1
|
Federico LB, Silva GM, de Fraga Dias A, Figueiró F, Battastini AMO, Dos Santos CBR, Costa LT, Rosa JMC, de Paula da Silva CHT. Identification of novel αβ-tubulin modulators with antiproliferative activity directed to cancer therapy using ligand and structure-based virtual screening. Int J Biol Macromol 2020; 165:3040-3050. [PMID: 33736292 DOI: 10.1016/j.ijbiomac.2020.10.136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/10/2020] [Accepted: 10/15/2020] [Indexed: 11/19/2022]
Abstract
Among several strategies related to cancer therapy targeting the modulation of αβ-tubulin has shown encouraging findings, more specifically when this is achieved by inhibitors located at the colchicine binding site. In this work, we aim to fish new αβ-tubulin modulators through a diverse and rational VS study, and thus, exhibiting the development of two VS pipelines. This allowed us to identify two compounds 5 and 9 that showed IC50 values of 19.69 and 21.97 μM, respectively, towards possible modulation of αβ-tubulin, such as assessed by in vitro assays in C6 glioma and HEPG2 cell lines. We also evaluated possible mechanisms of action of obtained hits towards the colchicine binding site of αβ-tubulin by using docking approaches. In addition, assessment of the stability of the active (5 and 9) and inactive compounds (3 and 13) within the colchicine binding site was carried out by molecular dynamics (MD) simulations, highlighting the solvent effect and revealing the compound 5 as the most stable in the complex. At last, deep analysis of these results provided some valuable insights on the importance of using mixed ligand- and structure-based strategies in VS campaigns, in order to achieve higher chemical diversity and biological effect as well.
Collapse
Affiliation(s)
- Leonardo Bruno Federico
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil.
| | - Guilherme Martins Silva
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901 Ribeirão Preto, SP, Brazil
| | - Amanda de Fraga Dias
- Graduate Program in Biological Sciences: Biochemistry, Institute of Health Sciences, Federal University of Rio Grande do Sul, Av. Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS 90035-003, Brazil
| | - Fabrício Figueiró
- Graduate Program in Biological Sciences: Biochemistry, Institute of Health Sciences, Federal University of Rio Grande do Sul, Av. Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS 90035-003, Brazil; Department of Biochemistry, Institute of Health Sciences, Federal University of Rio Grande do Sul, Av. Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS 90035-003, Brazil
| | - Ana Maria Oliveira Battastini
- Graduate Program in Biological Sciences: Biochemistry, Institute of Health Sciences, Federal University of Rio Grande do Sul, Av. Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS 90035-003, Brazil
| | - Cleydson Breno Rodrigues Dos Santos
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá, AP 68902-280, Brazil
| | - Luciano T Costa
- MolMod-CS, Institute of Chemistry, Federal Fluminense University, Outeiro de São João Batista, Niterói, Rio de Janeiro, Brazil
| | - Joaquín Maria Carmpos Rosa
- Department of Pharmaceutical and Organic Chemistry, Faculty of Pharmacy, Institute of Biosanitary Research ibs. GRANADA, University of Granada, 18071, Spain
| | - Carlos Henrique Tomich de Paula da Silva
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil; Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901 Ribeirão Preto, SP, Brazil
| |
Collapse
|
2
|
Pharmacology and in vivo efficacy of pyridine-pyrimidine amides that inhibit microtubule polymerization. Bioorg Med Chem Lett 2018; 28:934-941. [PMID: 29433928 DOI: 10.1016/j.bmcl.2018.01.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/23/2018] [Accepted: 01/25/2018] [Indexed: 10/18/2022]
Abstract
Microtubule-targeting agents are important tools in cancer treatment. Generating novel microtubule targeting agents with novel pharmacology could dramatically expand the utility of this class of drugs. Here we characterize the pharmacology of recently described small molecule microtubule polymerization inhibitors. Pharmacokinetic experiments show oral bioavailability through gastric absorption. In vitro assays designed to predict absorption, distribution, metabolism, and excretion (ADME) and safety reveal a scaffold that is metabolically stable, evades P-glycoprotein, does not inhibit CYP enzymes, occurs as a significant free fraction in serum, and has exceptionally high cellular permeability. Together with in vivo efficacy models, pharmacology supports further development as a treatment for solid tumors.
Collapse
|
3
|
Voitovich YV, Shegravina ES, Sitnikov NS, Faerman VI, Fokin VV, Schmalz HG, Combes S, Allegro D, Barbier P, Beletskaya IP, Svirshchevskaya EV, Fedorov AY. Synthesis and Biological Evaluation of Furanoallocolchicinoids. J Med Chem 2014; 58:692-704. [DOI: 10.1021/jm501678w] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Yuliya V. Voitovich
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| | - Ekaterina S. Shegravina
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| | - Nikolay S. Sitnikov
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| | - Vladimir I. Faerman
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| | - Valery V. Fokin
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| | - Hans-Gunther Schmalz
- Department
of Chemistry, University of Cologne, Greinstrasse 4, 50939 Koln, Germany
| | - Sebastien Combes
- CRCM,
CNRS UMR7258, Laboratory of Integrative Structural and Chemical Biology
(ISCB), INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Universit́e, UM105,
F-13009, Marseille, France
| | - Diane Allegro
- Centre de Recherche en Oncologie Biologique et en Oncopharmacologie,
CRO2 INSERM UMR 911, Faculte de Pharmacie, Universite d’Aix-Marseille, 27 Boulevard Jean Moulin, Marseille 13005, France
| | - Pascal Barbier
- Centre de Recherche en Oncologie Biologique et en Oncopharmacologie,
CRO2 INSERM UMR 911, Faculte de Pharmacie, Universite d’Aix-Marseille, 27 Boulevard Jean Moulin, Marseille 13005, France
| | - Irina P. Beletskaya
- Department
of Chemistry, M.V. Lomonosov Moscow State University, Vorobyevy Gory, 119992 Moscow, Russian Federation
| | - Elena V. Svirshchevskaya
- Laboratory
of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, GSP-7, Miklukho-Maklaya Street, 16/10, 117997 Moscow, Russian Federation
| | - Alexey Yu. Fedorov
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| |
Collapse
|
4
|
Abstract
Microtubules are dynamic filamentous cytoskeletal proteins composed of tubulin and are an important therapeutic target in tumour cells. Agents that bind to microtubules have been part of the pharmacopoeia of anticancer therapy for decades and until the advent of targeted therapy, microtubules were the only alternative to DNA as a therapeutic target in cancer. The screening of a range of botanical species and marine organisms has yielded promising new antitubulin agents with novel properties. In the current search for novel microtubule-binding agents, enhanced tumour specificity, reduced neurotoxicity and insensitivity to chemoresistance mechanisms are the three main objectives.
Collapse
Affiliation(s)
- Charles Dumontet
- INSERM 590, Faculté Rockefeller, 8 Avenue Rockefeller, 69008 Lyon, France and Université Lyon 1, ISPB, Lyon, F-69003, France.
| | | |
Collapse
|
5
|
Rubenstein SM, Baichwal V, Beckmann H, Clark DL, Frankmoelle W, Roche D, Santha E, Schwender S, Thoolen M, Ye Q, Jaen JC. Hydrophilic, pro-drug analogues of T138067 are efficacious in controlling tumor growth in vivo and show a decreased ability to cross the blood brain barrier. J Med Chem 2001; 44:3599-605. [PMID: 11606124 DOI: 10.1021/jm000478d] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The novel anticancer compound T138067 is an irreversible inhibitor of tubulin polymerization. Amides 3-6 were synthesized using standard methodologies and determined to be significantly less lipophilic than T138067 based on logP calculations. Tubulin polymerization and [(3)H]-T138067 competition assays revealed that these amides are pro-drugs for parent aniline 2. Amides 3-5 showed no detectable signs of crossing the blood brain barrier, while amide 6 was found in extremely small amounts (12 ng/g of brain tissue). Aniline 2, which was formed in vivo from these amides, was found in significantly smaller amounts (approximately 20 to >5000 times) in the brain than when 2 was administered directly. The in vivo efficacy of amide 6 approached that of T138067 and was better tolerated when administered to athymic nude mice bearing MX-1 human mammary tumor xenografts.
Collapse
Affiliation(s)
- S M Rubenstein
- Tularik Inc., Two Corporate Drive, South San Francisco, CA 94080, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Newell DR, Burtles SS, Fox BW, Jodrell DI, Connors TA. Evaluation of rodent-only toxicology for early clinical trials with novel cancer therapeutics. Br J Cancer 1999; 81:760-8. [PMID: 10555743 PMCID: PMC2374299 DOI: 10.1038/sj.bjc.6690761] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Preclinical toxicology studies are performed prior to phase I trials with novel cancer therapeutics to identify a safe clinical starting dose and potential human toxicities. The primary aim of this study was to evaluate the ability of rodent-only toxicology studies to identify a safe phase I trial starting dose. In addition, the ability of murine studies to predict the quantitative and qualitative human toxicology of cancer therapeutics was studied. Data for 25 cancer drugs were collated for which the preclinical and clinical routes and schedules of administration were either the same (22/25), or closely matched. The maximum tolerated dose/dose lethal to 10% of mice (MTD/LD10) was identified for 24 drugs, and in patients the maximum administered dose (MAD) was associated with dose-limiting toxicity (DLT) in initial clinical trials with 20 compounds. In addition, for 13 agents, the toxicity of the drug at one-tenth the mouse MTD/LD10 was also investigated in rats, following repeated administration (20 doses). A phase I trial starting dose of one-tenth the mouse MTD/LD10 (mg m(-2)) was, or would have been, safe for all 25 compounds. With the exception of nausea and vomiting, which cannot be assessed in rodents, other common DLTs were accurately predicted by the murine studies (i.e. 7/7 haematological and 3/3 neurological DLTs). For two of the 13 drugs studied in rats, repeated administration of one-tenth the mouse MTD/LD10 was toxic, leading to a reduction in the phase I trial starting dose; however, one-tenth the mouse MTD/LD10 was subsequently tolerated in patients. For the 20 drugs where clinical DLT was reached, the median ratio of the human MAD to the mouse MTD/LD10 was 2.6 (range 0.2-16) and the median ratio of the clinical starting dose to the MAD was 35 (range 2.3-160). In contrast, in 13 subsequent phase I trials with 11 of the initial 25 drugs, the median ratio of the clinical starting dose to the MAD was 2.8 (range 1.6-56), emphasizing the value of early clinical data in rapidly defining the dose range for therapeutic studies. For all 25 drugs studied, rodent-only toxicology provided a safe and rapid means of identifying the phase I trial starting dose and predicting commonly encountered DLTs. This study has shown that the routine use of a non-rodent species in preclinical toxicology studies prior to initial clinical trials with cancer therapeutics is not necessary.
Collapse
Affiliation(s)
- D R Newell
- Medical School, University of Newcastle, UK
| | | | | | | | | |
Collapse
|
7
|
Etievant C, Barret JM, Kruczynski A, Perrin D, Hill BT. Vinflunine (20',20'-difluoro-3',4'-dihydrovinorelbine), a novel Vinca alkaloid, which participates in P-glycoprotein (Pgp)-mediated multidrug resistance in vivo and in vitro. Invest New Drugs 1998; 16:3-17. [PMID: 9740539 DOI: 10.1023/a:1006022811895] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Vinflunine (VFL) is a novel derivative of vinorelbine (NVB, Navelbine), which has shown markedly superior antitumor activity to NVB, in various experimental animal models. To establish whether this new Vinca alkaloid participates in P-glycoprotein (Pgp)-mediated multidrug resistance (MDR), VFL-resistant murine P388 cells (P388/VFL) were established in vivo and used in conjunction with the well established MDR P388/ADR subline, to define the in vivo resistance profile for VFL. P388/VFL cells proved cross-resistant to drugs implicated in MDR (other Vinca alkaloids, doxorubicin, etoposide), but not to campothecin or cisplatin and showed an increased expression of Pgp, without any detectable alterations in topoisomerase II or in glutathione metabolism. The P388/ADR cells proved cross-resistant to VFL both in vivo and in vitro, and this VFL resistance was efficiently modulated by verapamil in vitro. Cellular transport experiments with tritiated-VFL revealed differential uptake by P388 sensitive and P388/ADR resistant cells, comparable with data obtained using tritiated-NVB. In various in vitro models of human MDR tumor cells, whilst full sensitivity was retained in cells expressing alternative non-Pgp-mediated MDR mechanisms, cross resistance was identified in Pgp-overexpressing cells. Differences were, however, noted in terms of the drug resistance profiles relative to the other Vinca, with tumor cell lines proving generally least cross-resistant to VFL. Overall, these results suggest that VFL, like other Vinca alkaloids, participates in Pgp-mediated MDR, with tumor cells selected for resistance to VFL overexpressing Pgp, yet MDR tumor cell lines proved generally less cross resistant to VFL relative to the other Vinca alkaloids.
Collapse
Affiliation(s)
- C Etievant
- Division de Cancerologie Experimentale I, Centre de Recherche Pierre Fabre, Castres, France
| | | | | | | | | |
Collapse
|