1
|
Dragulska SA, Poursharifi M, Chen Y, Wlodarczyk MT, Acosta Santiago M, Dottino P, Martignetti JA, Mieszawska AJ. Engineering and Validation of a Peptide-Stabilized Poly(lactic- co-glycolic) Acid Nanoparticle for Targeted Delivery of a Vascular Disruptive Agent in Cancer Therapy. Bioconjug Chem 2022; 33:2348-2360. [PMID: 36367382 DOI: 10.1021/acs.bioconjchem.2c00418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Developing a biocompatible and biodegradable nanoparticle (NP) carrier that integrates drug-loading capability, active targeting, and imaging modality is extremely challenging. Herein, we report an NP with a core of poly(lactic-co-glycolic) acid (PLGA) chemically modified with the drug combretastatin A4 (CA4), a vascular disrupting agent (VDA) in clinical development for ovarian cancer (OvCA) therapy. The NP is stabilized with a short arginine-glycine-aspartic acid-phenylalanine x3 (RGDFFF) peptide via self-assembly of the peptide on the PLGA surface. Importantly, the use of our RGDFFF coating replaces the commonly used polyethylene glycol (PEG) polymer that itself often induces an unwanted immunogenic response. In addition, the RGD motif of the peptide is well-known to preferentially bind to αvβ3 integrin that is implicated in tumor angiogenesis and is exploited as the NP's targeting component. The NP is enhanced with an optical imaging fluorophore label via chemical modification of the PLGA. The RGDFFF-CA4 NPs are synthesized using a nanoprecipitation method and are ∼75 ± 3.7 nm in diameter, where a peptide coating comprises a 2-3 nm outer layer. The NPs are serum stable for 72 h. In vitro studies using human umbilical cord vascular endothelial cells (HUVEC) confirmed the high uptake and biological activity of the RGDFFF-CA4 NP. NP uptake and viability reduction were demonstrated in OvCA cells grown in culture, and the NPs efficiently accumulated in tumors in a preclinical OvCA mouse model. The RGDFFF NP did not induce an inflammatory response when cultured with immune cells. Finally, the NP was efficiently taken up by patient-derived OvCA cells, suggesting a potential for future clinical applications.
Collapse
Affiliation(s)
- Sylwia A Dragulska
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York11210, United States
| | - Mina Poursharifi
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York11210, United States
| | - Ying Chen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, New York10029, United States
| | - Marek T Wlodarczyk
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York11210, United States
| | - Maxier Acosta Santiago
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York11210, United States
| | - Peter Dottino
- Department of Obstetrics/Gynecology & Reproductive Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, New York10029, United States
| | - John A Martignetti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, New York10029, United States.,Women's Health Research Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl., New York, New York10029, United States.,Rudy Ruggles Research Institute, Western Connecticut Health Network, 131 West St., Danbury, Connecticut06810, United States
| | - Aneta J Mieszawska
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York11210, United States
| |
Collapse
|
2
|
Mampaey G, Hellemans A, de Rooster H, Schipper T, Abma E, Broeckx BJG, Daminet S, Smets P. Assessment of Cardiotoxicity after a Single Dose of Combretastatin A4-Phosphate in Dogs Using Two-Dimensional Speckle-Tracking Echocardiography. Animals (Basel) 2022; 12:ani12213005. [PMID: 36359129 PMCID: PMC9658292 DOI: 10.3390/ani12213005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/21/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Simple Summary Combretastatin A4-phosphate is a chemotherapeutic drug which has been evaluated for treatment of solid canine tumors. Previous studies reported cardiotoxic effects based on changes in cardiac troponin I measurements, blood pressure, and electrocardiography. We evaluated the cardiotoxic effect by two-dimensional speckle tracking echocardiography. This advanced imaging technique analyzes global and regional myocardial function and is used as the gold-standard for the assessment of cardiac function in human patients receiving chemotherapy. We found that certain strain measurements were significantly decreased 24 h after the administration of combretastatin A4-phosphate and that these changes were correlated with an increase in cardiac troponin I. Our results suggest that two-dimensional speckle tracking may be useful for the early detection of cardiac dysfunction in canine cancer patients as well as promising during follow-up. Abstract Combretastatin A4-phosphate (CA4P) is a vascular disrupting agent that was recently described for the treatment of solid canine tumors. Conventional echocardiography and pulsed wave tissue Doppler imaging did not reveal cardiotoxicity in dogs, however, the gold standard for assessing myocardial damage in humans receiving cardiotoxic chemotherapeutics is two-dimensional speckle-tracking echocardiography. The current study evaluated the cardiotoxic effect of a single dose of CA4P in dogs using peak systolic strain measurements and the variability of these measurements. Echocardiographic examinations of seven healthy beagles and five canine cancer patients that received CA4P were retrospectively reviewed. Peak systolic regional longitudinal strain (LSt), peak systolic regional circumferential strain (CSt), and peak systolic regional radial strain (RSt) were measured before and 24 h after administration of CA4P. Peak systolic strain measurements were compared to serum cardiac troponin I (cTnI). To quantify intra- and inter-observer measurement variability, seven echocardiographic examinations were selected and each strain parameter was measured by three observers on three consecutive days. After CA4P administration, the median LSt and CSt values decreased by 21.8% (p = 0.0005) and 12.3% (p = 0.002), respectively, whereas the median RSt values were not significantly different (p = 0.70). The decrease in LSt was correlated with increased serum cTnI values (Spearman rho = −0.64, p = 0.02). The intra-observer coefficients of variation (CV) were 9%, 4%, and 13% for LSt, CSt, and RSt, respectively, while the corresponding interobserver CVs were 11%, 12%, and 20%. Our results suggest that regional peak systolic strain measurements may be useful for the early detection of cardiotoxicity that is caused by vascular disrupting agents and that LSt may be promising for the follow-up of canine cancer patients.
Collapse
Affiliation(s)
- Gitte Mampaey
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
- Correspondence:
| | - Arnaut Hellemans
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Hilde de Rooster
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Tom Schipper
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Eline Abma
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Bart J. G. Broeckx
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Sylvie Daminet
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Pascale Smets
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| |
Collapse
|
3
|
Cao Y, Ahmed AMQ, Du HH, Sun W, Lu X, Xu Z, Tao J, Cao QR. Combretastatin A4-loaded Poly (Lactic-co-glycolic Acid)/Soybean Lecithin Nanoparticles with Enhanced Drug Dissolution Rate and Antiproliferation Activity. Curr Drug Deliv 2022; 19:918-927. [PMID: 35139789 DOI: 10.2174/1567201819666220209093443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/24/2021] [Accepted: 12/12/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study aimed to prepare combretastatin A4 (CA4)-loaded nanoparticles (CA4 NPs) using poly(lactic-co-glycolic acid) (PLGA) and soybean lecithin (Lipoid S100) as carriers, and further evaluate the physicochemical properties and cytotoxicities of CA4 NPs against cancer cells. METHODS CA4 NPs were prepared using a solvent evaporation technique. The effects of formulations on CA4 NPs were investigated in terms of particle size, zeta potential, encapsulation efficacy, and drug loading. The physicochemical properties of CA4 NPs were characterized using transmission electron microscopy, X-ray powder diffraction, differential scanning calorimetry, and Fourier transform infrared spectra. The drug release from CA4NPs was performed using a dialysis method. In addition, the cytotoxicity of CA4NPs against human alveolar basal epithelial (A549) cells was also evaluated. RESULTS CA4 NPs prepared with a low organic/water phase ratio (1:20) and high drug/PLGA mass ratio (1:2.5) exhibited a uniform hydrodynamic particle size of 142 nm, the zeta potential of -1.66 mV, and encapsulation efficacy and drug loading of 92.1% and 28.3%, respectively. CA4 NPs showed a significantly higher release rate than pure CA4 in pH 7.4 phosphate-buffered solution with 0.5% Tween 80. It was found that the drug molecules could change from the crystal state to an amorphous form when loaded into the PLGA/Lipoid S100 matrix, and some molecular interactions could also occur between the drug and PLGA. Importantly, CA4 NPs showed a remarkably higher antiproliferation activity against A549 cancer cells compared to pure CA4. CONCLUSION These results suggested the promising potential of PLGA/Lipoid S100 nanoparticles as the drug delivery system of CA4 for effective cancer therapy.
Collapse
Affiliation(s)
- Yue Cao
- Department of Pharmacy, Beijing Health Vocational College, Beijing 100053, People's Republic of China
| | | | - Huan-Huan Du
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Wei Sun
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Xiaojuan Lu
- PharmaMax Pharmaceuticals, Ltd., China Medical City, Taizhou 225300, People's Republic of China
| | - Zhao Xu
- PharmaMax Pharmaceuticals, Ltd., China Medical City, Taizhou 225300, People's Republic of China
| | - Jing Tao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Qing-Ri Cao
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| |
Collapse
|
4
|
Wu Q, Wang Q, Wang Y, Huang J, Fang Y, Wu W, Wu W, Wu F, Yu X, Sun Y. A high-throughput and simultaneous determination of combretastatin A-4 phosphate and its metabolites in human plasma using HPLC-MS/MS: Application to a clinical pharmacokinetic study. Biomed Chromatogr 2021; 35:e5204. [PMID: 34165810 DOI: 10.1002/bmc.5204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 11/06/2022]
Abstract
To investigate the clinical pharmacokinetics of CA4P, a high-throughput high-performance liquid chromatography-tandem mass spectrometry assay with an identical positive electrospray ionization (ESI) mode was developed for the simultaneous determination of CA4P, its active metabolite CA4, and CA4 glucuronide in human plasma. CA4P and CA4 were easier to protonate in positive ESI mode, whereas CA4G was reported to produce deprotonated ion in negative ESI mode. Because the baseline separation of CA4P and CA4G could not be achieved, using MS positive/negative ion switching is not feasible. In this study, an abundant ammonium adduct ion of CA4G in ESI+ was observed as an ideal precursor ion. The final precursor/product transition pairs chosen for CA4P, CA4, and CA4G were at m/z 397/350, 317/286, and 510/317, respectively. To the best of our knowledge, it is the first report on the simultaneous quantification of CA4P, CA4, and CA4G in biological samples. The proposed method was validated, which showed a wide linear dynamic range, high selectivity and sensitivity, good repeatability, and a short run time. Compared with the literatures, the lower limits of quantification were five- and two-fold more sensitive for CA4G and CA4, respectively. Therefore, this method was successfully applied to the pharmacokinetic study of CA4P in phase I clinical trial.
Collapse
Affiliation(s)
- Qizhen Wu
- Laboratory of Phase I Clinical Trials, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qian Wang
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yixuan Wang
- Laboratory of Phase I Clinical Trials, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jingqiu Huang
- Laboratory of Phase I Clinical Trials, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yalin Fang
- Laboratory of Phase I Clinical Trials, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Weiyi Wu
- Laboratory of Phase I Clinical Trials, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wenying Wu
- Laboratory of Phase I Clinical Trials, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fanhong Wu
- Institute of Pharmaceutical Innovation, Shanghai Institute of Technology, Shanghai Engineering Research Center of Green Fluoro Pharmaceutical Technology, Shanghai, China
| | - Xiaodong Yu
- Shanghai Ecust Biomedicine Company Limited, Shanghai, China
| | - Yan Sun
- Laboratory of Phase I Clinical Trials, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Shiah HS, Chiang NJ, Lin CC, Yen CJ, Tsai HJ, Wu SY, Su WC, Chang KY, Wang CC, Chang JY, Chen LT. Phase I Dose-Escalation Study of SCB01A, a Microtubule Inhibitor with Vascular Disrupting Activity, in Patients with Advanced Solid Tumors. Oncologist 2020; 26:e567-e579. [PMID: 33245172 DOI: 10.1002/onco.13612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 11/15/2020] [Indexed: 11/10/2022] Open
Abstract
LESSONS LEARNED SCB01A is a novel microtubule inhibitor with vascular disrupting activity. This first-in-human study demonstrated SCB01A safety, pharmacokinetics, and preliminary antitumor activity. SCB01A is safe and well tolerated in patients with advanced solid malignancies with manageable neurotoxicity. BACKGROUND SCB01A, a novel microtubule inhibitor, has vascular disrupting activity. METHODS In this phase I dose-escalation and extension study, patients with advanced solid tumors were administered intravenous SCB01A infusions for 3 hours once every 21 days. Rapid titration and a 3 + 3 design escalated the dose from 2 mg/m2 to the maximum tolerated dose (MTD) based on dose-limiting toxicity (DLT). SCB01A-induced cellular neurotoxicity was evaluated in dorsal root ganglion cells. The primary endpoint was MTD. Safety, pharmacokinetics (PK), and tumor response were secondary endpoints. RESULTS Treatment-related adverse events included anemia, nausea, vomiting, fatigue, fever, and peripheral sensorimotor neuropathy. DLTs included grade 4 elevated creatine phosphokinase (CPK) in the 4 mg/m2 cohort; grade 3 gastric hemorrhage in the 6.5 mg/m2 cohort; grade 2 thromboembolic event in the 24 mg/m2 cohort; and grade 3 peripheral sensorimotor neuropathy, grade 3 elevated aspartate aminotransferase, and grade 3 hypertension in the 32 mg/m2 cohort. The MTD was 24 mg/m2 , and average half-life was ~2.5 hours. The area under the curve-dose response relationship was linear. Nineteen subjects were stable after two cycles. The longest treatment lasted 24 cycles. SCB01A-induced neurotoxicity was reversible in vitro. CONCLUSION The MTD of SCB01A was 24 mg/m2 every 21 days; it is safe and tolerable in patients with solid tumors.
Collapse
Affiliation(s)
- Her-Shyong Shiah
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Division of Hematology and Oncology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Nai-Jung Chiang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Jui Yen
- Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Jen Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shang-Yin Wu
- Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu-Chou Su
- Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | - Jang-Yang Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Oncology-Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, Kaohisung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
6
|
Tang Z, Xiong D, Song J, Ye M, Liu J, Wang Z, Zhang L, Xiao X. Antitumor Drug Combretastatin-A4 Phosphate Aggravates the Symptoms of Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice. Front Pharmacol 2020; 11:339. [PMID: 32265711 PMCID: PMC7106770 DOI: 10.3389/fphar.2020.00339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/06/2020] [Indexed: 12/17/2022] Open
Abstract
Ulcerative colitis (UC) is an idiopathic inflammatory bowel disease (IBD) that causes long-lasting inflammation and ulcers in the innermost lining of the colon and rectum. Previous studies demonstrated that resveratrol suppresses colitis and colon cancer associated with colitis by improving glucose metabolism, but resveratrol use is limited by its low oral bioavailability. Combretastatin-A4 phosphate (CA4P) is a vascular-disrupting agent with antitumor activity. CA4P is structurally similar to resveratrol, but whether CA4P has the same effect as resveratrol on UC is not clear. In this study, we examined the pharmacological effects of CA4P administration on dextran sulfate sodium (DSS)-induced inflammation in a mouse model of UC. C57BL/6 mice were administered 2.5% DSS in the drinking water to induce acute UC. CA4P (11 mg/kg/d) was injected intraperitoneally daily. The Disease Activity Index (DAI) score and histological score were evaluated to determine the severity of UC. Colon tissues and blood samples were collected for histological analyses. The results show that CA4P plus DSS significantly decreased colon length (P < 0.05 versus DSS+PBS group) and body weight (P < 0.001 versus PBS group), while increased spleen weight (P < 0.01 versus DSS+PBS group), DAI score (P < 0.01 versus DSS+PBS group), and histological score (P < 0.01 versus DSS+PBS group). Moreover, CA4P exacerbated the pathological features of colitis and significantly increased proinflammatory cytokines (IL-1β, IL-6, TNF-α) and inflammatory cells (neutrophil, lymphocyte, monocyte). These findings reveal that CA4P aggravates the symptoms of DSS-induced UC and provide a key reference for the potential of CA4P as an anticancer drug.
Collapse
Affiliation(s)
- Zhengshan Tang
- Hepatobiliary and Enteric Surgery Research Center, Xiangya Hospital, Central South University, Changsha, China
| | - Dehui Xiong
- School of Life Sciences & Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Jianhui Song
- School of Life Sciences & Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Jing Liu
- School of Life Sciences & Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Zi Wang
- Hepatobiliary and Enteric Surgery Research Center, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Zhang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Xiaojuan Xiao
- School of Life Sciences & Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| |
Collapse
|
7
|
Nowikow C, Fuerst R, Kauderer M, Dank C, Schmid W, Hajduch M, Rehulka J, Gurska S, Mokshyna O, Polishchuk P, Zupkó I, Dzubak P, Rinner U. Synthesis and biological evaluation of cis-restrained carbocyclic combretastatin A-4 analogs: Influence of the ring size and saturation on cytotoxic properties. Bioorg Med Chem 2019; 27:115032. [DOI: 10.1016/j.bmc.2019.07.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/19/2019] [Accepted: 07/28/2019] [Indexed: 10/26/2022]
|
8
|
Gill JH, Rockley KL, De Santis C, Mohamed AK. Vascular Disrupting Agents in cancer treatment: Cardiovascular toxicity and implications for co-administration with other cancer chemotherapeutics. Pharmacol Ther 2019; 202:18-31. [PMID: 31173840 DOI: 10.1016/j.pharmthera.2019.06.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 05/30/2019] [Indexed: 02/08/2023]
Abstract
Destruction of the established tumour vasculature by a class of compound termed Vascular Disrupting Agents (VDAs) is showing considerable promise as a viable approach for the management of solid tumours. VDAs induce a rapid shutdown and collapse of tumour blood vessels, leading to ischaemia and consequent necrosis of the tumour mass. Their efficacy is hindered by the persistence of a viable rim of tumour cells, supported by the peripheral normal vasculature, necessitating their co-administration with additional chemotherapeutics for maximal therapeutic benefit. However, a major limitation for the use of many cancer therapeutics is the development of life-threatening cardiovascular toxicities, with significant consequences for treatment response and the patient's quality of life. The aim of this review is to outline VDAs as a cancer therapeutic approach and define the mechanistic basis of cardiovascular toxicities of current chemotherapeutics, with the overall objective of discussing whether VDA combinations with specific chemotherapeutic classes would be good or bad in terms of cardiovascular toxicity.
Collapse
Affiliation(s)
- Jason H Gill
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK; School of Pharmacy, Faculty of Medical Sciences, Newcastle University, UK.
| | - Kimberly L Rockley
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK
| | - Carol De Santis
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK
| | - Asma K Mohamed
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK
| |
Collapse
|
9
|
Izumi Y, Takagi S. Vascular disrupting effect of combretastatin A-4 phosphate with inhibition of vascular endothelial cadherin in canine osteosarcoma-xenografted mice. Res Vet Sci 2019; 122:1-6. [DOI: 10.1016/j.rvsc.2018.10.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 11/24/2022]
|
10
|
Yu S, Wei S, Liu L, Qi D, Wang J, Chen G, He W, He C, Chen X, Gu Z. Enhanced local cancer therapy using a CA4P and CDDP co-loaded polypeptide gel depot. Biomater Sci 2019; 7:860-866. [DOI: 10.1039/c8bm01442f] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A CA4P and CDDP co-loaded polypeptide gel depot was prepared for enhanced local colon cancer treatment.
Collapse
|
11
|
Zhang C, Zhang X, Wang G, Peng Y, Zhang X, Wu H, Yu B, Sun J. Preclinical Pharmacokinetics of C118P, a Novel Prodrug of Microtubules Inhibitor and Its Metabolite C118 in Mice, Rats, and Dogs. Molecules 2018; 23:molecules23112883. [PMID: 30400617 PMCID: PMC6278385 DOI: 10.3390/molecules23112883] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/26/2018] [Accepted: 11/01/2018] [Indexed: 01/09/2023] Open
Abstract
C118P, a phosphate prodrug of C118, which is a novel microtubule protein inhibitor, is currently under Phase I clinical development in China for treating ovarian cancer and lung cancer. The preclinical pharmacokinetics of prodrug C118P and its metabolite C118 were extensively characterized in vivo in mice, rats, and dogs and in vitro to support the further development of C118P. The preclinical tissue distribution and excretion were investigated in rats. Plasma protein binding in mice, rat, and human, and hepatic microsomal metabolic stability in mice, rat, dog, monkey, and human, were also evaluated. The (AUC0-inf) and C30s of C118P at 50 mg/kg in rats and 6 mg/kg in dogs, and the C2min of C118 at 6 mg/kg in dogs increased less than the dosage increase, suggested nonlinear pharmacokinetic occurred at high dose. As a prodrug, C118P can be quickly hydrolyzed into C118 after an intravenous administration. The unbound C118 in plasma is slightly higher than C118P. C118P can hardly penetrate the tissue, while C118 can distribute widely into tissues. In tumor-bearing nude mice, the concentration of C118 is high in lung, ovary, and tumor, with an extended half-life in tumor. C118P is a promising candidate prodrug for further clinical development.
Collapse
Affiliation(s)
- Cang Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
- Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 211135, China.
| | - Xiaolan Zhang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Guangji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Ying Peng
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Xueyuan Zhang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Hui Wu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Boyang Yu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Jianguo Sun
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
12
|
Liu YW, De Keyzer F, Feng YB, Chen F, Song SL, Swinnen J, Bormans G, Oyen R, Huang G, Ni YC. Intra-individual comparison of therapeutic responses to vascular disrupting agent CA4P between rodent primary and secondary liver cancers. World J Gastroenterol 2018; 24:2710-2721. [PMID: 29991876 PMCID: PMC6034151 DOI: 10.3748/wjg.v24.i25.2710] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/01/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To compare therapeutic responses of a vascular-disrupting-agent, combretastatin-A4-phosphate (CA4P), among hepatocellular carcinomas (HCCs) and implanted rhabdomyosarcoma (R1) in the same rats by magnetic-resonance-imaging (MRI), microangiography and histopathology.
METHODS Thirty-six HCCs were created by diethylnitrosamine gavage in 14 rats that were also intrahepatically implanted with one R1 per rat as monitored by T2-/T1-weighted images (T2WI/T1WI) on a 3.0T clinical MRI-scanner. Vascular response and tumoral necrosis were detected by dynamic contrast-enhanced (DCE-) and CE-MRI before, 1 h after and 12 h after CA4P iv at 10 mg/kg (treatment group n = 7) or phosphate-buffered saline at 1.0 mL/kg (control group n = 7). Tumor blood supply was calculated by a semiquantitative DCE parameter of area under the time signal intensity curve (AUC30). In vivo MRI findings were verified by postmortem techniques.
RESULTS On CE-T1WIs, unlike the negative response in all tumors of control animals, in treatment group CA4P caused rapid extensive vascular shutdown in all R1-tumors, but mildly or spottily in HCCs at 1 h. Consequently, tumor necrosis occurred massively in R1-tumors but patchily in HCCs at 12 h. AUC30 revealed vascular closure (66%) in R1-tumors at 1 h (P < 0.05), followed by further perfusion decrease at 12 h (P < 0.01), while less significant vascular clogging occurred in HCCs. Histomorphologically, CA4P induced more extensive necrosis in R1-tumors (92.6%) than in HCCs (50.2%) (P < 0.01); tumor vascularity heterogeneously scored +~+++ in HCCs but homogeneously scored ++ in R1-tumors.
CONCLUSION This study suggests superior performance of CA4P in metastatic over primary liver cancers, which could guide future clinical applications of vascular-disrupting-agents.
Collapse
MESH Headings
- Angiography
- Animals
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/therapeutic use
- Carcinoma, Hepatocellular/blood supply
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Contrast Media/administration & dosage
- Diethylnitrosamine/toxicity
- Humans
- Liver/diagnostic imaging
- Liver/pathology
- Liver Neoplasms/blood supply
- Liver Neoplasms/chemically induced
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Liver Neoplasms, Experimental/chemically induced
- Liver Neoplasms, Experimental/diagnostic imaging
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/pathology
- Magnetic Resonance Imaging/methods
- Male
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/pathology
- Rats
- Rhabdomyosarcoma/blood supply
- Rhabdomyosarcoma/drug therapy
- Rhabdomyosarcoma/pathology
- Rhabdomyosarcoma/secondary
- Stilbenes/pharmacology
- Stilbenes/therapeutic use
- Treatment Outcome
- Tumor Microenvironment/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Ye-Wei Liu
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
- Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, China
| | | | - Yuan-Bo Feng
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Feng Chen
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Shao-Li Song
- Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Johan Swinnen
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Guy Bormans
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Raymond Oyen
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Gang Huang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, China
| | - Yi-Cheng Ni
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
13
|
El Bairi K, Amrani M, Afqir S. Starvation tactics using natural compounds for advanced cancers: pharmacodynamics, clinical efficacy, and predictive biomarkers. Cancer Med 2018; 7:2221-2246. [PMID: 29732738 PMCID: PMC6010871 DOI: 10.1002/cam4.1467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/21/2018] [Accepted: 02/28/2018] [Indexed: 02/05/2023] Open
Abstract
The high mortality associated with oncological diseases is mostly due to tumors in advanced stages, and their management is a major challenge in modern oncology. Angiogenesis is a defined hallmark of cancer and predisposes to metastatic invasion and dissemination and is therefore an important druggable target for cancer drug discovery. Recently, because of drug resistance and poor prognosis, new anticancer drugs from natural sources targeting tumor vessels have attracted more attention and have been used in several randomized and controlled clinical trials as therapeutic options. Here, we outline and discuss potential natural compounds as salvage treatment for advanced cancers from recent and ongoing clinical trials and real-world studies. We also discuss predictive biomarkers for patients' selection to optimize the use of these potential anticancer drugs.
Collapse
Affiliation(s)
- Khalid El Bairi
- Faculty of Medicine and PharmacyMohamed Ist UniversityOujdaMorocco
| | - Mariam Amrani
- Equipe de Recherche en Virologie et Onco‐biologieFaculty of MedicinePathology DepartmentNational Institute of OncologyUniversité Mohamed VRabatMorocco
| | - Said Afqir
- Department of Medical OncologyMohamed VI University HospitalOujdaMorocco
| |
Collapse
|
14
|
Porta-Sánchez A, Gilbert C, Spears D, Amir E, Chan J, Nanthakumar K, Thavendiranathan P. Incidence, Diagnosis, and Management of QT Prolongation Induced by Cancer Therapies: A Systematic Review. J Am Heart Assoc 2017; 6:JAHA.117.007724. [PMID: 29217664 PMCID: PMC5779062 DOI: 10.1161/jaha.117.007724] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background The cardiovascular complications of cancer therapeutics are the focus of the burgeoning field of cardio‐oncology. A common challenge in this field is the impact of cancer drugs on cardiac repolarization (ie, QT prolongation) and the potential risk for the life‐threatening arrhythmia torsades de pointes. Although QT prolongation is not a perfect marker of arrhythmia risk, this has become a primary safety metric among oncologists. Cardiologists caring for patients receiving cancer treatment should become familiar with the drugs associated with QT prolongation, its incidence, and appropriate management strategies to provide meaningful consultation in this complex clinical scenario. Methods and Results In this article, we performed a systematic review (using Preferred Reporting Items of Systematic Reviews and Meta‐Analyses (PRISMA) guidelines) of commonly used cancer drugs to determine the incidence of QT prolongation and clinically relevant arrhythmias. We calculated summary estimates of the incidence of all and clinically relevant QT prolongation as well as arrhythmias and sudden cardiac death. We then describe strategies to prevent, identify, and manage QT prolongation in patients receiving cancer therapy. We identified a total of 173 relevant publications. The weighted incidence of any corrected QT (QTc) prolongation in our systematic review in patients treated with conventional therapies (eg, anthracyclines) ranged from 0% to 22%, although QTc >500 ms, arrhythmias, or sudden cardiac death was extremely rare. The risk of QTc prolongation with targeted therapies (eg, small molecular tyrosine kinase inhibitors) ranged between 0% and 22.7% with severe prolongation (QTc >500 ms) reported in 0% to 5.2% of the patients. Arrhythmias and sudden cardiac death were rare. Conclusions Our systematic review demonstrates that there is variability in the incidence of QTc prolongation of various cancer drugs; however, the clinical consequence, as defined by arrhythmias or sudden cardiac death, remains rare.
Collapse
Affiliation(s)
- Andreu Porta-Sánchez
- Division of Cardiology, Department of Electrophysiology, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Cameron Gilbert
- Division of Cardiology, Department of Electrophysiology, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Danna Spears
- Division of Cardiology, Department of Electrophysiology, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Eitan Amir
- Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Joyce Chan
- Department of Pharmacy, University Health Network, Toronto, Ontario, Canada
| | - Kumaraswamy Nanthakumar
- Division of Cardiology, Department of Electrophysiology, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Paaladinesh Thavendiranathan
- Division of Cardiology, Peter Munk Cardiac Centre, Ted Rogers Program in Cardiotoxicity Prevention and Department of Medical Imaging, University Health Network University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Abstract
FtsZ, a homolog of tubulin, is found in almost all bacteria and archaea where it has a primary role in cytokinesis. Evidence for structural homology between FtsZ and tubulin came from their crystal structures and identification of the GTP box. Tubulin and FtsZ constitute a distinct family of GTPases and show striking similarities in many of their polymerization properties. The differences between them, more so, the complexities of microtubule dynamic behavior in comparison to that of FtsZ, indicate that the evolution to tubulin is attributable to the incorporation of the complex functionalities in higher organisms. FtsZ and microtubules function as polymers in cell division but their roles differ in the division process. The structural and partial functional homology has made the study of their dynamic properties more interesting. In this review, we focus on the application of the information derived from studies on FtsZ dynamics to study microtubule dynamics and vice versa. The structural and functional aspects that led to the establishment of the homology between the two proteins are explained to emphasize the network of FtsZ and microtubule studies and how they are connected.
Collapse
Affiliation(s)
- Rachana Rao Battaje
- Department of Biosciences and BioengineeringIndian Institute of Technology Bombay, Mumbai, India
| | - Dulal Panda
- Department of Biosciences and BioengineeringIndian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
16
|
Jaroch K, Karolak M, Górski P, Jaroch A, Krajewski A, Ilnicka A, Sloderbach A, Stefański T, Sobiak S. Combretastatins: In vitro structure-activity relationship, mode of action and current clinical status. Pharmacol Rep 2016; 68:1266-1275. [PMID: 27686966 DOI: 10.1016/j.pharep.2016.08.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/18/2016] [Accepted: 08/23/2016] [Indexed: 02/08/2023]
Abstract
For the first time combretastatins were isolated from African willow tree Combretum Caffrum. Subsequent studies have shown the impact of combretastatin A4 phosphate, a water-soluble prodrug, on endothelial cells in tumor vascular system. The same effect was not observed in the vascular system. This selectivity is associated with combretastatins mechanism of action: binding to colchicine domain of microtubules, which affects the cytoskeleton functionality of immature endothelial cells. At the same time, combretastatins directly induce cell death via apoptosis and/or mitotic catastrophe pathways. The combination of both elements makes combretastatin an anticancer compound of high efficiency. The cis-configuration is crucial for its biological activity. To date, many derivatives were synthesized. The attempts to resolve spontaneous isomerization to less active trans-stilbene derivative are still in progress. This issue seems to be overcome by incorporation of the ethene bridge with heterocyclic moiety in combretastatins structure. This modification retains the cis-configuration and prevents isomerization. Nevertheless, combretastatin A4 phosphate disodium is still the most potent compound of this group. The combination therapy, which is the most effective treatment, includes combretastatin A4 phosphate (CA4P) and conventional chemotherapeutics and/or radiotherapy. CA4P is relatively well tolerated giving adverse events of moderate severity, which includes: nausea, vomiting, headache, and tumor pain. The aforementioned effects subside on the day of drug administration or on the following day.
Collapse
Affiliation(s)
- Karol Jaroch
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland.
| | - Maciej Karolak
- Department of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Przemysław Górski
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Alina Jaroch
- Department and Institute of Nutrition and Dietetics, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland; Department and Clinic of Geriatrics, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Adrian Krajewski
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | | | - Anna Sloderbach
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Tomasz Stefański
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Poznan University of Medical Sciences, Poznań, Poland; Department of Crystallography, Faculty of Chemistry, Adam Mickiewicz University, Poznań, Poland
| | - Stanisław Sobiak
- Department of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| |
Collapse
|
17
|
Kalmuk J, Folaron M, Buchinger J, Pili R, Seshadri M. Multimodal imaging guided preclinical trials of vascular targeting in prostate cancer. Oncotarget 2016. [PMID: 26203773 PMCID: PMC4695192 DOI: 10.18632/oncotarget.4463] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The high mortality rate associated with castration-resistant prostate cancer (CRPC) underscores the need for improving therapeutic options for this patient population. The purpose of this study was to examine the potential of vascular targeting in prostate cancer. Experimental studies were carried out in subcutaneous and orthotopic Myc-CaP prostate tumors implanted into male FVB mice to examine the efficacy of a novel microtubule targeted vascular disrupting agent (VDA), EPC2407 (Crolibulin™). A non-invasive multimodality imaging approach based on magnetic resonance imaging (MRI), bioluminescence imaging (BLI), and ultrasound (US) was utilized to guide preclinical trial design and monitor tumor response to therapy. Imaging results were correlated with histopathologic assessment, tumor growth and survival analysis. Contrast-enhanced MRI revealed potent antivascular activity of EPC2407 against subcutaneous and orthotopic Myc-CaP tumors. Longitudinal BLI of Myc-CaP tumors expressing luciferase under the androgen response element (Myc-CaP/ARE-luc) revealed changes in AR signaling and reduction in intratumoral delivery of luciferin substrate following castration suggestive of reduced blood flow. This reduction in blood flow was validated by US and MRI. Combination treatment resulted in sustained vascular suppression, inhibition of tumor regrowth and conferred a survival benefit in both models. These results demonstrate the therapeutic potential of vascular targeting in combination with androgen deprivation against prostate cancer.
Collapse
Affiliation(s)
- James Kalmuk
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Current address: SUNY Upstate Medical University, Syracuse, NY, USA
| | - Margaret Folaron
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Molecular and Cellular Biophysics and Biochemistry, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Julian Buchinger
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Current address: University at Buffalo - School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Roberto Pili
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Mukund Seshadri
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Molecular and Cellular Biophysics and Biochemistry, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
18
|
Jalili M, Mozaffarpour Noori A, Sedaghat M, Safaie A. Efficacy of Intravenous Paracetamol Versus Intravenous Morphine in Acute Limb Trauma. Trauma Mon 2016; 21:e19649. [PMID: 27218042 PMCID: PMC4869432 DOI: 10.5812/traumamon.19649] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 07/23/2014] [Accepted: 08/23/2014] [Indexed: 11/16/2022] Open
Abstract
Background: Efficient pain management is one of the most important components of care in the field of emergency medicine. Objectives: This study was conducted to compare intravenous paracetamol and intravenous morphine sulfate for acute pain reduction in patients with limb trauma. Patients and Methods: In a randomized double-blinded clinical trial, all patients (aged 18 years and older) with acute limb trauma and a pain score of greater than 3/10 in the emergency department were recruited; they received either 1 g intravenous paracetamol or 0.1 mg/kg intravenous morphine sulfate over 15 minutes. The primary outcome was the pain score measured on a numerical rating scale at 0, 15 and 30 minutes after commencing drug administration. The requirement for rescue analgesia and the frequency of adverse reactions were also recorded. Results: Sixty patients randomly received either IV paracetamol (n = 30) or IV morphine (n = 30). The mean reduction in numerical rating scale pain intensity scores at 30 minutes was 3.86 (± 1.61) for paracetamol, and 2.16 (± 1.39) for morphine. However, pain relief was significantly higher in the paracetamol group compared to the morphine group (P < 0.001). Four patients in the paracetamol group and 15 patients in the morphine group needed rescue analgesia and the difference was significant (P = 0.05). Conclusions: Intravenous paracetamol appears to provide better analgesia than intravenous morphine in acute limb trauma. Further larger studies are required.
Collapse
Affiliation(s)
- Mohammad Jalili
- Department of Emergency Medicine, Tehran University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Mohammad Jalili, Department of Emergency Medicine, Imam Hospital, Keshavarz Boulevard, Tehran University of Medical Sciences, Tehran, IR Iran. Tel: +98-9125483998, Fax: +98-2166904848, E-mail:
| | - Ali Mozaffarpour Noori
- Department of Emergency Medicine, Zahedan University of Medical Sciences, Zahedan, IR Iran
| | - Mojtaba Sedaghat
- Department of Community Medicine, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Arash Safaie
- Department of Emergency Medicine, Tehran University of Medical Sciences, Tehran, IR Iran
| |
Collapse
|
19
|
Greene LM, Meegan MJ, Zisterer DM. Combretastatins: more than just vascular targeting agents? J Pharmacol Exp Ther 2015; 355:212-27. [PMID: 26354991 DOI: 10.1124/jpet.115.226225] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/25/2015] [Indexed: 01/23/2023] Open
Abstract
Several prodrugs of the naturally occurring combretastatins have undergone extensive clinical evaluation as vascular targeting agents (VTAs). Their increased selectivity toward endothelial cells together with their innate ability to rapidly induce vascular shutdown and inhibit tumor growth at doses up to 10-fold less than the maximum tolerated dose led to the clinical evaluation of combretastatins as VTAs. Tubulin is well established as the molecular target of the combretastatins and the vast majority of its synthetic derivatives. Furthermore, tubulin is a highly validated molecular target of many direct anticancer agents routinely used as front-line chemotherapeutics. The unique vascular targeting properties of the combretastatins have somewhat overshadowed their development as direct anticancer agents and the delineation of the various cell death pathways and anticancer properties associated with such chemotherapeutics. Moreover, the ongoing clinical trial of OXi4503 (combretastatin-A1 diphosphate) together with preliminary preclinical evaluation for the treatment of refractory acute myelogenous leukemia has successfully highlighted both the indirect and direct anticancer properties of combretastatins. In this review, we discuss the development of the combretastatins from nature to the clinic. The various mechanisms underlying combretastatin-induced cell cycle arrest, mitotic catastrophe, cell death, and survival are also reviewed in an attempt to further enhance the clinical prospects of this unique class of VTAs.
Collapse
Affiliation(s)
- Lisa M Greene
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (L.M.G., D.M.Z.), and School of Pharmacy and Pharmaceutical Sciences, Centre for Synthesis and Chemical Biology (M.J.M.), Trinity College Dublin, Dublin, Ireland
| | - Mary J Meegan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (L.M.G., D.M.Z.), and School of Pharmacy and Pharmaceutical Sciences, Centre for Synthesis and Chemical Biology (M.J.M.), Trinity College Dublin, Dublin, Ireland
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (L.M.G., D.M.Z.), and School of Pharmacy and Pharmaceutical Sciences, Centre for Synthesis and Chemical Biology (M.J.M.), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
20
|
Voitovich YV, Shegravina ES, Sitnikov NS, Faerman VI, Fokin VV, Schmalz HG, Combes S, Allegro D, Barbier P, Beletskaya IP, Svirshchevskaya EV, Fedorov AY. Synthesis and Biological Evaluation of Furanoallocolchicinoids. J Med Chem 2014; 58:692-704. [DOI: 10.1021/jm501678w] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Yuliya V. Voitovich
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| | - Ekaterina S. Shegravina
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| | - Nikolay S. Sitnikov
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| | - Vladimir I. Faerman
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| | - Valery V. Fokin
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| | - Hans-Gunther Schmalz
- Department
of Chemistry, University of Cologne, Greinstrasse 4, 50939 Koln, Germany
| | - Sebastien Combes
- CRCM,
CNRS UMR7258, Laboratory of Integrative Structural and Chemical Biology
(ISCB), INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Universit́e, UM105,
F-13009, Marseille, France
| | - Diane Allegro
- Centre de Recherche en Oncologie Biologique et en Oncopharmacologie,
CRO2 INSERM UMR 911, Faculte de Pharmacie, Universite d’Aix-Marseille, 27 Boulevard Jean Moulin, Marseille 13005, France
| | - Pascal Barbier
- Centre de Recherche en Oncologie Biologique et en Oncopharmacologie,
CRO2 INSERM UMR 911, Faculte de Pharmacie, Universite d’Aix-Marseille, 27 Boulevard Jean Moulin, Marseille 13005, France
| | - Irina P. Beletskaya
- Department
of Chemistry, M.V. Lomonosov Moscow State University, Vorobyevy Gory, 119992 Moscow, Russian Federation
| | - Elena V. Svirshchevskaya
- Laboratory
of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, GSP-7, Miklukho-Maklaya Street, 16/10, 117997 Moscow, Russian Federation
| | - Alexey Yu. Fedorov
- Department
of Organic Chemistry, Nizhny Novgorod State University, Gagarina
av. 23, Nizhny Novgorod 603950, Russian Federation
| |
Collapse
|
21
|
A combretastatin-mediated decrease in neutrophil concentration in peripheral blood and the impact on the anti-tumor activity of this drug in two different murine tumor models. PLoS One 2014; 9:e110091. [PMID: 25299269 PMCID: PMC4192533 DOI: 10.1371/journal.pone.0110091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 09/15/2014] [Indexed: 12/19/2022] Open
Abstract
The vascular disrupting agent combretastatin A-4 disodium phosphate (CA4P) induces fluctuations in peripheral blood neutrophil concentration. Because neutrophils have the potential to induce both vascular damage and angiogenesis we analyzed neutrophil involvement in the anti-tumoral effects of CA4P in C3H mammary carcinomas in CDF1 mice and in SCCVII squamous cell carcinomas in C3H/HeN mice. Flow cytometry analyses of peripheral blood before and up to 144 h after CA4P administration (25 and 250 mg/kg) revealed a decrease 1 h after treatment, followed by an early (3–6 h) and a late (>72 h) increase in the granulocyte concentration. We suggest that the early increase (3–6 h) in granulocyte concentration was caused by the initial decrease at 1 h and found that the late increase was associated with tumor size, and hence independent of CA4P. No alterations in neutrophil infiltration into the C3H tumor after CA4P treatment (25 and 250 mg/kg) were found. Correspondingly, neutrophil depletion in vivo, using an anti-neutrophil antibody, followed by CA4P treatment (25 mg/kg) did not increase the necrotic fraction in C3H tumors significantly. However, by increasing the CA4P dose to 250 mg/kg we found a significant increase of 359% in necrotic fraction when compared to neutrophil-depleted mice; in mice with no neutrophil depletion CA4P induced an 89% change indicating that the presence of neutrophils reduced the effect of CA4P. In contrast, neither CA4P nor 1A8 affected the necrotic fraction in the SCCVII tumors significantly. Hence, we suggest that the initial decrease in granulocyte concentration was caused by non-tumor-specific recruitment of neutrophils and that neutrophils may attenuate CA4P-mediated anti-tumor effect in some tumor models.
Collapse
|
22
|
Méndez-Callejas GM, Leone S, Tanzarella C, Antoccia A. Combretastatin A-4 induces p53 mitochondrial-relocalisation independent-apoptosis in non-small lung cancer cells. Cell Biol Int 2013; 38:296-308. [DOI: 10.1002/cbin.10199] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 10/04/2013] [Indexed: 12/16/2022]
Affiliation(s)
- Gina Marcela Méndez-Callejas
- Departament of Science University ‘Roma Tre’; V.le G. Marconi 446 00146 Rome Italy
- Universidad de Ciencias Aplicadas y Ambientales; Calle 222 55-37 Bogotá Colombia
| | - Stefano Leone
- Departament of Science University ‘Roma Tre’; V.le G. Marconi 446 00146 Rome Italy
| | - Caterina Tanzarella
- Departament of Science University ‘Roma Tre’; V.le G. Marconi 446 00146 Rome Italy
| | - Antonio Antoccia
- Departament of Science University ‘Roma Tre’; V.le G. Marconi 446 00146 Rome Italy
| |
Collapse
|
23
|
Feng D, Menger MD, Laschke MW. Vascular disrupting effects of combretastatin A4 phosphate on murine endometriotic lesions. Fertil Steril 2013; 100:1459-67. [DOI: 10.1016/j.fertnstert.2013.07.1967] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 11/16/2022]
|
24
|
Synthesis and anticancer activity of 2-benzylidene indanones through inhibiting tubulin polymerization. Bioorg Med Chem 2012; 20:3049-57. [DOI: 10.1016/j.bmc.2012.02.057] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 02/24/2012] [Accepted: 02/25/2012] [Indexed: 11/19/2022]
|
25
|
|
26
|
Erratum. Br J Clin Pharmacol 2011. [DOI: 10.1111/j.1365-2125.2011.04030.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|