Small Molecule Discoidin Domain Receptor Kinase Inhibitors and

Biological Functions of DDRs and Medical Implications ..... X-ray structures of cocrystals of DDR1 with drugs 3 and 7 have been reported and demonstra...
0 downloads 0 Views 3MB Size
Perspective pubs.acs.org/jmc

Small Molecule Discoidin Domain Receptor Kinase Inhibitors and Potential Medical Applications Miniperspective Yupeng Li,†,‡,§ Xiaoyun Lu,†,§ Xiaomei Ren,*,† and Ke Ding*,† †

State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China ‡ University of Chinese Academy of Sciences, No. 19 Yuquan Road, Beijing 100049, China ABSTRACT: Discoidin domain receptors (DDRs) are members of the transmembrane receptor tyrosine kinase (RTK) superfamily which are distinguished from others by the presence of a discoidin motif in the extracellular domain and their utilization of collagens as internal ligands. Two types of DDRs, DDR1 and DDR2, have been identified with distinct expression profiles and ligand specificities. These DDRs play important roles in the regulation of fundamental cellular process, such as proliferation, survival, differentiation, adhesion, and matrix remodeling. They have also been closely linked to a number of human diseases, including various fibrotic disorders, atherosclerosis, and cancer. As a consequence, DDRs have been considered as novel potential molecular targets for drug discovery and increasing efforts are being devoted to the identification of new small molecule inhibitors targeting the receptors. In this review, we offer a contemporary overview on the discovery of DDRs inhibitors and their potential medical application for the treatment of cancer and inflammation related disorders.



INTRODUCTION Discoidin domain receptors (DDRs) are members of the transmembrane receptor tyrosine kinase (RTK) superfamily discovered in the early 1990s. 1 They are structurally distinguished from other RTKs by the presence of a discoidin motif in the extracellular domain. Two types of DDRs, DDR1 and DDR2, with distinct expression profiles and ligand specificities have been identified. DDR1 has five splice variants, DDR1a, DDR1b, DDR1c, DDR1d, and DDR1e, generated by alternative splicing or deletion of exons, while DDR2 has only one isoform. DDR1a, DDR1b, and DDR1c encode full-length enzymatically active receptors,1g,2 but DDR1d and DDR1e are predicted as kinase-deficient receptors because of their truncated or kinase deficient cytosolic fragment (Figure 1).3 Typical RTKs use peptide-like growth factors as ligands, but DDRs are activated by various types of triple-helical collagens which are the most abundant components of the extracellular matrix (ECM).4 Specifically, DDR1 can bind to essentially almost all types of collagens identified to date, while DDR2 shows a preference for type I, II and III fibrillar collagens, and nonfibrillar type X collagen.4,5 In addition to their ligand specificities, DDR1 and DDR2 also display distinct expression profiles. DDR1 is widely expressed in epithelial cells in lung, kidney, colon, and brain, whereas DDR2 is primarily expressed in mesenchymal cells including fibroblasts, myofibroblasts, smooth muscle cells, and chondrocytes in kidney, skin, lung, heart, and connective tissues.1 © XXXX American Chemical Society

Studies have demonstrated that both DDR1 and DDR2 are important for the regulation of fundamental cellular processes, such as proliferation, survival, differentiation, adhesion, and matrix remodeling.6 Dysregulation of the receptors is closely related to a number of human diseases, including fibrotic disorders (e.g., renal fibrosis and pulmonary fibrosis, etc.), atherosclerosis, and cancer.6 Thus, DDR1 and DDR2 have been considered as novel potential molecular targets for drug discovery, and development of small molecule DDR inhibitors may lead to new and attractive therapeutic strategies for the treatment of cancer or various inflammation-related disorders. This review provides an updated overview on the discovery of DDR inhibitors and their potential application to the treatment of cancer and inflammation-related disorders. Audiences are also encouraged to read the recent comprehensive reviews to get extensive knowledge on the biological background of DDRs.6



STRUCTURAL DETAILS OF DDRS Similar to the structural arrangement of classic RTKs, DDR1 and DDR2 consist of three fundamental elements: an extracellular domain, a transmembrane domain, and an intracellular domain with intrinsic enzymatic activity (Figure 1). Distinctively, DDRs contain, in the extracellular region, a Received: August 10, 2014

A

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

Figure 1. Domain arrangements of DDR1 and DDR2. DDR1a, DDR1b, DDR1c, and DDR2 transmit signals normally by encoding full-length enzymatic active receptors, while DDR1d and DDR1e are kinase-deficient receptors as a result of their truncated or inactive kinase domains.1g,2,3

share high sequence and morphological homology with that of Abl (UniProtKB, P00519) or c-Kit (UniProtKB, P10721).11 Several cocrystal structures of DDR1 kinase domain with small molecule inhibitors have been reported (Table 1) and show that DDR1 kinase domain contains loops or motifs similar to those in other RTKs, including glycine-rich loop, A-loop, DFG motif, and C-helix (Figure 3).12 To date, there has been no report of a crystal structure of DDR2 kinase domain complexed with a small molecule ligand, but homology modeling suggests that the DDR2 kinase domain may adopt a DDR1-like conformational arrangement.13

unique discoidin motif, the DS domain which exhibits high structural similarity to discoidin I protein that was originally identified as a galactose-binding lectin in the slime mode Dictyostelium discoideum.7 A discoidin-like domain (DS-like domain) and an extracellular juxtamembrane region (EJXM) are also located in the extracellular region. The structure of a cocrystal of a DDR2−collagen complex reveals an important binding trench that is circumscribed by four conserved protruding loops, L1, L2, L4, and L6, at the protein−protein interface of the DS domain and is able to interact directly with collagen (Figure 2).8 In addition to the binding epitope, there are other loops regulating collagen recognition. Replacement of five solvent-exposed residues adjacent to the binding trench of DDR2 with the corresponding DDR1 substitutions results in significant ligand-selectivity alternatives.5 The transmembrane domain (TM) of DDRs provides a linkage between their extracellular and intracellular domains. Several repeated leucine zipper motifs in this region are critical for the self-associated dimerization which is required for the phosphotransferase activity of DDR kinases.9 It is notable that the intrinsic DDR dimerization is performed without recognizing ligand, which is in contrast to the ligand-induced dimerization of most of other RTKs.9 The intracellular domain contains an unexpectedly large intracellular juxtamembrane region (IJXM) and a kinase domain with a short C-terminal tail. Following collagen binding, the tyrosine residues in the IJXM are phosphorylated to serve as docking sites to which they attract nearby adaptor proteins.10 As with many other kinases, the intrinsic enzymatic activity of DDRs is determined by their kinase domain (KD). The kinase domains of DDR1 (UniProtKB, Q08345) and DDR2 (UniProtKB, Q16832)



BIOLOGICAL FUNCTIONS OF DDRS AND MEDICAL IMPLICATIONS 1. Roles in Development. Both DDR1 and DDR2 have been demonstrated to be crucial for the regulation of developmental and physiological processes.6,16 Generally, DDR1 is important for the biogenesis of multiple organs, whereas DDR2 is one of the key players for skeletetogenesis. DDR1-null mice were characterized to be smaller stature than normal littermates and lactational defect in pregnant female animals, but further investigation is needed to confirm the results.17 Defects in kidney and inner ear architecture were also observed for mice with lack of DDR1.18 DDR1 knockout mice display severe auditory function decrease and progressive morphological alterations,18b while the animals only show a localized matrix overproduction in the glomerular basement and no chronic renal disease development was observed.18c In transgenic mice deleting collagen II, the lack of endochondrial bone or epiphyseal growth plate in long bones has been reported.19 In view of the fact that DDR2 selectively recognizes B

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

Figure 2. Structural features of DDRs (PDB code 2WUH). (A) Overview structure of a DDR2−collagen cocrystal complex. (B) Binding details of DDR2 with collagen at the interface of the DS domain. Residues in the first chain of collagen are indicated in orange (V20, NLE21, and O24). Residues in the middle chain of collagen are shown in orange with asterisks (O23*, O24*, and O27*), and residues in DDR2 DS domain are in black (W52, D69, R105, I112, E113, M117, S173, M174, and N175). (C) Sequence alignment of DDR1 with DDR2.5,8

collagen II for activation, it would appear that DDR2 is important for chondrocyte proliferation,19 and this is further supported by the developmental defects such as dwarfism and shortened long bones exhibited in DDR2 (−/−) knockout mice.20 Moreover, transgenic mice with DDR2 overexpression were reported to display increased body length.21 In addition, DDR2 deletion or mis-sense mutation was shown to be closely related to autosomal recessive growth disorders such as Smallie (Slie) and human spondylo-meta-epiphyseal dysplasia which features short limbs and abnormal calcifications (SMED-SL).22 2. Regulation of Fundamental Cellular Processes. DDRs are highly implicated in fundamental cellular processes, including cell proliferation, migration, adhesion, and matrix remodeling. For example, DDR1a is important for promotion of leukocyte migration in three-dimensional collagen lattices.23 Dominant negative DDR1 inhibits neurite outgrowth both in vitro and in vivo, indicating a unique function of DDR1 in axon formation during cerebellar cortex histogenesis.24 DDR2 overexpression contributes greatly to the MMP-2 mediated proliferation and invasion of hepatic stellate cells,25 while DDR1 deletion leads to impaired ability of adhesion and migration.26 It has been demonstrated that DDR2 is required for normal fibroblast spreading and migration independent of adhesion ligand and collagen activation,27 and it plays a local and essential role in the proliferation of chondrocytes.28 Studies also suggest that DDR2 function is essential to the membrane dynamics which control the mechanical attachment of fibroblasts to the 3D collagen matrices.29 3. Correlation with Human Diseases. Dysregulation of DDR1 and DDR2 have been linked to a number of human diseases, including fibrotic disorders (e.g., renal fibrosis,

Table 1. Reported Crystal Structures of DDR−Ligand Complexes PDB code

DDR−ligand complex

refs

4AG4 3ZOS 4BKJ 4CKR 2WUH 2Z4F

DDR1/Fab DDR1/ponatinib (7) DDR1/imatinib (3) DDR1/DDR1-IN-1 (18) DDR2/collagen solution structure of DDR2 DS domain

8b 12 12 14 8a 15

Figure 3. Structure overview of cocrystal of DDR1 with drug 7 (PDB code 3ZOS).12

C

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

Figure 4. Domain distribution of somatic mutations of mutated DDR1 (red) and DDR2 (blue) identified in samples of NSCLC, AML, and SMED patients. Circles indicate “gain-of-function” mutants. The loss-of-function mutants are labeled as triangles, while mutants with undefined functions are labeled as squares.47−51

pulmonary fibrosis, and liver fibrosis), atherosclerosis, arthritis, and various types of cancer.6 For example, DDR1 overexpression has been identified as a novel biomarker of epithelial ovarian cancer.30 Clinicopathological parameter analysis in nonsmall-cell lung carcinoma patients (NSCLC) reveals a significant correlation between DDR1 overexpression and lymph node metastasis (p = 0.001), and DDR1 could be a prognostic marker for NSCLC patients.31 A global phosphoproteomic survey of tyrosine kinase activity in lung cancer cells identified DDR1 as an additional new activated kinase implicated in the tumorigenesis of NSCLC.32 Other studies also have demonstrated that high expression levels and/or mutations of DDR1 are frequently detected in multiple cancer cell lines and primary tumor tissues from lung,31,32,47 breast,33 brain,34 ovary,30 head and neck,35 liver,36 pancreas,37 prostate,38 and others.39 Silencing DDR1 by siRNA has been shown to reduce metastatic activity in lung cancer models and enhance chemosensitivity to genotoxic drugs in breast cancer cells.40 High level of DDR2 has also been determined in NSCLC,31d,47 nasopharyngeal carcinomas,35 thyroid carcinomas,35,41 and Hodgkin’s lymphoma, etc.42 Multivariate analysis of data from human breast cancer patients shows that DDR2 is a favorable independent predictor of recurrence and outcome for primary breast cancers.43 DDR2 overexpression also contributes greatly to metastasis, invasion, and migration of prostate cancer, and head and neck squamous cell carcinoma.44 Further mode of action (MOA) studies suggest that DDR2 can facilitate breast cancer metastasis via stabilization of the SNAIL1 protein.45 Downregulation or a host deficiency of DDR2 was demonstrated to obviously suppress the metastasis and tumor angiogenesis of various human cancers.46 Furthermore, “gain-of-function” mutations of DDR1 and/or DDR2 have been identified in various types of cancer cells and primary human NSCLC cancer tissues (Figure 4).47 Somatic mutations of DDR1 in lung neoplasms and cancer cells such as G1486T and A496S may contribute significantly to the development of lung cancer. 47a High-throughput DNA sequence analysis has revealed several novel somatic mutations of DDR1 such as N502S, A533S, and A803V in acute myeloid leukemia (AML), but additional studies are required to define the roles that these mutants play in AML pathogenesis.48 Some other mutations, including W385C, F866Y, F824W for DDR1 and R105S, N456S for DDR2 have also been reported in various cancer cells.11,31d More significantly, a recent Sanger sequencing of 20 primary lung squamous cell carcinoma (SCC) samples demonstrated that DDR2 is associated with one of the most frequently mutated genes. The corresponding mutations

include L63V, I120M, D125Y in the discoidin domain, C580Y, I638F, T765P, G774E/V in the kinase domain, and the missense mutations L239R, G253C, G505S, etc. Cancer cells with L239R and I638F mutations are selectively sensitive to DDR2 siRNAs and to a nonselective DDR2 inhibitor dasatinib (2).47d Two SCC patients with DDR2S768R mutation exhibit significant shrinkage of tumor size after treatment with drug 2.49 However, a recent proteomic study controversially suggested that the L63V and G505S mutants as well as wild type DDR2 might play a tumor-suppressive role in cancer cells induced by fibrillar collagen, indicating the complex role of DDR2 in lung cancer.50 In addition, mutation of N-glycosylation sites such as N211Q in DDR1 and N213Q in DDR2 that dysregulate the cell-surface expression of DDRs in their high molecular mass form, which is a functional isoform causing tyrosine autophosphorylation upon collagen stimulation, has also been identified.51 In addition to their significant involvement in human cancer, DDR1 and DDR2 are also closely related to fibrosis, atherosclerosis, and other inflammatory disorders or matrix degradations. Activation of DDR1 and DDR2 in human smooth muscle cells (SMCs) is important for enhancement of the activation of MMP, thus regulating extracellular matrix (ECM) remodeling in obstructive diseases of lungs and blood vessels.52 DDR1 deletion has been reported to alleviate bleomycin-induced lung inflammation and pulmonary fibrosis by blocking p38 MAPK activation.53 Use of DDR1/COL4A3 double-knockout models suggests that the collagen−DDR1 interaction plays an important role in the progressive renal fibrosis associated with Alport syndrome.18b,54a Further investigations also demonstrated that genetic deletion and/or antisense inhibition of DDR1 expression prevents the development of renal inflammation and fibrosis, blunted proteinuria, and preserved renal structure in different renal disease models (i.e., hypertensive nephropathy,54b tubular interstitial54c and glomerular nephritis54d), suggesting that pharmacological blockade of DDR1 may be an attractive strategy for efficient treatment of chronic kidney disease, an incurable today pathology. Silencing DDR2 expression was also demonstrated to decrease alcohol-induced liver injury and fibrosis in an early stage alcoholic liver disease mouse model.55 In view of its tissue distribution in articular cartilage, DDR2 reduction was clearly proved to attenuate the degeneration of osteoarthritis in knee joints.56 Studies also demonstrate that collagen I evoked DDR2 activation strongly promotes the release of inflammatory cytokines and co-stimulatory molecules via the NF-κB and JNK pathways in human monocyte derived dendritic cells.57 Consequently, the small molecule DDR inhibitor LCB 03-0110 D

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

Figure 5. Strategies for the development of novel DDR inhibitors.

DDR1 and DDR2. More importantly, compounds 7rh (17)67 and 1814 were recently discovered to inhibit selectively the kinase functions of DDR1, providing important research probes to further validate DDRs as new potential targets for drug discovery.

(13) was reported to significantly promote the wound healing process with no hypertrophic scar formation.58 Most recently, DDR2 point mutations such as T713I, I726R, R752C, and E113K have been reported to be causative genetic defects for the autosomal recessive human growth disorder SMED-SL.22





NATURAL ANTIBIOTIC 1 The protein−protein recognition of a triple-helical collagen with the DS domains of DDRs triggers a cascade of DDR signal transduction. Disruption of the DDR−collagen interaction represents a promising strategy to selectively suppress the DDR signal cascade. Using a self-constructed model of sf9 cells expressing DDR2 on type I collagen-coated wells, Yang et al. screened a drug compound library with 1040 chemicals and successfully identified the natural antibiotic 1 as an antagonist of the DDR2−collagen interaction (Figure 6).59 This

DDR INHIBITORS Given their significant contributions to the establishment and progression of human cancer and inflammation related disorders, DDRs are considered to be legitimate molecular targets for the discovery of new drugs. A variety of strategies have been adopted to develop inhibitors targeting the new class of kinases (Figure 5). For instance, the natural antibiotic actinomycin D (1) has been shown to effectively disrupt the DDR2−collagen interaction by binding to the DS domain of DDR2, the determinate region of the protein−protein interaction.59 Monoclonal antibodies including Fab 3E3,8b 48B3,26b and H-12660 have also been developed to bind to the DS-like domain of DDR1. It has been demonstrated that the phosphorylation of tyrosines located in the intracellular juxtamembrane of DDRs and the accumulation of signal adaptor proteins in this region are regulated by tyrosine kinases such as PDGFR, TrkA, Shc, and Src.61 Small molecule inhibitors of these kinases have been reported to interfere indirectly with the signaling pathway of DDRs. In addition, the phosphatase inhibitor pervanadate was also reported to induce rapid and significant autophosphorylation of DDR1 in the absence of ligand stimuli, implying that a phosphatase activator may suppress activation of DDRs.62 As members of the RTK superfamily, the enzymatic activity of DDRs is determined by their KDs. Consequently, small molecular inhibitors targeting the cytosolic kinase domain represent one of the most promising strategies for suppression of the biological functions of DDRs. This has been extensively validated by the inhibitors of many other RTKs such as BcrAbl,63 EGFR,64 or BTK.65 Because of the structural similarities in kinase domains,11 a number of well recognized kinase inhibitors such as the Bcr-Abl inhibitors 2, 3, nilotinib (4),11,68 bafetinib (5),69 and 7,12 the B-Raf inhibitor sorafenib (9), the VEGFR inhibitor pazopanib (10), the c-Met/VEGFR-2 inhibitor foretinib (11), and the p38 MAPK inhibitor BIRB796 (12)66 were found potently to inhibit the functions of both

Figure 6. Natural antibiotic 1, a novel DDR2−collagen interaction blocker.

compound selectively inhibits the activation of DDR2 stimulated by collagen I in HEK293 cells but in a parallel assay has little effect on the activation of DDR1b and also EFGR and IR kinases. However, the relatively weak inhibitory activity of compound 1 (IC50 = 9.0 μM in HEK293 cells) may restrict its further application as a biological tool for research in DDRs. E

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

Figure 7. Known kinase inhibitors with DDR inhibitory effects.

Figure 8. Cocrystal structure of drugs 3 (A, PDB code 4BKJ) and 7 (B, PDB code 3ZOS) with DDR1 kinase domain. The ligands are shown in stick model colored by C in purple, N in blue, and O in red, while the protein is shown in cartoon model. The yellow dash line indicates a hydrogen bond.



NONSELECTIVE INHIBITION OF DDRS BY KNOWN KINASE INHIBITORS A sequence alignment of human DDRs reveals that in the ATP binding domain they share ∼61% sequence identity with BcrAbl.11 Jarai et al. successfully developed a TR-FRET biochemical assay to identify three well-recognized type I or type II Bcr-Abl inhibitors 2, 3, and 4 which potently inhibit DDR1b and DDR2 with IC50 values of 0.5, 337, 43 nM and 1.4, 675, 55 nM, respectively.11 Further cell based investigations confirmed that the compounds potently suppress collageninduced autophosphorylation of DDR1b and DDR2 and inhibit MCP-1 release in monocytic cells in a DDR1 dependent manner.11 The strong DDR inhibitory effects of compounds 2, 3, and 4 were also independently validated by a global chemical proteomics approach. 68 In addition, drug 2 has been

demonstrated to display highly promising therapeutic efficacy in lung cancer cells harboring “gain-of-function” mutations of DDR2.47 In a clinical investigation two squamous cell carcinoma (SCC) patients with a DDR2S768R mutation were shown to have significant shrinkage of their tumors after the treatment with drug 2.49 Other type II Bcr-Abl inhibitors such as 5, 6, 7, and GZD824 (8) were also reported to display strong DDR inhibition (Figure 7).69 X-ray structures of cocrystals of DDR1 with drugs 3 and 7 have been reported and demonstrate that the compounds bind to inactive DDR1 with a type II binding mode similar to that of Bcr-Abl kinase (Figure 8).12 Upon binding of the inhibitor, a hydrophobic pocket is induced by extruding the C-helix and twisting the DFG motif in DDR1. Both of the inhibitors form a strong hydrogen bonding network with the E672, M704, V763, F

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

Figure 9. Selective DDR1/DDR2 kinase inhibitors.

EphA3, EphB4, VEGF receptor 2, misshapen/niks-related kinase 1, c-Abl, and RET. Further study is needed to demonstrate the correlation of the therapeutic efficacy to its DDR inhibition.

H764, and D784 residues of the protein, and other favorable van der Waals contacts are formed in the allosteric pocket. Although compound 7 fails to enjoy the key hydrogen bond interaction with the T701 gatekeeper residue which contributes greatly to the binding of drug 3 with DDR1, a strong hydrogenbond interaction is formed between M704 residue with the imidazo[1, 2-b]pyridazine moiety in 7, and this compensates for the loss of potency. In addition to the reported Bcr-Abl inhibitors, several other kinase inhibitors displaying potent DDR inhibitory effects have also been identified (Figure 7).66 For instance, drug 9, a B-Raf/ VGFR dual inhibitor approved for advanced hepatocellular carcinoma, was recently reported to bind to DDR1 and DDR2 with Kd values of 1.5 and 6.6 nM, respectively. A VEGFR inhibitor, 10 also exhibits off-target binding to DDR2 with a Kd value of 57 nM. Molecule 11 is a c-Met/VEGFR-2 dual inhibitor, but a kinase selectivity profiling study revealed that it binds potently to DDR1 with a Kd value of 0.2 nM. A reported p38 MAPK inhibitor 12 also tightly binds to DDR1 and DDR2 with Kd values of 1.9 and 33 nM, respectively. Recently, a thienopyridine, compound 13, was identified as a novel small molecule DDR inhibitor by random screening of a compound library followed by medicinal chemical optimization.58 This compound selectively suppresses the activated tyrosine kinase activity of DDR2 with an IC50 value of 6.0 nM, although it is clearly less potent with the unactivated kinase, with which its IC50 value is 145 nM. It also inhibits collageninduced DDR1b or DDR2 receptor autophosphorylation with IC50 values of 164 and 171 nM, respectively, in HEK293 cells which stably overexpress either DDR1b (HEK293-DDR1b) or DDR2 (HEK293-DDR2). Further biological investigation reveals that 13, by inactivation of α-smooth muscle actin, potently inhibits the proliferation and migration of primary dermal fibroblasts induced by transforming growth factor β1 (TGFβ1) and type I collagen, FAK, and Akt1. The compound also dose-dependently inhibits lipopolysaccharide (LPS) induced cell migration and the syntheses of inflammatory factors like nitric oxide (NO), inducible nitric oxide synthase (iNOS), cyclooxygenase 2 (COX-2), and tumor necrosis factor α (TNFα) in J774A.1 macrophage cells. In a rabbit ear wound healing model, topical treatment of 13 potently reduces the hypertrophic scar formation without delaying the wound closing process, suggesting the therapeutic potential of DDR inhibitors to treat diseases related to fibroinflammation. However, in a kinase profiling assay against 60 kinases, the compound displays broad inhibition against a panel of 20 tyrosine kinases including Btk, Syk, Tie2, FLT1, FLT3, FLT4,



SELECTIVE DDR KINASE INHIBITORS



COMPOUND 17 AND ITS ANALOGUES

To date, several classes of DDR kinase inhibitors have been identified. Most of them however display broad inhibitory activity against a panel of many other kinases, and selective DDR1 and/or DDR2 inhibitors are highly desirable for further validation of DDRs as drug targets. Sequence alignment of human DDRs demonstrated that they share high homology with many kinases in the ATP binding domain, and this highlights the challenge of identifying selective DDR inhibitors.11 Only recently, Ding et al. reported a pyrazolopyrimidine alkyne derivative 17 which selectively binds and inhibits the enzymatic function of DDR1.67 Compound 18 has also been identified as a selective DDR1 inhibitor by Gray’s group at the Dana Farber Cancer Institute.14

On the basis of the sequence similarity of DDR1/2 and Bcr-Abl in the kinase domain, Ding et al. conducted a focused screening against an internal library containing approximately 2000 kinase inhibitors and identified N-isopropyl-4-methyl-3-(2-(pyrazolo[1,5-a]pyrimidin-6-yl)ethynyl)benzamide 14 as a new potent DDR1 inhibitor with almost the same potency to c-Kit kinase (Figure 9).67 Structural optimization by replacing the isopropyl moiety with a meta-trifluoromethyl-substituted phenyl group gave compound 15, which has an improved DDR1 inhibitory activity and a diminished effect on c-Kit kinase. In spite of the introduction of a pharmaceutically acceptably hydrophilic (4methylpiperazin-1-yl)methyl group, compound 16 did not apparently lose its inhibitory activity against DDR1; the modification clearly restored the potency against Bcr-Abl. Further replacement of the “flag methyl” group with an ethyl group yielded a highly selective DDR1 inhibitor 17. This compound potently inhibits DDR1 kinase with an IC50 value of 6.8 nM but is significantly less active against DDR2, Bcr-Abl, and c-Kit with IC50 values of 101.4, 355 and >10 000 nM, respectively. Kinase profiling analysis using an Ambit kinome screening platform reveals that compound 17 possesses an excellent selective profile for DDR1. The new inhibitor 17 tightly binds to the ATP-binding site of DDR1 with a Kd value of 0.6 nM under an active-site-dependent competition binding assay,70 while it only displays ∼65% binding rates with 14 of the G

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

Figure 10. Other DDR kinases inhibitors.

enzymatic kinase assay, with IC50 values of 105 and 47 nM, respectively. They also exhibit strong blockage on the collagen induced autophosphorylation of DDR1 in U2OS cells with EC50 values of 86 and 9.0 nM, respectively (Figure 9).14 A KinomeScan profiling study was then conducted to demonstrate that inhibitor 18 displays a good selective profile on DDR1 with the S (1) score of 0.01 against 451 kinase at 1.0 μM, whereas compound 19 is considerably less selective, with an S (1) score of 0.07. Further studies demonstrated that inhibitor 18 shows significant synergy with PI3K or mTOR inhibitors to suppress the proliferation of cancer cells with DDR1 gain-of-function mutations and/or overexpression, but it is not active as single agents.14 The cocrystal X-ray structure of 18 with DDR1 confirms its type II binding mode with the protein. The indolinone motif of 18 forms two pairs of hydrogen bonds with Asp702 and Met704 in the hinge region, respectively, while the trifluoromethyl group binds deeply into a hydrophobic pocket directly adjacent to the ATP binding site. The amide group and the nitrogen in piperazine can also make additional hydrogen bond networks with Glu672, Asp784, Val763, and His764, respectively. A mutation of the gatekeeper residue T701 leads to a significant loss of potency of 18, while a G707A mutation in the hinge region results in 20- or 100-fold improvement in potency for compounds 18 and 19, respectively, further supporting the postulate of their binding to the DDR1 kinase domain.

other 395 nonmutated kinases evaluated at 100 nM which is about 160 times higher than its Kd value with DDR1. The offtarget kinases include Abl1, DDR2, EPHA8, HCK, LOK, MAK, PDGFRβ, Tie2, TRKb, TRBc, and ZAK, and the S (35) and S (10) selectivity scores are 0.035 and 0.008, respectively. Although the detail interaction of inhibitor 17 with DDR1 remains unclear, it is predicted that the compound binds to the receptor with a type II binding mode. Cell based investigation also reveals that 17 dose-dependently inhibits the activation of DDR1 and downstream signals and potently suppresses the proliferation, invasion, adhesion, and tumorigenicity of cancer cells with high level of DDR1. Preliminary pharmacokinetic studies were also conducted and demonstrated that the compound possesses promising PK profiles, with an oral bioavailability of 67.4% and T1/2 of 15.5 h.



INHIBITOR 18 AND THE DERIVATIVES Typically, a type II kinase inhibitor contains several key structural elements that engage in crucial interactions with the protein: (1) a heterocyclic moiety binding to the adenine pocket and participating in a hydrogen bonding network with residues in the hinge region (“hinge binding part”); (2) a hydrophobic group binding the back pocket induced by the DFG-out conformation (“tail motif”); (3) a suitable linker between the “head region” and the “tail motif”.14 On the basis of this pharmacophore model, scientists in Gray’s group designed and synthesized a collection of ∼100 type II inhibitors, among which compound 18 and DDR1-IN-2 (19) displayed significant inhibitory activity against DDR1 in an H

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

Figure 11. Chemical structures of new DDR inhibitors with unknown mechanism.



OTHER DDR KINASE INHIBITORS With the aim of discovering novel DDR inhibitors, Rauh et al. developed a new “fluorescent labels in kinases” (FLiK) assay to successfully identify a series of pyrazolourea compounds (20− 22) as new type II inhibitors of DDR2 with the DFG-out conformation (Figure 10).13 The DDR2 inhibitory effects of the compounds were further validated by an orthogonal activity-based assay. Compounds 20, 21, and 22 potently inhibit DDR2 with IC50 values of 0.065, 0.075, and 0.018 μM, respectively. The compounds also strongly inhibit DDR1 with IC50 values of 0.068, 0.235, and 0.039 μM, respectively. Significantly, compounds 20 and 22 also exhibit strong suppression against the T654M gatekeeper mutant of DDR2 with IC50 values of 2.0 and 1.0 nM, respectively. It is noteworthy that from an in vitro investigation in DDR2dependent lung cancer cell lines, the gatekeeper mutation T654I has been implied as a primary mechanism for the acquired resistance against DDR inhibitor therapy.71 Thus, the inhibitory effects of the compounds against DDR2T654I is eagerly awaited to demonstrate their potential to overcome DDR2 mutation induced resistance. The compounds may also provide useful probes with which to decipher the orchestrated network of signaling by DDRs and their relevance to tumor biology. However, further kinase profiling study reveals that the compounds display relatively broad inhibitory spectra against a panel of protein kinases. Furthermore, since no cocrystal structures of DDR2 with type II inhibitors are currently available, extensive investigations to demonstrate that binding modes of the compounds with wild type DDR2 and the resistant mutant are highly desirable. Recently, benzamide72 and quinazolinedione73 derivatives have also been identified as novel DDR1 inhibitors (Figure 10). A benzamide-derived compound 23 was found to exhibit inhibitory activity against DDR1 kinase with an IC50 value of 0.097 μM.72 It also suppresses the proliferation of U2OS cells harboring DDR1 overexpression with an IC50 value of 0.44 μM. The conformationally restrained quinazoline 24 also strongly inhibits the kinase activity of DDR1 with an IC50 value of 0.043 μM.73 It is encouraging that compounds 23 and 24 show promising in vivo antitumor efficacies with tumor growth inhibition (TGI) rates of ∼60% after oral administration in a mouse xenograft model of DDR1 overexpressed cancer cells. Benzylurea derivatives have also been identified by the Merck pharmaceutical company as potent DDR2 inhibitors (Figure 10).74 For instance, compound 25 suppresses the enzymatic activity of DDR2 with an IC50 value of 30 nM. Further work led to the discovery of more potent compounds 26, 27, and 28 which have IC50 values of 15, 20, and 6.0 nM, respectively. Biological investigation also revealed that compound 25 induces a dose-dependent reduction in the extent of thermal

hyperalgesia but not the carrageenan (CAR) induced swelling in rats, suggesting the potential application of DDR2 inhibitors as novel pain-reducing agents. However, further evidence demonstrating that the in vivo efficacies of the compounds are related to their DDR1 or DDR2 inhibition is eagerly awaited and no kinase selectivity profile of the compounds is available.



DDR INHIBITORS WITH UNKNOWN MECHANISM OF ACTION A family of polycyclic alkaloids 29−32 (discoipyrroles A−D, Figure 11), isolated from marine-derived Bacillus hunanensis, strain SNA-048, have recently been discovered to inhibit BR5 fibroblast migration in a 2D Matrigel matrix at 1.0 μM.75 The compounds also selectively inhibit the growth of HCC366 nonsmall-cell lung cancer cells harboring a “gain of function” mutation of DDR2 but are clearly less potent against A549 cancer cells with wild-type DDR2. Preliminary biological investigations suggest that compound 29 exhibits its biological effects through regulation of the glycosylation status of DDR2 during post-transcriptional modification. However, direct evidence demonstrating whether or how the compounds interact with DDR2 is still lacking.



PERSPECTIVE DDR1 and DDR2 are promising targets for discovery of drugs for the treatment of human cancer or inflammation-related diseases including renal fibrosis, pulmonary fibrosis, atherosclerosis, or osteoarthritis, etc. Several classes of small molecule DDR inhibitors with different selectivity profiles have been discovered. Among these, compound 2 in an early stage of clinical investigation has shown promising benefit for a small population of SCC patients with activating mutation of DDR2. Some other compounds also show promising therapeutic potential for various human cancers, would healing promotion, and antihyperalgesic effect. However, most of the reported DDR inhibitors display broad inhibition against a panel of protein kinases, and it is thus difficult to correlate their therapeutic efficacies with their DDR inhibition. Encouragingly, a few highly selective small DDR1 inhibitors have recently been identified. Compound 18 shows great synergistic effect with inhibitors of PI3K or mTOR in cancer cell growth suppression assays.14 Combination of inhibitor 17 with the first-line chemodrugs also shows a significantly improved therapeutic benefit for pancreatic cancer and NSCLC cancer in different animal models.76 These results intimate the promising potential for DDR-based therapy. However, the studies also demonstrate that almost all of the selective DDR1 inhibitors used alone fail to achieve significant inhibition of the in vitro growth of various cancer cell lines, suggesting that DDRs may not be the driving force for proliferation of the cancer cells. More biological I

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry investigations to demonstrate the precise roles of DDRs in human cancer establishment and development are highly desirable. It will be of special value to identify “activating mutation” of DDRs in the primary human cancer tissues. In addition, although biological investigation suggests that DDRs are important for a variety of inflammatory processes, there is still a lack of convincing evidence demonstrating the therapeutic application of selective DDR inhibitors against inflammation related disorders such as atherosclerosis, pulmonary fibrosis, or renal fibrosis. Nevertheless, given the fact that DDR overexpression and/or mutation contributes greatly to acquired resistance and plays critical roles in tumor invasion and metastasis for certain types of cancer,6,47 it is rational to postulate that combination of DDR inhibitors with other therapeutic agents may be a new tractable approach for novel anticancer therapy development. Despite the significant progress in DDR research area, from a medicinal chemist’s point of view, there are still many issues that are highly interesting for future investigations: (1) Is it feasible to design a selective DDR2 inhibitor over DDR1, since they share extremely high sequence identity in the ATP binding domain? A cocrystal structure will provide insight to the interaction of an inhibitor with DDR2 kinase domain. (2) Molecules selectively targeting the activating and/or drug induced DDR mutants, particularly the resistant gatekeeper mutants (i.e., DDR2T654I/M), will be of special value for future DDRs-based drug discovery. (3) DDR1/DDR2 inhibitors with preferred tissue distribution profile are highly desirable, since DDRs may function differently in various human tissues. (4) Will dual DDR1/DDR2 inhibition achieve synergistic effect or be antagonistic? (5) In vivo efficacy data of a selective DDR1 or DDR2 inhibitor are eagerly awaited for validating their therapeutic potential. (6) What are the optimal therapeutic indications and the corresponding disease models for DDR inhibitor therapies? In summary, biological investigations suggest DDRs as novel potential targets for drug discovery to treat human cancer and inflammatory disorders. A number of small molecule DDR inhibitors have been identified with different selectivity profiles, and some of these have been demonstrated to possess a promising therapeutic potential. However, given the fact that the precise role of DDRs in human diseases remains elusive, more explorations to further validate DDRs as the pharmaceutically accessible molecular targets are necessary. New selective DDR1 and/or DDR2 inhibitors are also highly valuable as research tools in future biological investigations.



of Biomedicine and Health, Chinese Academy of Sciences. He is currently a Postdoctoral Research Fellow working in the computeraided molecular design lab at Mayo Clinic under the supervision of Prof. Yuan-ping Pang. His research mainly focuses on the discovery and optimization of inhibitors targeting key kinase receptors as well as protein−protein interactions. Xiaoyun Lu is an Associate Research Professor in Medicinal Chemistry at Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences. She received her Ph.D. from China Pharmaceutical University in 2010 and then joined Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, as a Research Associate in Prof. Ke Ding’s group. She was promoted to Associate Research Professor in 2012. Her research mainly focuses on the rational design and synthesis of medicinal lead compounds targeting cancer and tuberculosis. Xiaomei Ren is an Associate Research Professor in Pharmacology at Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences. She got her Bachelor and Master degrees from China Pharmaceutical University and then worked as a Research Associate in Shanghai Institutes of Biological Sciences (Chinese Academy of Sciences), the University of Michigan (Ann Arbor) and Guangzhou Institutes of Biomedicine and Health and health (Chinese Academy of Sciences). She obtained her Ph.D. from Sun Yat-Sen University in 2013 and become an Associate Professor at Guangzhou Institutes of Biomedicine and Health (Chinese Academy of Sciences) in 2014. Her research mainly focuses on biological assay development and bioactivity evaluation of new anticancer molecules. Ke Ding got his Bachelor and Master degrees from China Pharmaceutical University in 1995 and 1998, respectively, and obtained his Ph.D. degree in Bioorganic Chemistry from Fudan University in 2001. After 5-year postdoctoral training at University of Michigan, Ann Arbor, he became a Professor of Medicinal Chemistry in Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences in 2006. Dr. Ding currently serves as an editorial board member for J. Med. Chem., ACS Med. Chem. Lett., and J. Chin. Pharm. Sci. Dr. Ding’s research interests mainly focus on design and synthesis of novel bioactive lead compounds for drug discovery. He has published more than 90 publications and is co-inventor of over 40 international patents, some of which have been licensed to international pharmaceutical companies.



ACKNOWLEDGMENTS We are thankful for a Strategic Collaboration grant between China and New Zealand from Ministry of Science and Technology of China (Grant 2014DFG32100), National Science Fund for Distinguished Young Scholars of China (Grant 81425021), and 100-Distinguished Scientist Award of Guangdong Province (Nanyue-Baijie Award) for financial support.

AUTHOR INFORMATION

Corresponding Authors

*X.R.: phone, 86-20-32015300; e-mail, [email protected]. cn. *K.D.: phone, +86-20-32015276; fax, +86-20-32015299; e-mail, [email protected].



ABBREVIATIONS USED DDR, discoidin domain receptor; RTK, receptor tyrosine kinase; ECM, extracellular matrix; DS, discoidin; EJXM, extracellular juxtamembrane; TM, transmembrane; IJXM, intracellular juxtamembrane; KD, kinase domain; DFG, AspPhe-Gly; SCC, squamous cell carcinoma; SMC, smooth muscle cell; GBM, glomerular basement membrane; MCP-1, monocyte chemoattractant protein 1; IR, insulin receptor; TGI, tumor growth inhibition; CAR, carrageenan; SMED-SL, spondylometa-epiphyseal dysplasia with short limbs and abnormal calcifications; i-NOS, inducible nitric oxide synthase; COX-2,

Author Contributions §

Y.L. and X.L. contributed equally to this work.

Notes

The authors declare no competing financial interest. Biographies Yupeng Li received his B.S. degree in Traditional Chinese Medicine in 2009 at Huazhong University of Science & Technology, Tongji Medical School, and completed his Ph.D. in Medicinal Chemistry in 2014 under the supervision of Prof. Ke Ding at Guangzhou Institutes J

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

receptor DDR2 is a receptor for type X collagen. Matrix Biol. 2006, 25, 355−364. (f) Agarwal, G.; Mihai, C.; Iscru, D. F. Interaction of discoidin domain receptor 1 with collagen type 1. J. Mol. Biol. 2007, 367, 443−455. (5) Xu, H.; Raynal, N.; Stathopoulos, S.; Myllyharju, J.; Farndale, R. W.; Leitinger, B. Collagen binding specificity of the discoidin domain receptors: binding sites on collagens II and III and molecular determinants for collagen IV recognition by DDR1. Matrix Biol. 2011, 30, 16−26. (6) (a) Valiathan, R. R.; Marco, M.; Leitinger, B.; Kleer, C. G.; Fridman, R. Discoidin domain receptor tyrosine kinases: new players in cancer progression. Cancer Metast. Rev. 2012, 31, 295−321. (b) Vogel, W. F.; Abdulhussein, R.; Ford, C. E. Sensing extracellular matrix: an update on discoidin domain receptor function. Cell Signal 2006, 18, 1108−1116. (c) Leitinger, B. Discoidin domain receptor functions in physiological and pathological conditions. Int. Rev. Cell Mol. Biol. 2014, 310, 39−87. (d) Iwai, L. K.; Luczynski, M. T.; Huang, P. H. Discoidin domain receptors: a proteomic portrait. Cell. Mol. Life Sci. 2014, 71, 3269−3279. (e) Borza, C. M.; Pozzi, A. Discoidin domain receptors in disease. Matrix Biol. 2014, 34, 185−192. (f) Kothiwale, S.; Borza, C. M.; Lowe, W.; Meiler, J. Discoidin domain receptor 1 (DDR1) kinase as target for structure-based drug discovery. Drug Discovery Today 2014, DOI: 10.1016/j.drudis.2014.09.025. (7) (a) Kiedzierska, A.; Smietana, K.; Czepczynska, H.; Otlewski, J. Structural similarities and functional diversity of eukaryotic discoidinlike domains. Biochim. Biophys. Acta 2007, 1774, 1069−1078. (b) Carafoli, F.; Hohenester, E. Collagen recognition and transmembrane signalling by discoidin domain receptors. Biochim. Biophys. Acta 2013, 1834, 2187−2194. (8) (a) Carafoli, F.; Bihan, D.; Stathopoulos, S.; Konitsiotis, A. D.; Kvansakul, M.; Farndale, R. W.; Leitinger, B.; Hohenester, E. Crystallographic insight into collagen recognition by discoidin domain receptor 2. Structure 2009, 17, 1573−1581. (b) Carafoli, F.; Mayer, M. C.; Shiraishi, K.; Pecheva, M. A.; Chan, L. Y.; Nan, R.; Leitinger, B.; Hohenester, E. Structure of the discoidin domain receptor 1 extracellular region bound to an inhibitory Fab fragment reveals features important for signaling. Structure 2012, 20, 688−697. (9) (a) Noordeen, N. A.; Carafoli, F.; Hohenester, E.; Horton, M. A.; Leitinger, B. A transmembrane leucine zipper is required for activation of the dimeric receptor tyrosine kinase DDR1. J. Biol. Chem. 2006, 281, 22744−22751. (b) Mihai, C.; Chotani, M.; Elton, T. S.; Agarwal, G. Mapping of DDR1 distribution and oligomerization on the cell surface by FRET microscopy. J. Mol. Biol. 2009, 385, 432−445. (c) Lemmon, M. A.; Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 2010, 141, 1117−1134. (10) Lemeer, S.; Bluwstein, A.; Wu, Z.; Leberfinger, J.; Muller, K.; Kramer, K.; Kuster, B. Phosphotyrosine mediated protein interactions of the discoidin domain receptor 1. J. Proteomics 2012, 75, 3465−3477. (11) Day, E.; Waters, B.; Spiegel, K.; Alnadaf, T.; Manley, P. W.; Buchdunger, E.; Walker, C.; Jarai, G. Inhibition of collagen-induced discoidin domain receptor 1 and 2 activation by imatinib, nilotinib and dasatinib. Eur. J. Pharmacol. 2008, 599, 44−53. (12) Canning, P.; Tan, L.; Chu, K.; Lee, S. W.; Gray, N. S.; Bullock, A. N. Structural mechanisms determining inhibition of the collagen receptor DDR1 by selective and multi-targeted type II kinase inhibitors. J. Mol. Biol. 2014, 426, 2457−2470. (13) Richters, A.; Nguyen, H. D.; Phan, T.; Simard, J. R.; Grütter, C.; Engel, J.; Rauh, D. Identification of type II and III DDR2 inhibitors. J. Med. Chem. 2014, 57, 4252−4262. (14) Kim, H. G.; Tan, L.; Weisberg, E. L.; Liu, F.; Canning, P.; Choi, H. G.; Ezell, S. A.; Wu, H.; Zhao, Z.; Wang, J.; Mandinova, A.; Griffin, J. D.; Bullock, A. N.; Liu, Q.; Lee, S. W.; Gray, N. S. Discovery of a potent and selective DDR1 receptor tyrosine kinase inhibitor. ACS Chem. Biol. 2013, 8, 2145−2150. (15) Ichikawa, O.; Osawa, M.; Nishida, N.; Goshima, N.; Nomura, N.; Shimada, I. Structural basis of the collagen-binding mode of discoidin domain receptor 2. EMBO J. 2007, 26, 4168−4176.

cyclooxygenase 2; JNK, c-Jun N-terminal kinase; PDGFR, platelet-derived growth factor receptor; TrkA, tropomycinrelated kinase A; BTK, Bruton tyrosine kinase; Syk, spleen tyrosine kinase; FLT, FMS-like tyrosine kinase; ZAK, zipper and sterile α motif containing kinase Amino Acids

Ala or A, alanine; Arg or R, arginine; Asn or N, asparagines; Asp or D, aspartic acid; Cys or C, cysteine; Gln or Q, glutamine; Glu or E, glutamic acid; Gly or G, glycine; His or H, histidine; Ile or I, isoleucine; Leu or L, leucine; Lys or K, lysine; Met or M, methionine; Phe or F, phenylalanine; Pro or P, proline; Ser or S, serine; Thr or T, threonine; Trp or W, tryptophan; Tyr or Y, tyrosine; Val or V, valine



REFERENCES

(1) (a) Di Marco, E.; Cutuli, N.; Guerra, L.; Cancedda, R.; De Luca, M. Molecular cloning of trkE, a novel trk-related putative tyrosine kinase receptor isolated from normal human keratinocytes and widely expressed by normal human tissues. J. Biol. Chem. 1993, 268, 24290− 24295. (b) Johnson, J. D.; Edman, J. C.; Rutter, W. J. A receptor tyrosine kinase found in breast carcinoma cells has an extracellular discoidin I-like domain. Proc. Natl. Acad. Sci. U. S. A. 1993, 90, 5677− 5681. (c) Karn, T.; Holtrich, U.; Brauninger, A.; Bohme, B.; Wolf, G.; Rubsamen-Waigmann, H.; Strebhardt, K. Structure, expression and chromosomal mapping of TKT from man and mouse: a new subclass of receptor tyrosine kinases with a factor VIII-like domain. Oncogene 1993, 8, 3433−3440. (d) Zerlin, M.; Julius, M. A.; Goldfarb, M. NEP: a novel receptor-like tyrosine kinase expressed in proliferating neuroepithelia. Oncogene 1993, 8, 2731−2739. (e) Lai, C.; Lemke, G. Structure and expression of the Tyro 10 receptor tyrosine kinase. Oncogene 1994, 9, 877−883. (f) Perez, J. L.; Shen, X.; Finkernagel, S.; Sciorra, L.; Jenkins, N. A.; Gilbert, D. J.; Copeland, N. G.; Wong, T. W. Identification and chromosomal mapping of a receptor tyrosine kinase with a putative phospholipid binding sequence in its ectodomain. Oncogene 1994, 9, 211−219. (g) Alves, F.; Vogel, W.; Mossie, K.; Millauer, B.; Hofler, H.; Ullrich, A. Distinct structural characteristics of discoidin I subfamily receptor tyrosine kinases and complementary expression in human cancer. Oncogene 1995, 10, 609− 618. (h) Shelling, A. N.; Butler, R.; Jones, T.; Laval, S.; Boyle, J. M.; Ganesan, T. S. Localization of an epithelial-specific receptor kinase (EDDR1) to chromosome 6q16. Genomics 1995, 25, 584−587. (i) Laval, S.; Butler, R.; Shelling, A. N.; Hanby, A. M.; Poulsom, R.; Ganesan, T. S. Isolation and characterization of an epithelial-specific receptor tyrosine kinase from an ovarian cancer cell line. Cell Growth Differ. 1994, 5, 1173−1183. (j) Perez, J. L.; Jing, S. Q.; Wong, T. W. Identification of two isoforms of the Cak receptor kinase that are coexpressed in breast tumor cell lines. Oncogene 1996, 12, 1469−1477. (2) Playford, M. P.; Butler, R. J.; Wang, X. C.; Katso, R. M.; Cooke, I. E.; Ganesan, T. S. The genomic structure of discoidin receptor tyrosine kinase. Genome Res. 1996, 6, 620−627. (3) Alves, F.; Saupe, S.; Ledwon, M.; Schaub, F.; Hiddemann, W.; Vogel, W. F. Identification of two novel, kinase-deficient variants of discoidin domain receptor 1: differential expression in human colon cancer cell lines. FASEB J. 2001, 15, 1321−1323. (4) (a) Shrivastava, A.; Radziejewski, C.; Campbell, E.; Kovac, L.; McGlynn, M.; Ryan, T. E.; Davis, S.; Goldfarb, M. P.; Glass, D. J.; Lemke, G.; Yancopoulos, G. D. An orphan receptor tyrosine kinase family whose members serve as nonintegrin collagen receptors. Mol. Cell 1997, 1, 25−34. (b) Vogel, W.; Gish, G. D.; Alves, F.; Pawson, T. The discoidin domain receptor tyrosine kinases are activated by collagen. Mol. Cell 1997, 1, 13−23. (c) Leitinger, B. Molecular analysis of collagen binding by the human discoidin domain receptors, DDR1 and DDR2. Identification of collagen binding sites in DDR2. J. Biol. Chem. 2003, 278, 16761−16769. (d) Leitinger, B.; Steplewski, A.; Fertala, A. The D2 period of collagen II contains a specific binding site for the human discoidin domain receptor, DDR2. J. Mol. Biol. 2004, 344, 993−1003. (e) Leitinger, B.; Kwan, A. P. The discoidin domain K

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry (16) (a) Leitinger, B. Transmembrane collagen receptors. Annu. Rev. Cell Dev. Biol. 2011, 27, 265−290. (b) Yeh, Y. C.; Lin, H. H.; Tang, M. J. A tale of two collagen receptors, integrin beta1 and discoidin domain receptor 1, in epithelial cell differentiation. Am. J. Physiol-Cell Physiol. 2012, 303, C1207−1217. (c) Fu, H. L.; Valiathan, R. R.; Arkwright, R.; Sohail, A.; Mihai, C.; Kumarasiri, M.; Mahasenan, K. V.; Mobashery, S.; Huang, P.; Agarwal, G.; Fridman, R. Discoidin domain receptors: unique receptor tyrosine kinases in collagen-mediated signaling. J. Biol. Chem. 2013, 288, 7430−7437. (17) (a) Vogel, W. F.; Aszodi, A.; Alves, F.; Pawson, T. Discoidin domain receptor 1 tyrosine kinase has an essential role in mammary gland development. Mol. Cell. Biol. 2001, 21, 2906−2917. (b) FaraciOrf, E.; McFadden, C.; Vogel, W. F. DDR1 signaling is essential to sustain Stat5 function during lactogenesis. J. Cell. Biochem. 2006, 97, 109−121. (18) (a) Torban, E.; Goodyer, P. The kidney and ear: emerging parallel functions. Annu. Rev. Med. 2009, 60, 339−353. (b) Meyer zum Gottesberge, A. M.; Gross, O.; Becker-Lendzian, U.; Massing, T.; Vogel, W. F. Inner ear defects and hearing loss in mice lacking the collagen receptor DDR1. Lab Invest. 2008, 88, 27−37. (c) Gross, O.; Beirowski, B.; Harvey, S. J.; McFadden, C.; Chen, D.; Tam, S.; Thorner, P. S.; Smyth, N.; Addicks, K.; Bloch, W.; Ninomiya, Y.; Sado, Y.; Weber, M.; Vogel, W. F. DDR1-deficient mice show localized subepithelial GBM thickening with focal loss of slit diaphragms and proteinuria. Kidney Int. 2004, 66, 102−111. (19) (a) Li, S. W.; Prockop, D. J.; Helminen, H.; Fassler, R.; Lapvetelainen, T.; Kiraly, K.; Peltarri, A.; Arokoski, J.; Lui, H.; Arita, M. Transgenic mice with targeted inactivation of the Col2 alpha 1 gene for collagen II develop a skeleton with membranous and periosteal bone but no endochondral bone. Genes Dev. 1995, 9, 2821− 2830. (b) Yang, C.; Li, S. W.; Helminen, H. J.; Khillan, J. S.; Bao, Y.; Prockop, D. J. Apoptosis of chondrocytes in transgenic mice lacking collagen II. Exp. Cell Res. 1997, 235, 370−373. (20) Labrador, J. P.; Azcoitia, V.; Tuckermann, J.; Lin, C.; Olaso, E.; Manes, S.; Bruckner, K.; Goergen, J. L.; Lemke, G.; Yancopoulos, G.; Angel, P.; Martinez, C.; Klein, R. The collagen receptor DDR2 regulates proliferation and its elimination leads to dwarfism. EMBO Rep. 2001, 2, 446−452. (21) Kawai, I.; Matsumura, H.; Fujii, W.; Naito, K.; Kusakabe, K.; Kiso, Y.; Kano, K. Discoidin domain receptor 2 (DDR2) regulates body size and fat metabolism in mice. Transgenic Res. 2014, 23, 165− 175. (22) (a) Borochowitz, Z.; Langer, L. O., Jr.; Gruber, H. E.; Lachman, R.; Katznelson, M. B.; Rimoin, D. L. Spondylo-meta-epiphyseal dysplasia (SMED), short limb-hand type: a congenital familial skeletal dysplasia with distinctive features and histopathology. Am. J. Med. Genet. 1993, 45, 320−326. (b) Kano, K.; Marin de Evsikova, C.; Young, J.; Wnek, C.; Maddatu, T. P.; Nishina, P. M.; Naggert, J. K. A novel dwarfism with gonadal dysfunction due to loss-of-function allele of the collagen receptor gene, Ddr2, in the mouse. Mol. Endocrinol. 2008, 22, 1866−1880. (c) Bargal, R.; Cormier-Daire, V.; Ben-Neriah, Z.; Le Merrer, M.; Sosna, J.; Melki, J.; Zangen, D. H.; Smithson, S. F.; Borochowitz, Z.; Belostotsky, R.; Raas-Rothschild, A. Mutations in DDR2 gene cause SMED with short limbs and abnormal calcifications. Am. J. Hum. Genet. 2009, 84, 80−84. (d) Ali, B. R.; Xu, H.; Akawi, N. A.; John, A.; Karuvantevida, N. S.; Langer, R.; Al-Gazali, L.; Leitinger, B. Trafficking defects and loss of ligand binding are the underlying causes of all reported DDR2 missense mutations found in SMED-SL patients. Hum. Mol. Genet. 2010, 19, 2239−2250. (e) Al-Anbouri, L.; Leitinger, B.; Ali, B. R. A novel mutation in DDR2 causing spondylometa-epiphyseal dysplasia with short limbs and abnormal calcifications (SMED-SL) results in defective intra-cellular trafficking. BMC Med. Genet. 2014, 15, 42 DOI: 10.1186/1471-2350-15-42. (23) Kamohara, H.; Yamashiro, S.; Galligan, C.; Yoshimura, T. Discoidin domain receptor 1 isoform-a (DDR1a) promotes migration of leukocytes in three-dimensional collagen lattices. FASEB J. 2001, 15, 2724−2726.

(24) Bhatt, R. S.; Tomoda, T.; Fang, Y.; Hatten, M. E. Discoidin domain receptor 1 functions in axon extension of cerebellar granule neurons. Gene. Dev. 2000, 14, 2216−2222. (25) Olaso, E.; Ikeda, K.; Eng, F. J.; Xu, L.; Wang, L. H.; Lin, H. C.; Friedman, S. L. DDR2 receptor promotes MMP-2-mediated proliferation and invasion by hepatic stellate cells. J. Clin. Invest. 2001, 108, 1369−1378. (26) (a) Hou, G.; Vogel, W. F.; Bendeck, M. P. Tyrosine kinase activity of discoidin domain receptor 1 is necessary for smooth muscle cell migration and matrix metalloproteinase expression. Circ. Res. 2002, 90, 1147−1149. (b) Ram, R.; Lorente, G.; Nikolich, K.; Urfer, R.; Foehr, E.; Nagavarapu, U. Discoidin domain receptor-1a (DDR1a) promotes glioma cell invasion and adhesion in association with matrix metalloproteinase-2. J. Neuro-Oncol. 2006, 76, 239−248. (27) Herrera-Herrera, M. L.; Quezada-Calvillo, R. DDR2 plays a role in fibroblast migration independent of adhesion ligand and collagen activated DDR2 tyrosine kinase. Biochem. Bioph. Res. Commun. 2012, 429, 39−44. (28) Kawai, I.; Hisaki, T.; Sugiura, K.; Naito, K.; Kano, K. Discoidin domain receptor 2 (DDR2) regulates proliferation of endochondral cells in mice. Biochem. Bioph. Res. Commun. 2012, 427, 611−617. (29) Kim, D.; You, E.; Min, N. Y.; Lee, K. H.; Kim, H. K.; Rhee, S. Discoidin domain receptor 2 regulates the adhesion of fibroblasts to 3D collagen matrices. Int. J. Mol. Med. 2013, 31, 1113−1118. (30) (a) Heinzelmann-Schwarz, V. A.; Gardiner-Garden, M.; Henshall, S. M.; Scurry, J.; Scolyer, R. A.; Davies, M. J.; Heinzelmann, M.; Kalish, L. H.; Bali, A.; Kench, J. G.; Edwards, L. S.; Vanden Bergh, P. M.; Hacker, N. F.; Sutherland, R. L.; O’Brien, P. M. Overexpression of the cell adhesion molecules DDR1, Claudin 3, and Ep-CAM in metaplastic ovarian epithelium and ovarian cancer. Clin. Cancer Res. 2004, 10, 4427−4436. (b) Quan, J.; Yahata, T.; Adachi, S.; Yoshihara, K.; Tanaka, K. Identification of receptor tyrosine kinase, discoidin domain receptor 1 (DDR1), as a potential biomarker for serous ovarian cancer. Int. J. Mol. Sci. 2011, 12, 971−982. (c) Colas, E.; Perez, C.; Cabrera, S.; Pedrola, N.; Monge, M.; Castellvi, J.; Eyzaguirre, F.; Gregorio, J.; Ruiz, A.; Llaurado, M.; Rigau, M.; Garcia, M.; Ertekin, T.; Montes, M.; Lopez-Lopez, R.; Carreras, R.; Xercavins, J.; Ortega, A.; Maes, T.; Rosell, E.; Doll, A.; Abal, M.; Reventos, J.; GilMoreno, A. Molecular markers of endometrial carcinoma detected in uterine aspirates. Int. J. Cancer 2011, 129, 2435−2444. (31) (a) Yang, S. H.; Baek, H. A.; Lee, H. J.; Park, H. S.; Jang, K. Y.; Kang, M. J.; Lee, D. G.; Lee, Y. C.; Moon, W. S.; Chung, M. J. Discoidin domain receptor 1 is associated with poor prognosis of nonsmall cell lung carcinomas. Oncol. Rep. 2010, 24, 311−319. (b) Miao, L.; Zhu, S.; Wang, Y.; Li, Y.; Ding, J.; Dai, J.; Cai, H.; Zhang, D.; Song, Y. Discoidin domain receptor 1 is associated with poor prognosis of non-small cell lung cancer and promotes cell invasion via epithelial-tomesenchymal transition. Med. Oncol. 2013, 30, 626. (c) Valencia, K.; Ormazabal, C.; Zandueta, C.; Luis-Ravelo, D.; Anton, I.; Pajares, M. J.; Agorreta, J.; Montuenga, L. M.; Martinez-Canarias, S.; Leitinger, B.; Lecanda, F. Inhibition of collagen receptor discoidin domain receptor1 (DDR1) reduces cell survival, homing, and colonization in lung cancer bone metastasis. Clin. Cancer Res. 2012, 18, 969−980. (d) Ford, C. E.; Lau, S. K.; Zhu, C. Q.; Andersson, T.; Tsao, M. S.; Vogel, W. F. Expression and mutation analysis of the discoidin domain receptors 1 and 2 in non-small cell lung carcinoma. Br. J. Cancer 2007, 96, 808− 814. (32) Rikova, K.; Guo, A.; Zeng, Q.; Possemato, A.; Yu, J.; Haack, H.; Nardone, J.; Lee, K.; Reeves, C.; Li, Y.; Hu, Y.; Tan, Z.; Stokes, M.; Sullivan, L.; Mitchell, J.; Wetzel, R.; Macneill, J.; Ren, J. M.; Yuan, J.; Bakalarski, C. E.; Villen, J.; Kornhauser, J. M.; Smith, B.; Li, D.; Zhou, X.; Gygi, S. P.; Gu, T. L.; Polakiewicz, R. D.; Rush, J.; Comb, M. J. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 2007, 131, 1190−1203. (33) (a) Barker, K. T.; Martindale, J. E.; Mitchell, P. J.; Kamalati, T.; Page, M. J.; Phippard, D. J.; Dale, T. C.; Gusterson, B. A.; Crompton, M. R. Expression patterns of the novel receptor-like tyrosine kinase, DDR, in human breast tumours. Oncogene 1995, 10, 569−575. (b) Turashvili, G.; Bouchal, J.; Baumforth, K.; Wei, W.; L

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

30, 1−9. (c) Duncan, J. S.; Whittle, M. C.; Nakamura, K.; Abell, A. N.; Midland, A. A.; Zawistowski, J. S.; Johnson, N. L.; Granger, D. A.; Jordan, N. V.; Darr, D. B.; Usary, J.; Kuan, P. F.; Smalley, D. M.; Major, B.; He, X.; Hoadley, K. A.; Zhou, B.; Sharpless, N. E.; Perou, C. M.; Kim, W. Y.; Gomez, S. M.; Chen, X.; Jin, J.; Frye, S. V.; Earp, H. S.; Graves, L. M.; Johnson, G. L. Dynamic reprogramming of the kinome in response to targeted MEK inhibition in triple-negative breast cancer. Cell 2012, 149, 307−321. (41) Rodrigues, R.; Roque, L.; Espadinha, C.; Pinto, A.; Domingues, R.; Dinis, J.; Catarino, A.; Pereira, T.; Leite, V. Comparative genomic hybridization, BRAF, RAS, RET, and oligo-array analysis in aneuploid papillary thyroid carcinomas. Oncol. Rep. 2007, 18, 917−926. (42) (a) Renne, C.; Willenbrock, K.; Kuppers, R.; Hansmann, M. L.; Brauninger, A. Autocrine- and paracrine-activated receptor tyrosine kinases in classic Hodgkin lymphoma. Blood 2006, 105, 4051−4059. (b) Willenbrock, K.; Kuppers, R.; Renne, C.; Brune, V.; Eckerle, S.; Weidmann, E.; Bräuninger, A.; Hansmann, M. L. Common features and differences in the transcriptome of large cell anaplastic lymphoma and classical Hodgkin’s lymphoma. Haematologica 2006, 91, 596−604. (43) (a) Ren, T.; Zhang, J.; Zhang, J.; Liu, X.; Yao, L. Increased expression of discoidin domain receptor 2 (DDR2): a novel independent prognostic marker of worse outcome in breast cancer patients. Med. Oncol. 2013, 30, 397. (b) Ren, T.; Zhang, W.; Liu, X.; Zhao, H.; Zhang, J.; Zhang, J.; Li, X.; Zhang, Y.; Bu, X.; Shi, M.; Yao, L.; Su, J. Discoidin domain receptor 2 (DDR2) promotes breast cancer cell metastasis and the mechanism implicates epithelial-mesenchymal transition programme under hypoxia. J. Pathol. 2014, DOI: 10.1002/ path.4415. (44) (a) Yan, Z.; Jin, S.; Wei, Z.; Huilian, H.; Zhanhai, Y.; Yue, T.; Juan, L.; Jing, L.; Libo, Y.; Xu, L. Discoidin domain receptor 2 facilitates prostate cancer bone metastasis via regulating parathyroid hormone-related protein. Biochim. Biophys. Acta 2014, 1842, 1350− 1363. (b) Xu, J.; Lu, W.; Zhang, S.; Zhu, C.; Ren, T.; Zhu, T.; Zhao, H.; Liu, Y.; Su, J. Overexpression of DDR2 contributes to cell invasion and migration in head and neck squamous cell carcinoma. Cancer Biol. Ther. 2014, 15, 612−622. (45) Zhang, K.; Corsa, C. A.; Ponik, S. M.; Prior, J. L.; PiwnicaWorms, D.; Eliceiri, K. W.; Keely, P. J.; Longmore, G. D. The collagen receptor discoidin domain receptor 2 stabilizes SNAIL1 to facilitate breast cancer metastasis. Nat. Cell Biol. 2013, 15, 677−687. (46) (a) Badiola, I.; Villacé, P.; Basaldua, I.; Olaso, E. Downregulation of discoidin domain receptor 2 in A375 human melanoma cells. Oncol. Rep. 2011, 26, 971−978. (b) Zhang, S.; Bu, X.; Zhao, H.; Yu, J.; Wang, Y.; Li, D.; Zhu, C.; Zhu, T.; Ren, T.; Liu, X.; Yao, L.; Su, J. A host deficiency of discoidin domain receptor 2 (DDR2) inhibits both tumour angiogenesis and metastasis. J. Pathol. 2014, 232, 436− 448. (47) (a) Ding, L.; Getz, G.; Wheeler, D. A.; Mardis, E. R.; McLellan, M. D.; Cibulskis, K.; Sougnez, C.; Greulich, H.; Muzny, D. M.; Morgan, M. B.; Fulton, L.; Fulton, R. S.; Zhang, Q.; Wend, M. C.; Lawrence, M. S.; Larson, D. E.; Chen, K.; Dooling, D. J.; Sabo, A.; Hawes, A. C.; Shen, H.; Jhangiani, S. N.; Lewis, L. R.; Hall, O.; Zhu, Y.; Mathew, T.; Ren, Y.; Yao, J.; Scherer, S. E.; Clerc, K.; Metcalf, G. A.; Ng, B.; Milosavljevic, A.; Gonzalez-Garay, M. L.; Osborne, J. R.; Meyer, R.; Shi, X.; Tang, Y.; Koboldt, D. C.; Lin, L.; Abbott, R.; Miner, T. L.; Pohl, C.; Fewell, G.; Haipek, C.; Schmidt, H.; Dunford-Shore, B. H.; Kraja, A.; Crosby, S. D.; Sawyer, C. S.; Vickery, T.; Sander, S.; Robinson, J.; Winckler, W.; Baldwin, J.; Chirieac, L. R.; Dutt, A.; Fennell, T.; Hanna, M.; Johnson, B. E.; Onofrio, R. C.; Thomas, R. K.; Tonon, G.; Weir, B. A.; Zhao, X.; Ziaugra, L.; Zody, M. C.; Giordano, T.; Orringer, M. B.; Roth, J. A.; Spitz, M. R.; Wistuba, I. I.; Ozenberger, B.; Good, P. J.; Chang, A. C.; Beer, D. G.; Watson, M. A.; Ladanyi, M.; Broderick, S.; Yoshizawa, A.; Travis, W. D.; Pao, W.; Province, M. A.; Weinstock, G. M.; Varmus, H. E.; Gabriel, S. B.; Lander, E. S.; Gibbs, R. A.; Meyerson, M.; Wilson, R. K. Somatic mutations affect key pathways in lung adenocarcinoma. Nature 2008, 455, 1069−1075. (b) Davies, H.; Hunter, C.; Smith, R.; Stephens, P.; Greenman, C.; Bignell, G.; Teague, J.; Butler, A.; Edkins, S.; Stevens, C.; Parker, A.; O’Meara, S.; Avis, T.; Barthorpe, S.; Brackenbury, L.;

Dziechciarkova, M.; Ehrmann, J.; Klein, J.; Fridman, E.; Skarda, J.; Srovnal, J.; Hajduch, M.; Murray, P.; Kolar, Z. Novel markers for differentiation of lobular and ductal invasive breast carcinomas by laser microdissection and microarray analysis. BMC Cancer 2007, 7, 55. (c) Neuhaus, B.; Buhren, S.; Bock, B.; Alves, F.; Vogel, W. F.; Kiefer, F. Migration inhibition of mammary epithelial cells by Syk is blocked in the presence of DDR1 receptors. Cell. Mol. Life Sci. 2011, 68, 3757− 3770. (34) (a) Weiner, H. L.; Rothman, M.; Miller, D. C.; Ziff, E. B. Pediatric brain tumors express multiple receptor tyrosine kinases including novel cell adhesion kinases. Pediatr. Neurosurg. 1996, 25, 64−71. discussion 71−72; (b) Weiner, H. L.; Huang, H.; Zagzag, D.; Boyce, H.; Lichtenbaum, R.; Ziff, E. B. Consistent and selective expression of the discoidin domain receptor-1 tyrosine kinase in human brain tumors. Neurosurgery 2000, 47, 1400−1409. (c) Yamanaka, R.; Arao, T.; Yajima, N.; Tsuchiya, N.; Homma, J.; Tanaka, R.; Sano, M.; Oide, A.; Sekijima, M.; Nishio, K. Identification of expressed genes characterizing long-term survival in malignant glioma patients. Oncogene 2006, 25, 5994−6002. (d) Yoshida, D.; Teramoto, A. Enhancement of pituitary adenoma cell invasion and adhesion is mediated by discoidin domain receptor-1. J. Neuro-Oncol. 2007, 82, 29−40. (35) (a) Squire, J. A.; Bayani, J.; Luk, C.; Unwin, L.; Tokunaga, J.; MacMillan, C.; Irish, J.; Brown, D.; Gullane, P.; Kamel-Reid, S. Molecular cytogenetic analysis of head and neck squamous cell carcinoma: by comparative genomic hybridization, spectral karyotyping, and expression array analysis. Head Neck 2002, 24, 874−887. (b) Hidalgo-Carcedo, C.; Hooper, S.; Chaudhry, S. I.; Williamson, P.; Harrington, K.; Leitinger, B.; Sahai, E. Collective cell migration requires suppression of actomyosin at cell-cell contacts mediated by DDR1 and the cell polarity regulators Par3 and Par6. Nat. Cell Biol. 2011, 13, 49−58. (36) (a) Shen, Q.; Cicinnati, V. R.; Zhang, X.; Iacob, S.; Weber, F.; Sotiropoulos, G. C.; Radtke, A.; Lu, M.; Paul, A.; Gerken, G.; Beckebaum, S. Role of microRNA-199a-5p and discoidin domain receptor 1 in human hepatocellular carcinoma invasion. Mol. Cancer 2010, 9, 227. (b) Gu, T. L.; Deng, X.; Huang, F.; Tucker, M.; Crosby, K.; Rimkunas, V.; Wang, Y.; Deng, G.; Zhu, L.; Tan, Z.; Hu, Y.; Wu, C.; Nardone, J.; MacNeill, J.; Ren, J.; Reeves, C.; Innocenti, G.; Norris, B.; Yuan, J.; Yu, J.; Haack, H.; Shen, B.; Peng, C.; Li, H.; Zhou, X.; Liu, X.; Rush, J.; Comb, M. J. Survey of tyrosine kinase signaling reveals ROS kinase fusions in human cholangiocarcinoma. PLoS One 2011, 6, e15640. (37) Couvelard, A.; Hu, J.; Steers, G.; O’Toole, D.; Sauvanet, A.; Belghiti, J.; Bedossa, P.; Gatter, K.; Ruszniewski, P.; Pezzella, F. Identification of potential therapeutic targets by gene-expression profiling in pancreatic endocrine tumors. Gastroenterol. 2006, 131, 1597−1610. (38) Shimada, K.; Nakamura, M.; Ishida, E.; Higuchi, T.; Yamamoto, H.; Tsujikawa, K.; Konishi, N. Prostate cancer antigen-1 contributes to cell survival and invasion though discoidin receptor 1 in human prostate cancer. Cancer Sci. 2008, 99, 39−45. (39) (a) Hajdu, M.; Singer, S.; Maki, R. G.; Schwartz, G. K.; Keohan, M. L.; Antonescu, C. R. IGF2 over-expression in solitary fibrous tumours is independent of anatomical location and is related to loss of imprinting. J. Pathol. 2010, 221, 300−307. (b) Chiaretti, S.; Li, X.; Gentleman, R.; Vitale, A.; Wang, K. S.; Mandelli, F.; Foà, R.; Ritz, J. Gene expression profiles of B-lineage adult acute lymphocytic leukemia reveal genetic patterns that identify lineage derivation and distinct mechanisms of transformation. Clin. Cancer Res. 2005, 11, 7209−7219. (40) (a) Das, S.; Ongusaha, P. P.; Yang, Y. S.; Park, J.-M.; Aaronson, S. A.; Lee, S. W. Discoidin domain receptor 1 receptor tyrosine kinase induces cyclooxygenase-2 and promotes chemoresistance through nuclear factor-κB pathway activation. Cancer Res. 2006, 66, 8123− 8130. (b) Miao, L.; Zhu, S.; Wang, Y.; Li, Y.; Ding, J.; Dai, J.; Cai, H.; Zhang, D.; Song, Y. Discoidin domain receptor 1 is associated with poor prognosis of non-small cell lung cancer and promotes cell invasion via epithelial-to-mesenchymal transition. Med. Oncol. 2013, M

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry

DDR2 and its lung cancer mutants. Biochem. J. 2013, 454, 501−513. (b) Wall, S. J.; Werner, E.; Werb, Z.; DeClerck, Y. A. Discoidin domain receptor 2 mediates tumor cell cycle arrest induced by fibrillar collagen. J. Biol. Chem. 2005, 280, 40187−40194. (51) (a) Yeung, D.; Chmielewski, D.; Mihai, C.; Agarwal, G. Oligomerization of DDR1 ECD affects receptor-ligand binding. J. Struct. Biol. 2013, 183, 495−500. (b) Fu, H.-L.; Valiathan, R. R.; Payne, L.; Kumarasiri, M.; Mahasenan, K. V.; Mobashery, S.; Huang, P.; Fridman, R. Glycosylation at ASN211 regulates the activation state of the discoidin domain receptor 1 (DDR1). J. Biol. Chem. 2014, DOI: 10.1074/jbc.M113.541102. (52) (a) Ferri, N.; Carragher, N. O.; Raines, E. W. Role of discoidin domain receptors 1 and 2 in human smooth muscle cell-mediated collagen remodeling: potential implications in atherosclerosis and lymphangioleiomyomatosis. Am. J. Pathol. 2004, 164, 1575−1585. (b) Xu, L.; Peng, H.; Wu, D.; Hu, K.; Goldring, M. B.; Olsen, B. R.; Li, Y. Activation of the discoidin domain receptor 2 induces expression of matrix metalloproteinase 13 associated with osteoarthritis in mice. J. Biol. Chem. 2005, 280, 548−555. (53) Avivi-Green, C.; Singal, M.; Vogel, W. F. Discoidin domain receptor 1-deficient mice are resistant to bleomycin-induced lung fibrosis. Am. J. Resp. Crit. Care 2006, 174, 420−427. (54) (a) Gross, O.; Girgert, R.; Beirowski, B.; Kretzler, M.; Kang, H. G.; Kruegel, J.; Miosge, N.; Busse, A. C.; Segerer, S.; Vogel, W. F.; Muller, G. A.; Weber, M. Loss of collagen-receptor DDR1 delays renal fibrosis in hereditary type IV collagen disease. Matrix Biol. 2010, 29, 346−356. (b) Flamant, M.; Placier, S.; Rodenas, A.; Curat, C. A.; Vogel, W. F.; Chatziantoniou, C.; Dussaule, J. C. Discoidin domain receptor 1 null mice are protected against hypertension-induced renal disease. J. Am. Soc. Nephrol. 2006, 17, 3374−3381. (c) Guerrot, D.; Kerroch, M.; Placier, S.; Vandermeersch, S.; Trivin, C.; Mael-Ainin, M.; Chatziantoniou, C.; Dussaule, J. C. Discoidin domain receptor 1 is a major mediator of inflammation and fibrosis in obstructive nephropathy. Am. J. Pathol. 2011, 179, 83−91. (d) Kerroch, M.; Guerrot, D.; Vandermeersch, S.; Placier, S.; Mesnard, L.; Jouanneau, C.; Rondeau, E.; Ronco, P.; Boffa, J. J.; Chatziantoniou, C.; Dussaule, J. C. Genetic inhibition of discoidin domain receptor 1 protects mice against crescentic glomerulonephritis. FASEB J. 2012, 26, 4079−4091. (55) Luo, Z.; Liu, H.; Sun, X.; Guo, R.; Cui, R.; Ma, X.; Yan, M. RNA interference against discoidin domain receptor 2 ameliorates alcoholic liver disease in rats. PLoS One 2013, 8, e55860. (56) Xu, L.; Servais, J.; Polur, I.; Kim, D.; Lee, P. L.; Chung, K.; Li, Y. Attenuation of osteoarthritis progression by reduction of discoidin domain receptor 2 in mice. Arthritis Rheum. 2010, 62, 2736−2744. (57) (a) Lee, J. E.; Kang, C. S.; Guan, X. Y.; Kim, B. T.; Kim, S. H.; Lee, Y. M.; Moon, W. S.; Kim, D. K. Discoidin domain receptor 2 is involved in the activation of bone marrow-derived dendritic cells caused by type I collagen. Biochem. Bioph. Res. Commun. 2007, 352, 244−250. (b) Poudel, B.; Yoon, D.-S.; Lee, J.-H.; Lee, Y.-M.; Kim, D.K. Collagen I enhances functional activities of human monocytederived dendritic cells via discoidin domain receptor 2. Cell. Immunol. 2012, 278, 95−102. (c) Poudel, B.; Ki, H.-H.; Lee, Y.-M.; Kim, D.-K. Induction of IL-12 production by the activation of discoidin domain receptor 2 via NF-κB and JNK pathway. Biochem. Bioph. Res. Commun. 2013, 434, 584−588. (58) Sun, X.; Phan, T. N.; Jung, S. H.; Kim, S. Y.; Cho, J. U.; Lee, H.; Woo, S. H.; Park, T. K.; Yang, B. S. LCB 03-0110, a novel pandiscoidin domain receptor/c-Src family tyrosine kinase inhibitor, suppresses scar formation by inhibiting fibroblast and macrophage activation. J. Pharmacol. Exp. Ther. 2012, 340, 510−519. (59) Siddiqui, K.; Kim, G. W.; Lee, D. H.; Shin, H. R.; Yang, E. G.; Lee, N. T.; Yang, B. S. Actinomycin D identified as an inhibitor of discoidin domain receptor 2 interaction with collagen through an insect cell based screening of a drug compound library. Biol. Pharm. Bull. 2009, 32, 136−141. (60) Castro-Sanchez, L.; Soto-Guzman, A.; Navarro-Tito, N.; Martinez-Orozco, R.; Salazar, E. P. Native type IV collagen induces cell migration through a CD9 and DDR1-dependent pathway in MDA-MB-231 breast cancer cells. Eur. J. Cell. Biol. 2010, 89, 843−852.

Buck, G.; Clements, J.; Cole, J.; Dicks, E.; Edwards, K.; Forbes, S.; Gorton, M.; Gray, K.; Halliday, K.; Harrison, R.; Hills, K.; Hinton, J.; Jones, D.; Kosmidou, V.; Laman, R.; Lugg, R.; Menzies, A.; Perry, J.; Petty, R.; Raine, K.; Shepherd, R.; Small, A.; Solomon, H.; Stephens, Y.; Tofts, C.; Varian, J.; Webb, A.; West, S.; Widaa, S.; Yates, A.; Brasseur, F.; Cooper, C. S.; Flanagan, A. M.; Green, A.; Knowles, M.; Leung, S. Y.; Looijenga, L. H.; Malkowicz, B.; Pierotti, M. A.; Teh, B. T.; Yuen, S. T.; Lakhani, S. R.; Easton, D. F.; Weber, B. L.; Goldstraw, P.; Nicholson, A. G.; Wooster, R.; Stratton, M. R.; Futreal, P. A. Somatic mutations of the protein kinase gene family in human lung cancer. Cancer Res. 2005, 65, 7591−7595. (c) Hammerman, P. S.; Sos, M. L.; Ramos, A. H.; Xu, C.; Dutt, A.; Zhou, W.; Brace, L. E.; Woods, B. A.; Lin, W.; Zhang, J.; Deng, X.; Lim, S. M.; Heynck, S.; Peifer, M.; Simard, J. R.; Lawrence, M. S.; Onofrio, R. C.; Salvesen, H. B.; Seidel, D.; Zander, T.; Heuckmann, J. M.; Soltermann, A.; Moch, H.; Koker, M.; Leenders, F.; Gabler, F.; Querings, S.; Ansen, S.; Brambilla, E.; Brambilla, C.; Lorimier, P.; Brustugun, O. T.; Helland, A.; Petersen, I.; Clement, J. H.; Groen, H.; Timens, W.; Sietsma, H.; Stoelben, E.; Wolf, J.; Beer, D. G.; Tsao, M. S.; Hanna, M.; Hatton, C.; Eck, M. J.; Janne, P. A.; Johnson, B. E.; Winckler, W.; Greulich, H.; Bass, A. J.; Cho, J.; Rauh, D.; Gray, N. S.; Wong, K. K.; Haura, E. B.; Thomas, R. K.; Meyerson, M. Mutations in the DDR2 kinase gene identify a novel therapeutic target in squamous cell lung cancer. Cancer Discovery 2011, 1, 78−89. (d) Miao, L.; Wang, Y.; Zhu, S.; Shi, M.; Li, Y.; Ding, J.; Yang, J.; Ye, Q.; Cai, H.; Zhang, D.; Liu, H.; Song, Y. Identification of novel driver mutations of the discoidin domain receptor 2 (DDR2) gene in squamous cell lung cancer of Chinese patients. BMC Cancer 2014, 24, 369 DOI: 10.1186/1471-2407-14-369. (e) Drilon, A.; Rekhtman, N.; Ladanyi, M.; Paik, P. Squamous-cell carcinomas of the lung: emerging biology, controversies, and the promise of targeted therapy. Lancet Oncol. 2012, 13, e418−426. (f) Sos, M. L.; Thomas, R. K. Genetic insight and therapeutic targets in squamous-cell lung cancer. Oncogene 2012, 31, 4811−4814. (48) (a) Loriaux, M. M.; Levine, R. L.; Tyner, J. W.; Frohling, S.; Scholl, C.; Stoffregen, E. P.; Wernig, G.; Erickson, H.; Eide, C. A.; Berger, R.; Bernard, O. A.; Griffin, J. D.; Stone, R. M.; Lee, B.; Meyerson, M.; Heinrich, M. C.; Deininger, M. W.; Gilliland, D. G.; Druker, B. J. High-throughput sequence analysis of the tyrosine kinome in acute myeloid leukemia. Blood 2008, 111, 4788−4796. (b) Tomasson, M. H.; Xiang, Z.; Walgren, R.; Zhao, Y.; Kasai, Y.; Miner, T.; Ries, R. E.; Lubman, O.; Fremont, D. H.; McLellan, M. D.; Payton, J. E.; Westervelt, P.; DiPersio, J. F.; Link, D. C.; Walter, M. J.; Graubert, T. A.; Watson, M.; Baty, J.; Heath, S.; Shannon, W. D.; Nagarajan, R.; Bloomfield, C. D.; Mardis, E. R.; Wilson, R. K.; Ley, T. J. Somatic mutations and germline sequence variants in the expressed tyrosine kinase genes of patients with de novo acute myeloid leukemia. Blood 2008, 111, 4797−4808. (49) (a) Pitini, V.; Arrigo, C.; Di Mirto, C.; Mondello, P.; Altavilla, G. Response to dasatinib in a patient with SQCC of the lung harboring a discoid-receptor-2 and synchronous chronic myelogenous leukemia. Lung Cancer 2013, 82, 171−172. (b) Haura, E. B.; Tanvetyanon, T.; Chiappori, A.; Williams, C.; Simon, G.; Antonia, S.; Gray, J.; Litschauer, S.; Tetteh, L.; Neuger, A.; Song, L.; Rawal, B.; Schell, M. J.; Bepler, G. Phase I/II study of the Src inhibitor dasatinib in combination with erlotinib in advanced non-small-cell lung cancer. J. Clin. Oncol. 2010, 28, 1387−1394. (c) Johnson, F. M.; Bekele, B. N.; Feng, L.; Wistuba, I.; Tang, X. M.; Tran, H. T.; Erasmus, J. J.; Hwang, L. L.; Takebe, N.; Blumenschein, G. R.; Lippman, S. M.; Stewart, D. J. Phase II study of dasatinib in patients with advanced non-small-cell lung cancer. J. Clin. Oncol. 2010, 28, 4609−4615. (d) Johnson, M. L.; Riely, G. J.; Rizvi, N. A.; Azzoli, C. G.; Kris, M. G.; Sima, C. S.; Ginsberg, M. S.; Pao, W.; Miller, V. A. Phase II trial of dasatinib for patients with acquired resistance to treatment with the epidermal growth factor receptor tyrosine kinase inhibitors erlotinib or gefitinib. J. Thorac. Oncol. 2011, 6, 1128−1131. (50) (a) Iwai, L. K.; Payne, L. S.; Luczynski, M. T.; Chang, F.; Xu, H.; Clinton, R. W.; Paul, A.; Esposito, E. A.; Gridley, S.; Leitinger, B.; Naegle, K. M.; Huang, P. H. Phosphoproteomics of collagen receptor networks reveals SHP-2 phosphorylation downstream of wild-type N

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX

Perspective

Journal of Medicinal Chemistry (61) (a) Foehr, E. D.; Tatavos, A.; Tanabe, E.; Raffioni, S.; Goetz, S.; Dimarco, E.; De Luca, M.; Bradshaw, R. A. Discoidin domain receptor 1 (DDR1) signaling in PC12 cells: activation of juxtamembrane domains in PDGFR/DDR/TrkA chimeric receptors. FASEB J. 2000, 14, 973−981. (b) Ikeda, K.; Wang, L. H.; Torres, R.; Zhao, H.; Olaso, E.; Eng, F. J.; Labrador, P.; Klein, R.; Lovett, D.; Yancopoulos, G. D.; Friedman, S. L.; Lin, H. C. Discoidin domain receptor 2 interacts with Src and Shc following its activation by type I collagen. J. Biol. Chem. 2002, 277, 19206−19212. (c) Yang, K.; Kim, J. H.; Kim, H. J.; Park, I. S.; Kim, I. Y.; Yang, B. S. Tyrosine 740 phosphorylation of discoidin domain receptor 2 by Src stimulates intramolecular autophosphorylation and Shc signaling complex formation. J. Biol. Chem. 2005, 280, 39058−39066. (62) L’Hote, C. G.; Thomas, P. H.; Ganesan, T. S. Functional analysis of discoidin domain receptor 1: effect of adhesion on DDR1 phosphorylation. FASEB J. 2002, 16, 234−236. (63) (a) Schenone, S.; Bruno, O.; Radi, M.; Botta, M. New insights into small-molecule inhibitors of Bcr-Abl. Med. Res. Rev. 2011, 31, 1− 41. (b) Lu, X. Y.; Cai, Q.; Ding, K. Recent developments in the third generation inhibitors of Bcr-Abl for overriding T315I mutation. Curr. Med. Chem. 2011, 18, 2146−2157. (c) Breccia, M.; Alimena, G. How to treat CML patients in the tyrosine kinase inhibitors era? From imatinib standard dose to second generation drugs front-line: Unmet needs, pitfalls and advantages. Cancer Lett. 2012, 322, 127−132. (64) (a) Ou, S. H. Second-generation irreversible epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs): a better mousetrap? A review of the clinical evidence. Crit. Rev. Oncol. Hemat. 2012, 83, 407−421. (b) Yang, X. W.; Yang, K.; Kuang, K. Y. The efficacy and safety of EGFR inhibitor monotherapy in non-small cell lung cancer: a systematic review. Curr. Oncol. Rep. 2014, 16, 390. (65) Lou, Y.; Owens, T. D.; Kuglstatter, A.; Kondru, R. K.; Goldstein, D. M. Bruton’s tyrosine kinase inhibitors: approaches to potent and selective inhibition, preclinical and clinical evaluation for inflammatory diseases and B cell malignancies. J. Med. Chem. 2012, 55, 4539−4550. (66) Davis, M. I.; Hunt, J. P.; Herrgard, S.; Ciceri, P.; Wodicka, L. M.; Pallares, G.; Hocker, M.; Treiber, D. K.; Zarrinkar, P. P. Comprehensive analysis of kinase inhibitor selectivity. Nat. Biotechnol. 2011, 29, 1046−1051. (67) Gao, M.; Duan, L.; Luo, J.; Zhang, L.; Lu, X.; Zhang, Y.; Zhang, Z.; Tu, Z.; Xu, Y.; Ren, X.; Ding, K. Discovery and optimization of 3(2-(Pyrazolo[1,5-a]pyrimidin-6-yl)ethynyl)benzamides as novel selective and orally bioavailable discoidin domain receptor 1 (DDR1) inhibitors. J. Med. Chem. 2013, 56, 3281−3295. (68) (a) Rix, U.; Hantschel, O.; Durnberger, G.; Remsing Rix, L. L.; Planyavsky, M.; Fernbach, N. V.; Kaupe, I.; Bennett, K. L.; Valent, P.; Colinge, J.; Kocher, T.; Superti-Furga, G. Chemical proteomic profiles of the Bcr-Abl inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinase targets. Blood 2007, 110, 4055−4063. (b) Li, J.; Rix, U.; Fang, B.; Bai, Y.; Edwards, A.; Colinge, J.; Bennett, K. L.; Gao, J.; Song, L.; Eschrich, S.; Superti-Furga, G.; Koomen, J.; Haura, E. B. A chemical and phosphoproteomic characterization of dasatinib action in lung cancer. Nat. Chem. Biol. 2010, 6, 291−299. (69) (a) Bantscheff, M.; Eberhard, D.; Abraham, Y.; Bastuck, S.; Boesche, M.; Hobson, S.; Mathieson, T.; Perrin, J.; Raida, M.; Rau, C.; Reader, V.; Sweetman, G.; Bauer, A.; Bouwmeester, T.; Hopf, C.; Kruse, U.; Neubauer, G.; Ramsden, N.; Rick, J.; Kuster, B.; Drewes, G. Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat. Biotechnol. 2007, 25, 1035−1044. (b) Rix, U.; Remsing Rix, L. L.; Terker, A. S.; Fernbach, N. V.; Hantschel, O.; Planyavsky, M.; Breitwieser, F. P.; Herrmann, H.; Colinge, J.; Bennett, K. L.; Augustin, M.; Till, J. H.; Heinrich, M. C.; Valent, P.; Superti-Furga, G. A comprehensive target selectivity survey of the BCR-ABL kinase inhibitor INNO-406 by kinase profiling and chemical proteomics in chronic myeloid leukemia cells. Leukemia 2010, 24, 44−50. (c) Ren, X.; Pan, X.; Zhang, Z.; Wang, D.; Lu, X.; Li, Y.; Wen, D.; Long, H.; Luo, J.; Feng, Y.; Zhuang, X.; Zhang, F.; Liu, J.; Leng, F.; Lang, X.; Bai, Y.; She, M.; Tu, Z.; Pan, J.; Ding, K. Identification of GZD824 as an orally bioavailable inhibitor that targets phosphorylated and nonphosphorylated breakpoint cluster region−

abelson (Bcr-Abl) kinase and overcomes clinically acquired mutationinduced resistance against imatinib. J. Med. Chem. 2013, 56, 879−894. (70) For more information about this assay, see www.discoverx.com. (71) Beauchamp, E. M.; Woods, B. A.; Dulak, A. M.; Tan, L.; Xu, C.; Gray, N. S.; Bass, A. J.; Wong, K. K.; Meyerson, M.; Hammerman, P. S. Acquired resistance to dasatinib in lung cancer cell lines conferred by DDR2 gatekeeper mutation and NF1 loss. Mol. Cancer. Ther. 2013, 13, 475−482. (72) Takeshi, M.; Satoshi, N.; Sousuke, H.; Hatsuo, K.; Kihito, H.; Hideaki, S.; Hiroshi, T.; Yoshito, N. Benzamide derivative. WO2013161851, 2013. (73) Takeshi, M.; Hatsuo, K.; Satoshi, N.; Sousuke, H.; Kihito, H.; Hideaki, S.; Hiroshi, T.; Toshiyuki, M. Quinazolinedione derivative. WO2013161853, 2013. (74) Wucherer-Plietker, M.; Werkmann, D.; Gigout, A.; Kuhn, D.; Sawatzky, E. New substituted aryl compounds are discoidin domain receptor tyrosine kinase 2 inhibitors used to treat e.g. osteoarthritis, hepatocirrhosis, traumatic cartilage injuries, allodynia, hyperalgesia, Alzheimer’s disease, scarring and rickets. WO2014032755, 2014. (75) Hu, Y.; Potts, M. B.; Colosimo, D.; Herrera-Herrera, M. L.; Legako, A. G.; Yousufuddin, M.; White, M. A.; MacMillan, J. B.; Discoipyrroles, A.-D. isolation, structure determination, and synthesis of potent migration inhibitors from Bacillus hunanensis. J. Am. Chem. Soc. 2013, 135, 13387−13392. (76) Aguilera, K. Y.; Goldstein, C. D.; Bradshaw, A. D.; Ding, K.; Brekken, R. A., SPARC as a regulator of collagen signaling in pancreatic cancer. Program Book, ASMB 2014 Biennial Meeting, Cleveland, OH, Oct 12−15, 2014; ASMB: Bethesda, MD, 2014; Abstract 137.

O

DOI: 10.1021/jm5012319 J. Med. Chem. XXXX, XXX, XXX−XXX