119 results on '"Giuseppe La Regina"'
Search Results
2. Targeting the Grb2 cSH3 domain: Design, synthesis and biological evaluation of the first series of modulators
- Author
-
Marianna Bufano, Michela Puxeddu, Marianna Nalli, Giuseppe La Regina, Angelo Toto, Francesca Romana Liberati, Alessio Paone, Francesca Cutruzzolà, Domiziana Masci, Chiara Bigogno, Giulio Dondio, Romano Silvestri, Stefano Gianni, and Antonio Coluccia
- Subjects
small molecules ,Anticancer ,Grb2-Gab2 system ,SH3 domain ,structure activity relationship ,Organic Chemistry ,Drug Discovery ,Molecular Biology ,Biochemistry - Published
- 2023
3. Single‐run chemo‐ and enantio‐selective high‐performance liquid chromatography separation of tramadol and its principal metabolite, O‐desmethyltramadol, using a chlorinated immobilized amylose‐based chiral stationary phase under multimodal elution conditions
- Author
-
Chiara Cantatore, Giuseppe La Regina, Rosella Ferretti, Romano Silvestri, and Roberto Cirilli
- Published
- 2022
4. An Innovation 10 Years in the Making: The Stories in the Pages of ACS Medicinal Chemistry Letters
- Author
-
Ryan A. Altman, Annalaura Brai, Jennifer Golden, Giuseppe La Regina, Zhengqiu Li, Terry W. Moore, William C. K. Pomerantz, Naomi S. Rajapaksa, and Ashley M. Adams
- Subjects
Organic Chemistry ,Drug Discovery ,Biochemistry - Abstract
[Image: see text] Innovation in medicinal chemistry has been at the heart of ACS Medicinal Chemistry Letters since the journal’s founding 10 years ago. In his inaugural editorial, Editor-in-Chief Dennis Liotta laid out a vision for the journal to become the “premier international journal for rapid communication of cutting-edge studies,” and, after 10 years, it has become exactly that. The great hope of drug discovery scientists is that their innovations will lead to new therapeutics to treat unmet medical needs. In the spirit of innovation and in celebration of the recent 10th anniversary of ACS Med. Chem. Lett., we highlight five therapeutics that were first reported or first comprehensively characterized within ACS Med. Chem. Lett.. This overview also serves to introduce the expansion of the scope of the Innovations article type to include Topical Innovations. With this extension, the journal hopes to provide a forum to showcase concise (rather than comprehensive) reviews of topics that are both timely and of great interest to the medicinal chemistry community. Moreover, these articles will emphasize the next steps to move the field toward new areas of interest in medicinal chemistry. Appropriate topics might include case studies of clinical candidates or approved drugs, new assay technologies in drug discovery, novel target classes, and innovative new approaches towards modulation of human physiology. Since its founding 10 years ago, ACS Med. Chem. Lett. has established itself as a venue for the rapid communication of studies in medicinal chemistry and drug discovery. There have been several drugs and clinical candidates that were first reported or first comprehensively characterized in ACS Med. Chem. Lett. In celebration of the 10th anniversary of ACS Med. Chem. Lett. this Topical Innovations article highlights five of these compounds: Ivosidenib, Siponimod, Glasdegib, Parsaclisib, and Dabrafenib.
- Published
- 2022
5. RS6077 induces mitotic arrest and selectively activates cell death in human cancer cell lines and in a lymphoma tumor in vivo
- Author
-
Jessica Sebastiani, Michela Puxeddu, Marianna Nalli, Ruoli Bai, Ludovica Altieri, Paola Rovella, Eugenio Gaudio, Daniela Trisciuoglio, Filippo Spriano, Patrizia Lavia, Cinzia Fionda, Domiziana Masci, Andrea Urbani, Chiara Bigogno, Giulio Dondio, Ernest Hamel, Francesco Bertoni, Romano Silvestri, and Giuseppe La Regina
- Subjects
Pharmacology ,Synthesis ,Lymphoma ,Mitotic arrest ,Pyrrole ,Tubulin ,Organic Chemistry ,Drug Discovery ,General Medicine - Abstract
We synthesized a new inhibitor of tubulin polymerization, the pyrrole (1-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-1H-pyrrol-3-yl)(3,4,5-trimethoxy-phenyl)methanone 6 (RS6077). Compound 6 inhibited the growth of multiple cancer cell lines, with IC
- Published
- 2023
6. Induction of Ferroptosis in Glioblastoma and Ovarian Cancers by a New Pyrrole Tubulin Assembly Inhibitor
- Author
-
Michela Puxeddu, Jianchao Wu, Ruoli Bai, Michele D’Ambrosio, Marianna Nalli, Antonio Coluccia, Simone Manetto, Alessia Ciogli, Domiziana Masci, Andrea Urbani, Cinzia Fionda, Sonia Coni, Rosa Bordone, Gianluca Canettieri, Chiara Bigogno, Giulio Dondio, Ernest Hamel, Te Liu, Romano Silvestri, and Giuseppe La Regina
- Subjects
Ovarian Neoplasms ,Mice ,Tubulin ,Cell Line, Tumor ,Drug Discovery ,Molecular Medicine ,Humans ,Animals ,Female ,Pyrroles ,Apoptosis ,Tubulin Modulators - Abstract
We synthesized new aroyl diheterocyclic pyrrole (ARDHEP)
- Published
- 2022
7. Emerging Direct Targeting β-Catenin Agents
- Author
-
Marianna Nalli, Domiziana Masci, Andrea Urbani, Giuseppe La Regina, and Romano Silvestri
- Subjects
Cell Nucleus ,Chemistry (miscellaneous) ,Neoplasms ,Organic Chemistry ,Drug Discovery ,Molecular Medicine ,Pharmaceutical Science ,Humans ,Antineoplastic Agents ,Physical and Theoretical Chemistry ,Wnt Signaling Pathway ,beta Catenin ,Analytical Chemistry - Abstract
Aberrant accumulation of β-catenin in the cell nucleus as a result of deregulation of the Wnt/β-catenin pathway is found in various types of cancer. Direct β-catenin targeting agents are being researched despite obstacles; however, specific β-catenin drugs for clinical treatments have not been approved so far. We focused on direct β-catenin targeting of potential therapeutic value as anticancer agents. This review provides recent advances on small molecule β-catenin agents. Structure-activity relationships and biological activities of reported inhibitors are discussed. This work provides useful knowledge in the discovery of β-catenin agents.
- Published
- 2022
8. In This Issue, Volume 13, Issue 10
- Author
-
William C. K. Pomerantz and Giuseppe La Regina
- Subjects
Organic Chemistry ,Drug Discovery ,Biochemistry - Published
- 2022
9. Discovery of novel human lactate dehydrogenase inhibitors: Structure-based virtual screening studies and biological assessment
- Author
-
Laura Di Magno, Antonio Coluccia, Marianna Bufano, Silvia Ripa, Giuseppe La Regina, Marianna Nalli, Fiorella Di Pastena, Gianluca Canettieri, Romano Silvestri, and Luigi Frati
- Subjects
Pharmacology ,L-Lactate Dehydrogenase ,Neoplasms ,Organic Chemistry ,Drug Discovery ,Humans ,General Medicine ,Lactic Acid ,Enzyme Inhibitors ,Glycolysis ,Oxidative Phosphorylation ,Cell Line - Abstract
Most cancer cells switch their metabolism from mitochondrial oxidative phosphorylation to aerobic glycolysis to generate ATP and precursors for the biosynthesis of key macromolecules. The aerobic conversion of pyruvate to lactate, coupled to oxidation of the nicotinamide cofactor, is a primary hallmark of cancer and is catalyzed by lactate dehydrogenase (LDH), a central effector of this pathological reprogrammed metabolism. Hence, inhibition of LDH is a potential new promising therapeutic approach for cancer. In the search for new LDH inhibitors, we carried out a structure-based virtual screening campaign. Here, we report the identification of a novel specific LDH inhibitor, the pyridazine derivative 18 (RS6212), that exhibits potent anticancer activity within the micromolar range in multiple cancer cell lines and synergizes with complex I inhibition in the suppression of tumor growth. Altogether, our data support the conclusion that compound 18 deserves to be further investigated as a starting point for the development of LDH inhibitors and for novel anticancer strategies based on the targeting of key metabolic steps.
- Published
- 2022
10. Anticancer Activity of (
- Author
-
Antonio, Coluccia, Marianna, Bufano, Giuseppe, La Regina, Michela, Puxeddu, Angelo, Toto, Alessio, Paone, Amani, Bouzidi, Giorgia, Musto, Nadia, Badolati, Viviana, Orlando, Stefano, Biagioni, Domiziana, Masci, Chiara, Cantatore, Roberto, Cirilli, Francesca, Cutruzzolà, Stefano, Gianni, Mariano, Stornaiuolo, and Romano, Silvestri
- Abstract
Wingless/integrase-11 (WNT)/β-catenin pathway is a crucial upstream regulator of a huge array of cellular functions. Its dysregulation is correlated to neoplastic cellular transition and cancer proliferation. Members of the Dishevelled (DVL) family of proteins play an important role in the transduction of WNT signaling by contacting its cognate receptor, Frizzled, via a shared PDZ domain. Thus, negative modulators of DVL1 are able to impair the binding to Frizzled receptors, turning off the aberrant activation of the WNT pathway and leading to anti-cancer activity. Through structure-based virtual screening studies, we identified racemic compound RS4690 (
- Published
- 2022
11. In This Issue, Volume 13, Issue 1
- Author
-
Giuseppe La Regina
- Subjects
Organic Chemistry ,Drug Discovery ,Biochemistry - Published
- 2022
12. Modulating undruggable targets to overcome cancer therapy resistance
- Author
-
Catherine Passirani, Anne Vessières, Giuseppe La Regina, Wolfgang Link, Romano Silvestri, Fundação para a Ciência e a Tecnologia (Portugal), Ministerio de Ciencia, Innovación y Universidades (España), and Agencia Estatal de Investigación (España)
- Subjects
Pharmacology ,Cell Nucleus ,Cancer Research ,Cytoplasmic and Nuclear ,Active Transport, Cell Nucleus ,Drug Resistance ,Receptors, Cytoplasmic and Nuclear ,Drug development ,Antineoplastic Agents ,Cancer ,Drug resistance ,Nanomedicine ,Therapeutic targets ,Drug Resistance, Neoplasm ,Humans ,Neoplasms ,Active Transport ,Infectious Diseases ,Oncology ,Receptors ,Neoplasm ,Pharmacology (medical) - Abstract
Many cancer patients frequently fail to respond to anti-cancer treatment due to therapy resistance which is the major obstacle towards curative cancer treatment. Therefore, identification of the molecular mechanisms underlying resistance is of paramount clinical and economic importance. The advent of targeted therapies based on a molecular understanding of cancer could serve as a model for strategies to overcome drug resistance. Accordingly, the identification and validation of proteins critically involved in resistance mechanisms represent a path towards innovative therapeutic strategies to improve the clinical outcome of cancer patients. In this review, we discuss emerging targets, small molecule therapeutics and drug delivery strategies to overcome therapy resistance. We focus on rational treatment strategies based on transcription factors, pseudokinases, nuclear export receptors and immunogenic cell death strategy. Historically, unliganded transcription factors and pseudokinases were considered undruggable while blocking the nuclear export e.g., through inhibition of the nuclear export receptor CRM1 was predicted as highly toxic. Recent success inhibiting Gli-1, HIF-1α, HIF-2α and reactivating the tumor suppressor transcription factors p53 and FOXO illustrates the feasibility and power of this targeting approach. Similarly, progress has been made in modulating the activity of pseudokinase proteins implicated in therapy resistance including members of the Tribbles protein family. On the other hand, the recent clinical approval of Selinexor, a specific inhibitor of CRM-1, a protein that mediates the transport of cargos with leucine-rich nuclear export signals and known to be a driver of drug resistance, represents the proof-of-concept for inhibiting the nuclear export as a feasible strategy to overcome therapy resistance. The ever-growing capacity to target resistance mechanisms with judiciously selected small molecules, some of which are being formulated within smart nanoparticles, will pave the way towards the improvement of the clinical outcome and realize the full potential of targeted therapies and immunotherapies., This work was supported by the FUNDAÇÃO PARA A CIÊNCIA E TECNOLOGIA (FCT) Research Center Grant UID/BIM/04773/2013, Centre for Biomedical Research (CBMR), and by the Spanish Ministry of Science, Innovation and Universities through Grant RTI2018-094629-B-I00 to WL.
- Published
- 2022
13. Anticancer Activity of (S)-5-Chloro-3-((3,5-dimethylphenyl)sulfonyl)-N-(1-oxo-1-((pyridin-4-ylmethyl)amino)propan-2-yl)-1H-indole-2-carboxamide (RS4690), a New Dishevelled 1 Inhibitor
- Author
-
Antonio Coluccia, Marianna Bufano, Giuseppe La Regina, Michela Puxeddu, Angelo Toto, Alessio Paone, Amani Bouzidi, Giorgia Musto, Nadia Badolati, Viviana Orlando, Stefano Biagioni, Domiziana Masci, Chiara Cantatore, Roberto Cirilli, Francesca Cutruzzolà, Stefano Gianni, Mariano Stornaiuolo, Romano Silvestri, Coluccia, Antonio, Bufano, Marianna, La Regina, Giuseppe, Puxeddu, Michela, Toto, Angelo, Paone, Alessio, Bouzidi, Amani, Musto, Giorgia, Badolati, Nadia, Orlando, Viviana, Biagioni, Stefano, Masci, Domiziana, Cantatore, Chiara, Cirilli, Roberto, Cutruzzolà, Francesca, Gianni, Stefano, Stornaiuolo, Mariano, and Silvestri, Romano
- Subjects
Cancer Research ,DVL1 ,Oncology ,WNT pathway ,cancer ,PDZ domain ,virtual screening - Abstract
Wingless/integrase-11 (WNT)/β-catenin pathway is a crucial upstream regulator of a huge array of cellular functions. Its dysregulation is correlated to neoplastic cellular transition and cancer proliferation. Members of the Dishevelled (DVL) family of proteins play an important role in the transduction of WNT signaling by contacting its cognate receptor, Frizzled, via a shared PDZ domain. Thus, negative modulators of DVL1 are able to impair the binding to Frizzled receptors, turning off the aberrant activation of the WNT pathway and leading to anti-cancer activity. Through structure-based virtual screening studies, we identified racemic compound RS4690 (1), which showed a promising selective DVL1 binding inhibition with an EC50 of 0.74 ± 0.08 μM. Molecular dynamic simulations suggested a different binding mode for the enantiomers. In the in vitro assays, enantiomer (S)-1 showed better inhibition of DVL1 with an EC50 of 0.49 ± 0.11 μM compared to the (R)-enantiomer. Compound (S)-1 inhibited the growth of HCT116 cells expressing wild-type APC with an EC50 of 7.1 ± 0.6 μM and caused a high level of ROS production. These results highlight (S)-1 as a lead compound for the development of new therapeutic agents against WNT-dependent colon cancer.
- Published
- 2022
14. RS6077 Induces Mitotic Arrest and Selectively Activates Cell Death in Human Cancer Cell Lines and in a Lymphoma Tumor In Vivo
- Author
-
Jessica Sebastiani, Michela Puxeddu, Marianna Nalli, Ruoli Bai, Ludovica Altieri, Paola Rovella, Eugenio Gaudio, Daniela Trisciuoglio, Filippo Spriano, Patrizia Lavia, Cinzia Fionda, Ernest Hamel, Francesco Bertoni, Romano Silvestri, and Giuseppe La Regina
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2022
15. A Novel Validated UHPLC Method for the Estimation of Rosuvastatin and Its Complete Impurity Profile in Tablet Formulations
- Author
-
Francesca Romana Mammone, Leo Zanitti, Michela Puxeddu, Giuseppe La Regina, Romano Silvestri, Anna Borioni, and Roberto Cirilli
- Subjects
rosuvastatin calcium salt ,impurities ,European pharmacopoeia ,UHPLC ,method validation ,reversed-phase ,tablets ,Chemistry (miscellaneous) ,Organic Chemistry ,Drug Discovery ,Molecular Medicine ,Pharmaceutical Science ,Physical and Theoretical Chemistry ,Analytical Chemistry - Abstract
A key step in the development of medicinal products is the research and validation of selective and sensitive analytical methods for the control of impurities from synthesis and degradation. As most impurities are similar in structure to the drug substance, the achievement of chemo-selective conditions is usually challenging. Herein, a direct and highly selective ultra-high-performance liquid chromatographic method for determining the assay and related substances content in medicinal products containing rosuvastatin calcium salt (RSV) is presented. RSV is used to treat high cholesterol levels and prevent heart attacks and strokes. The most engaging feature of this method was the baseline separation of all organic related substances listed in the European Pharmacopoeia (EP) monograph for the RSV tablets, achieved for the first time in less than 15 min using the Acquity BEH C18 (100 mm × 2.1 mm, 1.7 μm) column under reversed-phase isocratic conditions. The mobile phase adopted for the chemo-selective analysis does not contain buffers but instead contains trifluoroacetic as an acid additive. The chromatographic method was validated according to the guidelines of the International Conference on Harmonization (ICH) and proved to be linear, precise and accurate for determining the content of RSV and related chiral substances in tablet formulations.
- Published
- 2023
16. Emerging Therapeutic Agents for Colorectal Cancer
- Author
-
Marianna Nalli, Michela Puxeddu, Giuseppe La Regina, Stefano Gianni, and Romano Silvestri
- Subjects
emerging targets ,Pharmaceutical Science ,Organic chemistry ,Anticancer agents ,cancer ,CRC ,Review ,digestive system diseases ,Neoplasm Proteins ,Analytical Chemistry ,anticancer agents ,QD241-441 ,Chemistry (miscellaneous) ,Drug Discovery ,Humans ,Molecular Medicine ,Molecular Targeted Therapy ,Physical and Theoretical Chemistry ,Colorectal Neoplasms ,Protein Kinase Inhibitors ,neoplasms - Abstract
There are promising new therapeutic agents for CRC patients, including novel small-molecule inhibitors and immune checkpoint blockers. We focused on emerging CRC’s therapeutic agents that have shown the potential for progress in clinical practice. This review provides an overview of tyrosine kinase inhibitors targeting VEGF and KIT, BRAF and MEK inhibitors, TLR9 agonist, STAT3 inhibitors, and immune checkpoint blockers (PD1/PDL-1 inhibitors), for which recent advances have been reported. These new agents have the potential to provide benefits to CRC patients with unmet medical needs.
- Published
- 2021
17. Targeting PDZ domains as potential treatment for viral infections, neurodegeneration and cancer
- Author
-
Lorenzo Visconti, Stefano Gianni, Romano Silvestri, Giuseppe La Regina, Antonio Coluccia, Livia Pagano, Marianna Bufano, Angelo Toto, Caterina Nardella, Francesca Malagrinò, Marianna Nalli, Nardella, C., Visconti, L., Malagrino, Francesca., Pagano, L., Bufano, M., Nalli, M., Coluccia, A., La Regina, G., Silvestri, R., Gianni, S., and Toto, A.
- Subjects
PDZ Domain ,Inhibitor ,QH301-705.5 ,Immunology ,Protein domain ,PDZ domain ,PDZ Domains ,Ligand ,Review ,Computational biology ,Protein–protein interactions ,Biology ,Ligands ,General Biochemistry, Genetics and Molecular Biology ,Protein–protein interaction ,Neoplasms ,Cell polarity ,medicine ,Humans ,Biology (General) ,Ecology, Evolution, Behavior and Systematics ,Binding selectivity ,Inhibitors ,Protein ,Applied Mathematics ,Small molecules ,Neurodegeneration ,Proteins ,medicine.disease ,Virus Diseases ,Modeling and Simulation ,Neoplasm ,Signal transduction ,General Agricultural and Biological Sciences ,inhibitors ,small molecules ,protein–protein interactions ,Small molecule ,Function (biology) ,Human ,Protein Binding - Abstract
The interaction between proteins is a fundamental event for cellular life that is generally mediated by specialized protein domains or modules. PDZ domains are the largest class of protein–protein interaction modules, involved in several cellular pathways such as signal transduction, cell–cell junctions, cell polarity and adhesion, and protein trafficking. Because of that, dysregulation of PDZ domain function often causes the onset of pathologies, thus making this family of domains an interesting pharmaceutical target. In this review article we provide an overview of the structural and functional features of PDZ domains and their involvement in the cellular and molecular pathways at the basis of different human pathologies. We also discuss some of the strategies that have been developed with the final goal to hijack or inhibit the interaction of PDZ domains with their ligands. Because of the generally low binding selectivity of PDZ domain and the scarce efficiency of small molecules in inhibiting PDZ binding, this task resulted particularly difficult to pursue and still demands increasing experimental efforts in order to become completely feasible and successful in vivo.
- Published
- 2021
18. Discovery of a Novel Class of Norovirus Inhibitors with High Barrier of Resistance
- Author
-
Giuseppe La Regina, Jasper Rymenants, Romano Silvestri, Jana Van Dycke, Jessica Sebastiani, Michela Puxeddu, Eloise Mastrangelo, Marianna Nalli, Joana Rocha-Pereira, Jelle Matthijnssens, Delia Tarantino, and Johan Neyts
- Subjects
small molecule ,Pharmaceutical Science ,norovirus ,medicine.disease_cause ,Article ,Caliciviridae ,in vitro ,Norovirus ,Pharmacy and materia medica ,Drug Discovery ,medicine ,Potency ,Replicon ,EC50 ,Mutation ,biology ,Chemistry ,biology.organism_classification ,Small molecule ,Virology ,In vitro ,RS1-441 ,Molecular Medicine ,Medicine - Abstract
Human noroviruses (HuNoVs) are the most common cause of viral gastroenteritis resulting in ~219,000 deaths annually and a societal cost of ~USD60 billion. There are no antivirals or vaccines available to treat and/or prevent HuNoV. In this study, we performed a large-scale phenotypical antiviral screening using the mouse norovirus (MNV), which included ~1000 drug-like small molecules from the Drug Design and Synthesis Centre (Sapienza University, Rome). Compound 3-((3,5-dimethylphenyl)sulfonyl)-5-chloroindole-N-(phenylmethanol-4-yl)-2.carboxamide (compound 1) was identified as an inhibitor of MNV replication with an EC50 of 0.5 ± 0.1 µM. A series of 10 analogs were synthesized of which compound 6 showed an improved potency/selectivity (EC50 0.2 ± 0.1 µM) against MNV, good activity was also observed against the HuNoV GI replicon (EC50 1.2 ± 0.6 µM). Time-of-drug-addition studies revealed that analog 6 acts at a time point that coincides with the onset of viral RNA replication. After six months of selective pressure, two compound 6res variants were independently selected, both harboring one mutation in VPg and three mutations in the RdRp. After reverse engineering S131T and Y154F as single mutations into the MNV backbone, we did not find a markedly compound 6res phenotype. In this study, we present a class of novel norovirus inhibitors with a high barrier to resistance and in vitro antiviral activity.
- Published
- 2021
19. In This Issue, Volume 13, Issue 9
- Author
-
Giuseppe La Regina
- Subjects
Organic Chemistry ,Drug Discovery ,Biochemistry - Published
- 2022
20. In This Issue, Volume 12, Issue 3
- Author
-
Giuseppe La Regina
- Subjects
Organic Chemistry ,Drug Discovery ,Biochemistry - Published
- 2021
21. Discovery of pyrrole derivatives for the treatment of glioblastoma and chronic myeloid leukemia
- Author
-
Te Liu, Michela Puxeddu, Antonio Coluccia, Ruoli Bai, Maria Stefania Sinicropi, Chiara Tremolanti, Romano Silvestri, Eleonora Da Pozzo, Stefano Biagioni, Jessica Sebastiani, Giuseppe La Regina, Addolorata Coluccia, Marianna Bufano, Viviana Orlando, Jessica Ceramella, Claudia Martini, Ernest Hamel, Domenico Iacopetta, Hongliang Shen, Diego Brancaccio, and Marianna Nalli
- Subjects
Angiogenesis ,medicine.disease_cause ,Pyrrole ,01 natural sciences ,Polymerization ,Mice ,Synthesis ,Heterocyclic Compounds ,Tubulin ,hemic and lymphatic diseases ,Drug Discovery ,Tumor Cells, Cultured ,Glioblastoma ,Leukemia ,Mice, Inbred BALB C ,0303 health sciences ,Molecular Structure ,biology ,Chemistry ,Myeloid leukemia ,General Medicine ,Tubulin Modulators ,Female ,Stem cell ,Methane ,Tyrosine kinase ,Cell Survival ,Mice, Nude ,Antineoplastic Agents ,Structure-Activity Relationship ,03 medical and health sciences ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Animals ,Humans ,Cell Proliferation ,030304 developmental biology ,Pharmacology ,Dose-Response Relationship, Drug ,010405 organic chemistry ,Topoisomerase ,Organic Chemistry ,Neoplasms, Experimental ,medicine.disease ,0104 chemical sciences ,Imatinib mesylate ,Cancer research ,biology.protein ,Drug Screening Assays, Antitumor ,Carcinogenesis - Abstract
Long-term survivors of glioblastoma multiforme (GBM) are at high risk of developing second primary neoplasms, including leukemia. For these patients, the use of classic tyrosine kinase inhibitors (TKIs), such as imatinib mesylate, is strongly discouraged, since this treatment causes a tremendous increase of tumor and stem cell migration and invasion. We aimed to develop agents useful for the treatment of patients with GBM and chronic myeloid leukemia (CML) using an alternative mechanism of action from the TKIs, specifically based on the inhibition of tubulin polymerization. Compounds 7 and 25, as planned, not only inhibited tubulin polymerization, but also inhibited the proliferation of both GMB and CML cells, including those expressing the T315I mutation, at nanomolar concentrations. In in vivo experiments in BALB/cnu/nu mice injected subcutaneously with U87MG cells, in vivo, 7 significantly inhibited GBM cancer cell proliferation, in vivo tumorigenesis, and tumor growth, tumorigenesis and angiogenesis. Compound 7 was found to block human topoisomerase II (hTopoII) selectively and completely, at a concentration of 100 μM.
- Published
- 2021
22. RS-5645 attenuates inflammatory cytokine storm induced by SARS-CoV-2 spike protein and LPS by modulating pulmonary microbiota
- Author
-
Fangfang Dou, Giuseppe La Regina, Zhihua Yu, Jiulin Chen, Chuan Chen, Te Liu, Romano Silvestri, Zhangbin Gong, Changpeng Han, and Jianchao Wu
- Subjects
Lipopolysaccharides ,Male ,CD3 ,Anti-Inflammatory Agents ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Peripheral blood mononuclear cell ,Microbiology ,Interferon-gamma ,Mice ,4-(thiophen-3-yl)-1-(p-tolyl)-1H-pyrrol-3-yl)(3,4,5-trimethoxyphenyl)methanone (RS-5645) ,Antigens, CD ,medicine ,4-(thiophen-3-yl)-1-(p-tolyl)-1H-pyrrol-3-yl)(3 ,Animals ,Humans ,pulmonary microbiota ,SARS-CoV-2 (COVID-19) spike protein ,inflammatory factor storm ,Lung ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Mice, Inbred BALB C ,biology ,SARS-CoV-2 ,Chemistry ,Streptococcus ,Microbiota ,COVID-19 ,Translation (biology) ,Cell Biology ,Metabolism ,Ribosomal RNA ,medicine.disease ,5-trimethoxyphenyl)methanone (RS-5645) ,Disease Models, Animal ,Integrin alpha M ,Spike Glycoprotein, Coronavirus ,biology.protein ,Cytokine Release Syndrome ,Cytokine storm ,Research Paper ,Developmental Biology - Abstract
An inflammatory cytokine storm is considered an important cause of death in severely and critically ill COVID-19 patients, however, the relationship between the SARS-CoV-2 spike (S) protein and the host's inflammatory cytokine storm is not clear. Here, the qPCR results indicated that S protein induced a significantly elevated expression of multiple inflammatory factor mRNAs in peripheral blood mononuclear cells (PBMCs), whereas RS-5645 ((4-(thiophen-3-yl)-1-(p-tolyl)-1H-pyrrol-3-yl)(3,4,5-trimethoxyphenyl)methanone) attenuated the expression of the most inflammatory factor mRNAs. RS-5645 also significantly reduced the cellular ratios of CD45+/IFNγ+, CD3+/IFNγ+, CD11b+/IFNγ+, and CD56+/IFNγ+ in human PBMCs. In addition, RS-5645 effectively inhibited the activation of inflammatory cells and reduced inflammatory damage to lung tissue in mice. Sequencing results of 16S rRNA v3+v4 in mouse alveolar lavage fluid showed that there were 494 OTUs overlapping between the alveolar lavage fluid of mice that underwent S protein+ LPS-combined intervention (M) and RS-5645-treated mice (R), while R manifested 64 unique OTUs and M exhibited 610 unique OTUs. In the alveoli of group R mice, the relative abundances of microorganisms belonging to Porphyromonas, Rothia, Streptococcus, and Neisseria increased significantly, while the relative abundances of microorganisms belonging to Psychrobacter, Shimia, and Sporosarcina were significantly diminished. The results of KEGG analysis indicated that the alveolar microbiota of mice in the R group can increase translation and reduce the activity of amino acid metabolism pathways. COG analysis results indicated that the abundance of proteins involved in ribosomal structure and biogenesis related to metabolism was augmented in the alveolar microbiota of the mice in the R group, while the abundance of proteins involved in secondary metabolite biosynthesis was significantly reduced. Therefore, our research results showed that RS-5645 attenuated pulmonary inflammatory cell infiltration and the inflammatory storm induced by the S protein and LPS by modulating the pulmonary microbiota.
- Published
- 2021
23. RS4651 suppresses lung fibroblast activation via the TGF-β1/SMAD signalling pathway
- Author
-
Romano Silvestri, Anjian Xu, Giuseppe La Regina, Yanmeng Li, Chunting Tan, Shirong Li, Haoyan Wang, and Wenjie Qi
- Subjects
0301 basic medicine ,Male ,RS4651 ,Pulmonary fibrosis ,SMAD7 ,TGF-β1 ,α-SMA ,SMAD ,Smad2 Protein ,Bleomycin ,Cell Line ,Smad7 Protein ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Idiopathic pulmonary fibrosis ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,medicine ,Gene silencing ,Animals ,Humans ,Pharmacology ,Lung ,Chemistry ,Fibroblasts ,medicine.disease ,Actins ,Up-Regulation ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,030217 neurology & neurosurgery ,Transforming growth factor ,Signal Transduction - Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive disease resulting in respiratory failure with no efficient treatment options. We investigated the protective effect of RS4651 on pulmonary fibrosis in mice and the mechanism. Methods Intratracheal injection of bleomycin (BLM) was used to induce pulmonary fibrosis in mice. RS4561 was administered intraperitoneally at different doses. Histopathological changes were observed. The level of alpha-smooth muscle actin (α-SMA) were also tested. In vitro, the proliferation and migratory effects of RS4651 treatment on MRC-5 cells pre-treated with transforming growth factor (TGF-β1) were examined. RNA-sequencing was used to detect differentially expressed target genes. Then, the expression of α-SMA, pSMAD2 and SMAD7 were analysed during RS4651 treatment of MRC-5 cells with or without silencing by SMAD7 siRNA. Results Histopathological staining results showed decreased collagen deposition in RS4651 administered mice. Additionally, a lower level of α-SMA was also observed compared to the BLM group. The results of in vitro studies confirmed that RS4651 can inhibit the proliferation and migration, as well as α-SMA and pSMAD2 expression in MRC-5 cells treated with TGF-β1. RNA-sequencing data identified the target gene SMAD7. We found that RS4651 could upregulate SMAD7 expression and inhibit the proliferation and migration of MRC-5 cells via SMAD7, and RS4651 inhibition of α-SMA and pSMAD2 expression was blocked in SMAD7-siRNA MRC-5 cells. In vivo studies further confirmed that RS4651 could upregulate SMAD7 expression in BLM-induced lung fibrosis in mice. Conclusions Our data suggest that RS4651 alleviates BLM-induced pulmonary fibrosis in mice by inhibiting the TGF-β1/SMAD signalling pathway.
- Published
- 2021
24. Targeting the Interaction between the SH3 Domain of Grb2 and Gab2
- Author
-
Marianna Bufano, Lorenzo Visconti, Francesca Cutruzzolà, Alessio Paone, Francesca Troilo, Maria Chiara Magnifico, Romano Silvestri, Angelo Toto, Francesca Malagrinò, Antonio Coluccia, Michela Puxeddu, Stefano Gianni, Giuseppe La Regina, Malagrino, Francesca., Coluccia, A., Bufano, M., Regina, G., Puxeddu, M., Toto, A., Visconti, L., Paone, A., Magnifico, M. C., Troilo, F., Cutruzzola, F., Silvestri, R., and Gianni, S.
- Subjects
0301 basic medicine ,Scaffold protein ,MAPK/ERK pathway ,Models, Molecular ,Reproducibility of Result ,GAB2 ,Article ,SH3 domain ,Fluorescence ,src Homology Domains ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Humans ,lcsh:QH301-705.5 ,Inhibitory effect ,Adaptor Proteins, Signal Transducing ,GRB2 Adaptor Protein ,Kinetic ,Gab2 ,Virtual screening ,cancer cell lines ,biology ,Chemistry ,Reproducibility of Results ,kinetics ,virtual screening ,General Medicine ,Cell biology ,030104 developmental biology ,Lung cancer cell ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,biology.protein ,GRB2 ,cancer cell line ,Human ,Protein Binding - Abstract
Gab2 is a scaffolding protein, overexpressed in many types of cancers, that plays a key role in the formation of signaling complexes involved in cellular proliferation, migration, and differentiation. The interaction between Gab2 and the C-terminal SH3 domain of the protein Grb2 is crucial for the activation of the proliferation-signaling pathway Ras/Erk, thus representing a potential pharmacological target. In this study, we identified, by virtual screening, seven potential inhibitor molecules that were experimentally tested through kinetic and equilibrium binding experiments. One compound showed a remarkable effect in lowering the affinity of the C-SH3 domain for Gab2. This inhibitory effect was subsequently validated in cellula by using lung cancer cell lines A549 and H1299. Our results are discussed under the light of previous works on the C-SH3:Gab2 interaction.
- Published
- 2020
- Full Text
- View/download PDF
25. CXCR4 antagonism sensitizes cancer cells to novel indole-based MDM2/4 inhibitors in glioblastoma multiforme
- Author
-
Claudia Martini, Valeria La Pietra, Romano Silvestri, Linda Cerofolini, Simona Daniele, Michela Puxeddu, Sabrina Taliani, Chiara Cavallini, Martina Pedrini, Stefano Giuntini, Marianna Nalli, Vincenzo Maria D'Amore, Luciana Marinelli, Marco Fragai, Claudio Luchinat, Ettore Novellino, Deborah Pietrobono, Rebecca Piccarducci, Giuseppe La Regina, Pasquale Russomanno, Daniele, S., La Pietra, V., Piccarducci, R., Pietrobono, D., Cavallini, C., D'Amore, V. M., Cerofolini, L., Giuntini, S., Russomanno, P., Puxeddu, M., Nalli, M., Pedrini, M., Fragai, M., Luchinat, C., Novellino, E., Taliani, S., La Regina, G., Silvestri, R., Martini, C., and Marinelli, L.
- Subjects
0301 basic medicine ,p53 ,Benzylamines ,Indoles ,Cell Cycle Proteins ,Cyclams ,CXCR4 ,Benzylamine ,0302 clinical medicine ,Cell Movement ,Cell Cycle Protein ,Antineoplastic Combined Chemotherapy Protocols ,Proto-Oncogene Protein ,CXCR4 antagonist ,biology ,Chemistry ,GBM stem-Like cells (GSCs) ,Brain Neoplasms ,Drug Synergism ,Proto-Oncogene Proteins c-mdm2 ,Cyclam ,Neoplastic Stem Cells ,Mdm2 ,Stem cell ,Human ,Signal Transduction ,Receptors, CXCR4 ,Neurogenesis ,Brain Neoplasm ,03 medical and health sciences ,MDM4 ,Downregulation and upregulation ,MDM2 ,Glioma ,Neurosphere ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Spheroids, Cellular ,medicine ,Humans ,Neoplasm Invasiveness ,Cell Proliferation ,Pharmacology ,Neoplasm Invasivene ,Glioblastoma multiforme (GBM) ,Antineoplastic Combined Chemotherapy Protocol ,medicine.disease ,030104 developmental biology ,Indole ,Cancer cell ,biology.protein ,Cancer research ,Neurogenesi ,Neoplastic Stem Cell ,Tumor Suppressor Protein p53 ,Glioblastoma ,030217 neurology & neurosurgery - Abstract
Glioblastoma Multiforme (GBM) is a highly invasive primary brain tumour characterized by chemo- and radio-resistance and poor overall survival. GBM can present an aberrant functionality of p53, caused by the overexpression of the murine double minute 2 protein (MDM2) and its analogue MDM4, which may influence the response to conventional therapies. Moreover, tumour resistance/invasiveness has been recently attributed to an overexpression of the chemokine receptor CXCR4, identified as a pivotal mediator of glioma neovascularization. Notably, CXCR4 and MDM2-4 cooperate in promoting tumour invasion and progression. Although CXCR4 actively promotes MDM2 activation leading to p53 inactivation, MDM2-4 knockdown induces the downregulation of CXCR4 gene transcription. Our study aimed to assess if the CXCR4 signal blockade could enhance glioma cells' sensitivity to the inhibition of the p53-MDMs axis. Rationally designed inhibitors of MDM2/4 were combined with the CXCR4 antagonist, AMD3100, in human GBM cells and GBM stem-like cells (neurospheres), which are crucial for tumour recurrence and chemotherapy resistance. The dual MDM2/4 inhibitor RS3594 and the CXCR4 antagonist AMD3100 reduced GBM cell invasiveness and migration in single-agent treatment and mainly in combination. AMD3100 sensitized GBM cells to the antiproliferative activity of RS3594. It is noteworthy that these two compounds present synergic effects on cancer stem components: RS3594 inhibited the growth and formation of neurospheres, AMD3100 induced differentiation of neurospheres while enhancing RS3594 effectiveness preventing their proliferation/clonogenicity. These results confirm that blocking CXCR4/MDM2/4 represents a valuable strategy to reduce GBM proliferation and invasiveness, acting on the stem cell component too.
- Published
- 2020
26. Discovery of Zika Virus NS2B/NS3 Inhibitors That Prevent Mice from Life-Threatening Infection and Brain Damage
- Author
-
Romano Silvestri, Joachim J. Bugert, Eloise Mastrangelo, Jin-Ching Lee, Giuseppe La Regina, Juliane Nolte, Marianna Nalli, Benno Schreiner, Yu-Hsuan Wu, Antonio Coluccia, Tasneem Elamin, Michela Puxeddu, Frank Schwarze, Delia Tarantino, and Chih-Ku Wei
- Subjects
synthesis ,mouse model ,medicine.medical_treatment ,Brain damage ,01 natural sciences ,Biochemistry ,Zika virus ,Zika ,antivirals ,Drug Discovery ,medicine ,ZikV Infection ,zika virus ,Biological evaluation ,NS3 ,Protease ,biology ,010405 organic chemistry ,Organic Chemistry ,modeling ,biology.organism_classification ,Virology ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,NS2B/NS3 protease ,medicine.symptom - Abstract
[Image: see text] Zika virus (ZIKV) infection, which initially was endemic only in Africa and Asia, is rapidly spreading throughout Europe, Oceania, and the Americas. Although there have been enormous efforts, there is still no approved drug to treat ZIKV infection. Herein, we report the synthesis and biological evaluation of agents with noncompetitive mechanism of the ZIKV NS2B/NS3 protease inhibition through the binding to an allosteric site. Compounds 1 and 2 showed potent activity in both enzymatic and cellular assays. Derivative 1 efficiently reduced the ZIKV protein synthesis and the RNA replication and prevented the mice from life-threatening infection and the brain damage caused by ZIKV infection in a ZIKV mouse model.
- Published
- 2020
27. In This Issue, Volume 11, Issue 1
- Author
-
Giuseppe La Regina
- Subjects
Organic Chemistry ,Drug Discovery ,Biochemistry - Published
- 2020
28. New indolylarylsulfone non-nucleoside reverse transcriptase inhibitors show low nanomolar inhibition of single and double HIV-1 mutant strains
- Author
-
Elisabetta Cinquina, Emmanuele Crespan, Ombretta Turriziani, Myriam Catalano, Antonio Coluccia, Francesca Falasca, Roger Badia, Giuseppe La Regina, Cristina Limatola, Eva Riveira-Muñoz, Alessandro Brambilla, Romano Silvestri, Giovanni Maga, Marianna Nalli, Jorge I. Armijos Rivera, José A. Esté, and Domiziana Masci
- Subjects
Indoles ,Anti-HIV Agents ,Mutant ,Human immunodeficiency virus (HIV) ,Non-nucleoside reverse transcriptase inhibitor ,Indolylarylsulfone ,Microbial Sensitivity Tests ,Molecular Dynamics Simulation ,medicine.disease_cause ,01 natural sciences ,Nucleoside Reverse Transcriptase Inhibitor ,Cell Line ,03 medical and health sciences ,Structure-Activity Relationship ,Cell Line, Tumor ,Drug Discovery ,medicine ,Reverse transcriptase ,Humans ,Settore CHIM/08 - CHIMICA FARMACEUTICA ,Sulfones ,AIDS ,HIV-1 ,reverse transcriptase ,non-nucleoside reverse transcriptase inhibitor ,indolylarylsulfone ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Tumor ,Molecular Structure ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,virus diseases ,Drug Synergism ,General Medicine ,Virology ,HIV Reverse Transcriptase ,0104 chemical sciences ,Molecular Docking Simulation ,Drug Design ,Mutation ,Reverse Transcriptase Inhibitors ,Zidovudine ,Protein Binding - Abstract
We designed and synthesized 21 new indolylarylsulfones (IASs) as new HIV-1 NNRTIs. Among these, IAS 12 exhibited a remarkable antiviral activity against single and double mutants (K103N EC50 =
- Published
- 2020
29. Structure-activity relationship studies and in vitro and in vivo anticancer activity of novel 3-aroyl-1,4-diarylpyrroles against solid tumors and hematological malignancies
- Author
-
Giuseppe La Regina, Addolorata Coluccia, Carmela Mazzoccoli, Ruoli Bai, Claudia Martini, Cristina Mazzoni, Eleonora Da Pozzo, Romano Silvestri, Marianna Nalli, Michela Puxeddu, Te Liu, Hongliang Shen, Ernest Hamel, Chiara Cavallini, Viviana Orlando, Stefano Biagioni, and Antonio Coluccia
- Subjects
Injections, Subcutaneous ,Antineoplastic Agents ,Apoptosis ,01 natural sciences ,03 medical and health sciences ,Mice ,Structure-Activity Relationship ,In vivo ,pyrrole ,Drug Discovery ,medicine ,Tumor Cells, Cultured ,Structure–activity relationship ,Animals ,Humans ,Pyrroles ,030304 developmental biology ,Cell Proliferation ,Pharmacology ,0303 health sciences ,Mice, Inbred BALB C ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Cell growth ,Chemistry ,Brain Neoplasms ,Organic Chemistry ,Optical Imaging ,leukemia ,General Medicine ,In vitro ,0104 chemical sciences ,inhibitor ,Nilotinib ,tubulin ,Cell culture ,solid tumor ,Hematologic Neoplasms ,Clear cell carcinoma ,Cancer research ,Female ,Drug Screening Assays, Antitumor ,Glioblastoma ,Injections, Intraperitoneal ,medicine.drug - Abstract
Novel 3-aroyl-1,4-diarylpyrrole derivatives were synthesized to explore structure-activity relationships at the phenyls at positions 1 and 4 of the pyrrole. The presence of amino phenyl rings at positions 1 and 4 of the pyrrole ring were found to be a crucial requirement for potent antitumor activity. Several compounds strongly inhibited tubulin assembly through binding to the colchicine site. Compounds 42, 44, 48, 62 and 69 showed antitumor activity with low nanomolar IC50 values in several cancer cell lines. Compound 48 was generally more effective as an inhibitor of glioblastoma, colorectal and urinary bladder cancer cell lines; 69 consistently inhibited CML cell lines and demonstrated superiority in nilotinib and imatinib resistant LAMA84-R and KBM5-T315I cells. In animal models, compound 48 exhibited significant inhibition of the growth of T24 bladder carcinoma and ES-2 ovarian clear cell carcinoma tumors. Compounds 48 and 69 represent robust lead compounds for the design of new broad-spectrum anticancer agents active in different types of solid and hematological tumors.
- Published
- 2020
30. Sulfonamide inhibitors of beta-catenin signaling as anticancer agents with different output on c-Myc
- Author
-
Alessia Ciogli, Laura Di Magno, Romano Silvestri, Fiorella Di Pastena, Antonio Coluccia, Giuseppe La Regina, Simone Manetto, Marianna Nalli, Marella Maroder, Michela Puxeddu, and Gianluca Canettieri
- Subjects
Colorectal cancer ,Antineoplastic Agents ,anticancer ,01 natural sciences ,Biochemistry ,Proto-Oncogene Proteins c-myc ,chemistry.chemical_compound ,Drug Discovery ,sulfonamide ,beta-catenin ,medicine ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,IC50 ,Wnt Signaling Pathway ,beta Catenin ,Cell Proliferation ,Pharmacology ,Sulfonamides ,Trifluoromethyl ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Organic Chemistry ,Sulfonamide (medicine) ,Wnt signaling pathway ,Cancer ,medicine.disease ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Cell nucleus ,medicine.anatomical_structure ,chemistry ,Cancer research ,Molecular Medicine ,Signal transduction ,Drug Screening Assays, Antitumor ,medicine.drug - Abstract
The Wnt/β-catenin pathway is often found deregulated in cancer. The aberrant accumulation of β-catenin in the cell nucleus results in the development of various malignancies. Specific drugs against this signaling pathway for clinical treatments have not been approved yet. Herein we report inhibitors of β-catenin signaling of potential therapeutic value as anticancer agents. Ethyl 4-((4-(trifluoromethyl)phenyl)sulfonamido)benzoate (compound 14) inhibits the effect on Wnt reporter with an IC50 value of 7.0 μM, significantly reduces c-MYC levels, inhibits HCT116 colon cancer cell growth (IC50 20.2 μM), does not violate Lipinski and Veber rules, and shows predicted Caco-2 and MDCK cell permeability Papp >500 nm s-1 . Compound 14 seems to have potential for the development of new anticancer therapies.
- Published
- 2020
31. Discovery of new 1,1'-biphenyl-4-sulfonamides as selective subnanomolar human carbonic anhydrase II inhibitors
- Author
-
Michela Puxeddu, Romano Silvestri, Giuseppe La Regina, Marianna Nalli, Daniela Vullo, Alessio Nocentini, Claudiu T. Supuran, and Paola Gratteri
- Subjects
Biphenyl ,carbonic anhydrase ,inhibitor ,biphenyl ,subnanomolar ,glaucoma ,biology ,010405 organic chemistry ,Chemistry ,Carbonic anhydrase II ,Organic Chemistry ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,chemistry.chemical_compound ,Carbonic anhydrase ,Drug Discovery ,biology.protein - Abstract
[Image: see text] We report here the synthesis and human carbonic anhydrases (CA, EC 4.2.1.1) inhibitory properties of a series of 4′-substituted 1,1′-biphenyl-4-sulfonamides incorporating a 2″- or 3″-amino- or carboxyphenyl unit. Most compounds showed significant variations in their inhibition profiles against CA II and IX when compared to previously reported analogs 12–18 bearing a 4″-amino or a 4″-carboxy group. In particular, compounds 1–11 showed considerable improvement of the CA II inhibitory efficacy with K(I) values in the subnanomolar range (K(I)s spanning between 0.57 and 31.0 nM), a drop of activity against CA IX (K(I)s in the range 92.0 to 555.7 nM) and were as potent as 12–18 toward CA I (K(I)s in the range 5.9–217.7 nM). Docking and molecular dynamics were used to gain insights on the inhibition profiles. The reported inhibition data show that 1–11 have potential as novel agents to treat ocular pathologies, such as glaucoma, because of the potent and selective targeting of CA II, which is the isoform most implicated in this disease.
- Published
- 2020
32. New 6- and 7-heterocyclyl-1H-indole derivatives as potent tubulin assembly and cancer cell growth inhibitors
- Author
-
Giuseppe La Regina, Antonio Coluccia, Ruoli Bai, Carmela Mazzoccoli, Giulio Dondio, Ernest Hamel, Chiara Cavallini, Stefania Vultaggio, Romano Silvestri, Annalisa Verrico, Valentina Naccarato, Paola Rovella, Marianna Nalli, Eleonora Da Pozzo, Claudia Martini, Domiziana Masci, Valeria Famiglini, Ciro Mercurio, Mario Varasi, and Patrizia Lavia
- Subjects
0301 basic medicine ,Indoles ,Microtubule Tubulin Cancer cell Inhibitor Indole ,Cancer cell ,Drug Screening Assays ,Polymerization ,HeLa ,0302 clinical medicine ,Tubulin ,Drug Discovery ,Tumor Cells, Cultured ,Mic ,Cultured ,Molecular Structure ,biology ,Chemistry ,General Medicine ,Tubulin Modulators ,Tumor Cells ,inhibitor ,Biochemistry ,030220 oncology & carcinogenesis ,Indole ,Microtubule ,Cell Proliferation ,Cell Survival ,Dose-Response Relationship ,Structure-Activity Relationship ,Drug ,microtubule ,tubulin ,cancer cell, inhibitor, indole ,Antineoplastic Agents ,[object Object] ,03 medical and health sciences ,Humans ,Settore CHIM/08 - CHIMICA FARMACEUTICA ,Pharmacology ,Indole test ,Dose-Response Relationship, Drug ,Cell growth ,Organic Chemistry ,Antitumor ,biology.organism_classification ,030104 developmental biology ,Cell culture ,biology.protein ,Drug Screening Assays, Antitumor - Abstract
We designed new 3-arylthio- and 3-aroyl-1H-indole derivatives 3–22 bearing a heterocyclic ring at position 5, 6 or 7 of the indole nucleus. The 6- and 7-heterocyclyl-1H-indoles showed potent inhibition of tubulin polymerization, binding of colchicine to tubulin and growth of MCF-7 cancer cells. Compounds 13 and 19 inhibited a panel of cancer cells and the NCI/ADR-RES multidrug resistant cell line at low nanomolar concentrations. Compound 13 at 50 nM induced 77% G2/M in HeLa cells, and at 20 nM caused 50% stable arrest of mitosis. As an inhibitor of HepG2 cells (IC50 = 20 nM), 13 was 4-fold superior to 19. Compound 13 was a potent inhibitor of the human U87MG glioblastoma cells at nanomolar concentrations, being nearly one order of magnitude superior to previously reported arylthioindoles. The present results highlight 13 as a robust scaffold for the design of new anticancer agents.
- Published
- 2018
33. A Negative Allosteric Modulator of WNT Receptor Frizzled 4 Switches into an Allosteric Agonist
- Author
-
Giuseppe La Regina, Mariano Stornaiuolo, Antonella Accardo, Giovanni Battista Rossi, Sara Bottone, Nadia Badolati, Romano Silvestri, Ettore Novellino, Sara Passacantilli, Monica Dentice, Gennaro Riccio, Riccio, Gennaro, Bottone, Sara, Giuseppe La, Regina‡, Badolati, Nadia, Sara, Passacantilli‡, Giovanni Battista, Rossi§, Accardo, Antonella, Dentice, Monica, Silvestri, Romano, Novellino, Ettore, and Stornaiuolo, Mariano
- Subjects
0301 basic medicine ,Models, Molecular ,Frizzled ,Allosteric modulator ,Protein Conformation ,Allosteric regulation ,Primary Cell Culture ,Biochemistry ,Wnt-5a Protein ,Small Molecule Libraries ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,Structure-Activity Relationship ,Allosteric Regulation ,Heterotrimeric G protein ,Cell Line, Tumor ,Humans ,Computer Simulation ,Receptor ,Wnt Signaling Pathway ,Tissue homeostasis ,Chemistry ,Wnt signaling pathway ,Small molecule ,Heterotrimeric GTP-Binding Proteins ,Endocytosis ,Frizzled Receptors ,Recombinant Proteins ,Cell biology ,wnt receptor frizzled 4 ,allosteric agonist ,colon cancer cells ,030104 developmental biology ,HEK293 Cells ,Adenomatous Polyposis Coli ,Culture Media, Conditioned ,Neoplastic Stem Cells - Abstract
The WNT pathway interconnects a network of signaling events involved in a huge plethora of cellular processes, from organogenesis to tissue homeostasis. Despite its importance, the exiguity of organic drugs directly targeting the members of the Frizzled family of WNT receptors has hampered progress across the whole spectrum of biological fields in which the signaling is involved. We here present FzM1.8, a small molecule acting as an allosteric agonist of Frizzled receptor FZD4. FzM1.8 derives from FzM1, a negative allosteric modulator of the receptor. Replacement of FzM1 thiophene with a carboxylic moiety induces a molecular switch in the lead and transforms the molecule into an activator of WNT signaling. We here show that, in the absence of any WNT ligand, FzM1.8 binds to FZD4, promotes recruitment of heterotrimeric G proteins, and biases WNT signaling toward a noncanonical route that involves PI3K. Finally, in colon cancer cells, we prove that the FZD4/PI3K axis elicited by FzM1.8 preserves stemness and promotes proliferation of undifferentiated cells.
- Published
- 2018
34. Bax Activation Blocks Self-Renewal and Induces Apoptosis of Human Glioblastoma Stem Cells
- Author
-
Giuseppe La Regina, Luciana Marinelli, Barbara Costa, Chiara Giacomelli, Claudia Martini, Sabrina Taliani, Romano Silvestri, Simona Daniele, Federico Da Settimo, Ettore Novellino, Deborah Pietrobono, Mariateresa Giustiniano, Maria Letizia Trincavelli, Valeria La Pietra, Daniele, Simona, Pietrobono, Deborah, Costa, Barbara, Giustiniano, Mariateresa, LA PIETRA, Valeria, Giacomelli, Chiara, La Regina, Giuseppe, Silvestri, Romano, Taliani, Sabrina, Trincavelli, Maria Letizia, Da Settimo, Federico, Novellino, Ettore, Martini, Claudia, and Marinelli, Luciana
- Subjects
cancer stem cells ,0301 basic medicine ,cancer stem cell ,Cell Survival ,Physiology ,Cognitive Neuroscience ,Antineoplastic Agents ,Apoptosis ,Biology ,Bax activation ,Biochemistry ,Caspase 7 ,Central Nervous System Neoplasms ,03 medical and health sciences ,Bcl-2-associated X protein ,Bcl-2 family ,Cancer stem cell ,Cell Line, Tumor ,Temozolomide ,Humans ,glioblastoma ,pro-apoptotic proteins ,Cell Proliferation ,bcl-2-Associated X Protein ,Membrane Potential, Mitochondrial ,Caspase 3 ,Cell growth ,Cell Cycle ,Hydrazones ,Cell Biology ,General Medicine ,Cell cycle ,Cell biology ,Dacarbazine ,030104 developmental biology ,Neoplastic Stem Cells ,biology.protein ,Pyrazoles ,Drug Therapy, Combination ,Stem cell ,Glioblastoma - Abstract
Glioblastoma (GBM) is characterized by a poor response to conventional chemotherapeutic agents, attributed to the insurgence of drug resistance mechanisms and to the presence of a subpopulation of glioma stem cells (GSCs). GBM cells and GSCs present, among others, an overexpression of antiapoptotic proteins and an inhibition of pro-apoptotic ones, which help to escape apoptosis. Among pro-apoptotic inducers, the Bcl-2 family protein Bax has recently emerged as a promising new target in cancer therapy along with first BAX activators (BAM7, Compound 106, and SMBA1). Herein, a derivative of BAM-7, named BTC-8, was employed to explore the effects of Bax activation in different human GBM cells and in their stem cell subpopulation. BTC-8 inhibited GBM cell proliferation, arrested the cell cycle, and induced apoptosis through the induction of mitochondrial membrane permeabilization. Most importantly, BTC-8 blocked proliferation and self-renewal of GSCs and induced their apoptosis. Notably, BTC-8 was demonstrated to sensitize both GBM cells and GSCs to the alkylating agent Temozolomide. Overall, our findings shed light on the effects and the relative molecular mechanisms related to Bax activation in GBM, and they suggest Bax-targeting compounds as promising therapeutic tools against the GSC reservoir.
- Published
- 2017
35. Switching on the activity of 1,5-diaryl-pyrrole derivatives against drug-resistant ESKAPE bacteria: Structure-activity relationships and mode of action studies
- Author
-
Giuseppe La Regina, Romano Silvestri, Meir Touitou, Charlotte K. Hind, Anita Toscani, Antonio Coluccia, J. Mark Sutton, Xumin Wei, Mohammad Kaisarul Islam, Irene Conforti, Daniele Castagnolo, Melanie Clifford, Siham Memdouh, and Domiziana Masci
- Subjects
medicine.drug_class ,Cell Survival ,Antibiotics ,Drug Resistance ,Drug resistance ,Microbial Sensitivity Tests ,Antimicrobial resistance ,Pyrrole ,Gram-Positive Bacteria ,01 natural sciences ,DNA gyrase ,Microbiology ,Dose-Response Relationship ,03 medical and health sciences ,Structure-Activity Relationship ,Antibiotic resistance ,Levofloxacin ,Drug Discovery ,Drug Resistance, Bacterial ,Gram-Negative Bacteria ,medicine ,Settore CHIM/08 - CHIMICA FARMACEUTICA ,Humans ,Pyrroles ,antimicrobial resistance ,ESKAPE bacteria ,drug resistance ,pyrrole ,Mode of action ,030304 developmental biology ,Pharmacology ,0303 health sciences ,biology ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Wild type ,Bacterial ,General Medicine ,biology.organism_classification ,0104 chemical sciences ,Anti-Bacterial Agents ,Molecular Docking Simulation ,HEK293 Cells ,Drug ,Bacteria ,medicine.drug ,HeLa Cells - Abstract
Antibiotic resistance represents a major threat worldwide. Gram-positive and Gram-negative opportunistic pathogens are becoming resistant to all known drugs mainly because of the overuse and misuse of these medications and the lack of new antibiotic development by the pharmaceutical industry. There is an urgent need to discover structurally innovative antibacterial agents for which no pre-existing resistance is known. This work describes the identification, synthesis and biological evaluation of a novel series of 1,5-diphenylpyrrole compounds active against a panel of ESKAPE bacteria. The new compounds show high activity against both wild type and drug-resistant Gram + ve and Gram-ve pathogens at concentrations similar or lower than levofloxacin. Microbiology studies revealed that the plausible target of the pyrrole derivatives is the bacterial DNA gyrase, with the pyrrole derivatives displaying similar inhibitory activity to levofloxacin against the wild type enzyme and retaining activity against the fluoroquinolone-resistant enzyme.
- Published
- 2019
36. Small molecule inhibitors of KDM5 histone demethylases increase the radiosensitivity of breast cancer cells overexpressing Jarid1b
- Author
-
Antonio Coluccia, Romano Silvestri, Gianni Colotti, Adriano Mollica, Valerio Licursi, Luca Bombardi, Cecilia Mannironi, Simone Pippa, Rodolfo Negri, Enrico Cundari, Giuseppe La Regina, and Valentina Naccarato
- Subjects
Models, Molecular ,Jumonji Domain-Containing Histone Demethylases ,Radiation-Sensitizing Agents ,Pharmaceutical Science ,DNA damage ,breast cancer ,epigenetic drugs ,histone demethylase inhibitors ,Radiation Tolerance ,Analytical Chemistry ,Histones ,0302 clinical medicine ,small molecule, KDM5, JARID1B, cancer ,Drug Discovery ,KDM5 ,Cancer ,Histone Demethylases ,0303 health sciences ,Molecular Structure ,Chemistry ,Nuclear Proteins ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Chemistry (miscellaneous) ,030220 oncology & carcinogenesis ,MCF-7 Cells ,Molecular Medicine ,Female ,Cell Survival ,DNA repair ,small molecule ,Breast Neoplasms ,Article ,Small Molecule Libraries ,lcsh:QD241-441 ,03 medical and health sciences ,Breast cancer ,lcsh:Organic chemistry ,Cell Line, Tumor ,medicine ,Settore CHIM/08 - CHIMICA FARMACEUTICA ,Humans ,Radiosensitivity ,Physical and Theoretical Chemistry ,Clonogenic assay ,Cell Proliferation ,030304 developmental biology ,Organic Chemistry ,medicine.disease ,Repressor Proteins ,Comet assay ,JARID1B ,Small Molecules ,Drug Design ,Cancer research - Abstract
Background: KDM5 enzymes are H3K4 specific histone demethylases involved in transcriptional regulation and DNA repair. These proteins are overexpressed in different kinds of cancer, including breast, prostate and bladder carcinomas, with positive effects on cancer proliferation and chemoresistance. For these reasons, these enzymes are potential therapeutic targets. Methods: In the present study, we analyzed the effects of three different inhibitors of KDM5 enzymes in MCF-7 breast cancer cells over-expressing one of them, namely KDM5B/JARID1B. In particular we tested H3K4 demethylation (western blot), radio-sensitivity (cytoxicity and clonogenic assays) and damage accumulation (COMET assay and kinetics of H2AX phosphorylation). Results: we show that all three compounds with completely different chemical structures can selectively inhibit KDM5 enzymes and are capable of increasing sensitivity of breast cancer cells to ionizing radiation and radiation-induced damage. Conclusions: These findings confirm the involvement of H3K4 specific demethylases in the response to DNA damage, show a requirement of the catalytic function and suggest new strategies for the therapeutic use of their inhibitors.
- Published
- 2019
37. New indolylarylsulfones as highly potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors
- Author
-
Valeria Famiglini, Giuseppe La Regina, Roberto Cirilli, Andrea Brancale, Antonio Coluccia, Sveva Pelliccia, Giovanni Maga, Ettore Novellino, José A. Esté, Emmanuele Crespan, Roger Badia, Bonaventura Clotet, Romano Silvestri, Valeria, Famiglini, Giuseppe La Regina, Antonio, Coluccia, Pelliccia, Sveva, Andrea, Brancale, Giovanni, Maga, Emmanuele, Crespan, Roger, Badia, Bonaventura, Clotet, Est?, Jos? A., Roberto, Cirilli, Novellino, Ettore, and Romano, Silvestri
- Subjects
Models, Molecular ,RM ,Indoles ,Protein Conformation ,Mutant ,Human immunodeficiency virus (HIV) ,medicine.disease_cause ,Cell Line ,Nucleoside Reverse Transcriptase Inhibitor ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Broad spectrum ,Mutant strain ,Drug Discovery ,medicine ,Humans ,Sulfones ,QR355 ,Pharmacology ,Chemistry ,Organic Chemistry ,virus diseases ,General Medicine ,HIV Reverse Transcriptase ,Reverse transcriptase ,Biochemistry ,Docking (molecular) ,Drug Design ,QR180 ,HIV-1 ,Reverse Transcriptase Inhibitors ,Enantiomer - Abstract
New indolylarylsulfone HIV-1 NNRTIs were synthesized to evaluate unexplored substitutions of the benzyl/phenylethyl group linked at the indole-2-carboxamide. Against the NL4-3 HIV-1 WT strain, 17 out 20 compounds were superior to NVP and EFV. Several compounds inhibited the K103N HIV-1 mutant strain at nanomolar concentration and were superior to EFV. Some derivatives were superior to EFV against the Y181C and L100I HIV-1 mutant strains. Against the NL4-3 HIV-1 strain, the enantiomers 24 and 25 showed small differences of activity. In contrast, 24 turned out significantly more potent than 25 against the whole panel of mutant HIV-1 strains. The docking studies suggested that the difference in the observed inhibitory activities of 24 and 25 against the K03N mutation could be due to a kinetic rather than affinity differences.
- Published
- 2014
38. A New Case Manager for Diabetic Patients: A Pilot Observational Study of the Role of Community Pharmacists and Pharmacy Services in the Case Management of Diabetic Patients
- Author
-
Giuseppe La Regina, Dario Pandolfi, Micaela La Regina, Fulvio Glisenti, Raffaele La Regina, Paola Griggio, Lucio Beloni, and Nicola Stabile
- Subjects
medicine.medical_specialty ,Telemedicine ,health-system transformation ,pharmacist ,interprofessional cooperation ,Pharmacist ,lcsh:RS1-441 ,Context (language use) ,Pharmacy ,030226 pharmacology & pharmacy ,Article ,case management ,clinical pathway ,community pharmacy ,diabetes ,interprofessional care ,pharmacy services ,telemedicine ,lcsh:Pharmacy and materia medica ,03 medical and health sciences ,0302 clinical medicine ,Clinical pathway ,medicine ,Pharmacology (medical) ,030212 general & internal medicine ,General Pharmacology, Toxicology and Pharmaceutics ,Case report form ,health care economics and organizations ,Point of care ,business.industry ,Family medicine ,Observational study ,business - Abstract
The adherence of type 2 diabetes mellitus (DM2) patients with an individual care plan (ICP) is often not satisfactory, nor does it allow for a significant improvement in outcome, because of poor accessibility to services, poor integration of pathway articulations, poor reconciliation with the patient&rsquo, s life, or the lack of a constant reference person. The purpose of this study was to evaluate the contribution of community pharmacists and pharmacy services in improving adherence with periodic controls in DM2. The study was conducted at a rural pharmacy. A sample of 40 patients was calculated with respect to a historical cohort and subsequently enrolled. Clinical and personal data were collected in an electronic case report form. Pharmacists acting as a case manager followed patients carrying out their ICP developed by an attending physician. Some of the activities foreseen by the ICP, such as electrocardiogram, fundus examination, and self-analysis of blood and urine, were carried out directly in the pharmacy by the pharmacist through the use of telemedicine services and point of care units. Activities that could not be performed in the pharmacy were booked by the pharmacist at the accredited units. Examination results were electronically reported by the pharmacist to the attending physician. The primary endpoint was the variation in patient adherence with the ICP compared to a historical cohort. Secondary endpoints were variation in waiting time for the examinations, mean percentage change in glycated hemoglobin (HbA1c) and low-density lipoprotein (LDL) cholesterol levels and blood pressure, impact on healthcare-related costs, and perceived quality of care. Adherence to the ICP significantly increased. Waiting times were reduced and clinical outcomes improved with conceivable effects on costs. Patients appreciated the easier access to services. Community pharmacists and pharmacy services represent ideal actors and context that, integrated in the care network, can really favor ICP adherence and obtain daily morbidity reduction and cost savings through proper disease control and an early diagnosis of complications.
- Published
- 2020
39. Drug Design and Synthesis of First in Class PDZ1 Targeting NHERF1 Inhibitors as Anticancer Agents
- Author
-
Viviana Orlando, Marta Baiocchi, Maria Laura De Angelis, Romano Silvestri, Giuseppe La Regina, Stefano Biagioni, Candice Gautier, Valentina Naccarato, Gianluca Canettieri, Laura Di Magno, Antonio Coluccia, Stefano Gianni, Fiorella Di Pastena, Addolorata Coluccia, and Marianna Nalli
- Subjects
Drug ,Letter ,synthesis ,Colorectal cancer ,drug design ,media_common.quotation_subject ,PDZ domain ,01 natural sciences ,Biochemistry ,NHERF1 ,Drug Discovery ,medicine ,cancer ,media_common ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Wnt signaling pathway ,Cancer ,β-catenin ,Apoptotic death ,medicine.disease ,3. Good health ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Cancer research ,Phosphorylation - Abstract
Targeted approaches aiming at modulating NHERF1 activity, rather than its overall expression, would be preferred to preserve the normal functions of this versatile protein. We focused our attention on the NHERF1/PDZ1 domain that governs its membrane recruitment/displacement through a transient phosphorylation switch. We herein report the design and synthesis of novel NHERF1 PDZ1 domain inhibitors. These compounds have potential therapeutic value when used in combination with antagonists of β-catenin to augment apoptotic death of colorectal cancer cells refractory to currently available Wnt/β-catenin-targeted agents.
- Published
- 2018
40. Structure-Based Drug Design of Potent Pyrazole Derivatives against Rhinovirus Replication
- Author
-
Roberto Cirilli, Carmen Mirabelli, Romano Silvestri, Els Scheers, Giuseppe La Regina, Patrice Vanelle, Laurène Da Costa, Adriano Casulli, Thierry Terme, Antonio Coluccia, Carole Di Giorgio, Johan Neyts, Manon Roche, Université Grenoble Alpes [2016-2019] (UGA [2016-2019]), Institut de Chimie Radicalaire (ICR), Aix Marseille Université (AMU)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), Catholic University of Leuven - Katholieke Universiteit Leuven (KU Leuven), Dipartimento di Chimica e Tecnologie del Farmaco, Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome], Assistance Publique - Hôpitaux de Marseille (APHM), Institut méditerranéen de biodiversité et d'écologie marine et continentale (IMBE), Avignon Université (AU)-Aix Marseille Université (AMU)-Institut de recherche pour le développement [IRD] : UMR237-Centre National de la Recherche Scientifique (CNRS), Rega Institute for Medical Research [Leuven, België], Dipartimento del Farmaco, Istituto Superiore di Sanita [Rome], Department of Drug Chemistry and Technologies = Dipartimento di Chimica et Tecnologie del Farmaco [Roma], Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome]-Institut Pasteur, Fondation Cenci Bolognetti - Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP), Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome] (UNIROMA), Istituto Superiore di Sanità (ISS), Institut Pasteur, Fondation Cenci Bolognetti - Istituto Pasteur Italia, Fondazione Cenci Bolognetti, and Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome] (UNIROMA)
- Subjects
Male ,Models, Molecular ,0301 basic medicine ,Drug ,Rhinovirus infection ,Protein Conformation ,In vitro genotoxicity ,media_common.quotation_subject ,[SDV]Life Sciences [q-bio] ,RV ,Pyrazole ,Virus Replication ,medicine.disease_cause ,Antiviral Agents ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Discovery ,Enterovirus Infections ,medicine ,Animals ,Humans ,[CHIM]Chemical Sciences ,media_common ,VP1 protein ,heterocycles ,Micronucleus Tests ,Molecular Structure ,Chemistry ,molecular modelling studies ,rhinovirus ,pyrazole ,Virology ,Rats ,3. Good health ,capsid binder ,030104 developmental biology ,Drug Design ,Pyrazoles ,Molecular Medicine ,Structure based ,Rhinovirus ,chiral inhibitors ,HeLa Cells - Abstract
International audience; Rhinoviruses (RVs) have been linked to exacerbations of many pulmonary diseases, thus increasing morbidity and/or mortality in subjects at risk. Unfortunately, the wide variety of RV genotypes constitutes a major hindrance for the development of Rhinovirus replication inhibitors. In the current investigation, we have developed a novel series of pyrazole derivatives that potently inhibit the Rhinovirus replication. Compounds 10e and 10h behave as early stage inhibitors of Rhinovirus infection with a broad-spectrum activity against RV-A and RV-B species (EC 50 < 0.1 μM). We also evaluate the dynamics of the emerging resistance of these promising compounds and their in vitro genotoxicity. Molecular docking experiments shed light on the pharmacophoric elements interacting with residues of the drug-binding pocket.
- Published
- 2018
41. Towards modern anticancer agents that interact with tubulin
- Author
-
Antonio Coluccia, Giuseppe La Regina, Romano Silvestri, and Valentina Naccarato
- Subjects
Epothilones ,Vinca ,Pharmaceutical Science ,Antineoplastic Agents ,macromolecular substances ,02 engineering and technology ,030226 pharmacology & pharmacy ,microtubules ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Microtubule ,Tubulin ,Colchicine ,cancer ,tubulin interaction ,chemotherapeutics ,Animals ,Humans ,Cytotoxicity ,biology ,Chemistry ,021001 nanoscience & nanotechnology ,biology.organism_classification ,Tubulin Modulators ,Multiple drug resistance ,Biochemistry ,Tubulin Binding Agent ,biology.protein ,0210 nano-technology - Abstract
Tubulin is the primary target of an ever growing number of natural, semisynthetic and synthetic products as potential anticancer agents. The mechanisms of interaction of these molecules with tubulin are varied. These drug classes have shown to inhibit effectively several cancer types with IC50 from midmicromolar to low nanomolar concentrations. However, some limiting obstacles still remain, such as the development of multidrug resistance and cytotoxicity. We have reviewed recent advances in different classes of tubulin binding agents, including colchicine site agents, Vinca alkaloids, tryprostatins, moroidin, hemiasterlin, diazonamide, taxanes, epothilones and laulimalide.
- Published
- 2018
42. Small Molecule Inhibitors of KDM5 Histone Demethylases Increase Radio-Sensitivity of Breast Cancer Cells Over-Expressing JARID1B
- Author
-
Simone Pippa, Valentina Naccarato, Rodolfo Negri, Enrico Cundari, Romano Silvestri, Adriano Mollica, Luca Bombardi, Cecilia Mannironi, Valerio Licursi, Gianni Colotti, Giuseppe La Regina, and Antonio Coluccia
- Subjects
molecular_biology ,biology ,DNA damage ,Chemistry ,medicine.disease ,environment and public health ,Small molecule ,Radio sensitivity ,Breast cancer ,Cancer research ,biology.protein ,medicine ,Breast cancer cells ,Histone Demethylases ,JARID1B - Abstract
Background: KDM5 enzymes are H3K4 specific histone demethylases involved in transcriptional regulation and DNA repair. These proteins are over-expressed in different kinds of cancer, including breast, prostate and bladder carcinoma, with positive effects on cancer proliferation and chemo-resistance. For these reasons, these enzymes are potential therapeutic cancer targets. Methods: In the present study, we analyzed the effects of three different inhibitors of KDM5 enzymes in MCF-7 breast cancer cells over-expressing JARID1B. In particular we tested H3K4 demethylation (western blot); target gene transcription (RNAseq and real time PCR); radio-sensitivity (citoxicity and clonogenic assays) and damage accumulation (kinetics of H2AX phosphorylation). Results: we show that two compounds with completely different chemical structure can selectively inhibit KDM5 enzymes and that both compounds are capable of increasing sensitivity of breast cancer cells to ionizing radiation and H2AX phosphorylation. Conclusions: These findings confirm the involvement of H3K4 specific demethylases in DNA damage signaling and repair and suggest new strategies for the therapeutic use of their inhibitors.
- Published
- 2018
43. β-catenin knockdown promotes nherf1-mediated survival of colorectal cancer cells: implications for a double-targeted therapy
- Author
-
Michel Salzet, Valeria Famiglini, Concetta Saponaro, Luca Mologni, Giuseppe La Regina, Sara Sergio, Michele Maffia, Romano Silvestri, Candice Gautier, Valentina Naccarato, Addolorata Coluccia, Stefano Gianni, Antonio Coluccia, Carlo Gambacorti Passerini, Daniele Vergara, Maria De Luca, Cecilia Bucci, Isabelle Fournier, Daniela Bonetti, Istituto Tumori 'Giovanni Paolo II' [Bari], Liceo Classico Giovanni Paolo II [Lecce LE, Italie], University of Salento [Lecce], Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome], Laboratorio Pasteur [Istituto Pasteur-Fondazione Cenci Bolognetti, Rome], Institut Pasteur, Fondation Cenci Bolognetti - Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP), Università degli Studi di Milano-Bicocca [Milano] (UNIMIB), Protéomique, Réponse Inflammatoire, Spectrométrie de Masse (PRISM) - U 1192 (PRISM), Université de Lille-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome] (UNIROMA), Università degli Studi di Milano-Bicocca = University of Milano-Bicocca (UNIMIB), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), SALZET, Michel, Saponaro, C, Sergio, S, Coluccia, A, De Luca, M, La Regina, G, Mologni, L, Famiglini, V, Naccarato, V, Bonetti, D, Gautier, C, Gianni, S, Vergara, D, Salzet, M, Fournier, I, Bucci, C, Silvestri, R, Passerini, C, Maffia, M, Passerini, Cg, and Coluccia, Aml
- Subjects
0301 basic medicine ,Cancer Research ,Sodium-Hydrogen Exchangers ,Cell Survival ,[SDV]Life Sciences [q-bio] ,beta-catenin ,nherf1 ,colorectal cancer cells ,Apoptosis ,Biology ,medicine.disease_cause ,Small hairpin RNA ,03 medical and health sciences ,Transcription Factor 4 ,Genetic ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Genetics ,Gene Knockdown Techniques ,medicine ,Humans ,Molecular Biology ,beta Catenin ,Gene knockdown ,Sulfonamides ,Wnt signaling pathway ,Phosphoproteins ,3. Good health ,Gene Expression Regulation, Neoplastic ,[SDV] Life Sciences [q-bio] ,Protein Transport ,030104 developmental biology ,Catenin ,Mutation ,Cancer research ,KRAS ,Carcinogenesis ,Colorectal Neoplasms ,Chromatin immunoprecipitation - Abstract
International audience; Nuclear activated β-catenin plays a causative role in colorectal cancers (CRC) but remains an elusive therapeutic target. Using human CRC cells harboring different Wnt/β-catenin pathway mutations in APC/KRAS or β-catenin/KRAS genes, and both genetic and pharmacological knockdown approaches, we show that oncogenic β-catenin signaling negatively regulates the expression of NHERF1 (Na+/H+ exchanger 3 regulating factor 1), a PDZ-adaptor protein that is usually lost or downregulated in early dysplastic adenomas to exacerbate nuclear β-catenin activity. Chromatin immunoprecipitation (ChIP) assays demonstrated that β-catenin represses NHERF1 via TCF4 directly, while the association between TCF1 and the Nherf1 promoter increased upon β-catenin knockdown. To note, the occurrence of a cytostatic survival response in settings of single β-catenin-depleted CRC cells was abrogated by combining NHERF1 inhibition via small hairpin RNA (shRNA) or RS5517, a novel PDZ1-domain ligand of NHERF1 that prevented its ectopic nuclear entry. Mechanistically, dual NHERF1/β-catenin targeting promoted an autophagy-to-apoptosis switch consistent with the activation of Caspase-3, the cleavage of PARP and reduced levels of phospho-ERK1/2, Beclin-1, and Rab7 autophagic proteins compared with β-catenin knockdown alone. Collectively, our data unveil novel β-catenin/TCF-dependent mechanisms of CRC carcinogenesis, also offering preclinical proof of concept for combining β-catenin and NHERF1 pharmacological inhibitors as a mechanism-based strategy to augment apoptotic death of CRC cells refractory to current Wnt/β-catenin-targeted therapeutics.
- Published
- 2018
44. Discovery of 1,1′-Biphenyl-4-sulfonamides as a New Class of Potent and Selective Carbonic Anhydrase XIV Inhibitors
- Author
-
Peiwen Pan, Giuseppe La Regina, Elisa Nuti, Ludovica Monti, Vincenzo Alterio, Antonio Coluccia, Sveva Pelliccia, Claudiu T. Supuran, Romano Silvestri, Seppo Parkkila, Simona Maria Monti, Valeria Famiglini, Giuseppina De Simone, Armando Rossello, Daniela Vullo, La Regina, G., Coluccia, A., Famiglini, V., Pelliccia, S., Monti, L., Vullo, D., Nuti, E., Alterio, V., De Simone, G., Monti, S. M., Pan, P., Parkkila, S., Supuran, C. T., Rossello, A., and Silvestri, R.
- Subjects
Models, Molecular ,Carbonic Anhydrase Inhibitor ,Molecular model ,drug design ,Stereochemistry ,Crystallography, X-Ray ,Adduct ,Carbonic Anhydrase ,Structure-Activity Relationship ,Carbonic anhydrase ,Drug Discovery ,medicine ,Humans ,Structure–activity relationship ,Carbonic Anhydrase Inhibitors ,cristallography ,Carbonic Anhydrases ,chemistry.chemical_classification ,activators ,Sulfonamides ,biology ,IX ,targets ,Biphenyl Compounds ,Lyase ,Biphenyl compound ,Enzyme ,chemistry ,Biochemistry ,Biphenyl Compound ,biology.protein ,Molecular Medicine ,Acetazolamide ,Human ,medicine.drug - Abstract
New 1,1'-biphenylsulfonamides were synthesized and evaluated as inhibitors of the ubiquitous human carbonic anhydrase isoforms I, II, IX, XII, and XIV using acetazolamide (AAZ) as reference compound. The sulfonamides 1-21 inhibited all the isoforms, with Ki values in the nanomolar range of concentration, and were superior to AAZ against all of them. X-ray crystallography and molecular modeling studies on the adducts that compound 20, the most potent hCA XIV inhibitor of the series (Ki = 0.26 nM), formed with the five hCAs, provided insight into the molecular determinants responsible for the high affinity of this molecule toward the target enzymes. The results pave the way to the development of 1.1'-biphenylsulfonamides as a new class of highy potent hCA XIV inhibitors.
- Published
- 2015
45. Pharmacological folding chaperones act as allosteric ligands of Frizzled4
- Author
-
Agostino Bruno, Salvatore Di Maro, Serena F Generoso, Mariano Stornaiuolo, Mariateresa Giustiniano, Massimo Mallardo, Ettore Novellino, Daniela Sarnataro, Luciana Marinelli, Giuseppe La Regina, Sara Passacantilli, Sara Bottone, Romano Silvestri, Stefano Bonatti, Monica Dentice, Hilde Cassese, Generoso, Serena F, Giustiniano, Mariateresa, La Regina, Giuseppe, Bottone, Sara, Passacantilli, Sara, DI MARO, Salvatore, Cassese, Hilde, Bruno, Agostino, Mallardo, Massimo, Dentice, Monica, Silvestri, Romano, Marinelli, Luciana, Sarnataro, Daniela, Bonatti, Stefano, Novellino, Ettore, and Stornaiuolo, Mariano
- Subjects
Glycerol ,Molecular Chaperone ,Protein Folding ,Chemistry, Pharmaceutical ,Amino Acid Motifs ,Plasma protein binding ,Protein aggregation ,Ligands ,HeLa Cell ,cell-surface expression ,Mutagenesi ,Receptors, G-Protein-Coupled ,HEK293 Cell ,drosophila-melanogaster ,Effector ,Medicine (all) ,Frizzled Receptor ,Cell biology ,Co-chaperone ,Amino Acid Motif ,Protein folding ,dishevelled dep domain ,Allosteric Site ,Human ,Protein Binding ,signaling pathway ,Molecular Sequence Data ,Allosteric regulation ,Ligand ,Biology ,Cell Line, Tumor ,Humans ,Molecular Biology ,G protein-coupled receptor ,Base Sequence ,Dose-Response Relationship, Drug ,colorectal-cancer ,endoplasmic-reticulum ,structural basis ,Cell Biology ,Frizzled Receptors ,HEK293 Cells ,ephrogenic diabetes-insipidus ,Microscopy, Fluorescence ,Structural biology ,Mutagenesis ,Drug Design ,mass-spectrometry data ,receptor mutants ,HeLa Cells ,Molecular Chaperones - Abstract
Upon binding, ligands can chaperone their protein targets by preventing them from misfolding and aggregating. Thus, an organic molecule that works as folding chaperone for a protein might be its specific ligand, and, similarly, the chaperone potential could represent an alternative readout in a molecular screening campaign toward the identification of new hits. Here we show that small molecules selected for acting as pharmacological chaperones on a misfolded mutant of the Frizzled4 (Fz4) receptor bind and modulate wild-type Fz4, representing what are to our knowledge the first organic ligands of this until-now-undruggable GPCR. The novelty and the advantages of the screening platform, the allosteric binding site addressed by these new ligands and the mechanism they use to modulate Fz4 suggest new avenues for development of inhibitors of the Wnt-beta-catenin pathway and for drug discovery.
- Published
- 2015
46. Chiral Indolylarylsulfone Non-Nucleoside Reverse Transcriptase Inhibitors as New Potent and Broad Spectrum Anti-HIV-1 Agents
- Author
-
Francesca Romana Formica, Giuseppe La Regina, Roberto Cirilli, José A. Esté, Domiziana Masci, Alessandro Brambilla, Andrea Brancale, Emmanuele Crespan, Antonio Coluccia, Roger Badia, Eva Riveira-Muñoz, Myriam Catalano, Ettore Novellino, Romano Silvestri, Valeria Famiglini, Giovanni Maga, and Cristina Limatola
- Subjects
0301 basic medicine ,Lipopolysaccharides ,Efavirenz ,Indoles ,Stereochemistry ,Anti-HIV Agents ,Mutant ,Glutamic Acid ,Stereoisomerism ,Drug design ,RS ,Nucleoside Reverse Transcriptase Inhibitor ,03 medical and health sciences ,chemistry.chemical_compound ,Structure-Activity Relationship ,Non-Nucleoside Reverse Transcriptase Inhibitors ,chiral indolyarylsulfones ,non-nucleoside reverse transcriptase inhibitors ,drug design and discovery ,Drug Discovery ,Reverse transcriptase ,Structure–activity relationship ,Animals ,Humans ,Settore CHIM/08 - CHIMICA FARMACEUTICA ,Sulfones ,Cells, Cultured ,Alanine ,Neurons ,Molecular Structure ,Chemistry ,Glutamate receptor ,virus diseases ,Mice, Inbred C57BL ,AIDS ,Molecular Docking Simulation ,Chiral Indolylarylsulfones ,030104 developmental biology ,Neuroprotective Agents ,Mutation ,HIV-1 ,Molecular Medicine ,Reverse Transcriptase Inhibitors ,Microglia ,Enantiomer - Abstract
We designed and synthesized a series of chiral indolyarylsulfones (IASs) as new HIV-1 NNRTIs. The new IASs 8-37 showed potent inhibition of the HIV-1 WT NL4-3 strain and of the mutant K103N, Y181C, Y188L, and K103N-Y181C HIV-1 strains. Six racemic mixtures, 8, 23-25, 31, and 33, were separated at semipreparative level into their pure enantiomers. The (R)-8 enantiomer bearing the chiral (alpha-methylbenzyl) was superior to the (S)-counterpart. IAS derivatives bearing the (5) alanine unit, (S)-23, (S,R)-25, (S)-31, and (S)-33, were remarkably more potent than the corresponding (R)-enantiomers. Compound 23 protected hippocampal neuronal cells from the excitotoxic insult, while efavirenz (EFV) did not contrast the neurotoxic effect of glutamate. The present results highlight the chiral IASs as new NNRTIs with improved resistance profile against the mutant HIV-1 strains and reduced neurotoxic effects.
- Published
- 2017
47. Computer-Aided Identification and Lead Optimization of Dual Murine Double Minute 2 and 4 Binders: Structure-Activity Relationship Studies and Pharmacological Activity
- Author
-
Giuseppe La Regina, Claudio Luchinat, Sandro Cosconati, Linda Cerofolini, Diego Brancaccio, Mariateresa Giustiniano, Federico Da Settimo, Stefano Giuntini, Luciana Marinelli, Ettore Novellino, Anna Messere, Sveva Pelliccia, Simona Daniele, Claudia Martini, Deborah Pietrobono, Marco Fragai, Valeria La Pietra, Sabrina Taliani, Romano Silvestri, Giustiniano, Mariateresa, Daniele, Simona, Pelliccia, Sveva, La Pietra, Valeria, Pietrobono, Deborah, Brancaccio, Diego, Cosconati, Sandro, Messere, Anna, Giuntini, Stefano, Cerofolini, Linda, Fragai, Marco, Luchinat, Claudio, Taliani, Sabrina, La Regina, Giuseppe, Da Settimo, Federico, Silvestri, Romano, Martini, Claudia, Novellino, Ettore, and Marinelli, Luciana
- Subjects
0301 basic medicine ,High-Throughput Screening Assay ,Models, Molecular ,Magnetic Resonance Spectroscopy ,Antineoplastic Agents ,Apoptosis ,neuroblastoma cells ,Antineoplastic Agent ,03 medical and health sciences ,Structure-Activity Relationship ,0302 clinical medicine ,Proto-Oncogene Proteins ,Cell Line, Tumor ,Drug Discovery ,Structure–activity relationship ,Humans ,Receptor ,p53 protein ,Cell Proliferation ,Virtual screening ,MDM2/MDM4 binders ,Proto-Oncogene Protein ,biology ,Cell growth ,Chemistry ,Drug Discovery3003 Pharmaceutical Science ,Apoptosi ,Biological activity ,Proto-Oncogene Proteins c-mdm2 ,Genes, p53 ,High-Throughput Screening Assays ,030104 developmental biology ,Biochemistry ,Docking (molecular) ,Cell culture ,030220 oncology & carcinogenesis ,Drug Design ,biology.protein ,Neoplastic Stem Cells ,Mdm2 ,Computer-Aided Design ,Molecular Medicine ,Neoplastic Stem Cell ,Human - Abstract
The function of p53 protein, also known as "genome guardian", might be impaired by the overexpression of its primary cellular inhibitor, the murine double minute 2 protein (MDM2). However, the recent finding that MDM2-selective inhibitors induce high levels of its homologue MDM4, prompt us to identify, through a receptor-based virtual screening on an in house database, dual MDM2/MDM4 binders. Compound 1 turned out to possess an IC50 of 93.7 and of 4.6 nM on MDM2 and MDM4, respectively. A series of compounds were synthesized to optimize its activity on MDM2. As a result, compound 12 showed low nanomolar IC50 for both targets. NMR studies confirmed the pocket of binding of 12 as predicted by the Glide docking software. Notably, 12 was able to cause concentration-dependent inhibition of cell proliferation, yielding an IC50 value of 356 ± 21 nM in neuroblastoma SHSY5Y cells and proved even to efficiently block cancer stem cell growth. The function of p53 protein, also known as "genome guardian", might be impaired by the overexpression of its primary cellular inhibitor, the murine double minute 2 protein (MDM2). However, the recent finding that MDM2-selective inhibitors induce high levels of its homologue MDM4, prompt us to identify, through a receptor-based virtual screening on an in house database, dual MDM2/MDM4 binders. Compound 1 turned out to possess an IC50 of 93.7 and of 4.6 nM on MDM2 and MDM4, respectively. A series of compounds were synthesized to optimize its activity on MDM2. As a result, compound 12 showed low nanomolar IC50 for both targets. NMR studies confirmed the pocket of binding of 12 as predicted by the Glide docking software. Notably, 12 was able to cause concentration-dependent inhibition of cell proliferation, yielding an IC50 value of 356 ± 21 nM in neuroblastoma SHSY5Y cells and proved even to efficiently block cancer stem cell growth.
- Published
- 2017
48. Inhibition of dengue virus replication by novel inhibitors of RNA-dependent RNA polymerase and protease activities
- Author
-
Giuseppe La Regina, Sveva Pelliccia, John Hiscott, Yu-Hsuan Wu, Antonio Coluccia, Jin-Ching Lee, Valeria Famiglini, Romano Silvestri, Chin-Kai Tseng, Domiziana Masci, Pelliccia, S., Wu, Y. -H., Coluccia, A., La Regina, G., Tseng, C. -K., Famiglini, V., Masci, D., Hiscott, J., Lee, J. -C., and Silvestri, R.
- Subjects
0301 basic medicine ,Models, Molecular ,RdRp ,medicine.medical_treatment ,viruses ,synergy ,Dengue virus ,medicine.disease_cause ,Virus Replication ,Dengue Viru ,Dengue fever ,Dengue ,chemistry.chemical_compound ,Mice ,Models ,RNA polymerase ,Drug Discovery ,Enzyme Inhibitor ,Enzyme Inhibitors ,chemistry.chemical_classification ,ICR-suckling mouse ,Tumor ,Molecular Structure ,Microbial Sensitivity Test ,Serine Endopeptidases ,DENV inhibitors ,rdrp ,NS3 protease ,icr-suckling mouse ,virus diseases ,General Medicine ,Serine Endopeptidase ,DENV inhibitor ,Drug ,Research Paper ,Human ,Cell Survival ,030106 microbiology ,RNA-dependent RNA polymerase ,Microbial Sensitivity Tests ,Biology ,Antiviral Agents ,Cell Line ,Dose-Response Relationship ,03 medical and health sciences ,Structure-Activity Relationship ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Settore CHIM/08 - CHIMICA FARMACEUTICA ,Antiviral Agent ,Pharmacology ,NS3 ,Protease ,Dose-Response Relationship, Drug ,Animal ,lcsh:RM1-950 ,Molecular ,biochemical phenomena, metabolism, and nutrition ,Dengue Virus ,medicine.disease ,RNA-Dependent RNA Polymerase ,Virology ,030104 developmental biology ,Enzyme ,lcsh:Therapeutics. Pharmacology ,chemistry ,Viral replication - Abstract
Dengue virus (DENV) is the leading mosquito-transmitted viral infection in the world. With more than 390 million new infections annually, and up to 1 million clinical cases with severe disease manifestations, there continues to be a need to develop new antiviral agents against dengue infection. In addition, there is no approved anti-DENV agents for treating DENV-infected patients. In the present study, we identified new compounds with anti-DENV replication activity by targeting viral replication enzymes – NS5, RNA-dependent RNA polymerase (RdRp) and NS3 protease, using cell-based reporter assay. Subsequently, we performed an enzyme-based assay to clarify the action of these compounds against DENV RdRp or NS3 protease activity. Moreover, these compounds exhibited anti-DENV activity in vivo in the ICR-suckling DENV-infected mouse model. Combination drug treatment exhibited a synergistic inhibition of DENV replication. These results describe novel prototypical small anti-DENV molecules for further development through compound modification and provide potential antivirals for treating DENV infection and DENV-related diseases.
- Published
- 2017
49. 3-Aroyl-1,4-diarylpyrroles inhibit chronic myeloid leukemia cell growth through an interaction with tubulin
- Author
-
Vitalba Ruggieri, Romano Silvestri, Michele Maffia, Giorgio Ortar, Mario Varasi, Addolorata Coluccia, Carmela Mazzoccoli, Marianna Nalli, Sara Passacantilli, Antonio Coluccia, Concetta Saponaro, Ciro Mercurio, Tiziana Tataranni, Stefania Vultaggio, Ruoli Bai, Andrea Brancale, Valeria Famiglini, Giuseppe La Regina, Sara Sergio, Ernest Hamel, Valentina Naccarato, Claudia Piccoli, and Francesca Agriesti
- Subjects
0301 basic medicine ,synthesis ,medicine.drug_class ,Biology ,Pharmacology ,Biochemistry ,Tyrosine-kinase inhibitor ,RS ,03 medical and health sciences ,0302 clinical medicine ,chronic myeloid leukemia ,hemic and lymphatic diseases ,Drug Discovery ,3-aroyl-1 ,medicine ,Cytotoxic T cell ,cancer ,3-aroyl-1,4-diarylpyrrole ,tubulin ,Cell growth ,Organic Chemistry ,Myeloid leukemia ,Haematopoiesis ,030104 developmental biology ,Tubulin ,Imatinib mesylate ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,4-diarylpyrrole - Abstract
We designed 3-aroyl-1,4-diarylpyrrole (ARDAP) derivatives as potential anticancer agents having different\ud substituents at the 1- or 4-phenyl ring. ARDAP compounds exhibited potent inhibition of tubulin polymerization, binding of\ud colchicine to tubulin, and cancer cell growth. ARDAP derivative 10 inhibited the proliferation of BCR/ABL-expressing KU812\ud and LAMA84 cells from chronic myeloid leukemia (CML) patients in blast crisis and of hematopoietic cells ectopically\ud expressing the imatinib mesylate (IM)-sensitive KBM5-WT or its IM-resistant KBM5-T315I mutation. Compound 10 minimally\ud affected the proliferation of normal blood cells, indicating that it may be a promising agent to overcome broad tyrosine kinase\ud inhibitor resistance in relapsed/refractory CML patients. Compound 10 significantly decreased CML proliferation by inducing\ud G2/M phase arrest and apoptosis via a mitochondria-dependent pathway. ARDAP 10 augmented the cytotoxic effects of IM in\ud human CML cells. Compound 10 represents a robust lead compound to develop tubulin inhibitors with potential as novel treatments for CML.
- Published
- 2017
50. Mitotic cell death induction by targeting the mitotic spindle with tubulin-inhibitory indole derivative molecules
- Author
-
Erica Di Cesare, Antonio Coluccia, Romano Silvestri, Patrizia Lavia, Annalisa Verrico, Andrea Miele, Maria Giubettini, Paola Rovella, Enrico Cundari, Valeria Famiglini, and Giuseppe La Regina
- Subjects
Models, Molecular ,0301 basic medicine ,caspase-3 ,Indoles ,Time Factors ,Cell cycle checkpoint ,Cell Survival ,Blotting, Western ,Mitosis ,Apoptosis ,time-lapse imaging ,Spindle Apparatus ,macromolecular substances ,Cell fate determination ,tubulin inhibitors ,03 medical and health sciences ,0302 clinical medicine ,Tubulin ,Microtubule ,Cell Line, Tumor ,Humans ,mitotic spindle microtubules ,Cell Death ,Molecular Structure ,biology ,Tubulin Modulators ,mitotic cell death ,Cell Cycle Checkpoints ,mitotic spindle micortubules ,Spindle apparatus ,Cell biology ,030104 developmental biology ,Microscopy, Fluorescence ,Oncology ,Caspases ,030220 oncology & carcinogenesis ,Cancer cell ,MCF-7 Cells ,biology.protein ,HT29 Cells ,Research Paper ,HeLa Cells ,Protein Binding - Abstract
// Erica Di Cesare 1 , Annalisa Verrico 1 , Andrea Miele 1, 2 , Maria Giubettini 1, 3 , Paola Rovella 1 , Antonio Coluccia 4 , Valeria Famiglini 4 , Giuseppe La Regina 4 , Enrico Cundari 1 , Romano Silvestri 4 , Patrizia Lavia 1 1 Institute of Molecular Biology and Pathology, CNR National Research Council, c/o Department of Biology and Biotechnology, Sapienza Universita di Roma, Roma, Italy 2 Present address: IRBM Science Park, Advent Srl, Pomezia, Italy 3 Present address: EMBL, Heidelberg, Germany 4 Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technology, Sapienza Universita di Roma, Roma, Italy Correspondence to: Patrizia Lavia, email: patrizia.lavia@uniroma1.it Keywords: mitotic spindle microtubules, tubulin inhibitors, mitotic cell death, caspase-3, time-lapse imaging Received: April 13, 2016 Accepted: January 06, 2017 Published: February 01, 2017 ABSTRACT Tubulin-targeting molecules are widely used cancer therapeutic agents. They inhibit microtubule-based structures, including the mitotic spindle, ultimately preventing cell division. The final fates of microtubule-inhibited cells are however often heterogeneous and difficult to predict. While recent work has provided insight into the cell response to inhibitors of microtubule dynamics (taxanes), the cell response to tubulin polymerization inhibitors remains less well characterized. Arylthioindoles (ATIs) are recently developed tubulin inhibitors. We previously identified ATI members that effectively inhibit tubulin polymerization in vitro and cancer cell growth in bulk cell viability assays. Here we characterise in depth the response of cancer cell lines to five selected ATIs. We find that all ATIs arrest mitotic progression, yet subsequently yield distinct cell fate profiles in time-lapse recording assays, indicating that molecules endowed with similar tubulin polymerization inhibitory activity in vitro can in fact display differential efficacy in living cells. Individual ATIs induce cytological phenotypes of increasing severity in terms of damage to the mitotic apparatus. That differentially triggers MCL-1 down-regulation and caspase-3 activation, and underlies the terminal fate of treated cells. Collectively, these results contribute to define the cell response to tubulin inhibitors and pinpoint potentially valuable molecules that can increase the molecular diversity of tubulin-targeting agents.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.