Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cells ; 9(2)2020 02 13.
Article in English | MEDLINE | ID: mdl-32069895

ABSTRACT

The development of novel therapies or the improvement of currently used approaches to treat prostate cancer (PCa), the most frequently diagnosed male tumor in developed countries, is an urgent need. In this regard, the functional characterization of microRNAs, molecules shown to regulate a number of cancer-related pathways, is instrumental to their possible clinical exploitation. Here, we demonstrate the tumor-suppressive role of the so far uncharacterized miR-1272, which we found to be significantly down-modulated in PCa clinical specimens compared to normal tissues. Through a gain-of-function approach using miRNA mimics, we showed that miR-1272 supplementation in two PCa cell models (DU145 and 22Rv1) reverted the mesenchymal phenotype by affecting migratory and invasive properties, and reduced cell growth in vitro and in vivo in SCID mice. Additionally, by targeting HIP1 encoding the endocytic protein HIP1, miR-1272 balanced EGFR membrane turnover, thus affecting the downstream AKT/ERK pathways, and, ultimately, increasing PCa cell response to ionizing radiation. Overall, our results show that miR-1272 reconstitution can affect several tumor traits, thus suggesting this approach as a potential novel therapeutic strategy to be pursued for PCa, with the multiple aim of reducing tumor growth, enhancing response to radiotherapy and limiting metastatic dissemination.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Heterografts , Humans , Male , Mice , Mice, SCID , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transfection
2.
Cancers (Basel) ; 11(7)2019 Jul 19.
Article in English | MEDLINE | ID: mdl-31331120

ABSTRACT

Epithelioid sarcoma (ES) is a rare mesenchymal malignancy marked by SMARCB1/INI1 deficiency. Retrospective clinical data report on the activity of anthracycline- and gemcitabine-based regimens. EZH2 inhibitors are currently being tested in clinical trials. Since comparisons of these agents are unlikely to be prospectively evaluated in the clinics, we took advantage of an INI1-deficient proximal-type ES patient-derived xenograft (PDX ES-1) to comparatively assess its preclinical antitumor activity. Mice were treated with doxorubicin and ifosfamide, singly or in combination, gemcitabine, and the EZH2 inhibitor EPZ-011989. Comparable antitumor activity (max tumor volume inhibition: ~90%) was caused by gemcitabine, EPZ-011989, and the doxorubicin-ifosfamide combination. The integration of RNAseq data, generated on tumors obtained from untreated and EPZ-011989-treated mice, and results from functional studies, carried out on the PDX-derived ES-1 cell line, revealed autophagy induction as a possible survival mechanism in residual tumor cells following EPZ-011989 treatment and identified HMGA2 as a main player in this process. Our data support the clinical use of gemcitabine and the doxorubicin-ifosfamide combination, confirm EZH2 as a therapeutic target in proximal-type ES, and suggest autophagy as a cytoprotective mechanism against EZH2 inhibition.

3.
Cancer Lett ; 415: 187-197, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29225052

ABSTRACT

Synovial sarcoma (SS) is an aggressive tumor with propensity for lung metastases which significantly impact patients' prognosis. New therapeutic approaches are needed to improve treatment outcome. Targeting the heparanase/heparan sulfate proteoglycan system by heparin derivatives which act as heparanase inhibitors/heparan sulfate mimetics is emerging as a therapeutic approach that can sensitize the tumor response to chemotherapy. We investigated the therapeutic potential of a supersulfated low molecular weight heparin (ssLMWH) in preclinical models of SS. ssLMWH showed a potent anti-heparanase activity, dose-dependently inhibited SS colony growth and cell invasion, and downregulated the activation of receptor tyrosine kinases including IGF1R and IR. The combination of ssLMWH and the IGF1R/IR inhibitor BMS754807 synergistically inhibited proliferation of cells exhibiting IGF1R hyperactivation, also abrogating cell motility and promoting apoptosis in association with PI3K/AKT pathway inhibition. The drug combination strongly enhanced the antitumor effect against the CME-1 model, as compared to single agent treatment, abrogating orthotopic tumor growth and significantly repressing spontaneous lung metastatic dissemination in treated mice. These findings provide a strong preclinical rationale for developing drug regimens combining heparanase inhibitors/HS mimetics with IGF1R antagonists for treatment of metastatic SS.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Heparin, Low-Molecular-Weight/pharmacology , Pyrazoles/pharmacology , Receptors, Somatomedin/antagonists & inhibitors , Sarcoma, Synovial/drug therapy , Triazines/pharmacology , Xenograft Model Antitumor Assays , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Glucuronidase/antagonists & inhibitors , Glucuronidase/metabolism , Heparin, Low-Molecular-Weight/administration & dosage , Heparin, Low-Molecular-Weight/metabolism , Humans , Mice, SCID , Neoplasm Metastasis , Pyrazoles/administration & dosage , Receptor, IGF Type 1 , Receptors, Somatomedin/metabolism , Sarcoma, Synovial/metabolism , Sarcoma, Synovial/pathology , Sulfates , Triazines/administration & dosage
5.
Cancer Lett ; 395: 53-62, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28274892

ABSTRACT

Radiotherapy is one of the main treatment choices for non-metastatic prostate cancer (PCa), although development of radioresistance limits its effectiveness. Mounting evidence supports the ability of microRNAs to interfere with different radioresistance-associated pathways, suggesting their potential as radiosensitizers. Here, we demonstrate that reconstitution of miR-875-5p, whose expression is down-regulated in PCa clinical samples and directly correlates with that of E-cadherin, was able to enhance radiation response in PCa cell lines and xenografts through EGFR direct targeting. Consistent with the established role of EGFR in sustaining epithelial-to-mesenchymal transition (EMT) and promoting DNA repair following radiation-induced nuclear translocation, we found that miR-875-5p reconstitution in PCa cells counteracted EMT and impaired DNA lesion clearance. Down-regulation of the EMT-inducing transcription factor ZEB1, which also plays a role in homologous recombination-mediated repair of DNA lesions by regulating CHK1 expression, was found to be a major determinant of miR-875-5p-induced radiosensitization, as confirmed by phenocopy experiments showing that siRNA-mediated ZEB1 knock-down was able to reproduce the microRNA radiosensitizing effect. Overall, our data support the clinical interest in developing a novel therapeutic approach based on miR-875-5p reconstitution to increase PCa response to radiotherapy.


Subject(s)
Epithelial-Mesenchymal Transition , ErbB Receptors/physiology , MicroRNAs/physiology , Prostatic Neoplasms/radiotherapy , Zinc Finger E-box-Binding Homeobox 1/physiology , Animals , Cell Line, Tumor , DNA Repair , ErbB Receptors/genetics , Humans , Male , Mice , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Radiation Tolerance
6.
Eur J Med Chem ; 124: 840-851, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27643641

ABSTRACT

A series of [1,2]Oxazolo [5,4-e]isoindoles has been synthesized through a versatile and high yielding sequence. All the new structures showed in the 1HNMR spectra, the typical signal in the 8.34-8.47 ppm attributable to the H-3 of the [1,2]oxazole moiety. Among all derivatives, methoxy benzyl substituents at positions 3 and 4 or/and 5 were very effective in reducing the growth of different tumor cell lines, including diffuse malignant peritoneal mesothelioma (DMPM), an uncommon and rapidly malignancy poorly responsive to available therapeutic options. The most active compound 6j was found to impair tubulin polymerization, cause cell cycle arrest at G2/M phase and induce apoptosis in DMPM cells, making it as a new lead for the discovery of new potent antimitotic drugs.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Isoindoles/chemistry , Isoindoles/pharmacology , Protein Multimerization/drug effects , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Isoindoles/chemical synthesis , Isomerism , M Phase Cell Cycle Checkpoints/drug effects , Protein Structure, Quaternary , Tubulin/chemistry , Tubulin Modulators/chemical synthesis
7.
J Med Chem ; 59(15): 7223-38, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27428868

ABSTRACT

A series of 22 derivatives of the [1,2]oxazolo[5,4-e]isoindole system were synthesized through an efficient and versatile procedure that involves the annelation of the [1,2]oxazole moiety to the isoindole ring, producing derivatives with a wide substitution pattern. The structure-activity relationship indicates that the N-4-methoxybenzyl group appears crucial for potent activity. In addition, the presence of a 6-phenyl moiety is important and the best activity is reached with a 3,4,5-trimethoxy substituent. The most active compound, bearing both the structural features, was able to inhibit tumor cell proliferation at nanomolar concentrations when tested against the full NCI human tumor cell line panel. Interestingly, this compound was effective in reducing in vitro and in vivo cell growth, impairing cell cycle progression and inducing apoptosis, as a consequence of the inhibition of tubulin polymerization, in experimental models of diffuse malignant peritoneal mesothelioma (DMPM), a rapidly lethal disease, poorly responsive to conventional therapeutic strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Isoindoles/pharmacology , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Peritoneal Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Isoindoles/chemical synthesis , Isoindoles/chemistry , Lung Neoplasms/pathology , Mesothelioma/pathology , Mesothelioma, Malignant , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Peritoneal Neoplasms/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
8.
Bioorg Med Chem ; 23(21): 6785-93, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26481659

ABSTRACT

Among the natural histone deacetylase inhibitors (HDACi), the bicyclic depsipeptide macrolactone FK228 stands out for its unique chemical structure and mechanism of action. In order to expand the chemical diversity, exploiting the FK228 peculiar structure, we have synthesized a collection of 24 simplified novel analogs. A first series consists of bicyclic macrolactones, where the carboxy terminus of the natural compound was substituted by peptidomimetic aminomethylphenylacetic acid derivatives. These analogs, 7a-i, showed submicromolar cytotoxic activity, even though very low inhibitory activity against HDAC enzymes, suggesting that most probably they behave with a mechanism different from the natural compound. One of the most active members in the group, 7g, was evaluated in vivo and exhibited significant antitumor activity. This evidence supports that the activity is unrelated to HDAC inhibition and these compounds represent a novel series of promising active agents. Another analog series consists of monocyclic macrolactones, 9a-c and 10a-d which lack the disulfide bridge and bear the protected sulfur on the linear external chain; they showed similar cytotoxic activities compared to the natural compound, but proved to be very sensitive to the nature of the sulfur protection. In fact, when the sulfur was protected by an 1-octanoyl residue, like in 9b, the product displayed a one digit nanomolar activity. The results provide evidence that our approach may be followed to develop novel series of FK228 analogs.


Subject(s)
Depsipeptides/chemistry , Drug Design , Histone Deacetylase Inhibitors/chemical synthesis , Cell Survival/drug effects , Depsipeptides/chemical synthesis , Depsipeptides/toxicity , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/toxicity , Humans , Lactones/chemical synthesis , Lactones/chemistry , Lactones/toxicity , Microwaves , Solid-Phase Synthesis Techniques
9.
Oncotarget ; 6(15): 13119-32, 2015 May 30.
Article in English | MEDLINE | ID: mdl-25948791

ABSTRACT

Survivin, which is highly expressed and promotes cell survival in diffuse malignant peritoneal mesothelioma (DMPM), exclusively relies on exportin 1 (XPO1/CRM1) to be shuttled into the cytoplasm and perform its anti-apoptotic function. Here, we explored the efficacy of Selective Inhibitors of Nuclear Export (SINE), KPT-251, KPT-276 and the orally available, clinical stage KPT-330 (selinexor), in DMPM preclinical models. Exposure to SINE induced dose-dependent inhibition of cell growth, cell cycle arrest at G1-phase and caspase-dependent apoptosis, which were consequent to a decrease of XPO1/CRM1 protein levels and the concomitant nuclear accumulation of its cargo proteins p53 and CDKN1a. Cell exposure to SINE led to a time-dependent reduction of cytoplasmic survivin levels. In addition, after an initial accumulation, the nuclear protein abundance progressively decreased, as a consequence of an enhanced ubiquitination and proteasome-dependent degradation. SINE and the survivin inhibitor YM155 synergistically cooperated in reducing DMPM cell proliferation. Most importantly, orally administered SINE caused a significant anti-tumor effect in subcutaneous and orthotopic DMPM xenografts without appreciable toxicity. Overall, we have demonstrated a marked efficacy of SINE in DMPM preclinical models that may relay on the interference with survivin intracellular distribution and function. Our study suggests SINE-mediated XPO1/CRM1 inhibition as a novel therapeutic option for DMPM.


Subject(s)
Antineoplastic Agents/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , Karyopherins/antagonists & inhibitors , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Peritoneal Neoplasms/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Acrylamides/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Hydrazines/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mesothelioma/metabolism , Mesothelioma/pathology , Mesothelioma, Malignant , Mice , Mice, SCID , Neoplasm Proteins/metabolism , Oxadiazoles/pharmacology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Real-Time Polymerase Chain Reaction , Survivin , Thiazoles/pharmacology , Triazoles/pharmacology , Tumor Suppressor Protein p53/metabolism , Exportin 1 Protein
10.
Biochem Pharmacol ; 94(2): 79-90, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25600908

ABSTRACT

Non-Small Cell Lung Cancer (NSCLC) remains an aggressive and fatal disease with low responsiveness to chemotherapy, frequent drug resistance development and metastatic behavior. Platinum-based therapy is the standard of care for NSCLC with limited benefits. Since epigenetic alterations have been implicated in the aggressive behavior of lung cancer, the purpose of the present study was to examine the capability of the pan-histone deacetylase inhibitor SAHA and of ST3595, a novel hydroxamate-based compound, to interfere with the proliferative and invasive potential of NSCLC cells. We used two NSCLC cell lines (H460 and A549) and the cisplatin-resistant variants (H460/Pt and A549/Pt), to mimic a frequent clinical condition. The resistant models exhibited increased invasive properties as compared to parental cells, features associated with a wide modulation of the level of angiogenesis- and invasion-related factors in the cell conditioned media. The levels of urokinase-type plasminogen activator, IL-8, and macrophage migration inhibitory factor were increased in the conditioned media from both H460/Pt and A549/Pt cells. SAHA and ST3595 induced a strong inhibition of cell invasive properties, which was more marked after ST3595 exposure. Both HDAC inhibitors up-regulated the metastasis suppressor KiSS1 at the mRNA level. Forced expression of KiSS1 significantly decreased the invasive capability of drug-resistant cells. ST3595 displayed an anti-metastatic effect in tumors associated with decreased of phosphorylation of Src. Our data indicate that HDAC inhibitors are effective in NSCLC cell systems. The ability of ST3595 to counteract the invasive potential of resistant cells through mechanisms involving KiSS1 is an interesting novel finding.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Lung Neoplasms/pathology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Enzyme-Linked Immunosorbent Assay , Female , Humans , Kisspeptins/drug effects , Mice , Mice, Nude , Phenotype
11.
Oncotarget ; 5(12): 4516-28, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24980831

ABSTRACT

Target-specific agents used in melanoma are not curative, and chemokines are being implicated in drug-resistance to target-specific agents. Thus, the use of conventional agents in rationale combinations may result in optimization of therapy. Because histone deacetylases participate in tumor development and progression, the combination of the pan-inhibitor SAHA and temozolomide might provide a therapeutic advantage. Here, we show synergism between the two drugs in mutant BRAF cell lines, in association with decreased phosphorylation of cell survival proteins (e.g., C-Jun-N-terminal-kinase, JNK). In the spontaneous ret transgenic mouse melanoma model, combination therapy produced a significant disease onset delay and down-regulation of Chemokine (C-C motif) ligand 2 (CCL2), JNK, and of Myeloid-derived suppressor cell recruitment. Co-incubation with a CCL2-blocking-antibody enhanced in vitro cell sensitivity to temozolomide. Conversely, recombinant CCL2 activated JNK in human tumor melanoma cells. In keeping with these results, the combination of a JNK-inhibitor with temozolomide was synergistic. By showing that down-regulation of CCL2-driven signals by SAHA and temozolomide via JNK contributes to reduce melanoma growth, we provide a rationale for the therapeutic advantage of the drug combination. This combination strategy may be effective because of interference both with tumor cell and tumor microenvironment.


Subject(s)
Chemokines/metabolism , Dacarbazine/analogs & derivatives , Histone Deacetylase Inhibitors/metabolism , Melanoma/genetics , Animals , Apoptosis , Cell Survival , Dacarbazine/metabolism , Disease Models, Animal , Drug Synergism , Humans , Mice , Mice, Transgenic , Temozolomide , Tumor Microenvironment
12.
Mol Pharm ; 11(1): 283-93, 2014 Jan 06.
Article in English | MEDLINE | ID: mdl-24256025

ABSTRACT

Treatment of ovarian carcinoma often fails to be curative because of drug resistance, and many efforts are directed to overcome tumor cell resistance by increasing apoptosis induction. The potential of second mitochondria-derived activator of caspases (SMAC) mimetics (SMACm) has appeared in preclinical studies, but novel proapoptotic agents of this class with improved pharmacological profile are needed. To identify novel treatment options for ovarian carcinoma by interfering with antiapoptotic factors, in the present study a novel homodimeric SMACm (SM83) was employed in preclinical models both in vitro and in vivo. An investigation of the structural features of dimeric SM83 as compared to a closely related reference compound indicated slight differences, likely because of the interaction between one of the terminal phenyl groups and triazole rings of SM83 with the BIR2 domain. Although SM83 per se did not inhibit cell proliferation, it displayed a synergistic effect in combination with TNF-related apoptosis inducing ligand (TRAIL) in cell sensitivity assays. Because the tumor microenvironment is a reservoir of cytokines that may act in conjunction with SMACm to affect tumor growth, the activity of the novel compound was tested in vivo in ovarian carcinoma cells subcutaneously xenografted into immunodeficient mice. A significant tumor volume inhibition was observed together with activation of caspase 3 and apoptotic cell death. A biochemical analysis of tumor necrosis factor (TNF) and TRAIL content in specimens from xenografted mice indicated that SM83 downmodulated the levels of human TNF in plasma samples and tended to upmodulate human TRAIL levels in tumors. Thus, TRAIL appears to contribute to the antitumor activity of novel SMACm SM83 in subcutaneously grown ovarian carcinoma. Overall, our results indicate that SM83 is an attractive candidate for further development.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Apoptosis/drug effects , Biomimetic Materials/pharmacology , Carrier Proteins/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Mitochondrial Proteins/pharmacology , Ovarian Neoplasms/drug therapy , Animals , Blotting, Western , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Humans , Magnetic Resonance Spectroscopy , Mice , Molecular Dynamics Simulation , Ovarian Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
13.
J Clin Endocrinol Metab ; 99(2): 498-509, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24276455

ABSTRACT

CONTEXT: Tyrosine kinase inhibitors represent a new treatment option for patients with advanced medullary thyroid cancer (MTC). However, cures have not been achieved with current available agents used in monotherapy. OBJECTIVE: Because RET has been shown to negatively regulate CD95 death receptor activation in preclinical models of RET-dependent MTC, we investigated the potential of the combination approach with the RET-targeting tyrosine kinase inhibitor sunitinib and cisplatin to enhance apoptosis activation through the extrinsic pathway. DESIGN: The effects of sunitinib and cisplatin were examined in human MTC cell lines harboring oncogenic RET mutations. Experiments were designed to determine drug effects on RET signaling, cell growth, apoptosis, autophagy, and tumor growth in mice and to investigate the mechanisms of the drug interaction. RESULTS: Sunitinib and cisplatin synergistically inhibited the growth of MZ-CRC-1 cells harboring the RET M918T activating mutation. The combination enhanced apoptosis activation through CD95-mediated, caspase-8-dependent pathway. Moreover, sunitinib induced a severe perturbation of the autophagic flux characterized by autophagosome accumulation and a remarkable lysosomal dysfunction, which was further enhanced, with lysosomal leakage induction, by cisplatin. Administration of the drug combination to mice xenografted with MZ-CRC-1 cells improved the antitumor efficacy, as compared with single-agent treatments, inducing complete responses in 30% of the treated mice, a significant increase in caspase-3 activation (P < .01 vs cisplatin, and P < .0005 vs sunitinib) and apoptosis in tumor cells. CONCLUSIONS: Addition of cisplatin to sunitinib potentiates apoptotic cell death and has promising preclinical activity in MTCs harboring the RET M918T oncogene.


Subject(s)
Apoptosis/drug effects , Carcinoma, Medullary/drug therapy , Cisplatin/pharmacology , Indoles/pharmacology , Pyrroles/pharmacology , Thyroid Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Medullary/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/therapeutic use , Drug Synergism , Humans , Indoles/therapeutic use , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrroles/therapeutic use , Sunitinib , Thyroid Neoplasms/pathology
14.
Clin Cancer Res ; 20(4): 995-1006, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24327272

ABSTRACT

PURPOSE: Despite the frequent overexpression of epidermal growth factor receptor (EGFR) in squamous cell carcinoma (SCC), the efficacy of cetuximab alone is limited. Given the marked activity of namitecan, a hydrophilic camptothecin, against SCC models, the present study was performed to explore the efficacy of the cetuximab-namitecan combination in a panel of SCC models. EXPERIMENTAL DESIGN: We examined the antiproliferative and antitumor activities of the cetuximab-namitecan combination in four SCC models characterized by a different EGFR gene copy number/EGFR protein level. We also assessed the effects of the combination on EGFR expression at both mRNA and protein levels and investigated the molecular basis of the interaction between the two agents. RESULTS: Cetuximab and namitecan exhibited synergistic effects, resulting in potentiation of cell growth inhibition and, most importantly, enhanced therapeutic efficacy, with high cure rates in three SCC models characterized by high EGFR gene copy number, without increasing toxicity. The synergistic antitumor effect was also observed with the cetuximab-irinotecan combination. At the molecular level, the two agents produced a cooperative effect resulting in complete downregulation of EGFR. Interestingly, when singly administered, the camptothecin was able to strongly decrease EGFR expression mainly by transcriptional inhibition. CONCLUSIONS: Our results (i) demonstrate a marked efficacy of the cetuximab-namitecan combination, which reflects a complete abrogation of EGFR expression as a critical determinant of the therapeutic improvement, in SCC preclinical models, and (ii) suggest EGFR gene copy number as a possible marker to be used for patient selection in the clinical setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/genetics , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Line, Tumor , Cetuximab , DNA Copy Number Variations , Drug Synergism , ErbB Receptors/metabolism , Gene Dosage , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Xenograft Model Antitumor Assays
15.
Int J Cancer ; 134(12): 2789-97, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24242003

ABSTRACT

Altered degradation and deposition of extracellular matrix are hallmarks of tumor progression and response to therapy. From a microarray supervised analysis on a dataset of chemotherapy-treated breast carcinoma patients, maspin, a member of the serpin protease inhibitor family, has been the foremost variable identified in non-responsive versus responsive tumors. Accordingly, in a series of 52 human breast carcinomas, we detected high maspin expression in tumors that progressed under doxorubicin (DXR)-based chemotherapy. Our analysis of the role of maspin in response to chemotherapy in human MCF7 and MDAMB231 breast and SKOV3 ovarian carcinoma cells transfected to overexpress maspin and injected into mice showed that maspin overexpression led to DXR resistance through the maspin-induced collagen-enriched microenvironment and that an anti-maspin neutralizing monoclonal antibody reversed the collagen-dependent DXR resistance. Impaired diffusion and decreased DXR activity were also found in tumors derived from Matrigel-embedded cells, where abundant collagen fibers characterize the tumor matrix. Conversely, liposome-based DXR reached maspin-overexpressing tumor cells despite the abundant extracellular matrix and was more efficient in reducing tumor growth. Our results identify maspin-induced accumulation of collagen fibers as a cause of disease progression under DXR chemotherapy for breast cancer. Use of a more hydrophilic DXR formulation or of a maspin inhibitor in combination with chemotherapy holds the promise of more consistent responses to maspin-overexpressing tumors and dense-matrix tumors in general.


Subject(s)
Breast Neoplasms/metabolism , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Ovarian Neoplasms/metabolism , Serpins/metabolism , Tumor Microenvironment/drug effects , Animals , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/immunology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen/metabolism , Disease Progression , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Serpins/biosynthesis , Serpins/immunology
16.
Biochem Pharmacol ; 84(2): 163-71, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22525722

ABSTRACT

Namitecan (ST1968), a novel hydrophilic camptothecin analog of the 7-oxyiminomethyl series, was selected for clinical development on the basis of its promising preclinical efficacy. Since there is clinical evidence of efficacy of camptothecins against pediatric tumors, this study was performed to explore the antitumor and antiangiogenic activity of the camptothecin derivative in pediatric sarcoma models. With the exception of an undifferentiated rhabdomyosarcoma (A204), namitecan exhibited curative efficacy even at well-tolerated suboptimal doses in a panel of five models. The good therapeutic index of namitecan likely reflected a high and persistent drug accumulation at tumor site. The four responsive tumors were characterized by high topoisomerase I expression. In the RD/TE-671 rhabdomyosarcoma model the drug activity was associated with a marked antiangiogenic effect, which was consistent with the downregulation of proangiogenic factors, including VEGF, bFGF and the multifunctional chemokines CCL-2 and CXCL16. In agreement with this modulation, the combination of low doses of namitecan with other antiangiogenic agents, such as bevacizumab (a humanized anti-VEGF antibody) and sunitinib (a multitarget tyrosine kinase inhibitor effective against receptors implicated in the angiogenesis process), enhanced the antitumor effects. In conclusion, this preclinical study provides evidence of curative efficacy of namitecan at well-tolerated doses against pediatric sarcoma models, likely reflecting a contribution of antiangiogenic effects. Based on the promising therapeutic profile, namitecan is a good candidate for clinical evaluation in pediatric sarcomas.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Sarcoma/drug therapy , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Bevacizumab , Camptothecin/pharmacology , Chemokine CCL2/metabolism , Chemokine CXCL16 , Chemokines, CXC/metabolism , DNA Topoisomerases, Type I/metabolism , Female , Fibroblast Growth Factor 2/metabolism , Humans , Indoles/pharmacology , Mice , Mice, Nude , Pyrroles/pharmacology , Receptors, Scavenger/metabolism , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Sarcoma/metabolism , Sarcoma/pathology , Sunitinib , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...