Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
2.
Nat Cancer ; 5(7): 1102-1120, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38565920

ABSTRACT

The YAP-TEAD protein-protein interaction mediates YAP oncogenic functions downstream of the Hippo pathway. To date, available YAP-TEAD pharmacologic agents bind into the lipid pocket of TEAD, targeting the interaction indirectly via allosteric changes. However, the consequences of a direct pharmacological disruption of the interface between YAP and TEADs remain largely unexplored. Here, we present IAG933 and its analogs as potent first-in-class and selective disruptors of the YAP-TEAD protein-protein interaction with suitable properties to enter clinical trials. Pharmacologic abrogation of the interaction with all four TEAD paralogs resulted in YAP eviction from chromatin and reduced Hippo-mediated transcription and induction of cell death. In vivo, deep tumor regression was observed in Hippo-driven mesothelioma xenografts at tolerated doses in animal models as well as in Hippo-altered cancer models outside mesothelioma. Importantly this also extended to larger tumor indications, such as lung, pancreatic and colorectal cancer, in combination with RTK, KRAS-mutant selective and MAPK inhibitors, leading to more efficacious and durable responses. Clinical evaluation of IAG933 is underway.


Subject(s)
Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Transcription Factors , Xenograft Model Antitumor Assays , Humans , Animals , Transcription Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Mice , Cell Line, Tumor , Adaptor Proteins, Signal Transducing/metabolism , YAP-Signaling Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Signal Transduction/drug effects , TEA Domain Transcription Factors , ras Proteins/metabolism , Female , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
3.
Nat Med ; 28(10): 2162-2170, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36097219

ABSTRACT

Anti-BRAF/EGFR therapy was recently approved for the treatment of metastatic BRAFV600E colorectal cancer (mCRCBRAF-V600E). However, a large fraction of patients do not respond, underscoring the need to identify molecular determinants of treatment response. Using whole-exome sequencing in a discovery cohort of patients with mCRCBRAF-V600E treated with anti-BRAF/EGFR therapy, we found that inactivating mutations in RNF43, a negative regulator of WNT, predict improved response rates and survival outcomes in patients with microsatellite-stable (MSS) tumors. Analysis of an independent validation cohort confirmed the relevance of RNF43 mutations to predicting clinical benefit (72.7% versus 30.8%; P = 0.03), as well as longer progression-free survival (hazard ratio (HR), 0.30; 95% confidence interval (CI), 0.12-0.75; P = 0.01) and overall survival (HR, 0.26; 95% CI, 0.10-0.71; P = 0.008), in patients with MSS-RNF43mutated versus MSS-RNF43wild-type tumors. Microsatellite-instable tumors invariably carried a wild-type-like RNF43 genotype encoding p.G659fs and presented an intermediate response profile. We found no association of RNF43 mutations with patient outcomes in a control cohort of patients with MSS-mCRCBRAF-V600E tumors not exposed to anti-BRAF targeted therapies. Overall, our findings suggest a cross-talk between the MAPK and WNT pathways that may modulate the antitumor activity of anti-BRAF/EGFR therapy and uncover predictive biomarkers to optimize the clinical management of these patients.


Subject(s)
Colorectal Neoplasms , Ubiquitin-Protein Ligases , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , ErbB Receptors/genetics , Humans , Microsatellite Instability , Mutation , Proto-Oncogene Proteins B-raf/genetics , Ubiquitin-Protein Ligases/genetics
4.
Cancer Cell ; 39(11): 1531-1547.e10, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34624218

ABSTRACT

Cancer-associated fibroblasts (CAFs) are highly heterogeneous. With the lack of a comprehensive understanding of CAFs' functional distinctions, it remains unclear how cancer treatments could be personalized based on CAFs in a patient's tumor. We have established a living biobank of CAFs derived from biopsies of patients' non-small lung cancer (NSCLC) that encompasses a broad molecular spectrum of CAFs in clinical NSCLC. By functionally interrogating CAF heterogeneity using the same therapeutics received by patients, we identify three functional subtypes: (1) robustly protective of cancers and highly expressing HGF and FGF7; (2) moderately protective of cancers and highly expressing FGF7; and (3) those providing minimal protection. These functional differences among CAFs are governed by their intrinsic TGF-ß signaling, which suppresses HGF and FGF7 expression. This CAF functional classification correlates with patients' clinical response to targeted therapies and also associates with the tumor immune microenvironment, therefore providing an avenue to guide personalized treatment.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Fibroblast Growth Factor 7/genetics , Hepatocyte Growth Factor/genetics , Lung Neoplasms/pathology , Biopsy , Cancer-Associated Fibroblasts/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Precision Medicine , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Up-Regulation
5.
Nat Cancer ; 2(4): 414-428, 2021 04.
Article in English | MEDLINE | ID: mdl-34179825

ABSTRACT

Brain metastases are refractory to therapies that control systemic disease in patients with human epidermal growth factor receptor 2 (HER2+) breast cancer, and the brain microenvironment contributes to this therapy resistance. Nutrient availability can vary across tissues, therefore metabolic adaptations required for brain metastatic breast cancer growth may introduce liabilities that can be exploited for therapy. Here, we assessed how metabolism differs between breast tumors in brain versus extracranial sites and found that fatty acid synthesis is elevated in breast tumors growing in brain. We determine that this phenotype is an adaptation to decreased lipid availability in brain relative to other tissues, resulting in a site-specific dependency on fatty acid synthesis for breast tumors growing at this site. Genetic or pharmacological inhibition of fatty acid synthase (FASN) reduces HER2+ breast tumor growth in the brain, demonstrating that differences in nutrient availability across metastatic sites can result in targetable metabolic dependencies.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Brain Neoplasms/metabolism , Breast Neoplasms/drug therapy , Fatty Acid Synthases/genetics , Fatty Acids/therapeutic use , Female , Humans , Tumor Microenvironment
7.
NPJ Breast Cancer ; 5: 4, 2019.
Article in English | MEDLINE | ID: mdl-30675514

ABSTRACT

The effective treatment of cerebral metastases from HER2-positive breast cancer remains an unmet need. Recent studies indicate that activated astrocytes and brain endothelial cells exert chemoprotective effects on cancer cells through direct physical interaction. Here we report that the endothelin axis mediates protection of HER2-amplified brain metastatic breast cancers to the anti-HER2 antibody-drug conjugate ado-trastuzumab emtansine (T-DM1). Macitentan, a dual inhibitor of endothelin receptors A and B, improves the efficacy of T-DM1 against breast cancers grown in the brain. We show that direct contact of brain stroma with cancer cells is required for protection to T-DM1. Our data suggest that targeting the endothelin axis may be beneficial when anti-signaling agent and cytotoxic agent are combined. These findings may contribute to the development of therapeutic approaches with enhanced efficacy in the brain microenvironment.

8.
Cell Rep ; 21(11): 3298-3309, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29241554

ABSTRACT

Personalized cancer therapy is based on a patient's tumor lineage, histopathology, expression analyses, and/or tumor DNA or RNA analysis. Here, we aim to develop an in vitro functional assay of a patient's living cancer cells that could complement these approaches. We present methods for developing cell cultures from tumor biopsies and identify the types of samples and culture conditions associated with higher efficiency of model establishment. Toward the application of patient-derived cell cultures for personalized care, we established an immunofluorescence-based functional assay that quantifies cancer cell responses to targeted therapy in mixed cell cultures. Assaying patient-derived lung cancer cultures with this method showed promise in modeling patient response for diagnostic use. This platform should allow for the development of co-clinical trial studies to prospectively test the value of drug profiling on tumor-biopsy-derived cultures to direct patient care.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Lung Neoplasms/drug therapy , Neoplasms/drug therapy , Precision Medicine/methods , Primary Cell Culture/methods , Acrylamides , Aminopyridines , Anaplastic Lymphoma Kinase , Aniline Compounds , Biomarkers, Tumor/metabolism , Biopsy , Crizotinib , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride/therapeutic use , Feeder Cells/cytology , Fluorescent Antibody Technique/methods , Gene Expression , High-Throughput Screening Assays , Humans , Keratin-18/genetics , Keratin-18/metabolism , Keratin-8/genetics , Keratin-8/metabolism , Lactams , Lactams, Macrocyclic/therapeutic use , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mutation , Neoplasms/classification , Neoplasms/genetics , Neoplasms/pathology , Piperazines/therapeutic use , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Cells, Cultured
10.
Int J Pediatr Otorhinolaryngol ; 95: 133-138, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28576522

ABSTRACT

INTRODUCTION: Tumor immunotherapy have broadened therapeutic options for tumor treatment. The role of immune function in juvenile recurrent respiratory papillomatosis (JRRP) has not been investigated. Applying immunoblockade inhibitors as a novel disease treatment is unclear. Our study, for the first time, evaluates immune infiltration and immuno-suppressive molecule expression in JRRP. Our study provides insights in possibly treating this disease with tumor immunotherapies. We aimed to determine expression of programmed death-ligand 1 (PD-L1), a cancer escape protein, and presence of CD8+ T cell infiltration in tumor microenvironment. MATERIAL AND METHODS: Seven patients with JRRP (mean age: 7.43; age range 3-17) in this study routinely have their tumors surgical debulked at Massachusetts Eye and Ear Infirmary. Following surgery, samples were de-identified and sent to pathology where they were stained and analyzed. RESULTS: Six out of seven patients expressed PD-L1 on tumor cells to various extents. Three patients showed concurrent PD-L1 expression on tumor cells and abundant CD8+ tumor infiltrating lymphocytes as well as PD-L1+ stromal lymphocytes, while PD-L1 expression on tumor cells were not associated with CD8+ tumor infiltrating T cells nor PD-L1+ stromal lymphocytes in the other three patients. HPV 6/11 and p16 was detected in all the patients. There appeared to be no correlation between either PD-L1 expression and CD8+ infiltration and clinical severity as measured by both the number of surgeries per year or Derkay score. CONCLUSIONS: Despite a small cohort, the expression of p16 and HPV 6/11 in all of the patients confirms the tissues were HPV tumor cells. PD-L1 expression was detected in the vast majority of tumor samples, while inflammatory cell compartments showed a higher degree of variation. Expression of PD-L1 on tumor cells but not inflammatory cells raises the possibility of a tumor cell intrinsic manner of PD-L1 expression. In contrast, a group of patients showed PD-L1 positivity in both tumor and inflammatory cells along with abundant CD8+ tumor infiltrating lymphocytes, suggesting adoptive immune resistance in these tumors and potential benefits from tumor immunotherapy.


Subject(s)
B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Papillomavirus Infections/metabolism , Respiratory Tract Infections/metabolism , Adolescent , Child , Child, Preschool , Female , Genetic Heterogeneity , Humans , Immunohistochemistry , In Situ Hybridization , Male , Papillomavirus Infections/pathology , Respiratory Tract Infections/pathology
11.
Sci Transl Med ; 9(391)2017 05 24.
Article in English | MEDLINE | ID: mdl-28539475

ABSTRACT

Although targeted therapies are often effective systemically, they fail to adequately control brain metastases. In preclinical models of breast cancer that faithfully recapitulate the disparate clinical responses in these microenvironments, we observed that brain metastases evade phosphatidylinositide 3-kinase (PI3K) inhibition despite drug accumulation in the brain lesions. In comparison to extracranial disease, we observed increased HER3 expression and phosphorylation in brain lesions. HER3 blockade overcame the resistance of HER2-amplified and/or PIK3CA-mutant breast cancer brain metastases to PI3K inhibitors, resulting in marked tumor growth delay and improvement in mouse survival. These data provide a mechanistic basis for therapeutic resistance in the brain microenvironment and identify translatable treatment strategies for HER2-amplified and/or PIK3CA-mutant breast cancer brain metastases.


Subject(s)
Brain Neoplasms/metabolism , Breast Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptor, ErbB-3/metabolism , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/complications , Breast Neoplasms/drug therapy , Female , Mice , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-3/genetics
12.
Cell Res ; 26(9): 973-4, 2016 09.
Article in English | MEDLINE | ID: mdl-27514701

ABSTRACT

Astrocytes are emerging as essential regulators of brain metastasis progression. In a current issue of Nature, Chen et al. identify a novel mechanism of astrocyte-carcinoma interaction and exploit vulnerabilities therein to slow brain metastatic growth in pre-clinical models.


Subject(s)
Astrocytes , Brain Neoplasms , Brain , Gap Junctions , Humans , Nucleotides, Cyclic
13.
J Natl Cancer Inst ; 108(2)2016 Feb.
Article in English | MEDLINE | ID: mdl-26547932

ABSTRACT

BACKGROUND: Central nervous system (CNS) metastases represent a major problem in the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer because of the disappointing efficacy of HER2-targeted therapies against brain lesions. The antibody-drug conjugate ado-trastuzumab emtansine (T-DM1) has shown efficacy in trastuzumab-resistant systemic breast cancer. Here, we tested the hypothesis that T-DM1 could overcome trastuzumab resistance in murine models of brain metastases. METHODS: We treated female nude mice bearing BT474 or MDA-MB-361 brain metastases (n = 9-11 per group) or cancer cells grown in organotypic brain slice cultures with trastuzumab or T-DM1 at equivalent or equipotent doses. Using intravital imaging, molecular techniques and histological analysis we determined tumor growth, mouse survival, cancer cell apoptosis and proliferation, tumor drug distribution, and HER2 signaling. Data were analyzed with one-way analysis of variance (ANOVA), Kaplan-Meier analysis, and Coefficient of Determination. All statistical tests were two-sided. RESULTS: T-DM1 delayed the growth of HER2-positive breast cancer brain metastases compared with trastuzumab. These findings were consistent between HER2-driven and PI3K-driven tumors. The activity of T-DM1 resulted in a survival benefit (median survival for BT474 tumors: 28 days for trastuzumab vs 112 days for T-DM1, hazard ratio = 6.2, 95% confidence interval = 6.1 to 85.84, P < .001). No difference in drug distribution or HER2-signaling was revealed between the two groups. However, T-DM1 led to a statistically significant increase in tumor cell apoptosis (one-way ANOVA for ApopTag, P < .001), which was associated with mitotic catastrophe. CONCLUSIONS: T-DM1 can overcome resistance to trastuzumab therapy in HER2-driven or PI3K-driven breast cancer brain lesions due to the cytotoxicity of the DM1 component. Clinical investigation of T-DM1 for patients with CNS metastases from HER2-positive breast cancer is warranted.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/analysis , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Maytansine/analogs & derivatives , Receptor, ErbB-2/analysis , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Blotting, Western , Brain Neoplasms/chemistry , Breast Neoplasms/chemistry , Cell Proliferation/drug effects , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Kaplan-Meier Estimate , Maytansine/administration & dosage , Maytansine/pharmacology , Mice , Mice, Nude , Microarray Analysis , Microscopy, Electron , Odds Ratio , Trastuzumab , Xenograft Model Antitumor Assays
14.
Cancer Cell ; 28(1): 70-81, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26144315

ABSTRACT

We report the preclinical evaluation of PF-06463922, a potent and brain-penetrant ALK/ROS1 inhibitor. Compared with other clinically available ALK inhibitors, PF-06463922 displayed superior potency against all known clinically acquired ALK mutations, including the highly resistant G1202R mutant. Furthermore, PF-06463922 treatment led to regression of EML4-ALK-driven brain metastases, leading to prolonged mouse survival, in a superior manner. Finally, PF-06463922 demonstrated high selectivity and safety margins in a variety of preclinical studies. These results suggest that PF-06463922 will be highly effective for the treatment of patients with ALK-driven lung cancers, including those who relapsed on clinically available ALK inhibitors because of secondary ALK kinase domain mutations and/or brain metastases.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Drug Resistance, Neoplasm/drug effects , Lactams, Macrocyclic/administration & dosage , Neoplasms/drug therapy , Receptor Protein-Tyrosine Kinases/genetics , Aminopyridines , Anaplastic Lymphoma Kinase , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Lactams , Lactams, Macrocyclic/pharmacology , Mice , Mutation , NIH 3T3 Cells , Neoplasms/genetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrazoles , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
15.
Cancer Cell ; 27(2): 163-75, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25670078

ABSTRACT

Brain metastasis is an end stage in breast cancer progression. Traditional treatment options have minimal efficacy, and overall survival is on the order of months. The incidence of brain metastatic disease is increasing with the improved management of systemic disease and prolongation of survival. Unfortunately, the targeted therapies that control systemic disease have diminished efficacy against brain lesions. There are reasons to be optimistic, however, as emerging therapies have shown promise in preclinical and early clinical settings. This review discusses recent advances in breast cancer brain metastasis therapy and potential approaches for successful treatment.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Breast Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , Molecular Targeted Therapy , Pathology, Molecular , Receptor, ErbB-2/genetics
16.
J Natl Cancer Inst ; 107(4)2015 Apr.
Article in English | MEDLINE | ID: mdl-25710962

ABSTRACT

BACKGROUND: Matrix metalloproteinase (MMP) 14 may mediate tumor progression through vascular and immune-modulatory effects. METHODS: Orthotopic murine breast tumors (4T1 and E0771 with high and low MMP14 expression, respectively; n = 5-10 per group) were treated with an anti-MMP14 inhibitory antibody (DX-2400), IgG control, fractionated radiation therapy, or their combination. We assessed primary tumor growth, transforming growth factor ß (TGFß) and inducible nitric oxide synthase (iNOS) expression, macrophage phenotype, and vascular parameters. A linear mixed model with repeated observations, with Mann-Whitney or analysis of variance with Bonferroni post hoc adjustment, was used to determine statistical significance. All statistical tests were two-sided. RESULTS: DX-2400 inhibited tumor growth compared with IgG control treatment, increased macrophage numbers, and shifted the macrophage phenotype towards antitumor M1-like. These effects were associated with a reduction in active TGFß and SMAD2/3 signaling. DX-2400 also transiently increased iNOS expression and tumor perfusion, reduced tissue hypoxia (median % area: control, 20.2%, interquartile range (IQR) = 6.4%-38.9%; DX-2400: 1.2%, IQR = 0.2%-3.2%, P = .044), and synergistically enhanced radiation therapy (days to grow to 800mm(3): control, 12 days, IQR = 9-13 days; DX-2400 plus radiation, 29 days, IQR = 26-30 days, P < .001) in the 4T1 model. The selective iNOS inhibitor, 1400W, abolished the effects of DX-2400 on vessel perfusion and radiotherapy. On the other hand, DX-2400 was not capable of inducing iNOS expression or synergizing with radiation in E0771 tumors. CONCLUSION: MMP14 blockade decreased immunosuppressive TGFß, polarized macrophages to an antitumor phenotype, increased iNOS, and improved tumor perfusion, resulting in reduced primary tumor growth and enhanced response to radiation therapy, especially in high MMP14-expressing tumors.


Subject(s)
Amidines/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Benzylamines/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Macrophages/drug effects , Matrix Metalloproteinase 14/drug effects , Matrix Metalloproteinase 14/metabolism , Nitric Oxide Synthase Type II/drug effects , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Cell Line, Tumor , Dose Fractionation, Radiation , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunoglobulin G/blood , Macrophages/enzymology , Mammary Neoplasms, Experimental , Mice , Neovascularization, Pathologic , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Phenotype , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation
17.
Cancer Discov ; 4(1): 42-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24163374

ABSTRACT

Colorectal cancers harboring KRAS or BRAF mutations are refractory to current targeted therapies. Using data from a high-throughput drug screen, we have developed a novel therapeutic strategy that targets the apoptotic machinery using the BCL-2 family inhibitor ABT-263 (navitoclax) in combination with a TORC1/2 inhibitor, AZD8055. This combination leads to efficient apoptosis specifically in KRAS- and BRAF-mutant but not wild-type (WT) colorectal cancer cells. This specific susceptibility results from TORC1/2 inhibition leading to suppression of MCL-1 expression in mutant, but not WT, colorectal cancers, leading to abrogation of BIM/MCL-1 complexes. This combination strategy leads to tumor regressions in both KRAS-mutant colorectal cancer xenograft and genetically engineered mouse models of colorectal cancer, but not in the corresponding KRAS-WT colorectal cancer models. These data suggest that the combination of BCL-2/BCL-XL inhibitors with TORC1/2 inhibitors constitutes a promising targeted therapy strategy to treat these recalcitrant cancers.


Subject(s)
Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Morpholines/therapeutic use , Multiprotein Complexes/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Sulfonamides/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Mutant Strains , Mice, Nude , Morpholines/pharmacology , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras) , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors , ras Proteins/genetics
18.
Proc Natl Acad Sci U S A ; 109(45): E3119-27, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23071298

ABSTRACT

Brain metastases are a serious obstacle in the treatment of patients with human epidermal growth factor receptor-2 (HER2)-amplified breast cancer. Although extracranial disease is controlled with HER2 inhibitors in the majority of patients, brain metastases often develop. Because these brain metastases do not respond to therapy, they are frequently the reason for treatment failure. We developed a mouse model of HER2-amplified breast cancer brain metastasis using an orthotopic xenograft of BT474 cells. As seen in patients, the HER2 inhibitors trastuzumab and lapatinib controlled tumor progression in the breast but failed to contain tumor growth in the brain. We observed that the combination of a HER2 inhibitor with an anti-VEGF receptor-2 (VEGFR2) antibody significantly slows tumor growth in the brain, resulting in a striking survival benefit. This benefit appears largely due to an enhanced antiangiogenic effect: Combination therapy reduced both the total and functional microvascular density in the brain xenografts. In addition, the combination therapy led to a marked increase in necrosis of the brain lesions. Moreover, we observed even better antitumor activity after combining both trastuzumab and lapatinib with the anti-VEGFR2 antibody. This triple-drug combination prolonged the median overall survival fivefold compared with the control-treated group and twofold compared with either two-drug regimen. These findings support the clinical development of this three-drug regimen for the treatment of HER2-amplified breast cancer brain metastases.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Gene Amplification , Molecular Targeted Therapy , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Vessels/drug effects , Blood Vessels/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cell Death/drug effects , Cell Proliferation/drug effects , Diagnostic Imaging , Disease Models, Animal , Female , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Lapatinib , Mice , Necrosis , Neovascularization, Pathologic/drug therapy , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Survival Analysis , Trastuzumab , Treatment Outcome , Xenograft Model Antitumor Assays
19.
Am J Pathol ; 179(5): 2220-32, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21945411

ABSTRACT

Fibroblast growth factors (FGFs) participate in embryonic development, in maintenance of tissue homeostasis in the adult, and in various diseases. FGF-binding proteins (FGFBP) are secreted proteins that chaperone FGFs stored in the extracellular matrix to their receptor, and can thus modulate FGF signaling. FGFBP1 (alias BP1, FGF-BP1, or HBp17) expression is required for embryonic survival, can modulate FGF-dependent vascular permeability in embryos, and is an angiogenic switch in human cancers. To determine the function of BP1 in vivo, we generated tetracycline-regulated conditional BP1 transgenic mice. BP1-expressing adult mice are viable, fertile, and phenotypically indistinguishable from their littermates. Induction of BP1 expression increased mouse primary fibroblast motility in vitro, increased angiogenic sprouting into subcutaneous matrigel plugs in animals and accelerated the healing of excisional skin wounds. FGF-receptor kinase inhibitors blocked these effects. Healing skin wounds showed increased macrophage invasion as well as cell proliferation after BP1 expression. Also, BP1 expression increased angiogenesis during the healing of skin wounds as well as after ischemic injury to hindlimb skeletal muscles. We conclude that BP1 can enhance FGF effects that are required for the healing and repair of injured tissues in adult animals.


Subject(s)
Carrier Proteins/metabolism , Fibroblasts/metabolism , Neovascularization, Physiologic/physiology , Wound Healing/physiology , Animals , Carrier Proteins/genetics , Cell Movement , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Hindlimb/blood supply , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Ischemia/metabolism , Ischemia/physiopathology , Macrophages/physiology , Male , Mice , Mice, Transgenic , Recombinant Proteins , Skin/injuries , Transgenes/physiology
20.
Nat Rev Clin Oncol ; 8(6): 344-56, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21487419

ABSTRACT

Brain metastases are a serious obstacle in the treatment of patients with solid tumors and contribute to the morbidity and mortality of these cancers. It is speculated that the frequency of brain metastasis is increasing for several reasons, including improved systemic therapy and survival, and detection of metastases in asymptomatic patients. The lack of preclinical models that recapitulate the clinical setting and the exclusion of patients with brain metastases from most clinical trials have slowed progress. Molecular factors contributing to brain metastases are being elucidated, such as genes involved in cell adhesion, extravasation, metabolism, and cellular signaling. Furthermore, the role of the unique brain microenvironment is beginning to be explored. Although the presence and function of the blood-brain barrier in metastatic tumors is still poorly understood, it is likely that some tumor cells are protected from therapeutics by the blood-tumor barrier, creating a sanctuary site. This Review discusses what is known about the biology of brain metastases, what preclinical models are available to study the disease, and which novel therapeutic strategies are being studied in patients.


Subject(s)
Blood-Brain Barrier , Bone Neoplasms/secondary , Bone Neoplasms/therapy , Neoplasms/pathology , Neoplasms/therapy , Animals , Disease Models, Animal , Humans
SELECTION OF CITATIONS
SEARCH DETAIL