Size | Price | |
---|---|---|
500mg | ||
1g | ||
Other Sizes |
Fosbretabulin (CA4P; Zybrestat) is a naturally occuring cis-stilbene compound and is the water-soluble prodrug of CA-4. It that has to be activated to the active form, Combretastatin A4 (CA4), which is a highly potent MTA (microtubule-targeting agent) or tubulin inhibitor that acts as a potent anticancer agent that binds β-tubulin with a Kd of 0.4 μM in a cell-free assay. Fosbretabulin was originally isolated from African tree Combretum caffrum. Combretastatin A4 inhibits the polymerization of tubulin with an IC50 of 2.4 μM, and also disrupts tumor vasculature. Under in vivo conditions, the prodrug fosbretabulin is dephosphorylated and activated to its active form, combretastatin A4, which binds to tubulin dimers and prevents microtubule polymerization, resulting in mitotic arrest and apoptosis in endothelial cells.
Targets |
Microtubule; tubulin polymerization; β-tubulin (Kd = 0.4 μM)
|
---|---|
ln Vitro |
Forward scatter is greatly reduced and the proportion of Annexin-V bound cells is significantly increased when combretastatin A4 phosphate (≥ 50 μM) is used. The amount of hemolysis is not considerably increased by combretastatin A4 phosphate. Combretastatin A4 phosphate at concentrations of several hundred μM markedly increased Fluo3 fluorescence. When extracellular Ca2+ is removed, the effect of Combretastatin A4 phosphate (100 μM) on Annexin-V binding is greatly reduced but not completely eliminated. ROS and ceramide are not significantly increased by combretastatin A4 phosphate (≥ 50 μM), but it does dramatically lower GSH abundance and ATP levels [2]. Strong synergistic cytotoxicity was demonstrated by polymer capsules co-encapsulating doxorubicin-combretastatin-A4 phosphate (1:10) against human nasopharyngeal epithelial carcinoma (KB) cells [3]. The expression of these important molecules and the quantity of VM in 3-D cells are unaffected by pretreatment with combretastatin A4 phosphate [4].
A 48 hours exposure of human erythrocytes to Combretastatin A4/CA4P (≥ 50 µM) significantly increased the percentage of annexin-V-binding cells and significantly decreased forward scatter. Combretastatin A4/CA4P did not appreciably increase hemolysis. Hundred µM CA4P significantly increased Fluo3-fluorescence. The effect of CA4P (100 µM) on annexin-V-binding was significantly blunted, but not abolished, by removal of extracellular Ca2+. CA4P (≥ 50 µM) significantly decreased GSH abundance and ATP levels but did not significantly increase ROS or ceramide. Conclusions: Combretastatin A4CA4P triggers cell shrinkage and phospholipid scrambling of the erythrocyte cell membrane, an effect at least in part due to entry of extracellular Ca2+ and energy depletion.[2] In vitro model of three-dimensional cultures was used to test the effect of Combretastatin A4/CA4P on the tube formation of Walker 256 cells. Western blot analysis was conducted to assess the expression of hypoxia-inducible factor (HIF)-1α and VM-associated markers.Under hypoxic conditions for 48 h in vitro, W256 cells formed VM network associated with increased expression of VM markers. Pretreatment with CA4P did not influence the amount of VM in 3-D culture as well as the expression of these key molecules [4]. FosbretabuLin disodium suppresses the proliferation of leukemia P-388, pancreatic BXPC-3, neuroblast SK-N-SH, thyroid SW1736, lung-NSC NCI-H460, prostate DU-145, and pharyngeal FADU with EC50 of 0.0029, 0.23, and 0.00025, respectively., 0.00061, 0.00035, 0.00072, and 0.00045 μg/mL[8]. |
ln Vivo |
In rats, FosbretabuLin disodium (100 mg/kg; ip) decreases tumor blood flow and raises mean arterial blood pressure (MABP) one and six hours after injection. The potential for tumor vascular-targeting by using the tubulin destabilizing agent disodium combretastatin A-4 3-0-phosphate (CA-4-P) was assessed in a rat system. This approach aims to shut down the established tumor vasculature, leading to the development of extensive tumor cell necrosis. The early vascular effects of CA-4-P were assessed in the s.c. implanted P22 carcinosarcoma and in a range of normal tissues. Blood flow was measured by the uptake of radiolabeled iodoantipyrine, and quantitative autoradiography was used to measure spatial heterogeneity of blood flow in tumor sections. CA-4-P (100 mg/kg i.p.) caused a significant increase in mean arterial blood pressure at 1 and 6 h after treatment and a very large decrease in tumor blood flow, which-by 6 h-was reduced approximately 100-fold. The spleen was the most affected normal tissue with a 7-fold reduction in blood flow at 6 h. Calculations of vascular resistance revealed some vascular changes in the heart and kidney for which there were no significant changes in blood flow. Quantitative autoradiography showed that CA-4-P increased the spatial heterogeneity in tumor blood flow. The drug affected peripheral tumor regions less than central regions. Administration of CA-4-P (30 mg/kg) in the presence of the nitric oxide synthase inhibitor, N(omega)-nitro-L-arginine methyl ester, potentiated the effect of CA-4-P in tumor tissue. The combination increased tumor vascular resistance 300-fold compared with less than 7-fold for any of the normal tissues. This shows that tissue production of nitric oxide protects against the damaging vascular effects of CA-4-P. Significant changes in tumor vascular resistance could also be obtained in isolated tumor perfusions using a cell-free perfusate, although the changes were much less than those observed in vivo. This shows that the action of CA-4-P includes mechanisms other than those involving red cell viscosity, intravascular coagulation, and neutrophil adhesion. The uptake of CA-4-P and combretastatin A-4 (CA-4) was more efficient in tumor than in skeletal muscle tissue and dephosphorylation of CA-4-P to CA-4 was faster in the former. These results are promising for the use of CA-4-P as a tumor vascular-targeting agent [9].
Thirty minutes after the treatment, rats given 120 mg/10 mL/kg of Combretastatin A4 disodium phosphate had more DBP and MBP. Rats treated with Combretastatin A4 disodium phosphate 120 mg/10 mL/kg showed the following toxicokinetic characteristics for both Combretastatin A4 and its phosphate: Cmax, T1/2, and AUC0-inf values of Combretastatin A4 were 156± 13 μM, 5.87±1.69 h, and 89.4±10.1 h·μM[1]. W256 tumors showed a substantial intratumoral hypoxia following combretastatin A4 phosphate therapy, which was associated by an increase in VM development. Tumor growth was delayed with cercopetastatin A4 phosphate for a mere two days, however the growth of the tumor quickly resumed. Positive correlations were seen between the VM density and the tumor weight and volume on day 8. Through the HIF-1α/EphA2/PI3K/matrix metalloproteinase (MMP) signaling pathway, cercetastatin A4 phosphate stimulates hypoxia and VM formation in W256 tumors, which impairs tumor renewal [4]. In this study, we designed biodegradable polymersomes for co-delivery of an antiangiogenic drug Combretastatin A4 phosphate (CA4P) and doxorubicin (DOX) to collapse tumor neovasculature and inhibit cancer cell proliferation with the aim to achieve synergistic antitumor effects. The polymersomes co-encapsulating DOX and CA4P (Ps-DOX-CA4P) were prepared by solvent evaporation method using methoxy poly(ethylene glycol)-b-polylactide (mPEG-PLA) block copolymers as drug carriers. The resulting Ps-DOX-CA4P has vesicles shape with uniform sizes of about 50 nm and controlled co-encapsulation ratios of DOX to CA4P. More importantly, Ps-DOX-CA4P (1:10) showed strong synergistic cytotoxicity (combination index CI = 0.31) against human nasopharyngeal epidermal carcinoma (KB) cells. Furthermore, Ps-DOX-CA4P accumulated remarkably in KB tissues xenografts in nude mice. Consistent with these observations, Ps-DOX-CA4P (1:10) achieved significant antitumor potency because of fast tumor vasculature disruption and sustained tumor cells proliferation inhibition in vivo. The overall findings indicate that co-delivery of an antiangiogenic drug and a chemotherapeutic agent in polymersomes is a potentially promising strategy for cancer therapy. [3] In vivo, W256 tumors showed marked intratumoral hypoxia after Combretastatin A4/CA4P treatment, accompanied by increased VM formation. CA4P exhibited only a delay in tumor growth within 2 days but rapid tumor regrowth afterward. VM density was positively related to tumor volume and tumor weight at day 8. CA4P causes hypoxia which induces VM formation in W256 tumors through HIF-1α/EphA2/PI3K/matrix metalloproteinase (MMP) signaling pathway, resulting in the consequent regrowth of the damaged tumor [4]. |
Enzyme Assay |
In Vitro Tubulin Polymerization Assay[5,6]
According to the method described by Wang et al.,porcine brain tubulins (>97% pure) were mixed with general tubulin buffer (80 mM PIPES, 2.0 mM MgCl2, 0.5 mM EGTA, and 1 mM GTP) to reach a final concentration of 3 mg/mL at 4 °C. The tubulin polymerization assay was incubated at 37 °C in a SYNERGY 4 Microplate Reader immediately after mixing tubulin protein solution and the test compounds in a 96-well plate and monitored every 30 s for 65 min at 340 nm. The experiment was performed in duplicates with paclitaxel as a positive control for tubulin polymerization, and colchicine and ABI-274 as positive controls for tubulin depolymerization. SPR for Affinity Assay[5,6] Binding affinity with tubulin was analyzed using SPR technology in a Reichert4SPR system equipped with a dextran SPR sensor chip (Reichert Polycarboxylate Hydrogel Chip P/N 13206067). Then, 50 μg/mL tubulin was immobilized to the sensor chip surface to attain 12 000 μRIU. One of the four flow cells on the chip was left free as a negative control. 4v or colchicine at different concentrations was injected over the sensor chip surface for association analysis, followed by dissociation analysis. The experiment data were obtained at 25 °C with a running buffer PBST (8 mM Na2HPO4, 136 mM NaCl, 2 mM KH2PO4, 2.6 mM KCl, and 0.05% (v/v) Tween 20, pH 7.4). The equilibrium dissociation constant (KD) was calculated by a steady-state fitting mode with TraceDrawer software. |
Cell Assay |
Evaluation of cellular impedance [1]
Analysis of cellular impedance of hiPS-CMs using an xCELLigence Cardio Analyzer was performed with reference to and modification of the methods in earlier studies. Briefly, iCell hiPS-CMs were purchased from Cellular Dynamics International. hiPS-CMs were thawed and cultured in 96-well xCELLigence Cardio E-plates at 20,000 cells/well and 37°C in 5% CO2, using plating medium and maintenance medium specifically for iCell hiPS-CMs, according to the manufacturer’s protocol. During the incubation period, the impedance values were monitored continuously using an xCELLigence Cardio Analyzer according to the manufacturer’s instructions. Impedance was continuously sampled at 12.9 ms intervals and monitored at every measurement point with a 20 second sweep duration. After incubation for 14 days, test compounds (100 nM, 1 μM, and 10 μM CA4DP; 100 nM, 1 μM, and 10 μM Combretastatin A4/CA4; and 0.1% H2O for CA4DP or 0.1% DMSO for CA4 [vehicle]) (n = 3 well) were added to the culture. Then, impedance cell index (CI) and beating rate16, 18, 20 were calculated using dedicated software. Data for CI and beating rate were normalized by the value immediately before the addition of test compounds. The CI for 36 hours after administration was used to detect cytotoxic effects. The beating rate at 15 minutes, 3 hours, and 12 hours after administration were used to detect changes in contractility. Background/aims: Combretastatin A4 phosphate disodium (CA4P) is utilized for the treatment of malignancy. The substance has previously been shown to trigger suicidal cell death or apoptosis. Similar to apoptosis of nucleated cells, erythrocytes may enter suicidal death or eryptosis, characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. Stimulators of eryptosis include increase of cytosolic Ca2+ activity ([Ca2+]i), ceramide, oxidative stress and ATP depletion. The present study explored, whether CA4P induces eryptosis and, if so, to gain insight into mechanisms involved. Methods: Flow cytometry has been employed to estimate phosphatidylserine exposure at the cell surface from annexin-V-binding, cell volume from forward scatter, [Ca2+]i from Fluo3-fluorescence, reactive oxygen species (ROS) abundance from DCF fluorescence, glutathione (GSH) abundance from CMF fluorescence and ceramide abundance from fluorescent antibodies. In addition cytosolic ATP levels were quantified utilizing a luciferin-luciferase-based assay and hemolysis was estimated from hemoglobin concentration in the supernatant [2]. |
Animal Protocol |
Animal/Disease Models: Male BD9 rats (7-9 weeks) bearing the sc implanted P22 tumor[9]
Doses: 100 mg/kg Route of Administration: A single ip injections Experimental Results: Dramatically raised the MABP by about 30%, and decreased the heart rate at 1 h after administration. decreased the blood flow in the tumor. Evaluation of histopathological changes [1] A total of 14 rats were divided into four groups as described in Table 1. At 6 weeks of age, CA4DP/Combretastatin A4 (four doses of 30 or 60 mg/10 mL/kg at intervals of 24 hours or two doses of 120 mg/10 mL/kg at an interval of 72 hours) or saline (two doses at an interval of 72 hours) was administered via the caudal vein by bolus infusion. On the day after the last administration, the rats were anesthetized with isoflurane, and necropsy was performed. Also, one rat administered four doses of CA4DP 60 mg/10 mL/kg died unexpectedly before necropsy because of CA4DP toxicity. The cause of death was thought to be the cardiotoxicity of CA4DP because severe myocardial necrosis had been observed in this rat. After exsanguination, the hearts of the rats were removed and immediately fixed in 10% neutral phosphate-buffered formalin. The fixed hearts were cross-sectioned in two planes through the ventricles as described in a previous report7. The fixed hearts was embedded in paraffin and sectioned at a thickness of 4-6 μm. The specimens were stained with hematoxylin and eosin (HE). Observation of these specimens was performed using a light microscope. Evaluation of ECG data [1] Two rats were used (animal No. 1 and No. 2). At 5 weeks of age, a small telemetry device (weight = 3.9 g, volume = 1.9 cc) for transmitting ECG data was implanted in the dorsal subcutaneous region under anesthesia with pentobarbital sodium. Paired wire electrodes that came with the telemetry device were placed under the skin of the dorsal and ventral thorax to record the apex-base (A–B) lead ECG. One week after surgery, ECG signals were recorded from each rat in a cage that had been placed on a signal-receiving board. ECG data were continuously sampled at 1 ms intervals, and all data analyses of ECG-wave components were performed using an ECG processor analyzing system on a personal computer in series with an analog-digital converter; the ECG data were stored on an external hard disk. During the period of ECG recording, CA4DP/Combretastatin A4 50 mg/10 mL/kg was administered to both rats via the caudal vein by bolus infusion, 3 times at intervals of 24 hours. ECG was recorded until 12 hours after the third administration. The consecutive ECG waves for 4 seconds were averaged, and the ECG wave components (RR interval, QRS duration, PR interval, and QT interval) were analyzed. Evaluation of BP [1] A total of 9 rats were used. At 6 weeks of age, rats were anesthetized with isoflurane, and placed in a supine position. The femoral artery was exposed, and a polyethylene catheter filled with heparinized saline was inserted. The catheter was connected to transducer amplification equipment via a pressure transducer, and the arterial pressure was recorded. BP was continuously sampled at 1 ms intervals, and all data analyses were performed using an ECG processor analyzing system on a personal computer in series with an analog-digital converter. During the period of BP recording, CA4DP/Combretastatin A4 120 mg/10 mL/kg or saline 10 mL/kg was administered as a single dose via the caudal vein by bolus infusion (n = 5 for CA4DP and n = 4 for saline). BP was recorded until 30 minutes after administration. Consecutive BP waves for 4 seconds were averaged, and the BP components (systolic BP [SBP], diastolic BP [DBP], and mean BP [MBP]) and heart rate (HR) were analyzed. Toxicokinetic analysis [1] Rats were administered a single intravenous dose of CA4DP/Combretastatin A4 at 120 mg/10 mL/kg by bolus infusion (n = 3). Blood was taken via the jugular vein and collected in heparin-coated tubes at 10 minutes and 1, 3, 6, and 24 hours after administration. Plasma was separated by centrifugation immediately after sampling. After centrifugation, an aliquot of plasma was mixed with the equivalent volume of 1% formic acid and stored at −20°C. The thawed plasma samples were purified by solid-phase extraction, and the plasma concentrations of combretastatin A4 phosphate (free base of CA4DP; CA4P) and combretastatin A4 (the metabolite of CA4DP; CA4) were determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Toxicokinetic parameters [maximum concentration (Cmax), terminal half-life (T1/2), and area under the concentration-time curve from time zero to infinity (AUC0-inf)] were obtained by non-compartmental analysis using Phoenix WinNonlin 6.3. |
ADME/Pharmacokinetics |
Metabolism / Metabolites
Combretastatin A4 has known human metabolites that include (2S,3S,4S,5R)-3,4,5-trihydroxy-6-[2-methoxy-5-[(Z)-2-(3,4,5-trimethoxyphenyl)ethenyl]phenoxy]oxane-2-carboxylic acid. |
References |
|
Additional Infomation |
Fosbretabulin has been investigated for the treatment of Anaplastic Thyroid Cancer.
Fosbretabulin Disodium is the disodium salt of a water-soluble phosphate derivative of a natural stilbenoid phenol derived from the African bush willow (Combretum caffrum) with potential vascular disrupting and antineoplastic activities. Upon administration, the prodrug fosbretabulin is dephosphorylated to its active metabolite, the microtubule-depolymerizing agent combretastatin A4, which binds to tubulin dimers and prevents microtubule polymerization, resulting in mitotic arrest and apoptosis in endothelial cells. In addition, this agent disrupts the engagement of the endothelial cell-specific junctional molecule vascular endothelial-cadherin (VE-cadherin) and so the activity of the VE-cadherin/beta-catenin/Akt signaling pathway, which may result in the inhibition of endothelial cell migration and capillary tube formation. As a result of fosbretabulin's dual mechanism of action, the tumor vasculature collapses, resulting in reduced tumor blood flow and ischemic necrosis of tumor tissue. Fosbretabulin is a water-soluble prodrug derived from the African bush willow (Combretum caffrum) with antineoplastic activity. Fosbretabulin is dephosphorylated to its active metabolite, combretastatin A4, which binds to tubulin and inhibits microtubule polymerization, resulting in mitotic arrest and apoptosis in endothelial cells. As apoptotic endothelial cells detach from their substrata, tumor blood vessels collapse; the acute disruption of tumor blood flow may result in tumor necrosis. Combretastatin A4 is a stilbenoid. Combretastatin A4 has been reported in Combretum caffrum with data available. Combretastatin A-4 is an inhibitor of microtubule polymerization derived from the South African willow bush which causes mitotic arrest and selectively targets and reduces or destroys existing blood vessels, causing decreased tumor blood supply. See also: Fosbretabulin (annotation moved to). Combretastatin A-4 is an inhibitor of microtubule polymerization derived from the South African willow bush which causes mitotic arrest and selectively targets and reduces or destroys existing blood vessels, causing decreased tumor blood supply. Histopathological and electrocardiographic features of myocardial lesions induced by combretastatin A4 disodium phosphate (CA4DP) were evaluated, and the relation between myocardial lesions and vascular changes and the direct toxic effect of CA4DP on cardiomyocytes were discussed. We induced myocardial lesions by administration of CA4DP to rats and evaluated myocardial damage by histopathologic examination and electrocardiography. We evaluated blood pressure (BP) of CA4DP-treated rats and effects of CA4DP on cellular impedance-based contractility of human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs). The results revealed multifocal myocardial necrosis with a predilection for the interventricular septum and subendocardial regions of the apex of the left ventricular wall, injury of capillaries, morphological change of the ST junction, and QT interval prolongation. The histopathological profile of myocardial lesions suggested that CA4DP induced a lack of myocardial blood flow. CA4DP increased the diastolic BP and showed direct effects on hiPS-CMs. These results suggest that CA4DP induces dysfunction of small arteries and capillaries and has direct toxicity in cardiomyocytes. Therefore, it is thought that CA4DP induced capillary and myocardial injury due to collapse of the microcirculation in the myocardium. Moreover, the direct toxic effect of CA4DP on cardiomyocytes induced myocardial lesions in a coordinated manner.[1] The purpose of this study was to investigate the effect of combretastatin A4 phosphate (CA4P) on vasculogenic mimicry (VM) channel formation in vitro and in vivo after a single-dose treatment and the underlying mechanism involved in supporting VM. In vitro model of three-dimensional cultures was used to test the effect of CA4P on the tube formation of Walker 256 cells. Western blot analysis was conducted to assess the expression of hypoxia-inducible factor (HIF)-1α and VM-associated markers. W256 tumor-bearing rat model was established to demonstrate the effect of CA4P on VM formation and tumor hypoxia by double staining and a hypoxic marker pimonidazole. Anti-tumor efficacy of CA4P treatment was evaluated by tumor growth curve. Under hypoxic conditions for 48 h in vitro, W256 cells formed VM network associated with increased expression of VM markers. Pretreatment with CA4P did not influence the amount of VM in 3-D culture as well as the expression of these key molecules. In vivo, W256 tumors showed marked intratumoral hypoxia after CA4P treatment, accompanied by increased VM formation. CA4P exhibited only a delay in tumor growth within 2 days but rapid tumor regrowth afterward. VM density was positively related to tumor volume and tumor weight at day 8. CA4P causes hypoxia which induces VM formation in W256 tumors through HIF-1α/EphA2/PI3K/matrix metalloproteinase (MMP) signaling pathway, resulting in the consequent regrowth of the damaged tumor.[4] We recently reported the crystal structure of tubulin in complex with a colchicine binding site inhibitor (CBSI), ABI-231, having 2-aryl-4-benzoyl-imidazole (ABI). Based on this and additional crystal structures, here we report the structure-activity relationship study of a novel series of pyridine analogues of ABI-231, with compound 4v being the most potent one (average IC50 ∼ 1.8 nM) against a panel of cancer cell lines. We determined the crystal structures of another potent CBSI ABI-274 and 4v in complex with tubulin and confirmed their direct binding at the colchicine site. 4v inhibited tubulin polymerization, strongly suppressed A375 melanoma tumor growth, induced tumor necrosis, disrupted tumor angiogenesis, and led to tumor cell apoptosis in vivo. Collectively, these studies suggest that 4v represents a promising new generation of tubulin inhibitors. [5] Novel ABI-III compounds were designed and synthesized based on our previously reported ABI-I and ABI-II analogues. ABI-III compounds are highly potent against a panel of melanoma and prostate cancer cell lines, with the best compound having an average IC(50) value of 3.8 nM. They are not substrate of Pgp and thus may effectively overcome Pgp-mediated multidrug resistance. ABI-III analogues maintain their mechanisms of action by inhibition of tubulin polymerization.[6] A series of cis- and trans-stilbenes related to combretastatin A-4 (1a), with a variety of substituents at the 3'-position of the aryl B-ring, were synthesized and evaluated for inhibitory activity employing six human cancer cell lines (NCI-H460 lung carcinoma, BXPC-3 pancreas, SK-N-SH neuroblastoma, SW1736 thyroid, DU-145 prostate, and FADU pharynx-squamous sarcoma) as well as the P-388 murine lymphocyte leukemia cell line. Several of the cis-stilbene derivatives were significantly inhibitory against all cell lines used, with potencies comparable to that of the parent 1a. All were potent inhibitors of tubulin polymerization. The corresponding trans-stilbenes had little or no activity as tubulin polymerization inhibitors and were relatively inactive against the seven cancer cell lines. In terms of inhibition of both cancer cell growth and tubulin polymerization, the dimethylamino and bromo cis-stilbenes were the most potent of the new derivatives, the latter having biological activity approaching that of 1a. As part of the present study, the X-ray crystal structure of the 3'-O-phosphate of combretastatin A-4 (1b) was successfully elucidated. Compound 1b has been termed the "combretastatin A-4 prodrug", and it is currently undergoing clinical trials for the treatment of human cancer patients.[8] The potential for tumor vascular-targeting by using the tubulin destabilizing agent disodium combretastatin A-4 3-0-phosphate (CA-4-P) was assessed in a rat system. This approach aims to shut down the established tumor vasculature, leading to the development of extensive tumor cell necrosis. The early vascular effects of CA-4-P were assessed in the s.c. implanted P22 carcinosarcoma and in a range of normal tissues. Blood flow was measured by the uptake of radiolabeled iodoantipyrine, and quantitative autoradiography was used to measure spatial heterogeneity of blood flow in tumor sections. CA-4-P (100 mg/kg i.p.) caused a significant increase in mean arterial blood pressure at 1 and 6 h after treatment and a very large decrease in tumor blood flow, which-by 6 h-was reduced approximately 100-fold. The spleen was the most affected normal tissue with a 7-fold reduction in blood flow at 6 h. Calculations of vascular resistance revealed some vascular changes in the heart and kidney for which there were no significant changes in blood flow. Quantitative autoradiography showed that CA-4-P increased the spatial heterogeneity in tumor blood flow. The drug affected peripheral tumor regions less than central regions. Administration of CA-4-P (30 mg/kg) in the presence of the nitric oxide synthase inhibitor, N(omega)-nitro-L-arginine methyl ester, potentiated the effect of CA-4-P in tumor tissue. The combination increased tumor vascular resistance 300-fold compared with less than 7-fold for any of the normal tissues. This shows that tissue production of nitric oxide protects against the damaging vascular effects of CA-4-P. Significant changes in tumor vascular resistance could also be obtained in isolated tumor perfusions using a cell-free perfusate, although the changes were much less than those observed in vivo. This shows that the action of CA-4-P includes mechanisms other than those involving red cell viscosity, intravascular coagulation, and neutrophil adhesion. The uptake of CA-4-P and combretastatin A-4 (CA-4) was more efficient in tumor than in skeletal muscle tissue and dephosphorylation of CA-4-P to CA-4 was faster in the former. These results are promising for the use of CA-4-P as a tumor vascular-targeting agent.[9] |
Molecular Formula |
C18H21O8P
|
---|---|
Molecular Weight |
396.32834
|
Exact Mass |
396.097
|
Elemental Analysis |
C, 54.55; H, 5.34; O, 32.29; P, 7.82
|
CAS # |
222030-63-9
|
Related CAS # |
168555-66-6 (disodium);222030-63-9 (free acid);404886-32-4 ( tromethamine);
|
PubChem CID |
5351387
|
Appearance |
Typically exists as solid at room temperature
|
Density |
1.342g/cm3
|
Boiling Point |
611.8ºC at 760mmHg
|
Flash Point |
323.8ºC
|
Vapour Pressure |
7.99E-16mmHg at 25°C
|
Index of Refraction |
1.609
|
LogP |
3.362
|
Hydrogen Bond Donor Count |
2
|
Hydrogen Bond Acceptor Count |
8
|
Rotatable Bond Count |
8
|
Heavy Atom Count |
27
|
Complexity |
508
|
Defined Atom Stereocenter Count |
0
|
SMILES |
O=P(O)(OC1=CC(/C=C\C2=CC(OC)=C(OC)C(OC)=C2)=CC=C1OC)O
|
InChi Key |
WDOGQTQEKVLZIJ-WAYWQWQTSA-N
|
InChi Code |
InChI=1S/C18H21O8P/c1-22-14-8-7-12(9-15(14)26-27(19,20)21)5-6-13-10-16(23-2)18(25-4)17(11-13)24-3/h5-11H,1-4H3,(H2,19,20,21)/b6-5-
|
Chemical Name |
[2-methoxy-5-[(Z)-2-(3,4,5-trimethoxyphenyl)ethenyl]phenyl] dihydrogen phosphate
|
Synonyms |
Fosbretabulin; 222030-63-9; Combretastatin A-4 phosphate; Phosbretabulin; Fosbretabulin [INN]; combretastatin A4 phosphate; (Z)-2-Methoxy-5-(3,4,5-trimethoxystyryl)phenyl dihydrogen phosphate; fosbretabulina; Fosbretabulin disodium; 168555-66-6; Combretastatin A4 disodium phosphate; CA4DP; CA 4P; Combretastatin A4 Phosphate Disodium Salt; Fosbretabulin disodium [USAN]; CA-4DP;
|
HS Tariff Code |
2934.99.9001
|
Storage |
Powder -20°C 3 years 4°C 2 years In solvent -80°C 6 months -20°C 1 month |
Shipping Condition |
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
|
Solubility (In Vitro) |
May dissolve in DMSO (in most cases), if not, try other solvents such as H2O, Ethanol, or DMF with a minute amount of products to avoid loss of samples
|
---|---|
Solubility (In Vivo) |
Note: Listed below are some common formulations that may be used to formulate products with low water solubility (e.g. < 1 mg/mL), you may test these formulations using a minute amount of products to avoid loss of samples.
Injection Formulations
Injection Formulation 1: DMSO : Tween 80: Saline = 10 : 5 : 85 (i.e. 100 μL DMSO stock solution → 50 μL Tween 80 → 850 μL Saline)(e.g. IP/IV/IM/SC) *Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH ₂ O to obtain a clear solution. Injection Formulation 2: DMSO : PEG300 :Tween 80 : Saline = 10 : 40 : 5 : 45 (i.e. 100 μL DMSO → 400 μLPEG300 → 50 μL Tween 80 → 450 μL Saline) Injection Formulation 3: DMSO : Corn oil = 10 : 90 (i.e. 100 μL DMSO → 900 μL Corn oil) Example: Take the Injection Formulation 3 (DMSO : Corn oil = 10 : 90) as an example, if 1 mL of 2.5 mg/mL working solution is to be prepared, you can take 100 μL 25 mg/mL DMSO stock solution and add to 900 μL corn oil, mix well to obtain a clear or suspension solution (2.5 mg/mL, ready for use in animals). View More
Injection Formulation 4: DMSO : 20% SBE-β-CD in saline = 10 : 90 [i.e. 100 μL DMSO → 900 μL (20% SBE-β-CD in saline)] Oral Formulations
Oral Formulation 1: Suspend in 0.5% CMC Na (carboxymethylcellulose sodium) Oral Formulation 2: Suspend in 0.5% Carboxymethyl cellulose Example: Take the Oral Formulation 1 (Suspend in 0.5% CMC Na) as an example, if 100 mL of 2.5 mg/mL working solution is to be prepared, you can first prepare 0.5% CMC Na solution by measuring 0.5 g CMC Na and dissolve it in 100 mL ddH2O to obtain a clear solution; then add 250 mg of the product to 100 mL 0.5% CMC Na solution, to make the suspension solution (2.5 mg/mL, ready for use in animals). View More
Oral Formulation 3: Dissolved in PEG400  (Please use freshly prepared in vivo formulations for optimal results.) |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 2.5231 mL | 12.6157 mL | 25.2315 mL | |
5 mM | 0.5046 mL | 2.5231 mL | 5.0463 mL | |
10 mM | 0.2523 mL | 1.2616 mL | 2.5231 mL |
*Note: Please select an appropriate solvent for the preparation of stock solution based on your experiment needs. For most products, DMSO can be used for preparing stock solutions (e.g. 5 mM, 10 mM, or 20 mM concentration); some products with high aqueous solubility may be dissolved in water directly. Solubility information is available at the above Solubility Data section. Once the stock solution is prepared, aliquot it to routine usage volumes and store at -20°C or -80°C. Avoid repeated freeze and thaw cycles.
Calculation results
Working concentration: mg/mL;
Method for preparing DMSO stock solution: mg drug pre-dissolved in μL DMSO (stock solution concentration mg/mL). Please contact us first if the concentration exceeds the DMSO solubility of the batch of drug.
Method for preparing in vivo formulation::Take μL DMSO stock solution, next add μL PEG300, mix and clarify, next addμL Tween 80, mix and clarify, next add μL ddH2O,mix and clarify.
(1) Please be sure that the solution is clear before the addition of next solvent. Dissolution methods like vortex, ultrasound or warming and heat may be used to aid dissolving.
(2) Be sure to add the solvent(s) in order.