Size | Price | |
---|---|---|
500mg | ||
1g | ||
Other Sizes |
Vincristine (also known as Leurocristine) sulfate, a natural alkaloid isolated from the plant Vinca rosea Linn, is a potent inhibitor of microtubule polymerization by binding to tubulin with IC50 of 32 μM in a cell-free assay. Vincristine is extracted from leaves of the periwinkle plant Catharanthus roseus (L.) G. Don of the family Apocynaceae. Vincristine binds irreversibly to microtubules and spindle proteins in S phase of the cell cycle and interferes with the formation of the mitotic spindle, thereby arresting tumor cells in metaphase. This agent also depolymerizes microtubules and may also interfere with amino acid, cyclic AMP, and glutathione metabolism.
Targets |
Tubulin polymerization; Microtubule (Ki = 85 nM)
|
||||
---|---|---|---|---|---|
ln Vitro |
In vitro activity: Vincristine inhibits net addition of tubulin dimers at assembly ends of steady-state microtubules with Ki of 85 NM. At low concentrations, Vincristine stabilizes the spindle apparatus resulting in failure of the chromosomes to segregate leading to metaphase arrest and inhibition of mitosis. At higher concentrations, Vincristine may disrupt and induce total depolymerization of microtubules. Vincristine induces apoptosis in tumor cells and inhibits SH-SY5Y cell proliferation with IC50 of 0.1 μM. Vincristine induces mitotic arrest and promots the expression of caspase-3 and -9 and cyclin B, while decreasing the expression of cyclin D. Vincristine induced neurotoxicity is caused by interference with microtubule function, which results in blockage of axonal transport and thus in axonal degeneration.
Cell Assay: Cells are plated in 2 mL of medium in 35-mm plates at a concentration of about 5 × 104 cells/mL and grow for 24 h at 37 ℃ in an atmosphere of 5% CO2 and 95% air. Then medium is replaced with fresh medium lacking or containing 4 nM drug and proliferation is continued for 3 days. Cell counts are done each day in a Coulter Counter after detaching the cells with trypsin and EDTA. Axonal ultrastructural changes induced by three Vinca alkaloids, Vincristine, vinblastine, and desacetyl vinblastine amide, were studied in vitro at concentrations of 0.01, 0.05, and 0.1 mM in the cat vagus nerve. Disruption of microtubules, appearance of paracrystalline structures, and increase in neurofilaments were induced by all three agents at 0.1 mM. A new type of paracrystal with an electron-dense central core in each subunit was also observed with each drug. Whereas all three compounds affected unmyelinated fibers (vinblastine more so than the other two), only vinblastine significantly damaged the myelinated fibers. The greater effectiveness of vinblastine in causing these in vitro ultrastructural changes contrasts strikingly with the clinical in vivo situation in which vincristine is the most neurotoxic. This suggests that clinical neurotoxicity is associated with additional factors aside from the direct interaction of the Vinca alkaloids with microtubules or tubulin.[3] Cytotoxic effects of Vincristine and SAHA, alone and in combination, on human leukemic MOLT-4 cells [7] A 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay was performed to investigate the cytotoxicity of the microtubule-destabilizing agent vincristine and the HDACi vorinostat (SAHA) on human ALL MOLT-4 cells. We first tested the cytotoxic effect of SAHA and vincristine alone and in combination. As shown in Fig. 1a, there was no significant cytotoxicity at concentrations up to 500 nM of SAHA. However, SAHA had an IC50 of 840 nM for 48 h, when concentration reached the highest level (1000 nM). In addition, vincristine exhibited cytotoxicity against human leukemic MOLT-4 cells with an IC50 of 3.3 nM at 48 h (Fig. 1b). To determine whether an interaction between SAHA and vincristine took place, the cytotoxic potency of a combination assay was measured. Cells treated with 500 nM SAHA and various concentrations of vincristine (0.3 to 3 nM) significantly inhibited cell survival compared to each treatment alone (Fig. 1c). Effects of Vincristine in combination with SAHA on human T cell leukemic cell survival [7] To further explore the synergistic cytotoxic effects, we determined the effects on cell cycle distribution. As compared with SAHA, treatment with vincristine induced an increase in the G2/M phase of the cell cycle. In particular, the combination of vincristine plus SAHA caused an almost complete arrest of cells in the G2/M phase following short-term treatment (24 h) and a subsequent induction in the sub-G1 phase following long-term treatment (48 h) (Fig. 2a). Figure 2b shows the statistical results. Next, the combination index (CI) method was used to evaluate the synergistic combinations [25]. A CI value of >1.0, 1.0, and <1.0 indicates an antagonistic, additive, or synergistic interaction, respectively, between the drugs. In the G2/M phase, the CI values of vincristine (0.3, 1, and 3 nM) combined with 500 nM SAHA were 1.63, 0.72, and 0.32, respectively, and the CI values in the sub-G1 phase were 0.97, 0.77, and 0.28, respectively (Fig. 2c). And this synergistic combination effect also was noted in the other T cell leukemic cell line, CCRF-CEM (Fig. 2d), rather than in acute myeloid leukemic cells (Additional file 1: Figure S2). Moreover, vincristine (1 or 3 nM) combined with various concentrations of SAHA also shows synergistic effect (Additional file 2: Figure S1). These data indicate that vincristine and SAHA synergistically induced cell arrest in the G2/M phase and subsequently in the sub-G1 phase. Effects of SAHA in combination with Vincristine on mitotic arrest in human leukemic MOLT-4 cells [7] To further elucidate the synergistic effect mechanism on the G2/M phase of cell cycle progression, we investigated SAHA in combination with vincristine on tubulin polarization change and mitosis-related proteins. As shown in Fig. 3a, there were no obvious tubulin polarization changes following SAHA treatment under cell-free conditions. However, in combination with vincristine, a significant induction of microtubule depolymerization was observed (Fig. 3a). Additional file 3: Figure S3 shows a more comprehensive result, including various vincristine- and SAHA-alone in vitro tubulin polymerization assays. To understand the effects of microtubule dynamics on mitosis following drug treatment, the microtubule arrangement in human leukemic MOLT-4 cells was examined by β-tubulin staining. As shown in Fig. 3b(b), there was no significant change in microtubule distribution and cell morphology after SAHA treatment. In addition, at low vincristine concentrations, cells had accumulated at the metaphase stage of mitosis with abnormal spindles (Fig. 3b(c)). In this study, spindles with bipolar and multipolar organization, which had abnormal long astral microtubules and chromosomes, were found to be unequally distributed. Nevertheless, at a high vincristine concentration, microtubule depolymerization was observed (Fig. 3b(d)). In the present study, the vincristine and SAHA combination exerted more explicit effects than vincristine alone with regard to abnormal spindles and chromosomes (Fig. 3b and Additional file 4: Figure S4). These results suggest that SAHA potentiated the effects of vincristine due to inhibition of microtubule dynamics. Effects of SAHA in combination with Vincristine on the apoptotic pathway and HDAC activity in human leukemic MOLT-4 cells [7] Mitochondria play a crucial role both in the intrinsic and extrinsic apoptotic pathways. To test whether the vincristine/SAHA-mediated apoptotic pathway was associated with mitochondrial function, a change in mitochondrial transmembrane potential (Δψm) was assessed. As shown in Fig. 4a, treatment with SAHA or vincristine alone was insufficient to affect the mitochondrial membrane potential; however, this phenomenon was enhanced by co-treatment with SAHA in a time-dependent manner. The Bcl-2 protein family plays a regulatory role in controlling the mitochondrial apoptotic pathway. The data showed that the combination treatment more effectively downregulated the expression of the pro-survival members of the Bcl-2 family, such as Bcl-2, Bcl-xl, and Mcl-1, than did either treatment alone (Fig. 4b). HDAC6 inhibition was involved in Vincristine/SAHA-induced apoptosis [7] Previous findings have shown HDAC6-induced tubulin acetylation to affect the dynamics and function of microtubules [9–12]. As shown in Fig. 5a, SAHA, a pan-HDACi, induced tubulin acetylation; however, its combination with vincristine had no synergic effect. Tubastatin A, which is a specific HDAC6 inhibitor [26], was used to understand the role of HDAC6 in the vincristine/SAHA-treated cells. To evaluate the potential benefit of vincristine in combination with tubastatin A, the cytotoxicity of co-treatment was determined and the combination effects were analyzed. However, compared to tubastatin A alone, vincristine significantly enhanced the cytotoxicity of tubastatin A (Fig. 5b). Moreover, vincristine (1 and 3 nM) combined with various concentrations of tubastatin A induced cell accumulation at the G2/M phase followed by the sub-G1 phase (Fig. 5c). The CI values were <1 in combination of vincristine and tubastatin at the G2/M phase and sub-G1 phase (Fig. 5d). Co-treatment of vincristine and tubastatin revealed MPM2 and PARP activation consistent with the induction of apoptosis by western blot analysis (Fig. 5e). And vincristine and HDAC6 inhibitor combined synergism effect was further corroborated by the observation of vincristine and ACY1215 co-treatment in CCRF-CEM cells (Fig. 5f). These findings suggest that SAHA treatment may alter microtubule dynamics in cells through HDAC6 inhibition, even though the effect was insufficient to arrest cells in the G2/M phase. However, in combination with vincristine, which also had an effect on microtubules, SAHA caused extreme microtubule stress thus causing cell death. |
||||
ln Vivo |
Vincristine (3 mg/kg) administrated by a single i.p. injection to mice bearing bilateral subcutaneous xenografts Rh12 or Rh18, induces mean growth delay of >120 and >52 day, and repopulating fractions of 0.06% and 5%, respectively. Vincristine acts on subcutaneous colon 38 tumors in mice by host cell-mediated vascular effects as well as by direct tubulin-mediated cytotoxicity. Vincristine (5 mg/kg) reduces tumor blood flow of tumors by nearly 75%.
Vincristine (3 mg/kg, i.p.) given to mice receiving bilateral subcutaneous xenografts of Rh12 or Rh18, respectively, causes a mean growth delay of more than 120 and more than 52 days and repopulates fractions of 0.06% and 5%[5]. The antitumor activity of Vincristine and SAHA combination therapy in vivo [7] To evaluate whether the synergistic effect of Vincristine plus SAHA could be clinically relevant, the antitumor activity of this co-treatment in severe combined immunodeficiency mice bearing established MOLT-4 tumor xenografts was investigated. Once a tumor was palpable (approximately 100 mm3), mice were randomized into vehicle control and treatment groups (n = 6 per group). All mouse tumors were allowed to reach an endpoint volume of 2000 mm3, and in vivo antitumor efficacy was expressed as tumor growth delay (TGD; Fig. 6a). There were no improvements in TGD in mice treated with vincristine (0.1 mg/kg once weekly) or SAHA (50 mg/kg once daily) alone. However, log-rank analysis showed that the co-treatment exhibited significant antitumor activity in the MOLT-4 xenograft model (P = 0.0389). In addition, Kaplan–Meier curves displayed antitumor activity for the co-treatment group (vincristine, 0.025 mg/kg once weekly; SAHA, 200 mg/kg once daily) (Fig. 6b). Notably, the mice tolerated all of the treatments without overt signs of toxicity; no significant body weight difference or other adverse side effects were observed (Fig. 6d and Additional file 5: Figure S5). To correlate the in vivo antitumor effects with the mechanisms identified in vitro, intratumoral biomarkers were assessed by western blot analysis. Consistent with in vitro results, the combined treatment markedly induced caspase 3 activation and PARP cleavage in tumors, indicating elevated apoptosis (Fig. 6e). Taken together, these findings suggest that combination of vincristine and SAHA, both in vitro and in vivo, dramatically enhanced vincristine-induced cell death. Loss of the sense of touch in fingertips and toes is one of the earliest sensory dysfunctions in patients receiving chemotherapy with anti-cancer drugs such as Vincristine. However, mechanisms underlying this chemotherapy-induced sensory dysfunction is incompletely understood. Whisker hair follicles are tactile organs in non-primate mammals which are functionally equivalent to human fingertips. Here we used mouse whisker hair follicles as a model system and applied the pressure-clamped single-fiber recording technique to explore how vincristine treatment affect mechanoreceptors in whisker hair follicles. We showed that in vivo treatment of mice with vincristine impaired whisker tactile behavioral responses. The pressure-clamped single-fiber recordings made from whisker hair follicle afferent nerves showed that mechanical stimulations evoked three types of mechanical responses, rapidly adapting response (RA), slowly adapting type 1 response (SA1) and slowly adapting type 2 response (SA2). Vincristine treatment significantly reduced SA1 responses but did not significantly affect RA and SA2 responses. Our findings suggest that SA1 mechanoreceptors were selectively impaired by vincristine leading to the impairment of in vivo whisker tactile behavioral responses. [8] Tumor responsiveness to Vincristine (VCR) was determined in xenografts of human rhabdomyosarcoma (RMS), in sublines of RMS selected in vivo for VCR resistance, in a KB line (KB-ChR8-5) selected in vitro for colchicine resistance, and in a colon adenocarcinoma (GC3). Sensitivity to VCR was associated with prolonged retention of VCR by the tumors after a single i.p. injection, whereas in tumors with acquired or intrinsic VCR resistance the drug was eliminated more rapidly. The sensitive tumors with prolonged retention of drug also showed increased levels of mitotic accumulation for up to 72 hr following VCR administration. There were good correlations between VCR sensitivity, VCR retention and the proposed mechanism of VCR cytotoxicity-mitotic arrest. A model has been developed consistent with data obtained that can explain the responsiveness to VCR of a series of human tumor xenografts irrespective of their tissue of origin[4]. |
||||
Enzyme Assay |
In vitro tubulin polymerization assay [7]
To determine the microtubule polymerization of the indicated drugs in a cell-free condition, CytoDYNAMIX Screen 03 Kit was performed. General tubulin buffer, GTP stock (100 mM), and tubulin protein (10 mg/ml) were all prepared well following the protocol. A 96-well plate was placed in the spectrophotometer to prewarm at 37 °C for 30 min before detection. Then preparing the iced tubulin polymerization (TP) buffer, all mentioned processes were needed on the ice. Next, the drugs (2 μl) were added into each Eppendorf included with 85 μl TP buffer. The drugs must include DMSO (the control group), paclitaxel (10 μΜ), and Vincristine (10 μΜ). Paclitaxel and vincristine were used as positive controls. Paclitaxel would induce the microtubule polymerization; in contrast, vincristine would depolymerize the microtubules. Finally, 30 μl tubulin proteins was added into the Eppendorf and transferred to the prewarmed 96-well plate. The absorbance was measured by a spectrophotometer and recorded every 1 min for 30 min at 340 nm and 37 °C. VCR/Vincristine release profile in vitro from CS-ALG@TPGS-PL-GA-VCR NPs was researched using the classical dialysis bag method. Briefly, CS-ALG@TPGS-PLGA-VCR NPs (4 mg) were re-suspended in PBS (2 mL, 0.1% w/v Tween 80, pH 5.8 or pH 7.4) to simulate the cancer cytoplasmic environment (pH 5.8) and the physiological environment (pH 7.4). Subsequently, the re-suspension was transferred into a dialysis bag (molecular weight cut off 3 kDa; Millipore, Billerica, MA, USA), which was immersed in PBS (20 mL) and incubated in 37°C thermostatic water bath with 100 rpm shaking. At a designated time interval, 5 mL of the release solution was taken out to detect the concentration of VCR at 298 nm using an ultraviolet spectrophotometer. At the same time, isometric fresh PBS was added into the release medium [6]. Mitochondrial membrane potential [7] Rhodamine 123 was used to evaluate mitochondrial membrane potential. Rhodamine 123 is a kind of cationic fluorescent dye, which localizes in the mitochondria. Loss of mitochondrial membrane potential is associated with a lack of rhodamine 123 retention and a decrease of fluorescence intensity. Cells were treated with Vincristine, SAHA, or combination for the indicated time. Rhodamine 123 (final concentration 10 μM) was added and incubated for 30 min at 37 °C in the dark. Then, cells were harvested and rinsed with PBS. The fluorescence intensity was measured by FACScan Flow Cytometer and CellQuest. |
||||
Cell Assay |
Cell viability assay [7]
Cell viability was verified by MTT assay. Firstly, cells were seeded in a 24-well plate at a density of 4 × 105 cells/well in 1 ml culture medium and then treated with various concentrations of Vincristine or SAHA alone or a combination of both for 24 and 48 h. After treatment with drugs, 100 μl MTT solution (0.5 mg/ml in phosphate-buffered saline (PBS)) per well was added to the 24-well plate in the dark and the plate was incubated at 37 °C. The mitochondrial dehydrogenase of viable cells reduced MTT (yellow) to insoluble formazan dyes (purple). One hour later, the crystal formazan dyes were dissolved in the extraction buffer (0.1 M sodium acetate buffer, 100 μl/well). The absorbance was spectrophotometrically analyzed at 550 nm by an ELISA reader. Flow cytometry analysis [7] Evolution of the cell cycle histogram was performed by flow cytometry analysis to detect the changes in DNA content. Cells (1 × 106) were seeded in a 6-well plate in 2 ml fresh medium and treated with graded concentrations of Vincristine, SAHA, or combination for the indicated time. Then, cells were collected, washed with PBS and fixed with 70 % (v/v) ice cold ethanol at −20 °C for 30 min. The fixed cells were centrifuged to remove the ethanol, rinsed with PBS, resuspended in 0.1 ml DNA extraction buffer (0.2 M Na2HPO4-0.1 M citric buffer, pH 7.8) for 20 min, and subsequently stained with 500 μl PI solution (80 μg/ml propidium iodide, 100 μg/ml RNase A, and 1 % Triton X-100 in PBS) for 20 min at room temperature in the dark. Data were analyzed by FACScan Flow Cytometer and CellQuest software (Becton Dickinson). Immunofluorescence analysis [7] Microtubule distribution and morphology were detected by immunofluorescence. Cover slides were placed in the 24-well plate and coated with poly-d-lysine for 1 day at least to enhance the suspension cells attached to the cover slides. Cells were seeded into the 24-well plate (8 × 105 cells/well) and treated with Vincristine, SAHA, or both drugs for 24 h. The following experiments were performed at room temperature. The cells were fixed with 8 % paraformaldehyde in PBS for 15 min. After washing with PBS for several times, the cells were permeabilized with 0.1 % Triton X-100 in PBS for 10 min. Then, the cells were rinsed with PBS for 10 min three times. For blocking, 3 % BSA in PBS was used. After 1 h, the cells were washed with PBS and incubated with a primary β-tubulin antibody (1:200) for 2 h and FITC-conjugated anti-mouse IgG antibody (1:200) for 2 h. The mounting medium, which contains DAPI stain, was dropped onto the slides, and cover slides were recovered to the slides. Images were detected and captured with the ZEISS LSM 510 META confocal microscope. In vitro cytotoxicity of drug-loaded NPs [6] The cytotoxicity of free drugs, drug-loaded NPs against A549 cell, and A549/taxol cell was evaluated using the standard WST-1 assays. Briefly, A549 cell and A549/taxol cell were seeded onto 96-well plates at a density of 5.0×104 cells per well and cultured for overnight 24 h with 100 μL of DMEM medium or RPMI 1640 medium at 37°C in 5% CO2 atmosphere. Then, removing the original medium, tested cells were incubated, respectively, with fresh medium containing free DOX, free VCR/Vincristine, free (DOX plus VCR), CS-ALG@TPGS-PLGA-VCR NPs, CS-ALG-DOX@TPGS-PLGA NPs, and CS-ALG-DOX@TPGS-PLGA-VCR NPs at different concentrations. After 12 h and 24 h of incubation, 10 μL of WST-1 solution was added to each well and cultured for additional 4 h. Afterward, the cell viability was determined by measuring the absorbance at 450 nm using a microplate reader. The microtubule and the microfilament imaging analysis [6] A549 cell or A549/taxol cell was seeded on a coverslip in a 6-well plate (1×105 cells/well) and incubated overnight in a 5% CO2 incubator at 37°C for attachment. Then the cells were washed and cultured with TPGS-PLGA (0.2 mg/mL) NPs, free VCR/Vincristine (5 μg/mL), and TPGS-PLGA-VCR NPs (5 μg/mL VCR-loaded NPs) for a given time at 37°C, respectively. Cells without treatment were used as control group. Tested cells were washed with PBS and treated with Hoechst 33342 (10 mg/mL) for 20 min. Subsequently, the cells were fixed with 4% paraformaldehyde at room temperature for 10 min, permeated with 1% BSA in phosphate buffered saline containing 0.1% Tween-20 (PBST) for l h. The cells were then exposed to tubulin-tracker red (1:250, diluted in PBST containing 1% BSA, for the microtubule) or actin-tracker green (8 unit/mL, diluted in PBST containing 1% BSA, for the microfilament) at 4°C for 20 min in the dark room. Then, the original medium containing tubulin-tracker red or actin-tracker green was removed and the cells were washed with ice-cold PBS. Finally, the microtubule or the microfilament of the cells was visualized under a CLSM. Analysis of cell cycle [6] The cell cycle induced by empty NPs, free VCR/Vincristine, and drug-loaded NPs on A549/taxol cell was evaluated with cell cycle and apoptosis analysis kit. The cells were seeded at a density of 5×105 cells per well in 6-well plates for 24 h at 37°C, and then treated, respectively, with the fresh medium containing TPGS-PLGA NPs (0.2 mg/mL), free VCR (5 μg/mL), and TPGS-PLGA-VCR NPs (dose of VCR 5 μg/mL). The cells without treatment were used as control. After incubating for 24 h, the cells were harvested by trypsin digestion and centrifugation. Subsequently, the obtained cells were fixed with 70% cold ethanol and stored at 4°C for 24 h. Ultimately, the cells were centrifuged again, washed with cold PBS twice, and stained with 0.5 mL of the staining solution at 37°C for 30 min in the dark. The stained cells were analyzed using a flow cytometer system. Each experiment was performed in triplicates. |
||||
Animal Protocol |
|
||||
ADME/Pharmacokinetics |
Absorption, Distribution and Excretion
The liver is the major excretory organ in humans and animals. 80% of an injected dose of vincristine sulfate is excreted via feces. 10 - 20% is excreted via urine. Within 15 to 30 minutes after injection, over 90% of the drug is distributed from the blood into tissue, where it remains tightly, but not irreversibly, bound. Development of CNS leukemia in patients receiving vincristine and in hematological remission has been interpreted as evidence that ... /vincristine/ penetrates blood-brain barrier poorly. Vincristine ... can be infused into arterial blood supply of tumors in doses several times larger than those that can be admistered iv with comparable toxicity; thus either local uptake or destruction is very rapid. Vinca alkaloids appear to be excreted primarily by liver into bile. Urinary excretion ... over first few hr after injection was ... low in dogs and monkeys. In both ... the drug was distributed to most tissues, but highest concentrations ... found in lung, kidney, spleen, pancreas and liver. In monkeys, vincristine and its metabolites rapidly entered cerebrospinal fluid from plasma to form low concentrations of drug, which persisted for several days. Vincristine sulfate is unpredictably absorbed from the Gl tract. Following rapid iv injection of a 2 mg dose of vincristine in patients with normal renal and hepatic function, peak serum drug concentrations of approximately 0.19-0.89 uM occur immediately and the drug is rapidly cleared from serum. The area under the serum vincristine concentration time curve has been shown to be increased following continuous iv infusion compared with rapid iv injection of the drug when comparable doses are administered. Distribution of vincristine and its metabolites (and/or decomposition products) into human body tissues and fluids has not been fully characterized, but the drug is rapidly and apparently widely distributed following iv administration. Drug that is distributed into tissues is tightly but reversibly bound. Vincristine and its metabolites (and/or decomposition products) are rapidly and extensively distributed into bile, with peak biliary concentrations occurring within 2-4 hr after rapid iv injection of the drug. Vincristine and its metabolites (and/or decomposition products) cross the blood brain barrier poorly following rapid iv injection and generally do not appear in the CSF in cytotoxic concentrations. For more Absorption, Distribution and Excretion (Complete) data for VINCRISTINE (6 total), please visit the HSDB record page. Metabolism / Metabolites Hepatic. Cytochrome P450 isoenzymes of the CYP3A subfamily facilitate the metabolism of vincristine. After iv administration of ... (3)H vincristine, 69% of radioactivity was recovered in feces and 12% in urine over 72 hr period. Approx half ... was in form of metabolites, whose UV spectrum suggested that vincristine dimer was intact. Patients with biliary fistula showed extensive biliary excretion of intact drug (46.5%) & of metabolites (53.5%). Observations suggest that biliary-fecal route ... predominate in excretion ... . The metabolic fate of vincristine has not been clearly determined; the drug appears to be extensively metabolized, probably in the liver, but the extent of metabolism is not clear since the drug also apparently undergoes decomposition in vivo. Hepatic. Cytochrome P450 isoenzymes of the CYP3A subfamily facilitate the metabolism of vincristine. Route of Elimination: The liver is the major excretory organ in humans and animals. 80% of an injected dose of vincristine sulfate is excreted via feces. 10 - 20% is excreted via urine. Half Life: When intravenously injected into cancer patients, a triphasic serum decay patten was observed. The initial, middle, and terminal half-lives are 5 minutes, 2.3 hours, 85 hours respectively. The range of the terminal half-life is humans is 19 - 155 hours. Biological Half-Life When intravenously injected into cancer patients, a triphasic serum decay patten was observed. The initial, middle, and terminal half-lives are 5 minutes, 2.3 hours, 85 hours respectively. The range of the terminal half-life is humans is 19 - 155 hours. After iv administration of ... (3)H vincristine, triphasic decay was observed, with half-lives of 0.85, 7.4 and 164 min ... Following rapid iv injection of vincristine, serum concentrations of the drug appear to decline in a triphasic manner. The terminal elimination half-life of vincristine has ranged from 19-155 hr. |
||||
Toxicity/Toxicokinetics |
Effects During Pregnancy and Lactation
◉ Summary of Use during Lactation Most sources consider breastfeeding to be contraindicated during maternal antineoplastic drug therapy. It is probably impractical to resume breastfeeding after vincristine therapy because of the drug's long half-life. Chemotherapy may adversely affect the normal microbiome and chemical makeup of breastmilk. ◉ Effects in Breastfed Infants In a 4-month-old, neutropenia was probably caused by cyclophosphamide in a mother 9 days after the last of 6 weekly doses of 800 mg cyclophosphamide intravenously, 2 mg vincristine intravenously and daily doses of 30 mg of prednisolone orally. Neutropenia persisted at least 12 days and was accompanied by a brief episode of diarrhea. The contribution of vincristine to the neutropenia cannot be determined. A woman was diagnosed with B-cell lymphoma at 27 weeks of pregnancy. Labor was induced at 34 4/7 weeks and treatment was begun with a standard regimen of rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone in unspecified doses on a 21-day cycle, starting on day 2 postpartum. She pumped and discarded her milk and fed her infant donor milk for the first 10 days of each cycle and then breastfed her infant for the remaining 10 days before the next treatment cycle. The 10-day period of breastfeeding abstinence was determined by using about 3 half-lives of vincristine. After completion of 4 cycles of chemotherapy, her infant was reportedly healthy and developing without any complications. ◉ Effects on Lactation and Breastmilk Relevant published information was not found as of the revision date. Protein Binding ~75% |
||||
References | |||||
Additional Infomation |
Vincristine appears as a white crystalline solid. Melting point 218 °C. Used as an antineoplastic.
Vincristine is a vinca alkaloid with formula C46H56N4O10 found in the Madagascar periwinkle, Catharanthus roseus. It is used (commonly as the corresponding sulfate salt)as a chemotherapy drug for the treatment of leukaemia, lymphoma, myeloma, breast cancer and head and neck cancer. It has a role as a tubulin modulator, a microtubule-destabilising agent, a plant metabolite, an antineoplastic agent and a drug. It is a methyl ester, an acetate ester, a tertiary alcohol, a member of formamides, an organic heteropentacyclic compound, an organic heterotetracyclic compound, a tertiary amino compound and a vinca alkaloid. It is a conjugate base of a vincristine(2+). It derives from a hydride of a vincaleukoblastine. Vincristine is an antitumor vinca alkaloid isolated from Vinca Rosea. It is marketed under several brand names, many of which have different formulations such as Marqibo (liposomal injection) and Vincasar. Vincristine is indicated for the treatment of acute leukaemia, malignant lymphoma, Hodgkin's disease, acute erythraemia, and acute panmyelosis. vincristine sulfate is often chosen as part of polychemotherapy because of lack of significant bone–marrow suppression (at recommended doses) and of unique clinical toxicity (neuropathy). Vincristine has been reported in Taxus cuspidata, Ophioparma ventosa, and other organisms with data available. Vincristine is a natural alkaloid isolated from the plant Vinca rosea Linn. Vincristine binds irreversibly to microtubules and spindle proteins in S phase of the cell cycle and interferes with the formation of the mitotic spindle, thereby arresting tumor cells in metaphase. This agent also depolymerizes microtubules and may also interfere with amino acid, cyclic AMP, and glutathione metabolism; calmodulin-dependent Ca++ -transport ATPase activity; cellular respiration; and nucleic acid and lipid biosynthesis. (NCI04) Vincristine is only found in individuals that have used or taken this drug. It is an antitumor alkaloid isolated from Vinca Rosea. (Merck, 11th ed.) The antitumor activity of Vincristine is thought to be due primarily to inhibition of mitosis at metaphase through its interaction with tubulin. Like other vinca alkaloids, Vincristine may also interfere with: 1) amino acid, cyclic AMP, and glutathione metabolism, 2) calmodulin-dependent Ca2+-transport ATPase activity, 3) cellular respiration, and 4) nucleic acid and lipid biosynthesis. Vincristine is indicated for the treatment of acute leukaemia, malignant lymphoma, Hodgkin's disease, acute erythraemia, and acute panmyelosis. Vincristine sulfate is often chosen as part of polychemotherapy because of lack of significant bone marrow suppression (at recommended doses) and of unique clinical toxicity (neuropathy). An antitumor alkaloid isolated from VINCA ROSEA. (Merck, 11th ed.) See also: Vincristine Sulfate (has salt form). Drug Indication Treatment of acute lymphocytic leukemia (ALL), Hodgkin lymphoma, non-Hodgkin lymphomas, Wilms' tumor, neuroblastoma, rhabdomyosarcoma. Liposomal vincristine is indicated for the treatment of relapsed Philadelphia chromosome-negative (Ph-) acute lymphoblastic leukemia (ALL). FDA Label Mechanism of Action The antitumor activity of Vincristine is thought to be due primarily to inhibition of mitosis at metaphase through its interaction with tubulin. Like other vinca alkaloids, Vincristine may also interfere with: 1) amino acid, cyclic AMP, and glutathione metabolism, 2) calmodulin-dependent Ca2+-transport ATPase activity, 3) cellular respiration, and 4) nucleic acid and lipid biosynthesis. Vinca alkaloids are cell cycle specific agents ... /which/ block mitosis and produce metaphase arrest. Biological activities of these drugs can be explained by their ability to bind specifically tubulin, and to block ability of the protein to polymerize into microtubules ... through disruption of microtubules of mitotic apparatus, cell division is arrested in metaphase. In absence of intact mitotic spindle, chromosomes may disperse throughout cytoplasm ... or may occur in unusual groupings ... inability to segregate chromosomes correctly during mitosis presumably leads to cellular death. /Vinca alkaloids/ Although the mechanism of action has not been definitely established, vincristine appears to bind to or crystallize critical microtubular proteins of the mitotic spindle, thus preventing their proper polymerization and causing metaphase arrest. In high concentrations, the drug also exerts complex effects on nucleic acid and protein synthesis. Vincristine exerts some immunosuppressive activity. Vincristine Sulfate can cause developmental toxicity according to state or federal government labeling requirements. Vincristine sulfate appears as an anticancer drug. White to slightly yellow, amorphous or crystalline powder. Sensitive to light. Odorless. pH (0.1% solution) 3.5 - 4.5. (NTP, 1992) Vincristine Sulfate is the sulfate salt of a natural alkaloid isolated from the plant Catharanthus roseus (Vinca rosea L.) with antimitotic and antineoplastic activities. Vincristine binds irreversibly to microtubules and spindle proteins in S phase of the cell cycle and interferes with the formation of the mitotic spindle, thereby arresting tumor cells in metaphase. This agent also depolymerizes microtubules and may also interfere with amino acid, cyclic AMP, and glutathione metabolism; calmodulin-dependent Ca(2+)-activated ATPase activity; cellular respiration; and nucleic acid and lipid biosynthesis. Vincristine Sulfate Liposome is a sphingomyelin/cholesterol liposomal formulation of vincristine sulfate with potential antineoplastic activity. Vincristine, a vinca alkaloid isolated from the plant Vinca rosea, irreversibly binds to and stabilizes tubulin, thereby interrupting microtubule assembly/disassembly dynamics, thereby preventing the formation of the mitotic spindle and leading to cell cycle arrest in metaphase. Liposomal encapsulation prolongs bioavailability of vincristine, increases its delivery to tumor tissues and reduces its toxicity profile. Compared to standard liposomal delivery, sphingosomal drug delivery further increases circulation time of serum drug and enhances drug accumulation at tumor sites, thereby leading to a further increase in efficacy. An antitumor alkaloid isolated from VINCA ROSEA. (Merck, 11th ed.) See also: Vincristine (has active moiety). Vinblastine or colchicine, administered intraperitoneally to B6D2F1 mice with advanced subcutaneous colon 38 tumours, induced substantial tumour growth delays with progressive development of haemorrhagic necrosis beginning within 8 hours of treatment. Two multidrug-resistant P388 leukaemia sublines, refractory to vinblastine and vincristine when grown as intraperitoneal ascites, were sensitive to necrosis induction when grown as subcutaneous tumours. Vascular labelling with two fluorescent markers indicated that vincristine substantially reduced tumour blood flow within 4 hours after treatment. The effects of vinblastine, vincristine and colchicine were similar to those of tumour necrosis factor alpha in that: (a) similar tumour necrosis and blood flow changes were induced, (b) coadministration of the serotonin antagonist cyproheptidine prevented tumour necrosis and (c) plasma nitrate levels were elevated, indicative of the stimulation of oxidation of L-arginine to nitric oxide. The results suggest that vinca alkaloids and colchicine act on solid tumours by host cell-mediated vascular effects as well as by direct tubulin-mediated cytotoxicity.[5] Combination chemotherapy in clinical practice has been generally accepted as a feasible strategy for overcoming multidrug resistance (MDR). Here, we designed and successfully prepared a co-delivery system named S-D1@L-D2 NPs, where denoted some smaller nanoparticles (NPs) carrying a drug doxorubicin (DOX) were loaded into a larger NP containing another drug (vincristine [VCR]) via water-in-oil-in-water double-emulsion solvent diffusion-evaporation method. Chitosan-alginate nanoparticles carrying DOX (CS-ALG-DOX NPs) with a smaller diameter of about 20 nm formed S-D1 NPs; vitamin E D-α-tocopheryl polyethylene glycol 1000 succinate-modified poly(lactic-co-glycolic acid) nanoparticles carrying VCR (TPGS-PLGA-VCR NPs) with a larger diameter of about 200 nm constituted L-D2 NPs. Some CS-ALG-DOX NPs loaded into TPGS-PLGA-VCR NPs formed CS-ALG-DOX@TPGS-PLGA-VCR NPs. Under the acidic environment of cytosol and endosome or lysosome in MDR cell, CS-ALG-DOX@TPGS-PLGA-VCR NPs released VCR and CS-ALG-DOX NPs. VCR could arrest cell cycles at metaphase by inhibiting microtubule polymerization in the cytoplasm. After CS-ALG-DOX NPs escaped from endosome, they entered the nucleus through the nuclear pore and released DOX in the intra-nuclear alkaline environment, which interacted with DNA to stop the replication of MDR cells. These results indicated that S-D1@L-D2 NPs was a co-delivery system of intracellular precision release loaded drugs with pH-sensitive characteristics. S-D1@L-D2 NPs could obviously enhance the in vitro cytotoxicity and the in vivo anticancer efficiency of co-delivery drugs, while reducing their adverse effects. Overall, S-D1@L-D2 NPs can be considered an innovative platform for the co-delivery drugs of clinical combination chemotherapy for the treatment of MDR tumor. [6] Background Combination therapy is a key strategy for minimizing drug resistance, a common problem in cancer therapy. The microtubule-depolymerizing agent vincristine is widely used in the treatment of acute leukemia. In order to decrease toxicity and chemoresistance of vincristine, this study will investigate the effects of combination vincristine and vorinostat (suberoylanilide hydroxamic acid (SAHA)), a pan-histone deacetylase inhibitor, on human acute T cell lymphoblastic leukemia cells. Methods Cell viability experiments were determined by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay, and cell cycle distributions as well as mitochondria membrane potential were analyzed by flow cytometry. In vitro tubulin polymerization assay was used to test tubulin assembly, and immunofluorescence analysis was performed to detect microtubule distribution and morphology. In vivo effect of the combination was evaluated by a MOLT-4 xenograft model. Statistical analysis was assessed by Bonferroni’s t test. Results Cell viability showed that the combination of vincristine and SAHA exhibited greater cytotoxicity with an IC50 value of 0.88 nM, compared to each drug alone, 3.3 and 840 nM. This combination synergically induced G2/M arrest, followed by an increase in cell number at the sub-G1 phase and caspase activation. Moreover, the results of vincristine combined with an HDAC6 inhibitor (tubastatin A), which acetylated α-tubulin, were consistent with the effects of vincristine/SAHA co-treatment, thus suggesting that SAHA may alter microtubule dynamics through HDAC6 inhibition. Conclusion These findings indicate that the combination of vincristine and SAHA on T cell leukemic cells resulted in a change in microtubule dynamics contributing to M phase arrest followed by induction of the apoptotic pathway. These data suggest that the combination effect of vincristine/SAHA could have an important preclinical basis for future clinical trial testing.[7] In our study, SA1 mechanoreceptors were impaired by acutely exposing dissociated whisker hair follicles to vincristine in vitro. On the other hand, whisker tactile behavioral responses were impaired days after the in vivo vincristine treatment regimen. The delayed impairment of the sensory behavioral responses could be due to slow accumulation of vincristine in whisker hair follicles during the in vivo vincristine treatment. In addition, SA1 mechanoreceptors or Merkel discs are located deep in hair follicles and shielded by a layer of glassy membranes, which may not favor the diffusion of vincristine into Merkel disc regions during the in vivo vincristine treatment. Alternatively, whisker tactile responses may be compensated by RA and SA2 when the impairment of SA1 mechanoreceptors by vincristine was not very severe in the early stage of the in vivo vincristine treatment. Although the in vivo vincristine treatment regimen used in the present study did not impair RA and SA2 mechanoreceptors, it will be interesting to examine whether a prolonged in vivo vincristine treatment may eventually impair these two mechanoreceptors as well. It will also be interesting to examine whether and how long the vincristine-induced impairment of mechanoreceptors can be recovered following the termination of a vincristine treatment regimen.[8] |
Molecular Formula |
C46H56N4O10
|
---|---|
Molecular Weight |
824.9577
|
Exact Mass |
824.399
|
Elemental Analysis |
C, 66.97; H, 6.84; N, 6.79; O, 19.39
|
CAS # |
57-22-7
|
Related CAS # |
57-22-7;2068-78-2 (sulfate);
|
PubChem CID |
5978
|
Appearance |
Blades from methanol
|
Density |
1.4±0.1 g/cm3
|
Melting Point |
211-216ºC
|
Index of Refraction |
1.677
|
LogP |
2.82
|
Hydrogen Bond Donor Count |
3
|
Hydrogen Bond Acceptor Count |
12
|
Rotatable Bond Count |
10
|
Heavy Atom Count |
60
|
Complexity |
1750
|
Defined Atom Stereocenter Count |
9
|
SMILES |
CC[C@@]1(C[C@@H]2C[C@@](C3=C(CCN(C2)C1)C4=CC=CC=C4N3)(C5=C(C=C6C(=C5)[C@]78CCN9[C@H]7[C@@](C=CC9)([C@H]([C@@]([C@@H]8N6C=O)(C(=O)OC)O)OC(=O)C)CC)OC)C(=O)OC)O
|
InChi Key |
OGWKCGZFUXNPDA-XQKSVPLYSA-N
|
InChi Code |
InChI=1S/C46H56N4O10/c1-7-42(55)22-28-23-45(40(53)58-5,36-30(14-18-48(24-28)25-42)29-12-9-10-13-33(29)47-36)32-20-31-34(21-35(32)57-4)50(26-51)38-44(31)16-19-49-17-11-15-43(8-2,37(44)49)39(60-27(3)52)46(38,56)41(54)59-6/h9-13,15,20-21,26,28,37-39,47,55-56H,7-8,14,16-19,22-25H2,1-6H3/t28-,37+,38-,39-,42+,43-,44-,45+,46+/m1/s1
|
Chemical Name |
methyl (1R,9R,10S,11R,12R,19R)-11-acetyloxy-12-ethyl-4-[(13S,15S,17S)-17-ethyl-17-hydroxy-13-methoxycarbonyl-1,11-diazatetracyclo[13.3.1.04,12.05,10]nonadeca-4(12),5,7,9-tetraen-13-yl]-8-formyl-10-hydroxy-5-methoxy-8,16-diazapentacyclo[10.6.1.01,9.02,7.016,19]nonadeca-2,4,6,13-tetraene-10-carboxylate
|
Synonyms |
vincristine; 22-Oxovincaleukoblastine; Leurocristine; Vinkristin; Vincrystine; 57-22-7; Vincristina; leucristine;
|
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 | 1.2122 mL | 6.0609 mL | 12.1218 mL | |
5 mM | 0.2424 mL | 1.2122 mL | 2.4244 mL | |
10 mM | 0.1212 mL | 0.6061 mL | 1.2122 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.
Testing CC-486 (Oral Azacitidine) Plus the Standard Drug Therapy in Patients 75 Years or Older With Newly Diagnosed Diffuse Large B Cell Lymphoma
CTID: NCT04799275
Phase: Phase 2/Phase 3   Status: Recruiting
Date: 2024-11-27