1
|
Mahmoud Mohamed MM, Yang Z, Lum KY, Peschel G, Rosenbaum MA, Weber T, Coriani S, Gotfredsen CH, Ding L. Genome-Driven Discovery of Hygrocins in Streptomyces rapamycinicus. JOURNAL OF NATURAL PRODUCTS 2024; 87:1321-1329. [PMID: 38647518 DOI: 10.1021/acs.jnatprod.3c00767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Ansamycins, represented by the antituberculosis drug rifamycin, are an important family of natural products. To obtain new ansamycins, Streptomyces rapamycinicus IMET 43975 harboring an ansamycin biosynthetic gene cluster was fermented in a 50 L scale, and subsequent purification work led to the isolation of five known and four new analogues, where hygrocin W (2) belongs to benzoquinonoid ansamycins, and the other three hygrocins, hygrocins X-Z (6-8), are new seco-hygrocins. The structures of ansamycins (1-8) were determined by the analysis of spectroscopic (1D/2D NMR and ECD) and MS spectrometric data. The Baeyer-Villiger enzyme which catalyzed the ester formation in the ansa-ring was confirmed through in vivo CRISPR base editing. The discovery of these compounds further enriches the structural diversity of ansamycins.
Collapse
Affiliation(s)
- Manar Magdy Mahmoud Mohamed
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads 221, 2800 Kgs. Lyngby, Denmark
| | - Zhijie Yang
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads 221, 2800 Kgs. Lyngby, Denmark
| | - Kah Yean Lum
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads 221, 2800 Kgs. Lyngby, Denmark
| | - Gundela Peschel
- Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute (HKI), Beutenbergstraße 11a, 07745 Jena, Germany
| | - Miriam A Rosenbaum
- Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute (HKI), Beutenbergstraße 11a, 07745 Jena, Germany
| | - Tilmann Weber
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Sonia Coriani
- Department of Chemistry, Technical University of Denmark, Kemitorvet Building 207, 2800 Kgs. Lyngby, Denmark
| | - Charlotte H Gotfredsen
- Department of Chemistry, Technical University of Denmark, Kemitorvet Building 207, 2800 Kgs. Lyngby, Denmark
| | - Ling Ding
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads 221, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
2
|
Yang L, Li X, Zhao L, Hu W, Qian Y. Design, synthesis and bioactivity evaluation of novel monomethyl auristatin F analogues. Mol Divers 2024:10.1007/s11030-024-10873-1. [PMID: 38762686 DOI: 10.1007/s11030-024-10873-1] [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] [Received: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 05/20/2024]
Abstract
Monomethyl auristatin F (MMAF), a synthetic analogue of the natural compound dolastatin 10, has garnered significant attention in cancer research due to its high potency in vitro. While previous studies have focused on modifying the N-terminal extension of the amino group and the C-terminal modification of the carboxyl group, there has been limited exploration into modifying the P1 and P5 side chains. In this study, we substituted the valine residue at the P1 position with various natural or unnatural amino acids and introduced triazole functional groups at the P5 side chain. Compounds 11k and 18d exhibited excellent inhibition on tubulin. Additionally, compound 18d demonstrated enhanced cytotoxicity against HCT116 cells compared to the parent compound MMAF, suggesting its potential as a cytotoxic payload for further antibody-drug conjugates (ADCs) development.
Collapse
Affiliation(s)
- Lisheng Yang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xinglin Li
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lei Zhao
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Wenhao Hu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Yu Qian
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
3
|
Kaburagi Y, Kira K, Yahata K, Iso K, Sato Y, Matsuura F, Ohashi I, Matsumoto Y, Isomura M, Sasaki T, Fukuyama T, Miyashita Y, Azuma H, Iida D, Ishida T, Itano W, Matsuda M, Matsukura M, Murai N, Nagao S, Seki M, Yamamoto A, Yamamoto Y, Yoneda N, Watanabe Y, Kamada A, Kayano A, Tagami K, Asano O, Owa T, Kishi Y. Ten-Gram-Scale Total Synthesis of the Anticancer Drug Candidate E7130 to Supply Clinical Trials. Org Lett 2024; 26:2837-2842. [PMID: 38252895 DOI: 10.1021/acs.orglett.3c03663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
E7130 is a novel drug candidate with an exceedingly complex chemical structure of the halichondrin class, discovered by a total synthesis approach through joint research between the Kishi group at Harvard University and Eisai. Only 18 months after completion of the initial milligram-scale synthesis, ten-gram-scale synthesis of E7130 was achieved, providing the first good manufacturing practice (GMP) batch to supply clinical trials. This paper highlights the challenges in developing ten-gram-scale synthesis from the milligram-scale synthesis.
Collapse
Affiliation(s)
- Yosuke Kaburagi
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Kazunobu Kira
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Kenzo Yahata
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Kentaro Iso
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Yuki Sato
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Fumiyoshi Matsuura
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Isao Ohashi
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Yasunobu Matsumoto
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Minetaka Isomura
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Takeo Sasaki
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Takashi Fukuyama
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Yusuke Miyashita
- Kashima Plant, Eisai Co., Ltd., 22 Sunayama, Kamisu-shi, Ibaraki 314-0255, Japan
| | - Hiroshi Azuma
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Daisuke Iida
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Tasuku Ishida
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Wataru Itano
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Masaaki Matsuda
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Masayuki Matsukura
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Norio Murai
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Satoshi Nagao
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Masashi Seki
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Akihiko Yamamoto
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Yuji Yamamoto
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Naoki Yoneda
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Yuzo Watanabe
- Kashima Plant, Eisai Co., Ltd., 22 Sunayama, Kamisu-shi, Ibaraki 314-0255, Japan
| | - Atsushi Kamada
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Akio Kayano
- Kashima Plant, Eisai Co., Ltd., 22 Sunayama, Kamisu-shi, Ibaraki 314-0255, Japan
| | - Katsuya Tagami
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Osamu Asano
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Takashi Owa
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Yoshito Kishi
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
4
|
Yu Q, Zhou Y, Gao X, Pan S, Lin F, Li W. Gram-Scale Synthesis of the C14–C23 Fragment of Eribulin. Org Process Res Dev 2023. [DOI: 10.1021/acs.oprd.2c00370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Affiliation(s)
- Qiuhan Yu
- Department of Medicinal Chemistry School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P. R. of China
| | - Yueer Zhou
- Department of Medicinal Chemistry School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P. R. of China
| | - Xinai Gao
- Department of Medicinal Chemistry School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P. R. of China
| | - Shuheng Pan
- Department of Medicinal Chemistry School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P. R. of China
| | - Feng Lin
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, P. R. of China
| | - Wei Li
- Department of Medicinal Chemistry School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P. R. of China
| |
Collapse
|
5
|
Alpízar-Pedraza D, Veulens ADLN, Araujo EC, Piloto-Ferrer J, Sánchez-Lamar Á. Microtubules destabilizing agents binding sites in tubulin. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132723] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
6
|
Boichuk S, Syuzov K, Bikinieva F, Galembikova A, Zykova S, Gankova K, Igidov S, Igidov N. Computational-Based Discovery of the Anti-Cancer Activities of Pyrrole-Based Compounds Targeting the Colchicine-Binding Site of Tubulin. Molecules 2022; 27:2873. [PMID: 35566235 PMCID: PMC9101527 DOI: 10.3390/molecules27092873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 12/10/2022] Open
Abstract
Despite the tubulin-binding agents (TBAs) that are widely used in the clinic for cancer therapy, tumor resistance to TBAs (both inherited and acquired) significantly impairs their effectiveness, thereby decreasing overall survival (OS) and progression-free survival (PFS) rates, especially for the patients with metastatic, recurrent, and unresectable forms of the disease. Therefore, the development of novel effective drugs interfering with the microtubules' dynamic state remains a big challenge in current oncology. We report here about the novel ethyl 2-amino-1-(furan-2-carboxamido)-5-(2-aryl/tert-butyl-2-oxoethylidene)-4-oxo-4,5-dihydro-1H-pyrrole-3-carboxylates (EAPCs) exhibiting potent anti-cancer activities against the breast and lung cancer cell lines in vitro. This was due to their ability to inhibit tubulin polymerization and induce cell cycle arrest in M-phase. As an outcome, the EAPC-treated cancer cells exhibited a significant increase in apoptosis, which was evidenced by the expression of cleaved forms of PARP, caspase-3, and increased numbers of Annexin-V-positive cells. By using the in silico molecular modeling methods (e.g., induced-fit docking, binding metadynamics, and unbiased molecular dynamics), we found that EAPC-67 and -70 preferentially bind to the colchicine-binding site of tubulin. Lastly, we have shown that the EAPCs indicated above and colchicine utilizes a similar molecular mechanism to inhibit tubulin polymerization via targeting the T7 loop in the β-chain of tubulin, thereby preventing the conformational changes in the tubulin dimers required for their polymerization. Collectively, we identified the novel and potent TBAs that bind to the colchicine-binding site and disrupt the microtubule network. As a result of these events, the compounds induced a robust cell cycle arrest in M-phase and exhibited potent pro-apoptotic activities against the epithelial cancer cell lines in vitro.
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
- Department of Radiotherapy and Radiology, Russian Medical Academy of Continuous Professional Education, 125993 Moscow, Russia
- Biologically Active Terpenoids Laboratory, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Kirill Syuzov
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
| | - Firuza Bikinieva
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
| | - Svetlana Zykova
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| | - Ksenia Gankova
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| | - Sergei Igidov
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| | - Nazim Igidov
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| |
Collapse
|
7
|
Computational-Based Discovery of the Anti-Cancer Activities of Pyrrole-Based Compounds Targeting the Colchicine-Binding Site of Tubulin. Molecules 2022. [PMID: 35566235 DOI: 10.3390/molecules27092873.(] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Despite the tubulin-binding agents (TBAs) that are widely used in the clinic for cancer therapy, tumor resistance to TBAs (both inherited and acquired) significantly impairs their effectiveness, thereby decreasing overall survival (OS) and progression-free survival (PFS) rates, especially for the patients with metastatic, recurrent, and unresectable forms of the disease. Therefore, the development of novel effective drugs interfering with the microtubules' dynamic state remains a big challenge in current oncology. We report here about the novel ethyl 2-amino-1-(furan-2-carboxamido)-5-(2-aryl/tert-butyl-2-oxoethylidene)-4-oxo-4,5-dihydro-1H-pyrrole-3-carboxylates (EAPCs) exhibiting potent anti-cancer activities against the breast and lung cancer cell lines in vitro. This was due to their ability to inhibit tubulin polymerization and induce cell cycle arrest in M-phase. As an outcome, the EAPC-treated cancer cells exhibited a significant increase in apoptosis, which was evidenced by the expression of cleaved forms of PARP, caspase-3, and increased numbers of Annexin-V-positive cells. By using the in silico molecular modeling methods (e.g., induced-fit docking, binding metadynamics, and unbiased molecular dynamics), we found that EAPC-67 and -70 preferentially bind to the colchicine-binding site of tubulin. Lastly, we have shown that the EAPCs indicated above and colchicine utilizes a similar molecular mechanism to inhibit tubulin polymerization via targeting the T7 loop in the β-chain of tubulin, thereby preventing the conformational changes in the tubulin dimers required for their polymerization. Collectively, we identified the novel and potent TBAs that bind to the colchicine-binding site and disrupt the microtubule network. As a result of these events, the compounds induced a robust cell cycle arrest in M-phase and exhibited potent pro-apoptotic activities against the epithelial cancer cell lines in vitro.
Collapse
|
8
|
Suliman RS, Alghamdi SS, Ali R, Rahman I, Alqahtani T, Frah IK, Aljatli DA, Huwaizi S, Algheribe S, Alehaideb Z, Islam I. Distinct Mechanisms of Cytotoxicity in Novel Nitrogenous Heterocycles: Future Directions for a New Anti-Cancer Agent. Molecules 2022; 27:molecules27082409. [PMID: 35458609 PMCID: PMC9029529 DOI: 10.3390/molecules27082409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/30/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
Electron-rich, nitrogenous heteroaromatic compounds interact more with biological/cellular components than their non-nitrogenous counterparts. The strong intermolecular interactions with proteins, enzymes, and receptors confer significant biological and therapeutic properties to the imidazole derivatives, giving rise to a well-known and extensively used range of therapeutic drugs used for infections, inflammation, and cancer, to name a few. The current study investigates the anti-cancer properties of fourteen previously synthesized nitrogenous heterocycles, derivatives of imidazole and oxazolone, on a panel of cancer cell lines and, in addition, predicts the molecular interactions, pharmacokinetic and safety profiles of these compounds. Method: The MTT and CellTiter-Glo® assays were used to screen the imidazole and oxazolone derivatives on six cancer cell lines: HL60, MDA-MB-321, KAIMRC1, KMIRC2, MCF-10A, and HCT8. Subsequently, in vitro tubulin staining and imaging were performed, and the level of apoptosis was measured using the Promega ApoTox-Glo® triplex assay. Furthermore, several computational tools were utilized to investigate the pharmacokinetics and safety profile, including PASS Online, SEA Search, the QikProp tool, SwissADME, ProTox-II, and an in silico molecular docking study on tubulin to identify the critical molecular interactions. Results: In vitro analysis identified compounds 8 and 9 to possess the most significant potent cytotoxic activity on the HL60 and MDA-MB-231 cell lines, supported by PASS Online anti-cancer predictions with pa scores of 0.413 and 0.434, respectively. In addition, compound 9 induced caspase 3/7 dependent-apoptosis and interfered with tubulin polymerization in the MDA-MB-231 cell line, consistent with in silico docking results, identifying binding similarity to the native ligand colchicine. All the derivatives, including compounds 8 and 9, had acceptable pharmacokinetics; however, the safety profile was suboptimal for all the tested derivates except compound 4. Conclusion: The imidazole derivative compound 9 is a promising anti-cancer agent that switches on caspase-dependent apoptotic cell death and modulates microtubule function. Therefore, it could be a lead compound for further drug optimization and development.
Collapse
Affiliation(s)
- Rasha Saad Suliman
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia; (R.A.); (T.A.); (I.K.F.); (D.A.A.)
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh 14811, Saudi Arabia; (S.H.); (S.A.); (Z.A.); (I.I.)
- Correspondence: (R.S.S.); (S.S.A.); Tel.: +966-(11)-429-9570 (R.S.S.); +966-(11)-429-9516 (S.S.A.)
| | - Sahar Saleh Alghamdi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia; (R.A.); (T.A.); (I.K.F.); (D.A.A.)
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh 14811, Saudi Arabia; (S.H.); (S.A.); (Z.A.); (I.I.)
- Correspondence: (R.S.S.); (S.S.A.); Tel.: +966-(11)-429-9570 (R.S.S.); +966-(11)-429-9516 (S.S.A.)
| | - Rizwan Ali
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia; (R.A.); (T.A.); (I.K.F.); (D.A.A.)
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh 14811, Saudi Arabia; (S.H.); (S.A.); (Z.A.); (I.I.)
| | - Ishrat Rahman
- Department of Basic Dental Sciences, College of Dentistry, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Tariq Alqahtani
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia; (R.A.); (T.A.); (I.K.F.); (D.A.A.)
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh 14811, Saudi Arabia; (S.H.); (S.A.); (Z.A.); (I.I.)
| | - Ibrahim K. Frah
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia; (R.A.); (T.A.); (I.K.F.); (D.A.A.)
| | - Dimah A. Aljatli
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia; (R.A.); (T.A.); (I.K.F.); (D.A.A.)
| | - Sarah Huwaizi
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh 14811, Saudi Arabia; (S.H.); (S.A.); (Z.A.); (I.I.)
| | - Shatha Algheribe
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh 14811, Saudi Arabia; (S.H.); (S.A.); (Z.A.); (I.I.)
| | - Zeyad Alehaideb
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh 14811, Saudi Arabia; (S.H.); (S.A.); (Z.A.); (I.I.)
| | - Imadul Islam
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh 14811, Saudi Arabia; (S.H.); (S.A.); (Z.A.); (I.I.)
| |
Collapse
|
9
|
Khademi Z, Heravi MM. Applications of Claisen condensations in total synthesis of natural products. An old reaction, a new perspective. Tetrahedron 2022. [DOI: 10.1016/j.tet.2021.132573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
10
|
Szabó G, Schulz F, Manzano-Marín A, Toenshoff ER, Horn M. Evolutionarily recent dual obligatory symbiosis among adelgids indicates a transition between fungus- and insect-associated lifestyles. THE ISME JOURNAL 2022; 16:247-256. [PMID: 34294881 PMCID: PMC8692619 DOI: 10.1038/s41396-021-01056-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023]
Abstract
Adelgids (Insecta: Hemiptera: Adelgidae) form a small group of insects but harbor a surprisingly diverse set of bacteriocyte-associated endosymbionts, which suggest multiple replacement and acquisition of symbionts over evolutionary time. Specific pairs of symbionts have been associated with adelgid lineages specialized on different secondary host conifers. Using a metagenomic approach, we investigated the symbiosis of the Adelges laricis/Adelges tardus species complex containing betaproteobacterial ("Candidatus Vallotia tarda") and gammaproteobacterial ("Candidatus Profftia tarda") symbionts. Genomic characteristics and metabolic pathway reconstructions revealed that Vallotia and Profftia are evolutionary young endosymbionts, which complement each other's role in essential amino acid production. Phylogenomic analyses and a high level of genomic synteny indicate an origin of the betaproteobacterial symbiont from endosymbionts of Rhizopus fungi. This evolutionary transition was accompanied with substantial loss of functions related to transcription regulation, secondary metabolite production, bacterial defense mechanisms, host infection, and manipulation. The transition from fungus to insect endosymbionts extends our current framework about evolutionary trajectories of host-associated microbes.
Collapse
Affiliation(s)
- Gitta Szabó
- Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria.
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary.
| | - Frederik Schulz
- Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
- US Department of Energy (DOE) Joint Genome Institute, Berkeley, CA, USA
| | - Alejandro Manzano-Marín
- Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Elena Rebecca Toenshoff
- Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
| | - Matthias Horn
- Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| |
Collapse
|
11
|
Wordeman L, Vicente JJ. Microtubule Targeting Agents in Disease: Classic Drugs, Novel Roles. Cancers (Basel) 2021; 13:5650. [PMID: 34830812 PMCID: PMC8616087 DOI: 10.3390/cancers13225650] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Microtubule-targeting agents (MTAs) represent one of the most successful first-line therapies prescribed for cancer treatment. They interfere with microtubule (MT) dynamics by either stabilizing or destabilizing MTs, and in culture, they are believed to kill cells via apoptosis after eliciting mitotic arrest, among other mechanisms. This classical view of MTA therapies persisted for many years. However, the limited success of drugs specifically targeting mitotic proteins, and the slow growing rate of most human tumors forces a reevaluation of the mechanism of action of MTAs. Studies from the last decade suggest that the killing efficiency of MTAs arises from a combination of interphase and mitotic effects. Moreover, MTs have also been implicated in other therapeutically relevant activities, such as decreasing angiogenesis, blocking cell migration, reducing metastasis, and activating innate immunity to promote proinflammatory responses. Two key problems associated with MTA therapy are acquired drug resistance and systemic toxicity. Accordingly, novel and effective MTAs are being designed with an eye toward reducing toxicity without compromising efficacy or promoting resistance. Here, we will review the mechanism of action of MTAs, the signaling pathways they affect, their impact on cancer and other illnesses, and the promising new therapeutic applications of these classic drugs.
Collapse
Affiliation(s)
| | - Juan Jesus Vicente
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195, USA;
| |
Collapse
|
12
|
Misra SK, Pathak K. Naturally occurring heterocyclic anticancer compounds. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2021-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Naturally occurring heterocyclic scaffolds are key ingredients for the development of various therapeutics employed for biomedical applications. Heterocyclic pharmacophores are widely disseminated and have been befallen in almost all categories of drugs for the alleviation of myriad ailments including diabetes, neurodegenerative, psychiatric, microbial infections, disastrous cancers etc. Countless fused heterocyclic anticancerous templates are reported to display antimetabolite, antioxidant, antiproliferative, cytostatic etc. pharmacological actions via targeting different signaling pathways (cell cycle, PI-3kinase/Akt, p53, caspase extrinsic pathway etc.), overexpressive receptors (EGRF, HER2, EGF, VEGF etc.) and physiological enzymes (topoisomerase I and II, cyclin dependent kinase etc.). A compiled description on various natural sources (plants, microbes, marine) containing anticancer agents comprising heterocyclic ring specified with presence of nitrogen (vincristine, vinblastine, indole-3-carbinol, meridianins, piperine, lamellarins etc.), oxygen (paclitaxel, halichondrin B, quercetin, myricetin, kaempferol etc.) and sulphur atoms (brugine, fucoidan, carrageenan etc.) are displayed here along with their molecular level cytotoxic action and therapeutic applications.
Collapse
Affiliation(s)
- Shashi Kiran Misra
- University Institute of Pharmacy, Chhatrapati Shahu Ji Maharaj University , Kanpur , 208026 , India
| | - Kamla Pathak
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences , Saifai , Etawah , 206130 , Uttar Pradesh , India
| |
Collapse
|
13
|
Mertansine Inhibits mRNA Expression and Enzyme Activities of Cytochrome P450s and Uridine 5′-Diphospho-Glucuronosyltransferases in Human Hepatocytes and Liver Microsomes. Pharmaceutics 2020; 12:pharmaceutics12030220. [PMID: 32131538 PMCID: PMC7150891 DOI: 10.3390/pharmaceutics12030220] [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: 02/18/2020] [Revised: 02/28/2020] [Accepted: 03/01/2020] [Indexed: 11/16/2022] Open
Abstract
Mertansine, a tubulin inhibitor, is used as the cytotoxic component of antibody–drug conjugates (ADCs) for cancer therapy. The effects of mertansine on uridine 5′-diphospho-glucuronosyltransferase (UGT) activities in human liver microsomes and its effects on the mRNA expression of cytochrome P450s (CYPs) and UGTs in human hepatocytes were evaluated to assess the potential for drug–drug interactions (DDIs). Mertansine potently inhibited UGT1A1-catalyzed SN-38 glucuronidation, UGT1A3-catalyzed chenodeoxycholic acid 24-acyl-β-glucuronidation, and UGT1A4-catalyzed trifluoperazine N-β-d-glucuronidation, with Ki values of 13.5 µM, 4.3 µM, and 21.2 µM, respectively, but no inhibition of UGT1A6, UGT1A9, and UGT2B7 enzyme activities was observed in human liver microsomes. A 48 h treatment of mertansine (1.25–2500 nM) in human hepatocytes resulted in the dose-dependent suppression of mRNA levels of CYP1A2, CYP2B6, CYP3A4, CYP2C8, CYP2C9, CYP2C19, UGT1A1, and UGT1A9, with IC50 values of 93.7 ± 109.1, 36.8 ± 18.3, 160.6 ± 167.4, 32.1 ± 14.9, 578.4 ± 452.0, 539.5 ± 233.4, 856.7 ± 781.9, and 54.1 ± 29.1 nM, respectively, and decreased the activities of CYP1A2-mediated phenacetin O-deethylase, CYP2B6-mediated bupropion hydroxylase, and CYP3A4-mediated midazolam 1′-hydroxylase. These in vitro DDI potentials of mertansine with CYP1A2, CYP2B6, CYP2C8/9/19, CYP3A4, UGT1A1, and UGT1A9 substrates suggest that it is necessary to carefully characterize the DDI potentials of ADC candidates with mertansine as a payload in the clinic.
Collapse
|
14
|
Malik MS, Ahmed SA, Althagafi II, Ansari MA, Kamal A. Application of triazoles as bioisosteres and linkers in the development of microtubule targeting agents. RSC Med Chem 2020; 11:327-348. [PMID: 33479639 PMCID: PMC7580775 DOI: 10.1039/c9md00458k] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
The triazole ring system has emerged as an exciting prospect in the optimization studies of promising lead molecules in the quest for new drugs for clinical usage. Several marketed drugs possess these versatile moieties that are used in a wide range of medical indications. This stems from the unique intrinsic properties of triazoles, which impart stability to the basic pharmacophoric unit with an added advantage of being a bioisostere of different chemical functionalities. In the last decade, the use of triazoles as bioisosteres and linkers in the development of microtubule targeting agents has been extensively investigated. The present review highlights the advances in this promising area of drug discovery and development.
Collapse
Affiliation(s)
- M Shaheer Malik
- Department of Chemistry , Faculty of Applied Sciences , Umm Al-Qura University , 21955 Makkah , Saudi Arabia . ;
- Central Research Laboratories , Faculty of Applied Sciences , Umm Al-Qura University , 21955 Makkah , Saudi Arabia
| | - Saleh A Ahmed
- Department of Chemistry , Faculty of Applied Sciences , Umm Al-Qura University , 21955 Makkah , Saudi Arabia . ;
- Central Research Laboratories , Faculty of Applied Sciences , Umm Al-Qura University , 21955 Makkah , Saudi Arabia
- Chemistry Department , Faculty of Science , Assiut University , 71516 Assiut , Egypt
| | - Ismail I Althagafi
- Department of Chemistry , Faculty of Applied Sciences , Umm Al-Qura University , 21955 Makkah , Saudi Arabia . ;
- Central Research Laboratories , Faculty of Applied Sciences , Umm Al-Qura University , 21955 Makkah , Saudi Arabia
| | - Mohammed Azam Ansari
- Department of Epidemic Disease Research , Institute of Research and Medical Consultation , Imam AbdurRahman Bin Faisal University , 34212 Dammam , Saudi Arabia
| | - Ahmed Kamal
- School of Pharmaceutical Education and Research (SPER) , Jamia Hamdard , New Delhi-110062 , India . ; ; Tel: +91 11 26059665
| |
Collapse
|
15
|
Agarwal G, Carcache PJB, Addo EM, Kinghorn AD. Current status and contemporary approaches to the discovery of antitumor agents from higher plants. Biotechnol Adv 2020; 38:107337. [PMID: 30633954 PMCID: PMC6614024 DOI: 10.1016/j.biotechadv.2019.01.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 01/03/2019] [Accepted: 01/07/2019] [Indexed: 12/13/2022]
Abstract
Higher plant constituents have afforded clinically available anticancer drugs. These include both chemically unmodified small molecules and their synthetic derivatives currently used or those in clinical trials as antineoplastic agents, and an updated summary is provided. In addition, botanical dietary supplements, exemplified by mangosteen and noni constituents, are also covered as potential cancer chemotherapeutic agents. Approaches to metabolite purification, rapid dereplication, and biological evaluation including analytical hyphenated techniques, molecular networking, and advanced cellular and animal models are discussed. Further, enhanced and targeted drug delivery systems for phytochemicals, including micelles, nanoparticles and antibody drug conjugates (ADCs) are described herein.
Collapse
Affiliation(s)
- Garima Agarwal
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Peter J Blanco Carcache
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Ermias Mekuria Addo
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
16
|
Yaghoubi S, Karimi MH, Lotfinia M, Gharibi T, Mahi-Birjand M, Kavi E, Hosseini F, Sineh Sepehr K, Khatami M, Bagheri N, Abdollahpour-Alitappeh M. Potential drugs used in the antibody-drug conjugate (ADC) architecture for cancer therapy. J Cell Physiol 2019; 235:31-64. [PMID: 31215038 DOI: 10.1002/jcp.28967] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 05/20/2019] [Indexed: 01/04/2023]
Abstract
Cytotoxic small-molecule drugs have a major influence on the fate of antibody-drug conjugates (ADCs). An ideal cytotoxic agent should be highly potent, remain stable while linked to ADCs, kill the targeted tumor cell upon internalization and release from the ADCs, and maintain its activity in multidrug-resistant tumor cells. Lessons learned from successful and failed experiences in ADC development resulted in remarkable progress in the discovery and development of novel highly potent small molecules. A better understanding of such small-molecule drugs is important for development of effective ADCs. The present review discusses requirements making a payload appropriate for antitumor ADCs and focuses on the main characteristics of commonly-used cytotoxic payloads that showed acceptable results in clinical trials. In addition, the present study represents emerging trends and recent advances of payloads used in ADCs currently under clinical trials.
Collapse
Affiliation(s)
- Sajad Yaghoubi
- Department of Clinical Microbiology, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Majid Lotfinia
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran.,Core Research Lab, Kashan University of Medical Sciences, Kashan, Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Motahare Mahi-Birjand
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Esmaeil Kavi
- Department of Nursing, School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Fahimeh Hosseini
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Koushan Sineh Sepehr
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Khatami
- NanoBioelectrochemistry Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | |
Collapse
|
17
|
A landmark in drug discovery based on complex natural product synthesis. Sci Rep 2019; 9:8656. [PMID: 31209263 PMCID: PMC6572832 DOI: 10.1038/s41598-019-45001-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Despite their outstanding antitumour activity in mice, the limited supply from the natural sources has prevented drug discovery/development based on intact halichondrins. We achieved a total synthesis of C52-halichondrin-B amine (E7130) on a >10 g scale with >99.8% purity under GMP conditions. Interestingly, E7130 not only is a novel microtubule dynamics inhibitor but can also increase intratumoural CD31-positive endothelial cells and reduce α-SMA-positive cancer-associated fibroblasts at pharmacologically relevant compound concentrations. According to these unique effects, E7130 significantly augment the effect of antitumour treatments in mouse models and is currently in a clinical trial. Overall, our work demonstrates that a total synthesis can address the issue of limited material supply in drug discovery/development even for the cases of complex natural products.
Collapse
|
18
|
Taplin S, Vashisht K, Walles M, Calise D, Kluwe W, Bouchard P, Johnson R. Hepatotoxicity with antibody maytansinoid conjugates: A review of preclinical and clinical findings. J Appl Toxicol 2018; 38:600-615. [DOI: 10.1002/jat.3582] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Sarah Taplin
- Novartis Pharmaceuticals Inc.; East Hanover NJ USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Dicitrinone D, an antimitotic polyketide isolated from the marine-derived fungus Penicillium citrinum. Tetrahedron 2017. [DOI: 10.1016/j.tet.2017.08.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
20
|
Rossington SB, Hadfield JA, Shnyder SD, Wallace TW, Williams KJ. Tubulin-binding dibenz[c,e]oxepines: Part 2. Structural variation and biological evaluation as tumour vasculature disrupting agents. Bioorg Med Chem 2017; 25:1630-1642. [PMID: 28143677 DOI: 10.1016/j.bmc.2017.01.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 02/07/2023]
Abstract
5,7-Dihydro-3,9,10,11-tetramethoxybenz[c,e]oxepin-4-ol 1, prepared from a dibenzyl ether precursor via Pd-catalysed intramolecular direct arylation, possesses broad-spectrum in vitro cytotoxicity towards various tumour cell lines, and induces vascular shutdown, necrosis and growth delay in tumour xenografts in mice at sub-toxic doses. The biological properties of 1 and related compounds can be attributed to their ability to inhibit microtubule assembly at the micromolar level, by binding reversibly to the same site of the tubulin αβ-heterodimer as colchicine 2 and the allocolchinol, N-acetylcolchinol 4.
Collapse
Affiliation(s)
- Steven B Rossington
- School of Chemistry, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - John A Hadfield
- School of Environment and Life Sciences, University of Salford, Salford M5 4WT, UK
| | - Steven D Shnyder
- Institute of Cancer Therapeutics, University of Bradford, Richmond Road, Bradford BD7 1DP, UK
| | - Timothy W Wallace
- School of Chemistry, University of Manchester, Oxford Road, Manchester M13 9PL, UK.
| | - Kaye J Williams
- Manchester Pharmacy School, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
21
|
Olziersky AM, Labidi-Galy SI. Clinical Development of Anti-mitotic Drugs in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1002:125-152. [PMID: 28600785 DOI: 10.1007/978-3-319-57127-0_6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mitosis is one of the most fundamental processes of life by which a mammalian cell divides into two daughter cells. Mitosis has been an attractive target for anticancer therapies since fast proliferation was identified as one of the hallmarks of cancer cells. Despite efforts into developing specific inhibitors for mitotic kinases and kinesins, very few drugs have shown the efficiency of microtubule targeting-agents in cancer cells with paclitaxel being the most successful. A deeper translational research accompanying clinical trials of anti-mitotic drugs will help in identifying potent biomarkers predictive for response. Here, we review the current knowledge of mitosis targeting agents that have been tested so far in the clinics.
Collapse
Affiliation(s)
- Anna-Maria Olziersky
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - S Intidhar Labidi-Galy
- Department of Oncology, Geneva University Hospitals, Rue Gabrielle Perret-Gentil 4, Geneva, 1205, Switzerland.
| |
Collapse
|
22
|
Antúnez-Mojica M, Rodríguez-Salarichs J, Redondo-Horcajo M, León A, Barasoain I, Canales Á, Cañada FJ, Jiménez-Barbero J, Alvarez L, Díaz JF. Structural and Biochemical Characterization of the Interaction of Tubulin with Potent Natural Analogues of Podophyllotoxin. JOURNAL OF NATURAL PRODUCTS 2016; 79:2113-2121. [PMID: 27518758 DOI: 10.1021/acs.jnatprod.6b00428] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Four natural analogues of podophyllotoxin obtained from the Mexican medicinal plant Bursera fagaroides, namely, acetyl podophyllotoxin (2), 5'-desmethoxy-β-peltatin A methyl ether (3), 7',8'-dehydro acetyl podophyllotoxin (4), and burseranin (5), have been characterized, and their interactions with tubulin have been investigated. Cytotoxic activity measurements, followed by immunofluorescence microscopy and flow cytometry studies, demonstrated that these compounds disrupt microtubule networks in cells and cause cell cycle arrest in the G2/M phase in the A549 cell line. A tubulin binding assay showed that compounds 1-4 were potent assembly inhibitors, displaying binding to the colchicine site with Kb values ranging from 11.75 to 185.0 × 10(5) M(-1). In contrast, burseranin (5) was not able to inhibit tubulin assembly. From the structural perspective, the ligand-binding epitopes of compounds 1-3 have been mapped using STD-NMR, showing that B and E rings are the major points for interaction with the protein. The obtained results indicate that the inhibition of tubulin assembly of this family of compounds is more effective when there are at least two methoxyl groups at the E ring, along with a trans configuration of the lactone ring in the aryltetralin lignan core.
Collapse
Affiliation(s)
- Mayra Antúnez-Mojica
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos , Cuernavaca, Morelos 62209, México
| | - Javier Rodríguez-Salarichs
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas , Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Mariano Redondo-Horcajo
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas , Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Alejandra León
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos , Cuernavaca, Morelos 62209, México
| | - Isabel Barasoain
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas , Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Ángeles Canales
- Departamento de Química Orgánica I, Facultad Ciencias Químicas, Universidad Complutense de Madrid , Avenida Complutense s/n, 28040 Madrid, Spain
| | - F J Cañada
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas , Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE Parque Tecnológico de Bizkaia, Edif. 801A-1°, 48160 Derio-Bizkaia, Spain, and Ikerbasque, Basque Foundation for Science , Maria Diaz de Haro 3, 48009 Bilbao, Spain
| | - Laura Alvarez
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos , Cuernavaca, Morelos 62209, México
| | - J Fernando Díaz
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas , Ramiro de Maeztu 9, 28040 Madrid, Spain
| |
Collapse
|
23
|
Waight AB, Bargsten K, Doronina S, Steinmetz MO, Sussman D, Prota AE. Structural Basis of Microtubule Destabilization by Potent Auristatin Anti-Mitotics. PLoS One 2016; 11:e0160890. [PMID: 27518442 PMCID: PMC4982639 DOI: 10.1371/journal.pone.0160890] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/26/2016] [Indexed: 12/29/2022] Open
Abstract
The auristatin class of microtubule destabilizers are highly potent cytotoxic agents against several cancer cell types when delivered as antibody drug conjugates. Here we describe the high resolution structures of tubulin in complex with both monomethyl auristatin E and F and unambiguously define the trans-configuration of both ligands at the Val-Dil amide bond in their tubulin bound state. Moreover, we illustrate how peptidic vinca-site agents carrying terminal carboxylate residues may exploit an observed extended hydrogen bond network with the M-loop Arg278 to greatly improve the affinity of the corresponding analogs and to maintain the M-loop in an incompatible conformation for productive lateral tubulin-tubulin contacts in microtubules. Our results highlight a potential, previously undescribed molecular mechanism by which peptidic vinca-site agents maintain unparalleled potency as microtubule-destabilizing agents.
Collapse
Affiliation(s)
- Andrew B. Waight
- Department of Protein Sciences, Seattle Genetics, Inc., Bothell, WA, United States of America
| | - Katja Bargsten
- Department of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institut, Villigen PSI, Switzerland
| | - Svetlana Doronina
- Department of Protein Sciences, Seattle Genetics, Inc., Bothell, WA, United States of America
| | - Michel O. Steinmetz
- Department of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institut, Villigen PSI, Switzerland
| | - Django Sussman
- Department of Protein Sciences, Seattle Genetics, Inc., Bothell, WA, United States of America
- * E-mail: (DS); (AEP)
| | - Andrea E. Prota
- Department of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institut, Villigen PSI, Switzerland
- * E-mail: (DS); (AEP)
| |
Collapse
|
24
|
Aseyev O, Ribeiro JM, Cardoso F. Review on the clinical use of eribulin mesylate for the treatment of breast cancer. Expert Opin Pharmacother 2016; 17:589-600. [DOI: 10.1517/14656566.2016.1146683] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
25
|
Helfrich EJN, Piel J. Biosynthesis of polyketides by trans-AT polyketide synthases. Nat Prod Rep 2016; 33:231-316. [DOI: 10.1039/c5np00125k] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review discusses the biosynthesis of natural products that are generated bytrans-AT polyketide synthases, a family of catalytically versatile enzymes that represents one of the major group of proteins involved in the production of bioactive polyketides.
Collapse
Affiliation(s)
- Eric J. N. Helfrich
- Institute of Microbiology
- Eidgenössische Technische Hochschule (ETH) Zurich
- 8093 Zurich
- Switzerland
| | - Jörn Piel
- Institute of Microbiology
- Eidgenössische Technische Hochschule (ETH) Zurich
- 8093 Zurich
- Switzerland
| |
Collapse
|
26
|
Kiba T, Morii N, Takahashi H, Ozaki S, Atsumi M, Masumoto F, Shitakubo Y, Yamashiro H. Examination of the clinical efficacy of eribulin and trastuzumab in HER2-positive recurrent breast cancer. Mol Clin Oncol 2015; 4:47-50. [PMID: 26870356 DOI: 10.3892/mco.2015.668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/26/2015] [Indexed: 11/05/2022] Open
Abstract
There are limited studies reported that describe the efficacy of eribulin and trastuzumab in the treatment of recurrent breast cancer. The present study examined the therapeutic efficacy of eribulin and trastuzumab in the treatment of recurrent breast cancer. Between October 2011 and August 2013, 5 recurrent breast cancer patients who were treated with eribulin and trastuzumab were included in the study. The cancer stages in the 5 women who received this regimen were stage IIIB in 1 (20%) and stage IV in 4 (80%). The sites of recurrence were the lung in 3 patients, liver in 2, bone in 1, brain in 1, supraclavicular lymph nodes in 1, infraclavicular lymph nodes in 1 and mediastinal lymph nodes in 1. The median number of prior treatment regimens was 5 (range, 5-11). Complete response was achieved in 0 patients, 1 achieved partial response, 3 had stable disease, and 1 had progressive disease. The overall response rate was 20%, and the clinical benefit rate was 80%. Patients also reported grade 3/4 neutropenia (80.0%). However, hematological toxicity was reversible and manageable. The most common grade 3/4 nonhematological toxicities were fatigue (20.0%), peripheral neuropathy (20.0%) and appetite loss (20.0%). No patients withdrew from treatment, and favorable compliance was achieved in the study. The results indicated that eribulin and trastuzumab have the potential to be one of the drugs for treatment of recurrent breast cancer.
Collapse
Affiliation(s)
- Takayoshi Kiba
- Division of Modern Medical Technology, Institute for Clinical Research, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima 737-0023, Japan
| | - Nao Morii
- Department of Breast Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima 737-0023, Japan; Department of Breast Surgery, Tenri Hospital, Tenri, Nara 632-8552, Japan
| | - Hirotoshi Takahashi
- Department of Breast Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima 737-0023, Japan
| | - Shinji Ozaki
- Department of Breast Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima 737-0023, Japan
| | - Misao Atsumi
- Clinical Trial Management Office, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima 737-0023, Japan
| | - Fumi Masumoto
- Clinical Trial Management Office, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima 737-0023, Japan
| | - Yoshimi Shitakubo
- Division of Modern Medical Technology, Institute for Clinical Research, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima 737-0023, Japan
| | - Hiroyasu Yamashiro
- Department of Breast Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima 737-0023, Japan; Department of Breast Surgery, Tenri Hospital, Tenri, Nara 632-8552, Japan
| |
Collapse
|
27
|
Morgan RJ, Synold TW, Longmate JA, Quinn DI, Gandara D, Lenz HJ, Ruel C, Xi B, Lewis MD, Colevas AD, Doroshow J, Newman EM. Pharmacodynamics (PD) and pharmacokinetics (PK) of E7389 (eribulin, halichondrin B analog) during a phase I trial in patients with advanced solid tumors: a California Cancer Consortium trial. Cancer Chemother Pharmacol 2015; 76:897-907. [PMID: 26362045 DOI: 10.1007/s00280-015-2868-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 09/02/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND The California Cancer Consortium completed a phase I trial of E7389 (eribulin mesylate), an analog of the marine natural product halichondrin B. This trial was to determine the pharmacodynamics, pharmacokinetics, and MTD of E7389 administered by bolus injection weekly for 3 weeks out of four. METHODS This trial included a rapid titration design. Real-time pharmacokinetics were utilized to guide dose escalation. Initially, single-patient cohorts were enrolled with intra- and inter-patient dose doubling. The second phase was a standard 3 + 3 dose escalation schedule. At the MTD, a cohort of patients was enrolled for target validation studies (separate manuscript). The starting dose was 0.125 mg/m(2), and doses were doubled within and between patients in the first phase. Blood and urine sampling for E7389 pharmacokinetics was performed on doses 1 and 3 of cycle 1. Levels were determined using a LC/MS/MS assay. RESULTS Forty patients were entered. Thirty-eight were evaluable for toxicity and 35 for response. The rapid escalation ended with a grade 3 elevation of alkaline phosphatase at 0.5 mg/m(2)/week. The second phase ended at 2.0 mg/m(2)/week with dose-limiting toxicities of grades 3 and 4 febrile neutropenia. Other toxicities included hypoglycemia, hypophosphatemia, and fatigue. The MTD was 1.4 mg/m(2)/week. Responses included four partial responses (lung cancer [2], urothelial [1], and melanoma [1]). CONCLUSIONS E7389 was well tolerated in this trial with the major toxicity being myelosuppression. PD shows that E7389 induces significant morphologic changes (bundle formation) in the microtubules of peripheral blood mononuclear cells and tumor cells in vivo. The data suggest that lower intra-tumoral levels of β-tubulin III or higher intra-tumoral levels of MAP4 may correlate with response to E7389, while lower intra-tumoral levels of stathmin may be associated with progression. PK data reveal that E7389 exhibits a tri-exponential elimination from the plasma of patients receiving a rapid i.v. infusion. At sub-toxic doses, plasma concentrations of E7389 are maintained well above the levels required for activity in vitro for >72 h.
Collapse
Affiliation(s)
- Robert J Morgan
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd., Duarte, CA, 91010, USA.
| | - Timothy W Synold
- Department of Cancer Biology, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Jeffrey A Longmate
- Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - David I Quinn
- Division of Medical Oncology, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, 90033, USA
| | - David Gandara
- Division of Medical Oncology, University of California, Davis Cancer Center, Sacramento, CA, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, 90033, USA
| | - Christopher Ruel
- Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Bixin Xi
- Department of Cancer Biology, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Michael D Lewis
- Eisai Research Institute, Andover, MA, 01810, USA.,Edward P. Evans Foundation, Casanova, VA, 20139, USA
| | - A Dimitrios Colevas
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, 20892, USA.,Stanford Cancer Center, Stanford, CA, 94305, USA
| | - James Doroshow
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd., Duarte, CA, 91010, USA.,Division of Cancer Treatment and Diagnosis6, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Edward M Newman
- Department of Cancer Biology, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| |
Collapse
|
28
|
Peters C, Brown S. Antibody-drug conjugates as novel anti-cancer chemotherapeutics. Biosci Rep 2015; 35:e00225. [PMID: 26182432 PMCID: PMC4613712 DOI: 10.1042/bsr20150089] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/18/2015] [Accepted: 05/29/2015] [Indexed: 12/19/2022] Open
Abstract
Over the past couple of decades, antibody-drug conjugates (ADCs) have revolutionized the field of cancer chemotherapy. Unlike conventional treatments that damage healthy tissues upon dose escalation, ADCs utilize monoclonal antibodies (mAbs) to specifically bind tumour-associated target antigens and deliver a highly potent cytotoxic agent. The synergistic combination of mAbs conjugated to small-molecule chemotherapeutics, via a stable linker, has given rise to an extremely efficacious class of anti-cancer drugs with an already large and rapidly growing clinical pipeline. The primary objective of this paper is to review current knowledge and latest developments in the field of ADCs. Upon intravenous administration, ADCs bind to their target antigens and are internalized through receptor-mediated endocytosis. This facilitates the subsequent release of the cytotoxin, which eventually leads to apoptotic cell death of the cancer cell. The three components of ADCs (mAb, linker and cytotoxin) affect the efficacy and toxicity of the conjugate. Optimizing each one, while enhancing the functionality of the ADC as a whole, has been one of the major considerations of ADC design and development. In addition to these, the choice of clinically relevant targets and the position and number of linkages have also been the key determinants of ADC efficacy. The only marketed ADCs, brentuximab vedotin and trastuzumab emtansine (T-DM1), have demonstrated their use against both haematological and solid malignancies respectively. The success of future ADCs relies on improving target selection, increasing cytotoxin potency, developing innovative linkers and overcoming drug resistance. As more research is conducted to tackle these issues, ADCs are likely to become part of the future of targeted cancer therapeutics.
Collapse
Affiliation(s)
- Christina Peters
- School of Life Sciences, Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, U.K
| | - Stuart Brown
- School of Life Sciences, Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, U.K.
| |
Collapse
|
29
|
Kessler L, Falato C, Margolin S, Bergh J, Foukakis T. A retrospective safety and efficacy analysis of the first patients treated with eribulin for metastatic breast cancer in Stockholm, Sweden. Acta Oncol 2015; 54:522-9. [PMID: 25383448 DOI: 10.3109/0284186x.2014.973063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUNDS Eribulin is a non-taxane, microtubule dynamics inhibitor approved for the treatment of patients with metastatic breast cancer (MBC) in Europe in March 2011. MATERIAL AND METHODS For the purpose of an internal quality control, all patients with MBC treated with eribulin at Karolinska University Hospital were registered in a database. Clinical data were collected retrospectively for patients that were registered by August 2012 and safety and efficacy of eribulin were evaluated. Treatment toxicity including fatigue, neurotoxicity and infection was graded according to CTCAE v4.0. Objective response to treatment was investigated using routinely performed radiological assessments. When only clinical assessments were made, the evaluation of the treating physician was used. Furthermore, the efficacy of eribulin was investigated in different tumor subtypes. RESULTS Forty-eight patients who received at least one cycle of eribulin were identified. Most patients were heavily pretreated with a median of 3 (range 1-7) previous chemotherapy lines prior to eribulin. Median patient age was 56 years (range 35-74). At the end of the analysis, 23 patients were alive and two were still treated with eribulin. No hypersensitivity reactions and no toxic deaths were seen. Fatigue grade 3-4 was observed in three patients (6.3%). One patient experienced grade 4 neurotoxicity. Grade 3-4 neutropenia was documented in 18.8%, and three patients were treated for a grade 3 infection. Interestingly, three individuals developed Herpes zoster reactivation. One patient responded to treatment with complete remission, while 33.3% had a partial response. 48% of all patients had a clinical benefit (objective response or stable disease for more than six months). CONCLUSIONS Eribulin administered outside of a clinical trial in patients with advanced breast cancer was safe and well tolerated. A clinical benefit was seen in half of the cases. No statistically significant differences in objective response or survival were observed between histopathological subgroups.
Collapse
Affiliation(s)
- Luisa Kessler
- Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital , Stockholm , Sweden
| | | | | | | | | |
Collapse
|
30
|
Zhang X, Raghavan S, Ihnat M, Hamel E, Zammiello C, Bastian A, Mooberry SL, Gangjee A. The design, synthesis and biological evaluation of conformationally restricted 4-substituted-2,6-dimethylfuro[2,3-d]pyrimidines as multi-targeted receptor tyrosine kinase and microtubule inhibitors as potential antitumor agents. Bioorg Med Chem 2015; 23:2408-23. [PMID: 25882519 DOI: 10.1016/j.bmc.2015.03.061] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/13/2015] [Accepted: 03/20/2015] [Indexed: 01/19/2023]
Abstract
A series of eleven conformationally restricted, 4-substituted 2,6-dimethylfuro[2,3-d]pyrimidines was designed to explore the bioactive conformation required for dual inhibition of microtubule assembly and receptor tyrosine kinases (RTKs), and their biological activities are reported. All three rotatable single bonds in the lead compound 1 were sequentially restricted to address the role of each in SAR for microtubule and RTK inhibitory effects. Compounds 2, 3, 7 and 10 showed microtubule depolymerizing activity comparable to or better than the lead 1, some with nanomolar EC50 values. While compound 8 had no effect on microtubules, 8 and 10 both showed potent RTK inhibition with nanomolar IC50s. These compounds confirm that the bioactive conformation for RTK inhibition is different from that for tubulin inhibition. The tetrahydroquinoline analog 10 showed the most potent dual tubulin and RTK inhibitory activities (low nanomolar inhibition of EGFR, VEGFR2 and PDGFR-β). Compound 10 has highly potent activity against many NCI cancer cell lines, including several chemo-resistant cell lines, and could serve as a lead for further preclinical studies.
Collapse
Affiliation(s)
- Xin Zhang
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States
| | - Sudhir Raghavan
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States
| | - Michael Ihnat
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Cynthia Zammiello
- Department of Pharmacology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Anja Bastian
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Susan L Mooberry
- Department of Pharmacology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States.
| | - Aleem Gangjee
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States.
| |
Collapse
|
31
|
Jimmidi R, Guduru SKR, Arya P. Practical stereoselective synthesis of eribulin fragment toward building a hybrid macrocyclic toolbox. Org Lett 2015; 17:468-71. [PMID: 25583003 DOI: 10.1021/ol503464s] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A practical stereoselective synthesis to obtain the substituted furan ring as the substructure of eribulin is developed. An asymmetric syn-aldol and intramolecular oxy-Michael were two key steps in our approach. The functionalized furan derivatives were then utilized further to build the 14- and 12-membered macrocyclic diversity as trans- and cis-fused (C-29 and C-30) compounds. This is the first report of building a chemical toolbox with macrocyclic small molecules having trans- or cis-fused 14- or 12-membered rings containing the substructure of eribulin and its diastereomer.
Collapse
Affiliation(s)
- Ravikumar Jimmidi
- Dr. Reddy's Institute of Life Sciences (DRILS) , University of Hyderabad Campus, Hyderabad 500046, India
| | | | | |
Collapse
|
32
|
Negi AS, Gautam Y, Alam S, Chanda D, Luqman S, Sarkar J, Khan F, Konwar R. Natural antitubulin agents: importance of 3,4,5-trimethoxyphenyl fragment. Bioorg Med Chem 2014; 23:373-89. [PMID: 25564377 DOI: 10.1016/j.bmc.2014.12.027] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 12/11/2014] [Accepted: 12/12/2014] [Indexed: 01/29/2023]
Abstract
Microtubules are polar cytoskeletal filaments assembled from head-to-tail and comprised of lateral associations of α/β-tubulin heterodimers that play key role in various cellular processes. Because of their vital role in mitosis and various other cellular processes, microtubules have been attractive targets for several disease conditions and especially for cancer. Antitubulin is the most successful class of antimitotic agents in cancer chemotherapeutics. The target recognition of antimitotic agents as a ligand is not much explored so far. However, 3,4,5-trimethoxyphenyl fragment has been much highlighted and discussed in such type of interactions. In this review, some of the most important naturally occurring antimitotic agents and their interactions with microtubules are discussed with a special emphasis on the role of 3,4,5-trimethoxyphenyl unit. At last, some emerging naturally occurring antimitotic agents have also been tabulated.
Collapse
Affiliation(s)
- Arvind S Negi
- CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP), Kukrail Picnic Spot Road, PO CIMAP, Lucknow 226015, India.
| | - Yashveer Gautam
- CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP), Kukrail Picnic Spot Road, PO CIMAP, Lucknow 226015, India
| | - Sarfaraz Alam
- CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP), Kukrail Picnic Spot Road, PO CIMAP, Lucknow 226015, India
| | - Debabrata Chanda
- CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP), Kukrail Picnic Spot Road, PO CIMAP, Lucknow 226015, India
| | - Suaib Luqman
- CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP), Kukrail Picnic Spot Road, PO CIMAP, Lucknow 226015, India
| | - Jayanta Sarkar
- CSIR-Central Drug Research Institute (CSIR-CDRI), B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Feroz Khan
- CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP), Kukrail Picnic Spot Road, PO CIMAP, Lucknow 226015, India
| | - Rituraj Konwar
- CSIR-Central Drug Research Institute (CSIR-CDRI), B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| |
Collapse
|
33
|
A new tubulin-binding site and pharmacophore for microtubule-destabilizing anticancer drugs. Proc Natl Acad Sci U S A 2014; 111:13817-21. [PMID: 25114240 DOI: 10.1073/pnas.1408124111] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The recent success of antibody-drug conjugates (ADCs) in the treatment of cancer has led to a revived interest in microtubule-destabilizing agents. Here, we determined the high-resolution crystal structure of the complex between tubulin and maytansine, which is part of an ADC that is approved by the US Food and Drug Administration (FDA) for the treatment of advanced breast cancer. We found that the drug binds to a site on β-tubulin that is distinct from the vinca domain and that blocks the formation of longitudinal tubulin interactions in microtubules. We also solved crystal structures of tubulin in complex with both a variant of rhizoxin and the phase 1 drug PM060184. Consistent with biochemical and mutagenesis data, we found that the two compounds bound to the same site as maytansine and that the structures revealed a common pharmacophore for the three ligands. Our results delineate a distinct molecular mechanism of action for the inhibition of microtubule assembly by clinically relevant agents. They further provide a structural basis for the rational design of potent microtubule-destabilizing agents, thus opening opportunities for the development of next-generation ADCs for the treatment of cancer.
Collapse
|
34
|
Aeluri M, Chamakuri S, Dasari B, Guduru SKR, Jimmidi R, Jogula S, Arya P. Small Molecule Modulators of Protein–Protein Interactions: Selected Case Studies. Chem Rev 2014; 114:4640-94. [DOI: 10.1021/cr4004049] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Madhu Aeluri
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Srinivas Chamakuri
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Bhanudas Dasari
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Shiva Krishna Reddy Guduru
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Ravikumar Jimmidi
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Srinivas Jogula
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Prabhat Arya
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| |
Collapse
|
35
|
Harmrolfs K, Mancuso L, Drung B, Sasse F, Kirschning A. Preparation of new alkyne-modified ansamitocins by mutasynthesis. Beilstein J Org Chem 2014; 10:535-43. [PMID: 24605171 PMCID: PMC3943755 DOI: 10.3762/bjoc.10.49] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 01/29/2014] [Indexed: 11/23/2022] Open
Abstract
The preparation of alkyne-modified ansamitocins by mutasynthetic supplementation of Actinosynnema pretiosum mutants with alkyne-substituted aminobenzoic acids is described. This modification paved the way to introduce a thiol linker by Huisgen-type cycloaddition which can principally be utilized to create tumor targeting conjugates. In bioactivity tests, only those new ansamitocin derivatives showed strong antiproliferative activity that bear an ester side chain at C-3.
Collapse
Affiliation(s)
- Kirsten Harmrolfs
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ), Leibniz University Hannover, Schneiderberg 1b, 30167 Hannover, Germany
| | - Lena Mancuso
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ), Leibniz University Hannover, Schneiderberg 1b, 30167 Hannover, Germany
| | - Binia Drung
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ), Leibniz University Hannover, Schneiderberg 1b, 30167 Hannover, Germany
| | - Florenz Sasse
- Department of Chemical Biology, Helmholtz Center for Infectious Research (HZI), Inhoffenstraße 7, D-38124 Braunschweig, Germany
| | - Andreas Kirschning
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ), Leibniz University Hannover, Schneiderberg 1b, 30167 Hannover, Germany
| |
Collapse
|
36
|
Sapra P, Betts A, Boni J. Preclinical and clinical pharmacokinetic/pharmacodynamic considerations for antibody–drug conjugates. Expert Rev Clin Pharmacol 2014; 6:541-55. [DOI: 10.1586/17512433.2013.827405] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
37
|
Venghateri JB, Gupta TK, Verma PJ, Kunwar A, Panda D. Ansamitocin P3 depolymerizes microtubules and induces apoptosis by binding to tubulin at the vinblastine site. PLoS One 2013; 8:e75182. [PMID: 24124473 PMCID: PMC3790769 DOI: 10.1371/journal.pone.0075182] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 08/11/2013] [Indexed: 11/18/2022] Open
Abstract
Maytansinoid conjugates are currently under different phases of clinical trials and have been showing promising activity for various types of cancers. In this study, we have elucidated the mechanism of action of ansamitocin P3, a structural analogue of maytansine for its anticancer activity. Ansamitocin P3 potently inhibited the proliferation of MCF-7, HeLa, EMT-6/AR1 and MDA-MB-231 cells in culture with a half-maximal inhibitory concentration of 20±3, 50±0.5, 140±17, and 150±1.1 pM, respectively. Ansamitocin P3 strongly depolymerized both interphase and mitotic microtubules and perturbed chromosome segregation at its proliferation inhibitory concentration range. Treatment of ansamitocin P3 activated spindle checkpoint surveillance proteins, Mad2 and BubR1 and blocked the cells in mitotic phase of the cell cycle. Subsequently, cells underwent apoptosis via p53 mediated apoptotic pathway. Further, ansamitocin P3 was found to bind to purified tubulin in vitro with a dissociation constant (Kd) of 1.3±0.7 µM. The binding of ansamitocin P3 induced conformational changes in tubulin. A docking analysis suggested that ansamitocin P3 may bind partially to vinblastine binding site on tubulin in two different positions. The analysis indicated that the binding of ansamitocin P3 to tubulin is stabilized by hydrogen bonds. In addition, weak interactions such as halogen-oxygen interactions may also contribute to the binding of ansamitocin P3 to tubulin. The study provided a significant insight in understanding the antiproliferative mechanism of action of ansamitocin P3.
Collapse
Affiliation(s)
- Jubina B. Venghateri
- IITB-Monash Research Academy, Indian Institute of Technology Bombay, Mumbai, India
| | - Tilak Kumar Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Paul J. Verma
- Centre for Reproduction and Development, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Ambarish Kunwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
- * E-mail:
| |
Collapse
|
38
|
Yu MJ, Zheng W, Seletsky BM. From micrograms to grams: scale-up synthesis of eribulin mesylate. Nat Prod Rep 2013; 30:1158-64. [PMID: 23896896 DOI: 10.1039/c3np70051h] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The synthesis of eribulin mesylate from microgram to multi-gram scale is described in this Highlight. Key coupling reactions include formation of the C30a to C1 carbon-carbon bond and macrocyclic ring closure through an intramolecular Nozaki-Hiyama-Kishi reaction.
Collapse
|
39
|
Shablak A. Eribulin for advanced breast cancer: a drug evaluation. J Breast Cancer 2013; 16:12-5. [PMID: 23593076 PMCID: PMC3625759 DOI: 10.4048/jbc.2013.16.1.12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 03/07/2013] [Indexed: 11/30/2022] Open
Abstract
Eribulin is a synthetic microtubule dynamics inhibitor that was developed from a marine natural product halichondrin B. It exhibited in vitro and in vivo activities against a wide number of malignancies. A number of advanced phase trials showed improved survival following eribulin treatment in pretreated advanced breast cancer patients. This review provides an overview of the background to the therapeutic use of eribulin in oncology, including its pharmacology, pharmacokinetics, clinical efficacy, safety, and potential economic factors.
Collapse
Affiliation(s)
- Alaaeldin Shablak
- Department of Medical Oncology, Bradford Royal Infirmary, Bradford, UK
| |
Collapse
|
40
|
Noro JC, Kalaitzis JA, Neilan BA. Bioactive natural products from Papua New Guinea marine sponges. Chem Biodivers 2013; 9:2077-95. [PMID: 23081914 DOI: 10.1002/cbdv.201100292] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The discovery of novel natural products for drug development relies heavily upon a rich biodiversity, of which the marine environment is an obvious example. Marine natural product research has spawned several drugs and many other candidates, some of which are the focus of current clinical trials. The sponge megadiversity of Papua New Guinea is a rich but underexplored source of bioactive natural products. Here, we review some of the many natural products derived from PNG sponges with an emphasis on those with interesting biological activity and, therefore, drug potential. Many bioactive natural products discussed here appear to be derived from non-ribosomal peptide and polyketide biosynthesis pathways, strongly suggesting a microbial origin of these compounds. With this in mind, we also explore the notion of sponge-symbiont biosynthesis of these bioactive compounds and present examples to support the working hypothesis.
Collapse
Affiliation(s)
- Jeffery C Noro
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | | | | |
Collapse
|
41
|
Sapra P, Shor B. Monoclonal antibody-based therapies in cancer: advances and challenges. Pharmacol Ther 2013; 138:452-69. [PMID: 23507041 DOI: 10.1016/j.pharmthera.2013.03.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 02/19/2013] [Indexed: 12/25/2022]
Abstract
Conventional anticancer therapeutics often suffer from lack of specificity, resulting in toxicities to normal healthy tissues and poor therapeutic index. Antibody-mediated delivery of anticancer drugs or toxins to tumor cells through tumor selective or overexpressed antigens is progressively being recognized as an effective strategy for increasing the therapeutic index of anticancer drugs. In this review we focus on three therapeutic modalities in the field of antibody-mediated targeting, including antibody-drug conjugates (ADCs), immunotoxins (ITs) and immunoliposomes (ILs). Design considerations for development of each of the above therapeutic modalities are discussed. Furthermore, an overview of ADCs, ITs or ILs approved for use in clinical oncology and those currently in clinical development is provided. Challenges encountered by the field of antibody-based targeting are discussed and concepts around development of the next generation of antibody therapeutics are presented.
Collapse
Affiliation(s)
- Puja Sapra
- Bioconjugates Discovery and Development, Oncology Research Unit, Pfizer Worldwide Research and Development, 401 North Middletown Road, Pearl River, NY, 10965, USA.
| | | |
Collapse
|
42
|
|
43
|
Synthesis and evaluation of novel anti-proliferative pyrroloazepinone and indoloazepinone oximes derived from the marine natural product hymenialdisine. Eur J Med Chem 2012; 56:246-53. [PMID: 22995819 DOI: 10.1016/j.ejmech.2012.08.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 08/03/2012] [Accepted: 08/16/2012] [Indexed: 11/21/2022]
Abstract
The tetrahydroazepinone pharmacophore is a component of many interesting compounds, including several marine natural products, with anti-cancer properties. The synthesis and biological evaluation of a novel series of pyrroloazepinone and indoloazepinone oximes is reported. These compounds showed promising growth inhibition activity against four human cancer cell lines but did not significantly inhibit the cell cycle regulator cyclin dependent kinase 2. The most active compounds in this series displayed improved anti-proliferative activity over the related synthetic indoloazepine kenpaullone. The structure activity relationships exhibited by the azepinone pharmacophore suggests several novel lead compounds for anti-cancer drug discovery.
Collapse
|
44
|
Ranaivoson FM, Gigant B, Berritt S, Joullié M, Knossow M. Structural plasticity of tubulin assembly probed by vinca-domain ligands. ACTA CRYSTALLOGRAPHICA SECTION D: BIOLOGICAL CRYSTALLOGRAPHY 2012; 68:927-34. [DOI: 10.1107/s0907444912017143] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 04/18/2012] [Indexed: 11/10/2022]
|
45
|
Knobloch T, Dräger G, Collisi W, Sasse F, Kirschning A. Unprecedented deoxygenation at C-7 of the ansamitocin core during mutasynthetic biotransformations. Beilstein J Org Chem 2012; 8:861-9. [PMID: 23015834 PMCID: PMC3388874 DOI: 10.3762/bjoc.8.96] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 05/16/2012] [Indexed: 11/23/2022] Open
Abstract
We describe the unprecedented formation of six ansamitocin derivatives that are deoxygenated at C-7 of the ansamitocin core, obtained during fermentation experiments by employing a variety of Actinosynnema pretiosum mutants and mutasynthetic approaches. We suggest that the formation of these derivatives is based on elimination at C-7/C-8 followed by reduction(s) of the intermediate enone. In bioactivity tests, only ansamitocin derivatives bearing an ester side chain at C-3 showed strong antiproliferative activity.
Collapse
Affiliation(s)
- Tobias Knobloch
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ), Leibniz University Hannover, Schneiderberg 1b, 30167 Hannover, Germany
| | - Gerald Dräger
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ), Leibniz University Hannover, Schneiderberg 1b, 30167 Hannover, Germany
| | - Wera Collisi
- Department of Chemical Biology, Helmholtz Center for Infectious Research (HZI), Inhoffenstraße 7, D-38124 Braunschweig, Germany
| | - Florenz Sasse
- Department of Chemical Biology, Helmholtz Center for Infectious Research (HZI), Inhoffenstraße 7, D-38124 Braunschweig, Germany
| | - Andreas Kirschning
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ), Leibniz University Hannover, Schneiderberg 1b, 30167 Hannover, Germany
| |
Collapse
|
46
|
|
47
|
Scherlach K, Brendel N, Ishida K, Dahse HM, Hertweck C. Photochemical oxazole-nitrile conversion downstream of rhizoxin biosynthesis and its impact on antimitotic activity. Org Biomol Chem 2012; 10:5756-9. [PMID: 22453231 DOI: 10.1039/c2ob25250c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Through metabolic profiling of mutants and wild type of the endofungal bacterium Burkholderia rhizoxinica two novel rhizoxin derivatives with unusual nitrile substitutions were discovered. The nitrile groups result from a photochemical oxidative cleavage of the oxazolyl moiety. In vitro studies revealed that the photooxidation by singlet oxygen also takes place in the absence of a photosensitizer, and that also a thiazolyl-substituted rhizoxin analogue undergoes the same transformation. The resulting nitriles have antimitotic properties but are significantly less active than the parent compounds. These results highlight the impact of photoreactions onto the antiproliferative agent and encourage the introduction of bioisosteric groups that render the compound less susceptible towards photooxidation.
Collapse
Affiliation(s)
- Kirstin Scherlach
- Leibniz Institute for Natural Product Research and Infection Biology, HKI, Beutenbergstr. 11a, D-07745 Jena, Germany
| | | | | | | | | |
Collapse
|
48
|
Ortega V, Cortés J. Potential clinical applications of halichondrins in breast cancer and other neoplasms. BREAST CANCER (DOVE MEDICAL PRESS) 2012; 4:9-19. [PMID: 24367189 PMCID: PMC3846814 DOI: 10.2147/bctt.s12423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Halichondrin B is a large polyether macrolide found in a rare Japanese sponge, Halichondria okadai and has been shown to have anticancer activity. Eribulin mesylate is a completely synthetic analog of halichondrin B with a unique mechanism of action relative to other antimicrotubule agents. This new agent has demonstrated activity in preclinical studies, and it is being developed for the treatment of different tumor types. Eribulin has been approved by the United States Food and Drug Administration and the European Medicines Agency as late-line therapy for metastatic breast cancer patients previously treated with an anthracycline and a taxane. It has demonstrated superiority over other treatments in overall survival (OS) (hazard ratio: 0.81, P = 0.041), leading to its regulatory approbation for clinical practice use. Median OS for the eribulin-treated group was 13.1 months versus 10.6 months in the physician's treatment-of-choice group. Eribulin demonstrated a manageable toxicity profile. Most common adverse events associated with treatment were mild neutropenia and fatigue, mainly of grade 1 or 2. In contrast to other antimicrotubule agents, eribulin has a relatively low incidence of peripheral neuropathy and alopecia. Eribulin has been extensively studied in breast cancer and is currently being developed for treatment of other cancer types. Eribulin has demonstrated activity in Phase II trials in non-small cell lung cancer, pancreatic cancer, urothelial tract cancer, and sarcomas. Further studies in these cancers are ongoing. This article reviews pharmacology, mechanism of action, pharmacokinetics and efficacy of eribulin in breast cancer and other neoplasms.
Collapse
Affiliation(s)
- Vanesa Ortega
- Department of Oncology, Vall d’Hebrón University Hospital, Barcelona, Spain
| | - Javier Cortés
- Department of Oncology, Vall d’Hebrón University Hospital, Barcelona, Spain
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
49
|
Swami U, Chaudhary I, Ghalib MH, Goel S. Eribulin -- a review of preclinical and clinical studies. Crit Rev Oncol Hematol 2012; 81:163-84. [PMID: 21493087 PMCID: PMC3954568 DOI: 10.1016/j.critrevonc.2011.03.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 02/25/2011] [Accepted: 03/04/2011] [Indexed: 11/22/2022] Open
Abstract
Eribulin mesylate is a non-taxane, structurally simplified, completely synthetic, halichondrin B derivative with an end poisoning, microtubule inhibitory action. Preclinical studies have demonstrated activity in various cancer cell lines and synergistic action with gemcitabine, epirubicin, trastuzumab, cisplatin, docetaxel and vinorelbine. Eribulin has recently been approved by United States Food and Drug Administration as a third line therapy for metastatic breast cancer patients, who have previously been treated with an anthracycline and a taxane. It has also advanced to phase II trials in non-small cell lung cancer, pancreatic, prostate, bladder, head and neck cancers, sarcomas and ovarian and other gynecological tumors. Combination trials with carboplatin, gemcitabine, pemetrexed, cisplatin, and erlotinib are currently ongoing. Eribulin potentially has a low incidence of peripheral neuropathy. The predominant side effects are neutropenia and fatigue, which are manageable. This article reviews the available information on eribulin with respect to its clinical pharmacology, mechanism of action, pharmacokinetics, pharmacodynamics, metabolism, preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Umang Swami
- Albert Einstein College of Medicine, Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, Bronx, NY 10461, United States
| | - Imran Chaudhary
- Albert Einstein College of Medicine, Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, Bronx, NY 10461, United States
| | - Mohammad H. Ghalib
- Albert Einstein College of Medicine, Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, Bronx, NY 10461, United States
| | - Sanjay Goel
- Albert Einstein College of Medicine, Department of Medical Oncology, Montefiore Medical Center, 1825 Eastchester Road, Bronx, NY 10461, United States
| |
Collapse
|
50
|
Muñoz-Couselo E, Pérez-García J, Cortés J. Eribulin mesylate as a microtubule inhibitor for treatment of patients with metastatic breast cancer. Onco Targets Ther 2011; 4:185-92. [PMID: 22162924 PMCID: PMC3233277 DOI: 10.2147/ott.s16392] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Metastatic breast cancer (MBC) remains an incurable disease, with the goals of care aimed at maximizing the patient's duration and quality of life. Treatment options for MBC have become more efficacious and numerous. In addition to endocrine and chemotherapy agents, a number of targeted agents, including trastuzumab and bevacizumab, have further enhanced the landscape of therapeutic options. Eribulin mesylate (E7389) is a nontaxane microtubule dynamics inhibitor, and a structurally simplified synthetic analog of the natural marine product, halichondrin B, with a novel mechanism of action that has shown antitumor activity in pretreated MBC. Eribulin has shown a manageable tolerability profile in Phase I-II clinical trials and an improvement in overall survival compared with treatment of physician's choice, without relevant toxicities in a recently published Phase III trial. This review will focus on eribulin as a new active agent for MBC and its role in the management of breast disease.
Collapse
Affiliation(s)
- Eva Muñoz-Couselo
- Breast Cancer Unit, Vall d’Hebron Institute of Oncology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José Pérez-García
- Breast Cancer Unit, Vall d’Hebron Institute of Oncology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Javier Cortés
- Breast Cancer Unit, Vall d’Hebron Institute of Oncology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|