Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
JCI Insight ; 9(10)2024 May 22.
Article in English | MEDLINE | ID: mdl-38775153

ABSTRACT

Small cell lung cancer (SCLC) is the most aggressive lung cancer entity with an extremely limited therapeutic outcome. Most patients are diagnosed at an extensive stage. However, the molecular mechanisms driving SCLC invasion and metastasis remain largely elusive. We used an autochthonous SCLC mouse model and matched samples from patients with primary and metastatic SCLC to investigate the molecular characteristics of tumor metastasis. We demonstrate that tumor cell invasion and liver metastasis in SCLC are triggered by an Angiopoietin-2 (ANG-2)/Integrin ß-1-dependent pathway in tumor cells, mediated by focal adhesion kinase/Src kinase signaling. Strikingly, CRISPR-Cas9 KO of Integrin ß-1 or blocking Integrin ß-1 signaling by an anti-ANG-2 treatment abrogates liver metastasis formation in vivo. Interestingly, analysis of a unique collection of matched samples from patients with primary and metastatic SCLC confirmed a strong increase of Integrin ß-1 in liver metastasis in comparison with the primary tumor. We further show that ANG-2 blockade combined with PD-1-targeted by anti-PD-1 treatment displays synergistic treatment effects in SCLC. Together, our data demonstrate a fundamental role of ANG-2/Integrin ß-1 signaling in SCLC cells for tumor cell invasion and liver metastasis and provide a potentially new effective treatment strategy for patients with SCLC.


Subject(s)
Angiopoietin-2 , Integrin beta1 , Liver Neoplasms , Lung Neoplasms , Signal Transduction , Small Cell Lung Carcinoma , Animals , Female , Humans , Male , Mice , Angiopoietin-2/metabolism , Angiopoietin-2/genetics , Cell Line, Tumor , Integrin beta1/metabolism , Integrin beta1/genetics , Liver Neoplasms/secondary , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/drug therapy , Lung Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/drug therapy
2.
Cancer Sci ; 115(5): 1505-1519, 2024 May.
Article in English | MEDLINE | ID: mdl-38476010

ABSTRACT

The fibrotic tumor microenvironment is a pivotal therapeutic target. Nintedanib, a clinically approved multikinase antifibrotic inhibitor, is effective against lung adenocarcinoma (ADC) but not squamous cell carcinoma (SCC). Previous studies have implicated the secretome of tumor-associated fibroblasts (TAFs) in the selective effects of nintedanib in ADC, but the driving factor(s) remained unidentified. Here we examined the role of tissue inhibitor of metalloproteinase-1 (TIMP-1), a tumor-promoting cytokine overproduced in ADC-TAFs. To this aim, we combined genetic approaches with in vitro and in vivo preclinical models based on patient-derived TAFs. Nintedanib reduced TIMP-1 production more efficiently in ADC-TAFs than SCC-TAFs through a SMAD3-dependent mechanism. Cell culture experiments indicated that silencing TIMP1 in ADC-TAFs abolished the therapeutic effects of nintedanib on cancer cell growth and invasion, which were otherwise enhanced by the TAF secretome. Consistently, co-injecting ADC cells with TIMP1-knockdown ADC-TAFs into immunocompromised mice elicited a less effective reduction of tumor growth and invasion under nintedanib treatment compared to tumors bearing unmodified fibroblasts. Our results unveil a key mechanism underlying the selective mode of action of nintedanib in ADC based on the excessive production of TIMP-1 in ADC-TAFs. We further pinpoint reduced SMAD3 expression and consequent limited TIMP-1 production in SCC-TAFs as key for the resistance of SCC to nintedanib. These observations strongly support the emerging role of TIMP-1 as a critical regulator of therapy response in solid tumors.


Subject(s)
Adenocarcinoma of Lung , Cancer-Associated Fibroblasts , Indoles , Lung Neoplasms , Smad3 Protein , Tissue Inhibitor of Metalloproteinase-1 , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/drug effects , Humans , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Mice , Indoles/pharmacology , Indoles/therapeutic use , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/genetics , Smad3 Protein/metabolism , Cell Line, Tumor , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Female
3.
Front Oncol ; 13: 1145999, 2023.
Article in English | MEDLINE | ID: mdl-37234980

ABSTRACT

Background: Gastric adenocarcinoma (GAC) is the fourth leading cause of cancer death worldwide. Systemic chemotherapy is a preferred treatment option for advanced and recurrent GAC, but response rates and survival prolongation remain limited. Tumor angiogenesis plays a critical role in GAC growth, invasion and metastasis. We investigated the antitumor efficacy of nintedanib, a potent triple angiokinase inhibitor for VEGFR-1/2/3, PDGFR-α/ß and FGFR-1/2/3, alone or in combination with chemotherapy, in preclinical models of GAC. Methods: Animal survival studies were performed in peritoneal dissemination xenografts in NOD/SCID mice using human GAC cell lines MKN-45 and KATO-III. Tumor growth inhibition studies were performed in subcutaneous xenografts in NOD/SCID mice using human GAC cell lines MKN-45 and SNU-5. The mechanistic evaluation involved Immunohistochemistry analyses in tumor tissues obtained from subcutaneous xenografts. In vitro cell viability assays were performed using a colorimetric WST-1 reagent. Results: In MKN-45 GAC cell-derived peritoneal dissemination xenografts, animal survival was improved by nintedanib (33%), docetaxel (100%) and irinotecan (181%), while oxaliplatin, 5-FU and epirubicin had no effect. The addition of nintedanib to docetaxel (157%) or irinotecan (214%) led to a further extension in animal survival. In KATO-III GAC cell-derived xenografts carrying FGFR2 gene amplification, nintedanib extended survival by 209%. Again, the addition of nintedanib further enhanced the animal survival benefits of docetaxel (273%) and irinotecan (332%). In MKN-45 subcutaneous xenografts, nintedanib, epirubicin, docetaxel and irinotecan reduced tumor growth (range: 68-87%), while 5-FU and oxaliplatin had a smaller effect (40%). Nintedanib addition to all chemotherapeutics demonstrated a further reduction in tumor growth. Subcutaneous tumor analysis revealed that nintedanib attenuated tumor cell proliferation, reduced tumor vasculature and increased tumor cell death. Conclusion: Nintedanib showed notable antitumor efficacy and significantly improved taxane or irinotecan chemotherapy responses. These findings indicate that nintedanib, alone and in combination with a taxane or irinotecan, has the potential for improving clinical GAC therapy.

4.
Mol Cancer Ther ; 22(5): 616-629, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36805958

ABSTRACT

Bladder cancer is a highly prevalent tumor, requiring the urgent development of novel therapies, especially for locally advanced and metastatic disease. Nintedanib is a potent antifibrotic angio-kinase inhibitor, which has shown clinical efficacy in combination with chemotherapy in patients with locally advanced muscle-invasive bladder cancer. Nintedanib inhibits fibroblast growth factor receptors (FGFRs), validated targets in patients with bladder cancer harboring FGFR3/2 genetic alterations. Here, we aimed at studying its mechanisms of action to understand therapy resistance, identify markers predictive of response, and improve the design of future clinical trials. We have used a panel of genetically well-characterized human bladder cancer cells to identify the molecular and transcriptomic changes induced upon treatment with nintedanib, in vitro and in vivo, at the tumor and stroma cell levels. We showed that bladder cancer cells display an intrinsic resistance to nintedanib treatment in vitro, independently of their FGFR3 status. However, nintedanib has higher antitumor activity on mouse xenografts. We have identified PI3K activation as a resistance mechanism against nintedanib in bladder cancer and evidenced that the combination of nintedanib with the PI3K inhibitor alpelisib has synergistic antitumor activity. Treatment with this combination is associated with cell-cycle inhibition at the tumoral and stromal levels and potent nontumor cell autonomous effects on α-smooth muscle actin-positive tumor infiltrating cells and tumor vasculature. The combination of nintedanib with PI3K inhibitors not only reversed bladder cancer resistance to nintedanib but also enhanced its antiangiogenic effects.


Subject(s)
Lung Neoplasms , Urinary Bladder Neoplasms , Humans , Mice , Animals , Phosphatidylinositol 3-Kinases/therapeutic use , Lung Neoplasms/drug therapy , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Stromal Cells , Cell Line, Tumor
5.
J Pharmacol Exp Ther ; 384(3): 331-342, 2023 03.
Article in English | MEDLINE | ID: mdl-36241203

ABSTRACT

Vascular endothelial growth factor (VEGF) and angiopoietin (ANG)-2 have complementary roles in angiogenesis and promote an immunosuppressive tumor microenvironment. It is anticipated that the combination of VEGF and ANG2 blockade could provide superior activity to the blockade of either pathway alone and that the addition of VEGF/ANG2 inhibition to an anti-programmed cell death protein-1 (PD-1) antibody could change the tumor microenvironment to support T-cell-mediated tumor cytotoxicity. Here, we describe the pharmacologic and antitumor activity of BI 836880, a humanized bispecific nanobody comprising two single-variable domains blocking VEGF and ANG2, and an additional module for half-life extension in vivo. BI 836880 demonstrated high affinity and selectivity for human VEGF-A and ANG2, resulting in inhibition of the downstream signaling of VEGF/ANG2 and a decrease in endothelial cell proliferation and survival. In vivo, BI 836880 exhibited significant antitumor activity in all patient-derived xenograft models tested, showing significantly greater tumor growth inhibition (TGI) than bevacizumab (VEGF inhibition) and AMG386 (ANG1/2 inhibition) in a range of models. In a Lewis lung carcinoma syngeneic tumor model, the combination of PD-1 inhibition with VEGF inhibition showed superior efficacy versus the blockade of either pathway alone. TGI was further increased with the addition of ANG2 inhibition to VEGF/PD-1 blockade. VEGF/ANG2 inhibition had a strong antiangiogenic effect. Our data suggest that the blockade of VEGF and ANG2 with BI 836880 may offer improved antitumor activity versus the blockade of either pathway alone and that combining VEGF/ANG2 inhibition with PD-1 blockade can further enhance antitumor effects. SIGNIFICANCE STATEMENT: Vascular endothelial growth factor (VEGF) and angiopoietin (ANG)-2 play key roles in angiogenesis and have an immunosuppressive effect in the tumor microenvironment. This study shows that BI 836880, a bispecific nanobody targeting VEGF and ANG2, demonstrates substantial antitumor activity in preclinical models. Combining VEGF/ANG2 inhibition with the blockade of the PD-1 pathway can further improve antitumor activity.


Subject(s)
Neoplasms , Vascular Endothelial Growth Factor A , Humans , Vascular Endothelial Growth Factor A/metabolism , Angiopoietin-2/metabolism , Programmed Cell Death 1 Receptor , Vascular Endothelial Growth Factors/therapeutic use , Angiogenesis Inhibitors , Neoplasms/drug therapy , Cell Death , Angiopoietin-1 , Tumor Microenvironment
6.
Blood ; 137(15): 2070-2084, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33512435

ABSTRACT

The KIT D816V mutation is found in >80% of patients with systemic mastocytosis (SM) and is key to neoplastic mast cell (MC) expansion and accumulation in affected organs. Therefore, KIT D816V represents a prime therapeutic target for SM. Here, we generated a panel of patient-specific KIT D816V induced pluripotent stem cells (iPSCs) from patients with aggressive SM and mast cell leukemia to develop a patient-specific SM disease model for mechanistic and drug-discovery studies. KIT D816V iPSCs differentiated into neoplastic hematopoietic progenitor cells and MCs with patient-specific phenotypic features, thereby reflecting the heterogeneity of the disease. CRISPR/Cas9n-engineered KIT D816V human embryonic stem cells (ESCs), when differentiated into hematopoietic cells, recapitulated the phenotype observed for KIT D816V iPSC hematopoiesis. KIT D816V causes constitutive activation of the KIT tyrosine kinase receptor, and we exploited our iPSCs and ESCs to investigate new tyrosine kinase inhibitors targeting KIT D816V. Our study identified nintedanib, a US Food and Drug Administration-approved angiokinase inhibitor that targets vascular endothelial growth factor receptor, platelet-derived growth factor receptor, and fibroblast growth factor receptor, as a novel KIT D816V inhibitor. Nintedanib selectively reduced the viability of iPSC-derived KIT D816V hematopoietic progenitor cells and MCs in the nanomolar range. Nintedanib was also active on primary samples of KIT D816V SM patients. Molecular docking studies show that nintedanib binds to the adenosine triphosphate binding pocket of inactive KIT D816V. Our results suggest nintedanib as a new drug candidate for KIT D816V-targeted therapy of advanced SM.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Mastocytosis, Systemic/drug therapy , Point Mutation/drug effects , Proto-Oncogene Proteins c-kit/genetics , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/pathology , Tumor Cells, Cultured
7.
Mol Cancer Ther ; 20(1): 96-108, 2021 01.
Article in English | MEDLINE | ID: mdl-33037135

ABSTRACT

Activation of TRAILR2 has emerged as an important therapeutic concept in cancer treatment. TRAILR2 agonistic molecules have only had limited clinical success, to date, due either to lack of efficacy or hepatotoxicity. BI 905711 is a novel tetravalent bispecific antibody targeting both TRAILR2 and CDH17 and represents a novel liver-sparing TRAILR2 agonist specifically designed to overcome the disadvantages of previous strategies. Here, we show that BI 905711 effectively triggered apoptosis in a broad panel of CDH17-positive colorectal cancer tumor cells in vitro. Efficient induction of apoptosis was dependent on the presence of CDH17, as exemplified by the greater than 1,000-fold drop in potency in CDH17-negative cells. BI 905711 demonstrated single-agent tumor regressions in CDH17-positive colorectal cancer xenografts, an effect that was further enhanced upon combination with irinotecan. Antitumor efficacy correlated with induction of caspase activation, as measured in both the tumor and plasma. Effective tumor growth inhibition was further demonstrated across a series of different colorectal cancer PDX models. BI 905711 induced apoptosis in both a cis (same cell) as well as trans (adjacent cell) fashion, translating into significant antitumor activity even in xenograft models with heterogeneous CDH17 expression. In summary, we demonstrate that BI 905711 has potent and selective antitumor activity in CDH17-positive colorectal cancer models both in vitro and in vivo. The high prevalence of over 95% CDH17-positive tumors in patients with colorectal cancer, the molecule preclinical efficacy together with its potential for a favorable safety profile, support the ongoing BI 905711 phase I trial in colorectal cancer and additional CDH17-positive cancer types (NCT04137289).


Subject(s)
Antibodies, Bispecific/pharmacology , Apoptosis , Cadherins/metabolism , Colorectal Neoplasms/pathology , Liver/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Xenograft Model Antitumor Assays , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Humans , Liver/drug effects , Mice , Neoplasm Metastasis , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Remission Induction
8.
Clin Exp Metastasis ; 37(6): 637-648, 2020 12.
Article in English | MEDLINE | ID: mdl-32918638

ABSTRACT

Brain metastases (BM) are an ever-increasing challenge in oncology, threatening quality of life and survival of many cancer patients. The majority of BM originate from lung adenocarcinoma, and stage III patients have a risk of 40-50% to develop BM in the first years of disease onset. As therapeutic options are limited, prevention of their occurrence is an attractive concept. Here we investigated whether Nintedanib (BIBF 1120), a tyrosine kinase inhibitor (TKI) targeting the VEGF pathway approved for lung adenocarcinoma, and the dual anti-VEGF-A/Ang2 nanobody BI836880 have the potential to prevent BM formation. A mouse model of brain metastasis from lung adenocarcinoma was used in which tumor cells were injected intracardially. Metastases formation occurred inside and outside of the brain and was followed by MRI, IVIS, and immunohistochemistry. BM were reduced in volume and number by both Nintedanib and the dual anti-VEGF-A/Ang2 nanobody, which translated into improved survival. Both compounds were able to normalize cerebral blood vessels at the site of brain metastatic lesions. Extracranial metastases, however, were not reduced, and meningeal metastases only partially. Interestingly, unspecific control IgG also lead to brain vessel normalization and reduction of brain and meningeal metastases. This data indicates a brain-specific group effect of antiangiogenic compounds with respect to metastasis prevention, most likely by preventing an early angiogenic switch. Thus, Nintedanib and BI836880 are promising candidates for future BM preventive study concepts in lung adenocarcinoma patients.


Subject(s)
Adenocarcinoma of Lung/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/secondary , Brain Neoplasms/therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vesicular Transport Proteins/antagonists & inhibitors , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Angiogenesis Inhibitors/administration & dosage , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/prevention & control , Cell Line, Tumor , Humans , Immunoglobulin G/administration & dosage , Indoles/administration & dosage , Male , Mice , Mice, Inbred NOD , Mice, SCID , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/immunology , Vascular Endothelial Growth Factor A/immunology , Vesicular Transport Proteins/immunology , Xenograft Model Antitumor Assays
9.
Cancer Res ; 80(2): 276-290, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31694906

ABSTRACT

The tumor-promoting fibrotic stroma rich in tumor-associated fibroblasts (TAF) is drawing increased therapeutic attention. Intriguingly, a trial with the antifibrotic drug nintedanib in non-small cell lung cancer reported clinical benefits in adenocarcinoma (ADC) but not squamous cell carcinoma (SCC), even though the stroma is fibrotic in both histotypes. Likewise, we reported that nintedanib inhibited the tumor-promoting fibrotic phenotype of TAFs selectively in ADC. Here we show that tumor fibrosis is actually higher in ADC-TAFs than SCC-TAFs in vitro and patient samples. Mechanistically, the reduced fibrosis and nintedanib response of SCC-TAFs was associated with increased promoter methylation of the profibrotic TGFß transcription factor SMAD3 compared with ADC-TAFs, which elicited a compensatory increase in TGFß1/SMAD2 activation. Consistently, forcing global DNA demethylation of SCC-TAFs with 5-AZA rescued TGFß1/SMAD3 activation, whereas genetic downregulation of SMAD3 in ADC-TAFs and control fibroblasts increased TGFß1/SMAD2 activation, and reduced their fibrotic phenotype and antitumor responses to nintedanib in vitro and in vivo. Our results also support that smoking and/or the anatomic location of SCC in the proximal airways, which are more exposed to cigarette smoke particles, may prime SCC-TAFs to stronger SMAD3 epigenetic repression, because cigarette smoke condensate selectively increased SMAD3 promoter methylation. Our results unveil that the histotype-specific regulation of tumor fibrosis in lung cancer is mediated through differential SMAD3 promoter methylation in TAFs and provide new mechanistic insights on the selective poor response of SCC-TAFs to nintedanib. Moreover, our findings support that patients with ADC may be more responsive to antifibrotic drugs targeting their stromal TGFß1/SMAD3 activation. SIGNIFICANCE: This study implicates the selective epigenetic repression of SMAD3 in SCC-TAFs in the clinical failure of nintedanib in SCC and supports that patients with ADC may benefit from antifibrotic drugs targeting stromal TGFß1/SMAD3.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/genetics , Indoles/pharmacology , Lung Neoplasms/drug therapy , Smad3 Protein/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/surgery , Aged , Aged, 80 and over , Animals , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Cohort Studies , DNA Methylation/genetics , Epigenetic Repression , Female , Fibrosis , Gene Expression Regulation, Neoplastic , Humans , Indoles/therapeutic use , Lung/cytology , Lung/drug effects , Lung/pathology , Lung/surgery , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Mice , Middle Aged , Pneumonectomy , Promoter Regions, Genetic/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Tissue Array Analysis , Xenograft Model Antitumor Assays
10.
Angiogenesis ; 22(4): 535-546, 2019 11.
Article in English | MEDLINE | ID: mdl-31463627

ABSTRACT

In contrast to VEGF pathway-targeting antibodies, antiangiogenic tyrosine kinase inhibitors (TKIs) have failed to meet primary endpoints in almost all phase III clinical trials when combined with conventional chemotherapy. One exception is the combination of nintedanib and docetaxel as a second-line therapy for rapidly progressing advanced NSCLC. In addition to increased toxicity caused by this type of combination, thus necessitating drug dose reductions or treatment breaks, such phase III trial failures may also be related to the differential impact of host-mediated responses involving mobilization and tumor infiltration of bone marrow-derived cell populations (BMDCs), comprising both pro-angiogenic as well as immune effector cells. Herein, we evaluated two different antiangiogenic TKIs (sunitinib or nintedanib) and a VEGFR-2 antibody (DC101) either alone or combined with maximum tolerated dose paclitaxel for their differential impact on the BMDC host response, evaluating four different cell types. TKIs (in particular sunitinib) induced myelosuppression similar to paclitaxel, whereas DC101 had no such effect. Sunitinib also significantly decreased the number of tumor-infiltrating CD8 + T and B cells, MDSCs, and macrophages. In contrast, the effect of nintedanib on these BMDC populations was less marked, behaving closer to the VEGFR-2 antibody effects than sunitinib. The results raise the possibility that differences observed between antiangiogenic antibodies and TKIs in increasing chemotherapy efficacy could be related, at least in part, to differential effects on cells associated with local immunity within the tumor microenvironment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Bone Marrow Cells , Immune Tolerance/drug effects , Indoles/pharmacology , Sunitinib/pharmacology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Drug Screening Assays, Antitumor , Mice , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Rats
11.
J Biol Chem ; 294(27): 10428-10437, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31118272

ABSTRACT

RET is a transmembrane growth factor receptor. Aberrantly activated RET is found in several types of human cancer and is a target for treating RET aberration-associated cancer. Multiple clinically relevant RET protein-tyrosine kinase inhibitors (TKIs) have been identified, but how TKIs bind to RET is unknown except for vandetanib. Nintedanib is a RET TKI that inhibits the vandetanib-resistant RET(G810A) mutant. Here, we determined the X-ray co-crystal structure of RET kinase domain-nintedanib complex to 1.87 Å resolution and a RET(G810A) kinase domain crystal structure to 1.99 Å resolution. We also identified a vandetanib-resistant RET(L881V) mutation previously found in familial medullary thyroid carcinoma. Drug-sensitivity profiling of RET(L881V) revealed that it remains sensitive to nintedanib. The RET-nintedanib co-crystal structure disclosed that Leu-730 in RET engages in hydrophobic interactions with the piperazine, anilino, and phenyl groups of nintedanib, providing a structural basis for explaining that the p.L730V mutation identified in nine independently isolated cell lines resistant to nintedanib. Comparisons of RET-nintedanib, RET(G810A), and RET-vandetanib crystal structures suggested that the solvent-front Ala-810 makes hydrophobic contacts with a methyl group and aniline in nintedanib and blocks water access to two oxygen atoms of vandetanib, resulting in an energetic penalty for burying polar groups. Of note, even though the p.L881V mutation did not affect sensitivity to nintedanib, RET(L881V) was resistant to nintedanib analogs lacking a phenyl group. These results provide structural insights into resistance of RET mutants against the TKIs nintedanib and vandetanib.


Subject(s)
Indoles/chemistry , Piperidines/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-ret/chemistry , Quinazolines/chemistry , Animals , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Drug Resistance, Neoplasm , Humans , Hydrophobic and Hydrophilic Interactions , Indoles/metabolism , Mice , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Piperidines/metabolism , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Quinazolines/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
12.
Future Oncol ; 15(12): 1363-1383, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30758227

ABSTRACT

A substantial proportion of patients with nononcogene-addicted non-small-cell lung cancer (NSCLC) has 'aggressive disease', as reflected in short time to progression or lack of disease control with initial platinum-based chemotherapy. Recently, clinical correlates of aggressive disease behavior during first-line therapy have been shown to predict greater benefit from addition of nintedanib to second-line docetaxel in adenocarcinoma NSCLC. Positive predictive effects of aggressive disease have since been reported with other anti-angiogenic agents (ramucirumab and bevacizumab), while such features may negatively impact on outcomes with nivolumab in nonsquamous NSCLC with low PD-L1 expression. Based on a review of the clinical data, we recommend aggressive nonsquamous NSCLC should be defined by progression within <6-9 months of first-line treatment initiation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Lung/pathology , Patient Selection , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bevacizumab/therapeutic use , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Disease Progression , Disease-Free Survival , Docetaxel/therapeutic use , Humans , Indoles/therapeutic use , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Time Factors , Ramucirumab
13.
J Exp Clin Cancer Res ; 38(1): 16, 2019 Jan 11.
Article in English | MEDLINE | ID: mdl-30635009

ABSTRACT

BACKGROUND: Triple negative breast cancer (TNBC) is an aggressive malignancy with poor prognosis, in part because of the current lack of any approved molecularly targeted therapy. We evaluated various combinations of three different drugs: nintedanib, an antiangiogenic TKI targeting VEGF receptors, paclitaxel (PTX), or a PD-L1 antibody, using models of orthotopic primary or advanced metastatic TNBC involving a metastatic variant of the MDA-MB-231 human cell line (called LM2-4) in SCID mice and two mouse lines (EMT-6 and a drug-resistant variant, EMT-6/CDDP) in immunocompetent mice. These drugs were selected based on the following: PTX is approved for TNBC; nintedanib combined with docetaxel has shown phase III clinical trial success, albeit in NSCLC; VEGF can act as local immunosuppressive factor; and PD-L1 antibody plus taxane therapy was recently reported to have encouraging phase III trial benefit in TNBC. METHODS: Statistical analyses were performed with ANOVA followed by Tukey's Multiple Comparison Test or with Kruskal-Wallis test followed by Dunn's Multiple Comparison Test. Survival curves were analyzed using a Log-rank (Mantel Cox) test. Differences were considered statistically significant when p values were < 0.05. RESULTS: Toxicity analyses showed that nintedanib is well tolerated when administered 5-days ON 2-days OFF; PTX toxicity differed in mice, varied with cell lines used and may have influenced median survival in the metastatic EMT6/CDDP model; while toxicity of PD-L1 therapy depended on the cell lines and treatment settings tested. In the LM2-4 system, combining nintedanib with PTX enhanced overall antitumor efficacy in both primary and metastatic treatment settings. In immunocompetent mice, combining nintedanib or PTX with the PD-L1 antibody improved overall antitumor efficacy. Using the advanced metastatic EMT-6/CDDP model, optimal efficacy results were obtained using the triple combination. CONCLUSIONS: These results suggest circumstances where nintedanib plus PTX may be potentially effective in treating TNBC, and nintedanib with PTX may improve PD-L1 therapy of metastatic TNBC.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Indoles/pharmacology , Paclitaxel/pharmacology , Animals , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Kaplan-Meier Estimate , Mice , Molecular Targeted Therapy , Neoplasm Metastasis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
14.
Br J Cancer ; 120(1): 69-78, 2019 01.
Article in English | MEDLINE | ID: mdl-30377339

ABSTRACT

BACKGROUND: Immune infiltration is implicated in the development of acquired resistance to anti-angiogenic cancer therapy. We therefore investigated the correlation between neutrophil infiltration in metastasis of colorectal cancer (CRC) patients and survival after treatment with bevacizumab. Our study identifies CD177+ tumour neutrophil infiltration as an adverse prognostic factor for bevacizumab treatment. We further demonstrate that a novel anti-VEGF/anti-Ang2 compound (BI-880) can overcome resistance to VEGF inhibition in experimental tumour models. METHODS: A total of 85 metastatic CRC patients were stratified into cohorts that had either received chemotherapy alone (n = 39) or combined with bevacizumab (n = 46). Tumour CD177+ neutrophil infiltration was correlated to clinical outcome. The impact of neutrophil infiltration on anti-VEGF or anti-VEGF/anti-Ang2 therapy was studied in both xenograft and syngeneic tumour models by immunohistochemistry. RESULTS: The survival of bevacizumab-treated CRC patients in the presence of CD177+ infiltrates was significantly reduced compared to patients harbouring CD177- metastases. BI-880 treatment reduced the development of hypoxia associated with bevacizumab treatment and improved vascular normalisation in xenografts. Furthermore, neutrophil depletion or BI-880 treatment restored treatment sensitivity in a syngeneic tumour model of anti-VEGF resistance. CONCLUSIONS: Our findings implicate CD177 as a biomarker for bevacizumab and suggest VEGF/Ang2 inhibition as a strategy to overcome neutrophil associated resistance to anti-angiogenic treatment.


Subject(s)
Colorectal Neoplasms/drug therapy , Isoantigens/genetics , Neovascularization, Pathologic/drug therapy , Receptors, Cell Surface/genetics , Vascular Endothelial Growth Factor A/genetics , Vesicular Transport Proteins/genetics , Aged , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Bevacizumab/administration & dosage , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Female , GPI-Linked Proteins/genetics , Humans , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neutrophils/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays
15.
Br J Pharmacol ; 175(17): 3504-3515, 2018 09.
Article in English | MEDLINE | ID: mdl-29908090

ABSTRACT

BACKGROUND AND PURPOSE: Alterations in the tyrosine kinase enzyme RET are found in thyroid and lung cancer. While RET TK inhibitors (TKIs) are used to treat thyroid cancer and are in clinical trials for RET fusion-positive non-small cell lung cancer, the impact of mutations in the RET kinase domain on drug sensitivity is largely uncharacterized. EXPERIMENTAL APPROACH: We identified and analysed mutations in the RET kinase domain that conferred resistance to the TKIs cabozantinib, lenvatinib, vandetanib and nintedanib using RET kinase-dependent BaF3/KIF5B-RET (BaF3/KR) cells. We also examined the sensitivity of RET (M918T), a RET mutation prevalent in aggressive multiple endocrine neoplasia type 2B, to these TKIs in the context of BaF3/KR cells. KEY RESULTS: Fourteen mutations were analysed. Pan resistance to the four TKIs was found in six RET kinase domain mutations (L730I, V738A, V804L/M, Y806N, G810S). Seven RET kinase domain mutations (L730V, E732K, A807V, G810A, V871I, M918T, F998V) displayed selective resistance to one or more of these drugs. L730I/V and G810A/S had different drug resistance profiles. V871I, M918T and F998V mutations are located at distant sites away from the TKI binding pocket. CONCLUSIONS AND IMPLICATIONS: A panel of TKI-resistant RET mutations were identified, and their drug sensitivities were cross-profiled. The results provide a reference for selecting appropriate TKIs to inhibit RET kinase domain mutants. Besides changes in the drug-interacting residues, mutations at distant sites could exert long-range effects resulting in TKI resistance. Among the four TKIs analysed here, nintedanib remained unaffected by mutations at the three distant sites.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-ret/genetics , Antineoplastic Agents/therapeutic use , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics
16.
Clin Cancer Res ; 24(15): 3729-3740, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29724868

ABSTRACT

Purpose: Malignant pleural mesothelioma (MPM) is an aggressive thoracic tumor type with limited treatment options and poor prognosis. The angiokinase inhibitor nintedanib has shown promising activity in the LUME-Meso phase II MPM trial and thus is currently being evaluated in the confirmatory LUME-Meso phase III trial. However, the anti-MPM potential of nintedanib has not been studied in the preclinical setting.Experimental Design: We have examined the antineoplastic activity of nintedanib in various in vitro and in vivo models of human MPM.Results: Nintedanib's target receptors were (co)expressed in all the 20 investigated human MPM cell lines. Nintedanib inhibited MPM cell growth in both short- and long-term viability assays. Reduced MPM cell proliferation and migration and the inhibition of Erk1/2 phosphorylation were also observed upon nintedanib treatment in vitro Additive effects on cell viability were detected when nintedanib was combined with cisplatin, a drug routinely used for systemic MPM therapy. In an orthotopic mouse model of human MPM, survival of animals receiving nintedanib per os showed a favorable trend, but no significant benefit. Nintedanib significantly reduced tumor burden and vascularization and prolonged the survival of mice when it was administered intraperitoneally. Importantly, unlike bevacizumab, nintedanib demonstrated significant in vivo antivascular and antitumor potential independently of baseline VEGF-A levels.Conclusions: Nintedanib exerts significant antitumor activity in MPM both in vitro and in vivo These data provide preclinical support for the concept of LUME-Meso trials evaluating nintedanib in patients with unresectable MPM. Clin Cancer Res; 24(15); 3729-40. ©2018 AACR.


Subject(s)
Indoles/pharmacology , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Neovascularization, Pathologic/drug therapy , Pleural Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mesothelioma/genetics , Mesothelioma/pathology , Mesothelioma, Malignant , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Phosphorylation/drug effects , Pleural Neoplasms/genetics , Pleural Neoplasms/pathology , Xenograft Model Antitumor Assays
17.
J Pharmacol Exp Ther ; 364(3): 494-503, 2018 03.
Article in English | MEDLINE | ID: mdl-29263244

ABSTRACT

The triple-angiokinase inhibitor nintedanib is an orally available, potent, and selective inhibitor of tumor angiogenesis by blocking the tyrosine kinase activities of vascular endothelial growth factor receptor (VEGFR) 1-3, platelet-derived growth factor receptor (PDGFR)-α and -ß, and fibroblast growth factor receptor (FGFR) 1-3. Nintedanib has received regulatory approval as second-line treatment of adenocarcinoma non-small cell lung cancer (NSCLC), in combination with docetaxel. In addition, nintedanib has been approved for the treatment of idiopathic lung fibrosis. Here we report the results from a broad kinase screen that identified additional kinases as targets for nintedanib in the low nanomolar range. Several of these kinases are known to be mutated or overexpressed and are involved in tumor development (discoidin domain receptor family, member 1 and 2, tropomyosin receptor kinase A (TRKA) and C, rearranged during transfection proto-oncogene [RET proto oncogene]), as well as in fibrotic diseases (e.g., DDRs). In tumor cell lines displaying molecular alterations in potential nintedanib targets, the inhibitor demonstrates direct antiproliferative effects: in the NSCLC cell line NCI-H1703 carrying a PDGFRα amplification (ampl.); the gastric cancer cell line KatoIII and the breast cancer cell line MFM223, both driven by a FGFR2 amplification; AN3CA (endometrial carcinoma) bearing a mutated FGFR2; the acute myeloid leukemia cell lines MOLM-13 and MV-4-11-B with FLT3 mutations; and the NSCLC adenocarcinoma LC-2/ad harboring a CCDC6-RET fusion. Potent kinase inhibition does not, however, strictly translate into antiproliferative activity, as demonstrated in the TRKA-dependent cell lines CUTO-3 and KM-12. Importantly, nintedanib treatment of NCI-H1703 tumor xenografts triggered effective tumor shrinkage, indicating a direct effect on the tumor cells in addition to the antiangiogenic effect on the tumor stroma. These findings will be instructive in guiding future genome-based clinical trials of nintedanib.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Lung Neoplasms/pathology , Mice , Mutation , Oncogenes/genetics , Proto-Oncogene Mas , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
18.
Invest Ophthalmol Vis Sci ; 56(13): 7897-907, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26670826

ABSTRACT

PURPOSE: This study evaluated the effects of BIBF1120, a novel triple angiokinase inhibitor against pathological retinal neovascularization. METHODS: BIBF1120 effect on development of the normal retinal vasculature was evaluated in Sprague-Dawley rat pups. Two models of ischemic oxygen-induced retinopathy (OIR) and the aortic ring assay were used to assess the antiangiogenic effects of BIBF1120. In the vaso-obliteration model (VO), rat pups were exposed to 80% O2 from postnatal day (P) 5 to P10. In the preretinal neovascularization (NV) model, rat pups were exposed to cycling O2 (50% and 10%) from P1 to P14, followed by room air until P18. Animals were intravitreally or orally treated with BIBF1120. Retinal vasculature, VO, and NV were evaluated in retinal flat mounts. Retinal expression of VEGF, Delta-like ligand 4 (Dll4), Netrin-1, Ephrin-B2, and EphB4 was analyzed by quantitative PCR and Western blot analysis. RESULTS: BIBF1120 interfered with normal retinal vascular development and microvessel branching in the aortic assay. However, in VO model BIBF1120 did not accrue VO. On the contrary, in the NV model BIBF1120 accelerated normal retinal vascularization and robustly diminished preretinal neovascularization compared to vehicle (by ~80%). The expression levels of VEGF negative regulator Dll4 and repulsive cues EphrinB2 and EphB4 mRNA in the retina of vehicle-treated OIR animals were markedly increased compared to normoxia, but were normalized by BIBF1120. CONCLUSIONS: Data reveal efficacy of BIBF1120 on preretinal neovascularization and, of greater interest, on acceleration of normal vascularization, consistent with interference of major repulsive cues expressed in the retina during OIR. Accordingly, BIBF1120 appears to exhibit preferable properties compared to anti-VEGF therapies for the treatment of ischemic retinopathies.


Subject(s)
Indoles/therapeutic use , Neovascularization, Physiologic/drug effects , Retinal Neovascularization/drug therapy , Retinopathy of Prematurity/drug therapy , Animals , Animals, Newborn , Disease Models, Animal , Female , Rats , Rats, Sprague-Dawley , Retinal Neovascularization/pathology , Retinopathy of Prematurity/pathology
19.
Clin Cancer Res ; 21(21): 4856-67, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26206868

ABSTRACT

PURPOSE: Pancreatic neuroendocrine tumors (PNET) represent a rare but challenging heterogeneous group of cancers with an increasing incidence over the last number of decades. Herein, we report an in-depth evaluation of the new antiangiogenic small-molecule tyrosine kinase inhibitor (TKI) nintedanib in the preclinical Rip1Tag2 transgenic mouse model of neuroendocrine carcinoma of the pancreas (insulinoma). EXPERIMENTAL DESIGN: We have assessed the antiangiogenic and antitumor activity of nintedanib, in comparison with other antiangiogenic TKI, by treating Rip1Tag2 transgenic mice with different treatment schedules complemented with histopathologic, cell biologic, and biochemical analyses. RESULTS: Prolonged nintedanib treatment of Rip1Tag2 mice has led to a strong suppression of angiogenesis, accompanied by a reduced tumor burden, which translated into a significant prolongation of survival. Despite nintedanib's inhibitory action on perivascular cells, the blood vessels remaining after therapy displayed a considerably mature phenotype with tight perivascular cell coverage and preserved perfusion. Nintedanib treatment did not increase local tumor invasiveness or metastasis to the liver and pancreatic lymph nodes--a phenomenon that has been observed with antiangiogenic treatments of Rip1Tag2 transgenic mice in other laboratories. In contrast with the strong reduction in blood microvessel densities, nintedanib did not have any impact on tumor lymphangiogenesis. CONCLUSIONS: Based on our findings, we propose the clinical evaluation of the antiangiogenic drug nintedanib as a new treatment modality for PNET patients, notably in a direct comparison with already established therapeutic regimens, such as sunitinib.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Antineoplastic Agents/pharmacology , Carcinoma, Neuroendocrine/genetics , Indoles/pharmacology , Insulin/genetics , Pancreatic Neoplasms/genetics , Promoter Regions, Genetic , Animals , Apoptosis/drug effects , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Staging , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Phenotype , Signal Transduction/drug effects , Tumor Burden , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism
20.
J Med Chem ; 58(3): 1053-63, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25474320

ABSTRACT

Nintedanib (BIBF1120) is a potent, oral, small-molecule tyrosine kinase inhibitor, also known as a triple angiokinase inhibitor, inhibiting three major signaling pathways involved in angiogenesis. Nintedanib targets proangiogenic and pro-fibrotic pathways mediated by the VEGFR family, the fibroblast growth factor receptor (FGFR) family, the platelet-derived growth factor receptor (PDGFR) family, as well as Src and Flt-3 kinases. The compound was identified during a lead optimization program for small-molecule inhibitors of angiogenesis and has since undergone extensive clinical investigation for the treatment of various solid tumors, and in patients with the debilitating lung disease idiopathic pulmonary fibrosis (IPF). Recent clinical evidence from phase III studies has shown that nintedanib has significant efficacy in the treatment of NSCLC, ovarian cancer, and IPF. This review article provides a comprehensive summary of the preclinical and clinical research and development of nintedanib from the initial drug discovery process to the latest available clinical trial data.


Subject(s)
Drug Discovery , Idiopathic Pulmonary Fibrosis/drug therapy , Indoles/pharmacology , Indoles/therapeutic use , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Female , Humans , Indoles/chemical synthesis , Indoles/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...