Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Cancer Cell ; 39(9): 1227-1244.e20, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34297917

ABSTRACT

Fibroblasts display extensive transcriptional heterogeneity, yet functional annotation and characterization of their heterocellular relationships remains incomplete. Using mass cytometry, we chart the stromal composition of 18 murine tissues and 5 spontaneous tumor models, with an emphasis on mesenchymal phenotypes. This analysis reveals extensive stromal heterogeneity across tissues and tumors, and identifies coordinated relationships between mesenchymal and immune cell subsets in pancreatic ductal adenocarcinoma. Expression of CD105 demarks two stable and functionally distinct pancreatic fibroblast lineages, which are also identified in murine and human healthy tissues and tumors. Whereas CD105-positive pancreatic fibroblasts are permissive for tumor growth in vivo, CD105-negative fibroblasts are highly tumor suppressive. This restrictive effect is entirely dependent on functional adaptive immunity. Collectively, these results reveal two functionally distinct pancreatic fibroblast lineages and highlight the importance of mesenchymal and immune cell interactions in restricting tumor growth.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Carcinoma, Pancreatic Ductal/immunology , Endoglin/genetics , Pancreatic Neoplasms/immunology , Single-Cell Analysis/methods , Adaptive Immunity , Animals , Carcinoma, Pancreatic Ductal/genetics , Case-Control Studies , Cell Line, Tumor , Cell Plasticity , Endoglin/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Tumor Microenvironment
2.
Org Process Res Dev ; 25(1): 148-156, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33679122

ABSTRACT

The scale-up of a chiral bicyclic homopiperazine of pharmaceutical interest was investigated. The outcome and safety profile of a key batch ring-expansion step via Schmidt rearrangement was improved using continuous-flow chemistry. The selectivity of nitrogen insertion for the ring expansion was improved via an alternative photochemical oxaziridine rearrangement under mild conditions, which when converted to continuous-flow in a simple and efficient flow reactor allowed the first photochemical scale-up of a homopiperazine.

3.
J Med Chem ; 63(5): 2308-2324, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31430136

ABSTRACT

The lysyl oxidase (LOX) family of extracellular proteins plays a vital role in catalyzing the formation of cross-links in fibrillar elastin and collagens leading to extracellular matrix (ECM) stabilization. These enzymes have also been implicated in tumor progression and metastatic disease and have thus become an attractive therapeutic target for many types of invasive cancers. Following our recently published work on the discovery of aminomethylenethiophenes (AMTs) as potent, orally bioavailable LOX/LOXL2 inhibitors, we report herein the discovery of a series of dual LOX/LOXL2 inhibitors, as well as a subseries of LOXL2-selective inhibitors, bearing an aminomethylenethiazole (AMTz) scaffold. Incorporation of a thiazole core leads to improved potency toward LOXL2 inhibition via an irreversible binding mode of inhibition. SAR studies have enabled the discovery of a predictive 3DQSAR model. Lead AMTz inhibitors exhibit improved pharmacokinetic properties and excellent antitumor efficacy, with significantly reduced tumor growth in a spontaneous breast cancer genetically engineered mouse model.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Thiazoles/pharmacology , Amination , Amino Acid Oxidoreductases/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Female , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/pathology , Protein-Lysine 6-Oxidase/metabolism , Rats , Sulfinic Acids/chemistry , Sulfinic Acids/pharmacokinetics , Sulfinic Acids/pharmacology , Sulfinic Acids/therapeutic use , Thiazoles/chemistry , Thiazoles/pharmacokinetics , Thiazoles/therapeutic use
4.
Cancer Res ; 79(22): 5874-5883, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31604713

ABSTRACT

Increased stiffness in the extracellular matrix (ECM) contributes to tumor progression and metastasis. Therefore, stromal modulating therapies and accompanying biomarkers are being developed to target ECM stiffness. Magnetic resonance (MR) elastography can noninvasively and quantitatively map the viscoelastic properties of tumors in vivo and thus has clear clinical applications. Herein, we used MR elastography, coupled with computational histopathology, to interrogate the contribution of collagen to the tumor biomechanical phenotype and to evaluate its sensitivity to collagenase-induced stromal modulation. Elasticity (G d) and viscosity (G l) were significantly greater for orthotopic BT-474 (G d = 5.9 ± 0.2 kPa, G l = 4.7 ± 0.2 kPa, n = 7) and luc-MDA-MB-231-LM2-4 (G d = 7.9 ± 0.4 kPa, G l = 6.0 ± 0.2 kPa, n = 6) breast cancer xenografts, and luc-PANC1 (G d = 6.9 ± 0.3 kPa, G l = 6.2 ± 0.2 kPa, n = 7) pancreatic cancer xenografts, compared with tumors associated with the nervous system, including GTML/Trp53KI/KI medulloblastoma (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 7), orthotopic luc-D-212-MG (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 7), luc-RG2 (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 5), and luc-U-87-MG (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 8) glioblastoma xenografts, intracranially propagated luc-MDA-MB-231-LM2-4 (G d = 3.7 ± 0.2 kPa, G l = 2.2 ± 0.1 kPa, n = 7) breast cancer xenografts, and Th-MYCN neuroblastomas (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 5). Positive correlations between both elasticity (r = 0.72, P < 0.0001) and viscosity (r = 0.78, P < 0.0001) were determined with collagen fraction, but not with cellular or vascular density. Treatment with collagenase significantly reduced G d (P = 0.002) and G l (P = 0.0006) in orthotopic breast tumors. Texture analysis of extracted images of picrosirius red staining revealed significant negative correlations of entropy with G d (r = -0.69, P < 0.0001) and G l (r = -0.76, P < 0.0001), and positive correlations of fractal dimension with G d (r = 0.75, P < 0.0001) and G l (r = 0.78, P < 0.0001). MR elastography can thus provide sensitive imaging biomarkers of tumor collagen deposition and its therapeutic modulation. SIGNIFICANCE: MR elastography enables noninvasive detection of tumor stiffness and will aid in the development of ECM-targeting therapies.


Subject(s)
Breast Neoplasms/metabolism , Collagen/metabolism , Animals , Cell Line, Tumor , Elasticity , Elasticity Imaging Techniques/methods , Extracellular Matrix/metabolism , Female , Humans , Magnetic Resonance Imaging/methods , Mice , Phenotype
5.
Nat Commun ; 10(1): 3151, 2019 Jul 18.
Article in English | MEDLINE | ID: mdl-31320631

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
J Med Chem ; 62(12): 5863-5884, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31070916

ABSTRACT

Lysyl oxidase (LOX) is a secreted copper-dependent amine oxidase that cross-links collagens and elastin in the extracellular matrix and is a critical mediator of tumor growth and metastatic spread. LOX is a target for cancer therapy, and thus the search for therapeutic agents against LOX has been widely sought. We report herein the medicinal chemistry discovery of a series of LOX inhibitors bearing an aminomethylenethiophene (AMT) scaffold. High-throughput screening provided the initial hits. Structure-activity relationship (SAR) studies led to the discovery of AMT inhibitors with sub-micromolar half-maximal inhibitory concentrations (IC50) in a LOX enzyme activity assay. Further SAR optimization yielded the orally bioavailable LOX inhibitor CCT365623 with good anti-LOX potency, selectivity, pharmacokinetic properties, as well as anti-metastatic efficacy.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biological Availability , Cell Line, Tumor , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Humans , Mice , Neoplasm Metastasis/drug therapy , Structure-Activity Relationship , Thiophenes/chemistry , Thiophenes/pharmacokinetics , Thiophenes/pharmacology , Thiophenes/therapeutic use
7.
Nat Commun ; 8: 14909, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28416796

ABSTRACT

Lysyl oxidase (LOX) remodels the tumour microenvironment by cross-linking the extracellular matrix. LOX overexpression is associated with poor cancer outcomes. Here, we find that LOX regulates the epidermal growth factor receptor (EGFR) to drive tumour progression. We show that LOX regulates EGFR by suppressing TGFß1 signalling through the secreted protease HTRA1. This increases the expression of Matrilin2 (MATN2), an EGF-like domain-containing protein that traps EGFR at the cell surface to facilitate its activation by EGF. We describe a pharmacological inhibitor of LOX, CCT365623, which disrupts EGFR cell surface retention and delays the growth of primary and metastatic tumour cells in vivo. Thus, we show that LOX regulates EGFR cell surface retention to drive tumour progression, and we validate the therapeutic potential of inhibiting this pathway with the small molecule inhibitor CCT365623.


Subject(s)
Cell Membrane/metabolism , Disease Progression , ErbB Receptors/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Protein-Lysine 6-Oxidase/metabolism , Aminopropionitrile/chemistry , Aminopropionitrile/pharmacology , Animals , Biosensing Techniques , Cell Line, Tumor , Cell Membrane/drug effects , Cell Proliferation/drug effects , Dogs , Enzyme Activation , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , High-Temperature Requirement A Serine Peptidase 1/metabolism , Humans , Matrilin Proteins/metabolism , Mice , Models, Biological , Neoplasm Metastasis , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Rats , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
9.
Mol Cancer Ther ; 15(1): 172-83, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26487278

ABSTRACT

Sunitinib and pazopanib are antiangiogenic tyrosine kinase inhibitors (TKI) used to treat metastatic renal cell carcinoma (RCC). However, the ability of these drugs to extend progression-free and overall survival in this patient population is limited by drug resistance. It is possible that treatment outcomes in RCC patients could be improved by rationally combining TKIs with other agents. Here, we address whether inhibition of the Ras-Raf-MEK-ERK1/2 pathway is a rational means to improve the response to TKIs in RCC. Using a xenograft model of RCC, we found that tumors that are resistant to sunitinib have a significantly increased angiogenic response compared with tumors that are sensitive to sunitinib in vivo. We also observed significantly increased levels of phosphorylated ERK1/2 in the vasculature of resistant tumors, when compared with sensitive tumors. These data suggested that the Ras-Raf-MEK-ERK1/2 pathway, an important driver of angiogenesis in endothelial cells, remains active in the vasculature of TKI-resistant tumors. Using an in vitro angiogenesis assay, we identified that the MEK inhibitor (MEKI) trametinib has potent antiangiogenic activity. We then show that, when trametinib is combined with a TKI in vivo, more effective suppression of tumor growth and tumor angiogenesis is achieved than when either drug is utilized alone. In conclusion, we provide preclinical evidence that combining a TKI, such as sunitinib or pazopanib, with a MEKI, such as trametinib, is a rational and efficacious treatment regimen for RCC.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Animals , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Drug Synergism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Indoles/pharmacology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , Neovascularization, Pathologic/drug therapy , Pyrroles/pharmacology , Sunitinib , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Xenograft Model Antitumor Assays
10.
EMBO Mol Med ; 7(8): 1063-76, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26077591

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related mortality. Despite significant advances made in the treatment of other cancers, current chemotherapies offer little survival benefit in this disease. Pancreaticoduodenectomy offers patients the possibility of a cure, but most will die of recurrent or metastatic disease. Hence, preventing metastatic disease in these patients would be of significant benefit. Using principal component analysis (PCA), we identified a LOX/hypoxia signature associated with poor patient survival in resectable patients. We found that LOX expression is upregulated in metastatic tumors from Pdx1-Cre Kras(G12D/+) Trp53(R172H/+) (KPC) mice and that inhibition of LOX in these mice suppressed metastasis. Mechanistically, LOX inhibition suppressed both migration and invasion of KPC cells. LOX inhibition also synergized with gemcitabine to kill tumors and significantly prolonged tumor-free survival in KPC mice with early-stage tumors. This was associated with stromal alterations, including increased vasculature and decreased fibrillar collagen, and increased infiltration of macrophages and neutrophils into tumors. Therefore, LOX inhibition is able to reverse many of the features that make PDAC inherently refractory to conventional therapies and targeting LOX could improve outcome in surgically resectable disease.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Protein-Lysine 6-Oxidase/metabolism , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Humans , Hypoxia , Mice , Neoplasm Metastasis/prevention & control , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Treatment Outcome , Gemcitabine
11.
Cancer Cell ; 27(1): 85-96, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25500121

ABSTRACT

BRAF and MEK inhibitors are effective in BRAF mutant melanoma, but most patients eventually relapse with acquired resistance, and others present intrinsic resistance to these drugs. Resistance is often mediated by pathway reactivation through receptor tyrosine kinase (RTK)/SRC-family kinase (SFK) signaling or mutant NRAS, which drive paradoxical reactivation of the pathway. We describe pan-RAF inhibitors (CCT196969, CCT241161) that also inhibit SFKs. These compounds do not drive paradoxical pathway activation and inhibit MEK/ERK in BRAF and NRAS mutant melanoma. They inhibit melanoma cells and patient-derived xenografts that are resistant to BRAF and BRAF/MEK inhibitors. Thus, paradox-breaking pan-RAF inhibitors that also inhibit SFKs could provide first-line treatment for BRAF and NRAS mutant melanomas and second-line treatment for patients who develop resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Melanoma/drug therapy , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyrazines/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/pathology , Melanoma, Experimental , Mice , Mice, Nude , Xenograft Model Antitumor Assays
12.
Sci Signal ; 7(318): ra30, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24667377

ABSTRACT

Melanoma is a highly metastatic and lethal form of skin cancer. The protein kinase BRAF is mutated in about 40% of melanomas, and BRAF inhibitors improve progression-free and overall survival in these patients. However, after a relatively short period of disease control, most patients develop resistance because of reactivation of the RAF-ERK (extracellular signal-regulated kinase) pathway, mediated in many cases by mutations in RAS. We found that BRAF inhibition induces invasion and metastasis in RAS mutant melanoma cells through a mechanism mediated by the reactivation of the MEK (mitogen-activated protein kinase kinase)-ERK pathway, increased expression and secretion of interleukin 8, and induction of protease-dependent invasion. These events were accompanied by a cell morphology switch from predominantly rounded to predominantly elongated cells. We also observed similar responses in BRAF inhibitor-resistant melanoma cells. These data show that BRAF inhibitors can induce melanoma cell invasion and metastasis in tumors that develop resistance to these drugs.


Subject(s)
MAP Kinase Signaling System/physiology , Melanoma/physiopathology , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/physiopathology , Animals , Blotting, Western , Cell Shape/physiology , Dimethyl Sulfoxide , Drug Resistance, Neoplasm , Enzyme-Linked Immunosorbent Assay , Humans , Indoles/pharmacology , Interleukin-8/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Statistics, Nonparametric , Sulfonamides/pharmacology
13.
Mol Imaging Biol ; 16(2): 152-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23955100

ABSTRACT

PURPOSE: The purpose of this study is to evaluate if the differential exchange rates with bulk water between amine and amide protons can be exploited using chemical exchange saturation transfer magnetic resonance (CEST-MR) to monitor the release of glutamate induced by carboxypeptidase G2 (CPG2), an enzyme utilized in cancer gene therapy. PROCEDURES: Z spectra of solutions of the CPG2 substrate, 3,5-difluorobenzoyl-L-glutamate (amide), and glutamate (amine) were acquired at 11.7 T, 37 °C, across different pH (5-8). The ability of CEST-MR to monitor CPG2-mediated release of glutamate was assessed in extracts of CPG2-expressing cancer cells and purified solution of CPG2. RESULTS: The addition of CPG2 to a solution containing 3,5-difluorobenzoyl-L-glutamate led to a marked and progressively increasing CEST effect (+3 ppm), concomitant with the time-dependent release of glutamate induced by CPG2. CONCLUSION: CEST-MR allows the detection of CPG2 activity in vitro and supports the translation of CEST-MRI to assess CPG2-based gene therapy in vivo.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Prodrugs/metabolism , gamma-Glutamyl Hydrolase/metabolism , Benzoates/metabolism , Cell Line, Tumor , Humans , Mechlorethamine/chemistry , Mechlorethamine/metabolism , Prodrugs/chemistry , Signal Processing, Computer-Assisted , Solutions
14.
J Control Release ; 170(1): 120-31, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23688772

ABSTRACT

Current conventional treatments for cancer lack tumour selectivity resulting in the destruction of healthy tissue and severe adverse effects to the patient in addition to limiting the administration dose and efficacy. Hence, it is imperative that we seek alternative approaches to treat cancer that localise therapeutic agents to the site of the tumour and spare normal tissue. The use of bacteria in cancer therapy represents one such approach. Bacteria were first used as anti-cancer agents over a century ago. Today, this field has re-emerged from the past and is progressing at a rapid rate. Bacteria are used as anticancer agents either alone or in combination with conventional treatments and have been armed with an arsenal of therapeutic genes, which enhance their efficacy. Bacterial directed enzyme prodrug therapy (BDEPT) is one of the most promising approaches, which harnesses the tumour-specific location of bacteria to locally activate systemically administered 'prodrugs' within the tumour in order to induce selective tumour destruction. BDEPT is a relatively new concept. It was originally conceived more than 10years ago but it is only until recently that we witness a surge in activity in this field. In this review, we provide a full account of developments in the field of BDEPT since its inception. We share technical knowhow and discuss optimization strategies for vector and enzyme combinations, provide a clear view of the research landscape and suggest possible directions for the field.


Subject(s)
Antineoplastic Agents/administration & dosage , Bacteria , Enzyme Therapy/methods , Neoplasms/therapy , Prodrugs/administration & dosage , Animals , Humans
15.
Bioorg Med Chem ; 21(5): 1284-304, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23376011

ABSTRACT

The RAS-RAF-MEK-ERK pathway is hyperactivated in 30% of human cancers. BRAF is a serine-threonine kinase, belonging to this pathway that is mutated with high frequency in human melanoma and other cancers thus BRAF is an important therapeutic target in melanoma. We have designed inhibitors of BRAF based on 2,4,5-trisubstituted imidazoles with naphthyl and benzothiophene-4-substituents. Two compounds were discovered to be potent BRAF inhibitors: 1-(6-{2-[4-(2-dimethylamino-ethoxy)phenyl]-5-(pyridin-4-yl)-1H-imidazol-4-yl} benzo[b]thiophen-3-yl)-2,2,2-trifluoroethanol (1i) with BRAF IC(50)=190 nM and with cellular GI(50)=2100 nM, and 6-{2-[4-(2-dimethylamino-ethoxy)-phenyl]-5-pyridin-4-yl-3H-imidazol-4-yl}-naphthalen-1-ol (1q) with IC(50)=9 nM and GI(50)=220 nM.


Subject(s)
Imidazoles/chemistry , Naphthols/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Thiophenes/chemistry , Benzofurans/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Humans , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Melanoma/metabolism , Melanoma/pathology , Naphthols/chemical synthesis , Naphthols/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/pharmacology
16.
Expert Opin Ther Pat ; 23(2): 155-64, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23294221

ABSTRACT

INTRODUCTION: After its identification as an oncogene in 2002, mutant BRAF has become the target of a number of drug discovery programmes, primarily aimed at the treatment of late stage or unresectable melanoma. Some of the drugs thus developed, such as vemurafenib and dabrafenib, show impressive responses in melanoma patients harbouring a BRAF mutation. AREAS COVERED: This review summarises the patent literature on BRAF from 2006 to 2012, focusing on the specific areas of inhibitors of mutant BRAF, drug combinations including BRAF inhibitors, diagnostic methods for use with mutant BRAF inhibitors & diagnosis and treatment of mutant BRAF cancers resistant to BRAF inhibitors. EXPERT OPINION: Whilst these first-generation BRAF inhibitors initially mediate excellent responses in late stage or unresectable melanoma patients bearing the V600 mutation, resistance usually occurs and patients eventually relapse. The patent literature for new BRAF inhibitors and therapies reflects the desire to develop second-generation drugs able to overcome this resistance and combination treatments that increase the efficiency of current mutant BRAF inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Patents as Topic , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Drug Design , Drug Resistance, Neoplasm/genetics , Humans , Molecular Structure , Molecular Targeted Therapy , Mutation , Neoplasms/diagnosis , Neoplasms/enzymology , Neoplasms/genetics , Patient Selection , Precision Medicine , Predictive Value of Tests , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction/drug effects
17.
Cancer Discov ; 3(4): 458-469, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303902

ABSTRACT

UNLABELLED: NRAS mutations are common in human melanoma. To produce a mouse model of NRAS-driven melanoma, we expressed oncogenic NRAS (NRAS(G12D)) in mouse melanocytes. When NRAS(G12D) was expressed in the melanocytes of developing embryos, it induced melanocyte proliferation and congenital melanocytic lesions reminiscent of human blue nevi but did not induce cutaneous melanoma. Unexpectedly, however, it did induce early-onset primary melanoma of the central nervous system (CNS). The tumors were rapidly proliferating and caused neurologic symptoms, rapid health deterioration, and death. NRAS is not a common driver oncogene of primary melanoma of the CNS in adults, but we report two cases of primary melanoma of the CNS in children, both of which carried oncogenic mutations in NRAS. We conclude that acquisition of somatic mutations in NRAS in CNS melanocytes is a predisposing risk factor for primary melanoma of the CNS in children, and we present a mouse model of this disease. SIGNIFICANCE: We show that the acquisition of NRAS mutations in melanocytes during embryogenesis is a risk factor for early-onset melanoma of the CNS. We have developed a powerful mouse model to study this rare but devastating childhood disease, and to develop therapeutic approaches for its treatment.


Subject(s)
Central Nervous System Neoplasms/genetics , Genes, ras/genetics , Melanocytes/metabolism , Melanoma/genetics , Animals , Child , Child, Preschool , Female , Humans , Male , Mice , Mice, Inbred C57BL
18.
Cancer Discov ; 3(2): 158-67, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23242808

ABSTRACT

UNLABELLED: We generated cell lines resistant to BRAF inhibitors and show that the EGF receptor (EGFR)-SRC family kinase (SFK)-STAT3 signaling pathway was upregulated in these cells. In addition to driving proliferation of resistant cells, this pathway also stimulated invasion and metastasis. EGFR inhibitors cooperated with BRAF inhibitors to block the growth of the resistant cells in vitro and in vivo, and monotherapy with the broad specificity tyrosine kinase inhibitor dasatinib blocked growth and metastasis in vivo. We analyzed tumors from patients with intrinsic or acquired resistance to vemurafenib and observed increased EGFR and SFK activity. Furthermore, dasatinib blocked the growth and metastasis of one of the resistant tumors in immunocompromised mice. Our data show that BRAF inhibitor-mediated activation of EGFR-SFK-STAT3 signaling can mediate resistance in patients with BRAF-mutant melanoma. We describe 2 treatments that seem to overcome this resistance and could deliver therapeutic efficacy in patients with drug-resistant BRAF-mutant melanoma. SIGNIFICANCE: Therapies that target the driver oncogenes in cancer can achieve remarkable responses if patients are stratified for treatment. However, as with conventional therapies, patients often develop acquired resistance to targeted therapies, and a proportion of patients are intrinsically resistant and fail to respond despite the presence of an appropriate driver oncogene mutation. We found that the EGFR/SFK pathway mediated resistance to vemurafenib in BRAF -mutant melanoma and that BRAF and EGFR or SFK inhibition blocked proliferation and invasion of these resistant tumors, providing potentially effective therapeutic options for these patients.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dasatinib , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Female , Humans , Immunoblotting , Indoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Molecular Sequence Data , Mutation , Phosphorylation/drug effects , Proto-Oncogene Proteins B-raf/metabolism , Pyrimidines/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Thiazoles/pharmacology , Vemurafenib , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
20.
Angiogenesis ; 15(4): 623-41, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22843200

ABSTRACT

Sunitinib is a potent and clinically approved tyrosine kinase inhibitor that can suppress tumour growth by inhibiting angiogenesis. However, conflicting data exist regarding the effects of this drug on the growth of metastases in preclinical models. Here we use 4T1 and RENCA tumour cells, which both form lung metastases in Balb/c mice, to re-address the effects of sunitinib on the progression of metastatic disease in mice. We show that treatment of mice with sunitinib prior to intravenous injection of tumour cells can promote the seeding and growth of 4T1 lung metastases, but not RENCA lung metastases, showing that this effect is cell line dependent. However, increased metastasis occurred only upon administration of a very high sunitinib dose, but not when lower, clinically relevant doses were used. Mechanistically, high dose sunitinib led to a pericyte depletion effect in the lung vasculature that correlated with increased seeding of metastasis. By administering sunitinib to mice after intravenous injection of tumour cells, we demonstrate that while sunitinib does not inhibit the growth of 4T1 lung tumour nodules, it does block the growth of RENCA lung tumour nodules. This contrasting response was correlated with increased myeloid cell recruitment and persistent vascularisation in 4T1 tumours, whereas RENCA tumours recruited less myeloid cells and were more profoundly devascularised upon sunitinib treatment. Finally, we show that progression of 4T1 tumours in sunitinib treated mice results in increased hypoxia and increased glucose metabolism in these tumours and that this is associated with a poor outcome. Taken together, these data suggest that the effects of sunitinib on tumour progression are dose-dependent and tumour model-dependent. These findings have relevance for understanding how anti-angiogenic agents may influence disease progression when used in the adjuvant or metastatic setting in cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Indoles/therapeutic use , Neoplasm Metastasis/drug therapy , Pyrroles/therapeutic use , Animals , Mice , Mice, Inbred BALB C , Positron-Emission Tomography , Sunitinib , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL