1
|
Nardin S, Sacco G, Lagodin D'Amato A, Barcellini L, Rovere M, Santamaria S, Marconi S, Coco S, Genova C. Updates in pharmacotherapy for non-small cell lung cancer: a focus on emerging tubulin inhibitors. Expert Opin Pharmacother 2024; 25:1051-1069. [PMID: 38935538 DOI: 10.1080/14656566.2024.2369196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION The treatment landscape of non-small cell lung cancer (NSCLC) has seen significant advancements in recent years, marked by a shift toward target agents and immune checkpoint inhibitors (ICIs). However, chemotherapy remains a cornerstone of treatment, alone or in combination. Microtubule-targeting agents, such as taxanes and vinca alkaloids, play a crucial role in clinical practice in both early and advanced settings in NSCLC. AREA COVERED This review outlines the mechanisms of action, present significance, and prospective advancements of microtubule-targeting agents (MTAs), with a special highlight on new combinations in phase 3 trials. The online databases PubMed, Web of Science, Cochrane Library, and ClinicalTrials.gov were searched using the terms 'Microtubule-targeting agents' and 'non-small cell lung cancer' or synonyms, with a special focus over the last 5 years of publications. EXPERT OPINION Despite the emergence of immunotherapy, MTA remains crucial, often used alongside or after immunotherapy, especially in squamous cell lung cancer. Next-generation sequencing expands treatment options, but reliable biomarkers for immunotherapy are lacking. While antibody-drug conjugates (ADCs) show promise, managing toxicities remain vital. In the early stages, MTAs, possibly with ICIs, are standard, while ADCs may replace traditional chemotherapy in the advanced stages. Nevertheless, MTAs remain essential in subsequent lines or for patients with contraindications.
Collapse
Affiliation(s)
- Simone Nardin
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, Genoa, Italy
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Gianluca Sacco
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, Genoa, Italy
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Agostina Lagodin D'Amato
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, Genoa, Italy
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lucrezia Barcellini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, Genoa, Italy
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Matteo Rovere
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sara Santamaria
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Marconi
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Carlo Genova
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, Genoa, Italy
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
2
|
Joussaume A, Kanthou C, Pardo OE, Karayan-Tapon L, Benzakour O, Dkhissi F. The Vitamin K-Dependent Anticoagulant Factor, Protein S, Regulates Vascular Permeability. Curr Issues Mol Biol 2024; 46:3278-3293. [PMID: 38666935 PMCID: PMC11048934 DOI: 10.3390/cimb46040205] [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: 03/01/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Protein S (PROS1) is a vitamin K-dependent anticoagulant factor, which also acts as an agonist for the TYRO3, AXL, and MERTK (TAM) tyrosine kinase receptors. PROS1 is produced by the endothelium which also expresses TAM receptors, but little is known about its effects on vascular function and permeability. Transwell permeability assays as well as Western blotting and immunostaining analysis were used to monitor the possible effects of PROS1 on both endothelial cell permeability and on the phosphorylation state of specific signaling proteins. We show that human PROS1, at its circulating concentrations, substantially increases both the basal and VEGFA-induced permeability of endothelial cell (EC) monolayers. PROS1 induces p38 MAPK (Mitogen Activated Protein Kinase), Rho/ROCK (Rho-associated protein kinase) pathway activation, and actin filament remodeling, as well as substantial changes in Vascular Endothelial Cadherin (VEC) distribution and its phosphorylation on Ser665 and Tyr685. It also mediates c-Src and PAK-1 (p21-activated kinase 1) phosphorylation on Tyr416 and Ser144, respectively. Exposure of EC to human PROS1 induces VEC internalization as well as its cleavage into a released fragment of 100 kDa and an intracellular fragment of 35 kDa. Using anti-TAM neutralizing antibodies, we demonstrate that PROS1-induced VEC and c-Src phosphorylation are mediated by both the MERTK and TYRO3 receptors but do not involve the AXL receptor. MERTK and TYRO3 receptors are also responsible for mediating PROS1-induced MLC (Myosin Light Chain) phosphorylation on a site targeted by the Rho/ROCK pathway. Our report provides evidence for the activation of the c-Src/VEC and Rho/ROCK/MLC pathways by PROS1 for the first time and points to a new role for PROS1 as an endogenous vascular permeabilizing factor.
Collapse
Affiliation(s)
- Aurélie Joussaume
- Université de Poitiers, CHU de Poitiers, ProDiCeT, UR 24144 Poitiers, France; (A.J.); (O.B.)
| | - Chryso Kanthou
- Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield S10 2RX, UK;
| | - Olivier E. Pardo
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK;
| | - Lucie Karayan-Tapon
- Université de Poitiers, CHU de Poitiers, ProDiCeT, Laboratoire de Cancérologie Biologique, UR 24144 Poitiers, France;
| | - Omar Benzakour
- Université de Poitiers, CHU de Poitiers, ProDiCeT, UR 24144 Poitiers, France; (A.J.); (O.B.)
| | - Fatima Dkhissi
- Université de Poitiers, CHU de Poitiers, ProDiCeT, UR 24144 Poitiers, France; (A.J.); (O.B.)
| |
Collapse
|
3
|
Ng WH, Soo KC, Huynh H. Vinorelbine Improves the Efficacy of Sorafenib against Hepatocellular Carcinoma: A Promising Therapeutic Approach. Int J Mol Sci 2024; 25:1563. [PMID: 38338842 PMCID: PMC10855313 DOI: 10.3390/ijms25031563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading global cause of cancer-related mortality. Despite the widespread adoption of sorafenib as the standard HCC treatment, its efficacy is constrained, frequently encountering resistance. To augment the effectiveness of sorafenib, this study investigated the synergy of sorafenib and vinorelbine using 22 HCC patient-derived xenograft (PDX) models. In this study, mice bearing HCC tumors were treated with the vehicle, sorafenib (15 mg/kg), vinorelbine (3 mg/kg), and sorafenib-vinorelbine combination (Sora/Vino). Rigorous monitoring of the tumor growth and side effects coupled with comprehensive histological and molecular analyses was conducted. The overall survival (OS) of mice bearing HCC orthotopic tumors was also assessed. Our data showed a notable 86.4% response rate to Sora/Vino, surpassing rates of 31.8% for sorafenib and 9.1% for vinorelbine monotherapies. Sora/Vino significantly inhibited tumor growth, prolonged OS of mice bearing HCC orthotopic tumors (p < 0.01), attenuated tumor cell proliferation and angiogenesis, and enhanced necrosis and apoptosis. The combination therapy effectively suppressed the focal adhesion kinase (FAK) pathway, which is a pivotal player in cell proliferation, tumor angiogenesis, survival, and metastasis. The noteworthy antitumor activity in 22 HCC PDX models positions Sora/Vino as a promising candidate for early-phase clinical trials, leveraging the established use of sorafenib and vinorelbine in HCC and other cancers.
Collapse
Affiliation(s)
- Wai Har Ng
- Laboratory of Molecular Endocrinology, National Cancer Centre Singapore, Singapore 168583, Singapore;
| | - Khee Chee Soo
- Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore 168583, Singapore
| | - Hung Huynh
- Laboratory of Molecular Endocrinology, National Cancer Centre Singapore, Singapore 168583, Singapore;
| |
Collapse
|
4
|
Tochinai R, Nagashima Y, Sekizawa SI, Kuwahara M. Anti-tumor and cardiotoxic effects of microtubule polymerization inhibitors: The mechanisms and management strategies. J Appl Toxicol 2024; 44:96-106. [PMID: 37496236 DOI: 10.1002/jat.4521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023]
Abstract
Microtubule polymerization inhibitors (MPIs) have long been used as anticancer agents because they inhibit mitosis. Microtubules are thought to play an important role in the migration of tumor cells and the formation of tumor blood vessels, and new MPIs are being developed. Many clinical trials of novel MPIs have been conducted in humans, while some clinical studies in dogs have also been reported. More attempts to apply MPIs not only in humans but also in the veterinary field are expected to be made in the future. Meanwhile, MPIs have a risk of cardiotoxicity. In this paper, we review findings on the pharmacological effects and cardiotoxicity of MPIs, as well as the mechanisms of their cardiotoxicity. Cardiotoxicity of MPIs involves not only the direct effects of MPIs on cardiomyocytes but also their effects on vascular function. For example, hypertension induced by impaired vascular function also contributes to the exacerbation of myocardial damage, and blood pressure control may be useful in reducing cardiotoxicity. By combined administration of MPIs and other anticancer agents, MPI efficacy may be enhanced, thereby potentially allowing to keep MPI dosage low. Measurement of myocardial injury markers in blood and echocardiography may be useful for monitoring cardiotoxicity. In particular, two-dimensional speckle tracking may have high sensitivity for the early detection of MPI-induced cardiac dysfunction. The exploration of the potential of new MPIs while understanding their toxicity and how to deal with them will lead to the further development of cancer chemotherapy.
Collapse
Affiliation(s)
- Ryota Tochinai
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshiyasu Nagashima
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichi Sekizawa
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Masayoshi Kuwahara
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Zholudeva AO, Potapov NS, Kozlova EA, Lomakina ME, Alexandrova AY. Impairment of Assembly of the Vimentin Intermediate Filaments Leads to Suppression of Formation and Maturation of Focal Contacts and Alteration of the Type of Cellular Protrusions. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:184-195. [PMID: 38467554 DOI: 10.1134/s0006297924010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/01/2023] [Accepted: 12/03/2023] [Indexed: 03/13/2024]
Abstract
Cell migration is largely determined by the type of protrusions formed by the cell. Mesenchymal migration is accomplished by formation of lamellipodia and/or filopodia, while amoeboid migration is based on bleb formation. Changing of migrational conditions can lead to alteration in the character of cell movement. For example, inhibition of the Arp2/3-dependent actin polymerization by the CK-666 inhibitor leads to transition from mesenchymal to amoeboid motility mode. Ability of the cells to switch from one type of motility to another is called migratory plasticity. Cellular mechanisms regulating migratory plasticity are poorly understood. One of the factors determining the possibility of migratory plasticity may be the presence and/or organization of vimentin intermediate filaments (VIFs). To investigate whether organization of the VIF network affects the ability of fibroblasts to form membrane blebs, we used rat embryo fibroblasts REF52 with normal VIF organization, fibroblasts with vimentin knockout (REF-/-), and fibroblasts with mutation inhibiting assembly of the full-length VIFs (REF117). Blebs formation was induced by treatment of cells with CK-666. Vimentin knockout did not lead to statistically significant increase in the number of cells with blebs. The fibroblasts with short fragments of vimentin demonstrate the significant increase in number of cells forming blebs both spontaneously and in the presence of CK-666. Disruption of the VIF organization did not lead to the significant changes in the microtubules network or the level of myosin light chain phosphorylation, but caused significant reduction in the focal contact system. The most pronounced and statistically significant decrease in both size and number of focal adhesions were observed in the REF117 cells. We believe that regulation of the membrane blebbing by VIFs is mediated by their effect on the focal adhesion system. Analysis of migration of fibroblasts with different organization of VIFs in a three-dimensional collagen gel showed that organization of VIFs determines the type of cell protrusions, which, in turn, determines the character of cell movement. A novel role of VIFs as a regulator of membrane blebbing, essential for manifestation of the migratory plasticity, is shown.
Collapse
Affiliation(s)
- Anna O Zholudeva
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | - Nikolay S Potapov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Ekaterina A Kozlova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Maria E Lomakina
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | - Antonina Y Alexandrova
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia.
| |
Collapse
|
6
|
Chen ZH, Xu RM, Zheng GH, Jin YZ, Li Y, Chen XY, Tian YS. Development of Combretastatin A-4 Analogues as Potential Anticancer Agents with Improved Aqueous Solubility. Molecules 2023; 28:molecules28041717. [PMID: 36838705 PMCID: PMC9963121 DOI: 10.3390/molecules28041717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
Combretastatin A-4 (CA-4) is a potent tubulin polymerisation inhibitor. However, the clinical application of CA-4 is limited owing to its low aqueous solubility and the easy conversion of the olefin double bond from the more active cis- to the less active trans-configuration. Several structural modifications were investigated to improve the solubility of CA-4 derivatives. Among the compounds we synthesized, the kinetic solubility assay revealed that the solubility of compounds containing a piperazine ring increased the most, and the solubility of compounds 12a1, 12a2, 15 and 18 was increased 230-2494 times compared with that of the control compound (Z)-3-(4-aminophenyl)-2-(3,4,5-trimethoxyphenyl)acrylonitrile (9a). In addition, these synthesised stilbene nitriles had high anticancer cell (AGS, BEL-7402, MCF-7, and HCT-116) selectivity over L-02 and MCF-10A normal cells while maintaining micromolar activity against cancer cells. The most cytotoxic compound is 9a, and the IC50 value is 20 nM against HCT-116 cancer cells. Preliminary studies indicated that compound 12a1 had excellent plasma stability and moderate binding to rat plasma proteins, suggesting it is a promising lead compound for the development of an anticancer agent.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yu-Shun Tian
- Correspondence: ; Tel.: +864332436028; Fax: +864332435026
| |
Collapse
|
7
|
Elhemely MA, Belgath AA, El-Sayed S, Burusco KK, Kadirvel M, Tirella A, Finegan K, Bryce RA, Stratford IJ, Freeman S. SAR of Novel 3-Arylisoquinolinones: meta-Substitution on the Aryl Ring Dramatically Enhances Antiproliferative Activity through Binding to Microtubules. J Med Chem 2022; 65:4783-4797. [PMID: 35290041 PMCID: PMC9098178 DOI: 10.1021/acs.jmedchem.1c01936] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A set of meta-substituted 3-arylisoquinolinones have been identified that show substantial cytotoxicity in breast, liver, lung and colon cancer cell lines; these are up to 700-fold more active than the corresponding para analogues. These compounds were initially proposed as inhibitors of N-ribosyl dihydronicotinamide (NRH): quinone oxidoreductase 2 (NQO2) but were found to be inactive against the enzyme. Instead, COMPARE analysis suggested that 6-fluoro-3-(meta-fluorophenyl)isoquinolin-1(2H)-one (4) could mimic colchicine and interact with microtubules, a recognized target for cancer therapy. Subsequent docking, molecular dynamics simulations, and free energy analysis further suggested that compound 4 bound well into the colchicine-binding pocket of tubulin. Indeed, 4 suppressed tubulin polymerization, caused G2/M cell cycle arrest, and induced apoptosis. Also, 4 inhibited the formation of endothelial cell capillary-like tubes and further disrupted the structure of preestablished tubes; the effects were not observed with para analogue 5. In accordance with this, the computed free energy of binding of 5 to tubulin was lower in magnitude than that for 4 and appeared to arise in part from the inability of the para substituent to occupy a tubulin subpocket, which is possible in the meta orientation. In conclusion, the antiproliferative potential of the novel 3-arylisoquinolinones is markedly influenced by a subtle change in the structure (meta versus para). The meta-substituted isoquinolinone 4 is a microtubule-destabilizing agent with potential tumor-selectivity and antiangiogenic and vascular disrupting features.
Collapse
Affiliation(s)
- Mai A Elhemely
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, U.K.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Asma A Belgath
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, U.K
| | - Sherihan El-Sayed
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, U.K.,Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Kepa K Burusco
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, U.K
| | - Manikandan Kadirvel
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, U.K
| | - Annalisa Tirella
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, U.K.,BIOtech Center for Biomedical Technologies, Department of Industrial Engineering, University of Trento, Via delle Regole 101, Trento 38123, Italy
| | - Katherine Finegan
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, U.K
| | - Richard A Bryce
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, U.K
| | - Ian J Stratford
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, U.K
| | - Sally Freeman
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
8
|
Taninaka A, Ugajin S, Kurokawa H, Nagoshi Y, Kamiyanagi M, Takeuchi O, Matsui H, Shigekawa H. Direct analysis of the actin-filament formation effect in photodynamic therapy. RSC Adv 2022; 12:5878-5889. [PMID: 35424553 PMCID: PMC8981521 DOI: 10.1039/d1ra09291j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/11/2022] [Indexed: 11/30/2022] Open
Abstract
Photodynamic therapy (PDT) is a method in which a photosensitizer is administered in vivo and irradiated with light to generate reactive oxygen species (ROS), thereby causing the selective death of cancer cells. Since PDT is a noninvasive cancer treatment method with few adverse effects, it has attracted considerable attention and is increasingly used. In PDT, there are two dominant processes based on the actin filament (A-filament) formation effect: the destruction of cells by necrosis and vascular shutdown. Despite the importance of its fine control, the mechanism of the reaction process from the generation of reactive oxygen by photoinduction inducing the formation of A-filament and its polymerization to form stress fibers (S-fibers) has not yet been clarified because, for example, it has been difficult to directly observe and quantify such processes in living cells by conventional methods. Here, we have combined atomic force microscopy (AFM) with other techniques to reveal the mechanism of the A-filament and S-fiber formation processes that underlie the cell death process due to PDT. First, it was confirmed that activation of the small G protein RhoA, which is a signal that induces an increase in A-filament production, begins immediately after PDT treatment. The production of A-filament did not increase with increasing light intensity when the amount of light was large. Namely, the activation of RhoA reached an equilibrium state in about 1 min: however, the production of A-filament and its polymerization continued. The observed process corresponds well with the change in the amount of phosphorylated myosin-light chains, which induce A-filament polymerization. The increase in the elastic modulus of cells following the formation of S-fiber was confirmed by AFM for the first time. The distribution of generated A-filament and S-fiber was consistent with the photosensitizer distribution. PDT increases A-filament production, and when the ROS concentration is high, blebbing occurs and cells die, but when it is low, cell death does not occur and S-fiber is formed. That is, it is expected that vascular shutdown can be controlled efficiently by adjusting the amount of photosensitizer and the light intensity. We have combined atomic force microscopy with other techniques to reveal the mechanism of the actin filament and stress fibers formation processes that underlies the cell death process due to photodynamic therapy.![]()
Collapse
Affiliation(s)
- Atsushi Taninaka
- Faculty of Pure and Applied Sciences, University of Tsukuba 305-8573 Ibaraki Japan .,TAKANO Co., LTD. Miyada-mura, Kamiina-gun Nagano 399-4301 Japan
| | - Shunta Ugajin
- Faculty of Pure and Applied Sciences, University of Tsukuba 305-8573 Ibaraki Japan
| | - Hiromi Kurokawa
- Faculty of Medicine, University of Tsukuba 305-8575 Ibaraki Japan
| | - Yu Nagoshi
- Faculty of Pure and Applied Sciences, University of Tsukuba 305-8573 Ibaraki Japan
| | - Mayuka Kamiyanagi
- Faculty of Pure and Applied Sciences, University of Tsukuba 305-8573 Ibaraki Japan
| | - Osamu Takeuchi
- Faculty of Pure and Applied Sciences, University of Tsukuba 305-8573 Ibaraki Japan
| | - Hirofumi Matsui
- Faculty of Medicine, University of Tsukuba 305-8575 Ibaraki Japan
| | - Hidemi Shigekawa
- Faculty of Pure and Applied Sciences, University of Tsukuba 305-8573 Ibaraki Japan
| |
Collapse
|
9
|
Nasibullin I, Smirnov I, Ahmadi P, Vong K, Kurbangalieva A, Tanaka K. Synthetic prodrug design enables biocatalytic activation in mice to elicit tumor growth suppression. Nat Commun 2022; 13:39. [PMID: 35013295 PMCID: PMC8748823 DOI: 10.1038/s41467-021-27804-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/03/2021] [Indexed: 11/25/2022] Open
Abstract
Considering the intrinsic toxicities of transition metals, their incorporation into drug therapies must operate at minimal amounts while ensuring adequate catalytic activity within complex biological systems. As a way to address this issue, this study investigates the design of synthetic prodrugs that are not only tuned to be harmless, but can be robustly transformed in vivo to reach therapeutically relevant levels. To accomplish this, retrosynthetic prodrug design highlights the potential of naphthylcombretastatin-based prodrugs, which form highly active cytostatic agents via sequential ring-closing metathesis and aromatization. Structural adjustments will also be done to improve aspects related to catalytic reactivity, intrinsic bioactivity, and hydrolytic stability. The developed prodrug therapy is found to possess excellent anticancer activities in cell-based assays. Furthermore, in vivo activation by intravenously administered glycosylated artificial metalloenzymes can also induce significant reduction of implanted tumor growth in mice.
Collapse
Affiliation(s)
- Igor Nasibullin
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Ivan Smirnov
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University, 18 Kremlyovskaya street, Kazan, 420008, Russia
| | - Peni Ahmadi
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Kenward Vong
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Almira Kurbangalieva
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University, 18 Kremlyovskaya street, Kazan, 420008, Russia
| | - Katsunori Tanaka
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University, 18 Kremlyovskaya street, Kazan, 420008, Russia.
- GlycoTargeting Research Laboratory, RIKEN Baton Zone Program, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 O-okayama, Meguro-ku, Tokyo, 152-8552, Japan.
| |
Collapse
|
10
|
Sun M, Yuan M, Kang Y, Qin J, Zhang Y, Duan Y, Wang L, Yao Y. Identification of novel non-toxic and anti-angiogenic α-fluorinated chalcones as potent colchicine binding site inhibitors. J Enzyme Inhib Med Chem 2022; 37:339-354. [PMID: 34979843 PMCID: PMC8741257 DOI: 10.1080/14756366.2021.2014831] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
α-Fluorinated chalcones were prepared and evaluated for their cell growth inhibitory properties against six human cancer cell lines. The most potent chalcone 4c demonstrated excellent selective toxicity against cancer cells versus normal human cells, with IC50 values at nanomolar concentration ranges against 5 cancer cell lines. A further study revealed that 4c could bind to the colchicine site of tubulin, disrupt the cell microtubule networks, and effectively inhibit tubulin polymerisation. Cellular-based mechanism studies elucidated that 4c arrested MGC-803 cell cycle at G2/M phase. In addition, 4c dose-dependently caused Caspase-induced apoptosis of MGC-803 cells through mitochondrial dysfunction. Notably, compound 4c was found to inhibit the HUVECs tube formation, migration, and invasion in vitro. Furthermore, our data suggested that treatment with 4c significantly reduced MGC-803 cells metastasis and proliferation in vitro. Overall, this work showed that chalcone hybrid 4c is a potent inhibitor of tubulin assembly with prominent anti-angiogenesis and anti-cancer properties.
Collapse
Affiliation(s)
- Moran Sun
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, School of Pharmaceutical Sciences, Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Minghua Yuan
- Key Laboratory of Advanced Drug Preparation Technologies, School of Pharmaceutical Sciences, Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Yingying Kang
- Key Laboratory of Advanced Drug Preparation Technologies, School of Pharmaceutical Sciences, Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Jinling Qin
- Key Laboratory of Advanced Drug Preparation Technologies, School of Pharmaceutical Sciences, Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Yixin Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, School of Pharmaceutical Sciences, Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Longfei Wang
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yongfang Yao
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, School of Pharmaceutical Sciences, Ministry of Education, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Guo Y, Wang H, Gerberich JL, Odutola SO, Charlton-Sevcik AK, Li M, Tanpure RP, Tidmore JK, Trawick ML, Pinney KG, Mason RP, Liu L. Imaging-Guided Evaluation of the Novel Small-Molecule Benzosuberene Tubulin-Binding Agent KGP265 as a Potential Therapeutic Agent for Cancer Treatment. Cancers (Basel) 2021; 13:cancers13194769. [PMID: 34638255 PMCID: PMC8507561 DOI: 10.3390/cancers13194769] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/10/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Vascular-disrupting agents promise significant therapeutic efficacy against solid tumors by selectively damaging tumor-associated vasculature. Dynamic BLI and oxygen-enhanced multispectral optoacoustic tomography (OE-MSOT) were used to compare vascular shutdown following administration of KGP265. BLI signal and vascular oxygenation response (ΔsO2) to a gas breathing challenge were both significantly reduced within 2 h indicating vascular disruption, which continued over 24 h. Twice-weekly doses of KGP265 caused a significant growth delay in MDA-MB-231 human breast tumor xenografts and 4T1 syngeneic breast tumors growing orthotopically in mice. Abstract The selective disruption of tumor-associated vasculature represents an attractive therapeutic approach. We have undertaken the first in vivo evaluation of KGP265, a water-soluble prodrug of a benzosuberene-based tubulin-binding agent, and found promising vascular-disrupting activity in three distinct tumor types. Dose escalation in orthotopic MDA-MB-231-luc breast tumor xenografts in mice indicated that higher doses produced more effective vascular shutdown, as revealed by dynamic bioluminescence imaging (BLI). In syngeneic orthotopic 4T1-luc breast and RENCA-luc kidney tumors, dynamic BLI and oxygen enhanced multispectral optoacoustic tomography (OE-MSOT) were used to compare vascular shutdown following the administration of KGP265 (7.5 mg/kg). The BLI signal and vascular oxygenation response (ΔsO2) to a gas breathing challenge were both significantly reduced within 2 h, indicating vascular disruption, which continued over 24 h. A correlative histology confirmed increased necrosis and hemorrhage. Twice-weekly doses of KGP265 caused significant growth delay in both MDA-MB-231 and 4T1 breast tumors, with no obvious systemic toxicity. A combination with carboplatin produced significantly greater tumor growth delay than carboplatin alone, though significant carboplatin-associated toxicity was observed (whole-body weight loss). KGP265 was found to be effective at low concentrations, generating long-term vascular shutdown and tumor growth delay, thus providing strong rationale for further development, particularly in combination therapies.
Collapse
Affiliation(s)
- Yihang Guo
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha 410013, China
| | - Honghong Wang
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jeni L. Gerberich
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
| | - Samuel O. Odutola
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Amanda K. Charlton-Sevcik
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Maoping Li
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rajendra P. Tanpure
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Justin K. Tidmore
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Ralph P. Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: (R.P.M.); (L.L.)
| | - Li Liu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: (R.P.M.); (L.L.)
| |
Collapse
|
12
|
Zhao J, Stevens CH, Boyd AW, Ooi L, Bartlett PF. Role of EphA4 in Mediating Motor Neuron Death in MND. Int J Mol Sci 2021; 22:9430. [PMID: 34502339 PMCID: PMC8430883 DOI: 10.3390/ijms22179430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 11/26/2022] Open
Abstract
Motor neuron disease (MND) comprises a group of fatal neurodegenerative diseases with no effective cure. As progressive motor neuron cell death is one of pathological characteristics of MND, molecules which protect these cells are attractive therapeutic targets. Accumulating evidence indicates that EphA4 activation is involved in MND pathogenesis, and inhibition of EphA4 improves functional outcomes. However, the underlying mechanism of EphA4's function in MND is unclear. In this review, we first present results to demonstrate that EphA4 signalling acts directly on motor neurons to cause cell death. We then review the three most likely mechanisms underlying this effect.
Collapse
Affiliation(s)
- Jing Zhao
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia;
| | - Claire H. Stevens
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Andrew W. Boyd
- School of Medicine, University of Queensland, Brisbane, QLD 4072, Australia;
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Perry F. Bartlett
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia;
| |
Collapse
|
13
|
Liu L, O’Kelly D, Schuetze R, Carlson G, Zhou H, Trawick ML, Pinney KG, Mason RP. Non-Invasive Evaluation of Acute Effects of Tubulin Binding Agents: A Review of Imaging Vascular Disruption in Tumors. Molecules 2021; 26:2551. [PMID: 33925707 PMCID: PMC8125421 DOI: 10.3390/molecules26092551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor vasculature proliferates rapidly, generally lacks pericyte coverage, and is uniquely fragile making it an attractive therapeutic target. A subset of small-molecule tubulin binding agents cause disaggregation of the endothelial cytoskeleton leading to enhanced vascular permeability generating increased interstitial pressure. The resulting vascular collapse and ischemia cause downstream hypoxia, ultimately leading to cell death and necrosis. Thus, local damage generates massive amplification and tumor destruction. The tumor vasculature is readily accessed and potentially a common target irrespective of disease site in the body. Development of a therapeutic approach and particularly next generation agents benefits from effective non-invasive assays. Imaging technologies offer varying degrees of sophistication and ease of implementation. This review considers technological strengths and weaknesses with examples from our own laboratory. Methods reveal vascular extent and patency, as well as insights into tissue viability, proliferation and necrosis. Spatiotemporal resolution ranges from cellular microscopy to single slice tomography and full three-dimensional views of whole tumors and measurements can be sufficiently rapid to reveal acute changes or long-term outcomes. Since imaging is non-invasive, each tumor may serve as its own control making investigations particularly efficient and rigorous. The concept of tumor vascular disruption was proposed over 30 years ago and it remains an active area of research.
Collapse
Affiliation(s)
- Li Liu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Devin O’Kelly
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Regan Schuetze
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Graham Carlson
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Heling Zhou
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Ralph P. Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| |
Collapse
|
14
|
Role of protein S-Glutathionylation in cancer progression and development of resistance to anti-cancer drugs. Arch Biochem Biophys 2021; 704:108890. [PMID: 33894196 DOI: 10.1016/j.abb.2021.108890] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/16/2022]
Abstract
The survival, functioning and proliferation of mammalian cells are highly dependent on the cellular response and adaptation to changes in their redox environment. Cancer cells often live in an altered redox environment due to aberrant neo-vasculature, metabolic reprogramming and dysregulated proliferation. Thus, redox adaptations are critical for their survival. Glutathione plays an essential role in maintaining redox homeostasis inside the cells by binding to redox-sensitive cysteine residues in proteins by a process called S-glutathionylation. S-Glutathionylation not only protects the labile cysteine residues from oxidation, but also serves as a sensor of redox status, and acts as a signal for stimulation of downstream processes and adaptive responses to ensure redox equilibrium. The present review aims to provide an updated overview of the role of the unique redox adaptations during carcinogenesis and cancer progression, focusing on their dependence on S-glutathionylation of specific redox-sensitive proteins involved in a wide range of processes including signalling, transcription, structural maintenance, mitochondrial functions, apoptosis and protein recycling. We also provide insights into the role of S-glutathionylation in the development of resistance to chemotherapy. Finally, we provide a strong rationale for the development of redox targeting drugs for treatment of refractory/resistant cancers.
Collapse
|
15
|
Lombardi M, Gabrielli M, Adinolfi E, Verderio C. Role of ATP in Extracellular Vesicle Biogenesis and Dynamics. Front Pharmacol 2021; 12:654023. [PMID: 33790800 PMCID: PMC8006391 DOI: 10.3389/fphar.2021.654023] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/09/2021] [Indexed: 12/18/2022] Open
Abstract
Adenosine triphosphate (ATP) is among the molecules involved in the immune response. It acts as danger signal that promotes inflammation by activating both P2X and P2Y purinergic receptors expressed in immune cells, including microglia, and tumor cells. One of the most important receptors implicated in ATP-induced inflammation is P2X7 receptor (P2X7R). The stimulation of P2X7R by high concentration of ATP results in cell proliferation, inflammasome activation and shedding of extracellular vesicles (EVs). EVs are membrane structures released by all cells, which contain a selection of donor cell components, including proteins, lipids, RNA and ATP itself, and are able to transfer these molecules to target cells. ATP stimulation not only promotes EV production from microglia but also influences EV composition and signaling to the environment. In the present review, we will discuss the current knowledge on the role of ATP in the biogenesis and dynamics of EVs, which exert important functions in physiology and pathophysiology.
Collapse
Affiliation(s)
- Marta Lombardi
- CNR Institute of Neuroscience, Research Labs-University Milano-Bicocca, Vedano al Lambro, Italy
| | - Martina Gabrielli
- CNR Institute of Neuroscience, Research Labs-University Milano-Bicocca, Vedano al Lambro, Italy
| | - Elena Adinolfi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Claudia Verderio
- CNR Institute of Neuroscience, Research Labs-University Milano-Bicocca, Vedano al Lambro, Italy
| |
Collapse
|
16
|
Shiah HS, Chiang NJ, Lin CC, Yen CJ, Tsai HJ, Wu SY, Su WC, Chang KY, Wang CC, Chang JY, Chen LT. Phase I Dose-Escalation Study of SCB01A, a Microtubule Inhibitor with Vascular Disrupting Activity, in Patients with Advanced Solid Tumors. Oncologist 2020; 26:e567-e579. [PMID: 33245172 DOI: 10.1002/onco.13612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 11/15/2020] [Indexed: 11/10/2022] Open
Abstract
LESSONS LEARNED SCB01A is a novel microtubule inhibitor with vascular disrupting activity. This first-in-human study demonstrated SCB01A safety, pharmacokinetics, and preliminary antitumor activity. SCB01A is safe and well tolerated in patients with advanced solid malignancies with manageable neurotoxicity. BACKGROUND SCB01A, a novel microtubule inhibitor, has vascular disrupting activity. METHODS In this phase I dose-escalation and extension study, patients with advanced solid tumors were administered intravenous SCB01A infusions for 3 hours once every 21 days. Rapid titration and a 3 + 3 design escalated the dose from 2 mg/m2 to the maximum tolerated dose (MTD) based on dose-limiting toxicity (DLT). SCB01A-induced cellular neurotoxicity was evaluated in dorsal root ganglion cells. The primary endpoint was MTD. Safety, pharmacokinetics (PK), and tumor response were secondary endpoints. RESULTS Treatment-related adverse events included anemia, nausea, vomiting, fatigue, fever, and peripheral sensorimotor neuropathy. DLTs included grade 4 elevated creatine phosphokinase (CPK) in the 4 mg/m2 cohort; grade 3 gastric hemorrhage in the 6.5 mg/m2 cohort; grade 2 thromboembolic event in the 24 mg/m2 cohort; and grade 3 peripheral sensorimotor neuropathy, grade 3 elevated aspartate aminotransferase, and grade 3 hypertension in the 32 mg/m2 cohort. The MTD was 24 mg/m2 , and average half-life was ~2.5 hours. The area under the curve-dose response relationship was linear. Nineteen subjects were stable after two cycles. The longest treatment lasted 24 cycles. SCB01A-induced neurotoxicity was reversible in vitro. CONCLUSION The MTD of SCB01A was 24 mg/m2 every 21 days; it is safe and tolerable in patients with solid tumors.
Collapse
Affiliation(s)
- Her-Shyong Shiah
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Division of Hematology and Oncology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Nai-Jung Chiang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Jui Yen
- Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Jen Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shang-Yin Wu
- Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu-Chou Su
- Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | - Jang-Yang Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, Kaohisung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
17
|
Suzuki T, Tanaka M, Sasaki M, Ichikawa H, Nishie H, Kataoka H. Vascular Shutdown by Photodynamic Therapy Using Talaporfin Sodium. Cancers (Basel) 2020; 12:cancers12092369. [PMID: 32825648 PMCID: PMC7563359 DOI: 10.3390/cancers12092369] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 11/16/2022] Open
Abstract
Photodynamic therapy (PDT) is an attractive cancer treatment modality. Talaporfin sodium, a second-generation photosensitizer, results in lower systemic toxicity and relatively better selective tumor destruction than first-generation photosensitizers. However, the mechanism through which PDT induces vascular shutdown is unclear. In this study, the in vitro effects of talaporfin sodium-based PDT on human umbilical vein endothelial cells (HUVECs) were determined through cell viability and endothelial tube formation assays, and evaluation of the tubulin and F-actin dynamics and myosin light chain (MLC) phosphorylation. Additionally, the effects on tumor blood flow and tumor vessel destruction were assessed in vivo. In the HUVECs, talaporfin sodium-based PDT induced endothelial tube destruction and microtubule depolymerization, triggering the formation of F-actin stress fibers and a significant increase in MLC phosphorylation. However, pretreatment with the Rho-associated protein kinase (ROCK) inhibitor, Y27632, completely prevented PDT-induced stress fiber formation and MLC phosphorylation. The in vivo analysis and pathological examination revealed that the PDT had significantly decreased the tumor blood flow and the active area of the tumor vessel. We concluded that talaporfin sodium-based PDT induces the shutdown of existing tumor vessels via the RhoA/ROCK pathway by activating the Rho-GTP pathway and decreasing the tumor blood flow.
Collapse
Affiliation(s)
| | - Mamoru Tanaka
- Correspondence: ; Tel.: +81-52-853-8211; Fax: +81-52-852-0952
| | | | | | | | | |
Collapse
|
18
|
Zheng Y, Gong J, Zhen Y. Focal adhesion kinase is activated by microtubule-depolymerizing agents and regulates membrane blebbing in human endothelial cells. J Cell Mol Med 2020; 24:7228-7238. [PMID: 32452639 PMCID: PMC7339229 DOI: 10.1111/jcmm.15273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/01/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
Microtubule-depolymerizing agents can selectively disrupt tumor vessels via inducing endothelial membrane blebbing. However, the mechanism regulating blebbing is largely unknown. IMB5046 is a newly discovered microtubule-depolymerizing agent. Here, the functions of focal adhesion kinase (FAK) during IMB5046-induced blebbing and the relevant mechanism are studied. We found that IMB5046 induced membrane blebbing and reassembly of focal adhesions in human vascular endothelial cells. Both FAK inhibitor and knock-down expression of FAK inhibited IMB5046-induced blebbing. Mechanism study revealed that IMB5046 induced the activation of FAK via GEF-H1/ Rho/ ROCK/ MLC2 pathway. cRGD peptide, a ligand of integrin, also blocked IMB5046-induced blebbing. After activation, FAK further promoted the phosphorylation of MLC2. This positive feedback loop caused more intensive actomyosin contraction and continuous membrane blebbing. FAK inhibitor blocked membrane blebbing via inhibiting actomyosin contraction, and stimulated stress fibre formation via promoting the phosphorylation of HSP27. Conclusively, these results demonstrate that FAK is a molecular switch controlling endothelial blebbing and stress fibre formation. Our study provides a new molecular mechanism for microtubule-depolymerizing agents to be used as vascular disrupting agents.
Collapse
Affiliation(s)
- Yan‐Bo Zheng
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jian‐Hua Gong
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yong‐Su Zhen
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
19
|
The influence of hypoxia and energy depletion on the response of endothelial cells to the vascular disrupting agent combretastatin A-4-phosphate. Sci Rep 2020; 10:9926. [PMID: 32555222 PMCID: PMC7303175 DOI: 10.1038/s41598-020-66568-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/15/2020] [Indexed: 12/25/2022] Open
Abstract
Combretastatin A-4 phosphate (CA4P) is a microtubule-disrupting tumour-selective vascular disrupting agent (VDA). CA4P activates the actin-regulating RhoA-GTPase/ ROCK pathway, which is required for full vascular disruption. While hypoxia renders tumours resistant to many conventional therapies, little is known about its influence on VDA activity. Here, we found that active RhoA and ROCK effector phospho-myosin light chain (pMLC) were downregulated in endothelial cells by severe hypoxia. CA4P failed to activate RhoA/ROCK/pMLC but its activity was restored upon reoxygenation. Hypoxia also inhibited CA4P-mediated actinomyosin contractility, VE-cadherin junction disruption and permeability rise. Glucose withdrawal downregulated pMLC, and coupled with hypoxia, reduced pMLC faster and more profoundly than hypoxia alone. Concurrent inhibition of glycolysis (2-deoxy-D-glucose, 2DG) and mitochondrial respiration (rotenone) caused profound actin filament loss, blocked RhoA/ROCK signalling and rendered microtubules CA4P-resistant. Withdrawal of the metabolism inhibitors restored the cytoskeleton and CA4P activity. The AMP-activated kinase AMPK was investigated as a potential mediator of pMLC downregulation. Pharmacological AMPK activators that generate AMP, unlike allosteric activators, downregulated pMLC but only when combined with 2DG and/or rotenone. Altogether, our results suggest that Rho/ROCK and actinomyosin contractility are regulated by AMP/ATP levels independently of AMPK, and point to hypoxia/energy depletion as potential modifiers of CA4P response.
Collapse
|
20
|
Wong T, Narayanan S, Brown DP, Chen ZS. Synthesis and Cytotoxicity Studies of Stilbene Long-Chain Fatty Acid Conjugates. JOURNAL OF NATURAL PRODUCTS 2020; 83:1563-1570. [PMID: 32243160 DOI: 10.1021/acs.jnatprod.0c00027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A series of 16 conjugates of the tubulin polymerization inhibitor combretastatin A4 (CA-4) and other functionally related stilbene with four 18-carbon fatty acids, namely, stearic, oleic, linoleic, and linolenic acids, have been synthesized in good yields. These new derivatives have been evaluated against the KB-3-1 (human epidermoid carcinoma), NCI-H460 (human lung cancer), HEK293 (human embryonic kidney), and MCF-7 (human breast adenocarcinoma) cell lines for antiproliferative activity, with the exhibited cytotoxic activities comparable with those of CA-4 and colchicine. Compounds 22 and 23, CA-4 conjugates of linoleic and linolenic acids, respectively, were determined to have exhibited the most active in vitro assays, with compound 23 exhibiting very similar activity to the parent compound against the NCI-H460 cell line. Our studies further delineated the structurally required Z-geometry of the stilbene moiety and that conjugation of the less active E-stilbenes with the most active fatty acid had minimal or no improvement in their respective activities.
Collapse
Affiliation(s)
- Thomas Wong
- Department of Pharmaceutical Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Silpa Narayanan
- Department of Pharmaceutical Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - David P Brown
- Department of Chemistry, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| |
Collapse
|
21
|
Colchicine-Binding Site Inhibitors from Chemistry to Clinic: A Review. Pharmaceuticals (Basel) 2020; 13:ph13010008. [PMID: 31947889 PMCID: PMC7168938 DOI: 10.3390/ph13010008] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023] Open
Abstract
It is over 50 years since the discovery of microtubules, and they have become one of the most important drug targets for anti-cancer therapies. Microtubules are predominantly composed of the protein tubulin, which contains a number of different binding sites for small-molecule drugs. There is continued interest in drug development for compounds targeting the colchicine-binding site of tubulin, termed colchicine-binding site inhibitors (CBSIs). This review highlights CBSIs discovered through diverse sources: from natural compounds, rational design, serendipitously and via high-throughput screening. We provide an update on CBSIs reported in the past three years and discuss the clinical status of CBSIs. It is likely that efforts will continue to develop CBSIs for a diverse set of cancers, and this review provides a timely update on recent developments.
Collapse
|
22
|
Chen JL, Zhang H, Huang XQ, Wan HY, Li J, Fan XX, Luo KQ, Wang J, Zhu XM, Wang J. Antiangiogenesis-Combined Photothermal Therapy in the Second Near-Infrared Window at Laser Powers Below the Skin Tolerance Threshold. NANO-MICRO LETTERS 2019; 11:93. [PMID: 34138046 PMCID: PMC7770887 DOI: 10.1007/s40820-019-0327-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/14/2019] [Indexed: 05/16/2023]
Abstract
Photothermal agents with strong light absorption in the second near-infrared (NIR-II) region (1000-1350 nm) are strongly desired for successful photothermal therapy (PTT). In this work, titania-coated Au nanobipyramids (NBP@TiO2) with a strong plasmon resonance in the NIR-II window were synthesized. The NBP@TiO2 nanostructures have a high photothermal conversion efficiency of (93.3 ± 5.2)% under 1064-nm laser irradiation. They are also capable for loading an anticancer drug combretastatin A-4 phosphate (CA4P). In vitro PTT studies reveal that 1064-nm laser irradiation can efficiently ablate human lung cancer A549 cells and enhance the anticancer effect of CA4P. Moreover, the CA4P-loaded NBP@TiO2 nanostructures combined with PTT induce a synergistic antiangiogenesis effect. In vivo studies show that such CA4P-loaded NBP@TiO2 nanostructures under mild 1064-nm laser irradiation at an optical power density of 0.4 W cm-2, which is lower than the skin tolerance threshold value, exhibit a superior antitumor effect. This work presents not only the development of the NBP@TiO2 nanostructures as a novel photothermal agent responsive in the NIR-II window but also a unique combined chemo-photothermal therapy strategy for cancer therapy.
Collapse
Affiliation(s)
- Jian-Li Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Han Zhang
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People's Republic of China
| | - Xue-Qin Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Hong-Ye Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Jie Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Xing-Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, People's Republic of China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Targets Research and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Xiao-Ming Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China.
| | - Jianfang Wang
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People's Republic of China.
| |
Collapse
|
23
|
The Vascular Disrupting Agent CA4P Improves the Antitumor Efficacy of CAR-T Cells in Preclinical Models of Solid Human Tumors. Mol Ther 2019; 28:75-88. [PMID: 31672285 DOI: 10.1016/j.ymthe.2019.10.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/29/2019] [Accepted: 10/10/2019] [Indexed: 11/20/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy remains relatively ineffective against solid tumors due to inadequate infiltration and in vivo expansion of CAR-T cells. Unlike hematological malignancies, solid tumors have vascular barriers that hinder CAR-T cells from reaching the tumor site. Here, we demonstrated that combretastatin A-4 phosphate (CA4P), a vascular disrupting agent (VDA), can significantly improve the infiltration ability of CAR-T cells in solid tumors as evidenced by elevated levels of IFN-γ. Moreover, combined treatment with CA4P and CAR-T cells greatly increased the therapeutic efficiency of the CAR-T cells in subcutaneous ovarian cancer mouse xenograft models and patient-derived xenograft (PDX) models of colon and ovarian carcinoma. Our findings highlight CA4P as an effective antitumor agent candidate for combination with CAR-T cells in clinical applications to treat solid tumors.
Collapse
|
24
|
Zeng Q, Liu Y, Song Y, Feng B, Xu P, Shan B, Liao Z, Liu K, Zhong Y, Chen L, Su D. A UHPLC-MS/MS method coupled with simple and efficient alkaline hydrolysis for free and total determination of conjugate nanomedicine: Pharmacokinetic and biodistribution study of poly (l-glutamic acid)-graft-methoxy poly (ethylene glycol)/combretastatin A4. J Pharm Biomed Anal 2019; 169:215-224. [PMID: 30877933 DOI: 10.1016/j.jpba.2019.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 02/26/2019] [Accepted: 03/01/2019] [Indexed: 01/22/2023]
Abstract
Poly (l-glutamic acid)-Combretastatin A4 conjugate (PLG-CA4) is a novel nano-anticancer drug. For macromolecule conjugate nanomedicine, its pharmacology mechanism is closely related to the pharmacokinetic profiles in vivo. It is a great significance that evaluates this polymer drug combined by covalently bound via studying the pharmacokinetics and distribution characteristics. Therefore, it is urgent to develop a simple, accurate and practical analytical method for such conjugated polymers combined by covalently bound. In this study, a simple and complete alkali hydrolysis was designed and optimized for the total CA4 concentrations obtained from PLG-CA4. Ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method with multiple-reaction monitoring (MRM) mode and the internal standard (IS) were adopted to develop a sensitive and accurate method satisfied both free and total determination of PLG-CA4 in biosamples. The method was validated which showed good linearity over a wide concentration range (R2 > 0.99), and the intra- and inter-day assay variability was less than 15% for CA4. The mean extraction recoveries of CA4 from plasma were all more than 80.0%. Furthermore, the method was applied to the study of pharmacokinetics (PK) and tissue distribution of PLG-CA4 in tumor-bearing nude mice. PLG-CA4 significantly prolonged retention time and enhanced distribution of CA4 in tumor.
Collapse
Affiliation(s)
- Qiang Zeng
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Yali Liu
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Yonggui Song
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Bingwei Feng
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Pengfei Xu
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Baixi Shan
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Zhou Liao
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Kuangyi Liu
- SCIEX, Analytical Instrument Trading Co., Office Room 502, 5/F, Bldg 1, 518 North FU quan Road, IBP Changning District, Shanghai, 200335, PR China
| | - Youbao Zhong
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Lai Chen
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Dan Su
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China.
| |
Collapse
|
25
|
Sun CW, Wu LC, Wankhede M, Wang D, Thoerner J, Woody L, Sorg BS, Townes TM, Terman DS. Exogenous sickle erythrocytes combined with vascular disruption trigger disseminated tumor vaso-occlusion and lung tumor regression. JCI Insight 2019; 4:125535. [PMID: 30944254 DOI: 10.1172/jci.insight.125535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/14/2019] [Indexed: 12/31/2022] Open
Abstract
Hypoxic tumor niches are chief causes of treatment resistance and tumor recurrence. Sickle erythrocytes' (SSRBCs') intrinsic oxygen-sensing functionality empowers them to access such hypoxic niches wherein they form microaggregates that induce focal vessel closure. In search of measures to augment the scale of SSRBC-mediated tumor vaso-occlusion, we turned to the vascular disrupting agent, combretastatin A-4 (CA-4). CA-4 induces selective tumor endothelial injury, blood stasis, and hypoxia but fails to eliminate peripheral tumor foci. In this article, we show that introducing deoxygenated SSRBCs into tumor microvessels treated with CA-4 and sublethal radiation (SR) produces a massive surge of tumor vaso-occlusion and broadly propagated tumor infarctions that engulfs treatment-resistant hypoxic niches and eradicates established lung tumors. Tumor regression was histologically corroborated by significant treatment effect. Treated tumors displayed disseminated microvessels occluded by tightly packed SSRBCs along with widely distributed pimidazole-positive hypoxic tumor cells. Humanized HbS-knockin mice (SSKI) but not HbA-knockin mice (AAKI) showed a similar treatment response underscoring SSRBCs as the paramount tumoricidal effectors. Thus, CA-4-SR-remodeled tumor vessels license SSRBCs to produce an unprecedented surge of tumor vaso-occlusion and infarction that envelops treatment-resistant tumor niches resulting in complete tumor regression. Strategically deployed, these innovative tools constitute a major conceptual advance with compelling translational potential.
Collapse
Affiliation(s)
- Chiao-Wang Sun
- Department of Biochemistry and Molecular Genetics, University of Alabama School of Medicine at Birmingham, Birmingham, Alabama, USA
| | - Li-Chen Wu
- Department of Biochemistry and Molecular Genetics, University of Alabama School of Medicine at Birmingham, Birmingham, Alabama, USA
| | - Mamta Wankhede
- Department of Biomedical Engineering, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dezhi Wang
- Department of Pathology, University of Alabama School of Medicine at Birmingham, Birmingham, Alabama, USA
| | - Jutta Thoerner
- Histopathology Section, Hospital of the Monterey Peninsula, Monterey, California, USA
| | - Lawrence Woody
- Histopathology Section, Hospital of the Monterey Peninsula, Monterey, California, USA
| | - Brian S Sorg
- Cancer Diagnosis Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Tim M Townes
- Department of Biochemistry and Molecular Genetics, University of Alabama School of Medicine at Birmingham, Birmingham, Alabama, USA
| | - David S Terman
- Department of Biochemistry and Molecular Genetics, University of Alabama School of Medicine at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
26
|
Oxazole-Bridged Combretastatin A-4 Derivatives with Tethered Hydroxamic Acids: Structure⁻Activity Relations of New Inhibitors of HDAC and/or Tubulin Function. Int J Mol Sci 2019; 20:ijms20020383. [PMID: 30658435 PMCID: PMC6359144 DOI: 10.3390/ijms20020383] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/03/2019] [Accepted: 01/14/2019] [Indexed: 11/30/2022] Open
Abstract
New inhibitors of tubulin polymerization and/or histone deacetylase (HDAC) activity were synthesized by attaching alkyl tethered hydroxamic acid appendages of varying length to oxazole-bridged combretastatin A-4 analogous caps. While their antiproliferative and microtubule disrupting effect was most pronounced for derivatives with short spacers, HDAC inhibition was strongest for those with longer spacers. These findings were further supported by computational methods such as structure-based docking experiments exploring the target interactions of the derivatives with varying linkers. For instance, compounds featuring short four-atom spacers between cap and hydroxamic acid inhibited the growth of various cancer cell lines and human endothelial hybrid cells with IC50 values in the low nanomolar range. In line with their ability to inhibit the microtubule assembly, four- and five-atom spacered hydroxamic acids caused an accumulation of 518A2 melanoma cells in G2/M phase, whereas a compound featuring a six-atom spacer and performing best in HDAC inhibition, induced a G1 arrest in these cells. All these beneficial anticancer activities together with their selectivity for cancer cells over non-malignant cells, point out the great potential of these novel pleiotropic HDAC and tubulin inhibitors as drug candidates for cancer therapy.
Collapse
|
27
|
Brown AW, Holmes T, Fisher M, Tozer GM, Harrity JPA, Kanthou C. Evaluation of Sydnone-Based Analogues of Combretastatin A-4 Phosphate (CA4P) as Vascular Disrupting Agents for Use in Cancer Therapy. ChemMedChem 2018; 13:2618-2626. [PMID: 30281922 DOI: 10.1002/cmdc.201800567] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Indexed: 11/10/2022]
Abstract
The combretastatins have attracted significant interest as small-molecule therapies for cancer due to their ability to function as vascular disrupting agents. We have successfully prepared a range of combretastatin analogues that are based on a novel sydnone heterocycle core, and their potential as tubulin binders has been assessed in vitro and in vivo. The most potent candidate was found to disrupt microtubules and affect cellular morphology at sub-micromolar levels. Moreover, it was found to bind reversibly to tubulin and significantly increase endothelial cell monolayer permeability, in a similar manner to combretastatin A4. Surprisingly, the compound did not exhibit efficacy in vivo, possibly due to rapid metabolism.
Collapse
Affiliation(s)
- Andrew W Brown
- Department of Chemistry, The University of Sheffield, Dainton Building, Brook Hill, Sheffield, S3 7HF, UK.,Department of Oncology & Metabolism, The University of Sheffield, The Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Toby Holmes
- Department of Oncology & Metabolism, The University of Sheffield, The Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Matthew Fisher
- Department of Oncology & Metabolism, The University of Sheffield, The Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Gillian M Tozer
- Department of Oncology & Metabolism, The University of Sheffield, The Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Joseph P A Harrity
- Department of Chemistry, The University of Sheffield, Dainton Building, Brook Hill, Sheffield, S3 7HF, UK
| | - Chryso Kanthou
- Department of Oncology & Metabolism, The University of Sheffield, The Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| |
Collapse
|
28
|
Vicente-Blázquez A, González M, Álvarez R, Del Mazo S, Medarde M, Peláez R. Antitubulin sulfonamides: The successful combination of an established drug class and a multifaceted target. Med Res Rev 2018; 39:775-830. [PMID: 30362234 DOI: 10.1002/med.21541] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/02/2018] [Accepted: 09/06/2018] [Indexed: 12/13/2022]
Abstract
Tubulin, the microtubules and their dynamic behavior are amongst the most successful antitumor, antifungal, antiparasitic, and herbicidal drug targets. Sulfonamides are exemplary drugs with applications in the clinic, in veterinary and in the agrochemical industry. This review summarizes the actual state and recent progress of both fields looking from the double point of view of the target and its drugs, with special focus onto the structural aspects. The article starts with a brief description of tubulin structure and its dynamic assembly and disassembly into microtubules and other polymers. Posttranslational modifications and the many cellular means of regulating and modulating tubulin's biology are briefly presented in the tubulin code. Next, the structurally characterized drug binding sites, their occupying drugs and the effects they induce are described, emphasizing on the structural requirements for high potency, selectivity, and low toxicity. The second part starts with a summary of the favorable and highly tunable combination of physical-chemical and biological properties that render sulfonamides a prototypical example of privileged scaffolds with representatives in many therapeutic areas. A complete description of tubulin-binding sulfonamides is provided, covering the different species and drug sites. Some of the antimitotic sulfonamides have met with very successful applications and others less so, thus illustrating the advances, limitations, and future perspectives of the field. All of them combine in a mechanism of action and a clinical outcome that conform efficient drugs.
Collapse
Affiliation(s)
- Alba Vicente-Blázquez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Myriam González
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Sara Del Mazo
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Manuel Medarde
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| |
Collapse
|
29
|
Hura N, Sawant AV, Kumari A, Guchhait SK, Panda D. Combretastatin-Inspired Heterocycles as Antitubulin Anticancer Agents. ACS OMEGA 2018; 3:9754-9769. [PMID: 31459105 PMCID: PMC6644768 DOI: 10.1021/acsomega.8b00996] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/09/2018] [Indexed: 06/10/2023]
Abstract
Combretastatin (CA-4) and its analogues are undergoing several clinical trials for treating different types of tumors. In this work, the antiproliferative activity of a series of 2-aminoimidazole-carbonyl analogs of clinically relevant combretastatins A-4 (CA-4) and A-1 was evaluated using a cell-based assay. Among the compounds tested, C-13 and C-21 displayed strong antiproliferative activities against HeLa cells. C-13 inhibited the proliferation of lung carcinoma (A549) cells more potently than combretastatin A-4. C-13 also retarded the migration of A549 cells. Interestingly, C-13 displayed much stronger antiproliferative effects against breast carcinoma and skin melanoma cells compared to noncancerous breast epithelial and skin fibroblast cells. C-13 strongly disassembled cellular microtubules, perturbed the localization of EB1 protein, inhibited mitosis in cultured cells, and bound to tubulin at the colchicine site and inhibited the polymerization of reconstituted microtubules in vitro. C-13 treatment increased the level of reactive oxygen species and induced apoptosis via poly(ADP-ribose) polymerase-cleavage in HeLa cells. The results revealed the importance of the 2-aminoimidazole-carbonyl motif as a double bond replacement in combretastatin and indicated a pharmacodynamically interesting pattern of H-bond acceptors/donors and requisite syn-templated aryls.
Collapse
Affiliation(s)
- Neha Hura
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), S. A. S. Nagar, Mohali, Punjab 160062, India
| | - Avishkar V. Sawant
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Bombay, Mumbai 400076, India
| | - Anuradha Kumari
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Bombay, Mumbai 400076, India
| | - Sankar K. Guchhait
- Department
of Medicinal Chemistry, National Institute
of Pharmaceutical Education and Research (NIPER), S. A. S. Nagar, Mohali, Punjab 160062, India
| | - Dulal Panda
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
30
|
New (arene)ruthenium(II) complexes of 4‑aryl‑4H‑naphthopyrans with anticancer and anti-vascular activities. J Inorg Biochem 2018; 184:69-78. [DOI: 10.1016/j.jinorgbio.2018.03.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 03/06/2018] [Accepted: 03/22/2018] [Indexed: 01/06/2023]
|
31
|
Liu Y, Kim YJ, Siriwon N, Rohrs JA, Yu Z, Wanga P. Combination drug delivery via multilamellar vesicles enables targeting of tumor cells and tumor vasculature. Biotechnol Bioeng 2018; 115:1403-1415. [PMID: 29457630 DOI: 10.1002/bit.26566] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/16/2018] [Accepted: 02/08/2018] [Indexed: 12/15/2022]
Abstract
Blood vessel development is critical for the continued growth and progression of solid tumors and, therefore, makes an attractive target for improving cancer therapy. Indeed, vascular-targeted therapies have been extensively explored but they have shown minimal efficacy as monotherapies. Combretastatin A4 (CA-4) is a tubulin-binding vascular disrupting agent that selectively targets the established tumor endothelium, causing rapid vascular beak down. Despite its potent anticancer potential, the drug has dose-limiting side effects, particularly in the form of cardiovascular toxicity. Furthermore, its poor aqueous solubility and the resulting limited bioavailability hinder its antitumor activity in the clinic. To improve the therapeutic efficacy of CA-4, we investigated its application as a combination therapy with doxorubicin (Dox) in a tumor vasculature targeted delivery vehicle: peptide-modified cross-linked multilamellar liposomal vesicles (cMLVs). In vitro cell culture studies showed that a tumor vasculature-targeting peptide, RIF7, could facilitate higher cellular uptake of drug-loaded cMLVs, and consequently enhance the antitumor efficacy in both drug resistant B16 mouse melanoma and human MDA-MB-231 breast cancer cells. In vivo, upon intravenous injection, targeted cMLVs could efficiently deliver both Dox and CA-4 to significantly slow tumor growth through the specific interaction of the targeting peptide with its receptor on the surface of tumor vasculature. This study demonstrates the potential of our novel targeted combination therapy delivery vehicle to improve the outcome of cancer treatment.
Collapse
Affiliation(s)
- Yarong Liu
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California
| | - Yu J Kim
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California
| | - Natnaree Siriwon
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California
| | - Jennifer A Rohrs
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, Guangdong, China
| | - Pin Wanga
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California.,Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California.,Department of Biomedical Engineering, University of Southern California, Los Angeles, California
| |
Collapse
|
32
|
Liu Z, Li J, Li S, Li G, Sharpless KB, Wu P. SuFEx Click Chemistry Enabled Late-Stage Drug Functionalization. J Am Chem Soc 2018; 140:2919-2925. [PMID: 29451783 DOI: 10.1021/jacs.7b12788] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sulfur(VI) Fluoride Exchange (SuFEx) is a new family of click chemistry transformations which relies on readily available materials to produce compounds bearing the SVI-F motif. The potential of SuFEx in drug discovery has just started to be explored. We report the first method of SuFEx chemistry for the conversion of phenolic compounds to their respective arylfluorosulfate derivatives in situ in 96-well plates. This method is compatible with automated synthesis and screening to quickly assess the biological activities of the in situ generated, crude products. Using this method, we perform late-stage functionalization of a panel of known anticancer drugs to generate the corresponding arylfluorosulfates. These in situ generated arylfluorosulfates are directly tested in a cancer-cell growth inhibition assay in parallel with their phenolic precursors. We discover three arylfluorosulfates that exhibit improved anticancer cell proliferation activities compared to their phenol precursors. Among these three compounds, the fluorosulfate derivative of Fulvestrant possesses significantly enhanced activity to down-regulate estrogen receptor (ER) expression in ER+ breast cancer cell line MCF-7 and the fluorosulfate derivative of Combretastatin A4-a general anticancer drug currently being evaluated under clinical trials-exhibits a 70-fold increase in potency in the drug resistant colon cancer cell line HT-29.
Collapse
Affiliation(s)
- Zilei Liu
- Department of Molecular Medicine, The Scripps Research Institute , La Jolla, California 92037, United States.,Department of Chemistry, The Scripps Research Institute , La Jolla, California 92037, United States
| | - Jie Li
- Department of Molecular Medicine, The Scripps Research Institute , La Jolla, California 92037, United States.,Department of Chemistry, The Scripps Research Institute , La Jolla, California 92037, United States
| | - Suhua Li
- Department of Chemistry, The Scripps Research Institute , La Jolla, California 92037, United States
| | - Gencheng Li
- Department of Chemistry, The Scripps Research Institute , La Jolla, California 92037, United States
| | - K Barry Sharpless
- Department of Chemistry, The Scripps Research Institute , La Jolla, California 92037, United States
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute , La Jolla, California 92037, United States
| |
Collapse
|
33
|
Galmarini CM, Martin M, Bouchet BP, Guillen-Navarro MJ, Martínez-Diez M, Martinez-Leal JF, Akhmanova A, Aviles P. Plocabulin, a novel tubulin-binding agent, inhibits angiogenesis by modulation of microtubule dynamics in endothelial cells. BMC Cancer 2018; 18:164. [PMID: 29415678 PMCID: PMC5803861 DOI: 10.1186/s12885-018-4086-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/31/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Vascular supply of tumors is one of the main targets for cancer therapy. Here, we investigated if plocabulin (PM060184), a novel marine-derived microtubule-binding agent, presents antiangiogenic and vascular-disrupting activities. METHODS The effects of plocabulin on microtubule network and dynamics were studied on HUVEC endothelial cells. We have also studied its effects on capillary tube structures formation or destabilization in three-dimensional collagen matrices. In vivo experiments were performed on different tumor cell lines. RESULTS In vitro studies show that, at picomolar concentrations, plocabulin inhibits microtubule dynamics in endothelial cells. This subsequently disturbs the microtubule network inducing changes in endothelial cell morphology and causing the collapse of angiogenic vessels, or the suppression of the angiogenic process by inhibiting the migration and invasion abilities of endothelial cells. This rapid collapse of the endothelial tubular network in vitro occurs in a concentration-dependent manner and is observed at concentrations lower than that affecting cell survival. The in vitro findings were confirmed in tumor xenografts where plocabulin treatment induced a large reduction in vascular volume and induction of extensive necrosis in tumors, consistent with antivascular effects. CONCLUSIONS Altogether, these data suggest that an antivascular mechanism is contributing to the antitumor activities of plocabulin.
Collapse
Affiliation(s)
- Carlos M Galmarini
- R&D Area, PharmaMar S.A, Avda. de los Reyes 1, 28770 Colmenar Viejo, Madrid, Spain.
| | - Maud Martin
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584, CH, Utrecht, The Netherlands
| | - Benjamin Pierre Bouchet
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584, CH, Utrecht, The Netherlands
| | | | - Marta Martínez-Diez
- R&D Area, PharmaMar S.A, Avda. de los Reyes 1, 28770 Colmenar Viejo, Madrid, Spain
| | | | - Anna Akhmanova
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584, CH, Utrecht, The Netherlands
| | - Pablo Aviles
- R&D Area, PharmaMar S.A, Avda. de los Reyes 1, 28770 Colmenar Viejo, Madrid, Spain
| |
Collapse
|
34
|
Lei X, Chen M, Huang M, Li X, Shi C, Zhang D, Luo L, Zhang Y, Ma N, Chen H, Liang H, Ye W, Zhang D. Desacetylvinblastine Monohydrazide Disrupts Tumor Vessels by Promoting VE-cadherin Internalization. Am J Cancer Res 2018; 8:384-398. [PMID: 29290815 PMCID: PMC5743555 DOI: 10.7150/thno.22222] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/13/2017] [Indexed: 01/18/2023] Open
Abstract
Vinca alkaloids, the well-known tubulin-binding agents, are widely used for the clinical treatment of malignant tumors. However, little attention has been paid to their vascular disrupting effects, and the underlying mechanisms remain largely unknown. This study aims to investigate the vascular disrupting effect and the underlying mechanisms of vinca alkaloids. Methods: The capillary disruption assay and aortic ring assay were performed to evaluate the in vitro vascular disrupting effect of desacetylvinblastine monohydrazide (DAVLBH), a derivate of vinblastine, and the in vivo vascular disrupting effect was assessed on HepG2 xenograft model using magnetic resonance imaging, hematoxylin and eosin staining and immunohistochemistry. Tubulin polymerization, endothelial cell monolayer permeability, western blotting and immunofluorescence assays were performed to explore the underlying mechanisms of DAVLBH-mediated tumor vascular disruption. Results: DAVLBH has potent vascular disrupting activity both in vitro and in vivo. DAVLBH disrupts tumor vessels in a different manner than classical tubulin-targeting VDAs; it inhibits microtubule polymerization, promotes the internalization of vascular endothelial cadherin (VE-cadherin) and inhibits the recycling of internalized VE-cadherin to the cell membrane, thus increasing endothelial cell permeability and ultimately resulting in vascular disruption. DAVLBH-mediated promotion of VE-cadherin internalization and inhibition of internalized VE-cadherin recycling back to the cell membrane are partly dependent on inhibition of microtubule polymerization, and Src activation is involved in DAVLBH-induced VE-cadherin internalization. Conclusions: This study sheds light on the tumor vascular disrupting effect and underlying mechanisms of vinca alkaloids and provides new insight into the molecular mechanism of tubulin-targeting VDAs.
Collapse
|
35
|
Antivascular and antitumor properties of the tubulin-binding chalcone TUB091. Oncotarget 2017; 8:14325-14342. [PMID: 27224920 PMCID: PMC5362409 DOI: 10.18632/oncotarget.9527] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/01/2016] [Indexed: 12/28/2022] Open
Abstract
We investigated the microtubule-destabilizing, vascular-targeting, anti-tumor and anti-metastatic activities of a new series of chalcones, whose prototype compound is (E)-3-(3’’-amino-4’’-methoxyphenyl)-1-(5’-methoxy-3’,4’-methylendioxyphenyl)-2-methylprop-2-en-1-one (TUB091). X-ray crystallography showed that these chalcones bind to the colchicine site of tubulin and therefore prevent the curved-to-straight structural transition of tubulin, which is required for microtubule formation. Accordingly, TUB091 inhibited cancer and endothelial cell growth, induced G2/M phase arrest and apoptosis at 1-10 nM. In addition, TUB091 displayed vascular disrupting effects in vitro and in the chicken chorioallantoic membrane (CAM) assay at low nanomolar concentrations. A water-soluble L-Lys-L-Pro derivative of TUB091 (i.e. TUB099) showed potent antitumor activity in melanoma and breast cancer xenograft models by causing rapid intratumoral vascular shutdown and massive tumor necrosis. TUB099 also displayed anti-metastatic activity similar to that of combretastatin A4-phosphate. Our data indicate that this novel class of chalcones represents interesting lead molecules for the design of vascular disrupting agents (VDAs). Moreover, we provide evidence that our prodrug approach may be valuable for the development of anti-cancer drugs.
Collapse
|
36
|
Shepherd J, Fisher M, Welford A, McDonald DM, Kanthou C, Tozer GM. The protective role of sphingosine-1-phosphate against the action of the vascular disrupting agent combretastatin A-4 3- O-phosphate. Oncotarget 2017; 8:95648-95661. [PMID: 29221156 PMCID: PMC5707050 DOI: 10.18632/oncotarget.21172] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
Solid tumours vary in sensitivity to the vascular disrupting agent combretastatin A-4 3-O-phosphate (CA4P), but underlying factors are poorly understood. The signaling sphingolipid, sphingosine-1-phosphate (S1P), promotes vascular barrier integrity by promoting assembly of VE-cadherin/β-catenin complexes. We tested the hypothesis that tumour pre-treatment with S1P would render tumours less susceptible to CA4P. S1P (1μM) pretreatment attenuated an increase in endothelial cell (HUVEC) monolayer permeability induced by 10μM CA4P. Intravenously administered S1P (8mg/kg/hr for 20 minutes then 2mg/kg/hr for 40 minutes), reduced CA4P-induced (30mg/kg) blood flow shut-down in fibrosarcoma tumours in SCID mice (n≥7 per group), as measured by tumour retention of an intravenously administered fluorescent lectin. A trend towards in vivo protection was also found using laser Doppler flowmetry. Immunohistochemical staining of tumours ex vivo revealed disrupted patterns of VE-cadherin in vasculature of mice treated with CA4P, which were decreased by pretreatment with S1P. S1P treatment also stabilized N-cadherin junctions between endothelial cells and smooth muscle cells in culture, and stabilized tubulin filaments in HUVEC monolayers. We conclude that the rapid shutdown of tumour microvasculature by CA4P is due in part to disruption of adherens junctions and that S1P has a protective effect on both adherens junctions and the endothelial cell cytoskeleton.
Collapse
Affiliation(s)
- Joanna Shepherd
- Current/Present address: School of Clinical Dentistry, The University of Sheffield, Claremont Crescent, Sheffield, UK
| | - Matthew Fisher
- Tumour Microcirculation Group, The University of Sheffield, Department of Oncology and Metabolism, School of Medicine, Sheffield, UK
| | - Abigail Welford
- Tumour Microcirculation Group, The University of Sheffield, Department of Oncology and Metabolism, School of Medicine, Sheffield, UK
| | - Donald M McDonald
- UCSF Comprehensive Cancer Center, Cardiovascular Research Institute, and Department of Anatomy, University of California, San Francisco, CA, USA
| | - Chryso Kanthou
- Tumour Microcirculation Group, The University of Sheffield, Department of Oncology and Metabolism, School of Medicine, Sheffield, UK
| | - Gillian M Tozer
- Tumour Microcirculation Group, The University of Sheffield, Department of Oncology and Metabolism, School of Medicine, Sheffield, UK
| |
Collapse
|
37
|
Sherbet G. Suppression of angiogenesis and tumour progression by combretastatin and derivatives. Cancer Lett 2017; 403:289-295. [DOI: 10.1016/j.canlet.2017.06.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/10/2017] [Accepted: 06/28/2017] [Indexed: 12/17/2022]
|
38
|
Shen Y, Wu L, Qiu L. Water-Soluble Combretastatin A4 Phosphate Orally Delivered via Composite Nanoparticles With Improved Inhibition Effect Toward S180 Tumors. J Pharm Sci 2017; 106:3076-3083. [PMID: 28619603 DOI: 10.1016/j.xphs.2017.05.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/17/2017] [Accepted: 05/24/2017] [Indexed: 01/15/2023]
Abstract
Combretastatin A4 phosphate (CA4P) is a novel vascular disrupting agent for cancer therapy. However, frequent dosing and negative patient compliance have been encountered over CA4P by injection administration due to its quite short-term action and acute side effects. Therefore, it is significant to develop an oral formulation of CA4P. We established a novel method to prepare CA4P-loaded nanoparticles (CA4P-NPs) for oral administration by combining methoxy poly(ethylene glycol)-b-polylactide (PELA) and poly(d,l-lactic-co-glycolic acid) (PLGA) polymers. Transport study in vitro was evaluated on Madin-Darby canine kidney cell models, and antitumor effect evaluation in vivo was performed on S180 subcutaneous xenotransplanted tumor models in mice. The highest entrapment efficiency of CA4P-NPs was achieved when the weight ratio of PELA to PLGA was optimized to 1:1. The apparent permeability coefficient of CA4P-NPs was found to be 2.08-fold higher than that of free CA4P in transport study. CA4P-NPs reached an absolute bioavailability of 77.6% with the tumor inhibition ratio of 41.2% that was significantly superior to free CA4P. These results suggest a promising application of this composite nanoparticle for the oral delivery of water-soluble drugs.
Collapse
Affiliation(s)
- Yurun Shen
- Ministry of Educational (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Liping Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Liyan Qiu
- Ministry of Educational (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
39
|
Izumi Y, Aoshima K, Hoshino Y, Takagi S. Effects of combretastatin A-4 phosphate on canine normal and tumor tissue-derived endothelial cells. Res Vet Sci 2017; 112:222-228. [DOI: 10.1016/j.rvsc.2017.05.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/19/2017] [Accepted: 05/12/2017] [Indexed: 12/09/2022]
|
40
|
Hardin C, Shum E, Singh AP, Perez-Soler R, Cheng H. Emerging treatment using tubulin inhibitors in advanced non-small cell lung cancer. Expert Opin Pharmacother 2017; 18:701-716. [DOI: 10.1080/14656566.2017.1316374] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
41
|
Liu T, Zhang D, Song W, Tang Z, Zhu J, Ma Z, Wang X, Chen X, Tong T. A poly(l-glutamic acid)-combretastatin A4 conjugate for solid tumor therapy: Markedly improved therapeutic efficiency through its low tissue penetration in solid tumor. Acta Biomater 2017; 53:179-189. [PMID: 28167300 DOI: 10.1016/j.actbio.2017.02.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/30/2017] [Accepted: 02/01/2017] [Indexed: 01/13/2023]
Abstract
UNLABELLED Combretastatin A4 (CA4) is a leading agent in vascular disrupting strategies for tumor therapy. Although many small-molecule prodrugs of CA4 have been developed to improve its solubility, the overall therapeutic efficiency is moderate. A key reason for this is the reversible effect that CA4 has on tubulin as well as its rapid clearance from plasma and tissues. In this study, we proposed a poly(l-glutamic acid)-CA4 conjugate (PLG-CA4) nanomedicine to fulfill the requirements for fully liberating the potential of CA4 on tumor therapy. Enhanced accumulation and retention of CA4 in tumor tissue, especially, high distribution and gradual release around tumor blood vessels resulted in prolonged vascular disruption and markedly enhanced therapeutic efficiency. We examined and compared the therapeutic effect of PLG-CA4 and commercial combretastatin-A4 phosphate (CA4P) in a murine colon C26 tumor. PLG-CA4 showed significantly prolonged retention in plasma and tumor tissue. Most importantly, the PLG-CA4 was mainly distributed around the tumor vessels because of its low tissue penetration in solid tumor. Pathology tests showed that PLG-CA4 treatment resulted in persistent vascular disruption and tumor damage 72h after a single injection, this in contrast to CA4P treatment, which showed quick relapse at an equal dose. Tumor suppression tests showed that PLG-CA4 treatment resulted in a tumor suppression rate of 74%, which indicates a significant advantage when compared to tumor suppression rate of the CA4P group, which was 24%. This is the first time that an advantage of the polymeric CA4 nanomedicine with low tissue penetration for solid tumor therapy has been shown. Thus, the results presented in this study provide a new idea for enhancing the tumor therapeutic effect of vascular disrupting agents. STATEMENT OF SIGNIFICANCE Nanomedicine usually has low tissue penetration in solid tumors, which limits the efficacy of nanomedicine in most cases. But herein, we demonstrate a nanosized vascular disruptive agent (VDA) PLG-CA4 has supper advantages over small molecular combretastatin-A4 phosphate (CA4P) because the PLG-CA4 was mainly distributed around the tumor vessels due to its low tissue penetration in solid tumor.
Collapse
Affiliation(s)
- Tianzhou Liu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Dawei Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Jiaming Zhu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Zhiming Ma
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Xudong Wang
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Ti Tong
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China.
| |
Collapse
|
42
|
Tarade D, Ma D, Pignanelli C, Mansour F, Simard D, van den Berg S, Gauld J, McNulty J, Pandey S. Structurally simplified biphenyl combretastatin A4 derivatives retain in vitro anti-cancer activity dependent on mitotic arrest. PLoS One 2017; 12:e0171806. [PMID: 28253265 PMCID: PMC5333808 DOI: 10.1371/journal.pone.0171806] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 01/26/2017] [Indexed: 12/31/2022] Open
Abstract
The cis-stilbene, combretastatin A4 (CA4), is a potent microtubule targeting and vascular damaging agent. Despite promising results at the pre-clinical level and extensive clinical evaluation, CA4 has yet to be approved for therapeutic use. One impediment to the development of CA4 is an inherent conformational instability about the ethylene linker, which joins two aromatic rings. We have previously published preliminary data regarding structurally simplified biphenyl derivatives of CA4, lacking an ethylene linker, which retain anti-proliferative and pro-apoptotic activity, albeit at higher doses. Our current study provides a more comprehensive evaluation regarding the anti-proliferative and pro-apoptotic properties of biphenyl CA4 derivatives in both 2D and 3D cancerous and non-cancerous cell models. Computational analysis has revealed that cytotoxicity of CA4 and biphenyl analogues correlates with predicted tubulin affinity. Additional mechanistic evaluation of the biphenyl derivatives found that their anti-cancer activity is dependent on prolonged mitotic arrest, in a similar manner to CA4. Lastly, we have shown that cancer cells deficient in the extrinsic pathway of apoptosis experience delayed cell death following treatment with CA4 or analogues. Biphenyl derivatives of CA4 represent structurally simplified analogues of CA4, which retain a similar mechanism of action. The biphenyl analogues warrant in vivo examination to evaluate their potential as vascular damaging agents.
Collapse
Affiliation(s)
- Daniel Tarade
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Dennis Ma
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Christopher Pignanelli
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Fadi Mansour
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Daniel Simard
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Sean van den Berg
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - James Gauld
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - James McNulty
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Siyaram Pandey
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
- * E-mail:
| |
Collapse
|
43
|
Kong Y, Smith J, Li K, Cui J, Han J, Hou S, Brown ML. Development of a novel near-infrared fluorescent theranostic combretastain A-4 analogue, YK-5-252, to target triple negative breast cancer. Bioorg Med Chem 2017; 25:2226-2233. [PMID: 28284864 DOI: 10.1016/j.bmc.2017.02.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/15/2017] [Accepted: 02/21/2017] [Indexed: 01/05/2023]
Abstract
The treatment of triple negative breast cancer (TNBC) is a significant challenge to cancer research. The lack of hormone receptors limits the treatment options available to patients with this diagnosis, forcing them to endure prolonged radiation and chemotherapy. Anti-angiogenesis is a chemotherapeutic strategy that targets the vasculature of tumors. Combretastatin A-4 (CA-4) is a well-known vasculature-disrupting agent, which has been shown to effectively kill a variety of cancers through inhibition of tubulin polymerization. Due to its toxicity, small molecule analogues of CA-4 have been sought out. We have designed a novel dual action CA-4 prodrug, YK-5-252, which releases the drug through a disulfide bond cleavage mechanism and contains a near-infrared (NIR) fluorophore, which allows fluorescence monitoring of cleavage. This disulfide linkage causes CA-4 to become effective only when released by glutathione (GSH) reducing the toxicity of the drug while simultaneously releasing the NIR fluorophore. Therefore the prodrug, YK-5-252, represents a novel CA-4 analogue which has reduced toxicity and can be used for theranostics imaging.
Collapse
Affiliation(s)
- Yali Kong
- Drug Discovery & Development, Inova Schar Cancer Institute; Drug Discovery Center, Inova Center for Personalized Health, 3225 Gallows Rd, Fairfax, VA 22031, United States
| | - Jacqueline Smith
- Department of Natural Sciences, Bowie State University, 14000 Jericho Park Dr, Crawford 207A, Bowie, MD 20716, United States
| | - Kongwen Li
- Fairfax High School, 3501 Rebel Run, Fairfax, VA 22030, United States
| | - Jake Cui
- Thomas Jefferson High School for Science and Technology, 6560 Braddock Road, Alexandria 22312, United States
| | - John Han
- James Madison High School, 2500 James Madison Drive, Vienna, VA 22181, United States
| | - Shujie Hou
- Department of Oncology, Center for Drug Discovery, Georgetown University Medical Center, 3970 Reservoir Rd, Washington D.C. 20057, United States
| | - Milton L Brown
- Drug Discovery & Development, Inova Schar Cancer Institute; Drug Discovery Center, Inova Center for Personalized Health, 3225 Gallows Rd, Fairfax, VA 22031, United States.
| |
Collapse
|
44
|
Initiation of acute graft-versus-host disease by angiogenesis. Blood 2017; 129:2021-2032. [PMID: 28096092 DOI: 10.1182/blood-2016-08-736314] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/08/2017] [Indexed: 01/06/2023] Open
Abstract
The inhibition of inflammation-associated angiogenesis ameliorates inflammatory diseases by reducing the recruitment of tissue-infiltrating leukocytes. However, it is not known if angiogenesis has an active role during the initiation of inflammation or if it is merely a secondary effect occurring in response to stimuli by tissue-infiltrating leukocytes. Here, we show that angiogenesis precedes leukocyte infiltration in experimental models of inflammatory bowel disease and acute graft-versus-host disease (GVHD). We found that angiogenesis occurred as early as day+2 after allogeneic transplantation mainly in GVHD typical target organs skin, liver, and intestines, whereas no angiogenic changes appeared due to conditioning or syngeneic transplantation. The initiation phase of angiogenesis was not associated with classical endothelial cell (EC) activation signs, such as Vegfa/VEGFR1+2 upregulation or increased adhesion molecule expression. During early GVHD at day+2, we found significant metabolic and cytoskeleton changes in target organ ECs in gene array and proteomic analyses. These modifications have significant functional consequences as indicated by profoundly higher deformation in real-time deformability cytometry. Our results demonstrate that metabolic changes trigger alterations in cell mechanics, leading to enhanced migratory and proliferative potential of ECs during the initiation of inflammation. Our study adds evidence to the hypothesis that angiogenesis is involved in the initiation of tissue inflammation during GVHD.
Collapse
|
45
|
Brown AW, Fisher M, Tozer GM, Kanthou C, Harrity JPA. Sydnone Cycloaddition Route to Pyrazole-Based Analogs of Combretastatin A4. J Med Chem 2016; 59:9473-9488. [PMID: 27690431 DOI: 10.1021/acs.jmedchem.6b01128] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The combretastatins are an important class of tubulin-binding agents. Of this family, a number of compounds are potent tumor vascular disrupting agents (VDAs) and have shown promise in the clinic for cancer therapy. We have developed a modular synthetic route to combretastatin analogs based on a pyrazole core through highly regioselective alkyne cycloaddition reactions of sydnones. These compounds show modest to high potency against human umbilical vein endothelial cell proliferation. Moreover, evidence is presented that these novel VDAs have the same mode of action as CA4P and bind reversibly to β-tubulin, believed to be a key feature in avoiding toxicity. The most active compound from in vitro studies was taken forward to an in vivo model and instigated an increase in tumor cell necrosis.
Collapse
Affiliation(s)
- Andrew W Brown
- Department of Chemistry, University of Sheffield , Dainton Building, Brook Hill, Sheffield S3 7HF, U.K.,Department of Oncology and Metabolism, The Medical School, University of Sheffield , Beech Hill Road, Sheffield S10 2RX, U.K
| | - Matthew Fisher
- Department of Oncology and Metabolism, The Medical School, University of Sheffield , Beech Hill Road, Sheffield S10 2RX, U.K
| | - Gillian M Tozer
- Department of Oncology and Metabolism, The Medical School, University of Sheffield , Beech Hill Road, Sheffield S10 2RX, U.K
| | - Chryso Kanthou
- Department of Oncology and Metabolism, The Medical School, University of Sheffield , Beech Hill Road, Sheffield S10 2RX, U.K
| | - Joseph P A Harrity
- Department of Chemistry, University of Sheffield , Dainton Building, Brook Hill, Sheffield S3 7HF, U.K
| |
Collapse
|
46
|
Te Boekhorst V, Friedl P. Plasticity of Cancer Cell Invasion-Mechanisms and Implications for Therapy. Adv Cancer Res 2016; 132:209-64. [PMID: 27613134 DOI: 10.1016/bs.acr.2016.07.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer cell migration is a plastic and adaptive process integrating cytoskeletal dynamics, cell-extracellular matrix and cell-cell adhesion, as well as tissue remodeling. In response to molecular and physical microenvironmental cues during metastatic dissemination, cancer cells exploit a versatile repertoire of invasion and dissemination strategies, including collective and single-cell migration programs. This diversity generates molecular and physical heterogeneity of migration mechanisms and metastatic routes, and provides a basis for adaptation in response to microenvironmental and therapeutic challenge. We here summarize how cytoskeletal dynamics, protease systems, cell-matrix and cell-cell adhesion pathways control cancer cell invasion programs, and how reciprocal interaction of tumor cells with the microenvironment contributes to plasticity of invasion and dissemination strategies. We discuss the potential and future implications of predicted "antimigration" therapies that target cytoskeletal dynamics, adhesion, and protease systems to interfere with metastatic dissemination, and the options for integrating antimigration therapy into the spectrum of targeted molecular therapies.
Collapse
Affiliation(s)
- V Te Boekhorst
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - P Friedl
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Radboud University Medical Centre, Nijmegen, The Netherlands; Cancer Genomics Center (CGC.nl), Utrecht, The Netherlands.
| |
Collapse
|
47
|
Not flavone-8-acetic acid (FAA) but its murine metabolite 6-OH-FAA exhibits remarkable antivascular activities in vitro. Anticancer Drugs 2016; 27:398-406. [PMID: 26901071 DOI: 10.1097/cad.0000000000000341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Flavone-8-acetic acid (FAA) has been proved to be a potent vascular-disrupting agent in mice. Unfortunately, FAA did not produce any anticancer activity in clinical trials. Previously, we had reported that FAA is metabolized by mouse microsomes into six metabolites, whereas it was poorly metabolized by human microsomes, with fewer metabolites formed in lesser amounts. Especially, 6-OH-FAA was not formed by human microsomes. In this work, two major available metabolites, 4'-OH-FAA and 6-OH-FAA, were tested and compared with the parent compound FAA for their potential antivascular activities in vitro. The ability of the products to induce morphological changes, disrupt preformed capillaries of EA.hy926 endothelial cells and inhibit tubulin polymerization in vitro was assessed. The action mechanism was determined using the RhoA and Rac1 inhibitors. At 25 µg/ml, 6-OH-FAA induced morphological changes and membrane blebbing, whereas 300 µg/ml of FAA and 4'-OH-FAA slightly changed the morphology without inducing membrane blebbing. At 300 µg/ml, 6-OH-FAA produced morphological changes that were 2.1-6.9-fold greater than that produced by FAA and 4'-OH-FAA, an effect that was consistent with its much greater inhibitory effect on tubulin polymerization compared with FAA and 4'-OH-FAA. 6-OH-FAA significantly disrupted the EA.hy926 cell capillaries. 6-OH-FAA activities were prevented in EA.hy926 cells pretreated with RhoA, but not Rac1, inhibitor. In this short communication we report for the first time that, in vitro, 6-OH-FAA, a mouse-specific FAA metabolite, exhibits significantly stronger antivascular activities compared with FAA and 4'-OH-FAA, which are mediated through the RhoA kinase pathway.
Collapse
|
48
|
Porcù E, Salvador A, Primac I, Mitola S, Ronca R, Ravelli C, Bortolozzi R, Vedaldi D, Romagnoli R, Basso G, Viola G. Vascular disrupting activity of combretastatin analogues. Vascul Pharmacol 2016; 83:78-89. [DOI: 10.1016/j.vph.2016.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 05/11/2016] [Accepted: 05/21/2016] [Indexed: 01/11/2023]
|
49
|
Pérez-Pérez MJ, Priego EM, Bueno O, Martins MS, Canela MD, Liekens S. Blocking Blood Flow to Solid Tumors by Destabilizing Tubulin: An Approach to Targeting Tumor Growth. J Med Chem 2016; 59:8685-8711. [DOI: 10.1021/acs.jmedchem.6b00463] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
| | - Eva-María Priego
- Instituto de Química Médica (IQM-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Oskía Bueno
- Instituto de Química Médica (IQM-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | | | - María-Dolores Canela
- Instituto de Química Médica (IQM-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Sandra Liekens
- Rega
Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium
| |
Collapse
|
50
|
Driowya M, Leclercq J, Verones V, Barczyk A, Lecoeur M, Renault N, Flouquet N, Ghinet A, Berthelot P, Lebegue N. Synthesis of triazoloquinazolinone based compounds as tubulin polymerization inhibitors and vascular disrupting agents. Eur J Med Chem 2016; 115:393-405. [DOI: 10.1016/j.ejmech.2016.03.056] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 02/17/2016] [Accepted: 03/18/2016] [Indexed: 02/08/2023]
|