Skip to main content
Erschienen in: Cancer and Metastasis Reviews 3/2015

Open Access 01.09.2015 | Clinical

Fibroblast growth factor receptor signaling in hereditary and neoplastic disease: biologic and clinical implications

verfasst von: Teresa Helsten, Maria Schwaederle, Razelle Kurzrock

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 3/2015

Abstract

Fibroblast growth factors (FGFs) and their receptors (FGFRs) are transmembrane growth factor receptors with wide tissue distribution. FGF/FGFR signaling is involved in neoplastic behavior and also development, differentiation, growth, and survival. FGFR germline mutations (activating) can cause skeletal disorders, primarily dwarfism (generally mutations in FGFR3), and craniofacial malformation syndromes (usually mutations in FGFR1 and FGFR2); intriguingly, some of these activating FGFR mutations are also seen in human cancers. FGF/FGFR aberrations reported in cancers are mainly thought to be gain-of-function changes, and several cancers have high frequencies of FGFR alterations, including breast, bladder, or squamous cell carcinomas (lung and head and neck). FGF ligand aberrations (predominantly gene amplifications) are also frequently seen in cancers, in contrast to hereditary syndromes. There are several pharmacologic agents that have been or are being developed for inhibition of FGFR/FGF signaling. These include both highly selective inhibitors as well as multi-kinase inhibitors. Of note, only four agents (ponatinib, pazopanib, regorafenib, and recently lenvatinib) are FDA-approved for use in cancer, although the approval was not based on their activity against FGFR. Perturbations in the FGFR/FGF signaling are present in both inherited and malignant diseases. The development of potent inhibitors targeting FGF/FGFR may provide new tools against disorders caused by FGF/FGFR alterations.
Hinweise
Teresa Helsten and Maria Schwaederle contributed equally to this work.

1 Introduction

One of the most important advances in tumor biology is the recognition that cancer is frequently driven by inherited or acquired alterations in specific gene(s) or their products [1, 2]. Genomic alterations include changes in expression that can result from mutation, deletion, gene amplification, and/or translocation. Complicating matters, cancers often harbor multiple genetic alterations, but one or a few of these are thought to be primarily responsible for neoplastic behavior in any given tumor. These are the so-called “driver mutations,” while “passenger mutations” may have a more nuanced impact. A literature review suggests that over 1 % of human genes can be implicated as cancer drivers when they are mutated, with protein kinases comprising the largest subgroup of genes altered [3].
Among human signaling pathways, fibroblast growth factor (FGF)/fibroblast growth factor receptor (FGFR) is one of the pathways most enriched in non-synonymous mutations, including several candidate driver mutations [3]. A computational method designed to identify driver mutations within protein kinase datasets successfully identified multiple aberrations in the FGF/FGFR machinery [4]. In keeping with other genes implicated in neoplastic behavior, FGF/FGFR signaling is also involved in development, differentiation, growth, and survival mechanisms. Indeed, FGFR aberrations have been identified in both hereditary and neoplastic human diseases.
Most of the reported FGFR mutations that cause heritable human diseases are activating mutations which increase receptor signaling. These abnormalities are seen in craniofacial and skeletal syndromes such as the craniosynostoses [57] (Pfeiffer, Crouzon, Apert, Jackson-Weiss, Muenke, and Beare-Stevenson syndromes) and dwarfism syndromes [8, 9] (achondroplasia, thanatophoric dysplasia, and hypochondroplasia). The only reported inherited condition caused by loss of FGFR function is an autosomal dominant form of hereditary hypogonatotrophic hypogonadism 2 with or without anosmia [10, 11], which is caused by loss of function of FGFR1 [10, 12, 13] or a missense mutation in FGF8 [14]. Intriguingly, some of the same activating FGFR mutations seen in inherited syndromes are also seen in human cancers [14, 15]. Furthermore, FGF/FGFR aberrations reported in cancers are overwhelmingly thought to be gain-of-function changes, including gene amplifications and gene rearrangements [16].
The goal of identification and characterization of driver mutations in cancer is, ultimately, to create successful anti-cancer therapies with which to prosecute these tumors; several such therapies already exist, demonstrating proof of principle [17, 18]. Furthermore, for some gene targets, drugs may impact the course of cancer as well as non-malignant conditions that are driven by abnormalities in the cognate signal. JAK2 aberrations, for instance, are found in myelofibrosis, and JAK2 inhibitors such as ruxolitinib provide significant benefit in such patients [19]. At the same time, the JAK2 inhibitor tofacitinib can benefit patients with rheumatoid arthritis and is approved for that indication [20]. In the case of FGF/FGFR, multiple drugs targeting this pathway have entered the clinic [16]. Herein, we discuss the landscape of diseases that are driven by aberrant FGF/FGFR machinery.

2 Molecular biology of FGF/FGFR signaling

Fibroblast growth factors (FGFs) and their receptors (FGFRs) are evolutionarily conserved transmembrane growth factor receptors with wide tissue distribution in all vertebrates. FGFs and FGFRs share homologies among their respective groups and with other signaling molecules. FGFRs in particular are similar to other signaling receptors, including vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR), and other receptor tyrosine kinases [21]. However, there are important differences between the individual signaling molecules that allow for precise control of a full range of processes, including development, cell survival, differentiation, motility, angiogenesis, and carcinogenesis.

2.1 Receptors

In humans, there are five known FGFRs, called FGFR1–FGFR4 and FGFRL1 (also known as FGFR5). FGFR1–FGFR4 are typical growth factor receptor tyrosine kinases, with extracellular immunoglobulin (Ig)-like domains and intracellular tyrosine kinase domains, while FGFRL1 lacks the intracellular kinase domain and has less clear function(s)[22, 23]. Upon binding of their ligands, the typical receptors homo- or hetero-dimerize, leading to sequential phosphorylation of specific intracellular tyrosine residues and activation of an intracellular signaling cascade and gene transcription [24] (Fig. 1). The FGFR signaling pathway interacts with several other important intracellular pathways, including PI3K/Akt, Wnt, hedgehog, and bone morphogenic protein (BMP) [24, 25]. FGFR1–4 have different ligand specificities based on developmental aspects, tissue distribution, and RNA splicing variation [26]. For example, the FGFR2b isoform is predominantly expressed in epithelial cells, while the FGFR2c isoform is expressed predominantly in mesenchymal cells, and switching from FGFR2b to FGFR2c occurs during progression and invasion of prostate and bladder cancers [27].

2.2 Ligands

There are 18 human ligands for FGFRs (Fig. 1). They are FGF1 (acidic FGF), FGF2 (basic FGF), FGF3, FGF4, FGF5, FGF6, FGF7 (KGF), FGF8, FGF9, FGF10, FGF16, FGF17, FGF18, FGF19, FGF20, FGF21, FGF22, and FGF23. FGF11–14 are not ligands for FGFRs and are known as FGF homologous factors (FHF1–4) [28, 29]. There is no human FGF15 (FGF15 is the mouse equivalent of human FGF19) [30]. The FGFR ligands are secreted proteins that bind to the extracellular matrix, thereby restricting their influence to the tissue where they are produced (autocrine or paracrine function). However, three of the FGFs—FGF19, FGF21, and FGF23—bind less tightly to extracellular matrix heparin sulfates, so they are able to act systemically in an endocrine fashion (hormone-like), allowing them to spread from their production site into the circulation [30, 31]. Most FGFs are secreted proteins with cleavable amino terminal portions, but FGF9, FGF16, and FGF20 have non-cleavable secretion sequences, and FGF1 and FGF2 have no secretion sequences, although they are found in the extracellular compartment, suggesting an alternate process of release [29, 32]. Most FGFs are released from binding to the extracellular matrix via action of heparin sulfate proteoglycans and/or fibroblast growth factor–binding protein (FGFBP1)[33].

2.3 FGF/FGFR interaction and function

In general, FGFs are promiscuous, and most can bind to any of the four main FGFRs, although some of the FGF/FGFR ligand/receptor pairs differ in their affinities (Fig. 1c). FGFs interact with cell surface heparan sulfate proteoglycans (HSPGs) and with the transmembrane protein Klotho (for the hormone-like FGF19, 20, 23) to stabilize binding to FGFRs. Ornitz et al. [34, 35] systemically investigated affinities of FGFs for each FGFR isoform and showed, for example, that FGFR2b is a high-affinity receptor for FGF1, FGF7, FGF10, and FGF22, while FGFR2c has high affinity for FGF1, FGF2, FGF4, FGF6, FGF8, FGF9, and FGF20. But, differential expression of either FGFs or FGFRs in time or tissue may also contribute to tissue-specific effects of FGF/FGFR signaling. For example, FGF1 and FGF2 are expressed in both embryonic and adult tissues, while FGF7–FGF9 are predominantly expressed in developing or in restricted adult tissues [36].
The differences in activity/function of the different FGF/FGFR pairs are also highlighted by murine gene knockout experiments. Mice heterozygous for FGFR knockout mutations develop normally, so haplo-insufficiency is not likely to be a factor [37]. However, mice homozygous for FGFR1 or FGFR2 null mutations die in utero, and FGFR3-null mice develop normally other than overgrowth of cancellous bones and deafness [37]. Most of the FGFs have also been knocked out in mouse models, with varying phenotypic effects, including lethality (FGF4, FGF8, FGF9, FGF10, FGF19, FGF18, and FGF23), defects in neuromusculoskeletal development or function (FGF2, FGF3, FGF6, FGF7, FGF12, FGF14, FGF17, FGF18), cardiac defects (FGF2, FGF9, FGF16, FGF19), and no identifiable abnormality (FGF1) [29].

2.4 Activation of signaling

To signal, FGFs have to be released from the extracellular matrix by heparinases, proteases, or specific fibroblast growth factor–binding protein (FGFBP1). FGFBP1 are secreted heparin proteins that reversibly bind FGF1 and FGF2, releasing them from the extracellular matrix and increasing the local levels of free ligand available for receptor binding [38]. FGFBP1 is secreted by keratinocytes and human epidermal carcinomas, particularly squamous cell carcinomas [33, 38]. Its expression is seen in development, wound healing, cancer, and angiogenesis, and it is downregulated by pharmacologic agents in vitro [33]. The fibronectin-leucine-rich transmembrane protein 3 (FLRT3) is co-expressed with FGF8 during development, co-immunoprecipitates with FGF8/FGFRs, physically interacts with FGFRs via its fibronectin type III domain, and increases activity through the MAPK pathway, facilitating FGFR signaling [39, 40].

2.5 Inhibition of signaling

FGF/FGFR signaling is negatively regulated by several mechanisms. FGFR stimulation activates sprouty proteins (SPRY1–4), which in turn negatively feedbacks on FGF/FGFR/MAPK signaling by interacting with growth factor receptor bound protein 2 (GRB2), son of sevenless homolog 1 (SoS1), and/or RAF proto-oncogene serine/threonine-protein kinase (RAF1) [41, 42]. Intriguingly, sprouty proteins are thought to be general inhibitors of receptor tyrosine kinase growth factor signaling via inhibition of Ras-MAPK signaling, but their behavior differs among specific growth factor receptors. For example, sprouty protein inhibits FGFR signaling but potentiates epidermal growth factor receptor (EGFR) signaling [43]. The cytoplasmic domain of similar expression to FGF (SEF, also known as IL-17 receptor D) interacts with the cytoplasmic domain of FGFR and inhibits downstream signaling of the pathway [43]. Finally, MAP kinase phosphatases (MKPs) dephosphorylate ERKs and thereby downregulate the signaling pathway. MKP3 functions within the cytoplasm, whereas MKP1 is localized in the nucleus [43].
FGFRL1 is the atypical receptor that lacks an intracellular kinase domain. Like the other FGFRs, it is found in all vertebrates, is expressed in a wide variety of tissues, and binds to some FGF ligands. In vitro experiments show that FGFRL1 binds FGF3, FGF4, FGF8, FGF10, and FGF22, but not the other FGFs tested [44]. Unlike FGFR1–4, it is shed from cell membranes [44], suggesting that it may serve as a ligand trap to negatively regulate signaling [45]. Other regulatory mechanisms might include heterodimerization with other FGFRs to prevent trans-autophophorylation and thereby negatively regulating intracellular signaling or by increasing membrane turnover rates of the other FGFRs, but there is currently no evidence that these mechanisms exist [45]. The exact functions of FGFRL1 are not known. FGFRL1-null mice die in infancy with diaphragmatic defects and renal agenesis [45], and there are case reports of FGFRL1 mutations in human disease: craniosynostosis [46] and ovarian cancer [47].

3 FGF/FGFR aberrant signaling in human disease

3.1 Inherited syndromes

Several types of inherited syndromes are due to germline aberrations in FGFR. These include craniosynostosis syndromes and achondroplasia [48], mainly due to gain-of-function mutations, as well as loss-of-function anomalies associated with congenital hypogonadotropic hypogonadism (Table 1). Interestingly, although some of these aberrations are identical to those that, in somatic form, are associated with cancer, for the most part, the individuals with these hereditary syndromes have not been reported to have an increased incidence of cancer. Apart from case reports [4953], there is no epidemiologic evidence that people with craniosynostosis, dwarfism, or congenital hypogonadotropic hypogonadism are at increased risk for malignancy. This is possibly related to differential effects of activating FGFR mutations in cell or tissue type in the context of stage of development. It is possible, for instance, that there are as yet unknown anti-neoplastic compensatory effects in individuals with these disorders, perhaps stemming from the presence of germline FGFR aberrations during development as opposed to the appearance of somatic FGFR mutations in human cancers.
Table 1
Examples of FGFR aberrations in inherited syndromes
Gene chromosome aberration
Syndrome (OMIM number)
Clinical features
Examples of cancers in which an aberration is seen
Reference(s)
FGFR1 (Chr 8p)
P252R
Pfeiffer syndrome, type I (01600)
Type 1 “classic” Pfeiffer syndrome: mild manifestations, ibrachycephaly, midface hypoplasia, finger and toe abnormalities, normal intelligence, and generally good outcome
None reported
[8386] [87, 88]
Y372C
Osteoglophonic dysplasia (166250)
Craniosynostosis, telechanthus, facial hypoplasia, prominent supraorbital ridge, depressed nasal bridge, and rhizomelic dwarfism
None reported
[83, 89, 90]
FGFR2 (Chr 10q) S252W or P253R (most common)
Apert syndrome (101200)
Craniosynostosis, midface hypoplasia, syndactyly of the hands and feet, tendency to fusion of bony structures, varying mental deficiency, and hearing loss. Increased number and maturation of pre-osteoblasts
Endometrial cancers (S252W and P253R)
[7, 83, 91, 92] [93, 94]
Multiple mutations reported [95]
Crouzon syndrome (123500)
Craniosynostosis, hypertelorism, exophthalmos, external strabismus, parrot-beaked nose, short upper lip, hypoplastic maxilla, and relative mandibular prognathism
Gastric cancer (S267P)
[83, 9597] [98]
S252L, S267P
Pfeiffer syndrome, type 2 and 3 (101600)
Type 2: cloverleaf skull with “Pfeiffer” hands and feet, ankylosis of the elbows. Type 3: similar to Type 2, but without cloverleaf skull. Early demise is characteristic of types 2 and 3
Gastric cancer (S267P)
[84, 99, 100] [98]
FGFR3 (Chr 4p)
G380R, S279C G375C
Achondroplasia (100800)
Most frequent form of dwarfism: short stature, rhizomelic shortening of limbs, frontal bossing, midface hypoplasia, exaggerated lumbar lordosis, limitation of elbow extension, genu varum, and trident hand
Bladder, prostate, and testicular cancers (G380R)
[8, 83, 101103] [104, 105]
R248C, S249C, R373C, Ter807G/R/C, G370C, N540L, Q485R
Thanatophoric dysplasia I (TDI) (187600)
Severe dwarfism; usually fatal in the neonatal period. Curved short femurs with or without cloverleaf skull
Bladder (R248C, S249C, G370C), Prostate (S249C), Lung squamous (R248C, S249C), Head and Neck (S249C), Multiple Myeloma (R248C)
[83, 106108] [109111]
K650E
Thanatophoric dysplasia II (187601)
Severe dwarfism; usually fatal in the neonatal period. Straight, short femurs with cloverleaf skull
Multiple Myeloma, Bladder, Glioblastoma
[83, 112, 113] [56, 114]
N540K/T/S I538V, K650N/Q, L652Q, Y278C S84L
Hypochondroplasia (146000)
Dwarfism, lumbar lordosis, short and broad bones, and caudal narrowing of the interpediculate distance of the lumbar spine. Some resemblance to achondroplasia, but is much milder
Renal cell carcinoma (K650N)
[83, 115118] [56]
P250R
Muenke syndrome (602849)
Coronal synostosis, macrocephaly, midface hypoplasia, developmental delay. Variable phenotype
None reported
[5, 83, 86, 119]
Chr chromosome, FGF fibroblast growth factor, FGFR fibroblast growth factor receptor, OMIM Online Medelian Inheritance in Man (http://​www.​omim.​org/​)

3.2 Somatic aberrations in benign conditions

Somatic or acquired mutations in FGFR3 have been observed in benign skin conditions like seborrheic keratosis and epidermal nevi [54, 55]. These aberrations are activating and, when evaluated, are not seen in adjacent normal skin [55]. Many of these are identical to mutations that are also seen in bladder and/or cervical cancers.

3.3 Somatic aberrations in malignancies

Aberrations in FGFR and its ligands are common in malignancy (Tables 2, 3, and 4 and Figs. 2 and 3). Across malignancies, FGF anomalies are found in ≈14 % and FGFR in ≈7 % of malignancies [56] (FGF/FGFR in about 20 %).
Table 2
Specific examples of FGFR alterations in cancer
Activating
aberrations
Examples of disease(s) (most common)
Reference(s)
Amplifications
 FGFR1
Squamous cell carcinoma of lung, breast adenocarcinoma, bladder urothelial carcinoma, head and neck squamous cell carcinoma
[78, 120123] [124] [125, 126]
 FGFR2
Gastric adenocarcinoma
[127129]
 FGFR3
Uterine carcinosarcoma, ovarian cystadenocarcinoma, sarcoma
[56] [130] [56, 131]
 FGFR4
Kidney, renal clear cell carcinoma
[56]
Mutations
 FGFR1
Stomach adenocarcinoma, melanoma
[127] [56, 132]
 FGFR2
Uterine (endometrial carcinoma), melanomaa
[133] [56, 59, 132]
 FGFR3
High-grade bladder cancer, cervical cancer
[124, 134, 135] [136]
 FGFR4
Rhabdomyosarcoma, melanoma
[137, 138] [132]
Rearrangements
 FGFR1
8p11 myeloproliferative syndrome/fusions partners: BCR, ZNF198, CEP110, FGFR1OP1, FGFR1OP2, HERVK, TRIM24, LRRFIP1, MYO18A, CPSF6
[63]
Rhabdomyosarcoma/fusions partner: FOXO1
[64]
Glioblastoma/fusions partner: TACC1
[68]
Salivary gland/fusions partner: PLAG1
[139]
 FGFR2
Cholangiocarcinoma/fusions partners: AHCYL1,BICC1
[140, 141]
Breast/fusions partners: AFF3, CCDC6, CASP7
[141]
 FGFR3
Multiple myeloma/fusions partner: MMSET
[65]
Glioblastoma, bladder carcinoma, head and neck squamous cell carcinoma/fusions partner: TACC3
[68, 135, 141]
aFGFR2 mutations may be loss of function
Table 3
Examples of FGFR mutations and copy number alterations in cancer
Type of cancer
Approximate frequency
Approximate frequencies by FGF receptora
Comments
References
Bladder urothelial carcinoma
35 %
FGFR1 14 %FGFR3 19 %
FGFR2 3 %FGFR4 6 %
FGFR1 almost all amplifications
FGFR3 mostly amplification and mutations
[56, 124]
Lung, squamous cell carcinoma
27 %
FGFR1 18 %FGFR3 4 %
FGFR2 4 %FGFR4 2 %
FGFR1: Almost all are gene amplifications
[56, 142]
Uterine corpus endometrioid carcinoma
24 %
FGFR1 7 %FGFR3 5 %
FGFR2 14 %FGFR4 4 %
FGFR1 approximately 50 % amplification and 50 % mutations
FGFR2 almost all mutations
[56, 133]
Gastric adenocarcinoma
23 %
FGFR1 6 %FGFR3 4 %
FGFR2 10 %FGFR4 5 %
Approximately 50 % amplifications/deletions and 50 % mutations
[56, 127]
Breast adenocarcinoma
20 %
FGFR1 14 % FGFR3 2 %
FGFR2 3 %FGFR4 2 %
Almost all are amplifications
[56, 120, 143145]
Melanoma
20 %
FGFR1 5 %FGFR3 5 %
FGFR2 11 %FGFR4 5 %
FGFR2 mostly mutations
[56]
Ovarian serous cystadenocarcinoma
20 %
FGFR1 5 %FGFR3 8 %
FGFR2 4 %FGFR4 4 %
Almost all amplifications, rare mutations
[56, 130]
Head and neck squamous cell carcinoma
17 %
FGFR1 10 %FGFR3 4 %
FGFR2 1 %FGFR4 1 %
Majority of amplification with about 20 % deletion and mutations (each), and few fusions
[56, 146, 147]
Lung, adenocarcinoma
14 %
FGFR1 6 %FGFR3 2 %
FGFR2 4 %FGFR4 4 %
Approximately 50 % amplifications and 50 % mutations, with predominance of FGFR1 amplification
[56, 148150]
Prostate adenocarcinoma
11 %
FGFR1 6 %FGFR3 1 %
FGFR2 3 %FGFR4 1 %
Approximately 50 % amplification, 50 % deletions, mutations rare
[56, 151]
Renal cell carcinoma, clear cell
11 %
FGFR1 2 %FGFR3 1 %
FGFR2 < 1 %FGFR4 7 %
Majority amplifications
[56, 152]
Sarcoma
10 %
FGFR1 4 %FGFR3 4 %
FGFR2 1 %FGFR4 2 %
Majority amplifications (n = 2 deletions)
[56, 131]
Renal papillary cell
9 %
FGFR1 4 %FGFR3 2 %
FGFR2 1 %FGFR4 3 %
All mutations, only 2 cases had amplification
[56]
Colorectal adenocarcinoma
8 %
FGFR1 5 %FGFR3 1 %
FGFR2 1 %FGFR4 1 %
FGFR1 about 60 % amplification, rest mutations/deletion
[56, 153]
Glioblastoma
6 %
FGFR1 0 %FGFR3 2 %
FGFR2 3 %FGFR4 1 %
FGFR2 mostly deletions
[56, 154]
Adenoid cystic carcinoma
5 %
FGFR1 3 %FGFR3 0 %
FGFR2 0 %FGFR4 2 %
FGFR1 amplification and deletion (1 each)
FGFR4 mutation (n = 1)
[56, 155]
Brain, lower grade gliomas
5 %
FGFR1 0 %FGFR3 1 %
FGFR2 3 %FGFR4 1 %
Most are deletions, with few amplifications and mutations
[56, 156]
Acute myeloid leukemia
1 %
FGFR1 < 1 %FGFR3 0 %
FGFR2 0 %FGFR4 < 1 %
1 amplification, 1 deletion, no mutations
[56, 157]
Thyroid carcinoma
<1 %
FGFR1 0 %FGFR3 < 1 %
FGFR2 < 1 %FGFR4 < 1 %
Two amplifications, one mutation
[56]
See also Fig. 2 for illustration (bar graph)
FGF fibroblast growth factor, FGFR fibroblast growth factor receptor
aData extracted/analyzed based on cbioportal at http://​www.​cbioportal.​org/​public-portal (accessed November 2014). Most of the studies included >200 patients
Table 4
Examples of FGF ligand mutations and copy number aberrations in cancer
Type of cancer
Approximate frequency
Approximate frequencies by FGF liganda
Comments
Reference(s)
Head and neck squamous cell carcinoma
54 %
FGF3 28 %FGF12 19 %
FGF4 28 %FGF10 6 %
FGF19 28 %FGF23 5 %
Virtually all amplifications
[56]
Bladder urothelial carcinoma
47 %
FGF3 13 %FGF1711%
FGF4 12 %FGF10 9 %
FGF19 13 %FGF20 9 %
FGF3, FGF4, and FGF19 co-amplified in approximately 12 % of cases
FGF17 and 20 mostly deletions
[56, 124]
Stomach cancer
47 %
FGF3 7 %FGF12 8 %
FGF4 7 %FGF13 6 %
FGF19 7 %FGF14 5 %
FGF10 9 %FGF17 5 %
FGF3/4/19 co-amplified in 7 % of cases
FGF17 and FGF20 both deleted in 2 % of cases
[56, 127]
Lung, squamous cell carcinoma
46 %
FGF3 12 %FGF12 26 %
FGF4 12 %FGF10 7 %
FGF19 13 %
Virtually all are gene amplifications
[56, 142]
Cervical cancerb
42 %
FGF12 25 %
All are amplifications
[56]
Lung, adenocarcinoma
39 %
FGF10 11 %
FGF17 7 %
FGF20 7 %
FGF10 mostly amplifications
FGF17 and FGF20 mostly deletions
FGF3/4/19 co-amplified in 4 % of cases
[56, 148150]
Melanoma
38 %
FGF3 8 %
FGF4 6 %
FGF19 6 %
FGF3/4/19 co-amplified in about 7 % of cases
[56]
Ovarian cystadenocarcinoma
38 %
FGF3 5 %FGF6 5 %
FGF4 4 %FGF23 6 %
FGF19 4 %FGF12 13 %
Virtually all amplifications
[56, 130]
Breast adenocarcinoma
35 %
FGF3 15 %FGF17 6 %
FGF4 15 %FGF20 5 %
FGF19 15 %
High frequency of co-amplification of FGF3/4/19. Similar results with TCGA, Nature 2012 study (n = 482)
[56, 120, 143, 145]
Adenoid cystic carcinoma
27 %
FGF22 10 %
All others 5 %or less
Approximately 50 % deletions and 50 % amplifications, rare mutations
[56, 155]
Prostate adenocarcinoma
22 %
FGF17 8 %
FGF20 5 %
Majority are deletions, about 5 % cases are co-deleted FGF17/20
[56, 151]
Colorectal adenocarcinoma
17 %
All 5 % or less
Majority of mutations, less amplifications and rare deletion
[56, 153, 158]
See also Fig. 3 for illustration (bar graph)
FGF fibroblast growth factor
aIncluded FGFs with alteration frequency ≥5 % and at least 5 cases with the alteration. Extracted/analyzed in part based on cbioportal at http://​www.​cbioportal.​org/​public-portal (accessed November 2014)
bSquamous cell carcinoma and endocervical adenocarcinoma. Abbreviations: FGF = fibroblast growth factor. See also Fig. 3 for illustration (bar graph)

3.3.1 FGFR alterations

While almost any FGFR genes can be altered in many cancer types, some acquired genetic aberrations are more striking in prevalence or in cancer type(s) than others. The most common abnormalities reported to date are gene amplifications of FGFR1–3 (Table 2). These are generally assumed to represent activation or gain-of-function amplifications, but full molecular characterization of the effects of gene amplification within specific cellular or cancer microenvironment contexts is not fully available. The cancers in which FGFR gene amplifications are most frequent include bladder urothelial carcinomas (FGFR1), squamous cell lung cancer (FGFR1), head and neck squamous cell cancer (FGFR1), uterine carcinosarcoma (FGFR3), breast adenocarcinoma (FGFR1), and gastric adenocarcinoma (FGFR2) (Tables 2 and 3, and Fig. 2). In fact, FGFR2 was first identified in a gastric cancer cell line, and it is preferentially amplified/overexpressed in the diffuse type of gastric cancer, which correlates with poor prognosis, at least in a Japanese population [57]. More commonly, however, mutations in FGFR3 characterize bladder carcinoma. Indeed, activating mutations of fibroblast growth factor receptor-3 (FGFR3) have been described in approximately 75 % of low-grade papillary bladder tumors. In muscle-invasive disease, FGFR3 mutations are found in 20 % of tumors, but overexpression of FGFR3 is observed in about half of cases [58].
Loss-of-function alterations are relatively uncommon in cancer (a pattern that is also seen in hereditary disorders). However, there is at least one report indicating that some melanoma cell lines demonstrate FGFR2 loss of function [59].
FGFR1 amplification is prevalent in both squamous cell carcinoma of the lung and of the head and neck, suggesting a possible common underlying mechanism of carcinogenesis in these smoking-related carcinomas. Perhaps more importantly, development of FGFR1 inhibitors represents a viable targeted therapy for use in squamous cell lung cancers [60, 61].

3.3.2 FGF alterations

FGF abnormalities are for the most part amplifications (Fig. 3 and Table 4). There are few reported data regarding the mechanism(s) of action of these amplifications. Nevertheless, it seems reasonable to hypothesize that abundance of ligand could lead to increased receptor signaling, but the reality may be more complex, perhaps relating to the stoichiometric proportions of the different ligands present. Of note, three of the ligands, FGF3, FGF4, and FGF19, are frequently co-amplified on 11q13 [62]. This amplicon is present in several cancers, including breast, bladder, and squamous cell carcinoma of the lung and head and neck (Fig. 3 and Table 4).

3.3.3 FGFR rearrangements

Chromosomal translocation gives rise to chimeric gene products with aberrant function (Table 2). In general, fusion gene proteins result from the fusion of a “partner gene” with a tyrosine kinase domain derived from the FGFR family member gene. As a result of their constant dimerization state, they are constitutively active in the absence of ligand. The FGFR1 gene can be fused to other genes including BCR, ZNF198, CEP110, FGFR1OP1, FGFR1OP2, HERVK, TRIM24, LRRFIP1, and MYO18A, in the 8p11 myeloproliferative syndrome manifested by myeloproliferative neoplasms and peripheral blood eosinophilia without basophilia [63]; it is fused to the FOXO1 gene in alveolar rhabdomyosarcoma, and the FOXO1-FGFR1 fusion gene is amplified [64]. The FGFR3 gene is fused to the MMSET gene as a result of a t(4;14)(p16.3;q32) chromosomal translocation in 10–20 % of multiple myeloma [65]; it is fused to the ETV6 gene in peripheral T cell lymphoma with a t(4;12)(p16;p13) chromosomal translocation [66]. In glioblastoma multiforme, FGFR1 and FGFR3 genes are fused to neighboring TACC1 and TACC3 genes due to interstitial deletions, respectively [67, 68].

4 FGF/FGFR signaling inhibitors and cancer therapy

4.1 FDA approved drugs that target FGFR

Only four drugs (ponatinib, regorafenib, pazopanib, and more recently lenvatinib) are FDA-approved for use in human cancers (Table 5). Ponatinib is a multi-tyrosine kinase inhibitor that was approved for imatinib-resistant chronic myelogenous leukemia (CML) and Philadelphia chromosome-positive (CP) acute lymphoblastic leukemia (ALL). The efficacy results demonstrated a 54 % major cytogenetic response (MCyR) rate in patients with CP-CML, and seventy percent of patients with CP-CML with the T315I mutation in BCR-ABL achieved MCyR. In addition of targeting BCR-ABL, ponatinib can also inhibit members of the VEGFR, PDGFR, FGFR (IC50 = 2 nM for FGFR1), and SRC families of kinases, KIT, or RET, with IC50 between 0.1 and 20 nM. Of note, ponatinib was briefly taken off the market by the FDA because of concerns about cardiovascular side effects (clotting), but soon after, it was returned to the market with updated safety monitoring recommendations. Regorafenib was approved for imatinib-resistant gastrointestinal stromal tumor (GIST) and metastatic colorectal cancer, based on a statistically significant survival prolongation observed in patients randomized to receive regorafenib (6.4 vs 5.0 months in the placebo arm, P = 0.01). Regorafenib, and its active metabolites inhibit multiple membrane-bound and intracellular kinases including those in the RET, VEGFR1, VEGFR2, VEGFR3, KIT, PDGFR-α and -β, FGFR1-2, and Abl pathways. Pazopanib was approved for advanced renal cell carcinoma (based on a progression-free survival of 9.2 months compared to 4.2 months in the placebo arm), as well as soft tissue sarcomas (improved progression-free survival: 4.6 months versus 1.6 for patients who received placebo). Pazopanib is a multi-tyrosine kinase inhibitor whose targets include VEGFR1-3, PDGFR-α and -β, FGFR-1 and -3, and KIT. Lastly, lenvatinib is a multi-kinase inhibitor (targets including VEGFR1–3, FGFR1–4, PDGFR-α, KIT, and RET) indicated for the treatment of patients with locally recurrent or metastatic, progressive, radioactive iodine-refractory differentiated thyroid cancer. The recent approval in February 2015 was based on an improved progression-free survival (18.3 vs 3.6 months in the placebo group, P < 0.001). The approvals of these four agents (ponatinib, regorafenib, pazopanib, and lenvatinib), all three of which are multi-kinase inhibitors, were not based on activity against FGFR. Of interest, nintedanib is an FGFR inhibitor that is FDA-approved for a non-cancer indication—idiopathic pulmonary fibrosis (Table 5).
Table 5
Examples of drugs that inhibit FGF/FGFR pathway signaling
Drug
Company/type of drug
Examples of target(s)
FDA-approved
Yes/No
Examples of clinical development/trials/phase
References
ARQ087
ArQule/
selective FGFRs inhibitor
FGFR1/2/3
No
Phase I Dose Escalation Study
[159]
AZD4547
Astrazeneca/
selective FGFRs inhibitor
FGFR1/2/3
No
Phase II
Part of Lung-MAP
[128, 160, 161]
Brivanib
(BMS-540215)
BMS/
dual kinase inhibitor
FGFR1/2/3, VEGFR
No
Phase III hepatocellular; did not meet endpoint of survival non-inferiority
[121, 162164]
Danusertib
(PHA-739358)
Nerviano Medical Sciences/multi-kinase inhibitor
FGFR1, BCR-Abl, c-
RET, Aurora
No
Phase II in unselected prostate cancer showed minimal activity
[165, 166]
Debio1347
Debiopharm/
selective FGFRs inhibitors
FGFR1/2/3
No
Phase I (selecting patient with FGFRs alterations)
[70, 159]
Dovitinib
(TKI 258)
Novartis/
multi-kinase inhibitor
FGFR1/3, PDGFR, VEGFR, Flt3, c-kit
No
Phase III: renal cell carcinoma
(failed to meet the primary endpoint, unselected patients)
[167169]
FP-1039
(GSK3052230)
GlaxoSmithKline/
FGFsFGFs trap agent
Sequesters
No
Phase I (selecting patients with deregulated Fibroblast Growth Factor (FGF) Pathway Signaling)
[71]
JNJ-42756493
Janssen/
selective FGFRs inhibitors
FGFR1/2/3/4
No
Phase I
[159, 170]
Lenvatinib
(E7080)
Esai/
multi-kinase inhibitor
PDGFR
Yes
Phase III:FDA- approved for thyroid cancer (Feb 2015)
[171, 172]
Lucitanib
(E3810)
Clovis/
dual kinase inhibitor
FGFR1/2 and
VEGFR1/2/3
No
Phase II: ER-positive breast cancer
[69, 173175]
MGFR1877S (RG744)
Genentech/
monoclonal antibody
Anti-FGFR3
No
Phase I
[176]
MK2461
Merck/
multi-kinase inhibitor
FGFR1/2/3, PDGFR, c-Met, Flt1/3, Ron, Mer
No
Phase II
[177]
Nintedanib
(BIBF1120)
Boehringer Ingelheim/
multi-kinase inhibitor
FGFR1/2/3, VEGFR, PDGFR, flt3
Yes (not in cancer)
Phase III: ovarian, lung, FDA-approved for idiopathic
pulmonary fibrosis (Oct 2014)
[178180]
NVP-BGJ398
Novartis/
selective FGFRs inhibitor
FGFR1/2/3
No
Phase II
[181183]
Pazopanib
GlaxoSmithKline/
multi-kinase inhibitor
FGFR1/3, VEGFR1/2/3, PDGFR, c-Kit
Yes
Approved for advanced renal cell carcinoma and soft tissue sarcoma
[184, 185]
Ponatinib
Ariad/
multi-kinase inhibitor
FGFR1-4, BCR-Abl, PDGFR, VEGFR
Yes
Approved for T315I-positive chronic myelogenous leukemia and Ph-positive acute lymphoblastic leukemia
[186, 187]
Regorafenib
Bayer/
multi-kinase inhibitor
FGFR1/2, RET, VEGFR1/2/3 KIT, PDGFRs
Yes
Approved for advanced GIST and colorectal cancer (no FGFR selection)
[188, 189]
TAS120
Taiho Oncology/
selective FGFRs inhibitors
FGFR1/2/3/4
No
Phase I/II (selecting patient with FGFRs alterations)
[159, 190]
The IC50 (half-maximal inhibitory concentration) was <100 nmol/L for all the drugs included, except regorafenib
FGFR fibroblast growth factor receptor, GIST gastrointestinal stromal tumor

4.2 Experimental agents that target FGF/FGFR

There are several pharmacologic agents that have been or are being developed for inhibition of FGFR via targeting of the ATP binding site of the intracellular tyrosine kinase domain(s) (Table 5). The inhibition varies by their affinities for FGFR signaling, as most of these molecules are promiscuous kinase inhibitors (inhibiting VEGF, PDGF, and many other TKIs in addition to FGFR). Figure 1b gives examples of selective versus non-selective FGFR inhibitors. Of note, the dual kinase inhibitor (VEGFR/FGFR) lucitanib has shown activity in FGFR1-amplified breast cancer, with an overall response rate of 50 % [69]. There are also selective tyrosine kinase inhibitors (TKIs) available (Table 5 and Fig. 1b). While several of these agents are currently in clinical trials, none of the more highly selective FGFR inhibitors (e.g., BGJ398 or AZD4547) are currently FDA-approved. A recent study demonstrated that Debio 1347 (a selective orally available FGFR1–3 inhibitor) displayed preferential anti-tumor activity against cells with FGFR genetic alterations in a panel of 327 cancer cell lines and xenograft models [70]. Debio 1347 is currently under investigation for the treatment of patients harboring FGFR genetic alterations.
Of interest, other types of agents have been developed. As an example, FP-1039 is a soluble fusion protein, consisting of the extracellular domains of human FGFR1 linked to the Fc region of the human immunoglobulin G1; it is designed to bind multiple FGF ligands [71] (TRAP molecule).
A large number of other drugs and indications are being pursued. Some FGFR inhibitors have failed to meet their phase III endpoints (Table 5). The majority of the failed trials, however, have been performed in patient populations that were not biomarker-selected.

4.3 Resistance mechanisms

In a recent phase 1 study reporting on patients with FGFR1-amplified (identified by fluorescent/chromogenic in situ hybridization) advanced or metastatic lung squamous cell carcinoma (SCC) treated with the selective pan-FGFR inhibitor BGJ398, only a limited number of patients achieved relatively short-lived partial responses (2 of 17 patients, 11.7 %); responses lasted 3 and 8 months. These observations suggest the existence of resistance mechanisms. Sohl et al. [72] reported that resistance mutations at the “gatekeeper” residue may arise (FGFR1 V561M mutation confers a 38-fold increase in autophosphorylation and significant resistance to lucitanib), leading to tumor progression and explaining the non-durable responses. For instance, it has also been shown that the heterozygous gatekeeper mutation FGFR3 V555M appeared as a mechanism of acquired resistance to selective FGFR inhibitors [73]. Several other activating mutations were identified in FGFR2-expressing cells treated with high concentrations of dovitinib, and the multi-kinase inhibitor ponatinib inhibitory activity was affected by the V565I gatekeeper mutation [74]. In addition, a previously undescribed FGFR3 variant was identified as a key contributor to resistance in the MGH156-1A cell line derived from a patient with acquired resistance to EGFR TKIs [75], and follow-up studies clearly indicated that FGFR inhibitors re-sensitized these cells to EGFR inhibitors.
Besides secondary mutations in the kinase domain, resistance to FGFR kinase inhibitors may also occur through activation of alternative signaling pathways, as demonstrated by Harbinski et al. [76] who showed a broad and versatile potential for tyrosine kinase receptor from the FGFR, HER, and MET family to compensate for each other. Javidi-Sharifi et al. [77] suggest that some patients with gastrointestinal stromal tumor (GIST) treated with imatinib can develop a functional dependence on FGFR3, illustrated by the fact that the addition of the FGFR3 ligand FGF2 to GIST cells restored KIT phosphorylation during imatinib treatment. Furthermore, signaling crosstalk between KIT and FGFR3 activated the MAPK pathway to promote resistance to imatinib. FGFR amplification and overexpression have also been related to poor prognosis and endocrine resistance in breast cancer [78]. Of note, relationships between cyclins and FGF/FGFRs have also been reported at the protein level. For instance, a study showed that FGFR4 contributed to the maintenance of CCND1 via the mTOR translation pathway, and several other studies demonstrated cooperation between FGFR and CCND1 [79]. Finally, most patients with advanced cancer have complex molecular portfolios, and hence there may be multiple genomic drivers that are active and supplant the role of FGFR [8082]. In that context, it appears evident that identification of resistance mechanisms is crucial to crafting effective drug combinations.

5 Conclusion

The FGF/FGFR pathway is crucial to a variety of human diseases. There are five known FGFRs, FGFR1–FGFR4 and FGFRL1, and 18 human ligands for FGFRs. FGFR germline mutations (activating) can cause skeletal disorders, primarily dwarfism (generally mutations in FGFR3) and craniofacial malformation syndromes (usually mutations in FGFR1 and FGFR2). Loss-of-function mutations in FGF signaling are seen in congenital hypogonadotropic hypogonadism (including the Kallman syndrome variant with anosmia). Interestingly, many of the aberrations that cause the inherited skeletal disorders are also seen in human cancers.
The most common abnormalities in malignancies are gene amplifications of FGFR1–3 or of the FGF ligands. The cancers in which FGFR gene amplifications are most frequent include squamous cell lung cancer (FGFR1), head and neck squamous cell cancer (FGFR1), bladder (transitional cell) cancer (FGFR1), endometrial cancer (FGFR1), gastric adenocarcinoma (FGFR2), breast adenocarcinoma (FGFR1), and prostate adenocarcinoma (FGFR1). Point mutations are seen in all FGFRs but are less frequent in FGFs. For instance, mutations in FGFR3 are frequent in bladder carcinoma, and FGFR2 mutations in endometrial cancer, melanoma, and gastric tumors (Tables 2 and 3).
Interestingly, somatic mutations in FGFR3 have been observed in benign skin conditions such as seborrheic keratosis and epidermal nevi (but not in adjacent normal skin) [54]. FGFR rearrangements are also observed in certain cancers and characterize certain myeloproliferative disorders (Table 2).
Importantly, there are several pharmacologic agents that have been or are being developed for inhibition of FGFR kinases. These include both highly selective inhibitors as well as multi-kinase inhibitors. Ponatinib, regorafenib, pazopanib, and lenvatinib are already FDA-approved for cancer, albeit not on the basis of their FGFR activity. Very few studies in cancer have been aimed at an FGFR biomarker-selected population, and several of the FGFR inhibitors have failed phase III studies in unselected patients. A multi-kinase inhibitor (nintedanib), which suppresses FGFR1–3, was also recently FDA-approved for idiopathic pulmonary fibrosis.
Whether or not FGFR inhibitors could also be used to moderate the phenotype of inherited disorders due to FGFR activation is an intriguing question. Of interest, Garcia et al. [48] injected ia mouse model of achondroplasia with a soluble form of human FGFR3 (acting as a decoy receptor and preventing FGF from binding to mutant FGFR3), and effective maturation of growth plate chondrocytes was restored in the bones of treated mice. Of interest in this regard, individuals afflicted with the inherited disorders associated with FGFR aberrations, such as dwarfism, do not have an increased incidence of cancer, despite having mutations that are often identical to those somatic FGFR aberrations that characterize certain tumors. The secondary modulatory influences that mitigate the risk of cancer in these individuals are of interest. Whether or not treating them at an early age with FGFR inhibitors would increase the later risk of cancer if the inhibitors were withdrawn would need to be considered.
In summary, perturbations in the FGF/FGFR machinery appear to underlie a variety of inherited syndromes, as well as benign and malignant disorders. The advent of potent FGFR inhibitors provides important new agents in the armamentarium against diseases caused by FGF/FGFR abnormalities.

Acknowledgments

Conflicts of interest

Dr. Kurzrock is a founder of RScueRx and receives consulting fees from Sequenom. Dr. Helsten receives research funding from Pfizer, Genentech, Abbvie, Novartis, Lilly, Bayer, Merrimack, Clovis, and Esai. Dr. Schwaederle has no conflict of interest to report.

Author’s contributions

Conception and design: All authors
Collection and assembly of data: All authors
Manuscript writing: All authors
Final approval of manuscript: All authors

Research Support

This study was funded in part by the Joan and Irwin Jacobs Fund.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
3.
Zurück zum Zitat Greenman, C., Stephens, P., Smith, R., Dalgliesh, G. L., Hunter, C., Bignell, G., & Stratton, M. R. (2007). Patterns of somatic mutation in human cancer genomes. Nature, 446(7132), 153–158.PubMedCentralPubMedCrossRef Greenman, C., Stephens, P., Smith, R., Dalgliesh, G. L., Hunter, C., Bignell, G., & Stratton, M. R. (2007). Patterns of somatic mutation in human cancer genomes. Nature, 446(7132), 153–158.PubMedCentralPubMedCrossRef
4.
Zurück zum Zitat Torkamani, A., & Schork, N. J. (2008). Prediction of cancer driver mutations in protein kinases. Cancer Research, 68(6), 1675–1682.PubMedCrossRef Torkamani, A., & Schork, N. J. (2008). Prediction of cancer driver mutations in protein kinases. Cancer Research, 68(6), 1675–1682.PubMedCrossRef
5.
Zurück zum Zitat Muenke, M., Gripp, K. W., McDonald-McGinn, D. M., Gaudenz, K., Whitaker, L. A., Bartlett, S. P., & Wilkie, A. O. (1997). A unique point mutation in the fibroblast growth factor receptor 3 gene (FGFR3) defines a new craniosynostosis syndrome. American journal of human genetics, 60(3), 555–564.PubMedCentralPubMed Muenke, M., Gripp, K. W., McDonald-McGinn, D. M., Gaudenz, K., Whitaker, L. A., Bartlett, S. P., & Wilkie, A. O. (1997). A unique point mutation in the fibroblast growth factor receptor 3 gene (FGFR3) defines a new craniosynostosis syndrome. American journal of human genetics, 60(3), 555–564.PubMedCentralPubMed
6.
Zurück zum Zitat Robin, N. H., Falk, M. J., & Haldeman-Englert, C. R. (1993). FGFR-Related Craniosynostosis Syndromes. In R. A. Pagon, M. P. Adam, H. H. Ardinger, T. D. Bird, C. R. Dolan, C.-T. Fong, … K. Stephens (Eds.), GeneReviews(®). Seattle (WA): University of Washington, Seattle. Retrieved from http://www.ncbi.nlm.nih.gov/books/NBK1455/. Robin, N. H., Falk, M. J., & Haldeman-Englert, C. R. (1993). FGFR-Related Craniosynostosis Syndromes. In R. A. Pagon, M. P. Adam, H. H. Ardinger, T. D. Bird, C. R. Dolan, C.-T. Fong, … K. Stephens (Eds.), GeneReviews(®). Seattle (WA): University of Washington, Seattle. Retrieved from http://​www.​ncbi.​nlm.​nih.​gov/​books/​NBK1455/​.
7.
Zurück zum Zitat Anderson, J., Burns, H. D., Enriquez-Harris, P., Wilkie, A. O., & Heath, J. K. (1998). Apert syndrome mutations in fibroblast growth factor receptor 2 exhibit increased affinity for FGF ligand. Human molecular genetics, 7(9), 1475–1483.PubMedCrossRef Anderson, J., Burns, H. D., Enriquez-Harris, P., Wilkie, A. O., & Heath, J. K. (1998). Apert syndrome mutations in fibroblast growth factor receptor 2 exhibit increased affinity for FGF ligand. Human molecular genetics, 7(9), 1475–1483.PubMedCrossRef
8.
Zurück zum Zitat He, L., Horton, W., & Hristova, K. (2010). Physical basis behind achondroplasia, the most common form of human dwarfism. The Journal of biological chemistry, 285(39), 30103–30114.PubMedCentralPubMedCrossRef He, L., Horton, W., & Hristova, K. (2010). Physical basis behind achondroplasia, the most common form of human dwarfism. The Journal of biological chemistry, 285(39), 30103–30114.PubMedCentralPubMedCrossRef
9.
Zurück zum Zitat Rousseau, F., el Ghouzzi, V., Delezoide, A. L., Legeai-Mallet, L., Le Merrer, M., Munnich, A., & Bonaventure, J. (1996). Missense FGFR3 mutations create cysteine residues in thanatophoric dwarfism type I (TD1). Human molecular genetics, 5(4), 509–512.PubMedCrossRef Rousseau, F., el Ghouzzi, V., Delezoide, A. L., Legeai-Mallet, L., Le Merrer, M., Munnich, A., & Bonaventure, J. (1996). Missense FGFR3 mutations create cysteine residues in thanatophoric dwarfism type I (TD1). Human molecular genetics, 5(4), 509–512.PubMedCrossRef
10.
Zurück zum Zitat Dodé, C., Teixeira, L., Levilliers, J., Fouveaut, C., Bouchard, P., Kottler, M.-L., & Hardelin, J.-P. (2006). Kallmann syndrome: mutations in the genes encoding prokineticin-2 and prokineticin receptor-2. PLoS genetics, 2(10), e175.PubMedCentralPubMedCrossRef Dodé, C., Teixeira, L., Levilliers, J., Fouveaut, C., Bouchard, P., Kottler, M.-L., & Hardelin, J.-P. (2006). Kallmann syndrome: mutations in the genes encoding prokineticin-2 and prokineticin receptor-2. PLoS genetics, 2(10), e175.PubMedCentralPubMedCrossRef
12.
Zurück zum Zitat Dodé, C., Levilliers, J., Dupont, J.-M., De Paepe, A., Le Dû, N., Soussi-Yanicostas, N., & Hardelin, J.-P. (2003). Loss-of-function mutations in FGFR1 cause autosomal dominant Kallmann syndrome. Nature Genetics, 33(4), 463–465. doi:10.1038/ng1122.PubMedCrossRef Dodé, C., Levilliers, J., Dupont, J.-M., De Paepe, A., Le Dû, N., Soussi-Yanicostas, N., & Hardelin, J.-P. (2003). Loss-of-function mutations in FGFR1 cause autosomal dominant Kallmann syndrome. Nature Genetics, 33(4), 463–465. doi:10.​1038/​ng1122.PubMedCrossRef
14.
Zurück zum Zitat Trarbach, E. B., Abreu, A. P., Silveira, L. F. G., Garmes, H. M., Baptista, M. T. M., Teles, M. G., & Latronico, A. C. (2010). Nonsense mutations in FGF8 gene causing different degrees of human gonadotropin-releasing deficiency. The Journal of Clinical Endocrinology and Metabolism, 95(7), 3491–3496. doi:10.1210/jc.2010-0176.PubMedCentralPubMedCrossRef Trarbach, E. B., Abreu, A. P., Silveira, L. F. G., Garmes, H. M., Baptista, M. T. M., Teles, M. G., & Latronico, A. C. (2010). Nonsense mutations in FGF8 gene causing different degrees of human gonadotropin-releasing deficiency. The Journal of Clinical Endocrinology and Metabolism, 95(7), 3491–3496. doi:10.​1210/​jc.​2010-0176.PubMedCentralPubMedCrossRef
15.
Zurück zum Zitat Turner, N., & Grose, R. (2010). Fibroblast growth factor signalling: from development to cancer. Nature Reviews Cancer, 10(2), 116–129.PubMedCrossRef Turner, N., & Grose, R. (2010). Fibroblast growth factor signalling: from development to cancer. Nature Reviews Cancer, 10(2), 116–129.PubMedCrossRef
16.
Zurück zum Zitat Dienstmann, R., Rodon, J., Prat, A., Perez-Garcia, J., Adamo, B., Felip, E., & Tabernero, J. (2014). Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO, 25(3), 552–563. doi:10.1093/annonc/mdt419.CrossRef Dienstmann, R., Rodon, J., Prat, A., Perez-Garcia, J., Adamo, B., Felip, E., & Tabernero, J. (2014). Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO, 25(3), 552–563. doi:10.​1093/​annonc/​mdt419.CrossRef
17.
Zurück zum Zitat Baselga, J. (2006). Targeting tyrosine kinases in cancer: the second wave. Science, 312(5777), 1175–1178.PubMedCrossRef Baselga, J. (2006). Targeting tyrosine kinases in cancer: the second wave. Science, 312(5777), 1175–1178.PubMedCrossRef
19.
Zurück zum Zitat Verstovsek, S., Mesa, R. A., Gotlib, J., Levy, R. S., Gupta, V., DiPersio, J. F., & Kantarjian, H. M. (2012). A Double-Blind, Placebo-Controlled Trial of Ruxolitinib for Myelofibrosis. New England Journal of Medicine, 366(9), 799–807. doi:10.1056/NEJMoa1110557.PubMedCrossRef Verstovsek, S., Mesa, R. A., Gotlib, J., Levy, R. S., Gupta, V., DiPersio, J. F., & Kantarjian, H. M. (2012). A Double-Blind, Placebo-Controlled Trial of Ruxolitinib for Myelofibrosis. New England Journal of Medicine, 366(9), 799–807. doi:10.​1056/​NEJMoa1110557.PubMedCrossRef
20.
Zurück zum Zitat Lee, E. B., Fleischmann, R., Hall, S., Wilkinson, B., Bradley, J. D., Gruben, D., & van Vollenhoven, R. F. (2014). Tofacitinib versus Methotrexate in Rheumatoid Arthritis. New England Journal of Medicine, 370(25), 2377–2386. doi:10.1056/NEJMoa1310476.PubMedCrossRef Lee, E. B., Fleischmann, R., Hall, S., Wilkinson, B., Bradley, J. D., Gruben, D., & van Vollenhoven, R. F. (2014). Tofacitinib versus Methotrexate in Rheumatoid Arthritis. New England Journal of Medicine, 370(25), 2377–2386. doi:10.​1056/​NEJMoa1310476.PubMedCrossRef
21.
Zurück zum Zitat Hubbard, S. R., & Till, J. H. (2000). Protein tyrosine kinase structure and function. Annual review of biochemistry, 69, 373–398.PubMedCrossRef Hubbard, S. R., & Till, J. H. (2000). Protein tyrosine kinase structure and function. Annual review of biochemistry, 69, 373–398.PubMedCrossRef
22.
Zurück zum Zitat Kim, I., Moon, S.-O., Yu, K.-H., Kim, U.-H., & Koh, G. Y. (2001). A novel fibroblast growth factor receptor-5 preferentially expressed in the pancreas. Biochimica et Biophysica Acta (BBA) - Gene Structure and Expression, 1518(1–2), 152–156.CrossRef Kim, I., Moon, S.-O., Yu, K.-H., Kim, U.-H., & Koh, G. Y. (2001). A novel fibroblast growth factor receptor-5 preferentially expressed in the pancreas. Biochimica et Biophysica Acta (BBA) - Gene Structure and Expression, 1518(1–2), 152–156.CrossRef
23.
Zurück zum Zitat Sleeman, M., Fraser, J., McDonald, M., Yuan, S., White, D., Grandison, P., & Murison, J. G. (2001). Identification of a new fibroblast growth factor receptor, FGFR5. Gene, 271(2), 171–182.PubMedCrossRef Sleeman, M., Fraser, J., McDonald, M., Yuan, S., White, D., Grandison, P., & Murison, J. G. (2001). Identification of a new fibroblast growth factor receptor, FGFR5. Gene, 271(2), 171–182.PubMedCrossRef
24.
Zurück zum Zitat Klint, P., & Claesson-Welsh, L. (1999). Signal transduction by fibroblast growth factor receptors. Frontiers in bioscience : a journal and virtual library, 4, D165–77.CrossRef Klint, P., & Claesson-Welsh, L. (1999). Signal transduction by fibroblast growth factor receptors. Frontiers in bioscience : a journal and virtual library, 4, D165–77.CrossRef
25.
Zurück zum Zitat Jin, M., Du, X., & Chen, L. (2012). Cross-talk between FGF and other cytokine signalling pathways during endochondral bone development. Cell biology international, 36(8), 691–696.PubMedCrossRef Jin, M., Du, X., & Chen, L. (2012). Cross-talk between FGF and other cytokine signalling pathways during endochondral bone development. Cell biology international, 36(8), 691–696.PubMedCrossRef
26.
Zurück zum Zitat Horton, W. A. (2006). MOLECULAR PATHOGENESIS OF ACHONDROPLASIA. Growth Genet Horm. Horton, W. A. (2006). MOLECULAR PATHOGENESIS OF ACHONDROPLASIA. Growth Genet Horm.
27.
Zurück zum Zitat Katoh, M. (2008). Cancer genomics and genetics of FGFR2 (Review). International journal of oncology, 33(2), 233–237.PubMed Katoh, M. (2008). Cancer genomics and genetics of FGFR2 (Review). International journal of oncology, 33(2), 233–237.PubMed
28.
Zurück zum Zitat Olsen, S. K., Garbi, M., Zampieri, N., Eliseenkova, A. V., Ornitz, D. M., Goldfarb, M., & Mohammadi, M. (2003). Fibroblast Growth Factor (FGF) Homologous Factors Share Structural but Not Functional Homology with FGFs. Journal of Biological \ldots. Olsen, S. K., Garbi, M., Zampieri, N., Eliseenkova, A. V., Ornitz, D. M., Goldfarb, M., & Mohammadi, M. (2003). Fibroblast Growth Factor (FGF) Homologous Factors Share Structural but Not Functional Homology with FGFs. Journal of Biological \ldots.
29.
Zurück zum Zitat Itoh, N., & Ornitz, D. M. (2008). Functional evolutionary history of the mouse Fgf gene family. Developmental dynamics : an official publication of the American Association of Anatomists, 237(1), 18–27.CrossRef Itoh, N., & Ornitz, D. M. (2008). Functional evolutionary history of the mouse Fgf gene family. Developmental dynamics : an official publication of the American Association of Anatomists, 237(1), 18–27.CrossRef
30.
Zurück zum Zitat Potthoff, M. J., Kliewer, S. A., & Mangelsdorf, D. J. (2012). Endocrine fibroblast growth factors 15/19 and 21: from feast to famine. Genes & development, 26(4), 312–324.CrossRef Potthoff, M. J., Kliewer, S. A., & Mangelsdorf, D. J. (2012). Endocrine fibroblast growth factors 15/19 and 21: from feast to famine. Genes & development, 26(4), 312–324.CrossRef
31.
Zurück zum Zitat Fukumoto, S. (2008). Actions and mode of actions of FGF19 subfamily members. Endocrine journal, 55(1), 23–31.PubMedCrossRef Fukumoto, S. (2008). Actions and mode of actions of FGF19 subfamily members. Endocrine journal, 55(1), 23–31.PubMedCrossRef
33.
Zurück zum Zitat Abuharbeid, S., Czubayko, F., & Aigner, A. (2006). The fibroblast growth factor-binding protein FGF-BP. The international journal of biochemistry & cell biology, 38(9), 1463–1468.CrossRef Abuharbeid, S., Czubayko, F., & Aigner, A. (2006). The fibroblast growth factor-binding protein FGF-BP. The international journal of biochemistry & cell biology, 38(9), 1463–1468.CrossRef
34.
Zurück zum Zitat Zhang, X., Ibrahimi, O. A., Olsen, S. K., Umemori, H., Mohammadi, M., & Ornitz, D. M. (2006). Receptor specificity of the fibroblast growth factor family. The complete mammalian FGF family. The Journal of biological chemistry, 281(23), 15694–15700.PubMedCentralPubMedCrossRef Zhang, X., Ibrahimi, O. A., Olsen, S. K., Umemori, H., Mohammadi, M., & Ornitz, D. M. (2006). Receptor specificity of the fibroblast growth factor family. The complete mammalian FGF family. The Journal of biological chemistry, 281(23), 15694–15700.PubMedCentralPubMedCrossRef
35.
Zurück zum Zitat Ornitz, D. M., Xu, J., Colvin, J. S., McEwen, D. G., MacArthur, C. A., Coulier, F., & Goldfarb, M. (1996). Receptor specificity of the fibroblast growth factor family. The Journal of biological chemistry, 271(25), 15292–15297.PubMedCrossRef Ornitz, D. M., Xu, J., Colvin, J. S., McEwen, D. G., MacArthur, C. A., Coulier, F., & Goldfarb, M. (1996). Receptor specificity of the fibroblast growth factor family. The Journal of biological chemistry, 271(25), 15292–15297.PubMedCrossRef
36.
Zurück zum Zitat Itoh, N. (2007). The Fgf families in humans, mice, and zebrafish: their evolutional processes and roles in development, metabolism, and disease. Biological & pharmaceutical bulletin, 30(10), 1819–1825.CrossRef Itoh, N. (2007). The Fgf families in humans, mice, and zebrafish: their evolutional processes and roles in development, metabolism, and disease. Biological & pharmaceutical bulletin, 30(10), 1819–1825.CrossRef
37.
Zurück zum Zitat Hehr, U., & Muenke, M. (1999). Craniosynostosis syndromes: from genes to premature fusion of skull bones. Molecular genetics and metabolism, 68(2), 139–151.PubMedCrossRef Hehr, U., & Muenke, M. (1999). Craniosynostosis syndromes: from genes to premature fusion of skull bones. Molecular genetics and metabolism, 68(2), 139–151.PubMedCrossRef
38.
Zurück zum Zitat Wu, D. Q., Kan, M. K., Sato, G. H., Okamoto, T., & Sato, J. D. (1991). Characterization and molecular cloning of a putative binding protein for heparin-binding growth factors. The Journal of biological chemistry, 266(25), 16778–16785.PubMed Wu, D. Q., Kan, M. K., Sato, G. H., Okamoto, T., & Sato, J. D. (1991). Characterization and molecular cloning of a putative binding protein for heparin-binding growth factors. The Journal of biological chemistry, 266(25), 16778–16785.PubMed
39.
Zurück zum Zitat Böttcher, R. T., Pollet, N., Delius, H., & Niehrs, C. (2004). The transmembrane protein XFLRT3 forms a complex with FGF receptors and promotes FGF signalling. Nature cell biology, 6(1), 38–44.PubMedCrossRef Böttcher, R. T., Pollet, N., Delius, H., & Niehrs, C. (2004). The transmembrane protein XFLRT3 forms a complex with FGF receptors and promotes FGF signalling. Nature cell biology, 6(1), 38–44.PubMedCrossRef
40.
41.
Zurück zum Zitat Hacohen, N., Kramer, S., Sutherland, D., Hiromi, Y., & Krasnow, M. A. (1998). sprouty encodes a novel antagonist of FGF signaling that patterns apical branching of the Drosophila airways. Cell, 92(2), 253–263.PubMedCrossRef Hacohen, N., Kramer, S., Sutherland, D., Hiromi, Y., & Krasnow, M. A. (1998). sprouty encodes a novel antagonist of FGF signaling that patterns apical branching of the Drosophila airways. Cell, 92(2), 253–263.PubMedCrossRef
42.
Zurück zum Zitat Cabrita, M. A., & Christofori, G. (2008). Sprouty proteins, masterminds of receptor tyrosine kinase signaling. Angiogenesis, 11(1), 53–62.PubMedCrossRef Cabrita, M. A., & Christofori, G. (2008). Sprouty proteins, masterminds of receptor tyrosine kinase signaling. Angiogenesis, 11(1), 53–62.PubMedCrossRef
43.
Zurück zum Zitat Tsang, M., & Dawid, I. B. (2004). Promotion and attenuation of FGF signaling through the Ras-MAPK pathway. Science’s STKE : signal transduction knowledge environment, 2004(228), pe17. Tsang, M., & Dawid, I. B. (2004). Promotion and attenuation of FGF signaling through the Ras-MAPK pathway. Science’s STKE : signal transduction knowledge environment, 2004(228), pe17.
44.
Zurück zum Zitat Steinberg, F., Zhuang, L., Beyeler, M., Kälin, R. E., Mullis, P. E., Brändli, A. W., & Trueb, B. (2010). The FGFRL1 receptor is shed from cell membranes, binds fibroblast growth factors (FGFs), and antagonizes FGF signaling in Xenopus embryos. The Journal of biological chemistry, 285(3), 2193–2202.PubMedCentralPubMedCrossRef Steinberg, F., Zhuang, L., Beyeler, M., Kälin, R. E., Mullis, P. E., Brändli, A. W., & Trueb, B. (2010). The FGFRL1 receptor is shed from cell membranes, binds fibroblast growth factors (FGFs), and antagonizes FGF signaling in Xenopus embryos. The Journal of biological chemistry, 285(3), 2193–2202.PubMedCentralPubMedCrossRef
45.
Zurück zum Zitat Trueb, B. (2011). Biology of FGFRL1, the fifth fibroblast growth factor receptor. Cellular and molecular life sciences : CMLS, 68(6), 951–964.PubMedCrossRef Trueb, B. (2011). Biology of FGFRL1, the fifth fibroblast growth factor receptor. Cellular and molecular life sciences : CMLS, 68(6), 951–964.PubMedCrossRef
46.
Zurück zum Zitat Rieckmann, T., Zhuang, L., Flück, C. E., & Trueb, B. (2009). Characterization of the first FGFRL1 mutation identified in a craniosynostosis patient. Biochimica et biophysica acta, 1792(2), 112–121.PubMedCrossRef Rieckmann, T., Zhuang, L., Flück, C. E., & Trueb, B. (2009). Characterization of the first FGFRL1 mutation identified in a craniosynostosis patient. Biochimica et biophysica acta, 1792(2), 112–121.PubMedCrossRef
47.
Zurück zum Zitat Schild, C., & Trueb, B. (2005). Aberrant expression of FGFRL1, a novel FGF receptor, in ovarian tumors. International journal of molecular medicine, 16(6), 1169–1173.PubMed Schild, C., & Trueb, B. (2005). Aberrant expression of FGFRL1, a novel FGF receptor, in ovarian tumors. International journal of molecular medicine, 16(6), 1169–1173.PubMed
48.
Zurück zum Zitat Garcia, S., Dirat, B., Tognacci, T., Rochet, N., Mouska, X., Bonnafous, S., & Gouze, E. (2013). Postnatal soluble FGFR3 therapy rescues achondroplasia symptoms and restores bone growth in mice. Science Translational Medicine, 5(203), 203ra–124. doi:10.1126/scitranslmed.3006247.CrossRef Garcia, S., Dirat, B., Tognacci, T., Rochet, N., Mouska, X., Bonnafous, S., & Gouze, E. (2013). Postnatal soluble FGFR3 therapy rescues achondroplasia symptoms and restores bone growth in mice. Science Translational Medicine, 5(203), 203ra–124. doi:10.​1126/​scitranslmed.​3006247.CrossRef
49.
Zurück zum Zitat Cohen, M. M. (2003). Neoplasms associated with alterations in fibroblast growth factor receptors. American journal of medical genetics Part A, 119A(2), 97–100.PubMedCrossRef Cohen, M. M. (2003). Neoplasms associated with alterations in fibroblast growth factor receptors. American journal of medical genetics Part A, 119A(2), 97–100.PubMedCrossRef
50.
Zurück zum Zitat Andreou, A., Lamy, A., Layet, V., Cailliez, D., Gobet, F., Pfister, C., & Frebourg, T. (2006). Early-onset low-grade papillary carcinoma of the bladder associated with Apert syndrome and a germline FGFR2 mutation (Pro253Arg). American journal of medical genetics Part A, 140(20), 2245–2247.PubMedCrossRef Andreou, A., Lamy, A., Layet, V., Cailliez, D., Gobet, F., Pfister, C., & Frebourg, T. (2006). Early-onset low-grade papillary carcinoma of the bladder associated with Apert syndrome and a germline FGFR2 mutation (Pro253Arg). American journal of medical genetics Part A, 140(20), 2245–2247.PubMedCrossRef
51.
Zurück zum Zitat Rouzier, C., Soler, C., Hofman, P., Brennetot, C., Bieth, E., & Pedeutour, F. (2008). Ovarian dysgerminoma and Apert syndrome. Pediatric blood & cancer, 50(3), 696–698.CrossRef Rouzier, C., Soler, C., Hofman, P., Brennetot, C., Bieth, E., & Pedeutour, F. (2008). Ovarian dysgerminoma and Apert syndrome. Pediatric blood & cancer, 50(3), 696–698.CrossRef
52.
Zurück zum Zitat Barbosa, M., Almeida, M. do R., Reis-Lima, M., Pinto-Basto, J., & dos Santos, H. G. (2009). Muenke syndrome with osteochondroma. American journal of medical genetics. Part A, 149A(2), 260–261. Barbosa, M., Almeida, M. do R., Reis-Lima, M., Pinto-Basto, J., & dos Santos, H. G. (2009). Muenke syndrome with osteochondroma. American journal of medical genetics. Part A, 149A(2), 260–261.
53.
Zurück zum Zitat Bourdeaut, F., Miquel, C., Di Rocco, F., Grison, C., Richer, W., Brugieres, L., & Collet, C. (2013). Germline mutations in FGF receptors and medulloblastomas. American journal of medical genetics Part A, 161A(2), 382–385.PubMedCrossRef Bourdeaut, F., Miquel, C., Di Rocco, F., Grison, C., Richer, W., Brugieres, L., & Collet, C. (2013). Germline mutations in FGF receptors and medulloblastomas. American journal of medical genetics Part A, 161A(2), 382–385.PubMedCrossRef
54.
Zurück zum Zitat Logié, A., Dunois-Lardé, C., Rosty, C., Levrel, O., Blanche, M., Ribeiro, A., & Radvanyi, F. (2005). Activating mutations of the tyrosine kinase receptor FGFR3 are associated with benign skin tumors in mice and humans. Human molecular genetics, 14(9), 1153–1160.PubMedCrossRef Logié, A., Dunois-Lardé, C., Rosty, C., Levrel, O., Blanche, M., Ribeiro, A., & Radvanyi, F. (2005). Activating mutations of the tyrosine kinase receptor FGFR3 are associated with benign skin tumors in mice and humans. Human molecular genetics, 14(9), 1153–1160.PubMedCrossRef
55.
Zurück zum Zitat Hafner, C., van Oers, J. M. M., Vogt, T., Landthaler, M., Stoehr, R., Blaszyk, H., & Hartmann, A. (2006). Mosaicism of activating FGFR3 mutations in human skin causes epidermal nevi. Journal of Clinical Investigation, 116(8), 2201–2207.PubMedCentralPubMedCrossRef Hafner, C., van Oers, J. M. M., Vogt, T., Landthaler, M., Stoehr, R., Blaszyk, H., & Hartmann, A. (2006). Mosaicism of activating FGFR3 mutations in human skin causes epidermal nevi. Journal of Clinical Investigation, 116(8), 2201–2207.PubMedCentralPubMedCrossRef
57.
Zurück zum Zitat Hattori, Y., Itoh, H., Uchino, S., Hosokawa, K., Ochiai, A., Ino, Y., & Terada, M. (1996). Immunohistochemical detection of K-sam protein in stomach cancer. Clinical cancer research : an official journal of the American Association for Cancer Research, 2(8), 1373–1381. Hattori, Y., Itoh, H., Uchino, S., Hosokawa, K., Ochiai, A., Ino, Y., & Terada, M. (1996). Immunohistochemical detection of K-sam protein in stomach cancer. Clinical cancer research : an official journal of the American Association for Cancer Research, 2(8), 1373–1381.
58.
Zurück zum Zitat Gust, K. M., McConkey, D. J., Awrey, S., Hegarty, P. K., Qing, J., Bondaruk, J., & Black, P. C. (2013). Fibroblast growth factor receptor 3 is a rational therapeutic target in bladder cancer. Molecular Cancer Therapeutics, 12(7), 1245–1254.PubMedCentralPubMedCrossRef Gust, K. M., McConkey, D. J., Awrey, S., Hegarty, P. K., Qing, J., Bondaruk, J., & Black, P. C. (2013). Fibroblast growth factor receptor 3 is a rational therapeutic target in bladder cancer. Molecular Cancer Therapeutics, 12(7), 1245–1254.PubMedCentralPubMedCrossRef
59.
Zurück zum Zitat Gartside, M. G., Chen, H., Ibrahimi, O. A., Byron, S. A., Curtis, A. V., Wellens, C. L., & Pollock, P. M. (2009). Loss-of-function fibroblast growth factor receptor-2 mutations in melanoma. Molecular cancer research : MCR, 7(1), 41–54.PubMedCentralPubMedCrossRef Gartside, M. G., Chen, H., Ibrahimi, O. A., Byron, S. A., Curtis, A. V., Wellens, C. L., & Pollock, P. M. (2009). Loss-of-function fibroblast growth factor receptor-2 mutations in melanoma. Molecular cancer research : MCR, 7(1), 41–54.PubMedCentralPubMedCrossRef
60.
Zurück zum Zitat Heist, R. S., Mino-Kenudson, M., Sequist, L. V., Tammireddy, S., Morrissey, L., Christiani, D. C., & Iafrate, A. J. (2012). FGFR1 Amplification in Squamous Cell Carcinoma of The Lung. Journal of Thoracic Oncology, 7(12), 1775–1780.PubMedCentralPubMedCrossRef Heist, R. S., Mino-Kenudson, M., Sequist, L. V., Tammireddy, S., Morrissey, L., Christiani, D. C., & Iafrate, A. J. (2012). FGFR1 Amplification in Squamous Cell Carcinoma of The Lung. Journal of Thoracic Oncology, 7(12), 1775–1780.PubMedCentralPubMedCrossRef
61.
Zurück zum Zitat Weiss, J., Sos, M. L., Seidel, D., & Peifer, M. (2010). Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Science translational medicine, 2(62), 62ra93.PubMedCentralPubMedCrossRef Weiss, J., Sos, M. L., Seidel, D., & Peifer, M. (2010). Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Science translational medicine, 2(62), 62ra93.PubMedCentralPubMedCrossRef
62.
Zurück zum Zitat Albiges, L., Quidville, V., Valent, A., Mathieu, M. C., Drusch, F., Koscielny, S., … Andre, F. (2009). FGFR1 amplification and FGF gain in breast cancer. In San Antonio Breast Cancer Symposium. Institut Gustave Roussy, villejuif, France. Albiges, L., Quidville, V., Valent, A., Mathieu, M. C., Drusch, F., Koscielny, S., … Andre, F. (2009). FGFR1 amplification and FGF gain in breast cancer. In San Antonio Breast Cancer Symposium. Institut Gustave Roussy, villejuif, France.
64.
Zurück zum Zitat Liu, J., Guzman, M. A., Pezanowski, D., Patel, D., Hauptman, J., Keisling, M., & de Chadarévian, J.-P. (2011). FOXO1-FGFR1 fusion and amplification in a solid variant of alveolar rhabdomyosarcoma. Modern Pathology: An Official Journal of the United States and Canadian Academy of Pathology, Inc, 24(10), 1327–1335. doi:10.1038/modpathol.2011.98.CrossRef Liu, J., Guzman, M. A., Pezanowski, D., Patel, D., Hauptman, J., Keisling, M., & de Chadarévian, J.-P. (2011). FOXO1-FGFR1 fusion and amplification in a solid variant of alveolar rhabdomyosarcoma. Modern Pathology: An Official Journal of the United States and Canadian Academy of Pathology, Inc, 24(10), 1327–1335. doi:10.​1038/​modpathol.​2011.​98.CrossRef
65.
Zurück zum Zitat Grand, E. K., Chase, A. J., Heath, C., Rahemtulla, A., & Cross, N. C. P. (2004). Targeting FGFR3 in multiple myeloma: inhibition of t(4;14)-positive cells by SU5402 and PD173074. Leukemia, 18(5), 962–966. doi:10.1038/sj.leu.2403347.PubMedCrossRef Grand, E. K., Chase, A. J., Heath, C., Rahemtulla, A., & Cross, N. C. P. (2004). Targeting FGFR3 in multiple myeloma: inhibition of t(4;14)-positive cells by SU5402 and PD173074. Leukemia, 18(5), 962–966. doi:10.​1038/​sj.​leu.​2403347.PubMedCrossRef
66.
Zurück zum Zitat Yagasaki, F., Wakao, D., Yokoyama, Y., Uchida, Y., Murohashi, I., Kayano, H., & Bessho, M. (2001). Fusion of ETV6 to fibroblast growth factor receptor 3 in peripheral T-cell lymphoma with a t(4;12)(p16;p13) chromosomal translocation. Cancer Research, 61(23), 8371–8374.PubMed Yagasaki, F., Wakao, D., Yokoyama, Y., Uchida, Y., Murohashi, I., Kayano, H., & Bessho, M. (2001). Fusion of ETV6 to fibroblast growth factor receptor 3 in peripheral T-cell lymphoma with a t(4;12)(p16;p13) chromosomal translocation. Cancer Research, 61(23), 8371–8374.PubMed
67.
Zurück zum Zitat Katoh, M., & Katoh, M. (2003). Recombination cluster around FGFR2-WDR11-HTPAPL locus on human chromosome 10q26. International Journal of Molecular Medicine, 11(5), 579–583.PubMed Katoh, M., & Katoh, M. (2003). Recombination cluster around FGFR2-WDR11-HTPAPL locus on human chromosome 10q26. International Journal of Molecular Medicine, 11(5), 579–583.PubMed
68.
Zurück zum Zitat Singh, D., Chan, J. M., Zoppoli, P., Niola, F., Sullivan, R., Castano, A., & Iavarone, A. (2012). Transforming Fusions of FGFR and TACC Genes in Human Glioblastoma. Science, 337(6099), 1231–1235.PubMedCentralPubMedCrossRef Singh, D., Chan, J. M., Zoppoli, P., Niola, F., Sullivan, R., Castano, A., & Iavarone, A. (2012). Transforming Fusions of FGFR and TACC Genes in Human Glioblastoma. Science, 337(6099), 1231–1235.PubMedCentralPubMedCrossRef
69.
Zurück zum Zitat Soria, J.-C., DeBraud, F., Bahleda, R., Adamo, B., Andre, F., Dientsmann, R., & Tabernero, J. (2014). Phase I/IIa study evaluating the safety, efficacy, pharmacokinetics, and pharmacodynamics of lucitanib in advanced solid tumors. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO, 25(11), 2244–2251. doi:10.1093/annonc/mdu390.CrossRef Soria, J.-C., DeBraud, F., Bahleda, R., Adamo, B., Andre, F., Dientsmann, R., & Tabernero, J. (2014). Phase I/IIa study evaluating the safety, efficacy, pharmacokinetics, and pharmacodynamics of lucitanib in advanced solid tumors. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO, 25(11), 2244–2251. doi:10.​1093/​annonc/​mdu390.CrossRef
70.
Zurück zum Zitat Nakanishi, Y., Akiyama, N., Tsukaguchi, T., Fujii, T., Sakata, K., Sase, H., & Ishii, N. (2014). The Fibroblast Growth Factor Receptor Genetic Status as a Potential Predictor of the Sensitivity to CH5183284/Debio 1347, a Novel Selective FGFR Inhibitor. Molecular Cancer Therapeutics, 13(11), 2547–2558. doi:10.1158/1535-7163.MCT-14-0248.PubMedCrossRef Nakanishi, Y., Akiyama, N., Tsukaguchi, T., Fujii, T., Sakata, K., Sase, H., & Ishii, N. (2014). The Fibroblast Growth Factor Receptor Genetic Status as a Potential Predictor of the Sensitivity to CH5183284/Debio 1347, a Novel Selective FGFR Inhibitor. Molecular Cancer Therapeutics, 13(11), 2547–2558. doi:10.​1158/​1535-7163.​MCT-14-0248.PubMedCrossRef
71.
Zurück zum Zitat Bellovin, D. I., Palencia, S., Hestir, K., Lee, E., DeYoung, M. P., Brennan, T., & Baker, K. (2014). Abstract 5449: FP-1039/GSK3052230, an FGF ligand trap, enhances VEGF antagonist therapy in preclinical models of RCC and HCC. Cancer Research, 74(19 Supplement), 5449–5449. doi:10.1158/1538-7445.AM2014-5449.CrossRef Bellovin, D. I., Palencia, S., Hestir, K., Lee, E., DeYoung, M. P., Brennan, T., & Baker, K. (2014). Abstract 5449: FP-1039/GSK3052230, an FGF ligand trap, enhances VEGF antagonist therapy in preclinical models of RCC and HCC. Cancer Research, 74(19 Supplement), 5449–5449. doi:10.​1158/​1538-7445.​AM2014-5449.CrossRef
72.
Zurück zum Zitat Sohl, C. D., Ryan, M. R., Luo, B., Frey, K. M., & Anderson, K. S. (2015). Illuminating the Molecular Mechanisms of Tyrosine Kinase Inhibitor Resistance for the FGFR1 Gatekeeper Mutation: The Achilles’ Heel of Targeted Therapy. ACS chemical biology. doi:10.1021/acschembio.5b00014.PubMedCentralPubMed Sohl, C. D., Ryan, M. R., Luo, B., Frey, K. M., & Anderson, K. S. (2015). Illuminating the Molecular Mechanisms of Tyrosine Kinase Inhibitor Resistance for the FGFR1 Gatekeeper Mutation: The Achilles’ Heel of Targeted Therapy. ACS chemical biology. doi:10.​1021/​acschembio.​5b00014.PubMedCentralPubMed
73.
Zurück zum Zitat Chell, V., Balmanno, K., Little, A. S., Wilson, M., Andrews, S., Blockley, L., & Cook, S. J. (2013). Tumour cell responses to new fibroblast growth factor receptor tyrosine kinase inhibitors and identification of a gatekeeper mutation in FGFR3 as a mechanism of acquired resistance. Oncogene, 32(25), 3059–3070. doi:10.1038/onc.2012.319.PubMedCrossRef Chell, V., Balmanno, K., Little, A. S., Wilson, M., Andrews, S., Blockley, L., & Cook, S. J. (2013). Tumour cell responses to new fibroblast growth factor receptor tyrosine kinase inhibitors and identification of a gatekeeper mutation in FGFR3 as a mechanism of acquired resistance. Oncogene, 32(25), 3059–3070. doi:10.​1038/​onc.​2012.​319.PubMedCrossRef
74.
Zurück zum Zitat Byron, S. A., Chen, H., Wortmann, A., Loch, D., Gartside, M. G., Dehkhoda, F., & Pollock, P. M. (2013). The N550K/H mutations in FGFR2 confer differential resistance to PD173074, dovitinib, and ponatinib ATP-competitive inhibitors. Neoplasia (New York, N.Y.), 15(8), 975–988.PubMedCentralCrossRef Byron, S. A., Chen, H., Wortmann, A., Loch, D., Gartside, M. G., Dehkhoda, F., & Pollock, P. M. (2013). The N550K/H mutations in FGFR2 confer differential resistance to PD173074, dovitinib, and ponatinib ATP-competitive inhibitors. Neoplasia (New York, N.Y.), 15(8), 975–988.PubMedCentralCrossRef
75.
Zurück zum Zitat Crystal, A. S., Shaw, A. T., Sequist, L. V., Friboulet, L., Niederst, M. J., Lockerman, E. L., & Engelman, J. A. (2014). Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science, 346(6216), 1480–1486. doi:10.1126/science.1254721.PubMedCentralPubMedCrossRef Crystal, A. S., Shaw, A. T., Sequist, L. V., Friboulet, L., Niederst, M. J., Lockerman, E. L., & Engelman, J. A. (2014). Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science, 346(6216), 1480–1486. doi:10.​1126/​science.​1254721.PubMedCentralPubMedCrossRef
76.
Zurück zum Zitat Harbinski, F., Craig, V. J., Sanghavi, S., Jeffery, D., Liu, L., Sheppard, K. A., & Tiedt, R. (2012). Rescue screens with secreted proteins reveal compensatory potential of receptor tyrosine kinases in driving cancer growth. Cancer Discovery, 2(10), 948–959. doi:10.1158/2159-8290.CD-12-0237.PubMedCrossRef Harbinski, F., Craig, V. J., Sanghavi, S., Jeffery, D., Liu, L., Sheppard, K. A., & Tiedt, R. (2012). Rescue screens with secreted proteins reveal compensatory potential of receptor tyrosine kinases in driving cancer growth. Cancer Discovery, 2(10), 948–959. doi:10.​1158/​2159-8290.​CD-12-0237.PubMedCrossRef
77.
Zurück zum Zitat Javidi-Sharifi, N., Traer, E., Martinez, J., Gupta, A., Taguchi, T., Dunlap, J., & Tyner, J. W. (2015). Crosstalk between KIT and FGFR3 Promotes Gastrointestinal Stromal Tumor Cell Growth and Drug Resistance. Cancer Research, 75(5), 880–891. doi:10.1158/0008-5472.CAN-14-0573.PubMedCrossRef Javidi-Sharifi, N., Traer, E., Martinez, J., Gupta, A., Taguchi, T., Dunlap, J., & Tyner, J. W. (2015). Crosstalk between KIT and FGFR3 Promotes Gastrointestinal Stromal Tumor Cell Growth and Drug Resistance. Cancer Research, 75(5), 880–891. doi:10.​1158/​0008-5472.​CAN-14-0573.PubMedCrossRef
79.
Zurück zum Zitat Schwaederlé, M., Daniels, G. A., Piccioni, D. E., Fanta, P. T., Schwab, R. B., Shimabukuro, K. A., … Kurzrock, R. (2014). Cyclin alterations in diverse cancers: Outcome and co-amplification network. Oncotarget. Schwaederlé, M., Daniels, G. A., Piccioni, D. E., Fanta, P. T., Schwab, R. B., Shimabukuro, K. A., … Kurzrock, R. (2014). Cyclin alterations in diverse cancers: Outcome and co-amplification network. Oncotarget.
80.
Zurück zum Zitat Wheler, J. J., Parker, B. A., Lee, J. J., Atkins, J. T., Janku, F., Tsimberidou, A. M., … Kurzrock, R. (2014). Unique molecular signatures as a hallmark of patients with metastatic breast cancer: Implications for current treatment paradigms. Oncotarget. Wheler, J. J., Parker, B. A., Lee, J. J., Atkins, J. T., Janku, F., Tsimberidou, A. M., … Kurzrock, R. (2014). Unique molecular signatures as a hallmark of patients with metastatic breast cancer: Implications for current treatment paradigms. Oncotarget.
82.
Zurück zum Zitat Tsimberidou, A.-M., Iskander, N. G., Hong, D. S., Wheler, J. J., Falchook, G. S., Fu, S., & Kurzrock, R. (2012). Personalized medicine in a phase I clinical trials program: the MD Anderson Cancer Center initiative. Clinical cancer research: an official journal of the American Association for Cancer Research, 18(22), 6373–6383. doi:10.1158/1078-0432.CCR-12-1627.CrossRef Tsimberidou, A.-M., Iskander, N. G., Hong, D. S., Wheler, J. J., Falchook, G. S., Fu, S., & Kurzrock, R. (2012). Personalized medicine in a phase I clinical trials program: the MD Anderson Cancer Center initiative. Clinical cancer research: an official journal of the American Association for Cancer Research, 18(22), 6373–6383. doi:10.​1158/​1078-0432.​CCR-12-1627.CrossRef
84.
Zurück zum Zitat Cohen, M. M. (1993). Pfeiffer syndrome update, clinical subtypes, and guidelines for differential diagnosis. American journal of medical genetics, 45(3), 300–307.PubMedCrossRef Cohen, M. M. (1993). Pfeiffer syndrome update, clinical subtypes, and guidelines for differential diagnosis. American journal of medical genetics, 45(3), 300–307.PubMedCrossRef
86.
Zurück zum Zitat Ibrahimi, O. A., Zhang, F., Eliseenkova, A. V., Linhardt, R. J., & Mohammadi, M. (2004). Proline to arginine mutations in FGF receptors 1 and 3 result in Pfeiffer and Muenke craniosynostosis syndromes through enhancement of FGF binding affinity. Human Molecular Genetics, 13(1), 69–78. doi:10.1093/hmg/ddh011.PubMedCrossRef Ibrahimi, O. A., Zhang, F., Eliseenkova, A. V., Linhardt, R. J., & Mohammadi, M. (2004). Proline to arginine mutations in FGF receptors 1 and 3 result in Pfeiffer and Muenke craniosynostosis syndromes through enhancement of FGF binding affinity. Human Molecular Genetics, 13(1), 69–78. doi:10.​1093/​hmg/​ddh011.PubMedCrossRef
87.
Zurück zum Zitat Davies, H., Hunter, C., Smith, R., Stephens, P., Greenman, C., Bignell, G., … Futreal, P. A. (2005). Somatic Mutations of the Protein Kinase Gene Family in Human Lung Cancer. Davies, H., Hunter, C., Smith, R., Stephens, P., Greenman, C., Bignell, G., … Futreal, P. A. (2005). Somatic Mutations of the Protein Kinase Gene Family in Human Lung Cancer.
88.
Zurück zum Zitat Ruhe, J. E., Streit, S., Hart, S., Wong, C.-H., Specht, K., Knyazev, P., … Ullrich, A. (2007). Genetic Alterations in the Tyrosine Kinase Transcriptome of Human Cancer Cell Lines. Cancer Research. Ruhe, J. E., Streit, S., Hart, S., Wong, C.-H., Specht, K., Knyazev, P., … Ullrich, A. (2007). Genetic Alterations in the Tyrosine Kinase Transcriptome of Human Cancer Cell Lines. Cancer Research.
89.
Zurück zum Zitat White, K. E., Cabral, J. M., Davis, S. I., Fishburn, T., Evans, W. E., Ichikawa, S., & Econs, M. J. (2005). Mutations that cause osteoglophonic dysplasia define novel roles for FGFR1 in bone elongation. American journal of human genetics, 76(2), 361–367.PubMedCentralPubMedCrossRef White, K. E., Cabral, J. M., Davis, S. I., Fishburn, T., Evans, W. E., Ichikawa, S., & Econs, M. J. (2005). Mutations that cause osteoglophonic dysplasia define novel roles for FGFR1 in bone elongation. American journal of human genetics, 76(2), 361–367.PubMedCentralPubMedCrossRef
90.
Zurück zum Zitat Riminucci, M., Collins, M. T., Fedarko, N. S., Cherman, N., Corsi, A., White, K. E., & Gehron Robey, P. (2003). FGF-23 in fibrous dysplasia of bone and its relationship to renal phosphate wasting. Journal of Clinical Investigation, 112(5), 683–692.PubMedCentralPubMedCrossRef Riminucci, M., Collins, M. T., Fedarko, N. S., Cherman, N., Corsi, A., White, K. E., & Gehron Robey, P. (2003). FGF-23 in fibrous dysplasia of bone and its relationship to renal phosphate wasting. Journal of Clinical Investigation, 112(5), 683–692.PubMedCentralPubMedCrossRef
91.
Zurück zum Zitat Moloney, D. M., Slaney, S. F., Oldridge, M., Wall, S. A., Sahlin, P., Stenman, G., & Wilkie, A. O. (1996). Exclusive paternal origin of new mutations in Apert syndrome. Nature genetics, 13(1), 48–53.PubMedCrossRef Moloney, D. M., Slaney, S. F., Oldridge, M., Wall, S. A., Sahlin, P., Stenman, G., & Wilkie, A. O. (1996). Exclusive paternal origin of new mutations in Apert syndrome. Nature genetics, 13(1), 48–53.PubMedCrossRef
92.
Zurück zum Zitat Miraoui, H., Ringe, J., & Häupl, T. (2010). Increased EFG-and PDGFα-receptor signaling by mutant FGF-receptor 2 contributes to osteoblast dysfunction in Apert craniosynostosis. Human molecular \ldots. Miraoui, H., Ringe, J., & Häupl, T. (2010). Increased EFG-and PDGFα-receptor signaling by mutant FGF-receptor 2 contributes to osteoblast dysfunction in Apert craniosynostosis. Human molecular \ldots.
93.
Zurück zum Zitat Pollock, P. M., Gartside, M. G., Dejeza, L. C., Powell, M. A., Mallon, M. A., Davies, H., & Goodfellow, P. J. (2007). Frequent activating FGFR2 mutations in endometrial carcinomas parallel germline mutations associated with craniosynostosis and skeletal dysplasia syndromes. Oncogene, 26(50), 7158–7162.PubMedCentralPubMedCrossRef Pollock, P. M., Gartside, M. G., Dejeza, L. C., Powell, M. A., Mallon, M. A., Davies, H., & Goodfellow, P. J. (2007). Frequent activating FGFR2 mutations in endometrial carcinomas parallel germline mutations associated with craniosynostosis and skeletal dysplasia syndromes. Oncogene, 26(50), 7158–7162.PubMedCentralPubMedCrossRef
94.
Zurück zum Zitat Byron, S. A., Gartside, M., Powell, M. A., Wellens, C. L., Gao, F., Mutch, D. G., & Pollock, P. M. (2012). FGFR2 point mutations in 466 endometrioid endometrial tumors: relationship with MSI, KRAS, PIK3CA, CTNNB1 mutations and clinicopathological features. PLoS ONE, 7(2), e30801.PubMedCentralPubMedCrossRef Byron, S. A., Gartside, M., Powell, M. A., Wellens, C. L., Gao, F., Mutch, D. G., & Pollock, P. M. (2012). FGFR2 point mutations in 466 endometrioid endometrial tumors: relationship with MSI, KRAS, PIK3CA, CTNNB1 mutations and clinicopathological features. PLoS ONE, 7(2), e30801.PubMedCentralPubMedCrossRef
95.
Zurück zum Zitat Passos-Bueno, M. R., Wilcox, W. R., Jabs, E. W., Serti, A. L., Alonso, L. G., & Kitoh, H. (1999). Clinical spectrum of fibroblast growth factor receptor mutations. Human Mutation, 14(2), 115–125.PubMedCrossRef Passos-Bueno, M. R., Wilcox, W. R., Jabs, E. W., Serti, A. L., Alonso, L. G., & Kitoh, H. (1999). Clinical spectrum of fibroblast growth factor receptor mutations. Human Mutation, 14(2), 115–125.PubMedCrossRef
96.
Zurück zum Zitat Reardon, W., Winter, R. M., Rutland, P., Pulleyn, L. J., Jones, B. M., & Malcolm, S. (1994). Mutations in the fibroblast growth factor receptor 2 gene cause Crouzon syndrome. Nature genetics, 8(1), 98–103.PubMedCrossRef Reardon, W., Winter, R. M., Rutland, P., Pulleyn, L. J., Jones, B. M., & Malcolm, S. (1994). Mutations in the fibroblast growth factor receptor 2 gene cause Crouzon syndrome. Nature genetics, 8(1), 98–103.PubMedCrossRef
97.
Zurück zum Zitat Glaser, R. L., Jiang, W., Boyadjiev, S. A., Tran, A. K., Zachary, A. A., Van Maldergem, L., & Jabs, E. W. (2000). Paternal origin of FGFR2 mutations in sporadic cases of Crouzon syndrome and Pfeiffer syndrome. American journal of human genetics, 66(3), 768–777.PubMedCentralPubMedCrossRef Glaser, R. L., Jiang, W., Boyadjiev, S. A., Tran, A. K., Zachary, A. A., Van Maldergem, L., & Jabs, E. W. (2000). Paternal origin of FGFR2 mutations in sporadic cases of Crouzon syndrome and Pfeiffer syndrome. American journal of human genetics, 66(3), 768–777.PubMedCentralPubMedCrossRef
98.
Zurück zum Zitat Jang, J. H., Shin, K. H., & Park, J. G. (2001). Mutations in fibroblast growth factor receptor 2 and fibroblast growth factor receptor 3 genes associated with human gastric and colorectal cancers. Cancer Research, 61(9), 3541–3543.PubMed Jang, J. H., Shin, K. H., & Park, J. G. (2001). Mutations in fibroblast growth factor receptor 2 and fibroblast growth factor receptor 3 genes associated with human gastric and colorectal cancers. Cancer Research, 61(9), 3541–3543.PubMed
99.
Zurück zum Zitat Chokdeemboon, C., Mahatumarat, C., Rojvachiranonda, N., Tongkobpetch, S., Suphapeetiporn, K., & Shotelersuk, V. (2013). FGFR1 and FGFR2 mutations in Pfeiffer syndrome. The Journal of craniofacial surgery, 24(1), 150–152.PubMedCrossRef Chokdeemboon, C., Mahatumarat, C., Rojvachiranonda, N., Tongkobpetch, S., Suphapeetiporn, K., & Shotelersuk, V. (2013). FGFR1 and FGFR2 mutations in Pfeiffer syndrome. The Journal of craniofacial surgery, 24(1), 150–152.PubMedCrossRef
100.
Zurück zum Zitat Cornejo-Roldan, L. R., Roessler, E., & Muenke, M. (1999). Analysis of the mutational spectrum of the FGFR2 gene in Pfeiffer syndrome - Springer. Human genetics. Cornejo-Roldan, L. R., Roessler, E., & Muenke, M. (1999). Analysis of the mutational spectrum of the FGFR2 gene in Pfeiffer syndrome - Springer. Human genetics.
101.
Zurück zum Zitat Chen, L., Adar, R., Yang, X., Monsonego, E. O., Li, C., Hauschka, P. V., & Deng, C. X. (1999). Gly369Cys mutation in mouse FGFR3 causes achondroplasia by affecting both chondrogenesis and osteogenesis. Journal of Clinical Investigation, 104(11), 1517–1525.PubMedCentralPubMedCrossRef Chen, L., Adar, R., Yang, X., Monsonego, E. O., Li, C., Hauschka, P. V., & Deng, C. X. (1999). Gly369Cys mutation in mouse FGFR3 causes achondroplasia by affecting both chondrogenesis and osteogenesis. Journal of Clinical Investigation, 104(11), 1517–1525.PubMedCentralPubMedCrossRef
102.
Zurück zum Zitat Monsonego-Ornan, E., Adar, R., Feferman, T., Segev, O., & Yayon, A. (2000). The transmembrane mutation G380R in fibroblast growth factor receptor 3 uncouples ligand-mediated receptor activation from down-regulation. Molecular and cellular biology, 20(2), 516–522.PubMedCentralPubMedCrossRef Monsonego-Ornan, E., Adar, R., Feferman, T., Segev, O., & Yayon, A. (2000). The transmembrane mutation G380R in fibroblast growth factor receptor 3 uncouples ligand-mediated receptor activation from down-regulation. Molecular and cellular biology, 20(2), 516–522.PubMedCentralPubMedCrossRef
103.
Zurück zum Zitat Henderson, J. E., Naski, M. C., Aarts, M. M., Wang, D., Cheng, L., Goltzman, D., & Ornitz, D. M. (2000). Expression of FGFR3 with the G380R Achondroplasia Mutation Inhibits Proliferation and Maturation of CFK2 Chondrocytic Cells. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research, 15(1), 155–165.CrossRef Henderson, J. E., Naski, M. C., Aarts, M. M., Wang, D., Cheng, L., Goltzman, D., & Ornitz, D. M. (2000). Expression of FGFR3 with the G380R Achondroplasia Mutation Inhibits Proliferation and Maturation of CFK2 Chondrocytic Cells. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research, 15(1), 155–165.CrossRef
104.
Zurück zum Zitat Knowles, M. A. (2008). Novel therapeutic targets in bladder cancer: mutation and expression of FGF receptors. Future oncology (London, England), 4(1), 71–83.CrossRef Knowles, M. A. (2008). Novel therapeutic targets in bladder cancer: mutation and expression of FGF receptors. Future oncology (London, England), 4(1), 71–83.CrossRef
105.
Zurück zum Zitat Ahmad, I., Iwata, T., & Leung, H. Y. (2012). Mechanisms of FGFR-mediated carcinogenesis. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, 1823(4), 850–860.CrossRef Ahmad, I., Iwata, T., & Leung, H. Y. (2012). Mechanisms of FGFR-mediated carcinogenesis. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, 1823(4), 850–860.CrossRef
106.
Zurück zum Zitat Delezoide, A. L., Lasselin-Benoist, C., Legeai-Mallet, L., Brice, P., Senée, V., Yayon, A., & Bonaventure, J. (1997). Abnormal FGFR 3 expression in cartilage of thanatophoric dysplasia fetuses. Human molecular genetics, 6(11), 1899–1906.PubMedCrossRef Delezoide, A. L., Lasselin-Benoist, C., Legeai-Mallet, L., Brice, P., Senée, V., Yayon, A., & Bonaventure, J. (1997). Abnormal FGFR 3 expression in cartilage of thanatophoric dysplasia fetuses. Human molecular genetics, 6(11), 1899–1906.PubMedCrossRef
107.
Zurück zum Zitat Wilcox, W. R., Tavormina, P. L., Krakow, D., Kitoh, H., Lachman, R. S., Wasmuth, J. J., & Rimoin, D. L. (1998). Molecular, radiologic, and histopathologic correlations in thanatophoric dysplasia. American journal of medical genetics, 78(3), 274–281.PubMedCrossRef Wilcox, W. R., Tavormina, P. L., Krakow, D., Kitoh, H., Lachman, R. S., Wasmuth, J. J., & Rimoin, D. L. (1998). Molecular, radiologic, and histopathologic correlations in thanatophoric dysplasia. American journal of medical genetics, 78(3), 274–281.PubMedCrossRef
108.
Zurück zum Zitat Brodie, S. G., Kitoh, H., Lachman, R. S., Nolasco, L. M., Mekikian, P. B., & Wilcox, W. R. (1999). Platyspondylic lethal skeletal dysplasia, San Diego type, is caused by FGFR3 mutations. American journal of medical genetics, 84(5), 476–480.PubMedCrossRef Brodie, S. G., Kitoh, H., Lachman, R. S., Nolasco, L. M., Mekikian, P. B., & Wilcox, W. R. (1999). Platyspondylic lethal skeletal dysplasia, San Diego type, is caused by FGFR3 mutations. American journal of medical genetics, 84(5), 476–480.PubMedCrossRef
109.
Zurück zum Zitat Di Martino, E., Tomlinson, D. C., & Knowles, M. A. (2012). A Decade of FGF Receptor Research in Bladder Cancer: Past, Present, and Future Challenges. Advances in Urology, 2012, e429213. doi:10.1155/2012/429213.CrossRef Di Martino, E., Tomlinson, D. C., & Knowles, M. A. (2012). A Decade of FGF Receptor Research in Bladder Cancer: Past, Present, and Future Challenges. Advances in Urology, 2012, e429213. doi:10.​1155/​2012/​429213.CrossRef
112.
Zurück zum Zitat Webster, M. K., D’Avis, P. Y., Robertson, S. C., & Donoghue, D. J. (1996). Profound ligand-independent kinase activation of fibroblast growth factor receptor 3 by the activation loop mutation responsible for a lethal skeletal dysplasia, thanatophoric dysplasia type II. Molecular and cellular biology, 16(8), 4081–4087.PubMedCentralPubMedCrossRef Webster, M. K., D’Avis, P. Y., Robertson, S. C., & Donoghue, D. J. (1996). Profound ligand-independent kinase activation of fibroblast growth factor receptor 3 by the activation loop mutation responsible for a lethal skeletal dysplasia, thanatophoric dysplasia type II. Molecular and cellular biology, 16(8), 4081–4087.PubMedCentralPubMedCrossRef
113.
Zurück zum Zitat Lievens, P. M.-J., & Liboi, E. (2003). The thanatophoric dysplasia type II mutation hampers complete maturation of fibroblast growth factor receptor 3 (FGFR3), which activates signal transducer and activator of transcription 1 (STAT1) from the endoplasmic reticulum. The Journal of biological chemistry, 278(19), 17344–17349.PubMedCrossRef Lievens, P. M.-J., & Liboi, E. (2003). The thanatophoric dysplasia type II mutation hampers complete maturation of fibroblast growth factor receptor 3 (FGFR3), which activates signal transducer and activator of transcription 1 (STAT1) from the endoplasmic reticulum. The Journal of biological chemistry, 278(19), 17344–17349.PubMedCrossRef
114.
Zurück zum Zitat Otsuka, M., Mizuki, M., Fujita, J., Kang, S., & Kanakura, Y. (2011, January). Constitutively active FGFR3 with Lys650Glu mutation enhances bortezomib sensitivity in plasma cell malignancy. Anticancer Research. Otsuka, M., Mizuki, M., Fujita, J., Kang, S., & Kanakura, Y. (2011, January). Constitutively active FGFR3 with Lys650Glu mutation enhances bortezomib sensitivity in plasma cell malignancy. Anticancer Research.
115.
Zurück zum Zitat Walker, B. A., Murdoch, J. L., McKusick, V. A., Langer, L. O., & Beals, R. K. (1971). Hypochondroplasia. American journal of diseases of children (1960), 122(2), 95–104. Walker, B. A., Murdoch, J. L., McKusick, V. A., Langer, L. O., & Beals, R. K. (1971). Hypochondroplasia. American journal of diseases of children (1960), 122(2), 95–104.
116.
Zurück zum Zitat Bellus, G. A., McIntosh, I., Smith, E. A., Aylsworth, A. S., Kaitila, I., Horton, W. A., & Francomano, C. A. (1995). A recurrent mutation in the tyrosine kinase domain of fibroblast growth factor receptor 3 causes hypochondroplasia. Nature genetics, 10(3), 357–359.PubMedCrossRef Bellus, G. A., McIntosh, I., Smith, E. A., Aylsworth, A. S., Kaitila, I., Horton, W. A., & Francomano, C. A. (1995). A recurrent mutation in the tyrosine kinase domain of fibroblast growth factor receptor 3 causes hypochondroplasia. Nature genetics, 10(3), 357–359.PubMedCrossRef
117.
Zurück zum Zitat Bellus, G. A., Garber, A. T., Bryke, C. R., Weaver, C. A., Speiser, P. W., Webster, M. K., & Spector, E. B. (2000). FGFR3 Mutations K650N and K650Q Cause Hypochondroplasia. Genetics in Medicine, 2(1), 76–76.CrossRef Bellus, G. A., Garber, A. T., Bryke, C. R., Weaver, C. A., Speiser, P. W., Webster, M. K., & Spector, E. B. (2000). FGFR3 Mutations K650N and K650Q Cause Hypochondroplasia. Genetics in Medicine, 2(1), 76–76.CrossRef
118.
Zurück zum Zitat Heuertz, S., Le Merrer, M., Zabel, B., Wright, M., Legeai-Mallet, L., Cormier-Daire, V., & Bonaventure, J. (2006). Novel FGFR3 mutations creating cysteine residues in the extracellular domain of the receptor cause achondroplasia or severe forms of hypochondroplasia. European Journal of Human Genetics, 14(12), 1240–1247.PubMedCrossRef Heuertz, S., Le Merrer, M., Zabel, B., Wright, M., Legeai-Mallet, L., Cormier-Daire, V., & Bonaventure, J. (2006). Novel FGFR3 mutations creating cysteine residues in the extracellular domain of the receptor cause achondroplasia or severe forms of hypochondroplasia. European Journal of Human Genetics, 14(12), 1240–1247.PubMedCrossRef
119.
Zurück zum Zitat Abdel-Salam, G. M. H., Flores-Sarnat, L., El-Ruby, M. O., Parboosingh, J., Bridge, P., Eid, M. M., & Temtamy, S. A. (2011). Muenke syndrome with pigmentary disorder and probable hemimegalencephaly: An expansion of the phenotype. American journal of medical genetics Part A, 155A(1), 207–214.PubMedCrossRef Abdel-Salam, G. M. H., Flores-Sarnat, L., El-Ruby, M. O., Parboosingh, J., Bridge, P., Eid, M. M., & Temtamy, S. A. (2011). Muenke syndrome with pigmentary disorder and probable hemimegalencephaly: An expansion of the phenotype. American journal of medical genetics Part A, 155A(1), 207–214.PubMedCrossRef
121.
Zurück zum Zitat Shiang, C. Y., Qi, Y., Wang, B., Lazar, V., Wang, J., Fraser Symmans, W., & Pusztai, L. (2010). Amplification of fibroblast growth factor receptor-1 in breast cancer and the effects of brivanib alaninate. Breast Cancer Research and Treatment, 123(3), 747–755.PubMedCrossRef Shiang, C. Y., Qi, Y., Wang, B., Lazar, V., Wang, J., Fraser Symmans, W., & Pusztai, L. (2010). Amplification of fibroblast growth factor receptor-1 in breast cancer and the effects of brivanib alaninate. Breast Cancer Research and Treatment, 123(3), 747–755.PubMedCrossRef
122.
Zurück zum Zitat Brunello, E., Brunelli, M., Bogina, G., Caliò, A., Manfrin, E., Nottegar, A., & Bonetti, F. (2012). FGFR-1 amplification in metastatic lymph-nodal and haematogenous lobular breast carcinoma. Journal of experimental & clinical cancer research : CR, 31, 103.PubMedCentralCrossRef Brunello, E., Brunelli, M., Bogina, G., Caliò, A., Manfrin, E., Nottegar, A., & Bonetti, F. (2012). FGFR-1 amplification in metastatic lymph-nodal and haematogenous lobular breast carcinoma. Journal of experimental & clinical cancer research : CR, 31, 103.PubMedCentralCrossRef
123.
Zurück zum Zitat Elbauomy Elsheikh, S., Green, A. R., Lambros, M. B. K., Turner, N. C., Grainge, M. J., Powe, D., & Reis-Filho, J. S. (2007). FGFR1 amplification in breast carcinomas: a chromogenic in situ hybridisation analysis. Breast Cancer Research, 9(2), R23.PubMedCentralPubMedCrossRef Elbauomy Elsheikh, S., Green, A. R., Lambros, M. B. K., Turner, N. C., Grainge, M. J., Powe, D., & Reis-Filho, J. S. (2007). FGFR1 amplification in breast carcinomas: a chromogenic in situ hybridisation analysis. Breast Cancer Research, 9(2), R23.PubMedCentralPubMedCrossRef
125.
Zurück zum Zitat Freier, K., Schwaenen, C., Sticht, C., Flechtenmacher, C., Mühling, J., Hofele, C., & Joos, S. (2007). Recurrent FGFR1 amplification and high FGFR1 protein expression in oral squamous cell carcinoma (OSCC). Oral oncology, 43(1), 60–66.PubMedCrossRef Freier, K., Schwaenen, C., Sticht, C., Flechtenmacher, C., Mühling, J., Hofele, C., & Joos, S. (2007). Recurrent FGFR1 amplification and high FGFR1 protein expression in oral squamous cell carcinoma (OSCC). Oral oncology, 43(1), 60–66.PubMedCrossRef
126.
Zurück zum Zitat Goke, F., Franzen, A., Roopika, M., Schroeck, A., Kirsten, R., Boehm, D., & Perner, S. (2013). In 25th European Congress of Pathology. Portugal: Lisbon. Goke, F., Franzen, A., Roopika, M., Schroeck, A., Kirsten, R., Boehm, D., & Perner, S. (2013). In 25th European Congress of Pathology. Portugal: Lisbon.
127.
128.
Zurück zum Zitat Xie, L., Su, X., Zhang, L., Yin, X., Tang, L., Zhang, X., … Ji, Q. (2013). FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clinical cancer research : an official journal of the American Association for Cancer Research. Xie, L., Su, X., Zhang, L., Yin, X., Tang, L., Zhang, X., … Ji, Q. (2013). FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clinical cancer research : an official journal of the American Association for Cancer Research.
129.
Zurück zum Zitat Matsumoto, K., Arao, T., Hamaguchi, T., Shimada, Y., Kato, K., Oda, I., & Yamada, Y. (2012). FGFR2 gene amplification and clinicopathological features in gastric cancer. British Journal of Cancer, 106(4), 727–732.PubMedCentralPubMedCrossRef Matsumoto, K., Arao, T., Hamaguchi, T., Shimada, Y., Kato, K., Oda, I., & Yamada, Y. (2012). FGFR2 gene amplification and clinicopathological features in gastric cancer. British Journal of Cancer, 106(4), 727–732.PubMedCentralPubMedCrossRef
130.
Zurück zum Zitat Cancer Genome Atlas Research Network. (2011). Integrated genomic analyses of ovarian carcinoma. Nature, 474(7353), 609–615.CrossRef Cancer Genome Atlas Research Network. (2011). Integrated genomic analyses of ovarian carcinoma. Nature, 474(7353), 609–615.CrossRef
131.
Zurück zum Zitat Barretina, J., Taylor, B. S., Banerji, S., Ramos, A. H., Lagos-Quintana, M., Decarolis, P. L., & Singer, S. (2010). Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nature genetics, 42(8), 715–721.PubMedCentralPubMedCrossRef Barretina, J., Taylor, B. S., Banerji, S., Ramos, A. H., Lagos-Quintana, M., Decarolis, P. L., & Singer, S. (2010). Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nature genetics, 42(8), 715–721.PubMedCentralPubMedCrossRef
133.
Zurück zum Zitat Cancer Genome Atlas Research Network, Kandoth, C., Schultz, N., Cherniack, A. D., Akbani, R., Liu, Y., & Levine, D. A. (2013). Integrated genomic characterization of endometrial carcinoma. Nature, 497(7447), 67–73.CrossRef Cancer Genome Atlas Research Network, Kandoth, C., Schultz, N., Cherniack, A. D., Akbani, R., Liu, Y., & Levine, D. A. (2013). Integrated genomic characterization of endometrial carcinoma. Nature, 497(7447), 67–73.CrossRef
135.
Zurück zum Zitat Williams, S. V., Hurst, C. D., & Knowles, M. A. (2013). Oncogenic FGFR3 gene fusions in bladder cancer. Human molecular genetics, 22(4), 795–803.PubMedCentralPubMedCrossRef Williams, S. V., Hurst, C. D., & Knowles, M. A. (2013). Oncogenic FGFR3 gene fusions in bladder cancer. Human molecular genetics, 22(4), 795–803.PubMedCentralPubMedCrossRef
137.
Zurück zum Zitat Taylor, J. G., Cheuk, A. T., Tsang, P. S., Chung, J.-Y., Song, Y. K., Desai, K., & Khan, J. (2009). Identification of FGFR4-activating mutations in human rhabdomyosarcomas that promote metastasis in xenotransplanted models. The Journal of Clinical Investigation, 119(11), 3395–3407. doi:10.1172/JCI39703.PubMed Taylor, J. G., Cheuk, A. T., Tsang, P. S., Chung, J.-Y., Song, Y. K., Desai, K., & Khan, J. (2009). Identification of FGFR4-activating mutations in human rhabdomyosarcomas that promote metastasis in xenotransplanted models. The Journal of Clinical Investigation, 119(11), 3395–3407. doi:10.​1172/​JCI39703.PubMed
138.
Zurück zum Zitat Turkington, R. C., Longley, D. B., Allen, W. L., Stevenson, L., McLaughlin, K., Dunne, P. D., & Johnston, P. G. (2014). Fibroblast growth factor receptor 4 (FGFR4): a targetable regulator of drug resistance in colorectal cancer. Cell Death & Disease, 5(2), e1046. doi:10.1038/cddis.2014.10.CrossRef Turkington, R. C., Longley, D. B., Allen, W. L., Stevenson, L., McLaughlin, K., Dunne, P. D., & Johnston, P. G. (2014). Fibroblast growth factor receptor 4 (FGFR4): a targetable regulator of drug resistance in colorectal cancer. Cell Death & Disease, 5(2), e1046. doi:10.​1038/​cddis.​2014.​10.CrossRef
139.
Zurück zum Zitat Persson, F., Winnes, M., Andrén, Y., Wedell, B., Dahlenfors, R., Asp, J., & Stenman, G. (2008). High-resolution array CGH analysis of salivary gland tumors reveals fusion and amplification of the FGFR1 and PLAG1 genes in ring chromosomes. Oncogene, 27(21), 3072–3080. doi:10.1038/sj.onc.1210961.PubMedCrossRef Persson, F., Winnes, M., Andrén, Y., Wedell, B., Dahlenfors, R., Asp, J., & Stenman, G. (2008). High-resolution array CGH analysis of salivary gland tumors reveals fusion and amplification of the FGFR1 and PLAG1 genes in ring chromosomes. Oncogene, 27(21), 3072–3080. doi:10.​1038/​sj.​onc.​1210961.PubMedCrossRef
140.
Zurück zum Zitat Arai, Y., Totoki, Y., Hosoda, F., Shirota, T., Hama, N., Nakamura, H., & Shibata, T. (2014). Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology, 59(4), 1427–1434. doi:10.1002/hep.26890.PubMedCrossRef Arai, Y., Totoki, Y., Hosoda, F., Shirota, T., Hama, N., Nakamura, H., & Shibata, T. (2014). Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology, 59(4), 1427–1434. doi:10.​1002/​hep.​26890.PubMedCrossRef
141.
Zurück zum Zitat Wu, Y.-M., Su, F., Kalyana-Sundaram, S., Khazanov, N., Ateeq, B., Cao, X., … Chinnaiyan, A. M. (2013). Identification of Targetable FGFR Gene Fusions in Diverse Cancers. Cancer discovery. Wu, Y.-M., Su, F., Kalyana-Sundaram, S., Khazanov, N., Ateeq, B., Cao, X., … Chinnaiyan, A. M. (2013). Identification of Targetable FGFR Gene Fusions in Diverse Cancers. Cancer discovery.
142.
143.
Zurück zum Zitat Shah, S. P., Roth, A., Goya, R., Oloumi, A., Ha, G., Zhao, Y., & Aparicio, S. (2012). The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature, 486(7403), 395–399.PubMed Shah, S. P., Roth, A., Goya, R., Oloumi, A., Ha, G., Zhao, Y., & Aparicio, S. (2012). The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature, 486(7403), 395–399.PubMed
144.
Zurück zum Zitat Banerji, S., Cibulskis, K., Rangel-Escareno, C., Brown, K. K., Carter, S. L., Frederick, A. M., & Meyerson, M. (2012). Sequence analysis of mutations and translocations across breast cancer subtypes. Nature, 486(7403), 405–409.PubMedCentralPubMedCrossRef Banerji, S., Cibulskis, K., Rangel-Escareno, C., Brown, K. K., Carter, S. L., Frederick, A. M., & Meyerson, M. (2012). Sequence analysis of mutations and translocations across breast cancer subtypes. Nature, 486(7403), 405–409.PubMedCentralPubMedCrossRef
145.
Zurück zum Zitat Stephens, P. J., Tarpey, P. S., Davies, H., Van Loo, P., Greenman, C., Wedge, D. C., & Stratton, M. R. (2012). The landscape of cancer genes and mutational processes in breast cancer. Nature, 486(7403), 400–404.PubMedCentralPubMed Stephens, P. J., Tarpey, P. S., Davies, H., Van Loo, P., Greenman, C., Wedge, D. C., & Stratton, M. R. (2012). The landscape of cancer genes and mutational processes in breast cancer. Nature, 486(7403), 400–404.PubMedCentralPubMed
147.
Zurück zum Zitat Mountzios, G., Rampias, T., & Psyrri, A. (2014). The mutational spectrum of squamous-cell carcinoma of the head and neck: targetable genetic events and clinical impact. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. doi:10.1093/annonc/mdu143. Mountzios, G., Rampias, T., & Psyrri, A. (2014). The mutational spectrum of squamous-cell carcinoma of the head and neck: targetable genetic events and clinical impact. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. doi:10.​1093/​annonc/​mdu143.
149.
Zurück zum Zitat Imielinski, M., Berger, A. H., Hammerman, P. S., Hernandez, B., Pugh, T. J., Hodis, E., & Meyerson, M. (2012). Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell, 150(6), 1107–1120.PubMedCentralPubMedCrossRef Imielinski, M., Berger, A. H., Hammerman, P. S., Hernandez, B., Pugh, T. J., Hodis, E., & Meyerson, M. (2012). Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell, 150(6), 1107–1120.PubMedCentralPubMedCrossRef
150.
Zurück zum Zitat Ding, L., Getz, G., Wheeler, D. A., Mardis, E. R., McLellan, M. D., Cibulskis, K., & Wilson, R. K. (2008). Somatic mutations affect key pathways in lung adenocarcinoma. Nature, 455(7216), 1069–1075.PubMedCentralPubMedCrossRef Ding, L., Getz, G., Wheeler, D. A., Mardis, E. R., McLellan, M. D., Cibulskis, K., & Wilson, R. K. (2008). Somatic mutations affect key pathways in lung adenocarcinoma. Nature, 455(7216), 1069–1075.PubMedCentralPubMedCrossRef
151.
Zurück zum Zitat Taylor, B. S., Schultz, N., Hieronymus, H., Gopalan, A., Xiao, Y., Carver, B. S., & Gerald, W. L. (2010). Integrative genomic profiling of human prostate cancer. Cancer Cell, 18(1), 11–22.PubMedCentralPubMedCrossRef Taylor, B. S., Schultz, N., Hieronymus, H., Gopalan, A., Xiao, Y., Carver, B. S., & Gerald, W. L. (2010). Integrative genomic profiling of human prostate cancer. Cancer Cell, 18(1), 11–22.PubMedCentralPubMedCrossRef
153.
154.
Zurück zum Zitat McLendon, R., Friedman, A., Bigner, D., Van Meir, E. G., Brat, D. J., Mastrogianakis, M. G., & Thomson, E. (2008). Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature, 455(7216), 1061–1068.CrossRef McLendon, R., Friedman, A., Bigner, D., Van Meir, E. G., Brat, D. J., Mastrogianakis, M. G., & Thomson, E. (2008). Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature, 455(7216), 1061–1068.CrossRef
155.
Zurück zum Zitat Ho, A. S., Kannan, K., Roy, D. M., Morris, L. G. T., Ganly, I., Katabi, N., & Chan, T. A. (2013). The mutational landscape of adenoid cystic carcinoma. Nature genetics, 45(7), 791–798.PubMedCrossRef Ho, A. S., Kannan, K., Roy, D. M., Morris, L. G. T., Ganly, I., Katabi, N., & Chan, T. A. (2013). The mutational landscape of adenoid cystic carcinoma. Nature genetics, 45(7), 791–798.PubMedCrossRef
157.
Zurück zum Zitat Cancer Genome Atlas Research Network. (2013). Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. The New England journal of medicine, 368(22), 2059–2074.CrossRef Cancer Genome Atlas Research Network. (2013). Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. The New England journal of medicine, 368(22), 2059–2074.CrossRef
158.
Zurück zum Zitat Seshagiri, S., Stawiski, E. W., Durinck, S., Modrusan, Z., Storm, E. E., Conboy, C. B., & de Sauvage, F. J. (2012). Recurrent R-spondin fusions in colon cancer. Nature, 488(7413), 660–664.PubMedCentralPubMedCrossRef Seshagiri, S., Stawiski, E. W., Durinck, S., Modrusan, Z., Storm, E. E., Conboy, C. B., & de Sauvage, F. J. (2012). Recurrent R-spondin fusions in colon cancer. Nature, 488(7413), 660–664.PubMedCentralPubMedCrossRef
160.
Zurück zum Zitat Smyth, E. C., Turner, N. C., Popat, S., Morgan, S., Owen, K., Gillbanks, A., & Cunningham, D. (2013). FGFR: Proof-of-concept study of AZD4547 in patients with FGFR1 or FGFR2 amplified tumours. ASCO Meeting Abstracts, 31(15_suppl), TPS2626. Smyth, E. C., Turner, N. C., Popat, S., Morgan, S., Owen, K., Gillbanks, A., & Cunningham, D. (2013). FGFR: Proof-of-concept study of AZD4547 in patients with FGFR1 or FGFR2 amplified tumours. ASCO Meeting Abstracts, 31(15_suppl), TPS2626.
161.
Zurück zum Zitat Zhang, J., Zhang, L., Su, X., Li, M., Xie, L., Malchers, F., & Gavine, P. R. (2012). Translating the therapeutic potential of AZD4547 in FGFR1-amplified non-small cell lung cancer through the use of patient-derived tumor xenograft models. Clinical cancer research : an official journal of the American Association for Cancer Research, 18(24), 6658–6667.CrossRef Zhang, J., Zhang, L., Su, X., Li, M., Xie, L., Malchers, F., & Gavine, P. R. (2012). Translating the therapeutic potential of AZD4547 in FGFR1-amplified non-small cell lung cancer through the use of patient-derived tumor xenograft models. Clinical cancer research : an official journal of the American Association for Cancer Research, 18(24), 6658–6667.CrossRef
162.
Zurück zum Zitat Johnson, P. J., Qin, S., Park, J.-W., Poon, R. T. P., Raoul, J.-L., Philip, P. A., & Cheng, A.-L. (2013). Brivanib Versus Sorafenib As First-Line Therapy in Patients With Unresectable, Advanced Hepatocellular Carcinoma: Results From the Randomized Phase III BRISK-FL Study. Journal of Clinical Oncology, JCO, 2012(48), 4410. doi:10.1200/JCO.2012.48.4410. Johnson, P. J., Qin, S., Park, J.-W., Poon, R. T. P., Raoul, J.-L., Philip, P. A., & Cheng, A.-L. (2013). Brivanib Versus Sorafenib As First-Line Therapy in Patients With Unresectable, Advanced Hepatocellular Carcinoma: Results From the Randomized Phase III BRISK-FL Study. Journal of Clinical Oncology, JCO, 2012(48), 4410. doi:10.​1200/​JCO.​2012.​48.​4410.
163.
Zurück zum Zitat Huynh, H., Ngo, V. C., Fargnoli, J., Ayers, M., Soo, K. C., Koong, H. N., & Tran, E. (2008). Brivanib Alaninate, a Dual Inhibitor of Vascular Endothelial Growth Factor Receptor and Fibroblast Growth Factor Receptor Tyrosine Kinases, Induces Growth Inhibition in Mouse Models of Human Hepatocellular Carcinoma. Clinical Cancer Research, 14(19), 6146–6153.PubMedCrossRef Huynh, H., Ngo, V. C., Fargnoli, J., Ayers, M., Soo, K. C., Koong, H. N., & Tran, E. (2008). Brivanib Alaninate, a Dual Inhibitor of Vascular Endothelial Growth Factor Receptor and Fibroblast Growth Factor Receptor Tyrosine Kinases, Induces Growth Inhibition in Mouse Models of Human Hepatocellular Carcinoma. Clinical Cancer Research, 14(19), 6146–6153.PubMedCrossRef
164.
Zurück zum Zitat Siu, L. L., Shapiro, J. D., Jonker, D. J., Karapetis, C. S., Zalcberg, J. R., Simes, J., … NCIC Clinical Trials Group and AGITG. (2012). Phase III randomized trial of cetuximab (CET) plus either brivanib alaninate (BRIV) or placebo in patients (pts) with metastatic (MET) chemotherapy refractory K-RAS wild-type (WT) colorectal carcinoma (CRC): The NCIC Clinical Trials Group and AGITG CO.20 trial. ASCO Meeting Abstracts, 30(4_suppl), 386. Siu, L. L., Shapiro, J. D., Jonker, D. J., Karapetis, C. S., Zalcberg, J. R., Simes, J., … NCIC Clinical Trials Group and AGITG. (2012). Phase III randomized trial of cetuximab (CET) plus either brivanib alaninate (BRIV) or placebo in patients (pts) with metastatic (MET) chemotherapy refractory K-RAS wild-type (WT) colorectal carcinoma (CRC): The NCIC Clinical Trials Group and AGITG CO.20 trial. ASCO Meeting Abstracts, 30(4_suppl), 386.
165.
Zurück zum Zitat Meulenbeld, H. J., Bleuse, J. P., Vinci, E. M., Raymond, E., Vitali, G., Santoro, A., & de Wit, R. (2013). Randomized phase II study of danusertib in patients with metastatic castration-resistant prostate cancer after docetaxel failure. BJU international, 111(1), 44–52. doi:10.1111/j.1464-410X.2012.11404.x.PubMedCrossRef Meulenbeld, H. J., Bleuse, J. P., Vinci, E. M., Raymond, E., Vitali, G., Santoro, A., & de Wit, R. (2013). Randomized phase II study of danusertib in patients with metastatic castration-resistant prostate cancer after docetaxel failure. BJU international, 111(1), 44–52. doi:10.​1111/​j.​1464-410X.​2012.​11404.​x.PubMedCrossRef
166.
Zurück zum Zitat Cohen, R. B., Jones, S. F., Aggarwal, C., von Mehren, M., Cheng, J., Spigel, D. R., & Burris, H. A. (2009). A phase I dose-escalation study of danusertib (PHA-739358) administered as a 24-hour infusion with and without granulocyte colony-stimulating factor in a 14-day cycle in patients with advanced solid tumors. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research, 15(21), 6694–6701. doi:10.1158/1078-0432.CCR-09-1445.CrossRef Cohen, R. B., Jones, S. F., Aggarwal, C., von Mehren, M., Cheng, J., Spigel, D. R., & Burris, H. A. (2009). A phase I dose-escalation study of danusertib (PHA-739358) administered as a 24-hour infusion with and without granulocyte colony-stimulating factor in a 14-day cycle in patients with advanced solid tumors. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research, 15(21), 6694–6701. doi:10.​1158/​1078-0432.​CCR-09-1445.CrossRef
167.
Zurück zum Zitat Motzer, R. J., Porta, C., Vogelzang, N. J., Sternberg, C. N., Szczylik, C., Zolnierek, J., & Escudier, B. (2014). Dovitinib versus sorafenib for third-line targeted treatment of patients with metastatic renal cell carcinoma: an open-label, randomised phase 3 trial. The Lancet. Oncology, 15(3), 286–296. doi:10.1016/S1470-2045(14)70030-0.PubMedCrossRef Motzer, R. J., Porta, C., Vogelzang, N. J., Sternberg, C. N., Szczylik, C., Zolnierek, J., & Escudier, B. (2014). Dovitinib versus sorafenib for third-line targeted treatment of patients with metastatic renal cell carcinoma: an open-label, randomised phase 3 trial. The Lancet. Oncology, 15(3), 286–296. doi:10.​1016/​S1470-2045(14)70030-0.PubMedCrossRef
168.
Zurück zum Zitat Motzer, R. J., Porta, C., Bjarnason, G. A., Szcylik, C., Rha, S. Y., Esteban, E., … Escudier, B. J. (2012). Phase III trial of dovitinib (TKI258) versus sorafenib in patients with metastatic renal cell carcinoma after failure of anti-angiogenic (VEGF-targeted and mTOR inhibitor) therapies. ASCO Meeting Abstracts, 30(15_suppl), TPS4683. Motzer, R. J., Porta, C., Bjarnason, G. A., Szcylik, C., Rha, S. Y., Esteban, E., … Escudier, B. J. (2012). Phase III trial of dovitinib (TKI258) versus sorafenib in patients with metastatic renal cell carcinoma after failure of anti-angiogenic (VEGF-targeted and mTOR inhibitor) therapies. ASCO Meeting Abstracts, 30(15_suppl), TPS4683.
169.
Zurück zum Zitat Angevin, E., Lin, C., Pande, A. U., & Lopez, J. A. (2010). A phase I/II study of dovitinib (TKI258), a FGFR and VEGFR inhibitor, in patients (pts) with advanced or metastatic renal cell cancer: phase I results. ASCO Meeting \ldots. Angevin, E., Lin, C., Pande, A. U., & Lopez, J. A. (2010). A phase I/II study of dovitinib (TKI258), a FGFR and VEGFR inhibitor, in patients (pts) with advanced or metastatic renal cell cancer: phase I results. ASCO Meeting \ldots.
170.
Zurück zum Zitat Dienstmann, R., Bahleda, R., Adamo, B., Rodon, J., Varga, A., Gazzah, A., & Soria, J.-C. (2014). Abstract CT325: First in human study of JNJ-42756493, a potent pan fibroblast growth factor receptor (FGFR) inhibitor in patients with advanced solid tumors. Cancer Research, 74(19 Supplement), CT325–CT325. doi:10.1158/1538-7445.AM2014-CT325.CrossRef Dienstmann, R., Bahleda, R., Adamo, B., Rodon, J., Varga, A., Gazzah, A., & Soria, J.-C. (2014). Abstract CT325: First in human study of JNJ-42756493, a potent pan fibroblast growth factor receptor (FGFR) inhibitor in patients with advanced solid tumors. Cancer Research, 74(19 Supplement), CT325–CT325. doi:10.​1158/​1538-7445.​AM2014-CT325.CrossRef
171.
Zurück zum Zitat Schlumberger, M., Tahara, M., Wirth, L. J., Robinson, B., Brose, M. S., Elisei, R., … Sherman, S. I. (2014). A phase 3, multicenter, double-blind, placebo-controlled trial of lenvatinib (E7080) in patients with 131I-refractory differentiated thyroid cancer (SELECT) [abstract]. In Journal of clinical oncology : official journal of the American Society of Clinical Oncology (p. LBA6008). Schlumberger, M., Tahara, M., Wirth, L. J., Robinson, B., Brose, M. S., Elisei, R., … Sherman, S. I. (2014). A phase 3, multicenter, double-blind, placebo-controlled trial of lenvatinib (E7080) in patients with 131I-refractory differentiated thyroid cancer (SELECT) [abstract]. In Journal of clinical oncology : official journal of the American Society of Clinical Oncology (p. LBA6008).
172.
Zurück zum Zitat Tohyama, O., Matsui, J., Kodama, K., Hata-Sugi, N., Kimura, T., Okamoto, K., & Funahashi, Y. (2014). Antitumor Activity of Lenvatinib (E7080): An Angiogenesis Inhibitor That Targets Multiple Receptor Tyrosine Kinases in Preclinical Human Thyroid Cancer Models. Journal of Thyroid Research, 2014, e638747. doi:10.1155/2014/638747.CrossRef Tohyama, O., Matsui, J., Kodama, K., Hata-Sugi, N., Kimura, T., Okamoto, K., & Funahashi, Y. (2014). Antitumor Activity of Lenvatinib (E7080): An Angiogenesis Inhibitor That Targets Multiple Receptor Tyrosine Kinases in Preclinical Human Thyroid Cancer Models. Journal of Thyroid Research, 2014, e638747. doi:10.​1155/​2014/​638747.CrossRef
173.
Zurück zum Zitat Andre, F., Daly, F., Azim, H. A., Agrapart, V., Goulioti, T., Pinto, A. C. D. C. P., … Cortes, J. (2014). FINESSE: An open, three-cohort, phase II trial testing oral administration of lucitanib in patients with FGFR1-amplified or nonamplified estrogen receptor-positive metastatic breast cancer. Journal of Clinical Oncology, 32:5s(suppl; abstr TPS1134). Retrieved from http://meetinglibrary.asco.org/content/126911-144. Andre, F., Daly, F., Azim, H. A., Agrapart, V., Goulioti, T., Pinto, A. C. D. C. P., … Cortes, J. (2014). FINESSE: An open, three-cohort, phase II trial testing oral administration of lucitanib in patients with FGFR1-amplified or nonamplified estrogen receptor-positive metastatic breast cancer. Journal of Clinical Oncology, 32:5s(suppl; abstr TPS1134). Retrieved from http://​meetinglibrary.​asco.​org/​content/​126911-144.
174.
Zurück zum Zitat Bello, E., Taraboletti, G., Colella, G., Zucchetti, M., Forestieri, D., Licandro, S. A., & Damia, G. (2013). The Tyrosine Kinase Inhibitor E-3810 Combined with Paclitaxel Inhibits the Growth of Advanced-Stage Triple-Negative Breast Cancer Xenografts. Molecular Cancer Therapeutics, 12(2), 131–140.PubMedCrossRef Bello, E., Taraboletti, G., Colella, G., Zucchetti, M., Forestieri, D., Licandro, S. A., & Damia, G. (2013). The Tyrosine Kinase Inhibitor E-3810 Combined with Paclitaxel Inhibits the Growth of Advanced-Stage Triple-Negative Breast Cancer Xenografts. Molecular Cancer Therapeutics, 12(2), 131–140.PubMedCrossRef
175.
Zurück zum Zitat Bello, E., Colella, G., Scarlato, V., Oliva, P., Berndt, A., Valbusa, G., & Camboni, G. (2011). E-3810 is a potent dual inhibitor of VEGFR and FGFR that exerts antitumor activity in multiple preclinical models. Cancer Research, 71(4), 1396–1405.PubMedCrossRef Bello, E., Colella, G., Scarlato, V., Oliva, P., Berndt, A., Valbusa, G., & Camboni, G. (2011). E-3810 is a potent dual inhibitor of VEGFR and FGFR that exerts antitumor activity in multiple preclinical models. Cancer Research, 71(4), 1396–1405.PubMedCrossRef
176.
Zurück zum Zitat Carter, E. P., Fearon, A. E., & Grose, R. P. (n.d.). Careless talk costs lives: fibroblast growth factor receptor signalling and the consequences of pathway malfunction. Trends in Cell Biology. doi:10.1016/j.tcb.2014.11.003. Carter, E. P., Fearon, A. E., & Grose, R. P. (n.d.). Careless talk costs lives: fibroblast growth factor receptor signalling and the consequences of pathway malfunction. Trends in Cell Biology. doi:10.​1016/​j.​tcb.​2014.​11.​003.
177.
Zurück zum Zitat Pan, B.-S., Chan, G. K. Y., Chenard, M., Chi, A., Davis, L. J., Deshmukh, S. V., & Dinsmore, C. J. (2010). MK-2461, a novel multitargeted kinase inhibitor, preferentially inhibits the activated c-Met receptor. Cancer Research, 70(4), 1524–1533.PubMedCrossRef Pan, B.-S., Chan, G. K. Y., Chenard, M., Chi, A., Davis, L. J., Deshmukh, S. V., & Dinsmore, C. J. (2010). MK-2461, a novel multitargeted kinase inhibitor, preferentially inhibits the activated c-Met receptor. Cancer Research, 70(4), 1524–1533.PubMedCrossRef
178.
Zurück zum Zitat Hanna, N. H., Kaiser, R., Sullivan, R. N., Aren, O. R., Ahn, M.-J., Tiangco, B., … Kim, J. H. (2013). Lume-lung 2: A multicenter, randomized, double-blind, phase III study of nintedanib plus pemetrexed versus placebo plus pemetrexed in patients with advanced nonsquamous non-small cell lung cancer (NSCLC) after failure of first-line chemotherapy. ASCO Meeting Abstracts, 31(15_suppl), 8034. Hanna, N. H., Kaiser, R., Sullivan, R. N., Aren, O. R., Ahn, M.-J., Tiangco, B., … Kim, J. H. (2013). Lume-lung 2: A multicenter, randomized, double-blind, phase III study of nintedanib plus pemetrexed versus placebo plus pemetrexed in patients with advanced nonsquamous non-small cell lung cancer (NSCLC) after failure of first-line chemotherapy. ASCO Meeting Abstracts, 31(15_suppl), 8034.
179.
Zurück zum Zitat Richeldi, L., du Bois, R. M., Raghu, G., Azuma, A., Brown, K. K., Costabel, U., & Collard, H. R. (2014). Efficacy and Safety of Nintedanib in Idiopathic Pulmonary Fibrosis. New England Journal of Medicine, 370(22), 2071–2082. doi:10.1056/NEJMoa1402584.PubMedCrossRef Richeldi, L., du Bois, R. M., Raghu, G., Azuma, A., Brown, K. K., Costabel, U., & Collard, H. R. (2014). Efficacy and Safety of Nintedanib in Idiopathic Pulmonary Fibrosis. New England Journal of Medicine, 370(22), 2071–2082. doi:10.​1056/​NEJMoa1402584.PubMedCrossRef
180.
Zurück zum Zitat Kristensen, G., Harter, P., Trédan, O., Sailer, M. O., Bamias, A., Colombo, N., … Bois, A. D. (2014). Independent review of AGO-OVAR 12, a GCIG/ENGOT-Intergroup phase III trial of nintedanib (N) in first-line therapy for ovarian cancer (OC). Journal of Clinical Oncology, 32:5s(suppl; abstr 5556^). Retrieved from http://meetinglibrary.asco.org/content/130341-144. Kristensen, G., Harter, P., Trédan, O., Sailer, M. O., Bamias, A., Colombo, N., … Bois, A. D. (2014). Independent review of AGO-OVAR 12, a GCIG/ENGOT-Intergroup phase III trial of nintedanib (N) in first-line therapy for ovarian cancer (OC). Journal of Clinical Oncology, 32:5s(suppl; abstr 5556^). Retrieved from http://​meetinglibrary.​asco.​org/​content/​130341-144.
181.
Zurück zum Zitat Guagnano, V., Furet, P., Spanka, C., Bordas, V., Le Douget, M., Stamm, C., & Graus Porta, D. (2011). Discovery of 3-(2,6-dichloro-3,5-dimethoxy-phenyl)-1-{6-[4-(4-ethyl-piperazin-1-yl)-phenylamino]-pyrimidin-4-yl}-1-methyl-urea (NVP-BGJ398), a potent and selective inhibitor of the fibroblast growth factor receptor family of receptor tyrosine kinase. Journal of Medicinal Chemistry, 54(20), 7066–7083. doi:10.1021/jm2006222.PubMedCrossRef Guagnano, V., Furet, P., Spanka, C., Bordas, V., Le Douget, M., Stamm, C., & Graus Porta, D. (2011). Discovery of 3-(2,6-dichloro-3,5-dimethoxy-phenyl)-1-{6-[4-(4-ethyl-piperazin-1-yl)-phenylamino]-pyrimidin-4-yl}-1-methyl-urea (NVP-BGJ398), a potent and selective inhibitor of the fibroblast growth factor receptor family of receptor tyrosine kinase. Journal of Medicinal Chemistry, 54(20), 7066–7083. doi:10.​1021/​jm2006222.PubMedCrossRef
182.
Zurück zum Zitat Guagnano, V., Kauffmann, A., Wöhrle, S., Stamm, C., Ito, M., Barys, L., & Graus-Porta, D. (2012). FGFR genetic alterations predict for sensitivity to NVP-BGJ398, a selective pan-FGFR inhibitor. Cancer Discovery, 2(12), 1118–1133. doi:10.1158/2159-8290.CD-12-0210.PubMedCrossRef Guagnano, V., Kauffmann, A., Wöhrle, S., Stamm, C., Ito, M., Barys, L., & Graus-Porta, D. (2012). FGFR genetic alterations predict for sensitivity to NVP-BGJ398, a selective pan-FGFR inhibitor. Cancer Discovery, 2(12), 1118–1133. doi:10.​1158/​2159-8290.​CD-12-0210.PubMedCrossRef
183.
Zurück zum Zitat Konecny, G. E., Kolarova, T., O’Brien, N. A., Winterhoff, B., Yang, G., Qi, J., … Slamon, D. J. (2013). Activity of the Fibroblast Growth Factor Receptor Inhibitors Dovitinib (TKI258) and NVP-BGJ398 in Human Endometrial Cancer Cells. Molecular Cancer Therapeutics. Konecny, G. E., Kolarova, T., O’Brien, N. A., Winterhoff, B., Yang, G., Qi, J., … Slamon, D. J. (2013). Activity of the Fibroblast Growth Factor Receptor Inhibitors Dovitinib (TKI258) and NVP-BGJ398 in Human Endometrial Cancer Cells. Molecular Cancer Therapeutics.
184.
Zurück zum Zitat Motzer, R. J., Hutson, T. E., McCann, L., Deen, K., & Choueiri, T. K. (2014). Overall survival in renal-cell carcinoma with pazopanib versus sunitinib. The New England Journal of Medicine, 370(18), 1769–1770. doi:10.1056/NEJMc1400731.PubMedCrossRef Motzer, R. J., Hutson, T. E., McCann, L., Deen, K., & Choueiri, T. K. (2014). Overall survival in renal-cell carcinoma with pazopanib versus sunitinib. The New England Journal of Medicine, 370(18), 1769–1770. doi:10.​1056/​NEJMc1400731.PubMedCrossRef
185.
Zurück zum Zitat Kasper, B., Sleijfer, S., Litière, S., Marreaud, S., Verweij, J., Hodge, R. A., & van der Graaf, W. T. A. (2014). Long-term responders and survivors on pazopanib for advanced soft tissue sarcomas: subanalysis of two European Organisation for Research and Treatment of Cancer (EORTC) clinical trials 62043 and 62072. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO, 25(3), 719–724. doi:10.1093/annonc/mdt586.CrossRef Kasper, B., Sleijfer, S., Litière, S., Marreaud, S., Verweij, J., Hodge, R. A., & van der Graaf, W. T. A. (2014). Long-term responders and survivors on pazopanib for advanced soft tissue sarcomas: subanalysis of two European Organisation for Research and Treatment of Cancer (EORTC) clinical trials 62043 and 62072. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO, 25(3), 719–724. doi:10.​1093/​annonc/​mdt586.CrossRef
186.
Zurück zum Zitat O’Hare, T., Shakespeare, W. C., Zhu, X., Eide, C. A., Rivera, V. M., Wang, F., & Clackson, T. (2009). AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell, 16(5), 401–412.PubMedCentralPubMedCrossRef O’Hare, T., Shakespeare, W. C., Zhu, X., Eide, C. A., Rivera, V. M., Wang, F., & Clackson, T. (2009). AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell, 16(5), 401–412.PubMedCentralPubMedCrossRef
187.
Zurück zum Zitat Cortes, J. E., Kim, D.-W., Pinilla-Ibarz, J., le Coutre, P., Paquette, R., Chuah, C., & Investigators, P. A. C. E. (2013). A phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias. The New England Journal of Medicine, 369(19), 1783–1796. doi:10.1056/NEJMoa1306494.PubMedCrossRef Cortes, J. E., Kim, D.-W., Pinilla-Ibarz, J., le Coutre, P., Paquette, R., Chuah, C., & Investigators, P. A. C. E. (2013). A phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias. The New England Journal of Medicine, 369(19), 1783–1796. doi:10.​1056/​NEJMoa1306494.PubMedCrossRef
188.
Zurück zum Zitat Demetri, G. D., Reichardt, P., Kang, Y.-K., Blay, J.-Y., Rutkowski, P., Gelderblom, H., & GRID study investigators. (2013). Efficacy and safety of regorafenib for advanced gastrointestinal stromal tumours after failure of imatinib and sunitinib (GRID): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet, 381(9863), 295–302. doi:10.1016/S0140-6736(12)61857-1.PubMedCrossRef Demetri, G. D., Reichardt, P., Kang, Y.-K., Blay, J.-Y., Rutkowski, P., Gelderblom, H., & GRID study investigators. (2013). Efficacy and safety of regorafenib for advanced gastrointestinal stromal tumours after failure of imatinib and sunitinib (GRID): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet, 381(9863), 295–302. doi:10.​1016/​S0140-6736(12)61857-1.PubMedCrossRef
189.
Zurück zum Zitat Grothey, A., Van Cutsem, E., Sobrero, A., Siena, S., Falcone, A., Ychou, M., & CORRECT Study Group. (2013). Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet, 381(9863), 303–312. doi:10.1016/S0140-6736(12)61900-X.PubMedCrossRef Grothey, A., Van Cutsem, E., Sobrero, A., Siena, S., Falcone, A., Ychou, M., & CORRECT Study Group. (2013). Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet, 381(9863), 303–312. doi:10.​1016/​S0140-6736(12)61900-X.PubMedCrossRef
190.
Zurück zum Zitat Ochiiwa, H., Fujita, H., Itoh, K., Sootome, H., Hashimoto, A., Fujioka, Y., & Utsugi, T. (2013). Abstract A270: TAS-120, a highly potent and selective irreversible FGFR inhibitor, is effective in tumors harboring various FGFR gene abnormalities. Molecular Cancer Therapeutics, 12(11 Supplement), A270–A270. doi:10.1158/1535-7163.TARG-13-A270.CrossRef Ochiiwa, H., Fujita, H., Itoh, K., Sootome, H., Hashimoto, A., Fujioka, Y., & Utsugi, T. (2013). Abstract A270: TAS-120, a highly potent and selective irreversible FGFR inhibitor, is effective in tumors harboring various FGFR gene abnormalities. Molecular Cancer Therapeutics, 12(11 Supplement), A270–A270. doi:10.​1158/​1535-7163.​TARG-13-A270.CrossRef
191.
Zurück zum Zitat Powers, C. J., McLeskey, S. W., & Wellstein, A. (2000). Fibroblast growth factors, their receptors and signaling. Endocrine-Related Cancer, 7(3), 165–197.PubMedCrossRef Powers, C. J., McLeskey, S. W., & Wellstein, A. (2000). Fibroblast growth factors, their receptors and signaling. Endocrine-Related Cancer, 7(3), 165–197.PubMedCrossRef
Metadaten
Titel
Fibroblast growth factor receptor signaling in hereditary and neoplastic disease: biologic and clinical implications
verfasst von
Teresa Helsten
Maria Schwaederle
Razelle Kurzrock
Publikationsdatum
01.09.2015
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 3/2015
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-015-9579-8

Weitere Artikel der Ausgabe 3/2015

Cancer and Metastasis Reviews 3/2015 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.