51
|
Deng X, Li Z, Xiong R, Liu J, Liu R, Peng J, Chen Y, Lei X, Cao X, Zheng X, Xie Z, Tang G. FS-7 inhibits MGC-803 cells growth in vitro and in vivo via down-regulating glycolysis. Biomed Pharmacother 2019; 109:1659-1669. [DOI: 10.1016/j.biopha.2018.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 12/20/2022] Open
|
52
|
Choudhury H, Pandey M, Chin PX, Phang YL, Cheah JY, Ooi SC, Mak KK, Pichika MR, Kesharwani P, Hussain Z, Gorain B. Transferrin receptors-targeting nanocarriers for efficient targeted delivery and transcytosis of drugs into the brain tumors: a review of recent advancements and emerging trends. Drug Deliv Transl Res 2018; 8:1545-1563. [PMID: 29916012 DOI: 10.1007/s13346-018-0552-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Treatment of glioblastoma multiforme (GBM) is a predominant challenge in chemotherapy due to the existence of blood-brain barrier (BBB) which restricts delivery of chemotherapeutic agents to the brain together with the problem of drug penetration through hard parenchyma of the GBM. With the structural and mechanistic elucidation of the BBB under both physiological and pathological conditions, it is now viable to target central nervous system (CNS) disorders utilizing the presence of transferrin (Tf) receptors (TfRs). However, overexpression of these TfRs on the GBM cell surface can also help to avoid restrictions of GBM cells to deliver chemotherapeutic agents within the tumor. Therefore, targeting of TfR-mediated delivery could counteract drug delivery issues in GBM and create a delivery system that could cross the BBB effectively to utilize ligand-conjugated drug complexes through receptor-mediated transcytosis. Hence, approach towards successful delivery of antitumor agents to the gliomas has been making possible through targeting these overexpressed TfRs within the CNS and glioma cells. This review article presents a thorough analysis of current understanding on Tf-conjugated nanocarriers as efficient drug delivery system.
Collapse
Affiliation(s)
- Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia.
| | - Manisha Pandey
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Pei Xin Chin
- School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Yee Lin Phang
- School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Jeng Yuen Cheah
- School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Shu Chien Ooi
- School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Kit-Kay Mak
- School of Postgraduate Studies and Research, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Mallikarjuna Rao Pichika
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia.,Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Prashant Kesharwani
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Zahid Hussain
- Department of Pharmaceutics, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, 42300, Puncak Alam, Malaysia
| | - Bapi Gorain
- Faculty of Pharmacy, Lincoln University College, Petalling Jaya, 47301, Kuala Lumpur, Selangor, Malaysia
| |
Collapse
|
53
|
Vascular amounts and dispersion of caliber-classified vessels as key parameters to quantitate 3D micro-angioarchitectures in multiple myeloma experimental tumors. Sci Rep 2018; 8:17520. [PMID: 30504794 PMCID: PMC6269464 DOI: 10.1038/s41598-018-35788-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 11/05/2018] [Indexed: 12/26/2022] Open
Abstract
Blood vessel micro-angioarchitecture plays a pivotal role in tumor progression, metastatic dissemination and response to therapy. Thus, methods able to quantify microvascular trees and their anomalies may allow a better comprehension of the neovascularization process and evaluation of vascular-targeted therapies in cancer. To this aim, the development of a restricted set of indexes able to describe the arrangement of a microvascular tree is eagerly required. We addressed this goal through 3D analysis of the functional microvascular network in sulfo-biotin-stained human multiple myeloma KMS-11 xenografts in NOD/SCID mice. Using image analysis, we show that amounts, spatial dispersion and spatial relationships of adjacent classes of caliber-filtered microvessels provide a near-linear graphical “fingerprint” of tumor micro-angioarchitecture. Position, slope and axial projections of this graphical outcome reflect biological features and summarize the properties of tumor micro-angioarchitecture. Notably, treatment of KMS-11 xenografts with anti-angiogenic drugs affected position and slope of the specific curves without degrading their near-linear properties. The possibility offered by this procedure to describe and quantify the 3D features of the tumor micro-angioarchitecture paves the way to the analysis of the microvascular tree in human tumor specimens at different stages of tumor progression and after pharmacologic interventions, with possible diagnostic and prognostic implications.
Collapse
|
54
|
Schmitt F, Gold M, Rothemund M, Andronache I, Biersack B, Schobert R, Mueller T. New naphthopyran analogues of LY290181 as potential tumor vascular-disrupting agents. Eur J Med Chem 2018; 163:160-168. [PMID: 30503940 DOI: 10.1016/j.ejmech.2018.11.055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 10/04/2018] [Accepted: 11/22/2018] [Indexed: 10/27/2022]
Abstract
A series of 19 analogues of the antiproliferative naphthopyran LY290181 were prepared for structure-activity relationship studies. We found the best activities for test compounds bearing small substituents at the meta position of the phenyl ring. The mode of action of LY290181 and eight new analogues was studied in detail. The compounds were highly anti-proliferative with IC50 values in the sub-nanomolar to triple-digit nanomolar range. The new analogues led to G2/M arrest due to interruption of the microtubule dynamics. In 518A2 melanoma cells they caused a mitotic catastrophe which eventually led to apoptosis. The naphthopyrans also induced a disruption of the vasculature in the chorioallantoic membrane (CAM) of fertilized chicken eggs as well as in xenograft tumors in mice. In a preliminary therapy trial, the difluoro derivative 2b retarded the growth of resistant xenograft tumors in mice.
Collapse
Affiliation(s)
- Florian Schmitt
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Madeleine Gold
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Matthias Rothemund
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Ion Andronache
- University of Bucharest, Research Center for Integrated Analysis and Territorial Management, 4-12, Regina Elisabeta Avenue, Bucharest, 3rd District, 030018, Romania
| | - Bernhard Biersack
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Rainer Schobert
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany.
| | - Thomas Mueller
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle, Germany
| |
Collapse
|
55
|
Bian L, Gao M, Zhang D, Ji A, Su C, Duan X, Luo Q, Huang D, Feng Y, Ni Y, Yin Z, Jin Q, Zhang J. Synthesis and Biological Evaluation of Rhein-Based MRI Contrast Agents for in Vivo Visualization of Necrosis. Anal Chem 2018; 90:13249-13256. [DOI: 10.1021/acs.analchem.8b01868] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Li Bian
- Afliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
| | - Meng Gao
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
| | - Dongjian Zhang
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
| | - Aiyan Ji
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, P. R. China
| | - Chang Su
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, P. R. China
| | - Xinghua Duan
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, P. R. China
| | - Qi Luo
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, P. R. China
| | - Dejian Huang
- Afliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
| | - Yuanbo Feng
- Afliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
| | - Yicheng Ni
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
- Theragnostic Laboratory, Campus Gasthuisberg, KU Leuven, 3000 Leuven, Belgium
| | - Zhiqi Yin
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, P. R. China
| | - Qiaomei Jin
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
| | - Jian Zhang
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P. R. China
| |
Collapse
|
56
|
Höink A, Persigehl T, Kwiecien R, Balthasar M, Mesters R, Berdel W, Heindel W, Bremer C, Schwöppe C. Gadofosveset-enhanced MRI as simple surrogate parameter for real-time evaluation of the initial tumour vessel infarction by retargeted tissue factor tTF-NGR. Oncol Lett 2018; 17:270-280. [PMID: 30655764 PMCID: PMC6313167 DOI: 10.3892/ol.2018.9638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/22/2018] [Indexed: 01/16/2023] Open
Abstract
Truncated tissue factor (tTF)-NGR consists of the extracellular domain of the human TF and the binding motif NGR. tTF-NGR activates blood coagulation within the tumour vasculature following binding to CD13, and is overexpressed in the endothelial cells of tumour vessels, resulting in tumour vessel infarction and subsequent retardation/regression of tumour growth. The aim of the present study was to investigate gadofosveset-based real-time dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) in evaluating the initial therapeutic effects of the anti-vascular tTF-NGR approach. DCE-MRI (3.0 T) was performed in human U87-glioblastoma tumour-bearing nude mice. During a dynamic T1w GE-sequence, a gadolinium-based blood pool contrast agent (gadofosveset) was injected via a tail vein catheter. Following the maximum contrast intensity inside the tumour being obtained, tTF-NGR was injected (controls received NaCl) and the contrast behaviour of the tumour was monitored by ROI analysis. The slope difference of signal intensities between controls and the tTF-NGR group was investigated, as well as the differences between the average area under the curve (AUC) of the two groups. The association between intensity, group (control vs. tTF-NGR group) and time was analysed by fitting a linear mixed model. Following the injection of tTF-NGR, the signal intensity inside the tumours exhibited a statistically significantly stronger average slope decrease compared with the signal intensity of the tumours in the NaCl group. Furthermore, the initial average AUC values of mice treated with tTF-NGR were 5.7% lower than the average AUC of the control animals (P<0.05). Gadofosveset-enhanced MRI enables the visualization of the initial tumour response to anti-vascular treatment in real-time. Considering the clinical application of tTF-NGR, this method may provide a simple alternative parameter for monitoring the tumour response to vascular disrupting agents and certain vascular targeting agents in humans.
Collapse
Affiliation(s)
- Anna Höink
- Department of Clinical Radiology, University Hospital Münster, D-48149 Münster, Germany.,Department of Diagnostic and Interventional Radiology, University Hospital Cologne, D-50937 Cologne, Germany
| | - Thorsten Persigehl
- Department of Clinical Radiology, University Hospital Münster, D-48149 Münster, Germany.,Department of Diagnostic and Interventional Radiology, University Hospital Cologne, D-50937 Cologne, Germany
| | - Robert Kwiecien
- Institute of Biostatistics and Clinical Research, University of Münster, Germany
| | - Martin Balthasar
- Department of Diagnostic and Interventional Radiology, University Hospital Cologne, D-50937 Cologne, Germany
| | - Rolf Mesters
- Department of Medicine A - Haematology and Oncology, University Hospital Münster, D-48149 Münster, Germany
| | - Wolfgang Berdel
- Department of Medicine A - Haematology and Oncology, University Hospital Münster, D-48149 Münster, Germany
| | - Walter Heindel
- Department of Clinical Radiology, University Hospital Münster, D-48149 Münster, Germany
| | - Christoph Bremer
- Department of Clinical Radiology, University Hospital Münster, D-48149 Münster, Germany.,Department of Radiology, St. Franziskus-Hospital Münster, D-48145 Münster, Germany
| | - Christian Schwöppe
- Department of Radiology, St. Franziskus-Hospital Münster, D-48145 Münster, Germany
| |
Collapse
|
57
|
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: 4.0] [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
|
58
|
SPECT Imaging of Treatment-Related Tumor Necrosis Using Technetium-99m-Labeled Rhein. Mol Imaging Biol 2018; 21:660-668. [DOI: 10.1007/s11307-018-1285-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
59
|
Tomaszewski MR, Gehrung M, Joseph J, Quiros-Gonzalez I, Disselhorst JA, Bohndiek SE. Oxygen-Enhanced and Dynamic Contrast-Enhanced Optoacoustic Tomography Provide Surrogate Biomarkers of Tumor Vascular Function, Hypoxia, and Necrosis. Cancer Res 2018; 78:5980-5991. [PMID: 30115696 DOI: 10.1158/0008-5472.can-18-1033] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/22/2018] [Accepted: 08/13/2018] [Indexed: 11/16/2022]
Abstract
Measuring the functional status of tumor vasculature, including blood flow fluctuations and changes in oxygenation, is important in cancer staging and therapy monitoring. Current clinically approved imaging modalities suffer long procedure times and limited spatiotemporal resolution. Optoacoustic tomography (OT) is an emerging clinical imaging modality that may overcome these challenges. By acquiring data at multiple wavelengths, OT can interrogate hemoglobin concentration and oxygenation directly and resolve contributions from injected contrast agents. In this study, we tested whether two dynamic OT techniques, oxygen-enhanced (OE) and dynamic contrast-enhanced (DCE)-OT, could provide surrogate biomarkers of tumor vascular function, hypoxia, and necrosis. We found that vascular maturity led to changes in vascular function that affected tumor perfusion, modulating the DCE-OT signal. Perfusion in turn regulated oxygen availability, driving the OE-OT signal. In particular, we demonstrate for the first time a strong per-tumor and spatial correlation between imaging biomarkers derived from these in vivo techniques and tumor hypoxia quantified ex vivo Our findings indicate that OT may offer a significant advantage for localized imaging of tumor response to vascular-targeted therapies when compared with existing clinical DCE methods.Significance: Imaging biomarkers derived from optoacoustic tomography can be used as surrogate measures of tumor perfusion and hypoxia, potentially yielding rapid, multiparametric, and noninvasive cancer staging and therapeutic response monitoring in the clinic.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/20/5980/F1.large.jpg Cancer Res; 78(20); 5980-91. ©2018 AACR.
Collapse
Affiliation(s)
- Michal R Tomaszewski
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Marcel Gehrung
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Werner Siemens Imaging Center, Preclinical Imaging and Radiopharmacy, University of Tuebingen, Tuebingen, Germany
| | - James Joseph
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Isabel Quiros-Gonzalez
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan A Disselhorst
- Werner Siemens Imaging Center, Preclinical Imaging and Radiopharmacy, University of Tuebingen, Tuebingen, Germany
| | - Sarah E Bohndiek
- Department of Physics, University of Cambridge, Cambridge, United Kingdom.
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
60
|
Parhiz H, Shuvaev VV, Pardi N, Khoshnejad M, Kiseleva RY, Brenner JS, Uhler T, Tuyishime S, Mui BL, Tam YK, Madden TD, Hope MJ, Weissman D, Muzykantov VR. PECAM-1 directed re-targeting of exogenous mRNA providing two orders of magnitude enhancement of vascular delivery and expression in lungs independent of apolipoprotein E-mediated uptake. J Control Release 2018; 291:106-115. [PMID: 30336167 PMCID: PMC6477695 DOI: 10.1016/j.jconrel.2018.10.015] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 10/08/2018] [Accepted: 10/12/2018] [Indexed: 12/13/2022]
Abstract
Systemic administration of lipid nanoparticle (LNP)-encapsulated messenger RNA (mRNA) leads predominantly to hepatic uptake and expression. Here, we conjugated nucleoside-modified mRNA-LNPs with antibodies (Abs) specific to vascular cell adhesion molecule, PECAM-1. Systemic (intravenous) administration of Ab/LNP-mRNAs resulted in profound inhibition of hepatic uptake concomitantly with ~200-fold and 25-fold elevation of mRNA delivery and protein expression in the lungs compared to non-targeted counterparts. Unlike hepatic delivery of LNP-mRNA, Ab/LNP-mRNA is independent of apolipoprotein E. Vascular re-targeting of mRNA represents a promising, powerful, and unique approach for novel experimental and clinical interventions in organs of interest other than liver.
Collapse
Affiliation(s)
- Hamideh Parhiz
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Vladimir V Shuvaev
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Makan Khoshnejad
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Raisa Yu Kiseleva
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob S Brenner
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas Uhler
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Tuyishime
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | | | | | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Vladimir R Muzykantov
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
61
|
Maguire CJ, Chen Z, Mocharla VP, Sriram M, Strecker TE, Hamel E, Zhou H, Lopez R, Wang Y, Mason RP, Chaplin DJ, Trawick ML, Pinney KG. Synthesis of dihydronaphthalene analogues inspired by combretastatin A-4 and their biological evaluation as anticancer agents. MEDCHEMCOMM 2018; 9:1649-1662. [PMID: 30429970 PMCID: PMC6201230 DOI: 10.1039/c8md00322j] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022]
Abstract
The natural products colchicine and combretastatin A-4 (CA4) have provided inspiration for the discovery and development of a wide array of derivatives and analogues that inhibit tubulin polymerization through a binding interaction at the colchicine site on β-tubulin. A water-soluble phosphate prodrug salt of CA4 (referred to as CA4P) has demonstrated the ability to selectively damage tumor-associated vasculature and ushered in a new class of developmental anticancer agents known as vascular disrupting agents (VDAs). Through a long-term program of structure activity relationship (SAR) driven inquiry, we discovered that the dihydronaphthalene molecular scaffold provided access to small-molecule inhibitors of tubulin polymerization. In particular, a dihydronaphthalene analogue bearing a pendant trimethoxy aryl ring (referred to as KGP03) and a similar aroyl ring (referred to as KGP413) were potent inhibitors of tubulin polymerization (IC50 = 1.0 and 1.2 μM, respectively) and displayed low nM cytotoxicity against human cancer cell lines. In order to enhance water-solubility for in vivo evaluation, the corresponding phosphate prodrug salts (KGP04 and KGP152, respectively) were synthesized. In a preliminary in vivo study in a SCID-BALB/c mouse model bearing the human breast tumor MDA-MB-231-luc, a 99% reduction in signal was observed with bioluminescence imaging (BLI) 4 h after IP administration of KGP152 (200 mg kg-1) indicating reduced tumor blood flow. In a separate study, disruption of tumor-associated blood flow in a Fischer rat bearing an A549-luc human lung tumor was observed by color Doppler ultrasound following administration of KGP04 (15 mg kg-1).
Collapse
Affiliation(s)
- Casey J Maguire
- Department of Chemistry and Biochemistry , Baylor University , One Bear Place #97348 , Waco , TX 76798-7348 , USA . ; Tel: +(254) 710 4117
| | - Zhi Chen
- Department of Chemistry and Biochemistry , Baylor University , One Bear Place #97348 , Waco , TX 76798-7348 , USA . ; Tel: +(254) 710 4117
| | - Vani P Mocharla
- Department of Chemistry and Biochemistry , Baylor University , One Bear Place #97348 , Waco , TX 76798-7348 , USA . ; Tel: +(254) 710 4117
| | - Madhavi Sriram
- Department of Chemistry and Biochemistry , Baylor University , One Bear Place #97348 , Waco , TX 76798-7348 , USA . ; Tel: +(254) 710 4117
| | - Tracy E Strecker
- Department of Chemistry and Biochemistry , Baylor University , One Bear Place #97348 , Waco , TX 76798-7348 , USA . ; Tel: +(254) 710 4117
| | - Ernest Hamel
- Screening Technologies Branch , Developmental Therapeutics Program , Division of Cancer Treatment and Diagnosis , National Cancer Institute , Frederick National Laboratory for Cancer Research , National Institutes of Health , Frederick , MD 21702 , USA
| | - Heling Zhou
- Department of Radiology , The University of Texas Southwestern Medical Center , 5323 Harry Hines Boulevard , Dallas , TX 75390-9058 , USA
| | - Ramona Lopez
- Department of Radiology , The University of Texas Southwestern Medical Center , 5323 Harry Hines Boulevard , Dallas , TX 75390-9058 , USA
| | - Yifan Wang
- Department of Chemistry and Biochemistry , Baylor University , One Bear Place #97348 , Waco , TX 76798-7348 , USA . ; Tel: +(254) 710 4117
| | - Ralph P Mason
- Department of Radiology , The University of Texas Southwestern Medical Center , 5323 Harry Hines Boulevard , Dallas , TX 75390-9058 , USA
| | - David J Chaplin
- Department of Chemistry and Biochemistry , Baylor University , One Bear Place #97348 , Waco , TX 76798-7348 , USA . ; Tel: +(254) 710 4117
- Mateon Therapeutics, Inc. , 701 Gateway Boulevard, Suite 210 , South San Francisco , CA 94080 , USA
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry , Baylor University , One Bear Place #97348 , Waco , TX 76798-7348 , USA . ; Tel: +(254) 710 4117
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry , Baylor University , One Bear Place #97348 , Waco , TX 76798-7348 , USA . ; Tel: +(254) 710 4117
| |
Collapse
|
62
|
Molecular targeting of breast and colon cancer cells by PAR1 mediated apoptosis through a novel pro-apoptotic peptide. Apoptosis 2018; 23:679-694. [DOI: 10.1007/s10495-018-1485-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
63
|
Design, synthesis and characterization of potent microtubule inhibitors with dual anti-proliferative and anti-angiogenic activities. Eur J Med Chem 2018; 157:380-396. [DOI: 10.1016/j.ejmech.2018.07.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/30/2018] [Accepted: 07/16/2018] [Indexed: 11/30/2022]
|
64
|
Ghinea N. Vascular Endothelial FSH Receptor, a Target of Interest for Cancer Therapy. Endocrinology 2018; 159:3268-3274. [PMID: 30113652 DOI: 10.1210/en.2018-00466] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/30/2018] [Indexed: 11/19/2022]
Abstract
Improved molecular understanding of tumor microenvironment has resulted in the identification of various cancer cell targets for diagnostic and therapeutic interventions, including the receptor for the FSH, a glycoprotein hormone responsible for growth, maturation, and function of human reproductive system. The expression and localization of the FSH receptor (FSHR)-protein were associated with the tumor epithelial cells and/or with the peripheral tumor blood vessels. The available evidence indicates that in ovarian cancer, prostate cancer, and breast cancer, the tumor epithelial FSHR promotes proliferation, migration, and invasion of cancer cells. The vascular endothelial FSHR, detected in 11 types of solid tumors and 11 types of sarcomas, is involved in receptor-mediated transendothelial transport of FSH, tumor angiogenesis, and vascular remodeling. In contrast to intratumor vessels, which are abnormal and disorganized, the FSHR-positive blood microvessels are arranged in a hierarchical pattern: arterioles-capillaries-venules. The FSHR-positive blood vessels make connections between the intratumor vessels and the general blood circulation of patients. In this mini-review, I summarize these studies and discuss the rationale for developing a strategy for cancer therapy based on FSHR expressed on the luminal endothelial cell surface of blood vessels located in the peritumoral area rather than endothelial markers expressed in the core of tumors. Because FSHR is a common marker of peritumoral vessels, therapeutic agents coupled to anti-FSHR humanized antibodies should in principle be applicable to a wide range of tumor types.
Collapse
Affiliation(s)
- Nicolae Ghinea
- Inserm-Tumor Angiogenesis Team, Translational Research Department, Curie Institute, Paris, France
| |
Collapse
|
65
|
Heggannavar G, Hiremath CG, Achari DD, Pangarkar VG, Kariduraganavar MY. Development of Doxorubicin-Loaded Magnetic Silica-Pluronic F-127 Nanocarriers Conjugated with Transferrin for Treating Glioblastoma across the Blood-Brain Barrier Using an in Vitro Model. ACS OMEGA 2018; 3:8017-8026. [PMID: 30087932 PMCID: PMC6072239 DOI: 10.1021/acsomega.8b00152] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/05/2018] [Indexed: 05/14/2023]
Abstract
Brain glioma is the most lethal type of cancer, with extremely poor prognosis and high relapse. Unfortunately, the treatment of brain glioma is often limited because of the low permeability of anticancer drugs across the blood-brain barrier (BBB). To circumvent this, magnetic mesoporous nanoparticles were synthesized and loaded with doxorubicin as an anticancer agent. These nanoparticles were fabricated with Pluronic F-127 and subsequently conjugated with transferrin (Tf) to achieve the sustained release of the drug at the targeted site. The physicochemical properties of the conjugated nanoparticles were analyzed using different techniques. The magnetic saturation of the nanoparticles determined by a vibration sample magnetometer was found to be 26.10 emu/g. The cytotoxicity study was performed using the MTT assay at 48 and 96 h against the U87 cell line. The Tf-conjugated nanoparticles (DOX-MNP-MSN-PF-127-Tf) exhibited a significant IC50 value (0.570 μg/mL) as compared to the blank nanoparticles (121.98 μg/mL). To understand the transport mechanism of drugs across the BBB, an in vitro BBB model using human brain microvascular endothelial cells was developed. Among the nanoparticles, the Tf-conjugated nanoparticles demonstrated an excellent permeability across the BBB. This effect was predominant in the presence of an external magnetic field, suggesting that magnetic particles present in the matrix facilitated the uptake of drugs in U87 cells. Finally, it is concluded that nanoparticles conjugated with Tf effectively crossed the BBB. Thus, the developed nanocarriers can be considered as potential candidates to treat brain tumor.
Collapse
Affiliation(s)
| | - Chinmay G. Hiremath
- Department
of Studies in Chemistry, Karnatak University, Dharwad 580 003, India
| | - Divya D. Achari
- Department
of Studies in Chemistry, Karnatak University, Dharwad 580 003, India
| | | | | |
Collapse
|
66
|
Abma E, De Spiegelaere W, Vanderperren K, Stock E, Van Brantegem L, Cornelis I, Daminet S, Ni Y, Vynck M, Verstraete G, Smets P, de Rooster H. A single dose of intravenous combretastatin A4-phosphate is reasonably well tolerated and significantly reduces tumour vascularization in canine spontaneous cancers. Vet Comp Oncol 2018; 16:467-477. [PMID: 29797763 DOI: 10.1111/vco.12402] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/10/2018] [Accepted: 04/11/2018] [Indexed: 12/01/2022]
Abstract
Combretastatin A4-phosphate (CA4P) is an anti-tumour vascular targeting agent which selectively blocks tumour blood flow. Research on CA4P in rodent tumour models is extensive; however, knowledge of its effect on spontaneous cancer is scarce. This study was conducted in canine patients with spontaneous solid tumours. The goal was to assess the toxicity and efficacy of CA4P in various spontaneous tumour types. Eight dogs with spontaneous tumours were enrolled and treated with a single dose of 75 mg m-2 intravenous CA4P. The dogs were screened and monitored before and after injection. Pre- and post-treatment tumour blood flow was analysed in vivo by power Doppler ultrasound (PDUS) and contrast-enhanced ultrasound (CEUS). Vessel destruction and tumour necrosis were evaluated by histopathology. Clinically relevant toxicity was limited to one case of temporary tetraparesis; other adverse events were mild. Significant cardiovascular changes were mostly confined to changes in heart rate and cTnI levels. Macroscopic tumour size reduction was evident in 2 dogs. Based on PDUS and CEUS, CA4P induced a significant decrease in vascular index and tumour blood flow. Post-treatment, histopathology revealed a significant increase of necrotic tumoural tissue and a significant reduction in microvessel density in tumoural tissue. Anti-vascular and necrotizing effects of CA4P were documented in a variety of canine spontaneous cancers with only minimal side effects. This is the first study reporting the administration of CA4P to canine cancer patients with in vivo and ex vivo assessment, and a first step toward implementing CA4P in combination therapies in veterinary oncology patients. The use of CA4P in canine patients was approved and registered by the Belgian Federal Agency for Medicines and Health Products (FAMHP) (approval number 0002588, registration number 6518 ID 2F12).
Collapse
Affiliation(s)
- E Abma
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital Ghent, Ghent, Belgium
| | - W De Spiegelaere
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - K Vanderperren
- Department of Medical Imaging and Orthopedics of Small Animals, Faculty of Veterinary Medicine, University of Ghent, Ghent, Belgium
| | - E Stock
- Department of Medical Imaging and Orthopedics of Small Animals, Faculty of Veterinary Medicine, University of Ghent, Ghent, Belgium
| | - L Van Brantegem
- Department of Pathology, Bacteriology and Poultry Diseases, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - I Cornelis
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - S Daminet
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Y Ni
- Theragnostic Lab, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - M Vynck
- Department of Data Analysis and Mathematical Modeling, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - G Verstraete
- Laboratory of Pharmaceutical Technology, Ghent University, Ghent, Belgium
| | - P Smets
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - H de Rooster
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital Ghent, Ghent, Belgium
| |
Collapse
|
67
|
Subramanian S, Ugoya SO, Zhao Z, McRobb LS, Grau GE, Combes V, Inglis DW, Gauden AJ, Lee VS, Moutrie V, Santos ED, Stoodley MA. Stable thrombus formation on irradiated microvascular endothelial cells under pulsatile flow: Pre-testing annexin V-thrombin conjugate for treatment of brain arteriovenous malformations. Thromb Res 2018; 167:104-112. [PMID: 29803980 DOI: 10.1016/j.thromres.2018.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/19/2018] [Accepted: 05/14/2018] [Indexed: 01/30/2023]
Abstract
BACKGROUND Our goal is to develop a vascular targeting treatment for brain arteriovenous malformations (AVMs). Externalized phosphatidylserine has been established as a potential biomarker on the endothelium of irradiated AVM blood vessels. We hypothesize that phosphatidylserine could be selectively targeted after AVM radiosurgery with a ligand-directed vascular targeting agent to achieve localized thrombosis and rapid occlusion of pathological AVM vessels. OBJECTIVE The study aim was to establish an in vitro parallel-plate flow chamber to test the efficacy of a pro-thrombotic conjugate targeting phosphatidylserine. METHODS Conjugate was prepared by Lys-Lys cross-linking of thrombin with the phosphatidylserine-targeting ligand, annexin V. Cerebral microvascular endothelial cells were irradiated (5, 15, and 25 Gy) and after 1 or 3 days assembled in a parallel-plate flow chamber containing whole human blood and conjugate (1.25 or 2.5 μg/mL). Confocal microscopy was used to assess thrombus formation after flow via binding and aggregation of fluorescently-labelled platelets and fibrinogen. RESULTS AND CONCLUSIONS The annexin V-thrombin conjugate induced rapid thrombosis (fibrin deposition) on irradiated endothelial cells under shear stress in the parallel-plate flow device. Unconjugated, non-targeting thrombin did not induce fibrin deposition. A synergistic interaction between radiation and conjugate dose was observed. Thrombosis was greatest at the highest combined doses of radiation (25 Gy) and conjugate (2.5 μg/mL). The parallel-plate flow system provides a rapid method to pre-test pro-thrombotic vascular targeting agents. These findings validate the translation of the annexin V-thrombin conjugate to pre-clinical studies.
Collapse
Affiliation(s)
- S Subramanian
- Department of Clinical Medicine, Macquarie University, Sydney 2109, Australia
| | - S O Ugoya
- Department of Clinical Medicine, Macquarie University, Sydney 2109, Australia
| | - Z Zhao
- Department of Clinical Medicine, Macquarie University, Sydney 2109, Australia
| | - L S McRobb
- Department of Clinical Medicine, Macquarie University, Sydney 2109, Australia
| | - G E Grau
- Department of Pathology, University of Sydney, Sydney 2050, Australia
| | - V Combes
- University of Technology, School of Life Sciences, Sydney 2007, Australia
| | - D W Inglis
- School of Engineering, Macquarie University, Sydney 2109, Australia
| | - A J Gauden
- Department of Clinical Medicine, Macquarie University, Sydney 2109, Australia
| | - V S Lee
- Department of Clinical Medicine, Macquarie University, Sydney 2109, Australia
| | - V Moutrie
- Genesis Cancer Care, Macquarie University Hospital, Sydney 2109, Australia
| | - E D Santos
- Genesis Cancer Care, Macquarie University Hospital, Sydney 2109, Australia
| | - M A Stoodley
- Department of Clinical Medicine, Macquarie University, Sydney 2109, Australia.
| |
Collapse
|
68
|
NGR (Asn-Gly-Arg)-targeted delivery of coagulase to tumor vasculature arrests cancer cell growth. Oncogene 2018; 37:3967-3980. [PMID: 29662195 PMCID: PMC6053358 DOI: 10.1038/s41388-018-0213-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 01/25/2018] [Accepted: 02/02/2018] [Indexed: 11/17/2022]
Abstract
Induction of selective thrombosis and infarction in tumor-feeding vessels represents an attractive strategy to combat cancer. Here we took advantage of the unique coagulation properties of staphylocoagulase and genetically engineered it to generate a new fusion protein with novel anti-cancer properties. This novel bi-functional protein consists of truncated coagulase (tCoa) and an NGR (GNGRAHA) motif that recognizes CD13 and αvβ3 integrin receptors, targeting it to tumor endothelial cells. Herein, we report that tCoa coupled by its C-terminus to an NGR sequence retained its normal binding activity with prothrombin and avβ3 integrins, as confirmed in silico and in vitro. Moreover, in vivo biodistribution studies demonstrated selective accumulation of FITC-labeled tCoa-NGR fusion proteins at the site of subcutaneously implanted PC3 tumor xenografts in nude mice. Notably, systemic administration of tCoa-NGR to mice bearing 4T1 mouse mammary xenografts or PC3 human prostate tumors resulted in a significant reduction in tumor growth. These anti-tumor effects were accompanied by massive thrombotic occlusion of small and large tumor vessels, tumor infarction and tumor cell death. From these findings, we propose tCoa-NGR mediated tumor infarction as a novel and promising anti-cancer strategy targeting both CD13 and integrin αvβ3 positive tumor neovasculature.
Collapse
|
69
|
Wang L, Habib AA, Mintz A, Li KC, Zhao D. Phosphatidylserine-Targeted Nanotheranostics for Brain Tumor Imaging and Therapeutic Potential. Mol Imaging 2018; 16:1536012117708722. [PMID: 28654387 PMCID: PMC5470144 DOI: 10.1177/1536012117708722] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Phosphatidylserine (PS), the most abundant anionic phospholipid in cell membrane, is strictly confined to the inner leaflet in normal cells. However, this PS asymmetry is found disruptive in many tumor vascular endothelial cells. We discuss the underlying mechanisms for PS asymmetry maintenance in normal cells and its loss in tumor cells. The specificity of PS exposure in tumor vasculature but not normal blood vessels may establish it a useful biomarker for cancer molecular imaging. Indeed, utilizing PS-targeting antibodies, multiple imaging probes have been developed and multimodal imaging data have shown their high tumor-selective targeting in various cancers. There is a critical need for improved diagnosis and therapy for brain tumors. We have recently established PS-targeted nanoplatforms, aiming to enhance delivery of imaging contrast agents across the blood-brain barrier to facilitate imaging of brain tumors. Advantages of using the nanodelivery system, in particular, lipid-based nanocarriers, are discussed here. We also describe our recent research interest in developing PS-targeted nanotheranostics for potential image-guided drug delivery to treat brain tumors.
Collapse
Affiliation(s)
- Lulu Wang
- 1 Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Amyn A Habib
- 2 Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,3 North Texas VA Medical Center, Dallas, TX, USA
| | - Akiva Mintz
- 4 Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,5 Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - King C Li
- 4 Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,6 Clinical and Translational Science Institute, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Dawen Zhao
- 1 Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, NC, USA.,3 North Texas VA Medical Center, Dallas, TX, USA
| |
Collapse
|
70
|
Liang Y, Mafuvadze B, Besch-Williford C, Hyder SM. A combination of p53-activating APR-246 and phosphatidylserine-targeting antibody potently inhibits tumor development in hormone-dependent mutant p53-expressing breast cancer xenografts. BREAST CANCER-TARGETS AND THERAPY 2018; 10:53-67. [PMID: 29606888 PMCID: PMC5868596 DOI: 10.2147/bctt.s156285] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Between 30 and 40% of human breast cancers express a defective tumor suppressor p53 gene. Wild-type p53 tumor suppressor protein promotes cell-cycle arrest and apoptosis and inhibits vascular endothelial growth factor-dependent angiogenesis, whereas mutant p53 protein (mtp53) lacks these functions, resulting in tumor cell survival and metastasis. Restoration of p53 function is therefore a promising drug-targeted strategy for combating mtp53-expressing breast cancer. Methods In this study, we sought to determine whether administration of APR-246, a small-molecule drug that restores p53 function, in combination with 2aG4, an antibody that targets phosphatidylserine residues on tumor blood vessels and disrupts tumor vasculature, effectively inhibits advanced hormone-dependent breast cancer tumor growth. Results APR-246 reduced cell viability in mtp53-expressing BT-474 and T47-D human breast cancer cells in vitro, and significantly induced apoptosis in a dose-dependent manner. However, APR-246 did not reduce cell viability in MCF-7 breast cancer cells, which express wild-type p53. We next examined APR-246's anti-tumor effects in vivo using BT-474 and T47-D tumor xenografts established in female nude mice. Tumor-bearing mice were treated with APR-246 and/or 2aG4 and tumor volume followed over time. Tumor growth was more effectively suppressed by combination treatment than by either agent alone, and combination therapy completely eradicated some tumors. Immunohistochemistry analysis of tumor tissue sections demonstrated that combination therapy more effectively induced apoptosis and reduced cell proliferation in tumor xenografts than either agent alone. Importantly, combination therapy dramatically reduced the density of blood vessels, which serve as the major route for tumor metastasis, in tumor xenografts compared with either agent alone. Conclusion Based on our findings, we contend that breast tumor growth might effectively be controlled by simultaneous targeting of mtp53 protein and tumor blood vessels in mtp53-expressing cancers.
Collapse
Affiliation(s)
- Yayun Liang
- Deparment of Biomedical Sciences and Dalton Cardiovascular Research Center, Columbia, MO, USA
| | - Benford Mafuvadze
- Deparment of Biomedical Sciences and Dalton Cardiovascular Research Center, Columbia, MO, USA
| | | | - Salman M Hyder
- Deparment of Biomedical Sciences and Dalton Cardiovascular Research Center, Columbia, MO, USA
| |
Collapse
|
71
|
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: 4.2] [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
|
72
|
Seth A, Lee H, Cho MY, Park C, Korm S, Lee JY, Choi I, Lim YT, Hong KS. Combining vasculature disrupting agent and Toll-like receptor 7/8 agonist for cancer therapy. Oncotarget 2018; 8:5371-5381. [PMID: 28036266 PMCID: PMC5354915 DOI: 10.18632/oncotarget.14260] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/07/2016] [Indexed: 12/22/2022] Open
Abstract
This study evaluates the effect of combination of two different treatment regimens for solid tumor therapy: vasculature targeting agent and immune-stimulation. Poly lactide-co-glycolide (PLGA) nanoparticles were synthesized for intracellular delivery of toll-like receptor (TLR) 7/8 agonist—gardiquimod. Spherical and mono-disperse gardiquimod encapsulated PLGA nanoparticles (Gardi-PLGA), approximately 194 nm in size were formulated. Gardi-PLGA induced immune-stimulation, and vasculature disrupting agent (VDA)—5,6-Dimethylxanthenone-4-acetic acid (DMXAA) was used in combination to assessing the influence on bone marrow derived dendritic cells (BMDCs) and B16-F10 melanoma cells. The combination treatment significantly increased the levels of pro-inflammatory cytokines, indicating their activation in BMDCs, while melanoma cells remained viable. Further, mice melanoma model was established, and DMXAA was administered intraperitoneally and Gardi-PLGA was administered via an intra-tumoral injection. The combination treatments strategy significantly inhibited tumor growth as shown by tumor volume analysis, and the survival rate of the mice was found to be 63.6% (n = 11), after 54 days of tumor inoculation. Immunohistochemical findings of tumor sections treated with DMXAA confirmed the in vivo vasculature disruption. Thus, the inhibition of tumor growth can be attributed to the synergistic effect of immune stimulation caused by DC activation and vasculature disruption.
Collapse
Affiliation(s)
- Anushree Seth
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Republic of Korea.,Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea.,Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Hyunseung Lee
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Republic of Korea.,Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Mi Young Cho
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Republic of Korea.,Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Cheongsoo Park
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Sovannarith Korm
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Joo-Yong Lee
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Inpyo Choi
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kwan Soo Hong
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Republic of Korea.,Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea.,Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| |
Collapse
|
73
|
Targeted drug delivery for tumor therapy inside the bone marrow. Biomaterials 2018; 155:191-202. [DOI: 10.1016/j.biomaterials.2017.11.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/26/2017] [Accepted: 11/21/2017] [Indexed: 12/18/2022]
|
74
|
Recent Progress in Synthesis and Functionalization of Multimodal Fluorescent-Magnetic Nanoparticles for Biological Applications. APPLIED SCIENCES-BASEL 2018. [DOI: 10.3390/app8020172] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
There is a great interest in the development of new nanomaterials for multimodal imaging applications in biology and medicine. Multimodal fluorescent-magnetic based nanomaterials deserve particular attention as they can be used as diagnostic and drug delivery tools, which could facilitate the diagnosis and treatment of cancer and many other diseases. This review focuses on the recent developments of magnetic-fluorescent nanocomposites and their biomedical applications. The recent advances in synthetic strategies and approaches for the preparation of fluorescent-magnetic nanocomposites are presented. The main biomedical uses of multimodal fluorescent-magnetic nanomaterials, including biological imaging, cancer therapy and drug delivery, are discussed, and prospects of this field are outlined.
Collapse
|
75
|
Abma E, Peremans K, De Vos F, Bosmans T, Kitshoff AM, Daminet S, Ni Y, Dockx R, de Rooster H. Biodistribution and tolerance of intravenous iodine-131-labelled hypericin in healthy dogs. Vet Comp Oncol 2018; 16:318-323. [PMID: 29314561 DOI: 10.1111/vco.12381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/23/2017] [Accepted: 12/10/2017] [Indexed: 01/21/2023]
Abstract
Hypericin (Hyp) is a necrosis-avid compound that can be efficiently labelled with radioiodine for both diagnostic and therapeutic purposes. Before 131 I-Hyp can be considered as a clinically useful drug in a combination therapy for canine cancer patients, evaluation of its toxicity is necessary. The aim of this study was to investigate the biodistribution and tolerance of a single dose administration of 131 I-Hyp. Three healthy dogs were included. 131 I-Hyp at a dose of 0.2 mg/kg and an activity of 185 MBq was intravenously injected. The effects on physical, haematological and biochemical parameters were characterized and the biodistribution and elimination pattern, the effective half-life and dose rate were assessed. Drug-related adverse events were limited to mild gastrointestinal signs, resolving within 48 hours. No significant differences were found in blood haematology and serum biochemistry before and after treatment. Following administration, highest percentage of injected dose (%ID ± SD) was found in the liver (5.5 ± 0.33), the lungs (4.17 ± 0.14) and the heart (3.11 ± 0.78). After 24 hours, highest %ID was found in colon (4.25 ± 1.45) and liver (3.45 ± 0.60). Clearance from all organs was effective within 7 days. Effective half-life was established at 80 hours, and the dose rate fell below <20 μSv/h at 1 m within 1 day. The current study reveals that single dose treatment with 131 I-Hyp at the described dose is well tolerated by healthy dogs and supports the use of radioiodinated hypericin in a combination therapy for canine cancer patients.
Collapse
Affiliation(s)
- E Abma
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - K Peremans
- Department of Medical Imaging and Orthopaedics of Small Animals, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - F De Vos
- Laboratory of Pharmaceutical Technology, Ghent University, Ghent, Belgium
| | - T Bosmans
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - A M Kitshoff
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - S Daminet
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Y Ni
- Theragnostic Lab, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - R Dockx
- Department of Medical Imaging and Orthopaedics of Small Animals, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - H de Rooster
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
76
|
Synthesis and biological evaluation of N-substituted 3-oxo-1,2,3,4-tetrahydro-quinoxaline-6-carboxylic acid derivatives as tubulin polymerization inhibitors. Eur J Med Chem 2018; 143:8-20. [DOI: 10.1016/j.ejmech.2017.08.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 08/05/2017] [Accepted: 08/06/2017] [Indexed: 11/23/2022]
|
77
|
Eggers R, Philippi A, Altmeyer MO, Breinig F, Schmitt MJ. Primary T cells for mRNA-mediated immunotoxin delivery. Gene Ther 2018; 25:47-53. [PMID: 28937681 DOI: 10.1038/gt.2017.87] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/23/2017] [Accepted: 09/18/2017] [Indexed: 12/30/2022]
Abstract
Immune cells become increasingly attractive as delivery system for immunotoxins in cancer therapy to reduce the intrinsic toxicity and severe side effects of chimeric protein toxins. In this study, we investigated the potential of human primary T cells to deliver a secreted immunotoxin through transient messenger RNA (mRNA) transfection. The chimeric protein toxin was directed toward the neovasculature of cancer cells by fusing a truncated version of Pseudomonas exotoxin A (PE38) to human vascular endothelial growth factor (VEGF) and to the single chain variable fragment (scFv) of anti-Her2/neu. Protocols for the transient transfection of human embryonic kidney cells (HEK293) as well as activated primary human T cells were established. Transient transfection with mRNA coding for the immunotoxins e23-PE38, VEGF-PE38 and its attenuated variant VEGF-PE38D yielded efficient expression and secretion. Mass spectrometry analysis endorsed that a fraction of VEGF-PE38D was properly translocated into the endoplasmic reticulum. Furthermore, cytotoxic activity of immunotoxin secreting T cells toward cancer cells was confirmed in co-culture with ovarian adenocarcinoma cells in the presence of a bispecific antibody (bsAb), highlighting the potential of primary T cells for mRNA-mediated immunotoxin delivery.
Collapse
Affiliation(s)
- R Eggers
- Environmental safety group, Korea Institute of Science and Technology (KIST Europe), Saarbrücken, Germany
| | - A Philippi
- Environmental safety group, Korea Institute of Science and Technology (KIST Europe), Saarbrücken, Germany
| | - M O Altmeyer
- Environmental safety group, Korea Institute of Science and Technology (KIST Europe), Saarbrücken, Germany
| | - F Breinig
- Department of Biosciences, Molecular and Cell Biology and Center of Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
| | - M J Schmitt
- Department of Biosciences, Molecular and Cell Biology and Center of Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
| |
Collapse
|
78
|
Fathy M, Awale S, Nikaido T. Phosphorylated Akt Protein at Ser473 Enables HeLa Cells to Tolerate Nutrient-Deprived Conditions. Asian Pac J Cancer Prev 2017; 18:3255-3260. [PMID: 29286216 PMCID: PMC5980880 DOI: 10.22034/apjcp.2017.18.12.3255] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Despite angiogenesis, many tumours remain hypovascular and starved of nutrients while continuing to grow rapidly. The specific biochemical mechanisms associated with starvation resistance, austerity, may be new biological characters of cancer that are critical for cancer progression. Objective: This study aim was to investigate the effect of nutrient starvation on HeLa cells and the possible mechanism by which the cells are able to tolerate nutrient-deprived conditions. Methods: Nutrient starvation was achieved by culturing HeLa cells in nutrient-deprived medium (NDM) and cell survival was estimated by using cell counting kit-8. The effect of starvation on cell cycle distribution and the quantitative analysis of apoptotic cells were investigated by flow cytometry using propidium iodide staining. Western blotting was used to detect the expression levels of Akt and phosphorylated Akt at Ser473 (Ser473p-Akt) proteins. Results: HeLa cells displayed extremely long survival when cultured in NDM. The percentage of apoptotic HeLa cells was significantly increased by starvation in a time-dependent manner. A significant increase in the expression of Ser473p-Akt protein after starvation was also observed. Furthermore, it was found that Akt inhibitor III molecule inhibited the cells proliferation in a concentration- and time-dependent manner. Conclusion: Results of the present study provide evidence that Akt activation may be implicated in the tolerance of HeLa cells for nutrient starvation and may help to suggest new therapeutic strategies designed to prevent austerity of cervical cancer cells through inhibition of Akt activation.
Collapse
Affiliation(s)
- Moustafa Fathy
- Department of Regenerative Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.,Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | | | | |
Collapse
|
79
|
Design, synthesis, and biological evaluation of novel combretastatin A-4 thio derivatives as microtubule targeting agents. Eur J Med Chem 2017; 144:797-816. [PMID: 29291446 DOI: 10.1016/j.ejmech.2017.11.050] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/17/2017] [Accepted: 11/18/2017] [Indexed: 11/20/2022]
Abstract
A series of novel combretastatin A-4 (CA-4) thio derivatives containing different molecular cores, namely α-phenylcinnamic acids (core 1), (Z)-stilbenes (core 2), 4,5-disubstituted oxazoles (core 3), and 4,5-disubstituted N-methylimidazoles (core 4), as cis-restricted analogues were designed and synthesized. They were selected with the use of a parallel virtual screening protocol including the generation of a virtual combinatorial library based on an elaborated synthesis protocol of CA-4 analogues. The selected compounds were evaluated for antiproliferative activity against a panel of six human cancer cell lines (A431, HeLa, MCF7, MDA-MB-231, A549 and SKOV) and two human non-cancer cell lines (HaCaT and CCD39Lu). Moreover, the effect of the test compounds on the inhibition of tubulin polymerization in vitro was estimated. In the series studied here, oxazole-bridged analogues exhibited the most potent antiproliferative activity. Compounds 23a, 23e, and 23i efficiently inhibited tubulin polymerization with IC50 values of 0.86, 1.05, and 0.85 μM, respectively. Thio derivative 23i, when compared to its oxygen analogue 23j, showed a 5-fold higher inhibitory impact on tubulin polymerization. Compounds 23e and 23i, which showed both best cytotoxic and antitubulin activity, were further studied in terms of their effect on cell cycle distribution and proapoptotic activity. Compound 23e induced a statistically significant block of the cell cycle at the G2/M phase in A431, HaCaT, HeLa, MCF-7, MDA-MB-231, and SKOV-3 cells to an extent comparable to that observed in CA-4. In HeLa and SKOV-3 cells incubated with 23i, a concentration-dependent block of the G2/M phase was observed. The proapoptotic effect of 23e and 23i in A431, HaCaT, MCF-7, MDA-MB-231, and SKOV-3 was demonstrated with ELISA assay and double staining with Annexin V-FITC/PI. The results indicated that compound 23e and 23i may serve as novel lead compounds in research on more effective anticancer agents.
Collapse
|
80
|
Bähr O, Gross S, Harter PN, Kirches E, Mawrin C, Steinbach JP, Mittelbronn M. ASA404, a vascular disrupting agent, as an experimental treatment approach for brain tumors. Oncol Lett 2017; 14:5443-5451. [PMID: 29098034 PMCID: PMC5652230 DOI: 10.3892/ol.2017.6832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/07/2017] [Indexed: 01/06/2023] Open
Abstract
Malignant brain tumors, including gliomas, brain metastases and anaplastic meningiomas, are associated with poor prognosis, and represent an unmet medical need. ASA404 (DMXAA), a vascular disrupting agent, has demonstrated promising results in several preclinical tumor models and early phase clinical trials. However, two phase III trials in non-small cell lung cancer reported insufficient results. The aim of the present study was to determine the effects of ASA404 on brain tumors. The effects of ASA404 were evaluated in vitro and in vivo using subcutaneous, and orthotopical models for malignant glioma (U-87, LN-229, U-251, LN-308 and Tu-2449), brain metastasis (HT-29) and malignant meningioma (IOMM-Lee). The acute effects of ASA404 on tumor tissue were analyzed using conventional and immunohistochemical staining techniques [hematoxylin and eosin, MIB-1 antibody/proliferation maker protein Ki-67, cleaved caspase-8, stimulator of interferon genes (STING), ionized calcium-binding adapter molecule 1]. Furthermore, the sizes of subcutaneous tumors were measured and the symptom-free survival rates of animals with intracranial tumors receiving ASA404 treatment were analyzed. ASA404 demonstrated low toxicity in vitro, but exhibited strong effects on subcutaneous tumors 24 h following a single dose of ASA404 (25 mg/kg). ASA404 induced necrosis, hemorrhages and inhibited the proliferation, and growth of tumors in the subcutaneous glioma models. However, ASA404 failed to demonstrate comparable effects in any of the intracranial tumor models examined and did not result in a prolongation of survival. Expression of STING, the molecular target of ASA404, and infiltration of macrophages, the cells mediating ASA404 activity, did not differ between subcutaneous and intracranial tumors. In conclusion, ASA404 demonstrates clear efficacy in subcutaneous tumor models, but has no relevant activity in orthotopic brain tumor models. The expression of STING and infiltration with macrophages were not determined to be involved in the differential activity observed among tumor models. It is possible that the low penetration of ASA-404 into the brain prevents concentrations sufficient enough reaching the tumor in order to exhibit acute effects in vivo.
Collapse
Affiliation(s)
- Oliver Bähr
- Dr. Senckenberg Institute of Neurooncology, Goethe-University Hospital, Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefanie Gross
- Dr. Senckenberg Institute of Neurooncology, Goethe-University Hospital, Frankfurt, Germany
| | - Patrick N Harter
- Institute of Neurology (Edinger-Institute), Goethe-University, Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elmar Kirches
- Institute of Neuropathology, Otto-von-Guericke University, Magdeburg, Germany
| | - Christian Mawrin
- Institute of Neuropathology, Otto-von-Guericke University, Magdeburg, Germany
| | - Joachim P Steinbach
- Dr. Senckenberg Institute of Neurooncology, Goethe-University Hospital, Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michel Mittelbronn
- Institute of Neurology (Edinger-Institute), Goethe-University, Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Laboratoire National de Santé, Dudelange, Luxembourg.,Luxembourg Centre of Neuropathology (LCNP), Luxembourg City, Luxembourg.,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.,NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Strassen, Luxembourg
| |
Collapse
|
81
|
Federau C. Intravoxel incoherent motion MRI as a means to measure in vivo perfusion: A review of the evidence. NMR IN BIOMEDICINE 2017; 30. [PMID: 28885745 DOI: 10.1002/nbm.3780] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/19/2017] [Accepted: 07/07/2017] [Indexed: 05/07/2023]
Abstract
The idea that in vivo intravoxel incoherent motion magnetic resonance signal is influenced by blood motion in the microvasculature is exciting, because it suggests that local and quantitative perfusion information can be obtained in a simple and elegant way from a few diffusion-weighted images, without contrast injection. When the method was proposed in the late 1980s some doubts appeared as to its feasibility, and, probably because the signal to noise and image quality at the time was not sufficient, no obvious experimental evidence could be produced to alleviate them. Helped by the tremendous improvements seen in the last three decades in MR hardware, pulse design, and post-processing capabilities, an increasing number of encouraging reports on the value of intravoxel incoherent motion perfusion imaging have emerged. The aim of this article is to review the current published evidence on the feasibility of in vivo perfusion imaging with intravoxel incoherent motion MRI.
Collapse
Affiliation(s)
- Christian Federau
- Division of Diagnostic and Interventional Neuroradiology, Department of Radiology, University Hospital Basel, Petersgraben, Basle, Switzerland
| |
Collapse
|
82
|
Tsiambas E, Stamatelopoulos A, Karameris A, Panagiotou I, Rigopoulos D, Chatzimichalis A, Bouros D, Patsouris E. Simultaneous EGFR and VEGF Alterations in Non-Small Cell Lung Carcinoma Based on Tissue Microarrays. Cancer Inform 2017. [DOI: 10.1177/117693510700300026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Epidermal growth factor receptor (EGFR) overexpression is observed in significant proportions of non-small cell lung carcinomas (NSCLC). Furthermore, overactivation of vascular endothelial growth factor (VEGF) leads to increased angiogenesis implicated as an important factor in vascularization of those tumors. Patients and Methods Using tissue microarray technology, forty-paraffin ( n = 40) embedded, histologically confirmed primary NSCLCs were cored and re-embedded into a recipient block. Immunohistochemistry was performed for the determination of EGFR and VEGF protein levels which were evaluated by the performance of computerized image analysis. EGFR gene amplification was studied by chromogenic in situ hybridization based on the use of EGFR gene and chromosome 7 centromeric probes. Results EGFR overexpression was observed in 23/40 (57.5%) cases and was correlated to the stage of the tumors ( p = 0.001), whereas VEGF was overexpressed in 35/40 (87.5%) cases and was correlated to the stage of the tumors ( p = 0.005) and to the smoking history of the patients ( p = 0.016). Statistical significance was assessed comparing the protein levels of EGFR and VEGF ( p = 0.043, k = 0.846). EGFR gene amplification was identified in 2/40 (5%) cases demonstrating no association to its overall protein levels ( p = 0.241), whereas chromosome 7 aneuploidy was detected in 7/40 (17.5%) cases correlating to smoking history of the patients ( p = 0.013). Conclusions A significant subset of NSCLC is characterized by EGFR and VEGF simultaneous overexpression and maybe this is the eligible target group for the application of combined anti-EGFR/VEGF targeted therapies at the basis of genetic deregulation (especially gene amplification for EGFR).
Collapse
Affiliation(s)
- Evangelos Tsiambas
- Department of Pathology, Tissue Microarrays and Computerized Image Analysis Laboratory, 417 VA Hospital (NIMTS), Athens, Greece
- Departmentt of Pathology, Medical School, University of Athens, Greece
| | | | - Andreas Karameris
- Department of Pathology, Tissue Microarrays and Computerized Image Analysis Laboratory, 417 VA Hospital (NIMTS), Athens, Greece
- Departmentt of Pathology, Medical School, University of Athens, Greece
| | | | | | | | - Demosthenes Bouros
- Department of Respiratory Diseases, “Demokrition” University of Thrace, Alexandropole, Greece
| | | |
Collapse
|
83
|
Raoufi-Rad N, McRobb LS, Lee VS, Bervini D, Grace M, Ukath J, Mchattan J, Sreenivasan VKA, Duong TTH, Zhao Z, Stoodley MA. In vivo imaging of endothelial cell adhesion molecule expression after radiosurgery in an animal model of arteriovenous malformation. PLoS One 2017; 12:e0185393. [PMID: 28949989 PMCID: PMC5614630 DOI: 10.1371/journal.pone.0185393] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/12/2017] [Indexed: 12/18/2022] Open
Abstract
Focussed radiosurgery may provide a means of inducing molecular changes on the luminal surface of diseased endothelium to allow targeted delivery of novel therapeutic compounds. We investigated the potential of ionizing radiation to induce surface expression of intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) on endothelial cells (EC) in vitro and in vivo, to assess their suitability as vascular targets in irradiated arteriovenous malformations (AVMs). Cultured brain microvascular EC were irradiated by linear accelerator at single doses of 0, 5, 15 or 25 Gy and expression of ICAM-1 and VCAM-1 measured by qRT-PCR, Western, ELISA and immunocytochemistry. In vivo, near-infrared (NIR) fluorescence optical imaging using Xenolight 750-conjugated ICAM-1 or VCAM-1 antibodies examined luminal biodistribution over 84 days in a rat AVM model after Gamma Knife surgery at a single 15 Gy dose. ICAM-1 and VCAM-1 were minimally expressed on untreated EC in vitro. Doses of 15 and 25 Gy stimulated expression equally; 5 Gy was not different from the unirradiated. In vivo, normal vessels did not bind or retain the fluorescent probes, however binding was significant in AVM vessels. No additive increases in probe binding were found in response to radiosurgery at a dose of 15 Gy. In summary, radiation induces adhesion molecule expression in vitro but elevated baseline levels in AVM vessels precludes further induction in vivo. These molecules may be suitable targets in irradiated vessels without hemodynamic derangement, but not AVMs. These findings demonstrate the importance of using flow-modulated, pre-clinical animal models for validating candidate proteins for vascular targeting in irradiated AVMs.
Collapse
Affiliation(s)
- Newsha Raoufi-Rad
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Lucinda S. McRobb
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Vivienne S. Lee
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - David Bervini
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
- Neurosurgery Department, Inselspital, University of Bern, Bern, Switzerland
| | - Michael Grace
- Genesis Cancer Care, Macquarie University Hospital, Sydney, New South Wales, Australia
| | - Jaysree Ukath
- Genesis Cancer Care, Macquarie University Hospital, Sydney, New South Wales, Australia
| | - Joshua Mchattan
- Carestream Molecular Imaging, Sydney, New South Wales, Australia
| | - Varun K. A. Sreenivasan
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
- Department of Physics and Astronomy, Macquarie University, Sydney, New South Wales, Australia
| | - T. T. Hong Duong
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Zhenjun Zhao
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Marcus A. Stoodley
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
84
|
David A. Peptide ligand-modified nanomedicines for targeting cells at the tumor microenvironment. Adv Drug Deliv Rev 2017; 119:120-142. [PMID: 28506743 DOI: 10.1016/j.addr.2017.05.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/17/2017] [Accepted: 05/09/2017] [Indexed: 02/06/2023]
Abstract
Since their initial discovery more than 30years ago, tumor-homing peptides have become an increasingly useful tool for targeted delivery of therapeutic and diagnostic agents into tumors. Today, it is well accepted that cells at the tumor microenvironment (TME) contribute in many ways to cancer development and progression. Tumor-homing peptide-decorated nanomedicines can interact specifically with surface receptors expressed on cells in the TME, improve cellular uptake of nanomedicines by target cells, and impair tumor growth and progression. Moreover, peptide ligand-modified nanomedicines can potentially accumulate in the target tissue at higher concentrations than would small conjugates, thus increasing overall target tissue exposure to the therapeutic agent, enhance therapeutic efficacy and reduce side effects. This review describes the most studied peptide ligands aimed at targeting cells in the TME, discusses major obstacles and principles in the design of ligands for drug targeting and provides an overview of homing peptides in ligand-targeted nanomedicines that are currently in development for cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Ayelet David
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, and the Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
85
|
Siemann DW, Chaplin DJ, Horsman MR. Realizing the Potential of Vascular Targeted Therapy: The Rationale for Combining Vascular Disrupting Agents and Anti-Angiogenic Agents to Treat Cancer. Cancer Invest 2017; 35:519-534. [DOI: 10.1080/07357907.2017.1364745] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- D. W. Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | | | - M. R. Horsman
- Department of Experimental Clinical Oncology, Aarhus University, Denmark
| |
Collapse
|
86
|
Jahanban-Esfahlan R, Seidi K, Banimohamad-Shotorbani B, Jahanban-Esfahlan A, Yousefi B. Combination of nanotechnology with vascular targeting agents for effective cancer therapy. J Cell Physiol 2017; 233:2982-2992. [PMID: 28608554 DOI: 10.1002/jcp.26051] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/12/2017] [Indexed: 12/28/2022]
Abstract
As a young science, nanotechnology promptly integrated into the current oncology practice. Accordingly, various nanostructure particles were developed to reduce drug toxicity and allow the targeted delivery of various diagnostic and therapeutic compounds to the cancer cells. New sophisticated nanosystems constantly emerge to improve the performance of current anticancer modalities. Targeting tumor vasculature is an attractive strategy to fight cancer. Though the idea was swiftly furthered from basic science to the clinic, targeting tumor vasculature had a limited potential in patients, where tumors relapse due to the development of multiple drug resistance and metastasis. The aim of this review is to discuss the advantages of nanosystem incorporation with various vascular targeting agents, including (i) endogen anti-angiogenic agents; (ii) inhibitors of angiogenesis-related growth factors; (iii) inhibitors of tyrosine kinase receptors; (iv) inhibitors of angiogenesis-related signaling pathways; (v) inhibitors of tumor endothelial cell-associated markers; and (vi) tumor vascular disrupting agents. We also review the efficacy of nanostructures as natural vascular targeting agents. The efficacy of each approach in cancer therapy is further discussed.
Collapse
Affiliation(s)
- Rana Jahanban-Esfahlan
- Faculty of Advanced Medical Sciences, Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khaled Seidi
- Faculty of Advanced Medical Sciences, Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Banimohamad-Shotorbani
- Research Institute for Fundamental Sciences (RIFS), University of Tabriz, Tabriz, Iran.,Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Medicine, Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Medicine, Molecular Targeting Therapy Research Group, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
87
|
An atlas of bloodstream-accessible bone marrow proteins for site-directed therapy of acute myeloid leukemia. Leukemia 2017; 32:510-519. [DOI: 10.1038/leu.2017.208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/15/2022]
|
88
|
Abma E, Smets P, Daminet S, Cornelis I, De Clercq K, Ni Y, Vlerick L, de Rooster H. A dose-escalation study of combretastatin A4-phosphate in healthy dogs. Vet Comp Oncol 2017. [PMID: 28620942 DOI: 10.1111/vco.12327] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Combretastatin A4-Phosphate (CA4P) is a vascular disrupting agent revealing promising results in cancer treatments for humans. The aim of this study was to investigate the safety and adverse events of CA4P in healthy dogs as a prerequisite to application of CA4P in dogs with cancer. Ten healthy dogs were included. The effects of escalating doses of CA4P on physical, haematological and biochemical parameters, systolic arterial blood pressure, electrocardiogram, echocardiographic variables and general wellbeing were characterised. Three different doses were tested: 50, 75 and 100 mg m-2 . At all 3 CA4P doses, nausea, abdominal discomfort as well as diarrhoea were observed for several hours following administration. Likewise, a low-grade neutropenia was observed in all dogs. Doses of 75 and 100 mg m-2 additionally induced vomiting and elevation of serum cardiac troponine I levels. At 100 mg m-2 , low-grade hypertension and high-grade neurotoxicity were also observed. In healthy dogs, doses up to 75 mg m-2 seem to be well tolerated. The severity of the neurotoxicity observed at 100 mg m-2 , although transient, does not invite to use this dose in canine oncology patients.
Collapse
Affiliation(s)
- E Abma
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - P Smets
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - S Daminet
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - I Cornelis
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - K De Clercq
- Laboratory of Pharmaceutical Technology, Ghent University, Ghent, Belgium
| | - Y Ni
- Department of Radiology, KU Leuven, Leuven, Belgium
| | - L Vlerick
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - H de Rooster
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
89
|
Kulshrestha A, Katara GK, Ibrahim SA, Patil R, Patil SA, Beaman KD. Microtubule inhibitor, SP-6-27 inhibits angiogenesis and induces apoptosis in ovarian cancer cells. Oncotarget 2017; 8:67017-67028. [PMID: 28978013 PMCID: PMC5620153 DOI: 10.18632/oncotarget.17549] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 03/21/2017] [Indexed: 01/21/2023] Open
Abstract
In ovarian cancer (OVCA), treatment failure due to chemo-resistance is a serious challenge. It is therefore critical to identify new therapies that are effective against resistant tumors and have reduced side effects. We recently identified 4-H-chromenes as tubulin depolymerizing agents that bind to colchicine site of beta-tubulin. Here, we screened a chemical library of substituted 4-H-chromenes and identified SP-6-27 to exhibit most potent anti-proliferative activity towards a panel of human cisplatin sensitive and resistant OVCA cell lines with 50% inhibitory concentration (IC50; mean ± SD) ranging from 0.10 ± 0.01 to 0.84 ± 0.20 μM. SP-6-27 exhibited minimum cytotoxicity to normal ovarian epithelia. A pronounced decrease in microtubule density as well as G2/M cell cycle arrest was observed in SP-6-27 treated cisplatin sensitive/resistant OVCA cells. The molecular mechanism of SP-6-27 induced cell death revealed modulation in cell-cycle regulation by upregulation of growth arrest and DNA damage inducible alpha transcripts (GADD45). An enhanced intrinsic apoptosis was observed in OVCA cells through upregulation of Bax, Apaf-1, caspase-6, -9, and caspase-3. In vitro wound healing assay revealed reduced OVCA cell migration upon SP-6-27 treatment. Additionally, SP-6-27 and cisplatin combinatorial treatment showed enhanced cytotoxicity in chemo-sensitive/resistant OVCA cells. Besides effect on cancer cells, SP-6-27 further restrained angiogenesis by inhibiting capillary tube formation by human umbilical vein endothelial cells (HUVEC). Together, these findings show that the chromene analog SP-6-27 is a novel chemotherapeutic agent that offers important advantages for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Arpita Kulshrestha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, USA
| | - Safaa A Ibrahim
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, USA.,Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Renukadevi Patil
- Pharmaceutical Sciences Department, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, USA
| | - Shivaputra A Patil
- Pharmaceutical Sciences Department, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, USA
| |
Collapse
|
90
|
Seidi K, Jahanban-Esfahlan R, Zarghami N. Tumor rim cells: From resistance to vascular targeting agents to complete tumor ablation. Tumour Biol 2017; 39:1010428317691001. [DOI: 10.1177/1010428317691001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Current vascular targeting strategies pursue two main goals: anti-angiogenesis agents aim to halt sprouting and the formation of new blood vessels, while vascular disrupting agents along with coaguligands seek to compromise blood circulation in the vessels. The ultimate goal of such therapies is to deprive tumor cells out of oxygen and nutrients long enough to succumb cancer cells to death. Most of vascular targeting agents presented promising therapeutic potential, but the final goal which is cure is rarely achieved. Nevertheless, in both preclinical and clinical settings, tumors tend to grow back, featuring a highly invasive, metastatic, and extremely resistant form. This review highlights the critical significance of tumor rim cells as the main factor, determining therapy success with vascular targeting agents. We present an overview of different single and combination treatments with vascular targeting agents that enable efficient targeting of tumor rim cells and long-lasting tumor cure. Understanding the nature of tumor rim cells, how they establish, how they manage to survive of vascular targeting agents, and how they contribute in tumor refractoriness, may open new avenues to the development of beneficial strategies, capable to eliminate residual rim cells, and enable tumor ablation once and forever.
Collapse
Affiliation(s)
- Khaled Seidi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
91
|
Gangeh MJ, Hashim A, Giles A, Sannachi L, Czarnota GJ. Computer aided prognosis for cell death categorization and prediction in vivo using quantitative ultrasound and machine learning techniques. Med Phys 2017; 43:6439. [PMID: 27908167 DOI: 10.1118/1.4967265] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
PURPOSE At present, a one-size-fits-all approach is typically used for cancer therapy in patients. This is mainly because there is no current imaging-based clinical standard for the early assessment and monitoring of cancer treatment response. Here, the authors have developed, for the first time, a complete computer-aided-prognosis (CAP) system based on multiparametric quantitative ultrasound (QUS) spectroscopy methods in association with texture descriptors and advanced machine learning techniques. This system was used to noninvasively categorize and predict cell death levels in fibrosarcoma mouse tumors treated using ultrasound-stimulated microbubbles as novel endothelial-cell radiosensitizers. METHODS Sarcoma xenograft tumor-bearing mice were treated using ultrasound-stimulated microbubbles, alone or in combination with x-ray radiation therapy, as a new antivascular treatment. Therapy effects were assessed at 2-3, 24, and 72 h after treatment using a high-frequency ultrasound. Two-dimensional spectral parametric maps were generated using the power spectra of the raw radiofrequency echo signal. Subsequently, the distances between "pretreatment" and "post-treatment" scans were computed as an indication of treatment efficacy, using a kernel-based metric on textural features extracted from 2D parametric maps. A supervised learning paradigm was used to either categorize cell death levels as low, medium, or high using a classifier, or to "continuously" predict the levels of cell death using a regressor. RESULTS The developed CAP system performed at a high level for the classification of cell death levels. The area under curve of the receiver operating characteristic was 0.87 for the classification of cell death levels to both low/medium and medium/high levels. Moreover, the prediction of cell death levels using the proposed CAP system achieved a good correlation (r = 0.68, p < 0.001) with histological cell death levels as the ground truth. A statistical test of significance between individual treatment groups with the corresponding control group demonstrated that the predicted levels indicated the same significant changes in cell death as those indicated by the ground-truth levels. CONCLUSIONS The technology developed in this study addresses a gap in the current standard of care by introducing a quality control step that generates potentially actionable metrics needed to enhance treatment decision-making. The study establishes a noninvasive framework for quantifying levels of cancer treatment response developed preclinically in tumors using QUS imaging in conjunction with machine learning techniques. The framework can potentially facilitate the detection of refractory responses in patients to a certain cancer treatment early on in the course of therapy to enable switching to more efficacious treatments.
Collapse
Affiliation(s)
- M J Gangeh
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, Ontario M5G 2M9, Canada and Departments of Radiation Oncology, and Imaging Research - Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | - A Hashim
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | - A Giles
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | - L Sannachi
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, Ontario M5G 2M9, Canada and Departments of Radiation Oncology, and Imaging Research - Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | - G J Czarnota
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, Ontario M5G 2M9, Canada and Departments of Radiation Oncology, and Imaging Research - Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| |
Collapse
|
92
|
Deng J, Wang Y. Quantitative magnetic resonance imaging biomarkers in oncological clinical trials: Current techniques and standardization challenges. Chronic Dis Transl Med 2017; 3:8-20. [PMID: 29063052 PMCID: PMC5627686 DOI: 10.1016/j.cdtm.2017.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Indexed: 12/21/2022] Open
Abstract
Radiological imaging plays an important role in oncological trials to provide imaging biomarkers for disease staging, stratifying patients, defining dose setting, and evaluating the safety and efficacy of new candidate drugs and innovative treatment. This paper reviews the techniques of most commonly used quantitative magnetic resonance imaging (qMRI) biomarkers (dynamic contrast enhanced, dynamic susceptibility contrast, and diffusion weighted imaging) and their applications in oncological trials. Challenges of incorporating qMRI biomarkers in oncological trials are discussed including understanding biological mechanisms revealed by MRI biomarkers, consideration of rigorous trial design and standardized implementation of qMRI protocols.
Collapse
Affiliation(s)
- Jie Deng
- Department of Medical Imaging, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.,Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Beijing, 100044, China
| |
Collapse
|
93
|
Shaikh MV, Kala M, Nivsarkar M. Formulation and optimization of doxorubicin loaded polymeric nanoparticles using Box-Behnken design: ex-vivo stability and in-vitro activity. Eur J Pharm Sci 2017; 100:262-272. [DOI: 10.1016/j.ejps.2017.01.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/31/2016] [Accepted: 01/22/2017] [Indexed: 01/18/2023]
|
94
|
McRobb LS, Lee VS, Simonian M, Zhao Z, Thomas SG, Wiedmann M, Raj JVA, Grace M, Moutrie V, McKay MJ, Molloy MP, Stoodley MA. Radiosurgery Alters the Endothelial Surface Proteome: Externalized Intracellular Molecules as Potential Vascular Targets in Irradiated Brain Arteriovenous Malformations. Radiat Res 2017; 187:66-78. [PMID: 28054837 DOI: 10.1667/rr14518.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Stereotactic radiosurgery (SRS) is an established treatment for brain arteriovenous malformations (AVMs) that drives blood vessel closure through cellular proliferation, thrombosis and fibrosis, but is limited by a delay to occlusion of 2-3 years and a maximum treatable size of 3 cm. In this current study we used SRS as a priming tool to elicit novel protein expression on the endothelium of irradiated AVM vessels, and these proteins were then targeted with prothrombotic conjugates to induce rapid thrombosis and vessel closure. SRS-induced protein changes on the endothelium in an animal model of AVM were examined using in vivo biotin labeling of surface-accessible proteins and comparative proteomics. LC-MS/MS using SWATH acquisition label-free mass spectrometry identified 280 proteins in biotin-enriched fractions. The abundance of 56 proteins increased after irradiation of the rat arteriovenous fistula (20 Gy, ≥1.5-fold). A large proportion of intracellular proteins were present in this subset: 29 mitochondrial and 9 cytoskeletal. Three of these proteins were chosen for further validation based on previously published evidence for surface localization and a role in autoimmune stimulation: cardiac troponin I (TNNI3); manganese superoxide dismutase (SOD2); and the E2 subunit of the pyruvate dehydrogenase complex (PDCE2). Immunostaining of AVM vessels confirmed an increase in abundance of PDCE2 across the vessel wall, but not a measurable increase in TNNI3 or SOD2. All three proteins co-localized with the endothelium after irradiation, however, more detailed subcellular distribution could not be accurately established. In vitro, radiation-stimulated surface translocation of all three proteins was confirmed in nonpermeabilized brain endothelial cells using immunocytochemistry. Total protein abundance increased modestly after irradiation for PDCE2 and SOD2 but decreased for TNNI3, suggesting that radiation primarily affects subcellular distribution rather than protein levels. The novel identification of these proteins as surface exposed in response to radiation raises important questions about their potential role in radiation-induced inflammation, fibrosis and autoimmunity, but may also provide unique candidates for vascular targeting in brain AVMs and other vascular tissues.
Collapse
Affiliation(s)
- Lucinda S McRobb
- a Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Vivienne S Lee
- a Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Margaret Simonian
- a Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia.,c Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California
| | - Zhenjun Zhao
- a Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Santhosh George Thomas
- a Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Markus Wiedmann
- a Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Jude V Amal Raj
- a Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Michael Grace
- d Genesis Cancer Care, Macquarie University Hospital, Sydney, New South Wales, Australia
| | - Vaughan Moutrie
- d Genesis Cancer Care, Macquarie University Hospital, Sydney, New South Wales, Australia
| | - Matthew J McKay
- b Australian Proteome Analysis Facility, Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Mark P Molloy
- b Australian Proteome Analysis Facility, Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Marcus A Stoodley
- a Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| |
Collapse
|
95
|
Jahanban-Esfahlan R, Seidi K, Zarghami N. Tumor vascular infarction: prospects and challenges. Int J Hematol 2017; 105:244-256. [DOI: 10.1007/s12185-016-2171-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 12/21/2022]
|
96
|
Fruytier AC, Le Duff CS, Po C, Magat J, Bouzin C, Neveu MA, Feron O, Jordan BF, Gallez B. The Blood Flow Shutdown Induced by Combretastatin A4 Impairs Gemcitabine Delivery in a Mouse Hepatocarcinoma. Front Pharmacol 2016; 7:506. [PMID: 28066252 PMCID: PMC5179558 DOI: 10.3389/fphar.2016.00506] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 12/07/2016] [Indexed: 01/31/2023] Open
Abstract
In recent clinical studies, vascular disrupting agents (VDAs) are mainly used in combination with chemotherapy. However, an often overlooked concern in treatment combination is the VDA-induced impairment of chemotherapy distribution in the tumor. The work presented here investigated the impact of blood flow shutdown induced by Combretastatin A4 (CA4) on gemcitabine uptake into mouse hepatocarcinoma. At 2 h after CA4 treatment, using DCE-MRI, a significant decrease in the perfusion-relevant parameters Ktrans and Vp were observed in treated group compared with the control group. The blood flow shutdown was indeed confirmed by a histology study. In a third experiment, the total gemcitabine uptake was found to be significantly lower in treated tumors, as assessed in a separate experiment using ex vivo fluorine nuclear magnetic resonance spectroscopy. The amount of active metabolite gemcitabine triphosphate was also lower in treated tumors. In conclusion, the blood flow shutdown induced by VDAs can impact negatively on the delivery of small cytotoxic agents in tumors. The present study outlines the importance of monitoring the tumor vascular function when designing drug combinations.
Collapse
Affiliation(s)
- Anne-Catherine Fruytier
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain Brussels, Belgium
| | - Cecile S Le Duff
- Institute of Condensed Matter and Nanosciences, Université Catholique de Louvain Louvain-la-Neuve, Belgium
| | - Chrystelle Po
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain Brussels, Belgium
| | - Julie Magat
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain Brussels, Belgium
| | - Caroline Bouzin
- Institut de Recherche Expérimentale et Clinique, Pole of Pharmacology, Angiogenesis and Cancer Research Laboratory, Université Catholique de Louvain Brussels, Belgium
| | - Marie-Aline Neveu
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain Brussels, Belgium
| | - Olivier Feron
- Institut de Recherche Expérimentale et Clinique, Pole of Pharmacology, Angiogenesis and Cancer Research Laboratory, Université Catholique de Louvain Brussels, Belgium
| | - Benedicte F Jordan
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain Brussels, Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain Brussels, Belgium
| |
Collapse
|
97
|
Herdman CA, Strecker TE, Tanpure RP, Chen Z, Winters A, Gerberich J, Liu L, Hamel E, Mason RP, Chaplin DJ, Trawick ML, Pinney KG. Synthesis and Biological Evaluation of Benzocyclooctene-based and Indene-based Anticancer Agents that Function as Inhibitors of Tubulin Polymerization. MEDCHEMCOMM 2016; 7:2418-2427. [PMID: 28217276 PMCID: PMC5308454 DOI: 10.1039/c6md00459h] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The natural products colchicine and combretastatin A-4 (CA4) have been inspirational for the design and synthesis of structurally related analogues and spin-off compounds as inhibitors of tubulin polymerization. The discovery that a water-soluble phosphate prodrug salt of CA4 (referred to as CA4P) is capable of imparting profound and selective damage to tumor-associated blood vessels paved the way for the development of a new therapeutic approach for cancer treatment utilizing small-molecule inhibitors of tubulin polymerization that also act as vascular disrupting agents (VDAs). Combination of salient structural features associated with colchicine and CA4 led to the design and synthesis of a variety of fused aryl-cycloalkyl and aryl-heterocyclic compounds that function as inhibitors of tubulin polymerization. Prominent among these compounds is a benzosuberene analogue (referred to as KGP18), which demonstrates sub-nM cytotoxicity against human cancer cell lines and functions (when administered as a water-soluble prodrug salt) as a VDA in mouse models. Structure activity relationship considerations led to the evaluation of benzocyclooctyl [6,8 fused] and indene [6,5 fused] ring systems. Four benzocyclooctene and four indene analogues were prepared and evaluated biologically. Three of the benzocyclooctene analogues were active as inhibitors of tubulin polymerization (IC50 < 5 μM), and benzocyclooctene phenol 23 was comparable to KGP18 in terms of potency. The analogous indene-based compound 31 also functioned as an inhibitor of tubulin polymerization (IC50 = 11 μM) with reduced potency. The most potent inhibitor of tubulin polymerization from this group was benzocyclooctene analogue 23, and it was converted to its water-soluble prodrug salt 24 to assess its potential as a VDA. Preliminary in vivo studies, which utilized the MCF7-luc-GFP-mCherry breast tumor in a SCID mouse model, demonstrated that treatment with 24 (120 mg/kg) resulted in significant vascular shutdown, as evidenced by bioluminescence imaging at 4 h post administration, and that the effect continued at both 24 and 48 h. Contemporaneous studies with CA4P, a clinically relevant VDA, were carried out as a positive control.
Collapse
Affiliation(s)
- Christine A Herdman
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Tracy E Strecker
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Rajendra P Tanpure
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Zhi Chen
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Alex Winters
- Prognostic Imaging Research Laboratory, Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9058, United States
| | - Jeni Gerberich
- Prognostic Imaging Research Laboratory, Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9058, United States
| | - Li Liu
- Prognostic Imaging Research Laboratory, Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9058, United States
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Ralph P Mason
- Prognostic Imaging Research Laboratory, Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9058, United States
| | - David J Chaplin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States; Mateon Therapeutics, Inc., 701 Gateway Boulevard, Suite 210, South San Francisco, California 94080, United States
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| |
Collapse
|
98
|
Bukhvalova SY, Ivanov MA, Malysheva YB, Fedorov AY. Synthesis of polymethoxy-substituted triazolobenzoxazepines. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2016. [DOI: 10.1134/s1070428016100183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
99
|
Quantitative Evaluation of Tumor Early Response to a Vascular-Disrupting Agent with Dynamic PET. Mol Imaging Biol 2016; 17:865-73. [PMID: 25896816 DOI: 10.1007/s11307-015-0854-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE The purpose of this study is to evaluate the early response of tumors to a vascular-disrupting agent (VDA) VEGF121/recombinant toxin gelonin (rGel) using dynamic [(18)F]FPPRGD2 positron emission tomography (PET) and kinetic parameter estimation. PROCEDURES Two tumor xenograft models: U87MG (highly vascularized) and A549 (moderately vascularized), were selected, and both were randomized into treatment and control groups. Sixty-minute dynamic PET scans with [(18)F]FPPRGD2 that targets to integrin αvβ3 were performed at days 0 (baseline), 1, and 3 since VEGF121/rGel treatment started. Dynamic PET-derived binding potential (BPND) and parametric maps were compared with tumor uptake (%ID/g) and the static PET image at 1 h after the tracer administration. RESULTS The growth of U87MG tumor was obviously delayed upon VEGF121/rGel treatment. A549 tumor was not responsive to the same treatment. BPND of treated U87MG tumors decreased significantly at day 1 (p < 0.05), and the difference was more significant at day 3 (p < 0.01), compared with the control group. However, the tracer uptake (%ID/g) derived from static images at 1-h time point did not show significant difference between the treated and control tumors until day 3. Little difference in tracer uptake (%ID/g) or BPND was found between treated and control A549 tumors. Considering the tracer retention in tumor and the slower clearance due to damaged tumor vasculature after treatment, BPND representing the actual specific binding portion appears to be more sensitive and accurate than the semiquantitative parameters (such as %ID/g) derived from static images to assess the early response of tumor to VDA treatment. CONCLUSIONS Quantitative analysis based on dynamic PET with [(18)F]FPPRGD2 shows advantages in distinguishing effective from ineffective treatment during the course of VEGF121/rGel therapy at early stage and is therefore more sensitive in assessing therapy response than static PET.
Collapse
|
100
|
3D microtumors in vitro supported by perfused vascular networks. Sci Rep 2016; 6:31589. [PMID: 27549930 PMCID: PMC4994029 DOI: 10.1038/srep31589] [Citation(s) in RCA: 268] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/19/2016] [Indexed: 12/18/2022] Open
Abstract
There is a growing interest in developing microphysiological systems that can be used to model both normal and pathological human organs in vitro. This "organs-on-chips" approach aims to capture key structural and physiological characteristics of the target tissue. Here we describe in vitro vascularized microtumors (VMTs). This "tumor-on-a-chip" platform incorporates human tumor and stromal cells that grow in a 3D extracellular matrix and that depend for survival on nutrient delivery through living, perfused microvessels. Both colorectal and breast cancer cells grow vigorously in the platform and respond to standard-of-care therapies, showing reduced growth and/or regression. Vascular-targeting agents with different mechanisms of action can also be distinguished, and we find that drugs targeting only VEGFRs (Apatinib and Vandetanib) are not effective, whereas drugs that target VEGFRs, PDGFR and Tie2 (Linifanib and Cabozantinib) do regress the vasculature. Tumors in the VMT show strong metabolic heterogeneity when imaged using NADH Fluorescent Lifetime Imaging Microscopy and, compared to their surrounding stroma, many show a higher free/bound NADH ratio consistent with their known preference for aerobic glycolysis. The VMT platform provides a unique model for studying vascularized solid tumors in vitro.
Collapse
|