Abstract
As the upward spiral of novel cancer gene discoveries and novel molecular compounds continues to accelerate, a repetitive theme in molecular drug development remains the lack of activity of initially promising agents when given to patients in clinical trials. It is however invigorating that a few targeted agents directed against a select group of a few ‘cancer gene superfamilies’ have escaped this all to common fate, and have evolved into novel, clinically meaningful molecular therapy strategies. Targeting dysregulated signaling of the epidermal growth factor family of transmembrane receptors (Erbb family) has encompassed over the last decade an ever increasing role in personalized treatment approaches in an increasing number of human malignancies. Erbbs are receptor tyrosine kinases that are important regulators of several signaling pathways. Two of its family members (Erbb1/EGFR and Erbb2/HER2) have previously been shown to be somatically mutated of human cancers. To determine if this family is somatically mutated in melanoma, its sequences were recently analyzed and one of its members, Erbb4, was found to be somatically mutated in 19% of melanoma cases. Functional analysis of seven of its mutations was shown to increase its catalytic and transformation abilities as well as providing essential survival signals. Similar to other Erbb family members, mutant Erbb4 seems to confer “oncogene addiction” on melanoma cells, making it an attractive therapeutic target. Gaining further understanding into the oncogenic mechanism of Erbb4 may not only help in the development of targeted therapy in melanoma patients but might accelerate the acceptance of a novel taxonomy of cancer which is based on the genomic perturbations in cancer genes and cancer gene families and their response to targeted agents.
Keywords: Erbb, personalized medicine, somatic mutation, inhibitor, cancer
General Introduction
Despite an ever increasing number of novel cancer targets and an explosion of new molecular and biological agents currently emanating from preclinical studies, genotype-directed therapy targeting the epidermal growth factor family of transmembrane receptors (Erbb family) remains one of the few prime models of successful personalized medicine. The journey of targeting EGFR mutations acting as driving oncogenic signals in lung cancer has now reached population-based, large scale screening efforts treating patients with non-small cell lung cancer (NSCLC) with the ‘classical’ EGFR L858R and exon 19 deletion mutations with the small molecule tyrosine kinase (TKI) inhibitors erlotinib or gefitinib.1,2 Multiple studies have shown that the presence of activating EGFR mutations in NSCLC is associated with response to erlotinib and gefitinib treatment, improved progression-free, and median overall survival.3–8 If the favorable outcome of NSCLC harboring EGFR mutations treated with erlotinib and gefitinib is solely due to the impact of TKI treatment and if EGFR mutations are solely a predictive factor for TKI response, or if EGFR mutations are also a prognostic factor for NSCLC outcome and reflect a more favorable tumor biology remains, however, to be determined.9,10 In a recent large randomized study patients with EGFR mutations not treated with erlotinib or gefitinib had significantly longer survival rates than patients with wild-type EGFR tumors.11 The predictive, and possibly prognostic, impact of activating EGFR mutations on cancer outcome has now been further dissected by demonstrating a substantial heterogeneity of these mutations in respect to TKI responsiveness and clinical outcome: of the two most common activating EGFR kinase-domain mutations, in-frame exon 19 deletions (Class I; 44% prevalence of EGFR mutation harboring NSCLC), and EGFR L858R mutations (Class II; 41%) response rates to erlotinib and gefitinib are twice as high in tumors with exon 19 deletions (70–100%) compared to tumors harboring L858R mutations in exon 21 (30–67%).10,12,13 Similarly, patients with exon 19 deletion mutations treated with TKI had a median overall survival twice as long as patients with L858R mutations treated with gefitinib.12,13 The biochemical and structural correlates of this different clinical behavior of the two mutants have now also been elucidated: for example, EGFR L858R mutations have a 10-fold higher KM (μmol/L) and a two-fold lower Ki (nmol/L) for ATP than exon 19 deletion mutants,14 and structural data suggest that exon 19 deletions causing a greater shift of the αC helix narrowing the ATP-binding cleft of the kinase domain to a greater degree which increases the affinity of the mutated receptor for TKIs.15 Now, novel discoveries on the role of non-kinase domain mutations in the Erbb family and the role of Erb somatic mutations in other histologies is raising hopes for a true ‘dent’ on cancer mortality even higher.
Why are mutated Erbb receptor family members such a prime cancer target? All members of the Erbb receptor family, EGFR/Erbb1/HER1, Erbb2/HER2/neu, Erbb3/HER3, Erbb4/HER4, are known to play a pivotal role in cell-cell signaling and signal transduction regulating cell growth and development.16,17 Members of the Erbb family are structurally very similar membrane-spanning tyrosine kinase receptors composing of an extracellular domain subdivided into four subdomains, an α-helical transmembrane segment, and an intracellular protein kinase domain (Fig. 1A).18 Erbb receptor activation requires dimerization of two Erbb molecules which normally exist as inactive monomers.17,18 Ligand binding induces the release of intramolecular bonds (‘autoinhibition’) between the extracellular domains II and IV leading to a conformational twist which exposes the dimerization domain in subunit II of the receptor.18,19 Pairing with another receptor leads to activation of the kinase domain, phosphorylation of the other receptor, and consequently binding and activation of downstream proteins (Fig. 1A).18,19 There are, however, important differences between Erb receptors which are relevant in regard to their functional roles. Some of these are summarized in Table 1.19,20 Among these differences are the lack of an intrinsic kinase activity of Erbb3 and the ligand-independent activation of Erbb2/HER2 which normally exists in the ‘open’ form being constitutively available for pairing and signal transduction.17,21 In general, the nature of the activating ligand and the different homo- and heterodimer pairings are the major determinants which of the various downstream targets are activated.17,22 Regulation of Erb receptor mediated signaling also occurs at the post-receptor level via ubiquitination, mediation of endocytosis, or neddylation.17,22 Therefore, taking the multilayered regulatory complexity of the Erbb signaling network into account, it is not surprising that dysregulated Erbb signaling is causally linked with the development and progression of cancer, and cells afflicted by dysregulated Erbb signaling display at least four of the six hallmarks of cancer (metastasis, resistance to apoptosis, independence of growth signals, resistance to growth inhibitory signals).148 To date, well established mechanisms of Erb dys-regulation include the amplification of the Erbb2/HER2 gene locus, SNPs within the promoter region or intronic sequences of ERBB4,23–25 abnormal proteolytic cleavage of ligand and Erb isoforms,25 and most notably, activating somatic mutations in the EGFR receptor kinase domain.3 Now, large-scale, high-throughput genotyping efforts, including studies conducted by the ‘Tumor Sequencing Project’ (TSP) initiative or ‘The Cancer Genome Atlas’ (TCGA),26 have shifted the focus towards the role of gene perturbations in the non-kinase domain as well as mutations in other histologies.
Figure 1.
Somatic mutations of the extracellular, non-kinase domain region of Erb receptors cause activated Erb signal transduction via different mechanisms. (A) Ligand activation of Erb family receptors. Ligand-binding to subregions I and III of the ectodomain of the closed, ‘thethered’ from of the receptor prompts a conformational ‘switch’ of the molecule characterized by the formation of ligand-binding pocket formed by subregions I, II and III and a 180° twist and exposure of the dimerization region of subdomain II as a prerequisite for dimerization and signal transduction activation. (B) Mutations in highly conserved regions of domains II and IV disrupt intersurface, intramolecular bonds between domains II and IV which keep the receptor monomer in its ‘thethered’ and inactive state. The inability to form these interactions results in the loss of domain II and IV binding, and a shift in the equilibrium to the open form of the receptor which exposes the dimerization region of domain II leading to constitutive pairing and abnormal signal transduction. Mutations disrupting these interactions include e.g. EGFR A597T, Erbb3 G284R and Q298, and Erbb4 Y285C, R306S and D595V mutations in lung cancer, EGFR T263P, A289D/T/V and P596L mutations in glioblastoma multiforme, and Erbb4 D609N mutations in melanoma. (C) Activating somatic mutations might affect the ligand-binding domains increasing the affinity of the receptor for the ligand and resulting in abnormal receptor activation. Examples of somatic mutations potentially involved involved in increased ligand-binding are the EGFR domain I mutations R108K in glioblastoma and Erbb4 mutation Y111H in melanoma, and the domain III mutation R451F and S442F in the EGFR receptors of lung cancer and glioblastoma and the Erbb4 mutants R393W, P409L and E452K in melanoma.
Table 1.
Structural and functional differences between Erb receptor family members in respect to signal transduction
| Ligand | Homo-/ Heterodimer | Intrinsic kinase activity | Downstream signaling | Alternative splice sites | |
|---|---|---|---|---|---|
| EGFR | EGF, TGFα, EPR, HB-EGF, AR, BTC | Both | Yes | Adaptor proteins GRB2 and GAB1, STAT5, Shc | No |
| ERBB2 | No direct ligand identified; exists in active confirmation state | Both | Yes | Adaptor proteins GRB2 and GAB1 | No |
| ERBB3 | NRG1α, NRG1β, NRG2α, NRG2β, NGC | Heterodimer only | No | p85 subunit PI3K | No |
| ERBB4 | Heregulin, NRG1α, NRG1β, NRG2α, NRG2β, NRG3, NRG4, AR, HB-EGF, TR1, TR2 | Both | Yes | p85 subunit PI3K, GRB2, STAT5, Shc | JM-a/JM-b CYT-1/CYT-2 |
Abbreviations: AR, amphiregulin; BTC, netacellulin; EGF, epidermal growth factor; EPR, epiregulin; HB-EGF, heparin-binding epidermal growth factor; TGFα, transforming growth factor; NRG, neuregulin; NGC, neuroglycan; TR, tomoregulin.
Mechanisms of non-kinase domain mutations in the Erb receptor family and their role in the development and evolution of cancer
For Erb receptors to remain in the inactive, ‘tethered’ configuration requires the formation of intramolecular bonds between domains II and IV of the ectodomain, which buries the dimerization interface of domain II within a deep pocket (Fig. 1A).27–29 Residues 266 to 283 of domain II in the EGFR receptor form a small β-hairpin loop that interacts with the C-terminal portion of domain IV. This contact is mediated via a main-chain hydrogen bond between tyrosine residue Y270 in domain II and an aspartic acid residue at position 586 of domain IV, and three side-chain interactions between residues Y270 and K610, F275 and G587, and Q276 and H589 of domains II and IV.27,29 The integrity of these interactions mediated by these amino acid residues is essential for maintaining the ‘folded’, inactive form of Erb receptors as shown by the ‘tether-mutant’ EGFR forms having an up to 12-fold increased affinity for the ligand binding to domains I and III.18,27,30 These directly contacting residues in domains II and IV are highly conserved among all four receptors and various species with the exception of G587 and H589 in the Erbb2 receptor which are substituted with proline and phenylalanine residues in this receptor. As the substitution of the glycin at position 587 with a proline residue and the histidine at 589 with a phenylalanine residue prevents the formation of these critical hydrogen bonds, the receptor loses the close intramolecular contact between domains II and IV contact and its the ability to maintain in the “closed” and inactive confirmation (Fig. 1B).18 Thus, this loss of intramolecular bonds also explains the unique feature of the Erbb2 monomer which is constitutively present in the “open,” active form, available for dimerization and signal transduction in a ligand-independent fashion. The biological importance of these residues for controlled Erb signaling has now been further re-affirmed by the finding of somatic mutations occurring at or in close proximity of these critical positions across the Erb receptor family and across different cancers. Examples of mutations with similar positioning disrupting interactions between the interdomain surfaces of domains II and IV include EGFR A597T, Erbb3 G284R and Q298, and Erbb4 Y285C, R306S and D595V mutations in lung cancer,26 EGFR T263P, A289D/T/V and P596L mutations in glioblastoma multiforme,31 and Erbb4 M313I, E317K, E542K, R544W, E563K and D609N mutations in melanoma (Fig. 1B and Table 2).32 Recent crystallographic data from the Erbb4 receptor seem to support the activating mechanism of some of the above mutations in the Erb receptor molecule: for example, D609N which lies on position one of the tight β turn of domain IV opposite the juxtramembrane part, and which is conserved as Asp in EGFR, Erbb2 and Erbb3, forms the base of the ‘tether’ pocket. Disruption of this critical local structure might destabilize the ‘thether’. Both, R544W and E563K form salt bridges with other residues of domain IV (Glu 538 and R 519), and disruption of these bridges will likely affect the stability and local structure of domain IV favoring a shift towards the open, and active, configuration.33
Table 2.
Somatic mutations including missense, nonsense, deletions, insertions and duplications previously identified in the Erb receptors of various epithelial cancers
| EGFR | Erbb2 | Erbb3 | Erbb4 | ||
|---|---|---|---|---|---|
| Lung | P136T | S310F | G69R | N181S | 26, 126–135 |
| R451F | D769H | G284R | T244R | ||
| A597T | 774insAYVM | Q298 | Y285C | ||
| V651M | L755P | R306S | |||
| E709delT710 | 775insYVMA | V348L | |||
| G719S | G776V | D595V | |||
| G719C | G776L | H618P | |||
| G719A | G776V, Cins | E810K | |||
| E746delA750 | 776insYVMA | T926M | |||
| E746delT751insA | 779insVGS | D931Y | |||
| E746delS752insV | 781insGSP | K935I | |||
| L747delT751 | W906 | ||||
| L747delS752 | |||||
| L747delP753insS | |||||
| L747delA750insP | |||||
| L747delT751insP | |||||
| P753F | |||||
| 774insNPH | |||||
| S768I | |||||
| 770insASV | |||||
| 771insGL | |||||
| 771insSVD | |||||
| 773insTHP | |||||
| T790M | |||||
| L833V | |||||
| P848L | |||||
| L858R | |||||
| L861R | |||||
| L861Q | |||||
| Breast | L755delT759 | E872K | 57, 136, 137 | ||
| L755S | |||||
| S760A | |||||
| R896C | |||||
| Gastric | G776S | A773S | 57, 126, 136, 138 | ||
| K724N | Intron 19 2302-8del-7insT | ||||
| T733I | Intron 19 2302-8delT | ||||
| L755S | |||||
| L755S | |||||
| D769H | |||||
| Q799P | |||||
| V777L | |||||
| V777L | |||||
| T862A | |||||
| L869Q | |||||
| GBM | D46N | E914K | 31, 126 | ||
| G63R | |||||
| R108K | |||||
| T263P | |||||
| A289D | |||||
| A289T | |||||
| A289V | |||||
| R324L | |||||
| E330K | |||||
| S442F | |||||
| P596L | |||||
| G598V | |||||
| L861Q | |||||
| Ovarian | E746delA750 | N857S | 126, 139, 140 | ||
| E746delA750 | A771insM774dupAYVM | ||||
| Y772insA775dupYVMA | |||||
| HCC | H878Y | 141 | |||
| CRC | E749K | V777L | V721I | 57, 66, 136, 142 | |
| E762G | V777M | P854G | |||
| A767T | V842I | D861Y | |||
| I1030M | |||||
| Head and Neck | K745R | V773A | 143–146 | ||
| E746delA750 | |||||
| E746delT751insA | |||||
| L747delE749 | |||||
| L747delE749, A750P | |||||
| S784Y | |||||
| T790M | |||||
| F795S | |||||
| P848L | |||||
| L862P | |||||
| Esophageal | E746delA750 | 147 | |||
| T790M | |||||
| L858R | |||||
| Pancreas | DelE746-A750 | 138, 147 | |||
| DelE746-A750 | |||||
| C818Y | |||||
| Melanoma | L39F | 32 | |||
| Y111H | |||||
| M313I | |||||
| E317K | |||||
| S341L | |||||
| R393W | |||||
| P409L | |||||
| E452K | |||||
| R491K | |||||
| E542K | |||||
| R544W | |||||
| E563K | |||||
| D609N | |||||
| P700S | |||||
| E836K | |||||
| E872K | |||||
| G936R | |||||
| P1033S | |||||
| S1246N | |||||
| R1174Q |
‘Hotspots’ (mutations occurring in more than 10% of cases) are shaded in gray (Del, deletion; ins, insertion; dupl, duplication).
Mutations affecting the extracellular subdomains I and III, which are involved in ligand-binding, can result in increased affinity of the receptor to the ligand and abnormal Erb signaling.20,34 Ligand binding occurs via interactions with domain III, and to a lesser degree via domain I.19,27,28,30 Essential residues in the EGFR receptor are the highly conserved residues R108 and Y113 in domain I, and the conserved residues D379, Q408, H418, Q432 and H433 of domain III.27,28 Mutations observed in the Erb family assumed to increase ligand binding leading to abnormal receptor activation are the EGFR domain I R108K mutations in glioblastoma,31 EGFR P136T in lung cancer,26 Erbb4 Y111H in melanoma,32 and for domain III EGFR R451F in lung cancer,26 EGFR S442F in glioblastoma,20,31 and Erbb4 H374Q, R393W, P409L and E452K in malignant melanoma (Fig. 1C and Table 2).32,35 While at this point crystallographic structure data from the Erbb4 molecule do not suggest a direct involvement of these residues in ligand binding, all domain III mutations in this region including R393W, P409L and E452K involve residues which are exposed on the surface impacting on receptor solubility and domain arrangement.33 If altered domain arrangement is the cause for such enhanced ligand binding remains at this point speculative.
In general, the observed clustering of activating somatic gene mutations in the Erb family around functional subdomains and structurally critical ‘hotspots’ of multiple Erb receptors and across different tumor histologies are proof of the biologic importance of these mutations in the development and progression of cancer. The recent observation in NSCLC that mutations in the Erb receptors are mutually exclusive with mutations in other Erb pathway genes (e.g., PI3KCA, BRAF, KRAS) further affirms their ‘tumor- driving’ potential.
Downstream targets
The most commonly affected signaling pathways by aberrant Erb signal transduction are the PI3K-Akt and MAP kinase pathways. Following receptor dimerization, the amino terminal part of one of the tyrosine kinase domains interacts with the C-terminal lobe of the other in an asymmetrical fashion which leads to phosphorylation of the respective C-terminal tyrosine residues of the other, interacting receptor molecule. This event recruits a variety of adaptor and cell signaling proteins and initiates downstream signaling.
Erb dimer composition and ligand-binding determines which pathway will be activated, a process which is still only incompletely understood. EGFR and Erbb2 bind indirectly via the Erb growth factor bound adaptor proteins GRB2-Sos (sons of sevenless)-RAS complex or through an additional mediator GAB1 (GRB2-associated binding protein 1) to PI3K, whereas Erbb3 and 4 have direct binding sites for the p85 subunit of class IA PI3K. EGFR and Erbb4 have also been shown to bind to STAT5 and Shc proteins.36 Erbb3 contains six binding sites for the p85 subunit of PI3K, and it is suggested that Erbb3 and 4-mediated signaling occurs preferentially via the PI3K-Akt pathway, an observation supported by activation of the PI3K-Akt pathway in cancers with unregulated Erbb3 signaling, and the unfavorable clinical outcome of cancers with abnormal Erbb2/Erbb3 heterodimer formation and deregulated PI3K-Akt signaling.21,37 It has also been shown that inhibition of EGFR or Erbb2 signaling with tyrosine kinase inhibitors leads to frequent compensatory upregulation of Erbb3 because of negative Akt-mediated feedback signaling, and that the upregulation of Erbb2 and Erbb3 dimers following cetuximab treatment, a monoclonal antibody against EGFR, induces PI3K-Akt activation.37,38
The elucidation of Erb-mediated downstream signal transduction is of substantial clinical relevance: to date, with the exception of imatinib mesylate in the treatment of BCR-ABL-positive chronic myeloid leukemia and gefitinib and erlotinib in lung cancer and to a lesser degree in glioblastoma, no single agent targeted therapy has been shown to impact on survival rates in a clinically meaningful way. The future success of molecular therapy will largely depend on the ability of developing either more effective compounds or combinational approaches which effectively target escape pathways and the extensive crosstalk of cancer-driving pathways. Figure 3 shows the potential therapeutic targets and their respective, clinically advanced compounds which are currently available. Based on the current knowledge of Erb-mediated signaling such an approach would either pursue:
Figure 3.
Somatic mutations cluster around structurally and functionally unique residues and subdomains in the Erb receptor family. Somatic mutations in all members of the Erb family (EGFR, Erbb2/HER2, Erbb3, Erbb4) discovered in epithelial cancers are shown. The locations of the mutations are shown in reference to their amino acid position and the structural domains of the involved receptors. Triangles indicating missense or nonsense mutations, diamonds are indicating deletions, insertions or duplications. Colors of the different cancers are depicted on the right. Mutations involved in the disruption of the formation and function of the highly conserved Y270 residue of domain II and G587 and H589 of domain IV mediating the formation of intramolecular bonds and stabilization of the ‘tethered’ form are highlighted in grey.
Double inhibition of the upstream, mutated/dysregulated Erb receptor targeting different domains of the receptor (e.g., the combination of the monoclonal antibody trastuzumab and the tyrosine kinase inhibitor lapatinib or combination of trastuzumab and pertuzumab two anti-Erbb2 directed monoclonal antibodies targeting different functions of the receptor).39–42
Vertical pathway inhibition by targeting the upstream receptor and downstream targets of its effector pathway (e.g., combining trastuzumab or lapatinib with molecular compounds targeting the PI3K-Akt pathway as recently shown in preclinical models using the dual PI3K-mTOR inhibitor NVP-BEZ235).43,44
Horizontal pathway inhibition by targeting parallel, via intracellular crosstalk activated pathways. In the case of Erb-mediated signal transduction targeting the MAP kinase pathway would likely yield the highest reduction of proliferative Erb-mediated cell signaling (e.g., while up to date no direct inhibitors of RAS are available, targeting BRAF in combination with other agents has gained greater attention; e.g., phase I/II studies currently exploring the combination of lapatinib and sorafinib or erlotinib and sorafinib) (Phase I NCT00984425, Phase II NCT00696696).
Targeting two concomitant oncogenic mutations that are not interconnected. Examples are mutations/dys-regulation of the Erb receptor family and other major oncogenic events as e.g., controlling cell cycle progression with CDK inhibitors like flavoperidol, PD-002991, or roscivitine after loss of CDKN2A.45
Need for additional targets in malignant melanoma
Melanoma is the sixth leading cancer in both men and women. Metastatic melanoma has a poor prognosis with five-year survival of less than 5%. Melanoma ranks second in terms of loss of years of potential life.46 FDA-approved treatments for metastatic melanoma include aldesleukin (IL-2) and dacarbazine chemotherapy.47 Aldesleukin as the most efficacious therapy has an objective clinical response rate of about 16% and a complete response rate of 6%.48 It is the only treatment known to cure some patients with metastatic melanoma. Dacabarzine or temazolamide-based chemotherapy has a clinical response rate of up to 20% but almost no complete responders or long-term survivors.48 With the incidence of melanoma increasing in the United States, and the fact that the last time a novel treatment for melanoma was approved by the FDA dates back to 1996, novel and more effective treatment strategies for metastatic melanoma are clearly needed. To date, none of the few targeted single-agents evaluated without additional patients’ selection in melanoma in phase II trials has shown any clinically meaningful activity.49 This rather bleak outlook has only very recently changed with novel molecular agents specifically targeting tumors harboring the BRAFV600 mutation or the initial success of imatinib in melanoma harboring c-KIT mutations.50,51 While the uneven distribution of c-KIT harboring melanoma might hamper a more widespread use of c-KIT targeted therapy,52 the completion of the ongoing phase II trials testing compound PLX-4032, which inhibits mutant BRAFV600, the most common oncogene in melanoma, with a 1,000-fold greater specificity than wild type BRAF,53 is currently eagerly awaited. It is evident that for molecular therapy to succeed a much larger pool of both potential targets and effective compounds is necessary.
Mutational Analysis of the Tyrosine Kinome in Melanoma
Protein Tyrosine Kinases (PTKs) are ideal drug targets in that they have an important role in cell proliferation and apoptosis, they are amenable to pharmacological manipulation, and have been implicated in the pathogenesis of several cancer types.54,55 This gene family is therefore a particularly good source to derive novel potential targets in melanoma. One way to conclusively associate a gene in cancer pathogenesis is to identify somatic mutations in the particular gene and to test whether the discovered mutations alter the function of the derived protein. Identification of somatic mutations in the PTK gene family in melanoma is therefore highly warranted.
In order to do this, several reagents and tools are necessary. For example, a high quality melanoma tumor bank, established through the generation of early passage tumor cell lines, allows the detection of somatic mutations that are otherwise masked by surrounding normal tissue. Additionally, automated methods for high throughput amplification of regions of interest coupled with their sequencing permit a robust analysis of large areas in the genome. Finally, the comparisons of these tumor derived sequences to their normal counterparts results in the identification of somatic mutations.
These methods have recently been harnessed to the high throughput sequencing of the PTK family in melanoma.32 In order to evaluate whether any PTKs were genetically altered in melanoma, the kinase domain coding exons of all 86 PTKs were assessed in a panel of 29 melanoma samples. 593 exons were PCR amplified from melanoma derived genomic DNA and sequenced. All sequences were compared to the reference human genome and whenever a sequence alteration was identified, the same sequence was examined in genomic DNA derived from the normal tissue from the same patient. From this it was determined that 19 genes contained a total of 30 somatic (i.e. tumor-specific) mutations affecting the kinase domains of the protein.
The next step in this evaluation was to expand the analysis beyond the kinase domains of the 19 genes that were found to harbor somatic mutations in a larger melanoma tumor panel. To do this, all coding exons of each of the 19 genes were sequenced in a total of 80 samples leading to the identification of 99 non-synonymous somatic mutations. One commonly used method to determine whether somatic alterations in particular proteins have a role to play in tumorigenesis is to calculate the ratio of non-synonymous (NS) to synonymous (S) alterations, as it is expected that synonymous changes would have a neutral effect on structure and function of the respective protein. The NS/S ratio of the whole PTK screen was 5.8:1 which is significantly higher than the N/S ratio of 2.5:1 predicted for nonselected passenger mutations (p < 1 × 10−5) leading to the conclusion that a large panel of these alterations are driver mutations.56 Another way to determine whether mutations have a role in tumorgenesis is to functionally evaluate how these alterations affect the activity of the wild type version of the protein.
Out of the 19 genes that were found to harbor somatic mutations in the above screen, the most highly mutated was Erbb4, containing 24 non-synonymous somatic mutations affecting 19% of melanoma patients making Erbb4 the most highly mutated protein tyrosine kinase in melanoma. All but three of the alterations were heterozygous and no truncating or nonsense mutations were identified. One of the mutations identified in the kinase domain (E872K), was previously discovered in breast cancer,57 and two melanoma cases harbored mutations at the same position (hotspot E452K). Similarly to the kinome screen, Erbb4 had a NS:S ratio of 24:3 which is also significantly higher than the NS/S ratio predicted for non selected passenger mutations, thus suggesting that at least a portion of the discovered mutations are driver mutations and that Erbb4 is playing a role in tumorigenesis. A recurring theme in the identified mutations is a substitution of an acidic residue with a basic residue which might be due to the high frequency of C:G>T:A transitions, known as a UV signature found in melanomas.58 Interestingly, most of the tumors with Erbb4 mutations also contained mutations in either NRAS or BRAF, suggesting the Erbb4 operates via different pathways to the ones of NRAS and BRAF. A similar scenario was described for the well known oncogene PIK3CA in colorectal cancer where the majority of tumors with PIK3CA mutations also contained mutations in KRAS or BRAF.59 Importantly, at least 50% of the discovered mutations in Erbb4 were found in melanoma samples derived from patients who have not been treated with chemo or radiotherapy and two of the Erbb4 mutations (E452K and E872K) found in chemotherapy treated patients wre proven functionally to be activating.32 These alterations are summarized in Table 2.
Functional Evaluation of Erbb4 Mutations
To determine the biological effects of the identified Erbb4 mutations seven of the identified mutations were cloned and investigated in biochemical and biological assays. Since a total of 24 somatic mutations were discovered in the mutation screen several criteria were used to determine which of these to test. These included the proximity of the mutations to previously identified mutations in EGFR and Her2 on the Erbb4 protein crystal structure as well as the clustering of two of the mutations into two mini-hotspots (E452K and E872K)32 (Table 2).
To assess the biochemical effects of these alterations on Erbb4 kinase activity, wild type Erbb4 or the seven mutations were transiently transfected into HEK-293T cells and their autophosphorylation as well as kinase activity were evaluated. Expression of mutant Erbb4 resulted in significantly more kinase activity than wild-type protein.32 While the exact mechanism that lies behind the increased activity of mutations located in the extracellular domain remains in the majority of the mutations to be determined, crystallographic, structural and function data from mutations in similar positions in EGFR and Her2 suggest that the evaluated extracellular domain mutations may increase signaling activity due to a shift in the conformational equilibrium, increased ligand binding or by constitutive dimerization (Fig. 1B and C).
Functional assessment of the above Erbb4 mutations revealed them to show oncogenic activity: They transformed primary fibroblasts in culture, to a similar potency as oncogenic K-RasG12V. In addition, they induced increased foci formation and anchorage independent growth of melanoma cells.32 The seven evaluated mutations can therefore be regarded as ‘driver’ mutations and Erbb4 as an oncogene.
The dependence of melanoma cells on mutant Erbb4 signaling was initially established by performing shRNA knockdown of endogenous Erbb4 in melanoma cells that harbor either its wild type or mutant version. In each case in which Erbb4 was knocked down in cells containing mutant Erbb4, melanoma cells halted their proliferation, whereas cells maintained their growth rate when similar knockdowns were performed in cells containing wild type Erbb4. This ‘addiction’ of melanoma cell lines harboring mutated variants of the Erbb4 receptor to Erbb4 signal transduction established Erbb4 as an essential signal and dominant oncogene that allows for the growth of melanomas harboring these mutations. Indeed, to institute the potential relevance of mutant Erbb4 signaling to the clinic, the broadly acting Erbb inhibitor, lapatinib, was tested for its inhibition of cell proliferation of melanoma cells with or without the Erbb4 mutations. Strikingly, despite the fact that lapatinib is not Erbb4-specific, a clear growth inhibition of cells with Erbb4 mutations versus cells expressing wild type Erbb4 was observed,32 thus suggesting that cells with Erbb4 mutations are subject to ‘oncogene addiction’. The fact that response to lapatinib occurs despite the existence of other somatic mutations, strengthens the point that melanoma cells harboring Erbb4 mutations are dependent on its signaling. It is reasonable to expect that inhibition of co-existing oncogenes, such as Braf, will further increase the response and increased growth inhibition will be observed.
As several different Erbb4 isoforms exist which differ in their functions and their ability to promote growth,25 one of the interesting follow-up questions to this study is to test whether any of the splice site mutations that were discovered affect which Erbb4 isoform is expressed. Using a subset of the melanoma samples that were used in the mutational analysis study, it was shown that the melanoma samples exclusively express Erbb4 JM-a coupled to both types of its cytoplasmic isoforms, CYT-1 and CYT-2. No signal was observed for the Erbb4 JM-b isoform in any of the samples.60 This corroborates well with the studies described above for two reasons: (a) Prickett et al. reported that the Erbb4 mutations activate the AKT pathway specifically and the CYT-1 Erbb4 isoform is the isoform that is known to directly activate this pathway and (b) when the kinase activity of the Erbb4 mutants was evaluated, the JM-a CYT-1 Erbb4 isoform was used as the plasmid backbone to which the mutations were cloned. As this represents the Erbb4 isoform that is expressed by the melanoma cell lines, this makes the kinase results physiologically relevant.61
Notably, numerous different somatic mutations were identified that are widely distributed over the coding sequence and occur in all domains of Erbb4. Cleary, the seven mutations that were evaluated show gain of function phenotypes. However, since the identified mutations do not cluster into hot spot regions as seen in oncogenes such as PIK3CA, Kras, Braf and GNAQ,59,62–64 the oncogenic activity is probably due to the removal of negative regulatory domains. This is reminiscent of the mutations identified in FLT3.1,35 Clearly many questions remain unanswered regarding the effects of these mutations. For example, what effect do these mutations have on the binding of ligand, on Erbb4 structure, on its interaction with regulatory proteins. Do all the observed Erbb4 mutations results in increased kinase activity? Further biochemical and biological analysis will be required to fully investigate the effects of the identified alterations on Erbb4 function.
As mentioned above, Erbb4 has previously been described to contain mutations in various cancer types, including, gastric carcinomas (1.7%), colorectal carcinomas (0.68–2.9%), NSCLC (2.3–4.7%) and breast carcinomas (1.1%),26,57,65,66 and their biological impact has not always been consistent across these cancers. For example, analysis of some alterations suggested that Erbb4 may function as a tumor suppressor in breast cancer and prostate cancer.67,68 Another report describing the functional effects of Erbb4 mutations revealed two mutations to cause the loss of kinase activity when Erbb4 homodimerizes, but maintain Erbb4’s ability to heterodimerize and promote signaling via the ERK and AKT pathways. The findings of some Erbb4 mutations causing a loss of function is, however, not necessarily contradictory to the noted oncogenic effects of Erbb4 mutations. One important explanation for this is that as indicated above, Erbb4 encodes four different isoforms that promote various cellular responses,25,69 and different isoforms were used in the various cited studies. For example, in recent reports in which endogenous Erbb4 levels was downregulated in tumor cells, a growth promoting role for Erbb4 was observed strengthening the notion that Erbb4 acts as an oncogene.25,70
Somatic Mutations of the Erb Receptor Family in Other Cancers
Activating gene mutations in the Erbb tyrosine kinase receptor family have first been described in the EGFR/Erbb1 receptor which harbor these mutations in 10–30% of cases.4,5 The most common, biologically and clinically relevant, EGFR L858R and the exon 19 746 and 747 deletion variants, have now been found in no less than six different tumor types including lung, glioblastoma, ovarian, head and neck, esophageal and pancreas cancer (Fig. 2; Table 2),58 and their clinical value in NSCLC and GBM tumors is now firmly established as tumors harboring these mutations are commonly sensitive to the small molecule TKIs erlotinib and gefitinb.3,71–73 Similar to the activation mechanism of cyclin-dependent kinases, the A-loop of the intracellular kinase domain is in the active state extended and directed away from the catalytic cleft of the kinase domain in order to (1) enable and facilitate the interaction and transient binding of the phospharylation sites of the peptide substrate (commonly the C-terminal domain of the other, paired receptor) and (2) to stabilize and enhance ATP binding via an interaction of a hydrophilic glutamate residue from the C-loop of the kinase domain, which forms the backbone of the catalytic cleft, and a hydrophilic lysine residue.74,75 In the ‘closed’, inactive state, the A loop is ‘covering’ the cleft preventing docking and binding of peptide substrate, and the glutamate residue essential for ATP binding is not available as through an outward displacement the C-loop is rotated away separating the glutamate residue from the lysine. This conformational change is associated with a dramatic drop in the ATP affinity of the receptor.76,77 Mutations in the kinase domain, and possibly in the extracellular domain, disrupt this tightly regulated and balanced equilibrium. In the best studied example of EGFR L858R mutations, the substitution of the hydrophobic leucine with the hydrophilic arginine favors interactions with the surrounding ‘hydrophilic’ solvent inducing a conformational change which moves the A loop away from protecting the catalytic cleft from binding of a peptide substrate, destabilizes the inactive form, and facilitates formation of the ‘open’, active status.77,78 Since the degree, mechanism and pattern of these induced conformational alterations is different for each individual mutant, it is not surprising that different responses to allosteric inhibitors, like an increased sensitivity of cells with the L858 mutations to gefitinib over erlotinib, have been observed.79 Translation of this knowledge into a truly comprehensive molecular approach will ultimately require considering each distinct mutant as a unique cancer target and the design of a distinct and unique molecular agent for each distinct mutation.
Figure 2.
Post-receptor signal transduction pathways of Erb-receptor mediated signal transductions occurs mainly via the PI3K-Akt and MAPK kinase pathways (Erbb-mediated STAT signaling not shown). Potential targets and their inhibitors for combinational molecular and biological therapy strategies are shown.
Extensive searches have been performed to identify similar hotspots of activating mutations in other members of the Erbb family. Several independent studies have identified mutations in the kinase domains of the Erbb2/HER2 and Erbb4 receptors.31,71,80,81 The majority of kinase domain mutations in Erbb2 and Erbb4 are similar to the mutations seen in EGFR. They cluster around the two hotspots (‘classical’ EGFR mutations) and include exon 19 missense mutations, deletions, and insertions at positions 746 and 7474, and missense mutations at or in close proximity of L858R. Mutations in the kinase domain in Erbb2 and Erbb4 have been identified in lung, gastric, ovarian, colorectal, and head and neck cancers and are overall the most common type of activating somatic mutations affecting the Erbb family (Table 2). It is interesting that Erbb2 and Erbb4 receptor mutations affecting the kinase domain have a higher rate of missense mutations in exon 19 compared to the more common deletion variants at this location in the EGFR receptor (Table 2). This is particularly the case for gastric and colorectal cancer where no deletion or insertion mutations in the kinase domain of Erbb2 and 4 receptors have been identified. When comparing the predominantly affected Erb receptor within cancer types, it is striking that with the exception of colorectal cancer predominantly only one of the Erb receptors is affected in a near exclusive fashion. This is the EGFR receptor in lung cancer, glioblastoma, and head and neck cancers, the Erbb2 receptor in breast, gastric, and hepatocellular cancer, and Erbb4 in melanoma (Table 2). This receptor specific pattern of somatic mutation involvement points to a receptor preference for Erb signaling in individual tumors. If this receptor preference is due to an endogenous dominance of the affected receptor, or if oncogenic mutations transformed the affected Erb receptor into the driving Erb signaling molecule by providing a selection and growth advantage remains unresolved at this point. For a large number of mutations which do not involve one of the functional 4 subdomains of the ectodomain, the juxta- or transmembrane or kinase domains, or regulatory elements within the C-terminus harboring sites for effector proteins, adaptor molecules, or binding sites for proteins mediating receptor turnover, the exact mechanisms and impact onto Erb signaling remains to be determined. Also, it is important to note that there is great clinical, environmental, and ethnic variation of both the prevalence of activating mutations in Erb receptors and the response to TKI treatment of these EGFR mutations.5,82 For example, mutations are more commonly found in women, patients of Asian descend, never smokers and patients with adenocarcinomas of the lung. African Americans have a substantially lower frequency of EGFR mutations, and response to TKI treatment, and their mutation profile is significantly different from that of Asian or Caucasian cancer populations.82
Current and future strategies targeting aberrant Erb signaling
Current efforts on Erb-directed therapy exploiting the novel understanding of normal and aberrant Erb signaling focuses on (1) the development of improved, kinase-directed therapy targeting molecular subtypes of dysregulated Erb signaling, and (2) the development of second generation inhibitors to overcome resistance to Erb-directed targeted therapy.
The heterogeneity of activating Erb mutations, across the different domains of the individual Erb molecules, across the Erb receptor family itself, and across different tumor types will likely require a variety of different molecular interventions designed against the most prevalent Erb mutations to achieve maximum suppression of Erb signaling. To date, the monoclonal antibodies trastuzumab and cetuximab, and the TKIs gefitinib, erlotinib and lapatinib as regulatory approved agents form the backbone of Erb-directed targeted therapy (reviewed in ref. 83). The investigational monoclonal antibody pertuzumab, which exclusively binds to the dimerization region of subdomain II, aims to specifically target the dimerization requirement of Erb-mediated signal transduction,84,85 and might be of particular value in tumor harboring mutations in subdomains II and IV which lead to ‘unfolding’ of the closed receptor monomer, exposure of the dimerization domain, and enhanced signal transduction. Preclinical data with pertuzumab confirm the effective inhibition of Erbb2 dimerization,86,87 in particular Erbb-2-Erbb3 dimerization, and inhibition of Erb-mediated signal transduction that mediates tumorigenesis via activation of the PI3K-Akt pathway.16,86 While pertuzumab has shown substantial activity in human xenograft models derived from ovarian,88 prostate,89 lung90 and colorectal cancer91 cell lines most of them overexpressing Erbb2, clinical activity of this biological agent in an initial trial of non-selected patients with meta-static breast cancer has been disappointing.92 More recent trials, however, have confirmed activity and clinical benefit of pertuzumab in combination with trastuzumab in HER2 amplified metastatic breast cancer.39,41,42 These findings point not only to the need of selecting patients with activated Erbb2 signaling, but make pertuzumab potentially one of the first anti-Erb agents targeting a specific molecular feature of dysregulated Erb signaling. Improved ‘pharmacogenomic matching’ of the underlying genomic aberration and resulting structural alteration with the chosen anti-Erb targeted agent will ensure maximum reduction of oncogenic signaling and success of targeted therapy. This concept of exploiting molecular subtypes of an altered genotype of a receptor super family is in line with ongoing studies which combine trustuzumab and pertuzumab in breast cancer targeting different receptor domains of the Erbb2 molecule (Phase III; NCT00567190) or the addition of the TKI lapatinib to rescue trastuzumab-resistant breast cancer in patients with HER2-overexpressing tumors.40 The ideal agent against mutated Erb receptors, in particular for mutations like EGFR L858R and del747-P754insS, are TKIs with increased affinity to the mutated molecule compared to the wild type receptor. While such compounds have recently been described for the BRAFV600 mutation, a common oncogene in thyroid cancer and melanoma, where the novel compound PLX 4302 has a more than 1,000-fold increased sensitivity to mutant BRAFV600 than wild type BRAF kinase,53 similar compounds are currently not available for mutated Erb receptors. Current efforts on improved direct targeting of Erb kinase activity, which have completed phase I trials, include the recent development of TKIs like neratinib (HKI-272) and BIBW-2992,93 which bind irreversibly to Erb receptors or ARRY-334543 and TAK-285 which concomitantly inhibit multiple members of the Erb family.94,95
The other approach of improved targeting of mutant Erb signaling focuses on the evolving understanding of the resistance mechanisms of Erb-directed targeted therapy. Further development along this avenue is insofar more essential in respect to the durability of Erb-directed therapy as the clinical experience with single-agent targeted therapy uniformally tells that eventually all patients initially responding to TKIs will relapse and progress in their disease.96 Multiple trials supporting this notion have shown that initial differences in progression-free survival between the investigational arm of single-agent molecular therapy and the control cohort vanish with longer follow-up and no difference in overall survival was detected. The latest example hereto is the large, prospective randomized trial on adjuvant imatinib in patients with gastrointestinal stromal tumors (GIST).97
Resistance mechanisms against Erb-directed therapy include both the presence of an a priori resistance mechanism, most notably the presence of additional oncogenic mutations, or the acquaintance of de novo resistance during molecular therapy. The prime example of concomitant resistance-mediating genomic alterations in Erb-targeted therapy which has found entrance into clinical practice is the presence of KRAS mutations in NSCLS and colorectal cancer.11,98,99 For example, clinical benefit to cetuximab, an EGFR-directed monoclonal antibody, in colorectal cancer is not observed in tumors which harbor KRAS mutations.99 It is therefore recommended to test KRAS mutation status in patients considered for this biological agent prior to start of treatment, and cetuximab administration is discouraged in patients harboring KRAS mutations.100 Other examples of concurrent genomic alterations mediating a negative impact on the response to Erb-directed TKI therapy include absence of wild type BRAF in KRAS wild type colorectal cancer,101 concurrent PI3KCA mutations,102 amplification of wild type EGFR,103 PTEN deletions activating the PI3K-Akt pathway,100,104 and concomitant HER2 mutations.105
Equally clinically relevant, but frequently more common, are acquired resistance mechanisms during anti-Erb therapy.96 EGFR-mutant tumors in NSCLC previously treated with either gefitinib or erlotinib and initially responsive to these TKIs develop a secondary EGFR T790M mutation in more than half of the cases.106–108 EGFR T790M mutations increase the affinity for ATP to the receptor overcoming the competitive binding of the reversible allosteric inhibitors erlotinib and gefitinib.106–108 The development of similar mutations at analogous positions in the kinase domains have been observed in CML and GIST treated with imatinib (T315I in ABL, T674I in PDGFRα, and T670I in c-KIT).109–111 It is hoped that second-generation TKIs like neratinib (HKI-272) and BIBW-2992, which bind irreversibly to Erb receptors, including receptors which harbor T790M mutations, are able to overcome TKI resistance.93,108 Another strategy to overcome TKI resistance in patients treated previously with TKIs is the administration of heat shock protein inhibitors.112 By disrupting the heat shock protein function, which stabilizes Erb receptors, Erb molecules are more prone to ubiquitylation and degradation thereby reducing Erb signaling. The compound with the most advanced preclinical and clinical development is the heat shock protein 90 (Hsp90) inhibitor tanespimycin (17-AAG) which has now completed phase I trials.113 Another important, recently elucidated mechanism of TKI resistance is the amplification of the proto-oncogene MET which has been observed in 20% of NSCLCs treated with erlotinib or gefitinib.114–116 MET-mediated activation of downstream signaling, in particular activation of the PI3K-Akt, STAT3 and five pathways through activation of Erbb3 substitutes for the loss of EGFR-mediated signaling abrogated by TKI inhibition,117 and MET kinase inhibitors, like SU-11274, might be of particular clinical value in this scenario. Finally, it recently has been recognized that cancers with Erb mutations treated with TKI might usurp physiologic regulatory and feedback mechanism to develop resistance.37,118 Preclinical models of lung, colorectal and pancreatic cancer cell lines treated with erlotinib found compensatory upregulation of the insulin growth factor receptor 1 insulin receptor substrate (IGF-IR IRS-I) and a compensatory upregulation of Akt activity via IGF-IR.118 Addition of the IGF-IR inhibitor OSI-906 to erlotinib has shown promising activity in preclinical studies,118 a finding which is currently translated into the clinic.118
Personalized Therapy
Cancer treatment is currently undergoing a major paradigm shift in the U.S. Up to now, the majority of administered systemic cancer treatment, in particular cytotoxic therapy, is currently administered without any clinical benefit to patients disregarding the vast molecular heterogeneity of cancers. This shockingly high number will with the inevitable emerge of personalized medicine undoubtedly decrease, a goal now embraced by many lead organization like ASCO and the ACS.
There are two fundamental advantages of personalized cancer medicine: (1) to determine prior to treatment which patients will likely benefit from the planned therapeutic intervention, and preventing toxicities in patients who would derive no benefit from treatment, and (2) to further modulate additional therapies should the first-line therapy fail, the tumor become resistant or is limited by unacceptable toxicities.
The improved diagnostic and predictive value of genomic information enabling improved patient selection is expected to have one of the most profound impacts on the quality of cancer care in the forthcoming decade.119 To treat only patients which will likely benefit from treatment has prime examples, like the estrogen-receptor status for anti-estrogen therapy in breast cancer,120 SNP analysis of the UDP-glucoronosyltransferase 1 A1 gene for irinotecan toxicity,121,122 and Bcr-Abl fusion and c-KIT mutation analysis for imatinib therapy in CML and GIST to name a few. The advent of genomic technology for the analysis of human tumor samples has now added an unprecedended source of information to aid prognosis and clinical decisions.123 The dimensions and complexity which such data can capture provides an opportunity to predict clinically valid trends, like risk of recurrence or response to treatment, that can distinguish phenotypes in ways that traditional methods cannot.124,125 It is this variability in outcome not captured by current predictive and prognostic models based on classical clinicopathological data which currently deprives patients of personalized treatment allocations which optimally tailor selected treatment decisions to their individual tumor biology.
Therefore, a true commitment to personalized medicine will require us to abandon a so far histologically defined approach to cancer care, and embrace molecularly defined cancer subtypes and their different treatment algorithms for the selection of effective treatment strategies. It was initially thought that increased fragmentation and compartmentalization of cancer subtypes will hamper further development and progress of personalized cancer care. It was assumed that an ever increasing pool of cancer subgroups and subtypes with an ever decreasing size through further and further split-ups into novel molecular subgroups which arise from new molecular discoveries would make it impossible to conduct adequately powered clinical trials, obtain interest by pharmaceutical companies for very small markets, or even obtain enough clinical expertise in these rare phenotypes. Only recently has the pendulum shifted into the opposite direction now acknowledging the huge potential of a truly personalized approach. This includes not only the initial recognition of the huge savings of unnecessarily administered inefficient therapies including the saved costs of their associated toxicities but more so the potential of combining molecular subtypes, like Erb mutated cancers, across the boundaries of different histologies into a novel subclass of cancer negating some of the concerns raised above. To go a step further, such a classification should ideally not only be based on tumor genotype but also on the corresponding chemical signature associated with it. For example, the response to the TKI inhibitor lapatinib (currently approved for treatment of advanced breast cancer in combination with capacitabine or after previously failed treatment with either a taxane, anthracycline or trastuzumab in metastatic breast cancer) could define a class of dual-TKI responsive tumors comprising of Erbb2-amplified metastatic breast cancers, metastatic melanoma patients harboring somatic mutations of the Erbb4 gene which renders these tumors sensitive to lapatinib, and possibly—in the future—Erbb2-amplified gastric cancer or glioblastoma harboring the EGFRvIII deletion mutation (Fig. 4). As genotype-directed therapy is currently the fastest moving and dominating frontier in cancer medicine, the question will already not be if personalized medicine will make the conventional “one fits all” approach obsolete but when.
Figure 4.
A novel taxonomy of cancer based on genotype and the ‘chemotype’ response to the tyrosine kinase inhibitor lapatinib. Malignancies characterized by genetic dysregulation of the Erb receptor family (e.g., amplification of the Erbb2/HER2 gene in breast cancer, activating somatic gene mutations of the EGFR receptor in lung cancer and glioblastoma, or of Erbb4 in melanoma) and by response to the dual tyrosine kinase inhibitor lapatinib may form a novel genetically and clinically defined subgroup of cancer. Currently, Erbb2/HER2 positive metastatic breast cancer and malignant melanoma harboring Erbb4 mutations are malignancies that can be defined by genetically dysregulated Erb signaling and response to lapatinib. As genomic data and functional tests in other cancers emerge, glioblastoma multiforme harboring the EGFR deletion variant EGFRvIII (GBM EGFRvIII) and gastric cancer with Erbb2/HER2 amplification might be added to this subgroup in the near future (shaded background). Seize of rectangles reflect both prevalence of malignancy and proportion with dysregulated Erb signaling.
References
- 1.Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947–57. doi: 10.1056/NEJMoa0810699. [DOI] [PubMed] [Google Scholar]
- 2.Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, et al. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. 2009;361:958–67. doi: 10.1056/NEJMoa0904554. [DOI] [PubMed] [Google Scholar]
- 3.Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129–39. doi: 10.1056/NEJMoa040938. [DOI] [PubMed] [Google Scholar]
- 4.Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497–500. doi: 10.1126/science.1099314. [DOI] [PubMed] [Google Scholar]
- 5.Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci USA. 2004;101:13306–11. doi: 10.1073/pnas.0405220101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Takano T, Ohe Y, Sakamoto H, Tsuta K, Matsuno Y, Tateishi U, et al. Epidermal growth factor receptor gene mutations and increased copy numbers predict gefitinib sensitivity in patients with recurrent non-small-cell lung cancer. J Clin Oncol. 2005;23:6829–37. doi: 10.1200/JCO.2005.01.0793. [DOI] [PubMed] [Google Scholar]
- 7.Cortes-Funes H, Gomez C, Rosell R, Valero P, Garcia-Giron C, Velasco A, et al. Epidermal growth factor receptor activating mutations in Spanish gefitinib-treated non-small-cell lung cancer patients. Ann Oncol. 2005;16:1081–6. doi: 10.1093/annonc/mdi221. [DOI] [PubMed] [Google Scholar]
- 8.Cappuzzo F, Hirsch FR, Rossi E, Bartolini S, Ceresoli GL, Bemis L, et al. Epidermal growth factor receptor gene and protein and gefitinib sensitivity in non-small-cell lung cancer. J Natl Cancer Inst. 2005;97:643–55. doi: 10.1093/jnci/dji112. [DOI] [PubMed] [Google Scholar]
- 9.Han SW, Kim TY, Hwang PG, Jeong S, Kim J, Choi IS, et al. Predictive and prognostic impact of epidermal growth factor receptor mutation in non-small-cell lung cancer patients treated with gefitinib. J Clin Oncol. 2005;23:2493–501. doi: 10.1200/JCO.2005.01.388. [DOI] [PubMed] [Google Scholar]
- 10.Mitsudomi T, Kosaka T, Endoh H, Horio Y, Hida T, Mori S, et al. Mutations of the epidermal growth factor receptor gene predict prolonged survival after gefitinib treatment in patients with non-small-cell lung cancer with postoperative recurrence. J Clin Oncol. 2005;23:2513–20. doi: 10.1200/JCO.2005.00.992. [DOI] [PubMed] [Google Scholar]
- 11.Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst RS, et al. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol. 2005;23:5900–9. doi: 10.1200/JCO.2005.02.857. [DOI] [PubMed] [Google Scholar]
- 12.Riely GJ, Pao W, Pham D, Li AR, Rizvi N, Venkatraman ES, et al. Clinical course of patients with non-small cell lung cancer and epidermal growth factor receptor exon 19 and exon 21 mutations treated with gefitinib or erlotinib. Clin Cancer Res. 2006;12:839–44. doi: 10.1158/1078-0432.CCR-05-1846. [DOI] [PubMed] [Google Scholar]
- 13.Jackman DM, Yeap BY, Sequist LV, Lindeman N, Holmes AJ, Joshi VA, et al. Exon 19 deletion mutations of epidermal growth factor receptor are associated with prolonged survival in non-small cell lung cancer patients treated with gefitinib or erlotinib. Clin Cancer Res. 2006;12:3908–14. doi: 10.1158/1078-0432.CCR-06-0462. [DOI] [PubMed] [Google Scholar]
- 14.Carey KD, Garton AJ, Romero MS, Kahler J, Thomson S, Ross S, et al. Kinetic analysis of epidermal growth factor receptor somatic mutant proteins shows increased sensitivity to the epidermal growth factor receptor tyrosine kinase inhibitor, erlotinib. Cancer Res. 2006;66:8163–71. doi: 10.1158/0008-5472.CAN-06-0453. [DOI] [PubMed] [Google Scholar]
- 15.Kumar A, Petri ET, Halmos B, Boggon TJ. Structure and clinical relevance of the epidermal growth factor receptor in human cancer. J Clin Oncol. 2008;26:1742–51. doi: 10.1200/JCO.2007.12.1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–37. doi: 10.1038/35052073. [DOI] [PubMed] [Google Scholar]
- 17.Baselga J, Swain SM. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer. 2009;9:463–75. doi: 10.1038/nrc2656. [DOI] [PubMed] [Google Scholar]
- 18.Ferguson KM, Berger MB, Mendrola JM, Cho HS, Leahy DJ, Lemmon MA. EGF activates its receptor by removing interactions that autoinhibit ectodomain dimerization. Mol Cell. 2003;11:507–17. doi: 10.1016/s1097-2765(03)00047-9. [DOI] [PubMed] [Google Scholar]
- 19.Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, Denney DW, Jr, et al. Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab. Nature. 2003;421:756–60. doi: 10.1038/nature01392. [DOI] [PubMed] [Google Scholar]
- 20.Riese DJ, 2nd, Gallo RM, Settleman J. Mutational activation of ErbB family receptor tyrosine kinases: insights into mechanisms of signal transduction and tumorigenesis. Bioessays. 2007;29:558–65. doi: 10.1002/bies.20582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Pinkas-Kramarski R, Soussan L, Waterman H, Levkowitz G, Alroy I, Klapper L, et al. Diversification of Neu differentiation factor and epidermal growth factor signaling by combinatorial receptor interactions. EMBO J. 1996;15:2452–67. [PMC free article] [PubMed] [Google Scholar]
- 22.Carpenter G. ErbB-4: mechanism of action and biology. Exp Cell Res. 2003;284:66–77. doi: 10.1016/s0014-4827(02)00100-3. [DOI] [PubMed] [Google Scholar]
- 23.Rokavec M, Justenhoven C, Schroth W, Istrate MA, Haas S, Fischer HP, et al. A novel polymorphism in the promoter region of ERBB4 is associated with breast and colorectal cancer risk. Clin Cancer Res. 2007;13:7506–14. doi: 10.1158/1078-0432.CCR-07-0457. [DOI] [PubMed] [Google Scholar]
- 24.Murabito JM, Rosenberg CL, Finger D, Kreger BE, Levy D, Splansky GL, et al. A genome-wide association study of breast and prostate cancer in the NHLBI’s Framingham Heart Study. BMC Med Genet. 2007;8:6. doi: 10.1186/1471-2350-8-S1-S6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Maatta JA, Sundvall M, Junttila TT, Peri L, Laine VJ, Isola J, et al. Proteolytic cleavage and phosphorylation of a tumor-associated ErbB4 isoform promote ligand-independent survival and cancer cell growth. Mol Biol Cell. 2006;17:67–79. doi: 10.1091/mbc.E05-05-0402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069–75. doi: 10.1038/nature07423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Cho HS, Leahy DJ. Structure of the extracellular region of HER3 reveals an interdomain tether. Science. 2002;297:1330–3. doi: 10.1126/science.1074611. [DOI] [PubMed] [Google Scholar]
- 28.Garrett TP, McKern NM, Lou M, Elleman TC, Adams TE, Lovrecz GO, et al. Crystal structure of a truncated epidermal growth factor receptor extracellular domain bound to transforming growth factor alpha. Cell. 2002;110:763–73. doi: 10.1016/s0092-8674(02)00940-6. [DOI] [PubMed] [Google Scholar]
- 29.Ogiso H, Ishitani R, Nureki O, Fukai S, Yamanaka M, Kim JH, et al. Crystal structure of the complex of human epidermal growth factor and receptor extracellular domains. Cell. 2002;110:775–87. doi: 10.1016/s0092-8674(02)00963-7. [DOI] [PubMed] [Google Scholar]
- 30.Dawson JP, Bu Z, Lemmon MA. Ligand-induced structural transitions in ErbB receptor extracellular domains. Structure. 2007;15:942–54. doi: 10.1016/j.str.2007.06.013. [DOI] [PubMed] [Google Scholar]
- 31.Lee JC, Vivanco I, Beroukhim R, Huang JH, Feng WL, DeBiasi RM, et al. Epidermal growth factor receptor activation in glioblastoma through novel missense mutations in the extracellular domain. PLoS Med. 2006;3:485. doi: 10.1371/journal.pmed.0030485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Prickett TD, Agrawal NS, Wei X, Yates KE, Lin JC, Wunderlich JR, et al. Analysis of the tyrosine kinome in melanoma reveals recurrent mutations in ERBB4. Nat Genet. 2009;41:1127–32. doi: 10.1038/ng.438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Leahey D. Personal communication. 2010.
- 34.Jones JT, Akita RW, Sliwkowski MX. Binding specificities and affinities of egf domains for ErbB receptors. FEBS Lett. 1999;447:227–31. doi: 10.1016/s0014-5793(99)00283-5. [DOI] [PubMed] [Google Scholar]
- 35.Meshinchi S, Appelbaum FR. Structural and functional alterations of FLT3 in acute myeloid leukemia. Clin Cancer Res. 2009;15:4263–9. doi: 10.1158/1078-0432.CCR-08-1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Schulze WX, Deng L, Mann M. Phosphotyrosine interactome of the ErbB-receptor kinase family. Mol Syst Biol. 2005;1:2005–8. doi: 10.1038/msb4100012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Sergina NV, Rausch M, Wang D, Blair J, Hann B, Shokat KM, et al. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature. 2007;445:437–41. doi: 10.1038/nature05474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wheeler DL, Huang S, Kruser TJ, Nechrebecki MM, Armstrong EA, Benavente S, et al. Mechanisms of acquired resistance to cetuximab: role of HER (ErbB) family members. Oncogene. 2008;27:3944–56. doi: 10.1038/onc.2008.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Gelmon KAFP, Verma S, Wardley AM, Conte PF, Miles D, Gianni L, et al. Results of a phase II trial of trastuzumab (H) and pertuzumab (P) in patients (pts) with HER2-positive metastatic breast cancer (MBC) who had progressed during trastuzumab therapy. J Clin Oncol. 2008:26. doi: 10.1200/JCO.2009.24.2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.O’Shaughnessy JBK, Burstein H, Storniolo AM, Sledge G, Baselga J, Koehler M, et al. A randomized study of lapatinib alone or in combination with trastuzumab in heavily pretreated HER2+ metastatic breast cancer progressing on trastuzumab therapy. J Clin Oncol. 2008:26. doi: 10.1200/JCO.2008.21.4437. [DOI] [PubMed] [Google Scholar]
- 41.Baselga JCD, Miles D, Verma S, Climent M, Ross G, Gimenez V, Gelmon K. Objective response rate in a phase II multicenter trial of pertuzumab (P), a HER2 dimerization inhibiting monoclonal antibody, in combination with trastuzumab (T) in patients (pts) with HER2-positive metastatic breast cancer (MBC) which has progressed during treatment with T. J Clin Oncol. 2007;25:18. [Google Scholar]
- 42.Portera CC, Walshe JM, Rosing DR, Denduluri N, Berman AW, Vatas U, et al. Cardiac toxicity and efficacy of trastuzumab combined with pertuzumab in patients with [corrected] human epidermal growth factor receptor 2-positive metastatic breast cancer. Clin Cancer Res. 2008;14:2710–6. doi: 10.1158/1078-0432.CCR-07-4636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Serra V, Markman B, Scaltriti M, Eichhorn PJ, Valero V, Guzman M, et al. NVP-BEZ235, a dual PI3K/mTOR inhibitor, prevents PI3K signaling and inhibits the growth of cancer cells with activating PI3K mutations. Cancer Res. 2008;68:8022–30. doi: 10.1158/0008-5472.CAN-08-1385. [DOI] [PubMed] [Google Scholar]
- 44.Eichhorn PJ, Gili M, Scaltriti M, Serra V, Guzman M, Nijkamp W, et al. Phosphatidylinositol 3-kinase hyper-activation results in lapatinib resistance that is reversed by the mTOR/phosphatidylinositol 3-kinase inhibitor NVP-BEZ235. Cancer Res. 2008;68:9221–30. doi: 10.1158/0008-5472.CAN-08-1740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Johansson M, Persson JL. Cancer therapy: targeting cell cycle regulators. Anticancer Agents Med Chem. 2008;8:723–31. doi: 10.2174/187152008785914833. [DOI] [PubMed] [Google Scholar]
- 46.Desmond RA, Soong SJ. Epidemiology of malignant melanoma. Surg Clin North Am. 2003;83:1–29. doi: 10.1016/s0039-6109(02)00092-0. [DOI] [PubMed] [Google Scholar]
- 47.Gogas HJ, Kirkwood JM, Sondak VK. Chemotherapy for metastatic melanoma: time for a change? Cancer. 2007;109:455–64. doi: 10.1002/cncr.22427. [DOI] [PubMed] [Google Scholar]
- 48.Atkins MB, Kunkel L, Sznol M, Rosenberg SA. High-dose recombinant interleukin-2 therapy in patients with metastatic melanoma: long-term survival update. Cancer J Sci Am. 2000;6:11–4. [PubMed] [Google Scholar]
- 49.Haluska F, Pemberton T, Ibrahim N, Kalinsky K. The RTK/RAS/BRAF/PI3K pathways in melanoma: biology, small molecule inhibitors and potential applications. Semin Oncol. 2007;34:546–54. doi: 10.1053/j.seminoncol.2007.09.011. [DOI] [PubMed] [Google Scholar]
- 50.Flaherty KPI, Sosman J, Kim K, Ribas A, McArthur G, Lee RJ, et al. Phase I study of PLX4032: Proof of concept for V600E BRAF mutation as a therapeutic target in human cancer. J Clin Oncol. 2009;27:15. [Google Scholar]
- 51.Carvajal RDCP, Wolchok JD, Cane L, Teitcher JB, Lutzky J, Pavlick AC, et al. A phase II study of imatinib mesylate (IM) for patients with advanced melanoma harboring somatic alterations of KIT. J Clin Oncol. 2009;27:15. [Google Scholar]
- 52.Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol. 2006;24:4340–6. doi: 10.1200/JCO.2006.06.2984. [DOI] [PubMed] [Google Scholar]
- 53.Tsai J, Lee JT, Wang W, Zhang J, Cho H, Mamo S, et al. Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc Natl Acad Sci USA. 2008;105:3041–6. doi: 10.1073/pnas.0711741105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Futreal PA, Coin L, Marshall M, Down T, Hubbard T, Wooster R, et al. A census of human cancer genes. Nat Rev Cancer. 2004;4:177–83. doi: 10.1038/nrc1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Sawyers C. Targeted cancer therapy. Nature. 2004;432:294–7. doi: 10.1038/nature03095. [DOI] [PubMed] [Google Scholar]
- 56.Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, et al. The consensus coding sequences of human breast and colorectal cancers. Science. 2006;314:268–74. doi: 10.1126/science.1133427. [DOI] [PubMed] [Google Scholar]
- 57.Soung YH, Lee JW, Kim SY, Wang YP, Jo KH, Moon SW, et al. Somatic mutations of the ERBB4 kinase domain in human cancers. Int J Cancer. 2006;118:1426–9. doi: 10.1002/ijc.21507. [DOI] [PubMed] [Google Scholar]
- 58.Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, et al. Patterns of somatic mutation in human cancer genomes. Nature. 2007;446:153–8. doi: 10.1038/nature05610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554. doi: 10.1126/science.1096502. [DOI] [PubMed] [Google Scholar]
- 60.Elenius K. Personal communication. 2010.
- 61.Elenius K, Choi CJ, Paul S, Santiestevan E, Nishi E, Klagsbrun M. Characterization of a naturally occurring ErbB4 isoform that does not bind or activate phosphatidyl inositol 3-kinase. Oncogene. 1999;18:2607–15. doi: 10.1038/sj.onc.1202612. [DOI] [PubMed] [Google Scholar]
- 62.Bos JL, Fearon ER, Hamilton SR, Verlaande Vries M, van Boom JH, van der Eb AJ, et al. Prevalence of ras gene mutations in human colorectal cancers. Nature. 1987;327:293–7. doi: 10.1038/327293a0. [DOI] [PubMed] [Google Scholar]
- 63.Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–54. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
- 64.Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L, O’Brien JM, et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature. 2009;457:599–602. doi: 10.1038/nature07586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Davies H, Hunter C, Smith R, Stephens P, Greenman C, Bignell G, et al. Somatic mutations of the protein kinase gene family in human lung cancer. Cancer Res. 2005;65:7591–5. doi: 10.1158/0008-5472.CAN-05-1855. [DOI] [PubMed] [Google Scholar]
- 66.Parsons DW, Wang TL, Samuels Y, Bardelli A, Cummins JM, DeLong L, et al. Colorectal cancer: mutations in a signalling pathway. Nature. 2005;436:792. doi: 10.1038/436792a. [DOI] [PubMed] [Google Scholar]
- 67.Sundvall M, Korhonen A, Paatero I, Gaudio E, Melino G, Croce CM, et al. Isoform-specific monoubiquitination, endocytosis and degradation of alternatively spliced ErbB4 isoforms. Proc Natl Acad Sci USA. 2008;105:4162–7. doi: 10.1073/pnas.0708333105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Williams EE, Trout LJ, Gallo RM, Pitfield SE, Bryant I, Penington DJ, et al. A constitutively active ErbB4 mutant inhibits drug-resistant colony formation by the DU-145 and PC-3 human prostate tumor cell lines. Cancer Lett. 2003;192:67–74. doi: 10.1016/s0304-3835(02)00690-0. [DOI] [PubMed] [Google Scholar]
- 69.Muraoka-Cook RS, Sandahl MA, Strunk KE, Miraglia LC, Husted C, Hunter DM, et al. ErbB4 splice variants Cyt1 and Cyt2 differ by 16 amino acids and exert opposing effects on the mammary epithelium in vivo. Mol Cell Biol. 2009;29:4935–48. doi: 10.1128/MCB.01705-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Tang CK, Goldstein DJ, Payne J, Czubayko F, Alimandi M, Wang LM, et al. ErbB-4 ribozymes abolish neuregulin-induced mitogenesis. Cancer Res. 1998;58:3415–22. [PubMed] [Google Scholar]
- 71.Inoue A, Kobayashi K, Usui K, Maemondo M, Okinaga S, Mikami I, et al. First-line gefitinib for patients with advanced non-small-cell lung cancer harboring epidermal growth factor receptor mutations without indication for chemotherapy. J Clin Oncol. 2009;27:1394–400. doi: 10.1200/JCO.2008.18.7658. [DOI] [PubMed] [Google Scholar]
- 72.Mellinghoff IK, Wang MY, Vivanco I, Haas-Kogan DA, Zhu S, Dia EQ, et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med. 2005;353:2012–24. doi: 10.1056/NEJMoa051918. [DOI] [PubMed] [Google Scholar]
- 73.Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, Thongprasert S, et al. Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med. 2005;353:123–32. doi: 10.1056/NEJMoa050753. [DOI] [PubMed] [Google Scholar]
- 74.Tice DA, Biscardi JS, Nickles AL, Parsons SJ. Mechanism of biological synergy between cellular Src and epidermal growth factor receptor. Proc Natl Acad Sci USA. 1999;96:1415–20. doi: 10.1073/pnas.96.4.1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Huse M, Kuriyan J. The conformational plasticity of protein kinases. Cell. 2002;109:275–82. doi: 10.1016/s0092-8674(02)00741-9. [DOI] [PubMed] [Google Scholar]
- 76.Zhang X, Gureasko J, Shen K, Cole PA, Kuriyan J. An allosteric mechanism for activation of the kinase domain of epidermal growth factor receptor. Cell. 2006;125:1137–49. doi: 10.1016/j.cell.2006.05.013. [DOI] [PubMed] [Google Scholar]
- 77.Bose R, Zhang X. The ErbB kinase domain: structural perspectives into kinase activation and inhibition. Exp Cell Res. 2009;315:649–58. doi: 10.1016/j.yexcr.2008.07.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Yun CH, Boggon TJ, Li Y, Woo MS, Greulich H, Meyerson M, et al. Structures of lung cancer-derived EGFR mutants and inhibitor complexes: mechanism of activation and insights into differential inhibitor sensitivity. Cancer Cell. 2007;11:217–27. doi: 10.1016/j.ccr.2006.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Jiang J, Greulich H, Janne PA, Sellers WR, Meyerson M, Griffin JD. Epidermal growth factor-independent transformation of Ba/F3 cells with cancer-derived epidermal growth factor receptor mutants induces gefitinib-sensitive cell cycle progression. Cancer Res. 2005;65:8968–74. doi: 10.1158/0008-5472.CAN-05-1829. [DOI] [PubMed] [Google Scholar]
- 80.Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6. doi: 10.1126/science.1164368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Wood LD, Parsons DW, Jones S, Lin J, Sjoblom T, Leary RJ, et al. The genomic landscapes of human breast and colorectal cancers. Science. 2007;318:1108–13. doi: 10.1126/science.1145720. [DOI] [PubMed] [Google Scholar]
- 82.Leidner RS, Fu P, Clifford B, Hamdan A, Jin C, Eisenberg R, et al. Genetic abnormalities of the EGFR pathway in African American Patients with non-small-cell lung cancer. J Clin Oncol. 2009;27:5620–6. doi: 10.1200/JCO.2009.23.1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.McDermott U, Settleman J. Personalized cancer therapy with selective kinase inhibitors: an emerging paradigm in medical oncology. J Clin Oncol. 2009;27:5650–9. doi: 10.1200/JCO.2009.22.9054. [DOI] [PubMed] [Google Scholar]
- 84.Franklin MC, Carey KD, Vajdos FF, Leahy DJ, de Vos AM, Sliwkowski MX. Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell. 2004;5:317–28. doi: 10.1016/s1535-6108(04)00083-2. [DOI] [PubMed] [Google Scholar]
- 85.Hubbard SR. EGF receptor inhibition: attacks on multiple fronts. Cancer Cell. 2005;7:287–8. doi: 10.1016/j.ccr.2005.04.004. [DOI] [PubMed] [Google Scholar]
- 86.Agus DB, Akita RW, Fox WD, Lewis GD, Higgins B, Pisacane PI, et al. Targeting ligand-activated ErbB2 signaling inhibits breast and prostate tumor growth. Cancer Cell. 2002;2:127–37. doi: 10.1016/s1535-6108(02)00097-1. [DOI] [PubMed] [Google Scholar]
- 87.Baselga J. A new anti-ErbB2 strategy in the treatment of cancer: prevention of ligand-dependent ErbB2 receptor heterodimerization. Cancer Cell. 2002;2:93–5. doi: 10.1016/s1535-6108(02)00098-3. [DOI] [PubMed] [Google Scholar]
- 88.Mullen P, Cameron DA, Hasmann M, Smyth JF, Langdon SP. Sensitivity to pertuzumab (2C4) in ovarian cancer models: cross-talk with estrogen receptor signaling. Mol Cancer Ther. 2007;6:93–100. doi: 10.1158/1535-7163.MCT-06-0401. [DOI] [PubMed] [Google Scholar]
- 89.Mendoza N, Phillips GL, Silva J, Schwall R, Wickramasinghe D. Inhibition of ligand-mediated HER2 activation in androgen-independent prostate cancer. Cancer Res. 2002;62:5485–8. [PubMed] [Google Scholar]
- 90.Sakai K, Yokote H, Murakami-Murofushi K, Tamura T, Saijo N, Nishio K. Pertuzumab, a novel HER dimerization inhibitor, inhibits the growth of human lung cancer cells mediated by the HER3 signaling pathway. Cancer Sci. 2007;98:1498–503. doi: 10.1111/j.1349-7006.2007.00553.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Mann M, Sheng H, Shao J, Williams CS, Pisacane PI, Sliwkowski MX, et al. Targeting cyclooxygenase 2 and HER-2/neu pathways inhibits colorectal carcinoma growth. Gastroenterology. 2001;120:1713–9. doi: 10.1053/gast.2001.24844. [DOI] [PubMed] [Google Scholar]
- 92.Cortes JBJ, Kellokumpu-Lehtinen P, Bianchi G, Cameron D, Miles D, Salvagni S, et al. Open label, randomized, phase II study of pertuzumab (P) in patients (pts) with metastatic breast cancer (MBC) with low expression of HER2. J Clin Oncol. 2005;23:16. [Google Scholar]
- 93.Ocana A, Amir E. Irreversible pan-ErbB tyrosine kinase inhibitors and breast cancer: Current status and future directions. Cancer Treat Rev. 2009;35:685–91. doi: 10.1016/j.ctrv.2009.08.001. [DOI] [PubMed] [Google Scholar]
- 94.Lee PAD, Avrutskaya A, White A, Pheneger T, Winkler J. In vivo activity of ARRAY-334543, a potent, small molecule inhibitor of EGFR/ErbB-2 in combination with trastuzumab or docetaxol. Proceedings of the 99th Annual Meeting of the American Association for Cancer Research (AACR); 2008; AACR. 2008. [Google Scholar]
- 95.Iwahara ATT. In vivo antitumor efficacy of TAK-285, a novel ErbB1/ErbB2 dual kinase inhibitor. Proceedings of the 20th EORTC-NCI-AACR Symposium on Molecular Targets and Cancer Therapeutics; 2008. p. 311. [Google Scholar]
- 96.Engelman JA, Settleman J. Acquired resistance to tyrosine kinase inhibitors during cancer therapy. Curr Opin Genet Dev. 2008;18:73–9. doi: 10.1016/j.gde.2008.01.004. [DOI] [PubMed] [Google Scholar]
- 97.Dematteo RP, Ballman KV, Antonescu CR, Maki RG, Pisters PW, Demetri GD, et al. Adjuvant imatinib mesylate after resection of localised, primary gastrointestinal stromal tumour: a randomised, double-blind, placebo-controlled trial. Lancet. 2009;373:1097–104. doi: 10.1016/S0140-6736(09)60500-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Zhu CQ, da Cunha Santos G, Ding K, Sakurada A, Cutz JC, Liu N, et al. Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21. J Clin Oncol. 2008;26:4268–75. doi: 10.1200/JCO.2007.14.8924. [DOI] [PubMed] [Google Scholar]
- 99.Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ, Tu D, Tebbutt NC, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359:1757–65. doi: 10.1056/NEJMoa0804385. [DOI] [PubMed] [Google Scholar]
- 100.Loupakis F, Pollina L, Stasi I, Ruzzo A, Scartozzi M, Santini D, et al. PTEN expression and KRAS mutations on primary tumors and metastases in the prediction of benefit from cetuximab plus irinotecan for patients with metastatic colorectal cancer. J Clin Oncol. 2009;27:2622–9. doi: 10.1200/JCO.2008.20.2796. [DOI] [PubMed] [Google Scholar]
- 101.Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, et al. Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol. 2008;26:5705–12. doi: 10.1200/JCO.2008.18.0786. [DOI] [PubMed] [Google Scholar]
- 102.Sartore-Bianchi A, Martini M, Molinari F, Veronese S, Nichelatti M, Artale S, et al. PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies. Cancer Res. 2009;69:1851–7. doi: 10.1158/0008-5472.CAN-08-2466. [DOI] [PubMed] [Google Scholar]
- 103.Sholl LM, Yeap BY, Iafrate AJ, Holmes-Tisch AJ, Chou YP, Wu MT, et al. Lung adenocarcinoma with EGFR amplification has distinct clinicopathologic and molecular features in never-smokers. Cancer Res. 2009;69:8341–8. doi: 10.1158/0008-5472.CAN-09-2477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Frattini M, Saletti P, Romagnani E, Martin V, Molinari F, Ghisletta M, et al. PTEN loss of expression predicts cetuximab efficacy in metastatic colorectal cancer patients. Br J Cancer. 2007;97:1139–45. doi: 10.1038/sj.bjc.6604009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Wang SE, Narasanna A, Perez-Torres M, Xiang B, Wu FY, Yang S, et al. HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer Cell. 2006;10:25–38. doi: 10.1016/j.ccr.2006.05.023. [DOI] [PubMed] [Google Scholar]
- 106.Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2005;2:73. doi: 10.1371/journal.pmed.0020073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Costa DB, Schumer ST, Tenen DG, Kobayashi S. Differential responses to erlotinib in epidermal growth factor receptor (EGFR)-mutated lung cancers with acquired resistance to gefitinib carrying the L747S or T790M secondary mutations. J Clin Oncol. 2008;26:1182–4. doi: 10.1200/JCO.2007.14.9039. [DOI] [PubMed] [Google Scholar]
- 108.Gazdar AF. Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene. 2009;28:24–31. doi: 10.1038/onc.2009.198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Cools J, DeAngelo DJ, Gotlib J, Stover EH, Legare RD, Cortes J, et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome. N Engl J Med. 2003;348:1201–14. doi: 10.1056/NEJMoa025217. [DOI] [PubMed] [Google Scholar]
- 110.Tamborini E, Bonadiman L, Greco A, Albertini V, Negri T, Gronchi A, et al. A new mutation in the KIT ATP pocket causes acquired resistance to imatinib in a gastrointestinal stromal tumor patient. Gastroenterology. 2004;127:294–9. doi: 10.1053/j.gastro.2004.02.021. [DOI] [PubMed] [Google Scholar]
- 111.Antonescu CR, Besmer P, Guo T, Arkun K, Hom G, Koryotowski B, et al. Acquired resistance to imatinib in gastrointestinal stromal tumor occurs through secondary gene mutation. Clin Cancer Res. 2005;11:4182–90. doi: 10.1158/1078-0432.CCR-04-2245. [DOI] [PubMed] [Google Scholar]
- 112.Citri A, Kochupurakkal BS, Yarden Y. The achilles heel of ErbB-2/HER2: regulation by the Hsp90 chaperone machine and potential for pharmacological intervention. Cell Cycle. 2004;3:51–60. [PubMed] [Google Scholar]
- 113.Gaspar N, Sharp SY, Pacey S, Jones C, Walton M, Vassal G, et al. Acquired resistance to 17-allylamino-17-demethoxygeldanamycin (17-AAG, tanespimycin) in glioblastoma cells. Cancer Res. 2009;69:1966–75. doi: 10.1158/0008-5472.CAN-08-3131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Bean J, Brennan C, Shih JY, Riely G, Viale A, Wang L, et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci USA. 2007;104:20932–7. doi: 10.1073/pnas.0710370104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Cappuzzo F, Janne PA, Skokan M, Finocchiaro G, Rossi E, Ligorio C, et al. MET increased gene copy number and primary resistance to gefitinib therapy in non-small-cell lung cancer patients. Ann Oncol. 2009;20:298–304. doi: 10.1093/annonc/mdn635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Cappuzzo F, Marchetti A, Skokan M, Rossi E, Gajapathy S, Felicioni L, et al. Increased MET gene copy number negatively affects survival of surgically resected non-small-cell lung cancer patients. J Clin Oncol. 2009;27:1667–74. doi: 10.1200/JCO.2008.19.1635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–43. doi: 10.1126/science.1141478. [DOI] [PubMed] [Google Scholar]
- 118.Buck E, Eyzaguirre A, Rosenfeld-Franklin M, Thomson S, Mulvihill M, Barr S, et al. Feedback mechanisms promote cooperativity for small molecule inhibitors of epidermal and insulin-like growth factor receptors. Cancer Res. 2008;68:8322–32. doi: 10.1158/0008-5472.CAN-07-6720. [DOI] [PubMed] [Google Scholar]
- 119.Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer. 2009;9:28–39. doi: 10.1038/nrc2559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol. 2009;27:1160–7. doi: 10.1200/JCO.2008.18.1370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Innocenti F, Undevia SD, Iyer L, Chen PX, Das S, Kocherginsky M, et al. Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol. 2004;22:1382–8. doi: 10.1200/JCO.2004.07.173. [DOI] [PubMed] [Google Scholar]
- 122.Michael M, Thompson M, Hicks RJ, Mitchell PL, Ellis A, Milner AD, et al. Relationship of hepatic functional imaging to irinotecan pharmacokinetics and genetic parameters of drug elimination. J Clin Oncol. 2006;24:4228–35. doi: 10.1200/JCO.2005.04.8496. [DOI] [PubMed] [Google Scholar]
- 123.Golub TR. Genome-wide views of cancer. N Engl J Med. 2001;344:601–2. doi: 10.1056/NEJM200102223440809. [DOI] [PubMed] [Google Scholar]
- 124.Liu R, Wang X, Chen GY, Dalerba P, Gurney A, Hoey T, et al. The prognostic role of a gene signature from tumorigenic breast-cancer cells. N Engl J Med. 2007;356:217–26. doi: 10.1056/NEJMoa063994. [DOI] [PubMed] [Google Scholar]
- 125.Potti A, Mukherjee S, Petersen R, Dressman HK, Bild A, Koontz J, et al. A genomic strategy to refine prognosis in early-stage non-small-cell lung cancer. N Engl J Med. 2006;355:570–80. doi: 10.1056/NEJMoa060467. [DOI] [PubMed] [Google Scholar]
- 126.Stephens P, Hunter C, Bignell G, Edkins S, Davies H, Teague J, et al. Lung cancer: intragenic ERBB2 kinase mutations in tumours. Nature. 2004;431:525–6. doi: 10.1038/431525b. [DOI] [PubMed] [Google Scholar]
- 127.Shigematsu H, Takahashi T, Nomura M, Majmudar K, Suzuki M, Lee H, et al. Somatic mutations of the HER2 kinase domain in lung adenocarcinomas. Cancer Res. 2005;65:1642–6. doi: 10.1158/0008-5472.CAN-04-4235. [DOI] [PubMed] [Google Scholar]
- 128.Sasaki H, Endo K, Konishi A, Takada M, Kawahara M, Iuchi K, et al. EGFR Mutation status in Japanese lung cancer patients: genotyping analysis using LightCycler. Clin Cancer Res. 2005;11:2924–9. doi: 10.1158/1078-0432.CCR-04-1904. [DOI] [PubMed] [Google Scholar]
- 129.Sasaki H, Shimizu S, Endo K, Takada M, Kawahara M, Tanaka H, et al. EGFR and erbB2 mutation status in Japanese lung cancer patients. Int J Cancer. 2006;118:180–4. doi: 10.1002/ijc.21301. [DOI] [PubMed] [Google Scholar]
- 130.Blons H, Cote JF, Le Corre D, Riquet M, Fabre-Guilevin E, Laurent-Puig P, et al. Epidermal growth factor receptor mutation in lung cancer are linked to bronchioloalveolar differentiation. Am J Surg Pathol. 2006;30:1309–15. doi: 10.1097/01.pas.0000213285.65907.31. [DOI] [PubMed] [Google Scholar]
- 131.Lee JW, Soung YH, Kim SY, Nam SW, Park WS, Wang YP, et al. ERBB2 kinase domain mutation in the lung squamous cell carcinoma. Cancer Lett. 2006;237:89–94. doi: 10.1016/j.canlet.2005.05.026. [DOI] [PubMed] [Google Scholar]
- 132.Jeong EG, Soung YH, Lee JW, Lee SH, Nam SW, Lee JY, et al. ERBB3 kinase domain mutations are rare in lung, breast and colon carcinomas. Int J Cancer. 2006;119:2986–7. doi: 10.1002/ijc.22257. [DOI] [PubMed] [Google Scholar]
- 133.Marks JL, McLellan MD, Zakowski MF, Lash AE, Kasai Y, Broderick S, et al. Mutational analysis of EGFR and related signaling pathway genes in lung Adenocarcinomas identifies a novel somatic kinase domain mutation in FGFR4. PLoS One. 2007;2:426. doi: 10.1371/journal.pone.0000426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Bae NC, Chae MH, Lee MH, Kim KM, Lee EB, Kim CH, et al. EGFR, ERBB2 and KRAS mutations in Korean non-small cell lung cancer patients. Cancer Genet Cytogenet. 2007;173:107–13. doi: 10.1016/j.cancergencyto.2006.10.007. [DOI] [PubMed] [Google Scholar]
- 135.Gu D, Scaringe WA, Li K, Saldivar JS, Hill KA, Chen Z, et al. Database of somatic mutations in EGFR with analyses revealing indel hotspots but no smoking-associated signature. Hum Mutat. 2007;28:760–70. doi: 10.1002/humu.20512. [DOI] [PubMed] [Google Scholar]
- 136.Lee JW, Soung YH, Seo SH, Kim SY, Park CH, Wang YP, et al. Somatic mutations of ERBB2 kinase domain in gastric, colorectal and breast carcinomas. Clin Cancer Res. 2006;12:57–61. doi: 10.1158/1078-0432.CCR-05-0976. [DOI] [PubMed] [Google Scholar]
- 137.Tvorogov D, Sundvall M, Kurppa K, Hollmen M, Repo S, Johnson MS, et al. Somatic mutations of ErbB4: selective loss-of-function phenotype affecting signal transduction pathways in cancer. J Biol Chem. 2009;284:5582–91. doi: 10.1074/jbc.M805438200. [DOI] [PubMed] [Google Scholar]
- 138.Kubo T, Kuroda Y, Shimizu H, Kokubu A, Okada N, Hosoda F, et al. Resequencing and copy number analysis of the human tyrosine kinase gene family in poorly differentiated gastric cancer. Carcinogenesis. 2009;30:1857–64. doi: 10.1093/carcin/bgp206. [DOI] [PubMed] [Google Scholar]
- 139.Lassus H, Sihto H, Leminen A, Joensuu H, Isola J, Nupponen NN, et al. Gene amplification, mutation and protein expression of EGFR and mutations of ERBB2 in serous ovarian carcinoma. J Mol Med. 2006;84:671–81. doi: 10.1007/s00109-006-0054-4. [DOI] [PubMed] [Google Scholar]
- 140.Schilder RJ, Sill MW, Chen X, Darcy KM, Decesare SL, Lewandowski G, et al. Phase II study of gefitinib in patients with relapsed or persistent ovarian or primary peritoneal carcinoma and evaluation of epidermal growth factor receptor mutations and immunohistochemical expression: a Gynecologic Oncology Group Study. Clin Cancer Res. 2005;11:5539–48. doi: 10.1158/1078-0432.CCR-05-0462. [DOI] [PubMed] [Google Scholar]
- 141.Bekaii-Saab T, Williams N, Plass C, Calero MV, Eng C. A novel mutation in the tyrosine kinase domain of ERBB2 in hepatocellular carcinoma. BMC Cancer. 2006;6:278. doi: 10.1186/1471-2407-6-278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Nagahara H, Mimori K, Ohta M, Utsunomiya T, Inoue H, Barnard GF, et al. Somatic mutations of epidermal growth factor receptor in colorectal carcinoma. Clin Cancer Res. 2005;11:1368–71. doi: 10.1158/1078-0432.CCR-04-1894. [DOI] [PubMed] [Google Scholar]
- 143.Lee JW, Soung YH, Kim SY, Nam HK, Park WS, Nam SW, et al. Somatic mutations of EGFR gene in squamous cell carcinoma of the head and neck. Clin Cancer Res. 2005;11:2879–82. doi: 10.1158/1078-0432.CCR-04-2029. [DOI] [PubMed] [Google Scholar]
- 144.Cohen EE, Lingen MW, Martin LE, Harris PL, Brannigan BW, Haserlat SM, et al. Response of some head and neck cancers to epidermal growth factor receptor tyrosine kinase inhibitors may be linked to mutation of ERBB2 rather than EGFR. Clin Cancer Res. 2005;11:8105–8. doi: 10.1158/1078-0432.CCR-05-0926. [DOI] [PubMed] [Google Scholar]
- 145.Loeffler-Ragg J, Witsch-Baumgartner M, Tzankov A, Hilbe W, Schwentner I, Sprinzl GM, et al. Low incidence of mutations in EGFR kinase domain in Caucasian patients with head and neck squamous cell carcinoma. Eur J Cancer. 2006;42:109–11. doi: 10.1016/j.ejca.2005.08.034. [DOI] [PubMed] [Google Scholar]
- 146.Na II, Kang HJ, Cho SY, Koh JS, Lee JK, Lee BC, et al. EGFR mutations and human papillomavirus in squamous cell carcinoma of tongue and tonsil. Eur J Cancer. 2007;43:520–6. doi: 10.1016/j.ejca.2006.09.025. [DOI] [PubMed] [Google Scholar]
- 147.Kwak EL, Jankowski J, Thayer SP, Lauwers GY, Brannigan BW, Harris PL, et al. Epidermal growth factor receptor kinase domain mutations in esophageal and pancreatic adenocarcinomas. Clin Cancer Res. 2006;12:4283–7. doi: 10.1158/1078-0432.CCR-06-0189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]




