scispace - formally typeset
Search or ask a question

Showing papers by "Axel Ullrich published in 2006"


Journal ArticleDOI
TL;DR: The structural features of the kinase domain and the unique polo-box domain ofPLK1 that are most suited for drug development are addressed and the current understanding of the therapeutic potential of PLK1 is discussed.
Abstract: Human polo-like kinase 1 (PLK1) is essential during mitosis and in the maintenance of genomic stability. PLK1 is overexpressed in human tumours and has prognostic potential in cancer, indicating its involvement in carcinogenesis and its potential as a therapeutic target. The use of different PLK1 inhibitors has increased our knowledge of mitotic regulation and allowed us to assess their ability to suppress tumour growth in vivo. We address the structural features of the kinase domain and the unique polo-box domain of PLK1 that are most suited for drug development and discuss our current understanding of the therapeutic potential of PLK1.

799 citations


Journal ArticleDOI
TL;DR: The findings implicate Axl in gliomagenesis and validate it as a promising target for the development of approaches toward a therapy of these highly aggressive but, as yet, therapy-refractory, tumors.
Abstract: Malignant gliomas remain incurable brain tumors because of their diffuse-invasive growth. So far, the genetic and molecular events underlying gliomagenesis are poorly understood. In this study, we have identified the receptor tyrosine kinase Axl as a mediator of glioma growth and invasion. We demonstrate that Axl and its ligand Gas6 are overexpressed in human glioma cell lines and that Axl is activated under baseline conditions. Furthermore, Axl is expressed at high levels in human malignant glioma. Inhibition of Axl signaling by overexpression of a dominant-negative receptor mutant (AXL-DN) suppressed experimental gliomagenesis (growth inhibition >85%, P 72 days). A detailed analysis of the distinct hallmarks of glioma pathology, such as cell proliferation, migration, and invasion and tumor angiogenesis, revealed that inhibition of Axl signaling interfered with cell proliferation (inhibition 30% versus AXL-WT), glioma cell migration (inhibition 90% versus mock and AXL-WT, P < 0.05), and invasion (inhibition 62% and 79% versus mock and AXL-WT, respectively; P < 0.05). This study describes the identification, functional manipulation, in vitro and in vivo validation, and preclinical therapeutic inhibition of a target receptor tyrosine kinase mediating glioma growth and invasion. Our findings implicate Axl in gliomagenesis and validate it as a promising target for the development of approaches toward a therapy of these highly aggressive but, as yet, therapy-refractory, tumors.

265 citations


Journal ArticleDOI
TL;DR: Deletion of the mouse gene encoding Mig6 is shown to cause hyperactivation of endogenous epidermal growth factor receptor (EGFR) and sustained signaling through the mitogen-activated protein kinase (MAPK) pathway, resulting in overproliferation and impaired differentiation of Epidermal keratinocytes, indicating a tumor-suppressive role for Mig6.
Abstract: The growing number of recently identified negative feedback regulators of receptor tyrosine kinases (RTKs) highlights the importance of signal attenuation and modulation for correct signaling outcome. Mitogen-inducible gene 6 (Mig6 also known as RALT or Gene 33) is a multiadaptor protein thought to be involved in the regulation of RTK and stress signaling. Here, we show that deletion of the mouse gene encoding Mig6 (designated Errfi1, which stands for ERBB receptor feedback inhibitor 1) causes hyperactivation of endogenous epidermal growth factor receptor (EGFR) and sustained signaling through the mitogen-activated protein kinase (MAPK) pathway, resulting in overproliferation and impaired differentiation of epidermal keratinocytes. Furthermore, Errfi1-/- mice develop spontaneous tumors in various organs and are highly susceptible to chemically induced formation of skin tumors. A tumor-suppressive role for Mig6 is supported by our finding that MIG6 is downregulated in various human cancers. Inhibition of endogenous Egfr signaling with the Egfr inhibitor gefitinib (Iressa) or replacement of wild-type Egfr with the kinase-deficient protein encoded by the hypomorphic Egfr(wa2) allele completely rescued skin defects in Erffi1-/- mice. Carcinogen-induced tumors displayed by Errfi1-/- mice were highly sensitive to gefitinib. These results indicate that Mig6 is a specific negative regulator of Egfr signaling in skin morphogenesis and is a novel tumor suppressor of Egfr-dependent carcinogenesis.

250 citations


Journal ArticleDOI
TL;DR: Structural‐based mutagenesis, protein binding assays and receptor activation experiments demonstrate that both the major and minor Gas6 binding sites are required for productive transmembrane signalling.
Abstract: Receptor tyrosine kinases of the Axl family are activated by the vitamin K‐dependent protein Gas6. Axl signalling plays important roles in cancer, spermatogenesis, immunity, and platelet function. The crystal structure at 3.3 A resolution of a minimal human Gas6/Axl complex reveals an assembly of 2:2 stoichiometry, in which the two immunoglobulin‐like domains of the Axl ectodomain are crosslinked by the first laminin G‐like domain of Gas6, with no direct Axl/Axl or Gas6/Gas6 contacts. There are two distinct Gas6/Axl contacts of very different size, both featuring interactions between edge β‐strands. Structure‐based mutagenesis, protein binding assays and receptor activation experiments demonstrate that both the major and minor Gas6 binding sites are required for productive transmembrane signalling. Gas6‐mediated Axl dimerisation is likely to occur in two steps, with a high‐affinity 1:1 Gas6/Axl complex forming first. Only the minor Gas6 binding site is highly conserved in the other Axl family receptors, Sky/Tyro3 and Mer. Specificity at the major contact is suggested to result from the segregation of charged and apolar residues to opposite faces of the newly formed β‐sheet.

244 citations


Journal ArticleDOI
TL;DR: It is demonstrated that E2-induced EGFR transactivation in human breast cancer cells is driven via a novel signaling system controlled by the lipid kinase sphingosine kinase-1 (SphK1), which reveals a key role for SphK1 in the coupling of the signals between three membrane-spanning events induced by E2, S1P, and EGF.
Abstract: The transactivation of enhanced growth factor receptor (EGFR) by G protein–coupled receptor (GPCR) ligands is recognized as an important signaling mechanism in the regulation of complex biological processes, such as cancer development. Estrogen (E2), which is a steroid hormone that is intimately implicated in breast cancer, has also been suggested to function via EGFR transactivation. In this study, we demonstrate that E2-induced EGFR transactivation in human breast cancer cells is driven via a novel signaling system controlled by the lipid kinase sphingosine kinase-1 (SphK1). We show that E2 stimulates SphK1 activation and the release of sphingosine 1-phosphate (S1P), by which E2 is capable of activating the S1P receptor Edg-3, resulting in the EGFR transactivation in a matrix metalloprotease–dependent manner. Thus, these findings reveal a key role for SphK1 in the coupling of the signals between three membrane-spanning events induced by E2, S1P, and EGF. They also suggest a new signal transduction model across three individual ligand-receptor systems, i.e., “criss-cross” transactivation.

222 citations


Journal ArticleDOI
TL;DR: Her3 may represent a new prognostic factor in primary epithelial ovarian cancer and exploration of therapeutic strategies to block HER3 could be warranted.
Abstract: Background HER3 (erbB-3) is a member of the epidermal growth factor receptor (EGFR) family. After dimerization with other members of the EGFR family several signal transduction cascades can be activated, including phosphoinosite 3-kinase (PI3-K)/Akt and extracellular signal-regulated kinase (ERK1/2). Here, we studied a possible association between HER3 expression and prognosis in patients with ovarian cancer. Methods Tumor tissue of 116 consecutive patients diagnosed with primary epithelial ovarian cancer between 1986 and 1995 was analyzed immunohistochemically for HER3 expression. A possible influence of HER3 expression on survival was studied by multivariate Cox regression adjusting for established clinical prognostic factors. Results A positive HER3 expression was observed in 53.4% of the patients. HER3 expression was associated with decreased survival in proportional hazard modeling, including the International Federation of Gynecology and Obstetrics (FIGO) stage, histologic grade and type, residual disease, and age. After likelihood ratio forward as well as backward selection, only HER3 expression (hazard ratio, 1.71; 95% CI, 1.10 to 2.67; P .018), FIGO stage (hazard ratio, 4.78; 95% CI, 1.89 to 12.08; P .001), residual tumor (hazard ratio, 2.69; 95% CI, 1.40 to 5.17; P .003), and age (hazard ratio, 2.06; 95% CI, 1.17 to 3.65; P .013) were found to be significant. Kaplan-Meier plots demonstrated a clear influence of HER3 expression on survival time. Median survival time was 3.31 years (95% CI, 1.93 to 4.68) for patients with low HER3 expression, compared with only 1.80 years (95% CI, 0.83 to 2.78) for patients with HER3 overexpression (log-rank test P .0034).

209 citations


Journal ArticleDOI
TL;DR: It is demonstrated that endothelial cells of human malignant brain tumors also express guidance molecules, such as EphB4 and its ligand ephrinB2, which implies that both neoplastic and non‐neoplastic vascularization is driven not only by a vascular initiation program but also by aascular patterning program mediated by guidance molecules.
Abstract: Guidance molecules have attracted interest by demonstration that they regulate patterning of the blood vascular system during development. However, their significance during postnatal angiogenesis has remained unknown. Here, we demonstrate that endothelial cells of human malignant brain tumors also express guidance molecules, such as EphB4 and its ligand ephrinB2. To study their function, EphB4 variants were overexpressed in blood vessels of tumor xenografts. Our studies revealed that EphB4 acts as a negative regulator of blood vessel branching and vascular network formation, switching the vascularization program from sprouting angiogenesis to circumferential vessel growth. In parallel, EphB4 reduces the permeability of the tumor vascular system via activation of the angiopoietin-1/Tie2 system at the endothelium/pericyte interface. Furthermore, overexpression of EphB4 variants in blood vessels during (i) vascularization of non-neoplastic cell grafts and (ii) retinal vascularization revealed that these functions of EphB4 apply to postnatal, non-neoplastic angiogenesis in general. This implies that both neoplastic and non-neoplastic vascularization is driven not only by a vascular initiation program but also by a vascular patterning program mediated by guidance molecules.

158 citations


Journal ArticleDOI
TL;DR: The results show that the previously found association of FGFR4 Arg388 genotype with breast cancer progression is strongest in patients with adjuvant systemic therapy, particularly chemotherapy, and thus may reflect therapy resistance.
Abstract: Purpose A recent study presented first evidence that a single nucleotide polymorphism (SNP) at codon 388 of fibroblast growth factor receptor 4 (FGFR4) gene, causing a transmembrane domain missense mutation (Gly388Arg), is associated with disease outcome in node-positive breast cancer. This article addresses the clinical relevance of this SNP, FGFR4 genotype, phenotype, and HER2 regarding patient outcome and influence of adjuvant systemic therapy in a substantial primary breast cancer collective (n = 372; median follow-up, 94.5 months). Methods Polymerase chain reaction restriction fragment length polymorphism analysis of germ-line polymorphism was performed in uninvolved lymph nodes; FGFR4 and HER2 expression were assessed immunohistochemically in tissue microarrays. Results In 51% of patients, homo- or heterozygous Arg388 allele was present. No correlation existed between FGFR4 genotype and expression or HER2 status. In node-negative patients, FGFR4 genotype was not correlated with disease outcome. In n...

139 citations


Journal ArticleDOI
TL;DR: In this article, the MAM family prototype protein tyrosine phosphatases (RPTPs) are cleaved by three proteases: furin, ADAM 10, and -secretase.
Abstract: Several receptor protein tyrosine phosphatases (RPTPs) are cell adhesion molecules involved in homophilic interactions, suggesting that RPTP outside-in signaling is coupled to cell contact formation. However, little is known about the mechanisms by which cell density regulates RPTP function. We show that the MAM family prototype RPTP is cleaved by three proteases: furin, ADAM 10, and -secretase. Cell density promotes ADAM 10-mediated cleavage and shedding of RPTP. This is followed by -secretase-dependent intramembrane proteolysis of the remaining transmembrane part to release the phosphatase intracellular portion (PIC) from the membrane, thereby allowing its translocation to the nucleus. When cells were treated with leptomycin B, a nuclear export inhibitor, PIC accumulated in nuclear bodies. PIC is an active protein tyrosine phosphatase that binds to and dephosphorylates -catenin, an RPTP substrate. The expression of RPTP suppresses -catenin’s transcriptional activity, whereas the expression of PIC increases it. Notably, this increase required the phosphatase activity of PIC. Thus, both isoforms have acquired opposing roles in the regulation of -catenin signaling. We also found that RPTP, another MAM family member, undergoes -secretasedependent processing. Our results identify intramembrane proteolysis as a regulatory switch in RPTP signaling and implicate PIC in the activation of -catenin-mediated transcription. The phosphorylation of cellular proteins on tyrosine residues is reversible and regulated by the coordinated and competing actions of two enzyme families: protein tyrosine kinases and protein tyrosine phosphatases (PTPs). The PTP family is structurally diverse and includes both receptor-like and cytoplasmic enzymes. The majority of the receptor PTPs (RPTPs) contain two catalytic domains: a membrane-proximal domain (D1), which is responsible for mainly catalysis, and a membrane-distal domain (D2), which contains little or no phosphatase activity (45). The extracellular (E) portion exhibits broad structural variation. The MAM (meprin/A5 glycoprotein/PTPmu) family of RPTPs, including RPTP ,- ,- ,- , and PCP-2, are characterized by the presence of the MAM domain at their N termini (3, 6, 18, 29, 46). Additional structural features of the extracellular portions involve one immu

108 citations


Journal ArticleDOI
TL;DR: A novel, phosphotyrosine‐dependent fine‐tuning mechanism of Par3 in epithelial TJ assembly controlled by the EGF receptor‐SFK signaling pathway is suggested.
Abstract: The conserved polarity complex, comprising the partitioning-defective (Par) proteins Par3 and Par6, and the atypical protein kinase C, functions in various cell-polarization events and asymmetric cell divisions. However, little is known about whether and how external stimuli-induced signals may regulate Par3 function in epithelial cell polarity. Here, we found that Par3 was tyrosine phosphorylated through phosphoproteomic profiling of pervanadate-induced phosphotyrosine proteins. We also demonstrated that the tyrosine phosphorylation event induced by multiple growth factors including epidermal growth factor (EGF) was dependent on activation of Src family kinase (SFK) members c-Src and c-Yes. The tyrosine residue 1127 (Y1127) of Par3 was identified as the major EGF-induced phosphorylation site. Moreover, we found that Y1127 phosphorylation reduced the association of Par3 with LIM kinase 2 (LIMK2), thus enabling LIMK2 to regulate cofilin phosphorylation dynamics. Substitution of Y1127 for phenylalanine impaired the EGF-induced Par3 and LIMK2 dissociation and delayed epithelial tight junction (TJ) assembly considerably. Collectively, these data suggest a novel, phosphotyrosine-dependent fine-tuning mechanism of Par3 in epithelial TJ assembly controlled by the EGF receptor-SFK signaling pathway.

87 citations


Journal ArticleDOI
TL;DR: The Arg388 genotype and the protein expression of FGFR4 may be potential markers for progression of melanoma.
Abstract: A single nucleotide polymorphism in the gene for FGFR4 (−Arg388) has been associated with progression in various types of human cancer. Although fibroblast growth factors (FGFs) belong to the most important growth factors in melanoma, expression of FGF receptor subtype 4 has not been investigated yet. In this study, the protein expression of this receptor was analysed in 137 melanoma tissues of different progression stages by immunohistochemistry. FGFR4 protein was expressed in 45% of the specimens and correlated with pTNM tumour stages (UICC, P=0.023 and AJCC, P=0.046), presence of microulceration (P=0.009), tumour vascularity (P=0.001), metastases (P=0.025), number of primary tumours (P=0.022), overall survival (P=0.047) and disease-free survival (P=0.024). Furthermore, FGFR4 Arg388 polymorphism was analysed in 185 melanoma patients by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The Arg388 allele was detected in 45% of the melanoma patients and was significantly associated with tumour thickness (by Clark's level of invasion (P=0.004) and by Breslow in mm (P=0.02)) and the tumour subtype nodular melanoma (P=0.002). However, there was no correlation of the FGFR4 Arg388 allele with overall and disease-free survival. In conclusion, the Arg388 genotype and the protein expression of FGFR4 may be potential markers for progression of melanoma.

Journal ArticleDOI
TL;DR: It is concluded that FGFR4 G388 suppresses cell motility of invasive breast cancer cells by altering signalling pathways and the expression of genes that are required for metastasis.


Journal ArticleDOI
TL;DR: It is suggested that treatment with ET‐1 and EGF together induce a more potent anti‐adipogenic response, involving increased Erk1/2 phosphorylation and biphasic attenuation of Aktosphorylation.

Journal ArticleDOI
TL;DR: It is demonstrated that despite m EGFR expression in thymus, strong DTH response was induced by inoculation of mice with the mEGFR‐ECD, which stimulated a potent antimetastatic effect in the EGFR+ Lewis lung carcinoma model, while reproduction‐associated side effects were absent.
Abstract: The epidermal growth factor receptor (EGFR) plays a central role in regulating neoplastic processes. The EGFR overexpression in many human epithelial tumors has been correlated with disease progression and bad prognosis. Passive EGFR-directed immunotherapy, but not active specific approaches, has already been introduced in medical oncology practice. Then we wonder if mice immunization with the extracellular domain of murine EGFR (mEGFR-ECD) in adjuvants can circumvent tolerance to self EGFR, by inducing an immune response with consequent antitumor effect. The present study demonstrated that despite mEGFR expression in thymus, strong DTH response was induced by inoculation of mice with the mEGFR-ECD. This self-immunization, using both CFA and very small sized proteoliposomes from Neisseria meningitidis (VSSP), promoted highly specific IgG titers, predominantly IgG2a and IgG2b. Sera from mice immunized with mEGFR-ECD/VSSP not only recognized EGFR+ tumor cell lines by FACS, but also inhibited their in vitro growth, even in the absence of complement. Noteworthy, vaccination of mice with mEGFR-ECD/VSSP stimulated a potent antimetastatic effect in the EGFR+ Lewis lung carcinoma model, while reproduction-associated side effects were absent. Curiously, mice immunized with the human EGFR-ECD (Her1-ECD) in VSSP though induced highly specific IgG antibodies with strong in vitro cytotoxic effect over EGFR+ human cell lines, showed low cross-reactivity with the mEGFR-ECD. These results further encouraged the development of the Her1-ECD/VSSP vaccine project for patients with EGFR+ tumors. © 2006 Wiley-Liss, Inc.

Journal ArticleDOI
TL;DR: The epidermal growth factor receptor (EGFR) is identified as both an association partner of PTP-LAR, that mediates phosphorylation of the latter, as well as an inducer of LAR-cleavage, described as a means of regulating phosphatase activity downstream and thus under the control of EGFR-mediated signaling pathways.

Journal ArticleDOI
TL;DR: Functional characterization of the Ile322 and Ala573 PTP-PEST mutants revealed an enhancement of in vitro phosphatase activity, whereas the Lys709 variant showed reduced catalytic activity, and underscore the need for further characterizing PTP -PEST and its signaling pathways in context of this disease.

Book ChapterDOI
TL;DR: Methods to investigate GPCR-stimulated EGFR signal transactivation allowing the identification of both the EGF-like ligands and the metalloproteinases involved are described.
Abstract: Interreceptor cross-talk has emerged as a general concept in cellular signaling cascades Therein epidermal growth factor receptor (EGFR) signal transactivation represents the so far best investigated cross-talk mechanism comprising heterogeneous receptor families In this signaling process G protein-coupled receptor (GPCR) stimulation induces phosphorylation of the EGFR, combining the broad diversity of GPCRs with the potent signaling capacities of this receptor tyrosine kinase Early reports attributed this transactivation mechanism to solely intracellular pathways as no EGF-like ligands could be detected in conditioned media of GPCR agonist-stimulated cells However, Prenzel and colleagues demonstrated the involvement of metalloproteinase-mediated release of EGF-like ligands as the predominant mechanism of EGFR signal transactivation, providing a point of convergence for different intracellular effector proteins Since this discovery, numerous investigations revealed the broad relevance of metalloproteinase-mediated ligand-dependent EGFR signal transactivation for coupling GPCRs to various cellular signaling responses Here we describe methods to investigate GPCR-stimulated EGFR signal transactivation allowing the identification of both the EGF-like ligands and the metalloproteinases involved

Journal ArticleDOI
24 Apr 2006-Oncogene
TL;DR: It is demonstrated that CBP functions as a negative regulator of cell transformation and tumor cell growth through downregulation of Src activation, suggesting that Cbp might be broadly involved in RTKs-activated signaling pathways and tumorigenesis.
Abstract: Epidermal growth factor receptor (EGFR) and Src tyrosine kinase cooperate in regulating EGFR-mediated cell signaling and promoting cell transformation and tumorigenesis in pathological conditions. Activation of Src is tightly regulated by the C-terminal Src kinase (Csk). The Csk-binding protein (Cbp) is a ubiquitously expressed transmembrane protein. Its functions include suppression of T-cell receptor activation through recruiting Csk and inhibiting Src family kinase (SFK). However, a potential role of Cbp in EGF-induced cell activities has not been investigated. Here, we report that EGF-stimulation-induced Cbp tyrosine phosphorylation followed by Cbp-Csk association, in a SFK-dependent manner. Expression of wild-type (wt) Cbp remarkably suppressed EGF-induced activation of Src, ERK1/2, and Akt-1 enzymes, and NIH3T3 cell transformation, as well as colony formation of a breast cancer cell line (MDA-MB-468) in soft agar. In contrast, expression of CbpY317F or knockdown endogenous Cbp in NIH3T3 cells by RNA interference significantly enhanced EGF-induced activation of these enzymes and cell transformation. In addition, overexpression of multiple receptor tyrosine kinases (RTKs)-induced Cbp tyrosine phosphorylation. These results demonstrate that Cbp functions as a negative regulator of cell transformation and tumor cell growth through downregulation of Src activation, suggesting that Cbp might be broadly involved in RTKs-activated signaling pathways and tumorigenesis.

Journal ArticleDOI
TL;DR: Imatinib mesylate may affect particular signaling pathways that increase susceptibility to congestive heart failure and therefore may be a potential treatment option for patients with prior heart failure.
Abstract: Imatinib mesylate may affect particular signaling pathways that increase susceptibility to congestive heart failure.

Journal ArticleDOI
TL;DR: A QSAR method with automatic Variable Subset Selection (VSS) by Genetic Algorithm and goodness-of-prediction drivenQSAR model building is described, resulting an externally validated EGFR inhibitory model built from pIC50 values of a diverse structural set of 623 EGFR inhibitors.
Abstract: Epidermal Growth Factor Receptor (EGFR) is a high priority target in anticancer drug research. Thousands of very effective EGFR inhibitors have been developed in the last decade. The known inhibitors are originated from a very diverse chemical space but - without exception - all of them act at the Adenosine TriPhosphate (ATP) binding site of the enzyme. We have collected all of the diverse inhibitor structures and the relevant biological data obtained from comparable assays and built prediction oriented Quantitative Structure- Activity Relationship (QSAR) which models the ATP binding pockets interactive surface from the ligand side. We describe a QSAR method with automatic Variable Subset Selection (VSS) by Genetic Algorithm (GA) and goodness-of-prediction driven QSAR model building, resulting an externally validated EGFR inhibitory model built from pIC50 values of a diverse structural set of 623 EGFR inhibitors. Repeated Trainings/Evaluations (RTE) were used to obtain model fitness values and the effectiveness of VSS is amplified by using predictive ability scores of descriptors. Numerous models were generated by different methods and viable models were collected. Then, intensive RTE were applied to identify ultimate models for external validations. Finally, suitable models were validated by statistical tests. Since we use calculated molecular descriptors in the modeling, these models are suitable for virtual screening for obtaining novel potential EGFR inhibitors.

Book ChapterDOI
01 Jan 2006
TL;DR: Receptor tyrosine kinases are membrane-spanning proteins that possess a ligand-controlled intracellular kinase activity that regulates a wide variety of cellular processess as diverse as cell proliferation, apoptosis or cell migration and have been causally linked to cancer development and progression.
Abstract: Receptor tyrosine kinases (RTKs) are membrane-spanning proteins that possess a ligand-controlled intracellular kinase activity. They regulate a wide variety of cellular processess as diverse as cell proliferation, apoptosis or cell migration. Consequently, dysregulation of RTKs due to overexpression, mutation or autocrine stimulation has been causally linked to cancer development and progression. The advent of molecular cloning allowed the elucidationof the primary structure of the first RTK, the EGFR. Subsequent research in this field led to tremendous advances in understanding molecular signalling processes governing both physiological and pathophysiological behaviour of cells. These discoveries paved the way for the development of target-specific cancer therapeutics and opened up a new era of molecular targeted approaches in the treatment of human cancer. The approval of monoclonal antibodies such as Herceptin© for the treatment of breast cancer or small molecule inhibitors such as Gleevec© for gastrointestinal stromal tumors underlines both the power and success of this novel strategy.

Patent
24 May 2006
TL;DR: In this article, an agent that affects matrix-metalloproteinase 15 (MMP-15) activity and a method of diagnosing, preventing and/or treating a hyperproliferative disease such as precancerous or cancerous cell growth was presented.
Abstract: The present invention relates to an agent that affects matrix- metalloproteinase 15 (MMP-15) activity and a method of diagnosing, preventing and/or treating a hyperproliferative disease such as precancerous or cancerous cell growth.