB, Buckner, JC. Current strategies in treatment of oligodendroglioma: evolution of molecular signatures of response. Journal of Clinical Oncology 2006;24:1246–52.CrossRefGoogle Scholar
Dai, C, Celestino, JC, Okada, Y, et al. PDGF autocrine stimulation dedifferentiates cultured astrocytes and induces oligodendrogliomas and oligoastrocytomas from neural progenitors and astrocytes in vivo. Genes and Development 2001;15:1913–25.CrossRef
Riemenschneider, MJ, Perry, A, Reifenberger, G. Histological classification and molecular genetics of meningiomas. Lancet Neurology 2006;5:1045–54.CrossRef
Harada, T, Irving, RM, Xuereb, JH, et al. Molecular genetic investigation of the neurofibromatosis type 2 tumor suppressor gene in sporadic meningioma. Journal of Neurosurgery 1996;84:847–51.CrossRefGoogle ScholarPubMed
Ruttledge, MH, Xie, YG, Han, FY, et al. Deletions on chromosome 22 in sporadic meningioma. Genes, Chromosomes and Cancer 1994;10:122–30.CrossRef
Leone, PE, Bello, MJ, de Campos, JM, et al. NF2 gene mutations and allelic status of 1p, 14q and 22q in sporadic meningiomas. Oncogene 1999;18:2231–9.CrossRef
Merel, P, Hoang-Xuan, K, Sanson, M, et al. Predominant occurrence of somatic mutations of the NF2 gene in meningiomas and schwannomas. Genes, Chromosomes and Cancer 1995;13:211–16.CrossRef
Kalamarides, M, Niwa-Kawakita, M, Leblois, H, et al. Nf2 gene inactivation in arachnoidal cells is rate-limiting for meningioma development in the mouse. Genes and Development 2002;16:1060–5.CrossRef
Holland, EC. Progenitor cells and glioma formation. Current Opinion in Neurology 2001;14:683–8.CrossRef
Bachoo, RM, Maher, EA, Ligon, KL, et al. Epidermal growth factor receptor and Ink4a/Arf: convergent mechanisms governing terminal differentiation and transformation along the neural stem cell to astrocyte axis. Cancer Cell 2002;1:269–77.CrossRef
Phillips, HS, Kharbanda, S, Chen, R, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 2006;9:157–73.CrossRef
Xiao, A, Wu, H, Pandolfi, PP, Louis, DN, Van Dyke, T. Astrocyte inactivation of the pRb pathway predisposes mice to malignant astrocytoma development that is accelerated by PTEN mutation. Cancer Cell 2002;1:157–68.CrossRef
Jackson, EL, Garcia-Verdugo, JM, Gil-Perotin, S, et al. PDGFR alpha-positive B cells are neural stem cells in the adult SVZ that form glioma-like growths in response to increased PDGF signaling. Neuron 2006;51:187–99.CrossRef
Ligon, KL, Huillard, E, Mehta, S, et al. Olig2-regulated lineage-restricted pathway controls replication competence in neural stem cells and malignant glioma. Neuron 2007;53:503–17.CrossRef
Chen, J, Kwon, CH, Lin, L, Li, Y, Parada, LF. Inducible site-specific recombination in neural stem/progenitor cells. Genesis 2009;47:122–31.CrossRef
Taylor, MD, Poppleton, H, Fuller, C, et al. Radial glia cells are candidate stem cells of ependymoma. Cancer Cell 2005;8:323–35.CrossRef
Zlatescu, MC, TehraniYazdi, A, Sasaki, H, et al. Tumor location and growth pattern correlate with genetic signature in oligodendroglial neoplasms. Cancer Research 2001;61:6713–15.
Kros, JM, Pieterman, H, van Eden, CG, Avezaat, CJ. Oligodendroglioma: the Rotterdam-Dijkzigt experience. Neurosurgery 1994;34:959–66; discussion 966.
Rood, BR, MacDonald, T.J. Pediatric high-grade glioma: molecular genetic clues for innovative therapeutic approaches. Journal of Neurooncology 2005;75:267–72.CrossRefGoogle ScholarPubMed
Beier, D, Hau, P, Proescholdt, M, et al. CD133(+) and CD133(-) glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Research 2007;67:4010–15.CrossRef
Bao, S, Wu, Q, Sathornsumetee, S, et al. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Research 2006;66:7843–8.CrossRef
Bao, S, Wu, Q, McLendon, RE, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006;444:756–60.CrossRef
Li, Z, Bao, S, Wu, Q, et al. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell 2009;15:501–13.CrossRef
Bleau, AM, Hambardzumyan, D, Ozawa, T, et al. PTEN/PI3K/Akt pathway regulates the side population phenotype and ABCG2 activity in glioma tumor stem-like cells. Cell Stem Cell 2009;4:226–35.CrossRef
Holland, EC, Celestino, J, Dai, C, et al. Combined activation of Ras and Akt in neural progenitors induces glioblastoma formation in mice. Nature Genetics 2000;25:55–7.CrossRef
Uhrbom, L, Dai, C, Celestino, JC, et al. Ink4a-Arf loss cooperates with KRas activation in astrocytes and neural progenitors to generate glioblastomas of various morphologies depending on activated Akt. Cancer Research 2002;62:5551–8.
Ding, H, Roncari, L, Shannon, P, et al. Astrocyte-specific expression of activated p21-ras results in malignant astrocytoma formation in a transgenic mouse model of human gliomas. Cancer Research 2001;61:3826–36.
Wei, Q, Clarke, L, Scheidenhelm, DK, et al. High-grade glioma formation results from postnatal pten loss or mutant epidermal growth factor receptor expression in a transgenic mouse glioma model. Cancer Research 2006;66:7429–37.CrossRef
Charest, A, Wilker, EW, McLaughlin, ME, et al. ROS fusion tyrosine kinase activates a SH2 domain-containing phosphatase-2/phosphatidylinositol 3-kinase/mammalian target of rapamycin signaling axis to form glioblastoma in mice. Cancer Research 2006;66:7473–81.CrossRef
Zheng, H, Ying, H, Yan, H, et al. p53 and Pten control neural and glioma stem/progenitor cell renewal and differentiation. Nature 2008;455:1129–33.CrossRef
Marumoto, T, Tashiro, A, Friedmann-Morvinski, D, et al. Development of a novel mouse glioma model using lentiviral vectors. Nature Medicine 2009;15:110–16.CrossRef
Wang, Y, Yang, J, Zheng, H, et al. Expression of mutant p53 proteins implicates a lineage relationship between neural stem cells and malignant astrocytic glioma in a murine model. Cancer Cell 2009;15:514–26.CrossRef
Romer, JT, Kimura, H, Magdaleno, S, et al. Suppression of the Shh pathway using a small molecule inhibitor eliminates medulloblastoma in Ptc1(+/–)p53(–/–) mice. Cancer Cell 2004;6:229–40.CrossRef
Rudin, CM, Hann, CL, Laterra, J, et al. Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. New England Journal of Medicine 2009;361:1173–8.CrossRefGoogle ScholarPubMed
Yauch, RL, Dijkgraaf, GJ, Alicke, B, et al. Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science 2009;326:572–4.CrossRef
Fan, QW, Knight, ZA, Goldenberg, DD, et al. A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma. Cancer Cell 2006;9:341–9.CrossRef
Nicholas, MK, Lukas, RV, Jafri, NF, Faoro, L, Salgia, R. Epidermal growth factor receptor – mediated signal transduction in the development and therapy of gliomas. Clinical Cancer Research 2006;12:7261–70.CrossRef
Collins, VP. Mechanisms of disease: genetic predictors of response to treatment in brain tumors. Nature Clinical Practice Oncology 2007;4:362–74.CrossRef
Zhao, C, Blum, J, Chen, A, et al. Loss of beta-catenin impairs the renewal of normal and cml stem cells in vivo. Cancer Cell 2007;12:528–41.CrossRef
Yanagi, S, Kishimoto, H, Kawahara, K, et al. PTEN controls lung morphogenesis, bronchioalveolar stem cells, and onset of lung adenocarcinomas in mice. Journal of Clinical Investigation 2007;117:2929–40.CrossRefGoogle ScholarPubMed
Yilmaz, OH, Valdez, R, Theisen, BK, et al. PTEN dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature 2006;441:475–82.CrossRef
Liu, S, Dontu, G, Mantle, ID, et al. Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Research 2006;66:6063–71.CrossRef
Piccirillo, SG, Reynolds, BA, Zanetti, N, et al. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature 2006;444:761–5.CrossRef
Clement, V, Sanchez, P, de Tribolet, N, Radovanovic, I, Ruiz i, Altaba A. HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Current Biology 2007;17:165–72.CrossRef
Penuelas, S, Anido, J, Prieto-Sanchez, RM, et al. TGF-beta increases glioma-initiating cell self-renewal through the induction of LIF in human glioblastoma. Cancer Cell 2009;15:315–27.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Brain tumors
    • By Chang-Hyuk Kwon, Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Solid Tumor Program and Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA, Dennis K. Burns, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA, Luis F. Parada, Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.058
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Brain tumors
    • By Chang-Hyuk Kwon, Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Solid Tumor Program and Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA, Dennis K. Burns, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA, Luis F. Parada, Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.058
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Brain tumors
    • By Chang-Hyuk Kwon, Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Solid Tumor Program and Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA, Dennis K. Burns, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA, Luis F. Parada, Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.058
Available formats
×