Skip to main content
Molecular and Cellular Biology logoLink to Molecular and Cellular Biology
. 2014 May;34(10):1722–1732. doi: 10.1128/MCB.01592-13

Clinical Targeting of Mutated and Wild-Type Protein Tyrosine Kinases in Cancer

Justin M Drake a, John K Lee b,c, Owen N Witte a,d,e,f,
PMCID: PMC4019040  PMID: 24567371

Abstract

Clinical therapies for cancer have evolved from toxic, nontargeted agents to manageable, highly targeted therapies. Protein tyrosine kinases are a family of signaling molecules implicated in nearly every cancer type and are the foundation for the development of modern targeted agents. Recent genomic analyses have identified activating mutations, translocations, and amplifications of tyrosine kinases. Selective targeting of these genetically altered tyrosine kinases has resulted in significant clinical advances, including increased patient survival. This indicates that altered protein tyrosine kinases are the main drivers of many different cancers. However, lost during analyses of genetic lesions are the contributions of activated, wild-type kinases on tumor-dependent pathways. New approaches in phosphoproteomic technologies have identified several wild-type tyrosine kinase activation states, suggesting that non-genetically altered kinases can be essential “nodes” for signal transduction. Here, we summarize the evidence supporting the common mechanisms of protein tyrosine kinase activation in cancer and provide a personal perspective on the kinases BCR-ABL and BTK, as well as nonmutated kinase targets in prostate cancer, through our work. We outline the mechanisms of tyrosine kinase activation in the absence of direct mutation and discuss whether non-genetically altered tyrosine kinases or their associated downstream signaling pathways can be effectively targeted.

INTRODUCTION

The seminal finding of BCR-ABL tyrosine kinase activity (1) and the subsequent development of the small-molecule inhibitor imatinib (2) have pioneered an era of therapies targeted toward mutated tyrosine kinases in cancer. Advancements in genomic sequencing and in our understanding of tumor signaling pathways have revealed several mutated and nonmutated pathway-activated tyrosine kinases. In this minireview, we will describe the different mechanisms of tyrosine kinase activation and how therapies tailored to these kinases have transformed disease outcomes. In particular, we will highlight two main examples of tyrosine kinases investigated in our lab, BCR-ABL and Bruton's tyrosine kinase (BTK). We will also examine a shifting paradigm in which nonmutated tyrosine kinases are being considered primary targets for tyrosine kinase inhibitor (TKI) therapy in cancer. Finally, we will discuss how phosphoproteomic approaches can identify activation of nonmutated kinase pathways and the challenges that we face by clinically targeting activated wild-type tyrosine kinases.

TYROSINE KINASE SIGNALING

Since the discovery of v-SRC and v-ABL tyrosine phosphorylation 35 years ago (3, 4), considerable progress has been made in understanding how tyrosine phosphorylation contributes to normal cellular homeostasis and disease. Tyrosine phosphorylation is regulated by a family of enzymes known as tyrosine kinases. Tyrosine kinases are crucial mediators of normal cellular signal transduction functions, including cell proliferation, survival, migration, and apoptosis. Humans express at least 90 tyrosine kinases, including 58 receptor tyrosine kinases (RTKs) (5). RTKs are activated through binding of their extracellular domain to ligands, such as growth factors and cytokines. This ligand binding results in RTK dimerization/oligomerization (6) and subsequent tyrosine phosphorylation. The N-lobe of the tyrosine kinase domain adopts a conformational change upon activation, initiating 2 phases of tyrosine phosphorylation (7). The first phase of tyrosine autophosphorylation occurs in trans, and phosphotyrosine residues are phosphorylated within the activation loop of the tyrosine kinase domain (in a precise order) or juxtamembrane domain (7). The second phase of tyrosine autophosphorylation occurs on phosphotyrosines that are recognized by signaling molecules containing Src homology 2 (SH2) or phosphotyrosine binding (PTB) domains (8). The recruitment of these signaling molecules then initiates a cascade of RTK-specific pathways that regulates cell fate. Hence, the abundance of signaling molecules recruited by activated RTKs renders these kinases crucial nodes for transmission of cellular information.

Despite the large number of signaling molecules recruited by RTKs, regulation of tyrosine kinases is tightly controlled to prevent aberrant cellular activities, usually through receptor-mediated ubiquitylation and degradation (9). However, genetically altered mutations or pathway-activated mechanisms of tyrosine kinases can transform a cell from a normal state into a cancerous one. Over 50% of the 90 tyrosine kinases identified have been implicated in cancer despite the fact that tyrosine phosphorylation represents only 1% of the total phosphoproteome (10). The disproportionate contribution of tyrosine kinases to cancer further highlights the need to understand the signaling networks initiated by tyrosine kinases as well as how to effectively target them.

MAJOR MECHANISMS FOR GENETICALLY ALTERED AND NONMUTATED TYROSINE KINASE ACTIVATION IN CANCER

Constitutive signals from either mutated or wild-type tyrosine kinases result in hyperactive pathways leading to continued cancer cell growth and survival. This reliance on the hyperactive pathway, known as pathway addiction, has provided unique opportunities for targeted inhibition of tyrosine kinases and has resulted in clinical successes (Table 1). Genetically altered tyrosine kinases can act as drivers in cancer through activating mutations (1114), DNA translocations (1517), or DNA amplifications (18) (Fig. 1A to C). However, wild-type or nonmutated tyrosine kinases can also function as critical nodes for pathway activation (1922) (Fig. 1D and E).

TABLE 1.

Activating genetic alterations of tyrosine kinases and FDA-approved targeted inhibitors in human malignanciesa

Genetic alteration Kinase Disease(s) FDA-approved inhibitor(s)
Mutation(s)
    Exons 19, 21 EGFR NSCLC Erlotinib, afatinib
    Exons 8, 9, 11, 13, 17 c-KIT GIST Imatinib, sunitinib
    Exons 12, 14, 18 PDGFRα GIST Imatinib, sunitinib
    Exons 10, 11, 13, 14 RET Medullary thyroid carcinoma Vandetinib, cabozantinib
    V617F JAK2 Myeloproliferative neoplasms Ruxolitinib
Translocation(s)
    BCR-ABL1 fusion ABL1 CML Imatinib, dasatinib, nilotinib, bosutinib, ponatinib
    RET/PTC family fusions RET PTC Vandetinib
    EML4-ALK fusion ALK NSCLC Crizotinib
Amplification(s): increased copy no. and/or overexpression ERBB2 Breast cancer, gastric cancer Trastuzumab, lapatinib, ado-trastuzumab emtansine, pertuzumab
Pathway activation
    B-cell receptor signaling BTK CLL Ibrutinib
B-cell non-Hodgkin's lymphoma
    Angiogenesis VEGFR CCRCC, soft-tissue carcinoma Sorafenib, sunitinib, pazopanib, axitinib
a

Abbreviations: NSCLC, non-small-cell lung cancer; GIST, gastrointestinal stromal tumor; CML, chronic myelogenous leukemia; PTC, papillary thyroid carcinoma; CCRCC, clear cell renal cell carcinoma; CLL, chronic lymphocytic leukemia.

FIG 1.

FIG 1

Mechanisms of mutated and wild-type tyrosine kinase activation in cancer. Three prototypical mechanisms of genetically altered tyrosine kinase activation as evidenced through genetic alterations. Shown are examples of mutations (A), amplifications (B), or translocations (C) that render a kinase constitutively active and thus contribute to the addiction of tumor cells on pathways driven by the genetically altered kinase. In the absence of genetic alterations (D to F), nonmutated tyrosine kinases can be activated via many different mechanisms and contribute to pathway signaling. In all cases, this pathway addiction leads to increased tumor cell proliferation and survival. In some cases, highly potent therapies that inhibit (red) the altered protein kinase have resulted in robust clinical outcomes. RTK, receptor tyrosine kinase; Chr, chromosome; Ph1, Philadelphia chromosome; BCR, breakpoint cluster region; Ag, antigen; BTK, Bruton's tyrosine kinase; PTP1B, protein tyrosine phosphatase 1B.

Somatic activating mutations usually occur within the activation loop of the kinase domain (13, 14, 23). These mutations serve to stabilize the kinase in an active confirmation and to destabilize cis-inhibitory interactions. However, mutations of other domains within the tyrosine kinase can elicit constitutive activity. These include the juxtamembrane domain of KIT (13) or the cysteine-rich extracellular domain of RET (23). The cysteine-rich extracellular domain of RET forms intramolecular disulfide bonds with other cysteines in the kinase. Mutations in these cysteine residues increase aberrant disulfide bonding between partner RET molecules, resulting in dimerization and constitutive activity (24). Sequences within the juxtamembrane domain of KIT interact with several parts of the tyrosine kinase domain, maintaining the kinase in an inhibitory state. Mutations within the juxtamembrane domain of KIT disrupt these autoinhibitory interactions, resulting in a constitutively active kinase (13). Clinically, the different locations of the mutations for a particular kinase can dramatically influence patient survival (25). For example, in gastrointestinal stromal tumors (GIST), c-KIT mutations occur in several different exons (exon 9, 11, 13, or 17), and patients with mutations in exon 11 (juxtamembrane domain) respond favorably to imatinib therapy, with a longer median survival than that of patients with mutations in exon 9 (extracellular domain) (26). While the mechanism is not clear, the evidence paints a clear picture that segregating patients based on mutational status may be an important strategy for therapy decisions.

DNA translocations predominantly result in the molecular deregulation of a gene at one of the breakpoints or by creating a gene fusion. These chromosomal rearrangements include deregulation of two main classes of proteins: transcriptional regulators (frequency, 38 to 44%), such as MYC, ETV1, ERG, and RUNX1, and tyrosine kinases (frequency, 5 to 7%), including ABL, fibroblast growth factor receptor 1 (FGFR1), RET, and ALK (27, 28). Although several different tyrosine kinases are involved in gene fusions, a common feature is to maintain the catalytically active tyrosine kinase domain while removing the upstream inhibitory domains (28). Further, the breakpoint region is often conserved for each tyrosine kinase and usually located within one to three introns upstream of the tyrosine kinase domain (28). Finally, the same tyrosine kinase can have multiple fusion partners even within the same disease. These fusion partners contain coiled-coil or leucine zipper domains that result in dimerization or oligomerization of the rearranged kinase, leading to ligand-independent activation (28). However, other mechanisms, such as 5′ regulatory sequences that drive tyrosine kinase expression (29) or removal of the N-terminal myristoylation site of the BCR-ABL fusion, an important autoinhibitory element in ABL kinase (30), may also occur.

DNA amplifications, as observed with ERBB2 (HER2/neu) in breast cancer (18) or MET in lung cancer (31) and gastric cancer (32), result in constitutive kinase activity by increasing its dimerization at the cell surface. ERBB2 amplification/overexpression is found in 25 to 30% of breast cancers and is a significant predictor of overall survival and time to relapse in patients with breast cancer (18). In one study of younger women, ERRB2 amplification as measured by fluorescent in situ hybridization (FISH) was highly predictive of recurrence (33). Patients with low- or high-level amplifications demonstrated a 54% or 86% recurrence rate, respectively, compared to 17% in patients without amplification. Furthermore, ERRB2 amplification status was a stronger indicator of breast cancer recurrence risk than were classical clinical predictors such as lymph node status or tumor size. The identification of ERRB2 as a clinical biomarker and the elucidation of its role in the pathogenesis of a subset of breast cancers subsequently led to the development of the ERBB2-targeted antibody trastuzumab (Herceptin). A pivotal trial in patients with ERRB2-amplified metastatic breast cancer demonstrated that the addition of trastuzumab to chemotherapy was associated with a higher response rate, longer disease-free survival, increased median survival, and a 20% reduction in disease-related death (34).

An alternative mechanism of tyrosine kinase activation involves nonmutated pathway activation. Activation of wild-type tyrosine kinases includes BTK in hematopoietic malignancies (35) and vascular endothelial growth factor receptors 1 and 2 (VEGF1/2), SRC, and FGFR1 in adenocarcinomas (2022, 36). Since genetic mutations of these kinases are not frequently observed in these tumors, other mechanisms are postulated to regulate tyrosine kinase activity independently of constitutive, cell-autonomous activation. These include positive feedback loops whereby downstream mitogenic pathways positively influence tyrosine kinase activity (37); paracrine or autocrine stimulation by receptor tyrosine kinase ligands (21); increased or decreased phosphatase activity targeting pathway regulators or dephosphorylating the inhibitory sites of tyrosine kinases, resulting in activation (38); and decreased proteosomal degradation by stabilizing tyrosine kinase levels through heat shock protein 90 (HSP90) scaffold binding (39). While not exhaustive, these mechanisms shed light on several different ways that nonmutated tyrosine kinases can elicit pathway activation and emphasize the possibility that targeting these kinases or pathway components in cancer would be as beneficial as targeting mutated kinases in the clinical setting.

GENETICALLY ALTERED (BCR-ABL) AND WILD-TYPE (BTK) TYROSINE KINASES AS THERAPEUTIC TARGETS

From work done in our laboratory (1, 35, 4049), two well-characterized but distinct examples of tyrosine kinase activity in hematopoietic malignancies are the BCR-ABL activating translocation in chronic myelogenous leukemia (CML) and acute lymphoblastic leukemia (ALL) with the Philadelphia chromosome and pathway activation through BTK in B-cell malignancies. Below, we compare and contrast these two kinases and point out the crucial observation that tyrosine kinase activation drives cancer progression via either genetic alterations (i.e., mutations) or pathway activation (i.e., wild-type kinases).

THE DUAL FUNCTION OF TYROSINE KINASE c-ABL IN CELLULAR GROWTH AND TRANSFORMATION IS DEPENDENT ON ITS CELLULAR LOCALIZATION AND PHOSPHORYLATION STATUS

The ABL nonreceptor tyrosine kinases include ABL1 (c-ABL) and ABL2 (ARG). Both of these kinases have distinct and similar functions as indicated through genetic inactivation in mice (5052). Double knockout (ABL/ARG−/−) day 10.25 embryos displayed delayed closure of the neural tube as well as severe defects in the apical network of the actin cytoskeleton (50). These mice often die by embryonic day 10.5 with hemorrhage into the peritoneum or pericardial space (50). Further, global knockout of ABL in C57BL/6J mice revealed severe cardiac hypertrophy due to increased proliferation and perinatal lethality (53). Recently, c-ABL and ARG were found to play an important role in vascular function (54). A mouse expressing the endothelial tissue-specific knockout of the ABL gene was crossed with a global ARG−/− background, resulting in lethality at late embryonic and perinatal stages of development (54). This was accompanied by regions of vascular loss as evidenced by increased endothelial cell apoptosis, diminished TEK (TIE2) signaling, and loss of ANGPT1-mediated survival (54).

The structural organization of c-ABL is highly similar to that of SRC family kinases. They both include an SH3 domain, an SH2 domain, and a catalytic core. However, c-ABL has a unique C terminus relative to SRC family kinases and includes F- and G-actin binding domains and nuclear export, proline-rich, nuclear localization, and DNA binding sequences (55). c-ABL activity is regulated by intramolecular interactions primarily between the SH2 and SH3 domains, which maintain c-ABL in a closed, inactive conformation (56). c-ABL continuously shuttles between the nucleus and cytoplasm. The equilibrium between these subcellular locations dictates the biological function of c-ABL. Nuclear c-ABL functions as a regulator of gene expression and inhibitor of cellular growth, while cytosolic c-ABL is involved in cellular signaling and localization to cellular adhesion complexes (5759).

Interestingly, overexpression of c-ABL does not transform cells (60). However, the transformation capacity of c-ABL can be enhanced by several distinct mechanisms, including mutations in the kinase (61) or SH3 (62) domains, deletions of the SH3 domain (60), and translocations (47), which all maintain the tyrosine kinase activity of c-ABL (1). Tyrosine phosphorylation within the activation loop of c-ABL is critical for cellular localization and kinase activity driving cellular growth and transformation. Activation loop tyrosine phosphorylation can be triggered by activation of SRC family kinases (SFK) (63), epidermal growth factor receptor (EGFR) family members (64, 65), or insulin-like growth factor 1 receptor (IGF1R) (64), which then signals through downstream effectors such as extracellular signal-regulated kinase 5 (ERK5) (63) or Jun N-terminal protein kinase (JNK) or STAT3 (64), leading to increased tumor growth and proliferation. Indeed, tyrosine kinase inhibitors such as imatinib bind the c-ABL activation loop in its inactive conformation, thereby blocking the ability of the activation loop to bind ATP productively and rendering the kinase inactive (66).

CLINICAL TARGETING OF BCR-ABL GENE FUSION IN LEUKEMIA

The discovery of the Philadelphia (Ph) chromosome by Nowell and Hungerford provided evidence of a consistent genetic alteration in CML (67). This chromosomal abnormality mapped the cellular oncogene c-ABL from chromosome 9 to the breakpoint cluster region (BCR) of chromosome 22, creating the BCR-ABL fusion gene (68, 69). Phosphorylation of ABL kinase is ensured via translocation of BCR-ABL to the cytosol, which has constitutive kinase activity (1). Autophosphorylation and activation of the tyrosine kinase domain of BCR-ABL are accomplished by the formation of homotetrameric complexes mediated by the BCR coiled-coil domain (70). In addition to eliciting kinase activity, BCR-ABL also has kinase-independent functions that involve phosphorylation of Y177 in the BCR domain. This phosphorylation can act as a scaffold and recruits GRB2 to initiate downstream signaling pathways (71). Indeed, other kinases such as ERBB3 and PEAK1 (SGK269) function in a similar context, exhibiting elevated tyrosine phosphorylation in cancer cells behaving more as a scaffold rather than stimulating constitutive kinase activity (72, 73). However, it was the constitutive kinase activity of BCR-ABL that made it a very attractive target for inhibition. The chemical compound CGP 57148 was screened in vitro against a panel of kinases and was found to inhibit v-ABL with nanomolar affinity (74). This lead compound (now named imatinib [Gleevec]) was later shown to inhibit BCR-ABL tyrosine kinase activity (75) and was approved as a new treatment for patients harboring the BCR-ABL mutation in CML.

In CML, the severity of this disease can be classified into 3 phases: chronic, accelerated, and blast (or blast crisis) (76). Most patients are diagnosed in the chronic phase, are minimally symptomatic, and respond well to standard therapies. Over the course of 3 to 5 years, progression to the accelerated or blast phase occurs. Prior to 2001, the median survival rates were 3 to 5 years from the time of diagnosis and a bone marrow transplant was the only potentially curative treatment option. The introduction of imatinib in CML therapy has led to a major transformation in the natural history of the disease. In a 5-year follow-up of the initial phase III IRIS study, 93% of patients in chronic phase receiving imatinib did not progress to the accelerated phase or blast crisis, with an estimated overall survival rate near 90% (77). Based on a review of the outcomes associated with a diagnosis of CML since 1965 at the M. D. Anderson Cancer Center, the substantial impact of imatinib therapy on CML therapy is evident particularly in the chronic and accelerated phases of the disease (78). The 8-year survival rate of patients in chronic phase has climbed from 6% (before 1975) to 87% (since 2001). In addition, the 8-year survival rate of patients in accelerated phase has increased from <20% (before 1990) to 75% (since 2001). Furthermore, a recent study demonstrated that CML patients who are being treated with imatinib and have achieved a complete cytogenetic remission after 2 years have a 1% CML-related 8-year mortality rate that is not statistically different from the mortality rate of the general population (79).

The clinical success of imatinib in CML also brought into focus patients who were refractory to or who relapsed during imatinib therapy. Primary resistance to imatinib in chronic-phase CML is uncommon, with over 98% of patients demonstrating a complete hematologic response by 60 months in the IRIS trial (77). However, secondary resistance is problematic, as the failure rate at 60 months for patients receiving imatinib is 17%, with the majority of failures occurring within the first few years of treatment. Sequencing analysis of the blood and marrow samples from patients who relapsed after imatinib therapy demonstrated that the majority exhibited ABL kinase domain mutations that either disrupt amino acids in contact with imatinib or preclude the inactive conformational state of BCR-ABL necessary for imatinib interaction (80). Additional mechanisms of resistance have been identified and include the overexpression or amplification of BCR-ABL, activation of BCR-ABL-independent pathways, and alterations in drug influx/efflux (81, 82).

To overcome imatinib resistance, additional ABL kinase inhibitors, such as dasatinib, nilotinib, and bosutinib, were also developed as ATP-competitive inhibitors and showed high efficacy in first-line therapy and second-line therapy after failure of imatinib (8386). However, one clinically challenging mutation has been the T315I “gatekeeper mutation,” which confers resistance to nearly all BCR-ABL tyrosine kinase inhibitors (TKIs). Thr315 resides at the back of the ATP pocket, and replacement with a bulky hydrophobic isoleucine causes activation of the kinase through stabilization of the hydrophobic spine and increased interaction with ATP (87). A novel inhibitor named ponatinib that circumvents the T315I mutation and other mutations through structure-guided drug design has now been approved (88). Ponatinib forms a complex network of distributed binding interactions over multiple residues of the ABL kinase domain, which renders the complex less prone to disruption from point mutations (89). In a phase II trial in heavily pretreated patients with CML, ponatinib was highly active, with one-third of the patients demonstrating a major molecular response, indicating a multiple-log decrease in BCR-ABL transcript level (83). In addition, no single BCR-ABL mutation was identified that conferred resistance to ponatinib (83). Ponatinib received accelerated approval by the FDA for resistant CML but was subsequently placed on a partial hold due to significantly increased risks of serious venous and arterial thromboses appreciated after an additional 24-month clinical follow-up of the trial. However, given the therapeutic risk-to-benefit ratio, particularly for patients resistant to other TKI therapies, the sale and development of ponatinib were reauthorized with an amended boxed warning for patients.

ROLE OF BTK IN B-CELL LINEAGE DEVELOPMENT

The nonreceptor tyrosine kinase BTK is important for B-cell lineage development. A wide array of hereditary missense and nonsense mutations, insertions, and deletions spanning all domains of BTK lead to human X-linked agammaglobulinemia (XLA), which results in the arrest of early B-lymphoid development in humans (49, 90, 91). Patients with these mutations are susceptible to bacterial infections and enteroviral disease with a loss of plasma cells and reduction of serum IgG, IgM, and IgA (92). Gamma globulin therapy and antibiotics are mainstays in the treatments of these patients (92).

Analogous to human XLA, deletion or mutation of BTK in the CBA/N mouse leads to X-linked immunodeficiency (XID) (93, 94). The XID mutation results in a reduction of B-cell numbers to 30 to 50% of normal with the remaining B cells expressing an unusual IgM-high and IgD-low phenotype (95). Further, these mice have lower serum levels of IgM and IgG3, do not enter S phase after anti-IgM stimulation, and lack CD5+ B-1 B cells (95, 96). As a result, B cells do not respond appropriately to a variety of activation signals, including interleukin 5 (IL-5), IL-10, and CD40 (9799), and also fail to make antibodies to T-cell-independent type 2 antigens (95). Overall, the XID phenotype typically results in a milder form of XLA despite the observation of identical mutations (R28 residue) in patients with classical XLA (100). However, there are examples where patients display mild or asymptomatic XLA and yet harbor the R28 mutation (101, 102). This suggests that the genetic background of an individual is important in determining the severity of the XLA phenotype.

The evidence for BTK in B-cell lineage development indicates that this tyrosine kinase is critical for B-cell maturation and activation. Activation of wild-type BTK is triggered by B-cell receptor cross-linking, leading to the membrane localization of a fraction of BTK molecules (103). This translocation results in a 2-step process of BTK tyrosine phosphorylation, first by SRC family kinases followed by autophosphorylation within the SH2 domain of BTK (35, 40). Phosphorylation of BTK by SYK or LYN tyrosine kinases results in the phosphorylation and subsequent activation of phospholipase Cγ2 (PLCγ2) (104). PLCγ2 phosphorylation ultimately regulates downstream mitogen-activated protein kinase (MAPK)- and phosphoinositol-3-kinase (PI3K)-dependent signaling networks (105). These downstream signaling networks result in a diverse array of protumorigenic processes, including increased gene expression, cell proliferation, and survival (105).

WILD-TYPE BTK ACTIVATION AND THERAPEUTIC TARGETING IN LYMPHOMAS

B-cell receptor activation and subsequent signaling through BTK appear to be critical to the pathogenesis of certain B-cell malignancies. In chronic lymphocytic leukemia (CLL), autoantigen binding and activation of the B-cell receptor induce the proliferation of leukemic clones (106). More-convincing genetic evidence of the importance of chronic active B-cell receptor signaling has been delineated in the activated B-cell-like (ABC) subtype of diffuse large B-cell lymphoma (DLBCL), in which BTK and other B-cell receptor pathway components are essential for the survival of the lymphoma (107).

The importance of BTK function in B-cell malignancies has led to the development of the tyrosine kinase inhibitor ibrutinib (PCI-32765). This drug has received three Breakthrough Therapy designations from the FDA due to its activity and promise in treating multiple B-cell malignancies, including mantle cell lymphoma (MCL) (108), Waldenstrom's macroglobulinemia (109), and CLL (110).

Ibrutinib initially received accelerated approval in the United States in November 2013 for the indication of relapsed or refractory MCL based on a phase II study that demonstrated a 68% overall response rate and estimated progression-free survival of 13.9 months (108). Notably, few grade 3 or 4 toxicities were reported, indicating that the drug was well tolerated by patients. The high activity of ibrutinib has also been reported in patients with Waldenstrom's macroglobulinemia. In a phase II study of patients with relapsed or refractory Waldenstrom's macroglobulinemia, the overall response rate was 81%, paralleled by significant improvements in red blood cell indices and reductions in serum IgM levels (109). Perhaps the greatest overall impact of ibrutinib may be seen in CLL. In a phase Ib/II trial evaluating ibrutinib in patients with relapsed or refractory CLL, the overall response rate was 71% with minimal grade 3 or 4 toxicities (110). Strikingly, the subgroup of patients with the adverse 17p13.1 deletion, wherein loss of the TP53 locus confers resistance to cytotoxic chemotherapy and portends a poor prognosis, demonstrated a 68% overall response rate with ibrutinib therapy.

PHOSPHOPROTEOMIC AND KINOME siRNA SCREENING METHODS TO DETECT AND ASSESS FUNCTION OF ACTIVATED, WILD-TYPE TYROSINE KINASES AS THERAPEUTIC TARGETS IN CANCER

Clinically, tumors are primarily assessed using genetic sequencing technologies, therefore missing pathway-activated, nonmutated tyrosine kinases. However, the lack of predominant activating tyrosine kinase mutations in some epithelial cancers makes it difficult to pinpoint essential pathway drivers, and new approaches are needed to identify nonmutated, pathway-activated tyrosine kinases. To assess these kinases, phosphoproteomic enrichment strategies coupled with highly sensitive quantitative mass spectrometry (MS) and kinome-wide small interfering RNA (siRNA) screens have been utilized (37, 111125).

Kinome-wide siRNA screens have been used on numerous cancer cell lines to assess the behavior and effect of targeting each individual kinase within a controlled system. These screens have identified kinases that behave as regulators of hormonal signaling (122), survival and proliferation (120), mechanisms of escape during hormonal depletion (126, 127), regulators of downstream pathway activation (119, 123), or mechanisms of drug resistance (118, 121).

Phosphoproteomics provides valuable information about the activation states of tyrosine kinases independent of genomic alterations, thereby uncovering tyrosine kinases that may be driving essential pathways in cancers that next-generation whole-genome or exome sequencing may miss (117, 124, 125). A pioneering study investigating the phosphoproteomic landscape in non-small-cell lung cancer (NSCLC) tissues and cell lines identified over 50 different kinases and 2,500 downstream substrates (116). The authors further show that cell lines expressing heightened levels of the phosphorylated receptor tyrosine kinase (RTK) platelet-derived growth factor receptor alpha (PDGFRα) were sensitive to imatinib treatment while resistant cell lines were negative for PDGFRα expression. This study provides a proof of principle that kinase profiling may predict the sensitivity of tumors to selected TKIs based on RTK activation patterns. Since this publication, phosphoproteomics analyses have been performed on numerous epithelial and hematogenous cancer tissues and cell lines, including lung cancer (113, 115, 116), breast cancer (117, 128, 129), colorectal cancer (130), renal cancer (131), melanoma (132), prostate cancer (111, 125, 133, 134), CML (37), and acute myeloid leukemia (AML) (114).

We have implemented phosphoproteomics to investigate tyrosine kinase activity in both mouse models of prostate tumorigenesis where we express nontyrosine kinase oncogenes (111) and human metastatic castration-resistant prostate cancer (CRPC) tissues obtained at rapid autopsy (125). Despite the low overall mutation rates and rare events of activating mutations of tyrosine kinases in prostate cancer (135, 136), we observed numerous activated tyrosine kinases, including SRC, EGFR, ABL, and JAK2, as well as tyrosine phosphorylation of MAPK1/3 and STAT3 (111, 125). Our phosphoproteomic profiling analysis also revealed tyrosine phosphorylation signatures to be strikingly different between prostate cancer cell line-derived xenografts, treatment-naive prostate cancers, and metastatic CRPC (125, 133). This finding is interesting, as it would suggest that cell lines (at least in prostate cancer) do not accurately reflect the phosphorylation states of prostate cancer tissues isolated from patients. Most phosphoproteomic studies analyze cell lines perturbed by wild-type or mutated oncogenes implicated in the cancer type of interest and therefore limit analysis to a finite set of kinase alterations. While valuable, these analyses would not be useful for cancers that lack activating mutations of tyrosine kinases, and increased emphasis should be placed on analyzing patient tumors.

The biggest challenge arising from this and other phosphoprofiling studies will be to determine if the activated tyrosine kinases identified via phosphoproteomics are drivers of cancer in a fashion similar to that of genetically altered tyrosine kinases seen in other cancer types. In prostate cancer, the need for relevant in vivo models to assess the function of nonmutated activated tyrosine kinases is crucial to understanding the signaling pathways and nodes for appropriate therapy in this disease. In addition, the promiscuity of kinase inhibitors such as cabozantinib (VEGFR2, MET, or RET inhibitor) or sorafenib (RAF or VEGFR inhibitor) confounds the exact contribution of specific driver kinases in regulating survival and growth. However, this may be of therapeutic benefit, as multiple targets can be simultaneously inhibited with these agents, as suggested in CML (137). Finally, the evaluation of mechanisms of resistance to activated tyrosine kinases should also be assessed to predict combination or sequential therapies that could be tested clinically. This has been realized in certain cancers, such as BCR-ABL in CML, where sequential administration of new therapies circumvents common resistance mechanisms (83, 86).

CONCLUDING REMARKS

As we move toward the era of personalized medicine, the reliance on state-of-the-art technologies such as next-generation sequencing and phosphoproteomics will dramatically advance our knowledge base of activated kinases in cancer. Already, this has led to dramatic improvements in cancer therapy for diseases such as breast cancer, NSCLC, CML, and B-cell malignancies. Tyrosine kinase inhibitors are also less toxic and more potent than, and highly specific compared to, previous toxic, nonspecific chemotherapies such as alkylating agents, mitotic agents, or topoisomerase inhibitors. Combined, these methodologies will present a clearer picture of the activation states of genetically altered or nonmutated kinases for targeted therapy and will enable the systematic evaluation of resistance mechanisms during cancer treatment.

ACKNOWLEDGMENTS

We apologize to any contributor in this field, including those who performed all the clinical trial work in Table 1, whose papers were not cited due to space limitations required by the journal. We thank members of the O.N.W. laboratory for helpful comments and discussion on the manuscript.

J.M.D. is supported by the Department of Defense Prostate Cancer Research Program (W81XWH-11-1-0504). J.K.L. is supported by the UCLA Specialty Training and Advanced Research Program. O.N.W. is an investigator of the Howard Hughes Medical Institute. O.N.W. is partially supported by funds from the California Institute for Regenerative Medicine and from the Prostate Cancer Foundation Challenge Award for Defining Targets and Biomarkers in Prostate Cancer Stem Cells. O.N.W. is also partially supported by a Stand Up to Cancer-Prostate Cancer Foundation-Prostate Dream Team Translational Cancer Research Grant. This research grant is made possible by the generous support of the Movember Foundation. Stand Up To Cancer is a program of the Entertainment Industry Foundation administered by the American Association for Cancer Research.

Footnotes

Published ahead of print 24 February 2014

REFERENCES

  • 1.Konopka JB, Watanabe SM, Witte ON. 1984. An alteration of the human c-abl protein in K562 leukemia cells unmasks associated tyrosine kinase activity. Cell 37:1035–1042. 10.1016/0092-8674(84)90438-0 [DOI] [PubMed] [Google Scholar]
  • 2.Druker BJ, Sawyers CL, Kantarjian H, Resta DJ, Reese SF, Ford JM, Capdeville R, Talpaz M. 2001. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N. Engl. J. Med. 344:1038–1042. 10.1056/NEJM200104053441402 [DOI] [PubMed] [Google Scholar]
  • 3.Witte ON, Dasgupta A, Baltimore D. 1980. Abelson murine leukaemia virus protein is phosphorylated in vitro to form phosphotyrosine. Nature 283:826–831. 10.1038/283826a0 [DOI] [PubMed] [Google Scholar]
  • 4.Hunter T, Sefton BM. 1980. Transforming gene product of Rous sarcoma virus phosphorylates tyrosine. Proc. Natl. Acad. Sci. U. S. A. 77:1311–1315. 10.1073/pnas.77.3.1311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. 2002. The protein kinase complement of the human genome. Science 298:1912–1934. 10.1126/science.1075762 [DOI] [PubMed] [Google Scholar]
  • 6.Ullrich A, Schlessinger J. 1990. Signal transduction by receptors with tyrosine kinase activity. Cell 61:203–212. 10.1016/0092-8674(90)90801-K [DOI] [PubMed] [Google Scholar]
  • 7.Lemmon MA, Schlessinger J. 2010. Cell signaling by receptor tyrosine kinases. Cell 141:1117–1134. 10.1016/j.cell.2010.06.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schlessinger J, Lemmon MA. 2003. SH2 and PTB domains in tyrosine kinase signaling. Sci. STKE 2003:RE12. 10.1126/stke.2003.191.re12 [DOI] [PubMed] [Google Scholar]
  • 9.Kirkin V, Dikic I. 2007. Role of ubiquitin- and Ubl-binding proteins in cell signaling. Curr. Opin. Cell Biol. 19:199–205. 10.1016/j.ceb.2007.02.002 [DOI] [PubMed] [Google Scholar]
  • 10.Del Rosario AM, White FM. 2010. Quantifying oncogenic phosphotyrosine signaling networks through systems biology. Curr. Opin. Genet. Dev. 20:23–30. 10.1016/j.gde.2009.12.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Nakahara M, Isozaki K, Hirota S, Miyagawa J, Hase-Sawada N, Taniguchi M, Nishida T, Kanayama S, Kitamura Y, Shinomura Y, Matsuzawa Y. 1998. A novel gain-of-function mutation of c-kit gene in gastrointestinal stromal tumors. Gastroenterology 115:1090–1095. 10.1016/S0016-5085(98)70079-4 [DOI] [PubMed] [Google Scholar]
  • 12.Yamamoto Y, Kiyoi H, Nakano Y, Suzuki R, Kodera Y, Miyawaki S, Asou N, Kuriyama K, Yagasaki F, Shimazaki C, Akiyama H, Saito K, Nishimura M, Motoji T, Shinagawa K, Takeshita A, Saito H, Ueda R, Ohno R, Naoe T. 2001. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood 97:2434–2439. 10.1182/blood.V97.8.2434 [DOI] [PubMed] [Google Scholar]
  • 13.Corless CL, Barnett CM, Heinrich MC. 2011. Gastrointestinal stromal tumours: origin and molecular oncology. Nat. Rev. Cancer 11:865–878. 10.1038/nrc3143 [DOI] [PubMed] [Google Scholar]
  • 14.da Cunha Santos G, Shepherd FA, Tsao MS. 2011. EGFR mutations and lung cancer. Annu. Rev. Pathol. 6:49–69. 10.1146/annurev-pathol-011110-130206 [DOI] [PubMed] [Google Scholar]
  • 15.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. 2007. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 448:561–566. 10.1038/nature05945 [DOI] [PubMed] [Google Scholar]
  • 16.Sozzi G, Bongarzone I, Miozzo M, Borrello MG, Blutti MG, Pilotti S, Della Porta G, Pierotti MA. 1994. A t(10;17) translocation creates the RET/PTC2 chimeric transforming sequence in papillary thyroid carcinoma. Genes Chromosomes Cancer 9:244–250. 10.1002/gcc.2870090404 [DOI] [PubMed] [Google Scholar]
  • 17.Rowley JD. 1973. A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature 243:290–293. 10.1038/243290a0 [DOI] [PubMed] [Google Scholar]
  • 18.Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. 1987. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235:177–182. 10.1126/science.3798106 [DOI] [PubMed] [Google Scholar]
  • 19.Herman SE, Gordon AL, Hertlein E, Ramanunni A, Zhang X, Jaglowski S, Flynn J, Jones J, Blum KA, Buggy JJ, Hamdy A, Johnson AJ, Byrd JC. 2011. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood 117:6287–6296. 10.1182/blood-2011-01-328484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Cai H, Babic I, Wei X, Huang J, Witte ON. 2011. Invasive prostate carcinoma driven by c-Src and androgen receptor synergy. Cancer Res. 71:862–872. 10.1158/0008-5472.CAN-10-1605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Memarzadeh S, Xin L, Mulholland DJ, Mansukhani A, Wu H, Teitell MA, Witte ON. 2007. Enhanced paracrine FGF10 expression promotes formation of multifocal prostate adenocarcinoma and an increase in epithelial androgen receptor. Cancer Cell 12:572–585. 10.1016/j.ccr.2007.11.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Cartwright CA, Kamps MP, Meisler AI, Pipas JM, Eckhart W. 1989. pp60c-src activation in human colon carcinoma. J. Clin. Invest. 83:2025–2033. 10.1172/JCI114113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Jhiang SM. 2000. The RET proto-oncogene in human cancers. Oncogene 19:5590–5597. 10.1038/sj.onc.1203857 [DOI] [PubMed] [Google Scholar]
  • 24.Santoro M, Carlomagno F, Romano A, Bottaro DP, Dathan NA, Grieco M, Fusco A, Vecchio G, Matoskova B, Kraus MH, di Fiore PP. 1995. Activation of RET as a dominant transforming gene by germline mutations of MEN2A and MEN2B. Science 267:381–383. 10.1126/science.7824936 [DOI] [PubMed] [Google Scholar]
  • 25.Maleddu A, Pantaleo MA, Nannini M, Biasco G. 2011. The role of mutational analysis of KIT and PDGFRA in gastrointestinal stromal tumors in a clinical setting. J. Transl. Med. 9:75. 10.1186/1479-5876-9-75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Zhi X, Zhou X, Wang W, Xu Z. 2013. Practical role of mutation analysis for imatinib treatment in patients with advanced gastrointestinal stromal tumors: a meta-analysis. PLoS One 8:e79275. 10.1371/journal.pone.0079275 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mitelman F, Johansson B, Mertens F. 2004. Fusion genes and rearranged genes as a linear function of chromosome aberrations in cancer. Nat. Genet. 36:331–334. 10.1038/ng1335 [DOI] [PubMed] [Google Scholar]
  • 28.Shaw AT, Hsu PP, Awad MM, Engelman JA. 2013. Tyrosine kinase gene rearrangements in epithelial malignancies. Nat. Rev. Cancer 13:772–787. 10.1038/nrc3612 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Seo JS, Ju YS, Lee WC, Shin JY, Lee JK, Bleazard T, Lee J, Jung YJ, Kim JO, Yu SB, Kim J, Lee ER, Kang CH, Park IK, Rhee H, Lee SH, Kim JI, Kang JH, Kim YT. 2012. The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Res. 22:2109–2119. 10.1101/gr.145144.112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Nagar B, Hantschel O, Young MA, Scheffzek K, Veach D, Bornmann W, Clarkson B, Superti-Furga G, Kuriyan J. 2003. Structural basis for the autoinhibition of c-Abl tyrosine kinase. Cell 112:859–871. 10.1016/S0092-8674(03)00194-6 [DOI] [PubMed] [Google Scholar]
  • 31.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. 2007. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316:1039–1043. 10.1126/science.1141478 [DOI] [PubMed] [Google Scholar]
  • 32.Shibuya M, Yokota J, Ueyama Y. 1985. Amplification and expression of a cellular oncogene (c-myc) in human gastric adenocarcinoma cells. Mol. Cell. Biol. 5:414–418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Xing WR, Gilchrist KW, Harris CP, Samson W, Meisner LF. 1996. FISH detection of HER-2/neu oncogene amplification in early onset breast cancer. Breast Cancer Res. Treat. 39:203–212. 10.1007/BF01806187 [DOI] [PubMed] [Google Scholar]
  • 34.Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L. 2001. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344:783–792. 10.1056/NEJM200103153441101 [DOI] [PubMed] [Google Scholar]
  • 35.Rawlings DJ, Scharenberg AM, Park H, Wahl MI, Lin S, Kato RM, Fluckiger AC, Witte ON, Kinet JP. 1996. Activation of BTK by a phosphorylation mechanism initiated by SRC family kinases. Science 271:822–825. 10.1126/science.271.5250.822 [DOI] [PubMed] [Google Scholar]
  • 36.Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen I, Bycott PW, Baum CM, Figlin RA. 2007. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N. Engl. J. Med. 356:115–124. 10.1056/NEJMoa065044 [DOI] [PubMed] [Google Scholar]
  • 37.Rubbi L, Titz B, Brown L, Galvan E, Komisopoulou E, Chen SS, Low T, Tahmasian M, Skaggs B, Muschen M, Pellegrini M, Graeber TG. 2011. Global phosphoproteomics reveals crosstalk between Bcr-Abl and negative feedback mechanisms controlling Src signaling. Sci. Signal. 4:ra18. 10.1126/scisignal.2001314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Julien SG, Dube N, Hardy S, Tremblay ML. 2011. Inside the human cancer tyrosine phosphatome. Nat. Rev. Cancer 11:35–49. 10.1038/nrc2980 [DOI] [PubMed] [Google Scholar]
  • 39.Peschard P, Park M. 2003. Escape from Cbl-mediated downregulation: a recurrent theme for oncogenic deregulation of receptor tyrosine kinases. Cancer Cell 3:519–523. 10.1016/S1535-6108(03)00136-3 [DOI] [PubMed] [Google Scholar]
  • 40.Park H, Wahl MI, Afar DE, Turck CW, Rawlings DJ, Tam C, Scharenberg AM, Kinet JP, Witte ON. 1996. Regulation of Btk function by a major autophosphorylation site within the SH3 domain. Immunity 4:515–525. 10.1016/S1074-7613(00)80417-3 [DOI] [PubMed] [Google Scholar]
  • 41.Li T, Tsukada S, Satterthwaite A, Havlik MH, Park H, Takatsu K, Witte ON. 1995. Activation of Bruton's tyrosine kinase (BTK) by a point mutation in its pleckstrin homology (PH) domain. Immunity 2:451–460. 10.1016/1074-7613(95)90026-8 [DOI] [PubMed] [Google Scholar]
  • 42.Li Z, Wahl MI, Eguinoa A, Stephens LR, Hawkins PT, Witte ON. 1997. Phosphatidylinositol 3-kinase-gamma activates Bruton's tyrosine kinase in concert with Src family kinases. Proc. Natl. Acad. Sci. U. S. A. 94:13820–13825. 10.1073/pnas.94.25.13820 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Clark SS, McLaughlin J, Timmons M, Pendergast AM, Ben-Neriah Y, Dow LW, Crist W, Rovera G, Smith SD, Witte ON. 1988. Expression of a distinctive BCR-ABL oncogene in Ph1-positive acute lymphocytic leukemia (ALL). Science 239:775–777. 10.1126/science.3422516 [DOI] [PubMed] [Google Scholar]
  • 44.Lugo TG, Pendergast AM, Muller AJ, Witte ON. 1990. Tyrosine kinase activity and transformation potency of bcr-abl oncogene products. Science 247:1079–1082. 10.1126/science.2408149 [DOI] [PubMed] [Google Scholar]
  • 45.Satterthwaite AB, Willis F, Kanchanastit P, Fruman D, Cantley LC, Helgason CD, Humphries RK, Lowell CA, Simon M, Leitges M, Tarakhovsky A, Tedder TF, Lesche R, Wu H, Witte ON. 2000. A sensitized genetic system for the analysis of murine B lymphocyte signal transduction pathways dependent on Bruton's tyrosine kinase. Proc. Natl. Acad. Sci. U. S. A. 97:6687–6692. 10.1073/pnas.110146697 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Konopka JB, Watanabe SM, Singer JW, Collins SJ, Witte ON. 1985. Cell lines and clinical isolates derived from Ph1-positive chronic myelogenous leukemia patients express c-abl proteins with a common structural alteration. Proc. Natl. Acad. Sci. U. S. A. 82:1810–1814. 10.1073/pnas.82.6.1810 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ben-Neriah Y, Daley GQ, Mes-Masson AM, Witte ON, Baltimore D. 1986. The chronic myelogenous leukemia-specific P210 protein is the product of the bcr/abl hybrid gene. Science 233:212–214. 10.1126/science.3460176 [DOI] [PubMed] [Google Scholar]
  • 48.McLaughlin J, Chianese E, Witte ON. 1987. In vitro transformation of immature hematopoietic cells by the P210 BCR/ABL oncogene product of the Philadelphia chromosome. Proc. Natl. Acad. Sci. U. S. A. 84:6558–6562. 10.1073/pnas.84.18.6558 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tsukada S, Saffran DC, Rawlings DJ, Parolini O, Allen RC, Klisak I, Sparkes RS, Kubagawa H, Mohandas T, Quan S, Belmont JW, Cooper MD, Conley ME, Witte ON. 1993. Deficient expression of a B cell cytoplasmic tyrosine kinase in human X-linked agammaglobulinemia. Cell 72:279–290. 10.1016/0092-8674(93)90667-F [DOI] [PubMed] [Google Scholar]
  • 50.Koleske AJ, Gifford AM, Scott ML, Nee M, Bronson RT, Miczek KA, Baltimore D. 1998. Essential roles for the Abl and Arg tyrosine kinases in neurulation. Neuron 21:1259–1272. 10.1016/S0896-6273(00)80646-7 [DOI] [PubMed] [Google Scholar]
  • 51.Schwartzberg PL, Stall AM, Hardin JD, Bowdish KS, Humaran T, Boast S, Harbison ML, Robertson EJ, Goff SP. 1991. Mice homozygous for the ablm1 mutation show poor viability and depletion of selected B and T cell populations. Cell 65:1165–1175. 10.1016/0092-8674(91)90012-N [DOI] [PubMed] [Google Scholar]
  • 52.Tybulewicz VL, Crawford CE, Jackson PK, Bronson RT, Mulligan RC. 1991. Neonatal lethality and lymphopenia in mice with a homozygous disruption of the c-abl proto-oncogene. Cell 65:1153–1163. 10.1016/0092-8674(91)90011-M [DOI] [PubMed] [Google Scholar]
  • 53.Qiu Z, Cang Y, Goff SP. 2010. c-Abl tyrosine kinase regulates cardiac growth and development. Proc. Natl. Acad. Sci. U. S. A. 107:1136–1141. 10.1073/pnas.0913131107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chislock EM, Ring C, Pendergast AM. 2013. Abl kinases are required for vascular function, Tie2 expression, and angiopoietin-1-mediated survival. Proc. Natl. Acad. Sci. U. S. A. 110:12432–12437. 10.1073/pnas.1304188110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sirvent A, Benistant C, Roche S. 2008. Cytoplasmic signalling by the c-Abl tyrosine kinase in normal and cancer cells. Biol. Cell 100:617–631. 10.1042/BC20080020 [DOI] [PubMed] [Google Scholar]
  • 56.Hantschel O, Superti-Furga G. 2004. Regulation of the c-Abl and Bcr-Abl tyrosine kinases. Nat. Rev. Mol. Cell Biol. 5:33–44. 10.1038/nrm1280 [DOI] [PubMed] [Google Scholar]
  • 57.Sawyers CL, McLaughlin J, Goga A, Havlik M, Witte O. 1994. The nuclear tyrosine kinase c-Abl negatively regulates cell growth. Cell 77:121–131. 10.1016/0092-8674(94)90240-2 [DOI] [PubMed] [Google Scholar]
  • 58.Wetzler M, Talpaz M, Van Etten RA, Hirsh-Ginsberg C, Beran M, Kurzrock R. 1993. Subcellular localization of Bcr, Abl, and Bcr-Abl proteins in normal and leukemic cells and correlation of expression with myeloid differentiation. J. Clin. Invest. 92:1925–1939. 10.1172/JCI116786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Taagepera S, McDonald D, Loeb JE, Whitaker LL, McElroy AK, Wang JY, Hope TJ. 1998. Nuclear-cytoplasmic shuttling of C-ABL tyrosine kinase. Proc. Natl. Acad. Sci. U. S. A. 95:7457–7462. 10.1073/pnas.95.13.7457 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jackson P, Baltimore D. 1989. N-terminal mutations activate the leukemogenic potential of the myristoylated form of c-abl. EMBO J. 8:449–456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Jackson PK, Paskind M, Baltimore D. 1993. Mutation of a phenylalanine conserved in SH3-containing tyrosine kinases activates the transforming ability of c-Abl. Oncogene 8:1943–1956 [PubMed] [Google Scholar]
  • 62.Van Etten RA, Debnath J, Zhou H, Casasnovas JM. 1995. Introduction of a loss-of-function point mutation from the SH3 region of the Caenorhabditis elegans sem-5 gene activates the transforming ability of c-abl in vivo and abolishes binding of proline-rich ligands in vitro. Oncogene 10:1977–1988 [PubMed] [Google Scholar]
  • 63.Sirvent A, Boureux A, Simon V, Leroy C, Roche S. 2007. The tyrosine kinase Abl is required for Src-transforming activity in mouse fibroblasts and human breast cancer cells. Oncogene 26:7313–7323. 10.1038/sj.onc.1210543 [DOI] [PubMed] [Google Scholar]
  • 64.Srinivasan D, Sims JT, Plattner R. 2008. Aggressive breast cancer cells are dependent on activated Abl kinases for proliferation, anchorage-independent growth and survival. Oncogene 27:1095–1105. 10.1038/sj.onc.1210714 [DOI] [PubMed] [Google Scholar]
  • 65.Srinivasan D, Plattner R. 2006. Activation of Abl tyrosine kinases promotes invasion of aggressive breast cancer cells. Cancer Res. 66:5648–5655. 10.1158/0008-5472.CAN-06-0734 [DOI] [PubMed] [Google Scholar]
  • 66.Schindler T, Bornmann W, Pellicena P, Miller WT, Clarkson B, Kuriyan J. 2000. Structural mechanism for STI-571 inhibition of Abelson tyrosine kinase. Science 289:1938–1942. 10.1126/science.289.5486.1938 [DOI] [PubMed] [Google Scholar]
  • 67.Nowell PC, Hungerford DA. 1960. Chromosome studies on normal and leukemic human leukocytes. J. Natl. Cancer Inst. 25:85–109 [PubMed] [Google Scholar]
  • 68.de Klein A, van Kessel AG, Grosveld G, Bartram CR, Hagemeijer A, Bootsma D, Spurr NK, Heisterkamp N, Groffen J, Stephenson JR. 1982. A cellular oncogene is translocated to the Philadelphia chromosome in chronic myelocytic leukaemia. Nature 300:765–767. 10.1038/300765a0 [DOI] [PubMed] [Google Scholar]
  • 69.Heisterkamp N, Stam K, Groffen J, de Klein A, Grosveld G. 1985. Structural organization of the bcr gene and its role in the Ph' translocation. Nature 315:758–761. 10.1038/315758a0 [DOI] [PubMed] [Google Scholar]
  • 70.McWhirter JR, Galasso DL, Wang JY. 1993. A coiled-coil oligomerization domain of Bcr is essential for the transforming function of Bcr-Abl oncoproteins. Mol. Cell. Biol. 13:7587–7595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Pendergast AM, Quilliam LA, Cripe LD, Bassing CH, Dai Z, Li N, Batzer A, Rabun KM, Der CJ, Schlessinger J, Gishizky ML. 1993. BCR-ABL-induced oncogenesis is mediated by direct interaction with the SH2 domain of the GRB-2 adaptor protein. Cell 75:175–185. 10.1016/0092-8674(93)90689-N [DOI] [PubMed] [Google Scholar]
  • 72.Kelber JA, Klemke RL. 2010. PEAK1, a novel kinase target in the fight against cancer. Oncotarget 1:219–223 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kroiher M, Miller MA, Steele RE. 2001. Deceiving appearances: signaling by “dead” and “fractured” receptor protein-tyrosine kinases. Bioessays 23:69–76. [DOI] [PubMed] [Google Scholar]
  • 74.Buchdunger E, Zimmermann J, Mett H, Meyer T, Muller M, Druker BJ, Lydon NB. 1996. Inhibition of the Abl protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyrimidine derivative. Cancer Res. 56:100–104 [PubMed] [Google Scholar]
  • 75.Carroll M, Ohno-Jones S, Tamura S, Buchdunger E, Zimmermann J, Lydon NB, Gilliland DG, Druker BJ. 1997. CGP 57148, a tyrosine kinase inhibitor, inhibits the growth of cells expressing BCR-ABL, TEL-ABL, and TEL-PDGFR fusion proteins. Blood 90:4947–4952 [PubMed] [Google Scholar]
  • 76.Ross DW, Brunning RD, Kantarjian HM, Koeffler HP, Ozer H. 1993. A proposed staging system for chronic myeloid leukemia. Cancer 71:3788–3791. [DOI] [PubMed] [Google Scholar]
  • 77.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, IRIS Investigators. 2006. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N. Engl. J. Med. 355:2408–2417. 10.1056/NEJMoa062867 [DOI] [PubMed] [Google Scholar]
  • 78.Kantarjian H, O'Brien S, Jabbour E, Garcia-Manero G, Quintas-Cardama A, Shan J, Rios MB, Ravandi F, Faderl S, Kadia T, Borthakur G, Huang X, Champlin R, Talpaz M, Cortes J. 2012. Improved survival in chronic myeloid leukemia since the introduction of imatinib therapy: a single-institution historical experience. Blood 119:1981–1987. 10.1182/blood-2011-08-358135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Gambacorti-Passerini C, Antolini L, Mahon FX, Guilhot F, Deininger M, Fava C, Nagler A, Della Casa CM, Morra E, Abruzzese E, D'Emilio A, Stagno F, le Coutre P, Hurtado-Monroy R, Santini V, Martino B, Pane F, Piccin A, Giraldo P, Assouline S, Durosinmi MA, Leeksma O, Pogliani EM, Puttini M, Jang E, Reiffers J, Valsecchi MG, Kim DW. 2011. Multicenter independent assessment of outcomes in chronic myeloid leukemia patients treated with imatinib. J. Natl. Cancer Inst. 103:553–561. 10.1093/jnci/djr060 [DOI] [PubMed] [Google Scholar]
  • 80.Shah NP, Nicoll JM, Nagar B, Gorre ME, Paquette RL, Kuriyan J, Sawyers CL. 2002. 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 2:117–125. 10.1016/S1535-6108(02)00096-X [DOI] [PubMed] [Google Scholar]
  • 81.Illmer T, Schaich M, Platzbecker U, Freiberg-Richter J, Oelschlagel U, von Bonin M, Pursche S, Bergemann T, Ehninger G, Schleyer E. 2004. P-glycoprotein-mediated drug efflux is a resistance mechanism of chronic myelogenous leukemia cells to treatment with imatinib mesylate. Leukemia 18:401–408. 10.1038/sj.leu.2403257 [DOI] [PubMed] [Google Scholar]
  • 82.Hochhaus A, Kreil S, Corbin AS, La Rosee P, Muller MC, Lahaye T, Hanfstein B, Schoch C, Cross NC, Berger U, Gschaidmeier H, Druker BJ, Hehlmann R. 2002. Molecular and chromosomal mechanisms of resistance to imatinib (STI571) therapy. Leukemia 16:2190–2196. 10.1038/sj.leu.2402741 [DOI] [PubMed] [Google Scholar]
  • 83.Cortes JE, Kim DW, Pinilla-Ibarz J, le Coutre P, Paquette R, Chuah C, Nicolini FE, Apperley JF, Khoury HJ, Talpaz M, DiPersio J, DeAngelo DJ, Abruzzese E, Rea D, Baccarani M, Muller MC, Gambacorti-Passerini C, Wong S, Lustgarten S, Rivera VM, Clackson T, Turner CD, Haluska FG, Guilhot F, Deininger MW, Hochhaus A, Hughes T, Goldman JM, Shah NP, Kantarjian H, PACE Investigators. 2013. A phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias. N. Engl. J. Med. 369:1783–1796. 10.1056/NEJMoa1306494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cortes JE, Kim DW, Kantarjian HM, Brummendorf TH, Dyagil I, Griskevicius L, Malhotra H, Powell C, Gogat K, Countouriotis AM, Gambacorti-Passerini C. 2012. Bosutinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia: results from the BELA trial. J. Clin. Oncol. 30:3486–3492. 10.1200/JCO.2011.38.7522 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Saglio G, Kim DW, Issaragrisil S, le Coutre P, Etienne G, Lobo C, Pasquini R, Clark RE, Hochhaus A, Hughes TP, Gallagher N, Hoenekopp A, Dong M, Haque A, Larson RA, Kantarjian HM, ENESTnd Investigators. 2010. Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N. Engl. J. Med. 362:2251–2259. 10.1056/NEJMoa0912614 [DOI] [PubMed] [Google Scholar]
  • 86.Talpaz M, Shah NP, Kantarjian H, Donato N, Nicoll J, Paquette R, Cortes J, O'Brien S, Nicaise C, Bleickardt E, Blackwood-Chirchir MA, Iyer V, Chen TT, Huang F, Decillis AP, Sawyers CL. 2006. Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. N. Engl. J. Med. 354:2531–2541. 10.1056/NEJMoa055229 [DOI] [PubMed] [Google Scholar]
  • 87.Azam M, Seeliger MA, Gray NS, Kuriyan J, Daley GQ. 2008. Activation of tyrosine kinases by mutation of the gatekeeper threonine. Nat. Struct. Mol. Biol. 15:1109–1118. 10.1038/nsmb.1486 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Huang WS, Metcalf CA, Sundaramoorthi R, Wang Y, Zou D, Thomas RM, Zhu X, Cai L, Wen D, Liu S, Romero J, Qi J, Chen I, Banda G, Lentini SP, Das S, Xu Q, Keats J, Wang F, Wardwell S, Ning Y, Snodgrass JT, Broudy MI, Russian K, Zhou T, Commodore L, Narasimhan NI, Mohemmad QK, Iuliucci J, Rivera VM, Dalgarno DC, Sawyer TK, Clackson T, Shakespeare WC. 2010. Discovery of 3-[2-(imidazo[1,2-b]pyridazin-3-yl)ethynyl]-4-methyl-N-{4-[(4-methylpiperazin-1-yl)methyl]-3-(trifluoromethyl)phenyl}benzamide (AP24534), a potent, orally active pan-inhibitor of breakpoint cluster region-Abelson (BCR-ABL) kinase including the T315I gatekeeper mutant. J. Med. Chem. 53:4701–4719. 10.1021/jm100395q [DOI] [PubMed] [Google Scholar]
  • 89.Zhou T, Commodore L, Huang WS, Wang Y, Thomas M, Keats J, Xu Q, Rivera VM, Shakespeare WC, Clackson T, Dalgarno DC, Zhu X. 2011. Structural mechanism of the Pan-BCR-ABL inhibitor ponatinib (AP24534): lessons for overcoming kinase inhibitor resistance. Chem. Biol. Drug Des. 77:1–11. 10.1111/j.1747-0285.2010.01054.x [DOI] [PubMed] [Google Scholar]
  • 90.Vetrie D, Vorechovsky I, Sideras P, Holland J, Davies A, Flinter F, Hammarstrom L, Kinnon C, Levinsky R, Bobrow M, Smith CIE, Bentley DR. 1993. The gene involved in X-linked agammaglobulinaemia is a member of the src family of protein-tyrosine kinases. Nature 361:226–233. 10.1038/361226a0 [DOI] [PubMed] [Google Scholar]
  • 91.Valiaho J, Smith CI, Vihinen M. 2006. BTKbase: the mutation database for X-linked agammaglobulinemia. Hum. Mutat. 27:1209–1217. 10.1002/humu.20410 [DOI] [PubMed] [Google Scholar]
  • 92.Conley ME. 1992. Molecular approaches to analysis of X-linked immunodeficiencies. Annu. Rev. Immunol. 10:215–238. 10.1146/annurev.iy.10.040192.001243 [DOI] [PubMed] [Google Scholar]
  • 93.Rawlings DJ, Saffran DC, Tsukada S, Largaespada DA, Grimaldi JC, Cohen L, Mohr RN, Bazan JF, Howard M, Copeland NG, Jenkins NA, Witte ON. 1993. Mutation of unique region of Bruton's tyrosine kinase in immunodeficient XID mice. Science 261:358–361. 10.1126/science.8332901 [DOI] [PubMed] [Google Scholar]
  • 94.Thomas JD, Sideras P, Smith CI, Vorechovsky I, Chapman V, Paul WE. 1993. Colocalization of X-linked agammaglobulinemia and X-linked immunodeficiency genes. Science 261:355–358. 10.1126/science.8332900 [DOI] [PubMed] [Google Scholar]
  • 95.Maas A, Hendriks RW. 2001. Role of Bruton's tyrosine kinase in B cell development. Dev. Immunol. 8:171–181. 10.1155/2001/28962 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Rawlings DJ, Witte ON. 1995. The Btk subfamily of cytoplasmic tyrosine kinases: structure, regulation and function. Semin. Immunol. 7:237–246. 10.1006/smim.1995.0028 [DOI] [PubMed] [Google Scholar]
  • 97.Hitoshi Y, Sonoda E, Kikuchi Y, Yonehara S, Nakauchi H, Takatsu K. 1993. IL-5 receptor positive B cells, but not eosinophils, are functionally and numerically influenced in mice carrying the X-linked immune defect. Int. Immunol. 5:1183–1190. 10.1093/intimm/5.9.1183 [DOI] [PubMed] [Google Scholar]
  • 98.Go NF, Castle BE, Barrett R, Kastelein R, Dang W, Mosmann TR, Moore KW, Howard M. 1990. Interleukin 10, a novel B cell stimulatory factor: unresponsiveness of X chromosome-linked immunodeficiency B cells. J. Exp. Med. 172:1625–1631. 10.1084/jem.172.6.1625 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hasbold J, Klaus GG. 1994. B cells from CBA/N mice do not proliferate following ligation of CD40. Eur. J. Immunol. 24:152–157. 10.1002/eji.1830240123 [DOI] [PubMed] [Google Scholar]
  • 100.de Weers M, Mensink RG, Kraakman ME, Schuurman RK, Hendriks RW. 1994. Mutation analysis of the Bruton's tyrosine kinase gene in X-linked agammaglobulinemia: identification of a mutation which affects the same codon as is altered in immunodeficient xid mice. Hum. Mol. Genet. 3:161–166. 10.1093/hmg/3.1.161 [DOI] [PubMed] [Google Scholar]
  • 101.Qin X, Jiang LP, Tang XM, Wang M, Liu EM, Zhao XD. 2013. Clinical features and mutation analysis of X-linked agammaglobulinemia in 20 Chinese patients. World J. Pediatr. 9:273–277. 10.1007/s12519-013-0400-x [DOI] [PubMed] [Google Scholar]
  • 102.Conley ME, Mathias D, Treadaway J, Minegishi Y, Rohrer J. 1998. Mutations in btk in patients with presumed X-linked agammaglobulinemia. Am. J. Hum. Genet. 62:1034–1043. 10.1086/301828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Li T, Rawlings DJ, Park H, Kato RM, Witte ON, Satterthwaite AB. 1997. Constitutive membrane association potentiates activation of Bruton tyrosine kinase. Oncogene 15:1375–1383. 10.1038/sj.onc.1201308 [DOI] [PubMed] [Google Scholar]
  • 104.Takata M, Kurosaki T. 1996. A role for Bruton's tyrosine kinase in B cell antigen receptor-mediated activation of phospholipase C-gamma 2. J. Exp. Med. 184:31–40. 10.1084/jem.184.1.31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Tan SL, Liao C, Lucas MC, Stevenson C, DeMartino JA. 2013. Targeting the SYK-BTK axis for the treatment of immunological and hematological disorders: recent progress and therapeutic perspectives. Pharmacol. Ther. 138:294–309. 10.1016/j.pharmthera.2013.02.001 [DOI] [PubMed] [Google Scholar]
  • 106.Zwick C, Fadle N, Regitz E, Kemele M, Stilgenbauer S, Buhler A, Pfreundschuh M, Preuss KD. 2013. Autoantigenic targets of B-cell receptors derived from chronic lymphocytic leukemias bind to and induce proliferation of leukemic cells. Blood 121:4708–4717. 10.1182/blood-2012-08-447904 [DOI] [PubMed] [Google Scholar]
  • 107.Davis RE, Ngo VN, Lenz G, Tolar P, Young RM, Romesser PB, Kohlhammer H, Lamy L, Zhao H, Yang Y, Xu W, Shaffer AL, Wright G, Xiao W, Powell J, Jiang JK, Thomas CJ, Rosenwald A, Ott G, Muller-Hermelink HK, Gascoyne RD, Connors JM, Johnson NA, Rimsza LM, Campo E, Jaffe ES, Wilson WH, Delabie J, Smeland EB, Fisher RI, Braziel RM, Tubbs RR, Cook JR, Weisenburger DD, Chan WC, Pierce SK, Staudt LM. 2010. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature 463:88–92. 10.1038/nature08638 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wang ML, Rule S, Martin P, Goy A, Auer R, Kahl BS, Jurczak W, Advani RH, Romaguera JE, Williams ME, Barrientos JC, Chmielowska E, Radford J, Stilgenbauer S, Dreyling M, Jedrzejczak WW, Johnson P, Spurgeon SE, Li L, Zhang L, Newberry K, Ou Z, Cheng N, Fang B, McGreivy J, Clow F, Buggy JJ, Chang BY, Beaupre DM, Kunkel LA, Blum KA. 2013. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N. Engl. J. Med. 369:507–516. 10.1056/NEJMoa1306220 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Treon SP, Tripsas CK, Yang G, Cao Y, Xu L, Hunter Z, Cropper SJ, Mostyn P, Meid K, Warren D, Patterson C, Varma G, Laubach JP, Paba-Prada CE, Kunsman J, Ghobrial IM, Kanan S, Advani RH, Palomba ML. 2013. A prospective multicenter study of the Bruton's tyrosine kinase inhibitor ibrutinib in patients with relapsed or refractory Waldenstrom's macroglobulinemia, abstr 251. 55th ASH Annu. Meet. Expo. [Google Scholar]
  • 110.Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, Blum KA, Grant B, Sharman JP, Coleman M, Wierda WG, Jones JA, Zhao W, Heerema NA, Johnson AJ, Sukbuntherng J, Chang BY, Clow F, Hedrick E, Buggy JJ, James DF, O'Brien S. 2013. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 369:32–42. 10.1056/NEJMoa1215637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Drake JM, Graham NA, Stoyanova T, Sedghi A, Goldstein AS, Cai H, Smith DA, Zhang H, Komisopoulou E, Huang J, Graeber TG, Witte ON. 2012. Oncogene-specific activation of tyrosine kinase networks during prostate cancer progression. Proc. Natl. Acad. Sci. U. S. A. 109:1643–1648. 10.1073/pnas.1120985109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Rush J, Moritz A, Lee KA, Guo A, Goss VL, Spek EJ, Zhang H, Zha XM, Polakiewicz RD, Comb MJ. 2005. Immunoaffinity profiling of tyrosine phosphorylation in cancer cells. Nat. Biotechnol. 23:94–101. 10.1038/nbt1046 [DOI] [PubMed] [Google Scholar]
  • 113.Guha U, Chaerkady R, Marimuthu A, Patterson AS, Kashyap MK, Harsha HC, Sato M, Bader JS, Lash AE, Minna JD, Pandey A, Varmus HE. 2008. Comparisons of tyrosine phosphorylated proteins in cells expressing lung cancer-specific alleles of EGFR and KRAS. Proc. Natl. Acad. Sci. U. S. A. 105:14112–14117. 10.1073/pnas.0806158105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Hahn CK, Berchuck JE, Ross KN, Kakoza RM, Clauser K, Schinzel AC, Ross L, Galinsky I, Davis TN, Silver SJ, Root DE, Stone RM, DeAngelo DJ, Carroll M, Hahn WC, Carr SA, Golub TR, Kung AL, Stegmaier K. 2009. Proteomic and genetic approaches identify Syk as an AML target. Cancer Cell 16:281–294. 10.1016/j.ccr.2009.08.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Guo A, Villen J, Kornhauser J, Lee KA, Stokes MP, Rikova K, Possemato A, Nardone J, Innocenti G, Wetzel R, Wang Y, MacNeill J, Mitchell J, Gygi SP, Rush J, Polakiewicz RD, Comb MJ. 2008. Signaling networks assembled by oncogenic EGFR and c-Met. Proc. Natl. Acad. Sci. U. S. A. 105:692–697. 10.1073/pnas.0707270105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Rikova K, Guo A, Zeng Q, Possemato A, Yu J, Haack H, Nardone J, Lee K, Reeves C, Li Y, Hu Y, Tan Z, Stokes M, Sullivan L, Mitchell J, Wetzel R, Macneill J, Ren JM, Yuan J, Bakalarski CE, Villen J, Kornhauser JM, Smith B, Li D, Zhou X, Gygi SP, Gu TL, Polakiewicz RD, Rush J, Comb MJ. 2007. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 131:1190–1203. 10.1016/j.cell.2007.11.025 [DOI] [PubMed] [Google Scholar]
  • 117.Hochgrafe F, Zhang L, O'Toole SA, Browne BC, Pinese M, Porta Cubas A, Lehrbach GM, Croucher DR, Rickwood D, Boulghourjian A, Shearer R, Nair R, Swarbrick A, Faratian D, Mullen P, Harrison DJ, Biankin AV, Sutherland RL, Raftery MJ, Daly RJ. 2010. Tyrosine phosphorylation profiling reveals the signaling network characteristics of basal breast cancer cells. Cancer Res. 70:9391–9401. 10.1158/0008-5472.CAN-10-0911 [DOI] [PubMed] [Google Scholar]
  • 118.MacKeigan JP, Murphy LO, Blenis J. 2005. Sensitized RNAi screen of human kinases and phosphatases identifies new regulators of apoptosis and chemoresistance. Nat. Cell Biol. 7:591–600. 10.1038/ncb1258 [DOI] [PubMed] [Google Scholar]
  • 119.Scholl C, Frohling S, Dunn IF, Schinzel AC, Barbie DA, Kim SY, Silver SJ, Tamayo P, Wadlow RC, Ramaswamy S, Dohner K, Bullinger L, Sandy P, Boehm JS, Root DE, Jacks T, Hahn WC, Gilliland DG. 2009. Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells. Cell 137:821–834. 10.1016/j.cell.2009.03.017 [DOI] [PubMed] [Google Scholar]
  • 120.Tyner JW, Walters DK, Willis SG, Luttropp M, Oost J, Loriaux M, Erickson H, Corbin AS, O'Hare T, Heinrich MC, Deininger MW, Druker BJ. 2008. RNAi screening of the tyrosine kinome identifies therapeutic targets in acute myeloid leukemia. Blood 111:2238–2245. 10.1182/blood-2007-06-097253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Arora S, Bisanz KM, Peralta LA, Basu GD, Choudhary A, Tibes R, Azorsa DO. 2010. RNAi screening of the kinome identifies modulators of cisplatin response in ovarian cancer cells. Gynecol. Oncol. 118:220–227. 10.1016/j.ygyno.2010.05.006 [DOI] [PubMed] [Google Scholar]
  • 122.Giamas G, Filipovic A, Jacob J, Messier W, Zhang H, Yang D, Zhang W, Shifa BA, Photiou A, Tralau-Stewart C, Castellano L, Green AR, Coombes RC, Ellis IO, Ali S, Lenz HJ, Stebbing J. 2011. Kinome screening for regulators of the estrogen receptor identifies LMTK3 as a new therapeutic target in breast cancer. Nat. Med. 17:715–719. 10.1038/nm.2351 [DOI] [PubMed] [Google Scholar]
  • 123.Lu Y, Muller M, Smith D, Dutta B, Komurov K, Iadevaia S, Ruths D, Tseng JT, Yu S, Yu Q, Nakhleh L, Balazsi G, Donnelly J, Schurdak M, Morgan-Lappe S, Fesik S, Ram PT, Mills GB. 2011. Kinome siRNA-phosphoproteomic screen identifies networks regulating AKT signaling. Oncogene 30:4567–4577. 10.1038/onc.2011.164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Olsen JV, Blagoev B, Gnad F, Macek B, Kumar C, Mortensen P, Mann M. 2006. Global, in vivo, and site-specific phosphorylation dynamics in signaling networks. Cell 127:635–648. 10.1016/j.cell.2006.09.026 [DOI] [PubMed] [Google Scholar]
  • 125.Drake JM, Graham NA, Lee JK, Stoyanova T, Faltermeier CM, Sud S, Titz B, Huang J, Pienta KJ, Graeber TG, Witte ON. 2013. Metastatic castration-resistant prostate cancer reveals intrapatient similarity and interpatient heterogeneity of therapeutic kinase targets. Proc. Natl. Acad. Sci. U. S. A. 110:E4762–E4769. 10.1073/pnas.1319948110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Fox EM, Miller TW, Balko JM, Kuba MG, Sanchez V, Smith RA, Liu S, Gonzalez-Angulo AM, Mills GB, Ye F, Shyr Y, Manning HC, Buck E, Arteaga CL. 2011. A kinome-wide screen identifies the insulin/IGF-I receptor pathway as a mechanism of escape from hormone dependence in breast cancer. Cancer Res. 71:6773–6784. 10.1158/0008-5472.CAN-11-1295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Whitworth H, Bhadel S, Ivey M, Conaway M, Spencer A, Hernan R, Holemon H, Gioeli D. 2012. Identification of kinases regulating prostate cancer cell growth using an RNAi phenotypic screen. PLoS One 7:e38950. 10.1371/journal.pone.0038950 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Wolf-Yadlin A, Kumar N, Zhang Y, Hautaniemi S, Zaman M, Kim HD, Grantcharova V, Lauffenburger DA, White FM. 2006. Effects of HER2 overexpression on cell signaling networks governing proliferation and migration. Mol. Syst. Biol. 2:54. 10.1038/msb4100094 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Bose R, Molina H, Patterson AS, Bitok JK, Periaswamy B, Bader JS, Pandey A, Cole PA. 2006. Phosphoproteomic analysis of Her2/neu signaling and inhibition. Proc. Natl. Acad. Sci. U. S. A. 103:9773–9778. 10.1073/pnas.0603948103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Leroy C, Fialin C, Sirvent A, Simon V, Urbach S, Poncet J, Robert B, Jouin P, Roche S. 2009. Quantitative phosphoproteomics reveals a cluster of tyrosine kinases that mediates SRC invasive activity in advanced colon carcinoma cells. Cancer Res. 69:2279–2286. 10.1158/0008-5472.CAN-08-2354 [DOI] [PubMed] [Google Scholar]
  • 131.Suwaki N, Vanhecke E, Atkins KM, Graf M, Swabey K, Huang P, Schraml P, Moch H, Cassidy AM, Brewer D, Al-Lazikani B, Workman P, De-Bono J, Kaye SB, Larkin J, Gore ME, Sawyers CL, Nelson P, Beer TM, Geng H, Gao L, Qian DZ, Alumkal JJ, Thomas G, Thomas GV. 2011. A HIF-regulated VHL-PTP1B-Src signaling axis identifies a therapeutic target in renal cell carcinoma. Sci. Transl. Med. 3:85ra47. 10.1126/scitranslmed.3002004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Old WM, Shabb JB, Houel S, Wang H, Couts KL, Yen CY, Litman ES, Croy CH, Meyer-Arendt K, Miranda JG, Brown RA, Witze ES, Schweppe RE, Resing KA, Ahn NG. 2009. Functional proteomics identifies targets of phosphorylation by B-Raf signaling in melanoma. Mol. Cell 34:115–131. 10.1016/j.molcel.2009.03.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Grubb RL, Deng J, Pinto PA, Mohler JL, Chinnaiyan A, Rubin M, Linehan WM, Liotta LA, Petricoin EF, Wulfkuhle JD. 2009. Pathway biomarker profiling of localized and metastatic human prostate cancer reveal metastatic and prognostic signatures. J. Proteome Res. 8:3044–3054. 10.1021/pr8009337 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Lee BY, Hochgrafe F, Lin HM, Castillo L, Wu J, Raftery MJ, Martin Shreeve S, Horvath LG, Daly RJ. 2014. Phosphoproteomic profiling identifies focal adhesion kinase as a mediator of docetaxel resistance in castrate-resistant prostate cancer. Mol. Cancer Ther. 13:190–201. 10.1158/1535-7163.MCT-13-0225-T [DOI] [PubMed] [Google Scholar]
  • 135.Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, Quist MJ, Jing X, Lonigro RJ, Brenner JC, Asangani IA, Ateeq B, Chun SY, Siddiqui J, Sam L, Anstett M, Mehra R, Prensner JR, Palanisamy N, Ryslik GA, Vandin F, Raphael BJ, Kunju LP, Rhodes DR, Pienta KJ, Chinnaiyan AM, Tomlins SA. 2012. The mutational landscape of lethal castration-resistant prostate cancer. Nature 487:239–243. 10.1038/nature11125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, Kaushik P, Cerami E, Reva B, Antipin Y, Mitsiades N, Landers T, Dolgalev I, Major JE, Wilson M, Socci ND, Lash AE, Heguy A, Eastham JA, Scher HI, Reuter VE, Scardino PT, Sander C, Sawyers CL, Gerald WL. 2010. Integrative genomic profiling of human prostate cancer. Cancer Cell 18:11–22. 10.1016/j.ccr.2010.05.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Wong S, McLaughlin J, Cheng D, Zhang C, Shokat KM, Witte ON. 2004. Sole BCR-ABL inhibition is insufficient to eliminate all myeloproliferative disorder cell populations. Proc. Natl. Acad. Sci. U. S. A. 101:17456–17461. 10.1073/pnas.0407061101 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Molecular and Cellular Biology are provided here courtesy of Taylor & Francis

RESOURCES