Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biology (Basel) ; 10(4)2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33917111

ABSTRACT

The anti-Müllerian hormone (AMH) belongs to the TGF-ß family and plays a key role during fetal sexual development. Various reports have described the expression of AMH type II receptor (AMHRII) in human gynecological cancers including ovarian tumors. According to qRT-PCR results confirmed by specific In-Situ Hybridization (ISH) experiments, AMHRII mRNA is expressed in an extremely restricted number of normal tissues. By performing ISH on tissue microarray of solid tumor samples AMHRII mRNA was unexpectedly detected in several non-gynecological primary cancers including lung, breast, head and neck, and colorectal cancers. AMHRII protein expression, evaluated by immunohistochemistry (IHC) was detected in approximately 70% of epithelial ovarian cancers. Using the same IHC protocol on more than 900 frozen samples covering 18 different cancer types we detected AMHRII expression in more than 50% of hepato-carcinomas, colorectal, lung, and renal cancer samples. AMHRII expression was not observed in neuroendocrine lung tumor samples nor in non-Hodgkin lymphoma samples. Complementary analyses by immunofluorescence and flow cytometry confirmed the detection of AMHRII on a panel of ovarian and colorectal cancers displaying comparable expression levels with mean values of 39,000 and 50,000 AMHRII receptors per cell, respectively. Overall, our results suggest that this embryonic receptor could be a suitable target for treating AMHRII-expressing tumors with an anti-AMHRII selective agent such as murlentamab, also named 3C23K or GM102. This potential therapeutic intervention was confirmed in vivo by showing antitumor activity of murlentamab against AMHRII-expressing colorectal cancer and hepatocarcinoma Patient-Derived tumor Xenografts (PDX) models.

2.
Cancers (Basel) ; 13(8)2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33924378

ABSTRACT

AMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs but is also re-expressed in gynecologic cancers. Hence, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody currently in clinical trial. Low-fucosylated antibodies are known to increase the antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) potency of effector cells, but some preliminary results suggest a more global murlentamab-dependent activation of the immune system. In this context, we demonstrate here that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells, in the presence of unstimulated- or tumor-associated macrophage (TAM)-like macrophages, significantly promotes macrophage-mediated ADCC and shifts the whole microenvironment towards a pro-inflammatory and anti-tumoral status, thus triggering anti-tumor activity. We also report that murlentamab orients both unstimulated- and TAM-like macrophages to an M1-like phenotype characterized by a strong expression of co-stimulation markers, pro-inflammatory cytokines and chemokines, favoring T cell recruitment and activation. Moreover, we show that murlentamab treatment shifts CD4+ Th1/Th2 balance towards a Th1 response and activates CD8+ T cells. Altogether, these results suggest that murlentamab, through naïve macrophage orientation and TAM reprogrammation, stimulates the anti-tumor adaptive immune response. Those mechanisms might contribute to the sustained clinical benefit observed in advanced cancer patients treated with murlentamab. Finally, the enhanced murlentamab activity in combination with pembrolizumab opens new therapeutic perspectives.

3.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32503947

ABSTRACT

BACKGROUND: Besides the interest of an early detection of ovarian cancer, there is an urgent need for new predictive and prognostic biomarkers of tumor development and cancer treatment. In healthy patients, circulating blood monocytes are typically subdivided into classical (85%), intermediate (5%) and non-classical (10%) populations. Although these circulating monocyte subsets have been suggested as biomarkers in several diseases, few studies have investigate their potential as a predictive signature for tumor immune status,tumor growth and treatment adaptation. METHODS: In this study, we used a homogeneous cohort of 28 chemotherapy-naïve patients with ovarian cancer to evaluate monocyte subsets as biomarkers of the ascites immunological status. We evaluated the correlations between circulating monocyte subsets and immune cells and tumor burden in peritoneal ascites. Moreover, to validate the use of circulating monocyte subsets tofollow tumor progression and treatment response, we characterized blood monocytes from ovarian cancer patients included in a phase 1 clinical trial at baseline and following murlentamab treatment. RESULTS: We demonstrate here a robust expansion of the intermediate blood monocytes (IBMs) in ovarian cancer patients. We establish a significant positive correlation between IBM percentage and tumor-associate macrophages with a CCR2high/CD163high/CD206high/CD86lowprofile. Moreover, IBM expansion is associated with a decreased effector/regulatory T-cell ratio in ascites and with the presence of soluble immunosuppressive mediators. We also establish that IBM proportion positively correlates with the peritoneum tumor burden. Finally, the study of IBMs in patients with ovarian cancer under immunotherapy during the phase clinical trial supports IBMs to follow the evolution of tumor development and the treatment adaptation. CONCLUSIONS: This study, which links IBM level with immunosuppression and tumor burden in peritoneum, identifies IBMs as apotential predictive signature of ascites immune status and as a biomarker ofovarian cancer development and treatment response. TRIAL REGISTRATION NUMBER: EudraCT: 2015-004252-22 NCT02978755.


Subject(s)
Ascites/genetics , Biomarkers, Tumor/metabolism , Immunotherapy/methods , Lipopolysaccharide Receptors/metabolism , Monocytes/metabolism , Receptors, IgG/metabolism , Disease Progression , Female , Humans , Male , Tumor Microenvironment
4.
Cell Commun Signal ; 17(1): 106, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31443721

ABSTRACT

BACKGROUND: HER3/ErbB3 receptor deletion or blockade leads to tumor cell apoptosis, whereas its overexpression confers anti-cancer drug resistance through upregulation of protective mechanisms against apoptosis. We produced the anti-HER3 antibody 9F7-F11 that promotes HER3 ubiquitination and degradation via JNK1/2-dependent activation of the E3 ubiquitin ligase ITCH, and that induces apoptosis of cancer cells. Cellular FLICE-like inhibitory protein (c-FLIP) is a key regulator of apoptotic pathways. Here, we wanted to determine the mechanisms underlying the pro-apoptotic effect of 9F7-F11. METHODS: Anti-HER3 antibody-induced apoptosis was assessed by western blot, and by flow cytometry measurement of Annexin V/7-AAD-labelled tumor cells (BxPC3, MDA-MB-468 and DU145 cell lines). c-FLIP/ITCH interaction and subsequent degradation/ubiquitination were investigated by co-immunoprecipitation of ITCH-silenced vs scramble control cells. The relationship between ITCH-mediated c-FLIP degradation and antibody-induced apoptosis was examined by western blot and flow cytometry of tumor cells, after ITCH RNA interference or by pre-treatment with ITCH chemical inhibitor chlorimipramine (CI). RESULTS: Following incubation with 9F7-F11, cancer cell apoptosis occurs through activation of caspase-8, - 9 and - 3 and the subsequent cleavage of poly (ADP-ribose) polymerase (PARP). Moreover we showed that ubiquitination and proteasomal degradation of the anti-apoptotic protein c-FLIP was mediated by USP8-regulated ITCH recruitment. This effect was abrogated by ITCH- and USP8-specific RNA interference (siRNA), or by the ITCH chemical inhibitor CI. Specifically, ITCH silencing or CI blocked 9F7-F11-induced caspase-8-mediated apoptosis of tumor cells, and restored c-FLIP expression. ITCH-silencing or CI concomitantly abrogated HER3-specific antibody-induced apoptosis of Annexin V/7-AAD-labelled BxPC3 cells. 9F7-F11 favored the extrinsic apoptosis pathway by inducing TRAIL-R2/DR5 upregulation and TRAIL expression that promoted the formation of death-inducing signaling complex (DISC), leading to caspase-8-mediated apoptosis. Incubation with 9F7-F11 also induced BID cleavage, BAX upregulation and BIM expression, which initiated the caspase-9/3-mediated mitochondrial death pathway. The anti-HER3 antibody pro-apoptotic effect occurred concomitantly with downregulation of the pro-survival proteins c-IAP2 and XIAP. CONCLUSIONS: The allosteric non-neuregulin competing modulator 9F7-F11, sensitizes tumor cells to DR5/caspase-8-mediated apoptosis through ITCH-dependent downregulation of c-FLIP.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/metabolism , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , Humans , Signal Transduction
5.
Oncotarget ; 8(59): 99950-99965, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29245952

ABSTRACT

Müllerian inhibiting substance, also called anti-Müllerian hormone (AMH), inhibits proliferation and induces apoptosis of AMH type II receptor-positive tumor cells, such as human ovarian cancers (OCs). On this basis, a humanized glyco-engineered monoclonal antibody (3C23K) has been developed. The aim of this study was therefore to experimentally confirm the therapeutic potential of 3C23K in human OCs. We first determined by immunofluorescence, immunohistochemistry and cytofluorometry analyses the expression of AMHRII in patient's tumors and found that a majority (60 to 80% depending on the detection technique) of OCs were positive for this marker. We then provided evidence that the tumor stroma of OC is enriched in tumor-associated macrophages and that these cells are responsible for 3C23K-induced killing of tumor cells through ADCP and ADCC mechanisms. In addition, we showed that 3C23K reduced macrophages induced-T cells immunosuppression. Finally, we evaluated the therapeutic efficacy of 3C23K alone and in combination with a carboplatin-paclitaxel chemotherapy in a panel of OC Patient-Derived Xenografts. In those experiments, we showed that 3C23K significantly increased the proportion and the quality of chemotherapy-based in vivo responses. Altogether, our data support the potential interest of AMHRII targeting in human ovarian cancers and the evaluation of 3C23K in further clinical trials.

6.
Mol Cancer Ther ; 16(7): 1312-1323, 2017 07.
Article in English | MEDLINE | ID: mdl-28507002

ABSTRACT

Exploratory clinical trials using therapeutic anti-HER3 antibodies strongly suggest that neuregulin (NRG1; HER3 ligand) expression at tumor sites is a predictive biomarker of anti-HER3 antibody efficacy in cancer. We hypothesized that in NRG1-expressing tumors, where the ligand is present before antibody treatment, anti-HER3 antibodies that do not compete with NRG1 for receptor binding have a higher receptor-neutralizing action than antibodies competing with the ligand for binding to HER3. Using time-resolved-fluorescence energy transfer (TR-FRET), we demonstrated that in the presence of recombinant NRG1, binding of 9F7-F11 (a nonligand-competing anti-HER3 antibody) to HER3 is increased, whereas that of ligand-competing anti-HER3 antibodies (H4B-121, U3-1287, Ab#6, Mab205.10.2, and MOR09825) is decreased. Moreover, 9F7-F11 showed higher efficacy than antibodies that compete with the ligand for binding to HER3. Specifically, 9F7-F11 inhibition of cell proliferation and of HER3/AKT/ERK1/2 phosphorylation as well as 9F7-F11-dependent cell-mediated cytotoxicity were higher in cancer cells preincubated with recombinant NRG1 compared with cells directly exposed to the anti-HER3 antibody. This translated in vivo into enhanced growth inhibition of NRG1-expressing BxPC3 pancreatic, A549 lung, and HCC-1806 breast cell tumor xenografts in mice treated with 9F7-F11 compared with H4B-121. Conversely, both antibodies had similar antitumor effect in NRG1-negative HPAC pancreatic carcinoma cells. In conclusion, the allosteric modulator 9F7-F11 shows increased anticancer effectiveness in the presence of NRG1 and thus represents a novel treatment strategy for NRG1-addicted tumors. Mol Cancer Ther; 16(7); 1312-23. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/administration & dosage , Biomarkers, Tumor/immunology , Neoplasms/drug therapy , Neuregulin-1/genetics , Receptor, ErbB-3/immunology , A549 Cells , Animals , Antibodies, Anti-Idiotypic/administration & dosage , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Biomarkers, Tumor/genetics , Cell Proliferation/drug effects , Female , Fluorescence Resonance Energy Transfer , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neuregulin-1/immunology , Phosphorylation , Protein Binding , Receptor, ErbB-3/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Oncotarget ; 8(23): 37061-37079, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28427157

ABSTRACT

Ovarian cancer is the leading cause of death in women with gynecological cancers and despite recent advances, new and more efficient therapies are crucially needed. Müllerian Inhibiting Substance type II Receptor (MISRII, also named AMHRII) is expressed in most ovarian cancer subtypes and is a novel potential target for ovarian cancer immunotherapy. We previously developed and tested 12G4, the first murine monoclonal antibody (MAb) against human MISRII. Here, we report the humanization, affinity maturation and glyco-engineering steps of 12G4 to generate the Fc-optimized 3C23K MAb, and the evaluation of its in vivo anti-tumor activity. The epitopes of 3C23K and 12G4 were strictly identical and 3C23K affinity for MISRII was enhanced by a factor of about 14 (KD = 5.5 × 10-11 M vs 7.9 × 10-10 M), while the use of the EMABling® platform allowed the production of a low-fucosylated 3C23K antibody with a 30-fold KD improvement of its affinity to FcγRIIIa. In COV434-MISRII tumor-bearing mice, 3C23K reduced tumor growth more efficiently than 12G4 and its combination with carboplatin was more efficient than each monotherapy with a mean tumor size of 500, 1100 and 100 mm3 at the end of treatment with 3C23K (10 mg/kg, Q3-4D12), carboplatin (60 mg/kg, Q7D4) and 3C23K+carboplatin, respectively. Conversely, 3C23K-FcKO, a 3C23K form without affinity for the FcγRIIIa receptor, did not display any anti-tumor effect in vivo. These results strongly suggested that 3C23K mechanisms of action are mainly Fc-related. In vitro, antibody-dependent cytotoxicity (ADCC) and antibody-dependent cell phagocytosis (ADCP) were induced by 3C23K, as demonstrated with human effector cells. Using human NK cells, 50% of the maximal lysis was obtained with a 46-fold lower concentration of low-fucosylated 3C23K (2.9 ng/ml) than of 3C23K expressed in CHO cells (133.35 ng/ml). As 3C23K induced strong ADCC with human PBMC but almost none with murine PBMC, antibody-dependent cell phagocytosis (ADCP) was then investigated. 3C23K-dependent (100 ng/ml) ADCP was more active with murine than human macrophages (only 10% of living target cells vs. about 25%). These in vitro results suggest that the reduced ADCC with murine effectors could be partially balanced by ADCP activity in in vivo experiments. Taken together, these preclinical data indicate that 3C23K is a new promising therapeutic candidate for ovarian cancer immunotherapy and justify its recent introduction in a phase I clinical trial.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Ovarian Neoplasms/drug therapy , Receptors, Peptide/immunology , Receptors, Transforming Growth Factor beta/immunology , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/immunology , Apoptosis/drug effects , Apoptosis/immunology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Female , Glycosylation , Humans , Mice, Nude , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Protein Engineering
8.
PLoS One ; 6(8): e23597, 2011.
Article in English | MEDLINE | ID: mdl-21853156

ABSTRACT

F14512 is a novel anti-tumor molecule based on an epipodophyllotoxin core coupled to a cancer-cell vectoring spermine moiety. This polyamine linkage is assumed to ensure the preferential uptake of F14512 by cancer cells, strong interaction with DNA and potent inhibition of topoisomerase II (Topo II). The antitumor activity of F14512 in human tumor models is significantly higher than that of other epipodophyllotoxins in spite of a lower induction of DNA breakage. Hence, the demonstrated superiority of F14512 over other Topo II poisons might not result solely from its preferential uptake by cancer cells, but could also be due to unique effects on Topo II interactions with DNA. To further dissect the mechanism of action of F14512, we used Drosophila melanogaster mutants whose genetic background leads to an easily scored phenotype that is sensitive to changes in Topo II activity and/or localization. F14512 has antiproliferative properties in Drosophila cells and stabilizes ternary Topo II/DNA cleavable complexes at unique sites located in moderately repeated sequences, suggesting that the drug specifically targets a select and limited subset of genomic sequences. Feeding F14512 to developing mutant Drosophila larvae led to the recovery of flies expressing a striking phenotype, "Eye wide shut," where one eye is replaced by a first thoracic segment. Other recovered F14512-induced gain- and loss-of-function phenotypes similarly correspond to precise genetic dysfunctions. These complex in vivo results obtained in a whole developing organism can be reconciled with known genetic anomalies and constitute a remarkable instance of specific alterations of gene expression by ingestion of a drug. "Drosophila-based anticancer pharmacology" hence reveals unique properties for F14512, demonstrating the usefulness of an assay system that provides a low-cost, rapid and effective complement to mammalian models and permits the elucidation of fundamental mechanisms of action of candidate drugs of therapeutic interest in humans.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , Drosophila melanogaster/drug effects , Podophyllotoxin/analogs & derivatives , Polyamines/chemistry , Topoisomerase II Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Shape/drug effects , Chromosomal Position Effects/drug effects , DNA/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation/drug effects , Genes, Insect/genetics , Histones/genetics , Humans , Models, Animal , Phenotype , Podophyllotoxin/chemistry , Podophyllotoxin/pharmacology , Repetitive Sequences, Nucleic Acid/genetics , Suppression, Genetic/drug effects , Topoisomerase II Inhibitors/chemistry , X Chromosome/genetics
9.
Invest New Drugs ; 29(1): 9-21, 2011 Feb.
Article in English | MEDLINE | ID: mdl-19777159

ABSTRACT

We have exploited the polyamine transport system (PTS) to deliver selectively a spermine-drug conjugate, F14512 to cancer cells. This study was aimed to define F14512 anticancer efficacy against tumor models and to investigate whether fluorophor-labeled polyamine probes could be used to identify tumors expressing a highly active PTS and that might be sensitive to F14512 treatments. Eighteen tumor models were used to assess F14512 antitumor activity. Cellular uptake of spermine-based fluorescent probes was measured by flow cytometry in cells sampled from tumor xenografts by needle biopsy. The accumulation of the fluorescent probe within B16 tumors in vivo was assessed using infrared fluorescence imaging. This study has provided evidence of a major antitumor activity for F14512. Significant responses were obtained in 67% of the tumor models evaluated, with a high level of activity recorded in 33% of the responsive models. Complete tumor regressions were observed after i.v., i.p. or oral administrations of F14512 and its antitumor activity was demonstrated over a range of 2-5 dose levels, providing evidence of its good tolerance. The level of cellular fluorescence emitted by the fluorescent probes was higher in cells sampled from tumors sensitive to F14512 treatments than from F14512-refractory tumors. We suggest that these probes could be used to identify tumors expressing a highly active PTS and guide the selection of patients that might be treated with F14512. These results emphasize the preclinical interest of this novel molecule and support its further clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Podophyllotoxin/analogs & derivatives , Polyamines/metabolism , Xenograft Model Antitumor Assays , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Biological Transport/drug effects , Cell Death/drug effects , Cell Line, Tumor , Flow Cytometry , Fluorescence , Humans , Immunohistochemistry , Mice , Podophyllotoxin/chemistry , Podophyllotoxin/pharmacology , Spermine/metabolism
10.
Mol Cancer Ther ; 8(10): 2780-90, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19808979

ABSTRACT

Triptolide, a natural product extracted from the Chinese plant Tripterygium wilfordii, possesses antitumor properties. Despite numerous reports showing the proapoptotic capacity and the inhibition of NF-kappaB-mediated transcription by triptolide, the identity of its cellular target is still unknown. To clarify its mechanism of action, we further investigated the effect of triptolide on RNA synthesis in the human non-small cell lung cancer cell line A549. Triptolide inhibited both total RNA and mRNA de novo synthesis, with the primary action being on the latter pool. We used 44K human pan-genomic DNA microarrays and identified the genes primarily affected by a short treatment with triptolide. Among the modulated genes, up to 98% are down-regulated, encompassing a large array of oncogenes including transcription factors and cell cycle regulators. We next observed that triptolide induced a rapid depletion of RPB1, the RNA polymerase II main subunit that is considered a hallmark of a transcription elongation blockage. However, we also show that triptolide does not directly interact with the RNA polymerase II complex nor does it damage DNA. We thus conclude that triptolide is an original pharmacologic inhibitor of RNA polymerase activity, affecting indirectly the transcription machinery, leading to a rapid depletion of short-lived mRNA, including transcription factors, cell cycle regulators such as CDC25A, and the oncogenes MYC and Src. Overall, the data shed light on the effect of triptolide on transcription, along with its novel potential applications in cancers, including acute myeloid leukemia, which is in part driven by the aforementioned oncogenic factors.


Subject(s)
Diterpenes/chemistry , Down-Regulation/drug effects , Phenanthrenes/chemistry , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase I/antagonists & inhibitors , Transcription, Genetic/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Diterpenes/pharmacology , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oligonucleotide Array Sequence Analysis , Phenanthrenes/pharmacology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Time Factors , Tumor Suppressor Protein p53 , ets-Domain Protein Elk-1/metabolism
11.
Bioorg Med Chem Lett ; 19(9): 2474-7, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19332372

ABSTRACT

The synthesis of a series of conjugated spermine derivatives with benzoxadiazole, phenylxanthene or bodipy fluorophores is described. These fluorescent probes were used to identify the activity of the polyamine transport system (PTS). N(1)-Methylspermine NBD conjugate 5 proved to have the optimal fluorescence characteristics and was used to show a selectivity for PTS-proficient CHO versus PTS-deficient CHO-MG cells. It can therefore be used as a tool for the selection of cells sensitive to cytotoxic compounds vectored through the PTS.


Subject(s)
Boron Compounds/chemistry , Chemistry, Pharmaceutical/methods , Fluorescent Dyes/pharmacology , Oxadiazoles/chemical synthesis , Rhodamines/chemistry , Spermine/chemical synthesis , Animals , Biological Transport , CHO Cells , Cricetinae , Cricetulus , Drug Design , Fluorescent Dyes/chemistry , Humans , Oxadiazoles/pharmacology , Polyamines/chemistry , Spermine/analogs & derivatives , Spermine/chemistry , Spermine/pharmacology
12.
Anticancer Drugs ; 20(5): 364-72, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19322071

ABSTRACT

The anaplastic lymphoma kinase (ALK) is a validated target for the therapy of different malignancies. Aberrant expression of constitutively active ALK chimeric proteins has been implicated in the pathogenesis of anaplastic large-cell lymphoma (ALCL) and has been detected in other cancers such as inflammatory myofibroblastic tumors, diffuse large B-cell lymphomas, certain non-small-cell lung cancers, rhabdomyosarcomas, neuroblastomas and glioblastomas. In the course of a screening program aimed at identifying kinase inhibitors with novel scaffolds, the two pyridoisoquinoline derivatives F91873 and F91874, were identified as multikinase inhibitors with activity against ALK in a biochemical screen. F91873 and F91874 also inhibited nucleophosmin-ALK and signal transducer and activator of transcription 3 phosphorylation in the ALCL cell line COST with the same potency. Both F91873 and F91874 behaved as ATP noncompetitive inhibitors and inhibited cell proliferation of the ALK(+) ALCL cell lines COST, PIO, and Karpas299 ALCL. This growth inhibition effect was associated with a G1-phase cell cycle arrest. Furthermore, administration of F91874 to severe combined immunodeficient mice bearing COST tumor xenografts resulted in a significant antitumor efficacy at 15 mg/kg/day, illustrating the potential utility of such compounds in the treatment of ALK-related pathologies.


Subject(s)
Antineoplastic Agents/therapeutic use , Lymphoma, Large-Cell, Anaplastic/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinolizines/therapeutic use , Thiazoles/therapeutic use , Anaplastic Lymphoma Kinase , Animals , Antineoplastic Agents/chemical synthesis , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Female , G1 Phase/drug effects , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Lymphoma, Large-Cell, Anaplastic/enzymology , Lymphoma, Large-Cell, Anaplastic/pathology , Mice , Mice, Inbred ICR , Mice, SCID , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Structure, Tertiary , Quinolizines/chemical synthesis , Receptor Protein-Tyrosine Kinases , Recombinant Fusion Proteins/antagonists & inhibitors , Thiazoles/chemical synthesis , Xenograft Model Antitumor Assays
13.
Cancer Res ; 68(23): 9845-53, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047165

ABSTRACT

The polyamine transport system (PTS) is an energy-dependent machinery frequently overactivated in cancer cells with a high demand for polyamines. We have exploited the PTS to selectively deliver a polyamine-containing drug to cancer cells. F14512 combines an epipodophyllotoxin core-targeting topoisomerase II with a spermine moiety introduced as a cell delivery vector. The polyamine tail supports three complementary functions: (a) facilitate formulation of a water-soluble compound, (b) increase DNA binding to reinforce topoisomerase II inhibition, and (c) facilitate selective uptake by tumor cells via the PTS. F14512 is 73-fold more cytotoxic to Chinese hamster ovary cells compared with CHO-MG cells with a reduced PTS activity. A decreased sensitivity of L1210 leukemia cells to F14512 was observed in the presence of putrescine, spermidine, and spermine. In parallel, the spermine moiety considerably enhances the drug-DNA interaction, leading to a reinforced inhibition of topoisomerase II. The spermine tail of F14512 serves as a cell delivery vehicle as well as a DNA anchor, and this property translates at the cellular level into a distinct pharmacologic profile. Twenty-nine human solid or hematologic cell lines were used to characterize the high cytotoxic potential of F14512 (median IC50 of 0.18 micromol/L). Finally, the potent antitumor activity of F14512 in vivo was evidenced with a MX1 human breast tumor xenograft model, with partial and complete tumor regressions. This work supports the clinical development of F14512 as a novel targeted cytotoxic drug and sheds light on the concept of selective delivery of drugs to tumor cells expressing the PTS.


Subject(s)
Biogenic Polyamines/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Podophyllotoxin/analogs & derivatives , Topoisomerase II Inhibitors , Animals , Binding, Competitive , Biogenic Polyamines/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , DNA Damage , DNA Topoisomerases, Type II/biosynthesis , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA, Neoplasm/metabolism , Drug Delivery Systems , Etoposide/pharmacology , Female , Humans , Leukemia L1210/drug therapy , Leukemia L1210/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Neoplasms/enzymology , Neoplasms/genetics , Podophyllotoxin/pharmacology , Xenograft Model Antitumor Assays
14.
Semin Oncol ; 35(3 Suppl 3): S13-21, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18538174

ABSTRACT

Vinflunine is an innovative microtubule inhibitor of the vinca alkaloid class with distinct tubulin-binding properties. Preclinical evaluation of this novel microtubule inhibitor has shown superior antitumor activity against a broad spectrum of tumor types in vitro and in vivo, in comparison with other vinca alkaloids. The antitumor effect of vinflunine is largely attributable to its modulation of microtubule dynamics, and is mediated by its ability to induce apoptosis in target cells. At non-cytotoxic concentrations, vinflunine also exerts antiangiogenic and antivascular activity. The favorable preclinical profile of vinflunine, in addition to its synergism with a variety of other therapeutic modalities, justifies further clinical development of this compound.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Microtubules/drug effects , Vinblastine/analogs & derivatives , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Cycle/drug effects , Drug Screening Assays, Antitumor , Humans , Mice , Signal Transduction , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , Vinblastine/pharmacology , Vinblastine/therapeutic use
15.
Semin Oncol ; 35(3 Suppl 3): S22-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18538175

ABSTRACT

Antimitotic drugs targeting the microtubules, such as the taxanes and vinca alkaloids, are widely used in the treatment of neoplastic diseases. Development of drug resistance over time, however, limits the efficacy of these agents and poses a clinical challenge to long-term improvement of patient outcomes. Understanding the mechanism(s) of drug resistance becomes paramount to allowing for alternative, if not improved, therapeutic options that might circumvent this challenge. Vinflunine, a novel microtubule inhibitor, has shown superior preclinical antitumor activity, and displays a different pattern of resistance, compared with other agents in the vinca alkaloid class.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Drug Resistance, Neoplasm , Microtubules/drug effects , Tubulin Modulators/therapeutic use , Vinblastine/analogs & derivatives , Actins/metabolism , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Humans , Multidrug Resistance-Associated Proteins/metabolism , Proteomics/methods , Tubulin/genetics , Vinca Alkaloids/pharmacology
16.
Biochem Pharmacol ; 76(4): 453-62, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18577376

ABSTRACT

The ubiquitin-proteasome pathway plays a critical role in the degradation of proteins involved in tumor growth and has therefore become a target for cancer therapy. In order to discover novel inhibitors of this pathway, a cellular assay reporter of proteasome activity was established. Human DLD-1 colon cancer cells were engineered to express a 4 ubiquitin-luciferase (DLD-1 4Ub-Luc) reporter protein, rapidly degraded via the ubiquitin-proteasome pathway and designed DLD-1 4Ub-Luc cells. Following treatment with reference proteasome inhibitors, the 4Ub-Luc protein accumulated in DLD-1 4Ub-Luc cells and a 80-fold increase in luciferase-produced bioluminescence signal was measured, as compared to untreated cells. The screening of over 30,000 compounds using this DLD-1 4Ub-Luc assay led to the identification of physalin B as a novel inhibitor of the ubiquitin-proteasome pathway. Indeed, physalin B induced an increase in bioluminescence from DLD-1 4Ub-Luc cells, at concentrations also producing an accumulation of ubiquitinated proteins and inhibiting TNFalpha-induced NF-kappaB activation. Physalin B did not inhibit catalytic activities of purified proteasome and interfered with cellular proteasomal catalytic activities at 4- to 8-fold higher concentrations than that required to induce significant increase in bioluminescence and accumulation of ubiquitinated proteins in DLD-1 4Ub-Luc cells. Furthermore, physalin B proved to be cytotoxic, triggered apoptosis in DLD-1 4Ub-Luc cells and induced the proapoptotic protein NOXA, characteristic of the proteasome signaling pathway. Therefore, the use of the DLD-1 4Ub-Luc assay allowed the identification of a novel inhibitor of the ubiquitin-proteasome pathway that might interfere with proteasome functions in a different way from reference proteasome inhibitors.


Subject(s)
Apoptosis/drug effects , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Secosteroids/pharmacology , Ubiquitin/metabolism , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Plant Extracts/chemistry , Protease Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/physiology , Signal Transduction
17.
Bioorg Med Chem Lett ; 18(3): 1212-6, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18083028

ABSTRACT

Novel derivatives of the marine alkaloid bengacarboline have been synthesized. The seco derivatives 11 and 12 were evaluated for topoisomerase inhibition, DNA damages, cytotoxicity and cell cycle perturbation. The two synthetic analogs are more potent cytotoxic agents than bengacarboline and they both induce an accumulation of cells in the S phase of DNA synthesis. They do not function as topoisomerase inhibitors but trigger DNA damages in cells.


Subject(s)
Alkaloids/chemical synthesis , Alkaloids/pharmacology , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Carbolines/chemical synthesis , Carbolines/pharmacology , Topoisomerase II Inhibitors , Alkaloids/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/physiology , Carbolines/chemistry , Cell Cycle/drug effects , DNA Damage/drug effects , Drug Screening Assays, Antitumor , Humans , Marine Biology , Molecular Structure , Urochordata/chemistry
18.
Eur J Med Chem ; 39(12): 1029-38, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15571864

ABSTRACT

The synthesis of several acridine thioethers is described. These compounds were oxidized to give new sulfoxides and sulfones. Among 23 compounds prepared, 19 were tested in vitro against the human cancer cell lines panel of NCI screening. Activity is increased 5-10 times from sulfides to sulfoxides. Among substituted groups in the side chain, sulfur mustard, epoxy sulfide and sulfoxide displayed the most interesting activity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Sulfides/chemical synthesis , Sulfones/chemical synthesis , Sulfoxides/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Saccharomyces cerevisiae/drug effects , Structure-Activity Relationship , Sulfides/pharmacology , Sulfones/pharmacology , Sulfoxides/pharmacology
19.
Curr Med Chem Anticancer Agents ; 4(5): 389-91, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15379689

ABSTRACT

In recent years, efforts have been made to improve the selectivity of anti-cancer agents via the targeting of cancer-specific proteins or signalization pathways. Novel anticancer drugs inhibiting defined kinases, the proteasome, and selected growth factor receptors for examples have been developed with success for a few cancer types. But in parallel to these novel "soft" drugs, conventional "hard" cytotoxic molecules targeting DNA, topoisomerases or tubuline remain extensively used to treat solid tumors. This letter evokes the utility and limitations of the two drug categories and comments on new directions of the antitumor pharmacology taken to improve the efficacy of cancer chemotherapy and the development of new molecules.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Design , Enzyme Inhibitors/pharmacology , Multienzyme Complexes/antagonists & inhibitors , Neoplasms/drug therapy , Cysteine Endopeptidases , Humans , Pharmacology/methods , Proteasome Endopeptidase Complex
20.
Clin Cancer Res ; 10(9): 3156-68, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15131057

ABSTRACT

PURPOSE: The purpose of the study was to investigate the mechanisms associated with antitumor activity and resistance to F11782, a novel dual catalytic inhibitor of topoisomerases with DNA repair-inhibitory properties. EXPERIMENTAL DESIGN: For that purpose, an F11782-resistant P388 leukemia subline (P388/F11782) has been developed in vivo and characterized. RESULTS: Weekly subtherapeutic doses of F11782 (10 mg/kg) induced complete resistance to F11782 after 8 weekly passages. This resistant P388/F11782 subline retained some in vivo sensitivity to several DNA-topoisomerase II and/or I complex-stabilizing poisons and showed marked collateral sensitivity to cisplatin, topotecan, colchicine, and Vinca alkaloids, while proving completely cross-resistant only to merbarone and doxorubicin. Therefore, resistance to F11782 did not appear to be associated with a classic multidrug resistance profile, as further reflected by unaltered drug uptake and no overexpression of resistance-related proteins or modification of the glutathione-mediated detoxification process. In vivo resistance to F11782 was, however, associated with a marked reduction in topoisomerase IIalpha protein (87%) and mRNA (50%) levels, as well as a diminution of the catalytic activity of topoisomerase IIalpha. In contrast, only minor reductions in topoisomerases IIbeta and I levels were recorded. However, of major interest, nucleotide excision repair activity was decreased 3-fold in these P388/F11782 cells and was more specifically associated with a decreased (67%) level of XPG (human xeroderma pigmentosum group G complementing protein), an endonuclease involved in this DNA repair system. CONCLUSIONS: These findings suggest that both topoisomerase IIalpha and XPG are major targets of F11782 in vivo and further demonstrate the original mechanism of action of this novel compound.


Subject(s)
DNA Repair , DNA Topoisomerases, Type II/metabolism , Etoposide/analogs & derivatives , Naphthalenes/therapeutic use , Neoplasms, Experimental/drug therapy , Pyrans/therapeutic use , Animals , Antigens, Neoplasm , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Blotting, Northern , Catalysis/drug effects , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/therapeutic use , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/genetics , Etoposide/administration & dosage , Etoposide/therapeutic use , Leukemia/drug therapy , Leukemia/enzymology , Leukemia/pathology , Mice , Mice, Inbred DBA , Mutation, Missense , Naphthalenes/administration & dosage , Neoplasm Transplantation , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/therapeutic use , Pyrans/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...