1
|
Dhasmana A, Mishra AK, Khoja UB, Mishra S. Molecular structure, spectral analysis and chemical activity of sabizabulin: A computational study. J Mol Graph Model 2023; 125:108618. [PMID: 37678041 DOI: 10.1016/j.jmgm.2023.108618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/07/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023]
Abstract
In this study, a detailed computational spectroscopic investigation of sabizabulin, a small molecule known as a tubulin inhibitor with potential antineoplastic, antiviral, and anti-inflammatory activities, has been presented. Our work utilizes Density Functional Theory (DFT) calculations to explore molecular optimization, thermodynamic characteristics, and the analysis of normal modes with vibrational assignments. We calculate essential properties such as standard zero-point vibrational energy, entropy, dipole moment, etc., based on data extracted from the optimized molecular structure. Additionally, we examine Mulliken charges and the Molecular Electrostatic Potential (MEP) plot to comprehend the electronic distribution and chemical activity of sabizabulin. Our findings provide valuable insights into the spectroscopic properties of sabizabulin, highlighting its potential therapeutic applications. Our work aims to explore future research directions that could expand the understanding of sabizabulin's actions and enhance its applicability in medical treatments.
Collapse
Affiliation(s)
- Abhishek Dhasmana
- Department of Physics, Graphic Era Hill University, Dehradun, 248002, India
| | - Abhishek Kumar Mishra
- Department of Physics, Applied Science Cluster, School of Engineering, University of Petroleum and Energy Studies (UPES), Dehradun, 248007, Uttarakhand, India; Quantum Computing Centre, School of Computer Science, University of Petroleum and Energy Studies (UPES), Dehradun, 248007, Uttarakhand, India.
| | - Ummer Bashir Khoja
- Department of Allied Sciences, Graphic Era Deemed to be University, Clement Town, Dehradun, 248002, Uttarakhand, India
| | - Soni Mishra
- Department of Physics, Graphic Era Hill University, Dehradun, 248002, India.
| |
Collapse
|
2
|
Krutilina RI, Hartman KL, Oluwalana D, Playa HC, Parke DN, Chen H, Miller DD, Li W, Seagroves TN. Sabizabulin, a Potent Orally Bioavailable Colchicine Binding Site Agent, Suppresses HER2+ Breast Cancer and Metastasis. Cancers (Basel) 2022; 14:5336. [PMID: 36358755 PMCID: PMC9658816 DOI: 10.3390/cancers14215336] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 07/30/2023] Open
Abstract
HER2+ breast cancer accounts for 15% of all breast cancer cases. Current frontline therapy for HER2+ metastatic breast cancer relies on targeted antibodies, trastuzumab and pertuzumab, combined with microtubule inhibitors in the taxane class (paclitaxel or docetaxel). It is well known that the clinical efficacy of taxanes is limited by the development of chemoresistance and hematological and neurotoxicities. The colchicine-binding site inhibitors (CBSIs) are a class of promising alternative agents to taxane therapy. Sabizabulin (formerly known as VERU-111) is a potent CBSI that overcomes P-gp-mediated taxane resistance, is orally bioavailable, and inhibits tumor growth and distant metastasis in triple negative breast cancer (TNBC). Herein, we demonstrate the efficacy of sabizabulin in HER2+ breast cancer. In vitro, sabizabulin inhibits the proliferation of HER2+ breast cancer cell lines with low nanomolar IC50 values, inhibits clonogenicity, and induces apoptosis in a concentration-dependent manner. In vivo, sabizabulin inhibits breast tumor growth in the BT474 (ER+/PR+/HER2+) xenograft model and a HER2+ (ER-/PR-) metastatic patient-derived xenograft (PDX) model, HCI-12. We demonstrate that sabizabulin is a promising alternative agent to target tubulin in HER2+ breast cancer with similar anti-metastatic efficacy to paclitaxel, but with the advantage of oral bioavailability and lower toxicity than taxanes.
Collapse
Affiliation(s)
- Raisa I. Krutilina
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Kelli L. Hartman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Damilola Oluwalana
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Hilaire C. Playa
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Deanna N. Parke
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Duane D. Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Drug Discovery Center, College of Pharmacy, University of Tennessee, Memphis, TN 38103, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Drug Discovery Center, College of Pharmacy, University of Tennessee, Memphis, TN 38103, USA
| | - Tiffany N. Seagroves
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| |
Collapse
|
3
|
Indole derivatives (2010-2020) as versatile tubulin inhibitors: synthesis and structure-activity relationships. Future Med Chem 2021; 13:1795-1828. [PMID: 34468201 DOI: 10.4155/fmc-2020-0385] [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] [Indexed: 01/07/2023] Open
Abstract
Tubulin inhibitors are conjugates that interfere with the dynamic equilibrium of the polymerization and depolymerization of microtubules. Among all the reported conjugates, indole moiety is one of the most significant classes for the development of new drug candidates for cancer therapy. Due to their presence in a wide range of natural as well as synthetic antitubulin agents, indole has become a versatile scaffold in research, and various synthetic and semisynthetic indole-based antitubulin agents have been identified and reported. The present article focuses on the reported indole-based tubulin inhibitors of synthetic origin from last the decade. Synthesis, structure-activity relationships and biological activities of synthetic indole derivatives along with brief updates on their antitubulin activity are presented.
Collapse
|
4
|
Recent advances in research of colchicine binding site inhibitors and their interaction modes with tubulin. Future Med Chem 2021; 13:839-858. [PMID: 33821673 DOI: 10.4155/fmc-2020-0376] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Microtubules have been a concerning target of cancer chemotherapeutics for decades, and several tubulin-targeted agents, such as paclitaxel, vincristine and vinorelbine, have been approved. The colchicine binding site is one of the primary targets on microtubules and possesses advantages compared with other tubulin-targeted agents, such as inhibitors of tumor vessels and overcoming P-glycoprotein overexpression-mediated multidrug resistance. This study reviews and summarizes colchicine binding site inhibitors reported in recent years with structural studies via the crystal structures of complexes or computer simulations to discover new lead compounds. We are attempting to resolve the challenge of colchicine site agent research.
Collapse
|
5
|
Tantak MP, Malik M, Klingler L, Olson Z, Kumar A, Sadana R, Kumar D. Indolyl-α-keto-1,3,4-oxadiazoles: Synthesis, anti-cell proliferation activity, and inhibition of tubulin polymerization. Bioorg Med Chem Lett 2021; 37:127842. [PMID: 33556575 DOI: 10.1016/j.bmcl.2021.127842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/14/2021] [Accepted: 01/29/2021] [Indexed: 01/29/2023]
Abstract
A series of novel indolyl-α-keto-1,3,4-oxadiazole derivatives have been synthesized by employing molecular iodine-mediated oxidative cyclization of acylhydrazones. In vitro anti cell proliferation activity of these derivatives against various cancer cells lines such as human lymphoblast (U937), leukemia (Jurkat & SB) and human breast (BT474) was investigated. Among the synthesized indolyl-α-keto-1,3,4-oxadiazoles 19a-p, only one compound (19e) exhibited significant antiproliferative activity against a panel of cell lines. The compound 19e with 3,4,5-trimethoxyphenyl motif, endowed strong cytotoxicity against U937, Jurkat, BT474 and SB cancer cells with IC50 values of 7.1, 3.1, 4.1, and 0.8 µM, respectively. Molecular docking studies suggested a potential binding mode for 19e in the colchicine binding site of tubulin. When tested for in vitro tubulin polymerizaton, 19e inhibited tubulin polymezations (IC50 = 10.66 µM) and induced apoptosis through caspase 3/7 activation. Further, the derivative 19e did not cause necrosis when measured using lactate dehydrogenase assay.
Collapse
Affiliation(s)
- Mukund P Tantak
- Department: Department of Chemistry Birla Institute of Technology and Science, Pilani 333 031, India
| | - Monika Malik
- Department: Department of Chemistry Birla Institute of Technology and Science, Pilani 333 031, India
| | - Linus Klingler
- Department: Department of Natural Sciences, University of Houston - Downtown, Houston, TX 77002, USA
| | - Zachary Olson
- Department: Department of Natural Sciences, University of Houston - Downtown, Houston, TX 77002, USA
| | - Anil Kumar
- Department: Department of Chemistry Birla Institute of Technology and Science, Pilani 333 031, India
| | - Rachna Sadana
- Department: Department of Natural Sciences, University of Houston - Downtown, Houston, TX 77002, USA.
| | - Dalip Kumar
- Department: Department of Chemistry Birla Institute of Technology and Science, Pilani 333 031, India.
| |
Collapse
|
6
|
Deng S, Krutilina RI, Wang Q, Lin Z, Parke DN, Playa HC, Chen H, Miller DD, Seagroves TN, Li W. An Orally Available Tubulin Inhibitor, VERU-111, Suppresses Triple-Negative Breast Cancer Tumor Growth and Metastasis and Bypasses Taxane Resistance. Mol Cancer Ther 2019; 19:348-363. [PMID: 31645441 DOI: 10.1158/1535-7163.mct-19-0536] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/27/2019] [Accepted: 10/15/2019] [Indexed: 12/23/2022]
Abstract
Triple-negative breast cancer (TNBC) accounts for approximately 15% of breast cancer cases in the United States. TNBC has poorer overall prognosis relative to other molecular subtypes due to rapid onset of drug resistance to conventional chemotherapies and increased risk of visceral metastases. Taxanes like paclitaxel are standard chemotherapies that stabilize microtubules, but their clinical efficacy is often limited by drug resistance and neurotoxicities. We evaluated the preclinical efficacy of a novel, potent, and orally bioavailable tubulin inhibitor, VERU-111, in TNBC models. VERU-111 showed potent cytotoxicity against TNBC cell lines, inducing apoptosis and cell-cycle arrest in a concentration-dependent manner. VERU-111 also efficiently inhibited colony formation, cell migration, and invasion. Orally administered VERU-111 inhibited MDA-MB-231 xenograft growth in a dose-dependent manner, with similar efficacies to paclitaxel, but without acute toxicity. VERU-111 significantly reduced metastases originating from the mammary fat pad into lung, liver, and kidney metastasis in an experimental metastasis model. Moreover, VERU-111, but not paclitaxel, suppressed growth of luciferase-labeled, taxane-resistant, patient-derived metastatic TNBC tumors. In this model, VERU-111 repressed growth of preestablished axillary lymph node metastases and lung, bone, and liver metastases at study endpoint, whereas paclitaxel enhanced liver metastases relative to vehicle controls. Collectively, these studies strongly suggest that VERU-111 is not only a potent inhibitor of aggressive TNBC phenotypes, but it is also efficacious in a taxane-resistant model of metastatic TNBC. Thus, VERU-111 is a promising new generation of tubulin inhibitor for the treatment of TNBC and may be effective in patients who progress on taxanes.Results presented in this study demonstrate the efficacy of VERU-111 in vivo and provide strong rationale for future development of VERU-111 as an effective treatment for metastatic breast cancer.
Collapse
Affiliation(s)
- Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Raisa I Krutilina
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Qinghui Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Zongtao Lin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Deanna N Parke
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hilaire C Playa
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Tiffany N Seagroves
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee. .,Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee.
| |
Collapse
|
7
|
Bariwal J, Kumar V, Chen H, Bhattarai RS, Peng Y, Li W, Mahato RI. Nanoparticulate delivery of potent microtubule inhibitor for metastatic melanoma treatment. J Control Release 2019; 309:231-243. [PMID: 31330213 DOI: 10.1016/j.jconrel.2019.07.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/09/2019] [Accepted: 07/17/2019] [Indexed: 12/31/2022]
Abstract
Melanoma is the most aggressive type of skin cancer, which readily metastasizes through lymph nodes to the lungs, liver, and brain. Since the repeated administration of most chemotherapeutic drugs develops chemoresistance and severe systemic toxicities, herein we synthesized 2-(4-hydroxy-1H-indol-3-yl)-1H-imidazol-4-yl)(3,4,5-trimethoxyphenyl) methanone (abbreviated as QW-296), a novel tubulin destabilizing agent with little susceptible to transporter-mediated drug resistance. QW-296 disturbed the microtubule dynamics at the nanomolar concentration in A375 and B16F10 melanoma cells. QW-296 binding to colchicine-binding site on tubulin protein was confirmed by molecular modeling and tubulin polymerization assay. QW-296 significantly inhibited A375 and B16F10 cell proliferation, induced G2/M cell cycle arrest and led to apoptosis and cell death. To improve its aqueous solubility, QW-296 was encapsulated into methoxy poly(ethyleneglycol)-b-poly(carbonate-co-lactide) [mPEG-b-P(CB-co-LA)] polymeric nanoparticles by solvent evaporation, with the mean particle size of 122.0 ± 2.28 nm and drug loading of 3.70% (w/w). Systemic administration of QW-296 loaded nanoparticles into C57/BL6 albino mice bearing lung metastatic melanoma at the dose of 20 mg/kg 4 times a week for 1.5 weeks resulted in significant tumor regression and prolonged mouse median survival without significant change in mouse body weight. In conclusion, QW-296 loaded nanoparticles have the potential to treat metastatic melanoma.
Collapse
Affiliation(s)
- Jitender Bariwal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Hao Chen
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Rajan Sharma Bhattarai
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yang Peng
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
8
|
Arnst KE, Wang Y, Lei ZN, Hwang DJ, Kumar G, Ma D, Parke DN, Chen Q, Yang J, White SW, Seagroves TN, Chen ZS, Miller DD, Li W. Colchicine Binding Site Agent DJ95 Overcomes Drug Resistance and Exhibits Antitumor Efficacy. Mol Pharmacol 2019; 96:73-89. [PMID: 31043459 PMCID: PMC6553560 DOI: 10.1124/mol.118.114801] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 04/21/2019] [Indexed: 02/05/2023] Open
Abstract
Interfering with microtubule dynamics is a well-established strategy in cancer treatment; however, many microtubule-targeting agents are associated with drug resistance and adverse effects. Substantial evidence points to ATP-binding cassette (ABC) transporters as critical players in the development of resistance. Herein, we demonstrate the efficacy of DJ95 (2-(1H-indol-6-yl)-4-(3,4,5-trimethoxyphenyl)-1H-imidazo[4,5-c]pyridine), a novel tubulin inhibitor, in a variety of cancer cell lines, including malignant melanomas, drug-selected resistant cell lines, specific ABC transporter-overexpressing cell lines, and the National Cancer Institute 60 cell line panel. DJ95 treatment inhibited cancer cell migration, caused morphologic changes to the microtubule network foundation, and severely disrupted mitotic spindle formation of mitotic cells. The high-resolution crystal structure of DJ95 in complex with tubulin protein and the detailed molecular interactions confirmed its direct binding to the colchicine site. In vitro pharmacological screening of DJ95 using SafetyScreen44 (Eurofins Cerep-Panlabs) revealed no significant off-target interactions, and pharmacokinetic analysis showed that DJ95 was maintained at therapeutically relevant plasma concentrations for up to 24 hours in mice. In an A375 xenograft model in nude mice, DJ95 inhibited tumor growth and disrupted tumor vasculature in xenograft tumors. These results demonstrate that DJ95 is potent against a variety of cell lines, demonstrated greater potency to ABC transporter-overexpressing cell lines than existing tubulin inhibitors, directly targets the colchicine binding domain, exhibits significant antitumor efficacy, and demonstrates vascular-disrupting properties. Collectively, these data suggest that DJ95 has great potential as a cancer therapeutic, particularly for multidrug resistance phenotypes, and warrants further development. SIGNIFICANCE STATEMENT: Paclitaxel is a widely used tubulin inhibitor for cancer therapy, but its clinical efficacy is often limited by the development of multidrug resistance. In this study, we reported the preclinical characterization of a new tubulin inhibitor DJ95, and demonstrated its abilities to overcome paclitaxel resistance, disrupt tumor vasculature, and exhibit significant antitumor efficacy.
Collapse
Affiliation(s)
- Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Yuxi Wang
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Gyanendra Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Dejian Ma
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Deanna N Parke
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Qiang Chen
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Jinliang Yang
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Stephen W White
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Tiffany N Seagroves
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| |
Collapse
|
9
|
Arnst KE, Wang Y, Hwang DJ, Xue Y, Costello T, Hamilton D, Chen Q, Yang J, Park F, Dalton JT, Miller DD, Li W. A Potent, Metabolically Stable Tubulin Inhibitor Targets the Colchicine Binding Site and Overcomes Taxane Resistance. Cancer Res 2017; 78:265-277. [PMID: 29180476 DOI: 10.1158/0008-5472.can-17-0577] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 09/01/2017] [Accepted: 11/01/2017] [Indexed: 11/16/2022]
Affiliation(s)
- Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Yi Xue
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Terry Costello
- Department of Comparative Medicine, College of Medicine, the University of Tennessee Health Science Center, Memphis, Tennessee
| | - David Hamilton
- Department of Comparative Medicine, College of Medicine, the University of Tennessee Health Science Center, Memphis, Tennessee
| | - Qiang Chen
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - James T Dalton
- College of Pharmacy, The University of Michigan, Ann Arbor, Michigan
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee.
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee.
| |
Collapse
|
10
|
Mahal K, Biersack B, Schruefer S, Resch M, Ficner R, Schobert R, Mueller T. Combretastatin A-4 derived 5-(1-methyl-4-phenyl-imidazol-5-yl)indoles with superior cytotoxic and anti-vascular effects on chemoresistant cancer cells and tumors. Eur J Med Chem 2016; 118:9-20. [PMID: 27116710 DOI: 10.1016/j.ejmech.2016.04.045] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/15/2016] [Accepted: 04/16/2016] [Indexed: 12/27/2022]
Abstract
5-(1-Methyl-4-phenyl-imidazol-5-yl)indoles 5 were prepared and tested as analogs of the natural vascular-disrupting agent combretastatin A-4 (CA-4). The 3-bromo-4,5-dimethoxyphenyl derivative 5c was far more active than CA-4 with low nanomolar IC50 concentrations against multidrug-resistant KB-V1/Vbl cervix and MCF-7/Topo mamma carcinoma cells, and also against CA-4-resistant HT-29 colon carcinoma cells. While not interfering markedly with the polymerization of tubulin in vitro, indole 5c completely disrupted the microtubule cytoskeleton of cancer cells at low concentrations. It also destroyed real blood vessels, both in the chorioallantoic membrane (CAM) of fertilized chicken eggs and within tumor xenografts in mice, without harming embryo or mouse, respectively. Indole 5c was less toxic than CA-4 to endothelial cells, fibroblasts, and cardiomyocytes. In highly vascularized xenograft tumors 5c induced distinct discolorations and histological features typical of vascular-disrupting agents, such as disrupted vessel structures, hemorrhages, and extensive necrosis. In a first preliminary therapy trial, indole 5c retarded the growth of resistant xenograft tumors in mice. © 2016 Elsevier Science Ltd. All rights reserved.
Collapse
Affiliation(s)
- Katharina Mahal
- Organic Chemistry Laboratory, University Bayreuth, 95440 Bayreuth, Germany
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University Bayreuth, 95440 Bayreuth, Germany
| | | | - Marcus Resch
- Department of Molecular Structural Biology, Georg-August-University Göttingen, 37077 Göttingen, Germany
| | - Ralf Ficner
- Department of Molecular Structural Biology, Georg-August-University Göttingen, 37077 Göttingen, Germany
| | - Rainer Schobert
- Organic Chemistry Laboratory, University Bayreuth, 95440 Bayreuth, Germany.
| | - Thomas Mueller
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, 06120 Halle-Saale, Germany
| |
Collapse
|
11
|
Anticancer efficacy of unique pyridine-based tetraindoles. Eur J Med Chem 2015; 104:165-76. [DOI: 10.1016/j.ejmech.2015.09.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 09/16/2015] [Accepted: 09/24/2015] [Indexed: 02/06/2023]
|
12
|
Hwang DJ, Wang J, Li W, Miller DD. Structural Optimization of Indole Derivatives Acting at Colchicine Binding Site as Potential Anticancer Agents. ACS Med Chem Lett 2015; 6:993-7. [PMID: 26396686 DOI: 10.1021/acsmedchemlett.5b00208] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/06/2015] [Indexed: 12/18/2022] Open
Abstract
A new series of indole analogues based on our earlier lead compound, 2-(1H-indol-5-yl)-4-(3,4,5-trimethoxyphenyl)-1H-imidazo[4,5-c]pyridine (42), was prepared as tubulin inhibitors in an effort to find a molecule with improved cytotoxic potency and metabolic stability. A series of indolyl-imidazopyridines (IIP) were synthesized and exhibited potent tubulin polymerization inhibitory activity with potent IC50 values ranging from 3 to 175 nM against a panel of human melanoma and prostate cancer cell lines. Among these compounds, the 6-indolyl compound 43 showed improved cytotoxic potency (average IC50 of 9.75 nM vs 55.75 nM) and metabolic stability in human liver microsomes (half-life time was 56.3 min vs. 45.4 min) as compared to previously reported 42. It was also shown to be effective against P-glycoprotein (P-gp) mediated multiple drug resistance (MDR) and taxol resistance.
Collapse
Affiliation(s)
- Dong-Jin Hwang
- Department
of Pharmaceutical
Sciences, University of Tennessee, Health Science Center, Memphis, Tennessee 38163, United States
| | - Jin Wang
- Department
of Pharmaceutical
Sciences, University of Tennessee, Health Science Center, Memphis, Tennessee 38163, United States
| | - Wei Li
- Department
of Pharmaceutical
Sciences, University of Tennessee, Health Science Center, Memphis, Tennessee 38163, United States
| | - Duane D. Miller
- Department
of Pharmaceutical
Sciences, University of Tennessee, Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
13
|
Mahal K, Resch M, Ficner R, Schobert R, Biersack B, Mueller T. Effects of the tumor-vasculature-disrupting agent verubulin and two heteroaryl analogues on cancer cells, endothelial cells, and blood vessels. ChemMedChem 2014; 9:847-54. [PMID: 24678059 DOI: 10.1002/cmdc.201300531] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Indexed: 11/06/2022]
Abstract
Two analogues of the discontinued tumor vascular-disrupting agent verubulin (Azixa®, MPC-6827, 1) featuring benzo-1,4-dioxan-6-yl (compound 5 a) and N-methylindol-5-yl (compound 10) residues instead of the para-anisyl group on the 4-(methylamino)-2-methylquinazoline pharmacophore, were prepared and found to exceed the antitumor efficacy of the lead compound. They were antiproliferative with single-digit nanomolar IC50 values against a panel of nine tumor cell lines, while not affecting nonmalignant fibroblasts. Indole 10 surpassed verubulin in seven tumor cell lines including colon, breast, ovarian, and germ cell cancer cell lines. In line with docking studies indicating that compound 10 may bind the colchicine binding site of tubulin more tightly (Ebind =-9.8 kcal mol(-1) ) than verubulin (Ebind =-8.3 kcal mol(-1) ), 10 suppressed the formation of vessel-like tubes in endothelial cells and destroyed the blood vessels in the chorioallantoic membrane of fertilized chicken eggs at nanomolar concentrations. When applied to nude mice bearing a highly vascularized 1411HP germ cell xenograft tumor, compound 10 displayed pronounced vascular-disrupting effects that led to hemorrhages and extensive central necrosis in the tumor.
Collapse
Affiliation(s)
- Katharina Mahal
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth (Germany)
| | | | | | | | | | | |
Collapse
|
14
|
Indole molecules as inhibitors of tubulin polymerization: potential new anticancer agents. Future Med Chem 2013; 4:2085-115. [PMID: 23157240 DOI: 10.4155/fmc.12.141] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Agents that interfere with tubulin function have a broad anti-tumor spectrum and they represent one of the most significant classes of anticancer agents. In the past few years, several small synthetic molecules that have an indole nucleus as a core structure have been identified as tubulin inhibitors. Among these, several aroylindoles, arylthioindoles, diarylindoles and indolylglyoxyamides have shown good inhibition towards the tubulin polymerization. This article reviews the synthesis, biological activities and SARs of these main classes of indoles. Brief mention has also been made about the fused indole analogs as tubulin inhibitors.
Collapse
|
15
|
Orally bioavailable tubulin antagonists for paclitaxel-refractory cancer. Pharm Res 2012; 29:3053-63. [PMID: 22760659 DOI: 10.1007/s11095-012-0814-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 06/20/2012] [Indexed: 12/17/2022]
Abstract
PURPOSE To evaluate the efficacy and oral activity of two promising indoles, (2-(1H-indol-3-yl)-1H-imidazol-4-yl)(3,4,5-trimethoxyphenyl)methanone [compound II] and (2-(1H-indol-5-ylamino)-thiazol-4-yl)(3,4,5-trimethoxyphenyl)methanone [compound IAT], in paclitaxel- and docetaxel-resistant tumor models in vitro and in vivo. METHODS The in vitro drug-like properties, including potency, solubility, metabolic stability, and drug-drug interactions were examined for our two active compounds. An in vivo pharmacokinetic study and antitumor efficacy study were also completed to compare their efficacy with docetaxel. RESULTS Both compounds bound to the colchicine-binding site on tubulin, and inhibited tubulin polymerization, resulting in highly potent cytotoxic activity in vitro. While the potency of paclitaxel and docetaxel was compromised in a multidrug-resistant cell line that overexpresses P-glycoprotein, the potency of compounds II and IAT was maintained. Both compounds had favorable drug-like properties, and acceptable oral bioavailability (21-50 %) in mice, rats, and dogs. Tumor growth inhibition of greater than 100 % was achieved when immunodeficient mice with rapidly growing paclitaxel-resistant prostate cancer cells were treated orally at doses of 3-30 mg/kg of II or IAT. CONCLUSIONS These studies highlight the potent and broad anticancer activity of two orally bioavailable compounds, offering significant pharmacologic advantage over existing drugs of this class for multidrug resistant or taxane-refractory cancers.
Collapse
|
16
|
Li WS, Wang CH, Ko S, Chang TT, Jen YC, Yao CF, More SV, Jao SC. Synthesis and Evaluation of the Cytotoxicities of Tetraindoles: Observation that the 5-Hydroxy Tetraindole (SK228) Induces G2 Arrest and Apoptosis in Human Breast Cancer Cells. J Med Chem 2012; 55:1583-92. [DOI: 10.1021/jm2013425] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Wen-Shan Li
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Chie-Hong Wang
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Shengkai Ko
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
- Department of Chemistry, National Taiwan Normal University, Taipei 116, Taiwan
| | - Tzu Ting Chang
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Ya Ching Jen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Ching-Fa Yao
- Department of Chemistry, National Taiwan Normal University, Taipei 116, Taiwan
| | | | - Shu-Chuan Jao
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
17
|
Ahn S, Kearbey JD, Li CM, Duke CB, Miller DD, Dalton JT. Biotransformation of a novel antimitotic agent, I-387, by mouse, rat, dog, monkey, and human liver microsomes and in vivo pharmacokinetics in mice. Drug Metab Dispos 2011; 39:636-43. [PMID: 21233217 DOI: 10.1124/dmd.110.036673] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
3-(1H-Indol-2-yl)phenyl)(3,4,5-trimethoxyphenyl)methanone (I-387) is a novel indole compound with antitubulin action and potent antitumor activity in various preclinical models. I-387 avoids drug resistance mediated by P-glycoprotein and showed less neurotoxicity than vinca alkaloids during in vivo studies. We examined the pharmacokinetics and metabolism of I-387 in mice as a component of our preclinical development of this compound and continued interest in structure-activity relationships for antitubulin agents. After a 1 mg/kg intravenous dose, noncompartmental pharmacokinetic analysis in plasma showed that clearance (CL), volume of distribution at steady state (Vd(ss)), and terminal half-life (t(1/2)) of I-387 were 27 ml per min/kg, 5.3 l/kg, and 7 h, respectively. In the in vitro metabolic stability study, half-lives of I-387 were between 10 and 54 min by mouse, rat, dog, monkey, and human liver microsomes in the presence of NADPH, demonstrating interspecies variability. I-387 was most stable in rat liver microsomes and degraded quickly in monkey liver microsomes. Liquid chromatography-tandem mass spectrometry was used to identify phase I metabolites. Hydroxylation, reduction of a ketone group, and O-demethylation were the major metabolites formed by the liver microsomes of the five species. The carbonyl group of I-387 was reduced and identified as the most labile site in human liver microsomes. The results of these drug metabolism and pharmacokinetic studies provide the foundation for future structural modification of this pharmacophore to improve stability of drugs with potent anticancer effects in cancer patients.
Collapse
Affiliation(s)
- Sunjoo Ahn
- Division of Pharmaceutics, College of Pharmacy, Ohio State University, Columbus, Ohio, USA
| | | | | | | | | | | |
Collapse
|