Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-27T07:29:48.578Z Has data issue: false hasContentIssue false

10 - Oncologic Drugs

from II - Therapeutic Areas

Published online by Cambridge University Press:  05 June 2012

Russ B. Altman
Affiliation:
Stanford University, California
David Flockhart
Affiliation:
Indiana University
David B. Goldstein
Affiliation:
Duke University, North Carolina
Get access

Summary

Pharmacogenomics is of particular importance in oncology, a medical subspecialty characterized by rapidly lethal diseases and drugs with narrow therapeutic indices and significant toxicities. Identification of individuals likely to respond to or experience toxicity from a given chemotherapeutic agent, will have significant impact on outcomes, particularly in the field of oncology. Several models currently exist for discovery of pharmacogenomic markers in oncology. Phenotypic variations may range from variability in response as measured by survival or time to progression, to variability in toxicity in individuals treated with a particular agent. Measurements of toxicity can be a challenge to quantify in individuals because of interobserver variability. Lymphoblastoid cell lines (LCLs) and the NCI60 bank of tumor cell lines have been used as models for clinical phenotypes. To date, there are several examples of germline polymorphisms and somatic mutations that predict likelihood of response and/or toxicity from chemotherapeutic agents. A pattern of interethnic variability in response and toxicity has been observed for some chemotherapeutic agents, and the associated field of pharmacoethnicity is likely to contribute to our understanding of pharmacogenomics.

Pharmacogenomics has found extensive application in the field of oncology and is likely to remain an important tool in the race toward personalized medicine. Individualization of therapies is of particular importance in oncology because of several unique features of cancer treatment. First, most oncologic therapies have potential for organ toxicity and typically give rise to an array of potential life-threatening side effects. For example, taxanes are highly efficacious against malignancies of the lung, breast, ovary, and head and neck, but are also associated with significant toxicities such as myelosuppression and peripheral neuropathy. Identification of individuals unlikely to respond to taxane therapy a priori will be tremendously important in therapeutic decision making, because alternative therapies can be considered, thereby reducing the likelihood of unnecessary toxicity. Second, many oncologic diseases progress rapidly and are generally lethal in the absence of effective therapy. Consequently, prompt diagnosis and early institution of efficacious therapies is of paramount importance. In the absence of knowledge about predictors of response, individuals could be subjected to therapies to which their tumors might not respond, resulting in further disease progression. With more advanced disease and organ dysfunction, some therapies may no longer be given safely and may only serve a palliative rather than a curative role. For example, a five-year period of adjuvant tamoxifen therapy following successful treatment of early-stage estrogen receptor (ER)-positive breast cancer in a premenopausal woman is associated with a reduction in the rate of disease recurrence and mortality. A poor metabolizer phenotype results in insufficient conversion of tamoxifen to endoxifen and an increased risk of disease relapse and progression (1). Affected individuals may be better served by alternative antiestrogen maneuvers such as the combination of ovarian ablation and aromatase inhibitor therapy. Third, most chemotherapeutic agents have a fairly narrow therapeutic index (see Figure 10.1). The therapeutic index of a drug compares the dose that produces toxicity with the dose that produces the desired effect and, as such, provides a measure of the drug's safety. Given the significant likelihood of an adverse effect even within the therapeutic window, treatment with a particular agent is best reserved for individuals likely to respond, with the careful weighing of risks and benefits and informed decision making on the part of the patient. Finally, the expenses associated with oncologic therapies necessitate avoidance of therapy-related morbidity that further increases the likelihood of hospitalization and overall cost of care. For example, trastuzumab is an agent used in the treatment of HER2neu-positive breast cancer, is typically infused on a three-weekly schedule for at least one year, and may cost as much as $70,000 for a full course of therapy (2). Given all the aforementioned, it is not surprising that current pharmacogenomic research is dominated by investigation of variability in response to, and toxicity from, oncologic therapies (3).

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2012

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

Hoskins, JMCarey, LAMcLeod, HLCYP2D6 and tamoxifen: DNA matters in breast cancerNat Rev Cancer. 2009 9 576Google Scholar
Fleck, LMThe costs of caring: who pays? who profits? who panders?Hastings Cent Rep. 2006 36 13Google Scholar
Holmes, MVShah, TVickery, CSmeeth, LHingorani, ADCasas, JPFulfilling the promise of personalized medicine? Systematic review and field synopsis of pharmacogenetic studiesPLoS. 2009 4Google Scholar
Devereux, TRRisinger, JIBarrett, JC 1999 19
Olivier, MHollstein, MHainaut, PTP53 mutations in human cancers: origins, consequences, and clinical useCold Spring Harb Perspect Biol. 2010 2 1008Google Scholar
Chung, JHChoe, GJheon, SSung, S-WKim, TJLee, KWLee, JHLee, C-TEpidermal growth factor receptor mutation and pathologic-radiologic correlation between multiple lung nodules with ground-glass opacity differentiates multicentric origin from intrapulmonary spreadJ Thorac Oncol. 2009 4 1490Google Scholar
Bell, DWOur changing view of the genomic landscape of cancerJ Pathol. 2010 220 231Google Scholar
Ikediobi, ONSomatic pharmacogenomics in cancerPharmacogenomics J. 2008 8 305Google Scholar
Bernig, TChanock, SChallenges of SNP genotyping and genetic variation: its future role in diagnosis and treatment of cancerExpert Rev Mol Diagn. 2006 6 319Google Scholar
Yang, JJCheng, CYang, WPei, DCao, XFan, YPounds, SBNeale, GTrevino, LRFrench, DCampana, DDowning, JREvans, WEGenome-wide interrogation of germline genetic variation associated with treatment response in childhood acute lymphoblastic leukemiaJAMA. 2009 301 393Google Scholar
Soo, RAWang, LZNg, SSChong, PYYong, WPLee, SCLiu, JJChoo, TBTham, LSLee, HSGoh, BCSoong, RDistribution of gemcitabine pathway genotypes in ethnic Asians and their association with outcome in non-small cell lung cancer patientsLung Cancer. 2009 63 121Google Scholar
McWhinney, SRMcLeod, HLUsing germline genotype in cancer pharmacogenetic studiesPharmacogenomics. 2009 10 489Google Scholar
Trotti, AColevas, ADSetser, ARusch, VJacques, DBudach, VLanger, CMurphy, BCumberlin, RColeman, CNRubin, PCTCAE v3.0: development of a comprehensive grading system for the adverse effects of cancer treatmentSemin Radiat Oncol. 2003 13 176Google Scholar
Therasse, PArbuck, SGEisenhauer, EAWanders, JKaplan, RSRubinstein, LVerweij, JVan Glabbeke, Mvan Oosterom, ATChristian, MCGwyther, SGNew guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of CanadaJ Natl Cancer Inst. 2000 92 205Google Scholar
Santos, FPRavandi, FAdvances in treatment of chronic myelogenous leukemia – new treatment options with tyrosine kinase inhibitorsLeuk Lymphoma. 2009 50 16Google Scholar
Kantarjian, HSchiffer, CJones, DCortes, JMonitoring the response and course of chronic myeloid leukemia in the modern era of BCR-ABL tyrosine kinase inhibitors: practical advice on the use and interpretation of monitoring methodsBlood. 2008 111 1774Google Scholar
Thorn, CFKlein, TEAltman, RBPharmacogenomics and bioinformatics: PharmGKBPharmacogenomics. 2010 11 501Google Scholar
Welsh, MMangravite, LMedina, MWTantisira, KZhang, WHuang, RSMcLeod, HDolan, MEPharmacogenomic discovery using cell-based modelsPharmacol Rev. 2009 61 413Google Scholar
Shukla, SJDolan, MEUse of CEPH and non-CEPH lymphoblast cell lines in pharmacogenetic studiesPharmacogenomics. 2005 6 303Google Scholar
Masson, ERelling, MVSynold, TWLiu, QSchuetz, JDSandlund, JTPui, CHEvans, WEAccumulation of methotrexate polyglutamates in lymphoblasts is a determinant of antileukemic effects in vivo. A rationale for high-dose methotrexateJ Clin Invest. 1996 97 73Google Scholar
Hartford, CMDuan, SDelaney, SMMi, SKistner, EOLamba, JKHuang, RSDolan, MEPopulation-specific genetic variants important in susceptibility to cytarabine arabinoside cytotoxicityBlood. 2009 113 2145Google Scholar
Lennard, LLilleyman, JSVan Loon, JWeinshilboum, RMGenetic variation in response to 6-mercaptopurine for childhood acute lymphoblastic leukaemiaLancet. 1990 336 225Google Scholar
Hoskins, JMGoldberg, RMQu, PIbrahim, JGMcLeod, HLUGT1A1*28 genotype and irinotecan-induced neutropenia: dose mattersJ Natl Cancer Inst. 2007 99 1290Google Scholar
Goetz, MPRae, JMSuman, VJSafgren, SLAmes, MMVisscer, DWReynolds, CCouch, FJLingle, WLPharmacogenetics of tamoxifen biotransformation is associated with clinical outcomes of efficacy and hot flashesJ Clin Oncol. 2005 23 9312Google Scholar
Hartford, CMDolan, MEIdentifying genetic variants that contribute to chemotherapy-induced cytotoxicityPharmacogenomics. 2007 8 1159Google Scholar
Huang, RSRatain, MJPharmacogenetics and pharmacogenomics of anticancer agentsCA Cancer J Clin. 2009 59 42Google Scholar
Zhang, WDolan, MEImpact of the 1000 genomes project on the next wave of pharmacogenomic discoveryPharmacogenomics. 2010 11 249Google Scholar
International Human Genome Sequencing ConsortiumFinishing the euchromatic sequence of the human genomeNature. 2004 431 931Google Scholar
International HapMap ConsortiumThe International HapMap ProjectNature. 2003 426 789Google Scholar
Galmarini, CMThomas, XGraham, KEl Jafaari, ACros, EJordheim, LMackey, JRDumontet, CDeoxycytidine kinase and cN-II nucleotidase expression in blast cells predict survival in acute myeloid leukaemia patients treated with cytarabineBr J Haematol. 2003 122 53Google Scholar
Estey, EPlunkett, WDixon, DKeating, MMcCredie, KFreireich, EJVariables predicting response to high dose cytosine arabinoside therapy in patients with refractory acute leukemiaLeukemia. 1987 1 580Google Scholar
Raza, AGezer, SAnderson, JLykins, JBennett, JBrowman, GGoldberg, JLarson, RVogler, RPreisler, HDRelationship of [3H]Ara-C incorporation and response to therapy with high-dose Ara-C in AML patients: a Leukemia Intergroup studyExp Hematol. 1992 20 1194Google Scholar
Ling, PDHuls, HMIsolation and immortalization of lymphocytesCurr Protoc Mol Biol. 2005 22Google Scholar
Dolan, MENewbold, KGNagasubramanian, RWu, XRatain, MJCook, EHBadner, JAHeritability and linkage analysis of sensitivity to cisplatin-induced cytotoxicityCancer Res. 2004 64 4353Google Scholar
Watters, JWKraja, AMeucci, MAProvince, MAMcLeod, HLGenome-wide discovery of loci influencing chemotherapy cytotoxicityProc Natl Acad Sci USA. 2004 101 11809Google Scholar
Huang, RSDuan, SShukla, SJKistner, EOClark, TAChen, TXSchweitzer, ACBlume, JEDolan, MEIdentification of genetic variants contributing to cisplatin-induced cytotoxicity by use of a genomewide approachAm J Hum Genet. 2007 81 427Google Scholar
Duan, SBleibel, WKHuang, RSShukla, SJWu, XBadner, JADolan, MEMapping genes that contribute to daunorubicin-induced cytotoxicityCancer Res. 2007 67 5425Google Scholar
Choy, EYelensky, RBonakdar, SPlenge, RMSaxena, RDe Jager, PLShaw, SYWolfish, CSSlavik, JMCotsapas, CRivas, MDermitzakis, ETCahir-McFarland, EKieff, EHafler, DDaly, MJAltshuler, DGenetic analysis of human traits in vitro: drug response and gene expression in lymphoblastoid cell linesPLoS Genet. 2008 4 e1000287Google Scholar
Stark, ALZhang, WMi, SDuan, SO’Donnell, PHHuang, RSDolan, MEHeritable and non-genetic factors as variables of pharmacologic phenotypes in lymphoblastoid cell linesPharmacogenomics. 2010 10 505Google Scholar
Shoemaker, RHThe NCI60 human tumour cell line anticancer drug screenNat Rev Cancer. 2006 6 813Google Scholar
Jarjanazi, HKeifer, JSavas, SBriollais, LTuzmen, SPabalan, NIbrahim-Zada, IMousses, SOzcelik, HDiscovery of genetic profiles impacting response to chemotherapy: application to gemcitabineHum Mutat. 2008 29 461Google Scholar
Mori, SChang, JTAndrechek, ERPotti, ANevins, JRUtilization of genomic signatures to identify phenotype-specific drugsPLoS. 2009 4Google Scholar
Li, LFridley, BLKalari, KJenkins, GBatzler, AWeinshilboum, RMWang, LGemcitabine and arabinosylcytosin pharmacogenomics: genome-wide association and drug response biomarkersPLoS. 2009 4Google Scholar
Jensen, EVJordan, VThe estrogen receptor: a model for molecular medicineClin Cancer Res. 2003 9 1980Google Scholar
King, MCWieand, SHale, KLee, MWalsh, TOwens, KTait, JFord, LDunn, BKConstantino, JWickerham, LWolmark, NFisher, BTamoxifen and breast cancer incidence among women with inherited mutations in BRCA1 and BRCA2JAMA. 2001 286 2251Google Scholar
Ingelman-Sundberg, MSim, SCGomez, ARodriguez-Antona, CInfluence of cytochrome P450 polymorphisms on drug therapies: pharmacogenetic, pharmacoepigenetic and clinical aspectsPharmacol Ther. 2007 116 496Google Scholar
http://www.fda.gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/ucm083378.htm
Furuta, TPharmacogenomics in chemotherapy for GI tract cancerJ Gastroenterol. 2009 44 1016Google Scholar
Wong, SFCetuximab: an epidermal growth factor receptor monoclonal antibody for the treatment of colorectal cancerClin Ther. 2005 27 684Google Scholar
Ando, YSaka, HAsai, GSugiura, SShimokata, KKamataki, TUGT1A1 genotypes and glucuronidation of SN-38, the active metabolite of irinotecanAnn Oncol. 1998 9 845Google Scholar
Innocenti, FUndevia, SDIyer, LChen, PXDas, SKocherginsky, MKarrison, TJanisch, LRamirez, JRudin, CMVokes, EERatain, MJGenetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecanJ Clin Oncol. 2004 22 1382Google Scholar
Nagar, SRemmel, RPUridine disphosphoglucuronosyltransferase pharmacogenetics and cancerOncogene. 2006 25 1659Google Scholar
Ramchandani, RPWang, YBooth, BPIbrahim, AJohnson, JRRahman, AMehta, MInnocenti, FRatain, MJGobburu, JVThe role of SN-38 exposure, UGT1A1*28 polymorphism, and baseline bilirubin level in predicting severe irinotecan toxicityJ Clin Pharmacol. 2007 47 78Google Scholar
Walter, AJohnstone, ESwanton, CMidgley, RTomlinson, IKerr, DGenetic prognostic and predictive markers in colorectal cancerNat Rev Cancer. 2009 9 489Google Scholar
Yen, JLMcLeod, HLShould DPD analysis be required prior to prescribing fluoropyrimidinesEur J Cancer. 2007 43 1011Google Scholar
Van Kuilenburg, ABVreken, PAbeling, NGBakker, HDMeinsma, RVan Lenthe, HDe Abreu, RASmeitink, JAKayserili, HApak, MYGenotype and phenotype in patients with dihydropyrimidine dehydrogenase deficiencyHum Genet. 1999 104 1Google Scholar
Sahasranaman, SHoward, DRoy, SClinical pharmacology and pharmacogenetics of thiopurinesEur J Clin Pharmacol. 2008 64 753Google Scholar
O’Donnell, PHDolan, MECancer pharmacoethnicity: ethnic differences in susceptibility to the effects of chemotherapyClin Cancer Res. 2009 15 4806Google Scholar
Peters, EJKraja, ATLin, SJYen-Revollo, JLMarsh, SProvince, MAMcLeod, HLAssociation of thymidylate synthase variants with 5-fluorouracil cytotoxicityPharmacogenet Genomics. 2009 19 399Google Scholar
Bleibel, WKDuan, SHuang, RSKistner, EOShukla, SJWu, ZBadner, JADolan, MEIdentification of genomic regions contributing to etoposide-induced cytotoxicityHum Genet. 2009 125 173Google Scholar
Shukla, SJDuan, SBadner, JAWu, XDolan, MESusceptibility loci involved in cisplatin-induced cytotoxicity and apoptosisPharmacogenet Genomics. 2008 18 253Google Scholar
Huang, RSDuan, SKistner, EOBleibel, WKDelaney, SMFackenthal, DLDas, SDolan, MEGenetic variants contributing to daunorubicin-induced cytotoxicityCancer Res. 2008 68 3161Google Scholar
Huang, RSDuan, SKistner, EOHartford, CMDolan, MEGenetic variants associated with carboplatin-induced cytotoxicity in cell lines derived from AfricansMol Cancer Ther. 2008 7 3038Google Scholar
Morvan, VBellott, RMoisan, FMathoulin-Pelissier, SBonnet, JRobert, JRelationships between genetic polymorphisms and anticancer drug cytotoxicity vis-a-vis the NCI-60 panelPharmacogenomics. 2006 7 843Google Scholar
Puyo, SLe Morvan, VRobert, JImpact of EGFR gene polymorphisms on anticancer drug cytotoxicity in vitroMol Diagn Ther. 2008 12 225Google Scholar
Sasaki, SKobunai, TKitayama, JNagawa, HDNA methylation and sensitivity to antimetabolites in cancer cell linesOncol Rep. 2008 19 407Google Scholar
Liu, WHe, LRamirez, JRatain, MJInteractions between MDM2 and TP53 genetic alterations, and their impact on response to MDM2 inhibitors and other chemotherapeutic drugs in cancer cellsClin Cancer Res. 2009 15 7602Google Scholar
Klein, TEChang, JTCho MK, Easton, KLFergerson, RHewett, MLin, ZLiu, YLiu, SOliver, DERubin, DLShafa, FStuart, JMAltman, RBIntegrating genotype and phenotype information: an overview of the PharmGKB ProjectPharmacogenomics J. 2001 1 167Google Scholar

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.

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.

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.

Available formats
×