The Journal (2008) 8, 305–314 & 2008 Nature Publishing Group All rights reserved 1470-269X/08 $30.00 www.nature.com/tpj REVIEW

Somatic pharmacogenomics in cancer

ON Ikediobi Many of the initial examples of the clinical utility of pharmacogenetics were elucidated in the field of oncology. Those examples were largely based on Department of Clinical Pharmacy, School of the existence of germline genetic variation that influences the metabolism of Pharmacy, University of California, San Francisco, cytotoxic drugs. However, with the development of kinase inhibitors, drugs CA, USA designed to preferentially target altered proteins driving oncogenesis, pharmacogenetics in cancer has shifted to understanding the somatic Correspondence: Dr ON Ikediobi, Department of Clinical differences that determine response to these targeted agents. It is becoming Pharmacy, School of Pharmacy, University of increasingly clear that understanding the molecular genetics of cancer will California, 3333 California Street, Box 0613, lead to the further development of targeted therapeutics. Therefore, it is San Francisco, CA 94118, USA. imperative that pharmacogenomics researchers understand the motivations E-mail: [email protected] and challenges of developing targeted therapies to treat cancer as a paradigm for personalized medicine. However, much of the discussion in the pharmacogenomics community in cancer is still largely focused on the germline variants as predictors of drug toxicity. In light of that fact, this review presents a detailed discussion of the development of commonly used targeted therapies for the treatment of hematological and solid tumors, the somatic mutations that determine response to those therapies, and the mechanisms of drug resistance. The Pharmacogenomics Journal (2008) 8, 305–314; doi:10.1038/tpj.2008.8; published online 5 August 2008

Keywords: somatic; mutation; cancer; pharmacogenomics; kinase; inhibitor

Introduction

Pharmacogenomics is the study of the genetics of interindividual response to drugs and aims at molecular subsetting of patients for more effective therapy.1 The field of pharmacogenomics is especially important in oncology where most clinically used drugs have a narrow therapeutic window, that is the difference between the dose required to achieve the desired therapeutic effect and that causing toxicity, is small.2 Therefore, knowledge of genetic variations, inherited or acquired, that may predict differential response to cancer is key to personalized therapy. Genetic variations in drug effect are classified into two groups: those due to either pharmacokinetic or pharmacodynamic factors. The pharmacokinetic factors that influence drug effect involve the drug’s absorption, distribution, metabolism and excretion.3 In contrast, the pharmacodynamic factors that influence drug effect involve the transport of the drug into the cell and the interaction of the drug (ligand) and its target(s).3 Many of the initial examples from pharmacogenomics in cancer chemo- therapy center on understanding the inherited (germline) interindividual differences involved in (Table 1).2,4–7 However, there are also examples where acquired (somatic) mutations within the tumor DNA are Received 6 March 2008; revised 17 June 2008; accepted 3 July 2008; published online predictive of response to cancer chemotherapy. The elucidation of the signal- 5 August 2008 transduction networks that drive neoplastic transformation has led to rationally Somatic pharmacogenomics ON Ikediobi 306

Table 1 Standard chemotherapeutics and germline variants reported to modulate their action

Drug Tumor type Allelic polymorphism Drug action (references)

6-Mercaptopurine ALL TPMT*3A Enhanced toxicity 2,4 Irinotecan Colorectal UGT1A1*28 Enhanced toxicity 5 Tamoxifen Breast CYP2D6*4, *10 Decreased efficacy 6 5- Colorectal DPYD*2A Enhanced toxicity 7

Abbreviations: ALL, acute lymphoblastic leukemia; CYP2D6, cytochrome P450 2D6; DPYD, dihydropyrimidine dehydrogenase; TPMT, thiopurine methyltransferase; UGT1A1, uridine 50-diphosphate-glucuronosyl-transferase. Numbers in brackets indicate cited references.

Table 2 Kinase inhibitors and somatic mutations reported to modulate their drug action

Kinase inhibitor Tumor type product Somatic alterations

Imatinib CML, ALL, GIST BCR-ABL, KIT, PDGFRA Mutation, deletion, translocation CML BCR-ABL Mutation CML, ALL BCR-ABL Mutation Sunitinib GIST KIT, PDGFRA, FLT3, VEGFR2 Mutation Breast ERBB2 Amplification, overexpression Pertuzumab Breast ERBB2, EGFR, ERBB3 PTEN deletion, Akt overexpression Lapatinib Breast ERBB2, EGFR ERBB2 amplification Gefitinib NSCLC EGFR Mutation, amplification Erlotinib NSCLC EGFR Mutation, amplification

Abbreviations: Akt, serine/threonine protein kinase Akt; ALL, acute lymphoblastic leukemia; BCR-ABL, breakpoint cluster region-ABL; CML, chronic myeloid leukemia; ERBB2, v-erb-b2 erythroblastic leukemia viral oncogene homolog 2, neuro/glioblastoma-derived oncogene homolog (avian); ERBB3, v-erb-b2 erythroblastic leukemia viral oncogene homolog 3; FLT3, fms-related tyrosine kinase 3; GIST, gastrointestinal stromal tumor; KIT, v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homologue; NSCLC, non-small cell lung cancer; PDGFRA, platelet-derived growth factor receptor, a-polypeptide; PTEN, phosphatase and tensin homolog; VEGFR2, vascular endothelial growth factor receptor 2.

designed cancer therapeutics that target specific molecular within the tumor DNA but absent within the germline. In abnormalities.8 These targeted therapeutics, unlike tradi- this review, I have highlighted the most common examples tional cytotoxic cancer chemotherapeutics, do not have whereby somatic mutations within tumor DNA determine narrow therapeutic indices and therefore have less severe clinical response to kinase inhibitors (Table 2). side effects. The less severe side effects experienced by taking targeted therapies is as a result of their greater affinity for inhibiting the activity of somatic alterations of the target BCR-ABL, KIT, PDGFR kinase inhibitors kinase(s) that are present within the tumor DNA but absent within the germline. In addition, targeted therapeutics, The first example of the success of small-molecule targeted unlike traditional cytotoxic chemotherapy are available in therapies in is demonstrated with oral dosage forms for better ease of administration. , the first small-molecule kinase inhibitor approved Many of the currently known targeted therapeutic agents for clinical use that targets the breakpoint cluster region are protein kinase inhibitors. Kinases are key components of (BCR)-ABL protein tyrosine kinase for the treatment of cell signaling pathways involved in neoplastic transforma- chronic myeloid leukemia (CML). The BCR-ABL protein tion. that encode protein kinases are often over- results from the fusion of the BCR and nonreceptor protein expressed or activated as a result of mutations or tyrosine kinase ABL resulting from a reciprocal chromoso- chromosomal rearrangements in human tumors. Kinase mal translocation t(9;22) producing a shortened chromo- inhibitors are designed as adenosine triphosphate (ATP) some 22, called the Philadelphia (Ph) chromosome. The mimetics and therefore compete with ATP for binding at the resultant fusion protein has constitutive tyrosine kinase active site.8 When bound to the active site of the putative activity enabling the activation of several signaling path- kinase(s), kinase inhibitors reduce the activity of the ways such as Ras, PI3K-Akt and Jak-STAT leading to cell activated protein kinase(s), reducing the cellular oncogenic proliferation and survival.9 The BCR-ABL protein is asso- drive and inducing tumor regression. Interestingly, only a ciated predominantly not only with CML but also with subset of patients respond to these targeted therapies and acute lymphoblastic leukemia.9 Imatinib works by binding their response is governed by the presence of specific to and stabilizing an inactive conformation of the BCR-ABL somatic alterations of the target kinase that are present protein kinase.10,11 Because imatinib also has specificity for

The Pharmacogenomics Journal Somatic pharmacogenomics ON Ikediobi 307

the platelet-derived growth factor receptor, a-polypeptide malignancies harboring mutant forms of those proteins.24 (PDGFRA) and KIT protein kinases, it is also used in Another second generation ABL kinase inhibitor that treatment of malignancies associated with dysregulated demonstrates activity against imatinib resistance is dasati- forms of those proteins.11 nib. Dasatinib, although not specifically designed to target Although the initial response to imatinib is dramatic, imatinib-resistant forms of BCR-ABL, is a potent inhibitor of treatment failure quickly ensues. As is the case with the BCR-ABL, Src-family kinases, KIT and PDGFR.25,26 In con- treatment of CML patients with imatinib, patients initially trast to imatinib and nilotinib, dasatinib binds to the active experience complete cytogenetic and hematological remis- conformation of the ABL kinase.27 On the basis of promising sion. However because imatinib fails to deplete leukemic data from phase I and II trials of dasatinib, in patients with stem cells that harbor the BCR-ABL fusion protein,12 some imatinib-resistant CML and Ph-positive ALL patients, it was patients develop resistance to imatinib, particularly in the recently approved in the United States and Europe for the advanced phases of CML and Ph-positive acute lympho- treatment of adults in all phases of CML with imatinib blastic leukemia (ALL).13 Mechanisms of imatinib resistance resistance or intolerance.28 Dasatinib was also approved for include BCR-ABL amplification and overexpression of the treatment of patients with Ph-positive ALL with mRNA and protein.14,15 However, the most common imatinib resistance or intolerance.29 mechanism of resistance is the acquisition of somatic point Because imatinib also has specificity for KIT and PDGFRA mutations within the kinase domain of the ABL gene.16 The proteins, it is used to treat gastrointestinal tumors (GISTs) first point mutation identified as associated with imatinib that harbor mutant forms of those proteins. Most GISTs resistance was T315I.16 The crystal structure of an analogue harbor oncogenic KIT or PDGFRA receptor tyrosine kinase of imatinib bound to the ABL kinase domain revealed that the mutations.30 The KIT or PDGFRA gain-of-function muta- T315 residue was crucial for the interaction between imatinib tions are early events in GIST oncogenesis and lead to the and ABL.17 It was also found that a different mutation at the activation of the mitogen-activated protein kinase (MAPK) T315 residue (T315V) conferred constitutive kinase activity to and signal transducer and activator of transcription (STAT) ABL and was less sensitive to imatinib compared with wild- downstream signaling pathways involved in cell proliferation type ABL.18 The T315I point mutation impairs imatinib and survival, respectively.31 Imatinib, a potent inhibitor of binding thereby reducing its inhibition of ABL. KIT signaling, has recently become first-line treatment of There are more than 50 different point mutations in ABL metastatic GIST following in vitro studies suggesting a that are associated with imatinib resistance.13 However, therapeutic potential for imatinib in a human GIST cell line.32 most are rare and six amino-acid residues (Gly250, Tyr253, Before treatment with imatinib, surgical resection of primary Glu255, Thr315, Met351 and Phe359) thus far account for localized GIST was the only chance for cure.33 GISTs proved 60–70% of imatinib-resistant mutations.13 Two of the more refractory to standard chemotherapy and radiation. Recur- frequently detected ABL mutants, Y253F and E255K, have rence of disease was certain with a predicted 5-year survival of been shown to have high in vitro transforming potential.13 30%.34 Prospective trials of imatinib in metastatic GISTs have This in vitro finding is consistent with clinical findings that shown that approximately 80% of patients will respond to show P-loop mutations such as Y253F and E255K are imatinib and will achieve stable disease.35,36 In addition, 70% associated with a greater likelihood of progression to blast of metastatic GIST patients will have at least a 2-year disease- crisis and shorter overall survival in imatinib-treated free survival and 50% will be free of disease progression.37 patients.19 Another ABL mutant, T315I, is generally found As is the case with CML and imatinib response, specific in patients with advanced CML, and predicts a worse overall mutations of KIT and PDGFRA in GISTs predict response to survival compared with other ABL mutations in patients on imatinib. KIT mutations occur in up to 90% of GISTs and imatinib therapy.20 clinical response to imatinib is dependent, at least in part, Those drug-resistant BCR-ABL point mutations can be on the presence of specific KIT mutations.38 KIT exon 11 found in imatinib-naı¨ve CML or can be acquired during mutations are found in 75% of GISTs and result in the imatinib treatment. Acquired imatinib resistance involves abrogation of the juxtamembrane autoinhibition of the KIT the reemergence of BCR-ABL tyrosine kinase activity.21 This kinase.39 Patients with exon 11 KIT mutations have a higher suggests that the mutant BCR-ABL protein is still a viable response rate to imatinib treatment and longer time to target for inhibition by small-molecule inhibitors.21,22 To treatment failure compared with other KIT or PDGFRA this end, second generation ABL kinase inhibitors have been mutations.40 Patients without detectable KIT or PDGFRA designed and have been shown to inhibit the growth of mutation respond less frequently to imatinib treatment most mutant forms of BCR-ABL resistant to imatinib. One of compared with exon 11 KIT mutants.38 However, up to 39% those drugs, nilotinib, was specifically designed to target of those patients without KIT or PDGFRA mutations do imatinib-resistant forms of BCR-ABL, and is approximately respond to imatinib.41 As well, patients with the rare exon 30-fold more potent than imatinib as an ABL inhibitor.13 13 KIT mutation or exon 17 PDGFRA mutation may respond Phase I and II trials of nilotinib in patients with imatinib- to imatinib.40 These data suggest that imatinib treatment resistant CML in all phases of disease and patients with Ph- ought to be considered for all GIST patients, regardless of positive ALL demonstrate significant clinical response to KIT or PDGFRA mutation status.40 The only exception may nilotinib.23 Nilotinib also inhibits KIT and PDGFRB protein be patients with primary imatinib-resistant mutation of kinases and may therefore be used for the treatment of PDGFRA (D842V).40

The Pharmacogenomics Journal Somatic pharmacogenomics ON Ikediobi 308

The majority of metastatic GIST patients will develop acquired) have not been fully elucidated, however there are resistance to imatinib. The most common resistance numerous proposed mechanisms. Elucidating the molecular mechanisms involve the acquisition of secondary exon 13, mechanisms underlying primary or acquired trastuzumab 14 or 17 KIT mutations that prohibit imatinib binding.40 resistance is critical to improving the survival of metastatic Some of these secondary mutations are intrinsically breast cancer patients whose tumors overexpress ERBB2.55 imatinib-resistant, such as the frequently occurring V654A Altered downstream signaling from ERBB2 has been substitution.42 However, other mutations such as those shown to confer primary resistance to trastuzumab. ERBB2 involving the N822 residue are intrinsically imatinib signaling activates the PI3K signaling pathway. Constitutive sensitive but are associated with clinical imatinib resistance PI3K/AKT activity has been shown to inhibit trastuzumab- when coincident with an exon 11 KIT mutation.42 mediated cell-cycle arrest and apoptosis.56 An ERBB2 over- It is therefore crucial to develop more potent inhibitors of expressing breast cancer cell line, BT474, resistant to KIT and PDGFRA to potentially overcome imatinib resis- trastuzumab had elevated levels of phosphorylated AKT tance. Sunitinib, an inhibitor of KIT, PDGFRA, fms-related compared to the parent line.57 The resistant cells were tyrosine kinase 3 (FLT3) and vascular endothelial growth sensitive to a small-molecule inhibitor of PI3K.57 Patients factor receptor 2 (VEGFR2), has recently been approved for with phosphatase and tensin homolog (PTEN)-deficient, the treatment of imatinib-resistant GIST and patients unable ERBB2 overexpressing breast tumors have a poorer response to tolerate treatment with imatinib.43 In a randomized to trastuzumab-based therapy.58 Subsequently, it was shown phase III trial of sunitinib in patients who had progressed on that in phosphatase and tensin homolog (PTEN)-deficient imatinib therapy, sunitinib was found to prolong median cells, PI3K inhibitors rescued trastuzumab resistance in vitro time to tumor progression compared with placebo.44 and in vivo.58 Therefore, PTEN loss may serve as a predictor of However, sunitinib provides only a temporary benefit for trastuzumab resistance and PI3K inhibitors may be potential imatinib-resistant GIST patients, therefore more therapeutic therapies in PTEN-null trastuzumab-resistant tumors.49 options are needed. Preclinical studies in GIST cell lines have Novel therapeutic strategies are being employed to over- shown that treatment with heat-shock protein 90 inhibitors come resistance to trastuzumab. Pertuzumab is a mono- resulted in degradation of the KIT oncoprotein and may clonal ERBB2 antibody that represents a new class of drugs therefore be of benefit in imatinib-resistant GIST.45 called dimerization inhibitors.49 Pertuzumab can block signaling by other EGFR family receptors, as well as inhibit signaling in cells expressing normal ERBB2 levels. Pertuzu- ERBB2 kinase inhibitors mab sterically blocks dimerization of ERBB2 with EGFR and ERBB3, inhibiting signaling from ERBB2/EGFR and ERBB2/ Another example of the success of targeted therapies in the ERBB3 heterodimers.59 Pertuzumab is also able to disrupt treatment of cancer is that of trastuzumab, a monoclonal the interaction between ERBB2 and insulin-like growth antibody against v-erb-b2 erythroblastic leukemia viral factor-I receptor in trastuzumab-resistant cells.60 Trastuzu- oncogene homolog 2 (ERBB2) that was developed to treat mab and pertuzumab bind to different epitopes in the breast cancer patients whose tumors overexpress the extracellular domain of ERBB2.61,62 The combination of protein. ERBB2 protein tyrosine kinase amplification and trastuzumab and pertuzumab produced synergistic apopto- overexpression occurs in approximately 30% of metastatic sis in ERBB2 overexpressing trastuzumab-naı¨ve breast cancer breast cancers and leads to aberrant signaling through the cells,63 without any significant effect on the viability of PI3K and MAPK pathways, resulting in cell proliferation and trastuzumab-resistant breast cancer cells.60 evasion of apoptosis.46 Amplified ERBB2 in breast cancer Another alternative therapeutic agent against trastuzu- shows clinical correlates with earlier relapse and shorter mab-resistant tumors is lapatinib, a dual tyrosine kinase overall survival.47 Therefore, a drug, trastuzumab that could inhibitor that targets both EGFR and ERBB2. Binding of target ERBB2 protein in those cancers could increase lapatinib to EGFR and ERBB2 is reversible but its dissociation survival. Trastuzumab was the first genetic-based drug is much slower allowing for prolonged downregulation of approved for cancer therapy.48 Unlike the small-molecule receptor tyrosine phosphorylation.49 ERBB2 status and not kinase inhibitors, which are administered orally, trastuzu- EGFR status is a determinant of lapatinib activity.49 It has mab, a monoclonal antibody, is administered intravenously. been shown that the combination of lapatinib with Trastuzumab is active as a single agent after failure of other trastuzumab enhanced apoptosis of ERBB2 overexpressing chemotherapy regimens.49 However, primary resistance to breast cancer cells.64 Resistance to lapatinib seems to be trastuzumab monotherapy occurs in approximately 66–88% mediated by increased signaling from the estrogen receptor of ERBB2 overexpressing metastatic breast tumors.50,51 in estrogen receptor-positive ERBB2 overexpressing breast Trastuzumab with adjuvant chemotherapy (paclitaxel or cancers.65 This suggests that targeting of both the estrogen docetaxel) significantly improves disease-free and overall receptor and ERBB2 may be beneficial in that subset of survival in patients with early stage ERBB2 overexpressing cancer patients.65 A recent phase III trial of trastuzumab- breast cancers compared with trastuzumab monotherapy.52–54 resistant ERBB2 overexpressing breast cancer patients Invariably, the majority of patients who achieve an initial demonstrated that the combination of lapatinib and response to trastuzumab-based regimens develop resistance capecitabine resulted in longer median progression-free within 1 year.49 The mechanisms of resistance (primary or survival compared with capecitabine alone.66

The Pharmacogenomics Journal Somatic pharmacogenomics ON Ikediobi 309

ERBB2, a member of the EGFR family, is also over- cancers.88–93 Mutations outside of the kinase domain expressed, to a lesser degree, in lung cancers, specifically (extracellular domain) are rare in NSCLC,81–83 but are adenocarcinomas and large-cell carcinomas and is predictive common in gliomas94 and squamous cell lung cancers.95 of poorer outcomes.67–69 In addition, intragenic mutations Therefore, the use of EGFR inhibitors in subsets of those have also been found in the conserved kinase domain of the tumors may be a viable therapeutic option. ERBB2 gene in some lung cancers.70,71 The ERBB2 mutations In NSCLC, somatic mutations of EGFR are sometimes seem to occur exclusively in non-small cell lung cancer associated with amplification.96 Studies have shown that (NSCLC) of adenocarcinoma histology and are more patients with EGFR amplification were more likely to common in female patients and never smokers,71 similar respond to gefitinib or erlotinib and had longer median to the clinicophathologic manifestation of EGFR mutations. time to disease progression and overall survival compared to It was then thought that trastuzumab might be of clinical patients with normal EGFR copy number.97 However, it benefit for the treatment of ERBB2 mutant NSCLC. How- remains to be established whether amplification of EGFR ever, thus far, there has not been demonstrated the benefit (wildtype or mutant) contributes to lung cancer develop- of trastuzumab monotherapy or in combination with cancer ment and response to gefitinib or erlotinib.97 chemotherapeutics for the treatment of NSCLC with over- Thus far there are five mutations known to confer expressed ERBB2.72–75 sensitivity to EGFR tyrosine kinase inhibitors. The drug- sensitive mutations are: point mutations in exon 18 (G719A or G719C), point mutations of exon 21 (L858R and L861Q) EGFR kinase inhibitors and in-frame deletions of exon 19 that eliminates four amino acids (LREA).98 The most common of these drug- To further illustrate the importance of the EGFR family of sensitive mutations are the exon 19 in-frame deletion and proteins in cancer progression and treatment, I discuss the exon 21 missense amino-acid substitution (L858R) account- timely development of EGFR inhibitors and its subsequent ing for up to 90% of EGFR mutations in NSCLC.98 In clinical use. EGFR protein tyrosine kinase overexpression retrospective studies, the association between the presence has been implicated in numerous cancer types, is associated of EGFR mutation and sensitivity to gefitinib and erlotinib is with poor prognosis, and leads to enhanced signaling quite consistent showing 75% response rate for patients through antiapoptotic proteins, Akt and STAT.76–78 The fact with EGFR mutations compared to 10% response rate for that EGFR is overexpressed in many cancer types and patients with wild-type EGFR.99–103 Prospective trials have associated with poor prognosis made it a good candidate confirmed the association between EGFR mutation and for the development of targeted therapeutics. Therefore, two sensitivity to gefitinib and erlotinib. Those studies showed small-molecule inhibitors of EGFR, gefitinib and erlotinib, that 78% of patients with somatic exon 19 deletion or exon received speedy approval from the US Food and Drug 21 L858R mutation had radiographic responses to gefitinib Administration (FDA) in May 2003 and November 2004, and erlotinib.104–106 respectively. Gefitinib and erlotinib were approved as single- However, there is controversy around what prolongs drug therapy for the treatment of patients with advanced survival of NSCLC patients; treatment with EGFR inhibitors chemotherapy-refractory NSCLC.79,80 It was later observed, or presence of EGFR mutation? Large phase III retrospective in the clinic, that only a subset of NSCLC patients achieved trials have been conducted where NSCLC patients were dramatic response to these drugs. By molecular genetic randomized to receive either standard cytotoxic chemother- analyses, it was found that specific somatic mutations in the apy alone or standard chemotherapy in combination with conserved kinase domain of the EGFR gene are associated gefitinib or erlotinib.99,107 Those studies have reported that with sensitivity to gefitinib81,82 and erlotinib.83 Recently, patients with EGFR mutations have prolonged survival however, a large clinical trial comparing gefitinib with compared with patients with wild-type EGFR treated with placebo in NSCLC patients who had failed other courses of gefitinib or erlotinib. Interestingly, the studies have also chemotherapy showed no survival benefit from taking found that the prolonged survival may occur in the absence gefitinib.84 Following this report in 2005 the FDA restricted of treatment with gefitinib, erlotinib, surgery or standard use of gefitinib only to patients who had previously cancer chemotherapy.97 In the standard cytotoxic che- responded to the drug.85 In contrast, clinical trials have motherapy alone treatment arm, patients with EGFR muta- demonstrated that erlotinib improves survival in NSCLC tion had prolonged progression-free and overall survival patients whose cancers have progressed on other courses of compared with patients with wild-type EGFR.99,107 chemotherapy.86 Therefore, erlotinib is FDA approved for Even within the group of NSCLC patients with EGFR use in new NSCLC patients with chemotherapy-refractive mutations, there are reported differences in the clinical tumors. course between patients with exon 19 in-frame deletions Somatic mutations of EGFR kinase domain are most and patients with exon 21 L858R missense substitution. One common in NSCLC and have a distinct clinicophathologic study has shown that NSCLC patients with the L858R manifestation common in adenocarcinomas, East Asians, mutation treated with surgery alone have a prolonged women and never smokers.87 Rare mutations of EGFR are overall survival compared with NSCLC patients with exon found in head and neck cancers, cholangiosarcomas, colon 19 deletions.87 Another study has shown that after treat- cancers, ovarian cancers, esophageal cancers and pancreatic ment with gefitinib or erlotinib, NSCLC patients with EGFR

The Pharmacogenomics Journal Somatic pharmacogenomics ON Ikediobi 310

exon 19 deletions have a longer overall survival compared development of molecular diagnostics incorporating any with patients harboring the exon 21 L858R mutation.98 The one or combination of those biomarkers may be essential to molecular basis for such findings is yet unknown. guide the appropriate use of EGFR inhibitors. Therefore, it is no surprise that the mechanism of Adding to the complexity of predicting response to EFGR increased sensitivity of EGFR mutants to gefitinib and inhibitors, a novel drug-resistant gefitinib mutation has erlotinib is still unknown. However, it has been shown that recently been reported that does not involve mutation of the exon 19 in-frame deletions and exon 21 L858R missense EGFR. It was reported that gefitinib-resistant clones from an amino-acid substitution confer ligand-independent activa- EGFR-mutant lung cancer cell line displayed amplification tion and prolonged kinase activity after ligand stimula- of MET oncogene and maintained activation of ERBB3/PI3K/ tion,81,82 suggesting that inhibition of the aberrant EGFR AKT signaling in the presence of gefitinib.121 Following the signaling may be the predominant mechanism of action of initial observation in cell lines, a panel of 18 gefitinib or EGFR inhibitors. erlotinib-resistant primary lung tumors were assessed for As is the case with the aforementioned targeted therapies, MET amplification. MET amplification was found to occur in imatinib and trastuzumab, despite a dramatic initial re- 22% of those tumors implicating MET amplification in the sponse to gefitinib and erlotinib, NSCLC patients with EGFR development of resistance to gefitinib in a subset of lung mutations rarely achieve a complete response and resistance cancers.121 Therefore, developing targeted therapies against to treatment develops. There are thus far, three EGFR kinase amplified MET may be beneficial in the treatment of NSCLC domain mutations associated with drug resistance: an exon patients whose tumors bear the MET amplification and have 19 point mutation (D761Y), an exon 20 point mutation progressed on EGFR inhibitors. (T790M) and an exon 20 insertion (D770_N771insNPG).97 The most common of these drug-resistant mutations is the T790M reported to occur in about 50% of tumors after Discussion disease progression and has been predicted to block the binding of gefitinib or erlotinib to the kinase ATP binding It is clear from the above-mentioned examples, that somatic pocket.108,109 This mutation is analogous to the acquired mutations in cancer can be important in guiding the drug-resistant mutation, T315I, to imatinib seen in GIST and appropriate use of targeted therapies. As we continue to CML.97 Interestingly, the T790M mutation has been seen in understand the dysregulated pathways driving oncogenesis the germline and tumor DNA of family members with this will inevitably lead to the further development of hereditary bronchioloalveolar carcinoma.110 One of the targeted therapies. Elucidation of the myriad genomic family members with the T790M mutation did not respond abnormalities in cancer will be key to enhancing patient to treatment with gefitinib.110 In vitro data have suggested stratification for more accurate diagnosis and treatment. To that irreversible EGFR inhibitors may have activity in this end, a few groups are employing systematic high- patients with acquired resistance to gefitinib or erloti- throughput sequencing screens of cancer in search nib.111,112 To this end, a phase II trial of HKI-272, an of other mutated kinases driving oncogenesis. This ap- irreversible EGFR kinase inhibitor, is ongoing to determine proach has been fruitful in identifying more frequently the efficacy in patients who have progressed after initial mutated oncogenes; BRAF in melanoma,122 ERBB2 in lung treatment with gefitinib or erlotinib.97 cancer123 and PIK3CA in colorectal cancer.124 This has In addition to mutations of EGFR other molecular generated interest and investigation into the development parameters involved in the EGFR signaling cascade are and clinical use of small-molecule inhibitors of those kinases associated with activity of gefitinib or erlotinib. For (BRAF and PIK3CA) or their signaling partners for the example, it has been shown that increased expression of treatment of cancers.125–128 transforming growth factor-a and heregulin, ligands for Despite the initial success of targeted therapies in produ- EGFR and ERBB3, respectively, is associated with poor cing dramatic responses and tumor regression in genetically response to gefitinib.113,114 Perhaps competitive inhibitors defined subgroups of patients, drug resistance inevitably of those ligands may be a useful therapeutic intervention. It develops. The next challenge facing drug development has also been reported that increased copy number of experts is the design of targeted therapies against those another member of the EGFR family of proteins, ERBB2, in drug-resistant tumors. One way to overcome or abate the presence of EGFR mutation in NSCLC, is associated with resistance to single-targeted therapy may be to administer response to gefitinib.115 Paradoxically, however, NSCLC multiple kinase inhibitors or multikinase inhibitors to patients with intragenic ERBB2 mutations do not respond patients whose tumors may have more than a single to gefitinib or erlotinib.116 That finding suggests there is a mutated kinase. This goal may be achieved the more we distinct genetic context of EGFR mutant NSCLC that learn about the unique somatic mutation patterns of predicates response to EGFR inhibitors. Whereas down- individual tumors and have a plethora of targeted therapies stream of EGFR signaling, it has been observed that NSCLC with which to tailor appropriate treatment regimens. patients with KRAS mutations are resistant to gefitinib or The symbiosis between molecular genetic-based disease erlotinib,117 and increased phosphorylation of AKT is classification and genetic-based therapeutic intervention predictive of response to gefitinib or erlotinib.118–120 In serves as a paradigm for the future of personalized medicine light of those observations, one can envisage that the in cancer. Ultimately, however, drug response, even with

The Pharmacogenomics Journal Somatic pharmacogenomics ON Ikediobi 311

targeted therapies is not solely dependent on the tumor 20 Nicolini FE, Corm S, Le QH, Sorel N, Hayette S, Bories D et al. Mutation genetics. The germline genetics also plays a role, especially status and clinical outcome of 89 imatinib mesylate-resistant chronic in drug metabolism. Therefore, as we move forward with myelogenous leukemia patients: a retrospective analysis from the French intergroup of CML (Fi(phi)-LMC GROUP). Leukemia 2006; 20: implementing genetic-based tests for drug therapy in 1061–1066. oncology, it will be important to understand and consider 21 Barthe C, Gharbi MJ, Lagarde V, Chollet C, Cony-Makhoul P, Reiffers J the contribution of both somatic and germline genetics in et al. Mutation in the ATP-binding site of BCR-ABL in a patient with predicting drug response and toxicity. chronic myeloid leukaemia with increasing resistance to STI571. Br J Haematol 2002; 119: 109–111. 22 Branford S, Rudzki Z, Walsh S, Grigg A, Arthur C, Taylor K et al. High Duality of Interest frequency of point mutations clustered within the adenosine tripho- sphate-binding region of BCR/ABL in patients with chronic myeloid The author has no conflicts or duality of interests to disclose. leukemia or Ph-positive acute lymphoblastic leukemia who develop imatinib (STI571) resistance. Blood 2002; 99: 3472–3475. 23 Kantarjian H, Giles F, Wunderle L, Bhalla K, O’Brien S, Wassmann B References et al. Nilotinib in imatinib-resistant CML and Philadelphia chromo- some-positive ALL. N Engl J Med 2006; 354: 2542–2551. 1 Weinstein JN, Pommier Y. Transcriptomic analysis of the NCI-60 cancer 24 Manley PW, Cowan-Jacob SW, Mestan J. Advances in the structural cell lines. C R Biol 2003; 326: 909–920. biology, design and clinical development of Bcr-Abl kinase inhibitors 2 Weinshilboum R, Wang L. Pharmacogenomics: bench to bedside. Nat for the treatment of chronic myeloid leukaemia. Biochim Biophys Acta Rev Drug Discov 2004; 3: 739–748. 2005; 1754:3–13. 3 Lindpaintner K. The impact of pharmacogenetics and pharmacoge- 25 Das J, Chen P, Norris D, Padmanabha R, Lin J, Moquin RV et al. nomics on drug discovery. Nat Rev Drug Discov 2002; 1: 463–469. 2-aminothiazole as a novel kinase inhibitor template. Structure-activity 4 Goetz MP, Ames MM, Weinshilboum RM. Primer on medical relationship studies toward the discovery of N-(2-chloro-6-methylphenyl)- genomics. Part XII: pharmacogenomics—general principles with 2-[[6-[4-(2-hydroxyethyl)-1-piperazinyl)]-2-methyl-4-pyrimidinyl]amino)]- cancer as a model. Mayo Clin Proc 2004; 79: 376–384. 1,3-thiazole-5-carboxamide (dasatinib, BMS-354825) as a potent 5 Kim TW, Innocenti F. Insights, challenges, and future directions in pan-Src kinase inhibitor. J Med Chem 2006; 49: 6819–6832. irinogenetics. Ther Drug Monit 2007; 29: 265–270. 26 Melnick JS, Janes J, Kim S, Chang JY, Sipes DG, Gunderson D et al. An 6 Goetz MP, Kamal A, Ames MM. Tamoxifen pharmacogenomics: the efficient rapid system for profiling the cellular activities of molecular role of CYP2D6 as a predictor of drug response. Clin Pharmacol Ther libraries. Proc Natl Acad Sci USA 2006; 103: 3153–3158. 2008; 83: 160–166. 27 Tokarski JS, Newitt JA, Chang CY, Cheng JD, Wittekind M, Kiefer SE 7 Yen JL, McLeod HL. Should DPD analysis be required prior to et al. The structure of Dasatinib (BMS-354825) bound to activated ABL prescribing fluoropyrimidines? Eur J Cancer 2007; 43: 1011–1016. kinase domain elucidates its inhibitory activity against imatinib- 8 Sebolt-Leopold JS, English JM. Mechanisms of drug inhibition of resistant ABL mutants. Cancer Res 2006; 66: 5790–5797. signalling molecules. Nature 2006; 441: 457–462. 28 Cortes J, Rousselot P, Kim DW, Ritchie E, Hamerschlak N, Coutre S 9 Manley PW, Cowan-Jacob SW, Buchdunger E, Fabbro D, Fendrich G, et al. Dasatinib induces complete hematologic and cytogenetic Furet P et al. Imatinib: a selective tyrosine kinase inhibitor. Eur J Cancer responses in patients with imatinib-resistant or -intolerant chronic 2002; 38: S19–S27. myeloid leukemia in blast crisis. Blood 2007; 109: 3207–3213. 10 Roskoski Jr R. STI-571: an anticancer protein-tyrosine kinase inhibitor. 29 Talpaz M, Shah NP, Kantarjian H, Donato N, Nicoll J, Paquette R et al. Biochem Biophys Res Commun 2003; 309: 709–717. Dasatinib in imatinib-resistant Philadelphia chromosome-positive 11 Capdeville R, Buchdunger E, Zimmermann J, Matter A. Glivec (STI571, leukemias. N Engl J Med 2006; 354: 2531–2541. imatinib), a rationally developed, targeted anticancer drug. Nat Rev 30 Fletcher JA, Rubin BP. KIT mutations in GIST. Curr Opin Genet Dev Drug Discov 2002; 1: 493–502. 2007; 17: 3–7. 12 Michor F, Hughes TP, Iwasa Y, Branford S, Shah NP, Sawyers CL et al. 31 Corless CL, McGreevey L, Haley A, Town A, Heinrich MC. KIT Dynamics of chronic myeloid leukaemia. Nature 2005; 435: mutations are common in incidental gastrointestinal stromal tumors 1267–1270. one centimeter or less in size. Am J Pathol 2002; 160: 1567–1572. 13 Weisberg E, Manley PW, Cowan-Jacob SW, Hochhaus A, Griffin JD. 32 Tuveson DA, Willis NA, Jacks T, Griffin JD, Singer S, Fletcher CD et al. Second generation inhibitors of BCR-ABL for the treatment of imatinib- STI571 inactivation of the gastrointestinal stromal tumor c-KIT resistant chronic myeloid leukaemia. Nat Rev Cancer 2007; 7: 345–356. oncoprotein: biological and clinical implications. Oncogene 2001; 20: 14 le Coutre P, Tassi E, Varella-Garcia M, Barni R, Mologni L, Cabrita G 5054–5058. et al. Induction of resistance to the Abelson inhibitor STI571 in 33 Gold JS, Dematteo RP. Combined surgical and molecular therapy: the human leukemic cells through gene amplification. Blood 2000; 95: gastrointestinal stromal tumor model. Ann Surg 2006; 244: 176–184. 1758–1766. 34 DeMatteo RP, Lewis JJ, Leung D, Mudan SS, Woodruff JM, Brennan 15 Weisberg E, Griffin JD. Mechanism of resistance to the ABL tyrosine MF. Two hundred gastrointestinal stromal tumors: recurrence patterns kinase inhibitor STI571 in BCR/ABL-transformed hematopoietic cell and prognostic factors for survival. Ann Surg 2000; 231: 51–58. lines. Blood 2000; 95: 3498–3505. 35 Demetri GD. Identification and treatment of chemoresistant inoper- 16 Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, Rao PN et al. able or metastatic GIST: experience with the selective tyrosine kinase Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene inhibitor imatinib mesylate (STI571). Eur J Cancer 2002; 38: S52–S59. mutation or amplification. Science 2001; 293: 876–880. 36 Verweij J, van Oosterom A, Blay JY, Judson I, Rodenhuis S, van der 17 Schindler T, Bornmann W, Pellicena P, Miller WT, Clarkson B, Kuriyan J. Graaf W et al. Imatinib mesylate (STI-571 Glivec, Gleevec) is an active Structural mechanism for STI-571 inhibition of abelson tyrosine kinase. agent for gastrointestinal stromal tumours, but does not yield Science 2000; 289: 1938–1942. responses in other soft-tissue sarcomas that are unselected for a 18 Corbin AS, Buchdunger E, Pascal F, Druker BJ. Analysis of the structural molecular target. Results from an EORTC Soft Tissue and Bone basis of specificity of inhibition of the Abl kinase by STI571. J Biol Chem Sarcoma Group phase II study. Eur J Cancer 2003; 39: 2006–2011. 2002; 277: 32214–32219. 37 Verweij J, Casali PG, Zalcberg J, LeCesne A, Reichardt P, Blay JY et al. 19 Soverini S, Martinelli G, Rosti G, Bassi S, Amabile M, Poerio A et al. ABL Progression-free survival in gastrointestinal stromal tumours with high- mutations in late chronic phase chronic myeloid leukemia patients dose imatinib: randomised trial. Lancet 2004; 364: 1127–1134. with up-front cytogenetic resistance to imatinib are associated with a 38 Heinrich MC, Corless CL, Demetri GD, Blanke CD, von Mehren M, greater likelihood of progression to blast crisis and shorter survival: a Joensuu H et al. Kinase mutations and imatinib response in patients study by the GIMEMA Working Party on Chronic Myeloid Leukemia. with metastatic gastrointestinal stromal tumor. J Clin Oncol 2003; 21: J Clin Oncol 2005; 23: 4100–4109. 4342–4349.

The Pharmacogenomics Journal Somatic pharmacogenomics ON Ikediobi 312

39 Tarn C, Merkel E, Canutescu AA, Shen W, Skorobogatko Y, Heslin MJ 61 Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, Denney Jr DW et al. Analysis of KIT mutations in sporadic and familial gastrointestinal et al. Structure of the extracellular region of HER2 alone and in stromal tumors: therapeutic implications through protein modeling. complex with the Herceptin Fab. Nature 2003; 421: 756–760. Clin Cancer Res 2005; 11: 3668–3677. 62 Franklin MC, Carey KD, Vajdos FF, Leahy DJ, de Vos AM, Sliwkowski 40 Joensuu H. Gastrointestinal stromal tumor (GIST). Ann Oncol 2006a; MX. Insights into ErbB signaling from the structure of the ErbB2- 17: 280–286. pertuzumab complex. Cancer Cell 2004; 5: 317–328. 41 Heinrich MC, Corless CL. Gastric GI stromal tumors (GISTs): the 63 Nahta R, Hung MC, Esteva FJ. The HER-2-targeting antibodies role of surgery in the era of targeted therapy. J Surg Oncol 2005; 90: trastuzumab and pertuzumab synergistically inhibit the survival of 195–207. breast cancer cells. Cancer Res 2004; 64: 2343–2346. 42 Heinrich MC, Corless CL, Blanke CD, Demetri GD, Joensuu H, Roberts 64 Xia W, Gerard CM, Liu L, Baudson NM, Ory TL, Spector NL. PJ et al. Molecular correlates of imatinib resistance in gastrointestinal Combining lapatinib (GW572016), a small molecule inhibitor of stromal tumors. J Clin Oncol 2006; 24: 4764–4774. ErbB1 and ErbB2 tyrosine kinases, with therapeutic anti-ErbB2 43 Joensuu H. Sunitinib for imatinib-resistant GIST. Lancet 2006b; 368: antibodies enhances apoptosis of ErbB2-overexpressing breast cancer 1303–1304. cells. Oncogene 2005; 24: 6213–6221. 44 Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, 65 Xia W, Bacus S, Hegde P, Husain I, Strum J, Liu L et al. A model of Verweij J et al. Efficacy and safety of sunitinib in patients with advanced acquired autoresistance to a potent ErbB2 tyrosine kinase inhibitor and gastrointestinal stromal tumour after failure of imatinib: a randomised a therapeutic strategy to prevent its onset in breast cancer. Proc Natl controlled trial. Lancet 2006; 368: 1329–1338. Acad Sci USA 2006; 103: 7795–7800. 45 Bauer S, Yu LK, Demetri GD, Fletcher JA. Heat shock protein 90 66 Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T inhibition in imatinib-resistant gastrointestinal stromal tumor. Cancer et al. Lapatinib plus capecitabine for HER2-positive advanced breast Res 2006; 66: 9153–9161. cancer. N Engl J Med 2006; 355: 2733–2743. 46 Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE et al. 67 Azzoli CG, Krug LM, Miller VA, Kris MG, Mass R. Trastuzumab in the Studies of the HER-2/neu proto-oncogene in human breast and treatment of non-small cell lung cancer. Semin Oncol 2002; 29: 59–65. ovarian cancer. Science 1989; 244: 707–712. 68 Shi D, He G, Cao S, Pan W, Zhang HZ, Yu D et al. Overexpression of 47 Perren TJ. c-erbB-2 oncogene as a prognostic marker in breast cancer. the c-erbB-2/neu-encoded p185 protein in primary lung cancer. Mol Br J Cancer 1991; 63: 328–332. Carcinog 1992; 5: 213–218. 48 Roskoski Jr R. The ErbB/HER receptor protein-tyrosine kinases and 69 Brabender J, Danenberg KD, Metzger R, Schneider PM, Park J, Salonga cancer. Biochem Biophys Res Commun 2004; 319: 1–11. D et al. Epidermal growth factor receptor and HER2-neu mRNA 49 Nahta R, Esteva FJ. Herceptin: mechanisms of action and resistance. expression in non-small cell lung cancer Is correlated with survival. Clin Cancer Lett 2006; 232: 123–138. Cancer Res 2001; 7: 1850–1855. 50 Baselga J, Tripathy D, Mendelsohn J, Baughman S, Benz CC, Dantis L 70 Stephens P, Hunter C, Bignell G, Edkins S, Davies H, Teague J et al. et al. Phase II study of weekly intravenous trastuzumab (Herceptin) in Lung cancer: intragenic ERBB2 kinase mutations in tumours. Nature patients with HER2/neu-overexpressing metastatic breast cancer. 2004; 431: 525–526. Semin Oncol 1999; 26: 78–83. 71 Shigematsu H, Takahashi T, Nomura M, Majmudar K, Suzuki M, Lee H 51 Vogel CL, Cobleigh MA, Tripathy D, Gutheil JC, Harris LN, et al. Somatic mutations of the HER2 kinase domain in lung Fehrenbacher L et al. Efficacy and safety of trastuzumab as a single adenocarcinomas. Cancer Res 2005b; 65: 1642–1646. agent in first-line treatment of HER2-overexpressing metastatic breast 72 Gatzemeier U, Groth G, Butts C, Van Zandwijk N, Shepherd F, cancer. J Clin Oncol 2002; 20: 719–726. Ardizzoni A et al. Randomized phase II trial of gemcitabine-cisplatin 52 Seidman AD, Fornier MN, Esteva FJ, Tan L, Kaptain S, Bach A et al. with or without trastuzumab in HER2-positive non-small-cell lung Weekly trastuzumab and paclitaxel therapy for metastatic breast cancer. Ann Oncol 2004; 15: 19–27. cancer with analysis of efficacy by HER2 immunophenotype and gene 73 Langer CJ, Stephenson P, Thor A, Vangel M, Johnson DH. Trastuzumab amplification. J Clin Oncol 2001; 19: 2587–2595. in the treatment of advanced non-small-cell lung cancer: is there a 53 Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A et al. Use of chemotherapy plus a monoclonal antibody against HER2 role? Focus on Eastern Cooperative Oncology Group study 2598. J Clin for metastatic breast cancer that overexpresses HER2. N Engl J Med Oncol 2004; 22: 1180–1187. 2001; 344: 783–792. 74 Zinner RG, Glisson BS, Fossella FV, Pisters KM, Kies MS, Lee PM et al. 54 Esteva FJ, Valero V, Booser D, Guerra LT, Murray JL, Pusztai L et al. Trastuzumab in combination with cisplatin and gemcitabine in Phase II study of weekly docetaxel and trastuzumab for patients with patients with Her2-overexpressing, untreated, advanced non-small HER-2-overexpressing metastatic breast cancer. J Clin Oncol 2002; 20: cell lung cancer: report of a phase II trial and findings regarding 1800–1808. optimal identification of patients with Her2-overexpressing disease. 55 Nahta R, Yu D, Hung MC, Hortobagyi GN, Esteva FJ. Mechanisms of Lung Cancer 2004; 44: 99–110. disease: understanding resistance to HER2-targeted therapy in human 75 Clamon G, Herndon J, Kern J, Govindan R, Garst J, Watson D et al. Lack breast cancer. Nat Clin Pract Oncol 2006; 3: 269–280. of trastuzumab activity in nonsmall cell lung carcinoma with over- 56 Nahta R, Yuan LX, Zhang B, Kobayashi R, Esteva FJ. Insulin-like growth expression of erb-B2: 39810: a phase II trial of Cancer and Leukemia factor-I receptor/human epidermal growth factor receptor 2 hetero- Group B. Cancer 2005; 103: 1670–1675. dimerization contributes to trastuzumab resistance of breast cancer 76 Nicholson RI, Gee JM, Harper ME. EGFR and cancer prognosis. Eur J cells. Cancer Res 2005; 65: 11118–11128. Cancer 2001; 37(Suppl 4): S9–15. 57 Chan CT, Metz MZ, Kane SE. Differential sensitivities of trastuzumab 77 Sridhar SS, Seymour L, Shepherd FA. Inhibitors of epidermal-growth- (Herceptin)-resistant human breast cancer cells to phosphoinositide-3 factor receptors: a review of clinical research with a focus on non- kinase (PI-3K) and epidermal growth factor receptor (EGFR) kinase small-cell lung cancer. Lancet Oncol 2003; 4: 397–406. inhibitors. Breast Cancer Res Treat 2005; 91: 187–201. 78 Sridhar SS, Seymour L, Shepherd FA. Inhibitors of epidermal-growth- 58 Nagata Y, Lan KH, Zhou X, Tan M, Esteva FJ, Sahin AA et al. PTEN factor receptors: a review of clinical research with a focus on non- activation contributes to tumor inhibition by trastuzumab, and loss of small-cell lung cancer. Lancet Oncol 2003; 4: 397–406. PTEN predicts trastuzumab resistance in patients. Cancer Cell 2004; 6: 79 Kris MG, Natale RB, Herbst RS, Lynch Jr TJ, Prager D, Belani CP et al. 117–127. Efficacy of gefitinib, an inhibitor of the epidermal growth factor 59 Agus DB, Akita RW, Fox WD, Lewis GD, Higgins B, Pisacane PI et al. receptor tyrosine kinase, in symptomatic patients with non-small cell Targeting ligand-activated ErbB2 signaling inhibits breast and prostate lung cancer: a randomized trial. JAMA 2003; 290: 2149–2158. tumor growth. Cancer Cell 2002; 2: 127–137. 80 Fukuoka M, Yano S, Giaccone G, Tamura T, Nakagawa K, Douillard JY 60 Yakes FM, Chinratanalab W, Ritter CA, King W, Seelig S, Arteaga CL. et al. Multi-institutional randomized phase II trial of gefitinib Herceptin-induced inhibition of phosphatidylinositol-3 kinase and Akt for previously treated patients with advanced non-small-cell is required for antibody-mediated effects on p27, cyclin D1, and lung cancer (The IDEAL 1 Trial) (corrected). J Clin Oncol 2003; 21: antitumor action. Cancer Res 2002; 62: 4132–4141. 2237–2246.

The Pharmacogenomics Journal Somatic pharmacogenomics ON Ikediobi 313

81 Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, 100 Tsao MS, Sakurada A, Cutz JC, Zhu CQ, Kamel-Reid S, Squire J et al. Brannigan BW et al. Activating mutations in the epidermal growth Erlotinib in lung cancer—molecular and clinical predictors of outcome. factor receptor underlying responsiveness of non-small-cell lung N Engl J Med 2005; 353: 133–144. cancer to gefitinib. N Engl J Med 2004; 350: 2129–2139. 101 Han SW, Kim TY, Hwang PG, Jeong S, Kim J, Choi IS et al. Predictive 82 Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S et al. EGFR and prognostic impact of epidermal growth factor receptor mutation mutations in lung cancer: correlation with clinical response to gefitinib in non-small-cell lung cancer patients treated with gefitinib. J Clin therapy. Science 2004; 304: 1497–1500. Oncol 2005; 23: 2493–2501. 83 Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I et al. EGF 102 Mitsudomi T, Kosaka T, Endoh H, Horio Y, Hida T, Mori S et al. receptor gene mutations are common in lung cancers from ‘never Mutations of the epidermal growth factor receptor gene predict smokers’ and are associated with sensitivity of tumors to gefitinib and prolonged survival after gefitinib treatment in patients with non-small- erlotinib. Proc Natl Acad Sci USA 2004a; 101: 13306–13311. cell lung cancer with postoperative recurrence. J Clin Oncol 2005; 23: 84 AstraZeneca Pharmaceuticals. Gefitinib (Iressat) Marketing Authoriza- 2513–2520. tion Application Withdrawn in EU. Press Release, January 4, 2005. 103 Uramoto H, Sugio K, Oyama T, Ono K, Sugaya M, Yoshimatsu T et al. Available at http://www.astrazeneca.com/pressrelease/4442.aspx. Epidermal growth factor receptor mutations are associated with Accessed June 13, 2008. gefitinib sensitivity in non-small cell lung cancer in Japanese. Lung 85 US Food and Drug Administration. IRESSAs (ZD1839, gefitinib) Cancer 2006; 51: 71–77. Tablets. Oncologic Drugs Advisory Committee (ODAC) Meeting 104 Inoue A, Suzuki T, Fukuhara T, Maemondo M, Kimura Y, Morikawa N Briefing Document, March 4, 2005. Available at http://www.fda. et al. Prospective phase II study of gefitinib for chemotherapy-naive gov/ohrms/dockets/ac/05/briefing/2005-4095B2_01_01-AstraZeneca- patients with advanced non-small-cell lung cancer with epidermal Iressa.pdf. Accessed June 13, 2008.. growth factor receptor gene mutations. J Clin Oncol 2006; 24: 86 Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, 3340–3346. Thongprasert S et al. Erlotinib in previously treated non-small-cell lung 105 Sunaga N, Tomizawa Y, Yanagitani N, Iijima H, Kaira K, Shimizu K et al. cancer. N Engl J Med 2005; 353: 123–132. Phase II prospective study of the efficacy of gefitinib for the treatment 87 Shigematsu H, Lin L, Takahashi T, Nomura M, Suzuki M, Wistuba II of stage III/IV non-small cell lung cancer with EGFR mutations, et al. Clinical and biological features associated with epidermal growth irrespective of previous chemotherapy. Lung Cancer 2007; 56: factor receptor gene mutations in lung cancers. J Natl Cancer Inst 383–389. 2005a; 97: 339–346. 106 Sutani A, Nagai Y, Udagawa K, Uchida Y, Koyama N, Murayama Y 88 Guo M, Liu S, Lu F. Gefitinib-sensitizing mutations in esophageal et al. Gefitinib for non-small-cell lung cancer patients with epidermal carcinoma. N Engl J Med 2006; 354: 2193–2194. growth factor receptor gene mutations screened by peptide nucleic 89 Gwak GY, Yoon JH, Shin CM, Ahn YJ, Chung JK, Kim YA et al. Detection acid-locked nucleic acid PCR clamp. Br J Cancer 2006; 95: 1483–1489. of response-predicting mutations in the kinase domain of the 107 Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst epidermal growth factor receptor gene in cholangiocarcinomas. RS et al. Mutations in the epidermal growth factor receptor and in J Cancer Res Clin Oncol 2005; 131: 649–652. KRAS are predictive and prognostic indicators in patients with non- 90 Lee JW, Soung YH, Kim SY, Nam HK, Park WS, Nam SW et al. Somatic small-cell lung cancer treated with chemotherapy alone and in mutations of EGFR gene in squamous cell carcinoma of the head and combination with erlotinib. J Clin Oncol 2005; 23: 5900–5909. neck. Clin Cancer Res 2005; 11: 2879–2882. 108 Kobayashi S, Boggon TJ, Dayaram T, Janne PA, Kocher O, Meyerson M 91 Nagahara H, Mimori K, Ohta M, Utsunomiya T, Inoue H, Barnard GF et al. EGFR mutation and resistance of non-small-cell lung cancer to et al. Somatic mutations of epidermal growth factor receptor in gefitinib. N Engl J Med 2005a; 352: 786–792. colorectal carcinoma. Clin Cancer Res 2005; 11: 1368–1371. 109 Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF et al. 92 Schilder RJ, Sill MW, Chen X, Darcy KM, Decesare SL, Lewandowski G Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is et al. Phase II study of gefitinib in patients with relapsed or persistent associated with a second mutation in the EGFR kinase domain. PLoS ovarian or primary peritoneal carcinoma and evaluation of epidermal Med 2005a; 2: e73. growth factor receptor mutations and immunohistochemical expres- 110 Bell DW, Gore I, Okimoto RA, Godin-Heymann N, Sordella R, Mulloy R sion: a Gynecologic Oncology Group Study. Clin Cancer Res 2005; 11: et al. Inherited susceptibility to lung cancer may be associated 5539–5548. with the T790M drug resistance mutation in EGFR. Nat Genet 93 Kwak EL, Jankowski J, Thayer SP, Lauwers GY, Brannigan BW, Harris PL 2005a; 37: 1315–1316. et al. Epidermal growth factor receptor kinase domain mutations in 111 Kobayashi S, Ji H, Yuza Y, Meyerson M, Wong KK, Tenen DG et al. An esophageal and pancreatic adenocarcinomas. Clin Cancer Res 2006; alternative inhibitor overcomes resistance caused by a mutation of the 12: 4283–4287. epidermal growth factor receptor. Cancer Res 2005b; 65: 7096–7101. 94 Mellinghoff IK, Wang MY, Vivanco I, Haas-Kogan DA, Zhu S, Dia EQ 112 Kwak EL, Sordella R, Bell DW, Godin-Heymann N, Okimoto RA, et al. Molecular determinants of the response of glioblastomas to EGFR Brannigan BW et al. Irreversible inhibitors of the EGF receptor may kinase inhibitors. N Engl J Med 2005; 353: 2012–2024. circumvent acquired resistance to gefitinib. Proc Natl Acad Sci USA 95 Ji H, Zhao X, Yuza Y, Shimamura T, Li D, Protopopov A et al. Epidermal 2005; 102: 7665–7670. growth factor receptor variant III mutations in lung tumorigenesis and 113 Kakiuchi S, Daigo Y, Ishikawa N, Furukawa C, Tsunoda T, Yano S et al. sensitivity to tyrosine kinase inhibitors. Proc Natl Acad Sci USA 2006; Prediction of sensitivity of advanced non-small cell lung cancers to 103: 7817–7822. gefitinib (Iressa, ZD1839). Hum Mol Genet 2004; 13: 3029–3043. 96 Kaye FJ. A curious link between epidermal growth factor receptor 114 Zhou BB, Peyton M, He B, Liu C, Girard L, Caudler E et al. Targeting amplification and survival: effect of ‘allele dilution’ on gefitinib ADAM-mediated ligand cleavage to inhibit HER3 and EGFR pathways sensitivity? J Natl Cancer Inst 2005; 97: 621–623. in non-small cell lung cancer. Cancer Cell 2006; 10: 39–50. 97 Riely GJ, Politi KA, Miller VA, Pao W. Update on epidermal growth 115 Cappuzzo F, Varella-Garcia M, Shigematsu H, Domenichini I, factor receptor mutations in non-small cell lung cancer. Clin Cancer Res Bartolini S, Ceresoli GL et al. Increased HER2 gene copy number is 2006b; 12: 7232–7241. associated with response to gefitinib therapy in epidermal growth 98 Riely GJ, Pao W, Pham D, Li AR, Rizvi N, Venkatraman ES et al. factor receptor-positive non-small-cell lung cancer patients. J Clin Clinical course of patients with non-small cell lung cancer and Oncol 2005; 23: 5007–5018. epidermal growth factor receptor exon 19 and exon 21 mutations 116 Wang SE, Narasanna A, Perez-Torres M, Xiang B, Wu FY, Yang S et al. treated with gefitinib or erlotinib. Clin Cancer Res 2006a; 12: HER2 kinase domain mutation results in constitutive phosphorylation 839–844. and activation of HER2 and EGFR and resistance to EGFR tyrosine 99 Bell DW, Lynch TJ, Haserlat SM, Harris PL, Okimoto RA, Brannigan BW kinase inhibitors. Cancer Cell 2006; 10: 25–38. et al. Epidermal growth factor receptor mutations and gene 117 Pao W, Wang TY, Riely GJ, Miller VA, Pan Q, Ladanyi M et al. KRAS amplification in non-small-cell lung cancer: molecular analysis of the mutations and primary resistance of lung adenocarcinomas to gefitinib IDEAL/INTACT gefitinib trials. J Clin Oncol 2005b; 23: 8081–8092. or erlotinib. PLoS Med 2005b; 2: e17.

The Pharmacogenomics Journal Somatic pharmacogenomics ON Ikediobi 314

118 Sordella R, Bell DW, Haber DA, Settleman J. Gefitinib-sensitizing EGFR 124 Parsons DW, Wang TL, Samuels Y, Bardelli A, Cummins JM, DeLong L et al. mutations in lung cancer activate anti-apoptotic pathways. Science Colorectal cancer: mutations in a signaling pathway. Nature 2005; 2004; 305: 1163–1167. 436: 792. 119 Cappuzzo F, Magrini E, Ceresoli GL, Bartolini S, Rossi E, Ludovini V 125 Solit DB, Garraway LA, Pratilas CA, Sawai A, Getz G, Basso A et al. BRAF et al. Akt phosphorylation and gefitinib efficacy in patients with mutation predicts sensitivity to MEK inhibition. Nature 2006; 439: advanced non-small-cell lung cancer. J Natl Cancer Inst 2004; 96: 358–362. 1133–1141. 126 Niculescu-Duvaz I, Roman E, Whittaker SR, Friedlos F, Kirk R, Scanlon IJ 120 Pao W, Miller VA, Venkatraman E, Kris MG. Predicting sensitivity of et al. Novel inhibitors of the v-raf murine sarcoma viral oncogene non-small-cell lung cancer to gefitinib: is there a role for P-Akt? J Natl homologue B1 (BRAF) based on a 2,6-disubstituted pyrazine scaffold. Cancer Inst 2004b; 96: 1117–1119. J Med Chem 2008; 51: 3261–3274. 121 Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO 127 Ikediobi ON, Reimers M, Durinck S, Blower PE, Futreal PA, Stratton MR et al. MET amplification leads to gefitinib resistance in lung cancer by et al. In vitro differential sensitivity of melanomas to phenothiazines is activating ERBB3 signaling. Science 2007; 316: 1039–1043. based on the presence of codon 600 BRAF mutation. Mol Cancer Ther 122 Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S et al. Mutations 2008; 7: 1337–1346. oftheBRAFgeneinhumancancer.Nature 2002; 417: 949–954. 128 Yu K, Toral-Barza L, Shi C, Zhang WG, Zask A. Response and 123 Stephens P, Hunter C, Bignell G, Edkins S, Davies H, Teague J et al. determinants of cancer cell susceptibility to PI3K inhibitors: combined Lung cancer: intragenic ERBB2 kinase mutations in tumors. Nature targeting of PI3K and Mek1 as an effective anticancer strategy. Cancer 2004; 431: 525–526. Biol Ther 2008; 7: 307–315.

The Pharmacogenomics Journal