Abstract
Dysregulation of growth factor cell signaling is a major driver of most human cancers. This has led to development of numerous drugs targeting protein kinases, with demonstrated efficacy in the treatment of a wide spectrum of cancers. Despite their high initial response rates and survival benefits, the majority of patients eventually develop resistance to these targeted therapies. This review article discusses examples of established mechanisms of drug resistance to anticancer therapies, including drug target mutations or gene amplifications, emergence of alternate signaling pathways, and pharmacokinetic variation. This reveals a role for pharmacogenomic analysis to identify and monitor for resistance, with possible therapeutic strategies to combat chemoresistance.
Keywords: pharmacogenetics, pharmacogenomics, cancer, resistance, somatic mutations
Introduction
Cancer is a genetic disease that arises primarily from the accumulation of genetic changes in genes regulating cellular growth, proliferation, and survival. Gain of function alterations inducing hyperactivity of oncogenes or loss of function alterations leading to inactivation of tumor suppressor genes cause deregulation of cellular signaling, a fundamental trait of cancer cells. In healthy cells, homeostasis is conveyed via growth factors binding to cell surface receptors, primarily protein kinases, which then activate intracellular signaling pathways and regulate cell cycle progression (Hanahan and Weinberg, 2011). Deregulation of these signals results in uncontrollable cellular proliferation, metabolism, survival and, ultimately, cancer. Somatic (acquired, or tumor) mutations lead to constitutive activation of these signaling pathways. For example, mutations in the B-raf proto-oncogene, BRAF, a serine/threonine kinase, cause constitutive signaling through the mitogen-activated protein kinase (MAPK) pathway and are commonly observed in melanoma, colorectal (CRC), and papillary thyroid cancers (Araya et al., 2016). Identification of the genes and pathways deregulated in cancer, such as BRAF, has led to a rapid increase in the design and approval of therapies targeting these genetic drivers of oncogenesis. Targeted anti-cancer therapies function by interfering with specific molecular alterations that regulate cellular signaling and drive tumor growth. By binding to or inhibiting a known molecular driver, targeted therapies interrupt the signaling pathways, causing cellular deregulation and leading to cancer cell apoptosis or cell death. For example, vemurafenib selectively inhibits BRAF V600-mutated cancer cells, abrogating MAPK-mediated signaling, preventing proliferation of BRAF-mutated cells, and ultimately resulting in apoptosis (Tsai et al., 2008).
To optimize the use of targeted therapies, the genetic alterations causing pathway deregulation in each patient’s tumor must be identified. This is the modern concept of personalized cancer medicine (or precision medicine). Pharmacogenomics is the study of how genetic variations influence the response of an individual to drugs. In the context of cancer, there are two genomes relevant to predicting drug response or resistance: (1) germline, or inherited, genetics may affect drug exposure, potentially causing variability in efficacy and/or toxicity, and (2) somatic, or tumor, genetics are the acquired alterations that may initiate and perpetuate cellular deregulation. Generally, it is the somatic germline that is interrogated to identify alterations driving oncogenesis and to select targeted therapies. It should be emphasized that drug resistance phenomena continue to be a primary hindrance to curative chemotherapy of solid tumors and hematologic malignancies (Fletcher et al., 2016; Niewerth et al., 2015; Wicki et al., 2016). Hence, deciphering the molecular mechanisms underlying chemoresistance should enhance targeted individualized cancer medicine (Assaraf et al., 2014; Livney and Assaraf, 2013; Swanton et al., 2016).
Due to their critical role in regulating cellular signaling, > 20 protein kinase inhibitors (PKIs) have been developed and approved across a wide range of tumor types. Despite their overall high response rates, many patients for whom the drugs are indicated will not have any evidence of disease control, while others will have transient benefit followed by tumor growth. This lack of complete and durable responses is indicative of drug resistance phenomena. If the correct alteration driving tumor growth is not identified from the outset, intrinsic resistance may be observed. When the tumor cells evolve to overcome targeted inhibition, the patient develops acquired resistance and stops responding to therapies that were previously effective. In this review, we will elaborate on the various classifications of cancer drug resistance, provide examples of how pharmacogenomics plays a role in resistance to PKIs, and discuss possible therapeutic strategies to overcome cancer drug resistance.
Classifications of drug resistance mechanisms important in cancer
Drug resistance can be defined as the lack of therapeutic benefit or response to a medication. In the cancer setting, drug resistance is apparent with an increase in tumor size or metastasis (i.e. disease progression). Various mechanisms of chemoresistance can result in lack of complete or durable response to cancer therapies (Fig. 1). An overview of the common classifications is provided below. It is important to note that a single pharmacogenomic biomarker may represent multiple mechanisms of drug resistance.
Figure 1. Mechanisms of oncology drug resistance.
(A) Pharmacological resistance. Drug-drug interactions and germline pharmacogenetic variants can affect drug exposure at the tumor site. Pharmacological properties can affect drug penetration into the central nervous system. (B) Biological resistance. Somatic (acquired) mutations in the drug target can affect the drug’s ability to effectively inhibit oncogenesis. Somatic alterations downstream of the drug target can result in constitutive upregulation of oncogenic pathways. Genetic alterations may also activate alternative oncogenic signaling pathways. Some tumor types have been shown to transform into other tumor types (e.g., non-small cell lung cancer to small cell lung cancer). (C) Pharmacological drug resistance results from inadequate drug levels at the site of action, whereas biological drug resistance results despite adequate drug levels at the site of action. (D) Intrinsic resistance is the lack of even transitory clinical benefit – the tumor continues to progress despite treatment. (E) Acquired resistance is the lack of tumor response to medication despite initial benefit.
Pharmacological vs. biological resistance
Pharmacological resistance reflects inadequate drug exposure at the drug target, and can be caused by environmental factors (e.g., drug-drug interactions, non-compliance), germline pharmacogenomics (i.e., inter-individual variability in drug metabolism or pharmacokinetics) (Fig. 1A & 1C) as well as drug sequestration away from its target (Goler-Baron and Assaraf, 2011; Zhitomirsky and Assaraf, 2016). For example, addition of an antacid to alleviate gastroesophageal reflux caused by some PKI therapies will affect gastric absorption and exposure to the PKI, thereby leading to pharmacological resistance (Budha et al., 2012). Additionally, the observation that PKI-sensitive clones reemerge post-PKI discontinuation demonstrates the cytostatic, rather than cytotoxic, nature of some targeted therapies (Browning et al., 2013; Sequist et al., 2011). Therefore, inconsistent suppression of the drug target due to missed doses may lead to upregulation of the cancer-driving pathways and, ultimately, cancer progression. Biological resistance results from cancer cell evolution in the presence of adequate drug exposure (Fletcher et al., 2016; Liu et al., 2016; Niewerth et al., 2015). In the context of cancer, biological resistance can arise from somatic alterations in drug targets or pathways (Fig. 1B & 1C). Examples include genetic alterations in the drug target itself, activation of alternative signaling pathways (bypass tracks), alteration in signaling proteins downstream of the drug target, or phenotypic switch. Most known mechanisms of resistance to targeted cancer therapies are of the biological resistance subtype; hence, these will be the primary focus of this article.
Intrinsic vs. acquired resistance
Cancer drug resistance can also be classified based on timing during the course of treatment. Intrinsic resistance (also referred to as innate, inherent, or primary resistance) is the absence of discernible, even transitory beneficial effect from a medication. From the outset, there is neither cessation in tumor growth nor increase in survival benefits (Fig. 1D). Evidence of intrinsic resistance can be visualized in a waterfall plot, in which some patients fail to ever meet Response Evaluation Criteria in Solid Tumors (RECIST). For example, approximately 20% of BRAF V600-mutated melanoma patients do not respond to BRAF inhibitors (Hauschild et al., 2012), demonstrating intrinsic drug resistance. Mechanisms of intrinsic resistance can include germline or somatic alterations. Intrinsic resistance is a major challenge in cancer therapy, and it is important to elucidate the mechanisms conferring lack of therapeutic benefit. However, these mechanisms remain less well understood at this time.
Acquired or secondary resistance is the progression of disease after an initial benefit. In oncology, the tumor initially shrinks (responds) but eventually begins to increase in size (Fig. 1E). While intrinsic drug resistance can be due to germline or somatic mutations, acquired resistance is most commonly attributed to somatic mutations (an exception would be a new drug-drug interaction due to changes in therapy). Mechanisms that result in acquired drug resistance include genetic alterations in the drug target, activation of bypass tracks, alteration in downstream signaling proteins, phenotypic switch, drug efflux, drug catabolism as well as drug compartmentalization (Camidge et al., 2014; Gridelli et al., 2014; Liu et al., 2016).
Germline pharmacogenomics as a mechanism of pharmacological resistance
Germline pharmacogenomics can affect one’s inherent response to a medication or therapy. In the context of cancer, germline pharmacogenomics has most widely been associated with risk of developing adverse effects, rather than drug resistance (Hertz and Rae, 2015). Since adverse effects generally result from off-target (i.e., non-tumor) effects, it is logical that genetic variation in the germ cells throughout the body would confer risk to adverse events. However, due to the inherited nature of germline genetics, they may also play a role in one’s initial response to therapy. In fact, there are a few well-studied examples of how germline variation can confer intrinsic resistance to anticancer medications (Table 1).
Table 1.
Summary of FDA-approved protein kinase inhibitors and known mechanisms of resistance.
Class | Drug | Generation | Indication | Indication subgroup | Innate (primary) resistance | Acquired (secondary) resistance |
---|---|---|---|---|---|---|
EGFR inhibitors |
erlotinib | 1st | NSCLC | EGFR exon 19 deletions or exon 21 (L858R) substitution mutations |
EGFR wt, BIM deletion | secondary EGFR mutations (e.g., T790M), MET amplification, HER2 amplification (see Fig. 3) |
gefetinib | 1st | NSCLC | ||||
afatinib | 2nd | NSCLC | ||||
osimertinib | 3rd | NSCLC | EGFR T790M–positive | EGFR wt | loss of T790M, secondary EGFR mutations (e.g., C797S) |
|
ALK inhibitors | crizotinib | 1st | NSCLC | ALK+, recommended for pts with ROS1 translocation |
ALK wt | secondary ALK mutations, ALK fusion amplification, bypass signaling (e.g., EGFR, KIT, IGF1R) (see Fig. 3) |
ceritinib | 2nd | NSCLC | ALK+, resistant to crizotinib, ALK I1171N- positive |
ALK wt | secondary ALK mutations (e.g., G1202R) |
|
alectinib | 2nd | NSCLC | ALK+, resistant to crizotinib |
secondary ALK mutations (e.g., I1171N) |
||
BCR-ABL inhibitors |
imatinib | 1st | CML, ALL, MDS, GIST |
Ph+ | BIM deletion; KIT and PDGFRA wt (GIST) |
BCR-ABL1 T315I and others in heme; KIT and PDGFR secondary mutations in GIST |
bosutinib | 2nd | CML | ||||
dastinib | 2nd | CML, ALL | Ph+ | BCR-ABL1 T315I and others | ||
nilotinib | 2nd | CML | ||||
ponatinib | 3rd | CML,ALL | Ph+ for whom no other TKI is indicated, or T315I positive |
BCR-ABL1 compound mutations |
||
BTK inhibitor | ibrutinib | CLL, MCL, WM | C481S, point mutations in phospholipase Cy2 (PLCg2) |
Mutations in BTK and downstream |
||
HER2 inhibitor | lapatinib | breast | HER2+ | deregulation of PIK3CA pathway, AXL over-espression? |
||
BRAF inibitiors | dabrafenib | melanoma | BRAF V600E or V600K | RAC1 mutations, loss of PTEN or NF1, CCND1 over- expression, abundance of HGF |
re-activation of the MAPK pathway, activating mutations in NRAS, activating MEK 1/2 mutations, evelated CRAF (see Fig. 3) |
|
vemurafenib | melanoma | BRAF V600E | ||||
MEK inhibitors | trametinib | melanoma | BRAF V600E or V600K | |||
mTOR inhibitor |
everolimus | breast, pNET, RCC, angiolyolipoma, SEGA |
breast HER2- | TSC1/TSC2 mutations predict response |
||
EGFR monoclonal antibodies |
cetuximab | RAS mutations | ||||
panitumumab | ||||||
ER inhibitors | tamoxifen, fulvustrant | ESR1 mutations | ||||
HER2 inhibitors |
trastuzumab | breast, gastric | HER2+ | PIK3CA mutations possibly predictive of response in neoadjuvant setting (not predictive in adjuvant) |
||
SMO inhibitor | vismodegib | basal cell carcinoma |
SMO mutations |
Germline variation in TPMT as a pharmacogenomics predictor of response and adverse events
Germline variation in TPMT, the gene that encodes the thiopurine S-methyltransferase (TPMT) enzyme, is known to affect response to thiopurine drugs. Chemotherapeutic thiopurines include 6-mercaptopurine and 6-thioguanine, which are used in the treatment of pediatric and adult acute lymphocytic leukemia (ALL) (azathioprine is a prodrug of 6-mercaptopurine used in non-malignant conditions). Thiopurine drugs are inactive prodrugs that are bioactivated and metabolized via competing routes: (1) xanthine oxidase converts 6-mercaptopurine to an inactive metabolite, 6-thiouric acid; (2) hypoxanthine phosphoribosyltransferase (HPRT) converts thiopurines to activated nucleotide analogues, which can be incorporated into DNA or RNA, hence interfering with replication and transcription, resulting in cytotoxicity; and (3) TPMT inactivates thiopurines through methylation. The nucleotide analogues formed by HPRT are responsible for the efficacy and toxicity of thiopurine drugs, and insufficient TPMT activity results in upregulation of HPRT-mediated metabolism, conferring response. Patients with low TPMT activity have greater exposure to activated thioguanine nucleotides, resulting in the potential for greater efficacy, but also increased risk of severe toxicity (Lennard et al., 1997). This variation in activated nucleotide exposure is well-established as a predictor of treatment outcomes. Patients with TPMT loss-of-function variants have significantly lower rates of minimal residual disease positivity after receiving 6-mercaptopurine therapy when compared to wild type TPMT individuals (Stanulla et al., 2005). Increased risk of relapse has also been associated with wild type TPMT in children receiving 6-mercaptopurine, likely due to insufficient thioguanine nucleotide exposure, thus conferring a type of pharmacological resistance (Schmiegelow et al., 2009). Due to increased toxicity risk, decreased dosing for patients with TPMT variants has been recommended (Relling et al., 2011). However, it is unclear how this may affect relapse rates (Levinsen et al., 2014; Relling et al., 2006). A recent study reported that patients with 6-mercaptopurine non-adherence were at a 2.7-fold increased risk of relapse when compared to patients with a mean drug adherence rate of 95% or greater (p = 0.01), further emphasizing the importance of continuous drug exposure and adherence as a means to avoid development of drug resistance phenomena (Bhatia et al., 2015).
Germline alterations in BIM as a predictor of intrinsic pharmacological resistance
A common variant in BCL2L11 (also known as BIM), the gene that encodes the BCL2-like 11 protein, has been associated with intrinsic resistance to PKIs. BIM is a member of the B-cell CLL/lymphoma 2 (Bcl-2) family of genes and encodes a Bcl-2 homology domain 3 (BH3). BH3 activates cell death by either opposing the pro-survival members of the Bcl-2 family or by binding to the pro-apoptotic Bcl-2 family members and causing activation of their pro-apoptotic functions (Youle and Strasser, 2008). PKIs induce upregulation and stabilization of BIM through inhibition of the MAPK pathway, therefore, the activity of BIM is required for PKIs to induce apoptosis in kinase-driven cancers (Gong et al., 2007). Recently, a 2,903 bp germline deletion polymorphism in intron 2 of BIM was identified, which was associated with inferior responses to PKIs (i.e., imatinib, gefitinib, erlotinib, and afatinib) in chronic myeloid leukemia (CML), non-small cell lung cancer (NSCLC), and pediatric ALL patients (Lee et al., 2014; Ng et al., 2012; Soh et al., 2014). Functionally, this mutation results in alternative RNA splicing, leading to decreased production of BIM isoforms containing the essential BH3 domain.
Since its discovery, conflicting evidence of the ability of BIM variation to predict intrinsic resistance to PKIs has been documented (Chen et al., 2014; Cheng and Sawyers, 2012; Isobe et al., 2014). Two retrospective studies failed to observe an association between BIM genotype and response rates to PKIs in NSCLC patients (Lee et al., 2013; Lee et al., 2015a). However, a systematic review and meta-analysis of 951 patients supported the BIM deletion polymorphism as a predictor of shorter progression free survival (PFS) in NSCLC patients who were treated with PKIs (adjusted HR = 2.38, p < 0.001) (Nie et al., 2015). Another meta-analysis found that the BIM deletion polymorphism was associated with response rates (HR = 0.44, 95% CI = 0.27–0.7) and PFS (HR = 2.19, 95% CI = 1.7–2.8) in NSCLC, but not in CML (Ying et al., 2015). Further evidence indicating a lack of benefit or increased risk of harm in individuals carrying BIM deletions must be generated before this biomarker of intrinsic resistance can reasonably be implemented in clinical practice.
Methods to overcome BIM-related PKI resistance are already being explored. A preclinical study in NSCLC cell lines and xenograft models indicated that cells harboring the common BIM deletion had enhanced response to gefitinib when treated in combination with a histone deacetylase inhibitor, vorinostat (Nakagawa et al., 2013). Vorinostat functioned by increasing expression of BH3 in a dose-dependent manner, thus restoring sensitivity to tyrosine kinase inhibition. These findings further support the importance of BIM expression in PKI response and provide evidence to suggest that combination therapeutics may be a potential strategy to overcome this form of resistance.
Additional germline pharmacogenomic markers as predictors of drug resistance
One potential mechanism that can confer pharmacological resistance is decreased exposure at the drug target, which can result from drug-drug interactions or inter-individual genetic variability (Fig. 1A). There are a few well-established examples of germline genetics affecting exposure to anticancer therapies [reviewed in (Hertz and Rae, 2015)]. While outside the scope of this review, the importance of an established link between active drug exposure levels and clinical outcomes or adverse events must be noted. Drug exposure is predicted to affect drug efficacy or toxicity. However, discrete evidence must exist before clinical implementation is warranted (Gillis and Innocenti, 2014).
Somatic pharmacogenomics as a mechanism of drug resistance
Somatic mutations result in upregulation of oncogenic pathways, and their effects can be inhibited with the use of targeted therapies. Since 2003, over 20 PKIs have been approved to target various somatic alterations across a broad range of cancer types (including hematologic and solid malignancies), and more than 20 additional PKIs are currently in clinical trials (CenterWatch, 2016). Because these drugs target protein kinases, somatic alterations in the targets or pathways may confer resistance or response (Table 1). Some well-established examples of somatic genetic drivers of resistance to PKIs are discussed below.
RAS status as a predictor of intrinsic resistance to EGFR inhibition in CRC
The epidermal growth factor receptor (EGFR or ErbB-1) is a transmembrane protein kinase that binds epidermal growth factor, inducing dimerization and autophosporylation, which signals downstream pathways (i.e., MAPK and PI3K/Akt) that mediate cellular proliferation and survival (Fig.2). The EGFR signaling pathway plays a pivotal role in tumor growth and progression in many cancer types, including glioblastoma, NSCLC, head and neck cancers, and CRC. EGFR is overexpressed in approximately 50% of CRC patients, and is associated with disease progression and poor prognosis (Siena et al., 2009). Anti-EGFR monoclonal antibodies, such as cetuximab and panitumumab, were hypothesized to be effective in colorectal tumors over-expressing EGFR, and were initially U.S. Food and Drug Administration (FDA) approved for that indication. However, as monotherapy, the response rates to cetuximab and panitumumab were only 10% and 30%, respectively, indicating potential intrinsic resistance (Jonker et al., 2007; Van Cutsem et al., 2007). Retrospective analysis of phase III clinical study data revealed differential response to anti-EGFR monoclonal antibodies dependent on Kirsten rat sarcoma viral oncogene (KRAS) homolog (Amado et al., 2008). When stratified by KRAS mutation status, response rate to panitumumab in wild type individuals was 17%, whereas 0% of individuals with mutant KRAS responded. Similarly, KRAS mutations were associated with resistance and decreased overall survival (OS) in patients receiving cetuximab (Lievre et al., 2006). These observations provided evidence for KRAS as a biomarker of intrinsic resistance to EGFR-targeted monoclonal antibodies.
Figure 2. Example mechanisms of resistance to EGFR inhibitors and potential treatment strategies: The MAPK and PI3K/AKT pathways.
EGFR-mutated non-small cell lung cancers show initial response to EGFR inhibitors (e.g., erlotinib, gefetinib). Mechanisms of acquired resistance include secondary EGFR mutations, downstream mutations that result in EGFR-independent activation of MAPK or PI3K/AKT signaling pathways, or mutations in alternative protein kinases that bypass EGFR-mediated signaling through MAPK and/or PI3K/AKT pathways. Potential treatment strategies to combat acquired resistance to EGFR inhibitors include stronger inhibiton of EGFR (e.g., afatinib), combination therapy with a MEK (e.g., trametinib) or mTOR (e.g., sirolimus) inhibitor, or inhibition of bypass tracks.
KRAS is a member of the rat sarcoma virus (RAS) family of oncogenes, which also includes HRAS (Harvey rat sarcoma viral oncogene homolog) and NRAS (neuroblastoma RAS viral oncogene homolog). Mutations in RAS genes lead to the constitutive activation of the MAPK pathway independent of EGFR status. Interestingly, in CRC, mutations in KRAS are significant enough to negate EGFR inhibition. A prospective-retrospective analysis of 1,183 patients who received either FOLFOX-panitumumab or FOLFOX alone revealed that mutation status in both KRAS and NRAS were predictive of response to panitumumab (HR for progression or death in RAS wild type 0.72, 95% CI 0.58–0.99, p= 0.0004) (Douillard et al., 2013). Recently, the American Society of Clinical Oncology released a provisional clinical opinion update recommending NRAS and KRAS mutation testing in CRC patients who are candidates for anti-EGFR monoclonal antibodies, supporting the significance of this biomarker as a predictor of intrinsic drug resistance (Allegra et al., 2016). Additional potential markers of intrinsic resistance have since been identified in models of KRAS wild-type patient xenografts, including ERBB2 (HER2), FGFR1, PDGFRA, and MAP2K1 (MEK); secondary mutations in EGFR at the site of cetuximab binding were identified in acquired resistance (Bertotti et al., 2015). Further studies demonstrating effects on patient outcomes are needed before clinical implementation of these novel biomarkers can be recommended.
EGFR status as a mediator of resistance to EGFR PKIs in NSCLC
Mutations in EGFR are one of the most common cancer drivers identified in NSCLC tumors. Approximately 15% of NSCLC patients in the U.S. will have an EGFR mutation, and the incidence is approximately 35% in patients of Asian descent (Shi et al., 2014b). In the U.S., EGFR mutations are most prevalent in females (17.9% vs. 8%, p= 0.002), non-smokers (42% vs. 6.6%, p < 0.001), and adenocarcinomas (15.6%) (Bauml et al., 2013). The mutations are typically located in exons 18 to 21 of EGFR, the region that encodes the catalytic tyrosine kinase domain. Approximately 90% of the mutations are short exon 19 deletions or the L858R point mutation in exon 21, which result in enhanced EGFR signaling (Sharma et al., 2007). Mutations in EGFR are predictive of response to EGFR inhibitors, such as erlotinib and gefitinib, in NSCLC. However, despite 70–80% response rates to EGFR PKIs, a majority of patients will acquire resistance after 10–12 months (Mitsudomi et al., 2010; Zhou et al., 2011). Additionally, absence of initial EGFR mutations confers lower overall response rates in NSCLC patients, a relative intrinsic resistance (Morgensztern et al., 2015; Yang et al., 2015).
Acquired resistance to EGFR inhibitors in NSCLC is complex and heterogeneous, but ultimately all mechanisms drive sustained signaling through downstream cancer pathways (e.g., MAPK or PI3K/Akt pathways). Known mechanisms of drug resistance include secondary genetic alterations in EGFR, upregulation of parallel signaling pathways, or phenotypic transformation (Fig. 2). The most common mechanism of acquired resistance in EGFR-positive NSCLCs is the single-nucleotide mutation T790M, which occurs in approximately 50–60% of acquired resistance cases (Sharma et al., 2007). Also known as the gatekeeper residue, substitution of bulky methionine at this position causes steric hindrance and prevents EGFR inhibitors from binding and eliciting their pharmacologic effect. Other EGFR mutations have been identified in patients with acquired resistance, but their frequencies are much lower (Fig. 3). Another common mechanism of acquired resistance to EGFR PKIs is amplification of the MET proto-oncogene (MET), a receptor tyrosine kinase, which has been reported in up to 22% of resistant samples (Engelman et al., 2007). MET amplification drives resistance by inducing EGFR-independent phosphorylation of ERBB3 (HER3), which activates downstream PI3K/Akt signaling despite the presence of an EGFR inhibitor (Engelman et al., 2007). Amplifications of ERBB2 (HER2) and PIK3CA have also been identified in patients with acquired resistance, similarly functioning to activate shared pathways independent of EGFR (Sequist et al., 2011; Yu et al., 2013). Phenotypic transformation as a mechanism of acquired resistance occurs when the histology of the tumor transitions to small cell lung cancer (SCLC) or from epithelial to mesenchymal (EMT) subtype, and occurs in approximately 14% and 5%, respectively (Sequist et al., 2011; Takegawa et al., 2016). Interestingly, transformed SCLCs retain the primary EGFR activating mutations, but do not carry T790M or MET amplification. Not much is known about the mechanism of histological transformation, but currently it is recommended that these patients receive standard SCLC treatments (Oser et al., 2015).
Figure 3. Mechanisms of acquired resistance to protein kinase inhibitors.
(A) and (B) data from Camdige et al., 2014. (C) Data from Shi et al., 2013. The all-blue pie chart represents MAPK-mediated mechanisms of resistance. PI3K/AKT-mediated mechanisms include AKT1/3 mutations (3%), mutations in positive-regulatory genes, PIK3CA and PIK3CG, and mutations in negative-regulatory genes PIK3RS, PTEN, and PHLPP1.
Blue shades throughout correspond to alterations within the targeted oncogenic track; orange shades correspond to alterations regulating alternative or bypass tracks; yellow shades correspond to phenotypic transformations. NSCLC: non-small cell lung cancer, SCLC: small cell lung cancer, EMT: epithelial to mesenchymal subtype.
Numerous strategies are being investigated to overcome first generation EGFR inhibitor resistance due to secondary EGFR mutations or bypass track signaling. One strategy was to develop more potent inhibitors of EGFR. The FDA-approved second-generation EGFR inhibitor, afatinib, which irreversibly binds to and inhibits EGFR as well as HER2, HER3, and HER4. Despite its first-line indication and increased potency, afatinib has not demonstrated promise in the setting of acquired drug resistance. LUX-Lung 1, a phase 2b/3 trial of afatinib in patients who had progressed after treatment with an EGFR PKI, failed to meet its primary endpoint of OS (HR 1.08, 95% CI 0.86–1·35; p=0.74), and response rates were less than 10% (Miller et al., 2012). Demonstrating more promise than the second generation EGFR PKIs in combating acquired resistance are the third generation EGFR inhibitors: the recently approved osimertinib as well as rociletinib that is in clinical trials. Like the second-generation inhibitors, these are irreversible EGFR inhibitors. However, they have higher affinity for mutant EGFR, including T790M, than for wild type EGFR. In the pivotal phase 2 study of osimertinib in NSCLC patients who had progressed after treatment with a first generation PKI, response rates in T790M-positive patients were 61%. In a similar phase 1/2 study of rociletinib, patients with the T790M mutation had an objective response rate of 59% (Janne et al., 2015; Sequist et al., 2015).
Despite their therapeutic promise in acquired resistance to first generation EGFR PKIs, mechanisms of potential acquired resistance to third generation EGFR PKIs have already been reported. A study in patients who progressed on rociletinib reported that half of T790M-positive NCSLCs treated with rociletinib became T790-wild type at progression (Piotrowska et al., 2015). Loss of T790M was also observed in 27% patients who progressed on osimertinib, and 40% acquired a novel EGFR mutation, C797S, which is also resistant to rociletinib (Thress et al., 2015). A strategy that is being tested to overcome loss of T790M is combination therapy of an EGFR PKI with a monoclonal antibody that targets EGFR (e.g., NCT02496663). Recently, a preclinical study identified mutations and amplifications in NRAS and KRAS as mechanisms of acquired resistance to osimertinib (Eberlein et al., 2015). Due to their upstream signaling of MEK, alterations in NRAS and KRAS may confer response to MEK inhibitors. As such, clinical studies investigating combination therapy with third generation EGFR PKIs and MEK inhibitors are also underway (e.g., NCT02580708).
Therapeutic strategies targeting resistance mediated by upregulation of bypass signaling pathways are less established. Postulated strategies mainly consist of studies investigating dual inhibition of EGFR and known bypass tracks. In an ongoing phase 1b/2 clinical study of gefitinib plus a novel MET inhibitor (INC280) in patients who had progressed on a first-line EGFR PKI, partial responses were only observed in 6/41 (15%) individuals (Wu et al., 2014). A phase 1b study investigated concurrent inhibition of HER2 and EGFR with the combination of afatinib (dual HER2/EGFR inhibitor) and cetuximab in NSCLC patients who had progressed on gefitinib or erlotinib, and overall response rate (ORR) was 29%, and PFS was 4.7 months (Janjigian et al., 2014). A phase 2 clinical trial combining erlotinib with a PI3K inhibitor (BKM120) is currently ongoing in patients who acquired resistance to erlotinib (NCT01487265); the combination of afatinib and sirolimus is also being studied to overcome resistance due to mTOR (mechanistic target of rapamycin, a serine/threonine kinase) activation (NCT00993499), a downstream component of the PI3K/Akt pathway. These preliminary results suggest that dual inhibition of multiple signaling pathways may confer response in some individuals. However, an understanding of which individuals will benefit and durability of response is crucial.
Acquired resistance to ALK inhibitors
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase that is normally expressed in the nervous system and plays an important role in brain development. ALK rearrangements, mutations, and amplifications have been identified in numerous tumor types including anaplastic large cell lymphoma, neuroblastoma, and NSCLC. Chromosomal rearrangements resulting in gene fusions are the most common type of ALK alterations. Clinically, ALK alterations are known to be most actionable in NSCLC, where the most common rearrangement observed is echinoderm microtubule associated protein like 4 (EML4)-ALK (Soda et al., 2007). Multiple variants of EML4-ALK have been reported; however, they all encode the same cytoplasmic portion of ALK, but have different EML4 truncations (Choi et al., 2008). Fusions of ALK with other genes have also been described (e.g., KIF5B-ALK, TFG-ALK, and KLC1-ALK), but occur at much lower frequencies (Shaw and Engelman, 2013). These fusion proteins mediate ligand-independent dimerization of ALK, and like EGFR mutations, result in constitutive activation and downstream signaling through the MAPK and PI3K/Akt pathways (Soda et al., 2007). Interestingly, ALK gene arrangements are largely mutually exclusive with EGFR or KRAS mutations (Gainor et al., 2013). Cancers harboring ALK rearrangements, classified as ALK positive, derive clinical benefit from ALK PKI therapies.
Crizotinib is a first generation ALK PKI, which also inhibits the MET and ROS proto-oncogenes (MET and ROS1), which encode receptor tyrosine kinases. Crizotinib first received accelerated FDA approval for ALK-positive NSCLC in 2011 based on durable, objective response rates of 61% in a single-arm phase 1 study, and is now recommended first-line in ALK-rearranged NSCLC (Camidge et al., 2012). Crizotinib inhibits ALK phosphorylation and signal transduction through G1-S phase cell cycle arrest and induction of cellular apoptosis (Christensen et al., 2007). Despite high response rates (74% in the first-line phase 3 study), resistance to crizotinib develops 10–12 months after therapy initiation (Solomon et al., 2014).
As observed with EGFR inhibitors, acquired resistance to crizotinib is heterogeneous and complex (Fig. 3). Mechanisms of resistance that have been reported include somatic alterations (amplification and/or mutation) in ALK, activation of alternative signaling pathways, and genetic alterations in other important oncogenes. A case of phenotypic neuroendocrine transformation was recently reported in a patient who developed acquired resistance to crizotinib (Caumont et al., 2016). Unlike that observed in EGFR inhibitor resistance, only ~one-third of patients with crizotinib resistance harbor an ALK mutation, and there are numerous mutations that can drive resistance. Two of the most commonly observed mutations are L1196M and G1269. Amino acid 1196 is considered the gatekeeper residue of ALK and, similar to EGFR, it controls access to the active site; therefore, the bulky substitution of methionine causes steric hindrance, impeding crizotinib binding (Choi et al., 2010). Individual mutations have been shown to confer variable degrees of crizotinib resistance (Heuckmann et al., 2011; Katayama et al., 2012). Activation of alternative signaling pathways, or bypass tracks, can occur via genomic or non-genomic mechanisms. These alterations lead to constitutive activation of redundant downstream pathways, rendering crizotinib incapable of suppressing tumor growth. Non-genomic mechanisms of resistance include increased phosphorylation of other tyrosine kinases (e.g., EGFR, IGF-1R, and Src); genomic alterations include mutations in EGFR, c-KIT, MAPK, and KRAS (Isozaki et al., 2015) (Fig. 3).
Multiple potential strategies exist to combat crizotinib resistance. The most well established second-line therapeutic option in patients who progress after receiving crizotinib is second-generation ALK inhibitors. Ceritinib, an FDA-approved second-generation ALK inhibitor, is 20-fold more potent against ALK than crizotinib and also inhibits IGF-1R (insulin-like growth factor 1 receptor). In a phase 1 clinical study, among the 80 patients who had received crizotinib previously, there was a 56% response rate to ceritinib in patients with various ALK resistance mutations (Shaw et al., 2014). A preclinical study suggests that some crizotinib-resistance mutations (F1174C and G1202R) may not be sensitive to ceritinib and, in fact, may be mechanisms of acquired resistance to ceritinib as well (Friboulet et al., 2014). Alectinib, another second-generation ALK inhibitor with efficacy in patients who progressed on crizotinib received FDA-approval December 2015 (Shaw et al., 2016b). Interestingly, acquired mutations in ALK (I1171N and F1245C), have been reported to confer resistance to alectinib, but are susceptible to ceritinib (Kodityal et al., 2016; Ou et al., 2015). Interestingly, acquired resistance to lorlatinib (via ALK L1198F), a third-generation ALK inhibitor in clinical trials, has been reported to enhance crizotinib binding and resensitize resistant tumors (Shaw et al., 2016a). Findings such as these may be important when considering sequencing of therapy in resistant patients.
Strategies to overcome crizotinib resistance due to bypass track signaling are less well studied. However, rational combination therapies have been postulated. For example, a preclinical study in ALK-positive NSCLC cell lines with acquired IGF-1R upregulation demonstrated improved efficacy of combined ALK (crizotinib) and IGF-1R inhibition (OSI-906) (Lovly et al., 2014). In the case of EGFR-mediated mechanism of acquired resistance, combination therapy with an ALK and EGFR inhibitor has been suggested to be effective (Katayama et al., 2012; Yamaguchi et al., 2014). Other plausible combinations include ALK inhibition concurrently with MEK or Src inhibitors. Heat shock protein 90 (HSP90) inhibitors have also demonstrated efficacy against ALK-sensitive and ALK-mutant NSCLCs (Katayama et al., 2012; Sang et al., 2013). HSP90 is a molecular chaperone that aids in proper folding of specific target proteins; ALK fusion proteins are substrates of HSP90. The combination of crizotinib plus HSP90 inhibitors is currently in clinical trials (NCT01712217).
Acquired resistance to BCR-ABL inhibitors in hematologic malignancies
Imatinib is an inhibitor of the BCR-ABL fusion gene, also referred to as the Philadelphia chromosome, that is characteristic of CML and presents at lower frequencies in ALL and acute myeloid leukemia (AML). BCR-ABL is a constitutively active tyrosine kinase that functions by binding ATP and transferring a phosphate group to tyrosine residues to activate downstream signaling molecules. Imatinib competitively binds to the kinase pocket of BCR-ABL, inhibiting phosphorylation and downstream signaling (Savage and Antman, 2002; Signorovitch et al., 2014; Waller, 2014). The majority of CML patients will achieve clinically significant responses to imatinib. A complete and durable cytogenetic response is achieved in up to 80% of newly diagnosed patients and approximately 60% of patients with chronic-phase CML (Druker et al., 2006; Kantarjian et al., 2002). However, up to 27% of patients have been shown to develop resistance and relapse (Press et al., 2007).
Acquired resistance to imatinib occurs as a consequence of reactivation of BCR-ABL signaling, which can be due to BCR-ABL amplification, elimination of imatinib from the cell by multidrug efflux transporters, or, most commonly, by the development of BCR-ABL mutations (Gorre et al., 2001). The acquired mutations in BCR-ABL that have been associated with resistance to imatinib and other BCR-ABL inhibitors, such as nilotinib and dasatinib, are located in 12 key positions of ABL1 (Abelson murine leukemia viral oncogene, homolog 1). These result in amino acid substitutions that change the BCR-ABL binding site, altering the ability of the PKI to bind and inhibit downstream signaling (Zabriskie et al., 2014). The most commonly observed BCR-ABL mutation conferring acquired resistance is T315I, followed by E255K/V. Interestingly, multiple acquired mutations in BCR-ABL have been identified, and the degree of imatinib (and other BCR-ABL PKI) resistance is dependent on the location of the point mutation within the BCR-ABL binding site (Shah et al., 2002). Strategies to overcome acquired resistance to BCR-ABL PKIs are currently being explored.
The second generation PKIs, such as nilotinib and dasatinib, were designed with a higher affinity for BCR-ABL in attempts to combat imatinib resistance. While they are able to overcome some mutations observed in acquired imatinib resistance, they are ineffective against the common T315I mutation (Zabriskie et al., 2014). In 2012, a third generation BCR-ABL PKI, ponatinib, was approved. Ponatinib is unique in that it is a high affinity pan-BCR-ABL inhibitor with activity in T315I-positive CML. Despite its effectiveness against the acquired T315I mutation, emergence of compound mutations in BCR-ABL have been identified as conferring differential resistance to ponatinib (Zabriskie et al., 2014). Complex screening of resistance mutations and sensitivity to BCR-ABL PKIs is necessary to optimize therapy selection at the time of disease progression.
Acquired resistance to BCR-ABL inhibitors in solid tumors
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract, characterized by positive staining for KIT (the v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene). Approximately 95% of GISTs express KIT, 80% have mutations in KIT, and 10% have mutations in the platelet-derived growth factor receptor alpha (PDGFRA) (Joensuu et al., 2013). Wild type GISTs lack KIT expression as well as KIT and PDGFRA mutations; alterations identified in wild type patients include mutations in BRAF, RAS, NF1, and succinate dehydrogenase deficiency. Imatinib is recommended first-line in GIST due to its inhibition of KIT and PDGFRA. Furthermore, KIT and PDGFRA mutations are associated with response to imatinib, with 84% of mutation-positive patients achieving a partial response compared to 0% of wild type patients (Heinrich et al., 2003). Despite high initial response rates, approximately 80% of GIST patients will develop imatinib resistance within 12–36 months (Joensuu et al., 2013).
As observed in hematologic malignancies, acquired imatinib resistance in GIST is most commonly due to secondary alterations in the drug targets. However, because the oncogenic targets differ, so do the resistance alterations. A retrospective analysis of tumors from patients who progressed on the phase 2 B2222 imatinib trial indicated that 22 of 33 (67%) patients with acquired resistance had secondary mutations in KIT or PDGFRA (Heinrich et al., 2006). Interestingly, secondary KIT mutations were only observed in patients with primary KIT mutations, and the secondary PDGFRA mutation was identified in a patient with a primary PDGFRA mutation. The acquired mutations were all located within the ATP-binding pocket of KIT (e.g., T670I, the gatekeeper mutation) or the activation loop, thus inhibiting imatinib binding and inactivation of KIT-mediated signaling.
Current treatment strategies for acquired resistance to imatinib in GIST focus on more potent inhibition of KIT and PDGFRA. Sunitinib, a multi-targeted PKI with activity against KIT, PDGFRA, VEGF, and numerous other tyrosine kinases, is currently approved second-line in patients with imatinib-resistant disease. Sunitinib has activity against all resistant genotypes, but response rates are significantly higher in wild-type GIST (p= 0.04) and patients with mutations in the ATP-binding domain (p= 0.0005) when compared to patients with alterations in the activation loop (Heinrich et al., 2008). Efficacy of the third-line treatment, regorafenib, may be due to sufficient activity in patients with acquired alterations in the activation loop of KIT (George et al., 2012). Despite their increased specificity for resistant disease, phase 3 clinical trial response rates to sunitinib and regorafenib were only 7% and 4.5%, and time to progression was 27 and 17 weeks, respectively (Demetri et al., 2013; Demetri et al., 2006). These low response rates suggest that the majority of imatinib-resistant GISTs may be entirely resistant to KIT inhibition, require more potent inhibitors, or may require alternative inhibition strategies.
Mutations and amplifications of KIT are also observed in melanomas (~3%), with higher frequencies in mucosal (39%), acral (36%), and chronically sun-damaged (28%) subtypes, suggesting possible benefit from imatinib (Curtin et al., 2006). A phase 2 trial of imatinib in patients with KIT-positive metastatic melanoma demonstrated potential efficacy, with 53.5% of the patients achieving a response and a 6-month PFS rate of 36.6% (Guo et al., 2011). Another phase 2 trial identified differential response rates between melanoma patients with KIT mutations versus KIT amplification (54% vs. 0%, respectively) (Hodi et al., 2013). Retrospective data from patients with KIT-mutated melanoma demonstrated potential efficacy of sunitinib; of 4 evaluable patients, 3 (75%) responded to sunitinib (1 complete remission and 2 partial responses) (Minor et al., 2012). Additional studies support use of sunitinib in mucosal and acral subtypes of melanoma, but failed to show an association between KIT status and response (Buchbinder et al., 2015; Mahipal et al., 2012). Despite clinical benefit and relatively high initial response rates, most patients eventually progress on these therapies.
Similar to hematologic malignancies and GIST, the current strategy to overcome acquired resistance to KIT inhibition in melanoma is the use of more potent inhibitors. Nilotinib, the BCR-ABL PKI used in chemoresistant CML, also has activity against KIT, PDGFR, DDR, and several other protein kinases, with greater potency than imatinib. Recently, a phase 2 study of niltonib demonstrated potential efficacy in patients with acquired resistance to prior KIT inhibitors. The primary endpoint, 4-month disease control, was achieved in 27% of resistant patients (95% CI 8% – 56%), and two partial responses (18.2%, CI 3% – 47%) were observed (Carvajal et al., 2015). A similar phase 2 study of nilotinib in KIT-positive melanomas in a Korean population failed to meet its primary endpoint of response rate (overall response rate was 16.7%). However, 6 of the 7 responses observed occurred in patients with KIT mutations only (24% response rate in KIT-mutated melanoma), suggesting that nilotinib may provide benefit in this subgroup (Lee et al., 2015b). As in GIST, KIT status seems to be a biomarker of response to imatinib in melanoma, but resistance inevitably develops. Current strategies to combat resistance are the same as GIST, with stronger inhibition of KIT, but response rates are low and not durable. Further understanding of the mechanisms of resistance and means to effectively suppress drivers of drug resistance are crucial to achieve durable responses in GIST and melanoma patients.
Acquired resistance to BRAF inhibition
BRAF, a serine/threonine kinase, plays a key role in the MAPK signaling pathway, which affects cell division, differentiation, and growth. Mutations in BRAF have been associated with numerous cancers, including CRC, melanoma, thyroid carcinoma, NSCLC, and non-Hodgkin’s lymphoma. Most commonly observed are somatic mutations causing activation of BRAF, specifically a valine to glutamine or lysine substation at position 600 (V600E/K), and constitutive signaling through the MAPK pathway. The discovery of BRAF mutations in cancer led to development of drugs aimed at inhibiting this oncogenic driver. In 2011, the first selective BRAF inhibitor, vemurafenib, was FDA-approved for the first-line treatment of BRAF V600E-positive melanoma after interim review of a phase 3 randomized controlled trial that demonstrated improved OS (84% vs. 64%) and PFS (5.3 months vs. 1.6 months) (Chapman et al., 2011). Debrafenib, a second BRAF inhibitor, demonstrated similar efficacy (PFS 5.1 vs. 2.7 months, HR 0.30, p<0.0001) and, was approved for the treatment of BRAF-positive melanoma in 2013 (Hauschild et al., 2012). It was realized early that, despite high initial levels of response, efficacy of BRAF inhibitors alone is not durable, with most patients developing resistance within 6–8 months (McArthur et al., 2014).
Most mechanisms of acquired resistance to BRAF inhibitors involve reactivation of the MAPK pathway; unlike EGFR and ALK, no secondary or gatekeeper-type mutations have been identified as resistance drivers (Nazarian et al., 2010). Secondary resistance may be driven upstream (e.g., upregulation and activation of the other receptor tyrosine kinases), downstream (e.g., activating MEK1/2 mutations), at the level of BRAF (e.g., alternative splicing, BRAF V600E amplification), or elevated CRAF levels. Observed genetic alterations in the setting of acquired resistance include mutations that activate NRAS, MEK1, and MEK2 (Emery et al., 2009; Nazarian et al., 2010; Van Allen et al., 2014). Amplification of mitogen-activated protein kinase kinase kinase 8 (MAP3K8 or COT) is another mechanism of resistance, which results in RAF-independent activation of MEK and ERK signaling (Johannessen et al., 2010).
One potential strategy to circumvent or delay BRAF inhibitor resistance is dual inhibition of components of the MAPK-pathway. In a phase 3 clinical trial of BRAF and MEK inhibition vs. BRAF inhibition alone in melanoma, the combination of dabrafenib plus trametinib improved PFS when compared to debrafenib alone (9.3 vs. 8.8 months, HR 0.75, p= 0.03); significant improvements in response rates and OS were also observed (Long et al., 2014). Trametinib is now FDA approved as monotherapy or in combination with dabrafenib. Interestingly, the benefit of MEK inhibitors was not observed when administered as monotherapy in patients who had progressed after initial benefit from a BRAF inhibitor, suggesting that MEK inhibition alone is not sufficient to overcome BRAF resistance (Kim et al., 2013). Preclinical data suggests another potential strategy to delay or overcome resistance caused by increased MAPK signaling is concurrent or sequential inhibition of BRAF and ERK (Hatzivassiliou et al., 2012; Herrero et al., 2015; Wong et al., 2014). However, while dual inhibition of the MAPK pathway has shown benefit, acquired resistance occurs within 12 months. Known mechanisms of acquired resistance to BRAF plus MEK inhibition are similar to those observed with BRAF inhibitor monotherapy, and include amplification of BRAF V600, which activates CRAF and subsequently MAPK signaling, and acquired MEK1/2 mutations (Moriceau et al., 2015; Villanueva et al., 2013; Wagle et al., 2014). Preclinical studies investigating even broader combination therapy consisting of concurrent BRAF, MEK, and PI3K/mTOR inhibition have demonstrated potential efficacy in BRAF/MEK-induced resistance (Carlino et al., 2014; Moriceau et al., 2015; Villanueva et al., 2013). In addition, like ALK, BRAF V600E is a client of HSP90, and preclinical studies suggest that treatment with an HSP inhibitor may be another successful strategy to overcome BRAF and MEK inhibitor resistance (Smyth et al., 2014).
MAPK-independent mechanisms of acquired resistance to BRAF inhibition have also been identified, and dual inhibition has been proposed as a strategy to overcome this resistance. The most well studied bypass track of BRAF inhibitor resistance is activation through the PI3K/Akt signaling pathway. For example, increased expression of PDGFRB or IGF-1R has been observed in cell culture and patient xenograft models of secondary resistance. Over-activation of these receptors results in activation of alternate signaling pathways (e.g., PI3K/Akt), which can reduce the cancer cells’ dependency on MAPK signaling, rendering the cells resistant to BRAF-mediated inhibition (Nazarian et al., 2010; Villanueva et al., 2010). Secondary mutations in PI3K pathway regulatory genes, such as AKT1/3, PIK3CA, PIK3CG, PIK3R2, and PTEN have also been observed, further supporting combination therapy with PI3K inhibitors (Shi et al., 2014a). Preclinical data demonstrated efficacy of concurrent PI3K and MEK inhibition in BRAF resistant cell lines, and clinical trials of this combination are currently underway (NCT01363232, NCT01337765). Recently, expression of yes-associated protein 1 (YAP1), a member of the Hippo signaling pathway, was associated with resistance to BRAF and MEK inhibition, and preclinical studies demonstrated that triple therapy with a BRAF, MEK, and YAP inhibitor may be a promising strategy to increase response in the setting of resistance (Lin et al., 2015).
Conclusions
Targeted therapies are increasingly common and recommended first-line in some cancer types due to their impressive increases in response rates and survival benefits when compared to standard cytotoxic chemotherapy. However, even in the presence of a genetic biomarker predictive of response, not all patients will benefit from such therapies (intrinsic resistance), and for those who do, durable response rates are low (acquired resistance). Therefore, equally important as identifying targetable oncogenic alterations is the identification of biomarkers of intrinsic resistance and the ability to anticipate potential mechanisms of acquired resistance that may develop.
As discussed in this article, the most common mechanisms of acquired resistance to targeted therapies induce upregulation of the drug target or bypass signaling through the same or similar pathways, resulting in cancer progression. These patterns imply an evolutionary-like process in which the cells most fit to regulate cellular proliferation and survival are selected. The question of whether or not resistant cells are present at undetectable concentrations at diagnosis or whether they develop post-treatment remains unanswered. Specifically, does resistance arise from pre-existing clones or is there drug-induced selection pressure that drives acquisition of mutations? It is likely that both may be the case.
An important concept in the context of resistance to anticancer therapies is that a biomarker conferring response or resistance in one tumor type may or may not be predictive in all tumor types. For example, approximately 10% of CRCs carry BRAF V600 mutations, but are resistant to BRAF inhibition (Corcoran et al., 2015; Kopetz et al., 2015). However, in a recent “basket” study of vemurafenib in BRAF V600-mutated non-melanoma cancers (n = 7 tumor classifications, including CRC), patients with NSCLC and Erdheim-Chester disease or Langerhans’-cell histiocytosis met the predetermined overall response rate of >35%, suggesting that BRAF status may be important in those tumor types (Hyman et al., 2015). Recently, vemurafenib also demonstrated efficacy in BRAF-positive hairy cell leukemia (Tiacci et al., 2015). Another example is RAS-status as a predictor of intrinsic resistance to EGFR monoclonal antibodies in CRC, but not predictive of response to EGFR inhibition in NSCLC. Interestingly, secondary mutations in EGFR have been reported as mechanisms of acquired resistance in both tumor types.
Lack of efficacy across tumor types may be reflective of differential oncogenic drivers or compensatory resistance mechanisms. In CRC, data suggests that BRAF inhibition is overcome through increased EGFR-mediated signaling; this bypass track is not clinically relevant in melanoma due to low basal levels of EGFR in this cancer type (Corcoran et al., 2012). Therefore, dual BRAF and EGFR inhibition may be required to increase response rates in BRAF-mutant CRC (Atreya et al., 2015; Napolitano et al., 2015). The benefit of imatinib in hematologic malignancies with BCR-ABL translocations and solid tumors with KIT or PDGFRA mutations is a unique example of how different oncogenic drivers may respond to the same therapy, but result in different genetic mechanisms of acquired resistance.
Future directions
Treatment modalities to prevent and overcome drug resistance are critical to increase the rate of durable responses to cancer therapies (Fig. 4). To date, most strategies used in clinical practice involve sequential dosing once resistance develops (Fig. 4A). An alternative strategy is to predict and target known resistance pathways from the outset using combination therapy (Fig. 4B). The strength of this approach is supported by the increased benefit of dabrafenib plus trametinib vs. dabrafenib alone in BRAF-mutated melanoma (Long et al., 2015). However, while this prolongs duration of response, resistance inevitably develops. Studies indicate development of BRAF inhibitor dependence in melanoma cells, which may also be combatted with intermittent or continuous dosing (Das Thakur et al., 2013; Dooley et al., 2014) (Fig. 4C). Another possible strategy is pulse dosing, in which the targeted therapy is administered for a short time (maybe until progression), stopped temporarily, and then restarted (Fig. 4D). This method is supported by serial biopsies of EGFR-positive NSCLC resistance which demonstrated that patients may re-develop the T790M mutation after withdrawal and, therefore, respond to reinitiation of an EGFR inhibitor (Sequist et al., 2011). Finally, some patients with advanced disease may benefit from immunotherapy, but identification of this subset is much less understood.
Figure 4. Potential treatment strategies to combat anticancer drug resistance.
(A) Sequential dosing. Begin treatment with single targeted therapy, once resistance develops, switch to drug that targets resistant cells. (B) Combination therapy. Begin therapy with a medication that targets the identified oncogenic marker in combination with a drug targeting predicted resistance mechanisms. (C) Continuous therapy. Begin treatment with a single targeted therapy, once resistance develops, add medication that targets resistance mechanism. (D) Pulse dosing. Begin treatment with single targeted therapy and periodically administer medication that targets predicted resistance mechanism. (A) represents the most commonly used strategy to date. Some data suggest that (B), (C), and/or (D) may delay development of acquired resistance.
Despite overall response rates of 50% to 80% in clinical trials of targeted anticancer therapies, mechanisms of intrinsic resistance have yet to be fully elucidated. Intrinsic resistance may be attributed to germline genetics (e.g., BIM or TPMT) or somatic alterations (e.g., KRAS in CRC). Drug metabolism and germline variants that may affect exposure to cancer therapies are also an important consideration in the context of intrinsic resistance [reviewed in (Hertz and Rae, 2015; Kathawala et al., 2015)]. However, robust examples of intrinsic resistance to targeted anti-cancer therapies are lacking. Furthermore, mechanisms of resistance to PKIs that inhibit multiple targets, such as sorafenib and sunitinib, are even more challenging to elucidate due to the heterogeneity of their effects, and are even less studied. Another interesting gap in the area of resistance to anticancer therapies involves potential racial disparities. Certain alterations are known to be more common in some races (e.g., EFGR mutations in Asian populations), suggesting potential differences in racial trends of oncogenic drivers. Because clinical and genetic studies enroll a vast majority of Caucasian patients, these potential disparities in genetic oncogenesis between races, which can greatly affect response rates, have yet to be elucidated.
Acknowledgments
Research support: The DeBartolo Family Personalized Medicine Institute, the State of Florida Cancer Research Endowed Chair, the Collins Charitable Foundation, the American Foundation for Pharmaceutical Education, Khalid Ishaq Graduate Travel Stipend, and the Eshelman Foundation.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- Allegra CJ, Rumble RB, Hamilton SR, Mangu PB, Roach N, Hantel A, Schilsky RL. Extended RAS Gene Mutation Testing in Metastatic Colorectal Carcinoma to Predict Response to Anti-Epidermal Growth Factor Receptor Monoclonal Antibody Therapy: American Society of Clinical Oncology Provisional Clinical Opinion Update 2015. J Clin Oncol. 2016;34:179–185. doi: 10.1200/JCO.2015.63.9674. [DOI] [PubMed] [Google Scholar]
- Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, Juan T, Sikorski R, Suggs S, Radinsky R, Patterson SD, Chang DD. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26:1626–1634. doi: 10.1200/JCO.2007.14.7116. [DOI] [PubMed] [Google Scholar]
- Araya CL, Cenik C, Reuter JA, Kiss G, Pande VS, Snyder MP, Greenleaf WJ. Identification of significantly mutated regions across cancer types highlights a rich landscape of functional molecular alterations. Nat Genet. 2016;48:117–125. doi: 10.1038/ng.3471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Assaraf YG, Leamon CP, Reddy JA. The folate receptor as a rational therapeutic target for personalized cancer treatment. Drug Resist Updat. 2014;17:89–95. doi: 10.1016/j.drup.2014.10.002. [DOI] [PubMed] [Google Scholar]
- Atreya CE, Van Cutsem E, Bendell JC, Andre T, Schellens JHM, Gordon MS, McRee AJ, O’Dwyer PJ, Muro K, Tabernero J, Van Geel R, Sidhu R, Greger JG, Rangwala FA, Motwani M, Wu Y, Orford KW, Corcoran RB. Updated efficacy of the MEK inhibitor trametinib (T), BRAF inhibitor dabrafenib (D), and anti-EGFR antibody panitumumab (P) in patients (pts) with BRAF V600E mutated (BRAFm) metastatic colorectal cancer (mCRC) American Society of Clinical Oncology Annual Meeting; J Clin Oncol. 2015;33 (suppl; abstr 103) [Google Scholar]
- Bauml J, Mick R, Zhang Y, Watt CD, Vachani A, Aggarwal C, Evans T, Langer C. Frequency of EGFR and KRAS mutations in patients with non small cell lung cancer by racial background: do disparities exist? Lung Cancer. 2013;81:347–353. doi: 10.1016/j.lungcan.2013.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bertotti A, Papp E, Jones S, Adleff V, Anagnostou V, Lupo B, Sausen M, Phallen J, Hruban CA, Tokheim C, Niknafs N, Nesselbush M, Lytle K, Sassi F, Cottino F, Migliardi G, Zanella ER, Ribero D, Russolillo N, Mellano A, Muratore A, Paraluppi G, Salizzoni M, Marsoni S, Kragh M, Lantto J, Cassingena A, Li QK, Karchin R, Scharpf R, Sartore-Bianchi A, Siena S, Diaz LA, Jr, Trusolino L, Velculescu VE. The genomic landscape of response to EGFR blockade in colorectal cancer. Nature. 2015;526:263–267. doi: 10.1038/nature14969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhatia S, Landier W, Hageman L, Chen Y, Kim H, Sun CL, Kornegay N, Evans WE, Angiolillo AL, Bostrom B, Casillas J, Lew G, Maloney KW, Mascarenhas L, Ritchey AK, Termuhlen AM, Carroll WL, Wong FL, Relling MV. Systemic Exposure to Thiopurines and Risk of Relapse in Children With Acute Lymphoblastic Leukemia: A Children’s Oncology Group Study. JAMA Oncol. 2015;1:287–295. doi: 10.1001/jamaoncol.2015.0245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Browning ET, Weickhardt AJ, Camidge DR. Response to crizotinib rechallenge after initial progression and intervening chemotherapy in ALK lung cancer. J Thorac Oncol. 2013;8:e21. doi: 10.1097/JTO.0b013e31827a892c. [DOI] [PubMed] [Google Scholar]
- Buchbinder EI, Sosman JA, Lawrence DP, McDermott DF, Ramaiya NH, Van den Abbeele AD, Linette GP, Giobbie-Hurder A, Hodi FS. Phase 2 study of sunitinib in patients with metastatic mucosal or acral melanoma. Cancer. 2015;121:4007–4015. doi: 10.1002/cncr.29622. [DOI] [PubMed] [Google Scholar]
- Budha NR, Frymoyer A, Smelick GS, Jin JY, Yago MR, Dresser MJ, Holden SN, Benet LZ, Ware JA. Drug absorption interactions between oral targeted anticancer agents and PPIs: is pH-dependent solubility the Achilles heel of targeted therapy? Clin Pharmacol Ther. 2012;92:203–213. doi: 10.1038/clpt.2012.73. [DOI] [PubMed] [Google Scholar]
- Camidge DR, Bang YJ, Kwak EL, Iafrate AJ, Varella-Garcia M, Fox SB, Riely GJ, Solomon B, Ou SH, Kim DW, Salgia R, Fidias P, Engelman JA, Gandhi L, Janne PA, Costa DB, Shapiro GI, Lorusso P, Ruffner K, Stephenson P, Tang Y, Wilner K, Clark JW, Shaw AT. Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol. 2012;13:1011–1019. doi: 10.1016/S1470-2045(12)70344-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Camidge DR, Pao W, Sequist LV. Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nature reviews. Clinical oncology. 2014;11:473–481. doi: 10.1038/nrclinonc.2014.104. [DOI] [PubMed] [Google Scholar]
- Carlino MS, Todd JR, Gowrishankar K, Mijatov B, Pupo GM, Fung C, Snoyman S, Hersey P, Long GV, Kefford RF, Rizos H. Differential activity of MEK and ERK inhibitors in BRAF inhibitor resistant melanoma. Mol Oncol. 2014;8:544–554. doi: 10.1016/j.molonc.2014.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carvajal RD, Lawrence DP, Weber JS, Gajewski TF, Gonzalez R, Lutzky J, O’Day SJ, Hamid O, Wolchok JD, Chapman PB, Sullivan RJ, Teitcher JB, Ramaiya N, Giobbie-Hurder A, Antonescu CR, Heinrich MC, Bastian BC, Corless CL, Fletcher JA, Hodi FS. Phase II Study of Nilotinib in Melanoma Harboring KIT Alterations Following Progression to Prior KIT Inhibition. Clin Cancer Res. 2015;21:2289–2296. doi: 10.1158/1078-0432.CCR-14-1630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caumont C, Veillon R, Gros A, Laharanne E, Begueret H, Merlio JP. Neuroendocrine phenotype as an acquired resistance mechanism in ALK-rearranged lung adenocarcinoma. Lung Cancer. 2016;92:15–18. doi: 10.1016/j.lungcan.2015.12.001. [DOI] [PubMed] [Google Scholar]
- CenterWatch, Drug Information. 2016. [Google Scholar]
- Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, Hogg D, Lorigan P, Lebbe C, Jouary T, Schadendorf D, Ribas A, O’Day SJ, Sosman JA, Kirkwood JM, Eggermont AM, Dreno B, Nolop K, Li J, Nelson B, Hou J, Lee RJ, Flaherty KT, McArthur GA, Group B-S. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516. doi: 10.1056/NEJMoa1103782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X, Liu H, Xing H, Sun H, Zhu P. The BIM deletion polymorphism cannot account for intrinsic TKI resistance of Chinese individuals with chronic myeloid leukemia. Nat Med. 2014;20:1090. doi: 10.1038/nm.3652. [DOI] [PubMed] [Google Scholar]
- Cheng EH, Sawyers CL. In cancer drug resistance, germline matters too. Nat Med. 2012;18:494–496. doi: 10.1038/nm.2725. [DOI] [PubMed] [Google Scholar]
- Choi YL, Soda M, Yamashita Y, Ueno T, Takashima J, Nakajima T, Yatabe Y, Takeuchi K, Hamada T, Haruta H, Ishikawa Y, Kimura H, Mitsudomi T, Tanio Y, Mano H, Group ALKLCS. EML4-ALK mutations in lung cancer that confer resistance to ALK inhibitors. N Engl J Med. 2010;363:1734–1739. doi: 10.1056/NEJMoa1007478. [DOI] [PubMed] [Google Scholar]
- Choi YL, Takeuchi K, Soda M, Inamura K, Togashi Y, Hatano S, Enomoto M, Hamada T, Haruta H, Watanabe H, Kurashina K, Hatanaka H, Ueno T, Takada S, Yamashita Y, Sugiyama Y, Ishikawa Y, Mano H. Identification of novel isoforms of the EML4-ALK transforming gene in non-small cell lung cancer. Cancer Res. 2008;68:4971–4976. doi: 10.1158/0008-5472.CAN-07-6158. [DOI] [PubMed] [Google Scholar]
- Christensen JG, Zou HY, Arango ME, Li Q, Lee JH, McDonnell SR, Yamazaki S, Alton GR, Mroczkowski B, Los G. Cytoreductive antitumor activity of PF-2341066, a novel inhibitor of anaplastic lymphoma kinase and c-Met, in experimental models of anaplastic large-cell lymphoma. Mol Cancer Ther. 2007;6:3314–3322. doi: 10.1158/1535-7163.MCT-07-0365. [DOI] [PubMed] [Google Scholar]
- Corcoran RB, Atreya CE, Falchook GS, Kwak EL, Ryan DP, Bendell JC, Hamid O, Messersmith WA, Daud A, Kurzrock R, Pierobon M, Sun P, Cunningham E, Little S, Orford K, Motwani M, Bai Y, Patel K, Venook AP, Kopetz S. Combined BRAF and MEK Inhibition With Dabrafenib and Trametinib in BRAF V600-Mutant Colorectal Cancer. J Clin Oncol. 2015 doi: 10.1200/JCO.2015.63.2471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corcoran RB, Ebi H, Turke AB, Coffee EM, Nishino M, Cogdill AP, Brown RD, Della Pelle P, Dias-Santagata D, Hung KE, Flaherty KT, Piris A, Wargo JA, Settleman J, Mino-Kenudson M, Engelman JA. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2012;2:227–235. doi: 10.1158/2159-8290.CD-11-0341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol. 2006;24:4340–4346. doi: 10.1200/JCO.2006.06.2984. [DOI] [PubMed] [Google Scholar]
- Das Thakur M, Salangsang F, Landman AS, Sellers WR, Pryer NK, Levesque MP, Dummer R, McMahon M, Stuart DD. Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. Nature. 2013;494:251–255. doi: 10.1038/nature11814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Demetri GD, Reichardt P, Kang YK, Blay JY, Rutkowski P, Gelderblom H, Hohenberger P, Leahy M, von Mehren M, Joensuu H, Badalamenti G, Blackstein M, Le Cesne A, Schoffski P, Maki RG, Bauer S, Nguyen BB, Xu J, Nishida T, Chung J, Kappeler C, Kuss I, Laurent D, Casali PG G.s. investigators. Efficacy and safety of regorafenib for advanced gastrointestinal stromal tumours after failure of imatinib and sunitinib (GRID): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 2013;381:295–302. doi: 10.1016/S0140-6736(12)61857-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, Verweij J, McArthur G, Judson IR, Heinrich MC, Morgan JA, Desai J, Fletcher CD, George S, Bello CL, Huang X, Baum CM, Casali PG. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet. 2006;368:1329–1338. doi: 10.1016/S0140-6736(06)69446-4. [DOI] [PubMed] [Google Scholar]
- Dooley AJ, Gupta A, Bhattacharyya M, Middleton MR. Intermittent dosing with vemurafenib in BRAF V600E–mutant melanoma: review of a case series. Ther Adv Med Oncol. 2014;6:262–266. doi: 10.1177/1758834014548187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, Rivera F, Kocakova I, Ruff P, Blasinska-Morawiec M, Smakal M, Canon JL, Rother M, Williams R, Rong A, Wiezorek J, Sidhu R, Patterson SD. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369:1023–1034. doi: 10.1056/NEJMoa1305275. [DOI] [PubMed] [Google Scholar]
- Druker BJ, Guilhot F, O’Brien SG, Gathmann I, Kantarjian H, Gattermann N, Deininger MW, Silver RT, Goldman JM, Stone RM, Cervantes F, Hochhaus A, Powell BL, Gabrilove JL, Rousselot P, Reiffers J, Cornelissen JJ, Hughes T, Agis H, Fischer T, Verhoef G, Shepherd J, Saglio G, Gratwohl A, Nielsen JL, Radich JP, Simonsson B, Taylor K, Baccarani M, So C, Letvak L, Larson RA, Investigators I. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 2006;355:2408–2417. doi: 10.1056/NEJMoa062867. [DOI] [PubMed] [Google Scholar]
- Eberlein CA, Stetson D, Markovets AA, Al-Kadhimi KJ, Lai Z, Fisher PR, Meador CB, Spitzler P, Ichihara E, Ross SJ, Ahdesmaki MJ, Ahmed A, Ratcliffe LE, O’Brien EL, Barnes CH, Brown H, Smith PD, Dry JR, Beran G, Thress KS, Dougherty B, Pao W, Cross DA. Acquired Resistance to the Mutant-Selective EGFR Inhibitor AZD9291 Is Associated with Increased Dependence on RAS Signaling in Preclinical Models. Cancer Res. 2015;75:2489–2500. doi: 10.1158/0008-5472.CAN-14-3167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Emery CM, Vijayendran KG, Zipser MC, Sawyer AM, Niu L, Kim JJ, Hatton C, Chopra R, Oberholzer PA, Karpova MB, MacConaill LE, Zhang J, Gray NS, Sellers WR, Dummer R, Garraway LA. MEK1 mutations confer resistance to MEK and B-RAF inhibition. Proc Natl Acad Sci U S A. 2009;106:20411–20416. doi: 10.1073/pnas.0905833106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, Kosaka T, Holmes AJ, Rogers AM, Cappuzzo F, Mok T, Lee C, Johnson BE, Cantley LC, Janne PA. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–1043. doi: 10.1126/science.1141478. [DOI] [PubMed] [Google Scholar]
- Fletcher JI, Williams RT, Henderson MJ, Norris MD, Haber M. ABC transporters as mediators of drug resistance and contributors to cancer cell biology. Drug Resist Updat. 2016;26:1–9. doi: 10.1016/j.drup.2016.03.001. [DOI] [PubMed] [Google Scholar]
- Friboulet L, Li N, Katayama R, Lee CC, Gainor JF, Crystal AS, Michellys PY, Awad MM, Yanagitani N, Kim S, Pferdekamper AC, Li J, Kasibhatla S, Sun F, Sun X, Hua S, McNamara P, Mahmood S, Lockerman EL, Fujita N, Nishio M, Harris JL, Shaw AT, Engelman JA. The ALK inhibitor ceritinib overcomes crizotinib resistance in non-small cell lung cancer. Cancer Discov. 2014;4:662–673. doi: 10.1158/2159-8290.CD-13-0846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gainor JF, Varghese AM, Ou SH, Kabraji S, Awad MM, Katayama R, Pawlak A, Mino-Kenudson M, Yeap BY, Riely GJ, Iafrate AJ, Arcila ME, Ladanyi M, Engelman JA, Dias-Santagata D, Shaw AT. ALK rearrangements are mutually exclusive with mutations in EGFR or KRAS: an analysis of 1,683 patients with non-small cell lung cancer. Clin Cancer Res. 2013;19:4273–4281. doi: 10.1158/1078-0432.CCR-13-0318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- George S, Wang Q, Heinrich MC, Corless CL, Zhu M, Butrynski JE, Morgan JA, Wagner AJ, Choy E, Tap WD, Yap JT, Van den Abbeele AD, Manola JB, Solomon SM, Fletcher JA, von Mehren M, Demetri GD. Efficacy and safety of regorafenib in patients with metastatic and/or unresectable GI stromal tumor after failure of imatinib and sunitinib: a multicenter phase II trial. J Clin Oncol. 2012;30:2401–2407. doi: 10.1200/JCO.2011.39.9394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gillis NK, Innocenti F. Evidence required to demonstrate clinical utility of pharmacogenetic testing: the debate continues. Clin Pharmacol Ther. 2014;96:655–657. doi: 10.1038/clpt.2014.185. [DOI] [PubMed] [Google Scholar]
- Goler-Baron V, Assaraf YG. Structure and function of ABCG2-rich extracellular vesicles mediating multidrug resistance. PLoS One. 2011;6:e16007. doi: 10.1371/journal.pone.0016007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong Y, Somwar R, Politi K, Balak M, Chmielecki J, Jiang X, Pao W. Induction of BIM is essential for apoptosis triggered by EGFR kinase inhibitors in mutant EGFR-dependent lung adenocarcinomas. PLoS Med. 2007;4:e294. doi: 10.1371/journal.pmed.0040294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, Rao PN, Sawyers CL. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science. 2001;293:876–880. doi: 10.1126/science.1062538. [DOI] [PubMed] [Google Scholar]
- Gridelli C, Peters S, Sgambato A, Casaluce F, Adjei AA, Ciardiello F. ALK inhibitors in the treatment of advanced NSCLC. Cancer Treat Rev. 2014;40:300–306. doi: 10.1016/j.ctrv.2013.07.002. [DOI] [PubMed] [Google Scholar]
- Guo J, Si L, Kong Y, Flaherty KT, Xu X, Zhu Y, Corless CL, Li L, Li H, Sheng X, Cui C, Chi Z, Li S, Han M, Mao L, Lin X, Du N, Zhang X, Li J, Wang B, Qin S. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J Clin Oncol. 2011;29:2904–2909. doi: 10.1200/JCO.2010.33.9275. [DOI] [PubMed] [Google Scholar]
- Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- Hatzivassiliou G, Liu B, O’Brien C, Spoerke JM, Hoeflich KP, Haverty PM, Soriano R, Forrest WF, Heldens S, Chen H, Toy K, Ha C, Zhou W, Song K, Friedman LS, Amler LC, Hampton GM, Moffat J, Belvin M, Lackner MR. ERK inhibition overcomes acquired resistance to MEK inhibitors. Mol Cancer Ther. 2012;11:1143–1154. doi: 10.1158/1535-7163.MCT-11-1010. [DOI] [PubMed] [Google Scholar]
- Hauschild A, Grob JJ, Demidov LV, Jouary T, Gutzmer R, Millward M, Rutkowski P, Blank CU, Miller WH, Jr, Kaempgen E, Martin-Algarra S, Karaszewska B, Mauch C, Chiarion-Sileni V, Martin AM, Swann S, Haney P, Mirakhur B, Guckert ME, Goodman V, Chapman PB. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380:358–365. doi: 10.1016/S0140-6736(12)60868-X. [DOI] [PubMed] [Google Scholar]
- Heinrich MC, Corless CL, Blanke CD, Demetri GD, Joensuu H, Roberts PJ, Eisenberg BL, von Mehren M, Fletcher CD, Sandau K, McDougall K, Ou WB, Chen CJ, Fletcher JA. Molecular correlates of imatinib resistance in gastrointestinal stromal tumors. J Clin Oncol. 2006;24:4764–4774. doi: 10.1200/JCO.2006.06.2265. [DOI] [PubMed] [Google Scholar]
- Heinrich MC, Corless CL, Demetri GD, Blanke CD, von Mehren M, Joensuu H, McGreevey LS, Chen CJ, Van den Abbeele AD, Druker BJ, Kiese B, Eisenberg B, Roberts PJ, Singer S, Fletcher CD, Silberman S, Dimitrijevic S, Fletcher JA. Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol. 2003;21:4342–4349. doi: 10.1200/JCO.2003.04.190. [DOI] [PubMed] [Google Scholar]
- Heinrich MC, Maki RG, Corless CL, Antonescu CR, Harlow A, Griffith D, Town A, McKinley A, Ou WB, Fletcher JA, Fletcher CD, Huang X, Cohen DP, Baum CM, Demetri GD. Primary and secondary kinase genotypes correlate with the biological and clinical activity of sunitinib in imatinib-resistant gastrointestinal stromal tumor. J Clin Oncol. 2008;26:5352–5359. doi: 10.1200/JCO.2007.15.7461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herrero A, Pinto A, Colon-Bolea P, Casar B, Jones M, Agudo-Ibanez L, Vidal R, Tenbaum SP, Nuciforo P, Valdizan EM, Horvath Z, Orfi L, Pineda-Lucena A, Bony E, Keri G, Rivas G, Pazos A, Gozalbes R, Palmer HG, Hurlstone A, Crespo P. Small Molecule Inhibition of ERK Dimerization Prevents Tumorigenesis by RAS-ERK Pathway Oncogenes. Cancer Cell. 2015;28:170–182. doi: 10.1016/j.ccell.2015.07.001. [DOI] [PubMed] [Google Scholar]
- Hertz DL, Rae J. Pharmacogenetics of cancer drugs. Annu Rev Med. 2015;66:65–81. doi: 10.1146/annurev-med-053013-053944. [DOI] [PubMed] [Google Scholar]
- Heuckmann JM, Holzel M, Sos ML, Heynck S, Balke-Want H, Koker M, Peifer M, Weiss J, Lovly CM, Grutter C, Rauh D, Pao W, Thomas RK. ALK mutations conferring differential resistance to structurally diverse ALK inhibitors. Clin Cancer Res. 2011;17:7394–7401. doi: 10.1158/1078-0432.CCR-11-1648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hodi FS, Corless CL, Giobbie-Hurder A, Fletcher JA, Zhu M, Marino-Enriquez A, Friedlander P, Gonzalez R, Weber JS, Gajewski TF, O’Day SJ, Kim KB, Lawrence D, Flaherty KT, Luke JJ, Collichio FA, Ernstoff MS, Heinrich MC, Beadling C, Zukotynski KA, Yap JT, Van den Abbeele AD, Demetri GD, Fisher DE. Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. Clin Oncol. 2013;31:3182–3190. doi: 10.1200/JCO.2012.47.7836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hyman DM, Puzanov I, Subbiah V, Faris JE, Chau I, Blay JY, Wolf J, Raje NS, Diamond EL, Hollebecque A, Gervais R, Elez-Fernandez ME, Italiano A, Hofheinz RD, Hidalgo M, Chan E, Schuler M, Lasserre SF, Makrutzki M, Sirzen F, Veronese ML, Tabernero J, Baselga J. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N Engl J Med. 2015;373:726–736. doi: 10.1056/NEJMoa1502309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Isobe K, Hata Y, Tochigi N, Kaburaki K, Kobayashi H, Makino T, Otsuka H, Sato F, Ishida F, Kikuchi N, Hirota N, Sato K, Sano G, Sugino K, Sakamoto S, Takai Y, Shibuya K, Iyoda A, Homma S. Clinical significance of BIM deletion polymorphism in non-small-cell lung cancer with epidermal growth factor receptor mutation. J Thorac Oncol. 2014;9:483–487. doi: 10.1097/JTO.0000000000000125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Isozaki H, Takigawa N, Kiura K. Mechanisms of Acquired Resistance to ALK Inhibitors and the Rationale for Treating ALK-positive Lung Cancer. Cancers (Basel) 2015;7:763–783. doi: 10.3390/cancers7020763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Janjigian YY, Smit EF, Groen HJ, Horn L, Gettinger S, Camidge DR, Riely GJ, Wang B, Fu Y, Chand VK, Miller VA, Pao W. Dual inhibition of EGFR with afatinib and cetuximab in kinase inhibitor-resistant EGFR-mutant lung cancer with and without T790M mutations. Cancer Discov. 2014;4:1036–1045. doi: 10.1158/2159-8290.CD-14-0326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Janne PA, Yang JC, Kim DW, Planchard D, Ohe Y, Ramalingam SS, Ahn MJ, Kim SW, Su WC, Horn L, Haggstrom D, Felip E, Kim JH, Frewer P, Cantarini M, Brown KH, Dickinson PA, Ghiorghiu S, Ranson M. AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer. N Engl J Med. 2015;372:1689–1699. doi: 10.1056/NEJMoa1411817. [DOI] [PubMed] [Google Scholar]
- Joensuu H, Hohenberger P, Corless CL. Gastrointestinal stromal tumour. Lancet. 2013;382:973–983. doi: 10.1016/S0140-6736(13)60106-3. [DOI] [PubMed] [Google Scholar]
- Johannessen CM, Boehm JS, Kim SY, Thomas SR, Wardwell L, Johnson LA, Emery CM, Stransky N, Cogdill AP, Barretina J, Caponigro G, Hieronymus H, Murray RR, Salehi-Ashtiani K, Hill DE, Vidal M, Zhao JJ, Yang X, Alkan O, Kim S, Harris JL, Wilson CJ, Myer VE, Finan PM, Root DE, Roberts TM, Golub T, Flaherty KT, Dummer R, Weber BL, Sellers WR, Schlegel R, Wargo JA, Hahn WC, Garraway LA. COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. Nature. 2010;468:968–972. doi: 10.1038/nature09627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jonker DJ, O’Callaghan CJ, Karapetis CS, Zalcberg JR, Tu D, Au HJ, Berry SR, Krahn M, Price T, Simes RJ, Tebbutt NC, van Hazel G, Wierzbicki R, Langer C, Moore MJ. Cetuximab for the treatment of colorectal cancer. N Engl J Med. 2007;357:2040–2048. doi: 10.1056/NEJMoa071834. [DOI] [PubMed] [Google Scholar]
- Kantarjian H, Sawyers C, Hochhaus A, Guilhot F, Schiffer C, Gambacorti-Passerini C, Niederwieser D, Resta D, Capdeville R, Zoellner U, Talpaz M, Druker B, Goldman J, O’Brien SG, Russell N, Fischer T, Ottmann O, Cony-Makhoul P, Facon T, Stone R, Miller C, Tallman M, Brown R, Schuster M, Loughran T, Gratwohl A, Mandelli F, Saglio G, Lazzarino M, Russo D, Baccarani M, Morra E, International STICMLSG. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N Engl J Med. 2002;346:645–652. doi: 10.1056/NEJMoa011573. [DOI] [PubMed] [Google Scholar]
- Katayama R, Shaw AT, Khan TM, Mino-Kenudson M, Solomon BJ, Halmos B, Jessop NA, Wain JC, Yeo AT, Benes C, Drew L, Saeh JC, Crosby K, Sequist LV, Iafrate AJ, Engelman JA. Mechanisms of acquired crizotinib resistance in ALK-rearranged lung Cancers. Sci Transl Med. 2012;4:120ra117. doi: 10.1126/scitranslmed.3003316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kathawala RJ, Gupta P, Ashby CR, Jr, Chen ZS. The modulation of ABC transporter-mediated multidrug resistance in cancer: a review of the past decade. Drug Resist Updat. 2015;18:1–17. doi: 10.1016/j.drup.2014.11.002. [DOI] [PubMed] [Google Scholar]
- Kim KB, Kefford R, Pavlick AC, Infante JR, Ribas A, Sosman JA, Fecher LA, Millward M, McArthur GA, Hwu P, Gonzalez R, Ott PA, Long GV, Gardner OS, Ouellet D, Xu Y, DeMarini DJ, Le NT, Patel K, Lewis KD. Phase II study of the MEK1/MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. J Clin Oncol. 2013;31:482–489. doi: 10.1200/JCO.2012.43.5966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kodityal S, Elvin JA, Squillace R, Agarwal N, Miller VA, Ali SM, Klempner SJ, Ou SH. A novel acquired ALK F1245C mutation confers resistance to crizotinib in ALK-positive NSCLC but is sensitive to ceritinib. Lung Cancer. 2016;92:19–21. doi: 10.1016/j.lungcan.2015.11.023. [DOI] [PubMed] [Google Scholar]
- Kopetz S, Desai J, Chan E, Hecht JR, O’Dwyer PJ, Maru D, Morris V, Janku F, Dasari A, Chung W, Issa JJ, Gibbs P, James B, Powis G, Nolop KB, Bhattacharya S, Saltz L. Phase II Pilot Study of Vemurafenib in Patients With Metastatic BRAF-Mutated Colorectal Cancer. J Clin Oncol. 2015 doi: 10.1200/JCO.2015.63.2497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JH, Lin YL, Hsu WH, Chen HY, Chang YC, Yu CJ, Shih JY, Lin CC, Chen KY, Ho CC, Laio WY, Yang PC, Yang JC. Bcl-2-like protein 11 deletion polymorphism predicts survival in advanced non-small-cell lung cancer. J Thorac Oncol. 2014;9:1385–1392. doi: 10.1097/JTO.0000000000000238. [DOI] [PubMed] [Google Scholar]
- Lee JK, Shin JY, Kim S, Lee S, Park C, Kim JY, Koh Y, Keam B, Min HS, Kim TM, Jeon YK, Kim DW, Chung DH, Heo DS, Lee SH, Kim JI. Primary resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) in patients with non-small-cell lung cancer harboring TKI-sensitive EGFR mutations: an exploratory study. Ann Oncol. 2013;24:2080–2087. doi: 10.1093/annonc/mdt127. [DOI] [PubMed] [Google Scholar]
- Lee JY, Ku BM, Lim SH, Lee MY, Kim H, Kim M, Kim S, Jung HA, Sun JM, Ahn JS, Park K, Ahn MJ. The BIM Deletion Polymorphism and its Clinical Implication in Patients with EGFR-Mutant Non-Small-Cell Lung Cancer Treated with EGFR Tyrosine Kinase Inhibitors. J Thorac Oncol. 2015a;10:903–909. doi: 10.1097/JTO.0000000000000535. [DOI] [PubMed] [Google Scholar]
- Lee SJ, Kim TM, Kim YJ, Jang KT, Lee HJ, Lee SN, Ahn MS, Hwang IG, Lee S, Lee MH, Lee J. Phase II Trial of Nilotinib in Patients With Metastatic Malignant Melanoma Harboring KIT Gene Aberration: A Multicenter Trial of Korean Cancer Study Group (UN10-06) Oncologist. 2015b;20:1312–1319. doi: 10.1634/theoncologist.2015-0161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lennard L, Welch JC, Lilleyman JS. Thiopurine drugs in the treatment of childhood leukaemia: the influence of inherited thiopurine methyltransferase activity on drug metabolism and cytotoxicity. Br J Clin Pharmacol. 1997;44:455–461. doi: 10.1046/j.1365-2125.1997.t01-1-00607.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levinsen M, Rotevatn EO, Rosthoj S, Nersting J, Abrahamsson J, Appell ML, Bergan S, Bechensteen AG, Harila-Saari A, Heyman M, Jonsson OG, Maxild JB, Niemi M, Soderhall S, Schmiegelow K O. Nordic Society of Paediatric Haematology. Pharmacogenetically based dosing of thiopurines in childhood acute lymphoblastic leukemia: influence on cure rates and risk of second cancer. Pediatr Blood Cancer. 2014;61:797–802. doi: 10.1002/pbc.24921. [DOI] [PubMed] [Google Scholar]
- Lievre A, Bachet JB, Le Corre D, Boige V, Landi B, Emile JF, Cote JF, Tomasic G, Penna C, Ducreux M, Rougier P, Penault-Llorca F, Laurent-Puig P. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 2006;66:3992–3995. doi: 10.1158/0008-5472.CAN-06-0191. [DOI] [PubMed] [Google Scholar]
- Lin L, Sabnis AJ, Chan E, Olivas V, Cade L, Pazarentzos E, Asthana S, Neel D, Yan JJ, Lu X, Pham L, Wang MM, Karachaliou N, Cao MG, Manzano JL, Ramirez JL, Torres JM, Buttitta F, Rudin CM, Collisson EA, Algazi A, Robinson E, Osman I, Munoz-Couselo E, Cortes J, Frederick DT, Cooper ZA, McMahon M, Marchetti A, Rosell R, Flaherty KT, Wargo JA, Bivona TG. The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat Genet. 2015;47:250–256. doi: 10.1038/ng.3218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Li Q, Zhou L, Xie N, Nice EC, Zhang H, Huang C, Lei Y. Cancer drug resistance: redox resetting renders a way. Oncotarget. 2016 doi: 10.18632/oncotarget.8600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Livney YD, Assaraf YG. Rationally designed nanovehicles to overcome cancer chemoresistance. Adv Drug Deliv Rev. 2013;65:1716–1730. doi: 10.1016/j.addr.2013.08.006. [DOI] [PubMed] [Google Scholar]
- Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, Garbe C, Jouary T, Hauschild A, Grob JJ, Chiarion-Sileni V, Lebbe C, Mandala M, Millward M, Arance A, Bondarenko I, Haanen JB, Hansson J, Utikal J, Ferraresi V, Kovalenko N, Mohr P, Probachai V, Schadendorf D, Nathan P, Robert C, Ribas A, DeMarini DJ, Irani JG, Swann S, Legos JJ, Jin F, Mookerjee B, Flaherty K. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial. Lancet. 2015;386:444–451. doi: 10.1016/S0140-6736(15)60898-4. [DOI] [PubMed] [Google Scholar]
- Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, Garbe C, Jouary T, Hauschild A, Grob JJ, Chiarion Sileni V, Lebbe C, Mandala M, Millward M, Arance A, Bondarenko I, Haanen JB, Hansson J, Utikal J, Ferraresi V, Kovalenko N, Mohr P, Probachai V, Schadendorf D, Nathan P, Robert C, Ribas A, DeMarini DJ, Irani JG, Casey M, Ouellet D, Martin AM, Le N, Patel K, Flaherty K. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J Med. 2014;371:1877–1888. doi: 10.1056/NEJMoa1406037. [DOI] [PubMed] [Google Scholar]
- Lovly CM, McDonald NT, Chen H, Ortiz-Cuaran S, Heukamp LC, Yan Y, Florin A, Ozretic L, Lim D, Wang L, Chen Z, Chen X, Lu P, Paik PK, Shen R, Jin H, Buettner R, Ansen S, Perner S, Brockmann M, Bos M, Wolf J, Gardizi M, Wright GM, Solomon B, Russell PA, Rogers TM, Suehara Y, Red-Brewer M, Tieu R, de Stanchina E, Wang Q, Zhao Z, Johnson DH, Horn L, Wong KK, Thomas RK, Ladanyi M, Pao W. Rationale for co-targeting IGF-1R and ALK in ALK fusion-positive lung cancer. Nat Med. 2014;20:1027–1034. doi: 10.1038/nm.3667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahipal A, Tijani L, Chan K, Laudadio M, Mastrangelo MJ, Sato T. A pilot study of sunitinib malate in patients with metastatic uveal melanoma. Melanoma Res. 2012;22:440–446. doi: 10.1097/CMR.0b013e328358b373. [DOI] [PubMed] [Google Scholar]
- McArthur GA, Chapman PB, Robert C, Larkin J, Haanen JB, Dummer R, Ribas A, Hogg D, Hamid O, Ascierto PA, Garbe C, Testori A, Maio M, Lorigan P, Lebbe C, Jouary T, Schadendorf D, O’Day SJ, Kirkwood JM, Eggermont AM, Dreno B, Sosman JA, Flaherty KT, Yin M, Caro I, Cheng S, Trunzer K, Hauschild A. Safety and efficacy of vemurafenib in BRAF(V600E) and BRAF(V600K) mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol. 2014;15:323–332. doi: 10.1016/S1470-2045(14)70012-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller VA, Hirsh V, Cadranel J, Chen YM, Park K, Kim SW, Zhou C, Su WC, Wang M, Sun Y, Heo DS, Crino L, Tan EH, Chao TY, Shahidi M, Cong XJ, Lorence RM, Yang JC. Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trial. Lancet Oncol. 2012;13:528–538. doi: 10.1016/S1470-2045(12)70087-6. [DOI] [PubMed] [Google Scholar]
- Minor DR, Kashani-Sabet M, Garrido M, O’Day SJ, Hamid O, Bastian BC. Sunitinib therapy for melanoma patients with KIT mutations. Clin Cancer Res. 2012;18:1457–1463. doi: 10.1158/1078-0432.CCR-11-1987. [DOI] [PubMed] [Google Scholar]
- Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, Seto T, Satouchi M, Tada H, Hirashima T, Asami K, Katakami N, Takada M, Yoshioka H, Shibata K, Kudoh S, Shimizu E, Saito H, Toyooka S, Nakagawa K, Fukuoka M, West Japan Oncology G. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010;11:121–128. doi: 10.1016/S1470-2045(09)70364-X. [DOI] [PubMed] [Google Scholar]
- Morgensztern D, Politi K, Herbst RS. EGFR Mutations in Non-Small-Cell Lung Cancer: Find, Divide, and Conquer. JAMA Oncol. 2015;1:146–148. doi: 10.1001/jamaoncol.2014.278. [DOI] [PubMed] [Google Scholar]
- Moriceau G, Hugo W, Hong A, Shi H, Kong X, Yu CC, Koya RC, Samatar AA, Khanlou N, Braun J, Ruchalski K, Seifert H, Larkin J, Dahlman KB, Johnson DB, Algazi A, Sosman JA, Ribas A, Lo RS. Tunable-combinatorial mechanisms of acquired resistance limit the efficacy of BRAF/MEK cotargeting but result in melanoma drug addiction. Cancer Cell. 2015;27:240–256. doi: 10.1016/j.ccell.2014.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakagawa T, Takeuchi S, Yamada T, Ebi H, Sano T, Nanjo S, Ishikawa D, Sato M, Hasegawa Y, Sekido Y, Yano S. EGFR-TKI resistance due to BIM polymorphism can be circumvented in combination with HDAC inhibition. Cancer Res. 2013;73:2428–2434. doi: 10.1158/0008-5472.CAN-12-3479. [DOI] [PubMed] [Google Scholar]
- Napolitano S, Martini G, Rinaldi B, Martinelli E, Donniacuo M, Berrino L, Vitagliano D, Morgillo F, Barra G, De Palma R, Merolla F, Ciardiello F, Troiani T. Primary and Acquired Resistance of Colorectal Cancer to Anti-EGFR Monoclonal Antibody Can Be Overcome by Combined Treatment of Regorafenib with Cetuximab. Clin Cancer Res. 2015;21:2975–2983. doi: 10.1158/1078-0432.CCR-15-0020. [DOI] [PubMed] [Google Scholar]
- Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, Chen Z, Lee MK, Attar N, Sazegar H, Chodon T, Nelson SF, McArthur G, Sosman JA, Ribas A, Lo RS. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature. 2010;468:973–977. doi: 10.1038/nature09626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ng KP, Hillmer AM, Chuah CT, Juan WC, Ko TK, Teo AS, Ariyaratne PN, Takahashi N, Sawada K, Fei Y, Soh S, Lee WH, Huang JW, Allen JC, Jr, Woo XY, Nagarajan N, Kumar V, Thalamuthu A, Poh WT, Ang AL, Mya HT, How GF, Yang LY, Koh LP, Chowbay B, Chang CT, Nadarajan VS, Chng WJ, Than H, Lim LC, Goh YT, Zhang S, Poh D, Tan P, Seet JE, Ang MK, Chau NM, Ng QS, Tan DS, Soda M, Isobe K, Nothen MM, Wong TY, Shahab A, Ruan X, Cacheux-Rataboul V, Sung WK, Tan EH, Yatabe Y, Mano H, Soo RA, Chin TM, Lim WT, Ruan Y, Ong ST. A common BIM deletion polymorphism mediates intrinsic resistance and inferior responses to tyrosine kinase inhibitors in cancer. Nat Med. 2012;18:521–528. doi: 10.1038/nm.2713. [DOI] [PubMed] [Google Scholar]
- Nie W, Tao X, Wei H, Chen WS, Li B. The BIM deletion polymorphism is a prognostic biomarker of EGFR-TKIs response in NSCLC: A systematic review and meta-analysis. Oncotarget. 2015;6:25696–25700. doi: 10.18632/oncotarget.4678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niewerth D, Jansen G, Assaraf YG, Zweegman S, Kaspers GJ, Cloos J. Molecular basis of resistance to proteasome inhibitors in hematological malignancies. Drug Resist Updat. 2015;18:18–35. doi: 10.1016/j.drup.2014.12.001. [DOI] [PubMed] [Google Scholar]
- Oser MG, Niederst MJ, Sequist LV, Engelman JA. Transformation from non-small-cell lung cancer to small-cell lung cancer: molecular drivers and cells of origin. Lancet Oncol. 2015;16:e165–e172. doi: 10.1016/S1470-2045(14)71180-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ou SH, Greenbowe J, Khan ZU, Azada MC, Ross JS, Stevens PJ, Ali SM, Miller VA, Gitlitz B. I1171 missense mutation (particularly I1171N) is a common resistance mutation in ALK-positive NSCLC patients who have progressive disease while on alectinib and is sensitive to ceritinib. Lung Cancer. 2015;88:231–234. doi: 10.1016/j.lungcan.2015.02.005. [DOI] [PubMed] [Google Scholar]
- Piotrowska Z, Niederst MJ, Karlovich CA, Wakelee HA, Neal JW, Mino-Kenudson M, Fulton L, Hata AN, Lockerman EL, Kalsy A, Digumarthy S, Muzikansky A, Raponi M, Garcia AR, Mulvey HE, Parks MK, DiCecca RH, Dias-Santagata D, Iafrate AJ, Shaw AT, Allen AR, Engelman JA, Sequist LV. Heterogeneity Underlies the Emergence of EGFRT790 Wild-Type Clones Following Treatment of T790M–Positive Cancers with a Third-Generation EGFR Inhibitor. Cancer Discov. 2015;5:713–722. doi: 10.1158/2159-8290.CD-15-0399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Press RD, Galderisi C, Yang R, Rempfer C, Willis SG, Mauro MJ, Druker BJ, Deininger MW. A half-log increase in BCR-ABL RNA predicts a higher risk of relapse in patients with chronic myeloid leukemia with an imatinib-induced complete cytogenetic response. Clin Cancer Res. 2007;13:6136–6143. doi: 10.1158/1078-0432.CCR-07-1112. [DOI] [PubMed] [Google Scholar]
- Relling MV, Gardner EE, Sandborn WJ, Schmiegelow K, Pui CH, Yee SW, Stein CM, Carrillo M, Evans WE, Klein TE C. Clinical Pharmacogenetics Implementation. Clinical Pharmacogenetics Implementation Consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing. Clin Pharmacol Ther. 2011;89:387–391. doi: 10.1038/clpt.2010.320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Relling MV, Pui CH, Cheng C, Evans WE. Thiopurine methyltransferase in acute lymphoblastic leukemia. Blood. 2006;107:843–844. doi: 10.1182/blood-2005-08-3379. [DOI] [PubMed] [Google Scholar]
- Sang J, Acquaviva J, Friedland JC, Smith DL, Sequeira M, Zhang C, Jiang Q, Xue L, Lovly CM, Jimenez JP, Shaw AT, Doebele RC, He S, Bates RC, Camidge DR, Morris SW, El-Hariry I, Proia DA. Targeted inhibition of the molecular chaperone Hsp90 overcomes ALK inhibitor resistance in non-small cell lung cancer. Cancer Discov. 2013;3:430–443. doi: 10.1158/2159-8290.CD-12-0440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Savage DG, Antman KH. Imatinib mesylate--a new oral targeted therapy. N Engl J Med. 2002;346:683–693. doi: 10.1056/NEJMra013339. [DOI] [PubMed] [Google Scholar]
- Schmiegelow K, Forestier E, Kristinsson J, Soderhall S, Vettenranta K, Weinshilboum R, Wesenberg F. H. Nordic Society of Paediatric and Oncology, Thiopurine methyltransferase activity is related to the risk of relapse of childhood acute lymphoblastic leukemia: results from the NOPHO ALL-92 study. Leukemia. 2009;23:557–564. doi: 10.1038/leu.2008.316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sequist LV, Soria JC, Goldman JW, Wakelee HA, Gadgeel SM, Varga A, Papadimitrakopoulou V, Solomon BJ, Oxnard GR, Dziadziuszko R, Aisner DL, Doebele RC, Galasso C, Garon EB, Heist RS, Logan J, Neal JW, Mendenhall MA, Nichols S, Piotrowska Z, Wozniak AJ, Raponi M, Karlovich CA, Jaw-Tsai S, Isaacson J, Despain D, Matheny SL, Rolfe L, Allen AR, Camidge DR. Rociletinib in EGFR-mutated non-small-cell lung cancer. N Engl J Med. 2015;372:1700–1709. doi: 10.1056/NEJMoa1413654. [DOI] [PubMed] [Google Scholar]
- Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, Bergethon K, Shaw AT, Gettinger S, Cosper AK, Akhavanfard S, Heist RS, Temel J, Christensen JG, Wain JC, Lynch TJ, Vernovsky K, Mark EJ, Lanuti M, Iafrate AJ, Mino-Kenudson M, Engelman JA. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med. 2011;3:75ra26. doi: 10.1126/scitranslmed.3002003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shah NP, Nicoll JM, Nagar B, Gorre ME, Paquette RL, Kuriyan J, Sawyers CL. Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia. Cancer Cell. 2002;2:117–125. doi: 10.1016/s1535-6108(02)00096-x. [DOI] [PubMed] [Google Scholar]
- Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007;7:169–181. doi: 10.1038/nrc2088. [DOI] [PubMed] [Google Scholar]
- Shaw AT, Engelman JA. ALK in lung cancer: past, present, and future. J Clin Oncol. 2013;31:1105–1111. doi: 10.1200/JCO.2012.44.5353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shaw AT, Friboulet L, Leshchiner I, Gainor JF, Bergqvist S, Brooun A, Burke BJ, Deng YL, Liu W, Dardaei L, Frias RL, Schultz KR, Logan J, James LP, Smeal T, Timofeevski S, Katayama R, Iafrate AJ, Le L, McTigue M, Getz G, Johnson TW, Engelman JA. Resensitization to Crizotinib by the Lorlatinib ALK Resistance Mutation L1198F. N Engl J Med. 2016a;374:54–61. doi: 10.1056/NEJMoa1508887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shaw AT, Gandhi L, Gadgeel S, Riely GJ, Cetnar J, West H, Camidge DR, Socinski MA, Chiappori A, Mekhail T, Chao BH, Borghaei H, Gold KA, Zeaiter A, Bordogna W, Balas B, Puig O, Henschel V, Ou SH i. study. Alectinib in ALK-positive, crizotinib-resistant, non-small-cell lung cancer: a single-group, multicentre, phase 2 trial. Lancet Oncol. 2016b;17:234–242. doi: 10.1016/S1470-2045(15)00488-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shaw AT, Kim DW, Mehra R, Tan DS, Felip E, Chow LQ, Camidge DR, Vansteenkiste J, Sharma S, De Pas T, Riely GJ, Solomon BJ, Wolf J, Thomas M, Schuler M, Liu G, Santoro A, Lau YY, Goldwasser M, Boral AL, Engelman JA. Ceritinib in ALK-rearranged non-small-cell lung cancer. N Engl J Med. 2014;370:1189–1197. doi: 10.1056/NEJMoa1311107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi H, Hugo W, Kong X, Hong A, Koya RC, Moriceau G, Chodon T, Guo R, Johnson DB, Dahlman KB, Kelley MC, Kefford RF, Chmielowski B, Glaspy JA, Sosman JA, van Baren N, Long GV, Ribas A, Lo RS. Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy. Cancer Discov. 2014a;4:80–93. doi: 10.1158/2159-8290.CD-13-0642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi Y, Au JS, Thongprasert S, Srinivasan S, Tsai CM, Khoa MT, Heeroma K, Itoh Y, Cornelio G, Yang PC. A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology (PIONEER) J Thorac Oncol. 2014b;9:154–162. doi: 10.1097/JTO.0000000000000033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siena S, Sartore-Bianchi A, Di Nicolantonio F, Balfour J, Bardelli A. Biomarkers predicting clinical outcome of epidermal growth factor receptor-targeted therapy in metastatic colorectal cancer. J Natl Cancer Inst. 2009;101:1308–1324. doi: 10.1093/jnci/djp280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Signorovitch J, Ayyagari R, Reichmann WM, Wu EQ, Chen L. Major molecular response during the first year of dasatinib, imatinib or nilotinib treatment for newly diagnosed chronic myeloid leukemia: a network meta-analysis. Cancer Treat Rev. 2014;40:285–292. doi: 10.1016/j.ctrv.2013.09.004. [DOI] [PubMed] [Google Scholar]
- Smyth T, Paraiso KH, Hearn K, Rodriguez-Lopez AM, Munck JM, Haarberg HE, Sondak VK, Thompson NT, Azab M, Lyons JF, Smalley KS, Wallis NG. Inhibition of HSP90 by AT13387 delays the emergence of resistance to BRAF inhibitors and overcomes resistance to dual BRAF and MEK inhibition in melanoma models. Mol Cancer Ther. 2014;13:2793–2804. doi: 10.1158/1535-7163.MCT-14-0452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina K, Hatanaka H, Bando M, Ohno S, Ishikawa Y, Aburatani H, Niki T, Sohara Y, Sugiyama Y, Mano H. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561–566. doi: 10.1038/nature05945. [DOI] [PubMed] [Google Scholar]
- Soh SX, Lim JY, Huang JW, Jiang N, Yeoh AE, Ong ST. Multi-agent chemotherapy overcomes glucocorticoid resistance conferred by a BIM deletion polymorphism in pediatric acute lymphoblastic leukemia. PLoS One. 2014;9:e103435. doi: 10.1371/journal.pone.0103435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Solomon BJ, Mok T, Kim DW, Wu YL, Nakagawa K, Mekhail T, Felip E, Cappuzzo F, Paolini J, Usari T, Iyer S, Reisman A, Wilner KD, Tursi J, Blackhall F, Investigators P. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med. 2014;371:2167–2177. doi: 10.1056/NEJMoa1408440. [DOI] [PubMed] [Google Scholar]
- Stanulla M, Schaeffeler E, Flohr T, Cario G, Schrauder A, Zimmermann M, Welte K, Ludwig WD, Bartram CR, Zanger UM, Eichelbaum M, Schrappe M, Schwab M. Thiopurine methyltransferase (TPMT) genotype and early treatment response to mercaptopurine in childhood acute lymphoblastic leukemia. JAMA. 2005;293:1485–1489. doi: 10.1001/jama.293.12.1485. [DOI] [PubMed] [Google Scholar]
- Swanton C, Soria JC, Bardelli A, Biankin A, Caldas C, Chandarlapaty S, de Koning L, Dive C, Feunteun J, Leung SY, Marais R, Mardis ER, McGranahan N, Middleton G, Quezada SA, Rodon J, Rosenfeld N, Sotiriou C, Andre F. Consensus on precision medicine for metastatic cancers: a report from the MAP conference. Ann Oncol. 2016 doi: 10.1093/annonc/mdw192. [DOI] [PubMed] [Google Scholar]
- Takegawa N, Hayashi H, Iizuka N, Takahama T, Ueda H, Tanaka K, Takeda M, Nakagawa K. Transformation of ALK rearrangement-positive adenocarcinoma to small-cell lung cancer in association with acquired resistance to alectinib. Ann Oncol. 2016 doi: 10.1093/annonc/mdw032. [DOI] [PubMed] [Google Scholar]
- Thress KS, Paweletz CP, Felip E, Cho BC, Stetson D, Dougherty B, Lai Z, Markovets A, Vivancos A, Kuang Y, Ercan D, Matthews SE, Cantarini M, Barrett JC, Janne PA, Oxnard GR. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat Med. 2015;21:560–562. doi: 10.1038/nm.3854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tiacci E, Park JH, De Carolis L, Chung SS, Broccoli A, Scott S, Zaja F, Devlin S, Pulsoni A, Chung YR, Cimminiello M, Kim E, Rossi D, Stone RM, Motta G, Saven A, Varettoni M, Altman JK, Anastasia A, Grever MR, Ambrosetti A, Rai KR, Fraticelli V, Lacouture ME, Carella AM, Levine RL, Leoni P, Rambaldi A, Falzetti F, Ascani S, Capponi M, Martelli MP, Park CY, Pileri SA, Rosen N, Foa R, Berger MF, Zinzani PL, Abdel-Wahab O, Falini B, Tallman MS. Targeting Mutant BRAF in Relapsed or Refractory Hairy-Cell Leukemia. N Engl J Med. 2015;373:1733–1747. doi: 10.1056/NEJMoa1506583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai J, Lee JT, Wang W, Zhang J, Cho H, Mamo S, Bremer R, Gillette S, Kong J, Haass NK, Sproesser K, Li L, Smalley KS, Fong D, Zhu YL, Marimuthu A, Nguyen H, Lam B, Liu J, Cheung I, Rice J, Suzuki Y, Luu C, Settachatgul C, Shellooe R, Cantwell J, Kim SH, Schlessinger J, Zhang KY, West BL, Powell B, Habets G, Zhang C, Ibrahim PN, Hirth P, Artis DR, Herlyn M, Bollag G. Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc Natl Acad Sci U S A. 2008;105:3041–3046. doi: 10.1073/pnas.0711741105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Allen EM, Wagle N, Sucker A, Treacy DJ, Johannessen CM, Goetz EM, Place CS, Taylor-Weiner A, Whittaker S, Kryukov GV, Hodis E, Rosenberg M, McKenna A, Cibulskis K, Farlow D, Zimmer L, Hillen U, Gutzmer R, Goldinger SM, Ugurel S, Gogas HJ, Egberts F, Berking C, Trefzer U, Loquai C, Weide B, Hassel JC, Gabriel SB, Carter SL, Getz G, Garraway LA, Schadendorf D G. Dermatologic Cooperative Oncology Group of. The genetic landscape of clinical resistance to RAF inhibition in metastatic melanoma. Cancer Discov. 2014;4:94–109. doi: 10.1158/2159-8290.CD-13-0617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Cutsem E, Peeters M, Siena S, Humblet Y, Hendlisz A, Neyns B, Canon JL, Van Laethem JL, Maurel J, Richardson G, Wolf M, Amado RG. Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer. J Clin Oncol. 2007;25:1658–1664. doi: 10.1200/JCO.2006.08.1620. [DOI] [PubMed] [Google Scholar]
- Villanueva J, Infante JR, Krepler C, Reyes-Uribe P, Samanta M, Chen HY, Li B, Swoboda RK, Wilson M, Vultur A, Fukunaba-Kalabis M, Wubbenhorst B, Chen TY, Liu Q, Sproesser K, DeMarini DJ, Gilmer TM, Martin AM, Marmorstein R, Schultz DC, Speicher DW, Karakousis GC, Xu W, Amaravadi RK, Xu X, Schuchter LM, Herlyn M, Nathanson KL. Concurrent MEK2 mutation and BRAF amplification confer resistance to BRAF and MEK inhibitors in melanoma. Cell Rep. 2013;4:1090–1099. doi: 10.1016/j.celrep.2013.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M, Cipolla AK, Wubbenhorst B, Xu X, Gimotty PA, Kee D, Santiago-Walker AE, Letrero R, D’Andrea K, Pushparajan A, Hayden JE, Brown KD, Laquerre S, McArthur GA, Sosman JA, Nathanson KL, Herlyn M. Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell. 2010;18:683–695. doi: 10.1016/j.ccr.2010.11.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wagle N, Van Allen EM, Treacy DJ, Frederick DT, Cooper ZA, Taylor-Weiner A, Rosenberg M, Goetz EM, Sullivan RJ, Farlow DN, Friedrich DC, Anderka K, Perrin D, Johannessen CM, McKenna A, Cibulskis K, Kryukov G, Hodis E, Lawrence DP, Fisher S, Getz G, Gabriel SB, Carter SL, Flaherty KT, Wargo JA, Garraway LA. MAP kinase pathway alterations in BRAF-mutant melanoma patients with acquired resistance to combined RAF/MEK inhibition. Cancer Discov. 2014;4:61–68. doi: 10.1158/2159-8290.CD-13-0631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waller CF. Imatinib mesylate. Recent Results Cancer Res. 2014;201:1–25. doi: 10.1007/978-3-642-54490-3_1. [DOI] [PubMed] [Google Scholar]
- Wicki A, Mandala M, Massi D, Taverna D, Tang H, Hemmings BA, Xue G. Acquired Resistance to Clinical Cancer Therapy: A Twist in Physiological Signaling. Physiol Rev. 2016;96:805–829. doi: 10.1152/physrev.00024.2015. [DOI] [PubMed] [Google Scholar]
- Wong DJ, Robert L, Atefi MS, Lassen A, Avarappatt G, Cerniglia M, Avramis E, Tsoi J, Foulad D, Graeber TG, Comin-Anduix B, Samatar A, Lo RS, Ribas A. Antitumor activity of the ERK inhibitor SCH772984 [corrected] against BRAF mutant, NRAS mutant and wild-type melanoma. Mol Cancer. 2014;13:194. doi: 10.1186/1476-4598-13-194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu YL, Yang JCH, Kim DW, Su WC, Ahn MJ, Lee DH, Vansteenkiste JF, Zhang L, Felip E, Peng B, Gong Y, Zhao S, Amagasaki T, Akimov M, Tan DSW. Safety and efficacy of INC280 in combination with gefitinib (gef) in patients with EGFR-mutated (mut), MET-positive NSCLC: A single-arm phase lb/ll study. American Society of Clinical Oncology Annual Meeting; J Clin Oncol. 2014;32(5s) (suppl;abstr 8017) [Google Scholar]
- Yamaguchi N, Lucena-Araujo AR, Nakayama S, de Figueiredo-Pontes LL, Gonzalez DA, Yasuda H, Kobayashi S, Costa DB. Dual ALK and EGFR inhibition targets a mechanism of acquired resistance to the tyrosine kinase inhibitor crizotinib in ALK rearranged lung cancer. Lung Cancer. 2014;83:37–43. doi: 10.1016/j.lungcan.2013.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang JC, Wu YL, Schuler M, Sebastian M, Popat S, Yamamoto N, Zhou C, Hu CP, O’Byrne K, Feng J, Lu S, Huang Y, Geater SL, Lee KY, Tsai CM, Gorbunova V, Hirsh V, Bennouna J, Orlov S, Mok T, Boyer M, Su WC, Lee KH, Kato T, Massey D, Shahidi M, Zazulina V, Sequist LV. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol. 2015;16:141–151. doi: 10.1016/S1470-2045(14)71173-8. [DOI] [PubMed] [Google Scholar]
- Ying HQ, Chen J, He BS, Pan YQ, Wang F, Deng QW, Sun HL, Liu X, Wang SK. The effect of BIM deletion polymorphism on intrinsic resistance and clinical outcome of cancer patient with kinase inhibitor therapy. Sci Rep. 2015;5:11348. doi: 10.1038/srep11348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Youle RJ, Strasser A. The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 2008;9:47–59. doi: 10.1038/nrm2308. [DOI] [PubMed] [Google Scholar]
- Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, Kris MG, Miller VA, Ladanyi M, Riely GJ. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res. 2013;19:2240–2247. doi: 10.1158/1078-0432.CCR-12-2246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zabriskie MS, Eide CA, Tantravahi SK, Vellore NA, Estrada J, Nicolini FE, Khoury HJ, Larson RA, Konopleva M, Cortes JE, Kantarjian H, Jabbour EJ, Kornblau SM, Lipton JH, Rea D, Stenke L, Barbany G, Lange T, Hernandez-Boluda JC, Ossenkoppele GJ, Press RD, Chuah C, Goldberg SL, Wetzler M, Mahon FX, Etienne G, Baccarani M, Soverini S, Rosti G, Rousselot P, Friedman R, Deininger M, Reynolds KR, Heaton WL, Eiring AM, Pomicter AD, Khorashad JS, Kelley TW, Baron R, Druker BJ, Deininger MW, O’Hare T. BCR-ABL1 compound mutations combining key kinase domain positions confer clinical resistance to ponatinib in Ph chromosome-positive leukemia. Cancer Cell. 2014;26:428–442. doi: 10.1016/j.ccr.2014.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhitomirsky B, Assaraf YG. Lysosomes as mediators of drug resistance in cancer. Drug Resist Updat. 2016;24:23–33. doi: 10.1016/j.drup.2015.11.004. [DOI] [PubMed] [Google Scholar]
- Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, Zhang S, Wang J, Zhou S, Ren S, Lu S, Zhang L, Hu C, Hu C, Luo Y, Chen L, Ye M, Huang J, Zhi X, Zhang Y, Xiu Q, Ma J, Zhang L, You C. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011;12:735–742. doi: 10.1016/S1470-2045(11)70184-X. [DOI] [PubMed] [Google Scholar]