1
|
Ford JW, VanNatta JM, Mondal D, Lin CM, Deng Y, Bai R, Hamel E, Trawick ML, Pinney KG. Drug-Linker Constructs Bearing Unique Dual-Mechanism Tubulin Binding Payloads Tethered through Cleavable and Non-Cleavable Linkers. Tetrahedron 2025; 171:134350. [PMID: 39801742 PMCID: PMC11722312 DOI: 10.1016/j.tet.2024.134350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Antibody-drug conjugates (ADCs) have advanced as a mainstay among the most promising cancer therapeutics, offering enhanced antigen targeting and encompassing wide diversity in their linker and payload components. Small-molecule inhibitors of tubulin polymerization have found success as payloads in FDA approved ADCs and represent further promise in next-generation, pre-clinical and developmental ADCs. Unique dual-mechanism payloads (previously designed and synthesized in our laboratories) function as both potent antiproliferative agents and promising vascular disrupting agents capable of imparting selective and effective damage to tumor-associated microvessels. These payloads have been incorporated into a variety of drug-linker constructs utilizing the clinically relevant cathepsin B cleavable Val-Cit dipeptide linker, employed within several FDA approved ADCs, along with other non-cleavable constructs. Various synthetic strategies were evaluated to prepare these drug-linker constructs. Aniline-based payloads were incorporated utilizing the Val-Cit dipeptide linker similar to FDA approved ADCs such as Adcetris® (brentuximab vedotin). An additional self-immolative group, previously described in the literature for related model systems, was employed to tether the phenolic payloads. A variety of drug-linker constructs (with each bearing a unique dual mechanism payload) were synthesized and evaluated biologically for their enzyme-mediated release of payload and inhibition of tubulin polymerization. Following deactivation of the highly electrophilic maleimido terminus as its corresponding N-acetyl cysteine (NAC) derivative, the most promising construct (NAC-4) demonstrated approximately 90% release of an aniline-functionalized payload (1) upon treatment with cathepsins B or L over 90 minutes. Building on these promising results, future studies will examine the conjugation of drug-linker construct 4 to selected antibodies and engineered proteins and evaluate the biological activity of the resultant antibody-drug conjugates (ADCs).
Collapse
Affiliation(s)
- Jacob W Ford
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, Texas 76798-7348, United States
| | - Jennifer M VanNatta
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, Texas 76798-7348, United States
| | - Deboprosad Mondal
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, Texas 76798-7348, United States
| | - Chen-Ming Lin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, Texas 76798-7348, United States
| | - Yuling Deng
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, Texas 76798-7348, United States
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, Texas 76798-7348, United States
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, Texas 76798-7348, United States
| |
Collapse
|
2
|
Kumar A, Sharma P, Sheetal, Sharma N, Giri K, Das P. Pd-Catalyzed Tandem Approach for 1,2,3-Triazolo-azepine Fused Benzosuberenes and 1,2,3-Triazolobenzazepines Synthesis. J Org Chem 2025. [PMID: 39889210 DOI: 10.1021/acs.joc.4c02454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
Abstract
Herein, a highly efficient palladium-catalyzed tandem approach for the synthesis of 1,2,3-triazolo-azepine-fused benzosuberenes (TAABS) has been developed for the first time with vinyl bromide and internal alkynes as starting precursors. The given reaction proceeded under ligand- and additive-free conditions via sequential carbopalladation, followed by intramolecular electrophilic substitution. Also, the developed protocol has good functional group tolerance, wherein a range of sterically hindered TAABS analogues has been synthesized in appreciable yields. In addition, the developed methodology was also extended for the synthesis of 1,2.3-triazolobenzazepines from triazole-bearing aryl iodides and internal alkynes. The present protocol operates under relatively milder conditions with comparatively shorter reaction time. Furthermore, computational studies were also performed to validate the proposed mechanistic pathways. Additionally, the developed protocol is applicable to the gram-scale synthesis of TAABS analogues.
Collapse
Affiliation(s)
- Ashish Kumar
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, HP, India
- Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| | - Poonam Sharma
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, HP, India
- Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| | - Sheetal
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, HP, India
- Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| | - Navneet Sharma
- Department of Computational Sciences, Central University of Punjab, Bathinda 151401, India
| | - Kousik Giri
- Department of Computational Sciences, Central University of Punjab, Bathinda 151401, India
| | - Pralay Das
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, HP, India
- Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| |
Collapse
|
3
|
VanNatta JM, Niu H, Carlson GJ, Pinney KG. Application of Chlorosulfonyl Isocyanate (CSI) in the Synthesis of Fused Tetracyclic Ketone Ring Systems. J Org Chem 2024; 89:15636-15651. [PMID: 39388523 PMCID: PMC11708961 DOI: 10.1021/acs.joc.4c01714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Chlorosulfonyl isocyanate (CSI) is a complex reagent capable of facilitating numerous synthetic transformations, including lactam/lactone formation, sulfonylation, Friedel-Crafts-type acylations, and cycloadditions. Annulation reactions to form nitrogen-, oxygen-, and sulfur-bearing heterocycles have been observed with CSI; however, the application of CSI toward the generation of fused cyclic ketone ring systems has not been previously reported. A serendipitous discovery of the pertinence of CSI occurred during a structure-activity relationship campaign around our established lead benzosuberene-based molecule that functions as a potent inhibitor of tubulin polymerization. The benzylic olefin within this molecule represents a promising moiety for further functionalization. CSI was initially investigated as a reagent to effect transformation of this olefin to its corresponding β-lactam functionality, but instead resulted in an unexpected tetracyclic fused ring system in high yield (88%). This finding led to an exploration of the reactivity of CSI with various arenes. Benzosuberene analogues with varying functionalizations were synthesized and treated with CSI, with all examples resulting in a fused ring system except those bearing electron-withdrawing groups. Notably, simplified arene structures with fewer substituents were also observed to undergo cyclization under these conditions. This strategy represents a promising approach for the synthesis of appropriately functionalized tetracyclic ring systems.
Collapse
Affiliation(s)
- Jennifer M. VanNatta
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Haichan Niu
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Graham J. Carlson
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| |
Collapse
|
4
|
Lin W, Alix A, Guillot R, Gandon V, Bour C. Aluminum-Catalyzed Intramolecular Vinylation of Arenes by Vinyl Cations. Org Lett 2024; 26:3267-3272. [PMID: 38574281 DOI: 10.1021/acs.orglett.4c00889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
This study addresses the challenges associated with vinyl cation generation, a process that traditionally requires quite specific counterions. Described herein is a novel intramolecular vinylation of arenes catalyzed by aluminum(III) chloride, utilizing practical conditions and readily available vinyl triflates derived from 2-aceto-3-arylpropionates. Comprehensive experimental data support diverse carbocycle synthesis, exemplified by indenes and higher analogues. Control experiments verify the applicability of the vinylation protocol, and synthetic applications showcase a potent tubulin polymerization inhibitor with anticancer properties. Density functional theory computations reveal a Lewis-acid-driven mechanism involving triflate moiety abstraction to generate a reactive vinyl cation.
Collapse
Affiliation(s)
- Wenhua Lin
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, CNRS UMR 8182, Université Paris-Saclay, 17 Avenue des Sciences, 91400 Orsay, France
| | - Aurélien Alix
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, CNRS UMR 8182, Université Paris-Saclay, 17 Avenue des Sciences, 91400 Orsay, France
| | - Régis Guillot
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, CNRS UMR 8182, Université Paris-Saclay, 17 Avenue des Sciences, 91400 Orsay, France
| | - Vincent Gandon
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, CNRS UMR 8182, Université Paris-Saclay, 17 Avenue des Sciences, 91400 Orsay, France
| | - Christophe Bour
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, CNRS UMR 8182, Université Paris-Saclay, 17 Avenue des Sciences, 91400 Orsay, France
| |
Collapse
|
5
|
Ren W, Deng Y, Ward JD, Vairin R, Bai R, Wanniarachchi HI, Hamal KB, Tankoano PE, Tamminga CS, Bueno LMA, Hamel E, Mason RP, Trawick ML, Pinney KG. Synthesis and biological evaluation of structurally diverse 6-aryl-3-aroyl-indole analogues as inhibitors of tubulin polymerization. Eur J Med Chem 2024; 263:115794. [PMID: 37984295 PMCID: PMC11019941 DOI: 10.1016/j.ejmech.2023.115794] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 11/22/2023]
Abstract
The synthesis and evaluation of small-molecule inhibitors of tubulin polymerization remains a promising approach for the development of new therapeutic agents for cancer treatment. The natural products colchicine and combretastatin A-4 (CA4) inspired significant drug discovery campaigns targeting the colchicine site located on the beta-subunit of the tubulin heterodimer, but so far these efforts have not yielded an approved drug for cancer treatment in human patients. Interest in the colchicine site was enhanced by the discovery that a subset of colchicine site agents demonstrated dual functionality as both potent antiproliferative agents and effective vascular disrupting agents (VDAs). Our previous studies led to the discovery and development of a 2-aryl-3-aroyl-indole analogue (OXi8006) that inhibited tubulin polymerization and demonstrated low nM IC50 values against a variety of human cancer cell lines. A water-soluble phosphate prodrug salt (OXi8007), synthesized from OXi8006, displayed promising vascular disrupting activity in mouse models of cancer. To further extend structure-activity relationship correlations, a series of 6-aryl-3-aroyl-indole analogues was synthesized and evaluated for their inhibition of tubulin polymerization and cytotoxicity against human cancer cell lines. Several structurally diverse molecules in this small library were strong inhibitors of tubulin polymerization and of MCF-7 and MDA-MB-231 human breast cancer cells. One of the most promising analogues (KGP591) caused significant G2/M arrest of MDA-MB-231 cells, disrupted microtubule structure and cell morphology in MDA-MB-231 cells, and demonstrated significant inhibition of MDA-MB-231 cell migration in a wound healing (scratch) assay. A phosphate prodrug salt, KGP618, synthesized from its parent phenolic precursor, KGP591, demonstrated significant reduction in bioluminescence signal when evaluated in vivo against an orthotopic model of kidney cancer (RENCA-luc) in BALB/c mice, indicative of VDA efficacy. The most active compounds from this series offer promise as anticancer therapeutic agents.
Collapse
Affiliation(s)
- Wen Ren
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Yuling Deng
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Jacob D Ward
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Rebecca Vairin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD, 21702, United States.
| | - Hashini I Wanniarachchi
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Khagendra B Hamal
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Pouguiniseli E Tankoano
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Caleb S Tamminga
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Lorena M A Bueno
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD, 21702, United States.
| | - Ralph P Mason
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| |
Collapse
|
6
|
He YW, Ma WQ, Han Y, Sun J, Yan CG. Construction of Unique Spiro[dibenzo[ a, f]azulene-6,2'-indenes] via Unprecedented Annulation of ortho-C-H Bond of Benzylidene Group. J Org Chem 2023; 88:14911-14927. [PMID: 37870904 DOI: 10.1021/acs.joc.3c01246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
The domino reaction of alkyl and aryl isocyanides with two molecules of 2-arylidene-1,3-indanediones in acetonitrile at 80 °C resulted in unique functionalized spiro[dibenzo[a,f]azulene-6,2'-indenes] in good yields, in which the two 2-arylidene-1,3-indanediones acted as different building blocks to construct the polycyclic system. More importantly, the unprecedented anticipation of the ortho-position of benzylidene group to form a novel dibenzo[a,f]azulene ring through a formal [5 + 2] cycloaddition process was first observed. On the other hand, DABCO-promoted reaction of the isocyanides with two molecules of 2-arylidene-1,3-indanediones in acetonitrile at 80 °C afforded functionalized spiro[cyclopenta[a]-indene-2,2'-indene] derivatives.
Collapse
Affiliation(s)
- Yu-Wei He
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225002, China
| | - Wei-Qing Ma
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225002, China
| | - Ying Han
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225002, China
| | - Jing Sun
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225002, China
| | - Chao-Guo Yan
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225002, China
| |
Collapse
|
7
|
Ren W, Vairin R, Ward JD, Francis R, VanNatta J, Bai R, Tankoano PE, Deng Y, Hamel E, Trawick ML, Pinney KG. Structure Guided Design, Synthesis, and Biological Evaluation of Oxetane-Containing Indole Analogues. Bioorg Med Chem 2023; 92:117400. [PMID: 37556912 PMCID: PMC10848874 DOI: 10.1016/j.bmc.2023.117400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 08/11/2023]
Abstract
The oxetane functional group offers a variety of potential advantages when incorporated within appropriate therapeutic agents as a ketone surrogate. OXi8006, a 2-aryl-3-aroyl-indole analogue, functions as a small-molecule inhibitor of tubulin polymerization that has a dual mechanism of action as both an antiproliferative agent and a tumor-selective vascular disrupting agent. Replacement of the bridging ketone moiety in OXi8006 with an oxetane functional group has expanded structure activity relationship (SAR) knowledge and provided insights regarding oxetane incorporation within this class of molecules. A new synthetic method using an oxetane-containing tertiary alcohol subjected to Lewis acid catalyzed conditions led to successful Friedel-Crafts alkylation and yielded fourteen new oxetane-containing indole-based molecules. This synthetic approach represents the first method to successfully install an oxetane ring at the 3-position of a 2-aryl-indole system. Several analogues showed potent cytotoxicity (micromolar GI50 values) against human breast cancer cell lines (MCF-7 and MDA-MB-231) and a pancreatic cancer cell line (PANC-1), although they proved to be ineffective as inhibitors of tubulin polymerization. Molecular docking studies comparing colchicine with the OXi8006-oxetane analogue 5m provided a rationale for the differential interaction of these molecules with the colchicine site on the tubulin heterodimer.
Collapse
Affiliation(s)
- Wen Ren
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Rebecca Vairin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Jacob D Ward
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Ricardo Francis
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Jenny VanNatta
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Pouguiniseli E Tankoano
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Yuling Deng
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States.
| |
Collapse
|
8
|
Liu L, Schuetze R, Gerberich JL, Lopez R, Odutola SO, Tanpure RP, Charlton-Sevcik AK, Tidmore JK, Taylor EAS, Kapur P, Hammers H, Trawick ML, Pinney KG, Mason RP. Demonstrating Tumor Vascular Disrupting Activity of the Small-Molecule Dihydronaphthalene Tubulin-Binding Agent OXi6196 as a Potential Therapeutic for Cancer Treatment. Cancers (Basel) 2022; 14:4208. [PMID: 36077745 PMCID: PMC9454770 DOI: 10.3390/cancers14174208] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
The vascular disrupting activity of a promising tubulin-binding agent (OXi6196) was demonstrated in mice in MDA-MB-231 human breast tumor xenografts growing orthotopically in mammary fat pad and syngeneic RENCA kidney tumors growing orthotopically in the kidney. To enhance water solubility, OXi6196, was derivatized as its corresponding phosphate prodrug salt OXi6197, facilitating effective delivery. OXi6197 is stable in water, but rapidly releases OXi6196 in the presence of alkaline phosphatase. At low nanomolar concentrations OXi6196 caused G2/M cell cycle arrest and apoptosis in MDA-MB-231 breast cancer cells and monolayers of rapidly growing HUVECs underwent concentration-dependent changes in their morphology. Loss of the microtubule structure and increased bundling of filamentous actin into stress fibers followed by cell collapse, rounding and blebbing was observed. OXi6196 (100 nM) disrupted capillary-like endothelial networks pre-established with HUVECs on Matrigel®. When prodrug OXi6197 was administered to mice bearing orthotopic MDA-MB-231-luc tumors, dynamic bioluminescence imaging (BLI) revealed dose-dependent vascular shutdown with >80% signal loss within 2 h at doses ≥30 mg/kg and >90% shutdown after 6 h for doses ≥35 mg/kg, which remained depressed by at least 70% after 24 h. Twice weekly treatment with prodrug OXi6197 (20 mg/kg) caused a significant tumor growth delay, but no overall survival benefit. Similar efficacy was observed for the first time in orthotopic RENCA-luc tumors, which showed massive hemorrhage and necrosis after 24 h. Twice weekly dosing with prodrug OXi6197 (35 mg/kg) caused tumor growth delay in most orthotopic RENCA tumors. Immunohistochemistry revealed extensive necrosis, though with surviving peripheral tissues. These results demonstrate effective vascular disruption at doses comparable to the most effective vascular-disrupting agents (VDAs) suggesting opportunities for further development.
Collapse
Affiliation(s)
- Li Liu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Regan Schuetze
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeni L. Gerberich
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ramona Lopez
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Samuel O. Odutola
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | - Rajendra P. Tanpure
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | | | - Justin K. Tidmore
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | - Emily A.-S. Taylor
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | - Payal Kapur
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hans Hammers
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | - Ralph P. Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
9
|
Peng J, Miller M, Li BX, Xiao X. Design, Synthesis and Biological Evaluation of Prodrugs of 666-15 as Inhibitors of CREB-Mediated Gene Transcription. ACS Med Chem Lett 2022; 13:388-395. [PMID: 35300089 PMCID: PMC8919383 DOI: 10.1021/acsmedchemlett.1c00499] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 02/14/2022] [Indexed: 12/28/2022] Open
Abstract
cAMP-response element binding protein (CREB) is a transcription factor involved in multiple cancers. Chemical inhibitors of CREB represent potential anticancer agents. We previously identified 666-15 as a potent CREB inhibitor. While 666-15 showed efficacious anticancer activity in vivo through intraperitoneal (IP) injection, its oral bioavailability is limited. To increase its oral bioavailability, we describe synthesis and evaluation of prodrugs based on 666-15. The amino acid esters were attempted, but they were not stable for detailed characterization. The corresponding sulfate and phosphates were prepared. The sulfate of 666-15 was too stable to release 666-15 while the phosphates were converted into 666-15 with half-lives of ∼2 h. Phosphate 3 was also a potent CREB inhibitor with anti-breast cancer activity. Furthermore, compound 3 showed much improved oral bioavailability at 38%. These studies support that 3 can be used as an oral CREB inhibitor while IP administration of 666-15 is preferred for in vivo applications.
Collapse
Affiliation(s)
- Jiangling Peng
- Department
of Chemical Physiology and Biochemistry, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Mark Miller
- Department
of Chemical Physiology and Biochemistry, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Bingbing X. Li
- Department
of Chemical Physiology and Biochemistry, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Xiangshu Xiao
- Department
of Chemical Physiology and Biochemistry, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| |
Collapse
|
10
|
Wang C, Li Y, Liu Z, Wang Z, Liu Z, Man S, Zhang Y, Bao K, Wu Y, Guan Q, Zuo D, Zhang W. Design, synthesis and biological evaluation of 1-Aryl-5-(4-arylpiperazine-1-carbonyl)-1 H-tetrazols as novel microtubule destabilizers. J Enzyme Inhib Med Chem 2021; 36:549-560. [PMID: 33522315 PMCID: PMC8759721 DOI: 10.1080/14756366.2020.1759582] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A series of 1-aryl-5-(4-arylpiperazine-1-carbonyl)-1H-tetrazols as microtubule destabilizers were designed, synthesised and evaluated for anticancer activity. Based on bioisosterism, we introduced the tetrazole moiety containing the hydrogen-bond acceptors as B-ring of XRP44X analogues. The key intermediates ethyl 1-aryl-1H-tetrazole-5-carboxylates 10 can be simply and efficiently prepared via a microwave-assisted continuous operation process. Among the compounds synthesised, compound 6-31 showed noteworthy potency against SGC-7901, A549 and HeLa cell lines. In mechanism studies, compound 6-31 inhibited tubulin polymerisation and disorganised microtubule in SGC-7901 cells by binding to tubulin. Moreover, compound 6-31 arrested SGC-7901cells in G2/M phase. This study provided a new perspective for development of antitumor agents that target tubulin.
Collapse
Affiliation(s)
- Chao Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Yuelin Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Zi Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Zeyu Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Zihan Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Shuai Man
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Yujing Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Kai Bao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
11
|
Tang H, Liang Y, Cheng J, Ding K, Wang Y. Bifunctional chiral selenium-containing 1,4-diarylazetidin-2-ones with potent antitumor activities by disrupting tubulin polymerization and inducing reactive oxygen species production. Eur J Med Chem 2021; 221:113531. [PMID: 34044345 DOI: 10.1016/j.ejmech.2021.113531] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/05/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023]
Abstract
Organoselenium compounds have attracted growing interests as promising antitumor agents over recent years. Herein, four series of novel selenium-containing chiral 1,4-diarylazetidin-2-ones were asymmetrically synthesized and biologically evaluated for antitumor activities. Among them, compound 7 was found to be about 10-fold more potent than its prototype compound 1a, and compound 9a exhibited the most potent cytotoxicity against five human cancer cell lines, including a paclitaxel-resistant human ovarian cancer cell line A2780T, with IC50 values ranging from 1 to 3 nM. Mechanistic studies revealed that compound 9a worked by disrupting tubulin polymerization, inducing reactive oxygen species (ROS) production, decreasing mitochondrial membrane potential, blocking the cell cycle in the G2/M phase, inducing cellular apoptosis and suppressing angiogenesis. Additionally, compound 9a exhibited appropriate human-microsomal metabolic stability and physicochemical properties. Importantly, compound 9a was found to inhibit tumor growth effectively in a xenograft mice model with low toxicity profile, which rendered 9a a highly promising candidate for further pre-clinical development.
Collapse
Affiliation(s)
- Hairong Tang
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yuru Liang
- School of Pharmacy, Fudan University, Shanghai, 201203, China; State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Jiayi Cheng
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Kuiling Ding
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Yang Wang
- School of Pharmacy, Fudan University, Shanghai, 201203, China; Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
12
|
Liu L, O’Kelly D, Schuetze R, Carlson G, Zhou H, Trawick ML, Pinney KG, Mason RP. Non-Invasive Evaluation of Acute Effects of Tubulin Binding Agents: A Review of Imaging Vascular Disruption in Tumors. Molecules 2021; 26:2551. [PMID: 33925707 PMCID: PMC8125421 DOI: 10.3390/molecules26092551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor vasculature proliferates rapidly, generally lacks pericyte coverage, and is uniquely fragile making it an attractive therapeutic target. A subset of small-molecule tubulin binding agents cause disaggregation of the endothelial cytoskeleton leading to enhanced vascular permeability generating increased interstitial pressure. The resulting vascular collapse and ischemia cause downstream hypoxia, ultimately leading to cell death and necrosis. Thus, local damage generates massive amplification and tumor destruction. The tumor vasculature is readily accessed and potentially a common target irrespective of disease site in the body. Development of a therapeutic approach and particularly next generation agents benefits from effective non-invasive assays. Imaging technologies offer varying degrees of sophistication and ease of implementation. This review considers technological strengths and weaknesses with examples from our own laboratory. Methods reveal vascular extent and patency, as well as insights into tissue viability, proliferation and necrosis. Spatiotemporal resolution ranges from cellular microscopy to single slice tomography and full three-dimensional views of whole tumors and measurements can be sufficiently rapid to reveal acute changes or long-term outcomes. Since imaging is non-invasive, each tumor may serve as its own control making investigations particularly efficient and rigorous. The concept of tumor vascular disruption was proposed over 30 years ago and it remains an active area of research.
Collapse
Affiliation(s)
- Li Liu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Devin O’Kelly
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Regan Schuetze
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Graham Carlson
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Heling Zhou
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Ralph P. Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| |
Collapse
|
13
|
Serkova NJ, Glunde K, Haney CR, Farhoud M, De Lille A, Redente EF, Simberg D, Westerly DC, Griffin L, Mason RP. Preclinical Applications of Multi-Platform Imaging in Animal Models of Cancer. Cancer Res 2021; 81:1189-1200. [PMID: 33262127 PMCID: PMC8026542 DOI: 10.1158/0008-5472.can-20-0373] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/10/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022]
Abstract
In animal models of cancer, oncologic imaging has evolved from a simple assessment of tumor location and size to sophisticated multimodality exploration of molecular, physiologic, genetic, immunologic, and biochemical events at microscopic to macroscopic levels, performed noninvasively and sometimes in real time. Here, we briefly review animal imaging technology and molecular imaging probes together with selected applications from recent literature. Fast and sensitive optical imaging is primarily used to track luciferase-expressing tumor cells, image molecular targets with fluorescence probes, and to report on metabolic and physiologic phenotypes using smart switchable luminescent probes. MicroPET/single-photon emission CT have proven to be two of the most translational modalities for molecular and metabolic imaging of cancers: immuno-PET is a promising and rapidly evolving area of imaging research. Sophisticated MRI techniques provide high-resolution images of small metastases, tumor inflammation, perfusion, oxygenation, and acidity. Disseminated tumors to the bone and lung are easily detected by microCT, while ultrasound provides real-time visualization of tumor vasculature and perfusion. Recently available photoacoustic imaging provides real-time evaluation of vascular patency, oxygenation, and nanoparticle distributions. New hybrid instruments, such as PET-MRI, promise more convenient combination of the capabilities of each modality, enabling enhanced research efficacy and throughput.
Collapse
Affiliation(s)
- Natalie J Serkova
- Department of Radiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
- Animal Imaging Shared Resource, University of Colorado Cancer Center, Aurora, Colorado
| | - Kristine Glunde
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology, and the Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Chad R Haney
- Center for Advanced Molecular Imaging, Northwestern University, Evanston, Illinois
| | | | | | | | - Dmitri Simberg
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - David C Westerly
- Animal Imaging Shared Resource, University of Colorado Cancer Center, Aurora, Colorado
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lynn Griffin
- Department of Radiology, Veterinary Teaching Hospital, Colorado State University, Fort Collins, Colorado
| | - Ralph P Mason
- Department of Radiology, University of Texas Southwestern, Dallas, Texas
| |
Collapse
|
14
|
Zhu T, Wang SH, Li D, Wang SY, Liu X, Song J, Wang YT, Zhang SY. Progress of tubulin polymerization activity detection methods. Bioorg Med Chem Lett 2021; 37:127698. [PMID: 33468346 DOI: 10.1016/j.bmcl.2020.127698] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/05/2020] [Accepted: 11/14/2020] [Indexed: 12/13/2022]
Abstract
Tubulin, an important target in tumor therapy, is one of the hotspots in the field of antineoplastic drugs in recent years, and it is of great significance to design and screen new inhibitors for this target. Natural products and chemical synthetic drugs are the main sources of tubulin inhibitors. However, due to the variety of compound structure types, it has always been difficult for researchers to screen out polymerization inhibitors with simple operation, high efficiency and low cost. A large number of articles have reported the screening methods of tubulin inhibitors and their biological activity. In this article, the biological activity detection methods of tubulin polymerization inhibitors are reviewed. Thus, it provides a theoretical basis for the further study of tubulin polymerization inhibitors and the selection of methods for tubulin inhibitors.
Collapse
Affiliation(s)
- Ting Zhu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Sheng-Hui Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Dong Li
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shu-Yu Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xu Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Ya-Ting Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
15
|
Liao LS, Chen Y, Mo ZY, Hou C, Su GF, Liang H, Chen ZF. Ni(ii), Cu(ii) and Zn(ii) complexes with the 1-trifluoroethoxyl-2,9,10-trimethoxy-7-oxoaporphine ligand simultaneously target microtubules and mitochondria for cancer therapy. Inorg Chem Front 2021. [DOI: 10.1039/d0qi01463j] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Complexes 1–3 display potent anticancer activity against T-24 cell by disrupting mitochondria and microtubules. Furthermore, complex 1 exhibits almost same tumor growth inhibition activity in T-24 xenograft mouse model as cisplatin and paclitaxel.
Collapse
Affiliation(s)
- Lan-Shan Liao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources
- School of Chemistry and Pharmacy
- Guangxi Normal University
- Guilin 541004
- China
| | - Yin Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources
- School of Chemistry and Pharmacy
- Guangxi Normal University
- Guilin 541004
- China
| | - Zu-Yu Mo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources
- School of Chemistry and Pharmacy
- Guangxi Normal University
- Guilin 541004
- China
| | - Cheng Hou
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources
- School of Chemistry and Pharmacy
- Guangxi Normal University
- Guilin 541004
- China
| | - Gui-Fa Su
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources
- School of Chemistry and Pharmacy
- Guangxi Normal University
- Guilin 541004
- China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources
- School of Chemistry and Pharmacy
- Guangxi Normal University
- Guilin 541004
- China
| | - Zhen-Feng Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources
- School of Chemistry and Pharmacy
- Guangxi Normal University
- Guilin 541004
- China
| |
Collapse
|
16
|
Development of triazolothiadiazine derivatives as highly potent tubulin polymerization inhibitors: Structure-activity relationship, in vitro and in vivo study. Eur J Med Chem 2020; 208:112847. [DOI: 10.1016/j.ejmech.2020.112847] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022]
|
17
|
Winn BA, Devkota L, Kuch B, MacDonough MT, Strecker TE, Wang Y, Shi Z, Gerberich JL, Mondal D, Ramirez AJ, Hamel E, Chaplin DJ, Davis P, Mason RP, Trawick ML, Pinney KG. Bioreductively Activatable Prodrug Conjugates of Combretastatin A-1 and Combretastatin A-4 as Anticancer Agents Targeted toward Tumor-Associated Hypoxia. JOURNAL OF NATURAL PRODUCTS 2020; 83:937-954. [PMID: 32196334 PMCID: PMC7644341 DOI: 10.1021/acs.jnatprod.9b00773] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The natural products combretastatin A-1 (CA1) and combretastatin A-4 (CA4) function as potent inhibitors of tubulin polymerization and as selective vascular disrupting agents (VDAs) in tumors. Bioreductively activatable prodrug conjugates (BAPCs) can enhance selectivity by serving as substrates for reductase enzymes specifically in hypoxic regions of tumors. A series of CA1-BAPCs incorporating nor-methyl, mono-methyl, and gem-dimethyl nitrothiophene triggers were synthesized together with corresponding CA4-BAPCs, previously reported by Davis (Mol. Cancer Ther. 2006, 5 (11), 2886), for comparison. The CA4-gem-dimethylnitrothiophene BAPC 45 proved exemplary in comparison to its nor-methyl 43 and mono-methyl 44 congeners. It was stable in phosphate buffer (pH 7.4, 24 h), was cleaved (25%, 90 min) by NADPH-cytochrome P450 oxidoreductase (POR), was inactive (desirable prodrug attribute) as an inhibitor of tubulin polymerization (IC50 > 20 μM), and demonstrated hypoxia-selective activation in the A549 cell line [hypoxia cytotoxicity ratio (HCR) = 41.5]. The related CA1-gem-dimethylnitrothiophene BAPC 41 was also promising (HCR = 12.5) with complete cleavage (90 min) upon treatment with POR. In a preliminary in vivo dynamic bioluminescence imaging study, BAPC 45 (180 mg/kg, ip) induced a decrease (within 4 h) in light emission in a 4T1 syngeneic mouse breast tumor model, implying activation and vascular disruption.
Collapse
Affiliation(s)
- Blake A. Winn
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Laxman Devkota
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Bunnarack Kuch
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Matthew T. MacDonough
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Tracy E. Strecker
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Yifan Wang
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Zhe Shi
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Jeni L. Gerberich
- Predictive Imaging Research Laboratory, Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9058, United States
| | - Deboprosad Mondal
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Alejandro J. Ramirez
- Mass Spectrometry Center, Baylor University, One Bear Place #97046, Waco, Texas 76798-7046, United States
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - David J. Chaplin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
- Fast Biopharma Ltd., 10 Aston Park, Aston Rowant, OX49 5SW, United Kingdom
| | - Peter Davis
- Fast Biopharma Ltd., 10 Aston Park, Aston Rowant, OX49 5SW, United Kingdom
| | - Ralph P. Mason
- Predictive Imaging Research Laboratory, Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9058, United States
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| |
Collapse
|
18
|
Colchicine-Binding Site Inhibitors from Chemistry to Clinic: A Review. Pharmaceuticals (Basel) 2020; 13:ph13010008. [PMID: 31947889 PMCID: PMC7168938 DOI: 10.3390/ph13010008] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023] Open
Abstract
It is over 50 years since the discovery of microtubules, and they have become one of the most important drug targets for anti-cancer therapies. Microtubules are predominantly composed of the protein tubulin, which contains a number of different binding sites for small-molecule drugs. There is continued interest in drug development for compounds targeting the colchicine-binding site of tubulin, termed colchicine-binding site inhibitors (CBSIs). This review highlights CBSIs discovered through diverse sources: from natural compounds, rational design, serendipitously and via high-throughput screening. We provide an update on CBSIs reported in the past three years and discuss the clinical status of CBSIs. It is likely that efforts will continue to develop CBSIs for a diverse set of cancers, and this review provides a timely update on recent developments.
Collapse
|
19
|
Chang MY, Lai KX, Chang YL. In(OTf)3-catalyzed intramolecular hydroarylation of α-phenylallyl β-ketosulfones – synthesis of sulfonyl 1-benzosuberones and 1-tetralones. RSC Adv 2020; 10:18231-18244. [PMID: 35517185 PMCID: PMC9053753 DOI: 10.1039/d0ra01962c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 05/05/2020] [Indexed: 11/21/2022] Open
Abstract
In(OTf)3-catalyzed intramolecular hydroarylation of α-phenylallyl β-ketosulfones provides sulfonyl 1-benzosuberones and 1-tetralones in moderate to good yields in refluxing (CH2Cl)2 under open-vessel and easy-operation reaction conditions. A plausible mechanism is proposed and discussed. This highly regioselective protocol provides an atom-economic ring-closure route. In(OTf)3-catalyzed intramolecular hydroarylation of α-phenylallyl β-ketosulfones provides sulfonyl 1-benzosuberones and 1-tetralones in moderate to good yields in refluxing (CH2Cl)2 under open-vessel and easy-operation reaction conditions.![]()
Collapse
Affiliation(s)
- Meng-Yang Chang
- Department of Medicinal and Applied Chemistry
- Kaohsiung Medical University Hospital
- Kaohsiung Medical University
- Kaohsiung 80708
- Taiwan
| | - Kai-Xiang Lai
- Department of Medicinal and Applied Chemistry
- Kaohsiung Medical University Hospital
- Kaohsiung Medical University
- Kaohsiung 80708
- Taiwan
| | - Yu-Lun Chang
- Department of Medicinal and Applied Chemistry
- Kaohsiung Medical University Hospital
- Kaohsiung Medical University
- Kaohsiung 80708
- Taiwan
| |
Collapse
|
20
|
Zhou Y, Di B, Niu MM. Structure-Based Pharmacophore Design and Virtual Screening for Novel Tubulin Inhibitors with Potential Anticancer Activity. Molecules 2019; 24:E3181. [PMID: 31480625 PMCID: PMC6749218 DOI: 10.3390/molecules24173181] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/24/2022] Open
Abstract
Tubulin inhibitors have been considered as potential drugs for cancer therapy. However, their drug resistance and serious side-effects are the main reasons for clinical treatment failure. Therefore, there is still an urgent need to develop effective therapeutic drugs. Herein, a structure-based pharmacophore model was developed based on the co-crystallized structures of the tubulin with a high resolution. The model including one hydrogen-bond acceptor feature, two aromatic features, and one hydrophobic feature was further validated using the Gunner-Henry score method. Virtual screening was performed by an integrated protocol that combines drug-likeness analysis, pharmacophore mapping, and molecular docking approaches. Finally, five hits were selected for biological evaluation. The results indicated that all these hits at the concentration of 40 μM showed an inhibition of more than 50% against five human tumor cells (MCF-7, U87MG, HCT-116, MDA-MB-231, and HepG2). Particularly, hit 1 effectively inhibited the proliferation of these tumor cells, with inhibition rates of more than 80%. The results of tubulin polymerization and colchicine-site competition assays suggested that hit 1 significantly inhibited tubulin polymerization by binding to the colchicine site. Thus, hit 1 could be used as a potential chemotherapeutic agent for cancer treatment. This work also demonstrated the potential of our screening protocol to identify biologically active compounds.
Collapse
Affiliation(s)
- Yunjiang Zhou
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Bin Di
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China.
| | - Miao-Miao Niu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|